Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 14(8): 3348-61, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26074025

ABSTRACT

Muscle stem cells, or satellite cells, play an important role in the maintenance and repair of muscle tissue and have the capacity to proliferate and differentiate in response to physiological or environmental changes. Although they have been extensively studied, the key regulatory steps and the complex temporal protein dynamics accompanying the differentiation of primary human muscle cells remain poorly understood. Here, we demonstrate the advantages of applying a MS-based quantitative approach, stable isotope labeling by amino acids in cell culture (SILAC), for studying human myogenesis in vitro and characterize the fine-tuned changes in protein expression underlying the dramatic phenotypic conversion of primary mononucleated human muscle cells during in vitro differentiation to form multinucleated myotubes. Using an exclusively optimized triple encoding SILAC procedure, we generated dynamic expression profiles during the course of myogenic differentiation and quantified 2240 proteins, 243 of which were regulated. These changes in protein expression occurred in sequential waves and underlined vast reprogramming in key processes governing cell fate decisions, i.e., cell cycle withdrawal, RNA metabolism, cell adhesion, proteolysis, and cytoskeletal organization. In silico transcription factor target analysis demonstrated that the observed dynamic changes in the proteome could be attributed to a cascade of transcriptional events involving key myogenic regulatory factors as well as additional regulators not yet known to act on muscle differentiation. In addition, we created of a dynamic map of the developing myofibril, providing valuable insights into the formation and maturation of the contractile apparatus in vitro. Finally, our SILAC-based quantitative approach offered the possibility to follow the expression profiles of several muscle disease-associated proteins simultaneously and therefore could be a valuable resource for future studies investigating pathogenesis of degenerative muscle disorders as well as assessing new therapeutic strategies.


Subject(s)
Cell Differentiation , Muscle Fibers, Skeletal/metabolism , Proteome/metabolism , Proteomics/methods , Satellite Cells, Skeletal Muscle/metabolism , Amino Acids/metabolism , Blotting, Western , Cells, Cultured , Chromatography, Liquid , Cluster Analysis , Humans , Immunohistochemistry , Infant, Newborn , Isotope Labeling/methods , Kinetics , Muscle Fibers, Skeletal/cytology , Proteome/classification , Satellite Cells, Skeletal Muscle/cytology , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Time Factors
2.
Hum Mol Genet ; 22(15): 3152-64, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23575224

ABSTRACT

Dilated cardiomyopathy (DCM) associates left ventricular (LV) dilatation and systolic dysfunction and is a major cause of heart failure and cardiac transplantation. LMNA gene encodes lamins A/C, proteins of the nuclear envelope. LMNA mutations cause DCM with conduction and/or rhythm defects. The pathomechanisms linking mutations to DCM remain to be elucidated. We investigated the phenotype and associated pathomechanisms of heterozygous Lmna(ΔK32/+) (Het) knock-in mice, which carry a human mutation. Het mice developed a cardiac-specific phenotype. Two phases, with two different pathomechanisms, could be observed that lead to the development of cardiac dysfunction, DCM and death between 35 and 70 weeks of age. In young Het hearts, there was a clear reduction in lamin A/C level, mainly due to the degradation of toxic ΔK32-lamin. As a side effect, lamin A/C haploinsufficiency probably triggers the cardiac remodelling. In older hearts, when DCM has developed, the lamin A/C level was normalized and associated with increased toxic ΔK32-lamin expression. Crossing our mice with the Ub(G76V)-GFP ubiquitin-proteasome system (UPS) reporter mice revealed a heart-specific UPS impairment in Het. While UPS impairment itself has a clear deleterious effect on engineered heart tissue's force of contraction, it also leads to the nuclear aggregation of viral-mediated expression of ΔK32-lamin. In conclusion, Het mice are the first knock-in Lmna model with cardiac-specific phenotype at the heterozygous state. Altogether, our data provide evidence that Het cardiomyocytes have to deal with major dilemma: mutant lamin A/C degradation or normalization of lamin level to fight the deleterious effect of lamin haploinsufficiency, both leading to DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Haploinsufficiency , Heterozygote , Lamin Type A/genetics , Lamin Type A/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , Disease Models, Animal , Disease Progression , Female , Lamin Type A/chemistry , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Mutation , Myocardial Contraction/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism
3.
J Proteomics ; 88: 4-13, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23353020

ABSTRACT

With the rapid advance of MS-based proteomics one might think that 2D gel-based proteomics is dead. This is far from the truth. Current research has shown that there are still a number of places in the field of protein and molecular biology where 2D gels still play a leading role. The aim of this review is to highlight some of these applications. Examples from our own research as well as from other published works are used to illustrate the 2D gel driven research in the areas of: 1) de novo sequencing and protein identification from organisms with no or incomplete genome sequences available; 2) alternative detection methods for modification specific proteomics; 3) identification of protein isoforms and modified proteins. With an example of the glycoprotein TIMP-1 protein we illustrate the unique properties of 2D gels for the separation and characterisation of multiply modified proteins. We also show that careful analysis of experimental and theoretical protein mass and pI can lead to the identification of unanticipated protein variants modified by for example proteolytic cleavage. Together this shows that there is an important niche for 2D gel-based proteomics, which compliments traditional LC-MS techniques for specific protein research purposes.


Subject(s)
Proteomics/methods , Tissue Inhibitor of Metalloproteinase-1/analysis , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Electrophoresis, Gel, Two-Dimensional/methods , Humans
4.
Cell Mol Life Sci ; 70(12): 2159-74, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23344255

ABSTRACT

Duchenne muscular dystrophy results from loss of the protein dystrophin, which links the intracellular cytoskeletal network with the extracellular matrix, but deficiency in this function does not fully explain the onset or progression of the disease. While some intracellular events involved in the degeneration of dystrophin-deficient muscle fibers have been well characterized, changes in their secretory profile are undescribed. To analyze the secretome profile of mdx myotubes independently of myonecrosis, we labeled the proteins of mdx and wild-type myotubes with stable isotope-labeled amino acids (SILAC), finding marked enrichment of vesicular markers in the mdx secretome. These included the lysosomal-associated membrane protein, LAMP1, that co-localized in vesicles with an over-secreted cytoskeletal protein, myosin light chain 1. These LAMP1/MLC1-3-positive vesicles accumulated in the cytosol of mdx myotubes and were secreted into the culture medium in a range of abnormal densities. Restitution of dystrophin expression, by exon skipping, to some 30 % of the control value, partially normalized the secretome profile and the excess LAMP1 accumulation. Together, our results suggest that a lack of dystrophin leads to a general dysregulation of vesicle trafficking. We hypothesize that disturbance of the export of proteins through vesicles occurs before, and then concurrently with, the myonecrotic cascade and contributes chronically to the pathophysiology of DMD, thereby presenting us with a range of new potential therapeutic targets.


Subject(s)
Dystrophin/deficiency , Lysosomal Membrane Proteins/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Secretory Vesicles/metabolism , Actins/analysis , Amino Acids/metabolism , Animals , Blotting, Western , Cell Line , Chromatography, Liquid , Computational Biology , Immunoblotting , Isotope Labeling , Male , Membrane Proteins/metabolism , Mice , Microscopy, Electron, Transmission , Muscle Fibers, Skeletal/metabolism , Secretory Vesicles/ultrastructure , Statistics, Nonparametric , Tandem Mass Spectrometry
5.
PLoS One ; 7(12): e51865, 2012.
Article in English | MEDLINE | ID: mdl-23272181

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is a progressive muscle disorder linked to a contraction of the D4Z4 repeat array in the 4q35 subtelomeric region. This deletion induces epigenetic modifications that affect the expression of several genes located in the vicinity. In each D4Z4 element, we identified the double homeobox 4 (DUX4) gene. DUX4 expresses a transcription factor that plays a major role in the development of FSHD through the initiation of a large gene dysregulation cascade that causes myogenic differentiation defects, atrophy and reduced response to oxidative stress. Because miRNAs variably affect mRNA expression, proteomic approaches are required to define the dysregulated pathways in FSHD. In this study, we optimized a differential isotope protein labeling (ICPL) method combined with shotgun proteomic analysis using a gel-free system (2DLC-MS/MS) to study FSHD myotubes. Primary CD56(+) FSHD myoblasts were found to fuse into myotubes presenting various proportions of an atrophic or a disorganized phenotype. To better understand the FSHD myogenic defect, our improved proteomic procedure was used to compare predominantly atrophic or disorganized myotubes to the same matching healthy control. FSHD atrophic myotubes presented decreased structural and contractile muscle components. This phenotype suggests the occurrence of atrophy-associated proteolysis that likely results from the DUX4-mediated gene dysregulation cascade. The skeletal muscle myosin isoforms were decreased while non-muscle myosin complexes were more abundant. In FSHD disorganized myotubes, myosin isoforms were not reduced, and increased proteins were mostly involved in microtubule network organization and myofibrillar remodeling. A common feature of both FSHD myotube phenotypes was the disturbance of several caveolar proteins, such as PTRF and MURC. Taken together, our data suggest changes in trafficking and in the membrane microdomains of FSHD myotubes. Finally, the adjustment of a nuclear fractionation compatible with mass spectrometry allowed us to highlight alterations of proteins involved in mRNA processing and stability.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Phenotype , Proteome , Caveolae/metabolism , Cells, Cultured , Humans , Myoblasts/metabolism , Myosins/metabolism , Protein Isoforms , Proteomics
6.
J Proteomics ; 77: 344-56, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23000592

ABSTRACT

Efficient muscle regeneration requires cross talk between multiple cell types via secreted signaling molecules. However, as yet there has been no comprehensive analysis of this secreted signaling network in order to understand how it regulates myogenesis in humans. Using integrated proteomic and genomic strategies, we show that human muscle cells release not only soluble secreted proteins through conventional secretory mechanisms but also complex protein and nucleic acid cargos via membrane microvesicle shedding. The soluble secretome of muscle cells contains 253 conventionally secreted signaling proteins, including 43 previously implicated in myogenesis, while others are known to modulate various cell types thus implying a much broader role for myoblasts in muscle remodeling. We also isolated and characterized two types of secreted membrane-derived vesicles: nanovesicles harboring typical exosomal features and larger, morphologically distinct, microvesicles. While they share some common features, their distinct protein and RNA cargos suggest independent functions in myogenesis. We further demonstrate that both types of microvesicles can dock and fuse with adjacent muscle cells but also deliver functional protein cargo. Thus, the intercellular signaling networks invoked during muscle differentiation and regeneration may employ conventional soluble signaling molecules acting in concert with muscle derived microvesicles delivering their cargos directly into target cells.


Subject(s)
Cell Differentiation/physiology , Cell-Derived Microparticles/metabolism , Muscle Proteins/metabolism , Proteome/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Secretory Pathway/physiology , Cells, Cultured , Female , Humans , Infant, Newborn , Proteomics/methods , Satellite Cells, Skeletal Muscle/cytology
7.
Mol Cell Proteomics ; 11(8): 230-43, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22345510

ABSTRACT

Microparticles and exosomes are two of the most well characterized membrane-derived microvesicles released either directly from the plasma membrane or released through the fusion of intracellular multivesicular bodies with the plasma membrane, respectively. They are thought to be involved in many significant biological processes such as cell to cell communication, rescue from apoptosis, and immunological responses. Here we report for the first time a quantitative study of proteins from ß-cell-derived microvesicles generated after cytokine induced apoptosis using stable isotope labeled amino acids in cell culture combined with mass spectrometry. We identified and quantified a large number of ß-cell-specific proteins and proteins previously described in microvesicles from other cell types in addition to new proteins located to these vesicles. In addition, we quantified specific sites of protein phosphorylation and N-linked sialylation in proteins associated with microvesicles from ß-cells. Using pathway analysis software, we were able to map the most distinctive changes between microvesicles generated during growth and after cytokine stimulation to several cell death and cell signaling molecules including tumor necrosis factor receptor superfamily member 1A, tumor necrosis factor, α-induced protein 3, tumor necrosis factor-interacting kinase receptor-interacting serine-threonine kinase 1, and intercellular adhesion molecule 1.


Subject(s)
Cell-Derived Microparticles/metabolism , Cytokines/pharmacology , Exosomes/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Proteomics/methods , Amino Acids/metabolism , Animals , Apoptosis/drug effects , Carbon Isotopes , Cell Line, Tumor , Cell Membrane/metabolism , Cell-Derived Microparticles/ultrastructure , Chromatography, Liquid , Exosomes/ultrastructure , Glycoproteins/analysis , Insulin-Secreting Cells/pathology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Isotope Labeling/methods , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nitrogen Isotopes , Phosphoproteins/analysis , Rats , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/pharmacology
8.
Proteomics ; 6(16): 4646-61, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16858738

ABSTRACT

A proteomic strategy based upon the integrated use of SELDI-TOF/MS, 2-DE and MALDI-TOF/MS has been used to identify a panel of fast muscle protein markers: MLC1F, MLC3F, fast troponin C (STNC) and slow muscle markers: MLC1SB and MLC2v. MLC3F, MLC1F and STNC were virtually absent in the physiologically pure slow soleus muscle of kyphoscoliotic mutant mice compared to control BDmice, whereas MLC2v increased threefold. A SELDI-TOF/MS peak at 18,012 Da in spectra from strong anionic exchange protein array fractions of fast vastus muscle was confirmed as STNC by its specific depletion from crude extracts of vastus muscle using an anti-TNC mAb. SELDI-TOF/MS also identified MLC2F phosphorylation in crude muscle extracts after treatment with alkaline phosphatase. High probability protein identifications were achieved by SELDI-TOF/MS PMF based upon the resolution of large peptides formed by partial cleavage and high peptide coverage. When the pI from 2-D gels and molecular weight estimations from SELDI-TOF/MS were entered into the TagIdent algorithm, high probability protein identity predictions were obtained that were confirmed later by PMF. We confirm that SELDI-TOF/MS can be integrated with other proteomics techniques for the efficient analysis of protein expression changes and PTMs associated with physiological changes in skeletal muscle.


Subject(s)
Kyphosis/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle Proteins/metabolism , Proteome/metabolism , Scoliosis/metabolism , Animals , Biomarkers/metabolism , Electrophoresis, Gel, Two-Dimensional , Male , Mice , Mice, Mutant Strains , Protein Array Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Proteomics ; 6(10): 3096-108, 2006 May.
Article in English | MEDLINE | ID: mdl-16622832

ABSTRACT

Complex molecular changes associated with early stage human heart disease are poorly understood and prevent the development of effective treatments of human cardiac disease. Relatively minor structural changes in early disease may accompany some conditions such as arrhythmias. Our objective was to determine if significant proteomic changes occur in heart tissues in the absence of structural pathology. We used a proteomic "pipeline" based on Ciphergen SELDI-TOF/MS, gel electrophoresis and MALDI-TOF/MS. The kyphoscoliosis (ky) mouse carries a mutation in a putative transglutaminase causing a primary skeletal muscle disease. The ky protein is expressed usually in skeletal and cardiac muscle but its absence from the ky heart causes no structural pathology making it a good model of "occult" heart disease. We discovered 20 statistically validated biomarkers discriminating ky from normal hearts, one cardiac troponin-I was reduced by 40% in ky hearts. A 17% deficit was confirmed subsequently by Western blot. Thus, the proteome of ky hearts was abnormal, giving support to our contention that this SELDI-based analytical approach is capable of making a significant contribution to the analysis of complex proteomic changes in early stage human heart disease.


Subject(s)
Kyphosis/metabolism , Myocardium/metabolism , Proteome/biosynthesis , Scoliosis/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Heart Diseases/metabolism , Humans , Kyphosis/genetics , Mice , Mutation , Protein Array Analysis , Scoliosis/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transglutaminases/genetics , Troponin I/biosynthesis
10.
Proteomics ; 4(9): 2739-53, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15352248

ABSTRACT

Proteomic analysis of skeletal muscle presents particular challenges when trying to identify valid biomarkers of phenotypic change in small biopsies from genetically diverse human subjects. Currently, two-dimensional (2-D) gel electrophoresis and mass spectrometry are the chosen analytical strategies but 2-D gels are not appropriate for analyzing proteins less than 11 kDa, they can suffer from problems of reproducibility and in routine use are not a viable high-throughput technique. We have evaluated an integrated proteomic strategy employing Ciphergen ProteinChip arrays, one-dimensional polyacrylamide gel electrophoresis and mass spectrometry. Protein fingerprints characteristic of fast and slow contracting muscles from normal and kyphoscoliosis (ky) mutant mice were obtained from Ciphergen protein arrays. Eight statistically validated protein biomarkers have so far been identified capable of discriminating fast from slow muscle. Five of these showed further differential expression in ky versus normal BDL soleus muscles. Several biomarkers have been formally identified, and were myosin light chain isoforms shown previously to be expressed differentially by fast versus slow skeletal muscles. This integrated experimental approach using a model mouse muscle system shows the potential of Ciphergen protein array technology for proteomic analysis of small proteins in small muscle samples and its applicability for phenotypic characterization of skeletal muscle in general.


Subject(s)
Muscle Proteins/chemistry , Muscle, Skeletal/chemistry , Protein Array Analysis , Proteome/analysis , Proteomics/methods , Animals , Biomarkers , Humans , Hydrogen-Ion Concentration , Mice , Mice, Mutant Strains , Muscle Proteins/isolation & purification , Muscle Proteins/metabolism , Peptide Mapping , Protein Binding , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...