Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neuropathol Appl Neurobiol ; : e12952, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38124360

RESUMEN

AIMS: Limb-girdle congenital myasthenic syndrome (LG-CMS) is a genetically heterogeneous disorder characterized by muscle weakness and fatigability. The LG-CMS gene DPAGT1 codes for an essential enzyme of the glycosylation pathway, a posttranslational modification mechanism shaping the structure and function of proteins. In DPAGT1-related LG-CMS, reduced glycosylation of the acetylcholine receptor (AChR) reduces its localization at the neuromuscular junction (NMJ), and results in diminished neuromuscular transmission. LG-CMS patients also show tubular aggregates on muscle biopsy, but the origin and potential contribution of the aggregates to disease development are not understood. Here, we describe two LG-CMS patients with the aim of providing a molecular diagnosis and to shed light on the pathways implicated in tubular aggregate formation. METHODS: Following clinical examination of the patients, we performed next-generation sequencing (NGS) to identify the genetic causes, analysed the biopsies at the histological and ultrastructural levels, investigated the composition of the tubular aggregates, and performed experiments on protein glycosylation. RESULTS: We identified novel pathogenic DPAGT1 variants in both patients, and pyridostigmine treatment quantitatively improved muscle force and function. The tubular aggregates contained proteins of the sarcoplasmic reticulum (SR) and structurally conformed to the aggregates observed in tubular aggregate myopathy (TAM). TAM arises from overactivation of the plasma membrane calcium channel ORAI1, and functional studies on muscle extracts from our LG-CMS patients evidenced abnormal ORAI1 glycosylation. CONCLUSIONS: We expand the genetic variant spectrum of LG-CMS and provide a genotype/phenotype correlation for pathogenic DPAGT1 variants. The discovery of ORAI1 hypoglycosylation in our patients highlights a physiopathological link between LG-CMS and TAM.

3.
J Hepatol ; 75(6): 1420-1433, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34453962

RESUMEN

BACKGROUND & AIMS: Therapeutic targeting of injuries that require transient restoration of proteins by mRNA delivery is an attractive approach that, until recently, has remained poorly explored. In this study, we examined the therapeutic utility of mRNA delivery for liver fibrosis and cirrhosis. Specifically, we aimed to demonstrate the therapeutic efficacy of human hepatocyte nuclear factor alpha (HNF4A) mRNA in mouse models of fibrosis and cirrhosis. METHODS: We investigated restoration of hepatocyte functions by HNF4A mRNA transfection in vitro, and analyzed the attenuation of liver fibrosis and cirrhosis in multiple mouse models, by delivering hepatocyte-targeted biodegradable lipid nanoparticles (LNPs) encapsulating HNF4A mRNA. To identify potential mechanisms of action, we performed microarray-based gene expression profiling, single-cell RNA sequencing, and chromatin immunoprecipitation. We used primary liver cells and human liver buds for additional functional validation. RESULTS: Expression of HNF4A mRNA led to restoration of the metabolic activity of fibrotic primary murine and human hepatocytes in vitro. Repeated in vivo delivery of LNP-encapsulated HNF4A mRNA induced a robust inhibition of fibrogenesis in 4 independent mouse models of hepatotoxin- and cholestasis-induced liver fibrosis. Mechanistically, we discovered that paraoxonase 1 is a direct target of HNF4A and it contributes to HNF4A-mediated attenuation of liver fibrosis via modulation of liver macrophages and hepatic stellate cells. CONCLUSION: Collectively, our findings provide the first direct preclinical evidence of the applicability of HNF4A mRNA therapeutics for the treatment of fibrosis in the liver. LAY SUMMARY: Liver fibrosis and cirrhosis remain unmet medical needs and contribute to high mortality worldwide. Herein, we take advantage of a promising therapeutic approach to treat liver fibrosis and cirrhosis. We demonstrate that restoration of a key gene, HNF4A, via mRNA encapsulated in lipid nanoparticles decreased injury in multiple mouse models of fibrosis and cirrhosis. Our study provides proof-of-concept that mRNA therapy is a promising strategy for reversing liver fibrosis and cirrhosis.


Asunto(s)
Factor Nuclear 4 del Hepatocito/farmacología , Cirrosis Hepática/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Factor Nuclear 4 del Hepatocito/uso terapéutico , Ratones , ARN Mensajero/farmacología , ARN Mensajero/uso terapéutico
4.
Acta Neuropathol Commun ; 8(1): 154, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32887649

RESUMEN

Filamin C (FLNc) is mainly expressed in striated muscle cells where it localizes to Z-discs, myotendinous junctions and intercalated discs. Recent studies have revealed numerous mutations in the FLNC gene causing familial and sporadic myopathies and cardiomyopathies with marked clinical variability. The most frequent myopathic mutation, p.W2710X, which is associated with myofibrillar myopathy, deletes the carboxy-terminal 16 amino acids from FLNc and abolishes the dimerization property of Ig-like domain 24. We previously characterized "knock-in" mice heterozygous for this mutation (p.W2711X), and have now investigated homozygous mice using protein and mRNA expression analyses, mass spectrometry, and extensive immunolocalization and ultrastructural studies. Although the latter mice display a relatively mild myopathy under normal conditions, our analyses identified major mechanisms causing the pathophysiology of this disease: in comparison to wildtype animals (i) the expression level of FLNc protein is drastically reduced; (ii) mutant FLNc is relocalized from Z-discs to particularly mechanically strained parts of muscle cells, i.e. myotendinous junctions and myofibrillar lesions; (iii) the number of lesions is greatly increased and these lesions lack Bcl2-associated athanogene 3 (BAG3) protein; (iv) the expression of heat shock protein beta-7 (HSPB7) is almost completely abolished. These findings indicate grave disturbances of BAG3-dependent and -independent autophagy pathways that are required for efficient lesion repair. In addition, our studies reveal general mechanisms of lesion formation and demonstrate that defective FLNc dimerization via its carboxy-terminal domain does not disturb assembly and basic function of myofibrils. An alternative, more amino-terminally located dimerization site might compensate for that loss. Since filamins function as stress sensors, our data further substantiate that FLNc is important for mechanosensing in the context of Z-disc stabilization and maintenance.


Asunto(s)
Filaminas/genética , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Sarcómeros/patología , Animales , Técnicas de Sustitución del Gen , Homocigoto , Ratones , Mutación , Miopatías Estructurales Congénitas/metabolismo , Sarcómeros/metabolismo
5.
Brain ; 143(2): 452-466, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32040565

RESUMEN

Brody disease is an autosomal recessive myopathy characterized by exercise-induced muscle stiffness due to mutations in the ATP2A1 gene. Almost 50 years after the initial case presentation, only 18 patients have been reported and many questions regarding the clinical phenotype and results of ancillary investigations remain unanswered, likely leading to incomplete recognition and consequently under-diagnosis. Additionally, little is known about the natural history of the disorder, genotype-phenotype correlations, and the effects of symptomatic treatment. We studied the largest cohort of Brody disease patients to date (n = 40), consisting of 22 new patients (19 novel mutations) and all 18 previously published patients. This observational study shows that the main feature of Brody disease is an exercise-induced muscle stiffness of the limbs, and often of the eyelids. Onset begins in childhood and there was no or only mild progression of symptoms over time. Four patients had episodes resembling malignant hyperthermia. The key finding at physical examination was delayed relaxation after repetitive contractions. Additionally, no atrophy was seen, muscle strength was generally preserved, and some patients had a remarkable athletic build. Symptomatic treatment was mostly ineffective or produced unacceptable side effects. EMG showed silent contractures in approximately half of the patients and no myotonia. Creatine kinase was normal or mildly elevated, and muscle biopsy showed mild myopathic changes with selective type II atrophy. Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) activity was reduced and western blot analysis showed decreased or absent SERCA1 protein. Based on this cohort, we conclude that Brody disease should be considered in cases of exercise-induced muscle stiffness. When physical examination shows delayed relaxation, and there are no myotonic discharges at electromyography, we recommend direct sequencing of the ATP2A1 gene or next generation sequencing with a myopathy panel. Aside from clinical features, SERCA activity measurement and SERCA1 western blot can assist in proving the pathogenicity of novel ATP2A1 mutations. Finally, patients with Brody disease may be at risk for malignant hyperthermia-like episodes, and therefore appropriate perioperative measures are recommended. This study will help improve understanding and recognition of Brody disease as a distinct myopathy in the broader field of calcium-related myopathies.


Asunto(s)
Enfermedades Musculares/genética , Mutación/genética , Miotonía Congénita/genética , Retículo Sarcoplasmático/metabolismo , Adolescente , Adulto , ATPasas Transportadoras de Calcio/genética , Niño , Femenino , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Fenotipo , Adulto Joven
6.
Behav Brain Res ; 363: 199-215, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30599154

RESUMEN

Gait and postural control dysfunction are prototypical symptoms compromising quality of life for patients with Parkinson's disease (PD). Hallmarks of cellular pathology are dopaminergic degeneration and accumulation of the cytosolic protein alpha-synuclein, linked to impaired autophagy-lysosome pathway (ALP) clearance. Physical exercise improves gait in PD patients and motor function in rodent lesion models. Moreover, exercise is considered neuroprotective and ALP induction has been reported, e.g. in human skeletal muscle, rodent peripheral and cerebral tissues. A combined analysis of how distinct exercise paradigms affect motor and central biochemical aspects of PD could maximize benefits for patients. Here we examine the effect of 4 weeks treadmill exercise intervention in 7-8 month non-lesioned mice on a) distinct gait categories, b) ALP activity, c) dopaminergic and alpha-synuclein homeostasis. The study includes wild type, alpha-synuclein knockout, and mice exclusively expressing human alpha-synuclein. Parameters of gait regularity and stability, activity, and dynamic postural control during unforced walk, were assessed by an automated system (CatWalk XT). At baseline, alpha-synuclein mouse models exhibited irregular and less active gait, with impaired dynamic postural control, compared to wild type mice. Treadmill exercise particularly improved speed and stride length, while increasing dual diagonal versus three-paw body support in both the alpha-synuclein knockout and transgenic mice. Biochemical analyses showed higher striatal tyrosine hydroxylase immuno-reactivity and reduced higher-order alpha-synuclein species in the cerebral cortex. However, no significant cerebral ALP induction was measured. In summary, treadmill exercise improved gait activity and postural stability, and promoted dopaminergic and alpha-synuclein homeostasis, without robustly inducing cerebral ALP.


Asunto(s)
Marcha/fisiología , Condicionamiento Físico Animal/fisiología , Esfuerzo Físico/fisiología , Animales , Autofagia/fisiología , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/fisiología , Terapia por Ejercicio/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Neuroprotección , Enfermedad de Parkinson/fisiopatología , Postura/fisiología , Sustancia Negra/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/fisiología
8.
J Neurol ; 264(8): 1791-1803, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28712002

RESUMEN

Mutations in GFPT1 (glutamine-fructose-6-phosphate transaminase 1), a gene encoding an enzyme involved in glycosylation of ubiquitous proteins, cause a limb-girdle congenital myasthenic syndrome (LG-CMS) with tubular aggregates (TAs) characterized predominantly by affection of the proximal skeletal muscles and presence of highly organized and remodeled sarcoplasmic tubules in patients' muscle biopsies. We report here the first long-term clinical follow-up of 11 French individuals suffering from LG-CMS with TAs due to GFPT1 mutations, of which nine are new. Our retrospective clinical evaluation stresses an evolution toward a myopathic weakness that occurs concomitantly to ineffectiveness of usual CMS treatments. Analysis of neuromuscular biopsies from three unrelated individuals demonstrates that the maintenance of neuromuscular junctions (NMJs) is dramatically impaired with loss of post-synaptic junctional folds and evidence of denervation-reinnervation processes affecting the three main NMJ components. Moreover, molecular analyses of the human muscle biopsies confirm glycosylation defects of proteins with reduced O-glycosylation and show reduced sialylation of transmembrane proteins in extra-junctional area. Altogether, these results pave the way for understanding the etiology of this rare neuromuscular disorder that may be considered as a "tubular aggregates myopathy with synaptopathy".


Asunto(s)
Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/patología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Unión Neuromuscular/patología , Adolescente , Adulto , Anciano , Femenino , Estudios de Seguimiento , Glicosilación , Humanos , Persona de Mediana Edad , Músculo Esquelético/enzimología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Síndromes Miasténicos Congénitos/enzimología , Miopatías Estructurales Congénitas/tratamiento farmacológico , Miopatías Estructurales Congénitas/enzimología , Unión Neuromuscular/enzimología , Estudios Prospectivos , Estudios Retrospectivos , Adulto Joven
9.
Neurology ; 87(8): 799-805, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27440146

RESUMEN

OBJECTIVE: To assess the clinical, genetic, and myopathologic findings in 2 cousins with lack of desmin, the response to salbutamol in one patient, and the neuromuscular endplate pathology in a knock-in mouse model for recessive desminopathy. METHODS: We performed clinical investigations in the patients, genetic studies for linkage mapping, exome sequencing, and qPCR for transcript quantification, assessment of efficacy of (3-month oral) salbutamol administration by muscle strength assessment, 6-minute walking test (6MWT), and forced vital capacity, analysis of neuromuscular endplate pathology in a homozygous R349P desmin knock-in mouse by immunofluorescence staining of the hind limb muscles, and quantitative 3D morphometry and expression studies of acetylcholine receptor genes by quantitative PCR. RESULTS: Both patients had infantile-onset weakness and fatigability, facial weakness with bilateral ptosis and ophthalmoparesis, generalized muscle weakness, and a decremental response over 10% on repetitive nerve stimulation. Salbutamol improved 6MWT and subjective motor function in the treated patient. Genetic analysis revealed previously unreported novel homozygous truncating desmin mutation c.345dupC leading to protein truncation and consequent fast degradation of the mutant mRNA. In the recessive desminopathy mouse with low expression of the mutant desmin protein, we demonstrated fragmented motor endplates with increased surface areas, volumes, and fluorescence intensities in conjunction with increased α and γ acetylcholine receptor subunit expression in oxidative soleus muscle. CONCLUSIONS: The patients were desmin-null and had myopathy, cardiomyopathy, and a congenital myasthenic syndrome. The data from man and mouse demonstrate that the complete lack as well as the markedly decreased expression of mutant R349P desmin impair the structural and functional integrity of neuromuscular endplates.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Albuterol/farmacología , Cardiomiopatías , Desmina/genética , Placa Motora/patología , Distrofias Musculares , Unión Neuromuscular/patología , Adolescente , Agonistas de Receptores Adrenérgicos beta 2/administración & dosificación , Albuterol/administración & dosificación , Animales , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/genética , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Niño , Consanguinidad , Desmina/deficiencia , Modelos Animales de Enfermedad , Femenino , Genes Recesivos , Humanos , Masculino , Ratones , Distrofias Musculares/tratamiento farmacológico , Distrofias Musculares/genética , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Linaje
10.
Acta Neuropathol ; 132(3): 453-73, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27393313

RESUMEN

Secondary mitochondrial dysfunction is a feature in a wide variety of human protein aggregate diseases caused by mutations in different proteins, both in the central nervous system and in striated muscle. The functional relationship between the expression of a mutated protein and mitochondrial dysfunction is largely unknown. In particular, the mechanism how this dysfunction drives the disease process is still elusive. To address this issue for protein aggregate myopathies, we performed a comprehensive, multi-level analysis of mitochondrial pathology in skeletal muscles of human patients with mutations in the intermediate filament protein desmin and in muscles of hetero- and homozygous knock-in mice carrying the R349P desmin mutation. We demonstrate that the expression of mutant desmin causes disruption of the extrasarcomeric desmin cytoskeleton and extensive mitochondrial abnormalities regarding subcellular distribution, number and shape. At the molecular level, we uncovered changes in the abundancy and assembly of the respiratory chain complexes and supercomplexes. In addition, we revealed a marked reduction of mtDNA- and nuclear DNA-encoded mitochondrial proteins in parallel with large-scale deletions in mtDNA and reduced mtDNA copy numbers. Hence, our data demonstrate that the expression of mutant desmin causes multi-level damage of mitochondria already in early stages of desminopathies.


Asunto(s)
Desmina/genética , Filamentos Intermedios/patología , Mitocondrias/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/genética , Animales , Citoesqueleto/metabolismo , Citoesqueleto/patología , Desmina/metabolismo , Humanos , Filamentos Intermedios/genética , Ratones Transgénicos , Mitocondrias/patología , Enfermedades Musculares/patología , Mutación/genética
11.
Hum Mol Genet ; 24(25): 7207-20, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26472074

RESUMEN

Filamin C (FLNC) mutations in humans cause myofibrillar myopathy (MFM) and cardiomyopathy, characterized by protein aggregation and myofibrillar degeneration. We generated the first patient-mimicking knock-in mouse harbouring the most common disease-causing filamin C mutation (p.W2710X). These heterozygous mice developed muscle weakness and myofibrillar instability, with formation of filamin C- and Xin-positive lesions streaming between Z-discs. These lesions, which are distinct from the classical MFM protein aggregates by their morphology and filamentous appearance, were greatly increased in number upon acute physical exercise in the mice. This pathology suggests that mutant filamin influences the mechanical stability of myofibrillar Z-discs, explaining the muscle weakness in mice and humans. Re-evaluation of biopsies from MFM-filaminopathy patients with different FLNC mutations revealed a similar, previously unreported lesion pathology, in addition to the classical protein aggregates, and suggested that structures previously interpreted as aggregates may be in part sarcomeric lesions. We postulate that these lesions define preclinical disease stages, preceding the formation of protein aggregates.


Asunto(s)
Músculo Esquelético/patología , Miofibrillas/patología , Animales , Filaminas/genética , Genotipo , Ratones , Microscopía Electrónica , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Distrofias Musculares/genética , Miofibrillas/genética , Fenotipo
12.
Neuromuscul Disord ; 25(10): 773-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26248958

RESUMEN

Brody disease was first described as a benign pseudo-myotonic disorder with muscular stiffness, which increased with exercise. Biochemical and genetic studies have pointed out its close relationship to a functional defect of the fast-twitch sarcoplasmic reticulum Ca(++) ATPase pump (SERCA1) encoded by the ATP2A1 gene located on chromosome 16. The histopathological features in this form of myopathy were generally described as non-specific, i.e. moderate degree of type 2 fibre atrophy and excess of internal nuclei. We here present the clinical and histopathological features of a patient with Brody disease over a 19-year follow-up period. This patient had two heterozygous ATP2A1 mutations and complained about muscle stiffness immediately after effort. He had suffered from this since early childhood and exhibited clinical symptoms mimicking myotonia. Histological, ultrastructural and cytogenetic analyses revealed morphologically abnormal nuclei with polyploidy. In this report, we discuss the possible links between the consequences of the genetic abnormality and the peculiar aspect of the nuclei.


Asunto(s)
Núcleo Celular/patología , Músculo Esquelético/ultraestructura , Miotonía Congénita/patología , Adulto , Estudios de Seguimiento , Humanos , Masculino , Tono Muscular , Mutación , Miotonía Congénita/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
13.
Biochem Biophys Res Commun ; 463(4): 1210-7, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26086101

RESUMEN

Protein turnover and quality control by the proteasome is of paramount importance for cell homeostasis. Dysfunction of the proteasome is associated with aging processes and human diseases such as neurodegeneration, cardiomyopathy, and cancer. The regulation, i.e. activation and inhibition of this fundamentally important protein degradation system, is still widely unexplored. We demonstrate here that the evolutionarily highly conserved type II triple-A ATPase VCP and the proteasome inhibitor PSMF1/PI31 interact directly, and antagonistically regulate proteasomal activity. Our data provide novel insights into the regulation of proteasomal activity.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Proteínas de Ciclo Celular/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas/fisiología , Biopolímeros , Humanos , Proteína que Contiene Valosina
14.
Acta Neuropathol ; 129(2): 297-315, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25394388

RESUMEN

Mutations of the human desmin gene on chromosome 2q35 cause autosomal dominant, autosomal recessive and sporadic forms of protein aggregation myopathies and cardiomyopathies. We generated R349P desmin knock-in mice, which harbor the ortholog of the most frequently occurring human desmin missense mutation R350P. These mice develop age-dependent desmin-positive protein aggregation pathology, skeletal muscle weakness, dilated cardiomyopathy, as well as cardiac arrhythmias and conduction defects. For the first time, we report the expression level and subcellular distribution of mutant versus wild-type desmin in our mouse model as well as in skeletal muscle specimens derived from human R350P desminopathies. Furthermore, we demonstrate that the missense-mutant desmin inflicts changes of the subcellular localization and turnover of desmin itself and of direct desmin-binding partners. Our findings unveil a novel principle of pathogenesis, in which not the presence of protein aggregates, but disruption of the extrasarcomeric intermediate filament network leads to increased mechanical vulnerability of muscle fibers. These structural defects elicited at the myofiber level finally impact the entire organ and subsequently cause myopathy and cardiomyopathy.


Asunto(s)
Desmina/genética , Desmina/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miocardio/patología , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Citoesqueleto/metabolismo , Citoesqueleto/patología , Modelos Animales de Enfermedad , Escherichia coli , Técnicas de Sustitución del Gen , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Ratones Transgénicos , Debilidad Muscular/patología , Debilidad Muscular/fisiopatología , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Mutación Missense , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera
15.
J Med Genet ; 51(12): 824-33, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25326555

RESUMEN

BACKGROUND: Tubular aggregate myopathies (TAMs) are muscle disorders characterised by abnormal accumulations of densely packed single-walled or double-walled membrane tubules in muscle fibres. Recently, STIM1, encoding a major calcium sensor of the endoplasmic reticulum, was identified as a TAM gene. METHODS: The present study aims to define the clinical, histological and ultrastructural phenotype of tubular aggregate myopathy and to assess the STIM1 mutation spectrum. RESULTS: We describe six new TAM families harbouring one known and four novel STIM1 mutations. All identified mutations are heterozygous missense mutations affecting highly conserved amino acids in the calcium-binding EF-hand domains, demonstrating the presence of a mutation hot spot for TAM. We show that the mutations induce constitutive STIM1 clustering, strongly suggesting that calcium sensing and consequently calcium homoeostasis is impaired. Histological and ultrastructural analyses define a common picture with tubular aggregates labelled with Gomori trichrome and Nicotinamide adenine dinucleotide (NADH) tetrazolium reductase, substantiating their endoplasmic reticulum origin. The aggregates were observed in both fibre types and were often accompanied by nuclear internalisation and fibre size variability. The phenotypical spectrum ranged from childhood onset progressive muscle weakness and elevated creatine kinase levels to adult-onset myalgia without muscle weakness and normal CK levels. CONCLUSIONS: The present study expands the phenotypical spectrum of STIM1-related tubular aggregate myopathy. STIM1 should therefore be considered for patients with tubular aggregate myopathies involving either muscle weakness or myalgia as the first and predominant clinical sign.


Asunto(s)
Proteínas de la Membrana/genética , Músculo Esquelético/patología , Mutación , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/genética , Fenotipo , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Biopsia , Calcio/metabolismo , Línea Celular , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Proteínas de la Membrana/química , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Miopatías Estructurales Congénitas/metabolismo , Proteínas de Neoplasias/química , Linaje , Conformación Proteica , Alineación de Secuencia , Molécula de Interacción Estromal 1
16.
Neuromuscul Disord ; 24(11): 960-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25042397

RESUMEN

Disease processes and trauma affecting nerve-evoked muscle activity, motor neurons, synapses and myofibers cause different levels of muscle weakness, i.e., reduced maximal force production in response to voluntary activation or nerve stimulation. However, the mechanisms of muscle weakness are not well known. Using murine models of amyotrophic lateral sclerosis (SOD1(G93A) transgenic mice), congenital myasthenic syndrome (AChE knockout mice and Musk(V789M/-) mutant mice), Schwartz-Jampel syndrome (Hspg2(C1532YNEO/C1532YNEO) mutant mice) and traumatic nerve injury (Neurotomized wild-type mice), we show that the reduced maximal activation capacity (the ability of the nerve to maximally activate the muscle) explains 52%, 58% and 100% of severe weakness in respectively SOD1(G93A), Neurotomized and Musk mice, whereas muscle atrophy only explains 37%, 27% and 0%. We also demonstrate that the impaired maximal activation capacity observed in SOD1, Neurotomized, and Musk mice is not highly related to Hdac4 gene upregulation. Moreover, in SOD1 and Neurotomized mice our results suggest LC3, Fn14, Bcl3 and Gadd45a as candidate genes involved in the maintenance of the severe atrophic state. In conclusion, our study indicates that muscle weakness can result from the triggering of different signaling pathways. This knowledge may be helpful in designing therapeutic strategies and finding new drug targets for amyotrophic lateral sclerosis, congenital myasthenic syndrome, Schwartz-Jampel syndrome and nerve injury.


Asunto(s)
Potenciales Evocados Motores/fisiología , Neuronas Motoras/patología , Debilidad Muscular/etiología , Enfermedades Musculares/complicaciones , Miofibrillas/patología , Unión Neuroefectora/fisiopatología , Animales , Colinesterasas/genética , Modelos Animales de Enfermedad , Electromiografía , Potenciales Evocados Motores/genética , Regulación de la Expresión Génica/genética , Proteoglicanos de Heparán Sulfato/genética , Ratones , Ratones Transgénicos , Músculo Esquelético/fisiopatología , Enfermedades Musculares/genética , Mutación/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores del Factor de Necrosis Tumoral/genética , Superóxido Dismutasa/genética , Receptor de TWEAK
18.
Eur J Cell Biol ; 93(5-6): 252-66, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24598388

RESUMEN

Mutations in the human actin depolymerizing factor cofilin2 result in an autosomal dominant form of nemaline myopathy. Here, we report on the targeted ablation of murine cofilin2, which leads to a severe skeletal muscle specific phenotype within the first two weeks after birth. Apart from skeletal muscle, cofilin2 is also expressed in heart and CNS, however the pathology was restricted to skeletal muscle. The two close family members of cofilin2 - ADF and cofilin1 - were co-expressed in muscle, but unable to compensate for the loss of cofilin2. While primary myofibril assembly and muscle development were unaffected in cofilin2 mutant mice, progressive muscle degeneration was observed between postnatal days 3 and 7. Muscle pathology was characterized by sarcoplasmic protein aggregates, fiber size disproportion, mitochondrial abnormalities and internal nuclei. The observed muscle pathology differed from nemaline myopathy, but showed combined features of actin-associated myopathy and myofibrillar myopathy. In cofilin2 mutant mice, the postnatal expression pattern and turnover of sarcomeric α-actin isoforms were altered. Levels of smooth muscle α-actin were increased and remained high in developing muscles, suggesting that cofilin2 plays a crucial role during the exchange of α-actin isoforms during the early postnatal remodeling of the sarcomere.


Asunto(s)
Actinas/metabolismo , Cofilina 2/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Agregado de Proteínas/genética , Sarcómeros/metabolismo , Animales , Encéfalo/metabolismo , Cofilina 2/genética , Citoesqueleto/genética , Citoesqueleto/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/patología , Músculo Liso/crecimiento & desarrollo , Músculo Liso/metabolismo , Enfermedades Musculares/patología , Miocardio/metabolismo , Especificidad de Órganos
19.
J Neurol Neurosurg Psychiatry ; 85(3): 345-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23606733

RESUMEN

OBJECTIVE: Several families with characteristic features of hereditary myopathy with early respiratory failure (HMERF) have remained without genetic cause. This international study was initiated to clarify epidemiology and the genetic underlying cause in these families, and to characterise the phenotype in our large cohort. METHODS: DNA samples of all currently known families with HMERF without molecular genetic cause were obtained from 12 families in seven different countries. Clinical, histopathological and muscle imaging data were collected and five biopsy samples made available for further immunohistochemical studies. Genotyping, exome sequencing and Sanger sequencing were used to identify and confirm sequence variations. RESULTS: All patients with clinical diagnosis of HMERF were genetically solved by five different titin mutations identified. One mutation has been reported while four are novel, all located exclusively in the FN3 119 domain (A150) of A-band titin. One of the new mutations showed semirecessive inheritance pattern with subclinical myopathy in the heterozygous parents. Typical clinical features were respiratory failure at mid-adulthood in an ambulant patient with very variable degree of muscle weakness. Cytoplasmic bodies were retrospectively observed in all muscle biopsy samples and these were reactive for myofibrillar proteins but not for titin. CONCLUSIONS: We report an extensive collection of families with HMERF with five different mutations in exon 343 of TTN, which establishes this exon as the primary target for molecular diagnosis of HMERF. Our relatively large number of new families and mutations directly implies that HMERF is not extremely rare, not restricted to Northern Europe and should be considered in undetermined myogenic respiratory failure.


Asunto(s)
Enfermedades Genéticas Congénitas/epidemiología , Enfermedades Musculares/epidemiología , Insuficiencia Respiratoria/epidemiología , Adulto , Anciano , Conectina/genética , Exoma/genética , Femenino , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Ligamiento Genético/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Mutación/genética , Linaje , Fenotipo , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología
20.
Neuromuscul Disord ; 23(7): 587-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23746545

RESUMEN

Primary laminopathies caused by mutations in the LMNA gene typically display an extremely pleiotropic clinical presentation including cardiac, muscular and metabolic phenotypes. Additionally, many atypical laminopathies have been described combining features of two or more of the distinctive disorders or syndromes associated with LMNA mutations. We report on a 46-year-old female patient with a heterozygous p.R28W LMNA mutation, who presented with a novel clinical phenotype comprising severe limb-girdle muscular dystrophy, pronounced partial lipodystrophy, cardiac conduction defect, polycystic ovary disease and a metabolic syndrome with insulin-resistant diabetes mellitus and hypertriglyceridemia. On examination, her 23-year old daughter solely showed early signs of a LGMD phenotype.


Asunto(s)
Lamina Tipo A/genética , Distrofia Muscular de Cinturas/genética , Mutación/genética , Arritmias Cardíacas/genética , Síndrome de Brugada , Trastorno del Sistema de Conducción Cardíaco , Femenino , Predisposición Genética a la Enfermedad , Sistema de Conducción Cardíaco/anomalías , Heterocigoto , Humanos , Síndrome Metabólico/complicaciones , Persona de Mediana Edad , Distrofia Muscular de Cinturas/complicaciones , Distrofia Muscular de Cinturas/diagnóstico , Fenotipo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA