Search
2023 Volume 2
Article Contents
REVIEW   Open Access    

Advances in CRISPR/Cas technologies and their application in plants

  • # These authors contributed equally: Chao Dong, Yue Xi

More Information
  • Received Date: 16 November 2022
    Accepted Date: 26 January 2023
    Published Online: 17 February 2023
    Tropical Plants  2 Article number: 2 (2023)  |  Cite this article
  • Discovery of the Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated (CRISPR/Cas) system and its repurposing into a powerful genome editing tool has revolutionized genome engineering and generated excitement for innovative breeding technology. CRISPR/Cas can perform genetic operations such as targeted insertion, deletion and replacement of genes in plants by its simple two-component system comprising a Cas protein and a guide RNA. Here, we focus on the recent advances in CRISPR/Cas technologies that are available for genome editing, among which Cas9, Cas12, and Cas13 systems are widely used in the areas of botany and agriculture. We also describe the new high-precision genome editing tools, base editors and prime editors that are derived from the Cas9 system and beyond for altering the genome in living cells, without generating double-stranded breaks in DNA or requiring a donor. In addition, we summarize the differences between the different CRISPR/Cas systems and their broad applications in plants. We also discuss the challenges facing the use of CRISPR/Cas technologies and the future directions of CRISPR/Cas systems in plant genome editing.
  • 加载中
  • [1]

    Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. 1987. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. Journal of Bacteriology 169:5429−33

    doi: 10.1128/jb.169.12.5429-5433.1987

    CrossRef   Google Scholar

    [2]

    Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, et al. 2007. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315:1709−12

    doi: 10.1126/science.1138140

    CrossRef   Google Scholar

    [3]

    Cong L, Ran FA, Cox D, Lin S, Barretto R, et al. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science 339:819−23

    doi: 10.1126/science.1231143

    CrossRef   Google Scholar

    [4]

    Mali P, Yang L, Esvelt KM, Aach J, Guell M, et al. 2013. RNA-guided human genome engineering via Cas9. Science 339:823−26

    doi: 10.1126/science.1232033

    CrossRef   Google Scholar

    [5]

    Jacinto FV, Link W, Ferreira BI. 2020. CRISPR/Cas9-mediated genome editing: From basic research to translational medicine. Journal of Cellular and Molecular Medicine 24:3766−78

    doi: 10.1111/jcmm.14916

    CrossRef   Google Scholar

    [6]

    Jolany Vangah S, Katalani C, Booneh HA, Hajizade A, Sijercic A, et al. 2020. CRISPR-based diagnosis of infectious and noninfectious diseases. Biological Procedures Online 22:22

    doi: 10.1186/s12575-020-00135-3

    CrossRef   Google Scholar

    [7]

    Joung J, Ladha A, Saito M, Segel M, Bruneau R, et al. 2020. Point-of-care testing for COVID-19 using SHERLOCK diagnostics. MedRxiv Pre-print

    doi: 10.1101/2020.05.04.20091231

    CrossRef   Google Scholar

    [8]

    Li JF, Norville JE, Aach J, McCormack M, Zhang D, et al. 2013. Multiplex and homologous recombination–mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nature Biotechnology 31:688−91

    doi: 10.1038/nbt.2654

    CrossRef   Google Scholar

    [9]

    Nekrasov V, Staskawicz B, Weigel D, Jones JDG, Kamoun S. 2013. Targeted mutagenesis in the model plant Nicotiana benthamiana using Cas9 RNA-guided endonuclease. Nature Biotechnology 31:691−93

    doi: 10.1038/nbt.2655

    CrossRef   Google Scholar

    [10]

    Shan Q, Wang Y, Li J, Zhang Y, Chen K, et al. 2013. Targeted genome modification of crop plants using a CRISPR-Cas system. Nature Biotechnology 31:686−88

    doi: 10.1038/nbt.2650

    CrossRef   Google Scholar

    [11]

    Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, et al. 2015. Cpf1 is a single RNA-guided endonuclease of a Class 2 CRISPR-Cas system. Cell 163:759−71

    doi: 10.1016/j.cell.2015.09.038

    CrossRef   Google Scholar

    [12]

    Zong Y, Wang Y, Li C, Zhang R, Chen K, et al. 2017. Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nature Biotechnology 35:438−40

    doi: 10.1038/nbt.3811

    CrossRef   Google Scholar

    [13]

    Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, et al. 2019. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149−57

    doi: 10.1038/s41586-019-1711-4

    CrossRef   Google Scholar

    [14]

    Wan D, Guo Y, Cheng Y, Hu Y, Xiao S, et al. 2020. CRISPR/Cas9-mediated mutagenesis of VvMLO3 results in enhanced resistance to powdery mildew in grapevine (Vitis vinifera). Horticulture Research 7:116

    doi: 10.1038/s41438-020-0339-8

    CrossRef   Google Scholar

    [15]

    Xing S, Chen K, Zhu H, Zhang R, Zhang H, et al. 2020. Fine-tuning sugar content in strawberry. Genome Biology 21:230

    doi: 10.1186/s13059-020-02146-5

    CrossRef   Google Scholar

    [16]

    Wada N, Osakabe K, Osakabe Y. 2022. Expanding the plant genome editing toolbox with recently developed CRISPR-Cas systems. Plant Physiology 188:1825−37

    doi: 10.1093/plphys/kiac027

    CrossRef   Google Scholar

    [17]

    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

    [18]

    Wiedenheft B. 2014. US, Patent No. WO2014093479-A1

    [19]

    Wright AV, Nuñez JK, Doudna JA. 2016. Biology and applications of CRISPR systems: Harnessing nature's toolbox for genome engineering. Cell 164:29−44

    doi: 10.1016/j.cell.2015.12.035

    CrossRef   Google Scholar

    [20]

    Chylinski K, Makarova KS, Charpentier E, Koonin EV. 2014. Classification and evolution of type II CRISPR-Cas systems. Nucleic Acids Research 42:6091−105

    doi: 10.1093/nar/gku241

    CrossRef   Google Scholar

    [21]

    Shmakov S, Smargon A, Scott D, Cox D, Pyzocha N, et al. 2017. Diversity and evolution of class 2 CRISPR-Cas systems. Nature Reviews Microbiology 15:169−82

    doi: 10.1038/nrmicro.2016.184

    CrossRef   Google Scholar

    [22]

    Jiang F, Doudna JA. 2017. CRISPR-Cas9 structures and mechanisms. Annual Review of Biophysics 46:505−29

    doi: 10.1146/annurev-biophys-062215-010822

    CrossRef   Google Scholar

    [23]

    Shmakov S, Abudayyeh OO, Makarova KS, Wolf YI, Gootenberg JS, et al. 2015. Discovery and functional characterization of diverse class 2 CRISPR-Cas systems. Molecular Cell 60:385−97

    doi: 10.1016/j.molcel.2015.10.008

    CrossRef   Google Scholar

    [24]

    Xue C, Greene EC. 2021. DNA repair pathway choices in CRISPR-Cas9-mediated genome editing. Trends in Genetics 37:639−56

    doi: 10.1016/j.tig.2021.02.008

    CrossRef   Google Scholar

    [25]

    Yan WX, Hunnewell P, Alfonse LE, Carte JM, Keston-Smith E, et al. 2019. Functionally diverse type V CRISPR-Cas systems. Science 363:88−91

    doi: 10.1126/science.aav7271

    CrossRef   Google Scholar

    [26]

    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

    [27]

    Zetsche B, Heidenreich M, Mohanraju P, Fedorova I, Kneppers J, et al. 2017. Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nature Biotechnology 35:31−34

    doi: 10.1038/nbt.3737

    CrossRef   Google Scholar

    [28]

    Teng F, Cui T, Feng G, Guo L, Xu K, et al. 2018. Repurposing CRISPR-Cas12b for mammalian genome engineering. Cell Discovery 4:63

    doi: 10.1038/s41421-018-0069-3

    CrossRef   Google Scholar

    [29]

    Strecker J, Jones S, Koopal B, Schmid-Burgk J, Zetsche B, et al. 2019. Engineering of CRISPR-Cas12b for human genome editing. Nature Communications 10:212

    doi: 10.1038/s41467-018-08224-4

    CrossRef   Google Scholar

    [30]

    Ming M, Ren Q, Pan C, He Y, Zhang Y, et al. 2020. CRISPR-Cas12b enables efficient plant genome engineering. Nature Plants 6:202−8

    doi: 10.1038/s41477-020-0614-6

    CrossRef   Google Scholar

    [31]

    Selkova P, Vasileva A, Pobegalov G, Musharova O, Arseniev A, et al. 2020. Position of Deltaproteobacteria Cas12e nuclease cleavage sites depends on spacer length of guide RNA. RNA Biology 17:1472−79

    doi: 10.1080/15476286.2020.1777378

    CrossRef   Google Scholar

    [32]

    Burstein D, Harrington LB, Strutt SC, Probst AJ, Anantharaman K, et al. 2017. New CRISPR-Cas systems from uncultivated microbes. Nature 542:237−41

    doi: 10.1038/nature21059

    CrossRef   Google Scholar

    [33]

    Li Z, Zhang H, Xiao R, Han R, Chang L. 2021. Cryo-EM structure of the RNA-guided ribonuclease Cas12g. Nature Chemical Biology 17:387−93

    doi: 10.1038/s41589-020-00721-2

    CrossRef   Google Scholar

    [34]

    Karvelis T, Bigelyte G, Young JK, Hou Z, Zedaveinyte R, et al. 2020. PAM recognition by miniature CRISPR–Cas12f nucleases triggers programmable double-stranded DNA target cleavage. Nucleic Acids Research 48:5016−23

    doi: 10.1093/nar/gkaa208

    CrossRef   Google Scholar

    [35]

    Wu Z, Zhang Y, Yu H, Pan D, Wang Y, et al. 2021. Programmed genome editing by a miniature CRISPR-Cas12f nuclease. Nature Chemical Biology 17:1132−38

    doi: 10.1038/s41589-021-00868-6

    CrossRef   Google Scholar

    [36]

    Huang CJ, Adler BA, Doudna JA. 2022. A naturally DNase-free CRISPR-Cas12c enzyme silences gene expression. Molecular Cell 82:2148−2160.E4

    doi: 10.1016/j.molcel.2022.04.020

    CrossRef   Google Scholar

    [37]

    East-Seletsky A, O'Connell MR, Knight SC, Burstein D, Cate JH, et al. 2016. Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature 538:270−73

    doi: 10.1038/nature19802

    CrossRef   Google Scholar

    [38]

    Pickar-Oliver A, Gersbach CA. 2019. The next generation of CRISPR-Cas technologies and applications. Nature Reviews Molecular Cell Biology 20:490−507

    doi: 10.1038/s41580-019-0131-5

    CrossRef   Google Scholar

    [39]

    Cox DBT, Gootenberg JS, Abudayyeh OO, Franklin B, Kellner MJ, et al. 2017. RNA editing with CRISPR-Cas13. Science 358:1019−27

    doi: 10.1126/science.aaq0180

    CrossRef   Google Scholar

    [40]

    Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J, et al. 2017. RNA targeting with CRISPR-Cas13. Nature 550:280−84

    doi: 10.1038/nature24049

    CrossRef   Google Scholar

    [41]

    Aman R, Ali Z, Butt H, Mahas A, Aljedaani F, et al. 2018. RNA virus interference via CRISPR/Cas13a system in plants. Genome Biology 19:1

    doi: 10.1186/s13059-017-1381-1

    CrossRef   Google Scholar

    [42]

    Mohanraju P, Makarova KS, Zetsche B, Zhang F, Koonin EV, van der Oost J. 2016. Diverse evolutionary roots and mechanistic variations of the CRISPR-Cas systems. Science 353:aad5147

    doi: 10.1126/science.aad5147

    CrossRef   Google Scholar

    [43]

    Csörgő B, León LM, Chau-Ly IJ, Vasquez-Rifo A, Berry JD, et al. 2020. A compact Cascade-Cas3 system for targeted genome engineering. Nature methods 17:1183−90

    doi: 10.1038/s41592-020-00980-w

    CrossRef   Google Scholar

    [44]

    Young JK, Gasior SL, Jones S, Wang L, Navarro P, et al. 2019. The repurposing of type I-E CRISPR-Cascade for gene activation in plants. Communications Biology 2:383

    doi: 10.1038/s42003-019-0637-6

    CrossRef   Google Scholar

    [45]

    Osakabe K, Wada N, Miyaji T, Murakami E, Marui K, et al. 2020. Genome editing in plants using CRISPR type I-D nuclease. Communications Biology 3:648

    doi: 10.1038/s42003-020-01366-6

    CrossRef   Google Scholar

    [46]

    Walker FC, Chou-Zheng L, Dunkle JA, Hatoum-Aslan A. 2017. Molecular determinants for CRISPR RNA maturation in the Cas10-Csm complex and roles for non-Cas nucleases. Nucleic Acids Research 45:2112−23

    doi: 10.1093/nar/gkw891

    CrossRef   Google Scholar

    [47]

    Bari SMN, Walker FC, Cater K, Aslan B, Hatoum-Aslan A. 2017. Strategies for editing virulent staphylococcal phages using CRISPR-Cas10. ACS Synthetic Biology 6:2316−25

    doi: 10.1021/acssynbio.7b00240

    CrossRef   Google Scholar

    [48]

    Harrington LB, Burstein D, Chen JS, Paez-Espino D, Ma E, et al. 2018. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science 362:839−42

    doi: 10.1126/science.aav4294

    CrossRef   Google Scholar

    [49]

    Khan MZ, Haider S, Mansoor S, Amin I. 2019. Targeting plant ssDNA viruses with engineered miniature CRISPR-Cas14a. Trends in Biotechnology 37:800−4

    doi: 10.1016/j.tibtech.2019.03.015

    CrossRef   Google Scholar

    [50]

    Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT, et al. 2015. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523:481−5

    doi: 10.1038/nature14592

    CrossRef   Google Scholar

    [51]

    Kim HK, Lee S, Kim Y, Park J, Min S, et al. 2020. High-throughput analysis of the activities of xCas9, SpCas9-NG and SpCas9 at matched and mismatched target sequences in human cells. Nature Biomedical Engineering 4:111−24

    doi: 10.1038/s41551-019-0505-1

    CrossRef   Google Scholar

    [52]

    Hu X, Meng X, Liu Q, Li J, Wang K. 2018. Increasing the efficiency of CRISPR-Cas9-VQR precise genome editing in rice. Plant Biotechnology Journal 16:292−97

    doi: 10.1111/pbi.12771

    CrossRef   Google Scholar

    [53]

    Nishimasu H, Shi X, Ishiguro S, Gao L, Hirano S, et al. 2018. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 361:1259−62

    doi: 10.1126/science.aas9129

    CrossRef   Google Scholar

    [54]

    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

    [55]

    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

    [56]

    Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK. 2014. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nature Biotechnology 32:279−84

    doi: 10.1038/nbt.2808

    CrossRef   Google Scholar

    [57]

    Kocak DD, Josephs EA, Bhandarkar V, Adkar SS, Kwon JB, et al. 2019. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nature Biotechnology 37:657−66

    doi: 10.1038/s41587-019-0095-1

    CrossRef   Google Scholar

    [58]

    Ryan DE, Taussig D, Steinfeld I, Phadnis SM, Lunstad BD, et al. 2018. Improving CRISPR-Cas specificity with chemical modifications in single-guide RNAs. Nucleic Acids Research 46:792−803

    doi: 10.1093/nar/gkx1199

    CrossRef   Google Scholar

    [59]

    Bolukbasi MF, Gupta A, Oikemus S, Derr AG, Garber M, et al. 2015. DNA-binding-domain fusions enhance the targeting range and precision of Cas9. Nature Methods 12:1150−56

    doi: 10.1038/nmeth.3624

    CrossRef   Google Scholar

    [60]

    Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, et al. 2016. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529:490−95

    doi: 10.1038/nature16526

    CrossRef   Google Scholar

    [61]

    Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, et al. 2016. Rationally engineered Cas9 nucleases with improved specificity. Science 351:84−88

    doi: 10.1126/science.aad5227

    CrossRef   Google Scholar

    [62]

    Heigwer F, Kerr G, Boutros M. 2014. E-CRISP: fast CRISPR target site identification. Nature Methods 11:122−3

    doi: 10.1038/nmeth.2812

    CrossRef   Google Scholar

    [63]

    Lei Y, Lu L, Liu H, Li S, Xing F, et al. 2014. CRISPR-P: a web tool for synthetic single-guide RNA design of CRISPR-system in plants. Molecular Plant 7:1494−96

    doi: 10.1093/mp/ssu044

    CrossRef   Google Scholar

    [64]

    Haeussler M, Schönig K, Eckert H, Eschstruth A, Mianné J, et al. 2016. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biology 17:148

    doi: 10.1186/s13059-016-1012-2

    CrossRef   Google Scholar

    [65]

    Aquino-Jarquin G. 2021. Current advances in overcoming obstacles of CRISPR/Cas9 off-target genome editing. Molecular Genetics and Metabolism 134:77−86

    doi: 10.1016/j.ymgme.2021.08.002

    CrossRef   Google Scholar

    [66]

    Manghwar H, Li B, Ding X, Hussain A, Lindsey K, et al. 2020. CRISPR/Cas systems in genome editing: Methodologies and tools for sgRNA design, off-target evaluation, and strategies to mitigate off-target effects. Advanced Science 7:1902312

    doi: 10.1002/advs.201902312

    CrossRef   Google Scholar

    [67]

    Tang T, Yu X, Yang H, Gao Q, Ji H, et al. 2018. Development and validation of an effective CRISPR/Cas9 vector for efficiently isolating positive transformants and transgene-free mutants in a wide range of plant species. Frontiers in Plant Science 9:1533

    doi: 10.3389/fpls.2018.01533

    CrossRef   Google Scholar

    [68]

    Mao Y, Botella JR, Liu Y, Zhu J. 2019. Gene editing in plants: progress and challenges. National Science Review 6:421−37

    doi: 10.1093/nsr/nwz005

    CrossRef   Google Scholar

    [69]

    Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. 2016. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420−24

    doi: 10.1038/nature17946

    CrossRef   Google Scholar

    [70]

    Hua K, Han P, Zhu JK. 2022. Improvement of base editors and prime editors advances precision genome engineering in plants. Plant Physiology 188:1795−810

    doi: 10.1093/plphys/kiab591

    CrossRef   Google Scholar

    [71]

    Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, et al. 2017. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551:464−71

    doi: 10.1038/nature24644

    CrossRef   Google Scholar

    [72]

    Rees HA, Liu DR. 2018. Base editing: precision chemistry on the genome and transcriptome of living cells. Nature Reviews Genetics 19:770−88

    doi: 10.1038/s41576-018-0059-1

    CrossRef   Google Scholar

    [73]

    Zhang X, Zhu B, Chen L, Xie L, Yu W, et al. 2020. Dual base editor catalyzes both cytosine and adenine base conversions in human cells. Nature Biotechnology 38:856−60

    doi: 10.1038/s41587-020-0527-y

    CrossRef   Google Scholar

    [74]

    Xie J, Huang X, Wang X, Gou S, Liang Y, et al. 2020. ACBE, a new base editor for simultaneous C-to-T and A-to-G substitutions in mammalian systems. BMC Biology 18:131

    doi: 10.1186/s12915-020-00866-5

    CrossRef   Google Scholar

    [75]

    Li C, Zhang R, Meng X, Chen S, Zong Y, et al. 2020. Targeted, random mutagenesis of plant genes with dual cytosine and adenine base editors. Nature Biotechnology 38:875−82

    doi: 10.1038/s41587-019-0393-7

    CrossRef   Google Scholar

    [76]

    Kurt IC, Zhou R, Iyer S, Garcia SP, Miller BR, et al. 2021. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nature Biotechnology 39:41−46

    doi: 10.1038/s41587-020-0609-x

    CrossRef   Google Scholar

    [77]

    Zhao D, Li J, Li S, Xin X, Hu M, et al. 2021. Glycosylase base editors enable C-to-A and C-to-G base changes. Nature Biotechnology 39:35−40

    doi: 10.1038/s41587-020-0592-2

    CrossRef   Google Scholar

    [78]

    Liang Y, Xie J, Zhang Q, Wang X, Gou S, et al. 2022. AGBE: a dual deaminase-mediated base editor by fusing CGBE with ABE for creating a saturated mutant population with multiple editing patterns. Nucleic Acids Researh 50:5384−99

    doi: 10.1093/nar/gkac353

    CrossRef   Google Scholar

    [79]

    Abudayyeh OO, Gootenberg JS, Franklin B, Koob J, Kellner MJ, et al. 2019. A cytosine deaminase for programmable single-base RNA editing. Science 365:382−86

    doi: 10.1126/science.aax7063

    CrossRef   Google Scholar

    [80]

    Shimatani Z, Kashojiya S, Takayama M, Terada R, Arazoe T, et al. 2017. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nature Biotechnology 35:441−43

    doi: 10.1038/nbt.3833

    CrossRef   Google Scholar

    [81]

    Lu Y, Zhu J. 2017. Precise editing of a target base in the rice genome using a modified CRISPR/Cas9 system. Molecular Plant 10:523−25

    doi: 10.1016/j.molp.2016.11.013

    CrossRef   Google Scholar

    [82]

    Li J, Sun Y, Du J, Zhao Y, Xia L. 2017. Generation of Targeted Point Mutations in Rice by a Modified CRISPR/Cas9 System. Molecular Plant 10:526−29

    doi: 10.1016/j.molp.2016.12.001

    CrossRef   Google Scholar

    [83]

    Zuo E, Sun Y, Wei W, Yuan T, Ying W, et al. 2019. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364:289−92

    doi: 10.1126/science.aav9973

    CrossRef   Google Scholar

    [84]

    Jin S, Zong Y, Gao Q, Zhu Z, Wang Y, et al. 2019. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 364:292−95

    doi: 10.1126/science.aaw7166

    CrossRef   Google Scholar

    [85]

    Porto EM, Komor AC, Slaymaker IM, Yeo GW. 2020. Base editing: advances and therapeutic opportunities. Nature Reviews Drug Discovery 19:839−59

    doi: 10.1038/s41573-020-0084-6

    CrossRef   Google Scholar

    [86]

    Liu Y, Zhou J, Lan T, Zhou X, Yang Y, et al. 2022. Elimination of Cas9-dependent off-targeting of adenine base editor by using TALE to separately guide deaminase to target sites. Cell Discovery 8:28

    doi: 10.1038/s41421-022-00384-4

    CrossRef   Google Scholar

    [87]

    Lin Q, Zong Y, Xue C, Wang S, Jin S, et al. 2020. Prime genome editing in rice and wheat. Nature Biotechnology 38:582−85

    doi: 10.1038/s41587-020-0455-x

    CrossRef   Google Scholar

    [88]

    Tang X, Sretenovic S, Ren Q, Jia X, Li M, et al. 2020. Plant Prime Editors Enable Precise Gene Editing in Rice Cells. Molecular Plant 13:667−70

    doi: 10.1016/j.molp.2020.03.010

    CrossRef   Google Scholar

    [89]

    Li H, Li J, Chen J, Yan L, Xia L. 2020. Precise modifications of both exogenous and endogenous genes in rice by prime editing. Molecular Plant 13:671−74

    doi: 10.1016/j.molp.2020.03.011

    CrossRef   Google Scholar

    [90]

    Lin Q, Jin S, Zong Y, Yu H, Zhu Z, et al. 2021. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nature Biotechnology 39:923−7

    doi: 10.1038/s41587-021-00868-w

    CrossRef   Google Scholar

    [91]

    Nelson JW, Randolph PB, Shen SP, Everette KA, Chen PJ, et al. 2022. Engineered pegRNAs improve prime editing efficiency. Nature Biotechnology 40:402−10

    doi: 10.1038/s41587-021-01039-7

    CrossRef   Google Scholar

    [92]

    Liu Y, Yang G, Huang S, Li X, Wang X, et al. 2021. Enhancing prime editing by Csy4-mediated processing of pegRNA. Cell Research 31:1134−36

    doi: 10.1038/s41422-021-00520-x

    CrossRef   Google Scholar

    [93]

    Chen PJ, Hussmann JA, Yan J, Knipping F, Ravisankar P, et al. 2021. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 184:5635−5652.E29

    doi: 10.1016/j.cell.2021.09.018

    CrossRef   Google Scholar

    [94]

    Xu W, Yang Y, Yang B, Krueger CJ, Xiao Q, et al. 2022. A design optimized prime editor with expanded scope and capability in plants. Nature Plants 8:45−52

    doi: 10.1038/s41477-021-01043-4

    CrossRef   Google Scholar

    [95]

    Zong Y, Liu Y, Xue C, Li B, Li X, et al. 2022. An engineered prime editor with enhanced editing efficiency in plants. Nature Biotechnology 40:1394−402

    doi: 10.1038/s41587-022-01254-w

    CrossRef   Google Scholar

    [96]

    Zou J, Meng X, Liu Q, Shang M, Wang K, et al. 2022. Improving the efficiency of prime editing with epegRNAs and high-temperature treatment in rice. Science China Life sciences 65:2328−31

    doi: 10.1007/s11427-022-2147-2

    CrossRef   Google Scholar

    [97]

    Gao C. 2021. Genome engineering for crop improvement and future agriculture. Cell 184:1621−35

    doi: 10.1016/j.cell.2021.01.005

    CrossRef   Google Scholar

    [98]

    Hui S, Li H, Mawia AM, Zhou L, Cai J, et al. 2022. Production of aromatic three-line hybrid rice using novel alleles of BADH2. Plant Biotechnology Journal 20:59−74

    doi: 10.1111/pbi.13695

    CrossRef   Google Scholar

    [99]

    Wang F, Wang C, Liu P, Lei C, Hao W, et al. 2016. Enhanced rice blast resistance by CRISPR/Cas9-targeted mutagenesis of the ERF transcription factor gene OsERF922. PLoS One 11:e0154027

    doi: 10.1371/journal.pone.0154027

    CrossRef   Google Scholar

    [100]

    Lu Y, Wang J, Chen B, Mo S, Lian L, et al. 2021. A donor-DNA-free CRISPR/Cas-based approach to gene knock-up in rice. Nature Plants 7:1445−52

    doi: 10.1038/s41477-021-01019-4

    CrossRef   Google Scholar

    [101]

    Zhang P, Du H, Wang J, Pu Y, Yang C, et al. 2020. Multiplex CRISPR/Cas9-mediated metabolic engineering increases soya bean isoflavone content and resistance to soya bean mosaic virus. Plant Biotechnology Journal 18:1384−95

    doi: 10.1111/pbi.13302

    CrossRef   Google Scholar

    [102]

    Lorenzo CD, Debray K, Herwegh D, Develtere W, Impens L, et al. 2022. BREEDIT: a multiplex genome editing strategy to improve complex quantitative traits in maize. The Plant Cell 35:218−38

    doi: 10.1093/plcell/koac243

    CrossRef   Google Scholar

    [103]

    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

    [104]

    Nekrasov V, Wang C, Win J, Lanz C, Weigel D, Kamoun S. 2017. Rapid generation of a transgene-free powdery mildew resistant tomato by genome deletion. Scientific Reports 7:482

    doi: 10.1038/s41598-017-00578-x

    CrossRef   Google Scholar

    [105]

    Tripathi JN, Ntui VO, Shah T, Tripathi L. 2021. CRISPR/Cas9-mediated editing of DMR6 orthologue in banana (Musa spp.) confers enhanced resistance to bacterial disease. Plant Biotechnology Journal 19:1291−93

    doi: 10.1111/pbi.13614

    CrossRef   Google Scholar

    [106]

    Dong L, Li L, Liu C, Liu C, Geng S, et al. 2018. Genome editing and double-fluorescence proteins enable robust maternal haploid induction and identification in maize. Molecular Plant 11:1214−17

    doi: 10.1016/j.molp.2018.06.011

    CrossRef   Google Scholar

    [107]

    Yao L, Zhang Y, Liu C, Liu Y, Wang Y, et al. 2018. OsMATL mutation induces haploid seed formation in indica rice. Nature Plants 4:530−33

    doi: 10.1038/s41477-018-0193-y

    CrossRef   Google Scholar

    [108]

    Li X, Zhou W, Ren Y, Tian X, Lv T, et al. 2017. High-efficiency breeding of early-maturing rice cultivars via CRISPR/Cas9-mediated genome editing. Journal of Genetics and Genomics 44:175−78

    doi: 10.1016/j.jgg.2017.02.001

    CrossRef   Google Scholar

    [109]

    Gomez MA, Lin ZD, Moll T, Chauhan RD, Hayden L, et al. 2019. Simultaneous CRISPR/Cas9-mediated editing of cassava eIF4E isoforms nCBP-1 and nCBP-2 reduces cassava brown streak disease symptom severity and incidence. Plant Biotechnology Journal 17:421−34

    doi: 10.1111/pbi.12987

    CrossRef   Google Scholar

    [110]

    Braatz J, Harloff HJ, Mascher M, Stein N, Himmelbach A, Jung C. 2017. CRISPR-Cas9 targeted mutagenesis leads to simultaneous modification of different homoeologous gene copies in polyploid oilseed rape (Brassica napus). Plant Physiology 174:935−42

    doi: 10.1104/pp.17.00426

    CrossRef   Google Scholar

    [111]

    Ye M, Peng Z, Tang D, Yang Z, Li D, et al. 2018. Generation of self-compatible diploid potato by knockout of S-RNase. Nature Plants 4:651−4

    doi: 10.1038/s41477-018-0218-6

    CrossRef   Google Scholar

    [112]

    Zsögön A, Čermák T, Naves ER, Notini MM, Edel KH, et al. 2018. De novo domestication of wild tomato using genome editing. Nature Biotechnology 36:1211−16

    doi: 10.1038/nbt.4272

    CrossRef   Google Scholar

    [113]

    Lemmon ZH, Reem NT, Dalrymple J, Soyk S, Swartwood KE, et al. 2018. Rapid improvement of domestication traits in an orphan crop by genome editing. Nature Plants 4:766−70

    doi: 10.1038/s41477-018-0259-x

    CrossRef   Google Scholar

    [114]

    Yu H, Lin T, Meng X, Du H, Zhang J, et al. 2021. A route to de novo domestication of wild allotetraploid rice. Cell 184:1156−1170.E14

    doi: 10.1016/j.cell.2021.01.013

    CrossRef   Google Scholar

    [115]

    Schmitz RJ. 2014. Genetics. The secret garden—epigenetic alleles underlie complex traits. Science 343:1082−83

    doi: 10.1126/science.1251864

    CrossRef   Google Scholar

    [116]

    Miryeganeh M, Saze H. 2020. Epigenetic inheritance and plant evolution. Population Ecology 62:17−27

    doi: 10.1002/1438-390X.12018

    CrossRef   Google Scholar

    [117]

    Giordano A, Santo Domingo M, Quadrana L, Pujol M, Martín-Hernández AM, et al. 2022. CRISPR/Cas9 gene editing uncovers the roles of CONSTITUTIVE TRIPLE RESPONSE 1 and REPRESSOR OF SILENCING 1 in melon fruit ripening and epigenetic regulation. Journal of Experimental Botany 73:4022−33

    doi: 10.1093/jxb/erac148

    CrossRef   Google Scholar

    [118]

    Pan C, Sretenovic S, Qi Y. 2021. CRISPR/dCas-mediated transcriptional and epigenetic regulation in plants. Current Opinion in Plant Biology 60:101980

    doi: 10.1016/j.pbi.2020.101980

    CrossRef   Google Scholar

    [119]

    Liu X, Wu H, Ji X, Stelzer Y, Wu X, et al. 2016. Editing DNA Methylation in the Mammalian Genome. Cell 167:233−247.E17

    doi: 10.1016/j.cell.2016.08.056

    CrossRef   Google Scholar

    [120]

    Nuñez JK, Chen J, Pommier GC, Cogan JZ, Replogle JM, et al. 2021. Genome-wide programmable transcriptional memory by CRISPR-based epigenome editing. Cell 184:2503−2519.E17

    doi: 10.1016/j.cell.2021.03.025

    CrossRef   Google Scholar

    [121]

    Papikian A, Liu W, Gallego-Bartolome J, Jacobsen SE. 2019. Site-specific manipulation of Arabidopsis loci using CRISPR-Cas9 SunTag systems. Nature Communications 10:729

    doi: 10.1038/s41467-019-08736-7

    CrossRef   Google Scholar

    [122]

    Lee JE, Neumann M, Duro DI, Schmid M. 2019. CRISPR-based tools for targeted transcriptional and epigenetic regulation in plants. PloS one 14:e0222778

    doi: 10.1371/journal.pone.0222778

    CrossRef   Google Scholar

    [123]

    Tadić V, Josipović G, Zoldoš V, Vojta A. 2019. CRISPR/Cas9-based epigenome editing: An overview of dCas9-based tools with special emphasis on off-target activity. Methods 164-165:109−19

    doi: 10.1016/j.ymeth.2019.05.003

    CrossRef   Google Scholar

    [124]

    Qiu X, Zhu L, Zhu C, Ma J, Hou T, et al. 2018. Highly effective and low-cost MicroRNA detection with CRISPR-Cas9. ACS Synthetic Biology 7:807−13

    doi: 10.1021/acssynbio.7b00446

    CrossRef   Google Scholar

    [125]

    Wheatley MS, Yang Y. 2021. Versatile applications of the CRISPR/Cas toolkit in plant pathology and disease management. Phytopathology 111:1080−90

    doi: 10.1094/PHYTO-08-20-0322-IA

    CrossRef   Google Scholar

    [126]

    Zhang D, Guo J. 2011. The development and standardization of testing methods for genetically modified organisms and their derived products. Journal of Integrative Plant Biology 53:539−51

    doi: 10.1111/j.1744-7909.2011.01060.x

    CrossRef   Google Scholar

    [127]

    Abudayyeh OO, Gootenberg JS, Kellner MJ, Zhang F. 2019. Nucleic Acid Detection of Plant Genes Using CRISPR-Cas13. The CRISPR Journal 2:165−71

    doi: 10.1089/crispr.2019.0011

    CrossRef   Google Scholar

    [128]

    Mahas A, Hassan N, Aman R, Marsic T, Wang Q, et al. 2021. LAMP-coupled CRISPR-Cas12a module for rapid and sensitive detection of plant DNA viruses. Viruses 13:466

    doi: 10.3390/v13030466

    CrossRef   Google Scholar

    [129]

    Wheatley MS, Wang Q, Wei W, Bottner-Parker KD, Zhao Y, Yang Y. 2022. Cas12a-based diagnostics for potato purple top disease complex associated with infection by 'Candidatus Phytoplasma trifolii'-related strains. Plant Disease 106:2039−45

    doi: 10.1094/PDIS-09-21-2119-RE

    CrossRef   Google Scholar

    [130]

    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

    [131]

    Kunling CHEN CG. 2020. Genome-edited crops: how to move them from laboratory to market. Frontiers of Agricultural Science and Engineering 7:181−7

    doi: 10.15302/j-fase-2020332

    CrossRef   Google Scholar

  • Cite this article

    Dong C, Xi Y, Satheesh V, Lei M. 2023. Advances in CRISPR/Cas technologies and their application in plants. Tropical Plants 2:2 doi: 10.48130/TP-2023-0002
    Dong C, Xi Y, Satheesh V, Lei M. 2023. Advances in CRISPR/Cas technologies and their application in plants. Tropical Plants 2:2 doi: 10.48130/TP-2023-0002

Figures(3)  /  Tables(2)

Article Metrics

Article views(3381) PDF downloads(443)

Other Articles By Authors

REVIEW   Open Access    

Advances in CRISPR/Cas technologies and their application in plants

Tropical Plants  2 Article number: 2  (2023)  |  Cite this article

Abstract: Discovery of the Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated (CRISPR/Cas) system and its repurposing into a powerful genome editing tool has revolutionized genome engineering and generated excitement for innovative breeding technology. CRISPR/Cas can perform genetic operations such as targeted insertion, deletion and replacement of genes in plants by its simple two-component system comprising a Cas protein and a guide RNA. Here, we focus on the recent advances in CRISPR/Cas technologies that are available for genome editing, among which Cas9, Cas12, and Cas13 systems are widely used in the areas of botany and agriculture. We also describe the new high-precision genome editing tools, base editors and prime editors that are derived from the Cas9 system and beyond for altering the genome in living cells, without generating double-stranded breaks in DNA or requiring a donor. In addition, we summarize the differences between the different CRISPR/Cas systems and their broad applications in plants. We also discuss the challenges facing the use of CRISPR/Cas technologies and the future directions of CRISPR/Cas systems in plant genome editing.

    • CRISPR/Cas systems have emerged as one of the most popular and advanced tools for genome editing. As early as in 1987, CRISPR sequences were first discovered in the iap gene of Escherichia coli, which functions in providing resistance against bacteriophages and enhancing adaptive immune responses in prokaryotes[1,2]. Repurposing CRISPR/Cas systems as a gene editing tool in eukaryotic cells has revolutionized the field of genome engineering[3,4]. This technology has been rapidly expanding and applied to form translational medicine, diagnostics and therapeutics to other fields[57].

      In 2013, the CRISPR/Cas9 system was first established in Arabidopsis thaliana[8], Nicotiana benthamiana[9], rice (Oryza sativa) and wheat (Triticum aestivum)[10] showing prospects of wider application of CRISPR/Cas technologies in plants. Since then, continuous improvements in CRISPR/Cas systems, including CRISPR/Cpf1[11], base editors[12] and prime editors[13], have made plant genome editing easier and they have become more widely used as well as being low-cost and highly efficienct precision genetic manipulation tools. An ever-increasing number of crops and other plant species have been modified by genome editing for gene functional validation, trait improvement, and plant virus detection, among other uses[1416]. CRISPR/Cas technologies thus have the potential to change the landscape for conventional breeding by aiding the introduction of target traits more accurately, in less time. More importantly, CRISPR/Cas differs from transgenic breeding technology in the fact that there is no transfer of exogenous genes into the target genome, paving the way to reduced biosafety concerns. Here, we describe a series of CRISPR/Cas systems and summarize the recent advances in those systems and their applications in plant genome editing.

    • CRISPR/Cas systems exist in various species of bacteria and archaea providing adaptive immunity against viruses, and they vary in composition and mechanisms of action. The defense process is mainly accomplished by Cas proteins and CRISPR array through three sequential steps: adaptation, expression and interference[17]. CRISPR systems have been divided into two classes that were additionally grouped into six types based on the easily recognizable signature of Cas proteins[18,19] (Fig. 1). Typically, class 1 CRISPR/Cas systems (comprising types I, III, and IV; represent about 90% of the CRISPR-Cas loci) employ multi-subunit protein complexes to target DNA or RNA for processing and cleavage. By contrast, class 2 systems (comprising types II, V, and VI; represent about 10% of the CRISPR-Cas loci) employ a single multidomain protein in complex with crRNAs or alternative tracrRNA[20,21]. The extensive diversification of the CRISPR/Cas systems offers potential expansion of the gene editing toolbox from the original single system CRISPR/Cas9 to a rich toolbox.

      Figure 1. 

      The classification of CRISPR/Cas systems. Representative operons for each type are shown here. Gray represents Cas systems employing multi-subunit protein complexes to cut target DNA or RNA; blue represents Cas employing a single multidomain protein in complex for sequence cleavage.

    • CRISPR/Cas9 system belongs to the type II of class 2 CRISPR systems and is the most well-characterized, most popular and widely used system for genome editing in a number of organisms[20,22]. Among the CRISPR/Cas systems, the Cas9 protein was the first used system to introduce DNA double-strand break (DSB) in target DNA by an RNA duplex formed by crRNA and tracrRNA[17]. Cas9 nuclease contains two domains RuvC-like and HNH, which cleave the non-complementary and complementary strands of the target DNA, respectively[23] (Fig. 2). The dual RNA, crRNA and tracrRNA, designed a single guide RNA (sgRNA) together with the Cas9 protein which makes up the simplified two-component system. The binding of sgRNA by Cas9 in cells results in the formation of a ribonucleoprotein complex, which is then guided by 20 nt sgRNA to bind to protospacer adjacent motif (PAM, 5'-NGG-3') that achieves cleavage of target DNA. There are two major pathways for cellular DNA repair for DSBs caused by DNA cleavage[24]. One is homologous recombination (HR) that recombines and integrates donor DNA into the position of DSBs, and the second is non-homologous end joining (NHEJ) that is error-prone to induce insertion, deletion or substitution of individual bases. Under natural conditions in eukaryotic cells, HR is of low probability, whereas NHEJ happens more often. Therefore, gene modification and genome editing can be achieved through DSBs and subsequent DNA repair.

      Figure 2. 

      Composition of CRISPR/Cas system. CRISPR/Cas9, the Cas9 system is composed of a Cas9 protein and a sgRNA. The yellow arrow represents DNA double-strand break (DSB) induced by RuvC and HNH domains. CBE, a base editor is a fusion of catalytically deactivated Cas9 domain and deaminase domain. CBE deaminates cytosine to generate uracil (C-U), then converts the uracil into thymine (U-T) by DNA repair response, eventually effecting a C-T (or G-A) substitution. ABE deaminates adenosine to generate inosine (A-I), which is read as guanosine (G) by polymerase enzymes, eventually effecting a A-G (or T-C) substitution through DNA repair or replication. PE, Prime editor consist of two simple operational components, a fusion protein made by the fusion nickase Cas9 protein (nCas9) and reverse transcriptase (RT) and a pegRNA contains sgRNA, primer binding site and reverse transcription template.

    • The characteristics of Cas12 is similar to Cas9 with the difference that Cas12 proteins have multiple domains, but contain a single RuvC-like endonuclease domain[25]. Cas12a system, also known as Cpf1 because of its discovery in Prevotella and Francisella 1, belongs to type V-A of class 2[11]. Compared to recognizing to G-rich PAMs by the Cas9 system, Cas12a induces target DNA cleavage preferably by binding to 5′-TTTN-3′ (T-rich) PAMs (Table 1). Additionally, Cas12a cleaves target DNA using Cas12a-crRNA complex without the requirement of tracrRNA that produces a staggered DSB (sticky ends) compared to blunt ends produced by Cas9[11,26]. This unique ability in crRNA processing has been utilized for multiplex gene editing[27]. Thus, the discovery of Cas12a systems expands the CRISPR toolbox providing an alternative genome editing tool for other application scenarios.

      Table 1.  Comparative analysis between Cas9, Cas12, and Cas13.

      Cas9Cas12Cas13
      Class222
      TypeIIVVI
      SubtypesII-A,B,CV-A(Cas12a); V-B(Cas12b); V-C(C2c3)VI-A(Cas13a/C2c2); VI-B(Cas13b); VI-C(Cas13c);
      VI-D (Cas13d)
      TargetsdsDNAssDNARNA
      Nuclease domain(s)RuvC and HNHRuvCTwo HEPN
      Guide RNAcrRNA and tracrRNAcrRNAcrRNA
      Cleavage typeBlunt ended dsDNA break5′ overhang dsDNA breakssRNA
      Size1000−1600aa1100−1300aa900−1300aa
      Guide spacer18−24nt18−25nt~64nt
      PAMNGG (SpCas9)TTTN (FnCas12a)3′ non-G PFS (LasCas13a)
      NGN (SpCas9-NG)TYCV (LbCas12a-RR)D-(PS)-NAN/NNA (BzCas13b)
      NNG (ScCas9)TATV (LbCas12a-RVR)
      NNGRRT (SaCas9)
      NNNNGATT (NmCas9)
      NNNVRYM (CjCas9)
      NNAGAAW (StCas9)
      PAM-less (SpRY)
      Repair mechanismNHEJ and HDRNHEJ and HDRNo
      ApplicationsMost widely usedMajorly used in epigenetic editingMajorly used in disease detection and diagnostics

      In addition to the Cas12a system, the variants of Cas12, AaCas12b, BhCas12b, AacCas12b and BthCas12b derived from different bacteria, have been harnessed and engineered for editing in mammalian genomes and plant genomes with high targeting specificity[2830]. Cas12e (also known as CasX) derived from Deltaproteobacteria (DbCas12e) was proven to produce extensive 5ʹ-overhangs at cleaved targets in mammalian genomes[31]. Cas12d (also known as CasY) recognizes a 5ʹ-TA PAM and cleaves dsDNA[32]. Cas12g, smaller than other Cas12 effectors that only contain ~800 amino acids, is an RNA-guided ribonuclease with collateral RNase and single-strand DNase activities[25,33]. The characteristic structures of Cas12h and Cas12i are similar to that of Cas12a in that they use a single crRNA to target dsDNA without tracrRNA[33]. Cas12f is a miniature CRISPR/Cas system that cleaves dsDNA rely on PAM recognition, such as AsCas12f1 (422 amino acids), which recognizes 5ʹ T-rich PAMs and create staggered DSB to target DNA[34,35]. The most recent report found that Cas12c directly induces DSB in a unique mechanism without DNA cutting, which may be repurposed to represses transcription[36].

    • The Cas13 system, which belongs to type VI of class 2, is further classified into Cas13a (also known as C2c2), Cas13b, Cas13c and Cas13d[21]. Cas13 employs a single crRNA to cleave target single-strand RNA by its two HEPN domains (Table 1). Furthermore, Cas13 has two distinct catalytic activities; an RNA recognition and a cleavage activity[37,38]. These characteristics make Cas13 a versatile RNA editing tool and differs from other DNA manipulation tools. RNA Editing for Programmable A to I Replacement (REPAIR) platform was designed to edit nucleic acids at the RNA level in mammalian cells using the complex obtained by fusing adenosine deaminase acting on RNA type 2 (ADAR2) with catalytically-inactive Cas13 (dCas13)[39]. Cas13 orthologs, such as LwaCas13a, PspCas13b and LshCas13a, have been engineered for RNA knockdown and binding[40]. Among them, LshCas13a was optimized through plant codon and had been engineered to effect RNA interference targeting plant virual RNA sequences of Turnip Mosaic Virus (TuMV)[41]. Besides, the platform specific high-sensitivity enzymatic reporter unlocking (SHERLOCK) which is developed on CRISPR/Cas13a system, has made great achievements in the field of virus detection, especially in response to the recent SARS-CoV-2 outbreak[7].

    • Recently, additional CRISPR/Cas systems have been discovered and characterized, which differ from well-known Cas9, Cas12 and Cas13 systems. Cas3 system, which belongs to type I of class 1 CRISPR systems, recruiting CRISPR-associated complex for antiviral defense (Cascade) in trans for processive DNA degradation to programmable genome deletion and large-scale genome engineering[42,43]. Repurposing Cas3 systems to genome editing in Zea mays, shows its potential to become a powerful tool for plant genome streamlining[44]. Besides, a type I-D (TiD) system using Cas10d nuclease instead of Cas3 nuclease that engineered to generate small indels, long-range deletions and bi-directional DNA deletions in the tomato genome[45]. Cas10 system belongs to class 1 Type III CRISPR systems and the standout feature is its unique cas10 protein[46]. CRISPR/Cas10 system, a complex that is composed of multiple subunits and binds to crRNAs, has been used to edit phage genomes[47]. Cas14 is particularly unique for possessing a single-stranded DNA targeting activity and distinguishing single-nucleotide polymorphisms as its half the size of typical class 2 CRISPR effectors[48]. Thus, the CRISPR/Cas14 system has been engineered as an ideal tool for detecting ssDNA viruses in plant, and may be used widely in the diagnostics field[49].

    • Even CRISPR/Cas9, the most widely adopted CRISPR/Cas technology, has some flaws and negatives, such as PAM dependence, off-target effect and so on. Generally, the optimization of CRISPR/Cas9 system in plants mainly involves Cas9 protein, sgRNA and regulatory elements for Cas9 and gRNA (e.g. promoter, codon and terminator). To increase the targeting range, Cas9 protein has been engineered to recognize different PAMs, such as SpCas9-NG (NGN), xCas9 (NG, GAA and GAT), EQR-Cas9 (NGAG), VQR-SpCas9 (NGAN and NGCG) and VRER-SpCas9 (NGCG)[50,51]. A large number of Cas9 orthologs from other bacteria provide potential candidates for engineering Cas9 proteins with different PAM sites, such as StCas9 (NNAGAAW), CjCas9 (NNNVRYM) and SaCas9 (NNNRRT), among others[52,53]. In particular, using an engineered SpRY Cas9 variant made highly efficient genome editing target relaxed PAM sites in rice[54]. Similarly in Cas12 systems, different PAMs, such as TYCV, TATV, CCCC, and TATG, were also recognized by Cas12a variants to expand the scope of targeting[26,55].

      To decrease undesired mutagenesis at off-target sites, truncated gRNAs (17-18 nt), engineering a hairpin secondary structure on the 5' end of the sgRNA and chemical modification at the sgRNA sequence sites have been used and proven effective[5658]. The off-target effects were also effectively reduced by fusing Cas9 protein with programmable DNA-binding domain[59]. With the continuous efforts of researchers, high-fidelity variants (SpCas9-HF1), enhanced-specificity variants (eSpCas9) and several more new variants have been engineered to improve targeting specificity and precision[60,61]. In addition, many bioinformatics tools were developed to evaluate off-target effects and help design highly specific sgRNAs, including E-CRISP, CRISPR-P, and CRISPOR[6264] (Table 2).

      Table 2.  List of available web tools for designing CRISPR/Cas systems.

      NameAdvantageWeb address
      BE-DesignerProvide all possible base editor target sequencehttp://www.rgenome.net/be-designer/
      BEtargetDesign guide RNAs for base editing in plantshttp://skl.scau.edu.cn/betarget/
      Breaking-CasDesign gRNAs for CRISPR/Cas experimentshttp://bioinfogp.cnb.csic.es/tools/breakingcas
      Cas-AnalyzerAnalyze next generation sequencing datahttp://www.rgenome.net/cas-analyzer/
      Cas-DesignerDesign CRISPR targetshttp://www.rgenome.net/cas-designer/
      Cas-OFFinderSearch for potential off-target siteshttp://www.rgenome.net/cas-offinder
      CCTopPredict CRISPR/Cas9 targethttp://crispr.cos.uni-heidelberg.de
      CGDPredict the efficacy of gRNAshttp://big.hanyang.ac.kr:2195/CGD
      CHOPCHOP v3Identify CRISPR–Cas sgRNA targetshttps://chopchop.cbu.uib.no/
      COSMIDSearches genomes for potential off-target siteshttp://crispr.bme.gatech.edu
      CrisPamDetect PAMs for allele-specific targetinghttps://www.danioffenlab.com/crispam
      CRISPORFind guide RNAs and evaluate potential off-targetshttp://crispor.org
      CRISPRdirectSelect rational CRISPR/Cas targetshttp://crispr.dbcls.jp/.
      CRISPRscanDesign sgRNAs for CRISPR/Cas9 targeting in vivohttps://www.crisprscan.org/
      CRISPy-webDesign sgRNAs for microbial genomehttp://crispy.secondarymetabolites.org/
      CRISPR-ERADesign sgRNAs for gene regulationhttp://CRISPR-ERA.stanford.edu
      CRISPR-P 2.0Improved CRISPR-Cas9 tool for genome editing in plantshttp://cbi.hzau.edu.cn/CRISPR2/
      E-CRISPRDesign sgRNAs in a gene-by-gene fashionhttp://www.e-crisp.org/E-CRISP
      MMEJ-KODesign gRNAs for MMEJ-mediated fragment deletionhttp://skl.scau.edu.cn/mmejko/
      Off-SpotterIdentify all potential off-target siteshttps://cm.jefferson.edu/Off-Spotter/
      PhytoCRISP-ExOffer Cas9 target prediction using phytoplankton genomeshttp://www.phytocrispex.biologie.ens.fr/CRISP-Ex/
      The addresses of some web tools could not be resolved, this could be a temporarily unable service so they were not included.

      However, off-target effects are not so unacceptable in plants compared to that in clinical practice[65,66]. In fact, off-target effects in plants are rare. An off-target effects analysis using whole-genome sequencing found off-target mutations were negligible in rice plants edited by Cas9 and Cpf1[67]. Furthermore, any unwanted mutations or negative-effect mutations produced by off-target events could be segregated out through genetic crosses and during sexual reproduction[68]. In crop molecular breeding, as long as it can lead to good traits, it is a good goal, whether the mutation is caused by on-targets or off-targets. Therefore, off-target effects will not be a great hurdle for genome editing to be applied in plants, particularly crop plants, which play a major role in agriculture.

    • Base editing (BE) is a point mutation gene modification technology developed on the CRISPR/Cas9 system. Base editing induces R-loop formation to enable nucleotide substitutions without DSB and donor templates, thereby greatly reducing the random insertion, deletion, translocation and other genomic mutations caused by DNA repair[69,70]. A base editor is a fusion of catalytically deactivated Cas9 domain (Cas9 variants) and deaminase domain, usually divided into cytosine base editors (CBEs) and adenosine base editors (ABEs) according to their deaminase domain[71,72]. CBEs can only convert a C•G base pair to a T•A base pair, whereas ABEs convert an A•T base pair to a G•C base pair (Fig. 2). On the basis of the original prototype, a dual adenine and cytosine base editor (A&C-BEmax) and adenine and cytosine base editor (ACBE) were developed to simultaneously achieve C to T and A to G conersions in mammalian cells by fusing both cytosine and cytosine deaminases, whereas saturated targeted endogenous mutagenesis editors (STEMEs) were developed similar to rice[7375]. Moreover, C-to-G base editors (CGBE), glycosylase base editor (GBE) and UNG-nCas9-AID base editors were developed to convert C to G in human cells, C to G in mammalian cells, and C to A in bacteria, respectively[76,77]. AGBE, a new type of dual deaminase-mediated base editing system that fusing a CGBE with ABE, simultaneously generated four types of base conversions (A to G and C to G / C to T / C to A) as well as indels, thereby greatly extending the diversity of mutations in the same DNA strand at the target sites[78]. Besides being used to mediate specific DNA transition mutations, base editors are also used to directly edit RNA transcript adenosine to inosine (A to I) or cytidine to uridine (C to U) by using a catalytically dead Cas13 (dCas13) and an enzyme adenosine deaminase acting on RNA (ADAR)[39,79]. A large number of base editors have been generated and optimized for different plant species to obtain excellent character trait variations and help crop improvement. Plant based editing systems were first tested in rice, tomato, wheat and maize, proving the feasibility of introducing altered phenotypes[12,8082]. However, bystander mutations, indels, and off-target single nucleotide variations (SNV) were also observed in plants and animal cells after processing by BEs[83]. A higher number SNVs occur in the rice genome after editing by CBE compared to that after editing by ABE, which is supposed to be caused by random deamination[84,85]. Recently, a novel base editing system TAC9-ABE was developed for eliminating Cas9-dependent DNA off-target in human cells, which provides a potentially safe base editing tool for the breeding of gene edited plants and animals[86].

    • Prime editing (PE) is a major breakthrough beyond the CRISPR/Cas9 system, which can effectively generate the free conversion of all single bases, and can also effectively enable the precise insertion and deletion of multiple bases. Prime editor is significantly modified into two aspects; one, to transform Cas9 protein into nickase and then fuse it with reverse transcriptase to form a new protein, and two, to add a sequence including the primer binding site and the reverse transcription template at the 3' end of sgRNA. The newly formed sgRNA is referred to as pegRNA[13]. PE systems just consist of two simple operational components, the fusion protein and pegRNA (Fig. 2). Because of its simplicity, precision and versatility, PE was applied in plant research soon after its invention, especially in rice[8789]. However, the editing efficiency of prime editors in plants is not high and at some sites even lower than that edited by base editors[87]. Therefore, PE needs further improvement to be applied more efficiently in plants.

      To improve PE efficiency, several strategies have been used in plants including engineering pegRNAs and fusion protein, driving promoter replacement and optimizing editing conditions. A dual-pegRNA strategy greatly improved PE at most of the targets (13/15), which boosted PE efficiency 4.2-fold on average[90]. Through incorporating structured RNA motifs or fusing the 20-nt Csy4 recognition site at the 3' end of the pegRNA, PE efficiency was improved[91,92]. Strategic co-expressing MLH1dn to transiently inhibit DNA mismatch repair (MMR) enhances PE efficiency by at least 2.2-fold[93]. Replacing C-terminal reverse transcriptase Cas9 nickase fusion with N-terminal fusion could increase PE efficiency about 2- fold to 3-fold in rice[94]. Through engineering the fusion protein that removes the ribonuclease H domain of Moloney–murine leukemia virus reverse transcriptase (M-MLV RT) and incorporating a viral nucleocapsid protein with nucleic acid chaperone activity, can independently improve plant prime editing efficiency by ~1.8−3.4-fold in plant cells[95]. In addition, suitable high-temperature treatment could improve the PE efficiency in rice protoplast at least 2.8 fold[96]. While each modification at present independently improves PE efficiency several fold, combining these strategies may further improve the efficiency.

    • CRISPR/Cas technologies have developed to such an extent that they are transforming different fields of research, including agriculture and plant biology. CRISPR/Cas technology provides simple, highly efficient, low cost, and scalable tools for the research in plant biology and crop breeding, and has quickly been embraced by plant researchers. More importantly, the diversity of CRISPR/Cas tools is conducive to meeting the editing needs of arbitrary target sequences in different plant genomes.

    • Although CRISPR/Cas technology has been the preferred tool for plant genome editing and genetic operation for a short time, it has already demonstrated great application value, especially in crop genetic improvement. Genome editing provides a new toolkit for plant precision breeding and offers more crop improvement strategies[97]. CRISPR/Cas systems and CRISPR/Cas-derived base editors have emerged as powerful tools not only for DSB-mediated genome editing, but also for generating programmable single DNA base changes. Furthermore, prime editors work as another precise genome editing tool that can enable all base conversions and targeted precise insertions. The precise deletion and insertion of DNA or RNA has made it possible to manipulate gene functions and stack crop traits. New alleles of betaine aldehyde dehydrogenase 2 (OsBADH2) have been created using CRISPR/Cas9 systems, which improved aroma in non-fragrant rice varieties[98]. Producing CRISPR/Cas9 mutants with knocked out OsERF922 in rice, enhanced rice blast resistance[99]. Through editing the uORF of FvebZIPs1.1 in strawberry (Fragaria vesca) by A3A-PBE, a series of novel genotypes with a continuum of sugar content variations were generated that achieved fine-tuning of quantitative traits[15]. In addition, CRISPR/Cas-based systems could be used to knock-up target genes in rice by editing structural variations to create new genes and traits[100].

      Complex traits like growth and development are controlled by multiple genes that require multiplex genome editing and modification. CRISPR/Cas9 has been successfully used to knock-out flavanone-3-hydroxylase 1 (GmF3H1), -2 (GmF3H2) and flavone synthase II 1 (GmFNS II-1) in soya bean, leading to improved isoflavone content and resistance to soya bean mosaic virus (SMV)[101]. Utilizing CRISPR/Cas9-mediated system to directly knock-out 48 growth-related genes in maize created a batch of mutants with bigger leaf compared to the control[102]. Most of the important agronomic traits are significantly associated with single nucleotide polymorphisms (SNPs) and small indels, which are suitable for precise substitution and mutation using BEs and PEs[70]. Enhancing resistance to powdery mildew has been achieved through editing mildew resistance Locus O (MLO) family genes in various plant species, including wheat, tomato, and grapevine[14,103,104]. Enhancing broad-spectrum disease resistance to bacterial pathogens by using CRISPR/Cas9-mediated genome editing for DMR6 orthologue in banana[105]. To date, many traits including yield, quality, and biotic stress and abiotic stress resistance have been improved using CRISPR/Cas systems though knock-in or knock-out methods. Therefore, CRISPR/Cas technology is an effective approach, for improving haploid breeding in maize and rice[106,107], for shortening growth times in rice[108], for reducing cassava brown streak disease (CBSD) symptom severity and incidence in cassava[109], for increasing silique shatter resistance in Brassica napus[110] and overcoming self-incompatibility in diploid potato[111], to meet breeders’ requirements. Moreover, de novo domestication strategies with CRISPR/Cas genome editing technologies as the core, has been proposed and then applied in tomato, ground cherry and rice, which accelerated the process of domesticating crops[112114]. There is no doubt that it is an exciting route and direction for future crop genetic improvement.

    • Epigenetic mechanisms comprise DNA methylation, histone modifications, chromatin remodeling and RNA processing that play an essential role in the regulation of plant growth and development, underlie their complex traits and contribute to evolution[115,116]. Single cytosine methylome analyses of the CmROS1 knockout mutant that was induced by CRISPR/Cas9 systems revealed DNA methylation changes regulating fruit ripening in melon[117]. Constitutively expressing catalytically inactivated dCas9 fusion proteins further expand the application of CRISPR/Cas system for epigenetics studies without inducing DSBs[118]. Manipulated epigenetic modifications, such as CRISPR-mediated activation (CRISPRa) and CRISPR-mediated interference (CRISPRi), have already been achieved using dCas9-based tools in mammalian genome system[119]. Moreover, programmable epigenetic platform CRISPRoff triggers highly specific DNA methylation to silence genes, whereas CRISPRon removes DNA methylation to reverse silenced genes[120]. A dCas9-SunTag-VP64 system was adopted in Arabidopsis for activating DNA methylated genes, which provides a valuable tool for site-specific manipulation of the plant epigenome[121]. The MS2-CRISPR/dCas9 system was developed and validated, to enable manipulation of Arabidopsis flowering time by targeting modulate epigenetic status and transcriptional activity of FT gene[122]. Although only a few cases of CRISPR/dCas-mediated application were reported in plants and may be related to the off-target effects, it is conceivable that more studies will improve targeting specificity and efficiency of these epigenome editors and expand their application in plants in the future[123].

    • The CRISPR/Cas systems have been recognized for their promising applications in detecting different human and plant diseases and Cas9 have been used for miRNA detection[6,124]. In particular, the improvement of CRISPR/Cas12 and CRISPR/Cas13 has brought new dimensions to the field of disease diagnosis (Fig. 3). CRISPR/Cas toolkit reshape management of citrus greening disease through various strategies that precisely edit the host genome for improving resistance, rapidly detect the pathogen for early prevention and potentially use gene drive for insect population control[125]. The rapid, sensitive and specific nucleic acid detection is critical for agricultural applications, including trait detection, pest monitoring and pathogen identification during breeding process[126]. A modified version of the SHERLOCK platform has been used to rapidly quantify the level of glyphosate resistance genes in soybean mixtures and to detect multiple genes in a single reaction, which reveals its huge applications in detection and quantitation of genes in agriculture[127]. The geminiviruses, tomato leaf curl New Delhi virus (ToLCNDV, bipartite begomovirus) and tomato yellow leaf curl virus (TYLCV, monopartite begomovirus), can be both be detected in ~1 h by utilizing Cas12-mediated systems, making it suitable for point-of-use diagnostic applications[128]. By using the improvement of DNA endonuclease targeted CRISPR trans reporter (DETECTR), which is highly sensitive and specific, was used to detect potato witches' broom (PWB) and potato purple top (PPT) phytoplasmas[129]. The new strategy of these diseases-detection methods developed based on CRISPR/Cas technologies provides a practical and convenient approach for detecting early stage viral, bacterial and fungal infections in crop plants.

      Figure 3. 

      Application of CRISPR/Cas12 and CRISPR/Cas13 in nucleic acid detection. Cas12 collateral cleaves a reporter nucleic acid, which is composed of a single stranded DNA (ssDNA) bearing a fluorophore, and the fluorophore is released and leads to the emission of a fluorescent signal. Cas13 : crRNA complexes activate and cleave fluorescent RNA sensors for RNA target detection.

    • Despite the latest technological advances in CRISPR/Cas in plant genome editing, it is still not perfect and may create undesired changes in a genome. Most obviously, off-target effects, massive DNA deletions and other rearrangements still exist in CRISPR/Cas-based editing of plants, and further research to address this propensity is required. Besides, unlike mammalian cells, the plant cell wall makes efficient delivery of CRISPR reagents to plant cells challenging. The current delivery systems, biolistic bombardment and Agrobacterium-mediated delivery, are highly dependent on plant species, genotypes, and tissue-type specificity[130]. Also, plant tissue culture and regeneration still limit the efficiency of obtaining edited plants, especially for those plants that have not yet established genetic transformation and regeneration system, which is a great challenge. Thus, how to create edited plants without going through plant tissue culture that sidesteps the tissue culture bottleneck will be a key direction of future research in plants. Another challenge for plant genome editing is to identify and understand gene function before editing. Genes for desirable traits in many plants have not been mapped clearly. Despite these limitations, CRISPR/Cas systems have great potential for improving plant design and synthetic biology. For example, artificial DNA sequences, including promoters, genes, transcriptional regulatory elements, and genome assemblies, can be inserted into plant genomes to alter cell or plant behavior to generate novel functions and products. Given that CRISPR/Cas systems have been expanding as a molecular manipulation toolbox, misuse of this toolbox could be a risk and danger, so a scientific hierarchical regulatory system might contribute to accommodate both existing and future technologies[131]. CRISPR/Cas, like polymerase chain reaction (PCR), is an epoch-making discovery in the field of life science that will be worth mastering by every researcher. Therefore, with continuous technological advancement and interdisciplinary applications of big data, CRISPR/Cas systems and their derivations will be the most widely used toolbox in plants in the near future and will revolutionize the future of agriculture.

    • The authors would like to thank Dr. Yifu Tian at Hainan Yazhou Bay Seed Laboratory, for his valuable comments and suggestions to improve the quality of the figures by using biorender (https://biorender.com/) in this manuscript. The authors also thank Dr. Xinbo Li from Hainan Yazhou Bay Seed Laboratory for his good suggestions to improve the layout of this manuscript.

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

      • Received 16 November 2022; Accepted 26 January 2023; Published online 17 February 2023

      • # These authors contributed equally: Chao Dong, Yue Xi

      • Copyright: © 2023 by the author(s). Published by Maximum Academic Press on behalf of Hainan University. 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/.
    Figure (3)  Table (2) References (131)
  • About this article
    Cite this article
    Dong C, Xi Y, Satheesh V, Lei M. 2023. Advances in CRISPR/Cas technologies and their application in plants. Tropical Plants 2:2 doi: 10.48130/TP-2023-0002
    Dong C, Xi Y, Satheesh V, Lei M. 2023. Advances in CRISPR/Cas technologies and their application in plants. Tropical Plants 2:2 doi: 10.48130/TP-2023-0002

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return