Next Article in Journal
New Insights into Inflammatory Bowel Diseases from Proteomic and Lipidomic Studies
Next Article in Special Issue
PeptideWitch–A Software Package to Produce High-Stringency Proteomics Data Visualizations from Label-Free Shotgun Proteomics Data
Previous Article in Journal
High-Throughput Identification of the Rhodnius prolixus Midgut Proteome Unravels a Sophisticated Hematophagic Machinery
Previous Article in Special Issue
The Power of Three in Cannabis Shotgun Proteomics: Proteases, Databases and Search Engines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

What Room for Two-Dimensional Gel-Based Proteomics in a Shotgun Proteomics World?

1
Medizinisches Proteom-Center, Medical Faculty & Medical Proteome Analysis, Center for Proteindiagnostics (PRODI) Ruhr-University Bochum Gesundheitscampus, 4 44801 Bochum, Germany
2
CBM UMR CNRS5249, Université Grenoble Alpes, CEA, CNRS, 17 rue des Martyrs, CEDEX 9, 38054 Grenoble, France
3
Laboratory of Chemistry and Biology of Metals, UMR 5249, Université Grenoble Alpes, CNRS, 38054 Grenoble, France
*
Author to whom correspondence should be addressed.
Proteomes 2020, 8(3), 17; https://doi.org/10.3390/proteomes8030017
Submission received: 28 June 2020 / Revised: 2 August 2020 / Accepted: 4 August 2020 / Published: 6 August 2020
(This article belongs to the Special Issue Proteomics: Technologies and Their Applications)

Abstract

:
Two-dimensional gel electrophoresis was instrumental in the birth of proteomics in the late 1980s. However, it is now often considered as an outdated technique for proteomics—a thing of the past. Although this opinion may be true for some biological questions, e.g., when analysis depth is of critical importance, for many others, two-dimensional gel electrophoresis-based proteomics still has a lot to offer. This is because of its robustness, its ability to separate proteoforms, and its easy interface with many powerful biochemistry techniques (including western blotting). This paper reviews where and why two-dimensional gel electrophoresis-based proteomics can still be profitably used. It emerges that, rather than being a thing of the past, two-dimensional gel electrophoresis-based proteomics is still highly valuable for many studies. Thus, its use cannot be dismissed on simple fashion arguments and, as usual, in science, the tree is to be judged by the fruit.

1. Introduction

A decade after its introduction in the mid-1970s [1,2], two-dimensional electrophoresis was instrumental in the birth of proteomics [3] Since then, the exponential development of shotgun proteomics, pioneered at the turn of the century [4] and driven in great part by the gigantic increase in the sensitivity of mass spectrometers, has largely superseded two-dimensional (2D) gel-based proteomics. However, although often being referred to as an outdated method, 2D gel-based proteomics still has some merit, as recently reviewed [5,6,7]. The purpose of this review is to expand on this topic and emphasize the positive features that make 2D gel-based proteomics an option to consider in some circumstances.
First of all, the analysis depth is a question to consider, as 2D gel-based proteomics is often criticized on this ground. Modern 2D gels visualized with sensitive techniques (e.g., differential in gel electrophoresis (DIGE) or silver staining) often reach 2500–3000 individual spots. With an average of three spots per protein in eukaryotes [8], this corresponds to ca. 800 gene products. According to shotgun data, 800 proteins correspond themselves to 90% of the cellular protein mass [9], i.e., 90% of the energy invested in the cell in producing proteins. Thus, as a cost function, 2D gel-based proteomics is still a relevant approach, especially when the mechanistic details of the cellular responses, i.e., unveiling in detail the signaling pathways or networks at play, are not the core of the biological question.
Concisely, 2D gel electrophoresis is the only currently available technique that is able to separate complete proteins over a wide pI (isoelectric point) and molecular mass range, with a resolution high enough so that a spot corresponds most often to one major protein. With the onset of high sensitivity mass spectrometry (MS), every spot observed in a 2D map now leads to the identification of several proteins in it. This has been perceived as a concern of variable importance [10,11]. Recent research, carried out in the worst possible setup in terms of multiple identifications (i.e., coupling a high sensitivity MS to highly loaded 2D gels, where the Gaussian spreading of spots and the presence of streaks were maximized) has shown that the most abundant protein in a spot, as detected by MS, most often accounts for >75% of the total signal [12]. Although a previous paper had described that, in 30% of the cases, the most abundant protein accounted for less than 70% of the “protein intensity” [13]. It should be emphasized, however, that such calculations rely on MS-based indexes such as emPAI (exponentially modified protein abundance index), which have been shown to lack accuracy [14,15,16]. This shall not come as a surprise owing to the variations in peptide to mass signal yield, as detailed in the next section.
Furthermore, the corpus literature on identification of proteins from 2D gel spots by the peptide mass fingerprinting approach (reviewed in [17]) further exemplifies that unambiguous identifications are more the rule than the exception in two-dimensional gel electrophoresis (2DGE) proteomics.
Moreover, should a doubt persist, a third separation can always attempt to decipher, more precisely, the protein content of a spot of interest [18,19].
This ability to analyze complete proteins, with a view on their post-translational modifications, is of high theoretical and practical interest in proteomics. In an ideal world, to maximize the biological relevance of the method, proteomics should be able to analyze complete, native, folded proteins with their cohort of post-translational modifications and prosthetic groups. As a matter of fact, there are examples where the simple alteration of a loosely bound prosthetic group, such as an iron-sulfur cluster, can completely change the protein function at constant polypeptide chain [20].
This is unfortunately impossible with current technology, so that only a proxy to the real biological object of interest can be analyzed. In this context, a denatured protein, with its cohort of covalent post-translational modifications, is the best possible proxy, before the pool of all modified forms of the same protein (i.e., the level of the gene product), preceding itself digestion peptides produced from the proteins.
This being stated, the question that arises is to determine to which extent this theoretical advantage tells, knowing that 2D gel-based proteomics has a narrower analysis window than shotgun proteomics in terms of pI, MW (molecular weight), and protein hydrophobicity, is more sample consuming and slower to carry out. The purpose of this review is to investigate this question.

2. Peptides or Proteins—That Is the Question

Most MS methods in proteomics involve the analysis of peptides instead of proteins. This strategy is generally referred to as “bottom-up” analysis [21]. In this approach, the proteins are cleaved enzymatically or chemically into peptides, the mixture is analyzed by MS, then identified in a database search and, if required, quantified and then reclustered into “proteins” (in fact, on a gene product scale) in silico. In contrast, “top-down” proteomics is used to analyze proteins as a whole, as in the 2D-PAGE-based approaches or in newer MS-based approaches [22,23] (a comparison of the two workflows is shown in Figure 1). These strategies make it possible to obtain more complete information on proteoforms, which can be very relevant depending on the scientific question. The term “proteoform” describes all different molecular forms of a protein product of a single gene, including changes due to genetic variations, alternatively spliced RNA transcripts, and post-translational modifications [24].
Bottom-up strategies have evolved from the situation that it is much easier to analyze peptides by MS in high-throughput than proteins. Peptides have more similar physicochemical properties, such as solubility, hydrophobicity, and separation behavior among each other than proteins. After digestion and mass spectrometric measurement, characteristic peptide patterns are obtained for each protein, which are referred to as peptide mass fingerprints [25].
In tandem MS approaches, peptide ion mass spectra (MS1 spectra) are first generated. The peptide ions are then further fragmented inside the mass spectrometer, resulting in fragment ions mass spectra (MS/MS or MS2 spectra). In this way, more specific information is obtained for each peptide and a better identification of the peptides is possible [26]. The generated data are analyzed using database search algorithms, such as SEQUEST [27], Mascot [28], X! Tandem [29], etc., and further processed by analysis programs, such as MaxQuant [30], Proteome Discoverer [Thermo Scientific], or Progenesis QI [Nonlinear]. The results are then peptide spectrum matches (PSMs). In a next step, the obtained PSM data are reclustered into a gene product. This process is called protein inference [31].
Brain T. Chait writes in his 2006 article in Science that “Unfortunately, only a small fraction of tryptic peptides are normally detected…. The bottom-up approach is therefore suboptimal for determining modifications and alternative splice variants” [21] (i.e., proteoforms). This means that by analyzing the peptides, we only obtain a section of all information on the associated proteins and important information on proteoforms is lost.
Apart from the fact that not all peptides of a protein are measured and identified, and the protein information is, therefore, only fragmentary, there is an additional challenge in data analysis: it might not be possible to clearly determine which of the proteoforms present in the sample a peptide can be assigned. The bottom-up approach specifies that the measured peptide data are used to assign to a specific protein. However, a clear assignment between peptide and protein is only possible if the detected peptide is a unique peptide (i.e., if it is a peptide that only occurs in a single protein and is, therefore, clearly specific to that protein at a given time of knowledge). A significant number of peptides are not unique but shared by different proteins in the database, especially in eukaryotic organisms [32]. These shared peptides lead to sets of proteins (protein ambiguity groups), which are created, from the same (sub) set of measured peptides. Finally, without any unique peptide evidence, it cannot be determined, which of the proteins of an ambiguity group was/were actually present in the original sample. Some of the database search algorithms and programs try to solve this issue by using only unique peptides for inference or reporting protein groups or representatives as a result. There have been some developments in recent years that address the problem of protein inference [33,34,35,36,37,38]. It is important for us to emphasize this point again here and to draw the reader’s attention to the fact that the MS and MS/MS data evaluated may need to be viewed critically.
In addition, bottom-up analyses present additional challenges for label-free quantitative (LFQ) proteome analysis. The search algorithms or programs use different strategies to deduce the protein quantity from the peptide quantity. Ultimately, however, the aim is to draw conclusions from the measured intensities of the peptides about the intensity and hence the quantity of the protein and its variants. This means that peptides are regarded as representatives for the proteins. If not only unique peptides are included in this calculation, then there is a risk of incorrect quantification, since the intensities (and thus also quantities) of the shared peptides reflect those of several proteins/proteoforms. However, if only unique peptides are included in the quantitative analysis, of which there may be only a few, then the quantification of the protein may rely on a less reliable data basis. Other strategies approaching this problem use, e.g., the covariation of peptides’ abundances in all samples [39].
We would like to illustrate the problem with a concrete example. In a cerebrospinal fluid (CSF) study [40], which we partially published in 2018, and which aimed to evaluate potential published biomarkers for Parkinson’s disease (PD); we examined the protein “haptoglobin” (Hp). For Hp, two isoforms are described, where isoform 2 differs from the canonical sequence in that amino acids 38–96 are missing. In addition, various glycosylations and disulfide bridges are known, i.e., other proteoforms exist.
Even without consideration for its isoforms, this protein is described as a potential protein biomarker candidate for several neurological diseases such as PD [41,42,43], Alzheimer’s [44], multiple sclerosis [45], hypertrophic cardiomyopathy [46], as well as ovarian cancer [47,48], and many others. This protein had been described as a potential biomarker for PD, but different studies showed a different tendency with regard to regulation in CSF and/or serum [49]. We generally found a very high variability for the Hp in CSF. Our LFQ analysis also revealed that of the 21 peptides assigned to Hp (12 of them unique), some behaved very differently in terms of intensity within a sample (see Figure 2). From these and other data from our projects, which we have critically evaluated again afterwards, we have concluded that it is not always possible to draw clear conclusions about the amount of protein from the amount of peptides. Quantification at peptide level alone might be a possible solution to this problem. In this way, the protein inference step, which can lead to non-uniform assignments as described above, is avoided.
When looking at the Hp spot pattern in a 2D gel, we find that it is distributed over a large number of different spots across the gel [https://world-2dpage.expasy.org/swiss-2dpage/protein/ac=P00738]. A 2DGE-based study of ovarian cancer [47] clearly detected seven potentially disease-relevant well-separated Hp proteoforms (including modified alpha 1 and 2 isoforms) in ascitic fluid of patients. Authors found an association between the number of alpha isoforms and the disease stage of patients. Previously, several studies have already identified Hp and particularly the fucosylated forms in serum of patients and suggested its possible use as a diagnostic biomarker [50,51]. Indeed, only a top-down approach using 2DGE in combination with a fucosylation specific lectin assay allowed for the differential detection/identification of proteoforms, especially alpha subunit expression together with differential levels of fucosylation.
By contrast, and by design indeed, 2DGE proteomics is free from these issues. In this proteomic setup, the quantification step is not carried out by MS, but through the 2D gel images. As proteins and not peptides are separated in 2D gels, proteins are directly quantified on the images. Opposite to peptides, which show a high variability in their detectability and response factor (as exemplified by the different signals observed for different peptides arising from the same protein, which should be equimolecular except for shared peptides), there is an averaging/buffering effect in proteins that makes their quantification less variable from one protein to another.
Thus, besides the identification issues discussed earlier [10,11,12], the quantification issues in 2DGE proteomics lie mainly with the performances of the detection methods used, both in terms of sensitivity and linearity.
Regarding linearity, detection by fluorescence is clearly the best option, as fluorescence can be linear over several orders of magnitude, which is typically what proteomic analyses face. One of the most popular setups for fluorescence detection is the so-called DIGE setup (reviewed in [52]), but there are many different options for fluorescent detection and quantification of proteins after 2DGE, using covalent labelling of proteins or not [53], including detection of the classical Coomassie blue by fluorescence [54]. At equal dye, the comparison of the performances achieved by fluorescence [54] to those achieved by light absorption [55] clearly shows the power of fluorescent detection. Sensitive fluorescence detection requires however expensive hardware, so that classical detection by light absorption, which can be achieved on a classical scanner, is often favored. In this setup, organic dyes are plagued by a relatively low sensitivity [55], and are superseded in this respect by silver staining [56]. Silver staining however suffers from a low response curve [56], so that the quantitative differences highlighted by silver-stained 2D gels may be an under-representation of the true quantitative changes at the protein levels. This further emphasizes the fact that arbitrary thresholds should be avoided for quantitative proteomic analyses [57].
Incidentally, this combination of high sensitivity detection and homogeneous response of proteins makes 2DGE proteomics an attractive choice for a niche application in which the aim is not to compare different samples, but to quantitatively analyze one sample for its different proteins. One example of this case is represented by the analysis of therapeutic protein batches, where 2DGE has been successfully applied [58,59]. Indeed 2DGE is very good at detecting not only the predictable, such as contaminant host cell proteins, which can be analyzed by shotgun proteomics [60], but also the more difficultly-predictable, such as modifications of the therapeutic protein of interest, which can translate into the appearance of new protein spots.
In summary, it can be said that the current bottom-up strategies have their advantages and disadvantages. Even though it is easier to analyze peptides instead of proteins, the subsequent qualitative or quantitative data analysis must be critically reviewed and should ideally be inspected manually in order to avoid betting on the wrong horse in a subsequent extensive evaluation of the results. Indeed, important systemic technical biases have been documented for shotgun proteomics [61]. Conversely, the reliability of 2DGE proteomics, in this respect, is an often underlooked and underestimated advantage of the setup.

3. When an Unpredictable Subset of Proteins Is to Be Analyzed: The Example of Immunome/Allergome Studies

The first area in which 2D gel-based proteomics shows interesting performances are the studies where the point of interest is to decipher which proteins of a pathogen/allergen are targeted by the immune system of the host. The great advantage of 2D gel-based proteomics in this type of research is that it uses the host antibodies as an analytical reagent to detect the proteins of interest, and not as a preparative reagent to select the proteins of interest. Using antibodies as preparative reagents is associated with many problems associated with the constraints brought by the solid supports that must be used, which bind a wide variety of proteins, leading to a very extensive and severe background [62,63]. These problems do not happen when antibodies are used as analytical reagents because of the tricks (e.g., saturation with other proteins) that can be applied in this scheme. The combination of 2D gels to display the target organism proteins and 2D blots to highlight the proteins recognized by the host immune system, looping back to the target organism protein 2D gels to identify then the proteins of interest, has proven very efficient in many cases. Although theoretically straightforward, this process is practically not as easy as it may seem, as it requires a very rigorous matching of the 2D blot pattern on the gel pattern to avoid any mistake at this stage. This has led to the development of several methods, from the detection of the total protein pattern on the 2D blots prior and/or after the immunodetection (e.g., in [64]) to the partial blotting process, in which the very same 2D gel is used, both for generating the 2D reference pattern for MS identification and the 2D blot for immunodetection [65]. In the immune responses to pathogens, work has been carried out on bacteria [66,67,68,69,70], fungi [71,72,73], and parasites [74,75]. Such studies have always reached their initial goal of providing the major immunogens from the pathogen under study, and have sometimes resulted in valuable clinical advances [71,76,77].
The situation is very similar in the allergy field, where as a further refinement only the IgEs (immunoglobulin E) of the patients are selectively detected. The response to various allergens has been studied, as exemplified recently in [78,79,80,81], and reviewed earlier in [82].
Lastly, this approach has also been used in diseases with an autoimmunity component to identify autoantigens in various pathological contexts such as arthritis (e.g., in [83,84,85,86,87]), multiple sclerosis (e.g., in [88,89]), or other diseases [90,91].
These examples show the convenience and interest of 2D gel-based proteomics in the immunome/allergome field, where experimental schemes based on shotgun proteomics are plagued by the issues linked to antibody immobilization mentioned above.

4. Going to the Essence of Proteomics: Proteoforms and Post-Translational Modifications

If comprehensiveness is defined from a genetic/genomic standpoint, i.e., from the 1941 one gene-one protein dogma [92] and, thus, from the number of gene products identified, proteomics lacks comprehensiveness, even in its deepest shotgun versions, compared to transcriptomics, which has reached comprehensiveness through the use of deep sequencing [93]. Thus, the value of proteomics lies in the fact that it analyzes a better proxy of the cellular functions, i.e., proteins. In doing so, all proteomics setups integrate the regulations that take place at the translational level. However, it appears more and more shiningly that a great deal of regulations occur at the post-translational level, mostly through post-translational modifications (PTMs). There are literally dozens of different modifications, which are enzyme catalyzed for some of them but sometimes not [94,95,96]. Among the enzyme-catalyzed modifications, phosphorylation is the one described for the longest time and which role is best known. Glycosylation is also known for a very long time, but its daunting complexity [97] has made progress slower in understanding the full scope of its functions. More recently other modifications such as methylation [98,99], acetylation [100,101,102,103], or other acylations [104,105,106] have been described and modulate protein localization and/or function.
This increased recognition of the importance of PTMs has led to the concept of proteoform(s), i.e., a protein backbone bearing a precise combination of PTMs. Ideally, proteomics should focus on the study of proteoforms and not on gene products. This is easier said than done, owing to the number of technical and operational challenges that arise when studying proteoforms. In this context, any tool that is able to separate a protein into a subset of proteoforms even if this separation is not complete, is a step further in moving proteomics in this direction. Although some PTMs are electrophoretically neutral (e.g., Lys/Arg methylation, Cys acylation) many are not, such as Lys acylation or phosphorylation. Because of its resolution in the isoelectric focusing (IEF) dimension, which is where the electrophoretic impact of PTM is most easily seen, 2D electrophoresis is, therefore, able to separate many protein variants on the basis of their pI. Consequently, most proteins appear not as a single spot, but as a trail of spots, with an average of 3 spots/protein in mammalian cellular proteins [8]. This relatively low number shall not be taken as an average number of proteoforms, as IEF just counts the number of modifications but gives no information on their localization. For example, proteoforms bearing the same number of phosphorylations, but at different positions, will be merged into a single spot. In fact, full characterization of discrete protein spots have shown that a single spot can be a mixture of differently-modified proteins [107].
Despite this lack of resolving power when compared to ideality, the separation afforded by 2DGE is clearly an advantage when trying to bring proteomics closer to the proteoform world [5,6,7]. Consequently, there are literally thousands of scientific articles that bring together the keywords “2D electrophoresis” and “post-translational modifications”, and this article will of course not aim at citing all of them. It will rather cherry pick some papers that appear of interest in the dimensions selected and highlighted here.

4.1. Guidelines for Use

The advantage of being able to resolve proteins as several discrete spots brings its own set of challenges and issues that must be taken into account when using 2DGE in proteomics. With an average of three spots/protein [8], when 2DGE is used in differential proteomics, the most frequent situation is that only one (or a few) protein spots will change under the biological condition of interest, but not all the spots corresponding to the same protein. Murphy’s Law being what it is, the modulated spots are generally the weakest and the most modified, i.e., the farthest from the theoretical position of the protein. There is thus a danger to mistake the part for the whole, and to overclaim “the amount of protein X is changed” while the true situation is “the amount of form Y of protein X is changed”. In order to get a good appraisal of the real situation, it is, therefore, highly advisable to try to look for other forms of the protein of interest in the 2D gel space. This is more or less easy to do depending on the chemical characteristics of the proteins of interest, leading to the notion of separation cone in 2D electrophoresis. Indeed, depending on the number of charged amino acids that they contain, proteins are more or less “buffered” in the IEF dimension. Thus, the change brought by the same PTM will result in a different spacing in the IEF dimension depending on the protein. As an example, a high molecular weight protein such as matrin 3 contains many charged amino acids. Consequently, a PTM will lead to a minor spot spacing, so that the modified forms of the protein will appear as an easily recognizable train of spots [108]. Conversely, a lower molecular weight protein such as triosephosphate isomerase contains a lower number of charged amino acids, so that a single modification brings a much larger spacing on the 2D gels [109], which is much more difficult to take into account. Thus, as a first approximation, the spacing induced by PTM decreases when the molecular weight of the proteins increases, leading to the concept of separation cone. As always in the protein world, the situation is more complex, as proteins of the same molecular weight can have different charge densities and thus lead to a different spacing. Thus, a cautious and reasonable attitude when using 2DGE-based proteomics is, when an interesting spot is found, to try to figure out where the un-modulated major spots accounting for the bulk of the protein are, and to discuss the results accordingly.

4.2. Hypothesis Validation: Getting the Most from the 2DGE Data

Thus, the usual outcome of a 2DGE proteomic experiment is a list of modulated spots, and most often, the total amount of the protein of interest is not modulated in the experiment. This shall not mean the end of the story, as this situation is precisely what can be expected from a landscape where PTM play a major role in the modulation of protein activities, and this can be recognized upfront in very different perspectives, ranging from the description of modified protein forms in a therapeutic protein (e.g., in [59]) to functional differences (e.g., in [110]). However, the impact of PTM cannot be predicted a priori. Both for phosphorylation and for acetylation, sometimes the modifications increase the activity of the protein and sometimes it decreases it. Moreover, the classical LC-MS/MS (liquid chromatography tandem mass spectrometry) analysis of the spots carried out to identify the proteins of interest usually does not lead to the identification of the PTMs. In many cases, this is due to the fact that modifications are excluded from the analysis of the MS/MS spectra to limit the search space and the probability of false positives.
The absence of knowledge of the modification does not mean that the result is without biological interest. It just means that it shall be validated, directly or indirectly. In this respect, proteomic results are all equal, regardless of the setup used to produce them. By looking at quantitative changes, proteomics just exploit the homeostasis principle. As the composition of living systems tends to be constant, then if a perturbation of the system brings changes in the composition of the system, then these changes are relevant and important in the response to the perturbation. This ideal statement is tarnished by the fact that, as for all large-scale techniques, proteomics comes with its share of false positives and artifacts, so that a proteomic screen should be seen mostly as a hypothesis generator, and hypotheses must be validated.
This is most often a weak point of many proteomic papers, where the validation is kept to a minimum, to say the least, and most often consists of real-time quantitative polymerase chain reaction (RT-qPCR) and/or western blotting. This represents in a sense a kind of circular validation, in which a change of abundance detected by proteomics is just confirmed as being a change of abundance by another technique, without going much further in terms of biochemical knowledge. This may be due to the fact that validation is often research-intensive, and this poses in turn the problem of confidence in the proteomic results for validation.
One theoretical solution to this confidence problem would be to perform the proteomic analysis by two different techniques with different biases, and to consider what is found in common by both techniques as reliable results. This approach unfortunately does not apply to the combination 2DGE proteomics/shotgun proteomics. If the biases are different indeed, the two techniques target different analytes so that the results are not really comparable. There are, therefore, very few articles in which both approaches are used on the same samples, but these few are worth analyzing in detail.
In their work on schizophrenia [111], Martins de Souza et al. used both shotgun proteomics and 2DGE proteomics. As mentioned in their results, nine out of the ten differences that they found in 2DGE proteomics were not found by the shotgun proteomics screen. This should not come as a surprise in the light of the PTM effects described above. The reverse question is however worth being investigated. It is often stated that shotgun proteomics describes variations that are missed by 2DGE proteomics because of the greater analysis depth of shotgun proteomics. While this is certainly true in many cases, this study strongly suggests that it is not always the case. For example, phosphoglycerate mutase and transketolase are found modulated in schizophrenia by the shotgun proteomic screen, but not in 2DGE. These proteins belonging to the glycolysis and pentose phosphate pathways, respectively, they should be abundant proteins and detected easily in 2DGE, which they are indeed in other mammalian systems [112]. The fact that they were not detected in the 2DGE screen in the Martins de Souza et al. study [111] just means that they were not detected as variable, as 2DGE proteomics investigates only the proteins that change, and not all the detectable proteins, as shotgun proteomics does. There is thus an important discrepancy between the shotgun and 2DGE proteomics screen in the Martins de Souza et al. study concerning these two proteins, which are either a false positive of the shotgun proteomics or a false negative of the 2DGE proteomics. In the absence of enzyme activity data in the paper it is impossible to know which is correct.
Enzymes are indeed a very good illustrative case. They are regulated by modifications [100,113], so that they can be perceived as a good model for proteins in general, and the validation is often easy via the enzymatic activity. It can be argued that enzymes often belong to the “déjà vu in proteomics” [114], but this just points out their importance in stress response [115]. Indeed, the fact that the response is generic does not mean that it is unimportant. In this respect, it has been shown that preventing the induction of glyceraldehyde 3-phosphate dehydrogenase (GAPD), the epitome of a “déjà vu” protein, inhibits cell survival upon genotoxic stress [116].
Despite the relative easiness of validation through the activity, direct validation of proteomic results by this means is relatively scarce in the scientific literature. However, the few existing results often confirm that enzyme activities are altered even when the proteomic screen shows a change in one or a few spots corresponding to the enzyme, without necessarily a global change in the total amount of the protein [109,117,118,119,120,121]
Beyond these examples, an even more precise result can be reached when the enzyme activities are measured directly on the 2D gel spots after in gel renaturation, following the zymography principles [122,123,124]. Although kinetic measurements are difficult to carry out via such techniques, they allow mapping which spots bear the activity, which can be a very relevant information. As not all enzymes lend themselves to such in vitro renaturation and to gel assays, there are only a few examples in the literature [125,126,127,128,129].
Hopefully, validation of 2DGE proteomic results is not limited to enzymes, and indirect validations can be of high interest. As the name says, indirect validation does not validate directly the activity of the proteins of interest, but consequences on the function(s)/structure(s) in which these proteins play a role. One typical example is represented by the actin cytoskeleton. Actin being one of the most abundant cellular proteins, quantitative changes in the total actin amount are seldom observed. However, there are numerous proteins whose activities control the shape and dynamics of the actin cytoskeleton, and these proteins may change in their abundance/modification profile under various biological conditions. Consequently, a change in the structure of the actin cytoskeleton can be expected, and probed by confocal microscopy, as described in [130,131,132]. Detailed analysis of target proteins is also an interesting option [108,133]. Metabolic activity can also be a very good indirect validation (e.g., in [111,134,135]), and helps indeed in solving issues that are undecidable through proteomics and even enzyme activities. For example, when an increase in central metabolic enzymes is detected, does it mean that the overall energy consumption increases or that the system is trying to compensate for a defect? Measuring the glucose consumption [135] or the pyruvate level [111] solves these issues.
Another surprising benefit of 2DGE for hypothesis validation can be summarized by the motto from the architect Mies van der Rohe: “less is more”. For reasons that are linked to the analysis depth but also with some intrinsic yet not well-understood features of shotgun proteomics, this proteomic setup leads to very large lists of modulated proteins (often counting in hundreds) in differential proteomics. Such large lists are very difficult to handle by hand, so that computerized analysis of the data is the norm in shotgun proteomics. In addition to the problems caused by the variable quality of annotations, which percolate into the quality of the final results, computerized analysis is by essence collective, and the pathways that include a large number of proteins are selected over those with a low number of proteins. While normally robust, this approach leaves no room for serendipitous discoveries.
By contrast, 2DGE proteomics often yields lists of modulated proteins that count in tens instead of hundreds. Although computerized analysis can be applied to 2DGE proteomics with success (e.g., in [117,132,135]), such lists can be scrutinized for individual proteins, and sometimes a few proteins belonging to different pathways can lead to unraveling novel mechanisms. A good example is represented by pulling the thread from coactosin-like protein to cytoskeleton reorganization [132]. Proposal of mechanisms for zinc genotoxicity is also a good example where a few proteins can lead to mechanistic evidences, provided that substantial validation is carried out [131].

4.3. The Quest for PTMs: Unsupervised PTM Analysis as a Strength of 2DGE Proteomics

A step further in the use of the ability of 2DGE to separate modified forms of proteins resides in the identification of the modification(s) and of its (their) position(s) of the proteins. As mentioned above, taking modifications into account is one of the great strengths of proteomics over transcriptomics, for example. In order to perform this task that can be daunting, two strategies can be devised, which can be summarized as supervised vs. unsupervised search for modifications. In the supervised search, the type of modification that will be search for is defined upfront. For example, one can search for phosphorylations only, or for acetylations only, etc. There are two important advantages in such a supervised search. The first is that the search space for modified peptides, although greatly increased compared to unmodified peptides only, remains reasonable. The second and even more important advantage is that reagents able to select the class of modified peptides of interests become usable, with a great increase in the sensitivity of the approach. Such approaches have been reviewed [136,137,138,139], so that we will not enter into any further detail. Of course, the major limitation of the supervised search is that you only look at what you are allowed to look at.
Conversely, unsupervised PTM search sets no limit at the type of modifications that can be searched for, and relies on the ability of MS/MS to take into account any modification that brings a mass difference (i.e., any modification in fact). Such an approach is clearly out of reach by direct LC-MS/MS at a proteome-wide scale, because the search space becomes much too large. However, direct unsupervised identification of modifications has been instrumental in the discovery of new modifications, quite often using histones as target proteins. Histones offer the triple advantage of being very abundant in cells, bear only few sequence variations and easy to purify by acid extraction, leading to a small enough search space. It is therefore no surprise that modifications such as methylation, acetylation [140], propionylation, butyrylation [141], malonylation, succinylation [142], or crotonylation [143], have been identified first on histones in an unsupervised way and then searched for proteome-wide in a supervised way once the suitable reagents have been developed.
The histone example shows that the key is to restrict the search space by purifying the protein of interest. In this context, 2DGE can be viewed as a micropreparative tool able to separate protein modified forms, where modifications can be searched for in an unsupervised fashion by MS/MS. Thus, 2DGE has been instrumental in evidencing protein deamidation [144], but its abilities are not restricted to this modification. In this strategy, the name of the game is to separate a modified protein form, then to search for the modification(s) that may explain the observed change in the biochemical coordinates (most often the pI). It is perfectly possible to identify phosphorylations at this stage (e.g., in [108,120,145,146,147]), and sometimes phosphorylations that have been undetected by targeted phosphoproteomic approaches can be identified by this approach (e.g., in [147]). Of course deamidation can also be identified (e.g., in [148]) but less classical modifications can be detected too. For example, this combination of 2DGE and MS/MS has been instrumental for the identification of strong cysteine oxidation [149], a modification that strongly modifies protein function [150], for which no enrichment tool has been found to date but that has been described in several proteins [151,152,153,154,155]. Other modifications, such as succinylation, have been found by this approach [154]. Last, but certainly not least, extensive PTM mapping has been performed by this unsupervised approach in some cases [107,156].
Another case of non-classical PTM is represented by covalent adducts derived from toxic chemicals and modifying nucleophilic residues on proteins such as cysteine and lysine. There are cases where the chemical itself is known and electrophilic enough to react directly with amino acids, leading to possible supervised search strategies [157]. However, many other situations exist. For example, the chemical can be reactive but not lend itself to targeted approaches. More often, the chemical of interest itself is not reactive, but one of its metabolites is, which clearly complicates the situation. In such cases 2DGE proteomics can be an interesting solution to the problem, either by analyzing in detail the modified proteins to find the modification [158], or by detecting the modified protein forms via blotting and the use of an anti-hapten antibody [159,160], or using a radioactively-labelled chemical [161,162,163,164,165,166,167,168,169]. In such approaches, the precise identification of the modified peptides is often not carried out. Despite this relative lack of precision, these approaches provide the additional information of the source and gross chemical nature of the modifications, compared to classical 2DGE proteomics.
There are also intermediate situations between completely supervised searches for PTMs and completed unsupervised strategies. For example 2DGE can be used as a micropreparative tool to enrich into modified proteins prior to digestion and enrichment of modified peptides through dedicated approaches [170]. The reverse scheme, i.e., selection of modified proteins prior to analysis by 2DGE, has also been used [171], but with no precise identification of the modification sites. The same lack of precise site identifications is often observed in approaches using 2D blotting with modification specific antibodies (e.g., in [172,173]) even if now some authors make the required additional studies to map the modification sites on the target proteins [174].
Another interesting case is represented by protein carbonylation. Carbonylation of proteins can occur by a variety of mechanisms ranging from direct amino acid degradation [175,176] to conjugation to oxidized lipids [177] or unsaturated aldehydes [178]. These diverse chemistries make targeted approaches difficult. However, a popular way of identifying carbonylated proteins, without going into the details of the modified peptides, however, uses conjugation of the protein carbonyls to a hapten (usually biotin of dinitrophenyl) via the coupling of the carbonyl to a hydrazide or hydroxylamine moiety, then display of the proteins on a 2D gel, blotting and identification of the modified proteins by an anti hapten reagent (antibody or avidin). This approach has been used in a variety of biological situations (e.g., in [179,180])

4.4. The Case of Protein Truncation

Besides the generally taken into account case where chemical groups are grafted on the proteins, as mentioned above, an important but underestimated PTM is represented by protein truncation and cleavage. Maybe the epitome of this situation is represented by trypsin itself. After the classical activation of trypsinogen into trypsin [181], trypsin undergoes a series of self-cleavages [181], resulting into a protease with a low chymotrypsin activity [182] before final inactivation by autolysis. This simple example shows that besides regulating the half-lives of the proteins, cleavage can also alter their activity. There is therefore considerable interest in investigating protein cleavage and truncation [183], and dedicated shotgun approaches to do so have been proposed [184,185,186]. However, these N-terminomics protocols make some implicit assumptions, e.g., that the fragment containing the neo N-terminus after cleavage is stable enough to be detected, and that the neo N-terminal peptide has features (e.g., length, hydrophobicity) that make it detectable directly in LC-MS/MS. Complementary protocols looking for C-termini have been proposed [187], but are of much lower sensitivity.
By contrast, using 2DGE for the study of protein truncation makes the simple assumption that at least one of the protein fragments will fall in the separation space and will be sufficiently different from the parent protein to be separated from it. Noteworthy, 2D zymography has often given evidence for truncated proteins being enzymatically active [125,128,129]. Thus, 2DGE proteomics has been used to study protein cleavage, sometimes in a supervised way to study specifically the action of specific proteases [188,189,190], but more often in an unsupervised format to detect increased spots in the condition of interest, which are then identified as cleavage products (e.g., in [191,192]). This approach has been used with success in the food area to assess the post-mortem degradation of muscles, for example in meat [193,194] or in fish [195,196,197,198].
A summary of strengths and weaknesses of 2DGE-based and shotgun proteomics is given in Table 1.

5. 2DGE Proteomics in the Most Difficult Field: Clinical Proteomics

The field of clinical proteomics is huge, rewarding, but difficult, as proteomics faces the full force of human variability in this field. Indeed, biomarker discovery studies, independent from which method they use, while leading to a continuously growing number of potential candidates, most frequently originate in academic research settings. This is due to challenges in the transitioning of the results “from bench to bedside” which requires large-scale validation and clinical trials. Those studies are difficult to accomplish by a single academic lab or even institution [199] and require considerable resources. Overall, the complexity of translational research has turned out being the bottleneck for the implementation of clinical tests. Consequently, many potential markers are identified and published but require further investigation. A comprehensive overview for challenges in biomarker discovery studies is given in e.g., [200,201,202].
Indeed, in the subfield of cancer biomarker discovery, there are still some long-term success stories using 2DGE to report, although the following few selected examples are of course not a comprehensive review of the hundreds of publication in the field of 2DGE clinical proteomics:
The great potential of 2DGE in clinical research was already demonstrated by Charrier et al. in 1999 [203]. Authors were interested in the discrimination of prostate cancer (PCa) vs. benign prostate hyperplasia (BPH), which is a nonmalignant form of prostate disease. It was known that PSA (prostate specific antigen) the main marker for PCa is able to bind protease inhibitors in serum and this binding can be used for discrimination between PCa and BPH on basis of the free to total PSA ratio. Charrier and colleagues detected substantial numbers of cleaved (inactive) proteoforms of PSA being relevant for the diagnosis of PCa. An extension of the study to more than 90 patients showed that in BPH significantly more low-molecular weight PSA forms were present in serum compared to PCa allowing for a significantly improved diagnosis of PCa [204]. Those findings were only possible with top-down approaches like 2DGE. Ironically, these findings could not be translated into a classical biomarker kit because of the difficulty of the classical formats to handle proteoform and especially cleavage issues.
In addition to this example the group around Tadashi Kondo in Japan has used 2DGE in many cancer-related studies (reviewed in [205]). They investigated, e.g., esophageal cancer [206], lung cancer [207], and liver cancer [208,209,210]. For gastrointestinal stromal tumors (GIST) they could identify pfetin as a biomarker for postoperative recurrence [211]. Pfetin could be further verified in >500 cases in seven hospitals and a monoclonal antibody was developed for immunostaining and commercialization [212]. Clinical relevance and significance of pfetin has since been validated in prospective clinical trials [213].
Another example of the efforts required in validating results from 2DGE proteomics toward clinical applications is represented by the work carried out on cerebral strokes. From the first exploratory work using 2DGE proteomics [214], it took from one year to over a decade of work to reach an encouraging and relevant level of clinical validation [215,216,217,218,219].
We ourselves have worked in the field of clinical proteomics for years now. Some studies were also performed using 2DGE. The most successful studies were those conducted in close collaboration with excellent pathologists. One of the studies involved the identification and verification of a protein biomarker candidate for liver cirrhosis. Here we started with a 2DGE-based study of manually microdissected cirrhotic liver tissue from 7 patients identifying human microfibril–associated protein 4 (MFAP-4) as a potential diagnostic marker for hepatic cirrhosis [220]. In a next step, MFAP-4 could be verified as a biomarker in sera of patients with liver cirrhosis of different etiology and of different stages by Western-blotting and ELISA in >100 independent samples. MFAP-4 could be further confirmed in an extensive follow-up study 7 years later [221] focusing on its potential as biomarker for the differentiation of no to moderate (F0–F2) and severe fibrosis stages and cirrhosis. Here, MFAP-4 was verified in a retrospective study including n = 542 hepatitis C patients in serum using an AlphaLISA immunoassay. The clear result was that MFAP-4 in combination with existing tests leads to a more accurate non-invasive diagnosis of hepatic fibrosis and allows a cost-effective disease management in the era of new direct acting antivirals.
Complete success stories arising from proteomics in the field of clinics are difficult to report, either because the gap between what could be achieved at the academic level and what was required by the biomarker industry could not be closed (MFAP-4 example), or because the best clinical value was offered by a combination of markers, which is often felt as cost-ineffective [219], or because the relevant results offered by 2DGE proteomics at the proteoform level could not be easily transposed in a classical, user-friendly bioassay kit [204].

6. As a Conclusion: May Look Slow and Cumbersome, but Still Valuable If Not Irreplaceable

When trying to summarize, concisely, all the above examples, a few trends emerge on the utility of 2DGE proteomics in biological sciences. The outstanding ability of 2DGE proteomics is its ability to analyze proteoforms at low cost and with a relatively high efficiency, thereby moving away proteomics from areas where it falls in concurrence with sequencing-based transcriptomics, which is faster and much more comprehensive. The limitations of 2DGE proteomics in terms of protein hydrophobicity, molecular weight and pI extremes are well known for decades, but are also shared by MS-based top-down proteomic approaches, and are indeed linked to the physico-chemical behavior of proteins themselves. Consequently, the fact that electrophoresis is more efficient than chromatography to separate and analyze proteins (while the reverse is true for peptides) is as true now than it was at the end of the 20th century when it was published [222]. It is, therefore, no surprise that low-resolution variants of electrophoretic methods have been used as a prefractionation tool in MS-based top-down proteomics [223]. The second strength of 2DGE proteomics is its easy interface with MS, which has revolutionized the degree of detail and precision that can be reached with 2DGE proteomics, without the need of any peptide enrichment upfront (see for example [107]), but also with western blotting, which remains a very efficient tool in many proteomic studies, such as the immunome/allergome ones cited above. Moreover, a positive consequence of the fact that 2DGE proteomics takes proteoforms into account, and is, therefore, closer to the cellular physiology, and is felt downstream of the proteomic phase, when functional validation must be carried out. A perspective for future use of 2DGE would be as an enrichment tool prior to top-down MS; therefore, analyzing complete proteins eluted from the 2D gels instead of the peptides produced by in-gel digestion of the proteins. This would couple the superior separating power of 2DGE [222] with the exquisite details provided by top-down MS [224]. It must be underlined that this approach has been described in the past with some success [225,226]. However, it is less straightforward that it may seem. Protein elution from 2D gels require SDS (sodium-dodecyl-sulfate), which must be removed prior to the top-down MS [227], with serious risks of protein losses. Furthermore, an often neglected problem lies in the oxidative modifications of the proteins that can be brought by the electrophoretic process itself [228], which will complicate the downstream top-down analysis and requires special precautions to limit this artifact [228,229].
Nevertheless, and overall, the tradeoff between 2DGE proteomics and shotgun proteomics exchanges details and precision against speed and analysis depth in terms of number of gene products, and this is a choice to be seriously considered beyond fashion.

Funding

The authors were supported by P.U.R.E. (Protein Research Unit Ruhr within Europe) and ProDi (Center for protein diagnostics), Ministry of Innovation, Science and Research of North-Rhine Westphalia, Germany, by the “Deutsche Parkinson Gesellschaft”, by the H2020 project NISCI, (GA no. 681094) and by the ISF program of the European Union. This work was also funded by the CNRS and the University Grenoble Alpes.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

2Dtwo dimensional
2DGEtwo-dimensional gel electrophoresis
BPHbenign prostate hyperplasia
CSFcerebrospinal fluid
DIGEdifference in-gel electrophoresis
emPAIexponentially modified protein abundance index
GAPDHglyceraldehyde 3-phosphate dehydrogenase
Hphaptoglobin
IEFisoelectric focusing
IgEimmunoglobulin E
LC-MS/MSliquid chromatography tandem mass spectrometry
LFQlabel-free quantification
MFAP-4microfibril–associated protein 4
MSmass spectrometry
MS1peptide mass spectrum
MS2 or MS/MStandem mass spectrum
MWmolecular weight
PAGEpolyacrylamide gel electrophoresis
PCaprostate cancer
pIisoelectric point
PDParkinson’s disease
PSMpeptide spectrum match
PTMpost-translational modification
RT-qPCRreal-time quantitative polymerase chain reaction
SDSSodium-dodecyl-sulfate

References

  1. MacGillivray, A.J.; Rickwood, D. The heterogeneity of mouse-chromatin nonhistone proteins as evidenced by two-dimensional polyacrylamide-gel electrophoresis and ion-exchange chromatography. Eur. J. Biochem. 1974, 41, 181–190. [Google Scholar] [CrossRef] [PubMed]
  2. O’Farrell, P.H. High resolution two-dimensional electrophoresis of proteins. J. Biol. Chem. 1975, 250, 4007–4021. [Google Scholar] [PubMed]
  3. Rabilloud, T. Paleoproteomics explained to youngsters: How did the wedding of two-dimensional electrophoresis and protein sequencing spark proteomics on: Let there be light. J. Proteom. 2014, 107, 5–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yates, J.R.; McCormack, A.L.; Schieltz, D.; Carmack, E.; Link, A. Direct analysis of protein mixtures by tandem mass spectrometry. J. Protein Chem. 1997, 16, 495–497. [Google Scholar] [CrossRef] [PubMed]
  5. Rogowska-Wrzesinska, A.; Le Bihan, M.C.; Thaysen-Andersen, M.; Roepstorff, P. 2D gels still have a niche in proteomics. J. Proteom. 2013, 88, 4–13. [Google Scholar] [CrossRef] [PubMed]
  6. Oliveira, B.M.; Coorssen, J.R.; Martins-de-Souza, D. 2DE: The phoenix of proteomics. J. Proteom. 2014, 104, 140–150. [Google Scholar] [CrossRef]
  7. Zhan, X.; Li, B.; Zhan, X.; Schlüter, H.; Jungblut, P.R.; Coorssen, J.R. Innovating the Concept and Practice of Two-Dimensional Gel Electrophoresis in the Analysis of Proteomes at the Proteoform Level. Proteomes 2019, 7, 36. [Google Scholar] [CrossRef] [Green Version]
  8. Hoogland, C.; Mostaguir, K.; Sanchez, J.C.; Hochstrasser, D.F.; Appel, R.D. SWISS-2DPAGE, ten years later. Proteomics 2004, 4, 2352–2356. [Google Scholar] [CrossRef]
  9. Beck, M.; Schmidt, A.; Malmstroem, J.; Claassen, M.; Ori, A.; Szymborska, A.; Herzog, F.; Rinner, O.; Ellenberg, J.; Aebersold, R. The quantitative proteome of a human cell line. Mol. Syst. Biol. 2011, 7, 549. [Google Scholar] [CrossRef]
  10. Campostrini, N.; Areces, L.B.; Rappsilber, J.; Pietrogrande, M.C.; Dondi, F.; Pastorino, F.; Ponzoni, M.; Righetti, P.G. Spot overlapping in two-dimensional maps: A serious problem ignored for much too long. Proteomics 2005, 5, 2385–2395. [Google Scholar] [CrossRef]
  11. Hunsucker, S.W.; Duncan, M.W. Is protein overlap in two-dimensional gels a serious practical problem? Proteomics 2006, 6, 1374–1375. [Google Scholar] [CrossRef] [PubMed]
  12. Zhan, X.; Yang, H.; Peng, F.; Li, J.; Mu, Y.; Long, Y.; Cheng, T.; Huang, Y.; Li, Z.; Lu, M.; et al. How many proteins can be identified in a 2DE gel spot within an analysis of a complex human cancer tissue proteome? Electrophoresis 2018, 39, 965–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Thiede, B.; Koehler, C.J.; Strozynski, M.; Treumann, A.; Stein, R.; Zimny-Arndt, U.; Schmid, M.; Jungblut, P.R. High resolution quantitative proteomics of HeLa cells protein species using stable isotope labeling with amino acids in cell culture (SILAC), two-dimensional gel electrophoresis (2DE) and nano-liquid chromatograpohy coupled to an LTQ-OrbitrapMass spectrometer. Mol. Cell. Proteom. MCP 2013, 12, 529–538. [Google Scholar] [CrossRef] [Green Version]
  14. Li, J.J.; Bickel, P.J.; Biggin, M.D. System wide analyses have underestimated protein abundances and the importance of transcription in mammals. PeerJ 2014, 2, e270. [Google Scholar] [CrossRef] [Green Version]
  15. Liu, Y.; Beyer, A.; Aebersold, R. On the Dependency of Cellular Protein Levels on mRNA Abundance. Cell 2016, 165, 535–550. [Google Scholar] [CrossRef] [Green Version]
  16. Lawless, C.; Holman, S.W.; Brownridge, P.; Lanthaler, K.; Harman, V.M.; Watkins, R.; Hammond, D.E.; Miller, R.L.; Sims, P.F.G.; Grant, C.M.; et al. Direct and Absolute Quantification of over 1800 Yeast Proteins via Selected Reaction Monitoring. Mol. Cell. Proteom. MCP 2016, 15, 1309–1322. [Google Scholar] [CrossRef] [Green Version]
  17. Thiede, B.; Höhenwarter, W.; Krah, A.; Mattow, J.; Schmid, M.; Schmidt, F.; Jungblut, P.R. Peptide mass fingerprinting. Methods San Diego Calif. 2005, 35, 237–247. [Google Scholar] [CrossRef]
  18. Butt, R.H.; Coorssen, J.R. Postfractionation for enhanced proteomic analyses: Routine electrophoretic methods increase the resolution of standard 2D-PAGE. J. Proteome Res. 2005, 4, 982–991. [Google Scholar] [CrossRef]
  19. Colignon, B.; Raes, M.; Dieu, M.; Delaive, E.; Mauro, S. Evaluation of three-dimensional gel electrophoresis to improve quantitative profiling of complex proteomes. Proteomics 2013, 13, 2077–2082. [Google Scholar] [CrossRef]
  20. Haile, D.J.; Rouault, T.A.; Tang, C.K.; Chin, J.; Harford, J.B.; Klausner, R.D. Reciprocal control of RNA-binding and aconitase activity in the regulation of the iron-responsive element binding protein: Role of the iron-sulfur cluster. Proc. Natl. Acad. Sci. USA 1992, 89, 7536–7540. [Google Scholar] [CrossRef] [Green Version]
  21. Chait, B.T. CHEMISTRY: Mass Spectrometry: Bottom-Up or Top-Down? Science 2006, 314, 65–66. [Google Scholar] [CrossRef] [PubMed]
  22. Fornelli, L.; Toby, T.K.; Schachner, L.F.; Doubleday, P.F.; Srzentić, K.; DeHart, C.J.; Kelleher, N.L. Top-down proteomics: Where we are, where we are going? J. Proteom. 2018, 175, 3–4. [Google Scholar] [CrossRef] [PubMed]
  23. Toby, T.K.; Fornelli, L.; Kelleher, N.L. Progress in Top-Down Proteomics and the Analysis of Proteoforms. Annu. Rev. Anal. Chem. 2016, 9, 499–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. The Consortium for Top Down Proteomics; Smith, L.M.; Kelleher, N.L. Proteoform: A single term describing protein complexity. Nat. Methods 2013, 10. [Google Scholar] [CrossRef] [Green Version]
  25. Pappin, D.J.C.; Hojrup, P.; Bleasby, A.J. Rapid identification of proteins by peptide-mass fingerprinting. Curr. Biol. 1993, 3, 327–332. [Google Scholar] [CrossRef]
  26. Domon, B. Mass Spectrometry and Protein Analysis. Science 2006, 312, 212–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Eng, J.K.; McCormack, A.L.; Yates, J.R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass Spectrom. 1994, 5, 976–989. [Google Scholar] [CrossRef] [Green Version]
  28. Perkins, D.N.; Pappin, D.J.C.; Creasy, D.M.; Cottrell, J.S. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 1999, 20, 3551–3567. [Google Scholar] [CrossRef]
  29. Craig, R.; Beavis, R.C. TANDEM: Matching proteins with tandem mass spectra. Bioinformatics 2004, 20, 1466–1467. [Google Scholar] [CrossRef]
  30. Cox, J.; Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 2008, 26, 1367–1372. [Google Scholar] [CrossRef]
  31. Nesvizhskii, A.I.; Aebersold, R. Interpretation of Shotgun Proteomic Data: The Protein Inference Problem. Mol. Cell. Proteom. 2005, 4, 1419–1440. [Google Scholar] [CrossRef] [Green Version]
  32. Perez-Riverol, Y.; Sánchez, A.; Ramos, Y.; Schmidt, A.; Müller, M.; Betancourt, L.; González, L.J.; Vera, R.; Padron, G.; Besada, V. In silico analysis of accurate proteomics, complemented by selective isolation of peptides. J. Proteom. 2011, 74, 2071–2082. [Google Scholar] [CrossRef]
  33. Nesvizhskii, A.I.; Keller, A.; Kolker, E.; Aebersold, R. A Statistical Model for Identifying Proteins by Tandem Mass Spectrometry. Anal. Chem. 2003, 75, 4646–4658. [Google Scholar] [CrossRef] [PubMed]
  34. Searle, B.C. Scaffold: A bioinformatic tool for validating MS/MS-based proteomic studies. PROTEOMICS 2010, 10, 1265–1269. [Google Scholar] [CrossRef] [PubMed]
  35. Ma, C.; Yue, Q.X.; Guan, S.H.; Wu, W.Y.; Yang, M.; Jiang, B.H.; Liu, X.; Guo, D.A. Proteomic analysis of possible target-related proteins of cyclophosphamide in mice thymus. Food Chem. Toxicol. 2009, 47, 1841–1847. [Google Scholar] [CrossRef] [PubMed]
  36. Uszkoreit, J.; Maerkens, A.; Perez-Riverol, Y.; Meyer, H.E.; Marcus, K.; Stephan, C.; Kohlbacher, O.; Eisenacher, M. PIA: An Intuitive Protein Inference Engine with a Web-Based User Interface. J. Proteome Res. 2015, 14, 2988–2997. [Google Scholar] [CrossRef]
  37. Uszkoreit, J.; Perez-Riverol, Y.; Eggers, B.; Marcus, K.; Eisenacher, M. Protein Inference Using PIA Workflows and PSI Standard File Formats. J. Proteome Res. 2019, 18, 741–747. [Google Scholar] [CrossRef]
  38. Pfeuffer, J.; Sachsenberg, T.; Dijkstra, T.M.H.; Serang, O.; Reinert, K.; Kohlbacher, O. EPIFANY: A Method for Efficient High-Confidence Protein Inference. J. Proteome Res. 2020, 19, 1060–1072. [Google Scholar] [CrossRef]
  39. Zhang, B.; Pirmoradian, M.; Zubarev, R.; Käll, L. Covariation of Peptide Abundances Accurately Reflects Protein Concentration Differences. Mol. Cell. Proteom. 2017, 16, 936–948. [Google Scholar] [CrossRef] [Green Version]
  40. Schilde, L.M.; Kösters, S.; Steinbach, S.; Schork, K.; Eisenacher, M.; Galozzi, S.; Turewicz, M.; Barkovits, K.; Mollenhauer, B.; Marcus, K.; et al. Protein variability in cerebrospinal fluid and its possible implications for neurological protein biomarker research. PLoS ONE 2018, 13, e0206478. [Google Scholar] [CrossRef] [Green Version]
  41. Abdi, F.; Quinn, J.F.; Jankovic, J.; McIntosh, M.; Leverenz, J.B.; Peskind, E.; Nixon, R.; Nutt, J.; Chung, K.; Zabetian, C.; et al. Detection of biomarkers with a multiplex quantitative proteomic platform in cerebrospinal fluid of patients with neurodegenerative disorders. J. Alzheimers Dis. 2006, 9, 293–348. [Google Scholar] [CrossRef] [PubMed]
  42. Argüelles, S.; Venero, J.L.; García-Rodriguez, S.; Tomas-Camardiel, M.; Ayala, A.; Cano, J.; Machado, A. Use of haptoglobin and transthyretin as potential biomarkers for the preclinical diagnosis of Parkinson’s disease. Neurochem. Int. 2010, 57, 227–234. [Google Scholar] [CrossRef] [PubMed]
  43. Yazdani, S.; Mariosa, D.; Hammar, N.; Andersson, J.; Ingre, C.; Walldius, G.; Fang, F. Peripheral immune biomarkers and neurodegenerative diseases: A prospective cohort study with 20 years of follow-up. Ann. Neurol. 2019, 86, 913–926. [Google Scholar] [CrossRef] [PubMed]
  44. Yin, G.N.; Lee, H.W.; Cho, J.-Y.; Suk, K. Neuronal pentraxin receptor in cerebrospinal fluid as a potential biomarker for neurodegenerative diseases. Brain Res. 2009, 1265, 158–170. [Google Scholar] [CrossRef] [PubMed]
  45. Comabella, M.; Fernández, M.; Martin, R.; Rivera-Vallvé, S.; Borrás, E.; Chiva, C.; Julià, E.; Rovira, A.; Cantó, E.; Alvarez-Cermeño, J.C.; et al. Cerebrospinal fluid chitinase 3-like 1 levels are associated with conversion to multiple sclerosis. Brain 2010, 133, 1082–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Orenes-Piñero, E.; Hernández-Romero, D.; De Torre, C.; Vilchez, J.A.; Martínez, M.; Romero-Aniorte, A.I.; Climent, V.; García-Honrubia, A.; Valdés, M.; Marín, F. Identification and confirmation of haptoglobin as a potential serum biomarker in hypertrophic cardiomyopathy using proteomic approaches. Ann. Med. 2013, 45, 341–347. [Google Scholar] [CrossRef]
  47. Garibay-Cerdenares, O.; Hernández-Ramírez, V.; Osorio-Trujillo, J.; Hernández-Ortíz, M.; Gallardo-Rincón, D.; De León, D.C.; Encarnación-Guevara, S.; Villegas-Pineda, J.; Talamás-Rohana, P. Proteomic identification of fucosylated haptoglobin alpha isoforms in ascitic fluids and its localization in ovarian carcinoma tissues from Mexican patients. J. Ovarian Res. 2014, 7, 27. [Google Scholar] [CrossRef] [Green Version]
  48. Villegas-Pineda, J.C.; Garibay-Cerdenares, O.L.; Hernández-Ramírez, V.I.; Gallardo-Rincón, D.; De León, D.C.; Pérez-Montiel-Gómez, M.D.; Talamás-Rohana, P. Integrins and haptoglobin: Molecules overexpressed in ovarian cancer. Pathol. Res. Pract. 2015, 211, 973–981. [Google Scholar] [CrossRef]
  49. Halbgebauer, S.; Öckl, P.; Wirth, K.; Steinacker, P.; Otto, M. Protein biomarkers in Parkinson’s disease: Focus on cerebrospinal fluid markers and synaptic proteins: Protein Biomarkers in Parkinson’s Disease. Mov. Disord. 2016, 31, 848–860. [Google Scholar] [CrossRef]
  50. Fish, R.G.; Gill, T.S.; Adams, M.; Kerby, I. Serum haptoglobin and α1-acid glycoprotein as indicators of the effectiveness of cis-diamminedichloroplatinum (CDDP) in ovarian cancer patients—A preliminary report. Eur. J. Cancer Clin. Oncol. 1984, 20, 625–630. [Google Scholar] [CrossRef]
  51. Ahmed, N.; Barker, G.; Oliva, K.T.; Hoffmann, P.; Riley, C.; Reeve, S.; Smith, A.I.; Kemp, B.E.; Quinn, M.A.; Rice, G.E. Proteomic-based identification of haptoglobin-1 precursor as a novel circulating biomarker of ovarian cancer. Br. J. Cancer 2004, 91, 129–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. May, C.; Brosseron, F.; Chartowski, P.; Meyer, H.E.; Marcus, K. Differential proteome analysis using 2D-DIGE. Methods Mol. Biol. Clifton N.J. 2012, 893, 75–82. [Google Scholar] [CrossRef]
  53. Riederer, B.M. Non-covalent and covalent protein labeling in two-dimensional gel electrophoresis. J. Proteom. 2008, 71, 231–244. [Google Scholar] [CrossRef]
  54. Butt, R.H.; Coorssen, J.R. Coomassie blue as a near-infrared fluorescent stain: A systematic comparison with Sypro Ruby for in-gel protein detection. Mol. Cell. Proteom. MCP 2013, 12, 3834–3850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Dyballa, N.; Metzger, S. Fast and sensitive coomassie staining in quantitative proteomics. Methods Mol. Biol. Clifton N. J. 2012, 893, 47–59. [Google Scholar] [CrossRef]
  56. Rabilloud, T. Silver staining of 2D electrophoresis gels. Methods Mol. Biol. Clifton N. J. 2012, 893, 61–73. [Google Scholar] [CrossRef] [Green Version]
  57. Herrmann, A.G.; Searcy, J.L.; Le Bihan, T.; McCulloch, J.; Deighton, R.F. Total variance should drive data handling strategies in third generation proteomic studies. Proteomics 2013, 13, 3251–3255. [Google Scholar] [CrossRef] [Green Version]
  58. Sandomenico, A.; Severino, V.; Chambery, A.; Focà, A.; Focà, G.; Farina, C.; Ruvo, M. A comparative structural and bioanalytical study of IVIG clinical lots. Mol. Biotechnol. 2013, 54, 983–995. [Google Scholar] [CrossRef]
  59. Nebija, D.; Noe, C.; Urban, E.; Lachmann, B. Quality Control and Stability Studies with the Monoclonal Antibody, Trastuzumab: Application of 1D- vs. 2D-Gel Electrophoresis. Int. J. Mol. Sci. 2014, 15, 6399–6411. [Google Scholar] [CrossRef] [Green Version]
  60. Jawa, V.; Joubert, M.K.; Zhang, Q.; Deshpande, M.; Hapuarachchi, S.; Hall, M.P.; Flynn, G.C. Evaluating Immunogenicity Risk Due to Host Cell Protein Impurities in Antibody-Based Biotherapeutics. AAPS J. 2016, 18, 1439–1452. [Google Scholar] [CrossRef]
  61. Al Shweiki, M.R.; Mönchgesang, S.; Majovsky, P.; Thieme, D.; Trutschel, D.; Hoehenwarter, W. Assessment of Label-Free Quantification in Discovery Proteomics and Impact of Technological Factors and Natural Variability of Protein Abundance. J. Proteome Res. 2017, 16, 1410–1424. [Google Scholar] [CrossRef]
  62. Trinkle-Mulcahy, L.; Boulon, S.; Lam, Y.W.; Urcia, R.; Boisvert, F.M.; Vandermoere, F.; Morrice, N.A.; Swift, S.; Rothbauer, U.; Leonhardt, H.; et al. Identifying specific protein interaction partners using quantitative mass spectrometry and bead proteomes. J. Cell Biol. 2008, 183, 223–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Mellacheruvu, D.; Wright, Z.; Couzens, A.L.; Lambert, J.P.; St-Denis, N.A.; Li, T.; Miteva, Y.V.; Hauri, S.; Sardiu, M.E.; Low, T.Y.; et al. The CRAPome: A contaminant repository for affinity purification-mass spectrometry data. Nat. Methods 2013, 10, 730–736. [Google Scholar] [CrossRef] [Green Version]
  64. Chevallet, M.; Procaccio, V.; Rabilloud, T. A nonradioactive double detection method for the assignment of spots in two-dimensional blots. Anal. Biochem. 1997, 251, 69–72. [Google Scholar] [CrossRef]
  65. Kusch, K.; Uecker, M.; Liepold, T.; Möbius, W.; Hoffmann, C.; Neumann, H.; Werner, H.B.; Jahn, O. Partial Immunoblotting of 2D-Gels: A Novel Method to Identify Post-Translationally Modified Proteins Exemplified for the Myelin Acetylome. Proteomes 2017, 5, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Barrera, C.; Millon, L.; Rognon, B.; Quadroni, M.; Roussel, S.; Dalphin, J.C.; Court-Fortune, I.; Caillaud, D.; Jouneau, S.; Fellrath, J.M.; et al. Immunoreactive proteins of Saccharopolyspora rectivirgula for farmer’s lung serodiagnosis. Proteom. Clin. Appl. 2014, 8, 971–981. [Google Scholar] [CrossRef]
  67. Connolly, J.P.; Comerci, D.; Alefantis, T.G.; Walz, A.; Quan, M.; Chafin, R.; Grewal, P.; Mujer, C.V.; Ugalde, R.A.; DelVecchio, V.G. Proteomic analysis of Brucella abortus cell envelope and identification of immunogenic candidate proteins for vaccine development. Proteomics 2006, 6, 3767–3780. [Google Scholar] [CrossRef]
  68. Delvecchio, V.G.; Connolly, J.P.; Alefantis, T.G.; Walz, A.; Quan, M.A.; Patra, G.; Ashton, J.M.; Whittington, J.T.; Chafin, R.D.; Liang, X.; et al. Proteomic profiling and identification of immunodominant spore antigens of Bacillus anthracis, Bacillus cereus, and Bacillus thuringiensis. Appl. Environ. Microbiol. 2006, 72, 6355–6363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Fajardo Bonin, R.; Chapeaurouge, A.; Perales, J.; Da Silva, J.G.; Do Nascimento, H.J.; D’Alincourt Carvalho Assef, A.P.; Moreno Senna, J.P. Identification of immunogenic proteins of the bacterium Acinetobacter baumannii using a proteomic approach. Proteom. Clin. Appl. 2014, 8, 916–923. [Google Scholar] [CrossRef] [Green Version]
  70. Gaur, R.; Alam, S.I.; Kamboj, D.V. Immunoproteomic Analysis of Antibody Response of Rabbit Host Against Heat-Killed Francisella tularensis Live Vaccine Strain. Curr. Microbiol. 2017, 74, 499–507. [Google Scholar] [CrossRef] [PubMed]
  71. Pitarch, A.; Jimenez, A.; Nombela, C.; Gil, C. Decoding serological response to Candida cell wall immunome into novel diagnostic, prognostic, and therapeutic candidates for systemic candidiasis by proteomic and bioinformatic analyses. Mol. Cell. Proteom. 2006, 5, 79–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Pellon, A.; Ramirez-Garcia, A.; Buldain, I.; Antoran, A.; Rementeria, A.; Hernando, F.L. Immunoproteomics-Based Analysis of the Immunocompetent Serological Response to Lomentospora prolificans. J. Proteome Res. 2016, 15, 595–607. [Google Scholar] [CrossRef] [PubMed]
  73. Buldain, I.; Ramirez-Garcia, A.; Pellon, A.; Antoran, A.; Sevilla, M.J.; Rementeria, A.; Hernando, F.L. Cyclophilin and enolase are the most prevalent conidial antigens of Lomentospora prolificans recognized by healthy human salivary IgA and cross-react with Aspergillus fumigatus. Proteom. Clin. Appl. 2016, 10, 1058–1067. [Google Scholar] [CrossRef]
  74. Garcia-Lunar, P.; Regidor-Cerrillo, J.; Gutierrez-Exposito, D.; Ortega-Mora, L.; Alvarez-Garcia, G. First 2-DE approach towards characterising the proteome and immunome of Besnoitia besnoiti in the tachyzoite stage. Vet. Parasitol. 2013, 195, 24–34. [Google Scholar] [CrossRef] [PubMed]
  75. Reamtong, O.; Rujimongkon, K.; Sookrung, N.; Saeung, A.; Thiangtrongjit, T.; Sakolvaree, Y.; Thammapalo, S.; Loymek, S.; Chaicumpa, W. Immunome and immune complex-forming components of Brugia malayi identified by microfilaremic human sera. Exp. Parasitol. 2019, 200, 92–98. [Google Scholar] [CrossRef]
  76. Pitarch, A.; Nombela, C.; Gil, C. Prediction of the clinical outcome in invasive candidiasis patients based on molecular fingerprints of five anti-Candida antibodies in serum. Mol. Cell. Proteom. 2011, 10, M110.004010. [Google Scholar] [CrossRef] [Green Version]
  77. Pitarch, A.; Nombela, C.; Gil, C. Seroprofiling at the Candida albicans protein species level unveils an accurate molecular discriminator for candidemia. J. Proteom. 2016, 134, 144–162. [Google Scholar] [CrossRef]
  78. Singh, B.; Sharma, G.L.; Oellerich, M.; Kumar, R.; Singh, S.; Bhadoria, D.P.; Katyal, A.; Reichard, U.; Asif, A.R. Novel cytosolic allergens of Aspergillus fumigatus identified from germinating conidia. J. Proteome Res. 2010, 9, 5530–5541. [Google Scholar] [CrossRef]
  79. Ghosh, N.; Sircar, G.; Saha, B.; Pandey, N.; Bhattacharya, S.G. Search for Allergens from the Pollen Proteome of Sunflower (Helianthus annuus L.): A Major Sensitizer for Respiratory Allergy Patients. PLoS ONE 2015, 10, e0138992. [Google Scholar] [CrossRef] [Green Version]
  80. Saha, B.; Bhattacharya, S.G. Charting novel allergens from date palm pollen (Phoenix sylvestris) using homology driven proteomics. J. Proteom. 2017, 165, 1–10. [Google Scholar] [CrossRef]
  81. Park, K.H.; Lee, J.; Lee, J.Y.; Lee, S.C.; Sim, D.W.; Shin, J.U.; Park, C.O.; Lee, J.H.; Lee, K.H.; Jeong, K.Y.; et al. Sensitization to various minor house dust mite allergens is greater in patients with atopic dermatitis than in those with respiratory allergic disease. Clin. Exp. Allergy 2018, 48, 1050–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. De Angelis, M.; Di Cagno, R.; Minervini, F.; Rizzello, C.G.; Gobbetti, M. Two-dimensional electrophoresis and IgE-mediated food allergy. Electrophoresis 2010, 31, 2126–2136. [Google Scholar] [CrossRef] [PubMed]
  83. Matsuo, K.; Xiang, Y.; Nakamura, H.; Masuko, K.; Yudoh, K.; Noyori, K.; Nishioka, K.; Saito, T.; Kato, T. Identification of novel citrullinated autoantigens of synovium in rheumatoid arthritis using a proteomic approach. Arthritis Res. Ther. 2006, 8, R175. [Google Scholar] [CrossRef] [Green Version]
  84. Kinloch, A.; Lundberg, K.; Wait, R.; Wegner, N.; Lim, N.H.; Zendman, A.J.W.; Saxne, T.; Malmström, V.; Venables, P.J. Synovial fluid is a site of citrullination of autoantigens in inflammatory arthritis. Arthritis Rheum. 2008, 58, 2287–2295. [Google Scholar] [CrossRef]
  85. Goëb, V.; Thomas-L’Otellier, M.; Daveau, R.; Charlionet, R.; Fardellone, P.; Le Loët, X.; Tron, F.; Gilbert, D.; Vittecoq, O. Candidate autoantigens identified by mass spectrometry in early rheumatoid arthritis are chaperones and citrullinated glycolytic enzymes. Arthritis Res. Ther. 2009, 11, R38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Bussone, G.; Dib, H.; Tamby, M.C.; Broussard, C.; Federici, C.; Woimant, G.; Camoin, L.; Guillevin, L.; Mouthon, L. Identification of new autoantibody specificities directed at proteins involved in the transforming growth factor β pathway in patients with systemic sclerosis. Arthritis Res. Ther. 2011, 13, R74. [Google Scholar] [CrossRef] [Green Version]
  87. Biswas, S.; Sharma, S.; Saroha, A.; Bhakuni, D.S.; Malhotra, R.; Zahur, M.; Oellerich, M.; Das, H.R.; Asif, A.R. Identification of novel autoantigen in the synovial fluid of rheumatoid arthritis patients using an immunoproteomics approach. PLoS ONE 2013, 8, e56246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Mathey, E.K.; Derfuss, T.; Storch, M.K.; Williams, K.R.; Hales, K.; Woolley, D.R.; Al-Hayani, A.; Davies, S.N.; Rasband, M.N.; Olsson, T.; et al. Neurofascin as a novel target for autoantibody-mediated axonal injury. J. Exp. Med. 2007, 204, 2363–2372. [Google Scholar] [CrossRef] [Green Version]
  89. Derfuss, T.; Parikh, K.; Velhin, S.; Braun, M.; Mathey, E.; Krumbholz, M.; Kümpfel, T.; Moldenhauer, A.; Rader, C.; Sonderegger, P.; et al. Contactin-2/TAG-1-directed autoimmunity is identified in multiple sclerosis patients and mediates gray matter pathology in animals. Proc. Natl. Acad. Sci. USA 2009, 106, 8302–8307. [Google Scholar] [CrossRef] [Green Version]
  90. Privitera, D.; Corti, V.; Alessio, M.; Volontè, M.A.; Volontè, A.; Lampasona, V.; Comi, G.; Martino, G.; Franciotta, D.; Furlan, R.; et al. Proteomic identification of aldolase A as an autoantibody target in patients with atypical movement disorders. Neurol. Sci. Off. J. Ital. Neurol. Soc. Ital. Soc. Clin. Neurophysiol. 2013, 34, 313–320. [Google Scholar] [CrossRef]
  91. Kuwabara, Y.; Katayama, A.; Kurihara, S.; Orimo, H.; Takeshita, T. Immunoproteomic identification of anti-C9 autoimmune antibody in patients with seronegative obstetric antiphospholipid syndrome. PLoS ONE 2018, 13, e0198472. [Google Scholar] [CrossRef] [PubMed]
  92. Beadle, G.W.; Tatum, E.L. Genetic Control of Biochemical Reactions in Neurospora. Proc. Natl. Acad. Sci. USA 1941, 27, 499–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. McGettigan, P.A. Transcriptomics in the RNA-seq era. Curr. Opin. Chem. Biol. 2013, 17, 4–11. [Google Scholar] [CrossRef]
  94. Bischoff, R.; Schlüter, H. Amino acids: Chemistry, functionality and selected non-enzymatic post-translational modifications. J. Proteom. 2012, 75, 2275–2296. [Google Scholar] [CrossRef] [Green Version]
  95. Wagner, G.R.; Hirschey, M.D. Nonenzymatic protein acylation as a carbon stress regulated by sirtuin deacylases. Mol. Cell 2014, 54, 5–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Trub, A.G.; Hirschey, M.D. Reactive Acyl-CoA Species Modify Proteins and Induce Carbon Stress. Trends Biochem. Sci. 2018, 43, 369–379. [Google Scholar] [CrossRef]
  97. Thaysen-Andersen, M.; Packer, N.H. Advances in LC-MS/MS-based glycoproteomics: Getting closer to system-wide site-specific mapping of the N- and O-glycoproteome. Biochim. Biophys. Acta 2014, 1844, 1437–1452. [Google Scholar] [CrossRef]
  98. Zhang, X.; Huang, Y.; Shi, X. Emerging roles of lysine methylation on non-histone proteins. Cell. Mol. Life Sci. CMLS 2015, 72, 4257–4272. [Google Scholar] [CrossRef]
  99. Wesche, J.; Kühn, S.; Kessler, B.M.; Salton, M.; Wolf, A. Protein arginine methylation: A prominent modification and its demethylation. Cell. Mol. Life Sci. CMLS 2017, 74, 3305–3315. [Google Scholar] [CrossRef]
  100. Xiong, Y.; Guan, K.-L. Mechanistic insights into the regulation of metabolic enzymes by acetylation. J. Cell Biol. 2012, 198, 155–164. [Google Scholar] [CrossRef] [Green Version]
  101. Verdin, E.; Ott, M. 50 years of protein acetylation: From gene regulation to epigenetics, metabolism and beyond. Nat. Rev. Mol. Cell Biol. 2015, 16, 258–264. [Google Scholar] [CrossRef] [PubMed]
  102. Drazic, A.; Myklebust, L.M.; Ree, R.; Arnesen, T. The world of protein acetylation. Biochim. Biophys. Acta 2016, 1864, 1372–1401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Narita, T.; Weinert, B.T.; Choudhary, C. Functions and mechanisms of non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. 2019, 20, 156–174. [Google Scholar] [CrossRef] [PubMed]
  104. Hirschey, M.D.; Zhao, Y. Metabolic Regulation by Lysine Malonylation, Succinylation, and Glutarylation. Mol. Cell. Proteom. MCP 2015, 14, 2308–2315. [Google Scholar] [CrossRef] [Green Version]
  105. Hentschel, A.; Zahedi, R.P.; Ahrends, R. Protein lipid modifications—More than just a greasy ballast. Proteomics 2016, 16, 759–782. [Google Scholar] [CrossRef]
  106. Carrico, C.; Meyer, J.G.; He, W.; Gibson, B.W.; Verdin, E. The Mitochondrial Acylome Emerges: Proteomics, Regulation by Sirtuins, and Metabolic and Disease Implications. Cell Metab. 2018, 27, 497–512. [Google Scholar] [CrossRef] [Green Version]
  107. Seo, J.; Jeong, J.; Kim, Y.M.; Hwang, N.; Paek, E.; Lee, K.-J. Strategy for Comprehensive Identification of Post-translational Modifications in Cellular Proteins, Including Low Abundant Modifications: Application to Glyceraldehyde-3-phosphate Dehydrogenase. J. Proteome Res. 2008, 7, 587–602. [Google Scholar] [CrossRef]
  108. Niimori-Kita, K.; Tamamaki, N.; Koizumi, D.; Niimori, D. Matrin-3 is essential for fibroblast growth factor 2-dependent maintenance of neural stem cells. Sci. Rep. 2018, 8, 13412. [Google Scholar] [CrossRef]
  109. Dalzon, B.; Torres, A.; Diemer, H.; Ravanel, S.; Collin-Faure, V.; Pernet-Gallay, K.; Jouneau, P.-H.; Bourguignon, J.; Cianfrani, S.; Carrire, M.; et al. How reversible are the effects of silver nanoparticles on macrophages? A proteomic-instructed view. Environ. Sci. Nano 2019, 6, 3133–3157. [Google Scholar] [CrossRef] [Green Version]
  110. Sun, H.H.; Fukao, Y.; Ishida, S.; Yamamoto, H.; Maekawa, S.; Fujiwara, M.; Sato, T.; Yamaguchi, J. Proteomics Analysis Reveals a Highly Heterogeneous Proteasome Composition and the Post-translational Regulation of Peptidase Activity under Pathogen Signaling in Plants. J. Proteome Res. 2013, 12, 5084–5095. [Google Scholar] [CrossRef]
  111. Martins-de-Souza, D.; Maccarrone, G.; Wobrock, T.; Zerr, I.; Gormanns, P.; Reckow, S.; Falkai, P.; Schmitt, A.; Turck, C.W. Proteome analysis of the thalamus and cerebrospinal fluid reveals glycolysis dysfunction and potential biomarkers candidates for schizophrenia. J. Psychiatr. Res. 2010, 44, 1176–1189. [Google Scholar] [CrossRef] [PubMed]
  112. Chevallet, M.; Luche, S.; Diemer, H.; Strub, J.M.; Van Dorsselaer, A.; Rabilloud, T. Sweet silver: A formaldehyde-free silver staining using aldoses as developing agents, with enhanced compatibility with mass spectrometry. Proteomics 2008, 8, 4853–4861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ryšlavá, H.; Doubnerová, V.; Kavan, D.; Vaněk, O. Effect of posttranslational modifications on enzyme function and assembly. J. Proteom. 2013, 92, 80–109. [Google Scholar] [CrossRef] [PubMed]
  114. Petrak, J.; Ivanek, R.; Toman, O.; Cmejla, R.; Cmejlova, J.; Vyoral, D.; Zivny, J.; Vulpe, C.D. Deja vu in proteomics. A hit parade of repeatedly identified differentially expressed proteins. Proteomics 2008, 8, 1744–1749. [Google Scholar] [CrossRef]
  115. Wang, P.; Bouwman, F.G.; Mariman, E.C. Generally detected proteins in comparative proteomics—A matter of cellular stress response? Proteomics 2009, 9, 2955–2966. [Google Scholar] [CrossRef]
  116. Lenglet, G.; Depauw, S.; Mendy, D.; David-Cordonnier, M.-H. Protein recognition of the S23906-1–DNA adduct by nuclear proteins: Direct involvement of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Biochem. J. 2013, 452, 147–159. [Google Scholar] [CrossRef]
  117. Angeloni, C.; Turroni, S.; Bianchi, L.; Fabbri, D.; Motori, E.; Malaguti, M.; Leoncini, E.; Maraldi, T.; Bini, L.; Brigidi, P.; et al. Novel targets of sulforaphane in primary cardiomyocytes identified by proteomic analysis. PLoS ONE 2013, 8, e83283. [Google Scholar] [CrossRef] [Green Version]
  118. Armand, L.; Biola-Clier, M.; Bobyk, L.; Collin-Faure, V.; Diemer, H.; Strub, J.-M.; Cianferani, S.; Van Dorsselaer, A.; Herlin-Boime, N.; Rabilloud, T.; et al. Molecular responses of alveolar epithelial A549 cells to chronic exposure to titanium dioxide nanoparticles: A proteomic view. J. Proteom. 2016, 134, 163–173. [Google Scholar] [CrossRef]
  119. Luche, S.; Eymard-Vernain, E.; Diemer, H.; Van Dorsselaer, A.; Rabilloud, T.; Lelong, C. Zinc oxide induces the stringent response and major reorientations in the central metabolism of Bacillus subtilis. J. Proteom. 2016, 135, 170–180. [Google Scholar] [CrossRef] [Green Version]
  120. Chiasserini, D.; Davidescu, M.; Orvietani, P.L.; Susta, F.; Macchioni, L.; Petricciuolo, M.; Castigli, E.; Roberti, R.; Binaglia, L.; Corazzi, L. 3-Bromopyruvate treatment induces alterations of metabolic and stress-related pathways in glioblastoma cells. J. Proteom. 2017, 152, 329–338. [Google Scholar] [CrossRef]
  121. Dalzon, B.; Aude-Garcia, C.; Collin-Faure, V.; Diemer, H.; Béal, D.; Dussert, F.; Fenel, D.; Schoehn, G.; Cianférani, S.; Carrière, M.; et al. Differential proteomics highlights macrophage-specific responses to amorphous silica nanoparticles. Nanoscale 2017, 9, 9641–9658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Heeb, M.J.; Gabriel, O. Enzyme localization in gels. Methods Enzymol. 1984, 104, 416–439. [Google Scholar] [CrossRef]
  123. Gabriel, O.; Gersten, D.M. Staining for enzymatic activity after gel electrophoresis, I. Anal. Biochem. 1992, 203, 1–21. [Google Scholar] [CrossRef]
  124. Bischoff, K.M.; Shi, L.; Kennelly, P.J. The detection of enzyme activity following sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Anal. Biochem. 1998, 260, 1–17. [Google Scholar] [CrossRef] [PubMed]
  125. Manrow, R.E.; Dottin, R.P. Demonstration, by renaturation in O’Farrell gels, of heterogeneity in Dictyostelium uridine diphosphoglucose pyrophosphorylase. Anal. Biochem. 1982, 120, 181–188. [Google Scholar] [CrossRef]
  126. Durocher, Y.; Chapdelaine, A.; Chevalier, S. Identification of cytosolic protein tyrosine kinases of human prostate by renaturation after SDS/PAGE. Biochem. J. 1992, 284, 653–658. [Google Scholar] [CrossRef] [Green Version]
  127. Brochu, G.; Shah, G.M.; Poirier, G.G. Purification of poly (ADP-ribose) glycohydrolase and detection of its isoforms by a zymogram following one- or two-dimensional electrophoresis. Anal. Biochem. 1994, 218, 265–272. [Google Scholar] [CrossRef]
  128. Chen, S.; Meng, F.; Chen, Z.; Tomlinson, B.N.; Wesley, J.M.; Sun, G.Y.; Whaley-Connell, A.T.; Sowers, J.R.; Cui, J.; Gu, Z. Two-Dimensional Zymography Differentiates Gelatinase Isoforms in Stimulated Microglial Cells and in Brain Tissues of Acute Brain Injuries. PLoS ONE 2015, 10, e0123852. [Google Scholar] [CrossRef]
  129. Stroud, L.J.; Šlapeta, J.; Padula, M.P.; Druery, D.; Tsiotsioras, G.; Coorssen, J.R.; Stack, C.M. Comparative proteomic analysis of two pathogenic Tritrichomonas foetus genotypes: There is more to the proteome than meets the eye. Int. J. Parasitol. 2017, 47, 203–213. [Google Scholar] [CrossRef]
  130. Triboulet, S.; Aude-Garcia, C.; Armand, L.; Collin-Faure, V.; Chevallet, M.; Diemer, H.; Gerdil, A.; Proamer, F.; Strub, J.M.; Habert, A.; et al. Comparative proteomic analysis of the molecular responses of mouse macrophages to titanium dioxide and copper oxide nanoparticles unravels some toxic mechanisms for copper oxide nanoparticles in macrophages. PLoS ONE 2015, 10, e124496. [Google Scholar] [CrossRef] [Green Version]
  131. Aude-Garcia, C.; Dalzon, B.; Ravanat, J.L.; Collin-Faure, V.; Diemer, H.; Strub, J.M.; Cianferani, S.; Van Dorsselaer, A.; Carriere, M.; Rabilloud, T. A combined proteomic and targeted analysis unravels new toxic mechanisms for zinc oxide nanoparticles in macrophages. J. Proteom. 2016, 134, 174–185. [Google Scholar] [CrossRef] [PubMed]
  132. D’Anna, C.; Cigna, D.; Di Sano, C.; Di Vincenzo, S.; Dino, P.; Ferraro, M.; Bini, L.; Bianchi, L.; Di Gaudio, F.; Gjomarkaj, M.; et al. Exposure to cigarette smoke extract and lipopolysaccharide modifies cytoskeleton organization in bronchial epithelial cells. Exp. Lung Res. 2017, 43, 347–358. [Google Scholar] [CrossRef]
  133. Matsuda, Y.; Ishiwata, T.; Yoshimura, H.; Hagio, M.; Arai, T. Inhibition of nestin suppresses stem cell phenotype of glioblastomas through the alteration of post-translational modification of heat shock protein HSPA8/HSC. Cancer Lett. 2015, 357, 602–611. [Google Scholar] [CrossRef]
  134. Ostrowski, S.M.; Johnson, K.; Siefert, M.; Shank, S.; Sironi, L.; Wolozin, B.; Landreth, G.E.; Ziady, A.G. Simvastatin inhibits protein isoprenylation in the brain. Neuroscience 2016, 329, 264–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Gamberi, T.; Massai, L.; Magherini, F.; Landini, I.; Fiaschi, T.; Scaletti, F.; Gabbiani, C.; Bianchi, L.; Bini, L.; Nobili, S.; et al. Proteomic analysis of A2780/S ovarian cancer cell response to the cytotoxic organogold (III) compound Aubipy (c). J. Proteom. 2014, 103, 103–120. [Google Scholar] [CrossRef]
  136. Beltran, L.; Cutillas, P.R. Advances in phosphopeptide enrichment techniques for phosphoproteomics. Amino Acids 2012, 43, 1009–1024. [Google Scholar] [CrossRef]
  137. Carlson, S.M.; Gozani, O. Emerging technologies to map the protein methylome. J. Mol. Biol. 2014, 426, 3350–3362. [Google Scholar] [CrossRef] [Green Version]
  138. Ke, M.; Shen, H.; Wang, L.; Luo, S.; Lin, L.; Yang, J.; Tian, R. Identification, Quantification, and Site Localization of Protein Posttranslational Modifications via Mass Spectrometry-Based Proteomics. Adv. Exp. Med. Biol. 2016, 919, 345–382. [Google Scholar] [CrossRef] [PubMed]
  139. Diallo, I.; Seve, M.; Cunin, V.; Minassian, F.; Poisson, J.-F.; Michelland, S.; Bourgoin-Voillard, S. Current trends in protein acetylation analysis. Expert Rev. Proteomics 2019, 16, 139–159. [Google Scholar] [CrossRef] [PubMed]
  140. Allfrey, V.G.; Faulkner, R.; Mirsky, A.E. Acetylation and Methylation of Histones and Their Possible Role in the Regulation of Rna Synthesis. Proc. Natl. Acad. Sci. USA 1964, 51, 786–794. [Google Scholar] [CrossRef] [Green Version]
  141. Chen, Y.; Sprung, R.; Tang, Y.; Ball, H.; Sangras, B.; Kim, S.C.; Falck, J.R.; Peng, J.; Gu, W.; Zhao, Y. Lysine propionylation and butyrylation are novel post-translational modifications in histones. Mol. Cell. Proteom. MCP 2007, 6, 812–819. [Google Scholar] [CrossRef] [Green Version]
  142. Xie, Z.; Dai, J.; Dai, L.; Tan, M.; Cheng, Z.; Wu, Y.; Boeke, J.D.; Zhao, Y. Lysine succinylation and lysine malonylation in histones. Mol. Cell. Proteom. MCP 2012, 11, 100–107. [Google Scholar] [CrossRef] [Green Version]
  143. Tan, M.; Luo, H.; Lee, S.; Jin, F.; Yang, J.S.; Montellier, E.; Buchou, T.; Cheng, Z.; Rousseaux, S.; Rajagopal, N.; et al. Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell 2011, 146, 1016–1028. [Google Scholar] [CrossRef] [Green Version]
  144. Sarioglu, H.; Lottspeich, F.; Walk, T.; Jung, G.; Eckerskorn, C. Deamidation as a widespread phenomenon in two-dimensional polyacrylamide gel electrophoresis of human blood plasma proteins. Electrophoresis 2000, 21, 2209–2218. [Google Scholar] [CrossRef]
  145. Zhu, Y.; Li, G.; Dong, Y.; Zhou, H.H.; Kong, B.; Aleksunes, L.M.; Richardson, J.R.; Li, F.; Guo, G.L. Modulation of farnesoid X receptor results in post-translational modification of poly (ADP-ribose) polymerase 1 in the liver. Toxicol. Appl. Pharmacol. 2013, 266, 260–266. [Google Scholar] [CrossRef] [Green Version]
  146. Blundon, M.A.; Schlesinger, D.R.; Parthasarathy, A.; Smith, S.L.; Kolev, H.M.; Vinson, D.A.; Kunttas-Tatli, E.; McCartney, B.M.; Minden, J.S. Proteomic analysis reveals APC-dependent post-translational modifications and identifies a novel regulator of β-catenin. Development 2016, 143, 2629–2640. [Google Scholar] [CrossRef] [Green Version]
  147. Prudent, R.; Demoncheaux, N.; Diemer, H.; Collin-Faure, V.; Kapur, R.; Paublant, F.; Lafanechere, L.; Cianferani, S.; Rabilloud, T. A quantitative proteomic analysis of cofilin phosphorylation in myeloid cells and its modulation using the LIM kinase inhibitor Pyr. PLoS ONE 2018, 13, e0208979. [Google Scholar] [CrossRef] [Green Version]
  148. Iwai, K.; Shibukawa, Y.; Yamazaki, N.; Wada, Y. Transglutaminase 2-dependent Deamidation of Glyceraldehyde-3-phosphate Dehydrogenase Promotes Trophoblastic Cell Fusion. J. Biol. Chem. 2014, 289, 4989–4999. [Google Scholar] [CrossRef] [Green Version]
  149. Rabilloud, T.; Heller, M.; Gasnier, F.; Luche, S.; Rey, C.; Aebersold, R.; Benahmed, M.; Louisot, P.; Lunardi, J. Proteomics analysis of cellular response to oxidative stress. Evidence for in vivo overoxidation of peroxiredoxins at their active site. J. Biol. Chem. 2002, 277, 19396–19401. [Google Scholar] [CrossRef] [Green Version]
  150. Riquier, S.; Breton, J.; Abbas, K.; Cornu, D.; Bouton, C.; Drapier, J.-C. Peroxiredoxin post-translational modifications by redox messengers. Redox Biol. 2014, 2, 777–785. [Google Scholar] [CrossRef]
  151. Weber, H.; Engelmann, S.; Becher, D.; Hecker, M. Oxidative stress triggers thiol oxidation in the glyceraldehyde-3-phosphate dehydrogenase of Staphylococcus aureus. Mol. Microbiol. 2004, 52, 133–140. [Google Scholar] [CrossRef]
  152. Choi, J.; Rees, H.D.; Weintraub, S.T.; Levey, A.I.; Chin, L.-S.; Li, L. Oxidative modifications and aggregation of Cu, Zn-superoxide dismutase associated with Alzheimer and Parkinson diseases. J. Biol. Chem. 2005, 280, 11648–11655. [Google Scholar] [CrossRef] [Green Version]
  153. Hwang, N.R.; Yim, S.-H.; Kim, Y.M.; Jeong, J.; Song, E.J.; Lee, Y.; Lee, J.H.; Choi, S.; Lee, K.-J. Oxidative modifications of glyceraldehyde-3-phosphate dehydrogenase play a key role in its multiple cellular functions. Biochem. J. 2009, 423, 253–264. [Google Scholar] [CrossRef] [Green Version]
  154. Poschmann, G.; Seyfarth, K.; Besong Agbo, D.; Klafki, H.W.; Rozman, J.; Wurst, W.; Wiltfang, J.; Meyer, H.E.; Klingenspor, M.; Stuhler, K. High-fat diet induced isoform changes of the Parkinson’s disease protein DJ-1. J. Proteome Res. 2014, 13, 2339–2351. [Google Scholar] [CrossRef]
  155. Choi, J.E.; Lee, J.J.; Kang, W.; Kim, H.J.; Cho, J.H.; Han, P.L.; Lee, K.J. Proteomic Analysis of Hippocampus in a Mouse Model of Depression Reveals Neuroprotective Function of Ubiquitin C-terminal Hydrolase L1 (UCH-L1) via Stress-induced Cysteine Oxidative Modifications. Mol. Cell Proteom. 2018, 17, 1803–1823. [Google Scholar] [CrossRef] [Green Version]
  156. John, J.P.P.; Pollak, A.; Lubec, G. Complete sequencing and oxidative modification of manganese superoxide dismutase in medulloblastoma cells. Electrophoresis 2009, 30, 3006–3016. [Google Scholar] [CrossRef]
  157. Codreanu, S.G.; Liebler, D.C. Novel approaches to identify protein adducts produced by lipid peroxidation. Free Radic. Res. 2015, 49, 881–887. [Google Scholar] [CrossRef] [Green Version]
  158. Toyama, T.; Shinkai, Y.; Yazawa, A.; Kakehashi, H.; Kaji, T.; Kumagai, Y. Glutathione-mediated reversibility of covalent modification of ubiquitin carboxyl-terminal hydrolase L1 by 1,2-naphthoquinone through Cys152, but not Lys. Chem. Biol. Interact. 2014, 214, 41–48. [Google Scholar] [CrossRef]
  159. Asif, A.R.; Armstrong, V.W.; Voland, A.; Wieland, E.; Oellerich, M.; Shipkova, M. Proteins identified as targets of the acyl glucuronide metabolite of mycophenolic acid in kidney tissue from mycophenolate mofetil treated rats. Biochimie 2007, 89, 393–402. [Google Scholar] [CrossRef]
  160. Luo, J.; Hill, B.G.; Gu, Y.; Cai, J.; Srivastava, S.; Bhatnagar, A.; Prabhu, S.D. Mechanisms of acrolein-induced myocardial dysfunction: Implications for environmental and endogenous aldehyde exposure. Am. J. Physiol. Heart Circ. Physiol. 2007, 293, H3673–H3684. [Google Scholar] [CrossRef] [Green Version]
  161. Isbell, M.A.; Morin, D.; Boland, B.; Buckpitt, A.; Salemi, M.; Presley, J. Identification of proteins adducted by reactive naphthalene metabolites in vitro. Proteomics 2005, 5, 4197–4204. [Google Scholar] [CrossRef]
  162. Koen, Y.M.; Gogichaeva, N.V.; Alterman, M.A.; Hanzlik, R.P. A proteomic analysis of bromobenzene reactive metabolite targets in rat liver cytosol in vivo. Chem. Res. Toxicol. 2007, 20, 511–519. [Google Scholar] [CrossRef]
  163. Koen, Y.M.; Hajovsky, H.; Liu, K.; Williams, T.D.; Galeva, N.A.; Staudinger, J.L.; Hanzlik, R.P. Liver protein targets of hepatotoxic 4-bromophenol metabolites. Chem. Res. Toxicol. 2012, 25, 1777–1786. [Google Scholar] [CrossRef] [Green Version]
  164. Koen, Y.M.; Sarma, D.; Hajovsky, H.; Galeva, N.A.; Williams, T.D.; Staudinger, J.L.; Hanzlik, R.P. Protein targets of thioacetamide metabolites in rat hepatocytes. Chem. Res. Toxicol. 2013, 26, 564–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Ikehata, K.; Duzhak, T.G.; Galeva, N.A.; Ji, T.; Koen, Y.M.; Hanzlik, R.P. Protein targets of reactive metabolites of thiobenzamide in rat liver in vivo. Chem. Res. Toxicol. 2008, 21, 1432–1442. [Google Scholar] [CrossRef] [Green Version]
  166. Moro, S.; Chipman, J.K.; Antczak, P.; Turan, N.; Dekant, W.; Falciani, F.; Mally, A. Identification and Pathway Mapping of Furan Target Proteins Reveal Mitochondrial Energy Production and Redox Regulation as Critical Targets of Furan Toxicity. Toxicol. Sci. 2012, 126, 336–352. [Google Scholar] [CrossRef] [Green Version]
  167. Lame, M.W.; Jones, A.D.; Wilson, D.W.; Segall, H.J. Monocrotaline pyrrole targets proteins with and without cysteine residues in the cytosol and membranes of human pulmonary artery endothelial cells. Proteomics 2005, 5, 4398–4413. [Google Scholar] [CrossRef]
  168. Lame, M.W.; Jones, A.D.; Wilson, D.W.; Segall, H.J. Protein targets of 1,4-benzoquinone and 1,4-naphthoquinone in human bronchial epithelial cells. Proteomics 2003, 3, 479–495. [Google Scholar] [CrossRef]
  169. Van Laar, V.S.; Mishizen, A.J.; Cascio, M.; Hastings, T.G. Proteomic identification of dopamine-conjugated proteins from isolated rat brain mitochondria and SH-SY5Y cells. Neurobiol. Dis. 2009, 34, 487–500. [Google Scholar] [CrossRef] [Green Version]
  170. Gaviard, C.; Cosette, P.; Jouenne, T.; Hardouin, J. LasB and CbpD Virulence Factors of Pseudomonas aeruginosa Carry Multiple Post-Translational Modifications on Their Lysine Residues. J. Proteome Res. 2019, 18, 923–933. [Google Scholar] [CrossRef]
  171. Forthun, R.B.; Aasebo, E.; Rasinger, J.D.; Bedringaas, S.L.; Berven, F.; Selheim, F.; Bruserud, O.; Gjertsen, B.T. Phosphoprotein DIGE profiles reflect blast differentiation, cytogenetic risk stratification, FLT3/NPM1 mutations and therapy response in acute myeloid leukaemia. J. Proteom. 2018, 173, 32–41. [Google Scholar] [CrossRef] [PubMed]
  172. Drougat, L.; Olivier-Van Stichelen, S.; Mortuaire, M.; Foulquier, F.; Lacoste, A.-S.; Michalski, J.-C.; Lefebvre, T.; Vercoutter-Edouart, A.-S. Characterization of O-GlcNAc cycling and proteomic identification of differentially O-GlcNAcylated proteins during G1/S transition. Biochim. Biophys. Acta BBA Gen. Subj. 2012, 1820, 1839–1848. [Google Scholar] [CrossRef]
  173. Jiang, Z.; Cui, Y.; Wang, L.; Zhao, Y.; Yan, S.; Chang, X. Investigating citrullinated proteins in tumour cell lines. World J. Surg. Oncol. 2013, 11, 260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ishigami, A.; Masutomi, H.; Handa, S.; Nakamura, M.; Nakaya, S.; Uchida, Y.; Saito, Y.; Murayama, S.; Jang, B.; Jeon, Y.-C.; et al. Mass spectrometric identification of citrullination sites and immunohistochemical detection of citrullinated glial fibrillary acidic protein in Alzheimer’s disease brains. J. Neurosci. Res. 2015, 93, 1664–1674. [Google Scholar] [CrossRef] [PubMed]
  175. Amici, A.; Levine, R.L.; Tsai, L.; Stadtman, E.R. Conversion of amino acid residues in proteins and amino acid homopolymers to carbonyl derivatives by metal-catalyzed oxidation reactions. J. Biol. Chem. 1989, 264, 3341–3346. [Google Scholar]
  176. Hatasa, Y.; Chikazawa, M.; Furuhashi, M.; Nakashima, F.; Shibata, T.; Kondo, T.; Akagawa, M.; Hamagami, H.; Tanaka, H.; Tachibana, H.; et al. Oxidative Deamination of Serum Albumins by (-)-Epigallocatechin-3-O-Gallate: A Potential Mechanism for the Formation of Innate Antigens by Antioxidants. PLoS ONE 2016, 11, e153002. [Google Scholar] [CrossRef] [Green Version]
  177. Refsgaard, H.H.; Tsai, L.; Stadtman, E.R. Modifications of proteins by polyunsaturated fatty acid peroxidation products. Proc. Natl. Acad. Sci. USA 2000, 97, 611–616. [Google Scholar] [CrossRef] [Green Version]
  178. Hoff, H.F.; O’Neil, J. Structural and functional changes in LDL after modification with both 4-hydroxynonenal and malondialdehyde. J. Lipid Res. 1993, 34, 1209–1217. [Google Scholar]
  179. De Waal, E.M.; Liang, H.; Pierce, A.; Hamilton, R.T.; Buffenstein, R.; Chaudhuri, A.R. Elevated protein carbonylation and oxidative stress do not affect protein structure and function in the long-living naked-mole rat: A proteomic approach. Biochem. Biophys. Res. Commun. 2013, 434, 815–819. [Google Scholar] [CrossRef]
  180. Hu, W.; Culloty, S.; Darmody, G.; Lynch, S.; Davenport, J.; Ramirez-Garcia, S.; Dawson, K.A.; Lynch, I.; Blasco, J.; Sheehan, D. Toxicity of copper oxide nanoparticles in the blue mussel, Mytilus edulis: A redox proteomic investigation. Chemosphere 2014, 108, 289–299. [Google Scholar] [CrossRef] [Green Version]
  181. Perutka, Z.; Šebela, M. Pseudotrypsin: A Little-Known Trypsin Proteoform. Molecules 2018, 23, 2637. [Google Scholar] [CrossRef] [Green Version]
  182. Keil-Dlouhá, V.; Zylber, N.; Imhoff, J.-M.; Tong, N.-T.; Keil, B. Proteolytic activity of pseudotrypsin. FEBS Lett. 1971, 16, 291–295. [Google Scholar] [CrossRef] [Green Version]
  183. Fortelny, N.; Pavlidis, P.; Overall, C.M. The path of no return-Truncated protein N-termini and current ignorance of their genesis. PROTEOMICS 2015, 15, 2547–2552. [Google Scholar] [CrossRef] [Green Version]
  184. Staes, A.; Impens, F.; Van Damme, P.; Ruttens, B.; Goethals, M.; Demol, H.; Timmerman, E.; Vandekerckhove, J.; Gevaert, K. Selecting protein N-terminal peptides by combined fractional diagonal chromatography. Nat. Protoc. 2011, 6, 1130–1141. [Google Scholar] [CrossRef] [PubMed]
  185. Bertaccini, D.; Vaca, S.; Carapito, C.; Arsène-Ploetze, F.; Van Dorsselaer, A.; Schaeffer-Reiss, C. An Improved Stable Isotope N-Terminal Labeling Approach with Light/Heavy TMPP To Automate Proteogenomics Data Validation: dN-TOP. J. Proteome Res. 2013, 12, 3063–3070. [Google Scholar] [CrossRef] [PubMed]
  186. Marino, G.; Eckhard, U.; Overall, C.M. Protein Termini and Their Modifications Revealed by Positional Proteomics. ACS Chem. Biol. 2015, 10, 1754–1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Schilling, O.; Barré, O.; Huesgen, P.F.; Overall, C.M. Proteome-wide analysis of protein carboxy termini: C terminomics. Nat. Methods 2010, 7, 508–511. [Google Scholar] [CrossRef]
  188. Lee, A.Y.; Park, B.C.; Jang, M.; Cho, S.; Lee, D.H.; Lee, S.C.; Myung, P.K.; Park, S.G. Identification of caspase-3 degradome by two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization-time of flight analysis. Proteomics 2004, 4, 3429–3436. [Google Scholar] [CrossRef]
  189. Kim, C.; Yun, N.; Lee, Y.M.; Jeong, J.Y.; Baek, J.Y.; Song, H.Y.; Ju, C.; Youdim, M.B.H.; Jin, B.K.; Kim, W.-K.; et al. Gel-based protease proteomics for identifying the novel calpain substrates in dopaminergic neuronal cell. J. Biol. Chem. 2013, 288, 36717–36732. [Google Scholar] [CrossRef] [Green Version]
  190. Kim, C.; Oh, Y.J. A Novel 2-DE-Based Proteomic Analysis to Identify Multiple Substrates for Specific Protease in Neuronal Cells. Methods Mol. Biol. Clifton N.J. 2017, 1598, 229–245. [Google Scholar] [CrossRef]
  191. Le Naour, F.; Hohenkirk, L.; Grolleau, A.; Misek, D.E.; Lescure, P.; Geiger, J.D.; Hanash, S.; Beretta, L. Profiling Changes in Gene Expression during Differentiation and Maturation of Monocyte-derived Dendritic Cells Using Both Oligonucleotide Microarrays and Proteomics. J. Biol. Chem. 2001, 276, 17920–17931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Lüthi, A.U.; Cullen, S.P.; Martin, S.J. Chapter Seventeen Two-Dimensional Gel-Based Analysis of the Demolition Phase of Apoptosis. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2008; Volume 442, pp. 343–354. ISBN 978-0-12-374312-1. [Google Scholar]
  193. Marino, R.; Albenzio, M.; Della Malva, A.; Santillo, A.; Loizzo, P.; Sevi, A. Proteolytic pattern of myofibrillar protein and meat tenderness as affected by breed and aging time. Meat Sci. 2013, 95, 281–287. [Google Scholar] [CrossRef] [PubMed]
  194. López-Pedrouso, M.; Pérez-Santaescolástica, C.; Franco, D.; Fulladosa, E.; Carballo, J.; Zapata, C.; Lorenzo, J.M. Comparative proteomic profiling of myofibrillar proteins in dry-cured ham with different proteolysis indices and adhesiveness. Food Chem. 2018, 244, 238–245. [Google Scholar] [CrossRef] [PubMed]
  195. Terova, G.; Addis, M.F.; Preziosa, E.; Pisanu, S.; Pagnozzi, D.; Biosa, G.; Gornati, R.; Bernardini, G.; Roggio, T.; Saroglia, M. Effects of postmortem storage temperature on sea bass (Dicentrarchus labrax) muscle protein degradation: Analysis by 2-D DIGE and MS. Proteomics 2011, 11, 2901–2910. [Google Scholar] [CrossRef] [PubMed]
  196. Addis, M.F.; Pisanu, S.; Preziosa, E.; Bernardini, G.; Pagnozzi, D.; Roggio, T.; Uzzau, S.; Saroglia, M.; Terova, G. 2D DIGE/MS to investigate the impact of slaughtering techniques on postmortem integrity of fish filet proteins. J. Proteom. 2012, 75, 3654–3664. [Google Scholar] [CrossRef]
  197. Ethuin, P.; Marlard, S.; Delosière, M.; Carapito, C.; Delalande, F.; Van Dorsselaer, A.; Dehaut, A.; Lencel, V.; Duflos, G.; Grard, T. Differentiation between fresh and frozen-thawed sea bass (Dicentrarchus labrax) fillets using two-dimensional gel electrophoresis. Food Chem. 2015, 176, 294–301. [Google Scholar] [CrossRef]
  198. Deng, X.; Lei, Y.; Yu, Y.; Lu, S.; Zhang, J. The Discovery of Proteins Associated with Freshness of Coregonus Peled Muscle During Refrigerated Storage. J. Food Sci. 2019, 84, 1266–1272. [Google Scholar] [CrossRef]
  199. Pepe, M.S.; Etzioni, R.; Feng, Z.; Potter, J.D.; Thompson, M.L.; Thornquist, M.; Winget, M.; Yasui, Y. Phases of Biomarker Development for Early Detection of Cancer. JNCI J. Natl. Cancer Inst. 2001, 93, 1054–1061. [Google Scholar] [CrossRef] [Green Version]
  200. Rifai, N.; Gillette, M.A.; Carr, S.A. Protein biomarker discovery and validation: The long and uncertain path to clinical utility. Nat. Biotechnol. 2006, 24, 971–983. [Google Scholar] [CrossRef]
  201. Mischak, H.; Apweiler, R.; Banks, R.E.; Conaway, M.; Coon, J.; Dominiczak, A.; Ehrich, J.H.H.; Fliser, D.; Girolami, M.; Hermjakob, H.; et al. Clinical proteomics: A need to define the field and to begin to set adequate standards. PROTEOMICS Clin. Appl. 2007, 1, 148–156. [Google Scholar] [CrossRef]
  202. Hristova, V.A.; Chan, D.W. Cancer biomarker discovery and translation: Proteomics and beyond. Expert Rev. Proteom. 2019, 16, 93–103. [Google Scholar] [CrossRef] [PubMed]
  203. Charrier, J.P.; Tournel, C.; Michel, S.; Dalbon, P.; Jolivet, M. Two-dimensional electrophoresis of prostate-specific antigen in sera of men with prostate cancer or benign prostate hyperplasia. Electrophoresis 1999, 20, 1075–1081. [Google Scholar] [CrossRef]
  204. Charrier, J.P.; Tournel, C.; Michel, S.; Comby, S.; Jolivet-Reynaud, C.; Passagot, J.; Dalbon, P.; Chautard, D.; Jolivet, M. Differential diagnosis of prostate cancer and benign prostate hyperplasia using two-dimensional electrophoresis. Electrophoresis 2001, 22, 1861–1866. [Google Scholar] [CrossRef]
  205. Kondo, T. Cancer biomarker development and two-dimensional difference gel electrophoresis (2D-DIGE). Biochim. Biophys. Acta BBA Proteins Proteom. 2019, 1867, 2–8. [Google Scholar] [CrossRef] [PubMed]
  206. Uemura, N.; Nakanishi, Y.; Kato, H.; Saito, S.; Nagino, M.; Hirohashi, S.; Kondo, T. Transglutaminase 3 as a prognostic biomarker in esophageal cancer revealed by proteomics. Int. J. Cancer 2009, 124, 2106–2115. [Google Scholar] [CrossRef]
  207. Okano, T.; Kondo, T.; Fujii, K.; Nishimura, T.; Takano, T.; Ohe, Y.; Tsuta, K.; Matsuno, Y.; Gemma, A.; Kato, H.; et al. Proteomic Signature Corresponding to the Response to Gefitinib (Iressa, ZD1839), an Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor in Lung Adenocarcinoma. Clin. Cancer Res. 2007, 13, 799–805. [Google Scholar] [CrossRef] [Green Version]
  208. Yokoo, H.; Kondo, T.; Okano, T.; Nakanishi, K.; Sakamoto, M.; Kosuge, T.; Todo, S.; Hirohashi, S. Protein expression associated with early intrahepatic recurrence of hepatocellular carcinoma after curative surgery. Cancer Sci. 2007, 98, 665–673. [Google Scholar] [CrossRef]
  209. Orimo, T.; Ojima, H.; Hiraoka, N.; Saito, S.; Kosuge, T.; Kakisaka, T.; Yokoo, H.; Nakanishi, K.; Kamiyama, T.; Todo, S.; et al. Proteomic profiling reveals the prognostic value of adenomatous polyposis coli-end-binding protein 1 in hepatocellular carcinoma. Hepatology 2008, 48, 1851–1863. [Google Scholar] [CrossRef]
  210. Kimura, K.; Ojima, H.; Kubota, D.; Sakumoto, M.; Nakamura, Y.; Tomonaga, T.; Kosuge, T.; Kondo, T. Proteomic identification of the macrophage-capping protein as a protein contributing to the malignant features of hepatocellular carcinoma. J. Proteom. 2013, 78, 362–373. [Google Scholar] [CrossRef]
  211. Suehara, Y.; Kondo, T.; Seki, K.; Shibata, T.; Fujii, K.; Gotoh, M.; Hasegawa, T.; Shimada, Y.; Sasako, M.; Shimoda, T.; et al. Pfetin as a prognostic biomarker of gastrointestinal stromal tumors revealed by proteomics. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2008, 14, 1707–1717. [Google Scholar] [CrossRef] [Green Version]
  212. Kikuta, K.; Gotoh, M.; Kanda, T.; Tochigi, N.; Shimoda, T.; Hasegawa, T.; Katai, H.; Shimada, Y.; Suehara, Y.; Kawai, A.; et al. Pfetin as a prognostic biomarker in gastrointestinal stromal tumor: Novel monoclonal antibody and external validation study in multiple clinical facilities. Jpn. J. Clin. Oncol. 2010, 40, 60–72. [Google Scholar] [CrossRef]
  213. Kubota, D.; Okubo, T.; Saito, T.; Suehara, Y.; Yoshida, A.; Kikuta, K.; Tsuda, H.; Katai, H.; Shimada, Y.; Kaneko, K.; et al. Validation study on pfetin and ATP-dependent RNA helicase DDX39 as prognostic biomarkers in gastrointestinal stromal tumour. Jpn. J. Clin. Oncol. 2012, 42, 730–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Lescuyer, P.; Allard, L.; Zimmermann-Ivol, C.G.; Burgess, J.A.; Hughes-Frutiger, S.; Burkhard, P.R.; Sanchez, J.-C.; Hochstrasser, D.F. Identification of post-mortem cerebrospinal fluid proteins as potential biomarkers of ischemia and neurodegeneration. Proteomics 2004, 4, 2234–2241. [Google Scholar] [CrossRef]
  215. Allard, L.; Burkhard, P.R.; Lescuyer, P.; Burgess, J.A.; Walter, N.; Hochstrasser, D.F.; Sanchez, J.-C. PARK7 and nucleoside diphosphate kinase A as plasma markers for the early diagnosis of stroke. Clin. Chem. 2005, 51, 2043–2051. [Google Scholar] [CrossRef] [PubMed]
  216. Turck, N.; Vutskits, L.; Sanchez-Pena, P.; Robin, X.; Hainard, A.; Gex-Fabry, M.; Fouda, C.; Bassem, H.; Mueller, M.; Lisacek, F.; et al. A multiparameter panel method for outcome prediction following aneurysmal subarachnoid hemorrhage. Intensive Care Med. 2010, 36, 107–115. [Google Scholar] [CrossRef] [Green Version]
  217. Turck, N.; Robin, X.; Walter, N.; Fouda, C.; Hainard, A.; Sztajzel, R.; Wagner, G.; Hochstrasser, D.F.; Montaner, J.; Burkhard, P.R.; et al. Blood glutathione S-transferase-π as a time indicator of stroke onset. PLoS ONE 2012, 7, e43830. [Google Scholar] [CrossRef]
  218. Lagerstedt, L.; Egea-Guerrero, J.J.; Bustamante, A.; Rodríguez-Rodríguez, A.; El Rahal, A.; Quintana-Diaz, M.; García-Armengol, R.; Prica, C.M.; Andereggen, E.; Rinaldi, L.; et al. Combining H-FABP and GFAP increases the capacity to differentiate between CT-positive and CT-negative patients with mild traumatic brain injury. PLoS ONE 2018, 13, e0200394. [Google Scholar] [CrossRef]
  219. Posti, J.P.; Takala, R.S.K.; Lagerstedt, L.; Dickens, A.M.; Hossain, I.; Mohammadian, M.; Ala-Seppälä, H.; Frantzén, J.; van Gils, M.; Hutchinson, P.J.; et al. Correlation of Blood Biomarkers and Biomarker Panels with Traumatic Findings on Computed Tomography after Traumatic Brain Injury. J. Neurotrauma 2019, 36, 2178–2189. [Google Scholar] [CrossRef] [PubMed]
  220. Mölleken, C.; Sitek, B.; Henkel, C.; Poschmann, G.; Sipos, B.; Wiese, S.; Warscheid, B.; Broelsch, C.; Reiser, M.; Friedman, S.L.; et al. Detection of novel biomarkers of liver cirrhosis by proteomic analysis. Hepatology 2009, 49, 1257–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Bracht, T.; Mölleken, C.; Ahrens, M.; Poschmann, G.; Schlosser, A.; Eisenacher, M.; Stühler, K.; Meyer, H.E.; Schmiegel, W.H.; Holmskov, U.; et al. Evaluation of the biomarker candidate MFAP4 for non-invasive assessment of hepatic fibrosis in hepatitis C patients. J. Transl. Med. 2016, 14, 201. [Google Scholar] [CrossRef] [Green Version]
  222. Lottspeich, F.; Kellner, R. Microcharacterrization of Proteins, 1st ed.; Kellner, R., Lottspeich, F., Meyer, H.E., Eds.; Wiley: Hoboken, NJ, USA, 1999; ISBN 978-3-527-30084-6. [Google Scholar]
  223. Tran, J.C.; Zamdborg, L.; Ahlf, D.R.; Lee, J.E.; Catherman, A.D.; Durbin, K.R.; Tipton, J.D.; Vellaichamy, A.; Kellie, J.F.; Li, M.; et al. Mapping intact protein isoforms in discovery mode using top-down proteomics. Nature 2011, 480, 254–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Kelleher, N.L. Top-down proteomics. Anal. Chem. 2004, 76, 197A–203A. [Google Scholar] [CrossRef] [Green Version]
  225. Claverol, S.; Burlet-Schiltz, O.; Girbal-Neuhauser, E.; Gairin, J.E.; Monsarrat, B. Mapping and structural dissection of human 20 S proteasome using proteomic approaches. Mol. Cell. Proteom. MCP 2002, 1, 567–578. [Google Scholar] [CrossRef] [Green Version]
  226. Claverol, S.; Burlet-Schiltz, O.; Gairin, J.E.; Monsarrat, B. Characterization of Protein Variants and Post-Translational Modifications: ESI-MSn Analyses of Intact Proteins Eluted from Polyacrylamide Gels. Mol. Cell. Proteom. 2003, 2, 483–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Kachuk, C.; Stephen, K.; Doucette, A. Comparison of sodium dodecyl sulfate depletion techniques for proteome analysis by mass spectrometry. J. Chromatogr. A 2015, 1418, 158–166. [Google Scholar] [CrossRef] [PubMed]
  228. Sun, G.; Anderson, V.E. Prevention of artifactual protein oxidation generated during sodium dodecyl sulfate-gel electrophoresis. Electrophoresis 2004, 25, 959–965. [Google Scholar] [CrossRef] [PubMed]
  229. Rabilloud, T. Variations on a theme: Changes to electrophoretic separations that can make a difference. J. Proteom. 2010, 73, 1562–1572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Comparison of two-dimensional gel electrophoresis (2DGE) and shotgun MS-workflow. The main differences between the two workflows are the type of molecule separation and the method of quantification. While in the 2DGE approach, (A) the proteins are already separated; in the shotgun approach, (B) the proteins are first digested and then the resulting peptides are separated. In (A) proteins or proteoforms are detected, quantified, and identified, whereas in (B) the detection, identification and quantification is performed at the peptide level. The peptide data are then used for protein reclustering.
Figure 1. Comparison of two-dimensional gel electrophoresis (2DGE) and shotgun MS-workflow. The main differences between the two workflows are the type of molecule separation and the method of quantification. While in the 2DGE approach, (A) the proteins are already separated; in the shotgun approach, (B) the proteins are first digested and then the resulting peptides are separated. In (A) proteins or proteoforms are detected, quantified, and identified, whereas in (B) the detection, identification and quantification is performed at the peptide level. The peptide data are then used for protein reclustering.
Proteomes 08 00017 g001
Figure 2. Variability of haptoglobin peptides in cerebrospinal fluid (CSF). Two tryptic peptides (SPVGVQPILNEHTFCAGMSK, black, and VTSIQDWVQK, green) were selected as representatives for haptoglobin. Both peptides were detected in 36 CSF samples of healthy control subjects. In samples 11, 27, and 29 (marked with a red circle) the blue peptide was more intense than the green peptide. In all other samples, the intensity of the green peptide was similar or higher than the intensity of the blue peptide. Additionally, the difference in intensity of both peptides showed high variability in at least some of the samples (e.g., 31, 33–35, and 36, marked with a black circle). Summarized, the variability of intensity of some of the unique tryptic haptoglobin peptides has made a valid protein quantification on basis of the peptide amounts in this study cohort impossible.
Figure 2. Variability of haptoglobin peptides in cerebrospinal fluid (CSF). Two tryptic peptides (SPVGVQPILNEHTFCAGMSK, black, and VTSIQDWVQK, green) were selected as representatives for haptoglobin. Both peptides were detected in 36 CSF samples of healthy control subjects. In samples 11, 27, and 29 (marked with a red circle) the blue peptide was more intense than the green peptide. In all other samples, the intensity of the green peptide was similar or higher than the intensity of the blue peptide. Additionally, the difference in intensity of both peptides showed high variability in at least some of the samples (e.g., 31, 33–35, and 36, marked with a black circle). Summarized, the variability of intensity of some of the unique tryptic haptoglobin peptides has made a valid protein quantification on basis of the peptide amounts in this study cohort impossible.
Proteomes 08 00017 g002
Table 1. Strengths and weaknesses of 2DGE proteomics vs. shotgun proteomics.
Table 1. Strengths and weaknesses of 2DGE proteomics vs. shotgun proteomics.
2D Gel-based ProteomicsShotgun Proteomics
Sample consuming++(+) *+
Time consuming+++++
Analysis depth+++++
Separation/identification
Separation/detection of proteoforms ++++
Identification on protein level Multiple identificationsOnly by inference
from peptides
Detection of proteoforms +++-
Details at peptide level (e.g., sequence coverage)++++
Number of modulated proteins identified++++
Coupling with biochemical methods
Antibodies++++
Enzymes (zymography)+-
Robustness of quantification
Sensitivity+++++
Linearity++++
Need of validation++++++
* depending on 2D gel-based technique used.

Share and Cite

MDPI and ACS Style

Marcus, K.; Lelong, C.; Rabilloud, T. What Room for Two-Dimensional Gel-Based Proteomics in a Shotgun Proteomics World? Proteomes 2020, 8, 17. https://doi.org/10.3390/proteomes8030017

AMA Style

Marcus K, Lelong C, Rabilloud T. What Room for Two-Dimensional Gel-Based Proteomics in a Shotgun Proteomics World? Proteomes. 2020; 8(3):17. https://doi.org/10.3390/proteomes8030017

Chicago/Turabian Style

Marcus, Katrin, Cécile Lelong, and Thierry Rabilloud. 2020. "What Room for Two-Dimensional Gel-Based Proteomics in a Shotgun Proteomics World?" Proteomes 8, no. 3: 17. https://doi.org/10.3390/proteomes8030017

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