Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Lancet Neurol ; 22(10): 890-899, 2023 10.
Article En | MEDLINE | ID: mdl-37739572

BACKGROUND: Drug repurposing could provide novel treatment options for Duchenne muscular dystrophy. Because tamoxifen-an oestrogen receptor regulator-reduced signs of muscular pathology in a Duchenne muscular dystrophy mouse model, we aimed to assess the safety and efficacy of tamoxifen in humans as an adjunct to corticosteroid therapy over a period of 48 weeks. METHODS: We did a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial at 12 study centres in seven European countries. We enrolled ambulant boys aged 6·5-12·0 years with a genetically confirmed diagnosis of Duchenne muscular dystrophy and who were on stable corticosteroid treatment for more than 6 months. Exclusion criteria included ophthalmological disorders, including cataracts, and haematological disorders. We randomly assigned (1:1) participants using an online randomisation tool to either 20 mg tamoxifen orally per day or matched placebo, stratified by centre and corticosteroid intake. Participants, caregivers, and clinical investigators were masked to treatment assignments. Tamoxifen was taken in addition to standard care with corticosteroids, and participants attended study visits for examinations every 12 weeks. The primary efficacy outcome was the change from baseline to week 48 in scores on the D1 domain of the Motor Function Measure in the intention-to-treat population (defined as all patients who fulfilled the inclusion criteria and began treatment). This study is registered with ClinicalTrials.gov (NCT03354039) and is completed. FINDINGS: Between May 24, 2018, and Oct 14, 2020, 95 boys were screened for inclusion, and 82 met inclusion criteria and were initially enrolled into the study. Three boys were excluded after initial screening due to cataract diagnosis or revoked consent directly after screening, but before randomisation. A further boy assigned to the placebo group did not begin treatment. Therefore, 40 individuals assigned tamoxifen and 38 allocated placebo were included in the intention-to-treat population. The primary efficacy outcome did not differ significantly between tamoxifen (-3·05%, 95% CI -7·02 to 0·91) and placebo (-6·15%, -9·19 to -3·11; 2·90% difference, -3·02 to 8·82, p=0·33). Severe adverse events occurred in two participants: one participant who received tamoxifen had a fall, and one who received placebo suffered a panic attack. No deaths or life-threatening serious adverse events occurred. Viral infections were the most common adverse events. INTERPRETATION: Tamoxifen was safe and well tolerated, but no difference between groups was reported for the primary efficacy endpoint. Slower disease progression, defined by loss of motor function over time, was indicated in the tamoxifen group compared with the placebo group, but differences in outcome measures were neither clinically nor statistically significant. Currently, we cannot recommend the use of tamoxifen in daily clinical practice as a treatment option for boys with Duchenne muscular dystrophy due to insufficient clinical evidence. FUNDING: Thomi Hopf Foundation, ERA-Net, Swiss National Science Foundation, Duchenne UK, Joining Jack, Duchenne Parent Project, Duchenne Parent Project Spain, Fondation Suisse de Recherche sur les Maladies Musculaires, Association Monegasque contre les Myopathies.


Muscular Dystrophy, Duchenne , Male , Animals , Mice , Humans , Muscular Dystrophy, Duchenne/drug therapy , Double-Blind Method , Disease Models, Animal , Drug Repositioning , Ethnicity
2.
Mol Ther ; 29(8): 2514-2534, 2021 08 04.
Article En | MEDLINE | ID: mdl-33940157

Omics analyses are powerful methods to obtain an integrated view of complex biological processes, disease progression, or therapy efficiency. However, few studies have compared different disease forms and different therapy strategies to define the common molecular signatures representing the most significant implicated pathways. In this study, we used RNA sequencing and mass spectrometry to profile the transcriptomes and proteomes of mouse models for three forms of centronuclear myopathies (CNMs), untreated or treated with either a drug (tamoxifen), antisense oligonucleotides reducing the level of dynamin 2 (DNM2), or following modulation of DNM2 or amphiphysin 2 (BIN1) through genetic crosses. Unsupervised analysis and differential gene and protein expression were performed to retrieve CNM molecular signatures. Longitudinal studies before, at, and after disease onset highlighted potential disease causes and consequences. Main pathways in the common CNM disease signature include muscle contraction, regeneration and inflammation. The common therapy signature revealed novel potential therapeutic targets, including the calcium regulator sarcolipin. We identified several novel biomarkers validated in muscle and/or plasma through RNA quantification, western blotting, and enzyme-linked immunosorbent assay (ELISA) assays, including ANXA2 and IGFBP2. This study validates the concept of using multi-omics approaches to identify molecular signatures common to different disease forms and therapeutic strategies.


Gene Expression Profiling/methods , Myopathies, Structural, Congenital/drug therapy , Oligonucleotides, Antisense/therapeutic use , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proteomics/methods , Tamoxifen/therapeutic use , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Disease Models, Animal , Dynamin II/antagonists & inhibitors , Humans , Longitudinal Studies , Mass Spectrometry , Mice , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Sequence Analysis, RNA , Tumor Suppressor Proteins/antagonists & inhibitors
3.
Hum Mol Genet ; 28(12): 1931-1946, 2019 06 15.
Article En | MEDLINE | ID: mdl-30590522

Mutations in OCRL encoding the inositol polyphosphate 5-phosphatase OCRL (Lowe oculocerebrorenal syndrome protein) disrupt phosphoinositide homeostasis along the endolysosomal pathway causing dysfunction of the cells lining the kidney proximal tubule (PT). The dysfunction can be isolated (Dent disease 2) or associated with congenital cataracts, central hypotonia and intellectual disability (Lowe syndrome). The mechanistic understanding of Dent disease 2/Lowe syndrome remains scarce due to limitations of animal models of OCRL deficiency. Here, we investigate the role of OCRL in Dent disease 2/Lowe syndrome by using OcrlY/- mice, where the lethal deletion of the paralogue Inpp5b was rescued by human INPP5B insertion, and primary culture of proximal tubule cells (mPTCs) derived from OcrlY/- kidneys. The OcrlY/- mice show muscular defects with dysfunctional locomotricity and present massive urinary losses of low-molecular-weight proteins and albumin, caused by selective impairment of receptor-mediated endocytosis in PT cells. The latter was due to accumulation of phosphatidylinositol 4,5-bisphosphate PI(4,5)P2 in endolysosomes, driving local hyper-polymerization of F-actin and impairing trafficking of the endocytic LRP2 receptor, as evidenced in OcrlY/- mPTCs. The OCRL deficiency was also associated with a disruption of the lysosomal dynamic and proteolytic activity. Partial convergence of disease-pathways and renal phenotypes observed in OcrlY/- and Clcn5Y/- mice suggest shared mechanisms in Dent diseases 1 and 2. These studies substantiate the first mouse model of Lowe syndrome and give insights into the role of OCRL in cellular trafficking of multiligand receptors. These insights open new avenues for therapeutic interventions in Lowe syndrome and Dent disease.


Dent Disease/genetics , Endosomes/metabolism , Kidney Tubules, Proximal/metabolism , Lysosomes/metabolism , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Actins/metabolism , Animals , Cells, Cultured , Chloride Channels/genetics , Dent Disease/metabolism , Dent Disease/physiopathology , Disease Models, Animal , Endocytosis/genetics , Humans , Kidney/physiopathology , Kidney Tubules, Proximal/physiopathology , Locomotion/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/physiopathology , Phosphatidylinositol 4,5-Diphosphate/metabolism
4.
Nat Commun ; 9(1): 4848, 2018 11 19.
Article En | MEDLINE | ID: mdl-30451843

X-linked myotubular myopathy (XLMTM, also known as XLCNM) is a severe congenital muscular disorder due to mutations in the myotubularin gene, MTM1. It is characterized by generalized hypotonia, leading to neonatal death of most patients. No specific treatment exists. Here, we show that tamoxifen, a well-known drug used against breast cancer, rescues the phenotype of Mtm1-deficient mice. Tamoxifen increases lifespan several-fold while improving overall motor function and preventing disease progression including lower limb paralysis. Tamoxifen corrects functional, histological and molecular hallmarks of XLMTM, with improved force output, myonuclei positioning, myofibrillar structure, triad number, and excitation-contraction coupling. Tamoxifen normalizes the expression level of the XLMTM disease modifiers DNM2 and PI3KC2B, likely contributing to the phenotypic rescue. Our findings demonstrate that tamoxifen is a promising candidate for clinical evaluation in XLMTM patients.


Motor Activity/drug effects , Muscle, Skeletal/drug effects , Myopathies, Structural, Congenital/drug therapy , Protective Agents/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Tamoxifen/pharmacology , Animals , Class II Phosphatidylinositol 3-Kinases/genetics , Class II Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Disease Progression , Dynamin II/genetics , Dynamin II/metabolism , Electric Stimulation , Excitation Contraction Coupling/drug effects , Female , Gene Expression/drug effects , Genes, Lethal , Humans , Longevity/drug effects , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myofibrils/drug effects , Myofibrils/metabolism , Myofibrils/ultrastructure , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/metabolism , Myopathies, Structural, Congenital/pathology , Protein Tyrosine Phosphatases, Non-Receptor/deficiency
5.
Chimia (Aarau) ; 72(4): 238-240, 2018 Apr 25.
Article En | MEDLINE | ID: mdl-29720316

Drug discovery is a long, expensive and risky process. Evaluating drugs that have already been proved safe for use in humans and testing them for a new indication greatly reduces the time and monetary costs involved in finding treatments for life-threatening conditions. Here tamoxifen, a drug that is used for the treatment of breast cancer, is investigated in a mouse model of Duchenne muscular dystrophy. Tamoxifen was efficacious in countering the symptoms of the disease without affecting the underlying genetic cause. Based on these results, tamoxifen has been tested in other forms of muscle disease with success. Drug repurposing may not only be a cost-effective manner for treating a variety of diseases, it may also help us uncover common mechanisms between conditions that were previously thought to be unrelated.


Muscular Dystrophy, Duchenne/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Animals , Female , Humans , Male , Mice
6.
Mol Ther ; 26(4): 1093-1108, 2018 04 04.
Article En | MEDLINE | ID: mdl-29503200

Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease currently without cure. We investigated the use of the PiggyBac transposon for full-length dystrophin expression in murine mesoangioblast (MABs) progenitor cells. DMD murine MABs were transfected with transposable expression vectors for full-length dystrophin and transplanted intramuscularly or intra-arterially into mdx/SCID mice. Intra-arterial delivery indicated that the MABs could migrate to regenerating muscles to mediate dystrophin expression. Intramuscular transplantation yielded dystrophin expression in 11%-44% of myofibers in murine muscles, which remained stable for the assessed period of 5 months. The satellite cells isolated from transplanted muscles comprised a fraction of MAB-derived cells, indicating that the transfected MABs may colonize the satellite stem cell niche. Transposon integration site mapping by whole-genome sequencing indicated that 70% of the integrations were intergenic, while none was observed in an exon. Muscle resistance assessment by atomic force microscopy indicated that 80% of fibers showed elasticity properties restored to those of wild-type muscles. As measured in vivo, transplanted muscles became more resistant to fatigue. This study thus provides a proof-of-principle that PiggyBac transposon vectors may mediate full-length dystrophin expression as well as functional amelioration of the dystrophic muscles within a potential autologous cell-based therapeutic approach of DMD.


Cell- and Tissue-Based Therapy , DNA Transposable Elements , Gene Transfer Techniques , Genetic Vectors/genetics , Muscular Dystrophy, Duchenne/genetics , Myoblasts/metabolism , Myoblasts/transplantation , Animals , Cell Line , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Dystrophin/genetics , Fluorescent Antibody Technique , Gene Dosage , Gene Expression , Gene Order , Genes, Reporter , Male , Mice , Mice, Inbred mdx , Mice, SCID , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Phenotype , Transgenes , Transplantation, Autologous
7.
Front Physiol ; 6: 254, 2015.
Article En | MEDLINE | ID: mdl-26441673

Weight regain after caloric restriction results in accelerated fat storage in adipose tissue. This catch-up fat phenomenon is postulated to result partly from suppressed skeletal muscle thermogenesis, but the underlying mechanisms are elusive. We investigated whether the reduced rate of skeletal muscle contraction-relaxation cycle that occurs after caloric restriction persists during weight recovery and could contribute to catch-up fat. Using a rat model of semistarvation-refeeding, in which fat recovery is driven by suppressed thermogenesis, we show that contraction and relaxation of leg muscles are slower after both semistarvation and refeeding. These effects are associated with (i) higher expression of muscle deiodinase type 3 (DIO3), which inactivates tri-iodothyronine (T3), and lower expression of T3-activating enzyme, deiodinase type 2 (DIO2), (ii) slower net formation of T3 from its T4 precursor in muscles, and (iii) accumulation of slow fibers at the expense of fast fibers. These semistarvation-induced changes persisted during recovery and correlated with impaired expression of transcription factors involved in slow-twitch muscle development. We conclude that diminished muscle thermogenesis following caloric restriction results from reduced muscle T3 levels, alteration in muscle-specific transcription factors, and fast-to-slow fiber shift causing slower contractility. These energy-sparing effects persist during weight recovery and contribute to catch-up fat.

8.
PLoS One ; 9(10): e110708, 2014.
Article En | MEDLINE | ID: mdl-25329652

Elevation of intracellular Ca2+, excessive ROS production and increased phospholipase A2 activity contribute to the pathology in dystrophin-deficient muscle. Moreover, Ca2+, ROS and phospholipase A2, in particular iPLA2, are thought to potentiate each other in positive feedback loops. NADPH oxidases (NOX) have been considered as a major source of ROS in muscle and have been reported to be overexpressed in muscles of mdx mice. We report here on our investigations regarding the effect of diapocynin, a dimer of the commonly used NOX inhibitor apocynin, on the activity of iPLA2, Ca2+ handling and ROS generation in dystrophic myotubes. We also examined the effects of diapocynin on force production and recovery ability of isolated EDL muscles exposed to eccentric contractions in vitro, a damaging procedure to which dystrophic muscle is extremely sensitive. In dystrophic myotubes, diapocynin inhibited ROS production, abolished iPLA2 activity and reduced Ca2+ influx through stretch-activated and store-operated channels, two major pathways responsible for excessive Ca2+ entry in dystrophic muscle. Diapocynin also prevented force loss induced by eccentric contractions of mdx muscle close to the value of wild-type muscle and reduced membrane damage as seen by Procion orange dye uptake. These findings support the central role played by NOX-ROS in the pathogenic cascade leading to muscular dystrophy and suggest diapocynin as an effective NOX inhibitor that might be helpful for future therapeutic approaches.


Acetophenones/administration & dosage , Biphenyl Compounds/administration & dosage , Dystrophin/genetics , Muscular Dystrophy, Duchenne/drug therapy , NADPH Oxidases/antagonists & inhibitors , Phospholipases A2/metabolism , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Calcium Signaling/drug effects , Dystrophin/deficiency , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , NADPH Oxidases/metabolism , Phospholipases A2/biosynthesis , Reactive Oxygen Species/metabolism
9.
Am J Pathol ; 182(2): 485-504, 2013 Feb.
Article En | MEDLINE | ID: mdl-23332367

Duchenne muscular dystrophy (DMD) is a severe disorder characterized by progressive muscle wasting,respiratory and cardiac impairments, and premature death. No treatment exists so far, and the identification of active substances to fight DMD is urgently needed. We found that tamoxifen, a drug used to treat estrogen-dependent breast cancer, caused remarkable improvements of muscle force and of diaphragm and cardiac structure in the mdx(5Cv) mouse model of DMD. Oral tamoxifen treatment from 3 weeks of age for 15 months at a dose of 10 mg/kg/day stabilized myofiber membranes, normalized whole body force, and increased force production and resistance to repeated contractions of the triceps muscle above normal values. Tamoxifen improved the structure of leg muscles and diminished cardiac fibrosis by~ 50%. Tamoxifen also reduced fibrosis in the diaphragm, while increasing its thickness,myofiber count, and myofiber diameter, thereby augmenting by 72% the amount of contractile tissue available for respiratory function. Tamoxifen conferred a markedly slower phenotype to the muscles.Tamoxifen and its metabolites were present in nanomolar concentrations in plasma and muscles,suggesting signaling through high-affinity targets. Interestingly, the estrogen receptors ERa and ERb were several times more abundant in dystrophic than in normal muscles, and tamoxifen normalized the relative abundance of ERb isoforms. Our findings suggest that tamoxifen might be a useful therapy for DMD.


Antineoplastic Agents/therapeutic use , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Behavior, Animal/drug effects , Biomarkers/metabolism , Biomechanical Phenomena/drug effects , Body Weight/drug effects , Creatine Kinase/blood , Diaphragm/pathology , Diaphragm/physiopathology , Disease Models, Animal , Feeding Behavior/drug effects , Fibrosis , Mice , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/blood , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/physiopathology , Myocardium/pathology , Organ Size/drug effects , Receptors, Estrogen/metabolism , Tamoxifen/blood , Tamoxifen/pharmacology
10.
Histochem Cell Biol ; 137(6): 811-27, 2012 Jun.
Article En | MEDLINE | ID: mdl-22331205

In two separate previous studies, we reported that subcutaneous (sc) or oral administration of (-)-epigallocatechin-3-gallate (EGCG) limited the development of muscle degeneration of mdx mice, a mild phenotype model for Duchenne muscular dystrophy (DMD). However, it was not possible to conclude which was the more efficient route of EGCG administration because different strains of mdx mice, periods of treatment and methods of assessment were used. In this study, we investigated which administration routes and dosages of EGCG are the most effective for limiting the onset of dystrophic lesions in the same strain of mdx mice and applying the same methods of assessment. Three-week-old mdx mice were injected sc for 5 weeks with either saline or a daily average of 3 or 6 mg/kg EGCG. For comparison, age-matched mdx mice were fed for 5 weeks with either a diet containing 0.1% EGCG or a control diet. The effects of EGCG were assessed quantitatively by determining the activities of serum muscle-derived creatine kinase, isometric contractions of triceps surae muscles, integrated spontaneous locomotor activities, and oxidative stress and fibrosis in selected muscles. Oral administration of 180 mg/kg/day EGCG in the diet was found the most effective for significantly improving several parameters associated with muscular dystrophy. However, the improvements were slightly less than those observed previously for sc injection started immediately after birth. The efficacy of EGCG for limiting the development of dystrophic muscle lesions in mice suggests that EGCG may be of benefit for DMD patients.


Antioxidants/pharmacology , Catechin/analogs & derivatives , Tea/chemistry , Animals , Body Weight , Catechin/pharmacology , Disease Models, Animal , Eating , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Oxidative Stress , Polyphenols/pharmacology
11.
Am J Pathol ; 180(2): 749-62, 2012 Feb.
Article En | MEDLINE | ID: mdl-22192627

In Duchenne muscular dystrophy, the absence of dystrophin causes progressive muscle wasting and premature death. Excessive calcium influx is thought to initiate the pathogenic cascade, resulting in muscle cell death. Urocortins (Ucns) have protected muscle in several experimental paradigms. Herein, we demonstrate that daily s.c. injections of either Ucn 1 or Ucn 2 to 3-week-old dystrophic mdx(5Cv) mice for 2 weeks increased skeletal muscle mass and normalized plasma creatine kinase activity. Histological examination showed that Ucns remarkably reduced necrosis in the diaphragm and slow- and fast-twitch muscles. Ucns improved muscle resistance to mechanical stress provoked by repetitive tetanizations. Ucn 2 treatment resulted in faster kinetics of contraction and relaxation and a rightward shift of the force-frequency curve, suggesting improved calcium homeostasis. Ucn 2 decreased calcium influx into freshly isolated dystrophic muscles. Pharmacological manipulation demonstrated that the mechanism involved the corticotropin-releasing factor type 2 receptor, cAMP elevation, and activation of both protein kinase A and the cAMP-binding protein Epac. Moreover, both STIM1, the calcium sensor that initiates the assembly of store-operated channels, and the calcium-independent phospholipase A(2) that activates these channels were reduced in dystrophic muscle by Ucn 2. Altogether, our results demonstrate the high potency of Ucns for improving dystrophic muscle structure and function, suggesting that these peptides may be considered for treatment of Duchenne muscular dystrophy.


Acetylcysteine/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/metabolism , Erythromycin/analogs & derivatives , Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/drug therapy , Urocortins/pharmacology , Acetylcysteine/antagonists & inhibitors , Acetylcysteine/metabolism , Animals , Brefeldin A/pharmacology , Calcium/metabolism , Calcium Channels , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Erythromycin/antagonists & inhibitors , Erythromycin/metabolism , Group VI Phospholipases A2/metabolism , Injections, Intradermal , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle Strength/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Protein Kinase Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Stromal Interaction Molecule 1
12.
J Pineal Res ; 51(2): 163-71, 2011 Sep.
Article En | MEDLINE | ID: mdl-21486366

Duchenne muscular dystrophy (DMD) is a severe X-linked muscle-wasting disease caused by the absence of the cytoskeletal protein dystrophin. In addition to abnormal calcium handling, numerous studies point to a crucial role of oxidative stress in the pathogenesis of the disease. Considering the impressive results provided by antioxidants on dystrophic muscle structure and function, we investigated whether melatonin can protect the mdx(5Cv) mouse, an animal model for DMD. Male mdx(5Cv) mouse pups were treated with melatonin by daily intraperitoneal (i.p.) injection (30 mg/kg body weight) or by subcutaneous (s.c.) implant(s) (18 or 54 mg melatonin as Melovine® implants) from 17/18 to 28/29 days of age. Isometric force of the triceps surae was recorded at the end of the treatment. The i.p. treatment increased the phasic twitch tension of mdx(5Cv) mice. The maximal tetanic tension was ameliorated by 18 mg s.c. and 30 mg/kg i.p. treatments. Melatonin caused the dystrophic muscle to contract and relax faster. The force-frequency relationship of melatonin-treated dystrophic mice was shifted to the right. In accordance with improved muscle function, melatonin decreased plasma creatine kinase activity, a marker for muscle injury. Melatonin treatment increased total glutathione content and lowered the oxidized/reduced glutathione ratio, indicating a better redox status of the muscle. In light of the present investigation, the therapeutic potential of melatonin should be further considered for patients with DMD.


Antioxidants/pharmacology , Isometric Contraction/drug effects , Melatonin/pharmacology , Muscle Relaxation/drug effects , Muscle Strength/drug effects , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Animals , Creatinine/blood , Disease Models, Animal , Glutathione/blood , Humans , Isometric Contraction/genetics , Male , Mice , Mice, Inbred mdx , Muscle Relaxation/genetics , Muscle Strength/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Oxidation-Reduction/drug effects
13.
Biofactors ; 35(3): 279-94, 2009.
Article En | MEDLINE | ID: mdl-19322813

Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by the absence of the protein dystrophin. Because oxidative stress contributes to the pathogenesis of DMD, we investigated if a green tea polyphenol blend (GTP) and its major polyphenol (-)-epigallocatechin gallate (EGCg), could protect muscle cell primary cultures from oxidative damage induced by hydrogen peroxide (H(2)O(2)) in the widely used mdx mouse model. On-line fluorimetric measurements using an H(2)O(2)-sensitive probe indicated that GTP and EGCg scavenged peroxide in a concentration-dependent manner. A 48 h exposure to EGCg increased glutathione content but did not alter the expression of proteins involved in membrane stabilization and repair. Pretreatment of dystrophic cultures with GTP or EGCg 48 h before exposure to H(2)O(2) improved cell survival. Normal cultures were protected by GTP but not by EGCg. 67LR, a receptor for EGCg, was seven times more abundant in dystrophic compared with normal cultures. Altogether our results demonstrate that GTP and EGCg protect muscle cells by scavenging H(2)O(2) and by improving the glutathione balance. In addition, the higher levels of 67LR in dystrophic muscle cells compared with normal ones likely contribute to EGCg-mediated survival.


Flavonoids/pharmacology , Glutathione/metabolism , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Phenols/pharmacology , Receptors, Laminin/metabolism , Tea/chemistry , Animals , Animals, Newborn , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Survival/drug effects , Cells, Cultured , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Mutant Strains , Oxidative Stress/drug effects , Polyphenols , Protease Inhibitors/pharmacology
14.
Mol Ther ; 17(1): 19-25, 2009 Jan.
Article En | MEDLINE | ID: mdl-19002166

Dystrophin mediates a physical link between the cytoskeleton of muscle fibers and the extracellular matrix, and its absence leads to muscle degeneration and dystrophy. In this article, we show that the lack of dystrophin affects the elasticity of individual fibers within muscle tissue explants, as probed using atomic force microscopy (AFM), providing a sensitive and quantitative description of the properties of normal and dystrophic myofibers. The rescue of dystrophin expression by exon skipping or by the ectopic expression of the utrophin analogue normalized the elasticity of dystrophic muscles, and these effects were commensurate to the functional recovery of whole muscle strength. However, a more homogeneous and widespread restoration of normal elasticity was obtained by the exon-skipping approach when comparing individual myofibers. AFM may thus provide a quantification of the functional benefit of gene therapies from live tissues coupled to single-cell resolution.


Dystrophin/genetics , Genetic Therapy/methods , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Animals , Dependovirus/genetics , Elasticity , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Microscopy, Atomic Force , Muscular Dystrophy, Animal/physiopathology
15.
Virus Res ; 123(1): 30-9, 2007 Jan.
Article En | MEDLINE | ID: mdl-16956688

Coxsackieviruses A (CVA) are associated with several clinical manifestations such as aseptic meningitis and paralytic syndromes in humans. Most CVA are difficult-to-cultivate, which impedes their propagation and isolation from clinical material. Here, we tested the ability of cultivable (CVA-13, CVA-14), and difficult-to-cultivate (CVA-6, CVA-22) strains to infect primary cultures of skeletal muscle cells established from newborn mice. We found that such cultures sustained the multiplication of these CVA, as evidenced by the development of a cytopathic effect, already in the initial preparation or after passaging once. Cultures established for no more than 24h were sensitive to infection whereas older preparations were resistant. Using confocal microscopy after double-immunolabeling of the VP1 capsid protein and the muscle cell marker myosin, we demonstrated that only the myoblasts were infected, resulting in VP1 expression throughout their cytoplasm. Inoculation of infected cultures to suckling mice resulted in paralysis indicating that infection was productive. The nature of candidate receptors for virus entry in such cultures and the influence of cell culture conditions on the expression of these putative receptors are discussed. This work suggests that primary cultures of skeletal muscle cells could be used to propagate and isolate any CVA strain.


Enterovirus A, Human/growth & development , Enterovirus Infections/virology , Virus Cultivation/methods , Animals , Animals, Newborn , Capsid Proteins/metabolism , Cells, Cultured , Cytoplasm/metabolism , Mice , Muscle, Skeletal/virology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/virology
16.
J Cell Sci ; 119(Pt 18): 3733-42, 2006 Sep 15.
Article En | MEDLINE | ID: mdl-16926189

Duchenne muscular dystrophy is caused by deficiency of dystrophin and leads to progressive weakness. It has been proposed that the muscle degeneration occurring in this disease is caused by increased Ca2+ influx due to enhanced activity of cationic channels that are activated either by stretch of the plasma membrane (stretch-activated channels) or by Ca2+-store depletion (store-operated channels). Using both cytosolic Ca2+ measurements with Fura-2 and the manganese quench method, we show here that store-operated Ca2+ entry is greatly enhanced in dystrophic skeletal flexor digitorum brevis fibers isolated from mdx(5cv) mice, a mouse model of Duchenne muscular dystrophy. Moreover, we show for the first time that store-operated Ca2+ entry in these fibers is under the control of the Ca2+-independent phospholipase A2 and that the exaggerated Ca2+ influx can be completely attenuated by inhibitors of this enzyme. Enhanced store-operated Ca2+ entry in dystrophic fibers is likely to be due to a near twofold overexpression of Ca2+-independent phospholipase A2. The Ca2+-independent phospholipase A2 pathway therefore appears as an attractive target to reduce excessive Ca2+ influx and subsequent degeneration occurring in dystrophic fibers.


Calcium Signaling/physiology , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/enzymology , Muscular Dystrophy, Animal/chemically induced , Phospholipases A/metabolism , Anilides/pharmacology , Animals , Caffeine/pharmacology , Calcium Channels/metabolism , Calcium Signaling/drug effects , Group VI Phospholipases A2 , Ion Transport/drug effects , Manganese/metabolism , Melitten/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Models, Biological , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Naphthalenes/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Potassium Chloride/metabolism , Pyrones/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thiadiazoles/pharmacology
17.
Am J Physiol Cell Physiol ; 290(2): C616-25, 2006 Feb.
Article En | MEDLINE | ID: mdl-16403950

Duchenne muscular dystrophy is a frequent muscular disorder caused by mutations in the gene encoding dystrophin, a cytoskeletal protein that contributes to the stabilization of muscle fiber membrane during muscle activity. Affected individuals show progressive muscle wasting that generally causes death by age 30. In this study, the dystrophic mdx(5Cv) mouse model was used to investigate the effects of green tea extract, its major component (-)-epigallocatechin gallate, and pentoxifylline on dystrophic muscle quality and function. Three-week-old mdx(5Cv) mice were fed for either 1 or 5 wk a control chow or a chow containing the test substances. Histological examination showed a delay in necrosis of the extensor digitorum longus muscle in treated mice. Mechanical properties of triceps surae muscles were recorded while the mice were under deep anesthesia. Phasic and tetanic tensions of treated mice were increased, reaching values close to those of normal mice. The phasic-to-tetanic tension ratio was corrected. Finally, muscles from treated mice exhibited 30-50% more residual force in a fatigue assay. These results demonstrate that diet supplementation of dystrophic mdx(5Cv) mice with green tea extract or (-)-epigallocatechin gallate protected muscle against the first massive wave of necrosis and stimulated muscle adaptation toward a stronger and more resistant phenotype.


Antioxidants/pharmacology , Camellia sinensis/chemistry , Catechin/analogs & derivatives , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/metabolism , Plant Extracts/pharmacology , Plant Preparations/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/therapeutic use , Catechin/pharmacology , Catechin/therapeutic use , Diet , Disease Models, Animal , Free Radical Scavengers/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Pentoxifylline/pharmacology , Phytotherapy , Plant Extracts/therapeutic use , Plant Preparations/therapeutic use
18.
J Biol Chem ; 279(45): 47092-100, 2004 Nov 05.
Article En | MEDLINE | ID: mdl-15322099

In skeletal muscle cells, plasma membrane depolarization causes a rapid calcium release from the sarcoplasmic reticulum through ryanodine receptors triggering contraction. In Duchenne muscular dystrophy (DMD), a lethal disease that is caused by the lack of the cytoskeletal protein dystrophin, the cytosolic calcium concentration is known to be increased, and this increase may lead to cell necrosis. Here, we used myotubes derived from control and mdx mice, the murine model of DMD, to study the calcium responses induced by nicotinic acetylcholine receptor stimulation. The photoprotein aequorin was expressed in the cytosol or targeted to the plasma membrane as a fusion protein with the synaptosome-associated protein SNAP-25, thus allowing calcium measurements in a restricted area localized just below the plasma membrane. The carbachol-induced calcium responses were 4.5 times bigger in dystrophic myotubes than in control myotubes. Moreover, in dystrophic myotubes the carbachol-mediated calcium responses measured in the subsarcolemmal area were at least 10 times bigger than in the bulk cytosol. The initial calcium responses were due to calcium influx into the cells followed by a fast refilling/release phase from the sarcoplasmic reticulum. In addition and unexpectedly, the inositol 1,4,5-trisphosphate receptor pathway was involved in these calcium signals only in the dystrophic myotubes. This surprising involvement of this calcium release channel in the excitation-contraction coupling could open new ways for understanding exercise-induced calcium increases and downstream muscle degeneration in mdx mice and, therefore, in DMD.


Cell Membrane/metabolism , Egtazic Acid/analogs & derivatives , Inositol 1,4,5-Trisphosphate/physiology , Myocardium/metabolism , Receptors, Nicotinic/metabolism , Aequorin/chemistry , Animals , Calcium/chemistry , Calcium/metabolism , Calcium Channels/metabolism , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Cytosol/metabolism , Egtazic Acid/pharmacology , Green Fluorescent Proteins/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Muscle, Skeletal/metabolism , Necrosis , Nerve Tissue Proteins/metabolism , Phosphoproteins/chemistry , Plasmids/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcoplasmic Reticulum/metabolism , Synaptosomal-Associated Protein 25 , Time Factors , Transfection
19.
Dev Cell ; 4(5): 673-85, 2003 May.
Article En | MEDLINE | ID: mdl-12737803

The mechanisms that regulate the formation of multinucleated muscle fibers from mononucleated myoblasts are not well understood. We show here that extracellular matrix (ECM) receptors of the beta1 integrin family regulate myoblast fusion. beta1-deficient myoblasts adhere to each other, but plasma membrane breakdown is defective. The integrin-associated tetraspanin CD9 that regulates cell fusion is no longer expressed at the cell surface of beta1-deficient myoblasts, suggesting that beta1 integrins regulate the formation of a protein complex important for fusion. Subsequent to fusion, beta1 integrins are required for the assembly of sarcomeres. Other ECM receptors such as the dystrophin glycoprotein complex are still expressed but cannot compensate for the loss of beta1 integrins, providing evidence that different ECM receptors have nonredundant functions in skeletal muscle fibers.


Cell Fusion , Integrin beta1/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Sarcomeres/metabolism , Animals , Cell Death , Cell Division , Cell Movement , Cells, Cultured , Cytoskeleton/metabolism , Integrin beta1/genetics , Mice , Microscopy, Electron , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/ultrastructure , Myoblasts/ultrastructure
20.
Pflugers Arch ; 447(1): 14-22, 2003 Oct.
Article En | MEDLINE | ID: mdl-14976589

Under normal conditions in situ, muscle fibers and motoneurons, the main partners of motor units, are strongly dependent on each other. This interdependence hinders ex vivo studies of neuromuscular disorders where nervous or muscular components are considered separately. To allow in vitro access to complex nerve-muscle relationships, we developed a novel nerve-muscle co-culture system where mouse muscle innervation is assured by rat spinal cord explants. The degree of muscular maturation during co-culture was evaluated using the distribution of nicotinic acetylcholine receptors (AChRs) and their electrophysiological characteristics before and after innervation. In myotubes from non-innervated cultures, AChRs were diffusely distributed over the entire myotube surface. Their single-channel conductance (33.5+/-0.6 pS) and mean open time (8.1+/-0.7 ms) are characteristic of AChRs described in embryonic or denervated skeletal muscles. In innervated muscle fibers from co-cultures, AChRs appear as discrete aggregates and co-localize with synaptotagmin. In addition to the embryonic type currents, in innervated fibers AChR currents having high conductance (53.3+/-5.9 pS) and short mean open time (2.6+/-0.1 ms), characteristic of AChRs at mature neuromuscular junctions, were observed. Our data support the use of this new nerve-muscle co-culture system as a reliable model for the study of murine muscular differentiation and function.


Axons/physiology , Myoblasts, Skeletal/physiology , Receptors, Nicotinic/physiology , Action Potentials/physiology , Animals , Animals, Newborn , Cell Differentiation/physiology , Coculture Techniques , Mice , Mice, Inbred BALB C , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Myoblasts, Skeletal/cytology , Rats
...