Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Cachexia Sarcopenia Muscle ; 12(4): 973-982, 2021 08.
Article in English | MEDLINE | ID: mdl-34060717

ABSTRACT

BACKGROUND: The assessment of muscle mass is a key determinant of the diagnosis of sarcopenia. We introduce for the first time an ultrasound imaging method for diagnosing sarcopenia based on changes in muscle geometric proportions. METHODS: Vastus lateralis muscle fascicle length (Lf) and thickness (Tm) were measured at 35% distal femur length by ultrasonography in a population of 279 individuals classified as moderately active elderly (MAE), sedentary elderly (SE) (n = 109), mobility impaired elderly (MIE) (n = 43), and in adult young controls (YC) (n = 60). The ratio of Lf/Tm was calculated to obtain an ultrasound index of the loss of muscle mass associated with sarcopenia (USI). In a subsample of elderly male individuals (n = 76) in which corresponding DXA measurements were available (MAE, n = 52 and SE, n = 24), DXA-derived skeletal muscle index (SMI, appendicular limb mass/height2 ) was compared with corresponding USI values. RESULTS: For both young and older participants, USI values were found to be independent of sex, height and body mass. USI values were 3.70 ± 0.52 for YC, 4.50 ± 0.72 for the MAE, 5.05 ± 1.11 for the SE and 6.31 ± 1.38 for the MIE, all significantly different between each other (P < 0.0001). Based on the USI Z-scores, with reference to the YC population, the 219 elderly participants were stratified according to their muscle sarcopenic status. Individuals with USI values within a range of 3.70 < USI ≥ 4.23 were classified as non-sarcopenic (prevalence 23.7%), those with USI values within 4.23 < USI ≥ 4.76 were classified as pre-sarcopenic (prevalence 23.7%), those with USI values within 4.76 < USI ≥ 5.29 were classified as moderately sarcopenic (prevalence 15.1%), those with USI values within range 5.29 < USI ≥ 5.82 were classified as sarcopenic (prevalence 27.9%), and those with USI values >5.82 were classified as severely sarcopenic (prevalence 9.6%). The DXA-derived SMI was found to be significantly correlated with USI (r = 0.61, P < 0.0001). Notably, the USI cut-off value for moderate sarcopenia (4.76 a.u.) was found to coincide with the DXA cut-off value of sarcopenia (7.26 kg/m2 ). CONCLUSIONS: We propose a novel, practical, and inexpensive imaging marker of the loss of muscle mass associated with sarcopenia, called the ultrasound sarcopenic index (USI), based on changes in muscle geometric proportions. These changes provide a useful 'signature of sarcopenia' and allow the stratification of individuals according to the presence and severity of muscle sarcopenia. We are convinced that the USI will be a useful clinical tool for confirming the diagnosis of sarcopenia, of which the assessment of muscle mass is a key-component.


Subject(s)
Sarcopenia , Adult , Aged , Humans , Male , Muscle, Skeletal/diagnostic imaging , Prevalence , Quadriceps Muscle , Sarcopenia/diagnostic imaging , Sarcopenia/epidemiology , Ultrasonography
2.
J Cachexia Sarcopenia Muscle ; 9(6): 1063-1078, 2018 12.
Article in English | MEDLINE | ID: mdl-30216693

ABSTRACT

BACKGROUND: This study was performed to test the therapeutic potential of obestatin, an autocrine anabolic factor regulating skeletal muscle repair, to ameliorate the Duchenne muscular dystrophy (DMD) phenotype. METHODS AND RESULTS: Using a multidisciplinary approach, we characterized the ageing-related preproghrelin/GPR39 expression patterns in tibialis anterior (TA) muscles of 4-, 8-, and 18-week-old mdx mice (n = 3/group) and established the effects of obestatin administration at this level in 8-week-old mdx mice (n = 5/group). The findings were extended to in vitro effects on human immortalized DMD myotubes. An analysis of TAs revealed an age-related loss of preproghrelin expression, as precursor of obestatin, in mdx mice. Administration of obestatin resulted in a significant increase in tetanic specific force (33.0% ± 1.5%, P < 0.05), compared with control mdx mice. Obestatin-treated TAs were characterized by reduction of fibres with centrally located nuclei (10.0% ± 1.2%, P < 0.05) together with an increase in the number of type I fibres (25.2% ± 1.7%, P < 0.05) associated to histone deacetylases/myocyte enhancer factor-2 and peroxisome proliferator-activated receptor-gamma coactivator 1α axis, and down-regulation of ubiquitin E3-ligases by inactivation of FoxO1/4, indexes of muscle atrophy. Obestatin reduced the level of contractile damage and tissue fibrosis. These observations correlated with decline in serum creatine kinase (58.8 ± 15.2, P < 0.05). Obestatin led to stabilization of the sarcolemma by up-regulation of utrophin, α-syntrophin, ß-dystroglycan, and α7ß1-integrin proteins. These pathways were also operative in human DMD myotubes. CONCLUSIONS: These results highlight the potential of obestatin as a peptide therapeutic for preserving muscle integrity in DMD, thus allowing a better efficiency of gene or cell therapy in a combined therapeutic approach.


Subject(s)
Ghrelin/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/physiopathology , Phenotype , Animals , Biomarkers , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/diagnosis , Oxidation-Reduction/drug effects , Protein Biosynthesis/drug effects , Proteolysis , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sarcolemma/drug effects , Sarcolemma/metabolism
3.
Skelet Muscle ; 7(1): 20, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29017538

ABSTRACT

BACKGROUND: The hepatocyte growth factor (HGF) is required for the activation of muscle progenitor cells called satellite cells (SC), plays a role in the migration of proliferating SC (myoblasts), and is present as a soluble factor during muscle regeneration, along with extracellular matrix (ECM) molecules. In this study, we aimed at determining whether HGF is able to interact with ECM proteins, particularly laminin 111 and fibronectin, and to modulate human myoblast migration. METHODS: We evaluated the expression of the HGF-receptor c-Met, laminin, and fibronectin receptors by immunoblotting, flow cytometry, or immunofluorescence and used Transwell assays to analyze myoblast migration on laminin 111 and fibronectin in the absence or presence of HGF. Zymography was used to check whether HGF could modulate the production of matrix metalloproteinases by human myoblasts, and the activation of MAPK/ERK pathways was evaluated by immunoblotting. RESULTS: We demonstrated that human myoblasts express c-Met, together with laminin and fibronectin receptors. We observed that human laminin 111 and fibronectin have a chemotactic effect on myoblast migration, and this was synergistically increased when low doses of HGF were added. We detected an increase in MMP-2 activity in myoblasts treated with HGF. Conversely, MMP-2 inhibition decreased the HGF-associated stimulation of cell migration triggered by laminin or fibronectin. HGF treatment also induced in human myoblasts activation of MAPK/ERK pathways, whose specific inhibition decreased the HGF-associated stimulus of cell migration triggered by laminin 111 or fibronectin. CONCLUSIONS: We demonstrate that HGF induces ERK phosphorylation and MMP production, thus stimulating human myoblast migration on ECM molecules. Conceptually, these data state that the mechanisms involved in the migration of human myoblasts comprise both soluble and insoluble moieties. This should be taken into account to optimize the design of therapeutic cell transplantation strategies by improving the migration of donor cells within the host tissue, a main issue regarding this approach.


Subject(s)
Cell Movement , Extracellular Matrix/metabolism , Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System , Matrix Metalloproteinases/metabolism , Myoblasts/metabolism , Cells, Cultured , Humans , Integrin alpha5beta1/metabolism , Matrix Metalloproteinases/genetics , Myoblasts/drug effects , Myoblasts/physiology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Laminin/metabolism
4.
Mol Ther ; 25(10): 2345-2359, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28750736

ABSTRACT

Although cell-based therapy is considered a promising method aiming at treating different muscular disorders, little clinical benefit has been reported. One of major hurdles limiting the efficiency of myoblast transfer therapy is the poor survival of the transplanted cells. Any intervention upon the donor cells focused on enhancing in vivo survival, proliferation, and expansion is essential to improve the effectiveness of such therapies in regenerative medicine. In the present work, we investigated the potential role of obestatin, an autocrine peptide factor regulating skeletal muscle growth and repair, to improve the outcome of myoblast-based therapy by xenotransplanting primary human myoblasts into immunodeficient mice. The data proved that short in vivo obestatin treatment of primary human myoblasts not only enhances the efficiency of engraftment, but also facilitates an even distribution of myoblasts in the host muscle. Moreover, this treatment leads to a hypertrophic response of the human-derived regenerating myofibers. Taken together, the activation of the obestatin/GPR39 pathway resulted in an overall improvement of the efficacy of cell engraftment within the host's skeletal muscle. These data suggest considerable potential for future therapeutic applications and highlight the importance of combinatorial therapies.


Subject(s)
Ghrelin/metabolism , Ghrelin/pharmacology , Myoblasts/drug effects , Myoblasts/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Humans , Injections, Intramuscular , Mice , Mice, SCID , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism
5.
Hum Gene Ther ; 27(2): 117-26, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26652770

ABSTRACT

Cell-based therapy for muscular dystrophies was initiated in humans after promising results obtained in murine models. Early trials failed to show substantial clinical benefit, sending researchers back to the bench, which led to the discovery of many hurdles as well as many new venues to optimize this therapeutic strategy. In this review we summarize progress in preclinical cell therapy approaches, with a special emphasis on human cells potentially attractive for human clinical trials. Future perspectives for cell therapy in skeletal muscle are discussed, including the perspective of combined therapeutic approaches.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Muscular Dystrophies/therapy , Myoblasts/transplantation , Pericytes/transplantation , Pluripotent Stem Cells/transplantation , Satellite Cells, Skeletal Muscle/transplantation , Animals , Clinical Trials as Topic , Humans , Mice , Muscle Development , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Myoblasts/cytology , Myoblasts/physiology , Pericytes/cytology , Pericytes/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Transplantation, Autologous , Transplantation, Homologous , Treatment Failure
6.
Diabetes ; 64(9): 3121-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25695947

ABSTRACT

Inflammation and lipid accumulation are hallmarks of muscular pathologies resulting from metabolic diseases such as obesity and type 2 diabetes. During obesity, the hypertrophy of visceral adipose tissue (VAT) contributes to muscle dysfunction, particularly through the dysregulated production of adipokines. We have investigated the cross talk between human adipocytes and skeletal muscle cells to identify mechanisms linking adiposity and muscular dysfunctions. First, we demonstrated that the secretome of obese adipocytes decreased the expression of contractile proteins in myotubes, consequently inducing atrophy. Using a three-dimensional coculture of human myotubes and VAT adipocytes, we showed the decreased expression of genes corresponding to skeletal muscle contractility complex and myogenesis. We demonstrated an increased secretion by cocultured cells of cytokines and chemokines with interleukin (IL)-6 and IL-1ß as key contributors. Moreover, we gathered evidence showing that obese subcutaneous adipocytes were less potent than VAT adipocytes in inducing these myotube dysfunctions. Interestingly, the atrophy induced by visceral adipocytes was corrected by IGF-II/insulin growth factor binding protein-5. Finally, we observed that the skeletal muscle of obese mice displayed decreased expression of muscular markers in correlation with VAT hypertrophy and abnormal distribution of the muscle fiber size. In summary, we show the negative impact of obese adipocytes on muscle phenotype, which could contribute to muscle wasting associated with metabolic disorders.


Subject(s)
Adipocytes/metabolism , Contractile Proteins/metabolism , Intra-Abdominal Fat/cytology , Muscle Fibers, Skeletal/metabolism , Obesity, Morbid/metabolism , Adipocytes/immunology , Adult , Animals , Atrophy/immunology , Atrophy/metabolism , Coculture Techniques , Cytokines/immunology , Female , Gene Expression Regulation , Humans , Inflammation , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor II/pharmacology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Obese , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/pathology , Obesity, Morbid/immunology , Subcutaneous Fat/cytology , Subcutaneous Fat/immunology , Subcutaneous Fat/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
7.
Neuropathol Appl Neurobiol ; 41(3): 270-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25405809

ABSTRACT

Despite considerable progress to increase our understanding of muscle genetics, pathophysiology, molecular and cellular partners involved in muscular dystrophies and muscle ageing, there is still a crucial need for effective treatments to counteract muscle degeneration and muscle wasting in such conditions. This review focuses on cell-based therapy for muscle diseases. We give an overview of the different parameters that have to be taken into account in such a therapeutic strategy, including the influence of muscle ageing, cell proliferation and migration capacities, as well as the translation of preclinical results in rodent into human clinical approaches. We describe recent advances in different types of human myogenic stem cells, with a particular emphasis on myoblasts but also on other candidate cells described so far [CD133+ cells, aldehyde dehydrogenase-positive cells (ALDH+), muscle-derived stem cells (MuStem), embryonic stem cells (ES) and induced pluripotent stem cells (iPS)]. Finally, we provide an update of ongoing clinical trials using cell therapy strategies.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Muscular Diseases/therapy , Humans , Stem Cells
8.
Neuromuscul Disord ; 23(1): 75-83, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23058947

ABSTRACT

Muscle repair relies on coordinated activation and differentiation of satellite cells, a process that is unable to counterbalance progressive degeneration in sporadic inclusion body myositis (s-IBM). To explore features of myo regeneration, the expression of myogenic regulatory factors Pax7, MyoD and Myogenin and markers of regenerating fibers was analyzed by immunohistochemistry in s-IBM muscle compared with polymyositis, dermatomyositis, muscular dystrophy and age-matched controls. In addition, the capillary density and number of interstitial CD34(+) hematopoietic progenitor cells was determined by double-immunoflourescence staining. Satellite cells and regenerating fibers were significantly increased in s-IBM similar to other inflammatory myopathies and correlated with the intensity of inflammation (R>0.428). Expression of MyoD, visualizing activated satellite cells and proliferating myoblasts, was lower in s-IBM compared to polymyosits. In contrast, Myogenin a marker of myogenic cell differentiation was strongly up-regulated in s-IBM muscle. The microvascular architecture in s-IBM was distorted, although the capillary density was normal. Notably, CD34(+) hematopoietic cells were significantly increased in the interstitial compartment. Our findings indicate profound myo-endothelial remodeling of s-IBM muscle concomitant to inflammation. An altered expression of myogenic regulatory factors involved in satellite cell activation and differentiation, however, might reflect perturbations of muscle repair in s-IBM.


Subject(s)
Cell Differentiation , Cell Proliferation , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Myogenic Regulatory Factors/metabolism , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Dermatomyositis/metabolism , Dermatomyositis/pathology , Dermatomyositis/physiopathology , Endothelium/blood supply , Endothelium/pathology , Endothelium/physiopathology , Female , Humans , Male , Microvessels/pathology , Middle Aged , Muscle, Skeletal/blood supply , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , MyoD Protein/metabolism , Myogenin/metabolism , Myositis, Inclusion Body/physiopathology , PAX7 Transcription Factor/metabolism , Polymyositis/metabolism , Polymyositis/pathology , Polymyositis/physiopathology , Regeneration , Young Adult
9.
Am J Pathol ; 181(4): 1387-401, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22871573

ABSTRACT

In most cases facioscapulohumeral muscular dystrophy (FSHD) is caused by contraction of the D4Z4 repeat in the 4q subtelomere. This contraction is associated with local chromatin decondensation and derepression of the DUX4 retrogene. Its complex genetic and epigenetic cause and high clinical variability in disease severity complicate investigations on the pathogenic mechanism underlying FSHD. A validated cellular model bypassing the considerable heterogeneity would facilitate mechanistic and therapeutic studies of FSHD. Taking advantage of the high incidence of somatic mosaicism for D4Z4 repeat contraction in de novo FSHD, we have established a clonal myogenic cell model from a mosaic patient. Individual clones are genetically identical except for the size of the D4Z4 repeat array, being either normal or FSHD sized. These clones retain their myogenic characteristics, and D4Z4 contracted clones differ from the noncontracted clones by the bursts of expression of DUX4 in sporadic nuclei, showing that this burst-like phenomenon is a locus-intrinsic feature. Consequently, downstream effects of DUX4 expression can be observed in D4Z4 contracted clones, like differential expression of DUX4 target genes. We also show their participation to in vivo regeneration with immunodeficient mice, further expanding the potential of these clones for mechanistic and therapeutic studies. These cell lines will facilitate pairwise comparisons to identify FSHD-specific differences and are expected to create new opportunities for high-throughput drug screens.


Subject(s)
Models, Biological , Mosaicism , Muscle Cells/pathology , Muscle Contraction/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Muscular Dystrophy, Facioscapulohumeral/physiopathology , Repetitive Sequences, Nucleic Acid/genetics , Adult , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line, Transformed , Cell Proliferation , Clone Cells , Electrophoresis, Gel, Pulsed-Field , Gene Expression Regulation , Humans , Mice , Middle Aged , Muscle Cells/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Myoblasts/metabolism , Regeneration/genetics , Telomere/genetics
10.
Arthritis Res Ther ; 13(6): R207, 2011.
Article in English | MEDLINE | ID: mdl-22171690

ABSTRACT

INTRODUCTION: Chronic inflammation is a profound systemic modification of the cellular microenvironment which could affect survival, repair and maintenance of muscle stem cells. The aim of this study was to define the role of chronic inflammation on the regenerative potential of satellite cells in human muscle. METHODS: As a model for chronic inflammation, 11 patients suffering from rheumatoid arthritis (RA) were included together with 16 patients with osteoarthritis (OA) as controls. The mean age of both groups was 64 years, with more females in the RA group compared to the OA group. During elective knee replacement surgery, a muscle biopsy was taken from the distal musculus vastus medialis. Cell populations from four RA and eight OA patients were used for extensive phenotyping because these cell populations showed no spontaneous differentiation and myogenic purity greater than 75% after explantation. RESULTS: After mononuclear cell explantation, myogenic purity, viability, proliferation index, number of colonies, myogenic colonies, growth speed, maximum number of population doublings and fusion index were not different between RA and OA patients. Furthermore, the expression of proteins involved in replicative and stress-induced premature senescence and apoptosis, including p16, p21, p53, hTERT and cleaved caspase-3, was not different between RA and OA patients. Mean telomere length was shorter in the RA group compared to the OA group. CONCLUSIONS: In the present study we found evidence that chronic inflammation in RA does not affect the in vitro regenerative potential of human satellite cells. Identification of mechanisms influencing muscle regeneration by modulation of its microenvironment may, therefore, be more appropriate.


Subject(s)
Cell Differentiation , Cell Proliferation , Inflammation/pathology , Satellite Cells, Skeletal Muscle/pathology , Aged , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Humans , Hydrogen Peroxide/pharmacology , Inflammation/metabolism , Male , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Oxidants/pharmacology , Satellite Cells, Skeletal Muscle/metabolism , Telomerase/metabolism , Telomere/genetics , Tumor Suppressor Protein p53/metabolism
11.
Eur J Appl Physiol ; 109(2): 323-30, 2010 May.
Article in English | MEDLINE | ID: mdl-20101406

ABSTRACT

Measuring the DNA telomere length of skeletal muscle in experienced endurance runners may contribute to our understanding of the effects of chronic exposure to endurance exercise on skeletal muscle. This study compared the minimum terminal restriction fragment (TRF) length in the vastus lateralis muscle of 18 experienced endurance runners (mean age: 42 +/- 7 years) to those of 19 sedentary individuals (mean age: 39 +/- 10 years). The runners had covered almost 50,000 km in training and racing over 15 years. Minimum TRF lengths measured in the muscle of both groups were similar (P = 0.805) and within the normal range. Minimum TRF length in the runners, however, was inversely related to their years spent running (r = -0.63, P = 0.007) and hours spent training (r = -0.52, P = 0.035). Therefore, since exposure to endurance running may influence minimum TRF length, and by implication, the proliferative potential of the satellite cells, chronic endurance running may be seen as a stressor to skeletal muscle.


Subject(s)
Athletes , Muscle, Skeletal/metabolism , Physical Endurance , Running/physiology , Telomere/metabolism , Adult , Athletes/statistics & numerical data , Female , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Running/statistics & numerical data
12.
Mol Ther ; 17(10): 1771-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19623164

ABSTRACT

In recent years, numerous reports have identified in mouse different sources of myogenic cells distinct from satellite cells that exhibited a variable myogenic potential in vivo. Myogenic stem cells have also been described in humans, although their regenerative potential has rarely been quantified. In this study, we have investigated the myogenic potential of human muscle-derived cells based on the expression of the stem cell marker CD133 as compared to bona fide satellite cells already used in clinical trials. The efficiency of these cells to participate in muscle regeneration and contribute to the renewal of the satellite cell pool, when injected intramuscularly, has been evaluated in the Rag2(-/-) gammaC(-/-) C5(-/-) mouse in which muscle degeneration is induced by cryoinjury. We demonstrate that human muscle-derived CD133+ cells showed a much greater regenerative capacity when compared to human myoblasts. The number of fibers expressing human proteins and the number of human cells in a satellite cell position are all dramatically increased when compared to those observed after injection of human myoblasts. In addition, CD133+/CD34+ cells exhibited a better dispersion in the host muscle when compared to human myoblasts. We propose that muscle-derived CD133+ cells could be an attractive candidate for cellular therapy.


Subject(s)
Antigens, CD/immunology , Glycoproteins/immunology , Muscle Development/physiology , Muscle, Skeletal/cytology , Myoblasts/cytology , Peptides/immunology , Stem Cells/cytology , AC133 Antigen , Animals , Cell Differentiation/physiology , Cells, Cultured , Humans , Mice , Mice, Mutant Strains , Muscle Development/genetics , Muscle, Skeletal/immunology , Myoblasts/physiology , Stem Cells/immunology , Stem Cells/physiology
13.
Neuromuscul Disord ; 19(3): 199-206, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19185491

ABSTRACT

Oculo-pharyngeal muscular dystrophy (OPMD) is characterised by progressive eyelid drooping (ptosis) and difficulties with swallowing (dysphagia). In order to determine the role of growth factors, cytokines and chemokines in the physiopathology of muscle disease we have compared the level of expression of 174 factors in both the affected (cricopharyngeal) and non-affected (sternocleidomastoid) muscles of 8 OPMD patients by means of antibody arrays. Despite an important inter-individual variability the expression of sixty factors was found to be persistently different between affected and non-affected muscles. Many of the differentially expressed factors in our study are known to be involved in the formation of fibrosis in both the liver and the lung, indicating the possibility that treatments such as those used in hepatic fibrosis may have a beneficial effect in OPMD patients.


Subject(s)
Cytokines/metabolism , Fibrosis/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Oculopharyngeal/metabolism , Aged , Aged, 80 and over , Biomarkers/analysis , Biomarkers/metabolism , Chemokines/analysis , Chemokines/metabolism , Computational Biology/methods , Cytokines/analysis , Female , Fibrosis/immunology , Fibrosis/physiopathology , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/analysis , Male , Middle Aged , Muscle, Skeletal/immunology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Oculopharyngeal/immunology , Muscular Dystrophy, Oculopharyngeal/physiopathology , Pilot Projects , Protein Array Analysis/methods , Proteomics/methods , Software
14.
Am J Pathol ; 174(1): 216-27, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19095965

ABSTRACT

In myotonic dystrophy, muscleblind-like protein 1 (MBNL1) protein binds specifically to expanded CUG or CCUG repeats, which accumulate as discrete nuclear foci, and this is thought to prevent its function in the regulation of alternative splicing of pre-mRNAs. There is strong evidence for the role of the MBNL1 gene in disease pathology, but the roles of two related genes, MBNL2 and MBNL3, are less clear. Using new monoclonal antibodies specific for each of the three gene products, we found that MBNL2 decreased during human fetal development and myoblast culture, while MBNL1 was unchanged. In Duchenne muscular dystrophy muscle, MBNL2 was elevated in immature, regenerating fibres compared with mature fibres, supporting some developmental role for MBNL2. MBNL3 was found only in C2C12 mouse myoblasts. Both MBNL1 and MBNL2 were partially sequestered by nuclear foci of expanded repeats in adult muscle and cultured cells from myotonic dystrophy patients. In adult muscle nucleoplasm, both proteins were reduced in myotonic dystrophy type 1 compared with an age-matched control. In normal human myoblast cultures, MBNL1 and MBNL2 always co-distributed but their distribution could change rapidly from nucleoplasmic to cytoplasmic. Functional differences between MBNL1 and MBNL2 have not yet been found and may prove quite subtle. The dominance of MBNL1 in mature, striated muscle would explain why ablation of the mouse mbnl1 gene alone is sufficient to cause a myotonic dystrophy.


Subject(s)
Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , RNA-Binding Proteins/metabolism , Antibodies, Monoclonal/immunology , Antibody Specificity , Blotting, Western , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm , Electrophoresis, Polyacrylamide Gel , Fetus , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Muscle, Skeletal/embryology , Myoblasts/cytology , Myoblasts/metabolism , Protein Transport/physiology , RNA, Small Interfering , Transfection
15.
Transplantation ; 85(4): 566-75, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18347536

ABSTRACT

BACKGROUND: One major challenge in developing cell therapy for muscle diseases is to define the best condition for the recipient's muscle to niche donor cells. We have examined the efficiency of human myoblast transplantation in an immunodeficient animal model, after local irradiation, as well as the potential impact of laminin on myoblast behavior. METHODS: Human myoblasts were injected into preirradiated tibialis anterior muscles from immunodeficient mice. The donor cell engraftment, proliferation, and laminin content within the transplanted muscles were evaluated by immunocytochemistry. Additionally, the effect of laminin upon myoblast proliferation, migration, and survival was ascertained in vitro. RESULTS: Engraftment of human myoblasts into the skeletal muscle of immunodeficient Rag2-/gammac-/C5- mice presubjected to local irradiation provided the best niche for myoblast engraftment, as demonstrated by the number of viable and proliferating donor cells found in the host muscle. Local irradiation significantly enhanced laminin deposition within the recipient's muscle and donor cells were preferentially located in laminin-enriched areas. The same batch of myoblasts used for in vivo injections also responded to laminin in vitro with increased proliferation and cell survival, as well as an improved migratory response. CONCLUSIONS: We show that local irradiation enhances the laminin content in the host muscle microenvironment and provides a better engraftment of human myoblasts. In addition, laminin increases myoblast proliferation, survival, and migration in vitro. These data provide combined in vivo and in vitro evidence that laminin status should be taken into account when designing experimental and clinical cell therapy strategies for muscle disease.


Subject(s)
Laminin/physiology , Myoblasts/transplantation , Animals , Cell Culture Techniques , Cell Division/physiology , Cell Division/radiation effects , Cell Separation , DNA-Binding Proteins/deficiency , Graft Survival/physiology , Graft Survival/radiation effects , Humans , Mice , Mice, Knockout , Muscle, Skeletal/physiology , Muscle, Skeletal/radiation effects , Myoblasts/cytology , Transplantation, Heterologous
16.
Proteomics ; 8(2): 264-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18203276

ABSTRACT

In the present study, modifications in cytosolic expressed proteins during human myoblast differentiation were studied by dialysis-assisted 2-DE (DAGE, [1]). About 1000 spots were analysed on the 5th and 13th day of differentiation with a dynamic range of protein expression exceeding 1000-fold. During myogenic differentiation, the number of nonmatching spots as well as the extent of quantitative differences between matched spots significantly increased. Over one hundred differentially expressed spots were excised and identified by MALDI-TOF MS. The differentiation-associated expression pattern of eight proteins was validated by Western blot analysis. Differential expression of several proteins was demonstrated for the first time in human myotubes. Interestingly, Ingenuity pathway analysis grouped 30 of these proteins into two overlapping networks containing as principal nodes IGF-1 and tumour necrosis factor, two proteins known to play a crucial role in cytogenesis. Our results illustrate the large rearrangement of the proteome during the differentiation of human myoblasts and provide evidence for new partners involved in this complex process.


Subject(s)
Cell Differentiation , Dialysis/methods , Electrophoresis, Gel, Two-Dimensional/methods , Myoblasts/chemistry , Proteomics/methods , Blotting, Western , Cytosol/chemistry , Factor XIII/analysis , Guanine Nucleotide Dissociation Inhibitors/analysis , Heterogeneous-Nuclear Ribonucleoprotein K/analysis , Humans , STAT1 Transcription Factor/analysis , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stathmin/analysis , rho-Specific Guanine Nucleotide Dissociation Inhibitors
17.
J Gene Med ; 10(2): 217-24, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18074402

ABSTRACT

BACKGROUND: The most common form of congenital muscular dystrophy is caused by a deficiency in the alpha2 chain of laminin-211, a protein of the extracellular matrix. A wide variety of mutations, including 20 to 30% of nonsense mutations, have been identified in the corresponding gene, LAMA2. A promising approach for the treatment of genetic disorders due to premature termination codons (PTCs) is the use of drugs to force stop codon readthrough. METHODS: Here, we analyzed the effects of two compounds on a PTC in the LAMA2 gene that targets the mRNA to nonsense-mediated RNA decay, in vitro using a dual reporter assay, as well as ex vivo in patient-derived myotubes. RESULTS: We first showed that both gentamicin and negamycin promote significant readthrough of this PTC. We then demonstrated that the mutant mRNAs were strongly stabilized in patient-derived myotubes after administration of negamycin, but not gentamicin. Nevertheless, neither treatment allowed re-expression of the laminin alpha2-chain protein, pointing to problems that may have arisen at the translational or post-translational levels. CONCLUSIONS: Taken together, our results emphasize that achievement of a clinical benefit upon treatment with novel readthrough-inducing agents would require several favourable conditions including PTC nucleotide context, intrinsic and induced stability of mRNA and correct synthesis of a full-length active protein.


Subject(s)
Codon, Nonsense/genetics , Gentamicins/pharmacology , Laminin/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/genetics , RNA Stability/drug effects , Amino Acids, Diamino/pharmacology , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Myosins/metabolism , NIH 3T3 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Biol Cell ; 100(3): 189-99, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17988214

ABSTRACT

BACKGROUND INFORMATION: Aging of human skeletal muscle results in a decline in muscle mass and force, and excessive turnover of muscle fibres, such as in muscular dystrophies, further increases this decline. Although it has been shown in rodents, by cross-age transplantation of whole muscles, that the environment plays an important role in this process, the implication of proliferating aging of the muscle progenitors has been poorly investigated, particularly in humans, since the regulation of cell proliferation differs between rodents and humans. The myogenic differentiation of human myoblasts is regulated by the muscle-specific regulatory factors. Cross-talk between the muscle-specific regulatory factors and the cell cycle regulators is essential for differentiation. The aim of the present study was to determine the effects of replicative senescence on the myogenic programme of human myoblasts. RESULTS: We showed that senescent myoblasts, which could not re-enter the cell cycle, are still able to differentiate and form multinucleated myotubes. However, these myotubes are significantly smaller. The expression of muscle-specific regulatory factors and cell cycle regulators was analysed in proliferating myoblasts and compared with senescent cells. We have observed a delay and a decrease in the muscle-specific regulatory factors and the cyclin-dependent kinase inhibitor p57 during the early step of differentiation in senescent myoblasts, as well as an increase in the fibroblastic markers. CONCLUSIONS: Our results demonstrate that replicative senescence alters the expression of the factors triggering muscle differentiation in human myoblasts and could play a role in the regenerative defects observed in muscular diseases and during normal skeletal-muscle aging.


Subject(s)
Cellular Senescence/genetics , Muscle, Skeletal/embryology , Myoblasts/metabolism , Myogenic Regulatory Factors/metabolism , Adolescent , Aging/genetics , Aging/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Division/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Down-Regulation/genetics , Fetus , Humans , Muscle, Skeletal/growth & development , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/physiopathology , Myoblasts/cytology , Myogenic Regulatory Factors/genetics , Regeneration/genetics
19.
Genes Cells ; 12(9): 1035-48, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17825047

ABSTRACT

Nuclear speckles are storage sites for small nuclear RNPs (snRNPs) and other splicing factors. Current ideas about the role of speckles suggest that some pre-mRNAs are processed at the speckle periphery before being exported as mRNA. In myotonic dystrophy type 1 (DM1), the export of mutant DMPK mRNA is prevented by the presence of expanded CUG repeats that accumulate in nuclear foci. We now show that these foci accumulate at the periphery of nuclear speckles. In myotonic dystrophy type 2 (DM2), mRNA from the mutant ZNF9 gene is exported normally because the expanded CCUG repeats are removed during splicing. We now show that the nuclear foci formed by DM2 intronic repeats are widely dispersed in the nucleoplasm and not associated with either nuclear speckles or exosomes. We hypothesize that the expanded CUG repeats in DMPK mRNA are blocking a stage in its export pathway that would normally occur at the speckle periphery. Localization of the expanded repeats at the speckle periphery is not essential for their pathogenic effects because DM1 and DM2 are quite similar clinically.


Subject(s)
Cell Nucleus Structures/metabolism , Myotonic Dystrophy/genetics , Protein Serine-Threonine Kinases/genetics , RNA Splicing/physiology , RNA, Messenger/metabolism , Antibodies/immunology , Cell Nucleus/metabolism , Fetus/metabolism , HeLa Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , In Situ Hybridization , Introns , Models, Biological , Mutation , Myotonic Dystrophy/classification , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Splicing Factors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Trinucleotide Repeats
20.
Aging Cell ; 6(4): 515-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17559502

ABSTRACT

Cultured human myoblasts fail to immortalize following the introduction of telomerase. The availability of an immortalization protocol for normal human myoblasts would allow one to isolate cellular models from various neuromuscular diseases, thus opening the possibility to develop and test novel therapeutic strategies. The parameters limiting the efficacy of myoblast transfer therapy (MTT) could be assessed in such models. Finally, the presence of an unlimited number of cell divisions, and thus the ability to clone cells after experimental manipulations, reduces the risks of insertional mutagenesis by many orders of magnitude. This opportunity for genetic modification provides an approach for creating a universal donor that has been altered to be more therapeutically useful than its normal counterpart. It can be engineered to function under conditions of chronic damage (which are very different than the massive regeneration conditions that recapitulate normal development), and to overcome the biological problems such as cell death and failure to proliferate and migrate that limit current MTT strategies. We describe here the production and characterization of a human myogenic cell line, LHCN-M2, that has overcome replicative aging due to the expression of telomerase and cyclin-dependent kinase 4. We demonstrate that it functions as well as young myoblasts in xenotransplant experiments in immunocompromized mice under conditions of regeneration following muscle damage.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Cyclin-Dependent Kinase 4/metabolism , Muscle Development , Muscles/physiology , Myoblasts/physiology , Telomerase/metabolism , Animals , Cell Division , Cell Line , Genes, cdc , Humans , Mice , Muscular Dystrophies/therapy , Myoblasts/cytology , Myoblasts/enzymology , Myoblasts/transplantation , Regeneration , Satellite Cells, Skeletal Muscle/physiology , Telomere , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL