Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS Biol ; 22(6): e3002665, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38935589

RESUMO

Loss of synapses between spiral ganglion neurons and inner hair cells (IHC synaptopathy) leads to an auditory neuropathy called hidden hearing loss (HHL) characterized by normal auditory thresholds but reduced amplitude of sound-evoked auditory potentials. It has been proposed that synaptopathy and HHL result in poor performance in challenging hearing tasks despite a normal audiogram. However, this has only been tested in animals after exposure to noise or ototoxic drugs, which can cause deficits beyond synaptopathy. Furthermore, the impact of supernumerary synapses on auditory processing has not been evaluated. Here, we studied mice in which IHC synapse counts were increased or decreased by altering neurotrophin 3 (Ntf3) expression in IHC supporting cells. As we previously showed, postnatal Ntf3 knockdown or overexpression reduces or increases, respectively, IHC synapse density and suprathreshold amplitude of sound-evoked auditory potentials without changing cochlear thresholds. We now show that IHC synapse density does not influence the magnitude of the acoustic startle reflex or its prepulse inhibition. In contrast, gap-prepulse inhibition, a behavioral test for auditory temporal processing, is reduced or enhanced according to Ntf3 expression levels. These results indicate that IHC synaptopathy causes temporal processing deficits predicted in HHL. Furthermore, the improvement in temporal acuity achieved by increasing Ntf3 expression and synapse density suggests a therapeutic strategy for improving hearing in noise for individuals with synaptopathy of various etiologies.


Assuntos
Células Ciliadas Auditivas Internas , Neurotrofina 3 , Sinapses , Animais , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Sinapses/metabolismo , Sinapses/fisiologia , Neurotrofina 3/metabolismo , Neurotrofina 3/genética , Camundongos , Limiar Auditivo , Potenciais Evocados Auditivos/fisiologia , Reflexo de Sobressalto/fisiologia , Percepção Auditiva/fisiologia , Gânglio Espiral da Cóclea/metabolismo , Feminino , Masculino , Perda Auditiva Oculta
2.
Proc Natl Acad Sci U S A ; 121(31): e2315599121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39058581

RESUMO

Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing "synaptopathy" and hearing loss. Cocultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that, when P3-5 denervated organs of Corti are cocultured with SGNs, newly formed IHC/SGN synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When using older organs of Corti at P10-11, synaptic activity probed by deconvolution showed more mature release properties, closer to the specialized mode of IHC synaptic transmission crucial for coding the sound signal. This functional assessment of newly formed IHC synapses developed here, provides a powerful tool for testing approaches to improve synapse regeneration.


Assuntos
Gânglio Espiral da Cóclea , Sinapses , Animais , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/fisiologia , Sinapses/fisiologia , Camundongos , Células Ciliadas Auditivas Internas/fisiologia , Células Ciliadas Auditivas Internas/metabolismo , Transmissão Sináptica/fisiologia , Neurônios/fisiologia , Neurônios/metabolismo , Regeneração/fisiologia , Células Ciliadas Auditivas/fisiologia , Técnicas de Cocultura/métodos , Optogenética/métodos , Regeneração Nervosa/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Órgão Espiral/fisiologia , Órgão Espiral/citologia , Órgão Espiral/metabolismo
3.
Hum Mol Genet ; 33(10): 905-918, 2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38449065

RESUMO

Mutations in AIFM1, encoding for apoptosis-inducing factor (AIF), cause AUNX1, an X-linked neurologic disorder with late-onset auditory neuropathy (AN) and peripheral neuropathy. Despite significant research on AIF, there are limited animal models with the disrupted AIFM1 representing the corresponding phenotype of human AUNX1, characterized by late-onset hearing loss and impaired auditory pathways. Here, we generated an Aifm1 p.R450Q knock-in mouse model (KI) based on the human AIFM1 p.R451Q mutation. Hemizygote KI male mice exhibited progressive hearing loss from P30 onward, with greater severity at P60 and stabilization until P210. Additionally, muscle atrophy was observed at P210. These phenotypic changes were accompanied by a gradual reduction in the number of spiral ganglion neuron cells (SGNs) at P30 and ribbons at P60, which coincided with the translocation of AIF into the nucleus starting from P21 and P30, respectively. The SGNs of KI mice at P210 displayed loss of cytomembrane integrity, abnormal nuclear morphology, and dendritic and axonal demyelination. Furthermore, the inner hair cells and myelin sheath displayed abnormal mitochondrial morphology, while fibroblasts from KI mice showed impaired mitochondrial function. In conclusion, we successfully generated a mouse model recapitulating AUNX1. Our findings indicate that disruption of Aifm1 induced the nuclear translocation of AIF, resulting in the impairment in the auditory pathway.


Assuntos
Fator de Indução de Apoptose , Modelos Animais de Doenças , Perda Auditiva , Animais , Humanos , Masculino , Camundongos , Fator de Indução de Apoptose/genética , Fator de Indução de Apoptose/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/genética , Técnicas de Introdução de Genes , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Perda Auditiva/genética , Perda Auditiva/patologia , Perda Auditiva/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/metabolismo , Mutação , Transporte Proteico , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia
4.
J Neurosci ; 44(7)2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38176908

RESUMO

Early B-cell factor 1 (EBF1) is a basic helix-loop-helix transcription factor essential for the differentiation of various tissues. Our single-cell RNA sequencing data suggest that Ebf1 is expressed in the sensory epithelium of the mouse inner ear. Here, we found that the murine Ebf1 gene and its protein are expressed in the prosensory domain of the inner ear, medial region of the cochlear duct floor, otic mesenchyme, and cochleovestibular ganglion. Ebf1 deletion in mice results in incomplete formation of the spiral limbus and scala tympani, increased number of cells in the organ of Corti and Kölliker's organ, and aberrant course of the spiral ganglion axons. Ebf1 deletion in the mouse cochlear epithelia caused the proliferation of SOX2-positive cochlear cells at E13.5, indicating that EBF1 suppresses the proliferation of the prosensory domain and cells of Kölliker's organ to facilitate the development of appropriate numbers of hair and supporting cells. Furthermore, mice with deletion of cochlear epithelium-specific Ebf1 showed poor postnatal hearing function. Our results suggest that Ebf1 is essential for normal auditory function in mammals.


Assuntos
Orelha Interna , Rampa do Tímpano , Animais , Camundongos , Cóclea/metabolismo , Ducto Coclear , Mamíferos , Gânglio Espiral da Cóclea , Fatores de Transcrição/metabolismo
5.
Hum Genet ; 143(8): 979-993, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39066985

RESUMO

Gasdermin E (GSDME), a member of the gasdermin protein family, is associated with post-lingual hearing loss. All GSDME pathogenic mutations lead to skipping exon 8; however, the molecular mechanisms underlying hearing loss caused by GSDME mutants remain unclear. GSDME was recently identified as one of the mediators of programmed cell death, including apoptosis and pyroptosis. Therefore, in this study, we injected mice with GSDME mutant (MT) and examined the expression levels to assess its effect on hearing impairment. We observed loss of hair cells in the organ of Corti and spiral ganglion neurons. Further, the N-terminal release from the GSDME mutant in HEI-OC1 cells caused pyroptosis, characterized by cell swelling and rupture of the plasma membrane, releasing lactate dehydrogenase and cytokines such as interleukin-1ß. We also observed that the N-terminal release from GSDME mutants could permeabilize the mitochondrial membrane, releasing cytochromes and activating the mitochondrial apoptotic pathway, thereby generating possible positive feedback on the cleavage of GSDME. Furthermore, we found that treatment with disulfiram or dimethyl fumarate might inhibit pyroptosis and apoptosis by inhibiting the release of GSDME-N from GSDME mutants. In conclusion, this study elucidated the molecular mechanism associated with hearing loss caused by GSDME gene mutations, offering novel insights for potential treatment strategies.


Assuntos
Apoptose , Piroptose , Piroptose/genética , Animais , Camundongos , Mutação com Ganho de Função , Perda Auditiva/genética , Perda Auditiva/patologia , Humanos , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Órgão Espiral/metabolismo , Órgão Espiral/patologia , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Gasderminas
6.
Biochem Biophys Res Commun ; 704: 149704, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38430700

RESUMO

Ribbon synapses in the cochlear hair cells are subject to extensive pruning and maturation processes before hearing onset. Previous studies have highlighted the pivotal role of thyroid hormone (TH) in this developmental process, yet the detailed mechanisms are largely unknown. In this study, we found that the thyroid hormone receptor α (Thrα) is expressed in both sensory epithelium and spiral ganglion neurons in mice. Hypothyroidism, induced by Pax8 gene knockout, significantly delays the synaptic pruning during postnatal development in mice. Detailed spatiotemporal analysis of ribbon synapse distribution reveals that synaptic maturation involves not only ribbon pruning but also their migration, both of which are notably delayed in the cochlea of Pax8 knockout mice. Intriguingly, postnatal hyperthyroidism, induced by intraperitoneal injections of liothyronine sodium (T3), accelerates the pruning of ribbon synapses to the mature state without affecting the auditory functions. Our findings suggest that thyroid hormone does not play a deterministic role but rather controls the timing of cochlear ribbon synapse maturation.


Assuntos
Cóclea , Sinapses , Animais , Camundongos , Sinapses/fisiologia , Hormônios Tireóideos , Gânglio Espiral da Cóclea , Audição/fisiologia , Camundongos Knockout
7.
J Nanobiotechnology ; 22(1): 458, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39085923

RESUMO

Cochlear implants can directly activate the auditory system's primary sensory neurons, the spiral ganglion neurons (SGNs), via circumvention of defective cochlear hair cells. This bypass restores auditory input to the brainstem. SGN loss etiologies are complex, with limited mammalian regeneration. Protecting and revitalizing SGN is critical. Tissue engineering offers a novel therapeutic strategy, utilizing seed cells, biomolecules, and scaffold materials to create a cellular environment and regulate molecular cues. This review encapsulates the spectrum of both human and animal research, collating the factors contributing to SGN loss, the latest advancements in the utilization of exogenous stem cells for auditory nerve repair and preservation, the taxonomy and mechanism of action of standard biomolecules, and the architectural components of scaffold materials tailored for the inner ear. Furthermore, we delineate the potential and benefits of the biohybrid neural interface, an incipient technology in the realm of implantable devices. Nonetheless, tissue engineering requires refined cell selection and differentiation protocols for consistent SGN quality. In addition, strategies to improve stem cell survival, scaffold biocompatibility, and molecular cue timing are essential for biohybrid neural interface integration.


Assuntos
Regeneração Nervosa , Gânglio Espiral da Cóclea , Engenharia Tecidual , Alicerces Teciduais , Gânglio Espiral da Cóclea/citologia , Humanos , Engenharia Tecidual/métodos , Animais , Alicerces Teciduais/química , Neurônios , Implantes Cocleares , Células-Tronco/citologia , Diferenciação Celular
8.
Int J Mol Sci ; 25(10)2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38791192

RESUMO

The synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) are the most vulnerable structures in the noise-exposed cochlea. Cochlear synaptopathy results from the disruption of these synapses following noise exposure and is considered the main cause of poor speech understanding in noisy environments, even when audiogram results are normal. Cochlear synaptopathy leads to the degeneration of SGNs if damaged IHC-SGN synapses are not promptly recovered. Oxidative stress plays a central role in the pathogenesis of cochlear synaptopathy. C-Phycocyanin (C-PC) has antioxidant and anti-inflammatory activities and is widely utilized in the food and drug industry. However, the effect of the C-PC on noise-induced cochlear damage is unknown. We first investigated the therapeutic effect of C-PC on noise-induced cochlear synaptopathy. In vitro experiments revealed that C-PC reduced the H2O2-induced generation of reactive oxygen species in HEI-OC1 auditory cells. H2O2-induced cytotoxicity in HEI-OC1 cells was reduced with C-PC treatment. After white noise exposure for 3 h at a sound pressure of 118 dB, the guinea pigs intratympanically administered 5 µg/mL C-PC exhibited greater wave I amplitudes in the auditory brainstem response, more IHC synaptic ribbons and more IHC-SGN synapses according to microscopic analysis than the saline-treated guinea pigs. Furthermore, the group treated with C-PC had less intense 4-hydroxynonenal and intercellular adhesion molecule-1 staining in the cochlea compared with the saline group. Our results suggest that C-PC improves cochlear synaptopathy by inhibiting noise-induced oxidative stress and the inflammatory response in the cochlea.


Assuntos
Cóclea , Molécula 1 de Adesão Intercelular , Ruído , Estresse Oxidativo , Ficocianina , Sinapses , Animais , Estresse Oxidativo/efeitos dos fármacos , Cobaias , Ficocianina/farmacologia , Ficocianina/uso terapêutico , Cóclea/metabolismo , Cóclea/efeitos dos fármacos , Cóclea/patologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Ruído/efeitos adversos , Molécula 1 de Adesão Intercelular/metabolismo , Perda Auditiva Provocada por Ruído/tratamento farmacológico , Perda Auditiva Provocada por Ruído/metabolismo , Perda Auditiva Provocada por Ruído/patologia , Espécies Reativas de Oxigênio/metabolismo , Masculino , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Peróxido de Hidrogênio/metabolismo , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patologia , Antioxidantes/farmacologia , Linhagem Celular , Perda Auditiva Oculta
9.
J Neural Eng ; 21(2)2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38547528

RESUMO

Objective. Cochlear implants provide auditory perception to those with severe to profound sensorineural hearing loss: however, the quality of sound perceived by users does not approximate natural hearing. This limitation is due in part to the large physical gap between the stimulating electrodes and their target neurons. Therefore, directing the controlled outgrowth of processes from spiral ganglion neurons (SGNs) into close proximity to the electrode array could provide significantly increased hearing function.Approach.For this objective to be properly designed and implemented, the ability and limits of SGN neurites to be guided must first be determined. In this work, we engineer precise topographical microfeatures with angle turn challenges of various geometries to study SGN pathfinding and use live imaging to better understand how neurite growth is guided by these cues.Main Results.We find that the geometry of the angled microfeatures determines the ability of neurites to navigate the angled microfeature turns. SGN neurite pathfinding fidelity is increased by 20%-70% through minor increases in microfeature amplitude (depth) and by 25% if the angle of the patterned turn is made obtuse. Further, we see that dorsal root ganglion neuron growth cones change their morphology and migration to become more elongated within microfeatures. Our observations also indicate complexities in studying neurite turning. First, as the growth cone pathfinds in response to the various cues, the associated neurite often reorients across the angle topographical microfeatures. Additionally, neurite branching is observed in response to topographical guidance cues, most frequently when turning decisions are most uncertain.Significance.Overall, the multi-angle channel micropatterned substrate is a versatile and efficient system to assess neurite turning and pathfinding in response to topographical cues. These findings represent fundamental principles of neurite pathfinding that will be essential to consider for the design of 3D systems aiming to guide neurite growthin vivo.


Assuntos
Implantes Cocleares , Neuritos , Cones de Crescimento , Células Cultivadas , Neurônios , Gânglio Espiral da Cóclea
10.
Sci Rep ; 14(1): 9593, 2024 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-38671022

RESUMO

Moderate-to-profound sensorineural hearing loss in humans is treatable by electrically stimulating the auditory nerve (AN) with a cochlear implant (CI). In the cochlea, the modiolus presents a porous bony interface between the CI electrode and the AN. New bone growth caused by the presence of the CI electrode or neural degeneration inflicted by ageing or otological diseases might change the effective porosity of the modiolus and, thereby, alter its electrical material properties. Using a volume conductor description of the cochlea, with the aid of a 'mapped conductivity' method and an ad-hoc 'regionally kinetic' equation system, we show that even a slight variation in modiolus porosity or pore distribution can disproportionately affect AN stimulation. Hence, because of porosity changes, an inconsistent CI performance might occur if neural degeneration or new bone growth progress after implantation. Appropriate electrical material properties in accordance with modiolar morphology and pathology should be considered in patient-specific studies. The present first-of-its-kind in-silico study advocates for contextual experimental studies to further explore the utility of modiolus porous morphology in optimising the CI outcome.


Assuntos
Implantes Cocleares , Gânglio Espiral da Cóclea , Porosidade , Humanos , Nervo Coclear , Neurônios/fisiologia , Estimulação Elétrica , Perda Auditiva Neurossensorial/terapia , Perda Auditiva Neurossensorial/cirurgia , Cóclea
11.
Otol Neurotol ; 45(3): 326-333, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38238917

RESUMO

BACKGROUND: Brain-derived neurotrophic factor (BDNF) is an important factor in the development and neuroprotection of afferent auditory pathways. In this study, we investigated the expression of BDNF in the afferent auditory pathway after cochlear implantation (CI), hypothesizing that electrical stimulation after CI stimulates BDNF expression in the afferent auditory pathway. METHODS: Archival human temporal bones from eight patients with a history of CI and five patients with normal hearing (ages 65-93 years old) were studied. Temporal bone specimens were immunoreacted with rabbit polyclonal antibodies against BDNF and mouse monoclonal antibodies against pan-neurofilaments. In cases of unilateral CI, the BDNF expression was compared with the contralateral unimplanted ear and normal temporal bones without hearing loss. RESULTS: BDNF immunoreactivity (IR) localized to the spiral ganglion neurons (SGNs) somata and the surrounding satellite cells. BDNF-IR in the spiral ganglia was similar in the apical, middle, and basal hook regions. Neurofilament IR localized to SGN nerve fibers in both implanted and unimplanted cochleae. BDNF-IR in the SGN and satellite cells was significantly increased in the implanted specimens compared with the unimplanted specimens ( p < 0.05) and the normal hearing specimens ( p < 0.05). BDNF-IR expression was similar in the unimplanted cochlea and in the normal cochlea. BDNF protein expression was increased despite complete loss of the organ of Corti hair cells and supporting cells. Even in the cases of CI with a 6-mm first-generation electrode, BDNF expression was upregulated throughout the cochlea. CONCLUSIONS: BDNF expression in the SGN appears to be upregulated by the electrical stimulation from CI. This study provides evidence that the electrical stimulation from CI may stimulate the expression of BDNF, playing a neuroprotective role in the rehabilitation of hearing in the deafened ear.


Assuntos
Implante Coclear , Surdez , Camundongos , Animais , Humanos , Coelhos , Idoso , Idoso de 80 Anos ou mais , Gânglio Espiral da Cóclea/fisiologia , Fator Neurotrófico Derivado do Encéfalo , Cóclea , Neurônios
12.
Hear Res ; 442: 108935, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38113793

RESUMO

Sound information is transduced from mechanical vibration to electrical signals in the cochlea, conveyed to and further processed in the brain to form auditory perception. During the process, spiral ganglion neurons (SGNs) are the key cells that connect the peripheral and central auditory systems by receiving information from hair cells in the cochlea and transmitting it to neurons of the cochlear nucleus (CN). Decades of research in the cochlea greatly improved our understanding of SGN function under normal and pathological conditions, especially about the roles of different subtypes of SGNs and their peripheral synapses. However, it remains less clear how SGN central terminals or auditory nerve (AN) synapses connect to CN neurons, and ultimately how peripheral pathology links to structural alterations and functional deficits in the central auditory nervous system. This review discusses recent progress about the morphological and physiological properties of different subtypes of AN synapses and associated postsynaptic CN neurons, their changes during aging, and the potential mechanisms underlying age-related hearing loss.


Assuntos
Núcleo Coclear , Perda Auditiva , Humanos , Núcleo Coclear/patologia , Nervo Coclear , Neurônios/patologia , Sinapses/patologia , Gânglio Espiral da Cóclea/patologia , Cóclea/fisiologia
13.
Cell Rep ; 43(4): 114025, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38564333

RESUMO

Type I spiral ganglion neurons (SGNs) convey sound information to the central auditory pathway by forming synapses with inner hair cells (IHCs) in the mammalian cochlea. The molecular mechanisms regulating the formation of the post-synaptic density (PSD) in the SGN afferent terminals are still unclear. Here, we demonstrate that brain-specific angiogenesis inhibitor 1 (BAI1) is required for the clustering of AMPA receptors GluR2-4 (glutamate receptors 2-4) at the PSD. Adult Bai1-deficient mice have functional IHCs but fail to transmit information to the SGNs, leading to highly raised hearing thresholds. Despite the almost complete absence of AMPA receptor subunits, the SGN fibers innervating the IHCs do not degenerate. Furthermore, we show that AMPA receptors are still expressed in the cochlea of Bai1-deficient mice, highlighting a role for BAI1 in trafficking or anchoring GluR2-4 to the PSDs. These findings identify molecular and functional mechanisms required for sound encoding at cochlear ribbon synapses.


Assuntos
Cóclea , Audição , Densidade Pós-Sináptica , Receptores de AMPA , Receptores Acoplados a Proteínas G , Gânglio Espiral da Cóclea , Animais , Receptores de AMPA/metabolismo , Camundongos , Gânglio Espiral da Cóclea/metabolismo , Audição/fisiologia , Cóclea/metabolismo , Densidade Pós-Sináptica/metabolismo , Camundongos Knockout , Células Ciliadas Auditivas Internas/metabolismo , Camundongos Endogâmicos C57BL , Sinapses/metabolismo
14.
Neurosci Bull ; 40(8): 1093-1103, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38311706

RESUMO

GJB2 gene mutations are the most common causes of autosomal recessive non-syndromic hereditary deafness. For individuals suffering from severe to profound GJB2-related deafness, cochlear implants have emerged as the sole remedy for auditory improvement. Some previous studies have highlighted the crucial role of preserving cochlear neural components in achieving favorable outcomes after cochlear implantation. Thus, we generated a conditional knockout mouse model (Cx26-CKO) in which Cx26 was completely deleted in the cochlear supporting cells driven by the Sox2 promoter. The Cx26-CKO mice showed severe hearing loss and massive loss of hair cells and Deiter's cells, which represented the extreme form of human deafness caused by GJB2 gene mutations. In addition, multiple pathological changes in the peripheral auditory nervous system were found, including abnormal innervation, demyelination, and degeneration of spiral ganglion neurons as well as disruption of heminodes in Cx26-CKO mice. These findings provide invaluable insights into the deafness mechanism and the treatment for severe deafness in Cx26-null mice.


Assuntos
Conexina 26 , Conexinas , Surdez , Camundongos Knockout , Gânglio Espiral da Cóclea , Animais , Gânglio Espiral da Cóclea/patologia , Surdez/genética , Surdez/patologia , Conexinas/genética , Conexinas/deficiência , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/genética , Camundongos , Neurônios/patologia , Neurônios/metabolismo , Modelos Animais de Doenças , Degeneração Neural/patologia , Degeneração Neural/genética , Cóclea/patologia
15.
PLoS One ; 19(7): e0307973, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39058727

RESUMO

Although cochlear implants have become a well-established method for patients with sensory neural hearing loss, clinical results indicate that in some cases, corrosion of electrode contacts leads to high impedance that interferes with successful stimulation of the auditory nerve. As it is unclear whether corrosion products induce cell damage, we focused on cell culture models of the organ of Corti cell line (HEI-OC1), rat spiral ganglion cells (SGC) and rat organ of Corti explant (OCex) cultivated from neonatal rat cochleae to characterize the cytotoxicity of sodium hexachloroplatinate (IV) (Na2(PtCl6)). The oxidative activity in HEI-OC1 cells decreased with increasing Na2(PtCl6) concentrations between 8 and 16 ng/µl, and live cell staining with Calcein acetoxymethyl/Ethidium homodimer III revealed an increasing number of cells with disrupted membranes. Ultrastructural evidence of mitophagy followed by necroptosis was detected. Additionally, exposure of the SGC to 15-35 ng/µl Na2(PtCl6) dose-dependently reduced neuronal survival and neuritogenesis, as determined by neurofilament antigen staining. In parallel, staining glial cells and fibroblasts with specific antibodies confirmed the dose-dependent induction of cell death by Na2(PtCl6). Exposure of the OCex to 25-45 ng/µl Na2(PtCl6) resulted in severe concentration-dependent hair cell loss. Our data show for the first time that Na2(PtCl6) induces cell death in a concentration-dependent manner in inner ear cell types and tissues.


Assuntos
Morte Celular , Órgão Espiral , Gânglio Espiral da Cóclea , Animais , Ratos , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/citologia , Órgão Espiral/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos
16.
Hear Res ; 447: 109024, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38735179

RESUMO

Delayed loss of residual acoustic hearing after cochlear implantation is a common but poorly understood phenomenon due to the scarcity of relevant temporal bone tissues. Prior histopathological analysis of one case of post-implantation hearing loss suggested there were no interaural differences in hair cell or neural degeneration to explain the profound loss of low-frequency hearing on the implanted side (Quesnel et al., 2016) and attributed the threshold elevation to neo-ossification and fibrosis around the implant. Here we re-evaluated the histopathology in this case, applying immunostaining and improved microscopic techniques for differentiating surviving hair cells from supporting cells. The new analysis revealed dramatic interaural differences, with a > 80 % loss of inner hair cells in the cochlear apex on the implanted side, which can account for the post-implantation loss of residual hearing. Apical degeneration of the stria further contributed to threshold elevation on the implanted side. In contrast, spiral ganglion cell survival was reduced in the region of the electrode on the implanted side, but apical counts in the two ears were similar to that seen in age-matched unimplanted control ears. Almost none of the surviving auditory neurons retained peripheral axons throughout the basal half of the cochlea. Relevance to cochlear implant performance is discussed.


Assuntos
Limiar Auditivo , Implante Coclear , Implantes Cocleares , Gânglio Espiral da Cóclea , Implante Coclear/instrumentação , Implante Coclear/efeitos adversos , Humanos , Gânglio Espiral da Cóclea/patologia , Gânglio Espiral da Cóclea/fisiopatologia , Células Ciliadas Auditivas Internas/patologia , Fatores de Tempo , Sobrevivência Celular , Masculino , Audição , Perda Auditiva/fisiopatologia , Perda Auditiva/patologia , Perda Auditiva/cirurgia , Perda Auditiva/etiologia , Feminino , Células Ciliadas Auditivas/patologia , Idoso , Degeneração Neural , Pessoa de Meia-Idade , Osso Temporal/patologia , Osso Temporal/cirurgia
17.
Sci Rep ; 14(1): 15296, 2024 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961203

RESUMO

Blast wave exposure, a leading cause of hearing loss and balance dysfunction among military personnel, arises primarily from direct mechanical damage to the mechanosensory hair cells and supporting structures or indirectly through excessive oxidative stress. We previously reported that HK-2, an orally active, multifunctional redox modulator (MFRM), was highly effective in reducing both hearing loss and hair cells loss in rats exposed to a moderate intensity workday noise that likely damages the cochlea primarily from oxidative stress versus direct mechanical trauma. To determine if HK-2 could also protect cochlear and vestibular cells from damage caused primarily from direct blast-induced mechanical trauma versus oxidative stress, we exposed rats to six blasts of 186 dB peak SPL. The rats were divided into four groups: (B) blast alone, (BEP) blast plus earplugs, (BHK-2) blast plus HK-2 and (BEPHK-2) blast plus earplugs plus HK-2. HK-2 was orally administered at 50 mg/kg/d from 7-days before to 30-day after the blast exposure. Cochlear and vestibular tissues were harvested 60-d post-exposure and evaluated for loss of outer hair cells (OHC), inner hair cells (IHC), auditory nerve fibers (ANF), spiral ganglion neurons (SGN) and vestibular hair cells in the saccule, utricle and semicircular canals. In the untreated blast-exposed group (B), massive losses occurred to OHC, IHC, ANF, SGN and only the vestibular hair cells in the striola region of the saccule. In contrast, rats treated with HK-2 (BHK-2) sustained significantly less OHC (67%) and IHC (57%) loss compared to the B group. OHC and IHC losses were smallest in the BEPHK-2 group, but not significantly different from the BEP group indicating lack of protective synergy between EP and HK-2. There was no loss of ANF, SGN or saccular hair cells in the BHK-2, BEP and BEPHK-2 groups. Thus, HK-2 not only significantly reduced OHC and IHC damage, but completely prevented loss of ANF, SGN and saccule hair cells. The powerful protective effects of this oral MFRM make HK-2 an extremely promising candidate for human clinical trials.


Assuntos
Traumatismos por Explosões , Células Ciliadas Vestibulares , Gânglio Espiral da Cóclea , Animais , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/patologia , Ratos , Traumatismos por Explosões/prevenção & controle , Células Ciliadas Vestibulares/efeitos dos fármacos , Células Ciliadas Vestibulares/metabolismo , Masculino , Oxirredução , Ratos Sprague-Dawley , Cóclea/efeitos dos fármacos , Cóclea/patologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/patologia , Estresse Oxidativo/efeitos dos fármacos , Perda Auditiva Provocada por Ruído/prevenção & controle , Perda Auditiva Provocada por Ruído/patologia
18.
Sci Rep ; 14(1): 10910, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38740884

RESUMO

Transforming growth factor-ß (TGF-ß) signaling plays a significant role in multiple biological processes, including inflammation, immunity, and cell death. However, its specific impact on the cochlea remains unclear. In this study, we aimed to investigate the effects of TGF-ß signaling suppression on auditory function and cochlear pathology in mice with kanamycin-induced ototoxicity. Kanamycin and furosemide (KM-FS) were systemically administered to 8-week-old C57/BL6 mice, followed by immediate topical application of a TGF-ß receptor inhibitor (TGF-ßRI) onto the round window membrane. Results showed significant TGF-ß receptor upregulation in spiral ganglion neurons (SGNs) after KM-FA ototoxicity, whereas expression levels in the TGF-ßRI treated group remained unchanged. Interestingly, despite no significant change in cochlear TGF-ß expression after KM-FS ototoxicity, TGF-ßRI treatment resulted in a significant decrease in TGF-ß signaling. Regarding auditory function, TGF-ßRI treatment offered no therapeutic effects on hearing thresholds and hair cell survival following KM-FS ototoxicity. However, SGN loss and macrophage infiltration were significantly increased with TGF-ßRI treatment. These results imply that inhibition of TGF-ß signaling after KM-FS ototoxicity promotes cochlear inflammation and SGN degeneration.


Assuntos
Canamicina , Ototoxicidade , Transdução de Sinais , Fator de Crescimento Transformador beta , Animais , Camundongos , Cóclea/metabolismo , Cóclea/efeitos dos fármacos , Cóclea/patologia , Furosemida/farmacologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Canamicina/toxicidade , Camundongos Endogâmicos C57BL , Ototoxicidade/etiologia , Ototoxicidade/metabolismo , Ototoxicidade/patologia , Transdução de Sinais/efeitos dos fármacos , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Fator de Crescimento Transformador beta/metabolismo
19.
J Biomed Mater Res B Appl Biomater ; 112(7): e35439, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38923766

RESUMO

Sensorineural hearing loss (SNHL) is mainly caused by injury or loss of hair cells (HCs) and associated spiral ganglion neurons (SGNs) in the inner ear. At present, there is still no effective treatment for SNHL in clinic. Recently, advances in organoid bring a promising prospect for research and treatment of SNHL. Meanwhile, three-dimensional (3D) printing provides a tremendous opportunity to construct versatile organoids for tissue engineering and regenerative medicine. In this study, gelatin (Gel), sodium alginate (SA), and polyvinyl alcohol (PVA) were used to fabricate biomimetic scaffold through 3D printing. The organ of Corti derived from neonatal mice inner ear was seeded on the PVA/Gel/SA scaffold to construct organ of Corti organoid. Then, the organ of Corti organoid was used to study the potential protective effects of berberine sulfate on neomycin-juried auditory HCs and SGNs. The results showed that the PVA/Gel/SA biomimetic 3D scaffolds had good cytocompatibilities and mechanical properties. The constructed organoid could maintain organ of Corti activity well in vitro. In addition, the injury intervention results showed that berberine sulfate could significantly inhibit neomycin-induced HC and SGN damage. This study suggests that the fabricated organoid is highly biomimetic to the organ of Corti, which may provide an effective model for drug development, cell and gene therapy for SNHL.


Assuntos
Berberina , Órgão Espiral , Alicerces Teciduais , Animais , Órgão Espiral/efeitos dos fármacos , Camundongos , Berberina/farmacologia , Berberina/química , Alicerces Teciduais/química , Organoides/metabolismo , Organoides/efeitos dos fármacos , Impressão Tridimensional , Alginatos/química , Alginatos/farmacologia , Gelatina/química , Gelatina/farmacologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Engenharia Tecidual , Álcool de Polivinil/química , Álcool de Polivinil/farmacologia , Perda Auditiva Neurossensorial , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/metabolismo
20.
Rev. bras. otorrinolaringol ; 66(2): 155-158, Abr. 2000.
Artigo em Português | LILACS | ID: biblio-1022710

RESUMO

O objetivo deste estudo foi avaliara fisiopatologia da disacusia em pacientes portadores de diabetes mellitus (DM). Métodos: Estudos de casos e controles, estudos experimentais e artigos de revisão abordando a disacusia em DM foram identificados, usando o sistema de pesquisa MEDLINE, e incluídos no trabalho conforme sua consistência metodológica. Resultados: Os estudos têm demonstrado as lesões otoacústicas em diferentes localizações, variando desde lesões micro-angiopáticas até perda de células ciliares externas, ou comprometimento do gânglio espiral. Conclusões: As evidências sugerem a existência de dano neurossenssorial em DM. No entanto, mais estudos são necessários para elucidar os mecanismos fisiopatológicos das lesões e também para demonstrar se os danos se localizam em uma estrutura particular ou se refletem uma condição multifatorial.


The purpose of this study was to evaluate the pathophysiology of hearing loss occurring in diabetes mellitus (DM). Methods: Case-control studies, experimentals studies and review articles that have been demonstrated hearing loss in diabetes mellitus was identificated using a MEDLINE research, and included in the study according its methodologic consistence. Results: The studies have been demonstrate the lesions of the inner ear in different locations, varying since microangiophatic lesions until loss of external hair cells, or compromise of spiral ganglion. Conclusions: The evidence suggest the existence of sensorineural damage in diabetes mellitus. Nevertheless, more studies are necessary to elucidate the pathophysiologic mecanisms of lesions. lf the lesions occur in a particular region or reflect a multifactorial condition remains to clear.


Assuntos
Humanos , Estria Vascular/fisiologia , Gânglio Espiral da Cóclea , Diabetes Mellitus/terapia , Angiopatias Diabéticas/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA