Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 174(5): 1247-1263.e15, 2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30078710

RESUMO

Type I spiral ganglion neurons (SGNs) transmit sound information from cochlear hair cells to the CNS. Using transcriptome analysis of thousands of single neurons, we demonstrate that murine type I SGNs consist of subclasses that are defined by the expression of subsets of transcription factors, cell adhesion molecules, ion channels, and neurotransmitter receptors. Subtype specification is initiated prior to the onset of hearing during the time period when auditory circuits mature. Gene mutations linked to deafness that disrupt hair cell mechanotransduction or glutamatergic signaling perturb the firing behavior of SGNs prior to hearing onset and disrupt SGN subtype specification. We thus conclude that an intact hair cell mechanotransduction machinery is critical during the pre-hearing period to regulate the firing behavior of SGNs and their segregation into subtypes. Because deafness is frequently caused by defects in hair cells, our findings have significant ramifications for the etiology of hearing loss and its treatment.


Assuntos
Células Ciliadas Auditivas/fisiologia , Audição/fisiologia , Mecanotransdução Celular , Neurônios/fisiologia , Transdução de Sinais , Gânglio Espiral da Cóclea/fisiologia , Animais , Análise por Conglomerados , Marcadores Genéticos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Knockout , Mutação , Neuroglia/fisiologia , Análise de Sequência de RNA
2.
EMBO J ; 42(23): e114587, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37800695

RESUMO

Our sense of hearing enables the processing of stimuli that differ in sound pressure by more than six orders of magnitude. How to process a wide range of stimulus intensities with temporal precision is an enigmatic phenomenon of the auditory system. Downstream of dynamic range compression by active cochlear micromechanics, the inner hair cells (IHCs) cover the full intensity range of sound input. Yet, the firing rate in each of their postsynaptic spiral ganglion neurons (SGNs) encodes only a fraction of it. As a population, spiral ganglion neurons with their respective individual coding fractions cover the entire audible range. How such "dynamic range fractionation" arises is a topic of current research and the focus of this review. Here, we discuss mechanisms for generating the diverse functional properties of SGNs and formulate testable hypotheses. We postulate that an interplay of synaptic heterogeneity, molecularly distinct subtypes of SGNs, and efferent modulation serves the neural decomposition of sound information and thus contributes to a population code for sound intensity.


Assuntos
Cóclea , Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Internas/fisiologia , Som , Sinapses/fisiologia , Gânglio Espiral da Cóclea
3.
Proc Natl Acad Sci U S A ; 120(31): e2217033120, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37487063

RESUMO

Type I spiral ganglion neurons (SGNs) are the auditory afferents that transmit sound information from cochlear inner hair cells (IHCs) to the brainstem. These afferents consist of physiological subtypes that differ in their spontaneous firing rate (SR), activation threshold, and dynamic range and have been described as low, medium, and high SR fibers. Lately, single-cell RNA sequencing experiments have revealed three molecularly defined type I SGN subtypes. The extent to which physiological type I SGN subtypes correspond to molecularly defined subtypes is unclear. To address this question, we have generated mouse lines expressing CreERT2 in SGN subtypes that allow for a physiological assessment of molecular subtypes. We show that Lypd1-CreERT2 expressing SGNs represent a well-defined group of neurons that preferentially innervate the IHC modiolar side and exhibit a narrow range of low SRs. In contrast, Calb2-CreERT2 expressing SGNs preferentially innervate the IHC pillar side and exhibit a wider range of SRs, thus suggesting that a strict stratification of all SGNs into three molecular subclasses is not obvious, at least not with the CreERT2 tools used here. Genetically marked neuronal subtypes refine their innervation specificity onto IHCs postnatally during the time when activity is required to refine their molecular phenotype. Type I SGNs thus consist of genetically defined subtypes with distinct physiological properties and innervation patterns. The molecular subtype-specific lines characterized here will provide important tools for investigating the role of the physiologically distinct type I SGNs in encoding sound signals.


Assuntos
Tronco Encefálico , Células Ciliadas Vestibulares , Animais , Camundongos , Cóclea , Células Ciliadas Auditivas Internas , Neurônios
4.
Audiol Neurootol ; : 1-8, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38749408

RESUMO

INTRODUCTION: Noise-induced hearing loss is one of the most frequent recognized occupational diseases. The time course of the involved pathologies is still under investigation. Several studies have demonstrated an acute damage of the sensory tissue, but only few experiments investigated the degeneration of (type I) spiral ganglion neurons (SGNs), representing the primary neurons in the auditory system. The aim of the present study was to investigate the time course of SGN degeneration within a 7-day period after traumatic noise exposure starting immediately after trauma. METHODS: Young adult normal hearing mice were noise exposed for 3 h with a broadband noise (5-20 kHz) at 115 dB SPL. Auditory threshold shift was measured by auditory brainstem recordings, and SGN densities were analyzed at different time points during the first week after acoustic trauma. RESULTS: Significant reduction of SGN densities was detected and is accompanied by a significant hearing loss. Degeneration starts within hours after the applied trauma, further progressing within days post-exposure. DISCUSSION: Early neurodegeneration in the auditory periphery seems to be induced by direct overstimulation of the auditory nerve fibers. SGN loss is supposed to be a result of inflammatory responses and neural deprivation, leading to permanent hearing loss and auditory processing deficits.

5.
Dev Dyn ; 252(1): 124-144, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36284453

RESUMO

BACKGROUND: Proper connectivity between type I spiral ganglion neurons (SGNs) and inner hair cells (IHCs) in the cochlea is necessary for conveying sound information to the brain in mammals. Previous studies have shown that type I SGNs are heterogeneous in form, function and synaptic location on IHCs, but factors controlling their patterns of connectivity are not well understood. RESULTS: During development, cochlear supporting cells and SGNs express Semaphorin-3A (SEMA3A), a known axon guidance factor. Mice homozygous for a point mutation that attenuates normal SEMA3A repulsive activity (Sema3aK108N ) show cochleae with grossly normal patterns of IHC innervation. However, genetic sparse labeling and three-dimensional reconstructions of individual SGNs show that cochleae from Sema3aK108N mice lacked the normal synaptic distribution of type I SGNs. Additionally, Sema3aK108N cochleae show a disrupted distribution of GLUA2 postsynaptic patches around the IHCs. The addition of SEMA3A-Fc to postnatal cochleae led to increases in SGN branching, similar to the effects of inhibiting glutamate receptors. Ca2+ imaging studies show that SEMA3A-Fc decreases SGN activity. CONCLUSIONS: Contrary to the canonical view of SEMA3A as a guidance ligand, our results suggest SEMA3A may regulate SGN excitability in the cochlea, which may influence the morphology and synaptic arrangement of type I SGNs.


Assuntos
Células Ciliadas Auditivas , Semaforina-3A , Animais , Camundongos , Cóclea/metabolismo , Neurônios/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Gânglio Espiral da Cóclea/metabolismo
6.
Audiol Neurootol ; 28(1): 43-51, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36075188

RESUMO

INTRODUCTION: Loss of hair cells and degeneration of spiral ganglion neurons (SGN) lead to severe hearing loss or deafness. The successful use of a cochlear implant (CI) depends among other factors on the number of surviving SGN. Postoperative formation of fibrous tissue around the electrode array causes an increase in electrical impedances at the stimulating contacts. The use of immunophilin inhibitors may reduce the inflammatory processes without suppressing the immune response. Here, we report on in vitro experiments with different concentrations of immunophilin inhibitors MM284 and compound V20 regarding a possible application of these substances in the inner ear. METHODS: Standard cell lines (NIH/3T3 fibroblasts), freshly isolated SGN, and fibroblasts from neonatal rat cochleae (p3-5) were incubated with different concentrations of immunophilin inhibitors for 48 h. Metabolic activity of fibroblasts was investigated by MTT assay and cell survival by counting of immunochemically stained neurons and compared to controls. RESULTS: MM284 did not affect SGN numbers and neurite growth at concentrations of 4 × 10-5 mol/L and below, whereas V20 had no effect at 8 × 10-6 mol/L and below. Metabolic activity of fibroblasts was unchanged at these concentrations. CONCLUSION: Especially MM284 might be considered as a possible candidate for application within the cochlea.


Assuntos
Implantes Cocleares , Gânglio Espiral da Cóclea , Ratos , Animais , Imunofilinas/farmacologia , Cóclea , Neurônios , Fibroblastos
7.
Proc Natl Acad Sci U S A ; 117(7): 3828-3838, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32015128

RESUMO

Exposure to loud sound damages the postsynaptic terminals of spiral ganglion neurons (SGNs) on cochlear inner hair cells (IHCs), resulting in loss of synapses, a process termed synaptopathy. Glutamatergic neurotransmission via α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-type receptors is required for synaptopathy, and here we identify a possible involvement of GluA2-lacking Ca2+-permeable AMPA receptors (CP-AMPARs) using IEM-1460, which has been shown to block GluA2-lacking AMPARs. In CBA/CaJ mice, a 2-h exposure to 100-dB sound pressure level octave band (8 to 16 kHz) noise results in no permanent threshold shift but does cause significant synaptopathy and a reduction in auditory brainstem response (ABR) wave-I amplitude. Chronic intracochlear perfusion of IEM-1460 in artificial perilymph (AP) into adult CBA/CaJ mice prevented the decrease in ABR wave-I amplitude and the synaptopathy relative to intracochlear perfusion of AP alone. Interestingly, IEM-1460 itself did not affect the ABR threshold, presumably because GluA2-containing AMPARs can sustain sufficient synaptic transmission to evoke low-threshold responses during blockade of GluA2-lacking AMPARs. On individual postsynaptic densities, we observed GluA2-lacking nanodomains alongside regions with robust GluA2 expression, consistent with the idea that individual synapses have both CP-AMPARs and Ca2+-impermeable AMPARs. SGNs innervating the same IHC differ in their relative vulnerability to noise. We found local heterogeneity among synapses in the relative abundance of GluA2 subunits that may underlie such differences in vulnerability. We propose a role for GluA2-lacking CP-AMPARs in noise-induced cochlear synaptopathy whereby differences among synapses account for differences in excitotoxic susceptibility. These data suggest a means of maintaining normal hearing thresholds while protecting against noise-induced synaptopathy, via selective blockade of CP-AMPARs.


Assuntos
Cálcio/metabolismo , Cóclea/metabolismo , Perda Auditiva Provocada por Ruído/metabolismo , Ruído/efeitos adversos , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Animais , Potenciais Evocados Auditivos do Tronco Encefálico , Audição , Perda Auditiva Provocada por Ruído/etiologia , Perda Auditiva Provocada por Ruído/genética , Perda Auditiva Provocada por Ruído/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Receptores de AMPA/genética
8.
Int J Mol Sci ; 24(23)2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-38069416

RESUMO

Mammalian auditory hair cells transduce sound-evoked traveling waves in the cochlea into nerve stimuli, which are essential for hearing function. Pillar cells located between the inner and outer hair cells are involved in the formation of the tunnel of Corti, which incorporates outer-hair-cell-driven fluid oscillation and basilar membrane movement, leading to the fine-tuned frequency-specific perception of sounds by the inner hair cells. However, the detailed molecular mechanism underlying the development and maintenance of pillar cells remains to be elucidated. In this study, we examined the expression and function of brain-specific angiogenesis inhibitor 3 (Bai3), an adhesion G-protein-coupled receptor, in the cochlea. We found that Bai3 was expressed in hair cells in neonatal mice and pillar cells in adult mice, and, interestingly, Bai3 knockout mice revealed the abnormal formation of pillar cells, with the elevation of the hearing threshold in a frequency-dependent manner. Furthermore, old Bai3 knockout mice showed the degeneration of hair cells and spiral ganglion neurons in the basal turn. The results suggest that Bai3 plays a crucial role in the development and/or maintenance of pillar cells, which, in turn, are necessary for normal hearing function. Our results may contribute to understanding the mechanisms of hearing loss in human patients.


Assuntos
Cóclea , Audição , Proteínas de Membrana , Proteínas do Tecido Nervoso , Animais , Camundongos , Encéfalo , Cóclea/metabolismo , Células Ciliadas Auditivas Externas , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas de Membrana/genética
9.
Molecules ; 27(4)2022 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-35209205

RESUMO

Cochlear implants, like other active implants, rely on precise and effective electrical stimulation of the target tissue but become encapsulated by different amounts of fibrous tissue. The current study aimed at the development of a dual drug release from a PLLA coating and from the bulk material to address short-term and long-lasting release of anti-inflammatory drugs. Inner-ear cytocompatibility of drugs was studied in vitro. A PLLA coating (containing diclofenac) of medical-grade silicone (containing 5% dexamethasone) was developed and release profiles were determined. The influence of different coating thicknesses (2.5, 5 and 10 µm) and loadings (10% and 20% diclofenac) on impedances of electrical contacts were measured with and without pulsatile electrical stimulation. Diclofenac can be applied to the inner ear at concentrations of or below 4 × 10-5 mol/L. Release of dexamethasone from the silicone is diminished by surface coating but not blocked. Addition of 20% diclofenac enhances the dexamethasone release again. All PLLA coatings serve as insulator. This can be overcome by using removable masking on the contacts during the coating process. Dual drug release with different kinetics can be realized by adding drug-loaded coatings to drug-loaded silicone arrays without compromising electrical stimulation.


Assuntos
Anti-Inflamatórios , Materiais Revestidos Biocompatíveis/química , Implantes Cocleares , Dexametasona , Diclofenaco , Sistemas de Liberação de Medicamentos , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacocinética , Dexametasona/química , Dexametasona/farmacocinética , Diclofenaco/química , Diclofenaco/farmacocinética , Liberação Controlada de Fármacos , Ratos , Ratos Sprague-Dawley
10.
J Physiol ; 599(6): 1833-1854, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33450070

RESUMO

KEY POINTS: Sound information is transmitted by different subtypes of spiral ganglion neurons (SGN) from the ear to the brain. Selective damage of SGN peripheral synapses (cochlear synaptopathy) is widely recognized as one of the primary mechanisms of hearing loss, whereas the mechanisms at the SGN central synapses remain unclear. We report that different subtypes of SGN central synapses converge at different ratios onto individual target cochlear nucleus neurons with distinct physiological properties, and show biased morphological and physiological changes during age-related hearing loss (ARHL). The results reveal a new dimension in cochlear nucleus neural circuitry that systematically reassembles and processes auditory information from different SGN subtypes, which is altered during ageing and probably contributes to the development of ARHL. In addition to known cochlear synaptopathy, the present study shows that SGN central synapses are also pathologically changed during ageing, which collectively helps us better understand the structure and function of SGNs during ARHL. ABSTRACT: Sound information is transmitted from the cochlea to the brain by different subtypes of spiral ganglion neurons (SGN), which show varying degrees of vulnerability under pathological conditions. Selective cochlear synaptopathy, the preferential damage of certain subtypes of SGN peripheral synapses, has been recognized as one of the main mechanisms of hearing loss. The organization and function of the auditory nerve (AN) central synapses from different subtypes of SGNs remain unclear, including how different AN synapses reassemble onto individual neurons in the cochlear nucleus, as well as how they differentially change during hearing loss. Combining immunohistochemistry with electrophysiology, we investigated the convergence pattern and subtype-specific synaptopathy of AN synapses at the endbulb of Held, as well as the response properties of their postsynaptic bushy neurons in CBA/CaJ mice of either sex under normal hearing and age-related hearing loss (ARHL). We found that calretinin-expressing (type Ia ) and non-calretinin-expressing (type Ib /Ic ) endbulbs converged along a continuum of different ratios onto individual bushy neurons with varying physiological properties. Endbulbs degenerated during ageing in parallel with ARHL. Furthermore, the degeneration was more severe in non-calretinin-expressing synapses, which correlated with a gradual decrease in bushy neuron subpopulation predominantly innervated by these inputs. These synaptic and cellular changes were profound in middle-aged mice when their hearing thresholds were still relatively normal and prior to severe ARHL. Our findings suggest that biased AN central synaptopathy and the correlated shift in cochlear nucleus neuronal composition play significant roles in weakened auditory input and altered central auditory processing during ARHL.


Assuntos
Potenciais Evocados Auditivos do Tronco Encefálico , Perda Auditiva , Animais , Cóclea , Nervo Coclear , Camundongos , Camundongos Endogâmicos CBA , Gânglio Espiral da Cóclea , Sinapses
11.
J Neurophysiol ; 126(6): 2027-2038, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34788179

RESUMO

Cochlear synaptopathy is the noise-induced or age-related loss of ribbon synapses between inner hair cells (IHCs) and auditory-nerve fibers (ANFs), first reported in CBA/CaJ mice. Recordings from single ANFs in anesthetized, noise-exposed guinea pigs suggested that neurons with low spontaneous rates (SRs) and high thresholds are more vulnerable than low-threshold, high-SR fibers. However, there is extensive postexposure regeneration of ANFs in guinea pigs but not in mice. Here, we exposed CBA/CaJ mice to octave-band noise and recorded sound-evoked and spontaneous activity from single ANFs at least 2 wk later. Confocal analysis of cochleae immunostained for pre- and postsynaptic markers confirmed the expected loss of 40%-50% of ANF synapses in the basal half of the cochlea; however, our data were not consistent with a selective loss of low-SR fibers. Rather they suggested a loss of both SR groups in synaptopathic regions. Single-fiber thresholds and frequency tuning recovered to pre-exposure levels; however, response to tone bursts showed increased peak and steady-state firing rates, as well as decreased jitter in first-spike latencies. This apparent gain-of-function increased the robustness of tone-burst responses in the presence of continuous masking noise. This study suggests that the nature of noise-induced synaptic damage varies between different species and that, in mouse, the noise-induced hyperexcitability seen in central auditory circuits is also observed at the level of the auditory nerve.NEW & NOTEWORTHY Noise-induced damage to synapses between inner hair cells and auditory-nerve fibers (ANFs) can occur without permanent hair cell damage, resulting in pathophysiology that "hides" behind normal thresholds. Prior single-fiber neurophysiology in guinea pig suggested that noise selectively targets high-threshold ANFs. Here, we show that the lingering pathophysiology differs in mouse, with both ANF groups affected and a paradoxical gain-of-function in surviving low-threshold fibers, including increased onset rate, decreased onset jitter, and reduced maskability.


Assuntos
Doenças Cocleares/fisiopatologia , Nervo Coclear/fisiopatologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Gânglio Espiral da Cóclea/fisiopatologia , Sinapses/patologia , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos CBA
12.
Development ; 145(12)2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29784671

RESUMO

The cochlea is innervated by neurons that relay sound information from hair cells to central auditory targets. A subset of these are the type II spiral ganglion neurons, which have nociceptive features and contribute to feedback circuits providing neuroprotection in extreme noise. Type II neurons make a distinctive 90° turn towards the cochlear base to synapse with 10-15 outer hair cells. We demonstrate that this axon turning event requires planar cell polarity (PCP) signaling and is disrupted in Vangl2 and Celsr1 knockout mice, and that VANGL2 acts non-autonomously from the cochlea to direct turning. Moreover, VANGL2 is asymmetrically distributed at intercellular junctions between cochlear supporting cells, and in a pattern that could allow it to act directly as an axon guidance cue. Together, these data reveal a non-autonomous function for PCP signaling during axon guidance occurring in the tissue that is innervated, rather than the navigating growth cone.


Assuntos
Axônios/metabolismo , Células Ciliadas Auditivas Externas/fisiologia , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética , Gânglio Espiral da Cóclea/fisiologia , Animais , Polaridade Celular/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Nociceptividade/fisiologia , Ruído , Gânglio Espiral da Cóclea/embriologia
13.
Cell Biol Int ; 45(12): 2490-2498, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34435719

RESUMO

Age-related hearing loss (ARHL) is the most prevalent sensory deficit in the elderly, but its mechanism remains unclear. Scaffold protein prohibitin 2 (PHB2) has been widely involved in aging and neurodegeneration. However, the role of PHB2 in ARHL is undeciphered to date. To investigate the expression pattern and the role of PHB2 in ARHL, we used C57BL/6 mice and HEI-OC1 cell line as models. In our study, we have found PHB2 exists in the cochlea and is expressed in hair cells, spiral ganglion neurons, and HEI-OC1 cells. In mice with ARHL, mitophagy is reduced and correspondingly the expression level of PHB2 is decreased. Moreover, after H2 O2 treatment the mitophagy is activated and the PHB2 expression is increased. These findings indicate that PHB2 may exert an important role in ARHL through mitophagy. Findings from this study will be helpful for elucidating the mechanism underlying the ARHL and for providing a new target for ARHL treatment.


Assuntos
Envelhecimento/metabolismo , Cóclea/metabolismo , Perda Auditiva/metabolismo , Proibitinas/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitofagia/fisiologia , Neurônios/metabolismo , Presbiacusia/metabolismo , Gânglio Espiral da Cóclea/metabolismo
14.
Int J Mol Sci ; 21(18)2020 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-32933159

RESUMO

The administration of immune checkpoint inhibitors (ICIs) often leads to immune-related adverse events. However, their effect on auditory function is largely unexplored. Thorough preclinical studies have not been published yet, only sporadic cases and pharmacovigilance reports suggest their significance. Here we investigated the effect of anti-PD-1 antibody treatment (4 weeks, intraperitoneally, 200 µg/mouse, 3 times/week) on hearing function and cochlear morphology in C57BL/6J mice. ICI treatment did not influence the hearing thresholds in click or tone burst stimuli at 4-32 kHz frequencies measured by auditory brainstem response. The number and morphology of spiral ganglion neurons were unaltered in all cochlear turns. The apical-middle turns (<32 kHz) showed preservation of the inner and outer hair cells (OHCs), whilst ICI treatment mitigated the age-related loss of OHCs in the basal turn (>32 kHz). The number of Iba1-positive macrophages has also increased moderately in this high frequency region. We conclude that a 4-week long ICI treatment does not affect functional and morphological integrity of the inner ear in the most relevant hearing range (4-32 kHz; apical-middle turns), but a noticeable preservation of OHCs and an increase in macrophage activity appeared in the >32 kHz basal part of the cochlea.


Assuntos
Anticorpos Monoclonais/farmacologia , Limiar Auditivo/efeitos dos fármacos , Cóclea/efeitos dos fármacos , Células Ciliadas Auditivas Externas/efeitos dos fármacos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Audição , Inibidores de Checkpoint Imunológico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gânglio Espiral da Cóclea/efeitos dos fármacos
15.
J Cell Mol Med ; 23(8): 5098-5107, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31207045

RESUMO

The objective of this study was to elucidate whether paeoniflorin (PF) exerted an effect on cisplatin-induced spiral ganglion neuron (SGN) damage, with special attention given to the role of PINK1/BAD pathway in this process. Middle cochlear turn culture and C57BL/6 mice were utilized to identify the character of PF in vitro and in vivo. We found that cisplatin treatment led to SGN damage, in which reactive oxygen species (ROS) generation increased, PINK1 expression decreased, BAD accumulation on mitochondria raised and mitochondrial apoptotic pathway activated. Conversely, we demonstrated that PF pre-treatment obviously mitigated cisplatin-induced SGN damage. Mechanistic studies showed that PF could reduce ROS levels, increase PINK1 expression, decrease the BAD accumulation on mitochondria and, thus, alleviate the activated mitochondrial apoptosis in SGNs caused by cisplatin. Overall, the findings from this work reveal the important role of PF and provide another strategy against cisplatin-induced ototoxicity.


Assuntos
Cóclea/efeitos dos fármacos , Glucosídeos/farmacologia , Monoterpenos/farmacologia , Proteínas Quinases/genética , Gânglio Espiral da Cóclea/metabolismo , Proteína de Morte Celular Associada a bcl/genética , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cisplatino/efeitos adversos , Cisplatino/farmacologia , Cóclea/metabolismo , Cóclea/patologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Neurônios/efeitos dos fármacos , Neurônios/patologia , Substâncias Protetoras/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/patologia
16.
J Neurophysiol ; 122(5): 1962-1974, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31533018

RESUMO

Optogenetics comprise a promising alternative to electrical stimulation for characterization of neural circuits and for the next generation of neural prostheses. Optogenetic stimulation relies on expression of photosensitive microbial proteins in animal cells to initiate a flow of ions into the cells in response to visible light. Here, we generated a novel transgenic mouse model in which we studied the optogenetic activation of spiral ganglion neurons, the primary afferent neurons of the auditory system, and showed a strong optogenetic response, with a similar amplitude as the acoustically evoked response. A twofold increase in the level of channelrhodopsin expression significantly increased the photosensitivity at both the single cell and organismal levels but also partially compromised the native electrophysiological properties of the neurons. The importance of channelrhodopsin expression level to optogenetic stimulation, revealed by these quantitative measurements, will be significant for the characterization of neural circuitry and for the use of optogenetics in neural prostheses.NEW & NOTEWORTHY This study reveals a dose-response relationship between channelrhodopsin expression and optogenetic excitation. Both single cell and organismal responses depend on the expression level of the heterologous protein. Expression level of the opsin is thus an important variable in determining the outcome of an optogenetic experiment. These results are key to the implementation of neural prostheses based on optogenetics, such as next generation cochlear implants, which would use light to elicit a neural response to sound.


Assuntos
Channelrhodopsins/fisiologia , Cóclea/fisiologia , Fenômenos Eletrofisiológicos , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Neurônios Aferentes/fisiologia , Optogenética , Gânglio Espiral da Cóclea/fisiologia , Animais , Camundongos , Camundongos Transgênicos , Modelos Animais
17.
J Neurosci Res ; 97(11): 1414-1429, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31257632

RESUMO

The use of neurotrophic factors as therapeutic agents for neurodegenerative diseases is considered as an approach aimed at restoring and maintaining neuronal function in the peripheral and central nervous system. Since the neuroprotective effect is depending on chronic delivery of the neurotrophic factors a sustained application, e.g., via cell-based delivery is necessary. Human mesenchymal stem cells (hMSCs) were lentivirally modified to overexpress brain-derived neurotrophic factor (BDNF) and to express fluorescent marker genes for easy visualization. Since genetically modified cells should be site-specifically retained (e.g., by encapsulation) in the patients to avoid adverse effects the cells were additionally differentiated to chondrocytes to hypothetically improve their vitality and survival in a delivery matrix. Different polycations for lentiviral transduction were investigated for their efficiency. The success of differentiation was determined by analysis of chondrocyte marker genes and the neuroprotective effect of BDNF-overexpressing cells was exemplarily investigated on neurons of the peripheral auditory system. The genetically modified hMSCs overexpressed BDNF from under 1 to 125 ng ml-1  day-1 depending on the donor and transfection method. Using protamine sulfate the transfection efficacy was superior compared to the use of polybrene. The BDNF secreted by the MSCs was significantly neuroprotective in comparison to the relevant controls even though the produced mean concentrations were lower than the effective concentrations for recombinant industrially produced proteins described in literature. The presented system of BDNF-overexpressing hMSCs is neuroprotective and is therefore considered as a promising method for sustained delivery of proteins in therapeutically relevant amounts to degenerating neuronal structures.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Condrócitos/metabolismo , Engenharia Genética/métodos , Células-Tronco Mesenquimais/metabolismo , Fármacos Neuroprotetores , Fator Neurotrófico Derivado do Encéfalo/genética , Diferenciação Celular , Expressão Gênica , Vetores Genéticos/genética , Humanos , Lentivirus/genética , Neurônios/metabolismo
18.
Cell Tissue Res ; 378(1): 49-57, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31016387

RESUMO

The aim of this paper is to investigate the potential beneficial effects of taurine in cochlear neural stem cell (NSC) transplantation and elucidate the underlying molecular mechanism. The NSC cells were isolated from neonatal Balb/c mice and an auditory neuropathy gerbil model was established by microinjection of ouabain. The spiral ganglion neurons (SGN) were characterized with immunofluorescence stained with Tuj1 antibody. Cell proliferation was determined by BrdU incorporation assay and the morphologic index was measured under the light microscope. The relative protein level was determined by immunoblotting. The hearing of the animal model was scored by click- and tone burst-evoked auditory brainstem response (ABR). Here we consolidated our previous finding that taurine stimulated SGN density and the proliferation index, which were completely abolished by Shh inhibitor, cyclopamine. Transplantation of cochlear NSCs combined with taurine significantly improved ouabain-induced auditory neuropathy in gerbils. In addition, cyclopamine antagonized taurine's effect on glutamatergic and GABAergic neuron population via suppression of VGLUT1 and GAT1 expression. Mechanistically, taurine evidently activated the Sonic HedgeHog pathway and upregulated Shh, Ptc-1, Smo and Gli-1 proteins, which were specifically blockaded by cyclopamine. Here, for the first time demonstrated we that co-administration with taurine significantly improved NSC transplantation and the Shh pathway was identified in this beneficial effect.


Assuntos
Neurônios GABAérgicos/metabolismo , Perda Auditiva Central/cirurgia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/transplante , Gânglio Espiral da Cóclea/citologia , Taurina/farmacologia , Animais , Proliferação de Células , Neurônios GABAérgicos/citologia , Proteínas Hedgehog/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
19.
Synapse ; 73(5): e22087, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30592086

RESUMO

In mammals, hair cells and spiral ganglion neurons (SGNs) in the cochlea together are sophisticated "sensorineural" structures that transduce auditory information from the outside world into the brain. Hair cells and SGNs are joined by glutamatergic ribbon-type synapses composed of a molecular machinery rivaling in complexity the mechanoelectric transduction components found at the apical side of the hair cell. The cochlear hair cell ribbon synapse has received much attention lately because of recent and important findings related to its damage (sometimes termed "synaptopathy") as a result of noise overexposure. During development, ribbon synapses between type I SGNs and inner hair cells form in the time window between birth and hearing onset and is a process coordinated with type I SGN myelination, spontaneous activity, synaptic pruning, and innervation by efferents. In this review, we highlight new findings regarding the diversity of type I SGNs and inner hair cell synapses, and the molecular mechanisms of selective hair cell targeting. Also discussed are cell adhesion molecules and protein constituents of the ribbon synapse, and how these factors participate in ribbon synapse formation. We also note interesting new insights into the morphological development of type II SGNs, and the potential for cochlear macrophages as important players in protecting SGNs. We also address recent studies demonstrating that the structural and physiological profiles of the type I SGNs do not reach full maturity until weeks after hearing onset, suggesting a protracted development that is likely modulated by activity.


Assuntos
Neurogênese , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Sinapses/fisiologia , Animais , Células Ciliadas Auditivas/citologia , Células Ciliadas Auditivas/fisiologia , Humanos , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/fisiologia , Sinapses/ultraestrutura
20.
Adv Exp Med Biol ; 1130: 93-107, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30915703

RESUMO

In the auditory system, the primary sensory neurons, spiral ganglion neurons (SGNs), transmit complex acoustic information from hair cells to the second-order sensory neurons in the cochlear nucleus for sound processing, thus building the initial bridge between the physical world of sound and the perception of that sound. Cochlear SGN loss causes irreversible hearing impairment because this type of neural cell cannot regenerate. A better understanding of the molecular mechanisms of formation, structure, degeneration, and protection of SGNs will help to design potential therapeutic strategies for preservation and replacement of them in the cochlear implant recipient. In this review, we described and summarized the following about SGNs: (1) their cell biology and their peripheral and central connections, (2) mechanisms of their neuronal damage and their protection, and (3) the neural and synaptic mechanism of auditory neuropathy and current options for hearing rehabilitation from auditory neuropathy. The updates of the research progress and the significant issues on these topics were discussed.


Assuntos
Células Ciliadas Auditivas/citologia , Perda Auditiva Central/prevenção & controle , Neurônios/citologia , Gânglio Espiral da Cóclea/fisiopatologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA