Next Article in Journal
FGFR Inhibitors in Cholangiocarcinoma—A Novel Yet Primary Approach: Where Do We Stand Now and Where to Head Next in Targeting This Axis?
Previous Article in Journal
Nrf2 Deficiency Exacerbated CLP-Induced Pulmonary Injury and Inflammation through Autophagy- and NF-κB/PPARγ-Mediated Macrophage Polarization
Previous Article in Special Issue
Editing of a Novel Cd Uptake-Related Gene CUP1 Contributes to Reducing Cd Accumulations in Arabidopsis thaliana and Brassica napus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CRISPR/Cas Genome Editing Technologies for Plant Improvement against Biotic and Abiotic Stresses: Advances, Limitations, and Future Perspectives

1
Hubei Key Laboratory of Plant Pathology, Huazhong Agricultural University, Wuhan 430070, China
2
Hubei Hongshan Laboratory, National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan 430070, China
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(23), 3928; https://doi.org/10.3390/cells11233928
Submission received: 29 October 2022 / Revised: 28 November 2022 / Accepted: 1 December 2022 / Published: 5 December 2022
(This article belongs to the Special Issue Plant Genome Editing: State-of-the-Art and Perspectives in China)

Abstract

:
Crossbreeding, mutation breeding, and traditional transgenic breeding take much time to improve desirable characters/traits. CRISPR/Cas-mediated genome editing (GE) is a game-changing tool that can create variation in desired traits, such as biotic and abiotic resistance, increase quality and yield in less time with easy applications, high efficiency, and low cost in producing the targeted edits for rapid improvement of crop plants. Plant pathogens and the severe environment cause considerable crop losses worldwide. GE approaches have emerged and opened new doors for breeding multiple-resistance crop varieties. Here, we have summarized recent advances in CRISPR/Cas-mediated GE for resistance against biotic and abiotic stresses in a crop molecular breeding program that includes the modification and improvement of genes response to biotic stresses induced by fungus, virus, and bacterial pathogens. We also discussed in depth the application of CRISPR/Cas for abiotic stresses (herbicide, drought, heat, and cold) in plants. In addition, we discussed the limitations and future challenges faced by breeders using GE tools for crop improvement and suggested directions for future improvements in GE for agricultural applications, providing novel ideas to create super cultivars with broad resistance to biotic and abiotic stress.

1. Introduction

Plants produce food, fuel, and feed, which are essential in daily human and animal life for nourishment and growth. In the process of plant growth, they will be affected by a variety of biological stresses (bacteria, viruses, fungi, and insects) and abiotic stresses [1,2,3,4,5]. Due to continuous global climate change and anthropogenic activity, the impact of abiotic stresses on crop growth and development is becoming more serious. Abiotic stresses, including drought, salinity, waterlogging, heat/cold, and heavy metals, significantly reduce agricultural production worldwide. Therefore, the ability to breed new species that are tolerant to various stresses in order to reduce yield loss will be a sustainable way to overcome these obstacles and meet the growing needs of human beings. Different types of biotic stresses involve a complex interplay among pathogens and host plants based on the susceptibility/resistance of crop plants to any disease. The latest advances in molecular tools have provided insights into a wide array of pathogen infection mechanisms and their interactions with specific crop plants. The insertions/deletions (Indels) mutations by artificial or natural phenomena might be involved in altering these interactions and hinder the pathways involved in the mode of infection [6,7].
Traditional crop breeding such as crossbreeding is an effective method that has been widely used to modify various plant species. Crop productivity and varieties can be increased effectively through crop breeding programs. In modern agriculture, the key methodologies used for breeding purposes are transgenic breeding, mutation-breeding, and GE-mediated breeding for crop improvement [8]. Cross-breeding and genetic recombination require years to introduce desirable alleles and increase variability [8]. Transgenic breeding is easy and well-known, improving crop traits by the exogenous transformation of genes into economically important elite varieties greatly shortens the breeding time. Still, this method inserts specific genes into the genome at random locations through plant transformation, which results in varieties containing foreign DNA. Compared to crossbreeding, mutation-breeding, and traditional transgenic breeding, GE-mediated crop breeding is fast, efficient, and accurate (Figure 1). GE improves a targeted trait in a very fast and short time and exactly revising the target gene or regulatory sequence or changing the DNA and/or RNA bases in elite varieties. The current GE technique includes meganuclease (MegN), zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9) [7,9,10,11]. In 2013, genetic modifications through the CRISPR/Cas9 method were developed in plants and revolutionized the field by eliminating the barriers to targeted GE. The CRISPR system has been used in wheat, rice, tobacco, and Arabidopsis thaliana [12,13,14,15,16,17,18,19,20,21,22,23,24,25]. Till now, GE has been practiced in more than 50 plant species, and it will revolutionize plant breeding [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40] (Table 1).
Based on the composition of the CRISPR locus, this system has been divided into two classes: Class 1 requires multiple effector proteins with subtypes I, III, and IV, while class 2 requires only a single effector protein with subtypes II, V, and VI. The mode-of-action of GE by site-directed nucleases (SDNs) is that once present in a cell by insertion/expression and or transfection, the SDN is capable of cutting the genome at a targeted site. The cellular DNA-repair mechanisms fix the cut sites either by the non-homologous end joining (NHEJ) or by homology-directed repair (HDR). As NHEJ can be an error-prone process, indels can appear at the respective genomic site, leading to a loss-of-function edited gene sequence due to frameshift mutations. GE by using SDNs, can be categorized into three types: SDN-1 introduces small insertions or deletions which carry no additional or recombinant DNA. SDN-2 introduces short insertions or editing of a few base pairs by an external DNA-template sequence. The SDN-3, using a similar method to SDN-2, can be considered transgenic due to the insertion of large DNA pieces [62,63]. Since its introduction, in recent years, constant improvements have been made to make CRISPR systems easier and more suitable for different constraints, such as CRISPR/Cas9 [12,13,42,55], CRISPR/Cas12a [53,58,64,65,66], CRISPR/Cas12b [56], CRISPR/Cas13 [67,68], base editing tools [43,54,59,69,70,71,72], and CRISPR transcriptional activation (CRISPRa) [73,74,75,76,77] (Figure 2). A new form of GE technology, known as Prime Editing (PE) has recently been developed which is capable of achieving various forms of editing, for example, some base-to-base transfer, such as all transformations (C→T, G→A, A→G, and T→C) and transversion mutations (C→A, C→G, G→C, G→T, A→C, A→T, T→A, and T→G), as well as small indels without double-stranded breaks in the DNA. Since PE has enough versatility to accomplish specific forms of editing in the genome, it has great potential to grow superior crops for different purposes, including production, avoiding various biotic and abiotic stresses, and enhancing the quality of plant products [45,46,49,50,51,57,70,71,78,79,80].
CRISPR/Cas method has become the most popular among editing technologies and, thus far, has revealed the greatest potential to overcome the developing challenges (such as yield and biotic and abiotic stresses) of agriculture [9,81,82,83]. For example, mutations conferring resistance to various diseases in lettuce also exist [84]. Resistance against powdery mildew has been successfully acquired in barley by creating mutants at the mildew resistance locus o (MLO) [85]. The mutation at MLO is remarkable because it provides extraordinary, stable, and precise resistance for two decades against mildew without breakage of alleles; this long-lasting resistance is because of gene knockout [86,87]. Herein, we have summarized the recent developments and advances of CRISPR/Cas GE techniques to enhance crop resistance in biotic resistance (fungi, viruses, and bacteria) and abiotic (drought, salt, cold, and heat) resistance in sustainable agriculture, and discussed the advantages, limitations, and future prospects of the CRISPR/Cas system in modern agriculture.

2. CRISPR/Cas Technique for Disease Resistance

Biotic stresses, such as bacterial, viral, and fungal diseases, as well as herbivores, damage plant products every year, affecting 11% to 30% of worldwide agriculture production [88]. Plant defense against pathogens can reduce the effects of disease on plant growth and productivity, which is highly relevant to the lack of food availability in the world with the increasing population. Improvements in new methods or GE techniques have improved the new resistant crops, reducing yield losses due to plant defense. Until now, CRISPR/Cas techniques were mostly used against viral infection and for fungal and bacterial disease resistance (Figure 3). The CRISPR/Cas system has been used to develop resistance to many pathogen species [26,89] (Table 2).

2.1. CRISPR/Cas-Mediated Fungal Resistance in Plants

Many fungal pathogens cause lethal diseases in crop plants, such as rust, mildew, rot, and smut, which not only damage yield yearly in the biosphere but also damage the quality of the product. CRISPR/Cas has improved mycological resistance in various crop species based on the available information of the genomic mechanisms involved in crop-pathogen interactions. Defined candidate genes and gene products have provided the potential to increase plant defense against fungi [106,107]. In three crop varieties, RNA-guided Cas9 endonuclease was used to target MLO loci, such as tomato (Solanum lycopersicum), grapevine (Vitis vinifera), and wheat [47,100,105,110,111], and transgene-free plants have been generated [122]. An MLO encoded protein is localized in the cell membrane and contains seven transmembrane domains, which universally exist in all dicots and monocots [123]. Plants carrying loss-of-function alleles (mlo) of the MLO, such as A. thaliana, tomato, and barley, confer durable resistance against powdery mildew [124,125,126]. Using precision GE to target the MLO-B1 locus of the wheat genome to generate a 304K deletion Tamlo-R32 mutant maintains wheat growth and yield while providing robust powdery mildew resistance [86]. Out of three MLO home alleles, one (TaMLO-A1) has been mutated by CRISPR/Cas9 in triticum aestivum and displayed resilient resistance against Blumeria graminis f. sp. tritici infection [47]. The CRISPR/Cas-mediated transgene-free and self-pollinated tomato variety, which was developed by deleting the 48 bp fragment in the SlMlo1 gene (out of 16 important SlMlo genes), offers resistance against powdery mildew Oidium neolycopersici [105].
In grapevine, loss of VvMLO7 function by RNAi reduced sensitivity against powdery mildew Erysiphe necator [127]. In parallel, the knockout of VvMLO7 and VvMLO3 using CRISPR/Cas9 enhanced resistance to powdery mildew in grapevine [110,111]. In apple (Malus Domestica) protoplasts, RNP-based technology has been successfully used to edit three (DIPM-1, DIPM-2, and DIPM-4) genes to create resistance against fire blight Erwinia amylovora [110]. CRISPR/Cas9 scheme was used to target the VvWRKY52 transcription factor with four guide RNAs. The results showed 21% biallelic mutations in regenerated plants, and these plants confer resistance to the fungus Botrytis cinerea compared with monoallelic mutant plants [109]. To accelerate the GE application in woody plants, another approach based on transient leaf transformation together with disease assays was first demonstrated by researchers in Theobroma cacao [128]. Pathogenesis-Related 3 (NPR3) gene (the immune system suppressor) was targeted in cacao leaves, transiently by CRISPR/Cas9 system, so the leaves showed enhanced resistance against Phytophthora tropicalis. GE of a fungicide resistant gene PcMuORP1 by CRISPR/Cas9 elucidates a novel selection marker for Phytophthora (a genus of oomycetes) species [129]. In rice, CRISPR/Cas9-mediated disruption of OsSEC3A and OsERF922 genes confer resistance against rice blast disease [112,114]. In addition, the pi21 gene in rice also induced durable resistance to rice blast [116]. Furthermore, resistance to Magnaporthe oryzae disease in rice was enhanced by generating the OsSEC3A mutants and showed a pleiotropic type of phenotype with an increase in salicylic acid (SA) concentration, and several genes were induced related to SA- and pathogenesis related genes [112]. To conclude, all these successful fungal disease resistance results determined the advantage, efficacy, and potential of the CRISPR/Cas-based editing system to enhance resistance in crop plants.

2.2. CRISPR/Cas-Mediated Viral Resistance in Plants

Plant viruses are among the most common pathogens and cause hazardous diseases in a variety of economically important crops. There are five main groups based on viral genomes characters: sense-single-stranded-RNA (ssRNA+), antisense-single stranded-RNA (asRNA-), single-stranded-DNA (ssDNA), double-stranded-DNA (dsDNA), and double-stranded-RNA (dsRNA) viruses [130]. A rolling-circle amplification system is required to replicate the virus genome through recombination-mediated duplication or by a dsDNA replicative form [131]. Their genome holds a mutual fragment of 220 bp, which is prearranged in one (A, monopartite) or two (A and B, bipartite) constituents [132]. The Geminiviridae are a large family (over 360 species) of ssDNA plant viruses that cause significant losses to agriculturally and economically important crop plants worldwide [131], such as Malvaceae, Solanaceae, Fabaceae, Euphorbiaceae, and Cucurbitaceae [133]. The commercial term for a large genus of geminiviruses is Begomoviruses. Begomoviruses mostly produce diseases in dicotyledonous plants, for example, Nicotiana tabacum and sweet potato (Ipomoea batatas), and these viruses are mostly transmitted via the whitefly or leafhopper [103,134]. CRISPR/Cas9 system was used in Nicotiana benthamiana and A. thaliana to target two different geminiviruses: Bean yellow dwarf virus (BeYDV) and Beet severe curly top virus (BSCTV), respectively [91,94]. Recently, CRISPR/Cas9 techniques have also been applied to attain resistance against Begomoviruses [90]. In the (BSCTV) genome, 43 candidates were selected to target their coding and non-coding regions using CRISPR/Cas9 [94]. In inoculated leaves, virus accumulation was significantly reduced in all CRISPR/Cas9 constructs at variable levels. However, the highest resistance was observed in A. thaliana and N. tabacum to virus infection displaying the maximum expression level of sgRNAs and Cas9. Similar results have been detected by employing 11 sgRNAs in N. benthamiana, targeting the non-nucleotide sequence, Rep-binding sites, Rep motifs, and the hairpin of BeYDV [91], and decreased up to 87% load of the targeted viral. A tobacco rattle virus (TRV) vector was used to deliver the sgRNA molecules to the N. benthamiana, stably overexpressing the Cas9 endonuclease to target the Tomato yellow leaf curl virus (TYLCV) genome [90]. In that study, the CRISPR/Cas approach was effectively implemented to cleave and target the virus genome during duplication to confer resistance against TYLCV [90,100,135] (Table 2).
By using specific sgRNAs, several genome loci of TYLCV (non-coding and coding sequences) were targeted in their intergenic region (IR), the RCRII motif replication protein (Rep), and the viral capsid protein (CP). Targeting the IR stem-loop invariant structure showed the lowest viral accumulation and replication [90]. A similar CRISPR/Cas9 system was established to target the geminiviruses monopartite beet curly top virus (BCTV), and bipartite Merremia mosaic virus (MerMV), which possess a similar IR stem-loop sequence. CRISPR/Cas9 system-edited BCTV and MerMV viruses displayed tempered symptoms, indicating that combined resistance against various viral strains can be achieved by a single sgRNA specific for the conserved region of the pathogen.
The traditional SpCas9 system recognizes only dsDNA, so the defense against RNA-based viruses is difficult to attain. Nevertheless, the characterization and search for associated nucleases have steered to the discovery of LwaCas13a from (Leptotrichia wadei) and FnCas9 from (Francisella novicida), which have the ability to bind and cut the RNA [102]. FnCas9 was reported to demonstrate resistance against RNA viruses [93]. The sgRNAs designed to target the RNA of cucumber mosaic virus (CMV) and tobacco mosaic virus (TMV) in N. benthamiana and A. thaliana transgenic plants showed a significant reduction in TMV and CMV by 40–80% compared to wild-type (WT) plants [93]. It demonstrated that FnCas9-mediated application could be deliberated as a CRISPR interference (CRISPRi) apparatus, similar to the mitigation of gene expression by catalytically inactive proteins of SpCas9 [136]. A similar study was carried out with Cas13a for manipulating the RNA genome of turnip mosaic virus (TuMV) using RNA-guided ribonuclease [92]. The minimum spread and replication of TuMV was observed in tobacco leaves by using the most proficient virus interference, detected with CRISPR RNA excision of GFP2 and HC-Pro genes.
Furthermore, the pre-CRISPR RNA was processed by Cas13 (due to its innate ability) into functional CRISPR RNA to target many viral mRNAs simultaneously. This may provide an alternative system to improve its efficiency distinctly [92,97,137]. A second strategy is to achieve viral resistance by editing the specific plant genes that are responsible for virus resistance traits [52,95,115]. RNA viruses need plant host factors to preserve their normal life cycle, containing the eukaryotic translation initiation factors eIF4E, eIF4G, and eIF(iso)4E [138]. Host susceptibility gene eIF4E was targeted at two different sites to create resistance against plant potyviruses by CRISPR/Cas9 [52,98,99]. A similar approach in A. thaliana plants induced site-specific mutations at eIF(iso)4E locus and conferred complete resistance to single-stranded RNA potyvirus -TuMV by 1 bp deletions and 1 bp insertions without any off-target modification [95]. Recently, resistance to rice tungro spherical virus (RTSV) was developed by the mutagenesis in eIF4G alleles [115]. In addition, no negative effects on the growth of mutant plants were observed in studies by Macovei et al. and Pyott et al., although additional research should be conducted to verify and test the durability and efficacy of recessive resistance edited plants [95,115].

2.3. CRISPR/Cas-Mediated Bacterial Resistance in Plants

Many pathogenetic bacteria cause diseases in crops, and the crops show several types of symptoms [139]. Compared to fungal and viral resistance, few studies have been reported about the utilization of CRISPR/Cas against bacterial diseases in crop plant species. The Xanthomonas oryzae pv. Oryzae causes host gene expression to induce susceptibility by utilizing the type III transcription-activator-like effectors (TALEs) system. The X. oryzae pv. oryzae effector protein PthXo2 targets the host sucrose transporter gene OsSWEET13 and is recognized as a sensitive gene for pathogen progression. Disease susceptibility was conferred by transferring the indica rice IR24 OsSWEET13 allele to japonica rice Kitaake, while CRISPR/Cas9-mediated mutations in the allele offered resistance against bacterial blight [113]. Recently, a mutation in the promoter of three rice genes confers broad-spectrum resistance against bacterial blight in rice [117]. CRISPR/Cas9 was used to edit the promoter of the Xa13, a pluripotent gene for recessive resistance to bacterial blight in rice to obtain the highly resistant rice that does not affect agronomic traits [118]. Downy mildew resistance 6 (DMR6) is a well-known negative regulator of plant defense. In tomato, DMR6 ortholog SlDMR6-1 was reported to be up-expressed during Pseudomonas syringae pv. tomato pathogen progression and Phytophthora capsici infection [140]. By targeting the SlDMR6-1 (exon-3), the mutated plants conferred wide-spectrum resistance against P. capsici, Xanthomonas gardneri, P. syringae, and X. perforans [108,140,141]. The tomato bacterial speck disease (causal agent Pseudomonas syringae) causes stomatal opening using coronatine (COR) to facilitate bacterial progression. This stomatal response in A. thaliana relies on AtJAZ2 (Jasmonate ZIM-domain-2), a COR co-receptor. The JAZ2 does not have the C-terminal Jas domain (JAZ2Δjas) that avoids stomatal opening using COR [142]. The homologous gene of AtJAZ2 in tomato is SlJAZ2 [104]. Resistance against the model pathogen Pseudomonas syringae pv. tomato DC3000 (Pto) DC3000 was developed by targeting the dominant JAZ2 repressor- SlJAZ2Δjas by using CRISPR/Cas9 technology that prohibited stomatal opening. Improving and refining the CRISPR/Cas9 and CRISPR/Cas12a systems provide a new opportunity to edit perennial crops species such as citrus to introduce resistance against citrus greening disease [143].
After producing successful bacterial disease-resistant tomato and A. thaliana, the CRISPR/Cas9 system recently effectively produced citrus bacterial canker (CBC) (causal agent Xanthomonas citri subsp. citri (X. citri) resistant citrus plants. The X. citri is the most widespread disease in commercially cultivated citrus [41]. CBC resistance was firstly reported in Duncan grapefruit by altering the PthA4 effector binding elements in the promoter of the Lateral Organ Boundaries 1 (CsLOB1) gene [119]. A significant decline in Xcc symptoms was detected in the mutated lines with no additional phenotypic alterations confirming the link between CBC disease susceptibility and CsLOB1 promoter activity Citrus (Citrus sinensis) (Osbeck) Wanjincheng orange [120]. In Wanjincheng orange, editing of CsWRKY22 by CRISPR/Cas9 reduces susceptibility to X. citri [121]. CBC disease resistance was enhanced by deleting the EBEPthA4 sequence completely from both CsLOB1 alleles, and no additional changes were observed in plants with altered CsLOB1 promoter. Recently, the CRISPR/Cas9-FLP/FRT system has been successfully induced in apple cultivars to reduce fire blight susceptibility [144]. In conclusion, these fruitful results demonstrate that CRISPR/Cas has the potential to not only create bacterial resistance in annual and biennial crop species but also confer durable bacterial disease resistance in perennial crop plants.

3. CRISPR/Cas-Mediated Abiotic Stress Resistance in Plants

Abiotic stresses, such as salinity, drought, heavy metals, temperature, etc., pose a significant challenge to crop production and result in a substantial decrease in yield worldwide [145]. Climate change threatens agriculture and food security. Excessive greenhouse gas emissions are responsible for the frequent occurrence of high temperatures and drought stress in crop plants [146,147]. It is predicted that a 1 °C increase in atmospheric temperature will reduce the yield of maize, rice, and wheat by 21–31%, 10–20%, and 6%, respectively [147,148,149]. Notably, the negative effects of such abiotic stresses are more severe in South Asia and Africa, where food scarcity is already prevalent [146]. Thus, the breeding of climate-smart crops that can tolerate abiotic stresses would be a sustainable strategy for addressing these challenges.

3.1. CRISPR/Cas-Mediated Tolerance against Abiotic Stress in Plants

GE techniques, such as CRISPR/Cas systems, have significantly revolutionized crop improvement by enhancing resistance against abiotic stresses [145,150,151]. By activating or suppressing the target genes, GE technology is also an important tool for understanding the functions of genes involved in the resistance against abiotic stresses in plants [152,153]. CRISPR/Cas9 GE techniques have been applied to knockout the negative regulator of salt stress responses in the A. thaliana, Solanum lycopersicum, Triticum aestivum and Hordeum vulgare which are related to drought and salt stress tolerance [154,155,156,157,158,159,160,161]. Modified tomato variety lines showed highly severe symptoms on leaves (leaf wilting) in drought stress conditions compared to WT tomato plants. Knockout of Auxin Response Factor4 (SlARF4) in tomato using CRISPR/Cas9 exhibits strong salt tolerance [155]. Using CRISPR/Cas9 to generate OsDST varieties in indica mega rice cultivar MTU1010 is significant for improving drought and salt tolerance [162]. In tomato, another CRISPR/Cas9-mediated GE for heat tolerance has been accomplished by targeting the SlAGAMOUS-LIKE6 (SIAGL6) gene, resulting in enhanced fruit setting under heat stress [163]. Moreover, OsANN3 and OsMYB30 genes induce knockdown through CRISPR/Cas9 in japonica rice, which enhances the resistance mutant line against cold stress [164,165]. In a refined study, nuclease-deficient Cas9 (dCas9) or nickase Cas9 (nCas9) was fused to Petromyzon marinus cytidine deaminase (PmCDA1) to make point mutations in rice, showing resistance against herbicide in the edited plant lines [65]. In addition, mutagenesis in SiNPR1 by CRISPR/Cas9 was shown to minimize drought stress tolerance in tomato cultivars [166].
Reactive oxygen species (ROS) serve as signaling molecules to regulate gene expression and plant defense against viral pathogens and symbiotic nitrogen fixation between soil rhizobia and the plants [167,168,169]. However, overproduction of ROS, which is a typical response of plants to oxidative and abiotic stresses, can cause a variety of growth abnormalities, including a decrease in photosynthesis rate, increased cell death, and even male sterility, resulting in reduced crop yield [145]. Dozens of genes encoding antioxidant enzymes, such as glutathione S-transferases (GSTs), catalases (CATs), glutathione reductases (GRs), superoxide dismutase (SOD), and numerous peroxidases (PODs), are involved in the elimination of ROS molecules. These genes are known as the R genes and contribute to abiotic stress tolerance [170]. Molecular breeders and geneticists have identified a number of T genes related to abiotic stress tolerance and incorporated them into plants to achieve tolerance. The CRISPR/Cas9 system was recently used to develop genetic plants that constitutively overexpress the maize ARGOS8 gene by altering the natural promoter sides of the ARGOS8 gene with the GOS2 promoter [171] (Table 3). The ARGOS8 edited line showed vital alterations and improvement in grain production under field conditions using drought stress without any production drawback under natural conditions [171]. Knockout of the soybean flowering major gene GmPRR37 using CRISPR/Cas9 exhibited early flowering under natural long-day conditions, providing regionally adapted cultivars for specific regions [172]. Yield potential can be increased through manipulating an ARE1 ortholog related to nitrogen utilization efficiency in wheat by CRISPR/Cas9 [173]. Simultaneous knockout of BnaMAX1 alleles resulted in increased semi-dwarfing and branching phenotypes and more silique production, resulting in improved yield per plant relative to WT, which provides desirable germplasm for further breeding of high yield in rapeseed [174]. Moreover, contaminations of arable soils increased the heavy metals toxicity in crops. However, breeders have improved rice cultivars with a low level of arsenic, cadmium, and radioactive cesium by knocking out the OsARM1, OsNRAMP5, OsNRAMP1, and OsHAK1 genes [175,176,177,178,179]. Recently CRISPR/Cas9 knockout abscisic acid receptor gene (OsPYL) in rice showed increased grain yield in high-temperature stress tolerance and reduced pre-harvest developing plants compared with WT [180]. Additionally, targeted mutagenesis of the OsRR22 and OsmiR535 via CRISPR/Cas9 confers salinity tolerance, and the OsMPT3 gene is an important gene for osmotic regulation in rice [181,182,183].

3.2. CRISPR/Cas-Mediated Herbicide Resistance in Plants

Unwanted weeds grow everywhere in main field crops and compete with the uptake of nutrients and fertilizers. In this situation, the yield of main crops is significantly reduced, which imposes a huge loss to growers around the world. Key herbicides, such as chlorsulfuron, glufosinate, and glyphosate, as well as many selective herbicides, are involved in inhibiting the acetolactate synthase (ALS), 4-hydroxyphenylpyaunate dioxygenase (HPPD), acetyl coenzyme A carboxylase (ACCase), protoporphyrinogen oxidase (PPO), and phytoene desaturase (PDS) [192,193,194]. The use of excessive herbicides damages the crops due to low stress resistance against herbicide chemicals.
Currently, herbicides such as chlorsulfuron are widely used to target ALS1 and ALS2 genes [186]. The CRISPR/Cas9-edited ALS1 gene increases chlorsulfuron herbicides resistance in soybean crops [61]. The rice gene OsEPSPS (5-enolpyruvylshikimate-3-phosphate synthase) was replaced/knocked-in using CRISPR/Cas9 to confer glyphosate resistance in plants [185]. Similarly, point mutations were introduced via CRISPR/Cas9 into the rice ALS gene, conferring herbicide tolerance [48]. Moreover, CRISPR/Cas9 was used to induce the loss-of-function mutation in maize ALS2, conferring tolerance against the herbicide [184]. A CRISPR/Cas9-mediated C287T point mutation in ALS resulted in an amino acid substitution of A96V and two-point mutations (G590 and W483) in FTIP1e, conferring resistance against imazamox [65]. Further manipulation of the CRISPR/Cas system has led to the engineering of herbicide resistance, a single mutation in the ALS gene as the target for base-editing [191]. Sufficient herbicide resistance is conferred in rice by designing large-scale genomic inversion or duplication using CRISPR/Cas9 [187]. The base editors ABE, CBE, and PE have recently been used to improve herbicide resistance [44,80,188,190]. Novel transgene-free herbicide-resistant watermelon varieties were created by base-editing with the potential of immediate field application to facilitate broadleaved weed control [191]. Herbicide-resistant mutants were obtained by direct evolution of the rice OsACC gene through dual cytosine and adenine base editors STEME-1 and STEME-NG [188]. Moreover, the M268T mutation generated in the endogenous OsTubA2 gene by ABE endowed dinitroaniline herbicide resistance in rice without inducing fitness cost [44]. CBE was employed to target ZmALS1 and ZmALS2 generating sulfonylurea herbicide-resistant mutants in maize [80]. The A3A-PBE system was developed for conferring herbicide resistance in allotetraploid Brassica napus [190]. The above studies recommend that each crop needs a particular GE technique and perspective of genome-engineering to improve desired traits, agronomic traits, and yield under abiotic stress (Table 3).

4. CRISPR/Cas Systems of Advances, Limitations, and Prospective Applications

Although the CRISPR/Cas systems exhibit powerful ability in crop genetic improvement, some limitations still need to be overcome in this field [195,196,197,198,199,200]. SpCas9 requires a 5′-NGG-3ʹPAM immediately adjacent to the 20 nt DNA target sequence because it can only recognize NGG PAM sites, which limits its effectiveness. Although this restriction is vital, the goal is to turn off the gene through selective mutagenesis in any situation (Figure 4). Therefore, the main hard work to develop Cas9-like systems is underway, changing PAM sequences or causing the single CRISPR/Cas9 from Streptococcus pyogenes to identify other PAMs. For example, xCas9 is changed from SpCas9, which has been altered to identify a wide range of PAM sequences with GAT, NG, and GAA in mammalian cells [201]. Expanding the scope of CRISPR/Cas9-mediated GE in plants using an xCas9 and Cas9-NG hybrid [202]. A recently developed variant of SpCas9 can target an expanded set of NGN PAMs, and this enzyme was optimized for developing a near-PAMless SpCas9 variant named SpRY (NRN > NYN PAMs). SpRY nuclease and base-editor variants are capable of targeting almost all PAMs [203].
The delivery methods of CRISPR/Cas are divided into direct and indirect approaches. Direct methods include polyethylene glycol (PEG)-mediated delivery and bombardment-mediated delivery. Indirect methods include the floral dip method and Agrobacterium tumefaciens-mediated delivery. Direct gene delivery methods are mostly used for the transient expression of the genes. Indirect delivery based on Agrobacterium tumefaciens-mediated genetic transformation is mostly used in plants [204]. Nearly all GE tools in plants are based on tissue culture and the plant regeneration process. However, the regeneration of many plant species through tissue culture is a genotype dependent, time-consuming, cost-intensive, and laborious process. CRISPR/Cas GE is difficult and challenging in forest woody plants because of their lengthy propagation times, limited mutant resources, and low genetic transformation efficiency [205]. Therefore, to achieve efficient and rapid delivery of the CRISPR/Cas system to plants, the use of a suitable carrier can be considered depending on the purpose of delivery. Delivery vectors are available as plasmid and viral and non-viral vectors. Viral vectors that have been used in plants include bean yellow dwarf virus, tobacco mosaic virus, potato virus X, and cowpea mosaic virus [60,206]. However, the capacity of viral vectors limits the application of large fragment sequences or even large Cas proteins, and the use of viral vectors may stimulate the defense of the plant immune system. These non-viral vectors include a variety of materials, such as inorganic nanoparticles, carbon nanotubes, liposomes, protein- and peptide-based nanoparticles, and nanoscale polymeric materials. These novel non-viral vectors are very promising for future GE applications due to their small size, low toxicity, ability to maintain biological activity, and ability to cross many physical barriers in the domain. For citrus and grapes, Ribonucleoprotein RNP and nano-biotechnology transgene-free editing methods, and transient expression of CRISPR genes, can generate transgene-free and target gene edited plants. However, the efficiency is still low, and intensive labor is required in order to improve the current technology and develop new technologies [207].
CRISPR/Cas9 techniques can apply to other members of the kingdom Plantae, such as bryophytes, algae, and pteridophytes. The model species liverwort has emerged as an example of plant development, and the application of CRISPR/Cas9-mediated targeted mutagenesis studies has been used in the molecular breeding of liverwort Foliage (Marchantia polymorpha L.) [208]. Moreover, new fungus, bacteria, and virus species may be found in nature, or known ones may be sensibly changed [209].
CRISPR/Cas9 may introduce off-target mutations in plants [9,140]. Off-targets can lead to chromosomal rearrangements, causing damage at incompletely matched genomic loci, and limiting the use of GE for therapeutic purposes. Off-target effects may also lead to loss of functional gene activity, resulting in diverse physiological or signaling abnormalities [9]. Recently, whole-genome sequencing has been applied to recognize the cleavage at off-target sites by Cas9 or Cas12a system nucleases in A. thaliana [210], cotton [211] and rice [212]. Bioinformatics tools, such as CCTop (https://crispr.cos.uniheidelberg.de), Cas-OFFinder (http://www.rgenome.net/cas-offinder/), DISCOVER-Seq [213], Systemic evolution of ligands by exponential amplification (SELEX), Integrase-deficient lentivirus (IDLV) capture, High-throughput genomic translocation sequencing (HTGTS), and so on, have been established to manage with this issue [214]. In addition, significant advancements have been made to reduce off-target action of CRISPR/Cas9. For instance, HF-Cas9 [215], HypaCas9 [216], eSpCas9 [217], and Sniper Cas9 showed a significant reduction in off-target levels while absorbent on target action [218]. Improving current delivery methods and developing new methods will reduce barriers to the low-cost application of gene editing in crops (Figure 4). To increase the range of delivery methods, the Agrobacterium, vector and plant genes might be engineered to increase the efficacy of Agrobacterium tumefaciens-mediated transformation [219].
CRISPR/Cas9 system has minimal effects on the control of RNA and DNA viruses. Consequently, the advancement of an acceptable and effective CRISPR system is required to overcome such types of issues against viruses. Findings indicate that the Cas13 proteins (Cas13a, Cas13b, and Cas13c) have high potential as robust RNA regulators for RNA viruses [93]. For example, CRISPR/Cas13a conferred RNA virus resistance in monocot and dicot plants [182]. Targeted site gene editing was performed for designing eIF4E resistance alleles that play essential roles in resistance against virus [96,220], and altering the genes, which are responsible for increased metabolites (phytochemicals) that will boost abiotic and biotic stress tolerance in plants, such as drought stress tolerance, disease resistance (fungi, virus, bacteria, and phytoplasma), enhanced nutritional status, and reduced generation [221,222].
Homologous recombination (knock-in/gene replacement) mediated by CRISPR/Cas has been achieved in plants, but the editing efficiency is low [223,224]. Therefore, there is still a long way to go to achieve efficient gene knock-in by CRISPR/Cas-mediated homologous recombination in plants. The identification of more susceptible genes (S genes) in a crop genome with the new genomics strategy as the target of CRISPR systems can be achieved and remove unwanted traits [101]. On the contrary, more resistant genes (R genes) need to be cloned and knocked-in the crop genome by an improved CRISPR/Cas system via a homologous recombination-mediated DNA repair system. The molecular weight of the Cas9/Cas12a proteins is relatively large, so they cannot be packed into viral vectors for the direct delivery of Cas proteins into the plant cells without plant tissue culture. Scientists need to design several sgRNAs in one vector for multiple gene editing since the stress tolerance trait of the plant is determined by multiple genes. Accessibility of next-generation sequencing technologies will offer more adequate and accurate genome data for the assessment of target genes selection and sgRNA design in various cell types and plant species. The efficiency and accuracy of newly generated genome editing tools, including Base editors and Prime editor, are still far from satisfactory.

5. Conclusions and Future Perspective

Since the 1990s, various genetically modified organisms (GMO), including carrot, canola, Bt-cotton, Bt-potato, glyphosate-resistant soybean, and strawberry, have been approved to be released for beneficial usages, such as food, feed, and processing in many countries. With time, commercial cultivation of genetically modified (GM) food crops, for example, soybeans, corn, and cotton, has expanded in some countries, particularly the United States, Brazil, Argentina, India, Canada, and China. However, public skepticism about accepting GM crops is due to concerns that GM may have adverse effects on the environment or human health [225]. In China, a country with conservative attitudes towards crops and food, nearly 80% of the Chinese public accepts foods labeled as GM-free, about 40% accept GM-labeled foods, and those who are more aware of GM products are more likely to accept GM-labeled foods [226]. Using GE promises to produce crops with high yields, high quality, and good disease resistance. However, public attitudes toward GMOs suggest that people are initially unlikely to accept these plants [225]. People do not accept GE plants because they cannot tell the difference between GMO and GE plants [62,199]. GE plants alter plant traits by introducing small mutations such as deletions, insertions, and targeted mutations using CRISPR/Cas. These GE plants have resulted in significant improvements in their agronomic traits. The mutations produced by GE plants do not leave any foreign DNA behind. Additionally, the gRNA (guide RNA) used in the CRISPR/Cas system is not rDNA (recombinant DNA), so GM and GE are fundamentally different. In recent years, the use of CRISPR/Cas to generate transgene-free plants that obtain the expected agronomic traits without introducing any foreign DNA has been widely reported, thus exempting them from the definition and regulation of GMOs. Characterized by high target programmability, specificity, and robustness, CRISPR/Cas enables precise genetic manipulation of crop species, providing opportunities for creating germplasm with beneficial traits and developing novel, more sustainable agricultural systems [227]. In recent years, CRISPR/Cas has worked as a revolutionary tool with high efficiency to perform targeted GE, and it continues to progress rapidly through the invention of new CRISPR-based editing tools to achieve different goals of genome engineering, such as higher yield, pathogen-resistance, improved nutrients efficiency, and abiotic tolerance in crop species [189,228,229,230,231,232]. Many countries such as the United States, Canada, Brazil, Argentina, and Australia have exempted GE plants of SDN1-type and derived food and feed from their GMO legislation or allowed commercialization based on a simplified case-by-case procedure [233,234]. This will trigger the development of new plant varieties and a range of genome-edited plant products with minor genetic changes are expected to enter the global commodity market soon [63]. As science and technology advance, researchers will further develop various GE tools to meet people’s needs and produce high-quality and safe plants. The government must develop appropriate regulations to regulate the safety of GE plants. The government should also facilitate communication between the public and developers. If people understand the benefits of genome editing-mediated plant breeding and trust the regulations, such transgene-free plants can be gradually integrated into society. A sustainable future for agriculture can be imagined using this new and powerful GE tool.
As researchers, we must not avoid the challenges of providing clarity about CRISPR breeding methods, which promise to be significant for achieving public trust and developing regulatory policies to govern the use of the CRISPR system in agriculture. Whatever challenges remain, the newly developed CRISPR methods are just the tip of the iceberg. These powerful new plant breeding tools can provide a sustainable future for agriculture, and with that possibility comes a responsibility to alleviate the public and scientific worries regarding its usage.

Author Contributions

S.J. and F.D. planned and designed this review. Y.W., N.Z., Q.A., H.M., G.W. (Guanying Wang), L.Y. and X.D. wrote the manuscript and drew the figures. N.H., F.D., G.W. (Guoping Wang) and S.J. contributed to the critical revising of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Key Research and Development Program of China (2021YFD1400800), the National Natural Science Foundation of China (31872077), the Special projects for Foreign Cooperation of Yunnan Province (202003AD150014) to Dr. Ding. Hubei Hongshan Laboratory (2021hszd013) and Fundamental Research Funds for the Central Universities (2021ZKPY003) to Dr. Shuangxia Jin.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Borrelli, V.M.G.; Brambilla, V.; Rogowsky, P.; Marocco, A.; Lanubile, A. The Enhancement of Plant Disease Resistance Using CRISPR/Cas9 Technology. Front. Plant Sci. 2018, 9, 1245. [Google Scholar] [CrossRef] [PubMed]
  2. Rodriguez-Leal, D.; Lemmon, Z.H.; Man, J.; Bartlett, M.E.; Lippman, Z.B. Engineering Quantitative Trait Variation for Crop Improvement by Genome Editing. Cell 2017, 171, 470–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Shaibu, A.S.; Li, B.; Zhang, S.R.; Sun, J.M. Soybean cyst nematode-resistance: Gene identification and breeding strategies. Crop J. 2020, 8, 892–904. [Google Scholar] [CrossRef]
  4. Landa, B.B.; Saponari, M.; Feitosa-Junior, O.R.; Giampetruzzi, A.; Vieira, F.J.D.; Mor, E.; Robatzek, S. Xylella fastidiosa’s relationships: The bacterium, the host plants and the plant microbiome. New Phytol. 2022, 234, 1598–1605. [Google Scholar] [CrossRef]
  5. Dong, O.X.; Ronald, P.C. Genetic Engineering for Disease Resistance in Plants: Recent Progress and Future Perspectives. Plant Physiol. 2019, 180, 26–38. [Google Scholar] [CrossRef] [Green Version]
  6. Dracatos, P.M.; Haghdoust, R.; Singh, D.; Park, R.F. Exploring and exploiting the boundaries of host specificity using the cereal rust and mildew models. New Phytol. 2018, 218, 453–462. [Google Scholar] [CrossRef]
  7. Manghwar, H.; Lindsey, K.; Zhang, X.L.; Jin, S.X. CRISPR/Cas System: Recent Advances and Future Prospects for Genome Editing. Trends Plant Sci. 2019, 24, 1102–1125. [Google Scholar] [CrossRef] [Green Version]
  8. Scheben, A.; Wolter, F.; Batley, J.; Puchta, H.; Edwards, D. Towards CRISPR/Cas crops—Bringing together genomics and genome editing. New Phytol. 2017, 216, 682–698. [Google Scholar] [CrossRef] [Green Version]
  9. Manghwar, H.; Li, B.; Ding, X.; Hussain, A.; Lindsey, K.; Zhang, X.L.; Jin, S.X. CRISPR/Cas Systems in Genome Editing: Methodologies and Tools for sgRNA Design, Off-Target Evaluation, and Strategies to Mitigate Off-Target Effects. Adv. Sci. 2020, 7, 1902312. [Google Scholar] [CrossRef]
  10. Zhang, D.; Hussain, A.; Manghwar, H.; Xie, K.; Xie, S.; Zhao, S.; Larkin, R.M.; Qing, P.; Jin, S.; Ding, F. Genome editing with the CRISPR-Cas system: An art, ethics and global regulatory perspective. Plant Biotechnol. J. 2020, 18, 1651–1669. [Google Scholar] [CrossRef]
  11. Khalil, A.M. The genome editing revolution: Review. J. Genet. Eng. Biotechnol. 2020, 18, 68. [Google Scholar] [CrossRef] [PubMed]
  12. Shan, Q.W.; Wang, Y.P.; Li, J.; Zhang, Y.; Chen, K.L.; Liang, Z.; Zhang, K.; Liu, J.X.; Xi, J.J.; Qiu, J.L.; et al. Targeted genome modification of crop plants using a CRISPR-Cas system. Nat. Biotechnol. 2013, 31, 686–688. [Google Scholar] [CrossRef] [PubMed]
  13. Li, J.F.; Norville, J.E.; Aach, J.; McCormack, M.; Zhang, D.D.; Bush, J.; Church, G.M.; Sheen, J. Multiplex and homologous recombination-mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nat. Biotechnol. 2013, 31, 688–691. [Google Scholar] [CrossRef] [PubMed]
  14. Xing, H.L.; Dong, L.; Wang, Z.P.; Zhang, H.Y.; Han, C.Y.; Liu, B.; Wang, X.C.; Chen, Q.J. A CRISPR/Cas9 toolkit for multiplex genome editing in plants. BMC Plant Biol. 2014, 14, 327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hyun, Y.; Kim, J.; Cho, S.; Choi, Y.; Kim, J.S.; Coupland, G. Site-directed mutagenesis in Arabidopsis thaliana using dividing tissue-targeted RGEN of the CRISPR/Cas system to generate heritable null alleles. Planta 2015, 241, 271–284. [Google Scholar] [CrossRef] [Green Version]
  16. Steinert, J.; Schiml, S.; Fauser, F.; Puchta, H. Highly efficient heritable plant genome engineering using Cas9 orthologues from Streptococcus thermophilus and Staphylococcus aureus. Plant J. 2015, 84, 1295–1305. [Google Scholar] [CrossRef] [PubMed]
  17. Jiang, W.Z.; Zhou, H.B.; Bi, H.H.; Fromm, M.; Yang, B.; Weeks, D.P. Demonstration of CRISPR/Cas9/sgRNA-mediated targeted gene modification in Arabidopsis, tobacco, sorghum and rice. Nucleic Acids Res. 2013, 41, e188. [Google Scholar] [CrossRef]
  18. Yin, K.Q.; Han, T.; Liu, G.; Chen, T.Y.; Wang, Y.; Yu, A.Y.L.; Liu, Y.L. A geminivirus-based guide RNA delivery system for CRISPR/Cas9 mediated plant genome editing. Sci. Rep. 2015, 5, 14926. [Google Scholar] [CrossRef]
  19. Vazquez-Vilar, M.; Bernabe-Orts, J.M.; Fernandez-del-Carmen, A.; Ziarsolo, P.; Blanca, J.; Granell, A.; Orzaez, D. A modular toolbox for gRNA-Cas9 genome engineering in plants based on the GoldenBraid standard. Plant Methods 2016, 12, 10. [Google Scholar] [CrossRef] [Green Version]
  20. Gao, J.P.; Wang, G.H.; Ma, S.Y.; Xie, X.D.; Wu, X.W.; Zhang, X.T.; Wu, Y.Q.; Zhao, P.; Xia, Q.Y. CRISPR/Cas9-mediated targeted mutagenesis in Nicotiana tabacum. Plant Mol. Biol. 2015, 87, 99–110. [Google Scholar] [CrossRef]
  21. Mercx, S.; Tollet, J.; Magy, B.; Navarre, C.; Boutry, M. Gene Inactivation by CRISPR-Cas9 in Nicotiana tabacum BY-2 Suspension Cells. Front. Plant Sci. 2016, 7, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Miao, J.; Guo, D.S.; Zhang, J.Z.; Huang, Q.P.; Qin, G.J.; Zhang, X.; Wan, J.M.; Gu, H.Y.; Qu, L.J. Targeted mutagenesis in rice using CRISPR-Cas system. Cell Res. 2013, 23, 1233–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Xu, R.F.; Li, H.; Qin, R.Y.; Wang, L.; Li, L.; Wei, P.C.; Yang, J.B. Gene targeting using the Agrobacterium tumefaciens-mediated CRISPR-Cas system in rice. Rice 2014, 7, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhang, H.; Zhang, J.S.; Wei, P.L.; Zhang, B.T.; Gou, F.; Feng, Z.Y.; Mao, Y.F.; Yang, L.; Zhang, H.; Xu, N.F.; et al. The CRISPR/Cas9 system produces specific and homozygous targeted gene editing in rice in one generation. Plant Biotechnol. J. 2014, 12, 797–807. [Google Scholar] [CrossRef]
  25. Du, H.Y.; Zeng, X.R.; Zhao, M.; Cui, X.P.; Wang, Q.; Yang, H.; Cheng, H.; Yu, D.Y. Efficient targeted mutagenesis in soybean by TALENs and CRISPR/Cas9. J. Biotechnol. 2016, 217, 90–97. [Google Scholar] [CrossRef]
  26. Bisht, D.S.; Bhatia, V.; Bhattacharya, R. Improving plant-resistance to insect-pests and pathogens: The new opportunities through targeted genome editing. Semin. Cell Dev. Biol. 2019, 96, 65–76. [Google Scholar] [CrossRef]
  27. Gao, C. Genome engineering for crop improvement and future agriculture. Cell 2021, 184, 1621–1635. [Google Scholar] [CrossRef]
  28. Fauser, F.; Schiml, S.; Puchta, H. Both CRISPR/Cas-based nucleases and nickases can be used efficiently for genome engineering in Arabidopsis thaliana. Plant J. 2014, 79, 348–359. [Google Scholar] [CrossRef]
  29. Schiml, S.; Fauser, F.; Puchta, H. 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. 2014, 80, 1139–1150. [Google Scholar] [CrossRef]
  30. Zhao, Y.P.; Zhang, C.S.; Liu, W.W.; Gao, W.; Liu, C.L.; Song, G.Y.; Li, W.X.; Mao, L.; Chen, B.J.; Xu, Y.B.; et al. An alternative strategy for targeted gene replacement in plants using a dual-sgRNA/Cas9 design. Sci. Rep. 2016, 6, 23890. [Google Scholar] [CrossRef]
  31. Ron, M.; Kajala, K.; Pauluzzi, G.; Wang, D.X.; Reynoso, M.A.; Zumstein, K.; Garcha, J.; Winte, S.; Masson, H.; Inagaki, S.; et al. Hairy Root Transformation Using Agrobacterium rhizogenes as a Tool for Exploring Cell Type-Specific Gene Expression and Function Using Tomato as a Model. Plant Physiol. 2014, 166, 455–469. [Google Scholar] [CrossRef] [PubMed]
  32. Ito, Y.; Nishizawa-Yokoi, A.; Endo, M.; Mikami, M.; Toki, S. CRISPR/Cas9-mediated mutagenesis of the RIN locus that regulates tomato fruit ripening. Biochem. Biophys. Res. Commun. 2015, 467, 76–82. [Google Scholar] [CrossRef]
  33. Pan, C.T.; Ye, L.; Qin, L.; Liu, X.; He, Y.J.; Wang, J.; Chen, L.F.; Lu, G. CRISPR/Cas9-mediated efficient and heritable targeted mutagenesis in tomato plants in the first and later generations. Sci. Rep. 2016, 6, 24765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Shan, Q.W.; Wang, Y.P.; Li, J.; Gao, C.X. Genome editing in rice and wheat using the CRISPR/Cas system. Nat. Protoc. 2014, 9, 2395–2410. [Google Scholar] [CrossRef] [PubMed]
  35. Liang, Z.; Zhang, K.; Chen, K.L.; Gao, C.X. Targeted Mutagenesis in Zea mays Using TALENs and the CRISPR/Cas System. J. Genet. Genom. 2014, 41, 63–68. [Google Scholar] [CrossRef] [PubMed]
  36. Zhu, J.J.; Song, N.; Sun, S.L.; Yang, W.L.; Zhao, H.M.; Song, W.B.; Lai, J.S. Efficiency and Inheritance of Targeted Mutagenesis in Maize Using CRISPR-Cas9. J. Genet. Genom. 2016, 43, 25–36. [Google Scholar] [CrossRef]
  37. Feng, C.; Yuan, J.; Wang, R.; Liu, Y.; Birchler, J.A.; Han, F.P. Efficient Targeted Genome Modification in Maize Using CRISPR/Cas9 System. J. Genet. Genom. 2016, 43, 37–43. [Google Scholar] [CrossRef]
  38. Li, C.; Liu, C.; Qi, X.; Wu, Y.; Fei, X.; Mao, L.; Cheng, B.; Li, X.; Xie, C. RNA-guided Cas9 as an in vivo desired-target mutator in maize. Plant Biotechnol. J. 2017, 15, 1566–1576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Dong, L.; Li, L.; Liu, C.; Liu, C.; Geng, S.; Li, X.; Huang, C.; Mao, L.; Chen, S.; Xie, C. Genome Editing and Double-Fluorescence Proteins Enable Robust Maternal Haploid Induction and Identification in Maize. Mol. Plant 2018, 11, 1214–1217. [Google Scholar] [CrossRef] [Green Version]
  40. Lawrenson, T.; Shorinola, O.; Stacey, N.; Li, C.D.; Ostergaard, L.; Patron, N.; Uauy, C.; Harwood, W. Induction of targeted, heritable mutations in barley and Brassica oleracea using RNA-guided Cas9 nuclease. Genome Biol. 2015, 16, 258. [Google Scholar] [CrossRef]
  41. Jia, H.G.; Wang, N. Xcc-facilitated agroinfiltration of citrus leaves: A tool for rapid functional analysis of transgenes in citrus leaves. Plant Cell Rep. 2014, 33, 1993–2001. [Google Scholar] [CrossRef] [PubMed]
  42. Lowder, L.G.; Zhang, D.W.; Baltes, N.J.; Paul, J.W.; Tang, X.; Zheng, X.L.; Voytas, D.F.; Hsieh, T.F.; Zhang, Y.; Qi, Y.P. A CRISPR/Cas9 Toolbox for Multiplexed Plant Genome Editing and Transcriptional Regulation. Plant Physiol. 2015, 169, 971–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wang, M.; Xu, Z.; Gosavi, G.; Ren, B.; Cao, Y.; Kuang, Y.; Zhou, C.; Spetz, C.; Yan, F.; Zhou, X.; et al. Targeted base editing in rice with CRISPR/ScCas9 system. Plant Biotechnol. J. 2020, 18, 1645–1647. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, L.; Kuang, Y.; Yan, F.; Li, S.; Ren, B.; Gosavi, G.; Spetz, C.; Li, X.; Wang, X.; Zhou, X.; et al. Developing a novel artificial rice germplasm for dinitroaniline herbicide resistance by base editing of OsTubA2. Plant Biotechnol. J. 2021, 19, 5–7. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, Y.; Lin, Q.; Li, X.; Wang, F.; Chen, Z.; Wang, J.; Li, W.; Fan, F.; Tao, Y.; Jiang, Y.; et al. Fine-tuning the amylose content of rice by precise base editing of the Wx gene. Plant Biotechnol. J. 2021, 19, 11–13. [Google Scholar] [CrossRef]
  46. Tang, Y.; Abdelrahman, M.; Li, J.; Wang, F.; Ji, Z.; Qi, H.; Wang, C.; Zhao, K. CRISPR/Cas9 induces exon skipping that facilitates development of fragrant rice. Plant Biotechnol. J. 2021, 19, 642–644. [Google Scholar] [CrossRef]
  47. Wang, Y.P.; Cheng, X.; Shan, Q.W.; Zhang, Y.; Liu, J.X.; Gao, C.X.; Qiu, J.L. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat. Biotechnol. 2014, 32, 947–951. [Google Scholar] [CrossRef]
  48. Svitashev, S.; Schwartz, C.; Lenderts, B.; Young, J.K.; Cigan, A.M. Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat. Commun. 2016, 7, 13274. [Google Scholar] [CrossRef] [Green Version]
  49. Dong, L.; Qi, X.; Zhu, J.; Liu, C.; Zhang, X.; Cheng, B.; Mao, L.; Xie, C. Supersweet and waxy: Meeting the diverse demands for specialty maize by genome editing. Plant Biotechnol. J. 2019, 17, 1853–1855. [Google Scholar] [CrossRef]
  50. Wang, Y.; Liu, X.; Zheng, X.; Wang, W.; Yin, X.; Liu, H.; Ma, C.; Niu, X.; Zhu, J.K.; Wang, F. Creation of aromatic maize by CRISPR/Cas. J. Integr. Plant Biol. 2021, 63, 1664–1670. [Google Scholar] [CrossRef]
  51. Liu, L.; Gallagher, J.; Arevalo, E.D.; Chen, R.; Skopelitis, T.; Wu, Q.; Bartlett, M.; Jackson, D. Enhancing grain-yield-related traits by CRISPR-Cas9 promoter editing of maize CLE genes. Nat. Plants 2021, 7, 287–294. [Google Scholar] [CrossRef] [PubMed]
  52. Chandrasekaran, J.; Brumin, M.; Wolf, D.; Leibman, D.; Klap, C.; Pearlsman, M.; Sherman, A.; Arazi, T.; Gal-On, A. Development of broad virus resistance in non-transgenic cucumber using CRISPR/Cas9 technology. Mol. Plant Pathol. 2016, 17, 1140–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Li, B.; Rui, H.; Li, Y.; Wang, Q.; Alariqi, M.; Qin, L.; Sun, L.; Ding, X.; Wang, F.; Zou, J.; et al. Robust CRISPR/Cpf1 (Cas12a)-mediated genome editing in allotetraploid cotton (Gossypium hirsutum). Plant Biotechnol. J. 2019, 17, 1862–1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Qin, L.; Li, J.; Wang, Q.; Xu, Z.; Sun, L.; Alariqi, M.; Manghwar, H.; Wang, G.; Li, B.; Ding, X.; et al. High-efficient and precise base editing of C•G to T•A in the allotetraploid cotton (Gossypium hirsutum) genome using a modified CRISPR/Cas9 system. Plant Biotechnol. J. 2020, 18, 45–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wang, P.; Zhang, J.; Sun, L.; Ma, Y.; Xu, J.; Liang, S.; Deng, J.; Tan, J.; Zhang, Q.; Tu, L.; et al. High efficient multisites genome editing in allotetraploid cotton (Gossypium hirsutum) using CRISPR/Cas9 system. Plant Biotechnol. J. 2018, 16, 137–150. [Google Scholar] [CrossRef] [Green Version]
  56. Wang, Q.; Alariqi, M.; Wang, F.; Li, B.; Ding, X.; Rui, H.; Li, Y.; Xu, Z.; Qin, L.; Sun, L.; et al. The application of a heat-inducible CRISPR/Cas12b (C2c1) genome editing system in tetraploid cotton (G. hirsutum) plants. Plant Biotechnol. J. 2020, 18, 2436–2443. [Google Scholar] [CrossRef]
  57. Chen, Y.; Fu, M.; Li, H.; Wang, L.; Liu, R.; Liu, Z.; Zhang, X.; Jin, S. High-oleic acid content, nontransgenic allotetraploid cotton (Gossypium hirsutum L.) generated by knockout of GhFAD2 genes with CRISPR/Cas9 system. Plant Biotechnol. J. 2021, 19, 424–426. [Google Scholar] [CrossRef]
  58. Li, B.; Liang, S.; Alariqi, M.; Wang, F.; Wang, G.; Wang, Q.; Xu, Z.; Yu, L.; Naeem Zafar, M.; Sun, L.; et al. The application of temperature sensitivity CRISPR/LbCpf1 (LbCas12a) mediated genome editing in allotetraploid cotton (G. hirsutum) and creation of nontransgenic, gossypol-free cotton. Plant Biotechnol. J. 2021, 19, 221–223. [Google Scholar] [CrossRef]
  59. Wang, G.; Xu, Z.; Wang, F.; Huang, Y.; Xin, Y.; Liang, S.; Li, B.; Si, H.; Sun, L.; Wang, Q.; et al. Development of an efficient and precise adenine base editor (ABE) with expanded target range in allotetraploid cotton (Gossypium hirsutum). BMC Biol. 2022, 20, 45. [Google Scholar] [CrossRef]
  60. Li, B.; Fu, C.; Zhou, J.; Hui, F.; Wang, Q.; Wang, F.; Wang, G.; Xu, Z.; Che, L.; Yuan, D.; et al. Highly Efficient Genome Editing Using Geminivirus-Based CRISPR/Cas9 System in Cotton Plant. Cells 2022, 11, 2902. [Google Scholar] [CrossRef]
  61. Li, Z.S.; Liu, Z.B.; Xing, A.Q.; Moon, B.P.; Koellhoffer, J.P.; Huang, L.X.; Ward, R.T.; Clifton, E.; Falco, S.C.; Cigan, A.M. Cas9-Guide RNA Directed Genome Editing in Soybean. Plant Physiol. 2015, 169, 960–970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Gupta, S.; Kumar, A.; Patel, R.; Kumar, V. Genetically modified crop regulations: Scope and opportunity using the CRISPR-Cas9 genome editing approach. Mol. Biol. Rep. 2021, 48, 4851–4863. [Google Scholar] [CrossRef] [PubMed]
  63. Menz, J.; Modrzejewski, D.; Hartung, F.; Wilhelm, R.; Sprink, T. Genome Edited Crops Touch the Market: A View on the Global Development and Regulatory Environment. Front. Plant Sci. 2020, 11, 586027. [Google Scholar] [CrossRef] [PubMed]
  64. Zetsche, B.; Gootenberg, J.S.; Abudayyeh, O.O.; Slaymaker, I.M.; Makarova, K.S.; Essletzbichler, P.; Volz, S.E.; Joung, J.; van der Oost, J.; Regev, A.; et al. Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System. Cell 2015, 163, 759–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Shimatani, Z.; Kashojiya, S.; Takayama, M.; Terada, R.; Arazoe, T.; Ishii, H.; Teramura, H.; Yamamoto, T.; Komatsu, H.; Miura, K.; et al. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 441–443. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, Y.; Ren, Q.; Tang, X.; Liu, S.; Malzahn, A.A.; Zhou, J.; Wang, J.; Yin, D.; Pan, C.; Yuan, M.; et al. Expanding the scope of plant genome engineering with Cas12a orthologs and highly multiplexable editing systems. Nat. Commun. 2021, 12, 1944. [Google Scholar] [CrossRef]
  67. Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.T.; Kellner, M.J.; Regev, A.; et al. RNA targeting with CRISPR-Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef] [Green Version]
  68. Cox, D.B.T.; Gootenberg, J.S.; Abudayyeh, O.O.; Franklin, B.; Kellner, M.J.; Joung, J.; Zhang, F. RNA editing with CRISPR-Cas13. Science 2017, 358, 1019–1027. [Google Scholar] [CrossRef] [Green Version]
  69. Bharat, S.S.; Li, S.Y.; Li, J.Y.; Yan, L.; Xia, L.Q. Base editing in plants: Current status and challenges. Crop J. 2020, 8, 384–395. [Google Scholar] [CrossRef]
  70. Butt, H.; Rao, G.S.; Sedeek, K.; Aman, R.; Kamel, R.; Mahfouz, M. Engineering herbicide resistance via prime editing in rice. Plant Biotechnol. J. 2020, 18, 2370–2372. [Google Scholar] [CrossRef]
  71. Molla, K.A.; Sretenovic, S.; Bansal, K.C.; Qi, Y. Precise plant genome editing using base editors and prime editors. Nat. Plants 2021, 7, 1166–1187. [Google Scholar] [CrossRef] [PubMed]
  72. Nelson, J.W.; Randolph, P.B.; Shen, S.P.; Everette, K.A.; Chen, P.J.; Anzalone, A.V.; An, M.; Newby, G.A.; Chen, J.C.; Hsu, A.; et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 2022, 40, 402–410. [Google Scholar] [CrossRef] [PubMed]
  73. Li, Z.; Zhang, D.; Xiong, X.; Yan, B.; Xie, W.; Sheen, J.; Li, J.-F. A potent Cas9-derived gene activator for plant and mammalian cells. Nat. Plants 2017, 3, 930–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, Y.; Yin, C.; Zhang, T.; Li, F.; Yang, W.; Kaminski, R.; Fagan, P.R.; Putatunda, R.; Young, W.-B.; Khalili, K.; et al. CRISPR/gRNA-directed synergistic activation mediator (SAM) induces specific, persistent and robust reactivation of the HIV-1 latent reservoirs. Sci. Rep. 2015, 5, 16277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Pan, C.; Wu, X.; Markel, K.; Malzahn, A.A.; Kundagrami, N.; Sretenovic, S.; Zhang, Y.; Cheng, Y.; Shih, P.M.; Qi, Y. CRISPR–Act3.0 for highly efficient multiplexed gene activation in plants. Nat. Plants 2021, 7, 942–953. [Google Scholar] [CrossRef]
  76. Ding, X.; Yu, L.; Chen, L.; Li, Y.; Zhang, J.; Sheng, H.; Ren, Z.; Li, Y.; Yu, X.; Jin, S.; et al. Recent Progress and Future Prospect of CRISPR/Cas-Derived Transcription Activation (CRISPRa) System in Plants. Cells 2022, 11, 3045. [Google Scholar] [CrossRef]
  77. Park, J.J.; Dempewolf, E.; Zhang, W.Z.; Wang, Z.Y. RNA-guided transcriptional activation via CRISPR/dCas9 mimics overexpression phenotypes in Arabidopsis. PLoS ONE 2017, 12, e0179410. [Google Scholar] [CrossRef] [Green Version]
  78. Hassan, M.M.; Yuan, G.; Chen, J.-G.; Tuskan, G.A.; Yang, X. Prime Editing Technology and Its Prospects for Future Applications in Plant Biology Research. BioDesign Res. 2020, 2020, 9350905. [Google Scholar] [CrossRef]
  79. Zhang, Y.W.; Bai, Y.; Wu, G.H.; Zou, S.H.; Chen, Y.F.; Gao, C.X.; Tang, D.Z. Simultaneous modification of three homoeologs of TaEDR1 by genome editing enhances powdery mildew resistance in wheat. Plant J. 2017, 91, 714–724. [Google Scholar] [CrossRef] [Green Version]
  80. Li, Y.M.; Zhu, J.J.; Wu, H.; Liu, C.L.; Huang, C.L.; Lan, J.H.; Zhao, Y.M.; Xie, C.X. Precise base editing of non-allelic acetolactate synthase genes confers sulfonylurea herbicide resistance in maize. Crop J. 2020, 8, 449–456. [Google Scholar] [CrossRef]
  81. Bortesi, L.; Fischer, R. The CRISPR/Cas9 system for plant genome editing and beyond. Biotechnol. Adv. 2015, 33, 41–52. [Google Scholar] [CrossRef] [PubMed]
  82. Ma, X.L.; Zhu, Q.L.; Chen, Y.L.; Liu, Y.G. CRISPR/Cas9 Platforms for Genome Editing in Plants: Developments and Applications. Mol. Plant 2016, 9, 961–974. [Google Scholar] [CrossRef] [Green Version]
  83. Wang, S.Y.; Yang, Y.H.; Guo, M.; Zhong, C.Y.; Yan, C.J.; Sun, S.Y. Targeted mutagenesis of amino acid transporter genes for rice quality improvement using the CRISPR/Cas9 system. Crop J. 2020, 8, 457–464. [Google Scholar] [CrossRef]
  84. Christopoulou, M.; Wo, S.R.C.; Kozik, A.; McHale, L.K.; Truco, M.J.; Wroblewski, T.; Michelmore, R.W. Genome-Wide Architecture of Disease Resistance Genes in Lettuce. G3-Genes Genomes Genet. 2015, 5, 2655–2669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Miklis, M.; Consonni, C.; Bhat, R.A.; Lipka, V.; Schulze-Lefert, P.; Panstruga, R. Barley MLO modulates actin-dependent and actin-independent antifungal defense pathways at the cell periphery. Plant Physiol. 2007, 144, 1132–1143. [Google Scholar] [CrossRef] [Green Version]
  86. Li, S.; Lin, D.; Zhang, Y.; Deng, M.; Chen, Y.; Lv, B.; Li, B.; Lei, Y.; Wang, Y.; Zhao, L.; et al. Genome-edited powdery mildew resistance in wheat without growth penalties. Nature 2022, 602, 455–460. [Google Scholar] [CrossRef]
  87. Panstruga, R.; Schulze-Lefert, P. Live and let live: Insights into powdery mildew disease and resistance. Mol. Plant Pathol. 2002, 3, 495–502. [Google Scholar] [CrossRef]
  88. van Esse, H.P.; Reuber, T.L.; van der Does, D. Genetic modification to improve disease resistance in crops. New Phytol. 2020, 225, 70–86. [Google Scholar] [CrossRef] [Green Version]
  89. Arora, L.; Narula, A. Gene Editing and Crop Improvement Using CRISPR-Cas9 System. Front. Plant Sci. 2017, 8, 1932. [Google Scholar] [CrossRef] [Green Version]
  90. Ali, Z.; Abulfaraj, A.; Idris, A.; Ali, S.; Tashkandi, M.; Mahfouz, M.M. CRISPR/Cas9-mediated viral interference in plants. Genome Biol. 2015, 16, 238. [Google Scholar] [CrossRef]
  91. Baltes, N.J.; Hummel, A.W.; Konecna, E.; Cegan, R.; Bruns, A.N.; Bisaro, D.M.; Voytas, D.F. Conferring resistance to geminiviruses with the CRISPR-Cas prokaryotic immune system. Nat. Plants 2015, 1, 15145. [Google Scholar] [CrossRef]
  92. Aman, R.; Ali, Z.; Butt, H.; Mahas, A.; Aljedaani, F.; Khan, M.Z.; Ding, S.W.; Mahfouz, M. RNA virus interference via CRISPR/Cas13a system in plants. Genome Biol. 2018, 19, 1. [Google Scholar] [CrossRef]
  93. Zhang, T.; Zheng, Q.F.; Yi, X.; An, H.; Zhao, Y.L.; Ma, S.Q.; Zhou, G.H. Establishing RNA virus resistance in plants by harnessing CRISPR immune system. Plant Biotechnol. J. 2018, 16, 1415–1423. [Google Scholar] [CrossRef] [Green Version]
  94. Ji, X.; Zhang, H.; Zhang, Y.; Wang, Y.; Gao, C. Establishing a CRISPR-Cas-like immune system conferring DNA virus resistance in plants. Nat. Plants 2015, 1, 15144. [Google Scholar] [CrossRef]
  95. Pyott, D.E.; Sheehan, E.; Molnar, A. Engineering of CRISPR/Cas9-mediated potyvirus resistance in transgene-free Arabidopsis plants. Mol. Plant Pathol. 2016, 17, 1276–1288. [Google Scholar] [CrossRef] [Green Version]
  96. Bastet, A.; Zafirov, D.; Giovinazzo, N.; Guyon-Debast, A.; Nogue, F.; Robaglia, C.; Gallois, J.L. Mimicking natural polymorphism in eIF4E by CRISPR-Cas9 base editing is associated with resistance to potyviruses. Plant Biotechnol. J. 2019, 17, 1736–1750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Zhan, X.; Zhang, F.; Zhong, Z.; Chen, R.; Wang, Y.; Chang, L.; Bock, R.; Nie, B.; Zhang, J. Generation of virus-resistant potato plants by RNA genome targeting. Plant Biotechnol. J. 2019, 17, 1814–1822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Noureen, A.; Khan, M.Z.; Amin, I.; Zainab, T.; Mansoor, S. CRISPR/Cas9-Mediated Targeting of Susceptibility Factor eIF4E-Enhanced Resistance Against Potato Virus Y. Front. Genet. 2022, 13, 922019. [Google Scholar] [CrossRef] [PubMed]
  99. Lucioli, A.; Tavazza, R.; Baima, S.; Fatyol, K.; Burgyan, J.; Tavazza, M. CRISPR-Cas9 Targeting of the eIF4E1 Gene Extends the Potato Virus Y Resistance Spectrum of the Solanum tuberosum L. cv. Desirée. Front. Microbiol. 2022, 13, 873930. [Google Scholar] [CrossRef] [PubMed]
  100. Pramanik, D.; Shelake, R.M.; Park, J.; Kim, M.J.; Hwang, I.; Park, Y.; Kim, J.Y. CRISPR/Cas9-Mediated Generation of Pathogen-Resistant Tomato against Tomato Yellow Leaf Curl Virus and Powdery Mildew. Int. J. Mol. Sci. 2021, 22, 1878. [Google Scholar] [CrossRef]
  101. Santillán Martínez, M.I.; Bracuto, V.; Koseoglou, E.; Appiano, M.; Jacobsen, E.; Visser, R.G.F.; Wolters, A.A.; Bai, Y. CRISPR/Cas9-targeted mutagenesis of the tomato susceptibility gene PMR4 for resistance against powdery mildew. BMC Plant Biol. 2020, 20, 284. [Google Scholar] [CrossRef]
  102. Yu, Y.; Pan, Z.; Wang, X.; Bian, X.; Wang, W.; Liang, Q.; Kou, M.; Ji, H.; Li, Y.; Ma, D.; et al. Targeting of SPCSV-RNase3 via CRISPR-Cas13 confers resistance against sweet potato virus disease. Mol. Plant Pathol. 2022, 23, 104–117. [Google Scholar] [CrossRef]
  103. Kis, A.; Hamar, E.; Tholt, G.; Ban, R.; Havelda, Z. Creating highly efficient resistance against wheat dwarf virus in barley by employing CRISPR/Cas9 system. Plant Biotechnol. J. 2019, 17, 1004–1006. [Google Scholar] [CrossRef] [PubMed]
  104. Ortigosa, A.; Gimenez-Ibanez, S.; Leonhardt, N.; Solano, R. Design of a bacterial speck resistant tomato by CRISPR/Cas9-mediated editing of SlJAZ2. Plant Biotechnol. J. 2019, 17, 665–673. [Google Scholar] [CrossRef] [Green Version]
  105. Nekrasov, V.; Wang, C.M.; Win, J.; Lanz, C.; Weigel, D.; Kamoun, S. Rapid generation of a transgene-free powdery mildew resistant tomato by genome deletion. Sci. Rep. 2017, 7, 482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Silva, C.J.; van den Abeele, C.; Ortega-Salazar, I.; Papin, V.; Adaskaveg, J.A.; Wang, D.; Casteel, C.L.; Seymour, G.B.; Blanco-Ulate, B. Host susceptibility factors render ripe tomato fruit vulnerable to fungal disease despite active immune responses. J. Exp. Bot. 2021, 72, 2696–2709. [Google Scholar] [CrossRef] [PubMed]
  107. Jeon, J.E.; Kim, J.G.; Fischer, C.R.; Mehta, N.; Dufour-Schroif, C.; Wemmer, K.; Mudgett, M.B.; Sattely, E. A Pathogen-Responsive Gene Cluster for Highly Modified Fatty Acids in Tomato. Cell 2020, 180, 176–187.e119. [Google Scholar] [CrossRef]
  108. Thomazella, D.P.T.; Seong, K.; Mackelprang, R.; Dahlbeck, D.; Geng, Y.; Gill, U.S.; Qi, T.; Pham, J.; Giuseppe, P.; Lee, C.Y.; et al. Loss of function of a DMR6 ortholog in tomato confers broad-spectrum disease resistance. Proc. Natl. Acad. Sci. USA 2021, 118, e2026152118. [Google Scholar] [CrossRef]
  109. Wang, W.; Pan, Q.L.; He, F.; Akhunova, A.; Chao, S.M.; Trick, H.; Akhunov, E. Transgenerational CRISPR-Cas9 Activity Facilitates Multiplex Gene Editing in Allopolyploid Wheat. CRISPR J. 2018, 1, 65–74. [Google Scholar] [CrossRef] [Green Version]
  110. Malnoy, M.; Viola, R.; Jung, M.H.; Koo, O.J.; Kim, S.; Kim, J.S.; Velasco, R.; Kanchiswamy, C.N. DNA-Free Genetically Edited Grapevine and Apple Protoplast Using CRISPR/Cas9 Ribonucleoproteins. Front. Plant Sci. 2016, 7, 1904. [Google Scholar] [CrossRef] [PubMed]
  111. Wan, D.Y.; Guo, Y.; Cheng, Y.; Hu, Y.; Xiao, S.; Wang, Y.; Wen, Y.Q. CRISPR/Cas9-mediated mutagenesis of VvMLO3 results in enhanced resistance to powdery mildew in grapevine (Vitis vinifera). Hortic. Res. 2020, 7, 116. [Google Scholar] [CrossRef]
  112. Ma, J.; Chen, J.; Wang, M.; Ren, Y.L.; Wang, S.; Lei, C.L.; Cheng, Z.J. Sodmergen, Disruption of OsSEC3A increases the content of salicylic acid and induces plant defense responses in rice. J. Exp. Bot. 2018, 69, 1051–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhou, J.H.; Peng, Z.; Long, J.Y.; Sosso, D.; Liu, B.; Eom, J.S.; Huang, S.; Liu, S.Z.; Cruz, C.V.; Frommer, W.B.; et al. Gene targeting by the TAL effector PthXo2 reveals cryptic resistance gene for bacterial blight of rice. Plant J. 2015, 82, 632–643. [Google Scholar] [CrossRef]
  114. Wang, F.J.; Wang, C.L.; Liu, P.Q.; Lei, C.L.; Hao, W.; Gao, Y.; Liu, Y.G.; Zhao, K.J. Enhanced Rice Blast Resistance by CRISPR/Cas9-Targeted Mutagenesis of the ERF Transcription Factor Gene OsERF922. PLoS ONE 2016, 11, e0154027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Macovei, A.; Sevilla, N.R.; Cantos, C.; Jonson, G.B.; Slamet-Loedin, I.; Cermak, T.; Voytas, D.F.; Choi, I.R.; Chadha-Mohanty, P. Novel alleles of rice eIF4G generated by CRISPR/Cas9-targeted mutagenesis confer resistance to Rice tungro spherical virus. Plant Biotechnol. J. 2018, 16, 1918–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zhou, Y.; Xu, S.; Jiang, N.; Zhao, X.; Bai, Z.; Liu, J.; Yao, W.; Tang, Q.; Xiao, G.; Lv, C.; et al. Engineering of rice varieties with enhanced resistances to both blast and bacterial blight diseases via CRISPR/Cas9. Plant Biotechnol. J. 2022, 20, 876–885. [Google Scholar] [CrossRef]
  117. Oliva, R.; Ji, C.H.; Atienza-Grande, G.; Huguet-Tapia, J.C.; Perez-Quintero, A.; Li, T.; Eom, J.S.; Li, C.H.; Nguyen, H.; Liu, B.; et al. Broad-spectrum resistance to bacterial blight in rice using genome editing. Nat. Biotechnol. 2019, 37, 1344–1350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Li, C.; Li, W.; Zhou, Z.; Chen, H.; Xie, C.; Lin, Y. A new rice breeding method: CRISPR/Cas9 system editing of the Xa13 promoter to cultivate transgene-free bacterial blight-resistant rice. Plant Biotechnol. J. 2020, 18, 313–315. [Google Scholar] [CrossRef] [Green Version]
  119. Jia, H.; Orbovic, V.; Jones, J.B.; Wang, N. Modification of the PthA4 effector binding elements in Type I CsLOB1 promoter using Cas9/sgRNA to produce transgenic Duncan grapefruit alleviating XccΔpthA4:dCsLOB1.3 infection. Plant Biotechnol. J. 2016, 14, 1291–1301. [Google Scholar] [CrossRef]
  120. Peng, A.H.; Chen, S.C.; Lei, T.G.; Xu, L.Z.; He, Y.R.; Wu, L.; Yao, L.X.; Zou, X.P. Engineering canker-resistant plants through CRISPR/Cas9-targeted editing of the susceptibility gene CsLOB1 promoter in citrus. Plant Biotechnol. J. 2017, 15, 1509–1519. [Google Scholar] [CrossRef]
  121. Wang, L.; Chen, S.; Peng, A.; Xie, Z.; He, Y.; Zou, X. CRISPR/Cas9-mediated editing of CsWRKY22 reduces susceptibility to Xanthomonas citri subsp. citri in Wanjincheng orange (Citrus sinensis (L.) Osbeck). Plant Biotechnol. Rep. 2019, 13, 501–510. [Google Scholar] [CrossRef]
  122. Liu, Y.; Zeng, J.M.; Yuan, C.; Guo, Y.S.; Yu, H.Q.; Li, Y.P.; Huang, C.J. Cas9-PF, an early flowering and visual selection marker system, enhances the frequency of editing event occurrence and expedites the isolation of genome-edited and transgene-free plants. Plant Biotechnol. J. 2019, 17, 1191–1193. [Google Scholar] [CrossRef] [Green Version]
  123. Acevedo-Garcia, J.; Kusch, S.; Panstruga, R. Magical mystery tour: MLO proteins in plant immunity and beyond. New Phytol. 2014, 204, 273–281. [Google Scholar] [CrossRef] [PubMed]
  124. Piffanelli, P.; Ramsay, L.; Waugh, R.; Benabdelmouna, A.; D’Hont, A.; Hollricher, K.; Jorgensen, J.H.; Schulze-Lefert, P.; Panstruga, R. A barley cultivation-associated polymorphism conveys resistance to powdery mildew. Nature 2004, 430, 887–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Consonni, C.; Humphry, M.E.; Hartmann, H.A.; Livaja, M.; Durner, J.; Westphal, L.; Vogel, J.; Lipka, V.; Kemmerling, B.; Schulze-Lefert, P.; et al. Conserved requirement for a plant host cell protein in powdery mildew pathogenesis. Nat. Genet. 2006, 38, 716–720. [Google Scholar] [CrossRef] [PubMed]
  126. Bai, X.D.; Correa, V.R.; Toruno, T.Y.; Ammar, E.D.; Kamoun, S.; Hogenhout, S.A. AY-WB Phytoplasma Secretes a Protein That Targets Plant Cell Nuclei. Mol. Plant-Microbe Interact. 2009, 22, 18–30. [Google Scholar] [CrossRef] [Green Version]
  127. Pessina, S.; Lenzi, L.; Perazzolli, M.; Campa, M.; Dalla Costa, L.; Urso, S.; Vale, G.; Salamini, F.; Velasco, R.; Malnoy, M. Knockdown of MLO genes reduces susceptibility to powdery mildew in grapevine. Hortic. Res. 2016, 3, 16016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Fister, A.S.; Landherr, L.; Maximova, S.N.; Guiltinan, M.J. Transient Expression of CRISPR/Cas9 Machinery Targeting TcNPR3 Enhances Defense Response in Theobroma cacao. Front. Plant Sci. 2018, 9, 268. [Google Scholar] [CrossRef] [Green Version]
  129. Wang, W.; Xue, Z.; Miao, J.; Cai, M.; Zhang, C.; Li, T.; Zhang, B.; Tyler, B.M.; Liu, X. PcMuORP1, an Oxathiapiprolin-Resistance Gene, Functions as a Novel Selection Marker for Phytophthora Transformation and CRISPR/Cas9 Mediated Genome Editing. Front. Microbiol. 2019, 10, 2402. [Google Scholar] [CrossRef]
  130. Roossinck, M.J.; Martin, D.P.; Roumagnac, P. Plant Virus Metagenomics: Advances in Virus Discovery. Phytopathology 2015, 105, 716–727. [Google Scholar] [CrossRef]
  131. Hanley-Bowdoin, L.; Bejarano, E.R.; Robertson, D.; Mansoor, S. Geminiviruses: Masters at redirecting and reprogramming plant processes. Nat. Rev. Microbiol. 2013, 11, 777–788. [Google Scholar] [CrossRef] [PubMed]
  132. Fondong, V.N. Geminivirus protein structure and function. Mol. Plant Pathol. 2013, 14, 635–649. [Google Scholar] [CrossRef] [PubMed]
  133. Zaidi, S.S.E.A.; Tashkandi, M.; Mansoor, S.; Mahfouz, M.M. Engineering Plant Immunity: Using CRISPR/Cas9 to Generate Virus Resistance. Front. Plant Sci. 2016, 7, 1673. [Google Scholar] [CrossRef] [Green Version]
  134. Gilbertson, R.L.; Batuman, O.; Webster, C.G.; Adkins, S. Role of the Insect Supervectors Bemisia tabaci and Frankliniella occidentalis in the Emergence and Global Spread of Plant Viruses. Annu. Rev. Virol. 2015, 2, 67–93. [Google Scholar] [CrossRef] [PubMed]
  135. Ali, Z.; Ali, S.; Tashkandi, M.; Zaidi, S.S.E.A.; Mahfouz, M.M. CRISPR/Cas9-Mediated Immunity to Geminiviruses: Differential Interference and Evasion. Sci. Rep. 2016, 6, 26912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Larson, M.H.; Gilbert, L.A.; Wang, X.W.; Lim, W.A.; Weissman, J.S.; Qi, L.S. CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat. Protoc. 2013, 8, 2180–2196. [Google Scholar] [CrossRef] [Green Version]
  137. Noureen, A.; Zuhaib Khan, M.; Amin, I.; Zainab, T.; Ahmad, N.; Haider, S.; Mansoor, S. Broad-spectrum resistance against multiple PVY-strains by CRSIPR/Cas13 system in Solanum tuberosum crop. GM Crops Food 2022, 13, 97–111. [Google Scholar] [CrossRef] [PubMed]
  138. Sanfacon, H. Plant Translation Factors and Virus Resistance. Viruses 2015, 7, 3392–3419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Schloss, P.D.; Girard, R.A.; Martin, T.; Edwards, J.; Thrash, J.C. Status of the Archaeal and Bacterial Census: An Update. mBio 2016, 7, e00201-16. [Google Scholar] [CrossRef] [Green Version]
  140. Langner, T.; Kamoun, S.; Belhaj, K. CRISPR Crops: Plant Genome Editing Toward Disease Resistance. Annu. Rev. Phytopathol. 2018, 56, 479–512. [Google Scholar] [CrossRef]
  141. Paula de Toledo Thomazella, D.; Brail, Q.; Dahlbeck, D.; Staskawicz, B.J. CRISPR-Cas9 mediated mutagenesis of a DMR6 ortholog in tomato confers broad-spectrum disease resistance. bioRxiv 2016, 064824. [Google Scholar] [CrossRef] [Green Version]
  142. Gimenez-Ibanez, S.; Boter, M.; Ortigosa, A.; Garcia-Casado, G.; Chini, A.; Lewsey, M.G.; Ecker, J.R.; Ntoukakis, V.; Solano, R. JAZ2 controls stomata dynamics during bacterial invasion. New Phytol. 2017, 213, 1378–1392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Sun, L.F.; Nasrullah; Ke, F.Z.; Nie, Z.P.; Wang, P.; Xu, J.G. Citrus Genetic Engineering for Disease Resistance: Past, Present and Future. Int. J. Mol. Sci. 2019, 20, 5256. [Google Scholar] [CrossRef] [Green Version]
  144. Pompili, V.; Dalla Costa, L.; Piazza, S.; Pindo, M.; Malnoy, M. Reduced fire blight susceptibility in apple cultivars using a high-efficiency CRISPR/Cas9-FLP/FRT-based gene editing system. Plant Biotechnol. J. 2020, 18, 845–858. [Google Scholar] [CrossRef] [PubMed]
  145. Zafar, S.A.; Zaidi, S.S.; Gaba, Y.; Singla-Pareek, S.L.; Dhankher, O.P.; Li, X.; Mansoor, S.; Pareek, A. Engineering abiotic stress tolerance via CRISPR/ Cas-mediated genome editing. J. Exp. Bot. 2020, 71, 470–479. [Google Scholar] [CrossRef]
  146. Hasegawa, T.; Fujimori, S.; Havlik, P.; Valin, H.; Bodirsky, B.L.; Doelman, J.C.; Fellmann, T.; Kyle, P.; Koopman, J.F.L.; Lotze-Campen, H.; et al. Risk of increased food insecurity under stringent global climate change mitigation policy. Nat. Clim. Chang. 2018, 8, 699–703. [Google Scholar] [CrossRef] [Green Version]
  147. Asseng, S.; Ewert, F.; Martre, P.; Rotter, R.P.; Lobell, D.B.; Cammarano, D.; Kimball, B.A.; Ottman, M.J.; Wall, G.W.; White, J.W.; et al. Rising temperatures reduce global wheat production. Nat. Clim. Chang. 2015, 5, 143–147. [Google Scholar] [CrossRef]
  148. Yang, H.; Huang, T.Q.; Ding, M.Q.; Lu, D.L.; Lu, W.P. High Temperature during Grain Filling Impacts on Leaf Senescence in Waxy Maize. Agron. J. 2017, 109, 906–916. [Google Scholar] [CrossRef]
  149. Wang, Y.L.; Wang, L.; Zhou, J.X.; Hu, S.B.; Chen, H.Z.; Xiang, J.; Zhang, Y.K.; Zeng, Y.J.; Shi, Q.H.; Zhu, D.F.; et al. Research Progress on Heat Stress of Rice at Flowering Stage. Rice Sci. 2019, 26, 1–10. [Google Scholar] [CrossRef]
  150. Zong, Y.; Wang, Y.P.; Li, C.; Zhang, R.; Chen, K.L.; Ran, Y.D.; Qiu, J.L.; Wang, D.W.; Gao, C.X. Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 438–440. [Google Scholar] [CrossRef]
  151. Karunarathne, S.D.; Han, Y.; Zhang, X.Q.; Li, C. CRISPR/Cas9 gene editing and natural variation analysis demonstrate the potential for HvARE1 in improvement of nitrogen use efficiency in barley. J. Integr. Plant Biol. 2022, 64, 756–770. [Google Scholar] [CrossRef]
  152. Osakabe, Y.; Watanabe, T.; Sugano, S.S.; Ueta, R.; Ishihara, R.; Shinozaki, K.; Osakabe, K. Optimization of CRISPR/Cas9 genome editing to modify abiotic stress responses in plants. Sci. Rep. 2016, 6, 26685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Lou, D.; Wang, H.; Liang, G.; Yu, D. OsSAPK2 Confers Abscisic Acid Sensitivity and Tolerance to Drought Stress in Rice. Front. Plant Sci. 2017, 8, 993. [Google Scholar] [CrossRef] [Green Version]
  154. Wang, L.; Chen, L.; Li, R.; Zhao, R.R.; Yang, M.J.; Sheng, J.P.; Shen, L. Reduced Drought Tolerance by CRISPR/Cas9-Mediated SlMAPK3 Mutagenesis in Tomato Plants. J. Agric. Food Chem. 2017, 65, 8674–8682. [Google Scholar] [CrossRef] [PubMed]
  155. Bouzroud, S.; Gasparini, K.; Hu, G.; Barbosa, M.A.M.; Rosa, B.L.; Fahr, M.; Bendaou, N.; Bouzayen, M.; Zsögön, A.; Smouni, A.; et al. Down Regulation and Loss of Auxin Response Factor 4 Function Using CRISPR/Cas9 Alters Plant Growth, Stomatal Function and Improves Tomato Tolerance to Salinity and Osmotic Stress. Genes 2020, 11, 272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Tran, M.T.; Doan, D.T.H.; Kim, J.; Song, Y.J.; Sung, Y.W.; Das, S.; Kim, E.J.; Son, G.H.; Kim, S.H.; Van Vu, T.; et al. CRISPR/Cas9-based precise excision of SlHyPRP1 domain(s) to obtain salt stress-tolerant tomato. Plant Cell Rep. 2021, 40, 999–1011. [Google Scholar] [CrossRef] [PubMed]
  157. Alam, M.S.; Kong, J.; Tao, R.; Ahmed, T.; Alamin, M.; Alotaibi, S.S.; Abdelsalam, N.R.; Xu, J.H. CRISPR/Cas9 Mediated Knockout of the OsbHLH024 Transcription Factor Improves Salt Stress Resistance in Rice (Oryza sativa L.). Plants 2022, 11, 1184. [Google Scholar] [CrossRef]
  158. Liu, X.; Wu, D.; Shan, T.; Xu, S.; Qin, R.; Li, H.; Negm, M.; Wu, D.; Li, J. The trihelix transcription factor OsGTγ-2 is involved adaption to salt stress in rice. Plant Mol. Biol. 2020, 103, 545–560. [Google Scholar] [CrossRef]
  159. Li, P.; Li, Y.J.; Zhang, F.J.; Zhang, G.Z.; Jiang, X.Y.; Yu, H.M.; Hou, B.K. The Arabidopsis UDP-glycosyltransferases UGT79B2 and UGT79B3, contribute to cold, salt and drought stress tolerance via modulating anthocyanin accumulation. Plant J. 2017, 89, 85–103. [Google Scholar] [CrossRef] [Green Version]
  160. Zheng, M.; Lin, J.; Liu, X.; Chu, W.; Li, J.; Gao, Y.; An, K.; Song, W.; Xin, M.; Yao, Y.; et al. Histone acetyltransferase TaHAG1 acts as a crucial regulator to strengthen salt tolerance of hexaploid wheat. Plant Physiol. 2021, 186, 1951–1969. [Google Scholar] [CrossRef]
  161. Vlčko, T.; Ohnoutková, L. Allelic Variants of CRISPR/Cas9 Induced Mutation in an Inositol Trisphosphate 5/6 Kinase Gene Manifest Different Phenotypes in Barley. Plants 2020, 9, 195. [Google Scholar] [CrossRef] [Green Version]
  162. Santosh Kumar, V.V.; Verma, R.K.; Yadav, S.K.; Yadav, P.; Watts, A.; Rao, M.V.; Chinnusamy, V. CRISPR-Cas9 mediated genome editing of drought and salt tolerance (OsDST) gene in indica mega rice cultivar MTU1010. Physiol. Mol. Biol. Plants 2020, 26, 1099–1110. [Google Scholar] [CrossRef]
  163. Klap, C.; Yeshayahou, E.; Bolger, A.M.; Arazi, T.; Gupta, S.K.; Shabtai, S.; Usadel, B.; Salts, Y.; Barg, R. Tomato facultative parthenocarpy results from SlAGAMOUS-LIKE 6 loss of function. Plant Biotechnol. J. 2017, 15, 634–647. [Google Scholar] [CrossRef]
  164. Shen, C.X.; Que, Z.Q.; Xia, Y.M.; Tang, N.; Li, D.; He, R.H.; Cao, M.L. Knock out of the annexin gene OsAnn3 via CRISPR/Cas9-mediated genome editing decreased cold tolerance in rice. J. Plant Biol. 2017, 60, 539–547. [Google Scholar] [CrossRef]
  165. Zeng, Y.; Wen, J.; Zhao, W.; Wang, Q.; Huang, W. Rational Improvement of Rice Yield and Cold Tolerance by Editing the Three Genes OsPIN5b, GS3, and OsMYB30 With the CRISPR-Cas9 System. Front. Plant Sci. 2019, 10, 1663. [Google Scholar] [CrossRef] [Green Version]
  166. Li, R.; Liu, C.; Zhao, R.; Wang, L.; Chen, L.; Yu, W.; Zhang, S.; Sheng, J.; Shen, L. CRISPR/Cas9-Mediated SlNPR1 mutagenesis reduces tomato plant drought tolerance. BMC Plant Biol. 2019, 19, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Ribeiro, C.W.; Korbes, A.P.; Garighan, J.A.; Jardim-Messeder, D.; Carvalho, F.E.L.; Sousa, R.H.V.; Caverzan, A.; Teixeira, F.K.; Silveira, J.A.G.; Margis-Pinheiro, M. Rice peroxisomal ascorbate peroxidase knockdown affects ROS signaling and triggers early leaf senescence. Plant Sci. 2017, 263, 55–65. [Google Scholar] [CrossRef] [PubMed]
  168. Sinharoy, S.; Liu, C.; Breakspear, A.; Guan, D.; Shailes, S.; Nakashima, J.; Zhang, S.; Wen, J.; Torres-Jerez, I.; Oldroyd, G.; et al. A Medicago truncatula Cystathionine-β-Synthase-like Domain-Containing Protein Is Required for Rhizobial Infection and Symbiotic Nitrogen Fixation. Plant Physiol. 2016, 170, 2204–2217. [Google Scholar] [CrossRef] [Green Version]
  169. Wu, J.G.; Yang, R.X.; Yang, Z.R.; Yao, S.Z.; Zhao, S.S.; Wang, Y.; Li, P.C.; Song, X.W.; Jin, L.; Zhou, T.; et al. ROS accumulation and antiviral defence control by microRNA528 in rice. Nat. Plants 2017, 3, 16203. [Google Scholar] [CrossRef]
  170. Mittler, R. ROS Are Good. Trends Plant Sci. 2017, 22, 11–19. [Google Scholar] [CrossRef]
  171. Shi, J.R.; Gao, H.R.; Wang, H.Y.; Lafitte, H.R.; Archibald, R.L.; Yang, M.Z.; Hakimi, S.M.; Mo, H.; Habben, J.E. ARGOS8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnol. J. 2017, 15, 207–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Wang, L.; Sun, S.; Wu, T.; Liu, L.; Sun, X.; Cai, Y.; Li, J.; Jia, H.; Yuan, S.; Chen, L.; et al. Natural variation and CRISPR/Cas9-mediated mutation in GmPRR37 affect photoperiodic flowering and contribute to regional adaptation of soybean. Plant Biotechnol. J. 2020, 18, 1869–1881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Zhang, J.; Zhang, H.; Li, S.; Li, J.; Yan, L.; Xia, L. Increasing yield potential through manipulating of an ARE1 ortholog related to nitrogen use efficiency in wheat by CRISPR/Cas9. J. Integr. Plant Biol. 2021, 63, 1649–1663. [Google Scholar] [CrossRef]
  174. Zheng, M.; Zhang, L.; Tang, M.; Liu, J.; Liu, H.; Yang, H.; Fan, S.; Terzaghi, W.; Wang, H.; Hua, W. Knockout of two BnaMAX1 homologs by CRISPR/Cas9-targeted mutagenesis improves plant architecture and increases yield in rapeseed (Brassica napus L.). Plant Biotechnol. J. 2020, 18, 644–654. [Google Scholar] [CrossRef] [Green Version]
  175. Nieves-Cordones, M.; Mohamed, S.; Tanoi, K.; Kobayashi, N.I.; Takagi, K.; Vernet, A.; Guiderdoni, E.; Perin, C.; Sentenac, H.; Very, A.A. Production of low-Cs+ rice plants by inactivation of the K+ transporter OsHAK1 with the CRISPR-Cas system. Plant J. 2017, 92, 43–56. [Google Scholar] [CrossRef] [Green Version]
  176. Tang, L.; Mao, B.G.; Li, Y.K.; Lv, Q.M.; Zhang, L.P.; Chen, C.Y.; He, H.J.; Wang, W.P.; Zeng, X.F.; Shao, Y.; et al. Knockout of OsNramp5 using the CRISPR/Cas9 system produces low Cd-accumulating indica rice without compromising yield. Sci. Rep. 2017, 7, 14438. [Google Scholar] [CrossRef] [Green Version]
  177. Chang, J.D.; Huang, S.; Yamaji, N.; Zhang, W.; Ma, J.F.; Zhao, F.J. OsNRAMP1 transporter contributes to cadmium and manganese uptake in rice. Plant Cell Environ. 2020, 43, 2476–2491. [Google Scholar] [CrossRef]
  178. Chu, C.; Huang, R.; Liu, L.; Tang, G.; Xiao, J.; Yoo, H.; Yuan, M. The rice heavy-metal transporter OsNRAMP1 regulates disease resistance by modulating ROS homoeostasis. Plant Cell Environ. 2022, 45, 1109–1126. [Google Scholar] [CrossRef]
  179. Nazir, R.; Mandal, S.; Mitra, S.; Ghorai, M.; Das, N.; Jha, N.K.; Majumder, M.; Pandey, D.K.; Dey, A. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated genome-editing toolkit to enhance salt stress tolerance in rice and wheat. Physiol. Plant. 2022, 174, e13642. [Google Scholar] [CrossRef]
  180. Miao, C.B.; Xiao, L.H.; Huaa, K.; Zou, C.; Zhao, Y.; Bressan, R.A.; Zhu, J.K. Mutations in a subfamily of abscisic acid receptor genes promote rice growth and productivity. Proc. Natl. Acad. Sci. USA 2018, 115, 6058–6063. [Google Scholar] [CrossRef] [PubMed]
  181. Huang, S.C.; Xin, S.C.; Xie, G.Q.; Han, J.; Liu, Z.L.; Wang, B.; Zhang, S.Q.; Wu, Q.Y.; Cheng, X.G. Mutagenesis reveals that the rice OsMPT3 gene is an important osmotic regulatory factor. Crop J. 2020, 8, 465–479. [Google Scholar] [CrossRef]
  182. Zhang, A.N.; Liu, Y.; Wang, F.M.; Li, T.F.; Chen, Z.H.; Kong, D.Y.; Bi, J.G.; Zhang, F.Y.; Luo, X.X.; Wang, J.H.; et al. Enhanced rice salinity tolerance via CRISPR/Cas9-targeted mutagenesis of the OsRR22 gene. Mol. Breed. 2019, 39, 47. [Google Scholar] [CrossRef] [Green Version]
  183. Yue, E.; Cao, H.; Liu, B. OsmiR535, a Potential Genetic Editing Target for Drought and Salinity Stress Tolerance in Oryza sativa. Plants 2020, 9, 1337. [Google Scholar] [CrossRef]
  184. Svitashev, S.; Young, J.K.; Schwartz, C.; Gao, H.R.; Falco, S.C.; Cigan, A.M. Targeted Mutagenesis, Precise Gene Editing, and Site-Specific Gene Insertion in Maize Using Cas9 and Guide RNA. Plant Physiol. 2015, 169, 931–945. [Google Scholar] [CrossRef] [Green Version]
  185. Li, J.; Meng, X.B.; Zong, Y.; Chen, K.L.; Zhang, H.W.; Liu, J.X.; Li, J.Y.; Gao, C.X. Gene replacements and insertions in rice by intron targeting using CRISPR-Cas9. Nat. Plants 2016, 2, 16139. [Google Scholar] [CrossRef] [PubMed]
  186. Sun, Y.W.; Zhang, X.; Wu, C.Y.; He, Y.B.; Ma, Y.Z.; Hou, H.; Guo, X.P.; Du, W.M.; Zhao, Y.D.; Xia, L.Q. Engineering Herbicide-Resistant Rice Plants through CRISPR/Cas9-Mediated Homologous Recombination of Acetolactate Synthase. Mol. Plant 2016, 9, 628–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Lu, Y.; Wang, J.; Chen, B.; Mo, S.; Lian, L.; Luo, Y.; Ding, D.; Ding, Y.; Cao, Q.; Li, Y.; et al. A donor-DNA-free CRISPR/Cas-based approach to gene knock-up in rice. Nat. Plants 2021, 7, 1445–1452. [Google Scholar] [CrossRef]
  188. Li, C.; Zhang, R.; Meng, X.; Chen, S.; Zong, Y.; Lu, C.; Qiu, J.L.; Chen, Y.H.; Li, J.; Gao, C. Targeted, random mutagenesis of plant genes with dual cytosine and adenine base editors. Nat. Biotechnol. 2020, 38, 875–882. [Google Scholar] [CrossRef]
  189. Zhang, R.; Liu, J.X.; Chai, Z.Z.; Chen, S.; Bai, Y.; Zong, Y.; Chen, K.L.; Li, J.Y.; Jiang, L.J.; Gao, C.X. Generation of herbicide tolerance traits and a new selectable marker in wheat using base editing. Nat. Plants 2019, 5, 480–485. [Google Scholar] [CrossRef] [PubMed]
  190. Cheng, H.; Hao, M.; Ding, B.; Mei, D.; Wang, W.; Wang, H.; Zhou, R.; Liu, J.; Li, C.; Hu, Q. Base editing with high efficiency in allotetraploid oilseed rape by A3A-PBE system. Plant Biotechnol. J. 2021, 19, 87–97. [Google Scholar] [CrossRef]
  191. Tian, S.; Jiang, L.; Cui, X.; Zhang, J.; Guo, S.; Li, M.; Zhang, H.; Ren, Y.; Gong, G.; Zong, M.; et al. Engineering herbicide-resistant watermelon variety through CRISPR/Cas9-mediated base-editing. Plant Cell Rep. 2018, 37, 1353–1356. [Google Scholar] [CrossRef] [PubMed]
  192. Soares da Costa, T.P.; Hall, C.J.; Panjikar, S.; Wyllie, J.A.; Christoff, R.M.; Bayat, S.; Hulett, M.D.; Abbott, B.M.; Gendall, A.R.; Perugini, M.A. Towards novel herbicide modes of action by inhibiting lysine biosynthesis in plants. Elife 2021, 10, e69444. [Google Scholar] [CrossRef] [PubMed]
  193. Green, J.M. Current state of herbicides in herbicide-resistant crops. Pest Manag. Sci. 2014, 70, 1351–1357. [Google Scholar] [CrossRef]
  194. Hall, C.J.; Mackie, E.R.; Gendall, A.R.; Perugini, M.A.; Soares da Costa, T.P. Review: Amino acid biosynthesis as a target for herbicide development. Pest Manag. Sci. 2020, 76, 3896–3904. [Google Scholar] [CrossRef]
  195. Ding, D.; Chen, K.; Chen, Y.; Li, H.; Xie, K. Engineering Introns to Express RNA Guides for Cas9- and Cpf1-Mediated Multiplex Genome Editing. Mol. Plant 2018, 11, 542–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Liu, C.; Moschou, P.N. Phenotypic novelty by CRISPR in plants. Dev. Biol. 2018, 435, 170–175. [Google Scholar] [CrossRef]
  197. Negishi, K.; Kaya, H.; Abe, K.; Hara, N.; Saika, H.; Toki, S. An adenine base editor with expanded targeting scope using SpCas9-NGv1 in rice. Plant Biotechnol. J. 2019, 17, 1476–1478. [Google Scholar] [CrossRef]
  198. Li, Y.; Wu, X.; Zhang, Y.; Zhang, Q. CRISPR/Cas genome editing improves abiotic and biotic stress tolerance of crops. Front. Genome. Ed. 2022, 4, 987817. [Google Scholar] [CrossRef] [PubMed]
  199. Kaur, H.; Pandey, D.K.; Goutam, U.; Kumar, V. CRISPR/Cas9-mediated genome editing is revolutionizing the improvement of horticultural crops: Recent advances and future prospects. Sci. Hortic. 2021, 289, 110476. [Google Scholar] [CrossRef]
  200. Razzaq, M.K.; Akhter, M.; Ahmad, R.M.; Cheema, K.L.; Hina, A.; Karikari, B.; Raza, G.; Xing, G.; Gai, J.; Khurshid, M. CRISPR-Cas9 based stress tolerance: New hope for abiotic stress tolerance in chickpea (Cicer arietinum). Mol. Biol. Rep. 2022, 49, 8977–8985. [Google Scholar] [CrossRef]
  201. Hu, J.H.; Miller, S.M.; Geurts, M.H.; Tang, W.X.; Chen, L.W.; Sun, N.; Zeina, C.M.; Gao, X.; Rees, H.A.; Lin, Z.; et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 2018, 556, 57–63. [Google Scholar] [CrossRef]
  202. Niu, Q.; Wu, S.; Li, Y.; Yang, X.; Liu, P.; Xu, Y.; Lang, Z. Expanding the scope of CRISPR/Cas9-mediated genome editing in plants using an xCas9 and Cas9-NG hybrid. J. Integr. Plant Biol. 2020, 62, 398–402. [Google Scholar] [CrossRef] [Green Version]
  203. Walton, R.T.; Christie, K.A.; Whittaker, M.N.; Kleinstiver, B.P. Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science 2020, 368, 290–296. [Google Scholar] [CrossRef]
  204. Alghuthaymi, M.A.; Ahmad, A.; Khan, Z.; Khan, S.H.; Ahmed, F.K.; Faiz, S.; Nepovimova, E.; Kuča, K.; Abd-Elsalam, K.A. Exosome/Liposome-like Nanoparticles: New Carriers for CRISPR Genome Editing in Plants. Int. J. Mol. Sci. 2021, 22, 7456. [Google Scholar] [CrossRef]
  205. Fan, D.; Liu, T.T.; Li, C.F.; Jiao, B.; Li, S.; Hou, Y.S.; Luo, K.M. Efficient CRISPR/Cas9-mediated Targeted Mutagenesis in Populus in the First Generation. Sci. Rep. 2015, 5, 12217. [Google Scholar] [CrossRef] [Green Version]
  206. Varanda, C.M.; Félix, M.D.R.; Campos, M.D.; Patanita, M.; Materatski, P. Plant Viruses: From Targets to Tools for CRISPR. Viruses 2021, 13, 141. [Google Scholar] [CrossRef] [PubMed]
  207. He, Y.; Zhao, Y. Technological breakthroughs in generating transgene-free and genetically stable CRISPR-edited plants. aBIOTECH 2020, 1, 88–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Sugano, S.S.; Shirakawa, M.; Takagi, J.; Matsuda, Y.; Shimada, T.; Hara-Nishimura, I.; Kohchi, T. CRISPR/Cas9-mediated targeted mutagenesis in the liverwort Marchantia polymorpha L. Plant Cell Physiol. 2014, 55, 475–481. [Google Scholar] [CrossRef] [PubMed]
  209. Lacroix, B.; Citovsky, V. A Functional Bacterium-to-Plant DNA Transfer Machinery of Rhizobium etli. PLoS Pathog. 2016, 12, e1005502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Feng, Z.Y.; Mao, Y.F.; Xu, N.F.; Zhang, B.T.; Wei, P.L.; Yang, D.L.; Wang, Z.; Zhang, Z.J.; Zheng, R.; Yang, L.; et al. Multigeneration analysis reveals the inheritance, specificity, and patterns of CRISPR/Cas-induced gene modifications in Arabidopsis. Proc. Natl. Acad. Sci. USA 2014, 111, 4632–4637. [Google Scholar] [CrossRef]
  211. Li, J.; Manghwar, H.; Sun, L.; Wang, P.; Wang, G.; Sheng, H.; Zhang, J.; Liu, H.; Qin, L.; Rui, H.; et al. Whole genome sequencing reveals rare off-target mutations and considerable inherent genetic or/and somaclonal variations in CRISPR/Cas9-edited cotton plants. Plant Biotechnol. J. 2019, 17, 858–868. [Google Scholar] [CrossRef] [PubMed]
  212. Tang, X.; Liu, G.Q.; Zhou, J.P.; Ren, Q.R.; You, Q.; Tian, L.; Xin, X.H.; Zhong, Z.H.; Liu, B.L.; Zheng, X.L.; et al. A large-scale whole-genome sequencing analysis reveals highly specific genome editing by both Cas9 and Cpf1 (Cas12a) nucleases in rice. Genome Biol. 2018, 19, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Wienert, B.; Wyman, S.K.; Richardson, C.D.; Yeh, C.D.; Akcakaya, P.; Porritt, M.J.; Morlock, M.; Vu, J.T.; Kazane, K.R.; Watry, H.L.; et al. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq. Science 2019, 364, 286–2879. [Google Scholar] [CrossRef]
  214. Koo, T.; Lee, J.; Kim, J.S. Measuring and Reducing Off-Target Activities of Programmable Nucleases Including CRISPR-Cas9. Mol. Cells 2015, 38, 475–481. [Google Scholar] [CrossRef] [Green Version]
  215. Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.L.; Joung, J.K. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Chen, J.S.; Dagdas, Y.S.; Kleinstiver, B.P.; Welch, M.M.; Sousa, A.A.; Harrington, L.B.; Sternberg, S.H.; Joung, J.K.; Yildiz, A.; Doudna, J.A. Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature 2017, 550, 407–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Slaymaker, I.M.; Gao, L.Y.; Zetsche, B.; Scott, D.A.; Yan, W.X.; Zhang, F. Rationally engineered Cas9 nucleases with improved specificity. Science 2016, 351, 84–88. [Google Scholar] [CrossRef] [Green Version]
  218. Lee, J.K.; Jeong, E.; Lee, J.; Jung, M.; Shin, E.; Kim, Y.H.; Lee, K.; Jung, I.; Kim, D.; Kim, S.; et al. Directed evolution of CRISPR-Cas9 to increase its specificity. Nat. Commun. 2018, 9, 3048. [Google Scholar] [CrossRef] [Green Version]
  219. Ahmad, S.; Wei, X.J.; Sheng, Z.H.; Hu, P.S.; Tang, S.Q. CRISPR/Cas9 for development of disease resistance in plants: Recent progress, limitations and future prospects. Brief. Funct. Genom. 2020, 19, 26–39. [Google Scholar] [CrossRef] [PubMed]
  220. Bastet, A.; Lederer, B.; Giovinazzo, N.; Arnoux, X.; German-Retana, S.; Reinbold, C.; Brault, V.; Garcia, D.; Djennane, S.; Gersch, S.; et al. Trans-species synthetic gene design allows resistance pyramiding and broad-spectrum engineering of virus resistance in plants. Plant Biotechnol. J. 2018, 16, 1569–1581. [Google Scholar] [CrossRef]
  221. Rao, M.J.; Ding, F.; Wang, N.; Deng, X.X.; Xu, Q. Metabolic Mechanisms of Host Species Against Citrus Huanglongbing (Greening Disease). Crit. Rev. Plant Sci. 2018, 37, 496–511. [Google Scholar] [CrossRef]
  222. Xu, Z.Y.; Xu, X.M.; Gong, Q.; Li, Z.Y.; Li, Y.; Wang, S.; Yang, Y.Y.; Ma, W.X.; Liu, L.Y.; Zhu, B.; et al. Engineering Broad-Spectrum Bacterial Blight Resistance by Simultaneously Disrupting Variable TALE-Binding Elements of Multiple Susceptibility Genes in Rice. Mol. Plant 2019, 12, 1434–1446. [Google Scholar] [CrossRef] [Green Version]
  223. Puchta, H.; Fauser, F. Synthetic nucleases for genome engineering in plants: Prospects for a bright future. Plant J. 2014, 78, 727–741. [Google Scholar] [CrossRef]
  224. Lu, Y.M.; Tian, Y.F.; Shen, R.D.; Yao, Q.; Wang, M.G.; Chen, M.; Dong, J.S.; Zhang, T.G.; Li, F.; Lei, M.G.; et al. Targeted, efficient sequence insertion and replacement in rice. Nat. Biotechnol. 2020, 38, 1402–1407. [Google Scholar] [CrossRef]
  225. Ishii, T.; Araki, M. Consumer acceptance of food crops developed by genome editing. Plant Cell Rep. 2016, 35, 1507–1518. [Google Scholar] [CrossRef] [PubMed]
  226. Zhao, Y.; Deng, H.; Yu, C.; Hu, R. The Chinese public’s awareness and attitudes toward genetically modified foods with different labeling. NPJ Sci. Food 2019, 3, 17. [Google Scholar] [CrossRef] [Green Version]
  227. Zhu, H.; Li, C.; Gao, C. Applications of CRISPR–Cas in agriculture and plant biotechnology. Nat. Rev. Mol. Cell Biol. 2020, 21, 661–677. [Google Scholar] [CrossRef] [PubMed]
  228. Hussain, B.; Lucas, S.J.; Budak, H. CRISPR/Cas9 in plants: At play in the genome and at work for crop improvement. Brief. Funct. Genom. 2018, 17, 319–328. [Google Scholar] [CrossRef]
  229. Yin, K.Q.; Qiu, J.L. Genome editing for plant disease resistance: Applications and perspectives. Philos. Trans. R. Soc. B Biol. Sci. 2019, 374, 20180322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Barman, H.N.; Sheng, Z.H.; Fiaz, S.; Zhong, M.; Wu, Y.W.; Cai, Y.C.; Wang, W.; Jiao, G.A.; Tang, S.Q.; Wei, X.J.; et al. Generation of a new thermo-sensitive genic male sterile rice line by targeted mutagenesis of TMS5 gene through CRISPR/Cas9 system. BMC Plant Biol. 2019, 19, 109. [Google Scholar] [CrossRef]
  231. Sashidhar, N.; Harloff, H.J.; Potgieter, L.; Jung, C. Gene editing of three BnITPK genes in tetraploid oilseed rape leads to significant reduction of phytic acid in seeds. Plant Biotechnol. J. 2020, 18, 2241–2250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Chao, S.F.; Cai, Y.C.; Feng, B.B.; Jiao, G.A.; Sheng, Z.H.; Luo, J.; Tang, S.Q.; Wang, J.L.; Hu, P.S.; Wei, X.J. Editing of Rice Isoamylase Gene ISA1 Provides Insights into Its Function in Starch Formation. Rice Sci. 2019, 26, 77–87. [Google Scholar] [CrossRef]
  233. Spök, A.; Sprink, T.; Allan, A.C.; Yamaguchi, T.; Dayé, C. Towards social acceptability of genome-edited plants in industrialised countries? Emerging evidence from Europe, United States, Canada, Australia, New Zealand, and Japan. Front. Genome Ed. 2022, 4, 899331. [Google Scholar] [CrossRef] [PubMed]
  234. Schmidt, S.M.; Belisle, M.; Frommer, W.B. The evolving landscape around genome editing in agriculture: Many countries have exempted or move to exempt forms of genome editing from GMO regulation of crop plants. EMBO Rep. 2020, 21, e50680. [Google Scholar] [CrossRef]
Figure 1. Evolution of crop breeding techniques. Crossbreeding takes a great deal of time (8–10 years) to improve desirable characters/traits (in a particular species for disease tolerance or resistance) through crossing an elite variety line with a donor variety line and selecting the new outstanding offspring with desirable characters/traits. To introduce new progeny with desirable traits from the donor variety line to the elite variety line, the selected offspring must be backcrossed to the elite variety line for several years to remove undesirable related traits. In mutation breeding, mutations are used to improve traits in the time (6–7 years) of the genome through chemical treatment or physical irradiation to create novel genetic variations. Transgenic breeding is easy and well-known, improving crop traits within (4–6 years) by the exogenous transformation of genes into economically important elite varieties. Genome editing: improving a targeted trait in a very fast and short time (2–3 years) and exactly revising the target gene or regulatory sequence or changing the DNA and/or RNA bases in elite varieties.
Figure 1. Evolution of crop breeding techniques. Crossbreeding takes a great deal of time (8–10 years) to improve desirable characters/traits (in a particular species for disease tolerance or resistance) through crossing an elite variety line with a donor variety line and selecting the new outstanding offspring with desirable characters/traits. To introduce new progeny with desirable traits from the donor variety line to the elite variety line, the selected offspring must be backcrossed to the elite variety line for several years to remove undesirable related traits. In mutation breeding, mutations are used to improve traits in the time (6–7 years) of the genome through chemical treatment or physical irradiation to create novel genetic variations. Transgenic breeding is easy and well-known, improving crop traits within (4–6 years) by the exogenous transformation of genes into economically important elite varieties. Genome editing: improving a targeted trait in a very fast and short time (2–3 years) and exactly revising the target gene or regulatory sequence or changing the DNA and/or RNA bases in elite varieties.
Cells 11 03928 g001
Figure 2. The methodology of major CRISPR/Cas systems. (A) CRISPR/Cas9 induces double-stranded breaks (DSBs) in DNA strands. (B) CRISPR/Cas12a cleaves the target DNA and introduces DSBs. (C) CRISPR/Cas methods can achieve different research goals: (a–c) are results of non-homologous end-joining NHEJ, and (d,e) are results of the homology-directed repair HDR repair pathways using a donor DNA template. (DF) Base editing tools mainly include Cytidine Base Editor (CBE), Adenine Base Editor (ABE), and Prime Editor (PE). (D) CBE converts C-G base pairs to T-A base pairs at the target site. (E) ABE converts A-T base pairs to G-C base pairs at the target site. (F) PE is a new base editing system, which enables precise sequence substitution, insertion, and deletion. PE mainly consists of a Cas9 nickase (nCas9), an engineered reverse transcriptase (RT), and pegRNA. PegRNA includes PBS (Primer Binding Site) sequence and RT Template. (G) CRISPR/Cas13 consists of a Cas13, a crRNA, and a target RNA. Cas13:crRNA complexes bind target RNA and cleave the target RNA. (H) CRISPR transcriptional activation (CRISPRa) consists of a nuclease-deficient Cas9 (dCas9) and transcription activation domain (TAD). CRISPRa activates the transcription of single or multiple target genes.
Figure 2. The methodology of major CRISPR/Cas systems. (A) CRISPR/Cas9 induces double-stranded breaks (DSBs) in DNA strands. (B) CRISPR/Cas12a cleaves the target DNA and introduces DSBs. (C) CRISPR/Cas methods can achieve different research goals: (a–c) are results of non-homologous end-joining NHEJ, and (d,e) are results of the homology-directed repair HDR repair pathways using a donor DNA template. (DF) Base editing tools mainly include Cytidine Base Editor (CBE), Adenine Base Editor (ABE), and Prime Editor (PE). (D) CBE converts C-G base pairs to T-A base pairs at the target site. (E) ABE converts A-T base pairs to G-C base pairs at the target site. (F) PE is a new base editing system, which enables precise sequence substitution, insertion, and deletion. PE mainly consists of a Cas9 nickase (nCas9), an engineered reverse transcriptase (RT), and pegRNA. PegRNA includes PBS (Primer Binding Site) sequence and RT Template. (G) CRISPR/Cas13 consists of a Cas13, a crRNA, and a target RNA. Cas13:crRNA complexes bind target RNA and cleave the target RNA. (H) CRISPR transcriptional activation (CRISPRa) consists of a nuclease-deficient Cas9 (dCas9) and transcription activation domain (TAD). CRISPRa activates the transcription of single or multiple target genes.
Cells 11 03928 g002
Figure 3. Future applications of CRISPR/Cas in plants against the biotic and abiotic stress. CRISPR/Cas represents the future of genome editing technology and the potential use of the CRISPR/Cas system in various disciplines under biotic and abiotic stresses of agriculture. With the maturity of genome editing (GE) technology and the development of new GE tools, the application of CRISPR/Cas is becoming more and more extensive. CRISPR/Cas can now achieve gene knockout, knock-in, and knock-up in plants, replacing a single base to cause amino acid changes, etc. Therefore, CRISPR/Cas can be used to modify key genes of biotic and abiotic stresses, improving crop growth and development and coping with various environmental stresses to create more germplasm resources that meet human needs.
Figure 3. Future applications of CRISPR/Cas in plants against the biotic and abiotic stress. CRISPR/Cas represents the future of genome editing technology and the potential use of the CRISPR/Cas system in various disciplines under biotic and abiotic stresses of agriculture. With the maturity of genome editing (GE) technology and the development of new GE tools, the application of CRISPR/Cas is becoming more and more extensive. CRISPR/Cas can now achieve gene knockout, knock-in, and knock-up in plants, replacing a single base to cause amino acid changes, etc. Therefore, CRISPR/Cas can be used to modify key genes of biotic and abiotic stresses, improving crop growth and development and coping with various environmental stresses to create more germplasm resources that meet human needs.
Cells 11 03928 g003
Figure 4. Limitations of the current CRISPR/Cas system. Using the CRISPR/Cas system in plants requires Agrobacterium tumefaciens-mediated transformation, but it is a time-consuming, cost-intensive, and laborious process. The selection of target genes is very limited. On the one hand, the function of resistance genes is redundant, and knocking down a gene alone cannot achieve resistance. Conversely, the knockout of resistance genes is restricted by PAM, and sequences close to PAM must be selected. CRISPR/Cas may introduce random off-target mutations in the plant genome. The commercialization of CRISPR-edited crops has been disrupted as Cas proteins take many generations to be completely isolated and obtain transgene-free crops. Currently, the homologous recombination pathway (knock-in/gene replacement) is less efficient, and the efficiency of homologous donor sequence transformation into plant cells is low, resulting in low difficulty and efficiency of knock-in. Therefore, the use of CRISPR/Cas-mediated homologous recombination in plants still has a long way to go for efficient gene knock-in.
Figure 4. Limitations of the current CRISPR/Cas system. Using the CRISPR/Cas system in plants requires Agrobacterium tumefaciens-mediated transformation, but it is a time-consuming, cost-intensive, and laborious process. The selection of target genes is very limited. On the one hand, the function of resistance genes is redundant, and knocking down a gene alone cannot achieve resistance. Conversely, the knockout of resistance genes is restricted by PAM, and sequences close to PAM must be selected. CRISPR/Cas may introduce random off-target mutations in the plant genome. The commercialization of CRISPR-edited crops has been disrupted as Cas proteins take many generations to be completely isolated and obtain transgene-free crops. Currently, the homologous recombination pathway (knock-in/gene replacement) is less efficient, and the efficiency of homologous donor sequence transformation into plant cells is low, resulting in low difficulty and efficiency of knock-in. Therefore, the use of CRISPR/Cas-mediated homologous recombination in plants still has a long way to go for efficient gene knock-in.
Cells 11 03928 g004
Table 1. CRISPR/Cas system applied in major plant species.
Table 1. CRISPR/Cas system applied in major plant species.
Plants SpeciesCodon-OptimizationTarget GeneCas PromotersgRNA PromoterMutation Frequency (%)References
Arabidopsis thalianaArabidopsis codon optimizedADH1, TT4, and RTEL1PcUbi4-2AtU6-262.5–70.0 [28]
Arabidopsis thalianaArabidopsis codon-optimizedADH1PcUbi4-2AtU6HDR 42.8 [29,30]
Arabidopsis thalianaMaize codon-optimizedTRY, CPC, and ETC22_35SU6-26 and U6-2942–90 [14]
Arabidopsis thalianaHuman codon-optimizedFT and SPL4AtICU2AtU610.00–84.78 [15]
Arabidopsis thalianaStreptococcus thermophilus and Staphylococcus aurousADH1PcUbi4-2AtU6-266.1–98.5 [16]
Citrus sinensisHuman codon-optimizedCsPDSCaMV 35SCaMV 35 S3.2–3.9 [41]
Nicotiana benthamianaChlamydomonas reinhardtii codon-optimizedGFPCaMV 35SAtU6-26N/A [17]
Nicotiana benthamianaPlant codon-optimizedNbFLS2 and NbBAK135SAtU3 and AtU6N/A [42]
Nicotiana benthamianaPlant codon-optimizedNbPDS and NbIspH35SAtU6-2675–85 [18]
Nicotiana benthamianaPlant and human codon-optimizedXT35SU6-2611 [19]
Nicotiana tabacumPlant codon-optimizedNtPDS and NtPDR62_35SAtU6-2681.8–87.5 [20]
Nicotiana tabacumPlant codon-optimizedmCherry35S-PPDKU6N/A [21]
Oryza sativaRice codon-optimizedCAO1 and LAZY1OsUbiOsU383–92 [22]
Oryza sativaRice codon-optimizedOsPDS, OsMPK2, and OsBADH22_35SOsU6HDR7.1–50 [12]
Oryza sativaPlant codon-optimizedOsBEL2_35SAtU6-262–16 [23]
Oryza sativaRice codon-optimizedSWEET1a, SWEET1b, and SWEET11OsUbi1OsU612.5–100 [24]
Oryza sativaRice codon-optimizedOsCPK6, OsMPK16 and OsCPK7UbiN/A7.69–97.92 [43]
Oryza sativaPlant codon-optimizedOsTubA2UbiOsU612.7 [44]
Oryza sativaPlant codon-optimizedWxUbi-1OsU3N/A [45]
Oryza sativaPlant codon-optimizedOsBADH2UbiOsU6N/A [46]
Sorghum bicolorMonocot codon-optimized syntheticDsRED2Rice Actin 1OsU6N/A [17]
Solanum lycopersicumNicotiana codon-optimizedSHR and SCR35SAtU6N/A [31]
Solanum lycopersicumCodon-optimizedRINUbi4AtU6N/A [32]
Solanum lycopersicumHuman codon-optimizedSlPDS and SlPIF4CaMV 35SAtUBQ and AtU6-2672.7–100 [33]
Triticum aestivumRice codon-optimizedTaMLO2_35STaU626.5–38 [34]
Triticum aestivumPlant codon-optimizedTaMLOA1, TaMLOB1, and TaMLOD1Ub1TaU623–38 [47]
Triticum aestivumRice codon-optimizedTaLOX22_35STaU645 [34]
Zea maysPlant codon-optimizedZmIPK2_35SZmU316.4–19.1 [35]
Zea maysHuman and maize codon-optimizedZmHKT12_35SUbi1AtU6-26, OsU3, and TaU3N/A [14]
Zea maysMaize codon-optimizedPSY1ZmUbi2ZmU60.18–78.83 [36]
Zea maysHuman codon-optimizedZmzb72_35SZmU319–31 [37]
Zea maysMaize codon-optimizedLIG, MS26, and MS45UbiZmU6HDR0.13–3.9 [38,48]
Zea maysPlant codon-optimizedSHRUNKEN2, GBSS (WX)CaMV 35S Ubi and U6-2N/A [49]
Zea maysPlant codon-optimizedZmPLA1CaMV 35S Ubi and U6-287.06 [39]
Zea maysPlant codon-optimizedZmBADH2UbiZmU6N/A [50]
Zea maysPlant codon-optimizedZmFCP1 and ZmCLE7ZmUbiU6N/A [51]
Brassica oleraceaStreptococcus pyogenesBolC.GA4.a35SU6-2610 [40]
Cucumis sativusPlant codon-optimizedeIF4E35SAtU6N/A [52]
Cucumis sativusPlant codon-optimizedGmPDS11 and GmPDS18ZmUbiAtU6 and GmU611.7–48.1 [25]
Gossypium hirsutumPlant codon-optimizedGhCLAUbiGhU6-71–94.12 [53]
Gossypium hirsutumPlant codon-optimizedGhCLA and GhPEBPUbiGhU6-726.67–57.78 [54]
Gossypium hirsutumPlant codon-optimizedDsRed2 and GhCLA1UbiGhU666.7–100 [55]
Gossypium hirsutumPlant codon-optimizedGhCLAUbiGhU6-72.18–17.14 [56]
Gossypium hirsutumPlant codon-optimizedGhFAD2UbiGhU6-769.57 [57]
Gossypium hirsutumPlant codon-optimizedGhCLA and GhPGFUbiGhU6-768.4–89.7 [58]
Gossypium hirsutumPlant codon-optimizedGhCLA and GhPEBPCaMV 35SGhU6-764 [59]
Gossypium hirsutumPlant codon-optimizedGhCLACaMV 35S and UbiGhU6-744.6–97.2 [60]
Glycine maxSoybean codon-optimizedDD20 and DD43GmEF1A2GmU6HDR59–76 [61]
Table 2. CRISPR/Cas induced plant resistance against various diseases.
Table 2. CRISPR/Cas induced plant resistance against various diseases.
Plant SpeciesObjective GeneTransformation MethodCRISPR/Cas9 Induced Resistance against Plant PathogensReferences
Nicotiana benthamianaCP, Rep, and IRAgrobacterium tumefaciens-mediated transformationTomato Yellow Leaf Curl Virus (TYLCV) and Beet Curly Top Virus (BCTV) [90]
Nicotiana benthamianaLIR and Rep/RepAAgrobacterium tumefaciens-mediated transformationBean Yellow Dwarf Virus (BeYDV) [91]
Nicotiana benthamianaGFP1, GFP2, HC-Pro, and CPAgrobacterium tumefaciens-mediated transformationTurnip mosaic virus (TuMV) [92]
Nicotiana benthamiana and Arabidopsis thalianaORF1,2,3, CP and 30UTRAgrobacterium tumefaciens-mediated transformationCucumber mosaic virus (CMV) and Tobacco mosaic virus (TMV) [93]
Nicotiana benthamiana and Arabidopsis thalianaCP, Rep, and IRAgrobacterium tumefaciens-mediated transformationBean Yellow Dwarf Virus (BeYDV) [94]
Arabidopsis thalianaeIF(iso)4EAgrobacterium tumefaciens-mediated transformationTurnip mosaic virus (TuMV) [95]
Arabidopsis thalianaeIF4E1Agrobacterium tumefaciens-mediated transformationClover yellow vein virus (ClYVV) [96]
Solanum tuberosumP3, CI, NIb and CPAgrobacterium tumefaciens-mediated transformationPotato virus Y (PVY) [97]
Solanum tuberosumeIF4EAgrobacterium tumefaciens-mediated transformationPotato virus Y (PVY) [98]
Solanum tuberosumeIF4E1Protoplast transformationPotato virus Y (PVY) [99]
Solanum lycopersicumSlPelo and SlMlo1Agrobacterium tumefaciens-mediated transformationTomato yellow leaf curl virus (TYLCV) [100]
Solanum lycopersicumPMR4Agrobacterium tumefaciens-mediated transformationPowdery mildew (Oidium neolycopersici) [101]
Ipomoea batatasSPCSV-RNase3Agrobacterium tumefaciens-mediated transformationSweet potato chlorotic stunt virus (SPCSV) and sweet potato feathery mottle virus [102]
Hordeum vulgareRep, MP, and LIRAgrobacterium tumefaciens-mediated transformationWheat dwarf virus (WDV) [103]
Solanum lycopersicumJAZ2Agrobacterium tumefaciens-mediated transformationBacterial speck disease (Pseudomonas syringae pv. tomato DC3000) [104]
Solanum lycopersicumSlMlo1Agrobacterium tumefaciens-mediated transformationPowdery mildew (Oidium neolycopersici) [105]
Solanum lycopersicumPLAgrobacterium tumefaciens-mediated transformationFungal disease (Botrytis cinerea) [106]
Solanum lycopersicumACET1a and ACET1bAgrobacterium tumefaciens-mediated transformationFungal disease (Botrytis cinerea) [107]
Solanum lycopersicumSlDMR6Agrobacterium tumefaciens-mediated transformationBroad-spectrum disease resistance [108]
Vitis viniferaWRKY52Agrobacterium tumefaciens-mediated transformationGray mold (Botrytis cinerea) [109]
Vitis viniferaMLO-7PEG-mediated protoplast transformationPowdery mildew (Erysiphe necator) [110]
Vitis viniferaVvMLO3Agrobacterium tumefaciens-mediated transformationPowdery mildew (Erysiphe necator) [111]
Oryza sativaSEC3AProtoplast transformation with Cas9/gRNA expression binaryRice blast disease (Magnaporthe oryzae) [112]
Oryza sativaSWEET13Agrobacterium tumefaciens-mediated transformationBacterial blight (Xanthomonas oryzae p v.oryzae) [113]
Oryza sativaOsSWEET11 and OsSWEET14Agrobacterium tumefaciens-mediated transformationBacterial blight (Xanthomonas oryzae p v.oryzae) [17]
Oryza sativaOSERF922Agrobacterium tumefaciens-mediated transformationRice Blast Magnaporthe oryzae [114]
Oryza sativaeIF4GAgrobacterium tumefaciens-mediated transformationRice tungro spherical virus (RTSV) [115]
Oryza sativaBsr-d1, Pi21 and ERF922Agrobacterium tumefaciens-mediated transformationRice blast and bacterial blight [116]
Oryza sativaSWEET11, SWEET13, and SWEET14Agrobacterium tumefaciens-mediated transformationBacterial blight Xanthomonas oryzae pv. Oryzae [117]
Oryza sativaXa13promoterAgrobacterium tumefaciens-mediated transformationBacterial blight Xanthomonas oryzae pv. Oryzae [118]
Triticum aestivumTaMlo1Agrobacterium tumefaciens-mediated transformationPowdery mildew (Blumeria graminis f. sp. Tritici) [47]
Triticum aestivumTaEDR1Agrobacterium tumefaciens-mediated transformationPowdery mildew (Blumeria graminis f. sp. Tritici) [79]
Citrus sinensisLOB1Agrobacterium tumefaciens-mediated transformationCitrus canker (Xanthomonas citri subspecies citric) [119]
Citrus sinensisPhytoene desaturase (CsPDS CsLOB1) promoterAgrobacterium tumefaciens-mediated transformation(Carotenoid biosynthesis) Citrus canker resistance [120]
Citrus sinensisCsWRKY22Agrobacterium tumefaciens-mediated transformationCitrus canker Xanthomonas citri subsp. Citri [121]
Malus domesticaDIPM-1DIPM 2DIPM-4PEG-mediated protoplast transformationFire blight (Erwinia amylovora) [110]
Table 3. CRISPR/Cas induced resistance against abiotic stress.
Table 3. CRISPR/Cas induced resistance against abiotic stress.
Plant SpeciesObjective GeneTransformation MethodsCRISPR/Cas9 Induced Resistance in Plant against Herbicide and Abiotic StressReferences
Solanum lycopersicumSlMAPK3Agrobacterium tumefaciens-mediated transformationDrought resistance [154]
Solanum lycopersicumSlARF4Agrobacterium tumefaciens-mediated transformationSalinity and Osmotic tolerance [155]
Solanum lycopersicumSlHyPRP1Agrobacterium tumefaciens-mediated transformationsalt stress-tolerant [156]
Solanum lycopersicumSlAGAMOUS-LIKE 6Agrobacterium tumefaciens-mediated transformationHeat resistance [163]
Zea maysALS2Agrobacterium tumefaciens-mediated transformationHerbicide resistance [184]
Zea maysZmALS1, ZmALS2PEG-mediated protoplast transformationHerbicide resistance [80]
Zea maysARGOS8Agrobacterium tumefaciens-mediated transformationImprove yield under drought resistance [171]
Arabidopsis thalianaOST2Agrobacterium tumefaciens-mediated transformationReduced transpiration, stomatal closure, and abiotic stress [150]
Arabidopsis thalianaUGT79-B2, and B3Agrobacterium tumefaciens-mediated transformationOxidative stress, salt and cold tolerance [159]
Arabidopsis thalianaAVP1PEG-mediated transformationDrought tolerance [77]
Oryza sativaOsEPSPSParticle bombardment transformationglyphosate resistance [185]
Oryza sativaALSAgrobacterium tumefaciens-mediated transformationHerbicide tolerant [186]
Oryza sativaALS-FTIP1eAgrobacterium tumefaciens-mediated transformationImazamox herbicide resistance [65]
Oryza sativaOsSAPK2Agrobacterium tumefaciens-mediated transformationDrought tolerance [153]
Oryza sativaOsAnn3Agrobacterium tumefaciens-mediated transformationCold resistance [145]
Oryza sativaOsRR22Agrobacterium tumefaciens-mediated transformationSalinity tolerance [182]
Oryza sativaOsDSTAgrobacterium tumefaciens-mediated transformationDrought and salt tolerance [162]
Oryza sativaOsbHLH024Agrobacterium tumefaciens-mediated transformationSalt stress resistance [157]
Oryza sativaOsGTγ-2Agrobacterium tumefaciens-mediated transformationSalt stress resistance [158]
Oryza sativaOsmiR535Agrobacterium tumefaciens-mediated transformationDrought and salinity stress tolerance [183]
Oryza sativaPPO1 and HPPDPEG-mediated protoplast transformationHerbicide resistance [187]
Oryza sativaOsACCAgrobacterium tumefaciens-mediated transformationHerbicide resistance [188]
Oryza sativaOsTubA2Agrobacterium tumefaciens-mediated transformationDinitroaniline herbicide resistance [44]
Oryza sativaOsMYB30Agrobacterium tumefaciens-mediated transformationCold tolerance [165]
Oryza sativaOsHAK1Agrobacterium tumefaciens-mediated transformationHeavy metal pollution resistance [175]
Oryza sativaOsNRAMP5Agrobacterium tumefaciens-mediated transformationHeavy metal pollution resistance [176]
Oryza sativaOsNRAMP1Agrobacterium tumefaciens-mediated transformationHeavy metal pollution resistance [177,178]
Triticum aestivumTaALS-P174particle bombardment transformationHerbicide Resistance [189]
Triticum aestivumTaHAG1Agrobacterium tumefaciens-mediated transformationSalt tolerance [160]
Hordeum vulgareITPKAgrobacterium tumefaciens-mediated transformationSalt stress resistance [161]
Solanum lycopersicumSlNPR1Agrobacterium tumefaciens-mediated transformationDrought tolerance [166]
Brassica napusTwo BnaMAX1 homologsAgrobacterium tumefaciens-mediated transformationIncreases yield [174]
Brassica napusALSAgrobacterium tumefaciens-mediated transformationHerbicide resistance [190]
Glycine maxGmPRR37Agrobacterium tumefaciens-mediated transformationRegional adaptation [172]
Citrullus lanatusALSAgrobacterium tumefaciens-mediated transformationBensulfuron herbicide resistance [191]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, Y.; Zafar, N.; Ali, Q.; Manghwar, H.; Wang, G.; Yu, L.; Ding, X.; Ding, F.; Hong, N.; Wang, G.; et al. CRISPR/Cas Genome Editing Technologies for Plant Improvement against Biotic and Abiotic Stresses: Advances, Limitations, and Future Perspectives. Cells 2022, 11, 3928. https://doi.org/10.3390/cells11233928

AMA Style

Wang Y, Zafar N, Ali Q, Manghwar H, Wang G, Yu L, Ding X, Ding F, Hong N, Wang G, et al. CRISPR/Cas Genome Editing Technologies for Plant Improvement against Biotic and Abiotic Stresses: Advances, Limitations, and Future Perspectives. Cells. 2022; 11(23):3928. https://doi.org/10.3390/cells11233928

Chicago/Turabian Style

Wang, Yaxin, Naeem Zafar, Qurban Ali, Hakim Manghwar, Guanying Wang, Lu Yu, Xiao Ding, Fang Ding, Ni Hong, Guoping Wang, and et al. 2022. "CRISPR/Cas Genome Editing Technologies for Plant Improvement against Biotic and Abiotic Stresses: Advances, Limitations, and Future Perspectives" Cells 11, no. 23: 3928. https://doi.org/10.3390/cells11233928

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop