Next Article in Journal
Age-Dependent Changes in the Effects of Androgens on Female Metabolic and Body Weight Regulation Systems in Humans and Laboratory Animals
Next Article in Special Issue
Molecular Characterization and Pathogenicity of an Infectious cDNA Clone of Youcai Mosaic Virus on Solanum nigrum
Previous Article in Journal
Oxidative Stress in Xenograft Mouse Model Exposed to Dendrimers Decorated Polydopamine Nanoparticles and Targeted Chemo- and Photothermal Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Transcriptome and Metabolome Analyses Reveal That Jasmonic Acids May Facilitate the Infection of Cucumber Green Mottle Mosaic Virus in Bottle Gourd

Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(23), 16566; https://doi.org/10.3390/ijms242316566
Submission received: 24 October 2023 / Revised: 16 November 2023 / Accepted: 17 November 2023 / Published: 21 November 2023
(This article belongs to the Special Issue Advances in Plant Virus Diseases and Virus-Induced Resistance)

Abstract

:
Cucumber green mottle mosaic virus (CGMMV) is a typical seed-borne tobamovirus that mainly infects cucurbit crops. Due to the rapid growth of international trade, CGMMV has spread worldwide and become a significant threat to cucurbit industry. Despite various studies focusing on the interaction between CGMMV and host plants, the molecular mechanism of CGMMV infection is still unclear. In this study, we utilized transcriptome and metabolome analyses to investigate the antiviral response of bottle gourd (Lagenaria siceraria) under CGMMV stress. The transcriptome analysis revealed that in comparison to mock-inoculated bottle gourd, 1929 differently expressed genes (DEGs) were identified in CGMMV-inoculated bottle gourd. Among them, 1397 genes were upregulated while 532 genes were downregulated. KEGG pathway enrichment indicated that the DEGs were mainly involved in pathways including the metabolic pathway, the biosynthesis of secondary metabolites, plant hormone signal transduction, plant–pathogen interaction, and starch and sucrose metabolism. The metabolome result showed that there were 76 differentially accumulated metabolites (DAMs), of which 69 metabolites were up-accumulated, and 7 metabolites were down-accumulated. These DAMs were clustered into several pathways, including biosynthesis of secondary metabolites, tyrosine metabolism, flavonoid biosynthesis, carbon metabolism, and plant hormone signal transduction. Combining the transcriptome and metabolome results, the genes and metabolites involved in the jasmonic acid and its derivatives (JAs) synthesis pathway were significantly induced upon CGMMV infection. The silencing of the allene oxide synthase (AOS) gene, which is the key gene involved in JAs synthesis, reduced CGMMV accumulation. These findings suggest that JAs may facilitate CGMMV infection in bottle gourd.

1. Introduction

Cucumber green mottle mosaic virus (CGMMV) is a typical seed-borne plant virus belonging to the genus Tobamovirus. CGMMV is known to cause significant damage to cucurbit crops such as cucumber, watermelon, bottle gourd, squash, pumpkin, melons, and various gourd species [1,2,3,4,5,6]. Although the characteristic symptoms of CGMMV were first recorded in 1923 [7], it was not until 1935 that it was officially described in cucumber [8]. Since its initial discovery in England [8], CGMMV has rapidly spread globally due to the development of international trade [3,9,10,11,12,13,14,15,16,17]. In China, CGMMV was first detected in pumpkin in Guangxi province in 2005 [18], and has since caused widespread devastation to cucurbit crops across the country [19,20,21]. The marketable yield losses caused by CGMMV can be up to 50% and even 100% due to poor quality [12,21]. In 2006, CGMMV was listed as a national agricultural plant quarantine pest in China.
Phytohormones play a crucial role in plant development and response to abiotic and biotic stresses. Jasmonic acid (JA) and its derivatives (JAs) are important phytohormones that regulate various physiological processes, including plant growth, metabolism, and stress responses against pathogens and herbivores [22,23,24]. The active form of JA hormone, jasmonoyl-isoleucine (JA-Ile), is synthesized in the chloroplast from α-linoleic acid or hexadecatrienoic acid (16:3) and requires three reaction sites: chloroplast, peroxisome, and cytoplasm [25,26,27]. Various enzymes are involved in the chemical reactions of JAs, including allene oxide synthase (AOS), allene oxide cyclase (AOC), oxo-phytodienoic acid reductase (OPR3), jasmonate resistant 1 (JAR1), JA carboxyl methyltransferase (JMT), and JASSY protein [26,28]. Upon stimulation by biotic or abiotic factors, JA-Ile is quickly synthesized in the cytoplasm and transported to the nucleus by jasmonic transfer protein 1 (JAT1). In the nucleus, JA-Ile binds CORONATINE INSENSITIVE1 (COI1) and jasmonate ZIM-domain protein (JAZ), leading to ubiquitination and degradation by the 26S proteasome pathway. The degradation of JAZ releases the binding sites, which makes the transcription factors (TFs) activate the expression of JA-responsive genes [24].
JAs have also been reported to be involved in response to virus infection, but their function varies in response to different viruses [29,30,31,32,33]. The rice stripe virus coat protein (CP) induces the JAs signaling pathway, which further upregulates the expression of MYB TFs to active ARGONAUTE 18 (AGO18)-mediated RNA silencing and antiviral defense in rice [31,34]. The C2 protein encoded by tomato yellow leaf curl Sardinia virus (TYLCSV) affects the activity of the Skp1/Cullin/F-box (SCF) complex by interacting with the CSN5 protein, which further inhibits JAs signaling in Arabidopsis thaliana [35]. However, exogenously applied JA reduces the local resistance of N gene-containing tobacco to tobacco mosaic virus (TMV) and silencing of COI1, or AOS reduces TMV accumulation, demonstrating that JA negatively regulates the resistance to tobacco mosaic virus in tobacco [36]. But, the function of the JAs signaling pathway under CGMMV stress is still unclear.
CGMMV mainly infects cucurbit leaves, fruits, and seeds, causing mottling and mosaic symptoms on the leaves and fruit peel, brown necrotic lesions on stems and peduncles, and yellowing and spongy fruit flesh [10]. Previous studies using high-throughput deep sequencing have demonstrated that the infection of CGMMV in watermelon affects the expression of miRNAs and genes involved in cell wall modulation, the plant hormone signaling pathway, photosynthesis, primary and secondary metabolism, and intracellular transport [37,38]. An analysis of CGMMV-derived siRNAs indicated that different cucurbit species respond differently to CGMMV infection [39,40,41]. Transcriptome analysis of watermelon leaves and fruit under CGMMV stress revealed that DEGs are involved in photosynthesis, plant–pathogen interactions, secondary metabolism, and plant hormone signal transduction [38,42]. However, there has not been a study on the interaction between bottle gourd and CGMMV.
Previously, we found that CGMMV has been one of the main viruses threatening bottle gourd in Guangdong province, China [43]. The infectious cDNA clone of CGMMV was constructed and the infectivity in different cucurbit crops was analyzed [44]. In this study, we used the transcriptome and metabolome to analyze the antiviral response of bottle gourd with CGMMV infection. Then, we focused on the hormone pathway, especially the JAs signaling pathway.

2. Results

2.1. Inoculation and Virus Detection of CGMMV in Bottle Gourd

To inoculate bottle gourd plants, the cotyledons were infiltrated with Agrobacterium containing CGMMV infectious cDNA clones [44], while the mock plants were infiltrated with Agrobacterium containing an empty vector. At 12 days post inoculation (dpi), CGMMV infected plants developed obvious mosaic and mottling symptoms on the upper leaves compared to the mock plants (Figure 1A). The leaves of both the CGMMV-infected and mock plants were taken and subjected to RT-PCR and Western blot detection. RT-PCR using the CGMMV coat protein (CP) primer pair (Table S1) showed the expected band, and Western blot with CGMMV CP antibody also revealed a specific band of the expected size (Figure 1B).

2.2. Quality Control of RNA-Seq Data

To perform transcriptome and metabolome tests, the upper leaves of CGMMV-infected (ZLV12) and mock plants (ZLM12) were taken at 12 dpi, and three replicates were conducted in each treatment. The total RNA of all samples was extracted and subjected to deep sequencing using the Illumina HiSeq platform. The deep sequencing generated 298,190,450 raw reads and 291,823,170 (97.86%) clean reads after the removal of low-quality reads, contamination, and adapter sequences (Table 1). The clean reads encompassed about 43.76 Gb clean data, which were sufficient for gene expression analysis. Then, the clean data were mapped to the reference genome of bottle gourd (http://cucurbitgenomics.org/ftp/genome/BottleGourd/USVL1VR-Ls/) (accessed on 5 June 2022) using HISAT2 software (v2.1.0). The results showed that the unique mapped percentage of each replicate was over 92%, the Q20 percentage was over 97%, and the Q30 percentage was over 92%. The GC content of each replicate was over 44% (Table 1).

2.3. Gene Expression Analysis of Bottle Gourd in Response to CGMMV Infection

To identify the genes involved in response to CGMMV infection in bottle gourd, DEGs were analyzed using RNA-seq data. Different gene expression analyses between the mock and CGMMV treatments were performed using DESeq2 software (v1.22.2), which requires data of unnormalized reads. The genes with a false discovery rate (FDR) < 0.05 and |log2Fold Change| ≥ 1 were filtered as significant DEGs. A total of 1929 DEGs were identified between the mock and CGMMV treatments: 1397 DEGs were upregulated and 532 were downregulated (Figure 2A). The hierarchical clustering of DEGs in Figure 2B shows an overview of the transcriptome result.
To cluster these DEGs, Gene Ontology (GO) pathway enrichment was performed. In the biological process of GO enrichment, the pathways related to response to drugs, regulation of hormone levels, response to ethylene, hormone metabolic process, innate immune response, and response to oxidative stress were the most enriched (Supplementary Figure S1). The intrinsic component of plasma membrane was the only enriched pathway in the cellular component. In terms of the molecular function of GO, the significantly enriched pathways included tetrapyrrole binding and heme binding (Supplementary Figure S1).
Based on the GO analysis, the Kyoto Encyclopedia of Genes and Genomes (KEGG, https://www.genome.jp/kegg) (accessed on 5 June 2022) was used to further cluster the DEG pathways. Among the KEGG enrichment pathways, the metabolic pathway, the biosynthesis of secondary metabolites, plant hormone signal transduction, plant–pathogen interaction, and starch and sucrose metabolism were the most enriched (Figure 2C).

2.4. Real-Time RT-PCR Verification of Transcriptome Result

To verify the RNA-seq analysis, 20 DEGs were selected to analyze the expressions after virus infection. The thirteen upregulated DEGs were Lsi01G013470, Lsi02G017750, Lsi05G011760, Lsi04G015060, Lsi02G007470, Lsi02G018990, Lsi01G009350, Lsi02G021080, Lsi02G008430, Lsi11G012260, Lsi01G014560, Lsi11G005040, and Lsi05G018940. The seven downregulated DEGs were Lsi05G012660, Lsi04G002080, Lsi03G008210, Lsi09G006560, Lsi02G013460, Lsi11G000250, and Lsi03G014380. The annotation of these DEGs (Table 2) was acquired by searching the bottle gourd genome website (http://www.cucurbitgenomics.org/search/genome/13) (accessed on 5 June 2022) using these accession numbers. The qRT-PCR results showed that all of these selected DEGs exhibited a similar trend to the transcriptome analysis, suggesting that the transcriptome analysis was convincing (Figure 3).

2.5. Metabolite Accumulation Analysis of Bottle Gourd in Response to CGMMV Infection

To investigate the changes in metabolite accumulation under CGMMV stress, metabolome analysis was conducted between the mock and CGMMV-treated samples. The metabolites with |log2Fold Change| ≥ 1 were filtered as significant DAMs. A total of 76 DAMs were identified between the mock and CGMMV groups: 69 were significantly upregulated and 7 were significantly downregulated (Figure 4A). The most upregulated DAMs were 4-caffeoylquinic acid, acteoside, and malvidin-3,5-O-diglucoside (Table S2). The most downregulated DAMs were glucarate O-phosphoric acid, cyclic AMP, and scopoletin (7-Hydroxy-5-methoxycoumarin) (Table S2). The clustering heatmap of DAMs is shown in Figure 4B. To cluster these DAMs, KEGG pathway enrichment was performed based on the function of each DAM. KEGG showed that the biosynthesis of secondary metabolites, tyrosine metabolism, flavonoid biosynthesis, and the plant hormone signal transduction pathways were the most enriched (Figure 4C).

2.6. Joint Analysis of Transcriptome and Metabolome of Bottle Gourd in Response to CGMMV Infection

Based on the transcriptome and metabolome results, combined analysis was performed via KEGG enrichment. KEGG enrichment showed that plant hormone signal transduction, isoflavonoid biosynthesis, the biosynthesis of secondary metabolites, and alpha-linolenic acid metabolism were the most enriched pathways (Supplementary Figure S2).
Among these enriched pathways, the plant hormone signal transduction pathway was significantly enriched, as seen in transcriptome and metabolome analysis. The plant hormones consist of auxin, cytokinine, gibberellin, abscisic acid, ethylene, brassinosteroid, jasmonic acid, and salicylic acid. In the auxin signaling pathway, auxin response factor 5 (ARF5) (Lsi04G017260) was upregulated about two times more in CGMMV-infected plants compared with the mock plants. However, the auxin hormone showed no significant change upon CGMMV infection. In the brassinosteroid biosynthesis pathway, the expression of brassinosteroid insensitive 1 (BRI1) (Lsi11G000250) decreased by nearly 70% with CGMMV infection, while BRI1 kinase inhibitor 1 (BKI1) (Lsi08G014580), brassinosteroid insensitive 2 (BIN2) (Lsi01G002340), and brassinosteroid resistant 1/2 (BZR1/2) (Lsi07G002850) were induced about three times, two times, and two times, respectively (Figure 5). In addition, the expression of NPR1 and PR1 in the salicylic acid pathway showed no obvious response to CGMMV infection (Figure 5).
In summary, joint analysis of DEGs and DAMs showed that the jasmonic acid and brassinosteroid signal transduction pathways were remarkably induced, while changes in other hormone signaling pathways were not obvious.

2.7. Analysis of JAs signaling Pathway in Response to CGMMV Infection

Among the plant hormone signal transduction pathways, the stimulation of the JAs synthesis pathway is the most prominent. The genes involved in the JAs synthesis pathways include lipoxygenase (LOX), AOS, AOC, OPR3, JAR1, JMT, CYP94B3, and CYP94C1 (Figure 6A). qRT-PCR analysis showed that all these genes were upregulated upon CGMMV infection (Figure 6B). Moreover, the accumulation of JA and JA-Ile hormones was also remarkably increased (Figure 6C), demonstrating that the JAs signaling pathway was induced upon CGMMV infection.
In the JAs signaling pathway, JA-Ile will be transported to the nucleus from the cytoplasm to release the TFs to activate the expression of defensive genes. To identify the expression of TFs related with the JAs signaling pathway under CGMMV stress, the transcriptome data of TFs (Table S3) were screened and analyzed. To validate the RNA-seq results, we selected various types of TFs that may be involved in the JAs signaling pathway and confirmed their expression levels using qRT-PCR. As expected, the transcription factor Myc2 (Lsi09G016530), which can interact with JAZ protein, was induced about two times upon CGMMV infection, while another JAZ interacting transcription factor, Myc3 (Lsi07G011090), was upregulated more than 30 times (Figure 7A). TFs Myb13 (Lsi05G010160), Myb62 (Lsi02G001830), Myb77 (Lsi04G015930), and Myb86 (Lsi06G015850) were also induced via CGMMV stimulation, but the expression of Myb48 (Lsi10G013470) was reduced (Figure 7B). The TF NAC (Lsi08G004490) was also induced about nine times after CGMMV infection (Figure 7C). WRKY TFs play an important role in plant development, senescence, and response to abiotic and biotic stimuli. Compared with the mock control, the expression of WRKY18 (Lsi02G012150) was slightly higher, WRKY31 (Lsi05G014110) increased twice, and WRKY68 (Lsi06G014830) increased nearly tenfold, but WRKY6 (Lsi05G021100) decreased about twice (Figure 7D). ERF TFs are also modulated by the JAs signaling pathway. ERF-RAP2 (Lsi08G002460) increased nearly one thousand times, ERF1b (Lsi09G012390) increased more than 30 times, ERF1 (Lsi06G008160) and ERF2 (Lsi07G001170) increased about 6–7 times, and ERF2b (Lsi03G010920) increased about two times, but ERF6 (Lsi04G020490) decreased to about one third (Figure 7E).
Taken together, these results demonstrate that CGMMV infection activates the JAs signaling pathway and further induces the expression of TFs to initiate downstream gene expression.

2.8. The Function of the JAs Signaling Pathway in CGMMV Infection

To further identify the function of the JAs signaling pathway during CGMMV infection, we used CGMMV-based gene silencing as described previously [45]. Agrobacterium containing CGMMV-gfp, CGMMV-BgAOS, or CGMMV-Bgpds was infiltrated into the cotyledons of bottle gourd. At 12 dpi, plants inoculated with CGMMV-Bgpds showed obvious photobleaching phenotypes, indicating that pds was silenced by CGMMV-induced gene silencing (Figure 8A). Plants inoculated with CGMMV-gfp showed mottle and green symptoms, while CGMMV-BgAOS showed no obvious disease symptom (Figure 8A). Samples were taken and performed using Western blot and qRT-PCR detection. Compared with CGMMV-gfp, CP accumulation was substantially reduced in CGMMV-BgAOS (Figure 8B). qRT-PCR also showed that the mRNA level of the AOS gene was significantly reduced compared with CGMMV-gfp, though not much compared with the mock control (Figure 8C). These results indicated that the downregulation of AOS mRNA led to a decrease in CGMMV accumulation, implying that JAs may contribute to CGMMV infection in bottle gourd.

3. Discussion

CGMMV mainly infects cucurbit crops, causing serious damage to the cucurbit industry. Although the symptoms of CGMMV vary between different cucurbit species and cultivars, the classic symptoms are leaf mottling, green, mosaic, and fruit malformation. In a previous study, we found that CGMMV was one of the most prevalent viruses in bottle gourd in Guangdong province of China, causing serious damage to the bottle gourd industry [43]. High-throughput deep sequencing has been extensively used to investigate the interaction between biotic or abiotic stimuli and plants, facilitating the identification of host gene expressions using distinct treatments. Metabolome analysis is another potent methodology for examining changes in metabolites, which are regulated by gene expression and play a crucial role in plant growth, development, differentiation, and defense. Several studies have utilized RNA-seq or whole-genome bisulfite sequencing to identify the genes’ expressions or the DNA methylation level of watermelon leaves or fruit [38,42,46]. However, combined transcriptome and metabolome analysis of bottle gourd with CGMMV infection has not been reported. In this study, transcriptome and metabolome analysis revealed that DEGs and DAMs are mainly involved in pathways related to the biosynthesis of secondary metabolites and plant hormone signal transduction. The JAs signaling transduction pathway was found to be significantly activated, and silencing of the AOS gene, the key enzyme in the JAs synthesis pathway, decreased CGMMV accumulation, implying that JAs may play a role in facilitating CGMMV infection.
In our study, 1929 DEGs were found via transcriptome analysis: 1397 DEGs were upregulated and 532 were downregulated. GO and KEGG pathway enrichment showed that these DEGs are mainly involved in metabolic pathways, the biosynthesis of secondary metabolites, plant hormone signal transduction, and plant–pathogen interaction pathways. A previous study on the impact of CGMMV infection on watermelon leaves and fruit also demonstrated that the DEGs were involved in photosynthesis, plant–pathogen interactions, secondary metabolism, and plant hormone signal transduction [38,42]. These studies uncovered that the targeting of these pathways by CGMMV could potentially serve as the underlying cause of disease symptoms in cucurbit crops.
We found that CGMMV activates hormone signal transduction pathways, like JAs, ethylene, and brassinosteroid, while the response of the salicylic acid pathway was not obvious. JAs are phytohormones playing an important role in plant growth and development. JA and its derivates are synthesized in chloroplast, peroxisome, and cytoplasm. Transcriptome analysis showed that all of the genes involved in the JAs synthesis pathway were upregulated, and the accumulation of JA and the bioactive JA-Ile hormones was increased, demonstrating that the JAs signaling pathway was activated with CGMMV infection. After being transported to the nucleus, JAs activate the degradation of JAZ protein via the 26S proteasome pathway. A previous report showed that the cucumber mosaic virus (CMV) 2b protein interacts and represses the JAZ protein to manipulate JAs hormone signaling to attract insect vectors [32]. In this study, we found that the expression of JAZ was increased about three times in CGMMV-infected bottle gourd, and further experiments are needed to find out the reason.
The degradation of JAZ protein releases transcription factors (TFs) and initiates downstream gene expression. TFs involved in the JAs signaling pathway include Mycs, Mybs, NACs, WRKYs, and ERFs. Here, we have identified an increase in the expression of most TFs related to the JAs signaling pathway upon CGMMV infection. The expression of basic helix-loop-helix TF Myc2 and Myc3 increased about 2 times and 32 times, responding to CGMMV attack. Reports have shown that both Myc2 and Myc3 are the targets of the JAZ protein and regulate the expression of various subsets of JA-responsive genes [47,48,49]. Furthermore, we showed that some Myb TFs were also induced after CGMMV infection, like Myb13 (more than 4 times), Myb62 (more than 30 times), and Myb86 (10 times). Myb13 has been reported to be a transcriptional activator and enhances the expression of fructosyltransferase to synthesize fructans in wheat [50]. Transgenic overexpression of Myb48 improved the drought tolerance of Arabidopsis plants, indicating that Myb48 may be involved in the drought stress response [51]. Myb62 regulates the phosphate starvation response by affecting gibberellic acid (GA) biosynthesis [52]. Myb77 interacts with auxin response factors (ARFs) and is involved in the auxin response [53]. In wheat, the expression of Myb86 was induced by various hormones and cold treatments [54]. NAC TF was induced about nine times, but the function of NAC TF is largely unknown.
WRKY TFs represent another large TF family which consists of 89 members in Arabidopsis. Some WRKY TFs are regulated by the JAs signaling pathway, like WRKY22, WRKY50, WRKY57, WRKY70, and WRKY89 [55,56,57,58,59]. A previous study indicated that WRKY3 and WRKY6 are elicited upon herbivore feeding, and silencing WRKY3 or WRKY6 decreases the resistance of Nicotiana attenuate in herbivores [60]. However, in this study, we found that the expression of WRKY6 was reduced, demonstrating that WRKY6 may play a varied role in different plants under diverse stresses. Various studies have reported that WRKY18 was induced upon abiotic or biotic stress [61,62,63,64], and we also confirmed that CGMMV also facilitated WRKY18 expression. Moreover, we found that the expression of WRKY31 was upregulated with CGMMV infection. In apple, WRKY31 was induced after SA treatment, and the ectopic expression of WRKY31 increased the resistance of Arabidopsis and Nicotiana benthamiana to flg22 and Pseudomonas syringae tomato (Pst DC3000) [65]. Further, WRKY68 was induced up to 10 times with CGMMV infection, implying that WRKY68 may play an important role during viral infection. However, the function of WRKY68 under abiotic and biotic stress remains unclear.
Some ERF TFs are also induced via JAs signaling. We found that the expression levels of ERF1, ERF1b, ERF-AP2, ERF-2, and ERF-2b were significantly increased, indicating that these TFs may be involved in the antiviral response. ERF1 was identified as a member of the AP2 transcription factor family and was found to function dependently on JAs and/or ET during Botrytis cinerea infection [66]. We found that ERF-RAP2 was induced to nearly 1000 times upon CGMMV infection, indicating the importance of ERF-RAP2 during viral infection. A previous report has shown that RAP2 increases the resistance of Arabidopsis to Pseudomonas syringae [67]. ERF2 was also induced upon CGMMV infection, though not as much as ERF-RAP2. Studies showed that ERF2 was significantly upregulated in tomato plants infected with Stemphylium lycopersici, and SA and JA accumulation were lower in ERF2-silenced plants [68]. However, the expression of ERF6 was downregulated under CGMMV stress. On the contrary, a previous report showed that JA/ET-responsive genes were upregulated in ERF6 transgenic Arabidopsis plants, indicating that ERF6 may positively regulate the JAs signaling pathway [69]. Taken together, although a variety of reports have studied the functions of some TFs, the specific function of these TFs during CGMMV needs further research.
Phytohormones play an important role in the defense response to most biotic and abiotic stimuli, but viruses have evolved varied strategies to avoid host immunity. Although numerous studies have reported the interactions between plants and viruses, the role of phytohormones is still obscure. JAs signaling may play a protective role against plant viruses, like beet curly top virus (BCTV) and CMV [32,35]. In this study, we found that the silencing of AOS reduced CGMMV accumulation, implying that JAs signaling may play a negative role in the defense response of bottle gourd to CGMMV. Consistently, JAs also contribute to TMV infection in N gene-containing tobacco [36]. Further research may focus on the mechanism behind this, like the interactions between viral proteins and JAs signaling pathway components.

4. Materials and Methods

4.1. Plant Growth Conditions

Bottle gourd (Lagenaria siceraria cv. Pugua no. 1) plants were grown in a climate chamber with a 14 h/10 h light/dark photoperiod at 25 °C.

4.2. Virus Inoculation and Detection

The cotyledons of bottle gourd were infiltrated with Agrobacterium GV3101 containing pCB301-CGMMV infectious cDNA clone [44]. At 12 dpi, the upper leaves were taken and subjected to Western blot or RT-PCR with CGMMV CP antibody or CGMMV CP specific primer (Table S1).

4.3. Total RNA Extraction, cDNA Library Construction, and Deep Sequencing

At 12 dpi, approximately 0.1 g of the upper leaves from the mock plants and the CGMMV-infected bottle gourd, with three replicates each, was collected for RNA deep sequencing. Total RNA of the mock plants and CGMMV-infected bottle gourd was extracted using Trizol Reagent (Takara, Dalian, China). DNA contamination was digested using RNase-free rDNase (Transgene, Beijing, China). RNA concentration and integrity were evaluated using a Qubit 2.0 fluorescence spectrometer (Thermo Fisher, Waltham, MA, USA) and Agilent 2100 bioanalyzer (Agilent, Santa Clara, CA, USA). Then, the mRNA of the total RNA was enriched using Oligo dT magnetic beads before the construction of the cDNA library according to the manufacturer’s instructions (Illumina, San Diego, CA, USA). mRNA was fragmented by adding fragmentation buffer; then, the fragmentated mRNAs were used as templates for the synthesis of first-strand cDNA with random hexamers. Double-stranded cDNA was synthesized by adding reaction buffer, dNTPs, and DNA polymerase, followed by purifying with AMPure XP beads (Beckman Coulter, Brea, CA, USA). The purified double-stranded cDNA was end-repaired, and we added an “A” base and sequencing index adapter. The treated double-stranded cDNA was enriched with AMPure XP beads and formed the cDNA library. Deep sequencing was performed using the Illumina HiSeq 2000 platform (Illumina, USA) by Wuhan Metware Biotechnology Co., Ltd. (www.metware.cn, Wuhan, China).

4.4. Transcriptome Analysis

Raw data generated by deep sequencing were treated by removing the read adapter sequences and low-quality reads using Fastp [70]. Clean data were mapped to the bottle gourd genome (http://cucurbitgenomics.org/ftp/genome/BottleGourd/USVL1VR-Ls/) (accessed on 5 June 2022) using hierarchical indexing for spliced alignment of transcripts (HISAT) [71]. Stringtie [72] was used to reconstruct the transcriptome scripts and estimate the expression levels of each mRNA in different samples. Before comparing DEGs between different samples, transcripts were normalized by calculating fragments per kilobase of exon per million mapped reads (FPKM) using RSEM software (v.1.3.1) [73]. DESeq2 [74] was used to analyze the DEGs, with a false discovery rate (FDR) < 0.05 and |log2FC| ≥ 1. GO (http://www.geneontology.org/) (accessed on 5 June 2022) and KEGG (https://www.genome.jp/kegg) (accessed on 5 June 2022) pathway enrichment was used to analysis the DEGs.

4.5. RNA-Seq Validation Using qRT-PCR

To validate the transcriptome result, the total RNA of the mock and CGMMV-treated bottle gourd was extracted and the genomic DNA contamination was digested using RNase-free rDNase before reverse transcription using the PrimeScript™ RT reagent Kit with a gDNA Eraser (Takara, Dalian, China). Twenty DEGs were selected for qRT-PCR detection after the analysis of the transcriptome results. The primers used for real-time PCR of the 20 DEGs, the genes involved in JAs synthesis, and the TFs are listed in Table S1, and the Histone H3 gene of Lagenaria siceraria (LsH3) [75] was used as the reference gene. Real-time PCR was performed with TB green Premix Ex Taq (Takara, Japan) using the CFX96 Real-Time System (Bio-Rad, Hercules, CA, USA).

4.6. Metabolome Analysis

Sample preparation, extraction, and metabolite quantification were performed according to the protocol published previously [76] by Wuhan Metware Biotechnology Co., Ltd. Metabolite profiling was conducted using ultra-performance liquid chromatography (UPLC, Shim-pack UFLC Shimadzu CBM30A) and tandem mass spectrometry (MS) (Applied Biosystems 4500 QTRAP). Qualitative analysis of metabolites was performed based on the metware database, and quantitative analysis of metabolites was performed via multiple reaction monitoring (MRM) and triple quadrupole mass spectrometry [77,78]. Analyst 1.6.3 was used to analyze the MS data and |log2Fold Change| ≥ 1 were filtered as significant DAMs. The heatmap was drawn using R software (v.4.1.0).

4.7. CGMMV-Based Gene Silencing

The CGMMV infectious cDNA clone was constructed previously [44]. To construct the CGMMV-based gene silencing system, plasmid containing CGMMV infectious cDNA clone was modified, as reported previously [45]. Briefly, the full length of CGMMV was inserted into the pCB301 vector to make a pCB301-CGMMV infectious clone. Then, the sequence of CP promoter was repeated to start the transcription of downstream gene expression in the intergenic region of CP and MP ORF. The phytoene desaturase (PDS) is an essential enzyme during the plant carotenoid biosynthetic pathway, and silencing of PDS results in photobleaching phenotypes in Nicotiana benthamiana and cucurbit crops. Fragments of AOS and PDS of 300 bp in length were amplified from bottle gourd cDNA and inserted into the BamHI-digested CGMMV-infectious cDNA clone to make CGMMV-BgAOS and CGMMV-Bgpds. CGMMV-gfp was used as a positive control.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms242316566/s1.

Author Contributions

Z.L. and Z.H. conceived and designed the experiments. Z.L., Y.T. and L.Y. conducted the experiments. X.S., G.L. and S.D. analyzed the data. Z.L. and Z.H. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (32272509), the Project of Collaborative Innovation Center of GDAAS (XT202210), the Agricultural Competitive Industry Discipline Team Building Project of Guangdong Academy of Agricultural Sciences (202103TD and 202105TD), the Science and Technology Program of Guangzhou (SL2023A04J00751), the Young Talent Support Project of Guangzhou Association for Science and Technology (QT-2023-040), and the Special Fund for Scientific Innovation Strategy—Construction of High-Level Academy of Agriculture Science (R2023PY-JX011).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of Plant Protection Research Institute, Guangdong Academy of Agricultural Sciences.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article and supplementary.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Komuro, Y. Cucumber green mottle mosaic virus on cucumber and watermelon and melon necrotic spot virus on muskmelon. Jarq-Jpn. Agric. Res. Q. 1971, 6, 41–45. [Google Scholar]
  2. Antignus, Y.; Pearlsman, M.; Ben-Yoseph, R.; Cohen, S. Occurrence of a variant of cucumber green mottle mosaic virus in Israel. Phytoparasitica 1990, 18, 50–56. [Google Scholar] [CrossRef]
  3. Al-Shahwan, I.M.; Abdalla, O.A. A strain of cucumber green mottle mosaic virus (CGMMV) from bottle gourd in Saudi Arabia. J. Phytopathol. 1992, 134, 152–156. [Google Scholar] [CrossRef]
  4. Nagendran, K.; Aravintharaj, R.; Mohankumar, S.; Manoranjitham, S.K.; Naidu, R.A.; Karthikeyan, G. First report of cucumber green mottle mosaic virus in snake gourd (Trichosanthes cucumerina) in India. Plant Dis. 2015, 99, 559. [Google Scholar] [CrossRef]
  5. Sugiyama, M.; Ohara, T.; Sakata, Y. A new source of resistance to cucumber green mottle mosaic virus in melon. J. Jpn. Soc. Hortic. Sci. 2006, 75, 469–475. [Google Scholar] [CrossRef]
  6. Wang, S.M.; Chen, M.J. A new strain of cucumber mottle mosaic virus causing mosaic symptoms on bottlegourd in Taiwan. Plant Prot. Bull. 1985, 27, 105–110. [Google Scholar]
  7. Bewley, W.F. Diseases of Glasshouse Plants; E. Benn: London, UK, 1923. [Google Scholar]
  8. Ainsworth, G.C. Mosaic diseases of the cucumber. Ann. Appl. Biol. 1935, 22, 55–67. [Google Scholar] [CrossRef]
  9. Ali, A.; Hussain, A.; Ahmad, M. Occurrence and molecular characterization of cucumber green mottle mosaic virus in cucurbit crops of KPK, Pakistan. Braz. J. Microbiol. 2014, 45, 1247–1253. [Google Scholar] [CrossRef]
  10. Dombrovsky, A.; Tran-Nguyen, L.T.T.; Jones, R.A.C. Cucumber green mottle mosaic virus: Rapidly increasing global distribution, etiology, epidemiology, and management. Annu. Rev. Phytopathol. 2017, 55, 231–256. [Google Scholar] [CrossRef]
  11. Li, R.; Zheng, Y.; Fei, Z.; Ling, K.S. First complete genome sequence of an emerging cucumber green mottle mosaic virus isolate in North America. Genome Announc. 2015, 3, e00452-15. [Google Scholar] [CrossRef]
  12. Reingold, V.; Lachman, O.; Koren, A.; Dombrovsky, A. First report of cucumber green mottle mosaic virus (CGMMV) symptoms in watermelon used for the discrimination of non-marketable fruits in Israeli commercial fields. New Dis. Rep. 2013, 28, 11. [Google Scholar] [CrossRef]
  13. Rudnieva, T.O.; Budzanivs’ka, I.H.; Ryzhkova, A.; Shevchenko, T.P.; Dem’ianenko, F.P.; Polishchuk, V.P. Characteristics of green mottle mosaic virus isolates in cucumbers from different regions of Ukraine. Mikrobiol. Zhurnal 2005, 67, 96–103. [Google Scholar]
  14. Slavokhotova, A.A.; Andreeva, E.N.; Shiian, A.N.; Odintsova, T.I.; Pukhal’skii, V.A. Specifics of the coat protein gene in Russian strains of the cucumber green mottle mosaic virus. Genetika 2007, 43, 1461–1467. [Google Scholar] [CrossRef]
  15. Kehoe, M.A.; Webster, C.; Wang, C.; Jones, R.A.C.; Coutts, B.A. Occurrence of cucumber green mottle mosaic virus in Western Australia. Australas. Plant Pathol. 2022, 51, 1–8. [Google Scholar] [CrossRef]
  16. Ling, K.S.; Li, R.; Zhang, W. First report of cucumber green mottle mosaic virus infecting greenhouse cucumber in Canada. Plant Dis. 2014, 98, 701. [Google Scholar] [CrossRef]
  17. Tian, T.; Posis, K.; Maroon-Lango, C.J.; Mavrodieva, V.; Haymes, S.; Pitman, T.L.; Falk, B.W. First report of cucumber green mottle mosaic virus on melon in the United States. Plant Dis. 2014, 98, 1163. [Google Scholar] [CrossRef]
  18. Qin, B.X.; Cai, J.H.; Liu, Z.M.; Chen, Y.H.; Zhu, G.N.; Huang, F.X. Preliminary identification of a cucumber green mottle mosaic virus infecting pumpkin. Plant Quar. 2005, 19, 198–200. [Google Scholar]
  19. Wu, Y.H.; Li, L.M.; Zhao, X.X.; Wang, W.H.; Wang, L.; Cai, M. Pest risk analysis of invasion and spreading of cucumber green mottle mosaic virus in China. Plant Prot. 2010, 36, 33–36. [Google Scholar]
  20. Tian, Y.L.; Liu, D.M.; Zhang, Y.J.; Li, M.F.; Ma, Z.H. Bioassay and genomic studies on the two isolates of cucumber green mottle mosaic virus from Beijing and Shandong. Plant Quar. 2009, 23, 1–6. [Google Scholar]
  21. Chen, H.Y.; Zhao, W.J.; Cheng, Y.; Li, M.F.; Zhu, S.F. Molecular identification of the virus causing watermelon mosaic disease in Mid-Liaoning. Zhi Wu Bing Li Xue Bao 2006, 36, 306–309. [Google Scholar]
  22. Zhang, L.; Zhang, F.; Melotto, M.; Yao, J.; He, S.Y. Jasmonate signaling and manipulation by pathogens and insects. J. Exp. Bot. 2017, 68, 1371–1385. [Google Scholar] [CrossRef]
  23. Baldwin, I.T. An ecologically motivated analysis of plant-herbivore interactions in native tobacco. Plant Physiol. 2001, 127, 1449–1458. [Google Scholar] [CrossRef]
  24. Ghorbel, M.; Brini, F.; Sharma, A.; Landi, M. Role of jasmonic acid in plants: The molecular point of view. Plant Cell Rep. 2021, 40, 1471–1494. [Google Scholar] [CrossRef]
  25. Chini, A.; Monte, I.; Zamarreño, A.M.; Hamberg, M.; Lassueur, S.; Reymond, P.; Weiss, S.; Stintzi, A.; Schaller, A.; Porzel, A.; et al. An OPR3-independent pathway uses 4,5-didehydrojasmonate for jasmonate synthesis. Nat. Chem. Biol. 2018, 14, 171–178. [Google Scholar] [CrossRef]
  26. Ruan, J.; Zhou, Y.; Zhou, M.; Yan, J.; Khurshid, M.; Weng, W.; Cheng, J.; Zhang, K. Jasmonic acid signaling pathway in plants. Int. J. Mol. Sci. 2019, 20, 2479. [Google Scholar] [CrossRef]
  27. Wasternack, C.; Feussner, I. The oxylipin pathways: Biochemistry and function. Annu. Rev. Plant Biol. 2018, 69, 363–386. [Google Scholar] [CrossRef]
  28. Guan, L.; Denkert, N.; Eisa, A.; Lehmann, M.; Sjuts, I.; Weiberg, A.; Soll, J.; Meinecke, M.; Schwenkert, S. JASSY, a chloroplast outer membrane protein required for jasmonate biosynthesis. Proc. Natl. Acad. Sci. USA 2019, 116, 10568–10575. [Google Scholar] [CrossRef]
  29. Collum, T.D.; Culver, J.N. The impact of phytohormones on virus infection and disease. Curr. Opin. Virol. 2016, 17, 25–31. [Google Scholar] [CrossRef]
  30. Alazem, M.; Lin, N.-S. Roles of plant hormones in the regulation of host–virus interactions. Mol. Plant Pathol. 2015, 16, 529–540. [Google Scholar] [CrossRef]
  31. Yang, Z.; Huang, Y.; Yang, J.; Yao, S.; Zhao, K.; Wang, D.; Qin, Q.; Bian, Z.; Li, Y.; Lan, Y.; et al. Jasmonate signaling enhances RNA silencing and antiviral defense in rice. Cell Host Microbe 2020, 28, 89–103 e108. [Google Scholar] [CrossRef]
  32. Wu, D.; Qi, T.; Li, W.X.; Tian, H.; Gao, H.; Wang, J.; Ge, J.; Yao, R.; Ren, C.; Wang, X.B.; et al. Viral effector protein manipulates host hormone signaling to attract insect vectors. Cell Res. 2017, 27, 402–415. [Google Scholar] [CrossRef]
  33. Zhao, S.; Li, Y. Current understanding of the interplays between host hormones and plant viral infections. PLoS Pathog. 2021, 17, e1009242. [Google Scholar] [CrossRef]
  34. Wu, J.; Yang, Z.; Wang, Y.; Zheng, L.; Ye, R.; Ji, Y.; Zhao, S.; Ji, S.; Liu, R.; Xu, L.; et al. Viral-inducible argonaute18 confers broad-spectrum virus resistance in rice by sequestering a host microRNA. Elife 2015, 4, e05733. [Google Scholar] [CrossRef]
  35. Lozano-Duran, R.; Rosas-Diaz, T.; Gusmaroli, G.; Luna, A.P.; Taconnat, L.; Deng, X.W.; Bejarano, E.R. Geminiviruses subvert ubiquitination by altering CSN-mediated derubylation of SCF E3 ligase complexes and inhibit jasmonate signaling in Arabidopsis thaliana. Plant Cell 2011, 23, 1014–1032. [Google Scholar] [CrossRef]
  36. Oka, K.; Kobayashi, M.; Mitsuhara, I.; Seo, S. Jasmonic acid negatively regulates resistance to tobacco mosaic virus in tobacco. Plant Cell Physiol. 2013, 54, 1999–2010. [Google Scholar] [CrossRef]
  37. Sun, Y.; Niu, X.; Fan, M. Genome-wide identification of cucumber green mottle mosaic virus-responsive microRNAs in watermelon. Arch. Virol. 2017, 162, 2591–2602. [Google Scholar] [CrossRef]
  38. Li, X.; An, M.; Xia, Z.; Bai, X.; Wu, Y. Transcriptome analysis of watermelon (Citrullus lanatus) fruits in response to cucumber green mottle mosaic virus (CGMMV) infection. Sci. Rep. 2017, 7, 16747. [Google Scholar] [CrossRef]
  39. Li, Y.; Deng, C.; Shang, Q.; Zhao, X.; Liu, X.; Zhou, Q. Characterization of siRNAs derived from cucumber green mottle mosaic virus in infected cucumber plants. Arch. Virol. 2016, 161, 455–458. [Google Scholar] [CrossRef]
  40. Li, J.; Zheng, H.; Zhang, C.; Han, K.; Wang, S.; Peng, J.; Lu, Y.; Zhao, J.; Xu, P.; Wu, X.; et al. Different virus-derived siRNAs profiles between leaves and fruits in cucumber green mottle mosaic virus-infected Lagenaria siceraria plants. Front. Microbiol. 2016, 7, 1797. [Google Scholar] [CrossRef]
  41. Sun, Y.; Niu, X.; Cui, D.; Fan, M. High-throughput sequencing reveals vsiRNAs derived from cucumber green mottle mosaic virus -infected watermelon. Hortic. Plant J. 2017, 3, 17–22. [Google Scholar] [CrossRef]
  42. Sun, Y.; Fan, M.; He, Y. Transcriptome analysis of watermelon leaves reveals candidate genes responsive to cucumber green mottle mosaic virus infection. Int. J. Mol. Sci. 2019, 20, 610. [Google Scholar] [CrossRef]
  43. Li, Z.G.; Nong, Y.; Farooq, T.; Tang, Y.F.; She, X.M.; Yu, L.; Lan, G.B.; Zhou, X.P.; He, Z.F. Small RNA deep sequencing reveals the presence of multiple viral infections in cucurbit crops in Guangdong, China. J. Integr. Agric. 2022, 21, 1389–1400. [Google Scholar] [CrossRef]
  44. Li, Z.G.; Nong, Y.; Tang, Y.F.; She, X.M.; Yu, L.; Lan, G.B.; Deng, M.G.; He, Z.F. Molecular characteristic and pathogenicity analyses of cucumber green mottle mosaic virus (CGMMV) infecting bottle gourd in lianzhou, Guangdong. Zhongguo Nong Ye Ke Xue 2020, 53, 955–964. [Google Scholar]
  45. Liu, M.; Liang, Z.; Aranda, M.A.; Hong, N.; Liu, L.; Kang, B.; Gu, Q. A cucumber green mottle mosaic virus vector for virus-induced gene silencing in cucurbit plants. Plant Methods 2020, 16, 9. [Google Scholar] [CrossRef]
  46. Sun, Y.; Fan, M.; He, Y. DNA methylation analysis of the Citrullus lanatus response to cucumber greenmottle mosaic virus infection by whole-genome bisulfite sequencing. Genes 2019, 10, 344. [Google Scholar] [CrossRef]
  47. Boter, M.; Ruiz-Rivero, O.; Abdeen, A.; Prat, S. Conserved MYC transcription factors play a key role in jasmonate signaling both in tomato and Arabidopsis. Genes Dev. 2004, 18, 1577–1591. [Google Scholar] [CrossRef]
  48. Lorenzo, O.; Chico, J.M.; Sanchez-Serrano, J.J.; Solano, R. JASMONATE-INSENSITIVE1 encodes a MYC transcription factor essential to discriminate between different jasmonate-regulated defense responses in Arabidopsis. Plant Cell 2004, 16, 1938–1950. [Google Scholar] [CrossRef]
  49. Dombrecht, B.; Xue, G.P.; Sprague, S.J.; Kirkegaard, J.A.; Ross, J.J.; Reid, J.B.; Fitt, G.P.; Sewelam, N.; Schenk, P.M.; Manners, J.M.; et al. MYC2 differentially modulates diverse jasmonate-dependent functions in Arabidopsis. Plant Cell 2007, 19, 2225–2245. [Google Scholar] [CrossRef]
  50. Xue, G.P.; Kooiker, M.; Drenth, J.; McIntyre, C.L. TaMYB13 is a transcriptional activator of fructosyltransferase genes involved in beta-2,6-linked fructan synthesis in wheat. Plant J. 2011, 68, 857–870. [Google Scholar] [CrossRef]
  51. Wang, Y.; Wang, Q.; Liu, M.; Bo, C.; Wang, X.; Ma, Q.; Cheng, B.; Cai, R. Overexpression of a maize MYB48 gene confers drought tolerance in transgenic Arabidopsis plants. J. Plant Biol. 2017, 60, 612–621. [Google Scholar] [CrossRef]
  52. Devaiah, B.N.; Madhuvanthi, R.; Karthikeyan, A.S.; Raghothama, K.G. Phosphate starvation responses and gibberellic acid biosynthesis are regulated by the MYB62 transcription factor in Arabidopsis. Mol. Plant 2009, 2, 43–58. [Google Scholar] [CrossRef]
  53. Shin, R.; Burch, A.Y.; Huppert, K.A.; Tiwari, S.B.; Murphy, A.S.; Guilfoyle, T.J.; Schachtman, D.P. The Arabidopsis transcription factor MYB77 modulates auxin signal transduction. Plant Cell 2007, 19, 2440–2453. [Google Scholar] [CrossRef]
  54. Song, Y.; Yang, W.; Fan, H.; Zhang, X.; Sui, N. TaMYB86B encodes a R2R3-type MYB transcription factor and enhances salt tolerance in wheat. Plant Sci. 2020, 300, 110624. [Google Scholar] [CrossRef]
  55. Li, J.; Zhong, R.; Palva, E.T. WRKY70 and its homolog WRKY54 negatively modulate the cell wall-associated defenses to necrotrophic pathogens in Arabidopsis. PLoS ONE 2017, 12, e0183731. [Google Scholar] [CrossRef]
  56. Kloth, K.J.; Wiegers, G.L.; Busscher-Lange, J.; van Haarst, J.C.; Kruijer, W.; Bouwmeester, H.J.; Dicke, M.; Jongsma, M.A. AtWRKY22 promotes susceptibility to aphids and modulates salicylic acid and jasmonic acid signalling. J. Exp. Bot. 2016, 67, 3383–3396. [Google Scholar] [CrossRef]
  57. Gao, Q.M.; Venugopal, S.; Navarre, D.; Kachroo, A. Low oleic acid-derived repression of jasmonic acid-inducible defense responses requires the WRKY50 and WRKY51 proteins. Plant Physiol. 2011, 155, 464–476. [Google Scholar] [CrossRef]
  58. Jiang, Y.; Liang, G.; Yang, S.; Yu, D. Arabidopsis WRKY57 functions as a node of convergence for jasmonic acid- and auxin-mediated signaling in jasmonic acid-induced leaf senescence. Plant Cell 2014, 26, 230–245. [Google Scholar] [CrossRef]
  59. Jiang, Y.; Guo, L.; Liu, R.; Jiao, B.; Zhao, X.; Ling, Z.; Luo, K. Overexpression of poplar PtrWRKY89 in transgenic Arabidopsis leads to a reduction of disease resistance by regulating defense-related genes in salicylate- and jasmonate-dependent signaling. PLoS ONE 2016, 11, e0149137. [Google Scholar] [CrossRef]
  60. Skibbe, M.; Qu, N.; Galis, I.; Baldwin, I.T. Induced plant defenses in the natural environment: Nicotiana attenuata WRKY3 and WRKY6 coordinate responses to herbivory. Plant Cell 2008, 20, 1984–2000. [Google Scholar] [CrossRef]
  61. Chen, H.; Lai, Z.; Shi, J.; Xiao, Y.; Chen, Z.; Xu, X. Roles of arabidopsis WRKY18, WRKY40 and WRKY60 transcription factors in plant responses to abscisic acid and abiotic stress. BMC Plant Biol. 2010, 10, 281. [Google Scholar] [CrossRef]
  62. Abeysinghe, J.K.; Lam, K.M.; Ng, D.W. Differential regulation and interaction of homoeologous WRKY18 and WRKY40 in Arabidopsis allotetraploids and biotic stress responses. Plant J. 2019, 97, 352–367. [Google Scholar] [CrossRef]
  63. Pandey, S.P.; Roccaro, M.; Schon, M.; Logemann, E.; Somssich, I.E. Transcriptional reprogramming regulated by WRKY18 and WRKY40 facilitates powdery mildew infection of Arabidopsis. Plant J. 2010, 64, 912–923. [Google Scholar] [CrossRef]
  64. Yan, L.; Liu, Z.-Q.; Xu, Y.-H.; Lu, K.; Wang, X.-F.; Zhang, D.-P. Auto- and cross-repression of three Arabidopsis WRKY transcription factors WRKY18, WRKY40, and WRKY60 negatively involved in ABA signaling. J. Plant Growth Regul. 2012, 32, 399–416. [Google Scholar] [CrossRef]
  65. Zhao, X.Y.; Qi, C.H.; Jiang, H.; Zhong, M.S.; You, C.X.; Li, Y.Y.; Hao, Y.J. MdHIR4 transcription and translation levels associated with disease in apple are regulated by MdWRKY31. Plant Mol. Biol. 2019, 101, 149–162. [Google Scholar] [CrossRef]
  66. Lorenzo, O.; Piqueras, R.; Sanchez-Serrano, J.J.; Solano, R. ETHYLENE RESPONSE FACTOR1 integrates signals from ethylene and jasmonate pathways in plant defense. Plant Cell 2003, 15, 165–178. [Google Scholar] [CrossRef]
  67. Sun, F.; Liu, P.Q.; Xu, J.; Dong, H.S. Mutation in RAP2.6L, a transactivator of the ERF transcription factor family, enhances Arabidopsis resistance to Pseudomonas syringae. Physiol. Mol. Plant Pathol. 2010, 74, 295–302. [Google Scholar] [CrossRef]
  68. Yang, H.; Sun, Y.; Wang, H.; Zhao, T.; Xu, X.; Jiang, J.; Li, J. Genome-wide identification and functional analysis of the ERF2 gene family in response to disease resistance against Stemphylium lycopersici in tomato. BMC Plant Biol. 2021, 21, 72. [Google Scholar] [CrossRef]
  69. Moffat, C.S.; Ingle, R.A.; Wathugala, D.L.; Saunders, N.J.; Knight, H.; Knight, M.R. ERF5 and ERF6 play redundant roles as positive regulators of JA/Et-mediated defense against Botrytis cinerea in Arabidopsis. PLoS ONE 2012, 7, e35995. [Google Scholar] [CrossRef]
  70. Chen, S.; Zhou, Y.; Chen, Y.; Gu, J. fastp: An ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 2018, 34, i884–i890. [Google Scholar] [CrossRef]
  71. Kim, D.; Langmead, B.; Salzberg, S.L. HISAT: A fast spliced aligner with low memory requirements. Nat. Methods 2015, 12, 357–360. [Google Scholar] [CrossRef]
  72. Pertea, M.; Pertea, G.M.; Antonescu, C.M.; Chang, T.C.; Mendell, J.T.; Salzberg, S.L. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 2015, 33, 290–295. [Google Scholar] [CrossRef]
  73. Li, B.; Dewey, C.N. RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011, 12, 323. [Google Scholar] [CrossRef]
  74. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef]
  75. Zhang, C.; Zheng, H.; Wu, X.; Xu, H.; Han, K.; Peng, J.; Lu, Y.; Lin, L.; Xu, P.; Wu, X.; et al. Genome-wide identification of new reference genes for RT-qPCR normalization in CGMMV-infected Lagenaria siceraria. PeerJ 2018, 6, e5642. [Google Scholar] [CrossRef]
  76. Zhang, S.; Ying, H.; Pingcuo, G.; Wang, S.; Zhao, F.; Cui, Y.; Shi, J.; Zeng, H.; Zeng, X. Identification of potential metabolites mediating bird’s selective feeding on Prunus mira flowers. BioMed Res. Int. 2019, 2019, 1395480. [Google Scholar] [CrossRef]
  77. Fraga, C.G.; Clowers, B.H.; Moore, R.J.; Zink, E.M. Signature-discovery approach for sample matching of a nerve-agent precursor using liquid chromatography-mass spectrometry, XCMS, and chemometrics. Anal. Chem. 2010, 82, 4165–4173. [Google Scholar] [CrossRef]
  78. Chen, W.; Gong, L.; Guo, Z.; Wang, W.; Zhang, H.; Liu, X.; Yu, S.; Xiong, L.; Luo, J. A novel integrated method for large-scale detection, identification, and quantification of widely targeted metabolites: Application in the study of rice metabolomics. Mol. Plant 2013, 6, 1769–1780. [Google Scholar] [CrossRef]
Figure 1. Symptoms of CGMMV in bottle gourd and RT-PCR and Western blot detection of CGMMV. (A) Symptoms of CGMMV in bottle gourd. Agrobacterium strains containing CGMMV infectious cDNA clones or an empty vector were infiltrated into the cotyledons of bottle gourd. Photos were taken at 12 dpi. (B) At 12 dpi, the upper leaves of bottle gourd were taken and detected using CGMMV CP specific primer and CP antibody.
Figure 1. Symptoms of CGMMV in bottle gourd and RT-PCR and Western blot detection of CGMMV. (A) Symptoms of CGMMV in bottle gourd. Agrobacterium strains containing CGMMV infectious cDNA clones or an empty vector were infiltrated into the cotyledons of bottle gourd. Photos were taken at 12 dpi. (B) At 12 dpi, the upper leaves of bottle gourd were taken and detected using CGMMV CP specific primer and CP antibody.
Ijms 24 16566 g001
Figure 2. Transcriptome analysis of bottle gourd in response to CGMMV infection. (A) Volcano plot showing the DEGs revealed via RNA-seq analysis. Red, green, and black dots represent the upregulated, downregulated, and insignificant DEGs, respectively. (B) Clustering heatmap of the DEGs between ZLM12 and ZLV12. The abscissa of each sample represents three replicates. ZLM12 represents mock treatment and ZLV12 represents CGMMV treatment. (C) KEGG enrichment analysis of the DEGs.
Figure 2. Transcriptome analysis of bottle gourd in response to CGMMV infection. (A) Volcano plot showing the DEGs revealed via RNA-seq analysis. Red, green, and black dots represent the upregulated, downregulated, and insignificant DEGs, respectively. (B) Clustering heatmap of the DEGs between ZLM12 and ZLV12. The abscissa of each sample represents three replicates. ZLM12 represents mock treatment and ZLV12 represents CGMMV treatment. (C) KEGG enrichment analysis of the DEGs.
Ijms 24 16566 g002
Figure 3. qRT-PCR validation of the selected DEGs revealed via transcriptome analysis. Validation of 20 DEGs using qRT-PCR. The first 13 graphs show the upregulated DEGs, and the last 7 graphs show the downregulated DEGs. V, CGMMV-infected samples. M, mock samples. Annotation of the DEGs is shown in Table 2. Asterisks indicate the p-value between mock and CGMMV plants using Student’s t-test method. * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 3. qRT-PCR validation of the selected DEGs revealed via transcriptome analysis. Validation of 20 DEGs using qRT-PCR. The first 13 graphs show the upregulated DEGs, and the last 7 graphs show the downregulated DEGs. V, CGMMV-infected samples. M, mock samples. Annotation of the DEGs is shown in Table 2. Asterisks indicate the p-value between mock and CGMMV plants using Student’s t-test method. * p < 0.05, ** p < 0.01, *** p < 0.001.
Ijms 24 16566 g003
Figure 4. Metabolome analysis of bottle gourd in response to CGMMV infection. (A) Volcano plot showing the DAMs between the mock and CGMMV-infected samples. Red, green, and gray dots represent the upregulated, downregulated, and insignificant DAMs, respectively. (B) Clustering heatmap of the DEGs between ZLM12 and ZLV12. Each treatment contains three replicates. (C) KEGG enrichment analysis of the DAMs.
Figure 4. Metabolome analysis of bottle gourd in response to CGMMV infection. (A) Volcano plot showing the DAMs between the mock and CGMMV-infected samples. Red, green, and gray dots represent the upregulated, downregulated, and insignificant DAMs, respectively. (B) Clustering heatmap of the DEGs between ZLM12 and ZLV12. Each treatment contains three replicates. (C) KEGG enrichment analysis of the DAMs.
Ijms 24 16566 g004
Figure 5. Analysis of hormone signaling transduction in response to CGMMV infection. Joint analyses of hormone signaling pathway in response to CGMMV infection. Red blocks represent upregulated genes, green blocks represent downregulated genes, and blue blocks represent the genes which are not consistent between different replicates of each treatment. Red dot represents that the hormone is up-accumulated.
Figure 5. Analysis of hormone signaling transduction in response to CGMMV infection. Joint analyses of hormone signaling pathway in response to CGMMV infection. Red blocks represent upregulated genes, green blocks represent downregulated genes, and blue blocks represent the genes which are not consistent between different replicates of each treatment. Red dot represents that the hormone is up-accumulated.
Ijms 24 16566 g005
Figure 6. Analysis of DEGs and DAMs involved in JAs synthesis pathway with CGMMV infection. (A) Scheme of the JAs synthesis pathway. DEGs and DAMs identified via transcriptome and metabolome analyses are marked in red. (B) qRT-PCR verification of the DEGs involved in the JAs synthesis pathway. Asterisks indicate the significance of difference between mock and CGMMV plants using Student’s t-test method. *** p < 0.001. (C) Comparison of the accumulation of JA and JA-Ile in mock and CGMMV-treated bottle gourd. *** p < 0.001.
Figure 6. Analysis of DEGs and DAMs involved in JAs synthesis pathway with CGMMV infection. (A) Scheme of the JAs synthesis pathway. DEGs and DAMs identified via transcriptome and metabolome analyses are marked in red. (B) qRT-PCR verification of the DEGs involved in the JAs synthesis pathway. Asterisks indicate the significance of difference between mock and CGMMV plants using Student’s t-test method. *** p < 0.001. (C) Comparison of the accumulation of JA and JA-Ile in mock and CGMMV-treated bottle gourd. *** p < 0.001.
Ijms 24 16566 g006
Figure 7. Verification of the expressions of TFs using qRT-PCR. (A) Expressions of Myc TFs. Myc2, Lsi09G016530. Myc3, Lsi07G011090. (B) Expressions of Myb TFs. Myb13, Lsi05G010160. Myb48, Lsi10G013470. Myb62, Lsi02G001830. Myb77, Lsi04G015930. Myb86, Lsi06G015850. (C) Expressions of TF NAC (Lsi08G004490). (D) Expressions of WRKY TFs. WRKY6, Lsi05G021100. WRKY18, Lsi02G012150. WRKY31, Lsi05G014110. WRKY68, Lsi06G014830. (E) Expressions of ERF TFs. ERF1, Lsi06G008160. ERF1b, Lsi09G012390. ERF-RAP2, Lsi08G002460. ERF-2, Lsi07G001170. ERF-2b, Lsi03G010920. ERF-6, Lsi04G020490. The significance of difference between mock and CGMMV plants was tested via p-value using Student’s t-test method. * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 7. Verification of the expressions of TFs using qRT-PCR. (A) Expressions of Myc TFs. Myc2, Lsi09G016530. Myc3, Lsi07G011090. (B) Expressions of Myb TFs. Myb13, Lsi05G010160. Myb48, Lsi10G013470. Myb62, Lsi02G001830. Myb77, Lsi04G015930. Myb86, Lsi06G015850. (C) Expressions of TF NAC (Lsi08G004490). (D) Expressions of WRKY TFs. WRKY6, Lsi05G021100. WRKY18, Lsi02G012150. WRKY31, Lsi05G014110. WRKY68, Lsi06G014830. (E) Expressions of ERF TFs. ERF1, Lsi06G008160. ERF1b, Lsi09G012390. ERF-RAP2, Lsi08G002460. ERF-2, Lsi07G001170. ERF-2b, Lsi03G010920. ERF-6, Lsi04G020490. The significance of difference between mock and CGMMV plants was tested via p-value using Student’s t-test method. * p < 0.05, ** p < 0.01, *** p < 0.001.
Ijms 24 16566 g007
Figure 8. Effect of AOS gene silencing on CGMMV infection. (A) Agrobacterium strains containing empty vector (mock), CGMMV-gfp, CGMMV-BgAOS, or CGMMV-Bgpds were infiltrated into the cotyledons of bottle gourd. Photos were taken at 12 dpi. (B) Western blot detection of CGMMV accumulation with CP antibody. Rubisco was used as the equal loading control. (C) qRT-PCR analysis of AOS mRNA accumulation. The significance of difference between CGMMV-gfp and CGMMV-BgAOS was tested via p-value using Student’s t-test method. *** p < 0.001.
Figure 8. Effect of AOS gene silencing on CGMMV infection. (A) Agrobacterium strains containing empty vector (mock), CGMMV-gfp, CGMMV-BgAOS, or CGMMV-Bgpds were infiltrated into the cotyledons of bottle gourd. Photos were taken at 12 dpi. (B) Western blot detection of CGMMV accumulation with CP antibody. Rubisco was used as the equal loading control. (C) qRT-PCR analysis of AOS mRNA accumulation. The significance of difference between CGMMV-gfp and CGMMV-BgAOS was tested via p-value using Student’s t-test method. *** p < 0.001.
Ijms 24 16566 g008
Table 1. Overview of the RNA-seq data of bottle gourd.
Table 1. Overview of the RNA-seq data of bottle gourd.
SampleRaw ReadsClean ReadsClean BaseUnique Mapped ReadsQ20 (%)Q30 (%)GC Content (%)
ZLM12160,300,77459,025,2408.85 G55,742,483 (94.44%)97.4893.0244.80
ZLM12250,095,73249,062,6107.36 G46,436,613 (94.65%)97.4392.8745.17
ZLM12348,349,01647,355,4427.10 G45,101,108 (95.24%)97.6193.2644.86
ZLV12144,431,75443,343,2106.50 G41,374,202 (95.46%)97.7793.6144.56
ZLV12251,042,94650,073,4047.51 G46,079,504 (92.02%)97.5093.0244.48
ZLV12343,970,22842,963,2646.44 G40,631,748 (94.57%)97.4492.9244.92
Table 2. Annotation of the selected DEGs for qRT-PCR verification.
Table 2. Annotation of the selected DEGs for qRT-PCR verification.
Gene IDDescription
Lsi01G013470Aquaporin PIP2-2-like
Lsi02G017750Probable calcium-binding protein CML44
Lsi05G011760Probable membrane-associated kinase regulator 6
Lsi04G015060Cytochrome P450 81D1-like isoform X1
Lsi02G007470E3 ubiquitin-protein ligase RMA1H1-like
Lsi02G018990Heterodimeric geranylgeranyl pyrophosphate synthase small subunit, chloroplastic-like
Lsi01G009350Probable indole-3-acetic acid-amido synthetase GH3.3
Lsi02G021080Pectinesterase
Lsi02G008430Serine/threonine-protein kinase STY8
Lsi11G012260ACT-like tyrosine kinase family protein
Lsi01G014560NADPH-dependent aldo-keto reductase, chloroplastic-like
Lsi11G005040Disease resistance protein (TIR-NBS-LRR class)
Lsi05G018940Invertase/pectin methylesterase inhibitor family protein
Lsi05G012660Dehydration responsive element-binding protein 1
Lsi04G002080MATE efflux family protein
Lsi03G008210UDP-glycosyltransferase 74F1
Lsi09G006560DUF4228 domain protein
Lsi02G013460Scarecrow transcription factor family protein
Lsi11G000250Protein brassinosteroid insensitive 1
Lsi03G014380Seed maturation-like protein
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, Z.; Tang, Y.; Lan, G.; Yu, L.; Ding, S.; She, X.; He, Z. Transcriptome and Metabolome Analyses Reveal That Jasmonic Acids May Facilitate the Infection of Cucumber Green Mottle Mosaic Virus in Bottle Gourd. Int. J. Mol. Sci. 2023, 24, 16566. https://doi.org/10.3390/ijms242316566

AMA Style

Li Z, Tang Y, Lan G, Yu L, Ding S, She X, He Z. Transcriptome and Metabolome Analyses Reveal That Jasmonic Acids May Facilitate the Infection of Cucumber Green Mottle Mosaic Virus in Bottle Gourd. International Journal of Molecular Sciences. 2023; 24(23):16566. https://doi.org/10.3390/ijms242316566

Chicago/Turabian Style

Li, Zhenggang, Yafei Tang, Guobing Lan, Lin Yu, Shanwen Ding, Xiaoman She, and Zifu He. 2023. "Transcriptome and Metabolome Analyses Reveal That Jasmonic Acids May Facilitate the Infection of Cucumber Green Mottle Mosaic Virus in Bottle Gourd" International Journal of Molecular Sciences 24, no. 23: 16566. https://doi.org/10.3390/ijms242316566

APA Style

Li, Z., Tang, Y., Lan, G., Yu, L., Ding, S., She, X., & He, Z. (2023). Transcriptome and Metabolome Analyses Reveal That Jasmonic Acids May Facilitate the Infection of Cucumber Green Mottle Mosaic Virus in Bottle Gourd. International Journal of Molecular Sciences, 24(23), 16566. https://doi.org/10.3390/ijms242316566

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