Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
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
2.
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
3.
Brain ; 136(Pt 3): 957-70, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23404338

RESUMEN

Myotonic dystrophy type 1 is a complex multisystemic inherited disorder, which displays multiple debilitating neurological manifestations. Despite recent progress in the understanding of the molecular pathogenesis of myotonic dystrophy type 1 in skeletal muscle and heart, the pathways affected in the central nervous system are largely unknown. To address this question, we studied the only transgenic mouse line expressing CTG trinucleotide repeats in the central nervous system. These mice recreate molecular features of RNA toxicity, such as RNA foci accumulation and missplicing. They exhibit relevant behavioural and cognitive phenotypes, deficits in short-term synaptic plasticity, as well as changes in neurochemical levels. In the search for disease intermediates affected by disease mutation, a global proteomics approach revealed RAB3A upregulation and synapsin I hyperphosphorylation in the central nervous system of transgenic mice, transfected cells and post-mortem brains of patients with myotonic dystrophy type 1. These protein defects were associated with electrophysiological and behavioural deficits in mice and altered spontaneous neurosecretion in cell culture. Taking advantage of a relevant transgenic mouse of a complex human disease, we found a novel connection between physiological phenotypes and synaptic protein dysregulation, indicative of synaptic dysfunction in myotonic dystrophy type 1 brain pathology.


Asunto(s)
Conducta Animal/fisiología , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Adulto , Anciano , Animales , Western Blotting , Electroforesis en Gel Bidimensional , Electrofisiología , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofia Miotónica/complicaciones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Expansión de Repetición de Trinucleótido
4.
Proc Natl Acad Sci U S A ; 108(1): 260-5, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21173221

RESUMEN

Trinucleotide expansions cause disease by both protein- and RNA-mediated mechanisms. Unexpectedly, we discovered that CAG expansion constructs express homopolymeric polyglutamine, polyalanine, and polyserine proteins in the absence of an ATG start codon. This repeat-associated non-ATG translation (RAN translation) occurs across long, hairpin-forming repeats in transfected cells or when expansion constructs are integrated into the genome in lentiviral-transduced cells and brains. Additionally, we show that RAN translation across human spinocerebellar ataxia type 8 (SCA8) and myotonic dystrophy type 1 (DM1) CAG expansion transcripts results in the accumulation of SCA8 polyalanine and DM1 polyglutamine expansion proteins in previously established SCA8 and DM1 mouse models and human tissue. These results have implications for understanding fundamental mechanisms of gene expression. Moreover, these toxic, unexpected, homopolymeric proteins now should be considered in pathogenic models of microsatellite disorders.


Asunto(s)
Biosíntesis de Proteínas/genética , Ataxias Espinocerebelosas/genética , Expansión de Repetición de Trinucleótido/genética , Secuencia de Aminoácidos , Northern Blotting , Línea Celular , Clonación Molecular , Codón Iniciador/genética , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Lentivirus , Espectrometría de Masas , Datos de Secuencia Molecular , Mutagénesis , Distrofia Miotónica/genética , Péptidos/genética , Péptidos/metabolismo , Biosíntesis de Proteínas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
PLoS Genet ; 3(4): e52, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-17411343

RESUMEN

Trinucleotide repeat expansions are the genetic cause of numerous human diseases, including fragile X mental retardation, Huntington disease, and myotonic dystrophy type 1. Disease severity and age of onset are critically linked to expansion size. Previous mouse models of repeat instability have not recreated large intergenerational expansions ("big jumps"), observed when the repeat is transmitted from one generation to the next, and have never attained the very large tract lengths possible in humans. Here, we describe dramatic intergenerational CTG*CAG repeat expansions of several hundred repeats in a transgenic mouse model of myotonic dystrophy type 1, resulting in increasingly severe phenotypic and molecular abnormalities. Homozygous mice carrying over 700 trinucleotide repeats on both alleles display severely reduced body size and splicing abnormalities, notably in the central nervous system. Our findings demonstrate that large intergenerational trinucleotide repeat expansions can be recreated in mice, and endorse the use of transgenic mouse models to refine our understanding of triplet repeat expansion and the resulting pathogenesis.


Asunto(s)
Expansión de Repetición de Trinucleótido/genética , Alelos , Animales , Secuencia de Bases , Tamaño Corporal/genética , Inestabilidad Genómica/genética , Homocigoto , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Empalme del ARN/genética
6.
Elife ; 82019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31829940

RESUMEN

Cardiac conduction defects decrease life expectancy in myotonic dystrophy type 1 (DM1), a CTG repeat disorder involving misbalance between two RNA binding factors, MBNL1 and CELF1. However, how DM1 condition translates into conduction disorders remains poorly understood. Here we simulated MBNL1 and CELF1 misbalance in the Drosophila heart and performed TU-tagging-based RNAseq of cardiac cells. We detected deregulations of several genes controlling cellular calcium levels, including increased expression of straightjacket/α2δ3, which encodes a regulatory subunit of a voltage-gated calcium channel. Straightjacket overexpression in the fly heart leads to asynchronous heartbeat, a hallmark of abnormal conduction, whereas cardiac straightjacket knockdown improves these symptoms in DM1 fly models. We also show that ventricular α2δ3 expression is low in healthy mice and humans, but significantly elevated in ventricular muscles from DM1 patients with conduction defects. These findings suggest that reducing ventricular straightjacket/α2δ3 levels could offer a strategy to prevent conduction defects in DM1.


Asunto(s)
Canales de Calcio/biosíntesis , Trastorno del Sistema de Conducción Cardíaco/genética , Trastorno del Sistema de Conducción Cardíaco/fisiopatología , Regulación de la Expresión Génica , Distrofia Miotónica/complicaciones , Animales , Canales de Calcio/genética , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Humanos , Ratones
7.
Biochim Biophys Acta ; 1772(11-12): 1183-91, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17950578

RESUMEN

Myotonic dystrophy (DM1) is a dominant autosomal multisystemic disorder caused by the expansion of an unstable CTG trinucleotide repeat in the 3' untranslated region of the DMPK gene. Nuclear accumulation of the enlarged CUG-containing DMPK transcripts has a deleterious effect on the regulation of alternative splicing of some RNAs and has a central role in causing the symptoms of DM1. In particular, Insulin Receptor (IR) mRNA splicing defects have been observed in the muscle of DM1 patients. In this study, we have investigated IR splicing in insulin-responsive tissues (i.e. skeletal muscles, adipose tissue, liver) and pancreas and we have studied glucose metabolism in mice carrying the human genomic DM1 region with expanded (>350 CTG) or normal (20 CTG) repeats and in wild-type mice. Mice carrying DM1 expansions displayed a tissue- and age-dependent abnormal regulation of IR mRNA splicing in all the tissues that we investigated. Furthermore, these mice showed a basal hyperglycemia and glucose intolerance which disappeared with age. Our findings show that deregulation of IR splicing due to the DM1 mutation can occur in different mouse tissues, suggesting that CTG repeat expansions might also result in IR misplicing not only in muscles but also in other tissues in DM1 patients.


Asunto(s)
Distrofia Miotónica/genética , Receptor de Insulina/genética , Expansión de Repetición de Trinucleótido/genética , Envejecimiento , Empalme Alternativo/genética , Animales , Perfilación de la Expresión Génica , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Hipotálamo/metabolismo , Insulina/metabolismo , Secreción de Insulina , Ratones , Ratones Transgénicos , Proteínas Mutantes/metabolismo , Proteína Quinasa de Distrofia Miotónica , Especificidad de Órganos , Páncreas/enzimología , Páncreas/patología , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Insulina/metabolismo , Transgenes
8.
J Nucleic Acids ; 2013: 567435, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24455202

RESUMEN

An expanded CTG-repeat in the 3' UTR of the DMPK gene is responsible for myotonic dystrophy type I (DM1). Somatic and intergenerational instability cause the disease to become more severe during life and in subsequent generations. Evidence is accumulating that trinucleotide repeat instability and disease progression involve aberrant chromatin dynamics. We explored the chromatin environment in relation to expanded CTG-repeat tracts in hearts from transgenic mice carrying the DM1 locus with different repeat lengths. Using bisulfite sequencing we detected abundant CpG methylation in the regions flanking the expanded CTG-repeat. CpG methylation was postulated to affect CTCF binding but we found that CTCF binding is not affected by CTG-repeat length in our transgenic mice. We detected significantly decreased DMPK sense and SIX5 transcript expression levels in mice with expanded CTG-repeats. Expression of the DM1 antisense transcript was barely affected by CTG-repeat expansion. In line with altered gene expression, ChIP studies revealed a locally less active chromatin conformation around the expanded CTG-repeat, namely, decreased enrichment of active histone mark H3K9/14Ac and increased H3K9Me3 enrichment (repressive chromatin mark). We also observed binding of PCNA around the repeats, a candidate that could launch chromatin remodelling cascades at expanded repeats, ultimately affecting gene transcription and repeat instability.

9.
Neuromuscul Disord ; 23(12): 1016-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24139022

RESUMEN

Myotonic dystrophy is the most common adult muscle dystrophy. In view of emerging therapies, which use animal models as a proof of principle, the development of reliable outcome measures for in vivo longitudinal study of mouse skeletal muscle function is becoming crucial. To satisfy this need, we have developed a device to measure ankle dorsi- and plantarflexion torque in rodents. We present an in vivo 8-month longitudinal study of the contractile properties of the skeletal muscles of the DMSXL mouse model of myotonic dystrophy type 1. Between 4 and 12 months of age, we observed a reduction in muscle strength in the ankle dorsi- and plantarflexors of DMSXL compared to control mice although the strength per muscle cross-section was normal. Mild steady myotonia but no abnormal muscle fatigue was also observed in the DMSXL mice. Magnetic resonance imaging and histological analysis performed at the end of the study showed respectively reduced muscle cross-section area and smaller muscle fibre diameter in DMSXL mice. In conclusion, our study demonstrates the feasibility of carrying out longitudinal in vivo studies of muscle function over several months in a mouse model of myotonic dystrophy confirming the feasibility of this method to test preclinical therapeutics.


Asunto(s)
Músculo Esquelético/patología , Distrofia Miotónica/patología , Proteínas Serina-Treonina Quinasas/genética , Factores de Edad , Análisis de Varianza , Animales , Peso Corporal/genética , Modelos Animales de Enfermedad , Miembro Posterior/patología , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética , Ratones , Ratones Transgénicos , Fibras Musculares Esqueléticas/patología , Dinamómetro de Fuerza Muscular , Distrofia Miotónica/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Expansión de Repetición de Trinucleótido/genética
10.
Rare Dis ; 1: e25553, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25003003

RESUMEN

The toxicity of expanded transcripts in myotonic dystrophy type 1 (DM1) is mainly mediated by the disruption of alternative splicing. However, the detailed disease mechanisms in the central nervous system (CNS) have not been fully elucidated. In our recent study, we demonstrated that the accumulation of mutant transcripts in the CNS of a mouse model of DM1 disturbs splicing in a region-specific manner. We now discuss that the spatial- and temporal-regulated expression of splicing factors may contribute to the region-specific spliceopathy in DM1 brains. In the search for disease mechanisms operating in the CNS, we found that the expression of expanded CUG-containing RNA affects the expression and phosphorylation of synaptic vesicle proteins, possibly contributing to DM1 neurological phenotypes. Although mediated by splicing regulators with a described role in DM1, the misregulation of synaptic proteins was not associated with missplicing of their coding transcripts, supporting the view that DM1 mechanisms in the CNS have also far-reaching implications beyond the disruption of a splicing program.

11.
Neuromuscul Disord ; 20(5): 319-25, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20346670

RESUMEN

Myotonic dystrophy type 1 (DM1) is a neuromuscular disease caused by the expansion of a CTG repeat in the DMPK gene and characterised by progressive skeletal muscle weakness and wasting. To investigate the effects of the CTG expansion on the physiological function of the skeletal muscles, we have used a transgenic mouse model carrying the human DM1 region with 550 expanded CTG repeats. Maximal force is reduced in the skeletal muscles of 10-month-old but not in 3-month-old DM1 mice when compared to age-matched non-transgenic littermates. The progressive weakness observed in the DM1 mice is directly related to the reduced muscle mass and muscle fibre size. A significant increase in trypsin-like proteasome activity and Fbxo32 expression is also measured in the DM1 muscles indicating that an atrophic process mediated by the ubiquitin-proteasome pathway may contribute to the progressive muscle wasting and weakness in the DM1 mice.


Asunto(s)
Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Distrofia Miotónica/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Expansión de Repetición de Trinucleótido/genética , Ubiquitina/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fuerza de la Mano/fisiología , Humanos , Laminina/metabolismo , Ratones , Ratones Transgénicos , Proteínas Musculares/metabolismo , Fuerza Muscular/genética , Debilidad Muscular/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal/genética , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA