Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Pers Med ; 10(4)2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33333793

RESUMO

BACKGROUND: Pediatric epileptic encephalopathy and severe neurological disorders comprise a group of heterogenous diseases. We used whole-exome sequencing (WES) to identify genetic defects in pediatric patients. METHODS: Patients with refractory seizures using ≥2 antiepileptic drugs (AEDs) receiving one AED and having neurodevelopmental regression or having severe neurological or neuromuscular disorders with unidentified causes were enrolled, of which 54 patients fulfilled the inclusion criteria, were enrolled, and underwent WES. RESULTS: Genetic diagnoses were confirmed in 24 patients. In the seizure group, KCNQ2, SCN1A, TBCID 24, GRIN1, IRF2BPL, MECP2, OSGEP, PACS1, PIGA, PPP1CB, SMARCA4, SUOX, SZT2, UBE3A, 16p13.11 microdeletion, [4p16.3p16.1(68,345-7,739,782)X1, 17q25.1q25.3(73,608,322-81,041,938)X3], and LAMA2 were identified. In the nonseizure group, SCN2A, SPTBN2, DMD, and FBN1 were identified. Ten novel mutations were identified. The recurrent genes included SCN1A, KCNQ2, and TBCID24. Male pediatric patients had a significantly higher (57% vs. 29%; p < 0.05, odds ratio = 3.18) yield than their female counterparts. Seventeen genes were identified from the seizure groups, of which 82% were rare genetic etiologies for childhood seizure and did not appear recurrently in the case series. CONCLUSIONS: Wide genetic variation was identified for severe childhood seizures by WES. WES had a high yield, particularly in male infantile patients.

2.
Sci Rep ; 10(1): 13375, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32770121

RESUMO

Pediatric epilepsy caused by KCNQ2 mutations can manifest benign familial neonatal convulsions (BFNC) to neonatal-onset epileptic encephalopathy (EE). Patients might manifest mild to profound neurodevelopmental disabilities. We analysed c.853C > A (P285T) and three mutations that cause KCNQ2 protein changes in the 247 position: c.740C > T (S247L), c.740C > A (S247X), and c.740C > G (S247W). S247L, S247W, and P285T cause neonatal-onset EE and poor neurodevelopmental outcomes; S247X cause BFNC and normal outcome. We investigated the phenotypes correlated with human embryonic kidney 293 (HEK293) cell functional current changes. More cell-current changes and a worse conductance curve were present in the homomeric transfected S247X than in S247L, S247W, and P285T. But in the heteromeric channel, S247L, S247W and P285T had more current impairments than did S247X. The protein expressions of S247X were nonfunctional. The outcomes were most severe in S247L and S247W, and severity was correlated with heteromeric current. Current changes were more significant in cells with homomeric S247X, but currents were "rescued" after heteromeric transfection of KCNQ2 and KCNQ3. This was not the case in cells with S247L, S247W. Our findings support that homomeric current changes are common in KCNQ2 neonatal-onset EE and KCNQ2 BFNC; however, heteromeric functional current changes are correlated with long-term neurodevelopmental outcomes.


Assuntos
Canal de Potássio KCNQ2/metabolismo , Mutação com Perda de Função/genética , Transtornos do Neurodesenvolvimento/genética , Substituição de Aminoácidos/genética , Pré-Escolar , Eletroencefalografia , Epilepsia Neonatal Benigna/genética , Epilepsia Neonatal Benigna/metabolismo , Epilepsia Neonatal Benigna/fisiopatologia , Células HEK293 , Humanos , Lactente , Recém-Nascido , Canal de Potássio KCNQ2/genética , Transtornos do Neurodesenvolvimento/metabolismo , Transtornos do Neurodesenvolvimento/fisiopatologia
3.
Cells ; 9(5)2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32455934

RESUMO

Nonsyndromic hearing loss (NSHL) is of great clinical importance, and mutations in the GJB2 gene and the encoded human CONNEXIN 26 (CX26) protein play important roles in the genetic pathogenesis. The CX26 p.R184Q mutation was shown to be a dominant-negative effect in our previous study. Previously, we also demonstrated that zebrafish Cx30.3 is orthologous to human CX26. In the present study, we established transgenic zebrafish models with mutated Cx30.3 specifically expressed in the supporting cells of zebrafish inner ears driven by the agr2 promoter, to demonstrate and understand the mechanism by which the human CX26 R.184 mutation causes NSHL. Our results indicated that significant structural changes in the inner ears of transgenic lines with mutations were measured and compared to wild-type zebrafish. Simultaneously, significant alterations of transgenic lines with mutations in swimming behavior were analyzed with the zebrafish behavioral assay. This is the first study to investigate the functional results of the CX26 p.R184Q mutation with in vivo disease models. Our work supports and confirms the pathogenic role of the CX26 p.R184Q mutation in NSHL, with a hypothesized mechanism of altered interaction among amino acids in the connexins.


Assuntos
Conexina 26/genética , Conexinas/genética , Surdez/genética , Mutação/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Comportamento Animal , Bioensaio , Conexinas/química , Modelos Animais de Doenças , Orelha Interna/metabolismo , Orelha Interna/patologia , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Proteínas Mutantes/química
4.
Int J Mol Sci ; 21(7)2020 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-32235586

RESUMO

A previous study indicated that mutations in the transmembrane protease serine 3 (TMPRSS3) gene, which encodes a transmembrane serine protease, cause nonsyndromic hearing loss (NSHL). This was the first description of a serine protease involved in hearing loss (HL). In Taiwan, however, data on the TMPRSS3 gene's association with NSHL is still insufficient. In this study, we described 10 mutations of TMPRSS3 genes found in 14 patients after screening 230 children with NSHL. The prevalence of the TMPRSS3 mutation appeared to be 6.09% (14/230). Of the 10 mutations, three were missense mutations: c.239G>A (p.R80H), c.551T>C (p.L184S), and 1253C>T (p.A418V); three were silent mutations, and four were mutations in introns. To determine the functional importance of TMPRSS3 mutations, we constructed plasmids carrying TMPRSS3 mutations of p.R80H, p.L184S, and p.A418V. TMPRSS3 function can be examined by secretory genetic assay for site-specific proteolysis (sGASP) and Xenopus oocyte expression system. Our results showed that p.R80H, p.L184S, and p.A418V TMPRSS3 mutations gave ratios of 19.4%, 13.2%, and 27.6%, respectively, via the sGASP system. Moreover, these three TMPRSS3 mutations failed to activate the epithelial sodium channel (ENaC) in the Xenopus oocyte expression system. These results indicate that the p.R80H, p.L184S, and p.A418V missense mutations of TMPRSS3 resulted in greatly diminishing the proteolytic activity of TMPRSS3. Our study provides information for understanding the importance of TMPRSS3 in the NSHL of Taiwanese children and provides a novel molecular explanation for the role of TMPRSS3 in HL.


Assuntos
Surdez/genética , Proteínas de Membrana/genética , Mutação , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Adolescente , Adulto , Animais , Criança , Surdez/epidemiologia , Humanos , Íntrons , Mutação de Sentido Incorreto , Mutação Puntual , Taiwan/epidemiologia , Xenopus , Adulto Jovem
5.
Mol Genet Genomic Med ; 7(7): e00816, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31199083

RESUMO

BACKGROUND: Epilepsy caused by a KCNQ2 gene mutation usually manifests as neonatal seizures during the first week of life. The genotypes and phenotypes of KCNQ2 mutations are noteworthy. METHODS: The KCNQ2 sequencings done were selected from 131 nonconsanguineous pediatric epileptic patients (age range: 2 days to 18 years) with nonlesional epilepsy. RESULTS: Seven (5%) index patients had verified KCNQ2 mutations: c.387+1 G>T (splicing), c.1741 C>T (p.Arg581*), c.740 C>T p.(Ser247Leu), c.853 C>A p.(Pro285Thr), c.860 C>T p.(Thr287Ile), c.1294 C>T p.(Arg432Cys), and c.1627 G>A p.(Val543Met). We found, after their paternity had been confirmed, that three patients had de novo p.(Ser247Leu), p.(Pro285Thr), and p.(Thr287Ile) mutations and neonatal-onset epileptic encephalopathy; however, their frequent seizures remitted after they turned 6 months old. Those with the c.387+1G>T (splicing), (p.Arg581*), and p.(Val543Met) mutations presented with benign familial neonatal convulsions. In addition to their relatives, 14 patients had documented KCNQ2 mutations, and 12 (86%) had neonatal seizures. The seizures of all five patients treated with oxcarbazepine remitted. CONCLUSION: KCNQ2-related epilepsy led to varied outcomes (from benign to severe) in our patients. KCNQ2 mutations accounted for 13% of patients with seizure onset before 2 months old in our study. KCNQ2 mutations can cause different phenotypes in children. p.(Pro 285Thr) is a novel mutation, and the p.(Pro 285Thr), p.(Ser247Leu), and p.(Thr287Ile) variants can cause neonatal-onset epileptic encephalopathy.


Assuntos
Epilepsia/genética , Canal de Potássio KCNQ2/genética , Mutação de Sentido Incorreto , Fenótipo , Adolescente , Ondas Encefálicas , Criança , Pré-Escolar , Epilepsia/patologia , Feminino , Humanos , Lactente , Masculino
6.
Int J Med Sci ; 14(12): 1251-1256, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29104481

RESUMO

Primary open-angle glaucoma (POAG) is one of the most important disease in ophthalmology with high prevalence and risk of irreversible blindness. If diagnosed before the age of 35, it is usually categorized as juvenile open-angle glaucoma (JOAG). The WDR36 gene is reckoned as one of the major causative genes of POAG, and had been studied to be related to the pathogenesis of POAG in the literature. We have selected 61 JOAG patients and 61 JOAG-free individuals, and by next-generation sequencing method, the WDR36 gene of the subjects were analyzed. We identified 26 variations exclusively in JOAG group. Among these 26 variations, there were 3 noteworthy variations. First, a novel variation c.460-650A>G was found in our study which might cause premature termination of splicing of the conserved domain in WDR36; second, c.1494+1111G>T (rs13178997) had significantly different frequency in our JOAG patients compared to the reference frequency on NCBI; third, a variation c.710+30C>T (rs10038177) was found in our study, which had already been reported to be related to high-pressure glaucoma. We offer the profile of WDR36 in JOAG in Taiwan population, and we suggest that WDR36 gene is involved in the pathogenesis of JOAG as a subordinate modifier gene.


Assuntos
Povo Asiático/genética , Proteínas do Olho/genética , Glaucoma de Ângulo Aberto/epidemiologia , Glaucoma de Ângulo Aberto/genética , Adolescente , Adulto , Idade de Início , Feminino , Testes Genéticos/métodos , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Polimorfismo de Nucleotídeo Único , Splicing de RNA/genética , Análise de Sequência de DNA , Taiwan/epidemiologia , Adulto Jovem
7.
Int J Med Sci ; 14(3): 246-256, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28367085

RESUMO

In a previous study, we identified a novel missense mutation, p.W77S, in the GJC3 gene encoding connexin30.2/connexin31.3 (CX30.2/CX31.3) from patients with hearing loss. The functional alteration of CX30.2/CX31.3 caused by the p.W77S mutant of GJC3 gene, however, remains unclear. In the current study, our result indicated that the p.W77 is localized at the second membrane-spanning segments (TM2) and near border of the E1 domain of the CX30.2/CX31.3 protein and highly conserved (Conseq score = 8~9) in all species. The p.W77S missense mutation proteins in the intracellular distribution are different CX30.2/CX31.3WT and an accumulation of the mutant protein in the endoplasmic reticulum (ER) of the HeLa cell. Furthermore, co-expression of WT and p.W77S mutant chimerae proteins showed that the heteromeric connexon accumulated in the cytoplasm, thereby impairing the WT proteins' expression in the cell membranes. In addition, we found that CX30.2/CX31.3W77S missense mutant proteins were degraded by lysosomes and proteosomes in the transfected HeLa cell. Based on these findings, we suggest that p.W77S mutant has a dominant negative effect on the formation and function of the gap junction. These results give a novel molecular elucidation for the mutation of GJC3 in the development of hearing loss.


Assuntos
Conexinas/química , Conexinas/genética , Surdez/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Relação Estrutura-Atividade , Conexinas/metabolismo , Surdez/patologia , Retículo Endoplasmático/química , Retículo Endoplasmático/genética , Junções Comunicantes/genética , Junções Comunicantes/patologia , Regulação da Expressão Gênica , Células HeLa , Humanos , Lisossomos/genética , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/metabolismo , Domínios Proteicos , Proteólise
8.
J Child Neurol ; 32(8): 704-711, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28399683

RESUMO

We analyzed the KCNQ2 wild-type gene and 3 mutations to highlight the important association between the KCNQ2 phenotype and genotype. The clinical phenotypes of 3 mutations (p.E515D, p.V543 M, and p.R213Q) were compared. KCNQ2, wild-type, and mutant KCNQ2 alleles were transfected into HEK293 cells before whole-cell patch-clamp analysis. Neurodevelopmental outcomes were worst in patients with the p.R213Q mutation, better in patients with the p.E515D mutation, and best in patients with the novel p.V543 M mutation. The currents in p.E515D and in p.V543 M were significantly lower than in the wild type in homomeric and heteromeric transfected HEK293 cells ( P < .05). The opening threshold shifted to values that were more positive, and the maximal current induced by strong depolarization was higher in cells with the p.E515D and p.R213Q mutations. We provide evidence that genotype is involved in determining clinical phenotype, including the seizure frequency and outcome.


Assuntos
Epilepsia/genética , Canal de Potássio KCNQ3/genética , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Criança , Saúde da Família , Feminino , Genótipo , Células HEK293 , Humanos , Lactente , Masculino , Potenciais da Membrana/genética , Técnicas de Patch-Clamp , Fenótipo , Transfecção
9.
Pediatr Neonatol ; 58(4): 338-343, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28087260

RESUMO

BACKGROUND: We wanted to identify in children with epilepsy the factors associated with seizure control and recurrence after a 2-year remission. METHODS: We did a 5-year follow-up of epileptic children whose antiepileptic medication had been stopped. Bivariate and multivariate analyses were used to compare features of electroencephalograms (EEGs) and clinical findings. In this study, 43 patients with and 64 without a seizure recurrence (SR) were enrolled. RESULTS: Clinical features strongly associated with SR in the univariate analysis included a symptomatic etiology for seizures, a history of status epilepticus, treatment duration before stopping antiepileptic drugs, and abnormal EEG findings at the time of stopping antiepileptic drugs. CONCLUSION: We found that a history of status epilepticus, symptomatic partial epilepsy, treatment duration before stopping antiepileptic drugs, and an abnormal EEG when the medication was stopped are important predictors of SR. The risk factors of SR after discontinuing antiepileptic drugs have been investigated in several studies. However, a history of status epilepticus as a predictive factor is rarely mentioned.


Assuntos
Anticonvulsivantes/uso terapêutico , Epilepsia/tratamento farmacológico , Convulsões/tratamento farmacológico , Adolescente , Criança , Pré-Escolar , Eletroencefalografia , Epilepsia/etiologia , Feminino , Seguimentos , Humanos , Masculino , Análise Multivariada , Recidiva , Fatores de Risco , Convulsões/etiologia , Estado Epiléptico/complicações
10.
J Formos Med Assoc ; 116(9): 711-719, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28038823

RESUMO

BACKGROUND/PURPOSE: Pediatric epilepsy caused by a KCNQ2 gene mutation usually manifests as benign familial neonatal seizures (BFNS) during the 1st week of life. However, the exact mechanism, phenotype, and genotype of the KCNQ2 mutation are unclear. METHODS: We studied the KCNQ2 genotype from 75 nonconsanguineous patients with childhood epilepsy without an identified cause (age range: from 2 days to 18 years) and from 55 healthy adult controls without epilepsy. KCNQ2 mutation variants were transfected into HEK293 cells to investigate what functional changes they induced. RESULTS: Four (5%) of the patients had the E515D KCNQ2 mutation, which the computer-based PolyPhen algorithm predicted to be deleterious. Their seizure outcomes were favorable, but three had an intellectual disability. Two patients with E515D presented with continuous spikes and waves during slow-wave sleep (CSWS), and the other two presented with BFNS. We also analyzed 10 affected family members with the same KCNQ2 mutation: all had epilepsy (8 had BFNS and 2 had CSWS). A functional analysis showed that the recordings of the E515D currents were significantly different (p<0.05), which suggested that channels with KCNQ2 E515D variants are less sensitive to voltage and require stronger depolarization to reach opening probabilities than those with the wild type or N780T (a benign polymorphism). CONCLUSION: KCNQ2 mutations can cause various phenotypes in children: they lead to BFNS and CSWS. We hypothesize that patients with the KCNQ2 E515D mutation are susceptible to seizures.


Assuntos
Epilepsia Neonatal Benigna/genética , Canal de Potássio KCNQ2/genética , Mutação , Sono/fisiologia , Adolescente , Criança , Eletroencefalografia , Epilepsia Neonatal Benigna/fisiopatologia , Células HEK293 , Humanos , Lactente
11.
Pediatr Neonatol ; 56(6): 408-14, 2015 12.
Artigo em Inglês | MEDLINE | ID: mdl-26073370

RESUMO

BACKGROUND: Lenticulostriate vasculopathy is associated with various disorders, in particular cytomegalovirus infection, which can cause neurological consequences. We wanted to evaluate the association of lenticulostriate vasculopathy and cytomegalovirus infection. We retrospectively collected data on lenticulostriate vasculopathy from 858 neonatal ultrasonography scans. METHODS: Fifty-five patients with lenticulostriate vasculopathy were diagnosed. Lenticulostriate vasculopathy was classified as severe and mild according to the ultrasonographic findings. We analyzed gender, unilateral and bilateral lenticulostriate vasculopathy, mild and severe lenticulostriate vasculopathy, intrauterine growth retardation, and lenticulostriate vasculopathy associated with other brain malformations to determine whether they were correlated with cytomegalovirus infection. RESULTS: Neonatal cytomegalovirus infections correlated primarily with lenticulostriate vasculopathy that was associated with brain structure anomalies p < 0.0001, followed by severe lenticulostriate vasculopathy (p = 0.029). Cytomegalovirus urine polymerase chain reaction ratios were 69% for severe and 23% for mild lenticulostriate vasculopathy (p = 0.002; odds ratio = 7.33). Of 72 newborns with intrauterine growth retardation without lenticulostriate vasculopathy, 33 were analyzed for cytomegalovirus, of whom only one was positive, which was significantly different from the newborns with lenticulostriate vasculopathy (p = 0.003; odds ratio = 11.64). CONCLUSION: Lenticulostriate vasculopathy on neonatal ultrasonography is useful for predicting cytomegalovirus infection, particularly in severe lenticulostriate vasculopathy. When severe lenticulostriate vasculopathy is associated with a brain structure anomaly, cytomegalovirus infection should be considered. The outcomes for the cases in which cytomegalovirus infection was associated with other brain structure anomalies were significantly worse than the outcomes in cases associated with lenticulostriate vasculopathy only.


Assuntos
Doença Cerebrovascular dos Gânglios da Base/diagnóstico por imagem , Doença Cerebrovascular dos Gânglios da Base/virologia , Infecções por Citomegalovirus/complicações , Feminino , Humanos , Recém-Nascido , Masculino , Estudos Retrospectivos , Índice de Gravidade de Doença
12.
J Child Neurol ; 29(12): 1660-3, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24352160

RESUMO

The outcomes of children with cryptogenic seizures most probably arising from the frontal lobe are difficult to predict. We retrospectively collected data on 865 pediatric patients with epilepsy. In 78 patients with cryptogenic frontal lobe epilepsy, the age at first seizure was inversely correlated with the outcome, including the degree of intellectual disability/developmental delay (P = .002) and seizure frequency (P = .02) after adequate treatment. Intellectual disability was more prevalent in children with a first seizure at 0 to 3 years old (P = .002), and seizures were more frequent in those with a first seizure at 0 to 6 years old than at 7 to 16 years old (P = .026). For pediatric cryptogenic frontal lobe epilepsy, the age at first seizure is important and inversely correlated with outcome, including seizure frequency and intellectual disability.


Assuntos
Deficiências do Desenvolvimento/etiologia , Epilepsia do Lobo Frontal , Deficiência Intelectual/etiologia , Adolescente , Análise de Variância , Criança , Pré-Escolar , Eletroencefalografia , Epilepsia do Lobo Frontal/complicações , Epilepsia do Lobo Frontal/diagnóstico , Epilepsia do Lobo Frontal/psicologia , Feminino , Humanos , Lactente , Deficiência Intelectual/diagnóstico , Masculino , Pediatria , Prognóstico , Análise de Regressão , Estudos Retrospectivos
13.
Cell Biochem Biophys ; 66(2): 277-86, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23179405

RESUMO

Connexins (CXs), as a component of gap junction channel, are homologous four transmembrane-domain proteins, with numerous studies confirming their auditory functions. Among a cohort of patients having incurred non-syndromic hearing loss, we identified two novel missense mutations, p.R15G and p.L23H, in the GJC3 gene encoding CX30.2/CX31.3, as causally related to hearing loss in previous study. However, the functional alteration of CX30.2/CX31.3 caused by the mutant GJC3 gene remains unknown. In this study, we compared the intracellular distribution of mutant CX30.2/CX31.3 (p.R15G and p.L23H) with the wild-type (WT) protein in HeLa cells and the effect of the mutant protein had on those cells. Analytical results indicated that p.R15G and p.L23H mutant exhibited continuous staining along apposed cell membranes in the fluorescent localization assay, which is the same with the WT. Moreover, ATP release (hemichannel function) is less in HeLa cells carrying mutant GJC3 genes than those of WT expressing cells. We believe that although p.R15G and p.L23H mutants do not decrease the trafficking of CX proteins, mutations in GJC3 genes result in a loss of hemichannel function of CX30.2/CX31.3 protein, possibly causing hearing loss. Results of this study provide a novel molecular explanation for the role of GJC3 in hearing loss.


Assuntos
Conexinas/genética , Perda Auditiva/genética , Proteínas do Tecido Nervoso/genética , Sequência de Aminoácidos , Clonagem Molecular , Biologia Computacional , Conexinas/química , Bases de Dados Factuais , Células HeLa , Perda Auditiva/metabolismo , Perda Auditiva/patologia , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/química , Estrutura Terciária de Proteína , Alinhamento de Sequência
14.
PLoS One ; 6(6): e21473, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21731760

RESUMO

Dysfunctional gap junctions caused by GJB2 (CX26) and GJB6 (CX30) mutations are implicated in nearly half of nonsyndromic hearing loss cases. A recent study identified a heterozygous mutation, c.119C>T (p.A40V), in the GJB6 gene of patients with nonsyndromic hearing loss. However, the functional role of the mutation in hearing loss remains unclear. In this study, analyses of cell biology indicated that a p.A40V missense mutation of CX30 causes CX30 protein accumulation in the Golgi body rather than in the cytoplasmic membrane. The tet-on protein expression system was used for further study of mutant proteins in CX30 and CX30A40V co-expressions and in CX26 and CX30A40V co-expressions. The p.A40V missense mutation exerted a dominant negative effect on both normal CX30 and CX26, which impaired gap junction formation. Moreover, computer-assisted modeling suggested that this p.A40V mutation affects the intra molecular interaction in the hydrophobic core of Trp44, which significantly alters the efficiency of gap junction formation. These findings suggest that the p.A40V mutation in CX30 causes autosomal-dominant nonsyndromic hearing loss. These data provide a novel molecular explanation for the role of GJB6 in hearing loss.


Assuntos
Conexinas/genética , Perda Auditiva Neurossensorial/genética , Mutação de Sentido Incorreto/genética , Sequência de Aminoácidos , Biologia Computacional , Conexina 26 , Conexina 30 , Conexinas/química , Células HeLa , Humanos , Imuno-Histoquímica , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência
15.
Cell Biochem Biophys ; 61(1): 189-97, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21480002

RESUMO

Gap junctional intercellular communication has numerous functions, each of which meets the particular needs of organs, tissues, or groups of cells. Connexins (CXs) are homologous four-transmembrane-domain proteins that are the major components of gap junctions. CX30.2/CX31.3 (GJC3) is a relatively new member of the CX protein family. Until now, however, the functional characteristics of CX30.2/CX31.3 have been unclear. To elucidate the properties of CX30.2/CX31.3 channels, their subcellular localization in HeLa cells, their effectiveness in dye transfer, and function on channels were investigated. In the immunofluorescent assay, cells that express CX30.2/CX31.3-GFP exhibited continuous fluorescence along the apposed cell membranes, rather than punctated fluorescence in contacting membranes between two cells. Surprisingly, dyes that can be capable of being permeated by CX26 GJ, according to a scrape loading dye transfer assay in previous studies, are impermeated by CX30.2/CX31.3 GJ, suggesting a difference between the characteristics of CX30.2/CX31.3 GJ and CX26 GJ. Furthermore, a significant amount of ATP was released from the HeLa cells that stably expressed CX30.2/CX31.3, in a medium with low calcium ion concentration, suggesting a hemichannel-based function for CX30.2/CX31.3. Based on these findings, we suggest that CX30.2/CX31.3 shares functional properties with pannexin (hemi) channels rather than gap junction channels of other CXs.


Assuntos
Conexinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Trifosfato de Adenosina/metabolismo , Conexina 26 , Conexinas/genética , Conexinas/fisiologia , Corantes Fluorescentes/química , Junções Comunicantes/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/fisiologia
16.
Hum Genet ; 128(3): 303-13, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20593197

RESUMO

The crucial role of gap junctions, which are composed of connexin (CX) protein, in auditory functions has been confirmed by numerous studies. In this study, we investigate the prevalence and phenotype/genotype correlation of connexin (CX) gene family variants in a cohort of children with nonsyndromic hearing loss (HL). A total of 253 unrelated children with nonsyndromic HL were screened for the presence of variants in 6 genes of the CX gene family. The prevalence of CX gene variants in 253 patients was 19.7% (50/253). We found the frequency of a sloping audiometric configuration was significantly higher for children with GJB2 and GJB3 variants than for those with GJB4 and GJC3 variants (Adjusted OR = 4.89, p < 0.001). Conversely, the frequency of a flat audiometric configuration was significantly higher for children with GJB4 and GJC3 variants than for those with GJB2 and GJB3 variants (adjusted OR = 7.76, p < 0.001). The relative frequencies of multiplex families was significantly higher for children with GJB3 variants than for those with GJB2, GJB4, and GJC3 variants (Adjusted OR = 11.33, p = 0.003). Our results suggest the variants of GJC3, GJB4, and GJB3 may be the common genetic risk factor, after variants of GJB2, for the development of nonsyndromic HL in Taiwan. These data can be effectively applied to direct the clinical evaluation of children with CX gene variants.


Assuntos
Conexinas/genética , Variação Genética , Perda Auditiva/genética , Criança , Estudos de Coortes , Conexina 26 , Conexina 30 , Conexina 43/genética , Feminino , Estudos de Associação Genética , Testes Genéticos , Humanos , Masculino , Família Multigênica , Mutação , Proteínas do Tecido Nervoso/genética , Estudos Prospectivos , Taiwan
17.
Eur J Hum Genet ; 18(9): 1061-4, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20442751

RESUMO

Hearing impairment is the most common sensory disorder worldwide. In a recent study, the authors have shown that a heterozygous missense mutation, p.R184Q, in the connexin 26 (Cx26) is causally related to hearing loss. However, the functional change in the Cx26R184Q mutant remains unknown. This study compared the intracellular distribution and assembly of mutant Cx26R184Q with that of the wild-type (WT) Cx26 and Cx30WT in tet-on HeLa cells and the effect that the mutant protein had on those cells. Fluorescent localization assay of WT Cx26 showed the typical punctuate pattern of gap junction channel between neighboring expression cells. Conversely, the p.R184Q missense mutation resulted in accumulation of the Cx26 mutant protein in the Golgi apparatus rather than in the cytoplasmic membrane. Cx26R184Q coexpressed with either Cx26WT or Cx30WT showed perinuclear localization by bidirectional tet-on expression system, suggesting the impairment of the ability of both WT proteins to intracellular trafficking and targeting to the plasma membrane. Therefore, we proposed that Cx26R184Q has a dominant-negative effect on the function of WT Cx26 and Cx30.


Assuntos
Conexinas/genética , Genes Dominantes , Perda Auditiva/genética , Mutação , Sequência de Bases , Clonagem Molecular , Conexina 26 , Conexina 30 , Conexinas/metabolismo , Primers do DNA , Células HeLa , Humanos
18.
Int J Pediatr Otorhinolaryngol ; 74(5): 489-93, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20206386

RESUMO

OBJECTIVE: To determine whether variants of the TMIE gene are causes of nonsyndromic deafness in Taiwan. METHODS: A genetic survey was made from 370 individuals, with 250 nonsyndromic hearing loss and 120 normal hearing individuals. Genomic DNA was extracted from peripheral blood leukocytes and then subjected to PCR to amplify selected exons and flanking introns of the TMIE gene; the amplified products were screened for base variants by autosequence. Data from the two groups were then compared using Fisher's two-tailed exact test and Armitage's trend test. RESULTS: The analysis revealed 7 novel variants in the TMIE gene. Of the 7 variants, 5 variants were found in both nonsyndromic hearing loss and normal hearing group. Both allelic and genotype frequencies of these sequence changes did not differ significantly between patients and controls (P>0.05). However, a missense variant (c.257G>A) and one promoter variant (g.1-219A>T) were found in two patients with nonsyndromic hearing loss. Family study and microsatellite analysis found that c.257G>A variant is not inherited from his parents. The c.257G>A variant encodes a protein with glutamine at position 86 instead of arginine (p.R86Q), a residue that is conserved in mammals but different in fish, and predicted to be extracellular. CONCLUSIONS: Despite the fact that the frequency of TMIE variants in our study subjects was low, we suggested that c.257G>A (p.R86Q) variant is a de novo and may be as a risk factor for the development of hearing loss in Taiwanese.


Assuntos
Surdez/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Adolescente , Estudos de Casos e Controles , Criança , Pré-Escolar , Éxons , Frequência do Gene , Humanos , Repetições de Microssatélites , Linhagem , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Regiões Promotoras Genéticas/genética , Análise de Sequência de DNA , Taiwan , Adulto Jovem
19.
Hum Genet ; 127(5): 545-51, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20130915

RESUMO

Connexins (CXs), a large family of membrane proteins, are key components of gap junction channels. Among a cohort of patients with nonsyndromic hearing loss, we have recently identified three novel missense mutations in the GJA1 gene and GJA1 pseudogene (rhoGJA1) as likely being causally related to hearing loss. However, the functional alteration of CX43 caused by the mutations of GJA1 and rhoGJA1 gene remains unclear. This study compares the intracellular distribution and assembly of three CX43 mutants expressed in HeLa cells with their wild-type (WT) counterparts and the effects of the mutant proteins on those cells. Localization assay of WT CX43 reveals a typical punctuate fluorescence pattern of a gap junction channel between neighboring expression cells. Additionally, immunoblotting analysis of the transfectants confirms the production of mutant proteins, in which their distributions along appositional membranes are determined using immunofluorescent staining procedures. Furthermore, dye transfer assay results demonstrate that gap junctional intercellular communication is less in HeLa cells carrying mutant GJA1 or rhoGJA1 gene than in WT-expressing cells. The results of this study suggest that the three mutations in GJA1 or rhoGJA1 that we previously reported result in at least partial loss of normal functions carried out by CX43, which may form a basis for the mechanism contributing to hearing loss in patients.


Assuntos
Conexina 43/genética , Perda Auditiva/genética , Mutação , Pseudogenes , Western Blotting , Conexina 43/análise , Conexinas/genética , Proteínas do Olho/genética , Imunofluorescência , Corantes Fluorescentes , Células HeLa , Humanos , Imuno-Histoquímica , Isoquinolinas , Plasmídeos , Pseudogenes/genética , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Formos Med Assoc ; 109(1): 32-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20123584

RESUMO

BACKGROUND/PURPOSE: Mutations in the receptor tyrosine kinase RET gene are associated with Hirschsprung disease (HD), which is also known as congenital intestinal aganglionosis. We found an association with specific alleles in five single nucleotide polymorphism (SNP) sites of the RET gene in our HD patients. METHODS: We compared the association of specific RET SNP alleles in patients with severe GI disorders such as anorectal malformation (ARM) or pediatric intestinal pseudo-obstruction (IPO) to that in HD patients. Sixty-four HD, 23 ARM and 35 IPO patients were included. Genomic DNA extracted from blood samples was analyzed by polymerase chain reaction and DNA sequencing analysis. RESULTS: The allele distributions of all five RET SNPs in the HD patients deviated from those in the normal population (p < 0.05), whereas those of the ARM patients did not. The allele distributions of these RET SNPs in the IPO patients were all significantly different from those in the HD patients. Allele distributions of exon 2 and 13 in the IPO patients were also significantly different from those of the normal population. The frequencies of all the HD-predominant alleles were lower in the HD patients than the normal population, and were even lower in the IPO patients. CONCLUSION: This study strengthens the association of specific RET SNP alleles with typical HD in Taiwan.


Assuntos
Anormalidades do Sistema Digestório/genética , Doença de Hirschsprung/genética , Pseudo-Obstrução Intestinal/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Proto-Oncogênicas c-ret/genética , Alelos , DNA/genética , Éxons , Predisposição Genética para Doença , Genótipo , Hospitais Universitários , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Taiwan
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA