Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35041619

RESUMO

Defective primary cilia cause a range of diseases called ciliopathies, which include hearing loss (HL). Variants in the human oxysterol-binding protein like 2 (OSBPL2/ORP2) are responsible for autosomal dominant nonsyndromic HL (DFNA67). However, the pathogenesis of OSBPL2 deficiency has not been fully elucidated. In this study, we show that the Osbpl2-KO mice exhibited progressive HL and abnormal cochlear development with defective cilia. Further research revealed that OSBPL2 was located at the base of the kinocilia in hair cells (HCs) and primary cilia in supporting cells (SCs) and functioned in the maintenance of ciliogenesis by regulating the homeostasis of PI(4,5)P2 (phosphatidylinositol 4,5-bisphosphate) on the cilia membrane. OSBPL2 deficiency led to a significant increase of PI(4,5)P2 on the cilia membrane, which could be partially rescued by the overexpression of INPP5E. In addition, smoothened and GL13, the key molecules in the Sonic Hedgehog (Shh) signaling pathway, were detected to be downregulated in Osbpl2-KO HEI-OC1 cells. Our findings revealed that OSBPL2 deficiency resulted in ciliary defects and abnormal Shh signaling transduction in auditory cells, which helped to elucidate the underlying mechanism of OSBPL2 deficiency in HL.


Assuntos
DNA/genética , Células Ciliadas Auditivas/patologia , Perda Auditiva/genética , Proteínas Hedgehog/genética , Mutação , Receptores de Esteroides/genética , Animais , Análise Mutacional de DNA , Modelos Animais de Doenças , Células Ciliadas Auditivas/metabolismo , Perda Auditiva/metabolismo , Perda Auditiva/patologia , Proteínas Hedgehog/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Esteroides/metabolismo , Transdução de Sinais
2.
Biochem Biophys Res Commun ; 574: 1-7, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34418635

RESUMO

GPRASP2 is implicated in nervous system diseases, tumors and immune inflammation. In our previous study, G protein-coupled receptor associated sorting protein 2 (GPRASP2) was identified as a novel causal gene for X-linked recessive syndromic hearing loss (SHL). However, the role of GPRASP2 in auditory function has not been elucidated. The Gprasp2-knockout (KO) mouse HEI-OC1 auditory cells were constructed using CRISPR/Cas9-mediated gene editing. RNA-sequencing (RNA-seq) was used to investigate the differentially expressed genes (DEGs) and DEGs-enriched signaling pathways, which was verified by Western blot. Flow cytometry assay was used to examine cell apoptosis. The cytological pathology was evaluated by laser scanning confocal microscopy (LSCM) and transmission electron microscopy (TEM). Mitochondrial damage was observed in Gprasp2-KO HEI-OC1 cells. RNA-seq analysis suggested that Gprasp2-KO was implicated in the apoptosis process, which could be mediated by Hedgehog (Hh) signaling pathway. The key molecules in Hh signaling pathway (Smo, Gli1, Gli2) were detected to be down-regulated in Gprasp2-KO HEI-OC1 cells. The differential expression of apoptosis molecules (Bcl2, Bax, Caspase-3/cleaved-Caspase-3) indicated that Gprasp2-KO induced apoptosis in HEI-OC1 cells. The treatment of smoothened agonist (Purmorphamine, PUR) activated the Hh-Gli signaling pathway and reduced apoptosis in Gprasp2-KO HEI-OC1 cells. This study revealed that Gprasp2-disruption inhibited Hh signaling pathway and led to cell apoptosis in HEI-OC1 cells, which might provide the potential molecular mechanism of GPRASP2 mutation associated with human SHL.


Assuntos
Apoptose , Regulação para Baixo , Proteínas Hedgehog/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Células Cultivadas , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais
3.
iScience ; 23(7): 101252, 2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32650117

RESUMO

The accumulation of giant lipid droplets (LDs) increases the risk of metabolic disorders including obesity and insulin resistance. The lipolysis process involves the activation and transfer of lipase, but the molecular mechanism is not completely understood. The translocation of ATGL, a critical lipolysis lipase, from the ER to the LD surface is mediated by an energy catabolism complex. Oxysterol-binding protein-like 2 (OSBPL2/ORP2) is one of the lipid transfer proteins that regulates intracellular cholesterol homeostasis. A recent study has proven that Osbpl2-/- pigs exhibit hypercholesterolemia and obesity phenotypes with an increase in adipocytes. In this study, we identified that OSBPL2 links the endoplasmic reticulum (ER) with LDs, binds to COPB1, and mediates ATGL transport. We provide important insights into the function of OSBPL2, indicating that it is required for the regulation of lipid droplet lipolysis.

4.
Cell Death Dis ; 10(9): 627, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31427568

RESUMO

Oxysterol-binding protein like 2 (OSBPL2) was identified as a novel causal gene for autosomal dominant nonsyndromic hearing loss. However, the pathogenesis of OSBPL2 deficits in ADNSHL was still unclear. The function of OSBPL2 as a lipid-sensing regulator in multiple cellular processes suggested that OSBPL2 might play an important role in the regulation of cholesterol-homeostasis, which was essential for inner ear. In this study the potential roles of OSBPL2 in cholesterol biosynthesis and ROS production were investigated in Osbpl2-KO OC1 cells and osbpl2b-KO zebrafish. RNA-seq-based analysis suggested that OSBPL2 was implicated in cholesterol biosynthesis and AMPK signaling pathway. Furthermore, Osbpl2/osbpl2b-KO resulted in a reduction of AMPK activity and up-regulation of Srebp2/srebp2, Hmgcr/hmgcr and Hmgcs1/hmgcs1, key genes in the sterol biosynthetic pathway and associated with AMPK signaling. In addition, OSBPL2 was also found to interact with ATIC, key activator of AMPK. The levels of total cholesterol and ROS in OC1 cells or zebrafish inner ear were both increased in Osbpl2/osbpl2b-KO mutants and the mitochondrial damage was detected in Osbpl2-KO OC1 cells. This study uncovered the regulatory roles of OSBPL2 in cellular cholesterol biosynthesis and ROS production. These founds might contribute to the deep understanding of the pathogenesis of OSBPL2 mutation in ADNSHL.


Assuntos
Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Colesterol/biossíntese , Perda Auditiva/genética , Espécies Reativas de Oxigênio/metabolismo , Receptores de Esteroides/genética , Proteínas de Peixe-Zebra/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Deleção de Genes , Técnicas de Inativação de Genes , Células HEK293 , Perda Auditiva/metabolismo , Humanos , Receptores de Esteroides/metabolismo , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
5.
J Genet Genomics ; 46(8): 379-387, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31451425

RESUMO

Oxysterol binding protein like 2 (OSBPL2), an important regulator in cellular lipid metabolism and transport, was identified as a novel deafness-causal gene in our previous work. To resemble the phenotypic features of OSBPL2 mutation in animal models and elucidate the potential genotype-phenotype associations, the OSBPL2-disrupted Bama miniature (BM) pig model was constructed using CRISPR/Cas9-mediated gene editing, somatic cell nuclear transfer (SCNT) and embryo transplantation approaches, and then subjected to phenotypic characterization of auditory function and serum lipid profiles. The OSBPL2-disrupted pigs displayed progressive hearing loss (HL) with degeneration/apoptosis of cochlea hair cells (HCs) and morphological abnormalities in HC stereocilia, as well as hypercholesterolaemia. High-fat diet (HFD) feeding aggravated the development of HL and led to more severe hypercholesterolaemia. The dual phenotypes of progressive HL and hypercholesterolaemia resembled in OSBPL2-disrupted pigs confirmed the implication of OSBPL2 mutation in nonsydromic hearing loss (NSHL) and contributed to the potential linkage between auditory dysfunction and dyslipidaemia/hypercholesterolaemia.


Assuntos
Predisposição Genética para Doença , Perda Auditiva/genética , Hipercolesterolemia/genética , Mutação , Receptores de Esteroides/genética , Alelos , Animais , Sistemas CRISPR-Cas , Modelos Animais de Doenças , Transferência Embrionária , Estudos de Associação Genética , Ligação Genética , Loci Gênicos , Genótipo , Perda Auditiva/diagnóstico , Humanos , Hipercolesterolemia/diagnóstico , Fenótipo , Suínos , Porco Miniatura
6.
Exp Cell Res ; 383(2): 111512, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31356817

RESUMO

Previous studies have shown that oxysterol binding protein like 2 (OSBPL2) knockdown is closely related to cholesterol metabolism. However, whether there is a direct relation between OSBPL2 and cholesterol synthesis is unknown. This study explored the mechanism of OSBPL2 deficiency in the upregulation of squalene epoxidase (SQLE) and the subsequent accumulation of intracellular cholesterol and cholesteryl ester. Here, we constructed an OSBPL2-deleted HeLa cell line using CRISPR/Cas9 technology, screened differentially expressed genes and examined the transcriptional regulation of SQLE using a dual-luciferase reporter gene. RNA-seq analysis showed that SQLE was upregulated significantly and the dual luciferase reporter gene assay revealed that two new functional transcription factor binding sites of Sp1 transcription factor (SP1) and sterol regulatory element-binding transcription factor 2 (SREBF2) in the SQLE promoter participated in the SQLE transcription and expression. In addition, we also observed that OSBPL2 deletion inhibited the AMPK signalling pathway and that the inhibition of AMPK signalling promoted SP1 and SREBF2 entry into the nuclear to upregulate SQLE expression. Therefore, these data support that OSBPL2 deficiency upregulates SQLE expression and increases the accumulation of cholesterol and cholesteryl ester by suppressing AMPK signalling, which provides new evidence of the connection between OSBPL2 and cholesterol synthesis.


Assuntos
Adenilato Quinase/metabolismo , Ésteres do Colesterol/biossíntese , Colesterol/biossíntese , Receptores de Esteroides/genética , Fator de Transcrição Sp1/metabolismo , Esqualeno Mono-Oxigenase/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Deleção de Genes , Regulação Enzimológica da Expressão Gênica/genética , Células HEK293 , Células HeLa , Humanos , Redes e Vias Metabólicas/genética , Transporte Proteico/genética , Receptores de Esteroides/fisiologia , Esqualeno Mono-Oxigenase/metabolismo , Regulação para Cima/genética
7.
J Steroid Biochem Mol Biol ; 187: 17-26, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30391516

RESUMO

Oxysterol Binding Protein Like 2 (OSBPL2) is a lipid-binding protein implicated in various cellular processes. Previous studies have shown that depression of OSBPL2 significantly increases the level of cellular 25-hydroxycholesterol (25-OHC) which regulates the expression of lipid-metabolism-related genes. However, whether 25-OHC can regulate the expression of OSBPL2 remains unanswered. This study aimed to explore the molecular mechanism of 25-OHC regulating the expression of OSBPL2. Using dual-luciferase reporter assay, we found a decrease of nuclear transcription factor Y subunit alpha (NFYA) bound with OSBPL2 promoter when HeLa cells were treated with 25-OHC. Furthermore, transcriptome sequencing and RNA interference results revealed that the p53/sterol regulatory element binding transcription factor 2 (SREBF2) signaling pathway was involved in the NFYA-dependent transcription of OSBPL2 induced by 25-OHC. Based on these results, we concluded that pleomorphic adenoma gene 1 (PLAG1) and NFYA participated in the basal transcription of OSBPL2 and that 25-OHC decreased the transcription of OSBPL2 via the p53/SREBF2/NFYA signaling pathway. 25-OHC will accumulate over time in OSBPL2 knockdown cells. These results may provide a new insight into the deafness caused by OSBPL2 mutation.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Regulação para Baixo , Hidroxicolesteróis/metabolismo , Receptores de Esteroides/genética , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células HeLa , Humanos , Regiões Promotoras Genéticas
8.
Int J Pediatr Otorhinolaryngol ; 105: 6-11, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29447821

RESUMO

OBJECTIVE: The CLDN14 gene, encoding the tight junction protein Claudin-14, has been proposed as a candidate causative gene affecting autosomal recessive non-syndromic hearing loss (ARNSHL). Genetic analysis of nonsynonymous single-nucleotide variations (nsSNVs) in CLDN14 has been performed in different populations. The role of CLDN14 nsSNVs in contributing to hearing loss in Chinese populations would be investigated in this study. METHODS: Target screening for CLDN14 variations were conducted in 500 unrelated patients diagnosed with non-syndromic hearing loss (NSHL). RESULTS: No reported pathogenic CLDN14 nsSNVs in heterozygote or homozygote were detected in this study, however, we identified 4 heterozygous nsSNVs [c.11C > T, p.(Thr4Met); c.16G > A, p.(Val6Met); c.68T > C, p.(Ile23Thr); c.367A > C, p.(Thr123Pro)] in CLDN14. The 4 nsSNVs are located at claudin-14 transmembrane domains, but assessed to be poorly conservative and non-pathogenic via multiple in silico algorithms. The structure-based analysis also suggested that the 4 nsSNVs had less structural and functional impact on claudin-14. CONCLUSION: Our findings indicated that CLDN14 might not be a major causative gene for NSHL in Chinese populations, which would contribute to fully understanding the genetic cause of NSHL in the East Asian populations.


Assuntos
Claudinas/genética , Surdez/genética , Povo Asiático/genética , Feminino , Testes Genéticos , Humanos , Masculino , Mutação , Linhagem , Polimorfismo Genético
9.
Mol Med Rep ; 16(5): 5931-5937, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28849214

RESUMO

G protein­coupled receptor­associated sorting protein 2 (GPRASP2), a member of the GASP family, has been reported to be involved in the modulation of transcription. However, few studies have revealed the role of GPRASP2 in the development and progression of diseases. As a model organism, zebrafish have been widely used to investigate human diseases. In the present study, zebrafish armadillo repeat­containing 10 (armc10), an orthologous gene of human GPRASP2 was identified, and the spatial and temporal expression patterns of armc10 in zebrafish during early embryonic development were revealed. Bioinformatics analyses showed that ARMC10 protein sequences were highly conserved. Reverse transcription polymerase chain reaction analysis and whole mount in situ hybridization revealed that zebrafish armc10 was maternally expressed and was detected at a weak level up to 12 h post­fertilization (hpf), however, its expression increased to a high level at 24 hpf. At the 75% epiboly stage and 12 hpf, armc10 was widely expressed in the embryo. At 24 hpf, armc10 mRNA was expressed in the nervous system of the zebrafish head. When the embryo was 2 days old, the wide expression of armc10 was maintained in the nervous system of the zebrafish head. At 72 hpf, the mRNA expression of armc10 was located specifically in the otic vesicles in addition to the nervous system of the head. At 96 hpf, the expression of armc10 was maintained in the otic vesicles and the nervous system of the head. The results of the present study provided novel insight into the spatial and temporal mRNA expression of armc10 in zebrafish, for the further investigation of nervous system diseases.


Assuntos
Proteínas do Domínio Armadillo/genética , Proteínas de Transporte/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , Proteínas de Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Proteínas do Domínio Armadillo/metabolismo , Proteínas de Transporte/metabolismo , Sequência Conservada , Embrião não Mamífero , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
10.
J Med Genet ; 54(6): 426-430, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28096187

RESUMO

BACKGROUND: A substantial amount of nuclear genes have been identified to be implicated in genetic hearing loss, while X-linked hearing loss is genetically heterogeneous and relatively infrequent. OBJECTIVE: To identify the causative gene mutation in a five-generation Chinese family with an X-linked recessive syndromic hearing loss (SHL). METHODS: Targeted X-chromosome exome sequencing was conducted, and cosegregation analysis was performed in the members of the affected family. The in silico and expression studies were also performed. RESULTS: A 2-bp missense mutation (c.1717_1718GC>AA, p.A573N) in the G protein-coupled receptor associated sorting protein 2 (GPRASP2) gene was identified in four hemizygous male patients and two heterozygous female carriers, which was cosegregated with the clinical phenotypes in this family. In silico analysis supported that this gene mutation is functionally deleterious, and it was detected that homologous Gprasp2 was highly expressed in multiple structures of the mouse cochlea, which suggested that GPRASP2 might be the genetic cause for the described disease phenotypes. CONCLUSIONS: This study presented a novel X-linked SHL combined with unique and unrecognised clinical features, and a missense variation of GPRASP2 was first identified to be implicated in X-linked SHL.


Assuntos
Proteínas de Transporte/genética , Surdez/genética , Genes Ligados ao Cromossomo X/genética , Predisposição Genética para Doença/genética , Perda Auditiva/genética , Mutação de Sentido Incorreto/genética , Sequência de Aminoácidos , Povo Asiático/genética , Exoma/genética , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Linhagem , Fenótipo
11.
Int J Pediatr Otorhinolaryngol ; 84: 174-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27063776

RESUMO

OBJECTIVE: The mutated OSBPL2 (OMIM: 606731), encoding oxysterol binding protein-like protein 2, was recently identified as a novel causative gene for autosomal dominant nonsyndromic hearing loss (ADNSHL). We reported the expression patterns of Osbpl2 in zebrafish, in order to further understand the role of OSBPL2 in hearing formation and development. METHODS: Zebrafish was used as an animal model, and the expression of Osbpl2 was investigated by whole mount in situ hybridization. RESULTS: Bioinformatics analysis indicates that zebrafish has two homologues of Osbpl2 gene (Osbpl2a and Osbpl2b) and Osbpl2b is the orthologous gene of human OSBPL2. No expression of Osbpl2a and Osbpl2b mRNA is detected at 75% epiboly. The zygotical expression of the two genes has not been started at 11-somite stage. At 24h post-fertilization (hpf), both Osbpl2a and Osbpl2b are found at ventricle zone of brain, however, the expression level of Osbpl2a is higher than that of Osbpl2b. When embryos are 48hpf, the expression level of Osbpl2a and Osbpl2b becomes higher at the ventricle zone. At 72hpf, Osbpl2b is only found at liver primordium, while Osbpl2a is not detected anywhere obviously. At 96hpf, Osbpl2b is found at pharyngeal arches, liver, digestive tract and otic vesicle, while Osbpl2a remains undetected. CONCLUSION: Osbpl2b was demonstrated to be the orthologous gene of human OSBPL2, which has strong maternal expression, while Osbpl2a was detected without obvious maternal expression. This work would contribute to the further study of the molecular mechanism and function of OSBPL2 implicated with ADNSHL.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Perda Auditiva Neurossensorial/embriologia , Receptores de Esteroides/genética , Proteínas de Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Feminino , Marcadores Genéticos , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Humanos , Hibridização In Situ , Dados de Sequência Molecular , Receptores de Esteroides/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
12.
Medicine (Baltimore) ; 94(50): e2248, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26683941

RESUMO

Many SLC26A4 mutations have been identified in patients with nonsyndromic enlarged vestibular aqueduct (EVA). However, the roles of SLC26A4 genotypes and phenotypes in hereditary deafness remain unexplained. This study aims to perform a meta-analysis based on the PRISMA statement to evaluate the diagnostic value of SLC26A4 mutant alleles and their correlations with multiethnic hearing phenotypes in EVA patients. The systematic literature search of the PubMed, Wiley Online Library, EMBASE, Web of Science, and Science Direct databases was conducted in English for articles published before July 15, 2015. Two investigators independently reviewed retrieved literature and evaluated eligibility. Discrepancy was resolved by discussion and a third investigator. Quality of included studies was evaluated using Newcastle-Ottawa Quality Assessment Scale. Data were synthesized using random-effect or fixed-effect models. The effect sizes were estimated by measuring odds ratios (ORs) with 95% confidence interval (CI). Twenty-five eligible studies involved 2294 cases with EVA data. A total of 272 SLC26A4 variations were found in deafness with EVA and 26 mutations of SCL26A4 had higher frequency. The overall OR was 646.71 (95% CI: 383.30-1091.15, P = 0.000). A total of 22 mutants were considered statistically significant in all ethnicities (ORs >1, P < 0.05). In particular, 8 mutants were specificity of EVA phenotypes in mutations of SLC26A4 for Asia deafness populations (ORs >1, P < 0.05), 4 mutants for Europe and North America (ORs >1, P < 0.05), and the IVS7-2A>G mutations in SLC26A4 were found to have the highest frequency in deafness individuals with EVA phenotype (62.42%). Moreover, subgroups for studies limited to cases with EVA phenotype, 11 mutants relevant risks (RRs) were P < 0.05, especially for IVS7-2A>G bi-allelic mutants assayed in a deafness population (RR = 0.880, P = 0.000). Diagnostic accuracy of SLC26A4 mutation results also identified the significant association of IVS7-2A>G (AUC = 0.99, 95% CI: 0.97-0.99) and p.H723R (AUC = 0.99, 95% CI: 0.98-1.00) detecting deafness with EVA. To conclude, the IVS7-2A>G and H723R in SLC26A4 present a significant predicting value and discriminatory ability for clinical use on diagnosis of EVA within a deafness population.


Assuntos
Surdez/genética , Perda Auditiva Neurossensorial/genética , Proteínas de Membrana Transportadoras/genética , Mutação , Aqueduto Vestibular/anormalidades , Humanos , Transportadores de Sulfato
13.
Sci Rep ; 5: 14731, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26435059

RESUMO

Wolfram syndrome (WS) is a rare, progressive, neurodegenerative disorder that has an autosomal recessive pattern of inheritance. The gene for WS, wolfram syndrome 1 gene (WFS1), is located on human chromosome 4p16.1 and encodes a transmembrane protein. To date, approximately 230 mutations in WFS1 have been confirmed, in which nonsynonymous single nucleotide polymorphisms (nsSNPs) are the most common forms of genetic variation. Nonetheless, there is poor knowledge on the relationship between SNP genotype and phenotype in other nsSNPs of the WFS1 gene. Here, we analysed 395 nsSNPs associated with the WFS1 gene using different computational methods and identified 20 nsSNPs to be potentially pathogenic. Furthermore, to identify the amino acid distributions and significances of pathogenic nsSNPs in the protein of WFS1, its transmembrane domain was constructed by the TMHMM server, which suggested that mutations outside of the TMhelix could have more effects on protein function. The predicted pathogenic mutations for the nsSNPs of the WFS1 gene provide an excellent guide for screening pathogenic mutations.


Assuntos
Proteínas de Membrana/genética , Síndrome de Wolfram/genética , Simulação por Computador , Estudos de Associação Genética , Humanos , Proteínas de Membrana/química , Modelos Moleculares , Mutação de Sentido Incorreto , Fenótipo , Polimorfismo de Nucleotídeo Único , Estrutura Terciária de Proteína
14.
Chin Med J (Engl) ; 128(18): 2510-5, 2015 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-26365971

RESUMO

BACKGROUND: Nonsyndromic hearing loss (NSHL) is highly heterogeneous, in which more than 90 causative genes have currently been identified. DFNA5 is one of the deafness genes that known to cause autosomal dominant NSHL. Until date, only five DFNA5 mutations have been described in eight families worldwide. In this study, we reported the identification of a novel pathogenic mutation causing DFNA5 deafness in a five-generation Chinese family. METHODS: After detailed clinical evaluations of this family, the genomic DNA of three affected individuals was selected for targeted exome sequencing of 101 known deafness genes, as well as mitochondrial DNA and microRNA regions. Co-segregation analysis between the hearing loss and the candidate variant was confirmed in available family members by direct polymerase chain reaction (PCR)-Sanger sequencing. Real-time PCR (RT-PCR) was performed to investigate the potential effect of the pathogenic mutation on messenger RNA splicing. RESULTS: Clinical evaluations revealed a similar deafness phenotype in this family to that of previously reported DFNA5 families with autosomal dominant, late-onset hearing loss. Molecular analysis identified a novel splice site mutation in DFNA5 intron 8 (IVS8+1 delG). The mutation segregated with the hearing loss of the family and was absent in 120 unrelated control DNA samples of Chinese origin. RT-PCR showed skipping of exon 8 in the mutant transcript. CONCLUSIONS: We identified a novel DFNA5 mutation IVS8+1 delG in a Chinese family which led to skipping of exon 8. This is the sixth DFNA5 mutation relates to hearing loss and the second one in DFNA5 intron 8. Our findings provide further support to the hypothesis that the DFNA5-associated hearing loss represents a mechanism of gain-of-function.


Assuntos
Surdez/genética , Perda Auditiva Neurossensorial/genética , Perda Auditiva/genética , Adulto , Éxons/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Adulto Jovem
15.
Sci Rep ; 5: 10831, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26035154

RESUMO

A Chinese family was identified with clinical features of enlarged vestibular aqueduct syndrome (EVAS). The mutational analysis showed that the proband (III-2) had EVAS with bilateral sensorineural hearing loss and carried a rare compound heterozygous mutation of SLC26A4 (IVS7-2A>G, c.2167C>G), which was inherited from the same mutant alleles of IVS7-2A>G heterozygous father and c.2167C>G heterozygous mother. Compared with another confirmed pathogenic biallelic mutation in SLC26A4 (IVS7-2A>G, c.2168A>G), these two biallelic mutations shared one common mutant allele and the same codon of the other mutant allele, but led to different changes of amino acid (p.H723D, p.H723R) and both resulted in the deafness phenotype. Structure-modeling indicated that these two mutant alleles changed the shape of pendrin protein encoded by SLC26A4 with increasing randomness in conformation, and might impair pendrin's ability as an anion transporter. The molecular dynamics simulations also revealed that the stability of mutant pendrins was reduced with increased flexibility of backbone atoms, which was consistent with the structure-modeling results. These evidences indicated that codon 723 was a hot-spot region in SLC26A4 with a significant impact on the structure and function of pendrin, and acted as one of the genetic factors responsible for the development of hearing loss.


Assuntos
Códon , Surdez/diagnóstico , Surdez/genética , Heterozigoto , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Mutação , Fenótipo , Sequência de Aminoácidos , Análise Mutacional de DNA , Feminino , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Linhagem , Conformação Proteica , Relação Quantitativa Estrutura-Atividade , Alinhamento de Sequência , Transportadores de Sulfato
16.
Artigo em Inglês | MEDLINE | ID: mdl-25791913

RESUMO

AIMS: Compared with those in other head and neck regions, schwannomas in the nasal cavity or paranasal sinuses are rare. The aim of this study was to present the experience of the authors in 11 schwannoma cases of the sinonasal tract and pterygopalatine fossa over a decade. METHODS: A retrospective study from 2003 to 2014. RESULTS: Three female and 8 male patients from 22 to 61 years of age (mean age 42 years) were admitted. The most common complaints were unilateral nasal congestion. A total of 10 of the patients received surgery, including 6 functional endoscopic sinus surgeries (FESS). The postoperative course was generally uneventful. Among the patients, 10 remained regionally asymptomatic, and there has been no clinical or radiological evidence of recurrence or residual tumor. CONCLUSION: Surgical treatment is effective for schwannomas of the sinonasal tract and the pterygopalatine fossa with a low recurrence rate. Conducting CT and MRI (particularly fluid-attenuated inversion recovery) before surgery is mandatory. FESS could become the primary treatment of choice.


Assuntos
Cavidade Nasal/patologia , Neurilemoma/cirurgia , Neoplasias Nasais/patologia , Neoplasias dos Seios Paranasais/cirurgia , Seios Paranasais/patologia , Fossa Pterigopalatina/patologia , Adulto , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Cavidade Nasal/cirurgia , Recidiva Local de Neoplasia , Neoplasias Nasais/cirurgia , Neoplasias dos Seios Paranasais/patologia , Seios Paranasais/cirurgia , Estudos Retrospectivos , Tomografia Computadorizada por Raios X , Adulto Jovem
17.
Genet Med ; 17(3): 210-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25077649

RESUMO

PURPOSE: Various forms of hearing loss have genetic causes, but many of the responsible genes have not yet been identified. Here, we describe a large seven-generation Chinese family with autosomal dominant nonsyndromic hearing loss that has been excluded as being caused by known deafness gene mutations associated with autosomal dominant nonsyndromic hearing loss with the aim of identifying a novel causative gene involved in deafness. METHODS: Whole-exome sequencing was conducted in three affected family members, and cosegregation analysis was performed on other members of the family. RESULTS: Whole-exome sequencing and subsequent segregation analysis identified a heterozygous frameshift mutation (c.153_154delCT, p.Gln53Argfs*100) in the oxysterol binding protein-like 2 (OSBPL2) gene in 25 affected family members. The deletion mutation is predicted to lead to premature truncation of the OSBPL2 protein. Modeling and structure-based analysis support the theory that this gene deletion is functionally deleterious. Our finding was further confirmed by the detection of another missense mutation, a c.583C>A transversion (p.Leu195Met) in exon 7 of OSBPL2, in an additional sporadic case of deafness. CONCLUSION: Based on this study, OSBPL2 was identified as an excellent novel candidate gene for autosomal dominant nonsyndromic hearing loss; this study is the first to implicate OSBPL2 mutations in autosomal dominant nonsyndromic hearing loss.


Assuntos
Povo Asiático/genética , Cóclea/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Análise de Sequência de DNA/métodos , Animais , Povo Asiático/etnologia , China , Surdez/genética , Exoma , Feminino , Mutação da Fase de Leitura , Estudos de Associação Genética/métodos , Predisposição Genética para Doença , Humanos , Masculino , Camundongos , Mutação de Sentido Incorreto , Linhagem , Receptores de Esteroides/química
18.
Artigo em Inglês | MEDLINE | ID: mdl-29204539

RESUMO

OBJECTIVE: To explore the feasibility of the submental island flap in the repair of hypopharyngeal defects. METHODS: We collected wet specimens of fresh cadaveric heads from the Han Chinese adult population for applied anatomy of the submental island flap, and followed five patients with pyriform sinus carcinoma after reconstruction surgery using submental island flaps. RESULTS: We found that the average length and width of the submental island flaps were (65.20 ± 11.69) mm and (46.70 ± 6.59) mm, respectively. The skin flap in all five patients survived after surgery, and tracheal tubes and gastric tubes were removed 7-36 days after surgery. Patients were followed up for 24-42 months, pharyngeal flaps grew well, and speech and swallowing functions were satisfactory. CONCLUSION: The submental island flap is a preferred material for the repair of hypopharyngeal defects after hypopharyngeal carcinoma resection, because of good blood supply, easy harvesting, and high survival rate.

19.
Genetica ; 142(6): 555-62, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25447126

RESUMO

Connexins (Cxs) were first identified as subunit proteins of the intercellular membrane channels that cluster in the cell communication structures known as gap junctions. Mutations in the gap junction ß2 (GJB2) gene encoding connexin 26 (Cx26) have been linked to sporadic and hereditary hearing loss. In some cases, the mechanisms through which these mutations lead to hearing loss have been partly elucidated using cell culture systems and animal models. The goal of this study was to re-assess the pathogenic roles of the GJB2 mutations by combining comparative evolutionary studies. We used Bayesian phylogenetic analyses to determine the relationships among 35 orthologs and to calculate the ancestral sequences of these orthologs. By aligning sequences from the 35 orthologs and their ancestors and categorizing amino acid sites by degree of conservation, we used comparative evolutionary methods to determine potential functionally important amino acid sites in Cx26 and to identify missense changes that are likely to affect function. We identified six conserved regions in Cx26, five of which are located in the Connexin_CCC, and another is in the connexin super family domain. Finally, we identified 51 missense changes that are likely to disrupt function, and the probability of these changes occurring at hydrophilic amino acid residues was twice that of occurring at hydrophobic residues in the trans-membrane regions of Cx26. Our findings, which were obtained by combining comparative evolutionary methods to predict Cx26 mutant function, are consistent with the pathogenic characteristics of Cx26 mutants. This study provides a new pathway for studying the role of aberrant Cx26 in hereditary hearing loss.


Assuntos
Conexinas/genética , Evolução Molecular , Perda Auditiva Neurossensorial/genética , Mutação de Sentido Incorreto , Sequência de Aminoácidos , Animais , Teorema de Bayes , Conexina 26 , Análise Mutacional de DNA , Humanos , Dados de Sequência Molecular , Filogenia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
20.
J Transl Med ; 12: 311, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25388789

RESUMO

BACKGROUND: Hereditary hearing loss is genetically heterogeneous, and hundreds of mutations in than 60 genes are involved in this disease. Therefore, it is difficult to identify the causative gene mutations involved. In this study, we combined targeted genomic capture and massively parallel sequencing (MPS) to address this issue. METHODS: Using targeted genomic capture and MPS, 104 genes and three microRNA regions were selected and simultaneously sequenced in 23 unrelated probands of Chinese families with nonsyndromic hearing loss. The results were validated by Sanger sequencing for all available members of the probands' families. To analyze the possible pathogenic functional effects of the variants, three types of prediction programs (Mutation Taster, PROVEAN and SIFT) were used. A total of 195 healthy Chinese Han individuals were compared as controls to verify the novel causative mutations. RESULTS: Of the 23 probands, six had mutations in DFNA genes [WFS1 (n = 2), COCH, ACTG1, TMC1, and POU4F3] known to cause autosomal dominant nonsyndromic hearing loss. These included one novel in-frame indel mutation, three novel missense mutations and two reported missense mutations. Furthermore, one proband from a family with recessive DFNB carried two monoallelic mutations in the GJB2 and USH2A genes. All of these mutations co-segregated with the hearing loss phenotype in 36 affected individuals from 7 families and were predicted to be pathogenic. CONCLUSIONS: Mutations in uncommon deafness genes contribute to a portion of nonsyndromic deafness cases. In the future, critical gene mutations may be accurately and quickly identified in families with hereditary hearing loss by targeted genomic capture and MPS.


Assuntos
Genoma Humano , Perda Auditiva/genética , Conexina 26 , Conexinas , Etnicidade , Feminino , Humanos , Masculino , Mutação , Linhagem , Análise de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...