Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Antioxidants (Basel) ; 13(2)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38397748

RESUMO

The disruption of the synaptic connection between the sensory inner hair cells (IHCs) and the auditory nerve fiber terminals of the type I spiral ganglion neurons (SGN) has been observed early in several auditory pathologies (e.g., noise-induced or ototoxic drug-induced or age-related hearing loss). It has been suggested that glutamate excitotoxicity may be an inciting element in the degenerative cascade observed in these pathological cochlear conditions. Moreover, oxidative damage induced by free hydroxyl radicals and nitric oxide may dramatically enhance cochlear damage induced by glutamate excitotoxicity. To investigate the underlying molecular mechanisms involved in cochlear excitotoxicity, we examined the molecular basis responsible for kainic acid (KA, a full agonist of AMPA/KA-preferring glutamate receptors)-induced IHC synapse loss and degeneration of the terminals of the type I spiral ganglion afferent neurons using a cochlear explant culture from P3 mouse pups. Our results demonstrated that disruption of the synaptic connection between IHCs and SGNs induced increased levels of oxidative stress, as well as altered both mitochondrial function and neurotrophin signaling pathways. Additionally, the application of exogenous antioxidants and neurotrophins (NT3, BDNF, and small molecule TrkB agonists) clearly increases synaptogenesis. These results suggest that understanding the molecular pathways involved in cochlear excitotoxicity is of crucial importance for the future clinical trials of drug interventions for auditory synaptopathies.

2.
J Clin Med ; 12(3)2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36769387

RESUMO

Auditory neuropathy spectrum disorder (ANSD) refers to a range of hearing impairments characterized by an impaired transmission of sound from the cochlea to the brain. This defect can be due to a lesion or defect in the inner hair cell (IHC), IHC ribbon synapse (e.g., pre-synaptic release of glutamate), postsynaptic terminals of the spiral ganglion neurons, or demyelination and axonal loss within the auditory nerve. To date, the only clinical treatment options for ANSD are hearing aids and cochlear implantation. However, despite the advances in hearing-aid and cochlear-implant technologies, the quality of perceived sound still cannot match that of the normal ear. Recent advanced genetic diagnostics and clinical audiology made it possible to identify the precise site of a lesion and to characterize the specific disease mechanisms of ANSD, thus bringing renewed hope to the treatment or prevention of auditory neurodegeneration. Moreover, genetic routes involving the replacement or corrective editing of mutant sequences or defected genes to repair damaged cells for the future restoration of hearing in deaf people are showing promise. In this review, we provide an update on recent discoveries in the molecular pathophysiology of genetic lesions, auditory synaptopathy and neuropathy, and gene-therapy research towards hearing restoration in rodent models and in clinical trials.

3.
FASEB J ; 35(12): e22025, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34758144

RESUMO

Mepyramine, a first-generation antihistamine targeting the histamine H(1) receptor, was extensively prescribed to patients suffering from allergic reactions and urticaria. Serious adverse effects, especially in case of overdose, were frequently reported, including drowsiness, impaired thinking, convulsion, and coma. Many of these side effects were associated with the blockade of histaminergic or cholinergic receptors. Here we show that mepyramine directly inhibits a variety of voltage-gated sodium channels, including the Tetrodotoxin-sensitive isoforms and the main isoforms (Nav1.7, Nav1.8, and Nav1.9) of nociceptors. Estimated IC50 were within the range of drug concentrations detected in poisoned patients. Mepyramine inhibited sodium channels through fast- or slow-inactivated state preference depending on the isoform. Moreover, mepyramine inhibited the firing responses of C- and Aß-type nerve fibers in ex vivo skin-nerve preparations. Locally applied mepyramine had analgesic effects on the scorpion toxin-induced excruciating pain and produced pain relief in acute, inflammatory, and chronic pain models. Collectively, these data provide evidence that mepyramine has the potential to be developed as a topical analgesic agent.


Assuntos
Artrite Experimental/complicações , Gânglios Espinais/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.8/fisiologia , Nociceptores/efeitos dos fármacos , Dor/tratamento farmacológico , Pirilamina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Potenciais de Ação , Animais , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Antagonistas dos Receptores Histamínicos H1/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.8/química , Nociceptores/metabolismo , Nociceptores/patologia , Dor/etiologia , Dor/metabolismo , Dor/patologia
5.
Front Cell Neurosci ; 15: 658990, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33828461

RESUMO

Pituitary adenylyl cyclase-activating polypeptide (PACAP) is a member of the vasoactive intestinal polypeptide (VIP)-the secretin-glucagon family of neuropeptides. They act through two classes of receptors: PACAP type 1 (PAC1) and type 2 (VPAC1 and VPAC2). Among their pleiotropic effects throughout the body, PACAP functions as neuromodulators and neuroprotectors, rescuing neurons from apoptosis, mostly through the PAC1 receptor. To explore the potential protective effect of endogenous PACAP against Noise-induced hearing loss (NIHL), we used a knockout mouse model lacking PAC1 receptor expression (PACR1-/-) and a transgenic humanized mouse model expressing the human PAC1 receptor (TgHPAC1R). Based on complementary approaches combining electrophysiological, histochemical, and molecular biological evaluations, we show PAC1R expression in spiral ganglion neurons and in cochlear apical cells of the organ of Corti. Wild-type (WT), PAC1R-/-, and TgHPAC1R mice exhibit similar auditory thresholds. For most of the frequencies tested after acute noise damage, however, PAC1R-/- mice showed a larger elevation of the auditory threshold than did their WT counterparts. By contrast, in a transgene copy number-dependent fashion, TgHPAC1R mice showed smaller noise-induced elevations of auditory thresholds compared to their WT counterparts. Together, these findings suggest that PACAP could be a candidate for endogenous protection against noise-induced hearing loss.

6.
Front Cell Neurosci ; 15: 733004, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975405

RESUMO

NMDA receptors (NMDARs) populate the complex between inner hair cell (IHC) and spiral ganglion neurons (SGNs) in the developing and mature cochlea. However, in the mature cochlea, activation of NMDARs is thought to mainly occur under pathological conditions such as excitotoxicity. Ototoxic drugs such as aspirin enable cochlear arachidonic-acid-sensitive NMDAR responses, and induced chronic tinnitus was blocked by local application of NMDAR antagonists into the cochlear fluids. We largely ignore if other modulators are also engaged. In the brain, D-serine is the primary physiological co-agonist of synaptic NMDARs. Whether D-serine plays a role in the cochlea had remained unexplored. We now reveal the presence of D-serine and its metabolic enzymes prior to, and at hearing onset, in the sensory and non-neuronal cells of the cochlea of several vertebrate species. In vivo intracochlear perfusion of D-serine in guinea pigs reduces sound-evoked activity of auditory nerve fibers without affecting the receptor potentials, suggesting that D-serine acts specifically on the postsynaptic auditory neurons without altering the functional state of IHC or of the stria vascularis. Indeed, we demonstrate in vitro that agonist-induced activation of NMDARs produces robust calcium responses in rat SGN somata only in the presence of D-serine, but not of glycine. Surprisingly, genetic deletion in mice of serine racemase (SR), the enzyme that catalyzes D-serine, does not affect hearing function, but offers protection against noise-induced permanent hearing loss as measured 3 months after exposure. However, the mechanisms of activation of NMDA receptors in newborn rats may be different from those in adult guinea pigs. Taken together, these results demonstrate for the first time that the neuro-messenger D-serine has a pivotal role in the cochlea by promoting the activation of silent cochlear NMDAR in pathological situations. Thus, D-serine and its signaling pathway may represent a new druggable target for treating sensorineural hearing disorders (i.e., hearing loss, tinnitus).

7.
J Pain ; 22(4): 440-453, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33227509

RESUMO

Oral amitriptyline hydrochloride (amitriptyline) is ineffective against some forms of chronic pain and is often associated with dose-limiting adverse events. We evaluated the potential effectiveness of high-dose topical amitriptyline in a preliminary case series of chemotherapy-induced peripheral neuropathy patients and investigated whether local or systemic adverse events associated with the use of amitriptyline were present in these patients. We also investigated the mechanism of action of topically administered amitriptyline in mice. Our case series suggested that topical 10% amitriptyline treatment was associated with pain relief in chemotherapy-induced peripheral neuropathy patients, without the side effects associated with systemic absorption. Topical amitriptyline significantly increased mechanical withdrawal thresholds when applied to the hind paw of mice, and inhibited the firing responses of C-, Aß- and Aδ-type peripheral nerve fibers in ex vivo skin-saphenous nerve preparations. Whole-cell patch-clamp recordings on cultured sensory neurons revealed that amitriptyline was a potent inhibitor of the main voltage-gated sodium channels (Nav1.7, Nav1.8, and Nav1.9) found in nociceptors. Calcium imaging showed that amitriptyline activated the transient receptor potential cation channel, TRPA1. Our case series indicated that high-dose 10% topical amitriptyline could alleviate neuropathic pain without adverse local or systemic effects. This analgesic action appeared to be mediated through local inhibition of voltage-gated sodium channels. PERSPECTIVE: Our preliminary case series suggested that topical amitriptyline could provide effective pain relief for chemotherapy-induced peripheral neuropathy patients without any systemic or local adverse events. Investigation of the mechanism of this analgesic action in mice revealed that this activity was mediated through local inhibition of nociceptor Nav channels.


Assuntos
Amitriptilina/farmacologia , Analgésicos não Narcóticos/farmacologia , Antineoplásicos/efeitos adversos , Dor Nociceptiva/tratamento farmacológico , Nociceptores/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Canal de Cátion TRPA1/efeitos dos fármacos , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Canais de Sódio Disparados por Voltagem/efeitos dos fármacos , Administração Tópica , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Amitriptilina/administração & dosagem , Amitriptilina/efeitos adversos , Analgésicos não Narcóticos/administração & dosagem , Analgésicos não Narcóticos/efeitos adversos , Animais , Comportamento Animal/efeitos dos fármacos , Criança , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.7 , Canal de Sódio Disparado por Voltagem NAV1.8 , Canal de Sódio Disparado por Voltagem NAV1.9 , Bloqueadores do Canal de Sódio Disparado por Voltagem/administração & dosagem , Bloqueadores do Canal de Sódio Disparado por Voltagem/efeitos adversos , Adulto Jovem
8.
Nat Commun ; 10(1): 4253, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31534133

RESUMO

Medication-overuse headaches (MOH) occur with both over-the-counter and pain-relief medicines, including paracetamol, opioids and combination analgesics. The mechanisms that lead to MOH are still uncertain. Here, we show that abnormal activation of Nav1.9 channels by Nitric Oxide (NO) is responsible for MOH induced by triptan migraine medicine. Deletion of the Scn11a gene in MOH mice abrogates NO-mediated symptoms, including cephalic and extracephalic allodynia, photophobia and phonophobia. NO strongly activates Nav1.9 in dural afferent neurons from MOH but not normal mice. Abnormal activation of Nav1.9 triggers CGRP secretion, causing artery dilatation and degranulation of mast cells. In turn, released mast cell mediators potentiates Nav1.9 in meningeal nociceptors, exacerbating inflammation and pain signal. Analysis of signaling networks indicates that PKA is downregulated in trigeminal neurons from MOH mice, relieving its inhibitory action on NO-Nav1.9 coupling. Thus, anomalous activation of Nav1.9 channels by NO, as a result of chronic medication, promotes MOH.


Assuntos
Transtornos da Cefaleia Secundários/patologia , Transtornos de Enxaqueca/patologia , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Neurônios Aferentes/metabolismo , Óxido Nítrico/metabolismo , Triptaminas/efeitos adversos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Degranulação Celular/fisiologia , Células Cultivadas , Feminino , Transtornos da Cefaleia Secundários/induzido quimicamente , Hiperalgesia/fisiopatologia , Masculino , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.9/genética , Neurônios Aferentes/efeitos dos fármacos , Nociceptores/fisiologia , Dor/fisiopatologia , Uso Excessivo de Medicamentos Prescritos/efeitos adversos
9.
Cell Rep ; 11(7): 1067-78, 2015 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-25959819

RESUMO

Cold-triggered pain is essential to avoid prolonged exposure to harmfully low temperatures. However, the molecular basis of noxious cold sensing in mammals is still not completely understood. Here, we show that the voltage-gated Nav1.9 sodium channel is important for the perception of pain in response to noxious cold. Nav1.9 activity is upregulated in a subpopulation of damage-sensing sensory neurons responding to cooling, which allows the channel to amplify subthreshold depolarizations generated by the activation of cold transducers. Consequently, cold-triggered firing is impaired in Nav1.9(-/-) neurons, and Nav1.9 null mice and knockdown rats show increased cold pain thresholds. Disrupting Nav1.9 expression in rodents also alleviates cold pain hypersensitivity induced by the antineoplastic agent oxaliplatin. We conclude that Nav1.9 acts as a subthreshold amplifier in cold-sensitive nociceptive neurons and is required for the perception of cold pain under normal and pathological conditions.


Assuntos
Hiperalgesia/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.9/metabolismo , Percepção da Dor/fisiologia , Sensação Térmica/fisiologia , Animais , Temperatura Baixa , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptores/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
10.
Pflugers Arch ; 467(1): 109-19, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25416542

RESUMO

Physical contact with the external world occurs through specialized neural structures called mechanoreceptors. Cutaneous mechanoreceptors provide information to the central nervous system (CNS) about touch, pressure, vibration, and skin stretch. The physiological function of these mechanoreceptors is to convert physical forces into neuronal signals. Key questions concern the molecular identity of the mechanoelectric transducer channels and the mechanisms by which the physical parameters of the mechanical stimulus are encoded into patterns of action potentials (APs). Compelling data indicate that the biophysical traits of mechanosensitive channels combined with the collection of voltage-gated channels are essential to describe the nature of the stimulus. Recent research also points to a critical role of the auxiliary cell-nerve ending communication in encoding stimulus properties. This review describes the characteristics of ion channels responsible for translating mechanical stimuli into the neural codes that underlie touch perception and pain.


Assuntos
Gânglios Espinais/fisiologia , Mecanorreceptores/fisiologia , Mecanotransdução Celular/fisiologia , Fenômenos Fisiológicos da Pele , Pele/inervação , Tato/fisiologia , Potenciais de Ação/fisiologia , Vias Aferentes/fisiologia , Animais , Humanos , Modelos Biológicos , Percepção da Dor/fisiologia
11.
Methods Mol Biol ; 998: 159-70, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23529428

RESUMO

Mechanotransduction, the conversion of a mechanical stimulus into a biological response, constitutes the basis of a variety of physiological functions such as the senses of touch, balance, proprioception, blood pressure, and hearing. In vertebrates, mechanosensation is mediated by mechanosensory neurons, whose cell bodies are located in trigeminal and dorsal root ganglia. Here, we describe an in vitro model of mechanotransduction that provides an opportunity to explore the properties of mechanosensitive channels in mammalian sensory neurons. The mechano-clamp method allows applying local force on plasma membrane of whole-cell patch-clamped sensory neurons. This technique uses a mechanical probe driven by a computer-assisted piezoelectric microstage to repeatedly stimulate sensory neurons with accurate control of stimulus strength, duration, and speed.


Assuntos
Eletricidade , Fenômenos Mecânicos , Técnicas de Patch-Clamp/métodos , Células Receptoras Sensoriais/citologia , Animais , Fenômenos Eletrofisiológicos , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Masculino , Mecanotransdução Celular , Técnicas de Patch-Clamp/instrumentação , Ratos , Ratos Wistar
12.
Cell ; 150(3): 633-46, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863013

RESUMO

N-methyl-d-aspartate receptors (NMDARs) are located in neuronal cell membranes at synaptic and extrasynaptic locations, where they are believed to mediate distinct physiological and pathological processes. Activation of NMDARs requires glutamate and a coagonist whose nature and impact on NMDAR physiology remain elusive. We report that synaptic and extrasynaptic NMDARs are gated by different endogenous coagonists, d-serine and glycine, respectively. The regionalized availability of the coagonists matches the preferential affinity of synaptic NMDARs for d-serine and extrasynaptic NMDARs for glycine. Furthermore, glycine and d-serine inhibit NMDAR surface trafficking in a subunit-dependent manner, which is likely to influence NMDARs subcellular location. Taking advantage of this coagonist segregation, we demonstrate that long-term potentiation and NMDA-induced neurotoxicity rely on synaptic NMDARs only. Conversely, long-term depression requires both synaptic and extrasynaptic receptors. Our observations provide key insights into the operating mode of NMDARs, emphasizing functional distinctions between synaptic and extrasynaptic NMDARs in brain physiology.


Assuntos
Glicina/metabolismo , Plasticidade Neuronal , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Serina/metabolismo , Sinapses , Animais , Membrana Celular , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Neuroglia/metabolismo , Neurônios/citologia , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo
13.
Antioxid Redox Signal ; 16(3): 263-74, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21923553

RESUMO

AIMS: In our aging society, age-related hearing loss (ARHL) or presbycusis is increasingly important. Here, we study the mechanism of ARHL using the senescence-accelerated mouse prone 8 (SAMP8) which is a useful model to probe the effects of aging on biological processes. RESULTS: We found that the SAMP8 strain displays premature hearing loss and cochlear degeneration recapitulating the processes observed in human presbycusis (i.e., strial, sensory, and neural degeneration). The molecular mechanisms associated with premature ARHL in SAMP8 mice involve oxidative stress, altered levels of antioxidant enzymes, and decreased activity of Complexes I, II, and IV, which in turn lead to chronic inflammation and triggering of apoptotic cell death pathways. In addition, spiral ganglion neurons (SGNs) also undergo autophagic stress and accumulated lipofuscin. INNOVATION AND CONCLUSION: Our results provide evidence that targeting oxidative stress, chronic inflammation, or apoptotic pathways may have therapeutic potential. Modulation of autophagy may be another strategy. The fact that autophagic stress and protein aggregation occurred specifically in SGNs also offers promising perspectives for the prevention of neural presbycusis.


Assuntos
Autofagia , Cóclea/patologia , Inflamação/metabolismo , Presbiacusia/metabolismo , Potenciais de Ação , Fatores Etários , Animais , Apoptose , Cóclea/imunologia , Cóclea/inervação , Cóclea/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Fibroblastos/patologia , Células Ciliadas Auditivas Externas/patologia , Células Ciliadas Auditivas Externas/fisiologia , Lipofuscina/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Malondialdeído/metabolismo , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Órgão Espiral/metabolismo , Órgão Espiral/patologia , Presbiacusia/imunologia , Presbiacusia/patologia , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Estria Vascular/patologia
14.
Am J Hum Genet ; 83(2): 278-92, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18674745

RESUMO

Autosomal-dominant sensorineural hearing loss is genetically heterogeneous, with a phenotype closely resembling presbycusis, the most common sensory defect associated with aging in humans. We have identified SLC17A8, which encodes the vesicular glutamate transporter-3 (VGLUT3), as the gene responsible for DFNA25, an autosomal-dominant form of progressive, high-frequency nonsyndromic deafness. In two unrelated families, a heterozygous missense mutation, c.632C-->T (p.A211V), was found to segregate with DFNA25 deafness and was not present in 267 controls. Linkage-disequilibrium analysis suggested that the families have a distant common ancestor. The A211 residue is conserved in VGLUT3 across species and in all human VGLUT subtypes (VGLUT1-3), suggesting an important functional role. In the cochlea, VGLUT3 accumulates glutamate in the synaptic vesicles of the sensory inner hair cells (IHCs) before releasing it onto receptors of auditory-nerve terminals. Null mice with a targeted deletion of Slc17a8 exon 2 lacked auditory-nerve responses to acoustic stimuli, although auditory brainstem responses could be elicited by electrical stimuli, and robust otoacoustic emissions were recorded. Ca(2+)-triggered synaptic-vesicle turnover was normal in IHCs of Slc17a8 null mice when probed by membrane capacitance measurements at 2 weeks of age. Later, the number of afferent synapses, spiral ganglion neurons, and lateral efferent endings below sensory IHCs declined. Ribbon synapses remaining by 3 months of age had a normal ultrastructural appearance. We conclude that deafness in Slc17a8-deficient mice is due to a specific defect of vesicular glutamate uptake and release and that VGLUT3 is essential for auditory coding at the IHC synapse.


Assuntos
Sistemas de Transporte de Aminoácidos Acídicos/genética , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Surdez/genética , Células Ciliadas Auditivas/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/genética , Proteínas Vesiculares de Transporte de Glutamato/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/fisiologia , Animais , Mapeamento Cromossômico , Modelos Animais de Doenças , Genoma , Humanos , Desequilíbrio de Ligação , Camundongos , Camundongos Knockout , Modelos Genéticos , Mutação , Polimorfismo de Nucleotídeo Único , Proteínas Vesiculares de Transporte de Glutamato/fisiologia
15.
J Neurosci ; 28(29): 7313-23, 2008 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-18632935

RESUMO

Currently, many millions of people treated for various ailments receive high doses of salicylate. Consequently, understanding the mechanisms by which salicylate induces tinnitus is an important issue for the research community. Behavioral testing in rats have shown that tinnitus induced by salicylate or mefenamate (both cyclooxygenase blockers) are mediated by cochlear NMDA receptors. Here we report that the synapses between the sensory inner hair cells and the dendrites of the cochlear spiral ganglion neurons express NMDA receptors. Patch-clamp recordings and two-photon calcium imaging demonstrated that salicylate and arachidonate (a substrate of cyclooxygenase) enabled the calcium flux and the neural excitatory effects of NMDA on cochlear spiral ganglion neurons. Salicylate also increased the arachidonate content of the whole cochlea in vivo. Single-unit recordings of auditory nerve fibers in adult guinea pig confirmed the neural excitatory effect of salicylate and the blockade of this effect by NMDA antagonist. These results suggest that salicylate inhibits cochlear cyclooxygenase, which increased levels of arachidonate. The increased levels of arachidonate then act on NMDA receptors to enable NMDA responses to glutamate that inner hair cells spontaneously release. This new pharmacological profile of salicylate provides a molecular mechanism for the generation of tinnitus at the periphery of the auditory system.


Assuntos
Ácido Araquidônico/fisiologia , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Ácido Salicílico/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Ácido Araquidônico/metabolismo , Ácido Araquidônico/toxicidade , Cóclea/ultraestrutura , Ácido Glutâmico/farmacologia , Cobaias , Oxirredução/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/ultraestrutura , Ácido Salicílico/efeitos adversos , Zumbido/induzido quimicamente , Zumbido/metabolismo , Zumbido/fisiopatologia
16.
Hear Res ; 227(1-2): 19-27, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17079104

RESUMO

This report summarizes recent neuropharmacological data at the IHC afferent/efferent synaptic complex: the type of Glu receptors and transporter involved and the modulation of this fast synaptic transmission by the lateral efferents. Neuropharmacological data were obtained by coupling the recording of cochlear potentials and single unit of the auditory nerve with intra-cochlear applications of drugs (multi-barrel pipette). We also describe the IHC afferent/efferent functioning in pathological conditions. After acoustic trauma or ischemia, acute disruption of IHC-auditory dendrite synapses are seen. However, a re-growth of the nerve fibres and a re-afferentation of the IHC were completely done 5 days after injury. During this synaptic repair, multiple presynaptic bodies were commonly found, either linked to the membrane or "floating" in ectopic positions. In the meantime, the lateral efferents directly contact the IHCs. The demonstration that NMDA receptors blockade delayed the re-growth of neurites suggests a neurotrophic role of NMDA receptors in pathological conditions.


Assuntos
Nervo Coclear/metabolismo , Otopatias/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Plasticidade Neuronal , Regeneração , Sinapses/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Nervo Coclear/efeitos dos fármacos , Otopatias/induzido quimicamente , Otopatias/patologia , Otopatias/fisiopatologia , Potenciais Evocados Auditivos/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Humanos , Receptores de AMPA/metabolismo , Sinapses/efeitos dos fármacos , Fatores de Tempo
17.
Mol Pharmacol ; 71(3): 654-66, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17132689

RESUMO

We tested and characterized the therapeutic value of round window membrane-delivered (RWM) d-JNKI-1 peptide (Bonny et al., 2001) against sound trauma-induced hearing loss. Morphological characteristics of sound-damaged hair cell nuclei labeled by Hoechst staining show that apoptosis is the predominant mode of cell death after sound trauma. Analysis of the events occurring after sound trauma demonstrates that c-Jun N-terminal kinase (JNK)/stress-activated protein kinase activates a mitochondrial cell death pathway (i.e., activation of Bax, release of cytochrome c, activation of procaspases, and cleavage of fodrin). Fluorescein isothiocyanate (FITC)-conjugated d-JNKI-1 peptide applied onto an intact cochlear RWM diffuses through this membrane and penetrates cochlear tissues with the exception of the stria vascularis. A time sequence of fluorescence measurements demonstrates that FITC-labeled d-JNKI-1 remains in cochlear tissues for as long as 3 weeks. In addition to blocking JNK-mediated activation of a mitochondrial cell death pathway, RWM-delivered d-JNKI-1 prevents hair cell death and development of a permanent shift in hearing threshold that is caused by sound trauma in a dose-dependent manner (EC50 = 2.05 microM). The therapeutic window for protection of the cochlea from sound trauma with RWM delivery of d-JNKI-1 extended out to 12 h after sound exposure. These results show that the mitogen-activated protein kinase/JNK signaling pathway plays a crucial role in sound trauma-initiated hair cell death. Blocking this signaling pathway with RWM delivery of d-JNKI-1 may have significant therapeutic value as a therapeutic intervention to protect the human cochlea from the effects of sound trauma.


Assuntos
Apoptose/efeitos dos fármacos , Perda Auditiva Provocada por Ruído/tratamento farmacológico , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Mitocôndrias/patologia , Peptídeos/administração & dosagem , Animais , Proteínas de Transporte/metabolismo , Caspases/fisiologia , Citocromos c/metabolismo , Cobaias , Células Ciliadas Auditivas/patologia , Células Ciliadas Auditivas/ultraestrutura , Perda Auditiva Provocada por Ruído/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas dos Microfilamentos/metabolismo , Necrose , Peptídeos/farmacologia , Fosforilação , Transporte Proteico , Proteínas Proto-Oncogênicas c-jun/metabolismo , Janela da Cóclea/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
18.
J Neurochem ; 97(1): 190-200, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16524378

RESUMO

Dopamine, a neurotransmitter released by the lateral olivocochlear efferents, has been shown tonically to inhibit the spontaneous and sound-evoked activity of auditory nerve fibres. This permanent inhibition probably requires the presence of an efficient transporter to remove dopamine from the synaptic cleft. Here, we report that the dopamine transporter is located in the lateral efferent fibres both below the inner hair cells and in the inner spiral bundle. Perilymphatic perfusion of the dopamine transporter inhibitors nomifensine and N-[1-(2-benzo[b]thiophenyl)cyclohexyl]piperidine into the cochlea reduced the spontaneous neural noise and the sound-evoked compound action potential of the auditory nerve in a dose-dependent manner, leading to both neural responses being completely abolished. We observed no significant change in cochlear responses generated by sensory hair cells (cochlear microphonic, summating potential, distortion products otoacoustic emissions) or in the endocochlear potential reflecting the functional state of the stria vascularis. This is consistent with a selective action of dopamine transporter inhibitors on auditory nerve activity. Capillary electrophoresis with laser-induced fluorescence (EC-LIF) measurements showed that nomifensine-induced inhibition of auditory nerve responses was due to increased extracellular dopamine levels in the cochlea. Altogether, these results show that the dopamine transporter is essential for maintaining the spontaneous activity of auditory nerve neurones and their responsiveness to sound stimulation.


Assuntos
Nervo Coclear/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Dopamina/metabolismo , Audição/fisiologia , Neurônios Aferentes/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Estimulação Acústica , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Potenciais Microfônicos da Cóclea/efeitos dos fármacos , Potenciais Microfônicos da Cóclea/fisiologia , Nervo Coclear/citologia , Nervo Coclear/efeitos dos fármacos , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Vias Eferentes/efeitos dos fármacos , Vias Eferentes/fisiologia , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Cobaias , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Células Ciliadas Auditivas Internas/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Mecanotransdução Celular/fisiologia , Neurônios Aferentes/efeitos dos fármacos , Perilinfa/efeitos dos fármacos , Perilinfa/metabolismo , Gânglio Espiral da Cóclea/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
19.
Eur J Neurosci ; 22(5): 1109-19, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16176352

RESUMO

Ryanodine receptors (RyRs) are known to contribute to the regulation of free cytosolic calcium concentration. This family of intracellular calcium channels plays a significant role in calcium-induced-calcium-release (CICR), and have been implicated in calcium-dependent processes requiring exquisite spatio-temporal regulation. In order to characterize the importance of these intracellular calcium channels in cochlear physiology, we perfused the guinea pig cochlea with antagonistic concentrations of ryanodine. The distortion products of the cochlear microphonic and the compound action potential of the auditory nerve were reversibly inhibited by ryanodine (IC(50)=27.3 microm, Hill coefficient=1.9), indicating an action at the cochlear amplifier. Single auditory nerve fibre recordings showed that ryanodine slightly increased spontaneous firing rates by 22%, suggesting an excitatory effect of ryanodine. This paradoxical effect could be explained by an inhibitory action of ryanodine on presynaptic BK channels of inner hair cells (IHC). Indeed, perfusing iberiotoxin also increased the spontaneous firing activity of the auditory nerve fibres. Furthermore, whole-cell patch-clamp recordings demonstrated that ryanodine inhibits BK currents at the IHC level. Conversely, immunohistochemistry demonstrated a strong expression of RyR in IHCs and, more particularly, below the cuticular plate where membranous BK channels are highly expressed. Overall, the study demonstrated a key role for RyR and CICR in signal transduction at the IHCs. We therefore propose that coupled RyR--BK channels act to suppress the fast neurotransmission in IHCs.


Assuntos
Cóclea/citologia , Células Ciliadas Auditivas Internas/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Transmissão Sináptica , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Western Blotting/métodos , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Interações Medicamentosas , Estimulação Elétrica/métodos , Potenciais Evocados Auditivos/fisiologia , Cobaias , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Imuno-Histoquímica/métodos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Neurológicos , Técnicas de Patch-Clamp/métodos , Peptídeos/farmacologia , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos
20.
Neuroreport ; 16(10): 1087-90, 2005 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-15973153

RESUMO

Riluzole has been reported to protect against the deleterious effect of cerebral ischemia by blocking glutamatergic neurotransmission. Here, we investigated whether acoustic trauma-induced cochlear excitotoxicity could be attenuated by riluzole. Cumulative intracochlear perfusion of riluzole completely abolished single-nerve fiber activity in the guinea pig cochlea and the compound action potential of the auditory nerve. Guinea pigs treated with riluzole (100 microM) showed significantly less hearing threshold shift than untreated guinea pigs, and presented no sign of dendritic damage in the cochlea observable by electron microscopy. When coapplied with glutamate, riluzole did not prevent glutamate-induced swelling of auditory nerve dendrites, suggesting that the protective effect of riluzole was mediated principally by inhibition of glutamate release from sensory inner hair cells.


Assuntos
Estimulação Acústica/efeitos adversos , Perda Auditiva Provocada por Ruído/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Riluzol/uso terapêutico , Estimulação Acústica/métodos , Doença Aguda , Animais , Cóclea/efeitos dos fármacos , Cóclea/patologia , Cobaias , Perda Auditiva Provocada por Ruído/patologia , Fármacos Neuroprotetores/farmacologia , Ruído/efeitos adversos , Riluzol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA