Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
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 , Mice, Inbred C57BL , Ototoxicity , Signal Transduction , Spiral Ganglion , Transforming Growth Factor beta , Animals , Kanamycin/toxicity , Signal Transduction/drug effects , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Transforming Growth Factor beta/metabolism , Mice , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Cochlea/metabolism , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Furosemide/pharmacology , Male
2.
Otol Neurotol ; 45(5): 495-501, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38561601

ABSTRACT

HYPOTHESIS: Cyclodextrin (CDX)-induced serum prestin burst is not dependent on outer hair cell (OHC) loss. BACKGROUND: Serum prestin has been proposed as a biomarker for ototoxicity. We recently used an automated Western approach to quantify serum prestin changes in a newly introduced model of CDX ototoxicity. To gain insights into prestin as a biomarker, here we further characterize serum prestin in the CDX model. METHODS: Guinea pigs were treated with 750, 3,000, or 4,000 mg/kg CDX, and serum samples were obtained through up to 15 weeks after exposure. Serum prestin levels were quantified using automated Western, and hair cell counts were obtained. RESULTS: All three doses induced an N -glycosylated ~134-kDa prestin burst; however, only the 3,000 and 4,000 mg/kg resulted in robust OHC loss. Prestin levels returned to baseline where they remained up to 15 weeks in the absence of OHCs. CONCLUSION: The ~134-kDa prestin burst induced after CDX administration is N -glycosylated, representing a posttranslational modification of prestin. Serum prestin seems to be a promising biomarker when using therapeutics with ototoxic properties because it is not dependent on OHC loss as a necessary event, thus affording the opportunity for early detection and intervention.


Subject(s)
Hair Cells, Auditory, Outer , Animals , Guinea Pigs , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Biomarkers/blood , Biomarkers/metabolism , Ototoxicity/etiology , Sulfate Transporters/metabolism
3.
Sci Rep ; 14(1): 7889, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570541

ABSTRACT

Nobiletin, a citrus polymethoxy flavonoid with antiapoptotic and antioxidative properties, could safeguard against cisplatin-induced nephrotoxicity and neurotoxicity. Cisplatin, as the pioneer of anti-cancer drug, the severe ototoxicity limits its clinical applications, while the effect of nobiletin on cisplatin-induced ototoxicity has not been identified. The current study investigated the alleviating effect of nobiletin on cisplatin-induced ototoxicity and the underlying mechanisms. Apoptosis and ROS formation were evaluated using the CCK-8 assay, Western blotting, and immunofluorescence, indicating that nobiletin attenuated cisplatin-induced apoptosis and oxidative stress. LC3B and SQSTM1/p62 were determined by Western blotting, qPCR, and immunofluorescence, indicating that nobiletin significantly activated autophagy. Nobiletin promoted the nuclear translocation of NRF2 and the transcription of its target genes, including Hmox1, Nqo1, and ferroptosis markers (Gpx4, Slc7a11, Fth, and Ftl), thereby inhibiting ferroptosis. Furthermore, RNA sequencing analysis verified that autophagy, ferroptosis, and the NRF2 signaling pathway served as crucial points for the protection of nobiletin against ototoxicity caused by cisplatin. Collectively, these results indicated, for the first time, that nobiletin alleviated cisplatin-elicited ototoxicity through suppressing apoptosis and oxidative stress, which were attributed to the activation of autophagy and the inhibition of NRF2/GPX4-mediated ferroptosis. Our study suggested that nobiletin could be a prospective agent for preventing cisplatin-induced hearing loss.


Subject(s)
Ferroptosis , Flavones , Ototoxicity , Humans , Cisplatin/toxicity , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Ototoxicity/drug therapy , Ototoxicity/etiology , Prospective Studies , Phospholipid Hydroperoxide Glutathione Peroxidase/pharmacology , Autophagy
4.
Eur J Pediatr ; 183(6): 2625-2636, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492032

ABSTRACT

Neonates face heightened susceptibility to drug toxicity, often exposed to off-label medications with dosages extrapolated from adult or pediatric studies. Premature infants in Neonatal Intensive Care Units (NICUs) are particularly at risk due to underdeveloped pharmacokinetics and exposure to multiple drugs. The study aimed to survey commonly used medications with a higher risk of ototoxicity and nephrotoxicity in Spanish and Italian neonatal units. A prospective cross-sectional study was conducted in Italian and Spanish neonatal units using a web-based survey with 43 questions. A modified Delphi method involved experts refining the survey through online consensus. Ethical approval was obtained, and responses were collected from January to July 2023. The survey covered various aspects, including drug-related ototoxic and nephrotoxic management, hearing screening, and therapeutic drug monitoring. Responses from 131 participants (35.9% from Spain and 64.1% from Italy) revealed awareness of drug toxicity risks. Varied practices were observed in hearing screening protocols, and a high prevalence of ototoxic and nephrotoxic drug use, including aminoglycosides (100%), vancomycin (70.2%), loop diuretics (63.4%), and ibuprofen (62.6%). Discrepancies existed in guideline availability and adherence, with differences between Italy and Spain in therapeutic drug monitoring practices. CONCLUSIONS: The study underscores the need for clinical guidelines and uniform practices in managing ototoxic and nephrotoxic drugs in neonatal units. Awareness is high, but inconsistencies in practices indicate a necessity for standardization, including the implementation of therapeutic drug monitoring and the involvement of clinical pharmacologists. Addressing these issues is crucial for optimizing neonatal care in Southern Europe. WHAT IS KNOWN: • Neonates in intensive care face a high risk of nephrotoxicity and ototoxicity from drugs like aminoglycosides, vancomycin, loop diuretics, and ibuprofen. • Therapeutic drug monitoring is key for managing these risks, optimizing dosing for efficacy and minimizing side effects. WHAT IS NEW: • NICUs in Spain and Italy show high drug toxicity awareness but differ in ototoxic/nephrotoxic drug management. • Urgent need for standard guidelines and practices to address nephrotoxic risks from aminoglycosides, vancomycin, loop diuretics, and ibuprofen.


Subject(s)
Aminoglycosides , Intensive Care Units, Neonatal , Ototoxicity , Vancomycin , Humans , Italy , Infant, Newborn , Intensive Care Units, Neonatal/statistics & numerical data , Cross-Sectional Studies , Prospective Studies , Spain , Aminoglycosides/adverse effects , Ototoxicity/etiology , Vancomycin/adverse effects , Drug Monitoring/methods , Drug Monitoring/statistics & numerical data , Ibuprofen/adverse effects , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Surveys and Questionnaires , Female , Kidney Diseases/chemically induced , Kidney Diseases/epidemiology , Infant, Premature , Male
6.
Hear Res ; 445: 108995, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38518393

ABSTRACT

OBJECTIVE: The genotype-phenotype relationship in cisplatin-induced ototoxicity remains unclear. By assessing early shifts in distortion product otoacoustic emission (DPOAE) levels after initial cisplatin administration, we aimed to discriminate patients' susceptibility to cisplatin-induced ototoxicity and elucidate their genetic background. STUDY DESIGN: A prospective cross-sectional study. SETTING: Tertiary referral hospital in Japan. PATIENTS: Twenty-six patients with head and neck cancer were undergoing chemoradiotherapy with three cycles of 100 mg/m2 cisplatin. INTERVENTIONS: Repetitive pure-tone audiometry and DPOAE measurements, and blood sampling for DNA extraction were performed. Patients were grouped into early ototoxicity presence or absence based on whether DPOAE level shifts exceeded the corresponding reference limits of the 21-day test interval. MAIN OUTCOME MEASURES: Hearing thresholds after each cisplatin cycle, severity of other adverse events, and polymorphisms in cisplatin-induced ototoxicity-associated genes were compared. RESULTS: Early ototoxicity was present in 14 and absent in 12 patients. Ototoxicity presence on DPOAEs was associated with greater progression of hearing loss in frequencies ≥2 kHz throughout therapy and with higher ototoxicity grades compared with ototoxicity absence. Ototoxicity was further associated with grade ≥2 nausea. Ototoxicity presence was genetically associated with the GSTT1 null genotype and G-allele of NFE2L2 rs6721961, whereas ototoxicity absence was associated with the GSTM1 null genotype. Dose-dependent progression of hearing loss was the greatest in the combined genotype pattern of GSTT1 null and the T/G or G/G variants of rs6721961. CONCLUSION: Early DPOAE changes reflected genetic vulnerability to cisplatin-induced ototoxicity. Hereditary insufficiency of the antioxidant defense system causes severe cisplatin-induced hearing loss and nausea.


Subject(s)
Cisplatin , Hearing Loss , NF-E2-Related Factor 2 , Ototoxicity , Humans , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cross-Sectional Studies , Deafness/chemically induced , Hearing Loss/chemically induced , Hearing Loss/diagnosis , Hearing Loss/genetics , Nausea/chemically induced , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/pharmacology , Otoacoustic Emissions, Spontaneous , Ototoxicity/etiology , Ototoxicity/genetics , Polymorphism, Genetic , Prospective Studies
7.
Mol Ther ; 32(5): 1387-1406, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38414247

ABSTRACT

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.


Subject(s)
Cisplatin , Ferroptosis , Hearing Loss , Mice, Inbred C57BL , Mice, Knockout , Phospholipid Hydroperoxide Glutathione Peroxidase , Animals , Cisplatin/adverse effects , Ferroptosis/drug effects , Ferroptosis/genetics , Mice , Hearing Loss/chemically induced , Hearing Loss/genetics , Hearing Loss/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Disease Models, Animal , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Reactive Oxygen Species/metabolism , Lipid Peroxidation/drug effects , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Ototoxicity/etiology , Ototoxicity/metabolism , Antineoplastic Agents/adverse effects , Apoptosis/drug effects
8.
Therapie ; 79(2): 283-295, 2024.
Article in English | MEDLINE | ID: mdl-37957052

ABSTRACT

Sensorineural hearing loss (SNHL) is the most common type of hearing loss. Causes include degenerative changes in the sensory hair cells, their synapses and/or the cochlear nerve. As human inner ear hair cells have no capacity for regeneration, their destruction is irreversible and leads to permanent hearing loss. SNHL can be genetically inherited or acquired through ageing, exposure to noise or ototoxic drugs. Ototoxicity generally refers to damage to the structures and functions of the inner ear following exposure to specific drugs. Ototoxicity can be multifactorial, causing damage to cochlear hair cells or cells with homeostatic functions that modulate cochlear hair cell function. Clinical strategies to limit ototoxicity include identifying patients at risk, monitoring drug concentrations, performing serial hearing assessments and switching to less ototoxic therapy. This review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, using the PubMed® database. The search terms "ototoxicity", "hearing loss" and "drugs" were combined. We included studies published between September 2013 and June 2023, and focused on medicines and drugs used in hospitals. The review highlighted a number of articles reporting the main drug classes potentially involved: namely, immunosuppressants, antimalarials, vaccines, antibiotics, antineoplastic agents, diuretics, nonsteroidal anti-inflammatory drugs and analgesics. The presumed ototoxic mechanisms were described, together with the therapeutic and preventive options developed over the last ten years.


Subject(s)
Hearing Loss , Ototoxicity , Humans , Cochlea/physiology , Ototoxicity/etiology , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Anti-Bacterial Agents/adverse effects
9.
Ear Hear ; 45(2): 329-336, 2024.
Article in English | MEDLINE | ID: mdl-37700446

ABSTRACT

OBJECTIVES: To evaluate the extent of hearing loss among pottery workers in Mexico exposed to lead. DESIGN: The authors conducted a cross-sectional study including 315 adult pottery workers. Auditory function was evaluated by air conduction pure-tone audiometry (pure-tone average) and distortion-product otoacoustic emission (DPOAE) levels (amplitude and signal-to-noise ratio). Lead exposure was assessed with a single blood sample test and classified as low, medium, and high according to blood lead tertiles. Logistic regression models were calculated for the association between blood lead levels, pure-tone average, and DPOAE records. RESULTS: Median (25th-75th) blood lead levels were 14 µg/dL (7.5-22.6 µg/dL). The audiometric pattern and DPOAE records were similar across blood lead levels groups in all frequencies, and no statistically significant differences were found. Adjusted logistic regression models showed no increase in the odds for hearing thresholds >25 dB (HL) and DPOAE absence associated with blood lead levels, and no dose-response pattern was observed ( p > 0.05). CONCLUSIONS: Given the results from this cross-sectional study, no association was found between blood lead levels and hearing loss assessed with DPOAE. Future longitudinal work should consider chronic lead exposure estimates among underrepresented populations, which can potentially inform safer work practices to minimize the risk of ototoxicity.


Subject(s)
Deafness , Hearing Loss , Ototoxicity , Adult , Humans , Lead , Ototoxicity/etiology , Cross-Sectional Studies , Auditory Threshold/physiology , Otoacoustic Emissions, Spontaneous/physiology , Hearing Loss/chemically induced , Audiometry, Pure-Tone/methods
10.
J Appl Toxicol ; 44(2): 235-244, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37650462

ABSTRACT

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.


Subject(s)
Gentamicins , Ototoxicity , Humans , Gentamicins/metabolism , Reactive Oxygen Species/metabolism , PQQ Cofactor/pharmacology , PQQ Cofactor/therapeutic use , PQQ Cofactor/metabolism , Ototoxicity/etiology , Ototoxicity/prevention & control , Ototoxicity/metabolism , Hair Cells, Auditory/metabolism , Anti-Bacterial Agents/metabolism , Apoptosis
11.
Clin Otolaryngol ; 49(1): 1-15, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37818931

ABSTRACT

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


Subject(s)
Antineoplastic Agents , Hearing Loss , Neoplasms , Ototoxicity , Adult , Humans , Cisplatin/therapeutic use , Antineoplastic Agents/therapeutic use , Carboplatin/adverse effects , Ototoxicity/etiology , Ototoxicity/prevention & control , Ototoxicity/drug therapy , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Hearing Loss/drug therapy , Neoplasms/drug therapy , Neoplasms/chemically induced , Adrenal Cortex Hormones/therapeutic use , Randomized Controlled Trials as Topic
12.
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
13.
Int J Mol Sci ; 24(22)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38003734

ABSTRACT

Cisplatin is a commonly used chemotherapeutic agent with proven efficacy in treating various malignancies, including testicular, ovarian, cervical, breast, bladder, head and neck, and lung cancer. Cisplatin is also used to treat tumors in children, such as neuroblastoma, osteosarcoma, and hepatoblastoma. However, its clinical use is limited by severe side effects, including ototoxicity, nephrotoxicity, neurotoxicity, hepatotoxicity, gastrointestinal toxicity, and retinal toxicity. Cisplatin-induced ototoxicity manifests as irreversible, bilateral, high-frequency sensorineural hearing loss in 40-60% of adults and in up to 60% of children. Hearing loss can lead to social isolation, depression, and cognitive decline in adults, and speech and language developmental delays in children. Cisplatin causes hair cell death by forming DNA adducts, mitochondrial dysfunction, oxidative stress, and inflammation, culminating in programmed cell death by apoptosis, necroptosis, pyroptosis, or ferroptosis. Contemporary medical interventions for cisplatin ototoxicity are limited to prosthetic devices, such as hearing aids, but these have significant limitations because the cochlea remains damaged. Recently, the U.S. Food and Drug Administration (FDA) approved the first therapy, sodium thiosulfate, to prevent cisplatin-induced hearing loss in pediatric patients with localized, non-metastatic solid tumors. Other pharmacological treatments for cisplatin ototoxicity are in various stages of preclinical and clinical development. This narrative review aims to highlight the molecular mechanisms involved in cisplatin-induced ototoxicity, focusing on cochlear inflammation, and shed light on potential antioxidant and anti-inflammatory therapeutic interventions to prevent or mitigate the ototoxic effects of cisplatin. We conducted a comprehensive literature search (Google Scholar, PubMed) focusing on publications in the last five years.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Deafness , Hearing Loss , Osteosarcoma , Ototoxicity , Humans , Child , Cisplatin/adverse effects , Antineoplastic Agents/adverse effects , Ototoxicity/etiology , Ototoxicity/drug therapy , Hearing Loss/drug therapy , Osteosarcoma/drug therapy , Bone Neoplasms/drug therapy , Inflammation/drug therapy
14.
Cancer ; 129(24): 3952-3961, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37715631

ABSTRACT

BACKGROUND: High-dose carboplatin is an essential part of curative high-dose chemotherapy (HDCT) for patients with previously treated germ cell tumors (GCTs). Although hearing loss (HL) is a known side effect of HDCT, data on its severity and characteristics are limited. METHODS: Eligible patients received HDCT for GCTs from 1993 to 2017 and had audiograms before and after HDCT. HL severity was classified by American Speech-Language-Hearing Association criteria, and mean change in hearing threshold at each frequency (0.25-8 kHz) was estimated from pre- to post-HDCT and between HDCT cycles. RESULTS: Of 115 patients (median age, 32 years), 102 (89%) received three cycles of HDCT. Of 106 patients with normal hearing to mild HL in the speech frequencies (0.5-4 kHz) before HDCT, 70 (66%) developed moderate to profound HL in the speech frequencies after HDCT. Twenty-five patients (22%) were recommended for hearing aids after HDCT. Patients with moderate to profound HL isolated to the higher frequencies (6-8 kHz) before HDCT were more likely to develop moderate to profound HL in the speech frequencies after HDCT (94% vs. 61%; p = .01) and to be recommended for hearing aids (39% vs. 18%; p = .05). CONCLUSIONS: HL was frequent after HDCT for GCTs, with most patients developing at least moderate HL in the speech frequencies and approximately one in five recommended for hearing aids. Moderate to profound HL isolated to high frequencies at baseline was predictive of more clinically significant hearing impairment after HDCT. PLAIN LANGUAGE SUMMARY: Some patients with germ cell tumors, the most common malignancy in adolescent and young adult men, are not cured with standard-dose chemotherapy and require high-dose chemotherapy (HDCT). Using detailed hearing assessments of patients receiving HDCT, we found that most patients developed significant hearing loss and that one in five needed hearing aids. Thus, strategies to reduce this side effect are urgently needed, and all patients receiving HDCT should have a hearing test after therapy.


Subject(s)
Hearing Loss , Neoplasms, Germ Cell and Embryonal , Ototoxicity , Male , Adolescent , Young Adult , Humans , Adult , Carboplatin/adverse effects , Ototoxicity/etiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/etiology , Hearing Loss/chemically induced , Hearing Loss/epidemiology
15.
Arch Toxicol ; 97(11): 2955-2967, 2023 11.
Article in English | MEDLINE | ID: mdl-37608195

ABSTRACT

The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (HMGCR) gene encodes rate-limiting enzyme in cholesterol biosynthesis, which is related to cell proliferation and mitochondrial function. The present study was designed to explore the expression of HMGCR in murine cochlear hair cells and HEI-OC1 cells and the possible mechanisms underpinning the actions of HMGCR in cisplatin-induced ototoxicity, with special attention given to p38 mitogen-activated protein kinase (MAPK) activities in vitro. The expressions of HMGCR, p-p38, cleaved caspase-3 and LC3B was measured by immunofluorescence and western blot. JC-1 staining and MitoSOX Red were used to detect mitochondria membrane potential (MMP) and reactive oxygen species (ROS) levels respectively. The apoptosis of auditory cells was assessed by TUNEL staining and flow cytometry. Protein levels of bcl2/bax and beclin1 were examined by western blot. We found that HMGCR was widely expressed in the auditory cells, of both neonatal mice and 2-month-old mice, in cytoplasm, nucleus and stereocilia. Moreover, 30 µM cisplatin elicited the formation of ROS, which, in turn, led to HMGCR reduction, activating p38 kinase-related apoptosis and autophagy in auditory cells. Meanwhile, co-treatment with ROS scavenger at a concentration of 2 mM, N-acetyl-L-cysteine (NAC), could alleviate the aforementioned changes. In addition, HMGCR silencing resulted in higher p38 MAPK-mediated apoptosis and autophagy under cisplatin injury. Taken together, we demonstrate that, for the first time, that HMGCR is expressed in the cochlear. Furthermore, HMGCR exerts protective benefit on auditory cells against cisplatin-mediated injury stimulated by ROS, culminating in regulation of p38 MAPK-dependent apoptosis and autophagy.


Subject(s)
Ototoxicity , p38 Mitogen-Activated Protein Kinases , Animals , Mice , Cisplatin/toxicity , Ototoxicity/etiology , Ototoxicity/prevention & control , Reactive Oxygen Species , Signal Transduction , Hair Cells, Auditory
16.
Arch Toxicol ; 97(9): 2477-2493, 2023 09.
Article in English | MEDLINE | ID: mdl-37395757

ABSTRACT

The ototoxic side effect of cisplatin is a main cause of sensorineural hearing loss. This side effect limits the clinical application of cisplatin and affects patients' quality of life. This study was designed to investigate the effect of apelin-13 on cisplatin-induced C57BL/6 mice hearing loss model and explore the potential underlying molecular mechanisms. Mice were intraperitoneally injected with 100 µg/kg apelin-13 2 h before 3 mg/kg cisplatin injection for 7 consecutive days. Cochlear explants cultured in vitro were pretreated with 10 nM apelin-13 2 h prior to 30 µM cisplatin treatment for another 24 h. Hearing test and morphology results showed that apelin-13 attenuated cisplatin-induced mice hearing loss and protected cochlear hair cells and spiral ganglion neurons from damage. In vivo and in vitro experimental results showed that apelin-3 reduced cisplatin-induced apoptosis of hair cells and spiral ganglion neurons. In addition, apelin-3 preserved mitochondrial membrane potential and inhibited ROS production in cultured cochlear explants. Mechanistic studies showed that apelin-3 decreased cisplatin-induced cleaved caspase 3 expression but increased Bcl-2; inhibited the expression of pro-inflammatory factors TNF-a and IL-6; and increased STAT1 phosphorylation but decreased STAT3 phosphorylation. In conclusion, our results indicate that apelin-13 could be a potential otoprotective agent to prevent cisplatin-induced ototoxicity by inhibiting apoptosis, ROS production, TNF-α and IL-6 expression, and regulating phosphorylation of STAT1 and STAT3 transcription factors.


Subject(s)
Antineoplastic Agents , Hearing Loss , Ototoxicity , Mice , Animals , Cisplatin/toxicity , Antineoplastic Agents/toxicity , Apelin/toxicity , Ototoxicity/etiology , Ototoxicity/prevention & control , Reactive Oxygen Species/metabolism , Interleukin-6 , Quality of Life , Mice, Inbred C57BL , Hearing Loss/chemically induced , Apoptosis
17.
Otol Neurotol ; 44(7): e463-e470, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37317507

ABSTRACT

HYPOTHESIS: To examine the protective effects of infliximab (INF) against kanamycin (KM)-induced hearing loss. BACKGROUND: Tumor necrosis factor α blockers can reduce cellular inflammatory reactions and decrease cell death. METHODS: Thirty-six rats with normal hearing were randomly divided into six groups. The first group was injected with 400 mg/kg KM intramuscularly (IM), the second group with 7 mg/kg INF intraperitoneally (IP) and 400 mg/kg KM IM, the third group with 7 mg/kg INF IP and 200 mg/kg KM IM, and the fourth group with 1 mg/kg 6-methylprednisolone (MP) IP and 400 mg/kg KM IM. Group 5 was injected with 1 mg/kg MP IP and 200 mg/kg KM IM, and group 6 with saline IP once. Auditory brain-stem response (ABR) for hearing thresholds was performed on days 7 and 14. From the frozen sections of the cochlea, the area of the stria vascularis, the number of neurons in the spiral ganglion, the fluorescence intensity of hair cells (FIHC), postsynaptic density (PSD), and presynaptic ribbons (PSRs) were calculated. RESULTS: The KM-induced increase in hearing thresholds was detected on the 14th day. Hearing was only preserved in the group treated with INF after low-dose KM exposure but not in the groups that received high-dose KM. The FIHC, excitatory PSD, and PSR were preserved only in the INF-treated group after half-dose KM exposure. In MP groups, FIHC, excitatory PSD, and PSR were significantly lower than in the control group. CONCLUSIONS: Our results support that tumor necrosis factor-based inflammation may play a role in the ototoxicity mechanism.


Subject(s)
Kanamycin , Ototoxicity , Rats , Animals , Kanamycin/toxicity , Infliximab/pharmacology , Infliximab/therapeutic use , Ototoxicity/etiology , Ototoxicity/prevention & control , Cochlea/pathology , Stria Vascularis/pathology , Evoked Potentials, Auditory, Brain Stem
18.
Audiol Neurootol ; 28(6): 427-435, 2023.
Article in English | MEDLINE | ID: mdl-37379818

ABSTRACT

INTRODUCTION: Tetramethylpyrazine (TMP) is a chemical compound, which has been shown to possess numerous biological features such as anticoagulation, inhibition of platelet aggregation, anti-inflammation, capillary dilatation, improvement in microcirculation, and protection against reactive oxygen radicals. The aim of the present study was to investigate the protective effect of TMP against radiation-induced ototoxicity. MATERIALS AND METHODS: 40 rats were divided into four groups. The first group was irradiated for 5 days. The second group received a single dose of 140 mg/kg/day intraperitoneal TMP given to the rats 30 min before radiotherapy (RT) for 5 days. The third group received a single dose of 140 mg/kg/day i.p. TMP for 5 days, whereas the fourth group was administered saline. All rats underwent distortion product otoacoustic emission (DPOAE) and auditory brainstem response measurements before and after the application. The temporal bulla of animals was removed for immunohistopathological examination. RESULTS: Signal-noise ratio values were significantly decreased in the RT group for the frequencies of 2-32 kHz after RT (p < 0.05), whereas the difference was not significant in terms of pre- and posttreatment values for the other groups. Also in the RT group, the ABR thresholds were significantly increased after treatment. In H&E staining, the mean scores for outer hair cells (OHCs), stria vascularis (SV), and spiral ganglion (SG) injuries were significantly higher in RT and RT + TMP groups than in the other groups. The mean OHCs and SV injury scores were also significantly higher in the RT group than in the RT + TMP group (p < 0.05). The number of cochleas that showed cytoplasmic caspase-3 immunoreactivity in the OHC, SV, and SG was significantly higher in RT and RT + TMP groups than in the other groups. CONCLUSION: The findings of the present study suggest that TMP may have a therapeutic potential for preventing sensorineural hearing loss (SNHL) related to RT.


Subject(s)
Cisplatin , Ototoxicity , Pyrazines , Rats , Animals , Ototoxicity/etiology , Ototoxicity/prevention & control , Cochlea , Otoacoustic Emissions, Spontaneous , Evoked Potentials, Auditory, Brain Stem
19.
Int J Mycobacteriol ; 12(2): 168-174, 2023.
Article in English | MEDLINE | ID: mdl-37338479

ABSTRACT

Background: Recognized in 1994 as a global emergency by the World Health Organization, tuberculosis (TB) remains an ongoing health threat. In Cameroon, the mortality rate is estimated at 2.9%. Treatment of multidrug-resistant TB (MDR-TB) defined as the resistance to the two most effective antiTB drugs, and requires therapy of more than 7 drugs taken on a daily basis during 9-12 months. This study aimed to evaluate the safety profile of treatment regimens used for MDR-TB at Jamot Hospital of Yaounde (JHY). Methods: This was a retrospective cohort study of patients treated for MDR-TB at HJY from January 1, 2017, to December 31, 2019. Patients characteristics of the cohort, drugs regimen were collected and described. All possible adverse drug reactions (ADR) were described clinically and by severity grade. Results: During the study period, 107 patients were included, and 96 (89.7%) experienced at least one ADR. Most parts of the patients (90) experienced mild or moderate ADR. Hearing loss was the most frequent ADR, and led mostly in aminoglycosides dose reduction (n = 30, 96.7%). Gastrointestinal events were commonly observed during the study period. Conclusion: Our findings suggested that ototoxicity was a prominent safety issue during the study period. The implementation of the new short treatment regimen could be effective in reducing the burden of ototoxicity among MDR-TB patients. Nevertheless, new safety issues could emerge.


Subject(s)
Antitubercular Agents , Tuberculosis, Multidrug-Resistant , Humans , Antitubercular Agents/administration & dosage , Antitubercular Agents/adverse effects , Cameroon , Ototoxicity/etiology , Ototoxicity/prevention & control , Retrospective Studies , Tuberculosis, Multidrug-Resistant/drug therapy , Male , Female , Adult , Middle Aged , Cohort Studies , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Time Factors
20.
Ann Otol Rhinol Laryngol ; 132(12): 1686-1689, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37243327

ABSTRACT

INTRODUCTION: Aminoglycoside antibiotics such as gentamicin are bactericidal and effective against gram negative organisms and act synergistically against gram positive organisms, including Staphylococcus aureus. However, they have serious adverse effects such as nephrotoxicity and ototoxicity. Gentamicin ototoxicity may occur after a single dose and results in decreased vestibular function, which is frequently debilitating and often permanent. OBJECTIVE: To emphasize the risk of gentamicin ototoxicity and suggest alternative antibiotics in penicillin-allergic patients undergoing surgery. CASE SUMMARY: We present a case of a woman with preexisting Meniere's Disease who received gentamicin 400 mg perioperatively for a sigmoidectomy due to a penicillin allergy listed in the patient's medical record. The patient developed severe ototoxicity preventing her from working or driving. Physical examination was remarkable for a broad-based gait requiring assistance to walk and bilateral corrective saccades. Vestibular testing revealed high-grade bilateral vestibular loss associated with all semicircular canals, a considerable decline compared to her function 3 years prior. DISCUSSION: Gentamicin is indicated for surgical prophylaxis when a patient has a true allergy to penicillins and cannot receive cephalosporins, though alternatives exist. True allergies include IgE-mediated illness (anaphylaxis, bronchospasm, or urticaria 30-60 minutes after administration) or exfoliative reactions (Stevens-Johnson Syndrome or Toxic Epidermal Necrolysis). The authors encourage more prudent use of gentamicin, especially in patients susceptible for debilitating otologic insults, and offer recommendations for alternative agents prior to using gentamicin.


Subject(s)
Gentamicins , Ototoxicity , Female , Humans , Anti-Bacterial Agents/adverse effects , Gentamicins/adverse effects , Hypersensitivity , Ototoxicity/etiology , Penicillins/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...