Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38688704

RESUMEN

The hereditary sensory and autonomic neuropathies (HSANs) are a group of rare genetic disorders characterised by variable phenotypic expression affecting both sensory and autonomic dysfunction. Diagnosing these conditions can be a challenge as the presenting symptoms can be diverse and may overlap. This often leads to a delay in referral and diagnosis.Pain is often used by clinicians as a marker for systemic diseases. The key feature of HSAN conditions is the absence of pain perception and its consequences such as unexplained injuries. When a child presents with an unexplained injury, a diagnosis of non-accidental injuries must be considered, but rarely HSAN could be a possibility.The diagnosis of HSANs in children is both important and rare. This article aims to discuss an approach to the diagnosis and management of HSANs.

2.
Ann Neurol ; 88(2): 332-347, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32403198

RESUMEN

OBJECTIVE: A hitherto undescribed phenotype of early onset muscular dystrophy associated with sensorineural hearing loss and primary ovarian insufficiency was initially identified in 2 siblings and in subsequent patients with a similar constellation of findings. The goal of this study was to understand the genetic and molecular etiology of this condition. METHODS: We applied whole exome sequencing (WES) superimposed on shared haplotype regions to identify the initial biallelic variants in GGPS1 followed by GGPS1 Sanger sequencing or WES in 5 additional families with the same phenotype. Molecular modeling, biochemical analysis, laser membrane injury assay, and the generation of a Y259C knock-in mouse were done. RESULTS: A total of 11 patients in 6 families carrying 5 different biallelic pathogenic variants in specific domains of GGPS1 were identified. GGPS1 encodes geranylgeranyl diphosphate synthase in the mevalonate/isoprenoid pathway, which catalyzes the synthesis of geranylgeranyl pyrophosphate, the lipid precursor of geranylgeranylated proteins including small guanosine triphosphatases. In addition to proximal weakness, all but one patient presented with congenital sensorineural hearing loss, and all postpubertal females had primary ovarian insufficiency. Muscle histology was dystrophic, with ultrastructural evidence of autophagic material and large mitochondria in the most severe cases. There was delayed membrane healing after laser injury in patient-derived myogenic cells, and a knock-in mouse of one of the mutations (Y259C) resulted in prenatal lethality. INTERPRETATION: The identification of specific GGPS1 mutations defines the cause of a unique form of muscular dystrophy with hearing loss and ovarian insufficiency and points to a novel pathway for this clinical constellation. ANN NEUROL 2020;88:332-347.


Asunto(s)
Dimetilaliltranstransferasa/genética , Farnesiltransferasa/genética , Geraniltranstransferasa/genética , Pérdida Auditiva/genética , Distrofias Musculares/genética , Mutación/genética , Insuficiencia Ovárica Primaria/genética , Adolescente , Adulto , Animales , Femenino , Técnicas de Sustitución del Gen/métodos , Pérdida Auditiva/diagnóstico por imagen , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofias Musculares/diagnóstico por imagen , Linaje , Insuficiencia Ovárica Primaria/diagnóstico por imagen , Estructura Secundaria de Proteína , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos , Adulto Joven
3.
Am J Med Genet A ; 185(1): 15-25, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33029936

RESUMEN

Biallelic mutations in SNORD118, encoding the small nucleolar RNA U8, cause leukoencephalopathy with calcifications and cysts (LCC). Given the difficulty in interpreting the functional consequences of variants in nonprotein encoding genes, and the high allelic polymorphism across SNORD118 in controls, we set out to provide a description of the molecular pathology and clinical spectrum observed in a cohort of patients with LCC. We identified 64 affected individuals from 56 families. Age at presentation varied from 3 weeks to 67 years, with disease onset after age 40 years in eight patients. Ten patients had died. We recorded 44 distinct, likely pathogenic, variants in SNORD118. Fifty two of 56 probands were compound heterozygotes, with parental consanguinity reported in only three families. Forty nine of 56 probands were either heterozygous (46) or homozygous (three) for a mutation involving one of seven nucleotides that facilitate a novel intramolecular interaction between the 5' end and 3' extension of precursor-U8. There was no obvious genotype-phenotype correlation to explain the marked variability in age at onset. Complementing recently published functional analyses in a zebrafish model, these data suggest that LCC most often occurs due to combinatorial severe and milder mutations, with the latter mostly affecting 3' end processing of precursor-U8.


Asunto(s)
Calcinosis/genética , Estudios de Asociación Genética , Leucoencefalopatías/genética , ARN Nucleolar Pequeño/genética , Adolescente , Adulto , Anciano , Animales , Calcinosis/complicaciones , Calcinosis/patología , Niño , Preescolar , Consanguinidad , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Leucoencefalopatías/complicaciones , Leucoencefalopatías/patología , Masculino , Persona de Mediana Edad , Patología Molecular , Adulto Joven , Pez Cebra/genética
4.
Ann Neurol ; 86(2): 310-315, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31187502

RESUMEN

Distinct clinical syndromes have been associated with pathogenic MT-ATP6 variants. In this cohort study, we identified 125 individuals (60 families) including 88 clinically affected individuals and 37 asymptomatic carriers. Thirty-one individuals presented with Leigh syndrome and 7 with neuropathy ataxia retinitis pigmentosa. The remaining 50 patients presented with variable nonsyndromic features including ataxia, neuropathy, and learning disability. We confirmed maternal inheritance in 39 families and demonstrated that tissue segregation patterns and phenotypic threshold are variant dependent. Our findings suggest that MT-ATP6-related mitochondrial DNA disease is best conceptualized as a mitochondrial disease spectrum disorder and should be routinely included in genetic ataxia and neuropathy gene panels. ANN NEUROL 2019;86:310-315.


Asunto(s)
Variación Genética/genética , Enfermedades Mitocondriales/epidemiología , Enfermedades Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/diagnóstico , Reino Unido/epidemiología , Adulto Joven
5.
Muscle Nerve ; 57(4): 550-560, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29149770

RESUMEN

INTRODUCTION: X-linked myotubular myopathy (XLMTM), characterized by severe hypotonia, weakness, respiratory distress, and early mortality, is rare and natural history studies are few. METHODS: RECENSUS is a multicenter chart review of male XLMTM patients characterizing disease burden and unmet medical needs. Data were collected between September 2014 and June 2016. RESULTS: Analysis included 112 patients at six clinical sites. Most recent patient age recorded was ≤18 months for 40 patients and >18 months for 72 patients. Mean (SD) age at diagnosis was 3.7 (3.7) months and 54.3 (77.1) months, respectively. Mortality was 44% (64% ≤18 months; 32% >18 months). Premature delivery occurred in 34/110 (31%) births. Nearly all patients (90%) required respiratory support at birth. In the first year of life, patients underwent an average of 3.7 surgeries and spent 35% of the year in the hospital. DISCUSSION: XLMTM is associated with high mortality, disease burden, and healthcare utilization. Muscle Nerve 57: 550-560, 2018.


Asunto(s)
Miopatías Estructurales Congénitas/mortalidad , Nacimiento Prematuro/epidemiología , Respiración Artificial/estadística & datos numéricos , Procedimientos Quirúrgicos Operativos/estadística & datos numéricos , Adolescente , Factores de Edad , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Masculino , Mortalidad , Estudios Retrospectivos , Adulto Joven
6.
Hum Mutat ; 38(8): 970-977, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28544275

RESUMEN

We report here the first families carrying recessive variants in the MSTO1 gene: compound heterozygous mutations were identified in two sisters and in an unrelated singleton case, who presented a multisystem complex phenotype mainly characterized by myopathy and cerebellar ataxia. Human MSTO1 is a poorly studied protein, suggested to have mitochondrial localization and to regulate morphology and distribution of mitochondria. As for other mutations affecting genes involved in mitochondrial dynamics, no biochemical defects typical of mitochondrial disorders were reported. Studies in patients' fibroblasts revealed that MSTO1 protein levels were strongly reduced, the mitochondrial network was fragmented, and the fusion events among mitochondria were decreased, confirming the deleterious effect of the identified variants and the role of MSTO1 in modulating mitochondrial dynamics. We also found that MSTO1 is mainly a cytosolic protein. These findings indicate recessive mutations in MSTO1 as a new cause for inherited neuromuscular disorders with multisystem features.


Asunto(s)
Ataxia/genética , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Dinámicas Mitocondriales/fisiología , Enfermedades Musculares/genética , Mutación/genética , Ataxia/etiología , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Dinámicas Mitocondriales/genética , Enfermedades Musculares/etiología
7.
Brain ; 137(Pt 1): 44-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24253200

RESUMEN

Childhood onset motor neuron diseases or neuronopathies are a clinically heterogeneous group of disorders. A particularly severe subgroup first described in 1894, and subsequently called Brown-Vialetto-Van Laere syndrome, is characterized by progressive pontobulbar palsy, sensorineural hearing loss and respiratory insufficiency. There has been no treatment for this progressive neurodegenerative disorder, which leads to respiratory failure and usually death during childhood. We recently reported the identification of SLC52A2, encoding riboflavin transporter RFVT2, as a new causative gene for Brown-Vialetto-Van Laere syndrome. We used both exome and Sanger sequencing to identify SLC52A2 mutations in patients presenting with cranial neuropathies and sensorimotor neuropathy with or without respiratory insufficiency. We undertook clinical, neurophysiological and biochemical characterization of patients with mutations in SLC52A2, functionally analysed the most prevalent mutations and initiated a regimen of high-dose oral riboflavin. We identified 18 patients from 13 families with compound heterozygous or homozygous mutations in SLC52A2. Affected individuals share a core phenotype of rapidly progressive axonal sensorimotor neuropathy (manifesting with sensory ataxia, severe weakness of the upper limbs and axial muscles with distinctly preserved strength of the lower limbs), hearing loss, optic atrophy and respiratory insufficiency. We demonstrate that SLC52A2 mutations cause reduced riboflavin uptake and reduced riboflavin transporter protein expression, and we report the response to high-dose oral riboflavin therapy in patients with SLC52A2 mutations, including significant and sustained clinical and biochemical improvements in two patients and preliminary clinical response data in 13 patients with associated biochemical improvements in 10 patients. The clinical and biochemical responses of this SLC52A2-specific cohort suggest that riboflavin supplementation can ameliorate the progression of this neurodegenerative condition, particularly when initiated soon after the onset of symptoms.


Asunto(s)
Parálisis Bulbar Progresiva/genética , Pérdida Auditiva Sensorineural/genética , Mutación/genética , Receptores Acoplados a Proteínas G/genética , Adolescente , Encéfalo/patología , Parálisis Bulbar Progresiva/tratamiento farmacológico , Carnitina/análogos & derivados , Carnitina/sangre , Niño , Preescolar , Exoma/genética , Femenino , Genotipo , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Análisis por Micromatrices , Enfermedad de la Neurona Motora/fisiopatología , Examen Neurológico , Linaje , ARN/biosíntesis , ARN/genética , Riboflavina/uso terapéutico , Análisis de Secuencia de ADN , Nervio Sural/patología , Vitaminas/uso terapéutico , Adulto Joven
8.
J Neuromuscul Dis ; 11(2): 361-368, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38189761

RESUMEN

Background: Spinal muscular atrophy (SMA) is a progressive neuromuscular disease caused by mutations in Survival motor neuron 1 (SMN1) gene, leading to reduction in survival motor neuron protein (SMN), key for motor neuron survival and function in the brainstem and spinal cord. Risdiplam is an orally administered SMN2-splicing modifier which increases production of functional SMN protein. Risdiplam was offered in the UK under early access to medicines scheme (EAMS) to SMA type 1 and 2 patients aged 2 months and older, not suitable for authorised treatments from September 2020 to December 2021. Objective: To describe the largest paediatric European real-world set of data on patients' characteristics and short-term safety for risdiplam in Great Britain through EAMS. Methods: We collated data from SMA REACH UK a national clinical and research network for all patients enrolled onto EAMS and assessed all submitted adverse events. Results: Of the 92 patients; 78% were Type 2 SMA, mean age 10.9 years, range 0-17 years. 56 were treatment naïve, 33 previously treated; of these 25 had received nusinersen, 3 previous treatment unknown. Sixty adverse events (AEs) were reported occurring in 34 patients. The commonest were respiratory tract infections and gastrointestinal disturbance. Four life-threatening events were reported with 2 deaths and permanent cessation of risdiplam in 3 patients.Overall, 38/60 AEs were considered unrelated to risdiplam, 10/60 related to risdiplam and for 12/60 causality not specified. Conclusions: This study found a safety profile similar to clinical trials with no new safety concerns identified. With the restricted eligibility of onasemnogene abeparvovec and complications of nusinersen administration, EAMS allowed access or continued treatment to naïve patients or patients no longer suitable for approved medications. Collection of longitudinal data for this complex population is needed, to provide greater insights into risdiplam's role in addressing patients' needs into the future.


Asunto(s)
Compuestos Azo , Atrofia Muscular Espinal , Atrofias Musculares Espinales de la Infancia , Humanos , Niño , Recién Nacido , Lactante , Preescolar , Adolescente , Reino Unido , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Atrofias Musculares Espinales de la Infancia/tratamiento farmacológico , Pirimidinas/efectos adversos
9.
Neurol Clin Pract ; 14(3): e200298, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38932995

RESUMEN

Background and Objectives: Nusinersen has shown significant functional motor benefit in the milder types of spinal muscular atrophy (SMA). Less is known on the respiratory outcomes in patients with nusinersen-treated SMA. The aim of this study was to describe changes in respiratory function in pediatric patients with SMA type 2 and 3 on regular treatment with nusinersen within the iSMAc international cohort and to compare their trajectory with the natural history (NH) data published by the consortium in 2020. Methods: This is a 5-year retrospective observational study of pediatric SMA type 2 and nonambulant type 3 (age ≤18 years) treated with nusinersen. The primary objective was to compare the slopes of decline in forced vital capacity % predicted (FVC% pred.), FVC, and age when FVC dropped below 60% between the treated patients and a control group from the natural history cohort. Data on peak cough flow and the use of noninvasive ventilation (NIV) and cough assist were collected. Results: Data were available for 69 treated patients, 53 were SMA type 2 and 16 type 3. The mean (SD) age at first injection was 8.5 (3.2) and 9.7 (3.7) years, respectively. The median (interquartile range) treatment duration was 1 (0.7; 1.9) and 1.2 (0.9; 1.9) years, respectively. At the time of the first nusinersen injection, 24 of 52 (46%) patients with SMA type 2 and 2 of 16 (13%) patients with SMA type 3 were on NIV. Forty-three of 53 (81%) and 4 of 16 (25%) patients used cough device. FVC% pred. in treated patients with SMA type 2 declined annually by 2.3% vs 3.9% in NH (p = 0.08) and in treated patients with type 3 by 2.6% vs 3.4% NH (p = 0.59). Patients treated reached FVC <60% later than untreated (12.1 vs 10 years, p = 0.05). A higher percentage of treated vs untreated patients maintained FVC% pred. equal/above their baseline after 12 (65% vs 36%) and 24 (50% vs 24%) months, respectively. NIV use among treated did not significantly change throughout 1-year follow-up. Discussion: This study included the largest real-world cohort of pediatric patients with milder SMA types. The results suggest a positive role of nusinersen in delaying the respiratory decline in patients treated longer than 1 year when compared with natural history. Larger cohorts and longer observation are planned. Classification of Evidence: This study provided Class III evidence that nusinersen slows progression for patients with SMA types 2 and 3 compared with a natural history cohort.

10.
Lancet Reg Health Eur ; 37: 100817, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38169987

RESUMEN

Background: Real-world data on the efficacy and safety of onasemnogene abeparvovec (OA) in spinal muscular atrophy (SMA) are needed, especially to overcome uncertainties around its use in older and heavier children. This study evaluated the efficacy and safety of OA in patients with SMA type 1 in the UK, including patients ≥2 years old and weighing ≥13.5 kg. Methods: This observational cohort study used data from patients with genetically confirmed SMA type 1 treated with OA between May 2021 and January 2023, at 6 infusion centres in the United Kingdom. Functional outcomes were assessed using age-appropriate functional scales. Safety analyses included review of liver function, platelet count, cardiac assessments, and steroid requirements. Findings: Ninety-nine patients (45 SMA therapy-naïve) were treated with OA (median age at infusion: 10 [range, 0.6-89] months; median weight: 7.86 [range, 3.2-20.2] kg; duration of follow-up: 3-22 months). After OA infusion, mean ± SD change in CHOP-INTEND score was 11.0 ± 10.3 with increased score in 66/78 patients (84.6%); patients aged <6 months had a 13.9 points higher gain in CHOP-INTEND score than patients ≥2 years (95% CI, 6.8-21.0; P < 0.001). Asymptomatic thrombocytopenia (71/99 patients; 71.7%), asymptomatic troponin-I elevation (30/89 patients; 33.7%) and transaminitis (87/99 patients; 87.9%) were reported. No thrombotic microangiopathy was observed. Median steroid treatment duration was 97 (range, 28-548) days with dose doubled in 35/99 patients (35.4%). There were 22.5-fold increased odds of having a transaminase peak >100 U/L (95% CI, 2.3-223.7; P = 0.008) and 21.2-fold increased odds of steroid doubling, as per treatment protocol (95% CI, 2.2-209.2; P = 0.009) in patients weighing ≥13.5 kg versus <8.5 kg. Weight at infusion was positively correlated with steroid treatment duration (r = 0.43; P < 0.001). Worsening transaminitis, despite doubling of oral prednisolone, led to treatment with intravenous methylprednisolone in 5 children. Steroid-sparing immunosuppressants were used in 5 children to enable steroid weaning. Two deaths apparently unrelated to OA were reported. Interpretation: OA led to functional improvements and was well tolerated with no persistent clinical complications, including in older and heavier patients. Funding: Novartis Innovative Therapies AG provided a grant for independent medical writing services.

11.
Dev Med Child Neurol ; 55(2): 154-161, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23163885

RESUMEN

AIM: Genetic testing in the epilepsies is becoming an increasingly accessible clinical tool. Mutations in the sodium channel alpha 1 subunit (SCN1A) gene are most notably associated with Dravet syndrome. This is the first study to assess the impact of SCN1A testing on patient management from both carer and physician perspectives. METHOD: Participants were identified prospectively from referrals to the Epilepsy Genetics Service in Glasgow and contacted via their referring clinicians. Questionnaires exploring the consequences of SCN1A genetic testing for each case were sent to carers and physicians. RESULTS: Of the 244 individuals contacted, 182 (75%) carried a SCN1A mutation. Carers of 187 (77%) patients responded (90 females, 97 males; mean age at referral 4 y 10 mo; interquartile range 9 y 1 mo). Of those participants whose children tested positive for a mutation, 87% reported that genetic testing was helpful, leading to treatment changes resulting in fewer seizures and improved access to therapies and respite care. Out of 187 physicians, 163 responded (87%), of whom 48% reported that a positive test facilitated diagnosis earlier than with clinical and electroencephalography data alone. It prevented additional investigations in 67% of patients, altered treatment approach in 69%, influenced medication choice in 74%, and, through medication change, improved seizure control in 42%. INTERPRETATION: In addition to confirming a clinical diagnosis, a positive SCN1A test result influenced treatment choice and assisted in accessing additional therapies, especially in the very young.


Asunto(s)
Epilepsias Mioclónicas/diagnóstico , Epilepsia/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Adolescente , Niño , Preescolar , Electroencefalografía , Epilepsias Mioclónicas/genética , Epilepsia/diagnóstico , Femenino , Pruebas Genéticas , Humanos , Lactante , Masculino , Mutación , Pautas de la Práctica en Medicina , Encuestas y Cuestionarios
12.
Hum Mutat ; 33(6): 981-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473935

RESUMEN

Ryanodine receptor 1 (RYR1) mutations are a common cause of congenital myopathies associated with both dominant and recessive inheritance. Histopathological findings frequently feature central cores or multi-minicores, more rarely, type 1 predominance/uniformity, fiber-type disproportion, increased internal nucleation, and fatty and connective tissue. We describe 71 families, 35 associated with dominant RYR1 mutations and 36 with recessive inheritance. Five of the dominant mutations and 35 of the 55 recessive mutations have not been previously reported. Dominant mutations, typically missense, were frequently located in recognized mutational hotspot regions, while recessive mutations were distributed throughout the entire coding sequence. Recessive mutations included nonsense and splice mutations expected to result in reduced RyR1 protein. There was wide clinical variability. As a group, dominant mutations were associated with milder phenotypes; patients with recessive inheritance had earlier onset, more weakness, and functional limitations. Extraocular and bulbar muscle involvement was almost exclusively observed in the recessive group. In conclusion, our study reports a large number of novel RYR1 mutations and indicates that recessive variants are at least as frequent as the dominant ones. Assigning pathogenicity to novel mutations is often difficult, and interpretation of genetic results in the context of clinical, histological, and muscle magnetic resonance imaging findings is essential.


Asunto(s)
Mutación , Miopatías Estructurales Congénitas/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Niño , Preescolar , Femenino , Genes Dominantes , Genes Recesivos , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Linaje
13.
Pediatr Blood Cancer ; 58(3): 459-61, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21584931

RESUMEN

Post-transplant lymphoproliferative disorder (PTLD) in the central nervous system (CNS) is a very rare complication of haematopoietic stem cell transplant (HSCT) and has a dismal prognosis. We report the successful treatment of this disorder with intrathecal rituximab therapy in two children who developed isolated CNS PTLD after HSCT. These children had failed to respond to standard chemotherapy, intravenous rituximab and EBV specific cellular therapy.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Antineoplásicos/administración & dosificación , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trastornos Linfoproliferativos/tratamiento farmacológico , Activación Viral , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Central/patología , Preescolar , Infecciones por Virus de Epstein-Barr/etiología , Humanos , Inyecciones Espinales , Leucemia de Células B/tratamiento farmacológico , Leucemia de Células B/etiología , Leucemia de Células B/patología , Linfohistiocitosis Hemofagocítica/tratamiento farmacológico , Linfohistiocitosis Hemofagocítica/etiología , Linfohistiocitosis Hemofagocítica/patología , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/patología , Imagen por Resonancia Magnética , Masculino , Rituximab
14.
Arch Dis Child ; 107(2): 160-165, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34244165

RESUMEN

OBJECTIVE: To describe age and time at key stages in the Duchenne muscular dystrophy (DMD) prediagnosis pathway at selected centres to identify opportunities for service improvement. DESIGN: A multicentre retrospective national audit. SETTING: Nine tertiary neuromuscular centres across the UK and Ireland. A prior single-centre UK audit of 20 patients with no DMD family history provided benchmark criteria. PATIENTS: Patients with a definitive diagnosis of DMD documented within 3 years prior to December 2018 (n=122). MAIN OUTCOME MEASURES: Mean age (months) at four key stages in the DMD diagnostic pathway and mean time (months) of presentational and diagnostic delay, and time from first reported symptoms to definitive diagnosis. Type of symptoms was also recorded. RESULTS: Overall, mean age at definitive diagnosis, age at first engagement with healthcare professional (HCP) and age at first reported symptoms were 53.9±29.7, 49.9±28.9 and 36.4±26.8 months, respectively. The presentational delay and time to diagnosis were 21.1 (±21.1) and 4.6 (±7.9) months, respectively. The mean time from first reported symptoms to definitive diagnosis was 24.2±20.9. The percentages of patients with motor and/or non-motor symptoms recorded were 88% (n=106/121) and 47% (n=57/121), respectively. CONCLUSIONS: Majority of data mirrored the benchmark audit. However, while the time to diagnosis was shorter, a presentational delay was observed. Failure to recognise early symptoms of DMD could be a contributing factor and represents an unmet need in the diagnosis pathway. Methods determining how to improve this need to be explored.


Asunto(s)
Distrofia Muscular de Duchenne/diagnóstico , Edad de Inicio , Benchmarking , Preescolar , Vías Clínicas , Progresión de la Enfermedad , Humanos , Distrofia Muscular de Duchenne/patología , Estudios Retrospectivos
15.
Neurology ; 99(14): e1511-e1526, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36192182

RESUMEN

BACKGROUND AND OBJECTIVES: ATP1A3 is associated with a broad spectrum of predominantly neurologic disorders, which continues to expand beyond the initially defined phenotypes of alternating hemiplegia of childhood, rapid-onset dystonia parkinsonism, and cerebellar ataxia, areflexia, pes cavus, optic atrophy, sensorineural hearing loss syndrome. This phenotypic variability makes it challenging to assess the pathogenicity of an ATP1A3 variant found in an undiagnosed patient. We describe the phenotypic features of individuals carrying a pathogenic/likely pathogenic ATP1A3 variant and perform a literature review of all ATP1A3 variants published thus far in association with human neurologic disease. Our aim is to demonstrate the heterogeneous clinical spectrum of the gene and look for phenotypic overlap between patients that will streamline the diagnostic process. METHODS: Undiagnosed individuals with ATP1A3 variants were identified within the cohort of the Deciphering Developmental Disorders study with additional cases contributed by collaborators internationally. Detailed clinical data were collected with consent through a questionnaire completed by the referring clinicians. PubMed was searched for publications containing the term "ATP1A3" from 2004 to 2021. RESULTS: Twenty-four individuals with a previously undiagnosed neurologic phenotype were found to carry 21 ATP1A3 variants. Eight variants have been previously published. Patients experienced on average 2-3 different types of paroxysmal events. Permanent neurologic features were common including microcephaly (7; 29%), ataxia (13; 54%), dystonia (10; 42%), and hypotonia (7; 29%). All patients had cognitive impairment. Neuropsychiatric diagnoses were reported in 16 (66.6%) individuals. Phenotypes were extremely varied, and most individuals did not fit clinical criteria for previously published phenotypes. On review of the literature, 1,108 individuals have been reported carrying 168 different ATP1A3 variants. The most common variants are associated with well-defined phenotypes, while more rare variants often result in very rare symptom correlations, such as are seen in our study. Combined Annotation-Dependent Depletion (CADD) scores of pathogenic and likely pathogenic variants were significantly higher and variants clustered within 6 regions of constraint. DISCUSSION: Our study shows that looking for a combination of paroxysmal events, hyperkinesia, neuropsychiatric symptoms, and cognitive impairment and evaluating the CADD score and variant location can help identify an ATP1A3-related condition, rather than applying diagnostic criteria alone.


Asunto(s)
Ataxia Cerebelosa , Trastornos Distónicos , Ataxia Cerebelosa/genética , Trastornos Distónicos/genética , Hemiplejía/genética , Humanos , Mutación/genética , Fenotipo , ATPasa Intercambiadora de Sodio-Potasio/genética
16.
Pediatr Surg Int ; 27(6): 591-4, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21243366

RESUMEN

PURPOSE: The aim of this study was to determine the efficacy of thymectomy to induce remission in juvenile myasthenia gravis. METHODS: A retrospective review of all patients undergoing a thymectomy for the treatment of juvenile myasthenia gravis was performed at a single tertiary referral centre between 1997 and 2009 (N = 8). All cases were moderate to severe cases (Osserman stage ≥ 2a). All operations were open. Median follow-up was 18 months (range 1-77). Postoperative progress was assessed using the De Filippi classification of remission. MAIN RESULTS: Only two minor complications were reported. Mean operative time was 120 min (range 80-290 min). Mean postoperative stay was 17 days (range 3-52 days). Remission was observed in 5 of 8 patients (62%) at last known follow up. CONCLUSIONS: Spontaneous remission rates for myasthenia gravis are quoted to be between 20 and 29% while remission rates following thymectomy are 29-68% at 3 years. Although not all of our patients have had 3 years of follow up-the remission rate of 62% demonstrated by this study is encouraging. If reproducible over a larger series this provides evidence of the efficacy of thymectomy for treating juvenile myasthenia gravis in selected patients.


Asunto(s)
Miastenia Gravis/cirugía , Timectomía/métodos , Adolescente , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Masculino , Inducción de Remisión/métodos , Estudios Retrospectivos , Resultado del Tratamiento
17.
Neuromuscul Disord ; 31(1): 21-28, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33250374

RESUMEN

SLC5A7 encodes the presynaptic sodium-dependant high-affinity choline transporter 1 (CHT), which uptakes choline to the presynaptic nerve terminal following the breakdown of acetylcholine by the acetylcholinesterase within the synaptic cleft. We report 5 patients from three consanguineous families with congenital myasthenic syndrome type 20 caused by novel mutations in SLC5A7. The individuals from family 1 and 2 were homozygous for c.320G>A; (p.Arg107His) and c.886G>A (p.Ala296Thr), respectively, and their phenotype was characterised by recurrent apnoeic attacks early after birth and learning and speech difficulties in childhood. Individuals from family 3 were homozygous for c.1240T>A (p.Tyr414Asn) and suffered from more severe central and peripheral manifestations with lack of spontaneous movements and respiratory drive and overall minimal response to external stimuli. All individuals tested showed neurophysiological defects compatible with impaired neuromuscular transmission. Combined treatment with cholinesterase inhibitors and ß2-adrenergic agonists was beneficial in patients from family 1 and 2. Affected individuals from family 3 died from complications directly related to their underlying genetic condition. This report provides three novel pathogenic variants in SLC5A7 and highlights the variability in the clinical phenotype, severity and prognosis of this syndrome.


Asunto(s)
Proteínas de Transporte de Membrana/genética , Síndromes Miasténicos Congénitos/genética , Simportadores , Acetilcolinesterasa/genética , Antagonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Inhibidores de la Colinesterasa/uso terapéutico , Femenino , Homocigoto , Humanos , Lactante , Recién Nacido , Masculino , Mutación Missense , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Linaje , Fenotipo , Terminales Presinápticos , Sodio/metabolismo
18.
Mol Genet Metab ; 100(2): 207-12, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20226704

RESUMEN

Phospholipase associated neurodegeneration (PLAN) comprises a heterogeneous group of autosomal recessive neurological disorders caused by mutations in the PLA2G6 gene. Direct gene sequencing detects approximately 85% mutations in infantile neuroaxonal dystrophy. We report the novel use of multiplex ligation-dependent probe amplification (MLPA) analysis to detect novel PLA2G6 duplications and deletions. The identification of such copy number variants (CNVs) expands the PLAN mutation spectrum and may account for up to 12.5% of PLA2G6 mutations. MLPA should thus be employed to detect CNVs of PLA2G6 in patients who show clinical features of PLAN but in whom both disease-causing mutations cannot be identified on routine sequencing.


Asunto(s)
Fosfolipasas A2 Grupo VI/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Secuencia de Bases , Encéfalo/patología , Preescolar , Consanguinidad , Eliminación de Gen , Duplicación de Gen , Trastornos Heredodegenerativos del Sistema Nervioso/diagnóstico , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Mutación , Patología Molecular
19.
Pediatrics ; 146(3)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32826339

RESUMEN

Disorders of central and peripheral nervous system should be considered in floppy infants with ventilator dependence. Workup for neuromuscular disorders should be undertaken in infants with hypotonia, weakness, contractures, feeding difficulties, or failed attempts at extubation. We present the case of a preterm infant with hypotonia and ventilator dependence where despite a positive result, further investigations were undertaken because of lack of clinical correlation. The infant had a rare combination of 2 neuromuscular conditions: X-linked myotubular myopathy and Duchenne muscular dystrophy. One was the reason for immediate clinical manifestation and the other influenced the prognosis and decision-making in determining reorientation of care. This case demonstrates the value of interpretation of a positive result that did not explain the clinical picture and warranted consideration of further diagnosis. This case also emphasizes the importance of discussions with family about the prognosis of 2 conditions that influenced decision making.


Asunto(s)
Recien Nacido Prematuro , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/diagnóstico , Miopatías Estructurales Congénitas/complicaciones , Miopatías Estructurales Congénitas/diagnóstico , Resultado Fatal , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro/fisiología , Masculino , Distrofia Muscular de Duchenne/genética , Miopatías Estructurales Congénitas/genética
20.
Arch Dis Child ; 105(4): 332-338, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31484632

RESUMEN

PURPOSE: Individuals with X-linked myotubular myopathy (XLMTM) who survive infancy require extensive supportive care, including ventilator assistance, wheelchairs and feeding tubes. Half die before 18 months of age. We explored respiratory support and associated mortality risk in RECENSUS, particularly among patients ≤5 years old who received respiratory support at birth; this subgroup closely matches patients in the ASPIRO trial of gene therapy for XLMTM. DESIGN: RECENSUS is an international, retrospective study of patients with XLMTM. Descriptive and time-to-event analyses examined survival on the basis of age, respiratory support, tracheostomy use, predicted mutational effects and life-sustaining care. RESULTS: Outcomes for 145 patients were evaluated. Among 126 patients with respiratory support at birth, mortality was 47% overall and 59% among those ≤5 years old. Median survival time was shorter for patients ≤5 years old than for those >5 years old (2.2 years (IQR 0.7-5.6) vs 30.2 years (IQR 19.4-30.2)). The most common cause of death was respiratory failure (66.7%). Median survival time was longer for patients with a tracheostomy than for those without (22.8 years (IQR 8.7-30.2) vs 1.8 years (IQR 0.2-not estimable)). The proportion of patients living without a tracheostomy was 50% at age 6 months and 28% at age 2 years. Median survival time was longer with provision of life-sustaining care than without (19.4 years (IQR 3.1-not estimable) vs 0.2 years (IQR 0.1-2.1)). CONCLUSIONS: High mortality, principally due to respiratory failure, among patients with XLMTM ≤5 years old despite respiratory support underscores the need for early diagnosis, informed decision-making and disease-modifying therapies. TRIAL REGISTRATION NUMBER: NCT02231697.


Asunto(s)
Miopatías Estructurales Congénitas/mortalidad , Respiración Artificial/estadística & datos numéricos , Factores de Edad , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Miopatías Estructurales Congénitas/fisiopatología , Miopatías Estructurales Congénitas/terapia , Nacimiento Prematuro/epidemiología , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA