Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 602(16): 3995-4025, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39037943

RESUMO

The hair bundle of cochlear hair cells comprises specialized microvilli, the stereocilia, which fulfil the role of mechanotransduction. Genetic defects and environmental noise challenge the maintenance of hair bundle structure, critically contributing to age-related hearing loss. Stereocilia fusion is a major component of the hair bundle pathology in mature hair cells, but its role in hearing loss and its molecular basis are poorly understood. Here, we utilized super-resolution expansion microscopy to examine the molecular anatomy of outer hair cell stereocilia fusion in mouse models of age-related hearing loss, heightened endoplasmic reticulum stress and prolonged noise exposure. Prominent stereocilia fusion in our model of heightened endoplasmic reticulum stress, Manf (Mesencephalic astrocyte-derived neurotrophic factor)-inactivated mice in a background with Cadherin 23 missense mutation, impaired mechanotransduction and calcium balance in stereocilia. This was indicated by reduced FM1-43 dye uptake through the mechanotransduction channels, reduced neuroplastin/PMCA2 expression and increased expression of the calcium buffer oncomodulin inside stereocilia. Sparse BAIAP2L2 and myosin 7a expression was retained in the fused stereocilia but mislocalized away from their functional sites at the tips. These hair bundle abnormalities preceded cell soma degeneration, suggesting a sequela from stereociliary molecular perturbations to cell death signalling. In the age-related hearing loss and noise-exposure models, stereocilia fusion was more restricted within the bundles, yet both models exhibited oncomodulin upregulation at the fusion sites, implying perturbed calcium homeostasis. We conclude that stereocilia fusion is linked with the failure to maintain cellular proteostasis and with disturbances in stereociliary calcium balance. KEY POINTS: Stereocilia fusion is a hair cell pathology causing hearing loss. Inactivation of Manf, a component of the endoplasmic reticulum proteostasis machinery, has a cell-intrinsic mode of action in triggering outer hair cell stereocilia fusion and the death of these cells. The genetic background with Cadherin 23 missense mutation contributes to the high susceptibility of outer hair cells to stereocilia fusion, evidenced in Manf-inactivated mice and in the mouse models of early-onset hearing loss and noise exposure. Endoplasmic reticulum stress feeds to outer hair cell stereocilia bundle pathology and impairs the molecular anatomy of calcium regulation. The maintenance of the outer hair cell stereocilia bundle cohesion is challenged by intrinsic and extrinsic stressors, and understanding the underlying mechanisms will probably benefit the development of interventions to promote hearing health.


Assuntos
Caderinas , Células Ciliadas Auditivas Externas , Mecanotransdução Celular , Estereocílios , Animais , Estereocílios/metabolismo , Estereocílios/patologia , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patologia , Camundongos , Caderinas/metabolismo , Caderinas/genética , Estresse do Retículo Endoplasmático , Camundongos Endogâmicos C57BL , Masculino , Cálcio/metabolismo , Miosina VIIa/metabolismo , Feminino , Perda Auditiva/patologia , Perda Auditiva/genética , Perda Auditiva/metabolismo , Mutação de Sentido Incorreto , Proteínas de Ligação ao Cálcio
2.
Hum Genet ; 143(3): 311-329, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38459354

RESUMO

Identification of genes associated with nonsyndromic hearing loss is a crucial endeavor given the substantial number of individuals who remain without a diagnosis after even the most advanced genetic testing. PKHD1L1 was established as necessary for the formation of the cochlear hair-cell stereociliary coat and causes hearing loss in mice and zebrafish when mutated. We sought to determine if biallelic variants in PKHD1L1 also cause hearing loss in humans. Exome sequencing was performed on DNA of four families segregating autosomal recessive nonsyndromic sensorineural hearing loss. Compound heterozygous p.[(Gly129Ser)];p.[(Gly1314Val)] and p.[(Gly605Arg)];p[(Leu2818TyrfsTer5)], homozygous missense p.(His2479Gln) and nonsense p.(Arg3381Ter) variants were identified in PKHD1L1 that were predicted to be damaging using in silico pathogenicity prediction methods. In vitro functional analysis of two missense variants was performed using purified recombinant PKHD1L1 protein fragments. We then evaluated protein thermodynamic stability with and without the missense variants found in one of the families and performed a minigene splicing assay for another variant. In silico molecular modeling using AlphaFold2 and protein sequence alignment analysis were carried out to further explore potential variant effects on structure. In vitro functional assessment indicated that both engineered PKHD1L1 p.(Gly129Ser) and p.(Gly1314Val) mutant constructs significantly reduced the folding and structural stabilities of the expressed protein fragments, providing further evidence to support pathogenicity of these variants. Minigene assay of the c.1813G>A p.(Gly605Arg) variant, located at the boundary of exon 17, revealed exon skipping leading to an in-frame deletion of 48 amino acids. In silico molecular modeling exposed key structural features that might suggest PKHD1L1 protein destabilization. Multiple lines of evidence collectively associate PKHD1L1 with nonsyndromic mild-moderate to severe sensorineural hearing loss. PKHD1L1 testing in individuals with mild-moderate hearing loss may identify further affected families.


Assuntos
Surdez , Mutação de Sentido Incorreto , Linhagem , Receptores de Superfície Celular , Estereocílios , Animais , Feminino , Humanos , Masculino , Surdez/genética , Sequenciamento do Exoma , Genes Recessivos , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Modelos Moleculares , Receptores de Superfície Celular/genética , Estereocílios/metabolismo , Estereocílios/patologia , Estereocílios/genética
3.
J Biol Chem ; 296: 100243, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33372036

RESUMO

Cochlear hair cells each possess an exquisite bundle of actin-based stereocilia that detect sound. Unconventional myosin 15 (MYO15) traffics and delivers critical molecules required for stereocilia development and thus is essential for building the mechanosensory hair bundle. Mutations in the human MYO15A gene interfere with stereocilia trafficking and cause hereditary hearing loss, DFNB3, but the impact of these mutations is not known, as MYO15 itself is poorly characterized. To learn more, we performed a kinetic study of the ATPase motor domain to characterize its mechanochemical cycle. Using the baculovirus-Sf9 system, we purified a recombinant minimal motor domain (S1) by coexpressing the mouse MYO15 ATPase, essential and regulatory light chains that bind its IQ domains, and UNC45 and HSP90A chaperones required for correct folding of the ATPase. MYO15 purified with either UNC45A or UNC45B coexpression had similar ATPase activities (kcat = ∼ 6 s-1 at 20 °C). Using stopped-flow and quenched-flow transient kinetic analyses, we measured the major rate constants describing the ATPase cycle, including ATP, ADP, and actin binding; hydrolysis; and phosphate release. Actin-attached ADP release was the slowest measured transition (∼12 s-1 at 20 °C), although this did not rate-limit the ATPase cycle. The kinetic analysis shows the MYO15 motor domain has a moderate duty ratio (∼0.5) and weak thermodynamic coupling between ADP and actin binding. These findings are consistent with MYO15 being kinetically adapted for processive motility when oligomerized. Our kinetic characterization enables future studies into how deafness-causing mutations affect MYO15 and disrupt stereocilia trafficking necessary for hearing.


Assuntos
Surdez/genética , Chaperonas Moleculares/genética , Miosinas/genética , Estereocílios/genética , Adenosina Trifosfatases/genética , Animais , Surdez/patologia , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Audição/genética , Humanos , Cinética , Camundongos , Mutação/genética , Domínios Proteicos/genética , Estereocílios/patologia
4.
Hum Mol Genet ; 29(12): 2004-2021, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32167554

RESUMO

Perception of sound is initiated by mechanically gated ion channels at the tips of stereocilia. Mature mammalian auditory hair cells require transmembrane channel-like 1 (TMC1) for mechanotransduction, and mutations of the cognate genetic sequences result in dominant or recessive heritable deafness forms in humans and mice. In contrast, zebrafish lateral line hair cells, which detect water motion, require Tmc2a and Tmc2b. Here, we use standard and multiplex genome editing in conjunction with functional and behavioral assays to determine the reliance of zebrafish hearing and vestibular organs on Tmc proteins. Surprisingly, our approach using multiple mutant alleles demonstrates that hearing in zebrafish is not dependent on Tmc1, nor is it fully dependent on Tmc2a and Tmc2b. Hearing however is absent in triple-mutant zebrafish that lack Tmc1, Tmc2a and Tmc2b. These outcomes reveal a striking resemblance of Tmc protein reliance in the vestibular sensory epithelia of mammals to the maculae of zebrafish. Moreover, our findings disclose a logic of Tmc use where hearing depends on a complement of Tmc proteins beyond those employed to sense water motion.


Assuntos
Perda Auditiva Neurossensorial/genética , Audição/genética , Proteínas de Membrana/genética , Proteínas de Peixe-Zebra/genética , Animais , Surdez/genética , Surdez/patologia , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Perda Auditiva Neurossensorial/patologia , Humanos , Mecanotransdução Celular/genética , Camundongos , Mutação/genética , Estereocílios/genética , Estereocílios/patologia , Peixe-Zebra/genética
5.
J Neurosci Res ; 98(9): 1745-1763, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31762086

RESUMO

The aging cochlea is subjected to a number of pathological changes to play a role in the onset of age-related hearing loss (ARHL). Although ARHL has often been thought of as the result of the loss of hair cells, it is in fact a disorder with a complex etiology, arising from the changes to both the organ of Corti and its supporting structures. In this study, we examine two aging pathologies that have not been studied in detail despite their apparent prevalence; the fusion, elongation, and engulfment of cochlear inner hair cell stereocilia, and the changes that occur to the tectorial membrane (TM), a structure overlying the organ of Corti that modulates its physical properties in response to sound. Our work demonstrates that similar pathological changes occur in these two structures in the aging cochleae of both mice and humans, examines the ultrastructural changes that underlie stereocilial fusion, and identifies the lost TM components that lead to changes in membrane structure. We place these changes into the context of the wider pathology of the aging cochlea, and identify how they may be important in particular for understanding the more subtle hearing pathologies that precede auditory threshold loss in ARHL.


Assuntos
Envelhecimento/fisiologia , Cóclea/patologia , Perda Auditiva/etiologia , Estereocílios/patologia , Membrana Tectorial/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Cóclea/ultraestrutura , Feminino , Células Ciliadas Auditivas , Audição , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Pessoa de Meia-Idade , Órgão Espiral , Estereocílios/ultraestrutura , Membrana Tectorial/fisiologia , Membrana Tectorial/ultraestrutura
6.
PLoS Genet ; 13(3): e1006692, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28346477

RESUMO

Behavioural anomalies suggesting an inner ear disorder were observed in a colony of transgenic mice. Affected animals were profoundly deaf. Severe hair bundle defects were identified in all outer and inner hair cells (OHC, IHC) in the cochlea and in hair cells of vestibular macular organs, but hair cells in cristae were essentially unaffected. Evidence suggested the disorder was likely due to gene disruption by a randomly inserted transgene construct. Whole-genome sequencing identified interruption of the SorCS2 (Sortilin-related VPS-10 domain containing protein) locus. Real-time-qPCR demonstrated disrupted expression of SorCS2 RNA in cochlear tissue from affected mice and this was confirmed by SorCS2 immuno-labelling. In all affected hair cells, stereocilia were shorter than normal, but abnormalities of bundle morphology and organisation differed between hair cell types. Bundles on OHC were grossly misshapen with significantly fewer stereocilia than normal. However, stereocilia were organised in rows of increasing height. Bundles on IHC contained significantly more stereocilia than normal with some longer stereocilia towards the centre, or with minimal height differentials. In early postnatal mice, kinocilia (primary cilia) of IHC and of OHC were initially located towards the lateral edge of the hair cell surface but often became surrounded by stereocilia as bundle shape and apical surface contour changed. In macular organs the kinocilium was positioned in the centre of the cell surface throughout maturation. There was disruption of the signalling pathway controlling intrinsic hair cell apical asymmetry. LGN and Gαi3 were largely absent, and atypical Protein Kinase C (aPKC) lost its asymmetric distribution. The results suggest that SorCS2 plays a role upstream of the intrinsic polarity pathway and that there are differences between hair cell types in the deployment of the machinery that generates a precisely organised hair bundle.


Assuntos
Regulação da Expressão Gênica , Células Ciliadas Auditivas Internas/metabolismo , Proteínas do Tecido Nervoso/genética , Receptores de Superfície Celular/genética , Estereocílios/genética , Fatores Etários , Animais , Células Ciliadas Auditivas Internas/patologia , Perda Auditiva/genética , Perda Auditiva/metabolismo , Perda Auditiva/fisiopatologia , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Proteínas do Tecido Nervoso/metabolismo , Órgão Espiral/metabolismo , Órgão Espiral/fisiopatologia , Órgão Espiral/ultraestrutura , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estereocílios/metabolismo , Estereocílios/patologia
7.
Hum Mol Genet ; 26(3): 624-636, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031293

RESUMO

Usher syndrome (USH) is the most common cause of inherited deaf-blindness, manifested as USH1, USH2 and USH3 clinical types. The protein products of USH2 causative and modifier genes, USH2A, ADGRV1, WHRN and PDZD7, interact to assemble a multiprotein complex at the ankle link region of the mechanosensitive stereociliary bundle in hair cells. Defects in this complex cause stereociliary bundle disorganization and hearing loss. The four USH2 proteins also interact in vitro with USH1 proteins including myosin VIIa, USH1G (SANS), CIB2 and harmonin. However, it is unclear whether the interactions between USH1 and USH2 proteins occur in vivo and whether USH1 proteins play a role in USH2 complex assembly in hair cells. In this study, we identified a novel interaction between myosin VIIa and PDZD7 by FLAG pull-down assay. We further investigated the role of the above-mentioned four USH1 proteins in the cochlear USH2 complex assembly using USH1 mutant mice. We showed that only myosin VIIa is indispensable for USH2 complex assembly at ankle links, indicating the potential transport and/or anchoring role of myosin VIIa for USH2 proteins in hair cells. However, myosin VIIa is not required for USH2 complex assembly in photoreceptors. We further showed that, while PDZ protein harmonin is not involved, its paralogous USH2 proteins, PDZD7 and whirlin, function synergistically in USH2 complex assembly in cochlear hair cells. In summary, our studies provide novel insight into the functional relationship between USH1 and USH2 proteins in the cochlea and the retina as well as the disease mechanisms underlying USH1 and USH2.


Assuntos
Proteínas de Transporte/genética , Proteínas da Matriz Extracelular/genética , Miosinas/genética , Síndromes de Usher/genética , Animais , Proteínas de Transporte/química , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Proteínas da Matriz Extracelular/química , Células Ciliadas Auditivas/patologia , Humanos , Camundongos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Miosina VIIa , Miosinas/química , Domínios PDZ/genética , Retina/metabolismo , Retina/patologia , Estereocílios/genética , Estereocílios/metabolismo , Estereocílios/patologia , Síndromes de Usher/patologia
8.
EMBO Rep ; 18(11): 2015-2029, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28893864

RESUMO

Lipopolysaccharide-responsive beige-like anchor protein (LRBA) belongs to the enigmatic class of BEACH domain-containing proteins, which have been attributed various cellular functions, typically involving intracellular protein and membrane transport processes. Here, we show that LRBA deficiency in mice leads to progressive sensorineural hearing loss. In LRBA knockout mice, inner and outer hair cell stereociliary bundles initially develop normally, but then partially degenerate during the second postnatal week. LRBA deficiency is associated with a reduced abundance of radixin and Nherf2, two adaptor proteins, which are important for the mechanical stability of the basal taper region of stereocilia. Our data suggest that due to the loss of structural integrity of the central parts of the hair bundle, the hair cell receptor potential is reduced, resulting in a loss of cochlear sensitivity and functional loss of the fraction of spiral ganglion neurons with low spontaneous firing rates. Clinical data obtained from two human patients with protein-truncating nonsense or frameshift mutations suggest that LRBA deficiency may likewise cause syndromic sensorineural hearing impairment in humans, albeit less severe than in our mouse model.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas do Citoesqueleto/genética , Células Ciliadas Auditivas/metabolismo , Perda Auditiva Neurossensorial/genética , Proteínas de Membrana/genética , Fosfoproteínas/genética , Trocadores de Sódio-Hidrogênio/genética , Estereocílios/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Adulto , Animais , Proteínas do Citoesqueleto/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/patologia , Audição/fisiologia , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fosfoproteínas/metabolismo , Domínios Proteicos , Transdução de Sinais , Trocadores de Sódio-Hidrogênio/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Estereocílios/patologia
9.
J Neurosci ; 37(13): 3447-3464, 2017 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-28209736

RESUMO

Mutations in the Pejvakin (PJVK) gene are thought to cause auditory neuropathy and hearing loss of cochlear origin by affecting noise-induced peroxisome proliferation in auditory hair cells and neurons. Here we demonstrate that loss of pejvakin in hair cells, but not in neurons, causes profound hearing loss and outer hair cell degeneration in mice. Pejvakin binds to and colocalizes with the rootlet component TRIOBP at the base of stereocilia in injectoporated hair cells, a pattern that is disrupted by deafness-associated PJVK mutations. Hair cells of pejvakin-deficient mice develop normal rootlets, but hair bundle morphology and mechanotransduction are affected before the onset of hearing. Some mechanotransducing shorter row stereocilia are missing, whereas the remaining ones exhibit overextended tips and a greater variability in height and width. Unlike previous studies of Pjvk alleles with neuronal dysfunction, our findings reveal a cell-autonomous role of pejvakin in maintaining stereocilia architecture that is critical for hair cell function.SIGNIFICANCE STATEMENT Two missense mutations in the Pejvakin (PJVK or DFNB59) gene were first identified in patients with audiological hallmarks of auditory neuropathy spectrum disorder, whereas all other PJVK alleles cause hearing loss of cochlear origin. These findings suggest that complex pathogenetic mechanisms underlie human deafness DFNB59. In contrast to recent studies, we demonstrate that pejvakin in auditory neurons is not essential for normal hearing in mice. Moreover, pejvakin localizes to stereociliary rootlets in hair cells and is required for stereocilia maintenance and mechanosensory function of the hair bundle. Delineating the site of the lesion and the mechanisms underlying DFNB59 will allow clinicians to predict the efficacy of different therapeutic approaches, such as determining compatibility for cochlear implants.


Assuntos
Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Mecanotransdução Celular , Proteínas/metabolismo , Animais , Linhagem Celular , Audição , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Mutação/genética , Proteínas/genética , Estereocílios/metabolismo , Estereocílios/patologia
10.
Hum Mol Genet ; 25(10): 2045-2059, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26936824

RESUMO

Most clinical reports have suggested that patients with congenital profound hearing loss have recessive mutations in deafness genes, whereas dominant alleles are associated with progressive hearing loss (PHL). Jackson shaker (Ush1gjs) is a mouse model of recessive deafness that exhibits congenital profound deafness caused by the homozygous mutation of Ush1g/Sans on chromosome 11. We found that C57BL/6J-Ush1gjs/+ heterozygous mice exhibited early-onset PHL (ePHL) accompanied by progressive degeneration of stereocilia in the cochlear outer hair cells. Interestingly, ePHL did not develop in mutant mice with the C3H/HeN background, thus suggesting that other genetic factors are required for ePHL development. Therefore, we performed classical genetic analyses and found that the occurrence of ePHL in Ush1gjs/+ mice was associated with an interval in chromosome 10 that contains the cadherin 23 gene (Cdh23), which is also responsible for human deafness. To confirm this mutation effect, we generated C57BL/6J-Ush1gjs/+, Cdh23c.753A/G double-heterozygous mice by using the CRISPR/Cas9-mediated Cdh23c.753A>G knock-in method. The Cdh23c.753A/G mice harbored a one-base substitution (A for G), and the homozygous A allele caused moderate hearing loss with aging. Analyses revealed the complete recovery of ePHL and stereocilia degeneration in C57BL/6J-Ush1gjs/+ mice. These results clearly show that the development of ePHL requires at least two mutant alleles of the Ush1g and Cdh23 genes. Our results also suggest that because the SANS and CDH23 proteins form a complex in the stereocilia, the interaction between these proteins may play key roles in the maintenance of stereocilia and the prevention of ePHL.


Assuntos
Caderinas/genética , Perda Auditiva/genética , Mutação/genética , Proteínas do Tecido Nervoso/genética , Alelos , Sequência de Aminoácidos/genética , Animais , Cromossomos Humanos Par 10/genética , Modelos Animais de Doenças , Células Ciliadas Auditivas Externas/patologia , Perda Auditiva/patologia , Heterozigoto , Homozigoto , Humanos , Camundongos , Estereocílios/patologia
11.
Biochem Biophys Res Commun ; 503(4): 2646-2652, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30107916

RESUMO

Auditory hair cells play an essential role in hearing. These cells convert sound waves, mechanical stimuli, into electrical signals that are conveyed to the brain via spiral ganglion neurons. The hair cells are located in the organ of Corti within the cochlea. They assemble in a special arrangement with three rows of outer hair cells and one row of inner hair cells. The proper differentiation and preservation of auditory hair cells are essential for acquiring and maintaining hearing function, respectively. Many genetic regulatory mechanisms underlying hair-cell differentiation and maintenance have been elucidated to date. However, the role of epigenetic regulation in hair-cell differentiation and maintenance has not been definitively demonstrated. CTCF is an essential epigenetic component that plays a primary role in the organization of global chromatin architecture. To determine the role of CTCF in mammalian hair cells, we specifically deleted Ctcf in developing hair cells by crossing Ctcffl/fl mice with Gfi1Cre/+ mice. Gfi1Cre; Ctcffl/fl mice did not exhibit obvious developmental defects in hair cells until postnatal day 8. However, at 3 weeks, the Ctcf deficiency caused intermittent degeneration of the stereociliary bundles of outer hair cells, resulting in profound hearing impairment. At 5 weeks, most hair cells were degenerated in Gfi1Cre; Ctcffl/fl mice, and defects in other structures of the organ of Corti, such as the tunnel of Corti and Nuel's space, became apparent. These results suggest that CTCF plays an essential role in maintaining hair cells and hearing function in mammalian cochlea.


Assuntos
Fator de Ligação a CCCTC/genética , Epigênese Genética , Células Ciliadas Auditivas/metabolismo , Audição/fisiologia , Gânglio Espiral da Cóclea/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fator de Ligação a CCCTC/deficiência , Diferenciação Celular , Movimento Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/patologia , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurogênese/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Gânglio Espiral da Cóclea/patologia , Estereocílios/metabolismo , Estereocílios/patologia
12.
PLoS Genet ; 11(3): e1005137, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25816005

RESUMO

Hereditary hearing loss is a clinically and genetically heterogeneous disorder. More than 80 genes have been implicated to date, and with the advent of targeted genomic enrichment and massively parallel sequencing (TGE+MPS) the rate of novel deafness-gene identification has accelerated. Here we report a family segregating post-lingual progressive autosomal dominant non-syndromic hearing loss (ADNSHL). After first excluding plausible variants in known deafness-causing genes using TGE+MPS, we completed whole exome sequencing in three hearing-impaired family members. Only a single variant, p.Arg185Pro in HOMER2, segregated with the hearing-loss phenotype in the extended family. This amino acid change alters a highly conserved residue in the coiled-coil domain of HOMER2 that is essential for protein multimerization and the HOMER2-CDC42 interaction. As a scaffolding protein, HOMER2 is involved in intracellular calcium homeostasis and cytoskeletal organization. Consistent with this function, we found robust expression in stereocilia of hair cells in the murine inner ear and observed that over-expression of mutant p.Pro185 HOMER2 mRNA causes anatomical changes of the inner ear and neuromasts in zebrafish embryos. Furthermore, mouse mutants homozygous for the targeted deletion of Homer2 present with early-onset rapidly progressive hearing loss. These data provide compelling evidence that HOMER2 is required for normal hearing and that its sequence alteration in humans leads to ADNSHL through a dominant-negative mode of action.


Assuntos
Proteínas de Transporte/genética , Orelha Interna/metabolismo , Exoma/genética , Perda Auditiva Neurossensorial/genética , Animais , Proteínas de Transporte/biossíntese , Cóclea/metabolismo , Cóclea/patologia , Orelha Interna/patologia , Regulação da Expressão Gênica , Perda Auditiva Neurossensorial/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Arcabouço Homer , Humanos , Camundongos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Estereocílios/genética , Estereocílios/patologia , Peixe-Zebra , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
13.
Hum Mol Genet ; 24(1): 37-49, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25124451

RESUMO

Hearing relies on the mechanosensory inner and outer hair cells (OHCs) of the organ of Corti, which convert mechanical deflections of their actin-rich stereociliary bundles into electrochemical signals. Several actin-associated proteins are essential for stereocilia formation and maintenance, and their absence leads to deafness. One of the most abundant actin-bundling proteins of stereocilia is plastin 1, but its function has never been directly assessed. Here, we found that plastin 1 knock-out (Pls1 KO) mice have a moderate and progressive form of hearing loss across all frequencies. Auditory hair cells developed normally in Pls1 KO, but in young adult animals, the stereocilia of inner hair cells were reduced in width and length. The stereocilia of OHCs were comparatively less affected; however, they also showed signs of degeneration in ageing mice. The hair bundle stiffness and the acquisition of the electrophysiological properties of hair cells were unaffected by the absence of plastin 1, except for a significant change in the adaptation properties, but not the size of the mechanoelectrical transducer currents. These results show that in contrast to other actin-bundling proteins such as espin, harmonin or Eps8, plastin 1 is dispensable for the initial formation of stereocilia. However, the progressive hearing loss and morphological defects of hair cells in adult Pls1 KO mice point at a specific role for plastin 1 in the preservation of adult stereocilia and optimal hearing. Hence, mutations in the human PLS1 gene may be associated with relatively mild and progressive forms of hearing loss.


Assuntos
Células Ciliadas Auditivas Internas/patologia , Perda Auditiva/fisiopatologia , Glicoproteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Estereocílios/patologia , Fatores Etários , Animais , Perda Auditiva/genética , Perda Auditiva/patologia , Humanos , Camundongos , Camundongos Knockout , Mutação
14.
Hum Mol Genet ; 24(21): 6213-28, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26307081

RESUMO

Usher syndrome (USH) is the most common inherited deaf-blindness with the majority of USH causative genes also involved in nonsyndromic recessive deafness (DFNB). The mechanism underlying this disease variation of USH genes is unclear. Here, we addressed this issue by investigating the DFNB31 gene, whose mutations cause USH2D or DFNB31 depending on their position. We found that the mouse DFNB31 ortholog (Dfnb31) expressed different mRNA variants and whirlin protein isoforms in the cochlea and retina, where these isoforms played different roles spatially and temporally. Full-length (FL-) whirlin in photoreceptors and hair cell stereociliary bases is important for the USH type 2 protein complex, while FL- and C-terminal (C-) whirlins in hair cell stereociliary tips participate in stereociliary elongation. Mutations in the whirlin N-terminal region disrupted FL-whirlin isoform in the inner ear and retina but not C-whirlin in the inner ear, and led to retinal degeneration as well as moderate to severe hearing loss. By contrast, a mutation in the whirlin C-terminal region eliminated all normal whirlin isoforms but generated a truncated N-terminal whirlin protein fragment, which was partially functional in the retina and thus prevented retinal degeneration. Mice with this mutation had profound hearing loss. In summary, disruption of distinct whirlin isoforms by Dfnb31 mutations leads to a variety of phenotype configurations and may explain the mechanism underlying the different disease manifestations of human DFNB31 mutations. Our findings have a potential to improve diagnosis and treatment of USH disease and quality of life in USH patients.


Assuntos
Cóclea/metabolismo , Perda Auditiva Neurossensorial/metabolismo , Proteínas de Membrana/biossíntese , Retina/metabolismo , Síndromes de Usher/metabolismo , Animais , Células Ciliadas Auditivas/metabolismo , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Mutação , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/fisiologia , RNA Mensageiro , Estereocílios/patologia , Síndromes de Usher/genética , Síndromes de Usher/patologia
15.
Biochim Biophys Acta Gen Subj ; 1861(10): 2485-2493, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28571946

RESUMO

Severe auditory impairment observed in GM3 synthase-deficient mice and humans indicates that glycosphingolipids, especially sialic-acid containing gangliosides, are indispensable for hearing. Gangliosides associate with glycoproteins to form membrane microdomains, the composition of which plays a special role in maintaining the structural and functional integrity of hair cells. These microdomains, also called lipid rafts, connect with intracellular signaling and cytoskeletal systems to link cellular responses to environmental cues. During development, ganglioside species are expressed in distinctive spatial and temporal patterns throughout the cochlea. In both mice and humans, blocking particular steps of ganglioside metabolism produces distinctive neurological and auditory phenotypes. Thus each ganglioside species may have specific, non-overlapping functions within the cochlea, central auditory network, and brain.


Assuntos
Encéfalo/metabolismo , Surdez/metabolismo , Epilepsia/metabolismo , N-Acetilgalactosaminiltransferases/deficiência , Sialiltransferases/deficiência , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Surdez/genética , Surdez/patologia , Epilepsia/genética , Epilepsia/patologia , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Audição/genética , Humanos , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Knockout , N-Acetilgalactosaminiltransferases/genética , Organogênese/genética , Sialiltransferases/genética , Sialiltransferases/metabolismo , Transdução de Sinais , Estereocílios/metabolismo , Estereocílios/patologia
16.
Hum Mutat ; 37(10): 991-1003, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27375115

RESUMO

Deafness in humans is a common neurosensory disorder and is genetically heterogeneous. Across diverse ethnic groups, mutations of MYO15A at the DFNB3 locus appear to be the third or fourth most common cause of autosomal-recessive, nonsyndromic deafness. In 49 of the 67 exons of MYO15A, there are currently 192 recessive mutations identified, including 14 novel mutations reported here. These mutations are distributed uniformly across MYO15A with one enigmatic exception; the alternatively spliced giant exon 2, encoding 1,233 residues, has 17 truncating mutations but no convincing deafness-causing missense mutations. MYO15A encodes three distinct isoform classes, one of which is 395 kDa (3,530 residues), the largest member of the myosin superfamily of molecular motors. Studies of Myo15 mouse models that recapitulate DFNB3 revealed two different pathogenic mechanisms of hearing loss. In the inner ear, myosin 15 is necessary both for the development and the long-term maintenance of stereocilia, mechanosensory sound-transducing organelles that extend from the apical surface of hair cells. The goal of this Mutation Update is to provide a comprehensive review of mutations and functions of MYO15A.


Assuntos
Surdez/genética , Surdez/patologia , Mutação , Miosinas/genética , Miosinas/metabolismo , Processamento Alternativo , Animais , Surdez/metabolismo , Modelos Animais de Doenças , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Orelha Interna/patologia , Éxons , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Estereocílios/metabolismo , Estereocílios/patologia
17.
Gene Ther ; 23(5): 415-23, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26886463

RESUMO

Once inner ear hair cells (HCs) are damaged by drugs, noise or aging, their apical structures including the stereociliary arrays are frequently the first cellular feature to be lost. Although this can be followed by progressive loss of HC somata, a significant number of HC bodies often remain even after stereociliary loss. However, in the absence of stereocilia they are nonfunctional. HCs can sometimes be regenerated by Atoh1 transduction or Notch inhibition, but they also may lack stereociliary bundles. It is therefore important to develop methods for the regeneration of stereocilia, in order to achieve HC functional recovery. Espin is an actin-bundling protein known to participate in sterociliary elongation during development. We evaluated stereociliary array regeneration in damaged vestibular sensory epithelia in tissue culture, using viral vector transduction of two espin isoforms. Utricular HCs were damaged with aminoglycosides. The utricles were then treated with a γ-secretase inhibitor, followed by espin or control transduction and histochemistry. Although γ-secretase inhibition increased the number of HCs, few had stereociliary arrays. In contrast, 46 h after espin1 transduction, a significant increase in hair-bundle-like structures was observed. These were confirmed to be immature stereociliary arrays by scanning electron microscopy. Increased uptake of FM1-43 uptake provided evidence of stereociliary function. Espin4 transduction had no effect. The results demonstrate that espin1 gene therapy can restore stereocilia on damaged or regenerated HCs.


Assuntos
Células Ciliadas Auditivas Internas/ultraestrutura , Proteínas dos Microfilamentos/genética , Receptores Notch/genética , Regeneração/genética , Estereocílios/genética , Aminoglicosídeos/toxicidade , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Animais , Cóclea/efeitos dos fármacos , Cóclea/patologia , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Células Ciliadas Auditivas Internas/patologia , Humanos , Camundongos , Proteínas dos Microfilamentos/uso terapêutico , Microscopia Eletrônica de Varredura , Compostos de Piridínio/farmacologia , Compostos de Amônio Quaternário/farmacologia , Receptores Notch/antagonistas & inibidores , Estereocílios/patologia , Transdução Genética
18.
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
19.
Biochem Biophys Res Commun ; 479(4): 703-707, 2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27693694

RESUMO

The TPRN gene encodes taperin, which is prominently present at the taper region of hair cell stereocilia. Mutations in TPRN have been reported to cause autosomal recessive nonsyndromic deafness 79(DFNB 79). To investigate the role of taperin in pathogenesis of hearing loss, we generated TPRN knockout mice using TALEN technique. Sanger sequencing confirmed an 11 bp deletion at nucleotide 177-187 in exon 1 of TPRN, which results in a truncated form of taperin protein. Heterozygous TPRN+/- mice showed apparently normal auditory phenotypes to their wide-type (WT) littermates. Homozygous TPRN-/- mice exhibited progressive sensorineural hearing loss as reflected by auditory brainstem response to both click and tone burst stimuli at postnatal days 15 (P15), 30 (P30), and 60 (P60). Alex Fluor-594 phalloidin labeling showed no obvious difference in hair cell numbers in the cochlea between TPRN-/- mice and WT mice under light microscope. However, scanning electronic microscopy revealed progressive degeneration of inner hair cell stereocilia, from apparently normal at postnatal days 3 (P3) to scattered absence at P15 and further to substantial loss at P30. The outer hair cell stereocilia also showed progressive degeneration, though much less severe, Collectively, we conclude that taperin plays an important role in maintenance of hair cell stereocilia. Establishment of TPRN knockout mice enables further investigation into the function of this gene.


Assuntos
Surdez/genética , Surdez/patologia , Células Ciliadas Auditivas/ultraestrutura , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Proteínas/fisiologia , Estereocílios/patologia , Animais , Células Ciliadas Auditivas/metabolismo , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Proteínas/genética , Deleção de Sequência , Estereocílios/metabolismo
20.
J Neurosci ; 32(8): 2762-72, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22357859

RESUMO

Recent studies have shown that mutations in PTPRQ, a gene encoding a receptor-like inositol lipid phosphatase, cause recessive, nonsyndromic, hereditary hearing loss with associated vestibular dysfunction. Although null mutations in Ptprq cause the loss of high-frequency auditory hair cells and deafness in mice, a loss of vestibular hair cells and overt behavioral defects characteristic of vestibular dysfunction have not been described. Hair bundle structure and vestibular function were therefore examined in Ptprq mutant mice. Between postnatal days 5 and 16, hair bundles in the extrastriolar regions of the utricle in Ptprq(-/-) mice become significantly longer than those in heterozygous controls. This increase in length (up to 50%) is accompanied by the loss and fusion of stereocilia. Loss and fusion of stereocilia also occurs in the striolar region of the utricle in Ptprq(-/-) mice, but is not accompanied by hair bundle elongation. These abnormalities persist until 12 months of age but are not accompanied by significant hair cell loss. Hair bundle defects are also observed in the saccule and ampullae of Ptprq(-/-) mice. At ∼3 months of age, vestibular evoked potentials were absent from the majority (12 of 15) of Ptprq(-/-) mice examined, and could only be detected at high stimulus levels in the other 3 mutants. Subtle but distinct defects in swimming behavior were detected in most (seven of eight) mutants tested. The results reveal a distinct phenotype in the vestibular system of Ptprq(-/-) mice and suggest similar hair bundle defects may underlie the vestibular dysfunction reported in humans with mutations in PTPRQ.


Assuntos
Potenciais Evocados Auditivos/fisiologia , Células Ciliadas Auditivas/patologia , Células Ciliadas Auditivas/ultraestrutura , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/deficiência , Doenças Vestibulares , Estimulação Acústica/métodos , Actinas/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Potenciais Evocados Auditivos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Mutação/genética , Faloidina/metabolismo , Psicoacústica , Receptores Acoplados a Proteínas G/genética , Estereocílios/patologia , Estereocílios/ultraestrutura , Doenças Vestibulares/genética , Doenças Vestibulares/patologia , Doenças Vestibulares/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA