Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Exp Cell Res ; 406(2): 112766, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34364881

RESUMEN

Duchene muscular dystrophy leads to progressive muscle structural and functional decline due to chronic degenerative-regenerative cycles. Enhancing the regenerative capacity of dystrophic muscle provides potential therapeutic options. We previously demonstrated that the circadian clock repressor Rev-erbα inhibited myogenesis and Rev-erbα ablation enhanced muscle regeneration. Here we show that Rev-erbα deficiency in the dystrophin-deficient mdx mice promotes regenerative myogenic response to ameliorate muscle damage. Loss of Rev-erbα in mdx mice improved dystrophic pathology and muscle wasting. Rev-erbα-deficient dystrophic muscle exhibit augmented myogenic response, enhanced neo-myofiber formation and attenuated inflammatory response. In mdx myoblasts devoid of Rev-erbα, myogenic differentiation was augmented together with up-regulation of Wnt signaling and proliferative pathways, suggesting that loss of Rev-erbα inhibition of these processes contributed to the improvement in regenerative myogenesis. Collectively, our findings revealed that the loss of Rev-erbα function protects dystrophic muscle from injury by promoting myogenic repair, and inhibition of its activity may have therapeutic utilities for muscular dystrophy.


Asunto(s)
Diferenciación Celular , Músculo Esquelético/citología , Distrofia Muscular Animal/prevención & control , Distrofia Muscular de Duchenne/prevención & control , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Regeneración , Animales , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/etiología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/etiología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Vía de Señalización Wnt
2.
Mol Ther ; 29(3): 1070-1085, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33160075

RESUMEN

Gene editing is often touted as a permanent method for correcting mutations, but its long-term benefits in Duchenne muscular dystrophy (DMD) may depend on sufficiently high editing efficiencies to halt muscle degeneration. Here, we explored the persistence of dystrophin expression following recombinant adeno-associated virus serotype 6 (rAAV6):CRISPR-Cas9-mediated multi-exon deletion/reframing in systemically injected 2- and 11-week-old dystrophic mice and show that induction of low dystrophin levels persists for several months in cardiomyocytes but not in skeletal muscles, where myofibers remain susceptible to necrosis and regeneration. Whereas gene-correction efficiency in both muscle types was enhanced with increased ratios of guide RNA (gRNA)-to-nuclease vectors, obtaining high dystrophin levels in skeletal muscles via multi-exon deletion remained challenging. In contrast, when AAV-microdystrophin was codelivered with editing components, long-term gene-edited dystrophins persisted in both muscle types. These results suggest that the high rate of necrosis and regeneration in skeletal muscles, compared with the relative stability of dystrophic cardiomyocytes, caused the rapid loss of edited genomes. Consequently, stable dystrophin expression in DMD skeletal muscles will require either highly efficient gene editing or the use of cotreatments that decrease skeletal muscle degeneration.


Asunto(s)
Distrofina/genética , Edición Génica , Vectores Genéticos/administración & dosificación , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/prevención & control , Distrofia Muscular de Duchenne/prevención & control , Miocardio/metabolismo , Animales , Sistemas CRISPR-Cas , Dependovirus/genética , Modelos Animales de Enfermedad , Distrofina/metabolismo , Terapia Genética/métodos , Vectores Genéticos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Guía de Kinetoplastida
3.
Mol Ther ; 29(3): 1086-1101, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33221436

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe genetic disorder caused by mutations in the DMD gene. Absence of dystrophin protein leads to progressive degradation of skeletal and cardiac function and leads to premature death. Over the years, zebrafish have been increasingly used for studying DMD and are a powerful tool for drug discovery and therapeutic development. In our study, a birefringence screening assay led to identification of phosphodiesterase 10A (PDE10A) inhibitors that reduced the manifestation of dystrophic muscle phenotype in dystrophin-deficient sapje-like zebrafish larvae. PDE10A has been validated as a therapeutic target by pde10a morpholino-mediated reduction in muscle pathology and improvement in locomotion, muscle, and vascular function as well as long-term survival in sapje-like larvae. PDE10A inhibition in zebrafish and DMD patient-derived myoblasts were also associated with reduction of PITPNA expression that has been previously identified as a protective genetic modifier in two exceptional dystrophin-deficient golden retriever muscular dystrophy (GRMD) dogs that escaped the dystrophic phenotype. The combination of a phenotypic assay and relevant functional assessments in the sapje-like zebrafish enhances the potential for the prospective discovery of DMD therapeutics. Indeed, our results suggest a new application for a PDE10A inhibitor as a potential DMD therapeutic to be investigated in a mouse model of DMD.


Asunto(s)
Distrofina/metabolismo , Distrofia Muscular Animal/prevención & control , Distrofia Muscular de Duchenne/prevención & control , Mioblastos/efectos de los fármacos , Proteínas de Transferencia de Fosfolípidos/antagonistas & inhibidores , Hidrolasas Diéster Fosfóricas/química , Pirazoles/farmacología , Quinolinas/farmacología , Animales , Perros , Distrofina/genética , Humanos , Larva/efectos de los fármacos , Larva/genética , Larva/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Mioblastos/metabolismo , Mioblastos/patología , Proteínas de Transferencia de Fosfolípidos/genética , Proteínas de Transferencia de Fosfolípidos/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Pez Cebra
4.
Exp Cell Res ; 397(1): 112348, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33130178

RESUMEN

The muscle-intrinsic clock machinery is required for the maintenance of muscle growth, remodeling and function. Our previous studies demonstrated that the essential transcription activator of the molecular clock feed-back loop, Brain and Muscle Arnt-Like 1(Bmal1), plays a critical role in myogenic progenitor behavior to promote and regenerative myogenesis. Using genetic approaches targeting Bmal1 in the DMDmdx (mdx) dystrophic mouse model, here we report that the loss of Bmal1 function significantly accelerated dystrophic disease progression. In contrast to the mild dystrophic changes in mdx mice, the genetic loss-of-function of Bmal1 aggravated muscle damage in this dystrophic disease background, as indicated by persistently elevated creatine kinase levels, increased injury area and reduced muscle grip strength. Mechanistic studies revealed that markedly impaired myogenic progenitor proliferation and myogenic response underlie the defective new myofiber formation in the chronic dystrophic milieu. Taken together, our study identified the function of pro-myogenic clock gene Bmal1 in protecting against dystrophic damage, suggesting the potential for augmenting Bmal1 function to ameliorate dystrophic or degenerative muscle diseases.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Modelos Animales de Enfermedad , Desarrollo de Músculos , Músculo Esquelético/citología , Distrofia Muscular Animal/prevención & control , Distrofia Muscular de Duchenne/prevención & control , Regeneración , Factores de Transcripción ARNTL/genética , Animales , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología
5.
Am J Physiol Cell Physiol ; 318(6): C1083-C1091, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32208990

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe, progressive muscle-wasting disorder that leads to early death. The mdx mouse is a naturally occurring mutant model for DMD. It lacks dystrophin and displays peak muscle cell necrosis at ~28 days (D28), but in contrast to DMD, mdx mice experience muscle regeneration by D70. We hypothesized that matrix metalloproteinase-2 (MMP2) and/or MMP9 play key roles in the degeneration/regeneration phases in mdx mice. MMP2 abundance in muscle homogenates, measured by calibrated Western blotting, and activity, measured by zymogram, were lower at D70 compared with D28 in both mdx and wild-type (WT) mice. Importantly, MMP2 abundance was higher in both D28 and D70 mdx mice than in age-matched WT mice. The higher MMP2 abundance was not due to infiltrating macrophages, because MMP2 content was still higher in isolated muscle fibers where most macrophages had been removed. Prenatal supplementation with the amino acid taurine, which improved muscle strength in D28 mdx mice, produced approximately twofold lower MMP2 activity, indicating that increased MMP2 abundance is not required when muscle damage is attenuated. There was no difference in MMP9 abundance between age-matched WT and mdx mice (P > 0.05). WT mice displayed decreased MMP9 abundance as they aged. While MMP9 may have a role during age-related skeletal muscle growth, it does not appear essential for degeneration/regeneration cycles in the mdx mouse. Our findings indicate that MMP2 plays a more active role than MMP9 in the degenerative phases of muscle fibers in D28 mdx mice.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Distrofia Muscular de Duchenne/prevención & control , Efectos Tardíos de la Exposición Prenatal , Taurina/administración & dosificación , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Fuerza Muscular/efectos de los fármacos , Distrofia Muscular de Duchenne/enzimología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Necrosis , Embarazo , Factores de Tiempo , Regulación hacia Arriba
6.
Am J Pathol ; 185(12): 3349-60, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26465071

RESUMEN

Infiltration of immune cells and chronic inflammation substantially affect skeletal and cardiac muscle degeneration in Duchenne muscular dystrophy. In the immune system, extracellular adenosine triphosphate (ATP) released by dying cells is sensed as a danger associated molecular pattern through P2 purinergic receptors. Specifically, the P2X7 subtype has a prominent role in regulating immune system physiology and contributes to inflammasome activation also in muscle cells. Here, we show that in vivo blockade of the extracellular ATP/P2X purinergic signaling pathway by periodate-oxidized ATP delayed the progression of the dystrophic phenotype and dampened the local inflammatory response in mdx mice, a spontaneous mouse model of dystrophin deficiency. Reduced infiltration of leukocytes and macrophages and decreased expression of IL-6 were revealed in the muscles of periodate-oxidized ATP-treated mdx mice. Concomitantly, an increase in Foxp3(+) immunosuppressive regulatory T cells was observed and correlated with enhanced myofiber regeneration. Moreover, we detected reduced concentrations of profibrotic cytokines, including transforming growth factor-ß and connective tissue growth factor, in muscles of periodate-oxidized ATP-treated mdx mice. The improvement of inflammatory features was associated with increased strength and reduced necrosis, thus suggesting that pharmacologic purinergic antagonism altering the adaptive immune component in the muscle infiltrates might represent a promising therapeutic approach in Duchenne muscular dystrophy.


Asunto(s)
Músculo Esquelético/inmunología , Distrofia Muscular de Duchenne/inmunología , Receptores Purinérgicos P2X/fisiología , Linfocitos T Reguladores/inmunología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/farmacología , Adenosina Trifosfato/uso terapéutico , Animales , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/prevención & control , Condicionamiento Físico Animal , Antagonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Receptores Purinérgicos P2X/metabolismo , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos
7.
Muscle Nerve ; 54(1): 79-85, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26599341

RESUMEN

INTRODUCTION: We assessed prophylactic use of bisphosphonate (BP) in Duchenne muscular dystrophy (DMD) patients on glucocorticoid (GC) therapy. METHODS: Fifty-two DMD patients on daily GC were offered BP (oral risedronate). Patients were reviewed for tolerability, side effects, bone pain, and fracture frequency. Bone mineral density (BMD) was determined by annual dual energy X-ray absorptiometry. BP-treated patients were compared with 15 BP-naïve patients (untreated cohort). RESULTS: Side effects occurred in 9 patients. Thirty-six patients continued BP therapy for over 12 months (mean, 3.6 years). Five treated patients reported bone pain. Three treated patients suffered a vertebral fracture, significantly less than in the untreated cohort (5/15). Lumbar spine adjusted BMD Z-scores remained unchanged in treated patients and were significantly greater than in the untreated cohort. CONCLUSIONS: Prophylactic oral risedronate therapy was tolerated by most DMD patients. It appears to maintain BMD and may reduce fracture rate in DMD patients on GC. Muscle Nerve 54: 79-85, 2016.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Distrofia Muscular de Duchenne/prevención & control , Ácido Risedrónico/administración & dosificación , Administración Oral , Adolescente , Densidad Ósea , Niño , Estudios de Cohortes , Femenino , Humanos , Masculino
8.
Am J Pathol ; 178(1): 273-83, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224064

RESUMEN

The disease mechanisms underlying dystrophin-deficient muscular dystrophy are complex, involving not only muscle membrane fragility, but also dysregulated calcium homeostasis. Specifically, it has been proposed that calcium channels directly initiate a cascade of pathological events by allowing calcium ions to enter the cell. The objective of this study was to investigate the effect of chronically blocking calcium channels with the aminoglycoside antibiotic streptomycin from onset of disease in the mdx mouse model of Duchenne muscular dystrophy (DMD). Treatment in utero onwards delayed onset of dystrophic symptoms in the limb muscle of young mdx mice, but did not prevent degeneration and regeneration events occurring later in the disease course. Long-term treatment had a positive effect on limb muscle pathology, reduced fibrosis, increased sarcolemmal stability, and promoted muscle regeneration in older mice. However, streptomycin treatment did not show positive effects in diaphragm or heart muscle, and heart pathology was worsened. Thus, blocking calcium channels even before disease onset does not prevent dystrophy, making this an unlikely treatment for DMD. These findings highlight the importance of analyzing several time points throughout the life of the treated mice, as well as analyzing many tissues, to get a complete picture of treatment efficacy.


Asunto(s)
Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio/metabolismo , Calcio/metabolismo , Corazón/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/prevención & control , Animales , Diafragma/efectos de los fármacos , Diafragma/patología , Humanos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Miocardio/patología , Estreptomicina/uso terapéutico
9.
Mol Med ; 17(9-10): 917-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21607286

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked, lethal, degenerative disease that results from mutations in the dystrophin gene, causing necrosis and inflammation in skeletal muscle tissue. Treatments that reduce muscle fiber destruction and immune cell infiltration can ameliorate DMD pathology. We treated the mdx mouse, a model for DMD, with the immunosuppressant drug rapamycin (RAPA) both locally and systemically to examine its effects on dystrophic mdx muscles. We observed a significant reduction of muscle fiber necrosis in treated mdx mouse tibialis anterior (TA) and diaphragm (Dia) muscles 6 wks post-treatment. This effect was associated with a significant reduction in infiltration of effector CD4(+) and CD8(+) T cells in skeletal muscle tissue, while Foxp3(+) regulatory T cells were preserved. Because RAPA exerts its effects through the mammalian target of RAPA (mTOR), we studied the activation of mTOR in mdx TA and Dia with and without RAPA treatment. Surprisingly, mTOR activation levels in mdx TA were not different from control C57BL/10 (B10). However, mTOR activation was different in Dia between mdx and B10; mTOR activation levels did not rise between 6 and 12 wks of age in mdx Dia muscle, whereas a rise in mTOR activation level was observed in B10 Dia muscle. Furthermore, mdx Dia, but not TA, muscle mTOR activation was responsive to RAPA treatment.


Asunto(s)
Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/prevención & control , Sirolimus/farmacología , Animales , Western Blotting , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diafragma/efectos de los fármacos , Diafragma/metabolismo , Diafragma/patología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/prevención & control , Fosforilación/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo
10.
PLoS One ; 15(6): e0232654, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32559196

RESUMEN

Recently DNA sequencing analysis has played a vital role in the unambiguous diagnosis of clinically suspected patients with Duchenne Muscular Dystrophy (DMD). DMD is a monogenic, X-linked, recessive, degenerative pediatric neuromuscular disorder affecting males, invariably leading to fatal cardiopulmonary failure. Early and precise diagnosis of the disease is an essential part of an effective disease management strategy as care guidelines and prevention through counseling need to be initiated at the earliest particularly since therapies are now available for a subset of patients. In this manuscript we report the DMD gene mutational profiles of 961 clinically suspected male DMD patients, 99% of whom were unrelated. We utilized a molecular diagnostic approach which is cost-effective for most patients and follows a systematic process that sequentially involves identification of hotspot deletions using mPCR, large deletions and duplications using MLPA and small insertions/ deletions and point mutations using an NGS muscular dystrophy gene panel. Pathogenic DMD gene mutations were identified in 84% of patients. Our data compared well with the frequencies and distribution of deletions and duplications reported in the DMD gene in other published studies. We also describe a number of rare in-frame mutations, which appeared to be enriched in the 5' proximal hotspot region of the DMD gene. Furthermore, we identified a family with a rare non-contiguous deletion mutation in the DMD gene where three males were affected and two females were deemed carriers. A subset of patients with mutations in the DMD gene who are likely to benefit therapeutically from new FDA and EMA approved drugs were found in our cohort. Given that the burden of care for DMD patients invariably falls on the mothers, particularly in rural India, effective genetic counseling followed by carrier screening is crucial for prevention of this disorder. We analyzed the carrier status of consented female relatives of 463 probands to gauge the percentage of patients with familial disease. Our analysis revealed 43.7% of mothers with DMD gene mutations. Our comprehensive efforts, involving complete genetic testing coupled with compassionate genetic counseling provided to DMD patients and their families, are intended to improve the quality of life of DMD patients and to empower carrier females to make informed reproductive choices to impede the propagation of this deadly disease.


Asunto(s)
Distrofia Muscular de Duchenne/genética , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Familia , Femenino , Asesoramiento Genético , Heterocigoto , Humanos , Lactante , Recién Nacido , Masculino , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/prevención & control , Distrofia Muscular de Duchenne/terapia , Mutación , Fenotipo , Adulto Joven
11.
J Physiol ; 586(7): 2003-14, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18258657

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe degenerative muscle disease caused by a mutation in the gene encoding dystrophin, a protein linking the cytoskeleton to the extracellular matrix. In this study we investigated whether the antioxidant N-acetylcysteine (NAC) provided protection against dystrophic muscle damage in the mdx mouse, an animal model of DMD. In isolated mdx muscles, NAC prevented the increased membrane permeability and reduced the force deficit associated with stretch-induced muscle damage. Three-week-old mdx mice were treated with NAC in the drinking water for 6 weeks. Dihydroethidium staining showed that NAC treatment reduced the concentration of reactive oxygen species (ROS) in mdx muscles. This was accompanied by a significant decrease in centrally nucleated fibres in muscles from NAC-treated mdx mice. Immunoblotting showed that NAC treatment decreased the nuclear protein expression of NF-kappaB, a transcription factor involved in pro-inflammatory cytokine expression. Finally, we show that NAC treatment reduced caveolin-3 protein levels and increased the sarcolemmal expression of beta-dystroglycan and the dystrophin homologue, utrophin. Taken together, our findings suggest that ROS play an important role in the dystrophic pathogenesis, both in terms of activating damage pathways and in regulating the expression of some dystrophin-associated membrane proteins. These results offer the prospect that antioxidants such as NAC could have therapeutic potential for DMD patients.


Asunto(s)
Acetilcisteína/uso terapéutico , Depuradores de Radicales Libres/uso terapéutico , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/prevención & control , Acetilcisteína/farmacología , Animales , Caveolina 3/metabolismo , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Distrofina/genética , Distrofina/metabolismo , Depuradores de Radicales Libres/farmacología , Ratones , Ratones Endogámicos mdx , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/etiología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/prevención & control , Mutación/genética , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Am Heart J ; 154(3): 596-602, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17719312

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD), an X-linked disorder due to lack of dystrophin, is associated with muscle weakness and myocardial dysfunction. Although preliminary data support the efficacy of angiotensin-converting enzyme inhibitors on left ventricular (LV) function, our aim was to examine the long-term impact of a preventive treatment with perindopril on mortality in children with DMD. METHODS: Patients with DMD between the ages of 9.5 and 13 years presenting with normal LV ejection fraction were included in this prospective study. They were randomly assigned for 3 years to perindopril, 2 to 4 mg (group 1), or placebo (group 2) in a double-blind protocol, followed by open-label treatment with perindopril for up to 10 years. Survival rate at 10 years in each group is reported. RESULTS: There were 28 patients assigned to group 1 and 29 to group 2. Baseline characteristics were similar in both groups. At the end of the 10 years' follow-up period, survival status was available for all included patients: 26 (92.9%) of 28 patients in group 1 were alive at 10 years versus 19 (65.5%) of 29 in group 2 (P = .02). Kaplan-Meier cumulative survival was significantly lower in group 2 than in group 1 (P = .013). CONCLUSION: Early initiation of treatment with perindopril is associated with a lower mortality in patients with DMD with normal LV ejection fraction at study entry.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/mortalidad , Perindopril/uso terapéutico , Adolescente , Niño , Método Doble Ciego , Estudios de Seguimiento , Humanos , Distrofia Muscular de Duchenne/prevención & control , Tasa de Supervivencia , Factores de Tiempo
13.
Redox Rep ; 22(1): 26-34, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26866650

RESUMEN

OBJECTIVE: Duchenne muscular dystrophy (DMD) is a devastating muscle disease caused by a mutation in DMD encoding dystrophin. Oxidative stress accounts for dystrophic muscle pathologies in DMD. We examined the effects of molecular hydrogen in mdx mice, a model animal for DMD. METHODS: The pregnant mother started to take supersaturated hydrogen water (>5 ppm) ad libitum from E15.5 up to weaning of the offspring. The mdx mice took supersaturated hydrogen water from weaning until age 10 or 24 weeks when they were sacrificed. RESULTS: Hydrogen water prevented abnormal body mass gain that is commonly observed in mdx mice. Hydrogen improved the spontaneous running distance that was estimated by a counter-equipped running-wheel, and extended the duration on the rota-rod. Plasma creatine kinase activities were decreased by hydrogen at ages 10 and 24 weeks. Hydrogen also decreased the number of central nuclei of muscle fibers at ages 10 and 24 weeks, and immunostaining for nitrotyrosine in gastrocnemius muscle at age 24 weeks. Additionally, hydrogen tended to increase protein expressions of antioxidant glutathione peroxidase 1, as well as anti-apoptotic Bcl-2, in skeletal muscle at age 10 weeks. DISCUSSION: Although molecular mechanisms of the diverse effects of hydrogen remain to be elucidated, hydrogen potentially improves muscular dystrophy in DMD patients.


Asunto(s)
Hidrógeno/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , Western Blotting , Modelos Animales de Enfermedad , Distrofina/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/prevención & control , Estrés Oxidativo/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Neuromuscul Disord ; 26(12): 860-864, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27863875

RESUMEN

Our study objective was to survey female carriers for Duchenne and Becker muscular dystrophy to identify barriers to carrier testing and the impact of carrier risk knowledge on cardiac and reproductive health management. We surveyed women who have or had biological sons with Duchenne or Becker muscular dystrophy and were enrolled in the US DuchenneConnect patient registry, with questions assessing knowledge of carrier status and recurrence risk, knowledge of care standards for carriers, and barriers to testing. Of the 182 eligible respondents, 25% did not know their carrier status and 14% incorrectly classified themselves as not at risk. Cost of testing was the most commonly identified barrier to testing. Women reporting unknown carrier status were 13 times as likely to express uncertainty regarding their recurrence risk compared to women reporting positive carrier status. 37% of women at an increased risk for cardiomyopathy had never had an echocardiogram. Women who were certain of their positive carrier status were twice as likely to have had an echocardiogram in the last five years compared to women with unknown carrier status. Future research on reducing barriers to counseling and carrier testing, such as cost, may improve care standard adherence.


Asunto(s)
Pruebas Genéticas , Heterocigoto , Madres , Distrofia Muscular de Duchenne/genética , Adolescente , Adulto , Anciano , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/epidemiología , Cardiomiopatías/genética , Estudios Transversales , Femenino , Asesoramiento Genético , Predisposición Genética a la Enfermedad , Pruebas Genéticas/economía , Conocimientos, Actitudes y Práctica en Salud , Humanos , Difusión de la Información , Persona de Mediana Edad , Madres/psicología , Distrofia Muscular de Duchenne/prevención & control , Estudios Prospectivos , Hermanos , Adulto Joven
15.
Biol Trace Elem Res ; 167(1): 115-20, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25762099

RESUMEN

Oxidative stress and inflammatory processes strongly contribute to pathogenesis in Duchenne muscular dystrophy (DMD). Based on evidence that excess iron may increase oxidative stress and contribute to the inflammatory response, we investigated whether deferoxamine (DFX), a potent iron chelating agent, reduces oxidative stress and inflammation in the diaphragm (DIA) muscle of mdx mice (an experimental model of DMD). Fourteen-day-old mdx mice received daily intraperitoneal injections of DFX at a dose of 150 mg/kg body weight, diluted in saline, for 14 days. C57BL/10 and control mdx mice received daily intraperitoneal injections of saline only, for 14 days. Grip strength was evaluated as a functional measure, and blood samples were collected for biochemical assessment of muscle fiber degeneration. In addition, the DIA muscle was removed and processed for histopathology and Western blotting analysis. In mdx mice, DFX reduced muscle damage and loss of muscle strength. DFX treatment also resulted in a significant reduction of dystrophic inflammatory processes, as indicated by decreases in the inflammatory area and in NF-κB levels. DFX significantly decreased oxidative damage, as shown by lower levels of 4-hydroxynonenal and a reduction in dihydroethidium staining in the DIA muscle of mdx mice. The results of the present study suggest that DFX may be useful in therapeutic strategies to ameliorate dystrophic muscle pathology, possibly via mechanisms involving oxidative and inflammatory pathways.


Asunto(s)
Deferoxamina/farmacología , Inflamación/prevención & control , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/prevención & control , Estrés Oxidativo/efectos de los fármacos , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Deferoxamina/administración & dosificación , Diafragma/efectos de los fármacos , Diafragma/metabolismo , Femenino , Inflamación/metabolismo , Inyecciones Intraperitoneales , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/prevención & control , Distrofia Muscular de Duchenne/metabolismo , FN-kappa B/metabolismo
16.
Am J Clin Nutr ; 75(4): 749-53, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11916763

RESUMEN

BACKGROUND: Duchenne muscular dystrophy is a severe X-linked congenital disorder characterized by lethal muscle wasting caused by the absence of the structural protein dystrophin. OBJECTIVE: Because generation of reactive oxygen species appears to play an important role in the pathogenesis of this disease, we tested whether antioxidant green tea extract could diminish muscle necrosis in the mdx mouse dystrophy model. DESIGN: A diet supplemented with 0.01% or 0.05% green tea extract was fed to dams and neonates for 4 wk beginning on the day of birth. Muscle necrosis and regeneration were determined in stained cryosections of soleus and elongator digitorum longus muscles. Radical scavenging by green tea extract was determined in differentiated cultured C2C12 cells treated with tert-butylhydroperoxide, with the use of 2',7'-dichlorofluorescin diacetate as a radical detector. RESULTS: This feeding regimen significantly and dose-dependently reduced necrosis in the fast-twitch muscle elongator digitorum longus but at the doses tested had no effect on the slow-twitch soleus muscle. Green tea extract concentration-dependently decreased oxidative stress induced by tert-butylhydroperoxide treatment of cultured mouse C2C12 myotubes. The lower effective dose tested in mdx mice corresponds to approximately equal to 1.4 L (7 cups) green tea/d in humans. CONCLUSION: Green tea extract may improve muscle health by reducing or delaying necrosis in mdx mice by an antioxidant mechanism.


Asunto(s)
Antioxidantes/uso terapéutico , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/prevención & control , Extractos Vegetales/uso terapéutico , Especies Reactivas de Oxígeno , , Animales , Células Cultivadas , Dieta , Ratones , Músculo Esquelético/efectos de los fármacos , Necrosis
17.
Orv Hetil ; 143(52): 2881-3, 2002 Dec 29.
Artículo en Húngaro | MEDLINE | ID: mdl-12638315

RESUMEN

UNLABELLED: Preimplantation genetic diagnosis is introduced for prevention of genetic disorders. The combination of in vitro fertilization technique and single cell molecular genetic diagnosis allows only unaffected embryos to be selected for embryotransfer, providing a healthy pregnancy and so also avoiding the need for its possible termination. CASE REPORT: The authors report the first successful case of the clinical application of preimplantation genetic diagnosis in Hungary, resulting the birth of an unaffected baby. Preembryonal biopsy and subsequent fluorescent-PCR analysis of the blastomeres taken from the preembryos of a woman who is a carrier for Duchenne muscular dystrophy was performed. Sexing of the preembryos by X and Y chromosome specific primers were performed and three female preembryos were found. RESULT: All three preembryos were transferred into the uterus, which resulted a singleton pregnancy, resulting the birth of a healthy female baby. The authors are offering preimplantation genetic diagnosis with sexing for couples, who are at risk of having a child with X-linked diseases, or are carriers for the delta-F508 mutation of cystic fibrosis. CONCLUSIONS: The application of this diagnostic tool is also planned for other monogenic disorders.


Asunto(s)
Distrofia Muscular de Duchenne/prevención & control , Diagnóstico Preimplantación/métodos , Biopsia , Blastómeros , Femenino , Fluorescencia , Humanos , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/genética , Reacción en Cadena de la Polimerasa/métodos , Embarazo
18.
Rev. méd. Panamá ; 39(3): 100-105, 2019. ilus, tab
Artículo en Español | LILACS | ID: biblio-1100424

RESUMEN

Las distrofias musculares de origen genético son muy diversas y, tanto su diagnóstico preciso como su manejo, suponen un reto importante. En cuanto a este último aspecto, no obstante el desarrollo en proceso de nuevas estrategias a nivel molecular para su tratamiento, las herramientas con que se cuenta para este propósito son limitadas, y pocas veces pueden influir de manera efectiva para evitar el deterioro progresivo que muchos de estos pacientes experimentan. Además, las terapias de última generación no abarcan la gran diversidad de estas patologías y no se espera que estén disponibles a corto plazo para la mayoría de los pacientes. El propósito del artículo es mostrar el papel de las poliaminas, actores ubicuos en el metabolismo in­ tracelular tal vez poco conocidos; cómo están involucrados en los procesos fisiológicos y patológicos, y cómo también pudiesen estar involucrados en la fisiopatología de las distrofias musculares. Su inhi­bición controlada, mediante Difluorometilornitina (DFMO), pudiese constituir un mecanismo para en­ lentecer o eliminar el deterioro muscular de estos pacientes, al utilizarse como una herramienta dentro del arsenal de las ya existentes


Muscular dystrophies of genetic origin are very diverse and, both their precise diagnosis and their management represent an important challenge. Regarding this last aspect, despite the development in process of new strategies at the molecular level for its treatment, the tools available for this pur­ pose are limited, and can rarely influence effectively to avoid the progressive deterioration that many of these patients experience. In addition, the lates t­generation therapies do not cover the great diversity of these pathologies and are not expected to be available in the short term for most patients. The purpose of the article is to show the role of polyamines, ubiquitous actors in intracellular meta­ bolism, perhaps little known; how they are involved in physiological and pathological processes, and how they could also be involved in the physiopathology of muscular dystrophies. Its controlled inhi­ bition, by difluoromethylilitin (DFMO), could be a mechanism to slow or eliminate the muscle deterio­ ration of these patients, by being used as a tool within the arsenal of those already existing.


Asunto(s)
Humanos , Masculino , Femenino , Ornitina/farmacología , Poliaminas/farmacología , Distrofias Musculares/diagnóstico , Poliaminas/química , Compuestos Químicos , Distrofia Muscular de Duchenne/historia , Distrofia Muscular de Duchenne/prevención & control
19.
Science ; 345(6201): 1184-1188, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25123483

RESUMEN

Duchenne muscular dystrophy (DMD) is an inherited X-linked disease caused by mutations in the gene encoding dystrophin, a protein required for muscle fiber integrity. DMD is characterized by progressive muscle weakness and a shortened life span, and there is no effective treatment. We used clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9)-mediated genome editing to correct the dystrophin gene (Dmd) mutation in the germ line of mdx mice, a model for DMD, and then monitored muscle structure and function. Genome editing produced genetically mosaic animals containing 2 to 100% correction of the Dmd gene. The degree of muscle phenotypic rescue in mosaic mice exceeded the efficiency of gene correction, likely reflecting an advantage of the corrected cells and their contribution to regenerating muscle. With the anticipated technological advances that will facilitate genome editing of postnatal somatic cells, this strategy may one day allow correction of disease-causing mutations in the muscle tissue of patients with DMD.


Asunto(s)
Sistemas CRISPR-Cas , Distrofina/genética , Marcación de Gen/métodos , Distrofia Muscular de Duchenne/prevención & control , Animales , ADN/genética , Exones/genética , Terapia Genética/métodos , Células Germinativas , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA