Skip to main content

Metatranscriptome analysis of symptomatic bitter apple plants revealed mixed viral infections with a putative novel polerovirus

Abstract

Background

Next-generation Sequencing (NGS) combined with bioinformatic analyses constitutes a powerful approach for identifying and characterizing previously unknown viral genomes. In this study, leaf samples from bitter apple plants (Citrullus colocynthis (L.) Schrad) exhibiting symptoms such as dwarfing, leaf crinkling, and chlorosis were collected from the southern part of Kerman province, Iran.

Results

Putative infecting viruses were identified through de novo assembly of sequencing reads using various tools, followed by BLAST analysis. Complete genomes for Squash vein yellowing virus (SqVYV), Citrus-associated rhabdovirus (CiaRV), and a novel polerovirus-related strain termed Bitter apple aphid-borne yellows virus (BaABYV) were assembled and characterized. Additionally, a partial genome for Watermelon mosaic virus (WMV) was assembled. The genomic organization of the BaABYV was determined to be 5’-ORF0-ORF1-ORF1,2-ORF3a-ORF3-ORF3,5-ORF4-3’. Amino acid sequence identities for inferred proteins (P0 and P1, P1,2) with known poleroviruses were found to be the 90% species delineation limit, implying that BaABYV should be considered a new member of the genus Polerovirus. Recombination events were observed in the BaABYV and WMV strains; such events were not found in the CiaRV strain.

Conclusions

Molecular evidence from this study suggests that C. colocynthis is a reservoir host of several plant viruses. Among them, BaABYV is proposed as a new member of the genus Polerovirus. Furthermore, the CiaRV strain has been reported for the first time from Iran.

Peer Review reports

Background

In agricultural fields, weeds contribute to increase competition for essential resources like water and nutrients. Furthermore, they can serve as reservoirs for viruses that pose infection risks to neighboring crops [1]. One such plant is C. colocynthis, commonly referred to as bitter apple or colocynth, which belongs to the Cucurbitaceae family and is adapted to arid environments. The bitter apple fruit is valued for its medicinal benefits [2] and is extensively cultivated in southern Iran [3]. Previous studies have identified several viruses in C. colocynthis, including Cucurbit aphid-borne yellows virus (CABYV), Squash mosaic virus (SqMV), Papaya ring spot virus-type W (PRSV-W), Cucumber mosaic virus (CMV), Zucchini yellow mosaic virus (ZYMV), and Watermelon mosaic virus (WMV). These findings suggest that wild species like C. colocynthis can act as a viral reservoir affecting agricultural crops [4,5,6,7,8].

Research on viruses in wild plants offers multiple advantages, such as enhancing our understanding of viral diversity and host-virus interactions, conserving biodiversity, and providing insights into viral evolution [9]. Next-generation Sequencing (NGS) has revolutionized this field by enabling comprehensive sequencing of all viral genomes in a plant tissue sample [10]. This high-throughput technology sequences the entire viral genome after isolating its RNA or DNA from the plant tissue, facilitating rapid identification and characterization of known and unknown viruses [11,12,13]. In particular, metagenomics is a valuable NGS technique for detecting unidentified plant viruses, especially in asymptomatic hosts where traditional methods may not easily detect them. Metagenomics sequences all genetic material in a sample, providing a comprehensive snapshot of the viral community present [14].

Despite the bitter apple is native to west Asia including Iran, there has been notable gap in research identifying the viruses associated with these plants. A study by Sharifi et al. [8] made significant contributions in this area by detecting the WMV in bitter apple plants in the southern Kerman province, Jiroft region. However, this research did not explore the full spectrum of viruses potentially affecting these plants. In an effort to extend the findings of earlier research, our study aims to deepen the understanding of the virome associated with bitter apple plants. Using NGS technology, we focused on a group of bitter apple plants from the Jiroft region, all exhibiting virus-like symptoms. Our study led to discovery of a mixed viral infection in bitter apple plants, including the identification of a new type of a polerovirus.

Materials and methods

Sample collection

To investigate potential viruses infecting C. colocynthis, we collected samples with symptoms of viral diseases, including dwarfing, leaf crinkling, and mild chlorosis (Fig. 1) from a desert area in the Jiroft region (28°26’33.4"N 57°54’17.0” E) in July 2021. The specificity of our selection criteria led us to identify a limited number of samples (n = 7) that clearly exhibited these viral symptoms. These samples were immediately placed in plastic bags, flash-frozen in liquid nitrogen, and transferred to the Shahid Bahonar University laboratory. The plant materials utilized in our study were identified and verified in the herbarium (MIR-4307) by Dr. S.M. Mirtadzaddini from the Department of Biology, Faculty of Science at Shahid Bahonar University of Kerman.

They were stored at -80 °C pending further analysis.

Fig. 1
figure 1

A bitter apple plant exhibiting suspected viral symptoms, including dwarfing, leaf crinkling, and mild chlorosis, in its natural habitat (Jiroft region, Kerman province, Iran)

Total RNA extraction and sequencing

A pooled sample from leaves with relatively severe symptoms was ground using a mortar and pestle. Total RNA was extracted using the TOP Plant and Fungi RNA Purification Kit (mini-prep; Cat. No; TGK2004), following the manufacturer’s instructions (Topazgen, Iran). The integrity of the RNA was confirmed by the presence of distinct bands corresponding to 28 S ribosomal RNA (rRNA) (~ 4.8 kb), 18 S rRNA (~ 2.0 kb), and 5.8 S rRNA (~ 154 nt) on an agarose gel. High-quality RNA samples were purified using kit and concentrated prior to sequencing. Novogen (Beijing, China) constructed a paired-end sequencing library using the Illumina HiSeq 6000 sequencing platform.

Sequence analysis

The raw RNA-seq data were evaluated by FastQC v.0.11.9 [15]. Raw reads were processed using Cutadapt (Version 2.0.4) [16] to retain only those with a minimum length of 50 bases and a quality score exceeding 30; this step trimmed low-quality reads and adapter sequences. The sequencing data were then assembled using Trinity (v2.4.0) [17], SPAdes (v3.13.0) [18], and CLC Genomics Workbench (22.0.2) (CLC Bio, Aarhus, Denmark), with their default parameters. The assembled contigs were subjected to BLAST analysis against public nucleotide datasets and the Reference Viral DataBase (RVDB) (V 25.0) to identify potential viruses. The purified reads were mapped against the reference genome of the most closely related virus using NextGenMap 0.5.0 [19] to validate the results of de novo assembly approaches. Samtools flagstat (Version 2.0.4) [20] provided descriptive statistics for the BAM files and read counts for each viral reference genome.

Phylogenetic analysis

We compiled full-genome sequences of identified viruses and aligned them independently using MAFFT Version 7.0 [21]. The nearest non-target virus was selected as an outgroup based on BLASTN results. Potential recombinant isolates have been detected using seven recombination detection methods implemented in the RDP4 package (RDP, GENECONV, BootScan, MaxChi, Chimaera, SiScan, and 3Seq) [22]. Recombinant sequences were considered valid if detected by at least four of the methods and with a P-value less than 10− 6 under the default setting for linear sequences. These identified recombinants were excluded from the multiple sequence alignments (MSAs). Subsequently, we created a maximum likelihood phylogenetic tree (MLtree) using the IQtree program [23]. The IQ-tree automatically selected the best model using the ModelFinder program implemented in the IQ-tree tool. The reconstructed consensus ML tree was visualized using the Figtree v.1.4.3 (http://tree.bio.ed.ac.uk/software/figtree). Nucleotide pairwise distances were computed using the default option in MEGAX [24].

RT-PCR and sanger sequencing

Total RNA was extracted using a Spectrum Plant Total RNA Kit (Sigma Aldrich, USA), from the pooled sample, previously prepared for NGS analysis, which included seven bitter apple plants. For the reverse transcription (RT) reaction, a mixture was prepared consisting of 3 µL of reverse primer (20 pmol) (Table 1), 5 µL of RNA sample, and 8.5 µL of sterile deionized distilled water. The mixture was incubated at 95 °C for 1 min and cooled on ice for 3 min to denature the RNA. Subsequently, 4 µL of 5× M-MLV RT buffer, 2 µL of dNTPs mix (10 mM), 1 µL M-MLV reverse transcriptase (200 U µL), and 1 µL RNase inhibitor (20 U/µL) (Sinaclon, Iran) were added. The reaction mixture was then incubated at 42 °C for 60 min and terminated by heating at 65 °C for 10 min. For the RT-PCR, 2.5 µL of cDNA was mixed with 7.5 µL DEPC-treated water, 1.25 µL of 5× GoTag polymerase buffer, 2.5 µL of 10× MgCl2, 0.5 µL of each forward and reverse primer (20 pmol), 0.75 µL of dNTP mix (10 mM) and 0.125 µL of GoTag polymerase (2.5 U/µL) (Sinaclon, Iran). The RT-PCR program included an initial denaturation at 94 °C for 3 min, followed by 35 cycles of 30 s at 94 °C for denaturation, 30 s of annealing at temperatures specified in Tables 1 and 30s of extension at 72 °C, and concluded with a final extension step at 72 °C for 10 min.

RT-PCR products were analyzed by electrophoresis on a 1% agarose gel and visualized with ethidium bromide staining. The products were then purified using an Agarose Gel DNA Extraction Kit (Sangon, Shanghai, China) and sequenced directly with the Applied Biosystems 3500 Genetic Analyzer (Foster City, CA, USA) using the RT-PCR primers in the forward direction.

Table 1 PCR primers designed to detect and discriminate viruses in this study

Results

Data processing and assembly

After trimming the raw reads using the Cutadapt program, 98.90% of the reads were retained for further analysis. These trimmed reads were then assembled into contigs using three different de novo assembly methods: (i) Trinity, (ii) SPAdes, and (iii) CLC Genomic workbench. The assembly metrics, such as contig count, maximum length, total length, minimum length, average length and N50 values, for each method are compared and summarized in Appendix Table A1. Trinity generated the highest number of contigs (77,188), while SPAdes achieved the highest N50 value (1,834), indicating the assembly of contigs with superior quality. Despite SPAdes’s higher N50, the analysis proceeded with contigs assembled by Trinity. This decision was motivated by the more significant number of contigs from Trinity, which could offer additional data richness and uncover more insights into the virome. Moreover, it could be essential for identifying low-abundance viruses or novel variants that might be missed when focusing only on high-quality contigs.

Virus identification

All de novo assembled contigs from Trinity exceeding 3,500 bp were subjected to BLASTN for virus identification, searching against available nucleotide datasets and DataBase RVDB based on percentage of identity. Four contigs corresponding viruses within genera Potyvirus, Polerovirus, Rhabdovirus, and Ipomovirus were identified, with the details summarized in Table 2.

Table 2 The parameter values of BLASTN-aligned contigs and their two closest virus sequences

Characterization and phylogenetic analysis of the BaABYV-IR-1 strain

Computational analyses revealed that contig C-1 from the BaABYV-IR-1 strain has a nucleotide length of 5,816. It exhibited over 90% identity with the Pepo aphid-borne yellows virus (PABYV), which is classified within the genus Polerovirus (Table 2). To validate these findings, the trimmed reads from the BaABYV-IR-1 strain were mapped to the reference genomes of PABYV (NC_030225) and pumpkin polerovirus (PuPV) (NC_055513). The mapping confirmed that 0.01% of the trimmed reads, ranging from 3,744 to 3,790 nucleotides, were re-mapped onto the reference genomes. The bioinformatic analysis highlighted the typical ORF structure of the BaABYV-IR-1 strain (the contig C-1), revealing 7 ORFs characterized in Table 3.

Table 3 ORF organization of contig C-1 from BaABYV-IR-1, including putative proteins and closely related strains

The reconstructed ML phylogenetic tree included 29 sequences, comprising the whole genome of the BaABYV-IR-1 isolate and reference sequences of other poleroviruses (see Appendix Table A2). The sequence matrix had 9,671 characters, 6,781 distinct patterns, and 5,405 parsimony-informative, 1,530 singleton, and 2,736 constant sites. The best-fit model determined by Bayesian Information Criterion (BIC) was GTR + F + I + G4. According to the phylogenetic tree, BaABYV-IR-1 strain is closely grouped with both PuPV and PABYV, suggesting that it may represent a divergent lineage within the genus Polerovirus (Fig. 2a). The phylogeny distinction is further supported by the calculated pairwise distances, which are represented in a heatmap (Fig. 2b), where BaABYV-IR-1’s proximity to PABYV compared to PuPV is notable. Based on this result, estimated that the major and minor parents were likely to be BrYV and PeVYV (Fig. 2c). This region of recombination was identified in the BaABYV-IR-1 isolate within ORF5 (Fig. 2d).

Fig. 2
figure 2

(a) The ML phylogenetic tree, rooted with Groundnut enation virus (GEV), is based on polerovirus sequences including BaABYV-IR-1 and the reference sequences of poleroviruses. The bootstrap values below 100 are indicated in the main nodes. (b) Heatmap displaying the pairwise distance between polerovirus reference nucleotide sequences and BaABYV-IR-1, calculated by MEGA X. The color intensity corresponds to distance levels, with dark red for highest and dark green for the lowest distance. (c) Recombination event in the BaABYV-IR-1 genome, identified through RDP analysis including reference polerovirus sequences and BaABYV-IR-1. This event was detected by RDP, MaxChi, Chimaera, SiScan, and 3Seq with a P-value less than 10− 09. The estimated major and minor parents were BrYV and PeVYV. (d) The recombination region is demarcated in red from positions 4,166 to 4,890 within the BaABYV-IR-1 sequence in ORF5

Identification and phylogeny of SqVYV-IR-BA isolate

The findings indicated a high sequence similarity (98.42% identity) between contig C-2, designated Squash vein yellowing virus isolate IR-BA (SqVYV-IR-BA), and the reference genome of SqVYV, a member of the family Potyviridae (Table 2). Hence, following the ICTV guidelines, SqVYV-IR-BA is an isolate of the SqVYV species. To confirm these results, NextGenMap 0.5.0 was used to map trimmed reads of the SqVYV-IR-BA to the reference genome of SqVYV (NC_010521). Consequently, approximately 0.10% of the reads (42,715 nt) were successfully re-mapped to the reference genome. Table 4 provides a summary of the predicted ORF/region and amino acid length for each segment in the SqVYV-IR-BA isolate, including their start and stop nucleotide positions. Additionally, it features the identity percentages of nucleotides and amino acids, comparing sequences most similar to the SqVYV-IR-BA isolate with data sourced from GenBank.

Table 4 Genomic regions and ORF structure of SqVYV-IR-BA, and its similarity to some GenBank isolates

The reconstructed ML tree was based on a matrix including 16 sequences (the SqVYV-IR-BA complete genome and the reference sequences of ipomoviruses; see Appendix, Table A3), with 13,668 characters, 7,892 distinct patterns, and 7,344, 2,823 and 3,501 parsimony-informative, singleton, and constant sites, respectively. The best-fit model based on BIC was GTR + F + I + G4. Figure 3b displays a heatmap created from a matrix that includes the pairwise distances between the nucleotide sequences of reference ipomoviruses and the SqVYV-IR-BA sequence.

The ML tree revealed two distinct clades of SqYVV isolates: one containing isolates from the USA and the other from the Middle East (Fig. 3a). Isolate SqVYV-IR-BA clustered with isolates from Middle East (SqVY-Iraq and SqVY-IL), signifying potential geographical influence on the genetic variability (Fig. 3c). The recombination detection test did not identify SqVYV-IR-BA as a recombinant isolate (Fig. 3d).

Fig. 3
figure 3

(a) The maximum likelihood phylogenetic tree based on reference sequences of imopoviruses and isolate SqVYV-IR-BA, rooted by WMV. The bootstrap values < 100 are indicated in the main nodes. (b) Heatmap based on a matrix including the pairwise distance between the nucleotide sequence of reference sequences of ipomoviruses with the sequence of SqVYV-IR-BA calculated by MEGA X. The color intensity indicates the level of distance between the species. Dark red indicates the highest number, while dark green represents the lowest number in the heatmaps. (c) Recombination event in genome of SqVYV-Iraq isolate detected by RDP analysis of a matrix including reference sequences of ipomovirus, available SqVYV isolates, and SqVYV-IR-BA. This event has been detected by RDP, GENECONV, BootScan, MaxChi, SiScan, and 3Seq with a P-value less than 10− 09. The estimated major and minor parents were SqVYV-IR-BA and SqYV0-SM2008cHe. (d) The schematic presentation of the SqVYV-IR-BA genome. The recombination region is shown in red color in positions 1,178 -1,371 in the SqVYV-IR-BA sequence located in the P1a region

Identification and phylogeny analysis of WMV-IR-BA

Bioinformatic analysis identified a contig labeled C-3, designated as WMV-IR-BA, with a length of 3,757 nucleotides. This contig exhibited approximately 96% identity with WMV isolates CHI87-620 and VE10-099 which are documented in GenBank under accession numbers EU660580 and KC292915 respectively.

The contig known as WMV-IR-BA was found to contain an incomplete ORF encompassing four genomic regions: NIa-VPg (nuclear inclusion VPg protein), NIa-Pro (nuclear inclusion protein), NIb (nuclear inclusion b) and CP (coat protein gene). The contig was mapped to the reference genome of WMV (NC_006262) using NextGenMap 0.5.0, confirming the initial findings. Table 5 outlines the predicted ORF/region, amino acid length, and the positions of each segment in the WMV-IR-BA isolate. It also presents the identity percentages for nucleotides and amino acids, comparing sequences closely related to the WMV-IR-BA isolate as found in GenBank.

Table 5 ORF structure of the WMV-IR-BA isolate, and its similarity to some GenBank isolates

The reconstructed ML-tree included nucleotide sequences from the complete genome of WMV-IR-BA, 10 closely related WMV isolates, along with reference sequences from the Bean common mosaic virus (BCMV) subgroup of potyviruses (Appendix, Table A4). This selection was made due to the close relationship between WMV and other members of the BCMV subgroup [25]. The phylogenetic tree was based on a matrix including 33 sequences with 5,039 characters, 3,001 distinct patterns, 2,573, 510, and 1,956 parsimony-informative, singleton, and constant sites, respectively. The best-fit model for this dataset, based on BIC was GTR + F + I + G4. Within this framework, WMV-IR-BA and two isolates from South America formed a well-supported clade (Fig. 4a). The heatmap based on the distance matrix between WMV-IR-BA and other reference sequences of subgroup BCMV has been displayed in Fig. 4b. The WMV-IR-BA isolate was identified as a recombinant through RDP analysis, with major and minor parents traced back to France and South Korea (Fig. 4c). The recombination region was identified in the WMV-IR-BA isolate within the NIb and CP regions (Fig. 4d).

Fig. 4
figure 4

(a) The maximum likelihood phylogenetic tree based on partial sequence of the Watermelon mosaic virus isolate (WMV-IR-BA) using 10 WMV isolates, reference sequence of potyviruses and subgroup BCMV, rooted by LMV. The bootstrap values < 100 are indicated in the main nodes. (b) Heatmap based on a matrix including the pairwise distance between the nucleotide sequence of reference sequences of potyviruses, subgroup BCMV with the sequence of the WMV-IR-BA isolate calculated by MEGA X. The color intensity indicates the level of distance between the species. Dark red indicates the greatest divergence, while dark green indicates the least. (c) Recombination event in genome of the WMV-IR-BA isolate detected by RDP analysis of a matrix including reference sequences of potyviruses, 10 WMV isolates, and the WMV-IR-BA isolate. This event has been detected by GENECONV, BootScan, MaxChi, SiScan, and 3Seq with a P-value less than 10− 06. The estimated major and minor parents were WMV-C05-465 and Buan 4 − 1. (d) The schematic presentation of the WMV-IR-BA genome. The recombination region is in red color in positions 2,241-3,757 in the WMV-IR-BA sequence, located in the NIb and CP regions

Identification and phylogeny of the CiaRV-IR-BA isolate

The contig labeled C-4, referred to as citrus-associated rhabdovirus isolate IR-BA (CiaRV-IR-BA), has a length of 13,443 nucleotides. It has a maximum identity of 80.82% with the CiaRV (MT302545), classified in the genus Cytorhabdovirus within the family Rhabdoviridae [26]. The nucleotide and amino acid identity between CiaRV-IR-BA and the closest strain ranged between 80 and 90% and 85–95% respectively for nearly all ORFs. Notably, ORF4 exhibits a lower identity, with 73% at the nucleotide level and 65% (Table 6). For further analyses, the trimmed reads were mapped to the reference genome of CiaRV (MT302542) using NextGenMap 0.5.0. According to the results, 14,580 reads were re-mapped on the reference genome.

Table 6 Genomic regions and ORF structure of the CiaRV-IR-BA (the coting-4), and its similarity to selected GenBank isolates

The reconstructed ML tree was based on a matrix including 42 sequences, which encompassed the whole genome of CiaRV-IR-BA and the reference sequences of cytorhabdoviruses, along with 10 CiaRV isolates closely related to CiaRV-IR-BA (see Appendix, Table A5). This matrix contained 25,146 characters, 20,731 distinct patterns, and 17,115, 3,585, and 4,446 parsimony-informative, singleton, and constant sites, respectively. The optimal model for this dataset based on BIC, was GTR + F + I + G4.

The phylogenetic tree highlighted that CiaRV isolates, including the CiaRV-IR-BA, diverged significantly from other sequences, forming a unique clade, as shown in Fig. 5a. The heatmap based on the distance matrix between CiaRV-IR-BA and other reference sequences of the subgroup Cytorhabdovirus has been displayed in Fig. 5b. The CiaRV-IR-BA isolate is not a recombinant nor a parent of recombinants based on RDP analysis (Fig. 5b).

Fig. 5
figure 5

(a) The maximum likelihood phylogenetic tree based on the partial sequence of the isolate CiaRV-IR-BA, 10 CiaRV isolates, and the reference sequences of cytorhabdoviruses rooted by SYNV. The bootstrap values < 100 are indicated in the main nodes. (b) Heatmap based on a matrix including the pairwise distance between the nucleotide sequence of the reference sequences of cytorhabdoviruses with the sequence of CiaRV-IR-BA calculated by MEGA X. The color intensity indicates the level of the distance between the species. Dark red indicates the highest number, while dark green represents the lowest number in the heatmap

RT-PCR assay for verification of RNA-Seq data

In our RNASeq analysis, we primarily identified four large contigs corresponding to the four different viruses: BaABYV-IR, SqVYV-IR-BA, WMV-IR-BA, and CiaRV-IR-BA (Table 2). To confirm the results of RNA-Seq analysis, we conducted RT-PCR assays using virus-specific primers on the same pooled sample that previously prepared for NGS analysis, comprising seven bitter apple plants. As listed in Table 1, these primers were specifically designed to target distinct regions of each viral genome, thereby enabling their precise and unambiguous identification.

The RT-PCR successfully yielded amplified products of the expected sizes for each virus. Subsequent sequencing of these amplified products validated the presence of BaABYV-IR, SqVYV-IR-BA, WMV-IR-BA, and CiaRV-IR-BA in the bitter apple sample (sequencing data not provided).

Discussion

Metagenomics has emerged as a powerful tool for detecting viruses in plants, even when no symptoms are visible. It allows the simultaneous analysis of all genetic material in a sample, detecting both known and unknown viruses, as well as multiple viruses in a single sample [13]. Despite these advantages, the technique is not without limitations. One significant challenge is the lack of virion-enriched methods, which can hinder the acquisition of high-quality, representative viral samples, thus introducing bias into the data [12]. Moreover, the genetic diversity of viral communities often complicates data interpretation, especially for uncharacterized viruses not in databases. The computational tools for metagenomic analysis are continually evolving but require significant computational power [27].

To decrease these biases in our study, we implemented pool sampling by combining samples from available symptomatic plants, aiming to enhance virion enrichment. We also integrated a purification step to improve the quality of the genetic material for subsequent NGS analysis. Utilizing the Illumina HiSeq 6000 sequencing platform, known for its high accuracy and quality, we sought to improve detection accuracy. Furthermore, to ensure the reliability of our analysis, we employed three different de novo assembly tools and a mapping method to validate the generated contig.

In our study, we applied metagenomic analysis to examine viral presence in C. colocynthis, a wild plant species extensively cultivated in southern Iran. Building upon a previous research which identified WMV from bitter apple plants in the Jiroft region, our study aimed to explore a wider range of potential viruses in bitter apple plants showing viral symptoms in the Jiroft region. In addition to WMV, other studies in Iran have detected the presence of Papaya ring spot virus-type W and cucurbit aphid-borne yellows virus in this plant [45]. These findings indicate that this plant species can act as a viral reservoir affecting agricultural crops [4,5,6,7,8].

In current research, through de novo assembly, we identified several contigs with high similarity to known viruses in the genera Polerovirus, Impomovirus, Potyvirus, and Cytorhabdovirus. The species demarcation, as outlined by the ICTV, facilitated the classification of detected isolates. Notably, we identified a novel polerovirus species, BaABYV-IR-1, and documented the first occurrence of the Papaya cytorhabdovirus in Iran. These findings add to global inventory of recently identified Polerovirus and Cytorhabdovirus species [28,29,30,31,32,33]. The advancements in molecular biology and sequencing technologies has facilitated these discoveries [13]. The BaABYV, belonging to the genus Polerovirus classified within the family Solemoviriade, infects a variety of plant species including dicots and monocots. Its genome comprises a linear, single-stranded RNA containing ORFs 0, 1, 2, 3a, 3, 4 and 5 [3435]. Given that the translated ORFs 0, 1, and 2 of BaABYV-IR-1 exhibit amino acid sequence similarities ranging from 68 to 88% with publicly available viral species in the family Luteovoridae. By meeting the ICTV threshold, which necessitates over 10% divergence in amino acid sequences of any gene product for special delineation [36], BaABYV-IR-1 has been classified as a new virus species. This isolate has been deposited in the GenBank database under the accession number OR266512. Further investigations into the metagenomics data revealed the presence of three additional virus species.

The BLAST analysis of contig C2 demonstrates a remarkably high level of similarity, as per the taxonomic criteria defined by ICTV. Such findings conclusively categorize it within the genus Ipomovirus, pinpointing it as the SqVYV. The only Iranian SqVYV isolate previously recorded in the GenBank database (SqVYV-Ir, accession number KU953950) was notably clustered in the clade predominantly containing USA isolates. This presents an intriguing divergence compared to SqVYV-IR-BA and other isolates originating from the Middle East. Lacking additional published data on SqVYV-Ir precludes further analysis. SqVYV is phylogenetically linked as a sister group to both the Coccinia mottle virus (CocMoV) and Cucumber vein yellowing virus (CVYV). These affiliations are not merely taxonomical but also show up as similar symptoms in the host plants. All three viruses belong to the same genus and share a restricted host range, limited explicitly to plants in the Cucurbitaceae family [37]. This isolate was submitted to GenBank with the accession number OR232212.

Analysis of contig C3 revealed its relationship with the Potyvirus subgroup, elucidating its identification as a WMV isolate. Following the ICTV guidelines, the WMV-IR-BA isolate, with an identity exceeding 90% with existing WMV isolates, qualifies as a member of the WMV species within the family Potyviridae [3839] This isolate has also been submitted to GenBank under the accession number OR345349. The current study has undertaken the first examination of both Iranian isolates, SqVYV-IR-BA and WMV-IR-BA, regarding their phylogenetics and recombination attributes.

Cytorhabdoviruses are enveloped viruses with single-stranded, negative RNA genomes that infect a range of hosts, including plants, animals, and insects [40,41,42]. The genome typically spans approximately 12.2 to 14.5 kb and encodes a variety of proteins, including the structural proteins (nucleocapsid and envelope), enzymes (RNA polymerase and ribonucleoprotein), and accessory proteins (such as movement proteins or virulence factors) [40, 4344]. These viruses encapsulate their RNA within the nucleocapsid protein, which is then surrounded by a host-derived lipid membrane to complete virus particle formation [43, 45]. The analysis of contig C-4, designated as CiaRV-IR-BA, revealed it contains five major ORFs typical of rhabdoviruses, coding for nucleoprotein (N), phosphoprotein (P), a putative movement protein (P3), hypothetical protein (P4), matrix (M), glycoprotein (G), and an RNA-dependent RNA polymerase (L) [44]. Species demarcation within the genus Cytorhabdovirus is based on genome sequence identity below 75% and amino acid sequence identity under 80% across all cognate ORFs [43]. In the case of CiaRV-IR-BA, nearly all ORFs are more than 85.5% similar to existing CiaRV sequences. This suggests that CiaRV-IR-BA can be considered an isolate of the species Papaya cytorhabdovirus, as proposed by Zhang et al. [46]. However, the lower identity scores for ORF4 at 73 and 65% are notable, possibly pointing to a unique or fast-evolving protein [4748]. This variability could contribute to differential host specificity, virulence, or other ecologically significant traits [49]. The isolate has been submitted to GenBank with the accession number OR232213.

This research has elucidated the recombination dynamics within the genomes of BaABYV-IR-1 and WMV-IR-BA, emphasizing their potential implications for viral evolution and host adaptation. Recombination detection analysis revealed evidence of recombination in ORF5 of BaABYV-IR-1 genome, a gene that encodes for a movement protein crucial for viral replication and host adaption [50]. This suggests that BaABYV-IR-1 could potentially expand its host and vector range, a phenomenon observed in other poleroviruses, like the Soybean chlorotic leafroll virus (SbCLRV) [51]. The recombinant region spans positions 4,166 to 4,890 within the BaABYV-IR-1 genome, indicating a genetic exchange between Brassica yellows virus (BrYV) and Pepper vein yellows virus (PeVYV) as the major and minor parent, respectively. For WMV-IR-BA, the pinpointed recombination event within cistron NIb-CP, validates earlier studies labeling this region as a recombination hotspot [52]. This could have significant implications for the ability of the virus to adapt to new hosts and environments. Our study also examined recombination in SqVYV isolates. Remarkably, while SqVYV-IR-BA did not display recombination itself, it acted as a major parent in a recombination event in the P1b region for an isolate from Iraq, challenging the previously observed trend of high recombination rates in American isolates within P1a region [53]. The observed lower genetic diversity within SqVYV is an irregularity among ipomoviruses [53], potentially is attributable to negative selection pressures or a genetic bottleneck event similar to the evolutionary pattern of cucumber yellow vein virus in Spain [54]. In contrast to these findings, the CiaRV-IR-BA isolate did not exhibit any recombination nor it serve as a parent in any recombinant forms, hinting at a possibly stable evolutionary path that may be due to limited genetic diversity or host specificity. Our study not only sheds light on the recombination patterns of various viruses but also raises important questions about the evolutionary mechanisms that drive these events. The absence of recombination in CiaRV-IR-BA, for instance, could be indicative of a stable evolutionary lineage or could suggest that the virus has not yet been exposed to conditions that facilitate recombination [55,56,57]. These findings underscore the need for further research to understand the ecological and evolutionary dynamics that influence viral recombination, host adaptation, and the emergence of new viral strains.

Future research and implications

Our study provides robust validation of specific viruses in the bitter plant samples, thanks to the use of advanced techniques and cross-validation methods. This underscore the reliability of high-throughput sequencing methods like RNA-Seq in virological studies and paves the way for targeted interventions and a deeper understanding of virus-plant interactions. Given the strong amino acid sequence similarity and robust RNA-Seq data validation via RT-PCR, further research is essential. While, our study confirmed the presence of specific viruses using advanced techniques, it also highlighted several areas for future investigations. The limited geographic scope and sample size (n = 7) point to the need for more extensive research to achieve broader and more generalizable conclusions. Future studies should consider a larger sample size and geographic range, particularly beyond the Jiroft region.

A notable finding was the common viral symptoms such as dwarfing, leaf crinkling, and mild chlorosis observed in our samples, further complicated by mixed infections. This underscores the need for further research focused on isolating individual viruses and conducting biological assays to understand the role of each virus and its interaction with bitter apple plants. Additionally, the potential impact of these viruses on the medicinal properties of bitter apple remains an unexplored area of research.

The observed proximity of bitter apple to citrus trees in the Jiroft region, raises questions about the transmission of Citrus-associated rhabdovirus (CiaRV) to bitter apple plants, possibly through common vectors. While various insects like aphids, planthoppers, and leafhoppers are known to transmit cytorhabdoviruses [58], and whiteflies have been identified as vectors for Bean-associated cytorhabdovirus [59], the specific vector responsible for CiaRV transmission remains unidentified. Our observations suggest a potential transmission risk, but more research is needed to confirm this and to understand the virus transmission dynamics in this agroecosystem.

Additionally, our study is temporal scope offers opportunities for future research. We plan to investigate the viral community in bitter apple plants across different seasons and time periods, considering their potential as annual or perennial. This will be crucial for understanding seasonal changes in viral prevalence, which is vital for developing effective disease management strategies. The influence of local climate, marked by mild winters and hot, humid summers, on virus prevalence should also be considered in these studies.

Data availability

Sequencing data generated in this study have been deposited in the NCBI Sequence Read Archive database under accessions PRJNA1005066.

Abbreviations

NGS:

Next generation Sequencing

ICTV:

International Committee on Taxonomy of Viruses

MSAs:

Multiple sequence alignments

MLtree:

Maximum likelihood phylogenetic tree

RT:

Reverse Transcription

NCBI:

National Center for Biotechnology Information

RVDB:

Reference Viral DataBase

SqVYV:

Squash vein yellowing virus

CiaRV:

Citrus-associated rhabdovirus

BaABYV:

Bitter apple aphid-borne yellows virus

WMV:

Watermelon mosaic virus

CABYV:

Cucurbit aphid-borne yellows virus

SqMV:

Squash mosaic virus

PRSV-W:

Papaya ring spot virus-type W

CMV:

Cucumber mosaic virus

CocMoV:

Coccinia mottle virus

CVYV:

Cucumber vein yellowing virus

ZYMV:

Zucchini yellow mosaic virus

PABYV:

Pepo aphid-borne yellows virus

PuPV:

Pumpkin polerovirus

GEV:

Groundnut enation virus

BrYV:

Brassica yellows virus

PeVYV:

Pea enation mosaic virus

LMV:

Lettuce mosaic virus

BCMV:

Bean common mosaic virus

References

  1. Chao S, Wang H, Zhang S, Chen G, Mao C, Hu Y, Yu F, Wang S, Lv L, Chen L, Feng G. Novel RNA viruses discovered in weeds in Rice Fields. Viruses. 2022;14(11):2489.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wang Z, Hu H, Goertzen LR, McElroy JS, Dane F. Analysis of the Citrullus colocynthis transcriptome during water deficit stress. PLoS ONE. 2014;9(8):e104657.

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  3. Sanei M, Roozafzai F, Rostami Abousaidi S. Citrullus colocynthis: the most suggested herb in persian medicine for management of low-back pain. Res j Pharmacogn. 2020;7(1):79–86.

    Google Scholar 

  4. Naeimifar M, Pourrahim R, Zadehdabagh G. Natural infection of Citrullus colocynthis by papaya ringspot virus-W in Iran. Plant Dis. 2014;98(12):1748–8.

    Article  CAS  PubMed  Google Scholar 

  5. Vafaei SH, Mahmoodi M. Presence of recombinant strain of Cucurbit aphid borne yellows virus in Iran. Iran J Biotechnol. 2017;15(4):289.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Massumi H, Samei A, Hosseini-Pour A, Shaabanian M, Rahimian H. Occurrence, distribution, and relative incidence of seven viruses infecting greenhouse-grown cucurbits in Iran. Plant Dis. 2007;91(2):159–63.

    Article  PubMed  Google Scholar 

  7. Massumi H, Shaabanian M, Heydarnejad J, Hosseini Pour A, Rahimian H. Host range and phylogenetic analysis of Iranian isolates of Zucchini yellow mosaic virus. Plant Pathol. 2011:187–93.

  8. Sharifi M, Massumi H, Heydarnejad J, Hosseini Pour A, Shaabanian M, Rahimian H. Analysis of the biological and molecular variability of watermelon mosaic virus isolates from Iran. Virus Genes. 2008;37:304–13.

    Article  CAS  PubMed  Google Scholar 

  9. Hasiów-Jaroszewska B, Boezen D, Zwart MP. Metagenomic studies of viruses in weeds and wild plants: a powerful approach to characterise variable virus communities. Viruses. 2021;13(10):1939.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Zuckerman NS, Shulman LM. Next-Generation Sequencing in the Study of Infectious diseases. Infectious diseases. New York, NY: Springer US; 2023. pp. 35–56.

    Chapter  Google Scholar 

  11. Kasem S, Rice N, Henry RJ. DNA extraction from plant tissue. InPlant genotyping II: SNP technology. Wallingford UK: CABI; 2008. pp. 219–71.

    Google Scholar 

  12. Maree HJ, Fox A, Al Rwahnih M, Boonham N, Candresse T. Application of HTS for routine plant virus diagnostics: state of the art and challenges. Front. Plant Sci. 2018;9:1082.

    Google Scholar 

  13. Villamor DE, Ho T, Al Rwahnih M, Martin RR, Tzanetakis IE. High throughput sequencing for plant virus detection and discovery. Phytopathology. 2019;109(5):716–25.

    Article  CAS  PubMed  Google Scholar 

  14. Roossinck MJ, Martin DP, Roumagnac P. Plant virus metagenomics: advances in virus discovery. Phytopathology. 2015;105(6):716–27.

    Article  CAS  PubMed  Google Scholar 

  15. Andrews S. FastQC: a quality control tool for high throughput sequence data. 2010;370.

  16. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet j. 2011;17(1):10–2.

    Article  Google Scholar 

  17. Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, Adiconis X, Fan L, Raychowdhury R, Zeng Q, Chen Z. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat Biotechnol. 2011;29(7):644–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, Lesin VM, Nikolenko SI, Pham S, Prjibelski AD, Pyshkin AV. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19(5):455–77.

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sedlazeck FJ, Rescheneder P, Von Haeseler A. NextGenMap: fast and accurate read mapping in highly polymorphic genomes. Bioinformatics. 2013;1(21):2790–1.

    Article  Google Scholar 

  20. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25(16):2078–9. 1000 Genome Project Data Processing Subgroup.

  21. Katoh K, Standley DM. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol. 2013;30(4):772–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Martin DP, Murrell B, Golden M, Khoosal A, Muhire B. RDP4: detection and analysis of recombination patterns in virus genomes. Virus Evol. 2015;1(1):vev003.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Nguyen LT, Schmidt HA, Von Haeseler A, Minh BQ. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol. 2015;32(1):268–74.

    Article  CAS  PubMed  Google Scholar 

  24. Kumar S, Stecher G, Li M, Knyaz C, Tamura K. MEGA X: molecular evolutionary genetics analysis across computing platforms. Mol Biol Evol. 2018;35(6):1547.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Desbiez C, Lecoq H. The nucleotide sequence of watermelon mosaic virus (WMV, Potyvirus) reveals interspecific recombination between two related potyviruses in the 5′ part of the genome. Arch Virol. 2004;149:1619–32.

    Article  CAS  PubMed  Google Scholar 

  26. Jackson AO, Dietzgen RG, Goodin MM, Bragg JN, Deng M. Biology of plant rhabdoviruses. Annu Rev Phytopathol. 2005;43:623–60.

    Article  CAS  PubMed  Google Scholar 

  27. Roossinck MJ. Deep sequencing for discovery and evolutionary analysis of plant viruses. Virus Res. 2017;239:82–6.

    Article  CAS  PubMed  Google Scholar 

  28. Peng B, Kang B, Wu H, Liu L, Liu L, Fei Z, Hong N, Gu Q. Detection and genome characterization of a novel member of the genus Polerovirus from zucchini (Cucurbita pepo) in China. Arch Virol. 2019;164:2187–91.

    Article  CAS  PubMed  Google Scholar 

  29. Koeda S, Homma K, Kamitani M, Nagano AJ, Taniguchi M, Pohan N, Kesumawati E. Pepper vein yellows virus 9: a novel polerovirus isolated from Chili pepper in Indonesia. Arch Virol. 2020;165:3017–21.

    Article  PubMed  Google Scholar 

  30. Tokuda R, Watanabe K, Koinuma H, Okano Y, Nijo T, Yamamoto T, Suzuki M, Maejima K, Namba S, Yamaji Y. Complete genome sequence of a novel polerovirus infecting Cynanchum Rostellatum. Arch Virol. 2023;168(2):57.

    Article  CAS  PubMed  Google Scholar 

  31. Wu Y, Yang M, Yang H, Qiu Y, Xuan Z, Xing F, Cao M. Identification and molecular characterization of a novel cytorhabdovirus from rose plants (Rosa chinensis Jacq). Arch Virol. 2023;168(4):118.

    Article  CAS  PubMed  Google Scholar 

  32. Belete MT, Kim SE, Igori D, Ahn JK, Seo HK, Park YC, Moon JS. Complete genome sequence of daphne virus 1, a novel cytorhabdovirus infecting Daphne Odora. Arch Virol. 2023;168(5):141.

    Article  CAS  PubMed  Google Scholar 

  33. Medberry AN, Srivastava A, Diaz-Lara A, Rwahnih MA, Villamor DE, Tzanetakis IE. A Novel, divergent member of the Rhabdoviridae family infects strawberry. Plant Dis. 2023;107(3):620–3.

    Article  CAS  PubMed  Google Scholar 

  34. Krueger EN, Beckett RJ, Gray SM, Miller WA. The complete nucleotide sequence of the genome of Barley yellow dwarf virus-RMV reveals it to be a new Polerovirus distantly related to other yellow dwarf viruses. Front Microbiol. 2013;4:205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sõmera M, Sarmiento C, Truve E. Overview on sobemoviruses and a proposal for the creation of the family Sobemoviridae. Viruses. 2015;7(6):3076–115.

    Article  PubMed  PubMed Central  Google Scholar 

  36. https://ictv.global/report/chapter/solemoviridae/solemoviridae/polerovirus. Accessed 30 Aug 2023.

  37. Desbiez C, Verdin E, Tepfer M, Wipf-Scheibel C, Millot P, Dafalla G, Lecoq H. Characterization of a new cucurbit-infecting ipomovirus from Sudan. Arch Virol. 2016;161(10):2913–5.

    Article  CAS  PubMed  Google Scholar 

  38. Adams MJ, Antoniw JF, Fauquet CM. Molecular criteria for genus and species discrimination within the family Potyviridae. Arch Virol. 2005;150:459–79.

    Article  CAS  PubMed  Google Scholar 

  39. Inoue-Nagata AK, Jordan R, Kreuze J, Li F, López-Moya JJ, Mäkinen K, Ohshima K, Wylie SJ, ICTV Report Consortium. ICTV virus taxonomy profile: Potyviridae 2022. J Gen Virol. 2022;103(5):001738.

    Article  CAS  Google Scholar 

  40. Kuzmin IV, Novella IS, Dietzgen RG, Padhi A, Rupprecht CE. The rhabdoviruses: biodiversity, phylogenetics, and evolution. Infect Genet Evol. 2009;9(4):541–53.

    Article  CAS  PubMed  Google Scholar 

  41. Kuzmin IV, Walker PJ. Vector-borne rhabdoviruses. Arboviruses: Mol Biol Evol. 2016;71–88.

  42. Walker PJ, Dietzgen RG, Joubert DA, Blasdell KR. Rhabdovirus accessory genes. Virus Res. 2011;162(1–2):110–25.

    Article  CAS  PubMed  Google Scholar 

  43. Walker PJ, Blasdell KR, Calisher CH, Dietzgen RG, Kondo H, Kurath G, Longdon B, Stone DM, Tesh RB, Tordo N, Vasilakis N. ICTV virus taxonomy profile: Rhabdoviridae. J Gen Virol. 2018;99(4):447–8.

    Article  CAS  PubMed  Google Scholar 

  44. Walker PJ, Firth C, Widen SG, Blasdell KR, Guzman H, Wood TG, Paradkar PN, Holmes EC, Tesh RB, Vasilakis N. Evolution of genome size and complexity in the Rhabdoviridae. PLoS Pathog. 2015;11(2):e1004664.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Jayakar HR, Jeetendra E, Whitt MA. Rhabdovirus assembly and budding. Virus Res. 2004;106(2):117–32.

    Article  CAS  PubMed  Google Scholar 

  46. Zhang S, Huang A, Zhou X, Li Z, Dietzgen RG, Zhou C, Cao M. Natural defect of a plant rhabdovirus glycoprotein gene: a case study of virus–plant coevolution. Phytopathology. 2021;111(1):227–36.

    Article  CAS  PubMed  Google Scholar 

  47. Hughes AL. Adaptive evolution of genes and genomes. Oxford University Press; 1999.

  48. Nei M, Kumar S. Molecular Evolution and Phylogenetics. Oxford University Press; 2000.

  49. Woolhouse ME, Webster JP, Domingo E, Charlesworth B, Levin BR. Biological and biomedical implications of the co-evolution of pathogens and their hosts. Nat Genet. 2002;32(4):569–77.

    Article  CAS  PubMed  Google Scholar 

  50. LaTourrette K, Holste NM, Garcia-Ruiz H. Polerovirus genomic variation. Virus Evol. 2021;7(2):veab102.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Xu T, Lei L, Fu Y, Yang X, Luo H, Chen X, Wu X, Wang Y, Jia MA. Molecular characterization of a Novel Polerovirus infecting soybean in China. Viruses. 2022;14(7):1428.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Verma RK, Mishra M, Marwal A, Gaur RK. Identification, genetic diversity and recombination analysis of watermelon mosaic virus isolates. 3 Biotech. 2020;10:1–8.

    Article  Google Scholar 

  53. Webster CG, Adkins S. Low genetic diversity of squash vein yellowing virus in wild and cultivated cucurbits in the US suggests a recent introduction. Virus Res. 2012;163(2):520–7.

    Article  CAS  PubMed  Google Scholar 

  54. García-Arenal F, Fraile A, Malpica JM. Variability and genetic structure of plant virus populations. Annu Rev Phytopathol. 2001;39(1):157–86.

    Article  PubMed  Google Scholar 

  55. Simon-Loriere E, Holmes EC. Why do RNA viruses recombine? Nat Rev Microbiol. 2011;9(8):617–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Martin DP, Biagini P, Lefeuvre P, Golden M, Roumagnac P, Varsani A. Recombination in eukaryotic single stranded DNA viruses. Viruses. 2011;3(9):1699–738.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Holmes EC. Evolutionary history and phylogeography of human viruses. Annu Rev Microbiol. 2008;62:307–28.

    Article  CAS  PubMed  Google Scholar 

  58. Whitfield AE, Huot OB, Martin KM, Kondo H, Dietzgen RG. Plant rhabdoviruses—their origins and vector interactions. Curr Opin Virol. 2018;33:198–207.

    Article  CAS  PubMed  Google Scholar 

  59. Pinheiro-Lima B, Pereira-Carvalho RC, Alves-Freitas DM, Kitajima EW, Vidal AH, Lacorte C, Godinho MT, Fontenele RS, Faria JC, Abreu EF, Varsani A. Transmission of the bean-associated cytorhabdovirus by the whitefly Bemisia tabaci MEAM1. Viruses. 2020;12(9):1028.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by Research and Technology Institute of Plant Production (RTIPP), Shahid Bahonar University of Kerman, Kerman, Iran. The researchers are grateful to Dr. S.M. Mirtadzaddini in the Department of Biology, Faculty of Science, in Shahid Bahonar University of Kerman for his invaluable assistance in the plant identification.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

SG as a Ph.D student all tests in the lab was done by her and writing draft paper. HM as a supervisor monitoring all test and writing the paper. SHF as a advisor monitoring about phylogentic and recombination analysis and also writing. MM as a advisor monitoring on the analysis of dataset in RNA sequencing. JH as a advisor monitoring on the writing of the paper. AH monitoring on the writing of the paper. All authors reviewed the manuscript.

Corresponding author

Correspondence to Hossein Massumi.

Ethics declarations

Ethics approval and consent to participate

This study did not include using any animals, human or otherwise, and therefore did not require ethical approval.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

: Metatranscriptome analysis of symptomatic bitter apple plants revealed mixed viral infections with a putative novel polerovirus

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ghorani, S., Massumi, H., Farhangi, S.H. et al. Metatranscriptome analysis of symptomatic bitter apple plants revealed mixed viral infections with a putative novel polerovirus. BMC Genomics 25, 181 (2024). https://doi.org/10.1186/s12864-024-10057-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12864-024-10057-z

Keywords