Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
BMC Med Genomics ; 17(1): 32, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38254107

RESUMO

BACKGROUND: Mutations in MPZL2, the characteristic genetic etiology of autosomal recessive deafness loci 111 (DFNB111), cause non-syndromic and moderate sensorineural hearing loss. METHODS: In this study, we analyzed the phenotype and genotype of eight pedigrees consisting of 10 hearing loss patients with bi-allelic pathogenic or likely pathogenic variants in MPZL2. These patients were identified from a 3272 Chinese patient cohort who underwent genetic testing. RESULTS: Apart from symmetrical and moderate sensorineural hearing loss, the MPZL2-related phenotype was characterized by progressive hearing loss with variation in the onset age (congenital defect to onset at the young adult stage). We determined that in the Chinese population, the genetic load of MPZL2 defects was 0.24% (8/3272) in patients diagnosed with hearing loss and 7.02% (8/114) in patients diagnosed with hereditary moderate sensorineural hearing loss caused by STRC, OTOA, OTOG, OTOGL, TECTA, MPZL2 and others. Three known MPZL2 variants (c.220C > T (p.Gln74*), c.68delC (p.Pro23Leufs*2), c.463delG (p.Ala155Leufs*10)) and a novel start loss variant (c.3G > T (p.Met1?)) were identified. MPZL2 c.220C > T was identified as the hotspot variant in the Chinese population and even in East Asia compared with c.72delA (p.Ile24Metfs*22) in European and West Asia through allele frequency. CONCLUSIONS: We concluded that apart from moderate HL, progressive HL is another character of MPZL2-related HL. No specified variant was verified for the progression of HL, the penetrance and expressivity cannot be determined yet. A novel MPZL2 variant at the start codon was identified, enriching the variant spectrum of MPZL2. The hotspot variants of MPZL2 vary in different ethnicities. This study provides valuable data for the diagnosis, prognosis evaluation and genetic counseling of patients with moderate sensorineural hearing loss related to MPZL2.


Assuntos
Surdez , Perda Auditiva Neurossensorial , Humanos , Adulto Jovem , Povo Asiático/genética , Moléculas de Adesão Celular , China , Surdez/etnologia , Surdez/genética , Perda Auditiva Neurossensorial/etnologia , Perda Auditiva Neurossensorial/genética , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana
3.
BMC Med Genomics ; 15(1): 241, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36401330

RESUMO

Pathogenic variants in MYO15A are known to cause autosomal recessive nonsyndromic hearing loss (ARNSHL), DFNB3. We have previously reported on one ARNSHL family including two affected siblings and identified MYO15A c.5964+3G > A and c.8375 T > C (p.Val2792Ala) as the possible deafness-causing variants. Eight year follow up identified one new affected individual in this family, who also showed congenital, severe to profound sensorineural hearing loss. By whole exome sequencing, we identified a new splice-site variant c.5531+1G > C (maternal allele), in a compound heterozygote with previously identified missense variant c.8375 T > C (p.Val2792Ala) (paternal allele) in MYO15A as the disease-causing variants. The new affected individual underwent unilateral cochlear implantation at the age of 1 year, and 5 year follow-up showed satisfactory speech and language outcomes. Our results further indicate that MYO15A-associated hearing loss is good candidates for cochlear implantation, which is in accordance with previous report. In light of our findings and review of the literatures, 58 splice-site variants in MYO15A are correlated with a severe deafness phenotype, composed of 46 canonical splice-site variants and 12 non-canonical splice-site variants.


Assuntos
Surdez , Perda Auditiva , Humanos , Linhagem , Miosinas/genética , Surdez/genética , Perda Auditiva/genética , Fenótipo , Família , Genótipo
4.
Gene Expr Patterns ; 43: 119229, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34968768

RESUMO

BACKGROUND: IFNLR1 has been recently identified to be related to autosomal dominant nonsyndromic sensorineural hearing loss (ADNSHL). It is reported to be expressed in the inner ear of mice and the lateral line of zebrafish. However, it remains unclear how defects in this gene lead to hearing loss. OBJECTIVES: To elucidate the global gene expression changes in zebrafish when the expression of ifnlr1 is downregulated. METHODS: Transcriptome analysis was performed on ifnlr1 morpholino knockdown zebrafish and the control zebrafish using RNA-seq technology. RESULTS: The results show that 262 differentially expressed genes (DEGs) were up-regulated while 146 DEGs were down-regulated in the E4I4-Mo zebrafish larvae compared to the control-Mo. Six pathways were significantly enriched, including steroid biosynthesis pathway, adipocytokine signaling pathway, cytokine-cytokine receptor interaction pathway, p53 signaling pathway, AGE-RAGE signaling pathway in diabetic complications, and terpenoid backbone biosynthesis pathway. Among them, three pathways (steroid biosynthesis pathway, cytokine-cytokine receptor interaction pathway and p53 signaling pathway) are immune-associated. CONCLUSIONS: The transcriptome analysis results contribute to the groundwork for future research on the pathogenesis of IFNLR1-associated hearing loss.


Assuntos
Transcriptoma , Peixe-Zebra , Animais , Citocinas , Perfilação da Expressão Gênica , Imunidade , Receptores de Citocinas/genética , Esteroides , Proteína Supressora de Tumor p53/genética , Peixe-Zebra/genética
5.
Am J Hum Genet ; 105(4): 803-812, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31564438

RESUMO

Concurrent hearing and genetic screening of newborns is expected to play important roles not only in early detection and diagnosis of congenital deafness, which triggers intervention, but also in predicting late-onset and progressive hearing loss and identifying individuals who are at risk of drug-induced HL. Concurrent hearing and genetic screening in the whole newborn population in Beijing was launched in January 2012. This study included 180,469 infants born in Beijing between April 2013 and March 2014, with last follow-up on February 24, 2018. Hearing screening was performed using transiently evoked otoacoustic emission (TEOAE) and automated auditory brainstem response (AABR). For genetic testing, dried blood spots were collected and nine variants in four genes, GJB2, SLC26A4, mtDNA 12S rRNA, and GJB3, were screened using a DNA microarray platform. Of the 180,469 infants, 1,915 (1.061%) were referred bilaterally or unilaterally for hearing screening; 8,136 (4.508%) were positive for genetic screening (heterozygote, homozygote, or compound heterozygote and mtDNA homoplasmy or heteroplasmy), among whom 7,896 (4.375%) passed hearing screening. Forty (0.022%) infants carried two variants in GJB2 or SLC26A4 (homozygote or compound heterozygote) and 10 of those infants passed newborn hearing screening. In total, 409 (0.227%) infants carried the mtDNA 12S rRNA variant (m.1555A>G or m.1494C>T), and 405 of them passed newborn hearing screening. In this cohort study, 25% of infants with pathogenic combinations of GJB2 or SLC26A4 variants and 99% of infants with an m.1555A>G or m.1494C>T variant passed routine newborn hearing screening, indicating that concurrent screening provides a more comprehensive approach for management of congenital deafness and prevention of ototoxicity.


Assuntos
Testes Genéticos/métodos , Perda Auditiva/diagnóstico , Pequim , Teste em Amostras de Sangue Seco , Feminino , Predisposição Genética para Doença , Humanos , Recém-Nascido , Masculino
6.
Nat Commun ; 9(1): 3833, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30237406

RESUMO

Fast radio bursts (FRBs), bright transients with millisecond durations at ∼GHz and typical redshifts probably >0.8, are likely to be gravitationally lensed by intervening galaxies. Since the time delay between images of strongly lensed FRB can be measured to extremely high precision because of the large ratio ∼109 between the typical galaxy-lensing delay time [Formula: see text] (10 days) and the width of bursts [Formula: see text] (ms), we propose strongly lensed FRBs as precision probes of the universe. We show that, within the flat ΛCDM model, the Hubble constant H0 can be constrained with a ~0.91% uncertainty from 10 such systems probably observed with the square kilometer array (SKA) in <30 years. More importantly, the cosmic curvature can be model independently constrained to a precision of ∼0.076. This constraint can directly test the validity of the cosmological principle and break the intractable degeneracy between the cosmic curvature and dark energy.

7.
Biomed Res Int ; 2018: 5370802, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29850532

RESUMO

Hereditary nonsyndromic hearing loss is extremely heterogeneous. Mutations in the POU class 4 transcription factor 3 (POU4F3) are known to cause autosomal dominant nonsyndromic hearing loss linked to the loci of DFNA15. In this study, we describe a pathogenic missense mutation in POU4F3 in a four-generation Chinese family (6126) with midfrequency, progressive, and postlingual autosomal dominant nonsyndromic hearing loss (ADNSHL). By combining targeted capture of 129 known deafness genes, next-generation sequencing, and bioinformatic analysis, we identified POU4F3 c.602T>C (p.Leu201Pro) as the disease-causing variant. This variant cosegregated with hearing loss in other family members but was not detected in 580 normal controls or the ExAC database and could be classified as a "pathogenic variant" according to the American College of Medical Genetics and Genomics guidelines. We conclude that POU4F3 c.602T>C (p.Leu201Pro) is related to midfrequency hearing loss in this family. Routine examination of POU4F3 is necessary for the genetic diagnosis of midfrequency hearing loss.


Assuntos
Povo Asiático/genética , Perda Auditiva Neurossensorial/genética , Proteínas de Homeodomínio/genética , Mutação de Sentido Incorreto/genética , Fator de Transcrição Brn-3C/genética , Adulto , Idoso , Sequência de Aminoácidos , Sequência de Bases , Pré-Escolar , Análise Mutacional de DNA , Família , Feminino , Proteínas de Homeodomínio/química , Humanos , Pessoa de Meia-Idade , Linhagem , Fator de Transcrição Brn-3C/química
8.
J Med Genet ; 55(5): 298-306, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29453195

RESUMO

Background Hereditary sensorineural hearing loss is a genetically heterogeneous disorder. Objectives This study was designed to explore the genetic etiology of deafness in a large Chinese family with autosomal dominant, nonsyndromic, progressive sensorineural hearing loss (ADNSHL). Methods Whole exome sequencing and linkage analysis were performed to identify pathogenic mutation. Inner ear expression of Ifnlr1 was investigated by immunostaining in mice. ifnlr1 Morpholino knockdown Zebrafish were constructed to explore the deafness mechanism. Results We identified a cosegregating heterozygous missense mutation, c.296G>A (p.Arg99His) in the gene encoding interferon lambda receptor 1 (IFNLR1) - a protein that functions in the Jak/ STAT pathway- are associated with ADNSHL Morpholino knockdown of ifnlr1 leads to a significant decrease in hair cells and non-inflation of the swim bladder in late-stage zebrafish, which can be reversed by injection with normal Zebrafish ifnlr1 mRNA. Knockdown of ifnlr1 in zebrafish causes significant upregulation of cytokine receptor family member b4 (interleukin-10r2), jak1, tyrosine kinase 2, stat3, and stat5b in the Jak1/STAT3 pathway at the mRNA level. ConclusionIFNLR1 function is required in the auditory system and that IFNLR1 mutations are associated with ADNSHL. To the best of our knowledge, this is the first study implicating an interferon lambda receptor in auditory function.


Assuntos
Predisposição Genética para Doença , Perda Auditiva Neurossensorial/genética , Receptores de Citocinas/genética , Receptores de Interferon/genética , Animais , Técnicas de Silenciamento de Genes , Ligação Genética , Perda Auditiva Neurossensorial/fisiopatologia , Heterozigoto , Humanos , Janus Quinase 1/genética , Camundongos , Morfolinas , Mutação de Sentido Incorreto/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais , Sequenciamento do Exoma , Peixe-Zebra/genética
9.
Acta Otolaryngol ; 138(11): 993-997, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30776266

RESUMO

BACKGROUND: Safe cochlear implantation (CI) is challenging in patients with a canal wall down (CWD) mastoidectomy cavity. OBJECTIVES: We reviewed the outcomes of CI and proposed surgical management principles according to the presentation status of CWD mastoidectomy cavity. MATERIAL AND METHODS: The cases of eight patients (nine ears) with CWD mastoidectomy cavity who underwent CI were retrospectively reviewed. The basis of the surgical decision, postoperative complications, and postimplant auditory performance were analysed. RESULTS: In seven patients (eight ears), implantation was performed in a single stage; in six ears, the external auditory canal (EAC) was oversewn. In two patients with ossification, the electrode array was inserted into the scala tympani by drilling of the basal turn or in the second turn of the cochlea through a drill-out procedure. Seven patients had a follow-up of 12-50 months and one patient was lost to follow-up. None of the followed-up seven patients suffered complications. CONCLUSIONS: CI is safe and effective in patients with profound hearing loss after radical mastoidectomy. In patients with CWD mastoidectomy cavity, CI does not cause a higher rate of postoperative complications relative to standard CI procedures. The outcome is excellent and comparable to that in the general CI population.


Assuntos
Implante Coclear/métodos , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/cirurgia , Mastoidectomia/métodos , Segurança do Paciente , Adulto , Idoso , Audiometria/métodos , Terapia Combinada/métodos , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Medição de Risco , Estudos de Amostragem , Índice de Gravidade de Doença , Tomografia Computadorizada por Raios X/métodos , Resultado do Tratamento , Adulto Jovem
10.
Neural Plast ; 2017: 3192090, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28695016

RESUMO

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Mutations in the TMPRSS3 (transmembrane protease, serine 3) gene cause prelingual (DFNB10) or postlingual (DFNB8) deafness. In our previous study, three pathogenic mutations in TMPRSS3 were identified in one Chinese family. To evaluate the importance of TMPRSS3 mutations in recessive deafness among the Chinese, we screened 150 autosomal recessive nonsyndromic hearing loss (ARNSHL) families and identified 6 that carried seven causative TMPRSS3 mutations, including five novel mutations (c.809T>A, c.1151T>G, c.1204G>A, c.1244T>C, and c.1250G>A) and two previously reported mutations (c.323-6G>A and c.916G>A). Each of the five novel mutations was classified as severe, by both age of onset and severity of hearing loss. Together with our previous study, six families were found to share one pathogenic mutation (c.916G>A, p.Ala306Thr). To determine whether this mutation arose from a common ancestor, we analyzed six short tandem repeat (STR) markers spanning the TMPRSS3 gene. In four families, we observed linkage disequilibrium between p.Ala306Thr and STR markers. Our results indicate that mutations in TMPRSS3 account for about 4.6% (7/151) of Chinese ARNSHL cases lacking mutations in SLC26A4 or GJB2 and that the recurrent TMPRSS3 mutation p.Ala306Thr is likely to be a founder mutation.


Assuntos
Perda Auditiva Neurossensorial/genética , Proteínas de Membrana/genética , Mutação , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Adulto , Idade de Início , Criança , Pré-Escolar , China , Análise Mutacional de DNA , Feminino , Perda Auditiva Neurossensorial/diagnóstico , Humanos , Recém-Nascido , Masculino , Índice de Gravidade de Doença , Adulto Jovem
11.
Mol Med Rep ; 16(3): 2785-2790, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713909

RESUMO

The present study aimed to construct a lentiviral RNA interference (RNAi) vector targeting the transforming growth factor ß1 (TGFß1) gene of rats, in order to examine its effect on silencing of the TGFß1 gene and on the expression of collagen type 1 α1 (Col1a1) in HSC­T6 rat hepatic stellate cells. Three RNAi sites of the TGFß1 gene were selected according to its CDs sequence. Three pairs of small interfering RNA (siRNA) of these RNAi sites were synthesized and then transfected into HSC­T6 cells, respectively, to confirm the optimal siRNA sequence via reverse transcription­polymerase chain reaction analysis. Subsequently, shRNA targeting the sequence of the optimal siRNA was designed, synthesized and annealed to form a double­stranded structure. The annealed oligonucleotide fragment was cloned into pGreenPuro plasmids to establish the pGreenPuro/TGFß1 shRNA lentiviral vector, which was then transfected into 293T cells, following identification by restriction enzyme digestion and sequencing for the production of lentiviral particles exhibiting high reactivity. These particles were used to infect HSC­T6 cells, following which the expression of GFP in the transfected cells was observed under an inverted microscope. The effects on TGFß1 gene silencing and the expression levels of Colla1 were detected at the mRNA and protein levels. The results provided confirmation of the optimal siRNA sequence. Enzyme digestion and sequencing verified successful construction of the pGreenPuro/TGFß1 shRNA lentiviral vector. This lentiviral vector effectively silenced the TGFß1 gene in the HSC­T6 cells, and inhibited the expression of Col1a1 at the mRNA and protein levels. Taken together, the lentiviral RNAi vector targeting the TGFß1 gene of rats was successfully constructed, which effectively silenced the TGFß1 gene of the HSC­T6 cells and inhibited the expression of Col1a1.


Assuntos
Colágeno Tipo I/genética , Células Estreladas do Fígado/metabolismo , Interferência de RNA , Fator de Crescimento Transformador beta1/genética , Animais , Linhagem Celular , Cadeia alfa 1 do Colágeno Tipo I , Regulação da Expressão Gênica , Lentivirus/genética , RNA Interferente Pequeno/genética , Ratos
12.
Biomed Res Int ; 2017: 4707315, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28246597

RESUMO

Autosomal recessive hearing impairment with postlingual onset is rare. Exceptions are caused by mutations in the TMPRSS3 gene, which can lead to prelingual (DFNB10) as well as postlingual deafness (DFNB8). TMPRSS3 mutations can be classified as mild or severe, and the phenotype is dependent on the combination of TMPRSS3 mutations. The combination of two severe mutations leads to profound hearing impairment with a prelingual onset, whereas severe mutations in combination with milder TMPRSS3 mutations lead to a milder phenotype with postlingual onset. We characterized a Chinese family (number FH1523) with not only prelingual but also postlingual hearing impairment. Three mutations in TMPRSS3, one novel mutation c.36delC [p.(Phe13Serfs⁎12)], and two previously reported pathogenic mutations, c.916G>A (p.Ala306Thr) and c.316C>T (p.Arg106Cys), were identified. Compound heterozygous mutations of p.(Phe13Serfs⁎12) and p.Ala306Thr manifest as prelingual, profound hearing impairment in the patient (IV: 1), whereas the combination of p.Arg106Cys and p.Ala306Thr manifests as postlingual, milder hearing impairment in the patient (II: 2, II: 3, II: 5), suggesting that p.Arg106Cys mutation has a milder effect than p.(Phe13Serfs⁎12). We concluded that different combinations of TMPRSS3 mutations led to different hearing impairment phenotypes (DFNB8/DFNB10) in this family.


Assuntos
Povo Asiático/genética , Genes Recessivos , Predisposição Genética para Doença , Perda Auditiva/genética , Proteínas de Membrana/genética , Mutação/genética , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Idoso , Sequência de Aminoácidos , Audiometria , Sequência de Bases , Criança , Implantes Cocleares , Sequência Conservada , Análise Mutacional de DNA , Surdez/genética , Família , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Linhagem , Fenótipo
14.
Am J Med Genet A ; 167A(10): 2357-65, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26079994

RESUMO

Hereditary nonsyndromic hearing loss is extremely heterogeneous. Mutations in the transmembrane channel-like gene1 (TMC1) are known to cause autosomal dominant and recessive forms of nonsyndromic hearing loss linked to the loci of DFNA36 and DFNB7/11, respectively. We characterized a six-generation Chinese family (5315) with progressive, postlingual autosomal dominant nonsyndromic hearing loss (ADNSHL). By combining targeted capture of 82 known deafness genes, next-generation sequencing and bioinformatic analysis, we identified TMC1 c.1714G>A (p. D572N) as the disease-causing mutation. This mutation co-segregated with hearing loss in other family members and was not detected in 308 normal controls. In order to determine the prevalence of TMC1 c.1714G>A in Chinese ADNSHL families, we used DNA samples from 67 ADNSHL families with sloping audiogram and identified two families carry this mutation. To determine whether it arose from a common ancestor, we analyzed nine STR markers. Our results indicated that TMC1 c.1714G>A (p.D572N) account for about 4.4% (3/68) of ADNSHL in the Chinese population.


Assuntos
Biologia Computacional/métodos , Perda Auditiva Neurossensorial/genética , Proteínas de Membrana/genética , Mutação , Adulto , Povo Asiático , Audiometria , Sequência de Bases , Estudos de Casos e Controles , Criança , Análise Mutacional de DNA , Feminino , Expressão Gênica , Genes Dominantes , Loci Gênicos , Marcadores Genéticos , Perda Auditiva Neurossensorial/etnologia , Perda Auditiva Neurossensorial/patologia , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Dados de Sequência Molecular , Linhagem
15.
PLoS One ; 10(4): e0124757, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25919374

RESUMO

Mutations in PTPRQ are associated with deafness in humans due to defects of stereocilia in hair cells. Using whole exome sequencing, we identified responsible gene of family 1572 with autosomal recessively non-syndromic hearing loss (ARNSHL). We also used DNA from 74 familial patients with ARNSHL and 656 ethnically matched control chromosomes to perform extended variant analysis. We identified two novel compound heterozygous missense mutations, c. 3125 A>G p.D1042G (maternal allele) and c.5981 A>G p.E1994G (paternal allele), in the PTPRQ gene, as the cause of recessively inherited sensorineural hearing loss in family 1572. Both variants co-segregated with hearing loss phenotype in family 1572, but were absent in 74 familial patients. Heterozygosity for c. 3125 A>G was identified in two samples from unaffected Chinese individuals (656 chromosomes). Therefore, the hearing loss in this family was caused by two novel compound heterozygous mutations in PTPRQ.


Assuntos
Povo Asiático/genética , Genes Recessivos , Estudos de Associação Genética , Predisposição Genética para Doença , Perda Auditiva/genética , Mutação/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Sequência de Aminoácidos , Sequência de Bases , Criança , Análise Mutacional de DNA , Exoma/genética , Família , Feminino , Heterozigoto , Humanos , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Linhagem , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/química , Adulto Jovem
16.
PLoS One ; 9(7): e103415, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25080338

RESUMO

Usher syndrome is an autosomal recessive disease characterized by sensorineural hearing loss, age-dependent retinitis pigmentosa (RP), and occasionally vestibular dysfunction. The most severe form is Usher syndrome type 1 (USH1). Mutations in the MYO7A gene are responsible for USH1 and account for 29-55% of USH1 cases. Here, we characterized a Chinese family (no. 7162) with USH1. Combining the targeted capture of 131 known deafness genes, next-generation sequencing, and bioinformatic analysis, we identified two deleterious compound heterozygous mutations in the MYO7A gene: a reported missense mutation c.73G>A (p.G25R) and a novel nonsense mutation c.462C>A (p.C154X). The two compound variants are absent in 219 ethnicity-matched controls, co-segregates with the USH clinical phenotypes, including hearing loss, vestibular dysfunction, and age-dependent penetrance of progressive RP, in family 7162. Therefore, we concluded that the USH1 in this family was caused by compound heterozygous mutations in MYO7A.


Assuntos
Heterozigoto , Mutação , Miosinas/genética , Síndromes de Usher/genética , Sequência de Aminoácidos , Animais , China , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Miosina VIIa , Miosinas/química , Linhagem , Homologia de Sequência de Aminoácidos
17.
J Transl Med ; 11: 284, 2013 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-24206587

RESUMO

BACKGROUND: Inherited genetic defects play an important role in congenital hearing loss, contributing to about 60% of deafness occurring in infants. Hereditary nonsyndromic hearing loss is highly heterogeneous, and most patients with a presumed genetic etiology lack a specific molecular diagnosis. METHODS: By whole exome sequencing, we identified responsible gene of family 4794 with autosomal recessively nonsyndromic hearing loss (ARNSHL). We also used DNA from 56 Chinese familial patients with ARNSHL (autosomal recessive nonsyndromic hearing loss) and 108 ethnicity-matched negative samples to perform extended variants analysis. RESULTS: We identified MYO15A c.IVS25+3G>A and c.8375 T>C (p.V2792A) as the disease-causing mutations. Both mutations co-segregated with hearing loss in family 4794, but were absent in the 56 index patients and 108 ethnicity-matched controls. CONCLUSIONS: Our results demonstrated that the hearing loss of family 4794 was caused by novel compound heterozygous mutations in MYO15A.


Assuntos
Exoma , Genes Recessivos , Perda Auditiva/genética , Heterozigoto , Mutação , Miosinas/genética , Análise de Sequência , Adulto , Animais , Sequência de Bases , China , DNA/genética , Feminino , Perda Auditiva/fisiopatologia , Testes Auditivos , Humanos , Masculino , Dados de Sequência Molecular , Miosinas/química , Linhagem , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
18.
PLoS One ; 8(5): e63026, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23690975

RESUMO

Hereditary nonsyndromic hearing loss is highly heterogeneous and most patients with a presumed genetic etiology lack a specific diagnosis. It has been estimated that several hundred genes may be associated with this sensory deficit in humans. Here, we identified compound heterozygous mutations in the TMC1 gene as the cause of recessively inherited sensorineural hearing loss by using whole-exome sequencing in a family with two deaf siblings. Sanger sequencing confirmed that both siblings inherited a missense mutation, c.589G>A p.G197R (maternal allele), and a nonsense mutation, c.1171C>T p.Q391X (paternal allele), in TMC1. We also used DNA from 50 Chinese familial patients with ARNSHL and 208 ethnicity-matched negative samples to perform extended variants analysis. Both variants co-segregated in family 1953, which had the hearing loss phenotype, but were absent in 50 patients and 208 ethnicity-matched controls. Therefore, we concluded that the hearing loss in this family was caused by novel compound heterozygous mutations in TMC1.


Assuntos
Povo Asiático/genética , Perda Auditiva Neurossensorial/genética , Heterozigoto , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Linhagem , Adolescente , Sequência de Aminoácidos , Animais , Sequência de Bases , Análise Mutacional de DNA , Exoma/genética , Feminino , Perda Auditiva Neurossensorial/patologia , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Masculino , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Polimorfismo Genético , Ratos , Irmãos , Adulto Jovem
19.
Acta Otolaryngol ; 132(10): 1126-30, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22779753

RESUMO

Aberrant internal carotid artery (ICA) in the middle ear is a rare, dangerous vascular anomaly and conservative follow-up was usually adopted in most reported cases. Here we report the case of an 8-year-old girl with symptoms of objective pulsatile tinnitus and conductive hearing loss in the right ear. Otoscopic examination, computed tomography, and conventional angiography were performed. An aberrant ICA combined with a 'third mobile window' was suspected preoperatively and confirmed at exploratory surgery of the middle ear. The aberrant ICA was treated, and the pulsatile tinnitus disappeared and hearing recovered after the surgery. This case suggests that surgery is practical to relieve troublesome tinnitus and hearing loss in appropriate cases with aberrant ICA.


Assuntos
Artéria Carótida Interna/anormalidades , Perda Auditiva Condutiva/etiologia , Zumbido/etiologia , Tomografia Computadorizada por Raios X/métodos , Malformações Vasculares/complicações , Testes de Impedância Acústica/métodos , Audiometria de Tons Puros , Artéria Carótida Interna/diagnóstico por imagem , Criança , Feminino , Seguimentos , Perda Auditiva Condutiva/diagnóstico , Humanos , Angiografia por Ressonância Magnética/métodos , Procedimentos Cirúrgicos Otológicos/métodos , Otoscopia/métodos , Medição de Risco , Zumbido/diagnóstico , Resultado do Tratamento , Malformações Vasculares/diagnóstico por imagem , Malformações Vasculares/cirurgia
20.
Acta Otolaryngol ; 132(9): 951-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22668345

RESUMO

CONCLUSIONS: Intraoperative computed tomography (iCT)-guided cochlear implantation is practical and effective for correct electrode placement in the cochlea of patients with congenital inner ear and/or complex middle ear malformation. OBJECTIVES: The operation in patients with inner ear and/or complex middle ear malformation including abnormal facial nerve course is difficult. This study evaluated the efficacy of cochlear implantation under the guidance of iCT to insure correct electrode placement. METHODS: This was a prospective interventional case series. Ten patients with severe to profound sensorineural hearing loss due to ear malformations were enrolled, and iCT was used to confirm the right placement of electrodes. RESULTS: Intraoperative CT was performed three times in one patient, twice in two, and once in the others. Interruption of the surgical process for each iCT until resumption of surgery was 9.64 ± 0.63 min. iCT revealed incorrectly positioned cochlear implants in two patients, which were immediately corrected. There were no reoperations due to misplacement of electrodes. iCT helped locate the cochlea in the middle ear of one patient with an abnormal facial nerve course. The overall intervention rate based on iCT findings was 30%. LEVEL OF EVIDENCE: level 4.


Assuntos
Implante Coclear/métodos , Surdez/cirurgia , Orelha Interna/anormalidades , Orelha Média/anormalidades , Perda Auditiva Neurossensorial/congênito , Perda Auditiva Neurossensorial/cirurgia , Cirurgia Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Adulto , Criança , Pré-Escolar , China , Surdez/congênito , Surdez/diagnóstico por imagem , Surdez/fisiopatologia , Orelha Interna/diagnóstico por imagem , Orelha Interna/fisiopatologia , Orelha Interna/cirurgia , Orelha Média/diagnóstico por imagem , Orelha Média/fisiopatologia , Orelha Média/cirurgia , Eletrodos Implantados , Nervo Facial/anormalidades , Nervo Facial/fisiopatologia , Nervo Facial/cirurgia , Feminino , Perda Auditiva Neurossensorial/diagnóstico por imagem , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Lactente , Masculino , Complicações Pós-Operatórias/diagnóstico por imagem , Complicações Pós-Operatórias/fisiopatologia , Telemetria
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA