Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 177: 117025, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38941893

ABSTRACT

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.


Subject(s)
Activating Transcription Factor 6 , Apoptosis , Cisplatin , Ototoxicity , Unfolded Protein Response , Cisplatin/toxicity , Animals , Activating Transcription Factor 6/metabolism , Ototoxicity/prevention & control , Ototoxicity/etiology , Ototoxicity/pathology , Mice , Unfolded Protein Response/drug effects , Apoptosis/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Cell Line , Male , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss/chemically induced , Hearing Loss/metabolism , Hearing Loss/pathology , Hearing Loss/prevention & control , Mice, Inbred C57BL , Transcription Factor CHOP/metabolism
2.
Sci Rep ; 14(1): 10910, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740884

ABSTRACT

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.


Subject(s)
Kanamycin , Ototoxicity , Signal Transduction , Transforming Growth Factor beta , Animals , Mice , Cochlea/metabolism , Cochlea/drug effects , Cochlea/pathology , Furosemide/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Kanamycin/toxicity , Mice, Inbred C57BL , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Signal Transduction/drug effects , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Transforming Growth Factor beta/metabolism
3.
J Cancer Res Ther ; 19(Supplement): S0, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37147955

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Ototoxicity , Prostatic Neoplasms , Humans , Animals , Rats , Male , Antineoplastic Agents/toxicity , Antineoplastic Agents/metabolism , Ototoxicity/metabolism , Ototoxicity/pathology , Rats, Wistar , Cochlea/metabolism , Cochlea/pathology , Prostatic Neoplasms/pathology
4.
Biochem Pharmacol ; 197: 114904, 2022 03.
Article in English | MEDLINE | ID: mdl-34971589

ABSTRACT

Cisplatin is a widely used chemotherapeutic agent for the treatment of various tumors, but its side effects limit its application. Ototoxicity, a major adverse effect of cisplatin, causes irreversible sensorineural hearing loss. Unfortunately, there are no effective approaches to protect against this damage. Autophagy has been shown to exert beneficial effects in various diseases models. However, the role of autophagy in cisplatin-induced ototoxicity has been not well elucidated. In this study, we aimed to investigate whether the novel autophagy activator trehalose could prevent cisplatin-induced damage in the auditory cell line HEI-OC1 and mouse cochlear explants and to further explore its mechanisms. Our data demonstrated that trehalose alleviated cisplatin-induced hair cell (HC) damage by inhibiting apoptosis, attenuating oxidative stress and rescuing mitochondrial dysfunction. Additionally, trehalose significantly enhanced autophagy levels in HCs, and inhibiting autophagy with 3-methyladenine (3-MA) abolished these protective effects. Mechanistically, we showed that the effect of trehalose was attributed to increased nuclear translocation of transcription factor EB (TFEB), and this effect could be mimicked by TFEB overexpression and inhibited by TFEB gene silencing or treatment with cyclosporin A (CsA), a calcineurin inhibitor. Taken together, our findings suggest that trehalose and autophagy play a role in protecting against cisplatin-induced ototoxicity and that pharmacological enhancement of TFEB-mediated autophagy is a potential treatment for cisplatin-induced damage in cochlear HCs and HEI-OC1 cells.


Subject(s)
Antineoplastic Agents/toxicity , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cisplatin/toxicity , Hair Cells, Auditory/metabolism , Trehalose/pharmacology , Animals , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/agonists , Cell Line , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Mice , Mice, Inbred C57BL , Ototoxicity/pathology , Ototoxicity/prevention & control
5.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884516

ABSTRACT

Telmisartan (TM) has been proposed to relieve inflammatory responses by modulating peroxisome proliferator activator receptor-γ (PPARγ) signaling. This study aimed to investigate the protective effects of TM on kanamycin(KM)-induced ototoxicity in rats. Forty-eight, 8-week-old female Sprague Dawley rats were divided into four groups: (1) control group, (2) TM group, (3) KM group, and (4) TM + KM group. Auditory brainstem response was measured. The histology of the cochlea was examined. The protein expression levels of angiotensin-converting enzyme 2 (ACE2), HO1, and PPARγ were measured by Western blotting. The auditory threshold shifts at 4, 8, 16, and 32 kHz were lower in the TM + KM group than in the KM group (all p < 0.05). The loss of cochlear outer hair cells and spiral ganglial cells was lower in the TM + KM group than in the KM group. The protein expression levels of ACE2, PPARγ, and HO1 were higher in the KM group than in the control group (all p < 0.05). The TM + KM group showed lower expression levels of PPARγ and HO1 than the KM group.TM protected the cochlea from KM-induced injuries in rats. TM preserved hearing levels and attenuated the increase in PPARγ and HO1 expression levels in KM-exposed rat cochleae.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Kanamycin/toxicity , Ototoxicity/drug therapy , PPAR gamma/metabolism , Telmisartan/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Animals , Anti-Bacterial Agents/toxicity , Antihypertensive Agents/pharmacology , Auditory Threshold/drug effects , Cochlea/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Heme Oxygenase (Decyclizing)/genetics , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , PPAR gamma/genetics , Rats , Rats, Sprague-Dawley
6.
Med Sci Monit ; 27: e933278, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34657931

ABSTRACT

BACKGROUND Sodium salicylate (SS) induces excitotoxicity of spiral ganglion neurons (SGNs) by inhibiting the response of γ-aminobutyric acid type A receptors (GABAARs). Our previous studies have shown that SS can increase the internalization of GABAARs on SGNs, which involves dopamine D1-like receptors (D1Rs) and related signaling pathways. In this study, we aimed to explore the role of D1Rs and their downstream molecule protein kinase C (PKC) in the process of SS inhibiting GABAARs. MATERIAL AND METHODS The expression of D1Rs and GABARγ2 on rat cochlear SGNs cultured in vitro was tested by immunofluorescence. Then, the SGNs were exposed to SS, D1R agonist (SKF38393), D1R antagonist (SCH23390), clathrin/dynamin-mediated endocytosis inhibitor (dynasore), and PKC inhibitor (Bisindolylmaleimide I). Western blotting and whole-cell patch clamp technique were used to assess the changes of surface and total protein of GABARγ2 and GABA-activated currents. RESULTS Immunofluorescence showed that D1 receptors (DRD1) were expressed on SGNs. Data from western blotting showed that SS promoted the internalization of cell surface GABAARs, and activating D1Rs had the same result. Inhibiting D1Rs and PKC decreased the internalization of GABAARs. Meanwhile, the phosphorylation level of GABAARγ2 S327 affected by PKC was positively correlated with the degree of internalization of GABAARs. Moreover, whole-cell patch clamp recording showed that inhibition of D1Rs or co-inhibition of D1Rs and PKC attenuated the inhibitory effect of SS on GABA-activated currents. CONCLUSIONS D1Rs mediate the GABAAR internalization induced by SS via a PKC-dependent manner and participate in the excitotoxic process of SGNs.


Subject(s)
Ototoxicity/pathology , Protein Kinase C/metabolism , Receptors, Dopamine D1/metabolism , Receptors, GABA-A/metabolism , Sodium Salicylate/adverse effects , Spiral Ganglion/pathology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines , Cells, Cultured , Disease Models, Animal , Female , Humans , Hydrazones/pharmacology , Male , Models, Animal , Neurons/drug effects , Neurons/metabolism , Ototoxicity/etiology , Patch-Clamp Techniques , Primary Cell Culture , Rats , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Spiral Ganglion/cytology , Spiral Ganglion/drug effects
7.
Biochem Cell Biol ; 99(3): 385-395, 2021 06.
Article in English | MEDLINE | ID: mdl-34077275

ABSTRACT

Resveratrol is a non-flavonoid polyphenol compound that exists in many plants, and is considered an antitoxin. This study explores the effects from the regulation of miR-455-5p by resveratrol on cisplatin-induced ototoxicity via the PTEN-PI3K-AKT signaling pathway. For this, House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were transfected with miR-455-5p inhibitor and treated with cisplatin and resveratrol, then cell proliferation, apoptosis, and oxidative stress were evaluated. A mouse model of hearing loss was established, and these mice were treated with cisplatin, resveratrol, or cisplatin combined with resveratrol, by intraperitoneal injection. The auditory brainstem response (ABR) threshold was measured, and hair cells were examined using immunofluorescence staining. The expression levels of miR-455-5p, PTEN, and PI3K/Akt proteins were examined. The results from our in-vitro experiments indicate that resveratrol promoted viability and reduced apoptosis and oxidative stress in cisplatin-induced HEI-OC1 cells. Resveratrol upregulated miR-455-5p, downregulated PTEN, and activated the PI3K-Akt axis. These effects of resveratrol were reversed by knock-down of miR-455-5p. The results from our in-vivo experiments indicate that resveratrol protected hearing and inhibited the hair-cell injury caused by cisplatin ototoxicity. Resveratrol also upregulated miR-455-5p, downregulated PTEN, and activated the PTEN-PI3K-Akt axis in cochlear tissues from cisplatin-treated mice. These results indicate that resveratrol upregulates miR-455-5p to target PTEN and activate the PI3K-Akt signaling pathway to counteract cisplatin ototoxicity.


Subject(s)
Cisplatin/toxicity , MicroRNAs/genetics , Ototoxicity/drug therapy , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Resveratrol/pharmacology , Animals , Antineoplastic Agents/toxicity , Antioxidants/pharmacology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
8.
J Pediatr Hematol Oncol ; 43(7): e930-e934, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-33885038

ABSTRACT

The aim of this study is to evaluate risk factors contributing to the development of ototoxicity in children who received platinum-based chemotherapy for malignancies located in the head and neck region. Eighty-four children who received platinum-based chemotherapy were included. Audiologic evaluations were performed before and after each chemotherapy session through pure tone audiometry, distortion product otoacoustic emissions, and auditory brainstem response tests. Ototoxicity was evaluated using Brock, Muenster, and Chang classifications. Factors such as cranial irradiation, cumulative doses of cisplatin, age, sex, cotreatment with aminoglycosides, schedule of platinum, and type of chemotherapeutic agent were analyzed. Using χ2 tests, all risk factors were matched with the 3 ototoxicity classifications, and multivariate analyses were conducted using statistically significant risk factors. In univariate analyses, being between 5 and 12 years of age, cranial irradiation and being treated with both cisplatin and carboplatin were found to be related to ototoxicity in all 3 classifications. Logistic regression modeling analyses with these 3 risk factors showed that being between 5 and 12 years of age and being treated with both cisplatin and carboplatin significantly increased the risk of ototoxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cranial Irradiation/adverse effects , Head and Neck Neoplasms/drug therapy , Ototoxicity/etiology , Adolescent , Carboplatin/administration & dosage , Child , Child, Preschool , Cisplatin/administration & dosage , Female , Follow-Up Studies , Head and Neck Neoplasms/pathology , Humans , Infant , Male , Ototoxicity/pathology , Prognosis , Retrospective Studies , Risk Factors
9.
Biochem Pharmacol ; 186: 114513, 2021 04.
Article in English | MEDLINE | ID: mdl-33713642

ABSTRACT

Gentamicin (GM), an aminoglycoside antibiotic, is one commonly used clinical drugs with ototoxic side effects. One of the most principal mechanisms of its ototoxicity is that GM can activate caspase-mediated cell death pathways in the cochlea. Since the anti-apoptotic protein known as X-linked Inhibitor of Apoptosis Protein (XIAP) has been reported to directly bind to activated caspase protein and inhibit their activities, we hypothesized that it might protect cochlea hair cells from GM ototoxicity. To evaluate this hypothesis, postnatal day 2-3 (P2-3) transgenic (TG) mice, in which XIAP gene is over-expressed under a pure C57BL/6J genetic background was constructed. We first extracted the cochlea tissue of normal mice and treated them with different concentrations of GM, and the number of hair cells were observed to determine the concentration of GM used in subsequent experiments. Next, we used Western Blot experiment to examine the effect of GM on XIAP protein expression in normal mouse cochlea, and then Western Blot and RT-PCR experiments were used to identify the transgenic mice. Finally, immunofluorescence assays were used to detect the effect of GM on the expression of caspase protein and verify the protective effect of XIAP. We found that GM at a concentration of 0.5 mM significantly affected the function of cochlea hair cells, up-regulating the expression of cleaved-caspase-3 and cleaved-caspase-9 protein but down-regulating XIAP protein. In the cochlea tissues of TG mice, this effect of GM was suppressed, and the destruction of hair cells was significantly reduced, and the cleaved-caspase-3 and cleaved-caspase-9 proteins were significantly suppressed. These results suggested that XIAP reduces GM-induced ototoxicity and caspase-3/9 pathway is associated with this process.


Subject(s)
Caspase 3/metabolism , Caspase 9/metabolism , Gentamicins/toxicity , Hair Cells, Auditory/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Ototoxicity/metabolism , Animals , Dose-Response Relationship, Drug , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Ototoxicity/pathology , Ototoxicity/prevention & control , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Biomed Environ Sci ; 34(2): 110-118, 2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33685569

ABSTRACT

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.


Subject(s)
Ear, Inner/drug effects , Lateral Line System/drug effects , Toluene/toxicity , Animals , Ear, Inner/growth & development , Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Lateral Line System/growth & development , Locomotion/drug effects , Ototoxicity/etiology , Ototoxicity/pathology , Ototoxicity/physiopathology , Zebrafish
11.
Arch Biochem Biophys ; 701: 108752, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33675811

ABSTRACT

Hearing loss caused by ototoxic drugs is a kind of acquired hearing loss. Cisplatin is one of the most commonly used drugs and its main action sites are hair cells (HCs). Sorcin is a drug-resistant calcium-binding protein belonging to the small penta-EF-hand protein family. Sorcin is highly expressed in many tissues, including bone, heart, brain, lung, and skin tissues. Single-cell RNA sequencing showed that sorcin was expressed in the outer HCs of mice, but its role remained unknown. We also found that sorcin was highly expressed in the cytoplasm of cochlear HCs and HEI-OC1 cells. After cisplatin injury, the expression of sorcin in HCs and HEI-OC1 cells decreased significantly. SiRNA transfection technology was used to knock down the expression of sorcin. The results showed that the number of apoptotic cells, the expression of cleaved caspased-3, and the expression of Bax increased while the anti-apoptotic factor Bcl-2 decreased in the siRNA-Sorcin + CIS group. The observed increase in apoptosis was related to the increase of reactive oxygen species (ROS) and the destruction of the mitochondrial membrane potential (MMP). Finally, we found that the downregulated sorcin worked by activating the P-ERK1/2 signaling pathway. Overall, this study showed that sorcin can be used as a new target to prevent the ototoxicity of platinum drugs.


Subject(s)
Apoptosis/drug effects , Calcium-Binding Proteins/biosynthesis , Cisplatin/adverse effects , Hair Cells, Auditory/metabolism , MAP Kinase Signaling System/drug effects , Ototoxicity/metabolism , Animals , Apoptosis/genetics , Calcium-Binding Proteins/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Cisplatin/pharmacology , Gene Knockdown Techniques , Hair Cells, Auditory/pathology , MAP Kinase Signaling System/genetics , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice , Ototoxicity/genetics , Ototoxicity/pathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Biomed Pharmacother ; 137: 111281, 2021 May.
Article in English | MEDLINE | ID: mdl-33578233

ABSTRACT

BACKGROUND: Acrolein is a reactive aldehyde that forms during burning of wood and other fuels. It is also a product of lipid peroxidation (LPO) reactions and is present in cigarette smoke. Acrolein is known to cause oxidative stress and inflammatory nerve tissue damage. Lutein is a tetraterpenoid molecule with antioxidant and anti-inflammatory properties. There appear to be no studies on the effect of lutein on vestibulocochlear nerve damage induced by acrolein. The aim of this study was to investigate the effect of lutein on vestibulocochlear nerve damage induced by acrolein in rats using biochemical and histopathological methods. METHODS: The rats were divided into three groups (n = 6, for each group) a healthy control group (HG), an acrolein (ACR) group and a lutein and acrolein (LACR) group. In the LACR group, lutein was administered (1 mg/kg) via oral gavage. The ACR and HG groups received saline via oral gavage. Then, 1 h after the administration of lutein and saline, the LACR and ACR groups were treated with 3 mg/kg of acrolein via oral gavage. This procedure was repeated once a day for 30 days. RESULTS: The results of biochemical experiments showed that in the vestibulocochlear nerve tissues of the animals treated with acrolein, the levels of malondialdehyde, total oxidants, nuclear factor kappa b, tumor necrosis factor alpha and interleukin 1 beta significantly increased, whereas the levels of total glutathione and total antioxidants decreased as compared to those in the HG and LACR groups. In addition, severe histopathological damage was observed in vestibulocochlear nerve tissue of the acrolein group, whereas this damage was alleviated in the lutein group. CONCLUSION: Lutein protected vestibulocochlear nerve tissue from acrolein-associated oxidative and proinflammatory damage. This suggests that lutein might be useful in preventing or treating acrolein-induced ototoxicity.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Inflammation Mediators/metabolism , Lutein/pharmacology , Ototoxicity/prevention & control , Oxidative Stress/drug effects , Vestibulocochlear Nerve Diseases/prevention & control , Vestibulocochlear Nerve/drug effects , Acrolein , Animals , Disease Models, Animal , Male , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Rats, Wistar , Vestibulocochlear Nerve/metabolism , Vestibulocochlear Nerve/pathology , Vestibulocochlear Nerve Diseases/chemically induced , Vestibulocochlear Nerve Diseases/metabolism , Vestibulocochlear Nerve Diseases/pathology
13.
Toxicology ; 453: 152736, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33631298

ABSTRACT

Cisplatin-induced ototoxicity is one of the important reasons that limit the drug's clinical application, and its mechanism has not been fully elucidated so far. The aim of this study was to explore the attenuate effect of tauroursodeoxycholic acid (TUDCA), a proteostasis promoter, on cisplatin-induced ototoxicity in vivo and in vitro, and to explore its possible mechanism. Auditory brainstem response (ABR) was measured to identify the attenuate effects of TUDCA administered subcutaneously [500 mg/kg/d × 3d, cisplatin: 4.6 mg/kg/d × 3d, intraperitoneal injection (i.p.)] or trans-tympanically (0.5 mg/mL, cisplatin: 12 mg/kg, i.p. with a pump) in Sprague-Dawley (SD) rats subjected to cisplatin-induced hearing loss. The cochlear explants of neonatal rats and OC1 auditory hair cell-like cell lines cultured in vitro were used to observe the number of apoptotic cells and the endoplasmic reticulum (ER) stress in the control, cisplatin (5 µM for 48 h for cochlear explants, 10 µM for 24 h for OC1 cells), and cisplatin + TUDCA (1 mM for 24 h for cochlear explants, 1.6 mM for 24 h for OC1 cells) groups. Differences in the expression of key proteins in the ER protein quality control (ERQC) system were detected. The changes in the attenuate effect of TUDCA on cisplatin-induced ototoxicity after down-regulating calreticulin (CRT), UDP-glucose ceramide glucosyltransferase-like 1 (UGGT1), and OS9 ER lectin (OS9) were also measured. The effect of TUDCA (10 mM) on stabilizing unfolded or misfolded proteins (UFP/MFP) was analyzed in a cell-free 0.2 % bovine serum albumin (BSA) aggregation system in vitro. Both the subcutaneous and trans-tympanic TUDCA administration alleviated cisplatin-induced increase in ABR thresholds in rats. TUDCA was able to reduce cisplatin-induced apoptosis and alleviate ER stress in cochlear explants and OC1 cells. Under the cisplatin treatment, the expression levels of CRT, UGGT1, and OS9 in the auditory hair cell increased, and the expression of total ubiquitinated proteins decreased. TUDCA attenuated the effect of cisplatin on UGGT1 and OS9, and recovered the protein ubiquitination levels. After down-regulating CRT, UGGT1, or OS9, the protective effect of TUDCA decreased. In the cell-free experimental system, TUDCA inhibited the aggregation of denatured BSA molecules. In summary, TUDCA can attenuate cisplatin-induced ototoxicity, possibly by inhibiting the accumulation and aggregation of UFP/MFP and the associated ER stress.


Subject(s)
Cisplatin/toxicity , Endoplasmic Reticulum/drug effects , Ototoxicity/prevention & control , Protein Aggregates/drug effects , Protein Folding/drug effects , Taurochenodeoxycholic Acid/therapeutic use , Animals , Antineoplastic Agents/toxicity , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Endoplasmic Reticulum/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Male , Ototoxicity/pathology , Protein Aggregates/physiology , Rats , Rats, Sprague-Dawley , Taurochenodeoxycholic Acid/pharmacology
14.
Cell Death Dis ; 12(1): 3, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33414397

ABSTRACT

Macroautophagy/autophagy is a highly conserved self-digestion pathway that plays an important role in cytoprotection under stress conditions. Autophagy is involved in hepatotoxicity induced by acetaminophen (APAP) in experimental animals and in humans. APAP also causes ototoxicity. However, the role of autophagy in APAP-induced auditory hair cell damage is unclear. In the present study, we investigated autophagy mechanisms during APAP-induced cell death in a mouse auditory cell line (HEI-OC1) and mouse cochlear explant culture. We found that the expression of LC3-II protein and autophagic structures was increased in APAP-treated HEI-OC1 cells; however, the degradation of SQSTM1/p62 protein, the yellow puncta of mRFP-GFP-LC3 fluorescence, and the activity of lysosomal enzymes decreased in APAP-treated HEI-OC1 cells. The degradation of p62 protein and the expression of lysosomal enzymes also decreased in APAP-treated mouse cochlear explants. These data indicate that APAP treatment compromises autophagic degradation and causes lysosomal dysfunction. We suggest that lysosomal dysfunction may be directly responsible for APAP-induced autophagy impairment. Treatment with antioxidant N-acetylcysteine (NAC) partially alleviated APAP-induced autophagy impairment and apoptotic cell death, suggesting the involvement of oxidative stress in APAP-induced autophagy impairment. Inhibition of autophagy by knocking down of Atg5 and Atg7 aggravated APAP-induced ER and oxidative stress and increased apoptotic cell death. This study provides a better understanding of the mechanism responsible for APAP ototoxicity, which is important for future exploration of treatment strategies for the prevention of hearing loss caused by ototoxic medications.


Subject(s)
Acetaminophen/adverse effects , Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Ototoxicity , Oxidative Stress/drug effects , Animals , Cell Line , Mice , Mice, Inbred C57BL , Ototoxicity/metabolism , Ototoxicity/pathology
15.
Oxid Med Cell Longev ; 2020: 1782659, 2020.
Article in English | MEDLINE | ID: mdl-33343803

ABSTRACT

Ferroptosis is a recently discovered iron-dependent form of oxidative programmed cell death distinct from caspase-dependent apoptosis. In this study, we investigated the effect of ferroptosis in neomycin-induced hair cell loss by using selective ferroptosis inhibitor liproxstatin-1 (Lip-1). Cell viability was identified by CCK8 assay. The levels of reactive oxygen species (ROS) were determined by DCFH-DA and cellROX green staining. The mitochondrial membrane potential (ΔΨm) was evaluated by TMRM staining. Intracellular iron and lipid peroxides were detected with Mito-FerroGreen and Liperfluo probes. We found that ferroptosis can be induced in both HEI-OC1 cells and neonatal mouse cochlear explants, as evidenced by Mito-FerroGreen and Liperfluo staining. Further experiments showed that pretreatment with Lip-1 significantly alleviated neomycin-induced increased ROS generation and disruption in ΔΨm in the HEI-OC1 cells. In parallel, Lip-1 significantly attenuated neomycin-induced hair cell damage in neonatal mouse cochlear explants. Collectively, these results suggest a novel mechanism for neomycin-induced ototoxicity and suggest that ferroptosis inhibition may be a new clinical intervention to prevent hearing loss.


Subject(s)
Hair Cells, Auditory/metabolism , Neomycin/adverse effects , Ototoxicity/prevention & control , Quinoxalines/pharmacology , Spiro Compounds/pharmacology , Animals , Cell Line , Hair Cells, Auditory/pathology , Mice , Neomycin/pharmacology , Ototoxicity/metabolism , Ototoxicity/pathology , Reactive Oxygen Species/metabolism
16.
J Cell Mol Med ; 24(20): 12065-12081, 2020 10.
Article in English | MEDLINE | ID: mdl-32929878

ABSTRACT

Ferroptosis is a recently recognized form of non-apoptotic cell death caused by an iron-dependent accumulation of lipid hydroperoxides, which plays important roles in a wide spectrum of pathological conditions. The present study was aimed to investigate the impact of ferroptosis on cisplatin-induced sensory hair cell damage. Cell viability was determined by Cell Counting Kit-8 and lactase dehydrogenase assays. The reactive oxygen species (ROS) levels were evaluated by 2,7-Dichlorodi-hydrofluorescein diacetate (DCFH-DA) and MitoSox-Red staining. Mitochondrial membrane potential (MMP) was measured by tetramethylrhodamine methyl ester (TMRM) staining. Lipid peroxidation, intracellular and mitochondrial iron were detected by Liperfluo, C11-BODIPY581/591 , FerroOrange and Mito-FerroGreen, respectively. We found that cisplatin treatment not only markedly augmented ROS accumulation, decreased the MMP, but increased lipid peroxidation and iron accumulation in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells. Of note, treatment with the specific ferroptosis inhibitor ferrostatin-1 could effectively abrogate the cisplatin-induced toxicity and subsequent cell death. Specifically, the improvement of mitochondrial functions is important mechanisms for protective action of ferroptosis inhibitor against cisplatin-induced damages in HEI-OC1 cells. Moreover, inhibition of ferroptosis significantly protected murine cochlear hair cells against cisplatin damage. In addition, treatment murine cochlear hair cells with ferroptosis inducer, RSL3, significantly exacerbated cisplatin-induced damage, which could be alleviated by ROS inhibitor N-acetyl-L-cysteine. Collectively, our study indicated that ferroptosis inhibition could alleviate the cisplatin-induced ototoxicity via inactivation of lipid peroxide radical and improvement of mitochondrial function in hair cells.


Subject(s)
Cisplatin/adverse effects , Cytoprotection , Ferroptosis , Hair Cells, Auditory/pathology , Ototoxicity/pathology , Aldehydes/metabolism , Animals , Carbolines/pharmacology , Cell Line , Cell Survival/drug effects , Cyclohexylamines/pharmacology , Cytoprotection/drug effects , Ferroptosis/drug effects , Hair Cells, Auditory/drug effects , Iron/metabolism , Iron Overload/pathology , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Phenylenediamines/pharmacology , Reactive Oxygen Species/metabolism
17.
Eur J Pharmacol ; 887: 173338, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32781170

ABSTRACT

Hydroxytyrosol (HT), a polyphenol widely contained as an ester in olive fruits and olive leaves, exhibits a broad spectrum of effectiveness. The present study was designed to investigate the effect of HT alone as well as in the combination with cisplatin on the House Ear Institute-Organ of Corti 1 cells (HEI-OC1) and C57BL/6 cochlear hair cells in vitro. The cell viability was measured by cell counting kit-8 (CCK8) assay. The levels of reactive oxygen species were evaluated by Dichloro-dihydro-fluorescein diacetate (DCFH-DA) staining. The expression of phosphorylated Jun N-terminal kinase (p-JNK) and cleaved-caspase 3 was assessed by Western blotting. The apoptosis was detected by terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL) staining. The distribution of apoptosis inducing factor (AIF) was determined by immunofluorescent staining. HT alleviated the levels of reactive oxygen species in both untreated state and after cisplatin stimulus. However, HT at concentration of 100 µM decreased the cell viability of HEI-OC1 from 100 ± 17.38% in control group to 50.17 ± 1.89% and increased the expression of p-JNK and c-caspase 3 from 0.62 ± 0.10, 0.20 ± 0.050 in the control group to 1.24 ± 0.18, 0.85 ± 0.18 in the group treated with 30 µM cisplatin, as well as to 1.64 ± 0.14, 1.44 ± 0.12 in the group with 30 µM cisplatin +100 µM HT, respectively. Meanwhile, HT triggered AIF transferring to nuclei and, also, led to cochlear HCs arranging disorderly and missing. Moreover, HT elevated the expression of p-JNK and c-caspase 3 from 1.00 ± 0.27, 1.00 ± 0.26 in the control group to 2.23 ± 0.24, 22.87 ± 3.80 in the group with 30 µM cisplatin, and to 2.75 ± 0.23, 31.56 ± 3.86 in the group with 30 µM cisplatin+100 µM HT correspondingly. Taken together, data from this work reveal that HT itself possesses toxic effect on HCs mainly thorough AIF-dependent apoptosis, while, it aggravates the ototoxicity-caused by cisplatin via both JNK and AIF pathways related apoptosis. Findings from this work offer clear evidence that that HT might not be recommended to utilize for preventing cisplatin-induced ototoxicity.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cisplatin/toxicity , MAP Kinase Signaling System/drug effects , Ototoxicity/metabolism , Phenylethyl Alcohol/analogs & derivatives , Animals , Animals, Newborn , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/toxicity , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cells, Cultured , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Ototoxicity/pathology , Phenylethyl Alcohol/administration & dosage , Phenylethyl Alcohol/toxicity
18.
Asian Pac J Cancer Prev ; 21(7): 2155-2162, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32711445

ABSTRACT

BACKGROUND: Cisplatin is an alkylating agent that inhibits DNA replication and interferes with proliferation of cancer cells. However, the major limiting factor for its use is the possible development of adverse effects, including ototoxicity. Up till now, the mechanisms of this ototoxicity remain poorly understood. However, induction of oxidative stress and activation of the inflammatory cascade were suggested as contributing factors. PURPOSE: The aim of this study was to explore the effect of L-arginine on cisplatin-induced ototoxicity in rats. METHODS: Thirty male adult Wistar rats were divided into three equal groups as follows: control group; cisplatin group and cisplatin + L-arginine group. Auditory brainstem response (ABR), tissue oxidative stress parameters, total nitrate/nitrite, nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase-1 (HO-1) content, transforming growth factor beta 1 (TGF-ß1), tumor necrosis factor alpha (TNF-α) and interleukin 15 (IL-15) were assessed. Also, the cochlear tissues were subjected to histopathological and electron microscopic examination. RESULTS: Administration of L-arginine to cisplatin-treated rats induced significant decrease in the average ABR threshold shifts at all frequencies, tissue TGF-ß1, TNF-α and IL-15 associated with significant increase in tissue antioxidant enzymes, total nitrate/nitrite and Nrf2/HO-1 content compared to cisplatin group. Also, pretreatment of cisplatin-injected rats with L-arginine induced significant improvement of the histopathological and electron microscopic picture compared to cisplatin group. CONCLUSION: L-arginine may serve as a promising therapeutic modality for amelioration of cisplatin-induced ototoxicity.
.


Subject(s)
Arginine/pharmacology , Cisplatin/toxicity , Heme Oxygenase (Decyclizing)/metabolism , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Ototoxicity/prevention & control , Transforming Growth Factor beta1/metabolism , Animals , Antineoplastic Agents/toxicity , Antioxidants/metabolism , Male , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Oxidative Stress , Rats , Rats, Wistar , Signal Transduction
19.
J Int Adv Otol ; 16(2): 218-221, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32510456

ABSTRACT

OBJECTIVES: The aim of this study was to assess whether nivolumab is ototoxic in rats and whether this ototoxicity is dose-dependent. MATERIALS AND METHODS: Twelve rats were divided into two groups: Group 1 (control group, 6 rats, 12 ears) received intraperitoneal saline for 14 days. Group 2 (study group, 6 rats, 12 ears) and received two doses of 3 mg/kg intraperitoneal nivolumab within 14 days. Auditory brainstem responses (ABRs) were performed preoperatively and 4 and 8 weeks postoperatively. We compared between the groups, morphologic appearance of spiral ganglion cells and organ of Corti and density of spiral ganglion cells (measured with conventional light microscope connected to a personal computer). RESULTS: In our control group, both spiral ganglion and organ of corti had a normal morphological appearance. In our study group, spiral ganglion cells had a normal morphological appearance. However, some sections showed possibly mild degenerative changes in the organ of corti. Of 12 samples in the study group, four had a significant loss of density of spiral ganglion cells compared to the control group. The baseline ABR thresholds did not significantly differ between the groups (p=0.713). There was no statistically significant difference between the groups regarding ABR thresholds at week 4 (p=0.347). However, a statistically significant difference was observed in the ABR thresholds at week 8 (p=0.045). CONCLUSION: The results of our study showed that nivolumab treatment has ototoxic effects. Based on our results, we recommend monitoring the changes in the hearing ability of chemotherapy patients.


Subject(s)
Antineoplastic Agents, Immunological/toxicity , Nivolumab/toxicity , Ototoxicity/etiology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Organ of Corti/drug effects , Ototoxicity/pathology , Rats , Spiral Ganglion/drug effects
20.
J Tissue Eng Regen Med ; 14(8): 1149-1156, 2020 08.
Article in English | MEDLINE | ID: mdl-32593214

ABSTRACT

Gentamicin-induced cochlear hair cell ototoxicity, such as oxidative stress and apoptosis, could be attenuated by mouse inner ear stem cells (IESCs). However, it is still unclear whether such protective effects could be mediated by exosomes derived from IESCs (IESCs-ex). In the present study, HEI-OC1 cells were exposed to gentamicin (2 mM) to establish an ototoxicity model and further treated with exosomes isolated from miR-182-5p transferred or non-transferred IESCs. IESCs-ex improved HEI-OC1 cell viability, as assayed by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide method, and alleviated the oxidative stress response induced by the gentamicin treatment, as confirmed by measuring the malondialdehyde, superoxide dismutase, catalase, and glutathione peroxidase levels. IESCs-ex increased relative miR-182-5p expression and decreased FOXO3 expression in the gentamicin-exposed HEI-OC1 cells. Furthermore, exosomes derived from miR-182-5p mimics that were pre-treated with IESCs could increase miR-182-5p and Bcl-2 expressions and decrease FOXO3 and Bax expressions in gentamicin-exposed HEI-OC1 cells. All of these results indicate that IESCs-ex could attenuate gentamicin-induced HEI-OC1 cell apoptosis and oxidative stress through the miR-182-5p/FOXO3 axis.


Subject(s)
Ear, Inner/metabolism , Exosomes , Forkhead Box Protein O3/metabolism , Gentamicins/adverse effects , MicroRNAs/metabolism , Ototoxicity , Animals , Ear, Inner/pathology , Exosomes/metabolism , Exosomes/pathology , Exosomes/transplantation , Gentamicins/pharmacology , Mice , Ototoxicity/metabolism , Ototoxicity/pathology , Ototoxicity/therapy , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL