Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Talanta ; 258: 124404, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-36889190

RESUMEN

Tinnitus is a widespread and serious clinical and social problem. Although oxidative injury has been suggested to be one of pathological mechanisms in auditory cortex, whether this mechanism could be applied to inferior colliculus remains unclear. In this study, we used an online electrochemical system (OECS) integrating in vivo microdialysis with selective electrochemical detector to continuously monitor the dynamics of ascorbate efflux, an index of oxidative injury, in inferior colliculus of living rats during sodium salicylate-induced tinnitus. We found that OECS with a carbon nanotubes (CNTs)-modified electrode as the detector selectively responses to ascorbate, which is free from the interference from sodium salicylate and MK-801 that were used to induce tinnitus animal model and investigate the N-methyl-d-aspartate (NMDA) receptor mediated excitotoxicity, respectively. With the OECS, we found that the extracellular ascorbate level in inferior colliculus significantly increases after salicylate administration and such increase was suppressed by immediate injection of NMDA receptor antagonist MK-801. In addition, we found that salicylate administration significantly increases the spontaneous and sound stimuli evoked neural activity in inferior colliculus and that the increases were inhibited by the injection of MK-801. These results suggest that oxidative injury may occur in inferior colliculus following salicylate-induced tinnitus, which is closely relevant to the NMDA-mediated neuronal excitotoxicity. This information is useful for understanding the neurochemical processes in inferior colliculus involved in tinnitus and its related brain diseases.


Asunto(s)
Colículos Inferiores , Acúfeno , Técnicas Electroquímicas , Modelos Animales de Enfermedad , Salicilatos/toxicidad , Salicilato de Sodio/toxicidad , Maleato de Dizocilpina/farmacología , Acúfeno/inducido químicamente , Acúfeno/dietoterapia , Estrés Oxidativo , Ácido Ascórbico/metabolismo
2.
Environ Sci Pollut Res Int ; 29(60): 89954-89968, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35859240

RESUMEN

This study aimed to investigate the effectiveness of curcumin (CCM) against gentamicin (GEN) and sodium salicylates (NaS)-induced ototoxic effects in rats. For 15 consecutive days, seven rat groups were given 1 mL/rat physiological saline orally, 1 mL/rat olive oil orally, 50 mg/kg bwt CCM orally, 120 mg/kg bwt GEN intraperitoneally, 300 mg/kg bwt NaS intraperitoneally, CCM+GEN, or CCM+NaS. The distortion product otoacoustic emission measurements were conducted. The rats' hearing function and balance have been behaviorally assessed using auditory startle response, Preyer reflex, and beam balance scale tests. The serum lipid peroxidation and oxidative stress biomarkers have been measured. Immunohistochemical investigations of the apoptotic marker caspase-3 and the inflammatory indicator nuclear factor kappa (NF-κB) in cochlear tissues were conducted. GEN and NaS exposure resulted in deficit hearing and impaired ability to retain balance. GEN and NaS exposure significantly decreased the reduced glutathione level and catalase activity but increased malondialdehyde content. GEN and NaS exposure evoked pathological alterations in cochlear and vestibular tissues and increased caspase-3 and NF-κB immunoexpression. CCM significantly counteracted the GEN and NaS injurious effects. These outcomes concluded that CCM could be a naturally efficient therapeutic agent against GEN and NaS-associated ototoxic side effects.


Asunto(s)
Curcumina , Gentamicinas , Ototoxicidad , Salicilato de Sodio , Animales , Ratas , Caspasa 3 , Curcumina/farmacología , Gentamicinas/toxicidad , FN-kappa B , Salicilato de Sodio/toxicidad , Apoptosis
3.
Cells ; 10(5)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34069822

RESUMEN

Sodium salicylate (SA), a cyclooxygenase inhibitor, has been shown to increase insulin sensitivity and to suppress inflammation in obese patients and animal models. Transient receptor potential vanilloid 1 (TRPV1) is a nonselective cation channel expressed in afferent nerve fibers. Cyclooxygenase-derived prostaglandins are involved in the activation and sensitization of TRPV1. This study tested whether the metabolic and renal effects of SA were mediated by the TRPV1 channel. Wild-type (WT) and TRPV1-/- mice were fed a Western diet (WD) for 4 months and received SA infusion (120mg/kg/day) or vehicle for the last 4 weeks of WD feeding. SA treatment significantly increased blood pressure in WD-fed TRPV1-/- mice (p < 0.05) but not in WD-fed WT mice. Similarly, SA impaired renal blood flow in TRPV1-/- mice (p < 0.05) but not in WT mice. SA improved insulin and glucose tolerance in both WT and TRPV1-/- mice on WD (all p < 0.05). In addition, SA reduced renal p65 and urinary prostaglandin E2, prostaglandin F1α, and interleukin-6 in both WT and TRPV1-/- mice (all p < 0.05). SA decreased urine noradrenaline levels, increased afferent renal nerve activity, and improved baroreflex sensitivity in WT mice (all p < 0.05) but not in TRPV1-/- mice. Importantly, SA increased serum creatinine and urine kidney injury molecule-1 levels and decreased the glomerular filtration rate in obese WT mice (all p < 0.05), and these detrimental effects were significantly exacerbated in obese TRPV1-/- mice (all p < 0.05). Lastly, SA treatment increased urine albumin levels in TRPV1-/- mice (p < 0.05) but not in WT mice. Taken together, SA-elicited metabolic benefits and anti-inflammatory effects are independent of TRPV1, while SA-induced sympathetic suppression is dependent on TRPV1 channels. SA-induced renal dysfunction is dependent on intact TRPV1 channels. These findings suggest that SA needs to be cautiously used in patients with obesity or diabetes, as SA-induced renal dysfunction may be exacerbated due to impaired TRPV1 in obese and diabetic patients.


Asunto(s)
Inhibidores de la Ciclooxigenasa/toxicidad , Dieta Alta en Grasa , Enfermedades Renales/inducido químicamente , Riñón/efectos de los fármacos , Obesidad/tratamiento farmacológico , Salicilato de Sodio/toxicidad , Sistema Nervioso Simpático/efectos de los fármacos , Canales Catiónicos TRPV/deficiencia , Animales , Barorreflejo/efectos de los fármacos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Eliminación de Gen , Tasa de Filtración Glomerular/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Riñón/inervación , Riñón/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Enfermedades Renales/prevención & control , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Canales Catiónicos TRPV/genética
4.
Naunyn Schmiedebergs Arch Pharmacol ; 394(9): 1879-1891, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33937935

RESUMEN

Cyclooxygenase (COX) plays a critical role in synaptic plasticity. Therefore, long-term administration of acetylsalicylic acid (ASA) and its main metabolite, salicylate, as a COX inhibitor may impair synaptic plasticity and subsequently memory formation. Although different studies have tried to explain the effects of ASA and sodium salicylate (SS) on learning and memory, the results are contradictory and the mechanisms are not exactly known. The present study was designed to investigate the effects of long-term low-dose (equivalent to prophylactic dose) and short-term high-dose (equivalent to analgesic dose) administration of ASA and SS respectively, on spatial learning and memory and hippocampal synaptic plasticity. Animals were treated with a low dose of ASA (2 mg/ml solvated in drinking water, 6 weeks) or a high dose of SS, a metabolite of ASA, (300 mg/kg, 3 days, twice-daily, i.p). Spatial memory and synaptic plasticity were assessed by water maze performance and in vivo field potential recording from CA1, respectively. Animals treated with ASA but not SS showed a significant increase in escape latency and distance moved. Furthermore, in the probe test, animals treated with both drugs spent less time in the target quadrant zone. The paired-pulse ratio (PPR) at 20-ms inter-pulse intervals (IPI) as an index of short-term plasticity in both treated groups was significantly higher than of the control group. Interestingly, none of the administered drugs affected long-term potentiation (LTP). These data suggested that long-term inhibition of COX disrupted memory acquisition and retrieval. Interestingly, cognitive impairments happened along with short-term but not long-term synaptic plasticity disturbance.


Asunto(s)
Aspirina/toxicidad , Inhibidores de la Ciclooxigenasa/toxicidad , Salicilato de Sodio/toxicidad , Memoria Espacial/efectos de los fármacos , Animales , Aspirina/administración & dosificación , Inhibidores de la Ciclooxigenasa/administración & dosificación , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Wistar , Salicilato de Sodio/administración & dosificación , Aprendizaje Espacial/efectos de los fármacos
5.
Life Sci ; 265: 118824, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33278387

RESUMEN

Currently, antibiotics and salicylates are the most highly consumed medications worldwide. The side effects of these pharmaceuticals on the nervous system have been little investigated. Thus, this study aimed to examine the influence of the gentamicin (GM) and sodium salicylates (SS) on neurobehavioral functions, including locomotors function, memory, and sensorimotor functions together with gamma-aminobutyric acid (GABA) neurotransmitter levels. Also, oxidative stress, lipid peroxidation, and apoptotic indicators of brain tissue were assessed. Additionally, the histopathological architecture of brain tissues was investigated. This study also evaluated the curcumin (CUR) efficacy to counteract the GM or SS induced neurotoxic impacts in rats. For this purpose, seven groups were administered physiological saline (1 ml/rat; orally), olive oil (1 ml/rat; orally), CUR (50 mg/kg bwt; orally), GM (120 mg/kg bwt; intraperitoneally), SS (300 mg /kg bwt; intraperitoneally), CUR + GM, or CUR + SS for consecutive 15 days. The results revealed that GM and SS exposure evoked impaired memory, sensorimotor deficit functions, and depressive-like behavior together with the depletion of GABA. GM and SS exposure elevated malondialdehyde and Caspase-3 levels, but total antioxidant capacity and Bcl-2 levels were reduced. Besides, GM and SS exposure induced distinct pathological perturbations in cerebral cortices and hippocampus tissues. CUR significantly reversed the GM and SS harmful impacts. In conclusion, these findings verified that CUR could be a biologically efficient protective intervention against GM and SS induced neurotoxic impacts and neurobehavioral aberrations.


Asunto(s)
Curcumina/farmacología , Gentamicinas/toxicidad , Síndromes de Neurotoxicidad/prevención & control , Estrés Oxidativo/efectos de los fármacos , Salicilato de Sodio/toxicidad , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Síndromes de Neurotoxicidad/etiología , Ratas , Ratas Sprague-Dawley
6.
Biomed Pharmacother ; 126: 110036, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32172061

RESUMEN

OBJECTIVES: Although many studies have examined changes in gut microbiota composition in gastric carcinogenesis to clarify the mechanism of action of anticancer drugs, it is unclear whether animal models of gastric carcinogenesis adequately reflect the disease in humans. METHODS: To address this issue, the present study investigated changes in the gut microbiome profile of a rat model of gastric carcinogenesis established using a combination of N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), sodium salicylate, irregular fasting, and ranitidine. The rats were divided into control (Normal), chronic non-atrophic gastritis (CNAG), chronic atrophic gastritis (CAG), precancerous lesion of gastric cancer (PLGC), and gastric cancer (GC) groups according to histopathological features. Gut microbiome in gastric carcinogenesis profiling was performed by 16S rRNA gene sequencing of rat feces samples. RESULTS: We found that gut bacterial species richness increased whereas species diversity decreased during gastric carcinogenesis, with the most significant changes detected in the PLGC group. Gut microbiota community composition differed across groups, with the greatest similarities observed between CNAG and CAG groups and between PLGC and GC groups. There were significant differences in taxonomic representation at the phylum level: the PLGC group had the highest ratio of Firmicutes/Bacteroidetes whereas the GC group had the highest abundance of Proteobacteria and Actinobacteria. CONCLUSIONS: These results indicate that changes in the gut microbiome in a rat model of MNNG-induced gastric carcinogenesis are similar to those observed in humans, thus providing a useful tool for evaluating the efficacy and mechanism of action of novel monotherapies or drug combinations for the treatment of gastric carcinogenesis.


Asunto(s)
Bacterias/clasificación , Disbiosis/inducido químicamente , Microbioma Gastrointestinal/efectos de los fármacos , Neoplasias Gástricas/etiología , Animales , Bacterias/genética , Carcinogénesis , Heces/microbiología , Privación de Alimentos , Gastritis/inducido químicamente , Gastritis/complicaciones , Masculino , Metilnitronitrosoguanidina/toxicidad , ARN Bacteriano/aislamiento & purificación , ARN Ribosómico 16S/genética , Ranitidina/toxicidad , Ratas , Salicilato de Sodio/toxicidad
7.
Artículo en Inglés | MEDLINE | ID: mdl-31020389

RESUMEN

The purpose of this study was to observe the regulatory effects of GABAA (γ-aminobutyric acid A) receptor on the N-methyl-D-aspartate (NMDA) receptor during excitotoxicity in spiral ganglion neurons in the rat cochlea induced by sodium salicylate (SS). Western blot illustrated SS decreased the expression of NMDA receptor 2B subunit (NR2B) surface protein through affecting GABAA receptor, but the total protein content did not significantly change. Y1472 and S1480 are important phosphorylation sites in NR2B, SS downregulated the Fyn-dependent phosphorylation of Y1472 in a manner not related to the CK2 (Casein Kinase 2) dependent phosphorylation of S1480, thus regulating the surface distribution and internalization of NMDA receptor through GABAA receptor. These results suggest that the modified pattern of dynamic balance between excitation and inhibition by coactivation of the GABAA receptor can attenuate the excitatory NMDA receptor under the action of SS, via inhibiting the Fyn-dependent phosphorylation of Y1472.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Salicilato de Sodio/toxicidad , Ganglio Espiral de la Cóclea/efectos de los fármacos , Animales , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Ratas , Ratas Sprague-Dawley , Ganglio Espiral de la Cóclea/metabolismo
8.
Neurotox Res ; 35(4): 838-847, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30820888

RESUMEN

Sodium salicylate (SS) is one of the nonsteroidal anti-inflammatory drugs and widely used in clinical practice. Therefore, we aimed to investigate the potential ototoxicity mechanism of sodium salicylate: the influence of Ca2+/calmodulin-dependent protein kinase II (Ca2+/CaMKII) in interaction between NMDA receptors (NMDAR) and GABAA receptors (GABAAR) in rat cochlear spiral ganglion neurons (SGNs). After treatment with SS, NMDA, and an NMDAR inhibitor (APV), the changes of GABAAR ß3 (GABR ß3) mRNA, surface and total protein, and GABAAR currents in SGNs were assessed by quantitative PCR, Western blot, and whole-cell patch clamp. Mechanistically, SS and/or NMDA increased the GABR ß3 mRNA expression, while decreased GABR ß3 surface protein levels and GABAAR-mediated currents. Moreover, application of SS and/or NMDA showed promotion in phosphorylation levels at S383 of GABR ß3. Collectively, Ca2+ chelator (BAPTA) or Ca2+/CaMKII inhibitor (KN-93) reversed the effects of SS and/or NMDA on GABAAR. Therefore, we hypothesize that the interaction between NMDAR and GABAAR is involved in the SGNs damage induced by SS. In addition, the underlying molecular mechanism is related to Ca2+/CaMKII-mediated signaling pathway, which suggests that the interaction between calcium signal-regulated receptors mediates SS ototoxicity.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neuronas/efectos de los fármacos , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Salicilato de Sodio/toxicidad , Ganglio Espiral de la Cóclea/efectos de los fármacos , Animales , Neuronas/metabolismo , Ratas Sprague-Dawley , Ganglio Espiral de la Cóclea/metabolismo
9.
Hear Res ; 375: 44-52, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30795964

RESUMEN

Tinnitus may cause anxiety, depression, insomnia, which impair the quality of life of millions worldwide. However, the mechanism of tinnitus remains to be understood, it has been previously hypothesized that the activation of N-methyl-D-aspartate (NMDA) receptor is involved in the tinnitus processes and blockade of the NMDA receptor is regarded as a therapeutic strategy for tinnitus treatment even if the rescue treatment is still proved invalid in some cases. To demonstrate the therapeutic effect of the NMDA receptor blocker on tinnitus, we examined here the spontaneous firing rate (SFR) and the neurochemical dynamics in the auditory cortex (AC) of rats after sodium salicylate (SS) injection, which is a widely used model for tinnitus research. We also recorded their responses to MK-801 treatment. Electrophysiological studies showed that MK-801 significantly suppresses SFR in AC of rats with SS-induced tinnitus. In addition, by using a technique that combining in vivo microdialysis with an online electrochemical system (OECS) and a high-performance liquid chromatography (HPLC), we found that the levels of both glutamate and ascorbate in AC dramatically increased after SS injection and that MK-801 administration attenuated those response. Further studies found that MK-801 given at a time point of 30 min pre- or post-injection of SS were more effective than that given at a time point of 60 min post-SS injection, indicating that the time point of MK-801 intervention has a critical impact on the therapeutic effect. These findings suggest that MK-801 plays a neuroprotective role against hyperactivity during tinnitus induced by SS and that the therapeutic effect depends on the time point of MK-801 intervention, which would advance the studies on understanding of the therapeutic potential of NMDA receptor antagonist in tinnitus therapy.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Fármacos Neuroprotectores/farmacología , Acúfeno/tratamiento farmacológico , Animales , Ácido Ascórbico/metabolismo , Corteza Auditiva/fisiopatología , Modelos Animales de Enfermedad , Maleato de Dizocilpina/administración & dosificación , Esquema de Medicación , Potenciales Evocados Auditivos/efectos de los fármacos , Ácido Glutámico/fisiología , Humanos , Masculino , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Salicilato de Sodio/toxicidad , Acúfeno/inducido químicamente , Acúfeno/fisiopatología
10.
J Neurophysiol ; 121(3): 893-907, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30625004

RESUMEN

Electrophysiological and imaging studies from humans suggest that the phantom sound of tinnitus is associated with abnormal thalamocortical neural oscillations (dysrhythmia) and enhanced gamma band activity in the auditory cortex. However, these models have seldom been tested in animal models where it is possible to simultaneously assess the neural oscillatory activity within and between the thalamus and auditory cortex. To explore this issue, we used multichannel electrodes to examine the oscillatory behavior of local field potentials recorded in the rat medial geniculate body (MBG) and primary auditory cortex (A1) before and after administering a dose of sodium salicylate (SS) that reliably induces tinnitus. In the MGB, SS reduced theta, alpha, and beta oscillations and decreased coherence (synchrony) between electrode pairs in theta, alpha, and beta bands but increased coherence in the gamma band. Within A1, SS significantly increased gamma oscillations, decreased theta power, and decreased coherence between electrode pairs in theta and alpha bands but increased coherence in the gamma band. When coherence was measured between one electrode in the MGB and another in A1, SS decreased coherence in beta, alpha, and theta bands but increased coherence in the gamma band. SS also increased cross-frequency coupling between the phase of theta oscillations in the MGB and amplitude of gamma oscillations in A1. Altogether, our results suggest that SS treatment fundamentally alters the manner in which thalamocortical circuits communicate, leading to excessive cortical gamma power and synchronization, neurophysiological changes implicated in tinnitus. Our data provide support for elements of both the thalamocortical dysrhythmia (TD) and synchronization by loss of inhibition (SLIM) models of tinnitus, demonstrating that increased cortical gamma band activity is associated with both enhanced theta-gamma coupling as well as decreases alpha power/coherence between the MGB and A1. NEW & NOTEWORTHY There are no effective drugs to alleviate the phantom sound of tinnitus because the physiological mechanisms leading to its generation are poorly understood. Neural models of tinnitus suggest that it arises from abnormal thalamocortical oscillations, but these models have not been extensively tested. This article identifies abnormal thalamocortical oscillations in a drug-induced tinnitus model. Our findings open up new avenues of research to investigate whether cellular mechanisms underlying thalamocortical oscillations are causally linked to tinnitus.


Asunto(s)
Corteza Auditiva/fisiopatología , Ondas Encefálicas , Tálamo/fisiopatología , Acúfeno/fisiopatología , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Salicilato de Sodio/toxicidad , Acúfeno/etiología
11.
Congenit Anom (Kyoto) ; 59(6): 190-192, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30499140

RESUMEN

Thoracolumbar supernumerary ribs (TSRs) are classified as less severe skeletal anomalies in rat developmental toxicity studies, although their incidence is relatively high in rodent studies. To investigate the characteristics of the critical window for chemically-induced TSR, in this study, rats were administered 5-fluorocytocine (5-FC) or sodium salicylate (SAL) at one of three time periods on gestational day (GD) 9, early morning (7:00 am), midday (12:00 pm to 1:00 pm), or late afternoon (4:00 pm or 7:00 pm). The incidence of TSR and other anomalies were assessed in GD20 fetuses. A single treatment with both chemicals on GD9-induced TSR, with the incidence highest when administered at 7:00 Am, decreasing gradually when administered later. This trajectory was clearer in rats treated with 5-FC than with SAL. The critical period of TSR induction is shorter in rats administered 5-FC than SAL. The characteristics of the critical window may cause variability in the incidence of TSR observed in developmental toxicity studies.


Asunto(s)
Anomalías Inducidas por Medicamentos/fisiopatología , Feto/fisiopatología , Anomalías Musculoesqueléticas/fisiopatología , Costillas/fisiopatología , Animales , Feto/efectos de los fármacos , Flucitosina/toxicidad , Humanos , Anomalías Musculoesqueléticas/inducido químicamente , Ratas , Costillas/crecimiento & desarrollo , Salicilato de Sodio/toxicidad , Teratógenos/farmacología , Teratógenos/toxicidad
12.
Sheng Li Xue Bao ; 69(3): 285-290, 2017 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-28638920

RESUMEN

The aim of the present study was to observe whether dopamine receptor (DR) was involved in the effects of sodium salicylate (SS) on the expressions of N-methyl-D-aspartic acid (NMDA) and γ-aminobutyric acid (GABA) receptors in rat cochlear spiral ganglion neurons (SGNs). Forty-eight hours after primary culture of rat SGNs, immunofluorescence technique was applied to detect expressions of DR1 and DR2, the two subtypes of dopamine receptors. Western blot was performed to assess NMDA receptor NR1 subunit and GABAA receptor subunit α2 (GABRα2) protein expressions in the SGNs after the treatments of SS alone or in combination with DR antagonists. The results demonstrated that: (1) The DR1 and DR2 were expressed in the bodies and axons of the SGN; (2) After the treatment with SS, the surface protein expressions of GABRα2 and NR1 were decreased by 44.69% and 21.57%, respectively, while the total protein expressions showed no significant changes; (3) Neither SS + SCH23390 (DR1 antagonist) group nor SS + Eticlopride (DR2 antagonist) group showed significant differences in GABRα2 and NR1 surface protein expressions compared with the control group. These results suggest that SS regulates the surface GABAA and NMDA receptors trafficking on SGN, and the mechanism may involve DR mediation.


Asunto(s)
Receptores Dopaminérgicos/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Salicilato de Sodio/toxicidad , Ganglio Espiral de la Cóclea/efectos de los fármacos , Animales , Benzazepinas/farmacología , Células Cultivadas , Cóclea/citología , Neuronas/efectos de los fármacos , Ratas
13.
Eur J Neurosci ; 45(3): 398-409, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27862478

RESUMEN

Tinnitus chronically affects between 10-15% of the population but, despite its prevalence, the underlying mechanisms are still not properly understood. One experimental model involves administration of high doses of sodium salicylate, as this is known to reliably induce tinnitus in both humans and animals. Guinea pigs were implanted with chronic electrocorticography (ECoG) electrode arrays, with silver-ball electrodes placed on the dura over left and right auditory cortex. Two more electrodes were positioned over the cerebellum to monitor auditory brainstem responses (ABRs). We recorded resting-state and auditory evoked neural activity from awake animals before and 2 h following salicylate administration (350 mg/kg; i.p.). Large increases in click-evoked responses (> 100%) were evident across the whole auditory cortex, despite significant reductions in wave I ABR amplitudes (in response to 20 kHz tones), which are indicative of auditory nerve activity. In the same animals, significant decreases in 6-10 Hz spontaneous oscillations (alpha waves) were evident over dorsocaudal auditory cortex. We were also able to demonstrate for the first time that cortical evoked potentials can be inhibited by a preceding gap in background noise [gap-induced pre-pulse inhibition (PPI)], in a similar fashion to the gap-induced inhibition of the acoustic startle reflex that is used as a behavioural test for tinnitus. Furthermore, 2 h following salicylate administration, we observed significant deficits in PPI of cortical responses that were closely aligned with significant deficits in behavioural responses to the same stimuli. Together, these data are suggestive of neural correlates of tinnitus and oversensitivity to sound (hyperacusis).


Asunto(s)
Ritmo alfa , Corteza Auditiva/fisiología , Acúfeno/fisiopatología , Animales , Corteza Auditiva/efectos de los fármacos , Umbral Auditivo , Nervio Coclear/fisiología , Potenciales Evocados Auditivos , Femenino , Cobayas , Masculino , Inhibición Neural , Reflejo Acústico , Reflejo de Sobresalto , Salicilato de Sodio/toxicidad , Acúfeno/etiología , Vigilia
14.
Hear Res ; 332: 104-112, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26688177

RESUMEN

Rebound depolarization (RD) is a voltage response to the offset from pre-hyperpolarization of neuronal membrane potential, which manifests a particular form of the postsynaptic membrane potential response to inhibitory presynaptic inputs. We previously demonstrated that sodium salicylate (NaSal), a tinnitus inducer, can drastically suppress the RD in neurons of rat medial geniculate body (MGB) (Su et al, 2012; PLoS ONE 7, e46969). The purpose of the present study was to investigate the underlying cellular mechanism by using whole-cell patch-clamp recordings in rat MGB slices. NaSal (1.4 mM) had no effects on the current mediated by T-type Ca(2+) channels, indicating that it does not target these channels to suppress the RD. Instead, NaSal was shown to hyperpolarize the resting membrane potential to suppress the RD. NaSal had no effects on the current mediated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, indicating that it does not target these channels to hyperpolarize the resting membrane potential. NaSal induced an outward leak current that could be abolished by CGP55845, a GABAB receptor blocker, or respectively by Ba(2+) and Tertiapin-Q, blockers for G-protein-gated inwardly rectifying potassium (GIRK) channels, indicating that NaSal potentiates the GABAB-GIRK pathway to hyperpolarize the resting membrane potential. Our study demonstrates that NaSal targets GABAB receptors to alter functional behaviors of MGB neurons, which may be implicated in NaSal-induced tinnitus.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Cuerpos Geniculados/efectos de los fármacos , Receptores de GABA-B/efectos de los fármacos , Receptores de GABA-B/metabolismo , Transducción de Señal/efectos de los fármacos , Salicilato de Sodio/toxicidad , Animales , Femenino , Antagonistas de Receptores de GABA-B , Neuronas GABAérgicas/metabolismo , Cuerpos Geniculados/metabolismo , Técnicas In Vitro , Masculino , Potenciales de la Membrana , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Ratas Wistar
15.
Eur Arch Otorhinolaryngol ; 273(2): 325-32, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25636249

RESUMEN

Spontaneous neuronal activity in dorsal cochlear nucleus (DCN) may be involved in the physiological processes underlying salicylate-induced tinnitus. As a neuronal activity marker, immediate-early gene (IEG) expression, especially activity-dependent cytoskeletal protein (Arc/Arg3.1) and the early growth response gene-1 (Egr-1), appears to be highly correlated with sensory-evoked neuronal activity. However, their relationships with tinnitus induced by salicylate have rarely been reported in the DCN. In this study, we assessed the effect of acute and chronic salicylate treatment on the expression of N-methyl D-aspartate receptor subunit 2B (NR2B), Arg3.1, and Egr-1. We also observed ultrastructural alterations in the DCN synapses in an animal model of tinnitus. Levels of mRNA and protein expression of NR2B and Arg3.1 were increased in rats that were chronically administered salicylate (200 mg/kg, twice daily for 3, 7, or 14 days). These levels returned to baseline 14 days after cessation of treatment. However, no significant changes were observed in Egr-1 gene expression in any groups. Furthermore, rats subjected to long-term salicylate administration showed more presynaptic vesicles, thicker and longer postsynaptic densities, and increased synaptic interface curvature. Alterations of Arg3.1 and NR2B may be responsible for the changes in the synaptic ultrastructure. These changes confirm that salicylate can cause neural plasticity changes at the DCN level.


Asunto(s)
Núcleo Coclear/metabolismo , Regulación de la Expresión Génica , Genes Inmediatos-Precoces/genética , ARN Mensajero/genética , Acúfeno/genética , Animales , Núcleo Coclear/ultraestructura , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Microscopía Electrónica de Transmisión , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Salicilato de Sodio/toxicidad , Sinapsis/genética , Sinapsis/metabolismo , Acúfeno/inducido químicamente , Acúfeno/metabolismo
16.
Hear Res ; 332: 188-198, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26548740

RESUMEN

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.


Asunto(s)
Núcleo Coclear/efectos de los fármacos , Glicina/metabolismo , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Salicilato de Sodio/toxicidad , Transmisión Sináptica/efectos de los fármacos , Acúfeno/inducido químicamente , Animales , Núcleo Coclear/metabolismo , Núcleo Coclear/fisiopatología , Potenciales Evocados Auditivos , Técnicas In Vitro , Masculino , Neuronas/metabolismo , Ratas Wistar , Acúfeno/metabolismo , Acúfeno/fisiopatología
17.
Clin Lab ; 61(9): 1113-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26554229

RESUMEN

BACKGROUND: This study investigated the effects of monosialotetrahexosylganglioside (GM1) on the expression of N-methyl-D-aspartate receptor subunit 2B (NR2B) and phosphorylated (p)-cyclic AMP response element-binding protein (CREB) in the auditory cortex of rats with tinnitus. METHODS: Tinnitus-like behavior in rats was tested with the gap prepulse inhibition of acoustic startle paradigm. We then investigated the NR2B mRNA and protein and p-CREB protein levels in the auditory cortex of tinnitus rats compared with normal rats. RESULTS: Rats treated for 4 days with salicylate exhibited tinnitus. NR2B mRNA and protein and p-CREB protein levels were upregulated in these animals, with expression returning to normal levels 14 days after cessation of treatment; baseline levels of NR2B and p-CREB were also restored by GM1 administration. CONCLUSIONS: These data suggest that chronic salicylate administration induces tinnitus via upregulation of p-CREB and NR2B expression, and that GM1 can potentially be used to treat tinnitus.


Asunto(s)
Corteza Auditiva/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Gangliósido G(M1)/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/biosíntesis , Salicilato de Sodio/toxicidad , Acúfeno/tratamiento farmacológico , Animales , Corteza Auditiva/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos , Gangliósido G(M1)/farmacología , Masculino , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Fosforilación/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/genética , Acúfeno/inducido químicamente , Acúfeno/metabolismo
18.
Hear Res ; 295: 140-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22330978

RESUMEN

Animal models of tinnitus require a behavioral correlate thereof. Various conditioned response methods and gap-startle reflex methods are in use and the outcomes generally correspond with putative electrophysiological substrates of tinnitus. However, for salicylate-induced tinnitus there is discordance between the behavioral and electrophysiological test results. As a result, it is not clear what the various tests are reflecting. A review of the, mostly sub-cortical, neural circuits that underlie the behavioral responses suggests that cortical electrophysiological correlates do not necessarily have to correspond with behavioral ones. Human objective correlates of tinnitus point heavily into cortical network, but not just auditory cortex, correlates of tinnitus. Furthermore, the synaptic mechanisms underlying spontaneous firing rate changes may be different from those involved in driven neural activity.


Asunto(s)
Pérdida Auditiva/etiología , Hiperacusia/etiología , Acúfeno/etiología , Anestesia , Animales , Corteza Auditiva/fisiopatología , Conducta Animal , Condicionamiento Psicológico , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos , Pérdida Auditiva/fisiopatología , Pérdida Auditiva/psicología , Pérdida Auditiva Provocada por Ruido/etiología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Provocada por Ruido/psicología , Humanos , Hiperacusia/fisiopatología , Hiperacusia/psicología , Psicoacústica , Reflejo de Sobresalto , Salicilato de Sodio/toxicidad , Acúfeno/fisiopatología , Acúfeno/psicología
19.
Hear Res ; 295: 100-13, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23201030

RESUMEN

High doses of sodium salicylate (SS) have long been known to induce temporary hearing loss and tinnitus, effects attributed to cochlear dysfunction. However, our recent publications reviewed here show that SS can induce profound, permanent, and unexpected changes in the cochlea and central nervous system. Prolonged treatment with SS permanently decreased the cochlear compound action potential (CAP) amplitude in vivo. In vitro, high dose SS resulted in a permanent loss of spiral ganglion neurons and nerve fibers, but did not damage hair cells. Acute treatment with high-dose SS produced a frequency-dependent decrease in the amplitude of distortion product otoacoustic emissions and CAP. Losses were greatest at low and high frequencies, but least at the mid-frequencies (10-20 kHz), the mid-frequency band that corresponds to the tinnitus pitch measured behaviorally. In the auditory cortex, medial geniculate body and amygdala, high-dose SS enhanced sound-evoked neural responses at high stimulus levels, but it suppressed activity at low intensities and elevated response threshold. When SS was applied directly to the auditory cortex or amygdala, it only enhanced sound evoked activity, but did not elevate response threshold. Current source density analysis revealed enhanced current flow into the supragranular layer of auditory cortex following systemic SS treatment. Systemic SS treatment also altered tuning in auditory cortex and amygdala; low frequency and high frequency multiunit clusters up-shifted or down-shifted their characteristic frequency into the 10-20 kHz range thereby altering auditory cortex tonotopy and enhancing neural activity at mid-frequencies corresponding to the tinnitus pitch. These results suggest that SS-induced hyperactivity in auditory cortex originates in the central nervous system, that the amygdala potentiates these effects and that the SS-induced tonotopic shifts in auditory cortex, the putative neural correlate of tinnitus, arises from the interaction between the frequency-dependent losses in the cochlea and hyperactivity in the central nervous system.


Asunto(s)
Cóclea/efectos de los fármacos , Cóclea/fisiopatología , Salicilato de Sodio/toxicidad , Acúfeno/inducido químicamente , Acúfeno/fisiopatología , Potenciales de Acción/efectos de los fármacos , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiopatología , Animales , Corteza Auditiva/efectos de los fármacos , Corteza Auditiva/fisiopatología , Vías Auditivas/efectos de los fármacos , Vías Auditivas/fisiopatología , Cóclea/patología , Potenciales Evocados Auditivos/efectos de los fármacos , Cuerpos Geniculados/efectos de los fármacos , Cuerpos Geniculados/fisiopatología , Emisiones Otoacústicas Espontáneas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/patología , Ganglio Espiral de la Cóclea/fisiopatología , Acúfeno/patología
20.
PLoS One ; 7(10): e46969, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071681

RESUMEN

Sodium salicylate (NaSal), an aspirin metabolite, can cause tinnitus in animals and human subjects. To explore neural mechanisms underlying salicylate-induced tinnitus, we examined effects of NaSal on neural activities of the medial geniculate body (MGB), an auditory thalamic nucleus that provides the primary and immediate inputs to the auditory cortex, by using the whole-cell patch-clamp recording technique in MGB slices. Rats treated with NaSal (350 mg/kg) showed tinnitus-like behavior as revealed by the gap prepulse inhibition of acoustic startle (GPIAS) paradigm. NaSal (1.4 mM) decreased the membrane input resistance, hyperpolarized the resting membrane potential, suppressed current-evoked firing, changed the action potential, and depressed rebound depolarization in MGB neurons. NaSal also reduced the excitatory and inhibitory postsynaptic response in the MGB evoked by stimulating the brachium of the inferior colliculus. Our results demonstrate that NaSal alters neuronal intrinsic properties and reduces the synaptic transmission of the MGB, which may cause abnormal thalamic outputs to the auditory cortex and contribute to NaSal-induced tinnitus.


Asunto(s)
Cuerpos Geniculados/efectos de los fármacos , Neuronas/efectos de los fármacos , Salicilato de Sodio/toxicidad , Transmisión Sináptica/efectos de los fármacos , Acúfeno/fisiopatología , Estimulación Acústica , Potenciales de Acción/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/toxicidad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Cuerpos Geniculados/fisiopatología , Colículos Inferiores/efectos de los fármacos , Colículos Inferiores/fisiopatología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Receptores de GABA-A/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Reflejo de Sobresalto/efectos de los fármacos , Acúfeno/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...