Open access peer-reviewed chapter

Genomic Approaches in Wheat Breeding for Sustainable Production under Changing Climate

Written By

Zahid Manzoor, Junwei Liu, Muhammad Sheeraz Qadir, Muhammad Ahsan Jamil, Zeshan Hassan, Muhammad Shah Jahan and Amir Shakeel

Submitted: 08 March 2022 Reviewed: 30 March 2022 Published: 30 May 2022

DOI: 10.5772/intechopen.104751

From the Edited Volume

Wheat - Recent Advances

Edited by Mahmood-ur-Rahman Ansari

Chapter metrics overview

209 Chapter Downloads

View Full Metrics

Abstract

Wheat is the most important cereal crop, a great source of dietary protein. It is grown worldwide for its consumption in the form of different products. Wheat production faces a lot of biotic and abiotic stresses that hinder growth and yield. Changing climate is a worse scenario to be adopted for sustainable production. Food demand is rapidly increasing by a drastic increase in the world population. Conventional breeding techniques are time-consuming and ineffective in attaining high yield goals under changing climates. Next-generation sequencing revolutionized wheat breeding through molecular approaches for effective selection. The use of genomic approaches in wheat breeding is the need of time for sustainable production. Several genomic approaches, such as use of genome-wide markers for gene mapping, genomic selection and recurrent selection through QTL and meta-QTL analysis, markers-assisted selection in haploid breeding, heterosis breeding through genomic tools, and biotechnological tools, are currently used as modern techniques for developing climate-resilient wheat cultivars. This chapter illustrated the challenges of changing climate, molecular techniques in wheat breeding to develop climate-resilient genotypes, sustainable wheat production to cope with food demand, and future breeding strategies.

Keywords

  • genomic approaches
  • wheat breeding
  • sustainable goals
  • climate change
  • resilient cultivars
  • marker-assisted breeding

1. Introduction

The one-third population of the world mainly relies on wheat (Triticum aestivum L.) for their daily diet and it is becoming more important with a great increase in the world’s population [1, 2, 3]. Wheat is grown in an area of about 220 million hectares worldwide [4], with an average annual production of 729 million tons [5]. Wheat is also used as an industrial material and renewable feed resource [6]. Wheat demand is increasing due to increased population thought the world [5]; thus, it is influencing market prices [7], competition, and growing demand [8]. The considerable challenge in sustainable wheat production is to increase the yield with demand in continuously changing environmental conditions. It has been suggested that wheat yield should be increased by 1.7% per annum globally for the next 30 years [9]. Still, its production rate is 0.9% per annum [10], insufficient to meet global hunger and even in main wheat-producing countries, this percentage is gradually decreasing. The goal of sustainable wheat production is only possible to achieve by growing wheat in the best environmental condition since the world is facing the massive challenge of climate change; therefore, this could not be possible [11]. Yield and yield stability are highly affected by climate change [12].

Although agronomic practices and conventional breeding contributed to sustainable wheat production, now it is time to boost wheat production with the latest introduced technologies. So, breeders are looking for highly efficient methods to increase yield in a limited time [13]. Newly developed technologies, such as phenomics [14], marker-assisted selection (MAS) [15], genomic breeding [16], and biotechnological (cis-genic and transgenic) techniques [17], are promising technologies to fight hunger in the future.

Marker-assisted breeding is based on gene linkage and recombination events in meiosis [18]. Different molecular markers are being used to detect the variations in wheat germplasm [19] and resistant genes are being identified in various lines and used in marker-assisted breeding [20, 21]. Genomic selection (GS) is an advanced form of MAS [13]. Initially, a panel of genotypes, training population, is selected in GS, then genotyping is performed with genome-wide markers and lastly, phenotyping is done for the trait of interest. Genome-estimated breeding values (GEBVs) are calculated with the help of a training population for all genotypes included in the panel, newly developed lines, and the validation population [22]. GS is more effective based on computing more variations with the help of GEBVs without phenotyping [23]. Modified methods in GS significantly increase genetic gain and accuracy. Several recent genomic approaches have been illustrated in Figure 1.

Figure 1.

Schematic diagram for recent genomic approaches in wheat breeding.

This chapter focuses on genomic approaches in wheat breeding to combat hunger in changing climate. Genomic breeding in wheat is an efficient way to increase wheat production in changing climate and global warming scenarios. Using molecular markers in QTL-mapping and its combination with genome with association study (GWAS) could significantly improve yield to achieve sustainable wheat production goals.

Advertisement

2. Gene mapping through genome-wide markers

Initially, restriction fragment polymorphism (RFLP) markers were introduced to identify cultivars [24] and gene mapping, but the frequency of markers was not impressive due to the extremely low level of polymorphism for the D genome of bread wheat [25]. International Triticeae Mapping Initiative (ITMI) was very efficient because of generating high-density linkage groups [26]. With the advancement in technology, PCR-based markers were developed. There were two broad categories [27], simple sequence repeat (SSR) [28] and randomly amplified polymorphic DNA (RAPD), considered far better than RFLP markers. These markers proved to be time-saving and cost-effective, especially SSR markers were extensively used in wheat due to reproducibility. RAPD markers were used to make sequence characterized amplified regions (SCAR) or sequence-tagged sites (STS) markers [29] that were more reliable in wheat, for example, Lr24 and Lr2 QTL for Russian wheat aphid [30].

The first discovery of simple sequence repeats (SSR) markers, also known as microsatellites [28], opened a new era of wheat breeding due to their extensive use because they are highly reproducible, genome-specific, highly polymorphic, and relatively abundant [31]. The yield and yield-related traits in wheat were exploited with more resolution on their respective loci in the genome [32]. SSR markers also had limitations due to high cost, random distribution in the genome, finite motifs, and difficulty obtaining exact information [33]. Single nucleotide polymorphism (SNP) markers analyze variations at a single nucleotide level; therefore, they are not very effective in marker-assisted selection of wheat [34, 35, 36, 37].

Advertisement

3. Genome based breeding strategies in wheat

The reciprocal recurrent genomic selection in wheat is important to increase wheat yield [38]. Genomic selection of desired traits speeds up the breeding program with the accurate selection of traits of interest with the help of QTL and GWAS [39, 40]. When additive effects of focused QTL are not determined, they can be estimated by genome-wide prediction. The Ridge regression model has been proposed to be used in the genomic selection of desired traits with more accuracy and unbiased decisions [41, 42]. F∞ metric is implemented to determine additive effects and additive-by-additive epistasis [43]. The superior plants are selected for the next generation with the help of the estimation of additive effects in recurrent genomic selection [22]. The QTL study with the ridge regression model revealed that nonadditive effects were related to grain yield [44]; therefore, they are suggested to be included in the genome prediction model to increase wheat yield. Persistency in predicting models is mainly dependent upon the adequate size of parental population and linkage disequilibrium, size of training population, use of the statistical model to estimate markers effects in recurrent genomic selection, and density of markers [45, 46, 47].

Estimating haploid breeding and genomic breeding values are two important strategies for assessing long-term genetic gain and maintenance in recurrent genomic breeding [48]. Based on these values, optimal population value selection is performed to make blocks of genotypes exhibiting maximum haploid or genomic breeding values. Genotypes with maximum haploid value would fall in the block with minimal segregation. In contrast, minimal value haploid value will lead to minimal population value selection with a haplotype block with maximum segregation for the desired trait [48]. Genomic recurrent selection and its modified form reciprocal recurrent selection increase the efficacy of wheat breeding programs [49] to develop high-yielding, climate-resilient genotypes. Gene pyramiding is a novel concept in modern breeding to accumulate desired genes in a single genotype to develop an ideotype [50]. Implications of this breeding strategy via genomic breeding can be a way forward to achieve a landmark in wheat breeding programs for sustainable production in changing climate.

Advertisement

4. Marker-assisted selection

Genetic linkage between loci of the same chromosome and their recombination events during meiosis are the main basis of MAS [18]. The transfer of two loci together or separately in the next generation is dependent on how closely these loci are located on the chromosome [51]. There will be more chances to be inherited together if they are located closely on the same chromosome. Molecular markers are used to identify a specific region on the chromosome for a gene of interest [52]. Different alleles are detected in several lines, known as a polymorphism for a trait of interest in different lines. Molecular markers detect the presence of linked alleles on the base of genetic linkage [27]. Marker-assisted selection could prove a very effective technique for developing climate-resilient wheat cultivars in changing climates.

Single sequence repeat (SSR) markers were used to detect the cell membrane stability of wheat cultivated under drought stress. SSR markers were significantly linked with cell membrane stability, but the association was weak. SSR markers were suggested to detect increased frequency in progenies with drought tolerance [53] and used Xwmc273.3 marker was used to detect QTL associated with higher grain yield of wheat under drought conditions. Different wheat cultivars in irrigated and rainfed regions were selected with the help of QTL mapping and phenotypic selection based on higher yield in water stress conditions [54]. Marker-assisted backcross breeding was employed to detect and transfer three drought-tolerant QTLs in high-yielding cultivars. QTLs were detected in drought-tolerant cultivar HI5100 and HD2733 was used as a recurrent cultivar. They identified 29 lines having drought tolerant QTLs; further background selection resulted in five varieties for evaluation in the national breeding program [55]. QTL expression in common wheat for cold tolerance was detected in the region of CBF and Cor/Lea gene families located at 5AL [56]. Soriano et al. [40] performed a QTL meta-analysis to identify QTLs for biotic and abiotic stress tolerance in durum wheat. They identified 315 of 85 MQTL, while 71 corresponded to biotic stress and 127 to abiotic stresses.

Advertisement

5. Heterosis breeding through genome based strategies

Only <1% of the wheat cultivated area is under the cultivation of hybrid wheat due to the highly self-pollinated wheat and other technical problems. Meanwhile, genomic breeding has helped resolve the technical issues of hybrid wheat production. Male sterility is not well-understood in wheat because it is highly self-pollinated, and hybrid seed production is very cost-effective. Male sterility is of great importance in the hybridization of wheat. Male sterility II (ms2) has been identified in wheat for 40 years, but its corresponding genes were unknown. It was determined through mapped-based cloning experiments in 2017 that the promoter region of ms2 has TRIM element, activates Ms2 allele in anther, which induces male sterility in wheat [57, 58].

Furthermore, it was also investigated by map-based cloning studies that male sterility 1 (ms1) also prevailed in wheat [59, 60]. Functional analysis of MS1 has revealed a newly introduced protein in the wheat and Poaceae family. It is localized in mitochondria and plastids, associated with the phospholipid-binding activity to induce male sterility. The split gene system also inserts male sterility in wheat by expressing the phytotoxic gene barnase, controlled by two alleles and its activation induces male sterility. This system maintains male sterile female plants while, after crossing, sterile hybrids are produced because it does not need male storer lines and entirely relies on the genetically modified female plants [61].

The utilization of genomics to predict heterotic patterns is another strategy for hybridizing wheat. These patterns are used mainly to characterize parents and the hybrids population on a large scale. Heterotic patterns were used to assess 1604 wheat hybrids for disease resistance and morphological parameters. It was demonstrated that 69 hybrids performed better than the best commercial line by 7.2–10.7% in production [62]. Zhao et al. [63] described a three-way genome-based strategy to predict heterotic wheat patterns in 1604 hybrids and 135 parents. In another experiment, 135 parents and their 1604 hybrids were assessed through genomic prediction of heterotic patterns and a complete performance of hybrids was evaluated to suggest high yielding heterotic patterns.

Heterotic patterns were used to assess a well-defined population and their effectiveness, limitations, and representation were estimated to measure their success. Identifying and exploiting major genes is essential and helpful in hybridization, such as genes responsible for dwarf wheat and very important for selecting the parental population to make hybrids. Further, these genes greatly influence pollen mass and anther extrusion in wheat [64]. Hybrids D1b and B1b with reduced height showed poor anther extrusion due to the expression of genes related to dwarfness [65, 66]. While developing high-yielding cultivars with lodging resistance must be considered for sustainable goals. Using another gene Rht24 for male sterility in hybridization could be very effective because it has no effects on male floral parts and anther extrusion [67]. These studies predicted heterotic patterns via a quantitative genetic framework and laid the basis for the hybridization of wheat, having fine-tuned major genes of plant stature and floral traits [68]. The development of hybrid cultivars at a commercial scale could be feasible through genome-based prediction of these genes.

Advertisement

6. Developing climate-resilient cultivars through biotechnology

In the last 15 years, recombinant DNA technology, genetic manipulation technologies, and culturing methodologies have enabled the efficient transformation and development of transgenics in a wide variety of crop plants [69]. Moreover, transgenesis can be a supplementary technique for single-gene or transgenic plants development [70]. Despite traditional breeding, this method introduces only the cloned gene(s) of agronomic significance without the hazard of transferring any additional undesired genes from the donor [71]. In transgenesis, the backcrossing is unnecessary because the recipient plant/crop genotype is least affected [72]. Furthermore, this genetic transformation method opens the door to a vast array of genetic material, sourced from viruses and bacteria to fungus, insects to animals and human beings to unrelated plants, and even from chemical synthesis [73, 74, 75, 76]. Plant transgenics have been developed and tested for various crops [77], fruits, and trees with surprising speed and success. However, the breeders focus on the gradual enhancement of commercial cultivars by introducing cloned genes of important agronomic values. The gene-transfer methods and strategies have been used to create important agronomic features in numerous crop varieties.

The enormous size and structural complexity of the polyploid wheat genome initially hindered the genomic study. By the passage of time, the development of new genomic technologies has enabled the breeders to map the bread wheat and its ancestors. However, the introduction of modern genomic technologies like next-generation sequencing has resulted in draught genomes for bread wheat and its progenitors [78], paving the door for developing novel crop enhancement strategies. Diverse germplasms are evaluated in pre-breeding for several physiological, agronomical, and biochemical traits [79], then crossing [80] and high throughput phenotyping [81] are required, while marker-based next-generation sequencing [80], genomic prediction [82], and validation of climate-resilient lines [83] can be very helpful in developing high yielding cultivars in changing environment. The CRISPR-Cas (Clustered Regularly Interspaced Short Palindromic Repeats) technology makes breeding time conservative and helps to find and transfer the same gene of interest in the host wheat genotype [84]. These RNA-guided nucleases are used for genome editing and isolated from the microbial adaptive immune system [85]. A wheat line has been developed that has reduced gluten in the grain by using Cas9 protein with 20 nucleotides in its sequence [86]. A gene Mildew locus O (MLo) was inserted in the wheat genome to introduce resistance against powdery mildew, and this was the first successful attempt of CISPER/Cas9 technology in wheat [87]. Transcription activator-like effector nucleases, an earlier technology, were used with the combination of CRISPER/Cas9 to achieve this goal.

Since the development of CRISPR-Cas technology, wheat genes of agronomical and fundamental scientific interest have been targeted, such as -gliadin genes to reduce gluten grain content [88], TaGW2 to increase grain weight [89], TaZIP4-B2 to understand meiotic homologous crossover [90], TaQsd1 to minimize preharvest sprouting [91], TaMTL and CENH3 for haploid plant induction [92]. The Wheat CRISPR tool, which is freely accessible at https://crispr.bioinfo.nrc.ca/WheatCrispr/, identifies efficient sgRNAs that are anticipated high on-target and low off-target activity scores (enabling researchers to explore all potential sgRNAs inside a target gene or sequence of interest). The Wheat CRISPR tool considers hexaploidy in bread wheat, allowing the researcher to target either a single gene copy or all three homeologs by checking a box.

The recent approaches for modern wheat breeding in the era of genomic studies have been summarized in Table 1.

Breeding techniquesOutputReferences
Marker-assisted selection (600k SNP marker)Targeted genotyping and genetic improvement[15]
Marker-assisted breeding (SNPs)Evaluation of multiple elite traits[93]
Marker-assisted back-crossingDrought tolerance in bread wheat[55]
Marker-assisted back-crossingIntrogression of drought-tolerant QTLs[94]
Marker-assisted back-cross selectionImprovement in rust resistance through a selection of Yr59[95]
Marker-assisted back-crossingTransfer of recessive skr cross-ability trait[96]
Marker-assisted back-crossingAdaptation of a variety of Unnat PBW 343 in diverse environments[97]
Marker-assisted back-crossingEnhanced rust resistance
Two genes (Lr19/Sr25 and Lr24/Sr24) for leaf rust resistance
One gene (Yr15) for stripe rust resistance
[98]
Marker-assisted back-crossingDevelopment of near-isogenic lines for grain softness[99]
Marker-assisted back-crossingDevelopment of advanced lines for grain softness[100]
Marker-assisted recurrent selectionImproved crown rot resistance[101]
Marker-assisted recurrent selectionEnhanced genetic gains[49]
Reciprocal recurrent selectionHybridization[38]
S1 recurrent selection, early generation genomic selection, marker-assisted back-crossing, and gene pyramidingIntrogression of Ms3 gene for genetic male sterility in hybrid wheat[102]
Double haploid breedingDevelopment of thermos-sensitive genic male sterile lines[103]
Biotechnology (Horizontal gene transfer)Fhb7 from fungus expression in wheat for Fusarium head blight resistance[75]
CRISPR-Cas9-based multiplexed gene editingheritable mutations in the TaGW2, TaLpx-1, and TaMLO genes[104]
Development of the GlutEnSeq (Gluten gene Enrichment and Sequencing)Homozygous deletions for the α-gliadins on 6A and the γ-gliadins on 1B in two γ-irradiated lines of cultivar
Homozygous deletions of the γ-gliadins on 1B and heterozygous deletions for the α-gliadins on 6A in four Fielder CRISPR/Cas9 gliadin gene-edited lines
[105]
CRISPR/Cas9 system delivered via Agrobacterium tumefaciensObtained thirteen mutant lines by targeting seven sites of three genes (Pinb, waxy, and DA1)[106]

Table 1.

Recent approaches for modern wheat breeding in the era of genomic studies.

Advertisement

7. Conclusion and future perspective

Wheat is one of the most important food crops and a basic source of calories globally. The rising population necessitates an increase in wheat production for food security. On the other hand, its production faces great challenges under changing climate and global warming. Wheat production is still lower than its demand and conventional methods proved inefficient to cope with this gap. Modern plant breeding techniques need time to be adopted for sustainable wheat production. Genomic breeding and biotechnological tools are more precise and time-conserving techniques with maximum efficiency to increase wheat production. The discovery of molecular markers-initiated, marker-assisted breeding while QTL and meta-QTL analysis improved the technique’s efficacy. Genomic breeding is considered an advanced form of MAS and the genome-wide study provides quick backcrossing and recurrent selection in wheat breeding. Genomic heterotic patterns have been used in molecular hybridization, while different statistical models have also been made to make selection and hybridization more and more precise. Combining genomic knowledge with biotechnological tools makes it quick to breed wheat with sustainable goals in a limited time in changing climate. In the future, the adoption of genomic breeding and biotechnological techniques to develop climate-resilient wheat cultivars at commercial scales will only be the way to achieve sustainable wheat production.

Advertisement

Acknowledgments

We acknowledge all colleagues in the respective departments who guided and helped in the collection of material and outlines.

We do not have any funding source for this book chapter

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Acronyms and abbreviations

MAS(Marker-assisted selection)
GS(Genomic selection)
QTL(Quantitative trait loci)
GEBVs(Genome-estimated breeding values)
GWAS(genome-wide association study)
RFLP(Restriction fragment length polymorphism)
ITMI(International triticeae mapping initiative)
SSR(Simple sequence repeat)
RAPD(Random amplified polymorphic DNA)
SCAR(Sequence characterized amplified regions)
STS(Sequence tagged sites)
CRISPER(Clustered regularly interspaced short palindromic repeats)

References

  1. 1. Hassan NA. Afghan Wheat and Flour Market
  2. 2. Hossain A, Skalicky M, Brestic M, Maitra S, Ashraful Alam M, Syed MA, et al. Consequences and mitigation strategies of abiotic stresses in wheat (Triticum aestivum L.) under the changing climate. Agronomy. 2021;11(2):241
  3. 3. Jasrotia P, Kashyap PL, Bhardwaj AK, Kumar S, Singh G. Scope and applications of nanotechnology for wheat production: A review of recent advances. Wheat Barley and Research. 2018;10(1):1-14
  4. 4. Altaf A, Gull S, Zhu X, Zhu M, Rasool G, Ibrahim MEH, et al. Study of the effect of peg-6000 imposed drought stress on wheat (Triticum aestivum L.) cultivars using relative water content (RWC) and proline content analysis. Pakistan Journal of Agricultural Sciences. 2021;58(1):357-367
  5. 5. Khatodia S, Bhatotia K. Prospects of advanced genomics for development of climate resilient wheat genotypes. Ekin Journal of Crop Breeding and Genetics. 2019;5(1):54-55. DOI: 10.1007/978-3-319-59379-1
  6. 6. Barbieri V, Gualtieri ML, Siligardi C. Wheat husk: A renewable resource for bio-based building materials. Construction and Building Materials. 2020;251:118909. DOI: 10.1016/j.conbuildmat.2020.118909
  7. 7. Mahmood N, Arshad M, Kächele H, Ma H, Ullah A, Müller K. Wheat yield response to input and socioeconomic factors under changing climate: Evidence from rainfed environments of Pakistan. Science of the Total Environment. 2019;688:1275-1285. DOI: 10.1016/j.scitotenv.2019.06.266
  8. 8. Dong C, Yin Q , Lane KJ, Yan Z, Shi T, Liu Y, et al. Competition and transmission evolution of global food trade: A case study of wheat. Physica A: Statistical Mechanics and its Applications. 2018;509:998-1008. DOI: 10.1016/j.physa.2018.06.052
  9. 9. Tadesse W, Sanchez-Garcia M, Assefa SG, Amri A, Bishaw Z, Ogbonnaya FC, et al. Genetic gains in wheat breeding and its role in feeding the world. Crop Breeding Genetics and Genomics. 2019;1:e190005
  10. 10. Ray DK, Mueller ND, West PC, Foley JA. Yield trends are insufficient to double global crop production by 2050. PLOS ONE. 2013;8(6):e66428. DOI: 10.1371/journal.pone.0066428
  11. 11. Kahiluoto H, Kaseva J, Balek J, Olesen JE, Ruiz-Ramos M, Gobin A, et al. Decline in climate resilience of European wheat. Proceedings of the National Academy of Sciences. 2019;116(1):123-128. DOI: 10.1073/pnas.1804387115
  12. 12. Sehgal D, Rosyara U, Mondal S, Singh R, Poland J, Dreisigacker S. Incorporating genome-wide association mapping results into genomic prediction models for grain yield and yield stability in CIMMYT spring bread wheat. Frontiers in Plant Science. 2020;11:197. DOI: 10.3389/fpls.2020.00197
  13. 13. Larkin DL, Lozada DN, Mason RE. Genomic selection—considerations for successful implementation in wheat breeding programs. Agronomy. 2019;9(9):479. DOI: 10.3390/agronomy9090479
  14. 14. Robert P, Auzanneau J, Goudemand E, Oury F-X, Rolland B, Heumez E, et al. Phenomic selection in wheat breeding: Identification and optimisation of factors influencing prediction accuracy and comparison to genomic selection. Theoretical and Applied Genetics. 2022;35:895-914. DOI: 10.1007/s00122-021-04005-8
  15. 15. Sun C, Dong Z, Zhao L, Ren Y, Zhang N, Chen F. The Wheat 660K SNP array demonstrates great potential for marker-assisted selection in polyploid wheat. Plant Biotechnology Journal. 2020;18(6):1354-1360. DOI: 10.1111/pbi.13361
  16. 16. Rasheed A, Xia X. From markers to genome-based breeding in wheat. Theoretical and Applied Genetics. 2019;132(3):767-784. DOI: 10.1007/s00122-019-03286-4
  17. 17. Munawar S, Mustafa G, Khan MS, Joyia FA. Role of biotechnology in climate resilient agriculture. In: Environment, Climate, Plant and Vegetation Growth. Cham: Springer; 2020. pp. 339-365
  18. 18. Pandurangan S, Workman C, Nilsen K, Kumar S. Introduction to Marker-Assisted Selection in Wheat Breeding: Accelerated Breeding of Cereal Crops. Humana, New York: Springer; 2022. pp. 77-117. DOI: 10.1007/978-1-0716-1526-3_3
  19. 19. Rasheed A, Jin H, Xiao Y, Zhang Y, Hao Y, Zhang Y, et al. Allelic effects and variations for key bread-making quality genes in bread wheat using high-throughput molecular markers. Journal of Cereal Science. 2019;85:305-309. DOI: 10.1016/j.jcs.2018.12.004
  20. 20. Iquebal MA, Sharma P, Jasrotia RS, Jaiswal S, Kaur A, Saroha M, et al. RNAseq analysis reveals drought-responsive molecular pathways with candidate genes and putative molecular markers in root tissue of wheat. Scientific reports. 2019;9(1):1-18. DOI: 10.1038/s41598-019-49915-2
  21. 21. Zhang LY, Liu DC, Guo XL, Yang WL, Sun JZ, Wang DW, et al. Genomic distribution of quantitative trait loci for yield and yield-related traits in common wheat. Journal of Integrative Plant Biology. 2010;52(11):996-1007. DOI: 10.1111/j.1744-7909.2010.00967.x
  22. 22. Desta ZA, Ortiz R. Genomic selection: Genome-wide prediction in plant improvement. Trends in Plant Science. 2014;19(9):592-601. DOI: 10.1016/j.tplants.2014.05.006
  23. 23. Poland JA, Endelman J, Dawson J, Rutkoski J, Wu S, Manes Y, et al. Genomic selection in wheat breeding using genotyping-by-sequencing. Plant Genome. 2012;5(3):103-113. DOI: 10.3835/plantgenome2012.06.0006
  24. 24. Lin L-C, Tsai T-H, Kuo C-L. Chemical constituents comparison of Codonopsis tangshen Codonopsis pilosula var. modesta and Codonopsis pilosula. Natural Product Research. 2013;27(19):1812-1815
  25. 25. lhan D. Molecular marker technologies in wheat. In: Current Academic Studies in Science and Mathematics Sciences-II. Lyon, France: Livre de Lyon; 2021. pp. 17-31
  26. 26. Yu M, Mao SL, Hou DB, Chen GY, Pu ZE, Li W, et al. Analysis of contributors to grain yield in wheat at the individual quantitative trait locus level. Plant Breeding. 2018;137(1):35-49. DOI: 10.1111/pbr.12555
  27. 27. Nadeem MA, Nawaz MA, Shahid MQ , Doğan Y, Comertpay G, Yıldız M, et al. DNA molecular markers in plant breeding: Current status and recent advancements in genomic selection and genome editing. Biotechnology & Biotechnological Equipment. 2018;32(2):261-285. DOI: 10.1080/13102818.2017.1400401
  28. 28. Kikuchi T, Hara M, Terao K. Development of a microsatellite marker set applicable to genome-wide screening of cynomolgus monkeys (Macaca fascicularis). Primates. 2007;48(2):140-146. DOI: 10.1007/s10329-006-0008-z
  29. 29. Paran I, Michelmore RW. Development of reliable PCR-based markers linked to downy mildew resistance genes in lettuce. Theoretical and Applied Genetics. 1993;85(8):985-993. DOI: 10.1007/BF00215038
  30. 30. Feuillet C, Keller B. Molecular markers for disease resistance: The example wheat. Molecular Marker Systems in Plant Breeding and Crop Improvement. 2006;55:353-370. DOI: 10.1007/3-540-26538-4_21
  31. 31. Jaiswal S, Sheoran S, Arora V, Angadi UB, Iquebal MA, Raghav N, et al. Putative microsatellite DNA marker-based wheat genomic resource for varietal improvement and management. Frontiers in Plant Science. 2017;8:2009. DOI: 10.3389/fpls.2017.02009
  32. 32. Haque MS, Saha NR, Islam MT, Islam MM, Kwon S-J, Roy SK, et al. Screening for drought tolerance in wheat genotypes by morphological and SSR markers. Journal of Crop Science and Biotechnology. 2021;24(1):27-39. DOI: 10.1007/s12892-020-00036-7
  33. 33. Jiang Y, Zhao Y, Rodemann B, Plieske J, Kollers S, Korzun V, et al. Potential and limits to unravel the genetic architecture and predict the variation of Fusarium head blight resistance in European winter wheat (Triticum aestivum L.). Heredity. 2015;114(3):318-326. DOI: 10.1038/hdy.2014.104
  34. 34. Ren J, Sun D, Chen L, You FM, Wang J, Peng Y, et al. Genetic diversity revealed by single nucleotide polymorphism markers in a worldwide germplasm collection of durum wheat. International Journal of Molecular Sciences. 2013;14(4):7061-7088. DOI: 10.3390/ijms14047061
  35. 35. Rufo R, Alvaro F, Royo C, Soriano JM. From landraces to improved cultivars: Assessment of genetic diversity and population structure of Mediterranean wheat using SNP markers. PloS one. 2019;14(7):e0219867. DOI: 10.1371/journal.pone.0219867
  36. 36. Chao S, Zhang W, Akhunov E, Sherman J, Ma Y, Luo M-C, et al. Analysis of gene-derived SNP marker polymorphism in US wheat (Triticum aestivum L.) cultivars. Molecular Breeding. 2009;23(1):23-33. DOI: 10.1007/s11032-008-9210-6
  37. 37. Gao L, Jia J, Kong X. A SNP-based molecular barcode for characterization of common wheat. PloS one. 2016;11(3):e0150947. DOI: 10.1371/journal.pone.0150947
  38. 38. Rembe M, Zhao Y, Jiang Y, Reif JC. Reciprocal recurrent genomic selection: An attractive tool to leverage hybrid wheat breeding. Theoretical and Applied Genetics. 2019;132(3):687-698. DOI: 10.1007/s00122-018-3244-x
  39. 39. Gao L, Turner MK, Chao S, Kolmer J, Anderson JA. Genome wide association study of seedling and adult plant leaf rust resistance in elite spring wheat breeding lines. PLoS One. 2016;11(2):e0148671. DOI: 10.1371/journal.pone.0148671
  40. 40. Soriano JM, Colasuonno P, Marcotuli I, Gadaleta A. Meta-QTL analysis and identification of candidate genes for quality, abiotic and biotic stress in durum wheat. Scientific Reports. 2021;11(1):1-15. DOI: 10.1038/s41598-021-91446-2
  41. 41. Endelman JB. Ridge regression and other kernels for genomic selection with R package rrBLUP. The Plant Genome. 2011;4(3):250-255. DOI: 10.3835/plantgenome2011.08.0024
  42. 42. Heffner EL, Jannink JL, Sorrells ME. Genomic selection accuracy using multifamily prediction models in a wheat breeding program. The Plant Genome. 2011;4(1):65-75. DOI: 10.3835/plantgenome2010.12.0029
  43. 43. Zhao Y, Mette M, Gowda M, Longin C, Reif J. Bridging the gap between marker-assisted and genomic selection of heading time and plant height in hybrid wheat. Heredity. 2014;112(6):638-645. DOI: 10.1038/hdy.2014.1
  44. 44. Hill WG, Goddard ME, Visscher PM. Data and theory point to mainly additive genetic variance for complex traits. PLoS Genetics. 2008;4(2):e1000008. DOI: 10.1371/journal.pgen.1000008
  45. 45. Schopp P, Müller D, Technow F, Melchinger AE. Accuracy of genomic prediction in synthetic populations depending on the number of parents, relatedness, and ancestral linkage disequilibrium. Genetics. 2017;205(1):441-454. DOI: 10.1534/genetics.116.193243
  46. 46. Müller D, Schopp P, Melchinger AE. Persistency of prediction accuracy and genetic gain in synthetic populations under recurrent genomic selection. G3: Genes, Genomes, Genetics. 2017;7(3):801-811. DOI: 10.1534/g3.116.036582
  47. 47. Varona L, Legarra A, Toro MA, Vitezica ZG. Non-additive effects in genomic selection. Frontiers in Genetics. 2018;9(78). DOI: 10.3389/fgene.2018.00078
  48. 48. Müller D, Schopp P, Melchinger AE. Selection on expected maximum haploid breeding values can increase genetic gain in recurrent genomic selection. G3: Genes, Genomes, Genetics. 2018;8(4):1173-1181. DOI: 10.1534/g3.118.200091
  49. 49. Singh M, Nara U, Kumar A, Thapa S, Jaswal C, Singh H. Enhancing genetic gains through marker-assisted recurrent selection: From phenotyping to genotyping. Cereal Research Communications. 2021:1-16. DOI: 10.1007/s42976-021-00207-4
  50. 50. Ali Y, Khan MA, Atiq M, Hussain M. Novel gene pyramiding to combat rusts in global wheat varieties against prevalent virulence: A review. G3: Genes, Genomes, Genetics. 2018;34(4):797-810
  51. 51. Emebiri L, Singh S, Tan M-K, Singh PK, Fuentes-Dávila G, Ogbonnaya F. Unravelling the complex genetics of Karnal bunt (Tilletia indica) resistance in common wheat (Triticum aestivum) by genetic linkage and genome-wide association analyses. G3: Genes, Genomes, Genetics. 2019;9(5):1437-1447. DOI: 10.1534/g3.119.400103
  52. 52. Burow MD, Blake TK. Molecular Tools for the Study of Complex Traits: Molecular Dissection of Complex Traits. Boca Raton: CRC Press; 2019. pp. 13-30. DOI: 10.1201/9780429117770
  53. 53. Ciucă M, Todorvska E, Kolev S, Nicolae R, Guinea I, Saulescu N. Marker-assisted selection (MAS) for drought tolerance in wheat using markers associated with membrane stability. FUNDULEA. 2009;12:7-12
  54. 54. Gautam T, Saripalli G, Kumar A, Gahlaut V, Gadekar D, Oak M, et al. Introgression of a drought insensitive grain yield QTL for improvement of four Indian bread wheat cultivars using marker assisted breeding without background selection. Journal of Plant Biochemistry and Biotechnology. 2021;30(1):172-183. DOI: 10.1007/s13562-020-00553-0
  55. 55. Rai N, Bellundagi A, Kumar PK, Kalasapura Thimmappa R, Rani S, Sinha N, et al. Marker-assisted backcross breeding for improvement of drought tolerance in bread wheat (Triticum aestivum L. em Thell). Plant Breeding. 2018;137(4):514-526. DOI: 10.1111/pbr.12605
  56. 56. Motomura Y, Kobayashi F, Iehisa JC, Takumi S. A major quantitative trait locus for cold-responsive gene expression is linked to frost-resistance gene Fr-A2 in common wheat. Breeding Science. 2013;63(1):58-67. DOI: 10.1270/jsbbs.63.58
  57. 57. Ni F, Qi J, Hao Q , Lyu B, Luo M-C, Wang Y, et al. Wheat Ms2 encodes for an orphan protein that confers male sterility in grass species. Nature Communications. 2017;8(1):1-12. DOI: 10.1038/ncomms15121
  58. 58. Xia C, Zhang L, Zou C, Gu Y, Duan J, Zhao G, et al. A TRIM insertion in the promoter of Ms2 causes male sterility in wheat. Nature Communications. 2017;8(1):1-9. DOI: 10.1038/ncomms15407
  59. 59. Tucker EJ, Baumann U, Kouidri A, Suchecki R, Baes M, Garcia M, et al. Molecular identification of the wheat male fertility gene Ms1 and its prospects for hybrid breeding. Nature Communications. 2017;8(1):1-10. DOI: 10.1038/s41467-017-00945-2
  60. 60. Wang Z, Li J, Chen S, Heng Y, Chen Z, Yang J, et al. Poaceae-specific MS1 encodes a phospholipid-binding protein for male fertility in bread wheat. Proceedings of the National Academy of Sciences. 2017;114(47):12614-12619. DOI: 10.1073/pnas.1715570114
  61. 61. Kempe K, Rubtsova M, Gils M. Split-gene system for hybrid wheat seed production. Proceedings of the National Academy of Sciences. 2014;111(25):9097-9102. DOI: 10.1073/pnas.1402836111
  62. 62. Longin CFH, Gowda M, Mühleisen J, Ebmeyer E, Kazman E, Schachschneider R, et al. Hybrid wheat: Quantitative genetic parameters and consequences for the design of breeding programs. Theoretical and Applied Genetics. 2013;126(11):2791-2801. DOI: 10.1007/s00122-013-2172-z
  63. 63. Zhao Y, Li Z, Liu G, Jiang Y, Maurer HP, Würschum T, et al. Genome-based establishment of a high-yielding heterotic pattern for hybrid wheat breeding. Proceedings of the National Academy of Sciences. 2015;112(51):15624-15629. DOI: 10.1073/pnas.1514547112
  64. 64. Würschum T, Leiser WL, Langer SM, Tucker MR, Longin CFH. Phenotypic and genetic analysis of spike and kernel characteristics in wheat reveals long-term genetic trends of grain yield components. Theoretical and Applied Genetics. 2018;131(10):2071-2084. DOI: 10.1007/s00122-018-3133-3
  65. 65. Boeven PH, Longin CFH, Leiser WL, Kollers S, Ebmeyer E, Würschum T. Genetic architecture of male floral traits required for hybrid wheat breeding. Theoretical and Applied Genetics. 2016;129(12):2343-2357
  66. 66. Lu Q , Lillemo M, Skinnes H, He X, Shi J, Ji F, et al. Anther extrusion and plant height are associated with Type I resistance to Fusarium head blight in bread wheat line ‘Shanghai-3/Catbird’. Theoretical and Applied Genetics. 2013;126(2):317-1334. DOI: 10.1007/s00122-012-1981-9
  67. 67. Würschum T, Liu G, Boeven PH, Longin CFH, Mirdita V, Kazman E, et al. Exploiting the Rht portfolio for hybrid wheat breeding. Theoretical and Applied Genetics. 2018;131(7):1433-1442. DOI: 10.1007/s00122-018-3088-4
  68. 68. Jiang Y, Schmidt RH, Zhao Y, Reif JC. A quantitative genetic framework highlights the role of epistatic effects for grain-yield heterosis in bread wheat. Nature Genetics. 2017;49(12):1741-1746. DOI: 10.1038/ng.3974
  69. 69. Lassoued R, Macall DM, Smyth SJ, Phillips PW, Hesseln H. How should we regulate products of new breeding techniques? Opinion of surveyed experts in plant biotechnology. Biotechnology Reports. 2020;26:e00460. DOI: 10.1016/j.btre.2020.e00460
  70. 70. Kumar K, Gambhir G, Dass A, Tripathi AK, Singh A, Jha AK, et al. Genetically modified crops: Current status and future prospects. Planta. 2020;251(4):1-27. DOI: 10.1007/s00425-020-03372-8
  71. 71. Guo M, Ye J, Gao D, Xu N, Yang J. Agrobacterium-mediated horizontal gene transfer: Mechanism, biotechnological application, potential risk and forestalling strategy. Biotechnology Advances. 2019;37(1):259-270. DOI: 10.1016/j.biotechadv.2018.12.008
  72. 72. Li S, Lin D, Zhang Y, Deng M, Chen Y, Lv B, et al. Genome-edited powdery mildew resistance in wheat without growth penalties. Nature. 2022;602:455-460. DOI: 10.1038/s41586-022-04395-9
  73. 73. Ismagul A, Yang N, Maltseva E, Iskakova G, Mazonka I, Skiba Y, et al. A biolistic method for high-throughput production of transgenic wheat plants with single gene insertions. BMC Plant Biology. 2018;18(1):1-8. DOI: 10.1186/s12870-018-1326-1
  74. 74. Pallotta MA, Warner P, Kouidri A, Tucker EJ, Baes M, Suchecki R, et al. Wheat ms5 male-sterility is induced by recessive homoeologous A and D genome non-specific lipid transfer proteins. The Plant Journal. 2019;99(4):673-685. DOI: 10.1111/tpj.14350
  75. 75. Wang H, Sun S, Ge W, Zhao L, Hou B, Wang K, et al. Horizontal gene transfer of Fhb7 from fungus underlies Fusarium head blight resistance in wheat. Science. 2020;368(6493):5435
  76. 76. Zhang L, Gao C, Chen C, Zhang W, Huang X-Y, Zhao F-J. Overexpression of rice OsHMA3 in wheat greatly decreases cadmium accumulation in wheat grains. Environmental Science & Technology. 2020;54(16):10100-10108. DOI: 10.1021/acs.est.0c02877
  77. 77. Ohly H, Broadley M, Joy E, Khan M, McArdle H, Zaman M, et al. The BiZiFED project: Biofortified zinc flour to eliminate deficiency in Pakistan. Nutrition Bulletin. 2019;44(1):60-64. DOI: 10.1111/nbu.12362
  78. 78. Jia M, Guan J, Zhai Z, Geng S, Zhang X, Mao L, et al. Wheat functional genomics in the era of next generation sequencing: An update. The Crop Journal. 2018;6(1):7-14. DOI: 10.1016/j.cj.2017.09.003
  79. 79. Sharma S, Schulthess AW, Bassi FM, Badaeva ED, Neumann K, Graner A, et al. Introducing beneficial alleles from plant genetic resources into the wheat germplasm. Biology. 2021;10(10):982. DOI: 10.3390/biology10100982
  80. 80. Sukumaran S, Krishna H, Singh K, Mottaleb KA, Reynolds M. Progress and prospects of developing climate resilient wheat in South Asia using modern pre-breeding methods. Current Genomics. 2021;22(6):440-449. DOI: 10.2174/1389202922666210705125006
  81. 81. Rebetzke G, Jimenez-Berni J, Fischer R, Deery D, Smith D. High-throughput phenotyping to enhance the use of crop genetic resources. Plant Science. 2019;282:40-48. DOI: 10.1016/j.plantsci.2018.06.017
  82. 82. Bhatta M, Sandro P, Smith MR, Delaney O, Voss-Fels KP, Gutierrez L, et al. Need for speed: Manipulating plant growth to accelerate breeding cycles. Current Opinion in Plant Biology. 2021;60:101986. DOI: 10.1016/j.pbi.2020.101986
  83. 83. Reynolds MP, Lewis JM, Ammar K, Basnet BR, Crespo-Herrera L, Crossa J, et al. Harnessing translational research in wheat for climate resilience. Journal of Experimental Botany. 2021;72(14):5134-5157. DOI: 10.1093/jxb/erab256
  84. 84. Hahn F, Sanjurjo Loures L, Sparks CA, Kanyuka K, Nekrasov V. Efficient CRISPR/Cas-mediated targeted mutagenesis in spring and winter wheat varieties. Plants. 2021;10(7):1481. DOI: 10.3390/plants10071481
  85. 85. Schiml S, Puchta H. Revolutionizing plant biology: Multiple ways of genome engineering by CRISPR/Cas. Plant Methods. 2016;12(1):1-9. DOI: 10.1186/s13007-016-0103-0
  86. 86. Jouanin A, Gilissen LJ, Schaart JG, Leigh FJ, Cockram J, Wallington EJ, et al. CRISPR/Cas9 gene editing of gluten in wheat to reduce gluten content and exposure—Reviewing methods to screen for coeliac safety. Frontiers in Nutrition. 2020;7:51. DOI: 10.3389/fnut.2020.00051
  87. 87. Abe F, Haque E, Hisano H, Tanaka T, Kamiya Y, Mikami M, et al. Genome-edited triple-recessive mutation alters seed dormancy in wheat. Cell Reports. 2019;28(5):1362-1369. DOI: 10.1016/j.celrep.2019.06.090
  88. 88. Sánchez-León S, Gil-Humanes J, Ozuna CV, Giménez MJ, Sousa C, Voytas DF, et al. Low-gluten, nontransgenic wheat engineered with CRISPR/Cas9. Plant Biotechnology Journal. 2018;16(4):902-910. DOI: 10.1111/pbi.12837
  89. 89. Liang Z, Chen K, Li T, Zhang Y, Wang Y, Zhao Q , et al. Efficient DNA-free genome editing of bread wheat using CRISPR/Cas9 ribonucleoprotein complexes. Nature Communications. 2017;8(1):1-5. DOI: 10.1038/ncomms14261
  90. 90. Rey M-D, Martín AC, Smedley M, Hayta S, Harwood W, Shaw P, et al. Magnesium increases homoeologous crossover frequency during meiosis in ZIP4 (Ph1 gene) mutant wheat-wild relative hybrids. Frontiers in Plant Science. 2018;9:509. DOI: 10.3389/fpls.2018.00509
  91. 91. Kamiya Y, Abe F, Mikami M, Endo M, Kawaura K. A rapid method for detection of mutations induced by CRISPR/Cas9-based genome editing in common wheat. Plant Biotechnology. 2020;37(2):247-251. DOI: 10.5511/plantbiotechnology.20.0404b
  92. 92. Lv J, Yu K, Wei J, Gui H, Liu C, Liang D, et al. Generation of paternal haploids in wheat by genome editing of the centromeric histone CENH3. Nature Biotechnology. 2020;38(12):1397-1401. DOI: 10.1038/s41587-020-0728-4
  93. 93. Li CX, Xu WG, Guo R, Zhang JZ, Qi XL, Hu L, et al. Molecular marker assisted breeding and genome composition analysis of Zhengmai 7698, an elite winter wheat cultivar. Scientific Reports. 2018;8(1):1-8. DOI: 10.1038/s41598-017-18726-8
  94. 94. Todkar L, Singh GP, Jain N, Singh PK, Prabhu KV. Introgression of drought tolerance QTLs through marker assisted backcross breeding in wheat (Triticum aestivum L.). Indian Journal of Genetics and Plant Breeding. 2020;80(2):209-212
  95. 95. Zhang M, Fang T, Zhou X, Chen X, Li X, Feng J, et al. Combination of marker-assisted backcross selection of Yr59 and phenotypic selection to improve stripe rust resistance and agronomic performance in four elite wheat cultivars. Agronomy. 2022;12(2):497. DOI: 10.3390/agronomy12020497
  96. 96. Bouguennec A, Lesage VS, Gateau I, Sourdile P, Jahier J, Lonnet P. Transfer of recessive skr crossability trait into well-adapted French wheat cultivar Barok through marker-assisted backcrossing method. Cereal Research Communications. 2018;46(4):604-615. DOI: 10.1556/0806.46.2018.043
  97. 97. Ram H, Gupta N, Singh G, Kaur H, Kaur J, Srivastva P. Performance of marker-assisted backcross bread-wheat (Triticum aestivum) variety Unnat PBW 343 under diverse environments.
  98. 98. Singh A, Jaiswal JP, Badoni S. Enhancing rust resistance in wheat through marker assisted backcross breeding. Indian Journal of Genetics and Plant Breeding. 2018;78(1):19-25
  99. 99. Kumar TP, Ahlawat AK, Singh SK, Rathan ND, Rai A, Bajpai K, et al. Enhancing rust resistance in wheat through marker Quality evaluation of near isogenic lines of wheat developed through marker assisted backcross breeding for grain softness. Indian Journal of Genetics and Plant Breeding. 2022;82(1):56-64
  100. 100. Rai A, Mahendru-Singh A, Raghunandan K, Kumar TP, Sharma P, Ahlawat AK, et al. Marker-assisted transfer of PinaD1a gene to develop soft grain wheat cultivars. 3 Biotech. 2019;9(5):1-10
  101. 101. Rahman M, Davies P, Bansal U, Pasam R, Hayden M, Trethowan R. Marker-assisted recurrent selection improves the crown rot resistance of bread wheat. Molecular Breeding. Mar 2020;40(3):1-4. DOI: 10.1007/s11032-020-1105-1
  102. 102. Guttieri MJ. Ms3 dominant genetic male sterility for wheat improvement with molecular breeding. Crop Science. 2020;60(3):1362-1372. DOI: 10.1002/csc2.20091
  103. 103. Li H, Li S, Abdelkhalik S, Shahzad A, Gu J, Yang Z, et al. Development of thermo-photo sensitive genic male sterile lines in wheat using doubled haploid breeding. BMC Plant Biology. 2020;20(1):1
  104. 104. Wang W, Pan Q , He F, Akhunova A, Chao S, Trick H, et al. Transgenerational CRISPR-Cas9 activity facilitates multiplex gene editing in allopolyploid wheat. The CRISPR Journal. 2018;1(1):65-74. DOI: 10.1089/crispr.2017.0010
  105. 105. Jouanin A, Borm T, Boyd LA, Cockram J, Leigh F, Santos BA, et al. Development of the GlutEnSeq capture system for sequencing gluten gene families in hexaploid bread wheat with deletions or mutations induced by γ-irradiation or CRISPR/Cas9. Journal of Cereal Science. 2019;88:157-166. DOI: 10.1016/j.jcs.2019.04.008
  106. 106. Zhang S, Zhang R, Song G, Gao J, Li W, Han X, et al. Targeted mutagenesis using the Agrobacterium tumefaciens-mediated CRISPR-Cas9 system in common wheat. BMC Plant Biology. 2018;18(1):1-2. DOI: 10.1186/s12870-018-1496-x

Written By

Zahid Manzoor, Junwei Liu, Muhammad Sheeraz Qadir, Muhammad Ahsan Jamil, Zeshan Hassan, Muhammad Shah Jahan and Amir Shakeel

Submitted: 08 March 2022 Reviewed: 30 March 2022 Published: 30 May 2022