Next Article in Journal
Quantitative Trait Locus Mapping of Seed Vigor in Soybean under −20 °C Storage and Accelerated Aging Conditions via RAD Sequencing
Previous Article in Journal
Strategy to Develop and Evaluate a Multiplex RT-ddPCR in Response to SARS-CoV-2 Genomic Evolution
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Critical Review: Recent Advancements in the Use of CRISPR/Cas9 Technology to Enhance Crops and Alleviate Global Food Crises

1
Key Laboratory of Crops Physiology, Ecology and Genetic Breeding, Ministry of Education/College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
2
Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
3
Research Center on Ecological Sciences, Jiangxi Agricultural University, Nanchang 330045, China
4
Department of Agronomy, University of Agriculture, Faisalabad 38040, Pakistan
5
Biology Department, (Genetics and Molecular Biology Central Laboratory), Aljumum University College, Umm Al-Qura University, Makkah 24382, Saudi Arabia
6
University College of Dera Murad Jamali, Nasirabad 80700, Balochistan, Pakistan
7
College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
8
College of Humanity and Public Administration, Jiangxi Agricultural University, Nanchang 330045, China
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2021, 43(3), 1950-1976; https://doi.org/10.3390/cimb43030135
Submission received: 13 October 2021 / Revised: 4 November 2021 / Accepted: 5 November 2021 / Published: 11 November 2021
(This article belongs to the Section Molecular Plant Sciences)

Abstract

:
Genome editing (GE) has revolutionized the biological sciences by creating a novel approach for manipulating the genomes of living organisms. Many tools have been developed in recent years to enable the editing of complex genomes. Therefore, a reliable and rapid approach for increasing yield and tolerance to various environmental stresses is necessary to sustain agricultural crop production for global food security. This critical review elaborates the GE tools used for crop improvement. These tools include mega-nucleases (MNs), such as zinc-finger nucleases (ZFNs), and transcriptional activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR). Specifically, this review addresses the latest advancements in the role of CRISPR/Cas9 for genome manipulation for major crop improvement, including yield and quality development of biotic stress- and abiotic stress-tolerant crops. Implementation of this technique will lead to the production of non-transgene crops with preferred characteristics that can result in enhanced yield capacity under various environmental stresses. The CRISPR/Cas9 technique can be combined with current and potential breeding methods (e.g., speed breeding and omics-assisted breeding) to enhance agricultural productivity to ensure food security. We have also discussed the challenges and limitations of CRISPR/Cas9. This information will be useful to plant breeders and researchers in the thorough investigation of the use of CRISPR/Cas9 to boost crops by targeting the gene of interest.

1. Introduction

The global population continues to grow at an alarming rate [1], but there is an arithmetic increase in the number of food materials available [2]. The global population is expected to increase to 10 billion by 2050 [3,4]. The global population and the negative impact of climatic conditions may eventually create issues regarding food security. The condition may be further aggravated by a decline in the productive areas and reduced yield. Recent estimates from the International Rice Research Institute (IRRI) have indicated that every 7.7 s, one hectare of the fertile area is lost, and the effect may be more pronounced if the global temperature acceleration rate persists [5]. To maintain food security, crop yield capacity must nearly double, and highly tolerant cultivars against various stresses must be developed to achieve this goal [6].
An increase in agricultural crop production using the latest breeding techniques [7] is the primary concern regarding global food security [8]. However, conventional breeding techniques cannot ensure food safety and maximum food production [9,10,11,12]. Varietal improvement and crop development have been accomplished using traditional breeding techniques, such as hybridization, which to some extent, has enhanced the production of crops [13,14,15]. Genetic engineering has been the focus of research for several years in determining the function of genes. This involves using physical and biological mutagenesis and identification of biological contrivances to boost crop production. Owing to several issues, a 100% success ratio cannot be achieved [16]. In recent years, the actual yield has been recorded for the major cereal crops, such as rice in East Asia and wheat in Northwest Europe [17]. Additionally, aspects of the introduction of transgenes into the plant host genome raise public concerns regarding edible crops and are not always appreciated, owing to a lack of clarity on the methods used and consequent benefits. Therefore, the use of biotechnological tools for crop enhancement is of ultimate significance in overcoming these limitations [17].
Genetic engineering includes several methods, such as zinc-finger nucleases (ZFNs), transcriptional activator-like effector nucleases (TALENs), and CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats) [18]. ZFNs are defined as DNA cleavage proteins designed to split DNA structures at a given location. Second, TALENs generate breaks in double strands of targeted structures, activating the DNA response pathway that leads to genome breakage [19]. Although ZFNs and TALENs are commonly used for genome alterations in plants and animal cells (since 2002 and 2011, respectively), a few limitations exist that impede their successful use. The specificity of ZFNs is small and often results in off-target changes [20]. Construction of vectors is time and laborconsuming for ZFNs and TALENs [21]. Therefore, continuous progress in crop breeding programs would be vital in addressing these tasks and for developing food crops [22]. Recent developments in CRISPR/Cas9 tools have rendered directed and accurate genetic alteration of crops possible, increasing the speed of transition to crop improvement [23]. Only a few species have been studied thus far using this approach [24]. Since 2013, the focus shifted to the use of CRISPR/Cas9 and its variants. CRISPR is a DNA fragment that includes non-contiguous, small DNA repeats interspaced by variable sequence fragments or spacers. Therefore, this finding indicated that CRISPR/Cas9 could be responsible for the elicitation of adaptive immunity in prokaryotes [25,26] ISPR/Cas9 has the potential to cure diseases [27], and the use of this technique has a huge impact on plant biology [28]. The first reported application of CRISPR/Cas9 is an adaptive immune mechanism described in an experiment in 2007, wherein investigators considered the phenotype to be phage-resistant, and the addition or removal of specific spacers could alter the bacterial phenotype [29]. In the future, it will be easy to genetically alter plants for crops improvement [30]. The use of Cas9 (a protein known as biological scissors) sets new standards and revolutionizes genome editing, which will lead to new insights into agricultural crops improvement [31]. Here, we briefly reviewed the role of CRISPR/Cas9 applications in crop improvement, its limitations, and future prospects. CRISPR/Cas9 is currently being used for different aspects in plant breeding like, diseases resistance, drought resistance, salinity tolerance and genetic improvement of crops for various aspects. The grain yield and quality of crops is also improved by Cas9. The production of biomass has also been improved by using Cas9. Likewise, nutrients use efficiency and enhancement of morpho-physiological traits has been done using Cas9. Role of genome editing is shown in Figure 1.

2. Genome Editing Tools

Genome editing aims to revolutionize plant breeding and could help safeguard the global food supply chain [32]. Meganucleases are first-generation gene-editing tools and [33] were the first tools used for genome editing and have been classified by the presence of an approximately 12–40 bp recognition site. They are often known as the most defined restriction enzymes because of their precise nature and broad recognition site [34]; hence, they are also called homing endonuclease enzymes. Research studies have shown that the repair process of double-stranded breaks is caused by non-homologous end joining (NHEJ), responsible for the knockout of genes present in Arabidopsis and tobacco [35,36]. As DNA binding domains are attached to a catalytic area of meganucleases, separation is not possible, and therefore it is practically impossible to alter the meganucleases with other genome editing techniques [37]. There are several examples of studies in which meganucleases (MNs)were exploited in cotton [38] and maize [39] using alerted MNs; however, studies are warranted to enhance the methodology.
The second gene-editing tool includes ZFNs, which are more efficient and reliable owing to the generation of double-stranded breaks (DSBs) [40]. ZFNs were used as the first genome manipulation approach developed by the application of engineered nucleases. Detection of the Cys2-His2 Zinc-finger domain made this approach possible [19]. The structure of ZFNs consists of the following two main domains: (1) the DNA-binding domain containing 300 to 600 zinc-finger repeats [41]. Individually zinc-finger repeats can be observed between 9–18 bp; and (2) the DNA slice domain, considered a nonspecific domain of type 2 restriction endonucleases [42]. ZFNs comprise two monomers accredited to their particular object sequences reversely adjoining 5- and 6-bp DNA targets [43]. DNA is sliced by dimer enzymes comprising the FokI domain. The specific order of 24 to 30 bp is read by a zinc-finger domain that contains particular or occasional directing positions in the genome [44]. The genome-editing field has shown success and advancements in acquiring the capacity to control and alter the basic genomic marks.
TALENs have been widely used for many years. TALENs were developed by consolidating the FokI slice domain to the DNA-binding domains of the TALE proteins. These TALEs contain complex duplications of 34 amino acids for the effective alteration of a single base pair [45]. TALENs also cause modifications by creating DSBs that trigger initiation of pathways where editing occurs [19]. The central domain and the nuclear localization sequence are present in the TALEN system [46]. The proficiency of proteins to alter DNA was studied for the first time in 2007. Nevertheless, the DNA-binding domain comprises 34 amino acid repeat sequences, and a single nucleotide is detected in the DNA target region by each repeat, whereas individual constant sequences of ZFNs are observed as three nucleotides in the targeted DNA [47]. Few studies have been conducted using TALENs and ZFNs in plants; although these studies favor the application of TALENs, their gene-editing effectiveness is usually low. TALENs are more effective and reliable for genome editing programs [44]. A list of different crop traits improved by the usage of MNs, ZFNs, and TALENs is shown in Table 1.

2.1. CRISPR/Cas9 and Its Brief Overview

CRISPR/Cas9 genome editing technique is definedas clustered regularly interspaced short palindromic repeats, short, repeating variants of genetic material and found in most archaea as well as in many bacterial species. CRISPR/Cas9 and its related proteins develop a very strong protective system that safeguards plants against foreign agents like viruses, bacteria, and other elements. CRISPR/Cas9 is often used to mutate the targeted genes within the system [56] ISPR/Cas9 was discovered in DNA cleavage systems programmed by RNA, which has been found in bacteria and archaea. CRISPR/Cas9 is the most commonly used and well-studied CRISPR/Cas9 system [57,58]. Current recognition of the researchers for the development of a genome editing method using CRISPR/Cas9 by the Nobel Prize committee is an additional step nearer to developing and cultivating novel varieties of crops [59]. The system comprises two constituents, namely an endonuclease DNA (Cas9) and a target-specific RNA, called the single guide RNA (sgRNA) [57,58]. The basic steps to use Cas9 involves the existence of a protospacer adjacent motif (PAM) sequence near and directly pointed to a given target. Various spacer sequences are necessary for the application of Cas9 as a target; therefore, CRISPR/Cas9 is fast, effective, cost-effective, and adaptable. Parts of CRISPR/Cas9, either in DNA or RNA, are administered to plant cells for the CRISPR/Cas9-mediated manipulation of genomes to cut plant DNA in a predetermined series. In this manner, plant cells initiate processes to “patch” the break to maintain genome integrity, and this hints at the generation of numerous forms of alterations in the directed sequence. When the break is fixed via NHEJ/homology-directed repair (HDR), small deletions or insertions occur that can lead to the mutation of genes.
Otherwise, the accessibility of a homologous DNA template present around the targeted point can cause an HDR, which results in the insertion of a DNA template, thereby allowing accurate gene replacement or insertion [60]. Base manipulation is the newest addition to the features of this tool [61,62]. Until now, changes in A–G or C–T have been attained through the application of base manipulators [61,62], which has sparked significant interest in food crop improvement-related gene editors. The CRISPR/Cas9 technology can also be used with the dead Cas9/Cas12 a for gene control, epigenetic alteration, and chromosome imaging [23]. A comparison of the functions of different GE tools is described in Table 2.
Throughout the characterization of spacer absorption, motifs related to spacer precursors (protospacers), an invading bacteriophage, were isolated from DNA [25,63]. Such motifs are small extensions of trinucleotides, situated directly after the protospacers, which have a significant function in recognizing particular protospacers and in guiding the location of the spacers inserted in the protospacer repeat arrays [63]. Brouns, et al. [64] showed that a CRISPR RNA precursor transcribed from a CRISPR site was split into mature RNA molecules inside the repeated chain of proteins of Cas (to crRNA). Every crRNA comprises a spacer flanking short duplications of DNA and acts as a small RNA guide that helps proteins elicit an antiviral action [64]. CRISPR/Cas9 in Streptococcus thermophiles have been shown to slice plasmid DNA [65]. Such results support the molecular origin of adaptive immunity facilitated by the CRISPR/Cas9 system. Each of the genome-editing techniqueshas its advantages and drawbacks, as shown in Figure 2.
Table 2. Differences in functions of genome editing tools.
Table 2. Differences in functions of genome editing tools.
RoleZFNsTALENsMNsCRISPR/Cas9References
Efficacy of target recognitionHigherHigherHigherHigher[66]
Kind of ActionDouble-stranded break in target DNADouble-strandedbreak in target DNADirect conversions in targeted regionsDouble-stranded break in target DNA[67]
MutagenesisHigherMiddleMiddleLower[66]
MultiplexingDifficultDifficultDifficultPossible[68,69]
Target rangeUnlimitedUnlimitedUnlimitedLimited by PAM[70]
EffectsLowerLowerLowerLower[71]
CostHigherHigherHigherLow[72]
Crop ImprovementLowLowLowHigher[72]
RangeNarrowNarrowNarrowBroad[72]
Dimerization RequiredNot requiredNot requiredNot required[71]
TypesOneOneOneMany[71]
Future useMediumMediumMediumHigh[72]

2.2. CRISPR/Cas9 Mechanism and Function

Makarova, et al. [73] suggested that the immunity process comprises three phases facilitated by the CRISPR/Cas9 system, including adaptation, expression, and interference. In the adaptation phase, short plasmids are inserted as new spacers into the CRISPR/Cas9 series, whereas in the expression phase, crRNA transcription and maturation processes occur. Ultimately, the crRNAs direct Cas proteins in the interference process to match the splitters. Makarova et al. [73] suggested the hierarchical grouping of CRISPR/Cas into three main groups, namely I, II, and III, as described in a study on the origin of Cas proteins and CRISPR/Cas9. Type I and III systems involve the application of a complex of multiple Cas proteins as sign proteins, whereas Cas9 is a large multifunctional single sign protein in the Type II system and is accountable for both the development of crRNA and slicing of the target DNA [73]. The Type II system has a relatively easy architecture compared to that of the other systems and can be developed more effectively to act as a genome editing method. Mechanisms of genome editing using CRISPR/Cas9 involves, sgRNA and Cas9 make a complex, sgRNA unwinds DNA, and Cas9 cut the DNA, genome analysis, cloning of sgRNA, transformation and selection of plants, regeneration, extraction of genomic DNA and final step is analysis of sequence to confirm the results Figure 3.
Regarding the Type II system, a small trans-noncoding RNA (transactivating CRISPR/Cas9 RNA) was discovered in S. pyogenes, and it has been shown to direct the development of crRNAs through the pairing of bases with duplicated regions of transcripts of pre-crRNA via the action of RNase III and Cas9 [74]. Jinek et al. [57] found suggested application of a Type II system and synthesized a sgRNA via the fusion of crRNA and tracrRNA. The 5′ end 20-bp sgRNA sequence, a target binding DNA sequence, may be converted into any sequence [57]. The analysis described above indicated that three pairs of PAM bases [57,65] could be programmed with a sgRNA to produce DSB at a point from the nuclease of Cas9 from Sogenes (SpCas9). The sequencing and location of PAMs vary across various CRISPR/Cas systems [63]. A canonical PAM related to SpCas9 is 50-NGG-30 (N exemplifies one of the four nucleotides), and a 20bp target DNA sequence was accompanied by a PAM [57,63]. Cas9 nuclease comprises two domains identical to HNH and RuvC [73]. Jinek et al. [57] also found that the HNH and RuvC-like domains could complement the DNA strand and not the RNA reference, respectively. Therefore, this system is effective and programmable and acts similar to a model CRISPR/Cas9 system, which is most frequently used to alter genes, with the alterations facilitated by NHEJ/HDR. Single-guided RNA bioinformatics tools developed for CRISPR/Cas9 are listed in Table 3. The role of different RNAs in CRISPR/Cas9 is varied and highly studied. The guided RNA (mgRNA) is a particular RNA sequence thatidentifies the targeted regions of the DNA region and regulates the Cas9 nuclease enzyme for gene editing. Hence, it unwinds the DNA and aligns the gene with Cas9 protein, and causesdouble-stranded breaks. The second type of RNA is crRNA which describes the targeted DNA for Cas9protein, whereasthe role of tracrRNA is to work asa scaffold linking the crRNA molecule with Cas9 protein and assist insorting out of mature crRNAs from the pre-crRNAs developed from CRISPR/Cas9 arrays Table 3 [75].

3. Application of CRISPR/Cas9 for Crop Improvement

CRISPR/Cas9 technique has become the most widespreadand has many advantages, like time effectiveness, low cost of editing, extraordinary adaptability, and the ability for directing multiple genes reproductions instantaneously [86]. This gives extraordinary worth for breeding of numerous polyploid species of crops species, which are problematic to progress using classical approaches, and lets the group of a range of monoallelic as well as biallelic mutants, and a resulting allelic sequence of phenotypes, in the first generation, something that is characteristically not possible using classical breeding approaches. This technique is a more efficient and versatile technique of genome editing [87]. This technique, i.e., CRISPR/Cas9, is simple, efficient, and cost-effective relative to TALENS, ZFNs, and MNs, and it is used to edit multiple genomes at the same time [88,89]. Owing to its positive features, the CRISPR/Cas9 system has been used for several plant species [90,91], and its application provides the best solution to multiple issues encountered in plant breeding [92]. The CRISPR/Cas9 has been used to improve the monocots and dicots crops for a variety of traits like yield, quality, disease resistance, and resistance to climatic variations [93]. The CRISPR/Cas9 technique has been used to edit the genomes of cereal crops, such as wheat, maize, rice, cotton, and vegetables like tomatoes, and potatoes and fruits like banana and apple [94,95]. The most common use is the knockout of target genes that have been gained by induction of indels that result in a frameshift mutation. A large number of important traits have been introduced in crops using Cas9 and have been discussed in many reviews [96,97,98,99].

3.1. Use of CRISPR/Cas9 for Improvement of Yield and Quality

CRISPR/Cas9 creates revolutionary changes in crop species [100]. Several laboratories in the world have been established that use this powerful technique because of its potential. Here, we listed some of its uses that have been considered in the improvement of the yield and quality of crops. CRISPR/Cas9 is used to develop cultivars with high nutritional values and resistance characteristics and to create the largest milestone of genome editing in modern technology. A commercial oil was obtained from the crop seedsof Camelina sativa, which is widely cultivated because of its higher percentage of fatty acids. Moreover, the JAZI, a jasmonate-zim-domain protein, BRII brassinosteroid insensitive 1, and GA-insensitive genes were also altered, and CRISPR/Cas9 resulted in a 26% to 84% mutation frequency [17]. In the same species, a squamosal promotor binding protein and a flowering locus were manipulated using Cas9, and at the late flowering stage, the plants showed approximately a 90% mutation frequency [101]. A multiple CRISPR/Cas9 system combined with other tools was engineered and applied for the manipulation of six altered PYL genes of ABO receptors with a 13%–93% mutation frequency in the first generation [102]. The green fluorescent gene present in Nicotiana benthamiana with RNA-facilitated endonucleases was altered [90]. Later, this was delivered through the tobacco rattle virus to modulate the instructions to the plant genes to manufacture and manipulate transcriptional features [103].
The progenies of homozygous rice species were altered using CRISPR/Cas9, and the results showed a deletion in the gene cluster on the chromosome with heritable variations in genetic makeup [104]. Mutagenesis in three individuals in the aldehyde oxidase gene family of rice, OsAox1b, OsAox1a, and OsAox1c, in addition to the (abbreviation) OsBEL gene mediated by CRISPR/cas9, was observed. Subsequently, inherent alterations of genes in successive progenies were studied [105]. An ABA-inducible protein encoded by the barley gene HvPM19 was responsible for the upregulation of genes responsible for grain dormancy. As a result of mutations induced by CRISPR/Cas9 in HvPM19, a 10% mutation frequency was generated [106]. The role of two QTLs, Gn1a and GS3, for rice grain number and size, were also investigated by Cas9 [107]. Usman, et al. [108] studied the editing of OsSPL16 gene in rice by Cas9 and found an improvement in rice grain yield. CLBG1 mutagenesis by Cas9 in watermelon significantly reduced seed size and enhanced seed germination [109]. Cas9 can be used to improve every trait in crops, and this could lead to a green revolution in time. The base editing of the WAXY allele of granule bound starch synthase improved the cooking quality of rice using Cas9 [110]. Zhang, et al. [111] studied the disruption of MIR396f and MIR396e,which improves the yield of rice under low nitrogen conditions. The grain yield modulator miR156 regulates the seed dormancy in rice as studied in an experiment [112]. In a recent study, new rice mutants with high yield and aroma were generated by editing of three homologs (GL3.2, Os03g0568400, and Os03g0603100) of P450 and OsBADH2 using CRISPR/Cas9 in rice [113]. A high content of protein often reduces the rice cooking quality. By targeted mutagenesis of transporter genes of amino acids could bring significant variation among the mutants. The OsAAP6 and OsAAP10 are two mutants generated in rice high-yielding cultivars by Cas9, and these mutants showed significant improvement in the yield and cooking quality of rice [114]. From the above findings, we have reached a conclusion that further mutagenesis of amino acid genes could lead to the production of more high-yielding and good quality cultivars in rice. Several rice mutants with high-yielding attributes were generated by editing of GS3 gene by using Cas9. This study showed that complex traits of rice could be improved using CRISPR/Cas9 in rice [115]. Targeted mutagenesis of TaSBEIIa using CRISPR/Cas9 successfully produced high amylose wheat with a meaningfully improved resistant starch content [116]. Alteration of the composition of starch, its structure, and properties via editing of TaSBEIIa was studied in spring wheat using CRISPR/Cas9 [116].
The CRISPR/Cas9 technique was used for high-quality oil production, such as oleic acid from Brassica napus [117]. CRISPR is used in Camelina sativa by changing the function of the ALCATRAZ INDEHSCENT and JAGGED genes to increase pod shattering resistance [118,119]. In another study [120], the function of INDEHSCENT and ALCATRAZ genes were affected, which are essential to the dehiscence of fruits in Brassica species. Likewise, Zaman, et al. [121] studied the genome editing of JAGGED using Cas9 in Brassica napus and revealed that this gene was involved in the development of pod shattering resistance. Similarly, the role of ALCATRAZ in pod shattering resistance in Brassica napus was studied [86]. In vegetables, CRISPR/Cas9 was mainly used in tomatoes because of their economic significance and easy transformation capability. Targeted changes of the engineered gene SP5G in tomatoes resulted in early flowering and several brushes, which indicated an early harvest [122]. Liang et al. [54] reported the selective knockout of genes, including ZmIPK1A, ZmIPK, and ZmMRP4, which are involved in the synthesis of phytic acids in maize. The CRISPR/Cas9-based impediment of the SISG07 gene was studied, and the phenotype of the needle structure in tomatoes was observed [123]. The CRISPR/Cas9 also used to edit the apple protoplast for better taste and quality [124]. CRISPR/Cas9 was used to produce steroidal glycoalkaloids in certain varieties of potatoes to affect St16Dox. The study led to the generation of two lines of potatoes free of SGA with a deletion in the St16Dox gene [125]. Likewise, the GBSS gene that is responsible for the synthesis of starch was mutated using Cas9 in potatoes. An increase in amylopectin levels was observed in mutant lines [126]. The SnLazy1 locus, known as the Lazy1 ortholog in tomatoes, was manipulated by CRISPR/Cas9 with effective heritability of the snlazy-1 allele, and mutants showed a downward pattern of stem development [127]. Many techniques have been developed to improve their yield and quality regarding staple and fruit plants, such as citrus, grapes, and tomatoes, but CRISPR/Cas9 is the most useful and latest method. Earlier studies showed that Xanthomonas citri, which advanced the agro infusion method to deliver CRISPR/Cas9, affects the CsPDS genetic factors in leaves of sweet oranges [128]. Because of the greater efficiency of genome manipulation, the consumer can now eat edited fruits because this technique does not permit the entry of foreign genes into the genome. Previously, it was reported that the Cas9 technique could be used to edit genes in apple protoplasts, which has been applied in some other crops [124]. A ground cherry of a wild variety of tomatoes yielded higher and larger fruits [129]. Additionally, in apples, a mutation of PPO was reflected as transgene-free using Cas9, which is appropriate for human consumption [126]. The production of seedless fruit is always the primary goal of any breeding plan. In tomatoes, the targeting of SIAGL6 and SIIAA9 was studied, and parthenocarpy was achieved using Cas9 [127]. The MaGA20ox2 gene was knocked out in bananas, and the resulting mutation produced dwarf banana genotypes [130]. There are many uses of CRISPR/Cas9 for quality and yield enhancement, which are shown in Table 4.

3.2. Development of Disease Resistant Varieties Using CRISPR/Cas9

Plant diseases severely affect crop yields and quality [149];the primary causal agents of diseases are viruses, fungi, nematodes, insects, and bacteria, which cause severe reduction in crop yield. CRISPR/Cas9 is ow widely used for the development of disease-resistant crops [150]. The appearance of deadly outbreaks of these insects and other biotic stresses has become a major issue [151]. Understanding plant and pathogen relationships or interactions are of considerable importance in the defense of plants from these attacks [152]. In the wheat crop, there is an increasing demand for coeliac-safe wheat-based products. Coeliac safe wheat leads to reducing the risk of chronic disorders. These traits cannot be improved by traditional breeding methods. Recently, CRISPR/Cas9 has been used to edit the glutenins genes and produce coeliac safe cultivars. These techniques generate offspring with silenced and deleted gliadins, which may decrease the patients exposure to coeliac disorders (CD) epitopes [153]. Likewise, in another study, Verma, et al. [154] reviewed the role of CRISPR/Cas9 in developing strict-gluten free wheat, which can reduce the risk of coeliac disease (CD). They have concluded that CRISPR/Cas9 has been successfully edited the wheat genome. In the future, this technique would be more useful to develop coeliac-safe wheat rice, the gene Elf4g was mutated to increase resistance to thetungro spherical virus in rice [155]. In rice, bacterial leaf blight is a severe problem that causes huge losses in yield. This gene OsSWEET14 was targeted by Cas9 to induce resistance to the pathogen [156]. This shows that targeted mutagenesis of any susceptible gene could bring significant variation in plant resistance to any pathogen. For instance, the knockout of the OsERF922 gene was conducted by using Cas9, resulting in increased resistance against blast produced by Magnaporthe oryzae [157]. Similarly, the targeted mutation of SWEET1E has been performed using Cas9 to produce plants resistant to blight [158]. Therefore, genome editing has been successfully applied to investigate the plant and pathogen relationship, and CRISPR/Cas9 is a potent tool to develop disease-resistant cultivars by mutating the disease-causing gene. For instance, Cas9 was applied to create mutations in the promoter region of the gene CsLOBI, which causes citrus canker, and consequently, mutations would result in enhanced resistance to citrus canker. Two mutant lines, DLoB10 and DLoB9, exhibited higher frequencies of mutations. Frameshift mutations and changes in the function of CsLOBI resulted in increased resistance to Xanthomonas citri [159]. To increase the resistance of citrus against Xanthomonas, editing of the effector binding element using Cas9 in the promoter area of the CsLOBI gene was performed [160]. The Cas9 technique has been applied to edit the gene TaMLO [104] in wheat protoplasts, resulting in the development of enhanced powdery mildew resistance [50]. Similarly, powdery mildew resistance in wheat was developed by targeting EDRI homologs using Cas9 [161]. Likewise, MLO gene variants were generated in tomatoes, which increased powdery mildew resistance [162]. It was predicted that half of the plant diseases are caused by viruses, which cause substantial losses in crop yield and quality [163]. DNA virus amplicons considerably enhanced the targeting efficiency of the genome. In hexaploid wheat, replicons of geminivirus were used for Cas9 transient expression to counter dwarf virus, and approximately a 12-fold upregulation was noted in gene expression [164]. The geminivirus genome has been targeted by CRISPR/Cas9 and has been used to prevent viral growth [165,166]. Cas9 can be used to edit the viral DNA rather than cure the diseases caused by them [163]. Cas9-mediated alteration of the viral genome can be enhanced by using virus promoters to control sgRNA cassata expression [166]. Recently, a novel ortholog of Cas9 was found in Francisella novicida, which was used to manipulate the genome of RNA-based viruses. The FnCas9 gene helped to halt the replication process of TMV and the cucumber mosaic virus, conferring resistance against them [167]. The targeted mutagenesis of SIPeLo and SIMIO1 was done for trait introgression of tomato using Cas9 to confer resistance against leaf curl virus and powdery mildew [168]. Targeted mutagenesis of OsPETI in rice by Casp to improve the resistance to rice sheath blight was studied [169]. A novel diseases resistance paralog using Cas9 has been created in soybean. These novel paralogs confer diseases resistance [170]. Tobacco, an important cash crop, is severely affected by potato virus Y, which is due to the susceptibility of gene Ntab0942120. Through the use of CRISPR/Cas9 technique, this gene was knockout and transgene-free homozygous edited plants were generated which showed resistance to PVY [171]. In brassica. In an experiment,5genes were knocked out for resistance to Phytophthora in potatoes [138]. Thus, Cas9 is a powerful tool to enhance the genetic makeup of crops to increase their resistance against viruses and other casual agents. A list of crops developed using Cas9 against different diseases is shown in Table 5.

3.3. Development of Climate-Smart Crops Using CRISPR/Cas9

Various abiotic stresses have been tackled in major crops, such as rice, wheat, maize, cotton, and potatoes using Cas9. The plant breeding system has been modernized by developing climate-smart or abiotic stress-resistant crops. CRISPR/Cas9 has been used to modify any sequence and to bring any character to crops. Molecular breeders discovered many genes related to abiotic stress tolerance and engineered them into crops [180]. CRISPR/Cas9 generated slmapk3 protein gene mutants improved defense response to drought stress in tomatoes [181]. Two genes, TaDREB3 and TaDREB2, linked to abiotic stress resistance in wheat protoplasts, have been studied using Cas9. Using the T7 endonuclease assay, mutant gene expression was observed in approximately 70% of the transfected wheat protoplasts. Thus, all mutant plants showed tolerance to drought compared with wild-type plants [182]. A comprehensive review on improvement of drought stress tolerance in plantsusing Cas9 [183]. In rice, two mitogen-activated genes, OsMKP2 and betaine aldehyde dehydrogenase OsBADH2, were manipulated using the Cas9 approach. These genes were delivered into the host genome by protoplast transformation and the particle bombardment technique, which conferred resistance to various stresses [91]. The OsAnn3 gene of rice was edited against cold stress, and the role of this gene was studied in genome-edited plants [184]. The gene SAPK2 was altered to examine the stress mechanism in rice. A previous study showed that this gene enhanced drought and salinity tolerance in rice [185]. Drought tolerance has been developed by targeting the ARGOS8 gene to create new variants, and this is of remarkable significance in maize [186]. Cas9 induced mutagenesis of Leaf1,2 conferred drought tolerance by affecting the expression pattern of proteinand scavenging of ROS in rice [187]. Zhang et al. [188] enhanced the salinity tolerance in rice via targeted mutagenesis of OsRR22 gene. Chickpea is an important legume crop that is largely affected by drought stress. Previously only one study has been conducted to bring mutation in genes using CRISPR. Two genes 4CL and RVE7 were targeted by Cas9 to increase drought stress tolerance in chickpea. The knockout of these genes using Cas9 is a novel approach that led to the development of drought-tolerant varieties in Chickpea in the future [135]. Recently, de Melo, et al. [189] stated that AREB-1 activated Arabidopsis by CRISPR presented an enhanced drought tolerance than wild-type plants. CRISPR has also been used to introduce herbicide tolerance in crops. Kuang, et al. [190] studied the base-manipulation facilitated artificial evolution of OsALS1 in planta to develop new herbicide-tolerant rice germplasm.
Similarly, the Cas-based mutation of two genetic factors, Drb2a and Drb2b, was studied, and these genes controlled drought and salt tolerance in soybeans [191]. The mitogen-activated protein kinase gene that counteracts drought stress by safeguarding its membrane from oxidative stress and controlling the transcription of genes has been studied to reduce drought stress. The relationship of SIMPAK3 in drought stress has been studied in tomatoes by producing knockout variants in the SIMPAK3 gene for the development of drought tolerance via the Cas9 technique [181]. Many genes govern crop yield and stress tolerance. Several studies have been conducted to identify QTLs that govern significant characteristics in crop development systems. Such QTLs have been pyramided into superior lines; however, this is a complex methodfor QTLs closely linked and can produce deleterious effects if non-target regions are transferred. Cas9 can be applied to produce targeted mutagenesis. Using Cas9-based QTL editing approach, two QTLs, Gn1a and GS3, were investigated in rice varieties [107]. These studies indicate that the CRISPR/Cas9 technique has enormous potential for developing climate-adapted crops. The above studies showed that Cas9 could alter genes to facilitate resistance against many abiotic stresses, e.g., drought, high temperatures, heavy metals, as well as nutrient deficiencies [180]. A list of climate-smart crops is shown in Table 6.

4. Novel Breakthroughs

4.1. Production of Mutant Libraries

The efficient, practical investigation of all genetic factors of the genome of a sequenced crop is a considerable challenge. The construction of mutant libraries is an effective and reliable method [197,198]. CRISPR/Cas9 is a powerful technique for developing mutant libraries, and its targeting capability can be changed by altering the 18–20 bp target binding order in sgRNA. CRISPR/Cas9 application renders forward genetic screening studies possible. Development of mutant libraries using CRISPR/Cas9 was first reported in human cell culture [198], and this set the basis for CRISPR/Cas9 application to advance plant mutant libraries for high-throughput selection. In an experiment, pooled sgRNA was transformed into tomato plants, and mutants were developed [199]. In rice production, mutant libraries were constructed by two research groups, with loss-of-function mutants generated during the transformation of synthesized sgRNA libraries [197,200]. Certain phenotypic alterations, such as sterility and lethality, were observed when cultivated in the field [197,200]. The general use of CRISPR/Cas9 is valuable for constructing mutant libraries to study the genetic mechanism behindcrop improvement.

4.2. Base Editing

CRISPR/Cas 9 is now used to edit a single base, which is accountable for genetic variations in novel traits of crops, as shown by genome-wide association studies [201]. Hence, gene-editing techniques for point mutations are necessary. Without using a DNA repair template, one DNA base can be accurately changed into another DNA base using the CRISPR/Cas9 technique [62]. Cas9 should be fused with an enzyme with conversion capability for base editing. Imidazolinone-resistant rice was produced by base editing, and it is one of the best examples of this technique [202]. In the same manner, Arabidopsis was produced [203] by changing ALS and a cytidine amino acid-base changer. Similarly, multiple bases were changed in rice [204]. In this approach, the base change tool provides a novel direction for genome editing, widening its potential with specific nucleotide alterations at precise genomes.

4.3. Prime Editing

CRISPR has great efficacy to mutate genes but despite its ability to produce accurate base edits outside the four alteration mutations is still the main restraint. Prime editing is another technique of DBS, and it is used to enlarge the area and efficiency of genome editing [205]. This technique hires an engineered reverse transcriptase fused to Cas9 and a prime manipulating guide RNA [205]. This pegRNA varies from sgRNAs as it includes not only the guide order that can identify the objective spots but also a reverse transcriptase template spelling the preferred genetic deviations. Li, et al. [206] newly modified prime editors to bring point alterations, insertions, as well as deletions in rice. By using this method, all 12 kinds of base-to-base replacements, and numerous base replacements, insertions, as well as deletions, were noticed. They stated that the occurrence of prime editing brought by this prime editor was up to 21.8% [206]. Comparable conclusions have been stated by molecular breeders [206,207].

4.4. Transgene Free Editing of the Genome

Traditional techniques of genome alterations require the transfer and combination of DNA cassettes encoding altered parts into the host genome. DNA fragments are usually degenerated but produce harmful effects [208]. This technique increases the issues with the regulation of GM plants. Prolonged expression of editing enzymes and systems increases the number of off-target functions because of the greater number of nucleases in these organisms. DNA-free genome editing was first reported in Arabidopsis in the same manner as in tomato and rice via delivery of CRISPR/Cas9 RNPs into plant protoplasts [209]. However, methods for effective regeneration of protoplasts do not exist for maximum crops, which leads to the application of particle bombardment and thus facilitates DNA-free genome alteration approaches. In wheat embryos, CRISPR/Cas9 RNAs have been transferred by the particle bombardment technique [144].

4.5. Multiplex Genetic Engineering

Using multiple sgRNAs, multiple genomes can be affected simultaneously in any crop. Multiple traits can be incorporated into novel cultivars [210], and multiple individuals from multiple families can be targeted using this technique. This can be attained by combining various sgRNAs into one vector [211]. Clonal reproduction of rice hybrids was previously reported [211]. Hybrid heterozygosity was often fixed via CRISPR/Cas9 genome editing of three genes in three meiotic genes, namely REC8, PAIR1, and OSD1, to generate clonal tetraploid seeds. Hence, multiple GE approaches will provide a more rapid method of creating novel variations in varieties.

5. Utilization of CRISPR/Cas9 for Crop Domestication

Domestication of crops and plant breeding led to the development of crops with a high yield that is adjusted to native growing circumstances. Nevertheless, the rising human population faces a number of agricultural challenges, comprising climate alteration, variations in abiotic and biotic stresses, and damage of arable land, alongside a claim for more maintainable and defined agricultural practices. Relatives of modern cultivated and orphans’ crops are considered as an important source of novel variation. However, their low yield and undesirable look prevent their commercial cultivation [212]. Newly, the idea ofde novosubjugation via gene manipulation has been sightseen as a mechanism to bring the wild and orphan crops under domestication rapidly, and therefore advantage from recalled genetic difference and from the features of domesticated plants [213]. This is mainly promising; meanwhile, many classical domesticated genes are perfect candidates for Cas base editing of genes: they are well categorized, have simple genetic architecture, and are monogenetic in nature [214] milarly, ground cherry has alot of unwanted characteristics, like small fruit and a strong stem, which cause fruit dropping. CRISPR base gene-editing technique was used to mutate the gene SP5G, which caused a large number of fruits [129]. These studies prove that Cas-based gene editing can increase the speed of domestication and increase the worth and use of orphan crops.
CRISPR/Cas9 is now being used to domesticate wild plants to serve human needs. Ancient farmers began domesticating all main crops, including rice, wheat, and maize. Nevertheless, our descendants used only a restricted number of originator species during domestication and simply selected plants with better characteristics, such as high yield and ease of breeding, which resulted in a decline in the natural variability of plants. Recalling that genetic diversity is a main concern in the selection process, domestication of wild crops or plants may preserve this diversity. The CRISPR/Cas9 tool has been used to domesticate wild tomatoes, which are tolerant to stress but present with many defaults in fruit production [213]. Six QTLs that were significant for yield were manipulated in one study, and all lines showed enhanced fruit size and fruit number [213]. Many other crops, such as potatoes, bananas, and quinoa, are important locally, have good nutritional value, and are well adapted to local habitats. Despite these features, their low yield and fruit drop prevent their cultivation at larger scales. CRISPR/Cas9 is a powerful tool for manipulating genes and creating desirable crop features. This technique was recently applied to increase flower production and fruit size in ground cherries [129]. With the discovery of genes governing the domestication process, we are confident that we can engineer the genome and increase global food security. To improve the efficiency of genome editing using CRISPR/Cas9, here are the novel techniques by which we can improve it. The first step is we need to do an efficient screening of our desired traits that need to be edited. The knowledge of genetic information about desired traits is a prerequisite in genetic editing, whether it is polygenic or monogenetic. The selection of an efficient tool is important to get good results for editing. Off-target effects should get noticed, and it can be done by designing those sequences which have a close affinity towards each other. The challenges in the agricultural sector are posing a serious threat to crop production. There are many sources of genetic variation which can be utilized and explored using knockout methods to bring desirable changes to meet the need of agricultural crops. The more the efficacy of CRISPR/Cas9, the more the chances of efficient editing will increase, and the greater the desirable results. Novel strategies to improve genome editing using CRISPR/Cas9 are illustrated in Figure 4.
Domestication of crops and plant breeding led to the development of crops with high yields, which are adjusted to native growing circumstances. Nevertheless, the rising human population faces a number of agricultural encounters, comprising climate alteration, variations in abiotic and biotic stresses, and damage of arable land, alongside a claim for more maintainable and defined agricultural practices. Relatives of modern cultivated and orphans’ crops are considered as an important source of novel variation. However, their low yield and undesirable look prevent their commercial cultivation [212]. Newly, the idea ofde novosubjugation via gene manipulation has been sightseen as a mechanism to bring the wild and orphan crops under domestication rapidly, and therefore advantage from recalled genetic difference and from the features of domesticated plants [213]. This is mainly promising; meanwhile, many classical domesticated genes are perfect candidates for Cas base editing of genes: they are well categorized, have simple genetic architecture, and are monogenetic in nature [214].

6. Challenges and Limitations of CRISPR/Cas9 Application

CRISPR/Cas9 has several applications in plant breeding, but there are certain limitations. The main challenge is the availability of a small gene pool of important traits; hence, gene availability is important for this tool. Therefore, it is crucial to decode genomic sequence evidence and to investigate valuable genetic resources to improve major crops. It is important to remember that other challenges associated with this technique include the lack of efficient transformation techniques and plant regeneration from cultures that are complex and time-consuming processes. Biosafety issues hinder its application in crop development. Owing to improvements in identification methods, plant mutants with off-target properties can be recognized and detached by separation during successive crosses. In the future, off-target effects using these techniques may be overcome by sgRNAs with high attraction for directed sequences and the choice of Cas9 with high fidelity, comprising good experimental measures. The other chief concern is the issues related to the commercialization of genome-amended crops because these conditions are not encouraging. Recently, the Court of Justice of the European Union stated that genome-amended crops should focus on the same principles as other genetically improved crops [215]. This ruling may delay investment in this technique in the EU. CRISPR has many drawbacks as it does not occur naturally in plants, which means that CRISPR/Cas9 proteins must be moved into plant cells which is a time-consuming process [216] and occasionally needs optimization of codon if Cas9 is from dissimilar backgrounds [217]. The incompetent transfer of CRISPR/dCas9 into the plants is the main obstacle to understanding the prospective of this tool. This is mostly due to the resistance of plant tissue and the incapability of plant tissue to redevelop. Therefore, a new transfer technique like direct transfer of Cas9 constructs into plant apical meristem to circumvent tissue culture is needed [218].
With more comprehensive research and more developments in this genome expurgation tool, the CRISPR/Cas9 system may play a significant role in breeding new crop plants for progress toward a sustainable agricultural system that may support a rapidly increasing global population. The efficiency of this technique is restricted because of its large size; thus, it is not suitable for packing into viral vectors for delivery into somatic tissues. For efficient genome editing, the application of CRISPR/Cas9 would yield the best results. CRISPR/Cas9 application can induce numerous accidental off-target changes in the genome [219]. Nevertheless, new CRISPR/Cas9 variants have improved the editing effectiveness of target bases in the sequence desired by the identification of dissimilar PAMs [220]. Problems occur in the commercialization of transgenic crops, thus articulating challenges with CRISPR/Cas9 application in several countries, mainly because of the costs and restrictions imposed by the regulatory system for the field release of genetically altered organisms. Some of the main challenges of CRISPR/Cas9 delivery system are a lack of efficacy or lower efficiency of the delivery system, which limits its use on a larger scale of genome editing. CRISPR/Cas9 based bystander mutations can cause several dysregulations in plants, so this is a huge challenge that needs to be addressed. The presence of off-target effects can minimize our editing efficiency, and this should be reduced using a careful selection of off-target sequences. The off-target effects consist ofunplanned points mutations, deletions, insertions inversions, as well as translocations. Multiple studies using early CRISPR/Cas9 agents revealed that larger than 50% of RNA-guided endonuclease-induced mutations were not generating on-target. Several techniques have been developed to reduce these off-target effects including, biased and unbiased off-target identification, cytosine adenine base manipulators, prime editing as well as truncated called gRNAs [221]. The inefficient Cas9 protein delivery system makes it difficult to reduce the duration of genomic DNA. The unequal molar ratio of sgRNA and CRISPR/Cas9 hinders the use of /CRISPR/Cas9, which is a major limitation as shown in Figure 5.

7. Conclusions and Future Prospects

The goal of producing safe and low-cost crops to meet the increasing global demands of food using various practices may pose challenges. The use of modern techniques to boost crop varieties will be an essential feature. The use of advanced breeding methods allows scientists to rapidly manipulate genes and insert the gene of interest into the genome compared to classical breeding methods. CRISPR/Cas9 is an essential revolutionary tool for gene editing. Therefore, in the future, the use of this technique to enhance yield, quality, and disease resistance in crops may be a significant field of research. During the last 5 years, it has been applied dynamically in myriad plant systems for conducting practical studies, combating stress-induced responses, and increasing significant agronomic characteristics. However, multiple modifications to this tool should lead to an increase in target effectiveness, and most studies are introductory and warrant development. Nonetheless, CRISPR/Cas9-based genome manipulation will attain a reputation and will be a critical method to attain the generation of “suitably manipulated” plants to help achieve the zero-starvation objective and to realize sustainable food production for the increasing human population. The progress in modern breeding methods has been remarkably accepted as a novelty in our capability to alter genomes and has consequently confronted our consideration of existing regulatory rules. As GE tools are widely used in plants, the protection of GE plants is a problem that warrants discussion worldwide. The regulatory rules for new crop novelties must be multidimensional, precise, and capable of differentiating between GE and genetically modified (GM) crops. Innovative systems biology, next-generation sequencing, and the newest advances in functional genomic methods, cohesive with innovative CRISPR/Cas9 tools, will allow smart crop development with greater yield and enhanced features. The CRISPR/Cas9 tool and speed breeding programs can be used to ensure global food security. CRISPR/Cas9 based genome editing system has the advantage of mixing it with next-generation sequencing. Now researchers can conduct comprehensive mutational screening. Optimization and proper designing of gRNAs are very important at each phase to avoid or reduce the deleterious effects while doing off-target gene editing. Therefore, the use of the CRISPR/CAS9 library has several advantages like high multiplexing, specificity as well as high throughout targeting of a gene. To reduce or nullify negative results, it is important to do a quality check of the CRISPR library at each point during the screening procedure. Analysis of gene function by the above method is critical to recognize the function of genes. Newly exposed CRISPR/Cas9 methods and the development of novel tools are being uninterruptedly described, signifying that CRISPR/Cas9 toolbox for plant engineering will increase further in the future. This set of tools will deliver new methods to achieve defined genome editing without any bits of transgenes residual in genome-edited plants.

Author Contributions

A.R. prepared the draft, R.A.G. reviewed and improved the draft. M.U.H., A.M., S.Q., Q.U.Z., M.I., and M.A. improved the manuscript. M.B. improved the scientific figures, Z.W. supervised the study, reviewed the manuscript, and H.L. provided funding for this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The research was supported by the National Natural Science Foundation of China (31560350, 31760350, and 71963020), the National Key Research and Development Program of China (2018YFD0301102), the Key Research and Development Program of Jiangxi Province (20171ACF60018 and 20192ACB60003),Natural Science Foundation of Jiangxi (20181BAA208055 and 20202BABL205020), the Jiangxi Agriculture Research System (JXARS-18) and Training Program for Academic and Technical Leaders of Major Discipline in Jiangxi Province (20204BCJL22044).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. Barrett, C.B. Overcoming global food security challenges through science and solidarity. Ame. J. Agric. Econ. 2021, 103, 422–447. [Google Scholar] [CrossRef]
  2. Adhikari, P.; Poudel, M. CRISPR-Cas9 in agriculture: Approaches, applications, future perspectives, and associated challenges. Malay. J. Halal Res. 2020, 3, 6–16. [Google Scholar] [CrossRef]
  3. Clarke, J.L.; Zhang, P. Plant biotechnology for food security and bioeconomy. Plant Mol. Biol. 2013, 83, 1–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. FAOSTAT. Food and Agriculture Data. Available online: http://www.fao.org/faostat/en/#home (accessed on 25 May 2021).
  5. Stamm, P.; Ramamoorthy, R.; Kumar, P.P. Feeding the extra billions: Strategies to improve crops and enhance future food security. Plant Biotech. Rep. 2011, 5, 107–120. [Google Scholar] [CrossRef]
  6. Jones, J.D.; Witek, K.; Verweij, W.; Jupe, F.; Cooke, D.; Dorling, S.; Tomlinson, L.; Smoker, M.; Perkins, S.; Foster, S. Elevating crop disease resistance with cloned genes. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Khan, Z.; Khan, S.H.; Ahmad, A. Challenges and future perspective of CRISPR/Cas technology for crop improvement. In CRISPR Crops; Springer: Singapore, 2021; pp. 289–306. [Google Scholar]
  8. Razzaq, A.; Saleem, F.; Kanwal, M.; Mustafa, G.; Yousaf, S.; Imran Arshad, H.M.; Hameed, M.K.; Khan, M.S.; Joyia, F.A. Modern trends in plant genome editing: An inclusive review of the CRISPR/Cas9 toolbox. Int. J. Mol. Sci. 2019, 20, 4045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Rasheed, A.; Hassan, M.; Aamer, M.; Bian, J.; Xu, Z.; He, X.; Wu, Z. Iron toxicity, tolerance and quantitative trait loci mapping in rice; a review. Appl. Ecol. Environ. Res. 2020, 18, 7483–7498. [Google Scholar] [CrossRef]
  10. Rasheed, A.; Hassan, M.U.; Aamer, M.; Batool, M.; Sheng, F.; Ziming, W.; Huijie, L. A critical review on the improvement of drought stress tolerance in rice (Oryza sativa L.). Not. Bot. Horti Agrobot. Cluj-Napoca 2020, 48, 1756–1788. [Google Scholar] [CrossRef]
  11. Rasheed, A.; Fahad, S.; Hassan, M.U.; Tahir, M.M.; Aamer, M.; Wu, Z. A review on aluminum toxicity and quantitative trait loci maping in rice (Oryza sative L). Appl. Ecol. Environ. Res. 2020, 18, 3951–3961. [Google Scholar] [CrossRef]
  12. Rasheed, A.; Fahad, S.; Aamer, M.; Hassan, M.U.; Tahir, M.M.; Wu, Z. Role of genetic factors in regulating cadmium uptake, transport and accumulation mechanisms and quantitative trait loci mapping in rice. a review. Appl. Ecol. Environ. Res. 2020, 18, 4005–4023. [Google Scholar] [CrossRef]
  13. Rasheed, A.; Hassan, M.U.; Fahad, S.; Aamer, M.; Batool, M.; Ilyas, M.; Shang, F.; Wu, Z.; Li, H. Heavy metals stress and plants defense responses. In Sustainable Soil and Land Management and Climate Change; CRC Press: Boca Raton, FL, USA; Taylor and Francis Group: Abingdon, UK, 2021; pp. 57–82. [Google Scholar]
  14. Rasheed, A.; Wassan, G.M.; Khanzada, H.; Solangi, A.M.; Aamer, M.; Ruicai, H.; Jianmin, B.; Ziming, W. QTL underlying iron toxicity tolerance at seedling stage in backcross recombinant inbred lines (BRILs) population of rice using high density genetic map. Not. Bot. Horti Agrobot. Cluj-Napoca 2021, 49, 12158. [Google Scholar] [CrossRef]
  15. Rasheed, A.; Wassan, G.M.; Khanzada, H.; Solangi, A.M.; Han, R.; Li, H.; Bian, J.; Wu, Z. Identification of genomic regions at seedling related traits in response to aluminium toxicity using a new high-density genetic map in rice (Oryza sativa L.). Genet. Res. Crop Evol. 2021, 68, 1889–1903. [Google Scholar] [CrossRef]
  16. Samanta, M.K.; Dey, A.; Gayen, S. CRISPR/Cas9: An advanced tool for editing plant genomes. Trans. Res. 2016, 25, 561–573. [Google Scholar] [CrossRef]
  17. Jaganathan, D.; Ramasamy, K.; Sellamuthu, G.; Jayabalan, S.; Venkataraman, G. CRISPR for crop improvement: An update review. Front. Plant Sci. 2018, 9, 985. [Google Scholar] [CrossRef] [PubMed]
  18. Gillani, S.F.; Rasheed, A.; Majeed, Y.; Tariq, H.; Yunling, P. Recent advancements on use of CRISPR/Cas9 in maize yield and quality improvement. Not. Bot. Horti Agrobot. Cluj-Napoca 2021, 49, 12459. [Google Scholar] [CrossRef]
  19. Gaj, T.; Gersbach, C.A.; Barbas III, C.F. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotech. 2013, 31, 397–405. [Google Scholar] [CrossRef] [Green Version]
  20. Puchta, H. Applying CRISPR/Cas for genome engineering in plants: The best is yet to come. Curr. Opin. Plant Biol. 2017, 36, 1–8. [Google Scholar] [CrossRef]
  21. Tang, X.; Lowder, L.G.; Zhang, T.; Malzahn, A.A.; Zheng, X.; Voytas, D.F.; Zhong, Z.; Chen, Y.; Ren, Q.; Li, Q. A CRISPR–Cpf1 system for efficient genome editing and transcriptional repression in plants. Nat. Plants 2017, 3, 1–5. [Google Scholar]
  22. Boch, J.; Scholze, H.; Schornack, S.; Landgraf, A.; Hahn, S.; Kay, S.; Lahaye, T.; Nickstadt, A.; Bonas, U. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 2009, 326, 1509–1512. [Google Scholar] [CrossRef]
  23. Chen, K.; Wang, Y.; Zhang, R.; Zhang, H.; Gao, C. CRISPR/Cas genome editing and precision plant breeding in agriculture. Ann. Rev. Plant Biol. 2019, 70, 667–697. [Google Scholar] [CrossRef]
  24. Zhou, J.; Li, D.; Wang, G.; Wang, F.; Kunjal, M.; Joldersma, D.; Liu, Z. Application and future perspective of CRISPR/Cas9 genome editing in fruit crops. J. Integr. Plant Biol. 2020, 62, 269–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bolotin, A.; Quinquis, B.; Sorokin, A.; Ehrlich, S.D. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology 2005, 151, 2551–2561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Pourcel, C.; Salvignol, G.; Vergnaud, G. CRISPR elements in Yersinia pestis acquire new repeats by preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies. Microbiology 2005, 151, 653–663. [Google Scholar] [CrossRef] [Green Version]
  27. Leibowitz, M.L.; Papathanasiou, S.; Doerfler, P.A.; Blaine, L.J.; Sun, L.; Yao, Y.; Zhang, C.-Z.; Weiss, M.J.; Pellman, D. Chromothripsis as an on-target consequence of CRISPR–Cas9 genome editing. Nat. Genet. 2021, 53, 889–905. [Google Scholar] [CrossRef]
  28. Zhan, X.; Lu, Y.; Zhu, J.K.; Botella, J.R. Genome editing for plant research and crop improvement. J. Integr. Plant Biol. 2021, 63, 3–33. [Google Scholar] [CrossRef] [PubMed]
  29. Barrangou, R.; Fremaux, C.; Deveau, H.; Richards, M.; Boyaval, P.; Moineau, S.; Romero, D.A.; Horvath, P. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315, 1709–1712. [Google Scholar] [CrossRef]
  30. Sharma, S.; Vakhlu, J. Evolution and Biology of CRISPR System: A New Era Tool for Genome Editing in Plants. Bot. Rev. 2021, 1–22. [Google Scholar] [CrossRef]
  31. Sharma, A.; Badola, P.K.; Trivedi, P.K. CRISPR-Cas9 System for agriculture crop improvement. In Genome Engineering for Crop Improvement; Wiley: Hoboken, NJ, USA, 2021; pp. 97–111. [Google Scholar]
  32. Gao, C. Genome engineering for crop improvement and future agriculture. Cell 2021, 184, 1621–1635. [Google Scholar] [CrossRef]
  33. Namo, F.M.; Belachew, G.T. Genome editing technologies for crop improvement: Current status and future prospective. Plant Cell Biotech. Mol. Biol. 2021, 22, 1–19. [Google Scholar]
  34. Mishra, R.; Zhao, K. Genome editing technologies and their applications in crop improvement. Plant Biotech. Rep. 2018, 12, 57–68. [Google Scholar] [CrossRef]
  35. Townsend, J.A.; Wright, D.A.; Winfrey, R.J.; Fu, F.; Maeder, M.L.; Joung, J.K.; Voytas, D.F. High-frequency modification of plant genes using engineered zinc-finger nucleases. Nature 2009, 459, 442–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Zhang, F.; Maeder, M.L.; Unger-Wallace, E.; Hoshaw, J.P.; Reyon, D.; Christian, M.; Li, X.; Pierick, C.J.; Dobbs, D.; Peterson, T. High frequency targeted mutagenesis in Arabidopsis thaliana using zinc finger nucleases. Proc. Natl. Acad. Sci. USA 2010, 107, 12028–12033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Puchta, H. The repair of double-strand breaks in plants: Mechanisms and consequences for genome evolution. J. Exp. Bot. 2005, 56, 1–14. [Google Scholar] [CrossRef] [Green Version]
  38. D’Halluin, K.; Vanderstraeten, C.; Van Hulle, J.; Rosolowska, J.; Van Den Brande, I.; Pennewaert, A.; D’Hont, K.; Bossut, M.; Jantz, D.; Ruiter, R. Targeted molecular trait stacking in cotton through targeted double-strand break induction. Plant Biotech. J. 2013, 11, 933–941. [Google Scholar] [CrossRef] [Green Version]
  39. Djukanovic, V.; Smith, J.; Lowe, K.; Yang, M.; Gao, H.; Jones, S.; Nicholson, M.G.; West, A.; Lape, J.; Bidney, D. Male-sterile maize plants produced by targeted mutagenesis of the cytochrome P 450-like gene (MS 26) using a re-designed I–C reI homing endonuclease. Plant J. 2013, 76, 888–899. [Google Scholar] [CrossRef]
  40. Durai, S.; Mani, M.; Kandavelou, K.; Wu, J.; Porteus, M.H.; Chandrasegaran, S. Zinc finger nucleases: Custom-designed molecular scissors for genome engineering of plant and mammalian cells. Nucleic Acids Res. 2005, 33, 5978–5990. [Google Scholar] [CrossRef]
  41. Carlson, D.F.; Fahrenkrug, S.C.; Hackett, P.B. Targeting DNA with fingers and TALENs. Mol. Ther. Nucleic Acids 2012, 1, e3. [Google Scholar] [CrossRef]
  42. Carroll, D.; Morton, J.J.; Beumer, K.J.; Segal, D.J. Design, construction and in vitro testing of zinc finger nucleases. Nat. Protoc. 2006, 1, 1329–1341. [Google Scholar] [CrossRef]
  43. Minczuk, M.; Papworth, M.A.; Miller, J.C.; Murphy, M.P.; Klug, A. Development of a single-chain, quasi-dimeric zinc-finger nuclease for the selective degradation of mutated human mitochondrial DNA. Nucleic Acids Res. 2008, 36, 3926–3938. [Google Scholar] [CrossRef] [Green Version]
  44. Gaj, T.; Guo, J.; Kato, Y.; Sirk, S.J.; Barbas, C.F. Targeted gene knockout by direct delivery of zinc-finger nuclease proteins. Nat. Methods 2012, 9, 805–807. [Google Scholar] [CrossRef] [Green Version]
  45. Zhang, H.-X.; Zhang, Y.; Yin, H. Genome editing with mRNA encoding ZFN, TALEN, and Cas9. Mol. Ther. 2019, 27, 735–746. [Google Scholar] [CrossRef] [Green Version]
  46. Schornack, S.; Meyer, A.; Römer, P.; Jordan, T.; Lahaye, T. Gene-for-gene-mediated recognition of nuclear-targeted AvrBs3-like bacterial effector proteins. J. Plant Physiol. 2006, 163, 256–272. [Google Scholar] [CrossRef] [PubMed]
  47. Römer, P.; Hahn, S.; Jordan, T.; Strauss, T.; Bonas, U.; Lahaye, T. Plant pathogen recognition mediated by promoter activation of the pepper Bs3 resistance gene. Science 2007, 318, 645–648. [Google Scholar] [CrossRef] [Green Version]
  48. Shan, Q.; Zhang, Y.; Chen, K.; Zhang, K.; Gao, C. Creation of fragrant rice by targeted knockout of the Os BADH 2 gene using TALEN technology. Plant Biotech. J. 2015, 13, 791–800. [Google Scholar] [CrossRef] [PubMed]
  49. Králová, M.; Bergougnoux, V.; Frébort, I. CRISPR/Cas9 genome editing in ergot fungus Clavicepspurpurea. J. Biotechnol. 2021, 325, 341–354. [Google Scholar] [CrossRef]
  50. Wang, Y.; Cheng, X.; Shan, Q.; Zhang, Y.; Liu, J.; Gao, C.; Qiu, J.-L. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat. Biotech. 2014, 32, 947–951. [Google Scholar] [CrossRef]
  51. Gao, H.; Smith, J.; Yang, M.; Jones, S.; Djukanovic, V.; Nicholson, M.G.; West, A.; Bidney, D.; Falco, S.C.; Jantz, D. Heritable targeted mutagenesis in maize using a designed endonuclease. Plant J. 2010, 61, 176–187. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, Y.; Zhang, F.; Li, X.; Baller, J.A.; Qi, Y.; Starker, C.G.; Bogdanove, A.J.; Voytas, D.F. Transcription activator-like effector nucleases enable efficient plant genome engineering. Plant Physiol. 2013, 161, 20–27. [Google Scholar] [CrossRef] [Green Version]
  53. Gurushidze, M.; Hensel, G.; Hiekel, S.; Schedel, S.; Valkov, V.; Kumlehn, J. True-breeding targeted gene knock-out in barley using designer TALE-nuclease in haploid cells. PLoS ONE 2014, 9, e92046. [Google Scholar] [CrossRef] [Green Version]
  54. Liang, Z.; Zhang, K.; Chen, K.; Gao, C. Targeted mutagenesis in Zea mays using TALENs and the CRISPR/Cas system. J. Genet. Genom. 2014, 41, 63–68. [Google Scholar] [CrossRef]
  55. Kusano, H.; Onodera, H.; Kihira, M.; Aoki, H.; Matsuzaki, H.; Shimada, H. A simple Gateway-assisted construction system of TALEN genes for plant genome editing. Sci. Rep. 2016, 6, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Liu, Q.; Yang, F.; Zhang, J.; Liu, H.; Rahman, S.; Islam, S.; Ma, W.; She, M. Application of CRISPR/Cas9 in Crop quality improvement. Int. J. Mol. Sci. 2021, 22, 4206. [Google Scholar] [CrossRef] [PubMed]
  57. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  58. 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. 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]
  59. Turnbull, C.; Lillemo, M.; Hvoslef-Eide, T.A. Global Regulation of Genetically Modified Crops Amid the Gene Edited Crop Boom–A Review. Front. Plant Sci. 2021, 12, 258. [Google Scholar] [CrossRef] [PubMed]
  60. Symington, L.S.; Gautier, J. Double-strand break end resection and repair pathway choice. Ann. Rev. Genet. 2011, 45, 247–271. [Google Scholar] [CrossRef]
  61. Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of A• T to G• C in genomic DNA without DNA cleavage. Nature 2017, 551, 464–471. [Google Scholar] [CrossRef] [PubMed]
  62. Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420–424. [Google Scholar] [CrossRef] [Green Version]
  63. Mojica, F.J.; Díez-Villaseñor, C.; García-Martínez, J.; Almendros, C. Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology 2009, 155, 733–740. [Google Scholar] [CrossRef] [Green Version]
  64. Brouns, S.J.; Jore, M.M.; Lundgren, M.; Westra, E.R.; Slijkhuis, R.J.; Snijders, A.P.; Dickman, M.J.; Makarova, K.S.; Koonin, E.V.; Van Der Oost, J. Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 2008, 321, 960–964. [Google Scholar] [CrossRef] [Green Version]
  65. Garneau, J.E.; Dupuis, M.-È.; Villion, M.; Romero, D.A.; Barrangou, R.; Boyaval, P.; Fremaux, C.; Horvath, P.; Magadán, A.H.; Moineau, S. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 2010, 468, 67–71. [Google Scholar] [CrossRef] [PubMed]
  66. Rahim, J.; Gulzar, S.; Zahid, R.; Rahim, K. A Systematic review on the comparison of molecular gene editing tools. Int. J. Innov. Sci. Res. Tech. 2021, 6, 1–8. [Google Scholar]
  67. Eş, I.; Gavahian, M.; Marti-Quijal, F.J.; Lorenzo, J.M.; Khaneghah, A.M.; Tsatsanis, C.; Kampranis, S.C.; Barba, F.J. The application of the CRISPR-Cas9 genome editing machinery in food and agricultural science: Current status, future perspectives, and associated challenges. Biotech. Adv. 2019, 37, 410–421. [Google Scholar] [CrossRef] [PubMed]
  68. Sauer, N.J.; Mozoruk, J.; Miller, R.B.; Warburg, Z.J.; Walker, K.A.; Beetham, P.R.; Schöpke, C.R.; Gocal, G.F. Oligonucleotide-directed mutagenesis for precision gene editing. Plant Biotech. J. 2016, 14, 496–502. [Google Scholar] [CrossRef] [Green Version]
  69. Upadhyay, S.K.; Kumar, J.; Alok, A.; Tuli, R. RNA-guided genome editing for target gene mutations in wheat. G3 Genes Genomes Genet. 2013, 3, 2233–2238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Bhardwaj, A.; Nain, V. TALENs—An indispensable tool in the era of CRISPR: A mini review. J. Genet. Eng. Biotechnol. 2021, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
  71. Khandagale, K.; Nadaf, A. Genome editing for targeted improvement of plants. Plant Biotechnol. Rep. 2016, 10, 327–343. [Google Scholar] [CrossRef]
  72. Prajapat, R.K.; Mathur, M.; Upadhyay, T.K.; Lal, D.; Maloo, S.; Sharma, D. Genome Editing for Crop Improvement. In Crop Improvement; CRC Press: Boca Raton, FL, USA, 2021; pp. 111–123. [Google Scholar]
  73. Makarova, K.S.; Haft, D.H.; Barrangou, R.; Brouns, S.J.; Charpentier, E.; Horvath, P.; Moineau, S.; Mojica, F.J.; Wolf, Y.I.; Yakunin, A.F. Evolution and classification of the CRISPR–Cas systems. Nat. Rev. Microbiol. 2011, 9, 467–477. [Google Scholar] [CrossRef] [Green Version]
  74. Deltcheva, E.; Chylinski, K.; Sharma, C.M.; Gonzales, K.; Chao, Y.; Pirzada, Z.A.; Eckert, M.R.; Vogel, J.; Charpentier, E. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 2011, 471, 602–607. [Google Scholar] [CrossRef] [Green Version]
  75. Hoikkala, V.; Ravantti, J.; Díez-Villaseñor, C.; Tiirola, M.; Conrad, R.A.; McBride, M.J.; Moineau, S.; Sundberg, L.-R. Cooperation between different CRISPR-Cas types enables adaptation in an RNA-targeting system. Mbio 2021, 12, e03338-20. [Google Scholar] [CrossRef]
  76. Liao, C.; Beisel, C.L. The tracrRNA in CRISPR biology and technologies. Annu. Rev. Genet. 2021, 55. [Google Scholar] [CrossRef] [PubMed]
  77. Hsu, P.D.; Scott, D.A.; Weinstein, J.A.; Ran, F.A.; Konermann, S.; Agarwala, V.; Li, Y.; Fine, E.J.; Wu, X.; Shalem, O. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotech. 2013, 31, 827–832. [Google Scholar] [CrossRef] [PubMed]
  78. Carabias, A.; Fuglsang, A.; Temperini, P.; Pape, T.; Sofos, N.; Stella, S.; Erlendsson, S.; Montoya, G. Structure of the mini-RNA-guided endonuclease CRISPR-Cas12j3. Nat. Commun. 2021, 12, 4476. [Google Scholar] [CrossRef] [PubMed]
  79. Xie, S.; Shen, B.; Zhang, C.; Huang, X.; Zhang, Y. sgRNAcas9: A software package for designing CRISPR sgRNA and evaluating potential off-target cleavage sites. PLoS ONE 2014, 9, e100448. [Google Scholar] [CrossRef]
  80. Stemmer, M.; Thumberger, T.; del Sol Keyer, M.; Wittbrodt, J.; Mateo, J.L. CCTop: An intuitive, flexible and reliable CRISPR/Cas9 target prediction tool. PLoS ONE 2015, 10, e0124633. [Google Scholar] [CrossRef] [Green Version]
  81. Rastogi, A.; Murik, O.; Bowler, C.; Tirichine, L. PhytoCRISP-Ex: A web-based and stand-alone application to find specific target sequences for CRISPR/CAS editing. BMC Bioinf. 2016, 17, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wu, Z.; Zhang, Y.; Yu, H.; Pan, D.; Wang, Y.; Wang, Y.; Li, F.; Liu, C.; Nan, H.; Chen, W. Programmed genome editing by a miniature CRISPR-Cas12f nuclease. J. Nat. Chem. Biol. 2021, 17, 1132–1138. [Google Scholar] [CrossRef]
  83. Chari, R.; Yeo, N.C.; Chavez, A.; Church, G.M. sgRNA Scorer 2.0: A species-independent model to predict CRISPR/Cas9 activity. ACS Synth. Biol. 2017, 6, 902–904. [Google Scholar] [CrossRef] [Green Version]
  84. Sun, J.; Liu, H.; Liu, J.; Cheng, S.; Peng, Y.; Zhang, Q.; Yan, J.; Liu, H.-J.; Chen, L.-L. CRISPR-Local: A local single-guide RNA (sgRNA) design tool for non-reference plant genomes. Bioinformatics 2018, 35, 2501–2503. [Google Scholar] [CrossRef]
  85. Chen, W.; Zhang, G.; Li, J.; Zhang, X.; Huang, S.; Xiang, S.; Hu, X.; Liu, C. CRISPRlnc: A manually curated database of validated sgRNAs for lncRNAs. Nucleic Acids Res. 2019, 47, D63–D68. [Google Scholar] [CrossRef] [Green Version]
  86. Braatz, J.; Harloff, H.-J.; Mascher, M.; Stein, N.; Himmelbach, A.; Jung, C. CRISPR-Cas9 targeted mutagenesis leads to simultaneous modification of different homoeologous gene copies in polyploid oilseed rape (Brassica napus). Plant Physiol. 2017, 174, 935–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hsu, P.D.; Lander, E.S.; Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 2014, 157, 1262–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A. Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339, 819–823. [Google Scholar] [CrossRef] [Green Version]
  89. Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; DiCarlo, J.E.; Norville, J.E.; Church, G.M. RNA-guided human genome engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef] [Green Version]
  90. Nekrasov, V.; Staskawicz, B.; Weigel, D.; Jones, J.D.; Kamoun, S. Targeted mutagenesis in the model plant Nicotiana benthamiana using Cas9 RNA-guided endonuclease. Nat. Biotech. 2013, 31, 691–693. [Google Scholar] [CrossRef] [PubMed]
  91. Shan, Q.; Wang, Y.; Li, J.; Zhang, Y.; Chen, K.; Liang, Z.; Zhang, K.; Liu, J.; Xi, J.J.; Qiu, J.-L. Targeted genome modification of crop plants using a CRISPR-Cas system. Nat. Biotech. 2013, 31, 686–688. [Google Scholar] [CrossRef]
  92. Gao, C. The future of CRISPR technologies in agriculture. Nat. Rev. Mol. Cell Biol. 2018, 19, 275–276. [Google Scholar] [CrossRef] [PubMed]
  93. Ma, X.; Liu, Y.G. CRISPR/Cas9-based multiplex genome editing in monocot and dicot plants. Curr. Protoc. Mol. Biol. 2016, 115. [Google Scholar] [CrossRef] [PubMed]
  94. Ricroch, A.; Clairand, P.; Harwood, W. Use of CRISPR systems in plant genome editing: Toward new opportunities in agriculture. Emerg. Top. Life Sci. 2017, 1, 169–182. [Google Scholar]
  95. Zhang, D.; Li, Z.; Li, J.-F. Targeted gene manipulation in plants using the CRISPR/Cas technology. J. Genet. Genom. 2016, 43, 251–262. [Google Scholar] [CrossRef] [PubMed]
  96. Hua, K.; Zhang, J.; Botella, J.R.; Ma, C.; Kong, F.; Liu, B.; Zhu, J.-K. Perspectives on the application of genome-editing technologies in crop breeding. Mol. Plant 2019, 12, 1047–1059. [Google Scholar] [CrossRef] [Green Version]
  97. Schindele, A.; Dorn, A.; Puchta, H. CRISPR/Cas brings plant biology and breeding into the fast lane. Curr. Opin. Biotechnol. 2020, 61, 7–14. [Google Scholar] [CrossRef]
  98. Tan, Y.-y.; Du, H.; Wu, X.; Liu, Y.-h.; Jiang, M.; Song, S.-y.; Wu, L.; Shu, Q.-y. Gene editing: An instrument for practical application of gene biology to plant breeding. J. Zhejiang Univ.-Sci. B 2020, 21, 460–473. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, Y.; Malzahn, A.A.; Sretenovic, S.; Qi, Y. The emerging and uncultivated potential of CRISPR technology in plant science. Nat. Plants 2019, 5, 778–794. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, T.; Zhang, C.; Zhang, H.; Zhu, H. CRISPR/Cas9-mediated gene editing revolutionizes the improvement of horticulture food crops. J. Agric. Food Chem. 2021. [Google Scholar] [CrossRef] [PubMed]
  101. Hyun, Y.; Kim, J.; Cho, S.W.; 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] [PubMed] [Green Version]
  102. Zhang, Z.; Mao, Y.; Ha, S.; Liu, W.; Botella, J.R.; Zhu, J.-K. A multiplex CRISPR/Cas9 platform for fast and efficient editing of multiple genes in Arabidopsis. Plant Cell Rep. 2016, 35, 1519–1533. [Google Scholar] [CrossRef] [Green Version]
  103. Gao, J.; Wang, G.; Ma, S.; Xie, X.; Wu, X.; Zhang, X.; Wu, Y.; Zhao, P.; Xia, Q. CRISPR/Cas9-mediated targeted mutagenesis in Nicotiana tabacum. Plant Mol. Biol. 2015, 87, 99–110. [Google Scholar] [CrossRef]
  104. Zhang, H.; Zhang, J.; Wei, P.; Zhang, B.; Gou, F.; Feng, Z.; Mao, Y.; Yang, L.; Zhang, H.; Xu, N. The CRISPR/C as9 system produces specific and homozygous targeted gene editing in rice in one generation. Plant Biotech. J. 2014, 12, 797–807. [Google Scholar] [CrossRef]
  105. Xu, R.-F.; Li, H.; Qin, R.-Y.; Li, J.; Qiu, C.-H.; Yang, Y.-C.; Ma, H.; Li, L.; Wei, P.-C.; Yang, J.-B. Generation of inheritable and “transgene clean” targeted genome-modified rice in later generations using the CRISPR/Cas9 system. Sci. Rep. 2015, 5, 11491. [Google Scholar] [CrossRef] [Green Version]
  106. Lawrenson, T.; Shorinola, O.; Stacey, N.; Li, C.; Østergaard, 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] [PubMed] [Green Version]
  107. Shen, L.; Wang, C.; Fu, Y.; Wang, J.; Liu, Q.; Zhang, X.; Yan, C.; Qian, Q.; Wang, K. QTL editing confers opposing yield performance in different rice varieties. J. Integr. Plant Biol. 2018, 60, 89–93. [Google Scholar] [CrossRef] [PubMed]
  108. Usman, B.; Nawaz, G.; Zhao, N.; Liao, S.; Qin, B.; Liu, F.; Liu, Y.; Li, R. Programmed Editing of Rice (Oryza sativa L.) OsSPL16 gene using CRISPR/Cas9 improves grain yield by modulating the expression of pyruvate enzymes and cell cycle proteins. Int. J. Mol. Sci. 2021, 22, 249. [Google Scholar] [CrossRef]
  109. Wang, Y.; Wang, J.; Guo, S.; Tian, S.; Zhang, J.; Ren, Y.; Li, M.; Gong, G.; Zhang, H.; Xu, Y. CRISPR/Cas9-mediated mutagenesis of ClBG1 decreased seed size and promoted seed germination in watermelon. Hort. Res. 2021, 8, 1–12. [Google Scholar] [CrossRef]
  110. Monsur, M.B.; Ni, C.; Xiangjin, W.; Lihong, X.; Guiai, J.; Shaoqing, T.; Sreenivasulu, N.; Gaoneng, S.; Peisong, H. Improved eating and cooking quality of indica rice cultivar YK17 via adenine base editing of waxya allele of granule-bound starch synthase I (GBSS I). Rice Sci. 2021, 1, 427–430. [Google Scholar] [CrossRef]
  111. Zhang, J.; Zhou, Z.; Bai, J.; Tao, X.; Wang, L.; Zhang, H.; Zhu, J.-K. Disruption of MIR396e and MIR396f improves rice yield under nitrogen-deficient conditions. Nat. Sci. Rev. 2020, 7, 102–112. [Google Scholar] [CrossRef] [Green Version]
  112. Miao, C.; Wang, Z.; Zhang, L.; Yao, J.; Hua, K.; Liu, X.; Shi, H.; Zhu, J.-K. The grain yield modulator miR156 regulates seed dormancy through the gibberellin pathway in rice. Nat. Commun. 2019, 10, 3822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Usman, B.; Nawaz, G.; Zhao, N.; Liu, Y.; Li, R. Generation of high yielding and fragrant rice (Oryza sativa L.) Lines by CRISPR/Cas9 targeted mutagenesis of three homoeologs of cytochrome P450 gene family and OsBADH2 and transcriptome and proteome profiling of revealed changes triggered by mutations. Plants 2020, 9, 788. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, S.; Yang, Y.; Guo, M.; Zhong, C.; Yan, C.; Sun, S. 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]
  115. 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. 2020, 10, 1663. [Google Scholar] [CrossRef] [Green Version]
  116. Li, J.; Jiao, G.; Sun, Y.; Chen, J.; Zhong, Y.; Yan, L.; Jiang, D.; Ma, Y.; Xia, L. Modification of starch composition, structure and properties through editing of TaSBEIIa in both winter and spring wheat varieties by CRISPR/Cas9. Plant Biotech. J. 2020, 19, 937–951. [Google Scholar] [CrossRef] [PubMed]
  117. Okuzaki, A.; Ogawa, T.; Koizuka, C.; Kaneko, K.; Inaba, M.; Imamura, J.; Koizuka, N. CRISPR/Cas9-mediated genome editing of the fatty acid desaturase 2 gene in Brassica napus. Plant Physiol. Biochem. 2018, 131, 63–69. [Google Scholar] [CrossRef] [PubMed]
  118. Ellison, E.E.; Nagalakshmi, U.; Gamo, M.E.; Huang, P.-J.; Dinesh-Kumar, S.; Voytas, D.F. Multiplexed heritable gene editing using RNA viruses and mobile single guide RNAs. Nat. Plants 2020, 6, 620–624. [Google Scholar] [CrossRef] [PubMed]
  119. Morineau, C.; Bellec, Y.; Tellier, F.; Gissot, L.; Kelemen, Z.; Nogué, F.; Faure, J.D. Selective gene dosage by CRISPR-Cas9 genome editing in hexaploid Camelina sativa. Plant Botech. J. 2017, 15, 729–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Zhai, Y.; Cai, S.; Hu, L.; Yang, Y.; Amoo, O.; Fan, C.; Zhou, Y. CRISPR/Cas9-mediated genome editing reveals differences in the contribution of INDEHISCENT homologues to pod shatter resistance in Brassica napus L. Theor. Appl. Genet. 2019, 132, 2111–2123. [Google Scholar] [CrossRef]
  121. Zaman, Q.U.; Chu, W.; Hao, M.; Shi, Y.; Sun, M.; Sang, S.-F.; Mei, D.; Cheng, H.; Liu, J.; Li, C. CRISPR/Cas9-Mediated Multiplex Genome Editing of JAGGED Gene in Brassica napus L. Biomolecules 2019, 9, 725. [Google Scholar] [CrossRef] [Green Version]
  122. Soyk, S.; Müller, N.A.; Park, S.J.; Schmalenbach, I.; Jiang, K.; Hayama, R.; Zhang, L.; Van Eck, J.; Jiménez-Gómez, J.M.; Lippman, Z.B. Variation in the flowering gene SELF PRUNING 5G promotes day-neutrality and early yield in tomato. Nat. Genet. 2017, 49, 162–168. [Google Scholar] [CrossRef]
  123. Brooks, C.; Nekrasov, V.; Lippman, Z.B.; Van Eck, J. Efficient gene editing in tomato in the first generation using the clustered regularly interspaced short palindromic repeats/CRISPR-associated9 system. Plant Physiol. 2014, 166, 1292–1297. [Google Scholar] [CrossRef] [Green Version]
  124. Malnoy, M.; Viola, R.; Jung, M.-H.; Koo, O.-J.; Kim, S.; Kim, J.-S.; Velasco, R.; Nagamangala Kanchiswamy, C. DNA-free genetically edited grapevine and apple protoplast using CRISPR/Cas9 ribonucleoproteins. Front. Plant Sci. 2016, 7, 1904. [Google Scholar] [CrossRef]
  125. Toda, E.; Koiso, N.; Takebayashi, A.; Ichikawa, M.; Kiba, T.; Osakabe, K.; Osakabe, Y.; Sakakibara, H.; Kato, N.; Okamoto, T. An efficient DNA-and selectable-marker-free genome-editing system using zygotes in rice. Nat. Plants 2019, 5, 363. [Google Scholar] [CrossRef]
  126. Espley, R.V.; Leif, D.; Plunkett, B.; McGhie, T.; Henry-Kirk, R.; Hall, M.; Johnston, J.W.; Punter, M.P.; Boldingh, H.; Nardozza, S. Red to brown: An elevated anthocyanic response in apple drives ethylene to advance maturity and fruit flesh browning. Front. Plant Sci. 2019, 10, 1248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. 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 Sl AGAMOUS-LIKE 6 loss of function. Plant Biotech. J. 2017, 15, 634–647. [Google Scholar] [CrossRef] [PubMed]
  128. Jia, H.; Wang, N. Targeted genome editing of sweet orange using Cas9/sgRNA. PLoS ONE 2014, 9, e93806. [Google Scholar]
  129. Lemmon, Z.H.; Reem, N.T.; Dalrymple, J.; Soyk, S.; Swartwood, K.E.; Rodriguez-Leal, D.; Van Eck, J.; Lippman, Z.B. Rapid improvement of domestication traits in an orphan crop by genome editing. Nat. Plants 2018, 4, 766–770. [Google Scholar] [CrossRef] [PubMed]
  130. Naim, F.; Dugdale, B.; Kleidon, J.; Brinin, A.; Shand, K.; Waterhouse, P.; Dale, J. Gene editing the phytoene desaturase alleles of Cavendish banana using CRISPR/Cas9. Transg. Res. 2018, 27, 451–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. 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]
  132. Ueta, R.; Abe, C.; Watanabe, T.; Sugano, S.S.; Ishihara, R.; Ezura, H.; Osakabe, Y.; Osakabe, K. Rapid breeding of parthenocarpic tomato plants using CRISPR/Cas9. Sci. Rep. 2017, 7, 507. [Google Scholar] [CrossRef] [Green Version]
  133. Cai, Y.; Chen, L.; Liu, X.; Guo, C.; Sun, S.; Wu, C.; Jiang, B.; Han, T.; Hou, W. CRISPR/Cas9-mediated targeted mutagenesis of GmFT2a delays flowering time in soya bean. Plant Biotech. J. 2018, 16, 176–185. [Google Scholar] [CrossRef] [Green Version]
  134. 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 Biotech. J. 2020, 18, 2241–2250. [Google Scholar] [CrossRef] [Green Version]
  135. Badhan, S.; Ball, A.S.; Mantri, N. First report of CRISPR/Cas9 mediated DNA-free editing of 4CL and RVE7 genes in chickpea protoplasts. Int. J. Mol. Sci. 2021, 22, 396. [Google Scholar] [CrossRef]
  136. Zheng, M.; Zhang, L.; Tang, M.; Liu, J.; Liu, H.; Yang, H.; Fan, S.; Terzaghi, W.; Wang, H.; Hua, W. Knockout of two Bna MAX 1 homologs by CRISPR/Cas9-targeted mutagenesis improves plant architecture and increases yield in rapeseed (Brassica napus L.). Plant Biotech. J. 2020, 18, 644–654. [Google Scholar] [CrossRef] [Green Version]
  137. Li, A.; Jia, S.; Yobi, A.; Ge, Z.; Sato, S.J.; Zhang, C.; Angelovici, R.; Clemente, T.E.; Holding, D.R. Editing of an alpha-kafirin gene family increases, digestibility and protein quality in sorghum. Plant Physiol. 2018, 177, 1425–1438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Andersson, M.; Turesson, H.; Nicolia, A.; Fält, A.-S.; Samuelsson, M.; Hofvander, P. Efficient targeted multiallelic mutagenesis in tetraploid potato (Solanum tuberosum) by transient CRISPR-Cas9 expression in protoplasts. Plant Cell Rep. 2017, 36, 117–128. [Google Scholar] [CrossRef] [Green Version]
  139. Li, M.; Li, X.; Zhou, Z.; Wu, P.; Fang, M.; Pan, X.; Lin, Q.; Luo, W.; Wu, G.; Li, H. Reassessment of the four yield-related genes Gn1a, DEP1, GS3, and IPA1 in rice using a CRISPR/Cas9 system. Front. Plant Sci. 2016, 7, 377. [Google Scholar] [CrossRef] [Green Version]
  140. Tian, S.; Jiang, L.; Gao, Q.; Zhang, J.; Zong, M.; Zhang, H.; Ren, Y.; Guo, S.; Gong, G.; Liu, F. Efficient CRISPR/Cas9-based gene knockout in watermelon. Plant Cell Rep. 2017, 36, 399–406. [Google Scholar] [CrossRef]
  141. Čermák, T.; Baltes, N.J.; Čegan, R.; Zhang, Y.; Voytas, D.F. High-frequency, precise modification of the tomato genome. Genom. Biol. 2015, 16, 232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Li, R.; Fu, D.; Zhu, B.; Luo, Y.; Zhu, H. CRISPR/Cas9-mediated mutagenesis of lncRNA1459 alters tomato fruit ripening. Plant J. 2018, 94, 513–524. [Google Scholar] [CrossRef] [Green Version]
  143. Qi, W.; Zhu, T.; Tian, Z.; Li, C.; Zhang, W.; Song, R. High-efficiency CRISPR/Cas9 multiplex gene editing using the glycine tRNA-processing system-based strategy in maize. BMC Biotech. 2016, 16, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Zhang, Y.; Liang, Z.; Zong, Y.; Wang, Y.; Liu, J.; Chen, K.; Qiu, J.-L.; Gao, C. Efficient and transgene-free genome editing in wheat through transient expression of CRISPR/Cas9 DNA or RNA. Nat. Commun. 2016, 7, 12617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Kaur, N.; Alok, A.; Kaur, N.; Pandey, P.; Awasthi, P.; Tiwari, S. CRISPR/Cas9-mediated efficient editing in phytoene desaturase (PDS) demonstrates precise manipulation in banana cv. Rasthali genome. Funct. Integr. Genom. 2018, 18, 89–99. [Google Scholar] [CrossRef]
  146. Jiang, W.Z.; Henry, I.M.; Lynagh, P.G.; Comai, L.; Cahoon, E.B.; Weeks, D.P. Significant enhancement of fatty acid composition in seeds of the allohexaploid, Camelina sativa, using CRISPR/Cas9 gene editing. Plant. Biotech. J. 2017, 15, 648–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Martín-Pizarro, C.; Triviño, J.C.; Posé, D. Functional analysis of the TM6 MADS-box gene in the octoploid strawberry by CRISPR/Cas9-directed mutagenesis. J. Exp. Bot. 2019, 70, 885–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Ma, J.; Sun, S.; Whelan, J.; Shou, H. CRISPR/Cas9-Mediated knockout of GmFATB1 Significantly reduced the amount of saturated fatty acids in soybean seeds. Int. J. Mol. Sci. 2021, 22, 3877. [Google Scholar] [CrossRef]
  149. Singh, R. Genome Editing for Plant Disease Resistance. In Emerging Trends in Plant Pathology; Springer: Berlin/Heidelberg, Germany, 2021; pp. 577–590. [Google Scholar]
  150. Khan, Z.; Saboor, T.; Ashfaq, M.; Saddique, A.; Khanum, P. Disease resistance in crops through CRISPR/Cas. In CRISPR Crops; Springer: Berlin/Heidelberg, Germany, 2021; pp. 151–175. [Google Scholar]
  151. Al-Sadi, A.; Al-Moqbali, H.; Al-Yahyai, R.; Al-Said, F. AFLP data suggest a potential role for the low genetic diversity of acid lime (Citrus aurantifoliaSwingle) in Oman in the outbreak of witches’ broom disease of lime. Euphytica 2012, 188, 285–297. [Google Scholar] [CrossRef]
  152. Kettles, G.J.; Kanyuka, K. Dissecting the molecular interactions between wheat and the fungal pathogen Zymoseptoriatritici. Front. Plant Sci. 2016, 7, 508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Jouanin, A.; Gilissen, L.J.; Schaart, J.G.; Leigh, F.J.; Cockram, J.; Wallington, E.J.; Boyd, L.A.; Van Den Broeck, H.C.; Van der Meer, I.M.; America, A. CRISPR/Cas9 gene editing of gluten in wheat to reduce gluten content and exposure—reviewing methods to screen for coeliac safety. Front. Nutr. 2020, 7, 51. [Google Scholar] [CrossRef] [PubMed]
  154. Verma, A.K.; Mandal, S.; Tiwari, A.; Monachesi, C.; Catassi, G.N.; Srivastava, A.; Gatti, S.; Lionetti, E.; Catassi, C. Current status and perspectives on the application of CRISPR/Cas9 Gene-editing system to develop a low-gluten, non-transgenic wheat variety. Foods 2021, 10, 2351. [Google Scholar] [CrossRef]
  155. Macovei, A.; Sevilla, N.R.; Cantos, C.; Jonson, G.B.; Slamet-Loedin, I.; Čermák, 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 Biotech. J. 2018, 16, 1918–1927. [Google Scholar] [CrossRef] [Green Version]
  156. Sam, V.H.; Van, P.T.; Ha, N.T.; Ha, N.T.T.; Huong, P.T.T.; Hoi, P.X.; Phuong, N.D.; Le Quyen, C. Design and transfer of OsSWEET14-editing T-DNA construct to bac thom 7 rice cultivar. Acad. J. Biol. 2021, 43. [Google Scholar] [CrossRef]
  157. Wang, F.; Wang, C.; Liu, P.; Lei, C.; Hao, W.; Gao, Y.; Liu, Y.-G.; Zhao, K. Enhanced rice blast resistance by CRISPR/Cas9-targeted mutagenesis of the ERF transcription factor gene OsERF922. PLoS ONE 2016, 11, e0154027. [Google Scholar] [CrossRef]
  158. Zhou, J.; Peng, Z.; Long, J.; Sosso, D.; Liu, B.; Eom, J.S.; Huang, S.; Liu, S.; Vera Cruz, C.; Frommer, W.B. 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] [PubMed]
  159. Jia, H.; Zhang, Y.; Orbović, V.; Xu, J.; White, F.F.; Jones, J.B.; Wang, N. Genome editing of the disease susceptibility gene Cs LOB 1 in citrus confers resistance to citrus canker. Plant Biotechnol. J. 2017, 15, 817–823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Peng, A.; Chen, S.; Lei, T.; Xu, L.; He, Y.; Wu, L.; Yao, L.; Zou, X. Engineering canker-resistant plants through CRISPR/Cas9-targeted editing of the susceptibility gene Cs LOB 1 promoter in citrus. Plant Biotech. J. 2017, 15, 1509–1519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Zhang, Y.; Bai, Y.; Wu, G.; Zou, S.; Chen, Y.; Gao, C.; Tang, D. Simultaneous modification of three homoeologs of Ta EDR 1 by genome editing enhances powdery mildew resistance in wheat. Plant J. 2017, 91, 714–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Nekrasov, V.; Wang, C.; 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]
  163. 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] [PubMed] [Green Version]
  164. Gil-Humanes, J.; Wang, Y.; Liang, Z.; Shan, Q.; Ozuna, C.V.; Sánchez-León, S.; Baltes, N.J.; Starker, C.; Barro, F.; Gao, C. High-efficiency gene targeting in hexaploid wheat using DNA replicons and CRISPR/Cas9. Plant J. 2017, 89, 1251–1262. [Google Scholar] [CrossRef] [Green Version]
  165. 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] [Green Version]
  166. Ji, X.; Si, X.; Zhang, Y.; Zhang, H.; Zhang, F.; Gao, C. Conferring DNA virus resistance with high specificity in plants using virus-inducible genome-editing system. Genome Biol. 2018, 19, 197. [Google Scholar] [CrossRef] [Green Version]
  167. Zhang, T.; Zheng, Q.; Yi, X.; An, H.; Zhao, Y.; Ma, S.; Zhou, G. Establishing RNA virus resistance in plants by harnessing CRISPR immune system. Plant Biotech. J. 2018, 16, 1415–1423. [Google Scholar] [CrossRef] [Green Version]
  168. 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] [PubMed]
  169. Shah, P.R.; Varanavasiappan, S.; Kokiladevi, E.; Ramanathan, A.; Kumar, K. Genome editing of rice PFT1 gene to study its role in rice sheath blight disease resistance. Int. J. Curr. Microbiol. Appl. Sci. 2019, 8, 2356–2364. [Google Scholar] [CrossRef]
  170. Nagy, E.D.; Stevens, J.L.; Yu, N.; Hubmeier, C.S.; LaFaver, N.; Gillespie, M.; Gardunia, B.; Cheng, Q.; Johnson, S.; Vaughn, A.L. Novel disease resistance gene paralogs created by CRISPR/Cas9 in soy. Plant Cell Rep. 2021, 40, 1047–1058. [Google Scholar] [CrossRef] [PubMed]
  171. Ruyi, R.; Qiang, Z.; Futai, N.; Qiu, J.; Xiuqing, W.; Jicheng, W. Breeding for PVY resistance in tobacco LJ911 using CRISPR/Cas9 technology. Crop Breed. Appl. Biotech. 2021, 21, e31682116. [Google Scholar] [CrossRef]
  172. Zhang, Z.; Ge, X.; Luo, X.; Wang, P.; Fan, Q.; Hu, G.; Xiao, J.; Li, F.; Wu, J. Simultaneous editing of two copies of Gh14-3-3d confers enhanced transgene-clean plant defense against Verticillium dahliae in allotetraploid upland cotton. Front. Plant Sci. 2018, 9, 842. [Google Scholar] [CrossRef] [PubMed]
  173. Tashkandi, M.; Ali, Z.; Aljedaani, F.; Shami, A.; Mahfouz, M.M. Engineering resistance against Tomato yellow leaf curl virus via the CRISPR/Cas9 system in tomato. Plant Signal. Behav. 2018, 13, e1525996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kim, Y.-A.; Moon, H.; Park, C.-J. CRISPR/Cas9-targeted mutagenesis of Os8N3 in rice to confer resistance to Xanthomonas oryzae pv. oryzae. Rice 2019, 12, 1–13. [Google Scholar]
  175. Jiang, W.; Zhou, H.; Bi, 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]
  176. Ortigosa, A.; Gimenez-Ibanez, S.; Leonhardt, N.; Solano, R. Design of a bacterial speck resistant tomato by CRISPR/Cas9-mediated editing of Sl JAZ 2. Plant Biotech. J. 2019, 17, 665–673. [Google Scholar] [CrossRef] [Green Version]
  177. Sun, Q.; Lin, L.; Liu, D.; Wu, D.; Fang, Y.; Wu, J.; Wang, Y. CRISPR/Cas9-Mediated Multiplex Genome Editing of the BnWRKY11 and BnWRKY70 Genes in Brassica napus L. Int. J. Mol. Sci. 2018, 19, 2716. [Google Scholar] [CrossRef] [Green Version]
  178. Neequaye, M.; Stavnstrup, S.; Harwood, W.; Lawrenson, T.; Hundleby, P.; Irwin, J.; Troncoso-Rey, P.; Saha, S.; Traka, M.H.; Mithen, R. CRISPR-Cas9-mediated gene editing of MYB28 genes impair glucoraphanin accumulation of Brassica oleracea in the field. CRISPR J. 2021, 4, 416–426. [Google Scholar] [CrossRef]
  179. Gumtow, R.; Wu, D.; Uchida, J.; Tian, M. A Phytophthora palmivora extracellular cystatin-like protease inhibitor targets papain to contribute to virulence on papaya. Mol. Plant-Microbe Interact. 2018, 31, 363–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Razzaq, M.K.; Aleem, M.; Mansoor, S.; Khan, M.A.; Rauf, S.; Iqbal, S.; Siddique, K.H. Omics and CRISPR-Cas9 approaches for molecular insight, functional gene analysis, and stress tolerance development in crops. Int. J. Mol. Sci. 2021, 22, 1292. [Google Scholar] [CrossRef]
  181. Wang, L.; Chen, L.; Li, R.; Zhao, R.; Yang, M.; Sheng, J.; 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]
  182. Kim, D.; Alptekin, B.; Budak, H. CRISPR/Cas9 genome editing in wheat. Funct. Integr. Genom. 2018, 18, 31–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Joshi, R.K.; Bharat, S.S.; Mishra, R. Engineering drought tolerance in plants through CRISPR/Cas genome editing. 3 Biotech. 2020, 10, 400. [Google Scholar] [CrossRef] [PubMed]
  184. Shen, C.; Que, Z.; Xia, Y.; Tang, N.; Li, D.; He, R.; Cao, M. 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]
  185. 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] [PubMed] [Green Version]
  186. Shi, J.; Gao, H.; Wang, H.; Lafitte, H.R.; Archibald, R.L.; Yang, M.; Hakimi, S.M.; Mo, H.; Habben, J.E. ARGOS 8 variants generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant Biotech. J. 2017, 15, 207–216. [Google Scholar] [CrossRef] [Green Version]
  187. Liao, S.; Qin, X.; Luo, L.; Han, Y.; Wang, X.; Usman, B.; Nawaz, G.; Zhao, N.; Liu, Y.; Li, R. CRISPR/Cas9-induced mutagenesis of semi-rolled leaf1, 2 confers curled leaf phenotype and drought tolerance by influencing protein expression patterns and ros scavenging in rice (Oryza sativa L.). Agronomy 2019, 9, 728. [Google Scholar] [CrossRef] [Green Version]
  188. Zhang, A.; Liu, Y.; Wang, F.; Li, T.; Chen, Z.; Kong, D.; Bi, J.; Zhang, F.; Luo, X.; Wang, J. Enhanced rice salinity tolerance via CRISPR/Cas9-targeted mutagenesis of the OsRR22 gene. Mol. Breed. 2019, 39, 1–10. [Google Scholar] [CrossRef] [Green Version]
  189. de Melo, B.P.; Lourenço-Tessutti, I.T.; Paixão, J.F.R.; Noriega, D.D.; Silva, M.C.M.; de Almeida-Engler, J.; Fontes, E.P.B.; Grossi-de-Sa, M.F. Transcriptional modulation of AREB-1 by CRISPRa improves plant physiological performance under severe water deficit. Sci. Rep. 2020, 10, 16231. [Google Scholar] [CrossRef] [PubMed]
  190. Kuang, Y.; Li, S.; Ren, B.; Yan, F.; Spetz, C.; Li, X.; Zhou, X.; Zhou, H. Base-editing-mediated artificial evolution of OsALS1 in planta to develop novel herbicide-tolerant rice germplasms. Mol. Plant 2020, 13, 565–572. [Google Scholar] [CrossRef]
  191. Curtin, S.J.; Xiong, Y.; Michno, J.M.; Campbell, B.W.; Stec, A.O.; Čermák, T.; Starker, C.; Voytas, D.F.; Eamens, A.L.; Stupar, R.M. Crispr/cas9 and talen s generate heritable mutations for genes involved in small rna processing of glycine max and medicagotruncatula. Plant Biotech. J. 2018, 16, 1125–1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Corte, E.-D.; Mahmoud, L.M.; Moraes, T.S.; Mou, Z.; Grosser, J.W.; Dutt, M. Development of improved fruit, vegetable, and ornamental crops using the CRISPR/Cas9 genome editing technique. Plants 2019, 8, 601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Zhang, M.; Cao, Y.; Wang, Z.; Wang, Z.q.; Shi, J.; Liang, X.; Song, W.; Chen, Q.; Lai, J.; Jiang, C. A retrotransposon in an HKT1 family sodium transporter causes variation of leaf Na+ exclusion and salt tolerance in maize. New Phytol. 2018, 217, 1161–1176. [Google Scholar] [CrossRef] [Green Version]
  194. Korotkova, A.; Gerasimova, S.; Khlestkina, E. Current achievements in modifying crop genes using CRISPR/Cas system. VavilovskiiZh. Genet. Sel. 2019, 8, 14422. [Google Scholar] [CrossRef]
  195. Butler, N.M.; Baltes, N.J.; Voytas, D.F.; Douches, D.S. Geminivirus-mediated genome editing in potato (Solanum tuberosum L.) using sequence-specific nucleases. Front. Plant Sci. 2016, 7, 1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Zhao, Y.; Zhang, C.; Liu, W.; Gao, W.; Liu, C.; Song, G.; Li, W.-X.; Mao, L.; Chen, B.; Xu, Y. An alternative strategy for targeted gene replacement in plants using a dual-sgRNA/Cas9 design. Sci. Rep. 2016, 6, 1–11. [Google Scholar]
  197. Lu, Y.; Ye, X.; Guo, R.; Huang, J.; Wang, W.; Tang, J.; Tan, L.; Zhu, J.-K.; Chu, C.; Qian, Y. Genome-wide targeted mutagenesis in rice using the CRISPR/Cas9 system. Mol. Plant 2017, 10, 1242–1245. [Google Scholar] [CrossRef] [Green Version]
  198. Shalem, O.; Sanjana, N.E.; Hartenian, E.; Shi, X.; Scott, D.A.; Mikkelsen, T.S.; Heckl, D.; Ebert, B.L.; Root, D.E.; Doench, J.G. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 2014, 343, 84–87. [Google Scholar] [CrossRef] [Green Version]
  199. Jacobs, T.B.; Zhang, N.; Patel, D.; Martin, G.B. Generation of a collection of mutant tomato lines using pooled CRISPR libraries. Plant Physiol. 2017, 174, 2023–2037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Meng, X.; Yu, H.; Zhang, Y.; Zhuang, F.; Song, X.; Gao, S.; Gao, C.; Li, J. Construction of a genome-wide mutant library in rice using CRISPR/Cas9. Mol. Plant 2017, 10, 1238–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Henikoff, S.; Comai, L. Single-nucleotide mutations for plant functional genomics. Ann. Rev. Plant Biol. 2003, 54, 375–401. [Google Scholar] [CrossRef]
  202. 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. Biotech. 2017, 35, 441–443. [Google Scholar] [CrossRef] [PubMed]
  203. Chen, Y.; Wang, Z.; Ni, H.; Xu, Y.; Chen, Q.; Jiang, L. CRISPR/Cas9-mediated base-editing system efficiently generates gain-of-function mutations in Arabidopsis. Sci. China Life Sci. 2017, 60, 520–523. [Google Scholar] [CrossRef]
  204. Hua, K.; Tao, X.; Yuan, F.; Wang, D.; Zhu, J.-K. Precise A· T to G· C base editing in the rice genome. Mol. Plant 2018, 11, 627–630. [Google Scholar] [CrossRef] [Green Version]
  205. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157. [Google Scholar] [CrossRef]
  206. Li, H.; Li, J.; Chen, J.; Yan, L.; Xia, L. Precise modifications of both exogenous and endogenous genes in rice by prime editing. Mol. Plant 2020, 13, 671–674. [Google Scholar] [CrossRef]
  207. Tang, X.; Sretenovic, S.; Ren, Q.; Jia, X.; Li, M.; Fan, T.; Yin, D.; Xiang, S.; Guo, Y.; Liu, L. Plant prime editors enable precise gene editing in rice cells. Mol. Plant 2020, 13, 667–670. [Google Scholar] [CrossRef]
  208. Kim, S.; Kim, D.; Cho, S.W.; Kim, J.; Kim, J.-S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 2014, 24, 1012–1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Woo, J.W.; Kim, J.; Kwon, S.I.; Corvalán, C.; Cho, S.W.; Kim, H.; Kim, S.-G.; Kim, S.-T.; Choe, S.; Kim, J.-S. DNA-free genome editing in plants with preassembled CRISPR-Cas9 ribonucleoproteins. Nat. Biotech. 2015, 33, 1162–1164. [Google Scholar] [CrossRef]
  210. Yin, K.; Gao, C.; Qiu, J.-L. Progress and prospects in plant genome editing. Nat. Plants 2017, 3, 17107. [Google Scholar] [CrossRef] [PubMed]
  211. Wang, C.; Liu, Q.; Shen, Y.; Hua, Y.; Wang, J.; Lin, J.; Wu, M.; Sun, T.; Cheng, Z.; Mercier, R. Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat. Biotech. 2019, 37, 283–286. [Google Scholar] [CrossRef] [PubMed]
  212. Lyzenga, W.J.; Pozniak, C.J.; Kagale, S. Advanced domestication: Harnessing the precision of gene editing in crop breeding. Plant Biotech. J. 2021, 19, 660–670. [Google Scholar] [CrossRef] [PubMed]
  213. Zsögön, A.; Čermák, T.; Naves, E.R.; Notini, M.M.; Edel, K.H.; Weinl, S.; Freschi, L.; Voytas, D.F.; Kudla, J.; Peres, L.E.P. De novo domestication of wild tomato using genome editing. Nat. Biotech. 2018, 36, 1211–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Li, J.; Zhang, X.; Sun, Y.; Zhang, J.; Du, W.; Guo, X.; Li, S.; Zhao, Y.; Xia, L. Efficient allelic replacement in rice by gene editing: A case study of the NRT1. 1B gene. J. Integr. Plant Biol. 2018, 60, 536–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Callaway, E. CRISPR plants now subject to tough GM laws in European Union. Nature 2018, 560, 16–17. [Google Scholar] [CrossRef]
  216. Boettcher, M.; McManus, M.T. Choosing the right tool for the job: RNAi, TALEN, or CRISPR. Mol. Cell 2015, 58, 575–585. [Google Scholar] [CrossRef] [Green Version]
  217. Choudhary, E.; Lunge, A.; Agarwal, N. Strategies of genome editing in mycobacteria: Achievements and challenges. Tuberculosis 2016, 98, 132–138. [Google Scholar] [CrossRef]
  218. Karlson, C.K.S.; Noor, S.N.M.; Nolte, N.; Tan, B.C. CRISPR/dCas9-based systems: Mechanisms and applications in plant sciences. Plants 2021, 10, 2055. [Google Scholar] [CrossRef] [PubMed]
  219. Zhang, X.-H.; Tee, L.Y.; Wang, X.-G.; Huang, Q.-S.; Yang, S.-H. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol. Ther. Nucleic Acids 2015, 4, e264. [Google Scholar] [CrossRef] [PubMed]
  220. Hua, K.; Tao, X.; Han, P.; Wang, R.; Zhu, J.-K. Genome engineering in rice using Cas9 variants that recognize NG PAM sequences. Mol. Plant 2019, 12, 1003–1014. [Google Scholar] [CrossRef] [PubMed]
  221. Naeem, M.; Majeed, S.; Hoque, M.Z.; Ahmad, I.J.C. Latest developed strategies to minimize the off-target effects in CRISPR-Cas-mediated genome editing. Cells 2020, 9, 1608. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Application of genome editing in crops improvement. CRISPR/Cas9 is used for genetic improvement, increasing nutrients use efficiency, biomass production, and increasing disease resistance.
Figure 1. Application of genome editing in crops improvement. CRISPR/Cas9 is used for genetic improvement, increasing nutrients use efficiency, biomass production, and increasing disease resistance.
Cimb 43 00135 g001
Figure 2. A comparison of genome editing tools. Comparison of these tools shows their efficiency towards genome editing and also shows their limitations. Hence CRISPR/Cas9 technique is more accurate than other tools.
Figure 2. A comparison of genome editing tools. Comparison of these tools shows their efficiency towards genome editing and also shows their limitations. Hence CRISPR/Cas9 technique is more accurate than other tools.
Cimb 43 00135 g002
Figure 3. Mechanism of genome editing using Cas9. Cas9 gene-editing system involves complex of sgRNA and Cas9 protein, unwinding of DNA by sgRNA, cutting of gene by Cas9, use of analysis tools, cloning, and transformation, etc. This process needs no foreign element for editing.
Figure 3. Mechanism of genome editing using Cas9. Cas9 gene-editing system involves complex of sgRNA and Cas9 protein, unwinding of DNA by sgRNA, cutting of gene by Cas9, use of analysis tools, cloning, and transformation, etc. This process needs no foreign element for editing.
Cimb 43 00135 g003
Figure 4. Novel strategies to improve genome edit using Cas9. The editing efficiency of Cas9 could be enhanced by an efficient screening of targeted characters, exploration of genetic material via the knockout of genes, and the use of an ideal gene transformation system.
Figure 4. Novel strategies to improve genome edit using Cas9. The editing efficiency of Cas9 could be enhanced by an efficient screening of targeted characters, exploration of genetic material via the knockout of genes, and the use of an ideal gene transformation system.
Cimb 43 00135 g004
Figure 5. Challenges of CRISPR/Cas9 gene editing. Cas9 has low efficiency of transformation methods, lack of efficiency of the delivery system, and difficultyin achieving an equal molar ratio of Cas9 and sgRNA. These challenges hinder its use for further crops improvement on a larger scale.
Figure 5. Challenges of CRISPR/Cas9 gene editing. Cas9 has low efficiency of transformation methods, lack of efficiency of the delivery system, and difficultyin achieving an equal molar ratio of Cas9 and sgRNA. These challenges hinder its use for further crops improvement on a larger scale.
Cimb 43 00135 g005
Table 1. Use of MNs, ZFNs, and TALENs for crops improvement.
Table 1. Use of MNs, ZFNs, and TALENs for crops improvement.
CropToolGeneTraitReference
RiceTALENOsBADH2Fragment rice[48]
RyeCas9TrpEFungal resistance[49]
WheatTALENTaMLOResistance to powdery resistance[50]
MaizeMNsLGITargeted mutagenesis[51]
CottonEMNsEPSPSTolerance to herbicide[38]
TobaccoTALENsSur ADirected mutation[52]
BarleyTALENsTransgeneResistance to powdery resistance[53]
MaizeTALENsZMIPKPhytic acid biosynthesis Liang[54]
PotatoTALENsStGBSSQuality of tuber starch[55]
Table 3. RNA tools developed for CRISPR/Cas9.
Table 3. RNA tools developed for CRISPR/Cas9.
ToolFunctionYearReference
tracr RNAAct as reference point2021[76]
CRISPR designRNA construction for targeted regions and assessment of off target effects2013[77]
Cas12jGenome manipulation2021[78]
sgRNAcas9Speedy construction of sgRNA and fewer effects2014[79]
CCTopEstimate target sgRNA sequence on the basis of off-target influences2015[80]
phytoCRISP.ExCas9 target prediction2016[81]
Cas12Target recognition2021[82]
sgRNA Scorer 2.0sgRNA construction for many PAM locates2017[83]
CRISPR-LocalsgRNA construction for non-reference types2018[84]
CRISPRIncConstruct sgRNA for lncRNAs2019[85]
Table 4. Uses of CRISPR/Cas9 for quality and yield enhancement in crops.
Table 4. Uses of CRISPR/Cas9 for quality and yield enhancement in crops.
GeneCropsTraitTechniqueReference
CLEMaizeGrain yieldCRISPR/Cas9[131]
SLIAA9TomatoSeedless fruitCRIISPR/Cas9[132]
OsTB1RiceHigh grain yieldCRIISPR/Cas9[114]
GmFT2aSoybeanDelay in flowering timeCRIISPR/Cas9[133]
BnITPKRapeseedLow phytic acidCRIISPR/Cas9[134]
Bolc.GA4.aCabbageDwarf plantCRIISPR/Cas9[106]
OsAAP10RiceGood cooking qualityCRIISPR/Cas9[114]
RVE7ChickpeaGrain qualityCRIISPR/Cas9 B[135]
BnaMAXIRapeseedYieldCRISPR/Cas9[136]
Wholek1geneSorghumIncrease lysine contentCRIISPR/Cas9[137]
TaSBEIIaWheatHigh amylose contentCRIISPR/Cas9[116]
GBSSPotatoEnhance amylose contentCRIISPR/Cas9[138]
P450RiceHigh grain yieldCRIISPR/Cas9[113]
IPARiceEnhanced yieldCRIISPR/Cas9[139]
CIPDSWatermelonAlbino phenotypeCRIISPR/Cas9[140]
GS3RiceGrain yieldCRIISPR/Cas9[115]
ANTIRiceFruit colorCRIISPR/Cas9[141]
OsSPL16RiceGrain yieldCRIISPR/Cas9[108]
lncRNA1459TomatoProlong shelf lifeCRIISPR/Cas9[142]
PRLMaizeReduction in zein contentCRIISPR/Cas9[143]
GASR7WheatGrain weightCRIISPR/Cas9[144]
PDSBananaAlbino phenotypeCRIISPR/Cas9[145]
CsFAD2CamelinaOleic acidCRIISPR/Cas9[146]
FaTM6StrawberryFlower developmentCRIISPR/Cas9[147]
GmFATB1SoybeanLow saturated fatty acidCRIISPR/Cas9[148]
Table 5. A list of diseases resistant crops varieties made by using CRISPR/Cas9.
Table 5. A list of diseases resistant crops varieties made by using CRISPR/Cas9.
GeneChromosomal
Position
LocusPathogenCropFunctionTraitRepair ToolEditing ResultsReference
SIPeLo and SIMIO1 TomatoEnhance resistance to leaf curl virus Enhance resistance to leaf curl virus [168]
OsSWEET14 RiceBacterial leaf blightBacterial leaf blight resistance [156]
CsLOB1N/AN/AXanthomonas citriCitrusSusceptibility to citrus cankerResistance to citrus cankerNHEJKnockout[159]
Gh14-3-3dhttps://www.ncbi.nlm.nih.gov/nuccore/164652939 (accessed on 15 October 2020)N/AVerticillium dahliaeCottonNegative controller of resistance to diseaseVerticillium wilt resistanceNHEJKnock-in[172]
Ntab0942120 TobaccoResistance to potato virus YResistance to potato virus Y [171]
eLF4Gchr07:22114961..22123061 (+ strand)Os07g0555200Tungro spherical virusRiceStarting factor for initiationResistance to tungro spherical virusNHEJKnock out[155]
Rpsl SoybeanDiseases resistanceDiseases resistance [170]
Rp and Cp sequencesN/AN/AYellow leaf curl virusTomatoNegative controller of resistanceEnhanced resistance again leaf curl virusNHEJKnock out[173]
OsSWEET11chr08:26725952..26728794Os08g0535200Bacterial blightRiceResistance to bacterial blightResistance to bacterial blightNHEJKnock out[174]
OsSWEET14chr11:18171707..18174478Os11g0508600Bacterial blightRiceResistance to bacterial blightResistance to bacterial blightNHEJKnock out[175]
SiMLOlN/AN/A TomatoPowdery mildew resistance geneResistance to powdery mildewNHEJKnock out[162]
Jaz2Chr12: 2502581..2504643N/APseudomonas syringaeTomatoBacterial speck resistanceBacterial speck resistanceNHEJKnock out[176]
WRKY70LK032201:212360-212875BnaA09g35840D Brassica Resistance to pathogensNHEJKnock out[177]
MYB28 BrassicaGlucoraphanin accumulation NHEJKnock out[178]
S-genesN/AN/A PotatoResistance to PhytophthoraResistance to PhytophthoraNHEJKnock out[138]
TaMLO-A1Chromosome 4A: 519,570,414-519,575,284TraesCS4D02G318600 WheatResistance to mildewResistance to mildewNHEJKnock out[50]
RGA2, Ced93:6700445-6700466GSMUA_Achr3G09290_001Fusarium oxysporumBananaResistance to fusarium wiltResistance to fusarium wiltNHEJKnock out[162]
Mlo-7Chr13: (5335059..5339258VIT_00016304001 GrapeResistance to mildewResistance to mildewNHEJKnock out[124]
PpalEPIC8N/AN/APhytophthora palmivoraPapayaResistance to PhytophthoraResistance to PhytophthoraNHEJKnock out[179]
Table 6. Development of climate smart crops using CRISPR/Cas9.
Table 6. Development of climate smart crops using CRISPR/Cas9.
GeneChromosomal PositionLocusCropTraitsRepair
Pathway
Editing
Results
References
OsMYB30 RiceCold tolerance [115]
slmapk3 TomatoDrought tolerance [181]
OsALS1 RiceHerbicide tolerance [190]
4CL, RVE7 ChickpeaDrought tolerance [135]
SAPK2chr07:25717837..25722009 (+strand)Os07g0622000RiceTolerance to salinity and droughtNHEJKnockout[185]
TaDREB37D:27470774-27471448TraesCS7D02G052600WheatTolerance to droughtNHEJKnockout[182]
NPRI3:3740543-3741413Solyc03g026270.2TomatoTolerance to cold and drought stressNHEJKnockout[142,192]
ZmHKTIN/AN/AMaizeTolerance to salinity stressNHEJKnockout[193]
Drb2a12:5797459-5798459,
11:11249371-11250406
GLYMA_12G075700
GLYMA_11G145900
SoyabeanTolerance to drought and salinity stressNHEJKnockout[191]
OsRR22, OsPDSchr10:17076098..17081344 (- strand) chr03:4410090..4414779 (+ strand)Os10g0463400
Os03g0184000 (OsPDS)
RiceTolerance to salinity stressNHEJKnockout[188,194]
OsAOX1a,chr04:30287197..30289860Os04g0600200RiceDrought resistanceNHEJKnockout[104]
OsBADH2chr08:20379823..20385975Os08g0424500RiceAbiotic stress resistanceHDRKnockout[91]
ALS1chr3:8175606-8177917PGSC0003DMG400034102PotatoResistance to herbicideHDRKnockout[195]
MIR169aChr03:4358994-4359219AT3G13405ArabidopsisDrought resistanceHDRKnockout[196]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rasheed, A.; Gill, R.A.; Hassan, M.U.; Mahmood, A.; Qari, S.; Zaman, Q.U.; Ilyas, M.; Aamer, M.; Batool, M.; Li, H.; et al. A Critical Review: Recent Advancements in the Use of CRISPR/Cas9 Technology to Enhance Crops and Alleviate Global Food Crises. Curr. Issues Mol. Biol. 2021, 43, 1950-1976. https://doi.org/10.3390/cimb43030135

AMA Style

Rasheed A, Gill RA, Hassan MU, Mahmood A, Qari S, Zaman QU, Ilyas M, Aamer M, Batool M, Li H, et al. A Critical Review: Recent Advancements in the Use of CRISPR/Cas9 Technology to Enhance Crops and Alleviate Global Food Crises. Current Issues in Molecular Biology. 2021; 43(3):1950-1976. https://doi.org/10.3390/cimb43030135

Chicago/Turabian Style

Rasheed, Adnan, Rafaqat Ali Gill, Muhammad Umair Hassan, Athar Mahmood, Sameer Qari, Qamar U. Zaman, Muhammad Ilyas, Muhammad Aamer, Maria Batool, Huijie Li, and et al. 2021. "A Critical Review: Recent Advancements in the Use of CRISPR/Cas9 Technology to Enhance Crops and Alleviate Global Food Crises" Current Issues in Molecular Biology 43, no. 3: 1950-1976. https://doi.org/10.3390/cimb43030135

Article Metrics

Back to TopTop