Search
2025 Volume 4
Article Contents
REVIEW   Open Access    

Emerging nucleases in crop genome editing: towards intellectual property independence and technical flexibility

More Information
  • Received: 18 November 2024
    Revised: 14 March 2025
    Accepted: 31 March 2025
    Published online: 28 May 2025
    Seed Biology  4 Article number: e008 (2025)  |  Cite this article
  • As the global population grows and food security challenges increase, developing high-yield, stress-resistant crops have become crucial. Although CRISPR-Cas9 and CRISPR-Cas12a are the most widely utilized gene editing tools, their associated patent protections are notably stringent. This results in significant patent costs for commercializing breeding using these nucleases, highlighting the need for developing novel nucleases that possess autonomy of intellectual property (IP). In addition, novel nucleases exhibiting diverse recognition sites and hypercompact protein structures offer technical flexibility in gene editing. This review examines advancements in novel nuclease-based genome editing technologies and seeks to provide insights into the patent autonomy of plant genome editing, emphasizing the significant role of emerging compact nucleases in promoting sustainable agricultural practices and ensuring food security.
  • 加载中
  • [1] Flies EJ, Brook BW, Blomqvist L, Buettel JC. 2018. Forecasting future global food demand: a systematic review and meta-analysis of model complexity. Environment International 120:93−103 doi: 10.1016/j.envint.2018.07.019

    CrossRef   Google Scholar

    [2] Chen K, Wang Y, Zhang R, Zhang H, Gao C. 2019. CRISPR/Cas genome editing and precision plant breeding in agriculture. Annual Review of Plant Biology 70:667−97 doi: 10.1146/annurev-arplant-050718-100049

    CrossRef   Google Scholar

    [3] Huang X, Huang S, Han B, Li J. 2022. The integrated genomics of crop domestication and breeding. Cell 185:2828−39 doi: 10.1016/j.cell.2022.04.036

    CrossRef   Google Scholar

    [4] Puchta H. 2005. The repair of double-strand breaks in plants: mechanisms and consequences for genome evolution. Journal of Experimental Botany 56:1−14 doi: 10.1093/jxb/eri025

    CrossRef   Google Scholar

    [5] Wright DA, Townsend JA, Winfrey RJ Jr, Irwin PA, Rajagopal J, et al. 2005. High-frequency homologous recombination in plants mediated by zinc-finger nucleases. The Plant Journal 44:693−705 doi: 10.1111/j.1365-313X.2005.02551.x

    CrossRef   Google Scholar

    [6] Shukla VK, Doyon Y, Miller JC, DeKelver RC, Moehle EA, et al. 2009. Precise genome modification in the crop species Zea mays using zinc-finger nucleases. Nature 459:437−41 doi: 10.1038/nature07992

    CrossRef   Google Scholar

    [7] Townsend JA, Wright DA, Winfrey RJ, Fu F, Maeder ML, et al. 2009. High-frequency modification of plant genes using engineered zinc-finger nucleases. Nature 459:442−45 doi: 10.1038/nature07845

    CrossRef   Google Scholar

    [8] Zhang F, Maeder ML, Unger-Wallace E, Hoshaw JP, Reyon D, et al. 2010. High frequency targeted mutagenesis in Arabidopsis thaliana using zinc finger nucleases. Proceedings of the National Academy of Sciences of the United States of America 107:12028−33 doi: 10.1073/pnas.0914991107

    CrossRef   Google Scholar

    [9] Li S, Li J, He Y, Xu M, Zhang J, et al. 2019. Precise gene replacement in rice by RNA transcript-templated homologous recombination. Nature Biotechnology 37:445−50 doi: 10.1038/s41587-019-0065-7

    CrossRef   Google Scholar

    [10] Shan Q, Wang Y, Chen K, Liang Z, Li J, et al. 2013. Rapid and efficient gene modification in rice and Brachypodium using TALENs. Molecular Plant 6:1365−68 doi: 10.1093/mp/sss162

    CrossRef   Google Scholar

    [11] Zhang Y, Zhang F, Li X, Baller JA, Qi Y, et al. 2013. Transcription activator-like effector nucleases enable efficient plant genome engineering. Plant Physiology 161:20−27 doi: 10.1104/pp.112.205179

    CrossRef   Google Scholar

    [12] Sander JD, Dahlborg EJ, Goodwin MJ, Cade L, Zhang F, et al. 2011. Selection-free zinc-finger-nuclease engineering by context-dependent assembly (CoDA). Nature Methods 8:67−69 doi: 10.1038/nmeth.1542

    CrossRef   Google Scholar

    [13] Miao J, Guo D, Zhang J, Huang Q, Qin G, et al. 2013. Targeted mutagenesis in rice using CRISPR-Cas system. Cell Research 23:1233−36 doi: 10.1038/cr.2013.123

    CrossRef   Google Scholar

    [14] Svitashev S, Schwartz C, Lenderts B, Young JK, Mark Cigan A. 2016. Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nature Communications 7:13274 doi: 10.1038/ncomms13274

    CrossRef   Google Scholar

    [15] Shan Q, Wang Y, Li J, Gao C. 2014. Genome editing in rice and wheat using the CRISPR/Cas system. Nature Protocols 9:2395−410 doi: 10.1038/nprot.2014.157

    CrossRef   Google Scholar

    [16] Xing HL, Dong L, Wang ZP, Zhang HY, Han CY, et al. 2014. A CRISPR/Cas9 toolkit for multiplex genome editing in plants. BMC Plant Biology 14:327 doi: 10.1186/s12870-014-0327-y

    CrossRef   Google Scholar

    [17] Banakar R, Rai KM, Zhang F. 2022. CRISPR DNA- and RNP-mediated genome editing via Nicotiana benthamiana protoplast transformation and regeneration. In Protoplast Technology, eds Wang K, Zhang F. New York, NY: Humana. Volume 2464. pp. 65−82. doi: 10.1007/978-1-0716-2164-6_5
    [18] Sun X, Hu Z, Chen R, Jiang Q, Song G, et al. 2015. Targeted mutagenesis in soybean using the CRISPR-Cas9 system. Scientific Reports 5:10342 doi: 10.1038/srep10342

    CrossRef   Google Scholar

    [19] Lowder LG, Zhang D, Baltes NJ, Paul JW III, Tang X, et al. 2015. A CRISPR/Cas9 toolbox for multiplexed plant genome editing and transcriptional regulation. Plant Physiology 169:971−85 doi: 10.1104/pp.15.00636

    CrossRef   Google Scholar

    [20] Chen X, Zhong Z, Tang X, Yang S, Zhang Y, et al. 2024. Advancing PAM-less genome editing in soybean using CRISPR-SpRY. Horticulture Research 11:uhae160 doi: 10.1093/hr/uhae160

    CrossRef   Google Scholar

    [21] Contreras JL, Sherkow JS. 2017. CRISPR, surrogate licensing, and scientific discovery. Science 355:698−700 doi: 10.1126/science.aal4222

    CrossRef   Google Scholar

    [22] Ledford H. 2017. Bitter CRISPR patent war intensifies. Nature doi: 10.1038/nature.2017.22892

    CrossRef   Google Scholar

    [23] Kim YG, Cha J, Chandrasegaran S. 1996. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proceedings of the National Academy of Sciences of the United States of America 93:1156−60 doi: 10.1073/pnas.93.3.1156

    CrossRef   Google Scholar

    [24] Wolfe SA, Nekludova L, Pabo CO. 2000. DNA recognition by Cys2His2 zinc finger proteins. Annual Review of Biophysics 29:183−212 doi: 10.1146/annurev.biophys.29.1.183

    CrossRef   Google Scholar

    [25] Bitinaite J, Wah DA, Aggarwal AK, Schildkraut I. 1998. FokI dimerization is required for DNA cleavage. Proceedings of the National Academy of Sciences of the United States of America 95:10570−75 doi: 10.1073/pnas.95.18.10570

    CrossRef   Google Scholar

    [26] Symington LS, Gautier J. 2011. Double-strand break end resection and repair pathway choice. Annual Review of Genetics 45:247−71 doi: 10.1146/annurev-genet-110410-132435

    CrossRef   Google Scholar

    [27] Ainley WM, Sastry-Dent L, Welter ME, Murray MG, Zeitler B, et al. 2013. Trait stacking via targeted genome editing. Plant Biotechnology Journal 11:1126−34 doi: 10.1111/pbi.12107

    CrossRef   Google Scholar

    [28] Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, et al. 2010. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics 186:757−61 doi: 10.1534/genetics.110.120717

    CrossRef   Google Scholar

    [29] Christian M, Qi Y, Zhang Y, Voytas DF. 2013. Targeted mutagenesis of Arabidopsis thaliana using engineered TAL effector nucleases. G3 Genes| Genomes| Genetics 3:1697−705 doi: 10.1534/g3.113.007104

    CrossRef   Google Scholar

    [30] Cermak T, Doyle EL, Christian M, Wang L, Zhang Y, et al. 2011. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Research 39:e82 doi: 10.1093/nar/gkr218

    CrossRef   Google Scholar

    [31] Li T, Liu B, Spalding MH, Weeks DP, Yang B. 2012. High-efficiency TALEN-based gene editing produces disease-resistant rice. Nature Biotechnology 30:390−92 doi: 10.1038/nbt.2199

    CrossRef   Google Scholar

    [32] Luo M, Li H, Chakraborty S, Morbitzer R, Rinaldo A, et al. 2019. Efficient TALEN-mediated gene editing in wheat. Plant Biotechnology Journal 17:2026−28 doi: 10.1111/pbi.13169

    CrossRef   Google Scholar

    [33] Wang Y, Cheng X, Shan Q, Zhang Y, Liu J, et al. 2014. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nature Biotechnology 32:947−51 doi: 10.1038/nbt.2969

    CrossRef   Google Scholar

    [34] Hsu PD, Zhang F. 2012. Dissecting neural function using targeted genome engineering technologies. ACS Chemical Neuroscience 3:603−10 doi: 10.1021/cn300089k

    CrossRef   Google Scholar

    [35] Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. 2010. Genome editing with engineered zinc finger nucleases. Nature Reviews Genetics 11:636−46 doi: 10.1038/nrg2842

    CrossRef   Google Scholar

    [36] Doudna JA, Charpentier E. 2014. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346:1258096 doi: 10.1126/science.1258096

    CrossRef   Google Scholar

    [37] Makarova KS, Wolf YI, Alkhnbashi OS, Costa F, Shah SA, et al. 2015. An updated evolutionary classification of CRISPR-Cas systems. Nature Reviews Microbiology 13:722−36 doi: 10.1038/nrmicro3569

    CrossRef   Google Scholar

    [38] Makarova KS, Zhang F, Koonin EV. 2017. SnapShot: Class 1 CRISPR-Cas Systems. Cell 168:946−946.e1 doi: 10.1016/j.cell.2017.02.018

    CrossRef   Google Scholar

    [39] Makarova KS, Zhang F, Koonin EV. 2017. SnapShot: Class 2 CRISPR-Cas Systems. Cell 168:328−328.e1 doi: 10.1016/j.cell.2016.12.038

    CrossRef   Google Scholar

    [40] Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, et al. 2012. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337:816−21 doi: 10.1126/science.1225829

    CrossRef   Google Scholar

    [41] Horvath P, Barrangou R. 2010. CRISPR/Cas, the immune system of bacteria and archaea. Science 327:167−70 doi: 10.1126/science.1179555

    CrossRef   Google Scholar

    [42] Zong Y, Song Q, Li C, Jin S, Zhang D, et al. 2018. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A. Nature Biotechnology 36:950−53 doi: 10.1038/nbt.4261

    CrossRef   Google Scholar

    [43] Thakore PI, D'Ippolito AM, Song L, Safi A, Shivakumar NK, et al. 2015. Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nature Methods 12:1143−49 doi: 10.1038/nmeth.3630

    CrossRef   Google Scholar

    [44] Chen PJ, Liu DR. 2023. Prime editing for precise and highly versatile genome manipulation. Nature Reviews Genetics 24:161−77 doi: 10.1038/s41576-022-00541-1

    CrossRef   Google Scholar

    [45] Breunig CT, Köferle A, Neuner AM, Wiesbeck MF, Baumann V, et al. 2021. CRISPR tools for physiology and cell state changes: potential of transcriptional engineering and epigenome editing. Physiological Reviews 101:177−211 doi: 10.1152/physrev.00034.2019

    CrossRef   Google Scholar

    [46] Fan T, Cheng Y, Wu Y, Liu S, Tang X, et al. 2024. High performance TadA-8e derived cytosine and dual base editors with undetectable off-target effects in plants. Nature Communications 15:5103 doi: 10.1038/s41467-024-49473-w

    CrossRef   Google Scholar

    [47] Ge Z, Zheng L, Zhao Y, Jiang J, Zhang EJ, et al. 2019. Engineered xCas9 and SpCas9-NG variants broaden PAM recognition sites to generate mutations in Arabidopsis plants. Plant Biotechnology Journal 17:1865−67 doi: 10.1111/pbi.13148

    CrossRef   Google Scholar

    [48] Hua K, Tao X, Han P, Wang R, Zhu JK. 2019. Genome engineering in rice using Cas9 variants that recognize NG PAM sequences. Molecular Plant 12:1003−14 doi: 10.1016/j.molp.2019.03.009

    CrossRef   Google Scholar

    [49] Zeng D, Li X, Huang J, Li Y, Cai S, et al. 2020. Engineered Cas9 variant tools expand targeting scope of genome and base editing in rice. Plant Biotechnology Journal 18:1348−50 doi: 10.1111/pbi.13293

    CrossRef   Google Scholar

    [50] Ren Q, Sretenovic S, Liu S, Tang X, Huang L, et al. 2021. PAM-less plant genome editing using a CRISPR-SpRY toolbox. Nature Plants 7:25−33 doi: 10.1038/s41477-020-00827-4

    CrossRef   Google Scholar

    [51] Wu Y, Ren Q, Zhong Z, Liu G, Han Y, et al. 2022. Genome-wide analyses of PAM-relaxed Cas9 genome editors reveal substantial off-target effects by ABE8e in rice. Plant Biotechnology Journal 20:1670−82 doi: 10.1111/pbi.13838

    CrossRef   Google Scholar

    [52] Wang H, Ding J, Zhu J, Liu X, Xu R, et al. 2024. Developing a CRISPR/FrCas9 system for core promoter editing in rice. aBIOTECH 5:189−95 doi: 10.1007/s42994-024-00157-5

    CrossRef   Google Scholar

    [53] Huang TK, Armstrong B, Schindele P, Puchta H. 2021. Efficient gene targeting in Nicotiana tabacum using CRISPR/SaCas9 and temperature tolerant LbCas12a. Plant Biotechnology Journal 19:1314−24 doi: 10.1111/pbi.13546

    CrossRef   Google Scholar

    [54] He Y, Han Y, Ma Y, Liu S, Fan T, et al. 2024. Expanding plant genome editing scope and profiles with CRISPR-FrCas9 systems targeting palindromic TA sites. Plant Biotechnology Journal 22:2488−503 doi: 10.1111/pbi.14363

    CrossRef   Google Scholar

    [55] Steinert J, Schiml S, Fauser F, Puchta H. 2015. Highly efficient heritable plant genome engineering using Cas9 orthologues from Streptococcus thermophilus and Staphylococcus aureus. The Plant Journal 84:1295−305 doi: 10.1111/tpj.13078

    CrossRef   Google Scholar

    [56] Xu R, Qin R, Xie H, Li J, Liu X, et al. 2022. Genome editing with type II-C CRISPR-Cas9 systems from Neisseria meningitidis in rice. Plant Biotechnology Journal 20:350−59 doi: 10.1111/pbi.13716

    CrossRef   Google Scholar

    [57] Xu K, Ren C, Liu Z, Zhang T, Zhang T, et al. 2015. Efficient genome engineering in eukaryotes using Cas9 from Streptococcus thermophilus. Cellular and Molecular Life Sciences 72:383−99 doi: 10.1007/s00018-014-1679-z

    CrossRef   Google Scholar

    [58] Chen W, Ma J, Wu Z, Wang Z, Zhang H, et al. 2023. Cas12n nucleases, early evolutionary intermediates of type V CRISPR, comprise a distinct family of miniature genome editors. Molecular Cell 83:2768−2780.e6 doi: 10.1016/j.molcel.2023.06.014

    CrossRef   Google Scholar

    [59] Swarts DC, van der Oost J, Jinek M. 2017. Structural basis for guide RNA processing and seed-dependent DNA targeting by CRISPR-Cas12a. Molecular Cell 66:221−233.e4 doi: 10.1016/j.molcel.2017.03.016

    CrossRef   Google Scholar

    [60] Pacesa M, Pelea O, Jinek M. 2024. Past, present, and future of CRISPR genome editing technologies. Cell 187:1076−100 doi: 10.1016/j.cell.2024.01.042

    CrossRef   Google Scholar

    [61] Kumar J, Char SN, Weiss T, Liu H, Liu B, et al. 2023. Efficient protein tagging and cis-regulatory element engineering via precise and directional oligonucleotide-based targeted insertion in plants. The Plant Cell 35:2722−35 doi: 10.1093/plcell/koad139

    CrossRef   Google Scholar

    [62] Longo GMC, Sayols S, Kotini AG, Heinen S, Möckel MM, et al. 2025. Linking CRISPR–Cas9 double-strand break profiles to gene editing precision with BreakTag. Nature Biotechnology 43:608−22 doi: 10.1038/s41587-024-02238-8

    CrossRef   Google Scholar

    [63] Marino ND, Zhang JY, Borges AL, Sousa AA, Leon LM, et al. 2018. Discovery of widespread type I and type V CRISPR-Cas inhibitors. Science 362:240−42 doi: 10.1126/science.aau5174

    CrossRef   Google Scholar

    [64] Zhou J, Liu G, Zhao Y, Zhang R, Tang X, et al. 2023. An efficient CRISPR–Cas12a promoter editing system for crop improvement. Nature Plants 9:588−604 doi: 10.1038/s41477-023-01384-2

    CrossRef   Google Scholar

    [65] Tang X, Lowder LG, Zhang T, Malzahn AA, Zheng X, et al. 2017. A CRISPR-Cpf1 system for efficient genome editing and transcriptional repression in plants. Nature Plants 3:17018 doi: 10.1038/nplants.2017.18

    CrossRef   Google Scholar

    [66] Li S, Zhang X, Wang W, Guo X, Wu Z, et al. 2018. Expanding the scope of CRISPR/Cpf1-mediated genome editing in rice. Molecular Plant 11:995−98 doi: 10.1016/j.molp.2018.03.009

    CrossRef   Google Scholar

    [67] Zhong Z, Zhang Y, You Q, Tang X, Ren Q, et al. 2018. Plant genome editing using FnCpf1 and LbCpf1 nucleases at redefined and altered PAM sites. Molecular Plant 11:999−1002 doi: 10.1016/j.molp.2018.03.008

    CrossRef   Google Scholar

    [68] Su H, Wang Y, Xu J, Omar AA, Grosser JW, et al. 2023. Generation of the transgene-free canker-resistant Citrus sinensis using Cas12a/crRNA ribonucleoprotein in the T0 generation. Nature Communications 14:3957 doi: 10.1038/s41467-023-39714-9

    CrossRef   Google Scholar

    [69] Malzahn AA, Tang X, Lee K, Ren Q, Sretenovic S, et al. 2019. Application of CRISPR-Cas12a temperature sensitivity for improved genome editing in rice, maize, and Arabidopsis. BMC Biology 17:9 doi: 10.1186/s12915-019-0629-5

    CrossRef   Google Scholar

    [70] Gray BN, Spruill WM. 2017. CRISPR–Cas9 claim sets and the potential to stifle innovation. Nature Biotechnology 35:630−33 doi: 10.1038/nbt.3913

    CrossRef   Google Scholar

    [71] Starling S. 2017. CRISPR patent results. Nature Reviews Microbiology 15:194 doi: 10.1038/nrmicro.2017.21

    CrossRef   Google Scholar

    [72] Ledford H. 2016. Bitter fight over CRISPR patent heats up. Nature 529:265 doi: 10.1038/nature.2015.17961

    CrossRef   Google Scholar

    [73] Sherkow JS. 2015. Law, history and lessons in the CRISPR patent conflict. Nature Biotechnology 33:256−57 doi: 10.1038/nbt.3160

    CrossRef   Google Scholar

    [74] Charpentier E. 2017. China. Patent No. CN107603976A
    [75] Charpentier E. 2013. China. Patent No. CN104854241A
    [76] Zhang F. 2013. China. Patent No. CN105121648A
    [77] Zhang F. 2016. China. Patent No. CN108513582A
    [78] Zhang F. 2016. China. Patent No. CN109207477A
    [79] Tang X, Zhang Y. 2023. Beyond knockouts: fine-tuning regulation of gene expression in plants with CRISPR-Cas-based promoter editing. New Phytologist 239:868−74 doi: 10.1111/nph.19020

    CrossRef   Google Scholar

    [80] Liu S, He Y, Fan T, Zhu M, Qi C, et al. 2025. PAM-relaxed and temperature-tolerant CRISPR-Mb3Cas12a single transcript unit systems for efficient singular and multiplexed genome editing in rice, maize, and tomato. Plant Biotechnology Journal 23:156−73 doi: 10.1111/pbi.14486

    CrossRef   Google Scholar

    [81] Tang X, Liu G, Zhou J, Ren Q, You Q, et al. 2018. A large-scale whole-genome sequencing analysis reveals highly specific genome editing by both Cas9 and Cpf1 (Cas12a) nucleases in rice. Genome Biology 19:84 doi: 10.1186/s13059-018-1458-5

    CrossRef   Google Scholar

    [82] Bestas B, Wimberger S, Degtev D, Madsen A, Rottner AK, et al. 2023. A Type II-B Cas9 nuclease with minimized off-targets and reduced chromosomal translocations in vivo. Nature Communications 14:5474 doi: 10.1038/s41467-023-41240-7

    CrossRef   Google Scholar

    [83] Guo C, Ma X, Gao F, Guo Y. 2023. Off-target effects in CRISPR/Cas9 gene editing. Frontiers in Bioengineering and Biotechnology 11:1143157 doi: 10.3389/fbioe.2023.1143157

    CrossRef   Google Scholar

    [84] Yu T, Cui H, Li JC, Luo Y, Jiang G, et al. 2023. Enzyme function prediction using contrastive learning. Science 379:1358−63 doi: 10.1126/science.adf2465

    CrossRef   Google Scholar

    [85] Aliaga Goltsman DS, Alexander LM, Lin JL, Fregoso Ocampo R, Freeman B, et al. 2022. Compact Cas9d and HEARO enzymes for genome editing discovered from uncultivated microbes. Nature Communications 13:7602 doi: 10.1038/s41467-022-35257-7

    CrossRef   Google Scholar

    [86] Zhang Y, Ren Q, Tang X, Liu S, Malzahn AA, et al. 2021. Expanding the scope of plant genome engineering with Cas12a orthologs and highly multiplexable editing systems. Nature Communications 12:1944 doi: 10.1038/s41467-021-22330-w

    CrossRef   Google Scholar

    [87] Koonin EV, Makarova KS, Zhang F. 2017. Diversity, classification and evolution of CRISPR-Cas systems. Current Opinion in Microbiology 37:67−78 doi: 10.1016/j.mib.2017.05.008

    CrossRef   Google Scholar

    [88] Liu JJ, Orlova N, Oakes BL, Ma E, Spinner HB, et al. 2019. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 566:218−23 doi: 10.1038/s41586-019-0908-x

    CrossRef   Google Scholar

    [89] Takeda SN, Nakagawa R, Okazaki S, Hirano H, Kobayashi K, et al. 2021. Structure of the miniature type V-F CRISPR-Cas effector enzyme. Molecular Cell 81:558−570.e3 doi: 10.1016/j.molcel.2020.11.035

    CrossRef   Google Scholar

    [90] Duan Z, Liang Y, Sun J, Zheng H, Lin T, et al. 2024. An engineered Cas12i nuclease that is an efficient genome editing tool in animals and plants. The Innovation 5:100564 doi: 10.1016/j.xinn.2024.100564

    CrossRef   Google Scholar

    [91] Liu S, Sretenovic S, Fan T, Cheng Y, Li G, et al. 2022. Hypercompact CRISPR-Cas12j2 (CasΦ) enables genome editing, gene activation, and epigenome editing in plants. Plant Communications 3:100453 doi: 10.1016/j.xplc.2022.100453

    CrossRef   Google Scholar

    [92] Sun A, Li CP, Chen Z, Zhang S, Li DY, et al. 2023. The compact Casπ (Cas12l) 'bracelet' provides a unique structural platform for DNA manipulation. Cell Research 33:229−44 doi: 10.1038/s41422-022-00771-2

    CrossRef   Google Scholar

    [93] Al-Shayeb B, Skopintsev P, Soczek KM, Stahl EC, Li Z, et al. 2022. Diverse virus-encoded CRISPR-Cas systems include streamlined genome editors. Cell 185:4574−4586.e16 doi: 10.1016/j.cell.2022.10.020

    CrossRef   Google Scholar

    [94] Karvelis T, Druteika G, Bigelyte G, Budre K, Zedaveinyte R, et al. 2021. Transposon-associated TnpB is a programmable RNA-guided DNA endonuclease. Nature 599:692−96 doi: 10.1038/s41586-021-04058-1

    CrossRef   Google Scholar

    [95] Badon IW, Oh Y, Kim HJ, Lee SH. 2024. Recent application of CRISPR-Cas12 and OMEGA system for genome editing. Molecular Therapy 32:32−43 doi: 10.1016/j.ymthe.2023.11.013

    CrossRef   Google Scholar

    [96] Saito M, Xu P, Faure G, Maguire S, Kannan S, et al. 2023. Fanzor is a eukaryotic programmable RNA-guided endonuclease. Nature 620:660−68 doi: 10.1038/s41586-023-06356-2

    CrossRef   Google Scholar

    [97] Altae-Tran H, Kannan S, Demircioglu FE, Oshiro R, Nety SP, et al. 2021. The widespread IS200/IS605 transposon family encodes diverse programmable RNA-guided endonucleases. Science 374:57−65 doi: 10.1126/science.abj6856

    CrossRef   Google Scholar

    [98] Han D, Xiao Q, Wang Y, Zhang H, Dong X, et al. 2023. Development of miniature base editors using engineered IscB nickase. Nature Methods 20:1029−36 doi: 10.1038/s41592-023-01898-9

    CrossRef   Google Scholar

    [99] Zhu H, Li C, Gao C. 2020. Applications of CRISPR–Cas in agriculture and plant biotechnology. Nature Reviews Molecular Cell Biology 21:661−77 doi: 10.1038/s41580-020-00288-9

    CrossRef   Google Scholar

    [100] Lin Q, Zhu Z, Liu G, Sun C, Lin D, et al. 2021. Genome editing in plants with MAD7 nuclease. Journal of Genetics and Genomics 48:444−51 doi: 10.1016/j.jgg.2021.04.003

    CrossRef   Google Scholar

    [101] Bai M, Lin W, Peng C, Song P, Kuang H, et al. 2024. Expressing a human RNA demethylase as an assister improves gene-editing efficiency in plants. Molecular Plant 17:363−66 doi: 10.1016/j.molp.2024.02.010

    CrossRef   Google Scholar

    [102] McGaw C, Garrity AJ, Munoz GZ, Haswell JR, Sengupta S, et al. 2022. Engineered Cas12i2 is a versatile high-efficiency platform for therapeutic genome editing. Nature Communications 13:2833 doi: 10.1038/s41467-022-30465-7

    CrossRef   Google Scholar

    [103] Lv P, Su F, Chen F, Yan C, Xia D, et al. 2024. Genome editing in rice using CRISPR/Cas12i3. Plant Biotechnology Journal 22:379−85 doi: 10.1111/pbi.14192

    CrossRef   Google Scholar

    [104] Xie H, Song M, Cao X, Niu Q, Zhu J, et al. 2024. Breeding exceptionally fragrant soybeans for soy milk with strong aroma. Journal of Integrative Plant Biology 66:642−44 doi: 10.1111/jipb.13631

    CrossRef   Google Scholar

    [105] Bai Y, Liu S, Bai Y, Xu Z, Zhao H, et al. 2024. Application of CRISPR/Cas12i. 3 for targeted mutagenesis in broomcorn millet (Panicum miliaceum L.). Journal of Integrative Plant Biology 66:1544−47 doi: 10.1111/jipb.13669

    CrossRef   Google Scholar

    [106] Wang B, Smith SM, Li J. 2018. Genetic regulation of shoot architecture. Annual Review of Plant Biology 69:437−68 doi: 10.1146/annurev-arplant-042817-040422

    CrossRef   Google Scholar

    [107] Li Z, Zhong Z, Wu Z, Pausch P, Al-Shayeb B, et al. 2023. Genome editing in plants using the compact editor CasΦ. Proceedings of the National Academy of Sciences of the United States of America 120:e2216822120 doi: 10.1073/pnas.2216822120

    CrossRef   Google Scholar

    [108] Gong Z, Previtera DA, Wang Y, Botella JR. 2024. Geminiviral-induced genome editing using miniature CRISPR/Cas12j (CasΦ) and Cas12f variants in plants. Plant Cell Reports 43:71 doi: 10.1007/s00299-023-03092-9

    CrossRef   Google Scholar

    [109] Bai S, Cao X, Hu L, Hu D, Li D, et al. 2025. Engineering an optimized hypercompact CRISPR/Cas12j-8 system for efficient genome editing in plants. Plant Biotechnology Journal 23:1153−64 doi: 10.1111/pbi.14574

    CrossRef   Google Scholar

    [110] Karmakar S, Panda D, Panda S, Dash M, Saha R, et al. 2024. A miniature alternative to Cas9 and Cas12: transposon-associated TnpB mediates targeted genome editing in plants. Plant Biotechnology Journal 22:2950−53 doi: 10.1111/pbi.14416

    CrossRef   Google Scholar

    [111] Zhang R, Tang X, He Y, Li Y, Wang W, et al. 2024. IsDge10 is a hypercompact TnpB nuclease that confers efficient genome editing in rice. Plant Communications 5:101068 doi: 10.1016/j.xplc.2024.101068

    CrossRef   Google Scholar

    [112] Lv Z, Chen W, Fang S, Dong B, Wang X, et al. 2024. Targeted mutagenesis in Arabidopsis and medicinal plants using transposon-associated TnpB. Journal of Integrative Plant Biology 66:2083−86 doi: 10.1111/jipb.13758

    CrossRef   Google Scholar

    [113] Pietralla J, Capdeville N, Schindele P, Puchta H. 2024. Optimizing ErCas12a for efficient gene editing in Arabidopsis thaliana. Plant Biotechnology Journal 22:401−12 doi: 10.1111/pbi.14194

    CrossRef   Google Scholar

    [114] An Y, Geng Y, Yao J, Fu C, Lu M, et al. 2020. Efficient genome editing in Populus using CRISPR/Cas12a. Frontiers in Plant Science 11:593938 doi: 10.3389/fpls.2020.593938

    CrossRef   Google Scholar

    [115] Hsu CT, Lee WC, Cheng YJ, Yuan YH, Wu FH, et al. 2020. Genome editing and protoplast regeneration to study plant–pathogen interactions in the model plant Nicotiana benthamiana. Frontiers in Genome Editing 2:627803 doi: 10.3389/fgeed.2020.627803

    CrossRef   Google Scholar

    [116] Wang W, Tian B, Pan Q, Chen Y, He F, et al. 2021. Expanding the range of editable targets in the wheat genome using the variants of the Cas12a and Cas9 nucleases. Plant Biotechnology Journal 19:2428−41 doi: 10.1111/pbi.13669

    CrossRef   Google Scholar

    [117] He Y, Liu S, Chen L, Pu D, Zhong Z, et al. 2024. Versatile plant genome engineering using anti-CRISPR-Cas12a systems. Science China Life Sciences 67:2730−45 doi: 10.1007/s11427-024-2704-7

    CrossRef   Google Scholar

    [118] Hui F, Tang X, Li B, Alariqi M, Xu Z, et al. 2024. Robust CRISPR/Mb2Cas12a genome editing tools in cotton plants. iMeta 3:e209 doi: 10.1002/imt2.209

    CrossRef   Google Scholar

    [119] Ming M, Ren Q, Pan C, He Y, Zhang Y, et al. 2020. CRISPR–Cas12b enables efficient plant genome engineering. Nature Plants 6:202−08 doi: 10.1038/s41477-020-0614-6

    CrossRef   Google Scholar

    [120] Wang Q, Alariqi M, Wang F, Li B, Ding X, et al. 2020. The application of a heat-inducible CRISPR/Cas12b (C2c1) genome editing system in tetraploid cotton (G. hirsutum) plants. Plant Biotechnology Journal 18:2436−43 doi: 10.1111/pbi.13417

    CrossRef   Google Scholar

    [121] Wu F, Qiao X, Zhao Y, Zhang Z, Gao Y, et al. 2020. Targeted mutagenesis in Arabidopsis thaliana using CRISPR-Cas12b/C2c1. Journal of Integrative Plant Biology 62:1653−58 doi: 10.1111/jipb.12944

    CrossRef   Google Scholar

    [122] Haider S, Faiq A, Khan MZ, Mansoor S, Amin I. 2022. Fully Transient CRISPR/Cas12f system in plants capable of broad-spectrum resistance against Begomovirus. bioRxiv preprint doi: 10.1101/2022.06.07.495110

    CrossRef   Google Scholar

    [123] Sukegawa S, Nureki O, Toki S, Saika H. 2023. Genome editing in rice mediated by miniature size Cas nuclease SpCas12f. Frontiers in Genome Editing 5:1138843 doi: 10.3389/fgeed.2023.1138843

    CrossRef   Google Scholar

    [124] Xie H, Su F, Niu Q, Geng L, Cao X, et al. 2024. Knockout of miR396 genes increases seed size and yield in soybean. Journal of Integrative Plant Biology 66:1148−57 doi: 10.1111/jipb.13660

    CrossRef   Google Scholar

    [125] Zhao S, Han X, Zhu Y, Han Y, Liu H, et al. 2024. CRISPR/CasΦ2-mediated gene editing in wheat and rye. Journal of Integrative Plant Biology 66:638−41 doi: 10.1111/jipb.13624

    CrossRef   Google Scholar

    [126] Duan Z, Zhang X, Zhang JT, Li S, Liu R, et al. 2023. Molecular basis for DNA cleavage by the hypercompact Cas12j-SF05. Cell Discovery 9:117 doi: 10.1038/s41421-023-00612-5

    CrossRef   Google Scholar

    [127] Li Q, Wang Y, Hou Z, Zong H, Wang X, et al. 2024. Genome editing in plants using the TnpB transposase system. aBIOTECH 5:225−30 doi: 10.1007/s42994-024-00172-6

    CrossRef   Google Scholar

    [128] Chen Y, Hu Y, Wang X, Luo S, Yang N, et al. 2022. Synergistic engineering of CRISPR-Cas nucleases enables robust mammalian genome editing. The Innovation 3:100264 doi: 10.1016/j.xinn.2022.100264

    CrossRef   Google Scholar

    [129] Xiang G, Li Y, Sun J, Huo Y, Cao S, et al. 2024. Evolutionary mining and functional characterization of TnpB nucleases identify efficient miniature genome editors. Nature Biotechnology 42:745−57 doi: 10.1038/s41587-023-01857-x

    CrossRef   Google Scholar

    [130] Xu Y, Liu T, Wang J, Xiong B, Liu L, et al. 2023. Reprogramming an RNA-guided archaeal TnpB endonuclease for genome editing. Cell Discovery 9:112 doi: 10.1038/s41421-023-00615-2

    CrossRef   Google Scholar

    [131] Esvelt KM, Carlson JC, Liu DR. 2011. A system for the continuous directed evolution of biomolecules. Nature 472:499−503 doi: 10.1038/nature09929

    CrossRef   Google Scholar

    [132] Xu R, Li H, Qin R, Wang L, Li L, et al. 2014. Gene targeting using the Agrobacterium tumefaciens-mediated CRISPR-Cas system in rice. Rice 7:5 doi: 10.1186/s12284-014-0005-6

    CrossRef   Google Scholar

    [133] Ma X, Zhang Q, Zhu Q, Liu W, Chen Y, et al. 2015. A robust CRISPR/Cas9 system for convenient, high-efficiency multiplex genome editing in monocot and dicot plants. Molecular Plant 8:1274−84 doi: 10.1016/j.molp.2015.04.007

    CrossRef   Google Scholar

    [134] Ren C, Liu Y, Guo Y, Duan W, Fan P, et al. 2021. Optimizing the CRISPR/Cas9 system for genome editing in grape by using grape promoters. Horticulture Research 8:52 doi: 10.1038/s41438-021-00489-z

    CrossRef   Google Scholar

    [135] Ordon J, Bressan M, Kretschmer C, Dall'Osto L, Marillonnet S, et al. 2020. Optimized Cas9 expression systems for highly efficient Arabidopsis genome editing facilitate isolation of complex alleles in a single generation. Functional & Integrative Genomics 20:151−62 doi: 10.1007/s10142-019-00665-4

    CrossRef   Google Scholar

    [136] Diamos AG, Mason HS. 2018. Chimeric 3' flanking regions strongly enhance gene expression in plants. Plant Biotechnology Journal 16:1971−82 doi: 10.1111/pbi.12931

    CrossRef   Google Scholar

    [137] Grützner R, Martin P, Horn C, Mortensen S, Cram EJ, et al. 2021. High-efficiency genome editing in plants mediated by a Cas9 gene containing multiple introns. Plant Communications 2:100135 doi: 10.1016/j.xplc.2020.100135

    CrossRef   Google Scholar

    [138] Wang L, Han H. 2024. Strategies for improving the genome-editing efficiency of class 2 CRISPR/Cas system. Heliyon 10:e38588 doi: 10.1016/j.heliyon.2024.e38588

    CrossRef   Google Scholar

    [139] Krenek P, Samajova O, Luptovciak I, Doskocilova A, Komis G, et al. 2015. Transient plant transformation mediated by Agrobacterium tumefaciens: principles, methods and applications. Biotechnology Advances 33:1024−42 doi: 10.1016/j.biotechadv.2015.03.012

    CrossRef   Google Scholar

    [140] Cao X, Xie H, Song M, Lu J, Ma P, et al. 2023. Cut–dip–budding delivery system enables genetic modifications in plants without tissue culture. The Innovation 4:100345 doi: 10.1016/j.xinn.2022.100345

    CrossRef   Google Scholar

  • Cite this article

    Lin W, Li C, Li M, Guan Y. 2025. Emerging nucleases in crop genome editing: towards intellectual property independence and technical flexibility. Seed Biology 4: e008 doi: 10.48130/seedbio-0025-0007
    Lin W, Li C, Li M, Guan Y. 2025. Emerging nucleases in crop genome editing: towards intellectual property independence and technical flexibility. Seed Biology 4: e008 doi: 10.48130/seedbio-0025-0007

Tables(3)

Article Metrics

Article views(4692) PDF downloads(979)

Other Articles By Authors

REVIEW   Open Access    

Emerging nucleases in crop genome editing: towards intellectual property independence and technical flexibility

Seed Biology  4 Article number: e008  (2025)  |  Cite this article

Abstract: As the global population grows and food security challenges increase, developing high-yield, stress-resistant crops have become crucial. Although CRISPR-Cas9 and CRISPR-Cas12a are the most widely utilized gene editing tools, their associated patent protections are notably stringent. This results in significant patent costs for commercializing breeding using these nucleases, highlighting the need for developing novel nucleases that possess autonomy of intellectual property (IP). In addition, novel nucleases exhibiting diverse recognition sites and hypercompact protein structures offer technical flexibility in gene editing. This review examines advancements in novel nuclease-based genome editing technologies and seeks to provide insights into the patent autonomy of plant genome editing, emphasizing the significant role of emerging compact nucleases in promoting sustainable agricultural practices and ensuring food security.

    • By 2050, the world population is projected to surge to 9.6 billion, resulting in an increased demand for staple crops of over 60%[1]. There is an urgent need to develop high-yield, stress-resistant, and adaptable crops to achieve sustainable agricultural development and meet the needs of the growing population. Using genetic resources for crop improvement is one of the key strategies to address this challenge. Traditional crop improvement primarily relies on spontaneous genetic variations that occur in nature or artificial mutations induced through physical or chemical mutagens. However, these approaches are limited by low efficiency and time-consuming processes.

      Nuclease-mediated genome editing has been developing rapidly, representing a highly effective breeding technique[2,3]. Genome editing employs nucleases that generate double-strand breaks (DSBs) at specific genomic sites. These DSBs are then repaired by the host cell using two pathways: non-homologous end joining (NHEJ) or homologous recombination (HDR). This process allows for the introduction of mutations and enables precise modifications to the genome[4]. Pioneering genome editing technologies, such as zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), has successfully facilitated the goal of genome editing in various crops[511]. However, the applications of ZFN and TALEN systems are restricted due to the time-consuming assembly process, high costs, and low editing efficiency[11,12]. The CRISPR (clustered regularly interspaced short palindromic repeats)-Cas (CRISPR-associated proteins) system has emerged as a revolutionary genome editing tool. It offers significant advantages, including high efficiency, ease of design, low cost, and broad applicability, making it the predominant technology in gene editing. The Cas9 nuclease-based genome editing system has been effectively utilized in many plant species[1320]. Currently, research on crop improvement predominantly relies on the Cas9 nuclease. However, restrictions on its use are imposed by intellectual property rights as well as technical limitations. There is an urgent need to explore novel nucleases that offer intellectual property independence[21,22]. Moreover, novel nucleases that exhibit diverse recognition sites, hypercompact protein structures, and evolutionary distinction from Cas9 are being identified, which offers technical flexibility in gene editing. Such development significantly expands the toolkit of genome editing in breeding applications. However, the efficiency of novel compact nucleases in plant genome editing still requires enhancement for practical breeding.

      This review summarizes the development of nuclease-based gene editing technologies and explores the discovery and application of compact nucleases. With particular emphasis on plant genome editing, the work analyzes the unique opportunities and technical challenges confronting emerging compact nucleases in agricultural applications. Finally, the review proposes strategic implementation approaches and critical considerations for leveraging these novel compact editing systems to enhance editing efficiency and advance independent plant breeding initiatives.

    • Programmable gene editing technologies have advanced through three distinct generations of nucleases. The first generation is represented by Zinc Finger Nucleases (ZFN), which are programmable chimeric nucleases consisting of a zinc finger protein (ZFP) DNA-binding domain linked to the Fok I nuclease cleavage domain at the carboxyl terminus[23]. The ZFP comprises a series of Cys2-His2 zinc finger motifs, each capable of recognizing approximately three bp of DNA, allowing for the artificial design of specific DNA sequences[24]. The Fok I enzyme, in contrast, requires the formation of a heterodimer to exert effective cutting action 6−8 bp downstream of its recognition site[25,26]. Over the past two decades, ZFN technology has facilitated targeted modifications of endogenous genes in various crops[58,27]. The second generation of gene editing technology is the Transcription Activator-Like Effector Nuclease (TALEN) system. TALEN consists of Transcription Activator-Like (TAL) proteins that possess sequence-specific binding capabilities coupled with the Fok I nuclease, enabling targeted DNA cleavage. Each TAL monomer can recognize individual nucleotide target sites, thus offering enhanced specificity[28]. In plants, TALEN technology has been applied in genome editing of species such as tobacco[11], Arabidopsis[29,30], rice[10,31], and wheat[32,33]. ZFN and TALEN technologies rely on the customized design and synthesis of DNA sequence-specific protein modules. This process is laborious, time-consuming, and costly, limiting their broader applicability[34,35].

      The third-generation technology, comprised of CRISPR-Cas gene editing, is characterized by its remarkable simplicity, robust programmability, and exceptional targeting efficiency, making it widely utilized in the genome editing of both plants and animals[36]. The most commonly applied systems within this category are CRISPR-Cas9 and CRISPR-LbCas12a (or Cpf1). The CRISPR-Cas system is an RNA-mediated adaptive immune system in prokaryotes that classifies various systems based on sequence similarities between Cas genes and modes of adaptation and interference, encompassing two significant classes and six types[37]. Class 1 systems include types I, III, and IV, primarily found in bacteria and archaea, typically forming multi-subunit protein-crRNA effector complexes[38]. Class 2 systems, comprising types II, VI, and V, rely on a single crRNA-guided protein nuclease to achieve localization and cleavage at target sites[39].

      The engineered CRISPR-Cas9 system, derived from the Class 2 Type II CRISPR system, consists of two core components: the Cas9 endonuclease and a single-guide RNA (sgRNA). The sgRNA is a chimeric molecule formed by fusing crRNA (complementary to the target DNA) with tracrRNA (required for Cas9 activation), bypassing the need for RNase III-mediated processing in natural systems[40]. Cas9 contains two catalytic domains: the HNH domain cleaves the DNA strand complementary to the crRNA, while the RuvC-like domain cuts the non-complementary strand. This dual cleavage occurs three nucleotides upstream of the PAM sequence (typically 5'-NGG-3' on the non-target strand), generating blunt-ended double-strand breaks (DSBs)[36,41]. Various derivatives of Cas9, such as nCas9 and dCas9, further enhance CRISPR technology, enabling single-base editing, prime editing, transcriptional regulation, and epigenetic modifications[4246]. PAM-relaxed variants like xCas9, Cas9-NG, and SpRY significantly expand the regions of CRISPR-Cas9 targets within the plant genome, enriching the plant gene editing toolbox[4651]. On the other hand, exploring and utilizing SpCas9 ortholog (e.g., FrCas9, SaCas9, St1Cas9, NmCas9) further expands the options for nucleases in plant gene editing[5256]. Notably, St1Cas9 exhibits reduced off-target effects and cytotoxicity, making it advantageous for plant genome editing[57].

      The Cas12-type nuclease, classified as a Class II, Type V effector, can be subdivided into over twenty family proteins, ranging from Cas12a to Cas12n, each exhibiting distinct structural and functional characteristics[58]. CRISPR-Cas12a, a Class II Type V effector, differs fundamentally from Cas9 in both structure and function. It requires only a single crRNA for targeting and possesses intrinsic RNase activity to autonomously process precursor crRNA into mature forms, without the need for tracrRNA or auxiliary enzymes[59,60]. Cas12a recognizes T-rich PAM sequences (e.g., 5'-TTTN-3' for LbCas12a) located upstream of the target site on the non-complementary strand. Its single RuvC domain cleaves both DNA strands sequentially, generating staggered double-strand breaks with 5' overhangs 18−23 bp downstream of the PAM[6163]. This T-rich PAM preference enables efficient editing in AT-rich genomic regions (e.g., plant promoters)[64]. Cas12a system can process multiple crRNAs from a single transcript also facilitates multiplex genome editing, a critical feature for engineering plant polygenic traits[6568]. Currently, several types of Cas12a nucleases have been discovered and applied in plants. LbCas12a and AsCas12a are the most commonly used due to their high editing efficiency and strong applicability[69].

    • Gene editing technology based on the CRISPR-Cas system has emerged as a pivotal advancement in genetic engineering. There has been significant interest from various research teams and enterprises, which has led to a heightened focus on the intellectual property (IP) landscape related to this technology. Cas9 is the first nuclease utilized within the CRISPR framework, demonstrating a broader patent scope than the overall CRISPR technology[70]. Given the complexity of CRISPR-Cas patents in different countries, here we focus on the patent landscape in China as a representative.

      The foundational patent rights for CRISPR-Cas9 are held by two main groups: the CVC team (including the University of California, the University of Vienna, and the Emmanuelle Charpentier) and the Broad Institute team, which includes the Broad Institute, the Massachusetts Institute of Technology, and the President and Fellows of Harvard College. In 2013 (priority date in 2012), the CVC team filed a patent application for the CRISPR-Cas9 system, establishing a crucial foundation for the technology's subsequent development. The Broad Institute submitted corresponding patent applications in the same year, ultimately leading to a patent dispute with the University of California. Both parties claim rights to the CRISPR-Cas9 technology, leading to considerable overlap in the patents involved. After a decade of legal proceedings, the United States Patent and Trademark Office (USPTO) concluded in 2021 that specific patents held by the Broad Institute were valid and that the institute held rights to them. Nonetheless, disputes regarding the scope and legality of specific patents persist between the two entities[7173].

      The patent landscape for CRISPR-Cas9 system nucleases is quite complex, and the opportunities for operating freely within Cas9-based systems are limited. As a result, users of this technology must carefully assess patent risks and develop effective strategies to navigate these challenges. The patent issues surrounding Cas9 may pose significant restrictions on the commercial activities of emerging companies, particularly adversely affecting small enterprises that may find it challenging to afford the high costs of patent licensing. This situation could ultimately hinder technological development and application.Despite ongoing invalidation challenges to CRISPR-Cas9 related patents over the past years, Chinese patent authorities have consistently upheld the validity and broad claims of patent portfolios, particularly the enforceability of the divisional application CN201710585690.5 from the CVC team, encompassing rights related to sgRNA and its application, as shown in Table 1. Consequently, circumventing this patent while utilizing Cas9-sgRNA nucleases presents considerable challenges[74].

      Table 1.  Published patents of CRISPR-Cas9 and CRISPR-Cas12 systems in China.

      Assignee Application number Main claims Expiration date
      CVC team CN201380038920.6 The genome editing system is composed of Cas9 and sgRNA. 2033-03-15
      CN201710585690.5 The genome editing system is composed of Cas9 and sgRNA. 2033-03-15
      Broad Institute team CN201380070567.X The vector system for Cas9 and gRNA is utilized in eukaryotic cells. 2033-12-12

      It is important to note that the gRNA patent held by the CVC team is expressly limited to its use in conjunction with the Cas9 protein. This patent does not cover gRNA interacting with other nucleases, such as Cas12[7476]. Additionally, the scope of patent protection for crRNA associated with Cas12 proteins remains ambiguous. Given the diversity of Cas12 protein types, developing and applying novel Cas12-based systems for plant gene editing may represent an effective strategy for overcoming patent barriers. Currently, the earliest patent for LbCas12a (Cpf1) nucleases is held by the Broad Institute, primarily protecting the V-A type CRISPR system (Application No. CN201680035959.6) and a CRISPR system based on Cpf1 with a PAM of 5’T (Application No. CN201810911002.4)[77,78]. Additionally, several patents related to V-type nucleases, beyond the V-A type, have already been filed and granted in China, as detailed in Table 2.

      Table 2.  IP landscape of representative nucleases in China.

      Assignee Application number Claims Expiration date
      Cas12b Institute of Zoology, Chinese Academy of Sciences, and Beijing Institute of Stem Cell and Regenerative Medicine CN202110591698.9 Cas12b variants 2040-12-07
      chCas12b Fudan University CN202110606220.9 chCas12b (Cas12J-8) 2041-5-31
      Cas12d/CasY Longping Biotechnology (Hainan) Co., Ltd. CN202310237362.1 CasD 2043-03-13
      Cas12f/Cas14 Jilin Academy of Agricultural Sciences CN202210204370.1 CasF2 2042-03-03
      Cas12f (cas12i3) China Agricultural University CN201980014560.3 Cas12f 2039-10-29
      CN202110473640.4 Divisional Application, Under examination /
      CN202110473632.X Divisional Application 2039-10-29
      Bellagen Biotechnology Co., Ltd. CN202210603607.3 Cas12i3 variants with S7R, Y124R mutations 2042-05-31
      Bellagen Biotechnology Biotechnology Co., Ltd. CN202210314807.7 Cas12i3 variants with N417K, S638N mutations 2042-03-29
      Bellagen Biotechnology Co., Ltd. CN202310088437.4 Cas12i3 variants with D233R, D267R, N369R, S433R mutations 2043-02-02
      Bellagen Biotechnology Co., Ltd. CN202311039065.2 Fusion of Cas9-HNH with Cas12i3 2043-08-17
      Cas12i China Agricultural University CN201980027152.1 Cas12i (Cas12i.1) protein and application 2039-04-19
      Institute of Zoology, Chinese Academy of Sciences, and Beijing Institute of Stem Cell and Regenerative Medicine CN202011414384.3 Cas12i variants 2040-12-07
      Cas12i12 HuiEdit Therapeutics Co., Ltd. CN202111290670.8 Under examination /
      Bellagen Biotechnology Co., Ltd. CN202310096173.7 Cas12i12 variants with G179R, N269R, Q445R, V447R, E492R, T632R, A825R mutations, under examination /
      Cas12j/Casφ China Agricultural University CN201980014005.0 Cas12j.19 2039-11-15
      CN202110475316.6 Divisional Application of Cas12j.4 2039-11-15
      CN202110475336.3 Divisional Application, under examination /
      Cas12j.19 Bellagen Biotechnology Co., Ltd. CN202310092231.9 Cas12j.19 variant with E100R mutation 2043-02-03
      IscB HuidaGene Therapeutics Co., Ltd. CN202311617088.7 Novel IscB polypeptides and their applications 2043-11-30
      TnpB Beijing Coveland Biological Science Co., Ltd. CN202311230195.4 Mini-TnpB after functional domain streamlining (less than 380 AA) 2043-09-22
      Huazhong Agricultural University CN202310177144.3 TnpB identified from Archaea 2043-02-28
      Gs12-7 Huazhong Agricultural University CN202310086152.7 Nucleic acid endonucleases with high activity and temperature tolerance 2043-01-17
      Gs12-16/Gs12-18 Huazhong Agricultural University CN202310256014.9 Gs12-16 (PAM=NYYV), and Gs12-18 (PAM=YTYV) (V=A/C/G)(Y=C/T) 2043-03-16
      Gs12-10 Huazhong Agricultural University CN202310006257.7 Gs12-10 nuclease without PAM restrictions 2043-01-02
      Gs12-3/Gs12-5 Huazhong Agricultural University CN202310247616.8 Nucleases with a wider range of target sites 2043-03-10
      CasD Longping Biotechnology (Hainan) Co., Ltd. CN202310237362.1 CasD nuclease with 5'-TNN PAM 2043-03-13
      PaeCascade Sun Yat-sen University CN202010527709.2 Type I-F based CRISPR/Cas system 2040-06-11
      Cascade-Cas3 China Pharmaceutical University CN202211136496.6 Type I-B CRISPR-Cascade-Cas3 2042-09-19

      Regarding the novel compact nucleases of the Cas12 class currently explored, most related patents ar encompass the design of new CRISPR/Cas systems, the development of novel enzymes, and specific gene-editing methods applicable to certain plants or animals. Chinese patent applicants primarily include universities, research institutions, biotechnology companies, and various startups. Many enterprises seek to expand their technological barriers and safeguard their commercial interests through patent applications. Furthermore, with the growing global interest in CRISPR technology, research institutions, and companies are increasingly engaging in international collaboration and exchange, aiming to enhance their global influence in terms of intellectual property rights.

    • With the widespread application of Cas9 technology and the increasing expansion of associated patent protections, significant challenges to the autonomy of intellectual property rights for gene editing have emerged. The currently utilized CRISPR-Cas9 nuclease system consists of over 1,300 amino acids, necessitating integrating regulatory elements such as promoters and termination signals during vector delivery. This size surpasses the cargo capacity of viral delivery of transgene in plants. Moreover, the strict recognition of the protospacer adjacent motif (PAM) sequence (NGG) by Cas9 limits its targeting ability in adenine-thymine (A-T) rich regions, such as promoters[79]. Thus, editing crop promoters targeted by nucleases like Cas12a can enhance regional coverage, create genotype-rich mutants, achieve variations in crop quantitative traits, and enable the screening of desirable crop characteristics[64,80]. Additionally, during the precise targeting of recipient genomes, the CRISPR-Cas system may encounter off-target effects, and the off-target efficiency depends not only on nucleases, sgRNAs, and tissue culture processes[8183]. These challenges have significantly driven the exploration and application of novel compact nucleases. Consequently, more researchers are leveraging genomic prediction and bioinformatics tools to identify new compact nucleases with endonuclease activity and flexible intellectual property rights across diverse bacterial and archaeal species[84]. Building upon these findings, they aim to develop more comprehensive and adaptable programmable genome editing tools through synthetic biology approaches.

      Based on phylogenetic relationships and classification, the novel nucleases identified were categorized into Class II-Type II and Class II-Type V analogs. Within the second class of type II, several effectors rich in arginine and lysine have been successfully identified, including the Cas9d and Cas9C2 series, which consist of approximately 600 to 1,050 amino acids[85]. These effectors differ from conventional Cas9 by encoding a zinc finger within the HNH structural domain and incorporating multiple zinc-binding motifs in their recognition domain. Cas9d has exhibited an editing efficiency of up to 90% in mammalian cells, presenting a promising alternative for efficient nuclease-mediated genome editing[85].

      Additionally, the Type V Cas12 nuclease system encompasses a wide variety of variants characterized by their relatively small size. FnCas12a is particularly effective at low temperatures, allowing for efficient editing in cold conditions[69]. Additionally, Cas12a orthologs such as Lb5Cas12a, BsCas12a, Mb2Cas12a, TsCas12a, and MbCas12a nucleases have also been developed for plant gene editing[86]. With ongoing exploration, several novel compact nuclease systems have been discovered and applied, including CasX(Cas12e)[87,88], Cas14(Cas12f)[89], Cas12i[90], CasΦ(Cas12j)[91], and Casπ(Cas12l)[92]. It is noteworthy that in 2020, researchers identified the Casλ effector from a family of enzymes encoded by bacteriophages. This effector possesses a unique guide RNA (gRNA) capable of successfully inducing genome editing of endogenous genes in both human and plant cells[93].

      Recent phylogenetic analyses have identified Fazor and TnpB from the OMEGA (specific mobile element-guided activity) system as ancestors of CRISPR-Cas12, while IsCB is recognized as the ancestor of CRISPR-Cas9. Notably, these nucleases possess an extremely compact size that facilitates the delivery of adeno-associated viruses (AAV), suggesting they may substitute CRISPR-Cas9 in vivo gene editing, thereby offering promising applications. TnpB is categorized as a programmable RNA-guided functional nuclease, cleaving DNA near the 5' end of the transposon-associated motif (TAM) through reRNA (right element RNA), enabling precise targeting and editing of the genomic DNA[94]. Researchers have identified various TnpB variants with distinct characteristics that allow genome editing across multiple cell types[95]. Subsequently, based on AlphaFoldDB, the researchers identified a eukaryotic TnpB-IS200/IS605-like protein named Fanzor, which exhibits RNA-induced editable nuclease functionality in eukaryotes[96]. In addition, a compact RNA-inducible nuclease, IscB (~400 amino acids, primarily composed of a RuvC structural domain and an HNH nuclease structural domain), has been identified from IS200/IS605 superfamily transposons. It is associated with evolutionarily conserved non-coding RNAs. Phylogenetic analyses indicate that all extant Cas9 enzymes are derived from a single common ancestor, IscB[97]. The compact IscB exhibits low editing activity in eukaryotic cells, thus limiting its further applications. An efficient enIscB system was developed for mammals, enhancing the toolkit for eukaryotic genome editing[98]. Those novel nucleases within the OMEGA system generally exhibit an extremely compact size, providing significant delivery and vector assembly advantages.

    • Due to its unparalleled ability to precisely manipulate plant genomes, the CRISPR-Cas system has become a powerful tool in agriculture[99]. This technology facilitates the rational design of varietal traits, significantly accelerating the domestication process of various species and fundamentally transforming traditional breeding systems[2].

      With the ongoing exploration of the CRISPR-Cas system, novel compact nucleases have emerged in plant genome editing alongside the conventional Cas9 and Cas12a. These nucleases are characterized by their smaller size and greater flexibility, making them the focus of considerable interest (Table 3). Nucleases such as CasX, MAD7, and various Cas12 variants offer distinct advantages in targeting specific genomic sequences, thereby promoting precise modifications of plant genes.

      Table 3.  Representative novel nucleases for genome editing in plant.

      Nuclease Origin Editor type PAM/TAM Species Ref.
      MAD7 (ErCas12a) Eubacterium rectale Knockout/CBE YTTN Rice, wheat, soybean, arabidopsis [86,100,101,113]
      FnCas12a Francisella novicida Knockout TTV Rice, maize, arabidopsis, Populus, tobacco, wheat [67,69,114116]
      Lb5Cas12a Lachnospiraceae bacterium MA2020 Knockout TTTV Rice [86]
      BsCas12a Butyrivibrio sp. NC3005 Knockout VTTV Rice [86]
      MbCas12a Moraxella bovoculi Knockout TTTV Rice [86]
      Mb2Cas12a Moraxella bovoculi strain 57922 Knockout VTTV Rice and cotton [86,117,118]
      Mb2Cas12a-RVR Moraxella bovoculi strain 57922 Knockout TATV Rice [86]
      Mb3Cas12a Moraxella bovoculi AAX11 00205 Knockout NTTV Rice, maize, tomato [80]
      HkCas12a Helcococcus kunzii ATCC 51366 Knockout VTTV Rice [80]
      PrCas12a Pseudobutyrivibrio ruminis CF1b Knockout VTTV Rice [80]
      TsCas12a Thiomicrospira sp. XS5 Knockout TTTV Rice [86]
      AaCas12b Alicyclobacillus acidiphilus Knockout/CRISPRi/CRISPRa VTTV Rice [119]
      AacCas12b Alicyclobacillus acidoterrestris Knockout/CRISPRi/CRISPRa TTV Rice and cotton [119,120]
      BhCas12b Bacillus hisashii Knockout/CRISPRi/CRISPRa VTTV Rice [119]
      BthCas12b Bacillus thermoamylovorans Knockout/CRISPRi/CRISPRa ATTN Rice [119]
      BvCas12b Bacillus sp. V3-13 Knockout TTN Arabidopsis [121]
      BhCas12b v4 Bacillus hisashii variant Knockout TTN Arabidopsis [121]
      Cas12f
      Syntrophomonas palmitatica (Sp), Acidibacillus sulfuroxidans, and uncultured archaeon (Un1) Knockout TTTR Tobacco and rice [108,122,123]
      Cas12i3 (Cas-SF01) Lachnospiraceae bacterium Knockout NNTN Rice, pepper, soybean [90,104,124]
      CasΦ(Cas12j-2) Biggiephage Knockout/CRISPRa/Epigenome editor/CBE/ABE NTTV Arabidopsis, rice, tomato, tobacco, wheat, rye [91,107,108,125]
      Cas12j-SF05 Caudoviricetes Knockout TTN Rice [126]
      Cas12j-8 Biggiephage Knockout/CBE TTN Soybean and rice [109]
      TnpB-IsDra2 Deinococcus radiodurans Knockout/ABE/CRISPRa TTGAT Rice and arabidopsis [110112,127]
      TnpB-IsYmu1 Youngiibacter multivorans Knockout TTGAT Rice [111,127]
      TnpB-IsAam1 Anoxybacillus amylolyticus Knockout TTTAA Rice [111,127]
      TnpB-IsDge10 Deinococcus geothermalis Knockout TTAT Rice [111]
      enIscB Biggiephage Knockout NWRRNA Rice [111]

      The CRISPR-MAD7 is claimed to be a royalty-free nuclease derived from the Eubacterium rectale found in Madagascar. This nuclease shares about 76% nucleotide sequence similarity with its natural counterpart and consists of approximately 1,200 amino acids, while the amino acid homology with Acidaminococcus Cas12a (AsCpf1) is only 31%[86,100]. Nonetheless, it remains unclear whether CRISPR-MAD7, a Cas12a-type nuclease, is subject to existing LbCas12a or AsCas12a patents. MAD7 preferentially recognizes the YTTN protospacer adjacent motif (PAM), but its application in plants is limited. The CRISPR-MAD7 9 system has achieved editing efficiencies of 49.0% to 65.4% at OsALS and OsEPSPS sites in rice, though it has lower activity than other Cas12a nucleases[86,100]. In soybean root hairs, about 20% to 35% editing efficiency was achieved through co-expression of human FTO proteins[101].

      The application of the nuclease from the sub-type V-I system (Cas12i) represents a significant development in using emerging compact nucleases for plant genome editing. Compared to SpCas9 and Cas12a, Cas12i effectors are smaller (ranging from 1,033 to 1,093 amino acids), and their mature crRNA is shorter (40-43 nucleotides), making Cas12i particularly suitable for multiplex genome editing and virus-based delivery systems[102]. Structure-guided rational design and protein engineering were employed to optimize the previously uncharacterized Cas12i3 nuclease, leading to the development of Cas-SF01. This variant exhibits a broader PAM range, enabling effective recognition of NTTN sequences. An upgrade to the Cas-SF01HiFi system, achieved by incorporating the D876R modification, enhances its low off-target activity and high efficiency[90]. Approximately 77.2% editing efficiency was observed in stabilized transformation events at 12 targeting sites in monocotyledonous rice mediated by Cas-SF01, with an increased occurrence of homozygous, bi-allelic, and chimeric events in the E0 generation. Furthermore, the system demonstrated approximately 52.5% editing efficiency in dicot chili peppers and soybean root hairs, which is twice the editing efficiency of Cas9 (about 26.8%), underscoring its substantial potential in plant research and molecular breeding[90]. To enable the aggregation of multiple genetic traits in rice, the Cas12i3-based iMAGE serial system was constructed to edit multiple endogenous targets, achieving knockout efficiencies ranging from 38.4% to 47.3% at the OsNramp5 gene locus[103]. Additionally, an ABE system based on Cas12i3 was developed, which resulted in base conversions ranging from 4.0% to 7.3% in rice callus[103]. In soybean, targeting the aroma candidate genes GmBADH1 and GmBADH2 using Cas12i3 enabled the successful breeding of soybean varieties with enhanced aroma characteristics suitable for soybean milk production[104]. In addition to staple crops, the Cas12i3-based knockout of the PmBR2-a and PmBR2-b genes in broomcorn millet has successfully resulted in the production of plants exhibiting dwarf phenotypes, leading to denser planting and ultimately higher harvest indices[105,106]. Overall, Cas12i3 demonstrates remarkable performance in crop breeding, with Cas-SF01 achieving editing efficiencies at specific loci that meet or exceed those of Cas9. Furthermore, this nuclease reduces dependency on patent-constrained Cas12a systems, positioning it as a strategically valuable tool for modern crop breeding and commercialization.

      CasΦ (Cas12j), a nuclease (about 700-800 amino acids) derived from the family of large bacteriophages, represents another compact nuclease that has been explored and applied in plants. This nuclease can recognize T-rich PAM sequences and demonstrates heightened sensitivity to chromatin environments[107]. CasΦ-2 (also known as Cas12j-2) has been employed to target the Arabidopsis AtPDS locus for heritable edits for plant genome editing. Additionally, several variants of vCasΦ and nCasΦ have been developed to enhance editing efficiency[107]. Despite recent advances, the overall efficiency remains unsatisfactory. Cas12j has been demonstrated to facilitate gene editing in several species, including tomato, tobacco, and poplar[91,108]. Through the rational engineering of crRNA and Cas12j-8 proteins, a gene knockout and cytosine base editor system based on en4Cas12j-8/cRNA-Rz was developed, showing promising editing activity in rice and soybean[109].

      The TnpB nuclease from the OMEGA system, derived from the transposon family, is currently one of the most compact nucleases achieving genome editing in plants. Among these, the ISDra2 TnpB (about 400 amino acids, target adjacent motif (TAM: TTGAT) from Deinococcus radiodurans was the first to demonstrate targeted editing in rice protoplasts and regenerated callus, with editing efficiencies ranging from approximately 1.5% to 7.15%. Furthermore, employing a tandem tRNA-crRNA-HDV system achieved multiplex genome editing in rice, with a maximum editing efficiency of 14.84% ± 4.88% at the OsHMBPP locus[110]. Subsequent applications of the same strategy in Arabidopsis thaliana yielded an editing efficiency of around 1%[110]. As efforts to explore TnpB nucleases continue, additional variants such as ISAam1 (about 360 amino acids, TAM: TTTAA) and ISYmu1 (about 380 amino acids, TAM: TTGAT) have also been identified and utilized for editing in rice and Arabidopsis. However, the overall editing efficiency remains relatively low. Notably, the IsDge10 TnpB (about 390 amino acids, TAM: TTAT), isolated from Deinococcus geothermalis, demonstrated editing efficiencies ranging from 2.20% to 15.04% in rice protoplasts and from 4.2% to 25.0% in T0 regenerated seedlings. This TnpB also succeeded in editing seven target sites simultaneously in protoplasts using multiple target tandems, with efficiencies ranging from approximately 4.3% to 18.2%[111]. Moreover, IsDra2 has been utilized for genome editing in various medicinal plants, underscoring the promising applications of this novel compact nuclease in regulating secondary metabolite production in medicinal flora[112]. Currently, the efficiency of TnpB-like novel nucleases in genome editing across various plant species still presents significant opportunities for improvement. Additionally, TnpB's recognition of target TAM sequences shows considerable limitations. Therefore, mining novel TnpB nucleases with broader TAM recognition capabilities, or the engineering of existing nucleases to enhance genome editing efficiencies, is essential for advancing the utility of TnpB nucleases in plant biotechnology.

    • The discovery and efficient application of novel nucleases can significantly alleviate the constraints of intellectual property rights in plant breeding. Exploring novel nucleases and developing efficient editing systems are crucial for expanding a toolset of editing technologies with independent intellectual property rights. However, the successful application of novel nucleases derived from various sources, including microorganisms, in plants still faces challenges. One of the challenges includes the complex IP landscape surrounding CRISPR-Cas nucleases[128]. Furthermore, eukaryotic systems, such as plants, are inherently more complicated than prokaryotic systems, which presents additional challenges for applying novel nucleases in plants. These challenges include low editing efficiency, difficulties in delivery, and reduced activity of the nucleases.

      Bioinformatics and associated tools are leveraged to discover new nucleases with independence of intellectual property. For example, by developing a large-scale mining approach for TnpB-related targeted gene-editing systems, researchers managed to identify 33 TnpB proteins that have targeted editing functions in prokaryotic systems. Among them, five were found to be active in eukaryotic systems. This eventually led to developing the most minor viable micro gene editor with independent IP to date[129]. Subsequently, the first miniaturized novel nuclease system, SisTnpB1, derived from the archaeon Sulfolobus Islandicus REY15A, was characterized using a mining approach, and an efficient gene editing method was developed[130]. Moreover, based on AlphaFold Protein Structure Database (AlphaFold DB), the researchers discovered and validated the characterization of Fazor proteins as nucleases in the eukaryotic system[96]. These examples demonstrate that employing relevant algorithms to identify novel nucleases with endonuclease activity can effectively circumvent the patent restrictions associated with CRISPR-Cas9 and Cas12 systems, leading to the establishment of various functional editors suitable for eukaryotic systems.

      Engineering modifications can be utilized to boost the efficiency of genome editing when using new compact nucleases. Researchers have employed a variety of evolutionary approaches to improve the activity and specificity of these novel nucleases in host plants. One way to achieve this is through a process called directed evolution. This involves introducing random mutations into the nuclease genes and screening for enzyme variants performing exceptionally well under specific conditions. By doing so, researchers can obtain enzymes with enhanced activity. Several methods are used in directed evolution, such as extracellular mutagenesis, intracellular mutagenesis, and virus-assisted evolution. In 2011, the phage-assisted continuous evolution (PACE) system was reported, which has further advanced the field of nuclease engineering[131].

      Additionally, using advanced technologies such as X-ray crystallography and cryo-electron microscopy enables researchers to analyze the three-dimensional structures of nucleases. These structural insights provide critical guidance for selecting mutation sites, allowing researchers to optimize enzyme structures effectively to enhance their functionality. The application of computational modeling will enable researchers to predict the impact of mutations on enzyme activity, which not only accelerates the optimization process but also provides a theoretical basis for designing more efficient nucleases. Researchers can further refine enzyme designs by integrating experimental data with computational models, improving their effectiveness in plant genome editing[126].

      Different species exhibit unique codon preferences; implementing codon optimization can significantly influence the stability of heterologous mRNA in target species, thereby enhancing the expression levels of nucleases and ultimately improving gene editing efficiency[132,133]. In editing systems, optimizing nuclease expression levels relies not only on the design of coding regions but also on adjusting the combinations of non-coding region elements, such as selecting strong promoters for ubiquitous expression, varying the combinations of 5' and 3' untranslated regions (UTR), applying expression enhancers, and manipulating the number of introns within exogenous genes[134137]. Furthermore, the secondary structure stability of sgRNA and cutting efficiency also significantly impact gene editing outcomes. Optimization strategies, such as incorporating HDV elements, tandem transfer RNA (tRNA), and improving secondary structures, have considerably enhanced editing effectiveness[138]. Additionally, the chromatin environment surrounding the target gene sites affects the accessibility of the nuclease complex, and regulating chromatin openness can significantly enhance the editing efficiency of endogenous genes in plants[101]. Optimizing expression systems can substantially improve the expression abundance of heterologous CRISPR-Cas systems within plant tissues, thereby facilitating increases in gene editing efficiency.

      Enhancing the efficiency of plant genome editing primarily relies on effective delivery and transformation systems. While established methods such as Agrobacterium-mediated transformation, transformation of callus tissues with Agrobacterium tumefaciens, gene gun transformation, RNP transformation, and root-based cell transformation systems (CDB) are already in use, there is an urgent need to develop novel and efficient delivery strategies. These innovations are vital to maximizing the editing capabilities of the new nucleases[139,140].

      In summary, with the rapid advancement of genome editing technologies, exploring novel compact nucleases offers unprecedented opportunities for plant genome editing. Optimizing and advancing new tools based on these compact nucleases would help avoid the fundamental IP issues linked to Cas9 and Cas12 and enable the autonomy of core technologies in gene editing systems to reduce the technical barriers.

      • This work was supported by the National Key Research and Development Program of China (2023YFF1000203) to ML. We sincerely apologize to those researchers whose valuable contributions could not be cited in this work. Due to the limited space and scope of this review, we could not include all research on the emerging novel nuclease for plant gene editing.

      • The authors confirm their contributions to the paper: study conception and design: Guan Y, Li M; data collection: Lin W, Li C; draft manuscript preparation: Guan Y, Lin W, Li C. All authors reviewed the results and approved the final version of the manuscript.

      • All data generated or analyzed during this study are included in the published article.

      • The authors declare that they have no conflict of interest.

      • Copyright: © 2025 by the author(s). Published by Maximum Academic Press on behalf of Hainan Yazhou Bay Seed Laboratory. This article is an open access article distributed under Creative Commons Attribution License (CC BY 4.0), visit https://creativecommons.org/licenses/by/4.0/.
    Table (3) References (140)
  • About this article
    Cite this article
    Lin W, Li C, Li M, Guan Y. 2025. Emerging nucleases in crop genome editing: towards intellectual property independence and technical flexibility. Seed Biology 4: e008 doi: 10.48130/seedbio-0025-0007
    Lin W, Li C, Li M, Guan Y. 2025. Emerging nucleases in crop genome editing: towards intellectual property independence and technical flexibility. Seed Biology 4: e008 doi: 10.48130/seedbio-0025-0007

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return