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:

Targeted genome-modification tools and their advanced applications in crop breeding

Abstract

Crop improvement by genome editing involves the targeted alteration of genes to improve plant traits, such as stress tolerance, disease resistance or nutritional content. Techniques for the targeted modification of genomes have evolved from generating random mutations to precise base substitutions, followed by insertions, substitutions and deletions of small DNA fragments, and are finally starting to achieve precision manipulation of large DNA segments. Recent developments in base editing, prime editing and other CRISPR-associated systems have laid a solid technological foundation to enable plant basic research and precise molecular breeding. In this Review, we systematically outline the technological principles underlying precise and targeted genome-modification methods. We also review methods for the delivery of genome-editing reagents in plants and outline emerging crop-breeding strategies based on targeted genome modification. Finally, we consider potential future developments in precise genome-editing technologies, delivery methods and crop-breeding approaches, as well as regulatory policies for genome-editing products.

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: Precise DNA editing at the base pair level.
Fig. 2: Precise editing of large DNA segments.
Fig. 3: Delivery technologies used in plant targeted genome modification.
Fig. 4: Advanced applications of TGM in crop breeding.
Fig. 5: Prospects for TGM technologies in crop breeding.

Similar content being viewed by others

References

  1. Tilman, D., Balzer, C., Hill, J. & Befort, B. L. Global food demand and the sustainable intensification of agriculture. Proc. Natl Acad. Sci. USA 108, 20260–20264 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Meyer, R. S. & Purugganan, M. D. Evolution of crop species: genetics of domestication and diversification. Nat. Rev. Genet. 14, 840–852 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Holme, I. B., Gregersen, P. L. & Brinch-Pedersen, H. Induced genetic variation in crop plants by random or targeted mutagenesis: convergence and differences. Front. Plant. Sci. 10, 1468 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Chen, K., Wang, Y., Zhang, R., Zhang, H. & Gao, C. CRISPR/Cas genome eediting and precision plant breeding in agriculture. Annu. Rev. Plant. Biol. 70, 667–697 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Jinek, M. et al. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gaj, T., Gersbach, C. A. & Barbas, C. F. III ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31, 397–405 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  8. Liu, G., Lin, Q., Jin, S. & Gao, C. The CRISPR-Cas toolbox and gene editing technologies. Mol. Cell 82, 333–347 (2022).

    Article  CAS  PubMed  Google Scholar 

  9. Liu, Z. H. et al. Precise editing of methylated cytosine in Arabidopsis thaliana using a human APOBEC3Bctd–Cas9 fusion. Sci. China Life Sci. 65, 219–222 (2022).

    Article  CAS  PubMed  Google Scholar 

  10. Tang, S. et al. Targeted DNA demethylation produces heritable epialleles in rice. Sci. China Life Sci. 65, 753–756 (2022).

    Article  CAS  PubMed  Google Scholar 

  11. Gao, C. Genome engineering for crop improvement and future agriculture. Cell 184, 1621–1635 (2021).

    Article  CAS  PubMed  Google Scholar 

  12. Rao, Y., Yang, X., Pan, C., Wang, C. & Wang, K. Advance of clustered regularly interspaced short palindromic repeats–Cas9 system and its application in crop improvement. Front. Plant. Sci. 13, 839001 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Chen, Z., Debernardi, J. M., Dubcovsky, J. & Gallavotti, A. Recent advances in crop transformation technologies. Nat. Plants 8, 1343–1351 (2022).

    Article  CAS  PubMed  Google Scholar 

  14. Altpeter, F. et al. Advancing crop transformation in the era of genome editing. Plant. Cell 28, 1510–1520 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Fauser, F., Schiml, S. & Puchta, H. Both CRISPR/Cas-based nucleases and nickases can be used efficiently for genome engineering in Arabidopsis thaliana. Plant. J. 79, 348–359 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Chu, V. T. et al. Increasing the efficiency of homology-directed repair for CRISPR–Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 33, 543–548 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. Alanis-Lobato, G. et al. Frequent loss of heterozygosity in CRISPR–Cas9-edited early human embryos. Proc. Natl Acad. Sci. USA 118, e2004832117 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Shan, Q. et al. Targeted genome modification of crop plants using a CRISPR–Cas system. Nat. Biotechnol. 31, 686–688 (2013). This study marks the first application of CRISPR–Cas9 technology in plants, generating mutants with the desired edits.

    Article  CAS  PubMed  Google Scholar 

  20. 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  PubMed  PubMed Central  Google Scholar 

  21. 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 work combined deaminase with CRISPR for the first time to achieve precise cytosine editing without double-strand breaks at the single-base level.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, 6305 (2016).

    Article  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  24. Ren, B. et al. A CRISPR/Cas9 toolkit for efficient targeted base editing to induce genetic variations in rice. Sci. China Life Sci. 60, 516–519 (2017).

    Article  PubMed  Google Scholar 

  25. Zong, Y. et al. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A. Nat. Biotechnol. 36, 950–953 (2018).

    Article  CAS  Google Scholar 

  26. Jin, S. et al. Rationally designed APOBEC3B cytosine base editors with improved specificity. Mol. Cell 79, 728–740 e726 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Liang, Y., Chen, F., Wang, K. & Lai, L. Base editors: development and applications in biomedicine. Front. Med. 17, 359–387 (2023).

    Article  PubMed  Google Scholar 

  28. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Huang, J. et al. Discovery of deaminase functions by structure-based protein clustering. Cell 186, 3182–3195.e3114 (2023). This work used artificial-intelligence-assisted structural clustering methods for mining novel enzymes suitable for genome editing in eukaryote.

    Article  CAS  PubMed  Google Scholar 

  30. Cortizas, E. M. et al. UNG protects B cells from AID-induced telomere loss. J. Exp. Med. 213, 2459–2472 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kurt, I. C. et al. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat. Biotechnol. 39, 41–46 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Zhao, D. et al. Glycosylase base editors enable C-to-A and C-to-G base changes. Nat. Biotechnol. 39, 35–40 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Sretenovic, S. et al. Exploring C-To-G base editing in rice, tomato, and poplar. Front. Genome Edit. 3, 756766 (2021).

    Article  Google Scholar 

  34. 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 work created the first adenine base editor through extensive directed evolution and protein engineering.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  37. 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 

  38. Wei, C. et al. Efficient generation of homozygous substitutions in rice in one generation utilizing an rABE8e base editor. J. Integr. Plant. Biol. 63, 1595–1599 (2021).

    Article  CAS  PubMed  Google Scholar 

  39. Tong, H. et al. Programmable A-to-Y base editing by fusing an adenine base editor with an N-methylpurine DNA glycosylase. Nat. Biotechnol. 41, 1080–1084 (2023).

    Article  CAS  PubMed  Google Scholar 

  40. Chen, L. et al. Adenine transversion editors enable precise, efficient A*T-to-C*G base editing in mammalian cells and embryos. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01821-9 (2023).

  41. Wu, X. et al. Adenine base editor incorporating the N-methylpurine DNA glycosylase MPGv3 enables efficient A-to-K base editing in rice. Plant. Commun. 4, 100668 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tong, H. et al. Programmable deaminase-free base editors for G-to-Y conversion by engineered glycosylase. Natl Sci. Rev. 10, nwad143 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  43. He, Y. et al. Protein language models-assisted optimization of a uracil-N-glycosylase variant enables programmable T-to-G and T-to-C base editing. Mol. Cell 84, 1257–1270.e6 (2024).

    Article  CAS  PubMed  Google Scholar 

  44. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019). This work described the development of prime editing and precisely achieved all types of base substitutions and small insertions or deletions without double-strand breaks in eukaryotes for the first time.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zong, Y. et al. An engineered prime editor with enhanced editing efficiency in plants. Nat. Biotechnol. 40, 1394–1402 (2022). This work greatly increased the efficiency of prime editing in plants through protein engineering.

    Article  CAS  PubMed  Google Scholar 

  46. Ni, P. et al. Efficient and versatile multiplex prime editing in hexaploid wheat. Genome Biol. 24, 156 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Doman, J. L. et al. Phage-assisted evolution and protein engineering yield compact, efficient prime editors. Cell 186, 3983–4002.e3926 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ponnienselvan, K. et al. Addressing the dNTP bottleneck restricting prime editing activity. Preprint at bioRxiv https://doi.org/10.1101/2023.10.21.563443 (2023).

  49. Chen, P. J. et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 184, 5635–5652.e5629 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu, P. et al. Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nat. Commun. 12, 2121 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  52. Zhang, G. et al. Enhancement of prime editing via xrRNA motif-joined pegRNA. Nat. Commun. 13, 1856 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Li, X. et al. Enhancing prime editing efficiency by modified pegRNA with RNA G-quadruplexes. J. Mol. Cell Biol. 14, mjac022 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Li, X. et al. Highly efficient prime editing by introducing same-sense mutations in pegRNA or stabilizing its structure. Nat. Commun. 13, 1669 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Xu, W. et al. A design optimized prime editor with expanded scope and capability in plants. Nat. Plants 8, 45–52 (2022).

    Article  CAS  PubMed  Google Scholar 

  56. Yu, G. et al. Prediction of efficiencies for diverse prime editing systems in multiple cell types. Cell 186, 2256–2272.e2223 (2023).

    Article  CAS  PubMed  Google Scholar 

  57. Jiang, Y. et al. Optimized prime editing efficiently generates glyphosate-resistant rice plants carrying homozygous TAP–IVS mutation in EPSPS. Mol. Plant. 15, 1646–1649 (2022).

    Article  CAS  PubMed  Google Scholar 

  58. Liu, B. et al. Targeted genome editing with a DNA-dependent DNA polymerase and exogenous DNA-containing templates. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01947-w (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  59. da Silva, J. F. et al. Click editing enables programmable genome writing using DNA polymerases and HUH endonucleases. Preprint at bioRxiv https://doi.org/10.1101/2023.09.12.557440 (2023).

  60. Wang, S. et al. Precise, predictable multi-nucleotide deletions in rice and wheat using APOBEC–Cas9. Nat. Biotechnol. 38, 1460–1465 (2020).

    Article  CAS  PubMed  Google Scholar 

  61. Dong, O. X. et al. Marker-free carotenoid-enriched rice generated through targeted gene insertion using CRISPR–Cas9. Nat. Commun. 11, 1178 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gisler, B., Salomon, S. & Puchta, H. The role of double‐strand-break‐induced allelic homologous recombination in somatic plant cells. Plant. J. 32, 277–284 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Rönspies, M. et al. Massive crossover suppression by CRISPR–Cas-mediated plant chromosome engineering. Nat. Plants 8, 1153–1159 (2022).

    Article  PubMed  Google Scholar 

  64. Beying, N., Schmidt, C., Pacher, M., Houben, A. & Puchta, H. CRISPR–Cas9-mediated induction of heritable chromosomal translocations in Arabidopsis. Nat. Plants 6, 638–645 (2020).

    Article  CAS  PubMed  Google Scholar 

  65. Wang, J. et al. Efficient targeted insertion of large DNA fragments without DNA donors. Nat. Methods 19, 331–340 (2022).

    Article  CAS  PubMed  Google Scholar 

  66. Sun, C. et al. Precise integration of large DNA sequences in plant genomes using PrimeRoot editors. Nat. Biotechnol. 42, 316–327 (2023). This work achieved double-strand-break-independent precise targeted insertion of large DNA segments in plants.

    Article  PubMed  Google Scholar 

  67. Choi, J. et al. Precise genomic deletions using paired prime editing. Nat. Biotechnol. 40, 218–226 (2022).

    Article  CAS  PubMed  Google Scholar 

  68. Jiang, T., Zhang, X. O., Weng, Z. & Xue, W. Deletion and replacement of long genomic sequences using prime editing. Nat. Biotechnol. 40, 227–234 (2022).

    Article  CAS  PubMed  Google Scholar 

  69. Tansirichaiya, S., Rahman, M. A. & Roberts, A. P. The transposon registry. Mob. DNA 10, 40 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  70. O’Donnell, K. A. Advances in functional genetic screening with transposons and CRISPR/Cas9 to illuminate cancer biology. Curr. Opin. Genet. Dev. 49, 85–94 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Liu, P. et al. CRISPR-targeted transposable element insertion for efficient plant genome engineering. Preprint at Research Square https://doi.org/10.21203/rs.3.rs-2679086/v1 (2023).

  72. Pallares-Masmitja, M. et al. Find and cut-and-transfer (FiCAT) mammalian genome engineering. Nat. Commun. 12, 7071 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Klompe, S. E., Vo, P. L. H., Halpin-Healy, T. S. & Sternberg, S. H. Transposon-encoded CRISPR–Cas systems direct RNA-guided DNA integration. Nature 571, 219–225 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Strecker, J. et al. RNA-guided DNA insertion with CRISPR-associated transposases. Science 365, 48–53 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tou, C. J., Orr, B. & Kleinstiver, B. P. Precise cut-and-paste DNA insertion using engineered type V-K CRISPR-associated transposases. Nat. Biotechnol. 41, 968–979 (2023).

    Article  CAS  PubMed  Google Scholar 

  76. Lampe, G. D. et al. Targeted DNA integration in human cells without double-strand breaks using CRISPR-associated transposases. Nat. Biotechnol. 42, 87–98 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Wilkinson, M. E., Frangieh, C. J., Macrae, R. K. & Zhang, F. Structure of the R2 non-LTR retrotransposon initiating target-primed reverse transcription. Science 380, 301–308 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Hirano, N., Muroi, T., Takahashi, H. & Haruki, M. Site-specific recombinases as tools for heterologous gene integration. Appl. Microbiol. Biotechnol. 92, 227–239 (2011).

    Article  CAS  PubMed  Google Scholar 

  79. Anzalone, A. V. et al. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat. Biotechnol. 40, 731–740 (2022).

    Article  CAS  PubMed  Google Scholar 

  80. Yarnall, M. T. N. et al. Drag-and-drop genome insertion of large sequences without double-strand DNA cleavage using CRISPR-directed integrases. Nat. Biotechnol. 41, 500–512 (2023).

    Article  CAS  PubMed  Google Scholar 

  81. Wang, C. et al. dCas9-based gene editing for cleavage-free genomic knock-in of long sequences. Nat. Cell Biol. 24, 268–278 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Nasti, R. A. & Voytas, D. F. Attaining the promise of plant gene editing at scale. Proc. Natl Acad. Sci. USA 118, e2004846117 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ghogare, R., Ludwig, Y., Bueno, G. M., Slamet-Loedin, I. H. & Dhingra, A. Genome editing reagent delivery in plants. Transgen. Res. 30, 321–335 (2021).

    Article  CAS  Google Scholar 

  84. Svitashev, S., Schwartz, C., Lenderts, B., Young, J. K. & Mark Cigan, A. Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat. Commun. 7, 13274 (2016). This work provides a method of efficient transient genome editing using ribonucleoprotein complexes through particle bombardment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Liang, Z. et al. Efficient DNA-free genome editing of bread wheat using CRISPR/Cas9 ribonucleoprotein complexes. Nat. Commun. 8, 14261 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Liang, Z. et al. Genome editing of bread wheat using biolistic delivery of CRISPR/Cas9 in vitro transcripts or ribonucleoproteins. Nat. Protoc. 13, 413–430 (2018).

    Article  CAS  PubMed  Google Scholar 

  87. Zhang, Y. et al. Efficient and transgene-free genome editing in wheat through transient expression of CRISPR/Cas9 DNA or RNA. Nat. Commun. 7, 12617 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Qiu, F. et al. Transient expression of a TaGRF4–TaGIF1 complex stimulates wheat regeneration and improves genome editing. Sci. China Life Sci. 65, 731–738 (2022).

    Article  CAS  PubMed  Google Scholar 

  89. Lowe, K. et al. Morphogenic regulators Baby boom and Wuschel improve monocot transformation. Plant. Cell 28, 1998–2015 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Liu, J. et al. Genome-scale sequence disruption following biolistic transformation in rice and maize. Plant. Cell 31, 368–383 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Pan, C. et al. Boosting plant genome editing with a versatile CRISPR–Combo system. Nat. Plants 8, 513–525 (2022).

    Article  CAS  PubMed  Google Scholar 

  92. Debernardi, J. M. et al. A GRF–GIF chimeric protein improves the regeneration efficiency of transgenic plants. Nat. Biotechnol. 38, 1274–1279 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Maher, M. F. et al. Plant gene editing through de novo induction of meristems. Nat. Biotechnol. 38, 84–89 (2020). This work expressed developmental regulators and targeted genome-modification reagents, leading to de novo induction of meristematic tissues and edited plants obtained without the need for tissue culture.

    Article  CAS  PubMed  Google Scholar 

  94. Oh, Y., Kim, H. & Kim, S. G. Virus-induced plant genome editing. Curr. Opin. Plant. Biol. 60, 101992 (2021).

    Article  CAS  PubMed  Google Scholar 

  95. Ellison, E. E. et al. Multiplexed heritable gene editing using RNA viruses and mobile single guide RNAs. Nat. Plants 6, 620–624 (2020). This study used an RNA virus to express mobile single guide RNA, enabling efficient multiplex genome editing in plants without the need for tissue culture.

    Article  CAS  PubMed  Google Scholar 

  96. Čermák, T. et al. A multipurpose toolkit to enable advanced genome engineering in plants. Plant. Cell 29, 1196–1217 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Li, T. et al. Highly efficient heritable genome editing in wheat using an RNA virus and bypassing tissue culture. Mol. Plant. 14, 1787–1798 (2021).

    Article  CAS  PubMed  Google Scholar 

  98. Ma, X., Zhang, X., Liu, H. & Li, Z. Highly efficient DNA-free plant genome editing using virally delivered CRISPR–Cas9. Nat. Plants 6, 773–779 (2020).

    Article  CAS  PubMed  Google Scholar 

  99. Liu, Q., Zhao, C., Sun, K., Deng, Y. & Li, Z. Engineered biocontainable RNA virus vectors for non-transgenic genome editing across crop species and genotypes. Mol. Plant. 16, 616–631 (2023).

    Article  CAS  PubMed  Google Scholar 

  100. Park, S. Y., Shimizu, K., Brown, J., Aoki, K. & Westwood, J. H. Mobile host mRNAs are translated to protein in the associated parasitic plant Cuscuta campestris. Plants 11, 93 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  101. Zhang, W. et al. tRNA-related sequences trigger systemic mRNA transport in plants. Plant Cell 28, 1237–1249 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Jackson, S. D. & Hong, Y. Systemic movement of FT mRNA and a possible role in floral induction. Front. Plant. Sci. 3, 127 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Yang, L., Machin, F., Wang, S., Saplaoura, E. & Kragler, F. Heritable transgene-free genome editing in plants by grafting of wild-type shoots to transgenic donor rootstocks. Nat. Biotechnol. 41, 958–967 (2023). This work reported the first transgene-free and tissue-culture-free plant delivery by grafting, achieving heritable targeted mutagenesis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Woo, J. W. et al. DNA-free genome editing in plants with preassembled CRISPR–Cas9 ribonucleoproteins. Nat. Biotechnol. 33, 1162–1164 (2015).

    Article  CAS  PubMed  Google Scholar 

  105. Andersson, M. et al. Genome editing in potato via CRISPR–Cas9 ribonucleoprotein delivery. Physiol. Plant. 164, 378–384 (2018).

    Article  CAS  PubMed  Google Scholar 

  106. Liu, Y. et al. Establishment of a DNA-free genome editing and protoplast regeneration method in cultivated tomato (Solanum lycopersicum). Plant. Cell Rep. 41, 1843–1852 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Cao, X. et al. Cut-dip-budding delivery system enables genetic modifications in plants without tissue culture. Innovation 4, 100345 (2023).

    PubMed  Google Scholar 

  108. Raman, V. et al. Agrobacterium expressing a type III secretion system delivers Pseudomonas effectors into plant cells to enhance transformation. Nat. Commun. 13, 2581 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Soliman, A., Laurie, J., Bilichak, A. & Ziemienowicz, A. Applications of CPPs in genome editing of plants. Methods Mol. Biol. 2383, 595–616 (2022).

    Article  CAS  PubMed  Google Scholar 

  110. Kwak, S. Y. et al. Chloroplast-selective gene delivery and expression in planta using chitosan-complexed single-walled carbon nanotube carriers. Nat. Nanotechnol. 14, 447–455 (2019).

    Article  CAS  PubMed  Google Scholar 

  111. Wang, Z. P. et al. Efficient and genotype independent maize transformation using pollen transfected by DNA-coated magnetic nanoparticles. J. Integr. Plant. Biol. 64, 1145–1156 (2022).

    Article  CAS  PubMed  Google Scholar 

  112. Ran, Y., Liang, Z. & Gao, C. Current and future editing reagent delivery systems for plant genome editing. Sci. China Life Sci. 60, 490–505 (2017).

    Article  CAS  PubMed  Google Scholar 

  113. Li, C. et al. Targeted, random mutagenesis of plant genes with dual cytosine and adenine base editors. Nat. Biotechnol. 38, 875–882 (2020). This work generated dual cytosine and adenine base editors as mutagenesis editors that achieved near-saturated mutagenesis and facilitated directed evolution of plant endogenous genes.

    Article  CAS  PubMed  Google Scholar 

  114. Kuang, Y. et al. Base-editing-mediated artificial evolution of OsALS1 in planta to develop novel herbicide-tolerant rice germplasms. Mol. Plant. 13, 565–572 (2020).

    Article  CAS  PubMed  Google Scholar 

  115. Zhang, A. et al. Directed evolution rice genes with randomly multiplexed sgRNAs assembly of base editors. Plant Biotechnol. J. 21, 2597–2610 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zhang, C. et al. Artificial evolution of OsEPSPS through an improved dual cytosine and adenine base editor generated a novel allele conferring rice glyphosate tolerance. J. Integr. Plant. Biol. 65, 2194–2203 (2023).

    Article  CAS  PubMed  Google Scholar 

  117. Xu, R., Liu, X., Li, J., Qin, R. & Wei, P. Identification of herbicide resistance OsACC1 mutations via in planta prime-editing-library screening in rice. Nat. Plants 7, 888–892 (2021).

    Article  CAS  PubMed  Google Scholar 

  118. Romero, I. G., Ruvinsky, I. & Gilad, Y. Comparative studies of gene expression and the evolution of gene regulation. Nat. Rev. Genet. 13, 505–516 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Liu, L. et al. Enhancing grain-yield-related traits by CRISPR–Cas9 promoter editing of maize CLE genes. Nat. Plants 7, 287–294 (2021).

    Article  CAS  PubMed  Google Scholar 

  120. Cui, Y., Cao, Q., Li, Y., He, M. & Liu, X. Advances in cis-element- and natural variation-mediated transcriptional regulation and applications in gene editing of major crops. J. Exp. Bot. 74, 5441–5457 (2023).

    Article  CAS  PubMed  Google Scholar 

  121. Zhou, J. et al. An efficient CRISPR–Cas12a promoter editing system for crop improvement. Nat. Plants 9, 588–604 (2023).

    Article  CAS  PubMed  Google Scholar 

  122. Rodriguez-Leal, D., Lemmon, Z. H., Man, J., Bartlett, M. E. & Lippman, Z. B. Engineering quantitative trait variation for crop improvement by genome editing. Cell 171, 470–480.e478 (2017). This work demonstrates that targeting cis-regulatory elements in promoter regions can create a continuum of variation in genes for improving crop traits.

    Article  CAS  PubMed  Google Scholar 

  123. Aguirre, L., Hendelman, A., Hutton, S. F., McCandlish, D. M. & Lippman, Z. B. Idiosyncratic and dose-dependent epistasis drives variation in tomato fruit size. Science 382, 315–320 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Song, X. et al. Targeting a gene regulatory element enhances rice grain yield by decoupling panicle number and size. Nat. Biotechnol. 40, 1403–1411 (2022).

    Article  CAS  PubMed  Google Scholar 

  125. Wang, J., Liu, J. & Guo, Z. Natural uORF variation in plants. Trends Plant Sci. https://doi.org/10.1016/j.tplants.2023.07.005 (2023).

  126. Zhang, H. et al. Genome editing of upstream open reading frames enables translational control in plants. Nat. Biotechnol. 36, 894–898 (2018). This study demonstrates a strategy that manipulates crop quantitative traits by fine-tuning protein expression through the modification of upstream open reading frames.

    Article  CAS  PubMed  Google Scholar 

  127. Xing, S. et al. Fine-tuning sugar content in strawberry. Genome Biol. 21, 230 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Xue, C. et al. Tuning plant phenotypes by precise, graded downregulation of gene expression. Nat. Biotechnol. 41, 1758–1764 (2023).

    Article  CAS  PubMed  Google Scholar 

  129. Wang, H. et al. Genome editing of 3′ UTR-embedded inhibitory region enables generation of gene knock-up alleles in plants. Plant. Commun. https://doi.org/10.1016/j.xplc.2023.100745 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Meng, F. et al. Genomic editing of intronic enhancers unveils their role in fine-tuning tissue-specific gene expression in Arabidopsis thaliana. Plant. Cell 33, 1997–2014 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Guo, N. et al. Genome sequencing sheds light on the contribution of structural variants to Brassica oleracea diversification. BMC Biol. 19, 93 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Alonge, M. et al. Major impacts of widespread structural variation on gene expression and crop improvement in tomato. Cell 182, 145–161.e123 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Lu, Y. et al. A donor-DNA-free CRISPR/Cas-based approach to gene knock-up in rice. Nat. Plants 7, 1445–1452 (2021).

    Article  CAS  PubMed  Google Scholar 

  134. Li, S. et al. Genome-edited powdery mildew resistance in wheat without growth penalties. Nature 602, 455–460 (2022). This study utilized multiplexed genome editing to improve complex polyploid crops, while achieving a dual benefit of high yield and disease resistance.

    Article  CAS  PubMed  Google Scholar 

  135. Wang, Y. et al. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat. Biotechnol. 32, 947–951 (2014).

    Article  CAS  PubMed  Google Scholar 

  136. Zhou, Y. et al. Graph pangenome captures missing heritability and empowers tomato breeding. Nature 606, 527–534 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Zhang, J., Yu, H. & Li, J. De novo domestication: retrace the history of agriculture to design future crops. Curr. Opin. Biotechnol. 81, 102946 (2023).

    Article  CAS  PubMed  Google Scholar 

  138. Li, T. et al. Domestication of wild tomato is accelerated by genome editing. Nat. Biotechnol. 36, 1160–1163 (2018).

    Article  CAS  Google Scholar 

  139. Yu, H. et al. A route to de novo domestication of wild allotetraploid rice. Cell 184, 1156–1170.e1114 (2021).

    Article  CAS  PubMed  Google Scholar 

  140. Lemmon, Z. H. et al. Rapid improvement of domestication traits in an orphan crop by genome editing. Nat. Plants 4, 766–770 (2018).

    Article  CAS  PubMed  Google Scholar 

  141. Zsögön, A. et al. De novo domestication of wild tomato using genome editing. Nat. Biotechnol. 36, 1211–1216 (2018). This work is the first to demonstrate that genome editing can rapidly generate novel crops by de novo domestication of wild plant species.

    Article  Google Scholar 

  142. Smýkal, P., Nelson, M., Berger, J. & Von Wettberg, E. The impact of genetic changes during crop domestication. Agronomy 8, 26 (2018).

    Article  Google Scholar 

  143. Huang, X., Huang, S., Han, B. & Li, J. The integrated genomics of crop domestication and breeding. Cell 185, 2828–2839 (2022).

    Article  CAS  PubMed  Google Scholar 

  144. DeHaan, L. et al. Roadmap for accelerated domestication of an emerging perennial grain crop. Trends Plant. Sci. 25, 525–537 (2020).

    Article  CAS  PubMed  Google Scholar 

  145. Okada, A. et al. CRISPR/Cas9-mediated knockout of Ms1 enables the rapid generation of male-sterile hexaploid wheat lines for use in hybrid seed production. Plant Biotechnol. J. 17, 1905–1913 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Singh, M., Kumar, M., Albertsen, M. C., Young, J. K. & Cigan, A. M. Concurrent modifications in the three homeologs of Ms45 gene with CRISPR–Cas9 lead to rapid generation of male sterile bread wheat (Triticum aestivum L.). Plant. Mol. Biol. 97, 371–383 (2018).

    Article  CAS  PubMed  Google Scholar 

  147. Chen, G. et al. Gene editing to facilitate hybrid crop production. Biotechnol. Adv. 46, 107676 (2021).

    Article  CAS  PubMed  Google Scholar 

  148. Vernet, A. et al. High-frequency synthetic apomixis in hybrid rice. Nat. Commun. 13, 7963 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Marimuthu, M. P. et al. Synthetic clonal reproduction through seeds. Science 331, 876 (2011).

    Article  CAS  PubMed  Google Scholar 

  150. Wang, C. et al. Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat. Biotechnol. 37, 283–286 (2019).

    Article  CAS  PubMed  Google Scholar 

  151. Wei, X. et al. Synthetic apomixis with normal hybrid rice seed production. Mol. Plant. 16, 489–492 (2023).

    Article  CAS  PubMed  Google Scholar 

  152. Khanday, I., Skinner, D., Yang, B., Mercier, R. & Sundaresan, V. A male-expressed rice embryogenic trigger redirected for asexual propagation through seeds. Nature 565, 91–95 (2019). This work achieved apomixis in rice with the assistance of targeted genome modification, which can be utilized to fix hybrid vigour.

    Article  CAS  PubMed  Google Scholar 

  153. Song, M. et al. Simultaneous production of high-frequency synthetic apomixis with high fertility and improved agronomic traits in hybrid rice. Mol. Plant. 17, 4–7 (2024).

    Article  CAS  PubMed  Google Scholar 

  154. Kelliher, T. et al. MATRILINEAL, a sperm-specific phospholipase, triggers maize haploid induction. Nature 542, 105–109 (2017).

    Article  CAS  PubMed  Google Scholar 

  155. Liu, C. et al. A 4-bp insertion at ZmPLA1 encoding a putative phospholipase A generates haploid induction in maize. Mol. Plant. 10, 520–522 (2017).

    Article  CAS  PubMed  Google Scholar 

  156. Lv, J. et al. Generation of paternal haploids in wheat by genome editing of the centromeric histone CENH3. Nat. Biotechnol. 38, 1397–1401 (2020).

    Article  CAS  PubMed  Google Scholar 

  157. Li, Y. et al. Loss-of-function alleles of ZmPLD3 cause haploid induction in maize. Nat. Plants 7, 1579–1588 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Zhong, Y. et al. Mutation of ZmDMP enhances haploid induction in maize. Nat. Plants 5, 575–580 (2019).

    Article  PubMed  Google Scholar 

  159. Jiang, C. et al. A reactive oxygen species burst causes haploid induction in maize. Mol. Plant. 15, 943–955 (2022).

    Article  CAS  PubMed  Google Scholar 

  160. Kelliher, T. et al. One-step genome editing of elite crop germplasm during haploid induction. Nat. Biotechnol. 37, 287–292 (2019). This work integrated haploid induction technology with genome editing, and generated the HI-Edit system, thus achieving rapid improvement of a wide range of crop varieties.

    Article  CAS  PubMed  Google Scholar 

  161. Wang, B. et al. Development of a haploid-inducer-mediated genome-editing system for accelerating maize breeding. Mol. Plant. 12, 597–602 (2019).

    Article  PubMed  Google Scholar 

  162. Ye, M. et al. Generation of self-compatible diploid potato by knockout of S-RNase. Nat. Plants 4, 651–654 (2018).

    Article  CAS  PubMed  Google Scholar 

  163. Ma, C. et al. CRISPR/Cas9-mediated multiple gene editing in Brassica oleracea var. capitata using the endogenous tRNA-processing system. Hortic. Res. 6, 20 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Srivastava, V. & Thomson, J. Gene stacking by recombinases. Plant Biotechnol. J. 14, 471–482 (2016).

    Article  CAS  PubMed  Google Scholar 

  165. Zhu, Q. et al. Development of ‘purple endosperm rice’ by engineering anthocyanin biosynthesis in the endosperm with a high-efficiency transgene stacking system. Mol. Plant. 10, 918–929 (2017).

    Article  CAS  PubMed  Google Scholar 

  166. Shehryar, K. et al. Transgene stacking as effective tool for enhanced disease resistance in plants. Mol. Biotechnol. 62, 1–7 (2020).

    Article  CAS  PubMed  Google Scholar 

  167. Glasscock, C. J. et al. Computational design of sequence-specific DNA-binding proteins. Preprint at bioRxiv https://doi.org/10.1101/2023.09.20.558720 (2023).

  168. Watson, J. L. et al. De novo design of protein structure and function with RFdiffusion. Nature 620, 1089–1100 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Xu, C. et al. Computational design of transmembrane pores. Nature 585, 129–134 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Renard, D. & Tilman, D. National food production stabilized by crop diversity. Nature 571, 257–260 (2019).

    Article  CAS  PubMed  Google Scholar 

  171. Shalev, O. et al. Commensal Pseudomonas strains facilitate protective response against pathogens in the host plant. Nat. Ecol. Evol. 6, 383–396 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  172. Xu, S. et al. Fusarium fruiting body microbiome member Pantoea agglomerans inhibits fungal pathogenesis by targeting lipid rafts. Nat. Microbiol. 7, 831–843 (2022).

    Article  CAS  PubMed  Google Scholar 

  173. Jiang, D. et al. Highly efficient genome editing in Xanthomonas oryzae pv. oryzae through repurposing the endogenous type I‐C CRISPR–Cas system. Mol. Plant. Pathol. 23, 583–594 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  174. Zhang, X.-E. et al. Enabling technology and core theory of synthetic biology. Sci. China Life Sci. 66, 1742–1785 (2023).

    Article  PubMed  Google Scholar 

  175. Vora, Z., Pandya, J., Sangh, C. & Vaikuntapu, P. R. The evolving landscape of global regulations on genome-edited crops. J. Plant. Biochem. Biotechnol. 32, 831–845 (2023).

    Article  CAS  Google Scholar 

  176. Ahmad, A., Jamil, A. & Munawar, N. GMOs or non-GMOs? The CRISPR conundrum. Front. Plant. Sci. 14, 1232938 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  177. Lu, Y. & Zhu, J.-K. Precise editing of a target base in the rice genome using a modified CRISPR/Cas9 system. Mol. Plant. 10, 523–525 (2017).

    Article  CAS  PubMed  Google Scholar 

  178. Chen, Y. et al. CRISPR/Cas9-mediated base-editing system efficiently generates gain-of-function mutations in Arabidopsis. Sci. China Life Sci. 60, 520–523 (2017).

    Article  CAS  PubMed  Google Scholar 

  179. Tian, S. et al. Engineering herbicide-resistant watermelon variety through CRISPR/Cas9-mediated base-editing. Plant. Cell Rep. 37, 1353–1356 (2018).

    Article  CAS  PubMed  Google Scholar 

  180. Qin, L. et al. High‐efficient and precise base editing of C•G to T•A in the allotetraploid cotton (Gossypium hirsutum) genome using a modified CRISPR/Cas9 system. Plant Biotechnol. J. 18, 45–56 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  181. Li, J., Sun, Y., Du, J., Zhao, Y. & Xia, L. Generation of targeted point mutations in rice by a modified CRISPR/Cas9 System. Mol. Plant. 10, 526–529 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  183. Ren, B. et al. Improved base editor for efficiently inducing genetic variations in rice with CRISPR/Cas9-guided hyperactive hAID mutant. Mol. Plant. 11, 623–626 (2018).

    Article  CAS  PubMed  Google Scholar 

  184. Wang, M. et al. Optimizing base editors for improved efficiency and expanded editing scope in rice. Plant Biotechnol. J. 17, 1697–1699 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  185. Hua, K., Tao, X. & Zhu, J. K. Expanding the base editing scope in rice by using Cas9 variants. Plant Biotechnol. J. 17, 499–504 (2019).

    Article  PubMed  Google Scholar 

  186. Ren, B. et al. Cas9–NG greatly expands the targeting scope of the genome-editing toolkit by recognizing NG and other atypical PAMs in rice. Mol. Plant. 12, 1015–1026 (2019).

    Article  CAS  PubMed  Google Scholar 

  187. Veillet, F. et al. The Solanum tuberosum GBSSI gene: a target for assessing gene and base editing in tetraploid potato. Plant. Cell Rep. 38, 1065–1080 (2019).

    Article  CAS  PubMed  Google Scholar 

  188. Veillet, F. et al. Transgene-free genome editing in tomato and potato plants using Agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. Int. J. Mol. Sci. 20, 402 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  189. Wu, J. et al. Engineering herbicide‐resistant oilseed rape by CRISPR/Cas9‐mediated cytosine base‐editing. Plant Biotechnol. J. 18, 1857–1859 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Wang, M. et al. Targeted base editing in rice with CRISPR/ScCas9 system. Plant Biotechnol. J. 18, 1645–164 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  191. Zhang, C. et al. Expanding the base editing scope to GA and relaxed NG PAM sites by improved xCas9 system. Plant Biotechnol. J. 18, 884–886 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  192. Zeng, D. et al. PhieCBEs: plant high-efficiency cytidine base editors with expanded target range. Mol. Plant. 13, 1666–1669 (2020).

    Article  CAS  PubMed  Google Scholar 

  193. 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).

    Article  CAS  PubMed  Google Scholar 

  194. Hu, L. et al. Precise A∙T to G∙C base editing in the allotetraploid rapeseed (Brassica napus L.) genome. J. Cell Physiol. 237, 4544–4550 (2022).

    Article  CAS  PubMed  Google Scholar 

  195. Hua, K. et al. Simplified adenine base editors improve adenine base editing efficiency in rice. Plant Biotechnol. J. 18, 770–778 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Yan, D. et al. High-efficiency and multiplex adenine base editing in plants using new TadA variants. Mol. Plant. 14, 722–731 (2021).

    Article  CAS  PubMed  Google Scholar 

  197. Tan, J. et al. PhieABEs: a PAM‐less/free high‐efficiency adenine base editor toolbox with wide target scope in plants. Plant Biotechnol. J. 20, 934–943 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Xu, R. et al. Development of an efficient plant dual cytosine and adenine editor. J. Integr. Plant. Biol. 63, 1600–1605 (2021).

    Article  CAS  PubMed  Google Scholar 

  199. Tian, Y. et al. Efficient C‐to‐G editing in rice using an optimized base editor. Plant. Biotechnol. 20, 1238–1240 (2022).

    Article  CAS  Google Scholar 

  200. Cheng, Y. et al. CRISPR–Cas12a base editors confer efficient multiplexed genome editing in rice. Plant. Commun. 4, 100601 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Wang, D. et al. Developing a highly efficient CGBE base editor in watermelon. Hortic. Res. 10, uhad155 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  202. Li, Y. et al. Engineering a plant A-to-K base editor with improved performance by fusion with a transactivation module. Plant. Commun. 4, 100667 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Li, X. et al. Efficient and heritable A-to-K base editing in rice and tomato. Hortic. Res. 11, uhad250 (2024).

    Article  PubMed  Google Scholar 

  204. Zhong, D. et al. Targeted A‐to‐T and A‐to‐C base replacement in maize using an optimized adenine base editor. Plant Biotechnol. J. 22, 541–543 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  205. Lin, Q. et al. Prime genome editing in rice and wheat. Nat. Biotechnol. 38, 582–585 (2020).

    Article  CAS  PubMed  Google Scholar 

  206. Lin, Q. et al. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat. Biotechnol. 39, 923–927 (2021).

    Article  CAS  PubMed  Google Scholar 

  207. Butt, H. et al. Engineering herbicide resistance via prime editing in rice. Plant Biotechnol. J. 18, 2370–2372 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Hua, K., Jiang, Y., Tao, X. & Zhu, J. K. Precision genome engineering in rice using prime editing system. Plant Biotechnol. J. 18, 2167–2169 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  209. Xu, R. et al. Development of plant prime-editing systems for precise genome editing. Plant. Commun. 1, 100043 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  210. Jiang, Y.-Y. et al. Prime editing efficiently generates W542L and S621I double mutations in two ALS genes in maize. Genome Biol. 21, 257 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Lu, Y. et al. Precise genome modification in tomato using an improved prime editing system. Plant Biotechnol. J. 19, 415–417 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  212. Li, J. et al. Development of a highly efficient prime editor 2 system in plants. Genome Biol. 23, 161 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Li, H. et al. Multiplex precision gene editing by a surrogate prime editor in rice. Mol. Plant. 15, 1077–1080 (2022).

    Article  PubMed  Google Scholar 

  214. Zou, J. et al. Improving the efficiency of prime editing with epegRNAs and high-temperature treatment in rice. Sci. China Life Sci. 65, 2328–2331 (2022).

    Article  CAS  PubMed  Google Scholar 

  215. Liang, Z., Wu, Y., Guo, Y. & Wei, S. Addition of the T5 exonuclease increases the prime editing efficiency in plants. J. Genet. Genom. 50, 582–588 (2023).

    Article  Google Scholar 

  216. Gupta, A., Liu, B., Raza, S., Chen, Q.-J. & Yang, B. Modularly assembled multiplex prime editors for simultaneous editing of agronomically important genes in rice. Plant Commun. 5, 100471 (2023).

    Google Scholar 

  217. Gupta, A., Liu, B., Chen, Q. J. & Yang, B. High‐efficiency prime editing enables new strategies for broad‐spectrum resistance to bacterial blight of rice. Plant Biotechnol. J. 21, 1454–1464 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Liu, X. et al. Generating herbicide resistant and dwarf rice germplasms through precise sequence insertion or replacement. Plant Biotechnol. J. 22, 293–295 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  219. Wang, J. et al. Plant organellar genomes: much done, much more to do. Trends Plant. Sci. https://doi.org/10.1016/j.tplants.2023.12.014 (2024).

    Article  PubMed  Google Scholar 

  220. Kang, B.-C. et al. Chloroplast and mitochondrial DNA editing in plants. Nat. Plants 7, 899–905 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Kazama, T. et al. Curing cytoplasmic male sterility via TALEN-mediated mitochondrial genome editing. Nat. Plants 5, 722–730 (2019).

    Article  CAS  PubMed  Google Scholar 

  222. Kuwabara, K., Arimura, S.-i, Shirasawa, K. & Ariizumi, T. orf137 triggers cytoplasmic male sterility in tomato. Plant. Physiol. 189, 465–468 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Mok, B. Y. et al. A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing. Nature 583, 631–63 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Nakazato, I. et al. Targeted base editing in the plastid genome of Arabidopsis thaliana. Nat. Plants 7, 906–913 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Hu, J. et al. Strand-preferred base editing of organellar and nuclear genomes using CyDENT. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01910-9 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  226. Mok, Y. G., Hong, S., Bae, S. J., Cho, S. I. & Kim, J. S. Targeted A-to-G base editing of chloroplast DNA in plants. Nat. Plants 8, 1378–1384 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Yi, Z. et al. Strand-selective base editing of human mitochondrial DNA using mitoBEs. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01791-y (2023).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (32388201), the National Key Research and Development Program (2022YFF1002802), the Ministry of Agriculture and Rural Affairs of China, the Strategic Priority Research Program of the Chinese Academy of Sciences (Precision Seed Design and Breeding, XDA24020102), and the New Cornerstone Science Foundation. The authors thank K. T. Zhao, C. Xue, R. Liang, G. Liu, J. Hu, H. Li, Y. Li, F. Qiu, S. Li, Y. Lei and X. Jiang for their insightful comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.L., C.S. and C.G. researched the literature. All authors substantially contributed to discussions of the content and wrote the article. J.L. and C.G. reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Caixia Gao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Genetics thanks José R. Botella, who co-reviewed with Zheng (Tommy) Gong; Yuriko Osakabe; and Yiping Qi 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.

Glossary

Agrobacterium rhizogenes

A bacterium used for plant delivery. It can induce the formation of hairy roots in the infection site. It contains a root-inducing plasmid that carries a T-DNA segment capable of integrating into the plant genome. Typically, this T-DNA harbours the desired sequences intended for transfer into the plant genome.

Agrobacterium tumefaciens

A bacterium used for plant delivery. It contains a modified tumour-inducing plasmid that carries a T-DNA segment capable of integrating into the plant genome. Typically, this T-DNA harbours the desired sequences intended for transfer into the plant genome, as well as marker genes for selecting positive events.

CRISPR interference

(CRISPRi). CRISPR interference utilizes dCas9 either alone or with a transcription repressor to inhibit gene expression by targeting specific DNA sequences without altering the genetic code, offering precise control for studying gene functions and regulatory processes within cells.

Guide RNA

(gRNA). An RNA molecule used to direct Cas9 or similar enzymes to a specific DNA or RNA sequence for precise modification.

Hybrid vigour

A phenomenon in which the offspring of two different inbred lines or varieties exhibit improved traits compared to their parents, such as increased yield, growth or biotic or abiotic resistance. Also known as heterosis.

Non-targeted strand

The DNA strand that is not complementary to the guide RNA sequence. DNA nicking by PE2 and base deamination by base editors occur on the non-targeted DNA strand.

Particle bombardment

A genetic transformation technique, also known as gene gun or biolistic delivery, that involves loading exogenous DNA onto microscopic metal particles that are accelerated and propelled into plant cells or other target cells by compressed gas or physical force.

Protospacer adjacent motif

(PAM). A short DNA sequence immediately adjacent to the target site that is essential for the recognition and binding of Cas protein to the target DNA.

R-loop

A specific structure consisting of one DNA strand, its complementary DNA strand and an RNA strand located between them.

Targeted strand

The DNA strand that is complementary to the guide RNA sequence. DNA nicking by base editors occurs on the targeted DNA strand.

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

Li, B., Sun, C., Li, J. et al. Targeted genome-modification tools and their advanced applications in crop breeding. Nat Rev Genet (2024). https://doi.org/10.1038/s41576-024-00720-2

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41576-024-00720-2

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