Skip to main content

Analysis of proteomic changes in cassava cv. Kasetsart 50 caused by Sri Lankan cassava mosaic virus infection

Abstract

Background

Sri Lankan cassava mosaic virus (SLCMV) is a plant virus causing significant economic losses throughout Southeast Asia. While proteomics has the potential to identify molecular markers that could assist the breeding of virus resistant cultivars, the effects of SLCMV infection in cassava have not been previously explored in detail.

Results

Liquid Chromatography-Tandem Mass Spectrometry (LC/MS–MS) was used to identify differentially expressed proteins in SLCMV infected leaves, and qPCR was used to confirm changes at mRNA levels. LC/MS–MS identified 1,813 proteins, including 479 and 408 proteins that were upregulated in SLCMV-infected and healthy cassava plants respectively, while 109 proteins were detected in both samples. Most of the identified proteins were involved in biosynthetic processes (29.8%), cellular processes (20.9%), and metabolism (18.4%). Transport proteins, stress response molecules, and proteins involved in signal transduction, plant defense responses, photosynthesis, and cellular respiration, although present, only represented a relatively small subset of the detected differences. RT-qPCR confirmed the upregulation of WRKY 77 (A0A140H8T1), WRKY 83 (A0A140H8T7), NAC 6 (A0A0M4G3M4), NAC 35 (A0A0M5JAB4), NAC 22 (A0A0M5J8Q6), NAC 54 (A0A0M4FSG8), NAC 70 (A0A0M4FEU9), MYB (A0A2C9VER9 and A0A2C9VME6), bHLH (A0A2C9UNL9 and A0A2C9WBZ1) transcription factors. Additional upregulated transcripts included receptors, such as receptor-like serine/threonine-protein kinase (RSTK) (A0A2C9UPE4), Toll/interleukin-1 receptor (TIR) (A0A2C9V5Q3), leucine rich repeat N-terminal domain (LRRNT_2) (A0A2C9VHG8), and cupin (A0A199UBY6). These molecules participate in innate immunity, plant defense mechanisms, and responses to biotic stress and to phytohormones.

Conclusions

We detected 1,813 differentially expressed proteins infected cassava plants, of which 479 were selectively upregulated. These could be classified into three main biological functional groups, with roles in gene regulation, plant defense mechanisms, and stress responses. These results will help identify key proteins affected by SLCMV infection in cassava plants.

Peer Review reports

Background

After rice and maize, cassava (Manihot esculenta Crantz) is the third most widely cultivated crop in the world [1]. Apart from providing food, cassava is also used in the production of starch-based products, animal feed, ethanol, and biofuel. In Southeast Asia [2], cassava is grown as an industrial crop by more than 2 million households. In Thailand alone, 1.28 million hectares of farmland is used for the cultivation of this crop, providing work for more than 600,000 families in 50 provinces. Typically, the country produces 28–30 million tons of casava annually, and demand can exceed 40 million tons [3]. Cassava serves as food and it is also used in the production of starch-based products, animal feed, ethanol, and biofuel. Cultivar Kasetsart 50 (KU50) is one of the most important cassava cultivars in the world, cultivated in several Asian countries. In 2009, the adaptation of KU50 reached its peak; farmers grew this cultivar on 718,400 hectares of land, 52% of this being in Thailand. Its excellent yield, high starch content, good gemination and vigorous growth explain the increasing popularity of this cultivar. It also adapts well to different Agro-ecological zones [4, 5]. Unfortunately, in recent years the production of this valuable crop has been affected by pests and pathogens, reducing productivity.

Cassava mosaic disease (CMD) is the most important plant pest in African and Asia [6]. CMD first emerged in Southeast Asia in 2015 [2, 7]. Currently most CMD outbreaks are reported in mainland SEA, including Cambodia, Vietnam, Thailand, and Laos [7,8,9,10,11]. Uke, et al. reported that cassava root yield decreased by 16–33% and starch content by 22–28%, after infected cuttings were planted in Vietnam [10]. To date, all CMD outbreaks in SEA have been caused by a single viral species, Sri Lankan cassava mosaic virus (SLCMV) [12]. Although outbreaks affected SEA for 8 years, currently there are no commercially available SLCMV resistant cassava cultivars. Nonetheless, field trials and greenhouse testing indicated that KU50 showed some tolerance to CMD, with only mildly symptomatic infections and slow disease progression resulting in better preserved yields compared to other commercial cassava varieties [13, 14]. Consequently, the Department of Agriculture Extension, Thailand, has approved KU50 as an acceptable substitute for susceptible cassava varieties until more CMD resistant varieties are developed. SLCMV is a member of cassava mosaic geminiviruses (family Geminiviridae, genus Begomovirus). SLCMV is one of eleven cassava mosaic geminivirus species [15] that are primarily transmitted by a whitefly vector (Bemisia tabaci), although vegetative propagation also occurs, through the plantation of infected stems [16]. It appears, that the practice of vegetative cassava propagation is the main factor in disease spread in SEA.

Proteomics studies analyze proteins after post-translational modifications (PTMs), providing vital insight into biological functions [17]. PTMs are crucial for plant growth, development, and responses to biotic and abiotic stresses. As PTMs cannot be detected by genome sequencing or transcriptional analysis, proteomics has great utility in understanding the importance of these changes [18,19,20,21]. Proteomics approaches are also instrumental in studying interactions between plants and pathogens at the molecular level, providing insights into protein networks and pathways [22]. Thus, the approach holds great promise in improving our understanding of molecular responses in CMV infected cassava plants.

Diverse proteomic approaches have been utilized in the study of the interaction of plants and viral pathogens. Casado-Vela et al. [23] identified protein profiles involved in the defense of tomato plants against Tobacco mosaic virus (TMV). This work detected peptidases, endoglucanases, chitinases, proteins associated with the ascorbate–glutathione cycle, pathogenesis-related (PR) proteins, and antioxidant enzymes involved in antiviral plant responses. The comparison of tomato cultivars resistant and susceptible to Tomato yellow leaf curl virus (TYLCV) identified 86 differentially expressed proteins and classified them into seven functional groups involved in photosynthesis, protein metabolism, carbohydrate metabolism, energy and amino acid metabolism, detoxification and antioxidation, signal transduction, and chaperone roles. Proteomics analysis has also been used in cassava, to explore responses to abiotic stresses such as drought. However, only two proteomics studies have been performed to study CMV infection. Raghu et al. [24] identified 19 differentially expressed proteins from cassava leaves infected with ICMV and SLCMV using 2D gel electrophoresis and matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF) mass spectrometry. Peptide Mass Fingerprint (PMF) analysis indicated that eighteen of these proteins were relevant to plant growth and development and plant defense. A recent report, describing proteome mapping comparing plants susceptible or resistant to South African cassava mosaic virus (SACMV) identified 2,682 differentially expressed proteins during the systemic stage of the infection, and 2,817 differences during the stages of recovery. The key finding of that study was identifying the role of RPL10 protein in NIK1-mediated effector function, triggering plant immunity against the geminivirus infection [25].

Because in Thailand cassava is mostly propagated vegetatively, the use of potentially infected stems during this practice is the primary cause of CMV spread, and farmers also use this approach to replace other cassava cultivars with KU50. Despite previous proteomics studies investigating cassava leaves infected with ICMV and SLCMV, there is no available data on how infected KU50 plants respond to the virus. The objective of this study is to compare the proteome of healthy and infected KU50 leaves to identify changes induced by SLCMV infection. By identifying differentially expressed proteins during the infection, this study contributes significantly to our understanding of molecular mechanisms involved in the pathogenesis of SLCMV infection in cassava.

Materials and methods

Sample collection

Healthy and SLCMV infected cassava cv. KU50 stems were vegetatively propagated. The planting materials were supported by which obtained from Thai Tapioca Development Institute (TTDI), Nakhon Ratchasima, Thailand. The stems were cut into 15 cm lengths, containing 3–4 buds each, and planted in 20-cm diameter plastic pots in the green house of the Department of Plant Pathology, Faculty of Kasetsart University, Thailand. After 45 days of cultivation, the apex of healthy and infected stems was collected, pooling leaves from three plants into one sample. To stop all metabolic activity, the leaves were immediately flash frozen in liquid nitrogen, and the frozen samples were stored at -80 °C until protein extraction and nucleic acid extraction.

CMV detection by PCR

DNA was extracted from the frozen cassava leaves using the CTAB method [26]. Cassava tissues were ground to a powder in liquid nitrogen and 700 µL CTAB buffer was added to 200 mg of ground leaf power. This suspension was incubated at 65 °C for 30 min, then 700 μL chloroform:isoamyl alcohol mixture (24:1) was added to the tubes. DNA was precipitated by the addition of 700 μL isopropanol, and the tubes were incubated at -20 °C for 3 h. The pelleted DNA was washed twice in 70% ethanol and the pellet was dried at room temperature. The DNA was resuspended in ddH2O containing 100 μg/ml RNase (Thermo Fisher Scientific, Waltham, MA, USA) and stored at -20 °C. DNA samples were quantitated and checked for integrity by agarose gel electrophoresis and a Nanodrop spectrophotometer (NanoDrop Technologies, Thermo Scientific).

To identify infected plants, the AV1 gene of SLCMV was amplified using gene specific primers (forward: 5’-GTT GAA GGT ACT TAT TCC C-3’ and reverse: 5’-TAT TAA TAC GGT TGT AAA CGC-3’) [27]. Amplification was performed in a 25-μL reaction volume containing 1 × PCR buffer (PCR Biosystems, London, UK), 0.2 μM each of forward and reverse primers, and 50 ng of the genomic DNA. After an initial denaturation at 94 °C for 5 min 35 cycles were carried out, consisting of denaturation at 94 °C for 40 s, annealing at 55 °C for 40 s, and elongation at 72 °C for 40 s. The final elongation was carried out at 72 °C for 5 min. Amplified products were examined by gel electrophoresis on a 1.5% agarose TAE gel containing RedSafe Nucleic Acid Staining Solution (iNtRON Biotechnology, Sangdaewon, South Korea). Sanger sequencing was used to establish the sequence of the amplified product (Macrogen, The Netherlands), and the identity of the virus was confirmed by BLAST searches in the National Center for Biotechnology Information database (https://blast.ncbi.nlm.nih.gov/Blast.cgi).

Protein extraction

Cassava tissues were ground to a fine powder in liquid nitrogen, then 1 ml of 0.5% Sodium dodecyl sulfate was added to 100 mg tissue powder. After agitating for 1 h, tubes were centrifuged at 9,000 g for 15 min. The supernatant was transferred to fresh tube, mixed with 1,400 μL of cold acetone, and incubated at -20 °C. The mixture was centrifuged at 9,000 g for 15 min and the supernatant was discarded. The protein pellet was dried and stored at -80 °C.

Determination of protein concentration using the lowry method

The pellets were resuspended in 0.5% SDS and the protein concentration was determined by the Lowry method [28]. The absorbance at 750 nm (OD750) was measured and the protein concentration was calculated using a standard curve established using a serial dilution of BSA.

In solution digestion

Samples containing 5 µg of total protein were subjected to in-solution protease digestion. Samples were dissolved in 10 mM ammonium bicarbonate (AMBIC) buffer. Disulfide bonds were reduced by adding dithiothreitol (DTT) to a final concentration of 5 mM, followed by an incubation at 60ºC for 1 h. Finally, sulfhydryl groups were alkylated by incubating the samples for 45 min in AMBIC buffer containing 15 mM iodoacetamide, at room temperature, in the dark [29]. Next, samples were mixed with 50 ng/µL of sequencing grade trypsin (1:20 ratio) (Promega, Germany) and incubated at 37ºC overnight, to digest the protein content. Prior to LC–MS/MS analysis, the digested samples were dried and protonated with 0.1% formic acid.

Protein quantification

Tryptic peptide samples were prepared for injection into an Ultimate3000 Nano/Capillary LC System (Thermo Scientific, UK) coupled to a Hybrid quadrupole Q-Tof impact II™ (Bruker Daltonics) equipped with a Nano-captive spray ion source. Briefly, 1µL of peptide digest was enriched on a µ-Precolumn 300 µm i.d. X 5 mm C18 Pepmap 100, 5 µm, 100 A (Thermo Scientific, UK), separated on a 75-µm I.D. × 15 cm column packed with Acclaim PepMap RSLC C18, 2 μm, 100 Å, nanoViper (Thermo Scientific, UK). The C18 column was enclosed in a thermostatic oven set to 60 °C. Solvent A was 0.1% formic acid in water, solvent B was 0.1% formic acid in 80% acetonitrile. A gradient of 5–55% solvent B was used to elute the peptides at a constant flow rate of 0.30 µL/min over 30 min. Electrospray ionization was carried out at 1.6 kV using CaptiveSpray. Nitrogen was used as drying gas (flow rate approximately 50 L/h). Collision-induced-dissociation (CID) product ion mass spectra were obtained using nitrogen gas as the collision gas. Mass spectra [30] and MS/MS spectra were obtained in the positive-ion mode at 2 Hz over the range (m/z) 150–2200. The collision energy was adjusted to 10 eV as a function of the m/z value. The LC–MS analysis of each sample was performed in triplicate. The protein spectral data obtained in this study has been deposited at ProteomeXchange: PXD035792.

Bioinformatics and data analysis

The MaxQuant software package (version 2.0.3.0) was used to quantitate proteins in individual samples using the Andromeda search engine. This identifies peptide fragments by comparing MS/MS spectra to data deposited in the UniProt Manihot esculenta database [31]. Label-free quantitation with MaxQuant standard setting was performed allowing a maximum of two miss cleavages and a mass tolerance of 0.6 Daltons during the main search. These searches were set up based on trypsin being the digesting enzyme, carbamidomethylation of cysteine as a fixed modification, and the oxidation of methionine and acetylation of the protein N-terminus being variable modifications. Only peptides peaks containing a minimum of 7 amino acids and representing a single sequence motif were used in further analysis. Proteins were identified if at least two corresponding peptides were detected, with at least one of these being unique. Only proteins identified based on the above criteria were used for data analysis. False discovery rate (FDR) was set at 1% and was estimated by using reversed search sequences. The maximal number of modifications for any given peptide was set to 5.

For FASTA searches, the proteins present in the Manihot esculenta proteome were downloaded from the UniProt database. Potential contaminants, present in the contaminant FASTA file that comes with MaxQuant software, were automatically added to the search space. The MaxQuant ProteinGroups.txt file was loaded into the Perseus search platform (version 1.6.6.0) [31], and potential outliers that did not correspond to any UPS1 protein were removed from the data set. Maximum intensities were log2 transformed and pairwise comparisons were carried out via t-tests. Missing values were imputed in Perseus using a constant value (zero). Data visualization and statistical analyses were conducted using the MultiExperiment Viewer (MeV) of the TM4 software suit [32]. Protein organization and biological function were investigated according to the protein analysis through evolutionary relationships (Panther) protein classification [33] and Venn diagrams were used to visualize differences between protein lists derived from the various samples [34].

qPCR validation of differentially expressed genes

Total RNA was extracted from healthy and SLCMV infected leaf tissues using QIAzol Lysis Reagent (QIAGEN, Hilden, Germany) according to the manufacturer’s protocol. A RevertAid First Stand cDNA Synthesis Kit (ThermoFisher Scientific, USA) was used for first stand cDNA synthesis, primed by Oligo(dT)18, as recommend by the manufacturer. The qPCR reactions were performed using Hot FIREPol® EvaGreen® qPCR Mix Plus (Solis BioDyne,Tartu, Estonia). Reactions contained 10 ng cDNA template, 4 µL 5X HOT FIREPol® EvaGreen® qPCR Mix Plus, 0.2 µM primer mix, and nuclease-free H20 in 20 µL final volume. Amplification was carried out on a CFX Connect Real-Time PCR System (Bio-Rad, USA) using the following cycle parameters: initial denaturation (95ºC for 15 min), followed by 45 cycles of denaturation (95ºC for 30 s), annealing (60ºC for 30 s) and extension (72ºC for 30 s). Fluorescence was recorded at the end of each cycle and a melt curve analysis was carried out between 60ºC-95ºC with a 10 s hold at every 1ºC increment. Each reaction was set up in triplicates. Proteins uniquely upregulated in SLCMV infected cassava samples were analyzed by qPCR experiments. Most of these were associated with gene and protein processing, plant defense, and stress responses. The primers used in these experiments are listed in Supplementary Table S1. The reference gene to assess relative mRNA abundance was UBQ10 [25]. Fold changes in gene expression were analyzed using the log2 (ΔCT) method [35].

Results

SCMV symptoms and virus infection analysis

The characteristic symptoms of SLCMV infection were present in the infected plants after the emergence of the first leaf, with chlorotic mosaics and leaf deformation. PCR experiments using gene specific primers confirmed SLCMV as the causative agent of the visible symptoms. The ~ 900-bp amplicon was only seen when DNA from symptomatic leaves was analyzed (Supplementary Figure S1), and the sequence of the amplified product showed 99% identity with the Sri Lankan cassava mosaic virus isolated from Thailand (Supplementary Figure S2).

Protein identification and quantification

Samples from both healthy and infected plants contained three biotical replicates to reduce random sampling effects and increase the validity of proteins detections. The tandem mass spectra obtained matched the Manihot esculenta genome and protein annotation database. After normalizing spectral counts, the protein components of infected samples were compared with healthy leaves to identify differentially expressed molecules. Log2 change between paired values was calculated by affinity propagation to identify proteins showing significantly altered expression as a result of the infection. In total, 1,813 peptides were identified, 1,064 in the SLCMV infected KU50 samples and 947 in healthy plants. Venn diagram, showing the relationship between the 479 and 408 proteins detected in SLCMV-infected and healthy cassava plant, respectively is shown in Fig. 1. As shown in the Fig. 1, 109 of these proteins were present in both the infected and healthy samples.

Fig. 1
figure 1

Venn diagram showing the distribution and relationships of proteins in healthy and SLCMV infected KU50 plants

Gene Ontology Analysis

Analyzing the gene ontology classification of the detected proteins showed that 36% of the overexpressed proteins were membrane associated, 23.8% localized in the nucleus, and 14.6% to the cytoplasm. In contrast, proteins in healthy plants were up regulated on membranes (40.1%), nucleus (16.9%) and cytoplasm (19.5%). In addition, both samples contained proteins from other cellular components, such as chloroplast, cytosol, Golgi apparatus, mitochondria, microtube, endoplasmic reticulum, peroxisome, and vacuoles. The cellular localization of the detected molecules is shown in Fig. 2A.

Fig. 2
figure 2

Functional classification of differentially expressed proteins in the leaves of healthy or infected cassava cv. KU50. A Proportion of proteins in different functional classes based on cellular localization. B Functional grouping of biological processes. C Proportion of functional groups based on molecular function

In both healthy and SLCMV-infected cassava plants most of the identified proteins were involved in biosynthetic pathways and cellular or metabolic processes. The proportion of proteins associated with plant defense responses rose from 4.1% in healthy plants to 5.7% as a result of the infection, while the proportion of stress response proteins increased from 3.3% to 5.9%. Proteins related to transportation, signaling, photosynthesis, and respiration are shown in Fig. 2B, while those with a role in nucleotide binding, catalytic activity, transporter activity, structural role, and electron carrier activity are summarized in Fig. 2C. Overall, nucleotide binding proteins represented the most commonly identified protein category both in healthy and infected cells (60%). A heat map of differentially expressed proteins within nine biological functional groups is shown in Fig. 3, where a change in color from red to dark green corresponds to differential changes in gene expression abundance. In general, most upregulated proteins in infected KU50 samples were involved in respiration, plant defense, stress responses, metabolic processes, and biosynthetic pathways (Fig. 3).

Fig. 3
figure 3

Heatmap of the leaves of cassava cv. KU50 during SLCMV infection. Heatmaps represent each functional group of differentially expressed proteins a Respiration; b Stress response; c Plant defence response; d Metabolic processes and e Biosynthetic processes

Proteins differentially expressed in response to SLCMV infection

The identified differentially expressed proteins were associated with biosynthesis, plant defense, and stress responses. Within these categories, a subset of replication, transcription, and translation-related proteins were exclusively expressed in SLCMV-infected samples. There were 59 proteins that were overexpressed more than 17-fold in infected samples. As shown in Table 1, 8 of these were involved in replication, 43 in transcription, and 8 in translation. Interestingly, 21 of the most strikingly upregulated proteins were transcription factors, regulating immune mechanisms, stress responses, and the secretion of plant hormones, while another 20 were connected with plant defense responses. Finally, 26 proteins were previously documented to play a role in biotic plant stress. To confirm the findings of the proteomics analysis, we selected 77 differentially expressed proteins for qPCR-based quantitation. Samples were normalized using the expression of the UBQ10 gene in the same cDNA sample. These experiments identified 53 mRNAs that were upregulated in SLCMV infected plants (Fig. 4).

Table 1 Selected differentially expressed protein of SLCMV infected cassava cv. KU50
Fig. 4
figure 4

Validation of differential protein expression levels using RT-qPCR. Result represent the averages from three biological replicates and were normalized against the expression level of the endogenous UBQ10 gene

A total of 29 highly upregulated transcripts were related to biosynthetic processes, including the transcription factors NAC 6, NAC 22, NAC 35, NAC 54, NAC 70, WRKY 77, and WRKY 83. Furthermore, mRNA abundance and protein expression levels showed good correlation for these molecules. Regarding genes that were involved in transcription, translation, and protein synthesis DNA replication ATP-dependent helicase (A0A2C9VTN6) and replication termination factor 2 (A0A2C9UKD1) were also upregulated at transcript levels, showing good correlation with the proteomics data. However, transcripts levels for 16S rRNA m5C967 methyltransferase (A0A2C9UDI4), elongation factor 1-alpha (EF-1-alpha, O49169), HTH myb-type domain-containing protein (A0A2C9WJL4), peptidylprolyl isomerase (A0A251J9E4), and PWWP domain-containing protein (A0A2C9V4D0) were not upregulated at mRNA level, indicating discrepancies between proteomics and transcription data (Fig. 4). Furthermore, proteomics data showed that the WRKY 18 (A0A140H8M2), WRKY 24 (A0A140H8M8) and WRKY 29 (A0A140H8N3) transcription factors were down regulated in SLCMV infection.

qPCR experiments also confirmed changes in the abundance of mRNAs encoding plant defense molecules. The seven upregulated transcripts were cupin type-1 domain (A0A199UBY6), galectin domain (A0A2C9VQZ7), heavy metal-associated domain (HMA domain) (A0A2C9ULD8), Leucine rich repeat N-terminal domain (LRRNT_2 domain) (A0A2C9VHG8), occludin_ELL domain (A0A2C9W2D3), receptor-like serine/threonine-protein kinase (A0A2C9V7G1), and Toll/interleukin-1 receptor/resistance protein domain (TIR domain) (A0A2C9V5Q3). At the same time, AT-hook motif nuclear-localized protein, DCD domain, NB-ARC domain and, protein kinase domain were down-regulated according to qPCR and proteomics analysis (Fig. 4).

The expression of an additional 24 molecules was determined at mRNA level. Fourteen of these, annexin (A0A2C9UX68), catalase (A0A2C9WMD1), HSF_DOMAIN domain (A0A2C9WN68), peptidase_M28 domain (A0A2C9V3F4), peroxidase (A0A2C9UEX5), PMR5N domain (A0A2C9VL88), Rab-GAP TBC domain (A0A251KQD0), reticulon-like protein (A0A2C9VPT1), S-(hydroxymethyl) glutathione dehydrogenase (A0A2C9UNE1), SPRY domain (A0A2C9U854), START domain (A0A2C9WKA1), Thioredoxin domain (A0A2C9WFP1), and BEACH domain (A0A2C9UBJ8) showed elevate transcript abundance, in concordance with LC–MS/MS analysis. However, transcript levels of 1-amino-cyclopropane-1-carboxylic acid oxidase (A0SVL8), alpha-hydroxynitrile lyase (O49893), BAG domain (A0A2C9UMY9), CCHC-type domain (A0A2C9UUL9), Clp R domain (A0A2C9VMF9), formate dehydrogenase (A0A2C9U930), GUB_WAK_binding domain (A0A2C9UKE3), LEA_2 domain (A0A2C9WIP6), S-acyltransferase (A0A2C9WR35), and SHSP domain (A0A2C9V533) were found to be downregulated during qPCR, seemingly contradicting proteomics findings (Fig. 4).

Discussion

We explored the consequences of SLCMV infections in cassava plants at the level of the pant proteome. This approach enabled the documentation of changing protein expression levels in response to the infection and provided valuable insights into the molecular processes that occur in infected plants. Apart from their relevance to basic biological sciences, the findings may provide crucial information to aid the development of SLCMV tolerant commercial cultivars. Developing such resistant plants is critical for disease control and has very high economic significance. This is the first documentation of global changes in the cassava proteome, comparing healthy and SLCMV infected KU50 plants. Subsequent gene ontology analyses highlighted the most likely biological processes altered by this viral infection. During the analysis we paid special attention to proteins that have been implicated in geminivirus infestations in other plants, particularly those molecules involved in gene synthesis, and plant defense and stress responses.

The expression of an extensive subset of differentially expressed proteins was also quantitated using qPCR, including a set of 77 proteins upregulated during SLCMV infection. Out of this fairly comprehensive set of molecules, qPCR confirmed a corresponding increase in mRNA abundance in approximately 70% of the cases. However, proteomics findings and qPCR gene expression data showed contradictory outcomes in the remaining 30%. While this discordance may seem striking, several mechanisms have been reported in the literature that may explain such discrepancies. Post-transcriptional regulatory processes, such as different local translation efficiency and codon bias [36], altered translation elongation rate [37], differences in the half-life of proteins [38], or variations in poly A tail length [39, 40] have all been reported to result in discordant proteomics/transcriptomics results.

According to gene ontology analysis, the majority of differentially expressed proteins in SLCMV infected cassava cv. KU50 plants localized to the cell membrane, nucleus, cytoplasm, and chloroplast. Previous studies indicated that plant defense pathways involved transcriptional and post-transcriptional gene silencing, the ubiquitin-proteasomal degradation pathway, protein kinase signaling cascades, autophagy, and hypersensitive responses [41]. In the nucleus, geminivirus use the enzymatic machinery of the hosts to replicate their initially ssDNA genomes to double stranded forms that interact with histones in the form of minichromosomes. This elicits RNA-directed DNA methylation (RdDM) to suppress the virus via transcriptional gene silencing (TGS) [42,43,44]. Beyond their role in photosynthesis, chloroplasts also play an important role in plant immunity through the regulation of reactive oxygen species (ROS) and salicylic acid (SA) signaling. Chloroplasts also modulate biosynthetic pathways via the synthesis of phytophomones, gibberellic acid (GA), abscisic acid (ABA) and jasmonic acid (JA). During the infection, chloroplasts appear to be one of the major targets of pathogens [45, 46]. The communication between the chloroplast and the nucleus has been explored during biotic stresses. This process, pattern-triggered immunity (PTI) in thylakoid membrane, involves calcium sensing receptors (CAS) inducing the reprogramming of transcriptional activity, SA synthesis, and callose deposition, leading to pathogen resistance [47, 48]. In addition, nuclear transcription factors [45] and several cell surface membrane proteins, including receptor serine threonine kinase (RSTK), Toll/interleukin-1 receptor (TIR), and leucine rich repeat N-terminal domain (LRRNT 2) protein cascades, were reported to play a critical role in plant immunity [49].

Transcription factors (TFs) are keys regulators of mRNA synthesis, acting on cis-regulatory elements in promoter regions. Recent work indicated that TFs in the WRKY, AP2/ERF (Apetala2/Ethylene Responsive Factor), NAC (NAM, ATAF and CUC), MYB, bZIP (Basic leucine zipper domain), bHLH (Basic helix-loop-helix), NF-Y (Nuclear Factor Y), and CAMTA (CaM-binding transcription activator) TF families play a central role in regulating plant immune responses [50, 51]. We detected the upregulation of WRKY 77 and WRKY 83 in SLCMV infected cassava plants. Wei et al. [52] identified a group of Manihot esculenta (Me)WRKY transcription factors, which were shown to be involved in innate immunity, plant defense mechanisms, and biotic stress responses [53]. In agreement with our own observations, Freeborough et al. [54] showed that MeWRKY 83 was up regulated during the late stages of SLCMV infection in a susceptible cassava cultivar (T200). In our experiments, we also detected the overproduction of MeWRKY 83 both at protein and transcript level. Interestingly, a novel cluster of genes containing WRKY TF binding sequences was identified in some plants [55]. Because these genes appear to have a role in plant responses during infections, there is an urgent need to explore WRKY binding domain-containing genes in cassava, as these may aid the identification of CMD resistant variants.

We also detected the upregulation of NAC 6, NAC 35, NAC 22, NAC 54, and NAC 70 transcription factors in SLCMV infected leaves, while in healthy plants NAC genes were expressed at a low background levels. Using high throughput RNA sequencing, a previous study, comparing cassava brown streak virus sensitive and resistant plant variants also detected the increased expression of the members of the NAC family of TFs in resistant cultivars [56]. These TFs play a crucial role in responses to pathogens and abiotic stressors [57]. NAC 22 and NAC 35 were up regulated in response to salt, cold, and osmotic stress, or after treatment with ABA or H2O2 [58]. Our experiments suggest that NAC 54 and NAC 70 are upregulated in a more specific way, after an SLCMV infection. However, the exact role of NAC 54 and NAC 70 during a viral infection is currently unclear and will require further studies.

MYB TFs were also overexpressed during SLCMV infection. In Arabidopsis, the myeloblastosis response to pathogen attacks occurs via the activation of a hypersensitive cell death program, through the brassinosteroid pathway [59]. MYB plays an important role in signal transduction after exposure to SA [60], ABA [61], GA [62], and JA [63]. Recently, Wang et al. [64] reported that MeMYB26 was associated with drought resistance traits and biomass storage. Furthermore, the MeMYB pathway may show a crosstalk with MeWRKYs TFs under condition of stress in cassava [65]. As reported by Ramulifho and Rey [25] ribosomal protein L10 (RPL10) plays role in protecting cassava against geminivirus infections, with a MYB domain-containing TF activating RPL10 transcription in the nucleus. This mechanism inhibited gene translation by the host and had a profound inhibitory effect on geminiviral replication. The authors proposed that the MYB/RPL10 protein network contributed to the symptomatic recovery of the virus resistant TIM3 cassava cultivar after exposure to South African cassava mosaic virus infection. However, while the abundance of MYB protein also showed an increase in response to SLCMV in our studies, we were unable to detect any RPL10. This may indicate that MYB-driven responses may vary according to the strain of geminivirus causing the infection, or the cultivar being tested. Nonetheless, determining MYB and RPL10 expression could be a valuable tool in breeding of CMD resistant cultivars. The fact that RPL10 is encoded on chromosome 12 of cassava and that polymorphisms in this chromosome appear to correlate with CMD2 resistance [66, 67], lends further support to the potential importance of the MYB/RPL10 pathway.

Basic helix-loop-helix (bHLH) proteins are transcription factors representing one of the largest TF families [68]. In Arabidopsis, bHLHs can act both as transcriptional activators or repressors, influencing plant development and physiological processes, such as phytohormone [69] and secondary metabolite synthesis [70, 71]. JA regulates plant stress responses after pathogen attacks and wounding [72, 73]. Wang et al. [74] identified 96 bHLH genes in tomato plans that were either resistant or susceptible to yellow leaf cure virus (TYLCV). In our proteomics data, bHLH was overexpressed and the gene expression was also upregulated by a factor of 2 following SLCMV infection (Table 1 and Fig. 4). However, JA responses after SLCMV infestation or other viral infections will require further studies.

The 60S acidic ribosomal protein P0 (A0A2C9WNN0), 50S ribosomal protein L16 (B1NWI7), ribosomal_S7 domain-containing protein (A0A2C9V9X4), and ribosome biogenesis protein NOP53 (A0A2C9VL95) were also overexpressed in SLCMV infected KU50 (Table 1 and Supplementary Table S1). Several studies have reported the direct interaction of ribosomal proteins with plant viruses during infection. Such interactions were noted in infections caused by Soybean mosaic virus (SMV), Sugarcane mosaic virus (SCMV), Rice yellow mottle virus (RYMV), Papaya meleira virus (PMeV), Pepper mild mottle virus (PMMoV), Rice black-streaked dwarf virus (RBSDV), BNYVV, and Mungbean yellow mosaic India virus (MYMIV) [75, 76]. In addition, our experiments also showed the upregulation of elongation factor 1-alpha (EF-1-alpha) (O49169) and eukaryotic translation initiation factor 3 subunit D (eIF3d) (A0A2C9UXZ5) in response to SLCMV infection (Table 1). Transcription initiation and elongation factors interact with viral particles during viral protein synthesis, and have been shown to be differentially expressed during SCMV, RYMV, PMeV, RBSDV, and MYMIV infections [75,76,77,78,79,80]. Cho et al. [81] investigated how elongation factors and ribosomal proteins interacted with PVX stem loop 1 RNAs. Interestingly, in a resistant rice cultivar, translation elongation factor and RMYV co-purified, indicating particularly strong interactions [81]. Protein synthesis during viral replication and the interactions between viral nucleic acids and host proteins represent a unique field in biology. Our proteomics data provide an early insight into how elongation factor/ ribosomal protein interactions may influence SLCMV infections.

Our experimental data indicated the differential regulation of pathogen defense and stress response proteins, representing 5.9 and 5.7%, of all identified proteins, respectively (Fig. 2). Plants receptor serine threonine kinases (RSTK), Toll/interleukin-1 receptor (TIR), and leucine rich repeat N-terminal domain (LRRNT_2) were significantly upregulated in SLCMV infected plants, confirming the findings of previous studies [82]. The nucleotide-binding site leucine-rich repeat (NBS-LRR) family of receptors, for example receptor serine threonine kinase (RSTK) proteins, are involved in disease resistance and the regulation of plant development, as well as self-versus non-self-discrimination. RSTK is a transmembrane protein that, in associations with other protein families, regulates both disease resistance and developmental processes [83]. Plants respond to pathogen-associated molecular pattern molecules (PAMPs), using extracellular leucine-rich repeat (LRR) receptors for their recognition, serine/threonine kinases (STKs) for down-stream processing, and mitogen-activated protein kinases (MAPKs) for signal transduction. Pattern recognition receptors (PRRs) are activated upon detecting predetermined molecular structures, leading to the initiation of PAMP-triggered immunity (PTI) that can prevent the entry of most invading organisms [84]. Recent research has shown that viruses can both stimulate and repress PTI-like mechanisms [85,86,87,88,89,90,91,92,93,94,95,96,97,98,99]. In some instances, pathogen-derived molecular structures suppress protective mechanisms. This phenomenon is referred to as effector-triggered susceptibility (ETS) [86, 87]. To counteract ETS, a set of intracellular nucleotide-binding leucine-rich repeat (NLR) receptors have evolved, providing effector-triggered immunity [88]. These recognise virulence factors produced by the pathogen in a very precise manner and activate a second phase of defense responses [89].

The Search Tool for Interacting Chemicals (STITCH) algorithm was developed to investigate interactions between proteins and small molecules during biological processes [90]. In our work, we noted that the AT-hook motif nuclear-localized protein (AHL22) directly interacted with histone deacetylase 6 (HDA6), initiating a repression with a significant role in transcriptional regulation (Fig. 5). Previous work indicated that the AHL gene family is necessary for plant growth, development, and stress responses. In particular, AHL22 suppresses hypocotyl growth in Arabidopsis [91]. Taken together, this information suggests that the nuclear localization of an AT-hook motif protein in SLCMV infected cassava may indicate gene silencing. Protein kinase domain-containing protein (AT1G03740) is also involved in gene silencing though its interactions with RNA polymerase (NRPD1B), transcription elongation factor (AT2G34210), and the transcription factors KTF1 and GTA2 TFs (Fig. 5). Protein kinases represent one of the largest protein families in plants. Their central role in environmental stress responses, such as drought, high salinity, cold, and pathogen attacks has been amply demonstrated. In Arabidopsis, the serine/threonine protein kinase, TOUSLED (TSL) is required for antiviral RNA silencing and regulates RNA interference [92]. Wang et al. [93] identified the putative substrates of nine protein kinases that function in plant abiotic and biotic stress responses. This work contains more than 5,000 putative target sites controlling cellular activities. It is hoped that our results will represent the starting point for further studies on the mechanisms underlying plant responses to environmental stresses, including the mechanisms that involve gene silencing.

Fig. 5
figure 5

Prediction of protein induced gene silencing and L-serine metabolic process using STICH analysis

Serine/threonine-protein phosphatases use D-serine while thioredoxin domain-containing protein (TXND9) interacts with L-serine during metabolic processes. Serine metabolism in plant has been studied in the context of photorespiration, where serine is formed from two molecules of glycine. This serine becomes a precursor for glycine, formate, and glycolate accumulation during conditions of stress. Serin/threonine protein phosphatases, especially PP2A, fulfill multiple roles in plant cells during growth and stress rated signaling. Hormone-related signal transduction and oxidative stress signaling regulate the activity of a range of enzymes involved in key metabolic pathways [94]. proposed that this signaling pathway was essential in connecting carbon and nitrogen metabolism, preserving the cellular redox potential, and energy levels in plants under stress. Exposure to exogenous H2O2 replicates some of the consequences of Rhizoctonia cerealis infection by increasing the expression of PP2Ac [96]. There have been numerous studies showing the conflicting roles of Thioredoxins (TRXs) in viral pathogenesis and resistance. In Nicotiana tabacum, TRXh3 confers resistance to Tobacco mosaic virus (TMV) and Cucumber mosaic virus (CMV) [97] and in maize TRX mediates early resistance against the Sugarcane mosaic virus [98]. Using yeast-two-hybrid screening combined with invitro pull-down and biomolecular fluorescent complementation (BiFC) assays, Mathioudakis et al. [99] reported that thioredoxin (TRX) in tomato plants interacted with the pepino mosaic virus (PepMV) coat protein, triple gene block protein (TGBp1), heat shock cognate protein 70, and catalase 1 (CAT1). In our study, STICH analysis predicted the interaction of TRX combined with glutamate synthetase 1 and 2 (GLT1 and 2), potentially affecting glutamate biosynthesis.

Identifying viral and host components associated with infections and understanding the molecular mechanisms of plant defense systems, pathogen virulence, and susceptibility, are key areas of research in plant virology. The results presented here provide an overview of some of the key protein level changes induced by SLCMV infections in cassava plants. A better understanding of the fundamental processes involved in viral infections and host responses is critical for developing and selecting more resistant crops. An ideal defense mechanism would prevent the replication of the virus altogether, eliminating any chance of viral evolution. Additionally, a combinational or additive strategy, targeting the virus and its vector simultaneously, would hinder the infection even more, offering crops more durable protection.

Conclusions

Here we describe the comprehensive proteomics analysis of the leaves of SLCMV infected and healthy cassava KU50 cultivar plants. In the course of the work carried out, more than 1,813 proteins were detected, including 479 that were upregulated in infected plants. These SLCMV induced proteins could be classified into nine functional groups and exhibited a wide range of biological functions. Further studies of the differentially regulated molecules in the cell membrane, nucleus, and chloroplast will help the identification of key proteins involved in cassava-SLCMV infection, potentially aiding the development of resistant crops.

Availability of data and materials

The datasets generated and analyzed in the current study are available in ProteomeXchange; accession number PXD035792 and the data sets supporting the conclusions of this article are included with the article and its additional files.

References

  1. Cock JH, Connor DJ. Chapter 19 - Cassava. In: Sadras VO, Calderini DF, editors. Crop Physiology Case Histories for Major Crops. Academic Press; 2021. p. 588–633.

  2. Chi Y, Pan L-L, Bouvaine S, Fan Y-Y, Liu Y-Q, Liu S-S, et al. Differential transmission of Sri Lankan cassava mosaic virus by three cryptic species of the whitefly Bemisia tabaci complex. Virology. 2020;540:141–9.

    Article  CAS  Google Scholar 

  3. Office Agricultural Economics: Cassava Production Capacity information. In.; 2019.

  4. Legg J. Epidemiology of a Whitefly-Transmitted Cassava Mosaic Geminivirus Pandemic in Africa. In.; 2010. p 233–257.

  5. Praneetvatakul S, Vijitsrikomol K, Kitipadakul P. Beneficial research in cassava of Kasetsart University in research of Kasetsart University that benefit the farmers and society in the occasion of 72th anniversaty. In. Bangkok, Thailand: Kasetsart University; 2015.

  6. Herrera Campo BV, Hyman G, Bellotti A. Threats to cassava production: known and potential geographic distribution of four key biotic constraints. Food Secur. 2011;3:329.

    Article  Google Scholar 

  7. Wang HL, Cui XY, Wang XW, Liu SS, Zhang ZH, Zhou XP. First Report of Sri Lankan cassava mosaic virus Infecting Cassava in Cambodia. Plant Dis. 2016;100:1029.

    Article  Google Scholar 

  8. Minato N, Sok S, Chen S, Delaquis E, Phirun I, Le VX, et al. Surveillance for Sri Lankan cassava mosaic virus (SLCMV) in Cambodia and Vietnam one year after its initial detection in a single plantation in 2015. PLoS ONE. 2019;14: e0212780.

    Article  CAS  Google Scholar 

  9. Leiva AM, Siriwan W, Lopez-Alvarez D, Barrantes I, Hemniam N, Saokham K, et al. Nanopore-Based Complete Genome Sequence of a Sri Lankan Cassava Mosaic Virus Geminivirus Strain from Thailand. Microbiol Resour Announc. 2020;9:e01274-e11219.

    Article  CAS  Google Scholar 

  10. Uke A, Hoat TX, Quan MV, Liem NV, Ugaki M, Natsuaki KT. First Report of Sri Lankan Cassava Mosaic Virus Infecting Cassava in Vietnam. Plant Dis. 2018;102:2669.

    Article  Google Scholar 

  11. Chittarath K, Jimenez J, Vongphachanh P, Leiva AM, Sengsay S, Lopez-Alvarez D, et al. First Report of Cassava Mosaic Disease and Sri Lankan Cassava Mosaic Virus in Laos. Plant Dis. 2021;105:1861.

    Article  Google Scholar 

  12. Siriwan W, Jimenez J, Hemniam N, Saokham K, Lopez-Alvarez D, Leiva AM, et al. Surveillance and diagnostics of the emergent Sri Lankan cassava mosaic virus (Fam. Geminiviridae) in Southeast Asia. Virus Res. 2020;285:197959.

    Article  CAS  Google Scholar 

  13. Hemniam N, Saokham K, Roekwan S, Hunsawattanakul S, Thawinampan J, Siriwan W, Severity of cassava mosaic disease in resistance and commercial varieties by grafting. The 14th National Plant Protection Conference. Thailand. 2019;2019(794):799.

    Google Scholar 

  14. Malik AI, Sophearith S, Delaquis E, Cuellar WJ, Jimenez J, Newby JC. Susceptibility of Cassava Varieties to Disease Caused by Sri Lankan Cassava Mosaic Virus and Impacts on Yield by Use of Asymptomatic and Virus-Free Planting Material. Agronomy. 2022;12:1658.

    Article  CAS  Google Scholar 

  15. Legg JP, Lava Kumar P, Makeshkumar T, Tripathi L, Ferguson M, et al. Cassava virus diseases: biology, epidemiology, and management. Adv Virus Res. 2015;91:85–142.

    Article  CAS  Google Scholar 

  16. Storey HH, Nichols RF. STUDIES OF THE MOSAIC DISEASES OF CASSAVA. Ann Appl Biol. 1938;25:790–806.

    Article  Google Scholar 

  17. Nat NVK, Sanjeeva S, William Y, Nidhi S. Application of Proteomics to Investigate Plant-Microbe Interactions. Curr Proteom. 2007;4:28–43.

    Article  Google Scholar 

  18. Gygi SP, Rist B, Aebersold R. Measuring gene expression by quantitative proteome analysis. Curr Opin Biotechnol. 2000;11:396–401.

    Article  CAS  Google Scholar 

  19. Thurston G, Regan S, Rampitsch C, Xing T. Proteomic and phosphoproteomic approaches to understand plant–pathogen interactions. Physiol Mol Plant Pathol. 2005;66:3–11.

    Article  CAS  Google Scholar 

  20. Dubey H, Grover A. Current initiatives in proteomics research: The plant perspective. Curr Sci. 2001;80:262–9.

    CAS  Google Scholar 

  21. Eldakak M, Milad SIM, Nawar AI, Rohila JS. Proteomics: a biotechnology tool for crop improvement. Front Plant Sci. 2013;4:35–35.

    Article  Google Scholar 

  22. Zhou W, Eudes F, Laroche A. Identification of differentially regulated proteins in response to a compatible interaction between the pathogen Fusarium graminearum and its host. Triticum aestivum Proteomics. 2006;6:4599–609.

    Article  CAS  Google Scholar 

  23. Casado-Vela J, Sellés S, Martínez RB. Proteomic analysis of tobacco mosaic virus-infected tomato (Lycopersicon esculentum M.) fruits and detection of viral coat protein. Proteomics. 2006;6(S1):S196–206.

    Article  Google Scholar 

  24. Raghu D, Senthil N, Raveendran M, Karthikayan G, Pugalendhi L, Janavi G, et al. Proteomic Analysis of Cassava Mosaic Virus (CMV) Responsive Proteins in Cassava Leaf. Int J Curr Microbiol Appl Sci. 2019;8:2988–3005.

    Article  Google Scholar 

  25. Ramulifho E, Rey MEC. Proteome Mapping of South African Cassava Mosaic Virus-Infected Susceptible and Tolerant Landraces of Cassava. Proteomes. 2021;9:41.

    Article  CAS  Google Scholar 

  26. Doyle JJ. A rapid DNA isolation procedure for small quantities of fresh leaf tissue. Phytochem Bull. 1987;19:11–5.

    Google Scholar 

  27. Saokham K, Hemniam N, Roekwan S, Hunsawattanakul S, Thawinampan J, Siriwan W. Survey and molecular detection of Sri Lankan cassava mosaic virus in Thailand. PLoS ONE. 2021;16: e0252846.

    Article  CAS  Google Scholar 

  28. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193:265–75.

    Article  CAS  Google Scholar 

  29. Luna E, Pastor V, Robert J, Flors V, Mauch-Mani B, Ton J. Callose Deposition: A Multifaceted Plant Defense Response. Mol Plant Microbe Interact. 2011;24:183–93.

    Article  CAS  Google Scholar 

  30. Križnik M, Petek M, Dobnik D, Ramšak Ž, Baebler Š, Pollmann S, et al. Salicylic Acid Perturbs sRNA-Gibberellin Regulatory Network in Immune Response of Potato to Potato virus Y Infection. Front Plant Sci. 2017;8:2192.

    Article  Google Scholar 

  31. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods. 2016;13:731–40.

    Article  CAS  Google Scholar 

  32. Howe EA, Sinha R, Schlauch D, Quackenbush J. RNA-Seq analysis in MeV. Bioinformatics. 2011;27:3209–10.

    Article  CAS  Google Scholar 

  33. Mi H, Muruganujan A, Ebert D, Huang X, Thomas PD. PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res. 2019;47(D1):D419-d426.

    Article  CAS  Google Scholar 

  34. Bardou P, Mariette J, Escudié F, Djemiel C, Klopp C. jvenn: an interactive Venn diagram viewer. BMC Bioinformatics. 2014;15(1):293.

    Article  Google Scholar 

  35. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–8.

    Article  CAS  Google Scholar 

  36. Tuller T, Waldman YY, Kupiec M, Ruppin E. Translation efficiency is determined by both codon bias and folding energy. Proc Natl Acad Sci USA. 2010;107:3645–50.

    Article  CAS  Google Scholar 

  37. Gorochowski TE, Ignatova Z, Bovenberg RA, Roubos JA. Trade-offs between tRNA abundance and mRNA secondary structure support smoothing of translation elongation rate. Nucleic Acids Res. 2015;43:3022–32.

    Article  CAS  Google Scholar 

  38. Greenbaum D, Colangelo C, Williams K, Gerstein M. Comparing protein abundance and mRNA expression levels on a genomic scale. Genome Biol. 2003;4:117.

    Article  Google Scholar 

  39. de Sauvage F, Kruys V, Marinx O, Huez G, Octave JN. Alternative polyadenylation of the amyloid protein precursor mRNA regulates translation. EMBO J. 1992;11:3099–103.

    Article  Google Scholar 

  40. Jansen M, de Moor CH, Sussenbach JS, van den Brande JL. Translational control of gene expression. Pediatr Res. 1995;37:681–6.

    Article  CAS  Google Scholar 

  41. Gupta N, Reddy K, Bhattacharyya D, Chakraborty S. Plant responses to geminivirus infection: guardians of the plant immunity. Virol Jou. 2021;18:143.

    Article  CAS  Google Scholar 

  42. Abouzid AM, Frischmuth T, Jeske H. A putative replicative form of the abutilon mosaic virus (gemini group) in a chromatin-like structure. MGG. 1988;212:252–8.

    CAS  Google Scholar 

  43. Zarreen F, Chakraborty S. Epigenetic regulation of geminivirus pathogenesis: a case of relentless recalibration of defence responses in plants. J Exp Bot. 2020;71:6890–906.

    Article  CAS  Google Scholar 

  44. Vanitharani R, Chellappan P, Fauquet CM. Geminiviruses and RNA silencing. Trends Plant Sci. 2005;10:144–51.

    Article  CAS  Google Scholar 

  45. Yang F, Xiao K, Pan H, Liu J. Chloroplast: The Emerging Battlefield in Plant-Microbe Interactions. Front Plant Sci. 2021;12: 637853.

    Article  Google Scholar 

  46. Sowden Robert G, Watson Samuel J, Jarvis P. The role of chloroplasts in plant pathology. Essays Biochem. 2017;62:21–39.

    Google Scholar 

  47. Nomura H, Komori T, Kobori M, Nakahira Y, Shiina T. Evidence for chloroplast control of external Ca2+-induced cytosolic Ca2+ transients and stomatal closure. Plant J. 2008;53:988–98.

    Article  CAS  Google Scholar 

  48. Tang L, Yang G, Ma M, Liu X, Li B, Xie J, et al. An effector of a necrotrophic fungal pathogen targets the calcium-sensing receptor in chloroplasts to inhibit host resistance. Mol Plant Pathol. 2020;21:686–701.

    Article  CAS  Google Scholar 

  49. He Y, Zhou J, Shan L, Meng X. Plant cell surface receptor-mediated signaling - a common theme amid diversity. J Cell Sci. 2018;131:jcs209353.

    Article  Google Scholar 

  50. Nuruzzaman M, Sharoni AM, Kikuchi S. Roles of NAC transcription factors in the regulation of biotic and abiotic stress responses in plants. Front Microbiol. 2013;4:248.

    Article  Google Scholar 

  51. Noman A, Liu Z, Aqeel M, Zainab M, Khan MI, Hussain A, et al. Basic leucine zipper domain transcription factors: the vanguards in plant immunity. Biotechnol Lett. 2017;39:1779–91.

    Article  CAS  Google Scholar 

  52. Wei Y, Shi H, Xia Z, Tie W, Ding Z, Yan Y, et al. Genome-Wide Identification and Expression Analysis of the WRKY Gene Family in Cassava. Front Plant Sci. 2016;7:25.

    Article  Google Scholar 

  53. Bakshi M, Oelmüller R. WRKY transcription factors: Jack of many trades in plants. Plant Signal Behav. 2014;9: e27700.

    Article  CAS  Google Scholar 

  54. Freeborough W, Gentle N, Rey MEC. WRKY Transcription Factors in Cassava Contribute to Regulation of Tolerance and Susceptibility to Cassava Mosaic Disease through Stress Responses. Viruses. 1820;2021:13.

    Google Scholar 

  55. Du L, Chen Z. Identification of genes encoding receptor-like protein kinases as possible targets of pathogen- and salicylic acid-induced WRKY DNA-binding proteins in Arabidopsis. Plant J. 2000;24:837–47.

    Article  CAS  Google Scholar 

  56. Amuge T, Berger DK, Katari MS, Myburg AA, Goldman SL, Ferguson ME. A time series transcriptome analysis of cassava (Manihot esculenta Crantz) varieties challenged with Ugandan cassava brown streak virus. Sci Rep. 2017;7:9747.

    Article  CAS  Google Scholar 

  57. Olsen AN, Ernst HA, Leggio LL, Skriver K. NAC transcription factors: structurally distinct, functionally diverse. Trends Plant Sci. 2005;10:79–87.

    Article  CAS  Google Scholar 

  58. Hu W, Wei Y, Xia Z, Yan Y, Hou X, Zou M, et al. Genome-Wide Identification and Expression Analysis of the NAC Transcription Factor Family in Cassava. PLoS ONE. 2015;10: e0136993.

    Article  Google Scholar 

  59. Li L, Yu X, Thompson A, Guo M, Yoshida S, Asami T, Chory J, et al. Arabidopsis MYB30 is a direct target of BES1 and cooperates with BES1 to regulate brassinosteroid-induced gene expression. Plant J. 2009;582:275–86.

    Article  Google Scholar 

  60. Raffaele S, Rivas S, Roby D. An essential role for salicylic acid in AtMYB30-mediated control of the hypersensitive cell death program in Arabidopsis. FEBS Lett. 2006;580:3498–504.

    Article  CAS  Google Scholar 

  61. Abe H, Urao T, Ito T, Seki M, Shinozaki K, Yamaguchi-Shinozaki K. Arabidopsis AtMYC2 (bHLH) and AtMYB2 (MYB) function as transcriptional activators in abscisic acid signaling. Plant Cell. 2003;15:63–78.

    Article  CAS  Google Scholar 

  62. Murray F, Kalla R, Jacobsen J, Gubler F. A role for HvGAMYB in anther development. Plant J. 2003;33:481–91.

    Article  CAS  Google Scholar 

  63. Lee MW, Qi M, Yang Y. A novel jasmonic acid-inducible rice myb gene associates with fungal infection and host cell death. Mol Plant Microbe Interact. 2001;14:527–35.

    Article  CAS  Google Scholar 

  64. Wang B, Guo X, Zhao P, Liao W, Zeng C, Li K, et al. MeMYB26, a drought-responsive transcription factor in cassava (Manihot esculenta Crantz). Crop Breed. Apl. Biotechnol. 2021; 21:doi:https://doi.org/10.1590/1984-70332021v21n1a4.

  65. Ruan MB, Guo X, Wang B, Yang YL, Li WQ, Yu XL, et al. Genome-wide characterization and expression analysis enables identification of abiotic stress-responsive MYB transcription factors in cassava (Manihot esculenta). J Exp Bot. 2017;68:3657–72.

    Article  CAS  Google Scholar 

  66. Akano O, Dixon O, Mba C, Barrera E, Fregene M. Genetic mapping of a dominant gene conferring resistance to cassava mosaic disease. Theor Appl Genet. 2002;105:521–5.

    Article  CAS  Google Scholar 

  67. Wolfe MD, Rabbi IY, Egesi C, Hamblin M, Kawuki R, Kulakow P, et al. Genome-Wide Association and Prediction Reveals Genetic Architecture of Cassava Mosaic Disease Resistance and Prospects for Rapid Genetic Improvement. Plant Genome. 2016; 9: doi;https://doi.org/10.3835/plantgenome2015.11.0118.

  68. Riechmann JL, Heard J, Martin G, Reuber L, Jiang C, Keddie J, et al. Arabidopsis transcription factors: genome-wide comparative analysis among eukaryotes. Science. 2000;290:2105–10.

    Article  CAS  Google Scholar 

  69. Radoeva T, Lokerse AS, Llavata-Peris CI, Wendrich JR, Xiang D, Liao CY, et al. A Robust Auxin Response Network Controls Embryo and Suspensor Development through a Basic Helix Loop Helix Transcriptional Module. Plant Cell. 2019;31:52–67.

    Article  CAS  Google Scholar 

  70. Zhang HB, Bokowiec MT, Rushton PJ, Han SC, Timko MP. Tobacco transcription factors NtMYC2a and NtMYC2b form nuclear complexes with the NtJAZ1 repressor and regulate multiple jasmonate-inducible steps in nicotine biosynthesis. Mol Plant. 2012;5:73–84.

    Article  CAS  Google Scholar 

  71. Shen Q, Lu X, Yan T, Fu X, Lv Z, Zhang F, et al. The jasmonate-responsive AaMYC2 transcription factor positively regulates artemisinin biosynthesis in Artemisia annua. New Phytol. 2016;210:1269–81.

    Article  CAS  Google Scholar 

  72. Balbi V, Devoto A. Jasmonate signalling network in Arabidopsis thaliana: crucial regulatory nodes and new physiological scenarios. New Phytol. 2008;177:301–18.

    Article  CAS  Google Scholar 

  73. Yang D-L, Yao J, Mei C-S, Tong X-H, Zeng L-J, Li Q, et al. Plant hormone jasmonate prioritizes defense over growth by interfering with gibberellin signaling cascade. Proc Natl Acad Sci USA. 2012;109:E1192–200.

    Article  CAS  Google Scholar 

  74. Wang J, Hu Z, Zhao T, Yang Y, Chen T, Yang M, et al. Genome-wide analysis of bHLH transcription factor and involvement in the infection by yellow leaf curl virus in tomato (Solanum lycopersicum). BMC Genomics. 2015;16:39.

    Article  Google Scholar 

  75. Rodrigues SP, Ventura JA, Aguilar C, Nakayasu ES, Almeida IC, Fernandes PMB, et al. Proteomic analysis of papaya (Carica papaya L.) displaying typical sticky disease symptoms. PROTEOMICS. 2011;11:2592–602.

    Article  CAS  Google Scholar 

  76. Ventelon-Debout M, Delalande F, Brizard J-P, Diemer H, Van Dorsselaer A, Brugidou C. Proteome analysis of cultivar-specific deregulations of Oryza sativa indica and O. sativa japonica cellular suspensions undergoing Rice yellow mottle virus infection. PROTEOMICS. 2004;4:216–25.

    Article  CAS  Google Scholar 

  77. Wu L, Wang S, Chen X, Wang X, Wu L, Zu X, et al. Proteomic and Phytohormone Analysis of the Response of Maize (Zea mays L.) Seedlings to Sugarcane Mosaic Virus. PLOS ONE. 2013;8:e70295.

  78. Li K, Xu C, Zhang J. Proteome profile of maize (Zea Mays L.) leaf tissue at the flowering stage after long-term adjustment to rice black-streaked dwarf virus infection. Gene. 2011;485:106–13.

    Article  CAS  Google Scholar 

  79. Kundu S, Chakraborty D, Kundu A, Pal A. Proteomics approach combined with biochemical attributes to elucidate compatible and incompatible plant-virus interactions between Vigna mungo and Mungbean Yellow Mosaic India Virus. Proteome Sci. 2013;11:15.

    Article  CAS  Google Scholar 

  80. Xu Q, Ni H, Chen Q, Sun F, Zhou T, Lan Y, et al. Comparative proteomic analysis reveals the cross-talk between the responses induced by H2O2 and by long-term rice black-streaked dwarf virus infection in rice. PLoS ONE. 2013;8: e81640.

    Article  Google Scholar 

  81. Cho S-Y, Cho WK, Kim K-H. Identification of tobacco proteins associated with the stem-loop 1 RNAs of Potato virus X. Mol Cells. 2012;33:379–84.

    Article  CAS  Google Scholar 

  82. Goring DR, Walker JC. Self-Rejection–a New Kinase Connection. Science. 2004;303:1474–5.

    Article  CAS  Google Scholar 

  83. Morillo SA, Tax FE. Functional analysis of receptor-like kinases in monocots and dicots. Curr Opin Plant Biol. 2006;9:460–9.

    Article  CAS  Google Scholar 

  84. Bigeard J, Colcombet J, Hirt H. Signaling mechanisms in pattern-triggered immunity (PTI). Mol Plant. 2015;8:521–39.

    Article  CAS  Google Scholar 

  85. Teixeira RM, Ferreira MA, Raimundo GAS, Loriato VAP, Reis PAB, Fontes EPB. Virus perception at the cell surface: revisiting the roles of receptor-like kinases as viral pattern recognition receptors. Mol Plant Pathol. 2019;20:1196–202.

    Article  CAS  Google Scholar 

  86. Nicaise V, Candresse T. Plum pox virus capsid protein suppresses plant pathogen-associated molecular pattern (PAMP)-triggered immunity. Mol Plant Pathol. 2017;18:878–86.

    Article  CAS  Google Scholar 

  87. Kong J, Wei M, Li G, Lei R, Qiu Y, Wang C, et al. The cucumber mosaic virus movement protein suppresses PAMP-triggered immune responses in Arabidopsis and tobacco. Biochem Biophys Res Commun. 2018;498:395–401.

    Article  CAS  Google Scholar 

  88. Reyes MI, Nash TE, Dallas MM, Ascencio-Ibáñez JT, Hanley-Bowdoin L. Peptide aptamers that bind to geminivirus replication proteins confer a resistance phenotype to tomato yellow leaf curl virus and tomato mottle virus infection in tomato. J Virol. 2013;87:9691–706.

    Article  CAS  Google Scholar 

  89. Jones JDG, Dangl JL. The plant immune system. Nature. 2006;444:323–9.

    Article  CAS  Google Scholar 

  90. Szklarczyk D, Santos A, von Mering C, Jensen LJ, Bork P, Kuhn M. STITCH 5: augmenting protein-chemical interaction networks with tissue and affinity data. Nucleic acids res. 2016;44(D1):D380–4.

    Article  CAS  Google Scholar 

  91. Zhao J, Favero DS, Peng H, Neff MM. Arabidopsis thaliana AHL family modulates hypocotyl growth redundantly by interacting with each other via the PPC/DUF296 domain. Proc Natl Acad Sci USA. 2013;110:E4688–97.

    Article  CAS  Google Scholar 

  92. Uddin MN, Dunoyer P, Schott G, Akhter S, Shi C, Lucas WJ, et al. The protein kinase TOUSLED facilitates RNAi in Arabidopsis. Nucleic Acids Res. 2014;42:7971–80.

    Article  CAS  Google Scholar 

  93. Wang D, Zhang X, Yao X, Zhang P, Fang R, Ye J. A 7-Amino-Acid Motif of Rep Protein Essential for Virulence Is Critical for Triggering Host Defense Against Sri Lankan Cassava Mosaic Virus. Mol Plant Microbe Interact. 2020;33:78–86.

    Article  CAS  Google Scholar 

  94. Farkas I, Dombradi V, Miskei M, Szabados L, Koncz C. Arabidopsis PPP family of serine/threonine phosphatases. Trends Plant Sci. 2007;12:169–76.

    Article  CAS  Google Scholar 

  95. Igamberdiev AU, Kleczkowski LA. The Glycerate and Phosphorylated Pathways of Serine Synthesis in Plants: The Branches of Plant Glycolysis Linking Carbon and Nitrogen Metabolism. Front Plant Sci. 2018;9:318.

    Article  Google Scholar 

  96. Zhu X, Wang Y, Su Z, Lv L, Zhang Z. Silencing of the wheat protein phosphatase 2A catalytic subunit TaPP2Ac enhances host resistance to the necrotrophic pathogen Rhizoctonia cerealis. Front Plant Sci. 2018;9:1437.

    Article  Google Scholar 

  97. Sun L, Ren H, Liu R, Li B, Wu T, Sun F, et al. An h-Type Thioredoxin Functions in Tobacco Defense Responses to Two Species of Viruses and an Abiotic Oxidative Stress. Mol Plant Microbe Interact. 2010;23:1470–85.

    Article  CAS  Google Scholar 

  98. Liu Q, Liu H, Gong Y, Tao Y, Jiang L, Zuo W, et al. An Atypical Thioredoxin Imparts Early Resistance to Sugarcane Mosaic Virus in Maize. Mol Plant. 2017;10:483–97.

    Article  CAS  Google Scholar 

  99. Mathioudakis MM, Khechmar S, Owen CA, Medina V, Ben Mansour K, Tomaszewska W, et al. A Thioredoxin Domain-Containing Protein Interacts with Pepino mosaic virus Triple Gene Block Protein 1. Int J Mol Sci. 2018;19:3747.

    Article  Google Scholar 

Download references

Acknowledgements

This research was supported by Kasetsart University Research and Development (KURDI), FF(KU)18.65.

The authors confirm their contribution to the paper as follows: study conception and design: S. Wanwisa, R. Sittiruk and S. Aroonothai; data collection: Y. Author; analysis and interpretation of results: S. Wanwisa, R. Sittiruk, S. Charoenlappanit, V. Nattachai, H. Nuannapa, C. Somruthai and M. Srihunsa; draft manuscript preparation: S. Wanwisa. Author. All authors reviewed the results and approved the final version of the manuscript.

Funding

This research was supported by Kasetsart University Research and Development (KURDI), FF(KU)18.65.

Author information

Authors and Affiliations

Authors

Contributions

The authors confirm contribution to the paper as follows: study conception and design: S. Wanwisa, R. Sittiruk and S. Aroonothai; data collection: Y. Author; analysis and interpretation of results: S. Wanwisa, R. Sittiruk, S. Charoenlappanit, V. Nattachai, H. Nuannapa, C. Somruthai and M. Srihunsa; draft manuscript preparation: S. Wanwisa. Author. All authors reviewed the results and approved the final version of the manuscript.

Corresponding author

Correspondence to Wanwisa Siriwan.

Ethics declarations

Ethics approval and consent to participate

All experiments were performed in accordance to relevant guidelines and regulations.

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.

Supplementary Information

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

Siriwan, W., Hemniam, N., Vannatim, N. et al. Analysis of proteomic changes in cassava cv. Kasetsart 50 caused by Sri Lankan cassava mosaic virus infection. BMC Plant Biol 22, 573 (2022). https://doi.org/10.1186/s12870-022-03967-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12870-022-03967-1

Keywords