Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
N Engl J Med ; 373(13): 1251-61, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26398074

RESUMEN

A 50-year-old man with a history of cardiomyopathy and progressive muscle weakness was admitted with cardiogenic shock. Electroencephalography showed total suppression of cerebral activity; ventilator support was withdrawn, and he died. An autopsy was performed.


Asunto(s)
Músculo Esquelético/patología , Distrofia Miotónica/diagnóstico , Choque Cardiogénico/etiología , Diagnóstico Diferencial , Ecocardiografía , Electroencefalografía , Resultado Fatal , Paro Cardíaco/etiología , Humanos , Pulmón/diagnóstico por imagen , Pulmón/patología , Masculino , Persona de Mediana Edad , Debilidad Muscular/etiología , Distrofia Miotónica/complicaciones , Radiografía
2.
Nature ; 488(7409): 111-5, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22859208

RESUMEN

Antisense oligonucleotides (ASOs) hold promise for gene-specific knockdown in diseases that involve RNA or protein gain-of-function effects. In the hereditary degenerative disease myotonic dystrophy type 1 (DM1), transcripts from the mutant allele contain an expanded CUG repeat and are retained in the nucleus. The mutant RNA exerts a toxic gain-of-function effect, making it an appropriate target for therapeutic ASOs. However, despite improvements in ASO chemistry and design, systemic use of ASOs is limited because uptake in many tissues, including skeletal and cardiac muscle, is not sufficient to silence target messenger RNAs. Here we show that nuclear-retained transcripts containing expanded CUG (CUG(exp)) repeats are unusually sensitive to antisense silencing. In a transgenic mouse model of DM1, systemic administration of ASOs caused a rapid knockdown of CUG(exp) RNA in skeletal muscle, correcting the physiological, histopathologic and transcriptomic features of the disease. The effect was sustained for up to 1 year after treatment was discontinued. Systemically administered ASOs were also effective for muscle knockdown of Malat1, a long non-coding RNA (lncRNA) that is retained in the nucleus. These results provide a general strategy to correct RNA gain-of-function effects and to modulate the expression of expanded repeats, lncRNAs and other transcripts with prolonged nuclear residence.


Asunto(s)
Núcleo Celular/genética , Silenciador del Gen , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , ARN/antagonistas & inhibidores , ARN/genética , Alelos , Animales , Secuencia de Bases , Núcleo Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofia Miotónica/patología , Distrofia Miotónica/fisiopatología , Proteína Quinasa de Distrofia Miotónica , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , ARN/metabolismo , ARN Largo no Codificante , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , Ribonucleasa H/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Expansión de Repetición de Trinucleótido/genética
3.
J Pharmacol Exp Ther ; 355(2): 329-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26330536

RESUMEN

Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults. DM1 is caused by an expanded CTG repeat in the 3'-untranslated region of DMPK, the gene encoding dystrophia myotonica protein kinase (DMPK). Antisense oligonucleotides (ASOs) containing 2',4'-constrained ethyl-modified (cEt) residues exhibit a significantly increased RNA binding affinity and in vivo potency relative to those modified with other 2'-chemistries, which we speculated could translate to enhanced activity in extrahepatic tissues, such as muscle. Here, we describe the design and characterization of a cEt gapmer DMPK ASO (ISIS 486178), with potent activity in vitro and in vivo against mouse, monkey, and human DMPK. Systemic delivery of unformulated ISIS 486718 to wild-type mice decreased DMPK mRNA levels by up to 90% in liver and skeletal muscle. Similarly, treatment of either human DMPK transgenic mice or cynomolgus monkeys with ISIS 486178 led to up to 70% inhibition of DMPK in multiple skeletal muscles and ∼50% in cardiac muscle in both species. Importantly, inhibition of DMPK was well tolerated and was not associated with any skeletal muscle or cardiac toxicity. Also interesting was the demonstration that the inhibition of DMPK mRNA levels in muscle was maintained for up to 16 and 13 weeks post-treatment in mice and monkeys, respectively. These results demonstrate that cEt-modified ASOs show potent activity in skeletal muscle, and that this attractive therapeutic approach warrants further clinical investigation to inhibit the gain-of-function toxic RNA underlying the pathogenesis of DM1.


Asunto(s)
Distrofia Miotónica/tratamiento farmacológico , Proteína Quinasa de Distrofia Miotónica/metabolismo , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos/farmacología , Animales , Línea Celular , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/enzimología , Proteína Quinasa de Distrofia Miotónica/antagonistas & inhibidores , Proteína Quinasa de Distrofia Miotónica/genética , Oligonucleótidos/química , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Ann Neurol ; 74(6): 862-72, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23929620

RESUMEN

OBJECTIVE: To develop RNA splicing biomarkers of disease severity and therapeutic response in myotonic dystrophy type 1 (DM1) and type 2 (DM2). METHODS: In a discovery cohort, we used microarrays to perform global analysis of alternative splicing in DM1 and DM2. The newly identified splicing changes were combined with previous data to create a panel of 50 putative splicing defects. In a validation cohort of 50 DM1 subjects, we measured the strength of ankle dorsiflexion (ADF) and then obtained a needle biopsy of tibialis anterior (TA) to analyze splice events in muscle RNA. The specificity of DM-associated splicing defects was assessed in disease controls. The CTG expansion size in muscle tissue was determined by Southern blot. The reversibility of splicing defects was assessed in transgenic mice by using antisense oligonucleotides to reduce levels of toxic RNA. RESULTS: Forty-two splicing defects were confirmed in TA muscle in the validation cohort. Among these, 20 events showed graded changes that correlated with ADF weakness. Five other splice events were strongly affected in DM1 subjects with normal ADF strength. Comparison to disease controls and mouse models indicated that splicing changes were DM-specific, mainly attributable to MBNL1 sequestration, and reversible in mice by targeted knockdown of toxic RNA. Splicing defects and weakness were not correlated with CTG expansion size in muscle tissue. INTERPRETATION: Alternative splicing changes in skeletal muscle may serve as biomarkers of disease severity and therapeutic response in myotonic dystrophy.


Asunto(s)
Empalme Alternativo , Distrofia Miotónica/genética , Adolescente , Adulto , Anciano , Animales , Biomarcadores , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Trastornos Miotónicos/genética , Trastornos Miotónicos/patología , Trastornos Miotónicos/fisiopatología , Distrofia Miotónica/patología , Distrofia Miotónica/fisiopatología , Oligonucleótidos Antisentido/genética , Proteínas de Unión al ARN/genética , Índice de Severidad de la Enfermedad , Adulto Joven
5.
Mol Ther ; 21(2): 380-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23183533

RESUMEN

Myotonic dystrophy type 1 (DM1) is an RNA dominant disease caused by expression of DM protein kinase (DMPK) transcripts that contain an expanded CUG repeat (CUG(exp)). The toxic mRNA localizes to nuclear foci and sequesters proteins involved in the regulation of alternative splicing, such as, muscleblind-like 1 (MBNL1). Here, we used synthetic short interfering RNAs (siRNAs) to target CUG repeats and test the concept that inhibiting the expression of CUG(exp) RNA can mitigate features of DM1 in transgenic mice. Intramuscular injection and electroporation of siRNA resulted in ~70-80% downregulation of CUG(exp) transcripts. A limited survey of endogenous mouse transcripts that contain nonexpanded CUG or CAG repeats showed that most were not affected, though Txlnb containing (CUG)(9) was significantly reduced. By this strategy, the number and intensity of CUG(exp) nuclear foci were reduced and splicing of MBNL1-dependent exons was improved. These data suggest that the expanded CUG repeats are a potential target for allele-selective RNA interference.


Asunto(s)
Distrofia Miotónica/genética , Distrofia Miotónica/terapia , Interferencia de ARN , Expansión de Repetición de Trinucleótido , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Electromiografía , Exones , Técnica del Anticuerpo Fluorescente , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Datos de Secuencia Molecular , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
Nat Commun ; 14(1): 1956, 2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029100

RESUMEN

In myotonic dystrophy type 1 (DM1), deregulated alternative splicing of the muscle chloride channel Clcn1 causes myotonia, a delayed relaxation of muscles due to repetitive action potentials. The degree of weakness in adult DM1 is associated with increased frequency of oxidative muscle fibers. However, the mechanism for glycolytic-to-oxidative fiber type transition in DM1 and its relationship to myotonia are uncertain. Here we cross two mouse models of DM1 to create a double homozygous model that features progressive functional impairment, severe myotonia, and near absence of type 2B glycolytic fibers. Intramuscular injection of an antisense oligonucleotide for targeted skipping of Clcn1 exon 7a corrects Clcn1 alternative splicing, increases glycolytic 2B levels to ≥ 40% frequency, reduces muscle injury, and improves fiber hypertrophy relative to treatment with a control oligo. Our results demonstrate that fiber type transitions in DM1 result from myotonia and are reversible, and support the development of Clcn1-targeting therapies for DM1.

7.
Neurobiol Dis ; 45(1): 539-46, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21964252

RESUMEN

The genetic basis of oculopharyngeal muscular dystrophy (OPMD) is a short expansion of a polyalanine tract (normal allele: 10 alanines, mutant allele: 11-17 alanines) in the nuclear polyadenylate binding protein PABPN1 which is essential for controlling poly(A) tail length in messenger RNA. Mutant PABPN1 forms nuclear inclusions in OPMD muscle. To investigate the pathogenic role of mutant PABPN1 in vivo, we generated a ligand-inducible transgenic mouse model by using the mifepristone-inducible gene expression system. Induction of ubiquitous expression of mutant PABPN1 resulted in skeletal and cardiac myopathy. Histological changes of degenerative myopathy were preceded by nuclear inclusions of insoluble PABPN1. Downregulation of mutant PABPN1 expression attenuated the myopathy and reduced the nuclear burden of insoluble PABPN1. These results support association between mutant PABPN1 accumulation and degenerative myopathy in mice. Resolution of myopathy in mice suggests that the disease process in OPMD patients may be treatable.


Asunto(s)
Núcleo Celular/patología , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/patología , Alelos , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/metabolismo , Poli A/genética , Poli A/metabolismo
8.
Proc Natl Acad Sci U S A ; 106(33): 13915-20, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19667189

RESUMEN

Myotonic dystrophy type 1 (DM1) is caused by toxicity of an expanded, noncoding (CUG)n tract in DM protein kinase (DMPK) transcripts. According to current evidence the long (CUG)n segment is involved in entrapment of muscleblind (Mbnl) proteins in ribonuclear aggregates and stabilized expression of CUG binding protein 1 (CUGBP1), causing aberrant premRNA splicing and associated pathogenesis in DM1 patients. Here, we report on the use of antisense oligonucleotides (AONs) in a therapeutic strategy for reversal of RNA-gain-of-function toxicity. Using a previously undescribed mouse DM1 myoblast-myotube cell model and DM1 patient cells as screening tools, we have identified a fully 2'-O-methyl-phosphorothioate-modified (CAG)7 AON that silences mutant DMPK RNA expression and reduces the number of ribonuclear aggregates in a selective and (CUG)n-length-dependent manner. Direct administration of this AON in muscle of DM1 mouse models in vivo caused a significant reduction in the level of toxic (CUG)n RNA and a normalizing effect on aberrant premRNA splicing. Our data demonstrate proof of principle for therapeutic use of simple sequence AONs in DM1 and potentially other unstable microsatellite diseases.


Asunto(s)
Distrofia Miotónica/genética , Oligonucleótidos/genética , ARN/genética , Alelos , Animales , Proteínas CELF1 , Silenciador del Gen , Ratones , Modelos Genéticos , Músculo Esquelético/metabolismo , Mutación , Mioblastos/metabolismo , Distrofia Miotónica/terapia , Oligonucleótidos/química , Oligonucleótidos Antisentido/genética , Interferencia de ARN , Empalme del ARN , Proteínas de Unión al ARN/genética
9.
Mol Ther Nucleic Acids ; 23: 393-405, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33473325

RESUMEN

Patients with myotonic dystrophy type 1 (DM1) identify chronic fatigue as the most debilitating symptom, which manifests in part as prolonged recovery after exercise. Clinical features of DM1 result from pathogenic gain-of-function activity of transcripts containing an expanded microsatellite CUG repeat (CUGexp). In DM1 mice, therapies targeting the CUGexp transcripts correct the molecular phenotype, reverse myotonia, and improve muscle pathology. However, the effect of targeted molecular therapies on fatigue in DM1 is unknown. Here, we use two mouse models of DM1, age-matched wild-type controls, an exercise-activity assay, electrical impedance myography, and therapeutic antisense oligonucleotides (ASOs) to show that exaggerated exercise-induced fatigue progresses with age, is unrelated to muscle fiber size, and persists despite correction of the molecular phenotype for 3 months. In old DM1 mice, ASO treatment combined with an exercise training regimen consisting of treadmill walking 30 min per day 6 days per week for 3 months reverse all measures of fatigue. Exercise training without ASO therapy improves some measures of fatigue without correction of the molecular pathology. Our results highlight a key limitation of ASO monotherapy for this clinically important feature and support the development of moderate-intensity exercise as an adjuvant for targeted molecular therapies of DM1.

10.
J Clin Invest ; 117(12): 3952-7, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18008009

RESUMEN

In myotonic dystrophy (dystrophia myotonica [DM]), an increase in the excitability of skeletal muscle leads to repetitive action potentials, stiffness, and delayed relaxation. This constellation of features, collectively known as myotonia, is associated with abnormal alternative splicing of the muscle-specific chloride channel (ClC-1) and reduced conductance of chloride ions in the sarcolemma. However, the mechanistic basis of the chloride channelopathy and its relationship to the development of myotonia are uncertain. Here we show that a morpholino antisense oligonucleotide (AON) targeting the 3' splice site of ClC-1 exon 7a reversed the defect of ClC-1 alternative splicing in 2 mouse models of DM. By repressing the inclusion of this exon, the AON restored the full-length reading frame in ClC-1 mRNA, upregulated the level of ClC-1 mRNA, increased the expression of ClC-1 protein in the surface membrane, normalized muscle ClC-1 current density and deactivation kinetics, and eliminated myotonic discharges. These observations indicate that the myotonia and chloride channelopathy observed in DM both result from abnormal alternative splicing of ClC-1 and that antisense-induced exon skipping offers a powerful method for correcting alternative splicing defects in DM.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Canalopatías/tratamiento farmacológico , Canales de Cloruro/biosíntesis , Miotonía Congénita/tratamiento farmacológico , Distrofia Miotónica/tratamiento farmacológico , Oligodesoxirribonucleótidos Antisentido/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Empalme Alternativo/genética , Animales , Canalopatías/genética , Canalopatías/metabolismo , Canales de Cloruro/genética , Exones/genética , Ratones , Miotonía Congénita/genética , Miotonía Congénita/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Sitios de Empalme de ARN/genética , Sarcolema/genética , Sarcolema/metabolismo
11.
Ann Clin Transl Neurol ; 6(6): 1106-1112, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31211175

RESUMEN

In muscular dystrophies, identification of pathogenic pseudoexons involves sequencing of the target gene cDNA derived from muscle mRNA. Here we use a urine "liquid biopsy," droplet digital PCR, and sequencing of PCR products to identify a novel cryptic splice site in DMD intron 67 that causes dystrophinopathy. Pseudoexon inclusion is 35% in urine cells, 34% in urine extracellular RNA (exRNA), and 54% in muscle biopsy tissue, but absent in serum exRNA. Our results suggest that cryptic splice site use varies depending on the RNA source, and that urine RNA has the potential to substitute for muscle biopsies to identify DMD pseudoexons.


Asunto(s)
Biomarcadores/orina , Distrofina/genética , Distrofia Muscular de Duchenne/genética , ARN Mensajero/orina , Empalme Alternativo , Preescolar , Exones , Humanos , Mutación , Sitios de Empalme de ARN , ARN Mensajero/genética
12.
Nat Commun ; 9(1): 5227, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30531949

RESUMEN

During drug development, tissue samples serve as indicators of disease activity and pharmacodynamic responses. Reliable non-invasive measures of drug target engagement will facilitate identification of promising new treatments. Here we develop and validate a novel bi-transgenic mouse model of myotonic dystrophy type 1 (DM1) in which expression of either DsRed or GFP is determined by alternative splicing of an upstream minigene that is mis-regulated in DM1. Using a novel in vivo fluorescence spectroscopy system, we show that quantitation of the DsRed/GFP ratio provides an accurate estimation of splicing outcomes in muscle tissue of live mice that nearly doubles throughput over conventional fluorescence imaging techniques. Serial in vivo spectroscopy measurements in mice treated with a C16 fatty acid ligand conjugated antisense (LICA) oligonucleotide reveal a dose-dependent therapeutic response within seven days, confirm a several-week duration of action, and demonstrate a two-fold greater target engagement as compared to the unconjugated parent oligonucleotide.


Asunto(s)
Empalme Alternativo , Músculos/efectos de los fármacos , Distrofia Miotónica/tratamiento farmacológico , Oligonucleótidos Antisentido/farmacología , Animales , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Transgénicos , Microscopía Fluorescente , Músculos/metabolismo , Músculos/patología , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Evaluación de Resultado en la Atención de Salud/métodos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Espectrometría de Fluorescencia
13.
Nat Commun ; 9(1): 3906, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254196

RESUMEN

Urine contains extracellular RNA (exRNA) markers of urogenital cancers. However, the capacity of genetic material in urine to identify systemic diseases is unknown. Here we describe exRNA splice products in human urine as a source of biomarkers for the two most common forms of muscular dystrophies, myotonic dystrophy (DM) and Duchenne muscular dystrophy (DMD). Using a training set, RT-PCR, droplet digital PCR, and principal component regression, we identify ten transcripts that are spliced differently in urine exRNA from patients with DM type 1 (DM1) as compared to unaffected or disease controls, form a composite biomarker, and develop a predictive model that is 100% accurate in our independent validation set. Urine also contains mutation-specific DMD mRNAs that confirm exon-skipping activity of the antisense oligonucleotide drug eteplirsen. Our results establish that urine mRNA splice variants can be used to monitor systemic diseases with minimal or no clinical effect on the urinary tract.


Asunto(s)
Empalme Alternativo , Biomarcadores/orina , Distrofias Musculares/orina , Isoformas de ARN/orina , ARN Mensajero/orina , Animales , Expresión Génica , Humanos , Ratones Noqueados , Ratones Transgénicos , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/orina , Mutación , Distrofia Miotónica/diagnóstico , Distrofia Miotónica/genética , Distrofia Miotónica/orina , Pronóstico , Isoformas de ARN/genética , Isoformas de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sensibilidad y Especificidad
14.
Mol Ther Nucleic Acids ; 7: 465-474, 2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28624222

RESUMEN

Myotonic dystrophy type 1 (DM1), a dominant hereditary muscular dystrophy, is caused by an abnormal expansion of a (CTG)n trinucleotide repeat in the 3' UTR of the human dystrophia myotonica protein kinase (DMPK) gene. As a consequence, mutant transcripts containing expanded CUG repeats are retained in nuclear foci and alter the function of splicing regulatory factors members of the MBNL and CELF families, resulting in alternative splicing misregulation of specific transcripts in affected DM1 tissues. In the present study, we treated DMSXL mice systemically with a 2'-4'-constrained, ethyl-modified (ISIS 486178) antisense oligonucleotide (ASO) targeted to the 3' UTR of the DMPK gene, which led to a 70% reduction in CUGexp RNA abundance and foci in different skeletal muscles and a 30% reduction in the heart. Furthermore, treatment with ISIS 486178 ASO improved body weight, muscle strength, and muscle histology, whereas no overt toxicity was detected. This is evidence that the reduction of CUGexp RNA improves muscle strength in DM1, suggesting that muscle weakness in DM1 patients may be improved following elimination of toxic RNAs.

15.
Science ; 325(5938): 336-9, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19608921

RESUMEN

Genomic expansions of simple tandem repeats can give rise to toxic RNAs that contain expanded repeats. In myotonic dystrophy, the expression of expanded CUG repeats (CUGexp) causes abnormal regulation of alternative splicing and neuromuscular dysfunction. We used a transgenic mouse model to show that derangements of myotonic dystrophy are reversed by a morpholino antisense oligonucleotide, CAG25, that binds to CUGexp RNA and blocks its interaction with muscleblind-like 1 (MBNL1), a CUGexp-binding protein. CAG25 disperses nuclear foci of CUGexp RNA and reduces the overall burden of this toxic RNA. As MBNL1 is released from sequestration, the defect of alternative splicing regulation is corrected, thereby restoring ion channel function. These findings suggest an alternative use of antisense methods, to inhibit deleterious interactions of proteins with pathogenic RNAs.


Asunto(s)
Regiones no Traducidas 3'/metabolismo , Proteínas de Unión al ADN/metabolismo , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido , Regiones no Traducidas 3'/genética , Actinas/genética , Empalme Alternativo , Animales , Línea Celular , Núcleo Celular/metabolismo , Canales de Cloruro/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Distrofia Miotónica/metabolismo , Proteína Quinasa de Distrofia Miotónica , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Transcripción Genética
16.
Neurotherapeutics ; 5(4): 592-600, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19019311

RESUMEN

Myotonic dystrophy (DM) is a dominantly inherited neurodegenerative disorder for which there is no cure or effective treatment. Investigation of DM pathogenesis has identified a novel disease mechanism that requires development of innovative therapeutic strategies. It is now clear that DM is not caused by expression of a mutant protein. Instead, DM is the first recognized example of an RNA-mediated disease. Expression of the mutated gene gives rise to an expanded repeat RNA that is directly toxic to cells. The mutant RNA is retained in the nucleus, forming ribonuclear inclusions in affected tissue. A primary consequence of RNA toxicity in DM is dysfunction of two classes of RNA binding proteins, which leads to abnormal regulation of alternative splicing, or spliceopathy, of select genes. Spliceopathy now is known to cause myotonia and insulin resistance in DM. As our understanding of pathogenesis continues to improve, therapy targeted directly at the RNA disease mechanism will begin to replace the supportive care currently available. New pharmacologic approaches to treat myotonia and muscle wasting in DM type 1 are already in early clinical trials, and therapies designed to reverse the RNA toxicity have shown promise in preclinical models by correcting spliceopathy and eliminating myotonia. The well-defined ribonuclear inclusions may serve as convenient therapeutic targets to identify new agents that modify RNA toxicity. Continued development of appropriate model systems will allow testing of additional therapeutic strategies as they become available. Although DM is a decidedly complex disorder, its RNA-mediated disease mechanism may prove to be highly susceptible to therapy.


Asunto(s)
Distrofia Miotónica/terapia , Animales , Terapia Genética , Humanos , Músculo Esquelético/patología , Distrofia Miotónica/genética , Distrofia Miotónica/patología , ARN/antagonistas & inhibidores , ARN/genética
17.
Curr Opin Neurol ; 20(5): 572-6, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17885447

RESUMEN

PURPOSE OF REVIEW: The aim of this review is to highlight recent progress in elucidating the disease mechanism in myotonic dystrophy type 1 and type 2. RECENT FINDINGS: Research on myotonic dystrophy has led to the recognition of a novel RNA-mediated disease process. In myotonic dystrophy it is the RNA rather than protein product of a disease gene that has deleterious effects on muscle cells. These unusual RNAs, which contain a long expanse of CUG or CCUG repeats, have far reaching effects on cell function by influencing the biogenesis of other cellular RNAs. One aspect of RNA metabolism that is particularly affected is the regulation of alternative splicing. By this mechanism, effects of myotonic dystrophy repeat expansions impact many different pathways, triggering a complex set of signs and symptoms. SUMMARY: The genetic lesion in myotonic dystrophy does not eliminate an essential muscle protein. Instead, it induces a defect of RNA processing that is potentially reversible. The nature of this disease process raises the possibility that myotonic dystrophy, among genetic disorders, may be unusually susceptible to treatment using non-gene-therapy approaches.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Músculo Esquelético/fisiopatología , Distrofia Miotónica/genética , Distrofia Miotónica/fisiopatología , ARN/genética , Empalme Alternativo/genética , Animales , Proteínas CELF1 , Humanos , Músculo Esquelético/metabolismo , Mutación/genética , Distrofia Miotónica/metabolismo , Estabilidad del ARN/genética , Proteínas de Unión al ARN/genética
18.
Proc Natl Acad Sci U S A ; 103(2): 419-24, 2006 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-16387861

RESUMEN

Plasmid-mediated gene therapy can restore dystrophin expression in skeletal muscle in the mdx mouse, a model of Duchenne muscular dystrophy. However, sufficient long-term expression and distribution of dystrophin remain a hurdle for translating this technology into a viable treatment for Duchenne muscular dystrophy. To improve plasmid-mediated gene therapy for muscle diseases, we studied the effects of targeted plasmid integration using a phage integrase (phiC31) that can mediate the integration of suitably modified plasmids into the mammalian genome. Using a luciferase expression plasmid, we monitored plasmid gene expression noninvasively in living mice by bioluminescence imaging. Coinjection of an integrase plasmid resulted in 5- to 10-fold higher levels of sustained luciferase expression. Likewise, plasmid-mediated dystrophin expression in mdx muscle was enhanced by integration. Using a combination of dystrophin and luciferase plasmids, we analyzed the functional benefit of dystrophin expression in the dystrophic muscle. The expression of dystrophin slowed the loss of luciferase expression associated with muscle degeneration, and that protection was enhanced by targeted integration of the dystrophin plasmid. In the presence of integrase, dystrophin expression was distributed along a much greater length of individual fibers, and this was associated with increased protection against degenerative changes. These data demonstrate the importance of both the level and distribution of dystrophin expression to achieve therapeutic efficacy, and that the efficacy can be enhanced by targeted plasmid integration.


Asunto(s)
Marcación de Gen , Terapia Genética/métodos , Distrofias Musculares/genética , Distrofias Musculares/terapia , Plásmidos/genética , Plásmidos/metabolismo , Recombinación Genética/genética , Animales , ADN/genética , Distrofina/biosíntesis , Distrofina/genética , Distrofina/metabolismo , Regulación de la Expresión Génica , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Distrofias Musculares/metabolismo , Factores de Tiempo
19.
Proc Natl Acad Sci U S A ; 103(31): 11748-53, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16864772

RESUMEN

RNA-mediated pathogenesis is a recently developed disease model that proposes that certain types of mutant genes produce toxic transcripts that inhibit the activities of specific proteins. This pathogenesis model was proposed first for the neuromuscular disease myotonic dystrophy (DM), which is associated with the expansion of structurally related (CTG)(n) and (CCTG)(n) microsatellites in two unrelated genes. At the RNA level, these expansions form stable hairpins that alter the pre-mRNA splicing activities of two antagonistic factor families, the MBNL and CELF proteins. It is unclear which altered activity is primarily responsible for disease pathogenesis and whether other factors and biochemical pathways are involved. Here, we show that overexpression of Mbnl1 in vivo mediated by transduction of skeletal muscle with a recombinant adeno-associated viral vector rescues disease-associated muscle hyperexcitability, or myotonia, in the HSA(LR) poly(CUG) mouse model for DM. Myotonia reversal occurs concurrently with restoration of the normal adult-splicing patterns of four pre-mRNAs that are misspliced during postnatal development in DM muscle. Our results support the hypothesis that the loss of MBNL1 activity is a primary pathogenic event in the development of RNA missplicing and myotonia in DM and provide a rationale for therapeutic strategies designed either to overexpress MBNL1 or inhibit MBNL1 interactions with CUG and CCUG repeat expansions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Miotonía , Distrofia Miotónica , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido , Empalme Alternativo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Electromiografía , Exones , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Miotonía/genética , Miotonía/terapia , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores del ARN/metabolismo , Proteínas de Unión al ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA