Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Musculoskelet Disord ; 25(1): 35, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183077

RESUMEN

BACKGROUND: In facioscapulohumeral muscular dystrophy (FSHD), it is not known whether physical activity (PA) practiced at young age is associated with the clinical presentation of disease. To assess this issue, we performed a retrospective cohort study concerning the previous practice of sports and, among them, those with medium-high cardiovascular commitment in clinically categorized carriers of a D4Z4 reduced allele (DRA). METHODS: People aged between 18 and 60 were recruited as being DRA carriers. Subcategory (classical phenotype, A; incomplete phenotype, B; asymptomatic carriers, C; complex phenotype, D) and FSHD score, which measures muscle functional impairment, were assessed for all participants. Information on PAs was retrieved by using an online survey dealing with the practice of sports at a young age. RESULTS: 368 participants were included in the study, average age 36.6 years (SD = 9.4), 47.6% male. The FSHD subcategory A was observed in 157 (42.7%) participants with average (± SD) FSHD score of 5.8 ± 3.0; the incomplete phenotype (category B) in 46 (12.5%) participants (average score 2.2 ± 1.7) and the D phenotype in 61 (16.6%, average score 6.5 ± 3.8). Asymptomatic carriers were 104 (subcategory C, 28.3%, score 0.0 ± 0.2). Time from symptoms onset was higher for patients with A (15.8 ± 11.1 years) and D phenotype (13.3 ± 11.9) than for patients with B phenotype (7.3 ± 9.0). The practice of sports was associated with lower FSHD score (-17%) in participants with A phenotype (MR = 0.83, 95% CI = 0.73-0.95, p = 0.007) and by 33% in participants with D phenotype (MR = 0.67, 95% CI = 0.51-0.89, p = 0.006). Conversely, no improvement was observed in participants with incomplete phenotype with mild severity (B). CONCLUSIONS: PAs at a young age are associated with a lower clinical score in the adult A and D FSHD subcategories. These results corroborate the need to consider PAs at the young age as a fundamental indicator for the correct clinical stratification of the disease and its possible evolution.


Asunto(s)
Distrofia Muscular Facioescapulohumeral , Deportes , Adulto , Humanos , Masculino , Adolescente , Adulto Joven , Persona de Mediana Edad , Femenino , Distrofia Muscular Facioescapulohumeral/diagnóstico , Estudios Retrospectivos , Ejercicio Físico , Alelos
2.
Crit Rev Biochem Mol Biol ; 56(4): 360-372, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33823724

RESUMEN

Electron transfer flavoprotein dehydrogenase, also called ETF-ubiquinone oxidoreductase (ETF-QO), is a protein localized in the inner membrane of mitochondria, playing a central role in the electron-transfer system. Indeed, ETF-QO mediates electron transport from flavoprotein dehydrogenases to the ubiquinone pool. ETF-QO mutations are often associated with riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (RR-MADD, OMIM#231680), a multisystem genetic disease characterized by various clinical manifestations with different degrees of severity. In this review, we outline the clinical features correlated with ETF-QO deficiency and the benefits obtained from different treatments, such as riboflavin, L-carnitine and/or coenzyme Q10 supplementation, and a diet poor in fat and protein. Moreover, we provide a detailed summary of molecular and bioinformatic investigations, describing the mutations identified in ETFDH gene and highlighting their predicted impact on enzymatic structure and activity. In addition, we report biochemical and functional analysis, performed in HEK293 cells and patient fibroblasts and muscle cells, to show the relationship between the nature of ETFDH mutations, the variable impairment of enzyme function, and the different degrees of RR-MADD severity. Finally, we describe in detail 5 RR-MADD patients carrying different ETFDH mutations and presenting variable degrees of clinical symptom severity.


Asunto(s)
Flavoproteínas Transportadoras de Electrones , Proteínas Hierro-Azufre , Mitocondrias , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Animales , Carnitina/genética , Carnitina/metabolismo , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Humanos , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Mitocondrias/enzimología , Mitocondrias/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/enzimología , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/genética , Ubiquinona/metabolismo
3.
Mol Genet Metab ; 137(1-2): 213-222, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36155185

RESUMEN

Metabolic myopathies are characterized by the deficiency or dysfunction of essential metabolites or fuels to generate energy for muscle contraction; they most commonly manifest with neuromuscular symptoms due to impaired muscle development or functioning. We have summarized associations of signs and symptoms in 358 inherited metabolic diseases presenting with myopathies. This represents the tenth of a series of articles attempting to create and maintain a comprehensive list of clinical and metabolic differential diagnoses according to system involvement.


Asunto(s)
Enfermedades Metabólicas , Errores Innatos del Metabolismo , Enfermedades Musculares , Humanos , Enfermedades Musculares/metabolismo , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética , Enfermedades Metabólicas/genética
4.
Mol Cell Biochem ; 476(4): 1797-1811, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33452620

RESUMEN

Transportin3 (TNPO3) shuttles the SR proteins from the cytoplasm to the nucleus. The SR family includes essential splicing factors, such as SRSF1, that influence alternative splicing, controlling protein diversity in muscle and satellite cell differentiation. Given the importance of alternative splicing in the myogenic process and in the maintenance of healthy muscle, alterations in the splicing mechanism might contribute to the development of muscle disorders. Combining confocal, structured illumination and electron microscopy, we investigated the expression of TNPO3 and SRSF1 during myogenesis, looking at nuclear and cytoplasmic compartments. We investigated TNPO3 and its interaction with SRSF1 and we observed that SRSF1 remained mainly localized in the nucleus, while TNPO3 decreased in the cytoplasm and was strongly clustered in the nuclei of differentiated myotubes. In conclusion, combining different imaging techniques led us to describe the behavior of TNPO3 and SRSF1 during myogenesis, showing that their dynamics follow the myogenic process and could influence the proteomic network necessary during myogenesis. The combination of different high-, super- and ultra-resolution imaging techniques led us to describe the behavior of TNPO3 and its interaction with SRSF1, looking at nuclear and cytoplasmic compartments. These observations represent a first step in understanding the role of TNPO3 and SRFSF1 in complex mechanisms, such as myogenesis.


Asunto(s)
Núcleo Celular , Citoplasma , Desarrollo de Músculos , Factores de Empalme Serina-Arginina/metabolismo , beta Carioferinas/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Citoplasma/metabolismo , Citoplasma/ultraestructura , Ratones , Microscopía Confocal , Microscopía Electrónica
5.
Muscle Nerve ; 61(2): 253-257, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31729045

RESUMEN

BACKGROUND: Neutral lipid storage disease with myopathy (NLSDM) is a rare lipid metabolism disorder. In this study, we evaluated some circulating miRNAs levels in serum samples and the MRI of three affected siblings. METHODS: Three members of one NLSDM family were identified: two brothers and one sister. Muscles of lower and right upper extremities were studied by MRI. Expression profile of miRNAs, obtained from serum samples, was detected using qRT-PCR. RESULTS: Two brothers presented with progressive skeletal myopathy, while the sister had severe hepatosteatosis and diabetes. NLSDM patients showed a significant increase of muscle-specific miRNAs expression compared with healthy subjects. We found a correlation between hepatic damage and elevation of miRNAs expression profile of liver origin. CONCLUSIONS: The dysregulation of miRNAs might represent an indicator of skeletal and hepatic damage and it might be useful to monitor the progression of NLSDM.


Asunto(s)
Biomarcadores/sangre , Errores Innatos del Metabolismo Lipídico/sangre , Errores Innatos del Metabolismo Lipídico/genética , MicroARNs/sangre , Enfermedades Musculares/sangre , Enfermedades Musculares/genética , Edad de Inicio , Femenino , Humanos , Lipasa/genética , Errores Innatos del Metabolismo Lipídico/diagnóstico por imagen , Hígado/metabolismo , Hígado/patología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Músculo Esquelético/diagnóstico por imagen , Enfermedades Musculares/diagnóstico por imagen , Mutación/genética , Hermanos , Tomografía Computarizada por Rayos X
6.
Neurol Sci ; 41(10): 2953-2960, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32350671

RESUMEN

MiR-1 and myostatin are markers for muscle growth and regeneration. Myostatin has a key role in the regulation of muscle mass. Myotonic dystrophy type 1(DM1) patients have a disease-specific serum miRNA profile characterized by upregulation of miR-1, miR-206, miR-133a, and miR-133b (myomiRNAs).This study aims to evaluate the possible utility of myomiRs and myostatin as biomarkers of rehabilitation efficacy in DM1, supporting clinical outcomes that are often variable and related to the patient's clinical condition.In 9 genetically proven DM1 patients, we collected biological samples before (T0) and after (T1) exercise rehabilitation training as biological measurement. We measured serum myomiRNAs by qRT-PCR and myostatin by ELISA test. The clinical outcomes measures that we utilized during a 3-6 week rehabilitation controlled aerobic exercise period were the 6-min walking test (6MWT) that increased significantly of 53.5 m (p < 0.0004) and the 10-m walk test (10MWT) that decreased of 1.38 s.We observed, after physical rehabilitation, a significant downregulation of myomiRNAs and myostatin that occurred in parallel with the improvement of clinical functional outcome measures assessed as endurance and gait speed, respectively.The modulation of biomarkers may reflect muscle regeneration and increase muscle mass after aerobic exercise. miRNAs and myostatin might be considered as circulating biomarkers of DM1 rehabilitation. The efficacy of physical rehabilitation in counteracting molecular pathways responsible for muscle atrophy and disease progression and the role of these biomarkers in DM1 and other neuromuscular diseases warrant further investigation.


Asunto(s)
MicroARNs , Distrofia Miotónica , Biomarcadores , Humanos , Atrofia Muscular , Miostatina
7.
J Med Genet ; 56(5): 293-300, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30593463

RESUMEN

BACKGROUND: Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder, due to the loss of function of the survival motor neuron (SMN1) gene. The first treatment for the condition, recently approved, is based on the reduction of exon 7 skipping in mRNAs produced by a highly homologous gene (SMN2). The primary objective of the present study was to evaluate the applicability of the dosage of SMN gene produts in blood, as biomarker for SMA, and the safety of oral salbutamol, a beta2-adrenergic agonist modulating SMN2 levels. METHODS: We have performed a 1-year multicentre, double-blind, placebo-controlled study with salbutamol in 45 adult patients with SMA. Patients assumed 4 mg of salbutamol or placebo/three times a day. Molecular tests were SMN2 copy number, SMN transcript and protein levels. We have also explored the clinical effect, by the outcome measures available at the time of study design. RESULTS: Thirty-six patients completed the study. Salbutamol was safe and well tolerated. We observed a significant and progressive increase in SMN2 full-length levels in peripheral blood of the salbutamol-treated patients (p<0.00001). The exploratory analysis of motor function showed an improvement in most patients. CONCLUSIONS: Our data demonstrate safety and molecular efficacy of salbutamol. We provide the first longitudinal evaluation of SMN levels (both transcripts and protein) in placebo and in response to a compound modulating the gene expression: SMN transcript dosage in peripheral blood is reliable and may be used as pharmacodynamic marker in clinical trials with systemic compounds modifying SMN2levels. TRIAL REGISTRATION NUMBER: EudraCT no. 2007-001088-32.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Albuterol/uso terapéutico , Biomarcadores , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Atrofia Muscular Espinal/diagnóstico , Atrofia Muscular Espinal/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Resultado del Tratamiento , Adulto Joven
8.
Clin Neuropathol ; 39(3): 105-114, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32000889

RESUMEN

AIM: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease caused by the progressive degeneration of motor neurons. MicroRNAs are 17 - 27 nucleotide long molecules that regulate post-transcriptional mRNA expression. The aim of this study was to investigate the role of microRNAs in the skeletal muscle of ALS patients and correlate these results with the expression of histone deacetylase 4 (HDAC4) protein. MATERIALS AND METHODS: We measured the expression levels of muscle-specific microRNAs (miR-1, miR-133a, miR-133b, miR-206), inflammatory micro-RNAs (miR-27a, miR-221, miR-155), and HDAC4 protein content on western blotting in muscle biopsies obtained for diagnostic reasons in 18 ALS patients: 8 genetic forms (C9-ALS and SOD1-ALS), 5 sporadic cases (SALS), and 5 ALS cases affected only by upper motor neuron disease (UMN). RESULTS: In muscle of patients with genetic forms of ALS, we found a strong upregulation of miR-206, a muscle-specific miRNA involved in neuromuscular junction (NMJ), regeneration and muscle atrophy, and a decreased expression of HDAC4 protein levels, which is involved both in denervation and regulation of miR-206 in ALS pathophysiology. In these patients, we also observed an increase of inflammatory miRNAs. CONCLUSION: The different expression of miRNAs and HDAC4 in genetic ALS vs. SALS and UMN cases is likely to be correlated to different pathogenic mechanisms.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Histona Desacetilasas/genética , MicroARNs/genética , Músculo Esquelético/patología , Enfermedades Neurodegenerativas/patología , Proteínas Represoras/genética , Esclerosis Amiotrófica Lateral/genética , Femenino , Histona Desacetilasas/metabolismo , Humanos , Masculino , Enfermedades Neurodegenerativas/genética , Regulación hacia Arriba
9.
Int J Neurosci ; 130(12): 1272-1277, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32079439

RESUMEN

Purpose: We aimed at evaluating the feasibility of using MicroRNA (miR)-34a and miR-29b to detect inner ear damage in patients with mitochondrial disease (MD) and sensorineural hearing loss (SNHL).Material and Methods: Three patients with MD and SNHL and seven healthy control subjects were included in this case series. MD patients underwent pure tone audiometry (PTA), distortion product otoacoustic emission (DPOAE) and auditory brain response tests to investigate the specific cochlear and retrocochlear functions; control patients underwent PTA. MiR-34a and miR-29b were extracted from blood in all subjects included in the study. The expression of miR-34a and miR-29b in MD patients and healthy controls were statistically compared, then the expression of these two miRs was compared with DPOAE values.Results: In MD patients, miR-34a was significantly up-regulated compared to healthy controls; miR-34a and DPOAEs were negatively correlated. Conversely, miR-29b was up-regulated only in the youngest patient who suffered from the mildest forms of MD and SNHL, and negatively correlated with DPOAEs.Conclusion: In MD patients, miR-34a and miR-29b might be a marker of inner ear damage and early damage, respectively. Additional studies on larger samples are necessary to confirm these preliminary results.


Asunto(s)
Pérdida Auditiva Sensorineural/diagnóstico , Enfermedades del Laberinto/diagnóstico , MicroARNs/sangre , Enfermedades Mitocondriales/complicaciones , Factores de Edad , Biomarcadores/sangre , Pérdida Auditiva Sensorineural/sangre , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/fisiopatología , Pruebas Auditivas , Humanos , Enfermedades del Laberinto/sangre , Enfermedades del Laberinto/etiología , Enfermedades del Laberinto/fisiopatología , Enfermedades Mitocondriales/sangre , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/fisiopatología , Regulación hacia Arriba
10.
Int J Mol Sci ; 21(7)2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290091

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is characterized by incomplete penetrance and intra-familial clinical variability. The disease has been associated with the genetic and epigenetic features of the D4Z4 repetitive elements at 4q35. Recently, D4Z4 hypomethylation has been proposed as a reliable marker in the FSHD diagnosis. We exploited the Italian Registry for FSHD, in which FSHD families are classified using the Clinical Comprehensive Evaluation Form (CCEF). A total of 122 index cases showing a classical FSHD phenotype (CCEF, category A) and 110 relatives were selected to test with the receiver operating characteristic (ROC) curve, the diagnostic and predictive value of D4Z4 methylation. Moreover, we performed DNA methylation analysis in selected large families with reduced penetrance characterized by the co-presence of subjects carriers of one D4Z4 reduced allele with no signs of disease or presenting the classic FSHD clinical phenotype. We observed a wide variability in the D4Z4 methylation levels among index cases revealing no association with clinical manifestation or disease severity. By extending the analysis to family members, we revealed the low predictive value of D4Z4 methylation in detecting the affected condition. In view of the variability in D4Z4 methylation profiles observed in our large cohort, we conclude that D4Z4 methylation does not mirror the clinical expression of FSHD. We recommend that measurement of this epigenetic mark must be interpreted with caution in clinical practice.


Asunto(s)
Epigénesis Genética , Epigenómica , Estudios de Asociación Genética , Genotipo , Distrofia Muscular Facioescapulohumeral/diagnóstico , Distrofia Muscular Facioescapulohumeral/genética , Fenotipo , Alelos , Variación Biológica Poblacional , Metilación de ADN , Epigenómica/métodos , Familia , Predisposición Genética a la Enfermedad , Humanos , Linaje , Curva ROC
11.
Neurol Sci ; 40(3): 503-507, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30542963

RESUMEN

INTRODUCTION: The slow-channel congenital myasthenic syndrome (SCCMS) is a postsynaptic form of congenital myasthenic syndromes (CMSs), a clinically heterogeneous group of disorders caused by genetic defects leading to an abnormal signal transmission at the endplate. METHODS: We report clinical and molecular data of a multigenerational family in which the presentation of a progressive proximal-distal weakness with ocular involvement led to a number of different clinical diagnoses. RESULTS: A comprehensive genetic study which included whole-genome linkage analysis and whole-exome sequencing identified a heterozygous missense substitution (c.721C>T, p.L241F) in the ε subunit of the acetylcholine receptor (CHRNE) that was consistent with clinical weakness in all patients. DISCUSSION: SCCMS is characterized by a broad and heterogeneous clinical phenotype in which disease onset, symptoms, severity, and progression can be highly variable even between family members. The identification of a CHRNE mutation allowed to make the definitive diagnosis of CMS in this family and contributed to define the clinical spectrum of this disease.


Asunto(s)
Salud de la Familia , Mutación Missense/genética , Síndromes Miasténicos Congénitos/genética , Receptores Nicotínicos/genética , Adulto , Anciano , Electromiografía , Femenino , Ligamiento Genético , Humanos , Italia , Masculino , Persona de Mediana Edad , Músculo Esquelético/fisiopatología , Síndromes Miasténicos Congénitos/patología , Síndromes Miasténicos Congénitos/fisiopatología , Conducción Nerviosa/genética , Secuenciación del Exoma
12.
Am J Pathol ; 187(3): 654-664, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28082118

RESUMEN

Fibrosis is the main complication of muscular dystrophies. We identified moesin, a member of the ezrin-radixin-moesin family, in dystrophic muscles of mice representing Duchenne and congenital muscular dystrophies (DMD and CMD, respectively) and dysferlinopathy, but not in the wild type. High levels of moesin were also observed in muscle biopsy specimens from DMD, Ullrich CMD, and merosin-deficient CMD patients, all of which present high levels of fibrosis. The myofibroblasts, responsible for extracellular matrix protein synthesis, and the macrophages infiltrating the dystrophic muscles were the source of moesin. Moesin-positive cells were embedded within the fibrotic areas between the myofibers adjacent to the collagen type I fibers. Radixin was also synthesized by the myofibroblasts, whereas ezrin colocalized with the myofiber membranes. In animal models and patients' muscles, part of the moesin was in its active phosphorylated form. Inhibition of fibrosis by halofuginone, an antifibrotic agent, resulted in a major decrease in moesin levels in the muscles of DMD and CMD mice. In summary, the results of this study may pave the way for exploiting moesin as a novel target for intervention in MDs, and as part of a battery of biomarkers to evaluate treatment success in preclinical studies and clinical trials.


Asunto(s)
Distrofias Musculares/metabolismo , Adulto , Animales , Niño , Preescolar , Colágeno Tipo I/metabolismo , Proteínas del Citoesqueleto/metabolismo , Diafragma/efectos de los fármacos , Diafragma/metabolismo , Homocigoto , Humanos , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Proteínas de Microfilamentos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Quinazolinonas/farmacología
13.
Pharmacol Res ; 138: 43-56, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30219582

RESUMEN

Parkin, an E3 ubiquitin ligase and a Parkinson's disease (PD) related gene, translocates to impaired mitochondria and drives their elimination via autophagy, a process known as mitophagy. Mitochondrial pro-fusion protein Mitofusins (Mfn1 and Mfn2) were found to be a target for Parkin mediated ubiquitination. Mfns are transmembrane GTPase embedded in the outer membrane of mitochondria, which are required on adjacent mitochondria to mediate fusion. In mammals, Mfn2 also forms complexes that are capable of tethering mitochondria to endoplasmic reticulum (ER), a structural feature essential for mitochondrial energy metabolism, calcium (Ca2+) transfer between the organelles and Ca2+ dependent cell death. Despite its fundamental physiological role, the molecular mechanisms that control ER-mitochondria cross talk are obscure. Ubiquitination has recently emerged as a powerful tool to modulate protein function, via regulation of protein subcellular localization and protein ability to interact with other proteins. Ubiquitination is also a reversible mechanism, which can be actively controlled by opposing ubiquitination-deubiquitination events. In this work we found that in Parkin deficient cells and parkin mutant human fibroblasts, the tether between ER and mitochondria is decreased. We identified the site of Parkin dependent ubiquitination and showed that the non-ubiquitinatable Mfn2 mutant fails to restore ER-mitochondria physical and functional interaction. Finally, we took advantage of an established in vivo model of PD to demonstrate that manipulation of ER-mitochondria tethering by expressing an ER-mitochondria synthetic linker is sufficient to rescue the locomotor deficit associated to an in vivo Drosophila model of PD.


Asunto(s)
Retículo Endoplásmico/fisiología , GTP Fosfohidrolasas/fisiología , Mitocondrias/fisiología , Proteínas Mitocondriales/fisiología , Enfermedad de Parkinson/fisiopatología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Drosophila , Femenino , Fibroblastos/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Interferencia de ARN , ARN Interferente Pequeño/genética , Ubiquitinación
14.
Lipids Health Dis ; 17(1): 254, 2018 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-30424791

RESUMEN

BACKGROUND: Deficiency of electron transfer flavoprotein dehydrogenase (ETFDH) is associated with multiple acyl-CoA dehydrogenase deficiency (MADD). This disorder is an autosomal recessive lipid storage myopathy (LSM) that exhibits a wide range of clinical features, including myopathy, weakness and multisystem dysfunctions. Many patients with late onset of MADD improve when treated with riboflavin and are also referred to as RR-MADD (riboflavin-responsive multiple Acyl-CoA dehydrogenase disorder). METHODS: In this study, we report the clinical and genetic characterization of a novel RR-MADD patient. Biochemical data were obtained from analysis of muscle and plasma samples. DNA and RNA were extracted from peripheral blood, and sequence analysis and expression study of ETFDH gene were performed. Finally, the impact of mutations on ETFDH folding was evaluated using bioinformatic tools. RESULTS: Patient initially presented with vomiting, muscle weakness, and acidosis. Muscle biopsy revealed typical myopathological patterns of lipid storage myopathy and blood acylcarnitine profiles showed a combined elevation of long and medium chain acylcarnitines, supporting the diagnosis of RR-MADD. Molecular analysis of ETFDH gene revealed two heterozygous mutations, a novel splice variation in intron 10, c.1285 + 1G > A, and the previously reported c.560C > T missense mutation. RT-PCR analysis showed an alteration of ETFDH RNA splicing which in turn should lead to the production of a truncated protein. The in silico prediction analysis of ETFDH tridimensional structure demonstrated that the missense mutation resulted in instability and loss of protein activation, while the splice site variation induced a dramatic conformational change of the truncated protein. After MCT diet supplemented with carnitine and riboflavin, the patient showed significant biochemical and clinical improvement, in spite of severe molecular defect. CONCLUSION: This case report extends the spectrum of ETFDH mutations in MADD, providing further evidence that patients presenting at least one missense mutation in the FAD-binding domain may respond to either carnitine or riboflavin treatment, due to the recovery of some enzymatic activity.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Carnitina/uso terapéutico , Simulación por Computador , Análisis Mutacional de ADN , Quimioterapia Combinada , Flavoproteínas Transportadoras de Electrones/metabolismo , Femenino , Humanos , Proteínas Hierro-Azufre/metabolismo , Persona de Mediana Edad , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/enzimología , Músculo Esquelético/enzimología , Mutación Missense , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Conformación Proteica , Riboflavina/uso terapéutico
15.
Biochim Biophys Acta ; 1862(7): 1367-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27085974

RESUMEN

AIMS: Triglycerides droplets are massively stored in muscle in Lipid Storage Myopathies (LSM). We studied in muscle regulators of lipophagy, the expression of the transcription factor-EB (TFEB) (a master regulator of lysosomal biogenesis), and markers of autophagy which are induced by starvation and exert a transcriptional control on lipid catabolism. METHODS: We investigated the factors that regulate lipophagy in muscle biopsies from 6 patients with different types of LSM: 2 cases of riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (MADD), 1 case of primary carnitine deficiency (CD), 2 cases of neutral lipid storage myopathy (NLSD-M), 1 case of carnitine-palmitoyl-transferase-II (CPT) deficiency. RESULTS: Conventional morphology and electron microscopy documented the lipid accumulation and its dramatic resolution after treatment. Muscle immunofluorescence showed that while in MADD and NLSD-M there was a co-localized expression of TFEB and p62-SQSTM1 (marker of protein aggregates) in some atrophic fibers, in CD and CPT-II deficiency the reaction was almost normal. In regenerating fibers, TFEB localized in the cytoplasm (inactive form), whereas in atrophic fibers it localized in the nuclei (active form). Lipid-accumulated/atrophic fibers did not display p62-positive protein aggregates, indicating, together with the LC3-II (marker of autophagosomes) and p62-SQSTM1 analysis, that the autophagic flux is often preserved and lipophagy occurs. CONCLUSION: In atrophic and regenerating fibers of patients with NLSD-M we observed TFEB over-expression; in other conditions autophagy markers are increased, suggesting lipophagy active role on human lipid metabolism.


Asunto(s)
Cardiomiopatías/patología , Carnitina O-Palmitoiltransferasa/deficiencia , Carnitina/deficiencia , Hiperamonemia/patología , Errores Innatos del Metabolismo Lipídico/patología , Lipólisis , Errores Innatos del Metabolismo/patología , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/patología , Músculos/patología , Enfermedades Musculares/patología , Distrofias Musculares/patología , Adolescente , Adulto , Anciano , Autofagia , Cardiomiopatías/metabolismo , Carnitina/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Niño , Femenino , Humanos , Hiperamonemia/metabolismo , Errores Innatos del Metabolismo Lipídico/metabolismo , Masculino , Errores Innatos del Metabolismo/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Músculos/metabolismo , Enfermedades Musculares/metabolismo , Distrofias Musculares/metabolismo
16.
Muscle Nerve ; 55(1): 55-68, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27184587

RESUMEN

INTRODUCTION: Limb girdle muscular dystrophies (LGMDs) are characterized by high molecular heterogeneity, clinical overlap, and a paucity of specific biomarkers. Their molecular definition is fundamental for prognostic and therapeutic purposes. METHODS: We created an Italian LGMD registry that included 370 molecularly defined patients. We reviewed detailed retrospective and prospective data and compared each LGMD subtype for differential diagnosis purposes. RESULTS: LGMD types 2A and 2B are the most frequent forms in Italy. The ages at disease onset, clinical progression, and cardiac and respiratory involvement can vary greatly between each LGMD subtype. In a set of extensively studied patients, targeted next-generation sequencing (NGS) identified mutations in 36.5% of cases. CONCLUSION: Detailed clinical characterization combined with muscle tissue analysis is fundamental to guide differential diagnosis and to address molecular tests. NGS is useful for diagnosing forms without specific biomarkers, although, at least in our study cohort, several LGMD disease mechanisms remain to be identified. Muscle Nerve 55: 55-68, 2017.


Asunto(s)
Diagnóstico Diferencial , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/epidemiología , Adolescente , Adulto , Edad de Inicio , Anciano , Creatina Quinasa/sangre , Femenino , Estudios de Asociación Genética , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/complicaciones , Distrofia Muscular de Cinturas/genética , Sistema de Registros , Trastornos Respiratorios/etiología , Estadísticas no Paramétricas , Adulto Joven
17.
Neurol Sci ; 38(4): 619-625, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28078570

RESUMEN

We aimed to explore the cellular action of micro-RNAs that are non-coding-RNAs modulating gene expression, whose expression is dysregulated in myotonic dystrophy (DM1). Basic procedure was to measure the levels of muscle-specific myo-miRNAs (miR-1, miR-133a/b, miR-206) in muscle of 12 DM1 patients. Muscle fiber morphometry and a new grading of histopathological severity score were used to compare specific myo-miRNA level and fiber atrophy. We found that the levels of miR-1 and miR-133a/b were significantly decreased, while miR-206 was significantly increased as compared to controls. The histopathological score did not significantly correlate with the levels of myo-miRNAs, even if the lowest levels of miRNA-1 and miRNA-133a/b, and the highest levels of miRNA-206 were observed in patients with either severe histopathological scores or long disease duration. The histopathological score was inversely correlated with disease duration. Nowadays that DM1 muscle biopsies are scanty, since patients are usually diagnosed by genetic analysis, our study offers a unique opportunity to present miRNA expression profiles in muscle and correlate them to muscle morphology in this rare multisystem disorder. Our molecular and morphologic data suggest a post-transcriptional regulatory action of myo-miRNA in DM1, highlighting their potential role as biomarkers of muscle plasticity.


Asunto(s)
MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Miotónica/patología , Adulto , Atrofia , Biomarcadores/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Distrofia Miotónica/metabolismo , Índice de Severidad de la Enfermedad , Factores de Tiempo , Adulto Joven
18.
Biochim Biophys Acta ; 1852(4): 615-21, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24997454

RESUMEN

Metabolic myopathies are disorders of utilization of carbohydrates or fat in muscles. The acute nature of energy failure is manifested either by a metabolic crisis with weakness, sometimes associated with respiratory failure, or by myoglobinuria. A typical disorder where permanent weakness occurs is glycogenosis type II (GSDII or Pompe disease) both in infantile and late-onset forms, where respiratory insufficiency is manifested by a large number of cases. In GSDII the pathogenetic mechanism is still poorly understood, and has to be attributed more to structural muscle alterations, possibly in correlation to macro-autophagy, rather than to energetic failure. This review is focused on recent advances about GSDII and its treatment, and the most recent notions about the management and treatment of other metabolic myopathies will be briefly reviewed, including glycogenosis type V (McArdle disease), glycogenosis type III (debrancher enzyme deficiency or Cori disease), CPT-II deficiency, and ETF-dehydrogenase deficiency (also known as riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency or RR-MADD). The discovery of the genetic defect in ETF dehydrogenase confirms the etiology of this syndrome. Other metabolic myopathies with massive lipid storage and weakness are carnitine deficiency, neutral lipid storage-myopathy (NLSD-M), besides RR-MADD. Enzyme replacement therapy is presented with critical consideration and for each of the lipid storage disorders, representative cases and their response to therapy is included. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/deficiencia , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno , Proteínas Hierro-Azufre/deficiencia , Errores Innatos del Metabolismo Lipídico , Enfermedades Musculares , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/deficiencia , Animales , Enfermedad del Almacenamiento de Glucógeno/enzimología , Enfermedad del Almacenamiento de Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno/patología , Enfermedad del Almacenamiento de Glucógeno/terapia , Humanos , Errores Innatos del Metabolismo Lipídico/enzimología , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/patología , Errores Innatos del Metabolismo Lipídico/terapia , Enfermedades Musculares/enzimología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/terapia
19.
Muscle Nerve ; 54(5): 821-835, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27501525

RESUMEN

Dysferlin-deficient limb girdle muscular dystrophy type 2B, distal Miyoshi myopathy, and other less frequent phenotypes are a group of recessive disorders called dysferlinopathies. They are characterized by wide clinical heterogeneity. To diagnose dysferlinopathy, a clinical neuromuscular workup, including electrophysiological and muscle imaging investigations, is essential to support subsequent laboratory testing. Increased serum creatine kinase levels, distal or proximal muscle weakness, and myalgia with onset in the second or third decades are the main clinical features of the disease. In muscle biopsies, severe dysferlin deficiency by immunoblot or its abnormal localization by immunohistochemistry are the gold standard, as they have a high diagnostic value. Dysferlin testing on monocytes is a valuable alternative to muscle immunoblotting. Molecular techniques for gene mutation detection, such as next generation sequencing, have improved the genetic diagnosis, which is crucial for treatment and genetic counselling. Muscle Nerve 54: 821-835, 2016.


Asunto(s)
Distrofia Muscular de Cinturas/diagnóstico , Progresión de la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo
20.
Muscle Nerve ; 53(6): 981-3, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26800218

RESUMEN

INTRODUCTION: We report a patient in whom the diagnosis of a treatable disease was delayed for 30 years. METHODS: Recent discoveries of next generation sequencing (NGS) have allowed us to reconsider the diagnosis of limb girdle muscular dystrophy (LGMD) cases of unknown etiology. RESULTS: A 36-year-old man appeared to have LGMD with onset in shoulder girdle muscles, but all sarcolemmal and cytoskeletal proteins tested by immunoblotting and immunohistochemistry gave normal results. He developed respiratory insufficiency and became dependent on overnight ventilation at age 44. By NGS technology, 2 mutations in the GAA gene (intervening sequence 1 and a missense mutation in exon 11) allowed us to make a definite diagnosis of glycogenosis type II (Pompe disease) and start enzyme replacement therapy at age 71. CONCLUSIONS: Mild nondystrophic features on muscle biopsy and respiratory muscle involvement should suggest late-onset Pompe disease in patients with an unclassified LGMD phenotype. NGS may help make the diagnosis. Muscle Nerve 53: 981-983, 2016.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Anciano , Creatina Quinasa/metabolismo , Proteínas del Citoesqueleto/metabolismo , Electromiografía , Humanos , Masculino , Distrofia Muscular de Cinturas/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA