Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 110(8): 1356-1376, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37421948

RESUMO

By converting physical forces into electrical signals or triggering intracellular cascades, stretch-activated ion channels allow the cell to respond to osmotic and mechanical stress. Knowledge of the pathophysiological mechanisms underlying associations of stretch-activated ion channels with human disease is limited. Here, we describe 17 unrelated individuals with severe early-onset developmental and epileptic encephalopathy (DEE), intellectual disability, and severe motor and cortical visual impairment associated with progressive neurodegenerative brain changes carrying ten distinct heterozygous variants of TMEM63B, encoding for a highly conserved stretch-activated ion channel. The variants occurred de novo in 16/17 individuals for whom parental DNA was available and either missense, including the recurrent p.Val44Met in 7/17 individuals, or in-frame, all affecting conserved residues located in transmembrane regions of the protein. In 12 individuals, hematological abnormalities co-occurred, such as macrocytosis and hemolysis, requiring blood transfusions in some. We modeled six variants (p.Val44Met, p.Arg433His, p.Thr481Asn, p.Gly580Ser, p.Arg660Thr, and p.Phe697Leu), each affecting a distinct transmembrane domain of the channel, in transfected Neuro2a cells and demonstrated inward leak cation currents across the mutated channel even in isotonic conditions, while the response to hypo-osmotic challenge was impaired, as were the Ca2+ transients generated under hypo-osmotic stimulation. Ectopic expression of the p.Val44Met and p.Gly580Cys variants in Drosophila resulted in early death. TMEM63B-associated DEE represents a recognizable clinicopathological entity in which altered cation conductivity results in a severe neurological phenotype with progressive brain damage and early-onset epilepsy associated with hematological abnormalities in most individuals.


Assuntos
Encefalopatias , Deficiência Intelectual , Humanos , Encefalopatias/genética , Canais Iônicos/genética , Encéfalo , Deficiência Intelectual/genética , Fenótipo
2.
Am J Hum Genet ; 106(4): 549-558, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32169168

RESUMO

De novo variants (DNVs) cause many genetic diseases. When DNVs are examined in the whole coding regions of genes in next-generation sequencing analyses, pathogenic DNVs often cluster in a specific region. One such region is the last exon and the last 50 bp of the penultimate exon, where truncating DNVs cause escape from nonsense-mediated mRNA decay [NMD(-) region]. Such variants can have dominant-negative or gain-of-function effects. Here, we first developed a resource of rates of truncating DNVs in NMD(-) regions under the null model of DNVs. Utilizing this resource, we performed enrichment analysis of truncating DNVs in NMD(-) regions in 346 developmental and epileptic encephalopathy (DEE) trios. We observed statistically significant enrichment of truncating DNVs in semaphorin 6B (SEMA6B) (p value: 2.8 × 10-8; exome-wide threshold: 2.5 × 10-6). The initial analysis of the 346 individuals and additional screening of 1,406 and 4,293 independent individuals affected by DEE and developmental disorders collectively identified four truncating DNVs in the SEMA6B NMD(-) region in five individuals who came from unrelated families (p value: 1.9 × 10-13) and consistently showed progressive myoclonic epilepsy. RNA analysis of lymphoblastoid cells established from an affected individual showed that the mutant allele escaped NMD, indicating stable production of the truncated protein. Importantly, heterozygous truncating variants in the NMD(+) region of SEMA6B are observed in general populations, and SEMA6B is most likely loss-of-function tolerant. Zebrafish expressing truncating variants in the NMD(-) region of SEMA6B orthologs displayed defective development of brain neurons and enhanced pentylenetetrazole-induced seizure behavior. In summary, we show that truncating DNVs in the final exon of SEMA6B cause progressive myoclonic epilepsy.


Assuntos
Exoma/genética , Éxons/genética , Predisposição Genética para Doença/genética , Variação Genética/genética , Epilepsias Mioclônicas Progressivas/genética , Semaforinas/genética , Adolescente , Adulto , Alelos , Animais , Feminino , Heterozigoto , Humanos , Masculino , Degradação do RNAm Mediada por Códon sem Sentido/genética , Convulsões/genética , Adulto Jovem , Peixe-Zebra/genética
3.
Hum Genet ; 141(2): 283-293, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35031858

RESUMO

GRIA3 at Xq25 encodes glutamate ionotropic receptor AMPA type 3 (GluA3), a subunit of postsynaptic glutamate-gated ion channels mediating neurotransmission. Hemizygous loss-of-function (LOF) variants in GRIA3 cause a neurodevelopmental disorder (NDD) in male individuals. Here, we report a gain-of-function (GOF) variant at GRIA3 in a male patient. We identified a hemizygous de novo missense variant in GRIA3 in a boy with an NDD: c.1844C > T (p.Ala615Val) using whole-exome sequencing. His neurological signs, such as hypertonia and hyperreflexia, were opposite to those in previous cases having LOF GRIA3 variants. His seizures and hypertonia were ameliorated by carbamazepine, inhibiting glutamate release from presynapses. Patch-clamp recordings showed that the human GluA3 mutant (p.Ala615Val) had slower desensitization and deactivation kinetics. A fly line expressing a human GluA3 mutant possessing our variant and the Lurcher variant, which makes ion channels leaky, showed developmental defects, while one expressing a mutant possessing either of them did not. Collectively, these results suggest that p.Ala615Val has GOF effects. GRIA3 GOF variants may cause an NDD phenotype distinctive from that of LOF variants, and drugs suppressing glutamatergic neurotransmission may ameliorate this phenotype. This study should help in refining the clinical management of GRIA3-related NDDs.


Assuntos
Carbamazepina/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Mutação com Ganho de Função , Transtornos do Neurodesenvolvimento/tratamento farmacológico , Transtornos do Neurodesenvolvimento/genética , Receptores de AMPA/genética , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados , Pré-Escolar , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Humanos , Masculino , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Transtornos do Neurodesenvolvimento/metabolismo , Técnicas de Patch-Clamp , Fenótipo , Receptores de AMPA/química , Receptores de AMPA/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Genet Med ; 24(12): 2453-2463, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36305856

RESUMO

PURPOSE: Cerebellar hypoplasia and atrophy (CBHA) in children is an extremely heterogeneous group of disorders, but few comprehensive genetic studies have been reported. Comprehensive genetic analysis of CBHA patients may help differentiating atrophy and hypoplasia and potentially improve their prognostic aspects. METHODS: Patients with CBHA in 176 families were genetically examined using exome sequencing. Patients with disease-causing variants were clinically evaluated. RESULTS: Disease-causing variants were identified in 96 of the 176 families (54.5%). After excluding 6 families, 48 patients from 42 families were categorized as having syndromic associations with CBHA, whereas the remaining 51 patients from 48 families had isolated CBHA. In 51 patients, 26 aberrant genes were identified, of which, 20 (76.9%) caused disease in 1 family each. The most prevalent genes were CACNA1A, ITPR1, and KIF1A. Of the 26 aberrant genes, 21 and 1 were functionally annotated to atrophy and hypoplasia, respectively. CBHA+S was more clinically severe than CBHA-S. Notably, ARG1 and FOLR1 variants were identified in 2 families, leading to medical treatments. CONCLUSION: A wide genetic and clinical diversity of CBHA was revealed through exome sequencing in this cohort, which highlights the importance of comprehensive genetic analyses. Furthermore, molecular-based treatment was available for 2 families.


Assuntos
Exoma , Malformações do Sistema Nervoso , Criança , Humanos , Exoma/genética , Mutação , Malformações do Sistema Nervoso/genética , Atrofia/genética , Receptor 1 de Folato/genética , Cinesinas
5.
Hum Mutat ; 42(1): 66-76, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33131106

RESUMO

We report heterozygous CELF2 (NM_006561.3) variants in five unrelated individuals: Individuals 1-4 exhibited developmental and epileptic encephalopathy (DEE) and Individual 5 had intellectual disability and autistic features. CELF2 encodes a nucleocytoplasmic shuttling RNA-binding protein that has multiple roles in RNA processing and is involved in the embryonic development of the central nervous system and heart. Whole-exome sequencing identified the following CELF2 variants: two missense variants [c.1558C>T:p.(Pro520Ser) in unrelated Individuals 1 and 2, and c.1516C>G:p.(Arg506Gly) in Individual 3], one frameshift variant in Individual 4 that removed the last amino acid of CELF2 c.1562dup:p.(Tyr521Ter), possibly resulting in escape from nonsense-mediated mRNA decay (NMD), and one canonical splice site variant, c.272-1G>C in Individual 5, also probably leading to NMD. The identified variants in Individuals 1, 2, 4, and 5 were de novo, while the variant in Individual 3 was inherited from her mosaic mother. Notably, all identified variants, except for c.272-1G>C, were clustered within 20 amino acid residues of the C-terminus, which might be a nuclear localization signal. We demonstrated the extranuclear mislocalization of mutant CELF2 protein in cells transfected with mutant CELF2 complementary DNA plasmids. Our findings indicate that CELF2 variants that disrupt its nuclear localization are associated with DEE.


Assuntos
Proteínas CELF , Epilepsia , Deficiência Intelectual , Proteínas do Tecido Nervoso , Proteínas CELF/genética , Epilepsia/genética , Feminino , Heterozigoto , Humanos , Deficiência Intelectual/genética , Proteínas do Tecido Nervoso/genética , Sinais de Localização Nuclear/genética , Proteínas de Ligação a RNA/genética
6.
Epilepsy Behav ; 117: 107515, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33610462

RESUMO

PURPOSES: The purpose of the study was to investigate the positive and negative effects of perampanel (PER) treatment on the psychiatric and behavioral symptoms in patients with epilepsy and to evaluate factors associated with the psychiatric and behavioral changes caused by PER. METHODS: We retrospectively examined medical records of patients with epilepsy treated with PER in the Department of Psychiatry, Epilepsy Center, Nishiniigata Chuo National Hospital. Multiple regression analyses were performed with the psychiatric and behavioral prognoses as dependent variables and clinical characteristics of the patients as independent variables. RESULTS: Thirty-two of 135 patients (23.7%) had psychiatric and behavioral deterioration after the initiation of PER, whereas 22 patients (16.3%) showed improvement in psychiatric and behavioral symptoms after PER administration. Etiology of structural abnormalities, concomitant use of nitrazepam, and comorbidities of irritability and depression were significantly associated with increasing incidence of psychiatric and behavioral deterioration. Concomitant use of carbamazepine was significantly associated with decreasing incidence of psychiatric and behavioral deterioration. Suppression of awareness-impaired seizures by PER, concomitant use of carbamazepine, and comorbidities of insomnia, anxiety, and amnesia were significantly associated with an increasing incidence of psychiatric and behavioral improvement. Improvements in psychiatric symptoms by PER were associated with a reduction in the use of psychotropic drugs. In particular, about 1/4 of benzodiazepines had been discontinued. CONCLUSIONS: Perampanel therapy may aggravate or even ameliorate psychiatric and behavioral symptoms in patients with epilepsy. The psychiatric and behavioral prognoses after administration of PER vary depending on the type of psychiatric and behavioral comorbidities in patients with epilepsy. Psychiatric and behavioral symptoms may improve in patients with successful suppression of seizures by PER. Additionally, combination therapy consisting of PER and carbamazepine may be associated with good outcomes of psychiatric and behavioral symptoms in patients with epilepsy.


Assuntos
Anticonvulsivantes , Epilepsia , Adulto , Anticonvulsivantes/uso terapêutico , Sintomas Comportamentais , Epilepsia/complicações , Epilepsia/tratamento farmacológico , Humanos , Nitrilas , Piridonas/uso terapêutico , Estudos Retrospectivos , Resultado do Tratamento
7.
Epilepsy Behav ; 116: 107644, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33549477

RESUMO

PURPOSE: The present study evaluated whether patients with epilepsy who received both levetiracetam (LEV) and perampanel (PER) therapy showed side effects of irritability. The study also examined the relationship between patient characteristics and irritability when it occurred as a side effect. METHODS: We retrospectively examined medical records of 98 patients with epilepsy who were treated with both LEV and PER at the Department of Psychiatry in the Epilepsy Center of Nishiniigata Chuo National Hospital in Japan. We performed multiple regression analyses with the presence/absence of irritability due to LEV or PER as the dependent variables and clinical characteristics of the patients as independent variables. RESULTS: LEV and PER caused irritability in 7 and 17 of 98 patients, respectively. LEV- and PER-related irritability did not occur in the same patients. A logistic multiple regression analysis revealed that EEG findings of temporal focal epileptic discharge were significantly associated with increased incidence of irritability due to LEV. LEV-related irritability decreased significantly with higher dosages of LEV. Another logistic multiple regression analysis revealed that a psychiatric comorbidity of irritability and EEG findings of nontemporal focal epileptic discharge were significantly associated with increased incidence of irritability due to PER. CONCLUSIONS: LEV and PER cause irritability in different patient groups. Additionally, irritability as a side effect was present only at low dosages of LEV, but PER tended to cause irritability even at high dosages.


Assuntos
Epilepsia , Piracetam , Anticonvulsivantes/efeitos adversos , Epilepsia/tratamento farmacológico , Humanos , Japão , Levetiracetam/uso terapêutico , Nitrilas , Piracetam/efeitos adversos , Piridonas , Estudos Retrospectivos
8.
J Hum Genet ; 64(4): 313-322, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30655572

RESUMO

Casein kinase 2 (CK2) is a serine threonine kinase ubiquitously expressed in eukaryotic cells and involved in various cellular processes. In recent studies, de novo variants in CSNK2A1 and CSNK2B, which encode the subunits of CK2, have been identified in individuals with intellectual disability syndrome. In this study, we describe four patients with neurodevelopmental disorders possessing de novo variants in CSNK2A1 or CSNK2B. Using whole-exome sequencing, we detected two de novo variants in CSNK2A1 in two unrelated Japanese patients, a novel variant c.571C>T, p.(Arg191*) and a recurrent variant c.593A>G, p.(Lys198Arg), and two novel de novo variants in CSNK2B in Japanese and Malaysian patients, c.494A>G, p.(His165Arg) and c.533_534insGT, p.(Pro179Tyrfs*49), respectively. All four patients showed mild to profound intellectual disabilities, developmental delays, and various types of seizures. This and previous studies have found a total of 20 CSNK2A1 variants in 28 individuals with syndromic intellectual disability. The hotspot variant c.593A>G, p.(Lys198Arg) was found in eight of 28 patients. Meanwhile, only five CSNK2B variants were identified in five individuals with neurodevelopmental disorders. We reviewed the previous literature to verify the phenotypic spectrum of CSNK2A1- and CSNK2B-related syndromes.


Assuntos
Caseína Quinase II/genética , Deficiências do Desenvolvimento/genética , Convulsões/genética , Adolescente , Criança , Pré-Escolar , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Humanos , Lactente , Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Masculino , Mutação , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/fisiopatologia , Linhagem , Convulsões/complicações , Convulsões/fisiopatologia , Sequenciamento do Exoma
9.
Mol Genet Metab ; 124(1): 64-70, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29599076

RESUMO

We first characterized PPT1 and TPP1 enzymes in dried blood spots (DBS), plasma/serum, and leukocytes/lymphocytes using neuronal ceroid lipofuscinosis (NCL) 1 and 2 patients and control subjects. PPT1 enzyme had only one acid form in control DBS, plasma/serum, and leukocytes/lymphocytes and showed deficient activities in these samples from NCL 1 patients. Conversely, TPP1 enzymes in control DBS and leukocytes/lymphocytes consisted of two forms, an acidic form and a neutral form, whereas serum TPP1 enzyme had only a neutral form. In control subjects, the optimal pH of PPT1 enzyme in DBS, plasma/serum, and leukocytes/lymphocytes was 4.5 to 5.0 in the acidic form, whereas TPP1 enzyme in control DBS and leukocytes/lymphocytes was pH 4.5 and 6.5, respectively. In NCL 1 and 2, both PPT1 and TPP1 enzyme activities in DBS, plasma, and leukocytes/lymphocytes were markedly reduced in acidic pH, whereas heterozygotes of NCL 1 and 2 in the acidic form showed intermediate activities between patients and control subjects. In neutral conditions, pH 6.0, the PPT1 enzyme activities in NCL 1 patients showed rather higher residual activities and intermediate activities in heterozygotes in NCL 1, which was probably caused by mutated proteins in three cases with NCL 1 patients. TPP1 enzyme activities at neutral pH 6.5 to 7.0 in DBS and leukocytes/lymphocytes showed higher enzyme activities in NCL 2 patients and heterozygotes. The reason for the increases of neutral TPP1 enzyme activities at pH 6.5 to 7.0 in NCL 2 DBS and leukocytes/lymphocytes, is obscure, but possibly caused by secondary activation of neutral TPP1 enzyme due to the absence of the acidic form. Interestingly, TPP1 activity in serum only consisted of a neutral form, no acidic form, and was not deficient in any NCL 2 patient. Therefore, we can diagnose NCL 1 patients by plasma/serum enzyme assay of PPT1, but not diagnose NCL 2 by serum TPP1 enzyme assay. A pilot study of newborn screening of NCL 1 and 2 has been established by more than 1000 newborn DBS assays. Using this assay system, we will be able to perform newborn screening of NCL 1 and 2 by DBS.


Assuntos
Aminopeptidases/sangue , Dipeptidil Peptidases e Tripeptidil Peptidases/sangue , Leucócitos/química , Proteínas de Membrana/sangue , Triagem Neonatal/métodos , Lipofuscinoses Ceroides Neuronais/diagnóstico , Serina Proteases/sangue , Tioléster Hidrolases/sangue , Adulto , Criança , Pré-Escolar , Teste em Amostras de Sangue Seco/métodos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Recém-Nascido , Masculino , Mutação , Projetos Piloto , Tripeptidil-Peptidase 1
10.
J Hum Genet ; 63(3): 263-270, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29339779

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/genética , Doenças Cerebelares/diagnóstico , Doenças Cerebelares/genética , Proteínas do Citoesqueleto/genética , Genes Recessivos , Mutação , Retinose Pigmentar/diagnóstico , Retinose Pigmentar/genética , Adolescente , Idade de Início , Alelos , Atrofia , Linhagem Celular , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Linhagem , Fenótipo , Sequenciamento Completo do Genoma
11.
Clin Genet ; 94(6): 538-547, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30280376

RESUMO

N-methyl-d-aspartate (NMDA) receptors are glutamate-activated ion channels that are widely distributed in the central nervous system and essential for brain development and function. Dysfunction of NMDA receptors has been associated with various neurodevelopmental disorders. Recently, a de novo recurrent GRIN2D missense variant was found in two unrelated patients with developmental and epileptic encephalopathy. In this study, we identified by whole exome sequencing novel heterozygous GRIN2D missense variants in three unrelated patients with severe developmental delay and intractable epilepsy. All altered residues were highly conserved across vertebrates and among the four GluN2 subunits. Structural consideration indicated that all three variants are probably to impair GluN2D function, either by affecting intersubunit interaction or altering channel gating activity. We assessed the clinical features of our three cases and compared them to those of the two previously reported GRIN2D variant cases, and found that they all show similar clinical features. This study provides further evidence of GRIN2D variants being causal for epilepsy. Genetic diagnosis for GluN2-related disorders may be clinically useful when considering drug therapy targeting NMDA receptors.


Assuntos
Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/genética , Epilepsia/diagnóstico , Epilepsia/genética , Variação Genética , Receptores de N-Metil-D-Aspartato/genética , Adolescente , Alelos , Sequência de Aminoácidos , Encéfalo/anormalidades , Criança , Pré-Escolar , Eletroencefalografia , Feminino , Genótipo , Humanos , Masculino , Simulação de Dinâmica Molecular , Mutação , Conformação Proteica , Receptores de N-Metil-D-Aspartato/química , Relação Estrutura-Atividade
12.
Brain ; 140(9): 2322-2336, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-29050398

RESUMO

De novo in-frame deletions and duplications in the SPTAN1 gene, encoding the non-erythrocyte αII spectrin, have been associated with severe West syndrome with hypomyelination and pontocerebellar atrophy. We aimed at comprehensively delineating the phenotypic spectrum associated with SPTAN1 mutations. Using different molecular genetic techniques, we identified 20 patients with a pathogenic or likely pathogenic SPTAN1 variant and reviewed their clinical, genetic and imaging data. SPTAN1 de novo alterations included seven unique missense variants and nine in-frame deletions/duplications of which 12 were novel. The recurrent three-amino acid duplication p.(Asp2303_Leu2305dup) occurred in five patients. Our patient cohort exhibited a broad spectrum of neurodevelopmental phenotypes, comprising six patients with mild to moderate intellectual disability, with or without epilepsy and behavioural disorders, and 14 patients with infantile epileptic encephalopathy, of which 13 had severe neurodevelopmental impairment and four died in early childhood. Imaging studies suggested that the severity of neurological impairment and epilepsy correlates with that of structural abnormalities as well as the mutation type and location. Out of seven patients harbouring mutations outside the α/ß spectrin heterodimerization domain, four had normal brain imaging and three exhibited moderately progressive brain and/or cerebellar atrophy. Twelve of 13 patients with mutations located within the spectrin heterodimer contact site exhibited severe and progressive brain, brainstem and cerebellar atrophy, with hypomyelination in most. We used fibroblasts from five patients to study spectrin aggregate formation by Triton-X extraction and immunocytochemistry followed by fluorescence microscopy. αII/ßII aggregates and αII spectrin in the insoluble protein fraction were observed in fibroblasts derived from patients with the mutations p.(Glu2207del), p.(Asp2303_Leu2305dup) and p.(Arg2308_Met2309dup), all falling in the nucleation site of the α/ß spectrin heterodimer region. Molecular modelling of the seven SPTAN1 amino acid changes provided preliminary evidence for structural alterations of the A-, B- and/or C-helices within each of the mutated spectrin repeats. We conclude that SPTAN1-related disorders comprise a wide spectrum of neurodevelopmental phenotypes ranging from mild to severe and progressive. Spectrin aggregate formation in fibroblasts with mutations in the α/ß heterodimerization domain seems to be associated with a severe neurodegenerative course and suggests that the amino acid stretch from Asp2303 to Met2309 in the α20 repeat is important for α/ß spectrin heterodimer formation and/or αII spectrin function.


Assuntos
Encefalopatias/genética , Encéfalo/patologia , Proteínas de Transporte/genética , Epilepsia/genética , Proteínas dos Microfilamentos/genética , Adolescente , Atrofia/complicações , Atrofia/patologia , Encéfalo/anormalidades , Encefalopatias/complicações , Proteínas de Transporte/metabolismo , Células Cultivadas , Criança , Pré-Escolar , Progressão da Doença , Epilepsia/complicações , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas dos Microfilamentos/metabolismo , Modelos Moleculares , Mutação , Transtornos do Neurodesenvolvimento/complicações , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Agregação Patológica de Proteínas/metabolismo , Adulto Jovem
13.
BMC Pulm Med ; 17(1): 40, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28212655

RESUMO

BACKGROUND: Pulmonary alveolar proteinosis (PAP) is a rare lung disease characterized by surfactant accumulation, and is caused by disruption of granulocyte/macrophage colony-stimulating factor (GM-CSF) signaling. Abnormalities in CSF2 receptor alpha (CSF2RA) were reported to cause pediatric hereditary PAP. We report here the first case of CSF2RA-mutated, elderly-onset hereditary (h) PAP. CASE PRESENTATION: The patient developed dyspnea on exertion, and was diagnosed with PAP at the age of 77 years, based on findings from chest computed tomography scan and bronchoalveolar lavage. She tested negative for GM-CSF autoantibodies, with no underlying disease. Her serum GM-CSF level was elevated (91.3 pg/mL), indicating GM-CSF signaling impairment and genetic defects in the GM-CSF receptor. GM-CSF-stimulated phosphorylation in signal transducer and activator of transcription 5 (STAT5) was not observed, and GM-CSF-Rα expression was defective in her blood cells. Genetic screening revealed a homozygous, single-base C > T mutation at nt 508-a nonsense mutation that yields a stop codon (Q170X)-in exon 7 of CSF2RA. High-resolution analysis of single nucleotide polymorphism array confirmed a 22.8-Mb loss of heterozygosity region in Xp22.33p22.11, encompassing the CSF2RA gene. She was successfully treated with whole lung lavage (WLL), which reduced the serum levels of interleukin (IL)-2, IL-5, and IL-17, although IL-3 and M-CSF levels remained high. CONCLUSIONS: This is the first known report of elderly-onset hPAP associated with a CSF2RA mutation, which caused defective GM-CSF-Rα expression and impaired signaling. The analyses of serum cytokine levels during WLL suggested that GM-CSF signaling might be compensated by other signaling pathways, leading to elderly-onset PAP.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Proteinose Alveolar Pulmonar/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Idade de Início , Idoso , Autoanticorpos/sangue , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Feminino , Humanos , Interleucinas/sangue , Macrófagos Alveolares/imunologia , Mutação , Proteinose Alveolar Pulmonar/diagnóstico por imagem , Radiografia Torácica , Transdução de Sinais , Tomografia Computadorizada por Raios X
14.
Am J Hum Genet ; 93(3): 496-505, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23993195

RESUMO

Heterotrimeric G proteins, composed of α, ß, and γ subunits, can transduce a variety of signals from seven-transmembrane-type receptors to intracellular effectors. By whole-exome sequencing and subsequent mutation screening, we identified de novo heterozygous mutations in GNAO1, which encodes a Gαo subunit of heterotrimeric G proteins, in four individuals with epileptic encephalopathy. Two of the affected individuals also showed involuntary movements. Somatic mosaicism (approximately 35% to 50% of cells, distributed across multiple cell types, harbored the mutation) was shown in one individual. By mapping the mutation onto three-dimensional models of the Gα subunit in three different complexed states, we found that the three mutants (c.521A>G [p.Asp174Gly], c.836T>A [p.Ile279Asn], and c.572_592del [p.Thr191_Phe197del]) are predicted to destabilize the Gα subunit fold. A fourth mutant (c.607G>A), in which the Gly203 residue located within the highly conserved switch II region is substituted to Arg, is predicted to impair GTP binding and/or activation of downstream effectors, although the p.Gly203Arg substitution might not interfere with Gα binding to G-protein-coupled receptors. Transient-expression experiments suggested that localization to the plasma membrane was variably impaired in the three putatively destabilized mutants. Electrophysiological analysis showed that Gαo-mediated inhibition of calcium currents by norepinephrine tended to be lower in three of the four Gαo mutants. These data suggest that aberrant Gαo signaling can cause multiple neurodevelopmental phenotypes, including epileptic encephalopathy and involuntary movements.


Assuntos
Epilepsia/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Predisposição Genética para Doença , Mutação/genética , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Cálcio/metabolismo , Criança , Pré-Escolar , Eletroencefalografia , Epilepsia/patologia , Epilepsia/fisiopatologia , Exoma/genética , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Humanos , Lactente , Imageamento por Ressonância Magnética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenótipo , Transporte Proteico , Análise de Sequência de DNA , Transdução de Sinais/genética
15.
Ann Neurol ; 78(3): 375-86, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26018084

RESUMO

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.


Assuntos
Encéfalo/patologia , Malformações do Desenvolvimento Cortical do Grupo II/genética , Mutação/genética , Serina-Treonina Quinases TOR/genética , Adolescente , Adulto , Criança , Feminino , Células HEK293 , Humanos , Masculino , Malformações do Desenvolvimento Cortical/diagnóstico , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical do Grupo II/diagnóstico
16.
Hepatol Res ; 46(2): 166-73, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26287990

RESUMO

AIM: We evaluated the performance of automated tumor-feeder detection (AFD) software using cone-beam computed tomography technology in identifying tumor-feeders of extrahepatic collaterals. METHODS: AFD was prospectively used in superselective transarterial chemoembolization (TACE) or embolization (TAE) of extrahepatic collaterals for 29 hepatocellular carcinomas and one liver metastasis (mean tumor diameter ± standard deviation, 28 ± 15.6 mm) in 25 patients. The detectability of extrahepatic tumor-feeders with non-selective digital subtraction angiography (DSA) and AFD was evaluated and compared using a χ(2) -test. Tumor response of target lesions in each patient at 2-3 months after treatment was evaluated using the modified Response Evaluation Criteria in Solid Tumors. Complications were also evaluated. RESULTS: Of 46 tumor-feeders, non-selective DSA and AFD could identify 26 and 44, respectively (P < 0.001). Regarding the origin of tumor-feeders, both non-selective DSA and AFD could identify 14 of 15, six of seven and two of two tumor-feeders of the right inferior phrenic, omental and right renal capsular artery, respectively. In the cystic and left gastric or right colic artery, AFD could identify 13 of 13 and nine of nine tumor-feeders but non-selective DSA could identify only three of 13 and one of nine, respectively (P < 0.001). Complete response was obtained in 15 patients, partial response in six, stable disease in three and progressive disease in one. No severe complications developed except for right pleural effusion after embolization of the right inferior phrenic artery. CONCLUSION: AFD has a sufficient ability to identify extrahepatic tumor-feeders and may improve the safety and local effects of TACE/TAE of extrahepatic collaterals.

17.
Hepatol Res ; 46(9): 890-8, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26662842

RESUMO

AIM: To retrospectively evaluate the detectability of tumor recurrence with computed tomography (CT) and magnetic resonance imaging (MRI) after superselective conventional transcatheter arterial chemoembolization (cTACE) for hepatocellular carcinoma (HCC). METHODS: The detectability of tumor recurrence with CT and gadoxetate disodium-enhanced MRI obtained within 30 days (mean, 16.9 ± 10.1) were compared in 38 patients with recurrent HCC after superselective cTACE. Tumor recurrence was divided into local and distant recurrence. Local recurrence was also divided into intratumoral and peritumoral recurrence. RESULTS: Tumor recurrence (maximum diameter, 19.7 ± 10.1 mm) was demonstrated by images 12.4 ± 11.7 months after cTACE. CT could depict 16 (76.2%) of 21 intratumoral recurrences in 12 patients and 14 (53.8%) of 26 peritumoral recurrences in 11, as well as 39 (55.7%) of 70 distant recurrences in 15 (75%) of 20 patients. Arterial phase MRI could depict 20 (95.2%) of 21 intratumoral recurrences in 14 patients and all 26 (100%) peritumoral recurrences in 21, as well as 60 (85.7%) distant recurrences in all 20 (100%) patients. The detectability of tumor recurrence with MRI was significantly higher than that with CT (P = 0.00549). On MRI, pseudolesions were observed in five (13.2%) patients and artifacts in the arterial phase in five (13.2%). Regarding the diagnostic performance, CT was superior to MRI in two (5.3%) patients and MRI was superior to CT in 19 (50%). They were almost equal in 17 (44.7%). CONCLUSION: The detectability of tumor recurrence after superselective cTACE with gadoxetate disodium-enhanced MRI was superior to that of CT.

18.
J Hum Genet ; 60(4): 167-73, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25631096

RESUMO

Recent progress in genetic analysis reveals that a significant proportion of cryptogenic epileptic encephalopathies are single-gene disorders. Mutations in numerous genes for early-onset epileptic encephalopathies have been rapidly identified, including in SPTAN1, which encodes α-II spectrin. The aim of this review is to delineate SPTAN1 encephalopathy as a distinct clinical syndrome. To date, a total of seven epileptic patients with four different in-frame SPTAN1 mutations have been identified. The major clinical features of SPTAN1 mutations include epileptic encephalopathy with hypsarrhythmia, no visual attention, acquired microcephaly, spastic quadriplegia and severe intellectual disability. Brainstem and cerebellar atrophy and cerebral hypomyelination, as observed by magnetic resonance imaging, are specific hallmarks of this condition. A milder variant is characterized by generalized epilepsy with pontocerebellar atrophy. Only in-frame SPTAN1 mutations in the last two spectrin repeats in the C-terminal region lead to dominant negative effects and these specific phenotypes. The last two spectrin repeats are required for α/ß spectrin heterodimer associations and the mutations can alter heterodimer formation between the two spectrins. From these data we suggest that SPTAN1 encephalopathy is a distinct clinical syndrome owing to specific SPTAN1 mutations. It is important that this syndrome is recognized by pediatric neurologists to enable proper diagnostic work-up for patients.


Assuntos
Encefalopatias/diagnóstico , Encefalopatias/genética , Proteínas de Transporte/genética , Epilepsia/diagnóstico , Epilepsia/genética , Proteínas dos Microfilamentos/genética , Mutação , Idade de Início , Diagnóstico Diferencial , Eletroencefalografia , Genes Dominantes , Estudos de Associação Genética , Genótipo , Humanos , Imageamento por Ressonância Magnética , Fenótipo , Espectrina/genética , Espectrina/metabolismo , Síndrome
19.
Epilepsia ; 56(9): e121-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26140313

RESUMO

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.


Assuntos
Mutação/genética , Proteínas do Tecido Nervoso/genética , Canais de Potássio/genética , Espasmos Infantis/genética , Encéfalo/patologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Eletroencefalografia , Humanos , Lactente , Imageamento por Ressonância Magnética , Canais de Potássio Ativados por Sódio
20.
Epilepsia ; 56(6): 841-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25864721

RESUMO

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.


Assuntos
Encefalopatias/genética , Epilepsia/genética , Predisposição Genética para Doença/genética , Hipercinese/genética , Mutação de Sentido Incorreto/genética , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/genética , Transtorno de Movimento Estereotipado/genética , Adolescente , Encefalopatias/complicações , Criança , Pré-Escolar , Análise Mutacional de DNA , Eletroencefalografia , Epilepsia/complicações , Feminino , Humanos , Hipercinese/complicações , Imageamento por Ressonância Magnética , Masculino , Transtorno de Movimento Estereotipado/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA