Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Sci Rep ; 10(1): 1125, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980663

ABSTRACT

Arising from the ablation of the cytoskeletal protein dystrophin, Duchenne Muscular Dystrophy (DMD) is a debilitating and fatal skeletal muscle wasting disease underpinned by metabolic insufficiency. The inability to facilitate adequate energy production may impede calcium (Ca2+) buffering within, and the regenerative capacity of, dystrophic muscle. Therefore, increasing the metabogenic potential could represent an effective treatment avenue. The aim of our study was to determine the efficacy of adenylosuccinic acid (ASA), a purine nucleotide cycle metabolite, to stimulate metabolism and buffer skeletal muscle damage in the mdx mouse model of DMD. Dystrophin-positive control (C57BL/10) and dystrophin-deficient mdx mice were treated with ASA (3000 µg.mL-1) in drinking water. Following the 8-week treatment period, metabolism, mitochondrial density, viability and superoxide (O2-) production, as well as skeletal muscle histopathology, were assessed. ASA treatment significantly improved the histopathological features of murine DMD by reducing damage area, the number of centronucleated fibres, lipid accumulation, connective tissue infiltration and Ca2+ content of mdx tibialis anterior. These effects were independent of upregulated utrophin expression in the tibialis anterior. ASA treatment also increased mitochondrial viability in mdx flexor digitorum brevis fibres and concomitantly reduced O2- production, an effect that was also observed in cultured immortalised human DMD myoblasts. Our data indicates that ASA has a protective effect on mdx skeletal muscles.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Muscular Dystrophy, Animal/drug therapy , Adenosine Monophosphate/therapeutic use , Animals , Calcium/analysis , Cell Line, Transformed , Collagen/analysis , Drug Evaluation, Preclinical , Electron Transport/drug effects , Humans , Lipids/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mitochondria, Muscle/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/pathology , Myoblasts/metabolism , Organelle Biogenesis , Oxygen Consumption/drug effects , Superoxides/metabolism , Utrophin/biosynthesis , Utrophin/genetics
2.
Photochem Photobiol ; 96(1): 200-207, 2020 01.
Article in English | MEDLINE | ID: mdl-31733143

ABSTRACT

This study analyzed photobiomodulation therapy (PBMT) effects on regenerative, antioxidative, anti-inflammatory and angiogenic markers in the dystrophic skeletal muscle of mdx mice, the experimental model of Duchenne muscular dystrophy (DMD), during the acute phase of dystrophy disease. The following groups were set up: Ctrl (control group of normal wild-type mice; C57BL/10); mdx (untreated mdx mice); mdxPred (mdx mice treated with prednisolone) and mdxLA (mdx mice treated with PBMT). The PBMT was carried out using an Aluminum Gallium Arsenide (AIGaAs; IBRAMED® laserpulse) diode, 830 nm wavelength, applied on the dystrophic quadriceps muscle. The mdxLA group showed a degenerative and regenerative area reduction simultaneously with a MyoD level increase, ROS production and inflammatory marker reduction and up-regulation in the VEGF factor. In addition, PBMT presented similar effects to prednisolone treatment in most of the parameters analyzed. In conclusion, our results indicate that PBMT in the parameters selected attenuated the dystrophic phenotype of mdx mice, improving skeletal muscle regeneration; reducing the oxidative stress and inflammatory process; and up-regulating the angiogenic marker.


Subject(s)
Low-Level Light Therapy , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Phenotype , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Sci Rep ; 9(1): 12982, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506484

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked genetic disease characterized by progressive muscle wasting and weakness and premature death. Glucocorticoids (e.g. prednisolone) remain the only drugs with a favorable impact on DMD patients, but not without side effects. We have demonstrated that glycine preserves muscle in various wasting models. Since glycine effectively suppresses the activity of pro-inflammatory macrophages, we investigated the potential of glycine treatment to ameliorate the dystrophic pathology. Dystrophic mdx and dystrophin-utrophin null (dko) mice were treated with glycine or L-alanine (amino acid control) for up to 15 weeks and voluntary running distance (a quality of life marker and strong correlate of lifespan in dko mice) and muscle morphology were assessed. Glycine increased voluntary running distance in mdx mice by 90% (P < 0.05) after 2 weeks and by 60% (P < 0.01) in dko mice co-treated with prednisolone over an 8 week treatment period. Glycine treatment attenuated fibrotic deposition in the diaphragm by 28% (P < 0.05) after 10 weeks in mdx mice and by 22% (P < 0.02) after 14 weeks in dko mice. Glycine treatment augmented the prednisolone-induced reduction in fibrosis in diaphragm muscles of dko mice (23%, P < 0.05) after 8 weeks. Our findings provide strong evidence that glycine supplementation may be a safe, simple and effective adjuvant for improving the efficacy of prednisolone treatment and improving the quality of life for DMD patients.


Subject(s)
Disease Models, Animal , Glycine Agents/administration & dosage , Glycine/administration & dosage , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Prednisolone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology
4.
PLoS One ; 12(11): e0187317, 2017.
Article in English | MEDLINE | ID: mdl-29095865

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is a fatal muscle wasting disease manifested in young boys, for which there is no current cure. We have shown that the amino acid taurine is safe and effective at preventing dystropathology in the mdx mouse model for DMD. This study aimed to establish if treating growing mdx mice with a higher dose of taurine was more effective at improving strength and reducing inflammation and oxidative stress. Mice were treated with a dose of taurine estimated to be 16 g/kg/day, in drinking water from 1-6 weeks of age, after which in vivo and ex vivo muscle strength was assessed, as were measures of inflammation, oxidative stress and taurine metabolism. While the dose did decrease inflammation and protein oxidation in dystrophic muscles, there was no improvement in muscle strength (in contrast with benefits observed with the lower dose) and growth of the young mice was significantly restricted. We present novel data that a high taurine dose increases the cysteine content of both mdx liver and plasma, a possible result of down regulation of the taurine synthesis pathway in the liver (which functions to dispose of excess cysteine, which is toxic). These data caution that a high dose of taurine can have adverse effects and may be less efficacious than lower taurine doses. Therefore, monitoring of taurine dosage needs to be considered in future pre-clinical trials, in anticipation of using taurine as a clinical therapy for growing DMD boys (and other conditions).


Subject(s)
Growth , Muscular Dystrophy, Animal/drug therapy , Taurine/therapeutic use , Animals , Mice , Mice, Inbred mdx , Muscular Dystrophy, Animal/pathology , Oxidative Stress/drug effects
5.
Pol J Vet Sci ; 20(2): 387-394, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28865216

ABSTRACT

The aim of this study was to explain the correlations between selenium deficiency, hemostatic and biochemical disorders, and the progression of pathological changes in calves diagnosed with nutritional muscular dystrophy (NMD). The study was performed on 20 calves with supplementation of 8 ml selenium and vitamin E preparation and 20 calves with symptoms of NMD. Blood was sampled from calves aged 5, 12 and 19 days. On day 19, samples of the biceps femoris muscle were collected from 6 animals in each group for histopathological analysis. The following blood parameters were determined: PLT, PT, TT, APTT, fibrinogen and D-dimer concentrations, antithrombin III activity, glucose, selenium and vitamin E concentrations, activity of CK, LDH and GSH-Px. Muscle sections were stained with H&E and HBFP. Platelet counts were significantly lower in calves with symptoms of NMD. No significant differences in coagulation parameters were observed between the groups. Sick calves were diagnosed with hyperglycemia and elevation of CK and LDH activity. Selenium and vitamin E concentrations in the blood serum were significantly lower in the experimental group together with significant drop in GSH-Px activity. Changes characteristic of Zenker's necrosis were observed in a muscle of the sick animals. To our best knowledge this is the first study in which the attempt was made to explain the relationship between selenium deficiency and changes in the coagulation system in ruminants.


Subject(s)
Blood Coagulation Disorders/veterinary , Cattle Diseases/blood , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/etiology , Nutrition Disorders/veterinary , Selenium/deficiency , Animals , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/pathology , Cattle , Cattle Diseases/pathology , Glutathione Peroxidase/metabolism , Muscular Dystrophy, Animal/blood , Muscular Dystrophy, Animal/pathology , Nutrition Disorders/blood , Nutrition Disorders/etiology , Nutrition Disorders/pathology , Vitamin E/metabolism
6.
BMC Musculoskelet Disord ; 18(1): 20, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28103859

ABSTRACT

BACKGROUND: Inhibition of activin/myostatin pathway has emerged as a novel approach to increase muscle mass and bone strength. Duchenne muscular dystrophy (DMD) is a neuromuscular disorder that leads to progressive muscle degeneration and also high incidence of fractures. The aim of our study was to test whether inhibition of activin receptor IIB ligands with or without exercise could improve bone strength in the mdx mouse model for DMD. METHODS: Thirty-two mdx mice were divided to running and non-running groups and to receive either PBS control or soluble activin type IIB-receptor (ActRIIB-Fc) once weekly for 7 weeks. RESULTS: Treatment of mdx mice with ActRIIB-Fc resulted in significantly increased body and muscle weights in both sedentary and exercising mice. Femoral µCT analysis showed increased bone volume and trabecular number (BV/TV +80%, Tb.N +70%, P < 0.05) in both ActRIIB-Fc treated groups. Running also resulted in increased bone volume and trabecular number in PBS-treated mice. However, there was no significant difference in trabecular bone structure or volumetric bone mineral density between the ActRIIB-Fc and ActRIIB-Fc-R indicating that running did not further improve bone structure in ActRIIB-Fc-treated mice. ActRIIB-Fc increased bone mass also in vertebrae (BV/TV +20%, Tb.N +30%, P < 0.05) but the effects were more modest. The number of osteoclasts was decreased in histological analysis and the expression of several osteoblast marker genes was increased in ActRIIB-Fc treated mice suggesting decreased bone resorption and increased bone formation in these mice. Increased bone mass in femurs translated into enhanced bone strength in biomechanical testing as the maximum force and stiffness were significantly elevated in ActRIIB-Fc-treated mice. CONCLUSIONS: Our results indicate that treatment of mdx mice with the soluble ActRIIB-Fc results in a robust increase in bone mass, without any additive effect by voluntary running. Thus ActRIIB-Fc could be an attractive option in the treatment of musculoskeletal disorders.


Subject(s)
Activin Receptors, Type II/therapeutic use , Bone Density/drug effects , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne , Animals , Bone Resorption/pathology , Bone Resorption/prevention & control , Bone and Bones/drug effects , Bone and Bones/pathology , Combined Modality Therapy , Disease Models, Animal , Drug Evaluation, Preclinical , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Organ Size/drug effects , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Physical Conditioning, Animal , Solubility
7.
PLoS One ; 11(12): e0168293, 2016.
Article in English | MEDLINE | ID: mdl-27977770

ABSTRACT

Duchenne muscular dystrophy (DMD) results from a genetic lesion in the dystrophin gene and leads to progressive muscle damage. PGC-1α pathway activation improves muscle function and decreases histopathological injury. We hypothesized that mild disease found in the limb muscles of mdx mice may be responsive to quercetin-mediated protection of dystrophic muscle via PGC-1α pathway activation. To test this hypothesis muscle function was measured in the soleus and EDL from 14 month old C57, mdx, and mdx mice treated with quercetin (mdxQ; 0.2% dietary enrichment) for 12 months. Quercetin reversed 50% of disease-related losses in specific tension and partially preserved fatigue resistance in the soleus. Specific tension and resistance to contraction-induced injury in the EDL were not protected by quercetin. Given some functional gain in the soleus it was probed with histological and biochemical approaches, however, in dystrophic muscle histopathological outcomes were not improved by quercetin and suppressed PGC-1α pathway activation was not increased. Similar to results in the diaphragm from these mice, these data suggest that the benefits conferred to dystrophic muscle following 12 months of quercetin enrichment were underwhelming. Spontaneous activity at the end of the treatment period was greater in mdxQ compared to mdx indicating that quercetin fed mice were more active in addition to engaging in more vigorous activity. Hence, modest preservation of muscle function (specific tension) and elevated spontaneous physical activity largely in the absence of tissue damage in mdxQ suggests dietary quercetin may mediate protection.


Subject(s)
Cytoprotection/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/pathology , Quercetin/administration & dosage , Quercetin/pharmacology , Animals , Dietary Supplements , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Time Factors
8.
Amino Acids ; 48(11): 2635-2645, 2016 11.
Article in English | MEDLINE | ID: mdl-27444300

ABSTRACT

This study investigated the effect of taurine and ß-alanine supplementation on muscle function and muscle taurine transporter (TauT) protein expression in mdx mice. Wild-type (WT) and mdx mice (5 months) were supplemented with taurine or ß-alanine for 4 weeks, after which in vitro contractile properties, fatigue resistance and force recovery, and the expression of the TauT protein and proteins involved in excitation-contraction (E-C) coupling were examined in fast-twitch muscle. There was no difference in basal TauT protein expression or basal taurine content between mdx than WT muscle. Supplementation with taurine and ß-alanine increased and reduced taurine content, respectively, in muscle from WT and mdx mice but had no effect of TauT protein. Taurine supplementation reduced body and muscle mass, and enhanced fatigue resistance and force recovery in mdx muscle. ß-Alanine supplementation enhanced fatigue resistance in WT and mdx muscle. There was no difference in the basal expression of key E-C coupling proteins [ryanodine receptor 1 (RyR1), dihydropyridine receptor (DHPR), sarco(endo)plasmic reticulum Ca2+-ATPase 1 (SERCA1) or calsequestrin 1 (CSQ1)] between WT and mdx mice, and the expression of these proteins was not altered by taurine or ß-alanine supplementation. These findings suggest that TauT protein expression is relatively insensitive to changes in muscle taurine content in WT and mdx mice, and that taurine and ß-alanine supplementation may be viable therapeutic strategies to improve fatigue resistance of dystrophic skeletal muscle.


Subject(s)
Fatigue/metabolism , Gene Expression Regulation/drug effects , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins/biosynthesis , Muscle, Skeletal/metabolism , Taurine/pharmacology , beta-Alanine/pharmacology , Animals , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology
9.
Exp Physiol ; 100(1): 12-22, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25557727

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does dietary quercetin enrichment improve biochemical and histological outcomes in hearts from mdx mice? What is the main finding and what is its importance? Biochemical and histological findings suggest that chronic quercetin feeding of mdx mice may improve mitochondrial function and attenuate tissue pathology. Patients with Duchenne muscular dystrophy suffer from cardiac pathology, which causes up to 40% of all deaths because of fibrosis and cardiac complications. Quercetin is a flavonol with anti-inflammatory and antioxidant effects and is also an activator of peroxisome proliferator-activated receptor γ coactivator 1α capable of antioxidant upregulation, mitochondrial biogenesis and prevention of cardiac complications. We sought to determine the extent to which dietary quercetin enrichment prevents (experiment 1) and rescues cardiac pathology (experiment 2) in mdx mice. In experiment 1, 3-week-old mdx mice were fed control chow (C3w6m, n = 10) or chow containing 0.2% quercetin for 6 months (Q3w6m, n = 10). In experiment 2, 3-month-old mdx mice were fed control chow (C3m6m, n = 10) or 0.2% chow containing 0.2% quercetin for 6 months (Q3m6m, n = 10). Hearts were excised for histological and biochemical analyses. In experiment 1, Western blot targets for mitochondrial biogenesis (cytochrome c, P = 0.007) and antioxidant expression (superoxide dismutase 2, P = 0.014) increased in Q3w6m mice compared with C3w6m. Histology revealed increased utrophin (P = 0.025) and decreased matrix metalloproteinase 9 abundance (P = 0.040) in Q3w6m mice compared with C3w6m. In experiment 2, relative (P = 0.023) and absolute heart weights (P = 0.020) decreased in Q3m6m mice compared with C3m6m. Indications of damage (Haematoxylin- and Eosin-stained sections, P = 0.007) and Western blot analysis of transforming growth factor ß1 (P = 0.009) were decreased in Q3m6m mice. Six months of quercetin feeding increased a mitochondrial biomarker, antioxidant protein and utrophin and decreased matrix metalloproteinase 9 in young mice. Given that these adaptations are associated with attenuated cardiac pathology and damage, the present findings may indicate that dietary quercetin enrichment attenuates dystrophic cardiac pathology, but physiological confirmation is needed.


Subject(s)
Cardiomyopathies/drug therapy , Cardiomyopathies/prevention & control , Dietary Supplements , Mitochondria, Heart/drug effects , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Myocardium/pathology , Quercetin/pharmacology , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cytochromes c/metabolism , Cytoprotection , Disease Models, Animal , Matrix Metalloproteinase 9/metabolism , Mice, Inbred mdx , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Turnover/drug effects , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocardium/metabolism , Superoxide Dismutase/metabolism , Time Factors , Transforming Growth Factor beta1/metabolism , Utrophin/metabolism
10.
Hum Mol Genet ; 23(17): 4651-62, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24760771

ABSTRACT

Duchenne muscular dystrophy (DMD) is a common and relentlessly progressive muscle disease. Some interventions have been identified that modestly slow progression and prolong survival, but more meaningful therapies are lacking. The goal of this study is to identify new therapeutic pathways for DMD using a zebrafish model of the disease. To accomplish this, we performed a non-biased drug screen in sapje, a zebrafish line with a recessive nonsense mutation in dystrophin. We identified 6 positive hits (out of 640 total drugs tested) by their ability to prevent abnormal birefringence in sapje. Follow-up analyses demonstrated that fluoxetine, a selective serotonin reuptake inhibitor (SSRI), provided the most substantial benefit. Morpholino-based experimentation confirmed that modulation of the serotonin pathway alone can prevent the dystrophic phenotype, and transcriptomic analysis revealed changes in calcium homeostasis as a potential mechanism. In all, we demonstrate that monoamine agonists can prevent disease in a vertebrate model of DMD. Given the safe and widespread use of SSRIs in clinical practice, our study identifies an attractive target pathway for therapy development.


Subject(s)
Fluoxetine/therapeutic use , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Zebrafish/physiology , Animals , Base Sequence , Birefringence , Calcium/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Dystrophin/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Evans Blue/metabolism , Fluoxetine/pharmacology , Gene Expression Profiling , Gene Knockdown Techniques , Homeostasis/drug effects , Molecular Sequence Data , Morpholinos/pharmacology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Oligonucleotide Array Sequence Analysis , Phenotype , Serotonin Plasma Membrane Transport Proteins/metabolism , Stress, Mechanical , Survival Analysis , Zebrafish/embryology , Zebrafish/genetics
11.
PLoS One ; 8(7): e66617, 2013.
Article in English | MEDLINE | ID: mdl-23843959

ABSTRACT

BACKGROUND: In Duchenne muscular dystrophy (DMD), loss of the membrane stabilizing protein dystrophin results in myofiber damage. Microinjury to dystrophic myofibers also causes secondary imbalances in sarcolemmic ion permeability and resting membrane potential, which modifies excitation-contraction coupling and increases proinflammatory/apoptotic signaling cascades. Although glucocorticoids remain the standard of care for the treatment of DMD, there is a need to investigate the efficacy of other pharmacological agents targeting the involvement of imbalances in ion flux on dystrophic pathology. METHODOLOGY/PRINCIPAL FINDINGS: We designed a preclinical trial to investigate the effects of lansoprazole (LANZO) administration, a proton pump inhibitor, on the dystrophic muscle phenotype in dystrophin deficient (mdx) mice. Eight to ten week-old female mice were assigned to one of four treatment groups (n = 12 per group): (1) vehicle control; (2) 5 mg/kg/day LANZO; (3) 5 mg/kg/day prednisolone; and (4) combined treatment of 5 mg/kg/day prednisolone (PRED) and 5 mg/kg/day LANZO. Treatment was administered orally 5 d/wk for 3 months. At the end of the study, behavioral (Digiscan) and functional outcomes (grip strength and Rotarod) were assessed prior to sacrifice. After sacrifice, body, tissue and organ masses, muscle histology, in vitro muscle force, and creatine kinase levels were measured. Mice in the combined treatment groups displayed significant reductions in the number of degenerating muscle fibers and number of inflammatory foci per muscle field relative to vehicle control. Additionally, mice in the combined treatment group displayed less of a decline in normalized forelimb and hindlimb grip strength and declines in in vitro EDL force after repeated eccentric contractions. CONCLUSIONS/SIGNIFICANCE: Together our findings suggest that combined treatment of LANZO and prednisolone attenuates some components of dystrophic pathology in mdx mice. Our findings warrant future investigation of the clinical efficacy of LANZO and prednisolone combined treatment regimens in dystrophic pathology.


Subject(s)
Dystrophin/genetics , Lansoprazole/pharmacology , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/drug therapy , Proton Pump Inhibitors/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Synergism , Dystrophin/deficiency , Female , Gene Expression , Glucocorticoids/pharmacology , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Prednisolone/pharmacology
12.
PLoS One ; 8(3): e54922, 2013.
Article in English | MEDLINE | ID: mdl-23526927

ABSTRACT

Cellular therapy is a potential approach to improve the regenerative capacity of damaged or diseased skeletal muscle. However, its clinical use has often been limited by impaired donor cell survival, proliferation and differentiation following transplantation. Additionally, functional improvements after transplantation are all-too-often negligible. Because the host microenvironment plays an important role in the fate of transplanted cells, methods to modulate the microenvironment and guide donor cell behavior are warranted. The purpose of this study was to investigate whether the use of neuromuscular electrical stimulation (NMES) for 1 or 4 weeks following muscle-derived stem cell (MDSC) transplantation into dystrophic skeletal muscle can modulate the fate of donor cells and enhance their contribution to muscle regeneration and functional improvements. Animals submitted to 4 weeks of NMES after transplantation demonstrated a 2-fold increase in the number of dystrophin+ myofibers as compared to control transplanted muscles. These findings were concomitant with an increased vascularity in the MDSC+NMES group when compared to non-stimulated counterparts. Additionally, animals subjected to NMES (with or without MDSC transplantation) presented an increased maximal specific tetanic force when compared to controls. Although cell transplantation and/or the use of NMES resulted in no changes in fatigue resistance, the combination of both MDSC transplantation and NMES resulted in a faster recovery from fatigue, when compared to non-injected and non-stimulated counterparts. We conclude that NMES is a viable method to improve MDSC engraftment, enhance dystrophic muscle strength, and, in combination with MDSC transplantation, improve recovery from fatigue. These findings suggest that NMES may be a clinically-relevant adjunct approach for cell transplantation into skeletal muscle.


Subject(s)
Electric Stimulation Therapy/methods , Muscular Dystrophy, Animal/therapy , Myoblasts, Skeletal/transplantation , Animals , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Development , Muscle Strength , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Neuromuscular Junction/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Regeneration , Stem Cell Niche
13.
PLoS One ; 5(5): e10763, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20505827

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common, lethal disease of childhood. One of 3500 new-born males suffers from this universally-lethal disease. Other than the use of corticosteroids, little is available to affect the relentless progress of the disease, leading many families to use dietary supplements in hopes of reducing the progression or severity of muscle wasting. Arginine is commonly used as a dietary supplement and its use has been reported to have beneficial effects following short-term administration to mdx mice, a genetic model of DMD. However, the long-term effects of arginine supplementation are unknown. This lack of knowledge about the long-term effects of increased arginine metabolism is important because elevated arginine metabolism can increase tissue fibrosis, and increased fibrosis of skeletal muscles and the heart is an important and potentially life-threatening feature of DMD. METHODOLOGY: We use both genetic and nutritional manipulations to test whether changes in arginase metabolism promote fibrosis and increase pathology in mdx mice. Our findings show that fibrotic lesions in mdx muscle are enriched with arginase-2-expressing macrophages and that muscle macrophages stimulated with cytokines that activate the M2 phenotype show elevated arginase activity and expression. We generated a line of arginase-2-null mutant mdx mice and found that the mutation reduced fibrosis in muscles of 18-month-old mdx mice, and reduced kyphosis that is attributable to muscle fibrosis. We also observed that dietary supplementation with arginine for 17-months increased mdx muscle fibrosis. In contrast, arginine-2 mutation did not reduce cardiac fibrosis or affect cardiac function assessed by echocardiography, although 17-months of dietary supplementation with arginine increased cardiac fibrosis. Long-term arginine treatments did not decrease matrix metalloproteinase-2 or -9 or increase the expression of utrophin, which have been reported as beneficial effects of short-term treatments. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that arginine metabolism by arginase promotes fibrosis of muscle in muscular dystrophy and contributes to kyphosis. Our findings also show that long-term, dietary supplementation with arginine exacerbates fibrosis of dystrophic heart and muscles. Thus, commonly-practiced dietary supplementation with arginine by DMD patients has potential risk for increasing pathology when performed for long periods, despite reports of benefits acquired with short-term supplementation.


Subject(s)
Arginine/metabolism , Macrophages/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Myocardium/metabolism , Myocardium/pathology , Animals , Arginase/metabolism , Arginine/administration & dosage , Arginine/pharmacology , Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/pathology , Cytokines/metabolism , Dystrophin/deficiency , Dystrophin/metabolism , Fibrosis , Gene Deletion , Inflammation/complications , Inflammation/enzymology , Inflammation/pathology , Kyphosis/complications , Kyphosis/enzymology , Kyphosis/pathology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/pathology , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/enzymology , Muscular Dystrophy, Animal/complications , Muscular Dystrophy, Animal/enzymology , Nitric Oxide Synthase Type I/metabolism , Protein Transport/drug effects , Th2 Cells/drug effects
14.
Mol Cells ; 25(4): 531-7, 2008 Jun 30.
Article in English | MEDLINE | ID: mdl-18460899

ABSTRACT

Abnormal activation of nuclear factor kappa B (NF-kappaB) probably plays an important role in the pathogenesis of Duchenne's muscular dystrophy (DMD). In this report, we evaluated the efficacy of curcumin, a potent NF-kappaB inhibitor, in mdx mice, a mouse model of DMD. We found that it improved sarcolemmic integrity and enhanced muscle strength after intraperitoneal (i.p.) injection. Histological analysis revealed that the structural defects of myofibrils were reduced, and biochemical analysis showed that creatine kinase (CK) activity was decreased. We also found that levels of tumor necrosis factor alpha (TNF-alpha), interleukin-1 beta (IL-1beta) and inducible nitric oxide synthase (iNOS) in the mdx mice were decreased by curcumin administration. EMSA analysis showed that NF-kappaB activity was also inhibited. We thus conclude that curcumin is effective in the therapy of muscular dystrophy in mdx mice, and that the mechanism may involve inhibition of NF-kappaB activity. Since curcumin is a non-toxic compound derived from plants, we propose that it may be useful for DMD therapy.


Subject(s)
Muscle Strength/drug effects , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/drug therapy , NF-kappa B/antagonists & inhibitors , Phytotherapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Creatine Kinase/antagonists & inhibitors , Creatine Kinase/blood , Curcumin/administration & dosage , Humans , Injections, Intraperitoneal , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/blood , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Strength/physiology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/drug therapy , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood
15.
Biochem Pharmacol ; 56(6): 751-7, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9751080

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by dystrophin deficiency, which results in muscle necrosis and the upregulation of heat shock/stress proteins (HSP). We hypothesized that reactive oxygen species, and in particular hydroxyl radicals (.OH), participate in muscle necrosis and HSP expression. It was assumed that iron deprivation decreases .OH generation, restraining the disease process and reducing the oxidant-induced expression of HSP. The role of iron-catalyzed free radical reactions in the pathology of dystrophin-deficient muscle was evaluated in the murine model for Duchenne muscular dystrophy (mdx), by examining the effects of dietary deficiency and supplementation of iron on serum creatine kinase (CK), muscle morphology, lipid peroxidation and HSP levels in mice maintained on diets deficient in or supplemented with iron for 6 weeks. Iron-deprived mdx mice showed a significant decrease in the number of macrophage-invaded necrotic fibers and the expression of the 70-kDa heat shock protein (Hsp70). This suggests that the iron-dependent generation of .OH relates to muscle necrosis in the mdx mouse and modulates the expression of Hsp70 in vivo. In contrast, iron deprivation had no influence on other HSP or on lipid peroxidation in mdx mice, while maintenance on either diet caused a significant decrease in serum creatine kinase activity. The potential therapeutic effects of iron deprivation in mdx should be considered.


Subject(s)
Heat-Shock Proteins/biosynthesis , Iron Deficiencies , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Animals , Creatine Kinase/blood , Dietary Supplements , Disease Models, Animal , Genetic Linkage , Iron/blood , Iron, Dietary/administration & dosage , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Necrosis , X Chromosome
16.
Pediatr Res ; 40(3): 444-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8865282

ABSTRACT

A simple, reproducible test was used to quantify muscle weakness in mdx mice, an animal model of Duchenne muscular dystrophy. The effect of bedding on wheat kernels and of dietary supplementation of alpha-tocopherol on the progression of muscle weakness was investigated in mdx mice. When measured during the first 200 d of life, mdx mice developed muscle weakness, irrespective of bedding and diet. When kept on wood shavings and fed a conventional rodent diet, mdx mice showed progressive muscle weakness over the consecutive 200 d, and eventually showed a significant weight loss during the next 200-d observation period. Progression of muscle weakness and weight loss were almost completely prevented in mdx mice that were kept on wheat kernel bedding. In contrast, only incomplete maintenance of muscle strength and body weight was observed in mdx mice kept on wood shavings and fed the alpha-tocopherol-supplemented diet. It is concluded from these experiments that a component of wheat kernels other than alpha-tocopherol is essential to prevent the progression of muscle weakness in mdx mice.


Subject(s)
Aging/pathology , Muscle Weakness/prevention & control , Muscular Dystrophy, Animal/diet therapy , Seeds , Triticum , Vitamin E/therapeutic use , Animals , Biomarkers/chemistry , Disease Models, Animal , Disease Progression , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/pathology , Phenotype , Software , Statistics as Topic
17.
Nature ; 330(6150): 754-8, 1987.
Article in English | MEDLINE | ID: mdl-2447503

ABSTRACT

Duchenne muscular dystrophy (DMD) is a human X-linked biochemical defect resulting in the progressive wasting of skeletal muscle of affected individuals. It is the most common and is considered to be the most devastating of the muscular dystrophies, affecting about 1 in 3,500 live-born males. The gene that, when defective, results in this disorder was recently isolated. Using the cloned complementary DNA sequences corresponding to the DMD gene, antibodies have been produced that react with a protein species of relative molecular mass (Mr) approximately 400,000 (400K) which was absent in two DMD-affected individuals and in mdx mice. This protein species is called dystrophin because of its identification by molecular-genetic analysis of affected individuals. Here we show that dystrophin is associated with the triadic junctions in skeletal muscle, and is therefore probably involved with Ca2+ homoeostasis. We also show that the approximately 450K ryanodine receptor/sarcoplasmic reticulum Ca2+ channel, which has the large size and subcellular distribution characteristics of dystrophin, is an immunologically distinct protein species.


Subject(s)
Muscle Proteins/metabolism , Muscles/ultrastructure , Muscular Dystrophy, Animal/pathology , Animals , Ca(2+) Mg(2+)-ATPase/metabolism , Calcium/physiology , Calcium-Transporting ATPases/metabolism , Cell Compartmentation , Dystrophin , Immunosorbent Techniques , Ion Channels/physiology , Mice , Microsomes/metabolism , Receptors, Cholinergic/metabolism , Ryanodine Receptor Calcium Release Channel
18.
Muscle Nerve ; 10(5): 391-6, 1987 Jun.
Article in English | MEDLINE | ID: mdl-3614253

ABSTRACT

Dimethyl-4, 4'-dimethoxy-5, 6, 5'-6'-dimethylenedioxybiphenyl-2, 2'-dicarboxylate (DDB) is a synthetic analogue of Schizandrin C, an active compound isolated from a Chinese herb, Fructus schizandrae. We administered this compound to dystrophic hamsters in vivo for 31 days. This led to a 61% reduction of the calcium content, an 86% reduction of the area of calcium deposits, and a 52% reduction of the area of necrosis of cardiac muscle. However, skeletal muscle necrosis was not significantly improved. No clear change in plasma creatine kinase (CK) was observed. In an in vitro incubation study, the rate of CK release and tetanus tension of the extensor digitorum longus muscle of dystrophic hamsters were not substantially changed by the addition of DDB. This study suggests that DDB has some effect on cardiac necrosis, and that it might be useful for treatment of the cardiac involvement in patients with muscular dystrophy or other conditions with accompaning Ca accumulation.


Subject(s)
Dioxoles/therapeutic use , Muscular Dystrophy, Animal/drug therapy , Plant Extracts/therapeutic use , Animals , Calcium/metabolism , Creatine Kinase/blood , Cricetinae , Male , Muscle Contraction , Muscles/pathology , Muscles/physiopathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Myocardium/metabolism , Necrosis , Plants, Medicinal , Reference Values
19.
Differentiation ; 32(2): 181-4, 1986.
Article in English | MEDLINE | ID: mdl-3792705

ABSTRACT

The isolation and characterization of a myogenic cell line from C57BL/6J/dydy mice is described. This line (DyA4) maintains the morphological, biochemical and electrophysiological characteristics of the primary cultured cells, at least for 20 passages. The cells actively divide as long as they are subcultured in media supplemented with horse serum and embryo extract. If the cells are not subcultured for a few days, they fuse into multinucleated contracting myotubes, which readily synthesize specific muscle products such as acetylcholinesterase and acetylcholine receptor. This dystrophic cell line expresses in vitro the same altered phenotype that is characteristic of dystrophic muscle cells in primary cultures, namely reduced acetylcholine sensitivity and reduced acetylcholine receptor expression. Because they can be grown in large amounts, and represent a pure muscle cell population which express an altered phenotype in an in vitro aneural avascular environment, DyA4 cells provide a very useful model system for investigating the pathogenesis of murine muscular dystrophy.


Subject(s)
Muscles/pathology , Muscular Dystrophy, Animal/pathology , Acetylcholinesterase/isolation & purification , Acetylcholinesterase/metabolism , Animals , Cell Line , Cells, Cultured , Clone Cells , Mice , Mice, Inbred C57BL , Muscles/cytology , Muscles/enzymology , Muscular Dystrophy, Animal/enzymology , Phenotype
20.
J S Afr Vet Assoc ; 55(1): 39-40, 1984 Mar.
Article in Afrikaans | MEDLINE | ID: mdl-6545370

ABSTRACT

Six 1 1/2-month old ostrich chickens in the Upington district of the Cape Province developed lameness. Two died and pathological examination of one of them revealed lesions identical to those of white muscle disease in the larger muscle groups. Vitamin E-selenium therapy cured the other 4. The diet of the animals consisted mainly of lucerne (alfalfa).


Subject(s)
Bird Diseases/etiology , Muscular Dystrophy, Animal/etiology , Animals , Birds , Muscular Dystrophy, Animal/diet therapy , Muscular Dystrophy, Animal/pathology , Selenium/therapeutic use , Vitamin E/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL