Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Eur Heart J ; 44(48): 5064-5073, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37639473

RESUMEN

BACKGROUND AND AIMS: Emery-Dreifuss muscular dystrophy (EDMD) is caused by variants in EMD (EDMD1) and LMNA (EDMD2). Cardiac conduction defects and atrial arrhythmia are common to both, but LMNA variants also cause end-stage heart failure (ESHF) and malignant ventricular arrhythmia (MVA). This study aimed to better characterize the cardiac complications of EMD variants. METHODS: Consecutively referred EMD variant-carriers were retrospectively recruited from 12 international cardiomyopathy units. MVA and ESHF incidences in male and female variant-carriers were determined. Male EMD variant-carriers with a cardiac phenotype at baseline (EMDCARDIAC) were compared with consecutively recruited male LMNA variant-carriers with a cardiac phenotype at baseline (LMNACARDIAC). RESULTS: Longitudinal follow-up data were available for 38 male and 21 female EMD variant-carriers [mean (SD) ages 33.4 (13.3) and 43.3 (16.8) years, respectively]. Nine (23.7%) males developed MVA and five (13.2%) developed ESHF during a median (inter-quartile range) follow-up of 65.0 (24.3-109.5) months. No female EMD variant-carrier had MVA or ESHF, but nine (42.8%) developed a cardiac phenotype at a median (inter-quartile range) age of 58.6 (53.2-60.4) years. Incidence rates for MVA were similar for EMDCARDIAC and LMNACARDIAC (4.8 and 6.6 per 100 person-years, respectively; log-rank P = .49). Incidence rates for ESHF were 2.4 and 5.9 per 100 person-years for EMDCARDIAC and LMNACARDIAC, respectively (log-rank P = .09). CONCLUSIONS: Male EMD variant-carriers have a risk of progressive heart failure and ventricular arrhythmias similar to that of male LMNA variant-carriers. Early implantable cardioverter defibrillator implantation and heart failure drug therapy should be considered in male EMD variant-carriers with cardiac disease.


Asunto(s)
Cardiopatías , Insuficiencia Cardíaca , Distrofia Muscular de Emery-Dreifuss , Distrofia Muscular de Emery-Dreifuss Ligada a X , Humanos , Masculino , Femenino , Persona de Mediana Edad , Distrofia Muscular de Emery-Dreifuss Ligada a X/complicaciones , Estudios Retrospectivos , Arritmias Cardíacas/epidemiología , Arritmias Cardíacas/genética , Arritmias Cardíacas/complicaciones , Cardiopatías/complicaciones , Distrofia Muscular de Emery-Dreifuss/complicaciones , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/complicaciones , Mutación
2.
Ann Neurol ; 89(2): 280-292, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33159473

RESUMEN

OBJECTIVE: This study was undertaken to determine whether a low residual quantity of dystrophin protein is associated with delayed clinical milestones in patients with DMD mutations. METHODS: We performed a retrospective multicentric cohort study by using molecular and clinical data from patients with DMD mutations registered in the Universal Mutation Database-DMD France database. Patients with intronic, splice site, or nonsense DMD mutations, with available muscle biopsy Western blot data, were included irrespective of whether they presented with severe Duchenne muscular dystrophy (DMD) or milder Becker muscular dystrophy (BMD). Patients were separated into 3 groups based on dystrophin protein levels. Clinical outcomes were ages at appearance of first symptoms; loss of ambulation; fall in vital capacity and left ventricular ejection fraction; interventions such as spinal fusion, tracheostomy, and noninvasive ventilation; and death. RESULTS: Of 3,880 patients with DMD mutations, 90 with mutations of interest were included. Forty-two patients expressed no dystrophin (group A), and 31 of 42 (74%) developed DMD. Thirty-four patients had dystrophin quantities < 5% (group B), and 21 of 34 (61%) developed BMD. Fourteen patients had dystrophin quantities ≥ 5% (group C), and all but 4 who lost ambulation beyond 24 years of age were ambulant. Dystrophin quantities of <5%, as low as <0.5%, were associated with milder phenotype for most of the evaluated clinical outcomes, including age at loss of ambulation (p < 0.001). INTERPRETATION: Very low residual dystrophin protein quantity can cause a shift in disease phenotype from DMD toward BMD. ANN NEUROL 2021;89:280-292.


Asunto(s)
Distrofina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Adolescente , Corticoesteroides/uso terapéutico , Adulto , Edad de Inicio , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Western Blotting , Niño , Estudios de Cohortes , Progresión de la Enfermedad , Distrofina/genética , Humanos , Masculino , Limitación de la Movilidad , Mortalidad , Distrofia Muscular de Duchenne/terapia , Ventilación no Invasiva/estadística & datos numéricos , Oxadiazoles/uso terapéutico , Fenotipo , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Fusión Vertebral/estadística & datos numéricos , Volumen Sistólico , Traqueostomía/estadística & datos numéricos , Capacidad Vital , Adulto Joven
3.
Neuromuscul Disord ; 29(9): 678-683, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31474437

RESUMEN

Emery-Dreifuss muscular dystrophy is an early-onset, slowly progressive myopathy characterized by the development of multiple contractures, muscle weakness and cardiac dysfunction. We present here the case of a 65-year-old male patient with a 20 year history of slowly progressive camptocormia, bradycardia and shortness of breath. Examination showed severe spine extensor and neck flexor muscle weakness with slight upper limb proximal weakness. Cardiologic assessment revealed slow atrial fibrillation. Whole body MRI demonstrated adipose substitution of the paravertebral, limb girdle and peroneal muscles as well as the tongue. Emerin immunohistochemistry on patient muscle biopsy revealed the absence of nuclear envelope labeling confirmed by Western Blot. Genetic analysis showed a hemizygous duplication of 5 bases in exon 6 of the EMD, emerin, gene on the X chromosome. This is an unusual presentation of X-linked Emery-Dreifuss muscular dystrophy with adult onset, predominant axial muscles involvement and minimal joint contractures. Diagnosis was prompted by the analysis of emerin on muscle biopsy.


Asunto(s)
Proteínas de la Membrana/genética , Músculo Esquelético/diagnóstico por imagen , Distrofia Muscular de Emery-Dreifuss/genética , Proteínas Nucleares/genética , Edad de Inicio , Anciano , Fibrilación Atrial/fisiopatología , Músculos de la Espalda/diagnóstico por imagen , Bradicardia/fisiopatología , Contractura/fisiopatología , Músculo Deltoides/metabolismo , Músculo Deltoides/patología , Disnea/fisiopatología , Músculos Isquiosurales/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Músculos Masticadores/diagnóstico por imagen , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular Espinal/fisiopatología , Distrofia Muscular de Emery-Dreifuss/diagnóstico por imagen , Distrofia Muscular de Emery-Dreifuss/patología , Distrofia Muscular de Emery-Dreifuss/fisiopatología , Proteínas Nucleares/metabolismo , Índice de Severidad de la Enfermedad , Curvaturas de la Columna Vertebral/fisiopatología
4.
Skelet Muscle ; 8(1): 15, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29703249

RESUMEN

BACKGROUND: Duchenne (DMD) and Becker (BMD) muscular dystrophies are caused by mutations in the DMD gene coding for dystrophin, a protein being part of a large sarcolemmal protein scaffold that includes the neuronal nitric oxide synthase (nNOS). The nNOS was shown to play critical roles in a variety of muscle functions and alterations of its expression and location in dystrophic muscle fiber leads to an increase of the muscle fatigability. We previously revealed a decrease of nNOS expression in BMD patients all presenting a deletion of exons 45 to 55 in the DMD gene (BMDd45-55), impacting the nNOS binding site of dystrophin. Since several studies showed deregulation of microRNAs (miRNAs) in dystrophinopathies, we focused on miRNAs that could target nNOS in dystrophic context. METHODS: By a screening of 617 miRNAs in BMDd45-55 muscular biopsies using TLDA and an in silico study to determine which one could target nNOS, we selected four miRNAs. In order to select those that targeted a sequence of 3'UTR of NOS1, we performed luciferase gene reporter assay in HEK393T cells. Finally, expression of candidate miRNAs was modulated in control and DMD human myoblasts (DMDd45-52) to study their ability to target nNOS. RESULTS: TLDA assay and the in silico study allowed us to select four miRNAs overexpressed in muscle biopsies of BMDd45-55 compared to controls. Among them, only the overexpression of miR-31, miR-708, and miR-34c led to a decrease of luciferase activity in an NOS1-3'UTR-luciferase assay, confirming their interaction with the NOS1-3'UTR. The effect of these three miRNAs was investigated on control and DMDd45-52 myoblasts. First, we showed a decrease of nNOS expression when miR-708 or miR-34c were overexpressed in control myoblasts. We then confirmed that DMDd45-52 cells displayed an endogenous increased of miR-31, miR-708, and miR-34c and a decreased of nNOS expression, the same characteristics observed in BMDd45-55 biopsies. In DMDd45-52 cells, we demonstrated that the inhibition of miR-708 and miR-34c increased nNOS expression, confirming that both miRNAs can modulate nNOS expression in human myoblasts. CONCLUSION: These results strongly suggest that miR-708 and miR-34c, overexpressed in dystrophic context, are new actors involved in the regulation of nNOS expression in dystrophic muscle.


Asunto(s)
MicroARNs/genética , Distrofia Muscular de Duchenne/genética , Óxido Nítrico Sintasa de Tipo I/genética , Adolescente , Adulto , Anciano , Biopsia , Niño , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , MicroARNs/fisiología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Mioblastos/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
5.
Eur J Hum Genet ; 21(8): 855-63, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23299919

RESUMEN

The molecular basis underlying the clinical variability in symptomatic Duchenne muscular dystrophy (DMD) carriers are still to be precised. We report 26 cases of early symptomatic DMD carriers followed in the French neuromuscular network. Clinical presentation, muscular histological analysis and type of gene mutation, as well as X-chromosome inactivation (XCI) patterns using DNA extracted from peripheral blood or muscle are detailed. The initial symptoms were significant weakness (88%) or exercise intolerance (27%). Clinical severity varied from a Duchenne-like progression to a very mild Becker-like phenotype. Cardiac dysfunction was present in 19% of the cases. Cognitive impairment was worthy of notice, as 27% of the carriers are concerned. The muscular analysis was always contributive, revealing muscular dystrophy (83%), mosaic in immunostaining (81%) and dystrophin abnormalities in western blot analysis (84%). In all, 73% had exonic deletions or duplications and 27% had point mutations. XCI pattern was biased in 62% of the cases. In conclusion, we report the largest series of manifesting DMD carriers at pediatric age and show that exercise intolerance and cognitive impairment may reveal symptomatic DMD carriers. The complete histological and immunohistological study of the muscle is the key of the diagnosis leading to the dystrophin gene analysis. Our study shows also that cognitive impairment in symptomatic DMD carriers is associated with mutations in the distal part of the DMD gene. XCI study does not fully explain the mechanisms as well as the wide spectrum of clinical phenotype, though a clear correlation between the severity of the phenotype and inactivation bias was observed.


Asunto(s)
Distrofina/genética , Heterocigoto , Músculos/metabolismo , Distrofia Muscular de Duchenne/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Biopsia , Western Blotting , Niño , Preescolar , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Distrofina/metabolismo , Femenino , Francia/epidemiología , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Músculos/patología , Distrofia Muscular de Duchenne/epidemiología , Distrofia Muscular de Duchenne/patología , Mutación , Inactivación del Cromosoma X , Adulto Joven
6.
Eur J Hum Genet ; 19(6): 647-54, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21267004

RESUMEN

Mutation in ZMPSTE24 gene, encoding a major metalloprotease, leads to defective prelamin A processing and causes type B mandibuloacral dysplasia, as well as the lethal neonatal restrictive dermopathy syndrome. Phenotype severity is correlated with the residual enzyme activity of ZMPSTE24 and accumulation of prelamin A. We had previously demonstrated that a complete loss of function in ZMPSTE24 was lethal in the neonatal period, whereas compound heterozygous mutations including one PTC and one missense mutation were associated with type B mandibuloacral dysplasia. In this study, we report a 30-year longitudinal clinical survey of a patient harboring a novel severe and complex phenotype, combining an early-onset progeroid syndrome and a congenital myopathy with fiber-type disproportion. A unique homozygous missense ZMPSTE24 mutation (c.281T>C, p.Leu94Pro) was identified and predicted to produce two possible ZMPSTE24 conformations, leading to a partial loss of function. Western blot analysis revealed a major reduction of ZMPSTE24, together with the presence of unprocessed prelamin A and decreased levels of lamin A, in the patient's primary skin fibroblasts. These cells exhibited significant reductions in lifespan associated with major abnormalities of the nuclear shape and structure. This is the first report of MAD presenting with confirmed myopathic abnormalities associated with ZMPSTE24 defects, extending the clinical spectrum of ZMPSTE24 gene mutations. Moreover, our results suggest that defective prelamin A processing affects muscle regeneration and development, thus providing new insights into the disease mechanism of prelamin A-defective associated syndromes in general.


Asunto(s)
Anomalías Múltiples/genética , Acroosteólisis/genética , Lamina Tipo A/genética , Lipodistrofia/genética , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Distrofias Musculares/genética , Miopatías Estructurales Congénitas/genética , Proteínas Nucleares/genética , Progeria/genética , Precursores de Proteínas/genética , Anomalías Múltiples/fisiopatología , Acroosteólisis/complicaciones , Acroosteólisis/fisiopatología , Adulto , Secuencia de Aminoácidos , Técnicas de Cultivo de Célula , Femenino , Fibroblastos , Heterocigoto , Homocigoto , Humanos , Lamina Tipo A/metabolismo , Lipodistrofia/complicaciones , Lipodistrofia/fisiopatología , Mandíbula/anomalías , Mandíbula/fisiopatología , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Datos de Secuencia Molecular , Distrofias Musculares/complicaciones , Distrofias Musculares/fisiopatología , Mutación , Mutación Missense , Miopatías Estructurales Congénitas/complicaciones , Miopatías Estructurales Congénitas/fisiopatología , Proteínas Nucleares/metabolismo , Fenotipo , Progeria/complicaciones , Progeria/fisiopatología , Precursores de Proteínas/metabolismo
7.
Am J Hum Genet ; 85(3): 338-53, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19716112

RESUMEN

Emery-Dreifuss muscular dystrophy (EDMD) is a rare disorder characterized by early joint contractures, muscular dystrophy, and cardiac involvement with conduction defects and arrhythmias. So far, only 35% of EDMD cases are genetically elucidated and associated with EMD or LMNA gene mutations, suggesting the existence of additional major genes. By whole-genome scan, we identified linkage to the Xq26.3 locus containing the FHL1 gene in three informative families belonging to our EMD- and LMNA-negative cohort. Analysis of the FHL1 gene identified seven mutations, in the distal exons of FHL1 in these families, three additional families, and one isolated case, which differently affect the three FHL1 protein isoforms: two missense mutations affecting highly conserved cysteines, one abolishing the termination codon, and four out-of-frame insertions or deletions. The predominant phenotype was characterized by myopathy with scapulo-peroneal and/or axial distribution, as well as joint contractures, and associated with a peculiar cardiac disease characterized by conduction defects, arrhythmias, and hypertrophic cardiomyopathy in all index cases of the seven families. Heterozygous female carriers were either asymptomatic or had cardiac disease and/or mild myopathy. Interestingly, four of the FHL1-mutated male relatives had isolated cardiac disease, and an overt hypertrophic cardiomyopathy was present in two. Expression and functional studies demonstrated that the FHL1 proteins were severely reduced in all tested patients and that this was associated with a severe delay in myotube formation in the two patients for whom myoblasts were available. In conclusion, FHL1 should be considered as a gene associated with the X-linked EDMD phenotype, as well as with hypertrophic cardiomyopathy.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Musculares/genética , Distrofia Muscular de Emery-Dreifuss/genética , Mutación/genética , Adolescente , Adulto , Enfermedades Cardiovasculares/complicaciones , Diferenciación Celular , Niño , Preescolar , Cromosomas Humanos X/genética , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Técnica del Anticuerpo Fluorescente , Genes Ligados a X , Estudio de Asociación del Genoma Completo , Humanos , Immunoblotting , Proteínas con Dominio LIM , Escala de Lod , Enfermedades Pulmonares/complicaciones , Masculino , Persona de Mediana Edad , Distrofia Muscular de Emery-Dreifuss/complicaciones , Mioblastos/patología , Linaje , Isoformas de Proteínas/genética , Sarcómeros/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA