Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 19(9)2018 Sep 18.
Article in English | MEDLINE | ID: mdl-30231482

ABSTRACT

Intracellular signaling pathways, including the mammalian target of rapamycin (mTOR) and the mitogen-activated protein kinase (MAPK) pathway, are activated by exercise, and promote skeletal muscle hypertrophy. However, the mechanisms by which these pathways are activated by physiological stimulation are not fully understood. Here we show that extracellular ATP activates these pathways by increasing intracellular Ca2+ levels ([Ca2+]i), and promotes muscle hypertrophy. [Ca2+]i in skeletal muscle was transiently increased after exercise. Treatment with ATP induced the increase in [Ca2+]i through the P2Y2 receptor/inositol 1,4,5-trisphosphate receptor pathway, and subsequent activation of mTOR in vitro. In addition, the ATP-induced increase in [Ca2+]i coordinately activated Erk1/2, p38 MAPK and mTOR that upregulated translation of JunB and interleukin-6. ATP also induced an increase in [Ca2+]i in isolated soleus muscle fibers, but not in extensor digitorum longus muscle fibers. Furthermore, administration of ATP led to muscle hypertrophy in an mTOR- and Ca2+-dependent manner in soleus, but not in plantaris muscle, suggesting that ATP specifically regulated [Ca2+]i in slow muscles. These findings suggest that ATP and [Ca2+]i are important mediators that convert mechanical stimulation into the activation of intracellular signaling pathways, and point to the P2Y receptor as a therapeutic target for treating muscle atrophy.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Muscle, Skeletal/pathology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Hypertrophy/metabolism , Hypertrophy/pathology , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology
2.
Biochem Biophys Res Commun ; 505(1): 51-59, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30236982

ABSTRACT

Duchenne muscular dystrophy (DMD) and the less severe Becker muscular dystrophy (BMD) are due to mutations in the DMD gene. Previous reports show that in-frame deletion of exons 45-55 produces an internally shorted, but functional, dystrophin protein resulting in a very mild BMD phenotype. In order to elucidate the molecular mechanism leading to this phenotype, we generated exon 45-55 deleted dystrophin transgenic/mdx (Tg/mdx) mice. Muscular function of Tg/mdx mice was restored close to that of wild type (WT) mice but the localization of the neuronal type of nitric oxide synthase was changed from the sarcolemma to the cytosol. This led to hyper-nitrosylation of the ryanodine receptor 1 causing increased Ca2+ release from the sarcoplasmic reticulum. On the other hand, Ca2+ reuptake by the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) was restored to the level of WT mice, suggesting that the Ca2+ dysregulation had been compensated by SERCA activation. In line with this, expression of sarcolipin (SLN), a SERCA-inhibitory peptide, was upregulated in mdx mice, but strongly reduced in Tg/mdx mice. Furthermore, knockdown of SLN ameliorated the cytosolic Ca2+ homeostasis and the dystrophic phenotype in mdx mice. These findings suggest that SLN may be a novel target for DMD therapy.


Subject(s)
Dystrophin/metabolism , Muscle Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Proteolipids/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Dystrophin/genetics , Humans , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Mice, Transgenic , Muscle Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Phenotype , Proteolipids/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Transgenes/genetics
3.
Chimia (Aarau) ; 72(4): 238-240, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29720316

ABSTRACT

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.


Subject(s)
Muscular Dystrophy, Duchenne/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Animals , Female , Humans , Male , Mice
4.
Am J Pathol ; 187(5): 1147-1161, 2017 May.
Article in English | MEDLINE | ID: mdl-28315675

ABSTRACT

Inflammatory events occurring in dystrophic muscles contribute to the progression of Duchenne muscular dystrophy (DMD). Low-intensity training (LIT) attenuates the phenotype of mdx mice, an animal model for DMD. Therefore, we postulated that LIT could have anti-inflammatory properties. We assessed levels of inflammatory cytokines and infiltrated immune cells in gastrocnemius muscle of mdx mice after LIT. We detected high levels of complement component C5a, chemokine ligand (CCL) 2, CD68+ monocytes/macrophages, and proinflammatory M1 macrophages in muscles of mdx mice. LIT decreased CCL2 levels, increased CD68+ cell numbers, and shifted the macrophage population to the regenerative M2 type. We investigated whether inhibition of C5a or CCL2 with L-aptamers could mimic the effects of LIT. Although no effect of CCL2 inhibition was detected, treatment with the C5a inhibitor, NOX-D21, rescued the phenotype of nonexercised mdx mice, but not of exercised ones. In both cases, the level of CD68+ cells increased and macrophage populations leaned toward the inflammatory M1 type. In muscles of nonexercised treated mice, the level of IL-1 receptor antagonist increased, damage decreased, and fibers were switched toward the glycolytic fast type; in muscles of exercised mice, fibers were switched to the oxidative slow type. These results reveal the effects of LIT on the inflammatory status of mdx mice and suggest that NOX-D21 could be an anti-inflammatory drug for DMD.


Subject(s)
Complement C5a/antagonists & inhibitors , Muscular Dystrophy, Animal/metabolism , Physical Conditioning, Animal/physiology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aptamers, Nucleotide/pharmacology , Chemokine CCL2/antagonists & inhibitors , Cytokines/metabolism , Disease Models, Animal , Energy Metabolism/physiology , Forelimb , Macrophages/physiology , Male , Mice, Inbred mdx , Muscle Strength/physiology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/physiopathology , Myositis/physiopathology , Myositis/prevention & control , Phenotype , Swimming/physiology
5.
Front Physiol ; 6: 254, 2015.
Article in English | MEDLINE | ID: mdl-26441673

ABSTRACT

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.

6.
J Neuromuscul Dis ; 2(4): 325-342, 2015 Nov 22.
Article in English | MEDLINE | ID: mdl-27858750

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is caused by mutations in the gene coding for dystrophin and leads to muscle degeneration, wheelchair dependence and death by cardiac or respiratory failure. Physical exercise has been proposed as a palliative therapy for DMD to maintain muscle strength and prevent contractures for as long as possible. However, its practice remains controversial because the benefits of training may be counteracted by muscle overuse and damage.The effects of physical exercise have been investigated in muscles of dystrophin-deficient mdx mice and in patients with DMD. However, a lack of uniformity among protocols limits comparability between studies and translatability of results from animals to humans. In the present review, we summarize and discuss published protocols used to investigate the effects of physical exercise on mdx mice and DMD patients, with the objectives of improving comparability between studies and identifying future research directions.

7.
PLoS One ; 9(10): e110708, 2014.
Article in English | MEDLINE | ID: mdl-25329652

ABSTRACT

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.


Subject(s)
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
8.
Curr Opin Neurol ; 26(5): 577-84, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995279

ABSTRACT

PURPOSE OF REVIEW: The most encouraging recent advances regarding pharmacological agents for treating Duchenne muscular dystrophy (DMD) are summarized. Emphasis is given to compounds acting downstream of dystrophin, the protein lacking in DMD, on cellular pathways leading to pathological consequences. The author highlights the progress that may have the greatest potential for clinical use in DMD. RECENT FINDINGS: Modifying the transcripts of the mutated gene by exon skipping has led to expression of shortened dystrophins in DMD patients. Currently, the most promising potential drugs are the exon-skipping agents eteplirsen and drisapersen. Biglycan and SMTC1100 upregulate utrophin. The steroid receptor modulating compounds VBP15 and tamoxifen, and specific antioxidants appear promising agents for symptomatic therapy. SUMMARY: The past 18 months have seen a strong increase in the number of exciting reports on novel therapeutic agents for DMD. Exon-skipping agents have been fine-tuned to improve tissue delivery and stability. Impressive discoveries regarding pathogenic events in cellular signalling have revealed targets that were unknown in the context of DMD, thus enabling approaches that limit inflammation, fibrosis and necrosis. The targets are nuclear hormone receptors, NADPH-oxidases and Ca channels. Inhibition of NF-KB, transforming growth factor-alpha (TGF-α) and transforming growth factor-beta (TGF-ß)/myostatin production or action are also promising routes in counteracting the complex pathogenesis of DMD.


Subject(s)
Dystrophin/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Animals , Dystrophin/genetics , Humans , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Mutation/genetics , Signal Transduction/physiology , Utrophin/genetics , Utrophin/metabolism
9.
Channels (Austin) ; 7(3): 221-4, 2013.
Article in English | MEDLINE | ID: mdl-23584166

ABSTRACT

Mechanical load-induced intracellular signaling events are important for subsequent skeletal muscle hypertrophy. We previously showed that load-induced activation of the cation channel TRPV1 caused an increase in intracellular calcium concentrations ([Ca ( 2+) ]i) and that this activated mammalian target of rapamycin (mTOR) and promoted muscle hypertrophy. However, the link between mechanical load-induced intracellular signaling events, and the TRPV1-mediated increases in [Ca ( 2+) ]i are not fully understood. Here we show that administration of the TRPV1 agonist, capsaicin, induces phosphorylation of mTOR, p70S6K, S6, Erk1/2 and p38 MAPK, but not Akt, AMPK or GSK3ß. Furthermore, the TRPV1-induced phosphorylation patterns resembled those induced by mechanical load. Our results continue to highlight the importance of TRPV1-mediated calcium signaling in load-induced intracellular signaling pathways.


Subject(s)
Calcium Signaling , Muscle, Skeletal/pathology , Nitric Oxide Synthase Type I/metabolism , Peroxynitrous Acid/metabolism , TRPV Cation Channels/metabolism , Animals , Male
10.
Am J Pathol ; 182(2): 485-504, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23332367

ABSTRACT

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.


Subject(s)
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
11.
Diab Vasc Dis Res ; 10(1): 65-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22621918

ABSTRACT

Fluoroquinolone antibiotics cause rare, but clinically important, adverse events including hyperglycaemia and hypoglycaemia. The present study focuses on the possible effect of levofloxacin and moxifloxacin on the cardiovascular functions of rats with type I diabetes. Both antibiotics caused bradycardia. Levofloxacin but not moxifloxacin caused hypoglycaemia in diabetic rats and an increase in amplitude of the ST segment revealed by electrocardiogram (ECG) analysis of isolated hearts. In pressurized mesenteric arteries, levofloxacin did not affect the endothelium-derived hyperpolarising factor (EDHF) pathway or its main components, the small-conductance Ca(2+) activated potassium (SK(Ca)) and intermediate-conductance Ca(2+) activated potassium (IK(Ca)) channels. In moxifloxacin-treated rats, an increase in the EDHF response was observed, which was largely attributed to SK(Ca)-activation. In conclusion, levofloxacin and moxifloxacin use appeared to vary but with no evidence of impairment of the cardiovascular function. However, it is still possible that these antibiotics may produce different effects if there are co-morbidities and therefore their use must be with care.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aza Compounds/pharmacology , Cardiovascular System/drug effects , Diabetes Mellitus, Experimental/physiopathology , Hypoglycemia/chemically induced , Levofloxacin , Ofloxacin/pharmacology , Quinolines/pharmacology , Animals , Biological Factors , Blood Glucose/drug effects , Bradycardia/chemically induced , Diabetes Mellitus, Experimental/chemically induced , Electrocardiography , Endothelium, Vascular/drug effects , Fluoroquinolones/pharmacology , Heart/drug effects , Heart Rate/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/drug effects , Male , Mesenteric Arteries/drug effects , Moxifloxacin , Rats , Rats, Wistar , Small-Conductance Calcium-Activated Potassium Channels/drug effects , Streptozocin
12.
Nat Med ; 19(1): 101-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202294

ABSTRACT

Skeletal muscle atrophy occurs in aging and pathological conditions, including cancer, diabetes and AIDS. Treatment of atrophy is based on either preventing protein-degradation pathways, which are activated during atrophy, or activating protein-synthesis pathways, which induce muscle hypertrophy. Here we show that neuronal nitric oxide synthase (nNOS) regulates load-induced hypertrophy by activating transient receptor potential cation channel, subfamily V, member 1 (TRPV1). The overload-induced hypertrophy was prevented in nNOS-null mice. nNOS was transiently activated within 3 min after overload. This activation promoted formation of peroxynitrite, a reaction product of nitric oxide with superoxide, which was derived from NADPH oxidase 4 (Nox4). Nitric oxide and peroxynitrite then activated Trpv1, resulting in an increase of intracellular Ca(2+) concentration ([Ca(2+)](i)) that subsequently triggered activation of mammalian target of rapamycin (mTOR). Notably, administration of the TRPV1 agonist capsaicin induced hypertrophy without overload and alleviated unloading- or denervation-induced atrophy. These findings identify nitric oxide, peroxynitrite and [Ca(2+)](i) as the crucial mediators that convert a mechanical load into an intracellular signaling pathway and lead us to suggest that TRPV1 could be a new therapeutic target for treating muscle atrophy.


Subject(s)
Calcium Signaling , Muscle, Skeletal/pathology , Nitric Oxide Synthase Type I/metabolism , Peroxynitrous Acid/metabolism , TRPV Cation Channels/metabolism , Animals , Antipruritics/pharmacology , Calcium/analysis , Calcium Signaling/drug effects , Capsaicin/pharmacology , Cell Line , Hindlimb Suspension , Hypertrophy/metabolism , Hypertrophy/pathology , Ion Transport/drug effects , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects
13.
J Biol Chem ; 287(18): 14524-34, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22399301

ABSTRACT

We previously showed in vitro that calcium entry through Trpc1 ion channels regulates myoblast migration and differentiation. In the present work, we used primary cell cultures and isolated muscles from Trpc1(-/-) and Trpc1(+/+) murine model to investigate the role of Trpc1 in myoblast differentiation and in muscle regeneration. In these models, we studied regeneration consecutive to cardiotoxin-induced muscle injury and observed a significant hypotrophy and a delayed regeneration in Trpc1(-/-) muscles consisting in smaller fiber size and increased proportion of centrally nucleated fibers. This was accompanied by a decreased expression of myogenic factors such as MyoD, Myf5, and myogenin and of one of their targets, the developmental MHC (MHCd). Consequently, muscle tension was systematically lower in muscles from Trpc1(-/-) mice. Importantly, the PI3K/Akt/mTOR/p70S6K pathway, which plays a crucial role in muscle growth and regeneration, was down-regulated in regenerating Trpc1(-/-) muscles. Indeed, phosphorylation of both Akt and p70S6K proteins was decreased as well as the activation of PI3K, the main upstream regulator of the Akt. This effect was independent of insulin-like growth factor expression. Akt phosphorylation also was reduced in Trpc1(-/-) primary myoblasts and in control myoblasts differentiated in the absence of extracellular Ca(2+) or pretreated with EGTA-AM or wortmannin, suggesting that the entry of Ca(2+) through Trpc1 channels enhanced the activity of PI3K. Our results emphasize the involvement of Trpc1 channels in skeletal muscle development in vitro and in vivo, and identify a Ca(2+)-dependent activation of the PI3K/Akt/mTOR/p70S6K pathway during myoblast differentiation and muscle regeneration.


Subject(s)
Cell Differentiation/physiology , Myoblasts, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/physiology , Signal Transduction/physiology , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Enzyme Activation/physiology , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myoblasts, Skeletal/cytology , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , TRPC Cation Channels/genetics
14.
Histochem Cell Biol ; 137(6): 811-27, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22331205

ABSTRACT

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.


Subject(s)
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
15.
Am J Pathol ; 180(2): 749-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22192627

ABSTRACT

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.


Subject(s)
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
16.
J Pineal Res ; 51(2): 163-71, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21486366

ABSTRACT

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.


Subject(s)
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
17.
Biochem Biophys Res Commun ; 391(1): 401-6, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19914213

ABSTRACT

Duchenne muscular dystrophy is an inherited disease caused by the absence of dystrophin, a structural protein normally located under the sarcolemma of skeletal muscle fibers. Muscle degeneration occurring in this disease is thought to be partly caused by increased Ca(2+) entry through sarcolemmal cationic channels. Using the Mn(2+) quench method, we show here that Mn(2+) entry triggered by Ca(2+) store depletion but not basal Mn(2+) entry relies on Ca(2+)-independent PLA(2) (iPLA(2)) activity in dystrophic fibers isolated from a murine model of Duchenne muscular dystrophy, the mdx(5cv) mouse. iPLA(2) was found to be localized in the vicinity of the sarcolemma and consistently, the iPLA(2) lipid product lysophosphatidylcholine was found to trigger Ca(2+) entry through sarcolemmal channels, suggesting that it acts as an intracellular messenger responsible for store-operated channels opening in dystrophic fibers. Our results suggest that inhibition of iPLA(2) and lysophospholipid production may be of interest to reduce Ca(2+) entry and subsequent degeneration of dystrophic muscle.


Subject(s)
Calcium/metabolism , Lysophosphatidylcholines/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Phospholipases A2, Calcium-Independent/metabolism , Animals , Arachidonic Acid/pharmacology , Lysophosphatidylcholines/pharmacology , Manganese/metabolism , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Phospholipases A2, Calcium-Independent/antagonists & inhibitors , Sarcolemma/enzymology
18.
J Pineal Res ; 47(3): 238-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19664004

ABSTRACT

Oxidative stress-induced mitochondrial dysfunction plays a crucial role in the pathogenesis of a wide range of diseases including muscle disorders. In this study, we demonstrate that melatonin readily rescued mitochondria from oxidative stress-induced dysfunction and effectively prevented subsequent apoptosis of primary muscle cultures prepared from C57BL/6J mice. In particular, melatonin (10(-4)-10(-6) m) fully prevented myotube death induced by tert-butylhydroperoxide (t-BHP; 10 microm-24 hr) as assessed by acid phosphatase, caspase-3 activities and cellular morphological changes. Using fluorescence imaging, we showed that the mitochondrial protection provided by melatonin was associated with an inhibition of t-BHP-induced reactive oxygen species generation. In line with this observation, melatonin prevented t-BHP-induced mitochondrial depolarization and mitochondrial permeability transition pore (PTP) opening. This was associated with a highly reduced environment as reflected by an increased glutathione content and an increased ability to maintain mitochondrial pyridine nucleotides and glutathione in a reduced state. Using isolated mitochondria, in a similar manner as cyclosporin A, melatonin (10(-8)-10(-6) m) desensitized the PTP to Ca(2+) and prevented t-BHP-induced mitochondrial swelling, pyridine nucleotide and glutathione oxidation. In conclusion, our findings suggest that inhibition of the PTP essentially contributes to the protective effect of melatonin against oxidative stress in myotubes.


Subject(s)
Central Nervous System Depressants/pharmacology , Melatonin/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Muscle Cells , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Reactive Oxygen Species , tert-Butylhydroperoxide/pharmacology
19.
Biofactors ; 35(3): 279-94, 2009.
Article in English | MEDLINE | ID: mdl-19322813

ABSTRACT

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.


Subject(s)
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
20.
Mol Ther ; 17(1): 19-25, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19002166

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...