Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 49(4): 2240-2254, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33503262

RESUMEN

Biomarker-driven trials hold promise for therapeutic development in chronic diseases, such as muscular dystrophy. Myotonic dystrophy type 1 (DM1) involves RNA toxicity, where transcripts containing expanded CUG-repeats (CUGexp) accumulate in nuclear foci and sequester splicing factors in the Muscleblind-like (Mbnl) family. Oligonucleotide therapies to mitigate RNA toxicity have emerged but reliable measures of target engagement are needed. Here we examined muscle transcriptomes in mouse models of DM1 and found that CUGexp expression or Mbnl gene deletion cause similar dysregulation of alternative splicing. We selected 35 dysregulated exons for further study by targeted RNA sequencing. Across a spectrum of mouse models, the individual splice events and a composite index derived from all events showed a graded response to decrements of Mbnl or increments of CUGexp. Antisense oligonucleotides caused prompt reduction of CUGexp RNA and parallel correction of the splicing index, followed by subsequent elimination of myotonia. These results suggest that targeted splice sequencing may provide a sensitive and reliable way to assess therapeutic impact in DM1.


Asunto(s)
Empalme Alternativo , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , Análisis de Secuencia de ARN , Animales , Proteínas de Unión al ADN/genética , Exones , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones , Músculos/metabolismo , Músculos/fisiología , Distrofia Miotónica/metabolismo , Oligonucleótidos Antisentido , Proteínas de Unión al ARN/genética , Regeneración , Transcriptoma , Expansión de Repetición de Trinucleótido
2.
Muscle Nerve ; 66(3): 336-339, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35426155

RESUMEN

INTRODUCTION/AIMS: Remote study visits (RSVs) are emerging as important tools for clinical research. We tested the feasibility of using RSVs to evaluate patients with myotonic dystrophy type 1 (DM1), including remote quantitative assessment of muscle function, and we assessed correlations of remote assessments with patient-reported function. METHODS: Twenty three subjects with DM1 were consented remotely. Toolkits containing a tablet computer, grip dynamometer, and spirometer were shipped to participants. The tablets were loaded with software for video-conferencing and questionnaires about functional impairment, patient experience with technology, and willingness to participate in future remote studies. Grip strength, forced vital capacity, peak cough flow, timed-up-and-go (TUG), and grip myotonia (hand opening time) were determined during RSVs. We assessed correlations of remote assessments with patient-reported outcomes of muscle function and with CTG repeat size. RESULTS: All 23 subjects completed RSVs. 95% of participants were able to complete all components of the remote study. All toolkit components were returned upon completion. Grip strength and TUG demonstrated moderate to strong correlations with self-reported inventories of upper and lower extremity impairment, respectively (ρ = 0.7 and ρ = -0.52). A total of 91% of subjects expressed interest in participating in future RSVs. DISCUSSION: Results of this study support the feasibility of using portable devices and video-conferencing for remote collection of patient-reported outcomes and quantitative assessment of muscle function in DM1.


Asunto(s)
Miotonía , Distrofia Miotónica , Estudios de Factibilidad , Fuerza de la Mano , Humanos , Músculo Esquelético , Distrofia Miotónica/diagnóstico
3.
Muscle Nerve ; 66(4): 508-512, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35778789

RESUMEN

INTRODUCTION/AIMS: Disease progression in myotonic dystrophy (DM) is marked by milestone events when functional thresholds are crossed. DM type 2 (DM2) is considered less severe than DM type 1 (DM1), but it is unknown whether this applies uniformly to all features. We compared the age-dependent risk for milestone events in DM1 and DM2 and tested for associations with age of onset and sex. METHODS: We studied a large cohort of adult participants in a national registry of DM1 and DM2. Using annual surveys from participants, we ascertained milestone events for motor involvement (use of cane, walker, ankle brace, wheelchair, or ventilatory device), systemic involvement (diabetes, pacemaker, cancer), loss of employment due to DM, and death. RESULTS: Mean follow-up of registry participants (929 DM1 and 222 DM2 patients) was 7 years. Disability and motor milestones occurred at earlier ages in DM1 than in DM2. In contrast, the risk of diabetes was higher and tended to occur earlier in DM2 (hazard ratio [HR], 0.56; P ≤ .001). In DM1, the milestone events tended to occur earlier, and life expectancy was reduced, when symptoms began at younger ages. In DM1, men were at greater risk for disability (HR, 1.34; P ≤ .01), use of ankle braces (HR, 1.41; P = .02), and diabetes (HR, 2.2; P ≤ .0001), whereas women were at greater risk for needing walkers (HR, 0.68; P = .001) or malignancy (HR, 0.66; P ≤ .01). DISCUSSION: Milestone events recorded through registries can be used to assess long-term impact of DM in large cohorts. Except for diabetes, the age-related risk of milestone events is greater in DM1 than in DM2.


Asunto(s)
Diabetes Mellitus Tipo 2 , Distrofia Miotónica , Adulto , Estudios de Cohortes , Femenino , Humanos , Masculino , Distrofia Miotónica/diagnóstico , Sistema de Registros
4.
Muscle Nerve ; 65(5): 560-567, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35179228

RESUMEN

INTRODUCTION/AIMS: Myotonic dystrophy type 1 (DM1) is known to affect cognitive function, but the best methods to assess central nervous system involvement in multicenter studies have not been determined. In this study our primary aim was to evaluate the potential of computerized cognitive tests to assess cognition in DM1. METHODS: We conducted a prospective, longitudinal, observational study of 113 adults with DM1 at six sites. Psychomotor speed, attention, working memory, and executive functioning were assessed at baseline, 3 months, and 12 months using computerized cognitive tests. Results were compared with assessments of muscle function and patient reported outcomes (PROs), including the Myotonic Dystrophy Health Index (MDHI) and the 5-dimension EuroQol (EQ-5D-5L) questionnaire. RESULTS: Based on intraclass correlation coefficients, computerized cognitive tests had moderate to good reliability for psychomotor speed (0.76), attention (0.82), working memory speed (0.79), working memory accuracy (0.65), and executive functioning (0.87). Performance at baseline was lowest for working memory accuracy (P < .0001). Executive function performance improved from baseline to 3 months (P < .0001), without further changes over 1 year. There was a moderate correlation between poorer executive function and larger CTG repeat size (r = -0.433). There were some weak associations between PROs and cognitive performance. DISCUSSION: Computerized tests of cognition are feasible in multicenter studies of DM1. Poor performance was exhibited in working memory, which may be a useful variable in clinical trials. Learning effects may have contributed to the improvement in executive functioning. The relationship between PROs and cognitive impairment in DM1 requires further study.


Asunto(s)
Distrofia Miotónica , Adulto , Cognición , Computadores , Humanos , Estudios Longitudinales , Distrofia Miotónica/complicaciones , Distrofia Miotónica/diagnóstico , Estudios Prospectivos , Reproducibilidad de los Resultados
5.
Mol Cell ; 56(2): 311-322, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25263597

RESUMEN

Inhibition of muscleblind-like (MBNL) activity due to sequestration by microsatellite expansion RNAs is a major pathogenic event in the RNA-mediated disease myotonic dystrophy (DM). Although MBNL1 and MBNL2 bind to nascent transcripts to regulate alternative splicing during muscle and brain development, another major binding site for the MBNL protein family is the 3' untranslated region of target RNAs. Here, we report that depletion of Mbnl proteins in mouse embryo fibroblasts leads to misregulation of thousands of alternative polyadenylation events. HITS-CLIP and minigene reporter analyses indicate that these polyadenylation switches are a direct consequence of MBNL binding to target RNAs. Misregulated alternative polyadenylation also occurs in skeletal muscle in a mouse polyCUG model and human DM, resulting in the persistence of neonatal polyadenylation patterns. These findings reveal an additional developmental function for MBNL proteins and demonstrate that DM is characterized by misregulation of pre-mRNA processing at multiple levels.


Asunto(s)
Empalme Alternativo/genética , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Poliadenilación/genética , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3'/genética , Animales , Sitios de Unión/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Repeticiones de Microsatélite/genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Distrofia Miotónica/genética , Unión Proteica , Interferencia de ARN , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteínas de Unión al ARN/metabolismo
6.
Proc Natl Acad Sci U S A ; 115(16): 4234-4239, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29610297

RESUMEN

Expansions of simple sequence repeats, or microsatellites, have been linked to ∼30 neurological-neuromuscular diseases. While these expansions occur in coding and noncoding regions, microsatellite sequence and repeat length diversity is more prominent in introns with eight different trinucleotide to hexanucleotide repeats, causing hereditary diseases such as myotonic dystrophy type 2 (DM2), Fuchs endothelial corneal dystrophy (FECD), and C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Here, we test the hypothesis that these GC-rich intronic microsatellite expansions selectively trigger host intron retention (IR). Using DM2, FECD, and C9-ALS/FTD as examples, we demonstrate that retention is readily detectable in affected tissues and peripheral blood lymphocytes and conclude that IR screening constitutes a rapid and inexpensive biomarker for intronic repeat expansion disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Distrofia Endotelial de Fuchs/genética , Intrones/genética , Distrofia Miotónica/genética , Composición de Base , Biomarcadores , Humanos , Linfocitos/química , Músculo Esquelético/química , Miocardio/química , Especificidad de Órganos , Polimorfismo de Nucleótido Simple , Empalme del ARN , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Análisis de Matrices Tisulares
7.
Nucleic Acids Res ; 46(1): e1, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29036654

RESUMEN

Cellular accumulation of repetitive RNA occurs in several dominantly-inherited genetic disorders. Expanded CUG, CCUG or GGGGCC repeats are expressed in myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), or familial amyotrophic lateral sclerosis, respectively. Expanded repeat RNAs (ER-RNAs) exert a toxic gain-of-function and are prime therapeutic targets in these diseases. However, efforts to quantify ER-RNA levels or monitor knockdown are confounded by stable structure and heterogeneity of the ER-RNA tract and background signal from non-expanded repeats. Here, we used a thermostable group II intron reverse transcriptase (TGIRT-III) to convert ER-RNA to cDNA, followed by quantification on slot blots. We found that TGIRT-III was capable of reverse transcription (RTn) on enzymatically synthesized ER-RNAs. By using conditions that limit cDNA synthesis from off-target sequences, we observed hybridization signals on cDNA slot blots from DM1 and DM2 muscle samples but not from healthy controls. In transgenic mouse models of DM1 the cDNA slot blots accurately reflected the differences of ER-RNA expression across different transgenic lines, and showed therapeutic reductions in skeletal and cardiac muscle, accompanied by improvements of the DM1-associated splicing defects. TGIRT-III was also active on CCCCGG- and GGGGCC-repeats, suggesting that ER-RNA analysis is feasible for several repeat expansion disorders.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Intrones/genética , Distrofia Miotónica/genética , ADN Polimerasa Dirigida por ARN/genética , ARN/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Secuencia de Bases , Electroforesis Capilar , Electroforesis en Gel de Poliacrilamida , Estabilidad de Enzimas , Humanos , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Miotónica/metabolismo , Empalme del ARN , ADN Polimerasa Dirigida por ARN/metabolismo , Temperatura
8.
PLoS Genet ; 12(9): e1006316, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27681373

RESUMEN

Alternative splicing is a regulated process that results in expression of specific mRNA and protein isoforms. Alternative splicing factors determine the relative abundance of each isoform. Here we focus on MBNL1, a splicing factor misregulated in the disease myotonic dystrophy. By altering the concentration of MBNL1 in cells across a broad dynamic range, we show that different splicing events require different amounts of MBNL1 for half-maximal response, and respond more or less steeply to MBNL1. Motifs around MBNL1 exon 5 were studied to assess how cis-elements mediate the MBNL1 dose-dependent splicing response. A framework was developed to estimate MBNL concentration using splicing responses alone, validated in the cell-based model, and applied to myotonic dystrophy patient muscle. Using this framework, we evaluated the ability of individual and combinations of splicing events to predict functional MBNL concentration in human biopsies, as well as their performance as biomarkers to assay mild, moderate, and severe cases of DM.

9.
Biochemistry ; 57(14): 2094-2108, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29562132

RESUMEN

We report the development of a new class of nucleic acid ligands that is comprised of Janus bases and the MPγPNA backbone and is capable of binding rCAG repeats in a sequence-specific and selective manner via, inference, bivalent H-bonding interactions. Individually, the interactions between ligands and RNA are weak and transient. However, upon the installation of a C-terminal thioester and an N-terminal cystine and the reduction of disulfide bond, they undergo template-directed native chemical ligation to form concatenated oligomeric products that bind tightly to the RNA template. In the absence of an RNA target, they self-deactivate by undergoing an intramolecular reaction to form cyclic products, rendering them inactive for further binding. The work has implications for the design of ultrashort nucleic acid ligands for targeting rCAG-repeat expansion associated with Huntington's disease and a number of other related neuromuscular and neurodegenerative disorders.


Asunto(s)
Enfermedad de Huntington , ARN/química , Expansión de Repetición de Trinucleótido , Humanos , Ligandos , ARN/genética
10.
Hum Mol Genet ; 25(19): 4328-4338, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27522499

RESUMEN

Myotonic dystrophy type 1 (DM1) is a genetic disorder in which dominant-active DM protein kinase (DMPK) transcripts accumulate in nuclear foci, leading to abnormal regulation of RNA processing. A leading approach to treat DM1 uses DMPK-targeting antisense oligonucleotides (ASOs) to reduce levels of toxic RNA. However, basal levels of DMPK protein are reduced by half in DM1 patients. This raises concern that intolerance for further DMPK loss may limit ASO therapy, especially since mice with Dmpk gene deletion reportedly show cardiac defects and skeletal myopathy. We re-examined cardiac and muscle function in mice with Dmpk gene deletion, and studied post-maturity knockdown using Dmpk-targeting ASOs in mice with heterozygous deletion. Contrary to previous reports, we found no effect of Dmpk gene deletion on cardiac or muscle function, when studied on two genetic backgrounds. In heterozygous knockouts, the administration of ASOs reduced Dmpk expression in cardiac and skeletal muscle by > 90%, yet survival, electrocardiogram intervals, cardiac ejection fraction and muscle strength remained normal. The imposition of cardiac stress by pressure overload, or muscle stress by myotonia, did not unmask a requirement for DMPK. Our results support the feasibility and safety of using ASOs for post-transcriptional silencing of DMPK in muscle and heart.


Asunto(s)
Terapia Genética , Distrofia Miotónica/genética , Distrofia Miotónica/terapia , Proteína Quinasa de Distrofia Miotónica/biosíntesis , Oligonucleótidos Antisentido/administración & dosificación , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miocardio/metabolismo , Miocardio/patología , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica/genética , Oligonucleótidos Antisentido/genética , ARN/antagonistas & inhibidores , ARN/genética
11.
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
12.
Hum Mol Genet ; 24(7): 2035-48, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25504044

RESUMEN

Myotonic dystrophy type 1 (DM1), the most prevalent muscular dystrophy in adults, is characterized by progressive muscle wasting and multi-systemic complications. DM1 is the prototype for disorders caused by RNA toxicity. Currently, no therapies exist. Here, we identify that fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor receptor super-family, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and in tissues from DM1 patients, and that its expression correlates with severity of muscle pathology. This is associated with downstream signaling through the NF-κB pathways. In mice with RNA toxicity, genetic deletion of Fn14 results in reduced muscle pathology and better function. Importantly, blocking TWEAK/Fn14 signaling with an anti-TWEAK antibody likewise improves muscle histopathology and functional outcomes in affected mice. These results reveal new avenues for therapeutic development and provide proof of concept for a novel therapeutic target for which clinically available therapy exists to potentially treat muscular dystrophy in DM1.


Asunto(s)
Distrofia Miotónica/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Adulto , Animales , Anticuerpos/administración & dosificación , Citocina TWEAK , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/efectos de los fármacos , Receptor de TWEAK , Inhibidores del Factor de Necrosis Tumoral , Factores de Necrosis Tumoral/genética
13.
Nucleic Acids Res ; 43(6): 3318-31, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25753670

RESUMEN

Myotonic dystrophy type 1 (DM1) is an autosomal dominant multisystemic disorder caused by expansion of CTG triplet repeats in 3'-untranslated region of DMPK gene. The pathomechanism of DM1 is driven by accumulation of toxic transcripts containing expanded CUG repeats (CUG(exp)) in nuclear foci which sequester several factors regulating RNA metabolism, such as Muscleblind-like proteins (MBNLs). In this work, we utilized very short chemically modified antisense oligonucleotides composed exclusively of locked nucleic acids (all-LNAs) complementary to CUG repeats, as potential therapeutic agents against DM1. Our in vitro data demonstrated that very short, 8- or 10-unit all-LNAs effectively bound the CUG repeat RNA and prevented the formation of CUG(exp)/MBNL complexes. In proliferating DM1 cells as well as in skeletal muscles of DM1 mouse model the all-LNAs induced the reduction of the number and size of CUG(exp) foci and corrected MBNL-sensitive alternative splicing defects with high efficacy and specificity. The all-LNAs had low impact on the cellular level of CUG(exp)-containing transcripts and did not affect the expression of other transcripts with short CUG repeats. Our data strongly indicate that short all-LNAs complementary to CUG repeats are a promising therapeutic tool against DM1.


Asunto(s)
Empalme Alternativo , Distrofia Miotónica/terapia , Oligonucleótidos Antisentido/uso terapéutico , Oligonucleótidos/uso terapéutico , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Línea Celular , Modelos Animales de Enfermedad , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Proteínas Mutantes/genética , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Proteína Quinasa de Distrofia Miotónica/genética , Oligonucleótidos/genética , Oligonucleótidos Antisentido/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Expansión de Repetición de Trinucleótido
14.
Nucleic Acids Res ; 42(10): 6591-602, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24799433

RESUMEN

Myotonic dystrophy type 1 (DM1) is a dominantly inherited neuromuscular disorder resulting from expression of RNA containing an expanded CUG repeat (CUG(exp)). The pathogenic RNA is retained in nuclear foci. Poly-(CUG) binding proteins in the Muscleblind-like (MBNL) family are sequestered in foci, causing misregulated alternative splicing of specific pre-mRNAs. Inhibitors of MBNL1-CUG(exp) binding have been shown to restore splicing regulation and correct phenotypes in DM1 models. We therefore conducted a high-throughput screen to identify novel inhibitors of MBNL1-(CUG)12 binding. The most active compound was lomofungin, a natural antimicrobial agent. We found that lomofungin undergoes spontaneous dimerization in DMSO, producing dilomofungin, whose inhibition of MBNL1-(CUG)12 binding was 17-fold more potent than lomofungin itself. However, while dilomofungin displayed the desired binding characteristics in vitro, when applied to cells it produced a large increase of CUG(exp) RNA in nuclear foci, owing to reduced turnover of the CUG(exp) transcript. By comparison, the monomer did not induce CUG(exp) accumulation in cells and was more effective at rescuing a CUG(exp)-induced splicing defect. These results support the feasibility of high-throughput screens to identify compounds targeting toxic RNA, but also demonstrate that ligands for repetitive sequences may have unexpected effects on RNA decay.


Asunto(s)
Fenazinas/farmacología , Proteínas de Unión al ARN/metabolismo , ARN/química , ARN/metabolismo , Regiones no Traducidas 3' , Empalme Alternativo/efectos de los fármacos , Dimerización , Humanos , Fenazinas/química , Fenazinas/metabolismo , Estabilidad del ARN/efectos de los fármacos , Secuencias Repetitivas de Ácidos Nucleicos
15.
PLoS Genet ; 9(12): e1003866, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367268

RESUMEN

Slipped-strand DNAs, formed by out-of-register mispairing of repeat units on complementary strands, were proposed over 55 years ago as transient intermediates in repeat length mutations, hypothesized to cause at least 40 neurodegenerative diseases. While slipped-DNAs have been characterized in vitro, evidence of slipped-DNAs at an endogenous locus in biologically relevant tissues, where instability varies widely, is lacking. Here, using an anti-DNA junction antibody and immunoprecipitation, we identify slipped-DNAs at the unstable trinucleotide repeats (CTG)n•(CAG)n of the myotonic dystrophy disease locus in patient brain, heart, muscle and other tissues, where the largest expansions arise in non-mitotic tissues such as cortex and heart, and are smallest in the cerebellum. Slipped-DNAs are shown to be present on the expanded allele and in chromatinized DNA. Slipped-DNAs are present as clusters of slip-outs along a DNA, with each slip-out having 1-100 extrahelical repeats. The allelic levels of slipped-DNA containing molecules were significantly greater in the heart over the cerebellum (relative to genomic equivalents of pre-IP input DNA) of a DM1 individual; an enrichment consistent with increased allelic levels of slipped-DNA structures in tissues having greater levels of CTG instability. Surprisingly, this supports the formation of slipped-DNAs as persistent mutation products of repeat instability, and not merely as transient mutagenic intermediates. These findings further our understanding of the processes of mutation and genetic variation.


Asunto(s)
Anticuerpos Antinucleares/genética , ADN/genética , Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido/genética , Cromatina/genética , ADN/química , Humanos , Mutación , Distrofia Miotónica/patología , Conformación de Ácido Nucleico , Distribución Tisular
16.
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
17.
Hum Mol Genet ; 21(6): 1312-24, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22140091

RESUMEN

Myotonic dystrophy type 1 and type 2 (DM1 and DM2) are genetic diseases in which mutant transcripts containing expanded CUG or CCUG repeats cause cellular dysfunction by altering the processing or metabolism of specific mRNAs and miRNAs. The toxic effects of mutant RNA are mediated partly through effects on proteins that regulate alternative splicing. Here we show that alternative splicing of exon 29 (E29) of Ca(V)1.1, a calcium channel that controls skeletal muscle excitation-contraction coupling, is markedly repressed in DM1 and DM2. The extent of E29 skipping correlated with severity of weakness in tibialis anterior muscle of DM1 patients. Two splicing factors previously implicated in DM1, MBNL1 and CUGBP1, participated in the regulation of E29 splicing. In muscle fibers of wild-type mice, the Ca(V)1.1 channel conductance and voltage sensitivity were increased by splice-shifting oligonucleotides that induce E29 skipping. In contrast to human DM1, expression of CUG-expanded RNA caused only a modest increase in E29 skipping in mice. However, forced skipping of E29 in these mice, to levels approaching those observed in human DM1, aggravated the muscle pathology as evidenced by increased central nucleation. Together, these results indicate that DM-associated splicing defects alter Ca(V)1.1 function, with potential for exacerbation of myopathy.


Asunto(s)
Empalme Alternativo , Canales de Calcio Tipo L/fisiología , Calcio/metabolismo , Activación del Canal Iónico/fisiología , Debilidad Muscular/etiología , Trastornos Miotónicos/fisiopatología , Distrofia Miotónica/fisiopatología , Animales , Proteínas CELF1 , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Exones/genética , Regulación de la Expresión Génica , Humanos , Immunoblotting , Ratones , Ratones Transgénicos , Morfolinos/farmacología , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Técnicas de Placa-Clamp , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
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
19.
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
20.
Nucleic Acids Res ; 40(13): 6380-90, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492623

RESUMEN

The myotonic dystrophies (DM) are human diseases in which the accumulation of toxic RNA (CUG or CCUG) repeats in the cell causes sequestration of splicing factors, including MBNL1, leading to clinical symptoms such as muscle wasting and myotonia. We previously used Dynamic Combinatorial Chemistry to identify the first compounds known to inhibit (CUG)-MBNL1 binding in vitro. We now report transformation of those compounds into structures with activity in vivo. Introduction of a benzo[g]quinoline substructure previously unknown in the context of RNA recognition, as well as other modifications, provided several molecules with enhanced binding properties, including compounds with strong selectivity for CUG repeats over CAG repeats or CAG-CUG duplex RNA. Compounds readily penetrate cells, and improve luciferase activity in a mouse myoblast assay in which enzyme function is coupled to a release of nuclear CUG-RNA retention. Most importantly, two compounds are able to partially restore splicing in a mouse model of DM1.


Asunto(s)
Distrofia Miotónica/genética , Quinolinas/farmacología , ARN/química , Repeticiones de Trinucleótidos , Animales , Línea Celular , Técnicas Químicas Combinatorias , Cinética , Ratones , Quinolinas/química , Quinolinas/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA