Abstract
After being domesticated from teosinte, cultivated maize (Zea mays ssp. mays) spread worldwide and now is one of the most important staple crops. Due to its tremendous phenotypic and genotypic diversity, maize also becomes to be one of the most widely used model plant species for fundamental research, with many important discoveries reported by maize researchers. Here, we provide an overview of the history of maize domestication and key genes controlling major domestication-related traits, review the currently available resources for functional genomics studies in maize, and discuss the functions of most of the maize genes that have been positionally cloned and can be used for crop improvement. Finally, we provide some perspectives on future directions regarding functional genomics research and the breeding of maize and other crops.
Key words: maize, domestication, genomics, functional genomics, improvement
In this paper, we review the maize domestication process, available resources for genomic research, and positionally cloned genes underlying natural variation in important agronomic traits, and also discuss future directions for maize improvement.
Introduction
After Columbus arrived in the New World and brought maize to Europe, it subsequently spread throughout the world (Brandolini and Brandolini, 2009, Nunn and Nancy, 2010). Nowadays, its production exceeds that of wheat (Triticum aestivum) and rice (Oryza sativa) (http://www.fao.org). Indeed maize has become the most productive cereal crop and therefore has a massively important role, directly or indirectly, in feeding human beings. At the beginning of the last century, a controversy arose concerning the origin of maize. One hypothesis stated that maize is a species resulting from hybridization of a close relative of maize and Tripsacum; a second stated that the oldest wild maize is the ancestor of modern maize (Beadle, 1972). Subsequent genetic and archaeological evidence indicated that one subgroup of teosinte, Zea mays ssp. parviglumis, is the only ancestor of maize, a finding that is now widely accepted (Benz, 2001, Piperno and Flannery, 2001, Matsuoka et al., 2002, Vallebueno-Estrada et al., 2016). The divergence between Z. mays ssp. parviglumis and maize occurred about 9000 years ago around the Balsas River of southwest Mexico (Matsuoka et al., 2002).
Some key traits of teosinte are very different from those of maize, for example, its abundant branches and tillers, increased number of ears per plant, reduced number of kernels per ear (5–12 per ear for teosinte and several hundred for maize), and small kernels with a hardened fruitcase (reviewed in Doebley, 2004). How many loci were involved in the domestication of maize? More than 40 years ago, Beadle (1972) reported that he had planted a teosinte–maize F2 population consisting of 50 000 individuals and found that the frequency of parental types was ∼1 in 500, then estimated that there were four or five major loci involved in maize domestication. Later, Doebley and Stec (1993) mapped five major quantitative trait loci (QTLs) plus some minor-effect QTLs for key traits in which teosinte and maize differ. This result was consistent with Beadle's estimation and indicated that a small number of loci were responsible for the teosinte–maize morphological difference. Wright et al. (2005) investigated 774 genes and estimated that 2%–4% of maize genes were selected during maize domestication and subsequent improvement. According to recently released gene annotations of high-quality maize genomes, modern maize contains ∼39 000–42 000 protein-coding genes (Jiao et al., 2017, Springer et al., 2018, Sun et al., 2018, Yang et al., 2019a), indicating that ∼800–1700 protein-coding genes (39 000 × 2% ≈ 800; 42 000 × 4% ≈ 1700) underwent selection during the process of domestication. Enabled by the recent massive decrease in the cost of next-generation sequencing (NGS) technologies, population-scale genome resequencing has provided considerably more details about these selected regions. By resequencing improved maize varieties, landraces, and wild relatives, then scanning for selective sweeps, Hufford et al. (2012) identified 484 domestication loci, 107 of which were further selected during improvement, and 695 improvement loci covering ∼7.6% of the maize genome. These findings suggest that only a limited number of genes and a small proportion of the genome were selected during the maize domestication and improvement processes.
Among the candidate genes or loci identified to have undergone selection were some important domestication genes that had already been cloned (see Table 1). Teosinte branched 1 (tb1), which encodes a TEOSINTE BRANCHED1/CYCLOIDEA/PROLIFERATING CELL NUCLEAR ANTIGEN FACTOR (TCP) family transcription factor, was mapped with respect to key traits (i.e., inflorescence development and apical dominance) that distinguish teosinte from maize (Doebley et al., 1995). tb1 was then cloned by transposon tagging and shown to inhibit the outgrowth of axillary buds and enable the formation of female inflorescences (Doebley et al., 1997; reviewed in Doebley, 2004 and Doebley et al., 2006). The causal variant (a transposable element [TE]) of tb1 is located ∼60 kb upstream (according to the B73 V4 reference) of this gene (Figure 1A), which suggests that long-range chromatin interactions may have had an important role in maize domestication (Li et al., 2019b). This TE could enhance the expression of tb1 and then lead to repression of axillary growth and increased apical dominance through regulation of the expression of important development-related genes (such as cell-cycle genes pcna2 and mcm2) and direct/indirect gene-regulatory networks (Doebley et al., 2006, Studer et al., 2017). The role of tb1 in affecting apical dominance is not restricted to maize but has been conserved in other plant species. The rice homolog of tb1, OsTB1, also functions as a negative regulator of branching in rice (Takeda et al., 2003); RNAi of BRANCHED1 (BRC1) in Arabidopsis thaliana leads to a higher number of branches (Aguilar-Martínez et al., 2007); barley plants with the mutant allele of HvTB1 have a significantly higher number of tillers at the juvenile stage (Ramsay et al., 2011); and TaTB1 interacts with FLOWERING LOCUS T1 and regulates inflorescence architecture in bread wheat (Triticum aestivum) (Dixon et al., 2018). The conserved function of tb1 suggests its great potential for domesticating existing wild plants.
Table 1.
Key Domestication Genes Cloned in Maize.
| Gene | Phenotype | Functional annotation | Selection type | References |
|---|---|---|---|---|
| tb1 | Plant architecture | TCP transcription factor | Increased expression | Doebley et al., 1997, Studer et al., 2011 |
| gt1 | Plant architecture | Homeodomain leucine zipper | Increased expression | Whipple et al., 2011, Wills et al., 2013 |
| tru1 | Plant architecture | BTB/POZ ankyrin repeat protein | Increased expression | Dong et al., 2017 |
| tga1 | Hardened fruitcase | SBP-domain transcription factor | Protein function | Wang et al., 2005 |
| ZmSWEET4c | Seed filling | Hexose transporter | Increased expression | Sosso et al., 2015 |
| UB3 | Kernel row number | SBP-box transcription factor | Altered expression | |
| ids1/Ts6 | Kernel row number | AP2 transcription factor | Increased expression | Wang et al., 2019 |
|
ZmSh1-1 ZmSh1-5.1+ZmSh1-5.2 |
Shattering | YABBY transcription factor | Protein function | Lin et al., 2012 |
| ra1 | Inflorescence architecture | Transcription factor | Altered expression | Sigmon and Vollbrecht, 2010 |
Figure 1.
Genomic Sequence Variants in the tb1 Regions of Teosinte and Tropical and Temperate Maize Lines.
(A) The red rectangles indicate the position of tb1 and the blue rectangles indicate the position of the Hopscotch TE. This TE is the functional variant of tb1 and is absent in teosinte. This Hopscotch TE is located ∼60 kb upstream of tb1 in temperate line B73 (RefGen_v4, Jiao et al., 2017) and tropical line SK (Yang et al., 2019a).
(B–D) The increased expression levels of representative selected genes (tb1 in B, ZmSWEET4c in C, ra1 in D) in modern elite maize lines compared with teosinte, the ancestor of maize. The expression profile was obtained by analyzing RNA-seq data generated by Lemmon et al. (2014).
Another well-characterized maize domestication gene is tga1 (teosinte glume architecture 1), which encodes a squamosa-promoter binding protein (SBP) transcription factor. tga1 is responsible for the shift from kernels encased in a hardened fruitcase in teosinte to naked kernels exposed on the ear of modern maize. This QTL was mapped to a single Mendelian locus on chromosome 4 (Dorweiler et al., 1993) and was subsequently cloned (Wang et al., 2005). The causal variant of tga1 is a single-nucleotide polymorphism (SNP) in the first exon that leads to a single amino acid change (Lys to Asn) at the sixth amino acid of TGA1, a change that affects the binding of TGA1 to its targets (Wang et al., 2005). Interestingly, tga1 is regulated by tb1 through direct binding of TB1 to two GGNCCC motifs in the promoter region of tga1 (Studer et al., 2017).
In addition to tb1 and tga1, dozes of genes have been isolated that control key traits for the transition from teosinte to maize (Table 1). Most of these domestication genes are transcription factors, and their expression levels were upregulated during domestication (Figure 1B–1D). Thus, their altered expression likely affected the expression of large numbers of downstream genes, including additional transcription factors, as illustrated by the regulatory network in which tb1 and tga1 are involved (Studer et al., 2017). This suggests that selection at the level of a regulatory network might be much more efficient in driving phenotypic changes as compared with variations that affect the function of an individual protein (Liu et al., 2015c).
Recently, two studies using ChIA-PET (chromatin interaction analysis by paired-end tag sequencing) technology mapped genome-wide chromatin interactions and revealed their connections to gene-expression regulation (Li et al., 2019b, Peng et al., 2019), including the chromatin interactions in which TB1, UB3, ZmCCT9, Vgt1 involved. Likely, population-scale identification of chromatin interactions would allow the detection of many more important regulatory elements, providing a number of useful selection targets for future maize improvement.
Maize Functional Genomics Research Resources
Reference Genome Sequences
The first maize reference genome sequence (B73 RefGen_v1) was released in 2009 (Schnable et al., 2009) and relied on the sequencing of bacterial artificial chromosomes and fosmids; a subsequent release in 2017 was improved by the use of single-molecule technologies (Jiao et al., 2017). Since the release of the B73 reference genome sequence, it has been extensively used for maize functional genomics research. By comparing other inbred lines with B73, millions of SNPs and insertions/deletions (InDels; e.g., those of tb1 as illustrated in Figure 1) were identified, along with many structural variations (SVs) (examples of which are shown in Figure 2), presence/absence variation (PAV), expression presence/absence variation (ePAV), and copy-number variation (Springer et al., 2009, Lai et al., 2010, Jiao et al., 2012, Fu et al., 2013, Hirsch et al., 2014, Lu et al., 2015, Jin et al., 2016, Bukowski et al., 2018, Sun et al., 2018, Yang et al., 2019a).
Figure 2.
Examples of Large Structural Variations between Two Maize Lines, B73 and Mo17.
(A) An ∼2-Mb inversion on chromosome 7, as indicated by blue dots and dashed box.
(B) Two adjacent inversions covering ∼2 Mb on chromosome 1, as indicated by blue dots and dashed boxes.
(C) An ∼3-Mb deletion in Mo17, as compared with B73, on chromosome 6.
(D) An ∼3-Mb insertion in Mo17, as compared with B73, on chromosome 6.
The alignment was performed using MUMMER 3.23 (Kurtz et al., 2004) with a minimum match length of 1 kb.
The use of high-density SNPs, which were identified by resequencing maize lines and populations and mapping those sequences to the B73 reference genome, has greatly expanded our knowledge concerning maize domestication and development (Hufford et al., 2012) and the genetic basis of important traits such as flowering time (Buckler et al., 2009), disease resistance (Kump et al., 2011), oil and vitamin metabolism (Li et al., 2013, Wang et al., 2018a), plant height (Pan et al., 2017), and yield-related traits (Xiao et al., 2016, Liu et al., 2017c). Maize SNP arrays designed for molecular breeding by integrating SNPs associated with known genes and traits have been reported (Yan et al., 2010b, Xu et al., 2017). However, most resequencing research in maize and other crops has focused on identifying SNPs through mapping to a single reference genome, which limits the ability to make full use of genome data, detect SVs, and fully reveal the genetic diversity (Yang et al., 2019b). Recently, human graph-reference genomes, into which existing variations could be incorporated, have been used for alignments and genotyping (Garrison et al., 2018, Kim et al., 2019, Rakocevic et al., 2019). This strategy can take advantage of multiple genome sequences and reported variations and can increase the variant-calling accuracy, especially for SVs. We speculate that this graph-reference genome approach or other newly developed methods will soon be adopted by researchers studying maize and other crop genomes in order to integrate the increasing amounts of variation data and genome sequences that are becoming available.
A further disadvantage of resequencing studies is that most of them fail to detect associations between SVs and traits, despite growing evidence that SVs affect important agronomic traits, e.g., flowering time, plant height, kernel weight, disease resistance, and metabolic traits (Hirsch et al., 2014, Lu et al., 2015, Zuo et al., 2015, Jin et al., 2016). At a genome-wide level, it was demonstrated that 22% of SVs could not be detected using SNPs (Yang et al., 2019a). This indicates that more reference genome sequences are required to fully characterize the maize functional genome.
With the development of new sequencing technologies and the decreased costs of sequencing, several additional maize genome sequences have been reported since 2016. These include the genomes of PH207 (Hirsch et al., 2016), mexicana (Yang et al., 2017a), Mo17 (Yang et al., 2017a, Sun et al., 2018), W22 (Springer et al., 2018), HZS (Li et al., 2019a), and SK (Yang et al., 2019a). Among these released genomes, the B73, Mo17, W22, and SK genomes have high-quality assemblies (Table 2) and thus could be representative genomes for maize genomics research. The high-quality assemblies enabled by single-molecular sequencing, which are currently available for B73 Ref_V4, Mo17, and SK, have greater advantages in annotation of genome features and identification of important elements (such as promoters and TEs) and variations (especially structure variations) compared with assemblies based on short-read sequencing (Jiao et al., 2017, Sun et al., 2018, Yang et al., 2019a). Without the high-quality assembly of SK achieved by long-read sequencing, it would have taken tremendous effort to identify the large structure variations located upstream of ZmBAM1d, the gene underlying a major QTL affecting maize kernel size and weight (Yang et al., 2019a). There is no doubt that these genome sequences will greatly facilitate the revelation of maize genetic diversity, the connection between genetic variation and phenotypic variation, and the maize improvement process.
Table 2.
Summary of Released Maize (Teosinte) Genomes.
| Genome | Assembly size (Mb) | No. of scaffolds | Scaffold N50 | Contig N50 | No. of gene models | References |
|---|---|---|---|---|---|---|
| B73 | 2106 | 625 | 9.56 Mb | 1.18 Mb | 39 324 | Jiao et al., 2017 |
| Mo17 (CAU) | 2183 | 2560 | 10.2 Mb | 1.48 Mb | 38 620 | Sun et al., 2018 |
| W22 | 2134 | 306 | 35.52 Mb | 72.4 kb | 40 789 | Springer et al., 2018 |
| SK | 2161 | 708 | 73.24 Mb | 15.78 Mb | 43 271 | Yang et al., 2019a |
| PH207 | 2102 | 127 488 | 654 kb | 5.3 kb | 37 613 | Hirsch et al., 2016 |
| Mo17 (Yan) | 2042 | 48 268 | 3.00 Mb | 60.5 kb | 40 003 | Yang et al., 2017a |
| mexicana | 1204 | 107 418 | 108 kb | 26.6 kb | 31 387 | Yang et al., 2017a |
| HZS | 2209 | 12 | 223.93 Mb | 78.2 kb | 40 893 | Li et al., 2019a |
Mutant Libraries
Mutants are important resources for plant functional genomics research. Unlike Agrobacterium T-DNA insertion mutant libraries in Arabidopsis and rice (Alonso et al., 2003, Wu et al., 2003, Zhang et al., 2006), maize researchers mainly use transposon mutagenesis approaches to tag genes for cloning. Many genes for important agronomic traits have been cloned by transposon tagging. The most well known is tb1, a major domestication locus affecting tiller number and inflorescence development (Doebley et al., 1997). Several research programs have tagged maize genes with active Mu, including the Trait Utility System for Corn, Maize Targeted Mutagenesis database, Mu array, RescueMu, Photosynthetic Mutant Screen, and UniformMu (reviewed in Brutnell, 2002). Among these programs, UniformMu is currently the most widely used (McCarty et al., 2005). However, only ∼30% of maize genes have UniformMu insertions. Apart from transposon mutagenesis, targeting induced local lesions in genomes (TILLING) has also been used to construct maize mutant libraries (Till et al., 2004, Lu et al., 2018). In this approach, point mutations, caused by exposure to ethyl methanesulfonate, are detected with high-throughput methods or NGS. The mutant library constructed by Lu et al. (2018) covers ∼80% of maize genes (32 069 of 39 591), with each line containing on average 4500 mutations.
Nurtured and Natural Populations
As molecular markers were applied to construct linkage maps and to map QTLs for quantitative traits, many different kinds of mapping populations were generated, not only in maize but also in other crops, including classic F2, backcross, chromosome segment substitution line, recombinant inbred line (RIL), and multi-parent advanced generation intercross populations. In addition to these commonly used populations, maize researchers have constructed other types of high-resolution mapping populations that have been used extensively for genetic mapping. One such population is the intermated B73-Mo17 (IBM) population, a type of intermated RIL (IRIL) population. Unlike regular RIL populations, the F2 progenies of crosses between B73 and Mo17 were intermated for four generations before repeated selfing by single-seed descent to generate RILs. Because of the extra generations of intermating, the genetic map of this population has a 2.7-fold greater recombination fraction distance and a 3.86-fold longer map length (Lee et al., 2002), thus significantly improving the QTL mapping resolution. Another such population is a nested association mapping (NAM) population consisting of 25 RIL populations with a common parent line (B73) with a sequenced genome (Yu et al., 2008). This population captured ∼136 000 recombination events with three recombinations per gene on average. This genetic design took advantage of both linkage mapping and association mapping (McMullen et al., 2009). Both IBM and NAM populations have been used to dissect the genetic basis of complex traits, such as flowering time and disease resistance (Balint-Kurti et al., 2007, Buckler et al., 2009, Kump et al., 2011), and the population resources, including parental lines, are now publicly available (https://maizegdb.org/stock_catalog). Recently, Xiao et al. (2016) developed a random-open-parent association mapping population. This type of population has several advantages, the most impressive of which is that newly developed populations can be integrated into existing populations (reviewed in Xiao et al., 2017). It has been successfully used to dissect the genetic basis of cob traits (Xiao et al., 2016), kernel traits (Liu et al., 2017c), and plant architecture (Pan et al., 2017).
An association test between dwarf8 and flowering time was the first instance of association analysis being applied in plant genetics. This analysis tested associations between 123 polymorphic sites covering dwarf8 and flowering time in an association mapping population consisting of 92 maize inbred lines, and used 141 simple sequence repeat loci to control for population structure (Thornsberry et al., 2001). Thereafter, many maize natural populations were used for candidate-gene-association analyses (reviewed in Yan et al., 2011, Xiao et al., 2017, Liu and Yan, 2019), and association tests were subsequently extended to genome-wide association studies (GWASs) in maize and other plants including, but not limited to, Arabidopsis (Atwell et al., 2010), barley (Cockram et al., 2010), rice (Huang et al., 2011), and tomato (Sauvage et al., 2014). In maize, some natural populations have been widely used for GWAS research. One such population is the “Goodman” maize diversity panel, which contains 302 inbred lines and captures the diversity present in public breeding programs (Flint-Garcia et al., 2005). This panel has been genotyped using the Illumina MaizeSNP50 Array (Olukolu et al., 2013) and genotyping-by-sequencing (GBS; Romay et al., 2013), and is now covered by maize HapMap 3 (Bukowski et al., 2018). The “Goodman” panel has been used to dissect the genetic basis of ear rot resistance (Zila et al., 2013), iron homeostasis (Benke et al., 2015), aflatoxin resistance and important agronomic traits (Farfan et al., 2015), resistance to Fusarium verticillioides infection (Stagnati et al., 2019), and metabolite production (Zhou et al., 2019b). Another panel-based resource is the MaizeGo panel (http://www.maizego.org/Resources.html), which consists of 540 well-chosen lines and represents the largest association mapping population panel assembled for maize (Yang et al., 2011, Liu et al., 2017b). Many efforts to dissect the genetic architecture of complex traits have been carried out with this population or with a subset of this population, including analyses of expression traits (Fu et al., 2013, Liu et al., 2015c, Liu et al., 2017b), metabolite levels (Li et al., 2012, Li et al., 2013;; Wen et al., 2014, Wen et al., 2018, Deng et al., 2017, Wang et al., 2018a), agronomic traits (Yang et al., 2013, Yang et al., 2014), disease resistance (Chen et al., 2015, Ding et al., 2015, Li et al., 2019d), and drought tolerance (Liu et al., 2013, Mao et al., 2015, Wang et al., 2016, Xiang et al., 2017).
Databases and Datasets
The most valuable database for the maize research community is the Maize Genetics and Genomics Database (MaizeGDB; https://www.maizegdb.org/) (Portwood et al., 2019). This database was first released in the early 1990s and has since evolved into a “Swiss army knife” of datasets and tools for maize functional genomics research. First, it supports genome sequences of several maize inbred lines with a user-friendly genome browser. It also contains, among other features, large numbers of datasets covering stocks, polymorphisms, mutants and phenotypes, genetic maps, QTLs, gene annotations, and transcriptomic and proteomic data. Furthermore, it provides many tools for querying and visualizing datasets, such as SNPversity, which allows the visualization of the SNP diversity of a selected genomic region, and qTeller (https://qteller.maizegdb.org/), which allows the exploration of the expression of genes of interest. Finally, it provides additional services for the maize community such as the coordination of the annual maize genetics conference, recommendation of publications by the MaizeGDB Editorial Board, and handling of stock requests through the Maize Genetics Cooperation Stock Center (https://www.maizegdb.org/data_center/stock).
The maize community has generated large numbers of datasets, but in this review we will focus on population-scale genotypic variation and gene-expression datasets given their important proven connections with phenotypic variation. Accompanying the release of the maize B73 reference genome, the first-generation haplotype map of maize was also released. This HapMap 1 was constructed with 3.3 million SNPs and InDels identified within the founder lines of the NAM population (Gore et al., 2009). This HapMap subsequently evolved into HapMap 2 following the deep sequencing of 103 inbred lines covering 19 teosinte accessions, 23 landraces, and 60 improved lines, including the NAM founders of HapMap 1. HapMap 2 consists of 55 million SNPs, 21% of which are located in genic regions (Chia et al., 2012), and has been used to identify genomic regions that underwent selection during maize domestication and improvement (Hufford et al., 2012). More recently, maize HapMap 3 was built from NGS data from ∼1200 lines covering those used for HapMap 2, International Maize and Wheat Improvement Center (CIMMYT) lines, China Agricultural University (CAU) lines, and the Goodman panel. HapMap 3 contains ∼83 million polymorphisms (Bukowski et al., 2018). In addition to the HapMap populations, the MaizeGo panel has been genotyped with SNP arrays, RNA sequencing (RNA-seq), and GBS and also has a very high SNP density (2.65 million SNPs) (Yang et al., 2011, Fu et al., 2013, Liu et al., 2017b). All information about MaizeGo panel was integrated into a database called MODEM (http://modem.hzau.edu.cn/) (Liu et al., 2016).
Temporal and spatial gene-expression profiles are critical both for the functional analysis of genes and for understanding developmental processes. Most gene-expression profiling studies in maize have focused on maize seeds or seed parts at different stages of development in order to identify networks important for kernel and endosperm development and, thus, crop yield. Using deep RNA-seq, Fu et al. (2013) profiled the expression levels of 28 769 genes and 42 211 transcripts in kernels at 15 days after pollination (DAP) from 368 inbred lines, which were a subset of the MaizeGo panel. Similarly, Li et al. (2014) reported the temporal patterns of gene expression in kernels and endosperm during eight stages within 0–12 DAP; Chen et al. (2014) and Yi et al. (2019) quantified gene-expression levels in the embryo, endosperm, and whole seed at 0–38 DAP and 0–144 h after pollination, respectively. Recently, Zhan et al. (2015) used laser-capture microdissection technology to isolate five different endosperm compartments from B73 kernels at 8 DAP and then quantified the transcription level of genes within these compartments using RNA-seq. In addition to gene-expression quantification in kernel-related tissues, there have been other reports of gene-expression profiling in multiple tissues across the entire maize life cycle. These include 79 tissues from B73 (Stelpflug et al., 2016), 23 tissues from B73 (Walley et al., 2016), and 23 tissues from B73, Mo17, and their F1 hybrid (Zhou et al., 2019a). These diverse gene-expression profiles have already contributed much toward identifying networks for development and could also provide the first hints of the functions of genes of interest.
Genes Underlying Important Quantitative Traits
There are thousands of QTLs, and hundreds of genes have been identified that are important for maize agronomic traits. Among the resulting cloned genes, 464 “classic” genes were cloned based on visible morphological mutant phenotypes (Schnable and Freeling, 2011). Although these cloned mutant genes are functionally important for maize development, most of them cannot be used directly in maize-breeding programs due to their deleterious effects. In this review, we will pay much more attention to genes (Table 3) identified by positional cloning whose genetic polymorphisms are responsible for natural variation in important quantitative traits and are additionally promising candidates for marker-assisted selection (MAS).
Table 3.
Positionally Cloned Maize Genes Related to Important Quantitative Traits.
| Gene | Phenotype | Functional annotation | Causal variation | References |
|---|---|---|---|---|
| ZmRap2.7 | Flowering time | AP2-like transcription factor | MITE TE ∼70 kb upstream of this gene | Salvi et al., 2007, Castelletti et al., 2014 |
| ZmCCT10 | Flowering time Disease resistance |
CCT transcription factor | CACTA-like TE ∼2.5 kb upstream of this gene | Hung et al., 2012, Yang et al., 2013, Wang et al., 2017 |
| ZmCCT9 | Flowering time | CCT transcription factor | Harbinger-like TE ∼57 kb upstream of this gene | Huang et al., 2018 |
| ZmCLA4 | Leaf angle | Ortholog of LAZY1 in rice | – | Zhang et al., 2014 |
| ZmRAVL1 | Leaf angle | B3 domain–containing protein | 2-bp InDel ∼9.5 kb upstream of this gene | Tian et al., 2019 |
| brd1 | Leaf angle | Brassinosteroid C-6 oxidase | – | Tian et al., 2019 |
| ZmGA3ox2 | Plant height | GA3 β-hydroxylase | Two small InDels in the promoter | Teng et al., 2013 |
| qph1 | Plant height | ABC transporter | A mis-sense SNP | Xing et al., 2015 |
| UB3 | Kernel row number | SBP-box transcription factor | 1.2-kb transposon-containing insertion located ∼60 kb downstream of this gene | Liu et al., 2015b |
| ids1/Ts6 | Kernel row number | AP2 transcription factor | – | Wang et al., 2019 |
| ZmBAM1d | Kernel size and weight | CLV1/BAM-related receptor kinase-like protein | – | Yang et al., 2019a |
| DGAT1-2 | Oil content and composition | Diacylglycerol acyltransferase | Extra phenylalanine insertion | Zheng et al., 2008 |
| Zmfatb | Palmitic acid | Acyl-ACP thioesterase | 11-bp InDel in the sixth exon | Li et al., 2011 |
| ZmPORB2 | Tocopherol concentration | Protochlorophyllide oxidoreductase | A small InDel in 5’ UTR | Zhan et al., 2019 |
| ZmAuxRP1 | Disease resistance | Domain of unknown function 966 | – | Ye et al., 2019 |
| ZmPLA1/MTL/NLD | Haploid induction rate | Phospholipase A | A 4-bp insertion led to a frame shift | Gilles et al., 2017, Kelliher et al., 2017, Liu et al., 2017a |
| ZmDMP | Haploid induction rate | DUF679 domain membrane protein | One single-nucleotide change | Zhong et al., 2019 |
“–” indicates that a causal variant has not yet been confirmed.
Flowering Time-Related Genes
In the late 1990s, Vlăduţu et al. (1999) identified two tightly linked QTLs for flowering time and named them Vegetative to generative transition 1 and 2 (Vgt1 and Vgt2). Vgt1 was then mapped to a 1.3-cM interval (Salvi et al., 2002) and was finally cloned (Salvi et al., 2007). Vgt1 consists of a non-coding regulatory element located ∼70 kb upstream of an AP2-like transcription factor, ZmRap2.7. The role of ZmRap2.7 as a negative regulator of flowering time was confirmed in both overexpression and downregulation studies carried out by means of transgenesis (Salvi et al., 2007). Three polymorphisms (G/A/indel324, MITE, and ATindel434) in Vgt1 were found to be significantly associated with flowering time in two independent association mapping studies (Salvi et al., 2007, Ducrocq et al., 2008). The miniature inverted-repeat transposable element (MITE) insertion in Vgt1 was demonstrated to be highly methylated, and its flanking region was significantly more methylated compared with alleles without this MITE insertion (Castelletti et al., 2014). Allele-specific expression analysis of ZmRap2.7 in F1 plants showed that the two allele-specific transcripts exhibited significantly different expression levels (Castelletti et al., 2014).
ZmCCT is another major flowering-time QTL and was first mapped to a ∼170-kb region containing five putative genes in one parental line and eight in the other (Ducrocq et al., 2009). A CCT gene, ZmCCT10, which is homologous to rice Ghd7 (a large-effect locus for grain number, plant height, and heading date) (Xue et al., 2008), was proposed to be the candidate for this QTL (Ducrocq et al., 2009). This QTL was independently mapped to a ∼202-kb region that includes part of ZmCCT10, the only gene present in the QTL interval (Hung et al., 2012). The effect of ZmCCT10 on flowering time was confirmed by association analyses in two independent diverse panels (Hung et al., 2012, Yang et al., 2013) and also by transgenic analysis (Yang et al., 2013). The causal variation of ZmCCT is a 5122-bp CACTA-like TE located ∼2.5 kb upstream of ZmCCT10. This TE could lead to a change in the methylation level of the ZmCCT10 promoter that would affect the expression level of the gene (Yang et al., 2013). Another CCT gene, ZmCCT9, is the gene underlying qDTA9, a QTL for days to anthesis (DTA). This QTL was narrowed down to a ∼2.4-kb region located ∼57 kb upstream of ZmCCT9. A Harbinger-like TE was the most significant polymorphism associated with DTA based on an association analysis. In addition, knocking out ZmCCT9 with CRISPR/Cas9 resulted in earlier flowering under long-day conditions (Huang et al., 2018).
Interestingly, all three causal TEs underwent selection during maize adaptation to temperate regions. The frequency of the MITE allele of Vgt1 varies from 0.3 in tropical lines to 0.87 in the European and Northern elite lines, and there is a significant correlation between latitude and MITE frequency (Ducrocq et al., 2008). The frequency of the TE alleles of ZmCCT10 and ZmCCT9 increased from 51.4% and 10.3% in tropical lines to 85.4% and 79% in temperate lines, respectively (Huang et al., 2018). ZmCCT10, ZmCCT9, and CCT genes in other plants (such as rice Hd1 and Ghd7) have important roles in regulating flowering time, which indicates a conserved function among CCT genes. In maize there are 53 CCT genes, among which 15 are significantly associated with flowering time based on candidate-gene association analysis (Jin et al., 2018). These results underscore the substantial role of CCT genes and their related pathways in flowering-time manipulation.
Genes Regulating Plant Architecture
The use of dwarf and semi-dwarf cultivars in breeding programs was the key component of the first “Green Revolution.” This suggests the importance of plant architecture in crop yield improvement. Although many mutant genes for plant architecture have been cloned, only a few QTLs for leaf angle or plant height have been positionally cloned in maize.
ZmCLA4, an ortholog of rice OsLAZY1, is the functional gene responsible for a leaf-angle QTL, qLA4-1, with a semi-dominant effect of ∼15° (Zhang et al., 2014). ZmCLA4 was differentially expressed between plants with large and small leaf angles, with plants with larger leaf angles showing lower expression, indicating the negative role of ZmCLA4 in controlling leaf angle. This negative role was confirmed by RNAi of ZmCLA4 in maize and overexpression of ZmCLA4 in rice (Zhang et al., 2014). Rice plants that overexpressed ZmCLA4 had much smaller leaf angles than did controls, suggesting that ZmCLA4 has a conserved role in regulating leaf angle among different crops. This is supported by the finding that both mutant and RNAi lines of OsLAZY1 have larger tiller angles compared with their respective control lines (Li et al., 2007).
Recently, Tian et al. (2019) cloned two QTLs (Upright Plant Architecture1 and 2 [UPA1 and UPA2]) for upright plant architecture. The underlying genes for these two QTLs are brassinosteroid C-6 oxidase1 (brd1) and ZmRAVL1, which is a homolog of rice RAVL1. The functional variant in UPA2 is a 2-bp InDel located 9.5 kb upstream, which alters the binding affinity of the protein DRL encoded by drooping leaf1, another maize leaf-angle gene (Strable et al., 2017). While LG1, encoded by liguleless1, which is essential for leaf ligule and auricle development (Moreno et al., 1997), activates the expression of ZmRAVL1, the physical interaction between DRL1 and LG1 represses this activation (Tian et al., 2019). Interestingly, ZmRAVL1 regulates the expression level of brd1 and thus alters the endogenous brassinosteroid level and leaf angle (Tian et al., 2019).
qPH3.1 is a major QTL for plant height with a 10.0-cm additive effect and 3.7-cm dominant effect. This QTL was narrowed down to a 12.6-kb region, within which ZmGA3ox2 was shown to be responsible for both qPH3.1 and the dwarf-1 mutant (Teng et al., 2013). This gene, which encodes a gibberellin (GA) 3-oxidase, is expressed constitutively, and higher expression results in greater endogenous GA1 (a bioactive gibberellin) levels and taller plants. Association mapping results subsequently indicated that two 3-nucleotide repeats within this gene might be the causative polymorphism for plant-height variation (Teng et al., 2013).
qph1, another major QTL for plant height with a 17.7-cm additive effect and a 7.8-cm dominant effect, was mapped to a 1.59-kb region covered by the fifth exon of Br2. A mis-sense SNP leading to an amino acid change from Arg to Leu is the causal variant for this QTL (Xing et al., 2015). Mutants of maize Br2, which encodes an ATP-binding cassette transporter belonging to the multidrug-resistant class of P-glycoproteins, show shortening of the lower stalk internodes due to reduced auxin transport. In addition, an 882-bp duplication in exon 5 of the Br2 homolog in sorghum is responsible for the dw3 mutant phenotype and the instability of dw3 caused by unequal recombination at this duplication site (Multani et al., 2003).
Yield-Related Genes
Ear- and kernel-related traits are important components of maize yield. Two major QTLs (KRN4 and KRN1) have been cloned for kernel row number (KRN) and one (qHKW1) has been cloned for kernel size and weight.
KRN4 was mapped to a 3-kb intergenic region located ∼60 kb downstream of an SBP-box gene, UB3 (unbranched3), which negatively affects KRN (Chuck et al., 2014). A 1.2-kb PAV with a harbinger TE in this 3-kb region and a SNP (S35) in the third exon of UB3 additively contribute to KRN, and these two sites both underwent selection during maize improvement (Liu et al., 2015a). In both rice and maize, the UB3 protein binds to genes encoding inflorescence-related transcription factors that are involved in the cytokinin pathway, and UB3 additionally targets genes involved in the auxin pathway and CLV-WUS pathway in maize (Du et al., 2017).
krn1 was identified in an F2:3 population derived from crosses between the tropical line CML220 and landrace 1091AF, and had a phenotypic variation explained (PVE) value of 22%. This QTL was then fine-mapped to a 6.6-kb region, and the causal gene, which was confirmed by association analysis and two other independent mutation events, was shown to be a previously reported mutant gene, ids1/Ts6 (indeterminate spikelet 1/Tassel seed 6) (Wang et al., 2019). This gene is homologous to the wheat gene Q (Simons et al., 2006). Mutants of these two genes have similar phenotypes including altered spike length, spike density, grain size, and plant height, and both genes also underwent selection during the domestication of these two crop species (Debernardi et al., 2017, Xie et al., 2018, Wang et al., 2019).
qHKW1 accounts for 18.4% of the variation in hundred-kernel weight (HKW) in an RIL population derived from the modern elite maize inbred line ZHENG58 and the landrace SK (Raihan et al., 2016, Liu et al., 2017c). This QTL was fine-mapped to a ∼177-kb region containing only one gene, ZmBAM1d, which encodes a CLAVATA1 (CLV1)/BARELY ANY MERISTEM (BAM)-related receptor kinase-like protein. The increased expression of ZmBAM1d, which is caused both by disrupted chromatin interactions resulting from distal large InDels in the promoter and altered methylation levels in the promoter region, leads to enhanced HKW. A similar effect was noted in transgenic lines that overexpress ZmBAM1d, which show a ∼1.9-g increase in HKW (Yang et al., 2019a).
Disease Resistance-Related Genes
Disease is one of the largest contributors to maize yield loss. Strategies for cloning quantitative disease-resistance genes and the characterization of some of these cloned genes have been extensively reviewed in Yang et al. (2017b). Therefore, we focus here solely on two recently cloned disease-resistance genes, ZmCCT10 and ZmAuxRp1.
Two major QTLs (qRfg1 and qRfg2) conferring resistance to Gibberella stalk rot were identified, and qRfg1 was mapped to a ∼500-kb region (Yang et al., 2010a). qRfg1 was then narrowed down to a ∼170-kb region (Wang et al., 2017). Interestingly, ZmCCT10 was the gene underlying this QTL, and the CACTA-like TE, which had been shown to be the causal polymorphism for flowering time (Yang et al., 2013), was also the causal variant for stalk rot resistance. This TE results in altered histone modification and DNA methylation in the cis-regulatory region of ZmCCT10, which leads to changes in its expression and, thus, enhanced disease resistance (Wang et al., 2017).
qRfg2 was first mapped to a ∼300-kb region (Zhang et al., 2012) and then to a ∼2.6-kb region containing only one gene, ZmAuxRP1, which encodes a protein with a domain of unknown function (DUF966). RNAi of ZmAuxRP1 results in plants with greater resistance, whereas plants that overexpress this gene are more susceptible to Gibberella stalk rot (Ye et al., 2019). It turns out that this gene regulates resistance to Gibberella stalk rot by affecting the biosynthesis of both indole-3-acetic acid (IAA) and benzoxazinoids (Ye et al., 2019).
Nutrition-Related Genes
The 21st century has seen an increase in concerns about nutrition, as indicated by a PubMed search with the keywords “nutrition” or “metabolome,” which reveals a rapid increase in published studies since 2010. Searches with the phrase “maize nutrition” or “maize metabolome” produce similar results. Biofortification, the process of increasing nutrition in crops through plant breeding (Nestel et al., 2006), has been successfully applied in maize nutrition improvement through MAS for, for instance, protein content and provitamin A (Muthusamy et al., 2014, Zunjare et al., 2018). Selection of the recessive allele of Opaque2 increases the lysine content of kernels (Mertz et al., 1964) while favorable alleles of lcyE and crtRB1 enhance provitamin A levels in kernels (Harjes et al., 2008, Yan et al., 2010a).
A QTL (qHO6) that accounts for 11% and 9.5% of the variation in seed oil concentration and embryo oil concentration, respectively, was mapped to a 4.8-kb region. DGAT1-2, which encodes a type I acyl-coenzyme A:diacylglycerol acyltransferase, is the gene underlying this QTL (Zheng et al., 2008). Ectopic expression of the high-oil allele of this gene under the control of an embryo-specific promoter leads to a 27.9%, 26.1%, and 84.5% increase in seed oil, embryo oil, and oleic acid concentration, respectively, and to a 28.1% decrease in linoleic acid concentration. The causal polymorphism is the insertion of an extra phenylalanine at position 469 of DGAT1-2, which leads to the enhancement of its enzymatic activity (Zheng et al., 2008).
Palmitic acid (C16:0) is an important class of saturated fatty acids. Eight QTLs mapped using an RIL population derived from B73 (normal-oil line) and By804 (high-oil line) explain 65.3% of the total phenotypic variance in palmitic acid concentration, with pal9 having the largest effect (42.0% PVE) (Yang et al., 2010b). pal9 was then fine-mapped to a 90-kb region covering only one gene, Zmfatb, which encodes an acyl-ACP thioesterase. An 11-bp InDel in the last exon of Zmfatb is the causal variant detected by candidate-gene-association analysis, and this InDel introduces a premature stop codon that affects enzyme activity (Li et al., 2011).
ZmPORB2, encoding a protochlorophyllide oxidoreductase, was identified as the causal gene underlying a major QTL (qVE5) for tocopherol content in maize kernels mapped in three RIL populations (Wang et al., 2018a, Zhan et al., 2019). An InDel, called Indel058, located 59 bp upstream of the ATG of ZmPORB2, which allowed separation of diverse inbred lines into three groups (one group with no insertion, another two groups with 5- or 8-bp insertions), was the most significant variation detected by candidate-gene-association analysis and is likely the causal variant. Lines with no insertion had a much higher ZmPORB2 expression level and tocopherol concentration. Overexpression of this gene led to increased tocopherol content in both the leaf and kernel (Zhan et al., 2019), which suggests great potential in vitamin E biofortification.
These are by no means the only metabolic QTLs that are important for nutrition in maize, with several recent QTL studies based on maize introgression or RILs, or alternatively on association panels, profiling such nutritionally important compounds as essential amino acids, lipids, vitamins and flavonoids, and other antioxidants (Riedelsheimer et al., 2012, Wen et al., 2015, Wen et al., 2016). In a similar vein, although such studies have not yet been reported on a large scale in maize, a huge number of QTLs for taste have been found in studies in many species, most notably in tomato (Tieman et al., 2017, Gao et al., 2019, Zhao et al., 2019) and cucumber (Shang et al., 2014, Zhu et al., 2019). Encompassing both traits, the first papers studying the domestication of the plant metabolome have recently been published for wheat, tomato, and maize (Beleggia et al., 2016; Zhu et al., 2018; Li et al., 2019c, Xu et al., 2019). The first of these studies carried out in wheat demonstrated that a reduction in unsaturated fatty acids was associated with selection during domestication of emmer while changes in the amino acid content due to selection mark the domestication of durum wheat (Beleggia et al., 2016). Similarly, tomato domestication was characterized by a shift in the ratios of nutritional and antinutritional steroidal glycoalkaloids as well as alterations in phenylpropanoid content driven by the Asian preference for pink tomatoes and considerable changes in metabolism that were due to genes hitchhiking on those genes selected to improve tomato yield (Zhu et al., 2018). Two recent studies have carried out a detailed comparison of metabolism in large populations derived from maize-by-teosinte crosses, revealing large-scale changes in both primary (particularly in amino acids and lipids [Li et al., 2019c, Xu et al., 2019]) and secondary (particularly in alkaloids and terpenoids [Xu et al., 2019]) metabolism that occurred during domestication. A current complication in using insights derived from the metabolomics data described above for crop improvement is that often the metabolite function is not well characterized (Alseekh and Fernie, 2018). With increasing use of wild germplasm, such studies identifying the changes that occurred during domestication to make our food more palatable (Zhu et al., 2018) will, however, become essential in order to prevent the reintroduction of antinutritional compounds.
Lessons Learned from Characterization of Positionally Cloned Genes
One lesson we have learned from these positionally cloned genes is that SVs and TEs are of great importance in shaping plants and enhancing their fitness. SVs and TEs are causal variations not only for important QTLs (tb1, Vgt1, ZmCCT, ZmCCT9, KRN4, qHKW1) as discussed above, but also for other genes identified by GWASs (such as ZmNAC111 and ZmVPP1 for drought tolerance; Mao et al., 2015, Wang et al., 2016). TEs can lead to phenotypic plasticity in plants and humans through regulating gene expression (Wei and Cao, 2016). The second lesson is that both natural variants and mutant alleles of some cloned genes (ZmGA3ox2, Br2, UB3, ids1/Ts6) are responsible for variation in important quantitative traits. Other studies have had similar findings. For example, a weak allele of fea2 increases the KRN and the number of kernels per ear without changing ear length (Bommert et al., 2013). Liu et al. (2017b) found that some kernel-related mutant genes are enriched in QTLs for kernel traits and that SNPs in some of these genes are significantly associated with kernel traits, with the most significant SNPs being either located in the regulatory region or being synonymous substitutions. These findings together highlight the possibility of improving maize via a combination of knowledge concerning mutant genes and CRISPR/Cas9 technology, which, as has already been demonstrated (Eshed and Lippman, 2019, Fernie and Yan, 2019), could be used to generate considerably more favorable alleles.
The genetic basis of different traits varies substantially in maize. For flowering time and oil concentration, dozens of loci, each with a small effect, have been identified (Buckler et al., 2009, Li et al., 2013). Yield-related traits, such as plant architecture and ear and kernel traits, have a much more complex genetic architecture as indicated by the detection of several hundreds of loci (Xiao et al., 2016, Liu et al., 2017c, Pan et al., 2017). In contrast, there are fewer mapped QTLs for disease resistance. Taking gray leaf spot (GLS) as an example, only 16 loci were mapped in the NAM population, among which three had a large effect (Benson et al., 2015). This situation is not the same across crops. Unlike maize, rice heading and seed size and weight are regulated by only a handful of genes, each with a large effect (Huang et al., 2011). These differences suggest that the road to maize improvement should differ from that of rice, and researchers should select an appropriate method based on the genetic complexity of the desired traits. For traits mainly controlled by major QTLs, i.e., disease resistance, MAS would be a more efficient strategy, whereas genomic selection would be more suitable for traits with a much more complex genetic basis, such as flowering-time and yield-related traits (Figure 3A–3C).
Figure 3.
An Overview of Strategies for Rapid Gene Cloning and Maize Improvement.
(A–C) Genetic architecture of a target trait is dissected with, for example, linkage mapping, GWAS, QTG-seq, and QTG-seq. Different selection methods are then used based on the complexity of the genetic architecture of the target trait. For example, MAS is suitable for selection of GLS resistance, whereas GS is much more efficient for selection of hundred-kernel weight.
(D–G) After QTLs are mapped, candidate genes are proposed and the mutants for candidate genes are screened from a mutant library generated with CRISPR/Cas9. The causal gene is validated with these mutants. With causal variants and/or favorable alleles generated with CRISPR/Cas9 confirmed, they then can be applied directly in breeding programs. GWAS, genome-wide association study; MAS, marker-assisted selection; GS, genomic selection.
Perspectives
Based on the tremendous genotypic diversity revealed by resequencing studies in both maize and other plants (especially rice and Arabidopsis), we believe that more high-quality genome assemblies, including assemblies for teosinte, landraces, and tropical lines, are needed to comprehensively reveal maize variations. Another challenge is to rapidly clone and validate genes responsible for important agronomic traits. The step forward in methodology has greatly accelerated this process, especially the progress in transformation, CRISPR/Cas9 technology, statistical methods for speeding up QTL cloning (such as QTL sequencing [QTL-seq] and quantitative trait gene sequencing [QTG-seq] [Takagi et al., 2013, Zhang et al., 2019]), and haploid induction technologies. Using a combination of these methods to quickly map and validate candidate genes (with CRISPR/Cas9) could provide a revolutionary way to rapidly clone genes and generate favorable alleles that could be directly used in future breeding programs (Figure 3). In addition, many important QTLs, including tb1, UPA1, and UPA2, were mapped within populations derived from teosinte and cultivated maize, which indicates that teosinte is full of treasures waiting to be found. What studies with teosinte can provide is not only insights into domestication but also directions for future improvement and de novo domestication new crops (Fernie and Yan, 2019).
The Need for Additional Whole Genome Sequences from Diverse Maize Lines
Population-scale genome projects (the human 1000 Genomes Project, the Arabidopsis 1001 Genomes Project, and the rice 3000 Genomes Project) have shown that a handful of reference genome sequences is not enough to reveal the genetic variation within a species, especially low-frequency and rare variants that are also important components of the genetic basis of human diseases or plant traits. For example, according to the pilot phase results of the human 1000 Genomes Project, 55% of the ∼15 million SNPs identified are novel (1000 Genomes Project Consortium, 2010); According to pan-genome analysis results from the rice 3000 Genome Project, each rice accession. has at least 26.5% distributed gene families (Wang et al., 2018b).
The genetic diversity of maize is much higher than that of humans (more than 10-fold higher; Tenaillon et al., 2001). However, most of the maize genome sequences released to date are from temperate lines. Genome sequences from tropical lines, landraces, and teosintes have seldom been reported, although many known variations exist that are specific to teosinte or tropical lines (examples are shown in Figure 1). With the advancement of sequencing technologies, it will become much easier and also cheaper to assemble many more maize genome sequences to fully reveal genetic variations, including widespread genome-structure variations (representative inversions and InDels >1 Mb are illustrated in Figure 2), and their relationship to phenotypes.
Technologies
Although some maize genome sequences and abundant variations are already available, the functions of most genes and also the genes underlying many traits and biological processes remain unclear. In the past decades, genotype limitations and the low efficiency of maize genetic transformation made maize genetic analysis much slower than that of Arabidopsis and rice. However, the greatly increased transformation efficiency that results from ectopic expression of Baby Boom (Bbm) and Wuschel 2 (Wus2) and the use of promoters from Zm-PLTP and Zm-Axig1 (which encode a phospholipid transferase protein and an Aux/IAA transcription factor, respectively) opened the gates for efficient and genotype-independent maize transformation (Lowe et al., 2016, Lowe et al., 2018).
Rooted in these improved transformation approaches, CRISPR/Cas9 technology can be used to generate both multiple mutated alleles for a single gene and mutations of multiple genes in a single maize plant by integrating multiple guide RNAs into one vector and using just a single guide RNA targeting a conserved region of homologous genes, respectively (Doll et al., 2019). This would be a very convenient method for studying gene families with functional redundancy, as demonstrated in rice (Ma et al., 2015). Furthermore, the CRISPR/Cas9 system has been successfully used to construct genome-wide mutant libraries in rice (Lu et al., 2017, Meng et al., 2017). Lines in mutant libraries derived from TILLING or Mu tagging usually have many randomly mutated loci, and mutant loci caused by Mu insertions are sometimes not stable (Brutnell, 2002, Lu et al., 2018), which could exacerbate the difficulty in phenotype investigation and validation of gene function. In contrast, the CRISPR/Cas9 system introduces stable mutations precisely at specific sites as dictated by the single guide RNA with a much cleaner genetic background. There are also increasing reports about the application of CRISPR/Cas9 technology in maize. For example, Svitashev et al. (2016) delivered a Cas9-guide RNA ribonucleoprotein assembly into maize embryos and successfully obtained edited plants and achieved DNA-free genome editing; Feng et al. (2018) reported highly efficient genome editing using the dmc1 promoter combined with the U3 promoter to drive the CRISPR/Cas9 system. Thus, the technique shows great promise in both functional research and breeding of maize (Figure 3D–3G).
Traditional QTL mapping in populations derived from multiple generations of selfing, backcrossing, or intercrossing and positional cloning are time-consuming methods. There are two approaches, referred to as QTL-seq (Takagi et al., 2013) and QTG-seq (Zhang et al., 2019), that take advantage of both bulked segregant analysis and NGS to rapidly identify QTLs (Figure 3B). Using QTG-seq, researchers even mapped a plant-height QTL to a ∼300-kb region (Zhang et al., 2019). There is also another way to quickly construct populations and, hence, map QTLs, which depends on the application of doubled haploids. A haploid, which contains only one set of chromosomes, could become a diploid inbred line in only one generation after a chromosome-doubling process. One method widely used in maize to produce haploids is to cross plants with haploid inducer lines. A number of QTLs for haploid induction rate (HIR) have been reported, with qhir1 and qhir8 explaining most of the variation (66% and 20%, respectively) in this trait (Prigge et al., 2012, Hu et al., 2016). The cloning of these two loci and the dissection of the mechanisms underlying this process were reported recently (Gilles et al., 2017, Kelliher et al., 2017, Li et al., 2017, Liu et al., 2017a, Zhong et al., 2019). A 4-bp insertion and a nucleotide change in ZmPLA1 (also named MTL, NLD) and ZmDMP, respectively, led to increased HIR (Gilles et al., 2017, Kelliher et al., 2017, Liu et al., 2017a, Zhong et al., 2019). Li et al. (2017) found severe chromosome fragmentation in pollen of an inducer line using single nucleus sequencing technology, which might be the reason for haploid induction. These findings have great promise for accelerating the breeding process, and there are already reports about the genetic manipulation of ZmPLA1 in other crops to promote haploid induction, for example in rice and wheat (Yao et al., 2018, Liu et al., 2019).
Diversified Breeding Goals
Since the process of domestication of maize from its wild ancestors began several thousand years ago, yield has always been the central goal in breeding programs. In addition, there are now many new kinds of maize developed for specific purposes, the so-called “specialty corns,” such as silage corn, waxy corn, high-amylose corn, high-oil corn, high-quality-protein corn, sweet corn, and popcorn. The planting area of specialty corn has substantially increased since the early 20th century. For example, waxy corn was discovered in the 1900s in China, and its planting area now has reached ∼800 000 hm2 in China. Meanwhile, it has been reported that nearly 40% of maize produced in the United States is used to make ethanol for fuel (Ranum et al., 2014). These two observations reflect the increasingly diverse demands of maize breeding.
Based on the genomic changes during maize domestication and variations in positionally cloned maize genes that underlie QTLs for diverse traits, many traits are affected mainly by a limited number of genes. Taking the waxy endosperm trait as an example, mutation of only one gene (Wx) leads to an amylopectin content of nearly 100% in the endosperm (Sprague et al., 1943). Thus, it is possible that we could improve specific maize traits by manipulating only a handful of genes to meet modern demands for diverse maize types.
Reuse of Wild Relatives
It is estimated that maize retains 83% of the genetic diversity from its progenitor, Zea mays ssp. parviglumis (Hufford et al., 2012). This is actually a considerably higher proportion than found in analogous comparisons between other major crops and their wild species progenitors (Fernie et al., 2006), largely due to the fact that maize is an outcrossing species. Nevertheless, the teosintes represent important resources for pinpointing the genes/variations that are important for traits but have been lost during maize artificial selection (Li et al., 2019c, Tian et al., 2019). There are several ways to discover such genes/variations. First, traditional QTL mapping and positional cloning have been successfully used to clone genes within populations derived from crosses between maize and teosinte. For example, ZmCCT10 and ZmCCT9 were mapped and cloned using this method in a maize-teosinte BC2S3 population (Hung et al., 2012, Huang et al., 2018). Association mapping in a teosinte population has also been demonstrated to be a powerful approach by which to detect significant associations between genes and traits (Weber et al., 2007, Weber et al., 2008). However, these two studies performed candidate-gene-association analyses using <150 markers. It is expected that more loci, including some novel loci, would be detected by a GWAS with a much higher density of molecular markers. Recently, a k-mer-based association mapping method called association genetics with resistance gene-enrichment sequencing (AgRenSeq) was used to successfully discover and clone disease-resistance genes from crop wild relatives (Arora et al., 2019). Multi-omics data are also very powerful for identifying genes for traits (Liu et al., 2016, de Abreu E Lima et al., 2018, McLoughlin et al., 2018) The teosinte allele of cloned genes/variations could subsequently be directly applied in maize-breeding programs through introgression into modern elite maize lines.
An alternative possibility to rewilding maize would be to de novo domesticate teosinte, as was recently proposed by Fernie and Yan (2019), via either long-term backcrossing or short-term genome editing. The latter would require a successful transformation method for teosinte. As noted above, only a few hundred genes underwent selection during maize domestication. If we want to change only a few key traits to meet the aforementioned diverse breeding goals for maize, there may be as few as <100 genes that need to be edited. The resulting costs of this process—in terms of both time and money spent—will be greatly reduced relative to the previous process of human selection, which has already lasted for thousands of years.
Funding
This work was supported by the National Natural Science Foundation of China (31961133002, 31901549, and 31525017), and the Ministry of Agriculture of China (2018ZX0801004B).
Author Contributions
J.L., A.F., and J.Y. discussed and wrote the manuscript.
Acknowledgments
No conflict of interest declared.
Published: January 13, 2020
Footnotes
Published by the Plant Communications Shanghai Editorial Office in association with Cell Press, an imprint of Elsevier Inc., on behalf of CSPB and IPPE, CAS.
Contributor Information
Jie Liu, Email: jieliu@mail.hzau.edu.cn.
Jianbing Yan, Email: yjianbing@mail.hzau.edu.cn.
References
- 1000 Genomes Project Consortium A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073. doi: 10.1038/nature09534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Abreu E Lima F., Li K., Wen W., Yan J., Nikoloski Z., Willmitzer L., Brotman Y. Unraveling lipid metabolism in maize with time-resolved multi-omics data. Plant J. 2018;93:1102–1115. doi: 10.1111/tpj.13833. [DOI] [PubMed] [Google Scholar]
- Aguilar-Martínez J.A., Poza-Carrión C., Cubas P. Arabidopsis BRANCHED1 acts as an integrator of branching signals within axillary buds. Plant Cell. 2007;19:458–472. doi: 10.1105/tpc.106.048934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alonso J.M., Stepanova A.N., Leisse T.J., Kim C.J., Chen H., Shinn P., Stevenson D.K., Zimmerman J., Barajas P., Cheuk R. Genome-wide insertional mutagenesis of Arabidopsis thaliana. Science. 2003;301:653–657. doi: 10.1126/science.1086391. [DOI] [PubMed] [Google Scholar]
- Alseekh S., Fernie A.R. Metabolomics 20 years on: what have we learned and what hurdles remain? Plant J. 2018;94:933–942. doi: 10.1111/tpj.13950. [DOI] [PubMed] [Google Scholar]
- Arora S., Steuernagel B., Gaurav K., Chandramohan S., Long Y., Matny O., Johnson R., Enk J., Periyannan S., Singh N. Resistance gene cloning from a wild crop relative by sequence capture and association genetics. Nat. Biotechnol. 2019;37:139–143. doi: 10.1038/s41587-018-0007-9. [DOI] [PubMed] [Google Scholar]
- Atwell S., Huang Y.S., Vilhjálmsson B.J., Willems G., Horton M., Li Y., Meng D., Platt A., Tarone A.M., Hu T.T. Genome-wide association study of 107 phenotypes in Arabidopsis thaliana inbred lines. Nature. 2010;465:627–631. doi: 10.1038/nature08800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balint-Kurti P.J., Zwonitzer J.C., Wisser R.J., Carson M.L., Oropeza-Rosas M.A., Holland J.B., Szalma S.J. Precise mapping of quantitative trait loci for resistance to southern leaf blight, caused by Cochliobolus heterostrophus race O, and flowering time using advanced intercross maize lines. Genetics. 2007;176:645–657. doi: 10.1534/genetics.106.067892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beadle G.W. The mystery of maize. Field Museum Nat. Hist. Bull. 1972;43:2–11. [Google Scholar]
- Beleggia R., Rau D., Laidò G., Platani C., Nigro F., Fragasso M., De Vita P., Scossa F., Fernie A.R., Nikoloski Z. Evolutionary metabolomics reveals domestication-associated changes in tetraploid wheat kernels. Mol. Biol. Evol. 2016;33:1740–1753. doi: 10.1093/molbev/msw050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benke A., Urbany C., Stich B. Genome-wide association mapping of iron homeostasis in the maize association population. BMC Genet. 2015;16:1. doi: 10.1186/s12863-014-0153-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benson J.M., Poland J.A., Benson B.M., Stromberg E.L., Nelson R.J. Resistance to gray leaf spot of maize: genetic architecture and mechanisms elucidated through nested association mapping and near-isogenic line analysis. PLoS Genet. 2015;11:e1005045. doi: 10.1371/journal.pgen.1005045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benz B.F. Archaeological evidence of teosinte domestication from Guilá Naquitz, Oaxaca. Proc. Natl. Acad. Sci. U S A. 2001;98:2104–2106. doi: 10.1073/pnas.98.4.2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bommert P., Nagasawa N.S., Jackson D. Quantitative variation in maize kernel row number is controlled by the FASCIATED EAR2 locus. Nat. Genet. 2013;45:334–337. doi: 10.1038/ng.2534. [DOI] [PubMed] [Google Scholar]
- Brandolini A., Brandolini A. Maize introduction, evolution and diffusion in Italy. Maydica. 2009;54:233–242. [Google Scholar]
- Brutnell T.P. Transposon tagging in maize. Funct. Integr. Genom. 2002;2:4–12. doi: 10.1007/s10142-001-0044-0. [DOI] [PubMed] [Google Scholar]
- Buckler E.S., Holland J.B., Bradbury P.J., Acharya C.B., Brown P.J., Browne C., Ersoz E., Flint-Garcia S., Garcia A., Glaubitz J.C. The genetic architecture of maize flowering time. Science. 2009;325:714–718. doi: 10.1126/science.1174276. [DOI] [PubMed] [Google Scholar]
- Bukowski R., Guo X., Lu Y., Zou C., He B., Rong Z., Wang B., Xu D., Yang B., Xie C. Construction of the third-generation Zea mays haplotype map. Gigascience. 2018;7:1–12. doi: 10.1093/gigascience/gix134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castelletti S., Tuberosa R., Pindo M., Salvi S. A MITE transposon insertion is associated with differential methylation at the maize flowering time QTL Vgt1. G3 (Bethesda) 2014;4:805–812. doi: 10.1534/g3.114.010686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen J., Zeng B., Zhang M., Xie S., Wang G., Hauck A., Lai J. Dynamic transcriptome landscape of maize embryo and endosperm development. Plant Physiol. 2014;166:252–264. doi: 10.1104/pp.114.240689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen G., Wang X., Hao J., Yan J., Ding J. Genome-wide association implicates candidate genes conferring resistance to maize rough dwarf disease in maize. PLoS One. 2015;10:e0142001. doi: 10.1371/journal.pone.0142001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chia J.M., Song C., Bradbury P.J., Costich D., de Leon N., Doebley J., Elshire R.J., Gaut B., Geller L., Glaubitz J.C. Maize HapMap2 identifies extant variation from a genome in flux. Nat. Genet. 2012;44:803–807. doi: 10.1038/ng.2313. [DOI] [PubMed] [Google Scholar]
- Chuck G.S., Brown P.J., Meeley R., Hake S. Maize SBP-box transcription factors unbranched2 and unbranched3 affect yield traits by regulating the rate of lateral primordia initiation. Proc. Natl. Acad. Sci. U S A. 2014;111:18775–18780. doi: 10.1073/pnas.1407401112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cockram J., White J., Zuluaga D.L., Smith D., Comadran J., Macaulay M., Luo Z., Kearsey M.J., Werner P., Harrap D. Genome-wide association mapping to candidate polymorphism resolution in the unsequenced barley genome. Proc. Natl. Acad. Sci. U S A. 2010;107:21611–21616. doi: 10.1073/pnas.1010179107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Debernardi J.M., Lin H., Chuck G., Faris J.D., Dubcovsky J. microRNA172 plays a crucial role in wheat spike morphogenesis and grain threshability. Development. 2017;144:1966–1975. doi: 10.1242/dev.146399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deng M., Li D., Luo J., Xiao Y., Liu H., Pan Q., Zhang X., Jin M., Zhao M., Yan J. The genetic architecture of amino acids dissection by association and linkage analysis in maize. Plant Biotechnol. J. 2017;15:1250–1263. doi: 10.1111/pbi.12712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding J., Ali F., Chen G., Li H., Mahuku G., Yang N., Narro L., Magorokosho C., Makumbi D., Yan J. Genome-wide association mapping reveals novel sources of resistance to northern corn leaf blight in maize. BMC Plant Biol. 2015;15:206. doi: 10.1186/s12870-015-0589-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dixon L.E., Greenwood J.R., Bencivenga S., Zhang P., Cockram J., Mellers G., Ramm K., Cavanagh C., Swain S.M., Boden S.A. TEOSINTE BRANCHED1 regulates inflorescence architecture and development in bread wheat (Triticum aestivum) Plant Cell. 2018;30:563–581. doi: 10.1105/tpc.17.00961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doebley J. The genetics of maize evolution. Annu. Rev. Genet. 2004;38:37–59. doi: 10.1146/annurev.genet.38.072902.092425. [DOI] [PubMed] [Google Scholar]
- Doebley J., Stec A. Inheritance of the morphological differences between maize and teosinte: comparison of results for two F2 populations. Genetics. 1993;134:559–570. doi: 10.1093/genetics/134.2.559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doebley J., Stec A., Gustus C. Teosinte branched1 and the origin of maize: evidence for epistasis and the evolution of dominance. Genetics. 1995;141:333–346. doi: 10.1093/genetics/141.1.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doebley J., Stec A., Hubbard L. The evolution of apical dominance in maize. Nature. 1997;386:485–488. doi: 10.1038/386485a0. [DOI] [PubMed] [Google Scholar]
- Doebley J., Gaut B., Smith B. The molecular genetics of crop domestication. Cell. 2006;127:1309–1321. doi: 10.1016/j.cell.2006.12.006. [DOI] [PubMed] [Google Scholar]
- Doll N.M., Gilles L.M., Gérentes M.F., Richard C., Just J., Fierlej Y., Borrelli V.M.G., Gendrot G., Ingram G.C., Rogowsky P.M. Single and multiple gene knockouts by CRISPR-Cas9 in maize. Plant Cell Rep. 2019;38:487–501. doi: 10.1007/s00299-019-02378-1. [DOI] [PubMed] [Google Scholar]
- Dong Z., Li W., Unger-Wallace E., Yang J., Vollbrecht E., Chuck G. Ideal crop plant architecture is mediated by tassels replace upper ears1, a BTB/POZ ankyrin repeat gene directly targeted by TEOSINTE BRANCHED1. Proc. Natl. Acad. Sci. U S A. 2017;114:E8656–E8664. doi: 10.1073/pnas.1714960114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorweiler J., Stec A., Kermicle J., Doebley J. Teosinte glume architecture 1: a genetic locus controlling a key step in maize evolution. Science. 1993;262:233–235. doi: 10.1126/science.262.5131.233. [DOI] [PubMed] [Google Scholar]
- Du Y., Liu L., Li M., Fang S., Shen X., Chu J., Zhang Z. UNBRANCHED3 regulates branching by modulating cytokinin biosynthesis and signaling in maize and rice. New Phytol. 2017;214:721–733. doi: 10.1111/nph.14391. [DOI] [PubMed] [Google Scholar]
- Ducrocq S., Madur D., Veyrieras J.B., Camus-Kulandaivelu L., Kloiber-Maitz M., Presterl T., Ouzunova M., Manicacci D., Charcosset A. Key impact of Vgt1 on flowering time adaptation in maize: evidence from association mapping and ecogeographical information. Genetics. 2008;178:2433–2437. doi: 10.1534/genetics.107.084830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ducrocq S., Giauffret C., Madur D., Combes V., Dumas F., Jouanne S., Coubriche D., Jamin P., Moreau L., Charcosset A. Fine mapping and haplotype structure analysis of a major flowering time quantitative trait locus on maize chromosome 10. Genetics. 2009;183:1555–1563. doi: 10.1534/genetics.109.106922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eshed Y., Lippman Z.B. Revolutions in agriculture chart a course for targeted breeding of old and new crops. Science. 2019 doi: 10.1126/science.aax0025. [DOI] [PubMed] [Google Scholar]
- Farfan I.D., De La Fuente G.N., Murray S.C., Isakeit T., Huang P.C., Warburton M., Williams P., Windham G.L., Kolomiets M. Genome wide association study for drought, aflatoxin resistance, and important agronomic traits of maize hybrids in the sub-tropics. PLoS One. 2015;10:e0117737. doi: 10.1371/journal.pone.0117737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng C., Su H., Bai H., Wang R., Liu Y., Guo X., Liu C., Zhang J., Yuan J., Birchler J.A. High-efficiency genome editing using a dmc1 promoter-controlled CRISPR/Cas9 system in maize. Plant Biotechnol. J. 2018;16:1848–1857. doi: 10.1111/pbi.12920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernie A.R., Yan J. De Novo domestication: an alternative route toward new crops for the future. Mol. Plant. 2019;12:615–631. doi: 10.1016/j.molp.2019.03.016. [DOI] [PubMed] [Google Scholar]
- Fernie A.R., Tadmor Y., Zamir D. Natural genetic variation for improving crop quality. Curr. Opin. Plant Biol. 2006;9:196–202. doi: 10.1016/j.pbi.2006.01.010. [DOI] [PubMed] [Google Scholar]
- Flint-Garcia S.A., Thuillet A.C., Yu J., Pressoir G., Romero S.M., Mitchell S.E., Doebley J., Kresovich S., Goodman M.M., Buckler E.S. Maize association population: a high-resolution platform for quantitative trait locus dissection. Plant J. 2005;44:1054–1064. doi: 10.1111/j.1365-313X.2005.02591.x. [DOI] [PubMed] [Google Scholar]
- Fu J., Cheng Y., Linghu J., Yang X., Kang L., Zhang Z., Zhang J., He C., Du X., Peng Z. RNA sequencing reveals the complex regulatory network in the maize kernel. Nat. Commun. 2013;4:2832. doi: 10.1038/ncomms3832. [DOI] [PubMed] [Google Scholar]
- Gao L., Gonda I., Sun H., Ma Q., Bao K., Tieman D.M., Burzynski-Chang E.A., Fish T.L., Stromberg K.A., Sacks G.L. The tomato pan-genome uncovers new genes and a rare allele regulating fruit flavor. Nat. Genet. 2019;51:1044–1051. doi: 10.1038/s41588-019-0410-2. [DOI] [PubMed] [Google Scholar]
- Garrison E., Sirén J., Novak A.M., Hickey G., Eizenga J.M., Dawson E.T., Jones W., Garg S., Markello C., Lin M.F. Variation graph toolkit improves read mapping by representing genetic variation in the reference. Nat. Biotechnol. 2018;36:875–879. doi: 10.1038/nbt.4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilles L.M., Khaled A., Laffaire J.B., Chaignon S., Gendrot G., Laplaige J., Bergès H., Beydon G., Bayle V., Barret P. Loss of pollen-specific phospholipase NOT LIKE DAD triggers gynogenesis in maize. EMBO J. 2017;36:707–717. doi: 10.15252/embj.201796603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gore M.A., Chia J.M., Elshire R.J., Sun Q., Ersoz E.S., Hurwitz B.L., Peiffer J.A., McMullen M.D., Grills G.S., Ross-Ibarra J. A first-generation haplotype map of maize. Science. 2009;326:1115–1117. doi: 10.1126/science.1177837. [DOI] [PubMed] [Google Scholar]
- Harjes C.E., Rocheford T.R., Bai L., Brutnell T.P., Kandianis C.B., Sowinski S.G., Stapleton A.E., Vallabhaneni R., Williams M., Wurtzel E.T. Natural genetic variation in lycopene epsilon cyclase tapped for maize biofortification. Science. 2008;319:330–333. doi: 10.1126/science.1150255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirsch C.N., Foerster J.M., Johnson J.M., Sekhon R.S., Muttoni G., Vaillancourt B., Peñagaricano F., Lindquist E., Pedraza M.A., Barry K. Insights into the maize pan-genome and pan-transcriptome. Plant Cell. 2014;26:121–135. doi: 10.1105/tpc.113.119982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirsch C.N., Hirsch C.D., Brohammer A.B., Bowman M.J., Soifer I., Barad O., Shem-Tov D., Baruch K., Lu F., Hernandez A.G. Draft assembly of elite inbred line PH207 provides insights into genomic and transcriptome diversity in maize. Plant Cell. 2016;28:2700–2714. doi: 10.1105/tpc.16.00353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu H., Schrag T.A., Peis R., Unterseer S., Schipprack W., Chen S., Lai J., Yan J., Prasanna B.M., Nair S.K. The genetic basis of haploid induction in maize identified with a novel genome-wide association method. Genetics. 2016;202:1267–1276. doi: 10.1534/genetics.115.184234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang X., Zhao Y., Wei X., Li C., Wang A., Zhao Q., Li W., Guo Y., Deng L., Zhu C. Genome-wide association study of flowering time and grain yield traits in a worldwide collection of rice germplasm. Nat. Genet. 2011;44:32–39. doi: 10.1038/ng.1018. [DOI] [PubMed] [Google Scholar]
- Huang C., Sun H., Xu D., Chen Q., Liang Y., Wang X., Xu G., Tian J., Wang C., Li D. ZmCCT9 enhances maize adaptation to higher latitudes. Proc. Natl. Acad. Sci. U S A. 2018;115:E334–E341. doi: 10.1073/pnas.1718058115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hufford M.B., Xu X., van Heerwaarden J., Pyhäjärvi T., Chia J.M., Cartwright R.A., Elshire R.J., Glaubitz J.C., Guill K.E., Kaeppler S.M. Comparative population genomics of maize domestication and improvement. Nat. Genet. 2012;44:808–811. doi: 10.1038/ng.2309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hung H.Y., Shannon L.M., Tian F., Bradbury P.J., Chen C., Flint-Garcia S.A., McMullen M.D., Ware D., Buckler E.S., Doebley J.F. ZmCCT and the genetic basis of day-length adaptation underlying the postdomestication spread of maize. Proc. Natl. Acad. Sci. U S A. 2012;109:E1913–E1921. doi: 10.1073/pnas.1203189109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiao Y., Zhao H., Ren L., Song W., Zeng B., Guo J., Wang B., Liu Z., Chen J., Li W. Genome-wide genetic changes during modern breeding of maize. Nat. Genet. 2012;44:812–815. doi: 10.1038/ng.2312. [DOI] [PubMed] [Google Scholar]
- Jiao Y., Peluso P., Shi J., Liang T., Stitzer M.C., Wang B., Campbell M.S., Stein J.C., Wei X., Chin C.S. Improved maize reference genome with single-molecule technologies. Nature. 2017;546:524–527. doi: 10.1038/nature22971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin M., Liu H., He C., Fu J., Xiao Y., Wang Y., Xie W., Wang G., Yan J. Maize pan-transcriptome provides novel insights into genome complexity and quantitative trait variation. Sci. Rep. 2016;6:18936. doi: 10.1038/srep18936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin M., Liu X., Jia W., Liu H., Li W., Peng Y., Du Y., Wang Y., Yin Y., Zhang X. ZmCOL3, a CCT gene represses flowering in maize by interfering with the circadian clock and activating expression of ZmCCT. J. Integr. Plant Biol. 2018;60:465–480. doi: 10.1111/jipb.12632. [DOI] [PubMed] [Google Scholar]
- Kelliher T., Starr D., Richbourg L., Chintamanani S., Delzer B., Nuccio M.L., Green J., Chen Z., McCuiston J., Wang W. MATRILINEAL, a sperm-specific phospholipase, triggers maize haploid induction. Nature. 2017;542:105–109. doi: 10.1038/nature20827. [DOI] [PubMed] [Google Scholar]
- Kim D., Paggi J.M., Park C., Bennett C., Salzberg S.L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 2019;37:907–915. doi: 10.1038/s41587-019-0201-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kump K.L., Bradbury P.J., Wisser R.J., Buckler E.S., Belcher A.R., Oropeza-Rosas M.A., Zwonitzer J.C., Kresovich S., McMullen M.D., Ware D. Genome-wide association study of quantitative resistance to southern leaf blight in the maize nested association mapping population. Nat. Genet. 2011;43:163–168. doi: 10.1038/ng.747. [DOI] [PubMed] [Google Scholar]
- Kurtz S., Phillippy A., Delcher A.L., Smoot M., Shumway M., Antonescu C., Salzberg S.L. Versatile and open software for comparing large genomes. Genome Biol. 2004;5:R12. doi: 10.1186/gb-2004-5-2-r12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai J., Li R., Xu X., Jin W., Xu M., Zhao H., Xiang Z., Song W., Ying K., Zhang M. Genome-wide patterns of genetic variation among elite maize inbred lines. Nat. Genet. 2010;42:1027–1030. doi: 10.1038/ng.684. [DOI] [PubMed] [Google Scholar]
- Lee M., Sharopova N., Beavis W.D., Grant D., Katt M., Blair D., Hallauer A. Expanding the genetic map of maize with the intermated B73 x Mo17 (IBM) population. Plant Mol. Biol. 2002;48:453–461. doi: 10.1023/a:1014893521186. [DOI] [PubMed] [Google Scholar]
- Lemmon Z.H., Bukowski R., Sun Q., Doebley J.F. The role of cis regulatory evolution in maize domestication. PLoS Genet. 2014;10:e1004745. doi: 10.1371/journal.pgen.1004745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li P., Wang Y., Qian Q., Fu Z., Wang M., Zeng D., Li B., Wang X., Li J. LAZY1 controls rice shoot gravitropism through regulating polar auxin transport. Cell Res. 2007;17:402–410. doi: 10.1038/cr.2007.38. [DOI] [PubMed] [Google Scholar]
- Li L., Li H., Li Q., Yang X., Zheng D., Warburton M., Chai Y., Zhang P., Guo Y., Yan J. An 11-bp insertion in Zea mays fatb reduces the palmitic acid content of fatty acids in maize grain. PLoS One. 2011;6:e24699. doi: 10.1371/journal.pone.0024699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q., Yang X., Xu S., Cai Y., Zhang D., Han Y., Li L., Zhang Z., Gao S., Li J. Genome-wide association studies identified three independent polymorphisms associated with α-tocopherol content in maize kernels. PLoS One. 2012;7:e36807. doi: 10.1371/journal.pone.0036807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H., Peng Z., Yang X., Wang W., Fu J., Wang J., Han Y., Chai Y., Guo T., Yang N. Genome-wide association study dissects the genetic architecture of oil biosynthesis in maize kernels. Nat. Genet. 2013;45:43–50. doi: 10.1038/ng.2484. [DOI] [PubMed] [Google Scholar]
- Li G., Wang D., Yang R., Logan K., Chen H., Zhang S., Skaggs M.I., Lloyd A., Burnett W.J., Laurie J.D. Temporal patterns of gene expression in developing maize endosperm identified through transcriptome sequencing. Proc. Natl. Acad. Sci. U S A. 2014;111:7582–7587. doi: 10.1073/pnas.1406383111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X., Meng D., Chen S., Luo H., Zhang Q., Jin W., Yan J. Single nucleus sequencing reveals spermatid chromosome fragmentation as a possible cause of maize haploid induction. Nat. Commun. 2017;8:991. doi: 10.1038/s41467-017-00969-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li C., Song W., Luo Y., Gao S., Zhang R., Shi Z., Wang X., Wang R., Wang F., Wang J. The HuangZaoSi maize genome provides insights into genomic variation and improvement history of maize. Mol. Plant. 2019;12:402–409. doi: 10.1016/j.molp.2019.02.009. [DOI] [PubMed] [Google Scholar]
- Li E., Liu H., Huang L., Zhang X., Dong X., Song W., Zhao H., Lai J. Long-range interactions between proximal and distal regulatory regions in maize. Nat. Commun. 2019;10:2633. doi: 10.1038/s41467-019-10603-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li K., Wen W., Alseekh S., Yang X., Guo H., Li W., Wang L., Pan Q., Zhan W., Liu J. Large-scale metabolite quantitative trait locus analysis provides new insights for high-quality maize improvement. Plant J. 2019;99:216–230. doi: 10.1111/tpj.14317. [DOI] [PubMed] [Google Scholar]
- Li N., Lin B., Wang H., Li X., Yang F., Ding X., Yan J., Chu Z. Natural variation in ZmFBL41 confers banded leaf and sheath blight resistance in maize. Nat. Genet. 2019;51:1540–1548. doi: 10.1038/s41588-019-0503-y. [DOI] [PubMed] [Google Scholar]
- Lin Z., Li X., Shannon L.M., Yeh C.T., Wang M.L., Bai G., Peng Z., Li J., Trick H.N., Clemente T.E. Parallel domestication of the Shattering1 genes in cereals. Nat. Genet. 2012;44:720–724. doi: 10.1038/ng.2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H., Yan J. Crop genome-wide association study: a harvest of biological relevance. Plant J. 2019;97:8–18. doi: 10.1111/tpj.14139. [DOI] [PubMed] [Google Scholar]
- Liu S., Wang X., Wang H., Xin H., Yang X., Yan J., Li J., Tran L.S., Shinozaki K., Yamaguchi-Shinozaki K. Genome-wide analysis of ZmDREB genes and their association with natural variation in drought tolerance at seedling stage of Zea mays L. PLoS Genet. 2013;9:e1003790. doi: 10.1371/journal.pgen.1003790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu L., Du Y., Shen X., Li M., Sun W., Huang J., Liu Z., Tao Y., Zheng Y., Yan J. KRN4 controls quantitative variation in maize kernel row number. PLoS Genet. 2015;11:e1005670. doi: 10.1371/journal.pgen.1005670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu L., Jeffers D., Zhang Y., Ding M., Chen W., Kang M.S., Fan X. Introgression of the crtRB1 gene into quality protein maize inbred lines using molecular markers. Mol. Breed. 2015;35:154. doi: 10.1007/s11032-015-0349-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H., Wang X., Warburton M.L., Wen W., Jin M., Deng M., Liu J., Tong H., Pan Q., Yang X. Genomic, transcriptomic, and phenomic variation reveals the complex adaptation of modern maize breeding. Mol. Plant. 2015;8:871–884. doi: 10.1016/j.molp.2015.01.016. [DOI] [PubMed] [Google Scholar]
- Liu H., Wang F., Xiao Y., Tian Z., Wen W., Zhang X., Chen X., Liu N., Li W., Liu L. MODEM: multi-omics data envelopment and mining in maize. Database (Oxford) 2016 doi: 10.1093/database/baw117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C., Li X., Meng D., Zhong Y., Chen C., Dong X., Xu X., Chen B., Li W., Li L. A 4-bp insertion at ZmPLA1 encoding a putative phospholipase A generates haploid induction in maize. Mol. Plant. 2017;10:520–522. doi: 10.1016/j.molp.2017.01.011. [DOI] [PubMed] [Google Scholar]
- Liu H., Luo X., Niu L., Xiao Y., Chen L., Liu J., Wang X., Jin M., Li W., Zhang Q. Distant eQTL and non-coding sequences play critical roles in regulating gene expression and quantitative trait variation in maize. Mol. Plant. 2017;10:414–426. doi: 10.1016/j.molp.2016.06.016. [DOI] [PubMed] [Google Scholar]
- Liu J., Huang J., Guo H., Lan L., Wang H., Xu Y., Yang X., Li W., Tong H., Xiao Y. The conserved and unique genetic architecture of kernel size and weight in maize and rice. Plant Physiol. 2017;175:774–785. doi: 10.1104/pp.17.00708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu C., Zhong Y., Qi X., Chen M., Liu Z., Chen C., Tian X., Li J., Jiao Y., Wang D. Extension of the in vivo haploid induction system from diploid maize to hexaploid wheat. Plant Biotechnol. J. 2019 doi: 10.1111/pbi.13218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowe K., Wu E., Wang N., Hoerster G., Hastings C., Cho M.J., Scelonge C., Lenderts B., Chamberlin M., Cushatt J. Morphogenic regulators Baby boom and Wuschel improve monocot transformation. Plant Cell. 2016;28:1998–2015. doi: 10.1105/tpc.16.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowe K., La Rota M., Hoerster G., Hastings C., Wang N., Chamberlin M., Wu E., Jones T., Gordon-Kamm W. Rapid genotype "independent" Zea mays L. (maize) transformation via direct somatic embryogenesis. In Vitro Cell Dev. Biol. Plant. 2018;54:240–252. doi: 10.1007/s11627-018-9905-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu F., Romay M.C., Glaubitz J.C., Bradbury P.J., Elshire R.J., Wang T., Li Y., Li Y., Semagn K., Zhang X. High-resolution genetic mapping of maize pan-genome sequence anchors. Nat. Commun. 2015;6:6914. doi: 10.1038/ncomms7914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu Y., Ye X., Guo R., Huang J., Wang W., Tang J., Tan L., Zhu J.K., Chu C., Qian Y. Genome-wide targeted mutagenesis in rice using the CRISPR/Cas9 system. Mol. Plant. 2017;10:1242–1245. doi: 10.1016/j.molp.2017.06.007. [DOI] [PubMed] [Google Scholar]
- Lu X., Liu J., Ren W., Yang Q., Chai Z., Chen R., Wang L., Zhao J., Lang Z., Wang H. Gene-indexed mutations in maize. Mol. Plant. 2018;11:496–504. doi: 10.1016/j.molp.2017.11.013. [DOI] [PubMed] [Google Scholar]
- Ma X., Zhang Q., Zhu Q., Liu W., Chen Y., Qiu R., Wang B., Yang Z., Li H., Lin Y. A robust CRISPR/Cas9 system for convenient, high-efficiency multiplex genome editing in monocot and dicot plants. Mol. Plant. 2015;8:1274–1284. doi: 10.1016/j.molp.2015.04.007. [DOI] [PubMed] [Google Scholar]
- Mao H., Wang H., Liu S., Li Z., Yang X., Yan J., Li J., Tran L.S., Qin F. A transposable element in a NAC gene is associated with drought tolerance in maize seedlings. Nat. Commun. 2015;6:8326. doi: 10.1038/ncomms9326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuoka Y., Vigouroux Y., Goodman M.M., Sanchez G.J., Buckler E., Doebley J. A single domestication for maize shown by multilocus microsatellite genotyping. Proc. Natl. Acad. Sci. U S A. 2002;99:6080–6084. doi: 10.1073/pnas.052125199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCarty D.R., Settles A.M., Suzuki M., Tan B.C., Latshaw S., Porch T., Robin K., Baier J., Avigne W., Lai J. Steady-state transposon mutagenesis in inbred maize. Plant J. 2005;44:52–61. doi: 10.1111/j.1365-313X.2005.02509.x. [DOI] [PubMed] [Google Scholar]
- McLoughlin F., Augustine R.C., Marshall R.S., Li F., Kirkpatrick L.D., Otegui M.S., Vierstra R.D. Maize multi-omics reveal roles for autophagic recycling in proteome remodelling and lipid turnover. Nat. Plants. 2018;4:1056–1070. doi: 10.1038/s41477-018-0299-2. [DOI] [PubMed] [Google Scholar]
- McMullen M.D., Kresovich S., Villeda H.S., Bradbury P., Li H., Sun Q., Flint-Garcia S., Thornsberry J., Acharya C., Bottoms C. Genetic properties of the maize nested association mapping population. Science. 2009;325:737–740. doi: 10.1126/science.1174320. [DOI] [PubMed] [Google Scholar]
- Meng X., Yu H., Zhang Y., Zhuang F., Song X., Gao S., Gao C., Li J. Construction of a genome-wide mutant library in rice using CRISPR/Cas9. Mol. Plant. 2017;10:1238–1241. doi: 10.1016/j.molp.2017.06.006. [DOI] [PubMed] [Google Scholar]
- Mertz E.T., Bates L.S., Nelson O.E. Mutant gene that changes protein composition and increases lysine content of maize endosperm. Science. 1964;145:279–280. doi: 10.1126/science.145.3629.279. [DOI] [PubMed] [Google Scholar]
- Moreno M.A., Harper L.C., Krueger R.W., Dellaporta S.L., Freeling M. liguleless1 encodes a nuclear-localized protein required for induction of ligules and auricles during maize leaf organogenesis. Genes Dev. 1997;11:616–628. doi: 10.1101/gad.11.5.616. [DOI] [PubMed] [Google Scholar]
- Multani D.S., Briggs S.P., Chamberlin M.A., Blakeslee J.J., Murphy A.S., Johal G.S. Loss of an MDR transporter in compact stalks of maize br2 and sorghum dw3 mutants. Science. 2003;302:81–84. doi: 10.1126/science.1086072. [DOI] [PubMed] [Google Scholar]
- Muthusamy V., Hossain F., Thirunavukkarasu N., Choudhary M., Saha S., Bhat J.S., Prasanna B.M., Gupta H.S. Development of β-carotene rich maize hybrids through marker-assisted introgression of β-carotene hydroxylase allele. PLoS One. 2014;9:e113583. doi: 10.1371/journal.pone.0113583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nestel P., Bouis H.E., Meenakshi J.V., Pfeiffer W. Biofortification of staple food crops. J. Nutr. 2006;136:1064–1067. doi: 10.1093/jn/136.4.1064. [DOI] [PubMed] [Google Scholar]
- Nunn N., Nancy Q. The Columbian exchange: a history of disease, food, and ideas. J. Econ. Perspect. 2010;24:163–188. [Google Scholar]
- Olukolu B.A., Negeri A., Dhawan R., Venkata B.P., Sharma P., Garg A., Gachomo E., Marla S., Chu K., Hasan A. A connected set of genes associated with programmed cell death implicated in controlling the hypersensitive response in maize. Genetics. 2013;193:609–620. doi: 10.1534/genetics.112.147595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan Q., Xu Y., Li K., Peng Y., Zhan W., Li W., Li L., Yan J. The genetic basis of plant architecture in 10 maize recombinant inbred line populations. Plant Physiol. 2017;175:858–873. doi: 10.1104/pp.17.00709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng Y., Xiong D., Zhao L., Ouyang W., Wang S., Sun J., Zhang Q., Guan P., Xie L., Li W. Chromatin interaction maps reveal genetic regulation for quantitative traits in maize. Nat. Commun. 2019;10:2632. doi: 10.1038/s41467-019-10602-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piperno D.R., Flannery K.V. The earliest archaeological maize (Zea mays L.) from highland Mexico: new accelerator mass spectrometry dates and their implications. Proc. Natl. Acad. Sci. U S A. 2001;98:2101–2103. doi: 10.1073/pnas.98.4.2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Portwood J.L., Woodhouse M.R., Cannon E.K., Gardiner J.M., Harper L.C., Schaeffer M.L., Walsh J.R., Sen T.Z., Cho K.T., Schott D.A. MaizeGDB 2018: the maize multi-genome genetics and genomics database. Nucleic Acids Res. 2019;47:D1146–D1154. doi: 10.1093/nar/gky1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prigge V., Xu X., Li L., Babu R., Chen S., Atlin G.N., Melchinger A.E. New insights into the genetics of in vivo induction of maternal haploids, the backbone of doubled haploid technology in maize. Genetics. 2012;190:781–793. doi: 10.1534/genetics.111.133066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raihan M.S., Liu J., Huang J., Guo H., Pan Q., Yan J. Multi-environment QTL analysis of grain morphology traits and fine mapping of a kernel-width QTL in Zheng58 × SK maize population. Theor. Appl. Genet. 2016;129:1465–1477. doi: 10.1007/s00122-016-2717-z. [DOI] [PubMed] [Google Scholar]
- Rakocevic G., Semenyuk V., Lee W.P., Spencer J., Browning J., Johnson I.J., Arsenijevic V., Nadj J., Ghose K., Suciu M.C. Fast and accurate genomic analyses using genome graphs. Nat. Genet. 2019;51:354–362. doi: 10.1038/s41588-018-0316-4. [DOI] [PubMed] [Google Scholar]
- Ramsay L., Comadran J., Druka A., Marshall D.F., Thomas W.T., Macaulay M., MacKenzie K., Simpson C., Fuller J., Bonar N. INTERMEDIUM-C, a modifier of lateral spikelet fertility in barley, is an ortholog of the maize domestication gene TEOSINTE BRANCHED 1. Nat. Genet. 2011;43:169–172. doi: 10.1038/ng.745. [DOI] [PubMed] [Google Scholar]
- Ranum P., Peña-Rosas J.P., Garcia-Casal M.N. Global maize production, utilization, and consumption. Ann. N Y Acad. Sci. 2014;1312:105–112. doi: 10.1111/nyas.12396. [DOI] [PubMed] [Google Scholar]
- Riedelsheimer C., Czedik-Eysenberg A., Grieder C., Lisec J., Technow F., Sulpice R., Altmann T., Stitt M., Willmitzer L., Melchinger A.E. Genomic and metabolic prediction of complex heterotic traits in hybrid maize. Nat. Genet. 2012;44:217–220. doi: 10.1038/ng.1033. [DOI] [PubMed] [Google Scholar]
- Romay M.C., Millard M.J., Glaubitz J.C., Peiffer J.A., Swarts K.L., Casstevens T.M., Elshire R.J., Acharya C.B., Mitchell S.E., Flint-Garcia S.A. Comprehensive genotyping of the USA national maize inbred seed bank. Genome Biol. 2013;14:R55. doi: 10.1186/gb-2013-14-6-r55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salvi S., Tuberosa R., Chiapparino E., Maccaferri M., Veillet S., van Beuningen L., Isaac P., Edwards K., Phillips R.L. Toward positional cloning of Vgt1, a QTL controlling the transition from the vegetative to the reproductive phase in maize. Plant Mol. Biol. 2002;48:601–613. doi: 10.1023/a:1014838024509. [DOI] [PubMed] [Google Scholar]
- Salvi S., Sponza G., Morgante M., Tomes D., Niu X., Fengler K.A., Meeley R., Ananiev E.V., Svitashev S., Bruggemann E. Conserved noncoding genomic sequences associated with a flowering-time quantitative trait locus in maize. Proc. Natl. Acad. Sci. U S A. 2007;104:11376–11381. doi: 10.1073/pnas.0704145104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sauvage C., Segura V., Bauchet G., Stevens R., Do P.T., Nikoloski Z., Fernie A.R., Causse M. Genome-wide association in tomato reveals 44 candidate loci for fruit metabolic traits. Plant Physiol. 2014;165:1120–1132. doi: 10.1104/pp.114.241521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schnable J.C., Freeling M. Genes identified by visible mutant phenotypes show increased bias toward one of two subgenomes of maize. PLoS One. 2011;6:e17855. doi: 10.1371/journal.pone.0017855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schnable S.P., Ware D., Fulton R.S., Stein J.C., Wei F., Pasternak S., Liang C., Zhang J., Fulton L., Graves T.A. The B73 maize genome: complexity, diversity, and dynamics. Science. 2009;326:1112–1115. doi: 10.1126/science.1178534. [DOI] [PubMed] [Google Scholar]
- Shang Y., Ma Y., Zhou Y., Zhang H., Duan L., Chen H., Zeng J., Zhou Q., Wang S., Gu W. Biosynthesis, regulation, and domestication of bitterness in cucumber. Science. 2014;346:1084–1088. doi: 10.1126/science.1259215. [DOI] [PubMed] [Google Scholar]
- Sigmon B., Vollbrecht E. Evidence of selection at the ramosa1 locus during maize domestication. Mol. Ecol. 2010;19:1296–1311. doi: 10.1111/j.1365-294X.2010.04562.x. [DOI] [PubMed] [Google Scholar]
- Simons K.J., Fellers J.P., Trick H.N., Zhang Z., Tai Y.S., Gill B.S., Faris J.D. Molecular characterization of the major wheat domestication gene Q. Genetics. 2006;172:547–555. doi: 10.1534/genetics.105.044727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sosso D., Luo D., Li Q.B., Sasse J., Yang J., Gendrot G., Suzuki M., Koch K.E., McCarty D.R., Chourey P.S. Seed filling in domesticated maize and rice depends on SWEET-mediated hexose transport. Nat. Genet. 2015;47:1489–1493. doi: 10.1038/ng.3422. [DOI] [PubMed] [Google Scholar]
- Sprague G.F., Brimhall B., Hixon R.M. Some effects of the waxy gene in corn on properties of the endosperm starch. J. Am. Soc. Agron. 1943;35:817–822. [Google Scholar]
- Springer N.M., Ying K., Fu Y., Ji T., Yeh C.T., Jia Y., Wu W., Richmond T., Kitzman J., Rosenbaum H. Maize inbreds exhibit high levels of copy number variation (CNV) and presence/absence variation (PAV) in genome content. PLoS Genet. 2009;5:e1000734. doi: 10.1371/journal.pgen.1000734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Springer N.M., Anderson S.N., Andorf C.M., Ahern K.R., Bai F., Barad O., Barbazuk W.B., Bass H.W., Baruch K., Ben-Zvi G. The maize W22 genome provides a foundation for functional genomics and transposon biology. Nat. Genet. 2018;50:1282–1288. doi: 10.1038/s41588-018-0158-0. [DOI] [PubMed] [Google Scholar]
- Stagnati L., Lanubile A., Samayoa L.F., Bragalanti M., Giorni P., Busconi M., Holland J.B., Marocco A. A genome wide association study reveals markers and genes associated with resistance to fusarium verticillioides infection of seedlings in a maize diversity panel. G3 (Bethesda) 2019;9:571–579. doi: 10.1534/g3.118.200916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stelpflug S.C., Sekhon R.S., Vaillancourt B., Hirsch C.N., Buell C.R., de Leon N., Kaeppler S.M. An expanded maize gene expression atlas based on RNA sequencing and its use to explore root development. Plant Genome. 2016;9 doi: 10.3835/plantgenome2015.04.0025. [DOI] [PubMed] [Google Scholar]
- Strable J., Wallace J.G., Unger-Wallace E., Briggs S., Bradbury P.J., Buckler E.S., Vollbrecht E. Maize YABBY genes drooping leaf1 and drooping leaf2 regulate plant architecture. Plant Cell. 2017;29:1622–1641. doi: 10.1105/tpc.16.00477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Studer A., Zhao Q., Ross-Ibarra J., Doebley J. Identification of a functional transposon insertion in the maize domestication gene tb1. Nat. Genet. 2011;43:1160–1163. doi: 10.1038/ng.942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Studer A.J., Wang H., Doebley J.F. Selection during maize domestication targeted a gene network controlling plant and inflorescence architecture. Genetics. 2017;207:755–765. doi: 10.1534/genetics.117.300071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun S., Zhou Y., Chen J., Shi J., Zhao H., Zhao H., Song W., Zhang M., Cui Y., Dong X. Extensive intraspecific gene order and gene structural variations between Mo17 and other maize genomes. Nat. Genet. 2018;50:1289–1295. doi: 10.1038/s41588-018-0182-0. [DOI] [PubMed] [Google Scholar]
- Svitashev S., Schwartz C., Lenderts B., Young J.K., Mark Cigan A. Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat. Commun. 2016;7:13274. doi: 10.1038/ncomms13274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takagi H., Abe A., Yoshida K., Kosugi S., Natsume S., Mitsuoka C., Uemura A., Utsushi H., Tamiru M., Takuno S. QTL-seq: rapid mapping of quantitative trait loci in rice by whole genome resequencing of DNA from two bulked populations. Plant J. 2013;74:174–183. doi: 10.1111/tpj.12105. [DOI] [PubMed] [Google Scholar]
- Takeda T., Suwa Y., Suzuki M., Kitano H., Ueguchi-Tanaka M., Ashikari M., Matsuoka M., Ueguchi C. The OsTB1 gene negatively regulates lateral branching in rice. Plant J. 2003;33:513–520. doi: 10.1046/j.1365-313x.2003.01648.x. [DOI] [PubMed] [Google Scholar]
- Tenaillon M.I., Sawkins M.C., Long A.D., Gaut R.L., Doebley J.F., Gaut B.S. Patterns of DNA sequence polymorphism along chromosome 1 of maize (Zea mays ssp. mays L.) Proc. Natl. Acad. Sci. U S A. 2001;98:9161–9166. doi: 10.1073/pnas.151244298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teng F., Zhai L., Liu R., Bai W., Wang L., Huo D., Tao Y., Zheng Y., Zhang Z. ZmGA3ox2, a candidate gene for a major QTL, qPH3.1, for plant height in maize. Plant J. 2013;73:405–416. doi: 10.1111/tpj.12038. [DOI] [PubMed] [Google Scholar]
- Thornsberry J.M., Goodman M.M., Doebley J., Kresovich S., Nielsen D., Buckler E.S. Dwarf8 polymorphisms associate with variation in flowering time. Nat. Genet. 2001;28:286–289. doi: 10.1038/90135. [DOI] [PubMed] [Google Scholar]
- Tian J., Wang C., Xia J., Wu L., Xu G., Wu W., Li D., Qin W., Han X., Chen Q. Teosinte ligule allele narrows plant architecture and enhances high-density maize yields. Science. 2019;365:658–664. doi: 10.1126/science.aax5482. [DOI] [PubMed] [Google Scholar]
- Tieman D., Zhu G., Resende M.F., Jr., Lin T., Nguyen C., Bies D., Rambla J.L., Beltran K.S., Taylor M., Zhang B. A chemical genetic roadmap to improved tomato flavor. Science. 2017;355:391–394. doi: 10.1126/science.aal1556. [DOI] [PubMed] [Google Scholar]
- Till B.J., Reynolds S.H., Weil C., Springer N., Burtner C., Young K., Bowers E., Codomo C.A., Enns L.C., Odden A.R. Discovery of induced point mutations in maize genes by TILLING. BMC Plant Biol. 2004;4:12. doi: 10.1186/1471-2229-4-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vallebueno-Estrada M., Rodríguez-Arévalo I., Rougon-Cardoso A., Martínez González J., García Cook A., Montiel R., Vielle-Calzada J.P. The earliest maize from San Marcos Tehuacán is a partial domesticate with genomic evidence of inbreeding. Proc. Natl. Acad. Sci. U S A. 2016;113:14151–14156. doi: 10.1073/pnas.1609701113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vlăduţu C., McLaughlin J., Phillips R.L. Fine mapping and characterization of linked quantitative trait loci involved in the transition of the maize apical meristem from vegetative to generative structures. Genetics. 1999;153:993–1007. doi: 10.1093/genetics/153.2.993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walley J.W., Sartor R.C., Shen Z., Schmitz R.J., Wu K.J., Urich M.A., Nery J.R., Smith L.G., Schnable J.C., Ecker J.R. Integration of omic networks in a developmental atlas of maize. Science. 2016;353:814–818. doi: 10.1126/science.aag1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H., Nussbaum-Wagler T., Li B., Zhao Q., Vigouroux Y., Faller M., Bomblies K., Lukens L., Doebley J.F. The origin of the naked grains of maize. Nature. 2005;436:714–719. doi: 10.1038/nature03863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X., Wang H., Liu S., Ferjani A., Li J., Yan J., Yang X., Qin F. Genetic variation in ZmVPP1 contributes to drought tolerance in maize seedlings. Nat. Genet. 2016;48:1233–1241. doi: 10.1038/ng.3636. [DOI] [PubMed] [Google Scholar]
- Wang C., Yang Q., Wang W., Li Y., Guo Y., Zhang D., Ma X., Song W., Zhao J., Xu M. A transposon-directed epigenetic change in ZmCCT underlies quantitative resistance to Gibberella stalk rot in maize. New Phytol. 2017;215:1503–1515. doi: 10.1111/nph.14688. [DOI] [PubMed] [Google Scholar]
- Wang H., Xu S., Fan Y., Liu N., Zhan W., Liu H., Xiao Y., Li K., Pan Q., Li W. Beyond pathways: genetic dissection of tocopherol content in maize kernels by combining linkage and association analyses. Plant Biotechnol. J. 2018;16:1464–1475. doi: 10.1111/pbi.12889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang W., Mauleon R., Hu Z., Chebotarov D., Tai S., Wu Z., Li M., Zheng T., Fuentes R.R., Zhang F. Genomic variation in 3,010 diverse accessions of Asian cultivated rice. Nature. 2018;557:43–49. doi: 10.1038/s41586-018-0063-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J., Lin Z., Zhang X., Liu H., Zhou L., Zhong S., Li Y., Zhu C., Lin Z. krn1, a major quantitative trait locus for kernel row number in maize. New Phytol. 2019;223:1634–1646. doi: 10.1111/nph.15890. [DOI] [PubMed] [Google Scholar]
- Weber A., Clark R.M., Vaughn L., Sánchez-Gonzalez Jde J., Yu J., Yandell B.S., Bradbury P., Doebley J. Major regulatory genes in maize contribute to standing variation in teosinte (Zea mays ssp. parviglumis) Genetics. 2007;177:2349–2359. doi: 10.1534/genetics.107.080424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weber A.L., Briggs W.H., Rucker J., Baltazar B.M., de Jesús Sánchez-Gonzalez J., Feng P., Buckler E.S., Doebley J. The genetic architecture of complex traits in teosinte (Zea mays ssp. parviglumis): new evidence from association mapping. Genetics. 2008;180:1221–1232. doi: 10.1534/genetics.108.090134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei L., Cao X. The effect of transposable elements on phenotypic variation: insights from plants to humans. Sci. China Life Sci. 2016;59:24–37. doi: 10.1007/s11427-015-4993-2. [DOI] [PubMed] [Google Scholar]
- Wen W., Li D., Li X., Gao Y., Li W., Li H., Liu J., Liu H., Chen W., Luo J. Metabolome-based genome-wide association study of maize kernel leads to novel biochemical insights. Nat. Commun. 2014;5:3438. doi: 10.1038/ncomms4438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen W., Li K., Alseekh S., Omranian N., Zhao L., Zhou Y., Xiao Y., Jin M., Yang N., Liu H. Genetic determinants of the network of primary metabolism and their relationships to plant performance in a maize recombinant inbred line population. Plant Cell. 2015;27:1839–1856. doi: 10.1105/tpc.15.00208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen W., Brotman Y., Willmitzer L., Yan J., Fernie A.R. Broadening our portfolio in the genetic improvement of maize chemical composition. Trends Genet. 2016;32:459–469. doi: 10.1016/j.tig.2016.05.003. [DOI] [PubMed] [Google Scholar]
- Wen W., Jin M., Li K., Liu H., Xiao Y., Zhao M., Alseekh S., Li W., de Abreu E Lima F., Brotman Y. An integrated multi-layered analysis of the metabolic networks of different tissues uncovers key genetic components of primary metabolism in maize. Plant J. 2018;93:1116–1128. doi: 10.1111/tpj.13835. [DOI] [PubMed] [Google Scholar]
- Whipple C.J., Kebrom T.H., Weber A.L., Yang F., Hall D., Meeley R., Schmidt R., Doebley J., Brutnell T.P., Jackson D.P. Grassy tillers1 promotes apical dominance in maize and responds to shade signals in the grasses. Proc. Natl. Acad. Sci. U S A. 2011;108:E506–E512. doi: 10.1073/pnas.1102819108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wills D.M., Whipple C.J., Takuno S., Kursel L.E., Shannon L.M., Ross-Ibarra J., Doebley J.F. From many, one: genetic control of prolificacy during maize domestication. PLoS Genet. 2013;9:e1003604. doi: 10.1371/journal.pgen.1003604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright S.I., Bi I.V., Schroeder S.G., Yamasaki M., Doebley J.F., McMullen M.D., Gaut B.S. The effects of artificial selection on the maize genome. Science. 2005;308:1310–1314. doi: 10.1126/science.1107891. [DOI] [PubMed] [Google Scholar]
- Wu C., Li X., Yuan W., Chen G., Kilian A., Li J., Xu C., Li X., Zhou D.X., Wang S. Development of enhancer trap lines for functional analysis of the rice genome. Plant J. 2003;35:418–427. doi: 10.1046/j.1365-313x.2003.01808.x. [DOI] [PubMed] [Google Scholar]
- Xiang Y., Sun X., Gao S., Qin F., Dai M. Deletion of an endoplasmic reticulum stress response element in a ZmPP2C-A gene facilitates drought tolerance of maize seedlings. Mol. Plant. 2017;10:456–469. doi: 10.1016/j.molp.2016.10.003. [DOI] [PubMed] [Google Scholar]
- Xiao Y., Tong H., Yang X., Xu S., Pan Q., Qiao F., Raihan M.S., Luo Y., Liu H., Zhang X. Genome-wide dissection of the maize ear genetic architecture using multiple populations. New Phytol. 2016;210:1095–1106. doi: 10.1111/nph.13814. [DOI] [PubMed] [Google Scholar]
- Xiao Y., Liu H., Wu L., Warburton M., Yan J. Genome-wide association studies in maize: praise and stargaze. Mol. Plant. 2017;10:359–374. doi: 10.1016/j.molp.2016.12.008. [DOI] [PubMed] [Google Scholar]
- Xie Q., Li N., Yang Y., Lv Y., Yao H., Wei R., Sparkes D.L., Ma Z. Pleiotropic effects of the wheat domestication gene Q on yield and grain morphology. Planta. 2018;247:1089–1098. doi: 10.1007/s00425-018-2847-4. [DOI] [PubMed] [Google Scholar]
- Xing A., Gao Y., Ye L., Zhang W., Cai L., Ching A., Llaca V., Johnson B., Liu L., Yang X. A rare SNP mutation in Brachytic2 moderately reduces plant height and increases yield potential in maize. J. Exp. Bot. 2015;66:3791–3802. doi: 10.1093/jxb/erv182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu C., Ren Y., Jian Y., Guo Z., Zhang Y., Xie C., Fu J., Wang H., Wang G., Xu Y. Development of a maize 55 K SNP array with improved genome coverage for molecular breeding. Mol. Breed. 2017;37:20. doi: 10.1007/s11032-017-0622-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu G., Cao J., Wang X., Chen Q., Jin W., Li Z., Tian F. Evolutionary metabolomics identifies substantial metabolic divergence between maize and its wild ancestor, teosinte. Plant Cell. 2019;31:1990–2009. doi: 10.1105/tpc.19.00111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue W., Xing Y., Weng X., Zhao Y., Tang W., Wang L., Zhou H., Yu S., Xu C., Li X. Natural variation in Ghd7 is an important regulator of heading date and yield potential in rice. Nat. Genet. 2008;40:761–767. doi: 10.1038/ng.143. [DOI] [PubMed] [Google Scholar]
- Yan J., Kandianis C.B., Harjes C.E., Bai L., Kim E.H., Yang X., Skinner D.J., Fu Z., Mitchell S., Li Q. Rare genetic variation at Zea mays crtRB1 increases beta-carotene in maize grain. Nat. Genet. 2010;42:322–327. doi: 10.1038/ng.551. [DOI] [PubMed] [Google Scholar]
- Yan J., Yang X., Shah T., Sánchez-Villeda H., Li J., Warburton M., Zhou Y., Crouch J., Xu Y. High-throughput SNP genotyping with the GoldenGate assay in maize. Mol. Breed. 2010;25:441–451. [Google Scholar]
- Yan J., Warburton M., Crouch J. Association mapping for enhancing maize (Zea mays L.) genetic improvement. Crop Sci. 2011;51:433–449. [Google Scholar]
- Yang Q., Yin G., Guo Y., Zhang D., Chen S., Xu M. A major QTL for resistance to Gibberella stalk rot in maize. Theor. Appl. Genet. 2010;121:673–687. doi: 10.1007/s00122-010-1339-0. [DOI] [PubMed] [Google Scholar]
- Yang X., Guo Y., Yan J., Zhang J., Song T., Rocheford T., Li J.S. Major and minor QTL and epistasis contribute to fatty acid compositions and oil concentration in high-oil maize. Theor. Appl. Genet. 2010;120:665–678. doi: 10.1007/s00122-009-1184-1. [DOI] [PubMed] [Google Scholar]
- Yang X., Gao S., Xu S., Zhang Z., Prasanna B., Li L., Li J., Yan J. Characterization of a global germplasm collection and its potential utilization for analysis of complex quantitative traits in maize. Mol. Breed. 2011;28:511–526. [Google Scholar]
- Yang Q., Li Z., Li W., Ku L., Wang C., Ye J., Li K., Yang N., Li Y., Zhong T. CACTA-like transposable element in ZmCCT attenuated photoperiod sensitivity and accelerated the postdomestication spread of maize. Proc. Natl. Acad. Sci. U S A. 2013;110:16969–16974. doi: 10.1073/pnas.1310949110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang N., Lu Y., Yang X., Huang J., Zhou Y., Ali F., Wen W., Liu J., Li J., Yan J. Genome wide association studies using a new nonparametric model reveal the genetic architecture of 17 agronomic traits in an enlarged maize association panel. PLoS Genet. 2014;10:e1004573. doi: 10.1371/journal.pgen.1004573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang N., Xu X.W., Wang R.R., Peng W.L., Cai L., Song J.M., Li W., Luo X., Niu L., Wang Y. Contributions of Zea mays subspecies mexicana haplotypes to modern maize. Nat. Commun. 2017;8:1874. doi: 10.1038/s41467-017-02063-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Q., Balint-Kurti P., Xu M. Quantitative disease resistance: dissection and adoption in maize. Mol. Plant. 2017;10:402–413. doi: 10.1016/j.molp.2017.02.004. [DOI] [PubMed] [Google Scholar]
- Yang N., Liu J., Gao Q., Gui S., Chen L., Yang L., Huang J., Deng T., Luo J., He L. Genome assembly of a tropical maize inbred line provides insights into structural variation and crop improvement. Nat. Genet. 2019;51:1052–1059. doi: 10.1038/s41588-019-0427-6. [DOI] [PubMed] [Google Scholar]
- Yang N., Wu S., Yan J. Structural variation in complex genome: detection, integration and function. Sci. China Life Sci. 2019;62:1098–1100. doi: 10.1007/s11427-019-9664-4. [DOI] [PubMed] [Google Scholar]
- Yao L., Zhang Y., Liu C., Liu Y., Wang Y., Liang D., Liu J., Sahoo G., Kelliher T. OsMATL mutation induces haploid seed formation in indica rice. Nat. Plants. 2018;4:530–533. doi: 10.1038/s41477-018-0193-y. [DOI] [PubMed] [Google Scholar]
- Ye J., Zhong T., Zhang D., Ma C., Wang L., Yao L., Zhang Q., Zhu M., Xu M. The auxin-regulated protein ZmAuxRP1 coordinates the balance between root growth and stalk rot disease resistance in maize. Mol. Plant. 2019;12:360–373. doi: 10.1016/j.molp.2018.10.005. [DOI] [PubMed] [Google Scholar]
- Yi F., Gu W., Chen J., Song N., Gao X., Zhang X., Zhou Y., Ma X., Song W., Zhao H. High-temporal-resolution transcriptome landscape of early maize seed development. Plant Cell. 2019;31:974–992. doi: 10.1105/tpc.18.00961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu J., Holland J.B., McMullen M.D., Buckler E.S. Genetic design and statistical power of nested association mapping in maize. Genetics. 2008;178:539–551. doi: 10.1534/genetics.107.074245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan J., Thakare D., Ma C., Lloyd A., Nixon N.M., Arakaki A.M., Burnett W.J., Logan K.O., Wang D., Wang X. RNA sequencing of laser-capture microdissected compartments of the maize kernel identifies regulatory modules associated with endosperm cell differentiation. Plant Cell. 2015;27:513–531. doi: 10.1105/tpc.114.135657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan W., Liu J., Pan Q., Wang H., Yan S., Li K., Deng M., Li W., Liu N., Kong Q. An allele of ZmPORB2 encoding a protochlorophyllide oxidoreductase promotes tocopherol accumulation in both leaves and kernels of maize. Plant J. 2019;100:114–127. doi: 10.1111/tpj.14432. [DOI] [PubMed] [Google Scholar]
- Zhang J., Li C., Wu C., Xiong L., Chen G., Zhang Q., Wang S. RMD: a rice mutant database for functional analysis of the rice genome. Nucleic Acids Res. 2006;34:D745–D748. doi: 10.1093/nar/gkj016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D., Liu Y., Guo Y., Yang Q., Ye J., Chen S., Xu M. Fine-mapping of qRfg2, a QTL for resistance to Gibberella stalk rot in maize. Theor. Appl. Genet. 2012;124:585–596. doi: 10.1007/s00122-011-1731-4. [DOI] [PubMed] [Google Scholar]
- Zhang J., Ku L.X., Han Z.P., Guo S.L., Liu H.J., Zhang Z.Z., Cao L.R., Cui X.J., Chen Y.H. The ZmCLA4 gene in the qLA4-1 QTL controls leaf angle in maize (Zea mays L.) J. Exp. Bot. 2014;65:5063–5076. doi: 10.1093/jxb/eru271. [DOI] [PubMed] [Google Scholar]
- Zhang H., Wang X., Pan Q., Li P., Liu Y., Lu X., Zhong W., Li M., Han L., Li J. QTG-Seq accelerates QTL fine mapping through QTL partitioning and whole-genome sequencing of bulked segregant samples. Mol. Plant. 2019;12:426–437. doi: 10.1016/j.molp.2018.12.018. [DOI] [PubMed] [Google Scholar]
- Zhao J., Sauvage C., Zhao J., Bitton F., Bauchet G., Liu D., Huang S., Tieman D.M., Klee H.J., Causse M. Meta-analysis of genome-wide association studies provides insights into genetic control of tomato flavor. Nat. Commun. 2019;10:1534. doi: 10.1038/s41467-019-09462-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng P., Allen W.B., Roesler K., Williams M.E., Zhang S., Li J., Glassman K., Ranch J., Nubel D., Solawetz W. A phenylalanine in DGAT is a key determinant of oil content and composition in maize. Nat. Genet. 2008;40:367–372. doi: 10.1038/ng.85. [DOI] [PubMed] [Google Scholar]
- Zhong Y., Liu C., Qi X., Jiao Y., Wang D., Wang Y., Liu Z., Chen C., Chen B., Tian X. Mutation of ZmDMP enhances haploid induction in maize. Nat. Plants. 2019;5:575–580. doi: 10.1038/s41477-019-0443-7. [DOI] [PubMed] [Google Scholar]
- Zhou P., Hirsch C.N., Briggs S.P., Springer N.M. Dynamic patterns of gene expression additivity and regulatory variation throughout maize development. Mol. Plant. 2019;12:410–425. doi: 10.1016/j.molp.2018.12.015. [DOI] [PubMed] [Google Scholar]
- Zhou S., Kremling K., Bandillo N., Richter A., Zhang Y.K., Ahern K.R., Artyukhin A.B., Hui J.X., Younkin G.C., Schroeder F.C. Metabolome-scale genome-wide association studies reveal chemical diversity and genetic control of maize specialized metabolites. Plant Cell. 2019;31:937–955. doi: 10.1105/tpc.18.00772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu G., Gou J., Klee H., Huang S. Next-gen approaches to flavor-related metabolism. Annu. Rev. Plant Biol. 2019;70:187–212. doi: 10.1146/annurev-arplant-050718-100353. [DOI] [PubMed] [Google Scholar]
- Zhu G., Wang S., Huang Z. Rewiring of the fruit metabolome in tomato breeding. Cell. 2018;172:249–261. doi: 10.1016/j.cell.2017.12.019. [DOI] [PubMed] [Google Scholar]
- Zila C.T., Samayoa L.F., Santiago R., Butrón A., Holland J.B. A genome-wide association study reveals genes associated with fusarium ear rot resistance in a maize core diversity panel. G3 (Bethesda) 2013;3:2095–2104. doi: 10.1534/g3.113.007328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zunjare R.U., Hossain F., Muthusamy V., Baveja A., Chauhan H.S., Bhat J.S., Thirunavukkarasu N., Saha S., Gupta H.S. Development of biofortified maize hybrids through marker-assisted stacking of β-carotene hydroxylase, Lycopene-ε-cyclase and Opaque2 genes. Front Plant Sci. 2018;9:178. doi: 10.3389/fpls.2018.00178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo W., Chao Q., Zhang N., Ye J., Tan G., Li B., Xing Y., Zhang B., Liu H., Fengler K.A. A maize wall-associated kinase confers quantitative resistance to head smut. Nat. Genet. 2015;47:151–157. doi: 10.1038/ng.3170. [DOI] [PubMed] [Google Scholar]



