Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 28(2): 382-393, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31784415

RESUMEN

Multiple clinical trials employing recombinant adeno-associated viral (rAAV) vectors have been initiated for neuromuscular disorders, including Duchenne and limb-girdle muscular dystrophies, spinal muscular atrophy, and recently X-linked myotubular myopathy (XLMTM). Our previous work on a canine model of XLMTM showed that a single rAAV8-cMTM1 systemic infusion corrected structural abnormalities within the muscle and restored contractile function, with affected dogs surviving more than 4 years post injection. This remarkable therapeutic efficacy presents a unique opportunity to identify the downstream molecular drivers of XLMTM pathology and to what extent the whole muscle transcriptome is restored to normal after gene transfer. Herein, RNA-sequencing was used to examine the transcriptomes of the Biceps femoris and Vastus lateralis in a previously described canine cohort that showed dose-dependent clinical improvements after rAAV8-cMTM1 gene transfer. Our analysis confirmed several dysregulated genes previously observed in XLMTM mice but also identified transcripts linked to XLMTM pathology. We demonstrated XLMTM transcriptome remodeling and dose-dependent normalization of gene expression after gene transfer and created metrics to pinpoint potential biomarkers of disease progression and correction.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/genética , Transcriptoma , Animales , Biomarcadores , Modelos Animales de Enfermedad , Perros , Dosificación de Gen , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Transducción Genética
2.
Mol Ther ; 25(4): 839-854, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237839

RESUMEN

X-linked myotubular myopathy (XLMTM) results from MTM1 gene mutations and myotubularin deficiency. Most XLMTM patients develop severe muscle weakness leading to respiratory failure and death, typically within 2 years of age. Our objective was to evaluate the efficacy and safety of systemic gene therapy in the p.N155K canine model of XLMTM by performing a dose escalation study. A recombinant adeno-associated virus serotype 8 (rAAV8) vector expressing canine myotubularin (cMTM1) under the muscle-specific desmin promoter (rAAV8-cMTM1) was administered by simple peripheral venous infusion in XLMTM dogs at 10 weeks of age, when signs of the disease are already present. A comprehensive analysis of survival, limb strength, gait, respiratory function, neurological assessment, histology, vector biodistribution, transgene expression, and immune response was performed over a 9-month study period. Results indicate that systemic gene therapy was well tolerated, prolonged lifespan, and corrected the skeletal musculature throughout the body in a dose-dependent manner, defining an efficacious dose in this large-animal model of the disease. These results support the development of gene therapy clinical trials for XLMTM.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/genética , Animales , Biopsia , Dependovirus/clasificación , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perros , Marcha , Expresión Génica , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/farmacocinética , Inmunidad Celular , Inmunidad Humoral , Estimación de Kaplan-Meier , Fuerza Muscular , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Músculo Esquelético/ultraestructura , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/mortalidad , Miopatías Estructurales Congénitas/terapia , Proteínas Tirosina Fosfatasas no Receptoras/genética , Recuperación de la Función , Reflejo , Pruebas de Función Respiratoria , Distribución Tisular , Transgenes/genética , Transgenes/inmunología , Resultado del Tratamiento
3.
Muscle Nerve ; 56(5): 943-953, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28370029

RESUMEN

INTRODUCTION: X-linked myotubular myopathy (XLMTM), a devastating pediatric disease caused by the absence of the protein myotubularin, results from mutations in the MTM1 gene. While there is no cure for XLMTM, we previously reported effects of MTM1 gene therapy using adeno-associated virus (AAV) vector on muscle weakness and pathology in MTM1-mutant dogs. Here, we followed 2 AAV-infused dogs over 4 years. METHODS: We evaluated gait, strength, respiration, neurological function, muscle pathology, AAV vector copy number (VCN), and transgene expression. RESULTS: Four years following AAV-mediated gene therapy, gait, respiratory performance, neurological function and pathology in AAV-infused XLMTM dogs remained comparable to their healthy littermate controls despite a decline in VCN and muscle strength. CONCLUSIONS: AAV-mediated gene transfer of MTM1 in young XLMTM dogs results in long-term expression of myotubularin transgene with normal muscular performance and neurological function in the absence of muscle pathology. These findings support a clinical trial in patients. Muscle Nerve 56: 943-953, 2017.


Asunto(s)
Terapia Genética , Miopatías Estructurales Congénitas/terapia , Proteínas Tirosina Fosfatasas no Receptoras/uso terapéutico , Adenosina Trifosfatasas/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Femenino , Trastornos Neurológicos de la Marcha/etiología , Glucuronidasa/genética , Glucuronidasa/metabolismo , Humanos , Estudios Longitudinales , Microscopía Electrónica , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Mutación/genética , Miopatías Estructurales Congénitas/complicaciones , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/veterinaria , NAD/metabolismo , Examen Neurológico , Proteínas Tirosina Fosfatasas no Receptoras/genética , Trastornos Respiratorios/etiología , Transducción Genética
4.
Methods ; 99: 91-8, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26454198

RESUMEN

Current treatment options for patients with monogenetic congenital myopathies (MCM) ameliorate the symptoms of the disorder without resolving the underlying cause. However, gene therapies are being developed where the mutated or deficient gene target is replaced. Preclinical findings in animal models appear promising, as illustrated by gene replacement for X-linked myotubular myopathy (XLMTM) in canine and murine models. Prospective applications and approaches to gene replacement therapy, using these disorders as examples, are discussed in this review.


Asunto(s)
Terapia Genética , Miopatías Estructurales Congénitas/terapia , Animales , Técnicas de Transferencia de Gen , Humanos , Proteínas Musculares/genética , Miopatías Estructurales Congénitas/genética
5.
Muscle Nerve ; 54(6): 1007-1009, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27554703

RESUMEN

INTRODUCTION: The aim of this consensus statement is to provide a recommendation from AANEM experts on the clinical utility of genetic testing. It is not meant to recommend or endorse any specific genetic testing methodology or algorithm. METHODS: The AANEM Professional Practice Committee reached a consensus based on expert opinion on the utility of genetic testing in neuromuscular diseases and made recommendations on factors that physicians and patients should consider when deciding whether to proceed with such testing. RESULTS: Despite the costs of genetic testing, these tests can be both valuable and beneficial in the diagnosis and treatment of neuromuscular diseases in many situations. CONCLUSIONS: The AANEM believes that performing genetic testing to arrive at a specific molecular diagnosis is a critical step in providing high-quality care to neuromuscular patients. The cost of testing should not be a deterrent, as there are important clinical, safety, psychosocial, and research benefits. Muscle Nerve 54: 1007-1009, 2016.


Asunto(s)
Consenso , Pruebas Genéticas , Enfermedades Neuromusculares/diagnóstico , Enfermedades Neuromusculares/genética , Humanos , Sociedades Médicas/normas , Estados Unidos
6.
Hum Mol Genet ; 22(8): 1525-38, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23307925

RESUMEN

No effective treatment exists for patients with X-linked myotubular myopathy (XLMTM), a fatal congenital muscle disease caused by deficiency of the lipid phosphatase, myotubularin. The Mtm1δ4 and Mtm1 p.R69C mice model severely and moderately symptomatic XLMTM, respectively, due to differences in the degree of myotubularin deficiency. Contractile function of intact extensor digitorum longus (EDL) and soleus muscles from Mtm1δ4 mice, which produce no myotubularin, is markedly impaired. Contractile forces generated by chemically skinned single fiber preparations from Mtm1δ4 muscle were largely preserved, indicating that weakness was largely due to impaired excitation contraction coupling. Mtm1 p.R69C mice, which produce small amounts of myotubularin, showed impaired contractile function only in EDL muscles. Short-term replacement of myotubularin with a prototypical targeted protein replacement agent (3E10Fv-MTM1) in Mtm1δ4 mice improved contractile function and muscle pathology. These promising findings suggest that even low levels of myotubularin protein replacement can improve the muscle weakness and reverse the pathology that characterizes XLMTM.


Asunto(s)
Terapia de Reemplazo Enzimático , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Modelos Animales de Enfermedad , Fatiga/metabolismo , Fatiga/fisiopatología , Femenino , Humanos , Ratones , Debilidad Muscular/genética , Debilidad Muscular/terapia , Músculo Esquelético/fisiopatología , Músculos/enzimología , Músculos/metabolismo , Músculos/patología , Miopatías Estructurales Congénitas/enzimología , Miopatías Estructurales Congénitas/genética , Proteínas Tirosina Fosfatasas no Receptoras/biosíntesis , Proteínas Tirosina Fosfatasas no Receptoras/deficiencia
7.
Muscle Nerve ; 51(2): 287-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25354257

RESUMEN

INTRODUCTION: We describe a unique method that combines ultrasound and electromyography to guide intramuscular diaphragm injections in anesthetized large animals. METHODS: Ultrasound was used to visualize the diaphragm on each side of spontaneously breathing, anesthetized beagle dogs and cynomolgus macaques. An electromyography (EMG) needle was introduced and directed by ultrasound to confirm that the needle entered the muscular portion of the diaphragm, and methylene blue was injected. Injection accuracy was confirmed upon necropsy by tracking the spread of methylene blue. RESULTS: All methylene blue injections were confirmed to have been placed appropriately into the diaphragm. CONCLUSIONS: This study demonstrates the feasibility and accuracy of using ultrasound and EMG to guide injections and to reduce complications associated with conventional blind techniques. Ultrasound guidance can be used for clinical EMG of the diaphragm. Future applications may include targeted diaphragm injections with gene replacement therapy in neuromuscular diseases.


Asunto(s)
Diafragma/diagnóstico por imagen , Diafragma/fisiología , Electromiografía , Inyecciones Intramusculares/métodos , Inyecciones Intramusculares/normas , Ultrasonografía , Animales , Perros , Macaca fascicularis
8.
FASEB J ; 27(4): 1585-99, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23325319

RESUMEN

Mutations in the human SEPN1 gene, encoding selenoprotein N (SepN), cause SEPN1-related myopathy (SEPN1-RM) characterized by muscle weakness, spinal rigidity, and respiratory insufficiency. As with other members of the selenoprotein family, selenoprotein N incorporates selenium in the form of selenocysteine (Sec). Most selenoproteins that have been functionally characterized are involved in oxidation-reduction (redox) reactions, with the Sec residue located at their catalytic site. To model SEPN1-RM, we generated a Sepn1-knockout (Sepn1(-/-)) mouse line. Homozygous Sepn1(-/-) mice are fertile, and their weight and lifespan are comparable to wild-type (WT) animals. Under baseline conditions, the muscle histology of Sepn1(-/-) mice remains normal, but subtle core lesions could be detected in skeletal muscle after inducing oxidative stress. Ryanodine receptor (RyR) calcium release channels showed lower sensitivity to caffeine in SepN deficient myofibers, suggesting a possible role of SepN in RyR regulation. SepN deficiency also leads to abnormal lung development characterized by enlarged alveoli, which is associated with decreased tissue elastance and increased quasi-static compliance of Sepn1(-/-) lungs. This finding raises the possibility that the respiratory syndrome observed in patients with SEPN1 mutations may have a primary pulmonary component in addition to the weakness of respiratory muscles.


Asunto(s)
Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Enfermedades Musculares/genética , Selenoproteínas/deficiencia , Animales , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación/genética , Estrés Oxidativo/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Selenocisteína/genética , Selenoproteínas/metabolismo
9.
Muscle Nerve ; 50(3): 315-26, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24668768

RESUMEN

The centronuclear myopathies (CNMs) are a group of inherited neuromuscular disorders classified as congenital myopathies. While several causative genes have been identified, some patients do not harbor any of the currently known mutations. These diverse disorders have common histological features, which include a high proportion of centrally nucleated muscle fibers, and clinical attributes of muscle weakness and respiratory insufficiency. Respiratory problems in CNMs may manifest initially during sleep, but daytime symptoms, ineffective airway clearance, and hypoventilation predominate as more severe respiratory muscle dysfunction evolves. Respiratory muscle capacity can be evaluated using a variety of clinical tests selected with consideration for the age and baseline motor function of the patient. Similar clinical tests of respiratory function can also be incorporated into preclinical CNM canine models to offer insight for clinical trials. Because respiratory problems account for significant morbidity in patients, routine assessments of respiratory muscle function are discussed.


Asunto(s)
Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/fisiopatología , Pruebas de Función Respiratoria/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Debilidad Muscular/patología , Músculo Esquelético/fisiopatología , Miopatías Estructurales Congénitas/genética , Trastornos Respiratorios/etiología , Respiración Artificial
10.
Muscle Nerve ; 50(4): 607-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24861988

RESUMEN

INTRODUCTION: We tested the feasibility of using neuromuscular ultrasound for non-invasive real-time assessment of diaphragmatic structure and function in a canine model of X-linked myotubular myopathy (XLMTM). METHODS: Ultrasound images in 3 dogs [wild-type (WT), n=1; XLMTM untreated, n=1; XLMTM post-AAV8-mediated MTM1 gene replacement, n=1] were analyzed for diaphragm thickness, change in thickness with respiration, muscle echogenicity, and diaphragm excursion amplitude during spontaneous breathing. RESULTS: Quantitative parameters of diaphragm structure were different among the animals. WT diaphragm was thicker and less echogenic than the XLMTM control, whereas the diaphragm measurements of the MTM1-treated XLMTM dog were comparable to those of the WT dog. CONCLUSIONS: This pilot study demonstrates the feasibility of using ultrasound for quantitative assessment of the diaphragm in a canine model. In the future, ultrasonography may replace invasive measures of diaphragm function in canine models and in humans for non-invasive respiratory monitoring and evaluation of neuromuscular disease.


Asunto(s)
Diafragma/diagnóstico por imagen , Diafragma/fisiopatología , Miopatías Estructurales Congénitas/patología , Animales , Diafragma/patología , Modelos Animales de Enfermedad , Perros , Masculino , Miopatías Estructurales Congénitas/veterinaria , Proyectos Piloto , Ultrasonografía
11.
Proc Natl Acad Sci U S A ; 107(33): 14697-702, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20682747

RESUMEN

Mutations in the MTM1 gene encoding myotubularin cause X-linked myotubular myopathy (XLMTM), a well-defined subtype of human centronuclear myopathy. Seven male Labrador Retrievers, age 14-26 wk, were clinically evaluated for generalized weakness and muscle atrophy. Muscle biopsies showed variability in fiber size, centrally placed nuclei resembling fetal myotubes, and subsarcolemmal ringed and central dense areas highlighted with mitochondrial specific reactions. Ultrastructural studies confirmed the centrally located nuclei, abnormal perinuclear structure, and mitochondrial accumulations. Wild-type triads were infrequent, with most exhibiting an abnormal orientation of T tubules. MTM1 gene sequencing revealed a unique exon 7 variant in all seven affected males, causing a nonconservative missense change, p.N155K, which haplotype data suggest derives from a recent founder in the local population. Analysis of a worldwide panel of 237 unaffected Labrador Retrievers and 59 additional control dogs from 25 other breeds failed to identify this variant, supporting it as the pathogenic mutation. Myotubularin protein levels and localization were abnormal in muscles from affected dogs, and expression of GFP-MTM1 p.N155K in COS-1 cells showed that the mutant protein was sequestered in proteasomes, where it was presumably misfolded and prematurely degraded. These data demonstrate that XLMTM in Labrador Retrievers is a faithful genetic model of the human condition.


Asunto(s)
Enfermedades de los Perros/genética , Mutación , Miopatías Estructurales Congénitas/veterinaria , Proteínas Tirosina Fosfatasas no Receptoras/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Enfermedades de los Perros/patología , Perros , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Genotipo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Haplotipos , Humanos , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Linaje , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Homología de Secuencia de Aminoácido
12.
Lancet Neurol ; 22(12): 1125-1139, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37977713

RESUMEN

BACKGROUND: X-linked myotubular myopathy is a rare, life-threatening, congenital muscle disease observed mostly in males, which is caused by mutations in MTM1. No therapies are approved for this disease. We aimed to assess the safety and efficacy of resamirigene bilparvovec, which is an adeno-associated viral vector serotype 8 delivering human MTM1. METHODS: ASPIRO is an open-label, dose-escalation trial at seven academic medical centres in Canada, France, Germany, and the USA. We included boys younger than 5 years with X-linked myotubular myopathy who required mechanical ventilator support. The trial was initially in two parts. Part 1 was planned as a safety and dose-escalation phase in which participants were randomly allocated (2:1) to either the first dose level (1·3 × 1014 vector genomes [vg]/kg bodyweight) of resamirigene bilparvovec or delayed treatment, then, for later participants, to either a higher dose (3·5 × 1014 vg/kg bodyweight) of resamirigene bilparvovec or delayed treatment. Part 2 was intended to confirm the dose selected in part 1. Resamirigene bilparvovec was administered as a single intravenous infusion. An untreated control group comprised boys who participated in a run-in study (INCEPTUS; NCT02704273) or those in the delayed treatment cohort who did not receive any dose. The primary efficacy outcome was the change from baseline to week 24 in hours of daily ventilator support. After three unexpected deaths, dosing at the higher dose was stopped and the two-part feature of the study design was eliminated. Because of changes to the study design during its implementation, analyses were done on an as-treated basis and are deemed exploratory. All treated and control participants were included in the safety analysis. The trial is registered with ClinicalTrials.gov, NCT03199469. Outcomes are reported as of Feb 28, 2022. ASPIRO is currently paused while deaths in dosed participants are investigated. FINDINGS: Between Aug 3, 2017 and June 1, 2021, 30 participants were screened for eligibility, of whom 26 were enrolled; six were allocated to the lower dose, 13 to the higher dose, and seven to delayed treatment. Of the seven children whose treatment was delayed, four later received the higher dose (n=17 total in the higher dose cohort), one received the lower dose (n=7 total in the lower dose cohort), and two received no dose and joined the control group (n=14 total, including 12 children from INCEPTUS). Median age at dosing or enrolment was 12·1 months (IQR 10·0-30·9; range 9·5-49·7) in the lower dose cohort, 31·1 months (16·0-64·7; 6·8-72·7) in the higher dose cohort, and 18·7 months (10·1-31·5; 5·9-39·3) in the control cohort. Median follow-up was 46·1 months (IQR 41·0-49·5; range 2·1-54·7) for lower dose participants, 27·6 months (24·6-29·1; 3·4-41·0) for higher dose participants, and 28·3 months (9·7-46·9; 5·7-32·7) for control participants. At week 24, lower dose participants had an estimated 77·7 percentage point (95% CI 40·22 to 115·24) greater reduction in least squares mean hours per day of ventilator support from baseline versus controls (p=0·0002), and higher dose participants had a 22·8 percentage point (6·15 to 39·37) greater reduction from baseline versus controls (p=0·0077). One participant in the lower dose cohort and three in the higher dose cohort died; at the time of death, all children had cholestatic liver failure following gene therapy (immediate causes of death were sepsis; hepatopathy, severe immune dysfunction, and pseudomonal sepsis; gastrointestinal haemorrhage; and septic shock). Three individuals in the control group died (haemorrhage presumed related to hepatic peliosis; aspiration pneumonia; and cardiopulmonary failure). INTERPRETATION: Most children with X-linked myotubular myopathy who received MTM1 gene replacement therapy had important improvements in ventilator dependence and motor function, with more than half of dosed participants achieving ventilator independence and some attaining the ability to walk independently. Investigations into the risk for underlying hepatobiliary disease in X-linked myotubular myopathy, and the need for monitoring of liver function before gene replacement therapy, are ongoing. FUNDING: Astellas Gene Therapies.


Asunto(s)
Miopatías Estructurales Congénitas , Sepsis , Masculino , Niño , Humanos , Lactante , Preescolar , Francia , Terapia Genética/efectos adversos , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/terapia , Alemania , Resultado del Tratamiento
13.
Mamm Genome ; 23(1-2): 85-108, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22218699

RESUMEN

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


Asunto(s)
Modelos Animales de Enfermedad , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/terapia , Animales , Biomarcadores , Perros , Distrofina/deficiencia , Distrofina/genética , Articulaciones/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología
15.
Muscle Nerve ; 46(4): 588-91, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22987702

RESUMEN

INTRODUCTION: We established a colony of dogs that harbor an X-linked MTM1 missense mutation.Muscle from affected male dogs exhibits reduction and altered localization of the MTM1 gene product, myotubularin, and provides a model analogous to X-linked myotubular myopathy (XLMTM). METHODS: We studied hindlimb muscle function in age-matched canine XLMTM genotypes between ages 9 and 18 weeks. RESULTS: By the end of the study, affected dogs produce only ∼15% of the torque generated by normals or carriers (0.023 ± 0.005 vs. 0.152 ± 0.007 and 0.154 ± 0.003 N-m/kg body mass, respectively, P < 0.05) and are too weak to stand unassisted. At this age, XLMTM dogs also demonstrate an abnormally low twitch:tetanus ratio, a right-shifted torque-frequency relationship and an increase in torque during repetitive stimulation (P < 0.05). CONCLUSIONS: We hypothesize that muscle weakness results from impaired excitation-contraction (E-C) coupling. Interventions that improve E-C coupling might be translated from the XLMTM dog model to patients.


Asunto(s)
Músculo Esquelético/fisiopatología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/fisiopatología , Cromosoma X/genética , Animales , Modelos Animales de Enfermedad , Perros , Masculino , Mutación Missense , Proteínas Tirosina Fosfatasas no Receptoras/genética
16.
Stem Cell Reports ; 16(9): 2169-2181, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34019816

RESUMEN

Duchenne muscular dystrophy (DMD) is a rare X-linked recessive disease that is associated with severe progressive muscle degeneration culminating in death due to cardiorespiratory failure. We previously observed an unexpected proliferation-independent telomere shortening in cardiomyocytes of a DMD mouse model. Here, we provide mechanistic insights using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using traction force microscopy, we show that DMD hiPSC-CMs exhibit deficits in force generation on fibrotic-like bioengineered hydrogels, aberrant calcium handling, and increased reactive oxygen species levels. Furthermore, we observed a progressive post-mitotic telomere shortening in DMD hiPSC-CMs coincident with downregulation of shelterin complex, telomere capping proteins, and activation of the p53 DNA damage response. This telomere shortening is blocked by blebbistatin, which inhibits contraction in DMD cardiomyocytes. Our studies underscore the role of fibrotic stiffening in the etiology of DMD cardiomyopathy. In addition, our data indicate that telomere shortening is progressive, contraction dependent, and mechanosensitive, and suggest points of therapeutic intervention.


Asunto(s)
Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Contracción Miocárdica/genética , Miocitos Cardíacos/metabolismo , Acortamiento del Telómero/genética , Biomarcadores , Cardiomiopatías/etiología , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Diferenciación Celular , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Fibrosis , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Inmunofenotipificación , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Fenómenos Mecánicos , Distrofias Musculares/patología , Distrofia Muscular de Duchenne/etiología , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Contracción Miocárdica/efectos de los fármacos
17.
Muscle Nerve ; 42(1): 130-2, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20544944

RESUMEN

We tested the hypothesis that eccentric contractions (ECCs) rapidly induce greater-than-normal isometric torque drop in dystrophin-deficient golden retriever muscular dystrophy (GRMD) muscles. ECCs were imposed by forcibly stretching activated muscles. The results indicate that isometric torque drop was greater in GRMD versus controls (P < 0.0001). Our findings support the hypothesis that ECCs induce greater-than-normal isometric torque drop in GRMD muscles. The magnitude of ECC-induced isometric torque loss may be an ideal clinical endpoint in the GRMD model.


Asunto(s)
Distrofina/deficiencia , Distrofina/genética , Contracción Isométrica/genética , Contracción Isométrica/fisiología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatología , Animales , Perros , Estimulación Eléctrica , Heterocigoto , Músculo Esquelético/fisiología , Resistencia Física
18.
Cardiovasc Res ; 116(2): 368-382, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31049579

RESUMEN

AIMS: Heart failure invariably affects patients with various forms of muscular dystrophy (MD), but the onset and molecular sequelae of altered structure and function resulting from full-length dystrophin (Dp427) deficiency in MD heart tissue are poorly understood. To better understand the role of dystrophin in cardiomyocyte development and the earliest phase of Duchenne muscular dystrophy (DMD) cardiomyopathy, we studied human cardiomyocytes differentiated from induced pluripotent stem cells (hiPSC-CMs) obtained from the urine of a DMD patient. METHODS AND RESULTS: The contractile properties of patient-specific hiPSC-CMs, with no detectable dystrophin (DMD-CMs with a deletion of exon 50), were compared to CMs containing a CRISPR-Cas9 mediated deletion of a single G base at position 263 of the dystrophin gene (c.263delG-CMs) isogenic to the parental line of hiPSC-CMs from a healthy individual. We hypothesized that the absence of a dystrophin-actin linkage would adversely affect myofibril and cardiomyocyte structure and function. Cardiomyocyte maturation was driven by culturing long-term (80-100 days) on a nanopatterned surface, which resulted in hiPSC-CMs with adult-like dimensions and aligned myofibrils. CONCLUSIONS: Our data demonstrate that lack of Dp427 results in reduced myofibril contractile tension, slower relaxation kinetics, and to Ca2+ handling abnormalities, similar to DMD cells, suggesting either retarded or altered maturation of cardiomyocyte structures associated with these functions. This study offers new insights into the functional consequences of Dp427 deficiency at an early stage of cardiomyocyte development in both patient-derived and CRISPR-generated models of dystrophin deficiency.


Asunto(s)
Cardiomiopatías/etiología , Diferenciación Celular , Distrofina/deficiencia , Células Madre Pluripotentes Inducidas/metabolismo , Distrofia Muscular de Duchenne/complicaciones , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Miofibrillas/metabolismo , Señalización del Calcio , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Estudios de Casos y Controles , Línea Celular , Distrofina/genética , Humanos , Células Madre Pluripotentes Inducidas/ultraestructura , Cinética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Miocitos Cardíacos/ultraestructura , Miofibrillas/ultraestructura
19.
Neuromuscul Disord ; 18(5): 413-22, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18440815

RESUMEN

Null mutation of titin-cap (TCAP) causes limb-girdle muscular dystrophy type 2G (LGMD2G). LGMD2G patients develop muscle atrophy, and lose the ability to walk by their third decade. Previous findings suggest that TCAP regulates myostatin, a key regulator of muscle growth. We tested the hypothesis that TCAP knockdown with RNA interference will lead to differential expression of genes involved in muscle proliferation and differentiation, impairing muscle cell growth. mRNA from cultured cells treated with TCAP siRNA duplex constructs was analyzed using Northern blots and real-time RT-PCR. siRNA treatment decreased TCAP mRNA expression in differentiating muscle cells. Significant (p<0.05) decreases in mRNA were observed for myogenic regulatory factors. siRNA treatment also prevented development of the normal phenotype of muscle cells. Our findings suggest that TCAP knockdown with RNA interference alters normal muscle cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Musculares/fisiología , Mioblastos Esqueléticos/metabolismo , Interferencia de ARN , Animales , Secuencia de Bases , Northern Blotting , Diferenciación Celular/genética , Línea Celular , Proliferación Celular , Conectina , Técnica del Anticuerpo Fluorescente , Expresión Génica , Factor II del Crecimiento Similar a la Insulina/genética , Ratones , Datos de Secuencia Molecular , Proteínas Musculares/genética , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Proteína MioD/genética , Mioblastos Esqueléticos/citología , Miogenina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Stem Cell Reports ; 10(3): 942-955, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29478899

RESUMEN

Genetic deficiency of dystrophin leads to disability and premature death in Duchenne muscular dystrophy (DMD), affecting the heart as well as skeletal muscle. Here, we report that clinical-stage cardiac progenitor cells, known as cardiosphere-derived cells (CDCs), improve cardiac and skeletal myopathy in the mdx mouse model of DMD. Injection of CDCs into the hearts of mdx mice augments cardiac function, ambulatory capacity, and survival. Exosomes secreted by human CDCs reproduce the benefits of CDCs in mdx mice and in human induced pluripotent stem cell-derived Duchenne cardiomyocytes. Surprisingly, CDCs and their exosomes also transiently restored partial expression of full-length dystrophin in mdx mice. The findings further motivate the testing of CDCs in Duchenne patients, while identifying exosomes as next-generation therapeutic candidates.


Asunto(s)
Exosomas/fisiología , Distrofia Muscular de Duchenne/terapia , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Distrofina/metabolismo , Exosomas/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA