Next Article in Journal
Acknowledgment to Reviewers of Proteomes in 2021
Previous Article in Journal
Potential Tear Biomarkers for the Diagnosis of Parkinson’s Disease—A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Shotgun Proteomics as a Powerful Tool for the Study of the Proteomes of Plants, Their Pathogens, and Plant–Pathogen Interactions

1
New Town Research Laboratories, Tasmanian Institute of Agriculture, University of Tasmania, New Town, TAS 7008, Australia
2
Central Science Laboratory, University of Tasmania, Hobart, TAS 7001, Australia
*
Authors to whom correspondence should be addressed.
Proteomes 2022, 10(1), 5; https://doi.org/10.3390/proteomes10010005
Submission received: 20 December 2021 / Revised: 12 January 2022 / Accepted: 17 January 2022 / Published: 19 January 2022
(This article belongs to the Section Plant Proteomics)

Abstract

:
The interaction between plants and pathogenic microorganisms is a multifaceted process mediated by both plant- and pathogen-derived molecules, including proteins, metabolites, and lipids. Large-scale proteome analysis can quantify the dynamics of proteins, biological pathways, and posttranslational modifications (PTMs) involved in the plant–pathogen interaction. Mass spectrometry (MS)-based proteomics has become the preferred method for characterizing proteins at the proteome and sub-proteome (e.g., the phosphoproteome) levels. MS-based proteomics can reveal changes in the quantitative state of a proteome and provide a foundation for understanding the mechanisms involved in plant–pathogen interactions. This review is intended as a primer for biologists that may be unfamiliar with the diverse range of methodology for MS-based shotgun proteomics, with a focus on techniques that have been used to investigate plant–pathogen interactions. We provide a summary of the essential steps required for shotgun proteomic studies of plants, pathogens and plant–pathogen interactions, including methods for protein digestion, identification, separation, and quantification. Finally, we discuss how protein PTMs may directly participate in the interaction between a pathogen and its host plant.

1. Introduction

As a multifaceted process, plant–pathogen interactions have been extensively researched from both the pathogen and plant viewpoints. Plants are surrounded by many microorganisms, some of which can cause diseases and lead to negative impacts on yield, quality, and value [1]. Pathogen-derived molecules such as nucleic acids, secondary metabolites, and proteins are major factors in pathogenicity which allow the successful invasion and colonization of host tissues. Plants also produce molecules important in recognition of the pathogens and pathogen-derived molecules that can elicit defense responses. These responses need to be quick and specific to minimize the damage caused by pathogenic microorganisms [2]. An understanding of how pathogens and plants recognize each other and how they communicate pre- and post-infection is crucial in this field of investigation. The increase in genomic and transcriptome studies has advanced the understanding of the pathogenicity strategies employed by pathogens and the immune responses in plants [2,3,4,5]. However, these approaches have limitations and cannot monitor post-transcriptional processes [6]. Therefore, investigation at post-transcriptome levels (i.e., proteome and metabolome levels) is required for a better understanding of the pathogen–host interaction.
Proteomics is defined as the large-scale study of different proteins expressed by an organism [7] and has become a driver in discovering host-pathogen communication [8]. Study at the proteome level allows the concurrent study of the total proteome, its qualitative presence and quantitative abundance, variation within a population, and localization. The role of mass spectrometry (MS) in different aspects of plant biology, such as plant defense and signal transduction, has been reviewed in detail [9,10]. Technological advances in MS-based proteomics have significantly accelerated the characterization of pathogen–host protein interactions [11] and while gel-based methods for intact protein separation and identification remain in use for specialized applications [12], shotgun analysis is well established as the dominant proteomics platform [13]. The success of these approaches depends on various factors, including the methods used for isolation, digestion, separation, identification, and quantification of proteins [14]. In addition to improvements in MS sensitivity that continue to extend proteome depth, modern tools for bioinformatic analysis [15] and the availability of annotated genomes for many non-model organisms [16,17,18,19,20] has increased the potential to expand our understanding of plant–pathogen interaction. The purpose of this review is to summarize the research techniques of proteomics and posttranslational modification (PTM) analysis that can be used to uncover regulatory principles underlying plant host-pathogen interaction. We discuss methodological aspects, with emphasis on sample preparation, MS strategies, and PTMs, with a focus on the approaches that have already been successfully introduced in plant–pathogen studies.

2. Sample Preparation Prior to LC-MS/MS

An efficient sample preparation method for obtaining high-quality peptides for proteomic analysis of pathogens and plants represents a greater challenge than most other cell/tissue types [21]. Plant tissues with robust cell walls can be difficult to fully disrupt and contain relatively high levels of secondary metabolites that can interfere with proteome analysis [22]. In pathogens, sample preparation for proteomics can also be challenging. For example, obligate biotrophic pathogens are not culturable on the artificial media, and therefore it can be difficult to obtain pure samples of these pathogens [23]. In addition, many plant pathogens such as soilborne pathogens, produce long-lived resting spores, which are highly resistant to adverse environmental conditions [24]. Harsh conditions, including mechanical force, must be applied for the disruption of the thickened cell walls in these structures [25].

2.1. Protein Extraction

Protein extraction is one of the most critical steps in proteome analysis studies [26]. The ideal extraction method should first and foremost be robust in terms of reproducibility. Extraction conditions should minimize protein degradation and unwanted modifications, and solubilize the maximum number of proteins [27]. Plant tissues contain large amounts of compounds, including phenolics, organic acids, pigments, and polysaccharides that interfere with further steps of protein analysis [28]. There are several different methods to disrupt microorganisms and plant tissues and reduce protein degradation. Cell disruption for protein extraction can use chemical and/or physical approaches [29]. Snap freezing in liquid nitrogen, bead beating, the addition of organic solvents, thermolysis, and sonication are commonly used in plant and pathogen proteomics studies. Selecting the best cell/tissue disruption method depends on several factors and has been extensively reviewed before [27,30,31,32,33]. A perennial challenge for proteomic analysis of plant tissues is the interference of high abundance proteins such as the subunits of ribulose-1,5-bisphosphate carboxylase oxygenase (Rubisco), which can account for up to 50% of total protein in mature leaves of C3 plants [34]. Depletion of Rubisco is therefore potentially one method to extend proteome coverage by “unmasking” lower abundance proteins. Several methods have been developed for the depletion of highly abundant proteins [35,36,37], of which a few examples are presented here. Widjaja et al. [38] used a combination of Rubisco depletion and sub-proteome enrichment for the identification of low abundance proteins during Arabidopsis defense response. This approach enabled them to identify several low abundance proteins that differentially regulated post infection. Zhang et al. [39] developed a polyethyleneimine assisted Rubisco cleanup (PARC) method to improve proteomics coverage in rice plants. The results showed that PARC effectively removed Rubisco and improved protein identification. In another study of the plant proteome, a fractionation method using 10 mM of Ca2+ and 10 mM of phytate was used to precipitate Rubisco from soybean leaf protein extract [40]. This technique successfully removed 85% Rubisco enzyme from soybean leaf extract and enable identification of several new low abundance proteins.

2.2. Sample Cleanup

Plant and pathogen proteomics has currently moved from gel-based methods to gel-free shotgun (bottom-up) approaches [41,42,43]. Several new strategies for proteome sample preparation involving the removal of substances such as detergents, salts, and chaotropic agents which interfere with the protein digestion and MS analysis were introduced in recent years [44]. Traditional approaches for the removal of surfactants and other contaminants have been reviewed before [45,46]. More recent innovations include filter-aided sample preparation (FASP) [47], protein suspension trapping (S-Trap) [48], and single-pot solid-phase-enhanced sample preparation (SP3) [49]. In the FASP method, the sample lysate is applied to an ultrafiltration unit for washing out the low-mass contaminants and digesting the proteins on the membrane. Although many useful modifications of the classical FASP protocol have been published [47,50,51,52], there are a few disadvantages with the FASP method, which can limit the application of this method. The FASP methods are expensive and rather time-consuming. Moreover, the efficiency of the FASP filter for SDS removal has been doubted by the detection of traces of remaining SDS after FASP [53]. In the S-Trap (suspension trapping) method, the proteins are digested in a filter after removing contaminants in a short wash step. In contrast to the FASP method, which employs a molecular weight cut-off membrane, the S-Trap filter consists of a three-dimensional porous material. FASP requires hours of processing (approximately 3 h), while due to the large pore size, the total processing time is reduced to less than 15 min in the S-Trap [54]. In the protein analysis of bacterial whole-cell lysate, both S-trap and FASP methods yielded similar results regarding peptide and protein identifications [54]. The SP3 protocol consists of nonselective protein binding, where proteins are captured on the surface of magnetic beads. The beads are compatible with various organic solvents and detergents, including urea, SDS, and acetonitrile (ACN). The adaptability of the SP3 protocol to a 96-well platform provides a fast and efficient technique easily applicable for large-scale protein interactome analysis [55,56]. Ludwig et al. [57] also showed that S-Traps outperformed FASP and in-solution digest methods for colorectal cancer cell lysate regardless of lysis conditions. However, the potential for losses during wash steps if protein material does not completely aggregate onto magnetic beads remains the main limitation to the SP3 protocol. The solvent precipitation SP3 (SP4) method can be an efficient and effective alternative to SP3 [58], in which the magnetic beads are omitted, and brief centrifugation with or without an inert glass bead capture the aggregated protein. SP4 recovered equivalent or greater protein yields and improved reproducibility compered to SP3.
The selection of sample preparation method is highly dependent on tissue type, and no sample preparation method has been found to be applicable for all sample types. For example, Mikulášek et al. [59] showed that the SP3 workflow was the best sample preparation method (in comparison to FASP and S-Trap) for protein analysis of Arabidopsis leaves in terms of number of identifications, proteome coverage, number of missed cleavages, reduction of handling time, repeatability, and cost per assay. Similarly, Stoychev [60] found that the SP3 workflow resulted in over 30% increase in identified post-translational modifications of peptides and an approximately two-fold increase in peptide recovery compared to FASP. In contrast, the SP3 method was not very successful for the proteomic analysis of barley anthers [61]. In the protein analysis of resting spores in the obligate biotrophic plant pathogen Spongospora subterranea, the S-Trap method delivered a higher number of protein identifications with an improved reproducibility compared to the SP3 method [24,62].

3. MS Strategies

There are many options for MS-based proteomics, and decisions on what approaches to use are influenced by the equipment and expertise available as well as the specific research questions [63]. An untargeted proteome analysis that provides global-scale proteome changes would likely be chosen by the researcher aiming to quantify as many proteins as possible [64]. For the accurate quantification of a specific protein or small group of proteins, a targeted proteomics approach is preferred [65]. Deciding on the data acquisition and quantification methods is highly dependent on the experimental design and sample preparation [66].
Mass spectrometry using data-dependent acquisition (DDA) and, increasingly, data-independent acquisition (DIA) approaches have dominated the methodology for untargeted proteomics [67]. In DDA mode, the N most intense peptide precursors in a survey MS1 scan (10–25 most abundant peptides) at each point of the chromatographic gradient are identified and fragmented to acquire MS2 spectra. Each MS2 spectrum in DDA is effectively a single analyte and is matched to a protein database to identify specific peptides [68]. The well-established instrument operation, the option of label-dependent quantitation, data analysis, and processing pipelines are all benefits of DDA workflows [69,70]. However, due to the semi-stochastic sampling of lower abundance peptides, the inter-sample reproducibility in DDA is relatively low. The resulting “missing value” problem potentially limits the statistical analysis of all identified proteins across an experiment [64]. DIA methods represent an appealing alternative for DDA as all theoretical peptides in a sample are fragmented sequentially across mass windows of predefined m/z intervals. This provides quantitative data across the chromatographic peak at the MS2 level, which can be used in addition to MS1-level data for more precise peptide quantitation [71,72]. While the resulting MS2 spectra are highly multiplexed, an ever-expanding array of software solutions exist for MS2 spectrum deconvolution and peptide identification, using both spectrum-centric and peptide-centric approaches [73,74,75,76]. In addition to improvements in sensitivity and reproducibility, a further benefit of the DIA workflow is the ability to reanalyze previous DIA results as spectral libraries and algorithms are developed [77]. Parallel accumulation–serial fragmentation (PASEF) is another acquisition method that enhances sequencing speed and enables hundreds of MS/MS events per second at full sensitivity. In this method, synchronized scans in a trapped ion mobility device allow a 10-fold gain in sequencing speed without decreasing sensitivity [78]. A detailed description of the construction and operation of the PASEF has been published elsewhere [79,80,81]. This approach can improve protein identification in a complex interaction between plant and pathogen. Jin et al. [82] used a PASEF-MS/MS workflow to identify proteins associated with Fusarium crown rot resistance in wheat. A total of 9234 proteins were identified, including proteins associated with defense, photosynthesis, and cell wall formation.
Proteomic approaches can also be classified as label-based or label-free, which both have their own sets of strengths and limitations [83]. For intact protein analysis, the two-dimensional difference gel electrophoresis (2D-DIGE) method [84] uses protein labeling with cyanine fluorescent dyes for relative protein quantitation between two or more multiplexed samples. 2D-DIGE has been widely used in the study of plant proteomes [85,86,87] but mostly prior to the more widespread adoption of gel-free proteomics. In another early example of label-based analysis, this time analogous to the SILAC approach, Bindschedler et al. [88] developed a cost-effective method called hydroponic isotope labeling of entire plants (HILEP) for quantitative plant proteomics. In HILEP, the whole and mature plants are labeled with a stable isotope such as 15N. Zhang et al. [89] later used HILEP combined with phosphopeptide enrichment to study the phosphorylation events in auxin signaling in lateral root induction of Arabidopsis. The isobaric tag for relative and absolute quantitation (iTRAQ) is another label-based method that is used in plant proteomics [90], pathogen proteomics [91], and plant–pathogen interaction analysis [92]. One of the advantages of iTRAQ compared to HILEP is that iTRAQ allows for 4 or 8 comparisons, while HILEP is suitable for pair-wise comparisons [87]. In label-based quantitation, most often used in conjunction with DDA-MS, samples are differentially labeled with alternative differential mass tags, which allows the detection of peptides based on the change in the mass [64,93]. Sample multiplexing reduces variability, which can substantially minimize instrument time if “single shot” analysis is used. Alternatively, peptide labeling can be combined with off-line fractionation using an orthogonal separation approach (e.g., strong cation exchange) as a method to significantly extend proteome depth. The proteolytic, metabolic, and chemical labeling strategies are the most widely used labeling methods [94] and can be used for both absolute and relative quantification of proteins [95]. In contrast to label-based methods, label-free quantification of peptides is typically a more straightforward workflow that also does not require expensive labeling reagents. An overview of the experimental workflows for methods that can be used in shotgun proteomics is presented in Figure 1.
Several recent studies have employed DIA approaches for the analysis of the interaction between pathogens and their host plants. A DIA-MS workflow was used to profile the proteome of S. subterranea in resistant and susceptible potato cultivars [96]. The finding of this study illuminated the regulatory principles underlying Spongospora–potato interaction. The interaction between barley and Pyrenophora teres was also studied using DIA-MS [97]. Over 1000 proteins were quantified in which the increase in abundance of several classes of pathogenesis-related (PR) proteins was confirmed. A similar method demonstrated an increase in jasmonic acid biosynthesis and a decrease in photosynthesis-associated proteins in rice plants in response to pathogen infection [98]. More than 2000 proteins from tomato leaves infected by Pseudomonas syringae were identified using DIA-MS [99]. The significantly changed proteins belonged to immune response, redox processes, energy generation, and carbon fixation in the chloroplast. Fan et al. [100] employed a DIA-MS method to study the interaction between tomato and the hemibiotrophic oomycete pathogen Phytophthora infestans. Among the changed proteins, several were involved in plant defense responses, metabolic pathways, and signaling. The effect of Funneliformis mosseae in soybean roots was investigated using a transcriptomic and proteomic (DDA-MS) analysis. A total of 9488 proteins were identified, and the key pathways and differentially abundance proteins were involved in plant–pathogen interaction, phenylalanine metabolism, hormone signal transduction, and metabolic pathways [101]. Using a combination of a shotgun DDA-MS and a targeted DIA-MS, several peptides of potential markers for resistance to Peyronellaea pinodes, causing Ascochyta blight, were identified. This study revealed the importance of plant cell walls to hinder the growth of the pathogen within cells and redox response for the detoxification of fungal toxins [102]. Kerr et al. [103] used DIA-MS to analyze barley seed proteome during fungal infection. This study showed that oxalate oxidase was the only protein consistently increased in abundance in the infected plants.

4. Post-Translational Modifications

All living organisms need to respond to environmental changes quickly and efficiently using a strict regulatory system through molecular interactions of hundreds to thousands of biomolecules [104]. Due to multiple levels of regulation such as PTM and alternative splicing, a single gene can produce several different proteins, increasing proteome diversity [105]. PTMs act as molecular switches that can lead to dramatic changes in the regulation of molecular functions without changes in the transcriptome and proteome levels [106]. Protein phosphorylation is one of the most frequently studied PTMs and represents over 53% of all the PTMs based on the published experimental data [107]. Phosphorylation/dephosphorylation of proteins is a fast response that can switch on or off the cell processes or biological pathways. In eukaryotes, phosphorylation occurs on serine, tyrosine, and threonine [108]. MS-based proteomics has become the primary tool used to study protein phosphorylation. The simultaneous identification/quantitation of phosphopeptides and proteins has expanded our understanding of complicated biological systems and their regulation [109,110,111,112,113].
Similar to proteomics, a phosphopeptide analysis workflow starts with protein extraction and digestion (Figure 2). Phosphopeptide analysis needs one- or multi-stage enrichment strategies to achieve comprehensive coverage of phosphorylation events [114,115], and therefore requires more starting material, in the range of an order of magnitude compared with total peptide analysis. Enrichment methods include immobilized metal affinity chromatography (IMAC), metal oxide affinity chromatography (MOAC), titanium dioxide (TiO2) phosphopeptide enrichment, electrostatic repulsion hydrophilic interaction chromatography (ERLIC), and phosphopeptide precipitation [116,117,118,119]. Large-scale profiling of plant phosphoproteomes after infection by pathogens has revealed the dynamic phosphorylation events that regulate plant resistance or susceptibility to pathogens [110,120]. Quantitative phosphoproteomics analysis of Arabidopsis revealed the regulatory mechanisms of pathogen-associated molecular pattern immunity. The results of this study showed that some of the identified phosphosites are required for the production of reactive oxygen species during immunity against a virulent necrotrophic fungus [121]. Phosphoproteomics profiling of cotton roots in response to the soilborne plant pathogenic fungus Verticillium dahliae infection identified 92 and 38 specific phosphoproteins in the resistant and susceptible lines, respectively [122].
In addition to phosphorylation, large-scale profiling of other PTMs such as acetylation and ubiquitination during plant–pathogen interactions is becoming more common. In rice [123,124] and Arabidopsis [125,126,127], hundreds of ubiquitin-modified proteins were identified during the plant immune responses. The acetylome profiling of maize in response to Cochliobolus carbonum infection confirmed the hyperacetylation of several proteins, including chromatin remodeling enzymes and transcription factors [128]. Although recent studies have identified many acetylated proteins in diverse pathogens, the impact of these PTMs on the pathogenicity of plant pathogens is yet to be understood [129,130,131,132].

5. Bioinformatics

Molecular research routinely involves the application of computational methods to convert raw experimental data into condensed results for biological interpretation. In recent years, several software packages and online platforms have been developed to facilitate the analysis, interpretation, and visualization of proteomics data. The MaxQuant/Perseus platform is a user-friendly, interactive workflow environment and can provide complete documentation of computational methods, and has become one of the most popular software suites for biological interpretation of protein quantification, interaction, and PTM data [133]. Relative label-free quantification of shotgun proteomics data is one of the most used applications in biological science, including plant science, microbiology, and plant pathology. LFQ-Analyst, which is easy-to-use, has recently been created to perform differential expression analysis with “one-click” and to visualize proteomics data sets preprocessed with MaxQuant software [134]. Galaxy [135] and MetaboAnalyst [136] also provide various workflow management systems to analyze MS-based proteomics data. The PeptideWitch software package is a python-based web module for the label-free shotgun proteomics data visualization [137]. This software produces many statistical and graphical outputs including heatmaps, volcano plots, Venn diagrams, and p-value histograms. In addition to the technical advance in proteomics data analysis software, specific bioinformatics tools have been developed to study the relationships between plants and pathogens. For example, PhytoPath, which is a database on plant–pathogen interactions, provides genome-scale data from pathogens with information about plant infection phenotypes. To date, this database includes the genome information of 99 plants, 107 pathogens, and 350 interactions. The access to the complete assembly of genome and gene models of phytopathogens in PhytoPath gained using the Ensembl Genomes browser [138]. NIASGBdb, which links the genetic resources of plants and pathogens to plant disease information, is another example of plant–pathogen interactions databases [139].
Data sharing in MS-based proteomics is becoming a standard for proteomics researchers. However, any proteomics data set is only partially understood according to the availability of the currently available analytical tools (such as the algorithms for peak detection and quantitation and the proteome database that was searched). Therefore, there is an excellent opportunity for re-analyzing and reusing public data, particularly for benchmarking studies and evaluation of new bioinformatics software. While the majority of publicly available data sets correspond to human and other main model organisms, there has been a rapid expansion in proteomic datasets for non-model organisms in recent years [140]. The first proteomics resources were set up more than 15 years ago. The GPMDB [141], PeptideAtlas [142], and the PRoteomics IDEntifications (PRIDE Archive) [143] databases are a few examples of MS-based proteomics resources. PRIDE is the world’s largest MS-based proteomics data repository, with an average of around 500 datasets deposited per month during 2021 [144]. PRIDE stores datasets coming from all experimental proteomics approaches, including DDA and DIA proteomics. The PhosPhAt 4.0 is one of the most significant phosphorylation databases for Arabidopsis phosphorylation studies and contains phosphorylation sites identified in Arabidopsis by MS-based proteomics [145]. In addition, the PhosPhAt 4.0 database includes phosphorylation site prediction and kinase–target relationship retrieval, which provides researchers with more functionality for plant phosphoproteomics analysis. The availability of large MS datasets and bioinformatics tools has enabled the comparison of different proteomics studies. For example, Pinski et al. [146] used several previously published MS datasets to compare the bioinformatics predictions of the sub-cellular localization of cell wall proteins. Collectively, these public databases include valuable resources for research on humans, model organisms, non-model plants, and microorganisms. However, there is still a need for more comprehensive plant–pathogen genome and proteome databases [147].

6. Conclusions and Future Perspective

In summary, recent advances in MS-based proteomics and bioinformatics tools now enable the robust profiling of plant and pathogenic microbe samples to an unprecedented depth. Global proteome profiling using shotgun approaches during infection can identify specific proteins, molecular functions, and PTMs involved in plant disease resistance and susceptibility and pathogenicity processes. Therefore, proteomics will remain one of the fastest-growing areas in plant and pathogen research. Increasingly, studies that use “omics” technologies in combination (multi-omics approaches) enable cross-validation of data sets and the ability to filter out the most significant biological changes. Integration of shotgun proteomics with other omics approaches will further expand our understanding of biological mechanisms involved in host–pathogen interactions.
Proteomics analysis of mixtures containing different cell populations only provides a quantitative analysis of proteins that reflect the average variation in the whole cell population. Thus, the molecular changes in distinct subpopulations of rare cells will be missed in bulk sampling methods [148]. Techniques that address single cells’ molecular identity can help better understand uniqueness within the complexity of plant–pathogen interaction. The proteome of single cells can provide unique information about the processes taking place in the interaction between plants and pathogens, revealing signaling events that are taking place in a specific types of plant (or pathogen) cells. While single cell proteomics has a huge potential in developing a better understanding of the intricate connections between the host plant and its pathogen, it has yet to be applied to the analysis of such a complex interaction.
In addition, cross-linking mass spectrometry (XL-MS) has recently emerged to study protein interactomics on the system-wide level [149]. XL-MS is a unique technology capable of capturing the dynamic biological assemblies in their native environment and uncovering their physical interaction contacts [150,151]. Liu et al. [152] developed an in planta chemical cross-linking-based quantitative interactomics (IPQCX–MS) workflow in Arabidopsis to study protein–protein interactions. They identified 354 unique cross-linked peptides and showed that this workflow can identify hundreds of peptides cross-linked in vivo. Considering the exciting new developments in computational approaches, XL-MS can be expected to become one of the most versatile methods in the study of plant–pathogen interactions within the next ten years. Together, we believe that proteomic studies using the techniques presented in this review can guide breeding efforts to identify resistant cultivars and help with the development of antimicrobial agents, leading to improved global food security.

Author Contributions

Conceptualization, S.B., R.W., C.R.W., R.S.T. and D.S.N.; writing—original draft preparation, S.B.; writing—review and editing, R.W., C.R.W., R.S.T. and D.S.N.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Australian Research Council, Discovery Grant program, DP180103337.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Boyd, L.A.; Ridout, C.; O’Sullivan, D.M.; Leach, J.E.; Leung, H. Plant-pathogen interactions: Disease resistance in modern agriculture. Trends Genet. 2013, 29, 233–240. [Google Scholar] [CrossRef]
  2. Gupta, R.; Lee, S.E.; Agrawal, G.K.; Rakwal, R.; Park, S.; Wang, Y.; Kim, S.T. Understanding the plant-pathogen interactions in the context of proteomics-generated apoplastic proteins inventory. Front. Plant Sci. 2015, 6, 352. [Google Scholar] [CrossRef] [Green Version]
  3. Balotf, S.; Tegg, R.S.; Nichols, D.S.; Wilson, C.R. Spore Germination of the Obligate Biotroph Spongospora subterranea: Transcriptome Analysis Reveals Germination Associated Genes. Front. Microbiol. 2021, 12, 1557. [Google Scholar] [CrossRef]
  4. Hayden, K.J.; Garbelotto, M.; Knaus, B.J.; Cronn, R.C.; Rai, H.; Wright, J.W. Dual RNA-seq of the plant pathogen Phytophthora ramorum and its tanoak host. Tree Genet. Genom. 2014, 10, 489–502. [Google Scholar] [CrossRef] [Green Version]
  5. Schneider, D.J.; Collmer, A. Studying plant-pathogen interactions in the genomics era: Beyond molecular Koch’s postulates to systems biology. Annu. Rev. Phytopathol. 2010, 48, 457–479. [Google Scholar] [CrossRef] [Green Version]
  6. Haddad, N.; Johnson, N.; Kathariou, S.; Métris, A.; Phister, T.; Pielaat, A.; Tassou, C.; Wells-Bennik, M.H.; Zwietering, M.H. Next generation microbiological risk assessment—Potential of omics data for hazard characterisation. Int. J. Food Microbiol. 2018, 287, 28–39. [Google Scholar] [CrossRef] [PubMed]
  7. Wilkins, M.R.; Sanchez, J.-C.; Gooley, A.A.; Appel, R.D.; Humphery-Smith, I.; Hochstrasser, D.F.; Williams, K.L. Progress with Proteome Projects: Why all Proteins Expressed by a Genome Should be Identified and How to Do It. Biotechnol. Genet. Eng. Rev. 1996, 13, 19–50. [Google Scholar] [CrossRef] [Green Version]
  8. Lum, K.K.; Cristea, I.M. Proteomic approaches to uncovering virus–host protein interactions during the progression of viral infection. Expert Rev. Proteom. 2016, 13, 325–340. [Google Scholar] [CrossRef]
  9. Glinski, M.; Weckwerth, W. The role of mass spectrometry in plant systems biology. Mass Spectrom. Rev. 2006, 25, 173–214. [Google Scholar] [CrossRef] [PubMed]
  10. Chen, Y.; Weckwerth, W. Mass spectrometry untangles plant membrane protein signaling networks. Trends Plant Sci. 2020, 25, 930–944. [Google Scholar] [CrossRef]
  11. Meyer, M.D.; Ryck, J.D.; Goormachtig, S.; Van Damme, P. Keeping in Touch with Type-III Secretion System Effectors: Mass Spectrometry-Based Proteomics to Study Effector–Host Protein–Protein Interactions. Int. J. Mol. Sci. 2020, 21, 6891. [Google Scholar] [CrossRef] [PubMed]
  12. Prathi, N.B.; Palit, P.; Madhu, P.; Ramesh, M.; Laha, G.; Balachandran, S.; Madhav, M.S.; Sundaram, R.; Mangrauthia, S.K. Proteomic and transcriptomic approaches to identify resistance and susceptibility related proteins in contrasting rice genotypes infected with fungal pathogen Rhizoctonia solani. Plant Physiol. Biochem. 2018, 130, 258–266. [Google Scholar] [CrossRef] [PubMed]
  13. Sinitcyn, P.; Rudolph, J.D.; Cox, J. Computational methods for understanding mass spectrometry—Based shotgun proteomics data. Annu. Rev. Biomed. Data Sci. 2018, 1, 207–234. [Google Scholar] [CrossRef]
  14. Walther, T.C.; Mann, M. Mass spectrometry—Based proteomics in cell biology. J. Cell Biol. 2010, 190, 491–500. [Google Scholar] [CrossRef] [PubMed]
  15. Da Veiga Leprevost, F.; Haynes, S.E.; Avtonomov, D.M.; Chang, H.-Y.; Shanmugam, A.K.; Mellacheruvu, D.; Kong, A.T.; Nesvizhskii, A.I. Philosopher: A versatile toolkit for shotgun proteomics data analysis. Nat. Methods 2020, 17, 869–870. [Google Scholar] [CrossRef]
  16. Consortium, P.G.S. Genome sequence and analysis of the tuber crop potato. Nature 2011, 475, 189. [Google Scholar]
  17. Salanoubat, M.; Genin, S.; Artiguenave, F.; Gouzy, J.; Mangenot, S.; Arlat, M.; Billault, A.; Brottier, P.; Camus, J.; Cattolico, L. Genome sequence of the plant pathogen Ralstonia solanacearum. Nature 2002, 415, 497–502. [Google Scholar] [CrossRef] [Green Version]
  18. Poimala, A.; Vainio, E.J. Complete genome sequence of a novel toti-like virus from the plant-pathogenic oomycete Phytophthora cactorum. Arch. Virol. 2020, 165, 1679–1682. [Google Scholar] [CrossRef]
  19. Seo, S.; Pokhrel, A.; Coleman, J.J. The genome sequence of five genotypes of Fusarium oxysporum f. sp. vasinfectum: A resource for studies on Fusarium wilt of cotton. Mol. Plant-Microbe Interact. 2020, 33, 138–140. [Google Scholar] [CrossRef] [Green Version]
  20. Baroncelli, R.; Pensec, F.; Da Lio, D.; Boufleur, T.R.; Vicente, I.; Sarrocco, S.; Picot, A.; Baraldi, E.; Sukno, S.; Thon, M.R. Complete Genome Sequence of the plant pathogenic fungus Colletotrichum lupini. Mol. Plant-Microbe Interact. 2021, 34, 1461–1464. [Google Scholar] [CrossRef]
  21. Song, G.; Hsu, P.Y.; Walley, J.W. Assessment and refinement of sample preparation methods for deep and quantitative plant proteome profiling. Proteomics 2018, 18, 1800220. [Google Scholar] [CrossRef] [PubMed]
  22. Hussein, R.A.; El-Anssary, A.A. Plants secondary metabolites: The key drivers of the pharmacological actions of medicinal plants. Herb. Med. 2019, 1, 13. [Google Scholar]
  23. Shimizu, M.; Wariishi, H. Development of a sample preparation method for fungal proteomics. FEMS Microbiol. Lett. 2005, 247, 17–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Balotf, S.; Wilson, R.; Tegg, R.S.; Nichols, D.S.; Wilson, C.R. Optimisation of sporosori purification and protein extraction techniques for the biotrophic protozoan plant pathogen Spongospora subterranea. Molecules 2020, 25, 3109. [Google Scholar] [CrossRef]
  25. Horvatić, A.; Kuleš, J.; Guillemin, N.; Galan, A.; Mrljak, V.; Bhide, M. High-throughput proteomics and the fight against pathogens. Mol. Biosyst. 2016, 12, 2373–2384. [Google Scholar] [CrossRef] [PubMed]
  26. Patole, C.; Bindschedler, L.V. Plant proteomics: A guide to improve the proteome coverage. In Advances in Biological Science Research; Elsevier: Amsterdam, The Netherlands, 2019; pp. 45–67. [Google Scholar]
  27. Wu, X.; Gong, F.; Wang, W. Protein extraction from plant tissues for 2DE and its application in proteomic analysis. Proteomics 2014, 14, 645–658. [Google Scholar] [CrossRef] [PubMed]
  28. Charmont, S.; Jamet, E.; Pont-Lezica, R.; Canut, H. Proteomic analysis of secreted proteins from Arabidopsis thaliana seedlings: Improved recovery following removal of phenolic compounds. Phytochemistry 2005, 66, 453–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Hopkins, T. Physical and chemical cell disruption for the recovery of intracellular proteins. Bioprocess Technol. 1991, 12, 57–83. [Google Scholar]
  30. Wang, W.; Tai, F.; Chen, S. Optimizing protein extraction from plant tissues for enhanced proteomics analysis. J. Sep. Sci. 2008, 31, 2032–2039. [Google Scholar] [CrossRef] [PubMed]
  31. Thiellement, H.; Zivy, M.; Damerval, C.; Méchin, V. Plant Proteomics; Human Press: Totowa, NJ, USA, 2007. [Google Scholar]
  32. Niu, L.; Yuan, H.; Gong, F.; Wu, X.; Wang, W. Protein extraction methods shape much of the extracted proteomes. Front. Plant Sci. 2018, 9, 802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Fernandez-Acero, F.J.; Carbú, M.; Garrido, C.; Vallejo, I.; Cantoral, J.M. Proteomic advances in phytopathogenic fungi. Curr. Proteom. 2007, 4, 79–88. [Google Scholar] [CrossRef]
  34. Makino, A.; Mae, T.; Ohira, K. Changes in photosynthetic capacity in rice leaves from emergence through senescence. Analysis from ribulose-1, 5-bisphosphate carboxylase and leaf conductance. Plant Cell Physiol. 1984, 25, 511–521. [Google Scholar]
  35. Koroleva, O.A.; Bindschedler, L.V. Efficient strategies for analysis of low abundance proteins in plant proteomics. In Sample Preparation in Biological Mass Spectrometry; Springer: Berlin/Heidelberg, Germany, 2011; pp. 381–409. [Google Scholar]
  36. Righetti, P.G.; Boschetti, E. Low-abundance plant protein enrichment with peptide libraries to enlarge proteome coverage and related applications. Plant Sci. 2020, 290, 110302. [Google Scholar] [CrossRef] [PubMed]
  37. Gupta, R.; Wang, Y.; Agrawal, G.K.; Rakwal, R.; Jo, I.H.; Bang, K.H.; Kim, S.T. Time to dig deep into the plant proteome: A hunt for low-abundance proteins. Front. Plant Sci. 2015, 6, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Widjaja, I.; Naumann, K.; Roth, U.; Wolf, N.; Mackey, D.; Dangl, J.L.; Scheel, D.; Lee, J. Combining subproteome enrichment and Rubisco depletion enables identification of low abundance proteins differentially regulated during plant defense. Proteomics 2009, 9, 138–147. [Google Scholar] [CrossRef]
  39. Zhang, Y.; Gao, P.; Xing, Z.; Jin, S.; Chen, Z.; Liu, L.; Constantino, N.; Wang, X.; Shi, W.; Yuan, J.S. Application of an improved proteomics method for abundant protein cleanup: Molecular and genomic mechanisms study in plant defense. Mol. Cell. Proteom. 2013, 12, 3431–3442. [Google Scholar] [CrossRef] [Green Version]
  40. Krishnan, H.B.; Natarajan, S.S. A rapid method for depletion of Rubisco from soybean (Glycine max) leaf for proteomic analysis of lower abundance proteins. Phytochemistry 2009, 70, 1958–1964. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, W.-Q.; Jensen, O.N.; Møller, I.M.; Hebelstrup, K.H.; Rogowska-Wrzesinska, A. Evaluation of sample preparation methods for mass spectrometry-based proteomic analysis of barley leaves. Plant Methods 2018, 14, 1–13. [Google Scholar] [CrossRef] [PubMed]
  42. Bahmani, M.; O’Lone, C.E.; Juhász, A.; Nye-Wood, M.; Dunn, H.; Edwards, I.B.; Colgrave, M.L. Application of mass spectrometry-based proteomics to barley research. J. Agric. Food Chem. 2021, 69, 8591–8609. [Google Scholar] [CrossRef] [PubMed]
  43. Ball, B.; Bermas, A.; Carruthers-Lay, D.; Geddes-McAlister, J. Mass spectrometry-based proteomics of fungal pathogenesis, host–fungal interactions, and antifungal development. J. Fungi 2019, 5, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yang, Z.; Shen, X.; Chen, D.; Sun, L. Toward a universal sample preparation method for denaturing top-down proteomics of complex proteomes. J. Proteome Res. 2020, 19, 3315–3325. [Google Scholar] [CrossRef] [PubMed]
  45. Feist, P.; Hummon, A.B. Proteomic challenges: Sample preparation techniques for microgram-quantity protein analysis from biological samples. Int. J. Mol. Sci. 2015, 16, 3537–3563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wither, M.J.; Hansen, K.C.; Reisz, J.A. Mass spectrometry-based bottom-up proteomics: Sample preparation, LC-MS/MS analysis, and database query strategies. Curr. Protoc. Protein Sci. 2016, 86, 11–20. [Google Scholar] [CrossRef]
  47. Wiśniewski, J.R. Filter-aided sample preparation for proteome analysis. In Microbial Proteomics; Springer: Berlin/Heidelberg, Germany, 2018; pp. 3–10. [Google Scholar]
  48. Zougman, A.; Selby, P.J.; Banks, R.E. Suspension trapping (STrap) sample preparation method for bottom-up proteomics analysis. Proteomics 2014, 14. [Google Scholar] [CrossRef] [PubMed]
  49. Hughes, C.S.; Moggridge, S.; Müller, T.; Sorensen, P.H.; Morin, G.B.; Krijgsveld, J. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat. Protoc. 2019, 14, 68–85. [Google Scholar] [CrossRef] [PubMed]
  50. Nel, A.J.; Garnett, S.; Blackburn, J.M.; Soares, N.C. Comparative reevaluation of FASP and enhanced FASP methods by LC–MS/MS. J. Proteome Res. 2015, 14, 1637–1642. [Google Scholar] [CrossRef] [PubMed]
  51. Zhang, Z.; Dubiak, K.M.; Huber, P.W.; Dovichi, N.J. Miniaturized filter-aided sample preparation (MICRO-FASP) method for high throughput, ultrasensitive proteomics sample preparation reveals proteome asymmetry in Xenopus laevis embryos. Anal. Chem. 2020, 92, 5554–5560. [Google Scholar] [CrossRef]
  52. Lipecka, J.; Chhuon, C.; Bourderioux, M.; Bessard, M.A.; van Endert, P.; Edelman, A.; Guerrera, I.C. Sensitivity of mass spectrometry analysis depends on the shape of the filtration unit used for filter aided sample preparation (FASP). Proteomics 2016, 16, 1852–1857. [Google Scholar] [CrossRef] [PubMed]
  53. Yeung, Y.G.; Stanley, E.R. Rapid detergent removal from peptide samples with ethyl acetate for mass spectrometry analysis. Curr. Protoc. Protein Sci. 2010, 59, 11–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. HaileMariam, M.; Eguez, R.V.; Singh, H.; Bekele, S.; Ameni, G.; Pieper, R.; Yu, Y. S-Trap, an ultrafast sample-preparation approach for shotgun proteomics. J. Proteome Res. 2018, 17, 2917–2924. [Google Scholar] [CrossRef]
  55. Hayoun, K.; Gaillard, J.-C.; Pible, O.; Alpha-Bazin, B.; Armengaud, J. High-throughput proteotyping of bacterial isolates by double barrel chromatography-tandem mass spectrometry based on microplate paramagnetic beads and phylopeptidomics. J. Proteom. 2020, 226, 103887. [Google Scholar] [CrossRef] [PubMed]
  56. Gonzalez-Lozano, M.A.; Koopmans, F.; Paliukhovich, I.; Smit, A.B.; Li, K.W. A fast and economical sample preparation protocol for interaction proteomics analysis. Proteomics 2019, 19, 1900027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ludwig, K.R.; Schroll, M.M.; Hummon, A.B. Comparison of in-solution, FASP, and S-trap based digestion methods for bottom-up proteomic studies. J. Proteome Res. 2018, 17, 2480–2490. [Google Scholar] [CrossRef] [PubMed]
  58. Johnston, H.E.; Yadav, K.; Kirkpatrick, J.M.; Biggs, G.S.; Oxley, D.; Kramer, H.B.; Samant, R.S. Solvent Precipitation SP3 (SP4) enhances recovery for proteomics sample preparation without magnetic beads. bioRxiv 2021. [Google Scholar] [CrossRef]
  59. Mikulášek, K.; Konečná, H.; Potěšil, D.; Holánková, R.; Havliš, J.; Zdráhal, Z. SP3 Protocol for Proteomic Plant Sample Preparation Prior LC-MS/MS. Front. Plant Sci. 2021, 12, 369. [Google Scholar] [CrossRef] [PubMed]
  60. Stoyan Stoychev, P.N.; Mamputhaa, S.; Khumaloa, F.; Gerber, I.; van der Westhuyzena, C.; Jordaan, J. Universal Unbiased Pre-MS Clean-Up Using Magnetic HILIC Microparticles for SPE. Available online: https://resynbio.com/hilic/ (accessed on 25 February 2021).
  61. Lewandowska, D.; Zhang, R.; Colas, I.; Uzrek, N.; Waugh, R. Application of a sensitive and reproducible label-free proteomic approach to explore the proteome of individual meiotic-phase barley anthers. Front. Plant Sci. 2019, 10, 393. [Google Scholar] [CrossRef]
  62. Balotf, S.; Wilson, R.; Tegg, R.S.; Nichols, D.S.; Wilson, C.R. Quantitative proteomics provides an insight into germination-related proteins in the obligate biotrophic plant pathogen Spongospora subterranea. Environ. Microbiol. Rep. 2021, 13, 521–532. [Google Scholar] [CrossRef]
  63. Schubert, O.T.; Röst, H.L.; Collins, B.C.; Rosenberger, G.; Aebersold, R. Quantitative proteomics: Challenges and opportunities in basic and applied research. Nat. Protoc. 2017, 12, 1289–1294. [Google Scholar] [CrossRef] [PubMed]
  64. Elmore, J.M.; Griffin, B.D.; Walley, J.W. Advances in functional proteomics to study plant-pathogen interactions. Curr. Opin. Plant Biol. 2021, 63, 102061. [Google Scholar] [CrossRef] [PubMed]
  65. Peterson, A.C.; Russell, J.D.; Bailey, D.J.; Westphall, M.S.; Coon, J.J. Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol. Cell. Proteom. 2012, 11, 1475–1488. [Google Scholar] [CrossRef] [Green Version]
  66. Borràs, E.; Sabidó, E. What is targeted proteomics? A concise revision of targeted acquisition and targeted data analysis in mass spectrometry. Proteomics 2017, 17, 1700180. [Google Scholar] [CrossRef] [PubMed]
  67. Fernández-Costa, C.; Martínez-Bartolomé, S.; McClatchy, D.B.; Saviola, A.J.; Yu, N.-K.; Yates, J.R., III. Impact of the Identification Strategy on the Reproducibility of the DDA and DIA Results. J. Proteome Res. 2020, 19, 3153–3161. [Google Scholar] [CrossRef]
  68. Davies, V.; Wandy, J.; Weidt, S.; Van Der Hooft, J.J.; Miller, A.; Daly, R.; Rogers, S. Rapid Development of Improved Data-Dependent Acquisition Strategies. Anal. Chem. 2021, 93, 5676–5683. [Google Scholar] [CrossRef] [PubMed]
  69. Yu, F.; Teo, G.C.; Kong, A.T.; Haynes, S.E.; Avtonomov, D.M.; Geiszler, D.J.; Nesvizhskii, A.I. Identification of modified peptides using localization-aware open search. Nat. Commun. 2020, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  70. Verheggen, K.; Ræder, H.; Berven, F.S.; Martens, L.; Barsnes, H.; Vaudel, M. Anatomy and evolution of database search engines—A central component of mass spectrometry based proteomic workflows. Mass Spectrom. Rev. 2020, 39, 292–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Bruderer, R.; Bernhardt, O.M.; Gandhi, T.; Xuan, Y.; Sondermann, J.; Schmidt, M.; Gomez-Varela, D.; Reiter, L. Optimization of experimental parameters in data-independent mass spectrometry significantly increases depth and reproducibility of results. Mol. Cell. Proteom. 2017, 16, 2296–2309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chen, M.-X.; Zhang, Y.; Fernie, A.R.; Liu, Y.-G.; Zhu, F.-Y. SWATH-MS-based proteomics: Strategies and applications in plants. Trends Biotechnol. 2021, 39, 433–437. [Google Scholar] [CrossRef]
  73. Zhang, F.; Ge, W.; Ruan, G.; Cai, X.; Guo, T. Data-independent acquisition mass spectrometry-based proteomics and software tools: A glimpse in 2020. Proteomics 2020, 20, 1900276. [Google Scholar] [CrossRef]
  74. Gotti, C.; Roux-Dalvai, F.; Joly-Beauparlant, C.; Mangnier, L.; Leclercq, M.; Droit, A. Extensive and accurate benchmarking of DIA acquisition methods and software tools using a complex proteomic standard. J. Proteome Res. 2021, 20, 4801–4814. [Google Scholar] [CrossRef]
  75. Pino, L.K.; Just, S.C.; MacCoss, M.J.; Searle, B.C. Acquiring and analyzing data independent acquisition proteomics experiments without spectrum libraries. Mol. Cell. Proteom. 2020, 19, 1088–1103. [Google Scholar] [CrossRef] [Green Version]
  76. Sinitcyn, P.; Hamzeiy, H.; Salinas Soto, F.; Itzhak, D.; McCarthy, F.; Wichmann, C.; Steger, M.; Ohmayer, U.; Distler, U.; Kaspar-Schoenefeld, S. MaxDIA enables library-based and library-free data-independent acquisition proteomics. Nat. Biotechnol. 2021, 39, 1563–1573. [Google Scholar] [CrossRef] [PubMed]
  77. Ludwig, C.; Gillet, L.; Rosenberger, G.; Amon, S.; Collins, B.C.; Aebersold, R. Data-independent acquisition-based SWATH-MS for quantitative proteomics: A tutorial. Mol. Syst. Biol. 2018, 14, e8126. [Google Scholar] [CrossRef]
  78. Meier, F.; Beck, S.; Grassl, N.; Lubeck, M.; Park, M.A.; Raether, O.; Mann, M. Parallel accumulation-serial fragmentation (PASEF): Multiplying sequencing speed and sensitivity by synchronized scans in a trapped ion mobility device. J. Proteome Res. 2015, 14, 5378–5387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Fernandez-Lima, F.; Kaplan, D.; Park, M. Note: Integration of trapped ion mobility spectrometry with mass spectrometry. Rev. Sci. Instrum. 2011, 82, 126106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Michelmann, K.; Silveira, J.A.; Ridgeway, M.E.; Park, M.A. Fundamentals of trapped ion mobility spectrometry. J. Am. Soc. Mass Spectrom. 2014, 26, 14–24. [Google Scholar] [CrossRef] [PubMed]
  81. Charkow, J.; Röst, H.L. Trapped Ion Mobility Spectrometry Reduces Spectral Complexity in Mass Spectrometry-Based Proteomics. Anal. Chem. 2021, 93, 16751–16758. [Google Scholar] [CrossRef]
  82. Jin, J.-J.; Duan, S.-N.; Qi, Y.-Z.; Zhen, W.-C.; Jun, M. Identification of proteins associated with Fusarium crown rot resistance in wheat using label-free quantification analysis. J. Integr. Agric. 2021, 20, 3209–3221. [Google Scholar] [CrossRef]
  83. Anand, S.; Samuel, M.; Ang, C.-S.; Keerthikumar, S.; Mathivanan, S. Label-based and label-free strategies for protein quantitation. In Proteome Bioinformatics; Springer: Berlin/Heidelberg, Germany, 2017; pp. 31–43. [Google Scholar]
  84. Ünlü, M.; Morgan, M.E.; Minden, J.S. Difference gel electrophoresis. A single gel method for detecting changes in protein extracts. Electrophoresis 1997, 18, 2071–2077. [Google Scholar] [CrossRef] [PubMed]
  85. Arruda, S.C.C.; de Sousa Barbosa, H.; Azevedo, R.A.; Arruda, M.A.Z. Two-dimensional difference gel electrophoresis applied for analytical proteomics: Fundamentals and applications to the study of plant proteomics. Analyst 2011, 136, 4119–4126. [Google Scholar] [CrossRef] [Green Version]
  86. Guo, J.; Hao, G.; Hatt, S.; Wang, Z.; Francis, F. Host plant adaptability and proteomic differences of diverse Rhopalosiphum maidis (Fitch) lineages. Arch. Insect Biochem. Physiol. 2022, 109, e21853. [Google Scholar] [CrossRef]
  87. Bindschedler, L.V.; Cramer, R. Quantitative plant proteomics. Proteomics 2011, 11, 756–775. [Google Scholar] [CrossRef]
  88. Bindschedler, L.V.; Palmblad, M.; Cramer, R. Hydroponic isotope labelling of entire plants (HILEP) for quantitative plant proteomics; an oxidative stress case study. Phytochemistry 2008, 69, 1962–1972. [Google Scholar] [CrossRef]
  89. Zhang, H.; Zhou, H.; Berke, L.; Heck, A.J.; Mohammed, S.; Scheres, B.; Menke, F.L. Quantitative phosphoproteomics after auxin-stimulated lateral root induction identifies an SNX1 protein phosphorylation site required for growth. Mol. Cell. Proteom. 2013, 12, 1158–1169. [Google Scholar] [CrossRef] [Green Version]
  90. Zou, X.; Li, L.; Liao, F.; Chen, W. iTRAQ-based quantitative proteomic analysis reveals NtGNL1-dependent regulatory network underlying endosome trafficking for pollen tube polar growth. Plant Physiol. Biochem. 2021, 161, 200–209. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, R.; Jiang, W.; Liu, X.; Duan, Y.; Xiang, L.; Wang, Y.; Jiang, Y.; Shen, X.; Chen, X.; Yin, C. ITRAQ-based quantitative proteomic analysis of Fusarium moniliforme (Fusarium verticillioides) in response to Phloridzin inducers. Proteome Sci. 2021, 19, 1–13. [Google Scholar] [CrossRef] [PubMed]
  92. Feng, Z.-M.; Peng, G.; Zhao, J.-H.; Wang, G.-D.; Zhang, H.-M.; Cao, W.-L.; Xiang, X.; Zhang, Y.-F.; Rong, H.; Chen, Z.-X. iTRAQ-based quantitative proteomics analysis of defense responses triggered by the pathogen Rhizoctonia solani infection in rice. J. Integr. Agric. 2022, 21, 139–152. [Google Scholar] [CrossRef]
  93. Wang, F.-X.; Luo, Y.-M.; Ye, Z.-Q.; Cao, X.; Liang, J.-N.; Wang, Q.; Wu, Y.; Wu, J.-H.; Wang, H.-Y.; Zhang, M. iTRAQ-based proteomics analysis of autophagy-mediated immune responses against the vascular fungal pathogen Verticillium dahliae in Arabidopsis. Autophagy 2018, 14, 598–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Huang, A.; Tang, Y.; Shi, X.; Jia, M.; Zhu, J.; Yan, X.; Chen, H.; Gu, Y. Proximity labeling proteomics reveals critical regulators for inner nuclear membrane protein degradation in plants. Nat. Commun. 2020, 11, 1–10. [Google Scholar] [CrossRef] [PubMed]
  95. Domon, B.; Aebersold, R. Options and considerations when selecting a quantitative proteomics strategy. Nat. Biotechnol. 2010, 28, 710–721. [Google Scholar] [CrossRef]
  96. Balotf, S.; Wilson, R.; Tegg, R.S.; Nichols, D.S.; Wilson, C.R. In Planta Transcriptome and Proteome Profiles of Spongospora subterranea in Resistant and Susceptible Host Environments Illuminates Regulatory Principles Underlying Host–Pathogen Interaction. Biology 2021, 10, 840. [Google Scholar] [CrossRef] [PubMed]
  97. Hassett, K.; Ellwood, S.R.; Zulak, K.G.; Muria-Gonzalez, M.J. Analysis of apoplastic proteins expressed during net form net blotch of barley. J. Plant Dis. Prot. 2020, 127, 683–694. [Google Scholar] [CrossRef] [Green Version]
  98. Cheah, B.H.; Lin, H.-H.; Chien, H.-J.; Liao, C.-T.; Liu, L.-Y.D.; Lai, C.-C.; Lin, Y.-F.; Chuang, W.-P. SWAtH-MS-based quantitative proteomics reveals a uniquely intricate defense response in Cnaphalocrocis medinalis-resistant rice. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef] [PubMed]
  99. Fan, K.-T.; Wang, K.-H.; Chang, W.-H.; Yang, J.-C.; Yeh, C.-F.; Cheng, K.-T.; Hung, S.-C.; Chen, Y.-R. Application of data-independent acquisition approach to study the proteome change from early to later phases of tomato pathogenesis responses. Int. J. Mol. Sci. 2019, 20, 863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Fan, K.-T.; Hsu, Y.; Yeh, C.-F.; Chang, C.-H.; Chang, W.-H.; Chen, Y.-R. Quantitative Proteomics Reveals the Dynamic Regulation of the Tomato Proteome in Response to Phytophthora infestans. Int. J. Mol. Sci. 2021, 22, 4174. [Google Scholar] [CrossRef]
  101. Zhang, X.-Q.; Bai, L.; Sun, H.-B.; Yang, C.; Cai, B.-Y. Transcriptomic and proteomic analysis revealed the effect of funneliformis mosseae in soybean roots differential expression genes and proteins. J. Proteome Res. 2020, 19, 3631–3643. [Google Scholar] [CrossRef] [PubMed]
  102. Castillejo, M.-Á.; Fondevilla-Aparicio, S.; Fuentes-Almagro, C.; Rubiales, D. Quantitative analysis of target peptides related to resistance against Ascochyta blight (Peyronellaea pinodes) in pea. J. Proteome Res. 2020, 19, 1000–1012. [Google Scholar] [CrossRef] [PubMed]
  103. Kerr, E.D.; Phung, T.K.; Caboche, C.H.; Fox, G.P.; Platz, G.J.; Schulz, B.L. The intrinsic and regulated proteomes of barley seeds in response to fungal infection. Anal. Biochem. 2019, 580, 30–35. [Google Scholar] [CrossRef] [Green Version]
  104. Iqbal, Z.; Iqbal, M.S.; Hashem, A.; Abd_Allah, E.F.; Ansari, M.I. Plant defense responses to biotic stress and its interplay with fluctuating dark/light conditions. Front. Plant Sci. 2021, 12, 297. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, W.; Triplett, L.; Chen, X.-L. Emerging Roles of Posttranslational Modifications in Plant-Pathogenic Fungi and Bacteria. Annu. Rev. Phytopathol. 2021, 59, 99–124. [Google Scholar] [CrossRef]
  106. He, Z.; Huang, T.; Ao, K.; Yan, X.; Huang, Y. Sumoylation, phosphorylation, and acetylation fine-tune the turnover of plant immunity components mediated by ubiquitination. Front. Plant Sci. 2017, 8, 1682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Silva-Sanchez, C.; Li, H.; Chen, S. Recent advances and challenges in plant phosphoproteomics. Proteomics 2015, 15, 1127–1141. [Google Scholar] [CrossRef] [PubMed]
  108. Batalha, I.L.; Lowe, C.R.; Roque, A.C. Platforms for enrichment of phosphorylated proteins and peptides in proteomics. Trends Biotechnol. 2012, 30, 100–110. [Google Scholar] [CrossRef] [PubMed]
  109. Yang, F.; Melo-Braga, M.N.; Larsen, M.R.; Jørgensen, H.J.; Palmisano, G. Battle through signaling between wheat and the fungal pathogen Septoria tritici revealed by proteomics and phosphoproteomics. Mol. Cell. Proteom. 2013, 12, 2497–2508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Pang, Q.; Zhang, T.; Zhang, A.; Lin, C.; Kong, W.; Chen, S. Proteomics and phosphoproteomics revealed molecular networks of stomatal immune responses. Planta 2020, 252, 1–17. [Google Scholar] [CrossRef] [PubMed]
  111. 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. [Google Scholar] [CrossRef]
  112. Xing, T.; Laroche, A. Revealing plant defense signaling: Getting more sophisticated with phosphoproteomics. Plant Signal. Behav. 2011, 6, 1469–1474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Roustan, V.; Weckwerth, W. Quantitative phosphoproteomic and system-level analysis of TOR inhibition unravel distinct organellar acclimation in Chlamydomonas reinhardtii. Front. Plant Sci. 2018, 9, 1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhang, T.; Schneider, J.D.; Zhu, N.; Chen, S. Identification of MAPK substrates using quantitative phosphoproteomics. In Plant Pattern Recognition Receptors; Springer: Berlin/Heidelberg, Germany, 2017; pp. 133–142. [Google Scholar]
  115. Song, G.; Montes, C.; Walley, J.W. Quantitative profiling of protein abundance and phosphorylation state in plant tissues using tandem mass tags. In Plant Proteomics; Springer: Berlin/Heidelberg, Germany, 2020; pp. 147–156. [Google Scholar]
  116. Fíla, J.; Honys, D. Enrichment techniques employed in phosphoproteomics. Amino Acids 2012, 43, 1025–1047. [Google Scholar] [CrossRef] [Green Version]
  117. Low, T.Y.; Mohtar, M.A.; Lee, P.Y.; Omar, N.; Zhou, H.; Ye, M. Widening the bottleneck of phosphoproteomics: Evolving strategies for phosphopeptide enrichment. Mass Spectrom. Rev. 2021, 40, 309–333. [Google Scholar] [CrossRef]
  118. Qiu, W.; Evans, C.A.; Landels, A.; Pham, T.K.; Wright, P.C. Phosphopeptide enrichment for phosphoproteomic analysis—A tutorial and review of novel materials. Anal. Chim. Acta 2020, 1129, 158–180. [Google Scholar] [CrossRef]
  119. Wolschin, F.; Wienkoop, S.; Weckwerth, W. Enrichment of phosphorylated proteins and peptides from complex mixtures using metal oxide/hydroxide affinity chromatography (MOAC). Proteomics 2005, 5, 4389–4397. [Google Scholar] [CrossRef]
  120. Zhang, B.; Shao, L.; Wang, J.; Zhang, Y.; Guo, X.; Peng, Y.; Cao, Y.; Lai, Z. Phosphorylation of ATG18a by BAK1 suppresses autophagy and attenuates plant resistance against necrotrophic pathogens. Autophagy 2021, 17, 2093–2110. [Google Scholar] [CrossRef]
  121. Kadota, Y.; Liebrand, T.W.; Goto, Y.; Sklenar, J.; Derbyshire, P.; Menke, F.L.; Torres, M.A.; Molina, A.; Zipfel, C.; Coaker, G. Quantitative phosphoproteomic analysis reveals common regulatory mechanisms between effector-and PAMP-triggered immunity in plants. New Phytol. 2019, 221, 2160–2175. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, Y.; Shi, Y.; Zhao, L.; Wei, F.; Feng, Z.; Feng, H. Phosphoproteomics profiling of cotton (Gossypium hirsutum L.) roots in response to Verticillium dahliae inoculation. ACS Omega 2019, 4, 18434–18443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zhang, C.; Wei, Y.; Xu, L.; Wu, K.-C.; Yang, L.; Shi, C.-N.; Yang, G.-Y.; Chen, D.; Yu, F.-F.; Xie, Q. A Bunyavirus-inducible ubiquitin ligase targets RNA polymerase IV for degradation during viral pathogenesis in rice. Mol. Plant 2020, 13, 836–850. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, X.-L.; Xie, X.; Wu, L.; Liu, C.; Zeng, L.; Zhou, X.; Luo, F.; Wang, G.-L.; Liu, W. Proteomic analysis of ubiquitinated proteins in rice (Oryza sativa) after treatment with pathogen-associated molecular pattern (PAMP) elicitors. Front. Plant Sci. 2018, 9, 1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Grubb, L.E.; Derbyshire, P.; Dunning, K.E.; Zipfel, C.; Menke, F.L.; Monaghan, J. Large-scale identification of ubiquitination sites on membrane-associated proteins in Arabidopsis thaliana seedlings. Plant Physiol. 2021, 185, 1483–1488. [Google Scholar] [CrossRef]
  126. Liu, X.; Zhou, Y.; Du, M.; Liang, X.; Fan, F.; Huang, G.; Zou, Y.; Bai, J.; Lu, D. The calcium-dependent protein kinase CPK28 is targeted by the ubiquitin ligases ATL31 and ATL6 for proteasome-mediated degradation to fine-tune immune signaling in Arabidopsis. Plant Cell 2021, koab242. [Google Scholar] [CrossRef]
  127. Song, G.; Olatunji, D.; Montes, C.; Clark, N.M.; Pu, Y.; Kelley, D.R.; Walley, J.W. Quantitative proteomics reveals extensive lysine ubiquitination in the Arabidopsis root proteome and uncovers novel transcription factor stability states. bioRxiv 2021. [Google Scholar] [CrossRef]
  128. Walley, J.W.; Shen, Z.; McReynolds, M.R.; Schmelz, E.A.; Briggs, S.P. Fungal-induced protein hyperacetylation in maize identified by acetylome profiling. Proc. Natl. Acad. Sci. USA 2018, 115, 210–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Lv, B.; Yang, Q.; Li, D.; Liang, W.; Song, L. Proteome-wide analysis of lysine acetylation in the plant pathogen Botrytis cinerea. Sci. Rep. 2016, 6, 29313. [Google Scholar] [CrossRef] [Green Version]
  130. Li, D.; Lv, B.; Tan, L.; Yang, Q.; Liang, W. Acetylome analysis reveals the involvement of lysine acetylation in diverse biological processes in Phytophthora sojae. Sci. Rep. 2016, 6, 29897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Zhou, S.; Wu, C. Comparative acetylome analysis reveals the potential roles of lysine acetylation for DON biosynthesis in Fusarium graminearum. BMC Genom. 2019, 20, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Liao, X.; Li, Y.; Hu, Z.; Lin, Y.; Zheng, B.; Ding, J. Poplar acetylome profiling reveals lysine acetylation dynamics in seasonal bud dormancy release. Plant Cell Environ. 2021, 44, 1830–1845. [Google Scholar] [CrossRef] [PubMed]
  133. Tyanova, S.; Temu, T.; Sinitcyn, P.; Carlson, A.; Hein, M.Y.; Geiger, T.; Mann, M.; Cox, J. The Perseus computational platform for comprehensive analysis of (prote) omics data. Nat. Methods 2016, 13, 731–740. [Google Scholar] [CrossRef] [PubMed]
  134. Shah, A.D.; Goode, R.J.; Huang, C.; Powell, D.R.; Schittenhelm, R.B. LFQ-analyst: An easy-to-use interactive web platform to analyze and visualize label-free proteomics data preprocessed with MaxQuant. J. Proteome Res. 2019, 19, 204–211. [Google Scholar] [CrossRef]
  135. Afgan, E.; Baker, D.; Batut, B.; Van Den Beek, M.; Bouvier, D.; Čech, M.; Chilton, J.; Clements, D.; Coraor, N.; Grüning, B.A. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2018 update. Nucleic Acids Res. 2018, 46, W537–W544. [Google Scholar] [CrossRef] [Green Version]
  136. Chong, J.; Soufan, O.; Li, C.; Caraus, I.; Li, S.; Bourque, G.; Wishart, D.S.; Xia, J. MetaboAnalyst 4.0: Towards more transparent and integrative metabolomics analysis. Nucleic Acids Res. 2018, 46, W486–W494. [Google Scholar] [CrossRef] [Green Version]
  137. Handler, D.C.; Cheng, F.; Shathili, A.M.; Haynes, P.A. PeptideWitch–A software package to produce high-stringency proteomics data visualizations from label-free shotgun proteomics data. Proteomes 2020, 8, 21. [Google Scholar] [CrossRef] [PubMed]
  138. Pedro, H.; Maheswari, U.; Urban, M.; Irvine, A.G.; Cuzick, A.; McDowall, M.D.; Staines, D.M.; Kulesha, E.; Hammond-Kosack, K.E.; Kersey, P.J. PhytoPath: An integrative resource for plant pathogen genomics. Nucleic Acids Res. 2016, 44, D688–D693. [Google Scholar] [CrossRef] [Green Version]
  139. Takeya, M.; Yamasaki, F.; Uzuhashi, S.; Aoki, T.; Sawada, H.; Nagai, T.; Tomioka, K.; Tomooka, N.; Sato, T.; Kawase, M. NIASGBdb: NIAS Genebank databases for genetic resources and plant disease information. Nucleic Acids Res. 2010, 39, D1108–D1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Martens, L.; Vizcaíno, J.A. A golden age for working with public proteomics data. Trends Biochem. Sci. 2017, 42, 333–341. [Google Scholar] [CrossRef] [Green Version]
  141. Craig, R.; Cortens, J.P.; Beavis, R.C. Open source system for analyzing, validating, and storing protein identification data. J. Proteome Res. 2004, 3, 1234–1242. [Google Scholar] [CrossRef] [PubMed]
  142. Deutsch, E.W.; Lam, H.; Aebersold, R. PeptideAtlas: A resource for target selection for emerging targeted proteomics workflows. EMBO Rep. 2008, 9, 429–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Vizcaíno, J.A.; Csordas, A.; Del-Toro, N.; Dianes, J.A.; Griss, J.; Lavidas, I.; Mayer, G.; Perez-Riverol, Y.; Reisinger, F.; Ternent, T. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 2016, 44, D447–D456. [Google Scholar] [CrossRef] [PubMed]
  144. Perez-Riverol, Y.; Bai, J.; Bandla, C.; García-Seisdedos, D.; Hewapathirana, S.; Kamatchinathan, S.; Kundu, D.J.; Prakash, A.; Frericks-Zipper, A.; Eisenacher, M. The PRIDE database resources in 2022: A hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 2022, 50, D543–D552. [Google Scholar] [CrossRef]
  145. Xi, L.; Zhang, Z.; Schulze, W.X. PhosPhAt 4.0: An Updated Arabidopsis Database for Searching Phosphorylation Sites and Kinase-Target Interactions. In Plant Phosphoproteomics; Springer: Berlin/Heidelberg, Germany, 2021; pp. 189–202. [Google Scholar]
  146. Pinski, A.; Roujol, D.; Pouzet, C.; Bordes, L.; San Clemente, H.; Hoffmann, L.; Jamet, E. Comparison of mass spectrometry data ad bioinformatics predictions to assess the bona fide localization of proteins identified in cell wall proteomics studies. Plant Sci. 2021, 310, 110979. [Google Scholar] [CrossRef]
  147. Dong, A.-Y.; Wang, Z.; Huang, J.-J.; Song, B.-A.; Hao, G.-F. Bioinformatic tools support decision-making in plant disease management. Trends Plant Sci. 2021, 26, 953–967. [Google Scholar] [CrossRef]
  148. Ctortecka, C.; Mechtler, K. The rise of single-cell proteomics. Anal. Sci. Adv. 2021, 2, 84–94. [Google Scholar] [CrossRef]
  149. Yu, C.; Huang, L. Cross-linking mass spectrometry (XL-MS): An emerging technology for interactomics and structural biology. Anal. Chem. 2018, 90, 144. [Google Scholar] [CrossRef]
  150. Iacobucci, C.; Götze, M.; Sinz, A. Cross-linking/mass spectrometry to get a closer view on protein interaction networks. Curr. Opin. Biotechnol. 2020, 63, 48–53. [Google Scholar] [CrossRef] [PubMed]
  151. Piersimoni, L.; Kastritis, P.L.; Arlt, C.; Sinz, A. Cross-Linking Mass Spectrometry for Investigating Protein Conformations and Protein–Protein Interactions—A Method for All Seasons. Chem. Rev. 2021. [Google Scholar] [CrossRef] [PubMed]
  152. Liu, S.; Yu, F.; Hu, Q.; Wang, T.; Yu, L.; Du, S.; Yu, W.; Li, N. Development of in planta chemical cross-Linking-Based quantitative interactomics in arabidopsis. J. Proteome Res. 2018, 17, 3195–3213. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of different experimental workflows for shotgun proteomics.
Figure 1. Overview of different experimental workflows for shotgun proteomics.
Proteomes 10 00005 g001
Figure 2. An overview of the possible experimental workflows for shotgun phosphoproteomics.
Figure 2. An overview of the possible experimental workflows for shotgun phosphoproteomics.
Proteomes 10 00005 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Balotf, S.; Wilson, R.; Tegg, R.S.; Nichols, D.S.; Wilson, C.R. Shotgun Proteomics as a Powerful Tool for the Study of the Proteomes of Plants, Their Pathogens, and Plant–Pathogen Interactions. Proteomes 2022, 10, 5. https://doi.org/10.3390/proteomes10010005

AMA Style

Balotf S, Wilson R, Tegg RS, Nichols DS, Wilson CR. Shotgun Proteomics as a Powerful Tool for the Study of the Proteomes of Plants, Their Pathogens, and Plant–Pathogen Interactions. Proteomes. 2022; 10(1):5. https://doi.org/10.3390/proteomes10010005

Chicago/Turabian Style

Balotf, Sadegh, Richard Wilson, Robert S. Tegg, David S. Nichols, and Calum R. Wilson. 2022. "Shotgun Proteomics as a Powerful Tool for the Study of the Proteomes of Plants, Their Pathogens, and Plant–Pathogen Interactions" Proteomes 10, no. 1: 5. https://doi.org/10.3390/proteomes10010005

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

Article Metrics

Back to TopTop