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.

  • Article
  • Published:

Development of multiplexed orthogonal base editor (MOBE) systems

Abstract

Base editors (BEs) enable efficient, programmable installation of point mutations while avoiding the use of double-strand breaks. Simultaneous application of two or more different BEs, such as an adenine BE (which converts A·T base pairs to G·C) and a cytosine BE (which converts C·G base pairs to T·A), is not feasible because guide RNA crosstalk results in non-orthogonal editing, with all BEs modifying all target loci. Here we engineer both adenine BEs and cytosine BEs that can be orthogonally multiplexed by using RNA aptamer–coat protein systems to recruit the DNA-modifying enzymes directly to the guide RNAs. We generate four multiplexed orthogonal BE systems that enable rates of precise co-occurring edits of up to 7.1% in the same DNA strand without enrichment or selection strategies. The addition of a fluorescent enrichment strategy increases co-occurring edit rates up to 24.8% in human cells. These systems are compatible with expanded protospacer adjacent motif and high-fidelity Cas9 variants, function well in multiple cell types, have equivalent or reduced off-target propensities compared with their parental systems and can model disease-relevant point mutation combinations.

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: A schematic overview of multiplexed orthogonal base editing.
Fig. 2: Engineering of an ABE aptamer system.
Fig. 3: Engineering of a CBE aptamer system.
Fig. 4: Characterization of optimized CBE and ABE aptamer systems.
Fig. 5: On-target and crosstalk editing efficiencies of MOBE systems and parental BEs when multiplexing CBE and ABE.
Fig. 6: On-target and crosstalk editing efficiencies of MOBEs when performing multiplexed editing followed by FACS enrichment for episomal edits.

Similar content being viewed by others

Data availability

NGS data are available on the National Center for Biotechnology Information Sequencing Read Archive database under project number PRJNA836633 ref. 89. Plasmids from this study will be available at Addgene (plasmids 219934–219949; https://www.addgene.org/Alexis_Komor/). Homo sapiens Hg38 was used as the reference genome for MOBEnto quantification of co-occurring edits.

Code availability

Source code for MOBEnto (quantification of haplotypes from multiplexed genome editing using next-generation sequencing data) is available at https://github.com/Komorlab/MOBEnto (ref. 90).

References

  1. Green, R. C. et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet. Med. 15, 565–574 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rehm, H. L. et al. ClinGen—the clinical genome resource. N. Engl. J. Med. 372, 2235–2242 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Landrum, M. J. et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 46, D1062–D1067 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Starita, L. M. et al. Variant interpretation: functional assays to the rescue. Am. J. Hum. Genet. 101, 315–325 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shah, N. et al. Identification of misclassified clinvar variants via disease population prevalence. Am. J. Hum. Genet. 102, 609–619 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cooper, D. N., Krawczak, M., Polychronakos, C., Tyler-Smith, C. & Kehrer-Sawatzki, H. Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease. Hum. Genet. 132, 1077–1130 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rahit, K. M. T. H. & Tarailo-Graovac, M. Genetic modifiers and rare Mendelian disease. Genes 11, 239 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mukherjee, S. et al. Identifying digenic disease genes via machine learning in the Undiagnosed Diseases Network. Am. J. Hum. Genet. 108, 1946–1963 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Findlay, G. M. et al. Accurate classification of BRCA1 variants with saturation genome editing. Nature 562, 217–222 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765–771 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Turchiano, G. et al. Quantitative evaluation of chromosomal rearrangements in gene-edited human stem cells by CAST-Seq. Cell Stem Cell 28, 1136–1147.e5 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Leibowitz, M. L. et al. Chromothripsis as an on-target consequence of CRISPR–Cas9 genome editing. Nat. Genet. 53, 895–905 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ihry, R. J. et al. P53 inhibits CRISPR–Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939–946 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. 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  PubMed  PubMed Central  Google Scholar 

  21. 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  PubMed  PubMed Central  Google Scholar 

  22. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jiang, F. et al. Structures of a CRISPR–Cas9 R-loop complex primed for DNA cleavage. Science 351, 867–871 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sherry, S. T. et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 29, 308–311 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Webber, B. R. et al. Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors. Nat. Commun. 10, 5222 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Walton, R. T., Christie, K. A., Whittaker, M. N. & Kleinstiver, B. P. Unconstrained genome targeting with near-PAMless engineered CRISPR–Cas9 variants. Science 368, 290–296 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ting, P. Y. et al. Guide Swap enables genome-scale pooled CRISPR–Cas9 screening in human primary cells. Nat. Methods 15, 941–946 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Glaser, V. et al. Combining different CRISPR nucleases for simultaneous knock-in and base editing prevents translocations in multiplex-edited CAR T cells. Genome Biol. 24, 89 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Porto, E. M., Komor, A. C., Slaymaker, I. M. & Yeo, G. W. Base editing: advances and therapeutic opportunities. Nat. Rev. Drug Discov. 19, 839–859 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zalatan, J. G. et al. Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds. Cell 160, 339–350 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Ma, H. et al. Multiplexed labeling of genomic loci with dCas9 and engineered sgRNAs using CRISPRainbow. Nat. Biotechnol. 34, 528–530 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wang, S., Su, J.-H., Zhang, F. & Zhuang, X. An RNA–aptamer-based two-color CRISPR labeling system. Sci. Rep. 6, 26857 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fukunaga, K. & Yokobayashi, Y. Directed evolution of orthogonal RNA–RBP pairs through library-vs-library in vitro selection. Nucleic Acids Res. 50, 601–616 (2021).

    Article  PubMed Central  Google Scholar 

  37. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR–Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Multiplexable, locus-specific targeting of long RNAs with CRISPR–Display. Nat. Methods 12, 664–670 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Valegård, K. et al. The three-dimensional structures of two complexes between recombinant MS2 capsids and RNA operator fragments reveal sequence-specific protein–RNA interactions. J. Mol. Biol. 270, 724–738 (1997).

    Article  PubMed  Google Scholar 

  41. Tars, K., Fridborg, K., Bundule, M. & Liljas, L. The three-dimensional structure of bacteriophage PP7 from Pseudomonas aeruginosa at 3.7-Å resolution. Virology 272, 331–337 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Collantes, J. C. et al. Development and characterization of a modular CRISPR and RNA aptamer-mediated base editing system. CRISPR J. 4, 58–68 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, C. et al. SWISS: multiplexed orthogonal genome editing in plants with a Cas9 nickase and engineered CRISPR RNA scaffolds. Genome Biol. 21, 141 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Richter, M. F. et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat. Biotechnol. 38, 883–891 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nguyen Tran, M. T. et al. Engineering domain-inlaid SaCas9 adenine base editors with reduced RNA off-targets and increased on-target DNA editing. Nat. Commun. 11, 4871 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Gaudelli, N. M. et al. Directed evolution of adenine base editors with increased activity and therapeutic application. Nat. Biotechnol. 38, 892–900 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  49. Zhang, X. et al. Increasing the efficiency and targeting range of cytidine base editors through fusion of a single-stranded DNA-binding protein domain. Nat. Cell Biol. 22, 740–750 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 36, 843–848 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Thuronyi, B. W. et al. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat. Biotechnol. 37, 1070–1079 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Yu, Y. et al. Cytosine base editors with minimized unguided DNA and RNA off-target events and high on-target activity. Nat. Commun. 11, 2052 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, Y. et al. sgBE: a structure-guided design of sgRNA architecture specifies base editing window and enables simultaneous conversion of cytosine and adenosine. Genome Biol. 21, 222 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Fu, J. L., Kanno, T., Liang, S.-C., Matzke, A. J. M. & Matzke, M. GFP Loss-of-Function Mutations in Arabidopsis thaliana. G3 5, 1849–1855 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Martin, A. St. et al. A panel of eGFP reporters for single base editing by APOBEC–Cas9 editosome complexes. Sci. Rep. 9, 497 (2019)

    Article  PubMed  PubMed Central  Google Scholar 

  56. Di Blasi, R., Marbiah, M. M., Siciliano, V., Polizzi, K. & Ceroni, F. A call for caution in analysing mammalian co-transfection experiments and implications of resource competition in data misinterpretation. Nat. Commun. 12, 2545 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Vakulskas, C. A. et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat. Med. 24, 1216–1224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Standage-Beier, K. et al. A transient reporter for editing enrichment (TREE) in human cells. Nucleic Acids Res. 47, e120 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Doman, J. L., Raguram, A., Newby, G. A. & Liu, D. R. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat. Biotechnol. 38, 620–628 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. 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  PubMed  Google Scholar 

  61. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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  PubMed  PubMed Central  Google Scholar 

  63. Nachtegael, C. et al. Scaling up oligogenic diseases research with OLIDA: the Oligogenic Diseases Database. Database 2022, baac023 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Arbab, M. et al. Determinants of base editing outcomes from target library analysis and machine learning. Cell 182, 463–480.e30 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kim, N. et al. Deep learning models to predict the editing efficiencies and outcomes of diverse base editors. Nat. Biotechnol. 42, 484–497 (2023).

    Article  PubMed  Google Scholar 

  66. Kovalevich, J. & Langford, D. in Neuronal Cell Culture (eds Amini, S. & White, M. K.) Vol. 1078, 9–21 (Humana Press, 2013).

  67. Shipley, M. M., Mangold, C. A. & Szpara, M. L. Differentiation of the SH-SY5Y human neuroblastoma cell line. J. Vis. Exp. 17, 53193 (2016).

    Google Scholar 

  68. Garaffo, G. et al. The Dlx5 and Foxg1 transcription factors, linked via miRNA-9 and -200, are required for the development of the olfactory and GnRH system. Mol. Cell. Neurosci. 68, 103–119 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Yan, Y. et al. Endoplasmic reticulum stress-related calcium imbalance plays an important role on zinc oxide nanoparticles-induced failure of neural tube closure during embryogenesis. Environ. Int. 152, 106495 (2021).

    Article  CAS  PubMed  Google Scholar 

  70. Riordan, J. D. & Nadeau, J. H. From peas to disease: modifier genes, network resilience, and the genetics of health. Am. J. Hum. Genet. 101, 177–191 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 40, 402–410 (2022).

    Article  CAS  PubMed  Google Scholar 

  72. Yuan, Q. & Gao, X. Multiplex base- and prime-editing with drive-and-process CRISPR arrays. Nat. Commun. 13, 2771 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Di Blasi, R. et al. Resource-aware construct design in mammalian cells. Nat. Commun. 14, 3576 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Maynard, S. et al. Human embryonic stem cells have enhanced repair of multiple forms of DNA damage. Stem Cells 26, 2266–2274 (2008).

    Article  PubMed  Google Scholar 

  75. Wang, X. et al. Cas12a base editors induce efficient and specific editing with low DNA damage response. Cell Rep. 31, 107723 (2020).

    Article  CAS  PubMed  Google Scholar 

  76. Park, J.-C. et al. High expression of uracil DNA glycosylase determines C to T substitution in human pluripotent stem cells. Mol. Ther. Nucleic Acids 27, 175–183 (2022).

    Article  CAS  PubMed  Google Scholar 

  77. Park, J.-C., Kim, Y.-J., Kang, C. Y., Bae, S. & Cha, H.-J. Enhancing precise genome editing in human pluripotent stem cells through dual inhibition of dna damage response and repair pathways. Preprint at Research Square https://doi.org/10.21203/rs.3.rs-3028152/v1 (2023).

  78. Wang, C. et al. Microbial single-strand annealing proteins enable CRISPR gene-editing tools with improved knock-in efficiencies and reduced off-target effects. Nucleic Acids Res. 49, e36 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Najm, F. J. et al. Orthologous CRISPR–Cas9 enzymes for combinatorial genetic screens. Nat. Biotechnol. 36, 179–189 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. McCarty, N. S., Graham, A. E., Studená, L. & Ledesma-Amaro, R. Multiplexed CRISPR technologies for gene editing and transcriptional regulation. Nat. Commun. 11, 1281 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Vasquez, C. A., Cowan, Q. T. & Komor, A. C. Base editing in human cells to produce single‐nucleotide‐variant clonal cell lines. Curr. Protoc. Mol. Biol. 133, e129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Salomonsen, B., Mortensen, U. H. & Halkier, B. A. in DNA Cloning and Assembly Methods (eds Valla, S. & Lale, R.) vol. 1116, 59–72 (Humana Press, 2014).

  83. Adikusuma, F., Pfitzner, C. & Thomas, P. Q. Versatile single-step-assembly CRISPR–Cas9 vectors for dual gRNA expression. PLoS ONE 12, e0187236 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  84. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Rallapalli, K. L., Ranzau, B. L., Ganapathy, K. R., Paesani, F. & Komor, A. C. Combined theoretical, bioinformatic, and biochemical analyses of RNA editing by adenine base editors. CRISPR J. 5, 294–310 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Liao, Y., Smyth, G. K. & Shi, W. The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads. Nucleic Acids Res. 47, e47 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Cowan, Q. T. et al. Development of multiplexed orthogonal base editor (MOBE) systems. NCBI Bioproject https://www.ncbi.nlm.nih.gov/bioproject/PRJNA836633 (2024).

  90. Cowan, Q. T. et al. MOBEnto (quantification of haplotypes from multiplexed genome editing using next-generation sequencing data). GitHub https://github.com/Komorlab/MOBEnto (2024).

Download references

Acknowledgements

We are grateful to all members of the Komor laboratory for helpful discussions. We are thankful to G. Debelouchina for use of the Neon transfection system. We would also like to thank A. Nelson, N. Chi, C. Sawyer and A. Goren. This research was supported by the University of California, San Diego and the National Institutes of Health (NIH) through grant no. 1R35GM138317 (to A.C.K) and the Research Corporation for Science Advancement through award number 28385 (to A.C.K). Q.T.C. was supported by the Molecular Biophysics Training grant, NIH grant T32 GM008326. B.L.R. was supported by the Chemistry–Biology Interface Training Program, NIH grant T32 GM112584.

Author information

Authors and Affiliations

Authors

Contributions

Q.T.C. contributed to conceptualization of the research project, experimental design, data curation, data analysis and writing of the manuscript. S.G. contributed to data curation and data analysis. W.G. contributed to data analysis. B.L.R. contributed to experimental design. T.S.S. contributed to data analysis and supervision of the work. A.C.K. contributed to conceptualization of the research project, experimental design, data analysis, supervision of the work, writing of the manuscript and acquisition of the funding. All authors contributed to editing of the manuscript.

Corresponding author

Correspondence to Alexis C. Komor.

Ethics declarations

Competing interests

A.C.K. is a member of the SAB of Pairwise Plants, is an equity holder for Pairwise Plants and Beam Therapeutics, and receives royalties from Pairwise Plants, Beam Therapeutics and Editas Medicine via patents licensed from Harvard University. A.C.K.’s interests have been reviewed and approved by the University of California, San Diego, in accordance with its conflict of interest policies. A.C.K. and Q.T.C. are listed as inventors in a Regents of the University of California patent application (PCT/US23/66812) on the development and application of multiplexed orthogonal base editing systems in living cells. All other authors declare no competing financial interests.

Peer review

Peer review information

Nature Biotechnology thanks Nozomu Yachie and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 On-target and crosstalk editing efficiencies of MOBEs and parental BEs when targeted to two protospacers at distinct chromosomes.

(a–f) Editing efficiencies by the MOBE1-4 systems and the evoBE4/ABE8e and evoBE4/ABE8.20 combinations at the HIRA.0/HEK3.0 (A and D), HEK2.0/RNF2.0 (B and E), and HIRA.3/RNF2.0 (C and F) loci. HEK293T cells were treated as previously described in Fig. 5. (a–c) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target (y-axis, filled circles), C·G edited to T·A at the ABE target (y-axis, circles with X inset), C·G edited to T·A at the CBE target (x-axis, filled circles), and A·T edited to G·C at the CBE target (x-axis, circles with X inset). Dot plots and error bars represent the average and SEM for n = 3 biological replicates. (d–f) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target (on-target efficiencies only). Bar graphs and error bars represent the average and SEM for n = 3 biological replicates (each replicate is marked individually). Negative control (NC) samples are also shown, in which cells were treated identically but transfected with a non-targeting gRNA. (g) Orthogonality scores for the MOBE1-4 systems and the evoBE4/ABE8e and evoBE4/ABE8.20 combinations at the three protospacer combinations from (A) through (C). Plotted are the log2 of the ‘CBE orthogonality scores’, which we define for a given protospacer combination as the percent of total DNA sequencing reads with C·G edited to T·A at the CBE target divided by the percent of total DNA sequencing reads with C·G edited to T·A at the ABE target. The ‘ABE orthogonality scores’ are the A·T to G·C editing efficiency equivalent. Dot plots and error bars represent the average and propagation of uncertainty of the SEM for n = 3 biological replicates.

Extended Data Fig. 2 On-target and crosstalk editing efficiencies of MOBEs and parental BEs when targeted to two protospacers within the same locus.

(a–f) Editing efficiencies by the MOBE1-4 systems and the evoBE4/ABE8e and evoBE4/ABE8.20 combinations at the HEK3 (A and D), EMX1 (B and E), and RNF2 (C and F) loci. Cells were treated as previously described in Fig. 5. (a–c) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target (y-axis, solid circles), C·G edited to T·A at the ABE target (y-axis, circles with X inset), C·G edited to T·A at the CBE target (x-axis, solid circles), and A·T edited to G·C at the CBE target (x-axis, circles with X inset). Dot plots and error bars represent the average and SEM for n = 3 biological replicates. (d–f) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target (on-target efficiencies only). Bar graphs and error bars represent the average and SEM for n = 3 biological replicates at EMX1 and HEK3 or n = 4 biological replicates at RNF2 (each replicate is marked individually). Negative control (NC) samples are also shown, in which cells were treated identically but transfected with a non-targeting gRNA. (g) Orthogonality scores for the MOBE1-4 systems and the evoBE4/ABE8e and evoBE4/ABE8.20 combinations at the three protospacer combinations from (A) through (C). Plotted are the log2 of the ‘CBE orthogonality scores’, which we define for a given protospacer combination as the percent of total DNA sequencing reads with C·G edited to T·A at the CBE target divided by the percent of total DNA sequencing reads with C·G edited to T·A at the ABE target. The ‘ABE orthogonality scores’ are the A·T to G·C editing efficiency equivalent. Dot plots and error bars represent the average and propagation of uncertainty of the SEM for n = 3 biological replicates at EMX1 and HEK3 or n = 4 biological replicates at RNF2.

Extended Data Fig. 3 Quantification of indel rates for parental and MOBE systems.

(a, b) Indel introduction rates by the MOBE1-4 systems and the evoBE4/ABE8e and evoBE4/ABE8.20 combinations at the HIRA.0/HEK3.0, HEK2.0/RNF2.0, HIRA.3/RNF2.0 (A) distinct loci protospacer combinations and the HEK3, EMX1, and RNF2 (B) single-amplicon protospacer combinations. Cells were treated as previously described in Fig. 5. Plotted are the percent of total DNA sequencing reads with indels at each target site. Bar graphs and error bars represent the average and SEM for n = 3 biological replicates (for most sites) or n = 4 biological replicates at RNF2 (each replicate is marked individually). Negative control (NC) samples are also shown, in which cells were treated identically but transfected with a non-targeting gRNA.

Extended Data Fig. 4 Quantification of co-occurring orthogonal edits.

Cells were treated as previously described in Fig. 6. Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target, with no crosstalk edits when all multiplexing systems are targeted to two protospacers within the HEK3 (A), EMX1 (B), or RNF2 (C) locus. Bar graphs and error bars represent the average and SEM for n = 3 biological replicates at EMX1 and HEK3 or n = 4 biological replicates at RNF2 (each replicate is marked individually).

Extended Data Fig. 5 On-target and crosstalk editing efficiencies of MOBEs when targeted to two protospacers at distinct chromosomes followed by FACS enrichment for episomal edits.

(a–f) Editing efficiencies by the MOBE1-4 systems at the HIRA.0/HEK3.0 (A and D), HEK2.0/RNF2.0 (B and E), and HIRA.3/RNF2.0 (C and F) loci. HEK293T cells were treated as previously described in Fig. 6. (a-c) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target (y-axis, solid circles), C·G edited to T·A at the ABE target (y-axis, circles with X inset), C·G edited to T·A at the CBE target (x-axis, solid circles), and A·T edited to G·C at the CBE target (x-axis, circles with X inset). Dot plots and error bars represent the average and SEM for n = 3 biological replicates. (d-f) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target (on-target efficiencies only). Bar graphs and error bars represent the average and propagation of uncertainty of the SEM for n = 3 biological replicates (each replicate is marked individually). Negative control (NC) samples are also shown, in which cells were not transfected with any plasmid.

Extended Data Fig. 6 On-target and crosstalk editing efficiencies of MOBEs when targeted to two protospacers within the same locus followed by FACS enrichment for episomal edits.

(a-f) Editing efficiencies by the MOBE1-4 systems at the HEK3 (A and D), EMX1 (B and E), and RNF2 (C and F) loci. Cells were treated as previously described in Fig. 6. (a–c) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target (y-axis, solid circles), C·G edited to T·A at the ABE target (y-axis, circles with X inset), C·G edited to T·A at the CBE target (x-axis, solid circles), and A·T edited to G·C at the CBE target (x-axis, circles with X inset). Dot plots and error bars represent the average and SEM for n = 3 biological replicates at EMX1 and HEK3 or n = 4 biological replicates at RNF2. (d-f) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target (on-target efficiencies only). Bar graphs and error bars represent the average and SEM for n = 3 biological replicates at EMX1 and HEK3 or n = 4 biological replicates at RNF2 (each replicate is marked individually). Negative control (NC) samples are also shown, in which cells were not transfected with any plasmid.

Extended Data Fig. 7 MOBEs can be used in HeLa cells.

(a-c) Editing efficiencies by the MOBE2 and MOBE3 systems at the RNF2 (A), HEK3 (B), and HIRA.0/HEK3.0 (C) protospacer combinations. HeLa cells were transfected with plasmids encoding nCas9-NG-P2A-mCherry, tandem CP-deaminase fusions, both gRNA aptamers targeting endogenous loci, and the 2x-dead-GFP reporter. After 72 hours, the population of GFP+/mCherry+ ‘enriched’ cells was collected by FACS. Genomic loci of interest were then amplified and analyzed by NGS. Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target (y-axis, solid circles), C·G edited to T·A at the ABE target (y-axis, circles with X inset), C·G edited to T·A at the CBE target (x-axis, solid circles), and A·T edited to G·C at the CBE target (x-axis, circles with X inset). (d) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target, with no crosstalk edits when all multiplexing systems are targeted to two protospacers within the HEK3 or RNF2 loci. (e) Plotted are the percent of total DNA sequencing reads with indels at each target site. (f) Orthogonality scores for the MOBE systems at the two protospacer combinations from (A) through (C). Plotted are the log2 of the ‘CBE orthogonality scores’, which we define for a given protospacer combination as the percent of total DNA sequencing reads with C·G edited to T·A at the CBE target divided by the percent of total DNA sequencing reads with C·G edited to T·A at the ABE target. The ‘ABE orthogonality scores’ are the A·T to G·C editing efficiency equivalent. Dot plots and error bars represent the average and standard deviation for n = 3 biological replicates. Negative control (NC) samples are also shown, in which cells were treated identically but transfected with a non-targeting gRNA.

Extended Data Fig. 8 MOBEs can be used in SH-SY5Y cells.

(a-b) Editing efficiencies by the MOBE1 and MOBE3 systems at the HEK3 (A), and RNF2 (B) single-amplicon protospacer combinations. SH-SY5Y cells were electroporated with plasmids encoding nCas9-NG-P2A-mCherry, tandem CP-deaminase fusions, and both gRNA aptamers. After 72 hours, the population of transfected (mCherry+) cells was collected by FACS. Genomic loci of interest were then amplified and analyzed by NGS. Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target (y-axis, solid circles), C·G edited to T·A at the ABE target (y-axis, circles with X inset), C·G edited to T·A at the CBE target (x-axis, solid circles), and A·T edited to G·C at the CBE target (x-axis, circles with X inset). (c) Plotted are the percent of total DNA sequencing reads with A·T edited to G·C at the ABE target and C·G edited to T·A at the CBE target, with no crosstalk edits when all multiplexing systems are targeted to two protospacers within the HEK3 or RNF2 loci. (d) Plotted are the percent of total DNA sequencing reads with indels at each target site. (e) Orthogonality scores for the MOBE systems at the two protospacer combinations from (A) and (B). Plotted are the log2 of the ‘CBE orthogonality scores’, which we define for a given protospacer combination as the percent of total DNA sequencing reads with C·G edited to T·A at the CBE target divided by the percent of total DNA sequencing reads with C·G edited to T·A at the ABE target. The ‘ABE orthogonality scores’ are the A·T to G·C editing efficiency equivalent. Dot plots and error bars represent the average and standard deviation for n = 3 biological replicates. Negative control (NC) samples are also shown, in which cells were treated identically but transfected with a non-targeting gRNA.

Extended Data Fig. 9 MOBEs enable the installation of orthogonal point mutations known to cause Kallmann syndrome (OLI368).

(a) Schematic of the DNA and protein sequences of the OLI368 combination, FGFR1 c.2075A>G and DUSP6 c.545C>T, with MOBE protospacer and PAMs indicated. (b) Optimization of gRNA pairs with MOBE1 and MOBE3 to avoid bystander adenine editing in HEK293T cells. (c) Bulk editing efficiencies of the FGFR1 A7 protospacer combined with DUSP6 C7 and C8 protospacers with inset (D) to display crosstalk in HEK293T cells. (e) Editing efficiencies of MOBE3 with the optimal A7-C8 protospacer combination after enrichment with the all-in-one fluorescent reporter plasmid in HEK293T cells. (f) Editing efficiencies of MOBE3 (A7-C8 protospacers) sorted for nCas9-P2A-mCherry expression after electroporation in SH-SY5Y cells. (g) Orthogonality scores for the A7-C8 protospacer combination in all tested cell types. (h) Indel rates of MOBE3 with A7-C8 protospacer combination in all tested cell types. All values represent the mean and SEM of n = 3 or 4 independent biological replicates (shown individually).

Extended Data Fig. 10 MOBEs enable the installation of orthogonal point mutations known to cause isolated anencephaly (OLI736).

(a) Schematic of the DNA and protein sequences of the OLI736 combination, CELSR2 c.3800A > G and INVS c.725C > T, with MOBE protospacer and PAMs indicated. (b) On-target editing efficiencies of gRNA pair optimization with MOBE1 and MOBE3 in HEK293T cells. (c) Editing efficiencies of CELSR2 A6 protospacer combined with INVS C3 and C5 protospacers with inset (D) to display crosstalk in HEK293T cells. (e) Editing efficiencies of MOBE3 with the optimal A6-C5 protospacer combination sorted for nCas9-P2A-mCherry expression after electroporation in SH-SY5Y cells. (f) Orthogonality scores for the A6-C5 protospacer combination in both cell types. (g) Indel rates of MOBE3 with A6-C5 protospacer combination in both cell types. All values represent the mean and SEM of n = 3 independent biological replicates (shown individually).

Supplementary information

Supplementary Information

Supplementary Figs. 1–16 and Note 1.

Reporting Summary

Supplementary Data 1

Sequences of constructs, aptamers, protospacers and primers used.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cowan, Q.T., Gu, S., Gu, W. et al. Development of multiplexed orthogonal base editor (MOBE) systems. Nat Biotechnol (2024). https://doi.org/10.1038/s41587-024-02240-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-024-02240-0

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