Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
NMR Biomed ; 32(5): e4075, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30848538

RESUMEN

Duchenne Muscular Dystrophy (DMD) is a fatal X-linked genetic disorder. In DMD, the absence of the dystrophin protein causes decreased sarcolemmal integrity resulting in progressive replacement of muscle with fibrofatty tissue. The effects of lacking dystrophin on muscle and systemic metabolism are still unclear. Therefore, to determine the impact of the absence of dystrophin on metabolism, we investigated the metabolic and lipid profile at two different, well-defined stages of muscle damage and stabilization in mdx mice. We measured NMR-detectable metabolite and lipid profiles in the serum and muscles of mdx mice at 6 and 24 weeks of age. Metabolites were determined in muscle in vivo using 1 H MRI/MRS, in isolated muscles using 1 H-HR-MAS NMR, and in serum using high resolution 1 H/13 C NMR. Dystrophic mice were found to have a unique lipid saturation profile compared with control mice, revealing an age-related metabolic change. In the 6-week-old mdx mice, serum lipids were increased and the degree of lipid saturation changed between 6 and 24 weeks. The serum taurine-creatine ratio increased over the life span of mdx, but not in control mice. Furthermore, the saturation index of lipids increased in the serum but decreased in the tissue over time. Finally, we demonstrated associations between MRI-T2 , a strong indicator of inflammation/edema, with tissue and serum lipid profiles. These results indicate the complex temporal changes of metabolites in the tissue and serum during repetitive bouts of muscle damage and regeneration that occur in dystrophic muscle.


Asunto(s)
Envejecimiento/metabolismo , Lípidos/química , Metaboloma , Metabolómica , Distrofia Muscular Animal/metabolismo , Animales , Glucemia/análisis , Espectroscopía de Resonancia Magnética con Carbono-13 , Lípidos/sangre , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Análisis Multivariante , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/sangre , Análisis de Componente Principal
2.
PLoS One ; 14(1): e0211597, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30699200

RESUMEN

MicroRNAs (miRNAs) are non-coding small RNAs that regulate gene expression at the post-transcriptional level. Several miRNAs are exclusively expressed in skeletal muscle and participate in the regulation of muscle differentiation by interacting with myogenic factors. These miRNAs can be found at high levels in the serum of patients and animal models for Duchenne muscular dystrophy, which is expected to be useful as biomarkers for their clinical conditions. By miRNA microarray analysis, we identified miR-188 as a novel miRNA that is elevated in the serum of the muscular dystrophy dog model, CXMDJ. miR-188 was not muscle-specific miRNA, but its expression was up-regulated in skeletal muscles associated with muscle regeneration induced by cardiotoxin-injection in normal dogs and mice. Manipulation of miR-188 expression using antisense oligo and mimic oligo RNAs alters the mRNA expression of the myogenic regulatory factors, MRF4 and MEF2C. Our results suggest that miR-188 is a new player that participates in the gene regulation process of muscle differentiation and that it may serve as a serum biomarker reflecting skeletal muscle regeneration.


Asunto(s)
Biomarcadores/sangre , Regulación de la Expresión Génica , MicroARNs/genética , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Ratones , Músculo Esquelético/patología , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/patología , Mioblastos/citología , Mioblastos/metabolismo
3.
Appl Radiat Isot ; 143: 107-112, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30408633

RESUMEN

The elements Br, Ca, Cl, Cr, Fe, K, Mg, Na, P, Rb, S, and Zn were investigated in the whole blood samples of Golden Retriever dogs submitted to cell therapy (hASCs). These analyses were performed over 2 years using Neutron Activation Analysis and X-Ray Fluorescence techniques. The results were compared with control and untreated dog's. A significant increase was observed in K blood levels. There was also variation in blood levels of Br, Cr, Fe, Rb, S, and Zn.


Asunto(s)
Enfermedades de los Perros/sangre , Enfermedades de los Perros/terapia , Trasplante de Células Madre Mesenquimatosas/veterinaria , Metales/sangre , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/terapia , Animales , Análisis Químico de la Sangre/métodos , Estudios de Casos y Controles , Perros , Humanos , Iones/sangre , Masculino , Análisis de Activación de Neutrones/métodos , Potasio/sangre , Espectrometría por Rayos X/métodos
4.
Pol J Vet Sci ; 20(2): 387-394, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28865216

RESUMEN

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.


Asunto(s)
Trastornos de la Coagulación Sanguínea/veterinaria , Enfermedades de los Bovinos/sangre , Músculo Esquelético/patología , Distrofia Muscular Animal/etiología , Trastornos Nutricionales/veterinaria , Selenio/deficiencia , Animales , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/patología , Bovinos , Enfermedades de los Bovinos/patología , Glutatión Peroxidasa/metabolismo , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/patología , Trastornos Nutricionales/sangre , Trastornos Nutricionales/etiología , Trastornos Nutricionales/patología , Vitamina E/metabolismo
5.
Oxid Med Cell Longev ; 2017: 1987218, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28845212

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked genetic disease in which dystrophin gene is mutated, resulting in dysfunctional or absent dystrophin protein. The pathology of dystrophic muscle includes degeneration, necrosis with inflammatory cell invasion, regeneration, and fibrous and fatty changes. Nevertheless, the mechanisms by which the absence of dystrophin leads to muscle degeneration remain to be fully elucidated. An imbalance between oxidant and antioxidant systems has been proposed as a secondary effect of DMD. However, the significance and precise extent of the perturbation in redox signaling cascades is poorly understood. We report that mdx dystrophic mice are able to activate a compensatory antioxidant response at the presymptomatic stage of the disease. In contrast, increased circulating levels of IL-6 perturb the redox signaling cascade, even prior to the necrotic stage, leading to severe features and progressive nature of muscular dystrophy.


Asunto(s)
Interleucina-6/sangre , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/sangre , Transducción de Señal , Animales , Diafragma/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Necrosis , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
6.
Neuromuscul Disord ; 27(7): 635-645, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28554556

RESUMEN

Enzyme-linked and electrochemiluminescence immunoassays were developed for quantification of amino (N-) terminal fragments of the skeletal muscle protein titin (N-ter titin) and qualified for use in detection of urinary N-ter titin excretion. Urine from normal subjects contained a small but measurable level of N-ter titin (1.0 ± 0.4 ng/ml). A 365-fold increase (365.4 ± 65.0, P = 0.0001) in urinary N-ter titin excretion was seen in Duchene muscular dystrophy (DMD) patients. Urinary N-ter titin was also evaluated in dystrophin deficient rodent models. Mdx mice exhibited low urinary N-ter titin levels at 2 weeks of age followed by a robust and sustained elevation starting at 3 weeks of age, coincident with the development of systemic skeletal muscle damage in this model; fold elevation could not be determined because urinary N-ter titin was not detected in age-matched wild type mice. Levels of serum creatine kinase and serum skeletal muscle troponin I (TnI) were also low at 2 weeks, elevated at later time points and were significantly correlated with urinary N-ter titin excretion in mdx mice. Corticosteroid treatment of mdx mice resulted in improved exercise performance and lowering of both urinary N-ter titin and serum skeletal muscle TnI concentrations. Low urinary N-ter titin levels were detected in wild type rats (3.0 ± 0.6 ng/ml), while Dmdmdx rats exhibited a 556-fold increase (1652.5 ± 405.7 ng/ml, P = 0.002) (both at 5 months of age). These results suggest that urinary N-ter titin is present at low basal concentrations in normal urine and increases dramatically coincident with muscle damage produced by dystrophin deficiency. Urinary N-ter titin has potential as a facile, non-invasive and translational biomarker for DMD.


Asunto(s)
Conectina/orina , Distrofia Muscular de Duchenne/orina , Adolescente , Corticoesteroides/uso terapéutico , Factores de Edad , Animales , Estudios de Casos y Controles , Niño , Preescolar , Conectina/sangre , Creatina Quinasa/sangre , Estudios Transversales , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos mdx , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/orina , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/genética
7.
J Neuromuscul Dis ; 3(2): 247-260, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27854211

RESUMEN

BACKGROUND: Duchenne Muscular Dystrophy (DMD) is a severe, progressive, neuromuscular disorder of childhood. While a number of serum factors have been identified as potential biomarkers of DMD, none, as yet, are proteins within the dystrophin-associated glycoprotein (DAG) complex. OBJECTIVE: We have developed an immobilized serum ELISA assay to measure the expression of a constitutively cleaved and secreted component of the DAG complex, the N-terminal domain of α dystroglycan (αDG-N), and assayed relative expression in serum from muscular dystrophy patients and normal controls. METHODS: ELISAs of immobilized patient or mouse serum and Western blots were used to assess αDG-N expression. RESULTS: Immobilization of diluted serum on ELISA plates was important for this assay, as methods to measure serum αDG-N in solution were less robust. αDG-N ELISA signals were significantly reduced in DMD serum (27±3% decrease, n = 9, p < 0.001) relative to serum from otherwise normal controls (n = 38), and calculated serum αDG-N concentrations were reduced in DMD relative to normal (p < 0.01) and Becker Muscular Dystrophy (n = 11, p < 0.05) patient serum. By contrast, ELISA signals from patients with Inclusion Body Myositis were not different than normal (4±3% decrease, n = 8, p = 0.99). αDG-N serum signals were also significantly reduced in utrophin-deficient mdx mice as compared to mdx and wild type mice. CONCLUSIONS: Our results are the first demonstration of a component of the DAG complex as a potential serum biomarker in DMD. Such a serum measure could be further developed as a tool to help reflect overall muscle DAG complex expression or stability.


Asunto(s)
Distroglicanos/sangre , Distrofia Muscular de Duchenne/sangre , Animales , Biomarcadores/sangre , Western Blotting , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/sangre , Miositis por Cuerpos de Inclusión/sangre , Utrofina/genética
8.
Stem Cell Rev Rep ; 12(4): 448-53, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27193781

RESUMEN

The possibility to treat Duchenne muscular dystrophy (DMD), a lethal X-linked disorder, through cell therapy with mesenchymal stromal cells (MSCs) has been widely investigated in different animal models. However, some crucial questions need to be addressed before starting human therapeutic trials, particularly regarding its use for genetic disorders. How safe is the procedure? Are there any side effects following mesenchymal stem cell transplantation? To address these questions for DMD the best model is the golden retriever muscular dystrophy dog (GRMD), which is the closest model to the human condition displaying a much longer lifespan than other models. Here we report the follow-up of 5 GRMD dogs, which were repeatedly transplanted with human adipose-derived mesenchymal stromal cells (hASC), derived from different donors. Xenogeneic cell transplantation, which was done without immunosuppression, was well tolerated in all animals with no apparent long-term adverse effect. In the present study, we show that repeated heterologous stem-cell injection is a safe procedure, which is fundamental before starting human clinical trials.


Asunto(s)
Tejido Adiposo/citología , Enfermedades de los Perros/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Distrofia Muscular Animal/terapia , Animales , Células Cultivadas , Creatina Quinasa/sangre , Modelos Animales de Enfermedad , Enfermedades de los Perros/sangre , Perros , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Humanos , Inmunocompetencia , Distrofia Muscular Animal/sangre , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/terapia , Reproducibilidad de los Resultados , Factores de Tiempo , Trasplante Heterólogo , Resultado del Tratamiento
9.
Neuromuscul Disord ; 26(1): 73-84, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26494410

RESUMEN

Patients with Duchenne muscular dystrophy are at increased risk of decreased bone mineral density and bone fracture as a result of inactivity. To determine if antiresorptive bisphosphonates could improve bone quality and their effects on muscle we studied the Mdx mouse, treated with pamidronate during peak bone growth at 5 and 6 weeks of age, and examined the outcome at 13 weeks of age. Pamidronate increased cortical bone architecture and strength in femurs with increased resistance to fracture. While overall long bone growth was not affected by pamidronate, there was significant inhibition of remodeling in metaphyseal trabecular bone with evidence of residual calcified cartilage. Pamidronate treatment had positive effects on skeletal muscle in the Mdx mice with decreased serum and muscle creatine kinase and evidence of improved muscle histology and grip strength.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Huesos/efectos de los fármacos , Difosfonatos/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/patología , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Fenómenos Biomecánicos/efectos de los fármacos , Fenómenos Biomecánicos/genética , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Huesos/patología , Huesos/fisiología , Creatina Quinasa/sangre , Difosfonatos/farmacología , Modelos Animales de Enfermedad , Fluoresceínas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/genética , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/genética , Pamidronato , ARN Mensajero/metabolismo
10.
Int J Biochem Cell Biol ; 66: 141-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26239309

RESUMEN

The amino acid taurine is essential for the function of skeletal muscle and administration is proposed as a treatment for Duchenne Muscular Dystrophy (DMD). Taurine homeostasis is dependent on multiple processes including absorption of taurine from food, endogenous synthesis from cysteine and reabsorption in the kidney. This study investigates the cause of reported taurine deficiency in the dystrophic mdx mouse model of DMD. Levels of metabolites (taurine, cysteine, cysteine sulfinate and hypotaurine) and proteins (taurine transporter [TauT], cysteine deoxygenase and cysteine sulfinate dehydrogenase) were quantified in juvenile control C57 and dystrophic mdx mice aged 18 days, 4 and 6 weeks. In C57 mice, taurine content was much higher in both liver and plasma at 18 days, and both cysteine and cysteine deoxygenase were increased. As taurine levels decreased in maturing C57 mice, there was increased transport (reabsorption) of taurine in the kidney and muscle. In mdx mice, taurine and cysteine levels were much lower in liver and plasma at 18 days, and in muscle cysteine was low at 18 days, whereas taurine was lower at 4: these changes were associated with perturbations in taurine transport in liver, kidney and muscle and altered metabolism in liver and kidney. These data suggest that the maintenance of adequate body taurine relies on sufficient dietary intake of taurine and cysteine availability and metabolism, as well as retention of taurine by the kidney. This research indicates dystrophin deficiency not only perturbs taurine metabolism in the muscle but also affects taurine metabolism in the liver and kidney, and supports targeting cysteine and taurine deficiency as a potential therapy for DMD.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Taurina/metabolismo , Animales , Transporte Biológico , Cromatografía Líquida de Alta Presión , Cisteína/análogos & derivados , Cisteína/metabolismo , Modelos Animales de Enfermedad , Distrofina/deficiencia , Distrofina/metabolismo , Humanos , Immunoblotting , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular Animal/sangre , Distrofia Muscular de Duchenne/sangre , Taurina/análogos & derivados , Taurina/sangre , Taurina/deficiencia , Factores de Tiempo
11.
EBioMedicine ; 2(4): 285-93, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26137572

RESUMEN

The anti-inflammatory agents glucocorticoids (GC) are the only available treatment for Duchenne muscular dystrophy (DMD). However, long-term GC treatment causes muscle atrophy and wasting. Thus, targeting specific mediator of inflammatory response may be more specific, more efficacious, and with fewer side effects. The pro-inflammatory cytokine interleukin (IL) 6 is overproduced in patients with DMD and in the muscle of mdx, the animal model for human DMD. We tested the ability of inhibition of IL6 activity, using an interleukin-6 receptor (Il6r) neutralizing antibody, to ameliorate the dystrophic phenotype. Blockade of endogenous Il6r conferred on dystrophic muscle resistance to degeneration and alleviated both morphological and functional consequences of the primary genetic defect. Pharmacological inhibition of IL6 activity leaded to changes in the dystrophic muscle environment, favoring anti-inflammatory responses and improvement in muscle repair. This resulted in a functional homeostatic maintenance of dystrophic muscle. These data provide an alternative pharmacological strategy for treatment of DMD and circumvent the major problems associated with conventional therapy.


Asunto(s)
Músculos/patología , Músculos/fisiopatología , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Receptores de Interleucina-6/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Homeostasis , Inflamación/complicaciones , Inflamación/patología , Interleucina-6/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Distrofia Muscular Animal/sangre , Distrofia Muscular de Duchenne/sangre , Necrosis , Fenotipo , Receptores de Interleucina-6/metabolismo
12.
PLoS One ; 10(5): e0126585, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26010907

RESUMEN

Golden retriever muscular dystrophy (GRMD) provides the best animal model for characterizing the disease progress of the human disorder, Duchenne muscular dystrophy (DMD). The purpose of this study was to determine steroid hormone concentration profiles in healthy golden retriever dogs (control group - CtGR) versus GRMD-gene carrier (CaGR) and affected female dogs (AfCR). Therefore, a sensitive and specific analytical method was developed and validated to determine the estradiol, progesterone, cortisol, and testosterone levels in the canine serum by isotope dilution liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). To more accurately understand the dynamic nature of the serum steroid profile, the fluctuating levels of these four steroid hormones over the estrous cycle were compared across the three experimental groups using a multivariate statistical analysis. The concentration profiles of estradiol, cortisol, progesterone, and testosterone revealed a characteristic pattern for each studied group at each specific estrous phase. Additionally, several important changes in the serum concentrations of cortisol and estradiol in the CaGR and AfCR groups seem to be correlated with the status and progression of the muscular dystrophy. A comprehensive and quantitative monitoring of steroid profiles throughout the estrous cycle of normal and GRMD dogs were achieved. Significant differences in these profiles were observed between GRMD and healthy animals, most notably for estradiol. These findings contribute to a better understanding of both dog reproduction and the muscular dystrophy pathology. Our data open new venues for hormonal behavior studies in dystrophinopathies and that may affect the quality of life of DMD patients.


Asunto(s)
Enfermedades de los Perros/sangre , Enfermedades Genéticas Ligadas al Cromosoma X/sangre , Hormonas/sangre , Marcaje Isotópico/métodos , Distrofia Muscular Animal/sangre , Esteroides/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Cromatografía Liquida , Análisis Discriminante , Perros , Femenino , Análisis de Componente Principal , Reproducibilidad de los Resultados
13.
Hum Mol Genet ; 23(24): 6458-69, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027324

RESUMEN

It is expected that serum protein biomarkers in Duchenne muscular dystrophy (DMD) will reflect disease pathogenesis, progression and aid future therapy developments. Here, we describe use of quantitative in vivo stable isotope labeling in mammals to accurately compare serum proteomes of wild-type and dystrophin-deficient mdx mice. Biomarkers identified in serum from two independent dystrophin-deficient mouse models (mdx-Δ52 and mdx-23) were concordant with those identified in sera samples of DMD patients. Of the 355 mouse sera proteins, 23 were significantly elevated and 4 significantly lower in mdx relative to wild-type mice (P-value < 0.001). Elevated proteins were mostly of muscle origin: including myofibrillar proteins (titin, myosin light chain 1/3, myomesin 3 and filamin-C), glycolytic enzymes (aldolase, phosphoglycerate mutase 2, beta enolase and glycogen phosphorylase), transport proteins (fatty acid-binding protein, myoglobin and somatic cytochrome-C) and others (creatine kinase M, malate dehydrogenase cytosolic, fibrinogen and parvalbumin). Decreased proteins, mostly of extracellular origin, included adiponectin, lumican, plasminogen and leukemia inhibitory factor receptor. Analysis of sera from 1 week to 7 months old mdx mice revealed age-dependent changes in the level of these biomarkers with most biomarkers acutely elevated at 3 weeks of age. Serum analysis of DMD patients, with ages ranging from 4 to 15 years old, confirmed elevation of 20 of the murine biomarkers in DMD, with similar age-related changes. This study provides a panel of biomarkers that reflect muscle activity and pathogenesis and should prove valuable tool to complement natural history studies and to monitor treatment efficacy in future clinical trials.


Asunto(s)
Envejecimiento/sangre , Proteínas Sanguíneas/metabolismo , Distrofina/deficiencia , Distrofia Muscular Animal/sangre , Distrofia Muscular de Duchenne/sangre , Adolescente , Envejecimiento/genética , Envejecimiento/patología , Animales , Biomarcadores/sangre , Proteínas Sanguíneas/genética , Niño , Preescolar , Análisis por Conglomerados , Distrofina/genética , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Anotación de Secuencia Molecular , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Especificidad de la Especie
14.
J Am Anim Hosp Assoc ; 50(2): 130-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24446404

RESUMEN

This report describes a case of feline dystrophin-deficient muscular dystrophy (DDMD) with an atypical clinical presentation. A novel gene mutation is reported to be responsible for dystrophin-deficient hypertrophic muscular dystrophy. In an emergency setting, clinicians should be aware of muscular dystrophy in young cats and the importance of elevated creatine kinase (CK) activity. Muscular dystrophy is rare but can present both a diagnostic and therapeutic challenge in an emergency setting. Patients with muscular dystrophy have a progressive disease with no specific treatment and have an increased risk for death during their hospital stay.


Asunto(s)
Distrofia Muscular Animal/diagnóstico , Animales , Gatos , Creatina Quinasa/sangre , Diagnóstico Diferencial , Servicio de Urgencia en Hospital , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/patología
15.
Neuromuscul Disord ; 24(1): 63-73, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24295812

RESUMEN

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.


Asunto(s)
Distrofia Muscular Animal/fisiopatología , Trastornos Respiratorios/fisiopatología , Animales , Análisis de los Gases de la Sangre , Progresión de la Enfermedad , Perros , Femenino , Frecuencia Cardíaca , Masculino , Distrofia Muscular Animal/sangre , Trastornos Respiratorios/sangre , Frecuencia Respiratoria
16.
PLoS One ; 8(7): e69567, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23922741

RESUMEN

Duchenne Muscular Dystrophy (DMD) is caused by mutations in the DMD gene resulting in an absence of dystrophin in neurons and muscle. Respiratory failure is the most common cause of mortality and previous studies have largely concentrated on diaphragmatic muscle necrosis and respiratory failure component. Here, we investigated the integrity of respiratory control mechanisms in the mdx mouse model of DMD. Whole body plethysmograph in parallel with phrenic nerve activity recordings revealed a lower respiratory rate and minute ventilation during normoxia and a blunting of the hypoxic ventilatory reflex in response to mild levels of hypoxia together with a poor performance on a hypoxic stress test in mdx mice. Arterial blood gas analysis revealed low PaO2 and pH and high PaCO2 in mdx mice. To investigate chemosensory respiratory drive, we analyzed the carotid body by molecular and functional means. Dystrophin mRNA and protein was expressed in normal mice carotid bodies however, they are absent in mdx mice. Functional analysis revealed abnormalities in Dejours test and the early component of the hypercapnic ventilatory reflex in mdx mice. Together, these results demonstrate a malfunction in the peripheral chemosensory drive that would be predicted to contribute to the respiratory failure in mdx mice. These data suggest that investigating and monitoring peripheral chemosensory drive function may be useful for improving the management of DMD patients with respiratory failure.


Asunto(s)
Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/fisiopatología , Animales , Dióxido de Carbono/sangre , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos mdx , Oxígeno/sangre , Presión Parcial
17.
Am J Pathol ; 182(2): 485-504, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23332367

RESUMEN

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


Asunto(s)
Antineoplásicos/uso terapéutico , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/patología , Tamoxifeno/uso terapéutico , Animales , Antineoplásicos/farmacología , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Fenómenos Biomecánicos/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Creatina Quinasa/sangre , Diafragma/patología , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Conducta Alimentaria/efectos de los fármacos , Fibrosis , Ratones , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/fisiopatología , Miocardio/patología , Tamaño de los Órganos/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Tamoxifeno/sangre , Tamoxifeno/farmacología
18.
PLoS One ; 7(11): e48478, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23185260

RESUMEN

The GRMD (Golden retriever muscular dystrophy) dog has been widely used in pre-clinical trials targeting DMD (Duchenne muscular dystrophy), using in many cases a concurrent immune-suppressive treatment. The aim of this study is to assess if such a treatment could have an effect on the disease course of these animals. Seven GRMD dogs were treated with an association of cyclosporine A (immunosuppressive dosage) and prednisolone (2 mg/kg/d) during 7 months, from 2 to 9 months of age. A multi-parametric evaluation was performed during this period which allowed us to demonstrate that this treatment had several significant effects on the disease progression. The gait quality as assessed by 3D-accelerometry was dramatically improved. This was consistent with the evolution of other parameters towards a significant improvement, such as the clinical motor score, the post-tetanic relaxation and the serum CK levels. In contrast the isometric force measurement as well as the histological evaluation argued in favor of a more severe disease progression. In view of the disease modifying effects which have been observed in this study it should be concluded that immunosuppressive treatments should be used with caution when carrying out pre-clinical studies in this canine model of DMD. They also highlight the importance of using a large range of multi-parametric evaluation tools to reliably draw any conclusion from trials involving dystrophin-deficient dogs, which reproduce the complexity of the human disease.


Asunto(s)
Inmunosupresores/uso terapéutico , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Acelerometría , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Creatina Quinasa/sangre , Ciclosporina/farmacología , Ciclosporina/uso terapéutico , Modelos Animales de Enfermedad , Perros , Estudios de Seguimiento , Marcha/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Actividad Motora/efectos de los fármacos , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/complicaciones , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/fisiopatología , Análisis de Componente Principal , Tetania/sangre , Tetania/complicaciones , Tetania/fisiopatología
19.
Am J Pathol ; 181(2): 392-400, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22683468

RESUMEN

Exon skipping is capable of correcting frameshift and nonsense mutations in Duchenne muscular dystrophy. Phase 2 clinical trials in the United Kingdom and the Netherlands have reported induction of dystrophin expression in muscle of Duchenne muscular dystrophy patients by systemic administration of both phosphorodiamidate morpholino oligomers (PMO) and 2'-O-methyl phosphorothioate. Peptide-conjugated phosphorodiamidate morpholino offers significantly higher efficiency than phosphorodiamidate morpholino, with the ability to induce near-normal levels of dystrophin, and restores function in both skeletal and cardiac muscle. We examined 1-year systemic efficacy of peptide-conjugated phosphorodiamidate morpholino targeting exon 23 in dystrophic mdx mice. The LD(50) of peptide-conjugated phosphorodiamidate morpholino was determined to be approximately 85 mg/kg. The half-life of dystrophin expression was approximately 2 months in skeletal muscle, but shorter in cardiac muscle. Biweekly injection of 6 mg/kg peptide-conjugated phosphorodiamidate morpholino produced >20% dystrophin expression in all skeletal muscles and ≤5% in cardiac muscle, with improvement in muscle function and pathology and reduction in levels of serum creatine kinase. Monthly injections of 30 mg/kg peptide-conjugated phosphorodiamidate morpholino restored dystrophin to >50% normal levels in skeletal muscle, and 15% in cardiac muscle. This was associated with greatly reduced serum creatine kinase levels, near-normal histology, and functional improvement of skeletal muscle. Our results demonstrate for the first time that regular 1-year administration of peptide-conjugated phosphorodiamidate morpholino can be safely applied to achieve significant therapeutic effects in an animal model.


Asunto(s)
Distrofina/metabolismo , Morfolinos/uso terapéutico , Músculos/patología , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/fisiopatología , Péptidos/uso terapéutico , Recuperación de la Función/fisiología , Administración Intravenosa , Animales , Esquema de Medicación , Semivida , Dosificación Letal Mediana , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Morfolinos/administración & dosificación , Morfolinos/farmacología , Músculos/efectos de los fármacos , Músculos/fisiopatología , Distrofia Muscular Animal/sangre , Péptidos/administración & dosificación , Péptidos/farmacología , Factores de Tiempo
20.
Hum Mol Genet ; 20(24): 4978-90, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21949353

RESUMEN

Dp116 is a non-muscle isoform of dystrophin that assembles the dystrophin-glycoprotein complex (DGC), but lacks actin-binding domains. To examine the functional role of the DGC, we expressed the Dp116 transgene in mice lacking both dystrophin and utrophin (mdx:utrn(-/-)). Unexpectedly, expression of Dp116 prevented the most severe aspects of the mdx:utrn(-/-) phenotype. Dp116:mdx:utrn(-/-) transgenic mice had dramatic improvements in growth, mobility and lifespan compared with controls. This was associated with increased muscle mass and force generating capacity of limb muscles, although myofiber size and specific force were unchanged. Conversely, Dp116 had no effect on dystrophic injury as determined by muscle histopathology and serum creatine kinase levels. Dp116 also failed to restore normal fiber-type distribution or the post-synaptic architecture of the neuromuscular junction. These data demonstrate that the DGC is critical for growth and maintenance of muscle mass, a function that is independent of the ability to prevent dystrophic pathophysiology. Likewise, this is the first demonstration in skeletal muscle of a positive functional role for a dystrophin protein that lacks actin-binding domains. We conclude that both mechanical and non-mechanical functions of dystrophin are important for its role in skeletal muscle.


Asunto(s)
Distrofina/metabolismo , Longevidad , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/prevención & control , Animales , Fenómenos Biomecánicos , Creatina Quinasa/sangre , Distrofina/química , Esófago/patología , Femenino , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Contracción Muscular , Músculo Esquelético/ultraestructura , Distrofia Muscular Animal/sangre , Distrofia Muscular Animal/fisiopatología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Unión Neuromuscular/ultraestructura , Tamaño de los Órganos , Isoformas de Proteínas/metabolismo , Análisis de Supervivencia , Utrofina/deficiencia , Utrofina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA