Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 95
Filtrer
1.
Int J Mol Sci ; 23(5)2022 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-35269765

RÉSUMÉ

BMD is characterized by a marked heterogeneity of gene mutations resulting in many abnormal dystrophin proteins with different expression and residual functions. The smaller dystrophin molecules lacking a portion around exon 48 of the rod domain, named the D8 region, are related to milder phenotypes. The study aimed to determine which proteins might contribute to preserving muscle function in these patients. Patients were subdivided, based on the absence or presence of deletions in the D8 region, into two groups, BMD1 and BMD2. Muscle extracts were analyzed by 2-D DIGE, label-free LC-ESI-MS/MS, and Ingenuity pathway analysis (IPA). Increased levels of proteins typical of fast fibers and of proteins involved in the sarcomere reorganization characterize BMD2. IPA of proteomics datasets indicated in BMD2 prevalence of glycolysis and gluconeogenesis and a correct flux through the TCA cycle enabling them to maintain both metabolism and epithelial adherens junction. A 2-D DIGE analysis revealed an increase of acetylated proteoforms of moonlighting proteins aldolase, enolase, and glyceraldehyde-3-phosphate dehydrogenase that can target the nucleus promoting stem cell recruitment and muscle regeneration. In BMD2, immunoblotting indicated higher levels of myogenin and lower levels of PAX7 and SIRT1/2 associated with a set of proteins identified by proteomics as involved in muscle homeostasis maintenance.


Sujet(s)
Dystrophine , Myopathie de Duchenne , Dystrophine/génétique , Dystrophine/métabolisme , Exons/génétique , Humains , Muscles/métabolisme , Myopathie de Duchenne/génétique , Phénotype , Spectrométrie de masse en tandem
2.
Elife ; 102021 09 20.
Article de Anglais | MEDLINE | ID: mdl-34542403

RÉSUMÉ

Background: Spinal muscular atrophy (SMA) is a neuromuscular disorder characterized by the degeneration of the second motor neuron. The phenotype ranges from very severe to very mild forms. All patients have the homozygous loss of the SMN1 gene and a variable number of SMN2 (generally 2-4 copies), inversely related to the severity. The amazing results of the available treatments have made compelling the need of prognostic biomarkers to predict the progression trajectories of patients. Besides the SMN2 products, few other biomarkers have been evaluated so far, including some miRs. Methods: We performed whole miRNome analysis of muscle samples of patients and controls (14 biopsies and 9 cultures). The levels of muscle differentially expressed miRs were evaluated in serum samples (51 patients and 37 controls) and integrated with SMN2 copies, SMN2 full-length transcript levels in blood and age (SMA-score). Results: Over 100 miRs were differentially expressed in SMA muscle; 3 of them (hsa-miR-181a-5p, -324-5p, -451a; SMA-miRs) were significantly upregulated in the serum of patients. The severity predicted by the SMA-score was related to that of the clinical classification at a correlation coefficient of 0.87 (p<10-5). Conclusions: miRNome analyses suggest the primary involvement of skeletal muscle in SMA pathogenesis. The SMA-miRs are likely actively released in the blood flow; their function and target cells require to be elucidated. The accuracy of the SMA-score needs to be verified in replicative studies: if confirmed, its use could be crucial for the routine prognostic assessment, also in presymptomatic patients. Funding: Telethon Italia (grant #GGP12116).


Sujet(s)
Marqueurs biologiques/sang , microARN/génétique , Muscles squelettiques/métabolisme , Amyotrophie spinale/génétique , Adolescent , Adulte , Marqueurs biologiques/analyse , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , microARN/sang , microARN/métabolisme , Adulte d'âge moyen , Amyotrophie spinale/sang , Amyotrophie spinale/métabolisme , Transcriptome
3.
Genes Dis ; 8(1): 73-78, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-33569515

RÉSUMÉ

Neutral lipid storage disease with myopathy (NLSDM) is a rare autosomal recessive disorder, due to an enzymatic error of lipid metabolism. Patients present always with skeletal muscle myopathy and variable cardiac and hepatic involvement. NLSDM is caused by mutations in the PNPLA2 gene, which encodes the adipose triglyceride lipase (ATGL). Here we report the molecular characterization and clinical findings of two NLSDM siblings carrying the novel c.187+1G > C homozygous PNPLA2 mutation, localized in the splice site of intron 2. Molecular analyses revealed that neither aberrant PNPLA2 mRNA isoforms, nor ATGL mutated protein were detectable in patient's cells. Clinically, both patients presented early onset muscle weakness, in particular of proximal upper limb muscles. In almost 15 years, muscle damage affected also distal upper limbs. This is a NLSDM family, displaying a severe PNPLA2 mutation in two siblings with clinical presentation characterized by an early onset, but a slowly evolution of severe myopathy.

4.
Front Neurol ; 11: 646, 2020.
Article de Anglais | MEDLINE | ID: mdl-32849172

RÉSUMÉ

Background: Four main clinical phenotypes have been traditionally described in patients mutated in SCN4A, including sodium-channel myotonia (SCM), paramyotonia congenita (PMC), Hypokaliemic type II (HypoPP2), and Hyperkaliemic/Normokaliemic periodic paralysis (HyperPP/NormoPP); in addition, rare phenotypes associated with mutations in SCN4A are congenital myasthenic syndrome and congenital myopathy. However, only scarce data have been reported in literature on large patient cohorts including phenotypes characterized by myotonia and episodes of paralysis. Methods: We retrospectively investigated clinical and molecular features of 80 patients fulfilling the following criteria: (1) clinical and neurophysiological diagnosis of myotonia, or clinical diagnosis of PP, and (2) presence of a pathogenic SCN4A gene variant. Patients presenting at birth with episodic laryngospasm or congenital myopathy-like phenotype with later onset of myotonia were considered as neonatal SCN4A. Results: PMC was observed in 36 (45%) patients, SCM in 30 (37.5%), Hyper/NormoPP in 7 (8.7%), HypoPP2 in 3 (3.7%), and neonatal SCN4A in 4 (5%). The median age at onset was significantly earlier in PMC than in SCM (p < 0.01) and in Hyper/NormoPP than in HypoPP2 (p = 0.02). Cold-induced myotonia was more frequently observed in PMC (n = 34) than in SCM (n = 23) (p = 0.04). No significant difference was found in age at onset of episodes of paralysis among PMC and PP or in frequency of permanent weakness between PP (n = 4), SCM (n = 5), and PMC (n = 10). PP was more frequently associated with mutations in the S4 region of the NaV1.4 channel protein compared to SCM and PMC (p < 0.01); mutations causing PMC were concentrated in the C-terminal region of the protein, while SCM-associated mutations were detected in all the protein domains. Conclusions: Our data suggest that skeletal muscle channelopathies associated with mutations in SCN4A represent a continuum in the clinical spectrum.

5.
J Med Genet ; 56(5): 293-300, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30593463

RÉSUMÉ

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.


Sujet(s)
Agonistes des récepteurs béta-2 adrénergiques/usage thérapeutique , Salbutamol/usage thérapeutique , Marqueurs biologiques , Amyotrophie spinale/traitement médicamenteux , Amyotrophie spinale/génétique , Protéine-1 de survie du motoneurone/génétique , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Expression des gènes , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Mâle , Adulte d'âge moyen , Amyotrophie spinale/diagnostic , Amyotrophie spinale/métabolisme , Protéine-1 de survie du motoneurone/métabolisme , Protéine-2 de survie du motoneurone/génétique , Protéine-2 de survie du motoneurone/métabolisme , Résultat thérapeutique , Jeune adulte
7.
Trials ; 19(1): 291, 2018 May 10.
Article de Anglais | MEDLINE | ID: mdl-29793540

RÉSUMÉ

BACKGROUND: Trials in rare diseases have many challenges, among which are the need to set up multiple sites in different countries to achieve recruitment targets and the divergent landscape of clinical trial regulations in those countries. Over the past years, there have been initiatives to facilitate the process of international study set-up, but the fruits of these deliberations require time to be operationally in place. FOR-DMD (Finding the Optimum Steroid Regimen for Duchenne Muscular Dystrophy) is an academic-led clinical trial which aims to find the optimum steroid regimen for Duchenne muscular dystrophy, funded by the National Institutes of Health (NIH) for 5 years (July 2010 to June 2015), anticipating that all sites (40 across the USA, Canada, the UK, Germany and Italy) would be open to recruitment from July 2011. However, study start-up was significantly delayed and recruitment did not start until January 2013. METHOD: The FOR-DMD study is used as an example to identify systematic problems in the set-up of international, multi-centre clinical trials. The full timeline of the FOR-DMD study, from funding approval to site activation, was collated and reviewed. Systematic issues were identified and grouped into (1) study set-up, e.g. drug procurement; (2) country set-up, e.g. competent authority applications; and (3) site set-up, e.g. contracts, to identify the main causes of delay and suggest areas where anticipatory action could overcome these obstacles in future studies. RESULTS: Time from the first contact to site activation across countries ranged from 6 to 24 months. Reasons of delay were universal (sponsor agreement, drug procurement, budgetary constraints), country specific (complexity and diversity of regulatory processes, indemnity requirements) and site specific (contracting and approvals). The main identified obstacles included (1) issues related to drug supply, (2) NIH requirements regarding contracting with non-US sites, (3) differing regulatory requirements in the five participating countries, (4) lack of national harmonisation with contracting and the requirement to negotiate terms and contract individually with each site and (5) diversity of languages needed for study materials. Additionally, as with many academic-led studies, the FOR-DMD study did not have access to the infrastructure and expertise that a contracted research organisation could provide, organisations often employed in pharmaceutical-sponsored studies. This delay impacted recruitment, challenged the clinical relevance of the study outcomes and potentially delayed the delivery of the best treatment to patients. CONCLUSION: Based on the FOR-DMD experience, and as an interim solution, we have devised a checklist of steps to not only anticipate and minimise delays in academic international trial initiation but also identify obstacles that will require a concerted effort on the part of many stakeholders to mitigate.


Sujet(s)
Liste de contrôle , Essais cliniques comme sujet/méthodes , Études multicentriques comme sujet/méthodes , Myopathie de Duchenne/traitement médicamenteux , Maladies rares/traitement médicamenteux , Plan de recherche , Stéroïdes/administration et posologie , Budgets , Essais cliniques comme sujet/économie , Essais cliniques comme sujet/législation et jurisprudence , Contrats , Humains , Coopération internationale , Études multicentriques comme sujet/économie , Études multicentriques comme sujet/législation et jurisprudence , Myopathie de Duchenne/diagnostic , Myopathie de Duchenne/économie , Sélection de patients , Maladies rares/diagnostic , Maladies rares/économie , Plan de recherche/législation et jurisprudence , Soutien financier à la recherche comme sujet , Stéroïdes/effets indésirables , Stéroïdes/ressources et distribution , Facteurs temps , Résultat thérapeutique
8.
Nucleus ; 9(1): 292-304, 2018 01 01.
Article de Anglais | MEDLINE | ID: mdl-29693488

RÉSUMÉ

Among rare diseases caused by mutations in LMNA gene, Emery-Dreifuss Muscular Dystrophy type 2 and Limb-Girdle muscular Dystrophy 1B are characterized by muscle weakness and wasting, joint contractures, cardiomyopathy with conduction system disorders. Circulating biomarkers for these pathologies have not been identified. Here, we analyzed the secretome of a cohort of patients affected by these muscular laminopathies in the attempt to identify a common signature. Multiplex cytokine assay showed that transforming growth factor beta 2 (TGF ß2) and interleukin 17 serum levels are consistently elevated in the vast majority of examined patients, while interleukin 6 and basic fibroblast growth factor are altered in subgroups of patients. Levels of TGF ß2 are also increased in fibroblast and myoblast cultures established from patient biopsies as well as in serum from mice bearing the H222P Lmna mutation causing Emery-Dreifuss Muscular Dystrophy in humans. Both patient serum and fibroblast conditioned media activated a TGF ß2-dependent fibrogenic program in normal human myoblasts and tenocytes and inhibited myoblast differentiation. Consistent with these results, a TGF ß2 neutralizing antibody avoided fibrogenic marker activation and myogenesis impairment. Cell intrinsic TGF ß2-dependent mechanisms were also determined in laminopathic cells, where TGF ß2 activated AKT/mTOR phosphorylation. These data show that TGF ß2 contributes to the pathogenesis of Emery-Dreifuss Muscular Dystrophy type 2 and Limb-Girdle muscular Dystrophy 1B and can be considered a potential biomarker of those diseases. Further, the evidence of TGF ß2 pathogenetic effects in tenocytes provides the first mechanistic insight into occurrence of joint contractures in muscular laminopathies.


Sujet(s)
Différenciation cellulaire , Cellules musculaires/anatomopathologie , Dystrophie musculaire d'Emery-Dreifuss/sang , Dystrophie musculaire d'Emery-Dreifuss/anatomopathologie , Ténocytes/anatomopathologie , Facteur de croissance transformant bêta-2/sang , Adulte , Animaux , Cellules cultivées , Femelle , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Humains , Mâle , Souris , Souris knockout , Adulte d'âge moyen , Cellules musculaires/métabolisme , Dystrophie musculaire d'Emery-Dreifuss/génétique , Dystrophie musculaire d'Emery-Dreifuss/métabolisme , Ténocytes/métabolisme , Facteur de croissance transformant bêta-2/génétique , Facteur de croissance transformant bêta-2/métabolisme , Jeune adulte
9.
Toxicol In Vitro ; 50: 124-136, 2018 Aug.
Article de Anglais | MEDLINE | ID: mdl-29522793

RÉSUMÉ

Botulin toxin (BTX) is widely used for treating skeletal muscle spasticity. Experimental reports on BTX treatment were mainly focused on the neuromuscular junction, while relatively little is known about toxin effects on the muscle cell itself. We investigated possible impact of BTX type A on skeletal muscle cell transcriptome by microarray analysis in muscle-derived cell cultures (fibroblasts, myoblasts and myotubes) from controls and spastic patients, and results were then validated at transcript and protein level. BTX-A treatment of control cells induced major changes in the myogenic component of the transcriptome, whereas the same treatment had a negligible effect in the fibrogenic component. BTX-A treatment of cell cultures from spastic patients induced an increased number of genes differentially expressed both in the fibrogenic and myogenic components. Specifically, BTX-A had a major effect on cell cycle-related genes in myoblasts, on muscle contraction-related genes in myotubes, and on extracellular matrix-related genes in fibroblasts from spastic patients. Our findings show that in vitro BTX-A treatment differentially affects transcript expression in muscle cells from spastic patients compared to those from controls suggesting a direct effect of BTX-A on muscle-specific functional pathways.


Sujet(s)
Toxines botuliniques de type A/toxicité , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Spasticité musculaire/génétique , Agents neuromusculaires/toxicité , Muscle quadriceps fémoral/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Adolescent , Adulte , Biopsie , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Humains , Adulte d'âge moyen , Fibres musculaires squelettiques/métabolisme , Spasticité musculaire/métabolisme , Spasticité musculaire/anatomopathologie , Myoblastes squelettiques/effets des médicaments et des substances chimiques , Myoblastes squelettiques/métabolisme , Muscle quadriceps fémoral/métabolisme , Muscle quadriceps fémoral/anatomopathologie , Jeune adulte
10.
Sleep Med ; 39: 32-37, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-29157585

RÉSUMÉ

OBJECTIVE: Myotonic dystrophy type 1 (DM1) is a hereditary myopathy characterized by an autosomal dominant inheritance with important cardiovascular and autonomic deregulation. DM1 patients have a high prevalence of obstructive sleep apnea (OSA), but the effects of this comorbidity on cardiovascular autonomic control (CAC) are unknown. The present study aimed to investigate CAC during sleep-wake cycle in DM1 patients, taking into account the effects of OSA comorbidity. METHOD: Twenty-three patients with a diagnosis of DM1, and a control group, underwent a complete polysomnographic study (PSG). Electrocardiogram and respiration were extracted from PSG, divided according to the sleep stages, and analyzed using spectral analysis (SpA) of heart rate variability (HRV). SpA identified three components: very low frequency (VLF), low frequency (LF), a marker of sympathetic modulation, and high frequency (HF), a marker of vagal modulation. RESULTS: The results showed that in DM1 patients, the sympathovagal balance shifted towards a vagal predominance during non-rapid eye movement (NREM) sleep and a sympathetic predominance during rapid eye movement (REM) sleep. Second, this preserved cardiac autonomic modulation was not affected by the comorbidity with obstructive sleep apnea syndrome (OSAS). Third, in DM1 patients, OSAS comorbidity was associated with a reduction in HRV during the whole sleep-wake cycle. Lastly, in DM1 patients with OSA, cardiorespiratory coupling was reduced compared to controls. CONCLUSIONS: DM1 patients had preserved cardiac autonomic dynamics during NREM and REM sleep, and this phenomenon was not affected by the presence of OSA. However, the comorbidity with OSA was characterized by a reduction in total HRV, which is a marker of the ability of autonomic control to respond to stressors stimuli.


Sujet(s)
Système nerveux autonome/physiopathologie , Système cardiovasculaire/physiopathologie , Dystrophie myotonique/physiopathologie , Syndrome d'apnées obstructives du sommeil/complications , Adulte , Femelle , Rythme cardiaque/physiologie , Humains , Mâle , Polysomnographie , Syndrome d'apnées obstructives du sommeil/physiopathologie , Phases du sommeil/physiologie , Nerf vague/physiologie
11.
Biochim Biophys Acta Gene Regul Mech ; 1860(11): 1138-1147, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-28867298

RÉSUMÉ

The dystrophin gene (DMD) is the largest gene in the human genome, mapping on the Xp21 chromosome locus. It spans 2.2Mb and accounts for approximately 0,1% of the entire human genome. Mutations in this gene cause Duchenne and Becker Muscular Dystrophy, X-linked Dilated Cardiomyopathy, and other milder muscle phenotypes. Beside the remarkable number of reports describing dystrophin gene expression and the pathogenic consequences of the gene mutations in dystrophinopathies, the full scenario of the DMD transcription dynamics remains however, poorly understood. Considering that the full transcription of the DMD gene requires about 16h, we have investigated the activity of RNA Polymerase II along the entire DMD locus within the context of specific chromatin modifications using a variety of chromatin-based techniques. Our results unveil a surprisingly powerful processivity of the RNA polymerase II along the entire 2.2Mb of the DMD locus with just one site of pausing around intron 52. We also discovered epigenetic marks highlighting the existence of four novel cis­DNA elements, two of which, located within intron 34 and exon 45, appear to govern the architecture of the DMD chromatin with implications on the expression levels of the muscle dystrophin mRNA. Overall, our findings provide a global view on how the entire DMD locus is dynamically transcribed by the RNA pol II and shed light on the mechanisms involved in dystrophin gene expression control, which can positively impact on the optimization of the novel ongoing therapeutic strategies for dystrophinopathies.


Sujet(s)
Dystrophine/génétique , Dystrophine/métabolisme , Muscles squelettiques/métabolisme , Séquences d'acides nucléiques régulatrices , Adolescent , Adulte , Animaux , Cellules cultivées , Enfant , Enfant d'âge préscolaire , Épigenèse génétique/physiologie , Régulation de l'expression des gènes , Cellules HeLa , Humains , Souris , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Mutation , Jeune adulte
12.
Orphanet J Rare Dis ; 12(1): 90, 2017 05 12.
Article de Anglais | MEDLINE | ID: mdl-28499397

RÉSUMÉ

BACKGROUND: A small number of patients affected by Neutral Lipid Storage Diseases (NLSDs: NLSD type M with Myopathy and NLSD type I with Ichthyosis) have been described in various ethnic groups worldwide. However, relatively little is known about the progression and phenotypic variability of the disease in large specific populations. The aim of our study was to assess the natural history, disability and genotype-phenotype correlations in Italian patients with NLSDs. Twenty-one patients who satisfied the criteria for NLSDs were enrolled in a retrospective cross-sectional study to evaluate the genetic aspects, clinical signs at onset, disability progression and comorbidities associated with this group of diseases. RESULTS: During the clinical follow-up (range: 2-44 years, median: 17.8 years), two patients (9.5%, both with NLSD-I) died of hepatic failure, and a further five (24%) lost their ability to walk or needed help when walking after a mean period of 30.6 years of disease. None of the patients required mechanical ventilation. No patient required a heart transplant, one patient with NLSD-M was implanted with a cardioverter defibrillator for severe arrhythmias. CONCLUSION: The genotype/phenotype correlation analysis in our population showed that the same gene mutations were associated with a varying clinical onset and course. This study highlights peculiar aspects of Italian NLSD patients that differ from those observed in Japanese patients, who were found to be affected by a marked hypertrophic cardiopathy. Owing to the varying phenotypic expression of the same mutations, it is conceivable that some additional genetic or epigenetic factors affect the symptoms and progression in this group of diseases.


Sujet(s)
Érythrodermie ichtyosiforme congénitale/génétique , Erreurs innées du métabolisme lipidique/génétique , Muscles squelettiques/métabolisme , Maladies musculaires/génétique , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études transversales , Femelle , Études d'associations génétiques , Génotype , Humains , Érythrodermie ichtyosiforme congénitale/anatomopathologie , Italie , Triacylglycerol lipase/génétique , Métabolisme lipidique/génétique , Erreurs innées du métabolisme lipidique/anatomopathologie , Mâle , Adulte d'âge moyen , Maladies musculaires/anatomopathologie , Mutation/génétique , Phénotype , Ventilation artificielle , Études rétrospectives , Jeune adulte
13.
Mol Genet Metab Rep ; 10: 24-27, 2017 Mar.
Article de Anglais | MEDLINE | ID: mdl-28070494

RÉSUMÉ

To date, only few mutations in the mitochondrial DNA (mtDNA)-encoded ND2 subunit of Complex I have been reported, usually presenting a severe phenotype characterized by early onset encephalomyopathy and early death. In this report, we describe a new mutation in the MTND2 gene in a 21-year-old man with a mild myopathic phenotype characterized by exercise intolerance and increased plasma lactate at rest. Electromyography and brain NMR were normal, and no cardiac involvement was present. Muscle biopsy showed a massive presence of ragged red - COX-positive fibres, with enlarged mitochondria containing osmiophilic inclusions. Biochemical assays revealed a severe isolated complex I deficiency. We identified a novel, heteroplasmic mutation m.4831G > A in the MTND2 gene, causing the p.Gly121Asp substitution in the ND2 protein. The mutation was present in the 95% of mitochondrial genomes from patient's muscle tissue, at a lower level in cells from the urinary tract and at a lowest level in lymphocytes from patient's blood; the base substitution was absent in fibroblasts and in the tissues from proband's healthy mother and brother. The specific skeletal muscle tissue involvement can explain the childhood-onset and the relatively benign, exclusively myopathic course of the disease.

14.
Muscle Nerve ; 55(1): 55-68, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27184587

RÉSUMÉ

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.


Sujet(s)
Diagnostic différentiel , Dystrophies musculaires des ceintures/diagnostic , Dystrophies musculaires des ceintures/épidémiologie , Adolescent , Adulte , Âge de début , Sujet âgé , Creatine kinase/sang , Femelle , Études d'associations génétiques , Humains , Italie/épidémiologie , Mâle , Adulte d'âge moyen , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Dystrophies musculaires des ceintures/complications , Dystrophies musculaires des ceintures/génétique , Enregistrements , Troubles respiratoires/étiologie , Statistique non paramétrique , Jeune adulte
15.
J Appl Physiol (1985) ; 121(3): 699-708, 2016 09 01.
Article de Anglais | MEDLINE | ID: mdl-27445303

RÉSUMÉ

Aerobic training can be effective in patients with mitochondrial myopathies (MM) and McArdle's disease (McA). The aim of the study was to use noninvasive functional evaluation methods, specifically aimed at skeletal muscle oxidative metabolism, to evaluate the effects of an aerobic exercise training (cycle ergometer, 12 wk, 4 days/wk, ∼65-70% of maximal heart rate) in 6 MM and 7 McA. Oxygen uptake and skeletal muscle vastus lateralis fractional O2 extraction by near-infrared spectroscopy were assessed during incremental and low-intensity constant work rate (CWR) exercises before (BEFORE) and at the end (AFTER) of training. Peak O2 uptake increased significantly with training both in MM [14.7 ± 1.2 vs. 17.6 ± 1.4 ml·kg(-1)·min(-1) (mean ± SD)] and in McA (18.5 ± 1.8 ml·kg(-1)·min(-1) vs. 21.6 ± 1.9). Peak skeletal muscle fractional O2 extraction increased with training both in MM (22.0 ± 6.7 vs. 32.6 ± 5.9%) and in McA (18.5 ± 6.2 vs. 37.2 ± 7.2%). During low-intensity CWR in both MM and McA: V̇o2 kinetics became faster in AFTER, but only in the patients with slow V̇o2 kinetics in BEFORE; the transient overshoot in fractional O2 extraction kinetics disappeared. The level of habitual physical activity was not higher 3 mo after training (FOLLOW-UP vs. PRE). In MM and McA patients a home-based aerobic training program significantly attenuated the impairment of skeletal muscle oxidative metabolism and improved variables associated with exercise tolerance. Our findings indicate that in MM and McA patients near-infrared spectroscopy and V̇o2 kinetics can effectively detect the functional improvements obtained by training.


Sujet(s)
Traitement par les exercices physiques/méthodes , Tolérance à l'effort , Glycogénose de type V/physiopathologie , Glycogénose de type V/thérapie , Myopathies mitochondriales/physiopathologie , Myopathies mitochondriales/thérapie , Consommation d'oxygène , Adulte , Femelle , Services de soins à domicile , Humains , Mâle , Adulte d'âge moyen , Oxydoréduction , Oxygène/métabolisme , Résultat thérapeutique
16.
J Neurol ; 263(6): 1204-14, 2016 Jun.
Article de Anglais | MEDLINE | ID: mdl-27126453

RÉSUMÉ

Based on the 7-year experience of the Italian Clinical Network for FSHD, we revised the FSHD clinical form to describe, in a harmonized manner, the phenotypic spectrum observed in FSHD. The new Comprehensive Clinical Evaluation Form (CCEF) defines various clinical categories by the combination of different features. The inter-rater reproducibility of the CCEF was assessed between two examiners using kappa statistics by evaluating 56 subjects carrying the molecular marker used for FSHD diagnosis. The CCEF classifies: (1) subjects presenting facial and scapular girdle muscle weakness typical of FSHD (category A, subcategories A1-A3), (2) subjects with muscle weakness limited to scapular girdle or facial muscles (category B subcategories B1, B2), (3) asymptomatic/healthy subjects (category C, subcategories C1, C2), (4) subjects with myopathic phenotype presenting clinical features not consistent with FSHD canonical phenotype (D, subcategories D1, D2). The inter-rater reliability study showed an excellent concordance of the final four CCEF categories with a κ equal to 0.90; 95 % CI (0.71; 0.97). Absolute agreement was observed for categories C and D, an excellent agreement for categories A [κ = 0.88; 95 % CI (0.75; 1.00)], and a good agreement for categories B [κ = 0.79; 95 % CI (0.57; 1.00)]. The CCEF supports the harmonized phenotypic classification of patients and families. The categories outlined by the CCEF may assist diagnosis, genetic counseling and natural history studies. Furthermore, the CCEF categories could support selection of patients in randomized clinical trials. This precise categorization might also promote the search of genetic factor(s) contributing to the phenotypic spectrum of disease.


Sujet(s)
Dystrophie musculaire facio-scapulo-humérale/classification , Dystrophie musculaire facio-scapulo-humérale/diagnostic , Adulte , Âge de début , Sujet âgé , Famille , Femelle , Prédisposition génétique à une maladie , Humains , Italie , Mâle , Adulte d'âge moyen , Activité motrice , Force musculaire , Dystrophie musculaire facio-scapulo-humérale/anatomopathologie , Dystrophie musculaire facio-scapulo-humérale/physiopathologie , Examen neurologique , Biais de l'observateur , Phénotype , Enregistrements
17.
Muscle Nerve ; 54(5): 843-849, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-26970407

RÉSUMÉ

INTRODUCTION: The aim of this study was to assess the perceived effect of salbutamol in adult patients with spinal muscular atrophy and to evaluate the usefulness of the World Health Organization Disability Assessment Schedule II (WHODAS II) and Fatigue Severity Scale (FSS) for its measurement. METHODS: A longitudinal mixed methods study was performed. Ten patients were interviewed and completed WHODAS II and FSS questionnaires to assess disability and fatigue at 2 time-points. Inductive thematic analysis was used for qualitative data. The non-parametric Wilcoxon test was performed for quantitative analysis. RESULTS: All participants reported an improvement in their condition after salbutamol consumption. WHODAS II and FSS reliably captured changes in patients' disability and fatigue. CONCLUSIONS: The mixed methods design allowed us to identify the functional domains in which participants experienced effects of salbutamol. Patients were satisfied with the treatment as shown by decreased fatigue, improved functioning, and infrequent side effects. Muscle Nerve, 2016 Muscle Nerve 54: 843-849, 2016.


Sujet(s)
Salbutamol/usage thérapeutique , Bronchodilatateurs/usage thérapeutique , Amyotrophie spinale/traitement médicamenteux , Adulte , Femelle , Humains , Études longitudinales , Mâle , Adulte d'âge moyen , Indice de gravité de la maladie , Statistique non paramétrique , Enquêtes et questionnaires , Résultat thérapeutique
18.
Neuromuscul Disord ; 26(3): 189-96, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26916554

RÉSUMÉ

In Duchenne muscular dystrophy (DMD) little has been reported on the association between clinical outcome measures and patient health-related quality of life (HRQOL) tools. Our study evaluated the relationship between 12 month changes on the Generic Core Scales (GCS), the Multidimensional Fatigue Scale and the Neuromuscular Module of the PedsQL(TM) with several outcome measures (6 minute walk test, North Star Ambulatory Assessment and timed items) in ambulatory DMD. Ninety-eight ambulatory DMD in a multicentric setting were included in the study. At baseline, the PedsQL(TM) inventories correlated with almost all the functional measures On the Child Self-Report there was a significant decrease between baseline and 12 months on the PedsQL(TM) GCS and its first domain, in parallel with the decrement in the functional outcome measures. Correlation between the 12 month changes on the PedsQL(TM) inventories and functional measures were almost all negligible. Similar results were obtained on the Parent Proxy-Report. In conclusion, PedsQL(TM) correlates with the level of impairment at baseline, but this does not hold true when 12 month changes are considered. Further studies comparing different tools are needed to better elucidate the complexity of the relationship between HRQOL and functional performances.


Sujet(s)
Myopathie de Duchenne/diagnostic , Myopathie de Duchenne/psychologie , Qualité de vie , Adolescent , Enfant , Enfant d'âge préscolaire , Fatigue/diagnostic , Fatigue/étiologie , Humains , Études longitudinales , Mâle , Myopathie de Duchenne/complications , Études prospectives , Indice de gravité de la maladie , Enquêtes et questionnaires
19.
BMJ Open ; 6(1): e007798, 2016 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-26733561

RÉSUMÉ

OBJECTIVES: Facioscapulohumeral muscular dystrophy type 1 (FSHD1) has been genetically linked to reduced numbers (≤ 8) of D4Z4 repeats at 4q35. Particularly severe FSHD cases, characterised by an infantile onset and presence of additional extra-muscular features, have been associated with the shortest D4Z4 reduced alleles with 1-3 repeats (1-3 DRA). We searched for signs of perinatal onset and evaluated disease outcome through the systematic collection of clinical and anamnestic records of de novo and familial index cases and their relatives, carrying 1-3 DRA. SETTING: Italy. PARTICIPANTS: 66 index cases and 33 relatives carrying 1-3 DRA. OUTCOMES: The clinical examination was performed using the standardised FSHD evaluation form with validated inter-rater reliability. To investigate the earliest signs of disease, we designed the Infantile Anamnestic Questionnaire (IAQ). Comparison of age at onset was performed using the non-parametric Wilcoxon rank-sum or Kruskal-Wallis test. Comparison of the FSHD score was performed using a general linear model and Wald test. Kaplan-Meier survival analysis was used to estimate the age-specific cumulative motor impairment risk. RESULTS: No patients had perinatal onset. Among index cases, 36 (54.5%) showed the first signs by 10 years of age. The large majority of patients with early disease onset (26 out of 36, 72.2%) were de novo; whereas the majority of patients with disease onset after 10 years of age were familial (16, 53.3%). Comparison of the disease severity outcome between index cases with age at onset before and over 10 years of age, failed to detect statistical significance (Wald test p value=0.064). Of 61 index cases, only 17 (27.9%) presented extra-muscular conditions. Relatives carrying 1-3 DRA showed a large clinical variability ranging from healthy subjects, to patients with severe motor impairment. CONCLUSIONS: The size of the D4Z4 allele is not always predictive of severe clinical outcome. The high degree of clinical variability suggests that additional factors contribute to the phenotype complexity.


Sujet(s)
Allèles , Génotype , Dystrophie musculaire facio-scapulo-humérale/génétique , Protéines nucléaires/génétique , Phénotype , Adolescent , Adulte , Âge de début , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Nouveau-né , Italie , Estimation de Kaplan-Meier , Mâle , Protéines des microfilaments , Adulte d'âge moyen , Protéines de liaison à l'ARN , Enregistrements , Indice de gravité de la maladie , Jeune adulte
20.
Eur J Pediatr ; 175(8): 1113-8, 2016 Aug.
Article de Anglais | MEDLINE | ID: mdl-26780752

RÉSUMÉ

UNLABELLED: Mutations in SEPN1 cause selenoprotein N (SEPN)-related myopathy (SEPN-RM) characterized by early-onset axial and neck weakness, spinal rigidity, respiratory failure and histopathological features, ranging from mild dystrophic signs to a congenital myopathy pattern with myofibrillar disorganization. We report on clinical and instrumental features in three patients affected with a congenital myopathy characterized by prevalent neck weakness starting at different ages and mild myopathy, in whom we performed diagnosis of SEPN-RM. The patients presented myopathic signs since their first years of life, but the disease remained unrecognized because of a relatively benign myopathic course. In two cases, myopathic features were stable after 2 years of follow-up, but respiratory involvement worsened. The muscle MRI and muscle biopsy showed a typical pattern of SEPN-RM. Molecular diagnosis revealed two novel homozygous mutations in SEPN1, c.1176delA and c.726_727InsTCC. CONCLUSION: This report underlines the clinical diagnostic clues of early neck and axial weakness to suspect a SEPN-RM and the usefulness of muscle MRI in conjunction with clinical features to achieve the diagnosis. Our data confirm the slow progression of respiratory involvement in spite of the relatively stable course of myopathy. We report two previously undescribed mutations in SEPN1. WHAT IS KNOWN: • Mutations in SEPN1 cause myopathy characterized by early-onset axial and neck weakness spinal rigidity and respiratory failure. • SEPN-related myopathies have been initially associated with four distinct histopathological entities that however appear more mixed in recently described cases. What is New: • SEPN-related myopathies can remain unrecognized because of the normal early motor development and relatively benign myopathic course of the disease. • Our study adds two novel homozygous mutations to the number of reported pathogenic SEPN1 variants.


Sujet(s)
Protéines du muscle/génétique , Mutation , Myopathies congénitales structurales/diagnostic , Muscles du cou/malformations , Sélénoprotéines/génétique , Adolescent , Encéphale/imagerie diagnostique , Enfant , Enfant d'âge préscolaire , Électrocardiographie , Femelle , Humains , Imagerie par résonance magnétique , Mâle , Insuffisance respiratoire/étiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...