Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Hum Mol Genet ; 28(24): 4103-4112, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31628468

RESUMO

ELMOD3, an ARL2 GTPase-activating protein, is implicated in causing hearing impairment in humans. However, the specific role of ELMOD3 in auditory function is still far from being elucidated. In the present study, we used the CRISPR/Cas9 technology to establish an Elmod3 knockout mice line in the C57BL/6 background (hereinafter referred to as Elmod3-/- mice) and investigated the role of Elmod3 in the cochlea and auditory function. Elmod3-/- mice started to exhibit hearing loss from 2 months of age, and the deafness progressed with aging, while the vestibular function of Elmod3-/- mice was normal. We also observed that Elmod3-/- mice showed thinning and receding hair cells in the organ of Corti and much lower expression of F-actin cytoskeleton in the cochlea compared with wild-type mice. The deafness associated with the mutation may be caused by cochlear hair cells dysfunction, which manifests with shortening and fusion of inner hair cells stereocilia and progressive degeneration of outer hair cells stereocilia. Our finding associates Elmod3 deficiencies with stereocilia dysmorphologies and reveals that they might play roles in the actin cytoskeleton dynamics in cochlear hair cells, and thus relate to hearing impairment.


Assuntos
Surdez/enzimologia , Proteínas Ativadoras de GTPase/deficiência , Proteínas Ativadoras de GTPase/metabolismo , Perda Auditiva/enzimologia , Estereocílios/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Cóclea/enzimologia , Cóclea/metabolismo , Citoesqueleto/metabolismo , Surdez/genética , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/genética , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/fisiologia , Perda Auditiva/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Estereocílios/enzimologia
2.
Genet Med ; 21(12): 2744-2754, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31273342

RESUMO

PURPOSE: To determine the genetic etiology of deafness in a family (HN-SD01) with autosomal dominant nonsyndromic hearing loss (NSHL). METHODS: Stepwise genetic analysis was performed on family HN-SD01, including hotspot variant screening, exome sequencing, virtual hearing loss gene panel, and genome-wide linkage analysis. Targeted region sequencing was used to screen ABCC1 in additional cases. Cochlear expression of Abcc1 was evaluated by messenger RNA (mRNA) and protein levels. Computational prediction, immunofluorescence, real-time quantitative polymerase chain reaction, and flow cytometry were conducted to uncover functional consequences of candidate variants. RESULTS: Stepwise genetic analysis identified a heterozygous missense variant, ABCC1:c.1769A>G (p.Asn590Ser), cosegregating with phenotype in HN-SD01. Screening of ABCC1 in an additional 217 cases identified candidate pathogenic variants c.692G>A (p.Gly231Asp) in a sporadic case and c.887A>T (p.Glu296Val) in a familial proband. Abcc1 expressed in stria vascularis and auditory nerve of mouse cochlea. Immunofluorescence showed p.Asn590Ser distributed in cytomembrane and cytoplasm, while wild type was shown only in cytomembrane. Besides, it generated unstable mRNA and decreased efflux capacity of ABCC1. CONCLUSION: Stepwise genetic analysis is efficient to analyze the genetic etiology of NSHL. Variants in ABCC1 are linked with NSHL and suggest an important role of extruding pumps in maintaining cochlea function.


Assuntos
Surdez/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Adolescente , Adulto , Idoso , Animais , China , Cóclea/metabolismo , Surdez/etiologia , Surdez/metabolismo , Exoma , Família , Feminino , Ligação Genética , Testes Genéticos , Genótipo , Perda Auditiva/genética , Heterozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Análise de Sequência de DNA/métodos , Sequenciamento do Exoma
3.
Hum Genet ; 137(4): 329-342, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29713870

RESUMO

Autosomal dominant nonsyndromic hearing loss (ADNSHL) is a highly genetically heterogeneous disorder. Up to date only approximately 37 ADNSHL-causing genes have been identified. The goal of this study was to determine the causative gene in a five-generation Chinese family with ADNSHL. A Chinese family was ascertained. Simultaneously, two affected individuals and one normal hearing control from the family were analyzed by whole exome capture sequencing. To assess the functional effect of the identified variant, in-vitro studies were performed. novel missense variant, c.512A>G (p.His171Arg) in exon 8 of the ELMO domain-containing 3 (ELMOD3) gene, was identified as a causative variant in this family affected by late-onset and progressive ADNSHL. The variant was validated by Sanger sequencing and found to co-segregate with the phenotype within the pedigree and was absent in 500 ethnically matched unrelated normal hearing control subjects. To our knowledge, this is the first report of a family with ADNSHL caused by ELMOD3 mutation. Western blots and immunofluorescence staining demonstrated that p.His171Arg resulted in abnormal expression levels of ELMOD3 and abnormal subcellular localization. Furthermore, the analysis of the stability of the wild-type (WT) and mutant ELMOD3 protein shows that the decay of p.His171Arg is faster than that of the WT, suggesting a shorter halflife of the c.512A > G variant. A novel variant in the ELMOD3 gene, encoding a member of the engulfment and cell motility (ELMO) family of GTPase-activating proteins, was identified for the first time as responsible for ADNSHL.


Assuntos
Proteínas Ativadoras de GTPase/genética , Perda Auditiva Neurossensorial/genética , Adulto , Sequência de Aminoácidos/genética , Movimento Celular/genética , China/epidemiologia , Exoma/genética , Feminino , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Masculino , Mutação , Linhagem , Fenótipo
4.
J Hum Genet ; 63(6): 723-730, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29559740

RESUMO

X-linked inheritance is very rare and is estimated to account for only 1-5% of all nonsyndromic hearing loss cases. We found a multiplex family from China segregating with X-linked nonsyndromic hearing loss. After exclusive analysis of 10 common variations of three hearing loss-related genes, GJB2, mtDNA12srRNA and SLC26A4, a novel truncated variant of SMPX, c.87dupA (p.Gly30Argfs*12) (NCBI ClinVar Submission ID: SUB3136126), was identified by whole-exome sequencing. This variant was co-segregated with hearing loss in the entire family and was absent in 576 unrelated ethnically and geographically matched controls. We also detected a single nucleotide variation in two male controls with normal hearing, SMPX c.55A>G (p.Asn19Asp), which has been annotated as a rare variant in the Single Nucleotide Polymorphism (dbSNP) (rs759552778) and Exome Aggregation Consortium (ExAC) databases. This study has enriched the mutation spectrum of the SMPX gene.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/genética , Perda Auditiva Neurossensorial/genética , Proteínas Musculares/genética , Mutação , Adulto , Povo Asiático/genética , Estudos de Casos e Controles , China , Bases de Dados Genéticas , Feminino , Perda Auditiva Neurossensorial/etnologia , Humanos , Masculino , Linhagem , Polimorfismo de Nucleotídeo Único , Sequenciamento do Exoma , Adulto Jovem
5.
J Psychiatr Res ; 143: 113-122, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34487988

RESUMO

NRXN1 is involved in synaptogenesis and have been implicated in Autism spectrum disorders. However, many rare inherited missense variants of NRXN1 have not been thoroughly evaluated. Here, functional analyses in vitro and in Drosophila of three NRXN1 missense mutations, Y282H, L893V, and I1135V identified in ASD patients in our previous study were performed. Our results showed these three mutations interfered protein degradation compared with NRXN1-WT protein. Expressing human NRXN1 in Drosophila could lead to abnormal circadian rhythm and sleep behavior, and three mutated proteins caused milder phenotypes, indicating the mutations may change the function of NRXN1 slightly. These findings highlight the functional role of rare NRXN1 missense variants identified in autism patients, and provide clues for us to better understand the pathogenesis of abnormal circadian rhythm and sleep behavior of other organisms, including humans.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Animais , Transtorno do Espectro Autista/genética , Proteínas de Ligação ao Cálcio/genética , Drosophila/genética , Humanos , Mutação de Sentido Incorreto , Moléculas de Adesão de Célula Nervosa/genética , Proteólise , Sono/genética
6.
Front Genet ; 10: 639, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379920

RESUMO

Autosomal recessive non-syndromic hearing loss (ARNSHL) is a highly heterogeneous disease involving more than 70 pathogenic genes. However, most ARNSHL families have small-sized pedigrees with limited genetic information, rendering challenges for the molecular diagnosis of these patients. Therefore, we attempted to establish a strategy for identifying deleterious variants associated with ARNSHL by applying proband whole-exome sequencing (proband-WES). Aside from desiring to improve molecular diagnostic rates, we also aimed to search for novel deafness genes shared by patients with similar phenotype, making up for the deficiency of small ARNSHL families. In this study, 48.5% (16/33) families were detected the pathogenic variants in eight known deafness genes, including 10 novel variants identified in TMPRSS3 (MIM 605551), MYO15A (MIM 602666), TMC1 (MIM 606706), ADGRV1 (MIM 602851), and PTPRQ (MIM 603317). Apart from six novel variants with a truncating effect (nonsense, deletion, insertion, and splice-site), four novel missense variants were not found in 200 unrelated control population by using Sanger sequencing. It is important to note that none of novel genes were shared across different pedigrees, indicating that a larger sample size might be needed. Proband-WES is a cost-effective and precise way of identifying causative variants in nuclear families with ARNSHL. This economical strategy may be appropriated as a clinical application to provide molecular diagnostics, genetic counseling, and individualized health maintenance measures for patients with ARNSHL at hearing clinics.

7.
Gene ; 704: 113-120, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-30974196

RESUMO

Usher syndrome (USH) is a clinically common autosomal recessive disorder characterized by retinitis pigmentosa (RP) and sensorineural hearing loss with or without vestibular dysfunction. In this study, we identified a Hunan family of Chinese descent with two affected members clinically diagnosed with Usher syndrome type 3 (USH3) displaying hearing, visual acuity, and olfactory decline. Whole-exome sequencing (WES) identified a nonsense variant in ABHD12 gene that was confirmed to be segregated in this family by Sanger sequencing and exhibited a recessive inheritance pattern. In this family, two patients carried homozygous variant in the ABHD12 (NM_015600: c.249C>G). Mutation of ABHD12, an enzyme that hydrolyzes an endocannabinoid lipid transmitter, caused incomplete PHARC syndrome, as demonstrated in previous reports. Therefore, we also conducted a summary based on variants in ABHD12 in PHARC patients, and in PHARC patients showing that there was no obvious correlation between the genotype and phenotype. We believe that this should be considered during the differential diagnosis of USH. Our findings predicted the potential function of this gene in the development of hearing and vision loss, particularly with regard to impaired signal transmission, and identified a novel nonsense variant to expand the variant spectrum in ABHD12.


Assuntos
Códon sem Sentido , Monoacilglicerol Lipases/genética , Síndromes de Usher/genética , Adulto , Idoso , Ataxia/genética , Ataxia/patologia , Catarata/genética , Catarata/patologia , Criança , Família , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Homozigoto , Humanos , Masculino , Linhagem , Fenótipo , Polineuropatias/genética , Polineuropatias/patologia , Retinose Pigmentar/genética , Retinose Pigmentar/patologia , Síndromes de Usher/patologia , Sequenciamento do Exoma
8.
Int J Pediatr Otorhinolaryngol ; 115: 114-119, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30368370

RESUMO

OBJECTIVE: To evaluate the accuracy and validity of our protocol for prenatal diagnosis and genetic counseling in high-risk families at a clinic. METHODS: Fifteen unrelated families with recessive nonsyndromic hearing loss (NSHL) in their family history and a positive attitude towards prenatal diagnosis were recruited in the present study. According to genetic information for each family, Sanger sequencing, fluorescence polymerase chain reaction (PCR)-based congenital deafness gene detection kit and multiple PCR-based target gene capture and high-throughput sequencing were used. Genetic counseling was offered to all participating families by genetic counselors and otologists. Prenatal diagnosis was provided to families with detected pathogenic mutations and who were expected to participate in subsequent prenatal diagnosis. RESULTS: In this study, confirmed pathogenic mutations were detected in eight families, who were defined as high-risk families. These families all participated in prenatal diagnosis with positive attitudes. One novel variant (c.1687dupA) in the SLC264 gene was detected in a family. Through genetic counseling, the recurrence probability of NSHL in fetuses was 25% in six families, 0% in one family, and 50% in one family. The results of fetal DNA detection showed that one fetal variant was wild type, three were heterozygous mutations in SLC26A4, and one was a compound heterozygous mutation in SLC26A4. Two variants were heterozygous mutations in GJB2, and one was a homozygous mutation in GJB2. According to the test results for fetal DNA, prenatal diagnosis found that six fetuses had normal hearing, whereas two fetuses suffered from NSHL. After birth, six infants predicted to have normal hearing passed a newborn hearing screening test and two infants predicted to have NSHL were diagnosed with NSHL and received cochlear implants. CONCLUSION: Our protocol for prenatal diagnosis and genetic counseling provides detailed information that can assist couples in high-risk families in preparing for infant arrival and future family planning. For the affected neonates, prenatal diagnosis and genetic counseling achieve an "early screening, early diagnosis, early intervention" strategy.


Assuntos
Surdez/diagnóstico , Aconselhamento Genético/métodos , Diagnóstico Pré-Natal/métodos , Conexinas/genética , Análise Mutacional de DNA , Surdez/genética , Feminino , Audição , Testes Auditivos , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Homozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Linhagem , Reação em Cadeia da Polimerase/métodos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA