Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 163(4): 894-906, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26544938

RESUMEN

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.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Peroxisomas/metabolismo , Proteínas/metabolismo , Animales , Vías Auditivas , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Pérdida Auditiva Provocada por Ruido/patología , Humanos , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Estrés Oxidativo , Proteínas/genética
2.
Mol Ther ; 32(3): 800-817, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38243601

RESUMEN

Hearing loss is a major health concern affecting millions of people worldwide with currently limited treatment options. In clarin-2-deficient Clrn2-/- mice, used here as a model of progressive hearing loss, we report synaptic auditory abnormalities in addition to the previously demonstrated defects of hair bundle structure and mechanoelectrical transduction. We sought an in-depth evaluation of viral-mediated gene delivery as a therapy for these hearing-impaired mice. Supplementation with either the murine Clrn2 or human CLRN2 genes preserved normal hearing in treated Clrn2-/- mice. Conversely, mutated forms of CLRN2, identified in patients with post-lingual moderate to severe hearing loss, failed to prevent hearing loss. The ectopic expression of clarin-2 successfully prevented the loss of stereocilia, maintained normal mechanoelectrical transduction, preserved inner hair cell synaptic function, and ensured near-normal hearing thresholds over time. Maximal hearing preservation was observed when Clrn2 was delivered prior to the loss of transducing stereocilia. Our findings demonstrate that gene therapy is effective for the treatment of post-lingual hearing impairment and age-related deafness associated with CLRN2 patient mutations.


Asunto(s)
Células Ciliadas Auditivas , Pérdida Auditiva , Humanos , Animales , Ratones , Células Ciliadas Auditivas/metabolismo , Audición , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Estereocilios/metabolismo , Suplementos Dietéticos
3.
Int J Mol Sci ; 24(14)2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37511622

RESUMEN

Hyperacusis, i.e., an increased sensitivity to sounds, is described in several neurodevelopmental disorders (NDDs), including Fragile X Syndrome (FXS). The mechanisms underlying hyperacusis in FXS are still largely unknown and effective therapies are lacking. Big conductance calcium-activated potassium (BKCa) channels were proposed as a therapeutic target to treat several behavioral disturbances in FXS preclinical models, but their role in mediating their auditory alterations was not specifically addressed. Furthermore, studies on the acoustic phenotypes of FXS animal models mostly focused on central rather than peripheral auditory pathways. Here, we provided an extensive characterization of the peripheral auditory phenotype of the Fmr1-knockout (KO) mouse model of FXS at adulthood. We also assessed whether the acute administration of Chlorzoxazone, a BKCa agonist, could rescue the auditory abnormalities of adult mutant mice. Fmr1-KO mice both at 3 and 6 months showed a hyperacusis-like startle phenotype with paradoxically reduced auditory brainstem responses associated with a loss of ribbon synapses in the inner hair cells (IHCs) compared to their wild-type (WT) littermates. BKCa expression was markedly reduced in the IHCs of KOs compared to WT mice, but only at 6 months, when Chlorzoxazone rescued mutant auditory dysfunction. Our findings highlight the age-dependent and progressive contribution of peripheral mechanisms and BKCa channels to adult hyperacusis in FXS, suggesting a novel therapeutic target to treat auditory dysfunction in NDDs.


Asunto(s)
Síndrome del Cromosoma X Frágil , Hiperacusia , Animales , Ratones , Vías Auditivas/metabolismo , Clorzoxazona , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio , Ratones Noqueados
4.
J Neurosci ; 39(18): 3394-3411, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30833506

RESUMEN

Transmitter release at auditory inner hair cell (IHC) ribbon synapses involves exocytosis of glutamatergic vesicles during voltage activation of L-type Cav1.3 calcium channels. At these synapses, the fast and indefatigable release of synaptic vesicles by IHCs is controlled by otoferlin, a six-C2-domain (C2-ABCDEF) protein that functions as a high-affinity Ca2+ sensor. The molecular events by which each otoferlin C2 domain contributes to the regulation of the synaptic vesicle cycle in IHCs are still incompletely understood. Here, we investigate their role using a cochlear viral cDNA transfer approach in vivo, where IHCs of mouse lacking otoferlin (Otof-/- mice of both sexes) were virally transduced with cDNAs of various mini-otoferlins. Using patch-clamp recordings and membrane capacitance measurements, we show that the viral transfer of mini-otoferlin containing C2-ACEF, C2-EF, or C2-DEF partially restores the fast exocytotic component in Otof-/- mouse IHCs. The restoration was much less efficient with C2-ACDF, underlining the importance of the C2-EF domain. None of the mini-otoferlins tested restored the sustained component of vesicle release, explaining the absence of hearing recovery. The restoration of the fast exocytotic component in the transduced Otof-/- IHCs was also associated with a recovery of Ca2+ currents with normal amplitude and fast time inactivation, confirming that the C-terminal C2 domains of otoferlin are essential for normal gating of Cav1.3 channels. Finally, the reintroduction of the mini-otoferlins C2-EF, C2-DEF, or C2-ACEF allowed us to uncover and characterize for the first time a dynamin-dependent ultrafast endocytosis in IHCs.SIGNIFICANCE STATEMENT Otoferlin, a large six-C2-domain protein, is essential for synaptic vesicle exocytosis at auditory hair cell ribbon synapses. Here, we show that the viral expression of truncated forms of otoferlin (C2-EF, C2-DEF, and C2-ACEF) can partially rescue the fast and transient release component of exocytosis in mouse hair cells lacking otoferlin, yet cannot sustain exocytosis after long repeated stimulation. Remarkably, these hair cells also display a dynamin-dependent ultrafast endocytosis. Overall, our study uncovers the pleiotropic role of otoferlin in the hair cell synaptic vesicle cycle, notably in triggering both ultrafast exocytosis and endocytosis and recruiting synaptic vesicles to the active zone.


Asunto(s)
Endocitosis , Exocitosis , Células Ciliadas Auditivas/fisiología , Proteínas de la Membrana/fisiología , Transmisión Sináptica , Estimulación Acústica , Adenoviridae/fisiología , Animales , Calcio/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Vectores Genéticos , Masculino , Proteínas de la Membrana/genética , Ratones Noqueados , Vesículas Sinápticas/fisiología
5.
Proc Natl Acad Sci U S A ; 114(36): 9695-9700, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28835534

RESUMEN

Our understanding of the mechanisms underlying inherited forms of inner ear deficits has considerably improved during the past 20 y, but we are still far from curative treatments. We investigated gene replacement as a strategy for restoring inner ear functions in a mouse model of Usher syndrome type 1G, characterized by congenital profound deafness and balance disorders. These mice lack the scaffold protein sans, which is involved both in the morphogenesis of the stereociliary bundle, the sensory antenna of inner ear hair cells, and in the mechanoelectrical transduction process. We show that a single delivery of the sans cDNA by the adenoassociated virus 8 to the inner ear of newborn mutant mice reestablishes the expression and targeting of the protein to the tips of stereocilia. The therapeutic gene restores the architecture and mechanosensitivity of stereociliary bundles, improves hearing thresholds, and durably rescues these mice from the balance defects. Our results open up new perspectives for efficient gene therapy of cochlear and vestibular disorders by showing that even severe dysmorphogenesis of stereociliary bundles can be corrected.


Asunto(s)
Síndromes de Usher/genética , Síndromes de Usher/terapia , Animales , Animales Recién Nacidos , ADN Complementario/administración & dosificación , ADN Complementario/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Terapia Genética/métodos , Vectores Genéticos , Células Ciliadas Auditivas/patología , Células Ciliadas Auditivas/fisiología , Humanos , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Síndromes de Usher/fisiopatología , Vestíbulo del Laberinto/patología , Vestíbulo del Laberinto/fisiopatología
6.
J Neurosci ; 37(11): 2960-2975, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28193694

RESUMEN

The mechanisms orchestrating transient and sustained exocytosis in auditory inner hair cells (IHCs) remain largely unknown. These exocytotic responses are believed to mobilize sequentially a readily releasable pool of vesicles (RRP) underneath the synaptic ribbons and a slowly releasable pool of vesicles (SRP) at farther distance from them. They are both governed by Cav1.3 channels and require otoferlin as Ca2+ sensor, but whether they use the same Cav1.3 isoforms is still unknown. Using whole-cell patch-clamp recordings in posthearing mice, we show that only a proportion (∼25%) of the total Ca2+ current in IHCs displaying fast inactivation and resistance to 20 µm nifedipine, a l-type Ca2+ channel blocker, is sufficient to trigger RRP but not SRP exocytosis. This Ca2+ current is likely conducted by short C-terminal isoforms of Cav1.3 channels, notably Cav1.342A and Cav1.343S, because their mRNA is highly expressed in wild-type IHCs but poorly expressed in Otof-/- IHCs, the latter having Ca2+ currents with considerably reduced inactivation. Nifedipine-resistant RRP exocytosis was poorly affected by 5 mm intracellular EGTA, suggesting that the Cav1.3 short isoforms are closely associated with the release site at the synaptic ribbons. Conversely, our results suggest that Cav1.3 long isoforms, which carry ∼75% of the total IHC Ca2+ current with slow inactivation and confer high sensitivity to nifedipine and to internal EGTA, are essentially involved in recruiting SRP vesicles. Intracellular Ca2+ imaging showed that Cav1.3 long isoforms support a deep intracellular diffusion of Ca2+SIGNIFICANCE STATEMENT Auditory inner hair cells (IHCs) encode sounds into nerve impulses through fast and indefatigable Ca2+-dependent exocytosis at their ribbon synapses. We show that this synaptic process involves long and short C-terminal isoforms of the Cav1.3 Ca2+ channel that differ in the kinetics of their Ca2+-dependent inactivation and their relative sensitivity to the l-type Ca2+ channel blocker nifedipine. The short C-terminal isoforms, having fast inactivation and low sensitivity to nifedipine, mainly control the fast fusion of the readily releasable pool (RRP); that is, they encode the phasic exocytotic component. The long isoforms, with slow inactivation and great sensitivity to nifedipine, mainly regulate the vesicular replenishment of the RRP; that is, the sustained or tonic exocytosis.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/fisiología , Exocitosis/fisiología , Células Ciliadas Auditivas Internas/fisiología , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/clasificación , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/metabolismo
7.
J Neurosci ; 34(33): 10853-69, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25122888

RESUMEN

The hair cell ribbon synapses of the mammalian auditory and vestibular systems differ greatly in their anatomical organization and firing properties. Notably, vestibular Type I hair cells (VHC-I) are surrounded by a single calyx-type afferent terminal that receives input from several ribbons, whereas cochlear inner hair cells (IHCs) are contacted by several individual afferent boutons, each facing a single ribbon. The specificity of the presynaptic molecular mechanisms regulating transmitter release at these different sensory ribbon synapses is not well understood. Here, we found that exocytosis during voltage activation of Ca(2+) channels displayed higher Ca(2+) sensitivity, 10 mV more negative half-maximum activation, and a smaller dynamic range in VHC-I than in IHCs. VHC-I had a larger number of Ca(2+) channels per ribbon (158 vs 110 in IHCs), but their Ca(2+) current density was twofold smaller because of a smaller open probability and unitary conductance. Using confocal and stimulated emission depletion immunofluorescence microscopy, we showed that VHC-I had fewer synaptic ribbons (7 vs 17 in IHCs) to which Cav1.3 channels are more tightly organized than in IHCs. Gradual intracellular Ca(2+) uncaging experiments revealed that exocytosis had a similar intrinsic Ca(2+) sensitivity in both VHC-I and IHCs (KD of 3.3 ± 0.6 µM and 4.0 ± 0.7 µM, respectively). In otoferlin-deficient mice, exocytosis was largely reduced in VHC-I and IHCs. We conclude that VHC-I and IHCs use a similar micromolar-sensitive otoferlin Ca(2+) sensor and that their sensory encoding specificity is essentially determined by a different functional organization of Ca(2+) channels at their synaptic ribbons.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Exocitosis/fisiología , Células Ciliadas Vestibulares/fisiología , Proteínas de la Membrana/metabolismo , Órgano Espiral/fisiología , Sinapsis/fisiología , Animales , Cóclea/metabolismo , Cóclea/fisiología , Células Ciliadas Vestibulares/metabolismo , Ratones , Órgano Espiral/metabolismo
8.
Front Cell Neurosci ; 17: 1197611, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37538852

RESUMEN

Humans have six members of the ferlin protein family: dysferlin, myoferlin, otoferlin, fer1L4, fer1L5, and fer1L6. These proteins share common features such as multiple Ca2+-binding C2 domains, FerA domains, and membrane anchoring through their single C-terminal transmembrane domain, and are believed to play a key role in calcium-triggered membrane fusion and vesicle trafficking. Otoferlin plays a crucial role in hearing and vestibular function. In this review, we will discuss how we see otoferlin working as a Ca2+-dependent mechanical sensor regulating synaptic vesicle fusion at the hair cell ribbon synapses. Although otoferlin is also present in the central nervous system, particularly in the cortex and amygdala, its role in brain tissues remains unknown. Mutations in the OTOF gene cause one of the most frequent genetic forms of congenital deafness, DFNB9. These mutations produce severe to profound hearing loss due to a defect in synaptic excitatory glutamatergic transmission between the inner hair cells and the nerve fibers of the auditory nerve. Gene therapy protocols that allow normal rescue expression of otoferlin in hair cells have just started and are currently in pre-clinical phase. In parallel, studies have linked ferlins to cancer through their effect on cell signaling and development, allowing tumors to form and cancer cells to adapt to a hostile environment. Modulation by mechanical forces and Ca2+ signaling are key determinants of the metastatic process. Although ferlins importance in cancer has not been extensively studied, data show that otoferlin expression is significantly associated with survival in specific cancer types, including clear cell and papillary cell renal carcinoma, and urothelial bladder cancer. These findings indicate a role for otoferlin in the carcinogenesis of these tumors, which requires further investigation to confirm and understand its exact role, particularly as it varies by tumor site. Targeting this protein may lead to new cancer therapies.

9.
Hear Res ; 438: 108880, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37666034

RESUMEN

Transtympanic administration is used clinically for the injection of gentamicin and/or corticosteroids. This atraumatic route is based on passive diffusion through the round window membrane (RWM). The main limitation of this method is related to the clearance through the Eustachian tube, making the concentration of the therapeutic agent at the intracochlear level uncertain and limited. Moreover, this technique remains unsuitable for molecules of high molecular weight or in the case of gene therapies. The purpose was to study a new technique of intracochlear administration in an atraumatic, direct and controlled manner by laser-assisted bioprinting (LAB). LAB was used to deliver dexamethasone phosphate with thermosensitive hydrogel on the mouse RWM. After validation of the regularity and homogeneity of the pattern, the diffusion in vivo of the dexamethasone into the perilymph after LAB has been confirmed by ELISA. Auditory function measurements showed no hearing impairment suggesting that bioprinting does not induce significant cochlear damage. Hence, the present proof of concept study introduces a promising approach for inner ear drug delivery.


Asunto(s)
Bioimpresión , Animales , Ratones , Cóclea , Difusión , Sistemas de Liberación de Medicamentos , Rayos Láser
10.
J Neurophysiol ; 108(11): 3116-23, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22972963

RESUMEN

During development, synaptic exocytosis by cochlear hair cells is first initiated by patterned spontaneous Ca(2+) spikes and, at the onset of hearing, by sound-driven graded depolarizing potentials. The molecular reorganization occurring in the hair cell synaptic machinery during this developmental transition still remains elusive. We characterized the changes in biophysical properties of voltage-gated Ca(2+) currents and exocytosis in developing auditory hair cells of a precocial animal, the domestic chick. We found that immature chick hair cells (embryonic days 10-12) use two types of Ca(2+) currents to control exocytosis: low-voltage-activating, rapidly inactivating (mibefradil sensitive) T-type Ca(2+) currents and high-voltage-activating, noninactivating (nifedipine sensitive) L-type currents. Exocytosis evoked by T-type Ca(2+) current displayed a fast release component (RRP) but lacked the slow sustained release component (SRP), suggesting an inefficient recruitment of distant synaptic vesicles by this transient Ca(2+) current. With maturation, the participation of L-type Ca(2+) currents to exocytosis largely increased, inducing a highly Ca(2+) efficient recruitment of an RRP and an SRP component. Notably, L-type-driven exocytosis in immature hair cells displayed higher Ca(2+) efficiency when triggered by prerecorded native action potentials than by voltage steps, whereas similar efficiency for both protocols was found in mature hair cells. This difference likely reflects a tighter coupling between release sites and Ca(2+) channels in mature hair cells. Overall, our results suggest that the temporal characteristics of Ca(2+) entry through T-type and L-type Ca(2+) channels greatly influence synaptic release by hair cells during cochlear development.


Asunto(s)
Potenciales de Acción , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo T/metabolismo , Calcio/metabolismo , Cóclea/embriología , Exocitosis , Células Ciliadas Auditivas/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Embrión de Pollo , Cóclea/citología , Células Ciliadas Auditivas/citología , Mibefradil/farmacología , Neurogénesis , Nifedipino/farmacología , Transmisión Sináptica , Vesículas Sinápticas
11.
iScience ; 25(12): 105628, 2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36483015

RESUMEN

Hearing depends on fast and sustained calcium-dependent synaptic vesicle fusion at the ribbon synapses of cochlear inner hair cells (IHCs). The implication of the canonical neuronal SNARE complex in this exocytotic process has so far remained controversial. We investigated the role of SNAP-25, a key component of this complex, in hearing, by generating and analyzing a conditional knockout mouse model allowing a targeted postnatal deletion of Snap-25 in IHCs. Mice subjected to IHC Snap-25 inactivation after hearing onset developed severe to profound deafness because of defective IHC exocytosis followed by ribbon degeneration and IHC loss. Viral transfer of Snap-25 in these mutant mice rescued their hearing function by restoring IHC exocytosis and preventing synapses and hair cells from degeneration. These results demonstrate that SNAP-25 is essential for normal hearing function, most likely by ensuring IHC exocytosis and ribbon synapse maintenance.

12.
J Neurosci ; 30(40): 13281-90, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20926654

RESUMEN

In pre-hearing mice, vesicle exocytosis at cochlear inner hair cell (IHC) ribbon synapses is triggered by spontaneous Ca(2+) spikes. At the onset of hearing, IHC exocytosis is then exclusively driven by graded potentials, and is characterized by higher Ca(2+) efficiency and improved synchronization of vesicular release. The molecular players involved in this transition are still unknown. Here we addressed the involvement of synaptotagmins and otoferlin as putative Ca(2+) sensors in IHC exocytosis during postnatal maturation of the cochlea. Using cell capacitance measurements, we showed that Ca(2+)-evoked exocytosis in mouse IHCs switches from an otoferlin-independent to an otoferlin-dependent mechanism at postnatal day 4. During this early exocytotic period, several synaptotagmins (Syts), including Syt1, Syt2 and Syt7, were detected in IHCs. The exocytotic response as well as the release of the readily releasable vesicle pool (RRP) was, however, unchanged in newborn mutant mice lacking Syt1, Syt2 or Syt7 (Syt1(-/-), Syt2(-/-) and Syt7(-/-) mice). We only found a defect in RRP recovery in Syt1(-/-) mice which was apparent as a strongly reduced response to repetitive stimulations. In post-hearing Syt2(-/-) and Syt7(-/-) mutant mice, IHC synaptic exocytosis was unaffected. The transient expression of Syt1 and Syt2, which were no longer detected in IHCs after the onset of hearing, indicates that these two most common Ca(2+)-sensors in CNS synapses are not involved in mature IHCs. We suggest that otoferlin underlies highly efficient Ca(2+)-dependent membrane-membrane fusion, a process likely essential to increase the probability and synchrony of vesicle fusion events at the mature IHC ribbon synapse.


Asunto(s)
Cóclea/crecimiento & desarrollo , Exocitosis , Células Ciliadas Auditivas Internas/fisiología , Proteínas de la Membrana/fisiología , Sinaptotagmina II/fisiología , Sinaptotagmina I/fisiología , Animales , Calcio/fisiología , Señalización del Calcio/genética , Senescencia Celular/genética , Senescencia Celular/fisiología , Cóclea/citología , Capacidad Eléctrica , Exocitosis/genética , Femenino , Células Ciliadas Auditivas Internas/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Sinapsis/genética , Sinapsis/fisiología , Transmisión Sináptica/genética , Sinaptotagmina I/genética , Sinaptotagmina II/genética
13.
Front Aging Neurosci ; 13: 756449, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733152

RESUMEN

Age-related hidden hearing loss is often described as a cochlear synaptopathy that results from a progressive degeneration of the inner hair cell (IHC) ribbon synapses. The functional changes occurring at these synapses during aging are not fully understood. Here, we characterized this aging process in IHCs of C57BL/6J mice, a strain which is known to carry a cadherin-23 mutation and experiences early hearing loss with age. These mice, while displaying a large increase in auditory brainstem thresholds due to 50% loss of IHC synaptic ribbons at middle age (postnatal day 365), paradoxically showed enhanced acoustic startle reflex suggesting a hyperacusis-like response. The auditory defect was associated with a large shrinkage of the IHCs' cell body and a drastic enlargement of their remaining presynaptic ribbons which were facing enlarged postsynaptic AMPAR clusters. Presynaptic Ca2+ microdomains and the capacity of IHCs to sustain high rates of exocytosis were largely increased, while on the contrary the expression of the fast-repolarizing BK channels, known to negatively control transmitter release, was decreased. This age-related synaptic plasticity in IHCs suggested a functional potentiation of synaptic transmission at the surviving synapses, a process that could partially compensate the decrease in synapse number and underlie hyperacusis.

14.
J Neurosci ; 29(34): 10474-87, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19710301

RESUMEN

Otoferlin, a C2-domain-containing Ca(2+) binding protein, is required for synaptic exocytosis in auditory hair cells. However, its exact role remains essentially unknown. Intriguingly enough, no balance defect has been observed in otoferlin-deficient (Otof(-/-)) mice. Here, we show that the vestibular nerve compound action potentials evoked during transient linear acceleration ramps in Otof(-/-) mice display higher threshold, lower amplitude, and increased latency compared with wild-type mice. Using patch-clamp capacitance measurement in intact utricles, we show that type I and type II hair cells display a remarkable linear transfer function between Ca(2+) entry, flowing through voltage-activated Ca(2+) channels, and exocytosis. This linear Ca(2+) dependence was observed when changing the Ca(2+) channel open probability or the Ca(2+) flux per channel during various test potentials. In Otof(-/-) hair cells, exocytosis displays slower kinetics, reduced Ca(2+) sensitivity, and nonlinear Ca(2+) dependence, despite morphologically normal synapses and normal Ca(2+) currents. We conclude that otoferlin is essential for a high-affinity Ca(2+) sensor function that allows efficient and linear encoding of low-intensity stimuli at the vestibular hair cell synapse.


Asunto(s)
Calcio/metabolismo , Exocitosis/fisiología , Células Ciliadas Vestibulares/citología , Proteínas de la Membrana/fisiología , Sinapsis/fisiología , Aceleración , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Biofisica , Bloqueadores de los Canales de Calcio/farmacología , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Exocitosis/efectos de los fármacos , Exocitosis/genética , Células Ciliadas Vestibulares/clasificación , Células Ciliadas Vestibulares/efectos de los fármacos , Células Ciliadas Vestibulares/fisiología , Modelos Lineales , Proteínas de la Membrana/deficiencia , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Miosina VIIa , Miosinas/metabolismo , Técnicas de Placa-Clamp/métodos , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Receptores AMPA/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/genética , Sinapsis/ultraestructura , Tetrodotoxina/farmacología , Nervio Vestibular/fisiología
15.
Purinergic Signal ; 6(2): 201-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20806012

RESUMEN

Purinergic signaling in the mammalian cochleovestibular hair cells and afferent neurons is reviewed. The scope includes P2 and P1 receptors in the inner hair cells (IHCs) of the cochlea, the type I spiral ganglion neurons (SGNs) that convey auditory signals from IHCs, the vestibular hair cells (VHCs) in the vestibular end organs (macula in the otolith organs and crista in the semicircular canals), and the vestibular ganglion neurons (VGNs) that transmit postural and rotatory information from VHCs. Various subtypes of P2X ionotropic receptors are expressed in IHCs as well as P2Y metabotropic receptors that mobilize intracellular calcium. Their functional roles still remain speculative, but adenosine 5'-triphosphate (ATP) could regulate the spontaneous activity of the hair cells during development and the receptor potentials of mature hair cells during sound stimulation. In SGNs, P2Y metabotropic receptors activate a nonspecific cation conductance that is permeable to large cations as NMDG(+) and TEA(+). Remarkably, this depolarizing nonspecific conductance in SGNs can also be activated by other metabotropic processes evoked by acetylcholine and tachykinin. The molecular nature and the role of this depolarizing channel are unknown, but its electrophysiological properties suggest that it could lie within the transient receptor potential channel family and could regulate the firing properties of the afferent neurons. Studies on the vestibular partition (VHC and VGN) are sparse but have also shown the expression of P2X and P2Y receptors. There is still little evidence of functional P1 (adenosine) receptors in the afferent system of the inner ear.

16.
J Neurosci ; 28(8): 1798-803, 2008 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-18287496

RESUMEN

Immature cochlear outer hair cells (OHCs) make transient synaptic contacts (ribbon synapses) with type I afferent nerve fibers, but direct evidence of synaptic vesicle exocytosis is still missing. We thus investigated calcium-dependent exocytosis in murine OHCs at postnatal day 2 (P2)-P3, a developmental stage when calcium current maximum amplitude was the highest. By using time-resolved patch-clamp capacitance measurements, we show that voltage step activation of L-type calcium channels triggers fast membrane capacitance increase. Capacitance increase displayed two kinetic components, which are likely to reflect two functionally distinct pools of synaptic vesicles, a readily releasable pool (RRP; tau = 79 ms) and a slowly releasable pool (tau = 870 ms). The RRP size and maximal release rate were estimated at approximately 1200 vesicles and approximately 15,000 vesicles/s, respectively. In addition, we found a linear relationship between capacitance increase and calcium influx, like in mature inner hair cells (IHCs). These results give strong support to the existence of efficient calcium-dependent neurotransmitter release in immature OHCs. Moreover, we show that immature OHCs, just like immature IHCs, are able to produce regenerative calcium-dependent action potentials that could trigger synaptic exocytosis in vivo. Finally, the evoked membrane capacitance increases were abolished in P2-P3 OHCs from mutant Otof-/- mice defective for otoferlin, despite normal calcium currents. We conclude that otoferlin, the putative major calcium sensor at IHC ribbon synapses, is essential to synaptic exocytosis in immature OHCs too.


Asunto(s)
Calcio/fisiología , Exocitosis/fisiología , Células Ciliadas Auditivas Externas/fisiología , Proteínas de la Membrana/fisiología , Células Madre/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Exocitosis/efectos de los fármacos , Células Ciliadas Auditivas Externas/química , Células Ciliadas Auditivas Externas/citología , Proteínas de la Membrana/análisis , Ratones , Ratones Mutantes , Células Madre/química , Células Madre/citología
17.
Int J Dev Neurosci ; 27(1): 87-96, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18832023

RESUMEN

The small protein otospiralin has initially been identified as an inner ear specific molecule. However, compelling evidence from high throughput sequencing projects suggested that otospiralin is likely expressed in the central nervous system. Here, we tested this hypothesis using a combination of molecular biology, immunological, and histological techniques, and found that otospiralin is expressed in numerous regions of the central nervous system in mouse. In situ hybridization and immunohistochemistry revealed that otospiralin is widely expressed in neuronal cell bodies and glia. Ultrastructural observations in the cerebral cortex located the small protein in close proximity to membranous organelles in perikarya, the inner face of post-synaptic neuronal membranes, and in astrocytic processes. These results are in agreement with the predicted structure of the protein which revealed a single N-terminal transmembrane helix domain followed by a C-terminus cytosolic tail. Interestingly, 2 weeks after a mechanical trauma in the cerebral cortex, otospiralin expression increased in reactive astrocytes located within the vicinity of the site of injury, but not in neurons. Collectively, our observations suggest that otospiralin is possibly involved in signaling pathways, and could play a role in repair mechanisms subsequent to an injury in the central nervous system.


Asunto(s)
Encéfalo/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Proteínas/metabolismo , Animales , Encéfalo/citología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Gliosis/etiología , Gliosis/metabolismo , Gliosis/fisiopatología , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Regeneración Nerviosa/fisiología , Neuroglía/citología , Neuronas/citología , Orgánulos/metabolismo , Orgánulos/ultraestructura , Estructura Terciaria de Proteína/fisiología , Proteínas/genética , Membranas Sinápticas/metabolismo , Membranas Sinápticas/ultraestructura
18.
EMBO Mol Med ; 11(9): e10288, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31448880

RESUMEN

Hearing relies on mechanically gated ion channels present in the actin-rich stereocilia bundles at the apical surface of cochlear hair cells. Our knowledge of the mechanisms underlying the formation and maintenance of the sound-receptive structure is limited. Utilizing a large-scale forward genetic screen in mice, genome mapping and gene complementation tests, we identified Clrn2 as a new deafness gene. The Clrn2clarinet/clarinet mice (p.Trp4* mutation) exhibit a progressive, early-onset hearing loss, with no overt retinal deficits. Utilizing data from the UK Biobank study, we could show that CLRN2 is involved in human non-syndromic progressive hearing loss. Our in-depth morphological, molecular and functional investigations establish that while it is not required for initial formation of cochlear sensory hair cell stereocilia bundles, clarin-2 is critical for maintaining normal bundle integrity and functioning. In the differentiating hair bundles, lack of clarin-2 leads to loss of mechano-electrical transduction, followed by selective progressive loss of the transducing stereocilia. Together, our findings demonstrate a key role for clarin-2 in mammalian hearing, providing insights into the interplay between mechano-electrical transduction and stereocilia maintenance.


Asunto(s)
Pérdida Auditiva/metabolismo , Estereocilios/metabolismo , Adulto , Anciano , Animales , Estudios de Cohortes , Femenino , Células Ciliadas Auditivas/metabolismo , Audición , Pérdida Auditiva/genética , Pérdida Auditiva/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estereocilios/genética
19.
Int J Dev Neurosci ; 26(7): 723-32, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18682288

RESUMEN

Rho GTPases proteins are essential for cytoskeletal reorganization and play important roles in the development of neuronal dendrites and axons. Several studies have implicated two members of the Rho GTPase family Rho-A and Rac1 activities in the neuronal polarization and the formation of axons and dendrites. In order to correlate cellular expressions of Rho-A and Rac1 with neuronal polarity (axons versus dendrite formation) in the central nervous system, the cerebellum and immunochemical techniques have been chosen. In the adult cerebellar cortex differential pattern of distribution between Rho-A and Rac1 was observed. While Rac1 expression was restricted to Purkinje cell (somata, dendrites and axons), Rho-A was ubiquitously distributed within the cerebellar cortex. Rac1 was localized in the Purkinje cell dendritic arborization (largest and tiny dendrites) and in their axons. This pattern of distribution was also observed during the postnatal development and followed the dendritic morphogenesis of Purkinje cell. Rho-A was highly expressed in the adult Purkinje cells somata, in cells of the granular layer, in glia within the white matter and in axons. Intense staining was observed in Bergmann glia cell bodies and processes. In the developing cerebellum, Rho-A was highly present in cells of the external and internal granule layers and in the Purkinje cell layer. Bergmann glia cell bodies and processes had the most intense staining during the development. The present study reveals a high expression of Rac1 and Rho-A during Purkinje cell neurites outgrowth period which occurred after birth in the cerebellum. In addition Rho-A is highly expressed in granule cell progenitor cells present in the external granular layer and therefore may play an important role in granule cell progenitor migration.


Asunto(s)
Cerebelo/enzimología , Cerebelo/crecimiento & desarrollo , Neuritas/metabolismo , Neurogénesis/fisiología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Corteza Cerebelosa/citología , Corteza Cerebelosa/enzimología , Corteza Cerebelosa/crecimiento & desarrollo , Cerebelo/citología , Dendritas/enzimología , Dendritas/ultraestructura , Regulación del Desarrollo de la Expresión Génica/fisiología , Gerbillinae , Conos de Crecimiento/enzimología , Conos de Crecimiento/ultraestructura , Fibras Nerviosas Mielínicas/enzimología , Fibras Nerviosas Mielínicas/ultraestructura , Neuritas/ultraestructura , Neuroglía/citología , Neuroglía/enzimología , Células de Purkinje/citología , Células de Purkinje/enzimología , Células Madre/citología , Células Madre/enzimología
20.
Audiol Neurootol ; 13(1): 65-70, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17890859

RESUMEN

Streptococcus pneumoniae can induce local and systemic diseases such as meningitis, otitis media, and pneumonia. One third of these meningitis cases can be associated with irreversible sensorineural hearing loss whose mechanisms likely involves the exotoxin pneumolysin (PLY) that irreversibly damages cochlear hair cells (HCs). In the respiratory system and in neuron it has been demonstrated that zinc deficiency increases severity and mortality of such infections in animal models and in children. Moreover, zinc supplementation can decrease the severity of pneumococcal respiratory infections. The aim of our study was to assess the potential protective effect of zinc against PLY toxicity on HCs in culture. Our results showed that in the presence of zinc at concentration as low as 1 microM, the toxicity of PLY was largely reduced by about 50% for both inner and outer HCs. At 300 microM of zinc, protection significantly increased with 62 and 55.2% for IHCs and OHCs, respectively. Our results suggest that the protective effect of zinc is likely due to an inhibition of the toxin incorporation and aggregation into the plasma membrane, thus preventing calcium influx through the toxin pores. Our findings raise the possibility that treatments with zinc may help to prevent debilitating otological sequelae from pneumococcal infection.


Asunto(s)
Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/patología , Estreptolisinas/toxicidad , Oligoelementos/farmacología , Zinc/farmacología , Animales , Proteínas Bacterianas/toxicidad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Pérdida Auditiva/microbiología , Pérdida Auditiva/prevención & control , Técnicas de Cultivo de Órganos , Infecciones Neumocócicas/complicaciones , Ratas , Ratas Wistar , Streptococcus pneumoniae
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA