Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Top Companion Anim Med ; 42: 100500, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33249241

ABSTRACT

Muscular dystrophies in dogs and cats represent a heterogeneous group of inherited, sometimes congenital, but infrequently diagnosed, progressive neuromuscular disorders. A correct identification and characterization of canine and feline muscular dystrophies could increase diagnostic and treatment strategies for veterinary neurologists and could identify useful animal models for the study of human dystrophies. However, in dogs and cats, diagnosis of muscular dystrophies is challenging due to a nonspecific clinical phenotype and pathological lesions, thus is most likely underestimated. We performed immunofluorescence and Western blot techniques using a wide panel of antibodies against proteins involved in human dystrophies (dystrophin mid-rod and carboxyterminal domain, α, ß, γ, and δ-sarcoglycan, α-dystroglycan, caveolin-3, emerin, merosin, dysferlin, calpain-3, spectrin epitopes), on 9 canine and 3 feline muscle biopsies characterized by myopathic changes. Dystrophin deficiency was detected in 3 dogs and 2 novel canine muscular dystrophies have been identified, characterized by deficiency of caveolin-3 and calpain-3, respectively. In 2 cats, deficiency of ß-SG and carboxyterminal domain of dystrophin in all muscle fibers has been detected. Performing immunofluorescence and Western blot analyses with a wider panel of antibodies allowed a correct identification of muscular dystrophies in dogs and cats and provides a direction for subsequent targeted genetic testing.


Subject(s)
Cat Diseases , Dog Diseases , Dystrophin/metabolism , Muscular Dystrophies/metabolism , Sarcoglycans/genetics , Animals , Cats , Dogs , Immunohistochemistry/veterinary , Muscle, Skeletal , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Sarcoglycans/analysis , Sarcoglycans/deficiency
2.
Cell Death Dis ; 11(2): 127, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32071288

ABSTRACT

Muscular Dystrophies are severe genetic diseases due to mutations in structural genes, characterized by progressive muscle wasting that compromises patients' mobility and respiratory functions. Literature underlined oxidative stress and inflammation as key drivers of these pathologies. Interestingly among different myofiber classes, type I fibers display a milder dystrophic phenotype showing increased oxidative metabolism. This work shows the benefits of a cyanidin-enriched diet, that promotes muscle fiber-type switch and reduced inflammation in dystrophic alpha-sarcoglyan (Sgca) null mice having, as a net outcome, morphological and functional rescue. Notably, this benefit is achieved also when the diet is administered in dystrophic animals when the signs of the disease are seriously evident. Our work provides compelling evidence that a cyanidin-rich diet strongly delays the progression of muscular dystrophies, paving the way for a combinatorial approach where nutritional-based reduction of muscle inflammation and oxidative stress facilitate the successful perspectives of definitive treatments.


Subject(s)
Anthocyanins/administration & dosage , Dietary Supplements , Inflammation Mediators/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Oxidative Stress , Sarcoglycanopathies/diet therapy , Animals , Disease Models, Animal , Disease Progression , Female , Male , Mice, Knockout , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Organelle Biogenesis , Phenotype , Protein Carbonylation , Sarcoglycanopathies/genetics , Sarcoglycanopathies/metabolism , Sarcoglycanopathies/pathology , Sarcoglycans/deficiency , Sarcoglycans/genetics
3.
Lab Invest ; 100(6): 887-899, 2020 06.
Article in English | MEDLINE | ID: mdl-32060408

ABSTRACT

Genetic cardiomyopathy is a group of intractable cardiovascular disorders involving heterogeneous genetic contribution. This heterogeneity has hindered the development of life-saving therapies for this serious disease. Genetic mutations in dystrophin and its associated glycoproteins cause cardiomuscular dysfunction. Large animal models incorporating these genetic defects are crucial for developing effective medical treatments, such as tissue regeneration and gene therapy. In the present study, we knocked out the δ-sarcoglycan (δ-SG) gene (SGCD) in domestic pig by using a combination of efficient de novo gene editing and somatic cell nuclear transfer. Loss of δ-SG expression in the SGCD knockout pigs caused a concomitant reduction in the levels of α-, ß-, and γ-SG in the cardiac and skeletal sarcolemma, resulting in systolic dysfunction, myocardial tissue degeneration, and sudden death. These animals exhibited symptoms resembling human genetic cardiomyopathy and are thus promising for use in preclinical studies of next-generation therapies.


Subject(s)
Cardiomyopathies , Sarcoglycans , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Female , Frameshift Mutation/genetics , Gene Knockout Techniques , Male , Myocardium/chemistry , Myocardium/metabolism , Myocardium/pathology , Sarcoglycans/deficiency , Sarcoglycans/genetics , Swine
4.
Circ Heart Fail ; 12(4): e005418, 2019 04.
Article in English | MEDLINE | ID: mdl-30922066

ABSTRACT

BACKGROUND: Metformin is a popular antidiabetic agent that is also used to treat heart failure patients with type 2 diabetes mellitus. Several reports suggest that metformin may also have cardioprotective effects in patients without diabetes mellitus. In the present study, we investigated the possible therapeutic effect of metformin in heart failure and its underlying molecular mechanisms using a δ-sarcoglycan-deficient mouse model of dilated cardiomyopathy. METHODS AND RESULTS: Thirty-two-week-old δ-sarcoglycan-deficient mice exhibiting established cardiomyopathy with extensive left ventricular dilatation and dysfunction were administered saline or metformin (200 mg/kg per day) for 4 weeks using osmotic mini-pumps. Metformin partially reversed the left ventricular dilatation (reverse remodeling) and significantly improved cardiac function. The hearts of metformin-treated mice showed less fibrosis, less cardiomyocyte hypertrophy, and fewer degenerative subcellular changes than saline-treated mice. These effects were accompanied by restored expression of the sarcomeric proteins myosin heavy chain and troponin I, and their transcription factor, GATA-4. Autophagy was enhanced in the hearts from metformin-treated mice, as indicated by increase of myocardial microtubule-associated protein-1 LC-3 (light chain 3)-II levels and LC3-II/-I ratios as well as levels of cathepsin D and ATP. In addition, increased numbers of autophagic vacuoles and lysosomes were accompanied increased AMP-activated protein kinase activity and suppression of mammalian target of rapamycin phosphorylation. Finally, autophagic flux assays using short-term chloroquine treatment revealed that autophagy was activated in δ-sarcoglycan-deficient hearts and was further augmented by metformin treatment. CONCLUSIONS: Metformin is a beneficial pharmacological tool that mitigates heart failure caused by δ-sarcoglycan deficiency in association with enhanced autophagy.


Subject(s)
Autophagy/physiology , Cardiomyopathies/genetics , Sarcoglycans/deficiency , Ventricular Remodeling/genetics , Animals , Autophagy/genetics , Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Cardiomyopathy, Dilated/metabolism , Diabetes Mellitus, Type 2/metabolism , Heart Failure/genetics , Hypoglycemic Agents/therapeutic use , Metformin/pharmacology , Mice, Transgenic , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Ventricular Remodeling/drug effects
5.
PLoS One ; 14(1): e0210971, 2019.
Article in English | MEDLINE | ID: mdl-30673749

ABSTRACT

Galectin-3 is a ß-galactoside-binding lectin which is important in cell proliferation and apoptotic regulation. Recently, serum galectin-3 has been shown to have prognostic value as a biomarker in heart failure. Encephalomyocarditis virus (EMCV) can cause severe myocarditis, congestive heart failure and dilated cardiomyopathy as well as encephalitis in various animals including mice. The pathophysiological role of galectin-3 in acute myocarditis following viral infection is not fully understood. The goal of this study is to determine the cardiac localization and the time-course of galectin-3 expression in heart failure after viral inoculation with EMCV. At 12, 24, 48, 96 hours, 7 and 10 days after intraperitoneal EMCV inoculation, animals were examined histologically and analyzed for the expression of galectin-3 and Iba1. Galectin-3 was up-regulated in degenerated fibrotic lesions of cardiac tissues 96 hours after viral inoculation and were followed by myocardial fibrosis. At the same time, Iba1 positive macrophages were observed within the inflammatory sites. A time-course correlation between the number of galectin-3 positive cells and the cardiac area of degenerated fibrotic lesions was detected-serum galectin-3 increased at 96 hours and correlated well with the number of cardiac galectin-3 positive cells. Our results indicate that galectin-3 expression may be a useful biomarker of cardiac fibrotic degeneration in acute myocarditis following viral infection. In addition, measuring serum galectin-3 levels might be an early diagnostic method for detecting cardiac degeneration in acute myocarditis.


Subject(s)
Cardiovirus Infections/blood , Cardiovirus Infections/metabolism , Encephalomyocarditis virus , Galectin 3/blood , Galectin 3/metabolism , Myocarditis/blood , Myocarditis/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/metabolism , Cardiovirus Infections/pathology , Disease Models, Animal , Encephalomyocarditis virus/pathogenicity , Fibrosis , Immunohistochemistry , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Prognosis , Sarcoglycans/deficiency , Sarcoglycans/genetics
6.
Am J Pathol ; 189(2): 354-369, 2019 02.
Article in English | MEDLINE | ID: mdl-30448410

ABSTRACT

In muscular dystrophies, muscle membrane fragility results in a tissue-specific increase of danger-associated molecular pattern molecules (DAMPs) and infiltration of inflammatory cells. The DAMP extracellular ATP (eATP) released by dying myofibers steadily activates muscle and immune purinergic receptors exerting dual negative effects: a direct damage linked to altered intracellular calcium homeostasis in muscle cells and an indirect toxicity through the triggering of the immune response and inhibition of regulatory T cells. Accordingly, pharmacologic and genetic inhibition of eATP signaling improves the phenotype in models of chronic inflammatory diseases. In α-sarcoglycanopathy, eATP effects may be further amplified because α-sarcoglycan extracellular domain binds eATP and displays an ecto-ATPase activity, thus controlling eATP concentration at the cell surface and attenuating the magnitude and/or the duration of eATP-induced signals. Herein, we show that in vivo blockade of the eATP/P2X purinergic pathway by a broad-spectrum P2X receptor-antagonist delayed the progression of the dystrophic phenotype in α-sarcoglycan-null mice. eATP blockade dampened the muscular inflammatory response and enhanced the recruitment of forkhead box protein P3-positive immunosuppressive regulatory CD4+ T cells. The improvement of the inflammatory features was associated with increased strength, reduced necrosis, and limited expression of profibrotic factors, suggesting that pharmacologic purinergic antagonism, altering the innate and adaptive immune component in muscle infiltrates, might provide a therapeutic approach to slow disease progression in α-sarcoglycanopathy.


Subject(s)
Adenosine Triphosphate/immunology , Muscular Dystrophy, Animal , Myofibrils , Sarcoglycans/deficiency , T-Lymphocytes, Regulatory , Adenosine Triphosphate/genetics , Animals , Calcium/immunology , Chronic Disease , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Myofibrils/immunology , Myofibrils/pathology , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/immunology , Sarcoglycans/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
7.
FASEB J ; 33(2): 2047-2057, 2019 02.
Article in English | MEDLINE | ID: mdl-30216109

ABSTRACT

In skeletal muscle, extracellular matrix (ECM) remodeling can either support the complete regeneration of injured muscle or facilitate pathologic fibrosis and muscle degeneration. Muscular dystrophy (MD) is a group of genetic disorders that results in a progressive decline in muscle function and is characterized by the abundant deposition of fibrotic tissue. Unlike acute injury, where ECM remodeling is acute and transient, in MD, remodeling persists until fibrosis obstructs the regenerative efforts of diseased muscles. Thus, understanding how ECM is deposited and organized is critical in the context of muscle repair. Connective tissue growth factor (CTGF or CCN2) is a matricellular protein expressed by multiple cell types in response to tissue injury. Although used as a general marker of fibrosis, the cell type-dependent role of CTGF in dystrophic muscle has not been elucidated. To address this question, a conditional Ctgf myofiber and fibroblast-knockout mouse lines were generated and crossed to a dystrophic background. Only myofiber-selective inhibition of CTGF protected δ-sarcoglycan-null ( Sgcd-/-) mice from the dystrophic phenotype, and it did so by affecting collagen organization in a way that allowed for improvements in dystrophic muscle regeneration and function. To confirm that muscle-specific CTGF functions to mediate collagen organization, we generated mice with transgenic muscle-specific overexpression of CTGF. Again, genetic modulation of CTGF in muscle was not sufficient to drive fibrosis, but altered collagen content and organization after injury. Our results show that the myofibers are critical mediators of the deleterious effects associated with CTGF in MD and acutely injured skeletal muscle.-Petrosino, J. M., Leask, A., Accornero, F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle.


Subject(s)
Connective Tissue Growth Factor , Extracellular Matrix , Gene Expression Regulation , Muscle Fibers, Skeletal , Muscular Dystrophy, Animal , Animals , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Sarcoglycans/deficiency
8.
PLoS One ; 13(12): e0202838, 2018.
Article in English | MEDLINE | ID: mdl-30517097

ABSTRACT

OBJECTIVES: We hypothesized that systemic administration of high-mobility group box 1 fragment attenuates the progression of myocardial fibrosis and cardiac dysfunction in a hamster model of dilated cardiomyopathy by recruiting bone marrow mesenchymal stem cells thus causing enhancement of a self-regeneration system. METHODS: Twenty-week-old J2N-k hamsters, which are δ-sarcoglycan-deficient, were treated with systemic injection of high-mobility group box 1 fragment (HMGB1, n = 15) or phosphate buffered saline (control, n = 11). Echocardiography for left ventricular function, cardiac histology, and molecular biology were analyzed. The life-prolonging effect was assessed separately using the HMGB1 and control groups, in addition to a monthly HMGB1 group which received monthly systemic injections of high-mobility group box 1 fragment, 3 times (HMGB1, n = 11, control, n = 9, monthly HMGB1, n = 9). RESULTS: The HMGB1 group showed improved left ventricular ejection fraction, reduced myocardial fibrosis, and increased capillary density. The number of platelet-derived growth factor receptor-alpha and CD106 positive mesenchymal stem cells detected in the myocardium was significantly increased, and intra-myocardial expression of tumor necrosis factor α stimulating gene 6, hepatic growth factor, and vascular endothelial growth factor were significantly upregulated after high-mobility group box 1 fragment administration. Improved survival was observed in the monthly HMGB1 group compared with the control group. CONCLUSIONS: Systemic high-mobility group box 1 fragment administration attenuates the progression of left ventricular remodeling in a hamster model of dilated cardiomyopathy by enhanced homing of bone marrow mesenchymal stem cells into damaged myocardium, suggesting that high-mobility group box 1 fragment could be a new treatment for dilated cardiomyopathy.


Subject(s)
Bone Marrow Cells/metabolism , Cardiomyopathy, Dilated , Cell Self Renewal , HMGB1 Protein/pharmacology , Mesenchymal Stem Cells/metabolism , Sarcoglycans/deficiency , Ventricular Function, Left , Animals , Animals, Genetically Modified , Bone Marrow Cells/pathology , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Cricetinae , Disease Models, Animal , Echocardiography , Fibrosis , HMGB1 Protein/genetics , Mesenchymal Stem Cells/pathology , Ventricular Function, Left/drug effects , Ventricular Function, Left/genetics
9.
Free Radic Biol Med ; 129: 364-371, 2018 12.
Article in English | MEDLINE | ID: mdl-30312761

ABSTRACT

Generation of superoxide by xanthine oxidase can be stimulated under ischemic and aberrant calcium homeostasis. Because patients and mice with Duchenne muscular dystrophy (DMD) suffer from ischemia and excessive calcium influx, we tested the hypothesis that xanthine oxidase activity is elevated and contributes to disease pathology. Xanthine oxidase activity was measured by urinary isoxanthopterin in DMD patients at rest and in response to exercise. Urinary isoxanthopterin/creatinine was elevated compared to age-matched controls and Becker muscular dystrophy (BMD) patients. Concentrations were also increased after a six minute walk test in ambulatory patients. We also measured urinary isoxanthopterin in wildtype mice and a number of dystrophic mouse models; the DMD mouse model (mdx), mdx mice overexpressing a variety of transgenic miniaturized and chimeric skeletal muscle-specific dystrophins and utrophin and the ß-sarcoglycan deficient (Scgb-/-) mouse which represents type 2E human limb-girdle muscular dystrophy. Mdx and Scgb-/-mice had greater urinary isoxanthopterin/creatinine than wildtype mice while mdx mice expressing dystrophin or utrophin linking the extracellular matrix to the actin cytoskeleton were not different than wildtype. We also measured higher levels of urinary ortho-tyrosine in humans and mice deficient for dystrophin to confirm elevated oxidative stress. Surprisingly, mdx had lower xanthine oxidase protein levels and higher mRNA in gastrocnemius muscle compared to wildtype mice, however, the enzymatic activity of skeletal muscle xanthine oxidase was elevated above wildtype and a transgenic rescued mdx mouse (DysΔMTB-mdx). Downhill treadmill running also caused significant increases in mdx urinary isoxanthopterin that was prevented with the xanthine oxidase inhibitor allopurinol. Similarly, in vitro eccentric contraction-induced force drop of mdx muscle was attenuated by the allopurinol metabolite, oxypurinol. Together, our data suggests hyper-activity of xanthine oxidase in DMD, identifies xanthine oxidase activity as a contributing factor in eccentric contraction-induced force drop of dystrophin-deficient skeletal muscle and highlights the potential of isoxanthopterin as a noninvasive biomarker in DMD.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Duchenne/enzymology , Xanthine Oxidase/urine , Xanthopterin/urine , Adolescent , Allopurinol/pharmacology , Animals , Biomarkers/urine , Case-Control Studies , Creatinine/urine , Dystrophin/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Oxypurinol/pharmacology , Sarcoglycans/deficiency , Sarcoglycans/genetics , Tyrosine/urine , Utrophin/deficiency , Utrophin/genetics , Xanthine Oxidase/genetics , Young Adult
10.
Nutr Metab Cardiovasc Dis ; 28(11): 1188-1195, 2018 11.
Article in English | MEDLINE | ID: mdl-30143409

ABSTRACT

BACKGROUND AND AIMS: Several studies propose that (-)-epicatechin, a flavonol present in high concentration in the cocoa, has cardioprotective effects. This study aimed to evaluate the impact of (-)-epicatechin on the development of dilated cardiomyopathy in a δ sarcoglycan null mouse model. METHODS AND RESULTS: δ Sarcoglycan null mice were treated for 15 days with (-)-epicatechin. Histological and morphometric analysis of the hearts treated mutant mice showed significant reduction of the vasoconstrictions in the coronary arteries as well as fewer areas with fibrosis and a reduction in the loss of the ventricular wall. On the contrary, it was observed a thickening of this region. By Western blot analysis, it was shown, and increment in the phosphorylation level of eNOS and PI3K/AKT/mTOR/p70S6K proteins in the heart of the (-)-epicatechin treated animals. On the other hand, we observed a significantly decreased level of the atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) heart failure markers. CONCLUSION: All the results indicate that (-)-epicatechin has the potential to prevent the development of dilated cardiomyopathy of genetic origin and encourages the use of this flavonol as a pharmacological therapy for dilated cardiomyopathy and heart failure diseases.


Subject(s)
Cardiomyopathy, Dilated/prevention & control , Catechin/pharmacology , Myocytes, Cardiac/drug effects , Sarcoglycans/deficiency , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Atrial Natriuretic Factor/metabolism , Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Coronary Vessels/drug effects , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Disease Models, Animal , Fibrosis , Male , Mice, Knockout , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sarcoglycans/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Vasoconstriction/drug effects
11.
Cell Death Dis ; 9(1): 1, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29298988

ABSTRACT

Cell engraftment, survival and integration during transplantation procedures represent the crux of cell-based therapies. Thus, there have been many studies focused on improving cell viability upon implantation. We used severe oxidative stress to select for a mouse mesoangioblast subpopulation in vitro and found that this subpopulation retained self-renewal and myogenic differentiation capacities while notably enhancing cell survival, proliferation and migration relative to unselected cells. Additionally, this subpopulation of cells presented different resistance and recovery properties upon oxidative stress treatment, demonstrating select advantages over parental mesoangioblasts in our experimental analysis. Specifically, the cells were resistant to oxidative environments, demonstrating survival, continuous self-renewal and improved migration capability. The primary outcome of the selected cells was determined in in vivo experiments in which immunocompromised dystrophic mice were injected intramuscularly in the tibialis anterior with selected or non-selected mesoangioblasts. Resistant mesoangioblasts exhibited markedly enhanced survival and integration into the host skeletal muscle, accounting for a more than 70% increase in engraftment compared with that of the unselected mesoangioblast cell population and leading to remarkable muscle recovery. Thus, the positive effects of sorting on mesoangioblast cell behaviour in vitro and in vivo suggest that a selection step involving oxidative stress preconditioning may provide a novel methodology to select for resistant cells for use in regenerative tissue applications to prevent high mortality rates upon transplantation.


Subject(s)
Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Animals , Cell Cycle Checkpoints/drug effects , Cell Differentiation , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, SCID , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/therapy , Protein Isoforms/metabolism , Reactive Oxygen Species/metabolism , Sarcoglycans/deficiency , Sarcoglycans/genetics , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Neurol Neurosurg Psychiatry ; 89(1): 72-77, 2018 01.
Article in English | MEDLINE | ID: mdl-28889091

ABSTRACT

OBJECTIVES: To characterise the pattern and spectrum of involvement on muscle MRI in a large cohort of patients with sarcoglycanopathies, which are limb-girdle muscular dystrophies (LGMD2C-2F) caused by mutations in one of the four genes coding for muscle sarcoglycans. METHODS: Lower limb MRI scans of patients with LGMD2C-2F, ranging from severe childhood variants to milder adult-onset forms, were collected in 17 neuromuscular referral centres in Europe and USA. Muscle involvement was evaluated semiquantitatively on T1-weighted images according to a visual score, and the global pattern was assessed as well. RESULTS: Scans from 69 patients were examined (38 LGMD2D, 18 LGMD2C, 12 LGMD2E and 1 LGMD2F). A common pattern of involvement was found in all the analysed scans irrespective of the mutated gene. The most and earliest affected muscles were the thigh adductors, glutei and posterior thigh groups, while lower leg muscles were relatively spared even in advanced disease. A proximodistal gradient of involvement of vasti muscles was a consistent finding in these patients, including the most severe ones. CONCLUSIONS: Muscle involvement on MRI is consistent in patients with LGMD2C-F and can be helpful in distinguishing sarcoglycanopathies from other LGMDs or dystrophinopathies, which represent the most common differential diagnoses. Our data provide evidence about selective susceptibility or resistance to degeneration of specific muscles when one of the sarcoglycans is deficient, as well as preliminary information about progressive involvement of the different muscles over time.


Subject(s)
Magnetic Resonance Imaging/methods , Sarcoglycanopathies/genetics , Sarcoglycans/genetics , Adolescent , Adult , Child , Child, Preschool , Europe , Female , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Mutation , Phenotype , Sarcoglycans/deficiency , United States
13.
J Vasc Res ; 54(4): 235-245, 2017.
Article in English | MEDLINE | ID: mdl-28768281

ABSTRACT

BACKGROUND: Smooth muscle cells are important for atherosclerotic plaque stability. Their proper ability to communicate with the extracellular matrix is crucial for maintaining the correct tissue integrity. In this study, we have investigated the role of ß-sarcoglycan within the matrix-binding dystrophin-glycoprotein complex in the development of atherosclerosis. RESULTS: Atherosclerotic plaque development was significantly reduced in ApoE-deficient mice lacking ß-sarcoglycan, and their plaques contained an increase in differentiated smooth muscle cells. ApoE-deficient mice lacking ß-sarcoglycan showed a reduction in ovarian adipose tissue and adipocyte size, while the total weight of the animals was not significantly different. Western blot analysis of adipose tissues showed a decreased activation of protein kinase B, while that of AMP-activated kinase was increased in mice lacking ß-sarcoglycan. Analysis of plasma in ß-sarcoglycan-deficient mice revealed reduced levels of leptin, adiponectin, insulin, cholesterol, and triglycerides but increased levels of IL-6, IL-17, and TNF-α. CONCLUSIONS: Our results indicate that the dystrophin-glycoprotein complex and ß-sarcoglycan can affect the atherosclerotic process. Furthermore, the results show the effects of ß-sarcoglycan deficiency on adipose tissue and lipid metabolism, which may also have contributed to the atherosclerotic plaque reduction.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Sarcoglycans/deficiency , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipokines/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Dystrophin-Associated Protein Complex/metabolism , Female , Genetic Predisposition to Disease , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Sarcoglycans/genetics
14.
J Clin Invest ; 125(11): 4186-95, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26457733

ABSTRACT

Exon skipping uses antisense oligonucleotides as a treatment for genetic diseases. The antisense oligonucleotides used for exon skipping are designed to bypass premature stop codons in the target RNA and restore reading frame disruption. Exon skipping is currently being tested in humans with dystrophin gene mutations who have Duchenne muscular dystrophy. For Duchenne muscular dystrophy, the rationale for exon skipping derived from observations in patients with naturally occurring dystrophin gene mutations that generated internally deleted but partially functional dystrophin proteins. We have now expanded the potential for exon skipping by testing whether an internal, in-frame truncation of a transmembrane protein γ-sarcoglycan is functional. We generated an internally truncated γ-sarcoglycan protein that we have termed Mini-Gamma by deleting a large portion of the extracellular domain. Mini-Gamma provided functional and pathological benefits to correct the loss of γ-sarcoglycan in a Drosophila model, in heterologous cell expression studies, and in transgenic mice lacking γ-sarcoglycan. We generated a cellular model of human muscle disease and showed that multiple exon skipping could be induced in RNA that encodes a mutant human γ-sarcoglycan. Since Mini-Gamma represents removal of 4 of the 7 coding exons in γ-sarcoglycan, this approach provides a viable strategy to treat the majority of patients with γ-sarcoglycan gene mutations.


Subject(s)
Dystrophin-Associated Protein Complex/chemistry , Genetic Therapy , Muscular Dystrophies, Limb-Girdle/therapy , Oligonucleotides, Antisense/therapeutic use , Protein Engineering , Sarcoglycans/genetics , Animals , Codon, Nonsense/genetics , Diaphragm/metabolism , Diaphragm/pathology , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Exons , Fibrosis , HEK293 Cells , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/therapy , Mutation , Myocardium/metabolism , Myocardium/pathology , Oligonucleotides, Antisense/pharmacology , Protein Interaction Mapping , Protein Structure, Tertiary , RNA, Messenger/chemistry , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Sarcoglycans/biosynthesis , Sarcoglycans/chemistry , Sarcoglycans/deficiency , Sarcolemma/metabolism , Sequence Deletion
15.
Hum Mol Genet ; 23(25): 6903-15, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25106553

ABSTRACT

Muscular dystrophy (MD) is a disease characterized by skeletal muscle necrosis and the progressive accumulation of fibrotic tissue. While transforming growth factor (TGF)-ß has emerged as central effector of MD and fibrotic disease, the cell types in diseased muscle that underlie TGFß-dependent pathology have not been segregated. Here, we generated transgenic mice with myofiber-specific inhibition of TGFß signaling owing to expression of a TGFß type II receptor dominant-negative (dnTGFßRII) truncation mutant. Expression of dnTGFßRII in myofibers mitigated the dystrophic phenotype observed in δ-sarcoglycan-null (Sgcd(-/-)) mice through a mechanism involving reduced myofiber membrane fragility. The dnTGFßRII transgene also reduced muscle injury and improved muscle regeneration after cardiotoxin injury, as well as increased satellite cell numbers and activity. An unbiased global expression analysis revealed a number of potential mechanisms for dnTGFßRII-mediated protection, one of which was induction of the antioxidant protein metallothionein (Mt). Indeed, TGFß directly inhibited Mt gene expression in vitro, the dnTGFßRII transgene conferred protection against reactive oxygen species accumulation in dystrophic muscle and treatment with Mt mimetics protected skeletal muscle upon injury in vivo and improved the membrane stability of dystrophic myofibers. Hence, our results show that the myofibers are central mediators of the deleterious effects associated with TGFß signaling in MD.


Subject(s)
Muscular Dystrophies/genetics , Myofibrils/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Cobra Cardiotoxin Proteins/pharmacology , Crotoxin/pharmacology , Disease Models, Animal , Drug Combinations , Gene Expression Profiling , Gene Expression Regulation , Humans , Metallothionein/genetics , Metallothionein/metabolism , Mice , Mice, Transgenic , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Mutation , Myofibrils/drug effects , Myofibrils/pathology , Protein Serine-Threonine Kinases/deficiency , Reactive Oxygen Species/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Sarcoglycans/deficiency , Sarcoglycans/genetics , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Transforming Growth Factor beta/pharmacology , Transgenes
16.
Hum Mol Genet ; 23(25): 6722-31, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25070948

ABSTRACT

Disruption of the dystrophin complex causes muscle injury, dysfunction, cell death and fibrosis. Excess transforming growth factor (TGF) ß signaling has been described in human muscular dystrophy and animal models, where it is thought to relate to the progressive fibrosis that characterizes dystrophic muscle. We now found that canonical TGFß signaling acutely increases when dystrophic muscle is stimulated to contract. Muscle lacking the dystrophin-associated protein γ-sarcoglycan (Sgcg null) was subjected to a lengthening protocol to produce maximal muscle injury, which produced rapid accumulation of nuclear phosphorylated SMAD2/3. To test whether reducing SMAD signaling improves muscular dystrophy in mice, we introduced a heterozygous mutation of SMAD4 (S4) into Sgcg mice to reduce but not ablate SMAD4. Sgcg/S4 mice had improved body mass compared with Sgcg mice, which normally show a wasting phenotype similar to human muscular dystrophy patients. Sgcg/S4 mice had improved cardiac function as well as improved twitch and tetanic force in skeletal muscle. Functional enhancement in Sgcg/S4 muscle occurred without a reduction in fibrosis, suggesting that intracellular SMAD4 targets may be important. An assessment of genes differentially expressed in Sgcg muscle focused on those encoding calcium-handling proteins and responsive to TGFß since this pathway is a target for mediating improvement in muscular dystrophy. These data demonstrate that excessive TGFß signaling alters cardiac and muscle performance through the intracellular SMAD pathway.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Myocardium/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Body Weight , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation , Heart Function Tests , Humans , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/genetics , Mice , Mice, Knockout , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation , Myocardium/pathology , Phosphorylation , Sarcoglycans/deficiency , Sarcoglycans/genetics , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Transforming Growth Factor beta/genetics
17.
Pediatr Neurol ; 50(6): 640-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24742800

ABSTRACT

BACKGROUND: The sarcoglycan alpha gene, also known as the adhalin gene, is located on chromosome 17q21; mutations in this gene are associated with limb-girdle muscular dystrophy type 2D. We describe two Turkish siblings with findings consistent with limb-girdle muscular dystrophy type 2D. The evaluation excluded a dystrophinopathy, which is the most common form of muscular dystrophy. PATIENTS: Both siblings had very high levels of creatinine phosphokinase and negative molecular tests for deletions and duplications of the dystrophin gene. The older boy presented at 8 years of age with an inability to climb steps and an abnormal gait. His younger brother was 5 years old and had similar symptoms. The muscle biopsy evaluation was performed only in the older brother. RESULTS: The muscle biopsy showed dystrophic features as well as a deficiency in the expression of two different glycoproteins: the alpha sarcoglycan and the gamma sarcoglycan. Sarcolemmal expressions of dystrophin and other sarcoglycans (beta and delta) were diffusely present. DNA analysis demonstrated the presence of previously unknown homozygous mutations [c.226 C > T (p.L76 F)] in exon 3 in the sarcoglycan alpha genes of both siblings. Similar heterozygous point mutations at the same locus were found in both parents, but the genes of beta, delta, and gamma sarcoglycan were normal in the remaining family members. CONCLUSIONS: We describe two siblings with limb-girdle muscular dystrophy type 2D with a novel missense mutation. These patients illustrate that the differential diagnosis of muscular dystrophies is impossible with clinical findings alone. Therefore, a muscle biopsy and DNA analysis remain essential methods for diagnosis of muscle diseases.


Subject(s)
Mutation, Missense , Sarcoglycanopathies/genetics , Sarcoglycanopathies/physiopathology , Sarcoglycans/deficiency , Sarcoglycans/genetics , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , DNA Mutational Analysis , Diagnosis, Differential , Fathers , Humans , Male , Molecular Sequence Data , Mothers , Muscle, Skeletal/pathology , Sarcoglycanopathies/diagnosis , Sarcoglycanopathies/pathology , Siblings , Turkey
18.
Hum Gene Ther ; 25(8): 694-704, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24645914

ABSTRACT

The loss of dystrophin or its associated proteins results in the development of muscle wasting frequently associated with cardiomyopathy. Contractile cardiac tissue is injured and replaced by fibrous tissue or fatty infiltrates, leading to a progressive decrease of the contractile force and finally to end-stage heart failure. At the time symptoms appear, restoration of a functional allele of the causative gene might not be sufficient to prevent disease progression. Alterations in Ca(2+) transport and intracellular calcium levels have been implicated in many types of pathological processes, especially in heart disease. On the basis of a gene transfer strategy, we analyzed the therapeutic efficacy of primary gene correction in a δ-sarcoglycan (δ-SG)-deficient animal model versus gene transfer of the Ca(2+) pump hSERCA2a (human sarco-endoplasmic reticulum calcium ATPase 2a), at a symptomatic stage of heart disease. Our results strongly suggest that restoration of δ-SG at this stage of disease will not lead to improved clinical outcome. However, restoration of proper Ca(2+) handling by means of amplifying SERCA2a expression in the myocardium can lead to functional improvement. Abnormalities in Ca(2+) handling play an important role in disease progression toward heart failure, and increased SERCA2a levels appear to significantly improve cardiac contraction and relaxation. Beneficial effects persist at least over a period of 6 months, and the evolution of cardiac functional parameters paralleled those of normal controls. Furthermore, we demonstrate that a plasmid formulation based on amphiphilic block copolymers can provide a safe and efficient platform for myocardial gene therapies. The use of synthetic formulations for myocardial gene transfer might thus overcome one of the major hurdles linked to viral vectors, that is, repeat administrations.


Subject(s)
Genetic Therapy , Heart Failure/therapy , Sarcoglycans/deficiency , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Animals , Heart Failure/physiopathology , Hemodynamics , Humans , Male , Mesocricetus , Myocardial Contraction , Myocardium/pathology , Organ Specificity , Stroke Volume , Ventricular Pressure
19.
EMBO Mol Med ; 6(2): 239-58, 2014 02.
Article in English | MEDLINE | ID: mdl-24378569

ABSTRACT

Muscular dystrophies are severe genetic diseases for which no efficacious therapies exist. Experimental clinical treatments include intra-arterial administration of vessel-associated stem cells, called mesoangioblasts (MABs). However, one of the limitations of this approach is the relatively low number of cells that engraft the diseased tissue, due, at least in part, to the sub-optimal efficiency of extravasation, whose mechanisms for MAB are unknown. Leukocytes emigrate into the inflamed tissues by crossing endothelial cell-to-cell junctions and junctional proteins direct and control leukocyte diapedesis. Here, we identify the endothelial junctional protein JAM-A as a key regulator of MAB extravasation. We show that JAM-A gene inactivation and JAM-A blocking antibodies strongly enhance MAB engraftment in dystrophic muscle. In the absence of JAM-A, the exchange factors EPAC-1 and 2 are down-regulated, which prevents the activation of the small GTPase Rap-1. As a consequence, junction tightening is reduced, allowing MAB diapedesis. Notably, pharmacological inhibition of Rap-1 increases MAB engraftment in dystrophic muscle, which results into a significant improvement of muscle function offering a novel strategy for stem cell-based therapies.


Subject(s)
Cell Adhesion Molecules/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Muscular Dystrophies/therapy , Receptors, Cell Surface/metabolism , Signal Transduction , Stem Cell Transplantation , Stem Cells/cytology , rap1 GTP-Binding Proteins/metabolism , Animals , Cardiotoxins , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/deficiency , Cell Movement , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Muscular Dystrophies/surgery , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/deficiency , Sarcoglycans/deficiency , Sarcoglycans/metabolism
20.
Exp Physiol ; 99(4): 627-31, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24334334

ABSTRACT

New Findings What is the topic of this review? This symposium report summarizes autonomic, cardiac and skeletal muscle abnormalities in sarcoglycan-δ-deficient mice (Sgcd-/-), a mouse model of limb girdle muscular dystrophy, with emphasis on the roles of autonomic dysregulation and activation of the renin-angiotensin system at a young age. What advances does it highlight? The contributions of the autonomic nervous system and the renin-angiotensin system to the pathogenesis of muscular dystrophy are highlighted. Results demonstrate that autonomic dysregulation precedes and predicts later development of cardiac dysfunction in Sgcd-/- mice and that treatment of young Sgcd-/- mice with the angiotensin type 1 receptor antagonist losartan or with angiotensin-(1-7) abrogates the autonomic dysregulation, attenuates skeletal muscle pathology and increases spontaneous locomotor activity. Muscular dystrophies are a heterogeneous group of genetic muscle diseases characterized by muscle weakness and atrophy. Mutations in sarcoglycans and other subunits of the dystrophin-glycoprotein complex cause muscular dystrophy and dilated cardiomyopathy in animals and humans. Aberrant autonomic signalling is recognized in a variety of neuromuscular disorders. We hypothesized that activation of the renin-angiotensin system contributes to skeletal muscle and autonomic dysfunction in mice deficient in the sarcoglycan-δ (Sgcd) gene at a young age and that this early autonomic dysfunction contributes to the later development of left ventricular (LV) dysfunction and increased mortality. We demonstrated that young Sgcd-/- mice exhibit histopathological features of skeletal muscle dystrophy, decreased locomotor activity and severe autonomic dysregulation, but normal LV function. Autonomic regulation continued to deteriorate in Sgcd-/- mice with age and was accompanied by LV dysfunction and dilated cardiomyopathy at older ages. Autonomic dysregulation at a young age predicted later development of LV dysfunction and higher mortality in Sgcd-/- mice. Treatment of Sgcd-/- mice with the angiotensin type 1 receptor blocker losartan for 8-9 weeks, beginning at 3 weeks of age, decreased fibrosis and oxidative stress in skeletal muscle, increased locomotor activity and prevented autonomic dysfunction. Chronic infusion of the counter-regulatory peptide angiotensin-(1-7) resulted in similar protection. We conclude that activation of the renin-angiotensin system, at a young age, contributes to skeletal muscle and autonomic dysfunction in muscular dystrophy. We speculate that the latter is mediated via abnormal sensory nerve and/or cytokine signalling from dystrophic skeletal muscle to the brain and contributes to age-related LV dysfunction, dilated cardiomyopathy, arrhythmias and premature death. Therefore, correcting the early autonomic dysregulation and renin-angiotensin system activation may provide a novel therapeutic approach in muscular dystrophy.


Subject(s)
Autonomic Nervous System/physiopathology , Cardiomyopathies/physiopathology , Heart/innervation , Motor Activity , Muscle, Skeletal/innervation , Muscular Dystrophies, Limb-Girdle/physiopathology , Renin-Angiotensin System , Ventricular Dysfunction, Left/physiopathology , Angiotensin I/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/metabolism , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Disease Models, Animal , Genotype , Heart/drug effects , Humans , Mice, Knockout , Motor Activity/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/drug therapy , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Myocardium/pathology , Peptide Fragments/pharmacology , Phenotype , Renin-Angiotensin System/drug effects , Sarcoglycans/deficiency , Sarcoglycans/genetics , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Function, Left
SELECTION OF CITATIONS
SEARCH DETAIL
...