Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Genes Dev ; 34(15-16): 1089-1105, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32616519

RESUMEN

The circadian clock is encoded by a negative transcriptional feedback loop that coordinates physiology and behavior through molecular programs that remain incompletely understood. Here, we reveal rhythmic genome-wide alternative splicing (AS) of pre-mRNAs encoding regulators of peptidergic secretion within pancreatic ß cells that are perturbed in Clock-/- and Bmal1-/- ß-cell lines. We show that the RNA-binding protein THRAP3 (thyroid hormone receptor-associated protein 3) regulates circadian clock-dependent AS by binding to exons at coding sequences flanking exons that are more frequently skipped in clock mutant ß cells, including transcripts encoding Cask (calcium/calmodulin-dependent serine protein kinase) and Madd (MAP kinase-activating death domain). Depletion of THRAP3 restores expression of the long isoforms of Cask and Madd, and mimicking exon skipping in these transcripts through antisense oligonucleotide delivery in wild-type islets reduces glucose-stimulated insulin secretion. Finally, we identify shared networks of alternatively spliced exocytic genes from islets of rodent models of diet-induced obesity that significantly overlap with clock mutants. Our results establish a role for pre-mRNA alternative splicing in ß-cell function across the sleep/wake cycle.


Asunto(s)
Empalme Alternativo , Relojes Circadianos/genética , Exocitosis , Glucosa/metabolismo , Secreción de Insulina/genética , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/fisiología , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/fisiología , Células Cultivadas , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Homeostasis , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas Nucleares/fisiología , Obesidad/genética , Obesidad/metabolismo , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Factores de Transcripción/fisiología
4.
Dis Model Mech ; 13(2)2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31582396

RESUMEN

Limb-girdle muscular dystrophy type 2C is caused by autosomal recessive mutations in the γ-sarcoglycan (SGCG) gene. The most common SGCG mutation is a single nucleotide deletion from a stretch of five thymine residues in SGCG exon 6 (521ΔT). This founder mutation disrupts the transcript reading frame, abolishing protein expression. An antisense oligonucleotide exon-skipping method to reframe the human 521ΔT transcript requires skipping four exons to generate a functional, internally truncated protein. In vivo evaluation of this multi-exon skipping, antisense-mediated therapy requires a genetically appropriate mouse model. The human and mouse γ-sarcoglycan genes are highly homologous in sequence and gene structure, including the exon 6 region harboring the founder mutation. Herein, we describe a new mouse model of this form of limb-girdle muscular dystrophy generated using CRISPR/Cas9-mediated gene editing to introduce a single thymine deletion in murine exon 6, recreating the 521ΔT point mutation in Sgcg These mice express the 521ΔT transcript, lack γ-sarcoglycan protein and exhibit a severe dystrophic phenotype. Phenotypic characterization demonstrated reduced muscle mass, increased sarcolemmal leak and fragility, and decreased muscle function, consistent with the human pathological findings. Furthermore, we showed that intramuscular administration of a murine-specific multiple exon-directed antisense oligonucleotide cocktail effectively corrected the 521ΔT reading frame. These data demonstrate a molecularly and pathologically suitable model for in vivo testing of a multi-exon skipping strategy to advance preclinical development of this genetic correction approach.


Asunto(s)
Exones/genética , Edición Génica , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/terapia , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Fibrosis , Ratones Transgénicos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Mutación Puntual/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Sarcolema/metabolismo
5.
JCI Insight ; 4(6)2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30730308

RESUMEN

Myotonic dystrophy (DM) is the most common autosomal dominant muscular dystrophy and encompasses both skeletal muscle and cardiac complications. DM is nucleotide repeat expansion disorder in which type 1 (DM1) is due to a trinucleotide repeat expansion on chromosome 19 and type 2 (DM2) arises from a tetranucleotide repeat expansion on chromosome 3. Developing representative models of DM in animals has been challenging due to instability of nucleotide repeat expansions, especially for DM2, which is characterized by nucleotide repeat expansions often greater than 5,000 copies. To investigate mechanisms of human DM, we generated cellular models of DM1 and DM2. We used regulated MyoD expression to reprogram urine-derived cells into myotubes. In this myogenic cell model, we found impaired dystrophin expression, in the presence of muscleblind-like 1 (MBNL1) foci, and aberrant splicing in DM1 but not in DM2 cells. We generated induced pluripotent stem cells (iPSC) from healthy controls and DM1 and DM2 subjects, and we differentiated these into cardiomyocytes. DM1 and DM2 cells displayed an increase in RNA foci concomitant with cellular differentiation. iPSC-derived cardiomyocytes from DM1 but not DM2 had aberrant splicing of known target genes and MBNL sequestration. High-resolution imaging revealed tight association between MBNL clusters and RNA foci in DM1. Ca2+ transients differed between DM1- and DM2 iPSC-derived cardiomyocytes, and each differed from healthy control cells. RNA-sequencing from DM1- and DM2 iPSC-derived cardiomyocytes revealed distinct misregulation of gene expression, as well as differential aberrant splicing patterns. Together, these data support that DM1 and DM2, despite some shared clinical and molecular features, have distinct pathological signatures.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Proteína MioD/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/patología , Calcio/metabolismo , Línea Celular , Distrofina/metabolismo , Expresión Génica , Variación Genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Distrofia Miotónica/clasificación , Distrofia Miotónica/orina , Empalme del ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
JCI Insight ; 3(9)2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29720576

RESUMEN

Exon skipping uses chemically modified antisense oligonucleotides to modulate RNA splicing. Therapeutically, exon skipping can bypass mutations and restore reading frame disruption by generating internally truncated, functional proteins to rescue the loss of native gene expression. Limb-girdle muscular dystrophy type 2C is caused by autosomal recessive mutations in the SGCG gene, which encodes the dystrophin-associated protein γ-sarcoglycan. The most common SGCG mutations disrupt the transcript reading frame abrogating γ-sarcoglycan protein expression. In order to treat most SGCG gene mutations, it is necessary to skip 4 exons in order to restore the SGCG transcript reading frame, creating an internally truncated protein referred to as Mini-Gamma. Using direct reprogramming of human cells with MyoD, myogenic cells were tested with 2 antisense oligonucleotide chemistries, 2'-O-methyl phosphorothioate oligonucleotides and vivo-phosphorodiamidate morpholino oligomers, to induce exon skipping. Treatment with vivo-phosphorodiamidate morpholino oligomers demonstrated efficient skipping of the targeted exons and corrected the mutant reading frame, resulting in the expression of a functional Mini-Gamma protein. Antisense-induced exon skipping of SGCG occurred in normal cells and those with multiple distinct SGCG mutations, including the most common 521ΔT mutation. These findings demonstrate a multiexon-skipping strategy applicable to the majority of limb-girdle muscular dystrophy 2C patients.


Asunto(s)
Morfolinos/genética , Sarcoglicanopatías/genética , Sarcoglicanopatías/terapia , Sarcoglicanos/genética , Células Cultivadas , Reprogramación Celular , Exones , Fibroblastos/metabolismo , Terapia Genética , Humanos , Microscopía Fluorescente , Mutación , Cultivo Primario de Células , Empalme del ARN , Sistemas de Lectura , Sarcoglicanopatías/metabolismo , Sarcoglicanos/metabolismo , Transducción Genética , Orina/citología
7.
J Clin Invest ; 126(4): 1236-8, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26999602

RESUMEN

Exon skipping uses antisense oligonucleotides (ASOs) to alter transcript splicing for the purpose of rescuing or modulating protein expression. In this issue of the JCI, Lee and colleagues developed and evaluated an ASO-dependent method for treating certain molecularly defined diseases associated with alterations in lamin A/C (LMNA) splicing. Exon skipping by ASOs is gaining traction as a therapeutic strategy, and the use of ASOs is now being applied to bypass mutations and generate modified but functional proteins for an array of genetic disorders.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Lamina Tipo A/biosíntesis , Oligodesoxirribonucleótidos Antisentido/farmacología , Progeria/tratamiento farmacológico , Progeria/metabolismo , ARN Mensajero/metabolismo , Animales , Humanos
8.
Skelet Muscle ; 6: 32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27651888

RESUMEN

BACKGROUND: Cellular models of muscle disease are taking on increasing importance with the large number of genes and mutations implicated in causing myopathies and the concomitant need to test personalized therapies. Developing cell models relies on having an easily obtained source of cells, and if the cells are not derived from muscle itself, a robust reprogramming process is needed. Fibroblasts are a human cell source that works well for the generation of induced pluripotent stem cells, which can then be differentiated into cardiomyocyte lineages, and with less efficiency, skeletal muscle-like lineages. Alternatively, direct reprogramming with the transcription factor MyoD has been used to generate myotubes from cultured human fibroblasts. Although useful, fibroblasts require a skin biopsy to obtain and this can limit their access, especially from pediatric populations. RESULTS: We now demonstrate that direct reprogramming of urine-derived cells is a highly efficient and reproducible process that can be used to establish human myogenic cells. We show that this method can be applied to urine cells derived from normal individuals as well as those with muscle diseases. Furthermore, we show that urine-derived cells can be edited using CRISPR/Cas9 technology. CONCLUSIONS: With progress in understanding the molecular etiology of human muscle diseases, having a readily available, noninvasive source of cells from which to generate muscle-like cells is highly useful.


Asunto(s)
Reprogramación Celular , Desarrollo de Músculos , Enfermedades Musculares/fisiopatología , Proteína MioD/metabolismo , Orina/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Células Clonales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Edición Génica , Humanos , Enfermedades Musculares/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología
9.
J Neuromuscul Dis ; 1(2): 197-206, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26207203

RESUMEN

The New Directions in the Biology and Disease of Skeletal Muscle is a scientific meeting, held every other year, with the stated purpose of bringing together scientists, clinicians, industry representatives and patient advocacy groups to disseminate new discovery useful for treatment inherited forms of neuromuscular disease, primarily the muscular dystrophies. This meeting originated as a response the Muscular Dystrophy Care Act in order to provide a venue for the free exchange of information, with the emphasis on unpublished or newly published data. Highlights of this years' meeting included results from early phase clinical trials for Duchenne Muscular Dystrophy, progress in understanding the epigenetic defects in Fascioscapulohumeral Muscular Dystrophy and new mechanisms of muscle membrane repair. The following is a brief report of the highlights from the conference.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA