Next Article in Journal
Leucine Supplementation Improves Diastolic Function in HFpEF by HDAC4 Inhibition
Previous Article in Journal
Unveiling Mesenchymal Stem Cells’ Regenerative Potential in Clinical Applications: Insights in miRNA and lncRNA Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mass Spectrometry-Based Proteomic Technology and Its Application to Study Skeletal Muscle Cell Biology

by
Paul Dowling
1,2,
Dieter Swandulla
3 and
Kay Ohlendieck
1,2,*
1
Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
2
Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
3
Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany
*
Author to whom correspondence should be addressed.
Cells 2023, 12(21), 2560; https://doi.org/10.3390/cells12212560
Submission received: 6 October 2023 / Revised: 27 October 2023 / Accepted: 31 October 2023 / Published: 1 November 2023
(This article belongs to the Section Tissues and Organs)

Abstract

:
Voluntary striated muscles are characterized by a highly complex and dynamic proteome that efficiently adapts to changed physiological demands or alters considerably during pathophysiological dysfunction. The skeletal muscle proteome has been extensively studied in relation to myogenesis, fiber type specification, muscle transitions, the effects of physical exercise, disuse atrophy, neuromuscular disorders, muscle co-morbidities and sarcopenia of old age. Since muscle tissue accounts for approximately 40% of body mass in humans, alterations in the skeletal muscle proteome have considerable influence on whole-body physiology. This review outlines the main bioanalytical avenues taken in the proteomic characterization of skeletal muscle tissues, including top-down proteomics focusing on the characterization of intact proteoforms and their post-translational modifications, bottom-up proteomics, which is a peptide-centric method concerned with the large-scale detection of proteins in complex mixtures, and subproteomics that examines the protein composition of distinct subcellular fractions. Mass spectrometric studies over the last two decades have decisively improved our general cell biological understanding of protein diversity and the heterogeneous composition of individual myofibers in skeletal muscles. This detailed proteomic knowledge can now be integrated with findings from other omics-type methodologies to establish a systems biological view of skeletal muscle function.

1. Introduction

Modern cell biology is concerned with the detailed analysis of the structure and function of the basic cellular units of life and how they form highly complex structures in the form of cells, tissues, organs, bodily systems and individual organisms. This includes the detailed elucidation of protein expression levels, protein modifications and protein dynamics on the molecular level within the cellular context [1]. The application of mass spectrometry (MS)-based proteomics had a considerable impact on molecular cell biology [2], focusing especially on the analysis of cellular proteomes and organellar subproteomes [3]. The establishment of proteomics as both a cell biological concept, i.e., defining the entire dynamic set of proteins expressed by a given biological entity as a proteome [4], and a comprehensive bioanalytical approach, i.e., using large-scale protein separation techniques in combination with peptide/protein MS methods and bioinformatic software programs for high-throughput protein identification or the targeted characterization of isolated proteins [5], has revolutionized modern cell biology.
Of central importance for proteomic research is the biochemical definition of dynamic proteoforms being the basic units of a given proteome [6,7,8,9]. The proteolytic processing and post-translational modifications (PTMs) of individual protein species play a key role in their stability, biochemical functioning and subcellular localization [10]. Importantly, proteomics has reached single-cell resolution [11], giving unprecedented insights into the dynamic nature of the protein constituents that mediate cellular processes [12]. Major MS-based approaches in molecular cell biology include bottom-up proteomics and top-down proteomics [13]. Bottom-up proteomics aims at the peptide-based analysis of large and diverse protein populations [14,15], while top-down proteomics has a proteoform-centric focus and is concerned with the biochemical identification and detailed characterization of purified and intact protein species [16,17]. The bioanalytical combination of both methodologies can be strategically employed for improved data integration [18,19]. The skeletal muscle proteome project [20] was initiated in 2001 using initially top-down and gel-based separation methodology [21,22]. Subsequently, MS-based surveys have been widely applied to study almost all crucial aspects of skeletal muscle cell biology using both bottom-up and top-down proteomics [23,24]. This has led to the creation of a detailed proteomic map of different types of skeletal muscles [25,26,27,28] including predominantly fast-twitching versus slow-twitching muscles [29,30,31,32] and the exhaustive biochemical profiling of individual contractile fiber types [33,34,35,36].
A large number of extensive reviews have previously been published that cover specific biochemical and cell biological approaches and the underlying scientific scope of proteomic profiling with a focus on voluntary muscle tissues. These articles give a comprehensive overview of the many applications of MS-based analyses to improve our molecular and cellular understanding of skeletal muscle protein diversity, modifications and dynamics. Previous reviews have extensively covered the proteomic analysis of human skeletal muscle [20,37], the impact of exercise [38,39,40], disuse-related muscular atrophy [41], skeletal muscle plasticity [42,43], neuromuscular diseases [44,45] and frailty-associated muscle aging [46,47]. Building on these articles, we provide here a general synopsis of skeletal muscle proteomics and its scientific importance for the field of basic and applied myology. Examples of skeletal muscle proteomics are discussed, as well as an outline given on the impact of genomic, post-transcriptional and post-translational factors on muscle proteome diversity.

2. Mass Spectrometry-Based Proteomics to Study Skeletal Muscle Cell Biology

Expression changes, biochemical modifications and dynamic adaptations of proteins in their cellular context are of critical importance for understanding the molecular pathways that underlie fundamental biological processes. This positions MS-based proteomics at the core of the comprehensive bioanalysis of cell biological mechanisms, such as the excitation-contraction-relaxation cycle in skeletal muscles.

2.1. The Importance of Proteomics and the Concept of Proteoforms in the Biological Sciences

Since the initiation of proteome-wide research initiatives in the 1990s [48], MS techniques were widely applied for the comprehensive analysis of a variety of cells, tissues and organisms, including the human proteome [49,50,51,52], which had enormous consequences for the scientific scope of biological and biomedical research [53,54,55]. At the level of individual protein species, the term proteoform [6,7] best describes the basic unit of the proteome [56,57] and the central question remains of how many proteoforms exist within a given proteome [58,59,60]. Genomic variations at the level of allelic or locus heterogeneity, combined with post-transcriptional alternative RNA splicing events and extensive PTMs of proteins result in a highly complex and dynamic proteoform expression pattern [10]. Therefore, most single genes appear to be associated with a family of proteoforms rather than a sole protein product. The approximately 20,000 human genes are estimated to produce millions of distinct protein species [61]. This gives proteoform-focused research a central space in modern cell biology and promises to shed light on how protein heterogeneity is mediated by transcriptional/translational modifications in combination with the formation of higher-order protein structures and dynamic protein–protein interactions resulting in distinct biological phenotypes [62].
The rapid developments of novel proteomic technologies and applications has led to the establishment of many specialized approaches focusing on targeted biomarker proteomics [63,64], clinical proteomics [65,66], drug discovery and pharmaco-proteomics [67,68,69], proteo-genomics [70,71], membrane proteomics [72,73], extracellular matrix proteomics [74,75], native proteomics [76,77,78], thermal proteome profiling [79], the analysis of protein conformations and interactions via chemical cross-linking MS [80,81] and subcellular/organelle proteomics [82,83]. The more recently developed discipline of native MS focuses on the detailed top-down characterization of structural heterogeneity in protein isoforms under non-denaturing conditions, making this approach an important part of structural cell biology [84,85]. Together with genomics, transcriptomics, metabolomics, lipidomics, glycomics and cytomics, modern proteomics is embedded within a multi-omics research environment and plays an integral part in systems biology [86]. Techniques beyond MS include antibody-based screening methods for establishing a comprehensive protein expression atlas in all major tissue types [87] and next-generation proteomics [88] leading to a more comprehensive view of biological function through integromics research [89].

2.2. Large-Scale Protein Separation and Top-Down Proteomics

In the field of skeletal muscle proteomics, the isolation of native protein arrangements and intact proteoforms is of crucial importance for the detailed analysis of muscle protein function [23,24]. The initial protein separation and isolation from complex samples prior to MS analysis is usually carried out using subcellular fractionation procedures [90] involving (i) differential centrifugation or gradient ultra-centrifugation [91], (ii) affinity agglutination of distinct membranes [92], (iii) isoelectric focusing [93], (iv) one-dimensional gel electrophoresis [94], (v) two-dimensional gel electrophoresis [95] and/or (vi) reversed-phase liquid chromatography [96].
For gel-based top-down proteomics, which is routinely used for the isolation of intact proteoforms prior to controlled proteolysis and peptide MS analysis [6,97], the optimum application of two-dimensional gel electrophoresis (2D-GE) is of central importance [98,99,100]. Using isoelectric focusing (IEF) in the first dimension and sodium dodecyl sulphate-polyacrylamide slab gel electrophoresis (SDS-PAGE) in the second dimension [101,102,103], proteins can be efficiently separated by the unique combination of their overall charge and molecular size [104,105,106]. Recently, micro-needling was introduced to improve the time requirements for the IEF step in 2D-GE [107]. Proteins can be separated under both denatured or native conditions. In addition to using sodium dodecyl sulfate (SDS) exclusively in second dimension gels [108,109,110], 2D-gels can be operated with combinations of alternative detergents to improve the resolution of hydrophobic membrane proteins [111]. A specific method uses in the first dimension the cationic detergent benzyldime-thyl-n-hexadecylammonium chloride (BAC) and in the second dimension the anionic component SDS; therefore, this technique is abbreviated as BAC/SDS-PAGE [112]. Two-dimensional blue native polyacrylamide gel electrophoresis (BN-PAGE) employs native conditions [113,114] and is especially useful for studying supramolecular protein complexes present in mitochondria [115]. Diagonal non-reducing/reducing two-dimensional gel electrophoresis can be used to exploit both natural or modified differences between proteins and protein complexes during large-scale separation approaches following chemical cross-linking [116,117,118]. Suitable and highly sensitive dyes have to be used for the optimum visualization of two-dimensionally separated protein spots and subsequent sample picking for MS-based analysis [119,120,121].
Labelling of different proteoforms can be achieved using differential fluorescent tagging with CyDye2, CyDye3 and CyDye5 agents prior to 2D-GE [122,123]. High-throughput comparative studies can be performed via comparative two-dimensional fluorescence gel electrophoresis (CoFGE) [124,125]. Difference gel electrophoresis (2D-DIGE) can be carried out with 2-CyDye or 3-CyDye systems using saturation versus minimal labelling approaches [126,127]. Of note, 2D-GE, including the 2D-DIGE technique, can be employed effectively to study native protein configurations [128,129], as well as PTMs using specific chemical stains or blot overlay methods. Phosphoprotein gel stains, e.g., Pro-Q Diamond, provide a useful approach for selectively staining phosphoproteins in 2D gels [130]. By combining a standard DIGE or SYPRO Ruby experimental workflow with phospho-specific fluorescent Pro-Q Diamond staining [131], the sensitive and quantitative detection of phosphoproteins in 2D gels can be accomplished [132,133]. Other stains or blotting techniques routinely used include those targeting glycosylated proteins in 2D gels, e.g., Pro-Q Emerald [134] or peroxidase-conjugated wheat germ agglutinin [135], providing a rapid and very sensitive methodology for detecting glycoproteins of interest [136]. A more comprehensive description of PTM analyses is provided below. As an alternative to 2D-GE, multi-dimensional protein identification technology (MudPIT) can be employed, which is a non-gel technique for protein separation that uses two-dimensional liquid chromatography prior to MS-based analysis [137,138]. The initial preparative steps involved in the characterization of skeletal muscle proteins using top-down proteomics versus bottom-up proteomics are summarized in Figure 1.
Very large protein species that can often not be properly separated by conventional 2D-GE can be analyzed by combining 1D-GE, liquid chromatography and tandem MS (GeLC-MS/MS) methodology [139,140,141]. The stabilization of sensitive protein–protein interactions in skeletal muscle tissues can be supported by chemical cross-linking [142], a method that is becoming increasingly used to study dynamic protein conformations [80,81,84]. Especially the quantification and structural analysis of integral proteins [143] using MS-based methods is a challenging task [144,145,146]. High-resolution native MS techniques have been developed to study the dynamic nature of protein complexes [147,148,149].

2.3. Sample Preparation, Protein Digestion and Bottom-Up Proteomics

For bottom-up proteomics, a variety of sample preparation approaches have been developed for optimum protein extraction [150,151,152], including the widely employed filter-aided sample preparation (FASP) method that can be used to exchange buffers and remove incompatible detergents prior to MS-based analysis [153]. The routine bottom-up proteomic analysis pipeline can be divided into a series of consecutive steps [152] comprising (i) sample harvesting from complex cellular mixtures, (ii) sample pre-treatment, such as enrichment procedures using differential centrifugation or affinity isolation of protein constellations, or depletion procedures to enrich low-abundance proteins, (iii) initial sample protection from excess proteolysis using suitable protease inhibitor cocktails, (iv) efficient homogenization using mechanical grinding approaches or sample pulverization with liquid nitrogen, (v) protein extraction via precipitation or other biochemical methods, (vi) protein quantification using sensitive dye binding or other reliable assay systems, (vii) chemical reduction and alkylation of protein samples, (viii) protein digestion using enzymes such as trypsin for the generation of distinct peptide populations, (ix) peptide fractionation and MS analysis, (x) protein identification, and (xi) systems bioinformatic analysis of identified proteoforms and their PTMs [154,155,156]. Figure 2 compares the main differences in the preparation of protein samples between top-down proteomics and bottom-up proteomics, i.e., the isolation of intact proteoforms versus the enrichment of protein fractions.
Specific requirements of the starting material, i.e., cells, tissue specimens or biofluids, have to be taken into account for the most suitable extraction of protein populations. For the detection of muscle-derived marker proteins in biological fluids such as serum, saliva or urine, biofluids should be screened with optimized proteomics platforms [157] and usually require immuno-depletion, immuno-enrichment or offline fractionation techniques for the optimum isolation of low-abundance proteins [158,159,160]. In contrast, unfractionated and crude tissue samples can be conveniently prepared using FASP [161], In-StageTip (iST) [162], suspension trapping (S-Trap) [163], single-pot solid-phase-enhanced sample preparation (SP3) [164] or universal solid-phase protein preparation (USP3) [165] methodology. Importantly, the application of single-cell isolation is becoming more crucial for sample preparation in proteomics [166]. For the dependable screening of thousands of prioritized peptides with increased proteome depth, the prioritized Single-Cell ProtEomics (pSCoPE) analysis platform was developed [167]. Some of the bioanalytical issues and technical limitations encountered with the lysis and proteolytic digestion of cell or tissue samples [168] can be overcome with pressure cycling technology (PCT) [169].
Following isolation, the controlled digestion of proteins can be performed via in-solution [170,171], in-gel [172,173] or on-membrane [94,174,175] treatment with a suitable enzyme such as trypsin [176] or a great variety of other proteases used alone or in combination, including chymotrypsin, Glu-C, Asp-N, Arg-C, Lys-N and Lys-C [177,178,179]. Recently, a more rapid in-gel digestion protocol was developed to improve the GeLC-MS/MS technique [180], which can be applied to study very large proteins that cannot be separated by conventional 2D-GE [139]. Using BAC cross-linked gels, these separation media can be rapidly solubilized via chemical reduction. This new method has been named BAC-gel dissolution to digest PAGE-resolved objective proteins (BAC-DROP) [181].

2.4. Mass Spectrometric Protein Identification

Routine protein identification can be carried out using matrix-assisted laser desorption/ionization time-of-flight (MALDI-ToF) MS or electrospray ionization (ESI) MS analysis [182]. Liquid chromatography and the fragmentation of peptides can be employed to determine the amino acid sequence and PTMs of interest via tandem MS analysis (MS/MS) [183,184,185] and a variety of related MS methods [186,187], including single-cell MS [188]. A ToF-based analyzer determines efficiently the mass-to-charge ratio (m/z) using the time ions require to travel through an electric field under vacuum [189]. This is combined with the MALDI-mediated detection of peptides [190]. The optimized usage of reversed-phase liquid chromatography is crucial prior to the direct injection of peptides using ESI [191] or related techniques for MS analysis [192]. The detailed characterization of proteoforms can be carried out using Fourier-transform ion cyclotron resonance MS (FTMS) [193]. MS analyses can be performed with label-free quantification (LFQ) approaches [194,195] or quantitative label-based methods [196,197].
The acquisition of data sets in MS-based studies can be attained via data-dependent acquisition (DDA) [198], data-independent acquisition (DIA) [199] or targeted data acquisition (TDA) [200]. DIA-based methods are ideal for bottom-up proteomics focusing on the large-scale and LFQ analysis of proteoforms [201]. In the specialized biochemical discipline of targeted proteomics [202], multiplexed protein quantification can be carried out with the help of selected/multiple reaction monitoring (SRM/MRM) [203] and parallel reaction monitoring (PRM) techniques [204], which are characterized by high degrees of bioanalytical accuracy, specificity and reproducibility [205]. Targeted label-free data-acquisition can be performed with Sequential Window Acquisition of all Theoretical Mass Spectra (SWATH) [206,207]. Quantitative label-based methods used in MS-based analyses include stable isotope labelling using amino acids in cell culture (SILAC) [208,209], the isobaric tagging for relative and absolute quantitation (iTRAQ) technique [196,210], isotope-coded affinity tags (ICAT) [211,212], isotope-coded protein labelling (ICPL) [213] and isobaric tandem mass tagging (TMT) [197,214].
Advances in instrumentation and proteomic screening methodology are represented by BOXCAR, RTS-SPS-MS3, FAIMS, Evosep, TIMS, SOMAscan and PEA. BOXCAR is a data-independent acquisition method that fills multiple narrow mass-to-charge segments and allows for in-depth proteomic profiling [215]. RTS-SPS-MS3 is based on real-time search (RTS) synchronous precursor selection (SPS)-assisted acquisition, which improves proteomic coverage especially in multiplexed single-cell analyses [216]. FAIMS stands for high-field asymmetric ion mobility spectrometry. This method uses a mass spectrometer that is equipped with a front-end FAIMS interface that improves proteomic coverage [217]. Evosep is a liquid chromatography system for improved high-throughput peptide analysis [218]. TIMS uses trapped ion mobility spectrometry with parallel accumulation serial fragmentation (PASEF) operation mode and provides high analytical speed and sensitivity [219]. In addition, multiplexed proteomic assays have been developed for the parallel assessment of large numbers of proteins in biofluids using both modified aptamers (SOMAscan) and proximity extension assays (PEA) for alternative immuno-analysis [220,221]. Major methods used in the MS-based analysis of skeletal muscles and proteomic data acquisition are summarized in Figure 3.
Verification analyses to confirm the MS-based identification of proteomic changes on the biochemical and cellular level can be conveniently performed using standardized immunoblotting [222,223], enzyme assays [224,225], confocal microscopy [226] and histochemical analyses [227].

2.5. Microproteomic Analysis Using Laser Capture Microdissection

A key technique of molecular cell biology, laser-assisted microdissection [228], can be conveniently adapted for microproteomic applications [229] and used to study protein populations in small cellular regions of interest [230,231]. In general, the highly focused isolation of cellular structures employing direct microscopical visualization of tissues can be used to generate detailed systems biological maps through the bioanalysis of pure histological specimens [232]. Within a targeted microscopical area, the application of laser-assisted microdissection can be employed to determine spatial signatures of distinct cellular layers. Micro-dissected tissue specimens can be used to generate cDNA libraries, the systematic genotyping of DNA, RNA transcript profiling, detailed protein pathway analyses, advanced biomarker discovery and spatial proteomics [233]. Optimized microproteomic approaches have been instrumental for advanced MS-based analysis of cell type-specific properties and their cell biological microenvironment [234].
To avoid analyte loss during sample preparation for spatial proteomics, cellular specimens isolated via laser capture microdissection, or alternatively flow cytometry, should be properly prepared for the subsequent MS-based analysis. This can include carefully executed cell biological disruption methods, such as cellular lysis, protein extraction and sample clean-up, and optimized protein separation using liquid chromatographic or gel electrophoretic methodology [235]. Laser capture proteomics has been widely applied to carry out spatial analyses of diverse tissue types, including various skeletal muscle preparations [33,236] focusing, for example, on the analysis of myoferlin regulation during myofiber injury and regeneration [237], the determination of myosin content in individual human myofibers [238] and the proteomic assessment of the myotendinous junction [239].

2.6. Proteomic Analysis of Post-Translational Modifications

As already briefly outlined in the above section on the most commonly used MS approaches, proteomics is not only employed for the routine identification of proteoforms and the determination of abundance changes in individual proteins, but also represents an excellent method for the detailed analysis of PTMs [154,155]. Of note, major types of PTMs, such as phosphorylation, glycosylation, ubiquitination and methylation are essential for regulating protein stability, interactions and cellular localization [240]. Some PTMs can be added or removed to adapt and control the level of intracellular signaling cascade activation through the relationship between modifying and demodifying enzymes [241]. These enzymes include kinases, phosphatases, peroxidases, transferases, ligases and deubiquitylases, which can add or remove functional groups, with an estimated 5% of the proteome covering such enzymes. PTMs are increasingly investigated for their crucial role in physiology and pathophysiology [242]. Importantly, the detailed and specific detection of abnormal PTM events provides a potential reservoir of biomarkers for clinical utility, including early diagnosis, prognosis and to predict responses to therapeutic regimens in neuromuscular disease. A greater understanding of abnormal PTMs in specific proteins also provides an opportunity to accurately pursue such manifestations therapeutically and add to the already expanding list of drug targets in skeletal muscles. Accurate detection and analysis of PTMs is crucial for understanding their functional implications, and while technical difficulties limit these studies, recent advancements in these areas are now facilitating researchers in establishing a greater understanding of this complex area.
MS-based analysis of protein PTMs represents a sensitive and high-resolution approach that typically utilizes bottom-up proteomic approaches. The analytical approach relies on the controlled digestion of proteins for generating peptides using enzymes such as trypsin and/or lysyl endopeptidase (Lys-C) to perform this preparative action [154], as already outlined above. The enrichment of specifically modified peptides is an important consideration, as the creation of a pool of modified peptides, distinct from their unmodified counterparts, will reduce sample density and facilitate efficient PTM identification and quantification via MS. Peptide enrichment can be achieved using affinity strategies, targeting specific PTMs and concentrating their abundance in precise fractions. Affinity strategies can involve chromatographic separation based on unique chemical properties or purification based on antibody/protein domain recognition.
Immobilized metal affinity chromatography (IMAC) has been routinely used for phospho-peptide enrichment prior to MS analysis [243], with the use of positively charged metal ions including titanium (Ti4+), gallium (Ga3+) and iron (Fe3+) being central to this process [244]. Anti-phospho-tyrosine (pTyr) antibodies are frequently used to enrich tyrosine-phosphorylated peptides, as pTyr is significantly lower in abundance compared to phospho-serine (pSer) and phospho-threonine (pThr) [245]. Within advanced MS instrumentation, fragmentation of enriched peptides using approaches such as collision-induced dissociation (CID) or high-energy collision dissociation (HCD), can facilitate PTM identification. Phosphorylated peptides produce a characteristic neutral loss of the phosphate group, a prime example of this proteomic approach [246]. A similar antibody-based enrichment strategy can be used for the study of protein ubiquitination in human skeletal muscles, by utilizing antibodies that recognize a two glycine (GG) remnant on ubiquitinated peptides [247]. An overview of routine approaches used for the PTM analysis of skeletal muscle proteins is shown in Figure 4.
The quantification of PTMs can be performed using label-free approaches or label-based methods, with SILAC and TMT being routinely used. The phospho-proteomic analysis of human skeletal muscle quantified 8511 unique phosphorylation sites, with approximately 12% regulated due to physical exercise using iTRAQ or TMT combined with phospho-peptide enrichment [248]. A recent application of PTM analysis of human skeletal muscle proteins has focused on the modulatory effects of high-intensity interval training on contractile fibers [249]. The proteomic analysis of vastus lateralis muscle biopsies collected before and after training revealed distinct changes in the lysine-acetylome, an important PTM with respect to regulating process such as muscle metabolism, myofiber growth and muscle contraction [249].
For lower throughput PTM analyses, as compared to relatively swift MS-based approaches, immunoblotting is an appropriate technique facilitating the identification and quantification of PTMs by employing antibodies that specifically recognize modified protein targets. While this experimental technique is well validated, the generation of high-quality antibodies to PTMs is still demanding [250]. Protein sample probing using antibodies that are specific for both the modified antigen and its unmodified counterpart allows the level of PTMs to be evaluated for comparing specific sets of specimens. For example, phosphorylation in human skeletal muscle was successfully determined using antibody screening to study the effects of resistance exercise with a focus on type II myofibers [251]. The phosphorylation statuses of AMP-activated kinase (AMPK), eukaryotic initiation factor 4E-binding protein (4E-BP1), p70/p85-S6 protein kinase (S6K1) and ribosomal S6 protein (S6) could be determined using phospho-specific versus pan antibodies when comparing skeletal muscle biopsies before and immediately after exercise [251]. The pan class of antibodies recognizes a protein in its phosphorylated or unphosphorylated state, since it binds to an epitope that is distinct from the dynamically phosphorylated site. The availability of both phosphorylation-specific and pan antibodies can be exploited in enzyme-linked immunosorbent assays (ELISAs). Sandwich-based ELISAs employ two antibodies to detect phosphorylated proteins, with one antibody-binding total protein while the second antibody recognizes the phosphorylation site specifically. The results from these type of ELISAs can be quantitative when the phosphorylated protein of interest is available in purified form for standard curve generation. Changes in protein phosphorylation can then be used to investigate pre-selected proteoforms of physiological or pathophysiological importance. Notably, multiplex detection of phosphorylated proteins is feasible using sandwich-based antibody arrays. This methodological approach has recently been applied to study skeletal muscle wasting and protein turnover in pancreatic cancer cachexia [252].

3. Skeletal Muscles—The Proteomics Perspective

Muscle tissues account for approximately 40% of body mass, making contractile fibers one of the most abundant cell types in humans. Thus, cellular adaptations, biochemical changes, metabolic fluctuations or dysfunction of muscle fibers can exert a considerable influence on whole-body physiology [253].

3.1. Basic and Applied Myology

The human body contains approximately 650 individual skeletal muscles [254], which are involved in a large number of diverse biological functions, including (i) excitation-contraction-relaxation cycles to enable coordinated locomotion for body movements, (ii) postural maintenance of supportive structures to control muscle-skeletal balance and bodily protection, (iii) verbal and facial communication via the oral and facial musculature, (iv) respiration with the help of the diaphragm muscle, (v) regulation of body thermogenesis, (vi) provision of a protein reservoir for periods of starvation and (vii) metabolic and bioenergetic integration [255]. Figure 5 gives an overview of the extent of proteome-wide adaptations due to changed functional demands or pathophysiological alterations in the skeletal musculature.
The skeletal musculature is a highly adaptable organ system [42] that can be affected by changes in lifestyle, physical activity levels, side effects seen in traumatic injury, extensive pharmacotherapy and the natural aging process [38,46]. Acute or chronic myofiber degradation, inflammation and/or myofibrosis are involved in many primary muscle-associated diseases, such as muscular dystrophies [44,256], but also occur in association with various common pathologies, such as cancer, diabetes, obesity, heart disease or sepsis. Striking cellular changes in the skeletal musculature are observed during embryonic development, regenerative adult myogenesis, in response to different forms of physical exercise, disuse-related muscular atrophy, muscle fiber type specification, muscle transitions, primary neuromuscular disorders, muscle co-morbidities and sarcopenia of old age. Thus, a detailed understanding of normal muscle function and its cellular regulation including the dynamics of the skeletal muscle proteome, as compared to muscle wasting, is important for diverse biological and biomedical disciplines. This includes crucial aspects of lifestyle biology, health science and clinical medicine, including sports and exercise physiology [39,40], electro-stimulation therapy, research into skeletal muscle plasticity [43,257], disuse-related muscular atrophy and the effects of re-innervation [41,258,259], space flight medicine [260,261], neuromuscular diseases [44,45], frailty-associated muscle aging and biogerontology [47,262,263,264], forensic medicine [265,266] and the meat industry [267,268]. Important topics in muscle cell biology that are gaining steady interest are the regulation of skeletal muscle mass [269], myokine signaling [270] and muscle-bone-fat crosstalk [271]. Figure 6 summarizes important cell biological characteristics of skeletal muscles, including histological, physiological and biochemical properties of the two main types of contractile fibers, i.e., slow-twitching type I myofibers and predominantly fast-twitching type II myofibers [253,254].
In the adult human skeletal musculature, a mixture of slow oxidative type I, fast oxidative-glycolytic type IIa and fast glycolytic type IIx/d myofibers, plus hybrid fibers ranging from I/IIa to IIa/IIx, are found in most individual muscles [238,272]. Interestingly, in small rodents, the fiber type distribution patterns are more complex as compared to human voluntary muscles. Rodent muscles contain additional and extremely fast-twitching myofibers of the type IIb [273]. Excellent markers of the various subtypes of myofibers are represented by slow myosin heavy chain MyHC-1(beta) and the fast myosin heavy chains MyHC-2a, MyHC-2x and MyHC-2b [274]. The biochemical profiling of the contractile apparatus of skeletal muscles has recently been reviewed and this article lists the most abundant fiber type-specific markers, i.e., isoforms of myosin heavy chains, myosin light chains, troponins and tropomyosins, that were confirmed by proteomics [47]. The identification of individual fiber types using contractile apparatus proteins can be conveniently combined with reliable markers that belong to the Ca2+-handling complexes of skeletal muscles. This includes the slow SERCA2 and fast SERCA1 isoforms of the sarcoplasmic reticulum Ca2+-ATPase, and the slow CSQ2 and fast CSQ1 isoforms of the luminal Ca2+-binding protein calsequestrin (Figure 7).
The traditional determination of fiber type specification using relatively crude histological, histochemical or immuno-histological labelling techniques [226,227,275,276] has been superseded by high-throughput proteomic procedures that now focus on the biochemical profiling of distinct myosin proteoforms in single myofibers. This advanced approach for fiber type screening employs a proteomic analysis platform with a capillary liquid chromatography-MS gradient using a 96-well format [277]. This superb improvement of myofiber screening, named Proteomics-high-throughput-Fiber-Typing (ProFiT), shows the enormous impact MS has had already on skeletal muscle cell biology.

3.2. Proteomic Complexity in Skeletal Muscles

Within the various layers of biological organization in multi-cellular organisms, the relationship and flow of information from gene to RNA/mRNA to protein/proteoform to the cytome and functional physiome is highly complex, as outlined in Figure 8 for skeletal muscles. The underlying mechanisms that produce a considerably higher number of proteins per individual gene from the approximately 22,000 genes in the human genome are due to alternative promoter usage and alternative post-transcriptional RNA splicing mechanisms, as well as dynamic PTMs such as proteolytic cleavage of a protein product [54,55]. The combined involvedness of variations at the level of DNA, RNA and protein modifications are the underlying evolutionary drive that has produced a human proteome of astonishing complexity [278,279]. Especially high levels of alternative splicing contributes to the generation of proteomic diversity [280], which is also clearly the case in skeletal muscle tissues [281]. MS-based analysis can be instrumental to evaluate this considerable influence of RNA splicing on protein expression level in skeletal muscle fibers [23,24]. In humans, approximately 90% of transcripts of protein-coding genes have been estimated to undergo alternative splicing [278].
Some genes that contain protein-coding sequences for the production of muscle-specific components, such as the extremely large X-chromosomal DMD gene that encodes the Dp427-M isoform of the membrane cytoskeletal protein named dystrophin, have more than one promoter [282] (Figure 9). Thus, several protein products with tissue-specific expression patterns can derive from the sequence information that is stored in one individual gene. The production of precursor messenger RNA and alternative RNA splicing play a crucial part in the post-transcriptional regulation of gene expression. For example, the sarcoplasmic reticulum Ca2+-ATPase exists in the form of several protein isoforms with developmental and fiber type-specific expression patterns which derive through alternative RNA splicing from two genes [283] (Figure 9). Extensive PTMs are key mechanisms that produce distinct proteoforms, the functional protein units of cellular biological processes. An example is the dystroglycan complex which is part of the larger dystrophin complexome at the muscle fiber periphery [284]. The alpha/beta-dystroglycan complex is produced through proteolytic processing from one protein product that is encoded by the DAG1 gene [285,286]. The two protein products are biochemically very different protein species; one is a typical transmembrane-spanning glycoprotein of the sarcolemma, beta-dystroglycan, and the other is an extracellular laminin receptor located in the basal lamina, alpha-dystroglycan (Figure 9).

3.3. Proteomic Profiling of Skeletal Muscles

The main bioanalytical avenues taken in the proteomic characterization of skeletal muscles include top-down proteomics focusing on the characterization of intact proteoforms, bottom-up proteomics, which is a peptide-centric method concerned with the large-scale detection of proteins in complex mixtures, and subproteomics that examines the protein composition of distinct subcellular fractions [20,37]. MS-based studies have decisively improved our general cell biological understanding of protein diversity and the heterogeneous composition of individual fibers in skeletal muscles [36]. This detailed proteomic knowledge that was accumulated over the last two decades can now be integrated with findings from other omics-type methodologies, such as genomics, transcriptomics, lipidomics, metabolomics and cytomics, to establish a systems biological view of skeletal muscle function. Figure 10 gives an overview of the bioanalytically accessible parts of the highly dynamic skeletal muscle proteome and the main subcellular structures of myofibers.

3.3.1. The Status Quo of the Skeletal Muscle Proteome

Detailed information on the MS-based coverage of individual muscle proteins can be retrieved from multi-consensus files, which are usually provided as supplemental tables in the above- and below-quoted papers of major proteomic studies with a focus on skeletal muscles, or can be accessed from proteomic depositories and international databanks, such as ProteomeXchange (PX) [287] (http://www.proteomexchange.org). Currently, these detailed listings of MS-generated hits sum up to over 10,000 individual proteins that form the core proteome of skeletal muscles.
The continued expansion of the total number of identified muscle-associated proteins and the increased extent of established proteoform diversity has led to detailed proteomic maps of different types of skeletal muscles [25,26,27,28,288,289,290], including predominantly fast-twitching versus slow-twitching muscles [29,30,31,291,292,293,294]. Reliable fiber type-specific markers include fast versus slow isoforms of contractile proteins, such as myosin heavy chains, myosin light chains, troponins and tropomyosins, and Ca2+-regulatory proteins, including Ca2+-pumps, Ca2+-binding proteins and Ca2+-channels.
Established proteomic markers of adult fiber types [33,34,35,36] are:
  • Myosin-7 (MyHC-I) for slow type I myofibers;
  • Myosin-2 (MyHC-IIa) for fast type IIa myofibers;
  • Myosin-1 (MyHC-IIx/d) for fast type IIx/d myofibers;
  • Myosin-4 (MyHC-IIb) for extremely fast type IIb myofibers.
Frequently used markers of the contractile apparatus and its auxiliary filaments within the sarcomeric structure [25,26,27,290] include:
  • Myosin light chain MLC1/3 for the thick myosin-containing filament;
  • Alpha-actin ACTA for the thin actin-containing filament;
  • Tropomyosin isoform TPM2 for the tropomyosin complex;
  • Troponin subunit TnC for the troponin complex;
  • Titin for the sarcomere-spanning titin filament;
  • Myomesin MYOM-1 for the sarcomeric M-band;
  • Alpha-actinin ACTN2 for the Z-disk complex.

3.3.2. The Proteome of Specialized Cells and Structures within Skeletal Muscles

The proteome of intrafusal muscle spindles, which run anatomically in parallel to bulk extrafusal myofibers, are part of the proprioceptive system of the body that is intrinsically involved in controlling skeletal muscle length, coordinated movements and posture, was recently analyzed [295]. As compared to skeletal muscle fibers, the proprioceptive muscle system was shown to express higher levels of myosins-3/6/7/7b, collagen isoform COL-IV and sensory neuron proteins PIEZO2 and SLC17A7. Proteomic profiling was also employed to study the detailed molecular architecture of the neuromuscular junction with markers such as the nicotinic acetylcholine receptor, agrin, utrophin and the enzyme acetylcholinesterase [296,297]. Of crucial importance for the swift regeneration of damaged muscle fibers are myogenic stem cells, which are represented by the satellite cell population that is located between the sarcolemma and basal lamina in adult skeletal muscles. Satellite cell and activated muscle precursor cell proteomics has recently been carried out to identify biomarkers of fiber damage [298] and muscle aging [299].

3.3.3. The Subproteome of Skeletal Muscles

In addition to cataloguing surveys of total skeletal muscle extracts, subcellular studies have identified a large number of organellar-specific proteoforms in skeletal muscles [300,301]. Subproteomics is an important subdiscipline of protein biochemistry that focuses on the identification and characterization of proteins that specifically associate with particular organelles using MS-based proteomics [82,302,303]. Subproteomic studies of skeletal muscle preparations were directed towards the surveying of proteins that are highly enriched in the sarcolemma [92], sarcoplasmic reticulum [175,304], mitochondrion-sarcoplasmic reticulum linker complexes [305], mitochondria [306,307,308] and the sarcosol [309], as well as the class of giant muscle proteins, such as dystrophin, nebulin, obscurin, plectin, the ryanodine receptor and the half-sarcomere spanning filamentous component titin [310]. Of note, the more recent application of single-cell proteomics (SCP) for studying fast versus slow myofibers [36] has revealed further molecular features of the extensive complexity of fiber type-specific protein expression patterns [33,34,35,261,277,311,312,313].
Robust skeletal muscle markers that are closely associated with distinct subcellular structures [25,92,304,305,309] include:
  • Glycolytic enzymes for the sarcosol;
  • Dystroglycan beta-DG for the sarcolemma;
  • Caveolin-1 for surface caveolae structures;
  • Dysferlin for the sarcolemmal repair apparatus;
  • Talin for costameres involved in lateral force transmission;
  • Laminin for the basal lamina;
  • Collagen isoforms for the various layers of the extracellular matrix, including the epimysium, endomysium and perimysium;
  • Dystrophin isoform Dp427-M for the sub-plasmalemmal membrane cytoskeleton;
  • Alpha-1S subunit of the L-type Ca2+- channel for the transverse tubules;
  • SERCA-type Ca2+-ATPases for the longitudinal tubules of the sarcoplasmic reticulum;
  • Calsequestrin for the terminal cisternae region of the luminal sarcoplasmic reticulum;
  • Sarcalumenin for the lumen of the longitudinal tubules of the sarcoplasmic reticulum;
  • Ryanodine receptor Ca2+-release channel isoform RyR1 for the triad junction contact sites between transverse tubules and terminal cisternae of the sarcoplasmic reticulum;
  • 40S ribosomal protein SA for ribosomes;
  • Vesicular transport factor for the Golgi apparatus;
  • Lysosome-associated membrane glycoprotein 1 for lysosomes;
  • Catalase for peroxisomes;
  • Ubiquitin-conjugating enzyme E2 for proteasomes;
  • Emerin for the myonucleus.
Mitochondria are essential for the extensive bioenergetic needs of multi-cellular systems, including highly specialized skeletal muscles [314] and have been extensively characterized by proteomics [315,316].
In skeletal muscles, excellent mitochondrial marker proteins [301,306,308] include:
  • Voltage-dependent anion-selective channel protein VDAC1 for the mitochondrial outer membrane;
  • Glutathione transferase for dynamic contact sites between the mitochondrial inner and outer membranes;
  • Adenylate kinase isoform AK2 for the mitochondrial intermembrane space;
  • NADH dehydrogenase for the mitochondrial inner membrane complex I;
  • Succinate dehydrogenase for the mitochondrial inner membrane complex II;
  • Cytochrome b-c complex for the mitochondrial inner membrane complex III;
  • Cytochrome c oxidase for the mitochondrial inner membrane complex IV;
  • ATP synthase for the mitochondrial inner membrane complex V;
  • Isocitrate dehydrogenase for the mitochondrial matrix.

3.3.4. Major Classes of Proteins in Skeletal Muscles

Major protein classes of skeletal muscles that are routinely detected by proteomics, as judged by bioinformatic PANTHER analysis [317], include DNA metabolism protein, RNA metabolism protein, calcium-binding protein, cell adhesion molecule, cell junction protein, chaperone, chromatin/chromatin-binding or -regulatory protein, cytoskeletal protein, defense/immunity protein, extracellular matrix protein, gene-specific transcriptional regulator, intercellular signal molecule, membrane traffic protein, metabolite interconversion enzyme, protein modifying enzyme, protein-binding activity modulator, scaffold/adaptor protein, storage protein, structural protein, transfer/carrier protein, translational protein, transmembrane signal receptor and transporter [290].
Crucial intracellular fractions of myofibers, the cytoskeletal network, metabolite transportation and the abundant extracellular matrisome of skeletal muscle tissues have been covered by proteomic analysis [25,26,27,28,290] and have confirmed the suitability of the following protein markers:
  • Phosphofructokinase for the sarcosolic glycolytic pathway;
  • Fatty acid-binding proteins for metabolite transportation in the cytosol;
  • AlphaB-crystallin for the cellular stress response machinery;
  • Desmin for intermediate filaments;
  • Tubulins for microtubules;
  • Myoglobin for intracellular oxygen transportation;
  • Hemoglobin for external oxygen supply;
  • Serum albumin for osmotic balancing in the extracellular space;
  • Laminin-211 for the stabilization of the basal lamina;
  • Collagens, such as isoform COL-VI, for the extracellular matrix;
  • Periostin for the matricellular protein network;
  • Decorin for the proteoglycan matrix.

3.3.5. The Proteomic Profile of the Skeletal Muscle Secretome

Importantly, proteomics was applied to studying the muscle secretome, which can give crucial indications on the status of muscular changes in response to myogenesis, regeneration, disuse, neuromuscular disease, strenuous exercise, bioenergetic stress or aging [318,319,320,321].
Major muscle secretome components were identified as:
  • Matrisomal proteins: perlecan, biglycan, decorin, alpha-dystroglycan, fibronectin, laminin, mimecan, nidogen, periostin, prolargin, matrix metalloproteinases and a large number of collagen isoforms;
  • Cytokines and growth factors: angiopoietin, bone morphogenic protein, chemokines, cell adhesion molecules, complement factors, connective tissue growth factor, fibroblast growth factor, myostatin, insulin growth factor, transforming growth factor, tumor necrosis factor, and vascular endothelial growth factor;
  • Essential enzymes: major glycolytic enzymes such as aldolase, glyceraldehyde-3-phosphate dehydrogenase, triosephosphate isomerase and pyruvate kinase, alcohol dehydrogenase, aldose reductase, mitochondrial ATP synthase, creatine kinase, superoxide dismutase, glycogen phosphorylase and protein disulfide isomerase;
  • Enzyme inhibitors: cystatin, metalloproteinase inhibitors, various serpin isoforms and macroglobulin;
  • Major contractile proteins: myosin heavy chains, myosin light chains, myosin-binding proteins, actin, titin (fragments), troponins and tropomyosins;
  • Sarcomeric and non-sarcomeric cytoskeletal proteins: actin, F-actin capping proteins, alpha-actinin, cofilin, desmin, ezrin, filamin, myomesin, plectin, vimentin and vinculin.
Other muscle secretome proteins that are routinely detected by proteomics [321,322,323] include markers such as:
  • Agrin for the neuromuscular junction;
  • Annexins for the cellular repair mechanism;
  • Heat shock proteins of the subclasses HspA, HspB and HspC, and endoplasmin, for the class of molecular chaperones involved in the cellular stress response;
  • Calsequestrin, calmodulin and calreticulin for the Ca2+-handling apparatus;
  • Carbonic anhydrase isoform CA3 for the regulation of carbon dioxide metabolism;
  • Fatty acid-binding protein FABP3 for metabolite transportation.
In the context of biomarker research, the systematic cataloguing of the skeletal muscle secretome has established that many muscle proteins can be detected in suitable biofluids such as serum/plasma, urine or saliva for diagnostic purposes [324]. Muscle proteins that are associated with the intracellular fractions of myofibers can passively leak or be actively released into the systemic circulation due to strenuous exercise or traumatic muscle damage and then detected in biofluids. In addition to the above-listed proteomic markers CA3 and FABP3 [325], a variety of general muscle-derived serum markers are routinely assayed in single measurements or in combination. Muscle creatine kinase is the most frequently employed serum marker for the differential diagnosis of muscular abnormalities [45]. Other abundant and routinely used muscle damage markers include alanine amino transferase, aldolase, aspartate amino transferase, enolase, hydroxybutyrate dehydrogenase, lactate dehydrogenase, myosin light chain, troponin subunit TnI and myoglobin [326]. Although the 10 enzymes of glycolysis are present in almost all cell types in the body at high density, they nevertheless represent extremely abundant and distinct protein isoforms in the sarcosol of myofibers [309,327]. This makes specific muscle proteoforms of glycolytic enzymes, that can be routinely detected by advanced and high-throughput MS analysis, good candidates for establishing muscle-derived biomarkers of myofiber damage. This new array of markers for improved myofiber screening in health and disease exemplifies the vast impact proteomics has had on the cell biological characterization of skeletal muscle tissues over the last two decades of myology research.

3.3.6. Progress of Cataloguing the Skeletal Muscle Proteome

A list of major publications that have focused on various aspects of skeletal muscle proteomics is provided in Table 1. These select papers include both primary research articles and key review articles outlining crucial findings in the field of basic and applied myology. The proteomic information summarizes the methodological approaches taken to establish the core protein components of skeletal muscles. The proteomic studies and reviews listed include (i) the initiation of the skeletal muscle proteome project [21,22], (ii) the systematic cataloguing of human skeletal muscles [20,23,26,27,37,53], (iii) proteomic surveys of fast versus slow human muscles [29,292], (iv) the analysis of human single myofibers [35,36,313], (v) the systematic cataloguing of animal skeletal muscles [25,28,288,290,328,329,330], (vi) the differential proteomic surveys of fast versus slow mouse muscles [30,31,32], (vii) profiling of animal single myofibers [25,33,277,311,312], (viii) the mass spectrometric analysis of muscle spindles [295], (ix) the profiling of the neuromuscular junction [296,297], (x) proteomic profiling of subcellular fractions (sarcolemma, sarcoplasmic reticulum, mitochondria, sarcosol, giant muscle protein assemblies, contractile apparatus) [92,140,175,304,305,306,308,309,310], (xi) the systematic cataloguing of the muscle secretome [318,319,321], (xii) profiling of the effects of exercise on the skeletal muscle proteome [38,39,40,289,331,332], (xiii) proteomic analysis of muscular disuse atrophy [261,333,334,335,336], (xiv) profiling of muscle plasticity [43,257] and (xv) the proteomic screening of skeletal muscle aging [34,130,135,264,291,337,338,339,340,341,342,343,344].
The coverage of the entire set of muscle-associated proteoforms is currently limited by the technically achievable enrichment and separation of dynamic protein populations prior to MS-based analysis. Future advances in single-cell preparations [12,166,188] and more efficient protein isolation during both top-down and bottom-up proteomics, in combination with further progress in the sensitivity of peptide/protein mass spectrometry [167,194,215,216,217,218,219,345], will be instrumental for the comprehensive cataloguing of the entire skeletal muscle proteome.

4. Conclusions

MS-based proteomics has been successfully applied to the comprehensive profiling of skeletal muscles. The usage of both top-down and bottom-up proteomic approaches has improved our cell biological knowledge of muscle protein diversity and the considerably heterogeneous composition of fast- versus slow-twitching fibers in skeletal muscles. Major classes of muscle-associated proteins that were identified by proteomics, and frequently exist as fiber type-specific isoforms, include components that are located in diverse cellular structures, including (i) thin actin filaments of the I-band, (ii) thick myosin filaments of the A-band, (iii) auxiliary titin and nebulin filaments, (iv) sarcomeric M-line complexes, (v) sarcomeric Z-disk complexes, (vi) the non-sarcomeric cytoskeleton, (vii) metabolic enzymes and transporters of anaerobic energy metabolism, (viii) metabolic enzymes and transporters of oxidative energy metabolism, (ix) cellular signaling complexes, (x) membranous, cytosolic and luminal ion-handling complexes, (xi) excitation-contraction coupling apparatus, (xii) molecular chaperoning system, (xiii) satellite cells and activated myogenic precursors, (xiv) myokines and fiber-derived factors of the muscle secretome, (xv) elaborate layers of the extracellular matrix consisting of the basal lamina, endomysium, perimysium and epimysium, (xvi) muscle-associated capillaries and (xvii) innervating motor neurons and insulating glial Schwann cells forming functional motor units. In addition, major organellar markers of skeletal muscles have been identified and characterized by proteomics, including proteins located in the nucleus, sarcoplasmic reticulum, Golgi apparatus, ribosomes, lysosomes, peroxisomes, proteasomes, transverse tubules, sarcolemma and mitochondria. This detailed proteomic knowledge can now be used to establish a systems biological view of skeletal muscle function.

Author Contributions

Conceptualization, P.D., D.S. and K.O.; writing—original draft preparation, P.D. and K.O.; writing—review and editing, P.D., D.S. and K.O. All authors have read and agreed to the published version of the manuscript.

Funding

This manuscript was supported by the Kathleen Lonsdale Institute for Human Health Research at Maynooth University, the Irish Research Council (Ulysses/2024/5) and Science Foundation Ireland Infrastructure Award SFI-12/RI/2346/3.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors wish to thank Paula Meleady and Michael Henry from Dublin City University for their continued support of our skeletal muscle proteomics project.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AEAffinity enrichment
APAcetone precipitation
ACTAAlpha-actin
ACTNAlpha-actinin
BACBenzyldime-thyl-n-hexadecylammonium chloride
BAC-DROPBAC-gel dissolution to digest PAGE-resolved objective proteins
BNBlue native
CACarbonic anhydrase
CIDCollision-induced dissociation
CoFGEComparative two-dimensional fluorescence gel electrophoresis
COLCollagen
CSQCalsequestrin
DCDifferential centrifugation
DDAData-dependent acquisition
DGDystroglycan
DIAData-independent acquisition
DIGEDifference gel electrophoresis
ELISAEnzyme-linked immunosorbent assays
ESIElectrospray ionization
ETDElectron transfer dissociation
FABPFatty acid-binding proteins
FASPFilter-aided sample preparation
FAIMSHigh-field asymmetric ion mobility spectrometry
FTMSFourier-transform ion cyclotron resonance mass spectrometry
GEGel electrophoresis
GeLCGel electrophoresis-liquid chromatography
HCDHigh-energy collision dissociation
ICATIsotope-coded affinity tags
ICPLIsotope-coded protein labelling
IEFIsoelectric focusing
IMACImmobilized metal affinity chromatography
iSTIn-StageTip
iTRAQIsobaric tags for relative and absolute quantitation
LCLiquid chromatography
LFQLabel-free quantification
MALDIMatrix-assisted laser desorption/ionization
MLCMyosin light chain
MSMass spectrometry
MS/MSTandem mass spectrometry
MudPITMultidimensional protein identification technology
MWMolecular weight
MyHCMyosin heavy chain
MYOMMyomesin
m/zMass-to-charge ratio
nMSNative mass spectrometry
PAGEPolyacrylamide gel electrophoresis
PASEFParallel accumulation serial fragmentation
PCTPressure cycling technology
PEAProximity extension assays
pIIsoelectric point
PRMParallel reaction monitoring
ProFiTProteomics-high-throughput-Fiber-Typing
pSCoPEPrioritized single-cell proteomics
PTMPost-translational modification
RTSReal-time search
RyRRyanodine receptor
SDSSodium dodecyl sulfate
SERCASarcoplasmic or endoplasmic reticulum calcium ATPase
SILACStable isotope labelling by amino acids in cell culture
SOMASlow Off-Rate Modified Aptamers
SP3Single-pot solid-phase-enhanced sample preparation
SPSSynchronous precursor selection
SRSarcoplasmic reticulum
SRM/MRMSelected/Multiple Reaction Monitoring
S-TrapSuspension trapping
SWATH-MSSequential window acquisition of all theoretical mass spectra
TDATargeted data acquisition
TIMSTrapped ion mobility spectrometry
TMTTandem mass tag
TnTroponin
ToFTime-of-flight
TPMTropomyosin
UCUltracentrifugation
USP3Universal solid-phase protein preparation
VDACVoltage-dependent anion-selective channel protein
XL-MSCross-linking mass spectrometry

References

  1. Bludau, I.; Aebersold, R. Proteomic and interactomic insights into the molecular basis of cell functional diversity. Nat. Rev. Mol. Cell Biol. 2020, 21, 327–340. [Google Scholar] [CrossRef]
  2. Walther, T.C.; Mann, M. Mass spectrometry-based proteomics in cell biology. J. Cell Biol. 2010, 190, 491–500. [Google Scholar] [CrossRef]
  3. Jung, E.; Heller, M.; Sanchez, J.C.; Hochstrasser, D.F. Proteomics meets cell biology: The establishment of subcellular proteomes. Electrophoresis 2000, 21, 3369–3377. [Google Scholar] [CrossRef] [PubMed]
  4. Wheeler, C.H.; Berry, S.L.; Wilkins, M.R.; Corbett, J.M.; Ou, K.; Gooley, A.A.; Humphery-Smith, I.; Williams, K.L.; Dunn, M.J. Characterisation of proteins from two-dimensional electrophoresis gels by matrix-assisted laser desorption mass spectrometry and amino acid compositional analysis. Electrophoresis 1996, 17, 580–587. [Google Scholar] [CrossRef]
  5. Wilkins, M.R.; Sanchez, J.C.; Gooley, A.A.; Appel, R.D.; Humphery-Smith, I.; Hochstrasser, D.F.; Williams, K.L. Progress with proteome projects: Why all proteins expressed by a genome should be identified and how to do it. Biotechnol. Genet. Eng. Rev. 1996, 13, 19–50. [Google Scholar] [CrossRef] [PubMed]
  6. Carbonara, K.; Andonovski, M.; Coorssen, J.R. Proteomes Are of Proteoforms: Embracing the Complexity. Proteomes 2021, 9, 38. [Google Scholar] [CrossRef] [PubMed]
  7. Smith, L.M.; Kelleher, N.L.; Consortium for Top Down Proteomics. Proteoform: A single term describing protein complexity. Nat. Methods 2013, 10, 186–187. [Google Scholar] [CrossRef]
  8. Forgrave, L.M.; Wang, M.; Yang, D.; DeMarco, M.L. Proteoforms and their expanding role in laboratory medicine. Pract. Lab Med. 2021, 28, e00260. [Google Scholar] [CrossRef] [PubMed]
  9. Hollas, M.A.R.; Robey, M.T.; Fellers, R.T.; LeDuc, R.D.; Thomas, P.M.; Kelleher, N.L. The Human Proteoform Atlas: A FAIR community resource for experimentally derived proteoforms. Nucleic Acids Res. 2022, 50, D526–D533. [Google Scholar] [CrossRef]
  10. Aebersold, R.; Mann, M. Mass-spectrometric exploration of proteome structure and function. Nature 2016, 537, 347–355. [Google Scholar] [CrossRef]
  11. Vistain, L.F.; Tay, S. Single-Cell Proteomics. Trends Biochem. Sci. 2021, 46, 661–672. [Google Scholar] [CrossRef]
  12. Mund, A.; Brunner, A.D.; Mann, M. Unbiased spatial proteomics with single-cell resolution in tissues. Mol. Cell 2022, 82, 2335–2349. [Google Scholar] [CrossRef] [PubMed]
  13. Lermyte, F.; Tsybin, Y.O.; O’Connor, P.B.; Loo, J.A. Top or Middle? Up or Down? Toward a Standard Lexicon for Protein Top-Down and Allied Mass Spectrometry Approaches. J. Am. Soc. Mass Spectrom. 2019, 30, 1149–1157. [Google Scholar] [CrossRef] [PubMed]
  14. Manes, N.P.; Nita-Lazar, A. Application of targeted mass spectrometry in bottom-up proteomics for systems biology research. J. Proteom. 2018, 189, 75–90. [Google Scholar] [CrossRef]
  15. Miller, R.M.; Smith, L.M. Overview and considerations in bottom-up proteomics. Analyst 2023, 148, 475–486. [Google Scholar] [CrossRef]
  16. Habeck, T.; Lermyte, F. Seeing the complete picture: Proteins in top-down mass spectrometry. Essays Biochem. 2023, 67, 283–300. [Google Scholar] [CrossRef] [PubMed]
  17. Melby, J.A.; Roberts, D.S.; Larson, E.J.; Brown, K.A.; Bayne, E.F.; Jin, S.; Ge, Y. Novel Strategies to Address the Challenges in Top-Down Proteomics. J. Am. Soc. Mass Spectrom. 2021, 32, 1278–1294. [Google Scholar] [CrossRef]
  18. Ercan, H.; Resch, U.; Hsu, F.; Mitulovic, G.; Bileck, A.; Gerner, C.; Yang, J.W.; Geiger, M.; Miller, I.; Zellner, M. A Practical and Analytical Comparative Study of Gel-Based Top-Down and Gel-Free Bottom-Up Proteomics Including Unbiased Proteoform Detection. Cells 2023, 12, 747. [Google Scholar] [CrossRef]
  19. Schaffer, L.V.; Millikin, R.J.; Shortreed, M.R.; Scalf, M.; Smith, L.M. Improving Proteoform Identifications in Complex Systems Through Integration of Bottom-Up and Top-Down Data. J. Proteome Res. 2020, 19, 3510–3517. [Google Scholar] [CrossRef]
  20. Gonzalez-Freire, M.; Semba, R.D.; Ubaida-Mohien, C.; Fabbri, E.; Scalzo, P.; Højlund, K.; Dufresne, C.; Lyashkov, A.; Ferrucci, L. The Human Skeletal Muscle Proteome Project: A reappraisal of the current literature. J. Cachexia Sarcopenia Muscle 2017, 8, 5–18. [Google Scholar] [CrossRef] [PubMed]
  21. Sanchez, J.C.; Chiappe, D.; Converset, V.; Hoogland, C.; Binz, P.A.; Paesano, S.; Appel, R.D.; Wang, S.; Sennitt, M.; Nolan, A.; et al. The mouse SWISS-2D PAGE database: A tool for proteomics study of diabetes and obesity. Proteomics 2001, 1, 136–163. [Google Scholar] [CrossRef] [PubMed]
  22. Yan, J.X.; Harry, R.A.; Wait, R.; Welson, S.Y.; Emery, P.W.; Preedy, V.R.; Dunn, M.J. Separation and identification of rat skeletal muscle proteins using two-dimensional gel electrophoresis and mass spectrometry. Proteomics 2001, 1, 424–434. [Google Scholar] [CrossRef]
  23. Gelfi, C.; Vasso, M.; Cerretelli, P. Diversity of human skeletal muscle in health and disease: Contribution of proteomics. J. Proteom. 2011, 74, 774–795. [Google Scholar] [CrossRef]
  24. Ohlendieck, K. Skeletal muscle proteomics: Current approaches, technical challenges and emerging techniques. Skelet. Muscle 2011, 1, 6. [Google Scholar] [CrossRef]
  25. Deshmukh, A.S.; Murgia, M.; Nagaraj, N.; Treebak, J.T.; Cox, J.; Mann, M. Deep proteomics of mouse skeletal muscle enables quantitation of protein isoforms, metabolic pathways, and transcription factors. Mol. Cell. Proteom. 2015, 14, 841–853. [Google Scholar] [CrossRef]
  26. Højlund, K.; Yi, Z.; Hwang, H.; Bowen, B.; Lefort, N.; Flynn, C.R.; Langlais, P.; Weintraub, S.T.; Mandarino, L.J. Characterization of the human skeletal muscle proteome by one-dimensional gel electrophoresis and HPLC-ESI-MS/MS. Mol. Cell. Proteom. 2008, 7, 257–267. [Google Scholar] [CrossRef]
  27. Parker, K.C.; Walsh, R.J.; Salajegheh, M.; Amato, A.A.; Krastins, B.; Sarracino, D.A.; Greenberg, S.A. Characterization of human skeletal muscle biopsy samples using shotgun proteomics. J. Proteome Res. 2009, 8, 3265–3277. [Google Scholar] [CrossRef]
  28. Raddatz, K.; Albrecht, D.; Hochgräfe, F.; Hecker, M.; Gotthardt, M. A proteome map of murine heart and skeletal muscle. Proteomics 2008, 8, 1885–1897. [Google Scholar] [CrossRef]
  29. Capitanio, D.; Viganò, A.; Ricci, E.; Cerretelli, P.; Wait, R.; Gelfi, C. Comparison of protein expression in human deltoideus and vastus lateralis muscles using two-dimensional gel electrophoresis. Proteomics 2005, 5, 2577–2586. [Google Scholar] [CrossRef]
  30. Drexler, H.C.; Ruhs, A.; Konzer, A.; Mendler, L.; Bruckskotten, M.; Looso, M.; Günther, S.; Boettger, T.; Krüger, M.; Braun, T. On marathons and Sprints: An integrated quantitative proteomics and transcriptomics analysis of differences between slow and fast muscle fibers. Mol. Cell. Proteom. 2012, 11, M111.010801. [Google Scholar] [CrossRef]
  31. Gelfi, C.; Viganò, A.; De Palma, S.; Ripamonti, M.; Begum, S.; Cerretelli, P.; Wait, R. 2-D protein maps of rat gastrocnemius and soleus muscles: A tool for muscle plasticity assessment. Proteomics 2006, 6, 321–340. [Google Scholar] [CrossRef] [PubMed]
  32. Okumura, N.; Hashida-Okumura, A.; Kita, K.; Matsubae, M.; Matsubara, T.; Takao, T.; Nagai, K. Proteomic analysis of slow- and fast-twitch skeletal muscles. Proteomics 2005, 5, 2896–2906. [Google Scholar] [CrossRef]
  33. Eggers, B.; Schork, K.; Turewicz, M.; Barkovits, K.; Eisenacher, M.; Schröder, R.; Clemen, C.S.; Marcus, K. Advanced fiber type- specific protein profiles derived from adult murine skeletal muscle. Proteomes 2021, 9, 28. [Google Scholar] [CrossRef] [PubMed]
  34. Murgia, M.; Toniolo, L.; Nagaraj, N.; Ciciliot, S.; Vindigni, V.; Schiaffino, S.; Reggiani, C.; Mann, M. Single Muscle Fiber Proteomics Reveals Fiber-Type-Specific Features of Human Muscle Aging. Cell Rep. 2017, 19, 2396–2409. [Google Scholar] [CrossRef]
  35. Murgia, M.; Nogara, L.; Baraldo, M.; Reggiani, C.; Mann, M.; Schiaffino, S. Protein profile of fiber types in human skeletal muscle: A single-fiber proteomics study. Skelet. Muscle 2021, 11, 24. [Google Scholar] [CrossRef]
  36. Schiaffino, S.; Reggiani, C.; Murgia, M. Fiber type diversity in skeletal muscle explored by mass spectrometry-based single fiber proteomics. Histol. Histopathol. 2020, 35, 239–246. [Google Scholar] [CrossRef]
  37. Capitanio, D.; Moriggi, M.; Gelfi, C. Mapping the human skeletal muscle proteome: Progress and potential. Expert Rev. Proteom. 2017, 14, 825–839. [Google Scholar] [CrossRef] [PubMed]
  38. Cervone, D.T.; Moreno-Justicia, R.; Quesada, J.P.; Deshmukh, A.S. Mass spectrometry-based proteomics approaches to interrogate skeletal muscle adaptations to exercise. Scand. J. Med. Sci. Sports 2023. advance online publication. [Google Scholar] [CrossRef] [PubMed]
  39. Hesketh, S.J.; Stansfield, B.N.; Stead, C.A.; Burniston, J.G. The application of proteomics in muscle exercise physiology. Expert Rev. Proteom. 2020, 17, 813–825. [Google Scholar] [CrossRef]
  40. Petriz, B.A.; Gomes, C.P.; Almeida, J.A.; de Oliveira, G.P., Jr.; Ribeiro, F.M.; Pereira, R.W.; Franco, O.L. The Effects of Acute and Chronic Exercise on Skeletal Muscle Proteome. J. Cell. Physiol. 2017, 232, 257–269. [Google Scholar] [CrossRef] [PubMed]
  41. Cho, Y.; Ross, R.S. A mini review: Proteomics approaches to understand disused vs. exercised human skeletal muscle. Physiol. Genom. 2018, 50, 746–757. [Google Scholar] [CrossRef] [PubMed]
  42. Flueck, M. Plasticity of the muscle proteome to exercise at altitude. High Alt. Med. Biol. 2009, 10, 183–193. [Google Scholar] [CrossRef]
  43. Ohlendieck, K. Proteomic profiling of skeletal muscle plasticity. Muscles Ligaments Tendons J. 2012, 1, 119–126. [Google Scholar] [PubMed]
  44. Dowling, P.; Murphy, S.; Ohlendieck, K. Proteomic profiling of muscle fibre type shifting in neuromuscular diseases. Expert Rev. Proteom. 2016, 13, 783–799. [Google Scholar] [CrossRef] [PubMed]
  45. Ohlendieck, K. Proteomic identification of biomarkers of skeletal muscle disorders. Biomark. Med. 2013, 7, 169–186. [Google Scholar] [CrossRef]
  46. Danese, E.; Montagnana, M.; Lippi, G. Proteomics and frailty: A clinical overview. Expert Rev. Proteom. 2018, 15, 657–664. [Google Scholar] [CrossRef] [PubMed]
  47. Dowling, P.; Gargan, S.; Swandulla, D.; Ohlendieck, K. Fiber-Type Shifting in Sarcopenia of Old Age: Proteomic Profiling of the Contractile Apparatus of Skeletal Muscles. Int. J. Mol. Sci. 2023, 24, 2415. [Google Scholar] [CrossRef]
  48. Hochstrasser, D.F. Proteome in perspective. Clin. Chem. Lab. Med. 1998, 36, 825–836. [Google Scholar] [CrossRef]
  49. Adhikari, S.; Nice, E.C.; Deutsch, E.W.; Lane, L.; Omenn, G.S.; Pennington, S.R.; Paik, Y.K.; Overall, C.M.; Corrales, F.J.; Cristea, I.M.; et al. A high-stringency blueprint of the human proteome. Nat. Commun. 2020, 11, 5301. [Google Scholar] [CrossRef]
  50. Kim, M.S.; Pinto, S.M.; Getnet, D.; Nirujogi, R.S.; Manda, S.S.; Chaerkady, R.; Madugundu, A.K.; Kelkar, D.S.; Isserlin, R.; Jain, S.; et al. A draft map of the human proteome. Nature 2014, 509, 575–581. [Google Scholar] [CrossRef] [PubMed]
  51. Wang, D.; Eraslan, B.; Wieland, T.; Hallström, B.; Hopf, T.; Zolg, D.P.; Zecha, J.; Asplund, A.; Li, L.H.; Meng, C.; et al. A deep proteome and transcriptome abundance atlas of 29 healthy human tissues. Mol. Syst. Biol. 2019, 15, e8503. [Google Scholar] [CrossRef]
  52. Wilhelm, M.; Schlegl, J.; Hahne, H.; Gholami, A.M.; Lieberenz, M.; Savitski, M.M.; Ziegler, E.; Butzmann, L.; Gessulat, S.; Marx, H.; et al. Mass-spectrometry-based draft of the human proteome. Nature 2014, 509, 582. [Google Scholar] [CrossRef] [PubMed]
  53. Jiang, L.; Wang, M.; Lin, S.; Jian, R.; Li, X.; Chan, J.; Dong, G.; Fang, H.; Robinson, A.E.; GTEx Consortium; et al. A Quantitative Proteome Map of the Human Body. Cell 2020, 183, 269–283. [Google Scholar] [CrossRef] [PubMed]
  54. Omenn, G.S.; Lane, L.; Overall, C.M.; Cristea, I.M.; Corrales, F.J.; Lindskog, C.; Paik, Y.K.; Van Eyk, J.E.; Liu, S.; Pennington, S.R.; et al. Research on the Human Proteome Reaches a Major Milestone: >90% of Predicted Human Proteins Now Credibly Detected, According to the HUPO Human Proteome Project. J. Proteome Res. 2020, 19, 4735–4746. [Google Scholar] [CrossRef] [PubMed]
  55. Overall, C.M. The Human Proteome: 90% in the Light, 10% on the Dark Side. J. Proteome Res. 2020, 19, 4731–4734. [Google Scholar] [CrossRef]
  56. Dang, X.; Scotcher, J.; Wu, S.; Chu, R.K.; Tolić, N.; Ntai, I.; Thomas, P.M.; Fellers, R.T.; Early, B.P.; Zheng, Y.; et al. The first pilot project of the consortium for top-down proteomics: A status report. Proteomics 2014, 14, 1130–1140. [Google Scholar] [CrossRef] [PubMed]
  57. Schaffer, L.V.; Millikin, R.J.; Miller, R.M.; Anderson, L.C.; Fellers, R.T.; Ge, Y.; Kelleher, N.L.; LeDuc, R.D.; Liu, X.; Payne, S.H.; et al. Identification and Quantification of Proteoforms by Mass Spectrometry. Proteomics 2019, 19, e1800361. [Google Scholar] [CrossRef] [PubMed]
  58. Su, T.; Hollas, M.A.R.; Fellers, R.T.; Kelleher, N.L. Identification of Splice Variants and Isoforms in Transcriptomics and Proteomics. Annu. Rev. Biomed. Data Sci. 2023, 6, 357–376. [Google Scholar] [CrossRef]
  59. Schaffer, L.V.; Shortreed, M.R.; Cesnik, A.J.; Frey, B.L.; Solntsev, S.K.; Scalf, M.; Smith, L.M. Expanding Proteoform Identifications in Top-Down Proteomic Analyses by Constructing Proteoform Families. Anal. Chem. 2018, 90, 1325–1333. [Google Scholar] [CrossRef] [PubMed]
  60. Smith, L.M.; Agar, J.N.; Chamot-Rooke, J.; Danis, P.O.; Ge, Y.; Loo, J.A.; Paša-Tolić, L.; Tsybin, Y.O.; Kelleher, N.L.; The Consortium for Top-Down Proteomics. The Human Proteoform Project: Defining the human proteome. Sci. Adv. 2021, 7, eabk0734. [Google Scholar] [CrossRef]
  61. Drown, B.S.; Jooß, K.; Melani, R.D.; Lloyd-Jones, C.; Camarillo, J.M.; Kelleher, N.L. Mapping the Proteoform Landscape of Five Human Tissues. J. Proteome Res. 2022, 21, 1299–1310. [Google Scholar] [CrossRef]
  62. LeDuc, R.D.; Deutsch, E.W.; Binz, P.A.; Fellers, R.T.; Cesnik, A.J.; Klein, J.A.; Van Den Bossche, T.; Gabriels, R.; Yalavarthi, A.; Perez-Riverol, Y.; et al. Proteomics Standards Initiative’s ProForma 2.0: Unifying the Encoding of Proteoforms and Peptidoforms. J. Proteome Res. 2022, 21, 1189–1195. [Google Scholar] [CrossRef]
  63. Bader, J.M.; Albrecht, V.; Mann, M. MS-Based Proteomics of Body Fluids: The End of the Beginning. Mol. Cell. Proteom. 2023, 22, 100577. [Google Scholar] [CrossRef] [PubMed]
  64. Mendes, M.L.; Dittmar, G. Targeted proteomics on its way to discovery. Proteomics 2022, 22, e2100330. [Google Scholar] [CrossRef] [PubMed]
  65. Mann, S.P.; Treit, P.V.; Geyer, P.E.; Omenn, G.S.; Mann, M. Ethical Principles, Constraints and Opportunities in Clinical Proteomics. Mol. Cell. Proteom. 2021, 20, 100046. [Google Scholar] [CrossRef]
  66. Moore, J.L.; Patterson, N.H.; Norris, J.L.; Caprioli, R.M. Prospective on Imaging Mass Spectrometry in Clinical Diagnostics. Mol. Cell. Proteom. 2023, 22, 100576. [Google Scholar] [CrossRef]
  67. Meissner, F.; Geddes-McAlister, J.; Mann, M.; Bantscheff, M. The emerging role of mass spectrometry-based proteomics in drug discovery. Nat. Rev. Drug Discov. 2022, 21, 637–654. [Google Scholar] [CrossRef]
  68. Poulos, R.C.; Cai, Z.; Robinson, P.J.; Reddel, R.R.; Zhong, Q. Opportunities for pharmacoproteomics in biomarker discovery. Proteomics 2023, 23, e2200031. [Google Scholar] [CrossRef] [PubMed]
  69. Sadee, W.; Wang, D.; Hartmann, K.; Toland, A.E. Pharmacogenomics: Driving Personalized Medicine. Pharmacol. Rev. 2023, 75, 789–814. [Google Scholar] [CrossRef]
  70. Ang, M.Y.; Low, T.Y.; Lee, P.Y.; Wan Mohamad Nazarie, W.F.; Guryev, V.; Jamal, R. Proteogenomics: From next-generation sequencing (NGS) and mass spectrometry-based proteomics to precision medicine. Clin. Chim. Acta. 2019, 498, 38–46. [Google Scholar] [CrossRef]
  71. Low, T.Y.; Mohtar, M.A.; Ang, M.Y.; Jamal, R. Connecting Proteomics to Next-Generation Sequencing: Proteogenomics and Its Current Applications in Biology. Proteomics 2019, 19, e1800235. [Google Scholar] [CrossRef] [PubMed]
  72. Behnke, J.S.; Urner, L.H. Emergence of mass spectrometry detergents for membrane proteomics. Anal. Bioanal. Chem. 2023, 415, 3897–3909. [Google Scholar] [CrossRef] [PubMed]
  73. Danko, K.; Lukasheva, E.; Zhukov, V.A.; Zgoda, V.; Frolov, A. Detergent-Assisted Protein Digestion-On the Way to Avoid the Key Bottleneck of Shotgun Bottom-Up Proteomics. Int. J. Mol. Sci. 2022, 23, 13903. [Google Scholar] [CrossRef]
  74. Dowling, P.; Gargan, S.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Extracellular Matrix Proteomics: The mdx-4cv Mouse Diaphragm as a Surrogate for Studying Myofibrosis in Dystrophinopathy. Biomolecules 2023, 13, 1108. [Google Scholar] [CrossRef] [PubMed]
  75. Naba, A. Ten Years of Extracellular Matrix Proteomics: Accomplishments, Challenges, and Future Perspectives. Mol. Cell. Proteom. 2023, 22, 100528. [Google Scholar] [CrossRef]
  76. Reid, D.J.; Thibert, S.; Zhou, M. Dissecting the structural heterogeneity of proteins by native mass spectrometry. Protein Sci. 2023, 32, e4612. [Google Scholar] [CrossRef]
  77. van Schaick, G.; Haselberg, R.; Somsen, G.W.; Wuhrer, M.; Domínguez-Vega, E. Studying protein structure and function by native separation-mass spectrometry. Nat. Rev. Chem. 2022, 6, 215–231. [Google Scholar] [CrossRef] [PubMed]
  78. Wong, K.F.C.; Greatorex, R.E.; Gidman, C.E.; Rahman, S.; Griffiths, R.L. Surface-sampling mass spectrometry to study proteins and protein complexes. Essays Biochem. 2023, 67, 229–241. [Google Scholar] [CrossRef]
  79. Le Sueur, C.; Hammarén, H.M.; Sridharan, S.; Savitski, M.M. Thermal proteome profiling: Insights into protein modifications, associations, and functions. Curr. Opin. Chem. Biol. 2022, 71, 102225. [Google Scholar] [CrossRef]
  80. Piersimoni, L.; Kastritis, P.L.; Arlt, C.; Sinz, A. Cross-Linking Mass Spectrometry for Investigating Protein Conformations and Protein-Protein Interactions-A Method for All Seasons. Chem. Rev. 2022, 122, 7500–7531. [Google Scholar] [CrossRef]
  81. Tang, X.; Wippel, H.H.; Chavez, J.D.; Bruce, J.E. Crosslinking mass spectrometry: A link between structural biology and systems biology. Protein Sci. 2021, 30, 773–784. [Google Scholar] [CrossRef]
  82. Drissi, R.; Dubois, M.L.; Boisvert, F.M. Proteomics methods for subcellular proteome analysis. FEBS J. 2013, 280, 5626–5634. [Google Scholar] [CrossRef] [PubMed]
  83. Christopher, J.A.; Geladaki, A.; Dawson, C.S.; Vennard, O.L.; Lilley, K.S. Subcellular Transcriptomics and Proteomics: A Comparative Methods Review. Mol. Cell. Proteom. 2022, 21, 100186. [Google Scholar] [CrossRef]
  84. Graziadei, A.; Rappsilber, J. Leveraging crosslinking mass spectrometry in structural and cell biology. Structure 2022, 30, 37–54. [Google Scholar] [CrossRef] [PubMed]
  85. Westphall, M.S.; Lee, K.W.; Salome, A.Z.; Coon, J.J.; Grant, T. Mass spectrometers as cryoEM grid preparation instruments. Curr. Opin. Struct. Biol. 2023, 83, 102699. [Google Scholar] [CrossRef] [PubMed]
  86. Rajczewski, A.T.; Jagtap, P.D.; Griffin, T.J. An overview of technologies for MS-based proteomics-centric multi-omics. Expert Rev. Proteom. 2022, 19, 165–181. [Google Scholar] [CrossRef] [PubMed]
  87. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  88. Timp, W.; Timp, G. Beyond mass spectrometry, the next step in proteomics. Sci. Adv. 2020, 6, eaax8978. [Google Scholar] [CrossRef]
  89. Jackson, C.A.; Vogel, C. New horizons in the stormy sea of multimodal single-cell data integration. Mol. Cell. 2022, 82, 248–259. [Google Scholar] [CrossRef]
  90. Gargan, S.; Ohlendieck, K. Sample Preparation and Protein Determination for 2D-DIGE Proteomics. Methods Mol. Biol. 2023, 2596, 325–337. [Google Scholar] [CrossRef]
  91. Murphy, S.; Ohlendieck, K. Mass spectrometric identification of dystrophin, the protein product of the Duchenne muscular dystrophy gene, in distinct muscle surface membranes. Int. J. Mol. Med. 2017, 40, 1078–1088. [Google Scholar] [CrossRef]
  92. Murphy, S.; Zweyer, M.; Henry, M.; Meleady, P.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Proteomic analysis of the sarcolemma-enriched fraction from dystrophic mdx-4cv skeletal muscle. J. Proteom. 2019, 191, 212–227. [Google Scholar] [CrossRef] [PubMed]
  93. van Westering, T.L.E.; Johansson, H.J.; Hanson, B.; Coenen-Stass, A.M.L.; Lomonosova, Y.; Tanihata, J.; Motohashi, N.; Yokota, T.; Takeda, S.; Lehtiö, J.; et al. Mutation-independent Proteomic Signatures of Pathological Progression in Murine Models of Duchenne Muscular Dystrophy. Mol. Cell. Biochem. 2020, 19, 2047–2068. [Google Scholar] [CrossRef] [PubMed]
  94. Murphy, S.; Brinkmeier, H.; Krautwald, M.; Henry, M.; Meleady, P.; Ohlendieck, K. Proteomic profiling of the dystrophin complex and membrane fraction from dystrophic mdx muscle reveals decreases in the cytolinker desmoglein and increases in the extracellular matrix stabilizers biglycan and fibronectin. J. Muscle Res. Cell. Motil. 2017, 38, 251–268. [Google Scholar] [CrossRef]
  95. Capitanio, D.; Moriggi, M.; Torretta, E.; Barbacini, P.; De Palma, S.; Viganò, A.; Lochmüller, H.; Muntoni, F.; Ferlini, A.; Mora, M.; et al. Comparative proteomic analyses of Duchenne muscular dystrophy and Becker muscular dystrophy muscles: Changes contributing to preserve muscle function in Becker muscular dystrophy patients. J. Cachexia Sarcopenia Muscle 2020, 11, 547–563. [Google Scholar] [CrossRef]
  96. Matsumura, C.Y.; Menezes de Oliveira, B.; Durbeej, M.; Marques, M.J. Isobaric Tagging-Based Quantification for Proteomic Analysis: A Comparative Study of Spared and Affected Muscles from mdx Mice at the Early Phase of Dystrophy. PLoS ONE 2013, 8, e65831. [Google Scholar] [CrossRef] [PubMed]
  97. Woodland, B.; Necakov, A.; Coorssen, J.R. Optimized Proteome Reduction for Integrative Top-Down Proteomics. Proteomes 2023, 11, 10. [Google Scholar] [CrossRef] [PubMed]
  98. Marcus, K.; Lelong, C.; Rabilloud, T. What Room for Two-Dimensional Gel-Based Proteomics in a Shotgun Proteomics World? Proteomes 2020, 8, 17. [Google Scholar] [CrossRef] [PubMed]
  99. Westermeier, R. 2D gel-based Proteomics: There’s life in the old dog yet. Arch. Physiol. Biochem. 2016, 122, 236–237. [Google Scholar] [CrossRef]
  100. 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]
  101. Murphy, S.; Dowling, P.; Ohlendieck, K. Comparative Skeletal Muscle Proteomics Using Two-Dimensional Gel Electrophoresis. Proteomes 2016, 4, 27. [Google Scholar] [CrossRef] [PubMed]
  102. Oliveira, B.M.; Coorssen, J.R.; Martins-de-Souza, D. 2DE: The phoenix of proteomics. J. Proteom. 2014, 104, 140–150. [Google Scholar] [CrossRef]
  103. Rabilloud, T.; Chevallet, M.; Luche, S.; Lelong, C. Two-dimensional gel electrophoresis in proteomics: Past, present and future. J. Proteom. 2010, 73, 2064–2077. [Google Scholar] [CrossRef] [PubMed]
  104. Lee, P.Y.; Saraygord-Afshari, N.; Low, T.Y. The evolution of two-dimensional gel electrophoresis—From proteomics to emerging alternative applications. J. Chromatogr. A 2020, 1615, 460763. [Google Scholar] [CrossRef] [PubMed]
  105. Rabilloud, T.; Lelong, C. Two-dimensional gel electrophoresis in proteomics: A tutorial. J. Proteom. 2011, 74, 1829–1841. [Google Scholar] [CrossRef]
  106. Westermeier, R. Looking at proteins from two dimensions: A review on five decades of 2D electrophoresis. Arch. Physiol. Biochem. 2014, 120, 168–172. [Google Scholar] [CrossRef] [PubMed]
  107. Carbonara, K.; Coorssen, J.R. Sometimes faster can be better: Microneedling IPG strips enables higher throughput for integrative top-down proteomics. Proteomics 2023, 23, e2200307. [Google Scholar] [CrossRef] [PubMed]
  108. Carrette, O.; Burkhard, P.R.; Sanchez, J.C.; Hochstrasser, D.F. State-of-the-art two-dimensional gel electrophoresis: A key tool of proteomics research. Nat. Protoc. 2006, 1, 812–823. [Google Scholar] [CrossRef]
  109. Friedman, D.B.; Hoving, S.; Westermeier, R. Isoelectric focusing and two-dimensional gel electrophoresis. Methods Enzymol. 2009, 463, 515–540. [Google Scholar] [CrossRef]
  110. Görg, A.; Weiss, W.; Dunn, M.J. Current two-dimensional electrophoresis technology for proteomics. Proteomics 2004, 4, 3665–3685. [Google Scholar] [CrossRef]
  111. Yoneten, K.K.; Kasap, M.; Akpinar, G.; Kanli, A.; Karaoz, E. Comparative Proteomics Analysis of Four Commonly Used Methods for Identification of Novel Plasma Membrane Proteins. J. Membr. Biol. 2019, 252, 587–608. [Google Scholar] [CrossRef]
  112. Zahedi, R.P.; Moebius, J.; Sickmann, A. Two-dimensional BAC/SDS-PAGE for membrane proteomics. Subcell. Biochem. 2007, 43, 13–20. [Google Scholar] [CrossRef]
  113. Wittig, I.; Braun, H.P.; Schägger, H. Blue native PAGE. Nat. Protoc. 2006, 1, 418–428. [Google Scholar] [CrossRef] [PubMed]
  114. Fernandez-Vizarra, E.; Zeviani, M. Blue-Native Electrophoresis to Study the OXPHOS Complexes. Methods Mol. Biol. 2021, 2192, 287–311. [Google Scholar] [CrossRef]
  115. Sunderhaus, S.; Eubel, H.; Braun, H.P. Two-dimensional blue native/blue native polyacrylamide gel electrophoresis for the characterization of mitochondrial protein complexes and supercomplexes. Methods Mol. Biol. 2007, 372, 315–324. [Google Scholar] [CrossRef] [PubMed]
  116. Culligan, K.; Banville, N.; Dowling, P.; Ohlendieck, K. Drastic reduction of calsequestrin-like proteins and impaired calcium binding in dystrophic mdx muscle. J. Appl. Physiol. 2002, 92, 435–445. [Google Scholar] [CrossRef] [PubMed]
  117. Froemming, G.R.; Murray, B.E.; Ohlendieck, K. Self-aggregation of triadin in the sarcoplasmic reticulum of rabbit skeletal muscle. Biochim. Biophys. Acta. 1999, 1418, 197–205. [Google Scholar] [CrossRef] [PubMed]
  118. Maguire, P.B.; Briggs, F.N.; Lennon, N.J.; Ohlendieck, K. Oligomerization is an intrinsic property of calsequestrin in normal and transformed skeletal muscle. Biochem. Biophys. Res. Commun. 1997, 240, 721–727. [Google Scholar] [CrossRef]
  119. Panfoli, I.; Calzia, D.; Santucci, L.; Ravera, S.; Bruschi, M.; Candiano, G. A blue dive: From ‘blue fingers’ to ‘blue silver’. A comparative overview of staining methods for in-gel proteomics. Expert Rev. Proteom. 2012, 9, 627–634. [Google Scholar] [CrossRef] [PubMed]
  120. Noaman, N.; Abbineni, P.S.; Withers, M.; Coorssen, J.R. Coomassie staining provides routine (sub)femtomole in-gel detection of intact proteoforms: Expanding opportunities for genuine Top-down Proteomics. Electrophoresis 2017, 38, 3086–3099. [Google Scholar] [CrossRef] [PubMed]
  121. Sundaram, P. Protein Stains and Applications. Methods Mol. Biol. 2018, 1853, 1–14. [Google Scholar] [CrossRef]
  122. Arentz, G.; Weiland, F.; Oehler, M.K.; Hoffmann, P. State of the art of 2D DIGE. Proteom. Clin. Appl. 2015, 9, 277–288. [Google Scholar] [CrossRef]
  123. Ohlendieck, K. Comparative 3-Sample 2D-DIGE Analysis of Skeletal Muscles. Methods Mol. Biol. 2023, 2596, 127–146. [Google Scholar] [CrossRef] [PubMed]
  124. Hanneken, M.; König, S. Horizontal comparative fluorescence two-dimensional gel electrophoresis for improved spot coordinate detection. Electrophoresis 2014, 35, 1118–1121. [Google Scholar] [CrossRef]
  125. Ackermann, D.; König, S. Comparative Two-Dimensional Fluorescence Gel Electrophoresis. Methods Mol. Biol. 2023, 2596, 71–81. [Google Scholar] [CrossRef]
  126. Blundon, M.; Ganesan, V.; Redler, B.; Van, P.T.; Minden, J.S. Two-Dimensional Difference Gel Electrophoresis. Methods Mol. Biol. 2019, 1855, 229–247. [Google Scholar] [CrossRef] [PubMed]
  127. Ohlendieck, K. Top-Down Proteomics and Comparative 2D-DIGE Analysis. Methods Mol. Biol. 2023, 2596, 19–38. [Google Scholar] [CrossRef] [PubMed]
  128. Dani, D.; Dencher, N.A. Native-DIGE: A new look at the mitochondrial membrane proteome. Biotechnol. J. 2008, 3, 817–822. [Google Scholar] [CrossRef] [PubMed]
  129. Dani, D.; Dencher, N.A. Native DIGE for Quantitative and Functional Analysis of Protein Interactomes. Methods Mol. Biol. 2023, 2596, 53–69. [Google Scholar] [CrossRef]
  130. Gannon, J.; Staunton, L.; O’Connell, K.; Doran, P.; Ohlendieck, K. Phosphoproteomic analysis of aged skeletal muscle. Int. J. Mol. Med. 2008, 22, 33–42. [Google Scholar] [CrossRef]
  131. Tokutomi, Y.; Araki, N.; Kataoka, K.; Yamamoto, E.; Kim-Mitsuyama, S. Oxidation of Prx2 and phosphorylation of GRP58 by angiotensin II in human coronary smooth muscle cells identified by 2D-DIGE analysis. Biochem. Biophys. Res. Commun. 2007, 364, 822–830. [Google Scholar] [CrossRef]
  132. Morandell, S.; Stasyk, T.; Grosstessner-Hain, K.; Roitinger, E.; Mechtler, K.; Bonn, G.K.; Huber, L.A. Phosphoproteomics strategies for the functional analysis of signal transduction. Proteomics 2006, 6, 4047–4056. [Google Scholar] [CrossRef] [PubMed]
  133. Stasyk, T.; Huber, L.A. DIGE-Based Phosphoproteomic Analysis. Methods Mol. Biol. 2023, 2596, 97–104. [Google Scholar] [CrossRef]
  134. Krishnamoorthy, V.; Donofrio, A.J.; Martin, J.L. O-GlcNAcylation of αB-crystallin regulates its stress-induced translocation and cytoprotection. Mol. Cell. Biochem. 2013, 379, 59–68. [Google Scholar] [CrossRef] [PubMed]
  135. O’Connell, K.; Doran, P.; Gannon, J.; Ohlendieck, K. Lectin-based proteomic profiling of aged skeletal muscle: Decreased pyruvate kinase isozyme M1 exhibits drastically increased levels of N-glycosylation. Eur. J. Cell Biol. 2008, 87, 793–805. [Google Scholar] [CrossRef] [PubMed]
  136. Mehta-D’souza, P. Detection of Glycoproteins in Polyacrylamide Gels Using Pro-Q Emerald 300 Dye, a Fluorescent Periodate Schiff-Base Stain. Methods Mol. Biol. 2018, 1853, 115–119. [Google Scholar] [CrossRef]
  137. Elschenbroich, S.; Ignatchenko, V.; Sharma, P.; Schmitt-Ulms, G.; Gramolini, A.O.; Kislinger, T. Peptide separations by on-line MudPIT compared to isoelectric focusing in an off-gel format: Application to a membrane-enriched fraction from C2C12 mouse skeletal muscle cells. J. Proteome Res. 2009, 8, 4860–4869. [Google Scholar] [CrossRef] [PubMed]
  138. Banks, C.A.; Kong, S.E.; Washburn, M.P. Affinity purification of protein complexes for analysis by multidimensional protein identification technology. Protein Expr. Purif. 2012, 86, 105–119. [Google Scholar] [CrossRef] [PubMed]
  139. Murphy, S.; Henry, M.; Meleady, P.; Ohlendieck, K. Utilization of dried and long-term stored polyacrylamide gels for the advanced proteomic profiling of mitochondrial contact sites from rat liver. Biol. Methods Protoc. 2018, 3, bpy008. [Google Scholar] [CrossRef] [PubMed]
  140. Murphy, S.; Ohlendieck, K. Proteomic profiling of large myofibrillar proteins from dried and long-term stored polyacrylamide gels. Anal. Biochem. 2018, 543, 8–11. [Google Scholar] [CrossRef]
  141. Chen, C.; Wen, M.; Jin, Y. 1DE-MS Profiling for Proteoform-Correlated Proteomic Analysis, by Combining SDS-PAGE, Whole-Gel Slicing, Quantitative LC-MS/MS, and Reconstruction of Gel Distributions of Several Thousands of Proteins. J. Proteome Res. 2022, 21, 2311–2330. [Google Scholar] [CrossRef]
  142. Murphy, S.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Comparative gel-based proteomic analysis of chemically crosslinked complexes in dystrophic skeletal muscle. Electrophoresis 2018, 39, 1735–1744. [Google Scholar] [CrossRef]
  143. Vit, O.; Petrak, J. Integral membrane proteins in proteomics. How to break open the black box? J. Proteom. 2017, 153, 8–20. [Google Scholar] [CrossRef]
  144. Dafun, A.S.; Marcoux, J. Structural mass spectrometry of membrane proteins. Biochim. Biophys. Acta Proteins Proteom. 2022, 1870, 140813. [Google Scholar] [CrossRef]
  145. Kar, U.K.; Simonian, M.; Whitelegge, J.P. Integral membrane proteins: Bottom-up, top-down and structural proteomics. Expert Rev. Proteom. 2017, 14, 715–723. [Google Scholar] [CrossRef] [PubMed]
  146. Trötschel, C.; Poetsch, A. Current approaches and challenges in targeted absolute quantification of membrane proteins. Proteomics 2015, 15, 915–929. [Google Scholar] [CrossRef] [PubMed]
  147. Boeri Erba, E.; Signor, L.; Petosa, C. Exploring the structure and dynamics of macromolecular complexes by native mass spectrometry. J. Proteom. 2020, 222, 103799. [Google Scholar] [CrossRef] [PubMed]
  148. Santambrogio, C.; Ponzini, E.; Grandori, R. Native mass spectrometry for the investigation of protein structural (dis)order. Biochim. Biophys. Acta Proteins Proteom. 2022, 1870, 140828. [Google Scholar] [CrossRef]
  149. Tamara, S.; den Boer, M.A.; Heck, A.J.R. High-Resolution Native Mass Spectrometry. Chem. Rev. 2022, 122, 7269–7326. [Google Scholar] [CrossRef] [PubMed]
  150. Sielaff, M.; Kuharev, J.; Bohn, T.; Hahlbrock, J.; Bopp, T.; Tenzer, S.; Distler, U. Evaluation of FASP, SP3, and iST Protocols for Proteomic Sample Preparation in the Low Microgram Range. J. Proteome Res. 2017, 16, 4060–4072. [Google Scholar] [CrossRef]
  151. Wiśniewski, J.R.; Zougman, A.; Mann, M. Combination of FASP and StageTip-based fractionation allows in-depth analysis of the hippocampal membrane proteome. J. Proteome Res. 2009, 8, 5674–5678. [Google Scholar] [CrossRef] [PubMed]
  152. Duong, V.A.; Lee, H. Bottom-Up Proteomics: Advancements in Sample Preparation. Int. J. Mol. Sci. 2023, 24, 5350. [Google Scholar] [CrossRef] [PubMed]
  153. Wiśniewski, J.R.; Zougman, A.; Nagaraj, N.; Mann, M. Universal sample preparation method for proteome analysis. Nat. Methods 2009, 6, 359–362. [Google Scholar] [CrossRef] [PubMed]
  154. Brandi, J.; Noberini, R.; Bonaldi, T.; Cecconi, D. Advances in enrichment methods for mass spectrometry-based proteomics analysis of post-translational modifications. J. Chromatogr. A 2022, 1678, 463352. [Google Scholar] [CrossRef] [PubMed]
  155. Xie, Z.; Feng, Q.; Zhang, S.; Yan, Y.; Deng, C.; Ding, C.F. Advances in proteomics sample preparation and enrichment for phosphorylation and glycosylation analysis. Proteomics 2022, 22, e2200070. [Google Scholar] [CrossRef]
  156. Dowling, P.; Gargan, S.; Zweyer, M.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Protocol for the Bottom-Up Proteomic Analysis of Mouse Spleen. STAR Protoc. 2020, 1, 100196. [Google Scholar] [CrossRef] [PubMed]
  157. Woo, J.; Zhang, Q. A Streamlined High-Throughput Plasma Proteomics Platform for Clinical Proteomics with Improved Proteome Coverage, Reproducibility, and Robustness. J. Am. Soc. Mass Spectrom. 2023, 34, 754–762. [Google Scholar] [CrossRef] [PubMed]
  158. Gianazza, E.; Miller, I.; Palazzolo, L.; Parravicini, C.; Eberini, I. With or without you—Proteomics with or without major plasma/serum proteins. J. Proteom. 2016, 140, 62–80. [Google Scholar] [CrossRef] [PubMed]
  159. Lee, P.Y.; Osman, J.; Low, T.Y.; Jamal, R. Plasma/serum proteomics: Depletion strategies for reducing high-abundance proteins for biomarker discovery. Bioanalysis 2019, 11, 1799–1812. [Google Scholar] [CrossRef]
  160. Sarihan, M.; Bal Albayrak, M.G.; Kasap, M.; Akpinar, G.; Kocyigit, E. An experimental workflow for enrichment of low abundant proteins from human serum for the discovery of serum biomarkers. J. Biol. Methods. 2023, 10, e99010001. [Google Scholar] [CrossRef]
  161. Wiśniewski, J.R. Filter Aided Sample Preparation—A tutorial. Anal. Chim. Acta. 2019, 1090, 23–30. [Google Scholar] [CrossRef] [PubMed]
  162. Kulak, N.A.; Pichler, G.; Paron, I.; Nagaraj, N.; Mann, M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods 2014, 11, 319–324. [Google Scholar] [CrossRef] [PubMed]
  163. Elinger, D.; Gabashvili, A.; Levin, Y. Suspension Trapping (S-Trap) Is Compatible with Typical Protein Extraction Buffers and Detergents for Bottom-Up Proteomics. J. Proteome Res. 2019, 18, 1441–1445. [Google Scholar] [CrossRef]
  164. Hughes, C.S.; Moggridge, S.; Müller, T.; Sorensen, P.H.; Morin, G.B.; Krijgsveld, J. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat. Protoc. 2019, 14, 68–85. [Google Scholar] [CrossRef]
  165. Dagley, L.F.; Infusini, G.; Larsen, R.H.; Sandow, J.J.; Webb, A.I. Universal Solid-Phase Protein Preparation (USP3) for Bottom-up and Top-down Proteomics. J. Proteome Res. 2019, 18, 2915–2924. [Google Scholar] [CrossRef]
  166. Mansuri, M.S.; Williams, K.; Nairn, A.C. Uncovering biology by single-cell proteomics. Commun. Biol. 2023, 6, 381. [Google Scholar] [CrossRef] [PubMed]
  167. Huffman, R.G.; Leduc, A.; Wichmann, C.; Di Gioia, M.; Borriello, F.; Specht, H.; Derks, J.; Khan, S.; Khoury, L.; Emmott, E.; et al. Prioritized mass spectrometry increases the depth, sensitivity and data completeness of single-cell proteomics. Nat. Methods 2023, 20, 714–722. [Google Scholar] [CrossRef]
  168. Dapic, I.; Baljeu-Neuman, L.; Uwugiaren, N.; Kers, J.; Goodlett, D.R.; Corthals, G.L. Proteome analysis of tissues by mass spectrometry. Mass Spectrom. Rev. 2019, 38, 403–441. [Google Scholar] [CrossRef]
  169. Cai, X.; Xue, Z.; Wu, C.; Sun, R.; Qian, L.; Yue, L.; Ge, W.; Yi, X.; Liu, W.; Chen, C.; et al. High-throughput proteomic sample preparation using pressure cycling technology. Nat. Protoc. 2022, 17, 2307–2325. [Google Scholar] [CrossRef]
  170. Choksawangkarn, W.; Edwards, N.; Wang, Y.; Gutierrez, P.; Fenselau, C. Comparative study of workflows optimized for in-gel, in-solution, and on-filter proteolysis in the analysis of plasma membrane proteins. J. Proteome Res. 2012, 11, 3030–3034. [Google Scholar] [CrossRef]
  171. Glatter, T.; Ludwig, C.; Ahrné, E.; Aebersold, R.; Heck, A.J.; Schmidt, A. Large-scale quantitative assessment of different in-solution protein digestion protocols reveals superior cleavage efficiency of tandem Lys-C/trypsin proteolysis over trypsin digestion. J. Proteome Res. 2012, 11, 5145–5156. [Google Scholar] [CrossRef] [PubMed]
  172. Goodman, J.K.; Zampronio, C.G.; Jones, A.M.E.; Hernandez-Fernaud, J.R. Updates of the In-Gel Digestion Method for Protein Analysis by Mass Spectrometry. Proteomics 2018, 18, e1800236. [Google Scholar] [CrossRef] [PubMed]
  173. Gauci, V.J.; Wright, E.P.; Coorssen, J.R. Quantitative proteomics: Assessing the spectrum of in-gel protein detection methods. J. Chem. Biol. 2011, 4, 3–29. [Google Scholar] [CrossRef] [PubMed]
  174. Lewis, C.; Ohlendieck, K. Mass spectrometric identification of dystrophin isoform Dp427 by on-membrane digestion of sarcolemma from skeletal muscle. Anal. Biochem. 2010, 404, 197–203. [Google Scholar] [CrossRef]
  175. Staunton, L.; Ohlendieck, K. Mass spectrometric characterization of the sarcoplasmic reticulum from rabbit skeletal muscle by on-membrane digestion. Protein Pept. Lett. 2012, 19, 252–263. [Google Scholar] [CrossRef] [PubMed]
  176. Murphy, S.; Ohlendieck, K. Protein Digestion for 2D-DIGE Analysis. Methods Mol. Biol. 2023, 2596, 339–349. [Google Scholar] [CrossRef]
  177. Giansanti, P.; Tsiatsiani, L.; Low, T.Y.; Heck, A.J. Six alternative proteases for mass spectrometry-based proteomics beyond trypsin. Nat. Protoc. 2016, 11, 993–1006. [Google Scholar] [CrossRef] [PubMed]
  178. Tsiatsiani, L.; Heck, A.J. Proteom. beyond trypsin. FEBS J. 2015, 282, 2612–2626. [Google Scholar] [CrossRef]
  179. Zhang, X. Less is More: Membrane Protein Digestion Beyond Urea-Trypsin Solution for Next-level Proteomics. Mol. Cell. Proteom. 2015, 14, 2441–2453. [Google Scholar] [CrossRef] [PubMed]
  180. Goldman, A.R.; Beer, L.A.; Tang, H.Y.; Hembach, P.; Zayas-Bazan, D.; Speicher, D.W. Proteome Analysis Using Gel-LC-MS/MS. Curr. Protoc. Protein Sci. 2019, 96, e93. [Google Scholar] [CrossRef]
  181. Takemori, A.; Ishizaki, J.; Nakashima, K.; Shibata, T.; Kato, H.; Kodera, Y.; Suzuki, T.; Hasegawa, H.; Takemori, N. BAC-DROP: Rapid Digestion of Proteome Fractionated via Dissolvable Polyacrylamide Gel Electrophoresis and Its Application to Bottom-Up Proteomics Workflow. J. Proteome Res. 2021, 20, 1535–1543. [Google Scholar] [CrossRef] [PubMed]
  182. Domon, B.; Aebersold, R. Mass spectrometry and protein analysis. Science 2006, 312, 212–217. [Google Scholar] [CrossRef] [PubMed]
  183. Neagu, A.N.; Jayathirtha, M.; Baxter, E.; Donnelly, M.; Petre, B.A.; Darie, C.C. Applications of Tandem Mass Spectrometry (MS/MS) in Protein Analysis for Biomedical Research. Molecules 2022, 27, 2411. [Google Scholar] [CrossRef]
  184. Nesvizhskii, A.I.; Vitek, O.; Aebersold, R. Analysis and validation of proteomic data generated by tandem mass spectrometry. Nat. Methods 2007, 4, 787–797. [Google Scholar] [CrossRef] [PubMed]
  185. Yates, J.R.; Ruse, C.I.; Nakorchevsky, A. Proteomics by mass spectrometry: Approaches, advances, and applications. Annu. Rev. Biomed. Eng. 2009, 11, 49–79. [Google Scholar] [CrossRef] [PubMed]
  186. Lai, Y.H.; Wang, Y.S. Advances in high-resolution mass spectrometry techniques for analysis of high mass-to-charge ions. Mass Spectrom. Rev. 2022, 42, 2426–2445. [Google Scholar] [CrossRef] [PubMed]
  187. Yates, J.R., 3rd. Recent technical advances in proteomics. F1000Research 2019, 8, 351. [Google Scholar] [CrossRef]
  188. Tajik, M.; Baharfar, M.; Donald, W.A. Single-cell mass spectrometry. Trends Biotechnol. 2022, 40, 1374–1392. [Google Scholar] [CrossRef]
  189. Evangelista, A.J.; Ferreira, T.L. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry in the diagnosis of microorganisms. Future Microbiol. 2022, 17, 1409–1419. [Google Scholar] [CrossRef]
  190. Darie-Ion, L.; Whitham, D.; Jayathirtha, M.; Rai, Y.; Neagu, A.N.; Darie, C.C.; Petre, B.A. Applications of MALDI-MS/MS-Based Proteomics in Biomedical Research. Molecules 2022, 27, 6196. [Google Scholar] [CrossRef]
  191. Xu, X.; Jiang, X.; Shi, M.; Yin, L. Mass spectrometry-based techniques for single-cell analysis. Analyst 2023, 148, 3690–3707. [Google Scholar] [CrossRef] [PubMed]
  192. Lenčo, J.; Jadeja, S.; Naplekov, D.K.; Krokhin, O.V.; Khalikova, M.A.; Chocholouš, P.; Urban, J.; Broeckhoven, K.; Nováková, L.; Švec, F. Reversed-Phase Liquid Chromatography of Peptides for Bottom-Up Proteomics: A Tutorial. J. Proteome Res. 2022, 21, 2846–2892. [Google Scholar] [CrossRef] [PubMed]
  193. Tucholski, T.; Ge, Y. Fourier-transform ion cyclotron resonance mass spectrometry for characterizing proteoforms. Mass Spectrom. Rev. 2022, 41, 158–177. [Google Scholar] [CrossRef] [PubMed]
  194. Matzinger, M.; Mayer, R.L.; Mechtler, K. Label-free single cell proteomics utilizing ultrafast LC and MS instrumentation: A valuable complementary technique to multiplexing. Proteomics 2023, 23, e2200162. [Google Scholar] [CrossRef]
  195. Shah, A.D.; Goode, R.J.A.; Huang, C.; Powell, D.R.; Schittenhelm, R.B. LFQ-Analyst: An Easy-To-Use Interactive Web Platform To Analyze and Visualize Label-Free Proteomics Data Preprocessed with MaxQuant. J. Proteome Res. 2020, 19, 204–211. [Google Scholar] [CrossRef]
  196. Chen, X.; Sun, Y.; Zhang, T.; Shu, L.; Roepstorff, P.; Yang, F. Quantitative Proteomics Using Isobaric Labeling: A Practical Guide. Genom. Proteom. Bioinform. 2021, 19, 689–706. [Google Scholar] [CrossRef] [PubMed]
  197. Sivanich, M.K.; Gu, T.J.; Tabang, D.N.; Li, L. Recent advances in isobaric labeling and applications in quantitative proteomics. Proteomics 2022, 22, e2100256. [Google Scholar] [CrossRef]
  198. Fenaille, F.; Barbier Saint-Hilaire, P.; Rousseau, K.; Junot, C. Data acquisition workflows in liquid chromatography coupled to high resolution mass spectrometry-based metabolomics: Where do we stand? J. Chromatogr. A 2017, 1526, 1–12. [Google Scholar] [CrossRef]
  199. Kitata, R.B.; Yang, J.C.; Chen, Y.J. Advances in data-independent acquisition mass spectrometry towards comprehensive digital proteome landscape. Mass Spectrom. Rev. 2022, 42, 2324–2348. [Google Scholar] [CrossRef]
  200. Abdollahi, M.; Segura, P.A.; Beaudry, F. Is nontargeted data acquisition for target analysis (nDATA) in mass spectrometry a forward-thinking analytical approach? Biomed. Chromatogr. 2023, 37, e5531. [Google Scholar] [CrossRef]
  201. Distler, U.; Sielaff, M.; Tenzer, S. Label-Free Proteomics of Quantity-Limited Samples Using Ion Mobility-Assisted Data-Independent Acquisition Mass Spectrometry. Methods Mol. Biol. 2021, 2228, 327–339. [Google Scholar] [CrossRef]
  202. Kulyyassov, A.; Fresnais, M.; Longuespée, R. Targeted liquid chromatography-tandem mass spectrometry analysis of proteins: Basic principles, applications, and perspectives. Proteomics 2021, 21, e2100153. [Google Scholar] [CrossRef] [PubMed]
  203. Kontostathi, G.; Makridakis, M.; Bitsika, V.; Tsolakos, N.; Vlahou, A.; Zoidakis, J. Development and Validation of Multiple Reaction Monitoring (MRM) Assays for Clinical Applications. Methods Mol. Biol. 2019, 1959, 205–223. [Google Scholar] [CrossRef] [PubMed]
  204. Cho, B.G.; Gutierrez Reyes, C.D.; Goli, M.; Gautam, S.; Banazadeh, A.; Mechref, Y. Targeted N-Glycan Analysis with Parallel Reaction Monitoring Using a Quadrupole-Orbitrap Hybrid Mass Spectrometer. Anal. Chem. 2022, 94, 15215–15222. [Google Scholar] [CrossRef]
  205. Bhowmick, P.; Roome, S.; Borchers, C.H.; Goodlett, D.R.; Mohammed, Y. An Update on MRMAssayDB: A Comprehensive Resource for Targeted Proteomics Assays in the Community. J. Proteome Res. 2021, 20, 2105–2115. [Google Scholar] [CrossRef] [PubMed]
  206. Ludwig, C.; Gillet, L.; Rosenberger, G.; Amon, S.; Collins, B.C.; Aebersold, R. Data-independent acquisition-based SWATH-MS for quantitative proteomics: A tutorial. Mol. Syst. Biol. 2018, 14, e8126. [Google Scholar] [CrossRef]
  207. Sun, B.; Smialowski, P.; Aftab, W.; Schmidt, A.; Forne, I.; Straub, T.; Imhof, A. Improving SWATH-MS analysis by deep-learning. Proteomics 2023, 23, e2200179. [Google Scholar] [CrossRef]
  208. Deng, J.; Erdjument-Bromage, H.; Neubert, T.A. Quantitative Comparison of Proteomes Using SILAC. Curr. Protoc. Protein Sci. 2019, 95, e74. [Google Scholar] [CrossRef]
  209. Beller, N.C.; Hummon, A.B. Advances in stable isotope labeling: Dynamic labeling for spatial and temporal proteomic analysis. Mol. Omics 2022, 18, 579–590. [Google Scholar] [CrossRef] [PubMed]
  210. Xing, T.; Wang, C.; Zhao, X.; Dai, C.; Zhou, G.; Xu, X. Proteome Analysis Using Isobaric Tags for Relative and Absolute Analysis Quantitation (iTRAQ) Reveals Alterations in Stress-Induced Dysfunctional Chicken Muscle. J. Agric. Food Chem. 2017, 65, 2913–2922. [Google Scholar] [CrossRef]
  211. Chahrour, O.; Cobice, D.; Malone, J. Stable isotope labelling methods in mass spectrometry-based quantitative proteomics. J. Pharm. Biomed. Anal. 2015, 113, 2–20. [Google Scholar] [CrossRef]
  212. Wdowiak, A.P.; Duong, M.N.; Joyce, R.D.; Boyatzis, A.E.; Walkey, M.C.; Nealon, G.L.; Arthur, P.G.; Piggott, M.J. Isotope-Coded Maleimide Affinity Tags for Proteomics Applications. Bioconjug. Chem. 2021, 32, 1652–1666. [Google Scholar] [CrossRef] [PubMed]
  213. Lardenois, A.; Jagot, S.; Lagarrigue, M.; Guével, B.; Ledevin, M.; Larcher, T.; Dubreil, L.; Pineau, C.; Rouger, K.; Guével, L. Quantitative proteome profiling of dystrophic dog skeletal muscle reveals a stabilized muscular architecture and protection against oxidative stress after systemic delivery of MuStem cells. Proteomics 2016, 16, 2028–2042. [Google Scholar] [CrossRef] [PubMed]
  214. Rauniyar, N.; Yates, J.R., 3rd. Isobaric labeling-based relative quantification in shotgun proteomics. J. Proteome Res. 2014, 13, 5293–5309. [Google Scholar] [CrossRef]
  215. Meier, F.; Geyer, P.E.; Virreira Winter, S.; Cox, J.; Mann, M. BoxCar acquisition method enables single-shot proteomics at a depth of 10,000 proteins in 100 min. Nat. Methods 2018, 15, 440–448. [Google Scholar] [CrossRef]
  216. Furtwängler, B.; Üresin, N.; Motamedchaboki, K.; Huguet, R.; Lopez-Ferrer, D.; Zabrouskov, V.; Porse, B.T.; Schoof, E.M. Real-Time Search-Assisted Acquisition on a Tribrid Mass Spectrometer Improves Coverage in Multiplexed Single-Cell Proteomics. Mol. Cell. Proteom. 2022, 21, 100219. [Google Scholar] [CrossRef] [PubMed]
  217. Bekker-Jensen, D.B.; Martínez-Val, A.; Steigerwald, S.; Rüther, P.; Fort, K.L.; Arrey, T.N.; Harder, A.; Makarov, A.; Olsen, J.V. A Compact Quadrupole-Orbitrap Mass Spectrometer with FAIMS Interface Improves Proteome Coverage in Short LC Gradients. Mol. Cell. Proteom. 2020, 19, 716–729. [Google Scholar] [CrossRef]
  218. Krieger, J.R.; Wybenga-Groot, L.E.; Tong, J.; Bache, N.; Tsao, M.S.; Moran, M.F. Evosep One Enables Robust Deep Proteome Coverage Using Tandem Mass Tags while Significantly Reducing Instrument Time. J. Proteome Res. 2019, 18, 2346–2353. [Google Scholar] [CrossRef] [PubMed]
  219. Meier, F.; Park, M.A.; Mann, M. Trapped Ion Mobility Spectrometry and Parallel Accumulation-Serial Fragmentation in Proteomics. Mol. Cell. Proteom. 2021, 20, 100138. [Google Scholar] [CrossRef]
  220. Gold, L.; Walker, J.J.; Wilcox, S.K.; Williams, S. Advances in human proteomics at high scale with the SOMAscan proteomics platform. New Biotechnol. 2012, 29, 543–549. [Google Scholar] [CrossRef] [PubMed]
  221. Huang, J.; Chen, X.; Fu, X.; Li, Z.; Huang, Y.; Liang, C. Advances in Aptamer-Based Biomarker Discovery. Front. Cell Dev. Biol. 2021, 9, 659760. [Google Scholar] [CrossRef] [PubMed]
  222. Landsberger, M.; Brinkmeier, H. Immunoblot Analysis of DIGE-Based Proteomics. Methods Mol. Biol. 2023, 2596, 429–443. [Google Scholar] [CrossRef] [PubMed]
  223. Mishra, M.; Tiwari, S.; Gomes, A.V. Protein purification and analysis: Next generation Western blotting techniques. Expert Rev. Proteom. 2017, 14, 1037–1053. [Google Scholar] [CrossRef]
  224. Dowd, A. Elucidating Cellular Metabolism and Protein Difference Data from DIGE Proteomics Experiments Using Enzyme Assays. Methods Mol. Biol. 2023, 2596, 399–419. [Google Scholar] [CrossRef] [PubMed]
  225. Dowd, A. Enzyme Assay Methods to Validate DIGE Proteomics Data. Methods Mol. Biol. 2023, 2596, 421–428. [Google Scholar] [CrossRef]
  226. Zweyer, M.; Ohlendieck, K.; Swandulla, D. Verification of Protein Changes Determined by 2D-DIGE Based Proteomics Using Immunofluorescence Microscopy. Methods Mol. Biol. 2023, 2596, 445–464. [Google Scholar] [CrossRef]
  227. Zweyer, M.; Ohlendieck, K.; Swandulla, D. Histological and Histochemical Microscopy Used to Verify 2D-DIGE Pathoproteomics. Methods Mol. Biol. 2023, 2596, 465–480. [Google Scholar] [CrossRef]
  228. Espina, V.; Wulfkuhle, J.D.; Calvert, V.S.; VanMeter, A.; Zhou, W.; Coukos, G.; Geho, D.H.; Petricoin, E.F., 3rd; Liotta, L.A. Laser-capture microdissection. Nat. Protoc. 2006, 1, 586–603. [Google Scholar] [CrossRef]
  229. Longuespée, R.; Alberts, D.; Pottier, C.; Smargiasso, N.; Mazzucchelli, G.; Baiwir, D.; Kriegsmann, M.; Herfs, M.; Kriegsmann, J.; Delvenne, P.; et al. A laser microdissection-based workflow for FFPE tissue microproteomics: Important considerations for small sample processing. Methods 2016, 104, 154–162. [Google Scholar] [CrossRef] [PubMed]
  230. Guo, W.; Hu, Y.; Qian, J.; Zhu, L.; Cheng, J.; Liao, J.; Fan, X. Laser capture microdissection for biomedical research: Towards high-throughput, multi-omics, and single-cell resolution. J. Genet. Genom. 2023, 50, 641–651. [Google Scholar] [CrossRef]
  231. Lohani, V.; Akhiya, R.A.; Kundu, S.; Akhter, M.Q.; Bag, S. Single-Cell Proteomics with Spatial Attributes: Tools and Techniques. ACS Omega 2013, 8, 17499–17510. [Google Scholar] [CrossRef]
  232. Datta, S.; Malhotra, L.; Dickerson, R.; Chaffee, S.; Sen, C.K.; Roy, S. Laser capture microdissection: Big data from small samples. Histol. Histopathol. 2015, 30, 1255–1269. [Google Scholar] [CrossRef]
  233. Liotta, L.A.; Pappalardo, P.A.; Carpino, A.; Haymond, A.; Howard, M.; Espina, V.; Wulfkuhle, J.; Petricoin, E. Laser Capture Proteomics: Spatial tissue molecular profiling from the bench to personalized medicine. Expert Rev. Proteom. 2021, 18, 845–861. [Google Scholar] [CrossRef] [PubMed]
  234. Mao, Y.; Wang, X.; Huang, P.; Tian, R. Spatial proteomics for understanding the tissue microenvironment. Analyst 2021, 146, 3777–3798. [Google Scholar] [CrossRef]
  235. Alexovič, M.; Sabo, J.; Longuespée, R. Microproteomic sample preparation. Proteomics 2021, 21, e2000318. [Google Scholar] [CrossRef]
  236. Maerkens, A.; Olivé, M.; Schreiner, A.; Feldkirchner, S.; Schessl, J.; Uszkoreit, J.; Barkovits, K.; Güttsches, A.K.; Theis, V.; Eisenacher, M.; et al. New insights into the protein aggregation pathology in myotilinopathy by combined proteomic and immunolocalization analyses. Acta Neuropathol. Commun. 2016, 4, 8. [Google Scholar] [CrossRef]
  237. Demonbreun, A.R.; Lapidos, K.A.; Heretis, K.; Levin, S.; Dale, R.; Pytel, P.; Svensson, E.C.; McNally, E.M. Myoferlin regulation by NFAT in muscle injury, regeneration and repair. J. Cell Sci. 2010, 123, 2413–2422. [Google Scholar] [CrossRef] [PubMed]
  238. Stuart, C.A.; Stone, W.L.; Howell, M.E.; Brannon, M.F.; Hall, H.K.; Gibson, A.L.; Stone, M.H. Myosin content of individual human muscle fibers isolated by laser capture microdissection. Am. J. Physiol. Cell Physiol. 2016, 310, C381–C389. [Google Scholar] [CrossRef] [PubMed]
  239. Can, T.; Faas, L.; Ashford, D.A.; Dowle, A.; Thomas, J.; O’Toole, P.; Blanco, G. Proteomic analysis of laser capture microscopy purified myotendinous junction regions from muscle sections. Proteome Sci. 2014, 12, 25. [Google Scholar] [CrossRef] [PubMed]
  240. Keenan, E.K.; Zachman, D.K.; Hirschey, M.D. Discovering the landscape of protein modifications. Mol. Cell 2021, 81, 1868–1878. [Google Scholar] [CrossRef] [PubMed]
  241. Nishi, H.; Shaytan, A.; Panchenko, A.R. Physicochemical mechanisms of protein regulation by phosphorylation. Front. Genet. 2014, 5, 270. [Google Scholar] [CrossRef]
  242. Mann, M.; Jensen, O.N. Proteomic analysis of post-translational modifications. Nat. Biotechnol. 2003, 21, 255–261. [Google Scholar] [CrossRef]
  243. Ruprecht, B.; Koch, H.; Domasinska, P.; Frejno, M.; Kuster, B.; Lemeer, S. Optimized Enrichment of Phosphoproteomes by Fe-IMAC Column Chromatography. Methods Mol. Biol. 2017, 1550, 47–60. [Google Scholar] [CrossRef] [PubMed]
  244. Lyons, S.P.; Wilson, R.J.; Muoio, D.M.; Grimsrud, P.A. Proteomics and phosphoproteomics datasets of a muscle-specific STIM1 loss-of-function mouse model. Data Brief 2022, 42, 108051. [Google Scholar] [CrossRef] [PubMed]
  245. Hunter, T. The Croonian Lecture 1997. The phosphorylation of proteins on tyrosine: Its role in cell growth and disease. Philos. Trans. R. Soc. Lond. B Biol. Sci. 1998, 353, 583–605. [Google Scholar] [CrossRef] [PubMed]
  246. 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]
  247. Parker, B.L.; Kiens, B.; Wojtaszewski, J.F.P.; Richter, E.A.; James, D.E. Quantification of exercise-regulated ubiquitin signaling in human skeletal muscle identifies protein modification cross talk via NEDDylation. FASEB J. 2020, 34, 5906–5916. [Google Scholar] [CrossRef] [PubMed]
  248. Hoffman, N.J.; Parker, B.L.; Chaudhuri, R.; Fisher-Wellman, K.H.; Kleinert, M.; Humphrey, S.J.; Yang, P.; Holliday, M.; Trefely, S.; Fazakerley, D.J.; et al. Global Phosphoproteomic Analysis of Human Skeletal Muscle Reveals a Network of Exercise-Regulated Kinases and AMPK Substrates. Cell Metab. 2015, 22, 922–935. [Google Scholar] [CrossRef]
  249. Hostrup, M.; Lemminger, A.K.; Stocks, B.; Gonzalez-Franquesa, A.; Larsen, J.K.; Quesada, J.P.; Thomassen, M.; Weinert, B.T.; Bangsbo, J.; Deshmukh, A.S. High-intensity interval training remodels the proteome and acetylome of human skeletal muscle. eLife 2022, 11, e69802. [Google Scholar] [CrossRef]
  250. Hattori, T.; Koide, S. Next-generation antibodies for post-translational modifications. Curr. Opin. Struct. Biol. 2018, 51, 141–148. [Google Scholar] [CrossRef] [PubMed]
  251. Koopman, R.; Zorenc, A.H.; Gransier, R.J.; Cameron-Smith, D.; van Loon, L.J. Increase in S6K1 phosphorylation in human skeletal muscle following resistance exercise occurs mainly in type II muscle fibers. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E1245–E1252. [Google Scholar] [CrossRef] [PubMed]
  252. Mulder, S.E.; Dasgupta, A.; King, R.J.; Abrego, J.; Attri, K.S.; Murthy, D.; Shukla, S.K.; Singh, P.K. JNK signaling contributes to skeletal muscle wasting and protein turnover in pancreatic cancer cachexia. Cancer Lett. 2020, 491, 70–77. [Google Scholar] [CrossRef] [PubMed]
  253. Seaborne, R.A.E.; Ochala, J. The dawn of the functional genomics era in muscle physiology. J. Physiol. 2023, 601, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  254. Brooks, S.V.; Guzman, S.D.; Ruiz, L.P. Skeletal muscle structure, physiology, and function. Handb. Clin. Neurol. 2023, 195, 3–16. [Google Scholar] [CrossRef]
  255. Mukund, K.; Subramaniam, S. Skeletal muscle: A review of molecular structure and function, in health and disease. Wiley Interdiscip. Rev. Syst. Biol. Med. 2020, 12, e1462. [Google Scholar] [CrossRef] [PubMed]
  256. Ciciliot, S.; Rossi, A.C.; Dyar, K.A.; Blaauw, B.; Schiaffino, S. Muscle type and fiber type specificity in muscle wasting. Int. J. Biochem. Cell Biol. 2013, 45, 2191–2199. [Google Scholar] [CrossRef]
  257. Donoghue, P.; Doran, P.; Wynne, K.; Pedersen, K.; Dunn, M.J.; Ohlendieck, K. Proteomic profiling of chronic low-frequency stimulated fast muscle. Proteomics 2007, 7, 3417–3430. [Google Scholar] [CrossRef]
  258. Doering, T.M.; Thompson, J.M.; Budiono, B.P.; MacKenzie-Shalders, K.L.; Zaw, T.; Ashton, K.J.; Coffey, V.G. The muscle proteome reflects changes in mitochondrial function, cellular stress and proteolysis after 14 days of unilateral lower limb immobilization in active young men. PLoS ONE 2022, 17, e0273925. [Google Scholar] [CrossRef]
  259. Li, H.; Yuan, W.; Chen, Y.; Lin, B.; Wang, S.; Deng, Z.; Zheng, Q.; Li, Q. Transcription and proteome changes involved in re-innervation muscle following nerve crush in rats. BMC Genom. 2022, 23, 666. [Google Scholar] [CrossRef] [PubMed]
  260. Blottner, D.; Moriggi, M.; Trautmann, G.; Hastermann, M.; Capitanio, D.; Torretta, E.; Block, K.; Rittweger, J.; Limper, U.; Gelfi, C.; et al. Space Omics and Tissue Response in Astronaut Skeletal Muscle after Short and Long Duration Missions. Int. J. Mol. Sci. 2023, 24, 4095. [Google Scholar] [CrossRef]
  261. Murgia, M.; Ciciliot, S.; Nagaraj, N.; Reggiani, C.; Schiaffino, S.; Franchi, M.V.; Pišot, R.; Šimunič, B.; Toniolo, L.; Blaauw, B.; et al. Signatures of muscle disuse in spaceflight and bed rest revealed by single muscle fiber proteomics. PNAS Nexus 2022, 1, pgac086. [Google Scholar] [CrossRef]
  262. Deane, C.S.; Phillips, B.E.; Willis, C.R.G.; Wilkinson, D.J.; Smith, K.; Higashitani, N.; Williams, J.P.; Szewczyk, N.J.; Atherton, P.J.; Higashitani, A.; et al. Proteomic features of skeletal muscle adaptation to resistance exercise training as a function of age. Geroscience 2023, 45, 1271–1287. [Google Scholar] [CrossRef] [PubMed]
  263. Roberts, M.; Ruple, B.; Godwin, J.; McIntosh, M.; Chen, S.Y.; Kontos, N.; Agyin-Birikorang, A.; Michel, J.M.; Plotkin, D.; Mattingly, M.; et al. A novel deep proteomic approach in human skeletal muscle unveils distinct molecular signatures affected by aging and resistance training. bioRxiv 2023. [Google Scholar] [CrossRef]
  264. Théron, L.; Gueugneau, M.; Coudy, C.; Viala, D.; Bijlsma, A.; Butler-Browne, G.; Maier, A.; Béchet, D.; Chambon, C. Label-free quantitative protein profiling of vastus lateralis muscle during human aging. Mol. Cell. Proteom. 2014, 13, 283–294. [Google Scholar] [CrossRef] [PubMed]
  265. Battistini, A.; Capitanio, D.; Bailo, P.; Moriggi, M.; Tambuzzi, S.; Gelfi, C.; Piccinini, A. Proteomic analysis by mass spectrometry of postmortem muscle protein degradation for PMI estimation: A pilot study. Forensic Sci. Int. 2023, 349, 111774. [Google Scholar] [CrossRef]
  266. Brockbals, L.; Garrett-Rickman, S.; Fu, S.; Ueland, M.; McNevin, D.; Padula, M.P. Estimating the time of human decomposition based on skeletal muscle biopsy samples utilizing an untargeted LC-MS/MS-based proteomics approach. Anal. Bioanal. Chem. 2023, 415, 5487–5498. [Google Scholar] [CrossRef]
  267. Kaewsatuan, P.; Poompramun, C.; Kubota, S.; Yongsawatdigul, J.; Molee, W.; Uimari, P.; Molee, A. Thigh muscle metabolic response is linked to feed efficiency and meat characteristics in slow-growing chicken. Poult. Sci. 2023, 102, 102741. [Google Scholar] [CrossRef]
  268. Liu, Y.; Liu, Z.; Xing, T.; Li, J.; Zhang, L.; Jiang, Y.; Gao, F. Insight on the meat quality and carbonylation profile of breast muscle of broilers in response to chronic heat stress: A proteomic research. Food Chem. 2023, 423, 136437. [Google Scholar] [CrossRef] [PubMed]
  269. McGlory, C.; van Vliet, S.; Stokes, T.; Mittendorfer, B.; Phillips, S.M. The impact of exercise and nutrition on the regulation of skeletal muscle mass. J. Physiol. 2019, 597, 1251–1258. [Google Scholar] [CrossRef]
  270. Severinsen, M.C.K.; Pedersen, B.K. Muscle-Organ Crosstalk: The Emerging Roles of Myokines. Endocr. Rev. 2020, 41, 594–609. [Google Scholar] [CrossRef]
  271. Kirk, B.; Feehan, J.; Lombardi, G.; Duque, G. Muscle, Bone, and Fat Crosstalk: The Biological Role of Myokines, Osteokines, and Adipokines. Curr. Osteoporos. Rep. 2020, 18, 388–400. [Google Scholar] [CrossRef]
  272. Bottinelli, R.; Reggiani, C. Human skeletal muscle fibres: Molecular and functional diversity. Prog. Biophys. Mol. Biol. 2000, 73, 195–262. [Google Scholar] [CrossRef]
  273. Schiaffino, S. Fibre types in skeletal muscle: A personal account. Acta. Physiol. 2010, 199, 451–463. [Google Scholar] [CrossRef]
  274. Schiaffino, S.; Reggiani, C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 2011, 91, 1447–1531. [Google Scholar] [CrossRef] [PubMed]
  275. Murach, K.A.; Dungan, C.M.; Kosmac, K.; Voigt, T.B.; Tourville, T.W.; Miller, M.S.; Bamman, M.M.; Peterson, C.A.; Toth, M.J. Fiber typing human skeletal muscle with fluorescent immunohistochemistry. J. Appl. Physiol. 2019, 127, 1632–1639. [Google Scholar] [CrossRef]
  276. Sawano, S.; Mizunoya, W. History and development of staining methods for skeletal muscle fiber types. Histol. Histopathol. 2022, 37, 493–503. [Google Scholar] [CrossRef]
  277. Kallabis, S.; Abraham, L.; Müller, S.; Dzialas, V.; Türk, C.; Wiederstein, J.L.; Bock, T.; Nolte, H.; Nogara, L.; Blaauw, B.; et al. High-throughput proteomics fiber typing (ProFiT) for comprehensive characterization of single skeletal muscle fibers. Skelet. Muscle 2020, 10, 7. [Google Scholar] [CrossRef] [PubMed]
  278. Aebersold, R.; Agar, J.N.; Amster, I.J.; Baker, M.S.; Bertozzi, C.R.; Boja, E.S.; Costello, C.E.; Cravatt, B.F.; Fenselau, C.; Garcia, B.A.; et al. How many human proteoforms are there? Nat. Chem. Biol. 2018, 14, 206–214. [Google Scholar] [CrossRef]
  279. Tunyasuvunakool, K.; Adler, J.; Wu, Z.; Green, T.; Zielinski, M.; Žídek, A.; Bridgland, A.; Cowie, A.; Meyer, C.; Laydon, A.; et al. Highly accurate protein structure prediction for the human proteome. Nature 2021, 596, 590–596. [Google Scholar] [CrossRef] [PubMed]
  280. Gamazon, E.R.; Stranger, B.E. Genomics of alternative splicing: Evolution, development and pathophysiology. Hum Genet. 2014, 133, 679–687. [Google Scholar] [CrossRef] [PubMed]
  281. Nakka, K.; Ghigna, C.; Gabellini, D.; Dilworth, F.J. Diversification of the muscle proteome through alternative splicing. Skelet. Muscle 2018, 8, 8. [Google Scholar] [CrossRef] [PubMed]
  282. García-Cruz, C.; Aragón, J.; Lourdel, S.; Annan, A.; Roger, J.E.; Montanez, C.; Vaillend, C. Tissue- and cell-specific whole-transcriptome meta-analysis from brain and retina reveals differential expression of dystrophin complexes and new dystrophin spliced isoforms. Hum. Mol. Genet. 2023, 32, 659–676. [Google Scholar] [CrossRef] [PubMed]
  283. Dowling, P.; Swandulla, D.; Ohlendieck, K. Biochemical and proteomic insights into sarcoplasmic reticulum Ca2+-ATPase complexes in skeletal muscles. Expert Rev. Proteom. 2023, 20, 125–142. [Google Scholar] [CrossRef]
  284. Ohlendieck, K. Towards an understanding of the dystrophin-glycoprotein complex: Linkage between the extracellular matrix and the membrane cytoskeleton in muscle fibers. Eur. J. Cell Biol. 1996, 69, 1–10. [Google Scholar]
  285. Ibraghimov-Beskrovnaya, O.; Ervasti, J.M.; Leveille, C.J.; Slaughter, C.A.; Sernett, S.W.; Campbell, K.P. Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix. Nature 1992, 355, 696–702. [Google Scholar] [CrossRef] [PubMed]
  286. Murphy, S.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Chemical crosslinking analysis of β-dystroglycan in dystrophin-deficient skeletal muscle. HRB Open Res. 2018, 1, 17. [Google Scholar] [CrossRef]
  287. Deutsch, E.W.; Bandeira, N.; Perez-Riverol, Y.; Sharma, V.; Carver, J.J.; Mendoza, L.; Kundu, D.J.; Wang, S.; Bandla, C.; Kamatchinathan, S.; et al. The ProteomeXchange consortium at 10 years: 2023 update. Nucleic Acids Res. 2023, 51, D1539–D1548. [Google Scholar] [CrossRef] [PubMed]
  288. Burniston, J.G.; Connolly, J.; Kainulainen, H.; Britton, S.L.; Koch, L.G. Label-free profiling of skeletal muscle using high-definition mass spectrometry. Proteomics 2014, 14, 2339–2344. [Google Scholar] [CrossRef] [PubMed]
  289. Malik, Z.A.; Cobley, J.N.; Morton, J.P.; Close, G.L.; Edwards, B.J.; Koch, L.G.; Britton, S.L.; Burniston, J.G. Label-Free LC-MS Profiling of Skeletal Muscle Reveals Heart-Type Fatty Acid Binding Protein as a Candidate Biomarker of Aerobic Capacity. Proteomes 2013, 1, 290–308. [Google Scholar] [CrossRef]
  290. Murphy, S.; Zweyer, M.; Raucamp, M.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of the mouse diaphragm and refined mass spectrometric analysis of the dystrophic phenotype. J. Muscle Res. Cell. Motil. 2019, 40, 9–28. [Google Scholar] [CrossRef]
  291. Deshmukh, A.S.; Steenberg, D.E.; Hostrup, M.; Birk, J.B.; Larsen, J.K.; Santos, A.; Kjøbsted, R.; Hingst, J.R.; Schéele, C.C.; Murgia, M.; et al. Deep muscle-proteomic analysis of freeze-dried human muscle biopsies reveals fiber type-specific adaptations to exercise training. Nat. Commun. 2021, 12, 304. [Google Scholar] [CrossRef]
  292. Hadrévi, J.; Hellström, F.; Kieselbach, T.; Malm, C.; Pedrosa-Domellöf, F. Protein differences between human trapezius and vastus lateralis muscles determined with a proteomic approach. BMC Musculoskelet. Disord. 2011, 12, 181. [Google Scholar] [CrossRef]
  293. Tan, X.; He, Y.; He, Y.; Yan, Z.; Chen, J.; Zhao, R.; Sui, X.; Zhang, L.; Du, X.; Irwin, D.M.; et al. Comparative Proteomic Analysis of Glycolytic and Oxidative Muscle in Pigs. Genes 2023, 14, 361. [Google Scholar] [CrossRef] [PubMed]
  294. Wei, W.; Zha, C.; Jiang, A.; Chao, Z.; Hou, L.; Liu, H.; Huang, R.; Wu, W. A Combined Differential Proteome and Transcriptome Profiling of Fast- and Slow-Twitch Skeletal Muscle in Pigs. Foods 2022, 11, 2842. [Google Scholar] [CrossRef] [PubMed]
  295. Bornstein, B.; Heinemann-Yerushalmi, L.; Krief, S.; Adler, R.; Dassa, B.; Leshkowitz, D.; Kim, M.; Bewick, G.; Banks, R.W.; Zelzer, E. Molecular characterization of the intact mouse muscle spindle using a multi-omics approach. eLife 2023, 12, e81843. [Google Scholar] [CrossRef] [PubMed]
  296. Jones, R.A.; Harrison, C.; Eaton, S.L.; Llavero Hurtado, M.; Graham, L.C.; Alkhammash, L.; Oladiran, O.A.; Gale, A.; Lamont, D.J.; Simpson, H.; et al. Cellular and Molecular Anatomy of the Human Neuromuscular Junction. Cell Rep. 2017, 21, 2348–2356. [Google Scholar] [CrossRef]
  297. Mate, S.E.; Brown, K.J.; Hoffman, E.P. Integrated genomics and proteomics of the Torpedo californica electric organ: Concordance with the mammalian neuromuscular junction. Skelet. Muscle 2011, 1, 20. [Google Scholar] [CrossRef] [PubMed]
  298. Borok, M.; Didier, N.; Gattazzo, F.; Ozturk, T.; Corneau, A.; Rouard, H.; Relaix, F. Progressive and Coordinated Mobilization of the Skeletal Muscle Niche throughout Tissue Repair Revealed by Single-Cell Proteomic Analysis. Cells 2021, 10, 744. [Google Scholar] [CrossRef] [PubMed]
  299. Fernández-Lázaro, D.; Garrosa, E.; Seco-Calvo, J.; Garrosa, M. Potential Satellite Cell-Linked Biomarkers in Aging Skeletal Muscle Tissue: Proteomics and Proteogenomics to Monitor Sarcopenia. Proteomes 2022, 10, 29. [Google Scholar] [CrossRef] [PubMed]
  300. Vitorino, R.; Ferreira, R.; Neuparth, M.; Guedes, S.; Williams, J.; Tomer, K.B.; Domingues, P.M.; Appell, H.J.; Duarte, J.A.; Amado, F.M. Subcellular proteomics of mice gastrocnemius and soleus muscles. Anal. Biochem. 2007, 366, 156–169. [Google Scholar] [CrossRef]
  301. Dowling, P.; Gargan, S.; Swandulla, D.; Ohlendieck, K. Identification of Subproteomic Markers for Skeletal Muscle Profiling. Methods Mol. Biol. 2023, 2596, 291–302. [Google Scholar] [CrossRef] [PubMed]
  302. Lee, Y.H.; Tan, H.T.; Chung, M.C. Subcellular fractionation methods and strategies for proteomics. Proteomics 2010, 10, 3935–3956. [Google Scholar] [CrossRef] [PubMed]
  303. Plöscher, M.; Granvogl, B.; Reisinger, V.; Masanek, A.; Eichacker, L.A. Organelle proteomics. Methods Mol. Biol. 2009, 519, 65–82. [Google Scholar] [CrossRef]
  304. Liu, Z.; Du, X.; Yin, C.; Chang, Z. Shotgun proteomic analysis of sarcoplasmic reticulum preparations from rabbit skeletal muscle. Proteomics 2013, 13, 2335–2338. [Google Scholar] [CrossRef]
  305. Liu, Z.; Du, X.; Deng, J.; Gu, M.; Hu, H.; Gui, M.; Yin, C.C.; Chang, Z. The interactions between mitochondria and sarcoplasmic reticulum and the proteome characterization of mitochondrion-associated membrane from rabbit skeletal muscle. Proteomics 2015, 15, 2701–2704. [Google Scholar] [CrossRef] [PubMed]
  306. Anunciado-Koza, R.V.P.; Guntur, A.R.; Vary, C.P.; Gartner, C.A.; Nowak, M.; Koza, R.A. Purification of functional mouse skeletal muscle mitochondria using percoll density gradient centrifugation. BMC Res. Notes 2023, 16, 243. [Google Scholar] [CrossRef] [PubMed]
  307. Chae, S.; Kim, S.J.; Do Koo, Y.; Lee, J.H.; Kim, H.; Ahn, B.Y.; Ha, Y.C.; Kim, Y.H.; Jang, M.G.; Koo, K.H.; et al. A mitochondrial proteome profile indicative of type 2 diabetes mellitus in skeletal muscles. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [PubMed]
  308. Gonzalez-Franquesa, A.; Stocks, B.; Chubanava, S.; Hattel, H.B.; Moreno-Justicia, R.; Peijs, L.; Treebak, J.T.; Zierath, J.R.; Deshmukh, A.S. Mass-spectrometry-based proteomics reveals mitochondrial supercomplexome plasticity. Cell Rep. 2021, 35, 109180. [Google Scholar] [CrossRef] [PubMed]
  309. Maughan, D.W.; Henkin, J.A.; Vigoreaux, J.O. Concentrations of glycolytic enzymes and other cytosolic proteins in the diffusible fraction of a vertebrate muscle proteome. Mol. Cell. Proteom. 2005, 4, 1541–1549. [Google Scholar] [CrossRef]
  310. Murphy, S.; Dowling, P.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of giant skeletal muscle proteins. Expert Rev. Proteom. 2019, 16, 241–256. [Google Scholar] [CrossRef]
  311. Fomchenko, K.M.; Walsh, E.M.; Yang, X.; Verma, R.X.; Lin, B.L.; Nieuwenhuis, T.O.; Patil, A.H.; Fox-Talbot, K.; McCall, M.N.; Kass, D.A.; et al. Spatial Proteomic Approach to Characterize Skeletal Muscle Myofibers. J. Proteome Res. 2021, 20, 888–894. [Google Scholar] [CrossRef]
  312. Melby, J.A.; Brown, K.A.; Gregorich, Z.R.; Roberts, D.S.; Chapman, E.A.; Ehlers, L.E.; Gao, Z.; Larson, E.J.; Jin, Y.; Lopez, J.R.; et al. High sensitivity top-down proteomics captures single muscle cell heterogeneity in large proteoforms. Proc. Natl. Acad. Sci. USA 2023, 120, e2222081120. [Google Scholar] [CrossRef] [PubMed]
  313. Momenzadeh, A.; Jiang, Y.; Kreimer, S.; Teigen, L.E.; Zepeda, C.S.; Haghani, A.; Mastali, M.; Song, Y.; Hutton, A.; Parker, S.J.; et al. A Complete Workflow for High Throughput Human Single Skeletal Muscle Fiber Proteomics. J. Am. Soc. Mass Spectrom. 2023, 34, 1858–1867. [Google Scholar] [CrossRef] [PubMed]
  314. Schiaffino, S.; Reggiani, C.; Kostrominova, T.Y.; Mann, M.; Murgia, M. Mitochondrial specialization revealed by single muscle fiber proteomics: Focus on the Krebs cycle. Scand. J. Med. Sci. Sports 2015, 25 (Suppl. S4), 41–48. [Google Scholar] [CrossRef]
  315. Chen, X.; Li, J.; Hou, J.; Xie, Z.; Yang, F. Mammalian mitochondrial proteomics: Insights into mitochondrial functions and mitochondria-related diseases. Expert Rev. Proteom. 2010, 7, 333–345. [Google Scholar] [CrossRef] [PubMed]
  316. Gómez-Serrano, M.; Camafeita, E.; Loureiro, M.; Peral, B. Mitoproteomics: Tackling Mitochondrial Dysfunction in Human Disease. Oxid. Med. Cell. Longev. 2018, 2018, 1435934. [Google Scholar] [CrossRef] [PubMed]
  317. Mi, H.; Ebert, D.; Muruganujan, A.; Mills, C.; Albou, L.P.; Mushayamaha, T.; Thomas, P.D. PANTHER version 16: A revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res. 2021, 49, D394–D403. [Google Scholar] [CrossRef]
  318. Le Bihan, M.C.; Bigot, A.; Jensen, S.S.; Dennis, J.L.; Rogowska-Wrzesinska, A.; Lainé, J.; Gache, V.; Furling, D.; Jensen, O.N.; Voit, T.; et al. In-depth analysis of the secretome identifies three major independent secretory pathways in differentiating human myoblasts. J. Proteom. 2012, 77, 344–356. [Google Scholar] [CrossRef] [PubMed]
  319. Hartwig, S.; Raschke, S.; Knebel, B.; Scheler, M.; Irmler, M.; Passlack, W.; Muller, S.; Hanisch, F.G.; Franz, T.; Li, X.; et al. Secretome profiling of primary human skeletal muscle cells. Biochim. Biophys. Acta 2014, 1844, 1011–1017. [Google Scholar] [CrossRef]
  320. Leuchtmann, A.B.; Adak, V.; Dilbaz, S.; Handschin, C. The Role of the Skeletal Muscle Secretome in Mediating Endurance and Resistance Training Adaptations. Front. Physiol. 2021, 12, 709807. [Google Scholar] [CrossRef]
  321. Florin, A.; Lambert, C.; Sanchez, C.; Zappia, J.; Durieux, N.; Tieppo, A.M.; Mobasheri, A.; Henrotin, Y. The secretome of skeletal muscle cells: A systematic review. Osteoarthr. Cartil. Open 2020, 2, 100019. [Google Scholar] [CrossRef]
  322. Dowling, P.; Gargan, S.; Zweyer, M.; Sabir, H.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of carbonic anhydrase CA3 in skeletal muscle. Expert Rev. Proteom. 2021, 18, 1073–1086. [Google Scholar] [CrossRef] [PubMed]
  323. Dowling, P.; Gargan, S.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of fatty acid binding proteins in muscular dystrophy. Expert Rev. Proteom. 2020, 17, 137–148. [Google Scholar] [CrossRef] [PubMed]
  324. O’Sullivan, E.M.; Dowling, P.; Swandulla, D.; Ohlendieck, K. Proteomic Identification of Saliva Proteins as Noninvasive Diagnostic Biomarkers. Methods Mol. Biol. 2023, 2596, 147–167. [Google Scholar] [CrossRef] [PubMed]
  325. Burch, P.M.; Pogoryelova, O.; Goldstein, R.; Bennett, D.; Guglieri, M.; Straub, V.; Bushby, K.; Lochmüller, H.; Morris, C. Muscle-Derived Proteins as Serum Biomarkers for Monitoring Disease Progression in Three Forms of Muscular Dystrophy. J. Neuromuscul. Dis. 2015, 2, 241–255. [Google Scholar] [CrossRef]
  326. Brancaccio, P.; Lippi, G.; Maffulli, N. Biochemical markers of muscular damage. Clin. Chem. Lab. Med. 2010, 48, 757–767. [Google Scholar] [CrossRef]
  327. Ohlendieck, K. Proteomics of skeletal muscle glycolysis. Biochim. Biophys. Acta. 2010, 1804, 2089–21101. [Google Scholar] [CrossRef]
  328. Bouley, J.; Chambon, C.; Picard, B. Mapping of bovine skeletal muscle proteins using two-dimensional gel electrophoresis and mass spectrometry. Proteomics 2004, 4, 1811–1824. [Google Scholar] [CrossRef] [PubMed]
  329. Xu, Y.; Qian, H.; Feng, X.; Xiong, Y.; Lei, M.; Ren, Z.; Zuo, B.; Xu, D.; Ma, Y.; Yuan, H. Differential proteome and transcriptome analysis of porcine skeletal muscle during development. J. Proteom. 2012, 75, 2093–2108. [Google Scholar] [CrossRef]
  330. Wang, X.; Shi, T.; Zhao, Z.; Hou, H.; Zhang, L. Proteomic analyses of sheep (ovis aries) embryonic skeletal muscle. Sci. Rep. 2020, 10, 1750. [Google Scholar] [CrossRef]
  331. Camera, D.M.; Burniston, J.G.; Pogson, M.A.; Smiles, W.J.; Hawley, J.A. Dynamic proteome profiling of individual proteins in human skeletal muscle after a high-fat diet and resistance exercise. FASEB J. 2017, 31, 5478–5494. [Google Scholar] [CrossRef]
  332. Hesketh, S.J.; Sutherland, H.; Lisboa, P.J.; Jarvis, J.C.; Burniston, J.G. Adaptation of rat fast-twitch muscle to endurance activity is underpinned by changes to protein degradation as well as protein synthesis. FASEB J. 2020, 34, 10398–10417. [Google Scholar] [CrossRef]
  333. Isfort, R.J.; Wang, F.; Greis, K.D.; Sun, Y.; Keough, T.W.; Farrar, R.P.; Bodine, S.C.; Anderson, N.L. Proteomic analysis of rat soleus muscle undergoing hindlimb suspension-induced atrophy and reweighting hypertrophy. Proteomics 2002, 2, 543–550. [Google Scholar] [CrossRef]
  334. Moriggi, M.; Vasso, M.; Fania, C.; Capitanio, D.; Bonifacio, G.; Salanova, M.; Blottner, D.; Rittweger, J.; Felsenberg, D.; Cerretelli, P.; et al. Long term bed rest with and without vibration exercise countermeasures: Effects on human muscle protein dysregulation. Proteomics 2010, 10, 3756–3774. [Google Scholar] [CrossRef] [PubMed]
  335. Sun, H.; Qiu, J.; Chen, Y.; Yu, M.; Ding, F.; Gu, X. Proteomic and bioinformatic analysis of differentially expressed proteins in denervated skeletal muscle. Int. J. Mol. Med. 2014, 33, 1586–1596. [Google Scholar] [CrossRef] [PubMed]
  336. Lang, F.; Khaghani, S.; Türk, C.; Wiederstein, J.L.; Hölper, S.; Piller, T.; Nogara, L.; Blaauw, B.; Günther, S.; Müller, S.; et al. Single Muscle Fiber Proteomics Reveals Distinct Protein Changes in Slow and Fast Fibers during Muscle Atrophy. J. Proteome Res. 2018, 17, 3333–3347. [Google Scholar] [CrossRef] [PubMed]
  337. Gelfi, C.; Vigano, A.; Ripamonti, M.; Pontoglio, A.; Begum, S.; Pellegrino, M.A.; Grassi, B.; Bottinelli, R.; Wait, R.; Cerretelli, P. The human muscle proteome in aging. J. Proteome Res. 2006, 5, 1344–1353. [Google Scholar] [CrossRef] [PubMed]
  338. Doran, P.; Gannon, J.; O’Connell, K.; Ohlendieck, K. Aging skeletal muscle shows a drastic increase in the small heat shock proteins alphaB-crystallin/HspB5 and cvHsp/HspB7. Eur. J. Cell Biol. 2007, 86, 629–640. [Google Scholar] [CrossRef]
  339. Doran, P.; O’Connell, K.; Gannon, J.; Kavanagh, M.; Ohlendieck, K. Opposite pathobiochemical fate of pyruvate kinase and adenylate kinase in aged rat skeletal muscle as revealed by proteomic DIGE analysis. Proteomics 2008, 8, 364–377. [Google Scholar] [CrossRef]
  340. Staunton, L.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle. Int. J. Mol. Med. 2012, 30, 723–733. [Google Scholar] [CrossRef]
  341. Gueugneau, M.; Coudy-Gandilhon, C.; Gourbeyre, O.; Chambon, C.; Combaret, L.; Polge, C.; Taillandier, D.; Attaix, D.; Friguet, B.; Maier, A.B.; et al. Proteomics of muscle chronological ageing in post-menopausal women. BMC Genom. 2014, 15, 1165. [Google Scholar] [CrossRef] [PubMed]
  342. Ohlendieck, K. Two-CyDye-Based 2D-DIGE Analysis of Aged Human Muscle Biopsy Specimens. Methods Mol. Biol. 2023, 2596, 265–289. [Google Scholar] [CrossRef] [PubMed]
  343. Ebhardt, H.A.; Degen, S.; Tadini, V.; Schilb, A.; Johns, N.; Greig, C.A.; Fearon, K.C.H.; Aebersold, R.; Jacobi, C. Comprehensive proteome analysis of human skeletal muscle in cachexia and sarcopenia: A pilot study. J. Cachexia Sarcopenia Muscle 2017, 8, 567–582. [Google Scholar] [CrossRef] [PubMed]
  344. Ubaida-Mohien, C.; Lyashkov, A.; Gonzalez-Freire, M.; Tharakan, R.; Shardell, M.; Moaddel, R.; Semba, R.D.; Chia, C.W.; Gorospe, M.; Sen, R.; et al. Discovery proteomics in aging human skeletal muscle finds change in spliceosome, immunity, proteostasis and mitochondria. Elife 2019, 8, e49874. [Google Scholar] [CrossRef] [PubMed]
  345. Yu, H.; Tai, Q.; Yang, C.; Gao, M.; Zhang, X. Technological development of multidimensional liquid chromatography-mass spectrometry in proteome research. J. Chromatogr. A 2023, 1700, 464048. [Google Scholar] [CrossRef]
Figure 1. Overview of the initial preparative steps involved in top-down proteomics and bottom-up proteomics to study skeletal muscles. Abbreviations used: pI, isoelectric point; MW, molecular weight (in kDa).
Figure 1. Overview of the initial preparative steps involved in top-down proteomics and bottom-up proteomics to study skeletal muscles. Abbreviations used: pI, isoelectric point; MW, molecular weight (in kDa).
Cells 12 02560 g001
Figure 2. Comparison of the experimental steps involved in top-down proteomics versus bottom-up proteomics to study skeletal muscles. An overview of proteases used for the controlled protein digestion prior to peptide mass spectrometric analysis is provided. Abbreviations used: pI, isoelectric point; MW, molecular weight (in kDa).
Figure 2. Comparison of the experimental steps involved in top-down proteomics versus bottom-up proteomics to study skeletal muscles. An overview of proteases used for the controlled protein digestion prior to peptide mass spectrometric analysis is provided. Abbreviations used: pI, isoelectric point; MW, molecular weight (in kDa).
Cells 12 02560 g002
Figure 3. Overview of major mass spectrometric methods and data acquisition approaches that are routinely used in skeletal muscle proteomics (m/z, mass-to-charge ratio). The figure also lists key mass spectrometric methods employed for the structural analysis of native protein confirmation and the cellular analysis of protein expression levels.
Figure 3. Overview of major mass spectrometric methods and data acquisition approaches that are routinely used in skeletal muscle proteomics (m/z, mass-to-charge ratio). The figure also lists key mass spectrometric methods employed for the structural analysis of native protein confirmation and the cellular analysis of protein expression levels.
Cells 12 02560 g003
Figure 4. Overview of the complexity of post-translational modifications (PTMs) and how they are studied by mass spectrometric analysis (m/z, mass-to-charge ratio). Listed are major types of PTMs, including proteolytic cleavage, phosphorylation (P), glycosylation, acetylation (Ac), lipidation, disulfide bonding (S-S), sumoylation (SU), methylation (Me), hydroxylation (OH) and ubiquitination (Ub).
Figure 4. Overview of the complexity of post-translational modifications (PTMs) and how they are studied by mass spectrometric analysis (m/z, mass-to-charge ratio). Listed are major types of PTMs, including proteolytic cleavage, phosphorylation (P), glycosylation, acetylation (Ac), lipidation, disulfide bonding (S-S), sumoylation (SU), methylation (Me), hydroxylation (OH) and ubiquitination (Ub).
Cells 12 02560 g004
Figure 5. Overview of the extent of proteome-wide adaptations due to changed functional demands or pathophysiological alterations in the skeletal musculature. Major cell biological alterations occur during embryonic myogenesis, regenerative adult myogenesis, physical exercise, disuse atrophy, muscle fiber switching, muscular disorders and muscle aging.
Figure 5. Overview of the extent of proteome-wide adaptations due to changed functional demands or pathophysiological alterations in the skeletal musculature. Major cell biological alterations occur during embryonic myogenesis, regenerative adult myogenesis, physical exercise, disuse atrophy, muscle fiber switching, muscular disorders and muscle aging.
Cells 12 02560 g005
Figure 6. Summary of crucial cell biological characteristics of skeletal muscles. The figure includes a list of major histological, physiological and biochemical properties of the two main types of contractile fibers, i.e., slow-twitching type I myofibers and predominantly fast-twitching type II myofibers, as well as an inventory of the diverse functions of the skeletal musculature in the body.
Figure 6. Summary of crucial cell biological characteristics of skeletal muscles. The figure includes a list of major histological, physiological and biochemical properties of the two main types of contractile fibers, i.e., slow-twitching type I myofibers and predominantly fast-twitching type II myofibers, as well as an inventory of the diverse functions of the skeletal musculature in the body.
Cells 12 02560 g006
Figure 7. Overview of the main types of slow-twitching versus fast-twitching myofibers. The table at the right of the figure gives information on established marker proteins of fast versus slow myofibers. Abbreviations used: CSQ, calsequestrin; MyHC, myosin heavy chain; SERCA, sarcoplasmic or endoplasmic reticulum calcium ATPase; SR, sarcoplasmic reticulum.
Figure 7. Overview of the main types of slow-twitching versus fast-twitching myofibers. The table at the right of the figure gives information on established marker proteins of fast versus slow myofibers. Abbreviations used: CSQ, calsequestrin; MyHC, myosin heavy chain; SERCA, sarcoplasmic or endoplasmic reticulum calcium ATPase; SR, sarcoplasmic reticulum.
Cells 12 02560 g007
Figure 8. Diagrammatic presentation of the various layers of biological organization in skeletal muscle tissues. The figure summarizes the complex relationship and flow of information from gene to RNA/mRNA to protein/proteoform to the cytome and physiome in skeletal muscles. Proteomic diversity is due to alternative promoter usage, alternative post-transcriptional RNA splicing mechanisms and a large array of dynamic post-translational modifications (PTMs). For illustrative purposes, an electron microscopical image of rabbit skeletal muscle (biceps femoris) in the longitudinal direction is shown.
Figure 8. Diagrammatic presentation of the various layers of biological organization in skeletal muscle tissues. The figure summarizes the complex relationship and flow of information from gene to RNA/mRNA to protein/proteoform to the cytome and physiome in skeletal muscles. Proteomic diversity is due to alternative promoter usage, alternative post-transcriptional RNA splicing mechanisms and a large array of dynamic post-translational modifications (PTMs). For illustrative purposes, an electron microscopical image of rabbit skeletal muscle (biceps femoris) in the longitudinal direction is shown.
Cells 12 02560 g008
Figure 9. Proteomic complexity in skeletal muscles. The figure provides examples of the three main mechanisms that produce more than one specific proteoform per individual protein-coding gene, which are the underlying processes that produce a highly complex skeletal muscle proteome. As examples of post-transcriptional alternative RNA splicing, alternative promoter usage and post-translational modifications are shown the expression of the Ca2+-ATPase of the sarcoplasmic reticulum, the membrane cytoskeletal protein dystrophin and the sarcolemmal glycoprotein complex formed by the dystroglycans. Abbreviations used: DMD, Duchenne muscular dystrophy; PTMs, post-translational modifications; SERCA, sarcoplasmic or endoplasmic reticulum calcium ATPase.
Figure 9. Proteomic complexity in skeletal muscles. The figure provides examples of the three main mechanisms that produce more than one specific proteoform per individual protein-coding gene, which are the underlying processes that produce a highly complex skeletal muscle proteome. As examples of post-transcriptional alternative RNA splicing, alternative promoter usage and post-translational modifications are shown the expression of the Ca2+-ATPase of the sarcoplasmic reticulum, the membrane cytoskeletal protein dystrophin and the sarcolemmal glycoprotein complex formed by the dystroglycans. Abbreviations used: DMD, Duchenne muscular dystrophy; PTMs, post-translational modifications; SERCA, sarcoplasmic or endoplasmic reticulum calcium ATPase.
Cells 12 02560 g009
Figure 10. Overview of the bioanalytically accessible parts of the highly dynamic skeletal muscle proteome and the main subcellular structures of myofibers.
Figure 10. Overview of the bioanalytically accessible parts of the highly dynamic skeletal muscle proteome and the main subcellular structures of myofibers.
Cells 12 02560 g010
Table 1. List of major mass spectrometric studies focusing on the cataloguing of the skeletal muscle proteome *.
Table 1. List of major mass spectrometric studies focusing on the cataloguing of the skeletal muscle proteome *.
Proteomic AnalysisBioanalytical FocusTechnical ApproachReferences
Initiation of the skeletal muscle proteome project in the early 2000s; establishment of the mouse SWISS 2D-PAGE databaseGastrocnemius muscle, muscle tissues
(Mus musculus, Rattus norvegius)
2D-GE, MALDI-ToF MSSanchez et al. [21];
Yan et al. [22]
Systematic cataloguing of human skeletal musclesVastus lateralis muscle, diagnostic biopsies
(Homo sapiens)
1D-GE, LC-MS/MSHøjlund et al. [26]; Parker et al. [27]; Jiang et al. [53]
Differential proteomic surveys of fast versus slow human musclesVastus lateralis, trapezius and deltoideus muscles
(Homo sapiens)
2D-GE, MALDI-ToF MS, laser capture micro-
dissection
Capitanio et al. [29]; Stuart et al. [238]; Hadrévi et al. [292]
Profiling of human single myofibersVastus lateralis muscle
(Homo sapiens)
Single-myofiber proteomics, LC-MS/MSMurgia et al. [35];
Schiaffino et al. [36];
Momenzadeh et al. [313];
Description of the Human Skeletal Muscle Proteome ProjectOverview of MS-based proteomics used to analyze human skeletal musclesLiterature search; summary of the human muscle proteomeGonzalez-Freire et al. [20]; Gelfi et al. [23]; Capitanio et al. [37]
Systematic cataloguing of animal skeletal musclesGastrocnemius muscle, diaphragm muscle, C2C12 myoblast cell line
(Mus musculus, Rattus norvegius, Ovis aries, Sus scrofa, Bos taurus)
2D-GE, TMT, FASP, LC-MS/MSDeshmukh et al. [25];
Raddatz et al. [28];
Burniston et al. [288];
Murphy et al. [290];
Bouley et al. [328];
Xu et al. [329];
Wang et al. [330]
Differential proteomic surveys of fast versus slow mouse musclesGastrocnemius, soleus, tibialis anterior and extensor digitorum longus muscles
(Mus musculus)
2D-GE, SILAC, LC-MS/MS, ESI-MSDrexler et al. [30]; Gelfi et al. [31]; Okumura et al. [32]
Profiling of animal single myofibersSoleus, extensor digitorum longus, plantaris and vastus lateralis muscles
(Mus musculus, Rattus norvegicus)
Single-myofiber proteomics, ProFiT, FASP, LC-MS/MS, ESI-MSDeshmukh et al. [25];
Eggers et al. [33]; Kallabis et al. [277]; Fomchenko et al. [311]; Melby et al. [312]
Profiling of muscle spindlesSpindles from masseter muscle
(Mus musculus)
Dissection of muscle spindles, LC-MS/MSBornstein et al. [295]
Profiling of neuromuscular junctionLower limb muscles, electric organ
(Homo sapiens, Torpedo californica)
Dissection of neuromuscular junction region, TMT, LC-MS/MSJones et al. [296]; Mate et al. [297]
Profiling of myotendinous junction regionsGastrocnemius, soleus, extensor digitorium longus and tibialis anterior musclesProteomic analysis of laser capture microscopy, LC-MS/MSCan et al. [239]
Profiling of subcellular fractions
(sarcolemma, sarcoplasmic reticulum, mitochondria, sarcosol, giant muscle protein assemblies, contractile apparatus)
Various skeletal muscles
(Mus musculus, Oryctolagus cuniculus)
Enrichment and affinity isolation of subcellular fractions; LC-MS/MSMurphy et al. [92];
Murphy and Ohlendieck [140]; Staunton and Ohlendieck [175]; Vitorino et al. [300]; Liu et al. [304,305];
Anunciado-Koza et al. [306]; Chae et al. [307];
Maughan et al. [309];
Murphy et al. [310]
Systematic cataloguing of the muscle secretomeQuadriceps muscle, primary human muscle cells, myoblasts
(Homo sapiens)
1D-GE, 2D-GE, MALDI-ToF MS, ESI-MS, LS-MS/MS; literature reviewLe Bihan et al. [318];
Hartwig et al. [319]; Florin et al. [321]
Profiling of the effects of exercise on the skeletal muscle proteomeVastus lateralis and soleus muscles
(Homo sapiens, Rattus norvegius)
2D-GE, LC-MS/MS, ESI-MS, PTM analysis; literature reviewsCervone et al. [38];
Hesketh et al. [39]; Petriz et al. [40]; Hoffman et al. [248]; Hostrup et al. [249]; Koopman et al. [251];
Malik et al. [289]; Camera et al. [331]; Hesketh et al. [332]
Proteomic profiling of muscular disuse atrophyGastrocnemius, soleus and vastus lateralis muscles
(Homo sapiens, Rattus norvegius)
2D-GE, single-fiber proteomics, LC-MS/MSDoering et al. [258]; Li et al. [259]; Blottner et al. [260]; Murgia et al. [261]; Isfort et al. [333]; Moriggi et al. [334]; Sun et al., [335]; Lang et al. [336]
Proteomic profiling of muscle plasticityTibialis anterior muscle
(Oryctolagus cuniculus)
Muscle electro-stimulation, 2D-DIGE, ESI-MSOhlendieck [43];
Donoghue et al. [257]
Proteomic profiling of skeletal muscle agingVastus lateralis and gastrocnemius muscles, single myofibers, freeze-dried muscle specimens
(Homo sapiens, Mus musculus, Rattus norvegius)
2D-GE, 2D-DIGE, TMT, MALDI-ToF MS, ESI-MS, LC-MS/MSMurgia et al. [34]; Gannon et al. [130];
O’Connell et al. [135];
Théron et al. [264]; Deshmukh et al. [291];
Gelfi et al. [337];
Doran et al. [338,339]; Staunton et al. [340]; Gueugneau et al. [341]; Ohlendieck [342];
Ebhardt et al. [343];
Ubaida-Mohien et al. [344]
* Abbreviations used: DIGE, difference gel electrophoresis; ESI, electrospray ionization; FASP, filter-aided sample preparation; GE, gel electrophoresis; LC-MS/MS, liquid chromatography tandem mass spectrometry; MALDI-ToF, matrix-assisted laser desorption/ionization time-of-flight; MS, mass spectrometry; ProFiT, proteomics high-throughput fiber typing; PTM, post-translational modification; SILAC, stable isotope labeling with amino acids in cell culture; TMT, isobaric tandem mass tagging.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dowling, P.; Swandulla, D.; Ohlendieck, K. Mass Spectrometry-Based Proteomic Technology and Its Application to Study Skeletal Muscle Cell Biology. Cells 2023, 12, 2560. https://doi.org/10.3390/cells12212560

AMA Style

Dowling P, Swandulla D, Ohlendieck K. Mass Spectrometry-Based Proteomic Technology and Its Application to Study Skeletal Muscle Cell Biology. Cells. 2023; 12(21):2560. https://doi.org/10.3390/cells12212560

Chicago/Turabian Style

Dowling, Paul, Dieter Swandulla, and Kay Ohlendieck. 2023. "Mass Spectrometry-Based Proteomic Technology and Its Application to Study Skeletal Muscle Cell Biology" Cells 12, no. 21: 2560. https://doi.org/10.3390/cells12212560

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