Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(19): 4919-4938.e22, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34506722

RESUMEN

Replacing or editing disease-causing mutations holds great promise for treating many human diseases. Yet, delivering therapeutic genetic modifiers to specific cells in vivo has been challenging, particularly in large, anatomically distributed tissues such as skeletal muscle. Here, we establish an in vivo strategy to evolve and stringently select capsid variants of adeno-associated viruses (AAVs) that enable potent delivery to desired tissues. Using this method, we identify a class of RGD motif-containing capsids that transduces muscle with superior efficiency and selectivity after intravenous injection in mice and non-human primates. We demonstrate substantially enhanced potency and therapeutic efficacy of these engineered vectors compared to naturally occurring AAV capsids in two mouse models of genetic muscle disease. The top capsid variants from our selection approach show conserved potency for delivery across a variety of inbred mouse strains, and in cynomolgus macaques and human primary myotubes, with transduction dependent on target cell expressed integrin heterodimers.


Asunto(s)
Cápside/metabolismo , Dependovirus/metabolismo , Evolución Molecular Dirigida , Técnicas de Transferencia de Gen , Músculo Esquelético/metabolismo , Secuencia de Aminoácidos , Animales , Cápside/química , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Integrinas/metabolismo , Macaca fascicularis , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/terapia , Multimerización de Proteína , Proteínas Tirosina Fosfatasas no Receptoras/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/uso terapéutico , ARN Guía de Kinetoplastida/metabolismo , Recombinación Genética/genética , Especificidad de la Especie , Transgenes
2.
FASEB J ; 37(10): e23198, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37742307

RESUMEN

DOCK (dedicator of cytokinesis) is an 11-member family of typical guanine nucleotide exchange factors (GEFs) expressed in the brain, spinal cord, and skeletal muscle. Several DOCK proteins have been implicated in maintaining several myogenic processes such as fusion. We previously identified DOCK3 as being strongly upregulated in Duchenne muscular dystrophy (DMD), specifically in the skeletal muscles of DMD patients and dystrophic mice. Dock3 ubiquitous KO mice on the dystrophin-deficient background exacerbated skeletal muscle and cardiac phenotypes. We generated Dock3 conditional skeletal muscle knockout mice (Dock3 mKO) to characterize the role of DOCK3 protein exclusively in the adult muscle lineage. Dock3 mKO mice presented with significant hyperglycemia and increased fat mass, indicating a metabolic role in the maintenance of skeletal muscle health. Dock3 mKO mice had impaired muscle architecture, reduced locomotor activity, impaired myofiber regeneration, and metabolic dysfunction. We identified a novel DOCK3 interaction with SORBS1 through the C-terminal domain of DOCK3 that may account for its metabolic dysregulation. Together, these findings demonstrate an essential role for DOCK3 in skeletal muscle independent of DOCK3 function in neuronal lineages.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Distrofia Muscular de Duchenne , Humanos , Adulto , Animales , Ratones , Músculo Esquelético , Encéfalo , Ratones Noqueados , Glucosa , Proteínas del Tejido Nervioso , Factores de Intercambio de Guanina Nucleótido/genética
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.
Hum Mol Genet ; 28(2): 320-331, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30307508

RESUMEN

Facioscapulohumeral dystrophy type 1 (FSHD-1) is the most common autosomal dominant form of muscular dystrophy with a prevalence of ∼1 in 8000 individuals. It is considered a late-onset form of muscular dystrophy and leads to asymmetric muscle weakness in the facial, scapular, trunk and lower extremities. The prevalent hypothesis on disease pathogenesis is explained by misexpression of a germ line, primate-specific transcription factor DUX4-fl (double homeobox 4, full-length isoform) linked to the chromosome 4q35. In vitro and in vivo studies have demonstrated that very low levels of DUX4-fl expression are sufficient to induce an apoptotic and/or lethal phenotype, and therefore modeling of the disease has proved challenging. In this study, we expand upon our previously established injection model of DUX4 misexpression in zebrafish and describe a DUX4-inducible transgenic zebrafish model that better recapitulates the expression pattern and late onset phenotype characteristic of FSHD patients. We show that an induced burst of DUX4 expression during early development results in the onset of FSHD-like phenotypes in adulthood, even when DUX4 is no longer detectable. We also utilize our injection model to study long-term consequences of DUX4 expression in those that fail to show a developmental phenotype. Herein, we introduce a hypothesis that DUX4 expression during developmental stages is sufficient to induce FSHD-like phenotypes in later adulthood. Our findings point to a developmental role of DUX4 misexpression in the pathogenesis of FSHD and should be factored into the design of future therapies.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapulohumeral/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Contracción Muscular , Músculo Esquelético/embriología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal , Distrofia Muscular Facioescapulohumeral/embriología , Distrofia Muscular Facioescapulohumeral/etiología , Distrofia Muscular Facioescapulohumeral/genética , Pez Cebra , Proteínas de Pez Cebra/genética
5.
Mol Ther ; 28(1): 189-201, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31628052

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked muscle wasting disease that is caused by the loss of functional dystrophin protein in cardiac and skeletal muscles. DMD patient muscles become weakened, leading to eventual myofiber breakdown and replacement with fibrotic and adipose tissues. Inflammation drives the pathogenic processes through releasing inflammatory cytokines and other factors that promote skeletal muscle degeneration and contributing to the loss of motor function. Selective inhibitors of nuclear export (SINEs) are a class of compounds that function by inhibiting the nuclear export protein exportin 1 (XPO1). The XPO1 protein is an important regulator of key inflammatory and neurological factors that drive inflammation and neurotoxicity in various neurological and neuromuscular diseases. Here, we demonstrate that SINE compound KPT-350 can ameliorate dystrophic-associated pathologies in the muscles of DMD models of zebrafish and mice. Thus, SINE compounds are a promising novel strategy for blocking dystrophic symptoms and could be used in combinatorial treatments for DMD.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Carioferinas/antagonistas & inhibidores , Distrofia Muscular de Duchenne/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Pez Cebra/genética , Administración Oral , Animales , Biomarcadores/sangre , Citocinas/antagonistas & inhibidores , Citocinas/sangre , Modelos Animales de Enfermedad , Locomoción/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos DBA , Ratones Endogámicos mdx , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación , Proteínas de Pez Cebra/genética , Proteína Exportina 1
6.
PLoS Genet ; 14(3): e1007226, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29518074

RESUMEN

Gene expression in a tissue-specific context depends on the combined efforts of epigenetic, transcriptional and post-transcriptional processes that lead to the production of specific proteins that are important determinants of cellular identity. Ribosomes are a central component of the protein biosynthesis machinery in cells; however, their regulatory roles in the translational control of gene expression in skeletal muscle remain to be defined. In a genetic screen to identify critical regulators of myogenesis, we identified a DEAD-Box RNA helicase, DDX27, that is required for skeletal muscle growth and regeneration. We demonstrate that DDX27 regulates ribosomal RNA (rRNA) maturation, and thereby the ribosome biogenesis and the translation of specific transcripts during myogenesis. These findings provide insight into the translational regulation of gene expression in myogenesis and suggest novel functions for ribosomes in regulating gene expression in skeletal muscles.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Músculo Esquelético/fisiología , Biosíntesis de Proteínas , ARN Ribosómico/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestructura , Proliferación Celular/genética , ARN Helicasas DEAD-box/genética , Embrión no Mamífero , Ratones , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Mioblastos/citología , Mioblastos/fisiología , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo , ARN Ribosómico/genética , Regeneración/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
7.
Hum Mol Genet ; 27(9): 1608-1617, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29474540

RESUMEN

Centronuclear myopathies (CNM) are a subtype of congenital myopathies (CM) characterized by skeletal muscle weakness and an increase in the number of central myonuclei. We have previously identified three CNM probands, two with associated dilated cardiomyopathy, carrying striated preferentially expressed gene (SPEG) mutations. Currently, the role of SPEG in skeletal muscle function is unclear as constitutive SPEG-deficient mice developed severe dilated cardiomyopathy and died in utero. We have generated a conditional Speg-KO mouse model and excised Speg by crosses with striated muscle-specific cre-expressing mice (MCK-Cre). The resulting litters had a delay in Speg excision consistent with cre expression starting in early postnatal life and, therefore, an extended lifespan up to a few months. KO mice were significantly smaller and weaker than their littermate-matched controls. Histopathological skeletal muscle analysis revealed smaller myofibers, marked fiber-size variability, and poor integrity and low number of triads. Further, SPEG-deficient muscle fibers were weaker by physiological and in vitro studies and exhibited abnormal Ca2+ handling and excitation-contraction (E-C) coupling. Overall, SPEG deficiency in skeletal muscle is associated with fewer and abnormal triads, and defective calcium handling and excitation-contraction coupling, suggesting that therapies targeting calcium signaling may be beneficial in such patients.


Asunto(s)
Calcio/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Señalización del Calcio/fisiología , Femenino , Ratones , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética
8.
Eur J Appl Physiol ; 119(10): 2339-2348, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31468173

RESUMEN

PURPOSE: Changes in stiffness or extensibility of the muscle or muscle-tendon unit with aging could lead to impaired function and an increased vulnerability to injury. We aimed to investigate the passive force and viscoelastic properties of single muscle fibers in older adults. METHODS: Seven older adults (mean age 79.0 ± 3.8 years) and 10 young control (mean age 25.6 ± 4.5 years) were recruited. Biopsy specimens were obtained percutaneously from m. vastus lateralis and skinned single fibers were used for the experiments. Slack tests were performed to determine maximal force and maximal unloaded shortening velocity. Passive force was measured in pCa 9.0 solution using a stepwise stretch technique with increment of sarcomere length from 2.4 to 4.2 µm. Myosin heavy chain (MHC) isoform was determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Specific force was calculated as maximal force divided by cross-sectional area. Passive force, peak passive force, time to half stress relaxation (T1/2) and force decay index (a force time integral under a stress relaxation curve) were measured. RESULTS: No difference between the groups were found in specific force and shortening velocity. Passive force and peak passive force were greater in both MHC I and IIa fibers of older adults (p < 0.001, p = 0.012, respectively, at 4.2 mm SL). Force decay index was higher in older adults. (p = 0.001 at 4.2 µm SL). There were no significant differences in passive force and viscoelastic properties between fiber types. CONCLUSION: We demonstrated greater passive force and viscoelastic properties at the level of single fibers in older adults.


Asunto(s)
Envejecimiento/fisiología , Fibras Musculares Esqueléticas/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Elasticidad , Femenino , Humanos , Masculino , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Relajación Muscular , Cadenas Pesadas de Miosina/metabolismo , Viscosidad
9.
Physiol Genomics ; 48(11): 850-860, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27764767

RESUMEN

Sapje zebrafish lack the protein dystrophin and are the smallest vertebrate model of Duchenne muscular dystrophy (DMD). Their small size makes them ideal for large-scale drug discovery screens. However, the extent that sapje mimic the muscle dysfunction of higher vertebrate models of DMD is unclear. We used an optical birefringence assay to differentiate affected dystrophic sapje larvae from their unaffected siblings and then studied trunk muscle contractility at 4-7 days postfertilization. Preparation cross-sectional area (CSA) was similar for affected and unaffected larvae, yet tetanic forces of affected preparations were only 30-60% of normal. ANCOVA indicated that the linear relationship observed between tetanic force and CSA for unaffected preparations was absent in the affected population. Consequently, the average force/CSA of affected larvae was depressed 30-70%. Disproportionate reductions in twitch vs. tetanic force, and a slowing of twitch tension development and relaxation, indicated that the myofibrillar disorganization evident in the birefringence assay could not explain the entire force loss. Single eccentric contractions, in which activated preparations were lengthened 5-10%, resulted in tetanic force deficits in both groups of larvae. However, deficits of affected preparations were three- to fivefold greater at all strains and ages, even after accounting for any recovery. Based on these functional assessments, we conclude that the sapje mutant zebrafish is a phenotypically severe model of DMD. The severe contractile deficits of sapje larvae represent novel physiological endpoints for therapeutic drug screening.


Asunto(s)
Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Pez Cebra/fisiología , Animales , Modelos Animales de Enfermedad , Cinética , Contracción Muscular , Fibras Musculares de Contracción Rápida/patología , Fibras Musculares de Contracción Lenta/patología , Análisis de Regresión , Sarcómeros/metabolismo , Tetania/fisiopatología
10.
Hum Mol Genet ; 23(7): 1869-78, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24234649

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by a lack of the dystrophin protein and has no effective treatment at present. Zebrafish provide a powerful in vivo tool for high-throughput therapeutic drug screening for the improvement of muscle phenotypes caused by dystrophin deficiency. Using the dystrophin-deficient zebrafish, sapje, we have screened a total of 2640 compounds with known modes of action from three drug libraries to identify modulators of the disease progression. Six compounds that target heme oxygenase signaling were found to rescue the abnormal muscle phenotype in sapje and sapje-like, while upregulating the inducible heme oxygenase 1 (Hmox1) at the protein level. Direct Hmox1 overexpression by injection of zebrafish Hmox1 mRNA into fertilized eggs was found to be sufficient for a dystrophin-independent restoration of normal muscle via an upregulation of cGMP levels. In addition, treatment of mdx(5cv) mice with the PDE5 inhibitor, sildenafil, which was one of the six drugs impacting the Hmox1 pathway in zebrafish, significantly increased the expression of Hmox1 protein, thus making Hmox1 a novel target for the improvement of dystrophic symptoms. These results demonstrate the translational relevance of our zebrafish model to mammalian models and support the use of zebrafish to screen for new drugs to treat human DMD. The discovery of a small molecule and a specific therapeutic pathway that might mitigate DMD disease progression could lead to significant clinical implications.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Distrofina/genética , Hemo-Oxigenasa 1/biosíntesis , Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , GMP Cíclico/biosíntesis , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Distrofina/deficiencia , Hemo-Oxigenasa 1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , ARN Mensajero/genética , Transducción de Señal/genética , Citrato de Sildenafil , Sulfonas/farmacología , Regulación hacia Arriba , Pez Cebra/genética
11.
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
12.
Expert Opin Drug Discov ; 18(6): 629-641, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37183669

RESUMEN

INTRODUCTION: Zebrafish larvae are one of the few vertebrates amenable to large-scale drug discovery screens. Larval swimming behavior is often used as an outcome variable and many fields of study have developed assays for evaluating swimming performance. An unintended consequence of this wide interest is that details related to assay methodology and interpretation become scattered across the literature. The aim of this review is to consolidate this information, particularly as it relates to high-throughput approaches. AREAS COVERED: The authors describe larval swimming behaviors as this forms the basis for understanding their experimentally evoked swimming or spontaneous activity. Next, they detail how swimming activity can serve as an outcome variable, particularly in the multi-well formats used in large-scale screening studies. They also highlight biological and technical factors that can impact the sensitivity and variability of these measurements. EXPERT OPINION: Careful attention to animal husbandry, experimental design, data acquisition, and interpretation of results can improve screen outcomes by maximizing swimming activity while minimizing intra- and inter-larval variability. The development of more sensitive, quantitative methods of assessing swimming performance that can be incorporated into high-throughput workflows will be important in order to take full advantage of the zebrafish model.


Asunto(s)
Natación , Pez Cebra , Animales , Natación/fisiología , Pez Cebra/fisiología , Larva/fisiología , Descubrimiento de Drogas
13.
bioRxiv ; 2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36865261

RESUMEN

DOCK (dedicator of cytokinesis) is an 11-member family of typical guanine nucleotide exchange factors (GEFs) expressed in the brain, spinal cord, and skeletal muscle. Several DOCK proteins have been implicated in maintaining several myogenic processes such as fusion. We previously identified DOCK3 as being strongly upregulated in Duchenne muscular dystrophy (DMD), specifically in the skeletal muscles of DMD patients and dystrophic mice. Dock3 ubiquitous KO mice on the dystrophin-deficient background exacerbated skeletal muscle and cardiac phenotypes. We generated Dock3 conditional skeletal muscle knockout mice (Dock3 mKO) to characterize the role of DOCK3 protein exclusively in the adult muscle lineage. Dock3 mKO mice presented with significant hyperglycemia and increased fat mass, indicating a metabolic role in the maintenance of skeletal muscle health. Dock3 mKO mice had impaired muscle architecture, reduced locomotor activity, impaired myofiber regeneration, and metabolic dysfunction. We identified a novel DOCK3 interaction with SORBS1 through the C-terminal domain of DOCK3 that may account for its metabolic dysregulation. Together, these findings demonstrate an essential role for DOCK3 in skeletal muscle independent of DOCK3 function in neuronal lineages.

14.
Sci Rep ; 13(1): 7191, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37137956

RESUMEN

Age-related deficits in skeletal muscle function, termed sarcopenia, are due to loss of muscle mass and changes in the intrinsic mechanisms underlying contraction. Sarcopenia is associated with falls, functional decline, and mortality. Electrical impedance myography (EIM)-a minimally invasive, rapid electrophysiological tool-can be applied to animals and humans to monitor muscle health, thereby serving as a biomarker in both preclinical and clinical studies. EIM has been successfully employed in several species; however, the application of EIM to the assessment of zebrafish-a model organism amenable to high-throughput experimentation-has not been reported. Here, we demonstrated differences in EIM measures between the skeletal muscles of young (6 months of age) and aged (33 months of age) zebrafish. For example, EIM phase angle and reactance at 2 kHz showed significantly decreased phase angle (5.3 ± 2.1 versus 10.7 ± 1.5°; p = 0.001) and reactance (89.0 ± 3.9 versus 172.2 ± 54.8 ohms; p = 0.007) in aged versus young animals. Total muscle area, in addition to other morphometric features, was also strongly correlated to EIM 2 kHz phase angle across both groups (r = 0.7133, p = 0.01). Moreover, there was a strong correlation between 2 kHz phase angle and established metrics of zebrafish swimming performance, including turn angle, angular velocity, and lateral motion (r = 0.7253, r = 0.7308, r = 0.7857, respectively, p < 0.01 for all). In addition, the technique was shown to have high reproducibility between repeated measurements with a mean percentage difference of 5.34 ± 1.17% for phase angle. These relationships were also confirmed in a separate replication cohort. Together, these findings establish EIM as a fast, sensitive method for quantifying zebrafish muscle function and quality. Moreover, identifying the abnormalities in the bioelectrical properties of sarcopenic zebrafish provides new opportunities to evaluate potential therapeutics for age-related neuromuscular disorders and to interrogate the disease mechanisms of muscle degeneration.


Asunto(s)
Sarcopenia , Pez Cebra , Humanos , Animales , Impedancia Eléctrica , Reproducibilidad de los Resultados , Miografía/métodos , Músculo Esquelético/fisiología , Atrofia
15.
Elife ; 122023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37432316

RESUMEN

Ubiquitin-proteasome system (UPS) dysfunction is associated with the pathology of a wide range of human diseases, including myopathies and muscular atrophy. However, the mechanistic understanding of specific components of the regulation of protein turnover during development and disease progression in skeletal muscle is unclear. Mutations in KLHL40, an E3 ubiquitin ligase cullin3 (CUL3) substrate-specific adapter protein, result in severe congenital nemaline myopathy, but the events that initiate the pathology and the mechanism through which it becomes pervasive remain poorly understood. To characterize the KLHL40-regulated ubiquitin-modified proteome during skeletal muscle development and disease onset, we used global, quantitative mass spectrometry-based ubiquitylome and global proteome analyses of klhl40a mutant zebrafish during disease progression. Global proteomics during skeletal muscle development revealed extensive remodeling of functional modules linked with sarcomere formation, energy, biosynthetic metabolic processes, and vesicle trafficking. Combined analysis of klh40 mutant muscle proteome and ubiquitylome identified thin filament proteins, metabolic enzymes, and ER-Golgi vesicle trafficking pathway proteins regulated by ubiquitylation during muscle development. Our studies identified a role for KLHL40 as a regulator of ER-Golgi anterograde trafficking through ubiquitin-mediated protein degradation of secretion-associated Ras-related GTPase1a (Sar1a). In KLHL40-deficient muscle, defects in ER exit site vesicle formation and downstream transport of extracellular cargo proteins result in structural and functional abnormalities. Our work reveals that the muscle proteome is dynamically fine-tuned by ubiquitylation to regulate skeletal muscle development and uncovers new disease mechanisms for therapeutic development in patients.


Asunto(s)
Proteínas Musculares , Pez Cebra , Animales , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Pez Cebra/metabolismo , Proteoma/metabolismo , Músculo Esquelético/metabolismo , Ubiquitinación , Sarcómeros/metabolismo , Ubiquitina/metabolismo , Retículo Endoplásmico/metabolismo , Desarrollo de Músculos , Progresión de la Enfermedad
17.
J Sport Rehabil ; 21(2): 99-106, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22387809

RESUMEN

CONTEXT: Static stretching is commonly used during the treatment and rehabilitation of orthopedic injuries to increase joint range of motion (ROM) and muscle flexibility. Understanding the physiological adaptations that occur in the neuromuscular system as a result of long-term stretching may provide insight into the mechanisms responsible for changes in flexibility. OBJECTIVE: To examine possible neurological origins and adaptations in the Ia-reflex pathway that allow for increases in flexibility in ankle ROM, by evaluating the reduction in the synaptic transmission of Ia afferents to the motoneuron pool. DESIGN: Repeated-measures, case-controlled study. SETTING: Sports medicine research laboratory. PARTICIPANTS: 40 healthy volunteers with no history of cognitive impairment, neurological impairment, or lower extremity surgery or injury within the previous 12 mo. INTERVENTION: Presynaptic and postsynaptic mechanisms were evaluated with a chronic stretching pro- tocol. Twenty subjects stretched 5 times a wk for 6 wk. All subjects were measured at baseline, 3 wk, and 6 wk. MAIN OUTCOME MEASURES: Ankle-dorsiflexion ROM, Hmax:Mmax, presynaptic inhibition, and disynaptic reciprocal inhibition. RESULTS: Only ROM had a significant interaction between group and time, whereas the other dependent variables did not show significant differences. The experimental group had significantly improved ROM from baseline to 3 wk (mean 6.2 ± 0.9, P < .001), 3 wk to 6 wk (mean 5.0 ± 0.8, P < .001), and baseline to 6 wk (mean 11.2 ±0.9, P < .001). CONCLUSIONS: Ankle dorsiflexion increased by 42.25% after 6 wk of static stretching, but no significant neurological changes resulted at any point of the study, contrasting current literature. Significant neuromuscular origins of adaptation do not exist in the Ia-reflex-pathway components after a long-term stretching program as currently understood. Thus, any increases in flexibility are the result of other factors, potentially mechanical changes or stretch tolerance.


Asunto(s)
Adaptación Fisiológica/fisiología , Articulación del Tobillo/fisiología , Ejercicios de Estiramiento Muscular , Músculo Esquelético/fisiología , Sistema Nervioso Periférico/fisiología , Rango del Movimiento Articular/fisiología , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Factores de Tiempo , Adulto Joven
18.
JCI Insight ; 7(15)2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35763354

RESUMEN

Striated preferentially expressed protein kinase (SPEG), a myosin light chain kinase, is mutated in centronuclear myopathy (CNM) and/or dilated cardiomyopathy. No precise therapies are available for this disorder, and gene replacement therapy is not a feasible option due to the large size of SPEG. We evaluated the potential of dynamin-2 (DNM2) reduction as a potential therapeutic strategy because it has been shown to revert muscle phenotypes in mouse models of CNM caused by MTM1, DNM2, and BIN1 mutations. We determined that SPEG-ß interacted with DNM2, and SPEG deficiency caused an increase in DNM2 levels. The DNM2 reduction strategy in Speg-KO mice was associated with an increase in life span, body weight, and motor performance. Additionally, it normalized the distribution of triadic proteins, triad ultrastructure, and triad number and restored phosphatidylinositol-3-phosphate levels in SPEG-deficient skeletal muscles. Although DNM2 reduction rescued the myopathy phenotype, it did not improve cardiac dysfunction, indicating a differential tissue-specific function. Combining DNM2 reduction with other strategies may be needed to target both the cardiac and skeletal defects associated with SPEG deficiency. DNM2 reduction should be explored as a therapeutic strategy against other genetic myopathies (and dystrophies) associated with a high level of DNM2.


Asunto(s)
Dinamina II , Miopatías Estructurales Congénitas , Animales , Modelos Animales de Enfermedad , Dinamina II/genética , Ratones , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/terapia , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Fenotipo
19.
Life Sci Alliance ; 5(9)2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35512829

RESUMEN

miR-486 is a muscle-enriched microRNA, or "myomiR," that has reduced expression correlated with Duchenne muscular dystrophy (DMD). To determine the function of miR-486 in normal and dystrophin-deficient muscles and elucidate miR-486 target transcripts in skeletal muscle, we characterized mir-486 knockout mice (mir-486 KO). mir-486 KO mice developed disrupted myofiber architecture, decreased myofiber size, decreased locomotor activity, increased cardiac fibrosis, and metabolic defects were exacerbated in mir-486 KO:mdx 5cv (DKO) mice. To identify direct in vivo miR-486 muscle target transcripts, we integrated RNA sequencing and chimeric miRNA eCLIP sequencing to identify key transcripts and pathways that contribute towards mir-486 KO and dystrophic disease pathologies. These targets included known and novel muscle metabolic and dystrophic structural remodeling factors of muscle and skeletal muscle contractile transcript targets. Together, our studies identify miR-486 as essential for normal muscle function, a driver of pathological remodeling in dystrophin-deficient muscle, a useful biomarker for dystrophic disease progression, and highlight the use of multiple omic platforms to identify in vivo microRNA target transcripts.


Asunto(s)
Distrofina , MicroARNs , Animales , Distrofina/genética , Ratones , Ratones Endogámicos mdx , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Transcriptoma/genética
20.
Muscle Nerve ; 44(4): 563-70, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21922468

RESUMEN

INTRODUCTION: Skeletal muscles of mdx mice lack functional levels of dystrophin due to a mutation in Dmd exon 23. Morpholino antisense oligomers can induce expression of a truncated dystrophin by redirecting splicing to skip processing of exon 23. METHODS: We tested whether systemic administration of Vivo-Morpholino, an octaguanidine delivery moiety-Morpholino conjugate that targets exon 23 (VMO23), restored function to muscles of mdx mice. RESULTS: Extensor digitorum longus (EDL) muscles of mdx mice were weaker, less powerful, and showed greater functional deficits after eccentric contractions than normal. VMO23 treatment normalized EDL force and power of mdx mice and eliminated their exaggerated sensitivity to eccentric contractions. Diaphragm muscle strips from mdx mice also produced lower-than-normal force and power, and these variables were restored to normal, or near-normal, levels by VMO23 treatment. CONCLUSION: These results provide a functional basis for continuing development of VMO23 as a treatment for Duchenne muscular dystrophy.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Guanidina/farmacología , Morfolinas/farmacología , Músculo Esquelético/efectos de los fármacos , Distrofias Musculares/patología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Distrofina/metabolismo , Estimulación Eléctrica/métodos , Guanidina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Morfolinas/uso terapéutico , Contracción Muscular/efectos de los fármacos , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/fisiopatología , Distrofias Musculares/tratamiento farmacológico , Distrofias Musculares/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA