Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Hear Res ; 447: 109013, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38718672

RESUMEN

Cisplatin, a highly effective chemotherapeutic drug for various human cancers, induces irreversible sensorineural hearing loss as a side effect. Currently there are no highly effective clinical strategies for the prevention of cisplatin-induced ototoxicity. Previous studies have indicated that short-term cisplatin ototoxicity primarily affects the outer hair cells of the cochlea. Therefore, preventing the entry of cisplatin into hair cells may be a promising strategy to prevent cisplatin ototoxicity. This study aimed to investigate the entry route of cisplatin into mouse cochlear hair cells. The competitive inhibitor of organic cation transporter 2 (OCT2), cimetidine, and the sensory mechanoelectrical transduction (MET) channel blocker benzamil, demonstrated a protective effect against cisplatin toxicity in hair cells in cochlear explants. Sensory MET-deficient hair cells explanted from Tmc1Δ;Tmc2Δ mice were resistant to cisplatin toxicity. Cimetidine showed an additive protective effect against cisplatin toxicity in sensory MET-deficient hair cells. However, in the apical turn, cimetidine, benzamil, or genetic ablation of sensory MET channels showed limited protective effects, implying the presence of other entry routes for cisplatin to enter the hair cells in the apical turn. Systemic administration of cimetidine failed to protect cochlear hair cells from ototoxicity caused by systemically administered cisplatin. Notably, outer hair cells in MET-deficient mice exhibited no apparent deterioration after systemic administration of cisplatin, whereas the outer hair cells in wild-type mice showed remarkable deterioration. The susceptibility of mouse cochlear hair cells to cisplatin ototoxicity largely depends on the sensory MET channel both ex vivo and in vivo. This result justifies the development of new pharmaceuticals, such as a specific antagonists for sensory MET channels or custom-designed cisplatin analogs which are impermeable to sensory MET channels.


Asunto(s)
Antineoplásicos , Cimetidina , Cisplatino , Mecanotransducción Celular , Transportador 2 de Cátion Orgánico , Ototoxicidad , Cisplatino/toxicidad , Animales , Ototoxicidad/prevención & control , Ototoxicidad/metabolismo , Ototoxicidad/fisiopatología , Mecanotransducción Celular/efectos de los fármacos , Transportador 2 de Cátion Orgánico/metabolismo , Transportador 2 de Cátion Orgánico/genética , Transportador 2 de Cátion Orgánico/antagonistas & inhibidores , Cimetidina/farmacología , Antineoplásicos/toxicidad , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Células Ciliadas Auditivas Externas/metabolismo , Ratones Endogámicos C57BL , Ratones , Proteínas de la Membrana
2.
Biomed Res Int ; 2021: 5590973, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34095303

RESUMEN

Cisplatin is an efficacious anticancer agent, but its use is limited by ototoxicity and resultant irreversible sensorineural hearing loss. Cisplatin ototoxicity is associated with cochlear cell oxidative stress and mitochondrial damage. However, mitophagy is vital for maintaining mitochondrial quality and cellular metabolism. Accordingly, we investigated the role of mitophagy in regulating cisplatin-induced ototoxicity using the auditory cell line HEI-OC1. In this study, HEI-OC1 cells were treated with either cisplatin alone (10 µM, 0, 8, 16, and 24 h); cisplatin (10 µM, 24 h) post transfection with small-interfering (si)RNAs targeting mitophagy-associated mRNAs; cisplatin (10 µM, 24 h) succeeding pretreatment with the mitophagy suppressor, 3-methyladenine (3-MA; 5 or 10 mM, 6 h); or cisplatin (30 µM, 24 h) following pretreatment with the mitophagy promoter, carbonyl cyanide m-chlorophenylhydrazone (CCCP; 1 or 2 µM, 2 h). The viability of cells, expression of mitophagy marker, and mitochondrial functions were then assessed in these cells. Cell viability was determined by a water-soluble tetrazolium assay; expression of mitophagy-associated proteins PINK1, Parkin, BNIP3, FUNDC1, p62, and LC3B was analyzed by Western blotting, mitochondrial membrane potential by flow cytometry, intracellular ATP by spectrophotometry, and mitochondrial degradation by dual staining for mitochondria and autophagosomes or lysosomes. Our results showed that cisplatin gradually reduced the viable cell number over time, induced mitochondrial depolarization, decreased intracellular ATP concentration, and enhanced the expression of PINK1, Parkin, BNIP3, p62, and LC3B. In addition, Parkin and BNIP3 knockdown accelerated cisplatin-induced loss of cell viability, mitochondrial membrane potential, mitophagosome/lysosome formation, and reduction in intracellular ATP production. Pretreatment with 3-MA aggravated the cisplatin-induced cytotoxicity, while that with CCCP reversed this effect. Overall, our findings indicate that mitophagy protects HEI-OC1 cells against cisplatin-induced cell death. Consequently, we strongly believe that targeted promotion of mitophagy may confer protection against cisplatin-induced ototoxicity.


Asunto(s)
Cisplatino/efectos adversos , Mitofagia/fisiología , Ototoxicidad/fisiopatología , Apoptosis/efectos de los fármacos , Autofagosomas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Pérdida Auditiva Sensorineural/prevención & control , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Ototoxicidad/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Toxicol Lett ; 349: 115-123, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34089817

RESUMEN

Cisplatin, the most widely used platinum-based anticancer drug, often causes progressive and irreversible sensorineural hearing loss in cancer patients. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. Nicotinamide adenine dinucleotide (NAD+), a co-substrate for the sirtuin family and PARPs, has emerged as a potent therapeutic molecular target in various diseases. In our investigates, we observed that NAD+ level was changed in the cochlear explants of mice treated with cisplatin. Supplementation of a specific inhibitor (TES-1025) of α-amino-ß-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), a rate-limiting enzyme of NAD+de novo synthesis pathway, promoted SIRT1 activity, increased mtDNA contents and enhanced AMPK expression, thus significantly reducing hair cells loss and deformation. The protection was blocked by EX527, a specific SIRT1 inhibitor. Meanwhile, the use of NMN, a precursor of NAD+ salvage synthesis pathway, had shown beneficial effect on hair cell under cisplatin administration, effectively suppressing PARP1. In vivo experiments confirmed the hair cell protection of NAD+ modulators in cisplatin treated mice and zebrafish. In conclusion, we demonstrated that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway could both prevent ototoxicity of cisplatin. These results suggested that direct modulation of cellular NAD+ levels could be a promising therapeutic approach for protection of hearing from cisplatin-induced ototoxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Células Ciliadas Auditivas/efectos de los fármacos , Pérdida Auditiva/prevención & control , Audición/efectos de los fármacos , NAD/biosíntesis , Ototoxicidad/prevención & control , Sirtuina 1/metabolismo , Animales , Animales Modificados Genéticamente , Carboxiliasas/antagonistas & inhibidores , Carboxiliasas/metabolismo , Cisplatino , Modelos Animales de Enfermedad , Activación Enzimática , Células Ciliadas Auditivas/enzimología , Células Ciliadas Auditivas/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/enzimología , Pérdida Auditiva/fisiopatología , Sistema de la Línea Lateral/efectos de los fármacos , Sistema de la Línea Lateral/enzimología , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/patología , Ototoxicidad/enzimología , Ototoxicidad/etiología , Ototoxicidad/fisiopatología , Pez Cebra
4.
Biomed Environ Sci ; 34(2): 110-118, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33685569

RESUMEN

OBJECTIVE: The aim of this study was to explore the ototoxicity of toluene in the early development of zebrafish embryos/larvae. METHODS: Zebrafish were utilized to explore the ototoxicity of toluene. Locomotion analysis, immunofluorescence, and qPCR were used to understand the phenotypes and molecular mechanisms of toluene ototoxicity. RESULTS: The results demonstrated that at 2 mmol/L, toluene induced zebrafish larvae death at 120 hours post fertilization (hpf) at a rate of 25.79% and inhibited the rate of hatching at 72 hpf. Furthermore, toluene exposure inhibited the distance travelled and average swimming velocity of zebrafish larvae while increasing the frequency of movements. As shown by fluorescence staining of hair cells, toluene inhibited the formation of lateral line neuromasts and middle line 1 (Ml 1) neuromasts in 3 days post fertilization larvae in a concentration-dependent manner. Toluene altered the expression level of genes involved in ear development/function in zebrafish, among which the mRNA levels of cd164l2, tekt3, and pcsk5a were upregulated, while the level of otofb was downregulated, according to the qPCR results. CONCLUSION: This study indicated that toluene may affect the development of both the inner ear and lateral line systems in zebrafish, while the lateral line system may be more sensitive to toluene than the inner ear.


Asunto(s)
Oído Interno/efectos de los fármacos , Sistema de la Línea Lateral/efectos de los fármacos , Tolueno/toxicidad , Animales , Oído Interno/crecimiento & desarrollo , Embrión no Mamífero/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Sistema de la Línea Lateral/crecimiento & desarrollo , Locomoción/efectos de los fármacos , Ototoxicidad/etiología , Ototoxicidad/patología , Ototoxicidad/fisiopatología , Pez Cebra
5.
Arch Toxicol ; 95(3): 1003-1021, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495873

RESUMEN

The peripheral auditory and vestibular systems rely on sensorineural structures that are vulnerable to ototoxic agents that cause hearing loss and/or equilibrium deficits. Although attention has focused on hair cell loss as the primary pathology underlying ototoxicity, evidence from the peripheral vestibular system indicates that hair cell loss during chronic exposure is preceded by synaptic uncoupling from the neurons and is potentially reversible. To determine if synaptic pathology also occurs in the peripheral auditory system, we examined the extent, time course, and reversibility of functional and morphological alterations in cochleae from mice exposed to 3,3'-iminodipropionitrile (IDPN) in drinking water for 2, 4 or 6 weeks. Functionally, IDPN exposure caused progressive high- to low-frequency hearing loss assessed by measurement of auditory brainstem response wave I absolute thresholds and amplitudes. The extent of hearing loss scaled with the magnitude of vestibular dysfunction assessed behaviorally. Morphologically, IDPN exposure caused progressive loss of outer hair cells (OHCs) and synapses between the inner hair cells (IHCs) and primary auditory neurons. In contrast, IHCs were spared from ototoxic damage. Importantly, hearing loss consistent with cochlear synaptopathy preceded loss of OHCs and synapses and, moreover, recovered if IDPN exposure was stopped before morphological pathology occurred. Our observations suggest that synaptic uncoupling, perhaps as an early phase of cochlear synaptopathy, also occurs in the peripheral auditory system in response to IDPN exposure. These findings identify novel mechanisms that contribute to the earliest stages of hearing loss in response to ototoxic agents and possibly other forms of acquired hearing loss.


Asunto(s)
Cóclea/efectos de los fármacos , Pérdida Auditiva/inducido químicamente , Nitrilos/toxicidad , Ototoxicidad/etiología , Animales , Cóclea/patología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Células Ciliadas Auditivas Internas/efectos de los fármacos , Células Ciliadas Auditivas Internas/patología , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Pérdida Auditiva/fisiopatología , Masculino , Ratones , Ratones de la Cepa 129 , Nitrilos/administración & dosificación , Ototoxicidad/fisiopatología , Sinapsis/efectos de los fármacos , Sinapsis/patología , Factores de Tiempo
6.
Cancer Biol Ther ; 21(11): 990-993, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33121320

RESUMEN

Docetaxel is an important anti-microtubule agent used to treat a variety of solid tumors, including breast cancer; notably, docetaxel-containing regimens improve outcomes for patients in metastatic, adjuvant, and neoadjuvant settings. However, the effectiveness of docetaxel in clinical practice can be compromised by suboptimal management of side effects. Here, we report two cases of docetaxel-based chemotherapy regimens in patients who exhibited invasive ductal breast cancer and underwent two different clinical treatment approaches. A 58-year-old postmenopausal female received salvage treatment with 8 cycles of docetaxel (67 mg/m2), and a 74-year-old female received 1 cycle of docetaxel (100 mg/m2). The two patients exhibited considerable hearing loss two days later. Of note, both patients had no hearing loss symptoms prior to docetaxel. Thus, ototoxicity may be a side effect of docetaxel that should be considered during treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/complicaciones , Docetaxel/efectos adversos , Ototoxicidad/etiología , Antineoplásicos/farmacología , Docetaxel/farmacología , Femenino , Humanos , Persona de Mediana Edad , Ototoxicidad/fisiopatología
7.
Bull Exp Biol Med ; 169(4): 458-462, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32894392

RESUMEN

We studied possible otoprotective effect of drugs widely used for the correction of perinatal hypoxic brain damage in premature infants. The experiments were carried out on immature rabbits with an immature hearing organ. The auditory function was assessed by DPOAE and ABR methods in intact animals and rabbits treated with therapeutic doses of netromycin alone or in combination with the drugs that normalize metabolic processes in the brain (Cortexin, Cogitum, Elkar, vitamin B2, ATP, and cocarboxylase). It was found that the administered drugs produced an otoprotective effect and reduced the severity, but did not eliminate the ototoxic effect.


Asunto(s)
Ácido Aspártico/análogos & derivados , Carnitina/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Netilmicina/efectos adversos , Ototoxicidad/prevención & control , Sustancias Protectoras/farmacología , Adenosina Trifosfato/farmacología , Animales , Animales Recién Nacidos , Ácido Aspártico/farmacología , Carnitina/análogos & derivados , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Netilmicina/antagonistas & inhibidores , Emisiones Otoacústicas Espontáneas/efectos de los fármacos , Emisiones Otoacústicas Espontáneas/fisiología , Ototoxicidad/fisiopatología , Conejos , Riboflavina/farmacología , Tiamina Pirofosfato/farmacología
8.
FASEB J ; 34(10): 13978-13992, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32840016

RESUMEN

The chemotherapeutic agent cisplatin is renowned for its ototoxic effects. While hair cells in the cochlea are established targets of cisplatin, less is known regarding the afferent synapse, which is an essential component in the faithful temporal transmission of sound. The glutamate aspartate transporter (GLAST) shields the auditory synapse from excessive glutamate release, and its loss of function increases the vulnerability to noise, salicylate, and aminoglycosides. Until now, the involvement of GLAST in cisplatin-mediated ototoxicity remains unknown. Here, we test in mice lacking GLAST the effects of a low-dose cisplatin known not to cause any detectable change in hearing thresholds. When administered at nighttime, a mild hearing loss in GLAST KO mice was found but not at daytime, revealing a potential circadian regulation of the vulnerability to cisplatin-mediated ototoxicity. We show that the auditory synapse of GLAST KO mice is more vulnerable to cisplatin administration during the active phase (nighttime) when compared to WT mice and treatment during the inactive phase (daytime). This effect was not related to the abundance of platinum compounds in the cochlea, rather cisplatin had a dose-dependent impact on cochlear clock rhythms only after treatment at nighttime suggesting that cisplatin can modulate the molecular clock. Our findings suggest that the current protocols of cisplatin administration in humans during daytime may cause a yet undetectable damage to the auditory synapse, more so in already damaged ears, and severely impact auditory sensitivity in cancer survivors.


Asunto(s)
Antineoplásicos/toxicidad , Ritmo Circadiano , Cisplatino/toxicidad , Ototoxicidad/genética , Animales , Umbral Auditivo , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico , Transportador 1 de Aminoácidos Excitadores/deficiencia , Transportador 1 de Aminoácidos Excitadores/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ototoxicidad/etiología , Ototoxicidad/fisiopatología
9.
Int J Mol Sci ; 21(10)2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32429117

RESUMEN

Previous studies have described the effects of zingerone (ZO) on cisplatin (CXP)-induced injury to the kidneys, liver, and other organs but not to the cochlea. This study aimed to investigate the effects of ZO on CXP-induced ototoxicity. Eight-week-old Sprague-Dawley rats were used and divided into a control group, a CXP group, and a CXP + ZO group. Rats in the CXP group received 5 mg/kg/day CXP intraperitoneally for five days. Rats in the CXP + ZO group received 5 mg/kg/day CXP intraperitoneally for five days and 50 mg/kg/day ZO intraperitoneally for seven days. Auditory brainstem response thresholds (ABRTs) were measured before (day 0) and after (day 10) drug administration. Cochlear histology was examined using hematoxylin and eosin (H&E) staining and cochlear whole mounts. The expression levels of cytochrome P450 (CYP)1A1, CYP1B1, inducible nitric oxide synthase (iNOS), nuclear factor kappa B (NFκB), tumor necrosis factor alpha (TNFα), and interleukin 6 (IL6) were estimated using quantitative reverse transcription-polymerase chain reaction. The expression levels of heme oxygenase 1 (HO1) and caspase 3 were analyzed via Western blotting. The auditory thresholds at 4, 8, and 16 kHz were attenuated in the CXP + ZO group compared with the CXP group. The mRNA expression levels of CYP1A1, CYP1B1, iNOS, NFκB, TNFα, and IL6 were lower in the CXP + ZO group than in the CXP group. The protein expression levels of HO1 and caspase 3 were lower in the CXP + ZO group than in the CXP group. Cotreatment with ZO exerted otoprotective effects against CXP-induced cochlear injury via antioxidative and anti-inflammatory activities involving CYPs, iNOS, NFκB, and TNFα.


Asunto(s)
Cisplatino/efectos adversos , Guayacol/análogos & derivados , Ototoxicidad/tratamiento farmacológico , Sustancias Protectoras/uso terapéutico , Animales , Umbral Auditivo/efectos de los fármacos , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Cóclea/patología , Cóclea/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Guayacol/farmacología , Guayacol/uso terapéutico , Ototoxicidad/genética , Ototoxicidad/fisiopatología , Sustancias Protectoras/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley
10.
J Int Adv Otol ; 16(1): 77-86, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31287435

RESUMEN

OBJECTIVES: Nilotinib has very few side effects, including neutropenia, thrombocytopenia, cardiotoxicity, high pancreatic lipase, ischemia, and vascular occlusion. We aimed to investigate whether short-term administration of nilotinib had ototoxic effects in rats. MATERIALS AND METHODS: Wistar-albino rats were categorized into three groups: group C (administered 0.25 mL of distilled water, no nilotinib), group N-20 (administered 20 mg/kg/day of nilotinib dissolved in distilled water), and group N-50 (administered 50 mg/kg/day of nilotinib dissolved in distilled water). A single dose was administered once per day, at the same hour, over 21 days. Auditory brainstem response (ABR) thresholds were recorded on day 0 and day 21. RESULTS: There were no changes in ABR threshold values obtained on day 0 (baseline) and on day 21 across all three groups. A statistically significant difference was not found in terms of the mean latency of waves V and III, interpeak latency values of waves III-V, and amplitude ratios of waves III-V and V/Va at baseline and on day 21 across all three groups on within-group or between-group evaluation. CONCLUSION: Consequently, further studies are needed that involve different drug doses, prolonged administration of drugs, as well as distortion otoacoustic emission test for the evaluation of cochlear activation and ABR. Furthermore, histopathological studies are needed to indicate whether the cochlea is affected to prove that nilotinib has definitively no ototoxic effect.


Asunto(s)
Cóclea/efectos de los fármacos , Audición/efectos de los fármacos , Pirimidinas/efectos adversos , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Animales , Umbral Auditivo/fisiología , Cóclea/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Audición/fisiología , Masculino , Modelos Animales , Emisiones Otoacústicas Espontáneas/fisiología , Ototoxicidad/fisiopatología , Ototoxicidad/veterinaria , Pirimidinas/administración & dosificación , Ratas , Ratas Wistar
11.
Molecules ; 24(21)2019 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671767

RESUMEN

In this study, we investigated whether the curcuminoids, CLEFMA and EF24, improved cisplatin efficacy and reduced cisplatin ototoxicity. We used the lung cancer cell line, A549, to determine the effects of the curcuminoids and cisplatin on cell viability and several apoptotic signaling mechanisms. Cellular viability was measured using the MTT assay. A scratch assay was used to measure cell migration and fluorescent spectrophotometry to measure reactive oxygen species (ROS) production. Western blots and luminescence assays were used to measure the expression and activity of apoptosis-inducing factor (AIF), caspases-3/7, -8, -9, and -12, c-Jun N-terminal kinases (JNK), mitogen-activated protein kinase (MAPK), and proto-oncogene tyrosine-protein kinase (Src). A zebrafish model was used to evaluate auditory effects. Cisplatin, the curcuminoids, and their combinations had similar effects on cell viability (IC50 values: 2-16 µM) and AIF, caspase-12, JNK, MAPK, and Src expression, while caspase-3/7, -8, and -9 activity was unchanged or decreased. Cisplatin increased ROS yield (1.2-fold), and curcuminoid and combination treatments reduced ROS (0.75-0.85-fold). Combination treatments reduced A549 migration (0.51-0.53-fold). Both curcuminoids reduced auditory threshold shifts induced by cisplatin. In summary, cisplatin and the curcuminoids might cause cell death through AIF and caspase-12. The curcuminoids may potentiate cisplatin's effect against A549 migration, but may counteract cisplatin's effect to increase ROS production. The curcuminoids might also prevent cisplatin ototoxicity.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos de Bencilideno/uso terapéutico , Cisplatino/efectos adversos , Diarilheptanoides/uso terapéutico , Ototoxicidad/tratamiento farmacológico , Piperidonas/uso terapéutico , Células A549 , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Compuestos de Bencilideno/química , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Cisplatino/química , Diarilheptanoides/química , Diarilheptanoides/farmacología , Potenciales Evocados Auditivos/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Ototoxicidad/fisiopatología , Piperidonas/química , Proto-Oncogenes Mas , Pez Cebra
12.
JCI Insight ; 4(22)2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31723056

RESUMEN

BACKGROUNDBilateral loss of vestibular (inner ear inertial) sensation causes chronically blurred vision during head movement, postural instability, and increased fall risk. Individuals who fail to compensate despite rehabilitation therapy have no adequate treatment options. Analogous to hearing restoration via cochlear implants, prosthetic electrical stimulation of vestibular nerve branches to encode head motion has garnered interest as a potential treatment, but prior studies in humans have not included continuous long-term stimulation or 3D binocular vestibulo-ocular reflex (VOR) oculography, without which one cannot determine whether an implant selectively stimulates the implanted ear's 3 semicircular canals.METHODSWe report binocular 3D VOR responses of 4 human subjects with ototoxic bilateral vestibular loss unilaterally implanted with a Labyrinth Devices Multichannel Vestibular Implant System vestibular implant, which provides continuous, long-term, motion-modulated prosthetic stimulation via electrodes in 3 semicircular canals.RESULTSInitiation of prosthetic stimulation evoked nystagmus that decayed within 30 minutes. Stimulation targeting 1 canal produced 3D VOR responses approximately aligned with that canal's anatomic axis. Targeting multiple canals yielded responses aligned with a vector sum of individual responses. Over 350-812 days of continuous 24 h/d use, modulated electrical stimulation produced stable VOR responses that grew with stimulus intensity and aligned approximately with any specified 3D head rotation axis.CONCLUSIONThese results demonstrate that a vestibular implant can selectively, continuously, and chronically provide artificial sensory input to all 3 implanted semicircular canals in individuals disabled by bilateral vestibular loss, driving reflexive VOR eye movements that approximately align in 3D with the head motion axis encoded by the implant.TRIAL REGISTRATIONClinicalTrials.gov: NCT02725463.FUNDINGNIH/National Institute on Deafness and Other Communication Disorders: R01DC013536 and 2T32DC000023; Labyrinth Devices, LLC; and Med-El GmbH.


Asunto(s)
Vestibulopatía Bilateral , Estimulación Eléctrica/instrumentación , Prótesis Neurales , Reflejo Vestibuloocular/fisiología , Vestíbulo del Laberinto , Vestibulopatía Bilateral/fisiopatología , Vestibulopatía Bilateral/cirugía , Humanos , Ototoxicidad/fisiopatología , Ototoxicidad/cirugía , Diseño de Prótesis , Vestíbulo del Laberinto/fisiopatología , Vestíbulo del Laberinto/cirugía
13.
Audiol Neurootol ; 24(5): 253-257, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31661686

RESUMEN

BACKGROUND: The most common complaint of patients affected by chemotherapy-induced hearing loss is difficulty understanding speech in noisy environments despite the use of hearing aids. Cochlear dead regions, those areas with damaged or absent inner hair cells and dendrites, may account for this type of hearing loss. However, it is unknown whether this condition is associated with cisplatin agents. OBJECTIVE: The aim of this study was to determine whether cisplatin is associated with hearing loss and cochlear dead regions. METHODS: This prospective cross-sectional study was conducted in patients participating in routine audiological monitoring during and after chemotherapy treatment. Adults undergoing audiological evaluation who had completed chemotherapy treatment were invited to participate. Patients were divided into 3 groups according to pure tone thresholds. Group 1 patients had thresholds over 70 dB (HL) at 2,000 Hz and higher frequencies. Group 2 patients had thresholds below 70 dB (HL) up to 2,000 Hz. Patients in the control group had normal thresholds at all frequencies. The threshold equalizing noise test (TEN[HL]) was used to identify cochlear dead regions by repeating thresholds in the presence of TEN noise played from a compact disc. The presence of cochlear dead regions was established when the masked threshold was 10 dB or greater above the TEN level and 10 dB or greater above the absolute threshold at any frequency. RESULTS: Twelve patients were included in study group 1, 10 patients in study group 2, and 7 patients in the control group. Cochlear dead regions were present in all patients with hearing loss and in none of the control group. For groups 1 and 2, mean differences between absolute and masked thresholds were 21 and 16 dB at 500 Hz; 22 and 15 dB at 1,000 Hz; 31 and 17 dB at 2,000 Hz; 32 and 20 dB at 3,000 Hz; and 31 and 21 dB at 4,000 Hz, respectively. Nevertheless, analysis of variance testing with Bonferroni analysis showed a difference between groups 1 and 2 only at 2,000, 3,000, and 4,000 Hz. CONCLUSION: We found unresponsive or dead cochlear regions in patients who had undergone cisplatin chemotherapy even among patients with mild to moderate hearing loss.


Asunto(s)
Umbral Auditivo/fisiología , Cisplatino/efectos adversos , Cóclea/fisiopatología , Pérdida Auditiva/fisiopatología , Ototoxicidad/fisiopatología , Adolescente , Adulto , Anciano , Audiometría , Umbral Auditivo/efectos de los fármacos , Cóclea/efectos de los fármacos , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Adulto Joven
14.
Nat Commun ; 10(1): 4150, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515474

RESUMEN

Cisplatin is one of the most widely used chemotherapeutic drugs for the treatment of cancer. Unfortunately, one of its major side effects is permanent hearing loss. Here, we show that glutathione transferase α4 (GSTA4), a member of the Phase II detoxifying enzyme superfamily, mediates reduction of cisplatin ototoxicity by removing 4-hydroxynonenal (4-HNE) in the inner ears of female mice. Under cisplatin treatment, loss of Gsta4 results in more profound hearing loss in female mice compared to male mice. Cisplatin stimulates GSTA4 activity in the inner ear of female wild-type, but not male wild-type mice. In female Gsta4-/- mice, cisplatin treatment results in increased levels of 4-HNE in cochlear neurons compared to male Gsta4-/- mice. In CBA/CaJ mice, ovariectomy decreases mRNA expression of Gsta4, and the levels of GSTA4 protein in the inner ears. Thus, our findings suggest that GSTA4-dependent detoxification may play a role in estrogen-mediated neuroprotection.


Asunto(s)
Cisplatino/efectos adversos , Glutatión Transferasa/metabolismo , Ototoxicidad/enzimología , Animales , Umbral Auditivo/efectos de los fármacos , Capilares/patología , Cóclea/enzimología , Cóclea/patología , Cóclea/fisiopatología , Cruzamientos Genéticos , Daño del ADN/genética , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión Transferasa/deficiencia , Pérdida Auditiva/complicaciones , Pérdida Auditiva/enzimología , Pérdida Auditiva/fisiopatología , Masculino , Ratones Endogámicos CBA , Ototoxicidad/complicaciones , Ototoxicidad/patología , Ototoxicidad/fisiopatología , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/patología
15.
J Int Adv Otol ; 15(2): 237-246, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31347504

RESUMEN

OBJECTIVES: The aim of our study was to investigate the effects of folic acid on cisplatin-induced ototoxicity. MATERIALS AND METHODS: Thirty Wistar albino rats were divided into five groups. Group I received intraperitoneal cisplatin (IP) 10 mg/kg/day and IP folic acid 10 mg/kg/day; Group II received IP cisplatin 10 mg/kg/day and IP physiological saline; Group III received IP cisplatin 10 mg/kg/day and intratympanic (IT) folic acid 0.15 mL/day; Group IV received IP cisplatin 10 mg/kg/day and IT physiological saline; and Group V received IT folic acid 0.15 mL/day. Before and after drug administration, plasma homocysteine, folic acid levels, and auditory brainstem evoked responses (ABR) were measured. The rats were then sacrificed, and the inner ears were processed for electron microscopy. RESULTS: The differences of ABR thresholds in Group I compared to Group II were significantly smaller at 4 kHz, 8 kHz, and 16 kHz, whereas they were smaller but not statistically significant at 12 kHz in ABR. The differences of ABR thresholds in Group III compared to Group IV were significantly smaller at 12 kHz, and smaller but not statistically significant at 4 kHz, 8 kHz, and 16 kHz. Cisplatin treatment resulted in the degeneration of the cells of the organ of Corti, stria vascularis, and spiral ganglion. The cells of the organ of Corti, stria vascularis, and spiral ganglion showed a partially preserved morphology in both Group I and Group III. CONCLUSION: Our study results suggests that folic acid is a potential agent in preventing cisplatin-induced ototoxicity.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Ácido Fólico/farmacología , Ototoxicidad/prevención & control , Complejo Vitamínico B/farmacología , Animales , Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Cóclea/patología , Esquema de Medicación , Potenciales Evocados Auditivos/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Ácido Fólico/metabolismo , Células Ciliadas Auditivas/ultraestructura , Pérdida Auditiva/prevención & control , Homocisteína/metabolismo , Masculino , Microscopía Electrónica , Órgano Espiral/patología , Ototoxicidad/patología , Ototoxicidad/fisiopatología , Ratas Wistar , Umbral Sensorial/fisiología
16.
Arch Toxicol ; 93(5): 1385-1399, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30963202

RESUMEN

Individuals treated for multidrug-resistant tuberculosis (MDR-TB) with aminoglycosides (AGs) in resource-limited settings often experience permanent hearing loss. However, AG ototoxicity has never been conceptually integrated or causally linked to MDR-TB patients' pre-treatment health condition. We sought to develop a framework that examines the relationships between pre-treatment conditions and AG-induced hearing loss among MDR-TB-infected individuals in sub-Saharan Africa. The adverse outcome pathway (AOP) approach was used to develop a framework linking key events (KEs) within a biological pathway that results in adverse outcomes (AO), which are associated with chemical perturbation of a molecular initiating event (MIE). This AOP describes pathways initiating from AG accumulation in hair cells, sound transducers of the inner ear immediately after AG administration. After administration, the drug catalyzes cellular oxidative stress due to overproduction of reactive oxygen species. Since oxidative stress inhibits mitochondrial protein synthesis, hair cells undergo apoptotic cell death, resulting in irreversible hearing loss (AO). We identified the following pre-treatment conditions that worsen the causal linkage between MIE and AO: HIV, malnutrition, aging, noise, smoking, and alcohol use. The KEs are: (1) nephrotoxicity, pre-existing hearing loss, and hypoalbuminemia that catalyzes AG accumulation; (2) immunodeficiency and antioxidant deficiency that trigger oxidative stress pathways; and (3) co-administration of mitochondrial toxic drugs that hinder mitochondrial protein synthesis, causing apoptosis. This AOP clearly warrants the development of personalized interventions for patients undergoing MDR-TB treatment. Such interventions (i.e., choosing less ototoxic drugs, scheduling frequent monitoring, modifying nutritional status, avoiding poly-pharmacy) will be required to limit the burden of AG ototoxicity.


Asunto(s)
Aminoglicósidos/efectos adversos , Antituberculosos/efectos adversos , Ototoxicidad/etiología , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Rutas de Resultados Adversos , África del Sur del Sahara , Aminoglicósidos/administración & dosificación , Antituberculosos/administración & dosificación , Apoptosis/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/fisiopatología , Humanos , Ototoxicidad/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
17.
Hear Res ; 375: 66-74, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30827780

RESUMEN

Cisplatin-induced ototoxicity results in significant, permanent hearing loss in pediatric and adult cancer survivors. Elucidating the mechanisms underlying cisplatin-induced hearing loss as well as the development of therapies to reduce and/or reverse cisplatin ototoxicity have been impeded by suboptimal animal models. Clinically, cisplatin is most commonly administered in multi-dose, multi-cycle protocols. However, many animal studies are conducted using single injections of high-dose cisplatin, which is not reflective of clinical cisplatin administration protocols. Significant limitations of both high-dose, single-injection protocols and previous multi-dose protocols in rodent models include high mortality rates and relatively small changes in hearing sensitivity. These limitations restrict assessment of both long-term changes in hearing sensitivity and effects of potential protective therapies. Here, we present a detailed method for an optimized mouse model of cisplatin ototoxicity that utilizes a multi-cycle administration protocol that better approximates the type and degree of hearing loss observed clinically. This protocol results in significant hearing loss with very low mortality. This mouse model of cisplatin ototoxicity provides a platform for examining mechanisms of cisplatin-induced hearing loss as well as developing therapies to protect the hearing of cancer patients receiving cisplatin therapy.


Asunto(s)
Cisplatino/toxicidad , Ototoxicidad/etiología , Animales , Umbral Auditivo/efectos de los fármacos , Cisplatino/administración & dosificación , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Esquema de Medicación , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Femenino , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Humanos , Masculino , Ratones , Ratones Endogámicos CBA , Emisiones Otoacústicas Espontáneas/efectos de los fármacos , Ototoxicidad/patología , Ototoxicidad/fisiopatología
18.
Hear Res ; 374: 24-34, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30703625

RESUMEN

Auditory-nerve fibers are lost steadily with age and as a possible consequence of noise-induced glutamate excitotoxicity. Auditory-nerve loss in the absence of other cochlear pathologies is thought to be undetectable with a pure-tone audiogram while degrading real-world speech perception (hidden hearing loss). Perceptual deficits remain unclear, however, due in part to the limited behavioral capacity of existing rodent models to discriminate complex sounds. The budgerigar is an avian vocal learner with human-like behavioral sensitivity to many simple and complex sounds and the capacity to mimic speech. Previous studies in this species show that intracochlear kainic-acid infusion reduces wave 1 of the auditory brainstem response by 40-70%, consistent with substantial excitotoxic auditory-nerve damage. The present study used operant-conditioning procedures in trained budgerigars to quantify kainic-acid effects on tone detection across frequency (0.25-8 kHz; the audiogram) and as a function of duration (20-160 ms; temporal integration). Tone thresholds in control animals were lowest from 1 to 4 kHz and decreased with increasing duration as in previous studies of the budgerigar. Behavioral results in kainic-acid-exposed animals were as sensitive as in controls, suggesting preservation of the audiogram and temporal integration despite auditory-nerve loss associated with up to 70% wave 1 reduction. Distortion-product otoacoustic emissions were also preserved in kainic-acid exposed animals, consistent with normal hair-cell function. These results highlight considerable perceptual resistance of tone-detection performance with selective auditory-nerve loss. Future behavioral studies in budgerigars with auditory-nerve damage can use complex speech-like stimuli to help clarify aspects of auditory perception impacted by this common cochlear pathology.


Asunto(s)
Nervio Coclear/fisiopatología , Melopsittacus/fisiología , Estimulación Acústica , Animales , Audiometría de Tonos Puros , Percepción Auditiva/fisiología , Umbral Auditivo/fisiología , Conducta Animal/fisiología , Nervio Coclear/efectos de los fármacos , Nervio Coclear/lesiones , Condicionamiento Operante/fisiología , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Femenino , Humanos , Ácido Kaínico/toxicidad , Masculino , Emisiones Otoacústicas Espontáneas/fisiología , Ototoxicidad/fisiopatología , Psicoacústica
19.
Hear Res ; 374: 49-57, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30710792

RESUMEN

The blood-perilymph barrier serves a critical role by separating the components of blood from inner ear fluids, limiting traffic of cells, proteins and other solutes into the labyrinth, and allowing gas (O2-CO2) exchange. Inflammation produces changes in the blood-perilymph barrier resulting in increased vascular permeability. It is commonly thought that compromise of the blood-inner ear barrier would lead to hearing impairment through loss of the endocochlear potential (EP). In fact, the effect of increasing cochlear vascular permeability on hearing function and EP is poorly understood. We used a novel method to measure the integrity of the blood-perilymph barrier and demonstrated the effects of barrier compromise on ABR threshold and EP. We also investigated the contribution of CX3CR1 cochlear macrophages and CCR2 inflammatory monocytes to barrier function after systemic exposure to lipopolysaccharide (LPS). We found that systemic LPS induced a profound change in vascular permeability, which correlated with minimal change in ABR threshold and EP. Macrophage depletion using CX3CR1-DTR mice did not alter the baseline permeability of cochlear vessels and resulted in preservation of barrier function in LPS-treated animals. We conclude that cochlear macrophages are not required to maintain the barrier in normal mice and activated macrophages are a critical factor in breakdown of the barrier after LPS. CCR2 null mice demonstrated that LPS induction of barrier leakiness occurs in the absence of CCR2 expression. Thus, enhanced aminoglycoside ototoxicity after LPS can be linked to the expression of CCR2 in inflammatory monocytes, and not to preservation of the blood-perilymph barrier in CCR2 knockout mice.


Asunto(s)
Cóclea/irrigación sanguínea , Cóclea/fisiología , Macrófagos/fisiología , Monocitos/fisiología , Perilinfa/citología , Perilinfa/fisiología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/fisiología , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Cóclea/citología , Toxina Diftérica/toxicidad , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/fisiología , Lipopolisacáridos/toxicidad , Activación de Macrófagos/fisiología , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Ratones Transgénicos , Ototoxicidad/patología , Ototoxicidad/fisiopatología , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/fisiología
20.
Hear Res ; 374: 5-12, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30682699

RESUMEN

In the context of acquired sensorineural hearing loss (SNHL), cochlear hair cells have long been thought to be among the most vulnerable elements in mammalian cochleae. However, recent studies have indicated that the synaptic connection between inner hair cells (IHC) and spiral ganglion neurons (SGN) can be an important target for the treatment of SNHL. Our previous studies in patients with sudden SNHL demonstrated delayed and gradual hearing recovery following topical application of insulin-like growth factor 1 (IGF-1), suggesting that not only protective but also regenerative mechanisms may account for hearing recovery after treatment with IGF-1. We then hypothesized that IGF-1 has the potential to drive the regeneration of IHC-SGN synapses. To test this hypothesis, we investigated the effects of IGF-1 on IHC-SGN synapses using cochlear explant cultures from postnatal day 2 mice that had been damaged by exposure to the excitatory amino acids N-methyl-d-aspartate and kainate. Cochlear explants that lost IHC-SGN synapses upon exposure to excitatory amino acids were cultured with exogenous IGF-1 for an additional 48 h. We observed increased numbers of IHC-SGN synapses after exogenous IGF-1 application. Pharmacological inhibition of the IGF-1 receptor attenuated the restoration of IHC-SGN synapses by exogenous IGF-1. These findings indicated that IGF-1 induces regeneration of IHC-SGN synapses in cochlear explant cultures from postnatal day 2 mice. Therefore, in a future study we will perform in vivo experiments using adult mice to ascertain the effects of IGF-1 on the regeneration of IHC-SGN synapses.


Asunto(s)
Cóclea/efectos de los fármacos , Cóclea/inervación , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Regeneración Nerviosa/efectos de los fármacos , Animales , Cóclea/fisiología , Modelos Animales de Enfermedad , Células Ciliadas Auditivas Internas/efectos de los fármacos , Células Ciliadas Auditivas Internas/patología , Células Ciliadas Auditivas Internas/fisiología , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Pérdida Auditiva Sensorineural/patología , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Técnicas In Vitro , Factor I del Crecimiento Similar a la Insulina/fisiología , Ácido Kaínico/toxicidad , Ratones , Ratones Endogámicos ICR , N-Metilaspartato/toxicidad , Regeneración Nerviosa/fisiología , Ototoxicidad/tratamiento farmacológico , Ototoxicidad/patología , Ototoxicidad/fisiopatología , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/fisiología , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/patología , Ganglio Espiral de la Cóclea/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/patología , Sinapsis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA