Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38923193

RESUMEN

AIMS: To investigate how a change in body position with light-intensity physical activity (PA) 'snacks' (LIPAS, alternate sitting and standing, walking or standing continuously) compared with uninterrupted prolonged sitting affects glucose metabolism and heart rate variability (HRV) parameters in young adults with overweight and obesity. MATERIALS AND METHODS: We conducted a four-arm randomized controlled crossover trial. The following conditions were tested during an 8-h simulated workday: uninterrupted prolonged sitting (SIT), alternate sitting and standing (SIT-STAND; 2.5 h total), continuous standing (STAND), and continuous walking (1.0 mph; WALK). The primary outcome was to investigate how a change in body position (alternate sitting and standing, walking or standing continuously) compared with uninterrupted sitting affects mean 8-h glucose metabolism. Secondary outcomes included the effects on 2-h postprandial glucose concentrations, as well as on 8-h/24-h heart rate and HRV parameters, in the respective study arms. Capillary blood samples were drawn from an hyperemised earlobe in the fasted state and once every hour during each trial intervention by puncturing the earlobe with a lancet and collecting 20 µL of blood (Biosen S-Line Lab+; EKF diagnostics, Barleben, Germany). HRV was assessed for 24 h including the 8-h intervention phase, and a home phase by means of a Holter electrocardiogram. All participants received the same standardized non-relativised breakfast and lunch during the four trial visits. RESULTS: Seventeen individuals (eight women, mean age 23.4 ± 3.3 years, body mass index 29.7 ± 3.8 kg/m2, glycated haemoglobin level 34.8 ± 3.1 mmol/mol [5.4 ± 0.3%], body fat 31.8 ± 8.2%) completed all four trial arms. Compared with SIT (89.4 ± 6.8 mg/dL), 8-h mean glucose was lower in all other conditions (p < 0.05) and this was statistically significant compared with WALK (86.3 ± 5.2 mg/dL; p = 0.034). Two-hour postprandial glucose after breakfast was approximately 7% lower for WALK compared with SIT (p = 0.002). Furthermore, significant time × condition effects on HRV parameters favouring light-intensity walking were observed (p < 0.001). CONCLUSIONS: Replacement and interruption of prolonged sitting with light-intensity walking showed a significant blood glucose-lowering effect and improved HRV during an 8-h work environment in young adults with overweight and obesity.

2.
Ann Neurol ; 88(2): 332-347, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32403198

RESUMEN

OBJECTIVE: A hitherto undescribed phenotype of early onset muscular dystrophy associated with sensorineural hearing loss and primary ovarian insufficiency was initially identified in 2 siblings and in subsequent patients with a similar constellation of findings. The goal of this study was to understand the genetic and molecular etiology of this condition. METHODS: We applied whole exome sequencing (WES) superimposed on shared haplotype regions to identify the initial biallelic variants in GGPS1 followed by GGPS1 Sanger sequencing or WES in 5 additional families with the same phenotype. Molecular modeling, biochemical analysis, laser membrane injury assay, and the generation of a Y259C knock-in mouse were done. RESULTS: A total of 11 patients in 6 families carrying 5 different biallelic pathogenic variants in specific domains of GGPS1 were identified. GGPS1 encodes geranylgeranyl diphosphate synthase in the mevalonate/isoprenoid pathway, which catalyzes the synthesis of geranylgeranyl pyrophosphate, the lipid precursor of geranylgeranylated proteins including small guanosine triphosphatases. In addition to proximal weakness, all but one patient presented with congenital sensorineural hearing loss, and all postpubertal females had primary ovarian insufficiency. Muscle histology was dystrophic, with ultrastructural evidence of autophagic material and large mitochondria in the most severe cases. There was delayed membrane healing after laser injury in patient-derived myogenic cells, and a knock-in mouse of one of the mutations (Y259C) resulted in prenatal lethality. INTERPRETATION: The identification of specific GGPS1 mutations defines the cause of a unique form of muscular dystrophy with hearing loss and ovarian insufficiency and points to a novel pathway for this clinical constellation. ANN NEUROL 2020;88:332-347.


Asunto(s)
Dimetilaliltranstransferasa/genética , Farnesiltransferasa/genética , Geraniltranstransferasa/genética , Pérdida Auditiva/genética , Distrofias Musculares/genética , Mutación/genética , Insuficiencia Ovárica Primaria/genética , Adolescente , Adulto , Animales , Femenino , Técnicas de Sustitución del Gen/métodos , Pérdida Auditiva/diagnóstico por imagen , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofias Musculares/diagnóstico por imagen , Linaje , Insuficiencia Ovárica Primaria/diagnóstico por imagen , Estructura Secundaria de Proteína , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos , Adulto Joven
3.
Am J Hum Genet ; 100(3): 537-545, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28190459

RESUMEN

Congenital muscular dystrophies display a wide phenotypic and genetic heterogeneity. The combination of clinical, biochemical, and molecular genetic findings must be considered to obtain the precise diagnosis and provide appropriate genetic counselling. Here we report five individuals from four families presenting with variable clinical features including muscular dystrophy with a reduction in dystroglycan glycosylation, short stature, intellectual disability, and cataracts, overlapping both the dystroglycanopathies and Marinesco-Sjögren syndrome. Whole-exome sequencing revealed homozygous missense and compound heterozygous mutations in INPP5K in the affected members of each family. INPP5K encodes the inositol polyphosphate-5-phosphatase K, also known as SKIP (skeletal muscle and kidney enriched inositol phosphatase), which is highly expressed in the brain and muscle. INPP5K localizes to both the endoplasmic reticulum and to actin ruffles in the cytoplasm. It has been shown to regulate myoblast differentiation and has also been implicated in protein processing through its interaction with the ER chaperone HSPA5/BiP. We show that morpholino-mediated inpp5k loss of function in the zebrafish results in shortened body axis, microphthalmia with disorganized lens, microcephaly, reduced touch-evoked motility, and highly disorganized myofibers. Altogether these data demonstrate that mutations in INPP5K cause a congenital muscular dystrophy syndrome with short stature, cataracts, and intellectual disability.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Monoéster Fosfórico Hidrolasas/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Niño , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Femenino , Estudio de Asociación del Genoma Completo , Glicosilación , Trastornos del Crecimiento/genética , Humanos , Discapacidad Intelectual/genética , Masculino , Microcefalia/genética , Músculo Esquelético/metabolismo , Mutación , Linaje , Adulto Joven , Pez Cebra/genética
4.
Am J Hum Genet ; 98(1): 90-101, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26686765

RESUMEN

Clustered regularly interspaced short palindromic repeat (CRISPR) has arisen as a frontrunner for efficient genome engineering. However, the potentially broad therapeutic implications are largely unexplored. Here, to investigate the therapeutic potential of CRISPR/Cas9 in a diverse set of genetic disorders, we establish a pipeline that uses readily obtainable cells from affected individuals. We show that an adapted version of CRISPR/Cas9 increases the amount of utrophin, a known disease modifier in Duchenne muscular dystrophy (DMD). Furthermore, we demonstrate preferential elimination of the dominant-negative FGFR3 c.1138G>A allele in fibroblasts of an individual affected by achondroplasia. Using a previously undescribed approach involving single guide RNA, we successfully removed large genome rearrangement in primary cells of an individual with an X chromosome duplication including MECP2. Moreover, removal of a duplication of DMD exons 18-30 in myotubes of an individual affected by DMD produced full-length dystrophin. Our findings establish the far-reaching therapeutic utility of CRISPR/Cas9, which can be tailored to target numerous inherited disorders.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Enfermedades Genéticas Congénitas/terapia , Alelos , Expresión Génica , Enfermedades Genéticas Congénitas/genética , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia
5.
Hum Mol Genet ; 25(8): 1468-78, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26787513

RESUMEN

Defects in mRNA 3'end formation have been described to alter transcription termination, transport of the mRNA from the nucleus to the cytoplasm, stability of the mRNA and translation efficiency. Therefore, inhibition of polyadenylation may lead to gene silencing. Here, we choose facioscapulohumeral dystrophy (FSHD) as a model to determine whether or not targeting key 3' end elements involved in mRNA processing using antisense oligonucleotide drugs can be used as a strategy for gene silencing within a potentially therapeutic context. FSHD is a gain-of-function disease characterized by the aberrant expression of the Double homeobox 4 (DUX4) transcription factor leading to altered pathogenic deregulation of multiple genes in muscles. Here, we demonstrate that targeting either the mRNA polyadenylation signal and/or cleavage site is an efficient strategy to down-regulate DUX4 expression and to decrease the abnormally high-pathological expression of genes downstream of DUX4. We conclude that targeting key functional 3' end elements involved in pre-mRNA to mRNA maturation with antisense drugs can lead to efficient gene silencing and is thus a potentially effective therapeutic strategy for at least FSHD. Moreover, polyadenylation is a crucial step in the maturation of almost all eukaryotic mRNAs, and thus all mRNAs are virtually eligible for this antisense-mediated knockdown strategy.


Asunto(s)
Proteínas de Homeodominio/genética , Morfolinos/síntesis química , Distrofia Muscular Facioescapulohumeral/terapia , Oligonucleótidos Antisentido/síntesis química , Precursores del ARN/antagonistas & inhibidores , Regiones no Traducidas 3'/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Proteínas de Homeodominio/antagonistas & inhibidores , Humanos , Modelos Biológicos , Terapia Molecular Dirigida , Morfolinos/farmacología , Morfolinos/uso terapéutico , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/patología , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Poliadenilación/efectos de los fármacos , Precursores del ARN/química , Transducción de Señal
6.
Hum Mol Genet ; 25(16): 3555-3563, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27378686

RESUMEN

In preclinical models for Duchenne muscular dystrophy, dystrophin restoration during adeno-associated virus (AAV)-U7-mediated exon-skipping therapy was shown to decrease drastically after six months in treated muscles. This decline in efficacy is strongly correlated with the loss of the therapeutic AAV genomes, probably due to alterations of the dystrophic myofiber membranes. To improve the membrane integrity of the dystrophic myofibers at the time of AAV-U7 injection, mdx muscles were pre-treated with a single dose of the peptide-phosphorodiamidate morpholino (PPMO) antisense oligonucleotides that induced temporary dystrophin expression at the sarcolemma. The PPMO pre-treatment allowed efficient maintenance of AAV genomes in mdx muscles and enhanced the AAV-U7 therapy effect with a ten-fold increase of the protein level after 6 months. PPMO pre-treatment was also beneficial to AAV-mediated gene therapy with transfer of micro-dystrophin cDNA into muscles. Therefore, avoiding vector genome loss after AAV injection by PPMO pre-treatment would allow efficient long-term restoration of dystrophin and the use of lower and thus safer vector doses for Duchenne patients.


Asunto(s)
Distrofina/genética , Terapia Genética , Morfolinos/administración & dosificación , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Oligonucleótidos Antisentido/administración & dosificación , Animales , Dependovirus/genética , Exones/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Humanos , Ratones Endogámicos mdx , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Sarcolema/efectos de los fármacos , Sarcolema/patología
7.
Lancet ; 390(10101): 1489-1498, 2017 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-28728956

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is a severe, progressive, and rare neuromuscular, X-linked recessive disease. Dystrophin deficiency is the underlying cause of disease; therefore, mutation-specific therapies aimed at restoring dystrophin protein production are being explored. We aimed to assess the efficacy and safety of ataluren in ambulatory boys with nonsense mutation DMD. METHODS: We did this multicentre, randomised, double-blind, placebo-controlled, phase 3 trial at 54 sites in 18 countries located in North America, Europe, the Asia-Pacific region, and Latin America. Boys aged 7-16 years with nonsense mutation DMD and a baseline 6-minute walk distance (6MWD) of 150 m or more and 80% or less of the predicted normal value for age and height were randomly assigned (1:1), via permuted block randomisation (block size of four) using an interactive voice-response or web-response system, to receive ataluren orally three times daily (40 mg/kg per day) or matching placebo. Randomisation was stratified by age (<9 years vs ≥9 years), duration of previous corticosteroid use (6 months to <12 months vs ≥12 months), and baseline 6MWD (<350 m vs ≥350 m). Patients, parents and caregivers, investigational site personnel, PTC Therapeutics employees, and all other study personnel were masked to group allocation until after database lock. The primary endpoint was change in 6MWD from baseline to week 48. We additionally did a prespecified subgroup analysis of the primary endpoint, based on baseline 6MWD, which is reflective of anticipated rates of disease progression over 1 year. The primary analysis was by intention to treat. This study is registered with ClinicalTrials.gov, number NCT01826487. FINDINGS: Between March 26, 2013, and Aug 26, 2014, we randomly assigned 230 patients to receive ataluren (n=115) or placebo (n=115); 228 patients comprised the intention-to-treat population. The least-squares mean change in 6MWD from baseline to week 48 was -47·7 m (SE 9·3) for ataluren-treated patients and -60·7 m (9·3) for placebo-treated patients (difference 13·0 m [SE 10·4], 95% CI -7·4 to 33·4; p=0·213). The least-squares mean change for ataluren versus placebo in the prespecified subgroups was -7·7 m (SE 24·1, 95% CI -54·9 to 39·5; p=0·749) in the group with a 6MWD of less than 300 m, 42·9 m (15·9, 11·8-74·0; p=0·007) in the group with a 6MWD of 300 m or more to less than 400 m, and -9·5 m (17·2, -43·2 to 24·2; p=0·580) in the group with a 6MWD of 400 m or more. Ataluren was generally well tolerated and most treatment-emergent adverse events were mild to moderate in severity. Eight (3%) patients (n=4 per group) reported serious adverse events; all except one event in the placebo group (abnormal hepatic function deemed possibly related to treatment) were deemed unrelated to treatment. INTERPRETATION: Change in 6MWD did not differ significantly between patients in the ataluren group and those in the placebo group, neither in the intention-to-treat population nor in the prespecified subgroups with a baseline 6MWD of less than 300 m or 400 m or more. However, we recorded a significant effect of ataluren in the prespecified subgroup of patients with a baseline 6MWD of 300 m or more to less than 400 m. Baseline 6MWD values within this range were associated with a more predictable rate of decline over 1 year; this finding has implications for the design of future DMD trials with the 6-minute walk test as the endpoint. FUNDING: PTC Therapeutics.


Asunto(s)
Codón sin Sentido/genética , Distrofia Muscular de Duchenne/tratamiento farmacológico , Oxadiazoles/administración & dosificación , Adolescente , Niño , Método Doble Ciego , Distrofina/deficiencia , Distrofina/genética , Salud Global , Humanos , Masculino , Distrofia Muscular de Duchenne/genética , Resultado del Tratamiento , Caminata
8.
Mol Ther ; 25(9): 2038-2052, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28663100

RESUMEN

One of the most promising therapeutic approaches for familial amyotrophic lateral sclerosis linked to superoxide dismutase 1 (SOD1) is the suppression of toxic mutant SOD1 in the affected tissues. Here, we report an innovative molecular strategy for inducing substantial, widespread, and sustained reduction of mutant human SOD1 (hSOD1) levels throughout the body of SOD1G93A mice, leading to therapeutic effects in animals. Adeno-associated virus serotype rh10 vectors (AAV10) were used to mediate exon skipping of the hSOD1 pre-mRNA by expression of exon-2-targeted antisense sequences embedded in a modified U7 small-nuclear RNA (AAV10-U7-hSOD). Skipping of hSOD1 exon 2 led to the generation of a premature termination codon, inducing production of a deleted transcript that was subsequently degraded by the activation of nonsense-mediated decay. Combined intravenous and intracerebroventricular delivery of AAV10-U7-hSOD increased the survival of SOD1G93A mice injected either at birth or at 50 days of age (by 92% and 58%, respectively) and prevented weight loss and the decline of neuromuscular function. This study reports the effectiveness of an exon-skipping approach in SOD1-ALS mice, supporting the translation of this technology to the treatment of this as yet incurable disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Superóxido Dismutasa-1/genética , Edad de Inicio , Esclerosis Amiotrófica Lateral/mortalidad , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Animales , Modelos Animales de Enfermedad , Exones , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Ratones Transgénicos , Actividad Motora/genética , Oligonucleótidos Antisentido , Sitios de Empalme de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recuperación de la Función , Superóxido Dismutasa-1/metabolismo , Tasa de Supervivencia , Transducción Genética
9.
Hum Mol Genet ; 24(17): 4916-32, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26060189

RESUMEN

Therapy-responsive biomarkers are an important and unmet need in the muscular dystrophy field where new treatments are currently in clinical trials. By using a comprehensive high-resolution mass spectrometry approach and western blot validation, we found that two fragments of the myofibrillar structural protein myomesin-3 (MYOM3) are abnormally present in sera of Duchenne muscular dystrophy (DMD) patients, limb-girdle muscular dystrophy type 2D (LGMD2D) and their respective animal models. Levels of MYOM3 fragments were assayed in therapeutic model systems: (1) restoration of dystrophin expression by antisense oligonucleotide-mediated exon-skipping in mdx mice and (2) stable restoration of α-sarcoglycan expression in KO-SGCA mice by systemic injection of a viral vector. Following administration of the therapeutic agents MYOM3 was restored toward wild-type levels. In the LGMD model, where different doses of vector were used, MYOM3 restoration was dose-dependent. MYOM3 fragments showed lower inter-individual variability compared with the commonly used creatine kinase assay, and correlated better with the restoration of the dystrophin-associated protein complex and muscle force. These data suggest that the MYOM3 fragments hold promise for minimally invasive assessment of experimental therapies for DMD and other neuromuscular disorders.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Conectina/metabolismo , Distrofias Musculares/metabolismo , Proteómica , Adolescente , Adulto , Animales , Biomarcadores , Estudios de Casos y Controles , Niño , Preescolar , Conectina/sangre , Creatina Quinasa , Modelos Animales de Enfermedad , Humanos , Espectrometría de Masas , Ratones , Ratones Endogámicos mdx , Distrofias Musculares/sangre , Distrofias Musculares/terapia , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/metabolismo , Proteómica/métodos , Resultado del Tratamiento , Adulto Joven
10.
Nucleic Acids Res ; 43(W1): W571-5, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25883154

RESUMEN

Given a query list of genes or proteins, CellWhere produces an interactive graphical display that mimics the structure of a cell, showing the local interaction network organized into subcellular locations. This user-friendly tool helps in the formulation of mechanistic hypotheses by enabling the experimental biologist to explore simultaneously two elements of functional context: (i) protein subcellular localization and (ii) protein-protein interactions or gene functional associations. Subcellular localization terms are obtained from public sources (the Gene Ontology and UniProt-together containing several thousand such terms) then mapped onto a smaller number of CellWhere localizations. These localizations include all major cell compartments, but the user may modify the mapping as desired. Protein-protein interaction listings, and their associated evidence strength scores, are obtained from the Mentha interactome server, or power-users may upload a pre-made network produced using some other interactomics tool. The Cytoscape.js JavaScript library is used in producing the graphical display. Importantly, for a protein that has been observed at multiple subcellular locations, users may prioritize the visual display of locations that are of special relevance to their research domain. CellWhere is at http://cellwhere-myology.rhcloud.com.


Asunto(s)
Mapeo de Interacción de Proteínas , Proteínas/análisis , Programas Informáticos , Gráficos por Computador , Genes , Internet , Espacio Intracelular/química
11.
Nat Genet ; 40(9): 1113-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18711368

RESUMEN

Pontocerebellar hypoplasias (PCH) represent a group of neurodegenerative autosomal recessive disorders with prenatal onset, atrophy or hypoplasia of the cerebellum, hypoplasia of the ventral pons, microcephaly, variable neocortical atrophy and severe mental and motor impairments. In two subtypes, PCH2 and PCH4, we identified mutations in three of the four different subunits of the tRNA-splicing endonuclease complex. Our findings point to RNA processing as a new basic cellular impairment in neurological disorders.


Asunto(s)
Cerebelo/anomalías , Endorribonucleasas/genética , Mutación , Puente/anomalías , Encéfalo/metabolismo , Mapeo Cromosómico , Cromosomas Humanos Par 17 , Humanos , Modelos Moleculares , Polimorfismo de Nucleótido Simple , Síndrome
12.
Lancet ; 385(9979): 1748-1757, 2015 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-25907158

RESUMEN

BACKGROUND: Cardiorespiratory failure is the leading cause of death in Duchenne muscular dystrophy. Based on preclinical and phase 2 evidence, we assessed the efficacy and safety of idebenone in young patients with Duchenne muscular dystrophy who were not taking concomitant glucocorticoids. METHODS: In a multicentre phase 3 trial in Belgium, Germany, the Netherlands, Switzerland, France, Sweden, Austria, Italy, Spain, and the USA, patients (age 10-18 years old) with Duchenne muscular dystrophy were randomly assigned in a one-to-one ratio with a central interactive web response system with a permuted block design with four patients per block to receive idebenone (300 mg three times a day) or matching placebo orally for 52 weeks. Study personnel and patients were masked to treatment assignment. The primary endpoint was change in peak expiratory flow (PEF) as percentage predicted (PEF%p) from baseline to week 52, measured with spirometry. Analysis was by intention to treat (ITT) and a modified ITT (mITT), which was prospectively defined to exclude patients with at least 20% difference in the yearly change in PEF%p, measured with hospital-based and weekly home-based spirometry. This study is registered with ClinicalTrials.gov, number NCT01027884. FINDINGS: 31 patients in the idebenone group and 33 in the placebo group comprised the ITT population, and 30 and 27 comprised the mITT population. Idebenone significantly attenuated the fall in PEF%p from baseline to week 52 in the mITT (-3·05%p [95% CI -7·08 to 0·97], p=0·134, vs placebo -9·01%p [-13·18 to -4·84], p=0·0001; difference 5·96%p [0·16 to 11·76], p=0·044) and ITT populations (-2·57%p [-6·68 to 1·54], p=0·215, vs -8·84%p [-12·73 to -4·95], p<0·0001; difference 6·27%p [0·61 to 11·93], p=0·031). Idebenone also had a significant effect on PEF (L/min), weekly home-based PEF, FVC, and FEV1. The effect of idebenone on respiratory function outcomes was similar between patients with previous corticosteroid use and steroid-naive patients. Treatment with idebenone was safe and well tolerated with adverse event rates were similar in both groups. Nasopharyngitis and headache were the most common adverse events (idebenone, eight [25%] and six [19%] of 32 patients; placebo, nine [26%] and seven [21%] of 34 patients). Transient and mild diarrhoea was more common in the idebenone group than in the placebo group (eight [25%] vs four [12%] patients). INTERPRETATION: Idebenone reduced the loss of respiratory function and represents a new treatment option for patients with Duchenne muscular dystrophy. FUNDING: Santhera Pharmaceuticals.


Asunto(s)
Antioxidantes/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Trastornos Respiratorios/tratamiento farmacológico , Ubiquinona/análogos & derivados , Adolescente , Niño , Método Doble Ciego , Humanos , Masculino , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/fisiopatología , Ápice del Flujo Espiratorio , Trastornos Respiratorios/etiología , Trastornos Respiratorios/fisiopatología , Pruebas de Función Respiratoria , Músculos Respiratorios/fisiopatología , Resultado del Tratamiento , Ubiquinona/uso terapéutico
13.
J Hum Genet ; 61(6): 571-3, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26888483

RESUMEN

Hereditary spastic paraplegia (HSP) is an extremely heterogeneous disease caused by mutations of numerous genes leading to lower limb spasticity (pure forms) that can be accompanied by neurological symptoms (complex forms). Despite recent advances, many causal mutations in patients remain unknown. We identified a consanguineous family with the early-onset HSP. Whole-exome sequencing revealed homozygosity for a novel Atlastin GTPase 1 gene stop mutation in three affected siblings. Heterozygous parents and siblings were unaffected. This was unexpected as mutations in the Atlastin 1 gene are known to cause autosomal dominant HSP. But our study showed that Atlastin 1 mutations may cause autosomal recessively inherited paraplegia with an underlying loss-of-function mechanism. Hence, patients with recessive forms of HSP should also be tested for the Atlastin 1 gene.


Asunto(s)
Proteínas de Unión al GTP/genética , Genes Recesivos , Homocigoto , Proteínas de la Membrana/genética , Mutación , Paraplejía Espástica Hereditaria/diagnóstico , Paraplejía Espástica Hereditaria/genética , Adolescente , Alelos , Sustitución de Aminoácidos , Niño , Consanguinidad , Análisis Mutacional de ADN , Proteínas de Unión al GTP/química , Humanos , Masculino , Proteínas de la Membrana/química , Modelos Moleculares , Linaje , Fenotipo , Conformación Proteica , Hermanos
14.
Mol Ther ; 23(5): 918-930, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25619725

RESUMEN

The promising clinical results obtained for ocular gene therapy in recent years have paved the way for gene supplementation to treat recessively inherited forms of retinal degeneration. The situation is more complex for dominant mutations, as the toxic mutant gene product must be removed. We used spliceosome-mediated RNA trans-splicing as a strategy for repairing the transcript of the rhodopsin gene, the gene most frequently mutated in autosomal dominant retinitis pigmentosa. We tested 17 different molecules targeting the pre-mRNA intron 1, by transient transfection of HEK-293T cells, with subsequent trans-splicing quantification at the transcript level. We found that the targeting of some parts of the intron promoted trans-splicing more efficiently than the targeting of other areas, and that trans-splicing rate could be increased by modifying the replacement sequence. We then developed cell lines stably expressing the rhodopsin gene, for the assessment of phenotypic criteria relevant to the pathogenesis of retinitis pigmentosa. Using this model, we showed that trans-splicing restored the correct localization of the protein to the plasma membrane. Finally, we tested our best candidate by AAV gene transfer in a mouse model of retinitis pigmentosa that expresses a mutant allele of the human rhodopsin gene, and demonstrated the feasibility of trans-splicing in vivo. This work paves the way for trans-splicing gene therapy to treat retinitis pigmentosa due to rhodopsin gene mutation and, more generally, for the treatment of genetic diseases with dominant transmission.


Asunto(s)
Terapia Genética , ARN Mensajero/genética , Retinitis Pigmentosa/genética , Rodopsina/genética , Trans-Empalme , Animales , Sitios de Unión , Línea Celular Transformada , Expresión Génica , Genes Dominantes , Vectores Genéticos/genética , Humanos , Intrones , Ratones , Ratones Transgénicos , Fenotipo , Células Fotorreceptoras/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Transporte de Proteínas , Precursores del ARN/genética , Empalme del ARN , Retinitis Pigmentosa/terapia , Rodopsina/química , Rodopsina/metabolismo , Transducción Genética
15.
Neurogenetics ; 16(1): 33-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25231362

RESUMEN

Autosomal recessive forms of Charcot-Marie-Tooth disease (ARCMT) are rare but severe disorders of the peripheral nervous system. Their molecular basis is poorly understood due to the extensive genetic and clinical heterogeneity, posing considerable challenges for patients, physicians, and researchers. We report on the genetic findings from a systematic study of a large collection of 174 independent ARCMT families. Initial sequencing of the three most common ARCMT genes (ganglioside-induced differentiation protein 1­GDAP1, SH3 domain and tetratricopeptide repeats-containing protein 2­SH3TC2, histidine-triad nucleotide binding protein 1­HINT1) identified pathogenic mutations in 41 patients. Subsequently, 87 selected nuclear families underwent single nucleotide polymorphism (SNP) genotyping and homozygosity mapping, followed by targeted screening of known ARCMT genes. This strategy provided molecular diagnosis to 22% of the families. Altogether, our unbiased genetic approach identified pathogenic mutations in ten ARCMT genes in a total of 41.3% patients. Apart from a newly described founder mutation in GDAP1, the majority of variants constitute private molecular defects. Since the gene testing was independent of the clinical phenotype of the patients, we identified mutations in patients with unusual or additional clinical features, extending the phenotypic spectrum of the SH3TC2 gene. Our study provides an overview of the ARCMT genetic landscape and proposes guidelines for tackling the genetic heterogeneity of this group of hereditary neuropathies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/diagnóstico , Enfermedad de Charcot-Marie-Tooth/genética , Mutación , Mapeo Cromosómico , Análisis Mutacional de ADN , Femenino , Genes Recesivos , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas del Tejido Nervioso/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Proteínas/genética
16.
Am J Pathol ; 184(11): 2885-98, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25194663

RESUMEN

Duchenne muscular dystrophy (DMD) is a fatal, X-linked neuromuscular disease that affects 1 boy in 3500 to 5000 boys. The golden retriever muscular dystrophy dog is the best clinically relevant DMD animal model. Here, we used a high-thoughput miRNA sequencing screening for identification of candidate serum miRNA biomarkers in golden retriever muscular dystrophy dogs. We confirmed the dysregulation of the previously described muscle miRNAs, miR-1, miR-133, miR-206, and miR-378, and identified a new candidate muscle miRNA, miR-95. We identified two other classes of dysregulated serum miRNAs in muscular dystrophy: miRNAs belonging to the largest known miRNA cluster that resides in the imprinting DLK1-DIO3 genomic region and miRNAs associated with cardiac disease, including miR-208a, miR-208b, and miR-499. No simple correlation was identified between serum levels of cardiac miRNAs and cardiac functional parameters in golden retriever muscular dystrophy dogs. Finally, we confirmed a dysregulation of miR-95, miR-208a, miR-208b, miR-499, and miR-539 in a small cohort of DMD patients. Given the interspecies conservation of miRNAs and preliminary data in DMD patients, these newly identified dysregulated miRNAs are strong candidate biomarkers for DMD patients.


Asunto(s)
Cardiomiopatías/metabolismo , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Animales , Biomarcadores/metabolismo , Cardiomiopatías/genética , Niño , Preescolar , Modelos Animales de Enfermedad , Perros , Humanos , Masculino , MicroARNs/genética , Distrofia Muscular de Duchenne/genética
17.
NMR Biomed ; 28(9): 1150-62, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26215733

RESUMEN

Quantitative NMRI and (31)P NMRS indices are reported in the forearms of 24 patients with Duchenne muscular dystrophy (DMD) (6-18 years, 14 non-ambulant) amenable to exon 53 skipping therapy and in 12 age-matched male controls (CONT). Examinations carried out at 3 T comprised multi-slice 17-echo measurements of muscle water T2 and heterogeneity, three-point Dixon imaging of fat fraction in flexor and extensor muscles (FLEX, EXT), and non-localised spectroscopy of phosphate metabolites. We studied four imaging indices, eight metabolic ratios combining ATP, phosphocreatine, phosphomonoesters and phosphodiesters, the cytosolic inorganic phosphate (Pia ) and an alkaline (Pib) pool present in dystrophic muscle, and average pH. All indices differed between DMD and CONT, except for muscle water T2 . Measurements were outside the 95th percentile of age-matched CONT values in over 65% of cases for percentage fat signal (%F), and in 78-100% of cases for all spectroscopic indices. T2 was elevated in one-third of FLEX measurements, whereas %pixels > 39 ms and T2 heterogeneity were abnormal in one-half of the examinations. The FLEX muscles had higher fat infiltration and T2 than EXT muscle groups. All indices, except pH, correlated with patient age, although the correlation was negative for T2 . However, in non-ambulant patients, the correlation with years since loss of ambulation was stronger than the correlation with age, and the slope of evolution per year was steeper after loss of ambulation. All indices except Pi/gATP differed between ambulant and non-ambulant patients; however, T2 and %pixels > 39 ms were highest in ambulant patients, possibly owing to the greater extent of inflammatory processes earlier in the disease. All other indices were worse in non-ambulant subjects. Quantitative measurements obtained from patients at different disease stages covered a broad range of abnormalities that evolved with the disease, and metabolic indices were up to 10-fold above normal from the onset, thus establishing a variety of potential markers for future therapy.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Distrofia Muscular de Duchenne/metabolismo , Adenosina Trifosfato/metabolismo , Adolescente , Niño , Progresión de la Enfermedad , Antebrazo , Humanos , Masculino
18.
Mol Ther ; 22(11): 1923-35, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25200009

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disorder caused by mutations in the dystrophin gene, without curative treatment yet available. Our study provides, for the first time, the overall safety profile and therapeutic dose of a recombinant adeno-associated virus vector, serotype 8 (rAAV8) carrying a modified U7snRNA sequence promoting exon skipping to restore a functional in-frame dystrophin transcript, and injected by locoregional transvenous perfusion of the forelimb. Eighteen Golden Retriever Muscular Dystrophy (GRMD) dogs were exposed to increasing doses of GMP-manufactured vector. Treatment was well tolerated in all, and no acute nor delayed adverse effect, including systemic and immune toxicity was detected. There was a dose relationship for the amount of exon skipping with up to 80% of myofibers expressing dystrophin at the highest dose. Similarly, histological, nuclear magnetic resonance pathological indices and strength improvement responded in a dose-dependent manner. The systematic comparison of effects using different independent methods, allowed to define a minimum threshold of dystrophin expressing fibers (>33% for structural measures and >40% for strength) under which there was no clear-cut therapeutic effect. Altogether, these results support the concept of a phase 1/2 trial of locoregional delivery into upper limbs of nonambulatory DMD patients.


Asunto(s)
Dependovirus/genética , Distrofina/genética , Miembro Anterior/fisiopatología , Distrofia Muscular de Duchenne/terapia , ARN Nuclear Pequeño/genética , Animales , Estudios de Cohortes , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Exones , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Infusiones Intravenosas , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , ARN Nuclear Pequeño/metabolismo
19.
Nucleic Acids Res ; 41(17): 8391-402, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23861443

RESUMEN

RNA-based therapeutic approaches using splice-switching oligonucleotides have been successfully applied to rescue dystrophin in Duchenne muscular dystrophy (DMD) preclinical models and are currently being evaluated in DMD patients. Although the modular structure of dystrophin protein tolerates internal deletions, many mutations that affect nondispensable domains of the protein require further strategies. Among these, trans-splicing technology is particularly attractive, as it allows the replacement of any mutated exon by its normal version as well as introducing missing exons or correcting duplication mutations. We have applied such a strategy in vitro by using cotransfection of pre-trans-splicing molecule (PTM) constructs along with a reporter minigene containing part of the dystrophin gene harboring the stop-codon mutation found in the mdx mouse model of DMD. Optimization of the different functional domains of the PTMs allowed achieving accurate and efficient trans-splicing of up to 30% of the transcript encoded by the cotransfected minigene. Optimized parameters included mRNA stabilization, choice of splice site sequence, inclusion of exon splice enhancers and artificial intronic sequence. Intramuscular delivery of adeno-associated virus vectors expressing PTMs allowed detectable levels of dystrophin in mdx and mdx4Cv, illustrating that a given PTM can be suitable for a variety of mutations.


Asunto(s)
Distrofina/genética , Trans-Empalme , Animales , Dependovirus/genética , Distrofina/análisis , Exones , Vectores Genéticos , Genotipo , Humanos , Intrones , Ratones , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/química , Músculos/química , Distrofia Muscular de Duchenne/genética , Células 3T3 NIH , Sitios de Empalme de ARN , ARN Mensajero/análisis
20.
Nat Genet ; 38(8): 917-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16845398

RESUMEN

Aicardi-Goutières syndrome (AGS) presents as a severe neurological brain disease and is a genetic mimic of the sequelae of transplacentally acquired viral infection. Evidence exists for a perturbation of innate immunity as a primary pathogenic event in the disease phenotype. Here, we show that TREX1, encoding the major mammalian 3' --> 5' DNA exonuclease, is the AGS1 gene, and AGS-causing mutations result in abrogation of TREX1 enzyme activity. Similar loss of function in the Trex1(-/-) mouse leads to an inflammatory phenotype. Our findings suggest an unanticipated role for TREX1 in processing or clearing anomalous DNA structures, failure of which results in the triggering of an abnormal innate immune response.


Asunto(s)
Exodesoxirribonucleasas/genética , Trastornos Heredodegenerativos del Sistema Nervioso/enzimología , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Mutación , Fosfoproteínas/genética , Proteínas/genética , Animales , Secuencia de Bases , ADN/genética , Exodesoxirribonucleasas/deficiencia , Trastornos Heredodegenerativos del Sistema Nervioso/inmunología , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fosfoproteínas/deficiencia , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA