Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.443
Filter
1.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000202

ABSTRACT

The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) protein plays an essential role in the cisplatin (CDDP)-induced generation of reactive oxygen species (ROS). In this study, we evaluated the suitability of ultrasound-mediated lysozyme microbubble (USMB) cavitation to enhance NOX4 siRNA transfection in vitro and ex vivo. Lysozyme-shelled microbubbles (LyzMBs) were constructed and designed for siNOX4 loading as siNOX4/LyzMBs. We investigated different siNOX4-based cell transfection approaches, including naked siNOX4, LyzMB-mixed siNOX4, and siNOX4-loaded LyzMBs, and compared their silencing effects in CDDP-treated HEI-OC1 cells and mouse organ of Corti explants. Transfection efficiencies were evaluated by quantifying the cellular uptake of cyanine 3 (Cy3) fluorescein-labeled siRNA. In vitro experiments showed that the high transfection efficacy (48.18%) of siNOX4 to HEI-OC1 cells mediated by US and siNOX4-loaded LyzMBs significantly inhibited CDDP-induced ROS generation to almost the basal level. The ex vivo CDDP-treated organ of Corti explants of mice showed an even more robust silencing effect of the NOX4 gene in the siNOX4/LyzMB groups treated with US sonication than without US sonication, with a marked abolition of CDDP-induced ROS generation and cytotoxicity. Loading of siNOX4 on LyzMBs can stabilize siNOX4 and prevent its degradation, thereby enhancing the transfection and silencing effects when combined with US sonication. This USMB-derived therapy modality for alleviating CDDP-induced ototoxicity may be suitable for future clinical applications.


Subject(s)
Cisplatin , Hair Cells, Auditory , Microbubbles , Muramidase , NADPH Oxidase 4 , Ototoxicity , Reactive Oxygen Species , Cisplatin/pharmacology , Animals , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Mice , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Reactive Oxygen Species/metabolism , Ototoxicity/genetics , Muramidase/genetics , RNA, Small Interfering/genetics , Ultrasonic Waves , Gene Knockdown Techniques , Cell Line
2.
Sci Rep ; 14(1): 15296, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961203

ABSTRACT

Blast wave exposure, a leading cause of hearing loss and balance dysfunction among military personnel, arises primarily from direct mechanical damage to the mechanosensory hair cells and supporting structures or indirectly through excessive oxidative stress. We previously reported that HK-2, an orally active, multifunctional redox modulator (MFRM), was highly effective in reducing both hearing loss and hair cells loss in rats exposed to a moderate intensity workday noise that likely damages the cochlea primarily from oxidative stress versus direct mechanical trauma. To determine if HK-2 could also protect cochlear and vestibular cells from damage caused primarily from direct blast-induced mechanical trauma versus oxidative stress, we exposed rats to six blasts of 186 dB peak SPL. The rats were divided into four groups: (B) blast alone, (BEP) blast plus earplugs, (BHK-2) blast plus HK-2 and (BEPHK-2) blast plus earplugs plus HK-2. HK-2 was orally administered at 50 mg/kg/d from 7-days before to 30-day after the blast exposure. Cochlear and vestibular tissues were harvested 60-d post-exposure and evaluated for loss of outer hair cells (OHC), inner hair cells (IHC), auditory nerve fibers (ANF), spiral ganglion neurons (SGN) and vestibular hair cells in the saccule, utricle and semicircular canals. In the untreated blast-exposed group (B), massive losses occurred to OHC, IHC, ANF, SGN and only the vestibular hair cells in the striola region of the saccule. In contrast, rats treated with HK-2 (BHK-2) sustained significantly less OHC (67%) and IHC (57%) loss compared to the B group. OHC and IHC losses were smallest in the BEPHK-2 group, but not significantly different from the BEP group indicating lack of protective synergy between EP and HK-2. There was no loss of ANF, SGN or saccular hair cells in the BHK-2, BEP and BEPHK-2 groups. Thus, HK-2 not only significantly reduced OHC and IHC damage, but completely prevented loss of ANF, SGN and saccule hair cells. The powerful protective effects of this oral MFRM make HK-2 an extremely promising candidate for human clinical trials.


Subject(s)
Blast Injuries , Hair Cells, Vestibular , Spiral Ganglion , Animals , Spiral Ganglion/drug effects , Spiral Ganglion/pathology , Rats , Blast Injuries/prevention & control , Hair Cells, Vestibular/drug effects , Hair Cells, Vestibular/metabolism , Male , Oxidation-Reduction , Rats, Sprague-Dawley , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Oxidative Stress/drug effects , Hearing Loss, Noise-Induced/prevention & control , Hearing Loss, Noise-Induced/pathology
3.
Cell Death Dis ; 15(7): 531, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060244

ABSTRACT

Sensorineural hearing loss (SNHL) is the most common sensory deficit worldwide. Due to the heterogeneity of causes for SNHL, effective treatment options remain scarce, creating an unmet need for novel drugs in the field of otology. Cochlear implantation (CI) currently is the only established method to restore hearing function in profound SNHL and deaf patients. The cochlear implant bypasses the non-functioning sensory hair cells (HCs) and electrically stimulates the neurons of the cochlear nerve. CI also benefits patients with residual hearing by combined electrical and auditory stimulation. However, the insertion of an electrode array into the cochlea induces an inflammatory response, characterized by the expression of pro-inflammatory cytokines, upregulation of reactive oxygen species, and apoptosis and necrosis of HCs, putting residual hearing at risk. Here, we characterize the small molecule AC102, a pyridoindole, for its protective effects on residual hearing in CI. In a gerbil animal model of CI, AC102 significantly improves the recovery of hearing thresholds across multiple frequencies and confines the cochlear trauma to the directly mechanically injured area. In addition, AC102 significantly preserves auditory nerve fibers and inner HC synapses throughout the whole cochlea. In vitro experiments in an ethanol challenged HT22 cell-line revealed significant and dose-responsive anti-apoptotic effects following the treatment of with AC102. Further, AC102 treatment resulted in significant downregulation of the expression of pro-inflammatory cytokines in an organotypic ex vivo model of electrode insertion trauma (EIT). These results suggest that AC102's effects are likely elicited during the inflammatory phase of EIT and mediated by anti-apoptotic and anti-inflammatory properties, highlighting AC102 as a promising compound for hearing preservation during CI. Moreover, since the inflammatory response in CI shares similarities to that in other etiologies of SNHL, AC102 may be inferred as a potential general treatment option for various inner ear conditions.


Subject(s)
Cochlear Implantation , Disease Models, Animal , Gerbillinae , Hearing , Animals , Cochlear Implantation/methods , Hearing/drug effects , Cochlea/drug effects , Cochlea/pathology , Hearing Loss, Sensorineural , Indoles/pharmacology , Indoles/therapeutic use , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism
4.
Int Immunopharmacol ; 138: 112639, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38972209

ABSTRACT

Cisplatin (CDDP) is extensively utilized in the management of diverse types of cancers, but its ototoxicity cannot be ignored, and clinical interventions are not ideal. Histidine decarboxylase (HDC) is the exclusive enzyme for histamine synthesis. Anti-histamine receptor drugs are ubiquitously employed in the therapeutics of allergies and gastrointestinal diseases. Yet, the specific role of histamine and its signaling in the inner ear is not fully understood. This study utilized cisplatin treated mice and HEI-OC1 auditory hair cell line to establish a cisplatin-induced ototoxicity (CIO) model. Histidine decarboxylase knockout (HDC-/-) mice and histamine receptor 1 (H1R) antagonist were utilized to investigate the influence of HDC/histamine/H1R signaling on ototoxicity. The results identified HDC and H1R expression in mouse hair cells. Transcriptomics indicated that the expression levels of oxidative stress-related genes in the cochlea of HDC-/- mice increased. Furthermore, histamine deficiency or suppression of H1R signaling accelerated HC ferroptosis, a pivotal factor underlying the aggravation of CIO in vivo and in vitro, conversely, the supplementation of exogenous histamine reversed these deleterious effects. Mechanistically, this study revealed that the malfunction of HDC/histamine/H1R signaling induced upregulation of NRF2 expression, accompanied by the upregulation of ACSL4 and downregulation of GPX4 expression, which are major regulatory factors of ferroptosis. In summary, histamine deficiency may induce hair cell death by regulating the H1R pathway and exacerbate CIO. Our findings have indicated a potential therapeutic target for CIO.


Subject(s)
Cisplatin , Ferroptosis , Hair Cells, Auditory , Histamine , Histidine Decarboxylase , Mice, Knockout , Signal Transduction , Animals , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Histamine/metabolism , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Hair Cells, Auditory/metabolism , Mice , Ferroptosis/drug effects , Signal Transduction/drug effects , Ototoxicity , Receptors, Histamine H1/metabolism , Receptors, Histamine H1/genetics , Antineoplastic Agents/adverse effects , Mice, Inbred C57BL , Cell Line , Male , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics
5.
Food Chem Toxicol ; 190: 114792, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38849049

ABSTRACT

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.


Subject(s)
Amides , Apoptosis , Cisplatin , Ototoxicity , Pyridines , Zebrafish , Animals , Cisplatin/toxicity , Amides/pharmacology , Pyridines/pharmacology , Ototoxicity/prevention & control , Apoptosis/drug effects , Animals, Genetically Modified , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Larva/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , Disease Models, Animal
6.
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
7.
J Biomed Mater Res B Appl Biomater ; 112(7): e35439, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923766

ABSTRACT

Sensorineural hearing loss (SNHL) is mainly caused by injury or loss of hair cells (HCs) and associated spiral ganglion neurons (SGNs) in the inner ear. At present, there is still no effective treatment for SNHL in clinic. Recently, advances in organoid bring a promising prospect for research and treatment of SNHL. Meanwhile, three-dimensional (3D) printing provides a tremendous opportunity to construct versatile organoids for tissue engineering and regenerative medicine. In this study, gelatin (Gel), sodium alginate (SA), and polyvinyl alcohol (PVA) were used to fabricate biomimetic scaffold through 3D printing. The organ of Corti derived from neonatal mice inner ear was seeded on the PVA/Gel/SA scaffold to construct organ of Corti organoid. Then, the organ of Corti organoid was used to study the potential protective effects of berberine sulfate on neomycin-juried auditory HCs and SGNs. The results showed that the PVA/Gel/SA biomimetic 3D scaffolds had good cytocompatibilities and mechanical properties. The constructed organoid could maintain organ of Corti activity well in vitro. In addition, the injury intervention results showed that berberine sulfate could significantly inhibit neomycin-induced HC and SGN damage. This study suggests that the fabricated organoid is highly biomimetic to the organ of Corti, which may provide an effective model for drug development, cell and gene therapy for SNHL.


Subject(s)
Berberine , Organ of Corti , Tissue Scaffolds , Animals , Organ of Corti/drug effects , Mice , Berberine/pharmacology , Berberine/chemistry , Tissue Scaffolds/chemistry , Organoids/metabolism , Organoids/drug effects , Printing, Three-Dimensional , Alginates/chemistry , Alginates/pharmacology , Gelatin/chemistry , Gelatin/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Tissue Engineering , Polyvinyl Alcohol/chemistry , Polyvinyl Alcohol/pharmacology , Hearing Loss, Sensorineural , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism
8.
Hear Res ; 447: 109013, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718672

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Cimetidine , Cisplatin , Mechanotransduction, Cellular , Organic Cation Transporter 2 , Ototoxicity , Cisplatin/toxicity , Animals , Ototoxicity/prevention & control , Ototoxicity/metabolism , Ototoxicity/physiopathology , Mechanotransduction, Cellular/drug effects , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/antagonists & inhibitors , Cimetidine/pharmacology , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/metabolism , Mice, Inbred C57BL , Mice , Membrane Proteins
9.
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
10.
Environ Toxicol ; 39(7): 4058-4065, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38661261

ABSTRACT

Platinum-based antineoplastic drugs, including cisplatin, carboplatin, and oxaliplatin, are widely used in the treatment of various cancers. Ototoxicity is a common adverse effect of platinum-based drugs. Ototoxicity leads to irreversible hearing impairment. We hypothesize that different platinum-based drugs exhibit varying ototoxic concentrations, time effects, and ototoxic mechanisms. We tested this hypothesis by using a zebrafish model (pvalb3b: TagGFP) to assess the viability of hair cells collected from zebrafish larvae. Cisplatin, carboplatin, and oxaliplatin were administered at dosages of 100, 200, or 400 µM, and the ototoxic effects of these drugs were assessed 1, 2, or 3 h after administration. Fm4-64 and a TUNEL assay were used to label the membranes of living hair cells and to detect cell apoptosis, respectively. We observed that >50% of hair cells were damaged at 1 h after cisplatin (100 µM) exposure, and this ototoxic effect increased at higher dosages and over time. Owing to the smaller ototoxic effects of carboplatin and oxaliplatin, we conducted higher-strength and longer-duration experiments with these drugs. Neither carboplatin nor oxaliplatin was obviously ototoxic, even at 1600 µM and after 6 h. Moreover, only cisplatin damaged the membranes of the hair cells. Cell apoptosis and significantly increased antioxidant gene expression were observed in only the cisplatin group. In conclusion, cisplatin significantly damages sensory hair cells and has notable dosage and time effects. Carboplatin and oxaliplatin are less ototoxic than cisplatin, likely due to having different ototoxic mechanisms than cisplatin.


Subject(s)
Antineoplastic Agents , Apoptosis , Carboplatin , Cisplatin , Ototoxicity , Oxaliplatin , Zebrafish , Animals , Cisplatin/toxicity , Oxaliplatin/toxicity , Carboplatin/toxicity , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Hair Cells, Auditory/drug effects , Larva/drug effects
11.
Toxicol Appl Pharmacol ; 486: 116947, 2024 May.
Article in English | MEDLINE | ID: mdl-38688426

ABSTRACT

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.


Subject(s)
Apoptosis , Cisplatin , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Hair Cells, Auditory , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Cisplatin/toxicity , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Endoplasmic Reticulum Stress/drug effects , Mice , Apoptosis/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Cell Line , Antineoplastic Agents/toxicity , Male , Ototoxicity/prevention & control
12.
J Assoc Res Otolaryngol ; 25(3): 259-275, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38622383

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Cisplatin , Hearing Loss , Organophosphates , Oxazoles , Zebrafish , Animals , Cisplatin/toxicity , Cisplatin/adverse effects , Mice , Hearing Loss/prevention & control , Hearing Loss/chemically induced , Oxazoles/pharmacology , Organophosphates/toxicity , Antineoplastic Agents/toxicity , United States Food and Drug Administration , Drug Approval , Hair Cells, Auditory/drug effects , United States , Ototoxicity/prevention & control , Ototoxicity/etiology , Humans
13.
Arch Toxicol ; 98(6): 1827-1842, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563869

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents , Calcium Channel Blockers , Calcium , Gentamicins , Hair Cells, Auditory , Neomycin , Verapamil , Zebrafish , Animals , Calcium Channel Blockers/pharmacology , Calcium/metabolism , Verapamil/pharmacology , Neomycin/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Gentamicins/toxicity , Anti-Bacterial Agents/toxicity , Reactive Oxygen Species/metabolism , Ototoxicity/prevention & control , Aminoglycosides/toxicity , Lateral Line System/drug effects , Larva/drug effects , Hearing Loss/chemically induced , Hearing Loss/prevention & control
14.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673858

ABSTRACT

Hearing loss represents a multifaceted and pervasive challenge that deeply impacts various aspects of an individual's life, spanning psychological, emotional, social, and economic realms. Understanding the molecular underpinnings that orchestrate hearing loss remains paramount in the quest for effective therapeutic strategies. This review aims to expound upon the physiological, biochemical, and molecular aspects of hearing loss, with a specific focus on its correlation with diabetes. Within this context, phytochemicals have surfaced as prospective contenders in the pursuit of potential adjuvant therapies. These compounds exhibit noteworthy antioxidant and anti-inflammatory properties, which hold the potential to counteract the detrimental effects induced by oxidative stress and inflammation-prominent contributors to hearing impairment. Furthermore, this review offers an up-to-date exploration of the diverse molecular pathways modulated by these compounds. However, the dynamic landscape of their efficacy warrants recognition as an ongoing investigative topic, inherently contingent upon specific experimental models. Ultimately, to ascertain the genuine potential of phytochemicals as agents in hearing loss treatment, a comprehensive grasp of the molecular mechanisms at play, coupled with rigorous clinical investigations, stands as an imperative quest.


Subject(s)
Antioxidants , Hair Cells, Auditory , Hearing Loss, Sensorineural , Oxidative Stress , Phytochemicals , Oxidative Stress/drug effects , Humans , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Hearing Loss, Sensorineural/drug therapy , Hearing Loss, Sensorineural/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Cell Death/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
15.
Otolaryngol Head Neck Surg ; 170(5): 1421-1429, 2024 May.
Article in English | MEDLINE | ID: mdl-38314899

ABSTRACT

OBJECTIVE: Verification that blind and excessive use of antioxidants leads to antioxidant stress which exacerbates cochlear cell damage. STUDY DESIGN: Basic research. SETTING: The Third Affiliated Hospital of Sun Yat-Sen University. METHODS: We compared and quantified hair cell-like house ear institute-organ of corti 1 (HEI-OC1) cell density, cell viability, and apoptosis caused by different concentrations of N-acetylcysteine (NAC) via Hoechst staining, Cell Counting Kit 8, Hoechst with propidium iodide staining, and Annexin V with propidium iodide (PI) staining. Apoptosis induced by high concentrations of M40403 and coenzyme Q10 in cochlear explants was analyzed and compared by cochlear dissection and activated caspase 3 labeling. RESULTS: With the increase of NAC concentration (0-1000 µmol/L), cell density decreased consequently and reached the lowest at 1000 µmol/L (****P ≤ .0001). Cell viability is also declining (**P < .01). The number of Annexin V-fluorescein isothiocyanate-labeled cells and PI-labeled cells increased with increasing NAC concentration after treatment of HEI-OC1 cells for 48 hours. The proportion of apoptotic cells also rose (*P < .05, **P < .01). Cochlear hair cells (HCs) treated with low concentrations of M40403 and coenzyme Q10 for 48 hours showed no damage. When the concentrations of M40403 and coenzyme Q10 were increased (concentrations>30 µmol/L), HC damage began, followed by a dose-dependent increase in HC loss (*P < .001, **P < .0001). Activated caspase-3 was clearly apparent in cochlear explants treated with 50 µmol/L M40403 and coenzyme Q10 compared with cochlear explants without added M40403 and coenzyme Q10. CONCLUSION: These experimental results suggest that inappropriate application of antioxidants can cause severe damage to normal cochlear HCs.


Subject(s)
Acetylcysteine , Antioxidants , Apoptosis , Cell Survival , Oligopeptides , Oxidative Stress , Ubiquinone , Ubiquinone/analogs & derivatives , Antioxidants/pharmacology , Acetylcysteine/pharmacology , Ubiquinone/pharmacology , Ubiquinone/therapeutic use , Cell Survival/drug effects , Animals , Apoptosis/drug effects , Oxidative Stress/drug effects , Mice , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Cell Count
16.
Otol Neurotol ; 45(1): e49-e56, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38085767

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents , Gentamicins , Hair Cells, Auditory , Hydrocortisone , Ototoxicity , Animals , Humans , Aminoglycosides , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Apoptosis , Gentamicins/adverse effects , Gentamicins/toxicity , Hair Cells, Auditory/drug effects , Hydrocortisone/pharmacology , Mammals/metabolism , Ototoxicity/etiology , Ototoxicity/metabolism , Protein Synthesis Inhibitors , Reactive Oxygen Species/metabolism
17.
J Mol Med (Berl) ; 100(5): 797-813, 2022 05.
Article in English | MEDLINE | ID: mdl-35471608

ABSTRACT

Aminoglycoside antibiotics are lifesaving medicines, crucial for the treatment of chronic or drug resistant infections. However, aminoglycosides are toxic to the sensory hair cells in the inner ear. As a result, aminoglycoside-treated individuals can develop permanent hearing loss and vestibular impairment. There is considerable evidence that reactive oxygen species (ROS) production and the subsequent phosphorylation of c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (P38) drives apoptosis in aminoglycoside-treated hair cells. However, treatment strategies that directly inhibit ROS, JNK, or P38 are limited by the importance of these molecules for normal cellular function. Alternatively, the upstream regulator apoptosis signal-regulating kinase 1 (ASK1/MAP3K5) is a key mediator of ROS-induced JNK and P38 activation under pathologic but not homeostatic conditions. We investigated ASK1 as a mediator of drug-induced hair cell death using cochlear explants from Ask1 knockout mice, demonstrating that Ask1 deficiency attenuates neomycin-induced hair cell death. We then evaluated pharmacological inhibition of ASK1 with GS-444217 as a potential otoprotective therapy. GS-444217 significantly attenuated hair cell death in neomycin-treated explants but did not impact aminoglycoside efficacy against P. aeruginosa in the broth dilution test. Overall, we provide significant pre-clinical evidence that ASK1 inhibition represents a novel strategy for preventing aminoglycoside ototoxicity. KEY MESSAGES: ASK1 is an upstream, redox-sensitive regulator of P38 and JNK, which are known mediators of hair cell death. Ask1 knockout does not affect hair cell development in vivo, but significantly reduces aminoglycoside-induced hair cell death in vitro. A small-molecule inhibitor of ASK1 attenuates neomycin-induced hair cell death, and does not impact antibiotic efficacy in vitro. ASK1 may be a novel molecular target for preventing aminoglycoside-induced hearing loss.


Subject(s)
Aminoglycosides , Hair Cells, Auditory , Hearing Loss , MAP Kinase Kinase Kinase 5 , Aminoglycosides/adverse effects , Animals , Anti-Bacterial Agents/adverse effects , Apoptosis/drug effects , Cell Death/drug effects , 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 , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Mice , Neomycin/adverse effects , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35197290

ABSTRACT

Aminoglycosides (AGs) are commonly used antibiotics that cause deafness through the irreversible loss of cochlear sensory hair cells (HCs). How AGs enter the cochlea and then target HCs remains unresolved. Here, we performed time-lapse multicellular imaging of cochlea in live adult hearing mice via a chemo-mechanical cochleostomy. The in vivo tracking revealed that systemically administered Texas Red-labeled gentamicin (GTTR) enters the cochlea via the stria vascularis and then HCs selectively. GTTR uptake into HCs was completely abolished in transmembrane channel-like protein 1 (TMC1) knockout mice, indicating mechanotransducer channel-dependent AG uptake. Blockage of megalin, the candidate AG transporter in the stria vascularis, by binding competitor cilastatin prevented GTTR accumulation in HCs. Furthermore, cilastatin treatment markedly reduced AG-induced HC degeneration and hearing loss in vivo. Together, our in vivo real-time tracking of megalin-dependent AG transport across the blood-labyrinth barrier identifies new therapeutic targets for preventing AG-induced ototoxicity.


Subject(s)
Anti-Bacterial Agents/metabolism , Gentamicins/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Animals , Anti-Bacterial Agents/toxicity , Biological Transport , Cilastatin/pharmacology , Endolymph/metabolism , Gentamicins/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hearing/drug effects , Low Density Lipoprotein Receptor-Related Protein-2/antagonists & inhibitors , Mice , Stria Vascularis/metabolism
19.
Cell Mol Life Sci ; 79(2): 79, 2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35044530

ABSTRACT

The Hippo/Yes-associated protein (YAP) signaling pathway has been shown to be able to maintain organ size and homeostasis by regulating cell proliferation, differentiation, and apoptosis. The abuse of aminoglycosides is one of the main causes of sensorineural hearing loss (SSNHL). However, the role of the Hippo/YAP signaling pathway in cochlear hair cell (HC) damage protection in the auditory field is still unclear. In this study, we used the YAP agonist XMU-MP-1 (XMU) and the inhibitor Verteporfin (VP) to regulate the Hippo/YAP signaling pathway in vitro. We showed that YAP overexpression reduced neomycin-induced HC loss, while downregulated YAP expression increased HC vulnerability after neomycin exposure in vitro. We next found that activation of YAP expression inhibited C-Abl-mediated cell apoptosis, which led to reduced HC loss. Many previous studies have reported that the level of reactive oxygen species (ROS) is significantly increased in cochlear HCs after neomycin exposure. In our study, we also found that YAP overexpression significantly decreased ROS accumulation, while downregulation of YAP expression increased ROS accumulation. In summary, our results demonstrate that the Hippo/YAP signaling pathway plays an important role in reducing HC injury and maintaining auditory function after aminoglycoside exposure. YAP overexpression could protect against neomycin-induced HC loss by inhibiting C-Abl-mediated cell apoptosis and decreasing ROS accumulation, suggesting that YAP could be a novel therapeutic target for aminoglycosides-induced sensorineural hearing loss in the clinic.


Subject(s)
Anti-Bacterial Agents/adverse effects , Hair Cells, Auditory/drug effects , Hippo Signaling Pathway/drug effects , Neomycin/adverse effects , YAP-Signaling Proteins/metabolism , Animals , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Mice , Protective Factors , Protein Synthesis Inhibitors/adverse effects , Signal Transduction/drug effects
20.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119204, 2022 04.
Article in English | MEDLINE | ID: mdl-35026350

ABSTRACT

Cisplatin is a platinum-containing drug with ototoxicity commonly used clinically and has significant efficacy against a variety of solid tumors. One of the most important mechanisms of ototoxicity is that cisplatin induces apoptosis of hair cells. According to relevant literature, X-linked inhibitor of apoptosis protein (XIAP, anti-apoptotic protein) could inhibit the apoptotic pathway. We hypothesized that this protein might protect cochlear hair cells from cisplatin-induced injury. To figure it out, we treated cochlea of normal mice with various concentrations of cisplatin to observe the response and morphology of hair cells and determine a reasonable concentration. Next, Western Blot and quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) experiments were conducted to make an investigation about the expression of XIAP protein and mRNA. In addition, we constructed and identified XIAP overexpressing mice. Finally, we treated cochlear tissues of normal and overexpressing mice with cisplatin to investigate the cyto-protection of XIAP on hair cells, respectively. It was found that 50 µmol/L cisplatin resulted in significant loss and disorganization of hair cells, while simultaneously downregulating the protein and mRNA of XIAP. In XIAP overexpressing mice, the loss and disorganization of hair cells were significantly lessened. These results showed that XIAP can lessen cisplatin-induced hair cell loss and play a role in otoprotection.


Subject(s)
Cisplatin/pharmacology , Hair Cells, Auditory/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL