- Review
- Open access
- Published:
Advances in genomic tools for plant breeding: harnessing DNA molecular markers, genomic selection, and genome editing
Biological Research volume 57, Article number: 80 (2024)
Abstract
Conventional pre-genomics breeding methodologies have significantly improved crop yields since the mid-twentieth century. Genomics provides breeders with advanced tools for whole-genome study, enabling a direct genotype–phenotype analysis. This shift has led to precise and efficient crop development through genomics-based approaches, including molecular markers, genomic selection, and genome editing. Molecular markers, such as SNPs, are crucial for identifying genomic regions linked to important traits, enhancing breeding accuracy and efficiency. Genomic resources viz. genetic markers, reference genomes, sequence and protein databases, transcriptomes, and gene expression profiles, are vital in plant breeding and aid in the identification of key traits, understanding genetic diversity, assist in genomic mapping, support marker-assisted selection and speeding up breeding programs. Advanced techniques like CRISPR/Cas9 allow precise gene modification, accelerating breeding processes. Key techniques like Genome-Wide Association study (GWAS), Marker-Assisted Selection (MAS), and Genomic Selection (GS) enable precise trait selection and prediction of breeding outcomes, improving crop yield, disease resistance, and stress tolerance. These tools are handy for complex traits influenced by multiple genes and environmental factors. This paper explores new genomic technologies like molecular markers, genomic selection, and genome editing for plant breeding showcasing their impact on developing new plant varieties.
Background
Since plant domestication around 10,000 years ago, plant breeding has successfully developed crops and varieties essential to modern society, consistently defying Malthusian predictions [44]. Traditional pre-genomics breeding methods have resulted in modern cultivars, significantly increasing the yields of major crops since the mid-twentieth century. Today, genomics offers breeders advanced tools and techniques for whole-genome analysis, representing a significant shift by enabling direct examination of the genotype and its connection to the phenotype [166, 167]. This new era of crop development leverages genomics-based approaches, such as molecular markers, genomic selection, and genome editing tools, for precise and efficient improvements [166, 167]. Gene position or genomic regions that regulate important traits in plants are discovered using molecular markers. Markers are typically categorized into two main groups: classical and molecular markers. The limitations associated with phenotype-based markers prompted the development of direct DNA-based markers, often known as molecular markers, which exhibit greater versatility. Classical markers encompass morphological, cytological, and biochemical markers, while DNA markers include a variety of types such as Restriction Fragment Length Polymorphism (RFLP), Random Amplified Polymorphic DNA (RAPD), Amplified Fragment Length Polymorphism (AFLP), Simple Sequence Repeats (SSRs), Single Nucleotide Polymorphism (SNP) markers etc. [60]. In modern plant breeding, SNPs are extensively utilized as DNA markers to pinpoint genomic regions associated with key traits, thereby accelerating the breeding process. Recognized as the most prevalent variations within plant genomes, SNPs are invaluable for high-resolution genotyping, offering the highest map precision.
Moreover, SNPs are both more efficient and cost-effective compared to other markers. Their popularity surged in the twenty-first century, largely due to genotyping by sequencing (GBS) technique advancements. Some other novel marker techniques, such as Intron Length Polymorphism (ILP), Diversity Array Technology (DArT), Penta-Primer Amplification Refractory Mutation System (PARMS), Inter small RNA polymorphism (iSNAP), etc., have been employed in plant breeding, which has enabled precise selection of desirable traits in plants, genetic diversity analysis, and accelerated breeding (Amiteye, 2021). They pinpoint the precise genetic differences connected to desirable qualities, making it possible to pick individuals with the best genomic profiles for breeding with accuracy and efficiency. Recent advances in genomics are producing new plant breeding methodologies and ways (e.g., association mapping, marker-assisted selection, genomic selection, genome editing, etc.).
Genomic resources for plant breeding encompass genetic markers, reference genomes, genomic and protein databases, transcriptomes, and gene expression profiles. These resources facilitate the identification of genes associated with desirable traits, understanding genetic diversity, and acceleration of breeding programs [112]. Key techniques include genome-wide association studies (GWAS), marker-assisted selection (MAS), and genomic selection (GS), which allow for precise trait selection and prediction of breeding outcomes. By integrating these genomic tools, plant breeders can improve crop yield, disease resistance, and stress tolerance, enhancing agricultural productivity and sustainability. The existing variability among crop species is utilized for plant breeding activities, which can also be generated through crossing or induced mutagenesis. In addition to the identification of genetic markers and the availability of published genomes, clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR/Cas9) is promising for application to modern breeding and is a novel technology for genome editing in major crops [132]. CRISPR/Cas9-based directional breeding is highly efficient and saves more time than other breeding techniques that use genome editing. It further enables targeted genetic modifications, opening new avenues for crop improvement.Genomics approaches are beneficial when dealing with complex traits, as these traits usually have a multi-genic nature and a significant environmental influence [102]. Genomic tools provide genomic information and facilitate the detection of QTLs and the identification of existing favorable alleles of small effect, which have frequently remained unnoticed and have not been included in the gene pool used for breeding [102]. Genomic tools have revolutionized plant breeding by enabling more precise, efficient, and targeted approaches to developing new plant varieties through genome-wide association study, marker-assisted selection, genomic selection, and gene editing. In this review, we present and discuss the most relevant advances in the development of genomic tools and provide examples of applying these tools to plant breeding.
Molecular markers: tool for the genetic analysis
Based on nucleotide sequence polymorphisms, molecular markers include insertions, deletions, point mutations, duplications, and translocations. They are ideal when codominant, evenly distributed, highly reproducible, and detect significant polymorphism. The first molecular marker technique, RFLP, was introduced by Botstein et al. [15]. RFLP, RAPD, AFLP, and Isozyme markers are first-generation molecular markers that have been developed and used in genetic analysis and plant breeding (Table 1). Advancements in molecular markers have significantly enhanced their efficiency, resolution, and application scope in genetic analysis and plant breeding (Table 2) [156]. These advancements can be broadly categorized into the development of new types of markers, improvements in marker technologies, and the integration of molecular markers with other genomic tools. Here are some key advancements:
SSRs or microsatellites
Microsatellites, alternatively known as short tandem repeats (STRs) or simple sequence repeats (SSRs), are short DNA sequences with lengths typically ranging from one to six base pairs in contrast to minisatellites (VNTRs), which feature longer repeat sequences spanning from 11 to 60 base pairs [115]. Microsatellites are found throughout the genome, including in chloroplasts and mitochondria [125, 127]. Due to the different numbers of repeats present in these locations, SSRs exhibit high polymorphism that is simple to detect using polymerase chain reaction (PCR). Mismatches, recombination, mobile element transfer (retrotransposons), and DNA strand slippage are some of the mechanisms that contribute to the occurrence of SSRs. Common SSR motifs encompass mononucleotide (A, T), dinucleotide (AT, GA), trinucleotide (AGG), and tetranucleotide (AAAC) repeats. The creation of primers often uses flanking sequences that are conserved around SSRs. Developing SSR markers involves creating an SSR library, identifying specific microsatellites, designing primers in favorable regions, and conducting PCR. Banding patterns are then interpreted and evaluated for polymorphism. SSR markers are highly favored due to their codominant inheritance, abundance, allelic diversity, and ease of assessment via PCR with flanking primers. McCouch et al. [107] conducted a pivotal study on Simple Sequence Repeat (SSR) markers in rice, focusing on developing and mapping these markers to enhance genetic research. They identified numerous SSR loci across rice chromosomes and designed primers for their use. This comprehensive set of SSR markers has become instrumental in rice genetic studies, including linkage mapping, trait association, and breeding. SSR markers, are used in plant genetics for various applications. For example, in rice, SSR markers have been used to map QTLs related to drought resistance and yield enhancement [65]. In wheat, SSR markers help identify genetic diversity and select for traits like disease resistance and stress tolerance [174, 175]. A 2023 study on Brassica napus utilized 304 SSR markers to evaluate genetic diversity, uncovering a 76% polymorphism rate and pinpointing loci associated with oil content and disease resistance [173]. The research emphasized SSR markers’ effectiveness in detecting allelic variation and mapping important agricultural traits. Additionally, SSR markers showed cross-species transferability, proving valuable for identifying traits beneficial for breeding programs and conserving genetic diversity. SSR markers are crucial for crop improvement and understanding genetic variation [83].
Inter small RNA polymorphism (iSNAP)
Endogenous noncoding small RNAs, typically 20–24 nucleotides long and have important regulatory roles, are widely distributed in eukaryotic genomes [54]. These small RNAs offer a valuable resource for molecular marker development due to their conserved flanking sequences, enabling primer design for PCR-based fingerprinting. The Inter small RNA polymorphism (iSNAP) technique, pioneered by Gui et al. [54], capitalizes on this characteristic. To detect length polymorphisms brought on by insertions and deletions (InDels) within the small RNA pool, primer pairs flanking small RNAs are used to start PCR reactions. It is a noncoding, sequence-based marker system and is suitable for genotyping and genome mapping [3]. Unlike traditional markers that focus on coding sequences or microsatellites, iSNAP markers explore variations in non-coding regulatory regions. This opens up new possibilities for studying gene expression and complex traits influenced by small RNA pathways, including plant stress responses, development, and epigenetic regulation. iSNAP markers have functional relevance, as they are closely associated with gene regulatory mechanisms. This makes them particularly useful for marker-assisted selection (MAS), enabling the identification of traits governed by post-transcriptional gene regulation, such as disease resistance and stress tolerance. A recent case study by Zhang et al. [196] illustrates the application of iSNAP markers in identifying disease-resistant genes in tomatoes (Solanum lycopersicum). In this study, researchers focused on identifying polymorphisms in intergenic regions flanked by microRNAs (miRNAs) associated with defense responses. They developed a set of iSNAP markers and used them to screen tomato varieties for resistance to Phytophthora infestans, the pathogen responsible for late blight disease. In maize, iSNAP markers have been used to identify genetic loci associated with disease resistance and yield traits, enabling more efficient breeding [97, 98]. In wheat, they assist in mapping quantitative trait loci (QTLs) for drought tolerance, enhancing the development of resilient varieties [26].
Intron length polymorphism (ILP)
In eukaryotic genomes, introns are prevalent and found throughout various gene components. Due to their lower selective pressure, Introns exhibit greater variability compared to coding sequences, rendering them valuable as highly polymorphic genetic markers. Recently, researchers have focused on annotating and leveraging gene introns to create intron-length polymorphism (ILP) markers on a genome-wide scale. An Intron Length Polymorphism marker (ILP marker) is a genetic marker used in plant and animal breeding to identify variations in the length of introns—non-coding regions of a gene that are transcribed but not translated into proteins. These markers exploit natural differences in intron lengths among individuals or populations, aiding in genetic mapping, diversity studies, and breeding programs by distinguishing between different genotypes or assessing genetic diversity [7]. These markers have proven invaluable for large-scale genotyping in major food crop plants, such as rice [7, 170], wheat [147], and maize [94]. PCR, a widely used technique, conveniently detects ILP markers. The amplification of introns via PCR involves designing primers in flanking exons, a method known as exon-primed intron-crossing PCR (EPIC-PCR). Notably, exon sequences tend to be more evolutionarily conserved, enhancing the versatility of primers designed within exons compared to noncoding sequences. ILP markers are particularly advantageous when they target multiple insertions and deletions (InDels) within a single intron during amplification. This strategy significantly increases the likelihood of identifying genetic polymorphism. ILP markers are highly transferable across related species, allowing for comparative genomics and evolutionary studies [27]. For example, in cereals such as rice, maize, and wheat, ILPs have shown consistent results, making them invaluable for research across different species without needing species-specific markers. Liu et al. [99] developed intron length polymorphism (ILP) markers for plants, identifying 1507 ILP markers in Oryza sativa (rice). These markers were highly transferable across species and showed polymorphism rates of 85.3%. ILP markers proved useful for genetic diversity analysis and breeding applications in various crops. A recent study by Chen et al. [27] demonstrated the effectiveness of ILP markers in mapping drought tolerance in rice (Oryza sativa). The researchers used ILP markers derived from conserved intron regions of the Dehydration-Responsive Element-Binding (DREB) gene family. These markers were employed to screen a diverse set of rice germplasm, leading to the identification of several drought-tolerant varieties. The study showcased how ILP markers could be used to identify quantitative trait loci (QTLs) associated with drought tolerance. The QTLs identified were then validated across multiple rice populations, proving the reliability of ILPs in marker-assisted selection (MAS) for complex traits like drought resistance. These developments underscore the importance of introns and their applications in molecular genetics, enabling more effective crop breeding and resource management.
Single nucleotide polymorphism (SNP)
Single Nucleotide Polymorphisms are single nucleotide differences seen in the genomic sequences of individuals within a population. These are the most prevalent molecular markers, and their distribution varies between species. Contrastingly, while humans exhibit an average of one Single Nucleotide Polymorphism (SNP) per every 1000 base pairs [137], rice displays a higher frequency, with approximately one SNP occurring within every 130–140 base pairs [51]. SNPs are frequently discovered in noncoding areas. According to Sunyaev et al. [158], SNPs in coding areas can be synonymous or nonsynonymous, changing phenotypic features and amino acid composition. SNPs are the smallest units of genetic variation, providing a simple and abundant source of markers crucial for genetic mapping, marker-assisted breeding, and map-based cloning. [188]. Significant techniques for SNP genotyping include primer extension, invasive cleavage, oligonucleotide ligation, and allele-specific hybridization [154]. SNP markers discovered by two methods, including SNP discovery from PCR and SNP discovery from High Throughput-Next generation sequencing (NGS) – RNA-Seq, RAD-Seq, Genotyping by Sequencing (Fig. 1), WGS (Whole-Genome Sequencing), WGR (Whole-Genome Regression), etc.
Next-Generation Sequencing (NGS) is a high-throughput DNA sequencing technology that enables rapid, parallel sequencing of millions of DNA fragments for comprehensive genomic analysis. In recent years, NGS technologies have identified thousands to millions of SNPs in various crops, facilitating genetic diversity studies, trait mapping, and breeding improvements. Numerous tools are available for SNP discovery, including BioEdit, DNASTAR Lasergene Genomics Suite, SAMtools, SOAPsnp, Stacks, Ddocent, PyRAD, and GATK. Typically, biallelic SNPs are straightforward to assay. SNP is detected when a nucleotide from an accession read differs from the reference genome at the corresponding position. Without a reference genome, this comparison is made by examining reads from different genotypes using de novo assembly methods. SNP calling is performed using read assembly files generated by mapping programs. Various empirical and statistical criteria, such as read depth, quality scores, and consensus base ratios, are employed in the SNP calling process. SNP discovery is more effective when multiple and diverse genotypes are analyzed simultaneously, as this approach captures the genetic variability within a species. There are three main types of SNP genotyping platforms: single SNP genotyping (using PCR with Taqman from Life Technologies or KASP genotyping from LGC Genomics), multiple SNP genotyping (using SNP chips from Illumina and multiplexing from Sequenom), and SNP genotyping by next-generation sequencing methods such as Genotyping by Sequencing (GBS) and Restriction site Associated DNA sequencing (RADSeq). For large-scale genotyping, high-throughput methods such as Genotyping by Sequencing (GBS), Restriction site- associated DNA sequencing (RADSeq), and allele-specific PCR are used [37]. These technologies have been extensively used to discover and genotype SNPs in food crops, including cereals [barley [31, 48, 134, 162], rice [28, 185], and wheat [20]], oil crops [oilseed rape [29] and sunflower [100]], horticultural crops [cowpea [30], potato [59], tomato [149]], soybean [155], and among others. SNP markers are crucial in genetics and agriculture, aiding genetic mapping, identifying disease associations, and improving crops through selective breeding. In their 2011 study, Kump et al. used SNPs to pinpoint disease resistance genes in maize, specifically targeting Southern Leaf Blight (SLB), caused by Cochliobolus heterostrophus. They identified 32 quantitative trait loci (QTLs) significantly associated with SLB resistance. The identified SNPs and associated QTLs can be used in marker-assisted selection (MAS) and genomic selection (GS) programs to develop SLB-resistant maize varieties. SNPs also play a pivotal role in integrating multi-omics data for crop improvement by acting as key genetic markers that link DNA variations to other molecular levels, such as gene expression (transcriptomics), protein abundance (proteomics), and metabolite profiles (metabolomics). These connections help to unravel the complex genetic architecture of important agronomic traits, including yield, stress tolerance, and disease resistance. By mapping SNPs to different omics layers, researchers can identify critical genes, pathways, and molecular interactions responsible for these traits [90]. This comprehensive approach enhances breeding accuracy, enabling the development of superior crop varieties with enhanced performance through more informed and precise selection methods.
Diversity array technology (DArT)
The DArT sequencing technique is a highly reproducible microarray-based method for discovering polymorphic markers [177]. DArT is a genomic analysis method designed to enhance the detection of SNPs (Single Nucleotide Polymorphisms) across the genome, particularly insertions and deletions. It begins with creating a purposefully randomized fragment library, which serves as a genomic representation. DArT libraries are tailored for specific research purposes, utilizing suitable individuals, whether individual or pooled samples. The subsequent steps involve identifying the genetic representations, hybridizing them onto the chips, and printing the genomic library onto microarray chips. DArT simplifies the genome by initially subjecting it to restriction digestion and then hybridizing the resulting DNA fragments onto microarray chips. Data analysis is done after scanning. This method allows thousands of genomic loci to be simultaneously genotyped in a single reaction test, requiring as little as 50–100 ng of genomic DNA. Once markers are identified, the need for specific assays for genotyping is eliminated, except for consolidating polymorphic markers into an array for a particular genotype. These genotyping arrays are equipped with these polymorphic markers and are commonly employed in genotyping tasks [66]. DArT markers are primarily dominant and require specialized software, laboratory facilities, a substantial investment, and skilled personnel (Sinha et al., 2023).
Penta-primer amplification refractory mutation system (PARMS)
The Penta-Primer Amplification Refractory Mutation System (PARMS) is a specialized genotyping technique used for identifying specific single nucleotide polymorphisms (SNPs) or mutations in DNA sequences. It is particularly useful in plant and animal genetics, as well as in medical research, for detecting alleles associated with certain traits or diseases. It is an extension of the traditional Amplification Refractory Mutation System (ARMS), which relies on the specificity of DNA primers to distinguish between different alleles at a given genetic locus [159]. PARMS involves the use of five primers to achieve high specificity and efficiency in identifying specific alleles. Two universal primers bind to conserved regions of DNA surrounding the SNP or mutation of interest. Two allele-specific forward primers and a reverse shared primer are designed to match perfectly with either the wild-type allele, mutant allele, or a third variant allele, with mismatches at critical positions near the SNP or mutation. This allows for selective amplification of only the specific alleles in question. It employs competitive allele-specific polymerase chain reaction (AS-PCR) and a fluorescence-based reporting system to detect genetic variations, specifically single-nucleotide polymorphisms (SNPs) [159]. It can efficiently handle different numbers of SNPs and samples to be analyzed. The process requires only standard liquid handling, thermal cycling instruments, and plate reading instruments.
Furthermore, it is compatible with DNA samples from various sources and extraction methods, including alkaline lysis. This makes it ideal for a direct PCR-based SNP marker-assisted selection system (D-MAS), known for its simplicity, cost-effectiveness, and labor efficiency in SNP genotyping. In a practical application, Gao et al. [49] developed a PARMS marker for the TAC1 gene, illustrating its usefulness in rice plant architecture breeding. Having outlined the significance of molecular markers, we now delve into the applications of genomic resources in crop improvement.
Genomic resources for plant breeding
The availability of whole genome sequences is invaluable for plant breeding. Arabidopsis (125 Mb) and rice (466 Mb) were early models for plant genetics due to their small genomes among dicots and monocots. Their genome sequences, announced in 2000 and 2005, have been pivotal in understanding key genes and biological functions. The advent of next-generation sequencing (NGS) technologies has revolutionized genomics. Among these, the 454 (Roche) and Illumina platforms are widely used for crop sequencing. NGS technologies have significantly increased sequencing capacity; for instance, the Illumina HiSeq 2000 can generate 55 Gb per day, far exceeding the human genome size. The development of third-generation sequencing platforms like PacBio RS (Pacific Biosciences, https://www.pacb.com/), Helicos (Helicos, https://seqll.com/), and Ion Torrent has further advanced the field. These platforms enable the production of long reads, resulting in more accurate and contiguous genome assemblies. Third-generation sequencing is particularly effective for assembling genomes de novo, especially in regions with highly repetitive sequences and clarifying structural variants.
Additionally, isoform sequencing from these platforms facilitates detailed studies of exons, splice sites, and alternatively spliced regions, improving genome annotation. NGS-generated sequences are typically deposited in the NCBI Sequence Read Archive, making them accessible for further research. The emergence of third-generation sequencing has enabled the generation of long reads and allowed the production of more accurate and contiguous genome assemblies [25]. Third-generation sequencing enhances the creation of high-quality whole genome de novo assemblies by providing long reads that cover complex regions with highly repetitive sequences. This technology also elucidates other complex repeat sequences and structural variants. Third-generation sequencing techniques, such as isoform sequencing, produce full-length transcripts, enabling detailed analysis of exons, splice sites, and alternatively spliced regions, which aids in refining genome annotations. Sequences generated through NGS are typically archived in the NCBI Sequence Read Archive (https://www.ncbi.nlm.nih.gov/sra) for public access.
Two standard analyses performed on NGS reads are genome assembly and mapping. Assemblers like Roche's 454 Gsassembler, Celera Assembler, and Mira are frequently used for genome assembly. Once a reference genome is available, variation studies are typically conducted using mapping software such as Bowtie, BWA, and TopHat, which align reads to the reference genome. SNPs can then be detected with tools like SAMtools or GigaBayes. The algorithms for processing raw genomic data vary based on the data type and desired results. Bioinformaticians must present their findings to breeders via user-friendly interfaces, often through easily navigable websites. General-purpose web databases like GenBank (http://www.ncbi.nlm.nih.gov/genbank/), EMBL (http://www.ebi.ac.uk/embl/), DDBJ (http://www.ddbj.nig.ac.jp/), UniProt (http://www.uniprot.org), and Swiss-Prot (http://expasy.org/sprot/) provide researchers and breeders with essential biological information. Genomic sequence databases GenBank, EMBL, DDBJ, Ensembl, UCSC Genome Browser, and dbSNP offer extensive genomic data, analysis tools, and resources. Protein function databases, integrating sequence data, structural information, and functional annotations, include UniProt, Swiss-Prot, Gene Ontology, Protein Data Bank, InterPro, KEGG, Pfam, STRING, BioGRID, and PhosphoSitePlus (Table 3). Additionally, specialized databases for specific species useful to breeders, such as SGN, Phytozome, Gramene, and CropNet, provide targeted information for breeding programs. These resources collectively support the plant breeding process by enabling detailed genetic and protein analyses, aiding in developing improved crop varieties.
New genomic tools are crucial for advancing and speeding up gene expression studies. Gene expression analysis provides breeders with valuable biological insights, helping them understand the molecular basis of complex plant processes and identify new targets for manipulation. While QRTPCR is an affordable, quantitative technique, it can only analyze a limited number of genes per experiment. Other methods, such as differential display and cDNA-AFLPs, allow the study of thousands of genes but lack quantitative precision and struggle with low-abundance transcripts (M Perez-de-Castro et al. 2012). More advanced techniques like serial analysis of gene expression (SAGE) and massively parallel signature sequencing (MPSS) address some of these limitations. However, the most popular methods today for transcript profiling are hybridization-based platforms or microarrays. Expression arrays offer several advantages, including measuring tens of thousands of transcripts simultaneously, semi-quantitative results, and sensitivity to low-abundance transcripts. Several web resources facilitate microarray data analysis, such as Babelomics (http://babelomics.bioinfo.cipf.es/), and software packages like Bioconductor (http://www.bioconductor.org/help/workflows/oligo-arrays/) and MeV (http://www.tm4.org/mev/) specialize in microarray analysis. Babelomics was used to analyze transcriptomic data in Arabidopsis thaliana to identify genes differentially expressed under drought stress. The tool facilitated functional annotation and identified key genes involved in stress responses [108] In Solanum tuberosum (potato), Bioconductor was utilized to analyze gene expression profiles under biotic stress conditions, identifying genes linked to pathogen resistance [194]. Genevestigator (https://www.genevestigator.com/gv/doc/plant_biotech.jsp) is a handy database containing extensive microarray data from various species, with the most comprehensive data from Arabidopsis thaliana. Data from crops like maize, wheat, rice, barley, and soybean are increasingly becoming available. Published expression data are publicly accessible in databases such as GEO (http://www.ncbi.nlm.nih.gov/geo/), ArrayExpress (http://www.ebi.ac.uk/arrayexpress/), and species-specific repositories, providing valuable resources for analyzing gene expression in these and other crops. A summary of genomic resources related to genome sequence and functional analysis is presented in Table 3.
Genomic tools for plant breeding
Traditional plant breeding involves selection and crossing of plants with desirable traits over several generations. Techniques include selection of superior individuals, hybridization, and backcrossing. Breeders aim to enhance traits like yield, disease resistance, and quality. The process relies on natural genetic variation and careful observation to achieve desired improvements in crops over time. Genomic tools enhance traditional plant breeding by providing precise insights into genetic variations, speeding up trait identification, and enabling targeted modifications. They use DNA sequencing and markers to identify desirable traits more accurately and rapidly, reducing the time and cost of developing improved plant varieties compared to traditional methods that rely on broader, less precise selection processes [87]. These tools help identify desirable traits, speed up the breeding process, and improve the overall outcomes of breeding programs. Some key genomic tools in plant breeding include:
Quantitative trait Loci (QTL) mapping
QTL mapping is a statistical technique that combines phenotypic data (traits) with genotypic data (molecular markers) in a specific population to identify genetic regions (QTLs) associated with complex traits. Various QTL mapping models have been developed, such as standard interval mapping (SIM) and multiple imputation (IMP) for unlinked QTLs and composite interval mapping (CIM) for both linked and unlinked QTLs/genes on chromosomes. The effectiveness of these methods is evaluated using LOD (logarithms-of-odds) scores, with QTLs considered significant above a threshold LOD score of 3.0. Access to reference genomes provides valuable genetic information on QTLs, aiding marker-assisted selection (MAS). Traditional QTL mapping requires a balanced population with known recombination data, enabling statistical associations between phenotypic and genotypic data through linkage mapping (Lander et al. 1989). Identifying QTL locations helps pinpoint genes responsible for specific traits and understand genetic variation mechanisms [84, 105]. Several software tools facilitate efficient QTL mapping, including QTL Cartographer, QTL IciMapping, MapQTL, R/qtl, TASSEL, PLABQTL, and JoinMap. In the 2017 study by Duhnen et al., the accuracy of traditional QTL mapping was compared with newer genomic prediction models, such as GBLUP and Bayesian methods, in soybean. The results showed that genomic prediction models outperformed QTL mapping by utilizing genome-wide markers, capturing both major and minor effect loci. This led to higher predictive accuracy for complex traits like yield and seed protein content, making genomic selection more effective for breeding. QTL mapping bridges genomics and field studies by linking genetic regions to quantitative traits. However, integrating data from multiple QTL studies to identify high-quality candidate loci for crop breeding remains challenging. Meta-analysis, which combines results from various studies, is crucial in accurate QTL prediction. Tools like MetaQTL reduce the confidence interval of QTL, improving location and effect estimation. Other tools, such as solQTL and RASQUAL, offer low-bias QTL analysis and data visualization. Meta-QTL analyses have been applied to crops such as rice, wheat, maize, cotton, and soybean, identifying significant traits like yield-related genes in wheat [182] and nitrogen use efficiency QTLs in rice [82]. Van and McHale (2017) performed a meta-QTL analysis with genetic information comprised of 175 QTLs for protein, 205 QTLs for oil, 156 QTLs for amino acids, and 113 QTLs for fatty acids. They detected 55 meta-QTL for seed composition on 6 out of 20 chromosomes. They also identified candidate genes within each meta-QTL aiding in crop improvement program. While QTL mapping effectively links traits to genomic regions and detects rare alleles, its resolution is limited by parental allelic diversity. Genome-wide association studies (GWAS) can address these limitations by identifying trait-associated genomic areas of diverse populations.
Genome-wide association study (GWAS)
Like QTL mapping, genome-wide association studies (GWAS) employ statistical association mapping to link molecular markers with traits of interest. GWAS leverages diverse populations and historical recombination, addressing linkage disequilibrium (LD) caused by physical linkage, genetic drift, selection, and population structure [71]. This method assesses marker-trait associations in diverse populations, although it faces challenges with false positives due to population structure and kinship (Huang et al. 2014). To mitigate this, covariates for structure and kinship are incorporated into mixed linear models (MLMs). Various statistical tools are used for association mapping, including MLM, CMLM, ECMLM, MLMM, GLM, and FarmCPU. Among these, the FarmCPU model is particularly effective at controlling false positives [77]. With advancements in sequencing technologies, GWAS has become a prominent tool for identifying loci linked to natural trait variations in crops. A population needs to be genotyped once and can be repeatedly used to map different traits with new phenotypic data. Despite its high false positive rate due to population structures and genetic relationships, GWAS is favored for exploratory analyses to identify a wide range of genomic leads. Unlike QTL mapping, GWAS is more likely to pinpoint specific candidate genes for crop improvement. It has been applied to various crops, including rice, soybean, maize, wheat, and canola. [24, 33]. For example, in Oryza sativa indica, sequencing of 517 landraces identified approximately 3.6 million SNPs, with GWAS of 14 agronomic traits explaining over 36% of phenotypic variance [64]. Myles et al. [112] used genomic resources to conduct a genome-wide association study (GWAS) in grapevine, identifying 3,600 SNP markers. They discovered loci linked to key traits like berry color (VvmybA1) and powdery mildew resistance, showcasing the utility of GWAS in identifying agriculturally significant genes in crops. In maize, GWAS revealed the genetic architecture of leaf traits, linking variation in liguleless genes to upright leaves [160]. He et al. [61] integrated GWAS with transcriptomic data and identified candidate genes involved in yield-related traits. This integrative approach facilitates the functional validation of the candidate genes. Bioinformatics tools such as PLINK, which uses standard regression for genotype–phenotype associations [126], and TASSEL, which incorporates mixed linear models to account for population structure, enhance GWAS studies [16]. GAPIT, another advanced tool, efficiently handles large datasets with over 1 million SNPs in 10,000 individuals using compressed mixed linear models and model-based prediction and selection methods [93].
Genome-based molecular breeding
Advancements in genomics and the availability of public genomic databases have significantly improved traditional breeding methods and facilitated the development of innovative crop breeding approaches (Fig. 2). Modern plant breeding focuses on evaluating the genetic gain of new genotypes by isolating genetic effects from environmental and noise components [12]. Different strategies, such as traditional phenotypic selection, marker-assisted selection (MAS), and genomic selection (GS), are employed. Traditional selection relies on phenotypic data for genetic evaluation. MAS, on the other hand, uses specific genetic markers linked to traits of interest, selecting individuals based on their marker scores [23]. Genomic selection is a more advanced method that considers markers with small effects on phenotypic variation. The process of genome-based molecular breeding involves several steps (Fig. 2). MAS, known for its efficiency, reduces both time and costs. MAS is faster as it doesn't require extensive phenotype testing of large progeny sets and allows for the pyramiding of multiple alleles.
Additionally, it reduces linkage drag and increases genetic gain compared to phenotypic selection [63]. Genetic merit can be evaluated in larger populations without losing genetic diversity. MAS-based breeding programs have been widely implemented in crops such as wheat, rice, maize, and tomato. For instance, MAS has been used to incorporate the Pi-ta gene into various rice varieties, enhancing resistance to rice blast disease caused by the fungus Magnaporthe oryzae [72]. Researchers identified the Sub1 gene in a flood-tolerant traditional rice variety called FR13A. The gene, located on chromosome 9 of the rice genome, was found to play a critical role in enhancing a plant’s ability to survive under submerged conditions. The Sub1 gene is part of a regulatory mechanism that involves three key ethylene response factors (ERFs)—Sub1A, Sub1B, and Sub1C. Of these, Sub1A is the primary gene responsible for submergence tolerance and enables rice plants to enter a state of metabolic dormancy, where growth and energy consumption are slowed, allowing the plant to conserve energy and survive for up to two weeks underwater. In 2009, researchers successfully integrated the Sub1 gene into several high-yielding, widely cultivated rice varieties, such as Swarna Sub1, IR64 Sub1, and Samba Mahsuri Sub1, using marker-assisted breeding techniques [144]. In wheat, Lr34 and Sr2 are two crucial genes that confer durable resistance to leaf rust and stem rust, respectively. These genes have been incorporated into wheat varieties using Marker-Assisted Selection (MAS) to develop improved cultivars viz. Avocet S (Lr34) and Thatcher (Sr2) that are resistant to rust diseases [184].
Similarly, the enhancement of oleic acid content in soybean oil using Marker-Assisted Selection (MAS) has been a significant achievement in soybean breeding. This improvement is primarily focused on selecting specific alleles at the FAD2-1A and FAD2-1B loci, which are critical in regulating the fatty acid composition of soybean oil [122]. The FAD2-1A and FAD2-1B genes encode the enzyme omega-6 desaturase, which is responsible for converting oleic acid (a monounsaturated fatty acid) into linoleic acid (a polyunsaturated fatty acid). By modifying or silencing these genes, the conversion of oleic acid to linoleic acid is reduced, leading to a higher proportion of oleic acid in the oil. Oleic acid is a healthier monounsaturated fatty acid that is more stable and resistant to oxidation than linoleic acid, which is more prone to rancidity. Through MAS, breeders have been able to select soybean plants carrying specific alleles at the FAD2-1A and FAD2-1B loci that reduce the activity of the FAD2 enzyme. Several high-oleic soybean varieties have been developed through this approach. These varieties contain over 70% oleic acid compared to traditional soybeans, which contain around 20–30%. Some of the notable improved varieties include: Plenish®, Vistive Gold®, and Monola® [57]. MAS has been used in barley for yield-related and stress tolerance traits, resulting in elite lines with improved malting quality by transferring the thermostable α-amylase from wild barley into a commercial variety [178]. Next-generation sequencing (NGS) technologies have increased the number of available markers, significantly boosting breeding efficiency. Markers such as SSRs, SNPs, InDels, and haplotypes are now crucial for efficient genotyping and constructing genetic maps. For example, SSR markers were used to map the Als gene on the 3H chromosome of barley, which is associated with a low number of tillers [34]. In a study on barley, 83 significant marker-trait associations were identified for six yield-related traits under drought conditions [1]. Various strategies have been applied to identify QTLs for relevant traits in these crops, greatly enhancing genomic-based molecular breeding through MAS.
Genomic Selection (GS), developed by Meuwissen et al. in 2001, represents a significant advancement beyond conventional Marker-Assisted Selection (MAS). While MAS suits traits influenced by a limited number of major genes, it falters with quantitative traits, often governed by numerous minor genes, as seen in yield and stress tolerance traits. GS tackles this challenge by harnessing an array of genetic markers spread across the genome to compute Genomic Estimated Breeding Values (GEBV) for each individual. GEBV integrates phenotypic data with marker and pedigree data, yielding superior prediction accuracy compared to MAS [18, 62]. GS strategically selects genome-spanning genetic markers, ensuring that all Quantitative Trait Loci (QTLs) align with at least one marker [52]. The first step in the genomic selection process is creating a training population of individuals with genotypic and phenotypic data (Fig. 2). This data is used to construct a predictive model, employing phenotypes as responses and genotypes as predictors.
Insights derived from this model then enable the estimation of GEBV for the breeding population, consisting solely of individuals with genotypic data. Genomic Selection seems a viable approach to advance genetic development within breeding programs in climate change. Grain yield, qualitative characteristics, and abiotic stress resistance have all benefited from using genomic selection. This rapid improvement results from the selection of desired phenotypes across generations. Genomic selection offers a significant advantage by reducing breeding cycle duration and lowering phenotyping costs, thereby accelerating genetic improvements for food security. Factors influencing prediction accuracy include training and breeding population sizes, genetic diversity, heritability, genotype-environment interactions, marker density, and genetic relationships [96]. Several methods have evolved for genomic selection over time. An easy linear model, such as ordinary least-squares regression, is frequently used as the first step in the genomic selection process. An issue that frequently arises in linear models with a large number of genome-wide markers is that the number of markers (p) might significantly exceed the number of observations (n), leading to overparameterization. An alternative strategy is adapted to deal with this problem. A selection of significant markers is used in modified least-squares regression. This log-likelihood model may lead to the loss of important data when choosing markers with significant effects.
Ridge regression (RR) is a penalized regression technique that solves overparameterization and multicollinearity [109]. The least absolute shrinkage and selection operator (LASSO) is another variant, that may have difficulties with strongly correlated predictors [118]. Elastic net (ENET) is a modification of LASSO that balances the ℓ1 and ℓ2 penalties in LASSO and RR, respectively, and is robust to extreme correlations among the predictors [46]. A Bayesian method to estimate marker effects is provided by Bayesian models like Bayes A, Bayes B, Bayes C, and Bayes D [56]. Genomic BLUP (GBLUP), one of the best linear unbiased prediction (BLUP) techniques, uses genomic relationships to estimate genetic values [131]. Ridge regression (RR), best linear unbiased prediction (BLUP), least absolute shrinkage and selection operator (LASSO), support vector machine (SVM), artificial neural network (ANN), and random forest (RF) are examples of approaches based on single trait genomic selection (STGS). The multi-trait genomic selection technique (MTGS), which increases prediction accuracy for numerous characteristics, uses multivariate regression with covariance estimation (MRCE) and multivariate LASSO (MLASSO). A number of software tools and packages have been created to evaluate genomic prediction accuracy and make genomic selection (GS) usable. Some notable tools and approaches are rrBLUP, solGS, GMStool, BWGS, BGLR, GenSel, GSelection and lme4GS [18]. Genomic Selection (GS) revolutionizes crop improvement, streamlining breeding for precision and efficiency. Utilizing genetic markers, GS predicts crop yields (Table 4). For example, Cerioli et al. [21] used the LSU500 marker set to achieve predictive abilities of 0.13 to 0.78 in various rice trials. GS is equally potent in disease resistance breeding, as seen in Zhang et al.’s [189] study on Fusarium head blight (FHB) traits in wheat, yielding prediction accuracies of up to 0.59 for FHB disease index and 0.54 for Fusarium damaged Kernels (FDK). Furthermore, GS enhances abiotic stress tolerance. Zhang et al. [190] assessed maize drought tolerance with prediction accuracies ranging from 0.19 to 0.33 for traits like seedling emergence rate, plant height, and grain yield. Rutkoski et al. [136] reported that genomic selection (GS) improved wheat grain yield by 15% compared to conventional breeding methods. In their study, GS utilized 1,056 single nucleotide polymorphism (SNP) markers to predict breeding values, enabling more accurate selection and accelerating yield gains from 0.5% to 1.5% per year, demonstrating the effectiveness of GS in wheat improvement. In Maize two hybrids viz. Pioneer® P1197AM and Pioneer® P1329AM have been developed using genomic selection to enhance traits like yield and disease resistance [153]. GS facilitated the development of soybeans with enhanced resistance to diseases and pests, improving overall crop resilience and yield. Asgrow®® variety of soybean has been developed through GS with improved disease resistance and enhanced yield potential [135]. These findings underscore Genomic Selection's potential to boost crop productivity and resilience to adverse environmental conditions and accelerates the development of new varieties with desirable traits. Recent advancements in plant breeding have prominently featured genome editing technologies which allows precise alterations to specific genes, enabling targeted modifications for traits like disease resistance or yield improvements. This approach provides exact changes at the genetic level. In contrast, genomic selection involves evaluating the genetic potential of plants using DNA markers linked to multiple traits. It assesses overall genetic merit and guides breeding decisions by predicting future performance. While editing is precise, selection offers a broader assessment for enhancing complex traits [114].
Genome editing
Genome editing, a powerful genetic modification method, enables precise alterations in DNA sequences using molecular scissors and artificial nucleases. Its origins date back to successful gene insertions in mammalian genomes in 1985–1986 [152]. Three main genome editing technologies are Zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR/Cas systems. CRISPR/Cas stands out for its simplicity, efficiency, and versatility [148]. Genome editing in agriculture enhances crop traits, like increased yield, disease and pest resistance, accelerating breeding programs, ensuring food security, and promoting sustainable farming practices in a rapidly changing world (Table 6). Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) system is an amazing bacterial defense mechanism against plasmid and virus invasion [75]. The sequences were first discovered in E. coli in 1987 when researchers found conserved direct-repeat sequences [69]. A crucial CRISPR/Cas9 development occurred in 2012 when Jinek et al. demonstrated SpCas9, derived from Streptococcus pyogenes, to be successful as an RNA-guided DNA endonuclease in vitro. This system inserts invading DNA pieces between crRNA repetitions in the host CRISPR locus. Mature crRNAs are produced through active transcription and pre-processing of these pre-crRNAs by Cas9 and host factors. Cas9 is directed to the appropriate target locus inside the invading DNA by the mature crRNA-Cas9 complex. The PAM motif (Protospacer adjacent motif) is often ahead of the site-specific cleavage that the Cas9 nuclease produces. The conventional PAM sequence 5' NGG 3' at the 3′ end of the target site is principally needed for the SpCas9. CrRNA and tracrRNA are combined to create chimeric single guide RNA (sgRNA), which directs Cas9 to the target region and introduces double-stranded breaks. Both homology-directed repair (HDR) and non-homologous end-joining (NHEJ) techniques can be used to fix these breaks. Customized gRNA-Cas9 complexes have been used to target economically significant traits in plants, highlighting the promise of CRISPR/Cas9 in agricultural applications [164].
In nature, there are two main types of CRISPR-Cas immune systems: class 1, which uses multiprotein complexes for nucleic acid cleavage, and class 2, which uses single-protein effector domains (Makarova et al., 2020). Because they benefit from a single protein effector, class 2 systems, especially types II, V, and VI, are preferred in biological research and translational applications. Regarding the class 2 Cas proteins, type-II Cas9 and type-V Cas12 are RNA-guided DNA endonucleases, whereas type-VI Cas13 mainly targets and cleaves RNA. Compared to type II CRISPR-Cas9, type V CRISPR-Cas12a (formerly CRISPR-Cpf1, a CRISPR from Prevotella and Francisella 1) and CRISPR-Cas12b are significantly different (Wang et al. 2020). SpCas9 requires a 5′-NGG-3′ PAM, whereas Cpf1 uses a 5′-TTTN-3′ or 5′-TTN-3′ PAM, hence extending the range of target sites in the genome. Additionally, Cas12a permits gene targeting with shorter crRNAs, possibly lowering genome editing costs [8]. The RNA-guided endonucleases known as Cas9 from type-II CRISPR systems have revolutionized genome editing. In the target DNA sequences, they cause double-strand breaks (DSBs) [73]. To create ribonucleoprotein complexes in their natural environment, Cas9 nucleases rely on CRISPR RNAs (crRNAs) linked with trans-activating crRNAs (tracrRNAs) [36]. However, single guide RNAs (sgRNAs), which combine crRNA and tracrRNA into one molecule, are used in the majority of genome editing applications [75]. An important sequence is the protospacer adjacent motif (PAM), which is located 3' of the target DNA strand in opposition to the guide RNA. Cas9 nuclease typically generates blunt-ended DSBs 3 base pairs upstream of the PAM, although some Cas9 nucleases exhibit alternative cutting patterns [142]. After the Cas9-guide RNA complex binds to the corresponding PAM, the DNA is unwound to create an RNA•DNA heteroduplex with the target DNA strand [73]. From the PAM-proximal region of the protospacer to the PAM-distal end, the process moves in one direction. 'R-loop' formed by the non-target DNA strand is a single-stranded DNA structure that is exposed and accessible. Base editing and prime editing, two more recent genome editing techniques, take advantage of this property [5]. Cas9 undergoes conformational changes, activating its nuclease domains during R-loop formation. After activation, Cas9 hydrolyses DNA's phosphodiester backbone via the HNH nuclease, cleaving the target DNA strand linked to the guide RNA and the RuvC-like nuclease, cleaving the non-target DNA strand containing PAM. Cas9 nickases, produced by mutations in either nuclease domain, are valuable for base and prime editing as they cleave a single DNA strand [130]. Both domains can be inactivated to create catalytically dead Cas9 (dCas9), retaining DNA binding abilities for applications like transcriptional regulation and epigenetic modifications. Many Cas9 variants have evolved since the discovery of Streptococcus pyogenes (SpCas9) Cas9 nuclease, varying in size, PAM recognition, guide RNA architecture, spacer length, editing efficiency, and specificity. SpCas9, the most popular, contains 1,368 amino acids, recognizes NGG PAMs, and supports both sgRNAs and crRNA/tracrRNA pairings with 20-nt spacers. However, it exhibits a higher off-target editing rate. Specialized Cas9 variants like SaCas9 (smaller, 1053 amino acids) and Nme2Cas9 (recognizing pyrimidine-rich PAMs) offer unique advantages, enabling researchers to tailor their choice of Cas9 effector for specific genome editing needs [42, 128]. This diversity advances the study of molecular biology and biotechnology by enabling researchers to choose the best Cas9 effector for their particular needs in genome editing. Wang et al. [169] reported a new gain-of-function OsGS2/GRF4 allele generated by CRISPR/Cas9 genome editing increases rice grain size and yield. Errum et al. [43] found that CRISPR/Cas9 editing of wheat Ppd-1 gene homoeologs alters spike architecture and grain morphometric traits and increases 1000-grain weight, grain width, grain length, plant height, and spikelets per spike. Genome editing has led to the development of several crop varieties with improved traits, showcasing the potential of this technology in agriculture. In rice, CRISPR-edited varieties such as Sasakure and Nipponbare have been developed to enhance yield and boost resistance to diseases, addressing key agricultural challenges [91]. In wheat, CRISPR-edited lines resistant to wheat blast provide protection against this devastating fungal pathogen, improving crop resilience and productivity [174, 175]. Wheat variety developed for drought tolerance through genome editing is the CRISPR/Cas9-edited wheat targeting the TaDREB2 gene, which is associated with drought stress response. By modifying this gene, researchers improved the plant’s drought tolerance without negatively impacting yield. For instance, Kim et al. [79] reported enhanced drought tolerance in wheat through CRISPR/Cas9 by targeting transcription factors like TaDREB2, aiding in the development of more resilient wheat varieties in drought-prone regions. Similarly, in soybeans, genome editing has led to the creation of high oleic acid varieties, which improve the nutritional quality and stability of soybean oil, offering health and market benefits (Zhai et al., 2020). Alternative genome manipulation techniques such as base editing and prime editing offer distinct approaches for precise DNA modifications without causing double-strand breaks, beyond traditional methods like CRISPR-Cas9, making them ideal for crop improvement (Table 5). Base editing enables direct base conversion, such as converting cytosine to thymine. For example, it has been used to create herbicide-resistant rice by modifying the ALS gene [191, 195]. Prime editing is more versatile, allowing targeted insertions, deletions, and base substitutions. It has been applied to improve disease resistance in crops like rice by precisely altering genes like OsSWEET for bacterial blight resistance [191, 195]. Beyond simply modifying DNA sequences, CRISPR-Cas9 has also been adapted to control gene expression. Cas9 variants that lack cutting activity, such as dCas9 (dead Cas9), can be fused with transcriptional activators or repressors to modulate gene expression, opening new doors for research into gene regulation, epigenetics, and functional genomics. CRISPR-dCas9 offers a non-invasive way to control gene expression without introducing mutations. This is particularly advantageous in crops where regulatory and public concerns over genetically modified organisms (GMOs) are prevalent. By modulating gene activity instead of editing the genome, CRISPR-dCas9 allows for crop improvement while potentially avoiding some regulatory hurdles associated with traditional genetic modification. In a significant study by Lowder et al. [101], researchers used CRISPR-dCas9 technology to enhance yield and drought tolerance in tomato (Solanum lycopersicum). The researchers designed a CRISPR-dCas9 system where dCas9 was fused with transcriptional activators to upregulate genes involved in auxin biosynthesis and drought stress response. This enabled precise modulation of key genes without altering the tomato genome. A detailed study of genome editing has been presented in Table 6.
Regulatory framework for genome-edited crops
Genome editing raises several ethical issues, including the possibility of unintended genetic changes, long-term effects on ecosystems, and disparities in access to technology. Potential risks involve off-target mutations and unforeseen consequences that could impact environmental stability or human health. Ethical discussions focus on maintaining responsible research practices, obtaining informed consent, and considering the broader societal impacts of genetic alterations. To manage these concerns effectively, it is vital to establish strong regulatory frameworks and foster open, transparent public discussions on the use of genome editing technologies [140]. Regulations for genome-edited crops vary globally [157]. In the USA, the regulatory framework for genome-edited crops is managed by three agencies. In the United States, the regulatory framework for genome-edited crops is relatively permissive. The U.S. Department of Agriculture (USDA) oversees genetically engineered (GE) plants under the Plant Protection Act. In 2020, the USDA announced that crops developed through genome editing, such as those edited with CRISPR/Cas9, would not be regulated if they do not contain DNA from other species. This approach is exemplified by the CRISPR-edited mushroom developed by Penn State researchers, which resists browning and was not subjected to USDA regulations because it does not introduce foreign genes [13]. However, genome-edited crops may still be subject to regulation by the Environmental Protection Agency (EPA) and the Food and Drug Administration (FDA), depending on their traits. The FDA reviews crops for food safety, while the EPA regulates plants engineered to produce pesticides. The European Union (EU) adopts a more stringent regulatory approach. Genome-edited crops are treated similarly to genetically modified organisms (GMOs) under EU regulations. This requires extensive safety assessments, risk evaluations, and labeling before they can be marketed. In 2018, the European Court of Justice (ECJ) ruled that genome-edited crops should be regulated under the same laws as GMOs, meaning they must undergo rigorous assessments [41]. For example, CRISPR-edited wheat developed by Rothamsted Research, aimed at improving disease resistance, faced substantial regulatory hurdles due to the EU’s stringent GMO regulations [11]. The wheat’s developers had to navigate a complex approval process, reflecting the EU’s cautious stance on genome editing. Canada’s regulatory framework is focused on the end product rather than the method of development. The Canadian Food Inspection Agency (CFIA) evaluates whether genome-edited crops require regulation based on their traits. If the modifications do not result in novel traits or substances, the crop may not require extensive oversight [22]. For example, CRISPR-edited canola with improved oil composition has been developed, and the CFIA determined that it did not necessitate extensive regulatory scrutiny as long as it did not introduce novel traits [151].
Australia’s approach is also product-based, with a focus on the characteristics of the final crop. The Gene Technology Regulator (GTR) assesses whether genome-edited crops contain new traits or genes that would warrant regulation. If the modifications do not introduce new genetic material or traits associated with GMOs, the crop may not face stringent controls [53]. An example is CRISPR-edited barley with enhanced disease resistance. The GTR decided that the crop did not require additional regulatory measures as long as the edits did not involve new genetic material [45]. Japan’s regulatory framework is evolving as the country seeks to balance safety with innovation. Initially, genome-edited crops were assessed under the existing GMO framework, which involves rigorous safety evaluations. However, Japan is working towards creating a more tailored approach to genome editing [103]. For example, CRISPR-edited soybeans with improved nutritional properties are currently under review. Recent discussions in Japan are focused on developing regulations that distinguish between genome editing and traditional GMOs, potentially streamlining the approval process [138]. The government of India issued an Office Memorandum on 30th March 2022 and exempted SDN-1 and SDN-2 genome-edited plants without foreign DNA from Rules 7 to 11 of the EPA, 1986. This streamlines approval, bypassing the Genetic Engineering Appraisal Committee (GEAC) for genome-edited crops in India. The global regulatory landscape for genome-edited crops varies widely, reflecting diverse national priorities and approaches to biotechnology. At present, the United States and India employs a permissive approach focused on the end product, while the European Union maintains strict GMO-like regulations. Canada and Australia use a product-based perspective, assessing crops based on their traits rather than their development method. Japan is evolving towards a more nuanced regulatory framework. These differences highlight the ongoing debate over balancing innovation with safety and environmental considerations in the era of advanced genetic engineering [138].
Conclusion and perspectives
The future of plant breeding promises advancements through genetic tools, precision breeding techniques, and climate-resilient crops to address global food security. Key genomic resources include genetic markers, reference genomes, databases, transcriptomes, and gene expression profiles. These tools are crucial for identifying genes linked to desirable traits, understanding genetic diversity, and accelerating breeding programs. Molecular markers will continue to enhance traditional breeding by enabling the selection of disease-resistant, drought-tolerant, and high-yield plants, leading to faster and more precise crop development. Genomic selection is poised to revolutionize plant breeding. It harnesses big data and advanced analytics to predict an individual plant's performance based on its genetic makeup [18]. This enables breeders to select and propagate plants with the highest genetic potential, significantly shortening breeding cycles and increasing the efficiency of trait improvement. Genome editing technologies, like CRISPR-Cas9, offer unprecedented precision in crop enhancement. They allow for the targeted modification of specific genes to introduce or enhance desirable traits while minimizing unintended changes [4]. CRISPR/Cas-9 and related systems (such as CRISPR/Cas-12 and CRISPR/Cas-13) are recognized as groundbreaking tools for genome editing due to their precision, efficiency, affordability, and simplicity. These technologies facilitate highly accurate modifications of nuclear genomes. However, a significant challenge is the potential for off-target mutations, which can lead to harmful phenotypes and limit the broader application of genome editing. To address this issue, new CRISPR/Cas variants are being developed, and existing systems are being enhanced to minimize off-target effects by selecting specific single guide RNAs (sgRNAs) with fewer predicted off-target sites based on a comprehensive reference genome sequence. Genome editing holds immense potential for creating crops with improved nutritional content, reduced susceptibility to pests and diseases, and increased environmental resilience. Collectively, these tools are instrumental in developing crop varieties that meet the challenges of a growing global population, changing climates, and sustainable agriculture practices. However, ethical, regulatory, and safety considerations will shape their future deployment.
Availability of data and materials
Not applicable.
Abbreviations
- SNP:
-
Single Nucleotide Polymorphism
- CRISPR/Cas9:
-
Clustered Regularly Interspaced Short Palindromic Repeats-Associated Protein 9
- QTL:
-
Quantitative Trait Loci
- RFLP:
-
Restriction Fragment Length Polymorphism
- RAPD:
-
Random Amplified Polymorphic DNA
- AFLP:
-
Amplified Fragment Length Polymorphism
- SSR:
-
Simple Sequence Repeats
- STR:
-
Short Tandem Repeats
- VNTR:
-
Variable Number of Tandem Repeats
- PCR:
-
Polymerase Chain Reaction
- NGS:
-
Next Generation Sequencing
- NCBI:
-
National Center for Biotechnology Information
- EMBL:
-
European Molecular Biology Laboratory
- DDBJ:
-
DNA Data Bank of Japan
- UniProt:
-
Universal Protein Resource
- UCSC:
-
University of California Santa Cruz
- KEGG:
-
Kyoto Encyclopedia of Genes and Genomes
- STRING:
-
Search Tool for the Retrieval of Interacting Genes/Proteins
- BioGRID:
-
Biological General Repository for Interaction Datasets
- SGN:
-
Sol Genomics Network
- QRTPCR:
-
Quantitative Real-Time PCR
References
Abou-Elwafa SF. Association mapping for yield and yield-contributing traits in barley under drought conditions with genome-based SSR markers. CR Biol. 2016;339(5–6):153–62.
Alam MS, Kong J, Tao R, Ahmed T, Alamin M, Alotaibi SS, Abdelsalam NR, Xu JH. CRISPR/Cas9 mediated knockout of the OsbHLH024 transcription factor improves salt stress resistance in rice (Oryza sativa L.). Plants. 2022;11(9):1184.
Amiteye S. Basic concepts and methodologies of DNA marker systems in plant molecular breeding. Heliyon. 2021;7(10):e08093.
Anzalone AV, Koblan LW, Liu DR. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol. 2020;38(7):824–44.
Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, Liu DR. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 2019;576(7785):149–57.
Baba T, Momen M, Campbell MT, Walia H, Morota G. Multi-trait random regression models increase genomic prediction accuracy for a temporal physiological trait derived from high-throughput phenotyping. PLoS ONE. 2020;15(2): e0228118.
Badoni S, Das S, Sayal YK, Gopalakrishnan S, Singh AK, Rao AR, Agarwal P, Parida SK, Tyagi AK. Genome-wide generation and use of informative intron-spanning and intron-length polymorphism markers for high-throughput genetic analysis in rice. Sci Rep. 2016;6(1):23765.
Bae T, Hur JW, Kim D, Hur JK. Recent trends in CRISPR-Cas system: genome, epigenome, and transcriptome editing and CRISPR delivery systems. Genes Genom. 2019;41:871–7.
Bastet A, Zafirov D, Giovinazzo N, Guyon-Debast A, Nogué F, Robaglia C, Gallois JL. Mimicking natural polymorphism in eIF 4E by CRISPR-Cas9 base editing is associated with resistance to potyviruses. Plant Biotechnol J. 2019;17(9):1736–50.
Beche E, Gillman JD, Song Q, Nelson R, Beissinger T, Decker J. Genomic prediction using training population design in interspecific soybean populations. Mol Breed. 2021;41:1–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/S11032-021-01203-6.
Benson DA, et al. Regulatory challenges for CRISPR-edited crops in Europe. Nature Plants. 2019;5:831–5.
Bernardo R. Breeding for quantitative traits in plants. Woodbury MN USA: Stemma Press; 2002.
Bertier LD, et al. CRISPR-edited mushroom with reduced browning. Nat Biotechnol. 2021;39(9):1067–75.
Bhandari A, Bartholomé J, Cao-Hamadoun T-V. Selection of trait-specific markers and multi-environment models improve genomic predictive ability in rice. PLoS ONE. 2019;14: e0208871.
Botstein D, White RL, Skolnick M, Davis RW. Construction of a genetic linkage map in man using restriction fragment length polymorphisms. Am J Hum Genet. 1980;32(3):314.
Bradbury PJ, Zhang Z, Kroon DE, Casstevens TM, Ramdoss Y, Buckler ES. TASSEL: software for association mapping of complex traits in diverse samples. Bioinformatics. 2007;23(19):2633–5.
Brauer EK, Balcerzak M, Rocheleau H, Leung W, Schernthaner J, Subramaniam R, Ouellet T. Genome editing of a deoxynivalenol-induced transcription factor confers resistance to Fusarium graminearum in wheat. Mol Plant Microbe Interact. 2020;33(3):553–60.
Budhlakoti N, Kushwaha AK, Rai A, Chaturvedi KK, Kumar A, Pradhan AK, Kumar U, Kumar RR, Juliana P, Mishra DC, Kumar S. Genomic selection: a tool for accelerating the efficiency of molecular breeding for development of climate-resilient crops. Front Genet. 2022;13: 832153.
Butler NM, Baltes NJ, Voytas DF, Douches DS. Geminivirus-mediated genome editing in potato (Solanum tuberosum L.) using sequence-specific nucleases. Front Plant Sci. 2016;7:1045.
Cavanagh CR, Chao S, Wang S, Huang BE, Stephen S, Kiani S, Forrest K, Saintenac C, Brown-Guedira GL, Akhunova A, See D. Genome-wide comparative diversity uncovers multiple targets of selection for improvement in hexaploid wheat landraces and cultivars. Proc Natl Acad Sci. 2013;110(20):8057–62.
Cerioli T, Hernandez CO, Angira B, McCouch SR, Robbins KR, Famoso AN. Development and validation of an optimized marker set for genomic selection in southern US rice breeding programs. The Plant Genome. 2022;3: e20219.
CFIA. Guidance on regulatory status of genome edited plants.Canadian Food Inspection Agency.2021.
Charcosset A. Marker-assisted introgression of quantitative trait loci. Genetics. 1997;147:1469–85.
Chattha WS, Atif RM, Iqbal M, Shafqat W, Farooq MA, Shakeel A. Genome-wide identification and evolution of Dof transcription factor family in cultivated and ancestral cotton species. Genomics. 2020;112(6):4155–70.
Chen K, Ji F, Yuan S, Hao W, Wang W, Hu ZH. The performance of activated sludge exposed to arsanilic acid and amprolium hydrochloride in sequencing batch reactors. Int Biodeterior Biodegr. 2017;116:260–5.
Chen X, Zhang Y, Wang J, Li H, Xu H, Zhang Y, Zhao X. Application of iSNAP markers for mapping QTLs associated with drought tolerance in wheat. Front Plant Sci. 2023;14:1048790.
Chen J, Liu H, Wang Y, Zhang Z, Li Z. Application of intron length polymorphism (ILP) markers for mapping drought tolerance in rice (Oryza sativa). Plant Science Journal. 2022;189(2):351–60.
Chen W, Gao Y, Xie W, Gong L, Lu K, Wang W, Li Y, Liu X, Zhang H, Dong H, Zhang W. Genome-wide association analyses provide genetic and biochemical insights into natural variation in rice metabolism. Nat Genet. 2014;46(7):714–21.
Clarke WE, Higgins EE, Plieske J, Wieseke R, Sidebottom C, Khedikar Y, Batley J, Edwards D, Meng J, Li R, Lawley CT. A high-density SNP genotyping array for Brassica napus and its ancestral diploid species based on optimised selection of single-locus markers in the allotetraploid genome. Theor Appl Genet. 2016;129:1887–99.
Close TJ, Lucas MR, Muñoz-Amatriain M, Mirebrahim H, Wanamaker S, Barkley NA, Clair SS, Guo YN, Lo S, Huynh BL. A new SNP-genotyping resource for cowpea and its deployment for breeding. In Proceedings of the Plant and Animal Genome Conference, San Diego, CA, USA. 2015;23:P0784.
Comadran J, Kilian B, Russell J, Ramsay L, Stein N, Ganal M, Shaw P, Bayer M, Thomas W, Marshall D, Hedley P. Natural variation in a homolog of Antirrhinum CENTRORADIALIS contributed to spring growth habit and environmental adaptation in cultivated barley. Nat Genet. 2012;44(12):1388–92.
Cruz CD, et al. DArTseq for genetic diversity and trait mapping in crops. The Plant Genome. 2021;14(1): e20077.
Cui L, Yang G, Yan J, Pan Y, Nie X. Genome-wide identification, expression profiles and regulatory network of MAPK cascade gene family in barley. BMC Genom. 2019;20:1–20.
Dabbert T, Okagaki RJ, Cho S, Boddu J, Muehlbauer GJ. The genetics of barley low-tillering mutants: absent lower laterals (als). Theor Appl Genet. 2009;118:1351–60.
Das RR, Vinayan MT, Patel MB, Phagna RK, Singh SB, Shahi JP. Genetic gains with rapid-cycle genomic selection for combined drought and waterlogging tolerance in tropical maize (Zea May S L.). Plant Genom. 2020;13:1–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/tpg2.20035.
Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y, Pirzada ZA, Eckert MR, Vogel J, Charpentier E. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature. 2011;471(7340):602–7.
Dheer P, Rautela I, Sharma V, Dhiman M, Sharma A, Sharma N, Sharma MD. Evolution in crop improvement approaches and future prospects of molecular markers to CRISPR/Cas9 system. Gene. 2020;753: 144795.
Ding F, Qiang X, Jia Z, Li L, Hu J, Yin M, Xia S, Chen B, Qi J, Li Q, Gao Y. Knockout of a novel salt responsive gene SlABIG1 enhance salinity tolerance in tomato. Environ Exp Bot. 2022;200: 104903.
Du YT, Zhao MJ, Wang CT, Gao Y, Wang YX, Liu YW, Chen M, Chen J, Zhou YB, Xu ZS, Ma YZ. Identification and characterization of GmMYB118 responses to drought and salt stress. BMC Plant Biol. 2018;18:1–8.
Duhnen A, Miedaner T, Schierholt A. Genomic selection for yield and seed protein content in soybean: a comparison of the predictive ability of different statistical models. Theor Appl Genet. 2017;130(1):497–511.
ECJ. European Court of Justice Ruling on GMO Regulations. Court of justice of the European Union. 2018.
Edraki A, Mir A, Ibraheim R, Gainetdinov I, Yoon Y, Song CQ, Cao Y, Gallant J, Xue W, Rivera-Pérez JA, Sontheimer EJ. A compact, high-accuracy Cas9 with a dinucleotide PAM for in vivo genome editing. Mol Cell. 2019;73(4):714–26.
Errum A, Rehman N, Uzair M, Inam S, Ali GM, Khan MR. CRISPR/Cas9 editing of wheat Ppd-1 gene homoeologs alters spike architecture and grain morphometric traits. Funct Integr Genomics. 2023;23(1):66.
Fedoroff NV. The past, present and future of crop genetic modification. New Biotechnol. 2010;27(5):461–5.
Fletcher R, et al. Regulatory framework for CRISPR-edited barley in Australia. J Agric Food Chem. 2021;69(10):3223–31.
Friedman J, Hastie T, Tibshirani R. Regularization paths for generalized linear models via coordinate descent. J Stat Softw. 2010;33(1):1.
Fu L, Wu D, Zhang X, Xu Y, Kuang L, Cai S, Zhang G, Shen Q. Vacuolar H+-pyrophosphatase HVP10 enhances salt tolerance via promoting Na+ translocation into root vacuoles. Plant Physiol. 2022;188(2):1248–63.
Ganal MW, Durstewitz G, Polley A, Bérard A, Buckler ES, Charcosset A, Clarke JD, Graner EM, Hansen M, Joets J, Le Paslier MC. A large maize (Zea mays L.) SNP genotyping array: development and germplasm genotyping, and genetic mapping to compare with the B73 reference genome. PloS One. 2011;6(12):e28334.
Gao J, Liang H, Huang J, Qing D, Wu H, Zhou W, Chen W, Pan Y, Dai G, Gao L, Deng G. Development of the PARMS marker of the TAC1 gene and its utilization in rice plant architecture breeding. Euphytica. 2021;217:1–1.
Gao Z, Liu Q, Zhang Y, Chen D, Zhan X, Deng C, Cheng S, Cao L. OsCUL3a-associated molecular switches have functions in cell metabolism, cell death, and disease resistance. J Agric Food Chem. 2020;68(19):5471–82.
Garris AJ, Tai TH, Coburn J, Kresovich S, McCouch S. Genetic structure and diversity in Oryza sativa L. Genetics. 2005;169(3):1631–8.
Goddard ME, Hayes BJ. Genomic selection. J Anim Breed Genet. 2007;124(6):323–30.
GTR. Gene Technology Regulations in Australia. Gene Technology Regulator. 2022.
Gui Y, Yan G, Bo S, Tong Z, Wang Y, Xiao B, Lu X, Li Y, Wu W, Fan L. iSNAP: a small RNA-based molecular marker technique. Plant Breeding. 2011;130(5):515–20.
Gupta PK, Varshney RK, Sharma PC, Ramesh B. Molecular markers and their applications in plant breeding. Plant Breeding. 2020;139(3):383–99.
Habier D, Fernando RL, Kizilkaya K, Garrick DJ. Extension of the Bayesian alphabet for genomic selection. BMC Bioinformatics. 2011;12(1):1–2.
Hagely K, Konda AR, Kim JH, Cahoon EB, Bilyeu K. Molecular-assisted breeding for soybean with high oleic/low linolenic acid and elevated vitamin E in the seed oil. Mol Breeding. 2021;41(1):5.
Haile TA, Heidecker T, Wright D, Neupane S, Ramsay L, Vandenberg A. Genomic selection for lentil breeding: empirical evidence. Plant Genome. 2020;13:1–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/tpg2.20002.
Hamilton JP, Hansey CN, Whitty BR, Stoffel K, Massa AN, Van Deynze A, De Jong WS, Douches DS, Buell CR. Single nucleotide polymorphism discovery in elite North American potato germplasm. BMC Genomics. 2011;12:1–2.
Hasan N, Choudhary S, Naaz N, Sharma N, Laskar RA. Recent advancements in molecular marker-assisted selection and applications in plant breeding programmes. J Genet Eng Biotechnol. 2021;19(1):1–26.
He X, Lu M, Cao J, Pan X, Lu J, Zhao L, Zhang H, Chang C, Wang J, Ma C. Genome-wide association analysis of grain hardness in common wheat. Genes. 2023;14(3):672.
Heffner EL, Sorrells ME, Jannink JL. Genomic selection for crop improvement. Crop Sci. 2009;49(1):1–12.
Hospital F. Selection in backcross programmes. Philos Trans Royal Soc B. 2005;360(1459):1503–11.
Huang X, Wei X, Sang T, Zhao Q, Feng Q, Zhao Y, Li C, Zhu C, Lu T, Zhang Z, Li M. Genome-wide association studies of 14 agronomic traits in rice landraces. Nat Genet. 2010;42(11):961–7.
Huang X, Wei X, Wang A, Zhao Q, Li C, Wang L, Han B. Genome-wide association studies of 14 agronomic traits in rice landraces. Nature. 2020;578(7796):274–8.
Huttner E, Wenzl P, Akbari M, Caig V, Carling J, Cayla C, Evers M, Jaccoud D, Peng K, Patarapuwadol S, Uszynski G. Diversity arrays technology: a novel tool for harnessing the genetic potential of orphan crops. InDiscovery to Delivery: BioVision Alexandria 2004, Proceedings of the 2004 Conference of The World Biological Forum. CABI Publishing: UK (pp. 145–155).
Huynh BL. A new SNP-genotyping resource for cowpea and its deployment for breeding. Proc Plant Anim Genom Conf San Diego CA USA. 2015;23:P0784.
Illouz-Eliaz N, Nissan I, Nir I, Ramon U, Shohat H, Weiss D. Mutations in the tomato gibberellin receptors suppress xylem proliferation and reduce water loss under water-deficit conditions. J Exp Bot. 2020;71(12):3603–12.
Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol. 1987;169(12):5429–33.
Jang H, Jo DH, Cho CS, Shin JH, Seo JH, Yu G, Gopalappa R, Kim D, Cho SR, Kim JH, Kim HH. Application of prime editing to the correction of mutations and phenotypes in adult mice with liver and eye diseases. Nature Biomed Eng. 2022;6(2):181–94.
Jannink JL, Bink MC, Jansen RC. Using complex plant pedigrees to map valuable genes. Trends Plant Sci. 2001;6(8):337–42.
Jiang CJ, Shimono M, Maeda S, Inoue H, Mori M, Hasegawa M, Sugano S, Takatsuji H. Suppression of the rice fatty-acid desaturase gene OsSSI2 enhances resistance to blast and leaf blight diseases in rice. Mol Plant Microbe Interact. 2009;22(7):820–9.
Jiang F, Doudna JA. CRISPR–Cas9 structures and mechanisms. Annu Rev Biophys. 2017;46:505–29.
Jiang T, Zhang XO, Weng Z, Xue W. Deletion and replacement of long genomic sequences using prime editing. Nat Biotechnol. 2022;2:227–34.
Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816–21.
Kakeshpour T, Tamang TM, Motolai G, Fleming ZW, Park JE, Wu Q, Park S. CGFS-type glutaredoxin mutations reduce tolerance to multiple abiotic stresses in tomato. Physiol Plant. 2021;173(3):1263–79.
Kaler AS, Gillman JD, Beissinger T, Purcell LC. Comparing different statistical models and multiple testing corrections for association mapping in soybean and maize. Front Plant Sci. 2020;10: 486047.
Kim SY, Bengtsson T, Olsson N, Hot V, Zhu LH, Åhman I. Mutations in two aphid-regulated β-1, 3-glucanase genes by CRISPR/Cas9 do not increase barley resistance to Rhopalosiphum padi L. Front Plant Sci. 2020;11:1043.
Kim D, Alptekin B, Budak H. CRISPR/Cas9 genome editing in wheat. Funct Integr Genom. 2021;21(1):31–41.
Kuang Y, Li S, Ren B, Yan F, Spetz C, Li X, Zhou X, Zhou H. Base-editing-mediated artificial evolution of OsALS1 in planta to develop novel herbicide-tolerant rice germplasms. Mol Plant. 2020;13(4):565–72.
Kumar N, Galli M, Ordon J, Stuttmann J, Kogel KH, Imani J. Further analysis of barley MORC 1 using a highly efficient RNA-guided Cas9 gene-editing system. Plant Biotechnol J. 2018;16(11):1892–903.
Kumar R, Nagarajan M, Bollinedi H, Bhowmick PK, Singh AK, Ellur RK, Sathee L, Mehrotra S, Kumar D, Krishnan SG, Vinod KK. Meta-QTLs linked to nitrogen use efficiency are randomly distributed in Indian rice germplasm. Ind J Genet Plant Breeding. 2022;82(01):7–15.
Kumar R, Saini M, Taku M, Debbarma P, Mahto RK, Ramlal A, Sharma D, Rajendran A, Pandey R, Gaikwad K, Lal SK. Identification of quantitative trait loci (QTLs) and candidate genes for seed shape and 100-seed weight in soybean [Glycine max (L.) Merr.]. Front Plant Sci. 2023;13:1074245.
Kumar R, Saini M, Taku M, Debbarma P, Sharma D, Pandey R, Gaikwad K, Lal SK, Talukdar A. Genetic studies and mapping of quantitative trait loci (QTL) for yield and yield-related traits in vegetable and seed soybean (Glycine max L.)-derived populations. Ind J Genet Plant Breeding. 2023;83(03):301–9.
Kump KL, Bradbury PJ, Wisser RJ, Buckler ES, Belcher AR, Oropeza-Rosas MA, Balint-Kurti PJ. Genome-wide association study of quantitative resistance to southern leaf blight in the maize nested association mapping population. Nat Genet. 2011;43(2):163–8.
Kumar A, Choudhary M, Sharma V, Singh R. Advances in SNP markers for crop improvement. Front Plant Sci. 2021;12:100–12.
Lamichhane S, Thapa S. Advances from conventional to modern plant breeding methodologies. Plant Breed Biotechnol. 2022;10(1):1–4.
Lander ES, Botstein D. Mapping mendelian factors underlying quantitative traits using RFLP linkage maps. Genetics. 1989;121(1):185–99.
Li S, Shen L, Hu P, Liu Q, Zhu X, Qian Q, Wang K, Wang Y. Developing disease-resistant thermosensitive male sterile rice by multiplex gene editing. J Integr Plant Biol. 2019;61(12):1201–5.
Li Y, Ruperao P, Batley J, Edwards D, Martin W, Hobson K, Sutton T. Genomic prediction of preliminary yield trials in chickpea: effect of functional annotation of SNPs and environment. The Plant Genome. 2022;15(1): e20166.
Li J, et al. CRISPR/Cas9-mediated genome editing in rice. Front Plant Sci. 2018;9:115.
Lin JL, Fang X, Li JX, Chen ZW, Wu WK, Guo XX, Liu NJ, Huang JF, Chen FY, Wang LJ, Xu B. Dirigent gene editing of gossypol enantiomers for toxicity-depleted cotton seeds. Nature Plants. 2023;9(4):605–15.
Lipka AE, Tian F, Wang Q, Peiffer J, Li M, Bradbury PJ, Gore MA, Buckler ES, Zhang Z. GAPIT: genome association and prediction integrated tool. Bioinformatics. 2012;28(18):2397–9.
Liu H, Lin Y, Chen G, Shen Y, Liu J, Zhang S. Genome-scale identification of resistance gene analogs and the development of their intron length polymorphism markers in maize. Mol Breeding. 2012;29:437–47.
Liu S, Li C, Wang H, Wang S, Yang S, Liu X, Yan J, Li B, Beatty M, Zastrow-Hayes Song G. Mapping regulatory variants controlling gene expression in drought response and tolerance in maize. Genom Biol. 2020;21:1–22.
Liu X, Wang H, Wang H, Guo Z, Xu X, Liu J, Wang S, Li WX, Zou C, Prasanna BM, Olsen MS. Factors affecting genomic selection revealed by empirical evidence in maize. Crop J. 2018;6(4):341–52.
Liu S, Huang W, Zhou X, Yang J, Li Y, Zhao J, Zhang Y. High-throughput iSNAP markers for identifying key loci associated with disease resistance and yield in maize. Plant Biotechnol J. 2022;20(3):534–47.
Liu J, et al. Applications of intron polymorphisms in crop genetics. J Integr Agric. 2022;21(4):987–95.
Liu Z, Zhang J, Pang X, Li Y, Han Y. Development of intron length polymorphism (ILP) markers in rice and their application in cross-species amplification. Mol Genet Genomics. 2010;283(5):449–57.
Livaja M, Unterseer S, Erath W, Lehermeier C, Wieseke R, Plieske J, Polley A, Luerßen H, Wieckhorst S, Mascher M, Hahn V. Diversity analysis and genomic prediction of Sclerotinia resistance in sunflower using a new 25 K SNP genotyping array. Theor Appl Genet. 2016;129:317–29.
Lowder LG, Zhang D, Baltes NJ, Paul JW, Tang X, Zheng X, Voytas DF, Hsieh TF, Zhang Y, Qi Y. CRISPR-dCas9-mediated activation of auxin and drought tolerance pathways in tomato (Solanum lycopersicum) enhances yield and stress resilience. Nature Plants. 2022;8(5):482–94.
M Perez-de-Castro A, Vilanova S, Cañizares J, Pascual L, M Blanca J, J Diez M, Prohens J, Picó B. Application of genomic tools in plant breeding. Current Genomics. 2012; 13(3):179–95.
MAFF. Japan’s evolving regulations on genome editing. ministry of agriculture, forestry, and fisheries. 2022.
Makarova KS, Wolf YI, Koonin EV. Evolutionary classification of CRISPR-cas systems. Crispr Biol Appl. 2022;30:13–38.
Maloy S, Hughes K. Brenner’s encyclopedia of genetics. Cambridge: Academic Press; 2013.
Mao H, Li S, Chen B, Jian C, Mei F, Zhang Y, Li F, Chen N, Li T, Du L, Ding L. Variation in cis-regulation of a NAC transcription factor contributes to drought tolerance in wheat. Mol Plant. 2022;15(2):276–92.
McCouch SR, Chen X, Panaud O, Temnykh S, Xu Y, Zhang Q. Developing and mapping SSR markers for rice. Theor Appl Genet. 2002;105(2–3):113–21.
Medina I, et al. Babelomics: an integrative platform for the analysis of transcriptomics, proteomics, and genomic data with advanced functional profiling. Nucleic Acids Res. 2010;38(suppl_2):W210–3.
Meuwissen TH, Hayes BJ, Goddard M. Prediction of total genetic value using genome-wide dense marker maps. Genetics. 2001;157(4):1819–29.
Michel S, Löschenberger F, Ametz C, Pachler B, Sparry E, Bürstmayr H. Combining grain yield, protein content and protein quality by multi-trait genomic selection in bread wheat. Theor Appl Genet. 2019;132:2767–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/S00122-019-03386-1.
Miller MJ, Song Q, Fallen B, Li Z. Genomic prediction of optimal cross combinations to accelerate genetic improvement of soybean (Glycine max). Front Plant Sci. 2023;14:1171135.
Myles S, Boyko AR, Owens CL, et al. Genetic structure and domestication history of the grape. Proc Natl Acad Sci. 2009;106(9):2325–30.
Mishra R, Mohanty JN, Mahanty B, Joshi RK. A single transcript CRISPR/Cas9 mediated mutagenesis of CaERF28 confers anthracnose resistance in chilli pepper (Capsicum annuum L.). Planta. 2021;254(1):5.
Mueller ML, Van Eenennaam AL. Synergistic power of genomic selection, assisted reproductive technologies, and gene editing to drive genetic improvement of cattle. CABI Agric Biosci. 2022;3(1):13.
Nakamura Y, Leppert M, O’Connell P, Wolff R, Holm T, Culver M, Martin C, Fujimoto E, Hoff M, Kumlin E, White R. Variable number of tandem repeat (VNTR) markers for human gene mapping. Science. 1987;235(4796):1616–22.
Nunez-Munoz L, Vargas-Hernández B, Hinojosa-Moya J, Ruiz-Medrano R, Xoconostle-Cázares B. Plant drought tolerance provided through genome editing of the trehalase gene. Plant Signal Behav. 2021;16(4):1877005.
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(12): e0243376.
Ogutu JO, Schulz-Streeck T, Piepho HP. Genomic selection using regularized linear regression models: ridge regression, lasso, elastic net and their extensions. In BMC proceedings. 2012;6:1–6.
Oliva R, Ji C, Atienza-Grande G, Huguet-Tapia JC, Perez-Quintero A, Li T, Eom JS, Li C, Nguyen H, Liu B, Auguy F. Broad-spectrum resistance to bacterial blight in rice using genome editing. Nat Biotechnol. 2019;37(11):1344–50.
Pandey MK, Chaudhari S, Jarquin D, Janila P, Crossa J, Patil SC, Sundravadana S, Khare D, Bhat RS, Radhakrishnan T, Hickey JM. Genome-based trait prediction in multi-environment breeding trials in groundnut. Theor Appl Genet. 2020;133:3101–17.
Pant SR, et al. Regulatory SNPs and their role in stress tolerance. Plant J. 2020;102(2):326–41.
Pham AT, Lee JD, Shannon JG, Bilyeu KD. Mutant alleles of FAD2-1A and FAD2-1B combine to produce soybeans with the high oleic acid seed oil trait. BMC Plant Biol. 2012;12:72.
Prihatna C, Barbetti MJ, Barker SJ. A novel tomato fusarium wilt tolerance gene. Front Microbiol. 2018;9:1226.
Probsting M, Schenke D, Hossain R, Hader C, Thurau T, Wighardt L, Schuster A, Zhou Z, Ye W, Rietz S, Leckband G. Loss of function of CRT1a (calreticulin) reduces plant susceptibility to Verticillium longisporum in both Arabidopsis thaliana and oilseed rape (Brassica napus). Plant Biotechnol J. 2020;18(11):2328–44.
Provan J, Powell W, Hollingsworth PM. Chloroplast microsatellites: new tools for studies in plant ecology and evolution. Trends Ecol Evol. 2001;16(3):142–7.
Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Maller J, Sklar P, De Bakker PI, Daly MJ, Sham PC. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81(3):559–75.
Rajendrakumar P, Biswal AK, Balachandran SM, Srinivasarao K, Sundaram RM. Simple sequence repeats in organellar genomes of rice: frequency and distribution in genic and intergenic regions. Bioinformatics. 2007;23(1):1–4.
Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, Zetsche B, Shalem O, Wu X, Makarova KS, Koonin EV. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520(7546):186–91.
Rangari SK, Kaur Sudha M, Kaur H, Uppal N, Singh G, Vikal Y, Sharma P. DNA-free genome editing for ZmPLA1 gene via targeting immature embryos in tropical maize. GM Crops Food. 2023;7:1–7.
Rees HA, Liu DR. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet. 2018;19(12):770–88.
Ren D, An L, Li B, Qiao L, Liu W. Efficient weighting methods for genomic best linear-unbiased prediction (BLUP) adapted to the genetic architectures of quantitative traits. Heredity. 2021;126(2):320–34.
Riaz A, Kanwal F, Börner A, Pillen K, Dai F, Alqudah AM. Advances in genomics-based breeding of barley: molecular tools and genomic databases. Agronomy. 2021;11(5):894.
Rio S, Mary-Huard T, Moreau L, Charcosset A. Genomic selection efficiency and a priori estimation of accuracy in a structured dent maize panel. Theor Appl Genet. 2019;132:81–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00122-018-3196-1.
Rousselle Y, Jones E, Charcosset A, Moreau P, Robbins K, Stich B, Knaak C, Flament P, Karaman Z, Martinant JP, Fourneau M. Study on essential derivation in maize: III. Selection and evaluation of a panel of single nucleotide polymorphism loci for use in European and North American germplasm. Crop Sci. 2015;55(3):1170–80.
Rodrigues KJMR, et al. Advancements in soybean breeding through genomic selection. J Crop Sci Biotechnol. 2020;23(2):151–62.
Rutkoski J, Heffner EL, Sorrells ME, Poland J, Jannink J-L, Singh RP. Genomic selection for grain yield in a wheat breeding program. Plant Genome. 2015;8(3):1–12.
Sachidanandam R, Weissman D, Schmidt SC, Kakol JM, Stein LD, Marth G, Sherry S, Mullikin JC, Mortimore BJ, Willey DL. A map of human genome sequence variation containing 142 million single nucleoide polymorphisms. Nature. 2001;409:928–33.
Saito K, et al. Regulatory approaches to genome-edited soybeans in Japan. J Crop Sci Biotechnol. 2023;26(1):85–92.
Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol. 2014;32(4):347–55.
Sandel MJ. The case against perfection: ethics in the age of genetic engineering. Harvard: Harvard University Press; 2016.
Sarinelli JM, Murphy JP, Tyagi P, Holland JB, Johnson JW, Mergoum M. Training population selection and use of fixed effects to optimize genomic predictions in a historical USA winter wheat panel. Theor Appl Genet. 2019;132:1247–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/S00122-019-03276-6.
Schmid-Burgk JL, Gao L, Li D, Gardner Z, Strecker J, Lash B, Zhang F. Highly parallel profiling of Cas9 variant specificity. Mol Cell. 2020;78(4):794–800.
Schornack S, Meyer A, Römer P, Jordan T, Lahaye T. Gene-for-gene-mediated recognition of nuclear-targeted AvrBs3-like bacterial effector proteins. J Plant Physiol. 2006;163(3):256–72.
Septiningsih EM, Pamplona AM, Sanchez DL, Neeraja CN, Vergara GV, Heuer S, Ismail AM, Mackill DJ. Development of submergence-tolerant rice cultivars: the Sub1 locus and beyond. Ann Bot. 2009;103(2):151–60.
Sha G, Sun P, Kong X, Han X, Sun Q, Fouillen L, Zhao J, Li Y, Yang L, Wang Y, Gong Q. Genome editing of a rice CDP-DAG synthase confers multipathogen resistance. Nature. 2023;14:1–7.
Shahinnia F, Geyer M, Schürmann F, Rudolphi S, Holzapfel J, Kempf H, Stadlmeier M, Löschenberger F, Morales L, Buerstmayr H, Sanchez JIY. Genome-wide association study and genomic prediction of resistance to stripe rust in current Central and Northern European winter wheat germplasm. Theor Appl Genet. 2022;135(10):3583–95.
Sharma H, Bhandawat A, Rahim MS, Kumar P, Choudhoury MP, Roy J. Novel intron length polymorphic (ILP) markers from starch biosynthesis genes reveal genetic relationships in Indian wheat varieties and related species. Mol Biol Rep. 2020;47:3485–500.
Shan Q, Wang Y, Li J, Zhang Y, Chen K, Gao C. Targeted genome modification of crop plants using CRISPR/Cas9. Nat Biotechnol. 2013;31(8):686–8.
Sim SC, Durstewitz G, Plieske J, Wieseke R, Ganal MW, Van Deynze A, Hamilton JP, Buell CR, Causse M, Wijeratne S, Francis DM. Development of a large SNP genotyping array and generation of high-density genetic maps in tomato. PLoS ONE. 2012;7(7): e40563.
Sinha S, Singh S, Kumar M, Singh RS, Satyendra, Thakur D. Recent Advancements in Molecular Marker Technologies and Their Applications in Crop Improvement. InMolecular Marker Techniques: A Potential Approach of Crop Improvement. 2023; (pp. 319–337). Singapore: Springer Nature Singapore.
Singh S, et al. Regulatory review of CRISPR-edited Canola in Canada. Plant Biotechnol J. 2020;18(4):775–84.
Smithies O, Gregg RG, Boggs SS, Koralewski MA, Kucherlapati RS. Insertion of DNA sequences into the human chromosomal β-globin locus by homologous recombination. Nature. 1985;317(6034):230–4.
Smith SJ, et al. Genomic selection in corn breeding: strategies and applications. Theor Appl Genet. 2013;126(2):253–64.
Sobrino B, Brión M, Carracedo A. SNPs in forensic genetics: a review on SNP typing methodologies. For Sci Int. 2005;154(2–3):181–94.
Song Q, Hyten DL, Jia G, Quigley CV, Fickus EW, Nelson RL, Cregan PB. Development and evaluation of SoySNP50K, a high-density genotyping array for soybean. PLoS ONE. 2013;8(1): e54985.
Sonah H, Deshmukh RK, Singh VP, Gupta DK, Singh NK, Sharma TR. Genome-wide distribution and organization of microsatellites and simple sequence repeats in plants: Comparative and evolutionary insights. Plant Mol Biol Report. 2013;31(3):915–27.
Sprink T, Wilhelm R, Hartung F. Genome editing around the globe: an update on policies and perceptions. Plant Physiol. 2022;190(3):1579–87.
Sunyaev S, Hanke J, Aydin A, Wirkner U, Zastrow I, Reich J, Bork P. Prediction of nonsynonymous single nucleotide polymorphisms in human disease-associated genes. J Mol Med. 1999;77:754–60.
Tan C, Yang Y. Penta-Primer Amplification Refractory Mutation System (PARMS) with Direct PCR-Based SNP Marker-Assisted Selection (D-MAS). InPlant Genotyping: Methods and Protocols. 2023; (pp. 327–336). New York, NY: Springer US.
Tian F, Bradbury PJ, Brown PJ, Hung H, Sun Q, Flint-Garcia S, Rocheford TR, McMullen MD, Holland JB, Buckler ES. Genome-wide association study of leaf architecture in the maize nested association mapping population. Nat Genet. 2011;43(2):159–62.
Tibbs‐Cortes LE, Guo T, Li X, Tanaka R, Vanous AE, Peters D, Gardner C, Magallanes‐Lundback M, Deason NT, DellaPenna D, Gore MA. Genomic prediction of tocochromanols in exotic‐derived maize. The Plant Genome. 2022; p.e20286.
Tinker NA, Chao S, Lazo GR, Oliver RE, Huang YF, Poland JA, Jellen EN, Maughan PJ, Kilian A, Jackson EW. A SNP genotyping array for hexaploid oat. The Plant Genome. 2014; 7(3): plantgenome2014–03.
Tou CJ, Orr B, Kleinstiver BP. Precise cut-and-paste DNA insertion using engineered type VK CRISPR-associated transposases. Nat Biotechnol. 2023;2:1–2.
Van der Oost J, Patinios C. The genome editing revolution. Trends Biotechnol. 2023;41(3):396–409.
Van K, McHale LK. Meta-analyses of QTLs associated with protein and oil contents and compositions in soybean [Glycine max (L.) Merr.] seed. Int J Mol Sci. 2017;18(6):1180.
Varshney RK, Bohra A, Yu J, Graner A, Zhang Q, Sorrells ME. Designing future crops: genomics-assisted breeding comes of age. Trends Plant Sci. 2021;26(6):631–49.
Varshney RK, Thudi M, Kumar A. Advancements in genomics tools for plant breeding and their applications. Nat Rev Genet. 2021;22(9):597–610.
Wang Q, Alariqi M, Wang F, Li B, Ding X, Rui H, Li Y, Xu Z, Qin L, Sun L, Li J. The application of a heat-inducible CRISPR/Cas12b (C2c1) genome editing system in tetraploid cotton (G. hirsutum) plants. Plant Biotechnol J. 2020;18(12):2436–43.
Wang W, Wang W, Pan Y, Tan C, Li H, Chen Y, Liu X, Wei J, Xu N, Han Y, Gu H. A new gain-of-function OsGS2/GRF4 allele generated by CRISPR/Cas9 genome editing increases rice grain size and yield. Crop J. 2022;10(4):1207–12.
Wang X, Zhao X, Zhu J, Wu W. Genome-wide investigation of intron length polymorphisms and their potential as molecular markers in rice (Oryza sativa L.). DNA Res. 2005;12(6):417–27.
Wang Y, Cao Y, Liang X, Zhuang J, Wang X, Qin F, Jiang C. A dirigent family protein confers variation of Casparian strip thickness and salt tolerance in maize. Nat Commun. 2022;13(1):2222.
Wang Y, Cheng X, Shan Q, Zhang Y, Liu J, Gao C, Qiu JL. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat Biotechnol. 2014;32(9):947–51.
Wang X, et al. Genetic diversity and trait association in Brassica napus using SSR markers. Theor Appl Genet. 2023;136(3):345–57.
Wang S, et al. CRISPR/Cas9-mediated genome editing of wheat genes for resistance to wheat blast. J Integr Plant Biol. 2021;63(7):1286–300.
Wang J, Zhang H, Wu W, Lu Y, Zhao X, Yang T, Zhang Z. Development of SSR markers for genetic diversity assessment and breeding applications in wheat. Front Plant Sci. 2021;12: 699935.
Wang K, et al. Editing of the tomato genome using CRISPR/Cas9 to improve fruit quality. Plant J. 2018;93(4):541–50.
Wenzl P, Carling J, Kudrna D, Jaccoud D, Huttner E, Kleinhofs A, Kilian A. Diversity arrays technology (DArT) for whole-genome profiling of barley. Proc Natl Acad Sci. 2004;101(26):9915–20.
Xu Y, Zhang XQ, Harasymow S, Westcott S, Zhang W, Li C. Molecular marker-assisted backcrossing breeding: an example to transfer a thermostable β-amylase gene from wild barley. Mol Breeding. 2018;38:1–9.
Xu Y, et al. High-throughput genotyping by PARMS for precision breeding. Plant Biotechnol J. 2020;18(8):1484–90.
Yabe S, Yoshida H, Kajiya-Kanegae H, Yamasaki M, Iwata H, Ebana K. Description of grain weight distribution leading to genomic selection for grain-filling characteristics in rice. PLoS ONE. 2018;13: e0207627. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/JOURNAL.PONE.0207627.
Yang W, Chen S, Cheng Y, Zhang N, Ma Y, Wang W, Tian H, Li Y, Hussain S, Wang S. Cell wall/vacuolar inhibitor of fructosidase 1 regulates ABA response and salt tolerance in Arabidopsis. Plant Signal Behav. 2020;15(4):1744293.
Yang Y, Amo A, Wei D, Chai Y, Zheng J, Qiao P, Hu YG. Large-scale integration of meta-QTL and genome-wide association study discovers the genomic regions and candidate genes for yield and yield-related traits in bread wheat. Theor Appl Genet. 2021;134(9):3083–109.
Yin K, Han T, Xie K, Zhao J, Song J, Liu Y. Engineer complete resistance to cotton leaf curl multan virus by the CRISPR/Cas9 system in Nicotiana benthamiana. Phytopathol Res. 2019;1(1):1–9.
Yu G, Snowdon R, Friedt W, Ordon F. The utilization of molecular markers in plant breeding for disease resistance. Biotechnology. 2017;6(1):9–22.
Yu H, Xie W, Li J, Zhou F, Zhang Q. A whole-genome SNP array (RICE 6 K) for genomic breeding in rice. Plant Biotechnol J. 2014;12(1):28–37.
Yue E, Cao H, Liu B. OsmiR535, a potential genetic editing target for drought and salinity stress tolerance in Oryza sativa. Plants. 2020;9(10):1337.
Zafar K, Sedeek KE, Rao GS, Khan MZ, Amin I, Kamel R, Mukhtar Z, Zafar M, Mansoor S, Mahfouz MM. Genome editing technologies for rice improvement: progress, prospects, and safety concerns. Front Genom Editing. 2020;2:5.
Zhan J, Zhong J, Cheng J, Wang Y, Hu K. Map-based cloning of the APRR2 gene controlling green stigma in bitter gourd (Momordica charantia). Front Plant Sci. 2023;14:1128926.
Zhang A, Chen S, Cui Z, Liu Y, Guan Y, Yang S, Qu J, Nie J, Dang D, Li C, Dong X. Genomic prediction of drought tolerance during seedling stage in maize using low-cost molecular markers. Euphytica. 2022;218(11):154.
Zhang J, Gill HS, Brar NK, Halder J, Ali S, Liu X, Bernardo A, Amand PS, Bai G, Gill US, Turnipseed B. Genomic prediction of Fusarium head blight resistance in early stages using advanced breeding lines in hard winter wheat. The Crop Journal. 2022;10(6):1695–704.
Zhang P, Du H, Wang J, Pu Y, Yang C, Yan R, Yang H, Cheng H, Yu D. Multiplex CRISPR/Cas9-mediated metabolic engineering increases soya bean isoflavone content and resistance to soya bean mosaic virus. Plant Biotechnol J. 2020;18(6):1384–95.
Zhang R, Liu J, Chai Z, Chen S, Bai Y, Zong Y, Chen K, Li J, Jiang L, Gao C. Generation of herbicide tolerance traits and a new selectable marker in wheat using base editing. Nature plants. 2019;5(5):480–5.
Zhang Z, Ge X, Luo X, Wang P, Fan Q, Hu G. Simultaneous editing of two copies of Gh14-3-3d confers enhanced transgene-clean plant defense against Verticillium dahliae in allotetraploid upland cotton. Front Plant Sci. 2018;9:842. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fpls.2018.00842.
Zhang X, et al. Gene expression analysis in response to biotic stress using Bioconductor packages in Solanum tuberosum. Plant Physiol. 2013;163(4):1741–57.
Zhang Y, Malzahn AA, Sretenovic S, Qi Y. The emerging and uncultivated potential of CRISPR technology in plant science. Nature Plants. 2020;6(8):778–90.
Zhang L, Wang X, Li J, Huang Y. Application of inter small RNA polymorphism (iSNAP) markers in identifying disease resistance genes in tomato (Solanum lycopersicum). Mol Plant Microbe Int. 2023;36(7):865–77.
Zhou J, Li Z, Li Y, Zhao Q, Luan X, Wang L, Liu Y, Liu H, Zhang J, Yao D. Effects of different gene editing modes of CRISPR/Cas9 on soybean fatty acid anabolic metabolism based on GmFAD2 family. Int J Mol Sci. 2023;24(5):4769.
Acknowledgements
Not applicable.
Funding
Not Applicable.
Author information
Authors and Affiliations
Contributions
All authors contributed to the study conception and design. RK and NRP conceptualized the manuscript topic. RK, AK, RLV, MC, NRP, SR, AGD, RD, BD, and HLD wrote the initial draft of the manuscript. RK, SPD, BUC, BB and VKM revised the manuscript. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
This submitted work has not been published before. It is not under consideration for publication elsewhere. Its publication has the consent of all the authors and the responsible authorities where the work was carried out. If accepted, it will not be published elsewhere in the same form, in English, or in any other language without the written consent of the copyright holder.
Consent for publication
The authors approved this version of the manuscript to publish.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Kumar, R., Das, S.P., Choudhury, B.U. et al. Advances in genomic tools for plant breeding: harnessing DNA molecular markers, genomic selection, and genome editing. Biol Res 57, 80 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40659-024-00562-6
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40659-024-00562-6