Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Hear Res ; 447: 109022, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38705005

RESUMEN

The disruption of ribbon synapses in the cochlea impairs the transmission of auditory signals from the cochlear sensory receptor cells to the auditory cortex. Although cisplatin-induced loss of ribbon synapses is well-documented, and studies have reported nitration of cochlear proteins after cisplatin treatment, yet the underlying mechanism of cochlear synaptopathy is not fully understood. This study tests the hypothesis that cisplatin treatment alters the abundance of cochlear synaptosomal proteins, and selective targeting of nitrative stress prevents the associated synaptic dysfunction. Auditory brainstem responses of mice treated with cisplatin showed a reduction in amplitude and an increase in latency of wave I, indicating cisplatin-induced synaptic dysfunction. The mass spectrometry analysis of cochlear synaptosomal proteins identified 102 proteins that decreased in abundance and 249 that increased in abundance after cisplatin treatment. Pathway analysis suggested that the dysregulated proteins were involved in calcium binding, calcium ion regulation, synapses, and endocytosis pathways. Inhibition of nitrative stress by co-treatment with MnTBAP, a peroxynitrite scavenger, attenuated cisplatin-induced changes in the abundance of 27 proteins. Furthermore, MnTBAP co-treatment prevented the cisplatin-induced decrease in the amplitude and increase in the latency of wave I. Together, these findings suggest a potential role of oxidative/nitrative stress in cisplatin-induced cochlear synaptic dysfunction.


Asunto(s)
Cisplatino , Cóclea , Potenciales Evocados Auditivos del Tronco Encefálico , Proteómica , Sinapsis , Sinaptosomas , Cisplatino/toxicidad , Cisplatino/farmacología , Animales , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Cóclea/patología , Cóclea/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Sinapsis/patología , Sinaptosomas/metabolismo , Sinaptosomas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos CBA , Masculino , Ototoxicidad/metabolismo , Ototoxicidad/fisiopatología , Ratones
2.
Sci Rep ; 14(1): 10910, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740884

RESUMEN

Transforming growth factor-ß (TGF-ß) signaling plays a significant role in multiple biological processes, including inflammation, immunity, and cell death. However, its specific impact on the cochlea remains unclear. In this study, we aimed to investigate the effects of TGF-ß signaling suppression on auditory function and cochlear pathology in mice with kanamycin-induced ototoxicity. Kanamycin and furosemide (KM-FS) were systemically administered to 8-week-old C57/BL6 mice, followed by immediate topical application of a TGF-ß receptor inhibitor (TGF-ßRI) onto the round window membrane. Results showed significant TGF-ß receptor upregulation in spiral ganglion neurons (SGNs) after KM-FA ototoxicity, whereas expression levels in the TGF-ßRI treated group remained unchanged. Interestingly, despite no significant change in cochlear TGF-ß expression after KM-FS ototoxicity, TGF-ßRI treatment resulted in a significant decrease in TGF-ß signaling. Regarding auditory function, TGF-ßRI treatment offered no therapeutic effects on hearing thresholds and hair cell survival following KM-FS ototoxicity. However, SGN loss and macrophage infiltration were significantly increased with TGF-ßRI treatment. These results imply that inhibition of TGF-ß signaling after KM-FS ototoxicity promotes cochlear inflammation and SGN degeneration.


Asunto(s)
Kanamicina , Ratones Endogámicos C57BL , Ototoxicidad , Transducción de Señal , Ganglio Espiral de la Cóclea , Factor de Crecimiento Transformador beta , Animales , Kanamicina/toxicidad , Transducción de Señal/efectos de los fármacos , Ototoxicidad/etiología , Ototoxicidad/metabolismo , Ototoxicidad/patología , Factor de Crecimiento Transformador beta/metabolismo , Ratones , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/patología , Cóclea/metabolismo , Cóclea/efectos de los fármacos , Cóclea/patología , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Furosemida/farmacología , Masculino
3.
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
4.
Redox Rep ; 29(1): 2341470, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38629504

RESUMEN

Cisplatin is widely employed in clinical oncology as an anticancer chemotherapy drug in clinical practice and is known for its severe ototoxic side effects. Prior research indicates that the accumulation of reactive oxygen species (ROS) plays a pivotal role in cisplatin's inner ear toxicity. Hesperidin is a flavanone glycoside extracted from citrus fruits that has anti-inflammatory and antioxidant effects. Nonetheless, the specific pharmacological actions of hesperidin in alleviating cisplatin-induced ototoxicity remain elusive. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a critical mediator of the cellular oxidative stress response, is influenced by hesperidin. Activation of Nrf2 was shown to have a protective effect against cisplatin-induced ototoxicity. The potential of hesperidin to stimulate Nrf2 in attenuating cisplatin's adverse effects on the inner ear warrants further investigation. This study employs both in vivo and in vitro models of cisplatin ototoxicity to explore this possibility. Our results reveal that hesperidin mitigates cisplatin-induced ototoxicity by activating the Nrf2/NQO1 pathway in sensory hair cells, thereby reducing ROS accumulation, preventing hair cell apoptosis, and alleviating hearing loss.


Asunto(s)
Antineoplásicos , Hesperidina , Ototoxicidad , Humanos , Cisplatino/toxicidad , Hesperidina/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Ototoxicidad/tratamiento farmacológico , Ototoxicidad/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Línea Celular , Antineoplásicos/toxicidad , Células Ciliadas Auditivas/metabolismo , Apoptosis
5.
Chem Biol Interact ; 393: 110939, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38490643

RESUMEN

Cisplatin (CDDP) is broadly employed to treat different cancers, whereas there are no drugs approved by the Food and Drug Administration (FDA) for preventing its side effects, including ototoxicity. Quercetin (QU) is a widely available natural flavonoid compound with anti-tumor and antioxidant properties. The research was designed to explore the protective effects of QU on CDDP-induced ototoxicity and its underlying mechanisms in male C57BL/6 J mice and primary cultured pericytes (PCs). Hearing changes, morphological changes of stria vascularis, blood labyrinth barrier (BLB) permeability and expression of apoptotic proteins were observed in vivo by using the auditory brainstem response (ABR) test, HE staining, Evans blue staining, immunohistochemistry, western blotting, etc. Oxidative stress levels, mitochondrial function and endothelial barrier changes were observed in vitro by using DCFH-DA probe detection, flow cytometry, JC-1 probe, immunofluorescence and the establishment in vitro BLB models, etc. QU pretreatment activates the PI3K/AKT signaling pathway, inhibits CDDP-induced oxidative stress, protects mitochondrial function, and reduces mitochondrial apoptosis in PCs. However, PI3K/AKT specific inhibitor (LY294002) partially reverses the protective effects of QU. In addition, in vitro BLB models were established by coculturing PCs and endothelial cells (ECs), which suggests that QU both reduces the CDDP-induced apoptosis in PCs and improves the endothelial barrier permeability. On the whole, the research findings suggest that QU can be used as a novel treatment to reduce CDDP-induced ototoxicity.


Asunto(s)
Cisplatino , Ototoxicidad , Ratones , Animales , Masculino , Cisplatino/farmacología , Cisplatino/metabolismo , Pericitos/metabolismo , Quercetina/farmacología , Quercetina/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células Endoteliales/metabolismo , Ototoxicidad/metabolismo , Ratones Endogámicos C57BL , Estrés Oxidativo , Apoptosis
6.
Mol Ther ; 32(5): 1387-1406, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38414247

RESUMEN

Cisplatin-induced hearing loss is a common side effect of cancer chemotherapy in clinics; however, the mechanism of cisplatin-induced ototoxicity is still not completely clarified. Cisplatin-induced ototoxicity is mainly associated with the production of reactive oxygen species, activation of apoptosis, and accumulation of intracellular lipid peroxidation, which also is involved in ferroptosis induction. In this study, the expression of TfR1, a ferroptosis biomarker, was upregulated in the outer hair cells of cisplatin-treated mice. Moreover, several key ferroptosis regulator genes were altered in cisplatin-damaged cochlear explants based on RNA sequencing, implying the induction of ferroptosis. Ferroptosis-related Gpx4 and Fsp1 knockout mice were established to investigate the specific mechanisms associated with ferroptosis in cochleae. Severe outer hair cell loss and progressive damage of synapses in inner hair cells were observed in Atoh1-Gpx4-/- mice. However, Fsp1-/- mice showed no significant hearing phenotype, demonstrating that Gpx4, but not Fsp1, may play an important role in the functional maintenance of HCs. Moreover, findings showed that FDA-approved luteolin could specifically inhibit ferroptosis and alleviate cisplatin-induced ototoxicity through decreased expression of transferrin and intracellular concentration of ferrous ions. This study indicated that ferroptosis inhibition through the reduction of intracellular ferrous ions might be a potential strategy to prevent cisplatin-induced hearing loss.


Asunto(s)
Cisplatino , Ferroptosis , Pérdida Auditiva , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Animales , Cisplatino/efectos adversos , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Ratones , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Modelos Animales de Enfermedad , Receptores de Transferrina/metabolismo , Receptores de Transferrina/genética , Especies Reactivas de Oxígeno/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Ototoxicidad/etiología , Ototoxicidad/metabolismo , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167024, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38242180

RESUMEN

Oxidative stress is the common mechanism of sensorineural hearing loss (SNHL) caused by many factors, such as noise, drugs and ageing. Here, we used tert-butyl hydroperoxide (t-BHP) to cause oxidative stress damage in HEI-OC1 cells and in an in vitro cochlear explant model. We observed lipid peroxidation, iron accumulation, mitochondrial shrinkage and vanishing of mitochondrial cristae, which caused hair cell ferroptosis, after t-BHP exposure. Moreover, the number of TUNEL-positive cells in cochlear explants and HEI-OC1 cells increased significantly, suggesting that t-BHP caused the apoptosis of hair cells. Administration of deferoxamine (DFOM) significantly attenuated t-BHP-induced hair cell loss and disordered hair cell arrangement in cochlear explants as well as HEI-OC1 cell death, including via apoptosis and ferroptosis. Mechanistically, we found that DFOM treatment reduced t-BHP-induced lipid peroxidation, iron accumulation and mitochondrial pathological changes in hair cells, consequently mitigating apoptosis and ferroptosis. Moreover, DFOM treatment alleviated GSH depletion caused by t-BHP and activated the Nrf2 signalling pathway to exert a protective effect. Furthermore, we confirmed that the protective effect of DFOM mainly depended on its ability to chelate iron by constructing Fth1 knockout (KO), TfR1 KO and Nrf2 KO HEI-OC1 cell lines using CRISPR/Cas9 technology and a Flag-Fth1 (overexpression) HEI-OC1 cell line using the FlpIn™ System. Our findings suggest that DFOM is a potential drug for SNHL treatment due to its ability to inhibit apoptosis and ferroptosis by chelating iron and scavenging reactive oxygen species (ROS).


Asunto(s)
Deferoxamina , Ototoxicidad , Humanos , terc-Butilhidroperóxido/toxicidad , terc-Butilhidroperóxido/metabolismo , Deferoxamina/farmacología , Ototoxicidad/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Células Ciliadas Auditivas/metabolismo , Hierro/metabolismo
8.
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
9.
Otol Neurotol ; 45(1): e49-e56, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38085767

RESUMEN

BACKGROUND: The widespread use of aminoglycosides is a prevalent cause of sensorineural hearing loss. Patients receiving aminoglycosides usually have elevated levels of circulating stress hormones due to disease or physiological stress; however, whether the stress hormone cortisol impacts aminoglycoside-mediated injury of cochlear hair cells has not been fully investigated. METHODS: House Ear Institute-Organ of Corti 1 (HEI-OC1) cells with or without cortisol pretreatment were exposed to gentamicin, we investigated the effect of cortisol pretreatment on gentamicin ototoxicity by assessing cell viability. Molecular pathogenesis was explored by detecting apoptosis and oxidative stress. Meanwhile, by inhibiting glucocorticoid receptors (GR) and mineralocorticoid receptors (MR), the potential roles of receptor types in cortisol-mediated sensitization were evaluated. RESULTS: Cortisol concentrations below 75 µmol/l did not affect cell viability. However, pretreatment with 50 µmol/l cortisol for 24 hours sensitized hair cells to gentamicin-induced apoptosis. Further mechanistic studies revealed that cortisol significantly increased hair cell apoptosis and oxidative stress, and altered apoptosis-related protein expressions induced by gentamicin. In addition, blockade of either GR or MR attenuated cortisol-induced hair cell sensitization to gentamicin toxicity. CONCLUSION: Cortisol pretreatment increased mammalian hair cell susceptibility to gentamicin toxicity. Sensitization was related to the activation of the intrinsic apoptotic pathway and excessive generation of reactive oxygen species. Cortisol may exacerbate aminoglycoside ototoxicity.


Asunto(s)
Antibacterianos , Gentamicinas , Células Ciliadas Auditivas , Hidrocortisona , Ototoxicidad , Animales , Humanos , Aminoglicósidos , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Apoptosis , Gentamicinas/efectos adversos , Gentamicinas/toxicidad , Células Ciliadas Auditivas/efectos de los fármacos , Hidrocortisona/farmacología , Mamíferos/metabolismo , Ototoxicidad/etiología , Ototoxicidad/metabolismo , Inhibidores de la Síntesis de la Proteína , Especies Reactivas de Oxígeno/metabolismo
10.
Arch Biochem Biophys ; 748: 109766, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37813237

RESUMEN

AIMS: FAM134B, the initial endoplasmic reticulum (ER)-phagy receptor identified, facilitates ER-phagy during ER stress. The malfunction of FAM134B has been demonstrated to have a crucial role in the pathological mechanisms of diverse human ailments. However, the role of FAM134B-mediated ER-phagy in ototoxicity, particularly in cisplatin-induced ototoxicity, remains unclear. The present study endeavors to investigate whether FAM134B is expressed in House Ear Institute-Organ of Corti 1 (HEI-OC1) and C57BL/6 murine cochlear hair cells (HCs), and to explore its potential function in cisplatin-mediated ototoxicity, with the aim of discovering new insights that can mitigate or forestall the irreversible adverse effect of cisplatin. METHODS: Immunofluorescence (IF) staining was used to test the expression pattern of FAM134B, levels of C/EBP-homologous protein (CHOP), autophagy, and co-localization ratio of lysosomes and ER. Western blotting was employed to measure changes in expression levels of FAM134B, LC3B, ER stress-related proteins, LAMP1 and apoptotic mediators. Cell apoptosis was examined using transferase dUTP nick end labeling (TUNEL) assay and flow cytometry. RESULTS: In the present investigation, it was observed that FAM134B exhibited a diffuse expression pattern in the cytoplasm and nuclei of control HEI-OC1 cells. Following cisplatin administration, FAM134B was found to accumulate and form distinct dots around the nuclei, concomitant with increased levels of ER-phagy, ER stress, unfolded protein response (UPR), and cell apoptosis. Additionally, knockdown of FAM134B resulted in reduced ER-phagy, mitigated ER stress and UPR, and decreased apoptotic activity in HEI-OC1 cells following cisplatin exposure. CONCLUSIONS: Collectively, the findings of this study demonstrate that FAM134B-mediated ER-phagy enhances the susceptibility of HCs to ER stress and apoptosis in response to cisplatin-induced stress. This suggests a sequential progression of ER-phagy, ER stress and apoptosis following cisplatin stimulus, and implies the potential therapeutic benefit of inhibiting of FAM134B-mediated ER-phagy in the prevention of cisplatin-related ototoxicity.


Asunto(s)
Cisplatino , Ototoxicidad , Ratones , Humanos , Animales , Cisplatino/toxicidad , Ototoxicidad/metabolismo , Estrés del Retículo Endoplásmico , Células Ciliadas Auditivas/metabolismo , Autofagia , Retículo Endoplásmico/metabolismo , Apoptosis
11.
J Cell Sci ; 136(14)2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37334742

RESUMEN

Cisplatin is an effective platinum-based chemotherapeutic with several side effects, including ototoxicity. Cochlear cells have low rates of proliferation yet are highly susceptible to cisplatin. We hypothesised that cisplatin ototoxicity might be caused by cisplatin-protein interactions rather than cisplatin-DNA interactions. Two known cisplatin-binding proteins are involved in the stress granule (SG) response. SGs are a pro-survival mechanism involving formation of transient ribonucleoprotein complexes during stress. We examined the effects of cisplatin on SG dynamics and composition in cell lines derived from the cochlea and retinal pigment epithelium. Cisplatin-induced SGs are significantly diminished in size and quantity compared to arsenite-induced SGs and are persistent after 24 h recovery. Additionally, cisplatin pre-treated cells were unable to form a typical SG response to subsequent arsenite stress. Cisplatin-induced SGs had significant reductions in the sequestration of eIF4G and the proteins RACK1 and DDX3X. Live-cell imaging of Texas Red-conjugated cisplatin revealed its localisation to SGs and retention for at least 24 h. We show cisplatin-induced SGs have impaired assembly, altered composition and are persistent, providing evidence of an alternate mechanism for cisplatin-induced ototoxicity via an impaired SG response.


Asunto(s)
Arsenitos , Ototoxicidad , Humanos , Cisplatino/farmacología , Arsenitos/toxicidad , Arsenitos/metabolismo , Ototoxicidad/metabolismo , Gránulos de Estrés , Gránulos Citoplasmáticos/metabolismo
12.
Neurobiol Dis ; 183: 106176, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37263384

RESUMEN

Aminoglycoside antibiotics (AGAs) are widely used in life-threatening infections, but they accumulate in cochlear hair cells (HCs) and result in hearing loss. Increases in adenosine triphosphate (ATP) concentrations and P2X7 receptor expression were observed after neomycin treatment. Here, we demonstrated that P2X7 receptor, which is a non-selective cation channel that is activated by high ATP concentrations, may participate in the process through which AGAs enter hair cells. Using transgenic knockout mice, we found that P2X7 receptor deficiency protects HCs against neomycin-induced injury in vitro and in vivo. Subsequently, we used fluorescent gentamicin-Fluor 594 to study the uptake of AGAs and found fluorescence labeling in wild-type mice but not in P2rx7-/- mice in vitro. In addition, knocking-out P2rx7 did not significantly alter the HC count and auditory signal transduction, but it did inhibit mitochondria-dependent oxidative stress and apoptosis in the cochlea after neomycin exposure. We thus conclude that the P2X7 receptor may be linked to the entry of AGAs into HCs and is likely to be a therapeutic target for auditory HC protection.


Asunto(s)
Aminoglicósidos , Ototoxicidad , Animales , Ratones , Aminoglicósidos/toxicidad , Aminoglicósidos/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Ototoxicidad/metabolismo , Antibacterianos/toxicidad , Neomicina/toxicidad , Neomicina/metabolismo , Células Ciliadas Auditivas/metabolismo , Cóclea , Adenosina Trifosfato/metabolismo
13.
Hear Res ; 434: 108783, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37167889

RESUMEN

Sensorineural hearing loss (SNHL) can either be genetically inherited or acquired as a result of aging, noise exposure, or ototoxic drugs. Although the precise pathophysiological mechanisms underlying SNHL remain unclear, an overwhelming body of evidence implicates mitochondrial dysfunction and oxidative stress playing a central etiological role. With its high metabolic demands, the cochlea, particularly the sensory hair cells, stria vascularis, and spiral ganglion neurons, is vulnerable to the damaging effects of mitochondrial reactive oxygen species (ROS). Mitochondrial dysfunction and consequent oxidative stress in cochlear cells can be caused by inherited mitochondrial DNA (mtDNA) mutations (hereditary hearing loss and aminoglycoside-induced ototoxicity), accumulation of acquired mtDNA mutations with age (age-related hearing loss), mitochondrial overdrive and calcium dysregulation (noise-induced hearing loss and cisplatin-induced ototoxicity), or accumulation of ototoxic drugs within hair cell mitochondria (drug-induced hearing loss). In this review, we provide an overview of our current knowledge on the role of mitochondrial dysfunction and oxidative stress in the development of SNHL caused by genetic mutations, aging, exposure to excessive noise, and ototoxic drugs. We also explore the advancements in antioxidant therapies for the different forms of acquired SNHL that are being evaluated in preclinical and clinical studies.


Asunto(s)
Pérdida Auditiva Provocada por Ruido , Pérdida Auditiva Sensorineural , Ototoxicidad , Humanos , Ototoxicidad/metabolismo , Pérdida Auditiva Sensorineural/inducido químicamente , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Estrés Oxidativo , Pérdida Auditiva Provocada por Ruido/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , ADN Mitocondrial/uso terapéutico , Mitocondrias/metabolismo
14.
J Cancer Res Ther ; 19(Supplement): S0, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37147955

RESUMEN

Introduction: Chemotherapeutic agents can have both serious side effects and ototoxicity, which can be caused by direct toxic effects or by metabolic derangement by the agents. Cabazitaxel (CBZ) is a next-generation semi-synthetic taxane derivative that is effective in both preclinical models of human tumors that are sensitive or resistant to chemotherapy and in patients suffering from progressive prostate cancer despite docetaxel treatment. The primary aim of this study is to investigate the ototoxicity of CBZ in a rat model. Materials and Methods: : A total of 24 adult male Wistar-Albino rats were equally and randomly divided into four groups. CBZ (Jevtana, Sanofi-Aventis USA) was intraperitoneally administered to Groups 2, 3, and 4 at doses of 0.5, 1.0, and 1.5 mg/kg/week, respectively, for 4 consecutive weeks; Group 1 received only i.p. saline at the same time. At the end of the study, the animals were sacrificed and their cochlea removed for histopathological examination. Results: : Intraperitoneal administration of CBZ exerted an ototoxic effect on rats, and the histopathological results became worse in a dose-dependent manner (P < 0.05). Conclusion: : Our findings suggest that CBZ may be an ototoxic agent and can damage the cochlea. More clinical studies should be conducted to understand its ototoxicity.


Asunto(s)
Antineoplásicos , Ototoxicidad , Neoplasias de la Próstata , Humanos , Animales , Ratas , Masculino , Antineoplásicos/toxicidad , Antineoplásicos/metabolismo , Ototoxicidad/metabolismo , Ototoxicidad/patología , Ratas Wistar , Cóclea/metabolismo , Cóclea/patología , Neoplasias de la Próstata/patología
15.
Free Radic Biol Med ; 204: 177-183, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37119862

RESUMEN

Aminoglycoside, a medicinal category of antibiotics, are used in treatment of Gram-negative bacterial infections. Although they are the most widely-used antibiotics due to their high efficacy and low cost, several main adverse effects have been reported including nephrotoxicity and ototoxicity. Since drug-induced ototoxicity is one of the major etiological causes of acquired hearing loss, we examined cochlear hair cell damages caused by three aminoglycosides (amikacin, kanamycin, and gentamicin), and investigated protective property of an isoquinoline-type alkaloid, Berberine chloride (BC). Berberine, a well-known bioactive compound found from medicinal plants, has been known to have anti-inflammatory, antimicrobial effects. To determine protective effect of BC in aminoglycoside-induced ototoxicity, hair cell damages in aminoglycoside- and/or BC-treated hair cells using ex vivo organotypic culture system of mouse cochlea. Mitochondrial ROS levels and depolarization of mitochondrial membrane potential were analyzed, and TUNEL assay and immunostaining of cleaved caspase-3 were performed to detect apoptosis signals. As the results, it was found that BC significantly prevented aminoglycoside-induced hair cell loss and stereocilia degeneration by inhibiting excessive accumulation of mitochondrial ROS and subsequent loss of mitochondrial membrane potential. It eventually inhibited DNA fragmentation and caspase-3 activation, which were significant for all three aminoglycosides. This study is the first report suggested the preventative effect of BC against aminoglycoside-induced ototoxicity. Our data also suggests a possibility that BC has the potential to exert a protective effect against ototoxicity caused by various ototoxic drugs leading to cellular oxidative stress, not limited to aminoglycoside antibiotics.


Asunto(s)
Berberina , Ototoxicidad , Ratones , Animales , Aminoglicósidos/toxicidad , Aminoglicósidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ototoxicidad/etiología , Ototoxicidad/prevención & control , Ototoxicidad/metabolismo , Berberina/farmacología , Caspasa 3/genética , Caspasa 3/metabolismo , Cloruros , Antibacterianos/efectos adversos , Células Ciliadas Auditivas
16.
Neurobiol Dis ; 182: 106134, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37100209

RESUMEN

The vestibular ganglion contains primary sensory neurons that are postsynaptic to the transducing hair cells (HC) and project to the central nervous system. Understanding the response of these neurons to HC stress or loss is of great interest as their survival and functional competence will determine the functional outcome of any intervention aiming at repair or regeneration of the HCs. We have shown that subchronic exposure to the ototoxicant 3,3'-iminodipropionitrile (IDPN) in rats and mice causes a reversible detachment and synaptic uncoupling between the HCs and the ganglion neurons. Here, we used this paradigm to study the global changes in gene expression in vestibular ganglia using RNA-seq. Comparative gene ontology and pathway analyses of the data from both model species indicated a robust downregulation of terms related to synapses, including presynaptic and postsynaptic functions. Manual analyses of the most significantly downregulated transcripts identified genes with expressions related to neuronal activity, modulators of neuronal excitability, and transcription factors and receptors that promote neurite growth and differentiation. For choice selected genes, the mRNA expression results were replicated by qRT-PCR, validated spatially by RNA-scope, or were demonstrated to be associated with decreased expression of the corresponding protein. We conjectured that decreased synaptic input or trophic support on the ganglion neurons from the HC was triggering these expression changes. To support this hypothesis, we demonstrated decreased expression of BDNF mRNA in the vestibular epithelium after subchronic ototoxicity and also downregulated expression of similarly identified genes (e.g Etv5, Camk1g, Slc17a6, Nptx2, Spp1) after HC ablation with another ototoxic compound, allylnitrile. We conclude that vestibular ganglion neurons respond to decreased input from HCs by decreasing the strength of all their synaptic contacts, both as postsynaptic and presynaptic players.


Asunto(s)
Ototoxicidad , Roedores , Ratas , Ratones , Animales , Roedores/metabolismo , Ototoxicidad/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ADN/metabolismo
17.
Biochim Biophys Acta Mol Cell Res ; 1870(5): 119461, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36931607

RESUMEN

As an anticancer drug, cisplatin is widely used, but its clinical application is restricted due to its severe side effects of ototoxicity. Therefore, this study was dedicated to assessing the benefit of ginsenoside extract, 20(S)-Ginsenoside Rh1 (Rh1), on cisplatin-induced ototoxicity. HEI-OC1 cells and neonatal cochlear explants were cultured. Cleaved caspase-3, TUNEL, and MitoSOX Red were observed in vitro by immunofluorescence staining. CCK8 and LDH cytotoxicity assays were detected to measure cell viability and cytotoxicity. Our results showed that Rh1 significantly increased cell viability, reduced cytotoxicity, and alleviated cisplatin-induced apoptosis. In addition, Rh1 pretreatment decreased the excessive accumulation of intracellular reactive oxygen species. Mechanistic studies indicated that Rh1 pretreatment reversed the increase of apoptotic protein expression, accumulation of mitochondrial ROS, and activation of the MAPK signaling pathway. These results suggested that Rh1 can act as an antioxidant and anti-apoptotic agent against cisplatin-induced hearing loss by suppressing the excessive accumulation of mitochondrial ROS, activation of MAPK signaling pathway and apoptosis.


Asunto(s)
Ginsenósidos , Pérdida Auditiva , Ototoxicidad , Humanos , Recién Nacido , Cisplatino/efectos adversos , Ginsenósidos/efectos adversos , Ginsenósidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ototoxicidad/metabolismo , Células Ciliadas Auditivas , Pérdida Auditiva/inducido químicamente , Apoptosis , Sistema de Señalización de MAP Quinasas
18.
Adv Ther ; 40(4): 1357-1365, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36738370

RESUMEN

Aminoglycosides are a class of medications used to treat certain bacterial infections, specifically gram-negative aerobes. These drugs can be used alone as first-line treatments or in combination with other medications. There can be many different formulations of aminoglycosides including oral, inhalants, intravascular, intramuscular, or intraventricular. There are many distinctive types of aminoglycosides, and although they provide excellent coverage, they can have a wide variety of side effects. The most prevalent side effects of aminoglycosides are nephrotoxicity and ototoxicity. Aminoglycoside-induced nephrotoxicity is concerning because of the effects that abnormal creatinine levels can have on other drugs and the potential for neurotoxicity. Fortunately, changes in renal function are typically reversible. The kidney is affected by the drug's ability to enter the proximal tubule and cause a buildup of phospholipids in the lysosomes, inhibiting their function. Exposure to aminoglycosides in utero can result in permanent ototoxicity. The mechanism of ototoxicity is through the drug's ability to freely pass into hair cells and cause reactive oxygen species to damage the mitochondria, resulting in cell death. There is not a substantial amount of research regarding the prevention and treatment of adverse effects of aminoglycosides. Future research on the mediation or modulation of these pathophysiological processes would expand their usage in modern medicine.


Asunto(s)
Infecciones Bacterianas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Ototoxicidad , Humanos , Aminoglicósidos/efectos adversos , Aminoglicósidos/metabolismo , Ototoxicidad/etiología , Ototoxicidad/metabolismo , Antibacterianos/efectos adversos , Riñón
19.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769271

RESUMEN

Cisplatin-induced ototoxicity leads to hearing impairment, possibly through reactive oxygen species (ROS) production and DNA damage in cochlear hair cells (HC), although the exact mechanism is unknown. Avenanthramide-C (AVN-C), a natural, potent antioxidant, was evaluated in three study groups of normal adult C57Bl/6 mice (control, cisplatin, and AVN-C+cisplatin) for the prevention of cisplatin-induced hearing loss. Auditory brainstem responses and immunohistochemistry of outer hair cells (OHCs) were ascertained. Cell survival, ROS production, Phospho-H2AX-enabled tracking of DNA damage-repair kinetics, and expression levels of inflammatory cytokines (TNF-α, IL-1ß, IL6, iNOS, and COX2) were assessed using House Ear Institute-Organ of Corti 1 (HEI-OC1 Cells). In the in vivo mouse model, following cisplatin-induced damage, AVN-C decreased the hearing thresholds and sheltered all cochlear turns' OHCs. In HEI-OC1 cells, AVN-C preserved cell viability and decreased ROS production, whereas cisplatin enhanced both ROS levels and cell viability. In HEI-OC1 cells, AVN-C downregulated IL6, IL-1ß, TNF-α, iNOS, and COX2 production that was upregulated by cisplatin treatment. AVN-C attenuated the cisplatin-enhanced nuclear H2AX activation. AVN-C had a strong protective effect against cisplatin-induced ototoxicity through inhibition of ROS and inflammatory cytokine production and DNA damage and is thus a promising candidate for preventing cisplatin-induced sensorineural hearing loss.


Asunto(s)
Antineoplásicos , Pérdida Auditiva , Ototoxicidad , Ratones , Animales , Cisplatino/toxicidad , Cisplatino/metabolismo , Citocinas/metabolismo , Antineoplásicos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ototoxicidad/etiología , Ototoxicidad/metabolismo , Interleucina-6/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ciclooxigenasa 2/metabolismo , Línea Celular , Apoptosis , Células Ciliadas Auditivas/metabolismo , Estrés Oxidativo , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/prevención & control , Pérdida Auditiva/metabolismo , Daño del ADN
20.
Int J Mol Sci ; 24(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36834972

RESUMEN

Cisplatin is a widely used standard chemotherapy for various cancers. However, cisplatin treatment is associated with severe ototoxicity. Fucoidan is a complex sulfated polysaccharide mainly derived from brown seaweeds, and it shows multiple bioactivities such as antimicrobial, anti-inflammatory, anticancer, and antioxidant activities. Despite evidence of the antioxidant effects of fucoidan, research on its otoprotective effects remains limited. Therefore, the present study investigated the otoprotective effects of fucoidan in vitro using the mouse cochlear cell line UB/OC-2 to develop new strategies to attenuate cisplatin-induced ototoxicity. We quantified the cell membrane potential and analyzed regulators and cascade proteins in the apoptotic pathway. Mouse cochlear UB/OC-2 cells were pre-treated with fucoidan before cisplatin exposure. The effects on cochlear hair cell viability, mitochondrial function, and apoptosis-related proteins were determined via flow cytometry, Western blot analysis, and fluorescence staining. Fucoidan treatment reduced cisplatin-induced intracellular reactive oxygen species production, stabilized mitochondrial membrane potential, inhibited mitochondrial dysfunction, and successfully protected hair cells from apoptosis. Furthermore, fucoidan exerted antioxidant effects against oxidative stress by regulating the Nrf2 pathway. Therefore, we suggest that fucoidan may represent a potential therapeutic agent for developing a new otoprotective strategy.


Asunto(s)
Antineoplásicos , Ototoxicidad , Polisacáridos , Animales , Ratones , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Antioxidantes/farmacología , Apoptosis , Cisplatino/toxicidad , Ototoxicidad/tratamiento farmacológico , Ototoxicidad/metabolismo , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA