Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34050020

RESUMO

Genes that are primarily expressed in cochlear glia-like supporting cells (GLSs) have not been clearly associated with progressive deafness. Herein, we present a deafness locus mapped to chromosome 3p25.1 and an auditory neuropathy spectrum disorder (ANSD) gene, TMEM43, mainly expressed in GLSs. We identify p.(Arg372Ter) of TMEM43 by linkage analysis and exome sequencing in two large Asian families segregating ANSD, which is characterized by inability to discriminate speech despite preserved sensitivity to sound. The knock-in mouse with the p.(Arg372Ter) variant recapitulates a progressive hearing loss with histological abnormalities in GLSs. Mechanistically, TMEM43 interacts with the Connexin26 and Connexin30 gap junction channels, disrupting the passive conductance current in GLSs in a dominant-negative fashion when the p.(Arg372Ter) variant is introduced. Based on these mechanistic insights, cochlear implant was performed on three subjects, and speech discrimination was successfully restored. Our study highlights a pathological role of cochlear GLSs by identifying a deafness gene and its causal relationship with ANSD.


Assuntos
Códon sem Sentido , Conexinas/metabolismo , Genes Dominantes , Perda Auditiva Central/genética , Proteínas de Membrana/genética , Animais , Implante Coclear , Feminino , Perda Auditiva Central/metabolismo , Perda Auditiva Central/fisiopatologia , Perda Auditiva Central/cirurgia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linhagem , Percepção da Fala
2.
Hum Mutat ; 41(5): 913-920, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31944473

RESUMO

DNA polymerase δ, whose catalytic subunit is encoded by POLD1, is responsible for synthesizing the lagging strand of DNA. Single heterozygous POLD1 mutations in domains with polymerase and exonuclease activities have been reported to cause syndromic deafness as a part of multisystem metabolic disorder or predisposition to cancer. However, the phenotypes of diverse combinations of POLD1 genotypes have not been elucidated in humans. We found that five members of a multiplex family segregating autosomal recessive nonsyndromic sensorineural hearing loss (NS-SNHL) have revealed novel compound heterozygous POLD1 variants (p.Gly1100Arg and a presumptive null function variant, p.Ser197Hisfs*54). The recombinant p.Gly1100Arg polymerase δ showed a reduced polymerase activity by 30-40%, but exhibited normal exonuclease activity. The polymerase activity in cell extracts from the affected subject carrying the two POLD1 mutant alleles was about 33% of normal controls. We suggest that significantly decreased polymerase δ activity, but not a complete absence, with normal exonuclease activity could lead to NS-SNHL.


Assuntos
DNA Polimerase III/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Variação Genética , Perda Auditiva/diagnóstico , Perda Auditiva/genética , Adulto , Alelos , Sequência de Aminoácidos , Substituição de Aminoácidos , Biomarcadores , DNA Polimerase III/metabolismo , Ativação Enzimática , Feminino , Estudos de Associação Genética/métodos , Genótipo , Humanos , Masculino , Mutação , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Irmãos , Síndrome , Sequenciamento do Exoma
3.
J Med Genet ; 56(12): 818-827, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31473629

RESUMO

BACKGROUND: Diaphanous-related formin 1 (DIA1), which assembles the unbranched actin microfilament and microtubule cytoskeleton, is encoded by DIAPH1. Constitutive activation by the disruption of autoinhibitory interactions between the N-terminal diaphanous inhibitory domain (DID) and C-terminal diaphanous autoregulatory domain (DAD) dysregulates DIA1, resulting in both hearing loss and blood cell abnormalities. METHODS AND RESULTS: Here, we report the first constitutively active mutant in the DID (p.A265S) of humans with only hearing loss and not blood cell abnormality through whole exome sequencing. The previously reported DAD mutants and our DID mutant (p.A265S) shared the finding of diminished autoinhibitory interaction, abnormally upregulated actin polymerisation activity and increased localisations at the plasma membrane. However, the obvious defect in the DIA1-driven assembly of cytoskeleton 'during cell division' was only from the DAD mutants, not from p.A265S, which did not show any blood cell abnormality. We also evaluated the five DID mutants in the hydrophobic pocket since four of these five additional mutants were predicted to critically disrupt interaction between the DID and DAD. These additional pathogenic DID mutants revealed varying degrees of defect in the DIA1-driven cytoskeleton assembly, including nearly normal phenotype during cell division as well as obvious impaired autoinhibition, again coinciding with our key observation in DIA1 mutant (p.A265S) in the DID. CONCLUSION: Here, we report the first mutant in the DID of humans with only hearing loss. The differential cell biological phenotypes of DIA1 during cell division appear to be potential determinants of the clinical severity of DIAPH1-related cytoskeletopathy in humans.


Assuntos
Divisão Celular/genética , Citoesqueleto/genética , Forminas/genética , Perda Auditiva/genética , Citoesqueleto de Actina/genética , Citoesqueleto/patologia , Feminino , Estudos de Associação Genética , Perda Auditiva/patologia , Humanos , Masculino , Microtúbulos/genética , Proteínas Mutantes/genética , Mutação/genética , Domínios Proteicos/genética , Sequenciamento do Exoma
4.
Hum Mutat ; 40(5): 525-531, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30740825

RESUMO

Otoancorin (OTOA), encoded by OTOA, is required for the development of the tectorial membrane in the inner ear. Mutations in this gene cause nonsyndromic hearing loss (DFNB22). The molecular mechanisms underlying most DFNB22 remain poorly understood. Disruption of glycosylphosphatidylinositol (GPI) anchorage has been assumed to be the pathophysiology mandating experimental validation. From a Korean deaf family, we identified two trans OTOA variants (c.1320 + 5 G > C and p.Gln589ArgfsX55 [NM_144672.3]) . The pathogenic potential of c.1320 + 5 G > C was confirmed by a minigene splicing assay. To experimentally determine the GPI anchorage, wild-type (WT) and mutant OTOA harboring p.Gln589ArgfsX55 were expressed in HEK293T cells. The mutant OTOA with p.Gln589ArgfsX55 resulted in an uncontrolled release of OTOA into the medium in contrast with phosphatidylinositol-specific phospholipase C-induced controlled release of WT OTOA from the cell surface. Together, the results of this reverse translational study confirmed GPI-anchorage of OTOA and showed that downstream sequences from the 589th amino acid are critical for GPI-anchorage.


Assuntos
Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Variação Genética , Glicosilfosfatidilinositóis/metabolismo , Perda Auditiva/genética , Perda Auditiva/metabolismo , Alelos , Processamento Alternativo , Biomarcadores , Predisposição Genética para Doença , Genótipo , Perda Auditiva/diagnóstico , Humanos
5.
Cerebellum ; 18(3): 659-664, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30635863

RESUMO

Recent advances in next-generation sequencing technologies have uncovered the genetic backgrounds of various diseases. Type 1 sialidosis (OMIM#256550) is a rare autosomal recessive lysosomal storage disease caused by a mutation in the NEU1 (OMIM * 608272) gene. In this study, we aimed to review the previous reports of type 1 sialidosis and compare those with the first case of type 1 sialidosis in Korea. A 36-year-old woman presented with progressive ataxia, myoclonus, and seizure since the age of 12. Whole-exome sequencing revealed a pathogenic missense variant c.928G > A (p.D310N) and novel c.15_16del (p.P6Qfs*21) of the NEU1 gene as final causal candidate as compound heterozygotes. We reviewed the literature and selected the clinical reports of genetically confirmed type 1 sialidosis patients. A total of 45 patients in 17 reports were identified. Cherry-red spot, myoclonus, ataxia, and seizure were reported in 51.2%, 100.0%, 87.8%, and 73.7% of patients, respectively. Abnormalities of cognitive function, EEG, and brain MRI and visual symptoms were reported in 22.2%, 40.7%, 66.7%, and 70.2% of patients, respectively. Overall, our patient showed similar clinical features to previous type 1 sialidosis patients, but she did not complain of visual symptoms despite having cherry-red spots. We summarize the clinical features of type 1 sialidosis and report the first case of type 1 sialidosis with novel deletion variant in the NEU1 gene in the Korean population. Our study suggests the importance of ophthalmologic examinations in patients with myoclonus, ataxia, and seizure who do not complain of visual symptoms.


Assuntos
Mucolipidoses/genética , Neuraminidase/genética , Adulto , Feminino , Humanos , Mutação de Sentido Incorreto
6.
J Gene Med ; 20(6): e3019, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29607572

RESUMO

BACKGROUND: The MYO6 gene, if altered, can cause nonsyndromic hearing loss (NSHL) either in an autosomal dominant (AD) (DFNA22) or recessive form. The present study identified MYO6 variants in the cohort of Korean AD NSHL families and investigated the audiological phenotypes of DFNA22 with respect to suggesting clinical guides for the counseling of DFNA22. METHODS: A retrospective cohort study was performed on 81 AD NSHL families in two hospitals. Among them, five families (SH21, SB60, SB247, SB290 and SB305) segregating with MYO6 variant were genetically and clinically assessed. RESULTS: We identified two novel missense variants of MYO6: p.G223R (SB290) and p.T158R (SB305). A known heterozygous truncation variant, p.R205X, reported previously (SH21, SB60), was identified (SB247). The overall frequency of DFNA22 among such cases was 6.2%. Specifically, we found p.R205X from three of five DFNA22 families (60%). Five DFNA22 families demonstrated extremely diverse audiogram configurations and age of onset with even intrafamilial variations, whereas the severity of hearing loss mostly ranged within moderate. CONCLUSIONS: We report a recurring predominant allele and two new missense variants of MYO6, highlighting the significant contribution of MYO6 to AD NSHL in the Korean population. Extremely diverse audiological configurations of DFNA22 suggest that MYO6 should be considered in future genetic studies of patients with AD NSHL. Gradual progression with a good speech audiometry score could provide physicians with clinical insight with respect to advising patients to use hearing aids or consider middle ear implants, whereas, in the case of certain exceptional circumstances, physicians could provide patients with the option to consider a cochlear implant.


Assuntos
Perda Auditiva Neurossensorial/genética , Cadeias Pesadas de Miosina/genética , Idade de Início , Povo Asiático/genética , Audiologia/métodos , Códon sem Sentido , Estudos de Coortes , Feminino , Frequência do Gene , Estudos de Associação Genética , Perda Auditiva Neurossensorial/etiologia , Humanos , Masculino , Linhagem , Estudos Retrospectivos
7.
J Transl Med ; 16(1): 330, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30482216

RESUMO

BACKGROUND: While auditory neuropathy spectrum disorder (ANSD) is a heterogeneous disorder and its management quite varies depending upon the etiology, even including self-resolution, OTOF is an important molecular etiology of prelingual ANSD and has emerged as an attractive target for implementation of precision medicine in terms of timing and prognosis prediction of auditory rehabilitation. However, to date, the literature is lacking in the genotype-phenotype relationship of this gene as well as efficient molecular testing strategy in the clinic in many populations and to make things more complicated in Koreans, the most prevalent variant p.Arg1939Gln among Korean ANSD children frequently evaded detection by next generation sequencing (NGS), resulting in delayed genetic diagnosis and late cochlear implantation (CI). The aims of this study are to document the mutational and phenotypic spectrum of OTOF-related ANSD (DFNB9) in the Korean population, further establishing genotype-phenotype correlation and proposing a set of the most commonly found OTOF variants to be screened first. METHODS: Genetic diagnosis through the NGS-based sequencing was made on patients with ANSD in two tertiary hospitals. Genotype and phenotypes of eleven DFNB9 patients were reviewed. For data analysis, Mann-Whitney test and Fisher's exact test were applied. RESULTS: This study disclosed four prevalent variants in Koreans: p.Arg1939Gln with an allele frequency of 40.9%, p.Glu841Lys (13.6%), p.Leu1011Pro and p.Arg1856Trp (9.1%). Three novel variants (c.4227 + 5G > C, p.Gly1845Glu, and p.Pro1931Thr) were identified. Interestingly, a significant association of p.Arg1939Gln with worse ASSR thresholds was observed despite consistently no ABR response. Ten of 11 DFNB9 patients received CI for auditory rehabilitation, showing favorable outcomes with more rapid improvement on early-CI group (age at CI ≤ 18 mo.) than late-CI group. CONCLUSIONS: This study included the largest Korean DFNB9 cohort to date and proposed a set of the most frequent four OTOF variants, allowing the potential prioritization of exons during Sanger sequencing. Further, a significant association of p.Arg1939Gln homozygotes with poor residual hearing was observed. We may have to suspect p.Arg1939Gln homozygosity in cases of poor auditory thresholds in ANSD children with putative negative OTOF variants solely screened by NGS. Reciprocal feedback between bench and clinics regarding DFNB9 would complement each other.


Assuntos
Perda Auditiva Central/genética , Proteínas de Membrana/genética , Mutação/genética , Pesquisa Translacional Biomédica , Percepção Auditiva , Implante Coclear , Família , Feminino , Humanos , Masculino , Proteínas de Membrana/química , Linhagem , Fenótipo , Domínios Proteicos , República da Coreia
8.
J Gene Med ; 19(4)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28221712

RESUMO

BACKGROUND: Pathogenic variants of MYH14 are known to be associated (in either a syndromic or nonsyndromic manner) with hearing loss. Interestingly, all reported cases to date of MYH14-related nonsyndromic hearing loss with detailed phenotypes have demonstrated mild-to-moderate progressive hearing loss with postlingual onset. METHODS: In the present study, targeted resequencing (TRS) of known deafness genes was performed to identify the causative variant in two multiplex families segregating autosomal dominant (AD) inherited hearing loss. RESULTS: TRS uncovered two novel variants of MYH14 (c.A572G: p.Asp191Gly in the myosin head domain and c.C73T:p.Gln25* in exon 2) from two multiplex deafness Korean families. Notably, both probands showed phenotypes of congenital or prelingual severe hearing loss. It is remarkably uncommon to encounter such a severe-to-profound, prelingual, AD hearing loss. Given that the first variant, p. Asp191Gly, was the first documented missense allele discovered in the myosin head domain of this gene related to either congenital or prelingual severe nonsyndromic hearing loss, and also that the second variant, p. Gln25*, lead to a null allele, more severe phenotypes from our probands may have been the result of either genotype-phenotype correlation or genetic backgrounds, or both. CONCLUSIONS: In the present study, we report that MYH14 can manifest as nonsyndromic prelingual severe sensorineural hearing loss in an AD fashion in Koreans. The results of the present study suggest that further genetic studies of similar patients should consider MYH14 as a causative gene, and cochlear implantation during infant or early childhood should be indicated for those patients with certain MYH14 pathogenic variants.


Assuntos
Surdez/diagnóstico , Surdez/genética , Genes Dominantes , Estudos de Associação Genética , Mutação , Cadeias Pesadas de Miosina/genética , Miosina Tipo II/genética , Alelos , Sequência de Aminoácidos , Pré-Escolar , Análise Mutacional de DNA , Feminino , Humanos , Lactente , Masculino , Linhagem
9.
J Exp Bot ; 68(7): 1797-1809, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28369480

RESUMO

Proline metabolism is important for environmental responses, plant growth, and development. However, its precise roles in plant abiotic stress tolerance are not well understood. Mutants are valuable for the identification of new genes and for elucidating their roles in physiological mechanisms. We applied a suppressor mutation approach to identify novel genes involved in the regulation of proline metabolism in Arabidopsis. Using the atrzf1 (Arabidopsis thaliana ring zinc finger 1) mutant as a parental line for activation tagging mutagenesis, we selected several mutants with suppressed induction of proline accumulation under dehydration conditions. One of the selected mutants [proline content alterative 22 (pca22)] appeared to have reduced proline contents compared with the atrzf1 mutant under drought stress. Generally, pca22 mutant plants displayed suppressed atrzf1 insensitivity to dehydration and abscisic acid during early seedling growth. Additionally, the pca22 mutant exhibited shorter pollen tube length than wild-type (WT) and atrzf1 plants. Furthermore, PCA22-overexpressing plants were more sensitive to dehydration stress than the WT and RNAi lines. Green fluorescent protein-tagged PCA22 was localized to the cytoplasm of transgenic Arabidopsis cells. Collectively, these results suggest that pca22 acts as dominant suppressor mutant of atrzf1 in the abiotic stress response.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/fisiologia , Regulação da Expressão Gênica de Plantas , Peptídeos e Proteínas de Sinalização Intracelular/genética , Prolina/metabolismo , Estresse Fisiológico , Sequência de Aminoácidos , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Filogenia , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/metabolismo , Alinhamento de Sequência
10.
Ear Hear ; 38(5): e316-e324, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28841141

RESUMO

OBJECTIVES: About 60% of Korean pediatric cochlear implantees could be genetically diagnosed (GD) and we previously reported that a substantial portion of undiagnosed cases by deafness gene panel sequencing were predicted to have a nongenetic or complex etiology. We aimed to compare the outcomes of cochlear implantation (CI) in GD and genetically undiagnosed (GUD) patients and attempted to determine CI outcomes according to etiology. DESIGN: Ninety-three pediatric cochlear implantees underwent molecular genetic testing. Fifty-seven patients carried pathogenic variants and 36 patients remained GUD after panel sequencing of 204 known or potential deafness genes (TRS-204). Among them, 55 cochlear implantees with reliable speech evaluation results with a follow-up of longer than 24 months were recruited. Longitudinal changes in the audiologic performance were compared between the GD (n = 31) and GUD (n = 24) groups. The GD group was subdivided into cochlear implantee with SLC26A4 mutations (group 1) and cochlear implantee with other genetic etiology (group 2), and the GUD group was subdivided into groups 3 and 4, that is, patients with or without inner ear anomaly, respectively. RESULTS: Group 1 related to SLC26A4 mutations had the highest categories of auditory perception scores among all groups pre- and postoperatively. Group 4 with inner ear anomaly had the lowest categories of auditory perception scores. At 24 months post-CI, the group 2 with another genetic etiology had significantly better outcomes than molecularly undiagnosed group 3, which had with the same condition as group 2 except that the candidate gene was not detected. This finding was recapitulated when we limited cases to those that underwent CI before 24 months of age to minimize age-related bias at implantation. Furthermore, on extending the follow-up to 36 months postoperatively, this tendency became more prominent. Additionally, our preliminary clinical data suggest a narrower sensitive window period for good CI outcomes for implantees with OTOF mutation rather than the GJB2 and other genes. CONCLUSIONS: Current molecular genetic testing including deafness panel sequencing helps to predict the 2-year follow-up outcomes after CI in prelingually deafened children. GD cochlear implantees show better functional outcomes after CI than undiagnosed cochlear implantees as determined by deafness panel sequencing, suggesting a genotype-functional outcome correlation. The genetic testing may provide a customized optimal window period in terms of CI timing for favorable outcome according to genetic etiology.


Assuntos
Implante Coclear , Surdez/reabilitação , Percepção da Fala , Percepção Auditiva , Pré-Escolar , Implantes Cocleares , Surdez/genética , Feminino , Testes Genéticos , Genótipo , Humanos , Lactente , Masculino , Proteínas de Membrana Transportadoras/genética , Mutação , República da Coreia , Transportadores de Sulfato , Resultado do Tratamento
11.
Int J Mol Sci ; 18(11)2017 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-29072634

RESUMO

We performed targeted re-sequencing to identify the genetic etiology of early-onset postlingual deafness and encountered a frequent TMPRSS3 allele harboring two variants in a cis configuration. We aimed to evaluate the pathogenicity of the allele. Among 88 cochlear implantees with autosomal recessive non-syndromic hearing loss, subjects with GJB2 and SLC26A4 mutations were excluded. Thirty-one probands manifesting early-onset postlingual deafness were sorted. Through targeted re-sequencing, we detected two families with a TMPRSS3 mutant allele containing p.V116M and p.V291L in a cis configuration, p.[p.V116M; p.V291L]. A minor allele frequency was calculated and proteolytic activity was measured. A p.[p.V116M; p.V291L] allele demonstrated a significantly higher frequency compared to normal controls and merited attention due to its high frequency (4.84%, 3/62). The first family showed a novel deleterious splice site variant-c.783-1G>A-in a trans allele, while the other showed homozygosity. The progression to deafness was noted within the first decade, suggesting DFNB10. The proteolytic activity was significantly reduced, confirming the severe pathogenicity. This frequent mutant allele significantly contributes to early-onset postlingual deafness in Koreans. For clinical implication and proper auditory rehabilitation, it is important to pay attention to this allele with a severe pathogenic potential.


Assuntos
Alelos , Substituição de Aminoácidos , Surdez/genética , Proteínas de Membrana/genética , Mutação , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Sequência de Aminoácidos , Criança , Pré-Escolar , Surdez/diagnóstico , Feminino , Frequência do Gene , Estudos de Associação Genética , Perda Auditiva Neurossensorial/genética , Humanos , Masculino , Linhagem , Fenótipo , República da Coreia , Análise de Sequência de DNA
12.
J Gene Med ; 18(11-12): 353-358, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27886419

RESUMO

BACKGROUND: The symptoms of phosphoribosyl pyrophosphate synthetase 1 (PRPS1) deficiency diseases have been reported to be alleviated by medication. In the present study, we report biochemical data that favor PRPS1 deficiency-related hearing loss as a potential target for pharmaceutical treatment. METHODS: We recruited 42 probands from subjects aged less than 15 years with a moderate degree of nonsyndromic autosomal-recessive or sporadic sensorineural hearing loss (SNHL) in at least one side. Molecular genetic testing, including targeted exome sequencing (TES) of 129 genes for deafness, and in silico prediction were performed. RESULTS: A strong candidate variant (p.A82P) of PRPS1 is co-segregated with SNHL in X-linked recessive inheritance from one Korean multiplex SNHL family. Subsequent measurement of in vitro enzymatic activities of PRPS1 from erythrocytes of affected and unaffected family members, as well as unrelated normal controls, confirmed a pathogenic role of this variant. In detail, compared to normal hearing controls (0.23-0.26 nmol/ml/h), the proband, the affected sibling and their normal hearing mother demonstrated a significantly decreased PRPS1 enzymatic activity (0.07, 0.03 and 0.11 nmol/ml/h, respectively). This novel loss-of-function mutation of PRPS1 (p.A82P) is the ninth and sixth most reported mutation in the world and in Asia, respectively. CONCLUSIONS: DFNX1 was found to account for approximately 2.4% (1/42) of moderate SNHL in a Korean pediatric population. Confirmation of PRPS1 activity deficiency and an audiologic phenotype that initially begins in a milder form of SNHL, as in our family, should indicate the need for rigorous genetic screening as early as possible.


Assuntos
Surdez/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Perda Auditiva Neurossensorial/genética , Ribose-Fosfato Pirofosfoquinase/genética , Sequência de Bases , Estudos de Casos e Controles , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Mutação , Linhagem , República da Coreia , Sequenciamento do Exoma
13.
Mycoses ; 59(5): 274-80, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26786542

RESUMO

We investigated the distribution of Malassezia yeast in 120 Chinese (20 patients from each of six cities) and 20 Korean patients with scalp seborrheic dermatitis (SD) and dandruff (SD/D) using ITS1 and ITS2 polymerase chain reaction-restriction fragment length polymorphism. Bioactivity was studied by quantifying sebum lipid production by human primary sebocytes and inflammatory cytokine, interleukin-8 (IL-8) production was studied by exposing HaCaT keratinocytes with extracts of five standard Malassezia strains; M. globosa, M. restricta, M. sympodialis, M. dermatis and M. slooffiae. M. restricta and M. globosa were the most frequently encountered species from both Chinese and Korean patients. These two Malassezia species also promoted neutral lipid synthesis although the result was not statistically significant and induced significant increase in IL-8 production among the five Malassezia species studied. The study suggests a possible role of these organisms in the pathogenesis of SD/D.


Assuntos
Dermatite Seborreica/microbiologia , Interleucina-8/biossíntese , Malassezia/isolamento & purificação , Dermatoses do Couro Cabeludo/microbiologia , Sebo/metabolismo , Adulto , Idoso , Células Cultivadas , China , DNA Fúngico/isolamento & purificação , DNA Intergênico/análise , DNA Ribossômico/análise , Caspa/microbiologia , Feminino , Genoma Fúngico/genética , Humanos , Queratinócitos/citologia , Lipídeos/biossíntese , Malassezia/classificação , Malassezia/genética , Malassezia/imunologia , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , RNA Ribossômico/genética , Seul , População Urbana
14.
Eur Arch Otorhinolaryngol ; 273(5): 1123-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26015337

RESUMO

The EYA4 gene encodes a 640-amino-acid protein that serves as a transcription factor. This protein contains a highly conserved Eya domain (eya-HR) and a variable domain (eya-VR). Mutations of this gene are known to cause postlingual and progressive sensorineural hearing loss, either as non-syndromic (DFNA10) or syndromic hearing loss, depending on the location of truncation of the mutant protein. Since our previous report, we have recruited 14 families segregating autosomal dominant moderate SNHL. A thorough medical history and physical examination including evaluation of heart problems ruled out any syndromic features in these families. Screening of EYA4 was performed by targeted exome sequencing of 134 known deafness genes (TES-134) from the probands. After basic filtering of the variants, we identified one proband who carried a novel truncation mutation, c.1194delT (p.Met401TrpfsX3) of EYA4, making the frequency of DFNA10 to be 7.14 % (1/14) in Koreans. The variant co-segregated perfectly with a slightly down-sloping, moderate degree of SNHL in the family (SH117), and was not detected in any of the 592 normal control chromosomes. This variant is likely to generate protein products that are truncated just downstream of the eya-VR domain. None of the three affected family members showed any syndromic features, including cardiac problems, which was compatible with a previous genotype-phenotype correlation. The identification of a novel EYA4 truncation mutation associated with DFNA10, rather than syndromic hearing loss, supports a previously reported genotype-phenotype correlation in this gene. Considering its detection rate, EYA4 mutations should be suspected in hereditary moderate hearing loss with a corresponding audiologic configuration, and a cardiac examination should be included in the initial evaluation.


Assuntos
Povo Asiático/genética , Exoma/genética , Perda Auditiva Neurossensorial/genética , Mutação/genética , Transativadores/genética , Adulto , Estudos de Casos e Controles , Feminino , Estudos de Associação Genética , Humanos , Masculino , Linhagem , República da Coreia
15.
Eur Arch Otorhinolaryngol ; 273(10): 2993-3002, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26758463

RESUMO

Mutations of COCH can cause hearing loss and less frequently vestibular symptoms. However, vestibular phenotypes, especially in terms of the location of specific variants are not well documented yet. In this study, a retrospective and prospective cohort survey was performed in two tertiary referral hospitals to demonstrate vestibular phenotypes of DFNA9 subjects with a focus on the relationship with the location of COCH mutations. Two DFNA9 subjects were recruited from the previously collected cohort, each segregating p.G38D and p.C162Y of the COCH gene. Another two DFNA9 families were newly detected by targeted resequencing of known 129 deafness genes (TRS-129). These two families segregated the p.G38D variant of the COCH gene as the causative mutation, making p.G38D the most frequent COCH mutation in our Korean cohorts. Regarding the detailed clinical phenotype of the four DFNA9 families with documented vestibular phenotypes, we were able to classify them into two groups: one (p.C162Y variant) with a Meniere's disease (MD)-like phenotype and the other three (p.G38D variant) with significant bilateral vestibular loss without any definite MD symptoms. Distinct vestibular phenotypes depending on the location of COCH mutations were demonstrated, and this study correlates a genotype of p.G38D in COCH to the phenotype of bilateral total vestibular loss, therefore expanding the vestibular phenotypic spectrum of DFNA9 to range from bilateral vestibular loss without episodic vertigo to MD-like features with devastating episodic vertigo. In addition, the p.G38D variant of the COCH gene is suggested to be a frequent cause of progressive audiovestibular dysfunction in Koreans eventually requiring cochlear implantation.


Assuntos
DNA/genética , Proteínas da Matriz Extracelular/genética , Mutação , Doenças Vestibulares/genética , Adulto , Análise Mutacional de DNA , Proteínas da Matriz Extracelular/metabolismo , Feminino , Genótipo , Perda Auditiva Neurossensorial , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Estudos Prospectivos , Doenças Vestibulares/metabolismo
17.
J Transl Med ; 13: 263, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26264712

RESUMO

BACKGROUND: Despite the prevalence of CDH23 mutations in East Asians, its large size hinders investigation. The pathologic mutation p.P240L in CDH23 is common in East Asians. However, whether this mutation represents a common founder or a mutational hot spot is unclear. The prevalence of CDH23 mutations with prelingual severe-to-profound sporadic or autosomal recessive sensorineural hearing loss (arSNHL) is unknown in Koreans. METHODS: From September 2010 to October 2014, children with severe-to-profound sporadic or arSNHL without phenotypic markers, and their families, were tested for mutations in connexins GJB2, GJB6 and GJB3. Sanger sequencing of CDH23 p.P240L was performed on connexin-negative samples without enlarged vestibular aqueducts (EVA), followed by targeted resequencing of 129 deafness genes, including CDH23, unless p.P240L homozygotes were detected in the first screening. Four p.P240L-allele-linked STR markers were genotyped in 40 normal-hearing control subjects, and the p.P240L carriers in the hearing-impaired cohort, to identify the haplotypes. RESULTS: Four (3.1 %) of 128 children carried two CDH23 mutant alleles, and SLC26A4 and GJB2 accounted for 18.0 and 17.2 %, respectively. All four children showed profound nonsyndromic SNHL with minimal residual hearing. Interestingly, all had at least one p.P240L mutant allele. Analysis of p.P240L-linked STR markers in these children and other postlingual hearing-impaired adults carrying p.P240L revealed that p.P240L was mainly carried on a single haplotype. CONCLUSIONS: p.P240L contributed significantly to Korean pediatric severe arSNHL with a strong founder effect, with implications for future phylogenetic studies. Screening for p.P240L as a first step in GJB2-negative arSNHL Koreans without EVA is recommended.


Assuntos
Povo Asiático/genética , Caderinas/genética , Efeito Fundador , Predisposição Genética para Doença , Mutação/genética , Alelos , Proteínas Relacionadas a Caderinas , Criança , Cromossomos Humanos Par 10/genética , Estudos de Coortes , Conexina 26 , Conexinas , Surdez/genética , Feminino , Haplótipos/genética , Humanos , Masculino , Repetições de Microssatélites/genética , Linhagem
18.
Genet Med ; 17(11): 901-11, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25719458

RESUMO

PURPOSE: This study was designed to delineate genetic contributions, if any, to sporadic forms of mild to moderate sensorineural hearing loss (SNHL) not related to GJB2 mutations (DFNB1) in a pediatric population. METHODS: We recruited 11 non-DFNB1 simplex cases of mild to moderate SNHL in children. We applied whole-exome sequencing to all 11 probands. We used a filtering strategy assuming that de novo variants of known autosomal dominant (AD) deafness genes, biallelic mutations in autosomal recessive (AR) genes, monoallelic mutations in X chromosome genes for males, and digenic inheritance could be associated. Candidate variants first were prioritized with allele frequency in public databases and confirmed by a phase or a segregation test in each family. Additional information from the literature or public databases was used to identify strong candidate variants. RESULTS: Strong candidate variants were detected in 5 of 11 probands (45.4%). A diverse mode of inheritance implicated the sporadic occurrence of the phenotype. AR mutations in OTOGL and SERPINB6 and digenic inheritance involving two deafness genes, GPR98 and PDZ7, were detected. A de novo AD mutation also was detected in TECTA and MYH14. No syndromic feature was detected in individuals with GPR98/PDZ7 or MYH14 variants in our cohort at this moment. CONCLUSION: Mild to moderate pediatric SNHL, even if sporadic, features a strong genetic etiology and can manifest via diverse modes of inheritance. In addition, a multidisciplinary approach should be used for a correct diagnosis.


Assuntos
Exoma , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/genética , Sequenciamento de Nucleotídeos em Larga Escala , Padrões de Herança , Adolescente , Sequência de Aminoácidos , Criança , Pré-Escolar , Conexina 26 , Conexinas/genética , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , Feminino , Testes Auditivos , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Alinhamento de Sequência , Índice de Gravidade de Doença
19.
Audiol Neurootol ; 20(1): 17-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25413827

RESUMO

TECTA is a causative gene of autosomal dominant (DFNA8/A12) and autosomal recessive (DFNB 21) nonsyndromic sensorineural hearing loss (NSHL). Mutations in TECTA account for 4% of all autosomal dominant NSHL cases in some populations and are thus thought to be one of the major causes of autosomal dominant NSHL. A genotype-phenotype correlation for autosomal dominant mutations in the TECTA gene has been proposed. Two families (SB146 and SB149), which segregated moderate NSHL in an autosomal dominant fashion, were included in this study. We performed targeted resequencing of 134 known deafness genes (TRS-134) and bioinformatics analyses to find causative mutations for NSHL in these 2 families. Through TRS-134, we detected 2 novel mutations, i.e. c.3995G>T (p.C1332F) and c.5618C>T (p.T1873I), in the TECTA gene. These mutations cosegregated with NSHL in the studied families and were not detected in normal controls. The mutations c.3995G>T and c.5618C>T reside in the von Willebrand factor type D3-D4 (vWFD3-D4) interdomain of the zonadhesin (ZA) domain and the zona pellucida (ZP) domain, respectively. p.C1332F is the first mutation detected in the vWFD3-D4 interdomain of the ZA domain. The mutations p.C1332F and p.T1873I were associated with stable high-frequency and mid-frequency hearing loss, respectively. Notably, the cysteine residue mutated to phenylalanine in SB146 was not related to progression of sensorineural hearing loss, which argues against the previous hypothesis. Here we confirm a known genotype-phenotype correlation for the ZP domain and propose a hypothetical genotype-phenotype correlation which relates mutations in vWFD3-D4 to stable high-frequency NSHL in Koreans. This clinical feature makes subjects with the missense mutation in the vWFD3-D4 interdomain of TECTA potentially good candidates for middle ear implantation.


Assuntos
Proteínas da Matriz Extracelular/genética , Perda Auditiva Neurossensorial/genética , Mutação de Sentido Incorreto , Adulto , Idoso , Análise Mutacional de DNA , Feminino , Proteínas Ligadas por GPI/genética , Genes Dominantes , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo
20.
Audiol Neurootol ; 19(5): 319-26, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25358692

RESUMO

Mutations in the SLC26A4 gene, which encodes pendrin, cause congenital hearing loss as a manifestation of Pendred syndrome (PS) with an iodide organification defect or nonsyndromic enlarged vestibular aqueduct (NSEVA, DFNB4). There have been reports of differences between PS and NSEVA, including their auditory phenotypes and molecular genetic bases. For appropriate genetic diagnosis and counseling, it is important to functionally characterize SLC26A4 variants. In this study, we identified and evaluated a novel null mutation of SLC26A4 and report our method of assessing the pathogenic potential of mutations in SLC26A4, one of the most frequent causative genes of deafness in humans. A 3-year-old female with progressive sensorineural hearing loss and her parents were recruited. They underwent clinical, audiological, radiological and genetic evaluations, which revealed that the female patient had an enlarged vestibular aqueduct and an incomplete partition type II anomaly in the cochlea bilaterally. Sanger sequencing of the SLC26A4 gene was also performed. For a confirmatory genetic diagnosis, we first characterized the anion/base exchange ability of mutant pendrin products in HEK 293 cells and, if necessary, evaluated whether the mutant pendrin traffics to the plasma membrane in COS-7 cells. We also expressed a null function mutant, p.H723R, and a previously documented polymorphism, p.P542R, as controls. The pure tone average was 66 dB HL in the right ear and 75 dB HL in the left ear. Sequencing of SLC26A4 revealed a known pathogenic mutation (p.H723R) and a novel missense variant (p.V510D) as a compound heterozygote. When we expressed the p.V510D mutant pendrin in mammalian cells, the rate constants for Cl-/HCO3- exchange were 10.96±4.79% compared with those of wild-type pendrin. This figure was comparable to that of p.H723R, indicating p.V510D to be another pathogenic mutation with a null function. The p.V510D pendrin product was shown to be entrapped in the endoplasmic reticulum (ER) at 24-30 h after transfection, and not trafficked to the plasma membrane in COS-7 cells, suggesting retention in the ER and abnormal trafficking as the pathogenic mechanism. This was similar to p.H723R, which is a null function founder mutant in this population but is a candidate variant for future drug therapy to rescue the abnormal cell trafficking. Impaired cellular trafficking due to ER retention and abolished exchange activity of the newly detected p.V510D indicates the pathogenic potential of this variant. These missense variants may be good candidate variants for drug therapy if the intrinsic exchange activity is not damaged by the change.


Assuntos
Movimento Celular/genética , Perda Auditiva Neurossensorial/genética , Proteínas de Membrana Transportadoras/genética , Aqueduto Vestibular/anormalidades , Animais , Audiometria de Tons Puros , Células COS , Pré-Escolar , Chlorocebus aethiops/genética , Retículo Endoplasmático , Feminino , Células HEK293 , Perda Auditiva Neurossensorial/fisiopatologia , Heterozigoto , Humanos , Mutação de Sentido Incorreto , Linhagem , Análise de Sequência de DNA , Transportadores de Sulfato , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA