Next Article in Journal
Molecular Genotyping by 20K Gene Arrays (Genobait) to Unravel the Genetic Structure and Genetic Diversity of the Puccinia striiformis f. sp. tritici Population in the Eastern Xizang Autonomous Region
Previous Article in Journal
Phenotypic Characters and Inheritance Tendency of Agronomic Traits in F1 Progeny of Pear
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Large-Scale Rice Mutant Establishment and High-Throughput Mutant Manipulation Help Advance Rice Functional Genomics

1
State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou 311400, China
2
Department of Biology, College of Natural and Computational Sciences, Debre Tabor University, Debre Tabor P.O. Box 272, Ethiopia
3
School of Life Sciences, Hubei University, Wuhan 430062, China
4
College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
5
National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Plants 2025, 14(10), 1492; https://doi.org/10.3390/plants14101492
Submission received: 27 March 2025 / Revised: 8 May 2025 / Accepted: 13 May 2025 / Published: 16 May 2025
(This article belongs to the Special Issue Crop Improvement by Modern Breeding Strategies)

Abstract

:
Rice (Oryza sativa L.) is a stable food for over half of the world population, contributing 50–80% of the daily calorie intake. The completion of rice genome sequencing marks a significant milestone in understanding functional genomics, yet the systematic identification of gene functions remains a bottleneck for rice improvement. Large-scale mutant libraries in which the functions of genes are lost or gained (e.g., through chemical/physical treatments, T-DNA, transposons, RNAi, CRISPR/Cas9) have proven to be powerful tools for the systematic linking of genotypes to phenotypes. So far, using different mutagenesis approaches, a million mutant lines have been established and about 5–10% of the predicted rice gene functions have been identified due to the high demands of labor and low-throughput utilization. DNA-barcoding-based large-scale mutagenesis offers unprecedented precision and scalability in functional genomics. This review summarizes large-scale loss-of-function and gain-of-function mutant library development approaches and emphasizes the integration of DNA barcoding for pooled analysis. Unique DNA barcodes can be tagged to transposons/retrotransposons, DNA constructs, miRNA/siRNA, gRNA, and cDNA, allowing for pooling analysis and the assignment of functions to genes that cause phenotype alterations. In addition, the integration of high-throughput phenotyping and OMICS technologies can accelerate the identification of gene functions.

1. Introduction

Rice (Oryza sativa L.) is a stable food for over half of the world population, contributing 50–80% of the daily calorie intake [1]. As the most widely used model organism among major crops, rice is essential for genetic and physiological research [2] due to its small genome size (420 Mb) and synteny with other plants [3,4]. Traditional breeding struggles to address yields, stress tolerance, and nutritional traits, necessitating advanced mutagenesis. The annual rice production growth has slowed to the point that it is no longer able to keep up with the growing global population of consumers [5].
In the post-genome-sequencing era, the systematic identification of gene functions and understanding of the molecular mechanisms through which genes regulate important traits are the basis for rice genetic improvements [6]. The construction and phenotypic screening of random loss-of-function or gain-of-function mutant libraries have been regarded as one of the most promising ways to identify functional genes and genetically improve rice varieties [7,8].
According to the Food and Agriculture Organization of the United Nations/International Atomic Energy Agency [9], approximately 1351 mutant rice varieties, related to major agronomic and stress tolerance, have been developed using various mutagenesis techniques and have been released for cultivation, which has helped to address the growing challenges of rice production, including the need for improved agronomic traits, nutritional quality, and resilience to environmental stresses. Despite these advancements, the current global release of rice varieties is insufficient to meet the increasing demands of the growing population [5]. Thus, mutagenesis remains one of the most effective tools in functional genomics, facilitating the systematic identification of gene functions through two primary approaches: loss-of-function and gain-of-function mutagenesis [7,10,11,12].
This article examines the available mutant resources and highlights the potential of both loss-of-function and gain-of-function mutagenesis strategies in developing mutant libraries to advance rice functional genomics. Such mutagenesis approaches are essential for elucidating gene functions and enhancing critical traits in rice, a staple crop of considerable agricultural significance. The article also addresses the challenges associated with various mutagenesis techniques and underscores the importance of DNA barcoding for large-scale mutagenesis studies and OMICS technologies to accelerate rice functional genomics.

2. Approaches for Establishment of Loss-of-Function Mutants

2.1. Chemical and Physical Mutagenesis

To create genetic variability and identify the gene functions that cause changes in traits, random mutagenesis using chemicals, such as EMS, N-methyl-N-nitrosourea, sodium azide, and ENU, and physical agents, such as γ-rays, fast-neutron irradiation, ion beam radiation, and X-rays, are commonly employed to develop large-scale random mutations [13]. In chemical and physical mutagenesis, different plant parts, such as the embryo, seed, callus, and suspension culture, exhibit varying efficiencies, with seeds being the most used [10,12,13]. The dosage and duration of exposure to mutagens also significantly influence the efficiency of both chemical and physical mutagenesis [14,15,16,17].
Targeting induced local lesions in the genome (TILLING) reveals that 95% of chemical mutations are silent/missense, while physical mutagenesis often causes 15 bp deletions or chromosomal rearrangements [18]. For example, in fast neutron (FN)-mutagenized rice (Kitaake), there are 91,513 mutations affecting 32,307 genes, with 48% of these mutations having a single base change due to substitutions and deletions, while 66% of the mutations have other kinds of chromosomal rearrangements [19,20]. Moreover, chemical mutagens like N-methyl-N-nitrosourea (MNU) and ethyl methane sulfonate (EMS) are also essential tools for producing single-nucleotide polymorphisms with random distributions. In the indica rice variety IR64, for example, approximately 60,000 mutants induced via chemical and irradiation methods have facilitated significant advances in gene mining and functional characterization [21].
The genome-wide chips and high-throughput genotyping technologies indicate that there are larger chromosomal aberrations or deletions of essential genes in the mutant lines, limiting their effectiveness in rice mutagenomics [22]. While these methods facilitate large-scale mutagenesis, the identification of specific mutation sites and studies of gene functions across the genome through genetic mapping approaches can be laborious and time-consuming [23]. In contrast, T-DNA insertional mutagenesis offers a valuable alternative for generating loss-of-function mutants, as it tags mutations with identifiable inserted fragments, simplifying the process of mutation identification and functional analysis [24].

2.2. T-DNA Insertional Mutagenesis

In rice, the most widely used mutagenesis approach is insertional mutagenesis, where a segment of DNA is randomly inserted into the genome, leading to gene disruption and loss of function [25] (Figure 1). This DNA can be a transposable element (like Ac/Ds, Spm, and Tos17), which is a mobile genetic element capable of moving from one location to another within the genome when it has access to the necessary transposase enzyme [26,27], or it may consists of foreign DNA introduced into the cell [28]. This mechanism can lead to diverse genomic alterations, including deletions, insertions, and chromosomal translocations, thereby generating a wide range of mutant phenotypes [29]. Given the known sequence of the DNA insertional elements, the insertion sites in the mutants can be easily identified by using techniques like inverse PCR [30], thermal asymmetric interlaced PCR (TAIL PCR) [31], adapter ligation PCR [32], and DNA sequencing [30,33], which greatly facilitates the association of the phenotype with mutated genes. In 2017, researchers developed a barcode method for the pooled identification of transposon tagging sites using Illumina sequencing, and this can trace back the insertion sites to the preliminary individual clones of the transposon mutant library [34].
T-DNA insertional mutant libraries have been extensively constructed. These include 22,090 T-DNA insertion lines [28], 50,000 activation tagging lines of Nipponbare [35], 47,932 Kitaake activation tag lines [36], 171,000 T-DNA enhancer trap lines of different japonica backgrounds (Zhonghua 11, Zhonghua 15, Nipponbare) [37,38,39], and 22,665 Tainung 67 trifunctional T-DNA insertional lines [40]. The emphasis on japonica is due to its high transformation efficiency, enabling the large-scale generation of mutant resources [23,41]. Transposons have several advantages over T-DNA. In the Ac/Ds system, the Ds element preferentially transposes to genetically linked sites on the same chromosome, facilitating the establishment of an indexed insertion mutant library [42]. Tos17 is the most active endogenous retrotransposon for functional gene analyses [5]. In addition to T-DNA mutant lines, various transposon mutant resources have been established, including 50,000 mutant lines created through insertional mutagenesis [5]. A variety of genes have been identified through different mutagenesis approaches. For example, Tos17 helped to discover the photomorphogenesis regulator phyA [43]. Transposon-based mutagenesis, such as that with the Ac/Ds system, revealed key genes, including AID1 for another development trait [44], OsCYP96B4 for cell elongation and lipid metabolism [45], OsKS1 for germination and root growth [46], and OsMYOXIB for pollen development [47]. T-DNA insertion mutagenesis also uncovered important regulatory genes, such as RID1 as a master regulator of flowering induction [48], JMJ706 for floral development [49], and OsCHLH, which is involved in chlorophyll biosynthesis [50]. Other significant genes include ILA1 for mechanical tissue formation [51], DSM2 for the xanthophyll cycle and ABA synthesis [52], OsPMT16 for pistil development [53], MPK6 for early embryo development [54], DTC1 for programmed cell death in the tapetum [55], and OsPPDKB associated with floury endosperm formation [56]. Overall, these mutagenesis approaches have greatly enhanced our understanding of the genes associated with important traits in rice, offering valuable insights for rice improvement.
In addition to the disruption of gene functions, the integration of DNA insertional mutagens was also modified to identify regulatory elements of genes via gene trapping, enhancer trapping, and promoter trapping technologies [23,57]. In the enhancer trap method [58], a reporter gene is placed near an enhancer to reveal gene expression patterns and regulatory networks. Promoter traps [23] involve introducing a reporter gene without its own promoter, but alongside a transcription unit, enabling it to be activated by adjacent promoters. This approach is valuable for functional analyses. Additionally, in gene trapping [28], a reporter gene is integrated into the genome, and its expression indicates that a gene is disrupted.
Currently, only 5% to 10% of the corresponding genes have been identified from the millions of mutant lines, as the integration of transposons or T-DNA does not always correlate with a change in the phenotype [59]. In addition to the demands of huge labor and the high costs of insertional mutagenesis manipulation, achieving whole-genome saturation via insertional mutagenesis suffers greatly from the unequal distribution of insertions caused by T-DNA and transposons throughout the genome [25,60,61]. For example, T-DNA prefers to integrate into the 5′ upstream and 3′ downstream regulatory regions and noncoding genes. Hence, inducing the coding region to have a single insertion tag is very challenging [25,60,62]. Furthermore, insertional mutagenesis is restricted to the targeting of multiple genes within the same pathways in a single mutant line. This constraint hampers the identification of functionally redundant genes [63].

2.3. Large-Scale CRISPR/Cas9-Mediated Mutagenesis

Several rice mutant resources have been established globally; however, traditional loss-of-function mutagenesis approaches face challenges, such as random integration and the multiple individual mutants needed to achieve mutations [63,64,65]. The CRISPR/Cas9 genome editing system has emerged as a promising alternative, enabling targeted mutagenesis with low off-target effects [64]. CRISPR utilizes guide RNA (gRNA) to bind to the target DNA at the protospacer adjacent motif (PAM) and instructs Cas9 to perform specific cleavage [66,67] (Figure 2). The cell’s DNA repair mechanism then either uses nonhomologous end-joining (NHEJ) or homologous re-combination repair mechanisms [67], resulting in insertions and deletions (indels) that alter gene functions [68,69].
Initially, small-scale gene-knockout mutagenesis with CRISPR targets one or a few genes, which can be challenging due to the gene family [63]. However, CRISPR/Cas9 can target many DNA fragments, enabling the rapid construction of large-scale knockout mutant libraries for screening. High-throughput loss-of-function screening using this system is quickly being applied to plants [64,70,71] (Table 1). By using multiplexing and pooled gRNA libraries, multiple redundant genes can be simultaneously mutated across the genome [64,72].
Recently, a unique tag approach has been developed for the identification of gRNAs, facilitating pooled CRISPR library construction and simultaneous transformation. This new approach successfully generated 955 CRISPR/Cas9 mutant lines targeting the receptor like kinase (RLK)-family genes of rice, achieving a mutation frequency of 74.3% and enabling the identification of genes such as OsRLCK109, OsCERK1, OsMRLK13, OsRLCK253, OsWAK32, LOC_Os01g03370, LOC_Os01g04580, LOC_Os03g56160, LOC_Os07g35300, LOC_Os09g18360, and LOC_Os09g37834, involved in rice immunity [75]. Similarly, this approach has led to the development of 1319 OsNAC-gene-mutant rice lines (T0) with over 70% mutation frequency, aimed at uncovering the transcription factors (OsNAC30, OsNAC59, and OsNAC101) associated with rice immunity [76]. Large-scale CRISPR/Cas9 loss-of-function mutagenesis efforts have increasingly relied on pooled gRNA libraries to identify various gene functions. For instance, a 88,541 gRNA library generated 84,384 ZH1 (Oryza sativa L. ssp. Japonica) mutant lines (T0) with a mutation frequency of 83.9% [72]. Additionally, the library of 25,604 gRNAs produced 14,000 Zhonghua 11 (Oryza sativa L. ssp. japonica) mutant lines (T0) with 86.4% targeted mutations [81]. Furthermore, 2184 large-scale CRISPR/Cas9 mutant lines (T0) were developed by integrating 367 gRNAs targeting rice seed preferred genes, achieving a mutation frequency of 84.06% [82]. Despite these advancements, the number of mutant lines generated has remained low compared to other loss-of-function mutant libraries, and there is still a smaller number of mutant libraries. Therefore, the ongoing development of high-throughput, large-scale CRISPR/Cas9 mutant lines is essential for identifying new gene functions, ultimately contributing to rice improvement.

2.4. Large-Scale RNA-Interference-Mediated Mutagenesis

The RNA interference (RNAi) technique is an essential mutagenesis tool in functional genomics that allows for the analysis of target gene effects through suppression [83] (Figure 2). It begins with double-stranded RNA (dsRNA) or hairpin RNA (hpRNA), which are processed by Dicer or Dicer-like (DCL) proteins into siRNAs or miRNA. These then associate with an Argonaute (AGO) protein, forming active effector complexes, miRNP or RISC, which repress expression of the corresponding target mRNA [84,85].
To establish genome-wide long hpRNA mutant libraries targeting multiple transcripts simultaneously, an approach called a rolling cycle amplification-mediated hpRNA (RMHR) system has been developed to target multiple genes of the plant. Using the RMHR approach, 1500 hpRNA rice mutant lines (T0) were developed, and they exhibited more than 47.9% of distinct phenotypic changes compared to the wild-type [41]. This indicates that mutation occurrence at the exonic sequence is much higher compared with the chemical and T-DNA insertional mutagenesis approaches [22,23,25,86]. Thus, this approach of mutant library construction serves as an effective tool for large-scale genome-wide gene function analyses (Table 1). While loss-of-function mutagenesis approaches are effective, two key challenges remain: (1) functional redundancy (approximately 29% of rice genes belong to multiple families, where the knockout of a single gene may not result in a clear phenotype due to functional complementation or buffering against the mutations [87,88,89,90]), and (2) essential genes for which the complete knockout causes lethality due to defective gametes, embryos, and growth and development [35,91,92]. RNA interference (RNAi) enables the study of essential genes at specific developmental stages without lethal effects [81,93]. For redundant gene families, pooled CRISPR/Cas9 or RNAi libraries can simultaneously target multiple members, overcoming compensatory effects [41,64,82,93,94,95,96].

3. Approaches for Establishment of Gain-of-Function Mutants

3.1. Activation Tagging

Activation tagging is one of the efficient functional acquisition mutations for the development of new phenotypes and to understand the genes that cause the change. In this system, T-DNA or a transposable element containing enhancers activates the expression of nearby genes upon insertion into the genome (Figure 3). The enhancer typically consists of tetrameric repeats of the CaMV35S promoter, which can activate gene expression in either orientation or extend up to 10 kb from the insertion site in the genome [35,90,97].
Activation-tagged mutagenesis approaches have been utilized in various species, including Arabidopsis thaliana [98], rice [99], wheat [100], oat [101], tomato [102], and barley [103], for systematic gene function identification.
In rice, significant mutant lines have been developed from different cultivars via T-DNA or Ac-Ds transposon mutagenesis. For example, the Dongjin and Hwayong cultivars with T-DNA yielded 47,932 lines [36,104], while Tainung 67 produced 55,000 lines [105,106]. The Nipponbare cultivar alone generated 13,000 lines with T-DNA [107] and 638 lines with the Ac-Ds system [108], plus an additional 50,000 lines from other T-DNA treatments [35]. These resources have advanced rice functional genomics despite challenges in development. Several genes have been identified for rice improvements through activation tagging [97], such as GLR1, GLR2, XPB2, SEN1 for drought resistance [109,110], and SPL18/OsAT1 linked to enhanced blast resistance [107]. Activation tagging has also revealed potential sheath blight (ShB)-resistance genes encoding glycoside hydrolase and Class III chitinase in indica rice [111]. However, the enhancer in the activation tagging can activate multiple genes up to 12.5 kb from the insertion site, complicating the connection between specific genes and their phenotype due to the simultaneous transcription of multiple endogenous genes [78,90].

3.2. FOX (Full-Length cDNA Overexpression) Hunting

FOX hunting is another valuable gain-of-function mutagenesis approach. In these approaches, full-length (Fl) cDNA libraries are cloned between the strong promoter and a terminator, followed by transformation, to generate the FOX lines [79,80,112]. In studies utilizing Arabidopsis, mutation rates for the FOX approach range from approximately 10% to 17% [79,80]. In contrast, the activation methods yield lower mutation rates, at around 1% [78]. Thus, the FOX lines serve as a vital resource for the identification of new genes, the characterization of proteins, and conducting biochemical analyses [112,113,114,115].
In 2006, Ichikawa et al. [79] developed 15,000 FOX Arabidopsis lines driven by the dicot-specific promoter CaMV35S, and the lines exhibit stable phenotype mutations, unlike the mutant lines produced by the loss-of-function mutation techniques [79]. Similarly, the rice FOX-Arabidopsis population, developed with 13,000 rice FL-cDNAs and the CaMV 35S promoter, generated an impressive 23,715 FOX lines, with 11,000 integrated FL-cDNAs, resulting in an integration efficiency of 84.6% [112]. To further elucidate rice gene functions, 13,980 rice FL-cDNAs were integrated into rice using the maize Ubiquitin-1 promoter, which resulted in 11,582 integrated lines and achieved an integration efficiency of 39.1% [80]. Additionally, 13,823 rice FL-cDNAs were driven by the rice Actin-1 promoter, yielding 1920 FL-cDNA-integrated lines with a 13.8% integration efficiency [116]. There were methodological differences in the binary vector, promoter, and cloning methods in these two cases. Despite the widespread use of in vitro site-specific Gateway recombination technology for establishing full-length cDNA libraries across various organisms, the integration rates of full-length cDNAs are lower compared to restriction site cloning techniques using the pRiceFOX binary vector [80,117,118]. Since its invention, the FOX hunting system has primarily focused on model plants, like Arabidopsis and rice. However, this technique is now applicable to a wide range of interspecific and intraspecific species, regardless of the genome size [112], including woody plants [119,120] and cereal crops [80]. This broader application enhances our understanding of gene functions and improves the genome annotation quality [115,121].
For example, approximately 15,000 FOX rice lines derived from 1455 full-length cDNAs of the transcription factors of the cultivated and the wild species of wheat have been established. Of these, 14.88% exhibited visible phenotype changes, with five lines demonstrating enhanced tolerance to salt and osmotic stress [122]. In the same way, Fujita et al. [123] established an Arabidopsis FOX line using a mix of 43 salt-induced full-length cDNAs of transcription factors, leading to identification of the bZIP transcription factor-related gene AtZIP60, which plays a role in regulating the salt response. This implies that the FOX hunting approaches can effectively facilitate gene function identification within and across both wild and cultivated species. However, like other insertional mutagenesis techniques, this approach also requires high-throughput screening of the full-length cDNA, as well as high-throughput phenotyping and genotyping of mutant lines, to advance rice functional genomics (Table 1). The costs and the technical challenges associated with the existing full-length cDNA library synthesis for FOX hunting are also challenging.

4. Genetic Resources Required for a Gain of Function

4.1. Full-Length cDNA Library

So far, more than 60,000 full-length cDNAs, including about 38,000 full-length cDNAs from japonica cv. Nipponbare [124], 10,096 FL-cDNA sequences from indica variety (Guangluai 4) [125], and 10,828 from indica variety (Minghui 63) [126], and with only around 1888 putative full-length cDNA clones, have been completely sequenced from wild rice (Oryza rufipogon Griff. W1943) [127]. However, many existing FL-cDNAs lack detailed functional annotations and are insufficient for the functional characterization of all rice genes. Despite the efforts of full-length cDNA synthesis, it is still hindered by technical issues, such as incomplete transcripts, sequencing costs and errors, and difficulties in capturing alternative splicing events. Thus, these challenges require new approaches to cover and efficiently translate genomic data into functional insights.
In establishing large Fl-cDNA libraries in rice, different approaches yield varying success rates for full-length clones. For instance, oligo capping methods achieve over 90% full-length cDNA clones [124], while cap tagging yields approximately 70% [127]. In contrast, conventional oligo dT or adapter-oriented methods result in only 60% full-length cDNA clones [126]. The choice of the techniques is vital, as it directly impacts the quality and completeness of the resulting cDNA synthesis.
Alternatively, genomic DNA can be used directly as a template for single exon-containing genes, to enrich the amount of rice Fl-cDNA libraries, and offer significant promise for functional genomic studies and explorations of gene functions. This approach simplifies the process because the entire gene is contained within a single exon, eliminating the need for complex splicing or the handling of introns. The benefits of this method include reduced complexity and cost-effectiveness, as it minimizes the steps involved in isolating mRNA and converting it to cDNA. In the rice genome, more than 11,109 genes (19.9%) are monomorphic (single exon genes) and belong to a large gene family, like F-box proteins, ribosomal proteins, DEAD-box helicase, and so on [128]. The single-exon genes are much more beneficial for regulatory activities since they have better transcriptional fidelity due to the absence of splicing events [129,130].

4.2. Wild Relatives of Rice

It is noteworthy that there are currently very few, if any, libraries of rice lines available that are derived from wild species, which limits our ability to investigate novel gene functions from rice ancestors. Gain-of-function mutagenesis is a transformative approach to crop improvement that leverages the wild relatives’ genetic diversity and enhances specific traits in cultivated varieties [114,122,131]. Nowadays, climate variability and biotic and abiotic stress threaten rice production globally. Thus, the genetic variability of Oryza wild relatives will provide game-changing traits and genes to address the challenges of resistance and global agricultural production in current and future climates [132,133].
For an understanding of the function of wild rice, Lu et al. [127] has established 1,888 full-length cDNAs from wild rice (Oryza rufipogon Griff. W1943), and the homology sequence analysis showed that more than 96.8% of the wild rice full-length cDNA is highly homologous with the cultivated rice genome sequence and that 1% (17) of the full-length cDNA comprises unique rice genes that can be used for further study to advance rice functional genomics and breeding. Very recently, the functional screening of over 4000 wild apple (Malus sieversii) FOX Arabidopsis lines identified a total of 160 inserts appearing to be novel, with no or limited homology to the cultivated apple (Malus pumila) and Arabidopsis and mainly providing freezing resistance [134]. This implies that the wild genetic resource can also be used as a genetic donor for gain-of-function mutagenesis approaches for new gene discovery.

5. DNA Barcoding for Large-Scale Mutagenesis to Accelerate Functional Genomics

5.1. Fast and High-Throughput Genotyping

To utilize the mutants generated, genotyping the mutants is essential. Traditional technologies largely rely on PCR amplification and Sanger sequencing, which make the mutant genotyping time-consuming and tedious. Recently, DNA barcoding has emerged as a revolutionary method that enables the simultaneous sequencing of pooled samples from numerous genetic materials (Figure 4). These fixed-length-index nucleotide sequences allow for efficient multiplexing during sequencing [135], making it crucial for biodiversity studies across various taxa [136,137], detecting invasive species [138], understanding plant–pollinator interactions [139,140], and assessing water quality [137].
The future of functional genomics using barcoding techniques is promising, as unique sequences help to avoid redundancy and simplify identification [2,141]. Sample identifiers can be added via ligation or with a one-step PCR process [136,141,142], two-step PCR, offering flexibility with potential contamination risks [136,141,142], and tagged PCR approaches, which provides more design flexibility [136,143] while minimizing biases, although it risks tag-jumping errors [136].
Barcode libraries enhance functional genomics by allowing for the high-throughput screening of gene functions (Table 2). They consist of unique DNA sequences that enable the tracking of individual genes or genetic elements. Combining forward and reverse genetics with DNA barcoding is vital for understanding gene functions and interactions, especially in large-scale libraries for biomolecule identification [142]. Loss-of-function CRISPR/Cas9 mutant lines are significant for gene function discovery. High-throughput screening using sgRNA with unique identifiers has improved the efficiency in pooled mutant screening [144,145]. The barcode-based NGS approach successfully tracked mutations in maize, revealing genes with observable phenotype changes [146]. Additionally, a barcode-tagged insertion mutant library in yeast has enabled the high-throughput genotype analysis of 10,000 mutants [147].

5.2. High-Througput Phenotyping and OMICS Integration to Accelerate Functional Genomics

Many of the mutant lines are complemented by phenotypic information. The association of genetic and phenotypic data has propelled significant progress in all-inclusive analysis and enhancing our understanding of gene functions [24]. However, the phenotyping of mutants still depends on the manual scoring of visible traits. Thus, the ongoing large-scale developments of rice mutant resources have highlighted the necessity of high-throughput phenotyping to tackle the challenges of identifying the mutant phenotypes developed through loss- and gain-of-function mutations [40]. High-throughput phenotyping (phenomics) is being used for multidimensional phenotypes from cellular-to-organ-level assessments and can be extended to individual plants and entire fields for the identification of gene functions and new genes/QTLs and the understanding of the genetic architecture of a complex trait and environmental responses [154,155,156,157]. This approach can also increase the efficiency, reduce the labor, and analyze large populations to make associations between the genotype and phenotype [158].
Recently, the rapid advancement of the use of image-based approaches, implementation of robotic and conveyer belt and ground based, and aerial imaging have enhanced the invoice monitoring of plants [155]. For instance, Yang et al. [159] established a high-throughput nondestructive automatic phenotyping imaging device capable of monitoring fifteen agronomic traits, including plant compactness and grain projection areas of 5472 rice growing pots, leading to the identification of green revolution semi dwarf (SD1) genes through genome-wide association studies (GWASs). Additionally, high-throughput phenotyping relies heavily on imaging technologies, including multi-angle imaging for rice panicle numbers [154], structured light 3D imaging for filled and unfilled grain identification [160], and light emitting diode (LED) transmission imaging for rice yield trait characterization [161]. Additionally, 51 image-based traits related to drought responses were analyzed, using nondestructive high-throughput phenotyping and the identification of a new drought resistance gene (OsPP15) through genetic transformation experiments [162].
The integration of phenotype and OMICS technology improves the mutant characterization and identification of gene functions. For instance, the elemental profiling (Ionomics) of rice FOX Arabidopsis mutant lines revealed that OSZIP increases the Zn concentration [163], while metabolomic screening revealed that Os-LBD37/ASl39 is involved in nitrogen metabolic processes [113]. Additionally, the elemental profiling of T-DNA insertion mutant lines showed that OsNAC5 lowers iron and zinc concentrations in the leaves while increasing iron levels in the seeds compared to the wild-type [164]. Transcriptomic analyses revealed that the OsNRAMP5 gene is responsible for low Cd uptake and accumulation in EMS-mutagenized indica rice (9311) [165] and ion-beam irradiated seeds of japonica rice (Koshihikari) [166]. Furthermore, the ability to identify gene interactions further enhances the effectiveness of mutagenesis approaches. For example, loss-of-function studies, such as knocking out the OsMADS57 gene in an antisense manner, can demonstrate decreased tiller numbers in rice. However, proteomic and interatomic analyses reveal that complex interactions with OSTB1 proteins modulate rice tillering through DWARF14 [167]. The absence of such OMICS integration can challenge the understanding of compensatory mechanisms that may arise from the knockout.
Indeed, while gain-of-function and loss-of-function mutagenesis methods are valuable tools in rice functional genomics, their effectiveness for the advancement of rice functional genomics can substantially advance with the integration of high-throughput phenotyping, genotyping, and OMICS technologies.

6. Conclusions and Future Directions

The systematic identification of gene functions critically depends on phenotypic observations of loss-of-function or gain-of-function mutants. As demonstrated throughout this review, the establishment of large-scale mutant libraries has enabled genome-wide functional annotations at unprecedented scales. These resources have proven to be particularly valuable for overcoming the limitations of traditional single-gene approaches, allowing us to address complex biological questions regarding gene redundancy (e.g., via multiplexed CRISPR/Cas9 libraries), essential gene functions (e.g., via RNA interference and FOX hunting), and precise gene–trait relationships. Approximately 30% of rice genes belong to functionally overlapping families, obscuring the phenotypic effects in single-gene knockouts. The integration of high-throughput technologies has enhanced functional genomics. DNA barcoding coupled with next-generation sequencing has improved the efficiency of genotype–phenotype linkages, while high-throughput phenotyping helps for large-scale gene discovery.
Future research should prioritize the following: (1) development of scalable mutagenesis approaches. Multiplexed CRISPR/Cas9 mutagenesis enables the targeting of redundant gene families, while prime and base editing technologies can generate allelic series to study essential genes without lethal effects, overcoming the lethality hurdle of knockouts. These approaches will provide the necessary tools to achieve complete functional annotations of the rice genome; (2) conditional pooled RNAi knockdown to study lethal mutations. This approach complements CRISPR technologies by allowing for the temporal and spatial control of gene suppression, which is particularly valuable for investigating essential biological processes; (3) optimization of the DNA barcoding kit. This toolkit will be essential for large-scale library construction and mutant tracking. The use of multiplexed high-throughput sequencing will further enhance the capacity to manage and analyze ever expanding mutant collections for accelerating the rate of gene discovery; and (4) expanding the FOX libraries with wild rice (e.g., Oryza Rufipogon) cDNA to unlock novel alleles/genes. The utilization of wild genetic resources during mutant library establishment will help to discover potentially valuable alleles that have been lost during domestication processes. This approach may offer critical traits for tolerance and yield improvements. The integration of scalable mutagenesis approaches with high-throughput phenotyping (e.g., automated imaging) and multi-OMICS (e.g., metabolomic, ionomics) approaches will enable large-scale mutant characterization and accelerate the genomic discoveries and breeding applications.

Author Contributions

All the authors participated in the editing of this review. E.K.W. and Z.C. wrote the manuscript; M.L., D.X. and L.D.collected the data; L.L. and J.Z. (Jianwei Zhang) discussed the conceptualization; and J.Z. (Jian Zhang) and Z.L. reviewed and edited the manuscript. All the other authors provided suggestive comments on the review. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by the Nanfan special project of CAAS (YBXM2435) and Open Project Funding of the State Key Laboratory of Biocatalysis and Enzyme Engineering.

Acknowledgments

We thank all who have contributed to the establishment of rice FOX hunting libraries.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Solis, C.A.; Yong, M.T.; Vinarao, R.; Jena, K.; Holford, P.; Shabala, L.; Zhou, M.; Shabala, S.; Chen, Z.-H. Back to the wild: On a quest for donors toward salinity tolerant rice. Front. Plant Sci. 2020, 11, 323. [Google Scholar] [CrossRef] [PubMed]
  2. Hernández-Soto, A.; Echeverría-Beirute, F.; Abdelnour-Esquivel, A.; Valdez-Melara, M.; Boch, J.; Gatica-Arias, A. Rice breeding in the new era: Comparison of useful agronomic traits. Curr. Plant Biol. 2021, 27, 100211. [Google Scholar] [CrossRef]
  3. Mizuno, H.; Ito, K.; Wu, J.; Tanaka, T.; Kanamori, H.; Katayose, Y.; Sasaki, T.; Matsumoto, T. Identification and mapping of expressed genes, simple sequence repeats and transposable elements in centromeric regions of rice chromosomes. DNA Res. 2006, 13, 267–274. [Google Scholar] [CrossRef] [PubMed]
  4. Song, S.; Tian, D.; Zhang, Z.; Hu, S.; Yu, J. Rice genomics: Over the past two decades and into the future. Genom. Proteom. Bioinform. 2018, 16, 397–404. [Google Scholar] [CrossRef]
  5. Wang, C.; Han, B. Twenty years of rice genomics research: From sequencing and functional genomics to quantitative genomics. Mol. Plant 2022, 15, 593–619. [Google Scholar] [CrossRef]
  6. IRGS. The map-based sequence of the rice genome. Nature 2005, 436, 793–800. [Google Scholar] [CrossRef]
  7. Abe, K.; Ichikawa, H. Gene overexpression resources in cereals for functional genomics and discovery of useful genes. Front. Plant Sci. 2016, 7, 1359. [Google Scholar] [CrossRef]
  8. Lo, S.F.; Fan, M.J.; Hsing, Y.I.; Chen, L.J.; Chen, S.; Wen, I.C.; Liu, Y.L.; Chen, K.T.; Jiang, M.J.; Lin, M.K. Genetic resources offer efficient tools for rice functional genomics research. Plant Cell Environ. 2016, 39, 998–1013. [Google Scholar] [CrossRef]
  9. MVD. Joint FAO/IAEA Mutant Variety Database. 2022. Available online: https://mvd.iaea.org/ (accessed on 15 September 2024).
  10. Luz, V.K.d.; Silveira, S.F.d.S.; Fonseca, G.M.d.; Groli, E.L.; Figueiredo, R.G.; Baretta, D.; Kopp, M.M.; Magalhães, A.M.d.; Maia, L.C.d.; Oliveira, A.C.d. Identification of variability for agronomically important traits in rice mutant families. Bragantia 2016, 75, 41–50. [Google Scholar] [CrossRef]
  11. Manikandan, V.; Vanniarajan, C. Induced macromutational spectrum and frequency of viable mutants in M2 generation of rice (Oryza sativa L.). Int. J. Curr. Microbiol. Appl. Sci. 2017, 6, 1825–1834. [Google Scholar] [CrossRef]
  12. Oladosu, Y.; Rafii, M.Y.; Abdullah, N.; Hussin, G.; Ramli, A.; Rahim, H.A.; Miah, G.; Usman, M. Principle and application of plant mutagenesis in crop improvement: A review. Biotechnol. Biotechnol. Equip. 2016, 30, 1–16. [Google Scholar] [CrossRef]
  13. Viana, V.E.; Pegoraro, C.; Busanello, C.; Costa de Oliveira, A. Mutagenesis in rice: The basis for breeding a new super plant. Front. Plant Sci. 2019, 10, 1326. [Google Scholar] [CrossRef]
  14. Parry, M.A.; Madgwick, P.J.; Bayon, C.; Tearall, K.; Hernandez-Lopez, A.; Baudo, M.; Rakszegi, M.; Hamada, W.; Al-Yassin, A.; Ouabbou, H. Mutation discovery for crop improvement. J. Exp. Bot. 2009, 60, 2817–2825. [Google Scholar] [CrossRef] [PubMed]
  15. Shen, S.; Cheng, S.; Wu, D.; Xia, Y.; Shu, Q. Mutagenic Improvement of Shattering Characteristic of the Restorer Line of a Hybrid Rice “Xieyou 9308”. Rice Sci. 2004, 11, 222. [Google Scholar]
  16. Shen, S.; Wu, D.; Gao, M.; Xia, Y.; Shu, Q. An elite restorer mutant R3027 induced by the integral application of {gamma}-rays irradiation and somaclonal variation techniques. Acta Agric. Nucleatae Sin. 2003, 17, 165–170. [Google Scholar]
  17. Talebi, A.B.; Shahrokhifar, B. Ethyl methane sulphonate (EMS) induced mutagenesis in Malaysian rice (cv. MR219) for lethal dose determination. Am. J. Plant Sci. 2012, 3, 1661–1665. [Google Scholar] [CrossRef]
  18. Nawaz, Z.; Shu, Q. Molecular nature of chemically and physically induced mutants in plants: A review. Plant Genet. Resour. 2014, 12 (Suppl. S1), S74–S78. [Google Scholar] [CrossRef]
  19. Li, G.; Jain, R.; Chern, M.; Pham, N.T.; Martin, J.A.; Wei, T.; Schackwitz, W.S.; Lipzen, A.M.; Duong, P.Q.; Jones, K.C. The sequences of 1504 mutants in the model rice variety Kitaake facilitate rapid functional genomic studies. Plant Cell 2017, 29, 1218–1231. [Google Scholar] [CrossRef]
  20. Li, Y.; Xiao, J.; Chen, L.; Huang, X.; Cheng, Z.; Han, B.; Zhang, Q.; Wu, C. Rice functional genomics research: Past decade and future. Mol. Plant 2018, 11, 359–380. [Google Scholar] [CrossRef]
  21. Wu, J.-L.; Wu, C.; Lei, C.; Baraoidan, M.; Bordeos, A.; Madamba, M.R.S.; Ramos-Pamplona, M.; Mauleon, R.; Portugal, A.; Ulat, V.J. Chemical-and irradiation-induced mutants of indica rice IR64 for forward and reverse genetics. Plant Mol. Biol. 2005, 59, 85–97. [Google Scholar] [CrossRef]
  22. Krishnan, A.; Guiderdoni, E.; An, G.; Hsing, Y.I.; Han, C.D.; Lee, M.C.; Yu, S.M.; Upadhyaya, N.; Ramachandran, S.; Zhang, Q.; et al. Mutant resources in rice for functional genomics of the grasses. Plant Physiol. 2009, 149, 165–170. [Google Scholar] [CrossRef] [PubMed]
  23. Ram, H.; Soni, P.; Salvi, P.; Gandass, N.; Sharma, A.; Kaur, A.; Sharma, T.R. Insertional Mutagenesis Approaches and Their Use in Rice for Functional Genomics. Plants 2019, 8, 310. [Google Scholar] [CrossRef] [PubMed]
  24. Kuromori, T.; Takahashi, S.; Kondou, Y.; Shinozaki, K.; Matsui, M. Phenome analysis in plant species using loss-of-function and gain-of-function mutants. Plant Cell Physiol. 2009, 50, 1215–1231. [Google Scholar] [CrossRef]
  25. Wang, N.; Long, T.; Yao, W.; Xiong, L.; Zhang, Q.; Wu, C. Mutant resources for the functional analysis of the rice genome. Mol. Plant 2013, 6, 596–604. [Google Scholar] [CrossRef] [PubMed]
  26. Baharum, S.N.; Azizan, K.A. Metabolomics in systems biology. In Omics Applications for Systems Biology; Springer: Berlin/Heidelberg, Germany, 2018; pp. 51–68. [Google Scholar]
  27. Jalil, S.; Ali, Q.; Khan, A.U.; Nazir, M.M.; Ali, S.; Zulfiqar, F.; Javed, M.A.; Jin, X. Molecular and biochemical characterization of rice developed through conventional integration of nDart1-0 transposon gene. Sci. Rep. 2023, 13, 8139. [Google Scholar] [CrossRef]
  28. Jeon, J.S.; Lee, S.; Jung, K.H.; Jun, S.H.; Jeong, D.H.; Lee, J.; Kim, C.; Jang, S.; Yang, K.; Nam, J.; et al. T-DNA insertional mutagenesis for functional genomics in rice. Plant J. 2000, 22, 561–570. [Google Scholar] [CrossRef]
  29. Pucker, B.; Kleinbölting, N.; Weisshaar, B. Large scale genomic rearrangements in selected Arabidopsis thaliana T-DNA lines are caused by T-DNA insertion mutagenesis. BMC Genom. 2021, 22, 599. [Google Scholar] [CrossRef]
  30. Figueroa-Bossi, N.; Balbontín, R.; Bossi, L. Mapping transposon insertion sites by inverse polymerase chain reaction and Sanger sequencing. Cold Spring Harb. Protoc. 2024, 2024, pdb-prot108197. [Google Scholar]
  31. Sha, Y.; Li, S.; Pei, Z.; Luo, L.; Tian, Y.; He, C. Generation and flanking sequence analysis of a rice T-DNA tagged population. Theor. Appl. Genet. 2004, 108, 306–314. [Google Scholar] [CrossRef]
  32. Thole, V.; Worland, B.; Wright, J.; Bevan, M.W.; Vain, P. Distribution and characterization of more than 1000 T-DNA tags in the genome of Brachypodium distachyon community standard line Bd21. Plant Biotechnol.J. 2010, 8, 734–747. [Google Scholar] [CrossRef]
  33. Gentile, A.; D’Alessandro, L.; Medico, E. Gene trapping: A multi-purpose tool for functional genomics. Biotechnol. Genet. Eng 2003, 20, 77–100. [Google Scholar] [CrossRef] [PubMed]
  34. Serrania, J.; Johner, T.; Rupp, O.; Goesmann, A.; Becker, A. Massive parallel insertion site sequencing of an arrayed Sinorhizobium meliloti signature-tagged mini-Tn 5 transposon mutant library. J. Biotechnol. 2017, 257, 9–12. [Google Scholar] [CrossRef] [PubMed]
  35. Wan, S.; Wu, J.; Zhang, Z.; Sun, X.; Lv, Y.; Gao, C.; Ning, Y.; Ma, J.; Guo, Y.; Zhang, Q.; et al. Activation tagging, an efficient tool for functional analysis of the rice genome. Plant Mol. Biol. 2009, 69, 69–80. [Google Scholar] [CrossRef]
  36. Jeong, D.H.; An, S.; Park, S.; Kang, H.G.; Park, G.G.; Kim, S.R.; Sim, J.; Kim, Y.O.; Kim, M.K.; Kim, S.R. Generation of a flanking sequence-tag database for activation-tagging lines in japonica rice. Plant J. 2006, 45, 123–132. [Google Scholar] [CrossRef]
  37. Johnson, A.A.; Hibberd, J.M.; Gay, C.; Essah, P.A.; Haseloff, J.; Tester, M.; Guiderdoni, E. Spatial control of transgene expression in rice (Oryza sativa L.) using the GAL4 enhancer trapping system. Plant J. 2005, 41, 779–789. [Google Scholar] [CrossRef] [PubMed]
  38. Sallaud, C.; Gay, C.; Larmande, P.; Bès, M.; Piffanelli, P.; Piégu, B.; Droc, G.; Regad, F.; Bourgeois, E.; Meynard, D.; et al. High throughput T-DNA insertion mutagenesis in rice: A first step towards in silico reverse genetics. Plant J. 2004, 39, 450–464. [Google Scholar] [CrossRef]
  39. Zhang, J.; Li, C.; Wu, C.; Xiong, L.; Chen, G.; Zhang, Q.; Wang, S. RMD: A rice mutant database for functional analysis of the rice genome. Nucleic Acids Res. 2006, 34, D745–D748. [Google Scholar] [CrossRef]
  40. Chern, C.-G.; Fan, M.-J.; Yu, S.-M.; Hour, A.-L.; Lu, P.-C.; Lin, Y.-C.; Wei, F.-J.; Huang, S.-C.; Chen, S.; Lai, M.-H. A rice phenomics study—Phenotype scoring and seed propagation of a T-DNA insertion-induced rice mutant population. Plant Mol.Biol. 2007, 65, 427–438. [Google Scholar] [CrossRef]
  41. Wang, L.; Zheng, J.; Luo, Y.; Xu, T.; Zhang, Q.; Zhang, L.; Xu, M.; Wan, J.; Wang, M.B.; Zhang, C.; et al. Construction of a genomewide RNAi mutant library in rice. Plant Biotechnol. J. 2013, 11, 997–1005. [Google Scholar] [CrossRef]
  42. Upadhyaya, N.M.; Zhu, Q.H.; Bhat, R.S. Transposon insertional mutagenesis in rice. Methods Mol. Biol. 2011, 678, 147–177. [Google Scholar]
  43. Takano, M.; Kanegae, H.; Shinomura, T.; Miyao, A.; Hirochika, H.; Furuya, M. Isolation and characterization of rice phytochrome A mutants. Plant Cell 2001, 13, 521–534. [Google Scholar] [CrossRef] [PubMed]
  44. Zhu, Q.H.; Ramm, K.; Shivakkumar, R.; Dennis, E.S.; Upadhyaya, N.M. The ANTHER INDEHISCENCE1 gene encoding a single MYB domain protein is involved in anther development in rice. Plant Physiol. 2004, 135, 1514–1525. [Google Scholar] [CrossRef]
  45. Ramamoorthy, R.; Jiang, S.Y.; Ramachandran, S. Oryza sativa cytochrome P450 family member OsCYP96B4 reduces plant height in a transcript dosage dependent manner. PLoS ONE 2011, 6, e28069. [Google Scholar] [CrossRef] [PubMed]
  46. Margis-Pinheiro, M.; Zhou, X.-R.; Zhu, Q.-H.; Dennis, E.S.; Upadhyaya, N.M. Isolation and characterization of a Ds-tagged rice (Oryza sativa L.) GA-responsive dwarf mutant defective in an early step of the gibberellin biosynthesis pathway. Plant Cell Rep. 2005, 23, 819–833. [Google Scholar] [CrossRef]
  47. Jiang, S.-Y.; Cai, M.; Ramachandran, S. ORYZA SATIVA MYOSIN XI B controls pollen development by photoperiod-sensitive protein localizations. Dev.Bio. 2007, 304, 579–592. [Google Scholar] [CrossRef] [PubMed]
  48. Wu, C.; You, C.; Li, C.; Long, T.; Chen, G.; Byrne, M.E.; Zhang, Q. RID1, encoding a Cys2/His2-type zinc finger transcription factor, acts as a master switch from vegetative to floral development in rice. Proc. Natl. Acad. Sci. USA 2008, 105, 12915–12920. [Google Scholar] [CrossRef]
  49. Sun, Q.; Zhou, D.X. Rice jmjC domain-containing gene JMJ706 encodes H3K9 demethylase required for floral organ development. Proc. Natl. Acad. Sci. USA 2008, 105, 13679–13684. [Google Scholar] [CrossRef]
  50. Wei, S.; Hu, W.; Deng, X.; Zhang, Y.; Liu, X.; Zhao, X.; Luo, Q.; Jin, Z.; Li, Y.; Zhou, S.; et al. A rice calcium-dependent protein kinase OsCPK9 positively regulates drought stress tolerance and spikelet fertility. BMC Plant Biol. 2014, 14, 133. [Google Scholar] [CrossRef]
  51. Ning, J.; Zhang, B.; Wang, N.; Zhou, Y.; Xiong, L. Increased leaf angle1, a Raf-like MAPKKK that interacts with a nuclear protein family, regulates mechanical tissue formation in the Lamina joint of rice. Plant Cell 2011, 23, 4334–4347. [Google Scholar] [CrossRef]
  52. Du, H.; Wang, N.; Cui, F.; Li, X.; Xiao, J.; Xiong, L. Characterization of the beta-carotene hydroxylase gene DSM2 conferring drought and oxidative stress resistance by increasing xanthophylls and abscisic acid synthesis in rice. Plant Physiol. 2010, 154, 1304–1318. [Google Scholar] [CrossRef]
  53. Hasegawa, K.; Kamada, S.; Takehara, S.; Takeuchi, H.; Nakamura, A.; Satoh, S.; Iwai, H. Rice Putative Methyltransferase Gene OsPMT16 Is Required for Pistil Development Involving Pectin Modification. Front. Plant Sci. 2020, 11, 475. [Google Scholar] [CrossRef] [PubMed]
  54. Yi, J.; Lee, Y.S.; Lee, D.Y.; Cho, M.H.; Jeon, J.S.; An, G. OsMPK6 plays a critical role in cell differentiation during early embryogenesis in Oryza sativa. J. Exp. Bot. 2016, 67, 2425–2437. [Google Scholar] [CrossRef]
  55. Yi, J.; Moon, S.; Lee, Y.S.; Zhu, L.; Liang, W.; Zhang, D.; Jung, K.H.; An, G. Defective Tapetum Cell Death 1 (DTC1) Regulates ROS Levels by Binding to Metallothionein during Tapetum Degeneration. Plant Physiol. 2016, 170, 1611–1623. [Google Scholar] [CrossRef]
  56. Kang, H.G.; Park, S.; Matsuoka, M.; An, G. White-core endosperm floury endosperm-4 in rice is generated by knockout mutations in the C4-type pyruvate orthophosphate dikinase gene (OsPPDKB). Plant J. 2005, 42, 901–911. [Google Scholar] [CrossRef] [PubMed]
  57. Chang, Y.; Long, T.; Wu, C. Effort and contribution of T-DNA Insertion mutant library for rice functional genomics research in China: Review and perspective. J. Integr. Plant Biol. 2012, 54, 953–966. [Google Scholar] [CrossRef] [PubMed]
  58. Wu, C.; Li, X.; Yuan, W.; Chen, G.; Kilian, A.; Li, J.; Xu, C.; Li, X.; Zhou, D.X.; Wang, S. Development of enhancer trap lines for functional analysis of the rice genome. Plant J. 2003, 35, 418–427. [Google Scholar] [CrossRef]
  59. Wei, F.J.; Tsai, Y.C.; Hsu, Y.M.; Chen, Y.A.; Huang, C.T.; Wu, H.P.; Huang, L.T.; Lai, M.H.; Kuang, L.Y.; Lo, S.F.; et al. Lack of Genotype and Phenotype Correlation in a Rice T-DNA Tagged Line Is Likely Caused by Introgression in the Seed Source. PLoS ONE 2016, 11, e0155768. [Google Scholar] [CrossRef]
  60. Jiang, Y.; Cai, Z.; Xie, W.; Long, T.; Yu, H.; Zhang, Q. Rice functional genomics research: Progress and implications for crop genetic improvement. Biotechnol. Adv. 2012, 30, 1059–1070. [Google Scholar] [CrossRef]
  61. Zhang, J.; Guo, D.; Chang, Y.; You, C.; Li, X.; Dai, X.; Weng, Q.; Zhang, J.; Chen, G.; Li, X. Non-random distribution of T-DNA insertions at various levels of the genome hierarchy as revealed by analyzing 13 804 T-DNA flanking sequences from an enhancer-trap mutant library. Plant J. 2007, 49, 947–959. [Google Scholar] [CrossRef]
  62. Gong, W.; Zhou, Y.; Wang, R.; Wei, X.; Zhang, L.; Dai, Y.; Zhu, Z. Analysis of T-DNA integration events in transgenic rice. J. Plant Physiol. 2021, 266, 153527. [Google Scholar] [CrossRef]
  63. Hong, W.J.; Kim, Y.J.; Kim, E.J.; Kumar Nalini Chandran, A.; Moon, S.; Gho, Y.S.; Yoou, M.H.; Kim, S.T.; Jung, K.H. CAFRI-Rice: CRISPR applicable functional redundancy inspector to accelerate functional genomics in rice. Plant J. 2020, 104, 532–545. [Google Scholar] [CrossRef] [PubMed]
  64. Gaillochet, C.; Develtere, W.; Jacobs, T.B. CRISPR screens in plants: Approaches, guidelines, and future prospects. Plant Cell 2021, 33, 794–813. [Google Scholar] [CrossRef]
  65. Park, S.R.; Son, S. CRISPR/Cas9-based mutant library screening: The discovery of novel genes regulating immune responses in cotton and rice. Front. Plant Sci. 2024, 15, 1501092. [Google Scholar] [CrossRef]
  66. Arora, L.; Narula, A. Gene Editing and Crop Improvement Using CRISPR-Cas9 System. Front. Plant Sci. 2017, 8, 1932. [Google Scholar] [CrossRef]
  67. Lowder, L.G.; Zhou, J.; Zhang, Y.; Malzahn, A.; Zhong, Z.; Hsieh, T.F.; Voytas, D.F.; Zhang, Y.; Qi, Y. Robust Transcriptional Activation in Plants Using Multiplexed CRISPR-Act2.0 and mTALE-Act Systems. Mol. Plant 2018, 11, 245–256. [Google Scholar] [CrossRef] [PubMed]
  68. Char, S.N.; Li, R.; Yang, B. CRISPR/Cas9 for mutagenesis in rice. In Transgenic Plants: Methods and Protocols; Springer: Berlin/Heidelberg, Germany, 2019; pp. 279–293. [Google Scholar]
  69. Chen, F.; Chen, L.; Yan, Z.; Xu, J.; Feng, L.; He, N.; Guo, M.; Zhao, J.; Chen, Z.; Chen, H.; et al. Recent advances of CRISPR-based genome editing for enhancing staple crops. Front. Plant Sci. 2024, 15, 1478398. [Google Scholar] [CrossRef]
  70. Sun, L.; Alariqi, M.; Wang, Y.; Wang, Q.; Xu, Z.; Zafar, M.N.; Yang, G.; Jia, R.; Hussain, A.; Chen, Y.; et al. Construction of Host Plant Insect-Resistance Mutant Library by High-Throughput CRISPR/Cas9 System and Identification of A Broad-Spectrum Insect Resistance Gene. Adv. Sci. 2024, 11, e2306157. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, F.; Liang, S.; Wang, G.; Hu, T.; Fu, C.; Wang, Q.; Xu, Z.; Fan, Y.; Che, L.; Min, L.; et al. CRISPR-Cas9-mediated construction of a cotton CDPK mutant library for identification of insect-resistance genes. Plant Commun. 2024, 5, 101047. [Google Scholar] [CrossRef]
  72. Lu, Y.; Ye, X.; Guo, R.; Huang, J.; Wang, W.; Tang, J.; Tan, L.; Zhu, J.K.; Chu, C.; Qian, Y. Genome-wide Targeted Mutagenesis in Rice Using the CRISPR/Cas9 System. Mol. Plant 2017, 10, 1242–1245. [Google Scholar] [CrossRef]
  73. Till, B.J.; Cooper, J.; Tai, T.H.; Colowit, P.; Greene, E.A.; Henikoff, S.; Comai, L. Discovery of chemically induced mutations in rice by TILLING. BMC Plant Biol. 2007, 7, 19. [Google Scholar] [CrossRef]
  74. Kolesnik, T.; Szeverenyi, I.; Bachmann, D.; Kumar, C.S.; Jiang, S.; Ramamoorthy, R.; Cai, M.; Ma, Z.G.; Sundaresan, V.; Ramachandran, S. Establishing an efficient Ac/Ds tagging system in rice: Large-scale analysis of Ds flanking sequences. Plant J. 2004, 37, 301–314. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, K.; Ke, R.; Du, M.; Yi, Y.; Chen, Y.; Wang, X.; Yao, L.; Liu, H.; Hou, X.; Xiong, L.; et al. A FLASH pipeline for arrayed CRISPR library construction and the gene function discovery of rice receptor-like kinases. Mol. Plant 2022, 15, 243–257. [Google Scholar] [CrossRef]
  76. Son, S.; Song, G.; Nam, S.; Lee, G.; Im, J.; Lee, K.S.; Park, Y.J.; Suh, E.J.; Park, S.R. CRISPR/Cas9-mediated mutagenesis of rice NAC transcription factor genes results in altered innate immunity. Plant Physiol. 2024, 195, 1138–1142. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, L.; Luo, Y.Z.; Zhang, L.; Jiao, X.M.; Wang, M.B.; Fan, Y.L. Rolling circle amplification-mediated hairpin RNA (RMHR) library construction in plants. Nucleic Acids Res. 2008, 36, e149. [Google Scholar] [CrossRef] [PubMed]
  78. Weigel, D.; Ahn, J.H.; Blázquez, M.A.; Borevitz, J.O.; Christensen, S.K.; Fankhauser, C.; Ferrándiz, C.; Kardailsky, I.; Malancharuvil, E.J.; Neff, M.M. Activation tagging in Arabidopsis. Plant Physiol. 2000, 122, 1003–1014. [Google Scholar] [CrossRef]
  79. Ichikawa, T.; Nakazawa, M.; Kawashima, M.; Iizumi, H.; Kuroda, H.; Kondou, Y.; Tsuhara, Y.; Suzuki, K.; Ishikawa, A.; Seki, M. The FOX hunting system: An alternative gain-of-function gene hunting technique. Plant J. 2006, 48, 974–985. [Google Scholar] [CrossRef]
  80. Nakamura, H.; Hakata, M.; Amano, K.; Miyao, A.; Toki, N.; Kajikawa, M.; Pang, J.; Higashi, N.; Ando, S.; Toki, S. A genome-wide gain-of-function analysis of rice genes using the FOX-hunting system. Plant Mol. Biol. 2007, 65, 357–371. [Google Scholar] [CrossRef]
  81. Meng, X.; Yu, H.; Zhang, Y.; Zhuang, F.; Song, X.; Gao, S.; Gao, C.; Li, J. Construction of a Genome-Wide Mutant Library in Rice Using CRISPR/Cas9. Mol. Plant 2017, 10, 1238–1241. [Google Scholar] [CrossRef]
  82. Zhao, D.; Chen, S.; Han, Y.; Liu, G.; Liu, J.; Yang, Q.; Zhang, T.; Shen, J.; Fan, X.; Zhang, C.; et al. A CRISPR/Cas9-mediated mutant library of seed-preferred genes in rice. Plant Biotechnol. J. 2024, 22, 3012–3014. [Google Scholar] [CrossRef]
  83. Younis, A.; Siddique, M.I.; Kim, C.-K.; Lim, K.-B. RNA interference (RNAi) induced gene silencing: A promising approach of hi-tech plant breeding. Int. J.Biol. Sci. 2014, 10, 1150. [Google Scholar] [CrossRef]
  84. Guo, Q.; Liu, Q.; Smith, N.A.; Liang, G.; Wang, M.B. RNA Silencing in Plants: Mechanisms, Technologies and Applications in Horticultural Crops. Curr. Genom. 2016, 17, 476–489. [Google Scholar] [CrossRef]
  85. Malakondaiah, S.; Julius, A.; Ponnambalam, D.; Gunthoti, S.S.; Ashok, J.; Krishana, P.S.; Rebecca, J. Gene silencing by RNA interference: A review. Genome Instab. Dis. 2024, 5, 225–241. [Google Scholar] [CrossRef]
  86. Hirochika, H.; Guiderdoni, E.; An, G.; Hsing, Y.-I.; Eun, M.Y.; Han, C.-D.; Upadhyaya, N.; Ramachandran, S.; Zhang, Q.; Pereira, A. Rice mutant resources for gene discovery. Plant Mol. Biol. 2004, 54, 325–334. [Google Scholar] [CrossRef] [PubMed]
  87. Mei, Q.; Fu, Y.W.; Li, T.M.; Xuan, Y.H. Ac/Ds-Induced Receptor-like Kinase Genes Deletion Provides Broad-Spectrum Resistance to Bacterial Blight in Rice. Int. J. Mol. Sci. 2022, 23, 4561. [Google Scholar] [CrossRef]
  88. Huang, C.K.; Sie, Y.S.; Chen, Y.F.; Huang, T.S.; Lu, C.A. Two highly similar DEAD box proteins, OsRH2 and OsRH34, homologous to eukaryotic initiation factor 4AIII, play roles of the exon junction complex in regulating growth and development in rice. BMC Plant Biol. 2016, 16, 84. [Google Scholar] [CrossRef] [PubMed]
  89. Li, Z.; Woo, H.R.; Guo, H. Genetic redundancy of senescence-associated transcription factors in Arabidopsis. J. Exp. Bot. 2018, 69, 811–823. [Google Scholar] [CrossRef]
  90. Ling, J.; Li, R.; Nwafor, C.C.; Cheng, J.; Li, M.; Xu, Q.; Wu, J.; Gan, L.; Yang, Q.; Liu, C. Development of iFOX-hunting as a functional genomic tool and demonstration of its use to identify early senescence-related genes in the polyploid Brassica napus. Plant Biotechnol. J. 2018, 16, 591–602. [Google Scholar] [CrossRef]
  91. Cheng, H.; Gao, J.; Cai, H.; Zhu, J.; Huang, H. Gain-of-function in Arabidopsis (GAINA) for identifying functional genes in Hevea brasiliensis. SpringerPlus 2016, 5, 1853. [Google Scholar] [CrossRef]
  92. Pathak, B.; Maurya, C.; Faria, M.C.; Alizada, Z.; Nandy, S.; Zhao, S.; Jamsheer, K.M.; Srivastava, V. Targeting TOR and SnRK1 Genes in Rice with CRISPR/Cas9. Plants 2022, 11, 1453. [Google Scholar] [CrossRef]
  93. Zhao, S.; Luo, J.; Zeng, X.; Li, K.; Yuan, R.; Zhu, L.; Li, X.; Wu, G.; Yan, X. Rolling Circle Amplification (RCA)-Mediated Genome-Wide ihpRNAi Mutant Library Construction in Brassica napus. Int. J. Mol. Sci. 2020, 21, 7243. [Google Scholar] [CrossRef]
  94. Hauser, F.; Ceciliato, P.H.O.; Lin, Y.C.; Guo, D.; Gregerson, J.D.; Abbasi, N.; Youhanna, D.; Park, J.; Dubeaux, G.; Shani, E.; et al. A seed resource for screening functionally redundant genes and isolation of new mutants impaired in CO2 and ABA responses. J.Exp.Bot. 2019, 70, 641–651. [Google Scholar] [CrossRef] [PubMed]
  95. Hauser, F.; Chen, W.; Deinlein, U.; Chang, K.; Ossowski, S.; Fitz, J.; Hannon, G.J.; Schroeder, J.I. A genomic-scale artificial microRNA library as a tool to investigate the functionally redundant gene space in Arabidopsis. Plant Cell 2013, 25, 2848–2863. [Google Scholar] [CrossRef]
  96. Jover-Gil, S.; Paz-Ares, J.; Micol, J.L.; Ponce, M.R. Multi-gene silencing in Arabidopsis: A collection of artificial microRNAs targeting groups of paralogs encoding transcription factors. Plant J. 2014, 80, 149–160. [Google Scholar] [CrossRef] [PubMed]
  97. Moin, M.; Bakshi, A.; Saha, A.; Dutta, M.; Kirti, P. Gain-of-function mutagenesis approaches in rice for functional genomics and improvement of crop productivity. Brief. Funct. Genom 2017, 16, 238–247. [Google Scholar] [CrossRef]
  98. Deng, M.; Wang, Y.; Kuzma, M.; Chalifoux, M.; Tremblay, L.; Yang, S.; Ying, J.; Sample, A.; Wang, H.M.; Griffiths, R. Activation tagging identifies Arabidopsis transcription factor AtMYB68 for heat and drought tolerance at yield determining reproductive stages. Plant J. 2020, 104, 1535–1550. [Google Scholar] [CrossRef]
  99. Kota, V.R.; Gundra, S.R.; Vudem, D.R.; Pulugurtha, B.K.; Khareedu, V.R. Development of a large population of activation-tagged mutants in an elite indica rice variety. Plant Breed. 2020, 139, 328–343. [Google Scholar] [CrossRef]
  100. Plemmons, A.N.; Askins, A.R.; Mendoza, S.M.; Hancock, C.N. A Transposon-Based Activation Tag System for Functional Genomics in Cereals: Detection of Mping-Based Activation Tag Mobilization in Wheat. In Accelerated Breeding of Cereal Crops; Springer: Berlin/Heidelberg, Germany, 2022; pp. 195–207. [Google Scholar]
  101. Mahmoud, M.; Zhou, Z.; Kaur, R.; Bekele, W.; Tinker, N.A.; Singh, J. Toward the development of Ac/Ds transposon-mediated gene tagging system for functional genomics in oat (Avena sativa L.). Funct. Integr. Genom 2022, 22, 669–681. [Google Scholar] [CrossRef]
  102. Mathews, H.; Clendennen, S.K.; Caldwell, C.G.; Liu, X.L.; Connors, K.; Matheis, N.; Schuster, D.K.; Menasco, D.; Wagoner, W.; Lightner, J. Activation tagging in tomato identifies a transcriptional regulator of anthocyanin biosynthesis, modification, and transport. Plant Cell 2003, 15, 1689–1703. [Google Scholar] [CrossRef]
  103. Ayliffe, M.A.; Pryor, A.J. Activation tagging and insertional mutagenesis in barley. In Plant Reverse Genetics Methods and Protocols; Springer: Berlin/Heidelberg, Germany, 2011; pp. 107–128. [Google Scholar]
  104. An, S.; Park, S.; Jeong, D.-H.; Lee, D.-Y.; Kang, H.-G.; Yu, J.-H.; Hur, J.; Kim, S.-R.; Kim, Y.-H.; Lee, M. Generation and analysis of end sequence database for T-DNA tagging lines in rice. Plant Physiol. 2003, 133, 2040–2047. [Google Scholar] [CrossRef]
  105. Chen, S.; Jin, W.; Wang, M.; Zhang, F.; Zhou, J.; Jia, Q.; Wu, Y.; Liu, F.; Wu, P. Distribution and characterization of over 1000 T-DNA tags in rice genome. Plant J. 2003, 36, 105–113. [Google Scholar] [CrossRef]
  106. Hsing, Y.-I.; Chern, C.-G.; Fan, M.-J.; Lu, P.-C.; Chen, K.-T.; Lo, S.-F.; Sun, P.-K.; Ho, S.-L.; Lee, K.-W.; Wang, Y.-C. A rice gene activation/knockout mutant resource for high throughput functional genomics. Plant Mol. Biol. 2007, 63, 351–364. [Google Scholar] [CrossRef] [PubMed]
  107. Mori, M.; Tomita, C.; Sugimoto, K.; Hasegawa, M.; Hayashi, N.; Dubouzet, J.G.; Ochiai, H.; Sekimoto, H.; Hirochika, H.; Kikuchi, S. Isolation and molecular characterization of a Spotted leaf 18 mutant by modified activation-tagging in rice. Plant Mol. Biol. 2007, 63, 847–860. [Google Scholar] [CrossRef]
  108. Qu, S.; Desai, A.; Wing, R.; Sundaresan, V. A versatile transposon-based activation tag vector system for functional genomics in cereals and other monocot plants. Plant Physiol. 2008, 146, 189–199. [Google Scholar] [CrossRef] [PubMed]
  109. Dutta, M.; Moin, M.; Saha, A.; Dutta, D.; Bakshi, A.; Kirti, P. Gain-of-function mutagenesis through activation tagging identifies XPB2 and SEN1 helicase genes as potential targets for drought stress tolerance in rice. Theor. Appl. Genet. 2021, 134, 2253–2272. [Google Scholar] [CrossRef] [PubMed]
  110. Lu, G.; Wang, X.; Liu, J.; Yu, K.; Gao, Y.; Liu, H.; Wang, C.; Wang, W.; Wang, G.; Liu, M.; et al. Application of T-DNA activation tagging to identify glutamate receptor-like genes that enhance drought tolerance in plants. Plant Cell Rep. 2014, 33, 617–631. [Google Scholar] [CrossRef]
  111. Gandikota, M.; Krishnakanth Yadav, T.; Maram, R.R.; Kalluru, S.; Sena, M.B.; Siddiq, E.; Kalinati Narasimhan, Y.; Vemireddy, L.R.; Ghanta, A. Development of activation-tagged gain-of-functional mutants in indica rice line (BPT 5204) for sheath blight resistance. Mo. Biol. Rep. 2024, 51, 381. [Google Scholar] [CrossRef]
  112. Kondou, Y.; Higuchi, M.; Takahashi, S.; Sakurai, T.; Ichikawa, T.; Kuroda, H.; Yoshizumi, T.; Tsumoto, Y.; Horii, Y.; Kawashima, M. Systematic approaches to using the FOX hunting system to identify useful rice genes. Plant J. 2009, 57, 883–894. [Google Scholar] [CrossRef]
  113. Albinsky, D.; Kusano, M.; Higuchi, M.; Hayashi, N.; Kobayashi, M.; Fukushima, A.; Mori, M.; Ichikawa, T.; Matsui, K.; Kuroda, H. Metabolomic screening applied to rice FOX Arabidopsis lines leads to the identification of a gene-changing nitrogen metabolism. Mol. Plant 2010, 3, 125–142. [Google Scholar] [CrossRef]
  114. Higashi, Y.; Ohama, N.; Ishikawa, T.; Katori, T.; Shimura, A.; Kusakabe, K.; Yamaguchi-Shinozaki, K.; Ishida, J.; Tanaka, M.; Seki, M. HsfA1d, a protein identified via FOX hunting using Thellungiella salsuginea cDNAs improves heat tolerance by regulating heat-stress-responsive gene expression. Mol. Plant 2013, 6, 411–422. [Google Scholar] [CrossRef]
  115. Taji, T.; Sakurai, T.; Mochida, K.; Ishiwata, A.; Kurotani, A.; Totoki, Y.; Toyoda, A.; Sakaki, Y.; Seki, M.; Ono, H. Large-scale collection and annotation of full-length enriched cDNAs from a model halophyte, Thellungiella halophila. BMC Plant Biol. 2008, 8, 115. [Google Scholar] [CrossRef]
  116. Hakata, M.; Nakamura, H.; Iida-Okada, K.; Miyao, A.; Kajikawa, M.; Imai-Toki, N.; Pang, J.; Amano, K.; Horikawa, A.; Tsuchida-Mayama, T. Production and characterization of a large population of cDNA-overexpressing transgenic rice plants using Gateway-based full-length cDNA expression libraries. Breed. Sci. 2010, 60, 575–585. [Google Scholar] [CrossRef]
  117. Ogawa, Y.; Dansako, T.; Yano, K.; Sakurai, N.; Suzuki, H.; Aoki, K.; Noji, M.; Saito, K.; Shibata, D. Efficient and high-throughput vector construction and Agrobacterium-mediated transformation of Arabidopsis thaliana suspension-cultured cells for functional genomics. Plant Cell Physiol. 2008, 49, 242–250. [Google Scholar] [CrossRef]
  118. Papdi, C.; Abrahám, E.; Joseph, M.P.; Popescu, C.; Koncz, C.; Szabados, L. Functional identification of Arabidopsis stress regulatory genes using the controlled cDNA overexpression system. Plant Physiol. 2008, 147, 528–542. [Google Scholar] [CrossRef] [PubMed]
  119. Rauschendorfer, J.; Yordanov, Y.; Dobrev, P.; Vankova, R.; Sykes, R.; Külheim, C.; Busov, V. Overexpression of a developing xylem cDNA library in transgenic poplar generates high mutation rate specific to wood formation. Plant Biotechnol. J. 2020, 18, 1434–1443. [Google Scholar] [CrossRef]
  120. Kong, L.; Li, Z.; Song, Q.; Li, X.; Luo, K. Construction of a full-length cdna over-expressing library to identify valuable genes from Populus tomentosa. Int. J. Mol. Sci. 2021, 22, 3448. [Google Scholar] [CrossRef] [PubMed]
  121. Lin, L.; Cai, W.; Du, Z.; Zhang, W.; Xu, Q.; Sun, W.; Chen, M. Establishing a System for Functional Characterization of Full-Length cDNAs of Camellia sinensis. Int. J. Mol. Sci. 2019, 20, 5929. [Google Scholar] [CrossRef]
  122. Wu, J.; Zhang, Z.; Zhang, Q.; Liu, Y.; Zhu, B.; Cao, J.; Li, Z.; Han, L.; Jia, J.; Zhao, G. Generation of wheat transcription factor FOX rice lines and systematic screening for salt and osmotic stress tolerance. PLoS ONE 2015, 10, e0132314. [Google Scholar] [CrossRef]
  123. Fujita, M.; Mizukado, S.; Fujita, Y.; Ichikawa, T.; Nakazawa, M.; Seki, M.; Matsui, M.; Yamaguchi-Shinozaki, K.; Shinozaki, K. Identification of stress-tolerance-related transcription-factor genes via mini-scale Full-length cDNA Over-eXpressor (FOX) gene hunting system. Biochem. Biophys. Res. Commun. 2007, 364, 250–257. [Google Scholar] [CrossRef]
  124. Kikuchi, S.; Satoh, K.; Nagata, T.; Kawagashira, N.; Doi, K.; Kishimoto, N.; Yazaki, J.; Ishikawa, M.; Yamada, H.; Ooka, H. Collection, mapping, and annotation of over 28,000 cDNA clones from japonica rice. Science 2003, 301, 376–379. [Google Scholar] [CrossRef]
  125. Liu, X.; Lu, T.; Yu, S.; Li, Y.; Huang, Y.; Huang, T.; Zhang, L.; Zhu, J.; Zhao, Q.; Fan, D. A collection of 10,096 indica rice full-length cDNAs reveals highly expressed sequence divergence between Oryza sativa indica and japonica subspecies. Plant Mol. Biol. 2007, 65, 403–415. [Google Scholar] [CrossRef]
  126. Xie, K.; Zhang, J.; Xiang, Y.; Feng, Q.; Han, B.; Chu, Z.; Wang, S.; Zhang, Q.; Xiong, L. Isolation and annotation of 10828 putative full length cDNAs from indica rice. Sci. China Ser. C Life Sci. 2005, 48, 445–451. [Google Scholar] [CrossRef] [PubMed]
  127. Lu, T.; Yu, S.; Fan, D.; Mu, J.; Shangguan, Y.; Wang, Z.; Minobe, Y.; Lin, Z.; Han, B. Collection and comparative analysis of 1888 full-length cDNAs from wild rice Oryza rufipogon Griff. W1943. DNA Res. 2008, 15, 285–295. [Google Scholar] [CrossRef]
  128. Jain, M.; Khurana, P.; Tyagi, A.K.; Khurana, J.P. Genome-wide analysis of intronless genes in rice and Arabidopsis. Funct. Integr. Genom. 2008, 8, 69–78. [Google Scholar] [CrossRef] [PubMed]
  129. Aviña-Padilla, K.; Ramírez-Rafael, J.A.; Herrera-Oropeza, G.E.; Muley, V.Y.; Valdivia, D.I.; Díaz-Valenzuela, E.; García-García, A.; Varela-Echavarría, A.; Hernández-Rosales, M. Evolutionary perspective and expression analysis of intronless genes highlight the conservation of their regulatory role. Front. Genet. 2021, 12, 654256. [Google Scholar] [CrossRef]
  130. Shabalina, S.A.; Ogurtsov, A.Y.; Spiridonov, A.N.; Novichkov, P.S.; Spiridonov, N.A.; Koonin, E.V. Distinct patterns of expression and evolution of intronless and intron-containing mammalian genes. Mol. Biol. Evol. 2010, 27, 1745–1749. [Google Scholar] [CrossRef]
  131. Ariga, H.; Tanaka, T.; Ono, H.; Sakata, Y.; Hayashi, T.; Taji, T. CSP41b, a protein identified via FOX hunting using Eutrema salsugineum cDNAs, improves heat and salinity stress tolerance in transgenic Arabidopsis thaliana. Biochem. Biophys. Res. Commun. 2015, 464, 318–323. [Google Scholar] [CrossRef]
  132. Atwell, B.J.; Wang, H.; Scafaro, A.P. Could abiotic stress tolerance in wild relatives of rice be used to improve Oryza sativa? Plant Sci. 2014, 215–216, 48–58. [Google Scholar] [CrossRef] [PubMed]
  133. Bohra, A.; Kilian, B.; Sivasankar, S.; Caccamo, M.; Mba, C.; McCouch, S.R.; Varshney, R.K. Reap the crop wild relatives for breeding future crops. Trends Biotechnol. 2022, 40, 412–431. [Google Scholar] [CrossRef]
  134. Wisniewski, M.; Artlip, T.; Liu, J.; Ma, J.; Burchard, E.; Norelli, J.; Dardick, C. Fox hunting in wild apples: Searching for novel genes in Malus sieversii. Int. J. Mol. Sci. 2020, 21, 9516. [Google Scholar] [CrossRef]
  135. Kozich, J.J.; Westcott, S.L.; Baxter, N.T.; Highlander, S.K.; Schloss, P.D. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. App. Environ. Microbiol. 2013, 79, 5112–5120. [Google Scholar] [CrossRef]
  136. Bohmann, K.; Elbrecht, V.; Carøe, C.; Bista, I.; Leese, F.; Bunce, M.; Yu, D.W.; Seymour, M.; Dumbrell, A.J.; Creer, S. Strategies for sample labelling and library preparation in DNA metabarcoding studies. Mol. Ecol. Resour. 2022, 22, 1231–1246. [Google Scholar] [CrossRef] [PubMed]
  137. Cheng, Z.; Li, Q.; Deng, J.; Liu, Q.; Huang, X. The devil is in the details: Problems in DNA barcoding practices indicated by systematic evaluation of insect barcodes. Front. Ecol. Evol. 2023, 11, 1149839. [Google Scholar] [CrossRef]
  138. Pochon, X.; Bott, N.J.; Smith, K.F.; Wood, S.A. Evaluating detection limits of next-generation sequencing for the surveillance and monitoring of international marine pests. PLoS ONE 2013, 8, e73935. [Google Scholar] [CrossRef]
  139. Gous, A.; Swanevelder, D.Z.H.; Eardley, C.D.; Willows-Munro, S. Plant-pollinator interactions over time: Pollen metabarcoding from bees in a historic collection. Evol. Appl. 2019, 12, 187–197. [Google Scholar] [CrossRef] [PubMed]
  140. Lucas, A.; Bodger, O.; Brosi, B.J.; Ford, C.R.; Forman, D.W.; Greig, C.; Hegarty, M.; Jones, L.; Neyland, P.J.; de Vere, N. Floral resource partitioning by individuals within generalised hoverfly pollination networks revealed by DNA metabarcoding. Sci. Rep. 2018, 8, 5133. [Google Scholar] [CrossRef]
  141. Pjevac, P.; Hausmann, B.; Schwarz, J.; Kohl, G.; Herbold, C.; Loy, A.; Berry, D. An Economical and Flexible Dual Barcoding, Two-Step PCR Approach for Highly Multiplexed Amplicon Sequencing. Front. Microbiol. 2021, 12, 669776. [Google Scholar] [CrossRef]
  142. Lyons, E.; Sheridan, P.; Tremmel, G.; Miyano, S.; Sugano, S. Large-scale DNA barcode library generation for biomolecule identification in high-throughput screens. Sci. Rep. 2017, 7, 13899. [Google Scholar] [CrossRef]
  143. Carøe, C.; Bohmann, K. Tagsteady: A metabarcoding library preparation protocol to avoid false assignment of sequences to samples. Mol. Ecol. Resour. 2020, 20, 1620–1631. [Google Scholar] [CrossRef]
  144. Liu, Q.; Wang, C.; Jiao, X.; Zhang, H.; Song, L.; Li, Y.; Gao, C.; Wang, K. Hi-TOM: A platform for high-throughput tracking of mutations induced by CRISPR/Cas systems. Sci. China Life Sci. 2019, 62, 1–7. [Google Scholar] [CrossRef]
  145. Zhu, S.; Cao, Z.; Liu, Z.; He, Y.; Wang, Y.; Yuan, P.; Li, W.; Tian, F.; Bao, Y.; Wei, W. Guide RNAs with embedded barcodes boost CRISPR-pooled screens. Genome Biol. 2019, 20, 20. [Google Scholar] [CrossRef]
  146. Liu, H.-J.; Jian, L.; Xu, J.; Zhang, Q.; Zhang, M.; Jin, M.; Peng, Y.; Yan, J.; Han, B.; Liu, J. High-throughput CRISPR/Cas9 mutagenesis streamlines trait gene identification in maize. Plant Cell 2020, 32, 1397–1413. [Google Scholar] [CrossRef] [PubMed]
  147. Chen, B.-R.; Hale, D.C.; Ciolek, P.J.; Runge, K.W. Generation and analysis of a barcode-tagged insertion mutant library in the fission yeast Schizosaccharomyces pombe. BMC Genom. 2012, 13, 161. [Google Scholar] [CrossRef] [PubMed]
  148. Li, W.; Miller, D.; Liu, X.; Tosi, L.; Chkaiban, L.; Mei, H.; Hung, P.-H.; Parekkadan, B.; Sherlock, G.; Levy, S.F. Arrayed in vivo barcoding for multiplexed sequence verification of plasmid DNA and demultiplexing of pooled libraries. Nucleic Acids Res. 2024, 52, e47. [Google Scholar] [CrossRef]
  149. Tripathi, S.; Voogdt, C.G.P.; Bassler, S.O.; Anderson, M.; Huang, P.H.; Sakenova, N.; Capraz, T.; Jain, S.; Koumoutsi, A.; Bravo, A.M.; et al. Randomly barcoded transposon mutant libraries for gut commensals I: Strategies for efficient library construction. Cell Rep. 2024, 43, 113517. [Google Scholar] [CrossRef]
  150. Piffanelli, P.; Droc, G.; Mieulet, D.; Lanau, N.; Bès, M.; Bourgeois, E.; Rouvière, C.; Gavory, F.; Cruaud, C.; Ghesquière, A.; et al. Large-scale characterization of Tos17 insertion sites in a rice T-DNA mutant library. Plant Mol. Biol. 2007, 65, 587–601. [Google Scholar] [CrossRef] [PubMed]
  151. Uzbas, F.; Opperer, F.; Sönmezer, C.; Shaposhnikov, D.; Sass, S.; Krendl, C.; Angerer, P.; Theis, F.J.; Mueller, N.S.; Drukker, M. BART-Seq: Cost-effective massively parallelized targeted sequencing for genomics, transcriptomics, and single-cell analysis. Genome Biol. 2019, 20, 155. [Google Scholar] [CrossRef]
  152. Chen, W.; Wang, P.; Wang, L.; Zhang, D.; Han, M.; Han, M.; Song, L. Low-complexity and highly robust barcodes for error-rich single molecular sequencing. 3 Biotech 2021, 11, 78. [Google Scholar] [CrossRef]
  153. Srivathsan, A.; Meier, R. Scalable, Cost-Effective, and Decentralized DNA Barcoding with Oxford Nanopore Sequencing. Methods Mol. Biol. 2024, 2744, 223–238. [Google Scholar]
  154. Duan, L.; Huang, C.; Chen, G.; Xiong, L.; Liu, Q.; Yang, W. Determination of rice panicle numbers during heading by multi-angle imaging. Crop J. 2015, 3, 211–219. [Google Scholar] [CrossRef]
  155. Fahlgren, N.; Gehan, M.A.; Baxter, I. Lights, camera, action: High-throughput plant phenotyping is ready for a close-up. Curr. Opin. Plant Biol. 2015, 24, 93–99. [Google Scholar] [CrossRef]
  156. Pratap, A.; Gupta, S.; Nair, R.M.; Gupta, S.; Schafleitner, R.; Basu, P.; Singh, C.M.; Prajapati, U.; Gupta, A.K.; Nayyar, H. Using plant phenomics to exploit the gains of genomics. Agronomy 2019, 9, 126. [Google Scholar] [CrossRef]
  157. Song, P.; Wang, J.; Guo, X.; Yang, W.; Zhao, C. High-throughput phenotyping: Breaking through the bottleneck in future crop breeding. Crop J. 2021, 9, 633–645. [Google Scholar] [CrossRef]
  158. Jangra, S.; Chaudhary, V.; Yadav, R.C.; Yadav, N.R. High-throughput phenotyping: A platform to accelerate crop improvement. Phenomics 2021, 1, 31–53. [Google Scholar] [CrossRef] [PubMed]
  159. Yang, W.; Guo, Z.; Huang, C.; Duan, L.; Chen, G.; Jiang, N.; Fang, W.; Feng, H.; Xie, W.; Lian, X. Combining high-throughput phenotyping and genome-wide association studies to reveal natural genetic variation in rice. Nat. Commun. 2014, 5, 5087. [Google Scholar] [CrossRef]
  160. Qin, Z.; Zhang, Z.; Hua, X.; Yang, W.; Liang, X.; Zhai, R.; Huang, C. Cereal grain 3D point cloud analysis method for shape extraction and filled/unfilled grain identification based on structured light imaging. Sci. Rep. 2022, 12, 3145. [Google Scholar] [CrossRef]
  161. Sun, M.; Huang, S.; Lu, Z.; Wang, M.; Zhang, S.; Yang, K.; Tang, B.; Yang, W.; Huang, C. A novel method for intelligent analysis of rice yield traits based on LED transmission imaging and cloud computing. Measurement 2023, 217, 113017. [Google Scholar] [CrossRef]
  162. Guo, Z.; Yang, W.; Chang, Y.; Ma, X.; Tu, H.; Xiong, F.; Jiang, N.; Feng, H.; Huang, C.; Yang, P. Genome-wide association studies of image traits reveal genetic architecture of drought resistance in rice. Mol. Plant 2018, 11, 789–805. [Google Scholar] [CrossRef] [PubMed]
  163. Ricachenevsky, F.K.; Punshon, T.; Lee, S.; Oliveira, B.H.N.; Trenz, T.S.; Maraschin, F.d.S.; Hindt, M.N.; Danku, J.; Salt, D.E.; Fett, J. Elemental profiling of rice FOX lines leads to characterization of a new Zn plasma membrane transporter, OsZIP7. Front. Plant Sci. 2018, 9, 865. [Google Scholar] [CrossRef]
  164. Wairich, A.; Vitali, A.; Adamski, J.M.; Lopes, K.L.; Duarte, G.L.; Ponte, L.R.; Costa, H.K.; Menguer, P.K.; Santos, R.P.d.; Fett, J.P.J. Enhanced expression of OsNAC5 leads to up-regulation of OsNAC6 and changes rice (Oryza sativa L.) ionome. Genet. Mol. Biol. 2022, 46 (Suppl. S1), e20220190. [Google Scholar] [CrossRef]
  165. Cao, Z.Z.; Lin, X.Y.; Yang, Y.J.; Guan, M.Y.; Xu, P.; Chen, M.X. Gene identification and transcriptome analysis of low cadmium accumulation rice mutant (lcd1) in response to cadmium stress using MutMap and RNA-seq. BMC Plant Biol. 2019, 19, 250. [Google Scholar] [CrossRef]
  166. Ishikawa, S.; Ae, N.; Yano, M. Chromosomal regions with quantitative trait loci controlling cadmium concentration in brown rice (Oryza sativa). New Phytol. 2005, 168, 345–350. [Google Scholar] [CrossRef] [PubMed]
  167. Guo, S.; Xu, Y.; Liu, H.; Mao, Z.; Zhang, C.; Ma, Y.; Zhang, Q.; Meng, Z.; Chong, K. The interaction between OsMADS57 and OsTB1 modulates rice tillering via DWARF14. Nat. Commun. 2013, 4, 1566. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Insertional mutagenesis approaches to develop loss-of-function mutants. This figure illustrates three distinct approaches for developing loss-of-function mutants. (a) The T-DNA insertion method utilizes Agrobacterium tumefaciens harboring the binary vector to genetically transform rice, leading to the integration of T-DNA into the genome and the disruption of gene functions. (b) The Ac/Ds mutant lines are generated by crossing independently transformed Ac and Ds lines, which facilitates the disruption of gene functions in the subsequent generations. (c) The Tos17 insertion method involves tissue or callus culture activation to integrate Tos17 into the genome, resulting in the disruption of gene functions.
Figure 1. Insertional mutagenesis approaches to develop loss-of-function mutants. This figure illustrates three distinct approaches for developing loss-of-function mutants. (a) The T-DNA insertion method utilizes Agrobacterium tumefaciens harboring the binary vector to genetically transform rice, leading to the integration of T-DNA into the genome and the disruption of gene functions. (b) The Ac/Ds mutant lines are generated by crossing independently transformed Ac and Ds lines, which facilitates the disruption of gene functions in the subsequent generations. (c) The Tos17 insertion method involves tissue or callus culture activation to integrate Tos17 into the genome, resulting in the disruption of gene functions.
Plants 14 01492 g001
Figure 2. miRNA/siRNA-mediated RNA interference and CRISPR/Cas9-mediated mutagenesis approaches. This figure describes two advanced approaches for knockdown and loss-of-function mutagenesis. (a) The miRNA/siRNA mediated RNA silencing method involves genetic transformation using Agrobacterium that harbors pools of miRNA/siRNA constructs, enabling the generation of large-scale knockdown-mutant lines. (b) The CRISPR/Cas9-mediated mutagenesis approach utilizes genetic transformation with Agrobacterium containing pools of sgRNA constructs, facilitating the creation of large-scale knockout-mutant lines.
Figure 2. miRNA/siRNA-mediated RNA interference and CRISPR/Cas9-mediated mutagenesis approaches. This figure describes two advanced approaches for knockdown and loss-of-function mutagenesis. (a) The miRNA/siRNA mediated RNA silencing method involves genetic transformation using Agrobacterium that harbors pools of miRNA/siRNA constructs, enabling the generation of large-scale knockdown-mutant lines. (b) The CRISPR/Cas9-mediated mutagenesis approach utilizes genetic transformation with Agrobacterium containing pools of sgRNA constructs, facilitating the creation of large-scale knockout-mutant lines.
Plants 14 01492 g002
Figure 3. Gain-of-function mutagenesis approaches. This figure illustrates two key gain-of-function mutagenesis approaches. (a) Activation tagging: this approach involves the insertion of a strong enhancer construct, using Agrobacterium tumefaciens transformation, near a gene of interest, thereby enhancing its expression and resulting in the gain of function. BAR, glufosinate resistance; LB, left border; RB, right border. (b) FOX hunting. In this approach, full-length cDNA is cloned between a strong promoter and terminator and then transformed using Agrobacterium tumefaciens to drive the overexpression of specific genes. PUbi-1, ubiquitin promoter; Ter, terminator.
Figure 3. Gain-of-function mutagenesis approaches. This figure illustrates two key gain-of-function mutagenesis approaches. (a) Activation tagging: this approach involves the insertion of a strong enhancer construct, using Agrobacterium tumefaciens transformation, near a gene of interest, thereby enhancing its expression and resulting in the gain of function. BAR, glufosinate resistance; LB, left border; RB, right border. (b) FOX hunting. In this approach, full-length cDNA is cloned between a strong promoter and terminator and then transformed using Agrobacterium tumefaciens to drive the overexpression of specific genes. PUbi-1, ubiquitin promoter; Ter, terminator.
Plants 14 01492 g003
Figure 4. This figure illustrates a simplified dual-index barcoding approach, facilitating the simultaneous pooling of various sample types, including cDNA libraries for FOX hunting mutagenesis, gRNA for CRISPR/Cas9-mediated mutagenesis, and genomic DNA for identifying transposon or T-DNA insertion sites. This innovative design enhances the efficiency of next-generation sequencing (NGS) by allowing for the multiplexing of samples, thereby reducing costs and improving the throughput in genotyping. The color is to indicate the unique combinatorial barcoding method.
Figure 4. This figure illustrates a simplified dual-index barcoding approach, facilitating the simultaneous pooling of various sample types, including cDNA libraries for FOX hunting mutagenesis, gRNA for CRISPR/Cas9-mediated mutagenesis, and genomic DNA for identifying transposon or T-DNA insertion sites. This innovative design enhances the efficiency of next-generation sequencing (NGS) by allowing for the multiplexing of samples, thereby reducing costs and improving the throughput in genotyping. The color is to indicate the unique combinatorial barcoding method.
Plants 14 01492 g004
Table 1. Comparative analysis of mutagenesis approaches for large-scale mutant analysis.
Table 1. Comparative analysis of mutagenesis approaches for large-scale mutant analysis.
Mutagenesis
Tools
Mutation TypeAdvantagesChallengesPrecisionScalabilityReferences
Chemical (e.g., EMS)RandomSimple non-transgenic with high mutagenesis efficiencyRequires extensive screeningLowLow[73]
T-DNA InsertionRandomGenome-wide coverage; barcode-compatible.
Thousands of mutants analyzed in parallel
Random insertion disrupts non-target genes.Individual mutant screening via PCR or phenotypingLowHigh[24,28]
Transposons (e.g., Ac/Ds)RandomMobility enables regional mutagenesis.Reusable systemsRequires transposase control;
lower efficiency
Individual mutant screening via PCR and phenotyping
ModerateModerate[74]
CRISPR/Cas9Targeted knockouts/editingHigh precision; multiplex editing of redundant genesComplex design for large librariesHighHigh[64,75,76]
RNAiTargeted knockdownRapid; scalable with barcoded vectors (e.g., pooled RNAi screens) for redundant genesRequires efficient design for target silencingHighModerate[41,64,77]
Activation taggingGain-of-functionIdentifies dominant allelesrandom activation may cause pleiotropy
Low mutant frequency
LowModerate[35,78]
FOX huntingcDNA overexpressionPrecise overexpression.
Possible to use barcoding to trace back the cDNA clones; links phenotype to known genes
High cost of cDNA library construction.
Complex phenotyping
HighHigh[79,80]
Table 2. Important features of DNA barcode-based large-scale mutagenesis approaches to accelerate functional genomics.
Table 2. Important features of DNA barcode-based large-scale mutagenesis approaches to accelerate functional genomics.
Approaches for Integration of DNA BarcodingReferences
FOX huntingPooled cDNA clone libraries or mutants with unique barcodes can be synthesized using amplification primers or sequencing primers[148]
Activation taggingIntegrates unique barcodes into the T-DNA construct[24]
T-DNAEmbeds unique barcodes into the mutagenic T-DNA construct[24]
Transposon (Ac/Ds)Ds transposons tagged with barcodes for tracking reinsertion sites[24,149]
Retrotransposon (Tos17)Unique barcode embedded with long terminal repeat (LTR) for tracking insertion sites[150]
RNAiMultiplexed shRNA or amiRNA libraries with unique barcodes[41,77]
CRISPR/Cas9Multiplexed sgRNA libraries with unique barcodes[75,76,82]
Important features of DNA barcoding
Tracking and screeningScalable: sequencing identifies barcodes linked to samples[141,151]
ScalabilityThousands of mutants can be analyzed simultaneously[24,41,70,75,76,77,82]
PrecisionHigh: pooled libraries enable genome-wide screening (e.g., CRISPR-Cas9 with barcoded sgRNAs; and shRNA or amiRNA libraries with unique barcodes)[41,75,76,77,144]
ApplicationsLarge-scale functional genomics, pooled screens[24,41,70,75,76,77,82]
ChallengesRequires careful design and integration of DNA barcode and next-generation sequencing data analysis[136,152,153]
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

Wolella, E.K.; Cheng, Z.; Li, M.; Xia, D.; Zhang, J.; Duan, L.; Liu, L.; Li, Z.; Zhang, J. Large-Scale Rice Mutant Establishment and High-Throughput Mutant Manipulation Help Advance Rice Functional Genomics. Plants 2025, 14, 1492. https://doi.org/10.3390/plants14101492

AMA Style

Wolella EK, Cheng Z, Li M, Xia D, Zhang J, Duan L, Liu L, Li Z, Zhang J. Large-Scale Rice Mutant Establishment and High-Throughput Mutant Manipulation Help Advance Rice Functional Genomics. Plants. 2025; 14(10):1492. https://doi.org/10.3390/plants14101492

Chicago/Turabian Style

Wolella, Eyob Kassaye, Zhen Cheng, Mengyuan Li, Dandan Xia, Jianwei Zhang, Liu Duan, Li Liu, Zhiyong Li, and Jian Zhang. 2025. "Large-Scale Rice Mutant Establishment and High-Throughput Mutant Manipulation Help Advance Rice Functional Genomics" Plants 14, no. 10: 1492. https://doi.org/10.3390/plants14101492

APA Style

Wolella, E. K., Cheng, Z., Li, M., Xia, D., Zhang, J., Duan, L., Liu, L., Li, Z., & Zhang, J. (2025). Large-Scale Rice Mutant Establishment and High-Throughput Mutant Manipulation Help Advance Rice Functional Genomics. Plants, 14(10), 1492. https://doi.org/10.3390/plants14101492

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