Next Article in Journal
Revisiting the Role of Astrocytic MAOB in Parkinson’s Disease
Previous Article in Journal
E2F1 Expression and Apoptosis Initiation in Crayfish and Rat Peripheral Neurons and Glial Cells after Axonal Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CRISPR-Based Genome Editing: Advancements and Opportunities for Rice Improvement

1
State Key Laboratory of Rice Biology, China National Rice Research Institute, Hangzhou 310006, China
2
Department of Agricultural Biotechnology, Institute of Biotechnology, University of Gondar, Gondar P.O. Box 196, Ethiopia
3
Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
4
Zhejiang Key Laboratory of Super Rice Research, Hangzhou 311400, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(8), 4454; https://doi.org/10.3390/ijms23084454
Submission received: 3 March 2022 / Revised: 8 April 2022 / Accepted: 8 April 2022 / Published: 18 April 2022
(This article belongs to the Section Molecular Plant Sciences)

Abstract

:
To increase the potentiality of crop production for future food security, new technologies for plant breeding are required, including genome editing technology—being one of the most promising. Genome editing with the CRISPR/Cas system has attracted researchers in the last decade as a safer and easier tool for genome editing in a variety of living organisms including rice. Genome editing has transformed agriculture by reducing biotic and abiotic stresses and increasing yield. Recently, genome editing technologies have been developed quickly in order to avoid the challenges that genetically modified crops face. Developing transgenic-free edited plants without introducing foreign DNA has received regulatory approval in a number of countries. Several ongoing efforts from various countries are rapidly expanding to adopt the innovations. This review covers the mechanisms of CRISPR/Cas9, comparisons of CRISPR/Cas9 with other gene-editing technologies—including newly emerged Cas variants—and focuses on CRISPR/Cas9-targeted genes for rice crop improvement. We have further highlighted CRISPR/Cas9 vector construction model design and different bioinformatics tools for target site selection.

1. Introduction

Agriculture is currently facing many obstacles such as global climate change, a rapidly growing population, pests and pathogens, and many environmental threats. The growing population makes food security an urgent, pressing issue all over the world, and there it is forecast that global food-supply demand will double from 2005 to 2050 [1]. Rice is considered the main dietary component or primary source of food for a large portion of the world’s population. Several crops, including rice, have been domesticated and harvested for many years to meet the increasing demand for food. Nonetheless, attempts to guarantee food supplies by using traditional breeding methods have shown little promise. There is a critical need for more powerful technologies to improve foods’ nutritional content, increase abiotic stress tolerance, and enhance resistance to major biotic factors [2].
Recent advances in gene-editing technologies (GET) can contribute to coping with the complex problem of traditional breeding techniques, beginning a new age of crop enhancement. These methods allow us to alter genes of interest at specific locations and to provide new outlooks into crop functional genomics. A genome-editing system is a robust tool looking into the biological and biotechnological aspects of the characterization of plant genes and genetic modification. Recent genome-editing tools, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat (CRISPR)-associated endonuclease 9 (CRISPR/Cas9) techniques have opened up a new horizon for rice yield and quality enhancement [3,4,5]—of which, CRISPR/Cas9 has shown the most prominent potential for quick and effective genome editing in plant species. CRISPR/Cas9 genome editing as an advanced molecular biology tool enables us to carry out precise and efficient target modification, identifying novel prospects for evolving new plant variations using deletions, insertions, and substitutions, and hence encouraging the hastening of rice crop enhancement. So far, genome-editing tools employing CRISPR/Cas9 have been successfully exploited in rice for the improvement of drought tolerance [6], cold tolerance [7], salinity, and heavy metal stress [8,9].
More recently, newly emerged Cas variants such as CRISPR/Cpf1 (clustered regularly interspaced short palindromic repeat from Prevotella and Francisella 1) were developed in 2016. This technique is a more sophisticated and effective gene-editing technology than CRISPR/Cas9 [10]. As a new approach option that complements the HDR method, the strategy of a CRISPR/Cas9-based base editing enables the conversion of one base to another directly and permanently without a double-stranded break or donor DNA [11,12]. Researchers have successfully designed adenine base editors (ABE) and demonstrated the efficiency of A–T to G–C conversion using the ABE system in the rice genome [13]. Similarly, other scholars also confirmed the efficiency and precision of ABE by changing the A–T target into G–C without any other insertions/deletions (InDels) or substitution mutations in target sites, indicating its high efficiency in multiplex base editing [14]. Recent research has employed a fusion of CRISPR-Cas9 and activation-induced cytidine deaminase to develop multiple herbicide-resistant mutants [15,16,17].
Due to its small genome size, high transformation efficiency, accessibility of gene resources, and increased genomic synteny, rice is a model crop for functional and structural genomics studies. Thus, rice can be effectively exploited to examine multiple classes of rice genome modification in the CRISPR/Cas9 system [18,19], as well as to determine the function of functional genes [20,21,22]. To date, many studies have been released on the potential application of CRISPR/Cas9 in rice [23,24,25,26,27,28,29,30]. However, there is a need for an elaborative review, keeping in mind the rapid and extensive acceleration of new studies on the CRISPR/Cas9 genome-editing tool. Therefore, this article aims:
  • To discuss the current application of CRISPR/Cas9 to rice research that focuses on CRISPR/Cas9-targeted genes for rice crop improvement;
  • To discuss CRISPR/Cas9 components and mechanisms;
  • To highlight CRISPR/Cas9 vector construction model design and different online tools for target site design;
  • To compare CRISPR/Cas9 with other gene editing tools (ZFNs and TALENs), including the newly emerged Cas variants.

2. CRISPR/Cas9 System and Its Components

Initially, CRISPR/Cas9 was discovered as an adaptive immune system of archaea and bacteria. It was first reported by [31], and researchers discovered a means of exploiting it as a gene-editing technology. This technique can identify a particular site in a target gene in a highly efficient, unique, and flexible manner [32]. In roughly 50% of bacteria and 90% of archaea, CRISPR/Cas9 active immune mechanisms exist [33]. CRISPR/Cas9 is an endonuclease of DNA that splits the invading phage DNA into pieces and then incorporates it into the CRISPR set as a spacer. It was implemented efficiently in plants in 2013, and in five original research articles, the CRISPR/Cas9 scheme in rice was efficiently recorded [4,19,34,35,36]. Consequently, CRISPR/Cas9 technologies have proved to be an essential genome-editing method for rice.
Nowadays, the editing system of CRISPR/Cas9 is the most common mechanism in plant biology for the genome editing process [37,38] because of its uncomplicatedness, flexibility, and efficiency. Two CRISPR/Cas9 components: the Cas9 protein and a short RNA molecule (sgRNA), together form a ribonucleoprotein complex (Figure 1). The sgRNA is made up of 18 to 21 nucleotides that are designed to target specific sites in the genome (protospacer). A G-rich (5′-NGG-3′) protospacer-adjacent motif (PAM) should be found downstream of the target site [31]. tracrRNA (trans-activating CRISPR RNA) and crRNA (CRISPR-derived RNA) are the two principal components of guide RNA. In essence, crRNA has homology area that enables it to integrate with tracrRNA. The tracrRNA has a stem-loop shape associated with the Cas9 protein. The crRNA and tracrRNA should be designed as sgRNA in the CRISPR/Cas9 gene-editing scheme to direct Cas9 dsDNA cleavage at the targeted site [31]. Following the appropriate site identification by sgRNA based on the Watson–Crick base-pairing rule, the Cas9-sgRNA complex moves through the genome and produces a double-stranded break (DSB) [33]. During cleavage site repairing, the error-prone non-homologous end-joining (NHEJ) pathway regularly leaves a lesion in the form of a minor InDel or substitution upstream of PAM. Such mutations can induce frame-shift mutations in the coding sequence of the gene and cause a premature stop codon, resulting in a loss or gain of function in mutants. This achievement has unlocked numerous other possibilities for scientists to obtain more knowledge on plant biological systems [39].

3. Mechanism of the CRISPR/Cas9 System

In three steps, namely adaptation/acquisition, expression, and interference, CRISPR/Cas9 identifies and targets the genetic material of foreign DNA [40,41,42,43]. The adaptation/acquisition phase includes the recognition, invasion, and binding of donor DNA that are cut into small segments and combined within the CRISPR locus. Then, the CRISPR locus is transcribed to create crRNA that directs the intended effect or endonucleases to attack viral items by complementary base pairing [44,45]. Since the protospacer contains a G-rich base pair (5′-NGG-3′), PAMs are used as recognition motifs for the adaptation/acquisition of the targeted site. During the second step of the CRISPR/Cas9 execution mechanism, the lengthy Pre-crRNA is deliberately transcribed from the CRISPR nucleus and reproduced into crRNAs using Cas9 proteins. Recently, researchers revealed that tracrRNA is also involved in Streptococcus pyogenes pre-crRNA processing [46]. The tracrRNA is associated with the repeat crRNA site through complementary base pairing and allows precrRNA to be processed in crRNA [47]. The processed crRNAs join the associated antiviral defense complex of CRISPR and help to identify and parse a particular target area of donor DNA [47]. At the final steps, i.e., interference, the system needs a Cas9 protein [48,49] so that the sgRNA can guide the cleavage of the Cas9 protein complex from the particular target area; this creates immunity from pathogen attacks [40,41].

4. The Advantages and Drawbacks of the CRISPR/Cas9 System

Until recently, the CRISPR/Cas9 tool was thought to be the best option for genome editing (GE) in plants, but it still has some drawbacks that limit its widespread application (Figure 2).
The following are a list of the major issues with CRISPR/Cas9 and the advantages of CRISPR/Cas variants:
  • The CRISPR/Cas9 system’s large size limits its editing efficiency, and it is not suitable for packing into viral vectors for delivery to somatic tissues. For efficient plant GE, a smaller-sized CRISPR/Cas is required;
  • SpCas9 involves a 5′-NGG-3′ PAM next to a 20 nt DNA target site where it only distinguishes the NGG PAM sequence, which limits its effectiveness when compared to new CRISPR/Cas variants. NG-Cas9 is more active and there is a newer variant, SpRYCas9 that is almost PAM-less. The broad PAM compatibility of SpRY greatly expands the targeting scope of CRISPR-based tools in plant genome engineering;
  • CRISPR/Cas9 has the potential to incorporate a large number of off-target mutations into the genome. However, by identifying various PAMs, new CRISPR/Cas variants have achieved better editing efficiency (fewer off-target mutations) of target bases in the sequence of interest;
  • CRISPR/Cas9 generates mutations at non-specific loci that are homologous to target sites;
  • CRISPR/Cas9-made mutant plants via Agrobacterium-mediated transformation systems are more expensive, time consuming, and resource intensive. The use of tissue culture-free genome editing systems, on the other hand, has the potential to improve efficiency;
  • The commercialization of transgenic crops expressing CRISPR/Cas9 faces challenges in a number of countries, owing primarily to development costs and constraints imposed by regulatory systems for the field release of genetically modified organisms.

5. Comparison of CRISPER/Cas9 Tools with ZFN and TALEN

Genome-editing methods are critical not only to explore gene functions in biology tasks but also to enhance the characteristics of genetic engineering in biotechnology. As shown in Table 1, we summarized the comparisons of CRISPER/Cas9 with ZFNs and TALENs.
ZFNs and TALENs are composed of DNA-binding protein and the enzyme FokI, whereas CRISPR binds together with Cas9 protein in the CRISPR/Cas9 system [23,50]. The DNA-binding domain of ZFNs or TALEs is comprised of a series of tandem repeat units; each one identifies and binds to one or more nucleotide targets on one strand of DNA. In the case of ZFN, each zinc finger repeat targets a triplet (3 nt), and yet internal and external context-dependent effects of adjacent fingers can influence the efficiency and specificity of the target domain relationships [18,51]. There is no accurate relationship, and several strategies have been developed to screen for and construct more efficient and specific ZFNs. TALE domains, on the other hand, exhibit a more predictable one-to-one correspondence between repeat units and their single-nucleotide targets, and no screening is required for TALEN engineering. Despite the fact that hundreds of ZFNs and TALENs have been constructed and tested, the targeting efficiency of these engineered endonucleases varies greatly. Indeed, the efficiency of TALENs in the same work or even targeting the same locus can range from low to high, or even show no targeting activity. Unfortunately, predicting the efficiency of a given ZFN of TALEN is still complicated, owing to a lack of sufficient knowledge about the properties of these engineered endonucleases. Through the comparison of all the evaluated TALENs or ZFNs, it may be possible to extract common rules regarding the efficiencies of these engineered endonucleases by analyzing the rapidly expanding information on these engineered endonucleases from a variety of sources. The specificity and potential toxicity of TALENs, on the other hand, require further careful exploration. ZFN technology is more mature in some applications, such as gene therapy trials, and it has recently been reported that ZFNs can be delivered directly into cultured cells in the form of purified proteins. Other factors in the engineering of customizable engineered endonucleases for TALENs and ZFNs, such as FokI cleavage domain variants and the relationship between the length of linker peptides and the length of spacers between the two engineered endonuclease monomer-binding sites can also influence the efficiency and specificity of these engineered endonucleases.
CRISPR/Cas9 gene-editing technologies continue to be an excellent tool in revolutionizing many areas of genome processing with their ease, effectiveness, and accuracy in the cleavage of target sites compared to TALEN and ZFNs [52,53]. In contrast to ZFN and TALEN, the specificity of the target DNA in CRISPR/Cas9 can be altered by programming the sgRNA sequence, and multiple sgRNAs can work with the same Cas9 protein for diverse targets at the same time [54]. Several papers have noted that effective changes have been carried out on several plant genes, but these techniques are comparatively tedious, costly, and only a few genes have been changed so far [55]. A CRISPR/Cas9 mechanism that was established using a Cas9 endonuclease and an RNA guide complex has demonstrated significantly higher effectiveness in gene editing. Additionally, CRISPR/Cas9 enables target identification by using gRNAs rather than DNA binding, which makes this method more reliable compared to ZFNs and TALEN [56]. While the specificity of CRISPR/Cas9 still requires further investigation, the frequency of off-target mutations is already less than that of physical and chemical mutagenesis methods [57].

6. Comparison of CRISPR/Cas9 with the Newly Emerging CRISPR/Cas GE Tools

Many recent studies have focused on improving the CRISPR/Cas9 system’s efficiency and accuracy by modifying it. New CRISPR/Cas tools (spCas9-NG, base editing, prime editing, xCas9, Cpf1, Cas13, and Cas14) are now being used for GE. These recently developed CRISPR/Cas variants with their potential applications are displayed in Figure 3 and Table 2.
The common Streptococcus pyogene Cas9 (SpCas9) recognizes canonical NGG PAM, limiting the rice genome’s editable range. Many studies have attempted to address this limitation by evaluating whether other Cas effectors (Cpf1 for AT-rich PAMs) and engineered Cas9 variants (VQR for NGA PAMs and VRER for NGCG PAMs) could be used in conjunction with other PAMs for rice genome editing [58]. Researchers have used stable transgenic lines to assess the efficacy of xCas9 and SpCas9-NG for gene editing in rice [59]. xCas9 was found to efficiently induce mutations at target sites in rice with NG and GAT PAM sequences.
Two types of xCas9 variants (xCas9 3.6 & xCas9 3.7) were created using PCR site-directed mutagenesis [60]. By creating 18 target sites containing GAA, GAT, and NG PAMs in three rice endogenous genes—MOC1, D14, and PDS—the feasibility of genome editing with xCas9 variants in rice was investigated [60]. The findings suggested that by utilizing some previously reported non-canonical PAMs, xCas9 could broaden the scope of genome editing in rice. The wild-type SpCas9 in the pCas9 (OsU6) vector was replaced with xCas9 3.6 and 3.7, yielding vectors pCas9 3.6 (OsU6) and pCas9 3.7 (OsU6). Overall, xCas9 3.7 outperformed xCas9 3.6 for genome editing in rice, demonstrating that xCas9 variants have the potential to become versatile tools that will broaden our understanding of genome editing.
SpCas9-NG, a SpCas9 variant rationally designed based on the structure of the SpCas9-sgRNA–DNA complex, has recently been found to recognize relaxed NG PAM sequences [61]. xCas9 and SpCas9-NG both recognize more relaxed PAM sequences when compared to SpCas9 [60,61]. xCas9 and SpCas9-NG variants that recognize relaxed NG PAMs can work more efficiently in rice, and SpCas9-NG is better suited for performing base editing in rice. The created versatile GE tools have significantly broadened the target scope in rice, benefiting basic plant research and crop genetic improvement [60]. xCas9, in general, has higher DNA specificity and editing efficiency, reduced off-target activity, and a greater PAM compatibility, unlike SpCas9 [60].
As demonstrated for rice [60] and multiple base mutations in rice [62], prime editing allows for the specific induction of insertions (up to 15 nt) and deletions (up to 40 nt). The pegRNA and fusion protein are introduced to the target cell, and once inside, the fusion protein nicks the cell’s DNA at the target sequence, initiating reverse transcription of the template sequence found in the pegRNA. The pegRNA and fusion protein are introduced into the target cell, and once inside, the fusion protein nicks the cell’s DNA at the target sequence, causing reverse transcription of the DNA template contained in the pegRNA to begin. As a result, an edited strand of DNA and an unedited strand of DNA are produced. The unedited strand is first removed, and then the newly edited strand is annealed back together to form double-stranded DNA.
Cas13 is a newly discovered CRISPR effector that can target specific viral RNAs and endogenous RNAs in plant cells [63]. Cas13 has a high level of RNA target specificity and efficiency [64]. Cas13 has been used to direct ADAR2 deaminase in human cells for RNA modification (changing adenosine to inosine) in order to recover functional proteins and halt disease progression [65]. CRISPR/Cas13a has recently been identified as an entirely new CRISPR type that belongs to class II type VI. It is attributed with RNase activity due to the existence of higher eukaryote and prokaryote nucleotide-binding (HEPN) domains [65].
Cas12a (Cpf1) and Cas12b (C2c1) from the class 2 type V CRISPR/Cas system, in addition to the Cas9 proteins, have been repurposed for GE [66]. They have many characteristics that distinguish them from Cas9 proteins; Cas12a and Cas12b recognize AT-rich PAM sequences, in contrast to Cas9, which recognizes GC-rich PAM sequences [66]. Cpf1 is dependent on a T-rich PAM sequence at the 5′-end of the protospacer sequence (5′-TTTN-3′ or 5′-TTTV-3′; V can be A, C, G). While Cas9 generates blunt-ended DNA breaks, Cpf1 generates DSBs with staggered ends at the distal position of a PAM, which may provide additional advantages—particularly for knock-in strategies—and may increase the effectiveness of NHEJ-based gene insertion [67].
Furthermore, Cpf1 is regarded as a better GE tool than CRISPR/Cas9 as it only demands a 42 nt crRNA, whereas Cas9 demands a 100 nt gRNA; meanwhile, Cpf1-mediated GE needs short sgRNA sequences [68]. Cpf1 proteins, which have RNase activity, have been used to process crRNA arrays for GE in plants [69]. These characteristics improve insertion efficiency at the Cpf1-cleaved site [70]. Recently, an improved Acidaminococcus sp. Cas12a variant (enAsCas12a) with a significantly expanded targeting range was engineered, allowing the targeting of previously inaccessible PAMs. When compared to sites with canonical TTTV PAMs, enAsCas12a exhibits a two-fold increase in GE activity. Cas14a, in contrast to other known class II systems, does not require a flanking sequence (PAM) near the target site [71]. In vitro validation of the PAM requirement demonstrated that Cas14a can cleave target sites regardless of the different sequences adjacent to the targets of these different guides [72].
Base editing (BE) is a remarkably new and diverse GE system that incorporates accurate and highly predictable nucleotide changes at genomic targets without the need for donor DNA templates, DSBs, or reliance on HDR and NHEJ [73]. It is thought to be more effective than HDR-mediated base-pair substitution because it results in fewer unwanted mutations at the target locus [74]. BE3 [75], BE4 [76], targeted-AID [70], and dCpf1-BE [77] are among the BE systems currently used to edit plant and animal genetic material. Such systems use Cas9 or Cpf1 variants to recruit cytidine deaminases, which use DNA mismatch repair pathways to generate specific C to T alterations. Furthermore, when directed by sgRNAs to genomic targets in human cells, the adenine base editors, which were created by fusing an evolved tRNA adenosine deaminase with SpCas9 nickase (D10A), convert A–T to G–C [78].
Table 2. Different newly emerged CRISPR/Cas techniques with potential functions in associated hosts.
Table 2. Different newly emerged CRISPR/Cas techniques with potential functions in associated hosts.
Class NameSize(AA)PAMHostSpacer (bp)Cut SiteTargetRef
AacC2c11277T-rich PAMAlicyclobacillus acidoterrestris20Upstream of PAMDs DNA[79]
CjCas9984NNNNACAC and NNNRYACCampylobacter jejuni22Upstream of PAMDsDNA[80]
Cpf1TTTVPrevotella & Francisella 120Downstream of PAMDsDNA[67]
Cpf1(AsCpf1)13075′-TTTN-3′Acidaminococcus sp.24Downstream of PAMDsDNA[81]
Cas12a-Thymine-rich PAM sequencesAcidaminococcus sp.-Downstream of PAMDs DNA[69]
Cas131440Non-G nucleotide at the 3′orthologs; Leptotrichia shaii28-ssRNA[82]
Cas14400–700-Uncultivated archaea--ssDNA[71]
FnCas916295′-NGG-3′Francisella novicida20Upstream of PAMDs DNA[83]
Nme Cas910825′ NNNNGATT-3′Neisseria meningitidis24 and 20Upstream of PAMDsDNA[84]
SaCas910535ʹ-NNGRRT-3′Staphylococcus aureus21Upstream of PAMDsDNA[85]
SpCas913685′-NGG-3′Streptococcus pyogenes-Upstream of PAMDsDNA[31]
SpCas9-NG–-5′-NAC, NTG, NTT, and NCGS. pyogenes-Upstream of PAMDsDNA[86]
St1Cas91121NNAGAAWStreptococcus thermophilus20Upstream of PAMDsDNA[87]
St3Cas914095′-NGGNG-3′S. thermophilus20Upstream of PAMDsDNA[56]
xCas9GAA, GAT and NG-19–22Upstream of PAMDsDNA[88]
‘–’: information unavailable.

7. CRISPR/Cas9 Vector Design for Rice Genome Editing

CRISPR/Cas9 vector design and construction are comparatively easy and cheap. Components of the CRISPR/Cas9, namely crRNA and tracrRNA, together make up the sgRNA that directs Cas9 to implement target-specific DSBs. Effective CRISPR/Cas9 designing involves the transfer of both Cas9 and sgRNA proteins into target cells. They can be delivered via an agent (vectors) or directly (electroporation). The techniques of transmission in crops for gene editing using CRISPR/Cas9 approaches have been comprised of polyethylene glycol-mediated transformation, ballistics techniques, and Agrobacterium-mediated gene transformation [4,89,90]. For effective genome editing, it is essential to select ideal promoters to obtain the expression of sgRNA and Cas9 and codon-optimizedCas9. Codon-optimized SpCas9 variants in eukaryotic organisms have generally been used. Nevertheless, some studies have used the Cas9 option to optimize crop codons [4,35,89,90]. However, using RNPs or RNA, no optimization of elements is necessary for good expression (e.g., promoter, terminator, or codon optimization, etc.).
In many crops, the Cas9 expression is driven by by EF1A (Elongation factor 1-alpha), CMV (Cauliflower Mosaic Virus), or LTR (Long terminal repeats) promotors. Among these, 35S CMV (cauliflower mosaic virus) is the most common expression promoter [37]. The targeted gene for CRISPR/Cas9 needs a customized single guide RNA (sgRNA), which has a crRNA sequence and tracrRNA [31]. The crRNA structure gives the specificity of the targeted DNA, and it is feasible to design sgRNAs for multiplex gene editing with variable crRNAs. The crRNA region is a 20-nucleotide sequence homologous to the part of the gene that you are interested in, and that directs the activities of the Cas9 nuclease [91]. The target DNA sequence is, therefore, 20 bp followed by a PAM (NGG/NAG) sequence. However, DNA targets and crRNAs that show a discrepancy from the recognized 20 bp length have also been reported in a few studies, with length variations ranging from 19 to 22 bp [4,19,35,37]. Consequently, (N) 19–22 NGG or (N) 19–22 NAG is the target DNA sequence. Nevertheless, CRISPR/Cas9 may be restricted by the accessibility of the PAM site when targeting a specified sequence [92]. The rice genome demonstrates an abundance of potential PAM (1 in 10 bp) sites [35], which allows CRISPR technology to be used rapidly in the rice genome.
The U6 and U3 plant RNA III promoters have been used for sgRNA expression [37]. These promoters have to initiative nucleotide “G” and “A” transcriptions, respectively. Thus, the sgRNA crRNA structure for the promoter U6 is G (N) 19–22, and the promoter U3 is A (N) 19–22. The initiating G and A are often fused directly to the gRNA or are part of the target sequence [4,19,35,36,37,90].
Here we outline a procedure for performing targeted editing employing the CRISPR/Cas9 system (Figure 4). The first procedure is the selection of a target site to be placed at the short PAM sequence at its 3′ end or downstream of its protospacer. The choice of the target site is an essential focus of large-scale CRISPR/Cas9 technology applications. The choice of the appropriate target site(s) can achieve the lowest/no off-target effects at an important mutation site. Many bioinformatics online tools for the designing of sgRNA as well as the identification of off-target effects are available in model organisms, including rice crops [93].
Following the identification of a target site, the next step is to design the target site-related oligonucleotide or primers. A DNA fragment encoding the sgRNA scaffold, which is placed under the appropriate promoter to optimize expression, is fused into the developed primers. For cloning, appropriate sequences of adaptors should be included. The inverse primary should have a 5′-AAAC-3′ adaptor, while the forward primer must have 5′-GGCA-3 adaptors (Figure 4). Once the protospacer sequence is subcloned into an SK-sgRNA vector or cassette (an example vector for our case study), the next step is inserting the constructs into the Cas9 expression vector (pC1300 Cas9), followed by its delivery into the rice plant cells by appropriate methods such as Agrobacterium transformation, protoplast transformation, or callus bombardment.

8. Important Considerations for gRNA Selection

The best techniques for gRNA selection—through experiment development, execution, and evaluation, are always essential to the correct use of CRISPR technology. The best sgRNA thus relies a great deal on what one is attempting to do when developing sgRNAs for use in the CRISPR technique: gene knockout, a editing of a particular base, or gene expression modulation.
The following variables should be regarded for the design of sgRNAs:
  • The DSB should be introduced near to the 5′ end of the coding region or in the indispensable domains. If choosing a target sequence in the proximity of the adjacent protospacer motif (PAM), the 3′ end of the target sequence must have a PAM sequence (5′-NGG-3′; Figure 4). The target sequence (crRNA) should be upstream of the PAM structure, and the Cas9 nuclease will digest around three bases upstream of the PAM. For cleavages, the PAM sequence is compulsory; however, it is not a component of the sgRNA sequence and should, therefore, not be included in the sgRNA sequence itself.
  • When selecting the right target sequence for on-target activity, targets that have poly-T and very low or very high GC content (≤25% or ≥80%) have low editing efficiency. Similarly, target site(s) with eight or more adjacent nucleotides have low editing effectiveness and should be coupled to the sgRNA sequence [94].
  • Another consideration before the start of the CRISPR experiment is decreasing off-target effects. It is not always so important in what the location the gene target is, but the gRNA sequence must be designed to be highly active and minimize off-target sites. Potential off-target sites with higher scores may have a higher probability of being targeted by the sgRNA/Cas9 nuclease complex. Thus, the selected gRNA spacers/target sequence should have sufficient specificity to avoid off-target editing.
In general, designing the CRISPR/Cas9 vector for any experiment needs to balance the maximization of on-target activity while minimizing off-target activity, which sounds obvious.

9. Bioinformatics Tools Available for sgRNA Designing

Many bioinformatics tools for sgRNA designing with high specificity and off-target detection in model organisms have been created (Table 3). After considering SNP and/or InDel in the genome, a unique platform for selecting sgRNA with significant effects is required [95]. More recently, highly specific sgRNAs with few or no off-target results were executed in rice, and a CRISPR–GE platform has been developed [94].
At present, the technique is being used as a powerful and multipurpose technology for genome-engineering, such as editing “changing the genomic sequence” [56,96] and regulation “repressing or activating expression of genes” [97]. For accurate and programmable gene targeting, a single guide RNA (sgRNA) is needed [75]. Practical and specific gene editing demands a detailed layout of sgRNAs, which is still a significant task. In this review, we listed some of the computational tools (Table 3) that have been used to assist in the design of sgRNAs for the CRISPR/Cas9 editing system.

10. Applications of CRISPR/Cas9 Gene-Editing Technologies in Rice

To keep pace with food consumer demand, the potential increase of rice yield and quality depends on the integration of multiple modern breeding approaches, comprehensive agronomic practices, and appropriate social and economic policies to simulate crop production activities. Therefore, the enhancement of rice yield and quality using revolutionary breeding methods is instantly desirable in order to raise access to nutritious foods at a global level for the ever-increasing population. Recently, many research works have shown the application of CRISPR/Cas9 genome editing strategies in rice, covering several aspects from biotic stress resistance to abiotic stress tolerance, and the achievement of improved yield performance, biofortification, and improvements in quality (Table 4). In this paper, the potential applications of CRISPR/Cas9 in rice genome editing are taken into consideration in attaining a large number of objectives.

10.1. Improving Rice Yield and Quality

Improving the quality of food is important as many individuals depend on rice as their basic sustenance. However, improvements via conventional methods have limitations in terms of being time consuming and challenging, causing off-target changes, the involvement of undesirable traits, and their lower efficiency. CRISPR/Cas9 is a useful tool for knocking out negative regulators controlling yield-associated characteristics of rice, such as grain weight (OsGW5, OsGLW2), grain size (OsGS3), grain numbers (OsGn1a), tiller number (OsAAP3), and panicle size (OsDEP) [120,123,145,146]. The simultaneous knocking out of three rice grain weight genes, namely TGW6, GW2, and GW5, resulted in a significant enhancement of grain weight characteristics [22]. Since most yield-related components are quantitative and influenced by the environment, the knockout of individual variables may not be enough to boost yield. Recently, it was discovered that editing the OsSPL16 gene with CRISPR/Cas9 improves grain yield by modulating the expression of pyruvate enzymes and cell cycle proteins [147].
Improving rice grain quality characteristics via current advanced genome-editing tools is a rapid, feasible, and economical approach [148]. Several rice genes that determine grain quality appearance have been isolated. Grain appearance and chalkiness are among the quality traits that impact the market adequacy of rice [149,150]. The integration of functionally characterized genes such as OsSPL13, OsSPL16/GW8, Chalk5, and GW7 using CRISPR/Cas9 and the assessment of their relationships with other genes can significantly enhance our understanding of rice grain appearance and milling quality.
The Waxy gene in the endosperm is primarily responsible for the amylose content (AC), which determines the cooking and eating quality of rice grains [151]. Waxy was knocked out using CRISPR/Cas9, resulting in lower amylose content and, as a result, improved rice quality for food and high cooking efficiency [138,152]. Several studies have also been conducted to determine the roles of various enzymes or genes involved in transgenic Taichung65 rice plants comprising a Waxy antisense construction, which had lower AC; transgenic plants were also demonstrated to have lower AC levels in hybrids [153]. The SBEIIb gene has also been knocked out via CRISPR/Cas9 to create high amylose rice, which is an ideal food for patients with diet-associated noninfectious diseases [139]. The successful editing of the BADH2 gene, responsible for fragrant aromatic rice, involved the insertion of one nucleotide in the mutated line, which showed a significant amount of 2AP and enriched the fragrance of the rice [127]. Protoplast and particle-bombarded rice calli systems were used to validate sequence-specific CRISPR/Cas9-mediated genomic alteration of three rice genes, OsPDS, OsBADH2, and OsMPK2, which controlled the responses to various abiotic stress stimuli [4]. Editing rates of approximately 7% and 9% for OsBADH2 and OsPDS were observed, respectively. Similarly, the five carotenoid catabolic genes, namely; OsCYP97A4, OsDSM2, OsCCD4a, OsCCD4b, and OsCCD7, have been successfully knocked out to improve the quantity of β-carotene in rice endosperm [154].

10.2. CRISPR/Cas9 Systems in Developing Climate Resiliance

Climate-resilient farming for contesting biotic and abiotic stress is an upcoming crop enhancement of interest using genome editing approaches [155,156]. The CRISPR/Cas9 RNA-led method is regularly used to improve plants, but it has been considered in very few papers so far in producing climate-resilient plants. The primary influences on crop yield and performance are stresses. Many crops, including fungal, bacterial, and viral resistance and insects with enhanced biotic stress resistance, were acquired by the knockout of CRISPR/Cas9. Scientists have moved from proof-of-concept to more applicative uses of CRISPR/Cas9 in rice. Multiple disease resistance lines have been obtained via this technology. Blast is a critical fungal disease in rice, but resistance genes against this disease have been developed by targeting the OsERF922 gene [20]; these findings revealed that there was a considerable decrease in blast lesion formation under pathogen infection. Similarly, knocking out of the Bsrk-1 gene enhanced the resistance of rice against blast without compromising yield [128]. The knocking-out of OsSWEET13 resulted in crops that resist rice bacterial blight caused by Xanthomonas oryzae pv. oryzae (Xoo) [129]. A recent study [157] has discovered that OsCYP71A1 disruptively blocks serotonin biosynthesis with considerably higher concentrations of salicylic acid, giving resistance to planthoppers and stem borers–two of the most rice-damaging pests.
Contamination of arable lands is one of the abiotic stresses that have generated the need to avoid the buildup of poisonous heavy metals in plants. By knocking out OsARM1, OsNramp5, and OsHAK1, researchers were able to identify rice species with high cadmium and arsenic concentrations [8,136,158]. In 2018, a study of the OsPYL abscisic acid receptor family showed that the triple knockout of ply 1/4/6 by CRISPR/Cas9 produced improved grain output, enhanced tolerance to high temperatures, and lower pre-harvest sprouts [124]. The suppression of the cadmium (Cd) OsNramp5 transporter gene resulted in the growth of low-Cd hybrid rice populations. CD accumulation in roots, seeds, and shoots was reduced by mutant osnramp5 [8].
Scientists have also been able to generate CRISPR/Cas9-mediated herbicide-resistant rice plants [133]. Studies into herbicide resistance were started to guarantee public and environmental health, as both are affected by agrochemical use [159]. ALS1 is one of the essential enzymes accountable for herbicide resistance in rice. Its function was disturbed using CRISPR/Cas9 at multiple discrete points [133], and the results showed that homology-directed repair mediated by CRISPR/Cas9 was effective. The second exon of BEL was knocked out by CRISPR/Cas9 in the rice cultivar Nipponbare, which was associated with bentazon and herbicide resistance [21]. CRISPR/Cas9 was used to edit TIFY1b [160], a transcription factor, and the OsAnn3 gene [7], which considerably improved cold tolerance. However, a great potential remains to be utilized; there is a considerable deal of plant genome manipulation that has been carried out through genome editing using the CRISPR/Cas9 system. Resistance to imazapic (IMP) and imazethapyr (IMT) was conferred in rice mutant plants by CRISPR/Cas9-mediated knockout of the Acetolactate Synthase (OsALS) gene [161]. In general, the CRISRR/Cas9 system has been successfully used to improve abiotic stress tolerance in rice [162].

10.3. Practical Approach for Hybrid Seed Production

Hybrid breeding or heterosis is a powerful approach used in increasing the productivity and yield of crops. Evolving a male sterile maternal line is a precondition required for developing a high-quality hybrid variety [163]. Hence, marvelous achievements have been achieved via CRISPR/Cas9-mediated gene knockout in the development of rice male sterile lines, including thermosensitive male sterile tms5 lines [121,164], photosensitive genic male sterile csa rice [123], and TMS10 [122]. Rice photosensitive and thermosensitive male sterile lines have been developed to accelerate and exploit heterosis breeding [121,165]. However, hybrid sterility is a crucial bottleneck in the exploitation of heterosis in rice breeding. To overwhelm the generative barriers, SaF/SaMatSa and OgTPR1 at the S1 locus [166] were disrupted in japonica–indica hybrids. The recovery of male fertility in japonica–indica hybrids was demonstrated through the knockout of one or two copies of the Sc gene in the indica allele Sc-I [167]. TMS10, created by genetic crosses or genome editing, is a temperature-sensitive male sterile mutant applicable to hybrid seed production [122]. The results provide insights into how rice copes with adverse changes in temperature to attain normal male fertility and a new breeding line for rice hybrid seed production. Correspondingly, knockout of the ORF2 toxin gene, which is accountable for the newly revealed selfish gene suicide mechanism of rice, rescued the fertility of japonica–Indica hybrids [168].
Another obstacle for exploiting hybrid breeding is that heterosis is lost in the following generations because of genetic segregation. Besides this, the high cost of hybrid seed production hinders the application of heterosis in many crops, including rice. Two independent groups [169,170] used a multiplex CRISPR/Cas9 editing system to replace meiosis by mitosis in rice through the knockout of three essential meiotic genes: REC8, PAIR1, and OSD1. They developed asexual propagation lines either by simultaneous activation of BBM1 in the egg cell or by knocking out MTL, respectively, enabling the repair of heterozygosity of hybrids through clonal seed propagation. The study results showed that the MiMe phenotype could be quickly introduced into hybrid rice varieties using CRISPR/Cas9 genome-editing technologies.

10.4. Rapid Generation of Genetic Diversity

Furthermore, CRISPR/Cas9 applications include comprehensive genetic diversity studies. Germplasm is a fundamental tool for the enhancement of crops in sustainable farming. The main requirement of the plant breeder is the generation of trait-specific genetically varied relatives for improving the characteristics of the crop. The main objective of breeders is to concentrate studies into the collection, preservation, and identification of new elite germplasms, which eventually produce agriculturally superior cultivars with a broad genetic foundation in breeding programs.
Genetic diversity is an essential cause of plant characteristic enhancement. The creation of genetic variables in the gene pool is the precondition for the development of distinctive plant crops. Transgenes in the enhanced variations can be removed once the necessary changes have been made. Traditional plant improvement methods rely heavily on genetic differences between natural germplasms. The introduction of positive characteristics into the chosen germplasm necessitates a chronological overview, as well as a time and energy-intensive evaluation of the vast population [148].
Although mutation breeding methods can be significantly accelerated and directed towards more than 2250 new plant variants [171], subsequent modifications in the mutated genes are random. This is why plant biologists have sought methods that can easily edit required genes of interest. Hence, reverse genetic methods increase the progress of crop improvement through targeted gene editing, and the CRISPR/Cas9 gene-editing system is an advancing approach for the rapid introduction of population diversity in rice crop breeding and development [47].
CRISPR/Cas9 now makes it simple to edit the multiplex genome of a possibly limitless number of genes [172] and has been recently verified in Arabidopsis and rice [173,174,175]. Eight rice genes such as BADH2, DEP1, Gn1a, GS3, GW 2, Hd1, EP3, and LPA1 have been successfully edited using one binary vector [174]. They provided a strategy trial for the rapid generation of various breeding resources, targeting these agronomically essential rice genes, using CRISPR/Cas9 multiplex genome-editing technology. DEP1, EP3, Gn1a, GS3, and GW2 are yield-associated genes; LPA1 is a plant architecture gene; while BADH2 and Hd1 are associated with rice fragrance and photoperiod genes, respectively. The knockout of genes that have an enormous effect on grain yields, such as GS3, DEP1, GS5, GW2, Gnla, and TGW6, is an easy and direct method for average rice output. The mutants of these genes produce the desired, convincing phenotypes [120,176]. The thousand-grain weight (TGW) increased significantly through the development of triple rice mutants by the simultaneous knocking out of GW2, GW5, and TGW6 [22]. Four genes were recognized as panicle architecture, plant architecture, seed size, and grain number regulators by a study team centered in Guangzhou (China), demonstrating that various agronomically suitable characteristics can be rapidly enhanced in a single cultivar [120].
CRISPR/Cas9 could also provide an efficient pyramid breeding technique with the simultaneous alteration of several characteristics [23,177]. Consequently, CRISPR/Cas9 is a potential tool for feeding the world’s population and achieving the goal of zero global hunger [178]. The insertion of big sequences via NHEJ would enable transgenes to be introduced into a specified locus, which supports high-level transcripts and does not interfere with endogenous gene function. Site-specific nucleases allow the addition of many genes near the current transgenic nucleus to achieve focused molecular feature stacking. It also allows the introduction of several characteristics in plants that have a small degree of segregation, which is hard to achieve through standard breeding or even genetic engineering [179]. There is also a tremendous potential for CRISPR screens to map and interrogate gene regulatory networks at an unprecedented speed and scale. The implementation of CRISPR screens offers new opportunities to analyze plant genomes at deeper resolution and will greatly advance plant functional and synthetic biology [180]. Researchers [181] have developed genomic resources and efficient transformations in the orphan Solanaceae crop ‘groundcherry’ (Physalis pruinosa) and have used CRISPR–Cas9 to mutate orthologues of tomato domestication and improvement genes that control plant architecture, flower production, and fruit size, thereby improving these major productivity traits. Accordingly, translating knowledge from model crops enables the rapid creation of targeted allelic diversity and novel breeding germplasms in distantly related orphan crops. More importantly, promoting integrative ecological (biodiversity resources) genomic studies promises a better understanding of antagonistic co-evolutionary interactions, as well as the more efficient breeding utilization of resistant phenotypes. In this case, gene prediction may also go beyond pre-breeding efforts and feedback on restoration optimization [182].

11. Conclusions Remarks

The progress of science is dependent on new techniques, discoveries, and ideas, so the development of novel tools and techniques is essential for scientific advancement. Plant breeding has been transformed by the advancement of genome-editing technologies. Continuous innovation in crop breeding and genetics is essential in meeting challenges and achieving sustainable food production. CRISPR, a cutting-edge molecular biology technique, has already broadened our understanding of genome regulation and organization in living cells from various biological kingdoms. CRISPR is revolutionizing not only agriculture, but also industry, the environment, medicine, and other fields. Due to the relative specificity of each nuclease platform, the majority of research work using the CRISPR/Cas9 genome-editing technique has been groundwork/preliminary to date. In the future, the use of high-throughput methods that allow for the comprehensive profiling of off-target cleavage sites should provide insight into the target recognition stringency inherent in each system. As a result, more advancement is required to fully exploit the platform, which will result in increased on-target efficiency. Many strategies, including modifying Cas9 to recognize different PAM sequences, have been used to overcome this limitation. The xCas9 3.7 variant was created to broaden the scope of genome editing in rice. A new version of Cas9 (SpCas9) was also created that could recognize NG PAMs in rice. Many other such variants are ineffective in plants, emphasizing the need to develop more Cas9 variants capable of recognizing a wide range of PAMs. The limitation imposed by PAM specificity can also be overcome by using the newly discovered Cas14a system, which does not require PAM, but can only target ssDNA. As a result, the rapid advancement of research into epigenetic genome modifications in rice is a promising approach for future rice crop improvement. As the scope and power of CRISPR technologies expand, social and ethical concerns about their use grow, and the applications of these powerful tools deserve more thought.

Author Contributions

W.A.Z. contributed to this work by writing the article. M.T. revised the manuscript. Validations and funding acquisition were performed by Y.Z. and L.C. All authors have read and agreed to the published version of the manuscript.

Funding

The work was funded by the National Natural Science Foundation of China (31961143016, 31521064), the National Key Research and Development Program (2016YFD0101801), and the Agricultural Science and Technology Innovation Program of the Chinese Academy of Agricultural Science (CAAS-ASTIP-2013-CNRRI).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors state that there is no conflict of interest.

Abbreviations

2AP:2-Acetyl-1-Pyrroline; A: Adenine; ABE: Adenine Base Editors; APOBEC: apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like; bp: Base Pair; C: Cytosine; Cd: Cadmium; CMV: Cauliflower Mosaic Virus; CRISPR/Cpf1: Clustered Regularly Interspaced Short Palindromic Repeat From Prevotella And Francisella 1; CRISPR: Clustered Regularly Interspaced Short Palindromic Repeat; Cas9: CRISPR Associated Endonuclease 9; crRNA: CRISPR Derived RNA; DSBS: Double-Stranded Break; G: Guanine; GET: Gene-Editing Technologies; HDR: Homology-Directed Repair; InDel: Insertion and or Deletion; N: Nucleotide; NHEJ: Non-Homologous End Joining; PAM: Protospacer Adjacent Motif; T: Thiamine; TALENs Transcription activator-like Effector Nuclease; TracrRNA: Trans-Activating CRISPR RNA; ZFNs: Zinc Finger Nuclease.

References

  1. Tilman, D.; Balzer, C.; Hill, J.; Befort, B.L. Global food demand and the sustainable intensification of agriculture. Proc. Natl. Acad. Sci. USA 2011, 108, 20260–20264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Rashid, B.; Tariq, M.; Khalid, A.; Shams, F.; Ali, Q.; Ashraf, F.; Ghaffar, I.; Khan, M.I.; Rehman, R.; Husnain, T. Crop improvement: New approaches and modern techniques. Plant Gene Trait 2017, 8, 18–30. [Google Scholar] [CrossRef]
  3. Li, T.; Huang, S.; Zhao, X.; Wright, D.A.; Carpenter, S.; Spalding, M.H.; Weeks, D.P.; Yang, B. Modularly assembled designer TAL effector nucleases for targeted gene knockout and gene replacement in eukaryotes. Nucleic Acids Res. 2011, 39, 6315–6325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shan, Q.; Wang, Y.; Li, J.; Zhang, Y.; Chen, K.; Liang, Z.; Zhang, K.; Liu, J.; Xi, J.J.; Qiu, J.L.; et al. Targeted genome modification of crop plants using a CRISPR-Cas system. Nat. Biotechnol. 2013, 31, 686–688. [Google Scholar] [CrossRef] [PubMed]
  5. Wood, A.J.; Lo, T.W.; Zeitler, B.; Pickle, C.S.; Ralston, E.J.; Lee, A.H.; Amora, R.; Miller, J.C.; Leung, E.; Meng, X.; et al. Targeted genome editing across species using ZFNs and TALENs. Science 2011, 333, 307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lou, D.; Wang, H.; Liang, G.; Yu, D. OsSAPK2 confers abscisic acid sensitivity and tolerance to drought stress in rice. Front. Plant Sci. 2017, 8, 993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. 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 Biotechnol. 2017, 60, 539–547. [Google Scholar] [CrossRef]
  8. Tang, L.; Mao, B.; Li, Y.; Lv, Q.; Zhang, L.; Chen, C.; He, H.; Wang, W.; Zeng, X.; Shao, Y. Knockout of OsNramp5 using the CRISPR/Cas9 system produces low CD-accumulating indica rice without compromising yield. Sci. Rep. 2017, 7, 14438. [Google Scholar] [CrossRef] [Green Version]
  9. Bo, W.; Zhaohui, Z.; Huanhuan, Z.; Xia, W.; Binglin, L.; Lijia, Y.; Xiangyan, H.; Deshui, Y.; Xuelian, Z.; Chunguo, W.; et al. Targeted mutagenesis of NAC transcription factor gene, OsNAC041, leading to salt sensitivity in rice. Rice Sci. 2019, 26, 98–108. [Google Scholar] [CrossRef]
  10. Endo, A.; Masafumi, M.; Kaya, H.; Toki, S. Efficient targeted mutagenesis of rice and tobacco genomes using Cpf1 from Francisellanovicida. Sci. Rep. 2016, 6, 38169. [Google Scholar] [CrossRef] [Green Version]
  11. Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420. [Google Scholar] [CrossRef] [Green Version]
  12. Marx, V. Base editing a CRISPR way. Nat. Methods 2018, 15, 767–770. [Google Scholar] [CrossRef]
  13. Li, H.; Qin, R.; Liu, X. CRISPR/Cas9-mediated adenine base editing in rice genome. Rice Sci. 2019, 26, 125–128. [Google Scholar]
  14. Hua, K.; Tao, X.; Yuan, F.; Wang, D.; Zhu, J.K. Precise A·T to G·C base editing in the rice genome. Mol. Plant. 2018, 11, 627–630. [Google Scholar] [CrossRef] [Green Version]
  15. Lu, Y.; Zhu, J.K. Precise editing of a target base in the rice genome using a modified CRISPR/Cas9 system. Mol. Plant. 2017, 10, 523–525. [Google Scholar] [CrossRef] [Green Version]
  16. Shimatani, Z.; Kashojiya, S.; Takayama, M.; Terada, R.; Arazoe, T.; Ishii, H.; Teramura, H.; Yamamoto, T.; Komatsu, H.; Miura, K.; et al. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 441–443. [Google Scholar] [CrossRef]
  17. Zong, Y.; Wang, Y.P.; Li, C.; Zhang, R.; Chen, K.L.; Ran, Y.D.; Qiu, J.L.; Wang, D.; Gao, C. Precise base editing in rice, wheat, and maize with a Cas9-cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 438–440. [Google Scholar] [CrossRef]
  18. Li, T.; Liu, B.; Spalding, M.H.; Weeks, D.P.; Yang, B. High-efficiency TALEN-based gene editing, produces disease-resistant rice. Nat. Biotechnol. 2012, 30, 390–392. [Google Scholar] [CrossRef]
  19. Feng, Z.; Zhang, B.; Ding, W.; Liu, X.; Yang, D.-L.; Wei, P.; Cao, F.; Zhu, S.; Zhang, F.; Mao, Y.; et al. Efficient Genome Editing in Plants Using a CRISPR/Cas System. Cell Res. 2013, 23, 1229–1232. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, F.; Wang, C.; Liu, P.; Lei, C.; Hao, W.; Gao, Y.; Liu, Y.G.; Zhao, K. Enhanced rice blast resistance by CRISPR/Cas9- targeted mutagenesis of the ERF transcription factor gene OsERF922. PLoS ONE 2016, 11, e0154027. [Google Scholar] [CrossRef]
  21. Xu, R.; Li, H.; Qin, R.; Wang, L.; Li, L.; Wei, P.; Yang, J. Gene targeting using the Agrobacterium tumefaciens-mediated CRISPR-Cas system in rice. Rice 2014, 7, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Xu, R.; Yang, Y.; Qin, R.; Li, H.; Qiu, C.; Li, L.P.; Wei, P.; Yang, J. Rapid improvement of grain weight via highly efficient CRISPR/Cas9-mediated multiplex genome editing in rice. J. Genet. Genom. 2016, 43, 529–532. [Google Scholar] [CrossRef] [PubMed]
  23. Bortesi, L.; Fischer, R. The CRISPR/Cas9 system for plant genome editing and beyond. Biotechnol. Adv. 2015, 33, 41–52. [Google Scholar] [CrossRef] [PubMed]
  24. Khatodia, S.; Bhatotia, K.; Passricha, N.; Khurana, S.M.P.; Tuteja, N. The CRISPR/Cas genome-editing tool: Application in Improvement of Crops. Front. Plant Sci. 2016, 7, 506. [Google Scholar] [CrossRef] [Green Version]
  25. Arora, L.; Narula, A. Gene editing and crop improvement using the CRISPR-Cas9 system. Front. Plant Sci. 2017, 8, 1932. [Google Scholar] [CrossRef] [Green Version]
  26. Mohanta, T.K.; Bashir, T.; Hashem, A.; Abdallah, E.F.; Bae, H. Genome editing tools in plants. Genes 2017, 8, 399. [Google Scholar] [CrossRef] [Green Version]
  27. Zhang, K.; Raboanatahiry, N.; Zhu, B.; Li, M. Progress in genome editing technology and its application in plants. Front. Plant Sci. 2017, 8, 177. [Google Scholar] [CrossRef]
  28. Jaganathan, D.; Karthikeyan, R.; Gothandapani, S.; Shilpha, J.; Gayatri, V. CRISPR for crop improvement: An update review. Front. Plant Sci. 2018, 9, 985. [Google Scholar] [CrossRef]
  29. Borrelli, V.M.G.; Brambilla, V.; Rogowsky, P.; Marocco, A.; Lanubile, A. The enhancement of plant disease resistance using CRISPR/Cas9 technology. Front. Plant Sci. 2018, 9, 1245. [Google Scholar] [CrossRef]
  30. Mishra, R.; Zhao, K. Genome editing technologies and their applications in crop improvement. Plant Biotechnol. Rep. 2018, 12, 57–68. [Google Scholar] [CrossRef]
  31. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  32. Jinek, M.; Jiang, F.; Taylor, D.W.; Sternberg, S.H.; 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, 1247997. [Google Scholar] [CrossRef] [Green Version]
  33. Makarova, K.S.; Wolf, Y.; Alkhnbashi, O.S.; Costa, F.; Shah, S.; Saunders, S.; Barrangou, R.; Brouns, S.; Charpentier, E.; Haft, D.H.; et al. An updated evolutionary classification of CRISPR-Cas systems. Nat. Rev. Microbiol. 2015, 13, 722–736. [Google Scholar] [CrossRef] [Green Version]
  34. Jiang, W.Z.; Zhou, H.B.; Bi, H.H.; Fromm, M.; Yang, B.; Weeks, D.P. Demonstration of CRISPR/Cas9/sgRNA-mediated targeted gene modification in Arabidopsis, Tobacco, Sorghum, and rice. Nucleic Acids Res. 2013, 41, e188. [Google Scholar] [CrossRef]
  35. Miao, J.; Guo, D.; Zhang, J.; Huang, Q.; Qin, G.; Zhang, X.; Wan, J.; Gu, H.; Qu, L.J. Targeted mutagenesis in rice using CRISPR-Cas system. Cell Res. 2013, 23, 1233–1236. [Google Scholar] [CrossRef] [Green Version]
  36. Xie, K.; Yang, Y. RNA-guided genome editing in plants using a CRISPR–Cas system. Mol. Plant 2013, 6, 1975–1983. [Google Scholar] [CrossRef] [Green Version]
  37. Belhaj, K.; Chaparro Garcia, A.; Kamoun, S.; Patron, N.J.; Nekrasov, V. Editing plant genomes with CRISPR/Cas9. Curr. Opin. Biotechnol. 2015, 32, 76–84. [Google Scholar] [CrossRef]
  38. Weeks, D.P.; Spalding, M.H.; Yang, B. Use of designer nucleases for targeted gene and genome editing in plants. Plant Biotechnol. J. 2016, 14, 483–495. [Google Scholar] [CrossRef]
  39. Ricroch, A.; Clairand, P.; Harwood, W. Use of CRISPR systems in plant genome editing: Toward new opportunities in agriculture. Emerg. Top. Life Sci. 2017, 1, 169–182. [Google Scholar]
  40. Garneau, J.; Dupuis, M.; Villion, M.; Romero, D.; Barrangou, R.; Boyaval, P.; Fremaux, C.; Horvath, P.; Magadán, A.H.; Moineau, S. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 2010, 468, 67–71. [Google Scholar] [CrossRef]
  41. Marraffini, L.A.; Sontheimer, E.J. CRISPR interference: RNA-directed adaptive immunity in bacteria and archaea. Nat. Rev. Genet. 2010, 11, 181–190. [Google Scholar] [CrossRef]
  42. Horvath, P.; Barrangou, R. CRISPR/Cas, the immune system of bacteria and archaea. Science 2010, 327, 167–170. [Google Scholar] [CrossRef] [Green Version]
  43. Van der Oost, J.; Jore, M.M.; Westra, E.R.; Lundgren, M.; Brouns, S.J. CRISPR-based adaptive and heritable immunity in prokaryotes. Trends Biochem. Sci. 2009, 34, 401–407. [Google Scholar] [CrossRef] [PubMed]
  44. Barrangou, R.; Fremaux, C.; Deveau, H.; Richards, M.; Boyaval, P.; Moineau, S.; Romero, D.A.; Horvath, P. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315, 1709–1712. [Google Scholar] [CrossRef] [PubMed]
  45. Yosef, I.; Goren, M.G.; Qimron, U. Proteins and DNA elements essential for the CRISPR adaptation process in Escherichia coli. Nucleic Acids Res. 2012, 40, 5569–5576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Karvelis, T.; Gasiunas, G.; Miksys, A.; Barrangou, R.; Horvath, P.; Siksnys, V. crRNA and tracrRNA guide Cas9-mediated DNA interference in streptococcus thermophilus. RNA Biol. 2013, 10, 841–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Deltcheva, E.; Chylinski, K.; Sharma, C.M.; Gonzales, K.; Chao, Y.; Pirzada, Z.A.; Eckert, M.R.; Vogel, I.J.; Charpentier, E. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 2011, 471, 602–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Hale, C.R.; Zhao, P.; Olson, S.; Duff, M.O.; Graveley, B.R.; Wells, L. RNA-guided RNA cleavage by a CRISPR RNA-Cas protein complex. Cell 2009, 139, 945–956. [Google Scholar] [CrossRef] [Green Version]
  49. Zetsche, B.; Gootenberg, J.; Abudayyeh, O.; Slaymaker, J.; Makarova, K.; Essletzbichler, P.; Volz, S.E.; Joung, J.; van der Oost, J.; Regev, A.; et al. Cpf1 is a single RNA-guided endonuclease of Class 2 CRISPR-Cas system. Cell 2015, 163, 759–771. [Google Scholar] [CrossRef] [Green Version]
  50. Zhou, H.; Liu, B.; Weeks, D.P.; Spalding, M.H.; Yang, B. Large chromosomal deletions and heritable small genetic changes induced by CRISPR/Cas9 in rice. Nucleic Acids Res. 2014, 42, 10903–10914. [Google Scholar] [CrossRef]
  51. Zhang, F.; Maeder, M.L.; Unger-Wallace, E.; Hoshaw, J.P.; Reyon, D.; Christian, M.; Li, X.; Pierick, C.J.; Dobbs, D.; Peterson, T.; et al. High frequency targeted mutagenesis in arabidopsis thaliana Using zinc finger nucleases. Proc. Natl. Acad. Sci. USA 2010, 107, 12028–12033. [Google Scholar] [CrossRef] [Green Version]
  52. Gaj, T.; Gersbach, C.A.; Barbas, C.F. ZFN, TALEN, and CRISPR/Cas based methods for genome engineering. Trends Biotechnol. 2013, 31, 397–405. [Google Scholar] [CrossRef] [Green Version]
  53. Johnson, R.A.; Gurevich, V.; Filler, S.; Samach, A.; Levy, A.A. Comparative assessments of CRISPR-Cas nucleases cleavage efficiency in planta. Plant Mol Biol. 2015, 87, 143–156. [Google Scholar] [CrossRef]
  54. Wu, X.; Scott, D.A.; Kriz, A.J.; Chiu, A.C.; Hsu, P.D.; Dadon, D.B. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 2014, 32, 670–676. [Google Scholar] [CrossRef] [Green Version]
  55. Chen, K.; Gao, C. Targeted genome modification technologies and their applications in crop improvements. Plant Cell Rep. 2014, 33, 575–583. [Google Scholar] [CrossRef]
  56. Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339, 819–823. [Google Scholar] [CrossRef] [Green Version]
  57. Podevin, N.; Davies, H.V.; Hartung, F.; Nogue, F.; Casacuberta, J.M. Site-directed nucleases: A paradigm shift in predictable, knowledge-based plant breeding. Trends Biotechnol. 2013, 31, 375–383. [Google Scholar] [CrossRef]
  58. Gao, L.; Cox, D.; Yan, W.; Manteiga, J.; Schneider, M.W.; Yamano, T.; Nishimasu, H.; Nureki, O.; Crosetto, N.; Zhang, F. Engineered Cpf1 variants with altered PAM specificities. Nat. Biotechnol. 2017, 35, 789–792. [Google Scholar] [CrossRef] [Green Version]
  59. Hua, K.; Tao, X.; Han, P.; Wang, R.; Zhu, J.K. Genome engineering in rice using Cas9 variants that recognize NG PAM sequences. Mol. Plant 2019, 12, 1003–1014. [Google Scholar] [CrossRef]
  60. Hu, J.H.; Miller, S.M.; Geurts, M.H.; Tang, W.; Chen, L.; Sun, N.; Zeina, C.M.; Gao, X.; Rees, H.A.; Lin, Z. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 2018, 556, 57–63. [Google Scholar] [CrossRef]
  61. Nishimasu, H.; Shi, X.; Ishiguro, S.; Gao, L.; Hirano, S.; Okazaki, S.; Noda, T.; Abudayyeh, O.O.; Nishimasu, H.; Shi, X.; et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 2018, 361, 1259–1262. [Google Scholar] [CrossRef] [PubMed]
  62. Gehrke, J.M.; Cervantes, O.; Clement, M.K.; Wu, Y.; Zeng, J.; Bauer, D.E.; Pinello, L.; Joung, J.K. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat. Biotechnol. 2018, 36, 977–982. [Google Scholar] [CrossRef] [PubMed]
  63. Gehrke, F.; Puchta, H. The CRISPR/Cas revolution reaches the RNA world: Cas13, a new Swiss Army knife for plant biologists. Plant J. Cell Mol. Biol. 2018, 94, 767–775. [Google Scholar]
  64. Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.; Kellner, M.J.; Regev, A.; et al. RNA targeting with CRISPR-Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef] [Green Version]
  65. Gootenberg, J.S.; Abudayyeh, O.O.; Lee, J.W.; Essletzbichler, P.; Dy, A.J.; Joung, J.; Verdine, V.; Donghia, N.; Daringer, N.M.; Freije, C.A.; et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science 2017, 356, 438–442. [Google Scholar] [CrossRef] [Green Version]
  66. Teng, F.; Cui, T.; Feng, G.; Guo, L.; Xu, K.; Gao, Q.; Li, T.; Li, J.; Zhou, Q.; Li, W. Repurposing CRISPR-Cas12b for mammalian genome engineering. Cell Discov. 2018, 4, 63. [Google Scholar] [CrossRef]
  67. Moon, S.B.; Lee, J.M.; Kang, J.G.; Lee, N.E.; Ha, D.I.; Kim, D.Y.; Kim, S.H.; Yoo, K.; Kim, D.; Ko, J.H.; et al. Highly efficient genome editing by CRISPR-Cpf1 using CRISPR RNA with a uridinylate-rich 3′-overhang. Nat. Commun. 2018, 9, 3651. [Google Scholar] [CrossRef]
  68. Zaidi, S.S.; Mahfouz, M.M.; Mansoor, S. CRISPR-Cpf1: A New Tool for Plant Genome Editing. Trends Plant Sci. 2017, 22, 550–553. [Google Scholar] [CrossRef] [Green Version]
  69. Jeon, Y.; Choi, Y.H.; Jang, Y.; Yu, J.; Goo, J.; Lee, G.; Jeong, Y.K.; Lee, S.H.; Kim, I.S.; Kim, J.S.; et al. Direct observation of DNA target searching and cleavage by CRISPR-Cas12a. Nat. Commun. 2018, 9, 2777. [Google Scholar] [CrossRef] [Green Version]
  70. Safari, F.; Zare, K.; Negahdaripour, M.; Barekati-Mowahed, M.; Ghasemi, Y. CRISPR Cpf1 proteins: Structure, function and implications for genome editing. Cell Biosci. 2019, 9, 36. [Google Scholar] [CrossRef] [Green Version]
  71. Harrington, L.B.; Burstein, D.; Chen, J.S.; Paez-Espino, D.; Ma, E.; Witte, I.P.; Cofsky, J.C.; Kyrpides, N.C.; Banfield, J.F.; Doudna, J.A. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science 2018, 362, 839–842. [Google Scholar] [CrossRef] [Green Version]
  72. Khan, M.Z.; Haider, S.; Mansoor, S.; Amin, I. Targeting Plant ssDNA Viruses with Engineered Miniature CRISPR-Cas14a. Trends Biotechnol. 2019, 37, 800–804. [Google Scholar] [CrossRef]
  73. Hess, G.T.; Tycko, J.; Yao, D.; Bassik, M.C. Methods and Applications of CRISPR-Mediated Base Editing in Eukaryotic Genomes. Mol. Cell 2017, 68, 26–43. [Google Scholar] [CrossRef]
  74. Yang, B.; Li, X.; Lei, L.; Chen, J. APOBEC: From mutator to editor. J. Genet. Genom. 2017, 44, 423–437. [Google Scholar] [CrossRef]
  75. Doudna, J.A.; Charpentier, E. The new frontier of genome engineering with CRISPR-Cas9. Science 2014, 346, 1258096. [Google Scholar] [CrossRef]
  76. Komor, A.C.; Zhao, T.K.; Packer, S.M.; Gaudelli, M.N.; Waterbury, L.W.; Luke, W.; Koblan, W.K.Y.; Kim, B.Y.; Badran, H.A.; Liu, D.R. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci. Adv. 2017, 3, eaao4774. [Google Scholar] [CrossRef] [Green Version]
  77. Li, X.; Wang, Y.; Liu, Y.; Yang, B.; Wang, X.; Wei, J.; Lu, Z.; Zhang, Y.; Wu, J.; Huang, X.; et al. Base editing with a Cpf1-cytidine deaminase fusion. Nat. Biotechnol. 2018, 36, 324–327. [Google Scholar] [CrossRef]
  78. Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2017, 551, 464–471. [Google Scholar] [CrossRef]
  79. Liu, L.; Chen, P.; Wang, M.; Li, X.; Wang, J.; Yin, M.; Wang, Y. C2c1-sgRNA Complex Structure Reveals RNA-Guided DNA Cleavage Mechanism. Mol. Cell 2017, 65, 310–322. [Google Scholar] [CrossRef] [Green Version]
  80. Kim, E.; Koo, T.; Park, W.S.; Kim, D.; Kim, K.; Cho, Y.H.; Song, W.D.; Lee, L.K.; Jung, H.M.; Kim, S.; et al. In vivo genome editing with a small Cas9 orthologue derived from. Campylobacter jejuni. Nat. Commun. 2017, 8, 14500. [Google Scholar] [CrossRef] [Green Version]
  81. Yamano, T.; Nishimasu, H.; Zetsche, B.; Hirano, H.; Slaymaker, I.M.; Li, Y.; Fedorova, I.; Nakane, T.; Makarova, K.S.; Koonin, E.V.; et al. Crystal Structure of Cpf1 in Complex with Guide RNA and Target DNA. Cell 2016, 165, 949–962. [Google Scholar] [CrossRef] [PubMed]
  82. Garcia-Doval, C.; Jinek, M. Molecular architectures and mechanisms of Class 2 CRISPRassociated nucleases. Curr. Opin. Struct. Biol. 2017, 47, 157–166. [Google Scholar] [CrossRef] [PubMed]
  83. Hirano, H.; Gootenberg, J.S.; Horii, T.; Abudayyeh, O.O.; Kimura, M.; Hsu, P.D.; Nakane, T.; Ishitani, R.; Hatada, I.; Zhang, F.; et al. Structure and Engineering of Francisella novicida Cas9. Cell 2016, 164, 950–961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lee, C.M.; Cradick, T.J.; Bao, G. The Neisseria meningitidis CRISPR-Cas9 System Enables Specific Genome Editing in Mammalian Cells. Mol. Ther. J. Am. Soc. Gene Ther. 2016, 24, 645–654. [Google Scholar] [CrossRef] [Green Version]
  85. Nishimasu, H.; Cong, L.; Yan, W.X.; Ran, F.A.; Zetsche, B.; Li, Y.; Kurabayashi, A.; Ishitani, R.; Zhang, F.; Nureki, O. Crystal Structure of Staphylococcus aureus Cas9. Cell 2015, 162, 1113–1126. [Google Scholar] [CrossRef] [Green Version]
  86. Ren, B.; Liu, L.; Li, S.; Kuang, Y.; Wang, J.; Zhang, D.; Zhou, X.; Lin, H.; Zhou, H. Cas9-NG Greatly Expands the Targeting Scope of the Genome-Editing Toolkit by Recognizing NG and Other Atypical PAMs in Rice. Mol. Plant 2019, 12, 1015–1026. [Google Scholar] [CrossRef]
  87. Kleinstiver, B.P.; Prew, M.S.; Tsai, S.Q.; Topkar, V.V.; Nguyen, N.T.; Zheng, Z.; Gonzales, A.P.; Li, Z.; Peterson, R.T.; Yeh, J.R.; et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 2015, 523, 481–485. [Google Scholar] [CrossRef] [Green Version]
  88. Wang, J.; Meng, X.; Hu, X.; Sun, T.; Li, J.; Wang, K.; Yu, H. xCas9 expands the scope of genome editing with reduced efficiency in rice. Plant Biotechnol. J. 2019, 17, 709–711. [Google Scholar] [CrossRef] [Green Version]
  89. Gelvin, S.B. Agrobacterium-Mediated Plant Transformation: The Biology behind the “Gene-Jockeying” Tool. Microbiol. Mol. Biol. Rev. 2003, 67, 16–37. [Google Scholar] [CrossRef] [Green Version]
  90. Li, J.F.; Norville, J.E.; Aach, J.; McCormack, M.; Zhang, D.; Bush, J.; Church, G.M.; Sheen, J. Multiplex and homologous recombination-mediated genome editing in Arabidopsis and Nicotianabenthamiana using guide RNA and Cas9. Nat. Biotechnol. 2013, 31, 688–691. [Google Scholar] [CrossRef]
  91. Yang, H.; Wang, H.; Shivalila, C.S.; Cheng, A.W.; Shi, L.; Jaenisch, R. One-step generation of mice-carrying reported and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell 2013, 154, 1370–1379. [Google Scholar] [CrossRef] [Green Version]
  92. Wei, Y.; Terns, R.M.; Terns, M.P. Cas9 function, and host genome sampling in Type II-A CRISPR-Cas adaptation. Genes Dev. 2015, 29, 356–361. [Google Scholar] [CrossRef] [Green Version]
  93. Xiao, A.; Cheng, Z.; Kong, L.; Zhu, Z.; Lin, S.; Gao, G.; Zhang, B. CasOT: A Genome-wide Cas9/grna off-target searching Tool. Bioinformatics 2014, 30, 1180–1182. [Google Scholar] [CrossRef] [Green Version]
  94. Xie, X.; Ma, X.; Zhu, Q.; Zeng, D.; Li, G.; Liu, Y.G. CRISPR-GE: A convenient software toolkit for CRISPR-based genome editing. Mol. Plant 2017, 10, 1246–1249. [Google Scholar] [CrossRef] [Green Version]
  95. Ma, M.; Ye, A.Y.; Zheng, W.; Kong, L. A guide RNA sequence design platform for the CRISPR/Cas9 system for model organism genomes. Bio. Med. Res. Int. 2013, 2013, 270805. [Google Scholar] [CrossRef]
  96. Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; Dicarlo, J.E.; Norville, J.E.; Church, G.M. RNA-guided human genome engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef] [Green Version]
  97. Gilbert, L.; Horlbeck, M.A.; Adamson, B.; Villalta, J.E.; Chen, Y.; Whitehead, E.H.; Guimaraes, C.; Panning, B.; Ploegh, H.L.; Bassik, M.C.; et al. Genome-scale CRISPR-Mediated control of gene repression and activation. Cell 2014, 159, 647–661. [Google Scholar] [CrossRef] [Green Version]
  98. Naito, Y.; Hino, K.; Bono, H.; UiTei, K. CRISPRdirect: Software for designing CRISPR/Cas guide RNA with reduced off-target sites. Bioinformatics 2014, 31, 1120–1123. [Google Scholar] [CrossRef]
  99. Lei, Y.; Lu, L.; Liu, H.Y.; Li, S.; Xing, F.; Chen, L.L. CRISPR–P: A web tool for synthetic single-guide RNA design of CRISPR–system in plants. Mol. Plant 2014, 7, 1494–1496. [Google Scholar] [CrossRef] [Green Version]
  100. Pinello, L.; Canver, M.C.; Hoban, M.D.; Orkin, S.H.; Kohn, D.B.; Bauer, D.E.; Yuan, G.-C. Analyzing CRISPR Genome-editing experiments with CRISPResso. Nat. Biotechnol. 2016, 34, 695–697. [Google Scholar] [CrossRef] [Green Version]
  101. Doench, J.G.; Fusi, N.; Sullender, M.; Hegde, M.; Vaimberg, E.W.; Donovan, K.F.; Smith, I.; Tothova, Z.; Wilen, C.; Orchard, R.; et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 2016, 34, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Oliveros, J.C.; Franch, M.; Tabas–Madrid, D.; León San, D.; Montoliu, L.; Cubas, P.; Pazos, F. Breaking Cas-interactive design of guide RNAs for CRISPR–Cas experiments for Ensembl genomes. Nucleic Acids Res. 2016, 44, W267–W271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ma, J.; Köster, J.; Qin, Q.; Hu, S.; Li, W.; Chen, C.; Cao, Q.; Wang, J.; Mei, S.; Liu, Q.; et al. CRISPR-DO for genome-wide CRISPR design and 287 optimization. Bioinformatics 2016, 32, 3336–3338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Haeussler, M.; Schönig, K.; Eckert, H.; Eschstruth, A.; Mianné, J.; Renaud, J.B.; Schneider-Maunoury, S.; Shkumatava, A.; Teboul, L.; Kent, J.; et al. Evaluation of off-target and on-target scoring algorithms and integration into the guideRNA selection tool CRISPR. Genome Biol. 2016, 17, 148. [Google Scholar] [CrossRef]
  105. Zhu, H.; Misel, L.; Graham, M.; Robinson, M.L.; Liang, C. CT-Finder: A web service for CRISPR Optimal target prediction and visualization. Sci. Rep. 2016, 6, 25516. [Google Scholar] [CrossRef] [Green Version]
  106. Xie, S.; Shen, B.; Zhang, C.; Huang, X.; Zhang, Y. SgRNACas9: A software package for designing CRISPR SgRNA and evaluating potential off-target cleavage sites. PLoS ONE 2014, 9, e100448. [Google Scholar] [CrossRef]
  107. Hsu, P.D.; Scott, D.A.; Weinstein, J.A.; Ran, F.A.; Konermann, S.; Agarwala, V.; Li, Y.; Fine, E.J.; Wu, X.; Shalem, O.; et al. DNA targeting specificity of RNA–guided Cas9 nucleases. Nat. Biotechnol. 2013, 31, 827–832. [Google Scholar] [CrossRef]
  108. Brazelton, V.A.; Zarecor, S.; Wright, D.A.; Wang, Y.; Liu, J.; Chen, K.; Yang, B.; Lawrence-Dill, C.J. A quick guide to CRISPR sgRNA design tools. GM Crop. Food 2015, 6, 266–276. [Google Scholar] [CrossRef] [Green Version]
  109. Park, J.; Bae, S.; Kim, J.S. Cas–Designer: A web-based tool for the choice of CRISPR–Cas9 target sites. Bioinformatics 2015, 31, 4014–4016. [Google Scholar] [CrossRef] [Green Version]
  110. Bae, S.; Park, J.; Kim, J.S. Cas-OFFinder: A fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 2014, 30, 1473–1475. [Google Scholar] [CrossRef] [Green Version]
  111. Stemmer, M.; Thumberger, T.; del Sol Keyer, M.; Wittbrodt, J.; Mateo, J. CCTop: An intuitive, flexible, and reliable CRISPR/Cas9 target prediction tool. PLoS ONE 2015, 10, e0124633. [Google Scholar] [CrossRef] [Green Version]
  112. MacPherson, C.R.; Scherf, A. Flexible guide–RNA design for CRIS-PR applications using Protospacer Workbench. Nat. Biotechnol. 2015, 33, 805–806. [Google Scholar] [CrossRef] [Green Version]
  113. Xu, H.; Xiao, T.; Chen, C.H.; Li, W.; Meyer, C.A.; Wu, Q.; Wu, D.; Cong, L.; Zhang, F.; Liu, J.S.; et al. Sequence determinants of improved CRISPR sgRNA design. Genome Res. 2015, 25, 1147–1157. [Google Scholar] [CrossRef] [Green Version]
  114. Prykhozhij, S.V.; Rajan, V.; Gaston, D.; Berman, J.N. CRISPR multitargeter: A web tool to find common and unique CRISPR single guide RNA targets in a set of similar sequences. PLoS ONE 2015, 10, e0119372. [Google Scholar]
  115. Li, W.; Xu, H.; Xiao, T.; Cong, L.; Love, M.I.; Zhang, F.; Irizarry, R.A.; Liu, J.S.; Brown, M.; Liu, X.S. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 2014, 15, 554. [Google Scholar] [CrossRef]
  116. O’Brien, A.; Bailey, T.L. GT–Scan: Identifying unique genomic targets. Bioinformatics 2014, 30, 2673–2675. [Google Scholar] [CrossRef]
  117. Kaur, K.; Tandon, H.; Gupta, A.K.; Kumar, M. CrisprGE: Central hub of CRISPR/Cas-based genome editing. Database J. Biol. Databases Curation 2015, 2015, bav055. [Google Scholar] [CrossRef] [Green Version]
  118. Li, X.; Zhou, W.; Ren, Y.; Tian, X.; Lv, T.; Wang, Z.; Fang, J.; Chu, C.; Yang, J.; Bu, Q. High-efficiency breeding of early-maturing rice cultivars via CRISPR/Cas9-mediated genome editing. J. Genet. Genom. 2017, 44, 175–178. [Google Scholar] [CrossRef]
  119. Zhao, D.S.; Li, Q.F.; Zhang, C.Q.; Zhang, C.; Yang, Q.Q.; Pan, L.X.; Ren, X.Y.; Lu, J.; Gu, M.H.; Liu, Q.Q. GS9 acts as a transcriptional activator to regulate rice grain shape and appearance quality. Nat. Commun. 2018, 9, 1240. [Google Scholar] [CrossRef]
  120. Li, M.; Li, X.; Zhou, Z.; Wu, P.; Fang, M.; Pan, X.; Lin, Q.; Luo, W.; Wu, G.; Li, H. Reassessment of the four yield-related genes Gn1a, DEP1, GS3, and IPA1 in Rice Using a CRISPR/Cas9 System. Front. Plant Sci. 2016, 7, 377. [Google Scholar] [CrossRef] [Green Version]
  121. 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. Sci. Rep. 2016, 6, 37395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Yu, J.; Jiaojiao, H.; Yu, J.K.; Ming, S.; Zhen, Y.; Yi, H.; Fu, R.; Luo, Z.; Hu, J.; Liang, W.; et al. Two rice receptor-like kinases maintain male fertility under changing temperatures. Proc. Natl. Acad. Sci. USA 2017, 114, 12327–12332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Li, S.; Gao, F.; Xie, K.; Zeng, X.; Cao, Y.; Zeng, J.; He, Z.; Ren, Y.; Li, W.; Deng, Q.; et al. The OsmiR396c-OsGRF4-OsGIF1 regulatory module determines grain size and yield in rice. Plant Biotechnol. J. 2016, 14, 2134–2146. [Google Scholar] [CrossRef] [PubMed]
  124. Miao, C.; Xiao, L.; Hua, K.; Zou, C.; Zhao, Y.; Bressan, R.A.; Zhu, J.K. Mutations in a subfamily of abscisic acid receptor genes promote rice growth and productivity. Proc. Natl. Acad. Sci. USA 2018, 115, 6058–6063. [Google Scholar] [CrossRef] [Green Version]
  125. Chao, S.; Cai, Y.; Feng, B.; Jiao, G.; Sheng, Z.; Luo, J.; Tang, S.; Wang, J.; Wei, X.; Hu, P. Editing of the rice isoamylase gene ISA1 provides insights into its function in starch formation. Rice Sci. 2019, 26, 77–87. [Google Scholar]
  126. Mikami, M.; Toki, S.; Endo, M. Comparison of CRISPR/Cas9 expression constructs for efficient, targeted mutagenesis in rice. Plant Mol. Biol. 2015, 88, 561–572. [Google Scholar] [CrossRef] [Green Version]
  127. Shao, G.; Xie, L.; Jiao, G.; Wei, X.; Sheng, Z.; Tang, S.; Hu, P. CRISPR/CAS9-mediated editing of the fragrant gene Badh2 in rice. Chin. J. Rice Sci. 2017, 31, 216–222. [Google Scholar]
  128. Zhou, X.; Liao, H.; Chern, M.; Yin, J.; Chen, Y.; Wang, J.; Zhu, X.; Chen, Z.; Yuan, C.; Zhao, W.; et al. Loss of function of a rice TPR-domain RNA-binding protein confers broad-spectrum disease resistance. Proc. Natl. Acad. Sci. USA 2018, 115, 3174–3179. [Google Scholar] [CrossRef] [Green Version]
  129. Zhou, J.; Peng, Z.; Long, J.; Sosso, D.; Liu, B.; Eom, J.S.; Huang, S.; Liu, S.; Vera Cruz, C.; Frommer, W.B.; et al. Gene targeting by the TAL effector PthXo2 reveals cryptic resistance gene for bacterial blight of rice. Plant J. 2015, 82, 632–643. [Google Scholar] [CrossRef]
  130. Li, C.; Li, W.; Zhou, Z.; Chen, H.; Xie, C.; Lin, Y. A new rice breeding method: CRISPR/Cas9system editing of the Xa13promoter to cultivate transgene-free bacterial blight-resistant rice. Plant Biotechnol. J. 2019, 18, 313–315. [Google Scholar] [CrossRef] [Green Version]
  131. Macovei, A.; Sevilla, N.R.; Cantos, C.; Jonson, G.B.; Slamet-Loedin, I.; Cermak, T.; Voytas, D.F.; Choi, I.R.; Chadha-Mohanty, P. Novel alleles of rice eIF4G generated by CRISPR/Cas9-targeted mutagenesis confer resistance to Rice tungro spherical virus. Plant Biotechnol. J. 2018, 16, 1918–1927. [Google Scholar] [CrossRef] [Green Version]
  132. Li, J.; Meng, X.; Zong, Y.; Chen, K.; Zhang, H.; Liu, J.; Li, J.; Gao, C. Gene replacements and insertions in rice by intron targeting using CRISPR-Cas9. Nat. Plants 2016, 12, 16139. [Google Scholar] [CrossRef]
  133. Sun, Y.; Zhang, X.; Wu, C.; He, Y.; Ma, Y.; Hou, H.; Guo, X.; Du, W.; Zhao, Y.; Xia, L. Engineering herbicide-resistant rice plants through CRISPR/Cas9-mediated homologous recombination of acetolactate synthase. Mol. Plant 2016, 9, 628–631. [Google Scholar] [CrossRef] [Green Version]
  134. Zhang, H.; Zhang, J.; Wei, P.; Zhang, B.; Gou, F.; Feng, Z.; Mao, Y.; Yang, L.; Zhang, H.; Xu, N.; et al. The CRISPR/Cas9 system produces specific and homozygous targeted gene editing in rice in one generation. Plant Biotechnol. J. 2014, 12, 797–807. [Google Scholar] [CrossRef]
  135. Xu, R.F.; Li, H.; Qin, R.Y.; Li, J.; Qiu, C.H.; Yang, Y.C.; Ma, H.; Li, L.; Wei, P.C.; Yang, J.B. Generation of inheritable and “transgene clean” targeted genome-modified rice in later generations using the CRISPR/Cas9 system. Sci. Rep. 2015, 5, 11491. [Google Scholar] [CrossRef] [Green Version]
  136. Nieves-Cordones, M.; Mohamed, S.; Tanoi, K.; Kobayashi, N.I.; Takagi, K.; Vernet, A.; Guiderdoni, E.; Périn, C.; Sentenac, H.; Véry, A.A. Production of low- Cs+ rice plants by inactivation of the K+ transporter OsHAK1 with the CRISPR-Cas system. Plant J. 2017, 92, 43–56. [Google Scholar] [CrossRef] [Green Version]
  137. Mao, X.; Zheng, Y.; Xiao, K.; Wei, Y. Biochemical and biophysical research communications OsPRX2 contributes to stomatal closure and improves potassium de Fi Ciency tolerance in rice. Biochem. Biophys. Res. Commun. 2018, 495, 461–467. [Google Scholar] [CrossRef]
  138. Zhang, J.; Zhang, H.; Botella, J.R.; Zhu, J.K. Generation of new glutinous rice by CRISPR/Cas9-targeted mutagenesis of the Waxy gene in elite rice varieties. J. Integr. Plant Biol. 2018, 60, 369–375. [Google Scholar] [CrossRef]
  139. Sun, Y.; Jiao, G.; Liu, Z.; Zhang, X.; Li, J.; Guo, X.; Du, W.; Du, J.; Francis, F.; Zhao, Y.; et al. Generation of high-amylose rice through CRISPR/Cas9-mediated targeted mutagenesis of starch branching enzymes. Front Plant Sci. 2017, 8, 298. [Google Scholar] [CrossRef]
  140. Yang, Q.Q.; Zhang, C.Q.; Chan, M.L.; Zhao, D.S.; Chen, J.Z.; Wang, Q.; Li, Q.F.; Yu, H.X.; Gu, M.H.; Sun, S.S.; et al. Biofortification of rice with the essential amino acid lysine: Molecular characterization, nutritional evaluation, and field performance. J. Exp. Bot. 2016, 67, 4285–4296. [Google Scholar] [CrossRef] [Green Version]
  141. Yin, X.; Biswal, A.K.; Dionora, J.; Perdigon, K.M.; Balahadia, C.P.; Mazumdar, S.; Chater, C.; Lin, H.C.; Coe, R.A.; Kretzschmar, T.; et al. CRISPR-Cas9 and CRISPR-Cpf1 mediated targeting of a stomatal developmental gene EPFL9 in rice. Plant Cell Rep. 2017, 36, 745–757. [Google Scholar] [CrossRef] [PubMed]
  142. Masaki, E.; MasafumiMikami, S.T. Multigene knockout utilizing off-target mutations of the CRISPR/Cas9system in Rice. Plant Cell Physiol. 2015, 56, 41–47. [Google Scholar]
  143. Mikami, M.; Toki, S.; Endo, M. Parameters affecting the frequency of CRISPR/Cas9 mediated targeted mutagenesis in rice. Plant Cell Rep. 2015, 34, 1807–1815. [Google Scholar] [CrossRef] [PubMed]
  144. Mikami, M.; Toki, S.; Endo, M. Precision targeted mutagenesis via Cas9 paired nickases in rice. Plant Cell Physiol. 2016, 57, 1058–1068. [Google Scholar] [CrossRef] [Green Version]
  145. Liu, X.; Zhang, C.; Wang, X.; Liu, Q.; Yuan, D.; Pan, G.; Sun, S.S.; Tu, J. Development of high-lysine rice via endosperm-specific expression of a foreign LYSINE RICH PROTEIN gene. BMC Plant Biol. 2016, 16, 147. [Google Scholar] [CrossRef] [Green Version]
  146. Lu, K.; Wu, B.; Wang, J.; Zhu, W.; Nie, H.; Qian, J.; Huang, W.; Fang, Z. Blocking amino acid transporter OsAAP3 improves grain yield by promoting outgrowth buds and increasing tiller number in rice. Plant Biotechnol. J. 2018, 16, 1710–1722. [Google Scholar] [CrossRef] [Green Version]
  147. Usman, B.; Nawaz, G.; Zhao, N.; Liao, S.; Qin, B.; Liu, F.; Liu, Y.; Li, R. Programmed editing of rice (Oryza sativa L.) osspl16 gene using crispr/cas9 improves grain yield by modulating the expression of pyruvate enzymes and cell cycle proteins. Int. J. Mol. Sci. 2021, 22, 249. [Google Scholar] [CrossRef]
  148. Fiaz, S.; Ahmad, S.; Noor, M.A.; Wang, X.; Younas, A.; Riaz, A.; Riaz, A.; Ali, F.; Fiaz, S.; Ahmad, S.; et al. Applications of the CRISPR/Cas9system for rice grain quality improvement: Perspectives and opportunities. Int. J. Mol. Sci. 2019, 20, 888. [Google Scholar] [CrossRef] [Green Version]
  149. Bao, J. Genes and QTLs for rice grain quality improvement. In Rice-Germplasm, Genetics, and Improvement; InTech: London, UK, 2014. [Google Scholar]
  150. Li, Y.; Fan, C.; Xing, Y.; Yun, P.; Luo, L.; Yan, B.; Peng, B.; Xie, W.; Wang, G.; Li, X.; et al. Chalk5 encodes a vacuolar H+-translocating pyrophosphatase influencing grain chalkiness in rice. Nat. Genet. 2014, 46, 398. [Google Scholar] [CrossRef]
  151. Wang, Z.Y.; Zheng, F.Q.; Shen, G.Z.; Gao, J.P.; Snustad, D.P.; Li, M.G.; Zhang, J.L.; Hong, M.M. The amylose content in rice endosperm is related to the post-transcriptional regulation of the waxy gene. Plant J. 1995, 7, 613–622. [Google Scholar] [CrossRef]
  152. Ma, X.; Zhang, Q.; Zhu, Q.; Liu, W.; Chen, Y.; Qiu, R.; Wang, B.; Yang, Z.; Li, H.; Lin, Y.; et al. A robust CRISPR/Cas9 system for convenient, high-efficiency multiplex genome editing in monocot and dicot plants. Mol. Plant 2015, 8, 1274–1284. [Google Scholar] [CrossRef]
  153. Terada, R.; Nakajima, M.; Isshiki, M.; Okagaki, R.J.; Wessler, S.R.; Shimamoto, K. Antisense waxy genes wit highly active promoters effectively suppress waxy gene expression in transgenic rice. Plant Cell Physiol. 2000, 41, 881–888. [Google Scholar] [CrossRef] [Green Version]
  154. Yang, X.; Chen, L.; Yu, W. Knocking out of carotenoid catabolic genes in rice fails to boost carotenoid accumulation but reveals a mutation in strigolactone biosynthesis. Plant Cell Rep. 2017, 36, 1533–1545. [Google Scholar] [CrossRef]
  155. Kissoudis, C.; van de Wiel, C.; Visser, R.G.F.; van der Linden, G. Enhancing crop resilience to combined abiotic and biotic stress through the dissection of physiological and molecular crosstalk. Front. Plant Sci. 2014, 5, 207. [Google Scholar] [CrossRef] [Green Version]
  156. Jain, M. Function genomics of abiotic stress tolerance in plants: A CRISPR approach. Front. Plant Sci. 2015, 6, 375. [Google Scholar] [CrossRef] [Green Version]
  157. Lu, H.P.; Luo, T.; Fu, H.W.; Wang, L.; Tan, Y.Y.; Huang, J.Z.; Wang, Q.; Ye, G.Y.; Gatehouse, A.; Lou, Y.G.; et al. Resistance of rice to insect pests mediated by suppression of serotonin biosynthesis. Nat. Plants 2018, 4, 338–344. [Google Scholar] [CrossRef]
  158. Wang, F.Z.; Chen, M.X.; Yu, L.J.; Xie, L.J.; Yuan, L.B.; Qi, H.; Xiao, M.; Guo, W.; Chen, Z.; Yi, K.; et al. OsARM1, an R2R3 MYB transcription factor, is involved in the regulation of the response to arsenic stress in rice. Front. Plant Sci. 2017, 8, 1868. [Google Scholar] [CrossRef] [Green Version]
  159. Li, Z. Health risk characterization of maximum legal exposures for persistent organic pollutant (POP) pesticides in residential soil: An analysis. J. Environ. Manag. 2018, 205, 163–173. [Google Scholar] [CrossRef]
  160. Huang, X.Z.; Xiao, F.Z.; Li, J.R.; Zhao, D.G. Construction and analysis of tify1a and tify1b mutants in rice (Oryza sativa) based on CRISPR/Cas9 technology. J. Agric. Biotechnol. 2017, 25, 1003–1012. [Google Scholar]
  161. Wang, F.; Xu, Y.; Li, W.; Chen, Z.; Wang, J.; Fan, F.; Tao, Y.; Jiang, Y.; Zhu, Q.-H.; Yang, J. Creating a novel herbicide-tolerance OsALS allele using CRISPR/Cas9-mediated gene editing. Crop J. 2021, 9, 305–312. [Google Scholar] [CrossRef]
  162. Ahmad, S.; Sheng, Z.; Jalal, R.S.; Tabassum, J.; Ahmed, F.K.; Hu, S.; Shao, G.; Wei, X.; Abd-Elsalam, K.A.; Hu, P.; et al. CRISPR–Cas technology towards improvement of abiotic stress tolerance in plants. In CRISPR and RNAi Systems; Elsevier: Amsterdam, The Netherlands, 2021; pp. 755–772. [Google Scholar]
  163. 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, 667–697. [Google Scholar] [CrossRef] [PubMed]
  164. Barman, H.N.; Sheng, Z.; Fiaz, S.; Zhong, M.; Wu, Y.; Cai, Y.; Wang, W.; Jiao, G.; Tang, S.; Wei, X.; et al. Generation of thermo-sensitive genic sterile line by knockoutTMS5 gene based on CRISPR/Cas9system. BMC Plant Biol. 2019, 19, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Li, Q.; Zhang, D.; Chen, M.; Liang, W.; Wei, J.; Qi, Y.; Yuan, Z. Development of japonica photo-sensitive genic male sterile rice lines by editing carbon starved anther using CRISPR/Cas9. J. Genet. Genom. 2016, 43, 415–419. [Google Scholar] [CrossRef] [PubMed]
  166. Xie, Y.; Niu, B.; Long, Y.; Li, G.; Tang, J.; Zhang, Y.; Ren, D.; Liu, Y.; Chen, L. Suppression or knockout of SaF/SaM overcomes the Sa-mediated hybrid male sterility in rice. J. Integr. Plant Biol. 2017, 59, 669–679. [Google Scholar] [CrossRef] [Green Version]
  167. Shen, R.; Wang, L.; Liu, X.; Wu, J.; Jin, W.; Zhao, X.; Xie, X.; Zhu, Q.; Tang, H.; Li, Q.; et al. Genomic structural variation-mediated allelic suppression causes hybrid male sterility in rice. Nat. Commun. 2017, 8, 1310. [Google Scholar] [CrossRef] [Green Version]
  168. Yu, X.; Zhao, Z.; Zheng, X.; Zhou, J.; Kong, W.; Wang, P.; Bai, W.; Zheng, H.; Zhang, H.; Li, J.; et al. A selfish genetic element confers non-Mendelian inheritance in rice. Science 2018, 360, 1130–1132. [Google Scholar] [CrossRef] [Green Version]
  169. Khanday, I.; Skinner, D.; Yang, B.; Mercier, R.; Sundaresan, V. A male-expressed rice embryogenic trigger redirected for asexual propagation through seeds. Nature 2018, 565, 91–95. [Google Scholar] [CrossRef]
  170. 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, 283–286. [Google Scholar] [CrossRef]
  171. Jacobsen, E.; Schouten, H.J. Cisgenesis strongly improves introgression breeding and induced translocation breeding of plants. Trends Biotechnol. 2007, 25, 219–223. [Google Scholar] [CrossRef]
  172. Lowder, L.G.; Zhang, D.; Baltes, N.J.; Paul, J.W.; Tang, X.; Zheng, X. A CRISPR/Cas9 toolbox for multiplexed plant genome editing and transcriptional regulation. Plant Physiol. 2015, 169, 971–985. [Google Scholar] [CrossRef] [Green Version]
  173. Zhang, Z.; Mao, Y.; Ha, S.; Liu, W.; Botella, J.R.; Zhu, J.K. A multiplex CRISPR/Cas9 platform for fast and efficient editing of multiple genes in Arabidopsis. Plant Cell Rep. 2016, 35, 1519–1533. [Google Scholar] [CrossRef] [Green Version]
  174. Shen, L.; Hua, Y.; Fu, Y.; Li, J.; Liu, Q.; Jiao, X.; Xin, G.; Wang, J.; Wang, X.; Yan, C.; et al. Rapid generation of genetic diversity by multiplex CRISPR/Cas9 genome editing in rice. Sci. China Life Sci. 2017, 60, 506–515. [Google Scholar] [CrossRef]
  175. Manghwar, H.; Li, B.; Ding, X.; Hussain, A.; Lindsey, K.; Zhang, X.; Jin, S. CRISPR/Cas systems in genome editing: Methodologies and tools for sgrna design, off-target evaluation, and strategies to mitigate off-target effects. Adv. Sci. 2020, 7, 1902312. [Google Scholar] [CrossRef]
  176. Shen, L.; Wang, C.; Fu, Y.; Wang, J.; Liu, Q.; Zhang, X.; Yan, C.; Qian, Q.; Wang, K. QTL editing confers opposing yield performance in different rice varieties. J. Integr. Plant Biol. 2018, 60, 89–93. [Google Scholar] [CrossRef]
  177. Lu, Y.; Yifu, T.; Rundong, S.; Qi, Y.; Mugui, W.; Mei, C.; Dong, J.; Zhang, T.; Li, F.; Lei, M.; et al. Targeted, efficient sequence insertion and replacement in rice. Nat. Biotechnol. 2020, 32, 4264. [Google Scholar] [CrossRef]
  178. Ahmad, S.; Tang, L.; Shahzad, R.; Mawia, A.M.; Rao, G.S.; 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, 8307–8323. [Google Scholar] [CrossRef]
  179. Ainley, W.M.; Sastry Dent, L.; Welter, M.E.; Murray, M.G.; Zeitler, B.; Amora, R.; Corbin, D.R.; Miles, R.R.; Arnold, N.L.; Strange, T.L.; et al. Trait stacking via targeted genome editing. Plant Biotechnol. J. 2013, 11, 1126–1134. [Google Scholar] [CrossRef]
  180. Gaillochet, C.; Develtere, W.; Jacobs, T.B. CRISPR Screens in Plants: Approaches, Guidelines, and Future Prospects. Plant Cell 2021, 33, 794–813. [Google Scholar] [CrossRef]
  181. Zachary, H.L.; Nathan, T.R.; Justin, D.; Sebastian, S.; Kerry, E.S.; Daniel, R.; Joyce, V.E.; Zachary, B.L. Rapid improvement of domestication traits in an orphan crop by genome editing. Nat. Plants 2018, 4, 766–770. [Google Scholar]
  182. Arenas, S.; Cortés, A.J.; Mastretta-Yanes, A.; Jaramillo-Correa, J.P. Evaluating the accuracy of genomic prediction for the management and conservation of relictual natural tree populations. Tree Genet. Genomes 2021, 17, 12. [Google Scholar] [CrossRef]
Figure 1. Components of CRISPR/Cas9 system: (A). Genomic structures of the CRISPR/Cas system (top) and the engineered CRISPR/Cas9 system (bottom); (B). A schematic representation of the Cas9 protein structure. Domains includes REC (large recognition lobe) and RuvC (a nuclease domain), which is linked with an arginine-rich region. HNH is a second nuclease domain. PI is PAM-interacting domain; (C). The conformation of the Cas9–sgRNA complex in the process of DNA cleavage. The Cas9 endonuclease is targeted to DNA by a guide RNA which can be supplied as a two-part system consisting of crRNA and tracrRNA or as a single guide RNA, where the crRNA and tracrRNA are connected by a linker. Target recognition is facilitated by the protospacer-adjacent motif (PAM). Cleavage occurs on both strands (scissors) 3 bp upstream of the PAM.
Figure 1. Components of CRISPR/Cas9 system: (A). Genomic structures of the CRISPR/Cas system (top) and the engineered CRISPR/Cas9 system (bottom); (B). A schematic representation of the Cas9 protein structure. Domains includes REC (large recognition lobe) and RuvC (a nuclease domain), which is linked with an arginine-rich region. HNH is a second nuclease domain. PI is PAM-interacting domain; (C). The conformation of the Cas9–sgRNA complex in the process of DNA cleavage. The Cas9 endonuclease is targeted to DNA by a guide RNA which can be supplied as a two-part system consisting of crRNA and tracrRNA or as a single guide RNA, where the crRNA and tracrRNA are connected by a linker. Target recognition is facilitated by the protospacer-adjacent motif (PAM). Cleavage occurs on both strands (scissors) 3 bp upstream of the PAM.
Ijms 23 04454 g001
Figure 2. The advantages and disadvantages of the CRISPR/Cas9 system over other approaches for genome editing. (A). Conventional gene targeting. (B). ZNFs and TALENs. (C). CRISPR/SpCas9. (D). CRISPR/NmCas9. The red arrow indicates the corresponding gene editing method with its features, advantages, and disadvantages.
Figure 2. The advantages and disadvantages of the CRISPR/Cas9 system over other approaches for genome editing. (A). Conventional gene targeting. (B). ZNFs and TALENs. (C). CRISPR/SpCas9. (D). CRISPR/NmCas9. The red arrow indicates the corresponding gene editing method with its features, advantages, and disadvantages.
Ijms 23 04454 g002
Figure 3. Comparison of CRISPR/Cas9 with newly emerging CRISPR/Cas GE tools. (A) In the CRISPR/Cas9 system, Cas9 is a multicomponent protein and recognizes a G-rich PAM at the 3′ end of the target site. Both tracrRNA and crRNA are required to recruit Cas9. Then, the Cas9 creates a DSB, resulting in blunt ends. (B) In the CRISPR/Cas12a System, Cas12a is a single-component protein which recognizes T-rich PAM at the 5′ end of the target sequence; tracrRNA is not required. The DSB results in a 5′ overhang sticky ends with staggered cuts. (C) In the nuclear base-editing system, cytidine deaminase fused with dCas9 is used to target the desired site. There is no DSB, cytidine deaminase converts C directly into U, and during mismatch repair a C→ T substitution can be corrected when the modified strand is used as template. (D) Base editing in RNA. In the REPAIR system, “A-to-I” editing uses dCas13 fused to ADAR2. REPAIR uses 50- nucleotide RNA with a 50-nucleotide mRNA–gRNA duplex. “A–C” mismatch in the RNA–gRNA duplex determines the target A. RESCUE system editing “C-to-U.” The optimum results are achieved with a gRNA with a 30-nucleotide spacer. The target “C” is specified by an induced “C–C” or “C–U” mismatch in the mRNA–gRNA duplex. (E) Prime editing. (a) Nicking the desired DNA sequence at the PAM strand by the fusion protein, (b) the exposed 3′ hydroxyl group primes the reverse transcription (RT) of the RT template of the prime editing gRNA (pegRNA), (c) reverse transcription, (d) the branched intermediate form containing two flaps of DNA: a 3′ flap (containing the edited sequence), and a 5′ flap (containing the dispensable, unedited DNA sequence) followed by flap cleavage, and (e) ligation and mismatch repair; either incorporating the edited strand or removing it.
Figure 3. Comparison of CRISPR/Cas9 with newly emerging CRISPR/Cas GE tools. (A) In the CRISPR/Cas9 system, Cas9 is a multicomponent protein and recognizes a G-rich PAM at the 3′ end of the target site. Both tracrRNA and crRNA are required to recruit Cas9. Then, the Cas9 creates a DSB, resulting in blunt ends. (B) In the CRISPR/Cas12a System, Cas12a is a single-component protein which recognizes T-rich PAM at the 5′ end of the target sequence; tracrRNA is not required. The DSB results in a 5′ overhang sticky ends with staggered cuts. (C) In the nuclear base-editing system, cytidine deaminase fused with dCas9 is used to target the desired site. There is no DSB, cytidine deaminase converts C directly into U, and during mismatch repair a C→ T substitution can be corrected when the modified strand is used as template. (D) Base editing in RNA. In the REPAIR system, “A-to-I” editing uses dCas13 fused to ADAR2. REPAIR uses 50- nucleotide RNA with a 50-nucleotide mRNA–gRNA duplex. “A–C” mismatch in the RNA–gRNA duplex determines the target A. RESCUE system editing “C-to-U.” The optimum results are achieved with a gRNA with a 30-nucleotide spacer. The target “C” is specified by an induced “C–C” or “C–U” mismatch in the mRNA–gRNA duplex. (E) Prime editing. (a) Nicking the desired DNA sequence at the PAM strand by the fusion protein, (b) the exposed 3′ hydroxyl group primes the reverse transcription (RT) of the RT template of the prime editing gRNA (pegRNA), (c) reverse transcription, (d) the branched intermediate form containing two flaps of DNA: a 3′ flap (containing the edited sequence), and a 5′ flap (containing the dispensable, unedited DNA sequence) followed by flap cleavage, and (e) ligation and mismatch repair; either incorporating the edited strand or removing it.
Ijms 23 04454 g003
Figure 4. T-DNA vector with all the components necessary for Cas9-induced mutagenesis. The 20 bp protospacer sequences of each target site are subcloned or integrated between the sgRNA scaffold and the U3 promoter by ligation of primers into an AarI-digested SK-sgRNA vector. Then, this vector is again re-ligated with a pC1300-Cas9 vector by ligation of BamHI and KnpI-digested enzymes. The whole sgRNA cassette is then delivered into a pC1300-Cas9 vector (contains the Cas9 gene under the control of the 2 × 35S promoter) for plant transformation.
Figure 4. T-DNA vector with all the components necessary for Cas9-induced mutagenesis. The 20 bp protospacer sequences of each target site are subcloned or integrated between the sgRNA scaffold and the U3 promoter by ligation of primers into an AarI-digested SK-sgRNA vector. Then, this vector is again re-ligated with a pC1300-Cas9 vector by ligation of BamHI and KnpI-digested enzymes. The whole sgRNA cassette is then delivered into a pC1300-Cas9 vector (contains the Cas9 gene under the control of the 2 × 35S promoter) for plant transformation.
Ijms 23 04454 g004
Table 1. Comparison of CRISPER/Cas9 with ZFN and TALEN.
Table 1. Comparison of CRISPER/Cas9 with ZFN and TALEN.
Editing TechnologyDNA Binding DeterminantEndonucleaseTarget Length (bp)Off TargetingIntended EffectsUnintended Effects
CRISPR/Cas9crRNA/sgRNACas918–21VariableHighly specific, highly efficient, and multiple targeting sitesTarget selection limited by the requirement for PAM sequences; Off-target effects
ZFNZinc finger protein, FokI1018FokI18–36HighAny genomic sequence targeted; Fewer off-target effectsLow efficiency; Targets only a single site at one time
TALENsTranscription-activator like effectorFokI30–40LowTargets any genomic sequence; Off-target
effects are limited
Comparatively low inefficiency; Targets only a single site at one time; Sensitive to target DNA methylation
Table 4. A list of genes involved in a variety of agriculturally relevant parameters targeted by CRISPR/Cas9.
Table 4. A list of genes involved in a variety of agriculturally relevant parameters targeted by CRISPR/Cas9.
Application PerspectivesTargeted GenesMolecular FunctionsCas9 PromotersgRNA PromoterTransformation
Method
References
Yield and quality improvementGW2, GW5, and TGW6Improvement of grain weightOsUbi OsU3, OsU6, TaU3OsUbi OsU3, OsU6, TaU3Agrobacterium-mediated transformation[22]
Hd2, Hd4, and Hd5Early maturity of rice varietiesCas9 Pubi-HOsU3/U6a-[118]
GS9Yield ImprovementCaMV 35SOsU3Agrobacterium-mediated transformation[119]
OsGRF4Yield Improvement2 × 35SOsU6Agrobacterium-mediated transformation[120]
TMS5Photoperiod controlled male sterile lines-OsU3/U6Agrobacterium-mediated transformation[121]
TMS10Photo- and thermosensitive- Agrobacterium-mediated transformation[122]
CSAPhotoperiod-controlled male sterile lines- [123]
Gn1aGrain number; PaniclesZmUbiU6aAgrobacterium-mediated transformation[120]
DEP1Plant height; Erect panicles; Grain size
GS3Grain size
IPA1Plant height and tiller number
CCD7
PYLs
Increased tiller number; Improved growth and productivityOsUbiOsU3Agrobacterium-mediated transformation[124]
Lazy1Pronounced tiller spreadingOsUbi OsU3OsUbi OsU3Agrobacterium-mediated transformation[35]
OsPDS,
OsBADH2, Oso2g23823,
OsMPK2
Tolerance capacity against various abiotic stress factors2 × 35SOsU6Particle bombardment[4]
ISA1Quality improvementCaMV 35SOsU6Agrobacterium-mediated transformation[125]
YSAYoung albino seedlingsCaMV 35SOsU6–2Agrobacterium-mediated transformation[126]
PDSPhytoene Desaturase
DLDrooping Leaves
Chlorophyll A oxygenase (CAO I)Pale green leavesOsUbiOsU3Agrobacterium-mediated transformation[35]
ROCOutermost CellsCaMV 35SOsU6–2Agrobacterium-mediated transformation[19]
OsCYP97A4, OsDSM2,
OsCCD4a,
OsCCD4b, OsCCD7
Quality improvementCaMV 35SOsU3Agrobacterium-mediated transformation[126]
BADH2Enhanced fragranceCaMV 35SOsU3Agrobacterium-mediated transformation[127]
Biotic stress toleranceOsERF922Enhanced resistance to blast disease--Agrobacterium-mediated transformation[20]
OsAnn3Tolerance to cold stressCaMV 35SOsU6Agrobacterium-mediated transformation[7]
Bsrk1Disease resistanceCaMV 35SOsU6Agrobacterium-mediated transformation[128]
OsSWEET13Bacterial blight disease resistance---[129]
OsSWEET11, OsSWEET14Bacterial blight disease resistanceZmUbiOsU3Agrobacterium-mediated transformation[4,34]
Xa13Bacterial blight disease resistanceCaMV 35SOsU3 & OsU6Agrobacterium-mediated transformation[130]
eIF4GResistance to rice tungro spherical virusZmUbiTaU6Agrobacterium-mediated transformation[131]
OsMPK5Various abiotic stress tolerance and diseaseCaMV 35SOsU6Agrobacterium-mediated transformation[36]
Acetolactate synthase (ALS), DNA Ligase 4Disease resistance2 × 35SOsU6Expression plasmid vectors[10]
Abiotic stress toleranceBELHerbicide-resistant2 × 35SAtU6–26Agrobacterium-mediated transformation[21]
OsNAC041Salinity-- [9]
OsEPSPSGlyphosate-resistantCaMV 35SOsU3Agrobacterium-mediated transformation[132]
ALSHerbicide-resistant2 × 35SOsU6Agrobacterium-mediated transformation[133]
Bentazon Sensitive LethalPhenotypic analysis showed plants susceptible to bentazon--Agrobacterium-mediated transformation[21]
OsDERF1, OsPMS3,
OsEPSPS, OsMSH1,
OsMYB5
Drought toleranceCaMV 35S, OsUBQ1OsU6
OsU3
Agrobacterium-mediated transformation[134]
OsPYLDrought tolerance---[124]
OsAOX1a, OsAOX1b,
OsAOX1c
Various abiotic stress tolerance2 × 35SAtU6–26Agrobacterium-mediated transformation[135]
OsHAK-1Low cesium accumulationCaMV 35SOsU6aAgrobacterium-mediated transformation[136]
OsPRX2Potassium deficiency tolerance---[137]
OsSAPK2Drought tolerance---[6]
Nutritional improvementOsNramp5Low cadmium
content
CaMV 35SOsU6aAgrobacterium-mediated transformation[8]
ISA1StarchCaMV 35S Agrobacterium-mediated transformation[125]
OsWaxyAmylose synthaseCaMV 35SOsU6Agrobacterium-mediated transformation[138]
SBEIIb and SBEIGeneration of high amylose riceZmUbiOsU3Agrobacterium-mediated transformation[139]
lysC and dapALysine contentCaMV 35S-Agrobacterium-mediated transformation[140]
Stomatal densityOsEPFL9Regulates stomatal leaf density---[141]
Cyclin-dependent kinaseCDKA1, CDKA2, CDKB1-CaMV 35SOsU3Agrobacterium-mediated transformation[142]
CDKB2-2 × 35SOsU3Agrobacterium-mediated transformation[143]
Homologous pairing activityOsDMC1A, OsDMC1BDisrupted meiotic cDNA2 × 35SOsU3Agrobacterium-mediated transformation[144]
‘–’: information unavailable.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zegeye, W.A.; Tsegaw, M.; Zhang, Y.; Cao, L. CRISPR-Based Genome Editing: Advancements and Opportunities for Rice Improvement. Int. J. Mol. Sci. 2022, 23, 4454. https://doi.org/10.3390/ijms23084454

AMA Style

Zegeye WA, Tsegaw M, Zhang Y, Cao L. CRISPR-Based Genome Editing: Advancements and Opportunities for Rice Improvement. International Journal of Molecular Sciences. 2022; 23(8):4454. https://doi.org/10.3390/ijms23084454

Chicago/Turabian Style

Zegeye, Workie Anley, Mesfin Tsegaw, Yingxin Zhang, and Liyong Cao. 2022. "CRISPR-Based Genome Editing: Advancements and Opportunities for Rice Improvement" International Journal of Molecular Sciences 23, no. 8: 4454. https://doi.org/10.3390/ijms23084454

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

Article Metrics

Back to TopTop