Next Article in Journal
Using High-Throughput Phenotyping Analysis to Decipher the Phenotypic Components and Genetic Architecture of Maize Seedling Salt Tolerance
Next Article in Special Issue
Molecular Markers Help with Breeding for Agronomic Traits of Spring Wheat in Kazakhstan and Siberia
Previous Article in Journal
Comparative Mitogenome Analysis of Two Native Apple Snail Species (Ampullariidae, Pomacea) from Peruvian Amazon
Previous Article in Special Issue
Genetic Diversity in Natural Populations of Rhodiola Species of Different Adaptation Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Enhancement of Cereals Using Genomic Resources for Nutritional Food Security

by
Neeraj Chaudhary
1,
Romesh Kumar Salgotra
1 and
Bhagirath Singh Chauhan
2,*
1
School of Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, Chatha, Jammu 180009, Jammu and Kashmir, India
2
Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Gatton, QLD 4343, Australia
*
Author to whom correspondence should be addressed.
Genes 2023, 14(9), 1770; https://doi.org/10.3390/genes14091770
Submission received: 16 August 2023 / Revised: 4 September 2023 / Accepted: 6 September 2023 / Published: 7 September 2023

Abstract

:
Advances in genomics resources have facilitated the evolution of cereal crops with enhanced yield, improved nutritional values, and heightened resistance to various biotic and abiotic stresses. Genomic approaches present a promising avenue for the development of high-yielding varieties, thereby ensuring food and nutritional security. Significant improvements have been made within the omics domain, specifically in genomics, transcriptomics, and proteomics. The advent of Next-Generation Sequencing (NGS) techniques has yielded an immense volume of data, accompanied by substantial progress in bioinformatic tools for proficient analysis. The synergy between genomics and computational tools has been acknowledged as pivotal for unravelling the intricate mechanisms governing genome-wide gene regulation. Within this review, the essential genomic resources are delineated, and their harmonization in the enhancement of cereal crop varieties is expounded upon, with a paramount focus on fulfilling the nutritional requisites of humankind. Furthermore, an encompassing compendium of the available genomic resources for cereal crops is presented, accompanied by an elucidation of their judicious utilization in the advancement of crop attributes.

1. Introduction

The basic requisites furnished by food encompass proteins, minerals, carbohydrates, fiber, vitamins, lipids (macronutrients), as well as essential micronutrients, all of which furnish the energy imperative for bodily growth, function, and defense [1]. Belonging to the Poaceae family, all cereal crop species share this familial lineage, making it the fourth largest family of flowering plants [2]. The principal cereal crops, wheat, rice, maize, sorghum, finger millet, foxtail millet, barley, and other such counterparts, stand as pivotal sources of nourishment worldwide [3]. These grain cereals form the cornerstone of staple diets across many regions, and their utility extends beyond sustenance to encompass starch, animal feed, oils, sugar, and processed consumables, such as malts and alcoholic beverages. An impressive 50% of global calories stem from maize and wheat, while regions in Africa and Asia prominently rely on grains like sorghum and millets. As the global populace burgeons and standards of living surge, the demand for ample, high-quality, and diversified sustenance escalates, rendering the task of feeding an anticipated population of 10 billion by 2050 a formidable challenge.
The impending prospect of nourishing 10 billion individuals by 2050 looms as one of agriculture’s paramount challenges [4]. Concurrently, the specter of climate change further imperils food security, predicting heightened severity in the years ahead. Consequently, the augmentation of cereal crops to manifest enhanced productivity and climate resilience is imperative. Unveiling the genetic underpinnings of these traits assumes paramount importance, and herein lies the crux and promise of genetic and genomic strategies. These approaches are instrumental in delineating the genetic determinants responsible for these traits, further enabling their characterization and targeted integration into trait enhancement initiatives. Hence, genetics and genomics emerge as pivotal tools for dissecting the intricate tapestry of traits encompassing growth, development, and responses to environmental pressures. Unquestionably, the cereal research community has harnessed these tools to investigate crucial attributes inherent in cereal crops.
Anticipating that the world’s population will swell to around 11.2 billion by 2100 necessitates not only a surge in food production but also the establishment of secure and sustainable food systems, all while leveraging finite natural resources [5,6,7,8,9,10,11,12]. This challenge is intensified by the dwindling expanse of cultivable land, a consequence of industrialization and burgeoning infrastructure.
Cereal crops collectively constitute edible seed grains for global consumption, offering vital dietary supplements, such as vitamin E, vitamin B, magnesium, and zinc [13]. Among these, rice, wheat, and maize command a staggering 90% share of global cereal production, leaving the remaining 10% to encompass minor cereals like millets, barley, oats, and rye [14]. The advent of the Green Revolution in the 1960s bore witness to escalated yields of the two primary cereals, wheat and rice, curtailing food grain scarcities [15]. Over time, these predominant varieties were further optimized for enhanced yield and other agronomic traits, resulting in sustainable yields that could cater to growing nutritional demands. However, a protracted disregard for the nutritional components of these crops left a substantial void, ultimately fostering malnutrition in approximately one out of every three individuals worldwide [16]. Although traditional plant breeding played a pivotal role in improving cereal traits, addressing intricate attributes like enhanced selenium uptake in wheat to counter abiotic stresses, such as drought and heat tolerance, a noticeable gap remained in nutritional richness. Thus, the imperative to bridge this nutritional divide and ensure sustainable food security underscores the necessity of fortifying cereals using advanced genomic resources for genetic manipulations of major cereal crops.
The practice of fortifying cereal crops with key nutrients, such as vitamins (B, C, D, and E), iron, β-carotene, and zinc, has gained prominence as a strategy for augmenting their nutritional value [17]. Instances of successful nutritional enhancement employing genomic techniques and resources abound, encompassing endeavors, such as elevating the oligosaccharide and polysaccharide levels, enriching the iron content, boosting the vitamin E concentrations in maize, and engendering rice varieties replete with β-carotene and iron [18,19,20,21].
As the basis of future crop breeding, genetic resources stand as pivotal to future food security. Contemporary methods for germplasm characterization and assessment have yielded commendable results in managing crop genetic resources effectively. In parallel, the utilization of genomic resources and specialized germplasm subsets, including mini-core collections and reference sets, facilitates the identification of trait-specific germplasm, trait mapping, and allele mining, not only for resistance to diverse biotic and abiotic pressures, but also for beneficial agro-morphological traits.
The revolution in cereal genomics was inaugurated by the unveiling of draft genome sequences for rice, subsequently followed by sorghum, maize, pearl millet, barley, and wheat [22,23,24,25,26,27,28]. This repository of genome sequences, accompanied by an array of genomic resources, advanced mapping populations, and panels, has propelled genomics-assisted breeding within the realm of cereals [29]. These advancements have significantly advanced the exploration of the gene function vis-à-vis the target phenotypes.
Within the annals of botanical knowledge, an abundance of research material delves into the molecular and cellular mechanisms of rice, a model plant for cereal counterparts [30]. The intricacies of signal responses to sundry stresses in crops often hinge upon gene regulation. In this context, contemporary genomic resources, including molecular markers, whole-genome sequences, sequencing-based trait mapping, and germplasm sequencing, facilitate the exploration of gene regulation at the molecular level [31]. Despite tremendous leaps in genomics, avenues, such as transcriptomic and proteomic profiling, remain to be explored fully. The creation of novel crops endowed with enhanced resistance to diverse biotic and abiotic stresses, as well as other nutritional traits, stands as an essential pursuit. A wealth of stress-responsive genes is activated, engendering proteins that orchestrate physiological and biochemical pathways integral to stress tolerance [30,32]. Thus, the implication of genomic approaches with the ambit of systems biology augments our comprehension of intricate mechanisms and charts a course for future endeavors [33,34,35,36]. This review endeavors to elucidate recent strides in functional genomics, transcriptomic analyses, and proteomic investigations, shedding light on the mechanisms orchestrating stress tolerance and nutritional enhancement within diverse cereal crops. A synthesis of the transcriptomic, proteomic, and functional genomic approaches pertinent to crop advancement is succinctly portrayed (Figure 1).

2. Genomic Resources

In the pursuit of substantial advancements across various crop species, genomic tools have emerged as the cornerstone, offering essential inputs to breeding endeavors. A pivotal role is played by genomic resources, including linkage and genetic maps, molecular markers, and sequence data, in enhancing molecular breeding programs [37]. As the scientific landscape ascends to new heights, propelled by genomic paradigms and Next-Generation Sequencing (NGS), the analysis of genomes has reached unprecedented dimensions. NGS has permeated diverse domains, encompassing whole-genome sequencing (WGS), whole-genome resequencing (WGRS), de novo sequencing, transcriptomic analyses, genotyping-by-sequencing (GBS), and epigenetic research. These resources have culminated in the creation of several PCR-based markers. Among these markers, namely inter-simple sequence repeats (ISSRs), simple sequence repeats (SSRs), single-nucleotide polymorphisms (SNPs), and diversity array technologies (DArTs), are frequently harnessed to foster crop improvement [38]. While SSR markers find frequent utility in crop breeding [39], ISSRs excel in gauging genetic variation, conducting diversity analyses, and facilitating the molecular breeding of crop plants, owing to their elevated polymorphism levels. Dominating the marker landscape are SNPs, cherished for their facile availability, high-performance genotyping capabilities, and cost effectiveness [25]. The DArT marker system has, notably, surfaced as a robust tool for uncovering genetic diversity within germplasm collections and holds promise in molecular breeding studies [40]. The confluence of these resources expedites the identification of genes/QTLs and paves a well-defined trajectory toward molecular breeding, bolstering crop productivity and ensuring food security [25]. It is worth noting that the resolution of genetic variation closely aligns with the number of polymorphisms detected through the marker system [41].
Historically, the bulk of genomic and molecular research has concentrated on cereal crops, driven by the imperative of global food production [31]. However, the proliferation of NGS techniques has broadened the scope to encompass other crops, illuminating their capacities and prominence. Novel NGS platforms have facilitated the generation of expansive sequence data, giving rise to diverse genomic resources, such as restriction fragment length polymorphisms (RFLP) [42], randomly amplified polymorphic DNA (RAPD) [43], SSRs [44], cleaved amplified polymorphic sequences (CAPS) [45,46], amplified fragment length polymorphisms (AFLP) [47], SNPs [48], and DArT markers [49] (Figure 2). These molecular markers prove instrumental in diverse molecular studies, spanning genetic map construction, indirect selection, and the introgression or pyramiding of quantitative trait loci (QTLs) or genes into elite varieties via molecular breeding strategies like marker-assisted selection (MAS), marker-assisted backcross breeding (MABC), and genomic selection (GS). Notably, genome-wide association studies (GWAS), SNP arrays, and transcriptomic, metagenomic, epigenomic, and gene expression data have accelerated breeding cycles across multiple crops [50,51].
With the advent of high-throughput NGS techniques, genomics has empowered biotechnologists to sequence the genomes of nearly all major cereal crops, enabling the identification of genes/QTLs and facilitating the incorporation of desirable traits into diverse crops [52]. Consequently, researchers are inclined to explore gene upregulation/downregulation and analyze global shifts in gene expression by pinpointing corresponding RNAs. Within transcriptomics, the panorama of gene expression dynamics and functionality is unveiled. The realm of transcriptomics, replete with diverse coding and non-coding RNA molecules, bears witness to remarkable complexity [53]. Thanks to cost-effective, Sanger, and high-throughput NGS technologies, the field of transcriptomics has undergone a paradigm shift, post the completion of genome sequencing projects across various crops (Table 1) [30]. A plethora of sequence data has been generated by the sequencing of important cereal crops. This surge in knowledge has paved the way for downstream investigations in the domains of post-transcriptional and post-translational events [30]. Transcriptomic studies adopt a multitude of techniques, including microarrays, expressed sequence tags (ESTs), serial analysis of gene expression (SAGE), cap analysis of gene expression (CAGE), RNA-Seq/NGS, and de novo assembly. The microarray technique, reliant on hybridization, presents an economical and high-throughput approach, and played a pivotal role in pioneering transcriptomic research [54]. Utilizing a microarray, a DNA chip arrays multiple DNA fragments (probes/reporters) on small glass slides, offering an automated platform for parallel DNA sequence analysis. Thousands of labeled spots containing single-strand DNA/RNA molecules, labeled with fluorescent dye, populate the microarray glass slide. Upon introduction of a solution containing ssDNA or RNA fragments (targets), hybridization transpires, yielding fluorescence. This fluorescence is subsequently scanned and analyzed, furnishing data for further exploration. Presently, two primary microarray types have gained prominence: short oligonucleotide-based and cDNA-based microarrays. The application of DNA microarrays encompasses gene expression profiling, comparative genomic hybridization, SNP detection, and the detection of alternative splicing [55].
The domain of microarray study presents both quantity and quality challenges in regard to data acquisition. Degraded mRNAs can yield false microarray data, and sensitivity to the cross-hybridization of probes with non-target sequences can generate misleading outcomes [56]. The presence of high background noise due to sequence similarity can further complicate result determination. The specificity of probes in DNA microarrays is also influenced by their lengths, with longer probes exhibiting reduced specificity [57]. Additionally, microarray techniques exhibit a limited dynamic range of detection due to both background and saturation signals, necessitating prior knowledge of the sequences under examination [58,59].
Subsequent to microarray techniques, sequence-based methodologies emerged to provide a more comprehensive elucidation of the transcriptome by directly determining the transcript sequences. Complementary DNA (cDNA) libraries are constructed from isolated mRNAs in a manner that allows identification of the 5′ and 3′ ends of the message through sequencing the vector–insert joins in the plasmid. This approach, such as expressed sequence tags (ESTs) generation, facilitates the rapid and easy discovery of new genes, crucial for functional genomics and understanding the molecular mechanisms underpinning sustainable agricultural production [59,60]. However, limitations include the detection of predominantly abundant transcripts, relatively low throughput, and the inability to quantify transcripts [61]. The NCBI’s dbEST stands as a specialized division of GenBank, encompassing publicly available EST collections from various organisms [62].
To surmount the limitations posed by ESTs, tag-based methods such as serial analysis of gene expression (SAGE) and cap analysis of gene expression (CAGE) were developed. SAGE quantitatively estimates mRNA expression levels by measuring short (10–14 mer) sequences of transcribed messages, using them to infer specific transcripts. SAGE aids in detecting and quantifying gene activities in desired cells, providing insights into discovering novel genes. CAGE, conversely, collects 21 bp tags from the 5′ end of cDNAs to ascertain the 5′ end of transcripts and promoter locations [63]. While these techniques yield improved results, challenges persist, including splice isoforms, the inability to discover new genes, the requirement for a significant amount of RNA, labor-intensive procedures, and high sequencing costs. Nonetheless, the advent of high-throughput Next-Generation Sequencing (NGS) technologies, notably RNA sequencing (RNA-Seq), has effectively addressed these limitations, revealing transcriptomes with heightened sensitivity and accuracy [64,65].
RNA-Seq, a form of high-throughput NGS, sequences the RNA of a species that precisely determines each transcript, achieving remarkable sequencing depth [53]. This technique has surmounted previous limitations, elucidating molecular functions through single-cell gene expression and aiding in the identification of novel genes/QTLs [53,59]. NGS has elevated the scale of transcriptome sequencing, broadened sequence-based marker resources, and enabled de novo assembly for crop development, all at a reduced cost compared to phenotyping [66]. This approach also affords a high-resolution view of transcription, alternative splicing, and allele-specific expression. However, RNA-Seq analysis introduces computational challenges unique to this type of sequencing-based investigation [67].
The generation of diverse genomic resources through various technologies has precipitated the genomic revolution [31]. The application of these genomic resources accelerates breeding and other crop improvement programs. These resources furnish the foundation for the efficient utilization of genetic resources in the enhancement of cereals and other crops. For genetic improvement, aside from sequencing data, an array of genetic resources and biparental populations like doubled haploids (DHs), F2 or F2-derived F3 populations, recombinant inbred lines (RILs), backcrosses, near-isogenic lines (NILs), nested association mapping (NAM), and multiparent advanced generation inter-cross (MAGIC) populations are essential for linking gene(s)/QTLs to specific traits. The identified marker–trait associations can then be harnessed for crop enhancement or development using marker-assisted recurrent selection (MARS), marker-assisted backcross breeding (MABB), and genomic selection (GS) approaches [37,68].
Table 1. Important cereal crops sequenced using Sanger and NGS technologies.
Table 1. Important cereal crops sequenced using Sanger and NGS technologies.
CropBotanical NameGenome Size (Mb) and Sequencing MethodReference
Rice Oryza sativa ssp. japonica (Nipponbare)
Oryza sativa ssp. japonica (Nipponbare)
Oryza sativa ssp. Indica
(i)
420 Mb; Sanger, Whole-Genome Sequencing (WGS)
(ii)
389 Mb; Sanger, Bacterial Artificial Chromosome (BAC-by-BAC)
(iii)
466 Sanger, Whole-Genome Sequencing (WGS)
[22]
[69]
[70]
Maize Zea mays (Palomero Toluqueno) (popcorn)
Zea mays (B73)
(i)
2100 Mb; Sanger, Whole-Genome Sequencing (WGS)
(ii)
2300 Mb; Sanger, Bacterial Artificial Chromosome (BAC-by-BAC)
[71]
[24]
SorghumSorghum bicolor (L.) Moench 730 Mb; Sanger, Whole-Genome Sequencing (WGS)[23,72]
Foxtail milletSetaria italica515 Mb; Illumina, Whole-Genome Sequencing (WGS)[73]
Bread wheatTriticum aestivum17,000 Mb; 454, Whole-Genome Sequencing (WGS) [74]
BarleyHordeum vulgare5100 Mb; 454, Bacterial Artificial Chromosome (BAC-by-BAC) [26,75]
Finger milletEleusine coracana1200 Mb; Illumina, Whole-Genome Sequencing (WGS)[76,77]
Pearl milletCenchrus americanus1800 Mb; Illumina, Whole-Genome Sequencing (WGS) [25]
Proso milletPanicum miliaceum923 Mb; Illumina short-read coupled with Pac-Bio long-read sequencing [78]
Barnyard milletEchinochloa esculenta1.27 Mb; Illumina HiSeq platform [79]

3. Genomic Resources and Their Implications

Genomic resources play a crucial role in characterizing essential genes within a species, employing diverse approaches including structural and functional analyses, QTL/linkage mapping, and gene editing. These techniques have facilitated the genetic enhancement of cereal crops by deciphering complex trait architectures. Utilizing genomic resources in molecular breeding programs has effectively amplified cereal grain yield and crop productivity. Additionally, these resources optimize the utilization of genetic assets across different crop varieties (Figure 3).
Genomic resources have led to the identification and tagging of novel molecular markers through genome-wide association studies (GWAS), linking them to valuable agronomic and physiological traits. Furthermore, QTL mapping has become an instrumental approach for marker-assisted breeding (MAB), facilitating trait improvement and introgression. The downstream application of developed genomic resources has empowered plant breeders to introduce pivotal genes into various crops, particularly cereals. Notably, genomic resources have revolutionized the identification of significant genes/QTLs and their integration into diverse cereal crops.
This integration of crucial genes/QTLs has resulted in improved iterations of crop varieties that exhibit tolerance to environmental stresses. As a consequence, these enhanced varieties boast augmented yields and nutritional value. This substantial genetic progress is evident in crops such as rice, wheat, maize, and barley. In sum, genomic resources have significantly contributed to the genetic enhancement of multiple cereal crop traits [80,81].
In harnessing the genetic diversity present in cereal crop germplasm, genomic resources have played a pivotal role. For instance, in breeding programs, the inclusion of diverse genotypes in crossings is essential. Genomic resources prove invaluable in characterizing and identifying diverse parent candidates at the molecular level. Among various molecular breeding strategies, genomic selection (GS) has emerged as particularly fitting for improving intricate traits [82]. GS quantifies the collective impact of genome-wide markers to estimate the cumulative effects of all loci, yielding genomic estimated breeding values (GEBVs) that facilitate precise selection [83,84]. GS effectively captures alleles/QTLs governing traits with modest, heritable effects [85,86].
The following examples illustrate the deployment of essential genes/QTLs in grain cereal crops through methods such as MABC/MABB and GS, utilizing an array of genomic resources (Table 2).
Genomic resources have been pivotal in enhancing important agronomic traits via MABB. Examples include the TaSUS2-2B and TaZds-D1 genes impacting grain weight [81], the Rht1 and Rht2 semi-dwarfing genes in wheat germplasm [115], and genes like Lr34, Yr18, Pm38, and Sr45 enhancing disease resistance to rust and powdery mildew in wheat [118,119,120]. Additionally, genes like Psy1, Glu-B3, and GS3 influence traits such as yellow grain pigment, low gluten protein, and longitudinal grain elongation, respectively [121,122,123]. Similarly, Wx gene encoded intermediate amylose content in rice [124], Dwarf 8 gene for reduced plant height and early flowering in maize [125], and the sugary1 gene for sweetness in maize [126]. Other instances encompass genes for bacterial blight resistance and sweetness in rice, as well as genes governing traits in maize and barley [84,85,86].
The availability of genomic resources is pivotal for enhancing cereal and broader crop improvement endeavors. These resources not only facilitate the transfer of vital genes through molecular breeding, but also support studies on genetic diversity, genotype identification, population structure, and phylogenetics [127,128]. Multiple breeding approaches, including MAS, MABB, MARC, and GS, are leveraged for enhancing crop varieties to resist biotic and abiotic stresses, along with improving quality traits. The integration of genes/QTLs into diverse crop species through genomic resources has spawned the development and commercialization of various crop varieties, such as Improved Pusa Basmati 1, Improved Samba Mahsuri, Pusa 1121, Improved Tapaswini, and Ranbir Basmati [84,129,130,131,132]. Furthermore, numerous rice varieties and hybrid parental lines have been improved for resistance to bacterial blight (xa5, xa13, and Xa21), either independently or combined with genes/QTLs for other stresses and quality attributes [133].

4. Conclusions and Future Perspectives

The accessibility of genomic resources in conjunction with genotyping platforms has emerged as a cornerstone in advancing cereal crop varieties. Plant breeders are now empowered to unlock the genetic potential of cereal crop varieties by orchestrating native genes in various permutations through the utilization of diverse genomic resources. While molecular breeding techniques have been effectively harnessed for prominent cereal crops like rice, wheat, maize, and barley, considerable potential remains untapped for lesser-explored cereal varieties, such as sorghum, finger millet, and foxtail millet. A crucial avenue lies in establishing diverse bi-parental mapping populations to identify marker–trait associations in these underexplored crops.
In the future, the evolution of high-throughput genotyping and phenotyping techniques promises a comprehensive understanding of gene function and marker–trait correlations, enriching our capacity to further refine crop varieties. The advancement in genomic resources has revolutionized the enhancement of complex traits, a task previously arduous. Single-nucleotide polymorphism (SNP) markers have emerged as the preferred molecular markers, and the conversion of identified SNPs to cleaved amplified polymorphic sequences (CAPs) or KASPar assays, along with Illumina VeraCode, is poised to expand their application in cereal crop improvement programs.
The prospects for developing a plethora of genomic resources remain promising. The sequencing of genomes within and across species holds the potential to yield numerous novel resources. As we look ahead, the genetic enhancement of cereal crop varieties can be propelled through the integration of superior haplotypes into these varieties. Haplotype-based breeding approaches can pave the way for further advancements in cereal crop improvement.
In summary, the availability and utilization of genomic resources, coupled with innovative genotyping technologies, have ushered in a new era of cereal crop breeding. These resources not only refine existing crops, but also harbor the potential to rejuvenate neglected cereal varieties. As we continue to delve deeper into the realm of genomics, the canvas for cereal crop enhancement expands, promising increased yield, resilience, and nutritional value to meet the challenges of a growing global population and a changing environment.

Author Contributions

Conceptualization, C.N., S.R.K. and C.B.S.; writing—original draft preparation, C.N.; writing—review and editing, S.R.K. and C.B.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data has been included in the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tasgin, E. Macronutrients and micronutrients in nutrition. Int. J. Innov. Res. Rev. 2017, 1, 10–15. [Google Scholar]
  2. Salse, J.; Feuillet, C. Comparative genomics of cereals. In Genomics-Assisted Crop Improvement; Springer Science+Business Media: New York, NY, USA, 2007; pp. 177–205. [Google Scholar]
  3. Lata, C.; Shivhare, R. Engineering cereal crops for enhanced abiotic stress tolerance. Proc. Indian Natl. Sci. Acad. 2021, 87, 63–83. [Google Scholar] [CrossRef]
  4. Sharma, M.; Kishore, A.; Roy, D.; Joshi, K. A comparison of the Indian diet with the EAT-Lancet reference diet. BMC Public Health 2020, 20, 812. [Google Scholar] [CrossRef]
  5. Nawaz, M.A.; Chung, G. Genetic improvement of cereals and grain legumes. Genes 2020, 11, 1255. [Google Scholar] [CrossRef]
  6. Campbell, B.M.; Vermeulen, S.J.; Aggarwal, P.K.; Corner-Dollo, C.; Girvetz, E.; Loboguerrero, A.M.; Ramirez-Villegas, J.; Rosenstock, T.; Sebastian, L.; Thornton, P.K. Reducing risks to food security from climate change. Glob. Food Secur. 2016, 11, 34–43. [Google Scholar] [CrossRef]
  7. UNO United Nations Organization, Department of Economic and Social Affairs, Population Division. World Population Prospects 2019: Highlights. 2019. Available online: https://population.un.org/wpp/publications/files/wpp2019_highlights.pdf (accessed on 29 June 2023).
  8. Godfray, H.C.J.; Beddington, J.R.; Crute, I.R.; Haddad, L.; Lawrence, D.; Muir, J.F. Food security: The challenge of feeding 9 billion people. Science 2010, 327, 812–818. [Google Scholar] [CrossRef]
  9. Massawe, F.; Mayes, S.; Cheng, A. Crop diversity: An unexploited treasure trove for food security. Trends Plant Sci. 2016, 21, 365–368. [Google Scholar] [CrossRef]
  10. Varshney, R.K.; Bohra, A.; Yu, J.; Graner, A.; Zhang, Q.; Sorrells, M.E. Designing future crops: Genomics-assisted breeding comes of age. Trends Plant Sci. 2021, 26, 631–649. [Google Scholar] [CrossRef]
  11. Salgotra, R.K.; Sood, M.; Jasrotia, M. Underutilized Crops and Their Value Addition; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2021; p. 11788. [Google Scholar]
  12. McKevith, B. Nutritional aspects of cereals. Nutr. Bull. 2004, 29, 111–142. [Google Scholar] [CrossRef]
  13. Salgotra, R.K.; Gupta, M. Exploring the role of epigenetics in cereal and leguminous crops exposed to abiotic stress. In Epigenetics in Plants of Agronomic Importance: Fundamentals and Applications; Transcriptional Regulation and Chromatin Remodeling in Plants; Springer: Cham, Switzerland, 2019; pp. 149–170. [Google Scholar]
  14. Eliazer Nelson, A.R.L.; Ravichandran, K.; Antony, U. The impact of the Green Revolution on indigenous crops of India. J. Ethn. Foods 2019, 6, 8. [Google Scholar] [CrossRef]
  15. Bouis, H.E.; Saltzman, A. Improving nutrition through biofortification: A review of evidence from HarvestPlus, 2003 through 2016. Glob. Food Secur. 2017, 12, 49–58. [Google Scholar] [CrossRef]
  16. World Health Organization. Global Nutrition Report 2017: Nourishing the SDGs; Development Initiatives: Bristol, UK, 2017. [Google Scholar]
  17. Lyons, G.; Stangoulis, J.; Graham, R. High-selenium wheat: Biofortification for better health. Nutr. Res. Rev. 2003, 16, 45–60. [Google Scholar] [CrossRef]
  18. Buttriss, J. Nutrition and Food Processing; British Nutrition Foundation: London, UK, 1999. [Google Scholar]
  19. Henry, R.J. Biotechnology. In Cereals Processing Technology; Owens, G., Ed.; Woodhead Publishing: Abington, UK, 2001; pp. 53–76. [Google Scholar]
  20. Khush, G.S. Challenges for meeting the global food and nutrient needs in the new millenium. Proc. Nutr. Soc. 2001, 60, 15–26. [Google Scholar] [CrossRef]
  21. Lucca, P.; Hurrell, R.; Potrytkus, I. Fighting iron deficiency with iron-rich rice. J. Am. Coll. Nutr. 2002, 21, 184S–190S. [Google Scholar] [CrossRef]
  22. Goff, S.A.; Ricke, D.; Lan, T.H.; Presting, G.; Wang, R.; Dunn, M.; Glazebrook, J.; Sessions, A.; Oeller, P.; Varma, H.; et al. A draft sequence of the rice genome (Oryza sativa L. ssp. japonica). Science 2002, 296, 92–100. [Google Scholar] [CrossRef]
  23. Paterson, A.H.; Bowers, J.E.; Bruggmann, R.; Dubchak, I.; Grimwood, J.; Gundlach, H.; Haberer, G.; Hellsten, U.; Mitros, T.; Poliakov, A.; et al. The Sorghum bicolor genome and the diversification of grasses. Nature 2009, 457, 551–556. [Google Scholar] [CrossRef] [PubMed]
  24. Schnable, P.S.; Ware, D.; Fulton, R.S.; Stein, J.C.; Wei, F.; Pasternak, S.; Liang, C.; Zhang, J.; Fulton, L.; Graves, T.A.; et al. The B73 maize genome: Complexity, diversity, and dynamics. Science 2009, 326, 1112–1115. [Google Scholar] [CrossRef]
  25. Varshney, R.K.; Shi, C.; Thudi, M.; Mariac, C.; Wallace, J.; Qi, P.; Zhang, H.; Zhao, Y.; Wang, X.; Rathore, A.; et al. Pearl millet genome sequence provides a resource to improve agronomic traits in arid environments. Nat. Biotechnol. 2017, 35, 969–976. [Google Scholar] [CrossRef]
  26. Mayer, K.F.; Waugh, R.; Langridge, P.; Close, T.J.; Wise, R.P.; Graner, A.; Matsumoto, T.; Sato, K.; Schulman, A.; Muehlbauer, G.J.; et al. A physical, genetic and functional sequence assembly of the barley genome. Nature 2012, 491, 711–716. [Google Scholar]
  27. Mascher, M.; Gundlach, H.; Himmelbach, A.; Beier, S.; Twardziok, S.O.; Wicker, T.; Radchuk, V.; Dockter, C.; Hedley, P.E.; Russell, J.; et al. A chromosome conformation capture ordered sequence of the barley genome. Nature 2017, 544, 427–433. [Google Scholar] [CrossRef]
  28. International Wheat Genome Sequencing Consortium (IWGSC); Appels, R.; Eversole, K.; Stein, N.; Feuillet, C.; Keller, B.; Rogers, J.; Pozniak, C.J.; Choulet, F.; Distelfeld, A.; et al. Shifting the limits in wheat research and breeding using a fully annotated reference genome. Science 2018, 361, eaar7191. [Google Scholar] [CrossRef]
  29. Mochida, K.; Shinozaki, K. Genomics and bioinformatics resources for crop improvement. Plant Cell Physiol. 2010, 51, 497–523. [Google Scholar] [CrossRef] [PubMed]
  30. Rakkammal, K.; Priya, A.; Pandian, S.; Maharajan, T.; Rathinapriya, P.; Satish, L.; Ceasar, S.A.; Sohn, S.-I.; Ramesh, M. Conventional and Omics Approaches for Understanding the Abiotic Stress Response in Cereal Crops—An Updated Overview. Plants 2022, 11, 2852. [Google Scholar] [CrossRef] [PubMed]
  31. Salgotra, R.K.; Stewart, N.C. Functional markers for precision plant breeding. Int. J. Mol. Sci. 2020, 21, 4792. [Google Scholar] [CrossRef] [PubMed]
  32. Pandian, S.; Rakkammal, K.; Rency, A.S.; Muthuramalingam, P.; Pandian, S.K.; Ramesh, M. Abiotic stress and applications of omics approaches to develop stress tolerance in agronomic crops. In Agronomic Crops: Volume 3: Stress Responses and Tolerance; 2020; pp. 557–578. [Google Scholar]
  33. Zhan, X.; Lu, Y.; Zhu, J.; Botella, J.R. Genome editing for plant research and crop improvement. J. Integr. Plant Biol. 2021, 63, 3–33. [Google Scholar] [CrossRef] [PubMed]
  34. Salgotra, R.K.; Gupta, B.B.; Monika, S. Biotechnological interventions and their role in sustainable hill agriculture. J. Plant Sci. Res. 2015, 2, 1–8. [Google Scholar]
  35. Paes de Melo, B.; de Carpinetti, P.A.; Fraga, O.T.; Rodrigues-Silva, P.L.; Fioresi, V.S.; de Camargos, L.F.; da Silva Ferreira, M.F. Abiotic stresses in plants and their markers: A practice view of plant stress responses and programmed cell death mechanisms. Plants 2022, 11, 1100. [Google Scholar] [CrossRef] [PubMed]
  36. Yaqoob, U.; Jan, N.; Raman, P.V.; Siddique, K.H.M.; John, R. Crosstalk between brassinosteroid signaling, ROS signaling and phenylpropanoid pathway during abiotic stress in plants: Does It exist? Plant Stress 2022, 4, 100075. [Google Scholar] [CrossRef]
  37. Salgotra, R.K.; Stewart, C.N. Genetic augmentation of legume crops using genomic resources and genotyping platforms for nutritional food security. Plants 2022, 11, 1866. [Google Scholar] [CrossRef]
  38. Manzoni, C.; Kia, D.A.; Vandrovcova, J.; Hardy, J.; Wood, N.W.; Lewis, P.A.; Ferrari, R. Genome, transcriptome and proteome: The rise of omics data and their integration in biomedical sciences. Brief. Bioinform. 2018, 19, 286–302. [Google Scholar] [CrossRef]
  39. Gupta, P.K.; Varshney, R.K. The Development and Use of Microsatellite Markers for Genetic Analysis and Plant Breeding with the Emphasis on Bread Wheat. Euphytica 2000, 113, 163–185. [Google Scholar] [CrossRef]
  40. Cruz, V.M.V.; Kilian, A.; Dierig, D.A. Development of DArT marker platforms and genetic diversity assessment of the US collection of the new oilseed crop lesquerella and related species. PLoS ONE 2013, 8, 64062. [Google Scholar] [CrossRef]
  41. Sivprakash, K.R.; Prashanth, S.R.; Mohanty, B.P.; Parida, A. Genetic diversity of Black gram landraces as evaluated by AFLP markers. Curr. Sci. 2004, 86, 1411–1415. [Google Scholar]
  42. Botstein, D.; White, R.L.; Skolnick, M.; Davis, R.W. Construction of a genetic linkage map in man using restriction fragment length polymorphism. Am. J. Hum. Genet. 1980, 32, 314–331. [Google Scholar] [PubMed]
  43. Williams, J.G.; Kubelik, A.R.; Livak, K.; Rafalski, J.A.; Tingey, S.V. DNA polymorphisms amplified by arbitrary primers are useful as genetic markers. Nucleic Acids Res. 1990, 18, 6531–6535. [Google Scholar] [CrossRef]
  44. Tautz, D. Hypervariability of simple sequences as a general source of polymorphic DNA markers. Nucleic Acids Res. 1989, 17, 6463–6471. [Google Scholar] [CrossRef] [PubMed]
  45. Konieczny, A.; Ausubel, F.M. A procedure for mapping Arabidopsis mutations using co-dominant ecotype-specific PCR-based markers. Plant J. 1993, 4, 403–410. [Google Scholar] [CrossRef]
  46. Paran, I.; Michelmore, R.W. Development of reliable PCR based markers linked to downy mildew resistance genes in lettuce. Theor. Appl. Genet. 1993, 85, 985–993. [Google Scholar] [CrossRef] [PubMed]
  47. Vos, P.; Hogers, R.; Bleeker, M.; Reijans, M.; Lee, T.V.D.; Hornes, M.; Frijters, A.; Pot, J.; Paleman, J.; Kuiper, M. AFLP: A new technique for DNA fingerprinting. Nucleic Acids Res. 1995, 23, 4407–4414. [Google Scholar] [CrossRef]
  48. Gupta, P.K.; Roy, J.K.; Prasad, M. Single nucleotide polymorphisms: A new paradigm for molecular marker technology and DNA polymorphism detection with emphasis on their use in plants. Curr. Sci. 2001, 80, 524–535. [Google Scholar]
  49. Jaccoud, D.; Peng, K.; Feinstein, D.; Kilian, A. Diversity arrays: A solid state technology for sequence information independent genotyping. Nucleic Acids Res. 2001, 29, e25. [Google Scholar] [CrossRef] [PubMed]
  50. Varshney, R.K.; Mohan, S.M.; Gaur, P.M.; Gangarao, N.V.P.R.; Pandey, M.K.; Bohra, A.; Sawargaonkar, S.L.; Chitikineni, A.; Kimurto, P.K.; Janila, P.; et al. Achievements and prospects of genomics-assisted breeding in three legume crops of the semi-arid tropics. Biotechnol. Adv. 2013, 31, 1120–1134. [Google Scholar] [CrossRef]
  51. Pandey, M.K.; Roorkiwal, M.; Singh, V.; Ramalingam, A.; Kudapa, H.; Thudi, M.; Chitikineni, A.; Rathore, A.; Varshney, R.K. Emerging genomic tools for legume breeding: Current status and future prospects. Front. Plant Sci. 2016, 7, 455. [Google Scholar] [CrossRef]
  52. Salgotra, R.K.; Zargar, S.M. Rediscovery of Genetic and Genomic Resources for Future Food Security; Springer: Singapore, 2020. [Google Scholar]
  53. Wang, K.C.; Yang, Y.W.; Liu, B.; Sanyal, A.; Corces-Zimmerman, R.; Chen, Y.; Lajoie, B.R.; Protacio, A.; Flynn, R.A.; Gupta, R.A.; et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 2011, 472, 120–124. [Google Scholar] [CrossRef] [PubMed]
  54. Schena, M.; Shalon, D.; Davis, R.W.; Brown, P.O. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science 1995, 270, 467–470. [Google Scholar] [CrossRef] [PubMed]
  55. Govindarajan, R.; Duraiyan, J.; Kaliyappan, K.; Palanisamy, M. Microarray and its applications. J. Pharm. Bioallied Sci. 2012, 4, S310–S312. [Google Scholar] [PubMed]
  56. Russo, G.; Zegar, C.; Giordano, A. Advantages and limitations of microarray technology in human cancer. Oncogene 2003, 22, 6497–6507. [Google Scholar] [CrossRef] [PubMed]
  57. Loy, A.; Bodrossy, L. Highly parallel microbial diagnostics using oligonucleotide microarrays. Clin. Chim. Acta 2006, 363, 106–119. [Google Scholar] [CrossRef] [PubMed]
  58. Shendure, J.; Ji, H. Next-generation DNA sequencing. Nat. Biotechnol. 2008, 26, 1135–1145. [Google Scholar] [CrossRef] [PubMed]
  59. Sabreena; Nazir, M.; Ganai, B.A.; Mir, R.A.; Zargar, S.M. Genetics and Genomics Resources of Millets: Availability, Advancements, and Applications. In Neglected and Underutilized Crops-Towards Nutritional Security and Sustainability; Springer: Singapore, 2021; pp. 153–166. [Google Scholar]
  60. Afzal, M.; Alghamdi, S.S.; Migdadi, H.H.; Khan, M.A.; Mirza, S.B.; El-Harty, E. Legume genomics and transcriptomics: From classic breeding to modern technologies. Saudi J. Biol. Sci. 2020, 27, 543–555. [Google Scholar] [CrossRef] [PubMed]
  61. Adams, M.D.; Kelley, J.M.; Gocayne, J.D.; Dubnick, M.; Polymeropoulos, M.H.; Xiao, H.; Merril, C.R.; Wu, A.; Olde, B.; Moreno, R.F.; et al. Complementary DNA sequencing: Expressed sequence tags and human genome project. Science 1991, 252, 1651–1656. [Google Scholar] [CrossRef] [PubMed]
  62. Boguski, M.S.; Lowe, T.M.; Tolstoshev, C.M. dbEST–database for “expressed sequence tags”. Nat. Genet. 1993, 4, 332–333. [Google Scholar] [CrossRef] [PubMed]
  63. Kim, Y.C.; Wang, S.M. Decoding neuron transcriptome by SAGE. Encyclopedia of Neuroscience; Elsevier Ltd.: Amsterdam, The Netherlands, 2009; pp. 357–363. [Google Scholar]
  64. Martin, G.; Baurens, F.C.; Droc, G.; Rouard, M.; Cenci, A.; Kilian, A.; Hastie, A.; Doležel, J.; Aury, J.M.; Alberti, A.; et al. Improvement of the banana “Musa acuminata” reference sequence using NGS data and semi-automated bioinformatics methods. BMC Genom. 2016, 17, 1–12. [Google Scholar] [CrossRef]
  65. Ambrosino, L.; Colantuono, C.; Diretto, G.; Fiore, A.; Chiusano, M.L. Bioinformatics resources for plant abiotic stress responses: State of the art and opportunities in the fast evolving-omics era. Plants 2020, 9, 591. [Google Scholar] [CrossRef] [PubMed]
  66. Unamba, C.I.; Nag, A.; Sharma, R.K. Next generation sequencing technologies: The doorway to the unexplored genomics of non-model plants. Front. Plant Sci. 2015, 6, 1074. [Google Scholar] [CrossRef]
  67. Tofazzal Islam, T.I. CRISPR-Cas technology in modifying food crops. In CABI Reviews; CABI: Wallingford, UK, 2019; pp. 1–16. [Google Scholar]
  68. Katna, G.; Sood, V.K. Plant Genetic Resources, Traditional Knowledge and Their Use in Crop Improvement. In Plant Genetic Resources and Traditional Knowledge for Food Security; Springer: Singapore, 2015; pp. 23–38. [Google Scholar]
  69. Project, I.R.G.S. The Map-Based Sequence of the Rice Genome. Nature 2005, 436, 793–800. [Google Scholar]
  70. Yu, J.; Hu, S.; Wang, J.; Wong, G.K.S.; Li, S.; Liu, B.; Deng, Y.; Dai, L.; Zhou, Y.; Zhang, X.; et al. A draft sequence of the rice genome (Oryza sativa L. ssp. indica). Science 2002, 296, 79–92. [Google Scholar] [CrossRef] [PubMed]
  71. Vielle-Calzada, J.P.; Martínez de la Vega, O.; Hernández-Guzmán, G.; Ibarra-Laclette, E.; Alvarez-Mejía, C.; Vega-Arreguín, J.C.; Jiménez-Moraila, B.; Fernández-Cortés, A.; Corona-Armenta, G.; Herrera-Estrella, L. The palomero genome suggests metal effects on domestication. Science 2009, 326, 1078. [Google Scholar] [CrossRef] [PubMed]
  72. Cooper, E.A.; Brenton, Z.W.; Flinn, B.S.; Jenkins, J.; Shu, S.; Flowers, D.; Luo, F.; Wang, Y.; Xia, P.; Barry, K.; et al. A new reference genome for Sorghum bicolor reveals high levels of sequence similarity between sweet and grain genotypes: Implications for the genetics of sugar metabolism. BMC Genom. 2019, 20, 420. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, G.; Liu, X.; Quan, Z.; Cheng, S.; Xu, X.; Pan, S.; Xie, M.; Zeng, P.; Yue, Z.; Wang, W.; et al. Genome sequence of foxtail millet (Setaria Italica) provides insights into grass evolution and biofuel potential. Nat. Biotechnol. 2012, 30, 549–554. [Google Scholar] [CrossRef]
  74. Brenchley, R.; Spannagl, M.; Pfeifer, M.; Barker, G.L.A.; D’Amore, R.; Allen, A.M.; McKenzie, N.; Kramer, M.; Kerhornou, A.; Bolser, D.; et al. Analysis of the bread wheat genome using whole-genome shotgun sequencing. Nature 2012, 491, 705–710. [Google Scholar] [CrossRef] [PubMed]
  75. Sato, K. History and future perspectives of barley genomics. DNA Res. 2020, 27, dsaa023. [Google Scholar] [CrossRef] [PubMed]
  76. Hatakeyama, M.; Aluri, S.; Balachadran, M.T.; Sivarajan, S.R.; Patrignani, A.; Grüter, S.; Poveda, L.; Shimizu-Inatsugi, R.; Baeten, J.; Francoijs, K.J.; et al. Multiple hybrid de novo genome assembly of finger millet, an orphan allotetraploid Crop. DNA Res. 2018, 25, 39–47. [Google Scholar] [CrossRef] [PubMed]
  77. Hittalmani, S.; Mahesh, H.B.; Shirke, M.D.; Biradar, H.; Uday, G.; Aruna, Y.R.; Lohithaswa, H.C.; Mohanrao, A. Genome and transcriptome sequence of finger millet (Eleusine Coracana (L.) Gaertn.) provides insights into drought tolerance and nutraceutical properties. BMC Genom. 2017, 18, 465. [Google Scholar] [CrossRef] [PubMed]
  78. Shi, J.; Ma, X.; Zhang, J.; Zhou, Y.; Liu, M.; Huang, L.; Sun, S.; Zhang, X.; Gao, X.; Zhan, W.; et al. Chromosome conformation capture resolved near complete genome assembly of broomcorn millet. Nat. Commun. 2019, 10, 464. [Google Scholar] [CrossRef] [PubMed]
  79. Guo, L.; Qiu, J.; Ye, C.; Jin, G.; Mao, L.; Zhang, H.; Yang, X.; Peng, Q.; Wang, Y.; Jia, L.; et al. Echinochloa crus-galli genome analysis provides insight into its adaptation and invasiveness as a weed. Nat. Commun. 2017, 8, 1031. [Google Scholar] [CrossRef]
  80. Jones, E.; Chu, W.C.; Ayele, M.; Ho, J.; Bruggeman, E.; Yourstone, K.; Rafalski, A.; Smith, O.S.; McMullen, M.D.; Bezawada, C.; et al. Development of single nucleotide polymorphism (SNP) markers for use in commercial maize (Zea mays L.) germplasm. Mol. Breed. 2009, 24, 165–176. [Google Scholar] [CrossRef]
  81. Yang, Z.; Bai, Z.; Li, X.; Wang, P.; Wu, Q.; Yang, L.; Li, L.; Li, X. SNP identification and allelic-specific PCR markers development for TaGW2, a gene linked to wheat kernel weight. Theor. Appl. Genet. 2012, 125, 1057–1068. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, J.; Nakazaki, T.; Chen, S.; Chen, W.; Saito, H.; Tsukiyama, T.; Okumoto, Y.; Xu, Z.; Tanisaka, T. Identification and characterization of the erect-pose panicle gene EP conferring high grain yield in rice (Oryza sativa L.). Theor. Appl. Genet. 2009, 119, 85–91. [Google Scholar] [CrossRef]
  83. Spielmeyer, W.; Ellis, M.; Robertson, M.; Ali, S.; Lenton, J.R.; Chandler, P.M. Isolation of gibberellin metabolic pathway genes from barley and comparative mapping in barley, wheat and rice. Theor. Appl. Genet. 2004, 109, 847–855. [Google Scholar] [CrossRef] [PubMed]
  84. Raina, M.; Salgotra, R.K.; Pandotra, P.; Rathour, R.; Singh, K. Genetic enhancement for semi-dwarf and bacterial blight resistance with enhanced grain quality characteristics in traditional Basmati rice through marker-assisted selection. Comptes Rendus Biol. 2019, 342, 142–153. [Google Scholar] [CrossRef] [PubMed]
  85. Qi, Y.; Wang, L.; Gui, J.; Zhang, L.; Liu, Q.; Wang, J. Development and validation of a functional co-dominant SNP marker for the photoperiod thermo-sensitive genic male sterility pms3 (p/tms12-1) gene in rice. Breed. Sci. 2017, 67, 535–539. [Google Scholar] [CrossRef] [PubMed]
  86. Fu, D.; Szucs, P.; Yan, L.; Helguera, M.; Skinner, J.S.; Von Zitzewitz, J.; Hayes, P.M.; Dubcovsky, J. Large deletions within the first intron in VRN-1 are associated with spring growth habit in barley and wheat. Mol. Genet. Genom. 2005, 273, 54–65. [Google Scholar] [CrossRef] [PubMed]
  87. Rabbi, S.H.A.; Kumar, A.; Mohajeri Naraghi, S.; Simsek, S.; Sapkota, S.; Solanki, S.; Alamri, M.S.; Elias, E.M.; Kianian, S.; Missaoui, A.; et al. Genome-wide association mapping for yield and related traits under drought stressed and non-stressed environments in wheat. Front. Genet. 2021, 12, 649988. [Google Scholar] [CrossRef]
  88. Tomar, V.; Singh, D.; Dhillon, G.S.; Singh, R.P.; Poland, J.; Joshi, A.K.; Singh, P.K.; Bhati, P.K.; Kumar, S.; Rahman, M.; et al. New QTLs for spot blotch disease resistance in wheat (Triticum aestivum L.) using genome-wide association mapping. Front. Genet. 2021, 11, 613217. [Google Scholar] [CrossRef]
  89. He, X.; Juliana, P.; Kabir, M.R.; Roy, K.K.; Islam, R.; Marza, F.; Peterson, G.; Singh, G.P.; Chawade, A.; Joshi, A.K.; et al. Screening and mapping for head blast resistance in a panel of CIMMYT and South Asian bread wheat germplasm. Front. Genet. 2021, 12, 679162. [Google Scholar] [CrossRef]
  90. Pradhan, A.K.; Kumar, S.; Singh, A.K.; Budhlakoti, N.; Mishra, D.C.; Chauhan, D.; Mittal, S.; Grover, M.; Kumar, S.; Gangwar, O.P.; et al. Identification of QTLs/defense genes effective at seedling stage against prevailing races of wheat stripe rust in India. Front. Genet. 2020, 11, 572975. [Google Scholar] [CrossRef]
  91. Kokhmetova, A.; Sehgal, D.; Ali, S.; Atishova, M.; Kumarbayeva, M.; Leonova, I.; Dreisigacker, S. Genome-wide association study of tan spot resistance in a hexaploid wheat collection from Kazakhstan. Front. Genet. 2021, 11, 581214. [Google Scholar] [CrossRef]
  92. Xu, F.; Chen, S.; Yang, X.; Zhou, S.; Wang, J.; Zhang, Z.; Huang, Y.; Song, M.; Zhang, J.; Zhan, K.; et al. Genome-wide association study on root traits under different growing environments in wheat (Triticum aestivum L.). Front. Genet. 2021, 12, 646712. [Google Scholar] [CrossRef]
  93. Sehgal, D.; Mondal, S.; Crespo-Herrera, L.; Velu, G.; Juliana, P.; Huerta-Espino, J.; Shrestha, S.; Poland, J.; Singh, R.; Dreisigacker, S. Haplotype-based, genome-wide association study reveals stable genomic regions for grain yield in CIMMYT spring bread wheat. Front. Genet. 2020, 11, 589490. [Google Scholar] [CrossRef]
  94. Cao, J.; Shang, Y.; Xu, D.; Xu, K.; Cheng, X.; Pan, X.; Liu, X.; Liu, M.; Gao, C.; Yan, S.; et al. Identification and validation of new stable QTLs for grain weight and size by multiple mapping models in common wheat. Front. Genet. 2020, 11, 584859. [Google Scholar] [CrossRef]
  95. Adhikari, A.; Basnet, B.R.; Crossa, J.; Dreisigacker, S.; Camarillo, F.; Bhati, P.K.; Jarquin, D.; Manes, Y.; Ibrahim, A.M. Genome-wide association mapping and genomic prediction of anther extrusion in CIMMYT hybrid wheat breeding program via modeling pedigree, genomic relationship, and interaction with the environment. Front. Genet. 2020, 11, 586687. [Google Scholar] [CrossRef]
  96. Kaur, S.; Kaur, J.; Mavi, G.S.; Dhillon, G.S.; Sharma, A.; Singh, R.; Devi, U.; Chhuneja, P. Pyramiding of high grain weight with stripe rust and leaf rust resistance in elite Indian wheat cultivar using a combination of marker assisted and phenotypic selection. Front. Genet. 2020, 11, 593426. [Google Scholar] [CrossRef]
  97. Chen, L.; Wang, Q.; Tang, M.; Zhang, X.; Pan, Y.; Yang, X.; Gao, G.; Lv, R.; Tao, W.; Jiang, L.; et al. QTL mapping and identification of candidate genes for heat tolerance at the flowering stage in rice. Front. Genet. 2021, 11, 621871. [Google Scholar] [CrossRef]
  98. Lee, C.M.; Park, H.S.; Baek, M.K.; Jeong, O.Y.; Seo, J.; Kim, S.M. QTL mapping and improvement of pre-harvest sprouting resistance using japonica weedy rice. Front. Plant Sci. 2023, 14, 1194058. [Google Scholar] [CrossRef]
  99. Chen, H.; Xie, W.; He, H.; Yu, H.; Chen, W.; Li, J.; Yu, R.; Yao, Y.; Zhang, W.; He, Y.; et al. A high-density SNP genotyping array for rice biology and molecular breeding. Mol. Plant 2014, 7, 541–553. [Google Scholar] [CrossRef]
  100. Kim, K.W.; Nawade, B.; Nam, J.; Chu, S.H.; Ha, J.; Park, Y.J. Development of an inclusive 580K SNP array and its application for genomic selection and genome-wide association studies in rice. Front. Plant Sci. 2022, 13, 1036177. [Google Scholar] [CrossRef] [PubMed]
  101. Reyes, V.P.; Kitony, J.K.; Nishiuchi, S.; Makihara, D.; Doi, K. Utilization of genotyping-by-sequencing (GBS) for rice pre-breeding and improvement: A review. Life 2022, 12, 1752. [Google Scholar] [CrossRef] [PubMed]
  102. Bartholomé, J.; Prakash, P.T.; Cobb, J.N. Genomic Prediction: Progress and Perspectives for Rice Rice Improvement. In Genomic Prediction of Complex Traits: Methods and Protocols; Humana: New York, NY, USA, 2022; pp. 569–617. [Google Scholar]
  103. Subedi, S.R.; Sandhu, N.; Singh, V.K.; Sinha, P.; Kumar, S.; Singh, S.P.; Ghimire, S.K.; Pandey, M.; Yadaw, R.B.; Varshney, R.K.; et al. Genome-wide association study reveals significant genomic regions for improving yield, adaptability of rice under dry direct seeded cultivation condition. BMC Genom. 2019, 20, 471. [Google Scholar] [CrossRef]
  104. Daware, A.; Malik, A.; Srivastava, R.; Das, D.; Ellur, R.K.; Singh, A.K.; Tyagi, A.K.; Parida, S.K. Rice Pangenome Genotyping Array: An efficient genotyping solution for pangenome-based accelerated genetic improvement in rice. Plant J. 2023, 113, 26–46. [Google Scholar] [CrossRef] [PubMed]
  105. Qin, Y.; Cheng, P.; Cheng, Y.; Feng, Y.; Huang, D.; Huang, T.; Song, X.; Ying, J. QTL-Seq identified a major QTL for grain length and weight in rice using near isogenic F2 population. Rice Sci. 2018, 25, 121–131. [Google Scholar]
  106. Zhao, M.; Ma, Z.; Wang, L.; Tang, Z.; Mao, T.; Liang, C.; Gao, H.; Zhang, L.; He, N.; Fu, L.; et al. SNP-based QTL mapping for panicle traits in the japonica super rice cultivar Liaoxing 1. Crop J. 2020, 8, 769–780. [Google Scholar] [CrossRef]
  107. Ranganatha, H.M.; Lohithaswa, H.C.; Pandravada, A. Mapping and validation of major quantitative trait loci for resistance to northern corn leaf blight along with the determination of the relationship between resistances to multiple foliar pathogens of maize (Zea mays L.). Front. Genet. 2021, 11, 548407. [Google Scholar] [CrossRef]
  108. Adewale, S.A.; Badu-Apraku, B.; Akinwale, R.O.; Paterne, A.A.; Gedil, M.; Garcia-Oliveira, A.L. Genome-wide association study of Striga resistance in early maturing white tropical maize inbred lines. BMC Plant Biol. 2020, 20, 203. [Google Scholar] [CrossRef]
  109. Zebire, D.; Menkir, A.; Adetimirin, V.; Mengesha, W.; Meseka, S.; Gedil, M. Identifying suitable tester for evaluating Striga resistant lines using DArTseq markers and agronomic traits. PLoS ONE 2021, 16, e0253481. [Google Scholar] [CrossRef] [PubMed]
  110. Ma, J.; Cao, Y.; Wang, Y.; Ding, Y. Development of the maize 5.5 K loci panel for genomic prediction through genotyping by target sequencing. Front. Plant Sci. 2022, 13, 4544. [Google Scholar] [CrossRef]
  111. Yu, G.; Cui, Y.; Jiao, Y.; Zhou, K.; Wang, X.; Yang, W.; Xu, Y.; Yang, K.; Zhang, X.; Li, P.; et al. Comparison of sequencing-based and array-based genotyping platforms for genomic prediction of maize hybrid performance. Crop J. 2023, 11, 490–498. [Google Scholar] [CrossRef]
  112. Wang, M.; Zhang, R.; Zhao, Y.; Yao, J.; Li, W.; Yang, Z.; Sun, F.; Yang, X. Identifying QTL and candidate genes for prolificacy in maize. Crop J. 2023, 11, 531–539. [Google Scholar] [CrossRef]
  113. Gupta, M.; Choudhary, M.; Singh, A.; Sheoran, S.; Singla, D.; Rakshit, S. Meta-QTL analysis for mining of candidate genes and constitutive gene network development for fungal disease resistance in maize (Zea mays L.). Crop J. 2023, 11, 511–522. [Google Scholar] [CrossRef]
  114. Beyene, Y.; Semagn, K.; Crossa, J.; Mugo, S.; Atlin, G.N.; Tarekegne, A.; Meisel, B.; Sehabiague, P.; Vivek, B.S.; Oikeh, S.; et al. Improving maize grain yield under drought stress and non-stress environments in sub-Saharan Africa using marker-assisted recurrent selection. Crop Sci. 2016, 56, 344–353. [Google Scholar] [CrossRef]
  115. Ellis, M.; Spielmeyer, W.; Gale, K.; Rebetzke, J.; Richards, A. ‘‘Perfect’’ markers for the Rht-B1b and Rht-D1b dwarfing genes in wheat. Theor. Appl. Genet. 2002, 105, 1038–1042. [Google Scholar] [CrossRef] [PubMed]
  116. Mikołajczak, K.; Ogrodowicz, P.; Surma, M.; Adamski, T.; Kuczyńska, A. Introgression of LTP2 gene through marker assisted backcross in barley (Hordeum vulgare L.). Electron. J. Biotechnol. 2016, 24, 9–11. [Google Scholar] [CrossRef]
  117. Hernandez, J.; Steffenson, B.J.; Filichkin, T.; Fisk, S.P.; Helgerson, L.; Meints, B.; Vining, K.J.; Marshall, D.; del Blanco, A.; Chen, X.; et al. Introgression of rpg4/Rpg5 into barley germplasm provides insights into the genetics of resistance to Puccinia graminis f. sp. tritici race ttksk and resources for developing resistant cultivars. Phytopathology 2019, 109, 1018–1028. [Google Scholar] [CrossRef]
  118. Tommasini, L.; Yahiaoui, N.; Srichumpa, P.; Keller, B. Development of functional markers specific for seven Pm3 resistance alleles and their validation in the bread wheat gene pool. Theor. Appl. Genet. 2006, 114, 165–175. [Google Scholar] [CrossRef] [PubMed]
  119. Periyannan, S.; Bansal, U.; Bariana, H.; Deal, K.; Luo, M.; Dvorak, J.; Lagudah, E. Identification of a robust molecular marker for the detection of the stem rust resistance gene Sr45 in common wheat. Theor. Appl. Genet. 2014, 127, 947–955. [Google Scholar] [CrossRef] [PubMed]
  120. Lagudah, E.S.; Krattinger, S.G.; Herrera-Foessel, S.; Singh, R.P.; Huerta-Espino, J.; Spielmeyer, W.; Brown-Guedira, G.; Selter, L.L.; Keller, B. Gene specific markers for the wheat gene Lr34/Yr18/Pm38 which confers resistance to multiple fungal pathogens. Theor. Appl. Genet. 2009, 119, 889–898. [Google Scholar] [CrossRef] [PubMed]
  121. He, X.; Zhang, Y.; He, Z.; Wu, Y.P.; Xiao, Y.G.; Ma, C.X.; Xia, X.C. Characterization of phytoene synthase 1 gene (Psy1) located on common wheat chromosome 7A and development of a functional marker. Theor. Appl. Genet. 2008, 116, 213–221. [Google Scholar] [CrossRef]
  122. Wang, L.; Zhao, X.; He, Z.; Ma, W.; Appels, R.; Pena, R.J.; Xia, X.C. Characterization of low molecular-weight glutenin subunit Glu-B3 genes and development of STS markers in common wheat (T. aestivum L.). Theor. Appl. Genet. 2009, 118, 525–539. [Google Scholar] [CrossRef]
  123. Ramkumar, G.; Sivaranjani, A.; Pandey, M.K.; Sakthivel, K.; Shobha, N.R.; Sudarshan, I.; Prasad, G.S.V.; Neeraja, C.N.; Sundaram, R.M.; Viraktamath, B.C.; et al. Development of a PCR-based SNP marker system for effective selection of kernel length and kernel elongation in rice. Mol. Breed. 2010, 26, 735–740. [Google Scholar] [CrossRef]
  124. Zhou, L.; Chen, S.; Yang, G.; Zha, W.; Cai, H.; Li, S.; Chen, Z.; Liu, K.; Xu, H.; You, A. A perfect functional marker for the gene of intermediate amylose content Wx-in in rice (Oryza sativa L.). Crop Breed. Appl. Biotechnol. 2018, 18, 103–109. [Google Scholar] [CrossRef]
  125. Thornsberry, J.M.; Goodman, M.M.; Doebley, J.; Kresovich, S.; Nielsen, D.; Buckler, E.S. Dwarf8 polymorphisms associate with variation in flowering time. Nat. Gen. 2001, 28, 286–289. [Google Scholar] [CrossRef] [PubMed]
  126. Chhabra, R.; Hossain, F.; Muthusamy, V.; Baveja, A.; Mehta, B.K.; Zunjare, R.U. Development and validation of breeder-friendly functional markers of sugary1 gene encoding starch-debranching enzyme affecting kernel sweetness in maize (Zea mays). Crop Pasture Sci. 2019, 70, 868–875. [Google Scholar] [CrossRef]
  127. Dhutmal, R.R.; Mundhe, A.G.; More, A.W. Molecular marker techniques: A Review. Int. J. Curr. Microbiol. App. Sci. 2018, 6, 816–825. [Google Scholar]
  128. Amom, T.; Nongdam, P. The use of molecular marker methods in plants: A review. Int. J. Curr. Res. Rev. 2017, 9, 1–7. [Google Scholar]
  129. Joseph, M.; Gopalakrishnan, S.; Sharma, R.K.; Singh, V.P.; Singh, A.K.; Singh, N.K.; Mohapatra, T. Combining bacterial blight resistance and Basmati quality characteristics by phenotypic and molecular marker-assisted selection in rice. Mol. Breed. 2004, 13, 377–387. [Google Scholar] [CrossRef]
  130. Yugander, A.; Sundaram, R.M.; Singh, K.; Ladhalakshmi, D.; Subba Rao, L.V.; Madhav, M.S.; Badri, J.; Prasad, M.S.; Laha, G.S. Incorporation of the novel bacterial blight resistance gene Xa38 into the genetic background of elite rice variety ‘Improved Samba Mahsuri’. PLoS ONE 2018, 13, e0198260. [Google Scholar] [CrossRef]
  131. Ellur, R.K.; Khanna, A.; Gopala Krishnan, S.; Bhowmick, P.K.; Vinod, K.K.; Nagarajan, M.; Mondal, K.K.; Singh, N.K.; Singh, K.; Prabhu, K.V.; et al. Marker-aided incorporation of Xa38, a novel bacterial blight resistance gene, in PB1121 and comparison of its resistance spectrum with xa13 + Xa21. Sci. Rep. 2016, 6, 29188. [Google Scholar] [CrossRef]
  132. Das, G.; Rao, G.J.; Varier, M.; Prakash, A.; Prasad, D. Improved Tapaswini having four BB resistance genes pyramided with six genes/QTLs, resistance/tolerance to biotic and abiotic stresses in rice. Sci. Rep. 2018, 8, 2413. [Google Scholar] [CrossRef]
  133. Abdul Fiyaz, R.; Shivani, D.; Chaithanya, K.; Mounika, K.; Chiranjeevi, M.; Laha, G.S.; Viraktamath, B.C.; Subba Rao, L.V.; Sundaram, R.M. Genetic improvement of rice for bacterial blight resistance: Present status and future prospects. Rice Sci. 2022, 29, 118–132. [Google Scholar] [CrossRef]
Figure 1. Different genomic approaches used for cereal crop improvements.
Figure 1. Different genomic approaches used for cereal crop improvements.
Genes 14 01770 g001
Figure 2. Molecular markers used in MAS for cereal crop improvements.
Figure 2. Molecular markers used in MAS for cereal crop improvements.
Genes 14 01770 g002
Figure 3. A prototype of marker-assisted introgression/selection for high nutrition varieties in crops.
Figure 3. A prototype of marker-assisted introgression/selection for high nutrition varieties in crops.
Genes 14 01770 g003
Table 2. Genomic resources and genotyping platforms for the improvement of major cereal crops.
Table 2. Genomic resources and genotyping platforms for the improvement of major cereal crops.
CropMolecular Breeding ApproachesTraits ImprovedReferences
WheatQTL mapping and GWAS using Infinium 90 K SNP assayDrought tolerance[87]
GWAS using GBSSpot blotch resistance[88]
GWAS using Illumina Infinium 15 K BeadChipHead blast resistance[89]
GWAS using 35 K axiom® arraysStripe rust resistance[90]
GWAS using DArTseq technologyTan spot resistance[91]
GWAS using wheat 660 K SNP arrayIdentification of chromosomal regions of root traits[92]
Haplotype-based GWAS and genotyping by sequencingGrain yield[93]
QTL mapping (RIL population derived from specific locus amplified fragment sequencing (SLAF-seq))Grain weight and size[94]
GWAS using 20 K Infinitum iSelect
SNP array and genomic prediction of anther extrusion in hybrid breeding
Helps in rapid breeding for a particular trait[95]
Marker-assisted selection Pyramided genes of high grain weight, stripe rust, and leaf rust resistance[96]
RiceQTL mappingHeat tolerance[97]
QTL mappingPre-harvest sprouting resistance[98]
SNP genotyping array RiceSNP50Functional genomics and molecular breeding[99]
580 K SNP arrayGenomic selection and GWAS[100]
Genotyping-by-sequencingPre-breeding and improvement[101]
Genomic predictionRice improvement[102]
GWASAdaptability to dry direct-seeded rice (DDSR) system[103]
Rice pangenome genotyping array (RPGA) SNP genotypingGenetic improvement[104]
QTL-seq using NIL-F2Grain length and weight[103]
SNP-based QTL mappingPanicle traits[104]
MaizeQTL mappingNorthern corn leaf blight resistance[105]
GWASStriga resistance[106]
DArT seq SNP genotypingStriga resistance hybrid breeding[107]
5.5 K SNPs using genotyping by target sequencing (GBTS)Genomic prediction[108]
GBS, 40 K SNP array and target sequence captureGenomic prediction[109]
QTL mappingProlificacy trait[110]
Meta-QTL analysisFungal disease resistance[111]
Marker-assisted recurrent selection (MARS)Grain yield[112]
GWASEar rot resistance[113]
QTL/genomic region identificationBiotic stress resistance[114]
BarleyLTP2 through marker-assisted recurrent selection (MARS)Semi-dwarf[115]
Introgression of rpg4/Rpg5 gene through marker-assisted recurrent selection (MABB)Stem rust disease resistance[116]
Pearl milletIntrogression of qRSg1 and qRSg4 genes through marker-assisted selection (MAS)Downy mildew disease resistance[117]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chaudhary, N.; Salgotra, R.K.; Chauhan, B.S. Genetic Enhancement of Cereals Using Genomic Resources for Nutritional Food Security. Genes 2023, 14, 1770. https://doi.org/10.3390/genes14091770

AMA Style

Chaudhary N, Salgotra RK, Chauhan BS. Genetic Enhancement of Cereals Using Genomic Resources for Nutritional Food Security. Genes. 2023; 14(9):1770. https://doi.org/10.3390/genes14091770

Chicago/Turabian Style

Chaudhary, Neeraj, Romesh Kumar Salgotra, and Bhagirath Singh Chauhan. 2023. "Genetic Enhancement of Cereals Using Genomic Resources for Nutritional Food Security" Genes 14, no. 9: 1770. https://doi.org/10.3390/genes14091770

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

Article Metrics

Back to TopTop