Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur Heart J ; 42(20): 1976-1984, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-33748842

RESUMO

AIMS: To estimate the effect of prophylactic angiotensin-converting enzyme inhibitors (ACEi) on survival in Duchenne muscular dystrophy (DMD). METHODS AND RESULTS: We analysed the data from the French multicentre DMD Heart Registry (ClinicalTrials.gov: NCT03443115). We estimated the association between the prophylactic prescription of ACEi and event-free survival in 668 patients aged 8 to 13 years, with normal left ventricular function, using (i) a Cox model with intervention as a time-dependent covariate, (ii) a propensity-based analysis comparing ACEi treatment vs. no treatment, and (iii) a set of sensitivity analyses. The study outcomes were overall survival and hospitalizations for heart failure (HF) or acute respiratory failure. Among the 668 patients included in the DMD Heart Registry, 576 (mean age 6.1 ± 2.8 years) were eligible for this study, of whom 390 were treated with ACEi prophylactically. Death occurred in 53 patients (13.5%) who were and 60 patients (32.3%) who were not treated prophylactically with ACEi, respectively. In a Cox model with intervention as a time-dependent variable, the hazard ratio (HR) associated with ACEi treatment was 0.49 [95% confidence interval (CI) 0.34-0.72] and 0.47 (95% CI 0.31-0.17) for overall mortality after adjustment for baseline variables. In the propensity-based analysis, 278 patients were included in the treatment group and 834 in the control group, with 18.5% and 30.4% 12-year estimated probability of death, respectively. ACEi were associated with a lower risk of death (HR 0.39; 95% CI 0.17-0.92) and hospitalization for HF (HR 0.16; 95% CI 0.04-0.62). All other sensitivity analyses yielded similar results. CONCLUSION: Prophylactic ACEi treatment in DMD was associated with a significantly higher overall survival and lower rates of hospitalization for HF.


Assuntos
Insuficiência Cardíaca , Distrofia Muscular de Duchenne , Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Criança , Pré-Escolar , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/prevenção & controle , Humanos , Distrofia Muscular de Duchenne/tratamento farmacológico , Sistema de Registros , Resultado do Tratamento , Função Ventricular Esquerda
2.
Am J Hum Genet ; 99(3): 753-761, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27569547

RESUMO

The neuromuscular junction (NMJ) is one of the best-studied cholinergic synapses. Inherited defects of peripheral neurotransmission result in congenital myasthenic syndromes (CMSs), a clinically and genetically heterogeneous group of rare diseases with fluctuating fatigable muscle weakness as the clinical hallmark. Whole-exome sequencing and Sanger sequencing in six unrelated families identified compound heterozygous and homozygous mutations in SLC5A7 encoding the presynaptic sodium-dependent high-affinity choline transporter 1 (CHT), which is known to be mutated in one dominant form of distal motor neuronopathy (DHMN7A). We identified 11 recessive mutations in SLC5A7 that were associated with a spectrum of severe muscle weakness ranging from a lethal antenatal form of arthrogryposis and severe hypotonia to a neonatal form of CMS with episodic apnea and a favorable prognosis when well managed at the clinical level. As expected given the critical role of CHT for multisystemic cholinergic neurotransmission, autonomic dysfunctions were reported in the antenatal form and cognitive impairment was noticed in half of the persons with the neonatal form. The missense mutations induced a near complete loss of function of CHT activity in cell models. At the human NMJ, a delay in synaptic maturation and an altered maintenance were observed in the antenatal and neonatal forms, respectively. Increased synaptic expression of butyrylcholinesterase was also observed, exposing the dysfunction of cholinergic metabolism when CHT is deficient in vivo. This work broadens the clinical spectrum of human diseases resulting from reduced CHT activity and highlights the complexity of cholinergic metabolism at the synapse.


Assuntos
Apneia/genética , Mutação/genética , Miastenia Gravis/genética , Terminações Pré-Sinápticas/metabolismo , Simportadores/genética , Simportadores/metabolismo , Adolescente , Apneia/complicações , Apneia/metabolismo , Apneia/patologia , Artrogripose/complicações , Artrogripose/genética , Butirilcolinesterase/metabolismo , Criança , Pré-Escolar , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Análise Mutacional de DNA , Exoma/genética , Feminino , Genes Recessivos/genética , Células HEK293 , Heterozigoto , Homozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Hipotonia Muscular/genética , Debilidade Muscular/complicações , Debilidade Muscular/genética , Debilidade Muscular/patologia , Mutação de Sentido Incorreto/genética , Miastenia Gravis/complicações , Miastenia Gravis/metabolismo , Miastenia Gravis/patologia , Junção Neuromuscular/enzimologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Terminações Pré-Sinápticas/patologia , Simportadores/deficiência , Transmissão Sináptica
3.
Hum Genet ; 137(9): 753-768, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30167850

RESUMO

NALCN is a conserved cation channel, which conducts a permanent sodium leak current and regulates resting membrane potential and neuronal excitability. It is part of a large ion channel complex, the "NALCN channelosome", consisting of multiple proteins including UNC80 and UNC79. The predominant neuronal expression pattern and its function suggest an important role in neuronal function and disease. So far, biallelic NALCN and UNC80 variants have been described in a small number of individuals leading to infantile hypotonia, psychomotor retardation, and characteristic facies 1 (IHPRF1, OMIM 615419) and 2 (IHPRF2, OMIM 616801), respectively. Heterozygous de novo NALCN missense variants in the S5/S6 pore-forming segments lead to congenital contractures of the limbs and face, hypotonia, and developmental delay (CLIFAHDD, OMIM 616266) with some clinical overlap. In this study, we present detailed clinical information of 16 novel individuals with biallelic NALCN variants, 1 individual with a heterozygous de novo NALCN missense variant and an interesting clinical phenotype without contractures, and 12 individuals with biallelic UNC80 variants. We report for the first time a missense NALCN variant located in the predicted S6 pore-forming unit inherited in an autosomal-recessive manner leading to mild IHPRF1. We show evidence of clinical variability, especially among IHPRF1-affected individuals, and discuss differences between the IHPRF1- and IHPRF2 phenotypes. In summary, we provide a comprehensive overview of IHPRF1 and IHPRF2 phenotypes based on the largest cohort of individuals reported so far and provide additional insights into the clinical phenotypes of these neurodevelopmental diseases to help improve counseling of affected families.


Assuntos
Proteínas de Transporte/genética , Canalopatias/genética , Deficiências do Desenvolvimento/genética , Marcadores Genéticos , Variação Genética , Proteínas de Membrana/genética , Canais de Sódio/genética , Adolescente , Adulto , Canalopatias/patologia , Criança , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Feminino , Humanos , Lactente , Recém-Nascido , Canais Iônicos , Masculino , Fenótipo , Adulto Jovem
4.
J Cell Sci ; 127(Pt 13): 2873-84, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24806962

RESUMO

The mechanisms underlying the cell response to mechanical forces are crucial for muscle development and functionality. We aim to determine whether mutations of the LMNA gene (which encodes lamin A/C) causing congenital muscular dystrophy impair the ability of muscle precursors to sense tissue stiffness and to respond to mechanical challenge. We found that LMNA-mutated myoblasts embedded in soft matrix did not align along the gel axis, whereas control myoblasts did. LMNA-mutated myoblasts were unable to tune their cytoskeletal tension to the tissue stiffness as attested by inappropriate cell-matrix adhesion sites and cytoskeletal tension in soft versus rigid substrates or after mechanical challenge. Importantly, in soft two-dimensional (2D) and/or static three-dimensional (3D) conditions, LMNA-mutated myoblasts showed enhanced activation of the yes-associated protein (YAP) signaling pathway that was paradoxically reduced after cyclic stretch. siRNA-mediated downregulation of YAP reduced adhesion and actin stress fibers in LMNA myoblasts. This is the first demonstration that human myoblasts with LMNA mutations have mechanosensing defects through a YAP-dependent pathway. In addition, our data emphasize the crucial role of biophysical attributes of cellular microenvironment to the response of mechanosensing pathways in LMNA-mutated myoblasts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Lamina Tipo A/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Microambiente Celular/fisiologia , Humanos , Lamina Tipo A/genética , Microscopia Confocal , Mutação , Fosfoproteínas/genética , Transdução de Sinais , Fatores de Transcrição , Proteínas de Sinalização YAP
5.
Hum Mutat ; 35(7): 779-90, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24692096

RESUMO

Mutations affecting skeletal muscle isoforms of the tropomyosin genes may cause nemaline myopathy, cap myopathy, core-rod myopathy, congenital fiber-type disproportion, distal arthrogryposes, and Escobar syndrome. We correlate the clinical picture of these diseases with novel (19) and previously reported (31) mutations of the TPM2 and TPM3 genes. Included are altogether 93 families: 53 with TPM2 mutations and 40 with TPM3 mutations. Thirty distinct pathogenic variants of TPM2 and 20 of TPM3 have been published or listed in the Leiden Open Variant Database (http://www.dmd.nl/). Most are heterozygous changes associated with autosomal-dominant disease. Patients with TPM2 mutations tended to present with milder symptoms than those with TPM3 mutations, DA being present only in the TPM2 group. Previous studies have shown that five of the mutations in TPM2 and one in TPM3 cause increased Ca(2+) sensitivity resulting in a hypercontractile molecular phenotype. Patients with hypercontractile phenotype more often had contractures of the limb joints (18/19) and jaw (6/19) than those with nonhypercontractile ones (2/22 and 1/22), whereas patients with the non-hypercontractile molecular phenotype more often (19/22) had axial contractures than the hypercontractile group (7/19). Our in silico predictions show that most mutations affect tropomyosin-actin association or tropomyosin head-to-tail binding.


Assuntos
Estudos de Associação Genética , Doenças Musculares/congênito , Doenças Musculares/genética , Mutação , Tropomiosina/genética , Actinas/metabolismo , Adolescente , Adulto , Sequência de Aminoácidos , Criança , Pré-Escolar , Bases de Dados Genéticas , Feminino , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Doenças Musculares/diagnóstico , Fenótipo , Fosforilação , Ligação Proteica , Alinhamento de Sequência , Tropomiosina/química , Tropomiosina/metabolismo , Adulto Jovem
6.
Eur Respir J ; 39(5): 1206-12, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22135279

RESUMO

The aim of the study was to identify daytime predictors of nocturnal gas exchange anomalies in children with neuromuscular disease (NMD) and normal daytime gas exchange. Lung function tests, respiratory muscle evaluation and nocturnal gas exchange were obtained as part of routine evaluation. We included 52 consecutive children with Duchenne muscular dystrophy (n = 20), spinal muscular atrophy (n = 10) and other NMD (n = 22). 20 patients had nocturnal hypoxaemia, defined as minimal arterial oxygen saturation measured by pulse oximetry (S(p,O(2))) <90% for ≥ 2% of night time, and 22 had nocturnal hypercapnia, defined as maximal transcutaneous carbon dioxide tension (P(tc,CO(2))) >50 mmHg for ≥ 2% of night time. Forced vital capacity and helium functional residual capacity correlated with minimal nocturnal S(p,O(2)) (p = 0.009 and p = 0.01, respectively). Daytime pH correlated negatively with maximal nocturnal P(tc,CO(2)) (p=0.005) and daytime arterial carbon dioxide tension (P(a,CO(2))) correlated with the percentage of time with a P(tc,CO(2)) >50 mmHg (p = 0.02). Sniff nasal inspiratory pressure correlated with minimal nocturnal S(p,O(2)) (p = 0.02). Daytime P(a,CO(2)) was a weak predictor of nocturnal hypercapnia (sensitivity 80%; specificity 57%). Daytime lung function and respiratory muscle parameters correlate poorly with nocturnal hypoxaemia and hypercapnia in children with NMD and normal daytime gas exchange, which necessitates more systematic sleep studies in these children.


Assuntos
Ritmo Circadiano/fisiologia , Hipercapnia/fisiopatologia , Hipóxia/fisiopatologia , Doenças Neuromusculares/fisiopatologia , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Oximetria , Oxigênio/sangue , Polissonografia , Troca Gasosa Pulmonar/fisiologia , Testes de Função Respiratória , Músculos Respiratórios/fisiopatologia , Adulto Jovem
7.
Hum Mol Genet ; 18(20): 3779-94, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19602481

RESUMO

The presence of variable degrees of cognitive impairment, extending from severe mental retardation to specific deficits, in patients with dystrophinopathies is a well-recognized problem. However, molecular basis underlying mental retardation and its severity remain poorly understood and still a matter of debate. Here, we report one of the largest study based on the comparison of clinical, cognitive, molecular and expression data in a large cohort of 81 patients affected with Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) bearing mutations predicted to affect either all dystrophin products, including Dp71 or all dystrophin products, except Dp71. In addition to the consistent data defining molecular basis underlying mental retardation in DMD, we show that BMD patients with MR have mutations that significantly affect Dp71 expression or with mutations located in exons 75 and 76. We also show that mutations upstream to exon 62, with DMD phenotype, predicted to lead to a loss-of-function of all dystrophin products, except Dp71 isoform, are associated, predominantly, with normal or borderline cognitive performances. Altogether, these reliable phenotype-genotype correlations in combination with Dp71 mRNA and protein expression studies, strongly indicate that loss-of-function of all dystrophin products is systematically associated with severe form of MR, and Dp71 deficit is a factor that contributes in the severity of MR and may account for a shift of 2 SD downward of the intelligence quotient.


Assuntos
Distrofina/genética , Expressão Gênica , Deficiência Intelectual/genética , Distrofia Muscular de Duchenne/complicações , Mutação , Adolescente , Adulto , Sequência de Bases , Criança , Cognição , Estudos de Coortes , Distrofina/metabolismo , Feminino , Humanos , Deficiência Intelectual/metabolismo , Deficiência Intelectual/psicologia , Testes de Inteligência , Masculino , Dados de Sequência Molecular , Índice de Gravidade de Doença , Adulto Jovem
8.
Ann Neurol ; 68(4): 511-20, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20976770

RESUMO

OBJECTIVE: Mutations in the genes encoding the extracellular matrix protein collagen VI (ColVI) cause a spectrum of disorders with variable inheritance including Ullrich congenital muscular dystrophy, Bethlem myopathy, and intermediate phenotypes. We extensively characterized, at the clinical, cellular, and molecular levels, 49 patients with onset in the first 2 years of life to investigate genotype-phenotype correlations. METHODS: Patients were classified into 3 groups: early-severe (18%), moderate-progressive (53%), and mild (29%). ColVI secretion was analyzed in patient-derived skin fibroblasts. Chain-specific transcript levels were quantified by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), and mutation identification was performed by sequencing of complementary DNA. RESULTS: ColVI secretion was altered in all fibroblast cultures studied. We identified 56 mutations, mostly novel and private. Dominant de novo mutations were detected in 61% of the cases. Importantly, mutations causing premature termination codons (PTCs) or in-frame insertions strikingly destabilized the corresponding transcripts. Homozygous PTC-causing mutations in the triple helix domains led to the most severe phenotypes (ambulation never achieved), whereas dominant de novo in-frame exon skipping and glycine missense mutations were identified in patients of the moderate-progressive group (loss of ambulation). INTERPRETATION: This work emphasizes that the diagnosis of early onset ColVI myopathies is arduous and time-consuming, and demonstrates that quantitative RT-PCR is a helpful tool for the identification of some mutation-bearing genes. Moreover, the clinical classification proposed allowed genotype-phenotype relationships to be explored, and may be useful in the design of future clinical trials.


Assuntos
Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Doenças Musculares , Mutação/genética , Estatística como Assunto , Adolescente , Adulto , Células Cultivadas , Criança , Pré-Escolar , Europa (Continente) , Feminino , Fibroblastos/metabolismo , Testes Genéticos/métodos , Glicina/genética , Humanos , Masculino , Músculo Esquelético/metabolismo , Doenças Musculares/genética , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Fenótipo , Adulto Jovem
9.
Br J Nutr ; 105(10): 1486-91, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21272404

RESUMO

The life expectancy of patients with Duchenne muscular dystrophy (DMD) has increased. A cross-sectional study of DMD patients showed that 54 % of 13-year-old patients are obese and that 54 % of 18-year-old patients are underweight. We aimed to describe the natural evolution of weight status in DMD. This retrospective multi-centre audit collected body-weight measurements for seventy DMD patients born before 1992. The body-weight:age ratio (W:A) was used to evaluate weight status in reference to the Griffiths and Edwards chart. At the age of 13 years, 73 % were obese and 4 % were underweight. At maximal follow-up (age 15-26 years, mean 18·3 (sd 2·3) years), 47 % were obese and 34 % were underweight. Obesity at the age of 13 years was associated with later obesity, whereas normal weight status and underweight in 13-year-old patients predicted later underweight. A W:A ≥ 151 % in 13-year-old patients predicted later obesity, and a W:A ≤ 126·5 % predicted later underweight. Our audit provides the first longitudinal information about the spontaneous outcome of weight status in DMD. Patients (13 years old) with a W:A ≥ 151 % were more likely to become obese in late adolescence, but obesity prevented later underweight. These data suggest that mild obesity in 13-year-old DMD patients (W:A between 120 and 150 %) should not be discouraged because it prevents later underweight.


Assuntos
Peso Corporal , Auditoria Médica , Distrofia Muscular de Duchenne/fisiopatologia , Adolescente , Adulto , Criança , Pré-Escolar , Estudos Transversais , Seguimentos , Humanos , Desnutrição/complicações , Desnutrição/fisiopatologia , Distrofia Muscular de Duchenne/complicações , Obesidade/complicações , Obesidade/fisiopatologia , Estudos Retrospectivos , Adulto Jovem
10.
Brain Commun ; 3(3): fcab075, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34240052

RESUMO

Muscular dystrophies due to heterozygous pathogenic variants in LMNA gene cover a broad spectrum of clinical presentations and severity with an age of onset ranging from the neonatal period to adulthood. The natural history of these conditions is not well defined, particularly in patients with congenital or early onset who arguably present with the highest disease burden. Thus the definition of natural history endpoints along with clinically revelant outcome measures is essential to establishing both clinical care planning and clinical trial readiness for this patient group. We designed a large international cross-sectional retrospective natural history study of patients with genetically proven muscle laminopathy who presented with symptoms before two years of age intending to identify and characterize an optimal clinical trial cohort with pertinent motor, cardiac and respiratory endpoints. Quantitative statistics were used to evaluate associations between LMNA variants and distinct clinical events. The study included 151 patients (median age at symptom onset 0.9 years, range: 0.0-2.0). Age of onset and age of death were significantly lower in patients who never acquired independent ambulation compared to patients who achieved independent ambulation. Most of the patients acquired independent ambulation (n = 101, 66.9%), and subsequently lost this ability (n = 86; 85%). The age of ambulation acquisition (median: 1.2 years, range: 0.8-4.0) and age of ambulation loss (median: 7 years, range: 1.2-38.0) were significantly associated with the age of the first respiratory interventions and the first cardiac symptoms. Respiratory and gastrointestinal interventions occurred during first decade while cardiac interventions occurred later. Genotype-phenotype analysis showed that the most common mutation, p.Arg249Trp (20%), was significantly associated with a more severe disease course. This retrospective natural history study of early onset LMNA-related muscular dystrophy confirms the progressive nature of the disorder, initially involving motor symptoms prior to onset of other symptoms (respiratory, orthopaedic, cardiac and gastrointestinal). The study also identifies subgroups of patients with a range of long-term outcomes. Ambulatory status was an important mean of stratification along with the presence or absence of the p.Arg249Trp mutation. These categorizations will be important for future clinical trial cohorts. Finally, this study furthers our understanding of the progression of early onset LMNA-related muscular dystrophy and provides important insights into the anticipatory care needs of LMNA-related respiratory and cardiac manifestations.

11.
Neurol Genet ; 6(6): e520, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33134516

RESUMO

OBJECTIVE: To delineate the phenotype associated with biallelic ATAD1 variants. METHODS: We describe 2 new patients with ATAD1-related disorder diagnosed by whole-exome sequencing and compare their phenotype to 6 previous patients. RESULTS: Patients 1 and 2 had a similar distinctive phenotype comprising congenital stiffness of limbs, absent spontaneous movements, weak sucking, and hypoventilation. Both had absent brainstem evoked auditory responses (BEARs). Patient 1 carried the homozygous p.(His357Argfs*15) variant in ATAD1. In the light of the finding in patient 1, a second reading of exome data for patient 2 revealed the novel homozygous p.(Gly128Val) variant. CONCLUSIONS: Analysis of the phenotypes of these 2 patients and of the 6 previous cases showed that biallelic ATAD1 mutations are responsible for a unique congenital encephalopathy likely comprising absent BEAR, different from hyperekplexia and other conditions with neonatal hypertonia.

12.
Neurol Genet ; 6(6): e534, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33659639

RESUMO

OBJECTIVE: To report the identification of 2 new homozygous recessive mutations in the synaptotagmin 2 (SYT2) gene as the genetic cause of severe and early presynaptic forms of congenital myasthenic syndromes (CMSs). METHODS: Next-generation sequencing identified new homozygous intronic and frameshift mutations in the SYT2 gene as a likely cause of presynaptic CMS. We describe the clinical and electromyographic patient phenotypes, perform ex vivo splicing analyses to characterize the effect of the intronic mutation on exon splicing, and analyze the functional impact of this variation at the neuromuscular junction (NMJ). RESULTS: The 2 infants presented a similar clinical phenotype evoking first a congenital myopathy characterized by muscle weakness and hypotonia. Next-generation sequencing allowed to the identification of 1 homozygous intronic mutation c.465+1G>A in patient 1 and another homozygous frameshift mutation c.328_331dup in patient 2, located respectively in the 5' splice donor site of SYT2 intron 4 and in exon 3. Functional studies of the intronic mutation validated the abolition of the splice donor site of exon 4 leading to its skipping. In-frame skipping of exon 4 that encodes part of the C2A calcium-binding domain of SYT2 is associated with a loss-of-function effect resulting in a decrease of neurotransmitter release and severe pre- and postsynaptic NMJ defects. CONCLUSIONS: This study identifies new homozygous recessive SYT2 mutations as the underlying cause of severe and early presynaptic form of CMS expanding the genetic spectrum of recessive SYT2-related CMS associated with defects in neurotransmitter release.

13.
Front Pediatr ; 8: 4, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32133329

RESUMO

Spinal muscular atrophy type 1 (SMA-1) is a severe neurodegenerative disorder, which in the absence of curative treatment, leads to death before 1 year of age in most cases. Caring for these short-lived and severely impaired infants requires palliative management. New drugs (nusinersen) have recently been developed that may modify SMA-1 natural history and thus raise ethical concerns about the appropriate level of care for patients. The national Hospital Clinical Research Program (PHRC) called "Assessment of clinical practices of palliative care in children with Spinal Muscular Atrophy Type 1 (SMA-1)" was a multicenter prospective study conducted in France between 2012 and 2016 to report palliative practices in SMA-1 in real life through prospective caregivers' reports about their infants' management. Thirty-nine patients were included in the prospective PHRC (17 centers). We also studied retrospective data regarding management of 43 other SMA-1 patients (18 centers) over the same period, including seven treated with nusinersen, in comparison with historical data from 222 patients previously published over two periods of 10 years (1989-2009). In the latest period studied, median age at diagnosis was 3 months [0.6-10.4]. Seventy-seven patients died at a median 6 months of age[1-27]: 32% at home and 8% in an intensive care unit. Eighty-five percent of patients received enteral nutrition, some through a gastrostomy (6%). Sixteen percent had a non-invasive ventilation (NIV). Seventy-seven percent received sedative treatment at the time of death. Over time, palliative management occurred more frequently at home with increased levels of technical supportive care (enteral nutrition, oxygenotherapy, and analgesic and sedative treatments). No statistical difference was found between the prospective and retrospective patients for the last period. However, significant differences were found between patients treated with nusinersen vs. those untreated. Our data confirm that palliative care is essential in management of SMA-1 patients and that parents are extensively involved in everyday patient care. Our data suggest that nusinersen treatment was accompanied by significantly more invasive supportive care, indicating that a re-examination of standard clinical practices should explicitly consider what treatment pathways are in infants' and caregivers' best interest. This study was registered on clinicaltrials.gov under the reference NCT01862042 (https://clinicaltrials.gov/ct2/show/study/NCT01862042?cond=SMA1&rank=8).

14.
Neurology ; 95(11): e1512-e1527, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32796131

RESUMO

OBJECTIVE: To clarify the prevalence, long-term natural history, and severity determinants of SEPN1-related myopathy (SEPN1-RM), we analyzed a large international case series. METHODS: Retrospective clinical, histologic, and genetic analysis of 132 pediatric and adult patients (2-58 years) followed up for several decades. RESULTS: The clinical phenotype was marked by severe axial muscle weakness, spinal rigidity, and scoliosis (86.1%, from 8.9 ± 4 years), with relatively preserved limb strength and previously unreported ophthalmoparesis in severe cases. All patients developed respiratory failure (from 10.1±6 years), 81.7% requiring ventilation while ambulant. Histopathologically, 79 muscle biopsies showed large variability, partly determined by site of biopsy and age. Multi-minicores were the most common lesion (59.5%), often associated with mild dystrophic features and occasionally with eosinophilic inclusions. Identification of 65 SEPN1 mutations, including 32 novel ones and the first pathogenic copy number variation, unveiled exon 1 as the main mutational hotspot and revealed the first genotype-phenotype correlations, bi-allelic null mutations being significantly associated with disease severity (p = 0.017). SEPN1-RM was more severe and progressive than previously thought, leading to loss of ambulation in 10% of cases, systematic functional decline from the end of the third decade, and reduced lifespan even in mild cases. The main prognosis determinants were scoliosis/respiratory management, SEPN1 mutations, and body mass abnormalities, which correlated with disease severity. We propose a set of severity criteria, provide quantitative data for outcome identification, and establish a need for age stratification. CONCLUSION: Our results inform clinical practice, improving diagnosis and management, and represent a major breakthrough for clinical trial readiness in this not so rare disease.


Assuntos
Genótipo , Proteínas Musculares/genética , Doenças Musculares/diagnóstico por imagem , Doenças Musculares/genética , Selenoproteínas/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Doenças Musculares/patologia , Estudos Retrospectivos , Adulto Jovem
15.
Neurology ; 92(16): e1852-e1867, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30902907

RESUMO

OBJECTIVES: Because X-linked myotubular myopathy (XLMTM) is a rare neuromuscular disease caused by mutations in the MTM1 gene with a large phenotypic heterogeneity, to ensure clinical trial readiness, it was mandatory to better quantify disease burden and determine best outcome measures. METHODS: We designed an international prospective and longitudinal natural history study in patients with XLMTM and assessed muscle strength and motor and respiratory functions over the first year of follow-up. The humoral immunity against adeno-associated virus serotype 8 was also monitored. RESULTS: Forty-five male patients aged 3.5 months to 56.8 years were enrolled between May 2014 and May 2017. Thirteen patients had a mild phenotype (no ventilation support), 7 had an intermediate phenotype (ventilation support less than 12 hours a day), and 25 had a severe phenotype (ventilation support 12 or more hours a day). Most strength and motor function assessments could be performed even in very weak patients. Motor Function Measure 32 total score, grip and pinch strengths, and forced vital capacity, forced expiratory volume in the first second of exhalation, and peak cough flow measures discriminated the 3 groups of patients. Disease history revealed motor milestone loss in several patients. Longitudinal data on 37 patients showed that the Motor Function Measure 32 total score significantly decreased by 2%. Of the 38 patients evaluated, anti-adeno-associated virus type 8 neutralizing activity was detected in 26% with 2 patients having an inhibitory titer >1:10. CONCLUSIONS: Our data confirm that XLMTM is slowly progressive for male survivors regardless of their phenotype and provide outcome validation and natural history data that can support clinical development in this population. CLINICALTRIALSGOV IDENTIFIER: NCT02057705.


Assuntos
Miopatias Congênitas Estruturais/epidemiologia , Adolescente , Adulto , Criança , Pré-Escolar , Progressão da Doença , Seguimentos , Humanos , Lactente , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/fisiopatologia , Miopatias Congênitas Estruturais/terapia , Fenótipo , Estudos Prospectivos , Adulto Jovem
16.
Neurology ; 92(8): e852-e865, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30659139

RESUMO

OBJECTIVE: To genotypically and phenotypically characterize a large pediatric myotonic dystrophy type 1 (DM1) cohort to provide a solid frame of data for future evidence-based health management. METHODS: Among the 2,697 patients with genetically confirmed DM1 included in the French DM-Scope registry, children were enrolled between January 2010 and February 2016 from 24 centers. Comprehensive cross-sectional analysis of most relevant qualitative and quantitative variables was performed. RESULTS: We studied 314 children (52% females, with 55% congenital, 31% infantile, 14% juvenile form). The age at inclusion was inversely correlated with the CTG repeat length. The paternal transmission rate was higher than expected, especially in the congenital form (13%). A continuum of highly prevalent neurodevelopmental alterations was observed, including cognitive slowing (83%), attention deficit (64%), written language (64%), and spoken language (63%) disorders. Five percent exhibited autism spectrum disorders. Overall, musculoskeletal impairment was mild. Despite low prevalence, cardiorespiratory impairment could be life-threatening, and frequently occurred early in the first decade (25.9%). Gastrointestinal symptoms (27%) and cataracts (7%) were more frequent than expected, while endocrine or metabolic disorders were scarce. CONCLUSIONS: The pedDM-Scope study details the main genotype and phenotype characteristics of the 3 DM1 pediatric subgroups. It highlights striking profiles that could be useful in health care management (including transition into adulthood) and health policy planning.


Assuntos
Arritmias Cardíacas/fisiopatologia , Debilidade Muscular/fisiopatologia , Distrofia Miotônica/fisiopatologia , Insuficiência Respiratória/fisiopatologia , Adolescente , Arritmias Cardíacas/epidemiologia , Arritmias Cardíacas/etiologia , Criança , Pré-Escolar , Medicina Baseada em Evidências , Feminino , Deformidades do Pé/epidemiologia , Deformidades do Pé/etiologia , França/epidemiologia , Humanos , Lactente , Recém-Nascido , Masculino , Debilidade Muscular/epidemiologia , Debilidade Muscular/etiologia , Distrofia Miotônica/complicações , Distrofia Miotônica/epidemiologia , Distrofia Miotônica/genética , Sistema de Registros , Insuficiência Respiratória/epidemiologia , Insuficiência Respiratória/etiologia , Índice de Gravidade de Doença , Expansão das Repetições de Trinucleotídeos
17.
Neuromuscul Disord ; 18(6): 443-6, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18434154

RESUMO

Anti-MuSK antibodies have been reported in about 40-50% of patients with seronegative myasthenia gravis. Curiously, this condition has never been reported in association with fetal or transient neonatal myasthenia gravis, despite a known female predominance. We report the case of a 22-year-old woman who developed seronegative, mild steroid-responsive myasthenia gravis. When aged 26, she gave birth to a baby boy with neonatal myasthenia gravis characterized by hypotonia, stridor and sucking difficulties. Intubation was required for a few weeks. Anti-MuSK antibodies were assessed and found positive in both patients. Progressive hydramnios during the last trimester, with a decrease in spontaneous fetal mobility in the last weeks, long-lasting stridor, ptosis and occasional difficulties in swallowing liquids till two years of age, despite anti-MuSK antibodies becoming negative, suggest a fetal onset. The possible pathophysiology of this disorder, based on recent findings on the expression and function of MuSK protein, is reviewed.


Assuntos
Anticorpos/efeitos adversos , Miastenia Gravis/induzido quimicamente , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Adulto , Feminino , Humanos , Recém-Nascido , Relações Mãe-Filho
18.
Ann Neurol ; 62(6): 666-70, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17932957

RESUMO

We report four heterozygous dynamin 2 (DNM2) mutations in five centronuclear myopathy patients aged 1 to 15 years. They all presented with neonatal hypotonia with weak suckling. Thereafter, their phenotype progressively improved. All patients demonstrated muscle weakness prominent in the lower limbs, and most of them also presented with facial weakness, open mouth, arched palate, ptosis, and ophthalmoparesis. Electrophysiology showed only myopathic changes, and muscle biopsies showed central nuclei and type 1 fiber hypotrophy and predominance. Our results expand the phenotypic spectrum of dynamin 2-related centronuclear myopathy from the classic mild form to the more severe neonatal phenotype.


Assuntos
Dinamina II/genética , Mutação , Miopatias Congênitas Estruturais/epidemiologia , Miopatias Congênitas Estruturais/genética , Adolescente , Idade de Início , Aleitamento Materno , Criança , Pré-Escolar , Eletrofisiologia , Músculos Faciais/fisiopatologia , Heterozigoto , Humanos , Lactente , Recém-Nascido , Perna (Membro) , Hipotonia Muscular/complicações , Hipotonia Muscular/etiologia , Hipotonia Muscular/fisiopatologia , Debilidade Muscular/etiologia , Debilidade Muscular/patologia , Debilidade Muscular/fisiopatologia , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Mutação de Sentido Incorreto , Miopatias Congênitas Estruturais/complicações , Miopatias Congênitas Estruturais/fisiopatologia , Oftalmoplegia/etiologia
20.
Orphanet J Rare Dis ; 13(1): 209, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463562

RESUMO

BACKGROUND: Carpal tunnel syndrome (CTS) is a common complication of the mucopolysaccharidoses. In severe or attenuated mucopolysaccharidoses patients, clinical symptoms of CTS usually appear at a late stage of median nerve compression. Relying on CTS symptoms is often too late and there is a risk of axonal damage and further irreversible sequelae. Electroneurography is a powerful technique to detect the initial preclinical signs of median nerve compression. In a retrospective series of 13 children with mucopolysaccharidoses (10 Hunter, one Hurler-Scheie and 2 Hurler children), we describe the electroneurography progression of CTS (43 hand evaluations) and the severity of median nerve damage. RESULTS: The average age at mucopolysaccharidoses diagnosis was 33.6 months (11-66 months). Clinical signs of CTS appeared on average 44.6 months (0-73 months) after diagnosis of mucopolysaccharidoses. Electroneurography anomalies suggestive of CTS appeared as early as the age of 3.5 years and probably preceded clinical signs of CTS. Median nerve compression was bilateral and distal, initially on the sensory pathway then becoming motor-sensory. Beyond a threshold of 14 m/sec median distal motor nerve conduction velocity (MNCVd) and index of terminal latency (MNCVd/MNCVp) of 0.27, there was true distal conduction slowdown. CONCLUSIONS: To prevent irreversible sequelae of median nerve compression, we suggest annual electroneurography testing for mucopolysaccharidoses patients starting as early as 3 years of age, including both motor and sensory nerve pathways, on median and, in reference to the ulnar nerves, bilaterally at the wrist and the elbow. Timely surgical intervention can greatly improve the overall function and quality of life of these patients.


Assuntos
Mucopolissacaridoses/diagnóstico , Síndrome do Túnel Carpal/diagnóstico , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Mucopolissacaridoses/fisiopatologia , Condução Nervosa/fisiologia , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa