Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 159
1.
J Neurophysiol ; 127(1): 116-129, 2022 01 01.
Article En | MEDLINE | ID: mdl-34817286

Diverse physiological phenotypes in a neuronal population can broaden the range of computational capabilities within a brain region. The avian cochlear nucleus angularis (NA) contains a heterogeneous population of neurons whose variation in intrinsic properties results in electrophysiological phenotypes with a range of sensitivities to temporally modulated input. The low-threshold potassium conductance (GKLT) is a key feature of neurons involved in fine temporal structure coding for sound localization, but a role for these channels in intensity or spectrotemporal coding has not been established. To determine whether GKLT affects the phenotypical variation and temporal properties of NA neurons, we applied dendrotoxin-I (DTX), a potent antagonist of Kv1-type potassium channels, to chick brain stem slices in vitro during whole cell patch-clamp recordings. We found a cell-type specific subset of NA neurons that was sensitive to DTX: single-spiking NA neurons were most profoundly affected, as well as a subset of tonic-firing neurons. Both tonic I (phasic onset bursting) and tonic II (delayed firing) neurons showed DTX sensitivity in their firing rate and phenotypical firing pattern. Tonic III neurons were unaffected. Spike time reliability and fluctuation sensitivity measured in DTX-sensitive NA neurons was also reduced with DTX. Finally, DTX reduced spike threshold adaptation in these neurons, suggesting that GKLT contributes to the temporal properties that allow coding of rapid changes in the inputs to NA neurons. These results suggest that variation in Kv1 channel expression may be a key factor in functional diversity in the avian cochlear nucleus.NEW & NOTEWORTHY The dendrotoxin-sensitive voltage-gated potassium conductance typically associated with neuronal coincidence detection in the timing pathway for sound localization is demonstrated to affect spiking patterns and temporal input sensitivity in the intensity pathway in the avian auditory brain stem. The Kv1-family channels appear to be present in a subset of cochlear nucleus angularis neurons, regulate spike threshold dynamics underlying high-pass membrane filtering, and contribute to intrinsic firing diversity.


Action Potentials/physiology , Cochlear Nucleus/physiology , Neurons/physiology , Potassium Channel Blockers/pharmacology , Shaker Superfamily of Potassium Channels/metabolism , Action Potentials/drug effects , Animals , Chickens , Cochlear Nucleus/drug effects , Cochlear Nucleus/metabolism , Elapid Venoms/pharmacology , Neurons/drug effects , Patch-Clamp Techniques , Shaker Superfamily of Potassium Channels/drug effects
2.
Physiol Res ; 69(1): 181-190, 2020 02 19.
Article En | MEDLINE | ID: mdl-31852197

To examine whether exposure to sodium salicylate disrupts expression of vesicular glutamate transporter 3 (VGLUT3) and whether the alteration in expression corresponds to increased risk for tinnitus. Rats were treated with saline (control) or sodium salicylate (treated) Rats were examined for tinnitus by monitoring gap-pre-pulse inhibition of the acoustic startle reflex (GPIAS). Auditory brainstem response (ABR) was applied to evaluate hearing function after treatment. Rats were sacrificed after injection to obtain the cochlea, cochlear nucleus (CN), and inferior colliculus (IC) for examination of VGLUT3 expression. No significant differences in hearing thresholds between groups were identified (p>0.05). Tinnitus in sodium salicylate-treated rats was confirmed by GPIAS. VGLUT3 encoded by solute carrier family 17 members 8 (SLC17a8) expression was significantly increased in inner hair cells (IHCs) of the cochlea in treated animals, compared with controls (p<0.01). No significant differences in VGLUT3 expression between groups were found for the cochlear nucleus (CN) or IC (p>0.05). Exposure to sodium salicylate may disrupt SLC17a8 expression in IHCs, leading to alterations that correspond to tinnitus in rats. However, the CN and IC are unaffected by exposure to sodium salicylate, suggesting that enhancement of VGLUT3 expression in IHCs may contribute to the pathogenesis of tinnitus.


Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Hair Cells, Auditory, Inner/drug effects , Sodium Salicylate/adverse effects , Tinnitus/chemically induced , Vesicular Glutamate Transport Proteins/metabolism , Animals , Auditory Threshold/drug effects , Cochlear Nucleus/drug effects , Cochlear Nucleus/metabolism , Hair Cells, Auditory, Inner/metabolism , Inferior Colliculi/drug effects , Inferior Colliculi/metabolism , Male , Rats, Wistar
3.
Neurochem Int ; 124: 31-40, 2019 03.
Article En | MEDLINE | ID: mdl-30578839

Presbycusis has become a common sensory deficit in humans. Oxidative damage to mitochondrial DNA and mitochondrial dysfunction is strongly associated with the aging of the auditory system. A previous study established a mimetic rat model of aging using D-galactose (D-gal) and first reported that NADPH oxidase-dependent mitochondrial oxidative damage and apoptosis in the ventral cochlear nucleus (VCN) might contribute to D-gal-induced central presbycusis. In this study, we investigated the effects of apocynin, an NADPH oxidase inhibitor, on mitochondrial dysfunction and mitochondria-dependent apoptosis in the VCN of D-gal-induced aging model in rats. Our data showed that apocynin decreased NADPH oxidase activity, H2O2 levels, mitochondrial DNA common deletion, and 8-hydroxy-2-deoxyguanosine (8-OHdG) expression and increased total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px) activity in the VCN of D-gal-induced aging model in rats. Moreover, apocynin also decreased the protein levels of phospho-p47phox (p-p47phox), tumor necrosis factor alpha (TNFα), and uncoupling protein 2 (UCP2) in the VCN of D-gal-induced aging model in rats. Meanwhile, apocynin alleviated mitochondrial ultrastructure damage and enhanced ATP production and mitochondrial membrane potential (MMP) levels in the VCN of D-gal-induced aging model in rats. Furthermore, apocynin inhibited cytochrome c (Cyt c) translocation from mitochondria to the cytoplasm and suppressed caspase 3-dependent apoptosis in the VCN of D-gal-induced aging model in rats. Consequently, our findings suggest that neuronal survival promoted by an NADPH oxidase inhibitor is a potentially effective method to enhance the resistance of neurons to central presbycusis.


Acetophenones/pharmacology , Aging/drug effects , Cochlear Nucleus/drug effects , Galactose/toxicity , Mitochondria/drug effects , NADPH Oxidases/antagonists & inhibitors , Aging/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cochlear Nucleus/metabolism , Enzyme Inhibitors/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mitochondria/metabolism , NADPH Oxidases/metabolism , Rats , Rats, Sprague-Dawley
4.
J Membr Biol ; 251(5-6): 711-722, 2018 12.
Article En | MEDLINE | ID: mdl-30206647

ERG (ether-a-go-go-related gene) channels are the members of the voltage-dependent potassium channel family, which have three subtypes, as ERG1 (Kv 11.1), ERG2 (Kv 11.2), and ERG3 (Kv11.3). There is no information on ERG channels in the cochlear nucleus (CN) neurons, which is the first relay station of the auditory pathway. As occur in some of congenital long QT Syndromes (LQTS), mutation of the KCNQ11 genes for ERG channel has been reported to be accompanied by hearing loss. For that reason, we aimed to study biophysical properties and physiological importance, and contribution of ERG K+ currents to the formation of action potentials in the stellate and bushy neurons of the ventral cochlear nucleus (VCN). A total of 70 mice at 14-17 days old were used for this study. Electrophysiological characterization of ERG channels was performed using patch-clamp technique in the CN slices. In current clamp, ERG channel blockers, terfenadine (10 µM) and E-4031 (10 µM), were applied in both cell types. The activation, inactivation, and deactivation kinetics of the ERG channels were determined by voltage clamp. In conclusion, the findings obtained in the present study suggest that stellate and bushy neurons express ERG channels and ERG channels appear to contribute to setting action potential (AP) frequency, threshold for AP induction, and, possibly, resting membrane potentials in this cells.


Cochlear Nucleus/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Auditory Pathways/drug effects , Auditory Pathways/physiology , Cochlear Nucleus/drug effects , Electrophysiology , Ether-A-Go-Go Potassium Channels/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Piperidines/pharmacology , Pyridines/pharmacology , Stellate Ganglion/drug effects , Stellate Ganglion/metabolism , Terfenadine/pharmacology
5.
Hear Res ; 367: 97-105, 2018 09.
Article En | MEDLINE | ID: mdl-30081246

The responses of guinea pig cochlear nucleus neurons to in vivo iontophoretic application of various neurotransmitter agonists were recorded with extracellular multi-barrelled electrodes. Where possible, neurons were physiologically identified using strict criteria. Emphasis was placed on the action of cholinergic agonists in relation to the possible action of olivocochlear collateral innervation. Excitatory responses (increase in action potential firing) to glutamate were confirmed in a number of neuronal response types. Application of acetylcholine (ACh) or the broad spectrum cholinergic agonist carbachol produced reliable excitatory responses in about 47% of neurons (n = 29 out of 61 neurons). The remaining neurons were unresponsive to cholinergic agonists and no inhibitory responses were observed. Cholinergic responses were more common in dorsal cochlear nucleus (DCN) (73% of 30 neurons tested) than in ventral cochlear nucleus (VCN) (23% of 31 neurons). Of the total neuron sample in which cholinergic responses were investigated, 41 neurons were able to be categorized according to established acoustic response features. Excitatory responses to cholinergic agonists were seen in "Pauser-buildup" (Pb) and "Transient chopper" (Ct) response types. Primary-like neurons (PL and Pn) as well as "Onset chopper" (Oc) neurons (n = 6) were unresponsive to either ACh or carbachol. Oc neurons also did not show any effect on their acoustic responses. Robust cholinergic responses were also seen in several VCN and DCN neurons that were either unresponsive to sound, or had acoustic response properties that did not fit standard classification. The results suggest a relatively more robust cholinergic innervation of DCN compared to VCN. The excitatory cholinergic responses of some Ct neurons and the lack of effect on Oc neurons are consistent with previous results in mouse brain slice studies, but are in conflict with reports of medial olivocochlear collateral excitatory responses in onset-type neurons in vivo. The results also indicate that a number of neurons of unknown identity may also receive cholinergic input.


Acetylcholine/administration & dosage , Action Potentials/drug effects , Carbachol/administration & dosage , Cholinergic Agonists/administration & dosage , Cholinergic Neurons/drug effects , Cochlear Nucleus/drug effects , Acoustic Stimulation , Animals , Cholinergic Neurons/metabolism , Cochlear Nucleus/metabolism , Evoked Potentials, Auditory, Brain Stem , Guinea Pigs , Iontophoresis , Time Factors
6.
Hear Res ; 361: 36-44, 2018 04.
Article En | MEDLINE | ID: mdl-29453003

The purpose of this study was to test whether a Kv3 potassium channel modulator, AUT00063, has therapeutic potential for reversing noise-induced increases in spontaneous neural activity, a state that is widely believed to underlie noise-induced tinnitus. Recordings were conducted in noise exposed and control hamsters from dorsal cochlear nucleus (DCN) fusiform cells before and following intraperitoneal administration of AUT00063 (30 mg/kg). Fusiform cell spontaneous activity was increased in sound-exposed animals, approximating levels that were nearly 50% above those of controls. Administration of AUT00063 resulted in a powerful suppression of this hyperactivity. The first signs of this suppression began 13 min after AUT00063 administration, but activity continued to decline gradually until reaching a floor level which was approximately 60% of pre-drug baseline by 25 min after drug treatment. A similar suppressive effect of AUT00063 was observed in control animals, with onset of suppression first apparent at 13 min post-treatment, but continuing to decline toward a floor level that was 54% of pre-drug baseline and was reached 28 min after drug treatment. In contrast, no suppression of spontaneous activity was observed in animals given similar injections of vehicle (control) solution. The suppressive effect of AUT00063 was achieved without significantly altering heart rate and with minimal effects on response thresholds, supporting the interpretation that the reductions of hyperactivity were not a secondary consequence of a more general physiological suppression of the brain or auditory system. These findings suggest that Kv3 channel modulation may be an effective approach to suppressing spontaneous activity in the auditory system and may provide a future avenue for treatment of tinnitus resulting from exposure to intense sound.


Cochlear Nucleus/drug effects , Imidazoles/pharmacology , Membrane Transport Modulators/pharmacology , Noise/adverse effects , Pyrimidines/pharmacology , Shaw Potassium Channels/drug effects , Animals , Auditory Threshold/drug effects , Cochlear Nucleus/metabolism , Evoked Potentials, Auditory, Brain Stem/drug effects , Imidazoles/therapeutic use , Male , Mesocricetus , Pyrimidines/therapeutic use , Shaw Potassium Channels/metabolism , Time Factors , Tinnitus/drug therapy
7.
Neuroscience ; 376: 72-79, 2018 04 15.
Article En | MEDLINE | ID: mdl-29462704

The Dorsal Cochlear Nucleus (DCN) is a region which has been traditionally linked to the genesis of tinnitus, the constant perception of a phantom sound. Sodium salicylate, a COX-2 inhibitor, can induce tinnitus in high doses. Hyperactivity of DCN neurons is observed in several animal models of tinnitus, including salicylate-induced tinnitus. The DCN presents several forms of endocannabinoid (EC)-dependent synaptic plasticity and COX-2 can also participate in the oxidative degradation of ECs. We recently demonstrated that short-term perfusion of sodium salicylate and other inhibitors of both oxidative and hydrolytic EC degradation did not affect depolarization-induced suppression of excitation (DSE), a form of EC-dependent short-term synaptic plasticity. Here, we show that prolonged incubation with high doses of sodium salicylate (1.4 mM) enhances DSE of synapses onto glycinergic DCN interneurons but not those innervating glutamatergic DCN fusiform neurons. This effect was not reproduced with lower doses of salicylate (140 µM) or with ibuprofen, another inhibitor of COX-2. This effect was not observed in the presence of AM251, an antagonist/inverse agonist of cannabinoid CB1 receptors, showing that it was dependent on EC release. Finally we demonstrated that incubation with salicylate potentiated the increase in intracellular calcium during the depolarization. Our results point to an increased inhibition of DCN inhibitory CW neuron during depolarizations, probably by an enhanced EC release during the depolarizations, which is potentially significant for DCN hyperactivity and tinnitus generation.


Cyclooxygenase Inhibitors/adverse effects , Endocannabinoids/metabolism , Glycine/metabolism , Neurons/drug effects , Sodium Salicylate/adverse effects , Animals , Calcium/metabolism , Cochlear Nucleus/drug effects , Cochlear Nucleus/metabolism , Dose-Response Relationship, Drug , Ibuprofen/pharmacology , Male , Neurons/metabolism , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Synapses/drug effects , Synapses/metabolism , Tinnitus/chemically induced , Tinnitus/metabolism , Tissue Culture Techniques
8.
Brain Res ; 1682: 71-77, 2018 03 01.
Article En | MEDLINE | ID: mdl-29329983

It has previously been hypothesized that hyperactivity of central auditory neurons following exposure to intense noise is a consequence of synaptic alterations. Recent studies suggest the involvement of NMDA receptors in the induction of this hyperactive state. NMDA receptors can mediate long term changes in the excitability of neurons through their involvement in excitotoxic injury and long term potentiation and depression. In this study, we examined the effect of administering an NMDA receptor blocker on the induction of hyperactivity in the dorsal cochlear nucleus (DCN) following intense sound exposure. Our prediction was that if hyperactivity induced by intense sound exposure is dependent on NMDA receptors, then blocking these receptors by administering an NMDA receptor antagonist just before animals are exposed to intense sound should reduce the degree of hyperactivity that subsequently emerges. We compared the levels of hyperactivity that develop in the DCN after intense sound exposure to activity recorded in control animals that were not sound exposed. One group of animals to be sound exposed received intraperitoneal injection of MK-801 twenty minutes preceding the sound exposure, while the other group received injection of saline. Recordings performed in the DCN 26-28 days post-exposure revealed increased response thresholds and widespread increases in spontaneous activity in the saline-treated animals that had been sound exposed, consistent with earlier studies. The animals treated with MK-801 preceding sound exposure showed similarly elevated thresholds but an attenuation of hyperactivity in the DCN; the attenuation was most robust in the high frequency half of the DCN, but lower levels of hyperactivity were also found in the low frequency half. These findings suggest that NMDA receptors are an important component of the hyperactivity-inducing mechanism following intense sound exposure. They further suggest that blockade of NMDA receptors may offer a useful therapeutic approach to preventing induction of noise-induced hyperactivity-related hearing disorders, such as tinnitus and hyperacusis.


Cochlear Nucleus/drug effects , Dizocilpine Maleate/therapeutic use , Hyperkinesis/etiology , Hyperkinesis/prevention & control , Neuroprotective Agents/therapeutic use , Noise/adverse effects , Acoustic Stimulation , Animals , Cochlear Nucleus/physiology , Cricetinae , Disease Models, Animal , Drug Administration Schedule , Mesocricetus
9.
J Membr Biol ; 251(1): 163-178, 2018 02.
Article En | MEDLINE | ID: mdl-29379989

Major voltage-activated ionic channels of stellate cells in the ventral part of cochlear nucleus (CN) were largely characterized previously. However, it is not known if these cells are equipped with other ion channels apart from the voltage-sensitive ones. In the current study, it was aimed to study subunit composition and function of ATP-sensitive potassium channels (KATP) in stellate cells of the ventral cochlear nucleus. Subunits of KATP channels, Kir6.1, Kir6.2, SUR1, and SUR2, were expressed at the mRNA level and at the protein level in the mouse VCN tissue. The specific and clearly visible bands for all subunits but that for Kir6.1 were seen in Western blot. Using immunohistochemical staining technique, stellate cells were strongly labeled with SUR1 and Kir6.2 antibodies and moderately labeled with SUR2 antibody, whereas the labeling signals for Kir6.1 were too weak. In patch clamp recordings, KATP agonists including cromakalim (50 µM), diazoxide (0.2 mM), 3-Amino-1,2,4-triazole (ATZ) (1 mM), 2,2-Dithiobis (5-nitro pyridine) (DTNP) (330 µM), 6-Chloro-3-isopropylamino- 4H-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide (NNC 55-0118) (1 µM), 6-chloro-3-(methylcyclopropyl)amino-4H-thieno[3,2-e]-1,2,4-thiadiazine 1,1-dioxide (NN414) (1 µM), and H2O2 (0.88 mM) induced marked responses in stellate cells, characterized by membrane hyperpolarization which were blocked by KATP antagonists. Blockers of KATP channels, glibenclamide (0.2 mM), tolbutamide (0.1 mM) as well as 5-hydroxydecanoic acid (1 mM), and catalase (500 IU/ml) caused depolarization of stellate cells, increasing spontaneous action potential firing. In conclusion, KATP channels seemed to be composed dominantly of Kir 6.2 subunit and SUR1 and SUR2 and activation or inhibition of KATP channels regulates firing properties of stellate cells by means of influencing resting membrane potential and input resistance.


Cochlear Nucleus/drug effects , Cochlear Nucleus/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclic S-Oxides/pharmacology , Diazoxide/analogs & derivatives , Diazoxide/pharmacology , Hydrogen Peroxide , KATP Channels/agonists , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Mice , Mice, Inbred BALB C , Potassium Channels, Inwardly Rectifying/agonists , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Tolbutamide/pharmacology
10.
J Neurosurg ; 128(1): 296-303, 2018 01.
Article En | MEDLINE | ID: mdl-28298014

OBJECTIVE The risk of injury of the cochlear nerve during angle (CPA) surgery is high. Granulocyte colony-stimulating factor (G-CSF) has been found in various experimental models of peripheral and CNS injury to have a neuroprotective effect by inhibiting apoptosis and inflammation. However, to the authors' knowledge, the influence of G-CSF on cochlear nerve regeneration has not been reported. This study investigated the neuroprotective effect of G-CSF after a partial cochlear nerve lesion in rats. METHODS A lesion of the right cochlear nerve in adult male Sprague-Dawley rats was created using a water-jet dissector with a pressure of 8 bar. In the first group (G-CSF-post), G-CSF was administrated on Days 1, 3, and 5 after the surgery. The second group (G-CSF-pre/post) was treated with G-CSF 1 day before and 1, 3, and 5 days after applying the nerve injury. The control group received sodium chloride after nerve injury at the various time points. Brainstem auditory evoked potentials (BAEPs) were measured directly before and after nerve injury and on Days 1 and 7 to evaluate the acoustic function of the cochlear nerve. The animals were sacrificed 1 week after the operation, and their brains were fixed in formalin. Nissl staining of the cochlear nuclei was performed, and histological sections were analyzed with a light microscope and an image-processing program. The numbers of neurons in the cochlear nuclei were assessed. RESULTS The values for Waves 2 and 4 of the BAEPs decreased abruptly in all 3 groups in the direct postoperative measurement. Although the amplitude in the control group did not recover, it increased in both treatment groups. According to 2-way ANOVA, groups treated with G-CSF had a significant increase in BAEP Wave II amplitudes on the right side (p = 0.0401) after the applied cochlear nerve injury. With respect to Wave IV, a trend toward better recovery in the G-CSF groups was found, but this difference did not reach statistical significance. In the histological analysis, higher numbers of neurons were found in the G-CSF groups. In the statistical analysis, the difference in the numbers of neurons between the control and G-CSF-post groups reached significance (p = 0.0086). The difference in the numbers of neurons between the control and G-CSF-pre/post groups and between the G-CSF-post and G-CSF-pre/post groups did not reach statistical significance. CONCLUSIONS The use of G-CSF improved the function of the eighth cranial nerve and protected cochlear nucleus cells from destruction after a controlled partial injury of the nerve. These findings might be relevant for surgery that involves CPA tumors. The use of G-CSF in patients with a lesion in the CPA might improve postoperative outcomes.


Cochlear Nerve/drug effects , Cochlear Nerve/injuries , Cochlear Nucleus/drug effects , Cochlear Nucleus/injuries , Granulocyte Colony-Stimulating Factor/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cochlear Nerve/physiopathology , Cochlear Nucleus/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Male , Random Allocation , Rats, Sprague-Dawley
11.
Neurochem Int ; 114: 1-9, 2018 03.
Article En | MEDLINE | ID: mdl-29248694

Acrolein is a ubiquitous dietary and environmental pollutant, which can also be generated endogenously during cellular stress. However, the molecular mechanisms underlying acrolein-induced neurotoxicity, especially in ototoxicity conditions, have not been fully determined. In this study, we investigated the mechanisms on acrolein-induced toxicity in primary cultured cochlear nucleus neurons with focus on Sirt3, a mitochondrial deacetylase. We found that acrolein treatment induced neuronal injury and programmed cell death (PCD) in a dose dependent manner in cochlear nucleus neurons, which was accompanied by increased intracellular reactive oxygen species (ROS) generation and lipid peroxidation. Acrolein exposure also significantly reduced the mitochondrial membrane potential (MMP) levels, promoted cytochrome c release and decreased mitochondrial ATP production. In addition, increased ER tracker fluorescence and activation of ER stress factors were observed after acrolein treatment, and the ER stress inhibitors were shown to attenuate acrolein-induced toxicity in cochlear nucleus neurons. The results of western blot and RT-PCR showed that acrolein markedly decreased the expression of Sirt3 at both mRNA and protein levels, and reduced the activity of downstream mitochondrial enzymes. Furthermore, overexpression of Sirt3 by lentivirus transfection partially prevented acrolein-induced neuronal injury in cochlear nucleus neurons. These results demonstrated that acrolein induces mitochondrial dysfunction and ER stress in cochlear nucleus neurons, and Sirt3 acts as an endogenous protective factor in acrolein-induced ototoxicity.


Acrolein/toxicity , Cochlear Nucleus/metabolism , Cytoprotection/physiology , Neurons/metabolism , Oxidative Stress/physiology , Sirtuins/biosynthesis , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cochlear Nucleus/drug effects , Cochlear Nucleus/pathology , Cytoprotection/drug effects , Dose-Response Relationship, Drug , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Sirtuins/antagonists & inhibitors
12.
Braz. j. otorhinolaryngol. (Impr.) ; 83(2): 155-161, Mar.-Apr. 2017. graf
Article En | LILACS | ID: biblio-839425

Abstract Introduction: Salicylate at high doses induces tinnitus in humans and experimental animals. However, the mechanisms and loci of action of salicylate in inducing tinnitus are still not well known. The expression of Immediate Early Genes (IEG) is traditionally associated with long-term neuronal modifications but it is still not clear how and where IEGs are activated in animal models of tinnitus. Objectives: Here we investigated the expression of c-fos and Egr-1, two IEGs, in the Dorsal Cochlear Nucleus (DCN), the Inferior Colliculus (IC), and the Posterior Ventral Cochlear Nucleus (pVCN) of rats. Methods: Rats were treated with doses known to induce tinnitus in rats (300 mg/kg i.p. daily, for 3 days), and c-fos and Egr-1 protein expressions were analyzed using western blot and immunocytochemistry. Results: After administration of salicylate, c-fos protein expression increased significantly in the DCN, pVCN and IC when assayed by western blot. Immunohistochemistry staining showed a more intense labeling of c-fos in the DCN, pVCN and IC and a significant increase in c-fos positive nuclei in the pVCN and IC. We did not detect increased Egr-1 expression in any of these areas. Conclusion: Our data show that a high dose of salicylate activates neurons in the DCN, pVCN and IC. The expression of these genes by high doses of salicylate strongly suggests that plastic changes in these areas are involved in the genesis of tinnitus.


Resumo Introdução: Salicilato em doses elevadas induz zumbido nos seres humanos e em animais experimentais. No entanto, os mecanismos e loci de ação do salicilato na indução de zumbido ainda não são bem conhecidos. A expressão dos genes precoces imediatos (GPIs) está tradicionalmente associada a alterações neuronais em longo prazo, mas ainda não está claro como e onde os GPIs são ativados em modelos animais de zumbido. Objetivos: No presente estudo investigamos a expressão de c-fos e Egr-1, dois GPIs, no núcleo coclear dorsal (NCD), colículo inferior (CI) e núcleo coclear ventral posterior (NCVp) de ratos. Métodos: Os ratos foram tratados com doses que, conhecidamente, induzem zumbido em ratos (300 mg/kg IP/dia, por três dias) e as expressões das proteínas c-fos e Egr-1 foram analisadas por meio de Western blot e imunoistoquímica. Resultados: Após a administração de salicilato, a expressão da proteína c-fos aumentou significativamente no NCD, NCVp e CI, quando analisados por Western blot. A coloração imunoistoquímica mostrou uma marcação mais intensa de c-fos no NCD, NCVp e CI e um aumento significativo de núcleos positivos de c-fos no NCVp e CI. Não detectamos aumento da expressão de Egr-1 em qualquer dessas áreas. Conclusão: Nossos dados mostram que uma dose alta de salicilato ativa neurônios no NCD, NCVp e CI. A expressão desses genes por doses altas de salicilato sugere que as alterações plásticas nessas áreas estão envolvidas na gênese do zumbido.


Animals , Male , Rats , Inferior Colliculi/drug effects , Salicylates/pharmacology , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Cochlear Nucleus/drug effects , Salicylates/administration & dosage , Blotting, Western , Genes, fos/drug effects , Rats, Wistar , Dose-Response Relationship, Drug , Early Growth Response Protein 1/drug effects
13.
Hear Res ; 345: 57-68, 2017 03.
Article En | MEDLINE | ID: mdl-28065805

Low-voltage-activated K+ (gKL) and hyperpolarization-activated mixed cation conductances (gh) mediate currents, IKL and Ih, through channels of the Kv1 (KCNA) and HCN families respectively and give auditory neurons the temporal precision required for signaling information about the onset, fine structure, and time of arrival of sounds. Being partially activated at rest, gKL and gh contribute to the resting potential and shape responses to even small subthreshold synaptic currents. Resting gKL and gh also affect the coupling of somatic depolarization with the generation of action potentials. To learn how these important conductances are regulated we have investigated how genetic perturbations affect their expression in octopus cells of the ventral cochlear nucleus (VCN). We report five new findings: First, the magnitude of gh and gKL varied over more than two-fold between wild type strains of mice. Second, average resting potentials are not different in different strains of mice even in the face of large differences in average gKL and gh. Third, IKL has two components, one being α-dendrotoxin (α-DTX)-sensitive and partially inactivating and the other being α-DTX-insensitive, tetraethylammonium (TEA)-sensitive, and non-inactivating. Fourth, the loss of Kv1.1 results in diminution of the α-DTX-sensitive IKL, and compensatory increased expression of an α-DTX-insensitive, tetraethylammonium (TEA)-sensitive IKL. Fifth, Ih and IKL are balanced at the resting potential in all wild type and mutant octopus cells even when resting potentials vary in individual cells over nearly 10 mV, indicating that the resting potential influences the expression of gh and gKL. The independence of resting potentials on gKL and gh shows that gKL and gh do not, over days or weeks, determine the resting potential but rather that the resting potential plays a role in regulating the magnitude of either or both gKL and gh.


Auditory Pathways/metabolism , Cochlear Nucleus/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Kv1.1 Potassium Channel/genetics , Membrane Potentials , Potassium Channels/genetics , Animals , Auditory Pathways/cytology , Auditory Pathways/drug effects , Cochlear Nucleus/cytology , Cochlear Nucleus/drug effects , Gene Expression Regulation , Genotype , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/deficiency , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.1 Potassium Channel/deficiency , Membrane Potentials/drug effects , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Neuronal Plasticity , Patch-Clamp Techniques , Phenotype , Potassium Channel Blockers/pharmacology , Potassium Channels/deficiency , Time Factors
14.
Braz J Otorhinolaryngol ; 83(2): 155-161, 2017.
Article En | MEDLINE | ID: mdl-27174774

INTRODUCTION: Salicylate at high doses induces tinnitus in humans and experimental animals. However, the mechanisms and loci of action of salicylate in inducing tinnitus are still not well known. The expression of Immediate Early Genes (IEG) is traditionally associated with long-term neuronal modifications but it is still not clear how and where IEGs are activated in animal models of tinnitus. OBJECTIVES: Here we investigated the expression of c-fos and Egr-1, two IEGs, in the Dorsal Cochlear Nucleus (DCN), the Inferior Colliculus (IC), and the Posterior Ventral Cochlear Nucleus (pVCN) of rats. METHODS: Rats were treated with doses known to induce tinnitus in rats (300mg/kg i.p. daily, for 3 days), and c-fos and Egr-1 protein expressions were analyzed using western blot and immunocytochemistry. RESULTS: After administration of salicylate, c-fos protein expression increased significantly in the DCN, pVCN and IC when assayed by western blot. Immunohistochemistry staining showed a more intense labeling of c-fos in the DCN, pVCN and IC and a significant increase in c-fos positive nuclei in the pVCN and IC. We did not detect increased Egr-1 expression in any of these areas. CONCLUSION: Our data show that a high dose of salicylate activates neurons in the DCN, pVCN and IC. The expression of these genes by high doses of salicylate strongly suggests that plastic changes in these areas are involved in the genesis of tinnitus.


Cochlear Nucleus/drug effects , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Inferior Colliculi/drug effects , Salicylates/pharmacology , Animals , Blotting, Western , Dose-Response Relationship, Drug , Early Growth Response Protein 1/drug effects , Genes, fos/drug effects , Male , Rats , Rats, Wistar , Salicylates/administration & dosage
15.
Synapse ; 71(4)2017 04.
Article En | MEDLINE | ID: mdl-27997730

Neurons from the dorsal cochlear nucleus (DCN) present endocannabinoid (EC) dependent short-term synaptic plasticity in the form of depolarization-induced suppression of excitation (DSE). Postsynaptic calcium influx promotes EC synthesis and depression of neurotransmission. ECs can be degraded by a hydrolytic and an oxidative pathway, the latter via the enzyme cyclooxygenase 2 (COX-2). Hyperactivity in the DCN is related to the development of tinnitus, which can be induced by high doses of salicylate, a COX-2 inhibitor. Since EC-dependent plasticity in the DCN can affect its excitation-inhibition balance, we investigated the impact of inhibitors of both oxidative and hydrolytic EC metabolism on the DSE from the synapses between the parallel fibers and cartwheel neurons (PF-CW) in the DCN. We found that inhibitors of COX-2 (ibuprofen and indomethacin) did not alter DSE at the PF-CW synapse. Salicylate also did not alter DSE. However, we found that inhibitors of the hydrolytic pathway did not affect DSE magnitude, but surprisingly speeded DSE decay. We conclude that oxidative EC degradation in the PF-CW synapse is not relevant for termination of DSE and are probably not important for controlling this form of synaptic plasticity in the DCN PF-CW synapse. The lack of effect on DSE of high doses of salicylate also suggests that it is not acting by increasing DSE in the PF-CWC synapse. However, the counter intuitive effect of the hydrolytic inhibitors shows that increasing EC on this synapse have more complex effects on DSE.


Cochlear Nucleus/drug effects , Cyclooxygenase Inhibitors/pharmacology , Endocannabinoids/metabolism , Neuronal Plasticity , Neurons/drug effects , Synaptic Potentials , Animals , Calcium/metabolism , Cochlear Nucleus/cytology , Cochlear Nucleus/metabolism , Cochlear Nucleus/physiology , Cyclooxygenase 2/metabolism , Glycine/metabolism , Ibuprofen/pharmacology , Indomethacin/pharmacology , Male , Neurons/metabolism , Neurons/physiology , Rats , Rats, Wistar , Salicylates/pharmacology
16.
Hear Res ; 344: 13-23, 2017 02.
Article En | MEDLINE | ID: mdl-27838373

The neurochemical serotonin (5-hydroxytryptamine, 5-HT) is involved in a variety of behavioral functions including arousal, reward, and attention, and has a role in several complex disorders of the brain. In the auditory system, 5-HT fibers innervate a number of subcortical nuclei, yet the modulatory role of 5-HT in nearly all of these areas remains poorly understood. In this study, we examined spiking activity of neurons in the dorsal cochlear nucleus (DCN) following iontophoretic application of 5-HT. The DCN is an early site in the auditory pathway that receives dense 5-HT fiber input from the raphe nuclei and has been implicated in the generation of auditory disorders marked by neuronal hyperexcitability. Recordings from the DCN in awake mice demonstrated that iontophoretic application of 5-HT had heterogeneous effects on spiking rate, spike timing, and evoked spiking threshold. We found that 56% of neurons exhibited increases in spiking rate during 5-HT delivery, while 22% had decreases in rate and the remaining neurons had no change. These changes were similar for spontaneous and evoked spiking and were typically accompanied by changes in spike timing. Spiking increases were associated with lower first spike latencies and jitter, while decreases in spiking generally had opposing effects on spike timing. Cases in which 5-HT application resulted in increased spiking also exhibited lower thresholds compared to the control condition, while cases of decreased spiking had no threshold change. We also found that the 5-HT2 receptor subtype likely has a role in mediating increased excitability. Our results demonstrate that 5-HT can modulate activity in the DCN of awake animals and that it primarily acts to increase neuronal excitability, in contrast to other auditory regions where it largely has a suppressive role. Modulation of DCN function by 5-HT has implications for auditory processing in both normal hearing and disordered states.


Auditory Perception/drug effects , Behavior, Animal/drug effects , Cochlear Nucleus/drug effects , Receptors, Serotonin, 5-HT2/drug effects , Serotonergic Neurons/drug effects , Serotonin 5-HT2 Receptor Agonists/administration & dosage , Serotonin/administration & dosage , Acoustic Stimulation , Animals , Cochlear Nucleus/metabolism , Electroencephalography , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Iontophoresis , Male , Mice, Inbred CBA , Reaction Time/drug effects , Receptors, Serotonin, 5-HT2/metabolism , Serotonergic Neurons/metabolism , Serotonin/metabolism , Time Factors
17.
Hear Res ; 342: 134-143, 2016 12.
Article En | MEDLINE | ID: mdl-27773647

Many previous studies have shown significant neurotrophic effects of intracochlear delivery of BDNF in preventing degeneration of cochlear spiral ganglion (SG) neurons after deafness in rodents and our laboratory has shown similar results in developing cats deafened prior to hearing onset. This study examined the morphology of the cochlear nucleus (CN) in a group of neonatally deafened cats from a previous study in which infusion of BDNF elicited a significant improvement in survival of the SG neurons. Five cats were deafened by systemic injections of neomycin sulfate (60 mg/kg, SQ, SID) starting one day after birth, and continuing for 16-18 days until auditory brainstem response (ABR) testing demonstrated profound bilateral hearing loss. The animals were implanted unilaterally at about 1 month of age using custom-designed electrodes with a drug-delivery cannula connected to an osmotic pump. BDNF (94 µg/ml; 0.25 µl/hr) was delivered for 10 weeks. The animals were euthanized and studied at 14-23 weeks of age. Consistent with the neurotrophic effects of BDNF on SG survival, the total CN volume in these animals was significantly larger on the BDNF-treated side than on the contralateral side. However, total CN volume, both ipsi- and contralateral to the implants in these deafened juvenile animals, was markedly smaller than the CN in normal adult animals, reflecting the severe effects of deafness on the central auditory system during development. Data from the individual major CN subdivisions (DCN, Dorsal Cochlear Nucleus; PVCN, Posteroventral Cochlear Nucleus; AVCN, Anteroventral Cochlear Nucleus) also were analyzed. A significant difference was observed between the BDNF-treated and control sides only in the AVCN. Measurements of the cross-sectional areas of spherical cells showed that cells were significantly larger in the AVCN ipsilateral to the implant than on the contralateral side. Further, the numerical density of spherical cells was significantly lower in the AVCN ipsilateral to the implant than on the contralateral side, consistent with the larger AVCN volume observed with BDNF treatment. Together, findings indicate significant neurotrophic effects of intracochlear BDNF infusion on the developing CN.


Brain-Derived Neurotrophic Factor/administration & dosage , Cochlear Nucleus/drug effects , Cochlear Nucleus/pathology , Deafness/drug therapy , Deafness/pathology , Animals , Animals, Newborn , Cats , Cochlear Nucleus/growth & development , Deafness/chemically induced , Drug Delivery Systems , Evoked Potentials, Auditory, Brain Stem , Neomycin/toxicity , Organ Size/drug effects , Spiral Ganglion/drug effects , Spiral Ganglion/pathology
18.
Neurochem Res ; 41(6): 1343-53, 2016 Jun.
Article En | MEDLINE | ID: mdl-26886762

Lesion-induced cochlear damage can result in synaptic outgrowth in the ventral cochlear nucleus (VCN). Tinnitus may be associated with the synaptic outgrowth and hyperactivity in the VCN. However, it remains unclear how hearing loss triggers structural synaptic modifications in the VCN of rats subjected to salicylate-induced tinnitus. To address this issue, we evaluated tinnitus-like behavior in rats after salicylate treatment and compared the amplitude of the distortion product evoked otoacoustic emission (DPOAE) and auditory brainstem response (ABR) between control and treated rats. Moreover, we observed the changes in the synaptic ultrastructure and in the expression levels of growth-associated protein (GAP-43), brain-derived neurotrophic factor (BDNF), the microglial marker Iba-1 and glial fibrillary acidic protein (GFAP) in the VCN. After salicylate treatment (300 mg/kg/day for 4 and 8 days), analysis of the gap prepulse inhibition of the acoustic startle showed that the rats were experiencing tinnitus. The changes in the DPOAE and ABR amplitude indicated an improvement in cochlear sensitivity and a reduction in auditory input following salicylate treatment. The treated rats displayed more synaptic vesicles and longer postsynaptic density in the VCN than the control rats. We observed that the GAP-43 expression, predominantly from medial olivocochlear (MOC) neurons, was significantly up-regulated, and that BDNF- and Iba-1-immunoreactive cells were persistently decreased after salicylate administration. Furthermore, GFAP-immunoreactive astrocytes, which is associated with synaptic regrowth, was significantly increased in the treated groups. Our study revealed that reduced auditory nerve activity triggers synaptic outgrowth and hyperactivity in the VCN via a MOC neural feedback circuit. Structural synaptic modifications may be a reflexive process that compensates for the reduced auditory input after salicylate administration. However, massive increases in excitatory synapses in the VCN may represent a detrimental process that causes central hyperactivity, leading to tinnitus.


Cochlear Nucleus/ultrastructure , Feedback, Physiological , Hearing Loss/chemically induced , Nerve Net/ultrastructure , Salicylates/toxicity , Synapses/ultrastructure , Acoustic Stimulation/methods , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Cochlear Nucleus/drug effects , Cochlear Nucleus/metabolism , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Hearing Loss/metabolism , Hearing Loss/pathology , Male , Nerve Net/drug effects , Nerve Net/metabolism , Random Allocation , Rats , Rats, Wistar , Synapses/drug effects , Synapses/metabolism
19.
Neuroscience ; 316: 201-8, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26739326

Cochlea removal results in the death of 20-30% of neurons in the chick cochlear nucleus, nucleus magnocellularis (NM). Two potentially cytotoxic events, a dramatic rise in intracellular calcium concentration ([Ca(2+)]i) and a decline in the integrity of ribosomes are observed within 1h of deafferentation. Glutamatergic input from the auditory nerve has been shown to preserve NM neuron health by activating metabotropic glutamate receptors (mGluRs), maintaining both normal [Ca(2+)]i and ribosomal integrity. One interpretation of these results is that a common mGluR-activated signaling cascade is required for the maintenance of both [Ca(2+)]i and ribosomal integrity. This could happen if both responses are influenced directly by a common messenger, or if the loss of mGluR activation causes changes in one component that secondarily causes changes in the other. The present studies tested this common-mediator hypothesis in slice preparations by examining activity-dependent regulation of [Ca(2+)]i and ribosomes in the same tissue after selectively blocking group I mGluRs (1-Aminoindan-1,5-dicarboxylic acid (AIDA)) or group II mGluRs (LY 341495) during unilateral auditory nerve stimulation. Changes in [Ca(2+)]i of NM neurons were measured using fura-2 ratiometric calcium imaging and the tissue was subsequently processed for Y10B immunoreactivity (Y10B-ir), an antibody that recognizes a ribosomal epitope. The group I mGluR antagonist blocked the activity-dependent regulation of both [Ca(2+)]i and Y10B-ir, but the group II antagonist blocked only the activity-dependent regulation of Y10B-ir. That is, even when group II receptors were blocked, stimulation continued to maintain low [Ca(2+)]i, but it did not maintain Y10B-ir. These results suggest a dissociation in how calcium and ribosomes are regulated in NM neurons and that ribosomes can be regulated through a mechanism that is independent of calcium regulation.


Calcium/metabolism , Cochlear Nucleus/metabolism , Evoked Potentials, Auditory/physiology , Ribosomes/metabolism , Amino Acids/pharmacology , Analysis of Variance , Animals , Chick Embryo , Cochlear Nucleus/drug effects , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , In Vitro Techniques , Indans/pharmacology , Optical Imaging , Time Factors , Xanthenes/pharmacology
20.
Hear Res ; 332: 188-198, 2016 Feb.
Article En | MEDLINE | ID: mdl-26548740

High doses of salicylate induce reversible tinnitus in experimental animals and humans, and is a common tinnitus model. Salicylate probably acts centrally and induces hyperactivity in specific auditory brainstem areas like the dorsal cochlear nucleus (DCN). However, little is known about the effect of high doses of salicylate in synapses and neurons of the DCN. Here we investigated the effects of salicylate on the excitability and evoked and spontaneous neurotransmission in the main neurons (fusiform, cartwheel and tuberculoventral) and synapses of the DCN using whole cell recordings in slices containing the DCN. For this, we incubate the slices for at least 1 h in solution with 1.4 mM salicylate, and recorded action potentials and evoked and spontaneous synaptic currents in fusiform, cartwheel (CW) and putative tuberculoventral (TBV) neurons. We found that incubation with salicylate did not affect the firing of fusiform and TBV neurons, but decreased the spontaneous firing of cartwheel neurons, without affecting AP threshold or complex spikes. Evoked and spontaneous glutamatergic neurotransmission on the fusiform and CW neurons cells was unaffected by salicylate and evoked glycinergic neurotransmission on fusiform neurons was also unchanged by salicylate. On the other hand spontaneous glycinergic transmission on fusiform neurons was reduced in the presence of salicylate. We conclude that high doses of salicylate produces a decreased inhibitor drive on DCN fusiform neurons by reducing the spontaneous firing of cartwheel neurons, but this effect is not able to increase the excitability of fusiform neurons. So, the mechanisms of salicylate-induced tinnitus are probably more complex than simple changes in the neuronal firing and basal synaptic transmission in the DCN.


Cochlear Nucleus/drug effects , Glycine/metabolism , Neural Inhibition/drug effects , Neurons/drug effects , Sodium Salicylate/toxicity , Synaptic Transmission/drug effects , Tinnitus/chemically induced , Animals , Cochlear Nucleus/metabolism , Cochlear Nucleus/physiopathology , Evoked Potentials, Auditory , In Vitro Techniques , Male , Neurons/metabolism , Rats, Wistar , Tinnitus/metabolism , Tinnitus/physiopathology
...