Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Skelet Muscle ; 6: 14, 2016.
Article in English | MEDLINE | ID: mdl-27047655

ABSTRACT

BACKGROUND: Myostatin (Mstn) is a negative regulator of muscle growth whose inhibition promotes muscle growth and regeneration. Dystrophin-deficient mdx mice in which myostatin is knocked out or inhibited postnatally have a less severe phenotype with greater total mass and strength and less fibrosis and fatty replacement of muscles than mdx mice with wild-type myostatin expression. Dogs with golden retriever muscular dystrophy (GRMD) have previously been noted to have increased muscle mass and reduced fibrosis after systemic postnatal myostatin inhibition. Based partly on these results, myostatin inhibitors are in development for use in human muscular dystrophies. However, persisting concerns regarding the effects of long-term and profound myostatin inhibition will not be easily or imminently answered in clinical trials. METHODS: To address these concerns, we developed a canine (GRippet) model by crossbreeding dystrophin-deficient GRMD dogs with Mstn-heterozygous (Mstn (+/-)) whippets. A total of four GRippets (dystrophic and Mstn (+/-)), three GRMD (dystrophic and Mstn wild-type) dogs, and three non-dystrophic controls from two litters were evaluated. RESULTS: Myostatin messenger ribonucleic acid (mRNA) and protein levels were downregulated in both GRMD and GRippet dogs. GRippets had more severe postural changes and larger (more restricted) maximal joint flexion angles, apparently due to further exaggeration of disproportionate effects on muscle size. Flexors such as the cranial sartorius were more hypertrophied on magnetic resonance imaging (MRI) in the GRippets, while extensors, including the quadriceps femoris, underwent greater atrophy. Myostatin protein levels negatively correlated with relative cranial sartorius muscle cross-sectional area on MRI, supporting a role in disproportionate muscle size. Activin receptor type IIB (ActRIIB) expression was higher in dystrophic versus control dogs, consistent with physiologic feedback between myostatin and ActRIIB. However, there was no differential expression between GRMD and GRippet dogs. Satellite cell exhaustion was not observed in GRippets up to 3 years of age. CONCLUSIONS: Partial myostatin loss may exaggerate selective muscle hypertrophy or atrophy/hypoplasia in GRMD dogs and worsen contractures. While muscle imbalance is not a feature of myostatin inhibition in mdx mice, findings in a larger animal model could translate to human experience with myostatin inhibitors.


Subject(s)
Contracture/metabolism , Dystrophin/deficiency , Joints/metabolism , Muscular Dystrophy, Animal/metabolism , Myostatin/deficiency , Quadriceps Muscle/metabolism , Activin Receptors, Type II/metabolism , Animals , Animals, Genetically Modified , Biomechanical Phenomena , Contracture/genetics , Contracture/pathology , Contracture/physiopathology , Disease Models, Animal , Dogs , Dystrophin/genetics , Gait , Genetic Predisposition to Disease , Hybridization, Genetic , Joints/pathology , Joints/physiopathology , Magnetic Resonance Imaging , Muscle Strength , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Myostatin/genetics , PAX7 Transcription Factor/metabolism , Phenotype , Posture , Quadriceps Muscle/growth & development , Quadriceps Muscle/pathology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology
2.
Skelet Muscle ; 4: 18, 2014.
Article in English | MEDLINE | ID: mdl-25789154

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene and afflicts skeletal and cardiac muscles. Previous studies showed that DMD is associated with constitutive activation of NF-κB, and in dystrophin-deficient mdx and utrophin/dystrophin (utrn (-/-) ;mdx) double knock out (dko) mouse models, inhibition of NF-κB with the Nemo Binding Domain (NBD) peptide led to significant improvements in both diaphragm and cardiac muscle function. METHODS: A trial in golden retriever muscular dystrophy (GRMD) canine model of DMD was initiated with four primary outcomes: skeletal muscle function, MRI of pelvic limb muscles, histopathologic features of skeletal muscles, and safety. GRMD and wild type dogs at 2 months of age were treated for 4 months with NBD by intravenous infusions. Results were compared with those collected from untreated GRMD and wild type dogs through a separate, natural history study. RESULTS: Results showed that intravenous delivery of NBD in GRMD dogs led to a recovery of pelvic limb muscle force and improvement of histopathologic lesions. In addition, NBD-treated GRMD dogs had normalized postural changes and a trend towards lower tissue injury on magnetic resonance imaging. Despite this phenotypic improvement, NBD administration over time led to infusion reactions and an immune response in both treated GRMD and wild type dogs. CONCLUSIONS: This GRMD trial was beneficial both in providing evidence that NBD is efficacious in a large animal DMD model and in identifying potential safety concerns that will be informative moving forward with human trials.

3.
Neuromuscul Disord ; 24(1): 63-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24295812

ABSTRACT

Golden retriever muscular dystrophy (GRMD) is a well-established model of Duchenne muscular dystrophy. The value of this model would be greatly enhanced with practical tools to monitor progression of respiratory dysfunction during treatment trials. Arterial blood gas analysis, tidal breathing spirometry, and respiratory inductance plethysmography (RIP) were performed to determine if quantifiable abnormalities could be identified in unsedated, untrained, GRMD dogs. Results from 11 dogs with a mild phenotype of GRMD and 11 age-matched carriers were compared. Arterial blood gas analysis was successfully performed in all dogs, spirometry in 21 of 22 (95%) dogs, and RIP in 18 of 20 (90%) dogs. Partial pressure of carbon dioxide and bicarbonate concentration were higher in GRMD dogs. Tidal breathing peak expiratory flows were markedly higher in GRMD dogs. Abnormal abdominal motion was present in 7 of 10 (70%) GRMD dogs. Each technique provided objective, quantifiable measures that will be useful for monitoring respiratory function in GRMD dogs during clinical trials while avoiding the influence of sedation on results. Increased expiratory flows and the pattern of abdominal breathing are novel findings, not reported in people with Duchenne muscular dystrophy, and might be a consequence of hyperinflation.


Subject(s)
Muscular Dystrophy, Animal/physiopathology , Respiration Disorders/physiopathology , Animals , Blood Gas Analysis , Disease Progression , Dogs , Female , Heart Rate , Male , Muscular Dystrophy, Animal/blood , Respiration Disorders/blood , Respiratory Rate
4.
Phys Med Rehabil Clin N Am ; 23(1): 149-72, xii, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22239881

ABSTRACT

Mutations in the dystrophin gene cause Duchenne and Becker muscular dystrophy in humans and syndromes in mice, dogs, and cats. Affected humans and dogs have progressive disease that leads primarily to muscle atrophy. Mdx mice progress through an initial phase of muscle hypertrophy followed by atrophy. Cats have persistent muscle hypertrophy. Hypertrophy in humans has been attributed to deposition of fat and connective tissue (pseudohypertrophy). Increased muscle mass (true hypertrophy) has been documented in animal models. Muscle hypertrophy can exaggerate postural instability and joint contractures. Deleterious consequences of muscle hypertrophy should be considered when developing treatments for muscular dystrophy.


Subject(s)
Hypertrophy/etiology , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Animals , Contracture/etiology , Contracture/physiopathology , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Humans , Hypertrophy/metabolism , Hypertrophy/physiopathology , Kyphosis/etiology , Muscle Strength , Muscle, Skeletal/physiopathology , Muscular Dystrophies/complications , Muscular Dystrophies/drug therapy , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Myostatin/antagonists & inhibitors
5.
Mamm Genome ; 23(1-2): 85-108, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22218699

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder in which the loss of dystrophin causes progressive degeneration of skeletal and cardiac muscle. Potential therapies that carry substantial risk, such as gene- and cell-based approaches, must first be tested in animal models, notably the mdx mouse and several dystrophin-deficient breeds of dogs, including golden retriever muscular dystrophy (GRMD). Affected dogs have a more severe phenotype, in keeping with that of DMD, so may better predict disease pathogenesis and treatment efficacy. Various phenotypic tests have been developed to characterize disease progression in the GRMD model. These biomarkers range from measures of strength and joint contractures to magnetic resonance imaging. Some of these tests are routinely used in clinical veterinary practice, while others require specialized equipment and expertise. By comparing serial measurements from treated and untreated groups, one can document improvement or delayed progression of disease. Potential treatments for DMD may be broadly categorized as molecular, cellular, or pharmacologic. The GRMD model has increasingly been used to assess efficacy of a range of these therapies. A number of these studies have provided largely general proof-of-concept for the treatment under study. Others have demonstrated efficacy using the biomarkers discussed. Importantly, just as symptoms in DMD vary among patients, GRMD dogs display remarkable phenotypic variation. Though confounding statistical analysis in preclinical trials, this variation offers insight regarding the role that modifier genes play in disease pathogenesis. By correlating functional and mRNA profiling results, gene targets for therapy development can be identified.


Subject(s)
Disease Models, Animal , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Animals , Biomarkers , Dogs , Dystrophin/deficiency , Dystrophin/genetics , Joints/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology
6.
Mol Ther ; 20(3): 580-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22146342

ABSTRACT

Although restoration of dystrophin expression via exon skipping in both cardiac and skeletal muscle has been successfully demonstrated in the mdx mouse, restoration of cardiac dystrophin expression in large animal models of Duchenne muscular dystrophy (DMD) has proven to be a challenge. In large animals, investigators have focused on using intravenous injection of antisense oligonucleotides (AO) to mediate exon skipping. In this study, we sought to optimize restoration of cardiac dystrophin expression in the golden retriever muscular dystrophy (GRMD) model using percutaneous transendocardial delivery of recombinant AAV6 (rAAV6) to deliver a modified U7 small nuclear RNA (snRNA) carrying antisense sequence to target the exon splicing enhancers of exons 6 and 8 and correct the disrupted reading frame. We demonstrate restoration of cardiac dystrophin expression at 13 months confirmed by reverse transcription-PCR (RT-PCR) and immunoblot as well as membrane localization by immunohistochemistry. This was accompanied by improved cardiac function as assessed by cardiac magnetic resonance imaging (MRI). Percutaneous transendocardial delivery of rAAV6 expressing a modified U7 exon skipping construct is a safe, effective method for restoration of dystrophin expression and improvement of cardiac function in the GRMD canine and may be easily translatable to human DMD patients.


Subject(s)
Alternative Splicing , Dependovirus/genetics , Dystrophin/genetics , Genetic Vectors/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Animals , Cell Line , Disease Models, Animal , Dogs , Dystrophin/metabolism , Echocardiography , Exons , Fibrosis , Gene Expression , Gene Order , Gene Transfer Techniques , Genetic Vectors/pharmacokinetics , Genome, Viral , Humans , Magnetic Resonance Imaging , Muscular Dystrophy, Duchenne/diagnosis , Myocardium/pathology , RNA, Messenger/metabolism
7.
Hum Gene Ther ; 22(12): 1499-509, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21787232

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked recessive disease affecting 1 in 3,500 newborn boys for which there is no effective treatment or cure. One novel strategy that has therapeutic potential for DMD is inhibition of myostatin, a negative regulator of skeletal muscle mass that may also promote fibrosis. Therefore, our goal in this study was to evaluate systemic myostatin inhibition in the golden retriever model of DMD (GRMD). GRMD canines underwent liver-directed gene transfer of a self-complementary adeno-associated virus type 8 vector designed to express a secreted dominant-negative myostatin peptide (n = 4) and were compared with age-matched, untreated GRMD controls (n = 3). Dogs were followed with serial magnetic resonance imaging (MRI) for 13 months to assess cross-sectional area and volume of skeletal muscle, then euthanized so that tissue could be harvested for morphological and histological analysis. We found that systemic myostatin inhibition resulted in increased muscle mass in GRMD dogs as assessed by MRI and confirmed at tissue harvest. We also found that hypertrophy of type IIA fibers was largely responsible for the increased muscle mass and that reductions in serum creatine kinase and muscle fibrosis were associated with long-term myostatin inhibition in GRMD. This is the first report describing the effects of long-term, systemic myostatin inhibition in a large-animal model of DMD, and we believe that the simple and effective nature of our liver-directed gene-transfer strategy makes it an ideal candidate for evaluation as a novel therapeutic approach for DMD patients.


Subject(s)
Dependovirus/genetics , Genetic Vectors/therapeutic use , Liver/metabolism , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Myostatin/antagonists & inhibitors , Animals , Blotting, Western , Creatine Kinase/genetics , Creatine Kinase/metabolism , Dogs , Fibrosis/metabolism , Fibrosis/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Liver/pathology , Magnetic Resonance Imaging , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Myostatin/genetics , Myostatin/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism
8.
Methods Mol Biol ; 709: 105-23, 2011.
Article in English | MEDLINE | ID: mdl-21194024

ABSTRACT

Studies of canine models of Duchenne muscular dystrophy (DMD) provide insight regarding disease pathogenesis and treatment efficacy. To take maximal advantage, colonies of affected dogs must be maintained and outcome parameters developed. In this chapter, we review our 25 years of experience with the golden retriever muscular dystrophy (GRMD) model. Key challenges in colony development (breeding, neonatal death, and the risk of inbreeding) and representative functional measurements (tibiotarsal joint angle and torque force; and eccentric contraction decrement) are discussed.


Subject(s)
Muscle Strength/physiology , Muscle, Skeletal , Muscular Dystrophy, Animal/physiopathology , Animals , Breeding , Disease Models, Animal , Dogs , Dystrophin/deficiency , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/mortality , Muscular Dystrophy, Duchenne/physiopathology
9.
Front Pharmacol ; 2: 89, 2011.
Article in English | MEDLINE | ID: mdl-22291646

ABSTRACT

Calpains likely play a role in the pathogenesis of Duchenne muscular dystrophy (DMD). Accordingly, calpain inhibition may provide therapeutic benefit to DMD patients. In the present study, we sought to measure benefit from administration of a novel calpain inhibitor, C101, in a canine muscular dystrophy model. Specifically, we tested the hypothesis that treatment with C101 mitigates progressive weakness and severe muscle pathology observed in young dogs with golden retriever muscular dystrophy (GRMD). Young (6-week-old) GRMD dogs were treated daily with either C101 (17 mg/kg twice daily oral dose, n=9) or placebo (vehicle only, n =7) for 8 weeks. A battery of functional tests, including tibiotarsal joint angle, muscle/fat composition, and pelvic limb muscle strength were performed at baseline and every 2 weeks during the 8-week study. Results indicate that C101-treated GRMD dogs maintained strength in their cranial pelvic limb muscles (tibiotarsal flexors) while placebo-treated dogs progressively lost strength. However, concomitant improvement was not observed in posterior pelvic limb muscles (tibiotarsal extensors). C101 treatment did not mitigate force drop following repeated eccentric contractions and no improvement was seen in the development of joint contractures, lean muscle mass, or muscle histopathology. Taken together, these data do not support the hypothesis that treatment with C101 mitigates progressive weakness or ameliorates severe muscle pathology observed in young dogs with GRMD.

10.
Mol Ther ; 18(8): 1501-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20517298

ABSTRACT

Duchenne (DMD) and golden retriever (GRMD) muscular dystrophy are caused by genetic mutations in the dystrophin gene and afflict striated muscles. We investigated systemic gene delivery in 4-day-old GRMD dogs given a single intravenous injection of an AAV9 vector (1.5 x 10(14) vector genomes/kg) carrying a human codon-optimized human mini-dystrophin gene under control of the cytomegalovirus (CMV) promoter. One of the three treated dogs was euthanized 9 days later due to pre-existing conditions. Scattered mini-dystrophin-positive myofibers were seen by immunofluorescent (IF) staining in numerous muscles. At the end of the 16-week study, the other two dogs showed generalized muscle expression of mini-dystrophin in ~15% to nearly 100% of myofibers. Western blot and vector DNA quantitative PCR results agreed with the IF data. Delayed growth and pelvic limb muscle atrophy and contractures were seen several weeks after vector delivery. T-2 weighted magnetic resonance imaging (MRI) at 8 weeks showed increased signal intensity compatible with inflammation in several pelvic limb muscles. This marked early inflammatory response raised concerns regarding methodology. Use of the ubiquitous CMV promoter, extra-high vector dose, and marked expression of a human protein in canine muscles may have contributed to the pathologic changes seen in the pelvic limbs.


Subject(s)
Adenoviridae/genetics , Dystrophin/deficiency , Dystrophin/metabolism , Genetic Therapy/methods , Genetic Vectors/genetics , Injections, Intravenous/methods , Muscles/metabolism , Animals , Animals, Newborn , Blotting, Western , Dogs , Dystrophin/genetics , Female , Genetic Vectors/administration & dosage , Humans , Magnetic Resonance Imaging , Muscles/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/therapy
11.
Hum Gene Ther ; 20(1): 1-10, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18828709

ABSTRACT

Inhibition or blockade of myostatin, a negative growth factor of skeletal muscle, enhances muscle growth and therefore is considered a promising strategy for the treatment of muscle-wasting diseases such as the muscular dystrophies. Previously, we showed that myostatin blockade in both normal and dystrophin-deficient mdx mice by systemic delivery of the myostatin propeptide (MPRO) gene by an adeno-associated virus serotype 8 (AAV8) vector could enhance muscle growth and ameliorate dystrophic lesions. Here, we further investigate whether the muscle growth effect of myostatin blockade can be achieved in dogs by gene transfer. First, we cloned the canine MPRO gene, packaged it in the AAV8 vector, and showed robust muscle-enhancing effects after systemic delivery into neonatal mice. This vector was then further tested in two 3-month-old normal dogs (weighing 9.7 and 6.3 kg). The vector was delivered to one limb by hydrodynamic vein injection, and the contralateral limb served as a control. The delivery procedure was safe, without discernible adverse effects. AAV vector DNA and MPRO gene expression were detected by quantitative polymerase chain reaction, Western blotting, and immunofluorescence staining of muscle biopsies. Overexpression of MPRO resulted in enhanced muscle growth without a cytotoxic T lymphocytic immune response, as evidenced by larger myofibers in multiple muscles, increased muscle volume determined by magnetic resonance imaging, and the lack of CD4+ and CD8+ T cell infiltration in the vector-injected limbs. Our preliminary study thus supports further investigation of this therapeutic strategy in the dystrophin-deficient golden retriever muscular dystrophy dog model.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Muscle, Skeletal/growth & development , Myostatin/administration & dosage , Peptides/administration & dosage , Animals , Dependovirus/classification , Dependovirus/metabolism , Dogs , Gene Transfer Techniques , Genetic Vectors/genetics , Injections, Intravenous/methods , Male , Mice , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myostatin/genetics , Myostatin/metabolism , Myostatin/pharmacology , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Serotyping
12.
Muscle Nerve ; 30(6): 767-73, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15468337

ABSTRACT

Glucocorticoid use may provide short-term functional improvement in boys with Duchenne muscular dystrophy (DMD). We report functional and histopathologic changes following a 4-month course of daily oral prednisone in a canine model of DMD, termed golden retriever muscular dystrophy (GRMD). Muscle extension forces in GRMD dogs treated daily with 1 and 2 mg/kg prednisone measured 2.349 +/- 0.92 and 3.486 +/- 0.67 N/kg, respectively, compared to 1.927 +/- 0.63 N/kg in untreated GRMD controls (p < 0.05 for 2 mg/kg group); GRMD muscle flexion forces measured 0.435 +/- 0.13 and 0.303 +/- 0.08 N/kg, respectively, compared to 0.527 +/- 0.01 N/kg in untreated GRMD controls (p < 0.05 for both groups). Although cranial sartorius hypertrophy and tibiotarsal joint angles also tended to improve, myofiber calcification increased and fetal myosin expression decreased following prednisone. Thus, functional data indicate benefit but histopathologic changes following prednisone treatment in GRMD suggest possible deleterious consequences.


Subject(s)
Muscular Dystrophy, Animal/drug therapy , Prednisone/therapeutic use , Animals , Dogs , Isometric Contraction/drug effects , Isometric Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Prednisone/adverse effects , Prednisone/pharmacology
13.
Blood ; 104(13): 4311-8, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15328150

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene on the X-chromosome that result in skeletal and cardiac muscle damage and premature death. Studies in mice, including the mdx mouse model of DMD, have demonstrated that circulating bone marrow-derived cells can participate in skeletal muscle regeneration, but the potential clinical utility of treating human DMD by allogeneic marrow transplantation from a healthy donor remains unknown. To assess whether allogeneic hematopoietic cell transplantation (HCT) provides clinically relevant levels of donor muscle cell contribution in dogs with canine X-linked muscular dystrophy (c-xmd), 7 xmd dogs were given hematopoietic cell (HC) transplants from nonaffected littermates. Compared with the pretransplantation baseline, the number of dystrophin-positive fibers and the amount of wild-type dystrophin RNA did not increase after HCT, with observation periods ranging from 28 to 417 days. Similar results were obtained when the recipient dogs were given granulocyte colony-stimulating factor (G-CSF) after their initial transplantation to mobilize the cells. Despite successful allogeneic HCT and a permissive environment for donor muscle engraftment, there was no detectable contribution of bone marrow-derived cells to either skeletal muscle or muscle precursor cells assayed by clonal analyses at a level of sensitivity that should detect as little as 0.1% donor contribution.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Animals , Disease Models, Animal , Dogs , Female , Gene Expression Regulation/physiology , Immunosuppression Therapy/methods , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/therapy , Stem Cell Transplantation , Transplantation, Homologous , Whole-Body Irradiation
14.
Neuromuscul Disord ; 13(6): 493-500, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12899877

ABSTRACT

The degree of atrophy or hypertrophy of selected pelvic limb muscles was determined in the canine homologue of Duchenne muscular dystrophy. While most muscles were atrophied, the caudal and cranial sartorius were hypertrophied. Cranial sartorius weights were corrected for body weight and endomysial space to determine true muscle weights (g/kg; mean+/-SD) in three golden retriever muscular dystrophy age groups, 4-10 (Group 1; n=15), 13-26 (Group 2; n=4), and 33-66 (Group 3; n=4) months and grouped normal dogs (6-20 months; n=12). Group 1 golden retriever muscular dystrophy weights (2.2063+/-0.6884) were greater than those of normal dogs (1.2699+/-0.1966), indicating that young golden retriever muscular dystrophy dogs have true cranial sartorius muscle hypertrophy. Values of Group 2 (1.3758+/-0.5078) and Group 3 (0.5720+/-0.2423) golden retriever muscular dystrophy dogs were less than those of Group 1, suggesting that the cranial sartorius muscle atrophies over time. Given that cranial sartorius muscle weight correlated with tarsal joint angle in affected dogs (r=-0.817), the hypertrophied muscle may play a role analogous to iliotibial band tightness in Duchenne muscular dystrophy.


Subject(s)
Dog Diseases/pathology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Animals , Contracture/pathology , Dogs , Hindlimb , Hypertrophy , Myofibrils/pathology , Pelvis
15.
Arch Phys Med Rehabil ; 83(11): 1572-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12422328

ABSTRACT

OBJECTIVE: To test the hypothesis that eccentric contractions induce greater injury in dystrophic compared with normal canine muscle. DESIGN: Blinded cohort study. SETTING: Animal laboratory. ANIMALS: Ten dogs with a homologue to Duchenne muscular dystrophy (Golden retriever muscular dystrophy [GRMD]) and 10 normal littermates. INTERVENTIONS: Contractions induced in tibiotarsal flexors and extensors by sciatic nerve stimulation. Because more powerful extensors overrode flexors, eccentric contractions occurred in flexors. Concentric contractions were induced in contralateral flexors by peroneal nerve stimulation. MAIN OUTCOME MEASURE: Tibiotarsal flexion force 3 days after contractions. Muscle was examined for injury (esterase activity, Evans blue dye penetration) and regeneration (embryonic myosin isoform expression). RESULTS: Mean force deficit after eccentric flexor contractions was 43.3%+/-25.7% in GRMD dogs compared with 25.0%+/-18.4% in controls (P=.04, Wilcoxon rank-sum test). Concentric contractions induced force deficits in GRMD but not normal dogs; however, the difference between the 2 groups was not significant (P=.08, Wilcoxon rank-sum test). After concentric contractions in controls, force decrements correlated with esterase activity measured by area (r=.794, P=.006) and intensity (r=.697, P=.025, Spearman rank correlation). No other significant correlation was detected between force and biopsy data. CONCLUSIONS: Force data support the hypothesis that eccentric contractions induce greater injury in dystrophic compared with normal canine muscle. Phenotypic features of the dystrophic canine model used here are similar to those of humans with Duchenne's.


Subject(s)
Disease Models, Animal , Dog Diseases/physiopathology , Isometric Contraction , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Animals , Biomechanical Phenomena , Biopsy , Case-Control Studies , Dog Diseases/genetics , Dog Diseases/pathology , Dogs , Dystrophin/genetics , Electric Stimulation , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Mutation/genetics , Myofibrils/pathology , Necrosis , Peroneal Nerve , Phenotype , Random Allocation , Range of Motion, Articular , Sciatic Nerve , Single-Blind Method , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...