Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Development ; 2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39373109

RESUMO

Notch signaling patterns the cochlear organ of Corti, and patients with the JAG1/NOTCH2-related genetic disorder Alagille syndrome can thus experience hearing loss. We investigated the function of Jag1 in cochlear patterning and signaling using Jag1Ndr/Ndr mice, a model of Alagille syndrome. Jag1Ndr/Ndr mice exhibited expected vestibular and auditory deficits, a dose-dependent increase in ectopic inner hair cells, and a reduction in outer hair cells. Single cell RNA sequencing of the organ of Corti demonstrated a global dysregulation of genes associated with inner ear development and deafness. Analysis of individual cell types further revealed that Jag1 represses Notch activation in lateral supporting cells and demonstrated a function for Jag1 in gene regulation and development of outer hair cells. Surprisingly, ectopic "outer hair cell-like" cells were present in the medial compartment and pillar cell region of Jag1Ndr/Ndr cochleae, yet they exhibited location-dependent expression of the inner hair cell fate-determinant Tbx2, suggesting Jag1 is required for Tbx2 to drive inner hair cell commitment. This study thus identifies new roles for Jag1 in supporting cells, and in outer hair cell specification and positioning.

2.
Cell Mol Life Sci ; 81(1): 80, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38334784

RESUMO

Dominant optic atrophy (DOA) is one of the most prevalent forms of hereditary optic neuropathies and is mainly caused by heterozygous variants in OPA1, encoding a mitochondrial dynamin-related large GTPase. The clinical spectrum of DOA has been extended to a wide variety of syndromic presentations, called DOAplus, including deafness as the main secondary symptom associated to vision impairment. To date, the pathophysiological mechanisms underlying the deafness in DOA remain unknown. To gain insights into the process leading to hearing impairment, we have analyzed the Opa1delTTAG mouse model that recapitulates the DOAplus syndrome through complementary approaches combining morpho-physiology, biochemistry, and cellular and molecular biology. We found that Opa1delTTAG mutation leads an adult-onset progressive auditory neuropathy in mice, as attested by the auditory brainstem response threshold shift over time. However, the mutant mice harbored larger otoacoustic emissions in comparison to wild-type littermates, whereas the endocochlear potential, which is a proxy for the functional state of the stria vascularis, was comparable between both genotypes. Ultrastructural examination of the mutant mice revealed a selective loss of sensory inner hair cells, together with a progressive degeneration of the axons and myelin sheaths of the afferent terminals of the spiral ganglion neurons, supporting an auditory neuropathy spectrum disorder (ANSD). Molecular assessment of cochlea demonstrated a reduction of Opa1 mRNA level by greater than 40%, supporting haploinsufficiency as the disease mechanism. In addition, we evidenced an early increase in Sirtuin 3 level and in Beclin1 activity, and subsequently an age-related mtDNA depletion, increased oxidative stress, mitophagy as well as an impaired autophagic flux. Together, these results support a novel role for OPA1 in the maintenance of inner hair cells and auditory neural structures, addressing new challenges for the exploration and treatment of OPA1-linked ANSD in patients.


Assuntos
Surdez , Perda Auditiva Central , Atrofia Óptica Autossômica Dominante , Animais , Humanos , Camundongos , GTP Fosfo-Hidrolases/genética , Perda Auditiva Central/genética , Mutação , Atrofia Óptica Autossômica Dominante/genética
3.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34686590

RESUMO

Mammalian hearing depends on an amplification process involving prestin, a voltage-sensitive motor protein that enables cochlear outer hair cells (OHCs) to change length and generate force. However, it has been questioned whether this prestin-based somatic electromotility can operate fast enough in vivo to amplify cochlear vibrations at the high frequencies that mammals hear. In this study, we measured sound-evoked vibrations from within the living mouse cochlea and found that the top and bottom of the OHCs move in opposite directions at frequencies exceeding 20 kHz, consistent with fast somatic length changes. These motions are physiologically vulnerable, depend on prestin, and dominate the cochlea's vibratory response to high-frequency sound. This dominance was observed despite mechanisms that clearly low-pass filter the in vivo electromotile response. Low-pass filtering therefore does not critically limit the OHC's ability to move the organ of Corti on a cycle-by-cycle basis. Our data argue that electromotility serves as the primary high-frequency amplifying mechanism within the mammalian cochlea.


Assuntos
Células Ciliadas Auditivas Externas/fisiologia , Órgão Espiral/fisiologia , Estimulação Acústica , Animais , Cóclea/fisiologia , Eletrofisiologia , Feminino , Audição/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Mutantes , Modelos Biológicos , Proteínas Motores Moleculares/deficiência , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/fisiologia , Movimento/fisiologia , Dinâmica não Linear , Som , Tomografia de Coerência Óptica , Vibração
4.
Int J Mol Sci ; 25(13)2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-39000390

RESUMO

The motor protein prestin, found in the inner ear's outer hair cells (OHCs), is responsible for high sensitivity and sharp frequency selectivity in mammalian hearing. Some studies have suggested that prestin could be a serological biomarker for cochlear damage, as OHCs are highly vulnerable to damage from various sources. However, the reported data are inconsistent and lack appropriate negative controls. To investigate whether prestin can be used as a serological biomarker for cochlear damage or stress, we measured prestin quantities in the bloodstreams of mice using ELISA kits from different companies. Wildtype (WT) mice were exposed to different ototoxic treatments, including noise exposure and ototoxic reagents that rapidly kill OHCs. Prestin-knockout (KO) mice were used as a negative control. Our data show that some ELISA kits were not able to detect prestin specifically. The ELISA kit that could detect the prestin protein from cochlear homogenates failed to detect prestin in the bloodstream, despite there being significant damage to OHCs in the cochleae. Furthermore, the optical densities of the serum samples, which correlate to prestin quantities, were significantly influenced by hemolysis in the samples. In conclusion, Prestin from OHCs is not a sensitive and reliable serological biomarker for detecting cochlear damage in mice using ELISA.


Assuntos
Biomarcadores , Células Ciliadas Auditivas Externas , Proteínas Motores Moleculares , Animais , Biomarcadores/sangue , Camundongos , Células Ciliadas Auditivas Externas/patologia , Células Ciliadas Auditivas Externas/metabolismo , Proteínas Motores Moleculares/metabolismo , Proteínas Motores Moleculares/genética , Camundongos Knockout , Cóclea/patologia , Cóclea/metabolismo , Ensaio de Imunoadsorção Enzimática , Camundongos Endogâmicos C57BL
5.
J Neurosci ; 42(44): 8361-8372, 2022 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-36123119

RESUMO

The outer hair cells in the mammalian cochlea are cellular actuators essential for sensitive hearing. The geometry and stiffness of the structural scaffold surrounding the outer hair cells will determine how the active cells shape mammalian hearing by modulating the organ of Corti (OoC) vibrations. Specifically, the tectorial membrane and the Deiters cell are mechanically in series with the hair bundle and soma, respectively, of the outer hair cell. Their mechanical properties and anatomic arrangement must determine the relative motion among different OoC structures. We measured the OoC mechanics in the cochleas acutely excised from young gerbils of both sexes at a resolution fine enough to distinguish the displacement of individual cells. A three-dimensional finite element model of fully deformable OoC was exploited to analyze the measured data in detail. As a means to verify the computer model, the basilar membrane deformations because of static and dynamic stimulations were measured and simulated. Two stiffness ratios have been identified that are critical to understand cochlear physics, which are the stiffness of the tectorial membrane with respect to the hair bundle and the stiffness of the Deiters cell with respect to the outer hair cell body. Our measurements suggest that the Deiters cells act like a mechanical equalizer so that the outer hair cells are constrained neither too rigidly nor too weakly.SIGNIFICANCE STATEMENT Mammals can detect faint sounds thanks to the action of mammalian-specific receptor cells called the outer hair cells. It is getting clearer that understanding the interactions between the outer hair cells and their surrounding structures such as the tectorial membrane and the Deiters cell is critical to resolve standing debates. Depending on theories, the stiffness of those two structures ranges from negligible to rigid. Because of their perceived importance, their properties have been measured in previous studies. However, nearly all existing data were obtained ex situ (after they were detached from the outer hair cells), which obscures their interaction with the outer hair cells. We quantified the mechanical properties of the tectorial membrane and the Deiters cell in situ.


Assuntos
Células Ciliadas Auditivas Externas , Células Ciliadas Vestibulares , Masculino , Animais , Feminino , Órgão Espiral , Membrana Basilar , Membrana Tectorial , Cóclea , Gerbillinae
6.
J Neurosci ; 42(42): 7875-7884, 2022 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-36261265

RESUMO

Cochlear amplification enables the enormous dynamic range of hearing through amplifying cochlear responses to low- to moderate-level sounds and compressing them to loud sounds. Amplification is attributed to voltage-dependent electromotility of mechanosensory outer hair cells (OHCs) driven by changing voltages developed across their cell membranes. At low frequencies, these voltage changes are dominated by intracellular receptor potentials (RPs). However, OHC membranes have electrical low-pass filter properties that attenuate high-frequency RPs, which should potentially attenuate amplification of high-frequency cochlear responses and impede high-frequency hearing. We made in vivo intracellular and extracellular electrophysiological measurements from the organ of Corti of male and female mice of the CBA/J strain, with excellent high-frequency hearing, and from the CD-1 mouse strain, which has sensitive hearing below 12 kHz but loses high-frequency hearing within a few weeks postpartum. The CD-1 mouse strain was transfected with an A88V mutation of the connexin 30 gap-junction protein. By blocking the action of the GJ protein to reduce input resistance, the mutation increased the OHC extracellular RP (ERP) magnitude and rescued high-frequency hearing. However, by increasing the organ of Corti resistance, the mutation rescued high-frequency hearing through preserving the OHC extracellular RP (ERP) magnitude. We measured the voltage developed across the basolateral membranes of OHCs, which controls their electromotility, for low- to high-frequency sounds in male and female mice of the CD-1 strain that expressed the A88V mutation. We demonstrate that ERPs, not RPs, drive OHC motility and cochlear amplification at high frequencies because at high frequencies, ERPs are not frequency attenuated, exceed RPs in magnitude, and are appropriately timed to provide cochlear amplification.SIGNIFICANCE STATEMENT Cochlear amplification, which enables the enormous dynamic range of hearing, is attributed to voltage-dependent electromotility of the mechanosensory outer hair cells (OHCs) driven by sound-induced voltage changes across their membranes. OHC intracellular receptor potentials are electrically low-pass filtered, which should hinder high-frequency hearing. We measured the intracellular and extracellular voltages that control OHC electromotility in vivo in a mouse strain with impaired high-frequency hearing. A gap-junction mutation of the strain rescued high-frequency hearing, increased organ of Corti resistance, and preserved large OHC extracellular receptor potentials but reduced OHC intracellular receptor potentials and impaired low-frequency hearing. We concluded intracellular potentials drive OHC motility at low frequencies and extracellular receptor potentials drive OHC motility and cochlear amplification at high frequencies.


Assuntos
Cóclea , Células Ciliadas Auditivas Externas , Animais , Feminino , Masculino , Camundongos , Cóclea/fisiologia , Conexina 30/genética , Conexina 30/metabolismo , Células Ciliadas Auditivas Externas/fisiologia , Camundongos Endogâmicos CBA , Mutação/genética , Junções Comunicantes
7.
J Physiol ; 601(19): 4291-4308, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37642186

RESUMO

Cochlear outer hair cells (OHCs) are responsible for the exquisite frequency selectivity and sensitivity of mammalian hearing. During development, the maturation of OHC afferent connectivity is refined by coordinated spontaneous Ca2+ activity in both sensory and non-sensory cells. Calcium signalling in neonatal OHCs can be modulated by oncomodulin (OCM, ß-parvalbumin), an EF-hand calcium-binding protein. Here, we investigated whether OCM regulates OHC spontaneous Ca2+ activity and afferent connectivity during development. Using a genetically encoded Ca2+ sensor (GCaMP6s) expressed in OHCs in wild-type (Ocm+/+ ) and Ocm knockout (Ocm-/- ) littermates, we found increased spontaneous Ca2+ activity and upregulation of purinergic receptors in OHCs from Ocm-/- cochlea immediately following birth. The afferent synaptic maturation of OHCs was delayed in the absence of OCM, leading to an increased number of ribbon synapses and afferent fibres on Ocm-/- OHCs before hearing onset. We propose that OCM regulates the spontaneous Ca2+ signalling in the developing cochlea and the maturation of OHC afferent innervation. KEY POINTS: Cochlear outer hair cells (OHCs) exhibit spontaneous Ca2+ activity during a narrow period of neonatal development. OHC afferent maturation and connectivity requires spontaneous Ca2+ activity. Oncomodulin (OCM, ß-parvalbumin), an EF-hand calcium-binding protein, modulates Ca2+ signals in immature OHCs. Using transgenic mice that endogenously expressed a Ca2+ sensor, GCaMP6s, we found increased spontaneous Ca2+ activity and upregulated purinergic receptors in Ocm-/- OHCs. The maturation of afferent synapses in Ocm-/- OHCs was also delayed, leading to an upregulation of ribbon synapses and afferent fibres in Ocm-/- OHCs before hearing onset. We propose that OCM plays an important role in modulating Ca2+ activity, expression of Ca2+ channels and afferent innervation in developing OHCs.


Assuntos
Cálcio , Células Ciliadas Auditivas Externas , Camundongos , Animais , Células Ciliadas Auditivas Externas/fisiologia , Cálcio/metabolismo , Parvalbuminas/metabolismo , Cóclea/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Camundongos Transgênicos , Receptores Purinérgicos/metabolismo , Mamíferos/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(20): 11109-11117, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32358189

RESUMO

Outer hair cells (OHCs) play an essential role in hearing by acting as a nonlinear amplifier which helps the cochlea detect sounds with high sensitivity and accuracy. This nonlinear sound processing generates distortion products, which can be measured as distortion-product otoacoustic emissions (DPOAEs). The OHC stereocilia that respond to sound vibrations are connected by three kinds of extracellular links: tip links that connect the taller stereocilia to shorter ones and convey force to the mechanoelectrical transduction channels, tectorial membrane-attachment crowns (TM-ACs) that connect the tallest stereocilia to one another and to the overlying TM, and horizontal top connectors (HTCs) that link adjacent stereocilia. While the tip links have been extensively studied, the roles that the other two types of links play in hearing are much less clear, largely because of a lack of suitable animal models. Here, while analyzing genetic combinations of tubby mice, we encountered models missing both HTCs and TM-ACs or HTCs alone. We found that the tubby mutation causes loss of both HTCs and TM-ACs due to a mislocalization of stereocilin, which results in OHC dysfunction leading to severe hearing loss. Intriguingly, the addition of the modifier allele modifier of tubby hearing 1 in tubby mice selectively rescues the TM-ACs but not the HTCs. Hearing is significantly rescued in these mice with robust DPOAE production, indicating an essential role of the TM-ACs but not the HTCs in normal OHC function. In contrast, the HTCs are required for the resistance of hearing to damage caused by noise stress.


Assuntos
Células Ciliadas Auditivas Externas/fisiologia , Ruído , Emissões Otoacústicas Espontâneas/fisiologia , Som , Estimulação Acústica , Animais , Células Ciliadas Auditivas Externas/citologia , Perda Auditiva , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Modelos Animais , Emissões Otoacústicas Espontâneas/genética , Estereocílios/fisiologia , Membrana Tectorial
9.
J Neurosci ; 41(32): 6812-6821, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34253627

RESUMO

For normal cochlear function, outer hair cells (OHCs) require a precise control of intracellular Ca2+ levels. In the absence of regulatory elements such as proteinaceous buffers or extrusion pumps, OHCs degenerate, leading to profound hearing impairment. Influx of Ca2+ occurs both at the stereocilia tips and the basolateral membrane. In this latter compartment, two different origins for Ca2+ influx have been poorly explored: voltage-gated L-type Ca2+ channels (VGCCs) at synapses with Type II afferent neurons, and α9α10 cholinergic nicotinic receptors at synapses with medio-olivochlear complex (MOC) neurons. Using functional imaging in mouse OHCs, we dissected Ca2+ influx individually through each of these sources, either by applying step depolarizations to activate VGCC, or stimulating MOC axons. Ca2+ ions originated in MOC synapses, but not by VGCC activation, was confined by Ca2+-ATPases most likely present in nearby synaptic cisterns. Although Ca2+ currents in OHCs are small, VGCC Ca2+ signals were comparable in size to those elicited by α9α10 receptors, and were potentiated by ryanodine receptors (RyRs). In contrast, no evidence of potentiation by RyRs was found for MOC Ca2+ signals over a wide range of presynaptic stimulation strengths. Our study shows that despite the fact that these two Ca2+ entry sites are closely positioned, they differ in their regulation by intracellular cisterns and/or organelles, suggesting the existence of well-tuned mechanisms to separate the two different OHC synaptic functions.SIGNIFICANCE STATEMENT Outer hair cells (OHCs) are sensory cells in the inner ear operating under very special constraints. Acoustic stimulation leads to fast changes both in membrane potential and in the intracellular concentration of metabolites such as Ca2+ Tight mechanisms for Ca2+ control in OHCs have been reported. Interestingly, Ca2+ is crucial for two important synaptic processes: inhibition by efferent cholinergic neurons, and glutamate release onto Type II afferent fibers. In the current study we functionally imaged Ca2+ at these two different synapses, showing close positioning within the basolateral compartment of OHCs. In addition, we show differential regulation of these two Ca2+ sources by synaptic cisterns and/or organelles, which could result crucial for functional segregation during normal hearing.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/fisiologia , Sinapses/fisiologia , Animais , Canais de Cálcio/fisiologia , Feminino , Masculino , Camundongos
10.
J Neurophysiol ; 127(1): 313-327, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34907797

RESUMO

It is critical for hearing that the descending cochlear efferent system provides a negative feedback to hair cells to regulate hearing sensitivity and protect hearing from noise. The medial olivocochlear (MOC) efferent nerves project to outer hair cells (OHCs) to regulate OHC electromotility, which is an active cochlear amplifier and can increase hearing sensitivity. Here, we report that the MOC efferent nerves also could innervate supporting cells (SCs) in the vicinity of OHCs to regulate hearing sensitivity. MOC nerve fibers are cholinergic, and acetylcholine (ACh) is a primary neurotransmitter. Immunofluorescent staining showed that MOC nerve endings, presynaptic vesicular acetylcholine transporters (VAChTs), and postsynaptic ACh receptors were visible at SCs and in the SC area. Application of ACh in SCs could evoke a typical inward current and reduce gap junctions (GJs) between them, which consequently enhanced the direct effect of ACh on OHCs to shift but not eliminate OHC electromotility. This indirect, GJ-mediated inhibition had a long-lasting influence. In vivo experiments further demonstrated that deficiency of this GJ-mediated efferent pathway decreased the regulation of active cochlear amplification and compromised the protection against noise. In particular, distortion product otoacoustic emission (DPOAE) showed a delayed reduction after noise exposure. Our findings reveal a new pathway for the MOC efferent system via innervating SCs to control active cochlear amplification and hearing sensitivity. These data also suggest that this SC GJ-mediated efferent pathway may play a critical role in long-term efferent inhibition and is required for protection of hearing from noise trauma.NEW & NOTEWORTHY The cochlear efferent system provides a negative feedback to control hair cell activity and hearing sensitivity and plays a critical role in noise protection. We reveal a new efferent control pathway in which medial olivocochlear efferent fibers have innervations with cochlear supporting cells to control their gap junctions, therefore regulating outer hair cell electromotility and hearing sensitivity. This supporting cell gap junction-mediated efferent control pathway is required for the protection of hearing from noise.


Assuntos
Nervo Coclear/fisiopatologia , Células Ciliadas Auditivas Externas/fisiologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Neurônios Eferentes/fisiologia , Animais , Vias Eferentes/fisiopatologia , Feminino , Cobaias , Masculino
11.
Proc Natl Acad Sci U S A ; 116(51): 25948-25957, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31776257

RESUMO

The function of outer hair cells (OHCs), the mechanical actuators of the cochlea, involves the anchoring of their tallest stereocilia in the tectorial membrane (TM), an acellular structure overlying the sensory epithelium. Otogelin and otogelin-like are TM proteins related to secreted epithelial mucins. Defects in either cause the DFNB18B and DFNB84B genetic forms of deafness, respectively, both characterized by congenital mild-to-moderate hearing impairment. We show here that mutant mice lacking otogelin or otogelin-like have a marked OHC dysfunction, with almost no acoustic distortion products despite the persistence of some mechanoelectrical transduction. In both mutants, these cells lack the horizontal top connectors, which are fibrous links joining adjacent stereocilia, and the TM-attachment crowns coupling the tallest stereocilia to the TM. These defects are consistent with the previously unrecognized presence of otogelin and otogelin-like in the OHC hair bundle. The defective hair bundle cohesiveness and the absence of stereociliary imprints in the TM observed in these mice have also been observed in mutant mice lacking stereocilin, a model of the DFNB16 genetic form of deafness, also characterized by congenital mild-to-moderate hearing impairment. We show that the localizations of stereocilin, otogelin, and otogelin-like in the hair bundle are interdependent, indicating that these proteins interact to form the horizontal top connectors and the TM-attachment crowns. We therefore suggest that these 2 OHC-specific structures have shared mechanical properties mediating reaction forces to sound-induced shearing motion and contributing to the coordinated displacement of stereocilia.


Assuntos
Células Ciliadas Auditivas Externas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Estereocílios/metabolismo , Membrana Tectorial/metabolismo , Animais , Cóclea/citologia , Surdez/congênito , Surdez/genética , Surdez/metabolismo , Predisposição Genética para Doença , Células Ciliadas Auditivas Externas/citologia , Células Ciliadas Vestibulares/metabolismo , Perda Auditiva Neurossensorial/congênito , Perda Auditiva Neurossensorial/genética , Camundongos , Camundongos Knockout , Membrana Tectorial/citologia
12.
ORL J Otorhinolaryngol Relat Spec ; 83(4): 272-279, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33784680

RESUMO

OBJECTIVE: The aim of the study was to evaluate the association of conductive hearing loss (CHL) with the structural changes in the organ of Corti. METHODS: Twenty ears of 10 healthy adult Wistar albino rats were included in the study. The right ears (n = 10) of the animals served as controls (group 1), and no surgical intervention was performed in these ears. A tympanic membrane perforation without annulus removal was performed under operative microscope on the left ears (n = 5) in 5 of 10 animals (group 2). A tympanic membrane perforation with annulus removal was performed under operative microscope on the left ears (n = 5) of the remaining 5 animals (group 3). Auditory brainstem response testing was performed in the animals before the interventions. After 3 months, the animals were sacrificed, their temporal bones were removed, and inner ears were investigated using scanning electron microscopy (SEM). The organ of Corti was evaluated from the cochlear base to apex in the modiolar axis, and the parameters were scored semiquantitatively. RESULTS: In group 1, the pre- and post-intervention hearing thresholds were similar (p > 0.05). In group 2, a hearing decrease of at least 5 dB was encountered in all test frequencies (p > 0.05). In group 3, at the frequency range of 2-32 kHz, there was a significant hearing loss after 3 months (p < 0.01). After 3 months, the hearing thresholds in group 2 and 3 were higher than group 1 (p < 0.01). The hearing threshold in group 3 was higher than group 2 (p < 0.01). On SEM evaluation, the general cell morphology and stereocilia of the outer hair cells were preserved in all segments of the cochlea in group 1 with a mean SEM score of 0.2. There was segmental degeneration in the general cell morphology and outer hair cells in group 2 with a mean SEM score of 2.2. There was widespread degeneration in the general cell morphology and outer hair cells in group 3 with a mean SEM score of 3.2. The SEM scores of group 2 and 3 were significantly higher than group 1 (p < 0.05). The SEM scores of group 3 were significantly higher than group 2 (p < 0.05). CONCLUSION: CHL may be associated with an inner ear damage. The severity of damage appears to be associated with severity and duration of CHL. Early correction of CHL is advocated in order to reverse or prevent progression of the inner ear damage, which will enhance the success rates of hearing restoration surgeries. Subjective differences and compliance of the hearing aid users may be due to the impact of CHL on inner ear structures.


Assuntos
Cóclea , Perda Auditiva Condutiva , Animais , Limiar Auditivo , Potenciais Evocados Auditivos do Tronco Encefálico , Células Ciliadas Auditivas Externas , Audição , Perda Auditiva Condutiva/etiologia , Ratos
13.
J Biomech Eng ; 142(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32005999

RESUMO

Nobel Laureate von Békésy first presented traveling wave theory, which explains the vibration mechanism of the basilar membrane (BM) of cochlea in 1960, and thus the mysterious veil of passive phonoreceptive mechanism of human cochlea was unveiled. However, the interpretation of active phonoreceptive mechanism of human cochlea has been a major medical problem for mankind. The active mechanism can be reflected in structures and the perilymph where a series of complex coupling nonlinear motion process is observed in the cochlea. Because the cochlea is small and complex, vibration data of the whole BM are not yet available from existing experiments. To address the problem, first, the motion equations of the organ of Corti (OHC) are established, and the circuit equations of the outer hair cells (OHCs) in the perilymph and the relationship between the motion of the outer hair cells and the electromotile force are derived. Then the active feedback force on the BM is obtained. Finally, an analytical-numerical combination model, where both macrostructures and microstructures of cochlea are included, is established. The model not only vividly depicts the spatial helical body and biological materials of the cochlea but also reflects the fluid-solid coupling nonlinear motion of cochlear structures in the electrical environment. Thus, the active hearing mechanism of cochlea is revealed.


Assuntos
Membrana Basilar , Cóclea , Simulação por Computador , Células Ciliadas Auditivas Externas , Audição
14.
J Neurosci ; 38(24): 5495-5506, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29899032

RESUMO

The voltage-dependent protein SLC26a5 (prestin) underlies outer hair cell electromotility (eM), which is responsible for cochlear amplification in mammals. The electrical signature of eM is a bell-shaped nonlinear capacitance (NLC), deriving from prestin sensor-charge (Qp) movements, which peaks at the membrane voltage, Vh, where charge is distributed equally on either side of the membrane. Voltage dependencies of NLC and eM differ depending on interrogation frequency and intracellular chloride, revealing slow intermediate conformational transitions between anion binding and voltage-driven Qp movements. Consequently, NLC exhibits low-pass characteristics, substantially below prevailing estimates of eM frequency response. Here we study in guinea pig and mouse of either sex synchronous prestin electrical (NLC, Qp) and mechanical (eM) activity across frequencies under voltage clamp (whole cell and microchamber). We find that eM and Qp magnitude and phase correspond, indicating tight piezoelectric coupling. Electromechanical measures (both NLC and eM) show dual-Lorentzian, low-pass behavior, with a limiting (τ2) time constant at Vh of 32.6 and 24.8 µs, respectively. As expected for voltage-dependent kinetics, voltage excitation away from Vh has a faster, flatter frequency response, with our fastest measured τ2 for eM of 18.2 µs. Previous observations of ultrafast eM (τ ≈ 2 µs) were obtained at offsets far removed from Vh We hypothesize that trade-offs in eM gain-bandwith arising from voltage excitation at membrane potentials offset from Vh influence the effectiveness of cochlear amplification across frequencies.SIGNIFICANCE STATEMENT Of two types of hair cells within the organ of Corti, inner hair cells and outer hair cells, the latter evolved to boost sensitivity to sounds. Damage results in hearing loss of 40-60 dB, revealing amplification gains of 100-1000× that arise from voltage-dependent mechanical responses [electromotility (eM)]. eM, driven by the membrane protein prestin, may work beyond 70 kHz. However, this speed exceeds, by over an order of magnitude, kinetics of typical voltage-dependent membrane proteins. We find eM is actually low pass in nature, indicating that prestin bears kinetics typical of other membrane proteins. These observations highlight potential difficulties in providing sufficient amplification beyond a cutoff frequency near 20 kHz. Nevertheless, observed trade-offs in eM gain-bandwith may sustain cochlear amplification across frequency.


Assuntos
Percepção Auditiva/fisiologia , Células Ciliadas Auditivas Externas/fisiologia , Mecanotransdução Celular/fisiologia , Proteínas Motores Moleculares/fisiologia , Proteínas/fisiologia , Animais , Feminino , Cobaias , Cinética , Masculino , Camundongos
16.
Histochem Cell Biol ; 151(6): 461-474, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30963236

RESUMO

Cochlear hair cells are essential for the mechanotransduction of hearing. Sensorineural hearing loss can be irreversible because hair cells have a minimal ability to repair or regenerate themselves once damaged. In order to develop therapeutic interventions to prevent hair cell loss, it is necessary to understand the signaling pathway operating in cochlear hair cells and its alteration upon damage. Diacylglycerol kinase (DGK) regulates intracellular signal transduction through phosphorylation of lipidic second messenger diacylglycerol. We have previously reported characteristic expression and localization patterns of DGKs in various organs under pathophysiological conditions. Nevertheless, little is known about morphological and functional aspects of this enzyme family in the cochlea. First RT-PCR analysis reveals predominant mRNA expression of DGKα, DGKε and DGKζ. Immunohistochemical analysis shows that DGKζ localizes to the nuclei of inner hair cells (IHCs), outer hair cells (OHCs), supporting cells and spiral ganglion neurons in guinea pig cochlea under normal conditions. It is well known that loud noise exposure induces cochlear damage, thereby resulting in hair cell loss. In particular, OHCs are highly vulnerable to noise exposure than IHCs. We found that after 1 week of noise exposure DGKζ translocates from the nucleus to the cytoplasm in damage-sensitive OHCs and gradually disappears thereafter. In sharp contrast, DGKζ remains to the nucleus in damage-resistant IHCs. These results suggest that DGKζ cytoplasmic translocation is well correlated with cellular damage under noise-exposure stress conditions and is involved in delayed cell death in cochlear outer hair cells.


Assuntos
Cóclea/enzimologia , Diacilglicerol Quinase/análise , Ruído/efeitos adversos , Estresse Fisiológico , Animais , Cóclea/citologia , Cóclea/metabolismo , Diacilglicerol Quinase/metabolismo , Cobaias
17.
Biol Pharm Bull ; 42(1): 73-80, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30606991

RESUMO

It is well-known that outer hair cell (OHC) loss occurs in the cochlea of animal models of permanent hearing loss induced by intense noise exposure. Our earlier studies demonstrated the production of hydroxynonenal and peroxynitrite, as well as the disruption of gap junction-mediated intercellular communication (GJIC), in the cochlear spiral ligament prior to noise-induced sudden hearing loss. The goal of the present study was to evaluate the mechanism underlying cochlear OHC loss after sudden hearing loss induced by intense noise exposure. In organ of Corti explant cultures from mice, no significant OHC loss was observed after in vitro exposure to 4-hydroxynonenal (a product of lipid peroxidation), H2O2, SIN-1 (peroxynitrite generator), and carbenoxolone (a gap junction inhibitor). Interestingly, in vivo intracochlear carbenoxolone injection through the posterior semicircular canal caused marked OHC and hearing loss, as well as the disruption of gap junction-mediated intercellular communication in the cochlear spiral ligament. However, no significant OHC loss was observed in vivo in animals treated with 4-hydroxynonenal and SIN-1. Taken together, our data suggest that disruption of GJIC in the cochlear lateral wall structures is an important cause of cochlear OHC loss in models of hearing loss, including those induced by noise.


Assuntos
Estimulação Acústica/efeitos adversos , Comunicação Celular/fisiologia , Junções Comunicantes/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Perda Auditiva Provocada por Ruído/metabolismo , Ligamento Espiral da Cóclea/metabolismo , Aldeídos/toxicidade , Animais , Comunicação Celular/efeitos dos fármacos , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Junções Comunicantes/efeitos dos fármacos , Células Ciliadas Auditivas Externas/efeitos dos fármacos , Perda Auditiva Provocada por Ruído/induzido quimicamente , Perda Auditiva Provocada por Ruído/etiologia , Peróxido de Hidrogênio/toxicidade , Masculino , Camundongos , Técnicas de Cultura de Órgãos , Ligamento Espiral da Cóclea/efeitos dos fármacos
18.
Biochem Biophys Res Commun ; 500(2): 110-116, 2018 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-29571735

RESUMO

In hearing loss induced by aminoglycoside antibiotics, the outer hair cells (OHCs) in the basal turn are always more susceptible than OHCs in the apical turn, while the underlying mechanisms remain unknown. In this study, we reported that NAPDH oxidase 2 (NOX2) played an important role in the OHCs damage preferentially in the basal turn. Normally, NOX2 was evenly expressed in OHCs among different turns, at a relatively low level. However, after neomycin treatment, NOX2 was dominantly induced in OHCs in the basal turn. In vivo and in vitro studies demonstrated that inhibition of NOX2 significantly alleviated neomycin-induced OHCs damages, as seen from both the cleaved caspase-3 and TUNEL staining. Moreover, gp91 ds-tat delivery and DHE staining results showed that NOX2-derived ROS was responsible for neomycin ototoxicity. Taken together, our study shows that regional up-expression of NOX2 and subsequent increase of ROS in OHCs of the basal turn is an important factor contributing to the vulnerability of OHCs there, which should shed light on the prevention of hearing loss induced by aminoglycoside antibiotics.


Assuntos
Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patologia , NADPH Oxidase 2/metabolismo , Neomicina/efeitos adversos , Regulação para Cima , Animais , Apoptose/efeitos dos fármacos , Células Ciliadas Auditivas Externas/efeitos dos fármacos , Perda Auditiva/patologia , NADPH Oxidase 2/antagonistas & inibidores , Neomicina/administração & dosagem , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/efeitos dos fármacos
19.
Cell Mol Biol (Noisy-le-grand) ; 64(12): 2-10, 2018 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-30301494

RESUMO

The cochlea of guinea pigs was irradiated with different frequencies of bone-conducted ultrasound (BCU) at a specific dose to induce cochlear hair cell-specific injuries, in order to establish frequency-related cochlear hair cell-specific injury models. Cochlear near-field potentials were then evoked using BCU of different frequencies and intensities to explore the peripheral coding and recognition of BCU by the cochlea. The inner ears of guinea pigs were irradiated by 30 kHz at 100 db and 80 kHz at100 db BCU for 6h to create frequency-related, ultrasound-specific cochlear injury models. Then, 30 kHz and 80 kHz BCU of different intensities were used to evoke auditory brainstem response (ABR) thresholds, compound action potential (CAP) thresholds, and action potential (AP) intensity-amplitude input-output curves in the normal control group and the ultrasonic cochlear injury group. This allowed us to explore the coding and recognition of BCU frequencies and intensities by cochlear hair cells. Immunofluorescence assay of outer hair cell (OHC) Prestin and inner hair cell (IHC) Otofelin was performed to verify the injury models. Irradiation of guinea pig inner ears by 30 kHz and 80 kHz BCU at a specific dose induced hair cell injuries at different sites. Irradiation with low frequency BCU mainly induced OHC injury, whereas irradiation with high frequency BCU induced IHC injury; moreover, IHC injury was more serious than OHC injury. The 30 kHz-evoked ABR threshold was significantly higher in the 30 kHz ultrasonic cochlear injury group compared to the normal control group. The 30 kHz-evoked ABR threshold was significantly higher in the 30 kHz ultrasonic cochlear injury group compared to the 80 kHz ultrasonic cochlear injury group. The difference in the 80 kHz-evoked ABR thresholds were not significant between the 30 kHz and 80 kHz ultrasonic cochlear injury groups. The click- and 30 kHz-evoked AP intensity-amplitude curves for the 30 kHz ultrasonic cochlear injury group indicate that the AP amplitude evoked at the same intensity was higher in the 30 kHz-evoked group than the click-evoked group. The spatial positions of cochlear hair cells in guinea pigs had a coding function for the frequencies of low-frequency ultrasound. OHCs have an amplification effect on the coding of low-frequency ultrasonic intensities. The peripheral perception of high frequency BCU may not require the participation of cochlear hair cells.


Assuntos
Cóclea/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Ondas Ultrassônicas , Animais , Cóclea/efeitos da radiação , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos da radiação , Cobaias , Células Ciliadas Auditivas Internas/fisiologia , Células Ciliadas Auditivas Internas/efeitos da radiação
20.
Am J Otolaryngol ; 39(5): 594-598, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30025743

RESUMO

PURPOSE: To evaluate prestin as a biomarker for the identification of early ototoxicity. MATERIALS AND METHODS: Rats (n = 47) were randomly assigned to five groups: low-dose (LAG) or high-dose (HAG) amikacin (200 and 600 mg/kg/day, respectively, for 10 days), low-dose (LCIS)or high-dose (HCIS) cisplatin (single doses of 5 and 15 mg/kg, respectively, for 3 days), and control (n = 8). At the end of the experiment, measurement of distortion product-evoked otoacoustic emissions (DPOAE) were performed to evaluate hearing, then blood samples and both ear tissues were collected under anesthesia. Prestin levels were determined by ELISA. Cochlear damage was evaluated histologically using a 4-point scoring system. RESULTS: The mean serum prestin levels were 377.0 ±â€¯135.3, 411.3 ±â€¯73.1, 512.6 ±â€¯106.0, 455.0 ±â€¯74.2 and 555.3 ±â€¯47.9 pg/ml for control, LCIS, HCIS, LAG and HAG groups, respectively. There was significant difference between prestin levels of Control-LCIS-HCIS groups (p = 0.031) and prestin levels of Control-LAG-HAG groups (p = 0.003). There were also significant differences in prestin levels between the low- and high-dose cisplatin and amikacin groups (p = 0.028 and p = 0.011, respectively). Each group had significantly lower DPOAE results at 4, 6 and 8 kHz than control groups (p < 0.001). The LAG, HAG, LCIS and HCIS groups had significantly higher cochlear damage scores than the control group (p < 0.05). CONCLUSIONS: Higher doses of cisplatin and amikacin were associated with the greatest increases in serum prestin level and cochlear damage score. The results of this study suggest that prestin is a promising early indicator of cochlear damage.


Assuntos
Doenças Cocleares/sangue , Doenças Cocleares/diagnóstico , Células Ciliadas Auditivas Externas/patologia , Transportadores de Sulfato/sangue , Amicacina , Animais , Biomarcadores/sangue , Cisplatino , Doenças Cocleares/etiologia , Modelos Animais de Doenças , Masculino , Emissões Otoacústicas Espontâneas , Valor Preditivo dos Testes , Distribuição Aleatória , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa