The latest developments in CRISPR/Cas-mediated homology-directed repair technology for plants are summarized, with a focus on achievements, challenges, and future utility in crop improvement.
Abstract
Beneficial alleles derived from local landraces or related species, or even orthologs from other plant species, are often caused by differences of one or several single-nucleotide polymorphisms or indels in either the promoter region or the encoding region of a gene and often account for major differences in agriculturally important traits. Clustered regularly interspaced short palindromic repeats-associated endonuclease Cas9 system (CRISPR/Cas9)-mediated precision genome editing enables targeted allele replacement or insertion of flag or foreign genes at specific loci via homology-directed repair (HDR); however, HDR efficiency is low due to the intrinsic rare occurrence of HDR and insufficient DNA repair template in the proximity of a double-stranded break (DSB). Precise replacement of the targeted gene with elite alleles from landraces or relatives into a commercial variety through genome editing has been a holy grail in the crop genome editing field. In this update, we briefly summarize CRISPR/Cas-mediated HDR in plants. We describe diverse strategies to improve HDR efficiency by manipulating the DNA repair pathway, timing DSB induction, and donor delivery, and so on. Lastly, we outline open questions and challenges in HDR-mediated precision genome editing in both plant biological research and crop improvement.
Introduction
Rapid population growth, unsustainable agricultural practices, and climate change lead to degradation of natural resources and necessitate the attempts to sustainably increase crop production for global food security. Allelic differences, which have been selected during domestication and subsequent improvements, account for major differences in crop yield and other agriculturally important traits. Most of the valuable alleles, which are usually derived from local landraces or wild relatives, are often caused by differences of one or several single-nucleotide polymorphisms (SNPs), or insertion/deletions (indels) of a gene fragment in either the promoter region or the gene’s encoding region. Harnessing the genetic diversity and introducing the elite alleles into commercial cultivars has been a major goal in crop breeding programs. In crop breeding practice, introgression of the favorable alleles derived from either landraces or wild relatives into commercial varieties without any linkage drag from parental lines has been very difficult, laborious, and time-consuming. In general, it will take the breeders up to >10 years to introduce one beneficial allele into commercial cultivars by crossing and back-crossing. Nonetheless, it is very difficult or sometimes impossible to remove any undesired genes/agronomic traits derived from the parental lines by crossing if they are closely linked to the target genes. Thus, it will greatly accelerate crop improvement if we can introduce the elite alleles from landraces or the related species directly into the commercialized crop variety without introducing unwanted gene or DNA fragments in a short term.
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated endonuclease Cas9 system (CRISPR/Cas) has dominated the genome-editing field and demonstrated great potential either for plant functional genomics or crop improvement over the past few years. So far, three major CRISPR/Cas-mediated systems have been developed for precision genome editing in plants such as base editing (Komor et al., 2016; Gaudelli et al., 2017), prime editing (Anzalone et al., 2019), and homology-directed DNA repair (HDR)-mediated targeted gene replacement or insertion (Puchta and Fauser, 2013; Baltes et al., 2014; Čermák et al., 2015; Li et al., 2015; Shi et al., 2015; Butler et al., 2016; Endo et al., 2016; Sun et al., 2016a; Gil-Humanes et al., 2017; Shi et al., 2017; Wang et al., 2017a; Dahan-Meir et al., 2018; Li et al., 2018d, 2019b, 2020b; Miki et al., 2018; Peng et al., 2020; Lu et al., 2020b; Figure 1). Base editing enables single base transition within the editing window (Komor et al., 2016; Gaudelli et al., 2017; Figure 1A; Table 1). For precise single base substitution of an allele with single SNP, base editing, which is performed by engineered cytidine base editors (CBEs) and adenine base editors (ABEs), has emerged as an alternative tool to HDR-mediated gene replacement, facilitating precise editing of plant genome by transit single base to another without a double-stranded break (DSB) and DNA repair template (DRT) (Veillet et al., 2019). However, there are several limitations in the widespread use of base editors in plants, such as genome editing window, off-target effects, and so on (Table 1). Recently, a precision genome editing technology, prime editing, has been developed that directly mediates all 12 classes of point mutations and predefined indels of the target gene in human cells without requiring DSBs or DRT (Anzalone et al., 2019; Figure 1B;Table 1). Although prime editing could precisely install a wide range of sequences into DNA in plants, its precision editing efficiency remains low, at least for some target genes or plant species (Lin et al., 2020; Xu et al., 2020; Li et al., 2020a). Besides, due to the limited length of the reverse transcriptase (RT) template in the prime editing guide RNA (pegRNA), prime editing only enables small indels and base substitutions in mammalian cell lines as well as in plants (Anzalone et al., 2019; Li et al., 2020a; Wang et al., 2021; Table 1). In contrast, HDR or homologous recombination-based gene targeting enables the installation of all kinds of mutations or a fragment in a predefined manner, albeit at relatively lower efficiency (Puchta et al., 1996; Griffiths et al., 2006; Hiom, 2010; Symington and Gautier, 2011; Puchta and Fauser, 2013, 2014; Altpeter et al, 2016; Endo et al., 2016; Steinert et al., 2016; Sun et al., 2016b; Li and Xia, 2020; Fan et al., 2021; Figure 1C;Table 1). Targeted gene replacement is particularly important for introduction of a favorable allele from either landraces or wild relatives within two to three generations without introducing undesirable genes or traits. Thus, both limitations of base editing and prime editing and the advantages of HDR reiterate the necessities to further explore more efficient HDR technologies for precision genome editing in crop plants.
Table 1.
Gene Editing System | Advantages | Disadvantages |
---|---|---|
Base editing |
|
|
Prime editing |
|
|
HDR-mediated editing |
|
|
In this review, we provide an informative summary of the updated achievements of the CRISPR/Cas-mediated HDR in plants. We then outline diverse strategies to improve HDR efficiency by manipulating DNA repair pathway, timing the DSBs induction and donor delivery, and so on. At last, we discuss the existing challenges in precision genome editing by HDR and propose future perspectives of HDR in plant biological science as well as crop improvement.
CRISPR/Cas-mediated HDR in plants
CRISPR/Cas9-mediated HDR in plants
Once the DSB is produced by the CRISPR/Cas system, it can be repaired by nonhomologous end-joining (NHEJ) or HDR pathway. HDR is a precise repair pathway and is stimulated by the homologous DRT surrounding a DSB (Puchta, 1998; Puchta and Fauser, 2014; Baltes and Voytas, 2015; Li and Xia, 2020; Figure 1C). In general, there are three main repair mechanisms of HDR: single-strand annealing (SSA) model, synthesis-dependent strand annealing (SDSA) model and so-called DSB repair (DSBR) model (see Box 1 Figure). In comparison with mammalian cells, it remains challenging to achieve efficient precise genome editing by CRISPR/Cas-mediated HDR in plants because of four reasons: (1) HDR is mainly active in the late S and G2 phase of the cell cycle (Puchta and Fauser, 2014); (2) As the predominant pathway for DSBs repair in somatic cells, NHEJ competes with the HDR pathway (Symington and Gautier, 2011; Puchta and Fauser, 2014; Sun et al., 2016a); (3) spatially and temporally co-localizing the CRISPR reagents and DRT is crucial for successful HDR events (Endo et al., 2016); and (4) limited DRT delivered into plant cells for HDR due to the presence of cell wall (Li et al., 2019b). In fact, when foreign DNA is introduced into plant cells, there are typically 105 to 107 illegitimate recombination events along with one homologous recombination event (Puchta, 1998; Shukla et al., 2009). Even so, because of its great importance in crop improvement, CRISPR/Cas-mediated HDR has been attempted in various plant species (Baltes et al., 2014; Čermák et al., 2015; Li et al., 2015; Svitashev et al., 2015; Butler et al., 2016; Endo et al., 2016; Sauer et al., 2016; Sun et al., 2016a; Butt et al., 2017; Gil-Humanes et al., 2017; Paix et al., 2017; Shi et al., 2017; Yu et al., 2017; Wang et al., 2017b; Dahan-Meir et al., 2018; De Pater et al., 2018; Hahn et al., 2018; Miki et al., 2018; Shan et al., 2018; Wolter et al., 2018; Barone et al., 2020; Beying et al., 2020; Peng et al., 2020; Schmidt et al., 2020; Lu et al., 2020b). For example, herbicide resistant plants have been generated by replacement of the wild-type (WT) acetolactate synthase (ALS) gene with a mutated ALS gene containing desired mutations through CRISPR/Cas9-mediated HDR in rice (Oryza sativa L.), maize (Zea mays L.), tomato (Solanum lycopersicum), and Arabidopsis (Arabidopsis thaliana) (Svitashev et al., 2015; Endo et al., 2016; Sun et al., 2016a; Wolter et al., 2018; Table 2). Besides, alcobaca (ALC) and the crtiso gene in tomato were successfully modified by CRISPR/Cas9-mediated HDR (Yu et al., 2017; Dahan-Meir et al., 2018). Moreover, replacement of an nitrate-transporter gene (NRT1.1B) allele in a japonica rice with an elite allele from indica rice, improved rice nitrogen use efficiency (Li et al., 2018b). In maize, the grain yield was increased by replacing the native ARGOS8 promoter with the native GOS2 promoter (Shi et al., 2017). In addition, using CRISPR/Cas9 system, a kanamycin (neomycine phosphotransferase II [nptII]) resistance cassette was successfully integrated into alcohol dehydrogenase 1 (ADH1) gene in Arabidopsis (Schiml et al., 2014), and green fluorescent protein (GFP) gene and GFP expression cassette were inserted into glutathione S-transferase gene and putative DNA demethylase (ROS1) gene in rice and Arabidopsis (Wang et al., 2017a; Miki et al., 2018), respectively.
Table 2.
Nuclease | Plant species | Target gene | Donor | Length of donor/left arm/right arm | Transformation methods | References | Note |
---|---|---|---|---|---|---|---|
Cas9 | Arabidopsis | ADH1 | A kanamycin (nptII) resistance cassette | 3,167 bp/674 bp/673 bp | Agrobacterium | Schiml et al. (2014) | |
Cas9 | Arabidopsis | ALS | ALS with mutant sites | 1,542 bp/800 bp/742 bp | Agrobacterium | Wolter et al. (2018) | EC1.1 promoter |
Cas9 | Arabidopsis | ROS1/DME | GFP cassette/DME with mutant sites |
|
Agrobacterium | Miki et al. (2018) | AtDD45 promoter |
LbCas12a | Arabidopsis | ALS | ALS with mutant sites | 1,542 bp/800 bp/74 bp | Agrobacterium | Wolter and Puchta (2019) | EC1.1 promoter |
ttLbCas12a | Arabidopsis | ALS | ALS with mutant sites | 1,542 bp/637 bp/905 bp | Agrobacterium | Merker et al. (2020) | EC1.1 promoter |
Cas9 | Rice | ALS | ALS with mutant sites | −/-/- | Agrobacterium | Endo et al. (2016) | DNA ligase 4 knock out |
Cas9 | Rice | ALS | ALS with mutant sites | 410 bp/100 bp/46 bp | Biolistic | Sun et al. (2016a) | – |
FnCas12a/ LbCas12a | Rice | CAO | Chlorophyllide-a oxygenase (CAO) with mutant sites |
|
Biolistic | Begemann et al. (2017) | – |
Cas9 | Rice | NRT1.1B | Nitrate transporter NRT1.1B with mutant sites | 451 bp/100 bp/100 bp | Biolistic | Li et al. (2018a) | – |
LbCas12a | Rice | ALS | ALS with mutant sites | 635 bp/97 bp/121 bp | Biolistic | Li et al. (2018c) | – |
LbCas12a | Rice | ALS | ALS with mutant sites | 635 bp/97 bp/121 bp | Biolistic | Li et al. (2019b) | – |
Cas9 | Rice | SLR1/TT1/NRT1.1b/UBQ6 | Donor with mutant site/sequence with flag-tag fusion | −/-/- | Biolistic | Lu et al. (2020b) | – |
Cas9 | Rice | Genomic safe harbors | Carotenoid biosynthesis cassette | ∼5.4 kb/794 bp/816 bp | Biolistic | Dong et al. (2020) | – |
Cas9 | Rice | ALS/CCD7/HDT | ALS with mutant sites/CAROTENOID CCD7 with mutant site/C-terminal end with HA tag |
|
Agrobacterium | Ali et al. (2020) | – |
Cas9 | Tomato | ALC | ALC with mutant site | 708 bp/235 bp/450 bp | Agrobacterium | Yu et al. (2017) | Replicon |
Cas9 | Tomato | Crtiso | BeYDV replicons crtiso with mutant sites | 3,796 bp/1,778 bp/1,737 bp | Agrobacterium | Dahan-Meir et al. (2018) | Replicon |
LbCas12a | Tomato | ANT1 | BeYDV replicons anthocyanin mutant 1 (ANT1) and an NPTII expression cassette |
|
Agrobacterium | Vu et al. (2020) | Replicon |
ttLbCas12a | Tomato | ANT1/ SKT12/ EPSPS1 | BeYDV replicons ANT/SKT1.2/EPSPS1 expression cassette |
|
Agrobacterium | Vu et al. (2021) | Replicon |
Cas9 | Maize | ALS2 | ALS gene with mutant sites/dsRED expression cassette | 794 bp/508 bp/283 bp 5424 bp/1097 bp/1135 bp |
Agrobacterium | Svitashev et al. (2015) | – |
Cas9 | Maize | ARGOS8 | ARGOS8 promoter sequences | −/∼400 bp/∼400 bp | Agrobacterium | Shi et al. (2017) | – |
SaCas9 | Tobacco | ALS/SuRB | ALS gene with mutant sites/SuRB with mutant site |
|
Agrobacterium | Huang et al. (2021) | – |
Cas9 | Wheat | Ubiquitin | WDV replicons GFP/BFP/GFP+dsRed cassette |
|
Biolistic | Gil-Humanes et al. (2017) | Replicon |
One of the major reasons for low HDR efficiency is due to the limited DRT delivered into plant cells (Baltes et al., 2014). To enrich DRT template, an all-in-one vector containing Cas9, gRNAs and DRT, co-bombarded with a free DRT fragment, enabled efficient ALS gene replacement through CRISPR/Cas9-mediated HDR to generate herbicide-resistant rice plants (Sun et al., 2016a). Geminivirus replicons (GVRs) have been used to deliver sufficient DRTs into plant cells for CRISPR/Cas9-mediated HDR in tobacco (Nicotiana tabacum L.), tomato (cultivar MicroTom), rice, and wheat (Triticum aestivum L.), respectively (Baltes et al., 2014; Čermák et al., 2015; Butler et al., 2016; Gil-Humanes et al., 2017; Wang et al., 2017a; Dahan-Meir et al., 2018; Table 2). For example, in tobacco, bean yellow dwarf virus (BeYDV) replicon-based HDR strategy enhanced HDR frequencies (Baltes et al., 2014). In tomato cultivar MicroTom, GVR was also used to create heritable modifications (Čermák et al., 2015). In wheat, wheat dwarf virus (WDV) replicons enabled HDR events at an endogenous ubiquitin locus, albeit no stable wheat lines with precise modification were recovered (Gil-Humanes et al., 2017). By using WDV replicon, a GFP-2A-NPTII cassette was incorporated in a specific genomic locus in rice (Wang et al., 2017a). Furthermore, by taking advantage of the geminiviral replicon amplification, a fast-neutron-induced carotenoid isomerase (CRTISO) gene allele was repaired with the WT allele through HDR at an efficiency of 25% (Dahan-Meir et al., 2018). Besides, it was demonstrated that targeted recombination between homologous chromosomes can be achieved via CRISPR/Cas9-induced DSB repair in tomato (Solanum pimpinellifolium) and Arabidopsis, respectively, paving a way for the use of targeted gene conversion for precision breeding (Filler-Hayut et al., 2017; Ben Shlush et al., 2021; Filler-Hayut et al., 2021). Recently, Lu et al. reported a CRISPR/Cas9-mediated tandem repeat-HDR TR-HDR strategy by using a chemical modified donor with phosphorothioate linkages at the 5′- and 3′-ends of donor DNA strands for targeted gene replacement, and achieved replacement or precise modifications with a sequence of up to 130 bp at an efficiency of 6.1% (Lu et al., 2020b). However, it is worth mentioning that although the presence of free DRT itself may not be harmful, it may raise biosafety concerns in products commercialization because of the possible random integration of free DRT in the genome.
CRISPR/Cas12a-mediated HDR in plants
CRISPR/Cas12a has recently been reported as an alternative genome editing system in eukaryotes (Zetsche et al., 2015; Wang et al., 2017b). Cas12a is a class II type V CRISPR/Cas DNA endonuclease that is smaller than Cas9, and functions as an endonuclease to generate DSB and also acting as an endoribonuclease to process Cas12a-associated CRISPR repeats into mature CRISPR RNA (crRNA) (Fonfara et al., 2016; Zetsche et al., 2017). Cas12a extends target recognition in T-rich protospacer adjacent motif (PAM) sequences “TTTN,” and the cuts are located distal to the PAM site, generating staggered ends with overhangs of 4–5 nt upon cleavage (Zetsche et al., 2015). In addition, CRISPR/Cas12a has very low off-target effects in comparison with Cas9. Up to date, CRISPR/Cas12a has been exploited as an efficient and alternative genome editing tool in various organisms including plants such as rice, maize, tomato (Hongkwang cultivar, a local variety), cotton (Gossypium hirsutum), soybean (Glycine max), tobacco (Nicotiana attenuata), and so on (Begemann et al., 2017; Kim et al., 2017; Tang et al., 2017; Wang et al., 2017b; Xu et al., 2017; Bin Moon et al., 2018; Tang et al., 2018; Bernabé-Orts et al., 2019; Jia et al., 2019; Lee et al., 2019; Malzahn et al., 2019; Wolter and Puchta, 2019; Li et al., 2019a, 2020c; Merker et al., 2020; Schindele and Puchta, 2020; Vu et al., 2020; Huang et al., 2021). While the major applications of CRISPR/Cas12a in plants were to generate gene knock-out through NHEJ, successful HDR events by CRISPR/Cas12a system have been achieved in rice, tobacco, and Arabidopsis, respectively (Begemann et al., 2017; Li et al., 2018d, 2019b; Wolter and Puchta, 2019; Merker et al., 2020; Schindele and Puchta, 2020; Vu et al., 2020; Huang et al., 2021; Table 2).
Sufficient DRT and the temperature during tissue culture are two important factors affecting the efficacy of gene replacement by CRISPR/Cas12a (Begemann et al., 2017; Li et al., 2018d, 2019b; Malzahn et al., 2019; Huang et al., 2021). LbCas12a nucleases, delivered with a guide RNA and donor repair template (DRT), successfully generated precise gene insertions as well as indel mutations at the target site in the rice genome, and the targeting frequency was up to 8% (Begemann et al., 2017). Besides, co-delivery of all-in-one CRISPR/Cas12a vector with free DRT, enabled us to precisely replace the ALS gene with desired mutant version at relatively lower efficiency (Li et al., 2018d). Moreover, a transcript-templated HDR (TT-HDR) strategy has been demonstrated in yeast and human cells (Derr et al., 1991; Storici et al., 2007; Nowacki et al., 2008; Keskin et al., 2014). A chimeric single-guide RNA (sgRNA) might serve as both a guide RNA and a DRT (Butt et al., 2017). By coupling Cas12a to an array of crRNAs flanked with ribozymes, and a DRT flanked with ribozymes sequences, stable transgene-free lines with two desired mutations in ALS gene was successfully obtained in rice through TT-HDR, we successfully achieved targeted gene replacement in rice without free DRT, albeit at a relatively lower efficacy (Li et al., 2019b). In addition, Cas12a is sensitive to temperature treatment. High-temperature treatment during culture process can improve Cas12a-mediated genome editing in rice, maize, and Arabidopsis, respectively (Malzahn et al., 2019). In Arabidopsis, LbCas12a successfully mediated precise modification of ALS gene by HDR with a relatively higher efficiency, which greatly broadens the range of in planta HDR (Wolter and Puchta, 2019). Recently, an engineered temperature-insensitive LbCas12a (ttLbCas12a) with a D156R mutation was shown to be more efficient than the WT LbCas12a both for targeted mutagenesis and HDR in Arabidopsis, tobacco, and tomato (Hongkwang cultivar, a local variety), respectively (Merker et al., 2020; Schindele and Puchta, 2020; Huang et al., 2021; Vu et al., 2021; Table 2), demonstrating ttLbCas12a is an ideal tool in crop precision genome editing.
Manipulating DNA repair pathway and taking advantages of cell cycle to improve HDR efficiency
Two main repair pathways have been evolved in cells for DSB repair: NHEJ and HDR. NHEJ is the more predominant DSB repair mechanism, and it competes with the HDR pathway in cells (Symington and Gautier, 2011; Puchta and Fauser, 2014). It is therefore reasonable to enhance HDR by inhibiting main components involved in NHEJ pathway or overexpressing HDR-related proteins (Li and Xia, 2020; Figure 2A). For example, HDR efficiency was significantly increased by knocking out lig4 and PolQ and repressing Ku70 and Ku80 for inhibiting NHEJ pathway, respectively (Endo et al., 2016; Li et al., 2018a; Mara et al., 2019). Activation and interference of CRISPR (CRISPRa/i) by simultaneously activating CDK1 and inhibiting Ku80 significantly enhanced HDR occurrence (Ye et al., 2018). Furthermore, overexpression of RAD54, a protein involved in the HDR, led to an ∼10-fold increase of HDR efficiency in Arabidopsis (Shaked et al., 2005; Even-Faitelson et al., 2011). It was also demonstrated that overexpression of rice proteins RecQl4 (BLM counterpart) and/or Exo1 (Exo1 homolog) can enhance DSB processing, and thus could be applied in developing an efficient HDR system in rice (Kwon et al., 2012). In tobacco (Nicotiana plumbaginifolia), overexpression of a bacterial recombinant protein RecA in plants stimulates homologous recombination (Reiss et al., 2000). It has been demonstrated that overexpression of RAD51 can significantly increase CRISPR/Cas9-mediated homozygous knock-in efficiency in mouse embryos (Wilde et al., 2021). In addition, in human cell lines, SCR7 (the DNA ligase IV inhibitor) can improve efficiency of HDR-mediated genome editing (Chu et al., 2015; Maruyama et al., 2015). NU7441, a DNA-dependent protein kinase inhibitor, can improve the HDR efficiency in comparison to the control (Vu et al., 2021). These above strategies developed in various species including mammalian cells provide us some clues for performing HDR in crop plants, which can be evaluated and utilized in crop plants to further improve the HDR efficiency.
It has been documented that HDR is mainly active in the late S and G2 phases of the cell cycle (Hiom, 2010; Puchta and Fauser, 2014; Orthwein et al., 2015). Delivery of the CRISPR/Cas system into the cells in S phase can increase the HDR efficiency (Lin et al., 2014; Yang et al., 2016; Zhang et al., 2017). Fusion of Cas9 to the N-terminal region of human Geminin, Cas9-hGem (1/110), enabling this fusion protein as a substrate for the E3 ubiquitin ligase complex APC/Cdh1, and resulting in a cell-cycle-tailored expression with low levels in G1 but high expression in S/G2/M, increased the rate of HDR by up to 87% compared to WT Cas9 (Gutschner et al., 2016). In Arabidopsis, using the egg cell- and early embryo-specific promoter to drive the expression of Cas9 could improve the frequency of gene knock-ins and gene replacements via HDR (Miki et al., 2018; Wolter et al., 2018). It was therefore proposed that both the higher occurrence of HDR and tissue-specific Cas9 expression in late S and G2 phase of egg cells and early embryos, facilitated target gene replacement with improved HDR efficiency.
Challenges and future perspectives
Precise replacement of the target gene with the elite alleles for crop improvement
Precise replacement of the target gene with the elite alleles from landraces or relatives in commercial variety through HDR-mediated precision genome editing will greatly accelerate breeding process to breed for novel crop varieties which is otherwise time-consuming or impossible in conventional breeding practice (Li et al., 2021a). However, because of the intrinsic rare occurrence of HDR and insufficient DRT in the proximity of a DSB (Hiom, 2010; Symington and Gautier, 2011; Puchta and Fauser, 2014; Endo et al., 2016; Sun et al., 2016a; Li et al., 2019b), it still remains challenging to perform HDR in crop plants, especially in polyploid crop species such as wheat, which is a major staple food crop consumed by >30% of the world population (Petersen et al., 2006). To further improve the HDR efficiency, there are several open questions or challenges need to be addressed in the future. First, tethering sufficient exogenous DNA donor template in the proximity of a DSB will be a promising strategy to improve the occurrence of HDR in crop plants. For example, covalently tethering a single-stranded donor oligonucleotide (ssODN) to the Cas9/guide RNA ribonucleoprotein (RNP) complex via a fused HUH endonuclease 5, thus spatially and temporally co-localizing the CRISPR reagent and donor DNA, achieved improved HDR efficiency in human cells (Aird et al., 2018). In another study, the truncated Cas9 target sequences attached at terminal of the DRT, which could interact with Cas9 RNP and thus shuttle the templates to the nucleus, enhancing HDR efficiency approximately two- to four-fold in primary hematopoietic cells (Nguyen et al., 2020). Moreover, a chimeric protein Cas9-VirD2 which brings DRT in close proximity to the DSBs, facilitated the occurrence of HDR and enabled precise ALS and CLEAVAGE DIOXYGENASE 7 (OsCCD7) modification as well as in-frame fusions with the HA epitope at HISTONE DEACETYLASE (HDT) locus in rice (Ali et al., 2020). Second, the effect of the length of homologous arms on the efficacy of HDR need to be further investigated. For example, 600 bp homology arms in each side leads to high-level genome knock-in, with 97–100% of the donor insertion events being mediated by HDR in 293 T cells (Zhang et al., 2017). And linear DNAs (single and double-stranded DNA [dsDNA]) engage in a high-efficiency HDR mechanism that requires only ∼35 bp of homologous arm with the targeted site to introduce edits ranging from 1 to 1,000 bp (Paix et al., 2017). Moreover, a 2.7 kilobase (kb) homozygous gene replacement was achieved in up to 11% of iPSC without selection when the homology arm length was around 2 kb (Byrne et al., 2015). In general, in mammalian cells, the most routine way to do HDR is using long ssODNs as repair template (Hu et al., 2019; Gallagher and Haber, 2021). However, this remains challenging in plants because only a few plant species such as tobacco could recover plantlets from protoplast. Given the presence of cell wall in plant cells, it is relatively difficult to deliver both CRISPR/Cas RNP complex and ssODNs into plant cells. Besides, massive plantlets could be regenerated if there is no selection during tissue culture and regeneration processes. So far, only a few examples have been documented in crop plants such as in maize (Svitashev et al., 2016). In plants, different lengths of DRT have been used to achieve gene replacement or knock-in at a range of varied efficiencies (Table 2). It remains to be investigated the effect of the length of homologous arms and the target fragment to be replaced on HDR efficiency systemically. Third, both suppression of the genes involved in the NHEJ pathway or overexpression of related proteins such as RAD54 involved in homologous recombination enhanced the occurrence of HDR; it would be helpful to compare which strategy has a better performance. At last, it would be valuable to establish a routine CRISPR/Cas-mediated HDR system by combining diverse strategies to achieve efficient gene/allele replacement or gene tagging in crop plants without free DRT. Therefore, exploiting diverse strategies to enable the delivery of both CRISPR reagent and DNA repair donor template simultaneously inside the nuclei by an all-in-one vector would be a good choice. Retrons are bacterial phage-defense related operons composed of a specialized RT and a relevant noncoding RNA, which can be partially reverse transcribed by RT to produce a multicopy single-stranded DNA (ssDNA) (Yee et al., 1984; Millman et al., 2020). Recently, fusing retron RT to either the amino terminus or carboxy terminus of Cas9 with a XTEN linker to increase the spatial proximity between ssDNA and Cas9 and produce large amount of ssDNA in vivo for DSB repair, significantly increased the precision genome editing in human cells (Kong et al., 2021; Schubert et al., 2021). Retrons coupled with CRISPR can efficiently insert a GFP gene in yeast (saccharomyces) with efficiency up to 87% (Sharon et al., 2018). In comparison with conventional HDR, retron editing system enables precise gene editing without exogenous donor DNA, increases the abundance of donor msDNA in the vicinity of DSB stimulated by Cas9, thus improving the HDR efficacy, indicating it as an alternative, effective and promising tool in precision genome editing in crop plants. Therefore, in the years to come, it will be interesting to test the retron editing system for either target gene replacement or large fragment insertion in crop improvement.
Optimizing the plant regeneration and CRISPR/Cas delivery systems
To utilize CRISPR/Cas technology for precision genome editing in plants, efficient delivery of the CRISPR/Cas reagent and DRT into a plant cell is an essential step. So far, Agrobacterium tumefaciens-mediated transformation and particle bombardment are two commonly used methods to deliver gene editing reagents into the plant cells (Ghogare et al., 2021; Hayta et al., 2021; Smedley et al., 2021). Precision genome editing in plants is generally limited by low regeneration efficiency and few transformable genotypes. For example, no precisely edited stable wheat lines have been recovered because of its complex hexaploidy genomic background and in fact, most of modern wheat varieties are recalcitrant to genetic transformation and regeneration. To overcome this bottleneck, overexpression of a fusion protein composed of a wheat GROWTH-REGULATING FACTOR 4 (GRF4) and its cofactor GRF-INTERACTING FACTOR 1 (GIF1) has increased the transformation and regeneration efficiency in wheat, triticale (Triticum secale) and rice (Debernardi et al., 2020). Meanwhile, introduction of GRF5 and GRF5 orthologs into canola (Brassica napus), soybean and sunflower (Helianthus annuus L.) significantly increased genetic transformation of the explant tissue (Kong et al., 2020). In addition, regulators that related to somatic embryogenesis have been used to boost plant regeneration. In maize, overexpressing two regulators, WUSCHEL (Wus) and BABY BOOM (BBM), improved regeneration frequencies in various transformation-recalcitrant genotypes (Lowe et al., 2016, 2018). Besides, appropriate ectopic expression of developmental regulators including Wus2 and SHOOT MERISTEMLESS (STM), or MONOPTEROS (MP), could induce the formation of meristem-like structures in vivo in A. thaliana, and sidesteps tissue culture-based transformation for plant genome editing (Maher et al., 2020). To overcome the genotype dependence in wheat, a haploid induction (HI) editing technology, HI-Edit, was developed to enable direct editing of targeted gene not only in maize elite inbred lines, but also in an elite wheat germplasm by a single cross (Kelliher et al., 2019). Recently, an efficient genotype-independent strategy for genome editing in tobacco and wheat have been established by using RNA viral vectors, which sidestep the tissue culture process in genome editing, and helped to realize the promise of plant genome editing for the advancement of basic and applied plant research (Ellison et al., 2020; Li et al., 2021b). The development of RNA viral vectors-mediated genome editing methodology will undoubtedly bypass the genotype-dependent bottleneck in precision genome editing. However, delivery of CRISPR reagent either for gene knock-out or simultaneous delivery of both CRISPR reagent and DRT for precision genome editing in wheat by using this RNA viral system has failed in our laboratory, indicating this strategy might be extremely technically challenging, especially in wheat. Nevertheless, following the optimized regeneration system and development of diverse CRISPR reagent delivery systems for genome editing in plants, extension and validation of these strategies in precision genome editing through HDR will certainly expedite its utility in crop improvement (Figure 2B).
Gene tagging
Gene tagging is a powerful way to analyze the function of proteins in vivo. The development of CRISPR/Cas9-based HDR profoundly transformed our ability to directly tag interested genes at their endogenous loci, which pave a way for characterizing localization and behavior of target gene/protein in cells (Figure 2C). It has been demonstrated that overexpression or transient overexpression of fusion protein of a target protein with either a fluorescent protein or tag might not reflect the actual expression pattern of the endogenous gene (Beck et al., 2011). Similarly, an increased expression of a protein can break the balance between genes, resulting in severe impacts (Gibson et al., 2013). To overcome these drawbacks, inserting fluorescent genes or tags at endogenous genomic loci allows the preservation of a near-native cellular background. However, a slow progress has been made on endogenous genes tagging due to the less efficient HDR. So far, only a few successful cases have been reported, and in-locus tagging of proteins in plants has been challenging (Čermák et al., 2015; Shi et al., 2017; Dong et al., 2020; Lu et al., 2020b). In 2020, a rice protein tagging project (PRPT) was proposed, aiming to label all proteins in rice using a CRISPR/Cas9-based HDR (Lu et al., 2020a).
De novo domestication
Crop improvement by intensive artificial selection of yield components in conventional breeding practice has resulted in decreased stress resistances and loss of genetic diversity due to the linkage drag of deleterious genes/traits, which threatens the stability of yield and food security under the global climate change and frequent occurrences of natural disasters (Hickey et al., 2019). As a matter of fact, in breeding practice, breeders usually need to select the main yield traits while trade-off other agriculturally important traits to breed for a new variety. In contrast, wild relatives of crops could be rapidly re-domesticated using genome editing as proposed by Eshed and Lippman (2019). Also, precision genome editing can enable manipulation of a specific target gene for de novo domestication to facilitate the breeding process without affecting other genes (Figure 2D). Following the development of next-generation DNA sequencing technologies and omics, an immense amount of genomic data including full genome sequences, pan-genomic data, and transcriptomic and proteomic datasets have been generated for many crop species including the most important stable food species such as wheat and rice (Chen et al., 2011; Ikeda et al., 2013; IWGS, 2018; Ling et al., 2018; Li et al., 2018c; He et al., 2019). By combining the deep-sequenced genome data with the precision genome editing, it is possible to tailor crops or design future crops by precise editing of the key domestication genes alongside the development of the wild progenitors into current varieties for sustainable and climate-friendly agricultural development (Fernie and Yan, 2019). Although the successful domestication cases demonstrated so far are through gene knock-out (Zsögön et al., 2018; Li et al., 2018e; Yu et al., 2021), in the long run, it is possible to domesticate some wild relatives or landraces into modern crop varieties through HDR and thus to enrich the genetic diversity and facilitate crop improvement.
Concluding remarks
Although impressive progresses have been made in genome editing during the past few years, it remains very challenging to achieve precision genome editing through HDR in crop plants. Manipulating the DNA repair pathways, timing the DSBs induction and donor delivery, and development of diverse HDR strategies will benefit the improvement of HDR efficiency in crop plants. Furthermore, optimization of tissue culture and development of various delivery systems may facilitate increasing the recovery of HDR events at a better efficacy in crop plants. Following technical improvements and optimization, it is reasonable to expect that CRISPR/Cas-mediated HDR will become a routine platform for precise gene/allele replacement, gene tagging and crop domestication, and thus be widely used for fundamental plant biological research and breeding of diverse crop elite varieties for sustainable agriculture to ensure global food and ecological security.
Advances.
CRISPR/Cas-mediated HDR allows installation of all kinds of mutations without length limitation in a predefined manner, which enables introduction of a favorable allele from either landraces or wild relatives for crop improvement within two to three generations without introducing undesirable genes or traits. However, it remains challenging in plants, especially in crop plants.
HDR efficiency in plants could be increased by using egg-specific promoters to drive the expression of sgRNA, tethering the DRT to the DSB, enriching the availability of DRT surrounded by replicon or using TR-HDR.
CRISPR/Cas-mediated HDR expands our ability to tag genes of interest in locus in vivo, paving the way for characterizing localization and behavior of target genes/proteins in plant cells for fundamental biological research.
Outstanding questions.
How can we improve the occurrence of HDR by providing sufficient exogenous DRT in the proximity of a DSB?
Is suppressing the NHEJ pathway or overexpressing HDR-related proteins more effective for enhancing the occurrence of HDR?
How does length of homologous arms affect the efficacy of HDR?
How can we establish a routine CRISPR/Cas-mediated HDR system by combining diverse strategies to achieve efficient gene/allele replacement or gene tagging in crop plants without free donor repair template?
How can we de novo domesticate wild relatives and landraces into cultivated crops by CRISPR/Cas-mediated HDR technologies rather than gene knock-out in the future?
Box 1.
Acknowledgments
We apologize to those whose work we were unable to cite due to space and reference limitations. We thank Prof. Yunde Zhao, Section of Cell and Developmental Biology, University of California, San Diego, for his critical reading of this manuscript.
Funding
Part of the work in our lab mentioned in this review is funded by the National Natural Science Foundation of China (32188102), Hainan Yazhou Bay Seed Lab (B21HJ0215), National Key Research and Development Program of China (2020YFE0202300), Innovative Talents Supporting Program of Postdoc (BX20200375), and National Engineering Laboratory of Crop Molecular Breeding.
Conflict of interest statement. The authors declare no conflicts of interests.
Contributor Information
Jilin Chen, Institute of Crop Sciences (ICS), Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences/Hainan Yazhou Bay Seed Laboratory, Sanya 572024, China.
Shaoya Li, Institute of Crop Sciences (ICS), Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China.
Yubing He, State Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China.
Jingying Li, Institute of Crop Sciences (ICS), Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences/Hainan Yazhou Bay Seed Laboratory, Sanya 572024, China.
Lanqin Xia, Institute of Crop Sciences (ICS), Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China.
J.C., S.L., Y.H., and J.L. wrote the manuscript. L.X. revised the manuscript. All authors read and approved the final manuscript.
The author responsible for distribution of materials integral to the findings presented in this article in accordance with the policy described in the Instructions for Authors (https://academic.oup.com/plphys/pages/General-Instructions) is: Lanqin Xia (xialanqin@caas.cn).
References
- Aird EJ, Lovendahl KN, St Martin A, Harris RS, Gordon WR (2018) Increasing Cas9-mediated homology-directed repair efficiency through covalent tethering of DNA repair template. Commun Biol 1: 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ali Z, Shami A, Sedeek K, Kamel R, Alhabsi A, Tehseen M, Hassan N, Butt H, Kababji A, Hamdan S, Mahfouz M (2020) Fusion of the Cas9 endonuclease and the VirD2 relaxase facilitates homology-directed repair for precise genome engineering in rice. Commun Biol 3: 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Altpeter F, Springer NM, Bartley LE, Blechl AE, Brutnell TP, Citovsky V, Conrad LJ, Gelvin SB, Jackson DP, Kausch AP, et al. (2016) Advancing crop transformation in the era of genome editing. Plant Cell 28: 1510–1520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, et al. (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576: 149–157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baltes NJ, Gil-Humanes J, Cermak T, Atkins PA, Voytas DF (2014) DNA replicons for plant genome engineering. Plant Cell 26: 151–163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baltes NJ, Voytas DF (2015) Enabling plant synthetic biology through genome engineering. Trend Biotechnol 33: 120–131 [DOI] [PubMed] [Google Scholar]
- Barone P, Wu E, Lenderts B, Anand A, Gordon-Kamm W, Svitashev S, Kumar S (2020) Efficient gene targeting in maize using inducible CRISPR-Cas9 and marker-free donor template. Mol Plant 13: 1219–1227 [DOI] [PubMed] [Google Scholar]
- Beck M, Schmidt A, Malmstroem J, Claassen M, Ori A, Szymborska A, Herzog F, Rinner O, Ellenberg J, Aebersold R (2011) The quantitative proteome of a human cell line. Mol Syst Biol 7: 549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Begemann MB, Gray BN, January E, Gordon GC, He Y, Liu H, Wu X, Brutnell TP, Mockler TC, Oufattole M (2017) Precise insertion and guided editing of higher plant genomes using Cpf1 CRISPR nucleases. Sci Rep 7: 11606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ben Shlush I, Samach A, Melamed-Bessudo C, Ben-Tov D, Dahan-Meir T, Filler-Hayut S, Levy AA (2021) CRISPR/Cas9 induced somatic recombination at the CRTISO locus in tomato. Genes 12: 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernabé-Orts JM, Casas-Rodrigo I, Minguet EG, Landolfi V, Garcia-Carpintero V, Gianoglio S, Vázquez-Vilar M, Granell A, Orzaez D (2019) Assessment of Cas12a-mediated gene editing efficiency in plants. Plant Biotechnol J 17: 1971–1984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beying N, Schmidt C, Pacher M, Houben A, Puchta H (2020) CRISPR-Cas9-mediated induction of heritable chromosomal translocations in arabidopsis. Nat Plants 6: 1–8 [DOI] [PubMed] [Google Scholar]
- Bin Moon S, Lee JM, Kang JG, Lee NE, Ha DI, Kim DY, Kim SH, Yoo K, Kim D, Ko JH, et al. (2018) Highly efficient genome editing by CRISPR-Cpf1 using CRISPR RNA with a uridinylate-rich 3′-overhang. Nat Commun 9: 3651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butler NM, Baltes NJ, Voytas DF, Douches DS (2016) Geminivirus-mediated genome editing in potato (Solanum tuberosum L.) using sequence-specific nucleases. Front Plant Sci 7: 1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Butt H, Eid A, Ali Z, Atia MAM, Mokhtar MM, Hassan N, Lee CM, Bao G, Mahfouz MM (2017) Efficient CRISPR/Cas9-mediated genome editing using a chimeric single-guide RNA molecule. Front Plant Sci 8: 1441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Byrne SM, Luis O, Prashant M, John A, Church GM (2015) Multi-kilobase homozygous targeted gene replacement in human induced pluripotent stem cells. Nucleic Acids Res 43: e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Čermák T, Baltes NJ, Čegan R, Zhang Y, Voytas DF (2015) High-frequency, precise modification of the tomato genome. Genome Biol 16: 232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J, Liu Y, Ni J, Wang Y, Bai Y, Shi J, Gan J, Wu Z, Wu P (2011) OsPHF1 regulates the plasma membrane localization of low- and high-affinity inorganic phosphate transporters and determines inorganic phosphate uptake and translocation in rice. Plant Physiol 157: 269–278 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu VT, Weber T, Wefers B, Wurst W, Sander S, Rajewsky K, Kühn R (2015) Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat Biotechnol 33: 543–548 [DOI] [PubMed] [Google Scholar]
- Dahan-Meir T, Filler-Hayut S, Melamed-Bessudo C, Bocobza S, Czosnek H, Aharoni A, Levy AA (2018) Efficient in planta gene targeting in tomato using geminiviral replicons and the CRISPR/Cas9 system. Plant J 95: 5–16 [DOI] [PubMed] [Google Scholar]
- Debernardi JM, Tricoli DM, Ercoli MF, Hayta S, Ronald P, Palatnik JF, Dubcovsky J (2020) A GRF-GIF chimeric protein improves the regeneration efficiency of transgenic plants. Nat Biotechnol 38: 1274–1279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Pater S, Klemann B, Hooykaas PJJ (2018) True gene-targeting events by CRISPR/Cas-induced DSB repair of the PPO locus with an ectopically integrated repair template. Sci Rep 8: 3338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Derr LK, Strathern JN, Garfinkel DJ (1991) RNA-mediated recombination in S. cerevisiae. Cell 67: 355–364 [DOI] [PubMed] [Google Scholar]
- Dong OX, Yu S, Jain R, Zhang N, Duong PQ, Butler C, Li Y, Lipzen A, Martin JA, Barry KW, et al. (2020) Marker-free carotenoid-enriched rice generated through targeted gene insertion using CRISPR-Cas9. Nat Commun 11: 1178–1178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ellison EE, Nagalakshmi U, Gamo ME, Huang P-J, Dinesh-Kumar S, Voytas DF (2020) Multiplexed heritable gene editing using RNA viruses and mobile single guide RNAs. Nat Plants 6: 620–624 [DOI] [PubMed] [Google Scholar]
- Endo M, Mikami M, Toki S (2016) Biallelic gene targeting in rice. Plant Physiol 170: 667–677 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eshed Y, Lippman Z (2019) Revolutions in agriculture chart a course for targeted breeding of old and new crops. Science 366: 6466. [DOI] [PubMed] [Google Scholar]
- Even-Faitelson L, Samach A, Melamed-Bessudo C, Avivi-Ragolsky N, Levy AA (2011) Localized egg-cell expression of effector proteins for targeted modification of the Arabidopsis genome. Plant J 68: 929–937 [DOI] [PubMed] [Google Scholar]
- Fan C, Hao M, Jia Z, Neri C, Chen X, Chen W, Liu D, Lukaszewski AJ (2021) Some characteristics of crossing over in induced recombination between chromosomes of wheat and rye. Plant J 105: 1665–1676 [DOI] [PubMed] [Google Scholar]
- Fernie AR, Yan J (2019) De novo domestication: An alternative route toward new crops for the future. Mol Plant 12: 615–631 [DOI] [PubMed] [Google Scholar]
- Filler-Hayut S, Kniazev K, Melamed-Bessudo C, Levy AA (2021) Targeted inter-homologs recombination in Arabidopsis euchromatin and heterochromatin. Int J Mol Sci 22: 12096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filler-Hayut S, Melamed Bessudo C, Levy AA (2017) Targeted recombination between homologous chromosomes for precise breeding in tomato. Nat Commun 8: 15605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fonfara I, Richter H, Bratovič M, Le Rhun A, Charpentier E (2016) The CRISPR-associated DNA-cleaving enzyme Cpf1 also processes precursor CRISPR RNA. Nature 532: 517–521 [DOI] [PubMed] [Google Scholar]
- Gallagher DN, Haber JE (2021) Single-strand template repair: Key insights to increase the efficiency of gene editing. Curr Genet 67: 747–753 [DOI] [PubMed] [Google Scholar]
- Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, Liu DR (2017) Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551: 464–471 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghogare R, Ludwig Y, Bueno GM, Slamet-Loedin IH, Dhingra A (2021) Genome editing reagent delivery in plants. Transgen Res 30: 321–335 [DOI] [PubMed] [Google Scholar]
- Gibson TJ, Seiler M, Veitia RA (2013) The transience of transient overexpression. Nat Methods 10: 715–721 [DOI] [PubMed] [Google Scholar]
- Gil-Humanes J, Wang Y, Liang Z, Shan Q, Ozuna CV, Sánchez-León S, Baltes NJ, Starker C, Barro F, Gao C, et al. (2017) High-efficiency gene targeting in hexaploid wheat using DNA replicons and CRISPR/Cas9. Plant J 89: 1251–1262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griffiths S, Sharp R, Foote TN, Bertin I, Wanous M, Reader S, Colas I, Moore G (2006) Molecular characterization of Ph1 as a major chromosome pairing locus in polyploid wheat. Nature 439: 749–752 [DOI] [PubMed] [Google Scholar]
- Gutschner T, Haemmerle M, Genovese G, Draetta Giulio F, Chin L (2016) Post-translational regulation of Cas9 during G1 enhances homology-directed repair. Cell Rep 14: 1555–1566 [DOI] [PubMed] [Google Scholar]
- Hahn F, Eisenhut M, Mantegazza O, Weber APM (2018) Homology-directed repair of a defective glabrous gene in Arabidopsis with Cas9-based gene targeting. Front Plant Sci 9: 1–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayta S, Smedley MA, Clarke M, Forner M, Harwood WA (2021) An efficient Agrobacterium-mediated transformation protocol for hexaploid and tetraploid wheat. Curr Protocol 1: e58. [DOI] [PubMed] [Google Scholar]
- He F, Pasam R, Shi F, Kant S, Keeble-Gagnere G, Kay P, Forrest K, Fritz A, Hucl P, Wiebe K, et al. (2019) Exome sequencing highlights the role of wild-relative introgression in shaping the adaptive landscape of the wheat genome. Nat Genet 51: 896–904 [DOI] [PubMed] [Google Scholar]
- Hickey L, Hafeez A, Robinson H, Jackson S, Leal-Bertioli S, Tester M, Gao C, Godwin I, Hayes B, Wulff B (2019) Breeding crops to feed 10 billion. Nat Biotechnol 37: 744–754 [DOI] [PubMed] [Google Scholar]
- Hiom K (2010) Coping with DNA double strand breaks. DNA Repair 9: 1256–1263 [DOI] [PubMed] [Google Scholar]
- Hu Z, Zhou M, Wu Y, Li Z, Liu X, Wu L, Liang D (2019) ssODN-mediated in-frame deletion with CRISPR/Cas9 restores FVIII function in hemophilia A-Patient-Derived iPSCs and ECs. Mol Ther Nucleic Acid 17: 198–209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang TK, Armstrong B, Schindele P, Puchta H (2021) Efficient gene targeting in Nicotiana tabacum using CRISPR/SaCas9 and temperature tolerant LbCas12a. Plant Biotechnol J 19: 1314–1324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ikeda M, Miura K, Aya K, Kitano H, Matsuoka M (2013) Genes offering the potential for designing yield-related traits in rice. Curr Opin Plant Biol 16: 213–220 [DOI] [PubMed] [Google Scholar]
- IWGSC. (2018) Shifting the limits in wheat research and breeding using a fully annotated reference genome. Science 361: eaar7191. [DOI] [PubMed] [Google Scholar]
- Jia H, Orbović V, Wang N (2019) CRISPR-LbCas12a-mediated modification of citrus. Plant Biotechnol J 17: 1928–1937 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kan Y, Ruis B, Takasugi T, Hendrickson E (2017) Mechanisms of precise genome editing using oligonucleotide donors. Genome Res 27: 1099–1111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelliher T, Starr D, Su X, Tang G, Chen Z, Carter J, Wittich PE, Dong S, Green J, Burch E, et al. (2019) One-step genome editing of elite crop germplasm during haploid induction. Nat Biotechnol 37: 287–292 [DOI] [PubMed] [Google Scholar]
- Keskin H, Shen Y, Huang F, Patel M, Yang T, Ashley K, Mazin AV, Storici F (2014) Transcript-RNA-templated DNA recombination and repair. Nature 515: 436–439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim H, Kim ST, Ryu J, Kang BC, Kim JS, Kim SG (2017) CRISPR/Cpf1-mediated DNA-free plant genome editing. Nat Commun 8: 14406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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: 420–424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong J, Martin-Ortigosa S, Finer J, Orchard N, Gunadi A, Batts LA, Thakare D, Rush B, Schmitz O, Stuiver M, et al. (2020) Overexpression of the transcription factor GROWTH-REGULATING FACTOR5 improves transformation of dicot and monocot species. Front Plant Sci 11: 1389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong X, Wang Z, Zhang R, Wang X, Zhou Y, Shi L, Yang H (2021) Precise genome editing without exogenous donor DNA via retron editing system in human cells. Protein Cell 12: 899–902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon YI, Abe K, Osakabe K, Endo M, Nishizawa-Yokoi A, Saika H, Shimada H, Toki S (2012) Overexpression of OsRecQl4 and/or OsExo1 enhances DSB-induced homologous recombination in rice. Plant Cell Physiol 53: 2142–2152 [DOI] [PubMed] [Google Scholar]
- Lee K, Zhang Y, Kleinstiver BP, Guo JA, Aryee MJ, Miller J, Malzahn A, Zarecor S, Lawrence-Dill CJ, Joung JK, et al. (2019) Activities and specificities of CRISPR/Cas9 and Cas12a nucleases for targeted mutagenesis in maize. Plant Biotechnol J 17: 362–372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B, Rui H, Li Y, Wang Q, Alariqi M, Qin L, Sun L, Ding X, Wang F, Zou J, et al. (2019a) Robust CRISPR/Cpf1 (Cas12a)-mediated genome editing in allotetraploid cotton (Gossypium hirsutum). Plant Biotechnol J 17: 1862–1864 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G, Liu D, Zhang X, Quan R, Zhong C, Mo J, Huang Y, Wang H, Ruan X, Xu Z, et al. (2018a) Suppressing Ku70/Ku80 expression elevates homology-directed repair efficiency in primary fibroblasts. Int J Biochem Cell Biol 99: 154–160 [DOI] [PubMed] [Google Scholar]
- Li H, Li J, Chen J, Yan L, Xia L (2020a) Precise modifications of both exogenous and endogenous genes in rice by prime editing. Mol Plant 13: 671–674 [DOI] [PubMed] [Google Scholar]
- Li J, Li H, Chen J, Yan L, Xia L (2020b) Toward precision genome editing in crop plants. Mol Plant 13: 811–813 [DOI] [PubMed] [Google Scholar]
- Li J, Zhang X, Sun Y, Zhang J, Du W, Guo X, Li S, Zhao Y, Xia L (2018b) Efficient allelic replacement in rice by gene editing: a case study of the NRT1.1B gene. J Integr Plant Biol 60: 536–540 [DOI] [PubMed] [Google Scholar]
- Li S, Tian Y, Wu K, Ye Y, Yu J, Zhang J, Liu Q, Hu M, Li H, Tong Y, et al. (2018c) Modulating plant growth–metabolism coordination for sustainable agriculture. Nature 560: 595–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, Li J, He Y, Xu M, Zhang J, Du W, Zhao Y, Xia L (2019b) Precise gene replacement in rice by RNA transcript-templated homologous recombination. Nat Biotechnol 37: 445–450 [DOI] [PubMed] [Google Scholar]
- Li S, Li J, Zhang J, Du W, Fu J, Sutar S, Zhao Y, Xia L (2018d) Synthesis-dependent repair of Cpf1-induced double strand DNA breaks enables targeted gene replacement in rice. J Exp Bot 69: 4715–4721 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, Xia L (2020) Precise gene replacement in plants through CRISPR/Cas genome editing technology: Current status and future perspectives. aBIOTECH 1: 58–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, Zhang C, Li J, Yan L, Wang N, Xia L (2021a) Present and future prospects for wheat improvement through genome editing and advanced technologies. Plant Commun 2: 100211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li S, Zhang Y, Xia L, Qi Y (2020c) CRISPR-Cas12a enables efficient biallelic gene targeting in rice. Plant Biotechnol J 18: 1351–1353 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li T, Yang X, Yu Y, Si X, Zhai X, Zhang H, Dong W, Gao C, Xu C (2018e) Domestication of wild tomato is accelerated by genome editing. Nat Biotechnol 36: 1160–1163 [DOI] [PubMed] [Google Scholar]
- Li T, Hu J, Sun Y, Li B, Zhang D, Li W, Liu J, Li D, Gao C, Zhang Y (2021b) Highly efficient heritable genome editing in wheat using an rna virus and bypassing tissue culture. Mol Plant 14: 1–12 [DOI] [PubMed] [Google Scholar]
- Li Z, Liu ZB, Xing A, Moon BP, Koellhoffer JP, Huang L, Ward RT, Clifton E, Falco SC, Cigan AM (2015) Cas9-guide RNA directed genome editing in soybean. Plant Physiol 169: 960–970 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin Q, Zong Y, Xue C, Wang S, Jin S, Zhu Z, Wang Y, Anzalone AV, Raguram A, Doman JL, et al. (2020) Prime genome editing in rice and wheat. Nat Biotechnol 38: 582–585 [DOI] [PubMed] [Google Scholar]
- Lin S, Staahl BT, Alla RK, Doudna JA (2014) Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. Elife 3: e04766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ling HQ, Ma B, Shi X, Liu H, Dong L, Sun H, Cao Y, Gao Q, Zheng S, Li Y, et al. (2018) Genome sequence of the progenitor of wheat A subgenome Triticum urartu. Nature 557: 424–428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowe K, La Rota M, Hoerster G, Hastings C, Wang N, Chamberlin M, Wu E, Jones T, Gordon-Kamm W (2018) Rapid genotype “independent” Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev Biol Plant 54: 240–252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowe K, Wu E, Wang N, Hoerster G, Hastings C, Cho MJ, Scelonge C, Lenderts B, Chamberlin M, Cushatt J, et al. (2016) Morphogenic regulators baby boom and wuschel improve monocot transformation. Plant Cell 28: 1998–2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu Y, Ronald PC, Han B, Li J, Zhu JK (2020a) Rice protein tagging project: A call for international collaborations on genome-wide in-locus tagging of rice proteins. Mol Plant 13: 1663–1665 [DOI] [PubMed] [Google Scholar]
- Lu Y, Tian Y, Shen R, Yao Q, Wang M, Chen M, Dong J, Zhang T, Li F, Lei M, et al. (2020b) Targeted, efficient sequence insertion and replacement in rice. Nat Biotechnol 38: 1402–1407 [DOI] [PubMed] [Google Scholar]
- Maher MF, Nasti RA, Vollbrecht M, Starker CG, Clark MD, Voytas DF (2020) Plant gene editing through de novo induction of meristems. Nat Biotechnol 38: 84–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malzahn AA, Tang X, Lee K, Ren Q, Sretenovic S, Zhang Y, Chen H, Kang M, Bao Y, Zheng X, et al. (2019) Application of CRISPR-Cas12a temperature sensitivity for improved genome editing in rice, maize, and Arabidopsis. BMC Biol 17: 1–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mara K, Charlot F, Guyon-Debast A, Schaefer DG, Collonnier C, Grelon M, Nogué F (2019) POLQ plays a key role in the repair of CRISPR/Cas9-induced double-stranded breaks in the moss Physcomitrella patens. New Phytologist 222: 1380–1391 [DOI] [PubMed] [Google Scholar]
- Maruyama T, Dougan SK, Truttmann MC, Bilate AM, Ingram JR, Ploegh HL (2015) Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat Biotechnol 33: 538–542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merker L, Schindele P, Huang T-K, Wolter F, Puchta H (2020) Enhancing in planta gene targeting efficiencies in Arabidopsis using temperature-tolerant CRISPR/LbCas12a. Plant Biotechnol J 18: 2382–2384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miki D, Zhang W, Zeng W, Feng Z, Zhu JK (2018) CRISPR/Cas9-mediated gene targeting in Arabidopsis using sequential transformation. Nat Commun 9: 1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Millman A, Bernheim A, Stokar-Avihail A, Fedorenko T, Voichek M, Leavitt A, Oppenheimer-Shaanan Y, Sorek R (2020) Bacterial retrons function in anti-phage defense. Cell 183: 1551–1561 [DOI] [PubMed] [Google Scholar]
- Nguyen DN, Roth TL, Li PJ, Chen PA, Apathy R, Mamedov MR, Vo LT, Tobin VR, Goodman D, Shifrut E, et al. (2020) Polymer-stabilized Cas9 nanoparticles and modified repair templates increase genome editing efficiency. Nat Biotechnol 38: 44–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nowacki M, Vijayan V, Zhou Y, Schotanus K, Doak TG, Landweber LF (2008) RNA-mediated epigenetic programming of a genome-rearrangement pathway. Nature 451: 153–158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orthwein A, Noordermeer SM, Wilson MD, Landry S, Enchev RI, Sherker A, Munro M, Pinder J, Salsman J, Dellaire G, et al. (2015) A mechanism for the suppression of homologous recombination in G1 cells. Nature 528: 422–426 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Osman K, Higgins JD, Sanchez-Moran E, Armstrong SJ, Franklin FC (2011). Pathways to meiotic recombination in Arabidopsis thaliana. New Phytol 190: 523–544 [DOI] [PubMed] [Google Scholar]
- Paix A, Folkmann A, Goldman DH, Kulaga H, Grzelak MJ, Rasoloson D, Paidemarry S, Green R, Reed RR, Seydoux G (2017) Precision genome editing using synthesis-dependent repair of Cas9-induced DNA breaks. Proc Natl Acad Sci USA 114: E10745–E10754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paquet D, Kwart D, Chen A, Sproul A, Jacob S, Teo S, Olsen KM, Gregg A, Noggle S, Tessier-Lavigne M (2016) Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533: 125–129 [DOI] [PubMed] [Google Scholar]
- Peng FN, Zhang WX, Zeng WJ, Zhu JK, Miki D (2020) Gene targeting in Arabidopsis via an all-in-one strategy that uses a translational enhancer to aid Cas9 expression. Plant Biotechnol J 18: 892–894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petersen G, Seberg O, Yde M, Berthelsen K (2006) Phylogenetic relationships of Triticum and Aegilops and evidence for the origin of the A, B, and D genomes of common wheat (Triticum aestivum). Mol Phylogenet Evol 39: 70–82 [DOI] [PubMed] [Google Scholar]
- Puchta H, Dujon B, Hohn B (1996) Two different but related mechanisms are used in plants for the repair of genomic double-strand breaks by homologous recombination. Proc Natl Acad Sci USA 93: 5055–5060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puchta H (1998) Repair of genomic double-strand breaks in somatic plant cells by one-sided invasion of homologous sequences. Plant J 13: 331–339 [Google Scholar]
- Puchta H, Fauser F (2013) Gene targeting in plants: 25 years later. Int J Dev Biol 57: 629–637 [DOI] [PubMed] [Google Scholar]
- Puchta H, Fauser F (2014) Synthetic nucleases for genome engineering in plants: prospects for a bright future. Plant J 78: 727–741 [DOI] [PubMed] [Google Scholar]
- Reiss B, Schubert I, Köpchen K, Wendeler E, Schell J, Puchta H (2000) RecA stimulates sister chromatid exchange and the fidelity of double-strand break repair, but not gene targeting, in plants transformed by Agrobacterium. Proc Natl Acad Sci USA 97: 3358–3363 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richardson CD, Ray GJ, DeWitt MA, Curie GL, Corn JE (2016) Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat Biotechnol 34: 339–344 [DOI] [PubMed] [Google Scholar]
- Sauer NJ, Narváez-Vásquez J, Mozoruk J, Miller RB, Warburg ZJ, Woodward MJ, Mihiret YA, Lincoln TA, Segami RE, Sanders SL, et al. (2016) Oligonucleotide-mediated genome editing provides precision and function to engineered nucleases and antibiotics in plants. Plant Physiol 170: 1917–1928 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schiml S, Fauser F, Puchta H (2014) The CRISPR/Cas system can be used as nuclease for in planta gene targeting and as paired nickases for directed mutagenesis in Arabidopsis resulting in heritable progeny. Plant J 80: 1139–1150 [DOI] [PubMed] [Google Scholar]
- Schindele P, Puchta H (2020) Engineering CRISPR/LbCas12a for highly efficient, temperature-tolerant plant gene editing. Plant Biotechnol J 18: 1118–1120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt C, Fransz P, Rnspies M, Dreissig S, Puchta H (2020) Changing local recombination patterns in arabidopsis by CRISPR/Cass mediated chromosome engineering. Nat Commun 11: 4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schubert MG, Goodman DB, Wannier TM, Kaur D, Farzadfard F, Lu TK, Shipman SL, Church GM (2021) High-throughput functional variant screens via in vivo production of single-stranded DNA. Proc Natl Acad Sci USA 118: e2018181118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaked H, Melamed-Bessudo C, Levy AA (2005) High-frequency gene targeting in Arabidopsis plants expressing the yeast RAD54 gene. Proc Natl Acad Sci USA 102: 12265–12269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shan Q, Baltes NJ, Atkins P, Kirkland ER, Zhang Y, Baller JA, Lowder LG, Malzahn AA, Haugner JC, Seelig B, et al. (2018) ZFN, TALEN and CRISPR-Cas9 mediated homology directed gene insertion in Arabidopsis: A disconnect between somatic and germinal cells. J Genet Genom 45: 681–684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharon E, Chen S-AA, Khosla NM, Smith JD, Pritchard JK, Fraser HB (2018) Functional genetic variants revealed by massively parallel precise genome editing. Cell 175: 544–557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi J, Habben JE, Archibald RL, Drummond BJ, Chamberlin MA, Williams RW, Lafitte HR, Weers BP (2015) Overexpression of ARGOS genes modifies plant sensitivity to ethylene, leading to improved drought tolerance in both Arabidopsis and maize. Plant Physiol 169: 266–282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi J, Gao H, Wang H, Lafitte HR, Archibald RL, Yang M, Hakimi SM, Mo H, Habben JE (2017) ARGOS8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnol J 15: 207–216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shukla VK, Doyon Y, Miller JC, Dekelver RC, Moehle EA, Worden SE, Mitchell JC, Arnold NL, Gopalan S, Meng X (2009) Precise genome modification in the crop species Zea mays using zinc-finger nucleases. Nature 459: 437–441 [DOI] [PubMed] [Google Scholar]
- Smedley MA, Hayta S, Clarke M, Harwood WA (2021) CRISPR-Cas9 based genome editing in wheat. Curr Protocol 1: e65. [DOI] [PubMed] [Google Scholar]
- Steinert J, Schiml S, Puchta H (2016) Homology-based double-strand break-induced genome engineering in plants. Plant Cell Rep 35: 1429–1438 [DOI] [PubMed] [Google Scholar]
- Storici F, Bebenek K, Kunkel TA, Gordenin DA, Resnick MA (2007) RNA-templated DNA repair. Nature 447: 338–341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Y, Zhang X, Wu C, He Y, Ma Y, Hou H, Guo X, Du W, Zhao Y, Xia L (2016a) Engineering herbicide-resistant rice plants through CRISPR/Cas9-mediated homologous recombination of acetolactate synthase. Mol Plant 9: 628–631 [DOI] [PubMed] [Google Scholar]
- Sun Y, Li J, Xia L (2016b) Precise genome modification via sequence-specific nucleases-mediated gene targeting for crop improvement. Front Plant Sci 7: 1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Svitashev S, Young JK, Schwartz C, Gao H, Falco SC, Cigan AM (2015) Targeted mutagenesis, precise gene editing, and site-specific gene insertion in maize using Cas9 and guide RNA. Plant Physiol 169: 931–945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Svitashev S, Schwartz C, Lenderts B, Young JK, Cigan AM (2016) Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat Commun 7: 13274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Symington LS, Gautier J (2011) Double-strand break end resection and repair pathway choice. Ann Rev Genet 45: 247–271 [DOI] [PubMed] [Google Scholar]
- Tang X, Liu G, Zhou J, Ren Q, You Q, Tian L, Xin X, Zhong Z, Liu B, Zheng X, 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 Biol 19: 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang X, Lowder LG, Zhang T, Malzahn AA, Zheng X, Voytas DF, Zhong Z, Chen Y, Ren Q, Li Q, et al. (2017) A CRISPR-Cpf1 system for efficient genome editing and transcriptional repression in plants. Nat Plants 3: 17018. [DOI] [PubMed] [Google Scholar]
- Veillet F, Perrot L, Chauvin L, Kermarrec MP, Guyon-Debast A, Chauvin JE, Nogué F, Mazier M (2019) Transgene-free genome editing in tomato and potato plants using Agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. Int J Mol Sci 20: 402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vu T, Duong D, Tran MT, Yeon-Woo S, Kim JY (2021) Improvement of the LbCas12a-crRNA system for efficient gene targeting in Tomato. Front Plant Sci 12: 722552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vu TV, Sivankalyani V, Kim EJ, Doan DTH, Tran MT, Kim J, Sung YW, Park M, Kang YJ, Kim JY (2020) Highly efficient homology-directed repair using CRISPR/Cpf1-geminiviral replicon in tomato. Plant Biotechnol J 18: 2133–2143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, Kaya HB, Zhang N, Rai R, Willmann MR, Carpenter SCD, Read AC, Martin F, Fei Z, Leach JE, et al. (2021) Spelling changes and fluorescent tagging with prime editing vectors for plants. Front Genome Ed 3: 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang M, Lu Y, Botella JR, Mao Y, Hua K, Zhu JK (2017a) Gene targeting by homology-directed repair in rice using a geminivirus-based CRISPR/Cas9 system. Mol Plant 10: 1007–1010 [DOI] [PubMed] [Google Scholar]
- Wang M, Mao Y, Lu Y, Tao X, Zhu JK (2017b) Multiplex gene editing in rice using the CRISPR-Cpf1 system. Mol Plant 10: 1011–1013 [DOI] [PubMed] [Google Scholar]
- Wilde JJ, Aida T, del Rosario RCH, Kaiser T, Qi P, Wienisch M, Zhang Q, Colvin S, Feng G (2021) Efficient embryonic homozygous gene conversion via RAD51-enhanced interhomolog repair. Cell 184: 3267–3280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolter F, Klemm J, Puchta H (2018) Efficient in planta gene targeting in Arabidopsis using egg cell-specific expression of the Cas9 nuclease of Staphylococcus aureus. Plant J 94: 735–746 [DOI] [PubMed] [Google Scholar]
- Wolter F, Puchta H (2019) In planta gene targeting can be enhanced by the use of CRISPR/Cas12a. Plant J 100: 1083–1094 [DOI] [PubMed] [Google Scholar]
- Xu R, Qin R, Li H, Li D, Li L, Wei P, Yang J (2017) Generation of targeted mutant rice using a CRISPR-Cpf1 system. Plant Biotechnol J 15: 713–717 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu W, Zhang C, Yang Y, Zhao S, Kang G, He X, Song J, Yang J (2020) Versatile nucleotides substitution in plant using an improved prime editing system. Mol Plant 13: 675–678 [DOI] [PubMed] [Google Scholar]
- Yang D, Scavuzzo MA, Chmielowiec J, Sharp R, Bajic A, Borowiak M (2016) Enrichment of G2/M cell cycle phase in human pluripotent stem cells enhances HDR-mediated gene repair with customizable endonucleases. Sci Rep 6: 21264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye L, Wang C, Hong L, Sun N, Chen D, Chen S, Han F (2018) Programmable DNA repair with CRISPRa/i enhanced homology-directed repair efficiency with a single Cas9. Cell Discov 4: 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yee T, Furuichi T, Inouye S, Inouye M (1984) Multicopy single-stranded DNA isolated from a gram-negative bacterium, Myxococcus xanthus. Cell 38: 203–209 [DOI] [PubMed] [Google Scholar]
- Yu H, Lin T, Meng X, Du H, Zhang J, Liu G, Chen M, Jing Y, Kou L, Li X, et al. (2021) A route to de novo domestication of wild allotetraploid rice. Cell 184: 1156–1170 [DOI] [PubMed] [Google Scholar]
- Yu QH, Wang B, Li N, Tang Y, Yang S, Yang T, Xu J, Guo C, Yan P, Wang Q, et al. (2017) CRISPR/Cas9-induced targeted mutagenesis and gene replacement to generate long-shelf life tomato lines. Sci Rep 7: 11874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, Volz SE, Joung J, van der Oost J, Regev A, et al. (2015) Cpf1 is a single RNA-guided endonuclease of a Class 2 CRISPR-Cas system. Cell 163: 759–771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zetsche B, Heidenreich M, Mohanraju P, Fedorova I, Kneppers J, DeGennaro EM, Winblad N, Choudhury SR, Abudayyeh OO, Gootenberg JS, et al. (2017) Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nat Biotechnol 35: 31–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang JP, Li XL, Li GH, Chen W, Arakaki C, Botimer GD, Baylink D, Zhang L, Wen W, Fu YW, et al. (2017) Efficient precise knockin with a double cut HDR donor after CRISPR/Cas9-mediated double-stranded DNA cleavage. Genome Biol 18: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zsögön A, Čermák T, Naves ER, Notini MM, Edel KH, Weinl S, Freschi L, Voytas DF, Kudla J, Peres LEP (2018) De novo domestication of wild tomato using genome editing. Nat Biotechnol 36: 1211–1216 [DOI] [PubMed] [Google Scholar]