Next Article in Journal
RrWRKY1, a Transcription Factor, Is Involved in the Regulation of the Salt Stress Response in Rosa rugosa
Next Article in Special Issue
Trans-Kingdom RNA Dialogues: miRNA and milRNA Networks as Biotechnological Tools for Sustainable Crop Defense and Pathogen Control
Previous Article in Journal
Mechanisms of Cannabis Growth Promotion by Bacillus velezensis S141
Previous Article in Special Issue
Exploring Novel Genomic Loci and Candidate Genes Associated with Plant Height in Bulgarian Bread Wheat via Multi-Model GWAS
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unleashing the Potential of CRISPR/Cas9 Genome Editing for Yield-Related Traits in Rice

by
Archana Thiruppathi
1,
Shubham Rajaram Salunkhe
1,
Shobica Priya Ramasamy
2,
Rakshana Palaniswamy
1,
Veera Ranjani Rajagopalan
1,
Sakthi Ambothi Rathnasamy
1,
Senthil Alagarswamy
3,
Manonmani Swaminathan
4,
Sudha Manickam
1,* and
Raveendran Muthurajan
1,*
1
Department of Plant Biotechnology, Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore 641003, India
2
Department of Plant Breeding and Genetics, Centre for Plant Breeding and Genetics, Tamil Nadu Agricultural University, Coimbatore 641003, India
3
Department of Crop Physiology, Tamil Nadu Agricultural University, Coimbatore 641003, India
4
Department of Rice, Centre for Plant Breeding and Genetics, Tamil Nadu Agricultural University, Coimbatore 641003, India
*
Authors to whom correspondence should be addressed.
Plants 2024, 13(21), 2972; https://doi.org/10.3390/plants13212972
Submission received: 20 September 2024 / Revised: 18 October 2024 / Accepted: 22 October 2024 / Published: 24 October 2024

Abstract

:
Strategies to enhance rice productivity in response to global demand have been the paramount focus of breeders worldwide. Multiple factors, including agronomical traits such as plant architecture and grain formation and physiological traits such as photosynthetic efficiency and NUE (nitrogen use efficiency), as well as factors such as phytohormone perception and homeostasis and transcriptional regulation, indirectly influence rice grain yield. Advances in genetic analysis methodologies and functional genomics, numerous genes, QTLs (Quantitative Trait Loci), and SNPs (Single-Nucleotide Polymorphisms), linked to yield traits, have been identified and analyzed in rice. Genome editing allows for the targeted modification of identified genes to create novel mutations in rice, avoiding the unintended mutations often caused by random mutagenesis. Genome editing technologies, notably the CRISPR/Cas9 system, present a promising tool to generate precise and rapid modifications in the plant genome. Advancements in CRISPR have further enabled researchers to modify a larger number of genes with higher efficiency. This paper reviews recent research on genome editing of yield-related genes in rice, discusses available gene editing tools, and highlights their potential to expedite rice breeding programs.

1. Introduction

The rising global population poses a significant challenge to ensuring food security, demanding a collective effort to enhance productivity. Rice, being one of the major food crops, plays a pivotal role in providing nourishment for over fifty percent of the world’s population. To meet the increasing nutritional requirements, qualitative traits of rice like culm habits and quantitative traits such as grain yield, grain size, etc., must be improved [1]. Rice is also adopted as a model system in plant science due to its compact genome size, well-established molecular marker linkage maps, and the availability of efficient transformation technologies [2].
Various approaches have been utilized to enhance rice quality and quantity. Conventional breeding methods, such as mutational breeding and hybridization methods, have been used to improve rice varieties; however, they are considered tedious, time-consuming and inclined to human bias [3]. These approaches may introduce undesirable genes alongside targeted genes, and hybridization is restricted to plants within the same species. While other techniques like genetic engineering and molecular approaches have also shown promise in enhancing crop varieties, they come with certain limitations as well, such as ethical concerns, potential environmental risks, genetic instability, etc. In recent years, genome editing technologies have addressed the constraints associated with traditional breeding methods and can quickly introduce desirable traits into any plant species including rice, wheat, barley, cowpea, chickpea, cotton, tomato, etc., in a short time. Consequently, they hold significant potential to accelerate breeding programs [4].
Genome editing has ushered in a new era of genome engineering, facilitating the efficient and accurate modification of plant genomes in a remarkably short time frame [5]. The technique relies on site-specific nucleases (SSNs), encompassing mega-nucleases, zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced palindromic repeats (CRISPRs)/CRISPR-associated-Cas9 systems. These nucleases are utilized to induce double-strand breaks (DSBs) at targeted sites of DNA. The desired mutations can then be generated through the endogenous, error-prone non-homologous end joining (NHEJ) or homology-directed repair (HDR) pathway. Therefore, precise modifications to a target gene, including specific substitutions, insertions and deletions of a desired sequence, can be carried out with greater accuracy [6,7].
The CRISPR/Cas9 system is considered an effective genome editing tool due to its potential to generate novel modifications to a target gene with relative ease and high efficiency [8]. Another significant benefit of the CRISPR/Cas9 system is its ability to target multiple genes or specific sites within a gene, facilitating the creation of small or large deletions in the genome [9]. CRISPR/Cas9 has proven to be highly effective in enhancing agricultural traits, particularly agronomic traits, which ultimately result in increased crop yield. This review delves into recent genome editing research focusing on genes associated with yield traits and their alterations to enhance rice crop improvement. Additionally, it mentions the challenges posed by genome editing technologies and explores potential solutions to surpass them.

2. Yield and Its Component Traits

Grain yield, the culmination of various agronomic factors and genetic traits, stands as a foremost aim of rice agriculture and is influenced by numerous genes operating within complex signaling pathways. It can be determined by three primary traits: number of grains per panicle, number of panicles per plant (tillering ability of a plant), and grain weight [2]. Plant height can indirectly influence grain yield by affecting the ability of plants to absorb sunlight and compete for resources with neighboring plants [10]. Aside from these components, factors such as grain filling, panicle architecture, and leaf orientation play crucial roles in determining the rice grain yield (Figure 1). The extent and rate of grain filling are predictive of the final grain weight and are essential for both the grain quality and yield [11]. Nitrogen use efficiency (NUE) and photosynthetic efficiency are two physiological traits that greatly impact grain yield [12]. Phytohormone signaling, stress tolerance, diverse pathways for plant growth and developmental and transcriptional regulation are additional factors that could potentially influence rice grain yield. Therefore, agronomic traits, such as plant architecture (leaf angle, tiller angle, tiller number, plant height, panicle type, etc.) and grain formation (grain number, grain weight and grain filling), and the above-mentioned physiological traits are key factors that need to be considered to enhance the productivity of rice [13].

3. Agronomical Traits

3.1. Genes Controlling Rice Grain Number

The number of grains per panicle is an important trait for rice yield potential. It is primarily influenced by the type of panicle and the differentiation of branches, which are closely linked to various phytohormone pathways and vascular differentiation [14,15]. Studies have revealed that numerous genes from these pathways hold considerable promise for enhancing grain number and ultimately improving rice grain yield [16]. Gn1a codes for the enzyme cytokinin oxidase/dehydrogenase (OsCKX2), which is responsible for the degradation of cytokinin. When the expression level of OsCKX2 decreases, cytokinin levels increase in the inflorescence meristems, leading to a better reproductive organ and eventually resulting in a higher yield [17]. The CRISPR/Cas9 construct, aimed at targeting Gn1a, resulted in frame-shift mutants in Zhonghua 11 (rice cultivar) and showed increased plant height, panicle size and flower number per panicle [18].
“Seed size/seed number trade-off” is a well-known phenomenon observed not only in rice but also in many plant species. There exists a negative correlation between the size of seeds and their number, which is attributed to metabolic constraints [19]. When plants have limited resources available for reproduction, if they invest more resources to produce a larger number of seeds, then the seed size will tend to decrease, and vice versa [20]. The GSN1 gene encodes the mitogen-activated protein kinase (MAPK) phosphatase enzyme (OsMPK1); it is found to be “the molecular brake” precisely governing both grain size and grain number and changes in the expression of the gene directly influence the panicle type and grain shape. The characterization of the rice mutant grain size and number1 (gsn1) highlights that they exhibit larger grains but are fewer in number compared to the wild type due to the presence of disordered localized cell differentiation and proliferation. Thus, to put it another way, GSN1 promotes grain number but inhibits grain size. GSN1 directly interacts with OsMPK6 and inactivates the enzyme via dephosphorylation. Suppression of the MAPK cascade (OsMPKK10-OsMPK4-OsMPK6) genes (via CRISPR/Cas9 and RNAi), which participate in panicle morphogenesis, resulted in denser panicles with reduced grain size, thus effectively alleviating the phenotypes observed in the gsn1 mutant. Hence, GSN1 also acts as a negative regulator of the MAPK cascade, which influences the panicle architecture. Ultimately, the validation of the GSN1-MAPK module confirms its role in coordinating the trade-off between grain number and grain size in rice [21]. Another gene, OsPUB3, appears to coordinate the trade-off between grain weight and yield. When the OsPUB3 gene was knocked out, it reduced the grain weight but increased the number of grains per panicle and the number of panicles per plant, thus stabilizing the overall yield of a plant [22].
Spikelet number in a panicle is another important factor which influences the number of grains per panicle. Recently, it has been documented that the number of spikelets in a panicle can be controlled by modulating the expression level of Floral Organ Number (FON4). It was reported that fon4, a mutant allele generated via the CRISPR/Cas9 system, is responsible for forming lateral florets alongside the normal terminal florets. Thus, mutations in the FON4 gene have the capability to induce the formation of spikelets with multiple flowers, consequently resulting in the production of multiple seeds [23].
The 5′ untranslated region (UTR) significantly contributes to the regulation of gene expression [24]. FRIZZY PANICLE (FZP) encodes an AP2/ERF (ethylene response factor) transcription factor and is involved in the formation of panicle branching in rice [25]. The deletion of a specific 113bp segment (−157 to −45bp in UTR) upstream of the FZP gene showed increased grain numbers in rice [26].
DENSE ERECT PANICLE (DEP1), encoding the G protein ẟ subunit, has been a crucial trait in rice crop improvement due to its advantageous characteristics such as high yield, lodging tolerance conferred by robust stems, and efficient nitrogen utilization. DEP1 plays an important role in regulating panicle architecture, grain number, nitrogen absorption and stress tolerance through the G protein signaling pathway [27]. Mutants of DEP1 exhibit a modified plant architecture characterized by a reduced plant height and grain size, dense erect panicle, and increased grain number and density [18,28]. Table 1 lists additional genes governing the rice grain number, analyzed by the CRISPR/Cas9 system, in addition to those already stated.

3.2. Genes Controlling Rice Grain Weight

Grain weight in rice is predominantly influenced by grain size, which is defined by three dimensions: grain length, width and thickness, as well as the extent of filling [2]. These characteristics exert such a profound impact on grain weight that they all exhibit potential trade-offs and collectively control both the grain quality and quantity [31]. Recent advances have revealed numerous pathways that significantly influence both grain number and size, including G-protein signaling; the ubiquitin-proteasome degradation pathway; biosynthesis and signaling of brassinosteroids, cytokinins, and auxins; MAPK signaling; photosynthetic product accumulation; epigenetic pathways; and transcriptional regulation [32,33]. Therefore, manipulating the genes that participate in the above-mentioned pathways is an effective strategy for improving grain size and number, which eventually enhances rice grain yields.
GRAIN SIZE 3 (GS3) was the first major QTL (Quantitative Trait Locus) identified to influence the grain size and quality in rice [34]. GS3 encodes the γ subunit of heterotrimeric G-protein, which regulates cell proliferation and expansion of spikelet hull via the G-protein signaling pathway, thereby limiting grain elongation in rice [35]. The GS3 protein consists of four major domains: a plant-specific organ size regulation (OSR) domain located at the N-terminus, a transmembrane domain, a cysteine-rich domain from the tumor necrosis factor receptor/nerve growth factor receptor (TNFR/NGFR) family, and a von Willebrand factor type C (VWFC) located at the C-terminus. These domains play distinct roles in the regulation of grain size [34]. It has been reported that the gs3 allele, featuring a mutation in the OSR domain of the GS3 protein, leads to the production of longer grains. Conversely, a frameshift mutation lacking the TNFR and VWFC domains yielded the shortest grains, suggesting potential antagonistic effects of these domains in regulating grain size [34,36]. The frameshift mutants in the rice cultivar Zhonghua 11, generated via the CRISPR/Cas9 system, exhibited a substantial increase in grain size and had elongated awns on the husks compared to the wild type [18]. Thus, GS3 functions as a negative regulator of rice grain size [37,38].
The GW5 gene, encoding a calmodulin binding protein, exerts a greater impact on grain width and weight by influencing the brassinosteroid (BR) signaling pathway [39]. The GW5 protein localizes within the plasma membrane and interacts with GSK2 (glycogen synthase kinase2), effectively inhibiting its activity. This consequential interaction leads to the buildup of OsBZR1 (Oryza sativa BRASSINAZOLE RESISTANT1) and DLT (DWARF AND LOW TILLERING) proteins in their unphosphorylated states within the nucleus, facilitating the mediation of the BR signaling pathway. Ultimately, this accumulation activates genes involved in the pathway, thereby facilitating the growth responses, including grain width and grain weight in rice. Thus, GW5 acts as a positive regulator of BR signaling and knocking out the gene significantly increased grain weight and width [40]. Another gene, GW5L, a homolog of GW5, negatively regulates grain width and size in rice [41].
qTGW3 is another QTL that governs grain size and weight in rice. qTGW3 encodes OsSK41/OsGSK5, a member of the GLYCOGEN SYNTHASE KINASE3/SHAGGY-like family. OsSK41 effectively interacts with and activates OsARF4 (AUXIN REPONSE FACTOR 4), a gene involved in rice grain development. Loss-of-function of both OsSK41 and OsARF4 has resulted in a larger grain size in rice. This suggests that OsARF4 and OsSK41 might operate in the same pathway regulating grain size, and their expression patterns are also notably similar. However, further investigation is required to determine if the function of OsSK41 in grain development is entirely reliant on OsARF4 [42].
In another study, CRISPR/Cas9-based editing was used to generate two superior alleles of OsSPL4, which increased the grain number and the grain size, resulting in higher rice yield. OsSPL4 was also found to influence spikelet development by stimulating cell division, leading to the upregulation of various cell cycle and MADS-box genes in the mutated lines. Analysis of the co-expression network unveiled the involvement of numerous yield-related genes in the regulatory network of OsSPL4. Furthermore, OsSPL4 was observed to undergo cleavage by os-miR156 in living organisms, suggesting that the OsmiR156-OsSPL4 module may play a role in controlling rice grain size. In addition to the aforementioned genes, Table 2 lists a number of other genes that govern rice grain weight that have been analyzed by CRISPR/Cas9.

3.3. Genes Controlling Rice Tiller Number

Tillering in rice plants constitutes a fundamental determinant of grain yield and serves as a model system for investigating branching in monocotyledonous plants [64]. Additionally, it is an essential agronomic characteristic that governs tolerance to plant density and resistance to lodging [13]. Tillering formation can be divided into two process: the formation of a new AM (axillary meristem) on the leaf axil and its subsequent growth [65]. Each tiller has the ability to generate a panicle. Nonetheless, tillers that appear later in the growing season demonstrate incomplete grain filling, leading to an increase in straw biomass alone. Hence, it is essential to carefully control the number of tillers to maximize potential yield [10].
MONOCULM 1 (MOC1) and MONOCULM 3/TILLERS ABSENT 1/STERILE AND REDUCED TILLERING 1 (MOC3/TAB1/SRT1) are two important genes that are essential for the development of tillers in rice. They interact with FLORAL ORGAN NUMBER1 (FON1), the rice homolog of the CLAVATA1 gene, which regulates tiller bud outgrowth in rice. MOC3 directly engages with the promoter of FON1, facilitating its expression. In contrast, MOC1 does not establish a direct interaction with FON1; rather, it functions as a co-activator of MOC3 to promote FON1 expression. Mutants of MOC1 and MOC3 showed significantly reduced expression of FON1 in axillary meristems. Therefore, MOC1 and MOC3 genes positively regulate tillering in rice [66,67]. Genes governing rice tiller number, which have been examined using CRISPR/Cas9, are listed in Table 3.

3.4. Genes Controlling Biotic and Abiotic Stress Resistance in Rice

Worldwide, it is estimated that crop yield losses due to plant diseases range from 20% to 50% [77]. These losses result from various pathogens, including fungi, bacteria and viruses, and they considerably affect global agricultural productivity. The pathogens Magnaporthe oryzae and Xanthomonas oryzae pv. oryzae are responsible for causing rice blast and bacterial blight, respectively, both of which are highly destructive diseases that significantly reduce rice yields [78]. Additionally, abiotic stresses, including drought, salinity and extreme temperatures, are significant factors that hamper crop growth and development, further contributing to yield losses. Therefore, improving resistance to both biotic and abiotic stresses, using advanced techniques like CRISPR-Cas9, has proven to be the most efficient and cost-effective strategy for managing various plant stresses, ensuring sustainability and enhancing crop quality [79].
In a study, CRISPR/Cas9 was used for multiplex gene editing to simultaneously mutate three blast-resistant genes, such as Bsr-d1, Pi21 and ERF922, in an indica rice line (LK638S). The resulting single and triple mutants exhibited enhanced resistance to rice blast, with erf922 mutants showing the strongest resistance. Moreover, Pi21 and ERF922 mutants demonstrated improved resistance to bacterial blight. This increased resistance was attributed to the upregulation of salicylic acid (SA) and jasmonic acid (JA) pathways. Notably, the mutations had no adverse effects on key agricultural traits, highlighting the effectiveness of multiplex gene editing for developing disease-resistant rice varieties [80].
Abiotic stress adversely impacts plant growth and crop productivity by disrupting various biochemical, morphological, and physiological processes that are essential for plant development [81]. Targeting the OsPUB7 gene (a Plant U-box gene in Oryza sativa) with CRISPR/Cas9 resulted in increased resistance to drought and salinity stress in rice [82]. The OsPQT3 knockout mutants (ospqt3) demonstrate enhanced resistance to oxidative and salt stress, along with significantly improved agronomic performance, yielding more than the wild type when exposed to salt stress [83]. Additionally, several genes that contribute to biotic and abiotic stress resistance in rice, which have been studied using CRISPR-Cas9, are listed in Table 4.

3.5. Genes Controlling Herbicide Resistance in Rice

Another crucial trait that can significantly influence rice yield is herbicide resistance. The ability of rice plants to tolerate herbicides enables more efficient weed control, preventing weeds from competing for essential resources such as water, light and nutrients. Weeds are a major yield-limiting factor in rice production, often causing substantial yield losses if not effectively managed [97]. Moreover, herbicide-resistant rice varieties contribute to more sustainable farming practices by reducing the need for excessive herbicide applications and labor-intensive weed management techniques. Numerous studies have highlighted the adaptation of CRISPR/Cas technology to develop new herbicide-resistant lines [98], which are mentioned in Table 4.
The three polyamine uptake transporter (PUT) genes in rice, including OsPUT1, OsPUT2 and OsPUT3, homologous to the Arabidopsis AtRMV1, were successfully mutated using CRISPR/Cas9, resulting in increased resistance to the herbicide paraquat without any yield penalty [99]. A significant chromosomal fragment inversion (911 kb) and a 338 kb duplication were induced in the elite rice variety Jingeng 818 using CRISPR-Cas9. This process replaced the native promoters of PPOI and HPPD in rice, which are typically expressed at low levels in leaf tissue, with strong promoters of other linked genes in the mutant lines. As a result of these structural modifications, the OxPPOI and OxHPPD genes are highly expressed, conferring resistance to the herbicides FCD and bipyrazone [100].

4. Physiological Traits

Nitrogen, phosphorus and potassium are vital macronutrients for rice, with nitrogen fertilizer consumption at 15%, phosphorus at 13% and potassium at 11% of all fertilizers [101]. Photosynthesis in rice plants directly impacts yield by providing metabolic energy. Optimizing nutrient use efficiency (NUE) and enhancing photosynthetic efficiency are crucial for boosting productivity. Improved NUE maximizes nitrogen utilization, minimizing environmental losses, while enhanced photosynthesis drives growth and development by converting solar energy into chemical energy. Understanding the molecular mechanisms behind these processes is essential for sustainable increases in rice yield [12].

4.1. Genes Controlling NUE in Rice

Ammonium (NH4+) and nitrate (NO3) are the two major nitrogen forms that can be available to plants [102]. OsHHO3 belongs to the NIGT1/HHO subgroup within the GARP/G2-like transcription factor family found in rice, which comprises a nitrate-inducible NIGT1 protein and four nitrate non-inducible HHO proteins in rice [103]. The knockout of this gene via the CRISPR/Cas9 system generated seedlings with enhanced growth and increased shoot and root dry mass due to the increase in chlorophyll content and maximum quantum yield of photosystem II under N-deficient conditions. Transcriptome analysis of these mutant seedlings revealed an increase in the expression of genes encoding ammonium transporter (AMT) and nitrate transporter (NRT), as well as other genes related to nitrogen assimilation, in the roots of oshho3-KO mutants. Within these overexpressed genes, the upregulation of the three AMT1 genes, namely OsAMT1.1, OsAMT1.2 and OsAMT1.3, is very critical due to their influence on 95% of ammonium uptake in rice [104]. Thus, the knockout of OsHHO3, a transcriptional repressor of AMT1 genes, acts as negative regulator of nitrogen use efficiency in rice [105]. Table 5 enumerates the genes governing NUE, analyzed by CRISPR/Cas9.

4.2. Genes Controlling Photosynthetic Efficiency in Rice

Photosynthesis takes place in the chloroplast, whose function and development are governed by many genes. RNA editing plays a crucial role as a post-transcriptional process in plant organelles [108]. Among them, PPR (Pentatricopeptide repeat) is a major protein coded by the OsPPR9 gene, participating in RNA editing [109]. Knockout of the gene through the CRISPR/Cas9 system showed decreased expression of genes involved in chloroplast development and proteins related to photosynthesis. Additionally, OsPPR9 effectively interacts with OsMORF2 and OsMORF9, genes that encode multiple organellar RNA editing factor (MORF) proteins which are significant for RNA editing [110,111]. Table 6 lists the genes that regulate photosynthetic efficiency which have been examined by CRISPR/Cas9.

5. Targeting Regulatory DNA Regions for Improving Rice Yield

Genome editing tools have provided precise and efficient approaches for editing target genes, and substantial advancements have been achieved in editing the protein-coding part of the target gene. However, studies pertaining to the modifications of non-coding DNA regions with regulatory roles significantly trail behind. The non-coding parts of DNA, including those capable of being transcribed into miRNAs and long non-coding RNAs (lncRNAs), along with cis-regulatory elements (CREs) such as promoters, enhancers, silencers, transcription factor binding sites and introns, assume indispensable functions in the regulation of plant growth and development [117,118]. Promoters are crucial in regulating when, where, and how strongly genes are expressed. They contain regulatory elements that serve as binding sites for proteins such as transcription factors and RNA polymerases. Genome editing can modify promoter regions, allowing precise control over gene expression, which can impact rice traits like yield (Table 7), quality and biotic and abiotic stress resistance [119]. So, targeting these regions of a gene for editing will expand the application of genome editing tools in rice. Figure 2 highlights the yield-related genes edited by genome editing technologies.

6. CRISPR-Cas: Advantages over Other Gene Editing Tools

ZFNs and TALENs consist of a DNA-binding domain paired with the FokI enzyme, requiring extensive protein engineering to target new sequences. In contrast, the CRISPR/Cas9 system uses the Cas9 protein, which is guided by RNA to bind specific target sequences, allowing for simpler targeting by modifying the guide RNA (sgRNA) rather than engineering proteins [126]. Additionally, multiple sgRNAs can function with the same Cas9 protein, enabling simultaneous targeting of diverse DNA sequences, greatly enhancing the flexibility, efficiency, and versatility of genome editing with CRISPR/Cas9 [127]. CRISPR is compatible with various delivery methods, including plasmids, viral vectors, and ribonucleoprotein (RNP) complexes. CRISPR’s ability to be delivered as RNP complexes allows for a more immediate and transient expression of the editing compo-nents, which can reduce off-target effects and improve editing efficiency [128]. On the other hand, ZFNs and TALENs face limitations in delivery options and may have lower efficiency when delivered as proteins due to their larger size and complex assembly [129]. CRISPR/Cas9 demonstrates superior efficiency compared to ZFNs and TALENs, with reported editing activity ranging from 40 to 50% on average [130,131] and some studies showing rates as high as 73% in vitro [132]. Efficiency varies based on the specific Cas variant, sgRNA design, delivery method, and target site.
To broaden the versatility and to increase the range of applications for CRISPR technology, various advanced CRISPR-Cas tools, including base editing, prime editing, different Cas proteins, and multiple Cas9 orthologues, have been developed and successfully applied across a wide range of fields. Table 8 provides an overview of various Cas proteins, highlighting their distinct features and applications. For instance, Cas9 is widely used for its ability to introduce double-strand breaks in DNA, while Cas12a targets “AT”-rich regions in the genome, unlike Cas9, which typically targets GC-rich sequences. Cas13, on the other hand, specializes in RNA targeting, and Cas14 is known for its small size and potential for precise DNA editing. dCas9, a deactivated form of Cas9, is applied in gene regulation without cutting DNA, making it ideal for transcriptional modulation.
Base editing (BE) is an innovative and versatile genome editing (GE) system that allows for precise and highly predictable nucleotide modifications at genomic targets without the requirement for donor DNA templates, DSBs or dependence on HDR and NHEJ [138]. A base editor (BE) is formed by combining a catalytically inactive variant of Cas9 with a deaminase domain that targets either cytosine or adenine. Adenine base editors (ABEs) enable the conversion of an A-T base pair into a G-C pair, while cytosine base editors (CBEs) facilitate the transformation of a C-G base pair into a T-A pair [139]. Adenine base editing has been successfully applied to the yield gene OsSPL14 in rice, which is crucial for plant architecture and tiller number [140]. Prime editing is another GE technology that can effectively introduce all 12 possible types of point mutations, along with small insertions and deletions, in a precise and targeted way with advantageous editing to indel ratios. Prime editors are hybrid proteins that combine a Cas9 nickase domain (an inactive HNH nuclease) with an engineered reverse transcriptase domain [141,142].
Another key advantage of CRISPR/Cas9 and CRISPR/Cpf1 is their simplicity in multiplexing, especially when compared to ZFNs and TALENs [143]. Multiplexing using CRISPR allows for the simultaneous targeting of multiple genes or genomic loci within a single experiment, significantly enhancing the efficiency of genome editing. This approach enables researchers to study complex genetic interactions and perform comprehensive modifications, facilitating advancements in functional genomics and crop improvement. CRISPR/Cas9-based multiplex editing of quantitative trait loci (QTLs), specifically OsGS3, OsGW2 and OsGn1a, was performed across three elite rice varieties, J809, L237 and CNXJ, resulting in the successful generation of all seven combinations of single, double, and triple mutants for these targeted genes. Comprehensive analysis of these mutants revealed distinct effects on yield-related traits, including grain length, width, number, and 1000-grain weight. Notably, the triple mutants demonstrated a substantial increase in yield per panicle, with J809 achieving a remarkable 68% enhancement and L237 showing a 30% improvement [144]. Table 9 enumerates the advanced genome editing tools that have been utilized for the analysis of rice yield-related genes. Figure 3 presents the various gene editing tools including Cas variants, along with their advanced versions specifically available for use in plants, particularly rice. In summary, CRISPR-Cas technology provides significant advantages over other genome editing tools, including improved precision, simplified multiplexing and efficient mutation generation, making it a powerful platform for genetic research and crop enhancement.
CRISPR/Cas12a-RNP is an advanced genome editing system that utilizes the Cas12a protein complexed with ribonucleoprotein (RNP) to achieve precise modifications in plant genomes. This approach combines the targeting capabilities of CRISPR technology with the transient delivery of Cas12a and gRNA, allowing for efficient and specific editing while minimizing off-target effects. Unlike traditional DNA delivery methods, CRISPR/Cas12a-RNP is transgene-free and can enhance editing efficiency through controlled dosages and multiplexed targeting, making it a versatile tool for plant biotechnology [145,146]. To assess the efficiency of different genome editing systems, a comparative study was performed using three Cas9 nucleases (WT Cas9, HiFi Cas9 and Cas9 D10A nickase) along with two Cas12a nucleases (AsCas12a and LbCas12a) targeting the rice phytoene desaturase (PDS) gene. The results revealed that the delivery of WT Cas9, HiFi Cas9 and LbCas12a led to targeted mutagenesis, with LbCas12a demonstrating superior editing efficiency compared to both Cas9 variants. Editing with Cas9 primarily resulted in small insertions or deletions (indels) of 1–2 bp and larger deletions of 20–30 bp, frequently accompanied by the loss of the PAM site. Conversely, LbCas12a editing produced deletions ranging from 2 to 20 bp without affecting the PAM site. In summary, LbCas12a RNP complexes achieved a higher frequency of targeted mutagenesis at the OsPDS gene than AsCas12a or Cas9 RNPs [147].
Table 9. Different gene editing tools are employed to target rice yield genes.
Table 9. Different gene editing tools are employed to target rice yield genes.
GeneGene Editing ToolTraitReference
OsSPL14Base editingTiller number[140]
OsEPFL9CRISPR-LbCpf1 (Lachnospiracae bacterium Cpf1),
CRISPR-FnCpf1 (Francisella novicida Cpf1)
Stomatal development[148,149]
OsDEP1CRISPR-LbCpf1,
CRISPR-FnCpf1
Plant architecture and grain number[149,150,151]
OsGS3CRISPR-LbCpf1Grain size[152]
OsDEP1Prime editingPlant architecture and grain number[153]
OsSPL14Prime editingTiller number[153]
OsEPFL9CRISPR/Cas12a-RNPStomatal development[145]
NRT1.1BBase editingEnhanced nitrogen use efficiency[154]
C287Base editingHerbicide resistance[155]
GL2/OsGRF4, OsGRF3Base editingGrain size and yield[156]
OsACC-T1CRISPR–Cpf1-based base editingHerbicide resistance[157]
OsALSPrime editingHerbicide resistance[158,159]
TFIIAg5, OsSWEET11a, OsEPSPS1 and OsALS1Multiplexing—quadruple prime editingBroad spectrum resistance to bacterial blight and herbicide[160]
OsSPL13, OsSPL14 and OsGS2Multiplex prime editingMajor yield traits—grain size and weight, plant architecture, tiller number and NUE[160]
OsSWEET14Base editng (CBE)Resistance to bacterial blight[161]
OsDEP1, OsNRT1.1b, OsWaxyT1, OsWaxyT2 and OsWaxyT3Adenine base transition editor (ABE8e)Panicle architecture, NUE, starch biosynthesis[162]
OsYSA, OsNAL, OsMIR396e, and OsPYL6CRIPSR/Cas12i3-based multiplex direct repeat (DR)-spacer Array Genome Editing system (iMAGE)Chloroplast development, grain number, yield[163]
OsACCaseDeactivated Cas12i3 base editorResistant to sethoxydim herbicide[163]
OsGRF4Prime editingGrain yield[164]

7. Off-Target Effects of CRISPR/Cas9 on Plant Physiology

The physiological impacts of CRISPR on plants largely stem from off-target mutations, which are a significant concern when evaluating the safety of genome-edited crops. These unintended genetic modifications can affect plant phenotypes or interact unpredictably with their environment. Reducing off-target frequencies is essential, and mismatches between the seed sequence (the 12 base pairs adjacent to the PAM) and the target sequence are critical in achieving this reduction [165]. In a study, multiplex CRISPR/Cas9 was employed to mutate genes like OsPIN5b (affecting panicle length), GS3 (influencing grain size) and OsMYB30 (related to cold tolerance). The editing efficiency across target sites ranged between 42% and 66%. Furthermore, eight triple mutants were developed, six of which exhibited off-target effects [37]. These off-targets can be minimized by refining gRNA design to increase specificity (numerous sgRNA design tools, such as CGAT, CRISPR-P, CHOPCHOP and CRISPR, help identify specific sgRNA sequences to improve targeting and reduce off-target impacts) [166,167]. Additionally, novel genome editing systems are used, including nickase-Cas9, base editing (C to T, A to G), CRISPR-Cpf1-RNP (recombinant CRISPR-Cpf1 ribonucleoprotein), tru-gRNAs (shorter/truncated guide RNAs for on-target site), SpCsa9-HF1 (high-fidelity engineered variants SpCas9), eSpCsa9 (enhanced specificity of SpCas9), Hypa-Cas9 (hyper-accurate Cas9 variant), C2c2/Cas13, SauCas9, C2c1, evoCas9 (evolved Cas9) [168]. For instance, the cytidine base editor (CBE) developed mutants resistant to African Xanthomonas oryzae pv. oryzae (Xoo) strains by targeting the SWEET14 gene in rice with no off-targets [161]. Another study demonstrated that CRISPR-Cpf1 successfully induces targeted genome mutagenesis in rice for the OsPDS and OsBEL genes without any off-target effects. This indicates that, with careful selection of target sites, the CRISPR-Cpf1 system exhibits high specificity in vivo [169]. Similarly, a truncated gRNA (tru-gRNA)/Cas9 strategy successfully produced new alleles for the proton pump gene OST2 in Arabidopsis without off-target effects. Monitoring the expression of Cas9 and tru-gRNA revealed a high mutation rate of 32.8% in transgenic plants, with no off-target impacts observed under a constitutive promoter [170]. These systems have been employed to reduce off-target editing, although research on them in plants lags far behind.

8. Challenges and Future Prospects in Genome Editing

Genome editing has demonstrated its effectiveness as a tool for enhancing crops throughout the preceding decade. However, along with its beneficial features, it also presents certain challenges. Mitigating these challenges can expand their applications in plant breeding programs. The first challenge resides in the development of efficient transformation systems for various crop species. The second challenge is to reduce the requirement of a unique canonical NGG PAM (protospacer adjacent motif) site by the SpCas9 (wild-type) system, which limits the flexibility and target site of the system [171]. Nevertheless, the identification of alternative PAM sites (NAG, NGA, etc.) and Cas variants like SaCas9 (Staphylococcus aureus Cas9) can promote the applications of genome editing [172,173]. The development of Cas9 variants such as VQR (NGA PAM) and VRER (NGCG PAM) has broadened the range of genome editing in rice [174,175]. Aside from these, it has been reported that the wild-type SpCas9 in itself is effective in targeting both NGG and NAG PAM sites in rice with high efficiency and relatively low off-targets in rice [176]. SpRY, another new variant of SpCas, was created to greatly increase BE (base editing) editing capabilities to almost PAMless [132]. In rice, scientists have made attempts to use SpRYn-CBE (cytosine base editor) and SpRYn-ABE (adenine base editor) and revealed the effective editing activities of SpRY on various PAM sites, including NCN, NTH, NAG, NAC, NAB, NCR, NTK and NGV, thus presenting them as promising alternatives in genome editing [177]. All these developments will contribute to widening the scope of genome editing in most cereal crops, especially rice, by using different engineered Cas9 variants with distinct PAM specifics. However, there is still a need for the development of more Cas9 variants to target a diverse array of PAM sites, as not all Cas9 variants work well with the plant system.
The third challenge is to minimize off-targets, as these unintended modifications can result in various non-quantifiable cellular signaling changes and physiological impacts in plants. To minimize off-target effects, several strategies are employed, including optimizing the design of guide RNAs to enhance specificity, using Cas proteins with higher accuracy (such as Cas9 variants like SpCas9-HF1 or eSpCas9), employing paired nickases and utilizing high-fidelity base editors [165]. Following off-targets, the next issue to be addressed is the system’s precision, which can be attained by targeting genes using HDR. The efficacy of the HDR pathway is comparatively low in plants, and their constraint lies in the lack of an effective delivery system for DNA template repair [139]. The next challenge is gaining widespread acceptance of gene-edited crops among consumers and regulators. Despite their potential benefits, public concerns about safety and ethics persist. Addressing this requires transparent communication, robust regulatory frameworks, and clear demonstrations of benefits to farmers and consumers, alongside efforts to build trust through education [178]. In CRISPRa, CRISPR activation gene transcription is boosted by fusing dCas9 (catalytically inactivated Cas variants (dCas9)) with activators or using scaffold RNA to recruit them. In CRISPRi, CRISPR interference dCas9 binds the TSS with inhibitors like KRAB or SRDX, disrupting transcription factor and RNA polymerase activity to regulate gene expression [179,180]. Cas-CLOVER is one of the advanced gene editing technologies that can be effectively utilized for precise genome modifications and targeted gene editing in various organisms, including plants. CLOVER is a dual-guided system that induces double-strand breaks through the dimerization of its nuclease components, and this technique has been successfully applied in bananas [181,182]. Therefore, it holds potential for exploitation in rice as well. Multiplex genome editing has been used to alter many genes at once and to understand the interaction among genes, as many yield traits in rice are interconnected [183]. However, the number of targets that genome editing tools can manipulate simultaneously is limited. Sequential editing [184] can be conducted to overcome these constraints.

9. Conclusions

Genetic editing in rice, with specific emphasis on genes linked to yield characteristics, possesses the potential to accelerate the process of rice breeding, considering the advancement in genome editing techniques and the emergence of novel discoveries, indicating a pivotal role in hastening crop improvement. By leveraging genome editing tools, especially the CRISPR/Cas9 system, researchers can precisely modify the candidate genes governing yield determination. The benefits of genome editing tools include excising transgenes from the genome by employing genetic segregation, leaving the resulting gene-edited plants entirely indistinguishable from those produced through traditional breeding methods. Understanding the complex genetic networks involving major yield traits and their responses to various environmental factors by integrating them with different omics approaches can provide comprehensive insights into the molecular mechanisms underlying the yield genes and enable researchers to create novel alleles in rice research. Investigating the role of non-coding regions, such as promoters, enhancers and non-translated regions, in regulating yield genes can aid in understanding gene expression and the respective yield modifications. Comprehensive studies analyzing the simultaneous modification (multiplexing) of several characteristic genes to optimize rice yield are still lacking. Future research should prioritize comprehensive studies aimed at understanding epistatic interactions among different yield-related genes. Overall, integrating genome editing tools such as the CRISPR/Cas9 system into rice breeding programs offers a transformative pathway towards sustainable enhancement in rice production.

Author Contributions

R.M. and S.M.: Conceptualization; A.T.: literature collection, writing—original draft preparation; M.S., S.A., S.R.S., S.P.R., R.P., V.R.R. and S.A.R.: Reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank the Department of Plant Biotechnology, Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, for the facilities and support provided in preparing the article.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Zafar, K.; Sedeek, K.E.; Rao, G.S.; Khan, M.Z.; Amin, I.; Kamel, R.; Mukhtar, Z.; Zafar, M.; Mansoor, S.; Mahfouz, M.M. Genome editing technologies for rice improvement: Progress, prospects, and safety concerns. Front. Genome Ed. 2020, 2, 5. [Google Scholar] [CrossRef] [PubMed]
  2. Xing, Y.; Zhang, Q. Genetic and molecular bases of rice yield. Annu. Rev. Plant Biol. 2010, 61, 421–442. [Google Scholar] [CrossRef]
  3. Ahmar, S.; Gill, R.A.; Jung, K.-H.; Faheem, A.; Qasim, M.U.; Mubeen, M.; Zhou, W. Conventional and molecular techniques from simple breeding to speed breeding in crop plants: Recent advances and future outlook. Int. J. Mol. Sci. 2020, 21, 2590. [Google Scholar] [CrossRef] [PubMed]
  4. Tabassum, J.; Ahmad, S.; Hussain, B.; Mawia, A.M.; Zeb, A.; Ju, L. Applications and potential of genome-editing systems in rice improvement: Current and future perspectives. Agronomy 2021, 11, 1359. [Google Scholar] [CrossRef]
  5. Endo, M.; Toki, S. Genome Editing in Rice; Springer: Cham, Switzerland, 2020; Volume 13, pp. 1–2. [Google Scholar]
  6. Osakabe, Y.; Osakabe, K. Genome editing with engineered nucleases in plants. Plant Cell Physiol. 2015, 56, 389–400. [Google Scholar] [CrossRef]
  7. Symington, L.S.; Gautier, J. Double-strand break end resection and repair pathway choice. Annu. Rev. Genet. 2011, 45, 247–271. [Google Scholar] [CrossRef]
  8. Hussain, B.; Lucas, S.J.; Budak, H. CRISPR/Cas9 in plants: At play in the genome and at work for crop improvement. Brief. Funct. Genom. 2018, 17, 319–328. [Google Scholar] [CrossRef]
  9. Vats, S.; Kumawat, S.; Kumar, V.; Patil, G.B.; Joshi, T.; Sonah, H.; Sharma, T.R.; Deshmukh, R. Genome editing in plants: Exploration of technological advancements and challenges. Cells 2019, 8, 1386. [Google Scholar] [CrossRef] [PubMed]
  10. Sakamoto, T.; Matsuoka, M. Identifying and exploiting grain yield genes in rice. Curr. Opin. Plant Biol. 2008, 11, 209–214. [Google Scholar] [CrossRef]
  11. Yang, J.; Zhang, J. Grain-filling problem in ‘super’rice. J. Exp. Bot. 2010, 61, 1–5. [Google Scholar] [CrossRef]
  12. Li, G.; Tang, J.; Zheng, J.; Chu, C. Exploration of rice yield potential: Decoding agronomic and physiological traits. Crop J. 2021, 9, 577–589. [Google Scholar] [CrossRef]
  13. Zhong, Q.; Jia, Q.; Yin, W.; Wang, Y.; Rao, Y.; Mao, Y. Advances in cloning functional genes for rice yield traits and molecular design breeding in China. Front. Plant Sci. 2023, 14, 1206165. [Google Scholar] [CrossRef] [PubMed]
  14. Deveshwar, P.; Prusty, A.; Sharma, S.; Tyagi, A.K. Phytohormone-mediated molecular mechanisms involving multiple genes and QTL govern grain number in rice. Front. Genet. 2020, 11, 586462. [Google Scholar] [CrossRef] [PubMed]
  15. Terao, T.; Nagata, K.; Morino, K.; Hirose, T. A gene controlling the number of primary rachis branches also controls the vascular bundle formation and hence is responsible to increase the harvest index and grain yield in rice. Theor. Appl. Genet. 2010, 120, 875–893. [Google Scholar] [CrossRef]
  16. Lu, Y.; Chuan, M.; Wang, H.; Chen, R.; Tao, T.; Zhou, Y.; Xu, Y.; Li, P.; Yao, Y.; Xu, C. Genetic and molecular factors in determining grain number per panicle of rice. Front. Plant Sci. 2022, 13, 964246. [Google Scholar] [CrossRef] [PubMed]
  17. Ashikari, M.; Sakakibara, H.; Lin, S.; Yamamoto, T.; Takashi, T.; Nishimura, A.; Angeles, E.R.; Qian, Q.; Kitano, H.; Matsuoka, M. Cytokinin oxidase regulates rice grain production. Science 2005, 309, 741–745. [Google Scholar] [CrossRef]
  18. 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]
  19. Paul-Victor, C.; Turnbull, L.A. The effect of growth conditions on the seed size/number trade-off. PLoS ONE 2009, 4, e6917. [Google Scholar] [CrossRef]
  20. Gasparis, S.; Miłoszewski, M.M. Genetic Basis of Grain Size and Weight in Rice, Wheat, and Barley. Int. J. Mol. Sci. 2023, 24, 16921. [Google Scholar] [CrossRef]
  21. Guo, T.; Chen, K.; Dong, N.-Q.; Shi, C.-L.; Ye, W.-W.; Gao, J.-P.; Shan, J.-X.; Lin, H.-X. GRAIN SIZE AND NUMBER1 negatively regulates the OsMKKK10-OsMKK4-OsMPK6 cascade to coordinate the trade-off between grain number per panicle and grain size in rice. Plant Cell 2018, 30, 871–888. [Google Scholar] [CrossRef]
  22. Li, Z.-H.; Wang, S.-L.; Zhu, Y.-J.; Fan, Y.-Y.; Huang, D.-R.; Zhu, A.-K.; Zhuang, J.-Y.; Liang, Y.; Zhang, Z.-H. Control of Grain Shape and Size in Rice by Two Functional Alleles of OsPUB3 in Varied Genetic Background. Plants 2022, 11, 2530. [Google Scholar] [CrossRef] [PubMed]
  23. Ren, D.; Xu, Q.; Qiu, Z.; Cui, Y.; Zhou, T.; Zeng, D.; Guo, L.; Qian, Q. FON4 prevents the multi-floret spikelet in rice. Plant Biotechnol. J. 2019, 17, 1007. [Google Scholar] [CrossRef]
  24. Lakshmi Jayaraj, K.; Thulasidharan, N.; Antony, A.; John, M.; Augustine, R.; Chakravartty, N.; Sukumaran, S.; Uma Maheswari, M.; Abraham, S.; Thomas, G. Targeted editing of tomato carotenoid isomerase reveals the role of 5′ UTR region in gene expression regulation. Plant Cell Rep. 2021, 40, 621–635. [Google Scholar] [CrossRef]
  25. Li, J.; Zhang, L.; Elbaiomy, R.G.; Chen, L.; Wang, Z.; Jiao, J.; Zhu, J.; Zhou, W.; Chen, B.; Soaud, S.A. Evolution analysis of FRIZZY PANICLE (FZP) orthologs explored the mutations in DNA coding sequences in the grass family (Poaceae). PeerJ 2022, 10, e12880. [Google Scholar] [CrossRef]
  26. Chen, H.; Cai, Y.; Zhang, S.; Tang, W.; Fang, X.; Zhang, Y. Identification of a novel mutant allele, fzp-15, involved in panicle branch pattern of rice (Oryza sativa). Plant Breed. 2021, 140, 595–602. [Google Scholar] [CrossRef]
  27. Xu, H.; Zhao, M.; Zhang, Q.; Xu, Z.; Xu, Q. The DENSE AND ERECT PANICLE 1 (DEP1) gene offering the potential in the breeding of high-yielding rice. Breed. Sci. 2016, 66, 659–667. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, A.; Wang, F.; Kong, D.; Hou, D.; Huang, L.; Bi, J.; Zhang, F.; Luo, X.; Wang, J.; Liu, G. Mutation of DEP1 facilitates the improvement of plant architecture in Xian rice (Oryza sativa). Plant Breed. 2023, 142, 338–344. [Google Scholar] [CrossRef]
  29. Chu, H.; Qian, Q.; Liang, W.; Yin, C.; Tan, H.; Yao, X.; Yuan, Z.; Yang, J.; Huang, H.; Luo, D. The floral organ number4 gene encoding a putative ortholog of Arabidopsis CLAVATA3 regulates apical meristem size in rice. Plant Physiol. 2006, 142, 1039–1052. [Google Scholar] [CrossRef]
  30. Huang, L.; Hua, K.; Xu, R.; Zeng, D.; Wang, R.; Dong, G.; Zhang, G.; Lu, X.; Fang, N.; Wang, D. The LARGE2-APO1/APO2 regulatory module controls panicle size and grain number in rice. Plant Cell 2021, 33, 1212–1228. [Google Scholar] [CrossRef]
  31. Chen, K.; Łyskowski, A.; Jaremko, Ł.; Jaremko, M. Genetic and molecular factors determining grain weight in rice. Front. Plant Sci. 2021, 12, 605799. [Google Scholar] [CrossRef]
  32. Fan, Y.; Li, Y. Molecular, cellular and Yin-Yang regulation of grain size and number in rice. Mol. Breed. 2019, 39, 163. [Google Scholar] [CrossRef]
  33. Li, N.; Xu, R.; Li, Y. Molecular networks of seed size control in plants. Annu. Rev. Plant Biol. 2019, 70, 435–463. [Google Scholar] [CrossRef] [PubMed]
  34. Mao, H.; Sun, S.; Yao, J.; Wang, C.; Yu, S.; Xu, C.; Li, X.; Zhang, Q. Linking differential domain functions of the GS3 protein to natural variation of grain size in rice. Proc. Natl. Acad. Sci. USA 2010, 107, 19579–19584. [Google Scholar] [CrossRef] [PubMed]
  35. Sun, S.; Wang, L.; Mao, H.; Shao, L.; Li, X.; Xiao, J.; Ouyang, Y.; Zhang, Q. A G-protein pathway determines grain size in rice. Nat. Commun. 2018, 9, 851. [Google Scholar] [CrossRef]
  36. Fan, C.; Xing, Y.; Mao, H.; Lu, T.; Han, B.; Xu, C.; Li, X.; Zhang, Q. GS3, a major QTL for grain length and weight and minor QTL for grain width and thickness in rice, encodes a putative transmembrane protein. Theor. Appl. Genet. 2006, 112, 1164–1171. [Google Scholar] [CrossRef]
  37. Zeng, Y.; Wen, J.; Zhao, W.; Wang, Q.; Huang, W. Rational improvement of rice yield and cold tolerance by editing the three genes OsPIN5b, GS3, and OsMYB30 with the CRISPR–Cas9 system. Front. Plant Sci. 2020, 10, 1663. [Google Scholar] [CrossRef]
  38. Yuyu, C.; Aike, Z.; Pao, X.; Xiaoxia, W.; Yongrun, C.; Beifang, W.; Yue, Z.; Liaqat, S.; Shihua, C.; Liyong, C. Effects of GS3 and GL3. 1 for grain size editing by CRISPR/Cas9 in rice. Rice Sci. 2020, 27, 405–413. [Google Scholar] [CrossRef]
  39. Weng, J.; Gu, S.; Wan, X.; Gao, H.; Guo, T.; Su, N.; Lei, C.; Zhang, X.; Cheng, Z.; Guo, X. Isolation and initial characterization of GW5, a major QTL associated with rice grain width and weight. Cell Res. 2008, 18, 1199–1209. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, J.; Chen, J.; Zheng, X.; Wu, F.; Lin, Q.; Heng, Y.; Tian, P.; Cheng, Z.; Yu, X.; Zhou, K. GW5 acts in the brassinosteroid signalling pathway to regulate grain width and weight in rice. Nat. Plants 2017, 3, 17043. [Google Scholar] [CrossRef]
  41. Tian, P.; Liu, J.; Mou, C.; Shi, C.; Zhang, H.; Zhao, Z.; Lin, Q.; Wang, J.; Wang, J.; Zhang, X. GW5-Like, a homolog of GW5, negatively regulates grain width, weight and salt resistance in rice. J. Integr. Plant Biol. 2019, 61, 1171–1185. [Google Scholar] [CrossRef]
  42. Hu, Z.; Lu, S.-J.; Wang, M.-J.; He, H.; Sun, L.; Wang, H.; Liu, X.-H.; Jiang, L.; Sun, J.-L.; Xin, X. A novel QTL qTGW3 encodes the GSK3/SHAGGY-like kinase OsGSK5/OsSK41 that interacts with OsARF4 to negatively regulate grain size and weight in rice. Mol. Plant 2018, 11, 736–749. [Google Scholar] [CrossRef] [PubMed]
  43. Usman, B.; Zhao, N.; Nawaz, G.; Qin, B.; Liu, F.; Liu, Y.; Li, R. CRISPR/Cas9 guided mutagenesis of grain size 3 confers increased rice (Oryza sativa L.) grain length by regulating cysteine proteinase inhibitor and ubiquitin-related proteins. Int. J. Mol. Sci. 2021, 22, 3225. [Google Scholar] [CrossRef] [PubMed]
  44. Achary, V.M.M.; Reddy, M.K. CRISPR-Cas9 mediated mutation in GRAIN WIDTH and WEIGHT2 (GW2) locus improves aleurone layer and grain nutritional quality in rice. Sci. Rep. 2021, 11, 21941. [Google Scholar] [CrossRef] [PubMed]
  45. Qing, D.; Chen, W.; Huang, S.; Li, J.; Pan, Y.; Zhou, W.; Liang, Q.; Yuan, J.; Gan, D.; Chen, L. Editing of rice (Oryza sativa L.) OsMKK3 gene using CRISPR/Cas9 decreases grain length by modulating the expression of photosystem components. Proteomics 2023, 23, 2200538. [Google Scholar] [CrossRef]
  46. Ren, D.; Hu, J.; Xu, Q.; Cui, Y.; Zhang, Y.; Zhou, T.; Rao, Y.; Xue, D.; Zeng, D.; Zhang, G. FZP determines grain size and sterile lemma fate in rice. J. Exp. Bot. 2018, 69, 4853–4866. [Google Scholar] [CrossRef]
  47. Miao, C.; Wang, D.; He, R.; Liu, S.; Zhu, J.K. Mutations in MIR 396e and MIR 396f increase grain size and modulate shoot architecture in rice. Plant Biotechnol. J. 2020, 18, 491–501. [Google Scholar] [CrossRef]
  48. Tang, J.; Mei, E.; He, M.; Bu, Q.; Tian, X. Functions of OsWRKY24, OsWRKY70 and OsWRKY53 in regulating grain size in rice. Planta 2022, 255, 92. [Google Scholar] [CrossRef]
  49. Guo, M.; Liu, J.; Hou, L.; Zhao, S.; Zhang, N.; Lu, L.; Zhao, X. The mitochondria-localized protein OsNDB2 negatively regulates grain size and weight in rice. Crop J. 2022, 10, 1819–1824. [Google Scholar] [CrossRef]
  50. Shao, Y.; Zhou, H.-Z.; Wu, Y.; Zhang, H.; Lin, J.; Jiang, X.; He, Q.; Zhu, J.; Li, Y.; Yu, H. OsSPL3, an SBP-domain protein, regulates crown root development in rice. Plant Cell 2019, 31, 1257–1275. [Google Scholar] [CrossRef]
  51. Hu, J.; Huang, L.; Chen, G.; Liu, H.; Zhang, Y.; Zhang, R.; Zhang, S.; Liu, J.; Hu, Q.; Hu, F. The elite alleles of OsSPL4 regulate grain size and increase grain yield in rice. Rice 2021, 14, 90. [Google Scholar] [CrossRef]
  52. Wang, A.; Hou, Q.; Si, L.; Huang, X.; Luo, J.; Lu, D.; Zhu, J.; Shangguan, Y.; Miao, J.; Xie, Y. The PLATZ transcription factor GL6 affects grain length and number in rice. Plant Physiol. 2019, 180, 2077–2090. [Google Scholar] [CrossRef] [PubMed]
  53. Zhan, P.; Ma, S.; Xiao, Z.; Li, F.; Wei, X.; Lin, S.; Wang, X.; Ji, Z.; Fu, Y.; Pan, J. Natural variations in grain length 10 (GL10) regulate rice grain size. J. Genet. Genom. 2022, 49, 405–413. [Google Scholar] [CrossRef]
  54. Zhang, L.; Wang, R.; Xing, Y.; Xu, Y.; Xiong, D.; Wang, Y.; Yao, S. Separable regulation of POW1 in grain size and leaf angle development in rice. Plant Biotechnol. J. 2021, 19, 2517–2531. [Google Scholar] [CrossRef]
  55. Deng, X.; Han, X.; Yu, S.; Liu, Z.; Guo, D.; He, Y.; Li, W.; Tao, Y.; Sun, C.; Xu, P. OsINV3 and its homolog, OsINV2, control grain size in rice. Int. J. Mol. Sci. 2020, 21, 2199. [Google Scholar] [CrossRef]
  56. Zhan, P.; Wei, X.; Xiao, Z.; Wang, X.; Ma, S.; Lin, S.; Li, F.; Bu, S.; Liu, Z.; Zhu, H. GW10, a member of P450 subfamily regulates grain size and grain number in rice. Theor. Appl. Genet. 2021, 134, 3941–3950. [Google Scholar] [CrossRef]
  57. Zhong, J.; He, W.; Peng, Z.; Zhang, H.; Li, F.; Yao, J. A putative AGO protein, OsAGO17, positively regulates grain size and grain weight through OsmiR397b in rice. Plant Biotechnol. J. 2020, 18, 916–928. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, L.; Wang, D.; Yang, Z.; Jiang, S.; Qu, J.; He, W.; Liu, Z.; Xing, J.; Ma, Y.; Lin, Q. Roles of FERONIA-like receptor genes in regulating grain size and quality in rice. Sci. China Life Sci. 2021, 64, 294–310. [Google Scholar] [CrossRef]
  59. Shi, C.L.; Dong, N.Q.; Guo, T.; Ye, W.W.; Shan, J.X.; Lin, H.X. A quantitative trait locus GW6 controls rice grain size and yield through the gibberellin pathway. Plant J. 2020, 103, 1174–1188. [Google Scholar] [CrossRef]
  60. Bai, F.; Ma, H.; Cai, Y.; Shahid, M.Q.; Zheng, Y.; Lang, C.; Chen, Z.; Wu, J.; Liu, X.; Wang, L. Natural Allelic Variation in GRAIN SIZE AND WEIGHT 3 of Wild Rice Regulates the Grain Size and Weight. Plant Physiol. 2023, 193, 502–518. [Google Scholar] [CrossRef] [PubMed]
  61. Du, Z.; Huang, Z.; Li, J.; Bao, J.; Tu, H.; Zeng, C.; Wu, Z.; Fu, H.; Xu, J.; Zhou, D. qTGW12 a, a naturally varying QTL, regulates grain weight in rice. Theor. Appl. Genet. 2021, 134, 2767–2776. [Google Scholar] [CrossRef]
  62. Wen, Y.; Hu, P.; Fang, Y.; Tan, Y.; Wang, Y.; Wu, H.; Wang, J.; Wu, K.; Chai, B.; Zhu, L. GW9 determines grain size and floral organ identity in rice. Plant Biotechnol. J. 2023, 22, 915–928. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, S.-L.; Zhang, Z.-H.; Fan, Y.-Y.; Huang, D.-R.; Yang, Y.-L.; Zhuang, J.-Y.; Zhu, Y.-J. Control of Grain Weight and Size in Rice (Oryza sativa L.) by OsPUB3 Encoding a U-Box E3 Ubiquitin Ligase. Rice 2022, 15, 58. [Google Scholar] [CrossRef]
  64. Li, X.; Qian, Q.; Fu, Z.; Wang, Y.; Xiong, G.; Zeng, D.; Wang, X.; Liu, X.; Teng, S.; Hiroshi, F. Control of tillering in rice. Nature 2003, 422, 618–621. [Google Scholar] [CrossRef]
  65. Yan, Y.; Ding, C.; Zhang, G.; Hu, J.; Zhu, L.; Zeng, D.; Qian, Q.; Ren, D. Genetic and environmental control of rice tillering. Crop J. 2023, 11, 1287–1302. [Google Scholar] [CrossRef]
  66. Shao, G.; Lu, Z.; Xiong, J.; Wang, B.; Jing, Y.; Meng, X.; Liu, G.; Ma, H.; Liang, Y.; Chen, F. Tiller bud formation regulators MOC1 and MOC3 cooperatively promote tiller bud outgrowth by activating FON1 expression in rice. Mol. Plant 2019, 12, 1090–1102. [Google Scholar] [CrossRef] [PubMed]
  67. Liang, W.-h.; Shang, F.; Lin, Q.-t.; Lou, C.; Zhang, J. Tillering and panicle branching genes in rice. Gene 2014, 537, 1–5. [Google Scholar] [CrossRef]
  68. Gao, Q.; Li, G.; Sun, H.; Xu, M.; Wang, H.; Ji, J.; Wang, D.; Yuan, C.; Zhao, X. Targeted mutagenesis of the rice FW 2.2-like gene family using the CRISPR/Cas9 system reveals OsFWL4 as a regulator of tiller number and plant yield in rice. Int. J. Mol. Sci. 2020, 21, 809. [Google Scholar] [CrossRef]
  69. Taylor, M.R.; Reinders, A.; Ward, J.M. Transport Function of Rice Amino Acid Permeases (AAPs). Plant Cell Physiol. 2015, 56, 1355–1363. [Google Scholar] [CrossRef]
  70. Lu, K.; Wu, B.; Wang, J.; Zhu, W.; Nie, H.; Qian, J.; Huang, W.; Fang, Z. Blocking amino acid transporter Os AAP 3 improves grain yield by promoting outgrowth buds and increasing tiller number in rice. Plant Biotechnol. J. 2018, 16, 1710–1722. [Google Scholar] [CrossRef]
  71. Huang, W.; Nie, H.; Feng, F.; Wang, J.; Lu, K.; Fang, Z. Altered expression of OsNPF7. 1 and OsNPF7. 4 differentially regulates tillering and grain yield in rice. Plant Sci. 2019, 283, 23–31. [Google Scholar] [CrossRef]
  72. Song, X.; Lu, Z.; Yu, H.; Shao, G.; Xiong, J.; Meng, X.; Jing, Y.; Liu, G.; Xiong, G.; Duan, J. IPA1 functions as a downstream transcription factor repressed by D53 in strigolactone signaling in rice. Cell Res. 2017, 27, 1128–1141. [Google Scholar] [CrossRef]
  73. Butt, H.; Jamil, M.; Wang, J.Y.; Al-Babili, S.; Mahfouz, M. Engineering plant architecture via CRISPR/Cas9-mediated alteration of strigolactone biosynthesis. BMC Plant Biol. 2018, 18, 174. [Google Scholar] [CrossRef]
  74. Yang, X.; Chen, L.; He, J.; 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] [PubMed]
  75. Mo, T.; Wang, T.; Sun, Y.; Kumar, A.; Mkumbwa, H.; Fang, J.; Zhao, J.; Yuan, S.; Li, Z.; Li, X. The chloroplast pentatricopeptide repeat protein RCN22 regulates tiller number in rice by affecting sugar levels via the TB1-RCN22-RbcL module. Plant Commun. 2024, 101073. [Google Scholar] [CrossRef]
  76. Song, J.; Tang, L.; Fan, H.; Xu, X.; Peng, X.; Cui, Y.; Wang, J. Enhancing Yield and Improving Grain Quality in Japonica Rice: Targeted EHD1 Editing via CRISPR-Cas9 in Low-Latitude Adaptation. Curr. Issues Mol. Biol. 2024, 46, 3741–3751. [Google Scholar] [CrossRef]
  77. Ahmad, S.; Wei, X.; Sheng, Z.; Hu, P.; Tang, S. CRISPR/Cas9 for development of disease resistance in plants: Recent progress, limitations and future prospects. Brief. Funct. Genom. 2020, 19, 26–39. [Google Scholar] [CrossRef] [PubMed]
  78. Yin, X.; Zou, B.; Hong, X.; Gao, M.; Yang, W.; Zhong, X.; He, Y.; Kuai, P.; Lou, Y.; Huang, J. Rice copine genes Os BON 1 and Os BON 3 function as suppressors of broad-spectrum disease resistance. Plant Biotechnol. J. 2018, 16, 1476–1487. [Google Scholar] [CrossRef] [PubMed]
  79. Dhadge, K.; Walia, P. Applications of CRISPR/Cas9 for biotic and abiotic stress resistance of rice (Oryza sativa). J. Adv. Biol. Biotechnol. 2024, 27, 172–179. [Google Scholar] [CrossRef]
  80. Zhou, Y.; Xu, S.; Jiang, N.; Zhao, X.; Bai, Z.; Liu, J.; Yao, W.; Tang, Q.; Xiao, G.; Lv, C. Engineering of rice varieties with enhanced resistances to both blast and bacterial blight diseases via CRISPR/Cas9. Plant Biotechnol. J. 2022, 20, 876–885. [Google Scholar] [CrossRef]
  81. Jha, U.C.; Bohra, A.; Nayyar, H. Advances in “omics” approaches to tackle drought stress in grain legumes. Plant Breed. 2020, 139, 1–27. [Google Scholar] [CrossRef]
  82. Kim, M.-S.; Ko, S.-R.; Jung, Y.J.; Kang, K.-K.; Lee, Y.-J.; Cho, Y.-G. Knockout mutants of OsPUB7 generated using CRISPR/Cas9 revealed abiotic stress tolerance in rice. Int. J. Mol. Sci. 2023, 24, 5338. [Google Scholar] [CrossRef] [PubMed]
  83. Alfatih, A.; Wu, J.; Jan, S.U.; Zhang, Z.S.; Xia, J.Q.; Xiang, C.B. Loss of rice PARAQUAT TOLERANCE 3 confers enhanced resistance to abiotic stresses and increases grain yield in field. Plant Cell Environ. 2020, 43, 2743–2754. [Google Scholar] [CrossRef] [PubMed]
  84. Zhou, J.; Peng, Z.; Long, J.; Sosso, D.; Liu, B.; Eom, J.S.; Huang, S.; Liu, S.; Vera Cruz, C.; Frommer, W.B. Gene targeting by the TAL effector PthXo2 reveals cryptic resistance gene for bacterial blight of rice. Plant J. 2015, 82, 632–643. [Google Scholar] [CrossRef]
  85. 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]
  86. Oliva, R.; Ji, C.; Atienza-Grande, G.; Huguet-Tapia, J.C.; Perez-Quintero, A.; Li, T.; Eom, J.-S.; Li, C.; Nguyen, H.; Liu, B. Broad-spectrum resistance to bacterial blight in rice using genome editing. Nat. Biotechnol. 2019, 37, 1344–1350. [Google Scholar] [CrossRef]
  87. Foster, A.J.; Martin-Urdiroz, M.; Yan, X.; Wright, H.S.; Soanes, D.M.; Talbot, N.J. CRISPR-Cas9 ribonucleoprotein-mediated co-editing and counterselection in the rice blast fungus. Sci. Rep. 2018, 8, 14355. [Google Scholar] [CrossRef]
  88. Li, C.; Li, W.; Zhou, Z.; Chen, H.; Xie, C.; Lin, Y. A new rice breeding method: CRISPR/Cas9 system editing of the Xa13 promoter to cultivate transgene-free bacterial blight-resistant rice. Plant Biotechnol. J. 2020, 18, 313. [Google Scholar] [CrossRef]
  89. Macovei, A.; Sevilla, N.R.; Cantos, C.; Jonson, G.B.; Slamet-Loedin, I.; Čermák, T.; Voytas, D.F.; Choi, I.R.; Chadha-Mohanty, P. Novel alleles of rice eIF4G generated by CRISPR/Cas9-targeted mutagenesis confer resistance to Rice tungro spherical virus. Plant Biotechnol. J. 2018, 16, 1918–1927. [Google Scholar] [CrossRef] [PubMed]
  90. Arra, Y.; Auguy, F.; Stiebner, M.; Chéron, S.; Wudick, M.M.; Miras, M.; Schepler-Luu, V.; Köhler, S.; Cunnac, S.; Frommer, W.B. Rice Yellow Mottle Virus resistance by genome editing of the Oryza sativa L. ssp. japonica nucleoporin gene OsCPR5.1 but not OsCPR5.2. Plant Biotechnol. J. 2024, 22, 1299–1311. [Google Scholar]
  91. Ogata, T.; Ishizaki, T.; Fujita, M.; Fujita, Y. CRISPR/Cas9-targeted mutagenesis of OsERA1 confers enhanced responses to abscisic acid and drought stress and increased primary root growth under nonstressed conditions in rice. PLoS ONE 2020, 15, e0243376. [Google Scholar] [CrossRef]
  92. Santosh Kumar, V.; Verma, R.K.; Yadav, S.K.; Yadav, P.; Watts, A.; Rao, M.; Chinnusamy, V. CRISPR-Cas9 mediated genome editing of drought and salt tolerance (OsDST) gene in indica mega rice cultivar MTU1010. Physiol. Mol. Biol. Plants 2020, 26, 1099–1110. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, B.; Zhong, Z.; Wang, X.; Han, X.; Yu, D.; Wang, C.; Song, W.; Zheng, X.; Chen, C.; Zhang, Y. Knockout of the OsNAC006 transcription factor causes drought and heat sensitivity in rice. Int. J. Mol. Sci. 2020, 21, 2288. [Google Scholar] [CrossRef]
  94. Nawaz, G.; Han, Y.; Usman, B.; Liu, F.; Qin, B.; Li, R. Knockout of OsPRP1, a gene encoding proline-rich protein, confers enhanced cold sensitivity in rice (Oryza sativa L.) at the seedling stage. 3 Biotech 2019, 9, 254. [Google Scholar] [CrossRef] [PubMed]
  95. 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]
  96. 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, 2, 16139. [Google Scholar] [CrossRef]
  97. Hussain, A.; Ding, X.; Alariqi, M.; Manghwar, H.; Hui, F.; Li, Y.; Cheng, J.; Wu, C.; Cao, J.; Jin, S. Herbicide resistance: Another hot agronomic trait for plant genome editing. Plants 2021, 10, 621. [Google Scholar] [CrossRef]
  98. Romero, F.M.; Gatica-Arias, A. CRISPR/Cas9: Development and application in rice breeding. Rice Sci. 2019, 26, 265–281. [Google Scholar] [CrossRef]
  99. Lyu, Y.-S.; Cao, L.-M.; Huang, W.-Q.; Liu, J.-X.; Lu, H.-P. Disruption of three polyamine uptake transporter genes in rice by CRISPR/Cas9 gene editing confers tolerance to herbicide paraquat. Abiotech 2022, 3, 140–145. [Google Scholar] [CrossRef]
  100. Lu, Y.; Wang, J.; Chen, B.; Mo, S.; Lian, L.; Luo, Y.; Ding, D.; Ding, Y.; Cao, Q.; Li, Y. A donor-DNA-free CRISPR/Cas-based approach to gene knock-up in rice. Nat. Plants 2021, 7, 1445–1452. [Google Scholar] [CrossRef]
  101. Zhang, Z.; Gao, S.; Chu, C. Improvement of nutrient use efficiency in rice: Current toolbox and future perspectives. Theor. Appl. Genet. 2020, 133, 1365–1384. [Google Scholar] [CrossRef]
  102. Xu, G.; Fan, X.; Miller, A.J. Plant nitrogen assimilation and use efficiency. Annu. Rev. Plant Biol. 2012, 63, 153–182. [Google Scholar] [CrossRef] [PubMed]
  103. Ueda, Y.; Ohtsuki, N.; Kadota, K.; Tezuka, A.; Nagano, A.J.; Kadowaki, T.; Kim, Y.; Miyao, M.; Yanagisawa, S. Gene regulatory network and its constituent transcription factors that control nitrogen-deficiency responses in rice. New Phytol. 2020, 227, 1434–1452. [Google Scholar] [CrossRef]
  104. Konishi, N.; Ma, J.F. Three polarly localized ammonium transporter 1 members are cooperatively responsible for ammonium uptake in rice under low ammonium condition. New Phytol. 2021, 232, 1778–1792. [Google Scholar] [CrossRef]
  105. Liu, K.; Sakuraba, Y.; Ohtsuki, N.; Yang, M.; Ueda, Y.; Yanagisawa, S. CRISPR/Cas9-mediated elimination of OsHHO3, a transcriptional repressor of three AMMONIUM TRANSPORTER1 genes, improves nitrogen use efficiency in rice. Plant Biotechnol. J. 2023, 21, 2169. [Google Scholar] [CrossRef]
  106. Wakatabi, K.; Selvaraj, M.G.; Guzmán-Prada, D.A.; Cuásquer, J.B.; López-López, K.; Endo, M.; Ishitani, M. Selection and evaluation of gene-edited knockout mutants of AtAAP2 and AtCRF4 homologs of rice for agronomic nitrogen use efficiency (ANUE). Rev. Colomb. De Cienc. Hortícolas 2023, 17, e16120. [Google Scholar] [CrossRef]
  107. Zhu, Y.; Li, T.; Xu, J.; Wang, J.; Wang, L.; Zou, W.; Zeng, D.; Zhu, L.; Chen, G.; Hu, J. Leaf width gene LW5/D1 affects plant architecture and yield in rice by regulating nitrogen utilization efficiency. Plant Physiol. Biochem. 2020, 157, 359–369. [Google Scholar] [CrossRef] [PubMed]
  108. Yan, J.; Zhang, Q.; Yin, P. RNA editing machinery in plant organelles. Sci. China Life Sci. 2018, 61, 162–169. [Google Scholar] [CrossRef]
  109. Wang, Y.; Yang, Z.; Zhang, M.; Ai, P. A chloroplast-localized pentatricopeptide repeat protein involved in RNA editing and splicing and its effects on chloroplast development in rice. BMC Plant Biol. 2022, 22, 437. [Google Scholar] [CrossRef]
  110. Small, I.D.; Schallenberg-Rüdinger, M.; Takenaka, M.; Mireau, H.; Ostersetzer-Biran, O. Plant organellar RNA editing: What 30 years of research has revealed. Plant J. 2020, 101, 1040–1056. [Google Scholar] [CrossRef]
  111. Chen, C.-Z.; Wang, Y.-L.; He, M.-X.; Li, Z.-W.; Lan, S.; Qing, L.; Ren, D.-Y.; Jiang, H.; Li, Z.; Zhang, G.-H. OsPPR9 encodes a DYW-type PPR protein that affects editing efficiency of multiple RNA editing sites and is essential for chloroplast development. J. Integr. Agric. 2023, 22, 972–980. [Google Scholar] [CrossRef]
  112. Zheng, S.; Ye, C.; Lu, J.; Liufu, J.; Lin, L.; Dong, Z.; Li, J.; Zhuang, C. Improving the rice photosynthetic efficiency and yield by editing OsHXK1 via CRISPR/Cas9 system. Int. J. Mol. Sci. 2021, 22, 9554. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Q.; Wang, Y.; Xie, W.; Chen, C.; Ren, D.; Hu, J.; Zhu, L.; Zhang, G.; Gao, Z.; Guo, L. OsMORF9 is necessary for chloroplast development and seedling survival in rice. Plant Sci. 2021, 307, 110907. [Google Scholar] [CrossRef] [PubMed]
  114. Jung, Y.J.; Lee, H.J.; Yu, J.; Bae, S.; Cho, Y.-G.; Kang, K.K. Transcriptomic and physiological analysis of OsCAO1 knockout lines using the CRISPR/Cas9 system in rice. Plant Cell Rep. 2021, 40, 1013–1024. [Google Scholar] [CrossRef] [PubMed]
  115. Caddell, D.; Langenfeld, N.J.; Eckels, M.J.; Zhen, S.; Klaras, R.; Mishra, L.; Bugbee, B.; Coleman-Derr, D. Photosynthesis in rice is increased by CRISPR/Cas9-mediated transformation of two truncated light-harvesting antenna. Front. Plant Sci. 2023, 14, 1050483. [Google Scholar] [CrossRef]
  116. Rathnasamy, S.A.; Kambale, R.; Elangovan, A.; Mohanavel, W.; Shanmugavel, P.; Ramasamy, G.; Alagarsamy, S.; Marimuthu, R.; Rajagopalan, V.R.; Manickam, S. Altering Stomatal Density for Manipulating Transpiration and Photosynthetic Traits in Rice through CRISPR/Cas9 Mutagenesis. Curr. Issues Mol. Biol. 2023, 45, 3801–3814. [Google Scholar] [CrossRef]
  117. Chen, Y.-H.; Lu, J.; Yang, X.; Huang, L.-C.; Zhang, C.-Q.; Liu, Q.-Q.; Li, Q.-F. Gene editing of non-coding regulatory DNA and its application in crop improvement. J. Exp. Bot. 2023, 74, 6158–6175. [Google Scholar] [CrossRef]
  118. Basak, J.; Nithin, C. Targeting non-coding RNAs in plants with the CRISPR-Cas technology is a challenge yet worth accepting. Front. Plant Sci. 2015, 6, 1001. [Google Scholar] [CrossRef]
  119. Wu, B.; Luo, H.; Chen, Z.; Amin, B.; Yang, M.; Li, Z.; Wu, S.; Salmen, S.H.; Alharbi, S.A.; Fang, Z. Rice Promoter Editing: An Efficient Genetic Improvement Strategy. Rice 2024, 17, 55. [Google Scholar] [CrossRef]
  120. Li, H.; Zhang, Y.; Wu, C.; Bi, J.; Chen, Y.; Jiang, C.; Cui, M.; Chen, Y.; Hou, X.; Yuan, M. Fine-tuning OsCPK18/OsCPK4 activity via genome editing of phosphorylation motif improves rice yield and immunity. Plant Biotechnol. J. 2022, 20, 2258–2271. [Google Scholar] [CrossRef]
  121. Dong, S.; Dong, X.; Han, X.; Zhang, F.; Zhu, Y.; Xin, X.; Wang, Y.; Hu, Y.; Yuan, D.; Wang, J. OsPDCD5 negatively regulates plant architecture and grain yield in rice. Proc. Natl. Acad. Sci. USA 2021, 118, e2018799118. [Google Scholar] [CrossRef]
  122. 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. 2020, 22, 249. [Google Scholar] [CrossRef] [PubMed]
  123. Li, S.; Luo, Y.; Wei, G.; Zong, W.; Zeng, W.; Xiao, D.; Zhang, H.; Song, Y.; Hao, Y.; Sun, K. Improving yield-related traits by editing the promoter of the heading date gene Ehd1 in rice. Theor. Appl. Genet. 2023, 136, 239. [Google Scholar] [CrossRef] [PubMed]
  124. Usman, B.; Nawaz, G.; Zhao, N.; Liao, S.; Liu, Y.; Li, R. Precise editing of the OsPYL9 gene by RNA-guided Cas9 nuclease confers enhanced drought tolerance and grain yield in rice (Oryza sativa L.) by regulating circadian rhythm and abiotic stress responsive proteins. Int. J. Mol. Sci. 2020, 21, 7854. [Google Scholar] [CrossRef]
  125. Song, X.; Meng, X.; Guo, H.; Cheng, Q.; Jing, Y.; Chen, M.; Liu, G.; Wang, B.; Wang, Y.; Li, J. Targeting a gene regulatory element enhances rice grain yield by decoupling panicle number and size. Nat. Biotechnol. 2022, 40, 1403–1411. [Google Scholar] [CrossRef]
  126. 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]
  127. Wu, X.; Scott, D.A.; Kriz, A.J.; Chiu, A.C.; Hsu, P.D.; Dadon, D.B.; Cheng, A.W.; Trevino, A.E.; Konermann, S.; Chen, S. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 2014, 32, 670–676. [Google Scholar] [CrossRef] [PubMed]
  128. Yip, B.H. Recent advances in CRISPR/Cas9 delivery strategies. Biomolecules 2020, 10, 839. [Google Scholar] [CrossRef]
  129. González Castro, N.; Bjelic, J.; Malhotra, G.; Huang, C.; Alsaffar, S.H. Comparison of the feasibility, efficiency, and safety of genome editing technologies. Int. J. Mol. Sci. 2021, 22, 10355. [Google Scholar] [CrossRef]
  130. 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]
  131. Kim, N.; Kim, H.K.; Lee, S.; Seo, J.H.; Choi, J.W.; Park, J.; Min, S.; Yoon, S.; Cho, S.-R.; Kim, H.H. Prediction of the sequence-specific cleavage activity of Cas9 variants. Nat. Biotechnol. 2020, 38, 1328–1336. [Google Scholar] [CrossRef]
  132. Walton, R.T.; Christie, K.A.; Whittaker, M.N.; Kleinstiver, B.P. Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science 2020, 368, 290–296. [Google Scholar] [CrossRef] [PubMed]
  133. 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]
  134. Bandyopadhyay, A.; Kancharla, N.; Javalkote, V.S.; Dasgupta, S.; Brutnell, T.P. CRISPR-Cas12a (Cpf1): A versatile tool in the plant genome editing tool box for agricultural advancement. Front. Plant Sci. 2020, 11, 584151. [Google Scholar] [CrossRef]
  135. Liu, S.; Sretenovic, S.; Fan, T.; Cheng, Y.; Li, G.; Qi, A.; Tang, X.; Xu, Y.; Guo, W.; Zhong, Z. Hypercompact CRISPR–Cas12j2 (CasΦ) enables genome editing, gene activation, and epigenome editing in plants. Plant Commun. 2022, 3, 100453. [Google Scholar] [CrossRef] [PubMed]
  136. Kavuri, N.R.; Ramasamy, M.; Qi, Y.; Mandadi, K. Applications of CRISPR/Cas13-based RNA editing in plants. Cells 2022, 11, 2665. [Google Scholar] [CrossRef]
  137. 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]
  138. 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] [PubMed]
  139. Ahmed, S.; Zhang, Y.; Abdullah, M.; Ma, Q.; Wang, H.; Zhang, P. Current status, challenges, and future prospects of plant genome editing in China. Plant Biotechnol. Rep. 2019, 13, 459–472. [Google Scholar] [CrossRef]
  140. 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]
  141. Anzalone, A.V.; Koblan, L.W.; Liu, D.R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 2020, 38, 824–844. [Google Scholar] [CrossRef]
  142. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, H.; Zhang, J.; Lang, Z.; Botella, J.R.; Zhu, J.-K. Genome editing—Principles and applications for functional genomics research and crop improvement. Crit. Rev. Plant Sci. 2017, 36, 291–309. [Google Scholar] [CrossRef]
  144. Zhou, J.; Xin, X.; He, Y.; Chen, H.; Li, Q.; Tang, X.; Zhong, Z.; Deng, K.; Zheng, X.; Akher, S.A. Multiplex QTL editing of grain-related genes improves yield in elite rice varieties. Plant Cell Rep. 2019, 38, 475–485. [Google Scholar] [CrossRef]
  145. Zhang, Y.; Cheng, Y.; Fang, H.; Roberts, N.; Zhang, L.; Vakulskas, C.A.; Niedz, R.P.; Culver, J.N.; Qi, Y. Highly efficient genome editing in plant protoplasts by ribonucleoprotein delivery of CRISPR-Cas12a nucleases. Front. Genome Ed. 2022, 4, 780238. [Google Scholar] [CrossRef] [PubMed]
  146. Banakar, R.; Schubert, M.; Kurgan, G.; Rai, K.M.; Beaudoin, S.F.; Collingwood, M.A.; Vakulskas, C.A.; Wang, K.; Zhang, F. Efficiency, specificity and temperature sensitivity of Cas9 and Cas12a RNPs for DNA-free genome editing in plants. Front. Genome Ed. 2022, 3, 760820. [Google Scholar] [CrossRef] [PubMed]
  147. Banakar, R.; Schubert, M.; Collingwood, M.; Vakulskas, C.; Eggenberger, A.L.; Wang, K. Comparison of CRISPR-Cas9/Cas12a ribonucleoprotein complexes for genome editing efficiency in the rice phytoene desaturase (OsPDS) gene. Rice 2020, 13, 4. [Google Scholar] [CrossRef]
  148. 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. 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]
  149. Zhong, Z.; Zhang, Y.; You, Q.; Tang, X.; Ren, Q.; Liu, S.; Yang, L.; Wang, Y.; Liu, X.; Liu, B. Plant genome editing using FnCpf1 and LbCpf1 nucleases at redefined and altered PAM sites. Mol. Plant 2018, 11, 999–1002. [Google Scholar] [CrossRef]
  150. Tang, X.; Lowder, L.G.; Zhang, T.; Malzahn, A.A.; Zheng, X.; Voytas, D.F.; Zhong, Z.; Chen, Y.; Ren, Q.; Li, Q. A CRISPR–Cpf1 system for efficient genome editing and transcriptional repression in plants. Nat. Plants 2017, 3, 17018. [Google Scholar] [CrossRef]
  151. Malzahn, A.A.; Tang, X.; Lee, K.; Ren, Q.; Sretenovic, S.; Zhang, Y.; Chen, H.; Kang, M.; Bao, Y.; Zheng, X. Application of CRISPR-Cas12a temperature sensitivity for improved genome editing in rice, maize, and Arabidopsis. BMC Biol. 2019, 17, 9. [Google Scholar] [CrossRef]
  152. Xu, R.; Qin, R.; Li, H.; Li, J.; Yang, J.; Wei, P. Enhanced genome editing in rice using single transcript unit CRISPR-LbCpf1 systems. Plant Biotechnol. J. 2019, 17, 553. [Google Scholar] [CrossRef] [PubMed]
  153. Tang, X.; Sretenovic, S.; Ren, Q.; Jia, X.; Li, M.; Fan, T.; Yin, D.; Xiang, S.; Guo, Y.; Liu, L. Plant prime editors enable precise gene editing in rice cells. Mol. Plant 2020, 13, 667–670. [Google Scholar] [CrossRef] [PubMed]
  154. 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]
  155. Shimatani, Z.; Kashojiya, S.; Takayama, M.; Terada, R.; Arazoe, T.; Ishii, H.; Teramura, H.; Yamamoto, T.; Komatsu, H.; Miura, K. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 441–443. [Google Scholar] [CrossRef]
  156. Hao, L.; Ruiying, Q.; Xiaoshuang, L.; Shengxiang, L.; Rongfang, X.; Jianbo, Y.; Pengcheng, W. CRISPR/Cas9-mediated adenine base editing in rice genome. Rice Sci. 2019, 26, 125–128. [Google Scholar] [CrossRef]
  157. Li, X.; Wang, Y.; Liu, Y.; Yang, B.; Wang, X.; Wei, J.; Lu, Z.; Zhang, Y.; Wu, J.; Huang, X. Base editing with a Cpf1–cytidine deaminase fusion. Nat. Biotechnol. 2018, 36, 324–327. [Google Scholar] [CrossRef]
  158. Butt, H.; Rao, G.S.; Sedeek, K.; Aman, R.; Kamel, R.; Mahfouz, M. Engineering herbicide resistance via prime editing in rice. Plant Biotechnol. J. 2020, 18, 2370. [Google Scholar] [CrossRef] [PubMed]
  159. Lin, Q.; Zong, Y.; Xue, C.; Wang, S.; Jin, S.; Zhu, Z.; Wang, Y.; Anzalone, A.V.; Raguram, A.; Doman, J.L. Prime genome editing in rice and wheat. Nat. Biotechnol. 2020, 38, 582–585. [Google Scholar] [CrossRef]
  160. Gupta, A.; Liu, B.; Raza, S.; Chen, Q.-J.; Yang, B. Modularly assembled multiplex prime editors for simultaneous editing of agronomically important genes in rice. Plant Commun. 2024, 5, 100741. [Google Scholar] [CrossRef]
  161. Li, C.; Liu, B.; Dong, H.; Yang, B. Enhancing resistance to bacterial blight in rice using CRISPR-based base editing technology. Crop J. 2024, in press. [CrossRef]
  162. Li, Y.; Li, S.; Li, C.; Zhang, C.; Yan, L.; Li, J.; He, Y.; Guo, Y.; Xia, L. Fusion of a rice endogenous N-methylpurine DNA glycosylase to a plant adenine base transition editor ABE8e enables A-to-K base editing in rice plants. aBIOTECH 2024, 5, 127–139. [Google Scholar] [CrossRef] [PubMed]
  163. Lv, P.; Su, F.; Chen, F.; Yan, C.; Xia, D.; Sun, H.; Li, S.; Duan, Z.; Ma, C.; Zhang, H. Genome editing in rice using CRISPR/Cas12i3. Plant Biotechnol. J. 2024, 22, 379–385. [Google Scholar] [CrossRef]
  164. Zhong, Z.; Fan, T.; He, Y.; Liu, S.; Zheng, X.; Xu, Y.; Ren, J.; Yuan, H.; Xu, Z.; Zhang, Y. An improved plant prime editor for efficient generation of multiple-nucleotide variations and structural variations in rice. Plant Commun. 2024, 5, 100976. [Google Scholar] [CrossRef]
  165. Zhao, H.; Wolt, J.D. Risk associated with off-target plant genome editing and methods for its limitation. Emerg. Top. Life Sci. 2017, 1, 231–240. [Google Scholar] [PubMed]
  166. Brazelton, V.A., Jr.; 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 Crops Food 2015, 6, 266–276. [Google Scholar] [CrossRef]
  167. 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] [PubMed]
  168. Kadam, U.S.; Shelake, R.M.; Chavhan, R.L.; Suprasanna, P. Concerns regarding ‘off-target’activity of genome editing endonucleases. Plant Physiol. Biochem. 2018, 131, 22–30. [Google Scholar] [CrossRef] [PubMed]
  169. Xu, R.; Qin, R.; Li, H.; Li, D.; Li, L.; Wei, P.; Yang, J. Generation of targeted mutant rice using a CRISPR-Cpf1 system. Plant Biotechnol. J. 2017, 15, 713–717. [Google Scholar] [CrossRef]
  170. Osakabe, Y.; Watanabe, T.; Sugano, S.S.; Ueta, R.; Ishihara, R.; Shinozaki, K.; Osakabe, K. Optimization of CRISPR/Cas9 genome editing to modify abiotic stress responses in plants. Sci. Rep. 2016, 6, 26685. [Google Scholar] [CrossRef]
  171. Anders, C.; Niewoehner, O.; Duerst, A.; Jinek, M. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature 2014, 513, 569–573. [Google Scholar] [CrossRef]
  172. Kaya, H.; Mikami, M.; Endo, A.; Endo, M.; Toki, S. Highly specific targeted mutagenesis in plants using Staphylococcus aureus Cas9. Sci. Rep. 2016, 6, 26871. [Google Scholar] [CrossRef] [PubMed]
  173. 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.J. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 2015, 523, 481–485. [Google Scholar] [CrossRef] [PubMed]
  174. Hu, X.; Wang, C.; Fu, Y.; Liu, Q.; Jiao, X.; Wang, K. Expanding the range of CRISPR/Cas9 genome editing in rice. Mol. Plant 2016, 9, 943–945. [Google Scholar] [CrossRef]
  175. Hu, X.; Meng, X.; Liu, Q.; Li, J.; Wang, K. Increasing the efficiency of CRISPR-Cas9-VQR precise genome editing in rice. Plant Biotechnol. J. 2018, 16, 292–297. [Google Scholar] [CrossRef]
  176. Meng, X.; Hu, X.; Liu, Q.; Song, X.; Gao, C.; Li, J.; Wang, K. Robust genome editing of CRISPR-Cas9 at NAG PAMs in rice. Sci. China. Life Sci. 2018, 61, 122–125. [Google Scholar] [CrossRef]
  177. Zhang, C.; Wang, Y.; Wang, F.; Zhao, S.; Song, J.; Feng, F.; Zhao, J.; Yang, J. Expanding base editing scope to near-PAMless with engineered CRISPR/Cas9 variants in plants. Mol. Plant 2021, 14, 191–194. [Google Scholar] [CrossRef] [PubMed]
  178. El-Mounadi, K.; Morales-Floriano, M.L.; Garcia-Ruiz, H. Principles, applications, and biosafety of plant genome editing using CRISPR-Cas9. Front. Plant Sci. 2020, 11, 56. [Google Scholar] [CrossRef]
  179. Pan, C.; Sretenovic, S.; Qi, Y. CRISPR/dCas-mediated transcriptional and epigenetic regulation in plants. Curr. Opin. Plant Biol. 2021, 60, 101980. [Google Scholar] [CrossRef]
  180. Xu, L.; Sun, B.; Liu, S.; Gao, X.; Zhou, H.; Li, F.; Li, Y. The evaluation of active transcriptional repressor domain for CRISPRi in plants. Gene 2023, 851, 146967. [Google Scholar] [CrossRef]
  181. Tripathi, L.; Ntui, V.O.; Tripathi, J.N.; Norman, D.; Crawford, J. A new and novel high-fidelity genome editing tool for banana using Cas-CLOVER. Plant Biotechnol. J. 2023, 21, 1731. [Google Scholar] [CrossRef]
  182. Tripathi, L.; Ntui, V.O.; Tripathi, J.N. Application of CRISPR/Cas-based gene-editing for developing better banana. Front. Bioeng. Biotechnol. 2024, 12, 1395772. [Google Scholar] [CrossRef] [PubMed]
  183. Wu, W.Y.; Lebbink, J.H.; Kanaar, R.; Geijsen, N.; Van Der Oost, J. Genome editing by natural and engineered CRISPR-associated nucleases. Nat. Chem. Biol. 2018, 14, 642–651. [Google Scholar] [CrossRef] [PubMed]
  184. Demorest, Z.L.; Coffman, A.; Baltes, N.J.; Stoddard, T.J.; Clasen, B.M.; Luo, S.; Retterath, A.; Yabandith, A.; Gamo, M.E.; Bissen, J. Direct stacking of sequence-specific nuclease-induced mutations to produce high oleic and low linolenic soybean oil. BMC Plant Biol. 2016, 16, 225. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Key agronomic traits contributing to rice grain yield.
Figure 1. Key agronomic traits contributing to rice grain yield.
Plants 13 02972 g001
Figure 2. Applications of CRISPR/Cas9 in rice yield genes.
Figure 2. Applications of CRISPR/Cas9 in rice yield genes.
Plants 13 02972 g002
Figure 3. Different gene editing tools in rice. ZFNs—zinc finger nucleases, TALENs—transcription activator-like effector nucleases, CRISPR/Cas—clustered regularly interspaced palindromic repeat-associated Cas9 system, CRISPRa—CRISPR activation, CRISPRi—CRISPR interference, ABE—adenine base editing, CBE—cytosine base editing.
Figure 3. Different gene editing tools in rice. ZFNs—zinc finger nucleases, TALENs—transcription activator-like effector nucleases, CRISPR/Cas—clustered regularly interspaced palindromic repeat-associated Cas9 system, CRISPRa—CRISPR activation, CRISPRi—CRISPR interference, ABE—adenine base editing, CBE—cytosine base editing.
Plants 13 02972 g003
Table 1. CRISPR/Cas9 system for analyzing genes controlling rice grain number.
Table 1. CRISPR/Cas9 system for analyzing genes controlling rice grain number.
GeneCoding Product/ProteinModificationPathwayPhenotypeReference
Gn1aCytokinin oxidase/dehydrogenaseGene disruptionCytokinin biosynthesisIncreased panicle size and flower number per panicle[17,18]
GSN1 OsMPK1
(Mitogen-activated protein kinase (MAPK) phosphatase enzyme)
Gene disruptionMAPK signaling pathway Denser panicles and smaller grains [21]
FON4Receptor-like kinaseGene KnockoutCLV pathwayIncreased spikelet number per panicle[23,29]
FZPAP2/ERF transcription factorGene disruptionDifferent phytohormone-mediated signaling pathwaysIncreased grain numbers[26]
DEP1G-protein γ subunitGene disruptionG-protein signaling pathwayDense erect panicle and increased grain number and density[18,28]
LARGE2HECT-domain E3 ubiquitin ligase OsUPL2Gene disruptionFunctions with APO1 and APO2 (positively regulates grain number and panicle number)Large panicles with increased grain number[30]
Table 2. CRISPR/Cas9 system for analyzing genes controlling rice grain weight.
Table 2. CRISPR/Cas9 system for analyzing genes controlling rice grain weight.
GeneCoding Product/ProteinModificationPathwayPhenotypeReference
GS3G protein γ subunitGene disruption G-protein signalingIncreased grain size and quality[18,37,38,43]
GW2
(GRAIN WIDTH and WEIGHT2)
RING-type E3 ubiquitin ligaseGene knockoutUbiquitin-proteasome pathwayImproved grain filling and larger grain architecture[44]
GW5Calmodulin binding proteinGene knockoutBR signalingIncreased grain width and weight[40]
GW5LCalmodulin binding proteinGene knockoutBR signalingIncreased grain width and weight[41]
qTGW3OsSK41/OsGSK5Gene disruptionAuxin signalingLarger grain size[42]
OsMKK3Mitogen-Activated Protein Kinase Kinase 3Gene knockout *MAPK signalingDecreases grain length[45]
FZPERF domain Gene knockout *Ethylene biosynthesisSmaller grains and degenerated sterile lemmas[46]
MIR396e and MIR396fTranscription factorGene disruptionGA biosynthesisIncreased grain size and altered plant architecture[47]
OsWRKY53Transcription factorGene disruption *BA signaling and MAPK cascadesSmaller grains[48]
OsNDB2Type II NADPH dehydrogenaseGene knockoutAlternative respiratory pathway in mitochondria Increased grain size and 1000-grain weight [49]
OsSPL4SQUAMOSA PROMOTER BINDING PROTEIN-LIKEsGene disruptionTranscription factorIncreased grain number and size[3,50,51]
GL6Plant AT-rich sequence- and zinc-binding (PLATZ) transcription factorGene disruptionRNA polymerase III transcription machineryShort grains with increase in number[52]
GL10MADS56Gene knockout *Gibberellic acid (GA) signaling pathwayShorter grain length, lower grain weight and delayed flowering[53]
POW1
(PUT ON WEIGHT 1)
Homeodomain-like proteinGene disruptionBR pathwayIncreased grain size and leaf angle[54]
OsINV3 and OsINV2InvertaseGene knockout *Sucrose metabolismSmaller grain size[55]
GW10Cytochrome P450 subfamily 89A2 homology proteinGene knockoutBR pathwayIncreased grain number with smaller grains[56]
OsAGO17Argonaute (AGO) protein—component of the RNA-induced silencing complex (RISC)Gene knockout *sRNA pathwayDecreased grain size and weight[57]
FLR1, FLR2, FLR8
FLR15
FERONIA-like receptor protein kinases (FER)Gene disruption
Gene disruption *
FER pathwayLarger grain size
Smaller grain size
[58]
GW6 (GRAIN WIDTH 6)GA-regulated GAST family proteinGene knockout *Gibberellin pathwayReduced grain size and weight[59]
GSW3 (GRAIN SIZE AND WEIGHT 3)GTPase-regulated proteinGene knockoutGibberellin pathwayIncreased grain length, width and 1,000-grain weight[60]
qTGW12aMultidrug and toxic compound extrusion (MATE) transporterGene knockout *Various regulatory pathwaysReduction in grain weight[61]
GW9Nucleus-localized protein containing both C2H2 zinc finger (C2H2-ZnF) and VRN2-EMF2-FIS2-SUZ12 (VEFS) domainsGene knockout/disruptionGW2 ubiquitination pathwayLarge grains with increased plant height[62]
OsPUB3U-box E3 ubiquitin ligaseGene knockout *Ubiquitin-proteasome pathwayDecreased grain weight and size[63]
Note: * represents positive regulation.
Table 3. CRISPR/Cas9 system for analyzing genes controlling rice tiller number.
Table 3. CRISPR/Cas9 system for analyzing genes controlling rice tiller number.
GeneCoding Product/ProteinModificationPathwayPhenotypeReferences
MOC1GRAS family
transcription factor
Gene knockout *Transcription factorReduced tillering[66]
MOC3/
TAB1/SRT1
Homeobox domain
-containing protein
Gene knockout *Transcription factorReduced tillering[66]
OsFWL4Cysteine-rich proteinGene disruption Transcription factorIncreased tiller number and grain length[68]
OsAAP3Amino acid permeasesGene knockoutAmino acid transport
(regulate the concentrations of amino acids)
Increased tiller number[69,70]
OsNPF7.1 and OsNPF7.4Nitrate and di/tripeptide transporterGene knockout
(differential expression in the presence of nitrogen)
Nitrogen transportPlant architecture, NUE and tiller number[71]
OsPIN5bEndoplasmic reticulum localized proteinGene knockoutAuxin balance and transportLonger balance and transport tiller numbers[37]
IDEAL PLANT ARCHITECTURE1 (IPA1)/
OsSPL14
Squamosa promoter binding proteinGene disruptionStrigolactone signaling pathwayTiller number varied according to the changes induced in the OsmiR156 target region[18,72]
CCD7 (CAROTENOID CLEAVAGE DIOXYGENASE 7)Carotenoid cleavage dioxygenaseGene disruption, gene knockoutStrigolactone biosynthesisIncreased tillering and reduced height[73,74]
TB1 (TEOSINTE BRANCHED1)TEOSINTE BRANCHED1/CYCLOIDEA/PROLIFERATING CELL FACTORS (TCP) family transcription factorGene knockoutStrigolactone signaling pathwayDwarf phenotype with increased tiller numbers[75]
EHD1B-type response regulatorGene disruptionFlowering pathwayEnhanced yield and improved grain quality[76]
Note: * represents positive regulation.
Table 4. CRISPR/Cas9 system for analyzing genes controlling stress and herbicide resistance in rice.
Table 4. CRISPR/Cas9 system for analyzing genes controlling stress and herbicide resistance in rice.
TraitGeneGene Function/Coding ProductImproved PhenotypeReference
Biotic stress tolerance/resistanceOsSWEET13Sucrose transporter gene Bacterial blight disease resistance[84]
OsERF922Ethylene responsive factors Increased resistance to blast disease[85]
OsSWEET11,
OsSWEET13,
OsSWEET14
Sucrose transporter genesBroad-spectrum resistance to bacterial blight[86]
ALB1, RSY1Melanin biosynthetic polyketide synthaseRice blast resistance[87]
Xa13Recessive resistant allele of Os8N3, a member of the NODULIN3 (N3) gene familyBacterial blight disease resistance[88]
eIF4GTranslation initiation factor 4 gamma geneResistance to Rice Tungro Spherical Virus (RTSV)[89]
OsCPR5.1NucleoporinResistance to Rice Yellow Mottle Virus (RYMV)[90]
Abiotic stress tolerance/resistanceOsERA1β-subunit of farnesyltransferaseDrought tolerance[91]
OsDST (Drought and salt tolerance)DST proteinDrought and salt tolerance[92]
OsNAC006NAC transcription factorHeat tolerance[93]
OsPRP1Proline-rich proteinCold tolerance[94]
Herbicide resistanceOsALSAcetolactate synthaseSignificant tolerance to herbicides[95]
OsEPSPS5-enolpyruvylshikimate-3-phosphate synthaseResistance to glyphosate[96]
Table 5. CRISPR/Cas9 system for analyzing genes controlling NUE.
Table 5. CRISPR/Cas9 system for analyzing genes controlling NUE.
GeneCoding Product/ProteinModificationFunctionPhenotypeReference
AtAAP2
homolog of rice
AtCRF4
homolog of rice
Amino acid permease
Transcription factor
Gene knockoutN transportation
N uptake in roots
Shorter plants, increased panicle numbers, and dry biomass weight[106]
LW5/D1α subunit of G-proteinGene disruptionN uptake and transportAffect plant architecture and grain size by regulating N-transfer[107]
OsHHO3NIGT1 family proteinGene knockoutNitrate signalingEnhanced growth and increased shoot and root dry mass[105]
Table 6. CRISPR/Cas9 system for analyzing genes controlling photosynthetic efficiency.
Table 6. CRISPR/Cas9 system for analyzing genes controlling photosynthetic efficiency.
GeneCoding Product/
Protein
ModificationPathway/FunctionPhenotypeReferences
OsHXK1HexokinaseGene knockoutDifferent phytohormone-mediated signaling pathwaysHigh photosynthetic efficiency and yield[112]
OsPPR9DYW-PPR
(Pentatricopeptide repeat)
Gene disruption *RNA editingAffects chloroplast growth and development[111]
OsMORF9MORFs
(multiple organellar RNA editing factors)
Gene knockout *RNA editingBiogenesis of chloroplast ribosomes and chloroplast development[113]
OsCAO1Chlorophyllide a oxygenaseGene knockout *Chlorophyll degradation and ROS scavengingDegradation and ROS senescence[114]
CpSRP43, CpSRP54a, CpSRP54bCpSRP
(signal recognition particle)
Gene knockoutCpSRP pathwayIncreased photosynthesis per photon absorbed[115]
OsEPF1Epidermal patterning factorGene knockoutStomatal development and patterningEnhanced the stomatal conductance and photosynthetic efficiency[116]
Note: * represents positive regulation.
Table 7. Modifying regulatory gene regions to enhance rice yield.
Table 7. Modifying regulatory gene regions to enhance rice yield.
GeneModificationImproved PhenotypeReference
OsCPK18/OsCPK4Modification of the phosphorylation motif of OsCPK18/OsCPK4Improved disease resistance and yield[120]
OsPDCD5Improved plant architecture (plant height, panicle type, grain shape)Enhanced yield[121]
OsSPL16Upregulation of pyruvate enzymes and cell cycle enzymesLarger grain size and increased yield[122]
(Ehd1) Early heading date 1Modification of promoter at multiple sitesDelayed heading date and improved yield-related traits[123]
OsPYL9
(Pyrabactin resistance 1-like 9)
Upregulation of circadian rhythm and abiotic stress-responsive proteinsIncreased yield and drought tolerance[124]
FZPDeletion of −157 to −45 bp in UTR (untranslated region) upstream of FZP 5′ Grain number[26]
IPA1/OsSPL14Deletion of An-1 (transcription factor) binding site of IPA1
(54-base pair cis-regulatory region)
Balances the trade-off between grains per panicle and tiller numbers, leading to increased grain yield[125]
Table 8. Comparison of various Cas proteins.
Table 8. Comparison of various Cas proteins.
Cas ProteinHostTargetPAM SiteCut TypeUnique FeaturesReference
Cas9Streptococcus pyogenesDouble-strand DNA (dsDNA)NGGDSBVersatile, suitable for multiplexing[133]
Cas12a (Cpf1)Prevotella and Francisella 1dsDNATTTN (AT-rich region)Staggered cutStaggers DSBs and promotes HDR mechanism[134]
Cas12j2 (CasΦ)Huge phagesdsDNANTTVStaggered cutSmall and compact[135]
Cas13aLeptotrichia shahiiSingle-strand RNANoneRNA cleavageRNA targeting[136]
Cas14Uncultivated ArchaeaSingle-strand DNA (ssDNA)NonessDNA cleavageSmall and specific for ssDNA[137]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Thiruppathi, A.; Salunkhe, S.R.; Ramasamy, S.P.; Palaniswamy, R.; Rajagopalan, V.R.; Rathnasamy, S.A.; Alagarswamy, S.; Swaminathan, M.; Manickam, S.; Muthurajan, R. Unleashing the Potential of CRISPR/Cas9 Genome Editing for Yield-Related Traits in Rice. Plants 2024, 13, 2972. https://doi.org/10.3390/plants13212972

AMA Style

Thiruppathi A, Salunkhe SR, Ramasamy SP, Palaniswamy R, Rajagopalan VR, Rathnasamy SA, Alagarswamy S, Swaminathan M, Manickam S, Muthurajan R. Unleashing the Potential of CRISPR/Cas9 Genome Editing for Yield-Related Traits in Rice. Plants. 2024; 13(21):2972. https://doi.org/10.3390/plants13212972

Chicago/Turabian Style

Thiruppathi, Archana, Shubham Rajaram Salunkhe, Shobica Priya Ramasamy, Rakshana Palaniswamy, Veera Ranjani Rajagopalan, Sakthi Ambothi Rathnasamy, Senthil Alagarswamy, Manonmani Swaminathan, Sudha Manickam, and Raveendran Muthurajan. 2024. "Unleashing the Potential of CRISPR/Cas9 Genome Editing for Yield-Related Traits in Rice" Plants 13, no. 21: 2972. https://doi.org/10.3390/plants13212972

APA Style

Thiruppathi, A., Salunkhe, S. R., Ramasamy, S. P., Palaniswamy, R., Rajagopalan, V. R., Rathnasamy, S. A., Alagarswamy, S., Swaminathan, M., Manickam, S., & Muthurajan, R. (2024). Unleashing the Potential of CRISPR/Cas9 Genome Editing for Yield-Related Traits in Rice. Plants, 13(21), 2972. https://doi.org/10.3390/plants13212972

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