Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Med Genet A ; : e63656, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760879

RESUMEN

KIF1A-related disorders (KRDs) encompass recessive and dominant variants with wide clinical variability. Recent genetic investigations have expanded the clinical phenotypes of heterozygous KIF1A variants. However, there have been a few long-term observational studies of patients with heterozygous KIF1A variants. A retrospective chart review of consecutive patients diagnosed with spastic paraplegia at Miyagi Children's Hospital from 2016 to 2020 identified six patients with heterozygous KIF1A variants. To understand the long-term changes in clinical symptoms, we examined these patients in terms of their characteristics, clinical symptoms, results of electrophysiological and neuroimaging studies, and genetic testing. The median follow-up period was 30 years (4-44 years). This long-term observational study showed that early developmental delay and equinus gait, or unsteady gait, are the first signs of disease onset, appearing with the commencement of independent walking. In addition, later age-related progression was observed in spastic paraplegia, and the appearance of axonal neuropathy and reduced visual acuity were characteristic features of the late disease phenotype. Brain imaging showed age-related progression of cerebellar atrophy and the appearance of hyperintensity of optic radiation on T2WI and FLAIR imaging. Long-term follow-up revealed a pattern of steady progression and a variety of clinical symptoms, including spastic paraplegia, peripheral neuropathy, reduced visual acuity, and some degree of cerebellar ataxia. Clinical variability between patients was observed to some extent, and therefore, further studies are required to determine the phenotype-genotype correlation.

2.
Am J Hum Genet ; 102(2): 321-329, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29394991

RESUMEN

Early-onset epileptic encephalopathies, including West syndrome (WS), are a group of neurological disorders characterized by developmental impairments and intractable seizures from early infancy. We have now identified biallelic CNPY3 variants in three individuals with WS; these include compound-heterozygous missense and frameshift variants in a family with two affected siblings (individuals 1 and 2) and a homozygous splicing variant in a consanguineous family (individual 3). All three individuals showed hippocampal malrotation. In individuals 1 and 2, electroencephalography (EEG) revealed characteristic fast waves and diffuse sharp- and slow-wave complexes. The fast waves were clinically associated with seizures. CNPY3 encodes a co-chaperone in the endoplasmic reticulum and regulates the subcellular distribution and responses of multiple Toll-like receptors. The amount of CNPY3 in lymphoblastoid cells derived from individuals 1 and 2 was severely lower than that in control cells. Cnpy3-knockout mice exhibited spastic or dystonic features under resting conditions and hyperactivity and anxiolytic behavior during the open field test. Also, their resting EEG showed enhanced activity in the fast beta frequency band (20-35 Hz), which could mimic the fast waves in individuals 1 and 2. These data suggest that CNPY3 and Cnpy3 perform essential roles in brain function in addition to known Toll-like receptor-dependent immune responses.


Asunto(s)
Chaperonas Moleculares/genética , Mutación , Convulsiones/genética , Espasmos Infantiles/genética , Adolescente , Secuencia de Aminoácidos , Animales , Niño , Consanguinidad , Electroencefalografía , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Familia , Femenino , Expresión Génica , Heterocigoto , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Convulsiones/diagnóstico por imagen , Convulsiones/fisiopatología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Hermanos , Espasmos Infantiles/diagnóstico por imagen , Espasmos Infantiles/fisiopatología
3.
J Med Genet ; 56(6): 396-407, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842224

RESUMEN

BACKGROUND: Rett syndrome (RTT) is a characteristic neurological disease presenting with regressive loss of neurodevelopmental milestones. Typical RTT is generally caused by abnormality of methyl-CpG binding protein 2 (MECP2). Our objective to investigate the genetic landscape of MECP2-negative typical/atypical RTT and RTT-like phenotypes using whole exome sequencing (WES). METHODS: We performed WES on 77 MECP2-negative patients either with typical RTT (n=11), atypical RTT (n=22) or RTT-like phenotypes (n=44) incompatible with the RTT criteria. RESULTS: Pathogenic or likely pathogenic single-nucleotide variants in 28 known genes were found in 39 of 77 (50.6%) patients. WES-based CNV analysis revealed pathogenic deletions involving six known genes (including MECP2) in 8 of 77 (10.4%) patients. Overall, diagnostic yield was 47 of 77 (61.0 %). Furthermore, strong candidate variants were found in four novel genes: a de novo variant in each of ATPase H+ transporting V0 subunit A1 (ATP6V0A1), ubiquitin-specific peptidase 8 (USP8) and microtubule-associated serine/threonine kinase 3 (MAST3), as well as biallelic variants in nuclear receptor corepressor 2 (NCOR2). CONCLUSIONS: Our study provides a new landscape including additional genetic variants contributing to RTT-like phenotypes, highlighting the importance of comprehensive genetic analysis.


Asunto(s)
Secuenciación del Exoma , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Fenotipo , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Ontología de Genes , Redes Reguladoras de Genes , Estudios de Asociación Genética/métodos , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Polimorfismo de Nucleótido Simple
4.
Am J Hum Genet ; 99(4): 950-961, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27666374

RESUMEN

We describe four families with affected siblings showing unique clinical features: early-onset (before 1 year of age) progressive diffuse brain atrophy with regression, postnatal microcephaly, postnatal growth retardation, muscle weakness/atrophy, and respiratory failure. By whole-exome sequencing, we identified biallelic TBCD mutations in eight affected individuals from the four families. TBCD encodes TBCD (tubulin folding co-factor D), which is one of five tubulin-specific chaperones playing a pivotal role in microtubule assembly in all cells. A total of seven mutations were found: five missense mutations, one nonsense, and one splice site mutation resulting in a frameshift. In vitro cell experiments revealed the impaired binding between most mutant TBCD proteins and ARL2, TBCE, and ß-tubulin. The in vivo experiments using olfactory projection neurons in Drosophila melanogaster indicated that the TBCD mutations caused loss of function. The wide range of clinical severity seen in this neurodegenerative encephalopathy may result from the residual function of mutant TBCD proteins. Furthermore, the autopsied brain from one deceased individual showed characteristic neurodegenerative findings: cactus and somatic sprout formations in the residual Purkinje cells in the cerebellum, which are also seen in some diseases associated with mitochondrial impairment. Defects of microtubule formation caused by TBCD mutations may underlie the pathomechanism of this neurodegenerative encephalopathy.


Asunto(s)
Alelos , Encefalopatías/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Enfermedades Neurodegenerativas/genética , Adolescente , Edad de Inicio , Secuencia de Aminoácidos , Animales , Encefalopatías/patología , Encefalopatías/fisiopatología , Niño , Preescolar , Drosophila melanogaster/genética , Exoma , Femenino , Mutación del Sistema de Lectura/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Linaje , Sitios de Empalme de ARN/genética , Tubulina (Proteína)/metabolismo , Adulto Joven
5.
Brain ; 141(6): 1703-1718, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29668857

RESUMEN

V-type proton (H+) ATPase (v-ATPase) is a multi-subunit proton pump that regulates pH homeostasis in all eukaryotic cells; in neurons, v-ATPase plays additional and unique roles in synapse function. Through whole exome sequencing, we identified de novo heterozygous mutations (p.Pro27Arg, p.Asp100Tyr, p.Asp349Asn, p.Asp371Gly) in ATP6V1A, encoding the A subunit of v-ATPase, in four patients with developmental encephalopathy with epilepsy. Early manifestations, observed in all patients, were developmental delay and febrile seizures, evolving to encephalopathy with profound delay, hypotonic/dyskinetic quadriparesis and intractable multiple seizure types in two patients (p.Pro27Arg, p.Asp100Tyr), and to moderate delay with milder epilepsy in the other two (p.Asp349Asn, p.Asp371Gly). Modelling performed on the available prokaryotic and eukaryotic structures of v-ATPase predicted p.Pro27Arg to perturb subunit interaction, p.Asp100Tyr to cause steric hindrance and destabilize protein folding, p.Asp349Asn to affect the catalytic function and p.Asp371Gly to impair the rotation process, necessary for proton transport. We addressed the impact of p.Asp349Asn and p.Asp100Tyr mutations on ATP6V1A expression and function by analysing ATP6V1A-overexpressing HEK293T cells and patients' lymphoblasts. The p.Asp100Tyr mutant was characterized by reduced expression due to increased degradation. Conversely, no decrease in expression and clearance was observed for p.Asp349Asn. In HEK293T cells overexpressing either pathogenic or control variants, p.Asp349Asn significantly increased LysoTracker® fluorescence with no effects on EEA1 and LAMP1 expression. Conversely, p.Asp100Tyr decreased both LysoTracker® fluorescence and LAMP1 levels, leaving EEA1 expression unaffected. Both mutations decreased v-ATPase recruitment to autophagosomes, with no major impact on autophagy. Experiments performed on patients' lymphoblasts using the LysoSensor™ probe revealed lower pH of endocytic organelles for p.Asp349Asn and a reduced expression of LAMP1 with no effect on the pH for p.Asp100Tyr. These data demonstrate gain of function for p.Asp349Asn characterized by an increased proton pumping in intracellular organelles, and loss of function for p.Asp100Tyr with decreased expression of ATP6V1A and reduced levels of lysosomal markers. We expressed p.Asp349Asn and p.Asp100Tyr in rat hippocampal neurons and confirmed significant and opposite effects in lysosomal labelling. However, both mutations caused a similar defect in neurite elongation accompanied by loss of excitatory inputs, revealing that altered lysosomal homeostasis markedly affects neurite development and synaptic connectivity. This study provides evidence that de novo heterozygous ATP6V1A mutations cause a developmental encephalopathy with a pathomechanism that involves perturbations of lysosomal homeostasis and neuronal connectivity, uncovering a novel role for v-ATPase in neuronal development.


Asunto(s)
Encefalopatías/genética , Epilepsia/genética , Mutación/genética , ATPasas de Translocación de Protón Vacuolares/genética , Adolescente , Animales , Encéfalo/diagnóstico por imagen , Encefalopatías/complicaciones , Encefalopatías/patología , Células Cultivadas , Niño , Estudios de Cohortes , Epilepsia/complicaciones , Epilepsia/patología , Femenino , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Lisosomas/patología , Masculino , Modelos Moleculares , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Ratas , Sinapsis/metabolismo , Sinapsis/patología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Secuenciación del Exoma
6.
J Hum Genet ; 63(3): 263-270, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29339779

RESUMEN

Misato 1, mitochondrial distribution and morphology regulator (encoded by the MSTO1 gene), is involved in mitochondrial distribution and morphology. Recently, MSTO1 mutations have been shown to cause clinical manifestations suggestive of mitochondrial dysfunction, such as muscle weakness, short stature, motor developmental delay, and cerebellar atrophy. Both autosomal dominant and recessive modes of inheritance have been suggested. We performed whole-exome sequencing in two unrelated patients showing cerebellar atrophy, intellectual disability, and pigmentary retinopathy. Three novel mutations were identified: c.836 G > A (p.Arg279His), c.1099-1 G > A (p.Val367Trpfs*2), and c.79 C > T (p.Gln27*). Both patients had compound heterozygous mutations with a combination of protein-truncation mutation and missense mutation, the latter shared by them both. This survey of two patients with recessive and novel MSTO1 mutations provides additional clinical and genetic information on the pathogenicity of MSTO1 in humans.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cerebelosas/diagnóstico , Enfermedades Cerebelosas/genética , Proteínas del Citoesqueleto/genética , Genes Recesivos , Mutación , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/genética , Adolescente , Edad de Inicio , Alelos , Atrofia , Línea Celular , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Secuenciación Completa del Genoma
7.
J Hum Genet ; 62(6): 653-655, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28148925

RESUMEN

Cerebral, ocular, dental, auricular, skeletal (CODAS) syndrome is a rare autosomal recessive multisystem disorder caused by mutations in LONP1. It is characterized by intellectual disability, cataracts, delayed tooth eruption, malformed auricles and skeletal abnormalities. We performed whole-exome sequencing on a 12-year-old Japanese male with severe intellectual disability, congenital bilateral cataracts, spasticity, hypotonia with motor regression and progressive cerebellar atrophy with hyperintensity of the cerebellar cortex on T2-weighted images. We detected compound heterozygous mutation in LONP1. One allele contained a paternally inherited frameshift mutation (p.Ser100Glnfs*46). The other allele contained a maternally inherited missense mutation (p.Arg786Trp), which was predicted to be pathogenic by web-based prediction tools. The two mutations were not found in Exome Variant Server or our 575 in-house control exomes. Some features were not consistent with CODAS syndrome but overlapped with Marinesco-Sjögren syndrome, a multisystem disorder caused by a mutation in SIL1. An atypical mutation site may result in atypical presentation of the LONP1 mutation.


Asunto(s)
Proteasas ATP-Dependientes/genética , Anomalías Craneofaciales/genética , Anomalías del Ojo/genética , Trastornos del Crecimiento/genética , Luxación Congénita de la Cadera/genética , Discapacidad Intelectual/genética , Proteínas Mitocondriales/genética , Osteocondrodisplasias/genética , Degeneraciones Espinocerebelosas/genética , Anomalías Dentarias/genética , Niño , Anomalías Craneofaciales/fisiopatología , Exoma/genética , Anomalías del Ojo/fisiopatología , Mutación del Sistema de Lectura/genética , Predisposición Genética a la Enfermedad , Trastornos del Crecimiento/fisiopatología , Luxación Congénita de la Cadera/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Osteocondrodisplasias/fisiopatología , Dominios Proteicos/genética , Degeneraciones Espinocerebelosas/fisiopatología , Anomalías Dentarias/fisiopatología
8.
J Hum Genet ; 61(6): 527-31, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26888482

RESUMEN

Cerebellar atrophy is recognized in various types of childhood neurological disorders with clinical and genetic heterogeneity. Genetic analyses such as whole exome sequencing are useful for elucidating the genetic basis of these conditions. Pathological recessive mutations in Sep (O-phosphoserine) tRNA:Sec (selenocysteine) tRNA synthase (SEPSECS) have been reported in a total of 11 patients with pontocerebellar hypoplasia type 2, progressive cerebellocerebral atrophy or progressive encephalopathy, yet detailed clinical features are limited to only four patients. We identified two new families with progressive cerebellar atrophy, and by whole exome sequencing detected biallelic SEPSECS mutations: c.356A>G (p.Asn119Ser) and c.77delG (p.Arg26Profs*42) in family 1, and c.356A>G (p.Asn119Ser) and c.467G>A (p.Arg156Gln) in family 2. Their development was slightly delayed regardless of normal brain magnetic resonance imaging (MRI) in infancy. The progression of clinical symptoms in these families is evidently slower than in previously reported cases, and the cerebellar atrophy milder by brain MRI, indicating that SEPSECS mutations are also involved in milder late-onset cerebellar atrophy.


Asunto(s)
Alelos , Aminoacil-ARNt Sintetasas/genética , Mutación , Atrofias Olivopontocerebelosas/diagnóstico , Atrofias Olivopontocerebelosas/genética , Adolescente , Sustitución de Aminoácidos , Encéfalo/citología , Niño , Preescolar , Exoma , Femenino , Frecuencia de los Genes , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
9.
Ann Neurol ; 78(3): 375-86, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26018084

RESUMEN

OBJECTIVE: Focal cortical dysplasia (FCD) type IIb is a cortical malformation characterized by cortical architectural abnormalities, dysmorphic neurons, and balloon cells. It has been suggested that FCDs are caused by somatic mutations in cells in the developing brain. Here, we explore the possible involvement of somatic mutations in FCD type IIb. METHODS: We collected a total of 24 blood-brain paired samples with FCD, including 13 individuals with FCD type IIb, 5 with type IIa, and 6 with type I. We performed whole-exome sequencing using paired samples from 9 of the FCD type IIb subjects. Somatic MTOR mutations were identified and further investigated using all 24 paired samples by deep sequencing of the entire gene's coding region. Somatic MTOR mutations were confirmed by droplet digital polymerase chain reaction. The effect of MTOR mutations on mammalian target of rapamycin (mTOR) kinase signaling was evaluated by immunohistochemistry and Western blotting analyses of brain samples and by in vitro transfection experiments. RESULTS: We identified four lesion-specific somatic MTOR mutations in 6 of 13 (46%) individuals with FCD type IIb showing mutant allele rates of 1.11% to 9.31%. Functional analyses showed that phosphorylation of ribosomal protein S6 in FCD type IIb brain tissues with MTOR mutations was clearly elevated, compared to control samples. Transfection of any of the four MTOR mutants into HEK293T cells led to elevated phosphorylation of 4EBP, the direct target of mTOR kinase. INTERPRETATION: We found low-prevalence somatic mutations in MTOR in FCD type IIb, indicating that activating somatic mutations in MTOR cause FCD type IIb.


Asunto(s)
Encéfalo/patología , Malformaciones del Desarrollo Cortical del Grupo II/genética , Mutación/genética , Serina-Treonina Quinasas TOR/genética , Adolescente , Adulto , Niño , Femenino , Células HEK293 , Humanos , Masculino , Malformaciones del Desarrollo Cortical/diagnóstico , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical del Grupo II/diagnóstico
10.
Epilepsia ; 57(4): 566-73, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26918889

RESUMEN

OBJECTIVE: GABRA1 mutations have been identified in patients with familial juvenile myoclonic epilepsy, sporadic childhood absence epilepsy, and idiopathic familial generalized epilepsy. In addition, de novo GABRA1 mutations were recently reported in a patient with infantile spasms and four patients with Dravet syndrome. Those reports suggest that GABRA1 mutations are associated with infantile epilepsy including early onset epileptic encephalopathies. In this study, we searched for GABRA1 mutations in patients with infantile epilepsy to investigate the phenotypic spectrum of GABRA1 mutations. METHODS: In total, 526 and 145 patients with infantile epilepsy were analyzed by whole-exome sequencing and GABRA1-targeted resequencing, respectively. RESULTS: We identified five de novo missense GABRA1 mutations in six unrelated patients. A p.R112Q mutation in the long extracellular N-terminus was identified in a patient with infantile epilepsy; p.P260L, p.M263T, and p.M263I in transmembrane spanning domain 1 (TM1) were identified in three unrelated patients with West syndrome and a patient with Ohtahara syndrome, respectively; and p.V287L in TM2 was identified in a patient with unclassified early onset epileptic encephalopathy. Four of these mutations have not been observed previously. SIGNIFICANCE: Our study suggests that de novo GABRA1 mutations can cause early onset epileptic encephalopathies, including Ohtahara syndrome and West syndrome.


Asunto(s)
Mutación Missense/genética , Receptores de GABA-A/genética , Espasmos Infantiles/diagnóstico , Espasmos Infantiles/genética , Secuencia de Aminoácidos , Niño , Preescolar , Electroencefalografía , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Espasmos Infantiles/fisiopatología
11.
J Hum Genet ; 60(9): 509-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26016410

RESUMEN

Defects in the mitochondrial translation apparatus can impair energy production in affected tissues and organs. Most components of this apparatus are encoded by nuclear genes, including GFM2, which encodes a mitochondrial ribosome recycling factor. A few patients with mutations in some of these genes have been reported to date. Here, we present two female siblings with arthrogryposis multiplex congenita, optic atrophy and severe mental retardation. The younger sister had a progressive cerebellar atrophy and bilateral neuropathological findings in the brainstem. Although her cerebrospinal fluid (CSF) levels of lactate and pyruvate were not increased, brain magnetic resonance spectroscopy showed a lactate peak. Additionally, her CSF lactate/pyruvate and serum beta-hydroxybutyrate/acetoacetate ratios were high, and levels of oxidative phosphorylation in skin fibroblasts were reduced. We therefore diagnosed Leigh syndrome. Genomic investigation confirmed the presence of compound heterozygous GFM2 mutations (c.206+4A>G and c.2029-1G>A) in both siblings, causing aberrant splicing with premature stop codons (p.Gly50Glufs*4 and p.Ala677Leufs*2, respectively). These findings suggest that GFM2 mutations could be causative of a phenotype of Leigh syndrome with arthrogryposis multiplex congenita.


Asunto(s)
Artrogriposis/genética , Enfermedad de Leigh/genética , Proteínas Mitocondriales/genética , Factor G de Elongación Peptídica/genética , Artrogriposis/complicaciones , Secuencia de Bases , Niño , Resultado Fatal , Femenino , Heterocigoto , Humanos , Lactante , Enfermedad de Leigh/complicaciones , Linaje , Hermanos
12.
J Hum Genet ; 60(12): 739-42, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26354034

RESUMEN

Recently, de novo KIF1A mutations were identified in patients with intellectual disability, spasticity and cerebellar atrophy and/or optic nerve atrophy. In this study, we analyzed a total of 62 families, including 68 patients with genetically unsolved childhood cerebellar atrophy, by whole-exome sequencing (WES). We identified five de novo missense KIF1A mutations, including only one previously reported mutation (p.Arg316Trp). All the mutations are located in the motor domain of KIF1A. In all patients, initial symptom onset was during the infantile period, and included developmental delay in three patients and gait disturbance in two. Thereafter, they showed gait disturbances, exaggerated deep tendon reflexes, cerebellar symptoms and cerebellar atrophy on brain magnetic resonance imaging. Four patients showed lower limb spasticity, upper limb clumsiness and visual disturbances. Nerve conduction study revealed peripheral neuropathy in three patients. This study further delineates clinical features of de novo KIF1A mutations. Genetic testing of KIF1A should be considered in children with developmental delay, cerebellar atrophy and pyramidal features.


Asunto(s)
Enfermedades Cerebelosas/genética , Discapacidad Intelectual/genética , Cinesinas/genética , Espasticidad Muscular/genética , Mutación Missense , Trastornos de la Visión/genética , Sustitución de Aminoácidos , Enfermedades Cerebelosas/patología , Enfermedades Cerebelosas/fisiopatología , Femenino , Humanos , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Extremidad Inferior/patología , Extremidad Inferior/fisiopatología , Masculino , Espasticidad Muscular/patología , Espasticidad Muscular/fisiopatología , Trastornos de la Visión/patología , Trastornos de la Visión/fisiopatología
13.
J Hum Genet ; 60(4): 175-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25608832

RESUMEN

Whole-exome sequencing (WES) is becoming a standard tool for detecting nucleotide changes, and determining whether WES data can be used for the detection of copy-number variations (CNVs) is of interest. To date, several algorithms have been developed for such analyses, although verification is needed to establish if they fit well for the appropriate purpose, depending on the characteristics of each algorithm. Here, we performed WES CNV analysis using the eXome Hidden Markov Model (XHMM). We validated its performance using 27 rare CNVs previously identified by microarray as positive controls, finding that the detection rate was 59%, or higher (89%) with three or more targets. XHMM can be effectively used, especially for the detection of >200 kb CNVs. XHMM may be useful for deletion breakpoint detection. Next, we applied XHMM to genetically unsolved patients, demonstrating successful identification of pathogenic CNVs: 1.5-1.9-Mb deletions involving NSD1 in patients with unknown overgrowth syndrome leading to the diagnosis of Sotos syndrome, and 6.4-Mb duplication involving MECP2 in affected brothers with late-onset spasm and progressive cerebral/cerebellar atrophy confirming the clinical suspect of MECP2 duplication syndrome. The possibility of an 'exome-first' approach for clinical genetic investigation may be considered to save the cost of multiple investigations.


Asunto(s)
Variaciones en el Número de Copia de ADN , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Cadenas de Markov , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Algoritmos , Atrofia , Encefalopatías/genética , Encefalopatías/patología , Puntos de Rotura del Cromosoma , Duplicación Cromosómica , Biología Computacional/métodos , Femenino , Gigantismo/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Discapacidad Intelectual/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteínas Nucleares/genética , Sensibilidad y Especificidad , Eliminación de Secuencia
14.
Am J Med Genet A ; 167(7): 1593-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25931020

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder predominantly affecting females. Females with the MECP2 mutations exhibit a broad spectrum of clinical manifestations ranging from classical Rett syndrome to asymptomatic carriers. Mutations of genes encoding cyclin-dependent kinase-like 5 (CDKL5) and forkhead box G1 (FOXG1) are also found in early onset RTT variants. Here, we present the first report of a female patient with RTT-like phenotype caused by SHANK3 (SH3 and multiple ankylin repeat domain 3) mutation, indicating that the clinical spectrum of SHANK3 mutations may extend to RTT-like phenotype in addition to (severe) developmental delay, absence of expressive speech, autistic behaviors and intellectual disability.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Fenotipo , Síndrome de Rett/genética , Síndrome de Rett/patología , Adulto , Secuencia de Bases , Exoma/genética , Femenino , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa Multiplex , Análisis de Secuencia de ADN
15.
Epilepsia ; 56(9): e121-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26140313

RESUMEN

KCNT1 mutations have been found in epilepsy of infancy with migrating focal seizures (EIMFS; also known as migrating partial seizures in infancy), autosomal dominant nocturnal frontal lobe epilepsy, and other types of early onset epileptic encephalopathies (EOEEs). We performed KCNT1-targeted next-generation sequencing (207 samples) and/or whole-exome sequencing (229 samples) in a total of 362 patients with Ohtahara syndrome, West syndrome, EIMFS, or unclassified EOEEs. We identified nine heterozygous KCNT1 mutations in 11 patients: nine of 18 EIMFS cases (50%) in whom migrating foci were observed, one of 180 West syndrome cases (0.56%), and one of 66 unclassified EOEE cases (1.52%). KCNT1 mutations occurred de novo in 10 patients, and one was transmitted from the patient's mother who carried a somatic mosaic mutation. The mutations accumulated in transmembrane segment 5 (2/9, 22.2%) and regulators of K(+) conductance domains (7/9, 77.8%). Five of nine mutations were recurrent. Onset ages ranged from the neonatal period (<1 month) in five patients (5/11, 45.5%) to 1-4 months in six patients (6/11, 54.5%). A generalized attenuation of background activity on electroencephalography was seen in six patients (6/11, 54.5%). Our study demonstrates that the phenotypic spectrum of de novo KCNT1 mutations is largely restricted to EIMFS.


Asunto(s)
Mutación/genética , Proteínas del Tejido Nervioso/genética , Canales de Potasio/genética , Espasmos Infantiles/genética , Encéfalo/patología , Niño , Preescolar , Análisis Mutacional de ADN , Electroencefalografía , Humanos , Lactante , Imagen por Resonancia Magnética , Canales de potasio activados por Sodio
16.
Epilepsia ; 56(6): 841-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25864721

RESUMEN

OBJECTIVE: Recently, de novo mutations in GRIN1 have been identified in patients with nonsyndromic intellectual disability and epileptic encephalopathy. Whole exome sequencing (WES) analysis of patients with genetically unsolved epileptic encephalopathies identified four patients with GRIN1 mutations, allowing us to investigate the phenotypic spectrum of GRIN1 mutations. METHODS: Eighty-eight patients with unclassified early onset epileptic encephalopathies (EOEEs) with an age of onset <1 year were analyzed by WES. The effect of mutations on N-methyl-D-aspartate (NMDA) receptors was examined by mapping altered amino acids onto three-dimensional models. RESULTS: We identified four de novo missense GRIN1 mutations in 4 of 88 patients with unclassified EOEEs. In these four patients, initial symptoms appeared within 3 months of birth, including hyperkinetic movements in two patients (2/4, 50%), and seizures in two patients (2/4, 50%). Involuntary movements, severe developmental delay, and intellectual disability were recognized in all four patients. In addition, abnormal eye movements resembling oculogyric crises and stereotypic hand movements were observed in two and three patients, respectively. All the four patients exhibited only nonspecific focal and diffuse epileptiform abnormality, and never showed suppression-burst or hypsarrhythmia during infancy. A de novo mosaic mutation (c.1923G>A) with a mutant allele frequency of 16% (in DNA of blood leukocytes) was detected in one patient. Three mutations were located in the transmembrane domain (3/4, 75%), and one in the extracellular loop near transmembrane helix 1. All the mutations were predicted to impair the function of the NMDA receptor. SIGNIFICANCE: Clinical features of de novo GRIN1 mutations include infantile involuntary movements, seizures, and hand stereotypies, suggesting that GRIN1 mutations cause encephalopathy resulting in seizures and movement disorders.


Asunto(s)
Encefalopatías/genética , Epilepsia/genética , Predisposición Genética a la Enfermedad/genética , Hipercinesia/genética , Mutación Missense/genética , Proteínas del Tejido Nervioso/genética , Receptores de N-Metil-D-Aspartato/genética , Trastorno de Movimiento Estereotipado/genética , Adolescente , Encefalopatías/complicaciones , Niño , Preescolar , Análisis Mutacional de ADN , Electroencefalografía , Epilepsia/complicaciones , Femenino , Humanos , Hipercinesia/complicaciones , Imagen por Resonancia Magnética , Masculino , Trastorno de Movimiento Estereotipado/complicaciones
17.
Macromol Rapid Commun ; 36(6): 515-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25619869

RESUMEN

Microdevices designed for practical environmental pollution monitoring need to detect specific pollutants such as dioxins. Bisphenol A (BPA) has been widely used as a monomer for the synthesis of polycarbonate and epoxy resins. However, the recent discovery of its high potential ability to disrupt human endocrine systems has made the development of smart systems and microdevices for its detection and removal necessary. Molecule-responsive microsized hydrogels with ß-cycrodextrin (ß-CD) as ligands are prepared by photopolymerization using a fluorescence microscope. The molecule-responsive micro-hydrogels show ultra-quick shrinkage in response to target BPA. Furthermore, the flow rate of a microchannel is autonomously regulated by the molecule-responsive shrinking of their hydrogels as smart microvalves.


Asunto(s)
Hidrogeles/química , Polímeros/química , beta-Ciclodextrinas/química , Compuestos de Bencidrilo/química , Luz , Impresión Molecular , Fenoles/química , Polimerizacion/efectos de la radiación , Polímeros/síntesis química
18.
Neurogenetics ; 15(2): 85-92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24253414

RESUMEN

Defects of the human glycosylphosphatidylinositol (GPI) anchor biosynthetic pathway show a broad range of clinical phenotypes. A homozygous mutation in PIGN, a member of genes involved in the GPI anchor-synthesis pathway, was previously reported to cause dysmorphic features, multiple congenital anomalies, severe neurological impairment, and seizure in a consanguineous family. Here, we report two affected siblings with compound heterozygous PIGN mutations [c.808T >C (p.Ser270Pro) and c.963G >A] showing congenital anomalies, developmental delay, hypotonia, epilepsy, and progressive cerebellar atrophy. The c.808C >T mutation altered an evolutionarily conserved amino acid residue (Ser270), while reverse transcription-PCR and sequencing demonstrated that c.963G >A led to aberrant splicing, in which two mutant transcripts with premature stop codons (p.Ala322Valfs*24 and p.Glu308Glyfs*2) were generated. Expression of GPI-anchored proteins such as CD16 and CD24 on granulocytes from affected siblings was significantly decreased, and expression of the GPI-anchored protein CD59 in PIGN-knockout human embryonic kidney 293 cells was partially or hardly restored by transient expression of p.Ser270Pro and p.Glu308Glyfs*2 mutants, respectively, suggesting severe and complete loss of PIGN activity. Our findings confirm that developmental delay, hypotonia, and epilepsy combined with congenital anomalies are common phenotypes of PIGN mutations and add progressive cerebellar atrophy to this clinical spectrum.


Asunto(s)
Anomalías Múltiples/genética , Mutación , Fosfotransferasas/genética , Encéfalo/anomalías , Antígeno CD24/metabolismo , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/patología , Niño , Preescolar , Anomalías Congénitas/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Femenino , Proteínas Ligadas a GPI/metabolismo , Heterocigoto , Humanos , Masculino , Hipotonía Muscular/genética , Linaje , Receptores de IgG/metabolismo
19.
J Hum Genet ; 59(5): 292-5, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24621584

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder mostly caused by MECP2 mutations. We identified a de novo WDR45 mutation, which caused a subtype of neurodegeneration with brain iron accumulation, in a patient showing clinically typical RTT. The mutation (c.830+1G>A) led to aberrant splicing in lymphoblastoid cells. Sequential brain magnetic resonance imaging demonstrated that iron deposition in the globus pallidus and the substantia nigra was observed as early as at 11 years of age. Because the patient showed four of the main RTT diagnostic criteria, WDR45 should be investigated in patients with RTT without MECP2 mutations.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/genética , Hierro/metabolismo , Mutación , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Adolescente , Alelos , Empalme Alternativo , Encéfalo/patología , Análisis Mutacional de ADN , Exoma , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Imagen por Resonancia Magnética , Síndrome de Rett/diagnóstico
20.
Epilepsia ; 55(7): 994-1000, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24888894

RESUMEN

OBJECTIVE: De novo SCN8A mutations have been reported in patients with epileptic encephalopathy. Herein we report seven patients with de novo heterozygous SCN8A mutations, which were found in our comprehensive genetic analysis (target capture or whole-exome sequencing) for early onset epileptic encephalopathies (EOEEs). METHODS: A total of 163 patients with EOEEs without mutations in known genes, including 6 with malignant migrating partial seizures in infancy (MMPSI), and 60 with unclassified EOEEs, were analyzed by target capture (28 samples) or whole-exome sequencing (135 samples). RESULTS: We identified de novo SCN8A mutations in 7 patients: 6 of 60 unclassified EOEEs (10.0%), and one of 6 MMPSI cases (16.7%). The mutations were scattered through the entire gene: four mutations were located in linker regions, two in the fourth transmembrane segments, and one in the C-terminal domain. The type of the initial seizures was variable including generalized tonic-clonic, atypical absence, partial, apneic attack, febrile convulsion, and loss of tone and consciousness. Onset of seizures was during the neonatal period in two patients, and between 3 and 7 months of age in five patients. Brain magnetic resonance imaging (MRI) showed cerebellar and cerebral atrophy in one and six patients, respectively. All patients with SCN8A missense mutations showed initially uncontrollable seizures by any drugs, but eventually one was seizure-free and three were controlled at the last examination. All patients showed developmental delay or regression in infancy, resulting in severe intellectual disability. SIGNIFICANCE: Our data reveal that SCN8A mutations can cause variable phenotypes, most of which can be diagnosed as unclassified EOEEs, and rarely as MMPSI. Together with previous reports, our study further indicates that genetic testing of SCN8A should be considered in children with unclassified severe epilepsy.


Asunto(s)
Mutación Missense/genética , Canal de Sodio Activado por Voltaje NAV1.6/genética , Espasmos Infantiles/diagnóstico , Espasmos Infantiles/genética , Adolescente , Niño , Preescolar , Diagnóstico Precoz , Electroencefalografía/métodos , Epilepsia/complicaciones , Epilepsia/diagnóstico , Epilepsia/genética , Femenino , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/etiología , Discapacidad Intelectual/genética , Masculino , Fenotipo , Espasmos Infantiles/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA