Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 15(11): e17611, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37691516

RESUMO

Cingulin (CGN) is a cytoskeleton-associated protein localized at the apical junctions of epithelial cells. CGN interacts with major cytoskeletal filaments and regulates RhoA activity. However, physiological roles of CGN in development and human diseases are currently unknown. Here, we report a multi-generation family presenting with autosomal dominant non-syndromic hearing loss (ADNSHL) that co-segregates with a CGN heterozygous truncating variant, c.3330delG (p.Leu1110Leufs*17). CGN is normally expressed at the apical cell junctions of the organ of Corti, with enriched localization at hair cell cuticular plates and circumferential belts. In mice, the putative disease-causing mutation results in reduced expression and abnormal subcellular localization of the CGN protein, abolishes its actin polymerization activity, and impairs the normal morphology of hair cell cuticular plates and hair bundles. Hair cell-specific Cgn knockout leads to high-frequency hearing loss. Importantly, Cgn mutation knockin mice display noise-sensitive, progressive hearing loss and outer hair cell degeneration. In summary, we identify CGN c.3330delG as a pathogenic variant for ADNSHL and reveal essential roles of CGN in the maintenance of cochlear hair cell structures and auditory function.


Assuntos
Surdez , Perda Auditiva , Animais , Humanos , Camundongos , Proteínas do Citoesqueleto , Surdez/genética , Células Ciliadas Auditivas/metabolismo , Audição/fisiologia , Perda Auditiva/genética , Perda Auditiva/metabolismo
2.
PLoS Genet ; 16(9): e1009040, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32970669

RESUMO

Genetic hearing loss is a common health problem with no effective therapy currently available. DFNA15, caused by mutations of the transcription factor POU4F3, is one of the most common forms of autosomal dominant non-syndromic deafness. In this study, we established a novel mouse model of the human DFNA15 deafness, with a Pou4f3 gene mutation (Pou4f3Δ) identical to that found in a familial case of DFNA15. The Pou4f3(Δ/+) mice suffered progressive deafness in a similar manner to the DFNA15 patients. Hair cells in the Pou4f3(Δ/+) cochlea displayed significant stereociliary and mitochondrial pathologies, with apparent loss of outer hair cells. Progression of hearing and outer hair cell loss of the Pou4f3(Δ/+) mice was significantly modified by other genetic and environmental factors. Using Pou4f3(-/+) heterozygous knockout mice, we also showed that DFNA15 is likely caused by haploinsufficiency of the Pou4f3 gene. Importantly, inhibition of retinoic acid signaling by the aldehyde dehydrogenase (Aldh) and retinoic acid receptor inhibitors promoted Pou4f3 expression in the cochlear tissue and suppressed the progression of hearing loss in the mutant mice. These data demonstrate Pou4f3 haploinsufficiency as the main underlying cause of human DFNA15 deafness and highlight the therapeutic potential of Aldh inhibitors for treatment of progressive hearing loss.


Assuntos
Aldeído Desidrogenase/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Células Ciliadas Auditivas/patologia , Perda Auditiva/tratamento farmacológico , Perda Auditiva/etiologia , Proteínas de Homeodomínio/genética , Fator de Transcrição Brn-3C/genética , Animais , Benzaldeídos/farmacologia , Modelos Animais de Doenças , Haploinsuficiência/genética , Perda Auditiva/genética , Perda Auditiva/patologia , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Ruído/efeitos adversos , Quinolinas/farmacologia , Fator de Transcrição Brn-3C/metabolismo , Tretinoína/farmacologia , para-Aminobenzoatos/farmacologia
3.
Oncol Lett ; 15(3): 3472-3481, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29467869

RESUMO

Epithelial-mesenchymal transition (EMT) allows neoplastic cells to gain the invasive phenotype and become migratory, which is required for cancer progression and metastasis. In the present study, the expression of EMT-associated biomarkers and their association with clinicopathological parameters in laryngeal squamous cell carcinoma (LSCC) was investigated. E-cadherin, N-cadherin, ß-catenin and zinc finger E-box binding homeobox 2 (ZEB2) protein expression was evaluated with immunohistochemistry in a cohort of 76 patients with operable LSCC. The association between these transition markers, clinicopathological parameters and their prognostic impact in LSCC was analyzed. Immunohistochemical analysis revealed that EMT-associated proteins were differentially expressed between LSCC and adjacent non-neoplastic laryngeal tissue. Negative E-cadherin expression and positive N-cadherin, ß-catenin and ZEB2 expression were associated with a later tumor (T) stage, decreasing tumor differentiation and a reduced overall survival (OS) time (OS: E-cadherin, P=0.016; N-cadherin, P=0.003; ß-catenin, P=0.002; ZEB2, P=0.0003). E-cadherin/ß-catenin co-expression was significantly associated with the majority of clinicopathological parameters assessed, including lymph node metastases, T stage and tumor cell differentiation (P=0.004, P=0.005, and P<0.001, respectively). Multivariate analysis indicated that T stage and the positive expression of ß-catenin and ZEB2 were independent risk factors for OS in LSCC (P=0.014, P=0.025 and P=0.003, respectively). It was concluded that EMT mediates tumor progression, and reduces OS time in patients with LSCC. E-cadherin/ß-catenin co-expression may be associated with clinicopathological parameters. T stage, and the positive co-expression of ß-catenin and ZEB2 may be independent predictors of prognosis in LSCC.

4.
Mol Med Rep ; 16(6): 9011-9016, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28990112

RESUMO

Enlarged vestibular aqueduct (EVA)­associated hearing loss is frequently detected in individuals carrying the SLC26A4 mutation in the Chinese population. The present study aimed to identify the causative SLC26A4 coding mutations in a patient group with nonsyndromic hearing loss (NSHL) and EVA. Genomic DNA was extracted from blood samples obtained from 52 NSHL patients with EVA and from 60 normal controls. The mutation analysis for 20 coding exons of SLC26A4 was performed by direct sequencing. The results of the mutational analysis showed that there were two probands from two separate families suffering from bilateral sensorineural hearing loss with EVA, carrying the same novel compound heterozygous mutation of SLC26A4 (c.1644_1645insA and c.2168A>G). Other members of the two families had heterozygous mono­allelic mutations with normal hearing. However, neither of these mutations were detected in the 60 normal controls. These results are the first, to the best of our knowledge, to link the compound heterozygote mutation, c.1644_1645insA and c.2168A>G, in the SLC26A4 gene to NSHL patients with EVA. The two mutations identified in the present study were located in the anti­sigma factor antagonist domain, the core region for plasma membrane targeting of anion transporters, which suggested that the reduced or complete loss of SLC26A4 function was the direct cause of hearing loss in the two patients. These results provide a foundation for further elucidating the genetic factors responsible for EVA­associated NSHL.


Assuntos
Surdez/genética , Predisposição Genética para Doença , Proteínas de Membrana Transportadoras/genética , Mutação/genética , Aqueduto Vestibular/patologia , Audiometria de Tons Puros , Sequência de Bases , Estudos de Casos e Controles , Criança , Análise Mutacional de DNA , Surdez/diagnóstico por imagem , Éxons/genética , Família , Feminino , Heterozigoto , Humanos , Masculino , Linhagem , Transportadores de Sulfato , Aqueduto Vestibular/diagnóstico por imagem
5.
Mol Med Rep ; 9(4): 1185-90, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24535414

RESUMO

An eight­base pair (bp) deletion in the Pou4f3 gene in hair cells is associated with DFNA15, a hereditary form of hearing loss. To explore the pathological mechanisms underlying the development of DFNA15, the effect of the mutation in Pou4f3 on the activity of the myosin VI (Myo6) promoter, was investigated. The upstream regulatory sequence of Myo6 (2625 bp), consisting of an 1899 bp upstream sequence and a 727 bp intron 1 sequence, was amplified using polymerase chain reaction and subcloned into the pGL3­Basic vector expressing firefly luciferase. For verification of inserted fragments, plasmids were subjected to restriction analysis and then sequenced. HEK293T human embryonic kidney cells were transiently transfected with renilla luciferase­thymidine kinase vectors expressing Renilla luciferase and the Myo6 promoter­driven firefly luciferase expressing vectors along with pIRES2­enhanced green fluorescent protein (EGFP)­Pou4f3 (expressing wild­type Pou4f3) or pIRES2­EGFP­Pou4f3 (expressing the truncation mutant of Pou4f3). The relative luciferase activities were measured to determine the activity of the Myo6 promoter. The Myo6 promoter activity was not affected by co­expression of wild­type Pou4f3, as indicated by the comparable relative luciferase activities in the presence of the pIRES2­EGFP­Pou4f3 and the empty control vectors. However, co­expression of mutated Pou4f3 significantly inhibited the activity of the Myo6 promoter to almost half of that of the control (P<0.001). The data suggests that mutated Pou4f3 has a negative role in the promoter activity of Myo6, and by extension, the expression of myosin VI, and this may be an underlying mechanism of DFNA15 hearing loss.


Assuntos
Regulação da Expressão Gênica , Células Ciliadas Auditivas/metabolismo , Mutação/genética , Cadeias Pesadas de Miosina/genética , Fator de Transcrição Brn-3C/genética , Fator de Transcrição Brn-3C/metabolismo , Animais , Sequência de Bases , Genes Reporter , Vetores Genéticos/metabolismo , Humanos , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes/metabolismo
6.
Int J Mol Med ; 33(4): 905-12, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24481113

RESUMO

Myosin light chain kinase (MLCK) phosphorylates myosin regulatory light chains to facilitate its interaction with actin filaments and produce contractile activity. The outer hair cells (OHCs) in the ear contain large amounts of actin and a variety myosins. The stereociliary and somatic motility of OHCs are closely related to hearing. It appears likely that MLCK may play an important role in acoustic trans-duction. In this study, we analyzed, both in vivo and in vitro, the OHCs of mice bearing a specific deletion of the MLCK gene and the OHCs of control mice. The phenotype was assessed by auditory function [acoustic brainstem responses (ABRs) and distortion product otoacoustic emissions (DPOAEs)], inner ear morphology and histology. MLCK-deficient mice aged 6-7 months showed impaired hearing, a 5- to 10-dB sound pressure level (SPL) increase in the ABR thresholds, when responding to clicks and tones of different frequencies (8 and 16 kHz) (P<0.05). The DPOAE amplitudes of 3-month-old MLCK-deficient mice decreased significantly (>10 dB SPL) at low frequencies (4, 5 and 6 kHz). The OHCs in the MLCK-deficient mice increased with abnormal stereocilia. The staining of F-actin and the phosphorylation of the regulatory light chain in MLCK-deficient OHCs was weak. Our results indicate that MLCK may regulate the structure and the motility of stereocilia through F-actin polymerization.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Células Ciliadas Auditivas Externas/enzimologia , Células Ciliadas Auditivas Externas/fisiologia , Audição/fisiologia , Quinase de Cadeia Leve de Miosina/metabolismo , Animais , Limiar Auditivo/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Imunofluorescência , Células Ciliadas Auditivas Externas/citologia , Células Ciliadas Auditivas Externas/ultraestrutura , Camundongos , Camundongos Knockout , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/deficiência , Emissões Otoacústicas Espontâneas/fisiologia , Fosforilação
8.
Artigo em Chinês | MEDLINE | ID: mdl-23656815

RESUMO

OBJECTIVE: To investigate the function of myosin light chain kinase (MLCK) in hearing in mouse by generating inner hair cell-specific Mlck knockout mice and analyze the effect on their hearing. METHODS: Cross Mlck floxed mice with IHC-Cre mice, the genotype and knockout efficiency were confirmed by PCR. We used auditory brain stem response (ABR) to evaluate mice hearing function at different frequencies. RESULTS: Mlck knockout mice were selected by mice tail DNA genotyping and confirmed the deletion of the target gene by isolated inner hair cell DNA genotyping. Mlck-deficient mice showed impaired hearing with a rise in ABR threshold response to click and three different pure tones (8 kHz, 16 kHz, 32 kHz), and the rise was over 20 dB at high-frequency(32 kHz). Further analyses of waveforms showed that wave-I amplitudes on 60 dB SPL, 50 dB SPL and 40 dBSPL in response to tone (16 kHz) were less than control group(P < 0.05) on average, but the ratio of wave I/II and I/III were not difference (P > 0.05). CONCLUSIONS: Mlck is successfully deleted in inner hair cell-specific Mlck knockout mice. Mlck knockout mice display a significantly higher threshold in response to click and tones, especially in high-frequencies.


Assuntos
Potenciais Evocados Auditivos do Tronco Encefálico , Células Ciliadas Auditivas Internas/metabolismo , Perda Auditiva/genética , Quinase de Cadeia Leve de Miosina/genética , Animais , Audiometria de Tons Puros , Limiar Auditivo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
PLoS One ; 7(4): e34894, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22485190

RESUMO

The structural homeostasis of the cochlear hair cell membrane is critical for all aspects of sensory transduction, but the regulation of its maintenance is not well understood. In this report, we analyzed the cochlear hair cells of mice with specific deletion of myosin light chain kinase (MLCK) in inner hair cells. MLCK-deficient mice showed impaired hearing, with a 5- to 14-dB rise in the auditory brainstem response (ABR) thresholds to clicks and tones of different frequencies and a significant decrease in the amplitude of the ABR waves. The mutant inner hair cells produced several ball-like structures around the hair bundles in vivo, indicating impaired membrane stability. Inner hair cells isolated from the knockout mice consistently displayed less resistance to hypoosmotic solution and less membrane F-actin. Myosin light-chain phosphorylation was also reduced in the mutated inner hair cells. Our results suggest that MLCK is necessary for maintaining the membrane stability of inner hair cells.


Assuntos
Membrana Celular/enzimologia , Células Ciliadas Auditivas Internas/enzimologia , Homeostase , Quinase de Cadeia Leve de Miosina/fisiologia , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Membrana Celular/metabolismo , Epitélio/enzimologia , Epitélio/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Expressão Gênica , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/ultraestrutura , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cadeias Leves de Miosina/metabolismo , Miosina VIIa , Quinase de Cadeia Leve de Miosina/deficiência , Quinase de Cadeia Leve de Miosina/genética , Miosinas/metabolismo , Órgão Espiral/citologia , Pressão Osmótica , Fosforilação , Processamento de Proteína Pós-Traducional , Deleção de Sequência , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...