Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
RNA ; 27(9): 1046-1067, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34162742

RESUMO

RNA exosomopathies, a growing family of diseases, are linked to missense mutations in genes encoding structural subunits of the evolutionarily conserved, 10-subunit exoribonuclease complex, the RNA exosome. This complex consists of a three-subunit cap, a six-subunit, barrel-shaped core, and a catalytic base subunit. While a number of mutations in RNA exosome genes cause pontocerebellar hypoplasia, mutations in the cap subunit gene EXOSC2 cause an apparently distinct clinical presentation that has been defined as a novel syndrome SHRF (short stature, hearing loss, retinitis pigmentosa, and distinctive facies). We generated the first in vivo model of the SHRF pathogenic amino acid substitutions using budding yeast by modeling pathogenic EXOSC2 missense mutations (p.Gly30Val and p.Gly198Asp) in the orthologous S. cerevisiae gene RRP4 The resulting rrp4 mutant cells show defects in cell growth and RNA exosome function. Consistent with altered RNA exosome function, we detect significant transcriptomic changes in both coding and noncoding RNAs in rrp4-G226D cells that model EXOSC2 p.Gly198Asp, suggesting defects in nuclear surveillance. Biochemical and genetic analyses suggest that the Rrp4 G226D variant subunit shows impaired interactions with key RNA exosome cofactors that modulate the function of the complex. These results provide the first in vivo evidence that pathogenic missense mutations present in EXOSC2 impair the function of the RNA exosome. This study also sets the stage to compare exosomopathy models to understand how defects in RNA exosome function underlie distinct pathologies.


Assuntos
Exorribonucleases/genética , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Mutação de Sentido Incorreto , RNA Fúngico/genética , Proteínas de Ligação a RNA/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Nanismo/enzimologia , Nanismo/genética , Nanismo/patologia , Exorribonucleases/química , Exorribonucleases/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/química , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Fácies , Expressão Gênica , Glicina/química , Glicina/metabolismo , Perda Auditiva/enzimologia , Perda Auditiva/genética , Perda Auditiva/patologia , Humanos , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , RNA Fúngico/química , RNA Fúngico/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Retinose Pigmentar/enzimologia , Retinose Pigmentar/genética , Retinose Pigmentar/patologia , Saccharomyces cerevisiae/enzimologia , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Síndrome
2.
Hum Mol Genet ; 28(24): 4103-4112, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31628468

RESUMO

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


Assuntos
Surdez/enzimologia , Proteínas Ativadoras de GTPase/deficiência , Proteínas Ativadoras de GTPase/metabolismo , Perda Auditiva/enzimologia , Estereocílios/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Cóclea/enzimologia , Cóclea/metabolismo , Citoesqueleto/metabolismo , Surdez/genética , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/genética , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/fisiologia , Perda Auditiva/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Estereocílios/enzimologia
3.
Hum Mol Genet ; 23(6): 1591-601, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24191262

RESUMO

Methionine sulfoxide reductase B3 (MsrB3) is a protein repair enzyme that specifically reduces methionine-R-sulfoxide to methionine. A recent genetic study showed that the MSRB3 gene is associated with autosomal recessive hearing loss in human deafness DFNB74. However, the precise role of MSRB3 in the auditory system and the pathogenesis of hearing loss have not yet been determined. This work is the first to generate MsrB3 knockout mice to elucidate the possible pathological mechanisms of hearing loss observed in DFNB74 patients. We found that homozygous MsrB3(-/-) mice were profoundly deaf and had largely unaffected vestibular function, whereas heterozygous MsrB3(+/-) mice exhibited normal hearing similar to that of wild-type mice. The MsrB3 protein is expressed in the sensory epithelia of the cochlear and vestibular tissues, beginning at E15.5 and E13.5, respectively. Interestingly, MsrB3 is densely localized at the base of stereocilia on the apical surface of auditory hair cells. MsrB3 deficiency led to progressive degeneration of stereociliary bundles starting at P8, followed by a loss of hair cells, resulting in profound deafness in MsrB3(-/-) mice. The hair cell loss appeared to be mediated by apoptotic cell death, which was measured using TUNEL and caspase 3 immunocytochemistry. Taken together, our data suggest that MsrB3 plays an essential role in maintaining the integrity of hair cells, possibly explaining the pathogenesis of DFNB74 deafness in humans caused by MSRB3 deficiency.


Assuntos
Cóclea/patologia , Perda Auditiva/genética , Perda Auditiva/patologia , Metionina Sulfóxido Redutases/genética , Estereocílios/patologia , Animais , Apoptose , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/patologia , Perda Auditiva/enzimologia , Humanos , Metionina Sulfóxido Redutases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Estereocílios/metabolismo
4.
Hum Mol Genet ; 23(12): 3289-98, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24482543

RESUMO

Cyclic AMP (cAMP) production, which is important for mechanotransduction within the inner ear, is catalyzed by adenylate cyclases (AC). However, knowledge of the role of ACs in hearing is limited. Previously, a novel autosomal recessive non-syndromic hearing impairment locus DFNB44 was mapped to chromosome 7p14.1-q11.22 in a consanguineous family from Pakistan. Through whole-exome sequencing of DNA samples from hearing-impaired family members, a nonsense mutation c.3112C>T (p.Arg1038*) within adenylate cyclase 1 (ADCY1) was identified. This stop-gained mutation segregated with hearing impairment within the family and was not identified in ethnically matched controls or within variant databases. This mutation is predicted to cause the loss of 82 amino acids from the carboxyl tail, including highly conserved residues within the catalytic domain, plus a calmodulin-stimulation defect, both of which are expected to decrease enzymatic efficiency. Individuals who are homozygous for this mutation had symmetric, mild-to-moderate mixed hearing impairment. Zebrafish adcy1b morphants had no FM1-43 dye uptake and lacked startle response, indicating hair cell dysfunction and gross hearing impairment. In the mouse, Adcy1 expression was observed throughout inner ear development and maturation. ADCY1 was localized to the cytoplasm of supporting cells and hair cells of the cochlea and vestibule and also to cochlear hair cell nuclei and stereocilia. Ex vivo studies in COS-7 cells suggest that the carboxyl tail of ADCY1 is essential for localization to actin-based microvilli. These results demonstrate that ADCY1 has an evolutionarily conserved role in hearing and that cAMP signaling is important to hair cell function within the inner ear.


Assuntos
Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Perda Auditiva/patologia , Adenilil Ciclases/química , Animais , Células COS , Chlorocebus aethiops , Códon sem Sentido , Citoplasma/metabolismo , Orelha Interna/crescimento & desenvolvimento , Feminino , Perda Auditiva/enzimologia , Humanos , Células Labirínticas de Suporte/metabolismo , Masculino , Camundongos , Peixe-Zebra/genética
5.
Am J Hum Genet ; 93(1): 132-40, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23768514

RESUMO

Previously, DFNB89, a locus associated with autosomal-recessive nonsyndromic hearing impairment (ARNSHI), was mapped to chromosomal region 16q21-q23.2 in three unrelated, consanguineous Pakistani families. Through whole-exome sequencing of a hearing-impaired individual from each family, missense mutations were identified at highly conserved residues of lysyl-tRNA synthetase (KARS): the c.1129G>A (p.Asp377Asn) variant was found in one family, and the c.517T>C (p.Tyr173His) variant was found in the other two families. Both variants were predicted to be damaging by multiple bioinformatics tools. The two variants both segregated with the nonsyndromic-hearing-impairment phenotype within the three families, and neither mutation was identified in ethnically matched controls or within variant databases. Individuals homozygous for KARS mutations had symmetric, severe hearing impairment across all frequencies but did not show evidence of auditory or limb neuropathy. It has been demonstrated that KARS is expressed in hair cells of zebrafish, chickens, and mice. Moreover, KARS has strong localization to the spiral ligament region of the cochlea, as well as to Deiters' cells, the sulcus epithelium, the basilar membrane, and the surface of the spiral limbus. It is hypothesized that KARS variants affect aminoacylation in inner-ear cells by interfering with binding activity to tRNA or p38 and with tetramer formation. The identification of rare KARS variants in ARNSHI-affected families defines a gene that is associated with ARNSHI.


Assuntos
Perda Auditiva/enzimologia , Perda Auditiva/genética , Lisina-tRNA Ligase/genética , Mutação de Sentido Incorreto , Animais , Estudos de Casos e Controles , Galinhas , Cóclea/metabolismo , Cóclea/patologia , Biologia Computacional/métodos , Consanguinidade , Feminino , Ligação Genética , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Haplótipos , Homozigoto , Humanos , Lisina-tRNA Ligase/metabolismo , Masculino , Camundongos , Linhagem , Aminoacilação de RNA de Transferência , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
6.
J Med Genet ; 51(4): 275-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24556084

RESUMO

BACKGROUND: Congenital muscular dystrophies (CMD) with hypoglycosylation of α-dystroglycan are clinically and genetically heterogeneous disorders that are often associated with brain malformations and eye defects. Presently, 16 proteins are known whose dysfunction impedes glycosylation of α-dystroglycan and leads to secondary dystroglycanopathy. OBJECTIVE: To identify the cause of CMD with secondary merosin deficiency, hypomyelination and intellectual disability in two siblings from a consanguineous family. METHODS: Autozygosity mapping followed by whole exome sequencing and immunochemistry were used to discover and verify a new genetic defect in two siblings with CMD. RESULTS: We identified a homozygous missense mutation (c.325C>T, p.Q109*) in protein O-mannosyl kinase (POMK) that encodes a glycosylation-specific kinase (SGK196) required for function of the dystroglycan complex. The protein was absent from skeletal muscle and skin fibroblasts of the patients. In patient muscle, ß-dystroglycan was normally expressed at the sarcolemma, while α-dystroglycan failed to do so. Further, we detected co-localisation of POMK with desmin at the costameres in healthy muscle, and a substantial loss of desmin from the patient muscle. CONCLUSIONS: Homozygous truncating mutations in POMK lead to CMD with secondary merosin deficiency, hypomyelination and intellectual disability. Loss of desmin suggests that failure of proper α-dystroglycan glycosylation impedes the binding to extracellular matrix proteins and also affects the cytoskeleton.


Assuntos
Perda Auditiva/complicações , Deficiência Intelectual/complicações , Laminina/deficiência , Distrofias Musculares/congênito , Mutação/genética , Bainha de Mielina/patologia , Proteínas Quinases/genética , Adolescente , Sequência de Bases , Criança , Pré-Escolar , Análise Mutacional de DNA , Família , Feminino , Perda Auditiva/enzimologia , Perda Auditiva/genética , Humanos , Imuno-Histoquímica , Lactente , Recém-Nascido , Deficiência Intelectual/enzimologia , Deficiência Intelectual/genética , Masculino , Dados de Sequência Molecular , Músculo Esquelético/patologia , Distrofias Musculares/complicações , Distrofias Musculares/enzimologia , Distrofias Musculares/genética , Linhagem , Adulto Jovem
7.
Am J Hum Genet ; 87(2): 282-8, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20673864

RESUMO

Perrault syndrome is a recessive disorder characterized by ovarian dysgenesis in females, sensorineural deafness in both males and females, and in some patients, neurological manifestations. No genes for Perrault syndrome have heretofore been identified. A small family of mixed European ancestry includes two sisters with well-characterized Perrault syndrome. Whole-exome sequencing of genomic DNA from one of these sisters revealed exactly one gene with two rare functional variants: HSD17B4, which encodes 17beta-hydroxysteroid dehydrogenase type 4 (HSD17B4), also known as D-bifunctional protein (DBP). HSD17B4/DBP is a multifunctional peroxisomal enzyme involved in fatty acid beta-oxidation and steroid metabolism. Both sisters are compound heterozygotes for HSD17B4 c.650A>G (p.Y217C) (maternal allele) and HSB17B4 c.1704T>A (p.Y568X) (paternal allele). The missense mutation is predicted by structural analysis to destabilize the HSD17B4 dehydrogenase domain. The nonsense mutation leads to very low levels of HSD17B4 transcript. Expression of mutant HSD17B4 protein in a compound heterozygote was severely reduced. Mutations in HSD17B4 are known to cause DBP deficiency, an autosomal-recessive disorder of peroxisomal fatty acid beta-oxidation that is generally fatal within the first two years of life. No females with DBP deficiency surviving past puberty have been reported, and ovarian dysgenesis has not previously been associated with this illness. Six other families with Perrault syndrome have wild-type sequences of HSD17B4. These results indicate that Perrault syndrome and DBP deficiency overlap clinically; that Perrault syndrome is genetically heterogeneous; that DBP deficiency may be underdiagnosed; and that whole-exome sequencing can reveal critical genes in small, nonconsanguineous families.


Assuntos
17-Hidroxiesteroide Desidrogenases/genética , Ataxia/complicações , Disgenesia Gonadal/complicações , Disgenesia Gonadal/enzimologia , Perda Auditiva/complicações , Hidroliases/genética , Mutação/genética , Ovário/anormalidades , 17-Hidroxiesteroide Desidrogenases/química , Sequência de Aminoácidos , Ataxia/enzimologia , Ataxia/genética , Sequência de Bases , Análise Mutacional de DNA , Éxons/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Disgenesia Gonadal/genética , Perda Auditiva/enzimologia , Perda Auditiva/genética , Heterozigoto , Humanos , Hidroliases/química , Masculino , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteína Multifuncional do Peroxissomo-2 , Estrutura Secundária de Proteína , Síndrome
8.
Cell Mol Life Sci ; 69(14): 2429-34, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22562580

RESUMO

Cisplatin is a widely used chemotherapeutic agent that causes significant hearing loss. Previous studies have shown that cisplatin exposure is associated with increase in reactive oxygen species (ROS) in the cochlea. The inner ear expresses a unique isoform of NADPH oxidase, NOX3. This enzyme may be the primary source of ROS generation in the cochlea. The knockdown of NOX3 by pretreatment with siRNA prevented cisplatin ototoxicity, as demonstrated by preservation of hearing thresholds and inner ear sensory cells. Trans-tympanic NOX3 siRNA reduced the expression of NOX3 and biomarkers of cochlear damage, including transient receptor vanilloid 1 (TRPV1) channel and kidney injury molecule-1 (KIM-1) in cochlear tissues. In addition, siRNA against NOX3 reduced apoptosis as demonstrated by TUNEL staining, and prevented the increased expression of Bax and abrogated the decrease in Bcl2 expression following cisplatin administration. Trans-tympanic administration of siRNA directed against NOX3 may provide a useful method of attenuating cisplatin ototoxicity. In this paper, we review recent publications dealing with the role of NOX3 in ototoxicity and the effects of siRNA against cisplatin-induced hearing loss.


Assuntos
Perda Auditiva/enzimologia , Proteínas de Membrana/antagonistas & inibidores , NADPH Oxidases/antagonistas & inibidores , RNA Interferente Pequeno/metabolismo , Cisplatino/toxicidade , Orelha Interna/enzimologia , Perda Auditiva/induzido quimicamente , Perda Auditiva/patologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Paraquat/toxicidade , Interferência de RNA
9.
Proc Natl Acad Sci U S A ; 107(29): 13051-6, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20616061

RESUMO

A significantly increased risk for dominant sensorineural deafness in patients who have Hirschsprung disease (HSCR) caused by endothelin receptor type B and SOX10 has been reported. Despite the fact that c-RET is the most frequent causal gene of HSCR, it has not been determined whether impairments of c-Ret and c-RET cause congenital deafness in mice and humans. Here, we show that impaired phosphorylation of c-Ret at tyrosine 1062 causes HSCR-linked syndromic congenital deafness in c-Ret knockin (KI) mice. The deafness involves neurodegeneration of spiral ganglion neurons (SGNs) with not only impaired phosphorylation of Akt and NF-kappaB but decreased expression of calbindin D28k in inner ears. The congenital deafness involving neurodegeneration of SGNs in c-Ret KI mice was rescued by introducing constitutively activated RET. Taken together with our results for three patients with congenital deafness with c-RET-mediated severe HSCR, our results indicate that c-Ret and c-RET are a deafness-related molecule in mice and humans.


Assuntos
Perda Auditiva/complicações , Perda Auditiva/enzimologia , Doença de Hirschsprung/complicações , Doença de Hirschsprung/enzimologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Substituição de Aminoácidos/genética , Animais , Surdez/complicações , Surdez/enzimologia , Ativação Enzimática , Técnicas de Introdução de Genes , Perda Auditiva/congênito , Imuno-Histoquímica , Camundongos , Mutação/genética , NF-kappa B/metabolismo , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Neurônios/enzimologia , Neurônios/patologia , Neurônios/ultraestrutura , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Gânglio Espiral da Cóclea/enzimologia , Gânglio Espiral da Cóclea/patologia , Gânglio Espiral da Cóclea/ultraestrutura
10.
Int J Audiol ; 52(1): 23-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23190330

RESUMO

OBJECTIVE: The purpose of this review was to evaluate the current literature on phosphoribosylpyrophosphate synthetase 1 (PRPS1)-related diseases and their consequences on hearing function. DESIGN: A literature search of peer-reviewed, published journal articles was conducted in online bibliographic databases. STUDY SAMPLE: Three databases for medical research were included in this review. RESULTS: Mutations in PRPS1 are associated with a spectrum of non-syndromic to syndromic hearing loss. Hearing loss in male patients with PRPS1 mutations is bilateral, moderate to profound, and can be prelingual or postlingual, progressive or non-progressive. Audiogram shapes associated with PRPS1 deafness are usually residual and flat. Female carriers can have unilateral or bilateral hearing impairment. Gain of function mutations in PRPS1 cause a superactivity of the PRS-I protein whereas the loss-of-function mutations result in X-linked nonsyndromic sensorineural deafness type 2 (DFN2), or in syndromic deafness including Arts syndrome and X-linked Charcot-Marie-Tooth disease-5 (CMTX5). CONCLUSIONS: Lower residual activity in PRS-I leads to a more severe clinical manifestation. Clinical and molecular findings suggest that the four PRPS1 disorders discovered to date belong to the same disease spectrum. Dietary supplementation with S-adenosylmethionine (SAM) appeared to alleviate the symptoms of Arts syndrome patients, suggesting that SAM could compensate for PRS-I deficiency.


Assuntos
Perda Auditiva/genética , Audição/genética , Mutação , Ribose-Fosfato Pirofosfoquinase/genética , Suplementos Nutricionais , Feminino , Predisposição Genética para Doença , Perda Auditiva/diagnóstico , Perda Auditiva/tratamento farmacológico , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Hereditariedade , Humanos , Masculino , Fenótipo , S-Adenosilmetionina/uso terapêutico , Índice de Gravidade de Doença , Fatores Sexuais
11.
Clin Genet ; 82(1): 56-63, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21534946

RESUMO

Mutations in the TMPRSS3 gene are known to cause autosomal recessive non-syndromic hearing impairment (ARNSHI). After undergoing a genome scan, 10 consanguineous Pakistani families with ARNSHI were found to have significant or suggestive evidence of linkage to the TMPRSS3 region. In order to elucidate if the TMPRSS3 gene is responsible for ARNSHI in these families, the gene was sequenced using DNA samples from these families. Six TMPRSS3 variants were found to cosegregate in 10 families. None of these variants were detected in 500 control chromosomes. Four novel variants, three of which are missense [c.310G>A (p.Glu104Lys), c.767C>T (p.Ala256Val) and c.1273T>C (p.Cys425Arg)] and one nonsense [c.310G>T (p.Glu104Stop)], were identified. The pathogenicity of novel missense variants was investigated through bioinformatics analyses. Additionally, the previously reported deletion c.208delC (p.His70ThrfsX19) was identified in one family and the known mutation c.1219T>C (p.Cys407Arg) was found in five families, which makes c.1219T>C (p.Cys407Arg) as the most common TMPRSS3 mutation within the Pakistani population. Identification of these novel variants lends support to the importance of elements within the low-density lipoprotein receptor A (LDLRA) and serine protease domains in structural stability, ligand binding and proteolytic activity for proper TMPRSS3 function within the inner ear.


Assuntos
Orelha Interna/patologia , Perda Auditiva/genética , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Estudos de Casos e Controles , Cromossomos Humanos Par 21 , Consanguinidade , Orelha Interna/enzimologia , Éxons , Feminino , Genes Recessivos , Ligação Genética , Loci Gênicos , Perda Auditiva/enzimologia , Perda Auditiva/patologia , Humanos , Masculino , Modelos Moleculares , Mutação , Linhagem , Fenótipo , Estrutura Terciária de Proteína
12.
Biochim Biophys Acta ; 1802(2): 259-68, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19857571

RESUMO

The neuraminidase-1 (Neu1) knockout mouse model is a phenocopy of the lysosomal storage disease (LSD) sialidosis, characterized by multisystemic and neuropathic symptoms, including hearing loss. We have characterized the auditory defects in Neu1(-/-) mice and found that hearing loss involves both conductive and sensorineural components. Auditory brainstem response (ABR) thresholds were significantly elevated in Neu1(-/-) mice at P21 (48-55 dB), and hearing loss appeared progressive (53-66 dB at P60). At these ages Neu1(-/-) mice accumulated cerumen in the external ear canal and had a thickened mucosa and inflammation in the middle ear. In cochleae of adult wild-type mice, Neu1 was expressed in several cell types in the stria vascularis, the organ of Corti, and spiral ganglion. Progressive morphological abnormalities such as extensive vacuolization were detected in the Neu1(-/-) cochleae as early as P9. These early morphologic changes in Neu1(-/-) cochleae were associated with oversialylation of several lysosomal associated membrane proteins (Lamps) in the stria vascularis. A marked increase in the expression and apical localization of Lamp-1 in marginal cells of the stria vascularis predicts exacerbation of lysosomal exocytosis into the endolymph. Consequently, the endolymphatic potential in Neu1(-/-) mice was reduced by approximately 20 mV at ages P31-P44, which would cause dysfunction of transduction in sensory hair cells. This study suggests a molecular mechanism that contributes to hearing loss in sialidosis and identifies potential therapeutic targets.


Assuntos
Cóclea/fisiopatologia , Perda Auditiva/genética , Neuraminidase/deficiência , Neuraminidase/genética , Animais , Cóclea/patologia , Modelos Animais de Doenças , Orelha Média/patologia , Perda Auditiva/enzimologia , Doenças por Armazenamento dos Lisossomos/enzimologia , Doenças por Armazenamento dos Lisossomos/genética , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Vacúolos/patologia , Vacúolos/ultraestrutura
13.
Nat Cell Biol ; 5(5): 422-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12717441

RESUMO

Maintenance of the post-mitotic state in the post-natal mammalian brain is an active process that requires the cyclin-dependent kinase inhibitors (CKIs) p19Ink4d (Ink4d) and p27Kip1 (Kip1). In animals with targeted deletions of both Ink4d and Kip1, terminally differentiated, post-mitotic neurons are observed to re-enter the cell cycle, divide and undergo apoptosis. However, when either Ink4d or Kip1 alone are deleted, the post-mitotic state is maintained, suggesting a redundant role for these genes in mature neurons. In the organ of Corti--the auditory sensory epithelium of mammals--sensory hair cells and supporting cells become post-mitotic during embryogenesis and remain quiescent for the life of the animal. When lost as a result of environmental insult or genetic abnormality, hair cells do not regenerate, and this loss is a common cause of deafness in humans. Here, we report that targeted deletion of Ink4d alone is sufficient to disrupt the maintenance of the post-mitotic state of sensory hair cells in post-natal mice. In Ink4d-/- animals, hair cells are observed to aberrantly re-enter the cell cycle and subsequently undergo apoptosis, resulting in progressive hearing loss. Our results identify a novel mechanism underlying a non-syndromic form of progressive hearing loss in mice.


Assuntos
Apoptose/genética , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Células Ciliadas Auditivas/enzimologia , Perda Auditiva/enzimologia , Perda Auditiva/genética , Regeneração Nervosa/genética , Proteínas Supressoras de Tumor/deficiência , Animais , Caspase 3 , Caspases/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Divisão Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p19 , Inibidor de Quinase Dependente de Ciclina p27 , Dineínas , Feto , Imunofluorescência , Células Ciliadas Auditivas/ultraestrutura , Homeostase/genética , Camundongos , Camundongos Knockout , Miosina VIIa , Miosinas/metabolismo , Proteínas Supressoras de Tumor/genética
14.
J Med Genet ; 47(9): 643-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20472657

RESUMO

BACKGROUND: Moderate to severe prelingual hearing impairment (DFNB84) was observed in an extended consanguineous Palestinian kindred. All affected relatives shared a 12.5 MB homozygous haplotype on chromosome 12q21 with lod score 4.30. This homozygous region harbours the protein tyrosine phosphatase receptor Q gene PTPRQ, which is known to be essential to hearing in mouse. METHODS: Candidate genes in the 12.5 MB homozygous region were characterized genomically and sequenced in deaf and hearing relatives in the family. RESULTS: Sequence of PTPRQ in affected individuals in the extended kindred revealed c.1285C-->T, leading to p.Gln429Stop. This nonsense mutation co-segregated with hearing loss in the family and was homozygous in all affected relatives. The mutation did not appear among 288 Palestinian controls (576 chromosomes), all adults with normal hearing. No homozygous mutations in PTPRQ appeared in any of 218 other probands with hearing loss. CONCLUSION: Identification of the DFNB84 gene represents the first identification of PTPRQ mutation in human hearing loss.


Assuntos
Cílios/genética , Códon sem Sentido/genética , Loci Gênicos/genética , Perda Auditiva/enzimologia , Perda Auditiva/genética , Proteínas de Membrana/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Análise Mutacional de DNA , Família , Feminino , Humanos , Padrões de Herança/genética , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem
15.
Toxicol Lett ; 349: 115-123, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34089817

RESUMO

Cisplatin, the most widely used platinum-based anticancer drug, often causes progressive and irreversible sensorineural hearing loss in cancer patients. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. Nicotinamide adenine dinucleotide (NAD+), a co-substrate for the sirtuin family and PARPs, has emerged as a potent therapeutic molecular target in various diseases. In our investigates, we observed that NAD+ level was changed in the cochlear explants of mice treated with cisplatin. Supplementation of a specific inhibitor (TES-1025) of α-amino-ß-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), a rate-limiting enzyme of NAD+de novo synthesis pathway, promoted SIRT1 activity, increased mtDNA contents and enhanced AMPK expression, thus significantly reducing hair cells loss and deformation. The protection was blocked by EX527, a specific SIRT1 inhibitor. Meanwhile, the use of NMN, a precursor of NAD+ salvage synthesis pathway, had shown beneficial effect on hair cell under cisplatin administration, effectively suppressing PARP1. In vivo experiments confirmed the hair cell protection of NAD+ modulators in cisplatin treated mice and zebrafish. In conclusion, we demonstrated that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway could both prevent ototoxicity of cisplatin. These results suggested that direct modulation of cellular NAD+ levels could be a promising therapeutic approach for protection of hearing from cisplatin-induced ototoxicity.


Assuntos
Inibidores Enzimáticos/farmacologia , Células Ciliadas Auditivas/efeitos dos fármacos , Perda Auditiva/prevenção & controle , Audição/efeitos dos fármacos , NAD/biossíntese , Ototoxicidade/prevenção & controle , Sirtuína 1/metabolismo , Animais , Animais Geneticamente Modificados , Carboxiliases/antagonistas & inibidores , Carboxiliases/metabolismo , Cisplatino , Modelos Animais de Doenças , Ativação Enzimática , Células Ciliadas Auditivas/enzimologia , Células Ciliadas Auditivas/patologia , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Sistema da Linha Lateral/efeitos dos fármacos , Sistema da Linha Lateral/enzimologia , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Ototoxicidade/enzimologia , Ototoxicidade/etiologia , Ototoxicidade/fisiopatologia , Peixe-Zebra
16.
Hear Res ; 388: 107896, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31982642

RESUMO

The development of knockin mice with Cre recombinase expressed under the control of the promoter for choline acetyltransferase (ChAT) has allowed experimental manipulation of cholinergic circuits. However, currently available ChATCre mouse lines are on the C57BL/6J strain background, which shows early onset age-related hearing loss attributed to the Cdh23753A mutation (a.k.a., the ahl mutation). To develop ChATCre mice without accelerated hearing loss, we backcrossed ChATIRES-Cre mice with CBA/CaJ mice that have normal hearing. We used genotyping to obtain mice homozygous for ChATIRES-Cre and the wild-type allele at the Cdh23 locus (ChATCre,Cdh23WT). In the new line, auditory brainstem response thresholds were ∼20 dB lower than those in 9 month old ChATIRES-Cre mice at all frequencies tested (4-31.5 kHz). These thresholds were stable throughout the period of testing (3-12 months of age). We then bred ChATCre,Cdh23WT animals with Ai14 reporter mice to confirm the expression pattern of ChATCre. In these mice, tdTomato-labeled cells were observed in all brainstem regions known to contain cholinergic cells. We then stained the tissue with a neuron-specific marker, NeuN, to determine whether Cre expression was limited to neurons. Across several brainstem nuclei (pontomesencephalic tegmentum, motor trigeminal and facial nuclei), 100% of the tdTomato-labeled cells were double-labeled with anti-NeuN (n = 1896 cells), indicating Cre-recombinase was limited to neurons. Almost all of these cells (1867/1896 = 98.5%) also stained with antibodies against ChAT, indicating that reporter label was expressed almost exclusively in cholinergic neurons. Finally, an average 88.7% of the ChAT+ cells in these nuclei were labeled with tdTomato, indicating that the Cre is expressed in a large proportion of the cholinergic cells in these nuclei. We conclude that the backcrossed ChATCre,Cdh23WT mouse line has normal hearing and expresses Cre recombinase almost exclusively in cholinergic neurons. This ChATCre,Cdh23WT mouse line may provide an opportunity to manipulate cholinergic circuits without the confound of accelerated hearing loss associated with the C57BL/6J background. Furthermore, comparison with lines that do show early hearing loss may provide insight into possible cholinergic roles in age-related hearing loss.


Assuntos
Tronco Encefálico/enzimologia , Colina O-Acetiltransferase/metabolismo , Fibras Colinérgicas/enzimologia , Perda Auditiva/prevenção & controle , Audição , Integrases/metabolismo , Animais , Limiar Auditivo , Tronco Encefálico/fisiopatologia , Caderinas/genética , Colina O-Acetiltransferase/genética , Cruzamentos Genéticos , Proteínas de Ligação a DNA/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Técnicas de Introdução de Genes , Perda Auditiva/enzimologia , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Integrases/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Regiões Promotoras Genéticas , Especificidade da Espécie
17.
Hear Res ; 381: 107768, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31387070

RESUMO

The pupil dilation response is sensitive both to listening effort and the emotional significance of a task. We aimed to assess the influence of evaluative feedback on the pupil dilation response using a speech reception threshold (SRT) task. Besides the pupil dilation response, we acquired subjective ratings and two physiological biomarkers sensitive to stress: cortisol and alpha-amylase levels as determined in saliva samples. We included 34 participants with normal hearing (mean age = 52 years, age range 25-67 years) and 29 age-matched participants with mild-to-moderate hearing loss (mean age = 52 years, age range 23-64 years). Half of the participants performed a standard SRT test without feedback, and the other half performed an SRT test in which they did receive feedback and were urged to perform better. The SRT conditions targeted 50% and 71% correct reception of the sentences. Pupil size was recorded and saliva samples were obtained and participants rated their experience of the task. Participants with hearing loss performed more poorly on the SRT test than participants with normal hearing participants receiving feedback had better SRTs in the 71% intelligibility condition and higher peak pupil dilation in both intelligibility conditions than participants who did not receive feedback, irrespective of hearing status. Saliva cortisol level and alpha-amylase activity reflected the usual diurnal patterns but showed no effects of hearing status or feedback. Finally, participants who received feedback experienced more difficulties than those who did not receive feedback, irrespective of hearing status. This study underlines the importance of taking into account the influence of task instructions and feedback in a speech perception task as these factors may influence the experienced difficulties, listening effort, and task performance.


Assuntos
Retroalimentação Psicológica , Perda Auditiva/psicologia , Audição , Hidrocortisona/metabolismo , Pessoas com Deficiência Auditiva/psicologia , Pupila , Saliva/enzimologia , Percepção da Fala , alfa-Amilases/metabolismo , Adulto , Idoso , Atenção , Estudos de Casos e Controles , Feminino , Perda Auditiva/diagnóstico , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Inteligibilidade da Fala , Teste do Limiar de Recepção da Fala , Adulto Jovem
18.
Nat Commun ; 10(1): 4150, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515474

RESUMO

Cisplatin is one of the most widely used chemotherapeutic drugs for the treatment of cancer. Unfortunately, one of its major side effects is permanent hearing loss. Here, we show that glutathione transferase α4 (GSTA4), a member of the Phase II detoxifying enzyme superfamily, mediates reduction of cisplatin ototoxicity by removing 4-hydroxynonenal (4-HNE) in the inner ears of female mice. Under cisplatin treatment, loss of Gsta4 results in more profound hearing loss in female mice compared to male mice. Cisplatin stimulates GSTA4 activity in the inner ear of female wild-type, but not male wild-type mice. In female Gsta4-/- mice, cisplatin treatment results in increased levels of 4-HNE in cochlear neurons compared to male Gsta4-/- mice. In CBA/CaJ mice, ovariectomy decreases mRNA expression of Gsta4, and the levels of GSTA4 protein in the inner ears. Thus, our findings suggest that GSTA4-dependent detoxification may play a role in estrogen-mediated neuroprotection.


Assuntos
Cisplatino/efeitos adversos , Glutationa Transferase/metabolismo , Ototoxicidade/enzimologia , Animais , Limiar Auditivo/efeitos dos fármacos , Capilares/patologia , Cóclea/enzimologia , Cóclea/patologia , Cóclea/fisiopatologia , Cruzamentos Genéticos , Dano ao DNA/genética , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa Transferase/deficiência , Perda Auditiva/complicações , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Masculino , Camundongos Endogâmicos CBA , Ototoxicidade/complicações , Ototoxicidade/patologia , Ototoxicidade/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/patologia
19.
Hum Mutat ; 29(1): 130-41, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17918732

RESUMO

Building on our discovery that mutations in the transmembrane serine protease, TMPRSS3, cause nonsyndromic deafness, we have investigated the contribution of other TMPRSS family members to the auditory function. To identify which of the 16 known TMPRSS genes had a strong likelihood of involvement in hearing function, three types of biological evidence were examined: 1) expression in inner ear tissues; 2) location in a genomic interval that contains a yet unidentified gene for deafness; and 3) evaluation of hearing status of any available Tmprss knockout mouse strains. This analysis demonstrated that, besides TMPRSS3, another TMPRSS gene was essential for hearing and, indeed, mice deficient for Hepsin (Hpn) also known as Tmprss1 exhibited profound hearing loss. In addition, TMPRSS2, TMPRSS5, and CORIN, also named TMPRSS10, showed strong likelihood of involvement based on their inner ear expression and mapping position within deafness loci PKSR7, DFNB24, and DFNB25, respectively. These four TMPRSS genes were then screened for mutations in affected members of the DFNB24 and DFNB25 deafness families, and in a cohort of 362 sporadic deaf cases. This large mutation screen revealed numerous novel sequence variations including three potential pathogenic mutations in the TMPRSS5 gene. The mutant forms of TMPRSS5 showed reduced or absent proteolytic activity. Subsequently, TMPRSS genes with evidence of involvement in deafness were further characterized, and their sites of expression were determined. Tmprss1, 3, and 5 proteins were detected in spiral ganglion neurons. Tmprss3 was also present in the organ of Corti. TMPRSS1 and 3 proteins appeared stably anchored to the endoplasmic reticulum membranes, whereas TMPRSS5 was also detected at the plasma membrane. Collectively, these results provide evidence that TMPRSS1 and TMPRSS3 play and TMPRSS5 may play important and specific roles in hearing.


Assuntos
Perda Auditiva/genética , Proteínas de Membrana/genética , Serina Endopeptidases/genética , Animais , Orelha Interna/metabolismo , Perda Auditiva/enzimologia , Humanos , Imuno-Histoquímica , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Serina Endopeptidases/análise , Serina Endopeptidases/metabolismo
20.
Biochim Biophys Acta ; 1773(8): 1341-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17306896

RESUMO

c-Jun N-terminal kinases (JNKs), also referred to as stress-activated kinases (SAPKs), were initially characterized by their activation in response to cell stress such as UV irradiation. JNK/SAPKs have since been characterized to be involved in proliferation, apoptosis, motility, metabolism and DNA repair. Dysregulated JNK signaling is now believed to contribute to many diseases involving neurodegeneration, chronic inflammation, birth defects, cancer and ischemia/reperfusion injury. In this review, we present our current understanding of JNK regulation and their involvement in homeostasis and dysregulation in human disease.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sequência de Aminoácidos , Animais , Doença/etiologia , Feminino , Perda Auditiva/enzimologia , Humanos , Inflamação/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/química , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fígado/irrigação sanguínea , Fígado/lesões , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Dados de Sequência Molecular , Neoplasias/enzimologia , Defeitos do Tubo Neural/enzimologia , Doenças Neurodegenerativas/enzimologia , Gravidez , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA