Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 606
Filtrar
1.
Annu Rev Neurosci ; 42: 67-86, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-30699050

RESUMEN

The genetic approach, based on the study of inherited forms of deafness, has proven to be particularly effective for deciphering the molecular mechanisms underlying the development of the peripheral auditory system, the cochlea and its afferent auditory neurons, and how this system extracts the physical parameters of sound. Although this genetic dissection has provided little information about the central auditory system, scattered data suggest that some genes may have a critical role in both the peripheral and central auditory systems. Here, we review the genes controlling the development and function of the peripheral and central auditory systems, focusing on those with demonstrated intrinsic roles in both systems and highlighting the current underappreciation of these genes. Their encoded products are diverse, from transcription factors to ion channels, as are their roles in the central auditory system, mostly evaluated in brainstem nuclei. We examine the ontogenetic and evolutionary mechanisms that may underlie their expression at different sites.


Asunto(s)
Vías Auditivas/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes , Neurogénesis/genética , Animales , Vías Auditivas/crecimiento & desarrollo , Evolución Biológica , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Cóclea/fisiología , Ontología de Genes , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/fisiología , Trastornos de la Audición/genética , Humanos , Canales Iónicos/genética , Canales Iónicos/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Rombencéfalo/embriología , Rombencéfalo/crecimiento & desarrollo , Rombencéfalo/fisiología , Células Receptoras Sensoriales/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
2.
Development ; 151(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39120083

RESUMEN

In multicellular tissues, the size and shape of cells are intricately linked with their physiological functions. In the vertebrate auditory organ, the neurosensory epithelium develops as a mosaic of sensory hair cells (HCs), and their glial-like supporting cells, which have distinct morphologies and functional properties at different frequency positions along its tonotopic long axis. In the chick cochlea, the basilar papilla (BP), proximal (high-frequency) HCs, are larger than their distal (low-frequency) counterparts, a morphological feature essential for sound perception. Mitochondrial dynamics, which constitute the equilibrium between fusion and fission, regulate differentiation and functional refinement across a variety of cell types. We investigate this as a potential mechanism for regulating the shape of developing HCs. Using live imaging in intact BP explants, we identify distinct remodelling of mitochondrial networks in proximal compared with distal HCs. Manipulating mitochondrial dynamics in developing HCs alters their normal morphology along the proximal-distal (tonotopic) axis. Inhibition of the mitochondrial fusion machinery decreased proximal HC surface area, whereas promotion of fusion increased the distal HC surface area. We identify mitochondrial dynamics as a key regulator of HC morphology in developing inner ear epithelia.


Asunto(s)
Cóclea , Células Ciliadas Auditivas , Mitocondrias , Dinámicas Mitocondriales , Animales , Cóclea/embriología , Cóclea/citología , Cóclea/crecimiento & desarrollo , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Mitocondrias/metabolismo , Embrión de Pollo , Forma de la Célula , Pollos , Diferenciación Celular
3.
Proc Natl Acad Sci U S A ; 121(35): e2405217121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39172791

RESUMEN

Intercellular signaling mediated by evolutionarily conserved planar cell polarity (PCP) proteins aligns cell polarity along the tissue plane and drives polarized cell behaviors during tissue morphogenesis. Accumulating evidence indicates that the vertebrate PCP pathway is regulated by noncanonical, ß-catenin-independent Wnt signaling; however, the signaling components and mechanisms are incompletely understood. In the mouse hearing organ, both PCP and noncanonical Wnt (ncWnt) signaling are required in the developing auditory sensory epithelium to control cochlear duct elongation and planar polarity of resident sensory hair cells (HCs), including the shape and orientation of the stereociliary hair bundle essential for sound detection. We have recently discovered a Wnt/G-protein/PI3K pathway that coordinates HC planar polarity and intercellular PCP signaling. Here, we identify Wnt7b as a ncWnt ligand acting in concert with Wnt5a to promote tissue elongation in diverse developmental processes. In the cochlea, Wnt5a and Wnt7b are redundantly required for cochlear duct coiling and elongation, HC planar polarity, and asymmetric localization of core PCP proteins Fzd6 and Dvl2. Mechanistically, Wnt5a/Wnt7b-mediated ncWnt signaling promotes membrane recruitment of Daple, a nonreceptor guanine nucleotide exchange factor for Gαi, and activates PI3K/AKT and ERK signaling, which promote asymmetric Fzd6 localization. Thus, ncWnt and PCP signaling pathways have distinct mutant phenotypes and signaling components, suggesting that they act as separate, parallel pathways with nonoverlapping functions in cochlear morphogenesis. NcWnt signaling drives tissue elongation and reinforces intercellular PCP signaling by regulating the trafficking of PCP-specific Frizzled receptors.


Asunto(s)
Polaridad Celular , Proteínas Wnt , Vía de Señalización Wnt , Proteína Wnt-5a , Animales , Polaridad Celular/fisiología , Proteínas Wnt/metabolismo , Proteínas Wnt/genética , Proteína Wnt-5a/metabolismo , Proteína Wnt-5a/genética , Ratones , Vía de Señalización Wnt/fisiología , Cóclea/metabolismo , Cóclea/citología , Cóclea/crecimiento & desarrollo , Células Ciliadas Auditivas/metabolismo , Receptores Frizzled/metabolismo , Receptores Frizzled/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Morfogénesis
4.
Dev Dyn ; 253(8): 771-780, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38264972

RESUMEN

The sensory epithelium of the cochlea, the organ of Corti, has complex cytoarchitecture consisting of mechanosensory hair cells intercalated by epithelial support cells. The support cells provide important trophic and structural support to the hair cells. Thus, the support cells must be stiff yet compliant enough to withstand and modulate vibrations to the hair cells. Once the sensory cells are properly patterned, the support cells undergo significant remodeling from a simple epithelium into a structurally rigid epithelium with fluid-filled spaces in the murine cochlea. Cell adhesion molecules such as cadherins are necessary for sorting and connecting cells in an intact epithelium. To create the fluid-filled spaces, cell adhesion properties of adjoining cell membranes between cells must change to allow the formation of spaces within an epithelium. However, the dynamic localization of cadherins has not been properly analyzed as these spaces are formed. There are three cadherins that are reported to be expressed during the first postnatal week of development when the tunnel of Corti forms in the cochlea. In this study, we characterize the dynamic localization of cadherins that are associated with cytoskeletal remodeling at the contacting membranes of the inner and outer pillar cells flanking the tunnel of Corti.


Asunto(s)
Cadherinas , Cóclea , Animales , Cadherinas/metabolismo , Ratones , Epitelio/metabolismo , Cóclea/metabolismo , Cóclea/crecimiento & desarrollo , Cóclea/citología , Órgano Espiral/metabolismo , Órgano Espiral/citología , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/citología , Adhesión Celular/fisiología
5.
Development ; 148(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34061174

RESUMEN

During embryonic development, the otic epithelium and surrounding periotic mesenchymal cells originate from distinct lineages and coordinate to form the mammalian cochlea. Epithelial sensory precursors within the cochlear duct first undergo terminal mitosis before differentiating into sensory and non-sensory cells. In parallel, periotic mesenchymal cells differentiate to shape the lateral wall, modiolus and pericochlear spaces. Previously, Wnt activation was shown to promote proliferation and differentiation of both otic epithelial and mesenchymal cells. Here, we fate-mapped Wnt-responsive epithelial and mesenchymal cells in mice and found that Wnt activation resulted in opposing cell fates. In the post-mitotic cochlear epithelium, Wnt activation via ß-catenin stabilization induced clusters of proliferative cells that dedifferentiated and lost epithelial characteristics. In contrast, Wnt-activated periotic mesenchyme formed ectopic pericochlear spaces and cell clusters showing a loss of mesenchymal and gain of epithelial features. Finally, clonal analyses via multi-colored fate-mapping showed that Wnt-activated epithelial cells proliferated and formed clonal colonies, whereas Wnt-activated mesenchymal cells assembled as aggregates of mitotically quiescent cells. Together, we show that Wnt activation drives transition between epithelial and mesenchymal states in a cell type-dependent manner.


Asunto(s)
Cóclea/embriología , Epitelio/metabolismo , Células Madre Mesenquimatosas/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Desdiferenciación Celular , Diferenciación Celular , Proliferación Celular , Cóclea/citología , Cóclea/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Proteínas Wnt , beta Catenina/metabolismo
6.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32571852

RESUMEN

The cochlea, a coiled structure located in the ventral region of the inner ear, acts as the primary structure for the perception of sound. Along the length of the cochlear spiral is the organ of Corti, a highly derived and rigorously patterned sensory epithelium that acts to convert auditory stimuli into neural impulses. The development of the organ of Corti requires a series of inductive events that specify unique cellular characteristics and axial identities along its three major axes. Here, we review recent studies of the cellular and molecular processes regulating several aspects of cochlear development, such as axial patterning, cochlear outgrowth and cellular differentiation. We highlight how the precise coordination of multiple signaling pathways is required for the successful formation of a complete organ of Corti.


Asunto(s)
Cóclea/crecimiento & desarrollo , Animales , Percepción Auditiva , Diferenciación Celular , Cóclea/anatomía & histología , Cóclea/metabolismo , Células Ciliadas Auditivas/metabolismo , Mitosis , Órgano Espiral/anatomía & histología , Órgano Espiral/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal
7.
J Neurosci ; 41(4): 594-612, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33303678

RESUMEN

Spontaneous bursts of electrical activity in the developing auditory system arise within the cochlea before hearing onset and propagate through future sound-processing circuits of the brain to promote maturation of auditory neurons. Studies in isolated cochleae revealed that this intrinsically generated activity is initiated by ATP release from inner supporting cells (ISCs), resulting in activation of purinergic autoreceptors, K+ efflux, and subsequent depolarization of inner hair cells. However, it is unknown when this activity emerges or whether different mechanisms induce activity during distinct stages of development. Here we show that spontaneous electrical activity in mouse cochlea from both sexes emerges within ISCs during the late embryonic period, preceding the onset of spontaneous correlated activity in inner hair cells and spiral ganglion neurons, which begins at birth and follows a base to apex developmental gradient. At all developmental ages, pharmacological inhibition of P2Y1 purinergic receptors dramatically reduced spontaneous activity in these three cell types. Moreover, in vivo imaging within the inferior colliculus revealed that auditory neurons within future isofrequency zones exhibit coordinated neural activity at birth. The frequency of these discrete bursts increased progressively during the postnatal prehearing period yet remained dependent on P2RY1. Analysis of mice with disrupted cholinergic signaling in the cochlea indicate that this efferent input modulates, rather than initiates, spontaneous activity before hearing onset. Thus, the auditory system uses a consistent mechanism involving ATP release from ISCs and activation of P2RY1 autoreceptors to elicit coordinated excitation of neurons that will process similar frequencies of sound.SIGNIFICANCE STATEMENT In developing sensory systems, groups of neurons that will process information from similar sensory space exhibit highly correlated electrical activity that is critical for proper maturation and circuit refinement. Defining the period when this activity is present, the mechanisms responsible and the features of this activity are crucial for understanding how spontaneous activity influences circuit development. We show that, from birth to hearing onset, the auditory system relies on a consistent mechanism to elicit correlate firing of neurons that will process similar frequencies of sound. Targeted disruption of this activity will increase our understanding of how these early circuits mature and may provide insight into processes responsible for developmental disorders of the auditory system.


Asunto(s)
Vías Auditivas/crecimiento & desarrollo , Vías Auditivas/fisiología , Receptores Purinérgicos/fisiología , Adenosina Trifosfato/metabolismo , Animales , Señalización del Calcio/fisiología , Cóclea/crecimiento & desarrollo , Cóclea/fisiología , Femenino , Células Ciliadas Auditivas/fisiología , Células Ciliadas Auditivas Internas/fisiología , Colículos Inferiores/fisiología , Células Laberínticas de Soporte/fisiología , Masculino , Ratones , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/fisiología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/fisiología , Retina/fisiología , Ganglio Espiral de la Cóclea/fisiología
8.
Proc Natl Acad Sci U S A ; 116(13): 6415-6424, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30867284

RESUMEN

Ribbon synapses of cochlear inner hair cells (IHCs) undergo molecular assembly and extensive functional and structural maturation before hearing onset. Here, we characterized the nanostructure of IHC synapses from late prenatal mouse embryo stages (embryonic days 14-18) into adulthood [postnatal day (P)48] using electron microscopy and tomography as well as optical nanoscopy of apical turn organs of Corti. We find that synaptic ribbon precursors arrive at presynaptic active zones (AZs) after afferent contacts have been established. These ribbon precursors contain the proteins RIBEYE and piccolino, tether synaptic vesicles and their delivery likely involves active, microtubule-based transport pathways. Synaptic contacts undergo a maturational transformation from multiple small to one single, large AZ. This maturation is characterized by the fusion of ribbon precursors with membrane-anchored ribbons that also appear to fuse with each other. Such fusion events are most frequently encountered around P12 and hence, coincide with hearing onset in mice. Thus, these events likely underlie the morphological and functional maturation of the AZ. Moreover, the postsynaptic densities appear to undergo a similar refinement alongside presynaptic maturation. Blockwise addition of ribbon material by fusion as found during AZ maturation might represent a general mechanism for modulating ribbon size.


Asunto(s)
Cóclea/crecimiento & desarrollo , Células Ciliadas Auditivas Internas/fisiología , Células Ciliadas Vestibulares/fisiología , Sinapsis/fisiología , Animales , Cóclea/ultraestructura , Células Ciliadas Auditivas Internas/ultraestructura , Células Ciliadas Vestibulares/ultraestructura , Audición/fisiología , Ratones/embriología , Microscopía Electrónica , Modelos Animales , Sinapsis/ultraestructura , Vesículas Sinápticas , Tomografía
9.
J Neurosci ; 40(49): 9401-9413, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33127852

RESUMEN

During cochlear development, the Notch ligand JAGGED 1 (JAG1) plays an important role in the specification of the prosensory region, which gives rise to sound-sensing hair cells and neighboring supporting cells (SCs). While JAG1's expression is maintained in SCs through adulthood, the function of JAG1 in SC development is unknown. Here, we demonstrate that JAG1 is essential for the formation and maintenance of Hensen's cells, a highly specialized SC subtype located at the edge of the auditory epithelium. Using Sox2CreERT2/+::Jag1loxP/loxP mice of both genders, we show that Jag1 deletion at the onset of differentiation, at embryonic day 14.5, disrupted Hensen's cell formation. Similar loss of Hensen's cells was observed when Jag1 was deleted after Hensen's cell formation at postnatal day (P) 0/P1 and fate-mapping analysis revealed that in the absence of Jag1, some Hensen's cells die, but others convert into neighboring Claudius cells. In support of a role for JAG1 in cell survival, genes involved in mitochondrial function and protein synthesis were downregulated in the sensory epithelium of P0 cochlea lacking Jag1 Finally, using Fgfr3-iCreERT2 ::Jag1loxP/loxP mice to delete Jag1 at P0, we observed a similar loss of Hensen's cells and found that adult Jag1 mutant mice have hearing deficits at the low-frequency range.SIGNIFICANCE STATEMENT Hensen's cells play an essential role in the development and homeostasis of the cochlea. Defects in the biophysical or functional properties of Hensen's cells have been linked to auditory dysfunction and hearing loss. Despite their importance, surprisingly little is known about the molecular mechanisms that guide their development. Morphologic and fate-mapping analyses in our study revealed that, in the absence of the Notch ligand JAGGED1, Hensen's cells died or converted into Claudius cells, which are specialized epithelium-like cells outside the sensory epithelium. Confirming a link between JAGGED1 and cell survival, transcriptional profiling showed that JAGGED1 maintains genes critical for mitochondrial function and tissue homeostasis. Finally, auditory phenotyping revealed that JAGGED1's function in supporting cells is necessary for low-frequency hearing.


Asunto(s)
Cóclea/metabolismo , Proteína Jagged-1/metabolismo , Células Laberínticas de Soporte/fisiología , Animales , Supervivencia Celular , Cóclea/citología , Cóclea/crecimiento & desarrollo , Regulación hacia Abajo , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Proteína Jagged-1/genética , Masculino , Ratones , Ratones Noqueados , Embarazo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
10.
Development ; 145(24)2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30504125

RESUMEN

Morphogenesis of the inner ear epithelium requires coordinated deployment of several signaling pathways, and disruptions cause abnormalities of hearing and/or balance. The FGFR2b ligands FGF3 and FGF10 are expressed throughout otic development and are required individually for normal morphogenesis, but their prior and redundant roles in otic placode induction complicates investigation of subsequent combinatorial functions in morphogenesis. To interrogate these roles and identify new effectors of FGF3 and FGF10 signaling at the earliest stages of otic morphogenesis, we used conditional gene ablation after otic placode induction, and temporal inhibition of signaling with a secreted, dominant-negative FGFR2b ectodomain. We show that both ligands are required continuously after otocyst formation for maintenance of otic neuroblasts and for patterning and proliferation of the epithelium, leading to normal morphogenesis of both the cochlear and vestibular domains. Furthermore, the first genome-wide identification of proximal targets of FGFR2b signaling in the early otocyst reveals novel candidate genes for inner ear development and function.


Asunto(s)
Oído Interno/crecimiento & desarrollo , Oído Interno/metabolismo , Morfogénesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Linaje de la Célula , Proliferación Celular , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Doxiciclina/farmacología , Femenino , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 3 de Crecimiento de Fibroblastos/metabolismo , Ganglión/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrasas/metabolismo , Ligandos , Masculino , Ratones , Mutación/genética , Neuronas/citología , Neuronas/metabolismo , Factor de Transcripción PAX2/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Vestíbulo del Laberinto/crecimiento & desarrollo , Vestíbulo del Laberinto/metabolismo
11.
J Neurosci Res ; 99(2): 699-728, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33181864

RESUMEN

Neuronal diversity in the cochlea is largely determined by ion channels. Among voltage-gated channels, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels open with hyperpolarization and depolarize the cell until the resting membrane potential. The functions for hearing are not well elucidated and knowledge about localization is controversial. We created a detailed map of subcellular location and co-expression of all four HCN subunits across different mammalian species including CBA/J, C57Bl/6N, Ly5.1 mice, guinea pigs, cats, and human subjects. We correlated age-related hearing deterioration in CBA/J and C57Bl/6N with expression levels of HCN1, -2, and -4 in individual auditory neurons from the same cohort. Spatiotemporal expression during murine postnatal development exposed HCN2 and HCN4 involvement in a critical phase of hair cell innervation. The huge diversity of subunit composition, but lack of relevant heteromeric pairing along the perisomatic membrane and axon initial segments, highlighted an active role for auditory neurons. Neuron clusters were found to be the hot spots of HCN1, -2, and -4 immunostaining. HCN channels were also located in afferent and efferent fibers of the sensory epithelium. Age-related changes on HCN subtype expression were not uniform among mice and could not be directly correlated with audiometric data. The oldest mice groups revealed HCN channel up- or downregulation, depending on the mouse strain. The unexpected involvement of HCN channels in outer hair cell function where HCN3 overlaps prestin location emphasized the importance for auditory function. A better understanding may open up new possibilities to tune neuronal responses evoked through electrical stimulation by cochlear implants.


Asunto(s)
Envejecimiento/metabolismo , Cóclea/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Neuronas/metabolismo , Canales de Potasio/fisiología , Animales , Gatos , Cóclea/crecimiento & desarrollo , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Regulación de la Expresión Génica , Cobayas , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/biosíntesis , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Neuronas/ultraestructura , Canales de Potasio/biosíntesis , Canales de Potasio/genética , Fracciones Subcelulares/metabolismo
12.
Neural Plast ; 2021: 5511010, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34306061

RESUMEN

As a general sensory disorder, hearing loss was a major concern worldwide. Autophagy is a common cellular reaction to stress that degrades cytoplasmic waste through the lysosome pathway. Autophagy not only plays major roles in maintaining intracellular homeostasis but is also involved in the development and pathogenesis of many diseases. In the auditory system, several studies revealed the link between autophagy and hearing protection. In this review, we aimed to establish the correlation between autophagy and hair cells (HCs) from the aspects of ototoxic drugs, aging, and acoustic trauma and discussed whether autophagy could serve as a potential measure in the protection of HCs.


Asunto(s)
Autofagia , Pérdida Auditiva Sensorineural/prevención & control , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Autofagia/efectos de los fármacos , Cisplatino/toxicidad , Cóclea/irrigación sanguínea , Cóclea/crecimiento & desarrollo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Pérdida Auditiva Provocada por Ruido , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Isquemia/fisiopatología , Ratones , Ratones Noqueados , MicroARNs/genética , Estrés Oxidativo , Resveratrol/uso terapéutico , Privación de Sueño/complicaciones
13.
J Neurosci ; 39(18): 3360-3375, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30755493

RESUMEN

In the mature mammalian cochlea, inner hair cells (IHCs) are mainly innervated by afferent fibers that convey sound information to the CNS. During postnatal development, however, medial olivocochlear (MOC) efferent fibers transiently innervate the IHCs. The MOC-IHC synapse, functional from postnatal day 0 (P0) to hearing onset (P12), undergoes dramatic changes in the sensitivity to acetylcholine (ACh) and in the expression of key postsynaptic proteins. To evaluate whether there are associated changes in the properties of ACh release during this period, we used a cochlear preparation from mice of either sex at P4, P6-P7, and P9-P11 and monitored transmitter release from MOC terminals in voltage-clamped IHCs in the whole-cell configuration. The quantum content increased 5.6× from P4 to P9-P11 due to increases in the size and replenishment rate of the readily releasable pool of synaptic vesicles without changes in their probability of release or quantum size. This strengthening in transmission was accompanied by changes in short-term plasticity properties, which switched from facilitation at P4 to depression at P9-P11. We have previously shown that at P9-P11, ACh release is supported by P/Q- and N-type voltage-gated calcium channels (VGCCs) and negatively regulated by BK potassium channels activated by Ca2+ influx through L-type VGCCs. We now show that at P4 and P6-P7, release is mediated by P/Q-, R- and L-type VGCCs. Interestingly, L-type VGCCs have a dual role: they both support release and fuel BK channels, suggesting that at immature stages presynaptic proteins involved in release are less compartmentalized.SIGNIFICANCE STATEMENT During postnatal development before the onset of hearing, cochlear inner hair cells (IHCs) present spontaneous Ca2+ action potentials that release glutamate at the first auditory synapse in the absence of sound stimulation. The IHC Ca2+ action potential frequency pattern, which is crucial for the correct establishment and function of the auditory system, is regulated by the efferent medial olivocochlear (MOC) system that transiently innervates IHCs during this period. We show here that developmental changes in synaptic strength and synaptic plasticity properties at the MOC-IHC synapse upon MOC fiber activation at different frequencies might be crucial for tightly shaping the pattern of afferent activity during this critical period.


Asunto(s)
Cóclea/crecimiento & desarrollo , Células Ciliadas Auditivas Internas/fisiología , Sinapsis/fisiología , Transmisión Sináptica , Acetilcolina/metabolismo , Animales , Cóclea/metabolismo , Femenino , Potenciales Postsinápticos Inhibidores , Masculino , Ratones Endogámicos BALB C , Plasticidad Neuronal
14.
J Neurosci ; 39(36): 7037-7048, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31217330

RESUMEN

The auditory system in many mammals is immature at birth but precisely organized in adults. Spontaneous activity in the inner ear plays a critical role in guiding this maturation process. This is shaped by an efferent pathway that descends from the brainstem and makes transient direct synaptic contacts with inner hair cells. In this work, we used an α9 cholinergic nicotinic receptor knock-in mouse model (of either sex) with enhanced medial efferent activity (Chrna9L9'T, L9'T) to further understand the role of the olivocochlear system in the correct establishment of auditory circuits. Wave III of auditory brainstem responses (which represents synchronized activity of synapses within the superior olivary complex) was smaller in L9'T mice, suggesting a central dysfunction. The mechanism underlying this functional alteration was analyzed in brain slices containing the medial nucleus of the trapezoid body (MNTB), where neurons are topographically organized along a mediolateral (ML) axis. The topographic organization of MNTB physiological properties observed in wildtype (WT) was abolished in L9'T mice. Additionally, electrophysiological recordings in slices indicated MNTB synaptic alterations. In vivo multielectrode recordings showed that the overall level of MNTB activity was reduced in the L9'T The present results indicate that the transient cochlear efferent innervation to inner hair cells during the critical period before the onset of hearing is involved in the refinement of topographic maps as well as in setting the properties of synaptic transmission at a central auditory nucleus.SIGNIFICANCE STATEMENT Cochlear inner hair cells of altricial mammals display spontaneous electrical activity before hearing onset. The pattern and firing rate of these cells are crucial for the correct maturation of the central auditory pathway. A descending efferent innervation from the CNS contacts the hair cells during this developmental window. The present work shows that genetic enhancement of efferent function disrupts the orderly topographic distribution of biophysical and synaptic properties in the auditory brainstem and causes severe synaptic dysfunction. This work adds to the notion that the transient efferent innervation to the cochlea is necessary for the correct establishment of the central auditory circuitry.


Asunto(s)
Cóclea/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico , Núcleo Olivar/fisiología , Potenciales Sinápticos , Cuerpo Trapezoide/fisiología , Animales , Percepción Auditiva , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Femenino , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/fisiología , Masculino , Ratones , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Núcleo Olivar/crecimiento & desarrollo , Núcleo Olivar/metabolismo , Receptores Nicotínicos/genética , Cuerpo Trapezoide/crecimiento & desarrollo , Cuerpo Trapezoide/metabolismo
15.
J Neurosci ; 39(41): 8013-8023, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31462532

RESUMEN

Type II spiral ganglion neurons provide afferent innervation to outer hair cells of the cochlea and are proposed to have nociceptive functions important for auditory function and homeostasis. These neurons are anatomically distinct from other classes of spiral ganglion neurons because they extend a peripheral axon beyond the inner hair cells that subsequently makes a distinct 90 degree turn toward the cochlear base. As a result, patterns of outer hair cell innervation are coordinated with the tonotopic organization of the cochlea. Previously, it was shown that peripheral axon turning is directed by a nonautonomous function of the core planar cell polarity (PCP) protein VANGL2. We demonstrate using mice of either sex that Fzd3 and Fzd6 similarly regulate axon turning, are functionally redundant with each other, and that Fzd3 genetically interacts with Vangl2 to guide this process. FZD3 and FZD6 proteins are asymmetrically distributed along the basolateral wall of cochlear-supporting cells, and are required to promote or maintain the asymmetric distribution of VANGL2 and CELSR1. These data indicate that intact PCP complexes formed between cochlear-supporting cells are required for the nonautonomous regulation of axon pathfinding. Consistent with this, in the absence of PCP signaling, peripheral axons turn randomly and often project toward the cochlear apex. Additional analyses of Porcn mutants in which WNT secretion is reduced suggest that noncanonical WNT signaling establishes or maintains PCP signaling in this context. A deeper understanding of these mechanisms is necessary for repairing auditory circuits following acoustic trauma or promoting cochlear reinnervation during regeneration-based deafness therapies.SIGNIFICANCE STATEMENT Planar cell polarity (PCP) signaling has emerged as a complementary mechanism to classical axon guidance in regulating axon track formation, axon outgrowth, and neuronal polarization. The core PCP proteins are also required for auditory circuit assembly, and coordinate hair cell innervation with the tonotopic organization of the cochlea. This is a non-cell-autonomous mechanism that requires the formation of PCP protein complexes between cochlear-supporting cells located along the trajectory of growth cone navigation. These findings are significant because they demonstrate how the fidelity of auditory circuit formation is ensured during development, and provide a mechanism by which PCP proteins may regulate axon outgrowth and guidance in the CNS.


Asunto(s)
Cóclea/inervación , Receptores Frizzled/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Ganglio Espiral de la Cóclea/citología , Aciltransferasas/genética , Animales , Axones/fisiología , Axones/ultraestructura , Polaridad Celular , Cóclea/crecimiento & desarrollo , Femenino , Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Externas , Masculino , Proteínas de la Membrana/genética , Ratones , Mutación/genética , Órgano Espiral/crecimiento & desarrollo , Órgano Espiral/fisiología , Receptores Acoplados a Proteínas G/fisiología , Ganglio Espiral de la Cóclea/crecimiento & desarrollo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología
16.
Annu Rev Neurosci ; 35: 509-28, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22715884

RESUMEN

Cochlear inner hair cells (IHCs), the mammalian auditory sensory cells, encode acoustic signals with high fidelity by Graded variations of their membrane potential trigger rapid and sustained vesicle exocytosis at their ribbon synapses. The kinetics of glutamate release allows proper transfer of sound information to the primary afferent auditory neurons. Understanding the physiological properties and underlying molecular mechanisms of the IHC synaptic machinery, and especially its high temporal acuity, which is pivotal to speech perception, is a central issue of auditory science. During the past decade, substantial progress in high-resolution imaging and electrophysiological recordings, as well as the development of genetic approaches both in humans and in mice, has produced major insights regarding the morphological, physiological, and molecular characteristics of this synapse. Here we review this recent knowledge and discuss how it enlightens the way the IHC ribbon synapse develops and functions.


Asunto(s)
Células Ciliadas Auditivas Internas/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Cóclea/fisiología , Exocitosis/fisiología , Ácido Glutámico/metabolismo , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/metabolismo , Audición/fisiología , Humanos , Modelos Neurológicos , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Sinapsis/metabolismo , Sinapsis/ultraestructura , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/fisiología
17.
FASEB J ; 33(5): 5942-5956, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30753104

RESUMEN

Betaine-homocysteine S-methyltransferases (BHMTs) are methionine cycle enzymes that remethylate homocysteine; hence, their malfunction leads to hyperhomocysteinemia. Epidemiologic and experimental studies have revealed a correlation between hyperhomocysteinemia and hearing loss. Here, we have studied the expression of methionine cycle genes in the mouse cochlea and the impact of knocking out the Bhmt gene in the auditory receptor. We evaluated age-related changes in mouse hearing by recording auditory brainstem responses before and following exposure to noise. Also, we measured cochlear cytoarchitecture, gene expression by RNA-arrays and quantitative RT-PCR, and metabolite levels in liver and plasma by HPLC. Our results indicate that there is an age-dependent strain-specific expression of methionine cycle genes in the mouse cochlea and a further regulation during the response to noise damage. Loss of Bhmt did not cause an evident impact in the hearing acuity of young mice, but it produced higher threshold shifts and poorer recovery following noise challenge. Hearing loss was associated with increased cochlear injury, outer hair cell loss, altered expression of cochlear methionine cycle genes, and hyperhomocysteinemia. Our results suggest that BHMT plays a central role in the homeostasis of cochlear methionine metabolism and that Bhmt2 up-regulation could carry out a compensatory role in cochlear protection against noise injury in the absence of BHMT.-Partearroyo, T., Murillo-Cuesta, S., Vallecillo, N., Bermúdez-Muñoz, J. M., Rodríguez-de la Rosa, L., Mandruzzato, G., Celaya, A. M., Zeisel, S. H., Pajares, M. A., Varela-Moreiras, G., Varela-Nieto, I. Betaine-homocysteine S-methyltransferase deficiency causes increased susceptibility to noise-induced hearing loss associated with plasma hyperhomocysteinemia.


Asunto(s)
Betaína-Homocisteína S-Metiltransferasa/fisiología , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Pérdida Auditiva Provocada por Ruido/sangre , Homocisteína/sangre , Hiperhomocisteinemia/sangre , Animales , Apoptosis , Betaína-Homocisteína S-Metiltransferasa/genética , Cromatografía Líquida de Alta Presión , Femenino , Perfilación de la Expresión Génica , Genotipo , Audición , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pronóstico , Factores de Tiempo
18.
PLoS Genet ; 13(9): e1006967, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28892484

RESUMEN

The organ of Corti in the cochlea is a two-cell layered epithelium: one cell layer of mechanosensory hair cells that align into one row of inner and three rows of outer hair cells interdigitated with one cell layer of underlying supporting cells along the entire length of the cochlear spiral. These two types of epithelial cells are derived from common precursors in the four- to five-cell layered primordium and acquire functionally important shapes during terminal differentiation through the thinning process and convergent extension. Here, we have examined the role of Six1 in the establishment of the auditory sensory epithelium. Our data show that prior to terminal differentiation of the precursor cells, deletion of Six1 leads to formation of only a few hair cells and defective patterning of the sensory epithelium. Previous studies have suggested that downregulation of Sox2 expression in differentiating hair cells must occur after Atoh1 mRNA activation in order to allow Atoh1 protein accumulation due to antagonistic effects between Atoh1 and Sox2. Our analysis indicates that downregulation of Sox2 in the differentiating hair cells depends on Six1 activity. Furthermore, we found that Six1 is required for the maintenance of Fgf8 expression and dynamic distribution of N-cadherin and E-cadherin in the organ of Corti during differentiation. Together, our analyses uncover essential roles of Six1 in hair cell differentiation and formation of the organ of Corti in the mammalian cochlea.


Asunto(s)
Diferenciación Celular/genética , Células Ciliadas Auditivas/metabolismo , Proteínas de Homeodominio/genética , Órgano Espiral/crecimiento & desarrollo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cadherinas/genética , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Factor 8 de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Ratones , Morfogénesis/genética , Órgano Espiral/metabolismo , Factores de Transcripción SOXB1/genética
19.
Int J Mol Sci ; 21(4)2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32070057

RESUMEN

In mammals Homer1, Homer2 and Homer3 constitute a family of scaffolding proteins with key roles in Ca2+ signaling and Ca2+ transport. In rodents, Homer proteins and mRNAs have been shown to be expressed in various postnatal tissues and to be enriched in brain. However, whether the Homers are expressed in developing tissues is hitherto largely unknown. In this work, we used immunohistochemistry and in situ hybridization to analyze the expression patterns of Homer1, Homer2 and Homer3 in developing cephalic structures. Our study revealed that the three Homer proteins and their encoding genes are expressed in a wide range of developing tissues and organs, including the brain, tooth, eye, cochlea, salivary glands, olfactory and respiratory mucosae, bone and taste buds. We show that although overall the three Homers exhibit overlapping distribution patterns, the proteins localize at distinct subcellular domains in several cell types, that in both undifferentiated and differentiated cells Homer proteins are concentrated in puncta and that the vascular endothelium is enriched with Homer3 mRNA and protein. Our findings suggest that Homer proteins may have differential and overlapping functions and are expected to be of value for future research aiming at deciphering the roles of Homer proteins during embryonic development.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Andamiaje Homer/genética , Animales , Encéfalo/crecimiento & desarrollo , Diferenciación Celular/genética , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Glándulas Salivales/crecimiento & desarrollo , Glándulas Salivales/metabolismo , Transducción de Señal/genética , Diente/crecimiento & desarrollo , Diente/metabolismo
20.
J Neurosci ; 38(25): 5677-5687, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29789373

RESUMEN

Until postnatal day (P) 12, inner hair cells of the rat cochlea are invested with both afferent and efferent synaptic connections. With the onset of hearing at P12, the efferent synapses disappear, and afferent (ribbon) synapses operate with greater efficiency. This change coincides with increased expression of voltage-gated potassium channels, the loss of calcium-dependent electrogenesis, and the onset of graded receptor potentials driven by sound. The transient efferent synapses include near-membrane postsynaptic cisterns thought to regulate calcium influx through the hair cell's α9-containing and α10-containing nicotinic acetylcholine receptors. This influx activates small-conductance Ca2+-activated K+ (SK) channels. Serial-section electron microscopy of inner hair cells from two 9-d-old (male) rat pups revealed many postsynaptic efferent cisterns and presynaptic afferent ribbons whose average minimal separation in five cells ranged from 1.1 to 1.7 µm. Efferent synaptic function was studied in rat pups (age, 7-9 d) of either sex. The duration of these SK channel-mediated IPSCs was increased by enhanced calcium influx through L-type voltage-gated channels, combined with ryanodine-sensitive release from internal stores-presumably the near-membrane postsynaptic cistern. These data support the possibility that inner hair cell calcium electrogenesis modulates the efficacy of efferent inhibition during the maturation of inner hair cell synapses.SIGNIFICANCE STATEMENT Strict calcium buffering is essential for cellular function. This problem is especially acute for compact hair cells where increasing cytoplasmic calcium promotes the opposing functions of closely adjoining afferent and efferent synapses. The near-membrane postsynaptic cistern at efferent synapses segregates synaptic calcium signals by acting as a dynamic calcium store. The hair cell serves as an informative model for synapses with postsynaptic cisterns (C synapses) found in central neurons.


Asunto(s)
Señalización del Calcio/fisiología , Cóclea/inervación , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/fisiología , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Canales de Calcio Tipo L/metabolismo , Cóclea/citología , Cóclea/crecimiento & desarrollo , Femenino , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda