Next Article in Journal
Enhanced Electrophoretic Depletion of Sodium Dodecyl Sulfate with Methanol for Membrane Proteome Analysis by Mass Spectrometry
Next Article in Special Issue
Observations from the Proteomics Bench
Previous Article in Journal
The Mechanisms of Regulated Cell Death: Structural and Functional Proteomic Pathways Induced or Inhibited by a Specific Protein—A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction?

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
Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France
4
Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany
*
Author to whom correspondence should be addressed.
Proteomes 2024, 12(1), 4; https://doi.org/10.3390/proteomes12010004
Submission received: 5 December 2023 / Revised: 9 January 2024 / Accepted: 12 January 2024 / Published: 16 January 2024

Abstract

:
This perspective article is concerned with the question of how proteomics, which is a core technique of systems biology that is deeply embedded in the multi-omics field of modern bioresearch, can help us better understand the molecular pathogenesis of complex diseases. As an illustrative example of a monogenetic disorder that primarily affects the neuromuscular system but is characterized by a plethora of multi-system pathophysiological alterations, the muscle-wasting disease Duchenne muscular dystrophy was examined. Recent achievements in the field of dystrophinopathy research are described with special reference to the proteome-wide complexity of neuromuscular changes and body-wide alterations/adaptations. Based on a description of the current applications of top-down versus bottom-up proteomic approaches and their technical challenges, future systems biological approaches are outlined. The envisaged holistic and integromic bioanalysis would encompass the integration of diverse omics-type studies including inter- and intra-proteomics as the core disciplines for systematic protein evaluations, with sophisticated biomolecular analyses, including physiology, molecular biology, biochemistry and histochemistry. Integrated proteomic findings promise to be instrumental in improving our detailed knowledge of pathogenic mechanisms and multi-system dysfunction, widening the available biomarker signature of dystrophinopathy for improved diagnostic/prognostic procedures, and advancing the identification of novel therapeutic targets to treat Duchenne muscular dystrophy.

1. Introduction

The extraordinary cellular complexity and molecular heterogeneity of biological systems are formidable bioanalytical challenges for the systematic survey of dynamic processes during physiological adaptations under healthy conditions versus pathophysiological alterations in a diseased state. The multi-cellular organization of the average human body is estimated to contain over 400 different cell types that form a network of over 36 trillion cells, which in turn display trillions of molecules per average cellular unit, including diverse species of nucleic acids, proteins, carbohydrates, lipids, minerals and metabolites [1,2,3,4]. A meta-analysis of protein abundance distribution suggests that the average eukaryotic cell contains approximately 43 million proteins [5], making the systematic cataloging and differential analysis of all protein species in health and disease a daunting task in modern proteomics [6,7,8]. At the level of the hierarchical biological organization of highly complex biomolecular systems, the number of protein-coding genes has been determined to be approximately 20,000 individual human genes [9,10,11], which probably generate over a million dynamic proteoforms [12,13,14] due to extensive genomic variations, alternative RNA splicing and extensive post-translational modifications of protein products [15,16,17]. Proteoforms can be defined as the expressed variants of the protein products that are encoded by a single gene, whereby the different molecular forms are generated by genetic variations such as alternative promoter usage, alternative splicing of RNA transcripts due to mechanisms such as exon skipping, and extensive post-translational modifications, including proteolysis, phosphorylation and glycosylation [12].
This perspective article reviews and discusses the means by which proteomics can be employed in an optimum way to identify and characterize the individual proteoforms that are present in the technically accessible skeletal muscle proteome [18,19,20]. The main focus is on the proteomic survey of the molecular and cellular pathogenesis of a multi-system neuromuscular disorder [21,22], i.e., X-linked Duchenne muscular dystrophy (DMD) [23,24]. The abnormal expression of the full-length Dp427-M isoform of the membrane cytoskeletal protein dystrophin [25] that is encoded by one of the largest genes in the human genome, the DMD gene [26], is the underlying cause of this devastating disorder [27]. Although this monogenetic disease can be classified as a primary muscle-wasting disorder with a main defect in the membrane cytoskeleton [28], the progressive decline of contractile strength in Duchenne patients is accompanied by body-wide alterations [29,30,31] and multi-system dysfunction [32,33,34]. This review briefly summarizes key results from systematic proteomic studies with special reference to muscular dystrophy research. A description of the routine usage of top-down proteomics versus bottom-up proteomics in basic and applied myology, including a discussion of bioanalytical advantages versus technical challenges, is provided. Besides studying the molecular pathogenesis of dystrophinopathy and being an irreplicable tool for biomarker discovery, proteomics is also highly suitable for the identification of novel therapeutic targets and the elucidation of drug mechanisms [34].
Based on the high-throughput and large-scale methodology that is currently available for proteomic applications, a future systems biological approach is outlined for the holistic characterization of dystrophinopathy. This would include the amalgamation of findings from multi-omics studies, using mass spectrometry (MS)-based proteomics as the core discipline for conducting protein biochemical surveys, to achieve a high degree of integromic data handling [35]. A key bioanalytical aspect would be to assess pathophysiological crosstalk between individual organ proteomes in muscular dystrophy that could include the evaluation of the role of muscle–bone interactions, brain–muscle signaling, the metabolic liver–fat–muscle axis, the influence of muscular alterations on the kidneys, gastrointestinal tract and the cardio-respiratory system, as well as the linkage of muscle changes to immune responses. Verification studies to evaluate the validity of multi-omics-based networks and their involvement in disease processes would be carried out by robust and standardized physiological, molecular biological, biochemical, immunochemical and histological assays. In the long term, integrated proteomics could form the scientific basis for developing a more complex understanding of neuromuscular pathogenesis and associated multi-system dysregulation, as well as be helpful in identifying novel biomarker candidates for the improved diagnosis, prognosis and therapeutic monitoring of muscle diseases and their body-wide complications.

2. Mass Spectrometry-Based Proteomics: Top-Down versus Middle-Up/Down versus Bottom-Up Approaches

Proteomics can be used for the biochemical identification of individual protein species, the detailed characterization of peptides and proteins and their post-translational modifications, and the systematic cataloging of entire proteomes, as well as for comparative studies of complex protein mixtures [36,37,38]. Mass spectrometric surveys of proteins were initiated in the late 1990s [39,40,41] and have been greatly refined over recent decades to the current state of single-cell resolution [42,43,44]. Current proteomic analysis pipelines, which generally consist of sample preparation, protein extraction, efficient protein separation, different degrees of controlled protein fragmentation, mass spectrometric analysis, bioinformatic assessment and independent verification studies, can be categorized into two main types of approaches, i.e., top-down proteomics [45] versus bottom-up proteomics [46], plus an additional third category in the form of middle-up/down proteomics [47]. Figure 1 provides an overview of the major steps that are involved in routine proteomic analyses of proteins and their specific proteoforms.
The most frequently employed methods in skeletal muscle proteomics have recently been reviewed, including top-down approaches, bottom-up techniques, comparative studies and membrane protein analyses [48,49,50]. The main procedures are briefly summarized in the below subsections, which outline the basic rationale of top-down versus bottom-up proteomics, protein separation, sample handling, the importance of optimized protein digestion, mass spectrometric techniques and data acquisition. Detailed descriptions of the key techniques employed in MS-based proteomics are beyond the scope of this perspective article that instead focuses on the actual application of proteomics for a more in-depth understanding of the pathobiochemical aspects of the multi-system pathology of dystrophinopathy. Excellent reviews are available that provide a comprehensive and critical examination of the main analysis pipelines and most frequently applied methodologies in the field of proteomics research [45,46,47,51,52,53,54].

2.1. Top-Down Proteomic Approaches

Although liquid chromatography (LC) is currently the most frequently used method for large-scale and high-throughput protein separation prior to MS-based analysis, gel electrophoresis (GE) represents an excellent technique for the efficient preparation of isolated and intact proteoforms [55,56,57,58]. Within the portfolio of GE methodology, two-dimensional gel electrophoresis (2D-GE) is a significant and well-established technical platform [15,59,60,61] that allows users to perform a comprehensive top-down proteomic analysis [62,63,64]. Two-dimensional gel electrophoresis that uses isoelectric focusing (IEF) in the first dimension and standard sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) in the second dimension [65] enables the separation of complex proteomes from a variety of samples based on the unique combination of the isoelectric point (pI) and molecular mass of individual protein species [66]. These key parameters are then used to generate a map of proteins, representing changes in protein abundance levels of distinct proteoforms [15], and in some examples, post-translational modifications (PTMs) [67,68,69]. Two-dimensional gel electrophoresis facilitates the separation of hundreds to thousands of proteins on one gel [63], with some research groups having established protocols that allow for over 4000 proteins to be precisely separated with high accuracy [61,70]. Importantly, 2D-GE has been optimized to separate the skeletal muscle proteome [59,60,71].
A number of specialized gel stains are available, including SYPRO Ruby, Deep Purple, silver stain and Coomassie Brilliant Blue (CBB) for protein labeling [72,73,74,75,76], as well as protein species-selective procedures for the detection of glycoproteins, such as Pro-Q Emerald gel stain [77,78], or phosphoproteins, such as Pro-Q Diamond gel stain [79,80]. SYPRO Ruby is a highly sensitive (~1 ng) fluorescent stain that can accurately quantitate protein expression levels. Silver stain and CBB [72] are robust staining methods facilitating the quick visualization of results [73], while Pro-Q Emerald and Pro-Q Diamond are specific to glycosylated and phosphorylated proteins, respectively, conferring a degree of specialization with this approach. Trypsin is the gold standard for protein digestion [81]; however, alternative proteases, such as endoproteinase Glu-C, endoproteinase Asp-N or chymotrypsin, can help increase amino acid sequence coverage by generating unique peptides that are complementary to tryptic peptides [82,83,84,85].
The development of the differential imaging gel electrophoresis technique for increased sensitivity and reproducibility using fluorescent dyes proved to be a significant addition to sample analysis using 2D-GE [86,87,88]. Following optimized sample preparation [89], fluorescence two-dimensional difference gel electrophoresis (2D-DIGE) is based on labeling of protein within a sample with a different fluorophore (CyDye3, CyDye5 or CyDye2) that binds covalently with the epsilon amino group of lysine residues for minimal labeling [90]. Typically, the internal control is a combination of all samples that will be analyzed within a single experiment and is labeled with CyDye2. The CyDye3- and CyDye5-labeled samples can then be normalized to CyDye2 for the identification of protein spots with different abundance levels when comparing samples [91,92,93]. Software packages including DeCyder, SameSpots and Dymension 3 can be incorporated into the workflow to aid in the identification of significant proteins [94,95]. Saturation labeling using CyDye3 and CyDye5 fluorophores with maleimide chemistry can be employed to label all cysteine residues within the sample of interest [96], enhancing sensitivity compared to the minimal approach [90]. As an alternative to gel-based protein separation, multi-dimensional protein identification technology (MudPIT) [97] can be employed, which is based on two-dimensional liquid chromatography (2D-LC) followed by MS-based analysis [98]. Extremely large proteins, such as the class of giant muscle proteins (e.g., titin, nebulin, obscurin, plectin, dystrophin and the ryanodine receptor Ca2+-release channel) [99], which do not properly enter the second dimension of conventional 2D gels due to their high molecular mass, can be separated by agarose 2D-GE [100,101,102] or one-dimensional gradient gel electrophoresis–liquid chromatography (GeLC) [103,104,105]. Recently, Melby et al. [106] described a highly sensitive top-down proteomic approach for the characterization of single myofiber heterogeneity using a one-pot protein extraction and sample processing strategy. The fraction with extracted muscle proteins was separated by a low-flow capillary LC method, which was coupled to a microflow multi-emitter nanoelectrospray source for optimum ionization efficiency prior to the MS-based analysis of intact proteoforms [106].

2.2. Bottom-Up Proteomic Approaches

Bottom-up proteomics refers to the characterization of proteins by analysis of peptides created from the protein through proteolysis [46,107]. An important initial consideration is whether fractionation is necessary or whether a more global analysis is the best approach. Many strategies are available to fractionate samples from relatively small quantities of cells/tissues in a short period of time, generating cytoplasmic, plasma membrane, nuclear, mitochondrial and cytoskeletal fractions as an example of subproteomics [108,109,110]. Column chromatography is also a successful approach, separating proteins based on size, charge or affinity. Isolation of single cells for MS-based analysis has the ability to identify the emergence of cellular heterogeneity and distinct cellular mechanisms underlying pathophysiological processes [111] and has been successfully applied to the study of single myofibers [112].
For bottom-up proteomic approaches, both in-solution and filter-based approaches are routinely used for sample preparation [113,114]. The filter-aided sample preparation (FASP) technique [115] and the suspension trapping (S-Trap) method [116] are popular for peptide generation. Enrichment strategies can be used to isolate and enrich sub-populations of peptides based on specific chemical properties [117,118]. A description of the analysis pipelines for investigating PTMs specific to skeletal muscle cell biology has recently been collated [48]. Phosphoproteomics has become one of the most active research areas in proteomic studies with phosphopeptide enrichment being a critical step in the analysis [119,120]. In addition to dedicated antibodies (pTyr antibody), phosphopeptides are enriched by their selective interaction with metals in the form of chelated metal ions or metal oxides (ferric nitrilotriacetate/Fe-NTA and TiO2 immobilized resins) [121]. Protein ubiquitination is a dynamic multifaceted PTM involved in many key processes of physiology and pathophysiology. The enrichment of ubiquitinated peptides containing ubiquitin remnants can be achieved by antibody-based approaches that specifically recognize the di-glycine motive remaining after digestion with trypsin [122]. In bottom-up proteomics, 1D to 3D LC methods are routinely used approaches in separating peptides prior to MS analysis [123].

2.3. Mass Spectrometric Analysis and Data Acquisition Techniques

Untargeted label-free quantitation (LFQ) of proteins aims to determine the relative abundance of peptides/proteins when comparing multiple biological samples [124,125,126]. LFQ has been successfully integrated into single-cell proteomic workflows [127]. For LFQ, two major approaches are routinely used, i.e., spectral counting and measuring MS1 signal intensities. In contrast to LFQ-based analyses, quantitative label-based techniques [128] are carried out with isobaric tagging for relative and absolute quantitation (iTRAQ) [129], stable isotope labeling by amino acids in cell culture (SILAC) [130], isobaric tandem mass tagging (TMT) [131], isotope-coded protein labeling (ICPL) [132] and isotope-coded affinity tags (ICAT) [133]. With SILAC experiments, proteins are metabolically labeled by culturing cells in media containing normal and heavy isotope amino acids, which are distinguishable by MS when samples are mixed together in equal ratios prior to analysis [130]. Alternatively, in vivo SILAC can be used to study whole organisms, such as mouse models of dystrophinopathy [134]. Multiplexed quantitative proteomics using the TMT technique is an unbiased quantification approach that can be adopted when evaluating peptide/protein abundance levels in a multitude of sample types [135].
Within the MS instrument, fragmentation approaches include the use of collision-induced dissociation (CID), higher-energy collisional dissociation (HCD) and/or electron transfer dissociation (ETD), depending on the specific application [136]. CID has become a routine approach for the fragmentation of peptides for protocols involving LFQ, while ETD is seen as the method of choice for peptides carrying labile PTMs [137].
A number of data acquisition techniques are available when using MS analyses, the two most widely used are data-independent acquisition (DIA) [138] and data-dependent acquisition (DDA) [139]. During MS/MS analysis, MS2 spectra are produced from the fragmentation of a product ion in a particular m/z range, following the operation of MS1 in scan mode. In DIA, effectively all peptides are fragmented together, resulting in complex MS2 spectra, but the values are comprehensive across the run time. Spectral libraries are employed to extract information from the wide-ranging data, facilitating quantification at the MS2 level. Sequential Window Acquisition of all Theoretical Mass Spectra (SWATH) [140] is a specific variant of the DIA approach [141], facilitating a deep proteome analysis, where all ionized peptides that exist within a specified mass range are fragmented in a systematic manner [142].
Table 1 lists major proteomic techniques, as well as a variety of biochemical, immunochemical and cell biological methods that can be used to independently verify the MS-based identification of specific proteoforms or the findings of changes in protein abundance from comparative proteomic studies. These methods are frequently employed to study cell or tissue specimens derived from normal versus diseased skeletal muscles, including the following:
  • Two-dimensional gel electrophoresis (2D-GE) [55,56,57,58,59,60,61,62,63,64,65,66,71];
  • Differential imaging gel electrophoresis (2D-DIGE) [86,87,88,89,90,91,92,93];
  • Specialized gel-based methods for studying specific protein species [67,68,69,77,78,79,80];
  • Gel electrophoresis–liquid chromatography methods (GeLC) [103,104,105];
  • Protein microarrays [143,144,145,146];
  • Sample preparation for proteomic analysis [89,113,114,115,116];
  • Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry [70,147,148];
  • Surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) mass spectrometry [149,150];
  • Liquid chromatography–tandem mass spectrometry (LC-MS/MS) [54,151,152];
  • Label-free quantification (LFQ) mass spectrometry [124,125,126,127];
  • Isobaric tandem mass tagging (TMT) [128,131];
  • Stable isotope labeling by amino acids in cell culture (SILAC) [130,134];
  • Isobaric tagging for relative and absolute quantitation (iTRAQ) [128,129];
  • Native mass spectrometry [153,154,155];
  • Microproteomics using laser capture microdissection [156,157,158];
  • Single-fiber proteomics [112,159];
  • Enzyme-linked immunosorbent assay (ELISA) [160,161,162];
  • Immunoblot analysis [163,164,165];
  • Simoa bead-based immunoassay (SiMoA) [166,167,168];
  • Microscopical analysis [169,170,171,172] including imaging mass cytometry (IMC) that utilizes metal-labeled antibodies [173,174,175];
  • Flow cytometry [176,177,178];
  • Protein interaction assays [179,180,181];
  • Enzyme assays [182,183,184].
Table 1. Overview of key proteomic and biochemical techniques that are commonly used to study skeletal muscles in health and disease.
Table 1. Overview of key proteomic and biochemical techniques that are commonly used to study skeletal muscles in health and disease.
ApproachBioanalytical AdvantagesDisadvantages/LimitationsReferences
Two-dimensional gel electrophoresis (2D-GE)Two-dimensional gel electrophoresis has the ability to separate intact proteoforms. Preset conditions, such as pH ranges, size of the 2D gel and staining methods, can be adjusted to increase resolution. Straightforward to interface with many powerful biochemistry techniques including immunoblotting. Two-dimensional gels can be imaged using stains or fluorescent dyes, including labeling of PTMs.Two-dimensional gel electrophoresis exhibits a narrower dynamic range as compared to certain LC-based separation methods. Difficult to resolve very acidic or very basic proteins. Problematic analysis of very low- or extremely high-molecular-weight proteins.[55,56,57,58,59,60,61,62,63,64,65,66,71]
Differential imaging gel electrophoresis using fluorescence two-dimensional difference gel electrophoresis (2D-DIGE)Two-dimensional difference gel electrophoresis allows the simultaneous investigation and comparison of three different samples on one two-dimensional gel, thus reducing gel-to-gel variability. Normalization within an experiment can be carried out via the inclusion of an internal control (such as CyDye2) in all sample sets.A significant number of steps are involved, taking multiple days to complete. Multiple phenotype comparison is still a challenge using the 2D-DIGE technique.[86,87,88,89,90,91,92,93]
Gel electrophoresis–liquid chromatography (GeLC) methodsThe initial 1D-GE step using the GeLC-MS/MS technique allows for the efficient separation of extremely large proteins that do not properly separate in conventional 2D gels.GeLC-based methods are based on crowded 1D gel bands with the limited resolution of individual protein species.[103,104,105]
Protein microarraysMicroarray technology allows high throughput of samples. Different formats are available for general and targeted custom screening approaches. Systems can be arrayed as semi-quantitative or quantitative formats.High-quality antibodies are not available for all targets. Microarrays require two specific antibodies for each target from the specific sample of interest.[143,144,145,146]
Matrix-assisted laser desorption/ionization time-of-flight (MALDI-ToF) mass spectrometryMALDI-ToF MS is characterized by a simple operation mode and good mass accuracy, as well as high resolution and sensitivity for peptide mass fingerprinting (PMF). The method can be used for profiling and imaging of proteins directly using thin tissue sections (MALDI-IMS; imaging mass spectrometry)The sequence information provided by MALDI-ToF MS is generally not as comprehensive as that generated by LC-MS/MS. The method has reduced success rates for identifying proteins that are not in databases.[70,147,148]
Surface-enhanced laser desorption/ionization time-of-flight (SELDI-ToF) mass spectrometrySELDI-ToF MS allows for high-throughput analyses. The preanalytical sample preparation is rapid and streamlined due to the ability to achieve chromatographic separation using a variety of protein-chip surfaces.Results are generally based on peptides and smaller proteins (<30 kDa). Additional effort is required to identify peaks of interest. Relatively low resolution of MS scans and low sensitivity.[149,150]
Label-free quantification (LFQ) mass spectrometryLFQ MS analysis does not require expensive chemicals or metabolic tags, making it a cost-effective proteomic method. The time needed for sample preparation is significantly reduced due to a straightforward workflow as compared to labeling techniques.Factors such as the peptide or spectral count have limitations. Considerably more LC-MS time is needed for sample analysis. Low-abundance peptides may be underrepresented.[124,125,126,127]
Isobaric tandem mass tagging (TMT)The TMT method makes it possible to analyze a significant number of samples that can be labeled (18-plex). Specifically linked protocols, such as synchronous precursor selection (SPS), can be helpful in identifying and quantifying low-abundance proteins.During TMT experiments, replication in labeling procedures and batch effects cannot be completely uniform. The method is associated with a high cost of reagents.[128,131]
Stable isotope labeling by amino acids in cell culture (SILAC)For the SILAC approach, no in vitro labeling steps are necessary in the experimental procedures. Heavy and light amino acids share the same physico-chemical properties.SILAC has limited sample multiplexing capabilities and can only be carried out using cell culture or labeling of whole organisms.[130,134]
Multiplex enzyme-linked immunosorbent assay (ELISA)The ELISA method can be conveniently used for the verification of the proteomic identification of distinct protein species. Multiplex ELISA techniques use fewer wells and/or plates for sample handling and have increased throughput capabilities and the ability to develop custom panels.Identifying antibodies with high specificity is a challenge due to issues with cross-reactivity. Proteins that are present at different abundance levels make linearity over a wide range of concentrations difficult.[160,161,162]
Microscopical analysisHistological, histochemical and immunofluorescence microscopical studies can be employed to confirm proteomic results. Verification analyses can be carried out with both freshly dissected or frozen tissue samples for single-cell analysis. The techniques allow the subcellular localization of protein expression levels in a tissue sample with a fast turn-around time to achieve meaningful results. Of note, the recent development of imaging mass cytometry using metal-labeled antibodies has greatly improved the scope of microscopical investigations.Although these techniques provide data on the single-cell level, the optimization and quantifying results can be difficult. Immuno-histological studies can be subject to human error. Often, a highly trained histopathologist is needed for the proper interpretation of results. Imaging mass cytometry is associated with high costs due to the production of special antibodies.[169,170,171,172,173,174,175]
Flow cytometry (FC)FC allows simultaneous cell biological analysis with multiple parameters. The method identifies small populations of cells within complex samples and allows for the quantification of fluorescence intensities.For a successful analysis, the method requires the careful choosing of a suitable combination of fluorochrome conjugates. Complex instruments are prone to analytical problems.[176,177,178]

3. Skeletal Muscle Heterogeneity and Muscle Proteomics

Skeletal muscle heterogeneity, cellular complexity and multi-systems crosstalk present serious bioanalytical challenges for systematic proteomic and biochemical studies in basic and applied myology. The skeletal muscle proteome can be defined as the totality of all protein species, i.e., proteoforms and their dynamic PTMs, that are produced at a given time by the muscle-specific expression profile of the genome [185]. It is crucial to keep in mind that skeletal muscle tissues are markedly heterogeneous in their cellular composition. This cell biological fact has to be taken into account during omics-type analyses, including comparative skeletal muscle tissue proteomics [18]. Proteoform heterogeneity due to genomic, post-transcriptional and post-translational effects has a profound influence on skeletal muscle proteome diversity [19,186].

3.1. Tissue Heterogeneity and Cellular Complexity of Skeletal Muscles

Multi-nucleated myofibers contribute to 47% of the biomass in the average human body [4] and are involved in diverse biological functions, such as voluntary movements, posture, bioenergetic and metabolic integration, muscle–skeletal balance, bodily protection, the regulation of thermogenesis, respiration, communication and the provision of an abundant protein reservoir during extended periods of starvation [187,188,189]. The proper physiological functioning of the voluntary contractile system is based on highly coordinated interactions between the central and peripheral nervous systems on the one hand and the various subtypes of skeletal muscles that are associated with a network of capillaries, elaborate layers of extracellular matrix (ECM) and embedded satellite cell populations on the other hand [188]. Individual muscles usually contain a mixture of slow and fast fiber populations plus hybrid fibers. The main biological features that can be used to differentiate between the main myofiber types are histological, physiological, biophysical, metabolic and biochemical properties [190].
Slow-oxidative type I fibers are characterized by small-size motor neurons, relatively small cell diameter, high capillary density, prominent mitochondrial density, slow contraction time with low levels of force production, aerobic activity with high oxidative capacity based on fatty acid oxidation, and low glycolytic metabolism, which is associated with high resistance to fatigue [191]. In contrast, faster-contracting type II fibers exhibit larger-size motor neurons, larger cell diameter, lower capillary density, lower mitochondrial density, fast contraction time with high levels of force production, predominantly anaerobic activity with low-to-intermediate oxidative capacity, and high glycolytic activity based on the metabolization of glucose/glycogen, which is associated with low resistance to fatigue [191]. Type II fibers can be further subdivided into fast-oxidative/glycolytic type IIa and fast-glycolytic type IIx myofibers in mature human muscles [192]. The category of hybrids ranges from type I/IIa to type IIa/IIx myofibers. In small rodents, which are frequently used as animal models in muscular dystrophy research [193], a third subtype of extremely fast-twitching myofibers is present in their musculature, which is classified as type IIb [191].

3.2. Protein Markers of Myofiber Specification

Reliable biochemical/proteomic markers for the different myofiber types are represented by the isoforms of the contractile protein myosin II. Myosin heavy chains (MyHC) of the types MyHC-1/beta (MYH7 gene), MyHC-2a (MYH2 gene), MyHC-2x (MYH1 gene) and MyHC-2b (MYH4 gene) are superb indicators of the main fiber types I, IIa, IIx and IIb, respectively [190,191,192]. Developing and specialized types of skeletal muscles are also associated with particular myosin isoforms, such as MyHC-embryonic (MYH3 gene) and MyHC-neonatal (MYH8 gene) during myogenesis [194,195] and MyHC-eom (MYH13 gene) and MyHC-15 (MYH15 gene) in extraocular muscles [196,197]. A detailed review has recently outlined myosin isoform diversity in the contractile apparatus of skeletal muscles [49]. Fiber type specification is highly plastic, and together with the overall regulation of muscle tissue mass, is heavily influenced by neuromuscular activity levels, load bearing, diverse hormonal effects and nutritional supply [198,199,200]. A variety of histological, histochemical, immunofluorescence microscopical and biochemical methods is routinely used for fiber typing [169,170,171,172,201] but has recently been superseded by more sophisticated and high-throughput proteomic methodology [106,112,202,203].

3.3. Cellular Complexity of the Muscle Environment

The cellular environment of contractile fibers is highly complex and contains myogenic stem cells (MuSCs), mesenchymal stromal cells (MSCs), such as fibro/adipogenic progenitors (FAPs), and resident macrophages [204,205,206]. Muscle-specific satellite cells are positioned between the sarcolemma membrane and basal lamina. Following physical injury or disease, the MuSC pool is activated [207]. High levels of cellular proliferation are involved in the self-renewal of the satellite cell population, and differentiation produces myogenic precursor cells for repair mechanisms [208]. Biochemical markers of inactive satellite cells, activated myogenic cells, myoblasts and fused myotubes are differential expression patterns of CD34 and the transcription factors Pax7, Foxo, MyoG/myogenin, MyoD1 and Myf5 [209,210,211].
The complex cellular arrangement of the mature neuromuscular system creates a protected, regeneratable and highly flexible pool of motor units per individual skeletal muscle. The extrafusal myofiber population that forms a distinct motor unit usually receives its innervating signals from the axonal branches of a single α-motoneuron. Thus, the crucial muscle–nerve connections regulate the patterns of excitation–contraction–relaxation cycles. Coordinated contractions depend on the joined forces that are generated by all motor units forming a physiological motor pool within a single skeletal muscle [212]. Therefore, crude extracts from muscle biopsies are heterogeneous in composition. Of note, due to the specific nature of muscle tissues, which are characterized by large and elongated myofibers, a high abundance of sarcomeric structures and several layers of ECM, in combination with associated issues of subcellular fractionation and protein extraction procedures, only a near-to-complete coverage of the skeletal muscle proteome is currently possible [213].

3.4. Multi-System Interactions of Skeletal Muscles and the Muscle Secretome

Crucial physiological parameters of contractile muscle tissues are multi-systemic interactions that are provided by body-wide skeletal muscle signaling involving myokines [214], in combination with a diverse array of cytokines, hormones, osteokines, adipokines and growth factors [215,216,217]. Myokines can be defined as peptides or proteins that are released or secreted by skeletal muscles into the circulatory system and exert autocrine, paracrine and endocrine effects. Hence, these muscle-derived signaling factors influence the muscle itself at the local level, as well as trigger changes in short- or long-term distant cells/tissues/organs [214]. The secretome of skeletal muscles has been characterized by MS-based proteomics [218] and a large number of myokines have been identified in developing, regenerating and matured muscles [219,220,221]. Major patterns of organ crosstalk and signaling axes are summarized in Figure 2, including the brain, skeletal muscle, heart, bone, liver, gut and fatty tissue [214,215,216,217].
An excellent example of how crosstalk between skeletal muscles and other organ systems can trigger severe pathophysiological changes is muscle-associated rhabdomyolysis. This disorder can be triggered by diverse initiators, such as crush injury, sepsis, drug overdose or extreme physical exertion [222], as well as being based on genetic abnormalities [223]. During an episode of rhabdomyolysis, the disintegration of skeletal muscles releases a large number of proteins and electrolytes into the circulatory system, causing downstream dysfunction that may lead to kidney failure and heart fibrillation. Renal abnormalities are often linked to aggregates of large amounts of released muscle myoglobin and cardiac issues are associated with elevated levels of K+ ions in the circulatory system [224]. The abnormal brownish staining of urine due to the massive release of muscle proteins is a key diagnostic indicator of rhabdomyolysis. Therefore, the proteomic profiling of urine suggests itself as an ideal non-invasive diagnostic tool to monitor the extent of body-wide effects due to the release of muscle-associated proteins during an acute episode of rhabdomyolysis [225], which can be life-threatening [222].

3.5. Skeletal Muscle Proteomics

The proteomic profiling of skeletal muscles is concerned with both the detailed intra-proteomic analysis of the heterogenous cell types that are present within the neuromuscular system and inter-proteomic surveys focusing on the crosstalk between skeletal muscles and other organs via the circulatory system. Based on the dynamic nature of proteoforms as the basic units of the proteome [12,13,14,15], skeletal muscle proteomics attempts to increase our biochemical and pathophysiological knowledge of protein changes in health and disease [18,19,20,48]. However, since skeletal muscle tissues are heterogeneous in their cellular composition, this biological fact is reflected by the diverse constitution of the accessible muscle proteome [185]. Skeletal muscles contain both tissues that are difficult to homogenize and comprise a large amount of sarcomeric proteins, membrane-associated proteins and relatively insoluble proteins of the ECM, making the proteomic analysis of total extracts a difficult task. Proteome-wide effects on skeletal muscles are routinely studied using cultured muscle cells, human biopsy specimens and muscles derived from animal models [48]. The collection of mass spectrometrically identified and muscle-associated proteins consists of over 10,000 individual species. The members of the core muscle proteome have been established by the systematic survey of various skeletal muscle types [226,227,228,229,230,231,232] and focused analyses of slow versus fast subtypes of myofiber populations [233,234,235,236,237,238,239,240,241,242]. The MS-based analysis of skeletal muscle specimens is routinely carried out with crude total extracts or subcellular fractions using sophisticated biochemical separation methodology. Figure 3 summarizes the subdisciplines involved in the integrative proteomic analysis of the skeletal muscle system.
It is important to stress that changes in non-muscle phenotypes in muscular dystrophies are most likely due to a combination of both organ crosstalk via the circulatory system and intrinsic changes within individual tissues/organs. This is especially relevant to DMD, as it is known that the DMD gene contains several promoters that produce eight different tissue-specific dystrophin isoforms, as outlined in the below section on the genetic basis of dystrophinopathy. The tissue-specific expression of dystrophins can be differentially affected by various mutations in the DMD gene. Thus, not all non-muscle effects are due to organ crosstalk but can be based on mutation-specific alterations in non-muscle tissues.
The isolation of distinct muscle fractions most frequently involves the microdissection of cellular structures or optimized tissue homogenization followed by differential centrifugation, density gradient ultracentrifugation and/or biochemical affinity isolation approaches [48,49,50]. Affinity purification can be carried out with pharmacological agents, immobilized lectins or suitable antibodies. The most intensively studied subcellular fractions derived from myofibers are the sarcolemma, transverse tubules, sarcoplasmic reticulum (longitudinal tubules and terminal cisternae region), triad junctions, mitochondria (outer membrane, contact sites, inner membrane and matrix), nuclei, ribosomes, Golgi apparatus, lysosomes, peroxisomes, proteasome, sarcosol, the sarcomeric acto-myosin apparatus, the auxiliary titin and nebulin filament structures, the intracellular cytoskeletal network, costameres, and the various layers of the ECM, including the basal lamina, endomysium, perimysium and epimysium [213]. Single-myofiber proteomics, which presents a specialized form of single-cell proteomics [44] that is applied to the MS-based analysis of contractile fibers, is becoming increasingly important in the field of basic and applied myology [112,202,203,237,238,239].

4. The Pathoproteomic Profiling of Duchenne Muscular Dystrophy

4.1. The Genetic Basis of Dystrophinopathy

Primary abnormalities in the DMD gene are the underlying cause of dystrophinopathies [28], a group of progressive muscle-wasting diseases that include the severe Duchenne type of muscular dystrophy in early childhood [23,24] and the more benign and later-onset Becker’s muscular dystrophy [243]. This classifies dystrophinopathies as monogenetic diseases that are characterized primarily by chronic muscle wasting due to a main cellular defect in the membrane cytoskeleton. The degeneration of muscle fibers affects almost exclusively males due to the location of the defective gene within the Xp21.2 region on the short arm of the X-chromosome [26]. Diverse types of genetic abnormalities were shown to be associated with dystrophinopathies, including splice site mutations, nonsense point mutations, missense point mutations and mid-intronic mutations, as well as small and large insertions, small and large deletions and large duplications [244,245,246].
The almost complete loss of the full-length dystrophin isoform Dp427-M is the initial trigger that causes progressive myofiber degeneration. However, the 79-exon-spanning DMD gene with its 2.4 million bases has a highly complex genomic organization consisting of several promoter regions that are involved in the production of eight different and tissue-specific dystrophins [23]. Dystrophin proteins are represented by the full-length versions Dp427-M (muscle), Dp427-B (brain) and Dp427-P (Purkinje cells) and the shorter isoforms Dp260-R (retina), Dp140-B/K (brain/kidney), Dp116-S (Schwann cells), Dp71-G (ubiquitous) and Dp45 (nervous system) [247]. This diversity of tissue-specific dystrophin isoforms, which are affected differentially by particular mutations in the DMD gene, and secondary effects of muscle disintegration on other organs cause body-wide alterations in muscular dystrophy [29,30,31] and multi-system dysfunction [32,33,34], as discussed in detail in the below sections.

4.2. Pathoproteomics of Chronic Muscle Wasting in Dystrophinopathy

In skeletal muscle, the almost complete deficiency in the full-length dystrophin protein triggers an unstable membrane cytoskeleton and the collapse of the dystrophin-associated glycoprotein (DGC) complex, consisting of dystroglycans, sarcoglycans, sarcospan, dystrobrevins and syntrophins [248,249,250,251]. The disturbed trans-sarcolemmal linkage, which is usually provided by tight dystrophin/dystroglycan interactions within the DGC [252,253,254] causes the loss of the organizing dystrophin node [254]. In healthy muscles, the dystrophin-containing node of the sarcolemma is defined as the integrating structure of the intracellular cytoskeleton, the central provider of lateral force transmission, the plasmalemmal hub of fiber stabilization and a key point for cellular signaling mechanisms at the myofiber periphery. In muscular dystrophy, the collapse of the dystrophin node results in impaired sarcolemmal integrity, reduced lateral force transmission at weakened costamere structures, and pathophysiological Ca2+ influx into the sarcosol, which in turn triggers an increase in proteolytic degradation of muscle proteins [255,256,257]. The main feature of the cellular pathogenesis of DMD is progressive myonecrosis, which is accompanied by chronic inflammation, reactive myofibrosis and an impaired regenerative capacity due to satellite cell dysfunction, as recently reviewed [258]. Skeletal muscles are characterized by the presence of extremely large proteins, i.e., giant proteins such as titin and nebulin [99], as well as many distinctly hydrophobic membrane proteins [50] and complex layers of mostly insoluble ECM components [259]. This makes proteomic studies of total tissue extracts technically challenging. The extensive listings of proteome-wide changes in dystrophic muscles have previously been published in extensive reviews. These articles have summarized the findings from systematic proteomic surveys using both patient biopsy material and various animal models of dystrophinopathy [260,261,262,263,264,265].
Individual studies have confirmed complex proteomic changes due to myonecrosis and myofibrosis, including altered expression levels of proteins involved in the organization of the cytoskeleton, maintenance of the ECM, energy metabolism, the cellular stress response and the excitation–contraction–relaxation cycle [260,263]. Bioanalytical approaches have employed both top-down proteomics and bottom-up proteomics with crude extracts and select subcellular fractions. The main methods used for the screening of dystrophic muscle specimens included 2D-GE, 2D-DIGE, immuno-precipitation, affinity purification, chemical cross-linking, liquid chromatography, ICAT, SILAC, iTRAQ, MudPIT, MALDI-ToF MS and LC-MS/MS analysis [172,264,265]. Please see the above section on proteomic technology for details on these methods and their analytical advantages versus technical limitations. Proteins with a significant change in abundance were identified in a variety of skeletal muscle types [264]. The most prominent and reproducibly identified proteins include the adenylate kinase isoform AK1, annexins, small heat shock proteins, desmin, vimentin, tubulins, collagens, calsequestrin, B-type lamin, myoferlin, dysferlin, ferritin, carbonic anhydrase isoform CA3, the fatty acid-binding protein FABP3 and various contractile proteins [134,148,180,232,266,267,268,269,270,271,272,273,274,275,276,277,278,279,280,281,282,283,284,285,286,287,288,289,290,291,292]. Independent verification analyses using comparative immunoblotting, enzyme assays, histochemistry and immunofluorescence microscopy were employed to confirm proteome-wide changes in dystrophic skeletal muscles [172].

4.3. Pathoproteomics of Multi-System Dysfunction in Dystrophinopathy

Besides chronic skeletal muscle wasting [23,24] and impaired neuromuscular transmission [293], Duchenne patients suffer from multi-system dysfunction [29,30,31] involving a variety of tissue and organ systems [32,33,34]. These body-wide abnormalities include cardiomyopathy [294,295,296], respiratory failure [297,298,299,300], liver atrophy [301,302], renal failure [303,304,305,306], bladder dysfunction [307,308,309,310] and gastrointestinal complications [311,312,313], as well as bone fragility [314] and scoliosis [315,316,317]. A subset of Duchenne patients suffers from neurological deficiencies that manifest themselves as neurodevelopmental delays, emotional disturbances, mental retardation and behavioral problems [318,319,320,321,322]. The main non-skeletal muscle organ systems that are affected in dystrophinopathy include the following:
  • Central nervous system: cognitive impairments, attention deficit, altered emotions, impaired language, memory deficiencies and altered coordination;
  • Peripheral nervous system: abnormal transmission at nerve–muscle connections;
  • Cardio-respiratory system: late-onset cardiomyopathy, cardio-respiratory syndrome, respiratory insufficiency;
  • Liver: enlargement, steatosis, fibrosis, atrophy and ectopic fat deposition;
  • Renal system: kidney failure, cardio-renal syndrome, hyperfiltration, hypertension and ectopic fat deposition;
  • Bladder: dysfunction of the urinary tract and bladder;
  • Bone: increased risk of bone fragility;
  • Spine: high risk of development of scoliosis;
  • Gastrointestinal system: delayed gastric emptying and pancreatic dysregulation;
  • Immune system: hyperactivity causing chronic inflammation, spleen adaptations.
Although the detailed proteomic analysis of multi-system dysfunction in dystrophinopathy clearly lags behind the exhaustive MS-based analysis of dystrophic skeletal muscles [260,261,262,263,264,265], a small number of studies have been initiated to establish a more comprehensive picture of whole-body effects due to dystrophin deficiency. The pathophysiological status of non-skeletal muscle tissues has been mostly studied with the help of established animal models of dystrophinopathy and has included the investigation of the heart, liver, kidney, stomach/pancreas interface, spleen and brain.
Proteomic surveys of dystrophin-deficient hearts have been carried out by both top-down/gel-based proteomics and bottom-up proteomics [323,324,325]. Cardioproteomics is an established field within the systems biological multi-omics approach to determine the underlying mechanisms of heart disease [326,327,328,329]. Focusing on X-linked muscular dystrophy, biochemical, physiological and proteomic studies have revealed substantial changes in key components of the contractile apparatus, ion-regulatory elements, proteins involved in the maintenance of the cytoskeleton, components that are central to the stabilization of the basal lamina, molecular chaperones that mediate the cellular stress response, and proteins that are associated with oxidative and glycolytic energy metabolism in cardiomyocytes [95,330,331,332,333,334,335,336,337,338]. These alterations in the cardiac protein constellation agree with the cardiomyopathic complications seen in dystrophic patients [294,295,296]. In contrast to skeletal muscles, the DGC is not restricted to the sarcolemma in the heart but also localizes to the transverse tubular membrane system in cardiomyocytes [339]. These cell biological differences are reflected on the biochemical level by a slightly different composition of the dystrophin complexome in voluntary myofibers versus myocardial contractile cells [333,340]. This might explain why laminin is not majorly affected in dystrophic skeletal muscles but was found to be reduced in the basal lamina of dystrophic heart cells [335]. Protein perturbations in the dystrophin-lacking heart can be linked to muscle cell degradation, interstitial fibrosis and inflammation [323]. Importantly, the dystrophinopathy-related dysregulation of the heart and adaptations in the cardiovascular system are probably linked to detrimental changes in the overall circulatory system [30].
A late-onset pathophysiological effect of poor circulation due to a chronically weakened heart could trigger a lack of sufficient oxygen and nutritional supply to organs such as the liver and kidneys [32,33,34]. The proteomic screening of the liver from the mdx-4cv model of dystrophinopathy revealed changes in proteins involved in the metabolism of carbohydrates, fatty acids and amino acids, as well as components that are associated with the cellular stress response, the regulation of ion homeostasis and biotransformation [341]. This agrees with the observed liver abnormalities in Duchenne patients [301,302]. A striking finding was the MS-based demonstration of elevated levels of FABP5, a major member of the large family of fatty acid-binding proteins [342]. Increased expression of the FAPB5 isoform was confirmed by immunoblot analysis and confocal microscopy. Of note, Sudan Black staining labeled fatty deposits in the liver of dystrophic mice. These cellular changes in mdx-4cv hepatocytes agree with high levels of FABP5 and suggest the occurrence of altered patterns of fatty acid transportation and ectopic fat deposition in the liver in DMD [341]. In analogy to hepatic tissue, the proteomic analysis of serum from the same dystrophic mouse mutant was characterized by a high concentration of FABP5 [343,344], making it a potential serum biomarker candidate for the monitoring of hepatic alterations in association with dystrophinopathy [264,265,345].
In addition, the renal system can be severely impaired in some patients suffering from DMD and this might also be linked to abnormal circulation [303,304,305,306]. The proteomic analysis of mdx-4cv kidneys showed similar findings as already described for the liver in dystrophic mice, i.e., elevated levels of distinct fatty acid-binding proteins [346,347]. In the case of the kidneys, the FABP1 isoform was identified by MS-based analysis in association with ectopic fat deposits [346]. These results on the liver FABP5 and kidney FABP1 isoforms do not directly demonstrate a pathophysiological link between the increase in FABP levels and the accumulation of intracellular fat deposits. However, they agree with the disturbed fat metabolism in DMD. The FABP3 isoform is present at high levels in the heart and skeletal muscles, and proteomics has identified it as a robust biomarker of aerobic capacity in muscle [231]. Dystrophic skeletal and cardiac muscle tissues exhibit a drastically reduced concentration of FABP3 [232,274,275,276,277,278,279,290,335], which is mirrored by elevated FABP3 levels in the serum of Duchenne patients and mdx-type mouse models [348,349,350,351]. Hence, the analysis of serum FABP3, FAPB5 and FABP1 isoforms can be useful for evaluating the shedding or release of excess fatty acid-binding proteins from muscle tissues, the liver and kidneys, respectively, in the dystrophic phenotype [342]. Proteomic biomarker discovery for multi-system changes in dystrophinopathy is discussed in more detail in the below section.
An extensive immune response is observed in X-linked muscular dystrophy, causing chronic inflammation in the skeletal musculature [352,353,354] which presents an excellent opportunity for therapeutic interventions [355,356,357,358]. Both, the innate immune system and adaptive immune responses intermingle in a complex relationship in dystrophinopathy [359]. Besides activation of resident macrophages within the skeletal muscular system, cellular invasion by immune cells includes the movement of regulatory T cells, CD4+ T cells, CD8+ T cells, natural killer cells, eosinophils and monocytes into degenerating myofibers as a response to muscular dystrophy [360,361,362,363]. In muscular dystrophy, splenic abnormalities include morphological alterations to lymph nodes in the white pulp region of the spleen and adapted pools of splenic inflammatory monocytes, which are associated with drastically elevated levels of immune cell migration from the splenic reservoir to damaged muscles [364,365]. Since the spleen represents a key secondary lymphoid organ whose biological functions are involved in antigen detection, antibody production and the efficient removal of abnormal erythrocytes [366,367], splenic abnormalities may have body-wide consequences. The linkage between dystrophic skeletal muscles and the lymphoid system was assessed by proteomics [124,368]. Significant changes in the mdx-4cv spleen were identified in proteins that participate in cellular signaling, metabolic pathways and cytoarchitecture [368]. The spleen is probably involved in a pathophysiological crosstalk with dystrophin-deficient myofibers [34].
Gastrointestinal dysfunction in muscular dystrophy [29,311,312,313] was evaluated by an MS-based study focusing on proteome-wide changes at the interface between the pancreas and the muscular stomach wall of the mdx-4cv model of DMD. Lower levels of dystrophin and its associated glycoproteins, as well as laminin, filamin and titin, suggested a loss of cytoskeletal integrity leading to abnormal smooth muscle function in the gut [369]. The below section lists promising biomarker candidates of the above-discussed non-skeletal muscle tissues in X-linked muscular dystrophy.
Psychosocial and psychological care is an integral part of the management of Duchenne patients [31]. Since a sub-group of dystrophic children is afflicted by complex neurodevelopmental and neurological deficiencies, it was of interest to study potential changes in the dystrophin-deficient brain proteome. The mouse brain is frequently used in biochemical, proteomic and cell biological studies in order to characterize the potential involvement of the central nervous system in neuromuscular diseases [370]. Proteomic screening of the mdx-4cv brain revealed a decreased abundance of syntaxin-1B and the syntaxin-binding protein STXBP1, which are involved in synaptic vesicle docking mechanisms at the pre-synaptic membrane and the regulation of neurotransmitter release from neurons [371,372]. Key brain proteins with an increased abundance were identified as the glial fibrillary acidic protein (GFAP), the annexin isoform ANXA5, the neuron-specific enzyme ubiquitin carboxyl-terminal hydrolase isozyme L1 and the neuronal cytomatrix protein bassoon. GFAP is a member of the intermediate filament system and is tightly associated with astrocytes in the central nervous system [373,374,375]. Thus, increased levels of GFAP in the mdx-4cv brain, which were demonstrated by proteomics, comparative immunoblotting and immunofluorescence microscopy [371], strongly suggest the presence of astrogliosis being part of the neurodegenerative process in DMD [34]. However, astrogliosis appears to be a frequent occurrence in the brain in response to general tissue damage [373], as was recently shown by the proteomic analysis of the wobbler mouse model of amyotrophic lateral sclerosis [376]. This reduces the diagnostic usefulness of GFAP as a specific biomarker of muscular dystrophy-related changes in the central nervous system but does not limit its general suitability as an astrogliosis marker protein. Proteomics suggests nevertheless that neuronal disturbances and reactive astrogliosis might play a central role in the molecular pathogenesis of brain abnormalities in dystrophinopathy [371] that may lead to mental retardation, behavioral problems, cognitive impairments, emotional disturbances, attention deficit, impaired language, memory deficiencies and altered coordination in DMD [377,378,379].
Figure 4 gives an overview of the complexity of the multi-system pathology of dystrophinopathy [34] and how MS-based analyses can be used for interproteomic profiling. A crucial aspect of future muscular dystrophy research should be the establishment of a more comprehensive picture of pathophysiological inter-organ crosstalk between the various bodily systems in dystrophic patients. Combing findings from organ proteomics with biofluid proteomics could help us better understand the involvement of (i) skeletal muscle–bone interactions, (ii) the brain–muscle signaling axis, (iii) metabolic integration at the level of liver, fat and muscle, (iv) muscle disintegration in the context of renal dysfunction and gastrointestinal problems, (v) the cardio-respiratory system, and (vi) the immune system. The prediction of the severity of the disease phenotype in individual Duchenne patients and potential effects on the whole body based on specific mutations would be an important future tool for clinical work [380]. The relevance of proteomic analyses is summarized in Figure 4. Interproteomic profiling should lead to a better comprehension of the pathophysiological complexity of muscle-associated changes in combination with multi-system effects for biomarker discovery, as further discussed in the below section.

4.4. Proteomic Biomarkers of Muscular and Multi-System Changes in Dystrophinopathy

Besides testing novel pharmacological strategies [257,381,382,383,384] and immunomodulatory interventions [355,356,357,358,364], a variety of innovative therapeutic approaches are currently evaluated in the field of muscular dystrophy [385,386,387,388,389], including exon skipping [148,390,391,392,393], genomic editing [394,395,396], codon read-through [397], gene replacement with the help of adeno-associated viral vectors [398,399,400,401,402], dystrophin substitution with its autosomal homolog utrophin [403,404,405], and muscle stem cell therapy [406,407,408,409]. Hence, for the optimum pre-clinical testing of new therapies, the clinical evaluation of diverse patient cohorts during the various phases of clinal studies/trials, and long-term therapeutic monitoring, robust and specific biomarkers for the routine screening of the status of dystrophic patients are required [144,264,265,410,411]. Ideal protein biomarkers would be measurable in a non-invasive, or at least minimally invasive, way and be suitable for repeated sampling procedures [325,345,410,411,412].
Importantly, biomarker detection should not be overly influenced by age, gender, ethnicity, circadian rhythm, seasonal impact, co-morbidities and supportive treatments. Of note, the determination of biomarkers should not be susceptible to the generation of high levels of false positives and false negatives. The assay system should be optimized for a proper balance between specificity and sensitivity to measure the biomolecule of interest. The clinical application of a biomarker signature, rather than the usage of a single and often not completely reliable marker molecule, can be advantageous to cover more than one particular aspect of a complex pathophysiological process that may considerably change over time [413]. Thus, biomarkers that are highly suitable for initial screening and differential diagnostic purposes might not be ideal for prognosis, the testing of potentially adverse side effects and extended periods of therapeutic monitoring.
The main subtypes of proteomic biomarkers include the following:
  • Susceptibility markers: for risk assessment of potential disease initiation;
  • Diagnostic markers: for initial detection of a specific disease process;
  • Prognostic markers: for envisaging disease progression and adverse clinical events;
  • Predictive markers: for differential patient screening and individual sensitivities;
  • Pharmacodynamic markers: for assessing the body’s response to a specific treatment;
  • Therapeutic monitoring markers: for the repeated assessment of disease status following therapeutic intervention;
  • Safety-related biomarkers: for determining potential adverse side effects.
Of special interest are biofluid markers that can be easily accessed in a simple, cost-effective, safe, non-invasive and pain-free way, such as biomolecules that are present in sufficient abundance in urine or saliva, or at least be testable in a minimally invasive procedure using serum/plasma-associated biomarkers. The routine usage of these types of disease markers would have several advantages over invasive muscle biopsy procedures. Although the histological and histochemical analysis of muscle tissue biopsies is highly useful for determining the various cell biological aspects of pathological changes, the potential occurrence of fiber type shifting, reactive mechanisms such as fibrosis and alterations in cellular components of interest [169,170,171,172], needle or open biopsy procedures are often associated with higher costs, more complex harvesting and handling of tissue specimens, patient anxiety, tissue damage triggering inflammatory responses, potential infection and usually a lack of capacity for repeated sampling approaches [414,415,416,417].
Routinely used general muscle damage markers, assayed alone or in combination, include the muscle-specific isoform of creatine kinase, the carbonic anhydrase isoform CA3, troponin subunit TnI, myosin light chain MLC1, fatty acid-binding protein FABP3, myoglobin, aspartate transaminase, enolase, aldolase, lactate dehydrogenase, alanine aminotransaminase and hydroxybutyrate dehydrogenase [413,418,419,420]. An altered concentration of these types of proteins is often observed after crush injury or strenuous physical exercise but also in association with a variety of neuromuscular diseases, autoimmune disorders, toxic insults, body-wide inflammation, infectious diseases, sepsis, alcoholism and drug abuse. It is therefore imperative to identify more specific biomarkers of dystrophinopathy that are not changed by other types of physiological or pathobiochemical challenges to the neuromuscular system.
Figure 5 summarizes the findings from major biochemical and proteomic studies aimed at the identification of biomarker candidates that are characteristic of both tissue-related changes and the extent of the release of muscle proteins from dystrophic myofibers and their surrounding tissues and extracellular environment. The full names of abbreviated protein species are listed in the figure legend. The upper part of the diagram lists skeletal muscle tissue markers related to (i) the disintegration of sarcolemmal integrity and initiation of myonecrosis (members of the dystrophin complex, such as dystrophin, dystroglycans and sarcoglycans), (ii) abnormal Ca2+ homeostasis triggered by the influx of ions into the sarcosol through the damaged sarcolemma membrane and impaired luminal Ca2+ buffering (Ca2+-binding proteins calsequestrin and sarcalumenin), (iii) cycles of tissue regeneration and muscle repair (myoferlin, dysferlin, annexins, CD34 and cadherin-13), (iv) intracellular compensation of dystrophin deficiency by up-regulation of other types of cytoskeletal components (vimentin, desmin, and tubulin chains alpha-1B, alpha-1C, alpha-8 and beta-2A), (v) macrophage invasion and the triggering of chronic inflammation (cathepsin B, secreted phosphoprotein SPP1 and lysozyme), (vi) an increase in the cellular stress response by elevation of the abundance of small heat shock proteins (αB-crystallin/HspB5 and cardiovascular cvHsp/HspB7) and (vii) extensive reactive myofibrosis (matricellular protein periostin, collagens such as COL6, fibronectin, dermatopontin, biglycan, lumican and asporin).
Biochemical and proteomic studies have identified a large panel of potential muscle-derived biofluid markers, both as intact proteins or peptide fragments [421,422,423,424,425,426]. As listed in the lower panel of Figure 5, the myomesin isoform MYOM3, a marker component of the M-line in sarcomeres, was identified in the form of peptide fragments in serum samples [427] and various titin fragments were clearly detected in urine from both Duchenne patients and animal models of dystrophinpathy [428,429,430,431,432]. The half-sarcomere spanning protein titin of 3.8 MDa is the largest known protein in skeletal muscles [99] and its detection in urine indicates a massive disintegration of the auxiliary filaments of the sarcomere structure in dystrophinopathy [431]. The list of biofluid markers ranges from metabolic and modulating enzymes (adenylate kinase, creatine kinase, carbonic anhydrase, matrix metallo-proteinases, aspartate transaminase, alanine aminotransaminase, lactate dehydrogenase, malate dehydrogenase), metabolite transporters (fatty acid-binding proteins, myoglobin), sarcomeric proteins (troponins, myomesin, myosin light chains, titin fragments), signaling molecules (interleukins) to ECM proteins (fibronectin) (Figure 5). The passive shedding of large numbers of skeletal muscle proteins into the circulatory systems is indicative of the extent of sarcolemmal disintegration due to the collapse of the DGC. The overall changes in these biomarkers demonstrate the complexity of the cellular pathogenesis that is triggered by dystrophin deficiency and causes myofiber degeneration and loss of sarcolemmal integrity, followed by chronic inflammation and massive fibrosis of dystrophic muscles [422]. In our opinion, the most suitable minimally invasive and biofluid-associated biomarkers of DMD are represented by amino-terminal and carboxy-terminal TTN fragments of titin in urine, kallikrein KLK1 in saliva and the combination of CA3/FABP3/MYOM3/MDH2 in plasma/serum samples.
Multi-system changes in DMD are reflected by both proteomic changes in particular organs other than skeletal muscles and in biofluid markers that are characteristic of alterations in the heart, liver and kidneys. As already outlined above, striking proteomic changes in the brain included an increase in GFAP, an established marker of astrogliosis, as well as annexins and vimentin [371]. In contrast to dystrophic skeletal muscles that exhibited no major changes in laminin, this major component of the basal lamina was shown to be drastically reduced in a dystrophin-deficient heart [335]. The proteomic screening of the liver and kidney showed ectopic fat deposition in conjunction with elevated levels of the fatty acid-binding proteins FABP5 [341,342] and FABP1 [342,346], respectively. The analysis of the dystrophin-lacking stomach/pancreas interface revealed changes in the ECM protein repetin [369]. The proteome of the spleen was found to show increased levels of MMP9 and TGM2 in the dystrophic phenotype [368]. Figure 5 lists non-muscle biofluid markers that can be useful in screening dystrophinopathy-associated changes in the liver (glutamate dehydrogenase isoform GLUD1 and haptoglobin) and the heart (cardiac troponin subunit cTnI), as well as in carrying out the functional evaluation of the kidneys (cystatin-C/CST3 and uromodulin). Measurement of the serum carbonic anhydrase isoform CA3 alone versus the ratio of serum myoglobin to CA3 can be used to determine the loss of integrity of dystrophic skeletal muscles versus dystrophin-deficient heart muscle [423]. An interesting new source of non-invasive biomarkers is represented by the saliva proteome [412], whose analysis showed elevated levels of the kallikrein isoform Klk1 in the mdx-4cv model of DMD [344,433]. How future multi-omics research initiatives can build on these proteomic findings and establish a more comprehensive pathophysiological picture of dystrophinopathy is briefly outlined in the below section.

4.5. Integromics: Systems Biological Multi-Omics Analysis of Dystrophinopathy

For the future advancement of precision medicine and individualized patient treatments, the implementation of multi-omics biomarker signatures would be an advantage for the detailed evaluation of complex human disease initiation and progression [434]. Ideally, a wide range of multi-modal omics markers at the levels of the genome, transcriptome, epigenome, proteome, metabolome and cytome would be used in combination to decisively enhance the accuracy of diagnostic, prognostic and therapeutic monitoring procedures [435]. The cytome can be defined as the entire collection of dynamic cellular processes, incorporating both structural and functional parameters, that form the basis of all biochemical and physiological processes in the body [436,437,438]. In the field of neurological and neuromuscular disorders, this would be a strategic step forward to evolve evidence-based medicine to the next level of stratified approaches and establish personalized medical therapies via translational neuroscience [439].
In relation to studying the downstream effects of dystrophin deficiency, a holistic systems biological analysis of the complex pathogenesis of DMD would greatly enhance our insights into organ crosstalk and encompass an integrative multi-omics approach. This integromics strategy would ideally consist of genomics [440,441,442], transcriptomics [443,444,445], top-down proteomics [17,59,60,61], bottom-up proteomics [46,107], subproteomics [108,109,110,213], the proteomic evaluation of PTMs [117,291,446,447], metabolomics [448,449], lipidomics [450,451,452], glycomics [453], immunomics [454], secretomics [218,219,220,221,455] and high-throughput cytomics [158,172,456,457], as summarized in Figure 6.
Importantly, the integration of proteomic data sets from both bottom-up and top-down approaches would be extremely helpful in identifying and characterizing specific proteoforms in highly complex tissue systems [458,459]. This could especially help us better understand the enormous complexity of aberrant cellular signaling events that occur in DMD [460]. The wider application of spatial single-myofiber MS analysis would be extremely helpful with protein biochemical studies using techniques such as deep visual proteomics [461]. This method integrates high-content imaging with laser micro-dissection and multiplexed MS-based analyses at the single-cell level and has therefore been termed ‘single-cell Deep Visual Proteomics’ (scDVP) [462]. Besides metagenomics, an additional field of interest for studying the genome is epigenomics [463], which focuses on the systematic analysis of molecular modifications at the level of DNA that may alter the regulation of gene activity but are mitotically stable and are independent of the DNA sequence [464,465,466]. The determination of histone modifications, DNA methylation and the generation of modulatory non-coding RNAs can generate crucial proteogenomic data with considerable relevance to skeletal muscle development, repair and physiological functioning [467,468,469] and the treatment of muscular dystrophy [470,471,472].
Multi-omics approaches have a great potential to improve our understanding of complex human disease mechanisms [473] and establish systems biological concepts [474], including the systems biology of skeletal muscles [475]. The application of multi-omics has already been used to study crucial aspects of skeletal muscle cell biology in health and disease [476,477,478,479] and been applied to certain aspects of the field of dystrophinopathy research, including the integrative screening of dystrophic animal models [277,480,481,482], the evaluation of immune responses in muscular dystrophy [483], myogenic remodeling by human pluripotent stem cells [484], astrocyte-related abnormalities [485] and dystrophinopathy-associated cardiomyopathy [324,331]. The main techniques used for proteomics-centric and multi-omics studies have been recently reviewed by Rajczewski et al. [486]. Combining findings from these types of molecular and cellular analyses will be extremely helpful in predicting the trajectory of disease progression in clinical subtypes of DMD and determining the potential influence of environmental factors and lifestyle. To achieve a maximum yield of data from integrative analyses [35], multi-omics approaches will be assisted by employing big data analytic tools and using optimized machine learning and artificial intelligence approaches [487,488,489,490,491]. A crucial aspect of the development of novel therapies to treat dystrophinopathies is the elucidation of the underlying mechanisms that generate mild forms of DMD [492] or naturally protective phenotypes, such as the spared muscular systems of the tongue, intrinsic laryngeal muscles and extraocular muscles [196,493,494]. Multi-omics analyses of these specialized skeletal muscles that are only mildly affected, despite the fact of dystrophin deficiency, might identify novel targets for therapeutic intervention.

5. Conclusions

In this perspective article, we have attempted to address the question of how MS-based proteomic analyses can be employed to better comprehend the pathophysiological complexity and multi-systems involvement of DMD. Integrating findings from top-down proteomics, bottom-up proteomics and subproteomics could be used to establish a more precise picture of the molecular and cellular pathogenesis of this monogenetic disease of the neuromuscular system. Current limitations of MS-based studies of skeletal muscles are based on the fact that only the technically accessible proteome can be studied. Thus, the expansion of the measurable part of the total muscle proteome through improved tissue extraction and protein separation methodology would decisively widen the scope of muscular dystrophy research. Importantly, proteomics represents an essential method of modern systems biology, which is central to the multi-modal omics analysis of complex pathobiochemical mechanisms. The integration of data generation from traditional biological disciplines, such as histochemistry, physiology, biochemistry and molecular biology, with findings from high-throughput and large-scale bioanalytical approaches, including genomics, transcriptomics, proteomics and metabolomics, promises a genuine systems biological understanding of dystrophinopathies. In the future, multi-omics will be instrumental for a more detailed determination of the pathogenic mechanisms and multi-system dysfunction due to dystrophin deficiency. The establishment of a broad and reliable biomarker signature at all levels of biological organization, ranging from the genome to the physiome, will improve screening procedures, differential diagnostics and prognostic predictions, as well as expand the systematic identification of new therapeutic targets to treat dystrophinopathy. In the long term, if integrated proteomics and multi-omics approaches are properly established for studying detailed mechanisms of DMD and biomarkers are verified for their clinical suitability, this biomedical information can be consolidated to have a considerable clinical impact.

Author Contributions

Conceptualization, P.D., C.T., E.N., D.S. and K.O.; writing—original draft preparation, P.D. and K.O.; writing—review and editing, P.D., C.T., E.N., 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 and Campus France (Ulysses/2024/5), Science Foundation Ireland Infrastructure Award SFI-12/RI/2346/3, the Agence Nationale de la Recherche (ANR, «EpiMUSE» grant # ANR-22-CE14-0068), INSERM, Sorbonne University (project Emergence SU-16-R-EMR-60 Fib-Cell), and Association Institut de Myology and the Fondation Recherche Médicale (EQUIPE FRM EQU201903007784).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We would like to thank our many national and international colleagues for their support of the ongoing pathoproteomic analysis of muscular dystrophy.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Börner, K.; Teichmann, S.A.; Quardokus, E.M.; Gee, J.C.; Browne, K.; Osumi-Sutherland, D.; Herr, B.W., 2nd; Bueckle, A.; Paul, H.; Haniffa, M.; et al. Anatomical structures, cell types and biomarkers of the Human Reference Atlas. Nat. Cell Biol. 2021, 23, 1117–1128. [Google Scholar] [CrossRef]
  2. Bianconi, E.; Piovesan, A.; Facchin, F.; Beraudi, A.; Casadei, R.; Frabetti, F.; Vitale, L.; Pelleri, M.C.; Tassani, S.; Piva, F.; et al. An estimation of the number of cells in the human body. Ann. Hum. Biol. 2013, 40, 463–471. [Google Scholar] [CrossRef] [PubMed]
  3. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [PubMed]
  4. Hatton, I.A.; Galbraith, E.D.; Merleau, N.S.C.; Miettinen, T.P.; Smith, B.M.; Shander, J.A. The human cell count and size distribution. Proc. Natl. Acad. Sci. USA 2023, 120, e2303077120. [Google Scholar] [CrossRef] [PubMed]
  5. Ho, B.; Baryshnikova, A.; Brown, G.W. Unification of Protein Abundance Datasets Yields a Quantitative Saccharomyces cerevisiae Proteome. Cell Syst. 2018, 6, 192–205.e3. [Google Scholar] [CrossRef]
  6. 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]
  7. 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.e19. [Google Scholar] [CrossRef]
  8. 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]
  9. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef]
  10. Pertea, M.; Salzberg, S.L. Between a chicken and a grape: Estimating the number of human genes. Genome Biol. 2010, 11, 206. [Google Scholar] [CrossRef]
  11. Amaral, P.; Carbonell-Sala, S.; De La Vega, F.M.; Faial, T.; Frankish, A.; Gingeras, T.; Guigo, R.; Harrow, J.L.; Hatzigeorgiou, A.G.; Johnson, R.; et al. The status of the human gene catalogue. Nature 2023, 622, 41–47. [Google Scholar] [CrossRef] [PubMed]
  12. 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]
  13. 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] [PubMed]
  14. 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]
  15. Carbonara, K.; Andonovski, M.; Coorssen, J.R. Proteomes Are of Proteoforms: Embracing the Complexity. Proteomes 2021, 9, 38. [Google Scholar] [CrossRef]
  16. 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]
  17. 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.; Consortium for Top-Down Proteomics. The Human Proteoform Project: Defining the human proteome. Sci. Adv. 2021, 7, eabk0734. [Google Scholar] [CrossRef]
  18. 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]
  19. 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]
  20. 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]
  21. Dowling, J.J.; Weihl, C.C.; Spencer, M.J. Molecular and cellular basis of genetically inherited skeletal muscle disorders. Nat. Rev. Mol. Cell Biol. 2021, 22, 713–732. [Google Scholar] [CrossRef] [PubMed]
  22. Thompson, R.; Spendiff, S.; Roos, A.; Bourque, P.R.; Warman Chardon, J.; Kirschner, J.; Horvath, R.; Lochmüller, H. Advances in the diagnosis of inherited neuromuscular diseases and implications for therapy development. Lancet Neurol. 2020, 19, 522–532. [Google Scholar] [CrossRef]
  23. Duan, D.; Goemans, N.; Takeda, S.; Mercuri, E.; Aartsma-Rus, A. Duchenne muscular dystrophy. Nat. Rev. Dis. Primers 2021, 7, 13. [Google Scholar] [CrossRef]
  24. Bez Batti Angulski, A.; Hosny, N.; Cohen, H.; Martin, A.A.; Hahn, D.; Bauer, J.; Metzger, J.M. Duchenne muscular dystrophy: Disease mechanism and therapeutic strategies. Front. Physiol. 2023, 14, 1183101. [Google Scholar] [CrossRef] [PubMed]
  25. Hoffman, E.P. The discovery of dystrophin, the protein product of the Duchenne muscular dystrophy gene. FEBS J. 2020, 287, 3879–3887. [Google Scholar] [CrossRef]
  26. Roberts, R.G. Dystrophin, its gene, and the dystrophinopathies. Adv. Genet. 1995, 33, 177–231. [Google Scholar] [CrossRef]
  27. Fortunato, F.; Farnè, M.; Ferlini, A. The DMD gene and therapeutic approaches to restore dystrophin. Neuromuscul. Disord. 2021, 31, 1013–1020. [Google Scholar] [CrossRef]
  28. Mercuri, E.; Bönnemann, C.G.; Muntoni, F. Muscular dystrophies. Lancet 2019, 394, 2025–2038. [Google Scholar] [CrossRef]
  29. Birnkrant, D.J.; Bushby, K.; Bann, C.M.; Apkon, S.D.; Blackwell, A.; Brumbaugh, D.; Case, L.E.; Clemens, P.R.; Hadjiyannakis, S.; Pandya, S.; et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: Diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018, 17, 251–267. [Google Scholar] [CrossRef]
  30. Birnkrant, D.J.; Bushby, K.; Bann, C.M.; Alman, B.A.; Apkon, S.D.; Blackwell, A.; Case, L.E.; Cripe, L.; Hadjiyannakis, S.; Olson, A.K.; et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: Respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. 2018, 17, 347–361. [Google Scholar] [CrossRef]
  31. Birnkrant, D.J.; Bushby, K.; Bann, C.M.; Apkon, S.D.; Blackwell, A.; Colvin, M.K.; Cripe, L.; Herron, A.R.; Kennedy, A.; Kinnett, K.; et al. Diagnosis and management of Duchenne muscular dystrophy, part 3: Primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 2018, 17, 445–455. [Google Scholar] [CrossRef] [PubMed]
  32. Strehle, E.M.; Straub, V. Recent advances in the management of Duchenne muscular dystrophy. Arch. Dis. Child. 2015, 100, 1173–1177. [Google Scholar] [CrossRef] [PubMed]
  33. Messina, S.; Vita, G.L. Clinical management of Duchenne muscular dystrophy: The state of the art. Neurol. Sci. 2018, 39, 1837–1845. [Google Scholar] [CrossRef] [PubMed]
  34. Ohlendieck, K.; Swandulla, D. Complexity of skeletal muscle degeneration: Multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch. 2021, 473, 1813–1839. [Google Scholar] [CrossRef] [PubMed]
  35. 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] [PubMed]
  36. Aebersold, R.; Mann, M. Mass-spectrometric exploration of proteome structure and function. Nature 2016, 537, 347–355. [Google Scholar] [CrossRef]
  37. Mirza, S.P.; Olivier, M. Methods and approaches for the comprehensive characterization and quantification of cellular proteomes using mass spectrometry. Physiol. Genom. 2008, 33, 3–11. [Google Scholar] [CrossRef] [PubMed]
  38. Martinez-Val, A.; Guzmán, U.H.; Olsen, J.V. Obtaining Complete Human Proteomes. Annu. Rev. Genom. Hum. Genet. 2022, 23, 99–121. [Google Scholar] [CrossRef]
  39. 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]
  40. 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]
  41. Wilkins, M.R.; Pasquali, C.; Appel, R.D.; Ou, K.; Golaz, O.; Sanchez, J.C.; Yan, J.X.; Gooley, A.A.; Hughes, G.; Humphery-Smith, I.; et al. From proteins to proteomes: Large scale protein identification by two-dimensional electrophoresis and amino acid analysis. Biotechnology 1996, 14, 61–65. [Google Scholar] [CrossRef] [PubMed]
  42. Vistain, L.F.; Tay, S. Single-Cell Proteomics. Trends Biochem. Sci. 2021, 46, 661–672. [Google Scholar] [CrossRef] [PubMed]
  43. 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]
  44. 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] [PubMed]
  45. Habeck, T.; Lermyte, F. Seeing the complete picture: Proteins in top-down mass spectrometry. Essays Biochem. 2023, 67, 283–300. [Google Scholar] [CrossRef]
  46. Miller, R.M.; Smith, L.M. Overview and considerations in bottom-up proteomics. Analyst 2023, 148, 475–486. [Google Scholar] [CrossRef]
  47. 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]
  48. Dowling, P.; Swandulla, S.; Ohlendieck, K. Mass spectrometry-based proteomic technology and its application to study skeletal muscle cell biology. Cells 2023, 12, 2560. [Google Scholar] [CrossRef]
  49. 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]
  50. 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]
  51. Yates, J.R., 3rd. Recent technical advances in proteomics. F1000Research 2019, 8, 351. [Google Scholar] [CrossRef] [PubMed]
  52. Matthiesen, R.; Carvalho, A.S. Methods and Algorithms for Quantitative Proteomics by Mass Spectrometry. Methods Mol. Biol. 2020, 2051, 161–197. [Google Scholar] [CrossRef]
  53. Rozanova, S.; Barkovits, K.; Nikolov, M.; Schmidt, C.; Urlaub, H.; Marcus, K. Quantitative Mass Spectrometry-Based Proteomics: An Overview. Methods Mol. Biol. 2021, 2228, 85–116. [Google Scholar] [CrossRef]
  54. 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] [PubMed]
  55. Oliveira, B.M.; Coorssen, J.R.; Martins-de-Souza, D. 2DE: The phoenix of proteomics. J. Proteom. 2014, 104, 140–150. [Google Scholar] [CrossRef] [PubMed]
  56. Westermeier, R. 2D gel-based Proteomics: There’s life in the old dog yet. Arch. Physiol. Biochem. 2016, 122, 236–237. [Google Scholar] [CrossRef]
  57. 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]
  58. 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]
  59. Murphy, S.; Dowling, P.; Ohlendieck, K. Comparative Skeletal Muscle Proteomics Using Two-Dimensional Gel Electrophoresis. Proteomes 2016, 4, 27. [Google Scholar] [CrossRef]
  60. Dowling, P.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Characterization of Contractile Proteins from Skeletal Muscle Using Gel-Based Top-Down Proteomics. Proteomes 2019, 7, 25. [Google Scholar] [CrossRef]
  61. Zabel, C.; Klose, J. High-resolution large-gel 2DE. Methods Mol. Biol. 2009, 519, 311–338. [Google Scholar] [CrossRef] [PubMed]
  62. Friedman, D.B.; Hoving, S.; Westermeier, R. Isoelectric focusing and two-dimensional gel electrophoresis. Methods Enzymol. 2009, 463, 515–540. [Google Scholar] [CrossRef]
  63. 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]
  64. Rabilloud, T.; Lelong, C. Two-dimensional gel electrophoresis in proteomics: A tutorial. J. Proteom. 2011, 74, 1829–1841. [Google Scholar] [CrossRef] [PubMed]
  65. 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]
  66. 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]
  67. Agrawal, G.K.; Thelen, J.J. A high-resolution two dimensional Gel- and Pro-Q DPS-based proteomics workflow for phosphoprotein identification and quantitative profiling. Methods Mol. Biol. 2009, 527, 3–19. [Google Scholar] [CrossRef]
  68. Lourenço Dos Santos, S.; Baraibar, M.A.; Lundberg, S.; Eeg-Olofsson, O.; Larsson, L.; Friguet, B. Oxidative proteome alterations during skeletal muscle ageing. Redox Biol. 2015, 5, 267–274. [Google Scholar] [CrossRef]
  69. Rodríguez-Vázquez, R.; Mouzo, D.; Zapata, C. Phosphoproteome Analysis Using Two-Dimensional Electrophoresis Coupled with Chemical Dephosphorylation. Foods 2022, 11, 3119. [Google Scholar] [CrossRef]
  70. Zimny-Arndt, U.; Schmid, M.; Ackermann, R.; Jungblut, P.R. Classical proteomics: Two-dimensional electrophoresis/MALDI mass spectrometry. Methods Mol. Biol. 2009, 492, 65–91. [Google Scholar] [CrossRef]
  71. Reed, P.W.; Densmore, A.; Bloch, R.J. Optimization of large gel 2D electrophoresis for proteomic studies of skeletal muscle. Electrophoresis 2012, 33, 1263–1270. [Google Scholar] [CrossRef]
  72. Noaman, N.; Coorssen, J.R. Coomassie does it (better): A Robin Hood approach to total protein quantification. Anal. Biochem. 2018, 556, 53–56. [Google Scholar] [CrossRef] [PubMed]
  73. 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]
  74. Chevalier, F. Standard Dyes for Total Protein Staining in Gel-Based Proteomic Analysis. Materials 2010, 3, 4784–4792. [Google Scholar] [CrossRef] [PubMed]
  75. 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]
  76. Sundaram, P. Protein Stains and Applications. In Protein Gel Detection and Imaging; Methods in Molecular Biology; Humana Press: New York, NY, USA, 2018; Volume 1853, pp. 1–14. [Google Scholar] [CrossRef]
  77. 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]
  78. 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]
  79. 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]
  80. Steinberger, B.; Mayrhofer, C. Principles and examples of gel-based approaches for phosphoprotein analysis. Methods Mol. Biol. 2015, 1295, 305–321. [Google Scholar] [CrossRef]
  81. Murphy, S.; Ohlendieck, K. Protein Digestion for 2D-DIGE Analysis. Methods Mol. Biol. 2023, 2596, 339–349. [Google Scholar] [CrossRef]
  82. 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]
  83. Tsiatsiani, L.; Heck, A.J. Proteomics beyond trypsin. FEBS J. 2015, 282, 2612–2626. [Google Scholar] [CrossRef] [PubMed]
  84. 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]
  85. Dau, T.; Bartolomucci, G.; Rappsilber, J. Proteomics Using Protease Alternatives to Trypsin Benefits from Sequential Digestion with Trypsin. Anal. Chem. 2020, 92, 9523–9527. [Google Scholar] [CrossRef] [PubMed]
  86. Minden, J.S.; Dowd, S.R.; Meyer, H.E.; Stühler, K. Difference gel electrophoresis. Electrophoresis 2009, 30, S156–S161. [Google Scholar] [CrossRef] [PubMed]
  87. 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] [PubMed]
  88. 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]
  89. Gargan, S.; Ohlendieck, K. Sample Preparation and Protein Determination for 2D-DIGE Proteomics. Methods Mol. Biol. 2023, 2596, 325–337. [Google Scholar] [CrossRef]
  90. Ohlendieck, K. Top-Down Proteomics and Comparative 2D-DIGE Analysis. Methods Mol. Biol. 2023, 2596, 19–38. [Google Scholar] [CrossRef]
  91. Tonge, R.; Shaw, J.; Middleton, B.; Rowlinson, R.; Rayner, S.; Young, J.; Pognan, F.; Hawkins, E.; Currie, I.; Davison, M. Validation and development of fluorescence two-dimensional differential gel electrophoresis proteomics technology. Proteomics 2001, 1, 377–396. [Google Scholar] [CrossRef]
  92. Marouga, R.; David, S.; Hawkins, E. The development of the DIGE system: 2D fluorescence difference gel analysis technology. Anal. Bioanal. Chem. 2005, 382, 669–678. [Google Scholar] [CrossRef]
  93. Ohlendieck, K. Comparative 3-Sample 2D-DIGE Analysis of Skeletal Muscles. Methods Mol. Biol. 2023, 2596, 127–146. [Google Scholar] [CrossRef]
  94. Dowling, P. DIGE Analysis Software and Protein Identification Approaches. Methods Mol. Biol. 2023, 2596, 39–50. [Google Scholar] [CrossRef]
  95. Murphy, S.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Bioinformatic Analysis of the Subproteomic Profile of Cardiomyopathic Tissue. Methods Mol. Biol. 2023, 2596, 377–395. [Google Scholar] [CrossRef]
  96. Greengauz-Roberts, O.; Stöppler, H.; Nomura, S.; Yamaguchi, H.; Goldenring, J.R.; Podolsky, R.H.; Lee, J.R.; Dynan, W.S. Saturation labeling with cysteine-reactive cyanine fluorescent dyes provides increased sensitivity for protein expression profiling of laser-microdissected clinical specimens. Proteomics 2005, 5, 1746–1757. [Google Scholar] [CrossRef]
  97. 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]
  98. 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]
  99. 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]
  100. Hirabayashi, T. Agarose isoelectric focusing for the detection of many isoforms and high molecules in muscle protein analysis. Electrophoresis 2000, 21, 446–451. [Google Scholar] [CrossRef]
  101. Oh-Ishi, M.; Maeda, T. Disease proteomics of high-molecular-mass proteins by two-dimensional gel electrophoresis with agarose gels in the first dimension (Agarose 2-DE). J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2007, 849, 211–222. [Google Scholar] [CrossRef]
  102. Nakagawa, M.; Tomioka, Y.; Sakuma, C.; Kurosawa, Y.; Shibata, T.; Arakawa, T.; Akuta, T. Development of a novel two-dimensional gel electrophoresis protocol with agarose native gel electrophoresis. Electrophoresis 2023, 44, 1446–1460. [Google Scholar] [CrossRef]
  103. 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]
  104. 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] [PubMed]
  105. 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]
  106. 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]
  107. 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] [PubMed]
  108. Drissi, R.; Dubois, M.L.; Boisvert, F.M. Proteomics methods for subcellular proteome analysis. FEBS J. 2013, 280, 5626–5634. [Google Scholar] [CrossRef]
  109. Lee, Y.H.; Tan, H.T.; Chung, M.C. Subcellular fractionation methods and strategies for proteomics. Proteomics 2010, 10, 3935–3956. [Google Scholar] [CrossRef]
  110. Plöscher, M.; Granvogl, B.; Reisinger, V.; Masanek, A.; Eichacker, L.A. Organelle proteomics. Methods Mol. Biol. 2009, 519, 65–82. [Google Scholar] [CrossRef] [PubMed]
  111. Bennett, H.M.; Stephenson, W.; Rose, C.M.; Darmanis, S. Single-cell proteomics enabled by next-generation sequencing or mass spectrometry. Nat. Methods 2023, 20, 363–374. [Google Scholar] [CrossRef]
  112. 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] [PubMed]
  113. 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]
  114. Duong, V.A.; Lee, H. Bottom-Up Proteomics: Advancements in Sample Preparation. Int. J. Mol. Sci. 2023, 24, 5350. [Google Scholar] [CrossRef] [PubMed]
  115. 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]
  116. Zougman, A.; Selby, P.J.; Banks, R.E. Suspension trapping (STrap) sample preparation method for bottom-up proteomics analysis. Proteomics 2014, 14, 1006. [Google Scholar] [CrossRef] [PubMed]
  117. 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]
  118. 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]
  119. 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]
  120. Stasyk, T.; Huber, L.A. DIGE-Based Phosphoproteomic Analysis. Methods Mol. Biol. 2023, 2596, 97–104. [Google Scholar] [CrossRef]
  121. Ino, Y.; Kinoshita, E.; Kinoshita-Kikuta, E.; Akiyama, T.; Nakai, Y.; Nishino, K.; Osada, M.; Ryo, A.; Hirano, H.; Koike, T.; et al. Evaluation of four phosphopeptide enrichment strategies for mass spectrometry-based proteomic analysis. Proteomics 2022, 22, e2100216. [Google Scholar] [CrossRef]
  122. Sun, M.; Zhang, X. Current methodologies in protein ubiquitination characterization: From ubiquitinated protein to ubiquitin chain architecture. Cell Biosci. 2022, 12, 126. [Google Scholar] [CrossRef] [PubMed]
  123. Duong, V.A.; Park, J.M.; Lee, H. Review of Three-Dimensional Liquid Chromatography Platforms for Bottom-Up Proteomics. Int. J. Mol. Sci. 2020, 21, 1524. [Google Scholar] [CrossRef]
  124. 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]
  125. 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] [PubMed]
  126. 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] [PubMed]
  127. Ahmad, R.; Budnik, B. A review of the current state of single-cell proteomics and future perspective. Anal. Bioanal. Chem. 2023, 415, 6889–6899. [Google Scholar] [CrossRef] [PubMed]
  128. 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]
  129. 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]
  130. Deng, J.; Erdjument-Bromage, H.; Neubert, T.A. Quantitative Comparison of Proteomes Using SILAC. Curr. Protoc. Protein Sci. 2019, 95, e74. [Google Scholar] [CrossRef]
  131. 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]
  132. 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]
  133. 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]
  134. Rayavarapu, S.; Coley, W.; Cakir, E.; Jahnke, V.; Takeda, S.; Aoki, Y.; Grodish-Dressman, H.; Jaiswal, J.K.; Hoffman, E.P.; Brown, K.J.; et al. Identification of disease specific pathways using in vivo SILAC proteomics in dystrophin deficient mdx mouse. Mol. Cell. Proteom. 2013, 12, 1061–1073. [Google Scholar] [CrossRef]
  135. Alayi, T.D.; Tawalbeh, S.M.; Ogundele, M.; Smith, H.R.; Samsel, A.M.; Barbieri, M.L.; Hathout, Y. Tandem Mass Tag-Based Serum Proteome Profiling for Biomarker Discovery in Young Duchenne Muscular Dystrophy Boys. ACS Omega 2020, 5, 26504–26517. [Google Scholar] [CrossRef]
  136. Jünger, M.A.; Aebersold, R. Mass spectrometry-driven phosphoproteomics: Patterning the systems biology mosaic. Wiley Interdiscip. Rev. Dev. Biol. 2014, 3, 83–112. [Google Scholar] [CrossRef] [PubMed]
  137. Deng, N.; Zhang, J.; Zong, C.; Wang, Y.; Lu, H.; Yang, P.; Wang, W.; Young, G.W.; Wang, Y.; Korge, P.; et al. Phosphoproteome analysis reveals regulatory sites in major pathways of cardiac mitochondria. Mol. Cell. Proteom. 2011, 10, S1–S14. [Google Scholar] [CrossRef]
  138. Kitata, R.B.; Yang, J.C.; Chen, Y.J. Advances in data-independent acquisition mass spectrometry towards comprehensive digital proteome landscape. Mass Spectrom. Rev. 2023, 42, 2324–2348. [Google Scholar] [CrossRef]
  139. 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]
  140. 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]
  141. 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]
  142. Öhman, T.; Teppo, J.; Datta, N.; Mäkinen, S.; Varjosalo, M.; Koistinen, H.A. Skeletal muscle proteomes reveal downregulation of mitochondrial proteins in transition from prediabetes into type 2 diabetes. iScience 2021, 24, 102712. [Google Scholar] [CrossRef] [PubMed]
  143. Cretich, M.; Damin, F.; Chiari, M. Protein microarray technology: How far off is routine diagnostics? Analyst 2014, 139, 528–542. [Google Scholar] [CrossRef]
  144. Ayoglu, B.; Chaouch, A.; Lochmüller, H.; Politano, L.; Bertini, E.; Spitali, P.; Hiller, M.; Niks, E.H.; Gualandi, F.; Pontén, F.; et al. Affinity proteomics within rare diseases: A BIO-NMD study for blood biomarkers of muscular dystrophies. EMBO Mol. Med. 2014, 6, 918–936. [Google Scholar] [CrossRef] [PubMed]
  145. Cosemans, G.; Merckx, C.; De Bleecker, J.L.; De Paepe, B. Inducible Heat Shock Protein 70 Levels in Patients and the mdx Mouse Affirm Regulation during Skeletal Muscle Regeneration in Muscular Dystrophy. Front. Biosci. (Schol. Ed.) 2022, 14, 19. [Google Scholar] [CrossRef] [PubMed]
  146. Aparna, G.M.; Tetala, K.K.R. Recent Progress in Development and Application of DNA, Protein, Peptide, Glycan, Antibody, and Aptamer Microarrays. Biomolecules 2023, 13, 602. [Google Scholar] [CrossRef]
  147. Domon, B.; Aebersold, R. Mass spectrometry and protein analysis. Science 2006, 312, 212–217. [Google Scholar] [CrossRef]
  148. Doran, P.; Wilton, S.D.; Fletcher, S.; Ohlendieck, K. Proteomic profiling of antisense-induced exon skipping reveals reversal of pathobiochemical abnormalities in dystrophic mdx diaphragm. Proteomics 2009, 9, 671–685. [Google Scholar] [CrossRef]
  149. Clarke, M.S. Proteomic analysis of skeletal muscle tissue using SELDI-TOF MS: Application to disuse atrophy. Methods Mol. Biol. 2012, 818, 131–141. [Google Scholar] [CrossRef]
  150. Dare, T.O.; Davies, H.A.; Turton, J.A.; Lomas, L.; Williams, T.C.; York, M.J. Application of surface-enhanced laser desorption/ionization technology to the detection and identification of urinary parvalbumin-alpha: A biomarker of compound-induced skeletal muscle toxicity in the rat. Electrophoresis 2002, 23, 3241–3251. [Google Scholar] [CrossRef]
  151. 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]
  152. 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]
  153. 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]
  154. 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]
  155. 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] [PubMed]
  156. 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]
  157. 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]
  158. 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]
  159. 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]
  160. Tighe, P.J.; Ryder, R.R.; Todd, I.; Fairclough, L.C. ELISA in the multiplex era: Potentials and pitfalls. Proteom. Clin. Appl. 2015, 9, 406–422. [Google Scholar] [CrossRef]
  161. Tabatabaei, M.S.; Ahmed, M. Enzyme-Linked Immunosorbent Assay (ELISA). Methods Mol. Biol. 2022, 2508, 115–134. [Google Scholar] [CrossRef]
  162. Hakim, C.H.; Kumar, S.R.P.; Pérez-López, D.; Teixeira, J.; Herzog, R.W.; Duan, D. Assessment of the Gene Therapy Immune Response in the Canine Muscular Dystrophy Model. Methods Mol. Biol. 2023, 2587, 353–375. [Google Scholar] [CrossRef] [PubMed]
  163. Landsberger, M.; Brinkmeier, H. Immunoblot Analysis of DIGE-Based Proteomics. Methods Mol. Biol. 2023, 2596, 429–443. [Google Scholar] [CrossRef]
  164. 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]
  165. Carberry, S.; Ohlendieck, K. Gel electrophoresis-based proteomics of senescent tissues. Methods Mol. Biol. 2013, 1048, 229–246. [Google Scholar] [CrossRef] [PubMed]
  166. Mora, J.; Given Chunyk, A.; Dysinger, M.; Purushothama, S.; Ricks, C.; Osterlund, K.; Theobald, V. Next generation ligand binding assays-review of emerging technologies’ capabilities to enhance throughput and multiplexing. AAPS J. 2014, 16, 1175–1184. [Google Scholar] [CrossRef] [PubMed]
  167. Hendricks, R.; Baker, D.; Brumm, J.; Davancaze, T.; Harp, C.; Herman, A.; Büdingen, H.V.; Townsend, M.; Fischer, S.K. Establishment of neurofilament light chain Simoa assay in cerebrospinal fluid and blood. Bioanalysis 2019, 11, 1405–1418. [Google Scholar] [CrossRef]
  168. Cui, M.; Cheng, C.; Zhang, L. High-throughput proteomics: A methodological mini-review. Lab. Invest. 2022, 102, 1170–1181. [Google Scholar] [CrossRef]
  169. 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]
  170. 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]
  171. Sawano, S.; Mizunoya, W. History and development of staining methods for skeletal muscle fiber types. Histol. Histopathol. 2022, 37, 493–503. [Google Scholar] [CrossRef]
  172. Zweyer, M.; Sabir, H.; Dowling, P.; Gargan, S.; Murphy, S.; Swandulla, D.; Ohlendieck, K. Histopathology of Duchenne muscular dystrophy in correlation with changes in proteomic biomarkers. Histol. Histopathol. 2022, 37, 101–116. [Google Scholar] [CrossRef] [PubMed]
  173. Warren, C.; McDonald, D.; Capaldi, R.; Deehan, D.; Taylor, R.W.; Filby, A.; Turnbull, D.M.; Lawless, C.; Vincent, A.E. Decoding mitochondrial heterogeneity in single muscle fibres by imaging mass cytometry. Sci. Rep. 2020, 10, 15336. [Google Scholar] [CrossRef] [PubMed]
  174. Thirumal, S.; Jamzad, A.; Cotechini, T.; Hindmarch, C.T.; Graham, C.H.; Siemens, D.R.; Mousavi, P. TITAN: An end-to-end data analysis environment for the Hyperion™ imaging system. Cytometry A 2022, 101, 423–433. [Google Scholar] [CrossRef] [PubMed]
  175. Park, J.; Kim, J.; Lewy, T.; Rice, C.M.; Elemento, O.; Rendeiro, A.F.; Mason, C.E. Spatial omics technologies at multimodal and single cell/subcellular level. Genome Biol. 2022, 23, 256. [Google Scholar] [CrossRef] [PubMed]
  176. Manohar, S.M.; Shah, P.; Nair, A. Flow cytometry: Principles, applications and recent advances. Bioanalysis 2021, 13, 181–198. [Google Scholar] [CrossRef] [PubMed]
  177. Dell’Orso, S.; Juan, A.H.; Ko, K.D.; Naz, F.; Perovanovic, J.; Gutierrez-Cruz, G.; Feng, X.; Sartorelli, V. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions. Development 2019, 146, dev174177. [Google Scholar] [CrossRef]
  178. Li, N.; Parkes, J.E.; Spathis, R.; Morales, M.; Mcdonald, J.; Kendra, R.M.; Ott, E.M.; Brown, K.J.; Lawlor, M.W.; Nagaraju, K. The Effect of Immunomodulatory Treatments on Anti-Dystrophin Immune Response After AAV Gene Therapy in Dystrophin Deficient mdx Mice. J. Neuromuscul. Dis. 2021, 8, S325–S340. [Google Scholar] [CrossRef]
  179. Arora, B.; Tandon, R.; Attri, P.; Bhatia, R. Chemical Crosslinking: Role in Protein and Peptide Science. Curr. Protein Pept. Sci. 2017, 18, 946–955. [Google Scholar] [CrossRef]
  180. 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]
  181. Jayachandran, B.; Parvin, T.N.; Alam, M.M.; Chanda, K.; Mm, B. Insights on Chemical Crosslinking Strategies for Proteins. Molecules 2022, 27, 8124. [Google Scholar] [CrossRef]
  182. 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]
  183. O’Connell, K.; Ohlendieck, K. Proteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscle. Proteomics 2009, 9, 5509–5524. [Google Scholar] [CrossRef]
  184. Dowd, A. Enzyme Assay Methods to Validate DIGE Proteomics Data. Methods Mol. Biol. 2023, 2596, 421–428. [Google Scholar] [CrossRef]
  185. Ohlendieck, K. Skeletal muscle proteomics: Current approaches, technical challenges and emerging techniques. Skelet. Muscle 2011, 1, 6. [Google Scholar] [CrossRef]
  186. 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]
  187. Frontera, W.R.; Ochala, J. Skeletal muscle: A brief review of structure and function. Calcif. Tissue Int. 2015, 96, 183–195. [Google Scholar] [CrossRef]
  188. 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]
  189. 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]
  190. Schiaffino, S.; Reggiani, C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 2011, 91, 1447–1531. [Google Scholar] [CrossRef]
  191. Schiaffino, S. Fibre types in skeletal muscle: A personal account. Acta Physiol. 2010, 199, 451–463. [Google Scholar] [CrossRef]
  192. Bottinelli, R.; Reggiani, C. Human skeletal muscle fibres: Molecular and functional diversity. Prog. Biophys. Mol. Biol. 2000, 73, 195–262. [Google Scholar] [CrossRef]
  193. Swiderski, K.; Lynch, G.S. Murine models of Duchenne muscular dystrophy: Is there a best model? Am. J. Physiol. Cell Physiol. 2021, 321, C409–C412. [Google Scholar] [CrossRef]
  194. Schiaffino, S.; Gorza, L.; Pitton, G.; Saggin, L.; Ausoni, S.; Sartore, S.; Lømo, T. Embryonic and neonatal myosin heavy chain in denervated and paralyzed rat skeletal muscle. Dev. Biol. 1988, 127, 1–11. [Google Scholar] [CrossRef]
  195. Agarwal, M.; Sharma, A.; Kumar, P.; Kumar, A.; Bharadwaj, A.; Saini, M.; Kardon, G.; Mathew, S.J. Myosin heavy chain-embryonic regulates skeletal muscle differentiation during mammalian development. Development 2020, 147, dev184507. [Google Scholar] [CrossRef]
  196. Gargan, S.; Dowling, P.; Zweyer, M.; Reimann, J.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Mass Spectrometric Profiling of Extraocular Muscle and Proteomic Adaptations in the mdx-4cv Model of Duchenne Muscular Dystrophy. Life 2021, 11, 595. [Google Scholar] [CrossRef]
  197. Hoh, J.F.Y. Developmental, physiologic and phylogenetic perspectives on the expression and regulation of myosin heavy chains in mammalian skeletal muscles. J. Comp. Physiol. B 2023, 193, 355–382. [Google Scholar] [CrossRef]
  198. Pette, D.; Staron, R.S. Transitions of muscle fiber phenotypic profiles. Histochem. Cell Biol. 2001, 115, 359–372. [Google Scholar] [CrossRef]
  199. Ohlendieck, K. Proteomic profiling of skeletal muscle plasticity. Muscles Ligaments Tendons J. 2012, 1, 119–216. [Google Scholar]
  200. 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]
  201. 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]
  202. 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]
  203. 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]
  204. Yablonka-Reuveni, Z. The skeletal muscle satellite cell: Still young and fascinating at 50. J. Histochem. Cytochem. 2011, 59, 1041–1059. [Google Scholar] [CrossRef]
  205. Negroni, E.; Kondili, M.; Muraine, L.; Bensalah, M.; Butler-Browne, G.S.; Mouly, V.; Bigot, A.; Trollet, C. Muscle fibro-adipogenic progenitors from a single-cell perspective: Focus on their “virtual” secretome. Front. Cell Dev. Biol. 2022, 10, 952041. [Google Scholar] [CrossRef]
  206. Bernard, C.; Zavoriti, A.; Pucelle, Q.; Chazaud, B.; Gondin, J. Role of macrophages during skeletal muscle regeneration and hypertrophy-Implications for immunomodulatory strategies. Physiol. Rep. 2022, 10, e15480. [Google Scholar] [CrossRef]
  207. Engquist, E.N.; Zammit, P.S. The Satellite Cell at 60: The Foundation Years. J. Neuromuscul. Dis. 2021, 8, S183–S203. [Google Scholar] [CrossRef]
  208. Morgan, J.E.; Partridge, T.A. Muscle satellite cells. Int. J. Biochem. Cell Biol. 2003, 35, 1151–1156. [Google Scholar] [CrossRef]
  209. Buckingham, M.; Relaix, F. PAX3 and PAX7 as upstream regulators of myogenesis. Semin. Cell Dev. Biol. 2015, 44, 115–125. [Google Scholar] [CrossRef]
  210. Zammit, P.S. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin. Cell Dev. Biol. 2017, 72, 19–32. [Google Scholar] [CrossRef]
  211. Beauchamp, J.R.; Heslop, L.; Yu, D.S.; Tajbakhsh, S.; Kelly, R.G.; Wernig, A.; Buckingham, M.E.; Partridge, T.A.; Zammit, P.S. Expression of CD34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells. J. Cell Biol. 2000, 151, 1221–1234. [Google Scholar] [CrossRef]
  212. Khurram, O.U.; Pearcey, G.E.P.; Chardon, M.K.; Kim, E.H.; García, M.; Heckman, C.J. The Cellular Basis for the Generation of Firing Patterns in Human Motor Units. Adv. Neurobiol. 2022, 28, 233–258. [Google Scholar] [CrossRef]
  213. 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]
  214. Severinsen, M.C.K.; Pedersen, B.K. Muscle-Organ Crosstalk: The Emerging Roles of Myokines. Endocr. Rev. 2020, 41, 594–609. [Google Scholar] [CrossRef]
  215. 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]
  216. Lara-Castillo, N.; Johnson, M.L. Bone-Muscle Mutual Interactions. Curr. Osteoporos. Rep. 2020, 18, 408–421. [Google Scholar] [CrossRef]
  217. Gomarasca, M.; Banfi, G.; Lombardi, G. Myokines: The endocrine coupling of skeletal muscle and bone. Adv. Clin. Chem. 2020, 94, 155–218. [Google Scholar] [CrossRef]
  218. 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]
  219. 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]
  220. 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]
  221. 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]
  222. Baeza-Trinidad, R. Rhabdomyolysis: A syndrome to be considered. Rabdomiólisis: Un síndrome a tener en cuenta. Med. Clin. 2022, 158, 277–283. [Google Scholar] [CrossRef]
  223. Cabrera-Serrano, M.; Ravenscroft, G. Recent advances in our understanding of genetic rhabdomyolysis. Curr. Opin. Neurol. 2022, 35, 651–657. [Google Scholar] [CrossRef]
  224. Cabral, B.M.I.; Edding, S.N.; Portocarrero, J.P.; Lerma, E.V. Rhabdomyolysis. Dis. Mon. 2020, 66, 101015. [Google Scholar] [CrossRef]
  225. Carneiro, A.; Macedo-da-Silva, J.; Santiago, V.F.; de Oliveira, G.S.; Guimarães, T.; Mendonça, C.F.; de Oliveira Branquinho, J.L.; Lucena, C.V.; Osório, J.; Pernambuco, E. Urine proteomics as a non-invasive approach to monitor exertional rhabdomyolysis during military training. J. Proteom. 2022, 258, 104498. [Google Scholar] [CrossRef]
  226. 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]
  227. 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]
  228. 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]
  229. 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]
  230. 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]
  231. 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]
  232. 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] [PubMed]
  233. 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]
  234. 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]
  235. 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]
  236. 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] [PubMed]
  237. 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]
  238. 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] [PubMed]
  239. 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]
  240. 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]
  241. 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]
  242. 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]
  243. Straub, V.; Guglieri, M. An update on Becker muscular dystrophy. Curr. Opin. Neurol. 2023, 36, 450–454. [Google Scholar] [CrossRef] [PubMed]
  244. Flanigan, K.M.; Dunn, D.M.; von Niederhausern, A.; Soltanzadeh, P.; Gappmaier, E.; Howard, M.T.; Sampson, J.B.; Mendell, J.R.; Wall, C.; King, W.M.; et al. Mutational spectrum of DMD mutations in dystrophinopathy patients: Application of modern diagnostic techniques to a large cohort. Hum. Mutat. 2009, 30, 1657–1666. [Google Scholar] [CrossRef] [PubMed]
  245. Juan-Mateu, J.; Gonzalez-Quereda, L.; Rodriguez, M.J.; Baena, M.; Verdura, E.; Nascimento, A.; Ortez, C.; Baiget, M.; Gallano, P. DMD Mutations in 576 Dystrophinopathy Families: A Step Forward in Genotype-Phenotype Correlations. PLoS ONE 2015, 10, e0135189. [Google Scholar] [CrossRef]
  246. Bladen, C.L.; Salgado, D.; Monges, S.; Foncuberta, M.E.; Kekou, K.; Kosma, K.; Dawkins, H.; Lamont, L.; Roy, A.J.; Chamova, T.; et al. The TREAT-NMD DMD Global Database: Analysis of more than 7000 Duchenne muscular dystrophy mutations. Hum. Mutat. 2015, 36, 395–402. [Google Scholar] [CrossRef] [PubMed]
  247. Muntoni, F.; Torelli, S.; Ferlini, A. Dystrophin and mutations: One gene, several proteins, multiple phenotypes. Lancet Neurol. 2003, 2, 731–740. [Google Scholar] [CrossRef]
  248. 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]
  249. Ervasti, J.M.; Ohlendieck, K.; Kahl, S.D.; Gaver, M.G.; Campbell, K.P. Deficiency of a glycoprotein component of the dystrophin complex in dystrophic muscle. Nature 1990, 345, 315–319. [Google Scholar] [CrossRef]
  250. Ohlendieck, K.; Campbell, K.P. Dystrophin-associated proteins are greatly reduced in skeletal muscle from mdx mice. J. Cell Biol. 1991, 115, 1685–1694. [Google Scholar] [CrossRef]
  251. Ohlendieck, K.; Matsumura, K.; Ionasescu, V.V.; Towbin, J.A.; Bosch, E.P.; Weinstein, S.L.; Sernett, S.W.; Campbell, K.P. Duchenne muscular dystrophy: Deficiency of dystrophin-associated proteins in the sarcolemma. Neurology 1993, 43, 795–800. [Google Scholar] [CrossRef]
  252. Ervasti, J.M.; Campbell, K.P. A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J. Cell Biol. 1993, 122, 809–823. [Google Scholar] [CrossRef]
  253. Murphy, S.; Ohlendieck, K. The biochemical and mass spectrometric profiling of the dystrophin complexome from skeletal muscle. Comput. Struct. Biotechnol. J. 2015, 14, 20–27. [Google Scholar] [CrossRef]
  254. Dowling, P.; Gargan, S.; Murphy, S.; Zweyer, M.; Sabir, H.; Swandulla, D.; Ohlendieck, K. The Dystrophin Node as Integrator of Cytoskeletal Organization, Lateral Force Transmission, Fiber Stability and Cellular Signaling in Skeletal Muscle. Proteomes 2021, 9, 9. [Google Scholar] [CrossRef]
  255. Zabłocka, B.; Górecki, D.C.; Zabłocki, K. Disrupted Calcium Homeostasis in Duchenne Muscular Dystrophy: A Common Mechanism behind Diverse Consequences. Int. J. Mol. Sci. 2021, 22, 11040. [Google Scholar] [CrossRef]
  256. Dowling, P.; Gargan, S.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of impaired excitation-contraction coupling and abnormal calcium handling in muscular dystrophy. Proteomics 2022, 22, e2200003. [Google Scholar] [CrossRef] [PubMed]
  257. Dubinin, M.V.; Belosludtsev, K.N. Ion Channels of the Sarcolemma and Intracellular Organelles in Duchenne Muscular Dystrophy: A Role in the Dysregulation of Ion Homeostasis and a Possible Target for Therapy. Int. J. Mol. Sci. 2023, 24, 2229. [Google Scholar] [CrossRef] [PubMed]
  258. Dowling, P.; Swandulla, D.; Ohlendieck, K. Cellular pathogenesis of Duchenne muscular dystrophy: Progressive myofibre degeneration, chronic inflammation, reactive myofibrosis and satellite cell dysfunction. Eur. J. Transl. Myol. 2023, 33, 11856. [Google Scholar] [CrossRef] [PubMed]
  259. 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]
  260. Holland, A.; Carberry, S.; Ohlendieck, K. Proteomics of the dystrophin-glycoprotein complex and dystrophinopathy. Curr. Protein Pept. Sci. 2013, 14, 680–697. [Google Scholar] [CrossRef]
  261. Fuller, H.R.; Graham, L.C.; Llavero Hurtado, M.; Wishart, T.M. Understanding the molecular consequences of inherited muscular dystrophies: Advancements through proteomic experimentation. Expert Rev. Proteom. 2016, 13, 659–671. [Google Scholar] [CrossRef]
  262. Holland, A.; Murphy, S.; Dowling, P.; Ohlendieck, K. Pathoproteomic profiling of the skeletal muscle matrisome in dystrophinopathy associated myofibrosis. Proteomics 2016, 16, 345–366. [Google Scholar] [CrossRef] [PubMed]
  263. Carr, S.J.; Zahedi, R.P.; Lochmüller, H.; Roos, A. Mass spectrometry-based protein analysis to unravel the tissue pathophysiology in Duchenne muscular dystrophy. Proteom. Clin. Appl. 2018, 12, 1700071. [Google Scholar] [CrossRef] [PubMed]
  264. Dowling, P.; Murphy, S.; Zweyer, M.; Raucamp, M.; Swandulla, D.; Ohlendieck, K. Emerging proteomic biomarkers of X-linked muscular dystrophy. Expert Rev. Mol. Diagn. 2019, 19, 739–755. [Google Scholar] [CrossRef]
  265. Al-Khalili Szigyarto, C. Duchenne muscular dystrophy: Recent advances in protein biomarkers and the clinical application. Expert Rev. Proteom. 2020, 17, 365–375. [Google Scholar] [CrossRef]
  266. Ge, Y.; Molloy, M.P.; Chamberlain, J.S.; Andrews, P.C. Proteomic analysis of mdx skeletal muscle: Great reduction of adenylate kinase 1 expression and enzymatic activity. Proteomics 2003, 3, 1895–1903. [Google Scholar] [CrossRef] [PubMed]
  267. Doran, P.; Martin, G.; Dowling, P.; Jockusch, H.; Ohlendieck, K. Proteome analysis of the dystrophin-deficient MDX diaphragm reveals a drastic increase in the heat shock protein cvHSP. Proteomics 2006, 6, 4610–4621. [Google Scholar] [CrossRef] [PubMed]
  268. Doran, P.; Dowling, P.; Donoghue, P.; Buffini, M.; Ohlendieck, K. Reduced expression of regucalcin in young and aged mdx diaphragm indicates abnormal cytosolic calcium handling in dystrophin-deficient muscle. Biochim. Biophys. Acta 2006, 1764, 773–785. [Google Scholar] [CrossRef]
  269. Carberry, S.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Proteomics reveals drastic increase of extracellular matrix proteins collagen and dermatopontin in the aged mdx diaphragm model of Duchenne muscular dystrophy. Int. J. Mol. Med. 2012, 30, 229–234. [Google Scholar] [CrossRef]
  270. Gardan-Salmon, D.; Dixon, J.M.; Lonergan, S.M.; Selsby, J.T. Proteomic assessment of the acute phase of dystrophin deficiency in mdx mice. Eur. J. Appl. Physiol. 2011, 111, 2763–2773. [Google Scholar] [CrossRef]
  271. Guevel, L.; Lavoie, J.R.; Perez-Iratxeta, C.; Rouger, K.; Dubreil, L.; Feron, M.; Talon, S.; Brand, M.; Megeney, L.A. Quantitative proteomic analysis of dystrophic dog muscle. J. Proteome Res. 2011, 10, 2465–2478. [Google Scholar] [CrossRef]
  272. Yoon, J.H.; Johnson, E.; Xu, R.; Martin, L.T.; Martin, P.T.; Montanaro, F. Comparative proteomic profiling of dystroglycan-associated proteins in wild type, mdx, and Galgt2 transgenic mouse skeletal muscle. J. Proteome Res. 2012, 11, 4413–4424. [Google Scholar] [CrossRef] [PubMed]
  273. Ge, Y.; Molloy, M.P.; Chamberlain, J.S.; Andrews, P.C. Differential expression of the skeletal muscle proteome in mdx mice at different ages. Electrophoresis 2004, 25, 2576–2585. [Google Scholar] [CrossRef]
  274. Carberry, S.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Application of fluorescence two-dimensional difference in-gel electrophoresis as a proteomic biomarker discovery tool in muscular dystrophy research. Biology 2013, 2, 1438–1464. [Google Scholar] [CrossRef] [PubMed]
  275. 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]
  276. Holland, A.; Dowling, P.; Meleady, P.; Henry, M.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Label-free mass spectrometric analysis of the mdx-4cv diaphragm identifies the matricellular protein periostin as a potential factor involved in dystrophinopathy-related fibrosis. Proteomics 2015, 15, 2318–2331. [Google Scholar] [CrossRef] [PubMed]
  277. Roberts, T.C.; Johansson, H.J.; McClorey, G.; Godfrey, C.; Blomberg, K.E.; Coursindel, T.; Gait, M.J.; Smith, C.I.; Lehtiö, J.; El Andaloussi, S.; et al. Multi-level omics analysis in a murine model of dystrophin loss and therapeutic restoration. Hum. Mol. Genet. 2015, 24, 6756–6768. [Google Scholar] [CrossRef] [PubMed]
  278. Holland, A.; Henry, M.; Meleady, P.; Winkler, C.K.; Krautwald, M.; Brinkmeier, H.; Ohlendieck, K. Comparative Label-Free Mass Spectrometric Analysis of Mildly versus Severely Affected mdx Mouse Skeletal Muscles Identifies Annexin, Lamin, and Vimentin as Universal Dystrophic Markers. Molecules 2015, 20, 11317–11344. [Google Scholar] [CrossRef]
  279. Murphy, S.; Henry, M.; Meleady, P.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Simultaneous Pathoproteomic Evaluation of the Dystrophin-Glycoprotein Complex and Secondary Changes in the mdx-4cv Mouse Model of Duchenne Muscular Dystrophy. Biology 2015, 4, 397–423. [Google Scholar] [CrossRef]
  280. Fröhlich, T.; Kemter, E.; Flenkenthaler, F.; Klymiuk, N.; Otte, K.A.; Blutke, A.; Krause, S.; Walter, M.C.; Wanke, R.; Wolf, E.; et al. Progressive muscle proteome changes in a clinically relevant pig model of Duchenne muscular dystrophy. Sci. Rep. 2016, 6, 33362. [Google Scholar] [CrossRef]
  281. Turk, R.; Hsiao, J.J.; Smits, M.M.; Ng, B.H.; Pospisil, T.C.; Jones, K.S.; Campbell, K.P.; Wright, M.E. Molecular Signatures of Membrane Protein Complexes Underlying Muscular Dystrophy. Mol. Cell. Proteom. 2016, 15, 2169–2185. [Google Scholar] [CrossRef]
  282. 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]
  283. Gamberi, T.; Fiaschi, T.; Valocchia, E.; Modesti, A.; Mantuano, P.; Rolland, J.F.; Sanarica, F.; De Luca, A.; Magherini, F. Proteome analysis in dystrophic mdx mouse muscle reveals a drastic alteration of key metabolic and contractile proteins after chronic exercise and the potential modulation by anti-oxidant compounds. J. Proteom. 2018, 170, 43–58. [Google Scholar] [CrossRef] [PubMed]
  284. 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]
  285. 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]
  286. Capitanio, D.; Moriggi, M.; Torretta, E.; Barbacini, P.; De Palma, S.; Viganò, A.; Capitanio, D.; Moriggi, M.; Torretta, E.; Barbacini, P.; 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]
  287. Capitanio, D.; Moriggi, M.; Barbacini, P.; Torretta, E.; Moroni, I.; Blasevich, F.; Morandi, L.; Mora, M.; Gelfi, C. Molecular fingerprint of BMD patients lacking a portion in the rod domain of dystrophin. Int. J. Mol. Sci. 2020, 23, 2624. [Google Scholar] [CrossRef] [PubMed]
  288. 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. Proteom. 2020, 19, 2047–2068. [Google Scholar] [CrossRef] [PubMed]
  289. Stirm, M.; Fonteyne, L.M.; Shashikadze, B.; Lindner, M.; Chirivi, M.; Lange, A.; Kaufhold, C.; Mayer, C.; Medugorac, I.; Kessler, B.; et al. A scalable, clinically severe pig model for Duchenne muscular dystrophy. Dis. Models Mech. 2021, 14, dmm049285. [Google Scholar] [CrossRef]
  290. Gargan, S.; Dowling, P.; Zweyer, M.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteomic Identification of Markers of Membrane Repair, Regeneration and Fibrosis in the Aged and Dystrophic Diaphragm. Life 2022, 12, 1679. [Google Scholar] [CrossRef]
  291. Day, N.J.; Zhang, T.; Gaffrey, M.J.; Zhao, R.; Fillmore, T.L.; Moore, R.J.; Rodney, G.G.; Qian, W.J. A deep redox proteome profiling workflow and its application to skeletal muscle of a Duchenne Muscular Dystrophy model. Free Radic. Biol. Med. 2022, 193, 373–384. [Google Scholar] [CrossRef]
  292. Mucha, O.; Myszka, M.; Podkalicka, P.; Świderska, B.; Malinowska, A.; Dulak, J.; Łoboda, A. Proteome Profiling of the Dystrophic mdx Mice Diaphragm. Biomolecules 2023, 13, 1648. [Google Scholar] [CrossRef] [PubMed]
  293. Ng, S.Y.; Ljubicic, V. Recent insights into neuromuscular junction biology in Duchenne muscular dystrophy: Impacts, challenges, and opportunities. EBioMedicine 2020, 61, 103032. [Google Scholar] [CrossRef]
  294. Meyers, T.A.; Townsend, D. Cardiac Pathophysiology and the Future of Cardiac Therapies in Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2019, 20, 4098. [Google Scholar] [CrossRef] [PubMed]
  295. Florczyk-Soluch, U.; Polak, K.; Dulak, J. The multifaceted view of heart problem in Duchenne muscular dystrophy. Cell. Mol. Life Sci. 2021, 78, 5447–5468. [Google Scholar] [CrossRef] [PubMed]
  296. Lechner, A.; Herzig, J.J.; Kientsch, J.G.; Kohler, M.; Bloch, K.E.; Ulrich, S.; Schwarz, E.I. Cardiomyopathy as cause of death in Duchenne muscular dystrophy: A longitudinal observational study. ERJ Open Res. 2023, 9, 00176–02023. [Google Scholar] [CrossRef]
  297. Ricotti, V.; Selby, V.; Ridout, D.; Domingos, J.; Decostre, V.; Mayhew, A.; Eagle, M.; Butler, J.; Guglieri, M.; Van der Holst, M.; et al. Respiratory and upper limb function as outcome measures in ambulant and non- ambulant subjects with Duchenne muscular dystrophy: A prospective multicentre study. Neuromuscul. Disord. 2019, 29, 261–268. [Google Scholar] [CrossRef] [PubMed]
  298. Barnard, A.M.; Lott, D.J.; Batra, A.; Triplett, W.T.; Forbes, S.C.; Riehl, S.L.; Willcocks, R.J.; Smith, B.K.; Vandenborne, K.; Walter, G.A. Imaging respiratory muscle quality and function in Duchenne muscular dystrophy. J. Neurol. 2019, 266, 2752–2763. [Google Scholar] [CrossRef] [PubMed]
  299. Pennati, F.; LoMauro, A.; D’Angelo, M.G.; Aliverti, A. Non-Invasive Respiratory Assessment in Duchenne Muscular Dystrophy: From Clinical Research to Outcome Measures. Life 2019, 11, 947. [Google Scholar] [CrossRef]
  300. Fayssoil, A.; Chaffaut, C.; Ogna, A.; Stojkovic, T.; Lamothe, L.; Mompoint, D.; Meng, P.; Prigent, H.; Clair, B.; Behin, A.; et al. Echographic assessment of diaphragmatic function in Duchenne muscular dystrophy from childhood to adulthood. J. Neuromuscul. Dis. 2019, 6, 55–64. [Google Scholar] [CrossRef]
  301. Moriuchi, T.; Fujii, Y.; Kagawa, N.; Hizawa, K. Autopsy study on the weight of the heart, liver, kidney and brain in Duchenne muscular dystrophy. Tokushima J. Exp. Med. 1991, 38, 5–13. [Google Scholar]
  302. Naume, M.M.; Jørgensen, M.H.; Høi-Hansen, C.E.; Born, A.P.; Vissing, J.; Borgwardt, L.; Staerk, D.M.R.; Ørngreen, M.C. Metabolic assessment in children with neuromuscular disorders shows risk of liver enlargement, steatosis and fibrosis. Acta Paediatr. 2023, 112, 846–853. [Google Scholar] [CrossRef] [PubMed]
  303. Motoki, T.; Shimizu-Motohashi, Y.; Komaki, H.; Mori-Yoshimura, M.; Oya, Y.; Takeshita, E.; Ishiyama, A.; Saito, T.; Nakagawa, E.; Sugai, K.; et al. Treatable renal failure found in non-ambulatory Duchenne muscular dystrophy patients. Neuromuscul. Disord. 2015, 25, 754–757. [Google Scholar] [CrossRef] [PubMed]
  304. Braat, E.; Hoste, L.; De Waele, L.; Gheysens, O.; Vermeersch, P.; Goffin, K.; Pottel, H.; Goemans, N.; Levtchenko, E. Renal function in children and adolescents with Duchenne muscular dystrophy. Neuromuscul. Disord. 2015, 25, 381–387. [Google Scholar] [CrossRef]
  305. Kutluk, M.G.; Doğan, Ç.S. Kidney involvement and associated risk factors in children with Duchenne muscular dystrophy. Pediatr. Nephrol. 2020, 35, 1953–1958. [Google Scholar] [CrossRef]
  306. Villa, C.R.; Kaddourah, A.; Mathew, J.; Ryan, T.D.; Wong, B.L.; Goldstein, S.L.; Jefferies, J.L. Identifying evidence of cardio-renal syndrome in patients with Duchenne muscular dystrophy using cystatin C. Neuromuscul. Disord. 2016, 26, 637–642. [Google Scholar] [CrossRef] [PubMed]
  307. Caress, J.B.; Kothari, M.J.; Bauer, S.B.; Shefner, J.M. Urinary dysfunction in Duchenne muscular dystrophy. Muscle Nerve 1996, 19, 819–822. [Google Scholar] [CrossRef]
  308. MacLeod, M.; Kelly, R.; Robb, S.A.; Borzyskowski, M. Bladder dysfunction in Duchenne muscular dystrophy. Arch. Dis. Child. 2003, 88, 347–349. [Google Scholar] [CrossRef]
  309. Lionarons, J.M.; de Groot, I.J.M.; Fock, J.M.; Klinkenberg, S.; Vrijens, D.M.J.; Vreugdenhil, A.C.E.; Medici-van den Herik, E.G.; Cuppen, I.; Jaeger, B.; Niks, E.H.; et al. Prevalence of Bladder and Bowel Dysfunction in Duchenne Muscular Dystrophy Using the Childhood Bladder and Bowel Dysfunction Questionnaire. Life 2021, 11, 772. [Google Scholar] [CrossRef]
  310. Manokaran, R.K.; Aggarwala, S.; Kumar, R.; Gupta, A.K.; Chakrabarty, B.; Jauhari, P.; Pandey, R.M.; Gulati, S. Prevalence of smooth muscle dysfunction among children with Duchenne muscular dystrophy. Muscle Nerve 2020, 62, 699–704. [Google Scholar] [CrossRef]
  311. Jaffe, K.M.; McDonald, C.M.; Ingman, E.; Haas, J. Symptoms of upper gastrointestinal dysfunction in Duchenne muscular dystrophy: Case-control study. Arch. Phys. Med. Rehabil. 1990, 71, 742–744. [Google Scholar]
  312. Borrelli, O.; Salvia, G.; Mancini, V.; Santoro, L.; Tagliente, F.; Romeo, E.F.; Cucchiara, S. Evolution of gastric electrical features and gastric emptying in children with Duchenne and Becker muscular dystrophy. Am. J. Gastroenterol. 2005, 100, 695–702. [Google Scholar] [CrossRef] [PubMed]
  313. Lo Cascio, C.M.; Goetze, O.; Latshang, T.D.; Bluemel, S.; Frauenfelder, T.; Bloch, K.E. Gastrointestinal Dysfunction in Patients with Duchenne Muscular Dystrophy. PLoS ONE 2016, 11, e0163779. [Google Scholar] [CrossRef] [PubMed]
  314. Rufo, A.; Del Fattore, A.; Capulli, M.; Carvello, F.; De Pasquale, L.; Ferrari, S.; Pierroz, D.; Morandi, L.; De Simone, M.; Rucci, N.; et al. Mechanisms inducing low bone density in Duchenne muscular dystrophy in mice and humans. J. Bone Miner. Res. 2011, 26, 1891–1903. [Google Scholar] [CrossRef] [PubMed]
  315. Garg, S. Management of scoliosis in patients with Duchenne muscular dystrophy and spinal muscular atrophy: A literature review. J. Pediatr. Rehabil. Med. 2016, 9, 23–29. [Google Scholar] [CrossRef]
  316. Roberts, S.; Arshad, A.; Tsirikos, A.I. Surgical and long-term functional outcomes of patients with Duchenne muscular dystrophy following spinal deformity correction. World J. Orthop. 2023, 14, 411–426. [Google Scholar] [CrossRef]
  317. Arshad, A.; Tsirikos, A.I. Survival of patients with Duchenne muscular dystrophy who underwent spinal deformity correction. Dev. Med. Child Neurol. 2024, 66, 187–194. [Google Scholar] [CrossRef]
  318. Mehler, M.F. Brain dystrophin, neurogenetics and mental retardation. Brain Res. Rev. 2000, 32, 277–307. [Google Scholar] [CrossRef]
  319. Pane, M.; Lombardo, M.E.; Alfieri, P.; D’Amico, A.; Bianco, F.; Vasco, G.; Piccini, G.; Mallardi, M.; Romeo, D.M.; Ricotti, V.; et al. Attention deficit hyperactivity disorder and cognitive function in Duchenne muscular dystrophy: Phenotype-genotype correlation. J. Pediatr. 2012, 161, 705–709.e1. [Google Scholar] [CrossRef]
  320. Ricotti, V.; Mandy, W.P.; Scoto, M.; Pane, M.; Deconinck, N.; Messina, S.; Mercuri, E.; Skuse, D.H.; Muntoni, F. Neurodevelopmental, emotional, and behavioural problems in Duchenne muscular dystrophy in relation to underlying dystrophin gene mutations. Dev. Med. Child Neurol. 2016, 58, 77–84. [Google Scholar] [CrossRef]
  321. Parisi, L.; Di Filippo, T.; Glorioso, P.; La Grutta, S.; Epifanio, M.S.; Roccella, M. Autism spectrum disorders in children affected by Duchenne muscular dystrophy. Minerva Pediatr. 2018, 70, 233–239. [Google Scholar] [CrossRef]
  322. Pascual-Morena, C.; Cavero-Redondo, I.; Álvarez-Bueno, C.; Jiménez-López, E.; Saz-Lara, A.; Martínez-García, I.; Martínez-Vizcaíno, V. Global prevalence of intellectual developmental disorder in dystrophinopathies: A systematic review and meta-analysis. Dev. Med. Child Neurol. 2023, 65, 734–744. [Google Scholar] [CrossRef] [PubMed]
  323. Holland, A.; Ohlendieck, K. Proteomic profiling of the dystrophin-deficient mdx phenocopy of dystrophinopathy-associated cardiomyopathy. Biomed. Res. Int. 2014, 2014, 246195. [Google Scholar] [CrossRef] [PubMed]
  324. Gowran, A.; Brioschi, M.; Rovina, D.; Chiesa, M.; Piacentini, L.; Mallia, S.; Banfi, C.; Pompilio, G.; Santoro, R. Multiomic Approaches to Uncover the Complexities of Dystrophin-Associated Cardiomyopathy. Int. J. Mol. Sci. 2021, 22, 8954. [Google Scholar] [CrossRef] [PubMed]
  325. Lee-Gannon, T.; Jiang, X.; Tassin, T.C.; Mammen, P.P.A. Biomarkers in Duchenne Muscular Dystrophy. Curr. Heart Fail. Rep. 2022, 19, 52–62. [Google Scholar] [CrossRef]
  326. Matkovich, S.J. Multiomic approaches to delineate the pathogenesis of cardiac disease. Curr. Opin. Cardiol. 2019, 34, 246–253. [Google Scholar] [CrossRef] [PubMed]
  327. Sohag, M.M.H.; Raqib, S.M.; Akhmad, S.A. OMICS approaches in cardiovascular diseases: A mini review. Genom. Inform. 2021, 19, e13. [Google Scholar] [CrossRef]
  328. Sarhene, M.; Wang, Y.; Wei, J.; Huang, Y.; Li, M.; Li, L.; Acheampong, E.; Zhengcan, Z.; Xiaoyan, Q.; Yunsheng, X.; et al. Biomarkers in heart failure: The past, current and future. Heart Fail. Rev. 2019, 24, 867–903. [Google Scholar] [CrossRef] [PubMed]
  329. Joshi, A.; Rienks, M.; Theofilatos, K.; Mayr, M. Systems biology in cardiovascular disease: A multiomics approach. Nat. Rev. Cardiol. 2021, 18, 313–330. [Google Scholar] [CrossRef]
  330. Lohan, J.; Ohlendieck, K. Drastic reduction in the luminal Ca2+-binding proteins calsequestrin and sarcalumenin in dystrophin-deficient cardiac muscle. Biochim. Biophys. Acta 2004, 1689, 252–258. [Google Scholar] [CrossRef]
  331. Gulston, M.K.; Rubtsov, D.V.; Atherton, H.J.; Clarke, K.; Davies, K.E.; Lilley, K.S.; Griffin, J.L. A combined metabolomic and proteomic investigation of the effects of a failure to express dystrophin in the mouse heart. J. Proteome Res. 2008, 7, 2069–2077. [Google Scholar] [CrossRef]
  332. Lewis, C.; Jockusch, H.; Ohlendieck, K. Proteomic Profiling of the Dystrophin-Deficient MDX Heart Reveals Drastically Altered Levels of Key Metabolic and Contractile Proteins. J. Biomed. Biotechnol. 2010, 2010, 648501. [Google Scholar] [CrossRef] [PubMed]
  333. Johnson, E.K.; Zhang, L.; Adams, M.E.; Phillips, A.; Freitas, M.A.; Froehner, S.C.; Green-Church, K.B.; Montanaro, F. Proteomic analysis reveals new cardiac-specific dystrophin-associated proteins. PLoS ONE 2012, 7, e43515. [Google Scholar] [CrossRef]
  334. Holland, A.; Dowling, P.; Zweyer, M.; Swandulla, D.; Henry, M.; Clynes, M.; Ohlendieck, K. Proteomic profiling of cardiomyopathic tissue from the aged mdx model of Duchenne muscular dystrophy reveals a drastic decrease in laminin, nidogen and annexin. Proteomics 2013, 13, 2312–2323. [Google Scholar] [CrossRef] [PubMed]
  335. Murphy, S.; Dowling, P.; Zweyer, M.; Mundegar, R.R.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteomic analysis of dystrophin deficiency and associated changes in the aged mdx-4cv heart model of dystrophinopathy-related cardiomyopathy. J. Proteom. 2016, 145, 24–36. [Google Scholar] [CrossRef]
  336. Chung, H.S.; Kim, G.E.; Holewinski, R.J.; Venkatraman, V.; Zhu, G.; Bedja, D.; Kass, D.A.; Van Eyk, J.E. Transient receptor potential channel 6 regulates abnormal cardiac S-nitrosylation in Duchenne muscular dystrophy. Proc. Natl. Acad. Sci. USA 2017, 114, E10763–E10771. [Google Scholar] [CrossRef] [PubMed]
  337. Tamiyakul, H.; Kemter, E.; Kösters, M.; Ebner, S.; Blutke, A.; Klymiuk, N.; Flenkenthaler, F.; Wolf, E.; Arnold, G.J.; Fröhlich, T. Progressive Proteome Changes in the Myocardium of a Pig Model for Duchenne Muscular Dystrophy. iScience 2020, 23, 101516. [Google Scholar] [CrossRef]
  338. Jung, C.; Martins, A.S.; Niggli, E.; Shirokova, N. Dystrophic cardiomyopathy: Amplification of cellular damage by Ca2+ signalling and reactive oxygen species-generating pathways. Cardiovasc. Res. 2008, 77, 766–773. [Google Scholar] [CrossRef]
  339. Klietsch, R.; Ervasti, J.M.; Arnold, W.; Campbell, K.P.; Jorgensen, A.O. Dystrophin-glycoprotein complex and laminin colocalize to the sarcolemma and transverse tubules of cardiac muscle. Circ. Res. 1993, 72, 349–360. [Google Scholar] [CrossRef]
  340. Wang, H.; Marrosu, E.; Brayson, D.; Wasala, N.B.; Johnson, E.K.; Scott, C.S.; Yue, Y.; Hau, K.L.; Trask, A.J.; Froehner, S.C.; et al. Proteomic analysis identifies key differences in the cardiac interactomes of dystrophin and micro-dystrophin. Hum. Mol. Genet. 2021, 30, 1321–1336. [Google Scholar] [CrossRef]
  341. Murphy, S.; Zweyer, M.; Henry, M.; Meleady, P.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of liver tissue from the mdx-4cv mouse model of Duchenne muscular dystrophy. Clin. Proteom. 2018, 15, 34. [Google Scholar] [CrossRef]
  342. 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]
  343. Murphy, S.; Dowling, P.; Zweyer, M.; Henry, M.; Meleady, P.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of mdx-4cv serum reveals highly elevated levels of the inflammation-induced plasma marker haptoglobin in muscular dystrophy. Int. J. Mol. Med. 2017, 39, 1357–1370. [Google Scholar] [CrossRef] [PubMed]
  344. Murphy, S.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Dataset on the comparative proteomic profiling of mouse saliva and serum from wild type versus the dystrophic mdx-4cv mouse model of dystrophinopathy. Data Brief. 2018, 21, 1236–1245. [Google Scholar] [CrossRef] [PubMed]
  345. Murphy, S.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Proteomic serum biomarkers for neuromuscular diseases. Expert Rev. Proteom. 2018, 15, 277–291. [Google Scholar] [CrossRef]
  346. Dowling, P.; Zweyer, M.; Raucamp, M.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteomic and cell biological profiling of the renal phenotype of the mdx-4cv mouse model of Duchenne muscular dystrophy. Eur. J. Cell Biol. 2020, 99, 151059. [Google Scholar] [CrossRef] [PubMed]
  347. Dowling, P.; Zweyer, M.; Raucamp, M.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Dataset on the mass spectrometry-based proteomic profiling of the kidney from wild type and the dystrophic mdx-4cv mouse model of X-linked muscular dystrophy. Data Brief 2020, 28, 105067. [Google Scholar] [CrossRef] [PubMed]
  348. Hathout, Y.; Marathi, R.L.; Rayavarapu, S.; Zhang, A.; Brown, K.J.; Seol, H.; Gordish-Dressman, H.; Cirak, S.; Bello, L.; Nagaraju, K.; et al. Discovery of serum protein biomarkers in the mdx mouse model and cross-species comparison to Duchenne muscular dystrophy patients. Hum. Mol. Genet. 2014, 23, 6458–6469. [Google Scholar] [CrossRef]
  349. Coenen-Stass, A.M.; McClorey, G.; Manzano, R.; Betts, C.A.; Blain, A.; Saleh, A.F.; Gait, M.J.; Lochmüller, H.; Wood, M.J.; Roberts, T.C. Identification of novel, therapy-responsive protein biomarkers in a mouse model of Duchenne muscular dystrophy by aptamer-based serum proteomics. Sci. Rep. 2015, 5, 17014. [Google Scholar] [CrossRef]
  350. Spitali, P.; Hettne, K.; Tsonaka, R.; Charrout, M.; van den Bergen, J.; Koeks, Z.; Kan, H.E.; Hooijmans, M.T.; Roos, A.; Straub, V.; et al. Tracking disease progression non-invasively in Duchenne and Becker muscular dystrophies. J. Cachexia Sarcopenia Muscle 2018, 9, 715–726. [Google Scholar] [CrossRef]
  351. Parolo, S.; Marchetti, L.; Lauria, M.; Misselbeck, K.; Scott-Boyer, M.P.; Caberlotto, L.; Priami, C. Combined use of protein biomarkers and network analysis unveils deregulated regulatory circuits in Duchenne muscular dystrophy. PLoS ONE 2018, 13, e0194225. [Google Scholar] [CrossRef]
  352. Villalta, S.A.; Rosenberg, A.S.; Bluestone, J.A. The immune system in Duchenne muscular dystrophy: Friend or foe. Rare Dis. 2015, 3, e1010966. [Google Scholar] [CrossRef]
  353. Rosenberg, A.S.; Puig, M.; Nagaraju, K.; Hoffman, E.P.; Villalta, S.A.; Rao, V.A.; Wakefield, L.M.; Woodcock, J. Immune-mediated pathology in Duchenne muscular dystrophy. Sci. Transl. Med. 2015, 7, 299rv4. [Google Scholar] [CrossRef]
  354. Tidball, J.G.; Welc, S.S.; Wehling-Henricks, M. Immunobiology of Inherited Muscular Dystrophies. Compr. Physiol. 2018, 8, 1313–1356. [Google Scholar] [CrossRef] [PubMed]
  355. Farini, A.; Sitzia, C.; Cassani, B.; Cassinelli, L.; Rigoni, R.; Colleoni, F.; Fusco, N.; Gatti, S.; Bella, P.; Villa, C.; et al. Therapeutic Potential of Immunoproteasome Inhibition in Duchenne Muscular Dystrophy. Mol. Ther. 2014, 24, 1898–1912. [Google Scholar] [CrossRef] [PubMed]
  356. Giordano, C.; Mojumdar, K.; Liang, F.; Lemaire, C.; Li, T.; Richardson, J.; Divangahi, M.; Qureshi, S.; Petrof, B.J. Toll-like receptor 4 ablation in mdx mice reveals innate immunity as a therapeutic target in Duchenne muscular dystrophy. Hum. Mol. Genet. 2015, 24, 2147–6212. [Google Scholar] [CrossRef] [PubMed]
  357. Ouisse, L.H.; Remy, S.; Lafoux, A.; Larcher, T.; Tesson, L.; Chenouard, V.; Guillonneau, C.; Brusselle, L.; Vimond, N.; Rouger, K.; et al. Immunophenotype of a Rat Model of Duchenne’s Disease and Demonstration of Improved Muscle Strength After Anti-CD45RC Antibody Treatment. Front. Immunol. 2019, 10, 2131. [Google Scholar] [CrossRef] [PubMed]
  358. Tripodi, L.; Molinaro, D.; Fortunato, F.; Mella, C.; Cassani, B.; Torrente, Y.; Farini, A. Immunoproteasome Inhibition Ameliorates Aged Dystrophic Mouse Muscle Environment. Int. J. Mol. Sci. 2022, 23, 14657. [Google Scholar] [CrossRef]
  359. Tripodi, L.; Villa, C.; Molinaro, D.; Torrente, Y.; Farini, A. The Immune System in Duchenne Muscular Dystrophy Pathogenesis. Biomedicines 2021, 9, 1447. [Google Scholar] [CrossRef]
  360. Mojumdar, K.; Liang, F.; Giordano, C.; Lemaire, C.; Danialou, G.; Okazaki, T.; Bourdon, J.; Rafei, M.; Galipeau, J.; Divangahi, M.; et al. Inflammatory monocytes promote progression of Duchenne muscular dystrophy and can be therapeutically targeted via CCR2. EMBO Mol. Med. 2014, 6, 1476–1492. [Google Scholar] [CrossRef]
  361. Mojumdar, K.; Giordano, C.; Lemaire, C.; Liang, F.; Divangahi, M.; Qureshi, S.T.; Petrof, B.J. Divergent impact of Toll-like receptor 2 deficiency on repair mechanisms in healthy muscle versus Duchenne muscular dystrophy. J. Pathol. 2016, 239, 10–22. [Google Scholar] [CrossRef]
  362. Lozanoska-Ochser, B.; Benedetti, A.; Rizzo, G.; Marrocco, V.; Di Maggio, R.; Fiore, P.; Bouche, M. Targeting early PKCθ-dependent T-cell infiltration of dystrophic muscle reduces disease severity in a mouse model of muscular dystrophy. J. Pathol. 2018, 244, 323–333. [Google Scholar] [CrossRef]
  363. Lemos, J.P.; Tenório, L.P.G.; Mouly, V.; Butler-Browne, G.; Mendes-da-Cruz, D.A.; Savino, W.; Smeriglio, P. T cell biology in neuromuscular disorders: A focus on Duchenne Muscular Dystrophy and Amyotrophic Lateral Sclerosis. Front. Immunol. 2023, 14, 1202834. [Google Scholar] [CrossRef]
  364. Petrof, B.J.; Podolsky, T.; Bhattarai, S.; Tan, J.; Ding, J. Trained immunity as a potential target for therapeutic immunomodulation in Duchenne muscular dystrophy. Front. Immunol. 2023, 14, 1183066. [Google Scholar] [CrossRef]
  365. Rizzo, G.; Di Maggio, R.; Benedetti, A.; Morroni, J.; Bouche, M.; Lozanoska-Ochser, B. Splenic Ly6Chi monocytes are critical players in dystrophic muscle injury and repair. JCI Insight 2020, 5, 130807. [Google Scholar] [CrossRef]
  366. Mebius, R.E.; Kraal, G. Structure and function of the spleen. Nat. Rev. Immunol. 2005, 5, 606–616. [Google Scholar] [CrossRef]
  367. Lewis, S.M.; Williams, A.; Eisenbarth, S.C. Structure and function of the immune system in the spleen. Sci. Immunol. 2019, 4, eaau6085. [Google Scholar] [CrossRef]
  368. Dowling, P.; Gargan, S.; Zweyer, M.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteome-wide Changes in the mdx-4cv Spleen due to Pathophysiological Cross Talk with Dystrophin-Deficient Skeletal Muscle. iScience 2020, 23, 101500. [Google Scholar] [CrossRef]
  369. Dowling, P.; Gargan, S.; Zweyer, M.; Sabir, H.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of the interface between the stomach wall and the pancreas in dystrophinopathy. Eur. J. Transl. Myol. 2021, 31, 9627. [Google Scholar] [CrossRef]
  370. Dowling, P.; Zweyer, M.; Sabir, H.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Mass spectrometry-based proteomic characterization of the middle-aged mouse brain for animal model research of neuromuscular diseases. Eur. J. Transl. Myol. 2023, 33, 11553. [Google Scholar] [CrossRef]
  371. Murphy, S.; Zweyer, M.; Henry, M.; Meleady, P.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Label-free mass spectrometric analysis reveals complex changes in the brain proteome from the mdx-4cv mouse model of Duchenne muscular dystrophy. Clin. Proteom. 2015, 12, 27. [Google Scholar] [CrossRef]
  372. Murphy, S.; Ohlendieck, K. Proteomic Profiling of the Dystrophin-Deficient Brain. Methods Mol. Biol. 2018, 1687, 91–105. [Google Scholar] [CrossRef]
  373. Yang, Z.; Wang, K.K. Glial fibrillary acidic protein: From intermediate filament assembly and gliosis to neurobiomarker. Trends Neurosci. 2015, 38, 364–374. [Google Scholar] [CrossRef]
  374. Escartin, C.; Galea, E.; Lakatos, A.; O’Callaghan, J.P.; Petzold, G.C.; Serrano-Pozo, A.; Steinhäuser, C.; Volterra, A.; Carmignoto, G.; Agarwal, A.; et al. Reactive astrocyte nomenclature, definitions, and future directions. Nat. Neurosci. 2021, 24, 312–325. [Google Scholar] [CrossRef]
  375. Jurga, A.M.; Paleczna, M.; Kadluczka, J.; Kuter, K.Z. Beyond the GFAP-Astrocyte Protein Markers in the Brain. Biomolecules 2021, 11, 1361. [Google Scholar] [CrossRef]
  376. Murphy, S.; Schmitt-John, T.; Dowling, P.; Henry, M.; Meleady, P.; Swandulla, D.; Ohlendieck, K. Proteomic profiling of the brain from the wobbler mouse model of amyotrophic lateral sclerosis reveals elevated levels of the astrogliosis marker glial fibrillary acidic protein. Eur. J. Transl. Myol. 2023, 33, 11555. [Google Scholar] [CrossRef]
  377. Chesshyre, M.; Ridout, D.; Hashimoto, Y.; Ookubo, Y.; Torelli, S.; Maresh, K.; Ricotti, V.; Abbott, L.; Gupta, V.A.; Main, M.; et al. Investigating the role of dystrophin isoform deficiency in motor function in Duchenne muscular dystrophy. J. Cachexia Sarcopenia Muscle 2022, 13, 1360–1372. [Google Scholar] [CrossRef]
  378. De Stefano, M.E.; Ferretti, V.; Mozzetta, C. Synaptic alterations as a neurodevelopmental trait of Duchenne muscular dystrophy. Neurobiol. Dis. 2022, 168, 105718. [Google Scholar] [CrossRef]
  379. Wijekoon, N.; Gonawala, L.; Ratnayake, P.; Amaratunga, D.; Hathout, Y.; Mohan, C.; Steinbusch, H.W.M.; Dalal, A.; Hoffman, E.P.; de Silva, K.R.D. Duchenne Muscular Dystrophy from Brain to Muscle: The Role of Brain Dystrophin Isoforms in Motor Functions. J. Clin. Med. 2023, 12, 5637. [Google Scholar] [CrossRef]
  380. Andrews, J.G.; Galindo, M.K.; Thomas, S.; Mathews, K.D.; Whitehead, N. DMD Gene and Dystrophinopathy Phenotypes Associated with Mutations: A Systematic Review for Clinicians. J. Clin. Neuromuscul. Dis. 2023, 24, 171–187. [Google Scholar] [CrossRef]
  381. Hoffman, E.P. Pharmacotherapy of Duchenne Muscular Dystrophy. Handb. Exp. Pharmacol. 2020, 261, 25–37. [Google Scholar] [CrossRef]
  382. Grages, S.M.; Bell, M.; Berlau, D.J. New and emerging pharmacotherapy for duchenne muscular dystrophy: A focus on synthetic therapeutics. Expert Opin. Pharmacother. 2020, 21, 841–851. [Google Scholar] [CrossRef]
  383. Deng, J.; Zhang, J.; Shi, K.; Liu, Z. Drug development progress in duchenne muscular dystrophy. Front. Pharmacol. 2022, 13, 950651. [Google Scholar] [CrossRef]
  384. Rawls, A.; Diviak, B.K.; Smith, C.I.; Severson, G.W.; Acosta, S.A.; Wilson-Rawls, J. Pharmacotherapeutic Approaches to Treatment of Muscular Dystrophies. Biomolecules 2023, 13, 1536. [Google Scholar] [CrossRef]
  385. Fortunato, F.; Rossi, R.; Falzarano, M.S.; Ferlini, A. Innovative Therapeutic Approaches for Duchenne Muscular Dystrophy. J. Clin. Med. 2021, 10, 820. [Google Scholar] [CrossRef]
  386. Markati, T.; Oskoui, M.; Farrar, M.A.; Duong, T.; Goemans, N.; Servais, L. Emerging therapies for Duchenne muscular dystrophy. Lancet Neurol. 2022, 21, 814–829. [Google Scholar] [CrossRef]
  387. Chung Liang, L.; Sulaiman, N.; Yazid, M.D. A Decade of Progress in Gene Targeted Therapeutic Strategies in Duchenne Muscular Dystrophy: A Systematic Review. Front. Bioeng. Biotechnol. 2022, 10, 833833. [Google Scholar] [CrossRef]
  388. Chang, M.; Cai, Y.; Gao, Z.; Chen, X.; Liu, B.; Zhang, C.; Yu, W.; Cao, Q.; Shen, Y.; Yao, X.; et al. Duchenne muscular dystrophy: Pathogenesis and promising therapies. J. Neurol. 2023, 270, 3733–3749. [Google Scholar] [CrossRef]
  389. Duan, D. Duchenne Muscular Dystrophy Gene Therapy in 2023: Status, Perspective, and Beyond. Hum. Gene Ther. 2023, 34, 345–349. [Google Scholar] [CrossRef]
  390. Clemens, P.R.; Rao, V.K.; Connolly, A.M.; Harper, A.D.; Mah, J.K.; Smith, E.C.; McDonald, C.M.; Zaidman, C.M.; Morgenroth, L.P.; Osaki, H.; et al. Safety, Tolerability, and Efficacy of Viltolarsen in Boys With Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A Phase 2 Randomized Clinical Trial. JAMA Neurol. 2020, 77, 982–991. [Google Scholar] [CrossRef]
  391. Novak, J.S.; Spathis, R.; Dang, U.J.; Fiorillo, A.A.; Hindupur, R.; Tully, C.B.; Mázala, D.A.G.; Canessa, E.; Brown, K.J.; Partridge, T.A.; et al. Interrogation of Dystrophin and Dystroglycan Complex Protein Turnover After Exon Skipping Therapy. J. Neuromuscul. Dis. 2021, 8, S383–S402. [Google Scholar] [CrossRef]
  392. Eser, G.; Topaloğlu, H. Current Outline of Exon Skipping Trials in Duchenne Muscular Dystrophy. Genes 2022, 13, 1241. [Google Scholar] [CrossRef]
  393. Mercuri, E.; Seferian, A.M.; Servais, L.; Deconinck, N.; Stevenson, H.; Ni, X.; Zhang, W.; East, L.; Yonren, S.; Muntoni, F.; et al. Safety, tolerability and pharmacokinetics of eteplirsen in young boys aged 6-48 months with Duchenne muscular dystrophy amenable to exon 51 skipping. Neuromuscul. Disord. 2023, 33, 476–483. [Google Scholar] [CrossRef]
  394. Min, Y.L.; Bassel-Duby, R.; Olson, E.N. CRISPR Correction of Duchenne Muscular Dystrophy. Annu. Rev. Med. 2019, 70, 239–255. [Google Scholar] [CrossRef]
  395. Kupatt, C.; Windisch, A.; Moretti, A.; Wolf, E.; Wurst, W.; Walter, M.C. Genome editing for Duchenne muscular dystrophy: A glimpse of the future? Gene Ther. 2021, 28, 542–548. [Google Scholar] [CrossRef]
  396. Chemello, F.; Olson, E.N.; Bassel-Duby, R. CRISPR- Editing Therapy for Duchenne Muscular Dystrophy. Hum. Gene Ther. 2023, 34, 379–387. [Google Scholar] [CrossRef]
  397. Politano, L. Read-through approach for stop mutations in Duchenne muscular dystrophy. An update. Acta Myol. 2021, 40, 43–50. [Google Scholar] [CrossRef]
  398. Seto, J.T.; Ramos, J.N.; Muir, L.; Chamberlain, J.S.; Odom, G.L. Gene replacement therapies for duchenne muscular dystrophy using adeno-associated viral vectors. Curr. Gene Ther. 2012, 12, 139–151. [Google Scholar] [CrossRef]
  399. Nance, M.E.; Duan, D. Perspective on Adeno-Associated Virus Capsid Modification for Duchenne Muscular Dystrophy Gene Therapy. Hum. Gene Ther. 2015, 26, 786–800. [Google Scholar] [CrossRef]
  400. Duan, D. Systemic AAV Micro-dystrophin Gene Therapy for Duchenne Muscular Dystrophy. Mol. Ther. 2018, 26, 2337–2356. [Google Scholar] [CrossRef]
  401. Mendell, J.R.; Sahenk, Z.; Lehman, K.; Nease, C.; Lowes, L.P.; Miller, N.F.; Iammarino, M.A.; Alfano, L.N.; Nicholl, A.; Al-Zaidy, S.; et al. Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children with Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial. JAMA Neurol. 2020, 77, 1122–1131. [Google Scholar] [CrossRef]
  402. Matsuzaka, Y.; Hirai, Y.; Hashido, K.; Okada, T. Therapeutic Application of Extracellular Vesicles-Capsulated Adeno-Associated Virus Vector via nSMase2/Smpd3, Satellite, and Immune Cells in Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2022, 23, 1551. [Google Scholar] [CrossRef] [PubMed]
  403. Loro, E.; Sengupta, K.; Bogdanovich, S.; Whig, K.; Schultz, D.C.; Huryn, D.M.; Khurana, T.S. High-throughput identification of post-transcriptional utrophin up-regulators for Duchenne muscle dystrophy (DMD) therapy. Sci. Rep. 2020, 10, 2132. [Google Scholar] [CrossRef]
  404. Soblechero-Martín, P.; López-Martínez, A.; de la Puente-Ovejero, L.; Vallejo-Illarramendi, A.; Arechavala-Gomeza, V. Utrophin modulator drugs as potential therapies for Duchenne and Becker muscular dystrophies. Neuropathol. Appl. Neurobiol. 2021, 47, 711–723. [Google Scholar] [CrossRef]
  405. Vuorinen, A.; Wilkinson, I.V.L.; Chatzopoulou, M.; Edwards, B.; Squire, S.E.; Fairclough, R.J.; Bazan, N.A.; Milner, J.A.; Conole, D.; Donald, J.R.; et al. Discovery and mechanism of action studies of 4,6-diphenylpyrimidine-2-carbohydrazides as utrophin modulators for the treatment of Duchenne muscular dystrophy. Eur. J. Med. Chem. 2021, 220, 113431. [Google Scholar] [CrossRef] [PubMed]
  406. Galli, F.; Mouly, V.; Butler-Browne, G.; Cossu, G. Challenges in cell transplantation for muscular dystrophy. Exp. Cell Res. 2021, 409, 112908. [Google Scholar] [CrossRef]
  407. Boyer, O.; Butler-Browne, G.; Chinoy, H.; Cossu, G.; Galli, F.; Lilleker, J.B.; Magli, A.; Mouly, V.; Perlingeiro, R.C.R.; Previtali, S.C.; et al. Myogenic Cell Transplantation in Genetic and Acquired Diseases of Skeletal Muscle. Front. Genet. 2021, 12, 702547. [Google Scholar] [CrossRef]
  408. Ausems, C.R.M.; van Engelen, B.G.M.; van Bokhoven, H.; Wansink, D.G. Systemic cell therapy for muscular dystrophies: The ultimate transplantable muscle progenitor cell and current challenges for clinical efficacy. Stem Cell Rev. Rep. 2021, 17, 878–899. [Google Scholar] [CrossRef]
  409. Saleh, K.K.; Switzler, C.; Hicks, M.R.; Gane, L.; Gibbs, D.E.; Pyle, A.D. Duchenne muscular dystrophy disease severity impacts skeletal muscle progenitor cells systemic delivery. Front. Physiol. 2023, 14, 1190524. [Google Scholar] [CrossRef]
  410. Anaya-Segura, M.A.; García-Martínez, F.A.; Montes-Almanza, L.A.; Díaz, B.G.; Avila-Ramírez, G.; Alvarez-Maya, I.; Coral-Vazquez, R.M.; Mondragón-Terán, P.; Escobar-Cedillo, R.E.; García-Calderón, N.; et al. Non-Invasive Biomarkers for Duchenne Muscular Dystrophy and Carrier Detection. Molecules 2015, 20, 11154–11172. [Google Scholar] [CrossRef]
  411. Fortunato, F.; Ferlini, A. Biomarkers in Duchenne Muscular Dystrophy: Current Status and Future Directions. J. Neuromuscul. Dis. 2023, 10, 987–1002. [Google Scholar] [CrossRef]
  412. 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]
  413. Ohlendieck, K. Proteomic identification of biomarkers of skeletal muscle disorders. Biomark. Med. 2013, 7, 169–186. [Google Scholar] [CrossRef]
  414. Joyce, N.C.; Oskarsson, B.; Jin, L.W. Muscle biopsy evaluation in neuromuscular disorders. Med. Rehabil. Clin. N. Am. 2012, 23, 609–631. [Google Scholar] [CrossRef] [PubMed]
  415. Meola, G.; Bugiardini, E.; Cardani, R. Muscle biopsy. J. Neurol. 2012, 259, 601–610. [Google Scholar] [CrossRef]
  416. Shanely, R.A.; Zwetsloot, K.A.; Triplett, N.T.; Meaney, M.P.; Farris, G.E.; Nieman, D.C. Human skeletal muscle biopsy procedures using the modified Bergström technique. J. Vis. Exp. 2014, 10, 51812. [Google Scholar] [CrossRef]
  417. Nix, J.S.; Moore, S.A. What Every Neuropathologist Needs to Know: The Muscle Biopsy. J. Neuropathol. Exp. Neurol. 2020, 79, 719–733. [Google Scholar] [CrossRef] [PubMed]
  418. Brancaccio, P.; Lippi, G.; Maffulli, N. Biochemical markers of muscular damage. Clin. Chem. Lab. Med. 2010, 48, 757–767. [Google Scholar] [CrossRef]
  419. Lippi, G.; Schena, F.; Ceriotti, F. Diagnostic biomarkers of muscle injury and exertional rhabdomyolysis. Clin. Chem. Lab. Med. 2018, 57, 175–182. [Google Scholar] [CrossRef]
  420. 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]
  421. Szigyarto, C.A.; Spitali, P. Biomarkers of Duchenne muscular dystrophy: Current findings. Degener. Neurol. Neuromuscul. Dis. 2018, 8, 1–13. [Google Scholar] [CrossRef]
  422. Grounds, M.D.; Terrill, J.R.; Al-Mshhdani, B.A.; Duong, M.N.; Radley-Crabb, H.G.; Arthur, P.G. Biomarkers for Duchenne muscular dystrophy: Myonecrosis, inflammation and oxidative stress. Dis. Model Mech. 2020, 13, dmm043638. [Google Scholar] [CrossRef] [PubMed]
  423. 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]
  424. Rossi, R.; Johansson, C.; Heywood, W.; Vinette, H.; Jensen, G.; Tegel, H.; Jiménez-Requena, A.; Torelli, S.; Al-Khalili Szigyarto, C.; Ferlini, A. A Proof of Principle Proteomic Study Detects Dystrophin in Human Plasma: Implications in DMD Diagnosis and Clinical Monitoring. Int. J. Mol. Sci. 2023, 24, 5215. [Google Scholar] [CrossRef] [PubMed]
  425. Ogundele, M.; Zhang, J.S.; Goswami, M.V.; Barbieri, M.L.; Dang, U.J.; Novak, J.S.; Hoffman, E.P.; Nagaraju, K.; Cinrg-Dnhs Investigators; Hathout, Y. Validation of Chemokine Biomarkers in Duchenne Muscular Dystrophy. Life 2021, 11, 827. [Google Scholar] [CrossRef] [PubMed]
  426. Johansson, C.; Hunt, H.; Signorelli, M.; Edfors, F.; Hober, A.; Svensson, A.S.; Tegel, H.; Forstström, B.; Aartsma-Rus, A.; Niks, E.; et al. Orthogonal proteomics methods warrant the development of Duchenne muscular dystrophy biomarkers. Clin. Proteom. 2023, 20, 23. [Google Scholar] [CrossRef] [PubMed]
  427. Rouillon, J.; Poupiot, J.; Zocevic, A.; Amor, F.; Léger, T.; Garcia, C.; Camadro, J.M.; Wong, B.; Pinilla, R.; Cosette, J.; et al. Serum proteomic profiling reveals fragments of MYOM3 as potential biomarkers for monitoring the outcome of therapeutic interventions in muscular dystrophies. Hum. Mol. Genet. 2015, 24, 4916–4932. [Google Scholar] [CrossRef]
  428. Rouillon, J.; Zocevic, A.; Leger, T.; Garcia, C.; Camadro, J.M.; Udd, B.; Wong, B.; Servais, L.; Voit, T.; Svinartchouk, F. Proteomics profiling of urine reveals specific titin fragments as biomarkers of Duchenne muscular dystrophy. Neuromuscul. Disord. 2014, 24, 563–573. [Google Scholar] [CrossRef]
  429. Robertson, A.S.; Majchrzak, M.J.; Smith, C.M.; Gagnon, R.C.; Devidze, N.; Banks, G.B.; Little, S.C.; Nabbie, F.; Bounous, D.I.; DiPiero, J.; et al. Dramatic elevation in urinary amino terminal titin fragment excretion quantified by immunoassay in Duchenne muscular dystrophy patients and in dystrophin deficient rodents. Neuromuscul. Disord. 2017, 27, 635–645. [Google Scholar] [CrossRef]
  430. Gargan, S.; Dowling, P.; Zweyer, M.; Swandulla, D.; Ohlendieck, K. Identification of marker proteins of muscular dystrophy in the urine proteome from the mdx-4cv model of dystrophinopathy. Mol. Omics 2020, 16, 268–278. [Google Scholar] [CrossRef]
  431. Matsuo, M.; Awano, H.; Maruyama, N.; Nishio, H. Titin fragment in urine: A noninvasive biomarker of muscle degradation. Adv. Clin. Chem. 2019, 90, 1–23. [Google Scholar] [CrossRef]
  432. Ishii, M.N.; Nakashima, M.; Kamiguchi, H.; Zach, N.; Kuboki, R.; Baba, R.; Hirakawa, T.; Suzuki, K.; Quinton, M. Urine titin as a novel biomarker for Duchenne muscular dystrophy. Neuromuscul. Disord. 2023, 33, 302–308. [Google Scholar] [CrossRef]
  433. Murphy, S.; Zweyer, M.; Mundegar, R.R.; Swandulla, D.; Ohlendieck, K. Proteomic identification of elevated saliva kallikrein levels in the mdx-4cv mouse model of Duchenne muscular dystrophy. Biochem. Biophys. Rep. 2018, 18, 100541. [Google Scholar] [CrossRef] [PubMed]
  434. Olivier, M.; Asmis, R.; Hawkins, G.A.; Howard, T.D.; Cox, L.A. The Need for Multi-Omics Biomarker Signatures in Precision Medicine. Int. J. Mol. Sci. 2019, 20, 4781. [Google Scholar] [CrossRef] [PubMed]
  435. Wang, R.C.; Wang, Z. Precision Medicine: Disease Subtyping and Tailored Treatment. Cancers 2023, 15, 3837. [Google Scholar] [CrossRef]
  436. Bernas, T.; Grégori, G.; Asem, E.K.; Robinson, J.P. Integrating cytomics and proteomics. Mol. Cell. Proteom. 2006, 5, 2–13. [Google Scholar] [CrossRef]
  437. Gomase, V.S.; Tagore, S. Cytomics. Curr. Drug Metab. 2008, 9, 263–266. [Google Scholar] [CrossRef]
  438. Larance, M.; Lamond, A.I. Multidimensional proteomics for cell biology. Nat. Rev. Mol. Cell Biol. 2015, 16, 269–280. [Google Scholar] [CrossRef]
  439. O’Connor, L.M.; O’Connor, B.A.; Lim, S.B.; Zeng, J.; Lo, C.H. Integrative multi-omics and systems bioinformatics in translational neuroscience: A data mining perspective. J. Pharm. Anal. 2023, 13, 836–850. [Google Scholar] [CrossRef]
  440. Alto, S.I.; Chang, C.N.; Brown, K.; Kioussi, C.; Filtz, T.M. Gene Expression Profiling of Skeletal Muscles. Genes 2021, 12, 1718. [Google Scholar] [CrossRef]
  441. Erbe, L.S.; Hoffjan, S.; Janßen, S.; Kneifel, M.; Krause, K.; Gerding, W.M.; Döring, K.; Güttsches, A.K.; Roos, A.; Buena Atienza, E.; et al. Exome Sequencing and Optical Genome Mapping in Molecularly Unsolved Cases of Duchenne Muscular Dystrophy: Identification of a Causative X-Chromosomal Inversion Disrupting the DMD Gene. Int. J. Mol. Sci. 2023, 24, 14716. [Google Scholar] [CrossRef]
  442. Pluta, N.; von Moers, A.; Pechmann, A.; Stenzel, W.; Goebel, H.H.; Atlan, D.; Wolf, B.; Nanda, I.; Zaum, A.K.; Rost, S. Whole-Genome Sequencing Identified New Structural Variations in the DMD Gene That Cause Duchenne Muscular Dystrophy in Two Girls. Int. J. Mol. Sci. 2023, 24, 13567. [Google Scholar] [CrossRef]
  443. Baßler, K.; Günther, P.; Schulte-Schrepping, J.; Becker, M.; Biernat, P. A Bioinformatic Toolkit for Single-Cell mRNA Analysis. Methods Mol. Biol. 2019, 1979, 433–455. [Google Scholar] [CrossRef]
  444. Stark, R.; Grzelak, M.; Hadfield, J. RNA sequencing: The teenage years. Nat. Rev. Genet. 2019, 20, 631–656. [Google Scholar] [CrossRef]
  445. Rao, A.; Barkley, D.; França, G.S.; Yanai, I. Exploring tissue architecture using spatial transcriptomics. Nature 2021, 596, 211–220. [Google Scholar] [CrossRef]
  446. Sandonà, M.; Saccone, V. Post-translational Modification in Muscular Dystrophies. Adv. Exp. Med. Biol. 2022, 1382, 71–84. [Google Scholar] [CrossRef]
  447. Iwasaki, T.; Terrill, J.R.; Kawarai, K.; Miyata, Y.; Tagami, T.; Maeda, N.; Hasegawa, Y.; Watanabe, T.; Grounds, M.D.; Arthur, P.G. The location of protein oxidation in dystrophic skeletal muscle from the mdx mouse model of Duchenne muscular dystrophy. Acta Histochem. 2022, 124, 151959. [Google Scholar] [CrossRef]
  448. Xu, H.; Cai, X.; Xu, K.; Wu, Q.; Xu, B. The metabolomic plasma profile of patients with Duchenne muscular dystrophy: Providing new evidence for its pathogenesis. Orphanet J. Rare Dis. 2023, 18, 273. [Google Scholar] [CrossRef]
  449. Dabaj, I.; Ferey, J.; Marguet, F.; Gilard, V.; Basset, C.; Bahri, Y.; Brehin, A.C.; Vanhulle, C.; Leturcq, F.; Marret, S.; et al. Muscle metabolic remodelling patterns in Duchenne muscular dystrophy revealed by ultra-high-resolution mass spectrometry imaging. Sci. Rep. 2021, 11, 1906. [Google Scholar] [CrossRef]
  450. Tsonaka, R.; Signorelli, M.; Sabir, E.; Seyer, A.; Hettne, K.; Aartsma-Rus, A.; Spitali, P. Longitudinal metabolomic analysis of plasma enables modeling disease progression in Duchenne muscular dystrophy mouse models. Hum. Mol. Genet. 2020, 29, 745–755. [Google Scholar] [CrossRef]
  451. Milad, N.; White, Z.; Tehrani, A.Y.; Sellers, S.; Rossi, F.M.V.; Bernatchez, P. Increased plasma lipid levels exacerbate muscle pathology in the mdx mouse model of Duchenne muscular dystrophy. Skelet. Muscle 2017, 7, 19. [Google Scholar] [CrossRef]
  452. Tsonaka, R.; Seyer, A.; Aartsma-Rus, A.; Spitali, P. Plasma lipidomic analysis shows a disease progression signature in mdx mice. Sci. Rep. 2021, 11, 12993. [Google Scholar] [CrossRef]
  453. Rancourt, A.; Dufresne, S.S.; St-Pierre, G.; Lévesque, J.C.; Nakamura, H.; Kikuchi, Y.; Satoh, M.S.; Frenette, J.; Sato, S. Galectin-3 and N-acetylglucosamine promote myogenesis and improve skeletal muscle function in the mdx model of Duchenne muscular dystrophy. FASEB J. 2018, 32, 6445. [Google Scholar] [CrossRef] [PubMed]
  454. Bonaguro, L.; Schulte-Schrepping, J.; Ulas, T.; Aschenbrenner, A.C.; Beyer, M.; Schultze, J.L. A guide to systems-level immunomics. Nat. Immunol. 2022, 23, 1412–1423. [Google Scholar] [CrossRef] [PubMed]
  455. Wei, W.; Riley, N.M.; Yang, A.C.; Kim, J.T.; Terrell, S.M.; Li, V.L.; Garcia-Contreras, M.; Bertozzi, C.R.; Long, J.Z. Cell type-selective secretome profiling in vivo. Nat. Chem. Biol. 2021, 17, 326–334. [Google Scholar] [CrossRef]
  456. Coulton, G. Are histochemistry and cytochemistry ‘Omics’? J. Mol. Histol. 2004, 35, 603–613. [Google Scholar] [CrossRef]
  457. Robinson, J.P.; Ostafe, R.; Iyengar, S.N.; Rajwa, B.; Fischer, R. Flow Cytometry: The Next Revolution. Cells 2023, 12, 1875. [Google Scholar] [CrossRef] [PubMed]
  458. 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]
  459. 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]
  460. Starosta, A.; Konieczny, P. Therapeutic aspects of cell signaling and communication in Duchenne muscular dystrophy. Cell. Mol. Life Sci. 2021, 78, 4867–4891. [Google Scholar] [CrossRef]
  461. Mund, A.; Coscia, F.; Kriston, A.; Hollandi, R.; Kovács, F.; Brunner, A.D.; Migh, E.; Schweizer, L.; Santos, A.; Bzorek, M.; et al. Deep Visual Proteomics defines single-cell identity and heterogeneity. Nat. Biotechnol. 2022, 40, 1231–1240. [Google Scholar] [CrossRef]
  462. Rosenberger, F.A.; Thielert, M.; Strauss, M.T.; Schweizer, L.; Ammar, C.; Mädler, S.C.; Metousis, A.; Skowronek, P.; Wahle, M.; Madden, K.; et al. Spatial single-cell mass spectrometry defines zonation of the hepatocyte proteome. Nat. Methods 2023, 20, 1530–1536. [Google Scholar] [CrossRef] [PubMed]
  463. Kelsey, G.; Stegle, O.; Reik, W. Single-cell epigenomics: Recording the past and predicting the future. Science 2017, 358, 69–75. [Google Scholar] [CrossRef] [PubMed]
  464. Wang, K.C.; Chang, H.Y. Epigenomics: Technologies and Applications. Circ. Res. 2018, 122, 1191–1199. [Google Scholar] [CrossRef] [PubMed]
  465. Pinheiro, I.; Torres-Padilla, M.E.; Almouzni, G. Epigenomics in the single cell era, an important read out for genome function and cell identity. Epigenomics 2021, 13, 981–984. [Google Scholar] [CrossRef]
  466. Li, Y. Modern epigenetics methods in biological research. Methods 2021, 187, 104–113. [Google Scholar] [CrossRef]
  467. Robinson, D.C.L.; Dilworth, F.J. Epigenetic Regulation of Adult Myogenesis. Curr. Top. Dev. Biol. 2018, 126, 235–284. [Google Scholar] [CrossRef]
  468. Plaza-Diaz, J.; Izquierdo, D.; Torres-Martos, Á.; Baig, A.T.; Aguilera, C.M.; Ruiz-Ojeda, F.J. Impact of Physical Activity and Exercise on the Epigenome in Skeletal Muscle and Effects on Systemic Metabolism. Biomedicines 2022, 10, 126. [Google Scholar] [CrossRef]
  469. Massenet, J.; Gardner, E.; Chazaud, B.; Dilworth, F.J. Epigenetic regulation of satellite cell fate during skeletal muscle regeneration. Skelet. Muscle 2021, 11, 4. [Google Scholar] [CrossRef]
  470. Consalvi, S.; Saccone, V.; Mozzetta, C. Histone deacetylase inhibitors: A potential epigenetic treatment for Duchenne muscular dystrophy. Epigenomics 2014, 6, 547–560. [Google Scholar] [CrossRef]
  471. Schreyer, L.; Reilly, J.; McConkey, H.; Kerkhof, J.; Levy, M.A.; Hu, J.; Hnaini, M.; Sadikovic, B.; Campbell, C. The discovery of the DNA methylation episignature for Duchenne muscular dystrophy. Neuromuscul. Disord. 2023, 33, 5–14. [Google Scholar] [CrossRef]
  472. Sandonà, M.; Cavioli, G.; Renzini, A.; Cedola, A.; Gigli, G.; Coletti, D.; McKinsey, T.A.; Moresi, V.; Saccone, V. Histone Deacetylases: Molecular Mechanisms and Therapeutic Implications for Muscular Dystrophies. Int. J. Mol. Sci. 2023, 24, 4306. [Google Scholar] [CrossRef]
  473. Chen, C.; Wang, J.; Pan, D.; Wang, X.; Xu, Y.; Yan, J.; Wang, L.; Yang, X.; Yang, M.; Liu, G.P. Applications of multi-omics analysis in human diseases. MedComm 2023, 4, e315. [Google Scholar] [CrossRef] [PubMed]
  474. Tavassoly, I.; Goldfarb, J.; Iyengar, R. Systems biology primer: The basic methods and approaches. Essays Biochem. 2018, 62, 487–500. [Google Scholar] [CrossRef] [PubMed]
  475. Greising, S.M.; Gransee, H.M.; Mantilla, C.B.; Sieck, G.C. Systems biology of skeletal muscle: Fiber type as an organizing principle. Wiley Interdiscip. Rev. Syst. Biol. Med. 2012, 4, 457–473. [Google Scholar] [CrossRef] [PubMed]
  476. Heier, C.R.; Zhang, A.; Nguyen, N.Y.; Tully, C.B.; Panigrahi, A.; Gordish-Dressman, H.; Pandey, S.N.; Guglieri, M.; Ryan, M.M.; Clemens, P.R.; et al. Multi-Omics Identifies Circulating miRNA and Protein Biomarkers for Facioscapulohumeral Dystrophy. J. Pers. Med. 2020, 10, 236. [Google Scholar] [CrossRef]
  477. Liu, J.C.; Dong, S.S.; Shen, H.; Yang, D.Y.; Chen, B.B.; Ma, X.Y.; Peng, Y.R.; Xiao, H.M.; Deng, H.W. Multi-omics research in sarcopenia: Current progress and future prospects. Ageing Res. Rev. 2022, 76, 101576. [Google Scholar] [CrossRef] [PubMed]
  478. McCourt, J.L.; Stearns-Reider, K.M.; Mamsa, H.; Kannan, P.; Afsharinia, M.H.; Shu, C.; Gibbs, E.M.; Shin, K.M.; Kurmangaliyev, Y.Z.; Schmitt, L.R.; et al. Multi-omics analysis of sarcospan overexpression in mdx skeletal muscle reveals compensatory remodeling of cytoskeleton-matrix interactions that promote mechanotransduction pathways. Skelet. Muscle 2023, 13, 1. [Google Scholar] [CrossRef]
  479. 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]
  480. Van Pelt, D.W.; Kharaz, Y.A.; Sarver, D.C.; Eckhardt, L.R.; Dzierzawski, J.T.; Disser, N.P.; Piacentini, A.N.; Comerford, E.; McDonagh, B.; Mendias, C.L. Multiomics analysis of the mdx/mTR mouse model of Duchenne muscular dystrophy. Connect. Tissue Res. 2021, 62, 24–39. [Google Scholar] [CrossRef]
  481. Elasbali, A.M.; Al-Soud, W.A.; Adnan, M.; Alhassan, H.H.; Mohammad, T.; Hassan, M.I. Discovering Promising Biomarkers and Therapeutic Targets for Duchenne Muscular Dystrophy: A Multiomics Meta-Analysis Approach. Mol. Neurobiol. 2024. [Google Scholar] [CrossRef]
  482. Signorelli, M.; Tsonaka, R.; Aartsma-Rus, A.; Spitali, P. Multiomic characterization of disease progression in mice lacking dystrophin. PLoS ONE 2023, 18, e0283869. [Google Scholar] [CrossRef] [PubMed]
  483. Wu, X.; Dong, N.; Yu, L.; Liu, M.; Jiang, J.; Tang, T.; Zhao, H.; Fang, Q. Identification of immune-related features involved in Duchenne muscular dystrophy: A bidirectional transcriptome and proteome-driven analysis. Front. Immunol. 2022, 13, 1017423. [Google Scholar] [CrossRef] [PubMed]
  484. Mournetas, V.; Massouridès, E.; Dupont, J.B.; Kornobis, E.; Polvèche, H.; Jarrige, M.; Dorval, A.R.L.; Gosselin, M.R.F.; Manousopoulou, A.; Garbis, S.D.; et al. Myogenesis modelled by human pluripotent stem cells: A multi-omic study of Duchenne myopathy early onset. J. Cachexia Sarcopenia Muscle 2021, 12, 209–232. [Google Scholar] [CrossRef]
  485. Wijekoon, N.; Gonawala, L.; Ratnayake, P.; Dissanayaka, P.; Gunarathne, I.; Amaratunga, D.; Liyanage, R.; Senanayaka, S.; Wijesekara, S.; Gunasekara, H.H.; et al. Integrated genomic, proteomic and cognitive assessment in Duchenne Muscular Dystrophy suggest astrocyte centric pathology. Heliyon 2023, 9, e18530. [Google Scholar] [CrossRef] [PubMed]
  486. 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]
  487. Argelaguet, R.; Velten, B.; Arnol, D.; Dietrich, S.; Zenz, T.; Marioni, J.C.; Buettner, F.; Huber, W.; Stegle, O. Multi-Omics Factor Analysis-a framework for unsupervised integration of multi-omics data sets. Mol. Syst. Biol. 2018, 14, e8124. [Google Scholar] [CrossRef]
  488. Mann, M.; Kumar, C.; Zeng, W.F.; Strauss, M.T. Artificial intelligence for proteomics and biomarker discovery. Cell Syst. 2021, 12, 759–770. [Google Scholar] [CrossRef]
  489. Vera, C.D.; Zhang, A.; Pang, P.D.; Wu, J.C. Treating Duchenne Muscular Dystrophy: The Promise of Stem Cells, Artificial Intelligence, and Multi-Omics. Front Cardiovasc. Med. 2022, 9, 851491. [Google Scholar] [CrossRef]
  490. Torun, F.M.; Virreira Winter, S.; Doll, S.; Riese, F.M.; Vorobyev, A.; Mueller-Reif, J.B.; Geyer, P.E.; Strauss, M.T. Transparent Exploration of Machine Learning for Biomarker Discovery from Proteomics and Omics Data. J. Proteome Res. 2023, 22, 359–367. [Google Scholar] [CrossRef]
  491. Procopio, A.; Cesarelli, G.; Donisi, L.; Merola, A.; Amato, F.; Cosentino, C. Combined mechanistic modeling and machine-learning approaches in systems biology—A systematic literature review. Comput. Methods Programs Biomed. 2023, 240, 107681. [Google Scholar] [CrossRef]
  492. Fortunato, F.; Tonelli, L.; Farnè, M.; Selvatici, R.; Ferlini, A. DMD deletions underlining mild dystrophinopathies: Literature review highlights phenotype-related mutation clusters and provides insights about genetic mechanisms and prognosis. Front. Neurol. 2024, 14, 1288721. [Google Scholar] [CrossRef]
  493. Marques, M.J.; Ferretti, R.; Vomero, V.U.; Minatel, E.; Neto, H.S. Intrinsic laryngeal muscles are spared from myonecrosis in the mdx mouse model of Duchenne muscular dystrophy. Muscle Nerve 2007, 35, 349–353. [Google Scholar] [CrossRef] [PubMed]
  494. Lorena, M.D.S.V.; Santos, E.K.D.; Ferretti, R.; Nagana Gowda, G.A.; Odom, G.L.; Chamberlain, J.S.; Matsumura, C.Y. Biomarkers for Duchenne muscular dystrophy progression: Impact of age in the mdx tongue spared muscle. Skelet. Muscle 2023, 13, 16. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Outline of the main proteomic approaches that are routinely utilized to isolate, identify and characterize proteins and their individual proteoforms. Abbreviations used: 1D, one-dimensional; 2D, two-dimensional; BN-PAGE, blue native polyacrylamide gel electrophoresis; CBB, Coomassie Brilliant Blue; DC, differential centrifugation; DDA, data-dependent acquisition; DIA, data-independent acquisition; DIGE, difference gel electrophoresis; eFASP, enhanced filter-aided sample preparation; ELISA, enzyme-linked immunosorbent assay; EM, electron microscopy; ESI, electrospray ionization; FASP, filter-aided sample preparation; FT, Fourier-transform ion cyclotron resonance; GE, gel electrophoresis; GeLC, gel electrophoresis–liquid chromatography; IB, immunoblotting; ICAT, isotope-coded affinity tags; ICPL, isotope-coded protein labeling; IFM, immunofluorescence microscopy; IHC, immunohistochemistry; iST, In-StageTip; LC, liquid chromatography; IMAC, immobilized metal affinity chromatography; iTRAQ, isobaric tagging for relative and absolute quantitation; LFQ, label-free quantification; MALDI, matrix-assisted laser desorption/ionization; MS, mass spectrometry; MudPIT; multi-dimensional protein identification technology; PRM, parallel reaction monitoring; PTMs, post-translational modifications; SCP, single-cell proteomics; SILAC, stable isotope labeling by amino acids in cell culture; SiMoA, Simoa bead-based immunoassay; SP3, single-pot solid-phase-enhanced sample preparation; SRM/MRM, selected/multiple reaction monitoring; S-Trap, suspension trapping; SWATH, Sequential Window Acquisition of all Theoretical Mass Spectra; TDA, targeted data acquisition; TMT, isobaric tandem mass tagging; ToF, time-of-flight; USP3, universal solid-phase protein preparation; XL, cross-linking.
Figure 1. Outline of the main proteomic approaches that are routinely utilized to isolate, identify and characterize proteins and their individual proteoforms. Abbreviations used: 1D, one-dimensional; 2D, two-dimensional; BN-PAGE, blue native polyacrylamide gel electrophoresis; CBB, Coomassie Brilliant Blue; DC, differential centrifugation; DDA, data-dependent acquisition; DIA, data-independent acquisition; DIGE, difference gel electrophoresis; eFASP, enhanced filter-aided sample preparation; ELISA, enzyme-linked immunosorbent assay; EM, electron microscopy; ESI, electrospray ionization; FASP, filter-aided sample preparation; FT, Fourier-transform ion cyclotron resonance; GE, gel electrophoresis; GeLC, gel electrophoresis–liquid chromatography; IB, immunoblotting; ICAT, isotope-coded affinity tags; ICPL, isotope-coded protein labeling; IFM, immunofluorescence microscopy; IHC, immunohistochemistry; iST, In-StageTip; LC, liquid chromatography; IMAC, immobilized metal affinity chromatography; iTRAQ, isobaric tagging for relative and absolute quantitation; LFQ, label-free quantification; MALDI, matrix-assisted laser desorption/ionization; MS, mass spectrometry; MudPIT; multi-dimensional protein identification technology; PRM, parallel reaction monitoring; PTMs, post-translational modifications; SCP, single-cell proteomics; SILAC, stable isotope labeling by amino acids in cell culture; SiMoA, Simoa bead-based immunoassay; SP3, single-pot solid-phase-enhanced sample preparation; SRM/MRM, selected/multiple reaction monitoring; S-Trap, suspension trapping; SWATH, Sequential Window Acquisition of all Theoretical Mass Spectra; TDA, targeted data acquisition; TMT, isobaric tandem mass tagging; ToF, time-of-flight; USP3, universal solid-phase protein preparation; XL, cross-linking.
Proteomes 12 00004 g001
Figure 2. Body-wide signaling axes that involve the neuromuscular system. Shown are major patterns of organ crosstalk that involve voluntary muscles, the central nervous system, bone, liver, kidneys, the gastrointestinal tract, fatty tissue, the immune system, the lungs and the heart. At the level of the neuromuscular system, intensive signaling events occur between matured myofibers and their innervating motoneuron, as well as between contractile fibers and their environment consisting of the extracellular matrix and stem cells that can be activated to form myogenic precursors during repair processes. Major signaling factors that are involved in muscle adaptations, myofiber repair, muscle–bone interactions, activation of the muscle-associated immune response, metabolic regulation and bioenergetic processes include myokines, which constitute the dynamic skeletal muscle secretome, as well as a variety of hormones, growth factors, cytokines, osteokines and adipokines.
Figure 2. Body-wide signaling axes that involve the neuromuscular system. Shown are major patterns of organ crosstalk that involve voluntary muscles, the central nervous system, bone, liver, kidneys, the gastrointestinal tract, fatty tissue, the immune system, the lungs and the heart. At the level of the neuromuscular system, intensive signaling events occur between matured myofibers and their innervating motoneuron, as well as between contractile fibers and their environment consisting of the extracellular matrix and stem cells that can be activated to form myogenic precursors during repair processes. Major signaling factors that are involved in muscle adaptations, myofiber repair, muscle–bone interactions, activation of the muscle-associated immune response, metabolic regulation and bioenergetic processes include myokines, which constitute the dynamic skeletal muscle secretome, as well as a variety of hormones, growth factors, cytokines, osteokines and adipokines.
Proteomes 12 00004 g002
Figure 3. Overview of the main approaches used in integrative proteomics. Listed are organ-specific proteomics that focuses on the analysis of individual skeletal muscles, system-specific proteomics for the in-depth analysis of the nerve–muscle connection and motor units, and subproteomics that centers on the mass spectrometric characterization of distinct organelles and supramolecular protein assemblies. Organ-crosstalk proteomics is concerned with the analysis of the circulatory biofluid proteome and how the release of myokines affects other organ systems in the body.
Figure 3. Overview of the main approaches used in integrative proteomics. Listed are organ-specific proteomics that focuses on the analysis of individual skeletal muscles, system-specific proteomics for the in-depth analysis of the nerve–muscle connection and motor units, and subproteomics that centers on the mass spectrometric characterization of distinct organelles and supramolecular protein assemblies. Organ-crosstalk proteomics is concerned with the analysis of the circulatory biofluid proteome and how the release of myokines affects other organ systems in the body.
Proteomes 12 00004 g003
Figure 4. The pathoproteomic profile of multi-system changes in Duchenne muscular dystrophy. The diagram outlines the complexity of body-wide alterations due to dystrophin deficiency and illustrates how the systematic application of a comprehensive interproteomic profiling approach could help us better understand the multi-system dysfunction in dystrophinopathy.
Figure 4. The pathoproteomic profile of multi-system changes in Duchenne muscular dystrophy. The diagram outlines the complexity of body-wide alterations due to dystrophin deficiency and illustrates how the systematic application of a comprehensive interproteomic profiling approach could help us better understand the multi-system dysfunction in dystrophinopathy.
Proteomes 12 00004 g004
Figure 5. Overview of biomarker candidates of dystrophinopathy as determined by biochemical screening and mass spectrometry-based proteomics. Listed are both tissue-associated changes and biofluid-related alterations in specific protein species. Skeletal muscle tissue markers are categorized according to their involvement in myofiber degeneration, abnormal calcium handling, regeneration and repair mechanisms, compensatory processes, chronic inflammation and macrophage invasion, the cellular stress response and reactive myofibrosis. Individual muscle-derived biofluid markers were identified as intact proteins or fragments, as in the case of the giant protein titin. Non-skeletal muscle markers are listed for proteomic changes in the brain, heart, liver, kidney, stomach and spleen. Non-muscle biofluid markers are described for the differential analysis of changes in skeletal muscle versus the heart, the analysis of the liver and functional evaluation of the kidneys. Increases versus decreases in biomarkers are marked by green and red arrows, respectively. Abbreviations used: ACSM, acyl-CoA synthetase medium chain; ADAM, disintegrin and metalloproteinase domain-containing protein; AGER, advanced glycosylation end-product specific receptor; AK, adenylate kinase; ALT, alanine aminotransaminase; ANGP, angiopoietin; ASPN, asporin; AST, aspartate transaminase; ANXA, annexin A; BNG, biglycan; BOC, brother of CDO (CAM-related/down-regulated by oncogenes); C5-b-C6, complement components; CA, carbonic anhydrase; CALU, calumenin; CAMK, Ca2+/calmodulin-dependent protein kinase; CCL, C-C motif chemokine ligand; CD34, hematopoietic progenitor cell antigen CD34; CF, coagulation factor; CDH, cadherin; CKM, creatine kinase, muscle type; CNS, central nervous system; GLUD1, glutamate dehydrogenase; CNTN, contactin; COL, collagen; CSQ, calsequestrin; CSRP, cysteine- and glycine-rich protein; CST, cystatin; cTNI, cardiac troponin I; CTSB, cathepsin B; CXCL, C-X-C motif chemokine ligand; DES, desmin; DG, dystroglycan; Dp427, dystrophin; DPT, dermatopontin; DYSF, dysferlin; ETFA, electron transfer flavoprotein subunit alpha; FABP, fatty acid-binding protein; FGG, fibrinogen gamma chain; FN, fibronectin; FTH, ferritin heavy chain; GDF, growth differentiation factor; GFAP, glial fibrillary acidic protein; GP, glycoprotein; HP, haptoglobin; Hsp, heat shock protein; IL, interleukin; KLK, kallikrein; LAM, laminin; LAMP, lysosomal-associated membrane protein; LEP, leptin; LDH, lactate dehydrogenase; LUM, lumican; LYZ, lysozyme; MAPK, mitogen-activated protein kinase; MB, myoglobin; MDH, malate dehydrogenase; MLC, myosin light chain; MMP, matrix metallo-proteinase; MSTN, myoststin; MTTP, microsomal triglyceride transfer protein; MYOM, myomesin; MYOF, myoferlin; OMD, osteomodulin; POSTN, periostin; RPTN, repetin; SAR, sarcalumenin; SG, sarcoglycan; SPP, secreted phosphoprotein; ST, suppression of tumorigenicity; TGF, transforming growth factor; TGM, transglutaminase; TIMP, tissue inhibitor of metalloproteinase; TNNI, troponin subunit I; TNFR, tumor necrosis factor receptor; TTN, titin; TUBA/B, tubulin chains A and B; UMOD, uromodulin; VIM, vimentin.
Figure 5. Overview of biomarker candidates of dystrophinopathy as determined by biochemical screening and mass spectrometry-based proteomics. Listed are both tissue-associated changes and biofluid-related alterations in specific protein species. Skeletal muscle tissue markers are categorized according to their involvement in myofiber degeneration, abnormal calcium handling, regeneration and repair mechanisms, compensatory processes, chronic inflammation and macrophage invasion, the cellular stress response and reactive myofibrosis. Individual muscle-derived biofluid markers were identified as intact proteins or fragments, as in the case of the giant protein titin. Non-skeletal muscle markers are listed for proteomic changes in the brain, heart, liver, kidney, stomach and spleen. Non-muscle biofluid markers are described for the differential analysis of changes in skeletal muscle versus the heart, the analysis of the liver and functional evaluation of the kidneys. Increases versus decreases in biomarkers are marked by green and red arrows, respectively. Abbreviations used: ACSM, acyl-CoA synthetase medium chain; ADAM, disintegrin and metalloproteinase domain-containing protein; AGER, advanced glycosylation end-product specific receptor; AK, adenylate kinase; ALT, alanine aminotransaminase; ANGP, angiopoietin; ASPN, asporin; AST, aspartate transaminase; ANXA, annexin A; BNG, biglycan; BOC, brother of CDO (CAM-related/down-regulated by oncogenes); C5-b-C6, complement components; CA, carbonic anhydrase; CALU, calumenin; CAMK, Ca2+/calmodulin-dependent protein kinase; CCL, C-C motif chemokine ligand; CD34, hematopoietic progenitor cell antigen CD34; CF, coagulation factor; CDH, cadherin; CKM, creatine kinase, muscle type; CNS, central nervous system; GLUD1, glutamate dehydrogenase; CNTN, contactin; COL, collagen; CSQ, calsequestrin; CSRP, cysteine- and glycine-rich protein; CST, cystatin; cTNI, cardiac troponin I; CTSB, cathepsin B; CXCL, C-X-C motif chemokine ligand; DES, desmin; DG, dystroglycan; Dp427, dystrophin; DPT, dermatopontin; DYSF, dysferlin; ETFA, electron transfer flavoprotein subunit alpha; FABP, fatty acid-binding protein; FGG, fibrinogen gamma chain; FN, fibronectin; FTH, ferritin heavy chain; GDF, growth differentiation factor; GFAP, glial fibrillary acidic protein; GP, glycoprotein; HP, haptoglobin; Hsp, heat shock protein; IL, interleukin; KLK, kallikrein; LAM, laminin; LAMP, lysosomal-associated membrane protein; LEP, leptin; LDH, lactate dehydrogenase; LUM, lumican; LYZ, lysozyme; MAPK, mitogen-activated protein kinase; MB, myoglobin; MDH, malate dehydrogenase; MLC, myosin light chain; MMP, matrix metallo-proteinase; MSTN, myoststin; MTTP, microsomal triglyceride transfer protein; MYOM, myomesin; MYOF, myoferlin; OMD, osteomodulin; POSTN, periostin; RPTN, repetin; SAR, sarcalumenin; SG, sarcoglycan; SPP, secreted phosphoprotein; ST, suppression of tumorigenicity; TGF, transforming growth factor; TGM, transglutaminase; TIMP, tissue inhibitor of metalloproteinase; TNNI, troponin subunit I; TNFR, tumor necrosis factor receptor; TTN, titin; TUBA/B, tubulin chains A and B; UMOD, uromodulin; VIM, vimentin.
Proteomes 12 00004 g005
Figure 6. Integromic analysis of dysfunctions in Duchenne muscular dystrophy. Multi-omics-based investigations are envisaged to generate a more comprehensive understanding of the molecular and cellular complexity of the pathogenic mechanisms that are involved in dystrophinopathy.
Figure 6. Integromic analysis of dysfunctions in Duchenne muscular dystrophy. Multi-omics-based investigations are envisaged to generate a more comprehensive understanding of the molecular and cellular complexity of the pathogenic mechanisms that are involved in dystrophinopathy.
Proteomes 12 00004 g006
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.; Trollet, C.; Negroni, E.; Swandulla, D.; Ohlendieck, K. How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction? Proteomes 2024, 12, 4. https://doi.org/10.3390/proteomes12010004

AMA Style

Dowling P, Trollet C, Negroni E, Swandulla D, Ohlendieck K. How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction? Proteomes. 2024; 12(1):4. https://doi.org/10.3390/proteomes12010004

Chicago/Turabian Style

Dowling, Paul, Capucine Trollet, Elisa Negroni, Dieter Swandulla, and Kay Ohlendieck. 2024. "How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction?" Proteomes 12, no. 1: 4. https://doi.org/10.3390/proteomes12010004

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