Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Dev Biol ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38869222

RESUMO

Mutations in the gene encoding Tre2/Bub2/Cdc16 (TBC)1 domain family member 24 (TBC1D24) protein are associated with a variety of neurological disorders, ranging from non-syndromic hearing loss to drug-resistant lethal epileptic encephalopathy and DOORS syndrome [Deafness, Onychodystrophy, Osteodystrophy, intellectual disability (formerly referred to as mental Retardation), and Seizures]. TBC1D24 is a vesicle-associated protein involved in neural crest cell and neuronal migration, maturation, and neurotransmission. In the cochlea, TBC1D24 has been detected in auditory neurons, but few reliable and convergent data exist about the sensory epithelium. Here, the expression of TBC1D24 has been characterized via immunolabelling throughout the postnatal maturation of the mouse cochlear sensory epithelium. TBC1D24 was detected in glia-like non-sensory epithelial cells during early developmental stages. In contrast, TBC1D24 was virtually absent in adjacent sensory hair cells. This expression distinguishing non-sensory from sensory epithelial cells almost disappears around the onset of hearing. Until now, TBC1D24 was mainly described as a neuronal protein either in the brain or in the cochlea. The present observations suggest that TBC1D24 could also regulate vesicle trafficking in cochlear glia-like non-sensory epithelial cells. For a long time, research about epilepsy has been mainly neurocentric. However, there is now evidence proving that glial cell dysregulation contribute to pathogenesis of epilepsy and neurodevelopmental disorders. As a consequence, exploring the possibility that TBC1D24 could also have a role in glial cells of the central nervous system could help to gain insight into TBC1D24-related neurological pathogenesis.

2.
Dev Dyn ; 252(2): 239-246, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36106826

RESUMO

In the cochlea, connexin 26 (Cx26) and connexin 30 (Cx30) co-assemble into two types of homomeric and heteromeric gap junctions between adjacent non-sensory epithelial cells. These channels provide a mechanical coupling between connected cells, and their activity is critical to maintain cochlear homeostasis. Many of the mutations in GJB2 or GJB6, which encode Cx26 and Cx30 in humans, impair the formation of membrane channels and cause autosomal syndromic and non-syndromic hearing loss. Thus, deciphering the connexin trafficking pathways in situ should represent a major step forward in understanding the pathogenic significance of many of these mutations. A growing body of evidence now suggests that Cx26/Cx30 heteromeric and Cx30 homomeric channels display distinct assembly mechanisms. Here, we review the most recent advances that have been made toward unraveling the biogenesis and stability of these gap junctions in the cochlea.


Assuntos
Conexinas , Surdez , Humanos , Conexinas/genética , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Cóclea/metabolismo , Conexina 30/genética , Conexina 30/metabolismo , Surdez/genética
3.
Ibrain ; 8(4): 431-441, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37786584

RESUMO

Mutations and deletions in the gene or upstream of the gene encoding the POU3F4 transcription factor cause X-linked progressive deafness DFNX2 and additional neurodevelopmental disorders in humans. Hearing loss can be purely sensorineural or mixed, that is, with both conductive and sensorineural components. Affected males show anatomical abnormalities of the inner ear, which are jointly defined as incomplete partition type III. Current approaches to improve hearing and speech skills of DFNX2 patients do not seem to be fully effective. Owing to inner ear malformations, cochlear implantation is surgically difficult and may predispose towards severe complications. Even in cases where implantation is safely performed, hearing and speech outcomes remain highly variable among patients. Mouse models for DFNX2 deafness revealed that sensorineural loss could arise from a dysfunction of spiral ligament fibrocytes in the lateral wall of the cochlea, which leads to reduced endocochlear potential. Highly positive endocochlear potential is critical for sensory hair cell mechanotransduction and hearing. In this context, here, we propose to develop a therapeutic approach in male Pou3f4 -/y mice based on an adeno-associated viral (AAV) vector-mediated gene transfer in cochlear spiral ligament fibrocytes. Among a broad range of AAV vectors, AAV7 was found to show a strong tropism for the spiral ligament. Thus, we suggest that an AAV7-mediated delivery of Pou3f4 complementary DNA in the spiral ligament of Pou3f4 -/y mice could represent an attractive strategy to prevent fibrocyte degeneration and to restore normal cochlear functions and properties, including a positive endocochlear potential, before hearing loss progresses to profound deafness.

4.
Hear Res ; 409: 108311, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34311268

RESUMO

There is now growing evidence that hypercholesterolemia and high serum levels of low-density lipoproteins (LDL) predispose to sensorineural hearing loss. Circulating LDL-cholesterol is delivered to peripheral tissues via LDL receptor (LDLR) -mediated endocytosis. Recently, it has been shown that LDLR gene polymorphisms are associated with higher susceptibility to sudden deafness. These findings suggested that we should investigate the expression of LDLR from the postnatal maturation of the mouse cochlea until adulthood. In the cochlea of newborn mice, we observed that LDLR is mostly expressed in the lateral wall of the cochlea, especially in a band of cells directly facing the cochlear duct. Moreover, LDLR is expressed in the inner and outer hair cells, as well as in the adjacent greater epithelial ridge. In early postnatal stages, LDLR is expressed in the marginal cells of the immature stria vascularis, in the root cells of the spiral ligament, and in the adjacent outer sulcus cells. At the same time, LDLR begins to be expressed in the pillar cells of the immature organ of Corti. From the onset of hearing, LDLR is expressed in the marginal cells of the stria vascularis, in the outer sulcus cells, and in the capillaries of the adjacent spiral ligament. In the organ of Corti, LDLR is expressed in outer pillar cells and Deiters' cells, i.e. in the non-sensory supporting cells that directly surround the outer hair cells. These cells are believed to provide a mechanical coupling with the outer hair cells to modulate electromotility and cochlear amplification. In the stria vascularis of three-month-old mice, LDLR is further expressed in both marginal and intermediate cells. Overall, our results suggest that LDLR is mostly present in cochlear cells that are involved in endolymph homeostasis and cochlear amplification. Further functional studies will be needed to unravel how LDLR regulates extracellular and intracellular levels of cholesterol and lipoproteins in the cochlea, and how it could influence cochlear homeostasis.


Assuntos
Cóclea , Endolinfa , Homeostase , Animais , Camundongos , Receptores de LDL/genética , Estria Vascular
5.
Brain Res Bull ; 174: 153-160, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34139316

RESUMO

Chromosome 13q deletions encompassing EFNB2, which encodes the transmembrane protein ephrin-B2, are likely to cause syndromic forms of sensorineural hearing loss of unclear origin. Thus, unravelling the pathogenic mechanisms could help to improve therapeutic strategies. In the cochlea, adjacent non-sensory epithelial cells are connected via gap junction channels, the activity of which is critical to maintain cochlear homeostasis. Here we show that ephrin-B2 promotes the assembly of connexin 30 (Cx30) gap junction plaques (GJPs) between adjacent non-sensory Deiters' cells. An in situ proximity ligation assay revealed that ephrin-B2 preferentially interacts with Cx30 in the periphery of the GJPs, i.e. where newly synthesized connexin hemichannels accrue to the GJP. Moreover, we observed that heterozygous mice encoding an Efnb2 null allele display excessive clathrin-mediated internalization of Cx30 GJPs in early postnatal stages. Finally, an in vitro organotypic assay revealed that ectopic activation of ephrin-B2 reverse signalling promotes the internalization of Cx30 GJPs. These data argue in favor of a cell-autonomous, Eph receptor-independent role of ephrin-B2 in the assembly of Cx30 GJPs. According to recent observations, early GJP degradation could certainly play a role in the pathogenic process leading to progressive sensorineural hearing loss due to Efnb2/EFNB2 haploinsufficiency.


Assuntos
Cóclea/patologia , Sinapses Elétricas/patologia , Endocitose/genética , Efrina-B2/genética , Animais , Conexina 30/biossíntese , Conexina 30/genética , Efrina-B2/farmacologia , Haploinsuficiência , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Heterozigoto , Camundongos , Camundongos Knockout , Transdução de Sinais/genética
6.
Int J Mol Sci ; 22(3)2021 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-33498839

RESUMO

Unlike in most eukaryotic cells, the genetic information of budding yeast in the exponential growth phase is only present in the form of decondensed chromatin, a configuration that does not allow its visualization in cell nuclei conventionally prepared for transmission electron microscopy. In this work, we studied the distribution of chromatin and its relationships to the nucleolus using different cytochemical and immunocytological approaches applied to yeast cells subjected to hyperosmotic shock. Our results show that osmotic shock induces the formation of heterochromatin patches in the nucleoplasm and intranucleolar regions of the yeast nucleus. In the nucleolus, we further revealed the presence of osmotic shock-resistant DNA in the fibrillar cords which, in places, take on a pinnate appearance reminiscent of ribosomal genes in active transcription as observed after molecular spreading ("Christmas trees"). We also identified chromatin-associated granules whose size, composition and behaviour after osmotic shock are reminiscent of that of mammalian perichromatin granules. Altogether, these data reveal that it is possible to visualize heterochromatin in yeast and suggest that the yeast nucleus displays a less-effective compartmentalized organization than that of mammals.


Assuntos
Núcleo Celular/ultraestrutura , Cromatina/ultraestrutura , Pressão Osmótica , Saccharomyces cerevisiae/genética , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , Núcleo Celular/metabolismo , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Heterocromatina , Histocitoquímica , Microscopia Eletrônica de Transmissão , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura
7.
Hear Res ; 400: 108137, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33291008

RESUMO

In the cochlea, connexins 26 (Cx26) and 30 (Cx30) largely co-assemble into heteromeric gap junctions, which connect adjacent non-sensory epithelial cells. These channels are believed to ensure the rapid removal of K+ away from the base of sensory hair cells, resulting in K+ recycling back to the endolymph to maintain cochlear homeostasis. Many of the mutations in GJB2 and GJB6, which encode CX26 and CX30, impair the formation of membrane channels and cause autosomal hearing loss in humans. Although recent advances have been made, several important questions remain about connexin trafficking and gap junction biogenesis. Here we show that tricellular adherens junctions present at the crossroad between adjacent gap junction plaques, provide an unexpected cell surface delivery platform for Cx26/Cx30 oligomers. Using an in situ proximity ligation assay, we detected the presence of non-junctional Cx26/Cx30 oligomers within lipid raft-enriched tricellular junction sites. In addition, we observed that cadherin homophilic interactions are critically involved in microtubule-mediated trafficking of Cx26/Cx30 oligomers to the cell surface. Overall, our results unveil an unexpected role for tricellular junctions in the trafficking and assembly of membrane channels.


Assuntos
Junções Aderentes , Cóclea , Conexina 26/genética , Conexina 30 , Conexinas/genética , Junções Comunicantes , Humanos
8.
J Struct Biol ; 208(3): 107398, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31585176

RESUMO

The nucleolus is a multifunctional structure of the eukaryotic cell nucleus. However, its primary role is ribosome formation. Although the factors and mechanisms involved in ribogenesis are well conserved in eukaryotes, two types of nucleoli have been observed under the electron microscope: a tricompartmentalized nucleolus in amniotes and a bicompartmentalized nucleolus in other species. A recent study has also revealed that turtles, although belonging to amniotes, displayed a nucleolus with bipartite organization, suggesting that this reptile group may have carried out a reversion phenomenon during evolution. In this study, we examine in great detail the functional organization of the turtle nucleolus. In liver and spleen cells cultured in vitro, we confirm that the turtle nucleolus is mainly formed by two components: a fibrillar zone surrounded by a granular zone. We further show that the fibrillar zone includes densely-contrasted strands, which are positive after silver-stained Nucleolar Organizer Region (Ag-NOR) staining and DNA labelling. We also reveal that the dense strands condensed into a very compact mass within the fibrillar zone after a treatment with actinomycin D or 5,6-dichlorobenzimidazole riboside. Finally, by using pulse-chase experiments with BrUTP, three-dimensional image reconstructions of confocal optical sections, and electron microscopy analysis of ultrathin sections, we show that the topological and spatial dynamics of rRNA within the nucleolus extend from upstream binding factor (UBF)-positive sites in the fibrillar zone to the granular zone, without ever releasing the positive sites for the UBF. Together, these results seem to clearly indicate that the compartmentalization of the turtle nucleolus into two main components reflects a less orderly organization of ribosome formation.


Assuntos
Nucléolo Celular/genética , Nucléolo Celular/ultraestrutura , Tartarugas , Animais , Nucléolo Celular/efeitos dos fármacos , Células Cultivadas , Dactinomicina/farmacologia , Diclororribofuranosilbenzimidazol/farmacologia , Fígado/citologia , Microscopia Confocal , Região Organizadora do Nucléolo , RNA Ribossômico/metabolismo , Baço/citologia
9.
Proc Natl Acad Sci U S A ; 116(16): 8010-8017, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30936319

RESUMO

Noise overexposure causes oxidative stress, leading to auditory hair cell damage. Adaptive peroxisome proliferation involving pejvakin, a peroxisome-associated protein from the gasdermin family, has been shown to protect against this harmful oxidative stress. However, the role of pejvakin in peroxisome dynamics and homeostasis remains unclear. Here we show that sound overstimulation induces an early and rapid selective autophagic degradation of peroxisomes (pexophagy) in auditory hair cells from wild-type, but not pejvakin-deficient (Pjvk-/-), mice. Noise overexposure triggers recruitment of the autophagosome-associated protein MAP1LC3B (LC3B; microtubule-associated protein 1 light chain 3ß) to peroxisomes in wild-type, but not Pjvk-/-, mice. We also show that pejvakin-LC3B binding involves an LC3-interacting region within the predicted chaperone domain of pejvakin. In transfected cells and in vivo transduced auditory hair cells, cysteine mutagenesis experiments demonstrated the requirement for both C328 and C343, the two cysteine residues closest to the C terminus of pejvakin, for reactive oxygen species-induced pejvakin-LC3B interaction and pexophagy. The viral transduction of auditory hair cells from Pjvk-/- mice in vivo with both Pjvk and Lc3b cDNAs completely restored sound-induced pexophagy, fully prevented the development of oxidative stress, and resulted in normal levels of peroxisome proliferation, whereas Pjvk cDNA alone yielded only a partial correction of the defects. Overall, our results demonstrate that pexophagy plays a key role in noise-induced peroxisome proliferation and identify defective pexophagy as a cause of noise-induced hearing loss. They suggest that pejvakin acts as a redox-activated pexophagy receptor/adaptor, thereby identifying a previously unknown function of gasdermin family proteins.


Assuntos
Células Ciliadas Auditivas , Perda Auditiva Provocada por Ruído , Macroautofagia/fisiologia , Proteínas , Animais , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/fisiologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Perda Auditiva Provocada por Ruído/prevenção & controle , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/química , Proteínas/genética , Proteínas/metabolismo
10.
Dev Biol ; 449(1): 35-40, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30771305

RESUMO

In mammals, the functional development of the cochlea requires the tight regulation of multiple molecules and signalling pathways including fibroblast growth factors, bone morphogenetic proteins, Wnt and Notch signalling pathways. Over the last decade, the Eph/ephrin system also emerged as a key player of the development and function of the mammalian cochlea. In this review, we discuss the recent advances on the role of Eph/ephrin signalling in patterning the cochlear sensory epithelium and the complex innervation of mechanosensory hair cells by spiral ganglion neurons. Finally, we address the issue of a syndromic form of hearing loss caused by a deficient member of the Eph/ephrin family.


Assuntos
Cóclea/metabolismo , Efrinas/metabolismo , Mamíferos/metabolismo , Receptores da Família Eph/metabolismo , Transdução de Sinais , Animais , Orientação de Axônios , Humanos
11.
Hear Res ; 374: 69-75, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30732922

RESUMO

Hereditary hearing loss affects about 1 per 1000 children. Mutations in GJB2, which encodes the connexin 26 protein (Cx26) involved in cochlear homeostasis, are found in about 50% of patients with autosomal recessive non-syndromic hearing loss. Deciphering the trafficking pathway of cochlear Cx26 in situ should represent an advance in understanding the pathogenic significance of many of these mutations. Connexins trafficking and delivery to lipid raft-associated gap junction plaques usually requires successively microtubule and actin networks. Here we show that cochlear Cx26 exhibits an unusual trafficking pathway. We observed that Cx26 assembly occurs in non-lipid raft membrane domains and that junctional plaques are devoid of actin and associated zonula occludens proteins. Using cytoskeleton-disrupting drugs in organotypic culture, we found that cochlear Cx26 gap junction assembly requires microtubules but not actin filaments. Altogether, our data provide an unexpected insight into Cx26 trafficking pathway and gap junction assembly in the cochlea.


Assuntos
Cóclea/metabolismo , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Criança , Conexina 26 , Conexinas/genética , Surdez/genética , Surdez/metabolismo , Humanos , Técnicas In Vitro , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Mutação , Transporte Proteico , Proteínas da Zônula de Oclusão/metabolismo
12.
iScience ; 11: 246-257, 2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30639848

RESUMO

The cochlear sensory epithelium contains a functionally important triangular fluid-filled space between adjacent pillar cells referred to as the tunnel of Corti. However, the molecular mechanisms leading to local cell-cell separation during development remain elusive. Here we show that EphA4 associates with ADAM10 to promote the destruction of E-cadherin-based adhesions between adjacent pillar cells. These cells fail to separate from each other, and E-cadherin abnormally persists at the pillar cell junction in EphA4 forward-signaling-deficient mice, as well as in the presence of ADAM10 inhibitor. Using immunolabeling and an in situ proximity ligation assay, we found that EphA4 forms a complex with E-cadherin and its sheddase ADAM10, which could be activated by ephrin-B2 across the pillar cell junction to trigger the cleavage of E-cadherin. Altogether, our findings provide a new molecular insight into the regulation of adherens junctions, which might be extended to a variety of physiological or pathological processes.

13.
Mech Dev ; 155: 8-14, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30296578

RESUMO

Many of the mutations in GJB2 and GJB6, which encode connexins 26 and 30 (Cx26 and Cx30), impair the formation of membrane channels and cause autosomal syndromic and non-syndromic hearing loss. In cochlear non-sensory supporting cells, Cx26 and Cx30 form two types of homomeric and heteromeric gap junctions. The biogenesis processes of these channels occurring in situ remain largely unknown. Here we show that Cx30 homomeric and Cx26/Cx30 heteromeric gap junctions exhibit distinct assembly mechanisms in the cochlea. When expressed as homomeric channels, Cx30 preferentially interacts with ß-actin in the peripheral non-junctional membrane region, called perinexus, and strongly relies on the actin network for gap junction plaque assembly. In contrast, we found that Cx26/Cx30 heteromeric gap junction plaques are devoid of perinexus and associated actin network, and resist to actin-depolymerizating drug. This supports that Cx26/Cx30 oligomers could be directly delivered from the interior of the cell to the junctional plaque. Altogether, our data provide a novel insight in homomeric and heteromeric gap junction plaque assembly in the cochlea.


Assuntos
Cóclea/metabolismo , Conexina 30/metabolismo , Animais , Conexina 26/metabolismo , Surdez/metabolismo , Junções Comunicantes/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
14.
Cell ; 163(4): 894-906, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26544938

RESUMO

A deficiency in pejvakin, a protein of unknown function, causes a strikingly heterogeneous form of human deafness. Pejvakin-deficient (Pjvk(-/-)) mice also exhibit variable auditory phenotypes. Correlation between their hearing thresholds and the number of pups per cage suggest a possible harmful effect of pup vocalizations. Direct sound or electrical stimulation show that the cochlear sensory hair cells and auditory pathway neurons of Pjvk(-/-) mice and patients are exceptionally vulnerable to sound. Subcellular analysis revealed that pejvakin is associated with peroxisomes and required for their oxidative-stress-induced proliferation. Pjvk(-/-) cochleas display features of marked oxidative stress and impaired antioxidant defenses, and peroxisomes in Pjvk(-/-) hair cells show structural abnormalities after the onset of hearing. Noise exposure rapidly upregulates Pjvk cochlear transcription in wild-type mice and triggers peroxisome proliferation in hair cells and primary auditory neurons. Our results reveal that the antioxidant activity of peroxisomes protects the auditory system against noise-induced damage.


Assuntos
Perda Auditiva Provocada por Ruído/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peroxissomos/metabolismo , Proteínas/metabolismo , Animais , Vias Auditivas , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Perda Auditiva Provocada por Ruído/patologia , Humanos , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Estresse Oxidativo , Proteínas/genética
15.
Nat Commun ; 6: 7017, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25923646

RESUMO

In mammals, cochlear sensory hair cells that are responsible for hearing are postmitotic and are not replaced after loss. One of the most promising strategies to regenerate hair cells is to identify and inhibit the factors preventing the conversion of adjacent non-sensory supporting cells into hair cells. Here we demonstrate that mammalian hair cells can be directly generated from supporting cells by inhibition of ephrin-B2 signalling. Using either ephrin-B2 conditional knockout mice, shRNA-mediated gene silencing or soluble inhibitors, we found that downregulation of ephrin-B2 signalling at embryonic stages results in supporting cell translocation into hair cell layers and subsequent switch in cell identity from supporting cell to hair cell fate. As transdifferentiation is here a result of displacement across boundary, this original finding presents the interest that newly generated hair cells directly integrate either hair cell layer, then would be likely more rapidly able to fit into functional circuitry.


Assuntos
Transdiferenciação Celular , Cóclea/embriologia , Efrina-B2/metabolismo , Animais , Cóclea/metabolismo , Efrina-B2/antagonistas & inibidores , Células Ciliadas Auditivas , Camundongos
16.
Nat Commun ; 4: 1438, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23385583

RESUMO

Hearing requires an optimal afferent innervation of sensory hair cells by spiral ganglion neurons in the cochlea. Here we report that complementary expression of ephrin-A5 in hair cells and EphA4 receptor among spiral ganglion neuron populations controls the targeting of type I and type II afferent fibres to inner and outer hair cells, respectively. In the absence of ephrin-A5 or EphA4 forward signalling, a subset of type I projections aberrantly overshoot the inner hair cell layer and invade the outer hair cell area. Lack of type I afferent synapses impairs neurotransmission from inner hair cells to the auditory nerve. By contrast, radial shift of type I projections coincides with a gain of presynaptic ribbons that could enhance the afferent signalling from outer hair cells. Ephexin-1, cofilin and myosin light chain kinase act downstream of EphA4 to induce type I spiral ganglion neuron growth cone collapse. Our findings constitute the first identification of an Eph/ephrin-mediated mutual repulsion mechanism responsible for specific sorting of auditory projections in the cochlea.


Assuntos
Vias Aferentes/metabolismo , Efrina-A5/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Receptor EphA4/metabolismo , Transdução de Sinais , Animais , Limiar Auditivo , Efrina-A5/deficiência , Efrina-A5/genética , Regulação da Expressão Gênica , Cones de Crescimento/metabolismo , Imuno-Histoquímica , Camundongos , Microscopia Confocal , Modelos Biológicos , Receptor EphA4/genética , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/inervação , Gânglio Espiral da Cóclea/metabolismo , Transmissão Sináptica
17.
Am J Physiol Cell Physiol ; 301(4): C750-61, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21753183

RESUMO

In mammals, sensorineural deafness results from damage to the auditory receptors of the inner ear, the nerve pathways to the brain or the cortical area that receives sound information. In this review, we first focused on the cellular and molecular events taking part to spiral ganglion axon growth, extension to the organ of Corti, and refinement. In the second half, we considered the functional maturation of synaptic contacts between sensory hair cells and their afferent projections. A better understanding of all these processes could open insights into novel therapeutic strategies aimed to re-establish primary connections from sound transducers to the ascending auditory nerve pathways.


Assuntos
Cóclea/inervação , Mamíferos/anatomia & histologia , Neurônios Aferentes/citologia , Animais , Cóclea/crescimento & desenvolvimento , Audição/fisiologia , Neurônios Aferentes/fisiologia , Sinapses/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...