Next Article in Journal
Sterol Characteristics in Silkworm Brain and Various Tissues Characterized by Precise Sterol Profiling Using LC-MS/MS
Previous Article in Journal
Perioperative Cardioprotection by Remote Ischemic Conditioning
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptome Sequencing and Metabolome Analysis Reveal Genes Involved in Pigmentation of Green-Colored Cotton Fibers

1
The Key Laboratory of Oasis Eco-agriculture, Agriculture College, Shihezi University, Bei 5 Road, Shihezi 832003, China
2
CSIRO Agriculture and Food, GPO Box 1700, Canberra 2601, Australia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(19), 4838; https://doi.org/10.3390/ijms20194838
Submission received: 7 August 2019 / Revised: 19 September 2019 / Accepted: 27 September 2019 / Published: 29 September 2019
(This article belongs to the Section Biochemistry)

Abstract

:
Green-colored fiber (GCF) is the unique raw material for naturally colored cotton textile but we know little about the pigmentation process in GCF. Here we compared transcriptomes and metabolomes of 12, 18 and 24 days post-anthesis (DPA) fibers from a green fiber cotton accession and its white-colored fiber (WCF) near-isogenic line. We found a total of 2047 non-redundant metabolites in GCF and WCF that were enriched in 80 pathways, including those of biosynthesis of phenylpropanoid, cutin, suberin, and wax. Most metabolites, particularly sinapaldehyde, of the phenylpropanoid pathway had a higher level in GCF than in WCF, consistent with the significant up-regulation of the genes responsible for biosynthesis of those metabolites. Weighted gene co-expression network analysis (WGCNA) of genes differentially expressed between GCF and WCF was used to uncover gene-modules co-expressed or associated with the accumulation of green pigments. Of the 16 gene-modules co-expressed with fiber color or time points, the blue module associated with G24 (i.e., GCF at 24 DPA) was of particular importance because a large proportion of its genes were significantly up-regulated at 24 DPA when fiber color was visually distinguishable between GCF and WCF. A total of 56 hub genes, including the two homoeologous Gh4CL4 that could act in green pigment biosynthesis, were identified among the genes of the blue module that are mainly involved in lipid metabolism, phenylpropanoid biosynthesis, RNA transcription, signaling, and transport. Our results provide novel insights into the mechanisms underlying pigmentation of green fibers and clues for developing cottons with stable green colored fibers.

1. Introduction

Naturally colored cottons (NCCs) are varieties of cotton that produce fibers with non-white colors [1]. NCC accumulates its pigment during fiber development, requires no dyeing steps during fabric processing and manufacturing, there is thus no disposal of toxic dye waste [2,3,4], reducing manufacturing costs and being environmentally friendly [5,6]. However, the fiber color of NCCs are generally monotonous and unstable [7], which restricts large-scale production and utilization of NCCs.
Brown and green are the two most common colors observed in NCCs. Many studies have been carried out to understand the composition of pigments in colored fibers and the pathways involved in biosynthesis of pigments. The main pigment components in the brown-colored fiber (BCF) are proanthocyanidins or their derivatives generated from the flavonoid pathway. Several genes of the pathway that are related to pigment synthesis have been identified and characterized, such as genes encoding chalcone synthases (CHS), chalcone isomerase (CHI), flavanone 3-hydroxylases (F3H), flavonoid 3′, 5′-hydroxylases (F3′5′H), dihydroflavonol 4-reductases (DFR), leucoanthocyanidin reductases (LAR), anthocyanidin synthase (ANS), and anthocyanidin reductases (ANR) [4,8,9]. Genetically, at least 6 loci (Lc1 to Lc6) have been reported to be associated with BCF [7]. A recent study identified the gene underlying the major BCF locus, Lc1, to be a R2R3-type MYB transcription factor encoding TRANSPARENT TESTA 2 (TT2) that regulates the expression of genes of the flavonoid pathway [10]. The pigment components of the green-colored fibers (GCF) are more complex than that of BCF. Sections of GCF have concentric osmiophilic layers which not found in BCF, osmiophilic layers that are each separated by cellulosic material. Chemical analysis of the isolated cell walls from green fibers confirmed the presence of suberin layers with 65% of their total monomers being 22-hydroxydocosanoic acid [11,12]. GCF are suberized and contain a large proportion of wax. The unidentified components of the wax could be separated into a colorless fluorescent fraction, and a yellow-pigmented fraction. The colorless fraction contains caffeic acid esterified to fatty acids (mainly ω-hydroxy fatty acids) and glycerol in a molar ratio of 4:5:5 [13,14,15]. Two caffeic-acid derivatives have been isolated from the yellow components of the GCF extract [1,16]. In plants, caffeic acid derivatives are produced by the phenylpropanoid biosynthesis pathway. When the enzymatic activity of phenylalanine ammonia lyase (PAL) is inhibited, in vitro cultured fibers from ovules of GCF variety remained white, and the colorless caffeic-acid derivatives and yellow components could no longer be detected [14]. Additionally, inhibition of fatty acid elongation activity results in discontinuous suberin layers and reduced lamellae [17]. Zhao and Wang [18] reported that the contents of flavonoids, including flavone and flavanols, were higher in GCF than in WCF.
In this study, we compared transcriptomes and metabolomes of GCF and WCF, explored the gene regulatory networks of pigmentation in GCF, and found differentially accumulated metabolites and differentially expressed genes related to biosynthesis of phenylpropanoids and flavonoids. Additionally, we used weighted gene co-expression network analysis (WGCNA) to identify a gene module highly related to the green fiber color at 24 DPA. The module contained two candidate hub genes (Gh4CL4_At and Gh4CL4_Dt) encoding 4-coumarate:Coenzyme A ligase (4CL). Our findings provided new insights into the molecular mechanisms responsible for pigment biosynthesis in GCF.

2. Results

2.1. Overview of the Metabolomic Profiling

To compare GCF and WCF metabolites, datasets obtained from quadrapole time of flight–mass spectrometry (QTOF–MS) by electrospray ionization (ESI+ and ESI) were subjected to principal component analysis (PCA). The results showed that metabolites from different time points (12, 18 and 24 DPA) of GCF and WCF were clearly separated in the score plots, where the first principal component (PC1) was plotted against the second principal component (PC2). For the ESI+ mode, PC1 and PC2 represented 39.83% and 16.22% of the total variations, respectively (Figure 1A), and for the ESI mode, PC1 and PC2 accounted for 15.56% and 41.39% variations, respectively (Figure 1B). Plots from partial least squares discriminant analyses (PLS-DA) was further used to model the metabolite differences between GCF and WCF. At each time point, GCF and WCF were well separated in both ESI modes (Figure S1). These results suggested significant biochemical differences between GCF and WCF at 12–24 DPA.
To determine the effect of metabolites on fiber color, we did pairwise (GCF vs. WCF) comparison of the types of metabolites detected at each time point, and identified 852, 1011, and 1073 different metabolites in G12 vs W12, G18 vs W18 and G24 vs W24, respectively, representing a total of 2047 non-redundant metabolites. Of these metabolites, 95 were found to be commonly different at all three time points (Figure 1C). The number of different metabolites between GCF and WCF increased from 12 DPA to 24 DPA. At all three time points, more down-regulated metabolites and fewer up-regulated metabolites were observed in GCF than in WCF (Figure 1D).

2.2. Untargeted Metabolomic Analysis of Phenylpropanoid Metabolite Content

Significant metabolic difference was detected between GCF and WCF across all three time points. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of the 2047 differential ions peaks showed that they were enriched for 80 pathways, including phenylpropanoid pathway, biosynthesis of plant secondary metabolites, and cutin, suberin, and wax biosynthesis pathway (Figure 2). In the phenylpropanoid pathway, 30 different metabolites (Table S3) were detected between GCF and WCF, and 12 were up-regulated in G24 compared to W24. Sinapaldehyde was the most significantly up-regulated metabolite in G24, followed by caffeic acid, quercitrin, 5-hydroxyconiferaldehyde and ferulic acid. The amount of sinapaldehyde was >100 times higher in G24 than in W24 (Table 1).
Fourteen different metabolites involved in the biosynthesis of cutin, suberin, and wax were detected, and 10 were up-regulated in G24 compared to W24 (Table 2). These included three 22-carbon chain (C22) hydroxyalkanoic acids (22-hydroxydocosanoate, 22-oxo-docosanoate, docosanedioate), four 18-carbon chain (C18) hydroxyalkanoic acids (9,10-dihydroxystearate, 9,10-epoxy-18-hydroxystearate, cis-9,10-epoxystearic acid, 18-hydroxyoleate), and three 16-carbon chain (C16) hydroxyalkanoic acids (hexadecanedioate, 16-hydroxypalmitic acid, 16-oxo-palmitate). Docosanedioate was the most significantly up-regulated metabolite in G24, with an 11.85-fold increase compared to that in W24 (Table 2).

2.3. Global Transcriptome Changes During the Process of Fiber Pigmentation

To determine the global transcriptomic profile associated with fiber pigmentation, RNA-seq was performed using 12, 18, and 24 DPA RNA from green fiber cultivar C7 and its WCF near-isogenic line (C7-NIL). The Q30 percentage (sequences with sequencing error rate lower than 0.1%) was over 90%, and the average GC content of the RNA-seq reads was 44.7% (Table S4). After filtering, approximately 50 million clean reads were retained for each sample. We found that 92%–94% of the clean reads could be mapped to the G. hirsutum reference genome [19], of which 83%–86% were uniquely mapped. Approximately 90% of the clean reads mapped to the 70,478 annotated genes. The mapped reads were used to calculate gene expression levels based on the fragments per kilobase of transcript per million mapped reads (FPKM) that were further used in analysis of differentially expressed genes (DEGs) [20].
Using the criteria of |log2 (fold change)| > 2 and Padj < 0.05, we identified a total of 8467 DEGs between GCF and WCF at the three time points, including 1443 at 12 DPA, 4278 at 18 DPA, and 5473 at 24 DPA. Between GCF and WCF, there were more up-regulated genes than down-regulated ones at 12 and 18 DPA, whereas there were slightly more down-regulated genes than up-regulated ones at 24 DPA (Figure 3A). A total of 543 genes (332 up-regulated and 198 down-regulated) were differentially expressed at all the three time points (Figure 3B). Regarding the number of DEGs between different time points of each genotype (i.e., GCF or WCF), the highest was observed in G18 vs. G12 while the least was observed in W18 vs W12 (Figure 3A,C). Overall, a total of 13,438 non-redundant DEGs were found in the 7 comparisons (Figure 3D, Table S5).

2.4. Co-Expression Network Analysis Identified Pigmentation-Related Differentially Expressed Genes (DEGs)

To investigate the gene regulatory network during fiber development, and to identify specific gene modules that are associated with pigment formation, 13438 non-redundant DEGs were subjected to WGCNA. Modules were defined as clusters of highly interconnected genes, in which genes within the same cluster have high correlation coefficients. WGCNA analysis identified 16 distinct modules (labeled with different colors) shown in Figure 4A, in which major tree branches define the modules. The correlation coefficients between each module eigengene of the 16 distinct modules with each distinct sample (trait) are shown in Figure 4B (Table S6). Notably, 5 module–trait relationships (blue-G24, purple-W18, green-W24, red-G12 and magenta-G18) were highly significant (r > 0.8, p < 10−3; Figure 4B).
Pigment accumulation in naturally colored fibers starts at about 20 DPA [21]. Identification of a G24-specific module (the blue module) was thus particularly interesting. The majority genes of the blue module were significantly up-regulated in G24 but weakly expressed in WCF (Figure 5A). Of these up-regulated genes, those that were also up-regulated in G18 are of particularly interest, as they might be associated with pigment formation and accumulation. We also constructed gene networks of the blue-module genes using WGCNA and identified 56 hub genes based on the criteria of KME (eigengene connectivity) ≥0.99 and edge weight value ≥0.5. These genes were found to be involved in lipid metabolism, phenylpropanoid biosynthesis, RNA transcription, signaling, and transport (Figure 5B, Table 3). Previous studies have reported that green cotton fiber pigments are mainly hydroxycinnamic acid and its derivatives, which is synthesised by the phenylpropanoid pathway in plants. In this pathway, 4CL is the key gene which encodes 4-coumaric acid:coenzyme A ligase that catalyze hydroxycinnamic acids into corresponding CoA thiolesters and its derivatives [22]. Two of the candidate hub genes were homologs of Gh4CL4 (Gh_A10G0456 and Gh_D10G0473).
The 2705 DEGs of the blue module could be classified into 17 main groups/bins based on their annotated functions, including regulation of protein activity (10.22%), signaling (7.09%), transcriptional regulation (5.91%), transport (5.87%), cell wall (4.38%), and stress (4.31%) (Table S7). KEGG analysis indicated that these DEGs were highly enriched in the following pathways: pentose and glucuronate interconversions (p-value = 5.46 × 10−9, 37 genes), fatty acid metabolism (p_value = 3.45 × 10−7, 31 genes), cutin, suberin, and wax biosynthesis (p_value = 6.32 × 10−7, 17 genes), fatty acid elongation (p-value = 3.75 × 10−6, 18 genes), phenylpropanoid biosynthesis (p_value = 1.07 × 10−5, 44 genes), pyruvate metabolism (p_value = 1.72 × 10−5, 29 genes), biosynthesis of unsaturated fatty acids (p_value = 1.53 × 10−4, 16 genes), fatty acid biosynthesis (p_value = 3.96 × 10−4, 16 genes), flavonoid biosynthesis (p_value = 5.72 × 10−4, 11 genes) and biosynthesis of secondary metabolites (p_value = 8.91 × 10−4, 167 genes) (Figure 5C). Both metabolomic and transcriptomic analysis revealed significant changes in the pathways related to biosynthesis of phenylpropanoids, cutin, suberin, and wax, we therefore further analyzed selected genes of these two pathways.

2.5. Phenylpropanoid Pathway

One of the focuses of the present study was to understand the mechanisms underlying green pigmentation in cotton fibers based on comparative transcriptome analysis. To this end, we first used real-time quantitative polymerase chain reaction (RT-qPCR) to validate the expression changes obtained based on the RNA-seq data. Twelve genes from the phenylpropanoid pathway were selected for validation, and a strong correlation between the RNA-seq and qRT-PCR data was observed, indicating the reliability of the RNA-seq data (Figure S2). We then checked the relationship between the expression change of genes and the accumulation of their corresponding metabolites, and found a co-occurred relationship in G24 for the major genes and their metabolites of the phenylpropanoid pathway. To understand the regulatory network of phenylalanine and flavonoid metabolic pathways between GCF and WCF, we carried out Pearson correlation tests between relative quantitative changes of metabolites and transcripts according to Gianoulis et al. [23]. The result showed that 26 transcripts had correlation (R2 > 0.4) with 12 metabolites (Supplementary Table S8).
The phenylpropanoid pathway is one of the most important pathways associated with plant secondary metabolism, which includes two important branches, the phenylalanine and flavonoid metabolic pathways [24]. As shown in Figure 6, many genes and metabolites of the phenylalanine and flavonoid biosynthesis pathways were up-regulated in G24, including PAL (5 DEGs), cinnamate-3-hydrolase (C3H, 1 DEG), 4CL (4 DEGs), ferulate-5-hydroxylase (F5H, 2 DEGs), hydroxycinnamoyl-CoA shikimate/quinate hydroxycinnamoyl transferase (HCT, 3 DEGs), caffeoyl-CoA O-methyltransferase (CCoAOMT, 2 DEGs), CHS (3 DEGs), F3H (1 DEG), F3′H (1 DEG), F3′5′H (1 DEG), flavanol synthase (FLS, 1 DEG), and LAR (1 DEG).
Most of these genes were significantly up-regulated in both G18 and G24 (Table 4). The expression levels of four 4CL genes, Gh_A05G3997, Gh_A10G0456, Gh_D05G3934, and Gh_D10G0473, were 8.11-, 4.02-, 3.45-, and 5.04-times higher in G24 than in W24, respectively, which explains the high accumulation of 3,4-dihydroxystyrene in G24 (Figure 6). Caffeic acid was produced from p-coumaric acid, catalyzed by C3H (Gh_A13G2072, a 2.10-fold upregulation in G24), consistent with the increased accumulations of caffeic acid in G24. Aldehyde dehydrogenase catalyzes conversion of coniferyl aldehyde into ferulic acid, and its encoding gene ALDH (Gh_D07G0047) was upregulated 3.74 times in G24, consistent with increased ferulic acid (4.52-fold) content in G24. HCT catalyzes conversion of coumaroyl-CoA into caffeoyl quinic acid. Three HCT genes were up-regulated in G24, consequently the content of caffeoyl quinic acid increased 3.99-fold. As a result, the end-product of the phenylpropanoid pathway, sinapaldehyde, increased by 104.36-fold in G24 (Figure 6). Significantly up-regulated expression of the genes encoding CHS (Gh_A12G0367, Gh_D05G2280 and Gh_D12G0299), F3H (Gh_D09G1969), F3′H (Gh_A10G0500), and FLS that convers naringenin into quercitrin and leucocyanidin accounted for the significant differences of the metabolites catalyzed by these enzymes between GCF and WCF. LAR (Gh_D12G1686, 3.54-fold upregulation) catalyzes leucopelargonidin to afzelechin, and F3′5′H (Gh_A05G0557, 1.82-fold upregulation) is involved in the conversion of dihydrokaempferol into leucodelphinidin. Increased expression of these two genes consisted with high quantities of their catalyzed flavanols in G24 (Figure 6).

2.6. Lipid Metabolism Pathway

In the cutin, suberin, and wax biosynthesis pathway, all the detected metabolites, except hexadecanoic acid and (9Z)-octadecenoic acid, were increased in G24 compared to W24. Cytochrome P450 86A1 (CYP86A1) is a fatty acid ω-hydroxylase, and cytochrome P450 86B1 (CYP86B1) is required for biosynthesis of very long chain ω-hydroxyacid and α-, ω-dicarboxylic acid [25,26]. In G24, expression of two CYP86A1 genes up-regulated 14.95- and 11.39-fold, and two CYP86B1 genes up-regulated 7.36- and 6.05-fold. This could largely explain the high accumulation of 18-hydroxyoleate and 22-hydroxydocosanoate in G24. Omega-hydroxypalmitate O-feruloyl transferase (HHT) can catalyze the conversion of p-Coumaroyl-CoA and sinapoyl-CoA, but not feruloyl-CoA [27]. Notably, the four HHT genes were found to be significantly up-regulated in G24, consistent with significant increase of sinapaldehyde but no accumulation of 16-feruloyloxypalmitic acid in G24 compared to W24.
The GCF contains a large proportion of wax, and very long-chain fatty acids/alcohols provide precursors for wax synthesis [28]. Many genes involved in the formation of long-chain fatty acids, very long-chain fatty acids, alcohols, and wax were also up-regulated in GCF, such as β-ketoacyl CoA synthase (KCS), β-ketoacyl CoA reductase (KCR), fatty acyl-CoA reductase (FAR), and long chain acyl-CoA synthetase (LACS) (Table S9).

3. Discussion

3.1. Identifying Pigmentation-Related Metabolites in Green-Colored Fiber (GCF)

Understanding the molecular mechanisms controlling pigment formation in GCF is of great importance for developing cotton varieties with stable green-colored fibers and high fiber quality as green fiber color is thought to be unstable and associated with low fiber quality. Cinnamic acid and its derivatives have been identified as GCF pigments [14,29]. Additionally, two caffeic acid derivatives have been isolated from green fibers, and were found to be positively correlated with the degree of green color in cotton fibers [1,16]. Here, we detected significant accumulation of two types of cinnamic acids (caffeic acid and ferulic acid), and five types of cinnamic acid derivatives (caffeoyl quinic acid, 3,4-dihydroxystyrene, coniferylaldehyde, 5-hydroxy coniferaldehyde, and sinapaldehyde) in GCF. In plants, cinnamic acid and its derivatives are produced through the phenylpropanoid pathway. The first step of the pathway involves PAL, which catalyzes the deamination of phenylalanine to generate cinnamic acid [30]. High PAL activity is associated with the accumulation of anthocyanins and other phenolic compounds in fruit tissues of several plant species [31,32,33]. In Arabidopsis, the double mutant pal1/pal2 produced yellow seeds, as the uncolored proanthocyanidins were unable to undergo polymerization and oxidation to produce tannin pigments [30,34]. Compared to WCF, GCF showed significantly up-regulated expression level of PAL. Treating the in vitro cultured ovules from green-colored cotton with 2-aminoindan-2 phosphonic acid, a PAL inhibitor, made them remaining white [14], suggesting that the green pigments are synthesized via the phenylpropanoid pathway. The fiber color of GCF and WCF was visually distinguishable at ~24 DPA (Figure 7), but significant expression changes of the phenylpropanoid biosynthesis genes could be observed at 18 DPA (Figure 6), indicating that 18–24 DPA could be the critical time window for color transition.
More than 1000 flavonoids have been identified to date, some of which play key roles as pigments, in pathogen resistance, and in protection against oxidative stress [35,36,37,38]. Previous study showed that pigments extracted from green fibers include flavone and flavonols [18]. We detected anthocyanin, flavone, flavanonol, and flavanols in GCF, supporting involvement of flavonoids in green cotton pigmentation. Consistent with this observation, many genes, including CHS, F3H, F3′H, F3′5′H, FLS and LAR, involved in flavonoid biosynthesis were significantly up-regulated in GCF (Figure 6). CHS catalyzes the first reaction in anthocyanin biosynthesis and is required for production of the intermediate chalcone, the primary precursor for all classes of flavonoids [39]. The CHS expression level was found to be closely correlated with the biosynthesis of flavonoid [40,41]. F3′H and F3′5′H also play critical roles in the flavonoid biosynthetic pathway. Overexpression of F3′H contributed to anthocyanin accumulation, and F3′5′H expression was essential for anthocyanin biosynthesis [42,43,44]. LAR is present in both the anthocyanin and flavanone biosynthetic pathways. LAR belongs to the reductase–epimerase–dehydrogenase family and the short-chain dehydrogenase/reductase superfamily. Each LAR has a specific carboxy-terminal domain which may have different substrate specificity [45]. Based on our metabolome and transcriptome data, LAR activity favors production of afzelechin and gallocatechin from leucopelargonidin and leucodelphinidin, rather than catechin synthesis (Figure 6). Thus, leucodelphinidin is the dominant anthocyanin in GCF. The dihydroflavonols represent a branch point in flavonoid biosynthesis, as they are the only intermediates in the production of both the colored anthocyanins, through dihydroflavonol (DFR), and the colorless flavonols, through flavonol synthase (FLS) [46]. As a result, there is competition for the dihydroflavonol substrate. Up-regulation of FLS would increase biosynthesis of flavanols that might lead to down-regulation of DFR and reduced accumulation of anthocyanin. This is consistent with the previous result showing that the FLS expression was inhibited in white-colored petunia, which resulted in accumulation of colored anthocyanin and consequently pink flowers [46]. We did not find significant differences in the amount of colored anthocyanidins between GCF and WCF, suggesting that GCF is not colored by anthocyanidins. Overall, our results suggest that the GCF pigments are composed of cinnamic acid and its derivatives, with contributions from colorless anthocyanins, flavonols, and flavanols.

3.2. Identifying Gene-Expression Modules Associated with Pigmentation in GCF

Based on WGNCA, we identified developmental-stage-specific gene expression modules associated with pigmentation in GCF (Figure 4A,B). The most intriguing one is the blue module associated with G24, in which the two homoeologous Gh4CL4 genes [16] were identified as candidate hub genes (Figure 5B). 4CL is a key enzyme in the phenylpropanoid biosynthesis pathway, catalyzing the formation of CoA-esters of cinnamic acids and their derivatives, which are the substrates for biosynthesis of flavonoids, lignin, suberin, coumarin, and wall-bound phenolic compounds [22,47]. The result obtained in this study supported our previous finding that Gh4CL4 might be involved in the metabolism of caffeic and ferulic residues to affect pigmentation in GCF, because Gh4CL4 was preferably associated with these two substrates in GCF [16].
Notably, many genes involved in long-chain fatty acid, very long-chain fatty acid, alcohol, and wax synthesis were also enriched in G24 (Table S7). Long-chain fatty acids, very long-chain fatty acids, and alcohols are precursors of wax synthesis. In Arabidopsis, KCS2 and KCS20 are required to add two carbons to C22 fatty acids during cuticular wax and root suberin biosynthesis [48]. Suppressing KCR activity results in a reduction of cuticular wax production [49]. Overexpression of CER6 gene leads to increases epidermal wax in the stem [28]. Heterologous expression of the FAR gene can produce significant amounts of fatty alcohols in cuticle waxes of plant [50]. We found that all these genes were significantly up-regulated in GCF, consistent with their roles in accumulation of wax in GCF [15].
In G24-specific module (blue module), the hub genes (Table 3) of metal transporter related were also highly expressed in GCF. These include Fe2+ transport protein, copper-transporting ATPase, copper transporter, zinc transporter, metal transporter NRAMP, and heavy metal-associated isoprenylated plant protein (HIPP). The levels of Fe2+ and Cu2+ were higher in GCF than in WCF [51]. FeCl2 and CuSO4 have been used to treat green cotton fabric to deepen the shade, and Fe2+ seems to have a greater effect than Cu2+ [52]. It has been suggested that although non-ferrous metal ions, such as Cu2+ and Fe2+, are not components of pigments in GCF, they may chelate with pigment substances to form more stable structures that may alter the color of cotton fibers.

4. Materials and Methods

4.1. Plant Materials and Treatments

Upland cotton (G. hirsutum) cultivar Xincaimian No. 7 (C7) with GCF and C7-NIL were grown at the Shihezi University Experimental Station in Shihezi City (44°27′ N, 85°94′ E), Xinjiang Autonomous Region, China. GCF samples were collected from developing bolls at 12, 18, and 24 days post-anthesis (DPA), designated G12, G18, and G24, respectively. WCF samples were also collected at the same time and designated W12, W18, and W24 (Figure 7). The samples were frozen immediately in liquid nitrogen and stored at −80 °C until use.

4.2. Metabolite Extraction and Profiling

Fresh fiber samples (25 mg) were weighed, and put into Eppendorf tubes with 800 µL pre-cooled methanol and water solution in a 1:1 ratio, then grounded with a metal ball homogenizer (35 Hz for 5 min). The grounded samples were incubated at −20 °C for 2 h and centrifuged for 15 min at 4 °C at 2500 relative centrifugal field (RCF). A 550 µL aliquot of the supernatant was moved into a new Eppendorf tube for subsequent analysis.
A total of 30 samples (3 time points × 2 genotypes × 5 biological replicates) were prepared and analyzed according to the guidelines of the liquid chromatography–mass spectrometry (LC–MS) system. Firstly, all chromatographic separations were performed using an ultra-high performance liquid chromatography (UPLC) system (Waters, Milford, USA). An ACQUITY UPLC BEH C18 column (100 mm*2.1 mm, 1.7 μm, Waters, Milford, USA) was used for reversed phase separation. The column oven was maintained at 50 °C. The mobile phase contained solvent A (water + 0.1% formic acid), and solvent B (acetonitrile + 0.1% formic acid) with a flow rate of 0.4 mL/min. Gradient elution conditions were as follows: 0–2 min with 100% solvent A; 2–11 min with 0–100% solvent B; 11–13 min with 100% solvent B; 13–15 min, 0 to 100% solvent A. The injection volume for each sample was 10 µL.
A high-resolution tandem quadrapole time of flight (QTOF) mass spectrometer, Xevo G2 XS (Waters, UK), was used to detect metabolites eluted from the column. QTOF was operated in both positive electrospray ionization (ESI+) and negative electrospray ionization (ESI) modes. For ESI+, the capillary and sampling cone voltages were set at 2 kV and 40 V, respectively. For ESI, the capillary and sampling cone voltages were set at 1 kV and 40 V, respectively. Mass spectrometry data were acquired in the MSE centroid mode. TOF mass range was 50–1200 Da, and the scan time was 0.2 s. For the MS/MS detection, all precursors were fragmented using 20–40 eV, and the scan time was 0.2 s. During acquisition, the acquisition rate was set to 3 s to calibrate accuracy of mass measurements. Furthermore, in order to evaluate the stability of the LC-MS during acquisition, a quality control (QC) sample, which is a pool of all the samples, was acquired after every 10 samples.

4.3. Metabolite Identification and Quantification

Mass spectrum peaks were extracted using the commercial software Progenesis QI (version 2.2, Waters, Milford, USA). The metabolite structures, m/z values, retention time (RT), and the fragmentation pattern were identified based on the available metabolite databases. The mass ion peaks were selected using product ion scan during the first stage of mass spectrometry (MS1), and precursor ion scan during the second stage of mass spectrometry (MS2). The selected peaks were identified by MS2 fragment patterns. Total ion chromatograms (TICs) and extracted ion chromatograms (EICs or XICs) of QC samples that summarize the metabolic profiles of all samples were used to calculate the area under each peak.
Variable importance of the projection (VIP) scores of partial least-squares discriminant analysis (PLS-DA) was used to rank metabolites between the two upland cotton accessions. The VIP threshold was set to 1. In addition, ratio and t-test were also used as a univariate analysis to screen metabolites between samples. Samples with a ratio ≥ 1.2 or ≤ 0.83, p-value < 0.05 and VIP ≥ 1 were considered to be metabolites that were differentially present.

4.4. RNA Isolation and Illumina Sequencing

Total RNA was extracted from 12, 18 and 24 DPA fibers of C7 and C7-NIL using the Tiangen RNA Extraction Kits. Residue genomic DNA was degraded with DNase I (Promega, Beijing, China). RNA quantity and quality were determined by NanoDrop ND2000 spectrophotometer (NanoDrop Technologies, Wilmington, DE, USA), and Agilent Bioanalyzer 2100 system (Agilent Technologies, Palo Alto, CA, USA), respectively. RNA integrity was further determined by 1% agarose gel electrophoresis. A total of 3 µg total RNA per sample (in total 18 samples = 3 time points × 2 genotypes × 3 biological replicates) was used as input material for RNA-seq library preparation. RNA-seq (150 bp paired-end reads) was performed on an Illumina Hiseq 4000 platform. The whole set of annotated genes can be found in the National Center for Biotechnology Information (NCBI) SRA database (BioProject accession: PRJNA565616). Raw data (in FASTq format) were processed using Perl scripts to measure Q20, Q30, and GC content. After removing low-quality reads, the remaining cleaned data were used in all downstream analysis.

4.5. RNA Sequencing Data Analysis

The G. hirsutum reference genome and gene model annotation files were downloaded from https://www.cottongen.org (accessed on February 7, 2018) [19]. Indexes of the reference genome were built using Bowtie (version 2.2.3, Johns Hopkins University, Baltimore, Maryland, USA) and the paired-end clean RNA-seq reads were aligned to the reference genome using TopHat (version 2.0.12, Maryland University and California University, College Park and Oakland City, State of Maryland and California, USA) was used to count the number of reads mapped to each gene. The fragments per kilobase of transcript per million mapped reads (FPKM) of each gene was calculated and used to quantify the expression level of the annotated genes. Genes differentially expressed (DEG) between GCF and WCF (each with three biological replicates) were identified by DESeq packaged in R (1.18.0). DESeq can determine differential expression using a model based on negative binomial distribution. The resulting p-values were adjusted using the Benjamini and Hochberg’s approach to control the false discovery rate (FDR). Genes with an adjusted p-value < 0.05 were considered to be differentially expressed.

4.6. Assignment of MapMan Bins and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathways

The functions of DEGs were annotated based on their homology with Arabidopsis genes annotated by TAIR (The Arabidopsis Information Resource) and classified into MapMan bins using the Mercator pipeline for automated sequence annotation (http://mapman.gabipd.org/web/guest/app/mercator, accessed on March 5, 2018) [53]. Pathway enrichment was done using the KEGG orthology-based annotation system (KOBAS) based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database [54].

4.7. Gene Network Construction and Visualization

Co-expression networks were constructed using the WGCNA (v1.29, Department of Human Genetics, University of California, Los Angeles, CA, USA) package in R [55]. The modules were obtained using the automatic block-wise network construction approach. Modules were identified based on the default settings, with the exceptions of power, minModuleSize and merge CutHeight being set to 14, 30, and 0.25, respectively, and TOMType being selected. The eigengene value was calculated for each module and used to test for association with each sample. Total connectivity and intramodular connectivity (function soft Connectivity), KME (for modular membership, also known as eigengene-based connectivity), and p_value were calculated. Each of the module genes are shown in Table S1. The networks were visualized using Cytoscape [56].

4.8. Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR)

The same RNA samples used in RNA-seq were used in RT-qPCR. Three microliters of total RNA were used to synthesize cDNA by using oligo (dT) and M-MLV Reverse Transcriptase (Takara, Dalian, China) according to the manufacturer’s instructions. cDNAs were then loaded in a 96-well plate for qRT-PCR analysis with a Light Cycler® 480 II system (Roche, Switzerland) using the Power SYBR Green PCR Master Mixture (Roche, Switzerland), 10 µl reactions contained 1 µL of cDNA, 100 nM of each pair of target primers and 5 µL of SYBR Green PCR Master Mix. PCR conditions were as follows: 95 °C for 5 min, 40 cycles of 94 °C for 10 s, 60 °C for 10 s, and 72 °C for 10 s. Relative gene expression levels were analyzed according to the 2ΔΔCt method. The internal normalization gene was GhHistone 3. Primers were designed using National Center for Biotechnology Information PrimerBLAST tools (Available online: http://www.ncbi.nlm.nih.gov/tools/primer-blast/, accessed on March 27, 2018). Product specificity and reaction efficiencies were verified for each primer pair. Primer pairs are listed in Table S2.

Supplementary Materials

The following are available online at https://www.mdpi.com/1422-0067/20/19/4838/s1.

Author Contributions

J.S. and Y.-j.L. coordinated the project, conceived and designed experiments; S.S. and X.-p.X. conducted the bioinformatics work, analyzed the data and drafted the manuscript; Q.Z. contributed valuable discussion and language modification. All authors read and approved the final manuscript.

Funding

This work was supported by the National Key Research and Development Program of China (Grant No. 2016YFD0101900, 2017YFD0101600), the National Natural Science Foundation of China (Grant No. 31360347).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Ma, M.; Hussain, M.; Memon, H.; Zhou, W. Structure of pigment compositions and radical scavenging activity of naturally green-colored cotton fiber. Cellulose 2016, 23, 955–963. [Google Scholar] [CrossRef]
  2. Vreeland, J.M. The revival of colored cotton. Sci. Am. 1999, 280, 112–118. [Google Scholar] [CrossRef]
  3. Dutt, Y.; Wang, X.D.; Zhu, Y.G.; Li, Y.Y. Breeding for high yield and fibre quality in colored cotton. Plant. Breed. 2004, 123, 145–151. [Google Scholar] [CrossRef]
  4. Li, Y.J.; Zhang, X.Y.; Wang, F.X.; Yang, C.L.; Liu, F.; Xia, G.X.; Sun, J. A comparative proteomic analysis provides insights into pigment biosynthesis in brown color fiber. J. Proteom. 2013, 78, 374–388. [Google Scholar] [CrossRef] [PubMed]
  5. Murthy, M.S.S. Never say dye: The story of coloured cotton. Resonance 2001, 6, 29–35. [Google Scholar] [CrossRef]
  6. Yuan, S.; Hua, S.; Malik, W.; Bibi, N.; Wang, X. Physiological and biochemical dissection of fiber development in colored cotton. Euphytica 2012, 187, 215–226. [Google Scholar] [CrossRef]
  7. Kohel, R.J. Genetic Analysis of Fiber Color Variants in Cotton 1. Crop. Sci. 1985, 25, 793–797. [Google Scholar] [CrossRef]
  8. Xiao, Y.H.; Yan, Q.; Ding, H.; Luo, M.; Hou, L.; Zhang, M.; Yao, D.; Liu, H.S.; Li, X.; Zhao, J.; et al. Transcriptome and biochemical analyses revealed a detailed proanthocyanidin biosynthesis pathway in brown cotton fiber. PLoS ONE 2014, 9, e86344. [Google Scholar] [CrossRef]
  9. Feng, H.; Li, Y.; Wang, S.; Zhang, L.; Liu, Y.; Xue, F.; Sun, Y.; Wang, Y.; Sun, J. Molecular analysis of proanthocyanidins related to pigmentation in brown cotton fibre (gossypium hirsutum L.). J. Exp. Bot. 2014, 65, 5759–5769. [Google Scholar] [CrossRef]
  10. Yan, Q.; Wang, Y.; Li, Q.; Zhang, Z.; Ding, H.; Zhang, Y.; Liu, H.; Luo, M.; Liu, D.; Song, W.; et al. Up-regulation of GhTT2-3A in cotton fibres during secondary wall thickening results in brown fibres with improved quality. Plant. Biotechnol. J. 2018, 16, 1735–1747. [Google Scholar] [CrossRef]
  11. Ryser, U.; Meier, H.; Holloway, P.J. Identification and locailzation of suberin in the cell wall of green (Gossypium hirsutum L. Var. green lint). Protoplasma 1983, 117, 196–205. [Google Scholar] [CrossRef]
  12. Yatsu, L.Y.; Espelie, K.E.; Kolattukudy, P.E. Ultrastructural and chemical evidence that the cell wall of green cotton fber is suberized. Plant. Physiol. 1983, 73, 521–524. [Google Scholar] [CrossRef] [PubMed]
  13. Conrad, C.M. The high wax content of green lint cotton. Science 1941, 94, 113. [Google Scholar] [CrossRef] [PubMed]
  14. Schmutz, A.; Jenny, T.; Amrhein, N.; Ryser, U. Caffeic acid and glycerol are constituents of the suberin layers in green cotton fibres. Planta 1993, 189, 453–460. [Google Scholar] [CrossRef] [PubMed]
  15. Schmutz, A.; Jenny, T.; Ryser, U. A caffeoyl-fatty acid-glycerol ester from wax associated with green cotton fibre suberin. Phytochemistry 1994, 36, 1343–1346. [Google Scholar] [CrossRef]
  16. Feng, H.J.; Yang, Y.L.; Sun, S.C.; Li, Y.; Zhang, L.; Tian, J.; Zhu, Q.; Feng, Z.; Zhu, H.; Sun, J. Molecular analysis of caffeoyl residues related to pigmentation in green cotton fibers. J. Exp. Bot. 2017, 68, 4559–4569. [Google Scholar] [CrossRef]
  17. Schmutz, A.; Buchala, A.J.; Ryser, U. Changing the dimensions of suberin lamellae of green cotton fibers with a specific inhibitor of the endoplasmic reticulum-associated fatty acid elongases. Plant. Physiol. 1996, 110, 403–411. [Google Scholar] [CrossRef] [PubMed]
  18. Zhao, X.Q.; Wang, X.D. Composition analysis of pigment in colored cotton fiber. Acta Agron. Sin. 2005, 31, 456–462. [Google Scholar]
  19. Zhang, T.; Hu, Y.; Jiang, W.; Fang, L.; Guan, X.; Chen, J.; Zhang, J.; Saski, C.A.; Scheffler, B.E.; Stelly, D.M.; et al. Sequencing of allotetraploid cotton (gossypium hirsutum l. acc. tm-1) provides a resource for fiber improvement. Nat. Biotechnol. 2015, 33, 531–537. [Google Scholar] [CrossRef]
  20. Trapnell, C.; Williams, B.A.; Pertea, G.; Mortazavi, A.; Kwan, G.; Baren, M.J.; Salzberg, S.L.; Wold, B.J.; Pachter, L. Transcript assembly and quantifcation by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 2010, 28, 511–515. [Google Scholar] [CrossRef]
  21. Li, Y.J.; Wang, F.X.; Wang, Y.Q.; Liu, Y.C.; Zhang, X.Y.; Sun, Y.Q.; Sun, J. Identification of the proteins in green cotton fiber using a proteomics-based approach. Biotechnol. Lett. 2013, 35, 1519–1523. [Google Scholar] [CrossRef] [PubMed]
  22. Ehlting, J.; Buttner, D.; Wang, Q.; Douglas, C.J.; Somssich, I.E.; Kombrink, E. Three 4-coumarate: Coenzyme A ligases in Arabidopsis thaliana represent two evolutionarily divergent classes in angiosperms. Plant. J. 1999, 19, 9–20. [Google Scholar] [CrossRef] [PubMed]
  23. Gianoulis, T.A.; Griffin, M.A.; Spakowicz, D.J.; Dunican, B.F.; Alpha, C.J.; Sboner, A.; Sismour, A.M.; Kodira, C.; Egholm, M.; Church, G.M. Genomic Analysis of the Hydrocarbon-Producing, Cellulolytic, Endophytic Fungus Ascocoryne sarcoides. PLoS Genet. 2012, 8, e1002558. [Google Scholar] [CrossRef] [PubMed]
  24. Dixon, R.A.; Achnine, L.; Kota, P.; Liu, C.J.; Reddy, M.S.S.; Wang, L. The phenylpropanoid pathway and plant defence a genomics perspective. Mol. Plant. Pathol. 2002, 3, 371–390. [Google Scholar] [CrossRef] [PubMed]
  25. Compagnon, V.; Diehl, P.; Benveniste, I.; Meyer, D.; Schaller, H.; Schreiber, L.; Franke, R.; Pinot, F. CYP86B1 is required for very long chain omega-hydroxyacid and alpha, omega-dicarboxylic acid synthesis in root and seed suberin polyester. Plant. Physiol. 2009, 150, 1831–1843. [Google Scholar] [CrossRef] [PubMed]
  26. Rene, H.; Isabel, B.; Martina, B.; Franck, P.; Lukas, S.; Rochus, F. The arabidopsis cytochrome p450 cyp86a1 encodes a fatty acid ω-hydroxylase involved in suberin monomer biosynthesis. J. Exp. Bot. 2008, 59, 2347–2360. [Google Scholar]
  27. Lotfy, S.; Negrel, J.; Javelle, F. Formation of ω-feruloyloxypalmitic acid by an enzyme from wound-healing potato tuber discs. Phytochemistry 1994, 35, 1419–1424. [Google Scholar] [CrossRef]
  28. Hooker, T.S.; Millar, A.A.; Kunst, L. Significance of the expression of the CER6 condensing enzyme for cuticular wax production in Arabidopsis. Plant. Physiol. 2002, 129, 1568–1580. [Google Scholar] [CrossRef]
  29. Moire, L.; Schmutz, A.; Buchala, A.; Yan, B.; Stark, R.E.; Ryser, U. Glycerol is a suberin monomer: New experimental evidence for an old hypothesis. Plant. Physiol. 1999, 119, 1137–1146. [Google Scholar] [CrossRef]
  30. Huang, J.; Gu, M.; Lai, Z.; Fan, B.; Shi, K.; Zhou, Y.H.; Yu, J.Q.; Chen, Z.X. Functional analysis of the arabidopsis pal gene family in plant growth, development, and response to environmental stress. Plant. Physiol. 2010, 153, 1526–1538. [Google Scholar] [CrossRef]
  31. Blankenship, S.M.; Unrath, C.R. PAL and ethylene content during maturation of Red and Golden Delicious apples. Photochemistry 1988, 27, 1001–1003. [Google Scholar] [CrossRef]
  32. Kataoka, I.; Kubo, Y.; Sugiura, A.; Tomana, T. Changes in L-phenylalanine ammonia-lyase activity and anthocyanin synthesis during berry ripening of three grape cultivars. J. Jpn. Soc. Hort. Sci. 1983, 52, 273–279. [Google Scholar] [CrossRef]
  33. Cheng, G.W.; Breen, P.J. Activity of Phenylalanine Ammonia-Lyase (PAL) and Concentrations of Anthocyanins and Phenolics in Developing Strawberry Fruit. J. Amer. Soc. Hort. Sci. 1991, 116, 865–869. [Google Scholar] [CrossRef]
  34. Debeaujon, I.; Leon-Kloosterziel, K.M.; Koornneef, M. Influence of the testa on seed dormancy, germination, and longevity in Arabidopsis. Plant. Physiol. 2000, 122, 403–414. [Google Scholar] [CrossRef] [PubMed]
  35. Dixon, R.A.; Paiva, N.L. Stress-induced phenylpropanoid metabolism. Plant. Cell 1995, 7, 1085–1097. [Google Scholar] [CrossRef] [PubMed]
  36. Landry, L.G.; Chapple, C.C.S.; Last, R.L. Arabidopsis mutants lacking phenolic sunscreens exhibit enhanced ultraviolet-B injury and oxidative damage. Plant. Physiol. 1995, 109, 1159–1166. [Google Scholar] [CrossRef] [PubMed]
  37. Winkel-Shirley, B. Flavonoid biosynthesis: A colorful model for genetics, biochemistry, cell biology, and biotechnology. Plant. Physiol. 2001, 126, 485–493. [Google Scholar] [CrossRef]
  38. Tanaka, Y.; Sasaki, N.; Ohmiya, A. Biosynthesis of plant pigments:anthocyanins, betalains and carotenoids. Plant. J. 2008, 54, 733–749. [Google Scholar]
  39. Koes, R.E.; Spelt, C.E.; Mol, J.N.M. The chalcone synthase multigene family ofpetunia hybrida(v30): Differential, light-regulated expression during flower development and uv light induction. Plant. Mol. Biol. 1989, 12, 213–225. [Google Scholar] [CrossRef]
  40. Ohno, S.; Hosokawa, M.; Kojima, M.; Kitamura, Y.; Hoshino, A.; Tatsuzawa, F.; Doi, M.; Yazawa, S. Simultaneous post-transcriptional gene silencing of two different chalcone synthase genes resulting in pure white flowers in the octoploid dahlia. Planta 2011, 234, 945–958. [Google Scholar] [CrossRef] [Green Version]
  41. Ohno, S.; Hori, W.; Hosokawa, M.; Tatsuzawa, F.; Doi, M. Post-transcriptional silencing of chalcone synthase is involved in phenotypic lability in petals and leaves of bicolor dahlia (Dahlia variabilis) ‘Yuino’. Planta 2018, 247, 413–428. [Google Scholar] [CrossRef] [PubMed]
  42. Ueyama, Y.; Suzuki, K.; Fukuchi-Mizutani, M.; Fukui, Y.; Miyazaki, K.; Ohkawa, H.; Kusumi, T.; Tanaka, Y. Molecular and biochemical characterization of torenia flavonoid 3′-hydroxylase and flavone synthase II and modification of flower color by modulating the expression of these genes. Plant. Sci. 2002, 163, 253–263. [Google Scholar] [CrossRef]
  43. Han, Y.; Vimolmangkang, S.; Soria-Guerra, R.E.; Rosales-Mendoza, S.; Zheng, D.; Korban, S.S. Ectopic expression of apple F3′H genes contributes to anthocyanin accumulation in the arabidopsis tt7 mutant grown under nitrogen stress. Plant. Physiol. 2010, 153, 806–820. [Google Scholar] [CrossRef] [PubMed]
  44. Tanaka, Y.; Brugliera, F. Flower colour and cytochromes P450. Phytochem. Rev. 2006, 5, 283–291. [Google Scholar] [CrossRef]
  45. Tanner, G.J.; Francki, K.T.; Abrahams, S.; Watson, J.M.; Larkin, P.J.; Ashton, A.R. Proanthocyanidin biosynthesis in plants. Purification of legume leucoanthocyanidin reductase and molecular cloning of its cDNA. J. Biol. Chem. 2003, 278, 3147–3156. [Google Scholar] [CrossRef] [PubMed]
  46. Davies, K.M.; Schwinn, K.E.; Deroles, S.C.; Manson, D.G.; Lewis, D.H.; Bloor, S.J.; Bradley, J.M. Enhancing anthocyanin production by altering competition for substrate between flavonol synthase and dihydroflavonol reductase. Euphytica 2003, 131, 259–268. [Google Scholar] [CrossRef]
  47. Beuerle, T.; Pichersky, E. Enzymatic synthesis and purification of aromatic coenzyme a esters. Anal. Biochem. 2002, 302, 305–312. [Google Scholar] [CrossRef]
  48. Lee, S.B.; Jung, S.J.; Go, Y.S.; Kim, H.U.; Kim, J.K.; Cho, H.J.; Park, O.K.; Suh, M.C. Two Arabidopsis 3-ketoacyl CoA synthase genes, KCS20 and KCS2/DAISY, are functionally redundant in cuticular wax and root suberin biosynthesis, but differentially controlled by osmotic stress. Plant. J. 2009, 60, 462–475. [Google Scholar] [CrossRef]
  49. Beaudoin, F.; Wu, X.Z.; Li, F.; Haslam, R.P.; Markham, J.E.; Zheng, H. Functional characterization of the arabidopsis β-ketoacyl-coenzyme a reductase candidates of the fatty acid elongase. Plant. Physiol. 2009, 150, 1174–1191. [Google Scholar] [CrossRef]
  50. Doan, T.T.P.; Carlsson, A.S.; Hamberg, M.; Leif, B.; Stymne, S.; Olsson, P. Functional expression of five arabidopsis fatty acyl-coa reductase genes in escherichia coli. J. Plant. Physiol. 2009, 166, 787–796. [Google Scholar] [CrossRef]
  51. Yang, M.; Gu, L.Q.; Qian, H.S.; Zha, L.S. Comparative study on trace elements in natural colored cotton and white cotton. Spectrosc Spect Anal. 2008, 28, 203–205. [Google Scholar]
  52. Gu, Y.; Cai, X.J.; Li, M.S.; Zhou, W.L. An approach to the color properties of naturally colored cottons after metal salts treatment. J. Zhejiang Sci.-Tech. Univ. 2008, 25, 10–14. [Google Scholar]
  53. Thimm, O.; Bläsing, O.; Gibon, Y.; Nagel, A.; Meyer, S.; Krüger, P.; Selbig, J.; Müller, L.A.; Rhee, S.Y.; Stitt, M. Mapman: A user-driven tool to display genomics data sets onto diagrams of metabolic pathways and other biological processes. Plant. J. 2010, 37, 914–939. [Google Scholar] [CrossRef] [PubMed]
  54. Xie, C.; Mao, X.; Huang, J.; Ding, Y.; Wu, J.; Dong, S.; Kong, L.; Gao, G.; Li, C.Y.; Wei, L. Kobas 2.0: A web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res. 2011, 39, W316–W322. [Google Scholar] [CrossRef]
  55. Langfelder, P.; Horvath, S. WGCNA: An R package for weighted correlation network analysis. Bioinformatics 2008, 9, 559. [Google Scholar] [CrossRef]
  56. Kohl, M.; Wiese, S.; Warscheid, B. Cytoscape: Software for Visualization and Analysis of Biological Networks. Methods Mol. Biol. 2011, 696, 291–303. [Google Scholar]
Figure 1. Comparison of metabolites from different developmental stages of cotton fibers. (A) Principal component analysis (PCA) score plot derived from metabolite ions acquired using the electrospray ionization positive ion mode (ESI+). (B) PCA score plot derived from metabolite ions acquired using the electrospray ionization nagative ion mode (ESI). (C) Venn diagram showing different metabolites identified between green-colored fiber (GCF) and white-colored fiber (WCF). (D) The number of up- (red) and down-regulated (green) metabolites in each comparison.
Figure 1. Comparison of metabolites from different developmental stages of cotton fibers. (A) Principal component analysis (PCA) score plot derived from metabolite ions acquired using the electrospray ionization positive ion mode (ESI+). (B) PCA score plot derived from metabolite ions acquired using the electrospray ionization nagative ion mode (ESI). (C) Venn diagram showing different metabolites identified between green-colored fiber (GCF) and white-colored fiber (WCF). (D) The number of up- (red) and down-regulated (green) metabolites in each comparison.
Ijms 20 04838 g001
Figure 2. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation of all the different metabolites.
Figure 2. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation of all the different metabolites.
Ijms 20 04838 g002
Figure 3. Differentially expressed genes (DEGs) between GCF and WCF. (A) The total number of DEGs identified in each comparison. (B) Venn diagram showing DEGs between GCF and WCF in the three time points. (C) Venn diagram showing DEGs between different time points in GCF or WCF. (D) The total number of non-redundant DEGs between different time points as well as between GCF and WCF.
Figure 3. Differentially expressed genes (DEGs) between GCF and WCF. (A) The total number of DEGs identified in each comparison. (B) Venn diagram showing DEGs between GCF and WCF in the three time points. (C) Venn diagram showing DEGs between different time points in GCF or WCF. (D) The total number of non-redundant DEGs between different time points as well as between GCF and WCF.
Ijms 20 04838 g003
Figure 4. Weighted gene co-expression network analysis (WGCNA) of DEGs between GCF and WCF. (A) Hierarchical cluster tree showing co-expression modules identified by WGCNA. Each leaf in the tree represents one gene. Each major tree branch represents a distinct module, in total, there were 16 modules labeled by different colors. (B) Module-sample association relationships. Each row corresponds to a module, labeled by the same color as in (A). The number of genes in each module is shown next to the module name. Each column corresponds to a specific tissue. The correlation coefficient and p-value between the module and the sample or tissue are shown at the row-column intersection.
Figure 4. Weighted gene co-expression network analysis (WGCNA) of DEGs between GCF and WCF. (A) Hierarchical cluster tree showing co-expression modules identified by WGCNA. Each leaf in the tree represents one gene. Each major tree branch represents a distinct module, in total, there were 16 modules labeled by different colors. (B) Module-sample association relationships. Each row corresponds to a module, labeled by the same color as in (A). The number of genes in each module is shown next to the module name. Each column corresponds to a specific tissue. The correlation coefficient and p-value between the module and the sample or tissue are shown at the row-column intersection.
Ijms 20 04838 g004
Figure 5. Co-expression network analysis of a stage-specific module. (A) Heatmap showing genes in the blue module that were over-expressed at G24. (B) Correlation networks of hub genes in the blue module. The two homoeologous Gh4CL4 genes are shown in red. (C) The enriched KEGG pathways of the blue module genes.
Figure 5. Co-expression network analysis of a stage-specific module. (A) Heatmap showing genes in the blue module that were over-expressed at G24. (B) Correlation networks of hub genes in the blue module. The two homoeologous Gh4CL4 genes are shown in red. (C) The enriched KEGG pathways of the blue module genes.
Ijms 20 04838 g005
Figure 6. Transcript and metabolic profiling of genes in the phenylpropanoid and flavonoid biosynthetic pathways in cotton. PAL, phenylalanine ammonia-lyase; 4CL, 4-coumarate CoA ligase; C3H, cinnamate 3-hydroxylase; ALDH, Aldehyde dehydrogenase; HCT, shikimate O-hydroxycinnamoyltransferase; CCoAOMT, caffeoyl-coenzyme A O-methyltransferase; CHS, chalcone synthase; F3H, flavanone 3-hydroxylase; F3′H, flavanoid 3′-hydroxylase; DFR, dihydroflavonol 4-reductase; FLS, flavonol synthesis; LAR, leucocyanidin reductase.
Figure 6. Transcript and metabolic profiling of genes in the phenylpropanoid and flavonoid biosynthetic pathways in cotton. PAL, phenylalanine ammonia-lyase; 4CL, 4-coumarate CoA ligase; C3H, cinnamate 3-hydroxylase; ALDH, Aldehyde dehydrogenase; HCT, shikimate O-hydroxycinnamoyltransferase; CCoAOMT, caffeoyl-coenzyme A O-methyltransferase; CHS, chalcone synthase; F3H, flavanone 3-hydroxylase; F3′H, flavanoid 3′-hydroxylase; DFR, dihydroflavonol 4-reductase; FLS, flavonol synthesis; LAR, leucocyanidin reductase.
Ijms 20 04838 g006
Figure 7. Fiber phenotypes of the green-colored fibers (GCF) and white-colored fibers (WCF) at 12, 18, and 24 days post-anthesis (DPA).
Figure 7. Fiber phenotypes of the green-colored fibers (GCF) and white-colored fibers (WCF) at 12, 18, and 24 days post-anthesis (DPA).
Ijms 20 04838 g007
Table 1. Upregulated phenylpropanoids in G24 compared to W24.
Table 1. Upregulated phenylpropanoids in G24 compared to W24.
ComponentMetabolite NameLog2 (Fold Change)VIPExact Mass (m/z)
(G24/W24)
IntermediatesCaffeic acid16.063.55179.0336
Ferulic acid4.522.86193.049
Sinapaldehyde104.363.46207.0648
3,4-Dihydroxystyrene2.212.1136.0529
Coniferyl aldehyde1.351.12161.0601
5-Hydroxyconiferaldehyde4.522.86177.055
Caffeoyl quinic acid3.982.2355.0999
FlavoneNaringenin chalcone3.391.44271.0596
Flavanol(+)-Gallocatechin1.91.2305.066
FlavonolAfzelin3.832.24455.0991
Quercitrin6.672.64449.1087
AnthocyaninLeucodelphinidin1.541323.0751
VIP: Variable Importance in the Projection.
Table 2. Upregulated metabolites of the cutin, suberin, and wax biosynthesis pathway in G24 compared to W24.
Table 2. Upregulated metabolites of the cutin, suberin, and wax biosynthesis pathway in G24 compared to W24.
Metabolite NameLog2 (Fold Change)VIPExact Mass (m/z)
(G24/W24)
Docosanedioate11.852.5369.2993
22-Oxo-docosanoate1.851.37353.3067
22-Hydroxydocosanoate8.822.38355.3204
9,10-Dihydroxystearate3.032315.2526
9,10-Epoxy-18-hydroxystearate1.651.34313.2365
cis-9,10-Epoxystearic acid1.721.16297.2418
18-Hydroxyoleate2.011.61298.2513
Hexadecanedioate2.561.95285.2057
16-Hydroxypalmitic acid1.541.2271.2261
16-Oxo-palmitate1.451.01269.2104
Table 3. List of the blue module hub genes.
Table 3. List of the blue module hub genes.
Gene IDDescriptionKME Value
Lipid Metabolism
Gh_D11G1853Epoxide hydrolase 3, EPHX30.997
Gh_A11G1143Non-specific lipid-transfer protein-like protein, At2g13820 0.997
Gh_D02G0380Lipid binding protein0.997
Gh_D09G1221Fatty acyl-CoA reductase, FAR0.997
Gh_A05G1996Probable glucan endo-1,3-beta-glucosidase 1, BG1 0.997
Gh_D08G0086Fatty acyl-CoA reductase, FAR0.995
Gh_D11G1294Non-specific lipid-transfer protein-like protein, At2g138200.995
Gh_A08G1982Sterol 3-beta-Glucosyltransferase, UGT80A20.995
Gh_A11G0514Diacylglycerol O-acyltransferase 2, DGAT20.994
Gh_D01G06321-acyl-sn-glycerol-3-phosphate acyltransferase, AGPAT0.993
Gh_D01G2045Probable glycosyltransferase, At5g037950.993
Gh_D05G3800Probable glucan endo-1,3-beta-glucosidase, BG10.992
Gh_A13G0445 Lipid binding protein0.992
Gh_A09G1215Fatty acyl-CoA reductase, FAR0.992
Gh_D10G0915GDSL glycine (G), aspartic acid (D), serine (S) and leucine (L) esterase/lipase, At2g235400.991
Gh_D09G1967Xyloglucan Galactosyltransferase KATAMARI1 homolog, Os03g01448000.991
Gh_D08G2376Sterol 3-beta-Glucosyltransferase, UGT80A20.991
Gh_D11G1156Triacylglycerol lipase0.991
Gh_D07G1045GDSL esterase/lipase, At5g228100.990
Gh_A05G2906Lipid transfer-like protein, VAS0.990
Phenylpropanoid Biosynthesis
Gh_A10G04564-coumarate-CoA ligase, 4CL0.996
Gh_D10G04734-coumarate-CoA ligase, 4CL0.995
Gh_D03G1701Caffeoylshikimate esterase, CSE0.991
RNA
Gh_A13G1886Scarecrow-like protein, SCL30.995
Novel06214NAC domain protein0.994
Gh_D08G1424Probable WRKY transcription factor 43, WRKY430.994
Gh_D09G1008LOB domain-containing protein 1, LBD1 0.993
Gh_A06G1158Putative Myb family transcription factor, At1g146000.991
Gene IDDescriptionKME Value
Protein
Gh_A05G0511Aspartyl protease family protein 2, NEP20.998
Gh_A05G1182RING-H2 finger protein, ATL30.991
Gh_A11G1587Aspartic proteinase-like protein, At5g100800.990
Signalling
Gh_A11G2221Cysteine-rich repeat secretory protein, CRRSP30.992
Gh_D13G0421LRR receptor-like serine/threonine-protein kinase, LRR–RLK0.992
Gh_D07G1827Receptor-like protein kinase, FER0.991
Transport
Gh_A12G0165Nucleobase-ascorbate transporter, NAT0.997
Gh_D08G0448Heavy metal-associated isoprenylated plant protein, HIPP0.996
Gh_A07G2010Aquaporin, SIP0.994
Gh_D02G0459Protein NRT1/ PTR family, NPF0.992
Gh_D02G0450Phosphate transporter, PHO0.992
Gh_D06G1464Aquaporin, SIP0.992
Gh_D10G0695ADP,ATP carrier protein 1, chloroplastic, AATP0.991
Gh_D12G2828Nucleobase-ascorbate transporter, NAT0.991
Gh_D10G0695ADP,ATP carrier protein 1, chloroplastic, AATP0.991
Gh_D09G1048ABC transporter G family member 23, ABCG230.990
Hormone
Gh_A12G2038Probable aminotransferase 10, ACS100.990
Others
Gh_A07G1506Pentatricopeptide repeat-containing protein, At3g221500.994
Gh_D10G1737Uncharacterized protein0.994
Gh_D08G1326Uncharacterized protein0.994
Gh_A04G0984Periaxin, Prx0.993
Novel01048Uncharacterized protein0.992
Gh_A11G1731Uncharacterized protein0.992
Gh_D07G0827Condensin complex subunit0.992
Gh_D07G1315Uncharacterized protein0.992
Novel02826Uncharacterized protein0.991
Gh_A01G1050Uncharacterized protein0.991
Gh_D11G2795Uncharacterized protein0.990
Table 4. List of the phenylpropanoid pathway genes.
Table 4. List of the phenylpropanoid pathway genes.
Log2 (Fold Change G/W)AnnotationSymbol
12 DPA18 DPA24 DPA
Gh_A10G18350.749383.12883.1633Phenylalanine ammonia lyasePAL
Gh_A11G28910.276975.77916.3045PAL
Gh_D04G10780.181124.03665.2906PAL
Gh_D10G25280.768093.13113.7176PAL
Gh_D11G3277−0.00747716.02769.165PAL
Gh_A05G3997−0.499783.43558.11424-coumarate:CoA ligase4CL
Gh_A10G04563.84936.05064.02194CL
Gh_D05G3934−0.232082.44483.45084CL
Gh_D10G04731.54174.76045.04894CL
Gh_A13G20720.145361.62082.1007Coumarate 3-hydroxylaseC3H
Gh_A05G10050.51653.81583.4893Shikimate/quinate hydroxycinnamoyl transferaseHCT
Gh_D05G11230.556873.51512.4623HCT
Gh_Sca071998G010.73924.0523.1351HCT
Gh_D04G18184.37845.96123.5403Caffeoyl CoA O-methyltransfersaeCCoAOMT
Gh_D04G2028InfInfInfCCoAOMT
Gh_D07G00470.907544.24833.475Aldehyde dehydrogenaseALDH
Gh_D11G18052.57847.84031.8926Ferulate 5-hydroxylaseF5H
Gh_Sca170850G011.1641Inf1.5256F5H
Gh_A12G03671.57242.8194.867Chalcone and stilbene synthase family proteinCHS
Gh_D05G2280−1.4491.92415.4836CHS
Gh_D12G02991.26953.40814.445CHS
Gh_D09G19690.692490.867274.5918Flavanone 3-hydroxylaseF3H
Gh_A10G05000.401814.55261.7541Flavonoid 3′-hydroxylaseF3′H
Gh_A05G05572.50783.58651.8235Flavonoid 3′,5′-hydroxylaseF3′5′H
Gh_D04G19750.26368−3.0090.10962Flavonol synthaseFLS
Gh_D12G16860.305872.73143.453Leucoanthocyantin reducaseLAR
Inf represents that the gene is expressed in GCF, but no expression in WCF.

Share and Cite

MDPI and ACS Style

Sun, S.; Xiong, X.-p.; Zhu, Q.; Li, Y.-j.; Sun, J. Transcriptome Sequencing and Metabolome Analysis Reveal Genes Involved in Pigmentation of Green-Colored Cotton Fibers. Int. J. Mol. Sci. 2019, 20, 4838. https://doi.org/10.3390/ijms20194838

AMA Style

Sun S, Xiong X-p, Zhu Q, Li Y-j, Sun J. Transcriptome Sequencing and Metabolome Analysis Reveal Genes Involved in Pigmentation of Green-Colored Cotton Fibers. International Journal of Molecular Sciences. 2019; 20(19):4838. https://doi.org/10.3390/ijms20194838

Chicago/Turabian Style

Sun, Shichao, Xian-peng Xiong, Qianhao Zhu, Yan-jun Li, and Jie Sun. 2019. "Transcriptome Sequencing and Metabolome Analysis Reveal Genes Involved in Pigmentation of Green-Colored Cotton Fibers" International Journal of Molecular Sciences 20, no. 19: 4838. https://doi.org/10.3390/ijms20194838

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

Article Metrics

Back to TopTop