Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 31(1): 41-56, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34312665

RESUMEN

Alternative splicing has emerged as a fundamental mechanism for the spatiotemporal control of development. A better understanding of how this mechanism is regulated has the potential not only to elucidate fundamental biological principles, but also to decipher pathological mechanisms implicated in diseases where normal splicing networks are misregulated. Here, we took advantage of human pluripotent stem cells to decipher during human myogenesis the role of muscleblind-like (MBNL) proteins, a family of tissue-specific splicing regulators whose loss of function is associated with myotonic dystrophy type 1 (DM1), an inherited neuromuscular disease. Thanks to the CRISPR/Cas9 technology, we generated human-induced pluripotent stem cells (hiPSCs) depleted in MBNL proteins and evaluated the consequences of their losses on the generation of skeletal muscle cells. Our results suggested that MBNL proteins are required for the late myogenic maturation. In addition, loss of MBNL1 and MBNL2 recapitulated the main features of DM1 observed in hiPSC-derived skeletal muscle cells. Comparative transcriptomic analyses also revealed the muscle-related processes regulated by these proteins that are commonly misregulated in DM1. Together, our study reveals the temporal requirement of MBNL proteins in human myogenesis and should facilitate the identification of new therapeutic strategies capable to cope with the loss of function of these MBNL proteins.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofia Miotónica , Empalme Alternativo , Edición Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Desarrollo de Músculos/genética , Distrofia Miotónica/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
2.
Am J Pathol ; 192(11): 1604-1618, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36113555

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease caused by Dmd mutations, resulting in the absence of dystrophin in skeletal muscle, and a greater susceptibility to damage during contraction (exercise). The current study evaluated whether voluntary exercise impacts a Dmd exon skipping and muscle physiology in a severe DMD murine model. D2-mdx mice were intramuscularly injected with an adeno-associated virus (AAV) U7 snRNA to correct Dmd reading frame, and allowed to voluntary run on a wheel for 1 month. Voluntary running did not induce muscle fiber regeneration, as indicated by the percentage of centronucleated fibers, Myh3 and Myh4 expression, and maximal force production, and thus possibly did not compromise the gene therapy approach. Voluntary running did not impact the number of viral genomes and the expression of U7 and Dmd 1 month after injection of AAV-U7 injected just before exercise initiation, but reduced the amount of dystrophin in dystrophin-expressing fibers from 80% to 65% of the muscle cross-sectional area. In conclusion, voluntary running did not induce muscle damage and had no drastic detrimental effect on the AAV gene therapy exon skipping approach in a severe murine DMD model. Moreover, these results suggest considering exercise as an additional element in the design and conception of future therapeutic approaches for DMD.

3.
Neuropathol Appl Neurobiol ; 49(1): e12876, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36575942

RESUMEN

AIMS: Myotonic dystrophy type I (DM1) is one of the most frequent muscular dystrophies in adults. Although DM1 has long been considered mainly a muscle disorder, growing evidence suggests the involvement of peripheral nerves in the pathogenicity of DM1 raising the question of whether motoneurons (MNs) actively contribute to neuromuscular defects in DM1. METHODS: By using micropatterned 96-well plates as a coculture platform, we generated a functional neuromuscular model combining DM1 and muscleblind protein (MBNL) knock-out human-induced pluripotent stem cells-derived MNs and human healthy skeletal muscle cells. RESULTS: This approach led to the identification of presynaptic defects which affect the formation or stability of the neuromuscular junction at an early developmental stage. These neuropathological defects could be reproduced by the loss of RNA-binding MBNL proteins, whose loss of function in vivo is associated with muscular defects associated with DM1. These experiments indicate that the functional defects associated with MNs can be directly attributed to MBNL family proteins. Comparative transcriptomic analyses also revealed specific neuronal-related processes regulated by these proteins that are commonly misregulated in DM1. CONCLUSIONS: Beyond the application to DM1, our approach to generating a robust and reliable human neuromuscular system should facilitate disease modelling studies and drug screening assays.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofia Miotónica , Adulto , Humanos , Distrofia Miotónica/patología , Proteínas de Unión al ARN/metabolismo , Unión Neuromuscular/patología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas Motoras/patología
4.
Mol Ther ; 30(1): 75-91, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34371182

RESUMEN

CTG repeat expansion (CTGexp) is associated with aberrant alternate splicing that contributes to cardiac dysfunction in myotonic dystrophy type 1 (DM1). Excision of this CTGexp repeat using CRISPR-Cas resulted in the disappearance of punctate ribonuclear foci in cardiomyocyte-like cells derived from DM1-induced pluripotent stem cells (iPSCs). This was associated with correction of the underlying spliceopathy as determined by RNA sequencing and alternate splicing analysis. Certain genes were of particular interest due to their role in cardiac development, maturation, and function (TPM4, CYP2J2, DMD, MBNL3, CACNA1H, ROCK2, ACTB) or their association with splicing (SMN2, GCFC2, MBNL3). Moreover, while comparing isogenic CRISPR-Cas9-corrected versus non-corrected DM1 cardiomyocytes, a prominent difference in the splicing pattern for a number of candidate genes was apparent pertaining to genes that are associated with cardiac function (TNNT, TNNT2, TTN, TPM1, SYNE1, CACNA1A, MTMR1, NEBL, TPM1), cellular signaling (NCOR2, CLIP1, LRRFIP2, CLASP1, CAMK2G), and other DM1-related genes (i.e., NUMA1, MBNL2, LDB3) in addition to the disease-causing DMPK gene itself. Subsequent validation using a selected gene subset, including MBNL1, MBNL2, INSR, ADD3, and CRTC2, further confirmed correction of the spliceopathy following CTGexp repeat excision. To our knowledge, the present study provides the first comprehensive unbiased transcriptome-wide analysis of the differential splicing landscape in DM1 patient-derived cardiac cells after excision of the CTGexp repeat using CRISPR-Cas9, showing reversal of the abnormal cardiac spliceopathy in DM1.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofia Miotónica , Empalme Alternativo , Sistemas CRISPR-Cas , Proteínas de Unión a Calmodulina/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , Proteína Quinasa de Distrofia Miotónica/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma , Expansión de Repetición de Trinucleótido/genética
5.
Clin Exp Pharmacol Physiol ; 50(9): 749-756, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37381823

RESUMEN

Skeletal muscles in animal models of Duchenne muscular dystrophy (DMD) are more susceptible to contraction-induced functional loss, which is not related to fatigue. Valproic acid (VPA) reportedly improves serological and histological markers of damage in dystrophin-deficient murine muscle. Here, we tested whether VPA would reduce the susceptibility to contraction-induced functional loss in two murine DMD models. Adult female mdx (mild) and D2-mdx (severe) DMD murine models were administered VPA (240 mg/kg) or saline for 7 days. Some VPA-treated mdx mice also performed voluntary running in a wheel, which is known to reduce the susceptibility to contraction-induced functional loss; that is, isometric force drop following eccentric contractions. In situ muscle function was assessed before, during and after eccentric contractions. Muscle utrophin and desmin expression were also evaluated using immunoblotting. Interestingly, VPA reduced the isometric force drop following eccentric contractions in both murine models, without change in the relative eccentric maximal force and in the expression of utrophin and desmin. VPA for 7 days combined with voluntary running had no additive effect compared to VPA alone. Furthermore, VPA reduced the absolute isometric maximal force before eccentric contractions in both murine models. The results of our study indicated that VPA in both murine DMD models reduced the susceptibility to contraction-induced functional loss but increased muscle weakness.


Asunto(s)
Distrofia Muscular de Duchenne , Femenino , Animales , Ratones , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Ácido Valproico/farmacología , Ácido Valproico/metabolismo , Ratones Endogámicos mdx , Utrofina/metabolismo , Modelos Animales de Enfermedad , Desmina/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo
6.
J Physiol ; 598(17): 3667-3689, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32515007

RESUMEN

KEY POINTS: Desmin, similar to dystrophin, is associated with costameric structures bridging sarcomeres to the extracellular matrix. Deletion of the desmin gene in mdx mice [double knockout (DKO) mice] induces marked muscle weakness and fatigue resistance compared to mdx mice. Muscle fragility (higher susceptibility to contraction-induced injury) was also aggravated in DKO mice compared to mdx mice. By contrast to mdx mice, the DKO mice did not undergo muscle hypertrophy. Desmin cDNA transfer with adeno-associated virus in newborn mdx mice reduced muscle weakness. Overall, desmin plays important and beneficial roles in muscle wasting, performance and fragility in dystrophic muscle. ABSTRACT: Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease caused by dystrophin deficiency. Desmin, similar to dystrophin, is associated with costameric structures bridging sarcomeres to the extracellular matrix that contributes to muscle function. In the present study, we attempted to provide further insight into the roles of desmin, for which the expression is increased in the muscle from the mouse mdx DMD model. We show that a deletion of the desmin gene (Des) in mdx mice [double knockout (DKO) mice, mdx:desmin-/-] induces a marked muscle weakness; namely, a reduced absolute maximal force production and increased fatigue compared to that in mdx mice. Fragility (i.e. higher susceptibility to contraction-induced injury) was also aggravated in DKO mice compared to mdx mice, despite the promotion of supposedly less fragile muscle fibres in DKO mice, and this worsening of fragility was related to a decreased muscle excitability. Moreover, in contrast to mdx mice, the DKO mice did not undergo muscle hypertrophy, as indicated by smaller and fewer fibres, with a reduced percentage of centronucleated fibres, potentially explaining the severe muscle weakness. Notably, Desmin cDNA transfer with adeno-associated virus in newborn mdx mice improved specific maximal force normalized to muscle weight. Overall, desmin plays important and beneficial roles in muscle wasting, performance and fragility in dystrophic mdx mice, which differ, at least in part, from those observed in healthy muscle.


Asunto(s)
Músculo Esquelético , Distrofia Muscular de Duchenne , Animales , Desmina/genética , Modelos Animales de Enfermedad , Distrofina/genética , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/genética
7.
Mol Ther ; 27(8): 1372-1388, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31253581

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by a CTG repeat expansion located in the 3' UTR of the DMPK gene. Expanded DMPK transcripts aggregate into nuclear foci and alter the function of RNA-binding proteins, leading to defects in the alternative splicing of numerous pre-mRNAs. To date, there is no curative treatment for DM1. Here we investigated a gene-editing strategy using the CRISPR-Cas9 system from Staphylococcus aureus (Sa) to delete the CTG repeats in the human DMPK locus. Co-expression of SaCas9 and selected pairs of single-guide RNAs (sgRNAs) in cultured DM1 patient-derived muscle line cells carrying 2,600 CTG repeats resulted in targeted DNA deletion, ribonucleoprotein foci disappearance, and correction of splicing abnormalities in various transcripts. Furthermore, a single intramuscular injection of recombinant AAV vectors expressing CRISPR-SaCas9 components in the tibialis anterior muscle of DMSXL (myotonic dystrophy mouse line carrying the human DMPK gene with >1,000 CTG repeats) mice decreased the number of pathological RNA foci in myonuclei. These results establish the proof of concept that genome editing of a large trinucleotide expansion is feasible in muscle and may represent a useful strategy to be further developed for the treatment of myotonic dystrophy.


Asunto(s)
Edición Génica , Proteína Quinasa de Distrofia Miotónica/genética , ARN Nuclear , Expansión de Repetición de Trinucleótido , Empalme Alternativo , Animales , Secuencia de Bases , Sistemas CRISPR-Cas , Núcleo Celular , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Marcación de Gen , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , ARN Guía de Kinetoplastida , Transducción Genética
8.
Nucleic Acids Res ; 46(16): 8275-8298, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-29947794

RESUMEN

CRISPR/Cas9 is an attractive platform to potentially correct dominant genetic diseases by gene editing with unprecedented precision. In the current proof-of-principle study, we explored the use of CRISPR/Cas9 for gene-editing in myotonic dystrophy type-1 (DM1), an autosomal-dominant muscle disorder, by excising the CTG-repeat expansion in the 3'-untranslated-region (UTR) of the human myotonic dystrophy protein kinase (DMPK) gene in DM1 patient-specific induced pluripotent stem cells (DM1-iPSC), DM1-iPSC-derived myogenic cells and DM1 patient-specific myoblasts. To eliminate the pathogenic gain-of-function mutant DMPK transcript, we designed a dual guide RNA based strategy that excises the CTG-repeat expansion with high efficiency, as confirmed by Southern blot and single molecule real-time (SMRT) sequencing. Correction efficiencies up to 90% could be attained in DM1-iPSC as confirmed at the clonal level, following ribonucleoprotein (RNP) transfection of CRISPR/Cas9 components without the need for selective enrichment. Expanded CTG repeat excision resulted in the disappearance of ribonuclear foci, a quintessential cellular phenotype of DM1, in the corrected DM1-iPSC, DM1-iPSC-derived myogenic cells and DM1 myoblasts. Consequently, the normal intracellular localization of the muscleblind-like splicing regulator 1 (MBNL1) was restored, resulting in the normalization of splicing pattern of SERCA1. This study validates the use of CRISPR/Cas9 for gene editing of repeat expansions.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Mioblastos/metabolismo , Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido/genética , Células Cultivadas , Niño , Femenino , Humanos , Persona de Mediana Edad , Desarrollo de Músculos/genética , Distrofia Miotónica/metabolismo , Distrofia Miotónica/patología
9.
Am J Pathol ; 188(11): 2662-2673, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30142334

RESUMEN

Dystrophin deficiency in mdx mice, a model for Duchenne muscular dystrophy, leads to muscle weakness revealed by a reduced specific maximal force as well as fragility (ie, higher susceptibility to contraction-induced injury, as shown by a greater force decrease after lengthening contractions). Both symptoms could be improved with dystrophin restoration-based therapies and long-term (months) voluntary exercise. Herein, we evaluated the effect of short-term (1-week) voluntary wheel running. We found that running improved fragility of tibialis anterior muscle (TA), but not plantaris muscle, independently of utrophin up-regulation, without affecting weakness. Moreover, TA muscle excitability was also preserved by running, as shown by compound muscle action potential measurements after lengthening contractions. Of interest, the calcineurin inhibitor cyclosporin A prevented the effect of running on both muscle fragility and excitability. Cyclosporin also prevented the running-induced changes in expression of genes involved in excitability (Scn4a and Cacna1s) and slower contractile phenotype (Myh2 and Tnni1) in TA muscle. In conclusion, short-term voluntary exercise improves TA muscle fragility in mdx mice, without worsening weakness. Its effect was related to preserved excitability, calcineurin pathway activation, and changes in the program of genes involved in excitability and slower contractile phenotype. Thus, remediation of muscle fragility of Duchenne muscular dystrophy patients through appropriate exercise training deserves to be explored in more detail.


Asunto(s)
Calcineurina/metabolismo , Distrofia Muscular Animal/prevención & control , Condicionamiento Físico Animal , Animales , Ratones , Ratones Endogámicos mdx , Actividad Motora , Contracción Muscular , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología
10.
Nat Chem Biol ; 13(2): 188-193, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27941760

RESUMEN

Excluding the ribosome and riboswitches, developing small molecules that selectively target RNA is a longstanding problem in chemical biology. A typical cellular RNA is difficult to target because it has little tertiary, but abundant secondary structure. We designed allele-selective compounds that target such an RNA, the toxic noncoding repeat expansion (r(CUG)exp) that causes myotonic dystrophy type 1 (DM1). We developed several strategies to generate allele-selective small molecules, including non-covalent binding, covalent binding, cleavage and on-site probe synthesis. Covalent binding and cleavage enabled target profiling in cells derived from individuals with DM1, showing precise recognition of r(CUG)exp. In the on-site probe synthesis approach, small molecules bound adjacent sites in r(CUG)exp and reacted to afford picomolar inhibitors via a proximity-based click reaction only in DM1-affected cells. We expanded this approach to image r(CUG)exp in its natural context.


Asunto(s)
ARN/química , ARN/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Expansión de Repetición de Trinucleótido/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , ARN/genética , Empalme del ARN/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/síntesis química , Relación Estructura-Actividad
11.
Int J Mol Sci ; 20(8)2019 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-31010208

RESUMEN

Circular RNAs (circRNAs) constitute a recently re-discovered class of non-coding RNAs functioning as sponges for miRNAs and proteins, affecting RNA splicing and regulating transcription. CircRNAs are generated by "back-splicing", which is the linking covalently of 3'- and 5'-ends of exons. Thus, circRNA levels might be deregulated in conditions associated with altered RNA-splicing. Significantly, growing evidence indicates their role in human diseases. Specifically, myotonic dystrophy type 1 (DM1) is a multisystemic disorder caused by expanded CTG repeats in the DMPK gene which results in abnormal mRNA-splicing. In this investigation, circRNAs expressed in DM1 skeletal muscles were identified by analyzing RNA-sequencing data-sets followed by qPCR validation. In muscle biopsies, out of nine tested, four transcripts showed an increased circular fraction: CDYL, HIPK3, RTN4_03, and ZNF609. Their circular fraction values correlated with skeletal muscle strength and with splicing biomarkers of disease severity, and displayed higher values in more severely affected patients. Moreover, Receiver-Operating-Characteristics curves of these four circRNAs discriminated DM1 patients from controls. The identified circRNAs were also detectable in peripheral-blood-mononuclear-cells (PBMCs) and the plasma of DM1 patients, but they were not regulated significantly. Finally, increased circular fractions of RTN4_03 and ZNF609 were also observed in differentiated myogenic cell lines derived from DM1 patients. In conclusion, this pilot study identified circRNA dysregulation in DM1 patients.


Asunto(s)
Regulación de la Expresión Génica , Distrofia Miotónica/genética , ARN/genética , Adulto , Empalme Alternativo/genética , Estudios de Casos y Controles , Línea Celular , Femenino , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Miotónica/sangre , Reacción en Cadena de la Polimerasa , ARN/sangre , ARN Circular , Reproducibilidad de los Resultados
12.
Mol Ther ; 25(1): 24-43, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129118

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by (CTG⋅CAG)n-repeat expansion within the DMPK gene and thought to be mediated by a toxic RNA gain of function. Current attempts to develop therapy for this disease mainly aim at destroying or blocking abnormal properties of mutant DMPK (CUG)n RNA. Here, we explored a DNA-directed strategy and demonstrate that single clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-cleavage in either its 5' or 3' unique flank promotes uncontrollable deletion of large segments from the expanded trinucleotide repeat, rather than formation of short indels usually seen after double-strand break repair. Complete and precise excision of the repeat tract from normal and large expanded DMPK alleles in myoblasts from unaffected individuals, DM1 patients, and a DM1 mouse model could be achieved at high frequency by dual CRISPR/Cas9-cleavage at either side of the (CTG⋅CAG)n sequence. Importantly, removal of the repeat appeared to have no detrimental effects on the expression of genes in the DM1 locus. Moreover, myogenic capacity, nucleocytoplasmic distribution, and abnormal RNP-binding behavior of transcripts from the edited DMPK gene were normalized. Dual sgRNA-guided excision of the (CTG⋅CAG)n tract by CRISPR/Cas9 technology is applicable for developing isogenic cell lines for research and may provide new therapeutic opportunities for patients with DM1.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Inestabilidad Genómica , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido , Repeticiones de Trinucleótidos , Animales , Proteínas Bacterianas/genética , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Codón , Modelos Animales de Enfermedad , Endonucleasas/genética , Fibroblastos/metabolismo , Expresión Génica , Orden Génico , Sitios Genéticos , Humanos , Ratones , ARN Guía de Kinetoplastida , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia
13.
J Cell Sci ; 128(19): 3631-45, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26272918

RESUMEN

Twist-1 is mostly expressed during development and has been previously shown to control myogenesis. Because its regulation in muscle has not been fully exploited, the aim of this project was to identify micro (mi)RNAs in muscle that regulate Twist-1. miR-206, one of the most important muscle-specific miRNAs (myomiRs), was identified as a possible regulator of Twist-1 mRNA. Luciferase assays and transfections in human foetal myoblasts showed that Twist-1 is a direct target of miR-206 and that through this pathway muscle cell differentiation is promoted. We next investigated whether MyoD, a major myogenic transcription factor, regulates Twist-1 because it is known that MyoD induces expression of the miR-206 gene. We found that forced MyoD expression induced miR-206 upregulation and Twist-1 downregulation through binding to the miR-206 promoter, followed by increased muscle cell differentiation. Finally, experiments were performed in muscle cells from subjects with congenital myotonic dystrophy type 1, in which myoblasts fail to differentiate into myotubes. MyoD overexpression inhibited Twist-1 through miR-206 induction, which was followed by an increase in muscle cell differentiation. These results reveal a previously unidentified mechanism of myogenesis that might also play an important role in muscle disease.


Asunto(s)
MicroARNs/genética , Proteína MioD/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Inmunoprecipitación de Cromatina , Humanos , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Proteína MioD/genética , Mioblastos/citología , Mioblastos/metabolismo
14.
Muscle Nerve ; 55(2): 254-261, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27312354

RESUMEN

INTRODUCTION: The effect of constitutive inactivation of the gene encoding myostatin on the gain in muscle performance during postnatal growth has not been well characterized. METHODS: We analyzed 2 murine myostatin knockout (KO) models, (i) the Lee model (KOLee ) and (ii) the Grobet model (KOGrobet ), and measured the contraction of tibialis anterior muscle in situ. RESULTS: Absolute maximal isometric force was increased in 6-month-old KOLee and KOGrobet mice, as compared to wild-type mice. Similarly, absolute maximal power was increased in 6-month-old KOLee mice. In contrast, specific maximal force (relative maximal force per unit of muscle mass was decreased in all 6-month-old male and female KO mice, except in 6-month-old female KOGrobet mice, whereas specific maximal power was reduced only in male KOLee mice. CONCLUSIONS: Genetic inactivation of myostatin increases maximal force and power, but in return it reduces muscle quality, particularly in male mice. Muscle Nerve 55: 254-261, 2017.


Asunto(s)
Contracción Muscular/genética , Fuerza Muscular/genética , Músculo Esquelético/fisiología , Enfermedades Musculares/patología , Miostatina/deficiencia , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Enfermedades Musculares/genética , Miostatina/genética , Factores Sexuales
15.
Hum Mol Genet ; 23(15): 4125-33, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24659496

RESUMEN

While transfer of a protein encoded by a single nucleus to nearby nuclei in multinucleated cells has been known for almost 25 years, the biological consequences for gain-of-function diseases have not been considered. Here, we have investigated nuclear protein spreading and its potential consequences in two of the three most prevalent neuromuscular diseases. By performing co-cultures between diseased or control human myoblasts and murine C2C12 myoblasts, we demonstrate that in facioscapulohumeral dystrophy, although the transcription of the toxic protein DUX4 occurs in only a limited number of nuclei, the resulting protein diffuses into nearby nuclei within the myotubes, thus spreading aberrant gene expression. In myotonic dystrophy type 1, we observed that in human-mouse heterokaryons, the expression of a mutated DMPK from human nuclei titrates splicing factors produced by neighboring nuclei, inducing the mis-splicing of several pre-mRNAs in murine nuclei. In both cases, the spreading of the pathological phenotypes from one nucleus to another is observed, highlighting an additional mechanism that contributes to the dissemination and worsening of the muscle pathogenesis. These results indicate that nuclear protein spreading may be an important component of pathophysiology of gain of function muscular diseases which should be taken into consideration in the design of new therapeutic approaches.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Homeodominio/genética , Distrofia Muscular Facioescapulohumeral/genética , Mioblastos/metabolismo , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Transporte Activo de Núcleo Celular , Animales , Técnicas de Cocultivo , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapulohumeral/metabolismo , Distrofia Muscular Facioescapulohumeral/patología , Mioblastos/patología , Distrofia Miotónica/metabolismo , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica/metabolismo , Transporte de Proteínas , Empalme del ARN , Transcripción Genética
16.
Hum Mol Genet ; 23(6): 1551-62, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24179176

RESUMEN

Myotonic dystrophy (DM) is a multi-system neuromuscular disorder for which there is no treatment. We have developed a medium throughput phenotypic assay, based on the identification of nuclear foci in DM patient cell lines using in situ hybridization and high-content imaging to screen for potentially useful therapeutic compounds. A series of further assays based on molecular features of DM have also been employed. Two compounds that reduce and/or remove nuclear foci have been identified, Ro 31-8220 and chromomycin A3. Ro 31-8220 is a PKC inhibitor, previously shown to affect the hyperphosphorylation of CELF1 and ameliorate the cardiac phenotype in a DM1 mouse model. We show that the same compound eliminates nuclear foci, reduces MBNL1 protein in the nucleus, affects ATP2A1 alternative splicing and reduces steady-state levels of CELF1 protein. We demonstrate that this effect is independent of PKC activity and conclude that this compound may be acting on alternative kinase targets within DM pathophysiology. Understanding the activity profile for this compound is key for the development of targeted therapeutics in the treatment of DM.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Cromomicina A3/farmacología , Indoles/farmacología , Distrofia Miotónica/patología , Proteínas de Unión al ARN/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Empalme Alternativo , Animales , Proteínas CELF1 , Núcleo Celular/patología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Biblioteca de Péptidos , Proteínas de Unión al ARN/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Pez Cebra
17.
Am J Pathol ; 185(7): 2012-24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26009153

RESUMEN

There is fear that mechanical overloading (OVL; ie, high-force contractions) accelerates Duchenne muscular dystrophy. Herein, we determined whether short-term OVL combined with wheel running, short-term OVL combined with irradiation, and long-term OVL are detrimental for hind limb mdx mouse muscle, a murine model of Duchene muscular dystrophy exhibiting milder dystrophic features. OVL was induced by the surgical ablation of the synergic muscles of the plantaris muscle, a fast muscle susceptible to contraction-induced muscle damage in mdx mice. We found that short-term OVL combined with wheel and long-term OVL did not worsen the deficit in specific maximal force (ie, absolute maximal force normalized to muscle size) and histological markers of muscle damage (percentage of regenerating fibers and fibrosis) in mdx mice. Moreover, long-term OVL did not increase the alteration in calcium homeostasis and did not deplete muscle cell progenitors expressing Pax 7 in mdx mice. Irradiation before short-term OVL, which is believed to inhibit muscle regeneration, was not more detrimental to mdx than control mice. Interestingly, short-term OVL combined with wheel and long-term OVL markedly improved the susceptibility to contraction-induced damage, increased absolute maximal force, induced hypertrophy, and promoted a slower, more oxidative phenotype. Together, these findings indicate that OVL is beneficial to mdx muscle, and muscle regeneration does not mask the potentially detrimental effect of OVL.


Asunto(s)
Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Hipertrofia , Extremidad Inferior , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Actividad Motora , Contracción Muscular , Músculo Esquelético/efectos de la radiación , Mutación , Regeneración , Células Satélite del Músculo Esquelético/fisiología , Células Satélite del Músculo Esquelético/efectos de la radiación
18.
Hum Mol Genet ; 22(25): 5188-98, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23922231

RESUMEN

Myotonic dystrophy type 1 (DM1) is an RNA-mediated disorder caused by a non-coding CTG repeat expansion that, in particular, provokes functional alteration of CUG-binding proteins. As a consequence, several genes with misregulated alternative splicing have been linked to clinical symptoms. In our search for additional molecular mechanisms that would trigger functional defects in DM1, we took advantage of mutant gene-carrying human embryonic stem cell lines to identify differentially expressed genes. Among the different genes found to be misregulated by DM1 mutation, one strongly downregulated gene encodes a transcription factor, ZNF37A. In this paper, we show that this defect in expression, which derives from a loss of RNA stability, is controlled by the RNA-binding protein, CUGBP1, and is associated with impaired myogenesis-a functional defect reminiscent of that observed in DM1. Loss of the ZNF37A protein results in changes in the expression of the subunit α1 of the receptor for the interleukin 13. This suggests that the pathological molecular mechanisms linking ZNF37A and myogenesis may involve the signaling pathway that is known to promote myoblast recruitment during development and regeneration.


Asunto(s)
Empalme Alternativo/genética , Factores de Transcripción de Tipo Kruppel/genética , Desarrollo de Músculos/genética , Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido/genética , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Madre Embrionarias , Humanos , Subunidad alfa1 del Receptor de Interleucina-13/genética , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Mutación , Distrofia Miotónica/fisiopatología , Transducción de Señal/genética
19.
PLoS Genet ; 8(11): e1003043, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209425

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by an unstable CTG repeat expansion in the 3'UTR of the DM protein kinase (DMPK) gene. DMPK transcripts carrying CUG expansions form nuclear foci and affect splicing regulation of various RNA transcripts. Furthermore, bidirectional transcription over the DMPK gene and non-conventional RNA translation of repeated transcripts have been described in DM1. It is clear now that this disease may involve multiple pathogenic pathways including changes in gene expression, RNA stability and splicing regulation, protein translation, and micro-RNA metabolism. We previously generated transgenic mice with 45-kb of the DM1 locus and >300 CTG repeats (DM300 mice). After successive breeding and a high level of CTG repeat instability, we obtained transgenic mice carrying >1,000 CTG (DMSXL mice). Here we described for the first time the expression pattern of the DMPK sense transcripts in DMSXL and human tissues. Interestingly, we also demonstrate that DMPK antisense transcripts are expressed in various DMSXL and human tissues, and that both sense and antisense transcripts accumulate in independent nuclear foci that do not co-localize together. Molecular features of DM1-associated RNA toxicity in DMSXL mice (such as foci accumulation and mild missplicing), were associated with high mortality, growth retardation, and muscle defects (abnormal histopathology, reduced muscle strength, and lower motor performances). We have found that lower levels of IGFBP-3 may contribute to DMSXL growth retardation, while increased proteasome activity may affect muscle function. These data demonstrate that the human DM1 locus carrying very large expansions induced a variety of molecular and physiological defects in transgenic mice, reflecting DM1 to a certain extent. As a result, DMSXL mice provide an animal tool to decipher various aspects of the disease mechanisms. In addition, these mice can be used to test the preclinical impact of systemic therapeutic strategies on molecular and physiological phenotypes.


Asunto(s)
Músculo Esquelético , Distrofia Miotónica , Proteínas Serina-Treonina Quinasas/genética , Animales , Núcleo Celular/metabolismo , Endopeptidasas/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/fisiopatología , Distrofia Miotónica/genética , Distrofia Miotónica/fisiopatología , Proteína Quinasa de Distrofia Miotónica , Proteínas Serina-Treonina Quinasas/metabolismo , Empalme del ARN , Expansión de Repetición de Trinucleótido/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA