Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Sci Rep ; 14(1): 22382, 2024 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333235

RESUMEN

Cisplatin is a commonly used chemotherapy agent with a nearly universal side effect of sensorineural hearing loss. The cellular mechanisms underlying cisplatin ototoxicity are poorly understood. Efforts in drug development to prevent or reverse cisplatin ototoxicity have largely focused on pathways of oxidative stress and apoptosis. An effective treatment for cisplatin ototoxicity, sodium thiosulfate (STS), while beneficial when used in standard risk hepatoblastoma, is associated with reduced survival in disseminated pediatric malignancy, highlighting the need for more specific drugs without potential tumor protective effects. The unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways have been shown to be involved in the pathogenesis of noise-induced hearing loss and cochlear synaptopathy in vivo, and these pathways have been implicated broadly in cisplatin cytotoxicity. This study sought to determine whether the UPR can be targeted to prevent cisplatin ototoxicity. Neonatal cochlear cultures and HEK cells were exposed to cisplatin, and UPR marker gene expression and cell death measured. Treatment with ISRIB (Integrated Stress Response InhIBitor), a drug that activates eif2B and downregulates the pro-apoptotic PERK/CHOP pathway of the UPR, was tested for its ability to reduce apoptosis in HEK cells, hair-cell death in cochlear cultures, and hearing loss using an in vivo mouse model of cisplatin ototoxicity. Finally, to evaluate whether ISRIB might interfere with cisplatin chemoeffectiveness, we tested it in head and neck squamous cell carcinoma (HNSCC) cell-based assays of cisplatin cytotoxicity. Cisplatin exhibited a biphasic, non-linear dose-response of cell death and apoptosis that correlated with different patterns of UPR marker gene expression in HEK cells and cochlear cultures. ISRIB treatment protected against cisplatin-induced hearing loss and hair-cell death, but did not impact cisplatin's cytotoxic effects on HNSCC cell viability, unlike STS. These findings demonstrate that targeting the pro-apoptotic PERK/CHOP pathway with ISRIB can mitigate cisplatin ototoxicity without reducing anti-cancer cell effects, suggesting that this may be a viable strategy for drug development.


Asunto(s)
Cisplatino , Estrés del Retículo Endoplásmico , Ototoxicidad , Respuesta de Proteína Desplegada , Cisplatino/efectos adversos , Cisplatino/toxicidad , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Ototoxicidad/prevención & control , Ototoxicidad/metabolismo , Ototoxicidad/etiología , Humanos , Ratones , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HEK293 , Antineoplásicos/efectos adversos , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Cóclea/patología , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , eIF-2 Quinasa/metabolismo
2.
J Mol Med (Berl) ; 102(9): 1163-1174, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39110182

RESUMEN

Cisplatin is a chemotherapeutic agent widely used to treat solid tumors. However, it can also be highly ototoxic, resulting in high-frequency hearing loss. Cisplatin causes degeneration of hair cells (HCs) and spiral ganglion neurons (SGNs) in the inner ear, which are essential components of the hearing process and cannot be regenerated in mammals. As the affected cells primarily die by apoptosis, we tested several anti-apoptotic small molecules to protect these cells from drug-induced toxicity. We found that the general caspase inhibitor Emricasan could significantly counteract the toxic effects of cisplatin in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells, phoenix auditory cells, and primary SGNs. Importantly, the anti-cytotoxic effect in neuronal cells was even more pronounced than the effect of sodium thiosulfate (STS), which is currently the only approved prevention option for cisplatin-induced ototoxicity. Finally, we tested the protective effect of Emricasan treatment in the context of another ototoxic drug, i.e., the aminoglycoside antibiotic neomycin, and again found a significant increase in cell viability when the cultures were co-treated with Emricasan. These results suggest a promising strategy to prevent ototoxicity in patients by temporarily blocking the apoptotic pathway when applying cisplatin or aminoglycoside antibiotics. KEY MESSAGES: Anti-apoptotic small molecules can reduce cisplatin-induced toxicity. Emricasan can effectively exert its anti-apoptotic effect on cochlear cells. Strong protection from cisplatin- and neomycin-induced cytotoxicity with Emricasan. Sodium thiosulfate and Emricasan provide similar protective effects to cisplatin-treated cells. Emricasan is more potent than sodium thiosulfate in reducing neomycin-induced cytotoxicity.


Asunto(s)
Inhibidores de Caspasas , Cisplatino , Neomicina , Cisplatino/efectos adversos , Cisplatino/toxicidad , Cisplatino/farmacología , Animales , Neomicina/farmacología , Neomicina/toxicidad , Inhibidores de Caspasas/farmacología , Ratones , Apoptosis/efectos de los fármacos , Cóclea/efectos de los fármacos , Cóclea/citología , Supervivencia Celular/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ototoxicidad/etiología , Ototoxicidad/prevención & control , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Línea Celular , Células Cultivadas
3.
Biomed Pharmacother ; 178: 117174, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39098177

RESUMEN

Mangiferin(MGF) exhibits crucial biological roles, including antioxidant and anti-inflammatory functions. However, how to clearly elucidate the functioning mechanism of MGF for inhibiting cisplatin-induced hearing loss requires in-depth investigation. In this work, we aimed at gaining insight into how MGF functions as the protective agent against cisplatin-triggered ototoxicity using various assays. The variation for reactive oxygen species (ROS) concentrations was determined with MitoSOX-Red and 2',7'-Dichlorodihydrofluorescein diacetate staining (DCFH-DA). The protective function and corresponding mechanism of MGF in hair cell survival in the House Ear Institute-Organ of Corti (HEI-OC1) cell line were assessed using RNA sequencing (RNA-Seq). Our findings demonstrated that MGF significantly alleviated cisplatin-induced injury to hair cells in vitro, encompassing cell lines and cochlear explants, as well as in vivo models, including C57BL/6 J mice and zebrafish larvae. Mechanistic studies revealed that MGF reversed the increased accumulation of ROS and inhibited cell apoptosis through mitochondrial-mediated intrinsic pathway. Moreover, real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting data indicated MGF protected against cisplatin-mediated ototoxicity via the mitogen-activated protein kinase pathway (MAPK). These findings demonstrated MGF has significant potential promise in combating cisplatin-induced ototoxicity, offering a foundation for expanded investigation into therapeutic approaches for auditory protection.


Asunto(s)
Apoptosis , Cisplatino , Células Ciliadas Auditivas , Pérdida Auditiva Sensorineural , Ratones Endogámicos C57BL , Ototoxicidad , Especies Reactivas de Oxígeno , Xantonas , Pez Cebra , Cisplatino/toxicidad , Cisplatino/efectos adversos , Animales , Ototoxicidad/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Ratones , Pérdida Auditiva Sensorineural/inducido químicamente , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/prevención & control , Pérdida Auditiva Sensorineural/patología , Apoptosis/efectos de los fármacos , Xantonas/farmacología , Xantonas/uso terapéutico , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Antioxidantes/farmacología
4.
J Int Adv Otol ; 20(3): 189-195, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-39158163

RESUMEN

Different organs respond differently to cisplatin (CDDP)-induced toxicity. Oleuropein (OLE) is a natural phenolic antioxidant. The purpose of this study was to determine the potential protective effect of OLE against CDDP-induced ototoxicity by evaluating expression of genes associated with deoxyribonucleic acid (DNA) damage and repair in cochlear cells. House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were treated using CDDP, OLE, and OLE-CDDP. The water-soluble tetrazolium salt assay was used for monitoring cell viability. Deoxyribonucleic acid damage in cells due to the CDDP, OLE, and combination treatments was determined using a flow-cytometric kit. The change in the expression of 84 genes associated with CCDP, OLE, and OLE-CDDP treatments that induced DNA damage was tested using the reverse transcription polymerase chain reaction array. Changes ≥3-fold were considered significant. House Ear Institute-Organ of Corti 1 cell viability was significantly reduced by CDDP. The OLE-CDDP combination restored the cell viability. Cisplatin increased the H2AX ratio, while OLE-CDDP combination decreased it. Some of the DNA damage-associated genes whose expression was upregulated with CDDP were downregulated with OLE-CDDP, while the expression of genes such as Gadd45g and Rev1 was further downregulated. The expression of DNA repair-related Abl1, Dbd2, Rad52, and Trp53 genes was downregulated with CDDP, whereas their expression was upregulated with OLE-CDDP treatment. In cochlear cells, the OLE-CDDP combination downregulated DNA damage-associated gene expression relative to that upregulated mainly by CDDP. The results revealed that OLE has a potential protective effect on CDDP-induced ototoxicity in cochlear cells by altering the expression of DNA damage-related genes.


Asunto(s)
Supervivencia Celular , Cisplatino , Cóclea , Daño del ADN , Glucósidos Iridoides , Ototoxicidad , Cisplatino/toxicidad , Glucósidos Iridoides/farmacología , Daño del ADN/efectos de los fármacos , Animales , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Cóclea/patología , Supervivencia Celular/efectos de los fármacos , Ototoxicidad/prevención & control , Ratones , Iridoides/farmacología , Antineoplásicos/toxicidad , Antioxidantes/farmacología , Humanos , Línea Celular , Expresión Génica/efectos de los fármacos
5.
Otol Neurotol ; 45(9): 998-1005, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39186064

RESUMEN

HYPOTHESIS: Memantine, an N -methyl- d -aspartate receptor antagonist, is widely used to treat Alzheimer's disease and has been found to have potential neuroprotective effects. In this study, we evaluated the protective effects of memantine against cisplatin-induced ototoxicity. BACKGROUND: Cisplatin is a widely used anticancer drug for various cancers; however, its use is limited by its side effects, including ototoxicity. Several drugs have been developed to reduce cisplatin toxicity. In this study, we treated cisplatin-damaged cochlear hair cells with memantine and evaluated its protective effects. METHOD: House Ear Institute Organ of Corti 1 (HEI-OC1) cells and cochlear explants were treated with cisplatin or memantine. Cell viability, apoptotic patterns, reactive oxygen species (ROS) production, Bcl-2/caspase-3 activity, and cell numbers were measured to evaluate the anti-apoptotic and antioxidative effects of memantine. RESULT: Memantine treatment significantly improved cell viability and reduced cisplatin-induced apoptosis in auditory cells. Bcl-2/caspase-3 activity was also significantly increased, suggesting anti-apoptotic effects against cisplatin-induced ototoxicity. CONCLUSION: Our results suggest that memantine protects against cisplatin-induced ototoxicity in vitro, providing a potential new strategy for preventing hearing loss in patients undergoing cisplatin chemotherapy.


Asunto(s)
Antineoplásicos , Apoptosis , Supervivencia Celular , Cisplatino , Memantina , Ototoxicidad , Especies Reactivas de Oxígeno , Memantina/farmacología , Cisplatino/toxicidad , Cisplatino/efectos adversos , Animales , Ototoxicidad/prevención & control , Apoptosis/efectos de los fármacos , Antineoplásicos/toxicidad , Antineoplásicos/efectos adversos , Supervivencia Celular/efectos de los fármacos , Ratones , Especies Reactivas de Oxígeno/metabolismo , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/patología , Caspasa 3/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Fármacos Neuroprotectores/farmacología , Órgano Espiral/efectos de los fármacos , Órgano Espiral/patología , Cóclea/efectos de los fármacos , Cóclea/patología , Antagonistas de Aminoácidos Excitadores/farmacología , Línea Celular
6.
Braz J Biol ; 84: e279117, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39140499

RESUMEN

Cisplatin is an antineoplastic medicine used in the treatment for various types of cancer. Among its side effects is ototoxicity, which may result in a bilateral and irreversible hearing loss. The ototoxic effect in the pediatric population has a bigger impact as it compromises language acquisition. The discovery of drugs with otoprotective effects and the optimal way to administer them have become significant challenges in minimizing the impact of cisplatin regarding auditory function. The objective was to understand otoprotective drugs and their relevance in the preventive treatment to cisplatin-induced ototoxicity in childhood. An integrative review was conducted by consulting databases including PubMed, Bireme, MedLine, LILACS, SciELO, and ClinicalTrials.gov. The search strategy was performed by crossing descriptors (DeCS and MeSH) and free terms. Studies published in English, Spanish, and Portuguese were selected, with no publication year restrictions. Subsequently, articles were selected according to inclusion and exclusion criteria. A total of 736 articles were found in PubMed, 431 in Bireme, 425 in MedLine, 6 in LILACS, 0 in SciELO, and 4 in ClinicalTrials.gov. After document analysis, 12 articles were selected for full analysis. Evidence was found for 8 substances with potential otoprotective effects when used with cisplatin, which tend to minimize the impact of cisplatin regarding auditory function. The substances found were: Amifostine, Dexamethasone, Genistein, Ginkgo Biloba, Lycopene, N-acetylcysteine, Polydatin also Sodium Thiosulfate. In general, these drugs are applied before, during, or after cisplatin infusion, depending on the chosen drug, via intravenous, oral, or transtympanic injections, acting as antioxidant therapy. The biochemical effects of these substances are relevant to their potential otoprotective properties, including the inactivation of oxygen free radicals and electrophilic platinum species. The use of these substances can reduce ototoxicity, decreasing cisplatin-induced hearing loss and improving the confort of life, especially for children.


Asunto(s)
Antineoplásicos , Cisplatino , Ototoxicidad , Cisplatino/efectos adversos , Humanos , Ototoxicidad/prevención & control , Ototoxicidad/etiología , Niño , Antineoplásicos/efectos adversos , Sustancias Protectoras , Pérdida Auditiva/prevención & control , Pérdida Auditiva/inducido químicamente
7.
Immunopharmacol Immunotoxicol ; 46(5): 635-650, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39138615

RESUMEN

OBJECTIVE: The threat of hearing loss has become a universal reality. Gentamycin (GM) can lead to ototoxicity and may result in permanent hearing loss. This study aimed to elucidate whether the hypolipidemic drug Ezetimibe (EZE) has a possible underlying mechanism for protecting rats from GM-induced ototoxicity. METHODS AND RESULTS: 30 male Wister albino rats were separated into three groups, ten in each group: control, GM, and GM + EZE. At the end of the experiment, rats underwent hearing threshold evaluation via auditory brainstem response (ABR), carotid artery blood flow velocity (CBV), and resistance (CVR) measurement, in addition to a biochemical assessment of serum malondialdehyde (MDA), nitric oxide (NO), catalase (CAT), hemeOxygenase-1 (HO-1), and tumor necrosis factor-α (TNF-α). Also, real-time PCR was employed to quantify the levels of brain-derived neurotrophic factor (BDNF). Cochlea was also studied via histological and immunohistochemical methods. GM revealed a significant increase in CVR, MDA, NO, and TNF-α and a significant decrease in ABR, CBV, CAT, HO-1, and cochlear BDNF expression. EZE supplementation revealed a significant rise in ARB in addition to CBV and a decline in CVR and protected cochlear tissues via antioxidant, anti-inflammatory, and antiapoptotic mechanisms via downregulating Caspase-3 immunoreaction, upregulating proliferating cellular nuclear antigen (PCNA) immunoreaction, and upregulating of the cochlear BDNF expression. Correlations were significantly negative between BDNF and MDA, NO, TNF-α, COX 2, and caspase-3 immunoreaction and significantly positive with CAT, HO-1, and PCNA immunoreaction. DISCUSSION: EZE can safeguard inner ear tissues from GM via antioxidant, anti-inflammatory, and antiapoptotic mechanisms, as well as upregulation of BDNF mechanisms.


Asunto(s)
Antiinflamatorios , Antioxidantes , Factor Neurotrófico Derivado del Encéfalo , Ezetimiba , Gentamicinas , Ratas Wistar , Regulación hacia Arriba , Animales , Masculino , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Antioxidantes/farmacología , Ratas , Regulación hacia Arriba/efectos de los fármacos , Gentamicinas/toxicidad , Gentamicinas/efectos adversos , Antiinflamatorios/farmacología , Ezetimiba/farmacología , Ototoxicidad/prevención & control , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos
8.
Sci Adv ; 10(30): eadk9878, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39047106

RESUMEN

Cisplatin is a widely used anticancer drug with notable side effects including ototoxicity and nephrotoxicity. Macrophages, the major resident immune cells in the cochlea and kidney, are important drivers of both inflammatory and tissue repair responses. To investigate the roles of macrophages in cisplatin-induced toxicities, we used PLX3397, a U.S. Food and Drug Administration-approved inhibitor of the colony-stimulating factor 1 receptor, to eliminate tissue-resident macrophages. Mice treated with cisplatin alone had considerable hearing loss (ototoxicity) and kidney injury (nephrotoxicity). Macrophage ablation resulted in significantly reduced hearing loss and had greater outer hair cell survival. Macrophage ablation also protected against cisplatin-induced nephrotoxicity, as evidenced by markedly reduced tubular injury and fibrosis. Mechanistically, our data suggest that the protective effect of macrophage ablation against cisplatin-induced ototoxicity and nephrotoxicity is mediated by reduced platinum accumulation in both the inner ear and the kidney. Together, our data indicate that ablation of tissue-resident macrophages represents an important strategy for mitigating cisplatin-induced ototoxicity and nephrotoxicity.


Asunto(s)
Cisplatino , Macrófagos , Ototoxicidad , Cisplatino/efectos adversos , Cisplatino/toxicidad , Animales , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ototoxicidad/etiología , Ototoxicidad/prevención & control , Ratones , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/prevención & control , Antineoplásicos/efectos adversos , Antineoplásicos/toxicidad , Enfermedades Renales/inducido químicamente , Enfermedades Renales/prevención & control , Enfermedades Renales/patología , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Cóclea/patología , Ratones Endogámicos C57BL , Aminopiridinas , Pirroles
9.
Food Chem Toxicol ; 190: 114792, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38849049

RESUMEN

Cisplatin is an effective chemotherapy agent against various solid malignancies; however, it is associated with irreversible bilateral sensorineural hearing loss, emphasizing the need for drug development to prevent this complication, with the current options being very limited. Rho-associated coiled-coil-containing protein kinase (ROCK) is a serine-threonine protein kinase involved in various cellular processes, including apoptosis regulation. In this study, we used a transgenic zebrafish model (Brn3C: EGFP) in which hair cells within neuromasts are observed in green under fluorescent microscopy without the need for staining. Zebrafish larvae were exposed to cisplatin alone or in combination with various concentrations of Y-27632, a potent ROCK inhibitor. Hair cell counts, apoptosis assessments using the terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling assay, FM1-43FX labeling assay and behavioral analyses (startle response and rheotaxis) were performed to evaluate the protective effects of Y-27632 against cisplatin-induced ototoxicity. Cisplatin treatment reduced the number of hair cells in neuromasts, induced apoptosis, and impaired zebrafish larval behaviors. Y-27632 demonstrated a dose-dependent protective effect against cisplatin-induced hair cell loss and apoptosis. These findings suggest that Y-27632, as a ROCK inhibitor, mitigates cisplatin-induced hair cell loss and associated ototoxicity in zebrafish.


Asunto(s)
Amidas , Apoptosis , Cisplatino , Ototoxicidad , Piridinas , Pez Cebra , Animales , Cisplatino/toxicidad , Amidas/farmacología , Piridinas/farmacología , Ototoxicidad/prevención & control , Apoptosis/efectos de los fármacos , Animales Modificados Genéticamente , Antineoplásicos/toxicidad , Células Ciliadas Auditivas/efectos de los fármacos , Larva/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Modelos Animales de Enfermedad
10.
Biomed Pharmacother ; 177: 117025, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38941893

RESUMEN

As a broad-spectrum anticancer drug, cisplatin is widely used in the treatment of tumors in various systems. Unfortunately, several serious side effects of cisplatin limit its clinical application, the most common of which are nephrotoxicity and ototoxicity. Studies have shown that cochlear hair cell degeneration is the main cause of cisplatin-induced hearing loss. However, the mechanism of cisplatin-induced hair cell death remains unclear. The present study aimed to explore the potential role of activating transcription factor 6 (ATF6), an endoplasmic reticulum (ER)-localized protein, on cisplatin-induced ototoxicity in vivo and in vitro. In this study, we observed that cisplatin exposure induced apoptosis of mouse auditory OC-1 cells, accompanied by a significant increase in the expression of ATF6 and C/EBP homologous protein (CHOP). In cell or cochlear culture models, treatment with an ATF6 agonist, an ER homeostasis regulator, significantly ameliorated cisplatin-induced cytotoxicity. Further, our in vivo experiments showed that subcutaneous injection of an ATF6 agonist almost completely prevented outer hair cell loss and significantly alleviated cisplatin-induced auditory brainstem response (ABR) threshold elevation in mice. Collectively, our results revealed the underlying mechanism by which activation of ATF6 significantly improved cisplatin-induced hair cell apoptosis, at least in part by inhibiting apoptosis signal-regulating kinase 1 expression, and demonstrated that pharmacological activation of ATF6-mediated unfolded protein response is a potential treatment for cisplatin-induced ototoxicity.


Asunto(s)
Factor de Transcripción Activador 6 , Apoptosis , Cisplatino , Ototoxicidad , Respuesta de Proteína Desplegada , Cisplatino/toxicidad , Animales , Factor de Transcripción Activador 6/metabolismo , Ototoxicidad/prevención & control , Ototoxicidad/etiología , Ototoxicidad/patología , Ratones , Respuesta de Proteína Desplegada/efectos de los fármacos , Apoptosis/efectos de los fármacos , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Línea Celular , Masculino , Antineoplásicos/toxicidad , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Pérdida Auditiva/prevención & control , Ratones Endogámicos C57BL , Factor de Transcripción CHOP/metabolismo
11.
Int Immunopharmacol ; 138: 112464, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38917526

RESUMEN

BACKGROUND: Our previous research demonstrated that resveratrol counters DDP-induced ototoxicity by upregulating miR-455-5p, which targets PTEN. This study aimed to elucidate the underlying mechanisms involving GAS5 and DNA methyltransferase 1 (DNMT1) in resveratrol's protective action. METHODS: A luciferase reporter assay and RNA immunoprecipitation (RIP) assay were employed to study the binding between GAS5 and miR-455-5p, as well as between miR-455-5p and PTEN. HEI-OC1 cells treated with DDP were transfected with vectors for GAS5, si-GAS5, DNMT1, si-DNMT1, and miR-455-5p mimics, as well as PTEN. Subsequently, they were treated with resveratrol and exposed to DDP, both separately and in combination. The distribution of CpG islands in the GAS5 promoter was identified using MethyPrimer, and methylation-specific PCR (MSP) was conducted to determine the methylation levels of GAS5. Chromatin immunoprecipitation (ChIP) was utilized to examine the interaction between DNMT1 and GAS5. The viability of HEI-OC1 cells, catalase (CAT) activity, apoptosis, and ROS levels were assessed using the CCK-8 assay, CAT assay, TUNEL staining, and flow cytometry, respectively. An in vivo mouse model was developed to measure auditory brainstem response (ABR) thresholds, while RT-qPCR and Western blot analysis were employed to evaluate molecular levels. RESULTS: Our study discovered that GAS5 acts as a sponge for miR-455-5p, thereby increasing PTEN expression in DDP-treated HEI-OC1 cells. This process was reversed upon treatment with resveratrol. Importantly, DNMT1 promoted the methylation of the GAS5 promoter, leading to the suppression of GAS5 expression. This suppression enhanced the effectiveness of resveratrol in combating DDP-induced apoptosis and ROS in HEI-OC1 cells and amplified its protective effect against DDP's ototoxicity in vivo. CONCLUSIONS: Our research emphasizes the significance of the DNMT1/GAS5/miR-455-5p/PTEN axis as a promising new route to boost resveratrol's effectiveness against DDP-induced ototoxicity.


Asunto(s)
Cisplatino , ADN (Citosina-5-)-Metiltransferasa 1 , Epigénesis Genética , MicroARNs , Ototoxicidad , Fosfohidrolasa PTEN , ARN Largo no Codificante , Resveratrol , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , MicroARNs/genética , MicroARNs/metabolismo , Animales , Resveratrol/farmacología , Resveratrol/uso terapéutico , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ototoxicidad/prevención & control , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , Epigénesis Genética/efectos de los fármacos , Línea Celular , Masculino , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos
12.
Toxicol In Vitro ; 99: 105852, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38789064

RESUMEN

Cisplatin is an effective chemotherapeutic agent; however, ototoxicity is one of its negative effects that greatly limits the use of cisplatin in clinical settings. Previous research has shown that the most important process cisplatin damage to inner ear cells, such as hair cells (HCs), is the excessive production and accumulation of ROS. Schisandrin B (SchB), is a low-toxicity, inexpensive, naturally occurring antioxidant with a variety of pharmacological effects. Therefore, the potential antioxidant effects of SchB may be useful for cisplatin ototoxicity treatment. In this study, the effects of SchB on cochlear hair cell viability, ROS levels, and expression of apoptosis-related molecules were evaluated by CCK-8, immunofluorescence, flow cytometry, and qRT-PCR, as well as auditory brainstem response (ABR) and dysmorphic product otoacoustic emission (DPOAE) tests to assess the effects on inner ear function. The results showed that SchB treatment increased cell survival, prevented apoptosis, and reduced cisplatin-induced ROS formation. SchB treatment reduced the loss of cochlear HCs caused by cisplatin in exosome culture. In addition, SchB treatment attenuated cisplatin-induced hearing loss and HC loss in mice. This study demonstrates the ability of SchB to inhibit cochlear hair cell apoptosis and ROS generation and shows its potential therapeutic effect on cisplatin ototoxicity.


Asunto(s)
Antineoplásicos , Apoptosis , Supervivencia Celular , Cisplatino , Ciclooctanos , Células Ciliadas Auditivas Internas , Lignanos , Estrés Oxidativo , Compuestos Policíclicos , Especies Reactivas de Oxígeno , Cisplatino/toxicidad , Ciclooctanos/farmacología , Compuestos Policíclicos/farmacología , Compuestos Policíclicos/toxicidad , Animales , Apoptosis/efectos de los fármacos , Lignanos/farmacología , Estrés Oxidativo/efectos de los fármacos , Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Células Ciliadas Auditivas Internas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/farmacología , Antioxidantes/farmacología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Masculino , Ototoxicidad/prevención & control
13.
Hear Res ; 447: 109013, 2024 06.
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
14.
Arch Toxicol ; 98(6): 1827-1842, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38563869

RESUMEN

Aminoglycosides are commonly used antibiotics for treatment of gram-negative bacterial infections, however, they might act on inner ear, leading to hair-cell death and hearing loss. Currently, there is no targeted therapy for aminoglycoside ototoxicity, since the underlying mechanisms of aminoglycoside-induced hearing impairments are not fully defined. This study aimed to investigate whether the calcium channel blocker verapamil and changes in intracellular & extracellular calcium could ameliorate aminoglycoside-induced ototoxicity in zebrafish. The present findings showed that a significant decreased number of neuromasts in the lateral lines of zebrafish larvae at 5 days' post fertilization after neomycin (20 µM) and gentamicin (20 mg/mL) exposure, which was prevented by verapamil. Moreover, verapamil (10-100 µM) attenuated aminoglycoside-induced toxic response in different external calcium concentrations (33-3300 µM). The increasing extracellular calcium reduced hair cell loss from aminoglycoside exposure, while lower calcium facilitated hair cell death. In contrast, calcium channel activator Bay K8644 (20 µM) enhanced aminoglycoside-induced ototoxicity and reversed the protective action of higher external calcium on hair cell loss. However, neomycin-elicited hair cell death was not altered by caffeine, ryanodine receptor (RyR) agonist, and RyR antagonists, including thapsigargin, ryanodine, and ruthenium red. The uptake of neomycin into hair cells was attenuated by verapamil and under high external calcium concentration. Consistently, the production of reactive oxygen species (ROS) in neuromasts exposed to neomycin was also reduced by verapamil and high external calcium. Significantly, zebrafish larvae when exposed to neomycin exhibited decreased swimming distances in reaction to droplet stimulus when compared to the control group. Verapamil and elevated external calcium effectively protected the impaired swimming ability of zebrafish larvae induced by neomycin. These data imply that prevention of hair cell damage correlated with swimming behavior against aminoglycoside ototoxicity by verapamil and higher external calcium might be associated with inhibition of excessive ROS production and aminoglycoside uptake through cation channels. These findings indicate that calcium channel blocker and higher external calcium could be applied to protect aminoglycoside-induced listening impairments.


Asunto(s)
Antibacterianos , Bloqueadores de los Canales de Calcio , Calcio , Gentamicinas , Células Ciliadas Auditivas , Neomicina , Verapamilo , Pez Cebra , Animales , Bloqueadores de los Canales de Calcio/farmacología , Calcio/metabolismo , Verapamilo/farmacología , Neomicina/toxicidad , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Gentamicinas/toxicidad , Antibacterianos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Ototoxicidad/prevención & control , Aminoglicósidos/toxicidad , Sistema de la Línea Lateral/efectos de los fármacos , Larva/efectos de los fármacos , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/prevención & control
15.
J Assoc Res Otolaryngol ; 25(3): 259-275, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38622383

RESUMEN

PURPOSE: Cisplatin is a low-cost clinical anti-tumor drug widely used to treat solid tumors. However, its use could damage cochlear hair cells, leading to irreversible hearing loss. Currently, there appears one drug approved in clinic only used for reducing ototoxicity associated with cisplatin in pediatric patients, which needs to further explore other candidate drugs. METHODS: Here, by screening 1967 FDA-approved drugs to protect cochlear hair cell line (HEI-OC1) from cisplatin damage, we found that Tedizolid Phosphate (Ted), a drug indicated for the treatment of acute infections, had the best protective effect. Further, we evaluated the protective effect of Ted against ototoxicity in mouse cochlear explants, zebrafish, and adult mice. The mechanism of action of Ted was further explored using RNA sequencing analysis and verified. Meanwhile, we also observed the effect of Ted on the anti-tumor effect of cisplatin. RESULTS: Ted had a strong protective effect on hair cell (HC) loss induced by cisplatin in zebrafish and mouse cochlear explants. In addition, when administered systemically, it protected mice from cisplatin-induced hearing loss. Moreover, antitumor studies showed that Ted had no effect on the antitumor activity of cisplatin both in vitro and in vivo. RNA sequencing analysis showed that the otoprotective effect of Ted was mainly achieved by inhibiting phosphorylation of ERK. Consistently, ERK activator aggravated the damage of cisplatin to HCs. CONCLUSION: Collectively, these results showed that FDA-approved Ted protected HCs from cisplatin-induced HC loss by inhibiting ERK phosphorylation, indicating its potential as a candidate for preventing cisplatin ototoxicity in clinical settings.


Asunto(s)
Antineoplásicos , Cisplatino , Pérdida Auditiva , Organofosfatos , Oxazoles , Pez Cebra , Animales , Cisplatino/toxicidad , Cisplatino/efectos adversos , Ratones , Pérdida Auditiva/prevención & control , Pérdida Auditiva/inducido químicamente , Oxazoles/farmacología , Organofosfatos/toxicidad , Antineoplásicos/toxicidad , United States Food and Drug Administration , Aprobación de Drogas , Células Ciliadas Auditivas/efectos de los fármacos , Estados Unidos , Ototoxicidad/prevención & control , Ototoxicidad/etiología , Humanos
16.
Toxicol Appl Pharmacol ; 486: 116947, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38688426

RESUMEN

AIMS: SERCA2, one of the P-type pumps encoded by gene ATP2A2, is the only calcium reflux channel of the endoplasmic reticulum (ER) and participates in maintaining calcium homeostasis. The present study was designed to explore SERCA2 expression pattern in auditory hair cells and the possible mechanism underlying the effects of SERCA2 on cisplatin-induced ototoxicity. MAIN METHODS: The SERCA2 expression pattern in cochlea hair cells and HEI-OC1 cells was measured by Western blot (WB) and immunofluorescence staining. The apoptosis and its related factors were detected by TUNEL assay and WB. The expression levels of ER stress-related factors, ATF6, PERK, IRE1α, and GRP78, were measured via WB. As for the determination of SERCA2 overexpression and knockdown, plasmids and lentiviral vectors were constructed, respectively. KEY FINDINGS: We found that SERCA2 was highly expressed in cochlea hair cells and HEI-OC1 cells. Of note, the level of SERCA2 expression in neonatal mice was remarkably higher than that in adult mice. Under the exposure of 30 µM cisplatin, SERCA2 was down-regulated significantly compared with the control group. In addition, cisplatin administration triggered the occurrence of ER stress and apoptosis. Those events were reversed by overexpressing SERCA2. On the contrary, SERCA2 knockdown could aggravate the above processes. SIGNIFICANCE: The findings from the present study disclose, for the first time, that SERCA2 is abundantly expressed in cochlea hair cells, and the suppression of SERCA2 caused by cisplatin could trigger ER homeostasis disruption, thereby implying that SERCA2 might be a promising target to prevent cisplatin-induced cytotoxicity of hair cells.


Asunto(s)
Apoptosis , Cisplatino , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Células Ciliadas Auditivas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Cisplatino/toxicidad , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones , Apoptosis/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Línea Celular , Antineoplásicos/toxicidad , Masculino , Ototoxicidad/prevención & control
17.
Biochem Pharmacol ; 223: 116132, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38492782

RESUMEN

Cisplatin is an effective chemotherapeutic drug for different cancers, but it also causes severe and permanent hearing loss. Oxidative stress and mitochondrial dysfunction in cochlear hair cells (HCs) have been shown to be important in the pathogenesis of cisplatin-induced hearing loss (CIHL). CDGSH iron sulfur domain 1 (CISD1, also known as mitoNEET) plays a critical role in mitochondrial oxidative capacity and cellular bioenergetics. Targeting CISD1 may improve mitochondrial function in various diseases. However, the role of CISD1 in cisplatin-induced ototoxicity is unclear. Therefore, this study was performed to assess the role of CISD1 in cisplatin-induced ototoxicity. We found that CISD1 expression was significantly increased after cisplatin treatment in both HEI-OC1 cells and cochlear HCs. Moreover, pharmacological inhibition of CISD1 with NL-1 inhibited cell apoptosis and reduced mitochondrial reactive oxygen species accumulation in HEI-OC1 cells and cochlear explants. Inhibition of CISD1 with small interfering RNA in HEI-OC1 cells had similar protective effects. Furthermore, NL-1 protected against CIHL in adult C57 mice, as evaluated by the auditory brainstem response and immunofluorescent staining. Mechanistically, RNA sequencing revealed that NL-1 attenuated CIHL via the PI3K and MAPK pathways. Most importantly, NL-1 did not interfere with the antitumor efficacy of cisplatin. In conclusion, our study revealed that targeting CISD1 with NL-1 reduced reactive oxygen species accumulation, mitochondrial dysfunction, and apoptosis via the PI3K and MAPK pathways in HEI-OC1 cell lines and mouse cochlear explants in vitro, and it protected against CIHL in adult C57 mice. Our study suggests that CISD1 may serve as a novel target for the prevention of CIHL.


Asunto(s)
Antineoplásicos , Pérdida Auditiva , Enfermedades Mitocondriales , Ototoxicidad , Ratones , Animales , Cisplatino/toxicidad , Cisplatino/metabolismo , Antineoplásicos/toxicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ototoxicidad/prevención & control , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/prevención & control , Apoptosis , Proteínas de la Membrana/metabolismo , Proteínas de Unión a Hierro/farmacología
18.
Clin Otolaryngol ; 49(1): 1-15, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37818931

RESUMEN

OBJECTIVES: Ototoxicity is a common disabling side effect of platinum-based chemotherapy. This study aimed to assess the evidence on the management of platinum-induced ototoxicity in adult cancer patients. METHODS: Four databases were searched up to 1 November 2022. Original studies were included if they reported on a pharmacologic or non-pharmacologic intervention to prevent or treat platinum ototoxicity in adults. The articles' quality was assessed via two grading scales. RESULTS: Nineteen randomised controlled trials and five quasi-experimental studies with 1673 patients were analysed. Eleven interventions were identified, nine pharmacological and two non-pharmacological. Six of the interventions (sodium thiosulphate, corticoids, sertraline, statins, multivitamins and D-methionine) showed mild benefits in preventing cisplatin-induced ototoxicity. Only one trial assessed corticoids as a potential treatment. Overall, only six trials were deemed with a low risk of bias. The majority of studies inadequately documented intervention-related adverse effects, thereby limiting safety conclusions. CONCLUSIONS: Current interventions have mild benefits in preventing cisplatin-induced ototoxicity in adult cancer patients. Sodium thiosulphate is the most promising intervention as a preventive strategy. Rigorous, high-quality research is warranted, encompassing an evaluation of all potential symptoms and innovative treatment modalities.


Asunto(s)
Antineoplásicos , Pérdida Auditiva , Neoplasias , Ototoxicidad , Adulto , Humanos , Cisplatino/uso terapéutico , Antineoplásicos/uso terapéutico , Carboplatino/efectos adversos , Ototoxicidad/etiología , Ototoxicidad/prevención & control , Ototoxicidad/tratamiento farmacológico , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/prevención & control , Pérdida Auditiva/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Neoplasias/inducido químicamente , Corticoesteroides/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
19.
J Appl Toxicol ; 44(2): 235-244, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37650462

RESUMEN

Gentamicin (GM) is one of the commonly used antibiotics in the aminoglycoside class but is ototoxic, which constantly impacts the quality of human life. Pyrroloquinoline quinone (PQQ) as a redox cofactor produced by bacteria was found in soil and foods that exert an antioxidant and redox modulator. It is well documented that the PQQ can alleviate inflammatory responses and cytotoxicity. However, our understanding of PQQ in ototoxicity remains unclear. We reported that PQQ could protect against GM-induced ototoxicity in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells in vitro. To evaluate reactive oxygen species (ROS) production and mitochondrial function, ROS and JC-1 staining, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR) measurements in living cells, mitochondrial dynamics analysis was performed. GM-mediated damage was performed by reducing the production of ROS and inhibiting mitochondria biogenesis and dynamics. PQQ ameliorated the cellular oxidative stress and recovered mitochondrial membrane potential, facilitating the recovery of mitochondrial biogenesis and dynamics. Our in vitro findings improve our understanding of the GM-induced ototoxicity with therapeutic implications for PQQ.


Asunto(s)
Gentamicinas , Ototoxicidad , Humanos , Gentamicinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Cofactor PQQ/farmacología , Cofactor PQQ/uso terapéutico , Cofactor PQQ/metabolismo , Ototoxicidad/etiología , Ototoxicidad/prevención & control , Ototoxicidad/metabolismo , Células Ciliadas Auditivas/metabolismo , Antibacterianos/metabolismo , Apoptosis
20.
J Chin Med Assoc ; 87(1): 17-24, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37962398

RESUMEN

Aminoglycoside- and cisplatin-induced ototoxicity, which is a significant issue owing to the widespread use of these drugs in clinical practice, involves the entry of aminoglycosides and cisplatin into the endolymph and hair cells via specific channels or transporters, followed by reactive oxygen species (ROS) generation and hair cells apoptosis. Current strategies focalize primarily on interference with downstream ROS effects; however, recent evidence has demonstrated that inhibiting the uptake of aminoglycosides and cisplatin by hair cells is another promising strategy for tackling the upstream drug uptake pathway. With advances in structural biology, the conformations of certain aminoglycoside and cisplatin channels and transporters, such as the mechanoelectrical transduction channel and organic cation transporter-2, have been largely elucidated. These channels and transporters may become potential targets for the introduction of new otoprotective strategies. This review focuses on the strategies for inhibiting ototoxic drugs uptake by auditory hair cells and provides potential targets for recent developments in the field of otoprotection. Molecular dynamics (MD) simulations of these proteins could help identify the molecules that inhibit the uptake of aminoglycosides and cisplatin by hair cells. Integrating upstream drug uptake pathway targets and MD simulations may help dissect molecular mechanisms and develop novel otoprotective strategies for aminoglycoside- and cisplatin-induced ototoxicity.


Asunto(s)
Antineoplásicos , Ototoxicidad , Humanos , Cisplatino/toxicidad , Aminoglicósidos/efectos adversos , Antineoplásicos/toxicidad , Ototoxicidad/prevención & control , Especies Reactivas de Oxígeno , Antibacterianos/farmacología , Apoptosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...