Publication Cover
GM Crops & Food
Biotechnology in Agriculture and the Food Chain
Volume 14, 2023 - Issue 1
3,298
Views
4
CrossRef citations to date
0
Altmetric
Review

Putting CRISPR-Cas system in action: a golden window for efficient and precise genome editing for crop improvement

, , , , , ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1-27 | Received 11 Jan 2023, Accepted 24 May 2023, Published online: 08 Jun 2023

ABSTRACT

The daunting task of feeding an ever-growing population is an immense challenge for the contemporary scientific community, especially in view of the rapidly changing climate throughout the world. Amidst these threatening crises, we witness rapid development in genome editing (GE) technologies, revolutionizing the field of applied genomics and molecular breeding. Various GE tools have been developed during the last two decades, but the CRISPR/Cas system has most recently made a significant impact on crop improvement. The major breakthroughs of this versatile toolbox are genomic modifications like single base-substitutions, multiplex GE, gene regulation, screening mutagenesis, and enhancing the breeding of wild crop plants. Previously, this toolbox was used to modify genes related to significant traits such as biotic/abiotic resistance/tolerance, post-harvest traits, nutritional regulation, and to address self-incompatibility analysis-related challenges. In the present review, we have demonstrated the functional dynamics of CRISPR-based GE and its applicability in targeting genes to accomplish novel editing of crops. The compiled knowledge will provide a solid foundation for highlighting the primary source for applying CRISPR/Cas as a toolbox for enhancing crops, to achieve food and nutritional security.

1. Introduction

The need for technical advancements to feed the growing world population is inevitable, and plant scientists constantly deal with this challenge. The outstanding contribution of the Green revolution and new technical advances in plant breeding efforts are underway to overcome the challenge to achieve global food security under changing climate conditions.Citation1–6 Since ancient times, humans have improved crop plants by transferring novel traits from crossable relatives. The sole motive of this practice was to transfer desirable gene variants even though the process is time-consuming as it takes several years to transfer alleles of interest to desirable genotype. With the advancements, breeders learned to use chemical mutagens or radiations to generate mutants with the desired modifications in plants.Citation7 The yield and quality of crops have exceptionally increased by using spontaneous and induced mutations and transgenic methods.Citation8–16 Despite the availability of appropriate germplasm for crop improvement, these mutations have proven to be time-consuming and labor-intensive due to their low frequency and unpredictability.Citation7 The last decade witnessed a revolution in crop improvements with the introduction of various genome-editing tools.Citation17,Citation18 Previous reports have demonstrated that the effective strategies for targeted mutagenesis at the target loci can be customized using sequence-specific nucleases (SSNs), which generate double-strand breaks (DSBs) in DNA.Citation19,Citation20

The major drawback of transgenic crops lies in the tedious risk assessment and cost that may lead to less production and the application of traditional transgenic approaches. Further integration of foreign genes through illegitimate recombination may lead to sequence changes, gene disruption, random transgene integration, and production of irrelevant mutant proteins.Citation21,Citation22 In contrast, GE tools such as, CRISPR-Cas9-based GE, are the most amendable tools for the desirable editing of genomes in diverse range of plant species. Qualities such as, high efficacy, low off-target effects, simplicity in operation, cost effectiveness, and multiplex gene editing make them the most dominantly employed tools for plant modifications. In comparison to traditional transgenic approaches, the CRISPR/Cas9 system is incommensurable in the development of sustainable agriculture.Citation15,Citation23–27

Advanced GE tools such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR-Cas systems are employed to accomplish mutagenic effects in an organism, for inducing desirable changes in a predictable manner.Citation15,Citation17,Citation28–30 These techniques generate double-stranded breaks (DSBs) in the target DNA and induce deletions or insertions of nucleotides by the endogenous repair system via non-homologous end joining (NHEJ) or precise gene insertion or gene replacement by homologous recombination (HR), resulting in a loss-of-function or gain-of-function of the target genes.Citation31 For instance, to attain these changes in the target genome, earlier, scientists applied GE tools such as, ZFN and TALEN; both rely on artificial restriction enzymes that exert cleavage functions by the combination of the DNA- binding domains (DBDs) of ZFN and TALENs with the cleavage domains of Fok1.Citation32–34 Thus, through the protein-DNA interactions, both ZFN and TALEN identify their targeted sequences. Even though effective mutagenesis of ZFN and TALEN have been reported in many plant species, the difficult and prolonged DNA assembly procedure of ZFN/TALEN DNA- binding component has been challenging for scientists, consequently hindering their effective applications.Citation35,Citation36

In recent years, the advancements and developments in engineered CRISPR-Cas9 system has revolutionized targeted GE in plants and animals as it can recognize the target sites only via a single-guide RNA (sgRNA), unlike ZFN and TALEN.Citation37,Citation38 With the introduction of the CRISPR-toolbox, scientists are now better able to comprehend and read genomic structure better than ever before as well as edit it with previously unreachable precision.Citation39 The construction of the Cas9 RNA-guided engineered nuclease (RGEN) system is far more simple and easy, versatile, specific, and faster than assembling numerous ZFNs or TALENs for GE.Citation40,Citation41 Currently, the CRISPR/Cas9 system has emerged as a powerful tool for GE in a large variety of plant species, right from the model plant Arabidopsis and rice to other crop plants such as tetraploid potato, maize, hexaploid bread wheat, allotetraploid cotton, and tetraploid durum wheat.Citation35,Citation42–46 In addition, the CRISPR/Cas9 system was employed to target several homologous sites in Brassica napus genes, such as BnaRGA, BnaDA2, BnALC, BnJAG, BnSHP, BnCLV, and BnaFUL.Citation47–50 Even though employed in a wide range of plants, it still has not developed an easy and quick tool to generate first-generation mutated plants, which are important to gene functionalization and screening of mutations in a later generation.

Overall, the engineered CRISPR/Cas9 system involves two components, Cas9 protein expression cassettes and sgRNA. Several modifications mediated by CRISPR/Cas system include single base substitution, multiplex gene editing, regulation of gene transcription, and enhancing, screening mutagenesis, and enhancing the breeding of wild crop plants to enhance resilience to biotic as well abiotic stresses ().Citation27,Citation38,Citation51 The current review is aimed at comprehensively demonstrating the versatility of the CRISPR/Cas-based GE systems to accomplish efficient crop improvement for desirable traits to enhance the productivity, stress tolerance, and nutritional profile of crop plants. The following sections in this review will highlight the various mechanisms that CRISPR-based GE techniques work and the variety of approaches that they can be used to change plant genomes for the benefit of humanity’s existence. In addition, we have also critically mentioned the biosafety concerns over the dissemination of crops engineered through CRISPR-based tools.

Figure 1. Illustration for developing superior and elite crop plants resilient to biotic and abiotic stress factors, and enhancing the quality of crops by using CRISPR/Cas systems through single base substitution, multiplex gene editing, gene knockout, regulation of gene transcription, and enhancing and screening mutagenesis.

Figure 1. Illustration for developing superior and elite crop plants resilient to biotic and abiotic stress factors, and enhancing the quality of crops by using CRISPR/Cas systems through single base substitution, multiplex gene editing, gene knockout, regulation of gene transcription, and enhancing and screening mutagenesis.

2. Frontline Approach to Crop Improvement-CRISPR-Cas System-Construction and Mechanistic Approach

The development and advancements in the CRISPR/Cas GE tools have allowed them to emerge as crucial tools in biological research to understand the function of genes and in crop improvement.Citation52 The different classes of GE agents that are presently available for editing the genome include – nucleases, base editors, and prime editors. Each tool has its own simplicity, versatility, and limitations, and the major efforts and improvements in the editing capabilities of the tools have broadened and extended their targeting as well as enhanced specific editing abilities. The suitable class of editing agents used in experimental systems depends on the type of editing that is desired. The common editings yielded by CRISPR/Cas include (i) point mutations, that is, conversion of DNA base pairs, (ii) insertion of DNA base pairs, (iii) deletion of DNA base pairs or (iv) a combination of the above changes (including replacement of DNA base pairs).

2.1. CRISPR/Cas Nucleases: A Technical Overview

Naturally, by binding, CRISPR-Cas systems are used by bacteria and archaea as a part of their adaptive immunity to protect the host from foreign pathogens and to cleave foreign nucleic acids.Citation53 The mechanistic chemistry of CRISPR/Cas systems in practice is reprogrammed readily in order to target specific genomic sites by simply using the spacer sequences within a guide RNA molecule and by providing the similar target DNA protospacer sequence that is placed adjacent to a protospacer-adjacent motif (PAM), generating nonhomologous end joining (NHEJ) or homology-directed repair (HDR) ().Citation29,Citation54,Citation55 Furthermore, there are two main groups of naturally occurring CRISPR/Cas immune systems, viz., class-I and class-II. As far as class-1 is concerned, it uses the multiprotein complexes to cleave nucleic acids and is very rarely used for editing genomes. Whereas, class-II employs only one protein effector domain to cleave the nucleic acids. Due to this advantage, class-II is the most widely used CRISPR tool for biological research and translation applications.Citation56,Citation57 Class-II has been further divided into the three types, II, V, and VI; each of these types functions by employing different sets of Cas proteins. Among these, the Cas 9 variants from type – II and Cas 12 variants from type-V have the RNA-guided DNA endonuclease activity whereas Cas 13 of type VI possesses the targeting activity of RNA and the cleavage activity.

Figure 2. Schematics show the mechanistic workflow of the CRISPR/Cas system and its precise catalysis by inducing editing through NHEJ or HDR, employed to improve crops.

Figure 2. Schematics show the mechanistic workflow of the CRISPR/Cas system and its precise catalysis by inducing editing through NHEJ or HDR, employed to improve crops.

2.1.1. Cas9 Nucleases

The Cas9 protein, RNA-guided endonucleases, forms a part of the type-II CRISPR systems can generate DSBs in the target DNA.Citation58 The Cas9 nucleases and the CRISPR RNAs (crRNA) form the ribonucleoprotein complex to mediate its action on the target DNA sequence.Citation59 The Cas-9 GE uses the synthetic sgRNAs, a fusion product of crRNA and trans-activating CRISPR RNA (tracrRNA) molecules.Citation29 The Cas9 nuclease primarily makes the blunt-ended DSBs at the 3 bp upstream of the PAM sequences such as, 5′-NNGRR(N)-3´Citation60 and, in some cases, the Cas9, where alternative cutting patterns have been observed.Citation61,Citation62 The Cas9-guide RNA ribonucleoprotein complex recognizes the similar PAM sequences and induces unwinding of the target dsDNA to form RNA-DNA heteroduplex.Citation58,Citation63 Further, the guide RNA spacer displaces DNA at the nontarget sites, leading to the formation of the single-stranded “R-loop,” which is available to other molecules for interaction.Citation64–66 Subsequently, the formation of the R-loop and the conformation of Cas9 changes result in the activation of its nuclease domains.Citation29,Citation64,Citation66,Citation67 The mismatches formed between the guide RNA spacer and the target strand hinders the conformational changes in the Cas9 protein complex, thus restricting its activation.Citation68,Citation69 Lastly, after the nuclease activation, two distinct nuclease domains of Cas9 hydrolyze the phosphodiester backbone of DNA. Subsequently, RuvC nuclease domain cleaves the PAM-containing nontarget DNA strand and the HNH nuclease domain induces cleavage in the guide RNA-bound target DNA strand.Citation70,Citation71 Moreover, it is reported that the mutation induced in either of nuclease domains generates a Cas9 nickase, which is capable of cleaving only one DNA strand. On the other hand, inactivation of both the nuclease domains generates a catalytically dead Cas9 (dCas9).Citation23,Citation72,Citation73 Ever since the reports of programmed DNA cleavage by Cas9 nuclease from Streptococcus pyogenes (SpCas9) in vitroCitation70 and in mammalian cells,Citation23,Citation74–76 the Cas9 variants discovered and used for the GE orthologs are derived from microorganisms such as, Staphylococcus aureus,Citation77 Streptococcus thermophiles,Citation23,Citation75,Citation78 Neisseria meningitidis,Citation78–80 Campylobacter jejuni,Citation81 and other reported organisms.Citation82

2.1.2. Cas12 Nucleases

The Cas12 nucleases include numerous variations from the type-V CRISPR/Cas system.Citation83 They attain a single RuvC-like domain that induces cleavage in the target DNA in both strands. The first Cas12 nuclease widely used for GE applications is Cas12a, also known as Cpf1.Citation84 Cas12a facilitates multiplexed gene editing as it can use a dedicated RNase domain to process its single crRNAs.Citation85 Several Cas12 variants, including Cas12a, follow target site recognition and subsequent activation as well as cleavage of ssDNA or RNA.Citation86,Citation87 Although most Cas12 variants target and cleave DNA, other Cas12 effectors, such as Cas12g, are RNA-guided RNA-cleaving enzymes that exhibit both collateral RNase and DNase activity, upon activation.Citation86 Different from Cas9, the Cas12a identifies and recognizes the T-rich PAM sequence, resulting in the maturation of their own guide RNA and generates a staggered 5′ and 3′ ends breaks in PAM distal dsDNA.Citation84 For instance, resistance to high temperature was obtained by using Cas12b for editing GhCLA gene in the genome of cotton (Gossypium hirsutum).Citation88 Similarly, the LbCpf1 genome editing tool was used to induce DSBs for replacing wild type ALS with mutated ALS for obtaining herbicide resistance in japonica rice (Oryza sp.).Citation89 In rice, LbCas12a was used to mediate site-specific mutation in the Xa13 gene to develop resistance to bacterial leaf blight.Citation90 The abovementioned studies clearly indicate the versatile nature of the Cas12-based GE systems have great potential in improving the resilience of crop plants.

2.2. The Overview of Genome Editing (GE) of Specific Genomic Regions

2.2.1. Base Editors-A Narrow Window for CRISPR/Cas Based GE

Improved crop cultivars can be developed through CRISPR/Cas9-mediated mutagenesis to address growing food needs. In particular, single base editing through CRISPR/Cas9 could result in elite trait variants that hasten crop improvement programs. The base editing technology is an alternative genome-diting tool that generates point mutations precisely without making DSBs in the target region of genomes (). As an alternative, the Cas9 nickase (nCas9) is fused with the base editor and the guide RNA to target a specific DNA sequence. To date, two primary base editing tools have been reported, viz., cytosine base editors (CBEs) and adenine base editors (ABE).Citation91,Citation92 For instance, the nCas9, a CBEs causes a mutation in D10A, resulting in the inactivation of the RuvC domain. A wide range of crop genomes such as, rice, maize (Zea mays), wheat (Triticum aestivum), and potato (Solanum tuberosum) has been precisely edited by the CBE-mediated base editing technology to obtain the desired results.Citation91,Citation93

Figure 3. Ways toward precise editing: The prime editing mechanism involves the formation of a complex that comprises pegRNA to yield single-strand nick at a precise position in the target DNA PAM sequences, followed by the polymerization of newly edited DNA by reverse transcriptase by using pegRNA as a template. Subsequently, the sgRNA is employed to form the nick on the non-modified DNA strand to initiate repair-based insertion of editing in both the DNA strands. Base editied nCas9 is the preferred nuclease employed in dbase editing to generate single-stranded nick in the target DNA stranded, aided by gRNA using base deaminase to accomplish a single or more preferred nucleotide base exchanges.

Figure 3. Ways toward precise editing: The prime editing mechanism involves the formation of a complex that comprises pegRNA to yield single-strand nick at a precise position in the target DNA PAM sequences, followed by the polymerization of newly edited DNA by reverse transcriptase by using pegRNA as a template. Subsequently, the sgRNA is employed to form the nick on the non-modified DNA strand to initiate repair-based insertion of editing in both the DNA strands. Base editied nCas9 is the preferred nuclease employed in dbase editing to generate single-stranded nick in the target DNA stranded, aided by gRNA using base deaminase to accomplish a single or more preferred nucleotide base exchanges.

Adenine base editing is another potential CRISPR system that converts A-T to G-C. The adenosine deaminase activity of the enzyme helps in deaminating adenosine to produce inosine, which can then be paired with cytidine. The deaminated nucleotide is identified as guanine by the DNA polymerase during the process of DNA replication and repair mechanism.Citation94 The ABE systems work well in A. thaliana,Citation95 rice, wheat, and rapeseed (Brassica napus),Citation96 but showed lower efficiency in other organisms than its original counterpart, i.e., SpCas9 or SaCas9.Citation92,Citation97 Recently, several ABE variants have been developed for adenine base editing in mammalian cells, which may be useful for efficient editing in plants.Citation98,Citation99

2.2.2. Prime Editing-Extending the Dimensions of CRISPR/Cas Based GE

Prime editing in plants is a relatively new technique; yet, it has enormous potential for a wide range of plant gene editing applications. The editing system consists of nCas9 (H840A) with reverse transcriptase (RTase) and a prime editing guide RNA (pegRNA) (Figure-3). The process of prime editing allows for at least 12 kinds of base conversions in specific target genes at sites ranging from 3 bp upstream up to 29 downstream of the PAM sequence.Citation100 Two binding sites are critical to the recognition of pegRNA, associated with nCas9 (H840A), a spacer sequence by its 5` region, and a prime binding site (PBS) through 3` regions to edit the target DNA sequence. The PBS region acts as a primer for nCas9-linked RTase and the pegRNA is used as a template by RTase; it directly copies the genetic information from pegRNA to be incorporated into the target genome.Citation100 After the completion of the reverse transcription, the 5` and 3` unedited flaps equilibrate, trailed by the integration of the modified DNA into the target sequence in the genomic DNA through DNA repair system and ligation reactions.Citation100 Recently, prime editing was applied in wheat, rice, maize, and potato.Citation101 Consequently, it is pertinent that more reports displaying the wide applicability of prime editing GE system in crop plants are needed to accelerate the progress of this technology in the improvement of crop plants.

3. Crispr/cas-Widening the Editing Frame: From Single Gene Editing to Multiplex GE

In living systems, cellular processes are grossly controlled by a wide array of redundant genes. Consequently, mutating a gene may influence the desired phenotypes of an organism, even though in few cases, the mutated gene may be compensated by other genes of a similar family. Hence, it is pertinent to upgrade the functional diversity of GE systems to multiplex gene-editing systems for widening its applications. Against this motive, scientists have developed a CRISPR/Cas9-mediated multiplex GE system where several sgRNA cassettes are controlled by a single of multiple promoters loaded in a single vector.Citation43,Citation102,Citation103 Furthermore, efficient stacking of multiple traits in elite plant germplasm has been made achievable by multiplex genome editing, and this has a substantial impact on enhancing the complex agronomic attributes. For instance, two sgRNAs were designed to target two homologous of magnesium-chelatase subunit I (CHLI) that have a critical function in the mechanism of photosynthesis and subsequently, the disruption in both the genes resulted in albino phenotypes in Arabidopsis.Citation32 The application of multiplex GE was also validated through the disruption of four subunits of katanin p80 by using three sgRNAs, producing a dwarf phenotype of A. thaliana.Citation104 Tang and colleagues used a multiplexing system based on multiple sgRNAs with Cas9 proteins encoded as a single transcript for GE of various genes in plants.Citation105 Besides, for carrying multiplex GE, the CRISPR-Cpf1 system was widely used in crop plants.Citation106 Li et al. displayed the application of multiplex gene editing through the application of CRISPR/Cas9 by concurrent editing of 08 target sites in wheat genome.Citation103 For instance, the construct consisted of a single promotor controlling numerous repeated units of crRNA, recognized by Cpf1, to produce cleavage at the specific target sites.Citation106 These reports strongly suggest that multiplex gene editing by CRISPR systems are expedient tools for targeting multiple genes at once to reduce time cost in knowing the functions of the desired genes/gene families.

4. Generating High-Throughput Plant Mutant Libraries Using CRISPR Platform

Another way that CRISPR technology dominates the current trends of genomic research is by developing genome-wide high-speed screening approaches that can thoroughly analyze the influence of regulatory sequences or specific genes on the phenotype of interest.Citation107 Traditional plant mutant libraries relying on random mutations and generated through irradiation, ethyl methane sulfonate (EMS) mutagenesis, T-DNA insertions, and transposons require many generations to express loss-of-function mutants. CRISPR/Cas-based whole-genome-scale expression libraries can be constructed, according to computation, for extensive functional genomics and genetic improvement. By delivering the gRNA library, together with Cas9, into cells would generate a cell population to knock out each gene in the genome.Citation108 Recently, two independent research groups designed large scale CRISPR/Cas9-based knockout mutant libraries that covered most of the genes in rice plants.Citation109,Citation110 Lu and colleagues generated 90,000 transgenic rice lines by targeting 34,234 genes, while a single generation of homozygous mutants was obtained.Citation109 Similarly, 13000 genes expressed in rice shoot tissue were targeted, resulting in 14,000 independent T0 lines.Citation110,Citation111 Recently, Jacobs and colleagues constructed mutant expression libraries of leucine-rich repeat genes of subfamily ΧΙΙ (immunity-associated genes) in the tomato plant.Citation111

Recently, Liu, JianCitation112 used integrated CRISPR/Cas9-based high-throughput, targeted mutagenesis to target 743 candidate genes in maize, governing agronomic and nutritional traits.Citation112 It has been demonstrated that incorporating forward and reverse genetics through a focused mutagenesis library ensures that the essential agronomic genes with complex genomes are easily validated. They first designed an improved high-throughput GE platform in maize to target >1,000 maize candidate genes, potentially governing different agronomic traits, which could be applicable to other species with genomic complexity. Furthermore, to validate the phenotypes associated with genetic traits, this targeted mutagenesis workflow can be used as an unprecedented tool for dissecting classical genomic intervals and identifying novel genes, phenotypic modifications, and addressing the widely recognized gene redundancy issue in identifying gene function in plants. The knowledge-based targeted mutagenesis is certainly an effective and high-throughput approach to functional gene identification. This method can be exploited in other crops as well. Nevertheless, large-scale screens with a sgRNA library are eventually accessible to the plant community. Future advances in the high-throughput phenotyping approach, together with the emerging techniques, offering high transformation efficiency for a large array of crop species and improved sgRNA delivery efficiency by new DNA carriers without tissue culture would be critical for further large-scale exploration of mutants and precise crop breeding.Citation113 To enhance the utility and robustness of CRISPR-based screens, various scientific groups have refined and optimized screening approaches as well as developed computational tools that could help in designing large-scale CRISPR-based screens, including CRISPRa (CRISPR activation) and CRISPRi (CRISPR inhibition) tools.Citation114–116

5. Integrating Cis-Genesis and CRISPR/Cas Systems: Harnessing Precise Crop Breeding

Previously, CRISPR/Cas was used to target coding regions to generate null alleles. While this application notably enables heritable alleles for reverse genetic analysis, selection of loss-of- function mutations in coding sequences may culminate in deleterious or pleiotropic effects.Citation117,Citation118 On the other hand, gene expression modulation can use cis-engineering to be advantageous for crop improvement, with fewer detrimental pleiotropic effects.Citation119,Citation120 The key techniques for editing a gene or a genome are genetic mutations and transgene generation. Somatic hybridization enables the genetic material fusion between the two species separated by genetic barriers. Recent advances in sequencing platforms have led to the isolation of cis genes (genes from crossable species). The process of cis genesis transfers the entire gene, including its regulatory sequences in sense orientation, to sexually compatible species or a closely related species.Citation121,Citation122 In contrast, intragenesis refers to a process when coding and regulatory sequences of a gene are assembled either in antisense orientation of in sense orientation.Citation123 In theory, unlike intragenic, cis-genics can be accomplished by conventional breeding. The acceptance of cis-genic plants is more since genes are inserted from the crossable species, and the process is similar to conventional breeding. Nevertheless, in both cases, only the genes coding for traits to be improved should be ultimately traced in the regenerated crop plants, and the use of selectable marker genes can be avoided using various alternative technologies, such as cigenesis, steroid-inducible recombinase platform.Citation122,Citation124

Besides, cis-engineering through CRISPR/Cas-mediated have also succeeded in the epigenome. CRISPR/Cas9 has been used to modify the epigenome of an organism for switching OFF/ON specific gene modules. For instance, the dead Cas9 (dCas9), a catalytically inactive Cas9, has been fused to inhibitors and activators of transcription to target the specific promoters or enzymes/proteins that modify the chromatin to edit the epigenome of the target organism.Citation125,Citation126 However, only a few reports have been defined in Arabidopsis that show epiGE by DNA methylation modificationsCitation127 and histone acetylation.Citation128 Until now, there are only a few case studies under epiGE that focused attention on the applications of cis-engineering of DNA. Moreover, various recent articles have reviewed the application of cis-genic approach in enhancing the tolerance to diseases and improving good quality traits in different crops such as apple, grape, potato, poplar, and durum wheat.Citation122–124 Vanblaere et al.Citation129 reported the molecular characterization of cis-genic apple (Gala) lines with Rvi6 scab resistance genes, whereas, Würdig et al.Citation130 used cis-genesis based on the Flp/FRT recombinase system to develop apple cultivars with the same trait. Besides, Jo et al.Citation131 developed marker-free potato lines that are resistant to late blight from S. stoloniferum (Rpi-sto 1) and S. venturii (Rpi-vnt1.1) through Agrobacterium-mediated transformation by the cisgenic approach. Since the gene sequences transferred through cis-genesis are acquired from the same or closely related species, the information of their gene position, sequence, and function in the genomes is crucial. This information will offer timely access to a wider application of cis-genic technology instead of transgenesis. Rodríguez-Leal et al.Citation119 demonstrated that CRISPR/Cas9 engineering of promoters creates cis-regulatory alleles to enable quantitative variation for breeding of crops. This group edited promoters of genes that regulated three important characters in tomatoes (Solanum lycopersicum), i.e., inflorescence branching, fruit size, and plant architecture. Results showed that this method enabled rapid selection of unique alleles and fine manipulation of yield components in crop plants. Besides enhancing variation for different traits, this study provides a basis for analyzing complex associations between gene-regulatory alterations and quantitative traits. Furthermore, the cis-genic approach reduces both linkage drag and duration of selection, and the modification of genotype and phenotype characters remained largely unchanged. Remarkably, CRISPR/Cas9-driven sanitized genetic screens enable recovery of a pool of cis-regulatory alleles with a wide range of phenotypic effects, providing scope for increasing the genetic diversity in crops.Citation119

Data reveals CRISPR/Cas system is capable of targeting functional cis-regulatory elements for enhancing genetic variations in crop plants.Citation132 Until now, a few studies have been reported regarding CRE deletion or disruption to effectively control the target gene expression in crop plants. Duan, et al.Citation133 studied the expression patterns of all five OsRAVs under salt stress. Out of them, only RAV2 gene was transcriptionally controlled under enhanced salinity stress. CRISPR/Cas-mediated cis-engineering is used to mediate precise mutations in the GT-1 element of the OsRAV2 promoter. The results strongly specify that the GT-1 element switches the salt response of this gene. In another study, the promoter of barley (Hordeum vulgare) phytase gene, HvPAPhya, was targeted for three CREs, viz. Skn1, GCN4, and RY by CRISPR/Cas9 and TALENs. The plants with mutations at the targeted region indicated a considerable decline in phytase activity, showing the significance of the CREs for gene expression. Li et al.Citation134 used CRISPR/Cas9 editing to develop transgene-free bacterial blight resistance in rice by causing deletion in the promoter sequence of xa13. The Xa13 gene in rice normally controls bacterial blight disease resistance and anther development. Thus, this deletion strategy in rice improved disease resistance without affecting its fertility.

Presently, there are a few research articles published on the two-target site-directed deletion and the selection of editing sites in noncoding promoter regions. These gene-editing systems do not mutate coding regions to change their gene expression patterns. Instead, they simply edit the promoter regions that can be central to future genome engineering and breeding.Citation135,Citation136 Oliva R, et al.,Citation137 used CRISPR/Cas9 to edit TALE-binding elements (EBEs) of three SWEET gene promoters in rice, resulting in broad-spectrum bacterial blight resistant lines. Moreover, Xu et al.Citation138 employed the CRISPR/Cas9-based editing to engineer broad-spectrum resistance against rice bacterial blight by disrupting the EBEs of OsSWEET11 and OsSWEET14 S-genes. Three state-of-the-art studies in Duncan grapefruit (Citrus paradise Macf.) and Wanjincheng orange (Citrus sinensis Osbeck) validated the role of CRISPR/Cas editing of the PthA4 effector binding CREs (cis-regulatory elements) in the LATERAL ORGAN BOUNDARIES 1 (LOB1) promoter in alleviating canker resistance.Citation136,Citation139–141

The CRISPR/Cas-enabled cis-engineering was further exploited to alter known CREs in introns and downstream gene sequences.Citation142 YUCCA3 (YUC3) intron containing the CTCTGYTY motif was figured out in Arabidopsis using the chromatin immunoprecipitation sequencing (CHIP-seq) technique. It was found to be involved in engaging RELATIVE OF EARLY FLOWERING 6 (REF6) to its target site.Citation19 Removing four repeats of the CTCTGYTY motif blocked REF6 binding at the mutant loci. Also, CRE of 450 bp present in the 2nd intron of Arabidopsis AGAMOUS (AG) was excised with CRISPR/Cas9 to confirm its role as an activator of the AG gene expression. The excision of this CRE leads to early flowering after a 40% decline in its expression.Citation143 Promoter swapping and insertion is accomplished via HDR with unprecedented potential to improve different crop traits. However, the low efficiency of HDR in higher plants has been challenging.Citation144 Till date, only three cases have been investigated, wherein promoters were precisely introduced or swapped through CRISPR/Cas9-mediated HDR.Citation145–147 The 35S promoter was introduced to the upstream of the anthocyanin 1 (ANT1) gene, controlling its biosynthesis and enhancing the accumulation of anthocyanin tomato tissues.Citation145 In another study, HDR was exploited to insert and swap the GOS2 promoter in the 5′- UTR of ARGOS8 gene in maize. The mutated plants exhibited enhanced expression of ARGOS8 and improved grain yield in drought stress conditions.Citation146 Moreover, cassava (Manihot esculenta) resistance to glyphosate was created by a promoter swap of the ESPS gene.Citation147 These promising results reveal the prospect of harnessing CRISPR/Cas-enabled cis-engineering to improve various traits, for example, crop yield, quality, and stress tolerance. Besides, there is higher knowledge of transferred sequences in cis-genesis compared to conventional breeding.Citation123 Moreover, numerous reports confirmed a larger consumer acceptance of cis genic-derived products, unlike the corresponding transgenic ones.Citation123,Citation124,Citation148

6. Power of CRISPR/Cas for Enhancing Hybrid Breeding

Various recent studies have established the prospects of CRISPR to generate high-quality hybrid varieties, which are mostly based on the male-sterile maternal lines.Citation15,Citation149 A remarkable effort was made in generating male-sterile lines using CRISPR/Cas-mediated gene knockouts, including photosensitive male-sterile in CSA rice,Citation150,Citation151 ms45 wheat lines,Citation152 and thermosensitive male-sterile tms5 rice lines.Citation153 Shen, QueCitation154 demonstrated that male fertility could be rescued in Japonica indica hybrids by knocking out 1–2 Sc gene copies in the indica allele Sc-Ι. In the same way, Yu, MiaoCitation155 observed improved fertility in japonica-indica hybrids by knocking out the toxin gene ORF2, which is responsible for the felo-de-se of selfish-gene in rice. In another study, two genes, SaF/SaM and OgTPRI, at the sterility locus, Sa and S1, respectively, were disrupted to conquer the reproductive barriers in Japonica indica hybrids.Citation156,Citation157 Recently, GE was used to knockout three genes, REC8, PAIR1, and OSD1, involved in rice meiosis to substitute mitosis.Citation117 Asexual propagation lines were developed by two independent groups, either by knocking out the MTL geneCitation158 or by the activation of BBM1 in egg cells,allowing to fix hybrid heterozygosity via seed propagation.Citation159

7. Exploiting the Genetic Diversity of Wild Plants

Out of the more than 300,000 plant species available globally, only up to 200 plants are utilized at the commercial level. Moreover, only rice, maize, and wheat crops are the main energy sources for human consumption. Continuous modification and improvement of modern crops for thousands of years has led to a loss of diversity.Citation160 In addition, generating elite cultivars through further improvement by classical approaches could not always be the most prudent way for producing cultivars with novel traits, resistant to biotic/abiotic stresses. Witnessing the increased sequencing of crop genomes, GE tools offer one the efficient strategies to plant domestication by opening up the vast genetic diversity, from wild or semi-domesticated species, thus generating crops with improved polygenic traits and enhancing genetic diversity in plants. The long domestication process of modern cultivars results in loss of genetic diversity. A wild tomato plants exhibit a higher degree of tolerance to diverse stressors; thus a repository of tolerance genes to accomplish de novo-domestication through GE tools. For instance, accelerated de novo domestication of wild tomato using CRISPR/Cas9-mediated multiplex editing generated loss of function in four sets of genes coding for flowering time (SP5G; SELF PRUNING 5 G), plant architecture (SP; SELF PRUNING), fruit size (Sl CLV3; CLAVATA3, and Sl WUS; WUSCHEL).Citation161 The gene regulatory elements were targeted to create weak transcriptional alleles and target coding sequences. Moreover, CRISPR-edited plants displayed enlarged fruit size as well as synchronized flowering, determinating the plant architecture without losing tolerance to stresses.

Zsögön, ČermákCitation162 reported that de novo domestication was achieved in the ancestral tomato, Solanum pimpinellifolium, using the multiplex CRISPR/Cas9 approach, resulting in functional disruption of 06 domesticated genes coding for growth habit, nutritional traits, and yield components. They observed a ten-fold increase in fruit yield and a three-fold increase in fruit size within a single generation as compared to the wild plant. Moreover, fruit shape was found to be better and the nutritional quality increased by two-fold in lycopene content, which is equal to five-fold increase in translation compared to modern tomato cultivars. Rapid de novo domestication using the CRISPR/Cas approach was applied in an underutilized crop of Solanaceae family, ground cherry (Physalis pruinosa). A remarkable success was achieved by considering the efficient transformation procedure, gene annotation data, and reference genome. In turn, plants were produced with higher yields and increased fruit size.Citation163 In comparison to many other oilseeds, weed pennycress (Thlaspi arvense L., Brassicaceae) has a short life cycle, tolerance to extreme cold stress, and high seed oil content. It has homology with other advanced members of Brassicaceae.Citation164 Genes controlling seed dormancy (DOG1), oil content (DGAT genes), accumulation of glucosinolates (HAG1 and GTR2), and oil quality (FAE1 and FAE2) can be modified via the genome-editing technologies that should significantly contribute to the development of elite pennycress varieties. In addition, progenitors of teosinte (Zea mays spp. parviglumis), wild rice (Oryza rufipogon), and wild emmer wheat (Triticum dicoccoides) could be undertaken for re-domestication to promote agricultural diversity and decode a lot of problems facing sustainable agriculture.Citation117

Recently, Ivanizs, MonostoriCitation165 investigated the genetic diversity of Aegilops biuncialis, one of the treasured sources of agronomically important traits, which may considerably enable the introgression breeding of wheat. A correlation of the intraspecific variation in the heading time trait using analysis of variance (ANOVA) and principal component analysis (PCA) divulged four phenotypic groups, presenting association with the genetic structure and geographic distribution, apart from minor differences. The comprehensive study of genetic and phenologic differences provides an understanding of the adaptation of promising genotypes of A. biuncialis that could be exploited for wheat improvement. The CRISPR-Cas approach can be used to target agronomically important genes in A. biuncialis for wheat improvement. Thus, CRISPR-mediated de novo domestication of wild plants offers a new window for crop improvement.Citation166 Furthermore, exploiting wild crop relatives in the quest for a repository of allele mining could pave the way for developing germplasm for a wide range of crop varieties, having tolerance to biotic as well as abiotic stress factors.Citation167 Consequently, there is a huge scope for the use GE tools such as based on CRISPR/Cas mechanism for de novo domestication of neglected, semi-domesticated, and wild crop plants for food and national security.

8. A Combinatorial Approach to the Development of Disease-Resistant Crops

Induction of biotic stress to crop plants occurs majorly through virus, bacteria, fungi, nematodes, and insects. In addition, the constant increase in the number of new strains of microbial strains further complicates the battle against pathogens.Citation168,Citation169 Therefore, it is pertinent to unravel the plant interaction to protect agricultural crops from these deadly biotic stressors.Citation170 GE technologies such as CRISPR/Cas9 have been successfully used to understand the response of plants to pathogens and investigate the molecular basis of plant-pathogen interactions to develop disease-resistant crops (, ).Citation171,Citation172 For instance, disease-causing genes such as “S-genes” have been disrupted by using GE based on CRISPR/Cas9 systems to develop disease resistant crops. Similarly, resistance to rice blast and reduced blast lesions caused by Magnaporthe oryzae by the knockout of ethylene-responsive gene OsERF922 in plants were observed by using the CRISPR/Cas9 GE tool.Citation173 Likewise, targeted mutagenesis of SWEET13 gene produced bacterial blight-resistant plants upon application of CRISPR/Cas9 GE.Citation174 The improved resistance against Xanthomonas citri was due to the frame-shift mutation and disruption of the CsLOB1 gene.Citation175 Shan Q, et al.Citation176 applied the CRISPR/Cas9 technique in wheat protoplasts for editing the TaMLO geneCitation177, producing wheat lines resistant to powdery mildew due to infection of caused by Blumeria graminis f. sp. Tritici.Citation178 In a further study, resistance to powdery mildew was developed by mutating 03 homologs of the EDR1 gene by using multiplex GE by CRISPR/Cas9.Citation179 Similar results were obtained in tomato by generating mutation in the MLO gene using CRISPR/Cas9 to develop resistance against powdery mildew.Citation180 For enhancing virus resistance, sgRNAs and Cas9 were targeted in plant genomes for overexpression to inhibit the Gemini virus infection.Citation181,Citation182 To tackle wide range of viral diseases, CRISPR/Cas9 system has been widely applied to mutate the target viral genomes.Citation183 Francisella novicida (FnCas9) is a new discovered ortholog of Cas9 employed for the efficient editing of RNA virus genomes. This ortholog, FnCas9, has been successfully applied in tobacco mosaic virus and cucumber mosaic virus, inhibiting the replication and providing immunity against them.Citation184 Consequently, GE through CRISPR/Cas9 toolbox is a highly efficient and exceptional tool to improve the genetic makeup and to combat various pathogens.

Figure 4. Engineering crops for disease resistance. Schematics showing the examples of genes edited by the CRISPR/Cas system to enhance the resistance to bacteria, fungus, and virus..

Figure 4. Engineering crops for disease resistance. Schematics showing the examples of genes edited by the CRISPR/Cas system to enhance the resistance to bacteria, fungus, and virus..

Table 1. Role of CRISPR/Cas9 in the development of disease-resistant crops.

9. CRISPR/Cas9 to Produce Climate-Smart Crops

In order to cope with various abiotic stressors, the CRISPR/Cas9 technology is widely applied in major crop plants such as, rice, maize, wheat, tomato, cotton, soybean, and potato. Moreover, CRISPR/Cas9 tool has revolutionized the modernized plant-breeding strategies due to its critical role in producing climate-smart crops, resilient to a wide range of abiotic stressors.Citation15,Citation185–191 For instance, the functional studies of TaDREB3 and TaDREB2 genes located in wheat protoplast have been investigated by the CRISPR/Cas9-based GE.Citation186 Further, with a T7 endonuclease assay, approximately 70% of the transfected protoplasts have been confirmed to express mutated genes.Citation186 Similarly, in rice plants, the 03 genes, viz., phytoene desaturase (OsPDS), betaine aldehyde dehydrogenase (OsBADH2), and mitogen-activated protein kinase (OsMPK2) are edited by employing CRISPR/Cas9 GE tool. The transformation of CRISPR/Cas9 against the genes responsible for mediating stress tolerance is done by protoplast transformation and particle bombardment.Citation176 Plant annexins, including TdANN12, StANN1, BjANN1, and AnnAt8 play a major role in protecting plants from abiotic stressors. In addition, CRISPR/Cas9 system was employed to knockout the annexin gene OsAnn3 (a calcium-dependent lipid binding annexin) responsible for cold stress in rice.Citation154 Similarly, base editing system was used to validate the functions of SAPK2 gene using its critical role in salinity and drought stress.Citation192 In conclusion, CRISPR/Cas systems speed up the production of climate-resilient crop plants in the scenario of global climatic changes.

10. Organelle GE Through CRISPR/Cas System: An Overview

GE tools have emerged as the most dominant technique to develop important crops that have widespread applications in agriculture. Almost all research findings pertaining to GE of plant genomes are undoubtedly focused on plant nuclear genes. Scientists are still finding ways to modify the plastid and mitochondrial genomes since both plastome and mitome are vital for plants because of their much smaller size and because they contain nonredundant genes and genes for metabolic pathways.Citation193 Regardless of the progress in the editing of nuclear genomes, there is an important set of genes present in chloroplasts and mitochondria that plays key roles in photosynthesis, respiration, and development.Citation193,Citation194 These extra-nuclear nonredundant genes and metabolic pathways genes could be targeted for enhancing agricultural productivity.Citation195

Engineered nucleases are used to induce double-stranded DNA breaks in mitochondria.Citation196,Citation197 However, researchers need to introduce DNA templates into organelle genomes to repair and introduce desirable modifications at the break sites. For this purpose, methods and protocols are obligatory to isolate the stable organelle transformants. In the case of eukaryotes, these protocols are only available for chloroplasts, and the success rate in plastome engineering depends on the efficiency with which plastome transformation and regeneration of transplastomic plants is achieved. The engineering of the photosynthetic pathways for carbon metabolism and nitrogen fixation are two major challenges that plastome engineering can address. The key chloroplast-editing target gene, rbcL, encodes for the large catalytic subunit of Ribulose bisphosphate carboxylase/oxygenase, essential for carbon dioxide fixation, and is the focus for improvement.Citation198 Photosynthetic ability to reengineer genomes involves introducing and coordinating 20 genes or more to provide C4 photosynthesis in C3 plants. Photosynthetic efficiency can be enhanced by improving light capture, light energy conversion, CO2 uptake, and a prominent canopy.Citation195 Due to the variations in the nuclear and plastid genome characteristics, different engineering approaches came into existence.

The eukaryotic nuclear genome has the architecture, regulation, and processing of the eukaryotic genes. DNA insertion into the plant nuclear genome is most often achieved via random Agrobacterium-mediated transformation whereas plastome is an alternative compact option for targeted editing. Moreover, each gene has its own promoter, i.e., monocistronic gene structure is a general rule. On the contrary, the organelle genome is prokaryotic, and its genes are derived from operons with untranslated regions that play a pivotal role in gene expression. Foreign DNA is usually bombarded into plastome and mitochondria using a gene gun and is then incorporated at the target regions through homologous recombination. The type of genome to be edited relies on the choice of research or product. For example, single-gene governed traits that are achieved through either knockin or knockout are better targets for nuclear engineering, but plastomes and mitomes are better suited for manipulating metabolic pathways. Recently, it was observed that few noncoding RNA might possibly facilitate the import of foreign mRNA into plant chloroplasts.Citation199 Such RNA and protein delivery tools could open up the leeway of plant cell organelles’ DNA-free GE by unswervingly importing mRNA, protein, or CRISPR ribonucleoprotein complexes. The field of editing the plastome and mitome is a completely open challenge to scientists for modifying crop plants for food and nutritional security.

11. Biosafety Concerns of Gene-Edited Plants

The beginning of the editing of genomes generated not only enthusiasm but also debate, throwing up global regulatory and governance challenges, since it is evident that the use of physical as well as chemical mutagens involved in traditional plant breeding usually generates a large number of mutations in plants that are not desirable for the improvement of crops.Citation200 However, off target effects based on plant GE may not cause genetic complexities in plant breeding as the undesirable mutations are eliminated by backcrossing. CRISPR-Cas9 has been a flexible and robust tool for gene regulation and genome-editing in eukaryotic organisms, including plants.Citation32,Citation201 However, over time, it became apparent that the CRISPR-Cas9 system is affected by diverse factors such as sgRNA design, delivery methods, target site selection, Cas9 activity, off target effects, and the frequency rate of HDR.Citation32,Citation202–204 The most important apprehension is the vulnerability of producing undesirable gene alterations in plants, owing to the off target activity.Citation202–204 Furthermore, simultaneous gene editing by multiplexing will further create serious regulatory concerns, largely due to possibility of gene rearrangements.Citation205 The major concern to the CRISPR/Cas9 GE is off-target mutations that will have deleterious effects on thearget genome. Consequently, precision modifications by using CRISPR/Cas9 GE and its risk needs to be studied.Citation23,Citation38,Citation206 Off-target mutations can also threaten the environmental safety and integrity. This may happen by the transfer of off target mutations to other organisms. For instance, it is observed that, over time, the efficiency of GE in soybean embryos is enhanced by using the CRISPR/Cas9 system.

Gene drive is another worrying element as far as crop safety disquiets of GE technologies. The process of gene drive involves biased inheritance of genes from donor parents to the offspring.Citation207 The naturally existing gene drive is held in check by the naturally inbuilt mechanisms of plants.Citation207 It is reported that the CRISPR/Cas9 system may create highly efficient gene-driving genetic elements, making sure that researchers understand the clear containment and confinement conditions of the gene-drive possibilities. All the sam, we believe that there is limited possibility of gene drive in domesticated plants due to controlled breeding and a longer generation time.

Interestingly, DNA-free GE with in vitro preassembled CRISPR/Cas9 ribonucleoproteins (RNPs) prevented the off target mutations and transgene integration to unpredictable levels, thus improving the CRISPR-Cas9 specificity in plants.Citation42,Citation202,Citation204 However, the presence of cell wall makes it a challenging task for transformation through electroporation, lipofection, and microinjection in intact plant cells. However, in recent times, this problem has been solved by inserting Cas9 RNPs into protoplasts of lettuce, followed by tissue regeneration.Citation208 Moreover, Svitashev, SchwartzCitation209 used biolistic bombardment successfully to deliver Cas9 RNPs or in vitro transcripts (IVTs) into young embryos of wheat and maize. Hitherto, these transgene-free approaches are possible only with few plant varieties and species due to the high cost, less stability, and high technological inputs.Citation204,Citation210–212 Various research groups have done substantial work to decrease the off target activity of Cas9, together with improving gRNA design, delivery of RNPs, and protein engineering by systematically controlled Cas9 and gRNAs via an excess of environmental or chemicals or by employing artificial genetic circuits, which control the CRISPR function.Citation202,Citation204,Citation209 Similarly, improvements in base editing to enhance its specificity are achieved by restricting the deaminase activity outside Cas9 binding using various engineered deaminases or by using deaminase effectors to reduce its affinity to bind DNA.Citation91,Citation213

CRISPR/Cas9 technology also faces concerns linked to the Cas9 protein because it was revealed that an immune response was induced in mice through adeno-associated viral delivery of Cas9.Citation204,Citation214 Besides, Cas9 specificity and the limited number of targeted sites, owing to the PAM requirement, is a matter of concern.Citation215,Citation216 Moreover, functional characterization of gRNAs, prior to application, is significantly important because of the varying gRNA activities, thus making gRNA selection difficult.Citation216 In addition, the intervention of engineering proteins for establishing the feasibility of engineering Cas9 that changes PAM specificity, improves its fidelity, and identifies other important motifs, which are of great importance.Citation204,Citation217,Citation218 Moreover, the alterations to gRNA and Cas9 design, such as dimeric Cas9-FokI fusions, paired Cas9 nickases, and utilization of truncated sgRNAs (GN17-NGG or GN18-NGG), have significantly enhanced the on targeting activity of CRISPR/Cas nucleases.Citation219,Citation220 Additionally, Cas9 variants, other homologs from bacteria, or six novel programmable CRISPR-Cas nucleases of Class 2 system were uncovered in bacterial genomes using functional and computational analyses.Citation203,Citation221

12. The Regulatory Landscape of Genome-Edited Crops

Plants, whose genome was altered in a unique way from natural processes (mating and/or natural recombination), are referred to as genetically modified (GM).Citation222,Citation223 On the other hand, GE refers to DNA modifications that can hardly be distinguished from equal modifications achieved through conventional plant breeding or natural methods.Citation201,Citation204,Citation224 The regulation of genome-edited plants was comprehensively debated by policymakers, scientists, and regulatory authorities.Citation225–227 However, different countries have followed different patterns in generating, consuming, and regulating GM plants. At the same time, several countries ban their production and avoid consumption, whereas others adore the consumption and production of GM plants.Citation204 Owing to the escalating disagreement between scientific advancement and legal regulation, the German Research Foundation and Science Academies concludes that the principally process-based European regulatory method is not permissible. Consequently, it is not justifiable that budding risks can only stem from the organisms as a product of the breeding and not from the process itself.Citation228 Therefore, the report proposes, as a first and foremost step, to revise the European GE regulation in the short term. “In a second and long-term step, the legal agenda should be renovated not on the fundamental process but on the novel traits and characteristics of an organism that are related to the health, environment and nature conservation.Citation224,Citation229

However, genome-edited plants or animals are being regulated in some countries based on biosafety frameworks developed through specific legislation, even though some countries do not consider GE organisms as GMOs.Citation204,Citation230 Hurdles in the regulatory process of plant GE comprise market access and addressing the public’s concerns about its safety, without restraining the technological advances.Citation230,Citation231 Consequently, the marketing of gene-edited plants and their resulting products might circumvent the firm biosafety protocols that are mandatory for transgenic plants.Citation224,Citation232–234 In March 2018, the United States Department of Agriculture (USDA) stated that GE, in some cases, is the same as the conventional approach and thus does not involve any regulatory error in the American Regulatory context.Citation224,Citation235 The first CRISPR-edited crops commercialized in the USA without any regulatory protocols. These include mushrooms resistant to browning and a waxy corn composed entirely of amylopectin.Citation204,Citation235 The judgment about not to regulate established on the statement that no transgene was introduced during editing, and the subsequent modification did not encompass any pesticide or herbicide resistance.

In contrast, Canada has been faithful to the scientific values placed in its domestic regulatory modus operandi for GM plants, developed 25 years back. According to the Canadian policy, any genome-editing tool that produces a product having novelty is subjected to further regulatory checks related to allergic reactions and impacts on nontarget organisms.Citation236,Citation237 The first gene-edited products that are approved in Canada are potatoes with nondark spots and nonbrowning apples. Consent was provided after a prolonged assessment procedure that showed that gene-edited apples and potatoes did not cause ill effects to human health as compared to currently available apples and potatoes in the Canadian market.Citation235 Furthermore, Argentina has established a functional regulatory protocol for certifying genome-edited products.Citation204,Citation238 Regulatory bodies and policy makers have developed assessment protocols that are related to the Cartagena Protocol on Biosafety, depending on appraisals.Citation204,Citation239 If a product developed from a transgenic technology is DNA (transgene)-free, then this product can be categorized as non-transgenic. The almost same protocol is followed by Chile and Brazil. Both countries regulate gene-edited plants on a case-by-case evaluation and exclude them from biosafety protocols when the final product is without transgene. For now, European Union (EU) countries stay politically conflicted about GM crops.Citation235 Recently, the Court of Justice of the European Union (ECJ) ruled that gene-edited crops should be subjected to similar strict regulatory guidelines as GM organisms.Citation204,Citation239 The ECJ in its judgment stated that only mutagenesis techniques that are usually used in plant breeding and have an extended safety record are exempted from this regulation. Gene Technology Act (GT Act) was introduced in 2000 in Australia, specifying that a GMO is an organism that is created by altering its genes or genome. The Gene Technology Regulations were launched in 2001, in which schedule 1 postulates that organisms are generating from the genetic material exchange. Further, an amendment was made in October 2019 to schedule 1 that eliminated GMOs improved by CRISPR-Cas9 GE tools.Citation230

In 2016, the Hazardous Substances and New Organisms Act 1996 (HSNO Act) Act was amended in New Zealand in an article explaining that GE-based plant breeding is subject to the same regulations as applied to GMOs.Citation91 In India, the process of regulatory framework was established in 1989 for the purpose of generation and utilization of GMOs and their respective products through novel genetic tools. The Food Safety and Standards Authority of India (FSSAI) defined modified food or genetically engineered as “any food or food ingredient containing GM or engineered organisms obtained through gene technology, or food and food ingredients produced from but not containing GM or engineered organisms obtained through gene technology.” Thus, the regulation of all novel GE tools, including CRISPR-Cas9 GE technology, fall within the regulatory protocols.Citation240

Japan’s Ministry of Health, Labor, and Welfare (MHLW) has released a regulatory policy on March 27, 2019, for the handling of food products derived from GE technology. The policy established that genome-edited transgene-free foods are not classified as GMOs and are not subjected to regulatory assessments. The provision states that the GE technology based on single or few nucleotides can occur naturally. Such changes are difficult to differentiate from mutational changes that occur in traditional breeding techniques. The new MHLW’s policy also specifies that off target mutations in genome-edited foods are not a matter of concern as such mutations are also detected in crops produced by conventional breeding.Citation204,Citation241,Citation242

It appears that the choice to regulate or deregulate GM crops and foods by different countries is determined based on the type of existing GMO regulations. Those countries that have implemented a process-based regulatory protocol consider only those products that are developed by the regulated process. These products are primarily different and riskier than similar products produced by other methods that likely regulate foods, and GE crop plants coming under the GMO acts. In contrast, countries that follow a product-based regulatory protocol and regulate GE based on the final product characteristics instead of the process by which it was developed might not regulate foods, and GE crops under GMO rules. Countries such as, for example, Malaysia and Thailand, adopted both product- and process-based approaches, and they also possibly regulate foods and GE crops under the regulation of GMO laws.Citation224,Citation240

13. Conclusion and Future Directions

Over the last decade, we have seen a period of dramatic change in the rapidly developing GE technologies at the forefront of the effort to evolve crop plants for desirable traits. Large-scale production of low-cost, safe, and nutritious food by adopting a sustainable agricultural system is a major challenge for agricultural scientists in the current scenario. This goal can be achieved by using modern technologies to improve wild crops. Among these, genome-editing technologies are at the forefront to modify crop genomes to enhance production and create resilient features. The GE technologies employed for decades including ZFN, TALENS, Cre-LoX-P, and CRISPR/Cas system have been used for crop improvement to settle the issue of food security and nutritional quality.Citation243,Citation244 Many barriers to genome editing have been addressed by the use of CRISPR/Cas9 toolbox, which has also given crop improvement ideas a new start. In addition, they are integrating cis-genesis and harnessing precise crop breeding. Moreover, the power of CRISPR/Cas is validated by its applications to enhance hybrid breeding and enhance plants’ genetic diversity. The introduction of CRISPR/Cas GE provides scientists with an easy way to modulate target crop plants for a specific class of genes in a more efficient and precise manner. Wide usage and versatility of CRISPR/Cas GE tool is effectively practiced in functional genomic studies and molecular crop breeding programs, with incomparable results in terms of producing large crop varieties with special agronomic traits. Exploring the molecular phenomenon involved in stress biology of crop plants through CRISPR/Cas systems led to the development of superior and elite crop plants resilient to various biotic as well as abiotic stress factors.Citation120 Given that CRISPR/Cas systems are among the most important gene editing technology employed globally, the tool still has limitations like off targeting in plant genomes. This issue can be resolved by getting the whole genome sequence of the target plant and by designing sgRNAs that are highly tailored to the target sequences.Citation245 Other pitfalls include obstacles in the delivery of CRISPR cargoes and demands for devilment of the novel carriers for efficient transformation. Moreover, the crops developed through CRISPR/Cas systems have no involvement of exogenous DNA, and the editing of genomes is very much in compliance with the biosafety regulations. Due to the widespread use of CRISPR/Cas and its variants in figuring out the molecular mechanism behind the fundamental procedure involved in crop improvement, we can come to inference that it is a promising toolkit to engineer climate-resilient future crop plants.Citation252 Recently, Mobile CRISPR-RNA system has been introduced to produce transgene-free and genetically stable plants in difficult-to-propagate and near-extinct species.Citation252 The mobile CRISPR-RNA method expands the application of GE to low-regenerative plant species and could be applied to monocots through grafting. Future crop improvement methods should be based on maximum production within a short timespan and a species conservation initiative, without compromising on genetic diversity, and on quality characteristics by developing mutant plants via grafting. Despite several challenges, the current pace and utilization of CRISPR/Cas systems in crop improvement validates its expedition to serve humans by attaining food, nutrition and climate security.

Author Contributions

AT, MM, HY, SA, SMZ, SC, BAB, RAM, and AR conceived the idea, collected the literature, and participated in writing the manuscript. AT, BAB, AR, and RAM designed figures and table. AR, SA, RAM, SMZ, MSM, QUZ, and PVVP reviewed and edited the manuscript.

Disclosure statement

The authors do not have any conflict of interest.

Correction Statement

This article has been corrected with minor changes. These changes do not impact the academic content of the article.

Additional information

Funding

This research received no external funding.

References

  • Farooq MS, Uzair M, Raza A, Habib M, Xu Y, Yousuf M, Yang SH, Ramzan Khan M. Uncovering the Research Gaps to Alleviate the Negative Impacts of Climate Change on Food Security: a Review. Front Plant Sci. 2022;13:927535. doi:10.3389/fpls.2022.927535.
  • Scheben A, Yuan Y, Edwards D. Advances in genomics for adapting crops to climate change. Current Plant Biology. 2016;6:2–10. doi:10.1016/j.cpb.2016.09.001.
  • Kang Y, Khan S, Ma X. Climate change impacts on crop yield, crop water productivity and food security–A review. Progress In Natural Science. 2009;19(12):1665–74. doi:10.1016/j.pnsc.2009.08.001.
  • Rivero RM, Mittler R, Blumwald E, Zandalinas SI. Developing climate‐resilient crops: improving plant tolerance to stress combination. The Plant Journal. 2022;109(2):373–89. doi:10.1111/tpj.15483.
  • Zandalinas SI, Fritschi FB, Mittler R. Global warming, climate change, and environmental pollution: recipe for a multifactorial stress combination disaster. Trends Plant Sci. 2021;26(6):588–99. doi:10.1016/j.tplants.2021.02.011.
  • Raza A, Charagh S, Najafi-Kakavand S, Abbas S, Shoaib Y, Anwar S, Sharifi S, Lu G, Siddique KHM. Role of phytohormones in regulating cold stress tolerance: physiological and molecular approaches for developing cold-smart crop plants. Plant Stress. 2023;8:100152. doi:10.1016/j.stress.2023.100152.
  • Oladosu Y, Rafii MY, Abdullah N, Hussin G, Ramli A, Rahim HA, Miah G, Usman M. Principle and application of plant mutagenesis in crop improvement: a review. Biotechnol Biotechnol Equip. 2016;30(1):1–16. doi:10.1080/13102818.2015.1087333.
  • Nasti RA, Voytas DF. Attaining the promise of plant gene editing at scale. Proc Natl Acad Sci. 2021;118(22). doi:10.1073/pnas.2004846117.
  • Yu HQ, Sun FA, Feng WQ, Lu FZ, Li WC, Fu FL. The BES1/BZR1 transcription factors regulate growth, development and stress resistance in plants. Hereditas. 2019;41(3):206–14. doi:10.16288/j.yczz.18-253.
  • Yu H, Feng W, Sun F, Zhang Y, Qu J, Liu B, Lu F, Yang L, Fu F, Li W. Cloning and characterization of BES1/BZR1 transcription factor genes in maize. Plant Growth Regul. 2018;86(2):235–49. doi:10.1007/s10725-018-0424-2.
  • Yu H, Khalid MHB, Lu F, Sun F, Qu J, Liu B, Li W, Fu F. Isolation and identification of a vegetative organ-specific promoter from maize. Physiol Mol Biol Plants. 2019;25(1):277–87. doi:10.1007/s12298-018-0546-z.
  • Sun F, Ding L, Feng W, Cao Y, Lu F, Yang Q, Li W, Lu Y, Shabek N, Fu F, et al. Maize transcription factor ZmBES1/BZR1-5 positively regulates kernel size. J Exp Bot. 2021;72(5):1714–26. doi:10.1093/jxb/eraa544.
  • Sun F, Yu H, Qu J, Cao Y, Ding L, Feng W, Khalid MHB, Li W, Fu F. Maize ZmBES1/BZR1-5 decreases ABA sensitivity and confers tolerance to osmotic stress in transgenic Arabidopsis. Int J Mol Sci. 2020;21(3):996. doi:10.3390/ijms21030996.
  • Lu F, Li W, Peng Y, Cao Y, Qu J, Sun F, Yang Q, Lu Y, Zhang X, Zheng L, et al. ZmPP2C26 alternative splicing variants negatively regulate drought tolerance in maize. Front Plant Sci. 2022;13:871. doi:10.3389/fpls.2022.851531.
  • Yaqoob H, Tariq A, Bhat BA, Bhat KA, Nehvi IB, Raza A, Djalovic I, Prasad PV, Mir RA. Integrating genomics and genome editing for orphan crop improvement: a bridge between orphan crops and modern agriculture system. GM Crops & Food. 2023;14(1):1–20. doi:10.1080/21645698.2022.2146952.
  • Li M, Liu J-X, Deng Y-J, Xu Z-S, Xiong A-S. Heterologous expression of Arabidopsis thaliana rty gene in strawberry (Fragaria× ananassa Duch.) improves drought tolerance. BMC Plant Biol. 2021;21(1):1–20. doi:10.1186/s12870-021-03236-7.
  • Chen K, Gao C. Targeted genome modification technologies and their applications in crop improvements. Plant Cell Rep. 2014;33(4):575–83. doi:10.1007/s00299-013-1539-6.
  • Xia L, Wang K, Zhu JK. The power and versatility of genome editing tools in crop improvement. J Integr Plant Biol. 2021;63(9):1591. doi:10.1111/jipb.13160.
  • Lowder L, Malzahn A, Qi Y. Rapid evolution of manifold CRISPR systems for plant genome editing. Front Plant Sci. 2016;7:1683. doi:10.3389/fpls.2016.01683.
  • Zhu C, Bortesi L, Baysal C, Twyman RM, Fischer R, Capell T, Schillberg S, Christou P. Characteristics of genome editing mutations in cereal crops. Trends Plant Sci. 2017;22(1):38–52. doi:10.1016/j.tplants.2016.08.009.
  • Rischer H, Oksman-Caldentey K-M. Unintended effects in genetically modified crops: revealed by metabolomics? Trends Biotechnol. 2006;24(3):102–04. doi:10.1016/j.tibtech.2006.01.009.
  • Mubarik MS, MAJEED S, KHAN SH, DU X, FRELICHOWSKI JE, HINZE L, AZHAR MT. Reforming cotton genes: from elucidation of DNA structure to genome editing. Turk J Agric For. 2021;45(6):691–703. doi:10.3906/tar-2012-64.
  • Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121):819–23. doi:10.1126/science.1231143.
  • Zhang H, Zhang J, Lang Z, Botella JR, Zhu J-K. Genome editing—principles and applications for functional genomics research and crop improvement. CRC Crit Rev Plant Sci. 2017;36(4):291–309. doi:10.1080/07352689.2017.1402989.
  • 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. Nature Plants. 2019;5(4):363–68. doi:10.1038/s41477-019-0386-z.
  • Wurtzel ET, Vickers CE, Hanson AD, Millar AH, Cooper M, Voss-Fels KP, Nikel PI, Erb TJ. Revolutionizing agriculture with synthetic biology. Nat Plants. 2019;5(12):1207–10. doi:10.1038/s41477-019-0539-0.
  • Zaman QU, Li C, Cheng H, Hu Q. Genome editing opens a new era of genetic improvement in polyploid crops. null. 2019;7(2):141–50. doi:10.1016/j.cj.2018.07.004.
  • Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, Bogdanove AJ, Voytas DF. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics. 2010;186(2):757–61. doi:10.1534/genetics.110.120717.
  • Jinek M, Jiang F, Taylor DW, Sternberg SH, Kaya E, Ma E, Anders C, Hauer M, Zhou K, Lin S, et al. Structures of Cas9 endonucleases reveal RNA-mediated conformational activation. Science. 2014;343(6176). doi:10.1126/science.1247997.
  • Nidhi S, Anand U, Oleksak P, Tripathi P, Lal JA, Thomas G, Kuca K, Tripathi V. Novel CRISPR–Cas Systems: an Updated Review of the Current Achievements, Applications, and Future Research Perspectives. Int J Mol Sci. 2021;22(7):3327. doi:10.3390/ijms22073327.
  • Zhang K, Raboanatahiry N, Zhu B, Li M. Progress in genome editing technology and its application in plants. Front Plant Sci. 2017;8:177. doi:10.3389/fpls.2017.00177.
  • Mao Y, Zhang H, Xu N, Zhang B, Gou F, Zhu J-K. Application of the CRISPR–Cas system for efficient genome engineering in plants. Mol Plant. 2013;6(6):2008. doi:10.1093/mp/sst121.
  • Schornack S, Moscou MJ, Ward ER, Horvath DM. Engineering plant disease resistance based on TAL effectors. Annu Rev Phytopathol. 2013;51:383–406.
  • Lakota J. Synthetic Biology-Friend or Foe? What Kind of Threats Should We Expect? Вестник войск РХБ защиты. 2021;5:103–22.
  • Liang Z, Zhang K, Chen K, Gao C. Targeted mutagenesis in Zea mays using TALENs and the CRISPR/Cas system. Journal Of Genetics And Genomics. 2014;41(2):63–68. doi:10.1016/j.jgg.2013.12.001.
  • Mahfouz MM, Piatek A, Stewart CN Jr. Genome engineering via TALENs and CRISPR/Cas9 systems: challenges and perspectives. Plant Biotechnol J. 2014;12(8):1006–14. doi:10.1111/pbi.12256.
  • Li C, Brant E, Budak H, Zhang B. CRISPR/Cas: a Nobel Prize award-winning precise genome editing technology for gene therapy and crop improvement. J Zhejiang Univ-Sc B. 2021;22(4):253–84.
  • Zhu H, Li C, Gao C. Applications of CRISPR–Cas in agriculture and plant biotechnology. Nat Rev Mol Cell Biol. 2020;21(11):661–77. doi:10.1038/s41580-020-00288-9.
  • Walawage SL, Zaini PA, Mubarik MS, Martinelli F, Balan B, Caruso T, Leslie CA, Dandekar AM. Deploying genome editing tools for dissecting the biology of nut trees. Front Sustain Food Syst. 2019;3:100. doi:10.3389/fsufs.2019.00100.
  • Hassan MM, Zhang Y, Yuan G, De K, Chen J-G, Muchero W, Tuskan GA, Qi Y, Yang X. Construct design for CRISPR/Cas-based genome editing in plants. Trends Plant Sci. 2021;26(11):1133–52. doi:10.1016/j.tplants.2021.06.015.
  • Ding Y, Li H, Chen L-L, Xie K. Recent advances in genome editing using CRISPR/Cas9. Front Plant Sci. 2016;7:703. doi:10.3389/fpls.2016.00703.
  • 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(1):1–8. doi:10.1038/ncomms12617.
  • Xing H-L, Dong L, Wang Z-P, Zhang H-Y, Han C-Y, Liu B, Wang X-C, Chen Q-J. A CRISPR/Cas9 toolkit for multiplex genome editing in plants. BMC Plant Biol. 2014;14(1):1–12. doi:10.1186/s12870-014-0327-y.
  • Wang S, Zhang S, Wang W, Xiong X, Meng F, Cui X. Efficient targeted mutagenesis in potato by the CRISPR/Cas9 system. Plant Cell Rep. 2015;34(9):1473–76. doi:10.1007/s00299-015-1816-7.
  • 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(1):117–28. doi:10.1007/s00299-016-2062-3.
  • Gao W, Long L, Tian X, Xu F, Liu J, Singh PK, Botella JR, Song C. Genome editing in cotton with the CRISPR/Cas9 system. Front Plant Sci. 2017;8:1364. doi:10.3389/fpls.2017.01364.
  • 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(2):935–42. doi:10.1104/pp.17.00426.
  • Yang H, Wu J-J, Tang T, Liu K-D, Dai C. Crispr/cas9-mediated genome editing efficiently creates specific mutations at multiple loci using one sgRNA in Brassica napus. null. 2017;7(1):1–13. doi:10.1038/s41598-017-07871-9.
  • Zaman QU, Wen C, Yuqin S, Mengyu H, Desheng M, Jacqueline B, Baohong Z, Chao L, Qiong H. Characterization of SHATTERPROOF homoeologs and CRISPR-Cas9-mediated genome editing enhances pod-shattering resistance in Brassica napus L. The CRISPR Journal. 2021;4(3):360–70. doi:10.1089/crispr.2020.0129.
  • Zaman QU, Chu W, Hao M, Shi Y, Sun M, Sang S-F, Mei D, Cheng H, Liu J, Li C, et al. Crispr/cas9-mediated multiplex genome editing of JAGGED gene in Brassica napus L. Biomolecules. 2019;9(11):725. doi:10.3390/biom9110725.
  • 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. Curr Issues Mol Biol. 2021;43(3):1950–76. doi:10.3390/cimb43030135.
  • Jang G, Joung YH. Crispr/cas-mediated genome editing for crop improvement: current applications and future prospects. Plant Biotechnol Rep. 2019;13(1):1–10. doi:10.1007/s11816-018-0509-4.
  • Hille F, Richter H, Wong SP, Bratovič M, Ressel S, Charpentier E. The biology of CRISPR-Cas: backward and forward. Cell. 2018;172(6):1239–59. doi:10.1016/j.cell.2017.11.032.
  • Gasiunas G, Barrangou R, Horvath P, Siksnys V. Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci USA. 2012;109(39):E2579–E86. doi:10.1073/pnas.1208507109.
  • Nierzwicki L, Arantes PR, Saha A, Palermo G. Establishing the allosteric mecanism in CRISPR-Cas9. Wiley Interdiscip Rev Comput Mol Sci. 2020;e1503.
  • Koonin EV, Makarova KS, Zhang F. Diversity, classification and evolution of CRISPR-Cas systems. Curr Opin Microbiol. 2017;37:67–78.
  • 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(1):1–12. doi:10.1038/s41467-021-24707-3.
  • Jiang F, Doudna JA. CRISPR–Cas9 structures and mechanisms. Annu Rev Biophys. 2017;46(1):505–29. doi:10.1146/annurev-biophys-062215-010822.
  • Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y, Pirzada ZA, Eckert MR, Vogel J, Charpentier E. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature. 2011;471(7340):602–07. doi:10.1038/nature09886.
  • Garneau JE, Dupuis M-È, Villion M, Romero DA, Barrangou R, Boyaval P, Fremaux C, Horvath P, Magadán AH, Moineau S. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature. 2010;468(7320):67–71. doi:10.1038/nature09523.
  • Chen F, Ding X, Feng Y, Seebeck T, Jiang Y, Davis GD. Targeted activation of diverse CRISPR-Cas systems for mammalian genome editing via proximal CRISPR targeting. Nat Commun. 2017;8(1):1–12.
  • Schmid-Burgk JL, Gao L, Li D, Gardner Z, Strecker J, Lash B, Zhang F. Highly parallel profiling of Cas9 variant specificity. Mol Cell. 2020;78(4):794–800.e8. doi:10.1016/j.molcel.2020.02.023.
  • Anzalone AV, Koblan LW, Liu DR. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol. 2020;38:824–44.
  • Jiang F, Taylor DW, Chen JS, Kornfeld JE, Zhou K, Thompson AJ, Nogales E, Doudna JA. Structures of a CRISPR-Cas9 R-loop complex primed for DNA cleavage. Science. 2016;351(6275):867–71. doi:10.1126/science.aad8282.
  • Szczelkun MD, Tikhomirova MS, Sinkunas T, Gasiunas G, Karvelis T, Pschera P, Siksnys V, Seidel R. Direct observation of R-loop formation by single RNA-guided Cas9 and Cascade effector complexes. Proc Natl Acad Sci USA. 2014;111(27):9798–803. doi:10.1073/pnas.1402597111.
  • Pacesa M, Lin CH, et al. Structural basis for Cas9 off-target activity. Cell. 2022; 185(22): 4067–4081.
  • Jiang F, Zhou K, Ma L, Gressel S, Doudna JA. A Cas9–guide RNA complex preorganized for target DNA recognition. Science. 2015;348(6242):1477–81. doi:10.1126/science.aab1452.
  • Sternberg SH, LaFrance B, Kaplan M, Doudna JA. Conformational control of DNA target cleavage by CRISPR–Cas9. Nature. 2015;527(7576):110–13. doi:10.1038/nature15544.
  • Swartjes T, Staals RHJ, van der Oost J. Editor’s cut: dNA cleavage by CRISPR RNA-guided nucleases Cas9 and Cas12a. Biochem Soc Trans. 2020;48(1):207–19. doi:10.1042/BST20190563.
  • Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. science. Science. 2012;337(6096):816–21. doi:10.1126/science.1225829.
  • Babu K, Kathiresan V, Kumari P, Newsom S, Parameshwaran HP, Chen X, Liu J, Qin PZ, Rajan R. Coordinated Actions of Cas9 HNH and RuvC Nuclease Domains are Regulated by the Bridge Helix and the Target DNA Sequence. Biochemistry. 2021;60(49):3783–800. doi:10.1021/acs.biochem.1c00354.
  • Nishimasu H, Ran F, Hsu P, Konermann S, Shehata S, Dohmae N, Ishitani R, Zhang F, Nureki O. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. 2014;156(5):935–49. doi:10.1016/j.cell.2014.02.001.
  • Molla KA, Sretenovic S, Bansal KC, Qi Y. Precise plant genome editing using base editors and prime editors. Nat Plants. 2021;7(9):1166–87. doi:10.1038/s41477-021-00991-1.
  • Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM. RNA-guided human genome engineering via Cas9. Science. 2013;339(6121):823–26. doi:10.1126/science.1232033.
  • Cho SW, Kim S, Kim JM, Kim JS. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol. 2013;31(3):230–32.
  • Wang G, Li J. Review, analysis, and optimization of the CRISPR Streptococcus pyogenes Cas9 system. Medicine In Drug Discovery. 2021;9:100080.
  • Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, Zetsche B, Shalem O, Wu X, Makarova KS. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520(7546):186–91.
  • Esvelt KM, Mali P, Braff JL, Moosburner M, Yaung SJ, Church GM. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat Methods. 2013;10(11):1116–21. doi:10.1038/nmeth.2681.
  • Hou Z, Zhang Y, Propson NE, Howden SE, Chu L-F, Sontheimer EJ, Thomson JA. Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc Natl Acad Sci USA. 2013;110(39):15644–49. doi:10.1073/pnas.1313587110.
  • Edraki A, Mir A, Ibraheim R, Gainetdinov I, Yoon Y, Song C-Q, Cao Y, Gallant J, Xue W, Rivera-Pérez JA, et al. A compact, high-accuracy Cas9 with a dinucleotide PAM for in vivo genome editing. Mol Cell. 2019;73(4):714–26.e4. doi:10.1016/j.molcel.2018.12.003.
  • Kim E, Koo T, Park SW, Kim K, Cho HY, Song DW, Lee KJ, Jung, MH, Kim S. In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nat Commun. 2017;8(1):1–12.
  • Harrington LB, Paez-Espino D, Staahl BT, Chen JS, Doudna JA. A thermostable Cas9 with increased lifetime in human plasma. Nat Commun. 2017;8(1):1–8.
  • Makarova KS, Wolf YI, Iranzo J, Shmakov SA, Alkhnbashi OS, Brouns SJJ, Charpentier E, Cheng D, Haft DH, Horvath P, et al. Evolutionary classification of CRISPR–Cas systems: a burst of class 2 and derived variants. Nat Rev Microbiol. 2020;18(2):67–83. doi:10.1038/s41579-019-0299-x.
  • Zetsche B, Gootenberg J, Abudayyeh O, Slaymaker I, Makarova K, Essletzbichler P, Volz S, Joung J, van der Oost J, Regev A, et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. 2015;163(3):759–71. doi:10.1016/j.cell.2015.09.038.
  • Zetsche B, Heidenreich M, Mohanraju P, Fedorova I, Kneppers J, DeGennaro EM, Winblad N, Choudhury SR, Abudayyeh OO, Gootenberg JS, et al. Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nat Biotechnol. 2017;35(1):31–34. doi:10.1038/nbt.3737.
  • Yan WX, Hunnewell P, Alfonse LE, Carte JM, Keston-Smith E, Sothiselvam S, Garrity AJ, Chong S, Makarova KS, Koonin EV, et al. Functionally diverse type V CRISPR-Cas systems. Science. 2019;363(6422):88–91. doi:10.1126/science.aav7271.
  • Chen JS, Ma E, Harrington LB, Da Costa M, Tian X, Palefsky JM, Doudna JA. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science. 2018;360(6387):436–39. doi:10.1126/science.aar6245.
  • Wang Q, Alariqi M, Wang F, Li B, Ding X, Rui H, Li Y, Xu Z, Qin L, Sun L, et al. The application of a heat-inducible CRISPR/Cas12b (C2c1) genome editing system in tetraploid cotton (G. hirsutum) plants. Plant Biotechnol J. 2020;18(12):2436–43. doi:10.1111/pbi.13417.
  • Li S, Li J, Du W, Fu J, Sutar S, Zhao Y, Xia, L. Synthesis-dependent repair of Cpf1-induced double strand DNA breaks enables targeted gene replacement in rice. J Exp Bot. 2018;69(20):4715–21.
  • Yu K, Liu Z, Gui H, Geng L, Wei J, Liang D, Lv J, Xu J, Chen X. Highly efficient generation of bacterial leaf blight-resistant and transgene-free rice using a genome editing and multiplexed selection system. BMC Plant Biol. 2021;21(1):1–10. doi:10.1186/s12870-021-02979-7.
  • Shimatani Z, Kashojiya S, Takayama M, Terada R, Arazoe T, Ishii H, Teramura H, Yamamoto T, Komatsu H, Miura K, et al. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat Biotechnol. 2017;35(5):441–43. doi:10.1038/nbt.3833.
  • Li C, Zhang R, Meng X, Chen S, Zong Y, Lu C, Qiu J-L, Chen Y-H, Li J, Gao C. Targeted, random mutagenesis of plant genes with dual cytosine and adenine base editors. Nat Biotechnol. 2020;38(7):875–82. doi:10.1038/s41587-019-0393-7.
  • Jin S, Fei H, Zhu Z, Luo Y, Liu J, Gao S, Zhang F, Chen Y-H, Wang Y, Gao C. Rationally designed APOBEC3B cytosine base editors with improved specificity. Mol Cell. 2020;79(5):728–40.e6. doi:10.1016/j.molcel.2020.07.005.
  • Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, Liu DR. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 2017;551(7681):464–71. doi:10.1038/nature24644.
  • Kang B-C, Yun J-Y, Kim S-T, Shin Y, Ryu J, Choi M, Woo JW, Kim J-S. Precision genome engineering through adenine base editing in plants. Nature Plants. 2018;4(7):427–31. doi:10.1038/s41477-018-0178-x.
  • Li C, Zong Y, Wang Y, Jin S, Zhang D, Song Q, Zhang R, Gao C. Expanded base editing in rice and wheat using a Cas9-adenosine deaminase fusion. Genome Biol. 2018;19(1):1–9. doi:10.1186/s13059-018-1443-z.
  • Hua K, Tao X, Zhu JK. Expanding the base editing scope in rice by using Cas9 variants. Plant Biotechnol J. 2019;17(2):499–504. doi:10.1111/pbi.12993.
  • Gaudelli NM, Lam DK, Rees HA, Solá-Esteves NM, Barrera LA, Born DA, Edwards A, Gehrke JM, Lee S-J, Liquori AJ, et al. Directed evolution of adenine base editors with increased activity and therapeutic application. Nat Biotechnol. 2020;38(7):892–900. doi:10.1038/s41587-020-0491-6.
  • Richter MF, Zhao KT, Eton E, Lapinaite A, Newby GA, Thuronyi BW, Wilson C, Koblan LW, Zeng J, Bauer DE, et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat Biotechnol. 2020;38(7):883–91. doi:10.1038/s41587-020-0453-z.
  • Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 2019;576(7785):149–57. doi:10.1038/s41586-019-1711-4.
  • Jiang Y-Y, Chai Y-P, Lu M-H, Han X-L, Lin Q, Zhang Y, Zhang Q, Zhou Y, Wang X-C, Gao C, et al. Prime editing efficiently generates W542L and S621I double mutations in two ALS genes in maize. Genome Biol. 2020;21(1):1–10. doi:10.1186/s13059-020-02170-5.
  • Liu H, Ding Y, Zhou Y, Jin W, Xie K, Chen L-L. CRISPR-P 2.0: an improved CRISPR-Cas9 tool for genome editing in plants. Mol Plant. 2017;10(3):530–32. doi:10.1016/j.molp.2017.01.003.
  • Li J, Zhang S, Zhang R, Gao J, Qi Y, Song G, Li W, Li Y, Li G. Efficient multiplex genome editing by CRISPR/Cas9 in common wheat. Plant Biotechnol J. 2021;19(3):427. doi:10.1111/pbi.13508.
  • Wang C, Liu W, Wang G, Li J, Dong L, Han L, Wang Q, Tian J, Yu Y, Gao C, et al. KTN80 confers precision to microtubule severing by specific targeting of katanin complexes in plant cells. Embo J. 2017;36(23):3435–47. doi:10.15252/embj.201796823.
  • Tang X, Zheng X, Qi Y, Zhang D, Cheng Y, Tang A, Voytas D, Zhang Y. A single transcript CRISPR-Cas9 system for efficient genome editing in plants. Mol. Mol Plant. 2016;9(7):1088–91. doi:10.1016/j.molp.2016.05.001.
  • Wang M, Mao Y, Lu Y, Tao X, Zhu J-K. Multiplex gene editing in rice using the CRISPR-Cpf1 system. Mol Plant. 2017;10(7):1011–13. doi:10.1016/j.molp.2017.03.001.
  • Doench JG. Am I ready for CRISPR? A user’s guide to genetic screens. Nat Rev Genet. 2018;19(2):67–80. doi:10.1038/nrg.2017.97.
  • Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelsen TS, Heckl D, Ebert BL, Root DE, Doench JG, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014;343(6166):84–87. doi:10.1126/science.1247005.
  • 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(9):1242–45. doi:10.1016/j.molp.2017.06.007.
  • 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. Mol Plant. 2017;10(9):1238–41. doi:10.1016/j.molp.2017.06.006.
  • Jacobs TB, Zhang N, Patel D, Martin GB. Generation of a collection of mutant tomato lines using pooled CRISPR libraries. Plant Physiol. 2017;174(4):2023–37. doi:10.1104/pp.17.00489.
  • Liu H-J, Jian L, Xu J, Zhang Q, Zhang M, Jin M, Peng Y, Yan J, Han B, Liu J, et al. High-throughput CRISPR/Cas9 mutagenesis streamlines trait gene identification in maize. Plant Cell. 2020;32(5):1397–413. doi:10.1105/tpc.19.00934.
  • Nadakuduti SS, Enciso-Rodríguez F. Advances in genome editing with CRISPR systems and transformation technologies for plant DNA manipulation. Front Plant Sci. 2021;11:2267. doi:10.3389/fpls.2020.637159.
  • Doench JG, Hartenian E, Graham DB, Tothova Z, Hegde M, Smith I, Sullender M, Ebert BL, Xavier RJ, Root DE. Rational design of highly active sgRnas for CRISPR-Cas9–mediated gene inactivation. Nat Biotechnol. 2014;32(12):1262–67. doi:10.1038/nbt.3026.
  • Sanson KR, Hanna RE, Hegde M, Donovan KF, Strand C, Sullender ME, Vaimberg EW, Goodale A, Root DE, Piccioni F, et al. Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities. Nat Commun. 2018;9(1):1–15. doi:10.1038/s41467-018-07901-8.
  • Gaillochet C, Develtere W, Jacobs TB. CRISPR screens in plants: approaches, guidelines, and future prospects. Plant Cell. 2021;33(4):794–813. doi:10.1093/plcell/koab099.
  • Chen K, Wang Y, Zhang R, Zhang H, Gao C. CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu Rev Plant Biol. 2019;70(1):667–97. doi:10.1146/annurev-arplant-050718-100049.
  • Korotkova AM, Gerasimova SV, Khlestkina EK. Current achievements in modifying crop genes using CRISPR/Cas system. Vavilov J Genet Breed. 2019;23(1):29–37. doi:10.18699/VJ19.458.
  • Rodríguez-Leal D, Lemmon ZH, Man J, Bartlett ME, Lippman ZB. Engineering quantitative trait variation for crop improvement by genome editing. Cell. 2017;171(2):470–80.e8. doi:10.1016/j.cell.2017.08.030.
  • 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(8):4206. doi:10.3390/ijms22084206.
  • Sarmah BK, Gohain M, Borah BK, Acharjee S. Cisgenesis: engineering Plant Genome by Harnessing Compatible Gene Pools. Genome Engineering For Crop Improvement. 2021. p 193–216.
  • Giudice G, Moffa L, Varotto S, Cardone MF, Bergamini C, De Lorenzis G, Velasco R, Nerva L, Chitarra W. Novel and emerging biotechnological crop protection approaches. Plant Biotechnol J. 2021;19(8):1495–510. doi:10.1111/pbi.13605.
  • Holme IB, Wendt T, Holm PB. Intragenesis and cisgenesis as alternatives to transgenic crop development. Plant Biotechnol J. 2013;11(4):395–407. doi:10.1111/pbi.12055.
  • Cardi T, Varshney R. Cisgenesis and genome editing: combining concepts and efforts for a smarter use of genetic resources in crop breeding. Plant Breeding. 2016;135(2):139–47. doi:10.1111/pbr.12345.
  • Brocken DJ, Tark-Dame M, Dame RT. dCas9: a versatile tool for epigenome editing. Curr Issues Mol Biol. 2018;26(1):15–32. doi:10.21775/cimb.026.015.
  • Hilton IB, D'ippolito AM, Vockley CM, Thakore PI, Crawford GE, Reddy TE, Gersbach CA. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol. 2015;33(5):510–17.
  • Papikian A, Liu W, Gallego-Bartolomé J, Jacobsen SE. Site-specific manipulation of Arabidopsis loci using CRISPR-Cas9 SunTag systems. Nat Commun. 2019;10(1):1–11. doi:10.1038/s41467-019-08736-7.
  • Paixão JFR, Gillet F-X, Ribeiro TP, Bournaud C, Lourenço-Tessutti IT, Noriega DD, Melo BPD, de Almeida-Engler J, Grossi-de-Sa MF. Improved drought stress tolerance in Arabidopsis by CRISPR/dCas9 fusion with a Histone AcetylTransferase. 2019;9(1):1–9. doi:10.1038/s41598-019-44571-y.
  • Vanblaere T, Flachowsky H, Gessler C, Broggini GAL. Molecular characterization of cisgenic lines of apple ‘Gala’carrying the Rvi6 scab resistance gene. Plant Biotechnol J. 2014;12(1):2–9. doi:10.1111/pbi.12110.
  • Würdig J, Flachowsky H, Saß A, Peil A, Hanke M-V. Improving resistance of different apple cultivars using the Rvi6 scab resistance gene in a cisgenic approach based on the Flp/FRT recombinase system. Mol Breeding. 2015;35(3):1–18. doi:10.1007/s11032-015-0291-8.
  • Jo K-R, Kim C-J, Kim S-J, Kim T-Y, Bergervoet M, Jongsma MA, Visser RG, Jacobsen E, Vossen JH. Development of late blight resistant potatoes by cisgene stacking. BMC Biotechnol. 2014;14(1):1–10. doi:10.1186/1472-6750-14-50.
  • Crisp PA, Bhatnagar-Mathur P, Hundleby P, Godwin ID, Waterhouse PM, Hickey LT. Beyond the gene: epigenetic and cis-regulatory targets offer new breeding potential for the future. Curr Opin Biotechnol. 2022;73:88–94. doi:10.1016/j.copbio.2021.07.008.
  • Duan Y-B, Li J, Qin R-Y, Xu R-F, Li H, Yang Y-C, Ma H, Li L, Wei P-C, Yang J-B. Identification of a regulatory element responsible for salt induction of rice OsRAV2 through ex situ and in situ promoter analysis. Plant Mol Biol. 2016;90(1–2):49–62. doi:10.1007/s11103-015-0393-z.
  • Li C, Li W, Zhou Z, Chen H, Xie C, Lin Y. A new rice breeding method: cRISPR/Cas9 system editing of the Xa13 promoter to cultivate transgene‐free bacterial blight‐resistant rice. Plant Biotechnol J. 2020;18(2):313.
  • Schiml S, Puchta H. Revolutionizing plant biology: multiple ways of genome engineering by CRISPR/Cas. Plant Methods. 2016;12(1):1–9. doi:10.1186/s13007-016-0103-0.
  • 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 CsLOB1 promoter in citrus. Plant Biotechnol J. 2017;15(12):1509–19. doi:10.1111/pbi.12733.
  • Oliva R, Ji C, Atienza-Grande G, Huguet-Tapia JC, Perez-Quintero A, Li T, Eom J-S, Li C, Nguyen H, Liu B, et al. Broad-spectrum resistance to bacterial blight in rice using genome editing. Nat Biotechnol. 2019;37(11):1344–50. doi:10.1038/s41587-019-0267-z.
  • Xu Z, Xu X, Gong Q, Li Z, Li Y, Wang S, Yang Y, Ma W, Liu L, Zhu B, et al. Engineering broad-spectrum bacterial blight resistance by simultaneously disrupting variable TALE-binding elements of multiple susceptibility genes in rice. Mol Plant. 2019;12(11):1434–46. doi:10.1016/j.molp.2019.08.006.
  • Jia H, Orbovic V, Jones JB, Wang N. Modification of the PthA4 effector binding elements in Type I Cs LOB 1 promoter using Cas9/sg RNA to produce transgenic Duncan grapefruit alleviating XccΔpthA4: dCs LOB 1.3 infection. Plant Biotechnol J. 2016;14(5):1291–301. doi:10.1111/pbi.12495.
  • Jia H, Wang N. Generation of homozygous canker‐resistant citrus in the T0 generation using CRISPR‐SpCas9p. Plant Biotechnol J. 2020;18(10):1990. doi:10.1111/pbi.13375.
  • Jia H, Omar AA, Orbović V, Wang N. Biallelic editing of the LOB1 promoter via CRISPR/Cas9 creates canker-resistant ‘Duncan’grapefruit. Phytopathology. 2022;112(2):308–314.
  • Li Q, Sapkota M, van der Knaap E. Perspectives of CRISPR/Cas-mediated cis-engineering in horticulture: unlocking the neglected potential for crop improvement. Horticul Res. 2020;7(1):1–11. doi:10.1038/s41438-020-0258-8.
  • Yan W, Chen D, Kaufmann K. Efficient multiplex mutagenesis by RNA-guided Cas9 and its use in the characterization of regulatory elements in the AGAMOUS gene. Plant Methods. 2016;12(1):1–9. doi:10.1186/s13007-016-0125-7.
  • Liu M, Rehman S, Tang X, Gu K, Fan Q, Chen D, Ma W. Methodologies for improving HDR efficiency. Front Genet. 2019;9:691. doi:10.3389/fgene.2018.00691.
  • Čermák T, Baltes NJ, Čegan R, Zhang Y, Voytas DF. High-frequency, precise modification of the tomato genome. Genome Biol. 2015;16(1):1–15. doi:10.1186/s13059-015-0796-9.
  • Shi J, Gao H, Wang H, Lafitte HR, Archibald RL, Yang M, Hakimi SM, Mo H, Habben JE. ARGOS 8 variants generated by CRISPR‐Cas9 improve maize grain yield under field drought stress conditions. Plant Biotechnol J. 2017;15(2):207–16. doi:10.1111/pbi.12603.
  • Hummel AW, Chauhan RD, Cermak T, Mutka AM, Vijayaraghavan A, Boyher A, Starker CG, Bart R, Voytas DF, Taylor NJ. Allele exchange at the EPSPS locus confers glyphosate tolerance in cassava. Plant Biotechnol J. 2018;16(7):1275–82. doi:10.1111/pbi.12868.
  • Mohanta TK, Bashir T, Hashem A, Abd_allah E, Bae H. Genome editing tools in plants. Genes. 2017;8(12):399. doi:10.3390/genes8120399.
  • Ahmad S, Tang L, Shahzad R, Mawia AM, Rao GS, Jamil S, Wei C, Sheng Z, Shao G, Wei X, et al. CRISPR-based crop improvements: a way forward to achieve zero hunger. J Agric Food Chem. 2021;69(30):8307–23. doi:10.1021/acs.jafc.1c02653.
  • Zhou H, He M, Li J, Chen L, Huang Z, Zheng S, Zhu L, Ni E, Jiang D, Zhao B, et al. Development of commercial thermo-sensitive genic male sterile rice accelerates hybrid rice breeding using the CRISPR/Cas9-mediated TMS5 editing system. . 2016;6(1):1–12. doi:10.1038/srep37395.
  • Rao MJ, Wang L. CRISPR/Cas9 technology for improving agronomic traits and future prospective in agriculture. Planta. 2021;254(4):1–16. doi:10.1007/s00425-021-03716-y.
  • Singh M, Kumar M, Albertsen MC, Young JK, Cigan AM. Concurrent modifications in the three homeologs of Ms45 gene with CRISPR-Cas9 lead to rapid generation of male sterile bread wheat (Triticum aestivum L.). Plant Mol Biol. 2018;97(4–5):371–83. doi:10.1007/s11103-018-0749-2.
  • Barman HN, Sheng Z, Fiaz S, Zhong M, Wu Y, Cai Y, Wang W, Jiao G, Tang S, Wei X, et al. Generation of a new thermo-sensitive genic male sterile rice line by targeted mutagenesis of TMS5 gene through CRISPR/Cas9 system. BMC Plant Biol. 2019;19(1):1–9. doi:10.1186/s12870-019-1715-0.
  • 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(6):539–47. doi:10.1007/s12374-016-0400-1.
  • Yu J, Miao J, Zhang Z, Xiong H, Zhu X, Sun X, Pan Y, Liang Y, Zhang Q, Abdul Rehman RM, et al. Alternative splicing of Os LG 3b controls grain length and yield in japonica rice. Plant Biotechnol J. 2018;16(9):1667–78. doi:10.1111/pbi.12903.
  • Xie Y, Niu B, Long Y, Li G, Tang J, Zhang Y, Ren D, Liu YG, Chen L. Suppression or knockout of SaF / SaM overcomes the Sa-mediated hybrid male sterility in rice. J Integr Plant Biol. 2017;59(9):669–79. doi:10.1111/jipb.12564.
  • Xie Y, Xu P, Huang J, Ma S, Xie X, Tao D, Chen L, Liu Y-G. Interspecific hybrid sterility in rice is mediated by OgTPR1 at the S1 locus encoding a peptidase-like protein. Mol Plant. 2017;10(8):1137–40. doi:10.1016/j.molp.2017.05.005.
  • Wang C, Liu Q, Shen Y, Hua Y, Wang J, Lin J, Wu M, Sun T, Cheng Z, Mercier R, et al. Clonal seeds from hybrid rice by simultaneous genome engineering of meiosis and fertilization genes. Nat Biotechnol. 2019;37(3):283–86. doi:10.1038/s41587-018-0003-0.
  • Khanday I, Skinner D, Yang B, Mercier R, Sundaresan V. A male-expressed rice embryogenic trigger redirected for asexual propagation through seeds. Nature. 2019;565(7737):91–95. doi:10.1038/s41586-018-0785-8.
  • Østerberg JT, Xiang W, Olsen LI, Edenbrandt AK, Vedel SE, Christiansen A, Landes X, Andersen MM, Pagh P, Sandøe P, et al. Accelerating the domestication of new crops: feasibility and approaches. Trends Plant Sci. 2017;22(5):373–84. doi:10.1016/j.tplants.2017.01.004.
  • Fernie AR, Yan J. De Novo domestication: an alternative route toward new crops for the future. Mol Plant. 2019;12(5):615–31. doi:10.1016/j.molp.2019.03.016.
  • Zsögön A, Čermák T, Naves ER, Notini MM, Edel KH, Weinl S, Freschi L, Voytas DF, Kudla J, Peres LEP. De Novo domestication of wild tomato using genome editing. Nat Biotechnol. 2018;36(12):1211–16. doi:10.1038/nbt.4272.
  • Lemmon ZH, Reem NT, Dalrymple J, Soyk S, Swartwood KE, Rodriguez-Leal D, Van Eck J, Lippman ZB. Rapid improvement of domestication traits in an orphan crop by genome editing. Nature Plants. 2018;4(10):766–70. doi:10.1038/s41477-018-0259-x.
  • Sedbrook JC, Phippen WB, Marks MD. New approaches to facilitate rapid domestication of a wild plant to an oilseed crop: example pennycress (Thlaspi arvense L.). Plant Sci. 2014;227:122–32. doi:10.1016/j.plantsci.2014.07.008.
  • Ivanizs L, Monostori I, Farkas A, Megyeri M, Mikó P, Türkösi E, Gaál E, Lenykó-Thegze A, Szőke-Pázsi K, Szakács É, et al. Unlocking the genetic diversity and population structure of a wild gene source of wheat, Aegilops biuncialis Vis., and its relationship with the heading time. Front Plant Sci. 2019;10:1531. doi:10.3389/fpls.2019.01531.
  • Pramanik D, Shelake RM, Kim MJ, Kim J-Y. CRISPR-mediated engineering across the central dogma in plant biology for basic research and crop improvement. Mol Plant. 2021;14(1):127–50. doi:10.1016/j.molp.2020.11.002.
  • González Guzmán M, Cellini F, Fotopoulos V, Balestrini R, Arbona V. New approaches to improve crop tolerance to biotic and abiotic stresses. Physiol Plant. 2022;174(1). doi:10.1111/ppl.13547.
  • Al-Sadi AM, Al-Moqbali HS, Al-Yahyai RA, Al-Said FA. AFLP data suggest a potential role for the low genetic diversity of acid lime (Citrus aurantifolia Swingle) in Oman in the outbreak of witches’ broom disease of lime. Euphytica. 2012;188(2):285–97. doi:10.1007/s10681-012-0728-7.
  • Collinge DB, Sarrocco S. Transgenic approaches for plant disease control: status and prospects 2021. null. 2022;71(1):207–25. doi:10.1111/ppa.13443.
  • Kettles GJ, Kanyuka K. Dissecting the molecular interactions between wheat and the fungal pathogen Zymoseptoria tritici. Front Plant Sci. 2016;7:508. doi:10.3389/fpls.2016.00508.
  • Tyagi S, Kumar R, Kumar V, Won SY, Shukla P. Engineering disease resistant plants through CRISPR-Cas9 technology. GM Crops & Food. 2021;12(1):125–44. doi:10.1080/21645698.2020.1831729.
  • Alphonse V, Marimuthu J, Murugan K. CRISPR/Cas system for the development of disease resistance in horticulture crops. CRISPR And RNAi Systems. 2021;107–28. Elsevier.
  • 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(4):e0154027. doi:10.1371/journal.pone.0154027.
  • Zhou J, Peng Z, Long J, Sosso D, Liu B, Eom J-S, Huang S, Liu S, Vera Cruz C, Frommer WB, et al. Gene targeting by the TAL effector PthXo2 reveals cryptic resistance gene for bacterial blight of rice. The Plant Journal. 2015;82(4):632–43. doi:10.1111/tpj.12838.
  • Jia H, Zhang Y, Orbović V, Xu J, White FF, Jones JB, Wang N. Genome editing of the disease susceptibility gene Cs LOB 1 in citrus confers resistance to citrus canker. Plant Biotechnol J. 2017;15(7):817–23. doi:10.1111/pbi.12677.
  • Shan Q, Wang Y, Li J, Zhang Y, Chen K, Liang Z, Zhang K, Liu J, Xi JJ, Qiu J-L, et al. Targeted genome modification of crop plants using a CRISPR-Cas system. Nat Biotechnol. 2013;31(8):686–88. doi:10.1038/nbt.2650.
  • Zhang H, Zhang J, Wei P, Zhang B, Gou F, Feng Z, Mao Y, Yang L, Zhang H, Xu N, et al. The CRISPR/C as 9 system produces specific and homozygous targeted gene editing in rice in one generation. Plant Biotechnol J. 2014;12(6):797–807. doi:10.1111/pbi.12200.
  • 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 Biotechnol. 2014;32(9):947–51. doi:10.1038/nbt.2969.
  • 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. The Plant Journal. 2017;91(4):714–24. doi:10.1111/tpj.13599.
  • 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. . 2017;7(1):1–6. doi:10.1038/s41598-017-00578-x.
  • 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(1):1–7. doi:10.1186/s13059-018-1580-4.
  • Ali Z, Abulfaraj A, Idris A, Ali S, Tashkandi M, Mahfouz MM. Crispr/cas9-mediated viral interference in plants. Genome Biol. 2015;16(1):1–11. doi:10.1186/s13059-015-0799-6.
  • Zaidi SS-E-A, Tashkandi M, Mansoor S, Mahfouz MM. Engineering plant immunity: using CRISPR/Cas9 to generate virus resistance. Front Plant Sci. 2016;7:1673.
  • 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 Biotechnol J. 2018;16(8):1415–23.
  • Farooq MU, Bashir MF, Khan M, Iqbal B, Ali Q. Role of crispr to improve abiotic stress tolerance in crop plants. Biol Clin Sci Res J. 2021;2021(1). doi:10.54112/bcsrj.v2021i1.69.
  • Kim D, Alptekin B, Budak H. CRISPR/Cas9 genome editing in wheat. Funct Integr Genomics. 2018;18(1):31–41. doi:10.1007/s10142-017-0572-x.
  • Yadav R, Thankappan R, Kumar A. Novel Approaches for Genome Editing to Develop Climate Smart Crops. Microbiomes And The Global Climate Change. 2021;267–91. Springer.
  • Raza A, Tabassum J, Fakhar AZ, Sharif R, Chen H, Zhang C, Ju L, Fotopoulos V, Siddique KHM, Singh RK, et al. Smart reprograming of plants against salinity stress using modern biotechnological tools. Crit Rev Biotechnol. 2022:1–28. doi:10.1080/07388551.2022.2093695
  • Raza A, Mubarik MS, Sharif R, Habib M, Jabeen W, Zhang C, Chen H, Chen Z-H, Siddique KHM, Zhuang W, et al. Developing drought‐smart, ready‐to‐grow future crops. Plant Genome. 2023;16(1):e20279. doi:10.1002/tpg2.20279.
  • Raza A, Charagh S, García-Caparrós P, Rahman MA, Ogwugwa VH, Saeed F, Jin W. Melatonin-mediated temperature stress tolerance in plants. GM Crops & Food. 2022;13(1):196–217. doi:10.1080/21645698.2022.2106111.
  • Raza A, Tabassum J, Kudapa H, Varshney RK. Can omics deliver temperature resilient ready-to-grow crops? Crit Rev Biotechnol. 2021;41(8):1209–32. doi:10.1080/07388551.2021.1898332.
  • 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. doi:10.3389/fpls.2017.00993.
  • Li S, Chang L, Zhang J. Advancing organelle genome transformation and editing for crop improvement. Plant Commun. 2021;2(2):100141. doi:10.1016/j.xplc.2021.100141.
  • Okie JG, Smith VH, Martin-Cereceda M. Major evolutionary transitions of life, metabolic scaling and the number and size of mitochondria and chloroplasts. Proc R Soc B Biol Sci. 2016;283283(1831):20160611. doi:10.1098/rspb.2016.0611.
  • Ort DR, Merchant SS, Alric J, Barkan A, Blankenship RE, Bock R, Croce R, Hanson MR, Hibberd JM, Long SP, et al. Redesigning photosynthesis to sustainably meet global food and bioenergy demand. Proc Natl Acad Sci USA. 2015;112(28):8529–36. doi:10.1073/pnas.1424031112.
  • Bacman SR, Williams SL, Pinto M, Peralta S, Moraes CT. Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTalens. Nat Med. 2013;19(9):1111–13. doi:10.1038/nm.3261.
  • Kang B-C, Bae S-J, Lee S, Lee JS, Kim A, Lee H, Baek G, Seo H, Kim J, Kim J-S. Chloroplast and mitochondrial DNA editing in plants. Nat Plants. 2021;7(7):899–905. doi:10.1038/s41477-021-00943-9.
  • Lin C-P, Ko C-Y, Kuo C-I, Liu M-S, Schafleitner R, Chen LFO. Transcriptional slippage and RNA editing increase the diversity of transcripts in chloroplasts: insight from deep sequencing of Vigna radiata genome and transcriptome. PLos One. 2015;10(6):e0129396. doi:10.1371/journal.pone.0129396.
  • Gómez G, Pallás V, Zhang B. Noncoding RNA mediated traffic of foreign mRNA into chloroplasts reveals a novel signaling mechanism in plants. PLos One. 2010;5(8):e12269. doi:10.1371/journal.pone.0012269.
  • Salvi S, Druka A, Milner SG, Gruszka, D. Induced genetic variation, TILLING and NGS-based cloning. Biotechnological Approaches To Barley Improvement. 2014;287–310. Springer.
  • Mushtaq M, Sakina A, Wani SH, Shikari AB, Tripathi P, Zaid A, Galla A, Abdelrahman M, Sharma M, Singh AK, et al. Harnessing genome editing techniques to engineer disease resistance in plants. Front Plant Sci. 2019;10:550. doi:10.3389/fpls.2019.00550.
  • Liang Z, Chen K, Zhang Y, Liu J, Yin K, Qiu J-L, Gao C. Genome editing of bread wheat using biolistic delivery of CRISPR/Cas9 in vitro transcripts or ribonucleoproteins. Nat Protoc. 2018;13(3):413–30. doi:10.1038/nprot.2017.145.
  • Pineda M, Lear A, Collins JP, Kiani S. Safe CRISPR: challenges and possible solutions. Trends Biotechnol. 2019;37(4):389–401. doi:10.1016/j.tibtech.2018.09.010.
  • El-Mounadi K, Morales-Floriano ML, Garcia-Ruiz H. Principles, applications, and biosafety of plant genome editing using CRISPR-Cas9. Front Plant Sci. 2020;11:56. doi:10.3389/fpls.2020.00056.
  • Corrigan-Curay J, O’Reilly M, Kohn DB, Cannon PM, Bao G, Bushman FD, Carroll D, Cathomen T, Joung JK, Roth D, et al. Genome editing technologies: defining a path to clinic: genomic editing: establishing preclinical toxicology standards, bethesda, maryland 10 June 2014. Mol Ther. 2015;23(5):796–806. doi:10.1038/mt.2015.54.
  • Ma Y, Zhang L, Huang X. Genome modification by CRISPR/Cas9. FEBS J. 2014;281(23):5186–93. doi:10.1111/febs.13110.
  • National Academies of Sciences, Engineering and Medicine. Gene drives on the horizon: advancing science, navigating uncertainty, and aligning research with public values. Washington (DC): National Academies Press (US);2016. PMID: 27536751.
  • Woo JW, Kim J, Kwon SI, Corvalán C, Cho SW, 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 Biotechnol. 2015;33(11):1162–64. doi:10.1038/nbt.3389.
  • Svitashev S, Schwartz C, Lenderts B, Young JK, Mark Cigan A. Genome editing in maize directed by CRISPR–Cas9 ribonucleoprotein complexes. Nat Commun. 2016;7(1):1–7. doi:10.1038/ncomms13274.
  • Subburaj S, Chung SJ, Lee C, Ryu S-M, Kim DH, Kim J-S, Bae S, Lee G-J. Site-directed mutagenesis in Petunia× hybrida protoplast system using direct delivery of purified recombinant Cas9 ribonucleoproteins. Plant Cell Rep. 2016;35(7):1535–44. doi:10.1007/s00299-016-1937-7.
  • Murovec J, Guček K, Bohanec B, Avbelj M, Jerala R. DNA-free genome editing of Brassica oleracea and B. rapa protoplasts using CRISPR-Cas9 ribonucleoprotein complexes. Front Plant Sci. 2018;9:1594. doi:10.3389/fpls.2018.01594.
  • Mao Y, Zhang Z, Feng Z, Wei P, Zhang H, Botella JR, Zhu J-K. Development of germ‐line‐specific CRISPR‐Cas9 systems to improve the production of heritable gene modifications in Arabidopsis. Plant Biotechnol J. 2016;14(2):519–32. doi:10.1111/pbi.12468.
  • Molla KA, Yang Y. Crispr/cas-mediated base editing: technical considerations and practical applications. Trends Biotechnol. 2019;37(10):1121–42. doi:10.1016/j.tibtech.2019.03.008.
  • Chew WL, Tabebordbar M, Cheng JKW, Mali P, Wu EY, Ng AHM, Zhu K, Wagers AJ, Church GM. A multifunctional AAV–CRISPR–Cas9 and its host response. Nat Methods. 2016;13(10):868–74. doi:10.1038/nmeth.3993.
  • Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK, Sander JD. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol. 2013;31(9):822–26. doi:10.1038/nbt.2623.
  • Char SN, Unger-Wallace E, Frame B, Briggs SA, Main M, Spalding MH, Vollbrecht E, Wang K, Yang B. Heritable site-specific mutagenesis using TALENs in maize. Plant Biotechnol J. 2015;13(7):1002–10. doi:10.1111/pbi.12344.
  • Kleinstiver BP, Tsai SQ, Prew MS, Nguyen NT, Welch MM, Lopez JM, McCaw ZR, Aryee MJ, Joung JK. Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells. Nat Biotechnol. 2016;34(8):869–74. doi:10.1038/nbt.3620.
  • Leenay RT, Beisel CL. Deciphering, communicating, and engineering the CRISPR PAM. J Mol Biol. 2017;429(2):177–91. doi:10.1016/j.jmb.2016.11.024.
  • Wyvekens N, Topkar VV, Khayter C, Joung JK, Tsai SQ. Dimeric CRISPR RNA-guided FokI-dCas9 nucleases directed by truncated gRnas for highly specific genome editing. Hum Gene Ther. 2015;26(7):425–31. doi:10.1089/hum.2015.084.
  • O’Geen H, Abigail SY, Segal DJ. How specific is CRISPR/Cas9 really? Curr Opin Chem Biol. 2015;29:72–78.
  • Nakade S, Yamamoto T, Sakuma T. Cas9, Cpf1 and C2c1/2/3―What’s next? Bioengineered. 2017;8(3):265–73. doi:10.1080/21655979.2017.1282018.
  • Duensing N, Sprink T, Parrott WA, Fedorova M, Lema MA, Wolt JD, Bartsch D. Novel features and considerations for ERA and regulation of crops produced by genome editing. Frontiers In Bioengineering And Biotechnology. 2018;6:79.
  • Friedrichs S, Takasu Y, Kearns P, Dagallier B, Oshima R, Schofield J, Moreddu C. Meeting report of the OECD conference on “genome editing: applications in agriculture—implications for health, environment and regulation. Springer; 2019.
  • Schiemann J, Robienski J, Schleissing S, Spök A, Sprink T, Wilhelm RA. Editorial: plant Genome Editing – Policies and Governance. Front Plant Sci. 2020;11:284. doi:10.3389/fpls.2020.00284.
  • Ammann K. Selected Innovative Solutions for the Regulation of GM Crops in Times of Gene Editing. Plant Biotechnology: Progress In Genomic Era. 2019;3–41. Springer.
  • Wolt JD, Wang K, Yang B. The regulatory status of genome‐edited crops. Plant Biotechnol J. 2016;14(2):510–18. doi:10.1111/pbi.12444.
  • Wolt JD, Wolf C. Policy and governance perspectives for regulation of genome edited crops in the United States. Front Plant Sci. 2018;9:1606. doi:10.3389/fpls.2018.01606.
  • Kok EJ, Glandorf DCM, Prins TW, Visser RGF. Food and environmental safety assessment of new plant varieties after the European Court decision: process-triggered or product-based? Trends in Food Sci Technol. 2019;88:24–32. doi:10.1016/j.tifs.2019.03.007.
  • Okoli AS, Blix T, Myhr AI, Xu W, Xu X. Sustainable use of CRISPR/Cas in fish aquaculture: the biosafety perspective. Transgenic Res. 2021;31(1):1–21. doi:10.1007/s11248-021-00274-7.
  • Eckerstorfer MF, Engelhard M, Heissenberger A, Simon S, Teichmann H. Plants developed by new genetic modification techniques—comparison of existing regulatory frameworks in the EU and non-EU countries. Front Bioeng Biotechnol. 2019;7:26. doi:10.3389/fbioe.2019.00026.
  • Kupferschmidt K. EU verdict on CRISPR crops dismays scientists. Washington, DC, USA: American Association for the Advancement of Science; 2018. doi:10.1126/science.361.6401.435.
  • Tuteja N, Verma S, Sahoo RK, Raveendar S, Reddy IBL. Recent advances in development of marker-free transgenic plants: regulation and biosafety concern. J Biosci. 2012;37(1):167–97. doi:10.1007/s12038-012-9187-5.
  • Greer SL, Trump B. Regulation and regime: the comparative politics of adaptive regulation in synthetic biology. Policy Sci. 2019;52(4):505–24. doi:10.1007/s11077-019-09356-0.
  • Molinari HBC, Silva N, Prado G, Lopes Filho J.V, Vieira L, Hugo Bruno Correa Molinari CNPAE, Letícia RV, Silva NVE, Prado GS, Lopes Filho JH. CRISPR technology in plant genome editing: biotechnology applied to agriculture. Embrapa Agroenergia-Livro científico (ALICE). 2021.
  • Waltz E. CRISPR-edited crops free to enter market, skip regulation. Nat Biotechnol. 2016;34(6):582–582. doi:10.1038/nbt0616-582.
  • Davison J, Ammann K. New GMO regulations for old: determining a new future for EU crop biotechnology. GM Crops & Food. 2017;8(1):13–34. doi:10.1080/21645698.2017.1289305.
  • Arpaia S, Birch ANE, Kiss J, van Loon JJA, Messéan A, Nuti M, Perry JN, Sweet JB, Tebbe CC. Assessing environmental impacts of genetically modified plants on non-target organisms: the relevance of in planta studies. Sci Total Environ. 2017;583:123–32. doi:10.1016/j.scitotenv.2017.01.039.
  • Whelan AI, Lema MA. Regulatory framework for gene editing and other new breeding techniques (NBTs) in Argentina. GM Crops & Food. 2015;6(4):253–65. doi:10.1080/21645698.2015.1114698.
  • Callaway E. CRISPR plants now subject to tough GM laws in European Union. Nature. 2018;560(7716):16–17. doi:10.1038/d41586-018-05814-6.
  • Kumar A. Application, Regulation, Ethical Concerns and Governance of Genome-Editing Technologies: an Overview. Asian Biotechnology & Development Review. 2019;21.
  • Menz J, et al. Genome edited crops touch the market: a view on the global development and regulatory environment. Front Plant Sci. 2020;21: p 11.
  • Jaffe G. The regulation of genome-edited crops, in Genome editing for precision crop breeding. Tylor & Francis, UK, London: Burleigh Dodds Science Publishing; 2021. pp. 279–321.
  • Zhang D, Zhang Z, Unver T, Zhang B. CRISPR/Cas: a powerful tool for gene function study and crop improvement. J Adv Res. 2021;29:207–21. doi:10.1016/j.jare.2020.10.003.
  • Tussipkan D, Manabayeva SA. Employing CRISPR/Cas Technology for the Improvement of Potato and Other Tuber Crops. Front Plant Sci. 2021;12:12. doi:10.3389/fpls.2021.747476.
  • Hahn F, Nekrasov V. CRISPR/Cas precision: do we need to worry about off-targeting in plants? Plant Cell Rep. 2019;38(4):437–41. doi:10.1007/s00299-018-2355-9.
  • Macovei A, Sevilla NR, Cantos C, Jonson GB, Slamet‐Loedin I, Čermák T, Voytas DF, Choi I-R, Chadha‐Mohanty P. Novel alleles of rice eIF4G generated by CRISPR/Cas9‐targeted mutagenesis confer resistance to Rice tungro spherical virus. Plant Biotechnol J. 2018;16(11):1918–27. doi:10.1111/pbi.12927.
  • Alqahantani N, Alotaibi B, Alshumrani R, Bamhrez M, Unver T, Tombuloglu H. CRISPR Applications in Crops. Oil Crop Genomics. 2021;367–81. Springer.
  • Ortigosa A, Gimenez-Ibanez S, Leonhardt N, Solano R. Design of a bacterial speck resistant tomato by CRISPR/Cas9-mediated editing of SlJAZ2. Plant Biotechnol J. 2019;17(3):665–73. doi:10.1111/pbi.13006.
  • 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. doi:10.3389/fpls.2018.00842.
  • Li R, Liu C, Zhao R, Wang L, Chen L, Yu W, Zhang S, Sheng J, Shen L. Crispr/cas9-Mediated SlNPR1 mutagenesis reduces tomato plant drought tolerance. BMC Plant Biol. 2019;19(1):1–13. doi:10.1186/s12870-018-1627-4.
  • Chandrasekaran J, Brumin M, Wolf D, Leibman D, Klap C, Pearlsman M, Sherman A, Arazi T, Gal-On A. Development of broad virus resistance in non‐transgenic cucumber using CRISPR/Cas9 technology. Mol Plant Pathol. 2016;17(7):1140–53. doi:10.1111/mpp.12375.
  • Zaman Q U, Raza A, Gill R Ali, Hussain M Azhar, Wang H Feng and Varshney R K. 2023. New possibilities for trait improvement via mobile CRISPR-RNA. Trends Biotechnol.2023. doi: 10.1016/j.tibtech.2023.05.001.