Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Toxicol Lett ; 313: 11-18, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31220555

RESUMEN

Previous study reported that either selective GSK-3ß inhibitor or up-regulating autophagy can alleviate cisplatin-induced ototoxicity. Other studies indicate that the activity of GSK-3ß is closely associated with the autophagy level. The purpose of this study is to primarily explore the role of autophagy in the alleviation effect of GSK-3ß inhibition on cisplatin-induced ototoxicity in vivo and in vitro. We observed the autophagy changes induced by GSK-3ß inhibitor in outer hair cells (OHCs) in a cisplatin-induced ototoxicity rat model. In addition, autophagy inhibitor 3-MA was used in vitro experiments to observe the influence of autophagy inhibition on the cell protection effect due to GSK-3ß inactivation. The relationship among autophagy, GSK-3ß and cell damage were inferred. Negative regulation of GSK-3ß significantly enhanced autophagy and alleviated cisplatin-induced hearing loss, OHC death in vivo and apoptosis in vitro. The autophagy inhibitor 3-MA inverted the protective effect of negative regulation of GSK-3ß. These results indicated that enhancing autophagy may be a key downstream effect of GSK-3ß inhibition in the alleviation of cisplatin-induced ototoxicity both in vivo and in vitro.


Asunto(s)
Autofagia/efectos de los fármacos , Cisplatino , Enfermedades del Oído/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Ciliadas Auditivas/efectos de los fármacos , Cloruro de Litio/farmacología , Animales , Fatiga Auditiva/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Enfermedades del Oído/inducido químicamente , Enfermedades del Oído/enzimología , Enfermedades del Oído/patología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Células Ciliadas Auditivas/enzimología , Células Ciliadas Auditivas/patología , Masculino , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
2.
Int J Audiol ; 57(12): 917-924, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30382794

RESUMEN

OBJECTIVE: Identify hearing effects of a single course of intravenous (IV) aminoglycoside antibiotics (AGs) therapy in adult cystic fibrosis (CF) patients. Determine whether the change is large enough to enable a proof-of-concept study of a new drug preventing AG-associated hearing loss. DESIGN: Retrospective case review of CF patients with sequential audiograms ± an intervening course of IV AG therapy. STUDY SAMPLE: 84 patients with no intervening IV AG treatment, 38 patients undergoing a single course of IV AGs. RESULTS: Using ASHA ototoxicity metrics, 45% of adult CF patients in the Single-IV group met the criteria for ototoxicity compared to 23% of the No-IV patients. Other hearing metrics including the average maximal threshold shift (TS) and average high frequency TS showed highly significant differences between groups. Testing only participants with mild or greater pre-therapy high frequency hearing loss further increased the differences between the two groups by every metric tested. CONCLUSION: Adult CF patients exposed to a single course of IV AGs have significantly greater TS than patients without IV AG exposure. Patients with mild to moderate hearing loss prior to AG-IVs are at increased risk of developing ototoxicity from subsequent parenteral AG therapy.


Asunto(s)
Aminoglicósidos/efectos adversos , Antibacterianos/efectos adversos , Fibrosis Quística/tratamiento farmacológico , Trastornos de la Audición/inducido químicamente , Audición/efectos de los fármacos , Administración Intravenosa , Adolescente , Adulto , Anciano , Aminoglicósidos/administración & dosificación , Antibacterianos/administración & dosificación , Fatiga Auditiva/efectos de los fármacos , Fibrosis Quística/diagnóstico , Fibrosis Quística/microbiología , Femenino , Trastornos de la Audición/diagnóstico , Trastornos de la Audición/fisiopatología , Trastornos de la Audición/psicología , Pruebas Auditivas , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento , Adulto Joven
3.
Hear Res ; 370: 16-21, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30253329

RESUMEN

Cisplatin is a potent chemotherapeutic compound for which ototoxicity is a significant side effect. Cisplatin has shown sensitivity to circadian time, in that cisplatin is most effective as an anti-tumor compound, and least nephrotoxic, when given in the active (dark) period of the light-dark cycle in rodents. The objective of the study was to determine the sensitivity of cisplatin ototoxicity to circadian time. Fifty-seven Fischer 344/NHsd rats were exposed to 12 mg/kg cisplatin by intra-peritoneal injection at one of six time points on a 12 h light-12 h dark cycle: 2, 6, or 10 h after light onset or 2, 6, or 10 h after light offset. Cochlear injury was evaluated using auditory brainstem response threshold shifts and postmortem outer hair cell counts. All animals experienced threshold shift in the highest frequencies tested (30 and 40 kHz). The animals exposed to cisplatin at 6 h after light onset (the inactive period) had significantly higher mid-frequency threshold shifts and outer hair cell losses than the groups exposed during the dark hours. The results indicate that cisplatin is less likely to cause ototoxicity in the Fischer 344/NHsd rat when given during the active period. This finding is consistent with the lower nephrotoxicity that has been detected in cisplatin-exposed animals treated during the dark hours, and the magnitude of differences in threshold shifts between the light and dark exposure indicates that circadian timing has a significant impact on susceptibility to cisplatin ototoxicity.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Cóclea/efectos de los fármacos , Enfermedades Cocleares/prevención & control , Cronoterapia de Medicamentos , Animales , Antineoplásicos/toxicidad , Fatiga Auditiva/efectos de los fármacos , Cisplatino/toxicidad , Cóclea/patología , Cóclea/fisiopatología , Enfermedades Cocleares/inducido químicamente , Enfermedades Cocleares/patología , Enfermedades Cocleares/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Masculino , Fotoperiodo , Ratas Endogámicas F344 , Factores de Tiempo
4.
Hear Res ; 368: 92-108, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30006113

RESUMEN

The hybrid or electric-acoustic stimulation cochlear implant is indicated in patients with a residual hearing at low frequencies. It provides electric and acoustic stimulation for compensating for high- and low-frequency sounds, respectively. However, the implantation procedure damages the cochlea, resulting in loss of the residual-hearing and diminished effects of the acoustic-hearing in several patients. To prevent hearing loss after implantation, corticosteroids have been used clinically although their effects are limited. As an alternative to corticosteroids, insulin-like growth factor 1 (IGF1) has shown potent effects in various types of cochlear injury. In this study, the effects of IGF1 on hearing preservation were examined after cochlear implantation to a normal-hearing guinea pig model. The electrode was inserted in an atraumatic way through the round window membrane of guinea pigs with the application of a gelatin-sponge soaked with IGF1 or saline. The auditory brainstem response (ABR) was recorded pre-operatively, immediately after cochlear implantation, and 7, 14, 28, and 56 days after electrode insertion. In comparison to the control group, the IGF1-treated group showed better hearing preservation at low frequencies, 7 days after surgery. IGF1 application was effective at low frequencies (2 and 4 kHz) throughout the period of examination. Histological studies revealed that outer hair cell numbers, in the IGF1-treated group, were maintained in the cochlear region responsible for low-frequency hearing (upper midbasal turn) and that there was less fibrous tissue formation around the electrode. Both the outer hair cell counts and the extent of fibrosis significantly correlated with the ABR threshold shifts at low frequencies. These results indicate that IGF1 might attenuate loss of low-frequency hearing after cochlear implantation, suggesting its possible clinical use in soft surgeries involving cochlear implants with electric-acoustic stimulation for hearing preservation.


Asunto(s)
Cóclea/efectos de los fármacos , Implantación Coclear/instrumentación , Implantes Cocleares , Audición/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Animales , Fatiga Auditiva/efectos de los fármacos , Cóclea/lesiones , Cóclea/patología , Cóclea/fisiopatología , Implantación Coclear/efectos adversos , Portadores de Fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Fibrosis , Gelatina/química , Cobayas , Masculino , Modelos Animales , Tapones Quirúrgicos de Gaza , Factores de Tiempo
5.
Hear Res ; 358: 10-21, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29304389

RESUMEN

Aminoglycoside antibiotics are used widely in medicine despite their ototoxic side-effects. Oxidative stress and inflammation are key mechanisms determining the extent and severity of the damage. Here we evaluate the protective effect of a treatment with resveratrol plus N-acetylcysteine on the ototoxic actions of kanamycin and furosemide in the rat. Resveratrol (10 mg/kg) and N-acetylcysteine (400 mg/kg) were administered together to Wistar rats on 5 consecutive days. The second day, a concentrated solution of kanamycin and furosemide was placed on the round window to induce ototoxicity. Hearing was assessed by recording auditory brainstem responses before and 5, 16 and 23 days after the beginning of the treatment. Cochlear samples were taken at day 5 (end of the treatment) and at day 23, and targeted PCR arrays or RT-qPCR were performed to analyze oxidative balance and inflammation related genes, respectively. In addition, the cytoarchitecture and the presence of apoptosis, oxidative stress and inflammation markers were evaluated in cochlear sections. Results indicate that administration of resveratrol plus N-acetylcysteine reduced the threshold shifts induced by ototoxic drugs at high frequencies (≈10 dB), although this protective effect fades after the cessation of the treatment. Gene expression analysis showed that the treatment modulated the expression of genes involved in the cellular oxidative (Gpx1, Sod1, Ccs and Noxa1) and inflammatory (Il1b, Il4, Mpo and Ncf) responses to injury. Thus, co-administration of resveratrol and NAC, routinely used individually in patients, could reduce the ototoxic secondary effects of aminoglycosides.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Tronco Encefálico/efectos de los fármacos , Cóclea/efectos de los fármacos , Pérdida Auditiva/prevención & control , Audición/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Resveratrol/farmacología , Animales , Apoptosis/efectos de los fármacos , Fatiga Auditiva/efectos de los fármacos , Tronco Encefálico/fisiopatología , Cóclea/metabolismo , Cóclea/patología , Citoprotección , Modelos Animales de Enfermedad , Quimioterapia Combinada , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Furosemida , Regulación de la Expresión Génica , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/metabolismo , Pérdida Auditiva/fisiopatología , Mediadores de Inflamación/metabolismo , Kanamicina , Masculino , Estrés Oxidativo/genética , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos
6.
Hear Res ; 356: 104-115, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29089185

RESUMEN

Sustained local delivery of drugs to the inner ear may be required for future regenerative and protective strategies. The round window is surgically accessible and a promising delivery route. To be viable, a delivery system should not cause hearing loss. This study determined the effect on hearing of placing a drug-delivery microcatheter on to the round window, and delivering either artificial perilymph (AP) or brain-derived neurotrophic factor (BDNF) via this catheter with a mini-osmotic pump. Auditory brainstem responses (ABRs) were monitored for 4 months after surgery, while the AP or BDNF was administered for the first month. The presence of the microcatheter - whether dry or when delivering AP or BDNF for 4 weeks - was associated with an increase in ABR thresholds of up to 15 dB, 16 weeks after implantation. This threshold shift was, in part, delayed by the delivery of BDNF. We conclude that the chronic presence of a microcatheter in the round window niche causes hearing loss, and that this is exacerbated by delivery of AP, and ameliorated temporarily by delivery of BDNF.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Cateterismo/instrumentación , Catéteres de Permanencia , Catéteres Venosos Centrales , Sistemas de Liberación de Medicamentos/instrumentación , Pérdida Auditiva/tratamiento farmacológico , Audición/efectos de los fármacos , Ventana Redonda/efectos de los fármacos , Estimulación Acústica , Animales , Audiometría de Tonos Puros , Fatiga Auditiva/efectos de los fármacos , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Cobayas , Pérdida Auditiva/diagnóstico por imagen , Pérdida Auditiva/etiología , Pérdida Auditiva/fisiopatología , Bombas de Infusión Implantables , Microscopía Confocal , Perilinfa/química , Recuperación de la Función , Ventana Redonda/diagnóstico por imagen , Ventana Redonda/fisiopatología , Factores de Tiempo , Tomografía de Coherencia Óptica
7.
Hear Res ; 327: 257-64, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26117408

RESUMEN

INTRODUCTION: Steroids have been shown to reduce the hearing threshold shifts associated with cochlear implantation. Previous studies have examined only the administration of steroids just prior to surgery. The aim of this study is to examine the role of extended preoperative systemic steroids in hearing preservation cochlear implantation. METHODS: An animal model of cochlear implantation was used. 24 Hartley strain guinea pigs with a mean weight of 768 g and normal hearing were randomised into a control group, a second group receiving a single dose of systemic dexamethasone one day prior to surgery, and a third group receiving a daily dose of systemic dexamethasone for 5 days prior to surgery. A specially designed cochlear implant electrode by Med-EL (Innsbruck) was inserted through a dorsolateral approach to an insertion depth of 5 mm and left in-situ. Auditory brain stem responses at 8 kHz, 16 kHz and 32 kHz were measured preoperatively, and 1 week, 1 month and 2 months postoperatively. Cochlear histopathology was examined at the conclusion of the study. RESULTS: At 1-week post operative, both groups receiving dexamethasone prior to implantation had smaller threshold shifts across all frequencies and which was significant at 32 kHz (p < 0.05). There were no differences among the three groups in the area of electrode related fibrosis. Spiral ganglion neuron (SGN) density was significantly higher in the group receiving steroids for 5 days, but only in the basal cochlear turn. DISCUSSION: This is study demonstrates the benefits of extended preoperative systemic steroids on hearing outcomes and SGN density in an animal model of cochlear implantation surgery.


Asunto(s)
Cóclea/efectos de los fármacos , Implantación Coclear/efectos adversos , Implantes Cocleares/efectos adversos , Dexametasona/administración & dosificación , Glucocorticoides/administración & dosificación , Pérdida Auditiva/prevención & control , Audición/efectos de los fármacos , Estimulación Acústica , Animales , Fatiga Auditiva/efectos de los fármacos , Cóclea/patología , Cóclea/fisiopatología , Implantación Coclear/instrumentación , Modelos Animales de Enfermedad , Esquema de Medicación , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Fibrosis , Cobayas , Pérdida Auditiva/etiología , Pérdida Auditiva/patología , Pérdida Auditiva/fisiopatología , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/patología , Factores de Tiempo
8.
Noise Health ; 16(72): 265-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25209035

RESUMEN

A potential consequence of exposure to noise is a temporary reduction in auditory sensitivity known as temporary threshold shift (TTS), which mainly depends on the intensity and duration of exposure to the noise. Recovery time is related to the amount of initial hearing loss, and the most recovery takes place during the first 15 min following exposure. This study evaluated the efficacy in otoprotection against noise-induced hearing loss of an orally administrated food supplement containing coenzyme Q 10 -Ter. This water-soluble formulation of coenzyme Q 10 shows better bioavailability than the native form and has been found to have a protective effect on outer hair cells after exposure to noise in animal models. Thirty volunteers were enrolled, and the right ear of each subject was exposed to a narrow-band noise centered at 3 kHz for 10 min at the intensity of 90 dB HL. In the 30 subjects enrolled, TTS was evaluated after 2, 15, and 30 min and the recovery time was recorded in each subject. The longest recovery time was 45 min. Among the 18 subjects who underwent a second test after treatment with Q-Ter, the mean recovery time was 31.43 min. The results of the present study show that 30 days' treatment with Q-Ter can aid faster recovery after exposure to noise (P < 0.0001). The reduction in the recovery time following treatment can be explained by Q-Ter-mediated improvement of the outer hair cells' response to oxidative stress.


Asunto(s)
Fatiga Auditiva/efectos de los fármacos , Suplementos Dietéticos , Ruido/efectos adversos , Recuperación de la Función/efectos de los fármacos , Ubiquinona/análogos & derivados , Estimulación Acústica , Adulto , Audiometría de Tonos Puros , Disponibilidad Biológica , Femenino , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/fisiología , Humanos , Masculino , Ruido en el Ambiente de Trabajo/efectos adversos , Recuperación de la Función/fisiología , Factores de Tiempo , Acúfeno/tratamiento farmacológico , Acúfeno/fisiopatología , Ubiquinona/administración & dosificación
9.
Environ Toxicol Pharmacol ; 36(2): 463-471, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23792233

RESUMEN

All-trans retinoic acid (ATRA) is reported to reduce hair cell loss and hearing deterioration caused by noise-induced hearing loss (NIHL). The present study investigates the involvement of peroxiredoxin 6 (Prdx 6) in ATRA-mediated protection of temporary threshold shift of hearing. Mice fed with ATRA before or after exposure to white noise showed a faster recovery than untreated controls within 1 week, with a concomitant increase of cochlear Prdx 6 expression. Treatment of mouse auditory cells with ATRA induced Prdx 6 expression. A putative retinoic acid (RA)-response element (RARE) was identified in a murine Prdx 6 promoter region. Prdx 6 promoter activities were elevated in wild-type reporter plasmid-transfected cells, whereas no significant change in activity was in those with RARE-disrupted mutant reporter. RA receptor α (RARα) functions as a transactivator of Prdx 6 gene expression. These findings suggest that ATRA-induced Prdx 6 expression may be associated with rapid recovery from temporary NIHL.


Asunto(s)
Fatiga Auditiva/efectos de los fármacos , Cóclea/efectos de los fármacos , Pérdida Auditiva Provocada por Ruido/metabolismo , Audición/efectos de los fármacos , Peroxiredoxina VI/metabolismo , Tretinoina/farmacología , Animales , Sitios de Unión , Línea Celular , Cóclea/metabolismo , Cóclea/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Pérdida Auditiva Provocada por Ruido/etiología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Ratones , Ratones Endogámicos BALB C , Mutación , Peroxiredoxina VI/genética , Regiones Promotoras Genéticas , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Recuperación de la Función , Elementos de Respuesta , Receptor alfa de Ácido Retinoico , Transducción de Señal , Factores de Tiempo , Activación Transcripcional , Transfección , Regulación hacia Arriba
10.
Eur Arch Otorhinolaryngol ; 270(4): 1513-20, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23179931

RESUMEN

Noise exposure causes an excessive reactive oxygen species (ROS) generation as an unwanted byproduct of high metabolic activity. Oxidative stress and antioxidative protective mechanisms have been therefore proposed as the most interesting issues in the development of noise-induced hearing loss. The aim of this study was to examine changes in superoxide dismutase (SOD), catalase (CAT) and the auditory brainstem response (ABR) in the cochlea of C57BL/6 mice 1, 7 and 14 days after exposure to 4 kHz octave band noise at the intensity of 110 dB SPL for 8 h. The evaluation of three D-methionine (D-met) doses (100, 200 and 400 mg/kg) has been performed in order to choose an optimal concentration displaying most effectively its antioxidant and thereby otoprotective functions. Administering D-met at the dose of 400 mg/kg resulted in a significant decrease in threshold shift (TS) independently of the evaluation time after exposure to noise. SOD activity was strongly supported by the same concentration (400 mg/kg) of D-met. This effect was seen not shortly, but 7 and 14 days after exposure to noise. CAT activity was induced only by noise and it reached the peak levels 7 days after exposure. D-Met at the doses of 200 and 400 mg/kg significantly decreased noise-induced changes in CAT activity. The findings of this study indicate that the protective effect depends on the concentration of D-met and can be fully expressed only when the drug is administered in the dose 400 mg/kg.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/tratamiento farmacológico , Pérdida Auditiva Provocada por Ruido/fisiopatología , Metionina/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Animales , Fatiga Auditiva/efectos de los fármacos , Fatiga Auditiva/efectos de la radiación , Catalasa/sangre , Cóclea/efectos de los fármacos , Cóclea/enzimología , Relación Dosis-Respuesta a Droga , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/sangre
11.
J Am Acad Audiol ; 23(5): 332-40, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22533976

RESUMEN

BACKGROUND: Interindividual variation in cochlear vulnerability to noise and ototoxins must in part reflect allelic variation in genes that largely remain unknown. Work in our laboratory has shown that young adult CBA/J mice are more vulnerable to cochlear noise injury than are similar-aged mice of other well-studied strains such as C57BL/6J (B6). Conversely, young CBA/J mice are dramatically protected against noise exposure by low-dose kanamycin (KM) treatment, while B6 mice are not. Genetic differences that distinguish these two strains may include genes that help establish the early "sensitive period" in mammals, as well as genes that shape innate protective responses to stress. These genes may have human homologs that exert similar influences and thereby partly govern individual risk of acquired hearing loss. PURPOSE: We hypothesize that young CBA/J and B6 mice carry different alleles at unknown loci that mediate their characteristic sensitivities to noise and responses to kanamycin. The first step in any experimental genetic analysis of two divergent populations is to examine F1 hybrids formed from these. Accordingly, we evaluated both noise vulnerability and the extent of protection from noise by low-dose KM in 6-wk-old F1 hybrids derived from a B6 × CBA/J cross. STUDY SAMPLE: The study included 52 CBA/J, 59 C57BL/6J (B6), and 45 (B6 × CBA/J) F1 hybrid mice, aged 6 wk at time of noise exposure. Both genders were included. INTERVENTION: For experiments aimed at noise vulnerability, B6 and F1 mice were exposed to loud broadband noise (4-45 kHz, 110 dB SPL) for varying durations, and the resulting noise-induced permanent threshold shifts (NIPTSs, measured 2 wk postnoise) were compared with previous data from CBA/J mice. For experiments aimed at KM-based "protectability," CBA/J, B6, and F1 mice received either kanamycin (300 mg/kg, sc) or saline twice daily for 10 days and then were noise exposed for 30 min, followed by measurement of NIPTS at 2 wk postnoise. DATA COLLECTION AND ANALYSIS: Data comprised auditory brainstem response (ABR) thresholds examined by two-way ANOVA (threshold × frequency, group) and derived metrics for NIPTS, plotted versus noise duration. RESULTS: The "threshold" noise exposure duration for NIPTS in F1 hybrid mice was similar to that in CBA/J. Like CBA/J mice, F1 mice were also significantly protected from noise by KM although the protection appeared less robust than in the CBA/J parent strain. B6 mice appeared harmed by KM alone, even without noise exposure. None of the experimental groups provided any evidence for synergistic interactions between noise and KM. CONCLUSIONS: Our data support the hypothesis that young CBA/J and B6 mice carry different alleles that underlie their divergent responses to KM and sensitivities to noise exposure. While the number and type of genes remain unknown, they are worth pursuing because they establish completely novel hearing phenotypes with potential relevance to humans. Our results lay the foundation for mapping of the underlying genes, and ultimately gene identification.


Asunto(s)
Alelos , Antibacterianos/farmacología , Pérdida Auditiva Provocada por Ruido/genética , Hibridación Genética/genética , Kanamicina/farmacología , Ratones Endogámicos C57BL/genética , Ratones Endogámicos CBA/genética , Ruido/efectos adversos , Animales , Fatiga Auditiva/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Femenino , Inyecciones Subcutáneas , Masculino , Ratones , Espectrografía del Sonido
12.
Acta Otolaryngol ; 127(5): 459-69, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17453470

RESUMEN

CONCLUSION: This study demonstrated differential gene expression profiles in salicylate ototoxicity with oligonucleotide microarray. This study may also provide basic information on candidate genes associated with hearing loss and/or tinnitus or recovery after salicylate-induced cochlear dysfunction. OBJECTIVES: Salicylate ototoxicity is accompanied by temporary hearing loss and tinnitus. The purpose of the present study was to evaluate the gene expression profiles in the mouse cochlea with salicylate ototoxicity using DNA microarray. MATERIALS AND METHODS: The subject mice were injected intraperitoneally with 400 mg/kg of sodium salicylate; an approximate 30 dB threshold shift that was observed by auditory brainstem response was achieved 3 h after an injection of sodium salicylate and the hearing threshold returned to within normal range at 3 days. Differential gene expression profiles at 3 h after salicylate injection in comparison to the normal cochlea were analyzed with DNA microarray technology. RESULTS: No ultrastructural changes in the mice cochlea were observed by TEM at 3 h after salicylate injection. Microarray revealed that 87 genes were up-regulated twofold or more in the mouse cochlea with salicylate ototoxicity in comparison to the normal cochlea. Among these genes, increased expression levels of 30 functional genes were confirmed by semi-quantitative RT-PCR.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Cóclea/efectos de los fármacos , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Salicilato de Sodio/toxicidad , Animales , Fatiga Auditiva/efectos de los fármacos , Cóclea/patología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/efectos de los fármacos
13.
Acta Otolaryngol ; 126(11): 1140-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17050305

RESUMEN

CONCLUSION: Physiological and morphological assessments indicated that low-dose and long-term caroverine delivery might be a new approach to protect against impulse noise-induced hearing loss. BACKGROUND: Although the exact mechanisms by which impulse noise causes hearing loss are still unclear, there is accumulating evidence that increased reactive oxygen species (ROS) production and excessive glutamate released from the inner hair cells lead to hair cell loss and consequently hearing loss. Caroverine is an antagonist of two glutamate receptors, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptors in the inner ear, as well as an antioxidant. MATERIALS AND METHODS: In this study, caroverine was delivered subcutaneously using an osmotic pump. This kind of delivery has the advantage, via continuous, long-term and low dose drug administration, of avoiding systemic side effects. RESULTS: It was shown that caroverine could significantly protect the cochlea against impulse noise trauma.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/prevención & control , Quinoxalinas/farmacología , Animales , Antioxidantes/farmacología , Fatiga Auditiva/efectos de los fármacos , Fatiga Auditiva/fisiología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/patología , Tronco Encefálico/fisiopatología , Relación Dosis-Respuesta a Droga , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Células Ciliadas Auditivas Internas/efectos de los fármacos , Células Ciliadas Auditivas Internas/patología , Células Ciliadas Auditivas Internas/fisiopatología , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Células Ciliadas Auditivas Externas/fisiopatología , Pérdida Auditiva Provocada por Ruido/patología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Inyecciones Subcutáneas , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/fisiología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología
14.
Acta Otolaryngol ; 123(1): 14-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12625567

RESUMEN

OBJECTIVE: The protective effect of brain-derived neurotrophic factor (BDNF) against the ototoxicity resulting from exposure of Pseudomonas aeruginosa exotoxin A (PaExoA) to the middle ear was analyzed. The combined effect of BDNF and N(G)-nitro-L-arginine methyl ester (L-NAME) was also investigated. MATERIAL AND METHODS: Six groups of albino rats were instilled through the tympanic membrane into the round window niche with the following solutions: saline; PaExoA; BDNF; L-NAME; PaExoA + BDNF; and PaExoA + BDNF + L-NAME. Frequency-specific (2-31.5 kHz) auditory brainstem responses were used to obtain the hearing thresholds before and 2, 5 and 15 days after instillation. RESULTS: PaExoA penetrated from the middle ear into the cochlea, causing initially mixed hearing loss, followed by persistent sensorineural hearing loss. This impairment was blocked by BDNF at 6, 8 and 10 kHz on Day 2 and at 8 kHz on Day 5. L-NAME given in combination with BDNF did not show any additional protective effect. There were no significant differences in the thickness of the round window membrane between control ears and those in each instillation group. CONCLUSION: Our results suggest that BDNF may protect against cochlear damage caused by PaExoA in the middle turns of the ear.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Toxinas Bacterianas/toxicidad , Factor Neurotrófico Derivado del Encéfalo/farmacología , Oído Medio/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Exotoxinas/toxicidad , Pérdida Auditiva Sensorineural/fisiopatología , Fármacos Neuroprotectores/farmacología , Percepción de la Altura Tonal/efectos de los fármacos , Factores de Virulencia/toxicidad , Animales , Audiometría de Tonos Puros , Fatiga Auditiva/efectos de los fármacos , Umbral Auditivo/efectos de los fármacos , Nervio Coclear/efectos de los fármacos , Masculino , NG-Nitroarginina Metil Éster/farmacología , Ventana Redonda/efectos de los fármacos , Exotoxina A de Pseudomonas aeruginosa
15.
Eur Arch Otorhinolaryngol ; 257(1): 10-6, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10664038

RESUMEN

The effect of magnesium (Mg) on noise-induced hearing loss was investigated in two groups of adult pigmented guinea pigs maintained either on optimal or suboptimal (physiologically high or low) Mg produced by different diets. The total Mg concentrations of the perilymph (PL), cerebrospinal fluid, blood plasma and red blood cells were measured by atomic absorption spectrometry and were found to differ significantly between the two groups (P < 0.01). One ear of each animal was exposed to either a single shooting impulse at a peak pressure level of 187 dB or two impulse noise series at a rate of 1/s and peak pressure levels of 150 dB (1,000 impulses) and 167 dB (2,280 impulses), respectively. Temporary (TTS) and permanent (PTS) hearing threshold shifts in anesthetized animals were measured 2 h and 1 week after the noise exposure, using auditory brain stem response (ABR) audiometry at a frequency range from 3.75 to 30 kHz. Exposure to the single noise impulse resulted in a mean TTS that was significantly lower in the high Mg group than that in the low Mg group (P < 0.05), although no substantial PTS was observed in either group. In the animals exposed to 150 dB noise, the TTS showed a tendency towards an Mg-related reduction at the higher frequencies. A small difference in PTS was found between the low Mg and high Mg groups, but was not significant. Exposure to the 167-dB noise series caused a considerable TTS, which was significantly lower in the high Mg group at 7.5 and 15 kHz than in the low Mg group (P < 0.05). The mean PTS showed a significant difference between the two Mg groups over the whole frequency range (P < 0.05) and was found to correlate negatively with the total Mg concentrations of both PL and plasma (P < 0.05). Moreover, the high Mg group showed a faster recovery from the hearing threshold shift than the low Mg group. The present findings show that preventive oral Mg supplements can significantly reduce the rate of acoustic trauma caused by high-level impulse noise exposure in the guinea pig.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/prevención & control , Cloruro de Magnesio/farmacología , Animales , Fatiga Auditiva/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Cobayas , Magnesio/sangre , Espectrografía del Sonido
16.
Audiology ; 38(6): 312-20, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10582532

RESUMEN

Auditory brainstem responses (ABRs) were recorded in 60 male or female, anxious or anxiety-free university students, before and after separated or simultaneous intake of alprazolam and exposure to noise. A significant increase of the latencies of the ABRs was found when subjects took alprazolam. This effect is consistent with the presence of gamma-aminobutyric acid (GABA), one of the neurotransmitters at terminals of cochlear efferent fibres A significant increase of the latencies was observed after noise alone. In subjects taking alprazolam when they are exposed to noise, the effect of noise on the ABR latencies is reduced, but not abolished. The effects of alprazolam on the ABR are consistent with the presence of GABA in the medulla and pons. Significant effects of noise upon III-V and I-V intervals suggest that auditory 'fatigue' may involve a retrocochlear component. Differences due to sex appear to be abolished by anxiety.


Asunto(s)
Alprazolam/farmacología , Ansiolíticos/farmacología , Fatiga Auditiva/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Ruido/efectos adversos , Adolescente , Adulto , Ansiedad/fisiopatología , Fatiga Auditiva/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Femenino , Humanos , Masculino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiopatología , Puente/efectos de los fármacos , Puente/fisiología , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Valores de Referencia , Ácido gamma-Aminobutírico/fisiología
17.
Acta Otolaryngol ; 118(1): 52-5, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9504163

RESUMEN

In a previously established animal model a standardized drill-induced injury to the body of the incus was applied, and the effects on hearing were characterized by electrocochleography. In a placebo-controlled, randomized, blinded study methylprednisolone showed no protective effect in reducing or improving the auditory threshold shifts, which occurred within seconds after drilling and remained stable throughout the 5-week observation period. Therefore the otologic surgeon must pay close attention to avoiding any contact of a rotating burr with an ossicle in an intact ossicular chain.


Asunto(s)
Antiinflamatorios/farmacología , Pérdida Auditiva Sensorineural/fisiopatología , Yunque/lesiones , Metilprednisolona/farmacología , Animales , Audiometría de Respuesta Evocada , Fatiga Auditiva/efectos de los fármacos , Método Doble Ciego , Esquema de Medicación , Cobayas , Yunque/efectos de los fármacos , Yunque/fisiopatología , Inyecciones Intraperitoneales , Complicaciones Intraoperatorias/fisiopatología , Instrumentos Quirúrgicos
18.
J Acoust Soc Am ; 104(4): 2261-71, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10491690

RESUMEN

Mongolian gerbils were exposed to either alpha-ketoglutarate, salicylate, or an 8-kHz pure tone. Cochlear microphonic (CM) was recorded from the round window in response to 68 and 88 dB SPL Gaussian noise. A nonlinear systems identification technique provided the frequency-domain parameters of a third-order polynomial model characterizing cochlear mechano-electric transduction (MET). A series of physiologic indices were derived from further exploration of the model. Exposure to the 8-kHz pure tone and round window application of salicylate resulted in different changes in the polynomial parameters and physiologic indices even though the threshold shifts were similar. A general reduction of CM magnitude was found after the tone exposure, and an increase at low-mid frequencies was demonstrated in the salicylate group especially at the lower signal level. The slope of the MET curve was reduced by the acoustic overstimulation. The root or the operating point of the MET was shifted in opposite directions after the two treatments. Sound-pressure levels that saturate MET expanded in the tone exposure group and narrowed in the salicylate group. The signal level also had effects on these indices.


Asunto(s)
Cóclea/fisiopatología , Potenciales Microfónicos de la Cóclea/fisiología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Salicilatos/toxicidad , Animales , Fatiga Auditiva/efectos de los fármacos , Fatiga Auditiva/fisiología , Cóclea/efectos de los fármacos , Potenciales Microfónicos de la Cóclea/efectos de los fármacos , Gerbillinae , Pérdida Auditiva de Alta Frecuencia/inducido químicamente , Pérdida Auditiva de Alta Frecuencia/fisiopatología , Pérdida Auditiva Provocada por Ruido/inducido químicamente , Ácidos Cetoglutáricos/farmacología , Protectores contra Radiación/farmacología
19.
Audiol Neurootol ; 1(6): 339-58, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-9390814

RESUMEN

When anaesthetized animals are maintained over a long period, crossed-cochlear suppressive and enhancement-in-noise effects mediated by the olivocochlear bundle (OCB), as well as some OCB neuronal responses, show time-dependent variations. The present study determined if there were any such changes in OCB-mediated crossed-cochlear protection against compound action potential (CAP) threshold losses caused by a standard loud sound exposure at 11 kHz, presented under conditions either not evoking OCB-mediated protection (i.e. monaural exposure) or evoking protection (binaural exposure). Maintaining animals for periods up to approximately 30 h from initial anaesthetization resulted in non-significant changes in pre-exposure CAP thresholds. There were also only small changes over select frequency ranges in threshold losses caused by the monaural or binaural loud sound, after a single exposure as well as when the testing of OCB function was extended to examine effects after dual successive exposures, the latter result being determined by application of a previously described additivity model. The features of OCB-mediated protection also showed good stability over the long maintenance. These results are discussed as providing further circumstantial evidence that protection is mediated by a different OCB subcomponent to that/those responsible for other OCB-mediated crossed-cochlear effects. In general, the results show that the barbiturate anaesthetic used here does not significantly modulate the crossed-cochlear OCB effect of protection, even though it has been shown elsewhere to significantly depress other crossed-cochlear OCB effects.


Asunto(s)
Fatiga Auditiva/fisiología , Nervio Coclear/fisiopatología , Núcleo Coclear/fisiopatología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Núcleo Olivar/fisiopatología , Anestesia General , Animales , Fatiga Auditiva/efectos de los fármacos , Gatos , Nervio Coclear/efectos de los fármacos , Núcleo Coclear/efectos de los fármacos , Vías Eferentes/efectos de los fármacos , Vías Eferentes/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Hipnóticos y Sedantes/farmacología , Núcleo Olivar/efectos de los fármacos , Pentobarbital/farmacología
20.
Laryngoscope ; 106(6): 772-6, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8656966

RESUMEN

Recent data suggest that diltiazem reduces noise-induced hearing loss. Our study was designed to replicate and extend the results of Maurer et al. by using the gerbil as a model. In experiment A, subjects received diltiazem (30 mg/kg/day intraperitoneally) or saline for 3 days. After peripheral thresholds were measured, each subject was exposed to a 4-kHz tone (90-dB sound pressure level) for 20 minutes. Similar amounts of temporary threshold shifts (ITS) were measured in the saline and diltiazem groups. In experiment B, subjects were given saline or diltiazem (30 mg/kg/day intraperitoneally) for 3 days and then exposed to an octave band of noise centered at 4 kHz for 5 days, during which time the subjects continued to receive the drug or saline. The TTS and permanent threshold shifts were similar in the two groups. Measures of cochlear nonlinearities also showed no effect of diltiazem, suggesting that diltiazem does not protect the ear from the effects of noise.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Diltiazem/farmacología , Pérdida Auditiva Provocada por Ruido/prevención & control , Animales , Fatiga Auditiva/efectos de los fármacos , Fatiga Auditiva/fisiología , Gerbillinae , Pérdida Auditiva Provocada por Ruido/fisiopatología , Inyecciones Intraperitoneales , Emisiones Otoacústicas Espontáneas/efectos de los fármacos , Emisiones Otoacústicas Espontáneas/fisiología , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA