Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Methods Mol Biol ; 2596: 245-263, 2023.
Article En | MEDLINE | ID: mdl-36378444

The global analysis of the proteome is an important tool in cell biology. Comparative proteomic evaluations can identify and compare the composition, dynamics, and modifications between different samples. Comparing tissue proteomes under different conditions is crucial for advancing the biomedical field. Fluorescence two-dimensional difference gel electrophoresis (2D-DIGE) is a sensitive and robust biochemical method that can compare multiple protein samples over a broad dynamic range on the same analytical gel and can be used to establish differentially expressed protein profiles between different sample groups. 2D-DIGE involves fluorescently labeling protein samples with CyDye flours, via a two-dye or a three-dye system, pre-separation by isoelectric point, and molecular weight. DIGE circumvents gel-to-gel variability by multiplexing samples to a single gel and through the use of a pooled internal standard for normalization, thus enabling accurate high-resolution analysis of differences in protein abundance between samples. This chapter discusses 2D-DIGE as a comparative tissue proteomic technique and describes in detail the experimental steps required for comparative proteomic analysis employing both options of two-dye and three-dye DIGE minimal labeling.


Proteomics , Testis , Male , Humans , Proteomics/methods , Two-Dimensional Difference Gel Electrophoresis/methods , Proteome , Isoelectric Point , Electrophoresis, Gel, Two-Dimensional/methods
2.
JCI Insight ; 6(12)2021 06 22.
Article En | MEDLINE | ID: mdl-34032637

Evolutionarily conserved signaling intermediate in Toll pathways (ECSIT) is a protein with roles in early development, activation of the transcription factor NF-κB, and production of mitochondrial reactive oxygen species (mROS) that facilitates clearance of intracellular bacteria like Salmonella. ECSIT is also an important assembly factor for mitochondrial complex I. Unlike the murine form of Ecsit (mEcsit), we demonstrate here that human ECSIT (hECSIT) is highly labile. To explore whether the instability of hECSIT affects functions previously ascribed to its murine counterpart, we created a potentially novel transgenic mouse in which the murine Ecsit gene is replaced by the human ECSIT gene. The humanized mouse has low levels of hECSIT protein, in keeping with its intrinsic instability. Whereas low-level expression of hECSIT was capable of fully compensating for mEcsit in its roles in early development and activation of the NF-κB pathway, macrophages from humanized mice showed impaired clearance of Salmonella that was associated with reduced production of mROS. Notably, severe cardiac hypertrophy was manifested in aging humanized mice, leading to premature death. The cellular and molecular basis of this phenotype was delineated by showing that low levels of human ECSIT protein led to a marked reduction in assembly and activity of mitochondrial complex I with impaired oxidative phosphorylation and reduced production of ATP. Cardiac tissue from humanized hECSIT mice also showed reduced mitochondrial fusion and more fission but impaired clearance of fragmented mitochondria. A cardiomyocyte-intrinsic role for Ecsit in mitochondrial function and cardioprotection is also demonstrated. We also show that cardiac fibrosis and damage in humans correlated with low expression of human ECSIT. In summary, our findings identify a role for ECSIT in cardioprotection, while generating a valuable experimental model to study mitochondrial dysfunction and cardiac pathophysiology.


Adaptor Proteins, Signal Transducing , Cardiomegaly , Myocardium , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Humans , Macrophages/metabolism , Mice , Mitochondria/metabolism , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/genetics , NF-kappa B/metabolism
3.
J Clin Invest ; 129(11): 4739-4744, 2019 11 01.
Article En | MEDLINE | ID: mdl-31479430

Antisense oligonucleotides (ASOs) targeting pathologic RNAs have shown promising therapeutic corrections for many genetic diseases including myotonic dystrophy (DM1). Thus, ASO strategies for DM1 can abolish the toxic RNA gain-of-function mechanism caused by nucleus-retained mutant DMPK (DM1 protein kinase) transcripts containing CUG expansions (CUGexps). However, systemic use of ASOs for this muscular disease remains challenging due to poor drug distribution to skeletal muscle. To overcome this limitation, we test an arginine-rich Pip6a cell-penetrating peptide and show that Pip6a-conjugated morpholino phosphorodiamidate oligomer (PMO) dramatically enhanced ASO delivery into striated muscles of DM1 mice following systemic administration in comparison with unconjugated PMO and other ASO strategies. Thus, low-dose treatment with Pip6a-PMO-CAG targeting pathologic expansions is sufficient to reverse both splicing defects and myotonia in DM1 mice and normalizes the overall disease transcriptome. Moreover, treated DM1 patient-derived muscle cells showed that Pip6a-PMO-CAG specifically targets mutant CUGexp-DMPK transcripts to abrogate the detrimental sequestration of MBNL1 splicing factor by nuclear RNA foci and consequently MBNL1 functional loss, responsible for splicing defects and muscle dysfunction. Our results demonstrate that Pip6a-PMO-CAG induces long-lasting correction with high efficacy of DM1-associated phenotypes at both molecular and functional levels, and strongly support the use of advanced peptide conjugates for systemic corrective therapy in DM1.


Cell-Penetrating Peptides/pharmacology , Muscle, Skeletal/metabolism , Myotonic Dystrophy , Myotonin-Protein Kinase , Oligodeoxyribonucleotides, Antisense , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Mice , Muscle, Skeletal/pathology , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
4.
Methods Mol Biol ; 1664: 185-202, 2018.
Article En | MEDLINE | ID: mdl-29019134

Comparative tissue proteomics aims to analyze alterations of the proteome in response to a stimulus. Two-dimensional difference gel electrophoresis (2D-DIGE) is a modified and advanced form of 2D gel electrophoresis. DIGE is a powerful biochemical method that compares two or three protein samples on the same analytical gel, and can be used to establish differentially expressed protein levels between healthy normal and diseased pathological tissue sample groups. Minimal DIGE labeling can be used via a 2-dye system with Cy3 and Cy5 or a 3-dye system with Cy2, Cy3, and Cy5 to fluorescently label samples with CyDye flours pre-electrophoresis. DIGE circumvents gel-to-gel variability by multiplexing samples to a single gel and through the use of a pooled internal standard for normalization. This form of quantitative high-resolution proteomics facilitates the comparative analysis and evaluation of tissue protein compositions. Comparing tissue groups under different conditions is crucially important for advancing the biomedical field by characterization of cellular processes, understanding pathophysiological development and tissue biomarker discovery. This chapter discusses 2D-DIGE as a comparative tissue proteomic technique and describes in detail the experimental steps required for comparative proteomic analysis employing both options of 2-dye and 3-dye DIGE minimal labeling.


Fluorescent Dyes , Proteomics , Testis/metabolism , Two-Dimensional Difference Gel Electrophoresis , Animals , Chromatography, Liquid , Humans , Image Processing, Computer-Assisted , Isoelectric Focusing , Male , Mass Spectrometry , Proteome , Proteomics/methods , Staining and Labeling , Two-Dimensional Difference Gel Electrophoresis/methods
5.
Proteomics ; 16(2): 345-66, 2016 Jan.
Article En | MEDLINE | ID: mdl-26256116

The gradual accumulation of collagen and associated proteins of the extracellular matrix is a crucial myopathological parameter of many neuromuscular disorders. Progressive tissue damage and fibrosis play a key pathobiochemical role in the dysregulation of contractile functions and often correlates with poor motor outcome in muscular dystrophies. Following a brief introduction into the role of the extracellular matrix in skeletal muscles, we review here the proteomic profiling of myofibrosis and its intrinsic role in X-linked muscular dystrophy. Although Duchenne muscular dystrophy is primarily a disease of the membrane cytoskeleton, one of its most striking histopathological features is a hyperactive connective tissue and tissue scarring. We outline the identification of novel factors involved in the modulation of the extracellular matrix in muscular dystrophy, such as matricellular proteins. The establishment of novel proteomic markers will be helpful in improving the diagnosis, prognosis, and therapy monitoring in relation to fibrotic substitution of contractile tissue. In the future, the prevention of fibrosis will be crucial for providing optimum conditions to apply novel pharmacological treatments, as well as establish cell-based approaches or gene therapeutic interventions. The elimination of secondary abnormalities in the matrisome promises to reduce tissue scarring and the loss of skeletal muscle elasticity.


Extracellular Matrix Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Animals , Biomarkers/metabolism , Gene Expression Profiling , Humans , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Proteomics
6.
Molecules ; 20(6): 11317-44, 2015 Jun 19.
Article En | MEDLINE | ID: mdl-26102067

The primary deficiency in the membrane cytoskeletal protein dystrophin results in complex changes in dystrophic muscles. In order to compare the degree of secondary alterations in differently affected subtypes of skeletal muscles, we have conducted a global analysis of proteome-wide changes in various dystrophin-deficient muscles. In contrast to the highly degenerative mdx diaphragm muscle, which showed considerable alterations in 35 distinct proteins, the spectrum of mildly to moderately dystrophic skeletal muscles, including interosseus, flexor digitorum brevis, soleus, and extensor digitorum longus muscle, exhibited a smaller number of changed proteins. Compensatory mechanisms and/or cellular variances may be responsible for differing secondary changes in individual mdx muscles. Label-free mass spectrometry established altered expression levels for diaphragm proteins associated with contraction, energy metabolism, the cytoskeleton, the extracellular matrix and the cellular stress response. Comparative immunoblotting verified the differences in the degree of secondary changes in dystrophin-deficient muscles and showed that the up-regulation of molecular chaperones, the compensatory increase in proteins of the intermediate filaments, the fibrosis-related increase in collagen levels and the pathophysiological decrease in calcium binding proteins is more pronounced in mdx diaphragm as compared to the less severely affected mdx leg muscles. Annexin, lamin, and vimentin were identified as universal dystrophic markers.


Annexins/isolation & purification , Dystrophin/isolation & purification , Lamins/isolation & purification , Muscular Dystrophy, Duchenne/diagnosis , Vimentin/isolation & purification , Animals , Annexins/biosynthesis , Dystrophin/biosynthesis , Gene Expression Regulation , Humans , Lamins/biosynthesis , Mass Spectrometry , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Proteome , Vimentin/biosynthesis
7.
Proteomics ; 15(13): 2318-31, 2015 Jul.
Article En | MEDLINE | ID: mdl-25737063

Proteomic profiling plays a decisive role in the identification of novel biomarkers of muscular dystrophy and the elucidation of new pathobiochemical mechanisms that underlie progressive muscle wasting. Building on the findings of recent comparative analyses of tissue samples and body fluids from dystrophic animals and patients afflicted with Duchenne muscular dystrophy, we have used here label-free MS to study the severely dystrophic diaphragm from the not extensively characterized mdx-4cv mouse. This animal model of progressive muscle wasting exhibits less dystrophin-positive revertant fibers than the conventional mdx mouse, making it ideal for the future monitoring of experimental therapies. The pathoproteomic signature of the mdx-4cv diaphragm included a significant increase in the fibrosis marker collagen and related extracellular matrix proteins (asporin, decorin, dermatopontin, prolargin) and cytoskeletal proteins (desmin, filamin, obscurin, plectin, spectrin, tubulin, vimentin, vinculin), as well as decreases in proteins of ion homeostasis (parvalbumin) and the contractile apparatus (myosin-binding protein). Importantly, one of the most substantially increased proteins was identified as periostin, a matricellular component and apparent marker of fibrosis and tissue damage. Immunoblotting confirmed a considerable increase of periostin in the dystrophin-deficient diaphragm from both mdx and mdx-4cv mice, suggesting an involvement of this matricellular protein in dystrophinopathy-related fibrosis.


Cell Adhesion Molecules/metabolism , Diaphragm/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Computational Biology , In Vitro Techniques , Mice , Mice, Inbred mdx , Tandem Mass Spectrometry
8.
Proteomics ; 15(4): 632-48, 2015 Feb.
Article En | MEDLINE | ID: mdl-24909132

The highly complex and species-selective mechanism of fertilization is a central theme of developmental biology. Gametogenesis, sperm activation, and egg-sperm recognition are fundamental biological processes, warranting detailed studies into the molecular composition of gametes. Biological MS has been instrumental for the comprehensive itemizing of gamete proteomes. The protein constellation of sperm cells and its subcellular structures has been established for a variety of animal species. Spermatogenesis and the crucial activation of sperm cells as a prerequisite of successful fertilization and physiological adaptations to external stressors was investigated using proteomics, as well as the underlying mechanisms of male infertility with respect to proteome-wide alterations. This review outlines recent achievements of sperm proteomics and exemplifies the usefulness of gel-based surveys by outlining the comparative analysis of abnormal spermatozoa in globozoospermia. Besides label-free MS techniques and cell-based labeling methodology, high-resolution fluorescence 2DE has been shown to be highly suitable as a proteomic biomarker discovery tool in sperm protein research. The appropriateness of novel protein markers for improving our understanding of normal spermatogenesis and sperm activation versus the molecular pathogenesis of male infertility will be discussed. New biomarker candidates might be useful to improve diagnostic, prognostic, and therapeutic aspects of infertility.


Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Spermatozoa/chemistry , Spermatozoa/metabolism , Animals , Humans , Male , Proteome/analysis , Spermatogenesis
9.
Biosci Rep ; 34(4)2014 Jul 01.
Article En | MEDLINE | ID: mdl-24895011

While the long-term physiological adaptation of the neuromuscular system to changed functional demands is usually reflected by unilateral skeletal muscle transitions, the progressive degeneration of distinct motor neuron populations is often associated with more complex changes in the abundance and/or isoform expression pattern of contractile proteins and metabolic enzymes. In order to evaluate these intricate effects of primary motor neuronopathy on the skeletal muscle proteome, label-free MS was employed to study global alterations in the WR (wobbler) mouse model of progressive neurodegeneration. In motor neuron disease, fibre-type specification and the metabolic weighting of bioenergetic pathways appear to be strongly influenced by both a differing degree of a subtype-specific vulnerability of neuromuscular synapses and compensatory mechanisms of fibre-type shifting. Proteomic profiling confirmed this pathobiochemical complexity of disease-induced changes and showed distinct alterations in 72 protein species, including a variety of fibre-type-specific isoforms of contractile proteins, metabolic enzymes, metabolite transporters and ion-regulatory proteins, as well as changes in molecular chaperones and various structural proteins. Increases in slow myosin light chains and the troponin complex and a decrease in fast MBP (myosin-binding protein) probably reflect the initial preferential loss of the fast type of neuromuscular synapses in motor neuron disease.


Contractile Proteins/metabolism , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Animals , Mass Spectrometry/methods , Mice , Molecular Chaperones/metabolism , Protein Isoforms/metabolism , Proteome/metabolism , Proteomics/methods
10.
Front Aging Neurosci ; 6: 109, 2014.
Article En | MEDLINE | ID: mdl-24917816

Primary abnormalities in the dystrophin gene cause X-linked muscular dystrophy, a highly progressive muscle wasting disorder of childhood. A spontaneous animal model of Duchenne muscular dystrophy is the mdx mouse, which presents a highly interesting phenotype that exhibits considerable variations in the degree of fiber degeneration in different subtypes of muscles. The idea that aging exacerbates the dystrophic mdx phenotype, as previously indicated by a large number of biochemical and cell biological studies, was clearly confirmed by comparative muscle proteomics. Here we outline recent findings of age-dependent changes in the dystrophin-deficient muscle proteome and contrast these results with the previously established proteomic profile of sarcopenic muscle. Besides comparable perturbations of various biochemical functions, especially striking are similarities in the cellular stress response associated with a drastic up-regulation of small αB-crystallin-like heat shock proteins. Hence, the comparison of large-scale proteomic data sets of natural muscle aging with dystrophic sarcopenia promises to shed light on the differential effect of sarcopenia of old age vs. senescent abnormalities on a mutant dystrophic background.

11.
Biomed Res Int ; 2014: 246195, 2014.
Article En | MEDLINE | ID: mdl-24772416

Cardiorespiratory complications are frequent symptoms of Duchenne muscular dystrophy, a neuromuscular disorder caused by primary abnormalities in the dystrophin gene. Loss of cardiac dystrophin initially leads to changes in dystrophin-associated glycoproteins and subsequently triggers secondarily sarcolemmal disintegration, fibre necrosis, fibrosis, fatty tissue replacement, and interstitial inflammation. This results in progressive cardiac disease, which is the cause of death in a considerable number of patients afflicted with X-linked muscular dystrophy. In order to better define the molecular pathogenesis of this type of cardiomyopathy, several studies have applied mass spectrometry-based proteomics to determine proteome-wide alterations in dystrophinopathy-associated cardiomyopathy. Proteomic studies included both gel-based and label-free mass spectrometric surveys of dystrophin-deficient heart muscle from the established mdx animal model of dystrophinopathy. Comparative cardiac proteomics revealed novel changes in proteins associated with mitochondrial energy metabolism, glycolysis, signaling, iron binding, antibody response, fibre contraction, basal lamina stabilisation, and cytoskeletal organisation. This review summarizes the importance of studying cardiomyopathy within the field of muscular dystrophy research, outlines key features of the mdx heart and its suitability as a model system for studying cardiac pathogenesis, and discusses the impact of recent proteomic findings for exploring molecular and cellular aspects of cardiac abnormalities in inherited muscular dystrophies.


Cardiomyopathies/genetics , Dystrophin/genetics , Muscular Dystrophy, Duchenne/complications , Protein Biosynthesis , Animals , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Dystrophin/deficiency , Gene Expression Profiling , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Phenotype , Proteomics
12.
J Med Microbiol ; 63(Pt 2): 258-262, 2014 Feb.
Article En | MEDLINE | ID: mdl-24248990

Rosacea is a chronic inflammatory condition that affects the skin of the face and the eyes. The aetiology of rosacea is not clearly established but increasing evidence suggests a potential role for bacteria in the induction of the condition. A role for Bacillus oleronius, originally isolated from within a Demodex folliculorum mite, in the aetiology of the condition has been suggested. The aim of the study was to determine whether a correlation existed between the level of sebum and the density of D. folliculorum in the skin of erythematotelangiectatic rosacea patients, and the reactivity of these patients' sera to proteins of B. oleronius. Serum reactivity to the 62 and 83 kDa B. oleronius proteins was found in 82.6 % (62/75) of the rosacea patients and in 26.9 % (14/52) of controls (P = 0.0016). In the group of rosacea patients whose sera reacted to B. oleronius proteins, the level of sebum was statistically lower than in controls (P = 0.01). The density of D. folliculorum on the face of Bacillus positive rosacea patients was statistically higher than controls (P = 0.0001). Rosacea patients demonstrated increased Demodex populations on their faces and reduced sebum levels. Their sera also showed reactivity to B. oleronius proteins, suggesting a potential role for this bacterium in the aetiology of rosacea.


Acari/growth & development , Acari/microbiology , Antibodies, Bacterial/blood , Bacillus/immunology , Rosacea/pathology , Sebum/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Middle Aged , Rosacea/microbiology , Rosacea/parasitology , Young Adult
13.
Proteomics ; 14(7-8): 839-52, 2014 Apr.
Article En | MEDLINE | ID: mdl-24115398

In human globozoospermia, round-headed spermatozoa lack an acrosome and therefore cannot properly interact with oocytes. In the wobbler (WR) mouse, an L967Q missense mutation in the vesicular protein-sorting factor VPS54 causes motor neuron degeneration and globozoospermia. Although electron microscopy of WR testis shows all major components of spermatogenesis, they appear in a deranged morphology that prevents the formation of the acrosome. In order to determine proteome-wide changes, affected testes were analysed by 2D-DIGE and MS. The concentration of 8 proteins was increased and that of 35 proteins decreased as compared to wild type. Mass spectrometric analysis identified proteins with an altered concentration to be associated with metabolite transport, fatty acid metabolism, cellular interactions, microtubule assembly and stress response (chaperones Hsp70-2 and Hsp90α). Minor changes were observed for proteins involved in cell redox homeostasis, cytoskeleton formation, PTMs, detoxification and metabolism. The most dramatically decreased protein in WR testis was identified as fatty acid binding protein FABP3, as confirmed by immunoblot analysis. We conclude that a missense mutation in VPS54, an essential component of the Golgi-associated retrograde protein complex, not only prevents the formation of an acrosome but also initiates a cascade of metabolic abnormalities and a stress reaction.


Infertility, Male/genetics , Membrane Proteins/genetics , Proteomics , Vesicular Transport Proteins/genetics , Acrosome/metabolism , Acrosome/pathology , Animals , Disease Models, Animal , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Humans , Infertility, Male/pathology , Male , Mice , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Mutation, Missense , Spermatogenesis/genetics
14.
J Neuromuscul Dis ; 1(1): 15-40, 2014.
Article En | MEDLINE | ID: mdl-27858666

The optimization of large-scale screening procedures of pathological specimens by genomic, proteomic and metabolic methods has drastically increased the bioanalytical capability for swiftly identifying novel biomarkers of inherited disorders, such as neuromuscular diseases. X-linked muscular dystrophy represents the most frequently inherited muscle disease and is characterized by primary abnormalities in the membrane cytoskeletal protein dystrophin. Mass spectrometry-based proteomics has been widely employed for the systematic analysis of dystrophin-deficient muscle tissues, using patient samples and animal models of dystrophinopathy. Both, gel-based methods and label-free mass spectrometric techniques have been applied in comparative analyses and established a large number of altered proteins that are associated with muscle contraction, energy metabolism, ion homeostasis, cellular signaling, the cytoskeleton, the extracellular matrix and the cellular stress response. Although these new indicators of muscular dystrophy have increased our general understanding of the molecular pathogenesis of dystrophinopathy, their application as new diagnostic or prognostic biomarkers would require the undesirable usage of invasive methodology. Hence, to reduce the need for diagnostic muscle biopsy procedures, more recent efforts have focused on the proteomic screening of suitable body fluids, such as plasma, serum or urine, for the identification of changed concentration levels of muscle-derived peptides, protein fragments or intact proteins. The occurrence of muscular dystrophy-related protein species in biofluids will be extremely helpful for the future development of cost-effective and non-invasive diagnostic procedures. Novel biomarker signatures of dystrophinopathies will be indispensible for the swift evaluation of innovative therapeutic approaches, such as exon skipping, codon-read-through or stem cell therapy.

15.
Curr Protein Pept Sci ; 14(8): 680-97, 2013 Dec.
Article En | MEDLINE | ID: mdl-24106963

The largest human gene is represented by the X-chromosomal dystrophin gene of 2.4 million bases, which encodes for the membrane cytoskeletal protein dystrophin. The dystrophin isoform Dp427 has a subsarcolemmal location and forms a supramolecular membrane assembly with a variety of glycoproteins. In healthy muscle fibres, dystrophin acts as an actin-binding protein that links the cytoskeleton via the α/ß-dystroglycan complex to the extracellular matrix protein laminin. This trans-sarcolemmal complex is believed to stabilize the muscle surface and thus prevents membrane rupturing during excitation-contraction-relaxation cycles. In the highly progressive muscle wasting disease Duchenne muscular dystrophy, the primary deficiency in dystrophin causes a drastic reduction in dystrophin-associated glycoproteins, which renders muscle fibres more susceptible to necrosis. Following the biochemical and cell biological characterization of the dystrophin-glycoprotein complex, several mass spectrometry-based proteomic studies have investigated global changes in dystrophin-deficient muscle tissues. This review briefly outlines the basic domain structure of Dp427 and the composition of the dystrophin-associated glycoprotein complex from skeletal muscle. A detailed discussion of recent proteomic analyses of the purified dystrophin-glycoprotein complex is included, as well as a summary of mass spectrometric surveys of dystrophic specimens. The study of these new areas of muscle proteomics tends to improve our understanding of the normal function of dystrophin in contractile fibres and better define the molecular mechanism of X-linked muscular dystrophy.


Dystrophin/metabolism , Glycoproteins/metabolism , Muscular Dystrophies/metabolism , Proteomics/methods , Animals , Dystrophin/analysis , Glycoproteins/analysis , Humans , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Protein Isoforms/analysis , Protein Isoforms/metabolism , Protein Structure, Tertiary
16.
Expert Rev Proteomics ; 10(3): 239-57, 2013 Jun.
Article En | MEDLINE | ID: mdl-23777215

In skeletal muscle fibers, the excitation-contraction-relaxation cycle is a highly evolved process that is mediated by the contractile proteins - myosin and actin - and the regulatory elements - troponin and tropomyosin. Contractile fibers exhibit enormous complexity and heterogeneity on the molecular level, which is reflected by the diversity of protein isoforms that constitute the actomyosin apparatus. The main components of the contractile apparatus exist in high abundance and are relatively soluble, making them ideal candidates for a systematic analysis by liquid chromatography or gel electrophoresis-based proteomics. This review discusses the proteomic profiling of contractile components in adapting, degenerating and aging skeletal muscle tissues. The proteomic identification of altered contractile proteins may be useful for the establishment of biomarker signatures that can be applied in the examination of the physiological adaptability, cellular plasticity and pathological susceptibility of the neuromuscular system.


Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Proteomics/methods , Animals , Humans , Mass Spectrometry , Meat , Muscle Development , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology
17.
Proteomics ; 13(15): 2312-23, 2013 Aug.
Article En | MEDLINE | ID: mdl-23713012

The majority of patients afflicted with Duchenne muscular dystrophy develop cardiomyopathic complications, warranting large-scale proteomic studies of global cardiac changes for the identification of new protein markers of dystrophinopathy. The aged heart from the X-linked dystrophic mdx mouse has been shown to exhibit distinct pathological aspects of cardiomyopathy. In order to establish age-related alterations in the proteome of dystrophin-deficient hearts, cardiomyopathic tissue from young versus aged mdx mice was examined by label-free LC-MS/MS. Significant age-dependent alterations were established for 67 proteins, of which 28 proteins were shown to exhibit a lower abundance and 39 proteins were found to be increased in their expression levels. Drastic changes were demonstrated for 17 proteins, including increases in Ig chains and transferrin, and drastic decreases in laminin, nidogen and annexin. An immunblotting survey of young and old wild-type versus mdx hearts confirmed these proteomic findings and illustrated the effects of natural aging versus dystrophin deficiency. These proteome-wide alterations suggest a disintegration of the basal lamina structure and cytoskeletal network in dystrophin-deficient cardiac fibres, increased levels of antibodies in a potential autoimmune reaction of the degenerating heart, compensatory binding of excess iron and a general perturbation of metabolic pathways in dystrophy-associated cardiomyopathy.


Annexins/metabolism , Laminin/metabolism , Membrane Glycoproteins/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myocardium/metabolism , Age Factors , Animals , Annexins/analysis , Cardiomyopathies/metabolism , Humans , Laminin/analysis , Male , Membrane Glycoproteins/analysis , Mice , Mice, Inbred mdx , Myocardium/chemistry , Proteome/analysis , Proteome/metabolism , Random Allocation , Rats, Wistar
...