Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 543
Filter
1.
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
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.
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
4.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206364

ABSTRACT

Umbilical cord-derived mesenchymal stromal cells (UCMSCs) have potential applications in regenerative medicine. UCMSCs have been demonstrated to repair tissue damage in many inflammatory and degenerative diseases. We have previously shown that UCMSC exosomes reduce nerve injury-induced pain in rats. In this study, we characterized UCMSC exosomes using RNA sequencing and proteomic analyses and investigated their protective effects on cisplatin-induced hearing loss in mice. Two independent experiments were designed to investigate the protective effects on cisplatin-induced hearing loss in mice: (i) chronic intraperitoneal cisplatin administration (4 mg/kg) once per day for 5 consecutive days and intraperitoneal UCMSC exosome (1.2 µg/µL) injection at the same time point; and (ii) UCMSC exosome (1.2 µg/µL) injection through a round window niche 3 days after chronic cisplatin administration. Our data suggest that UCMSC exosomes exert protective effects in vivo. The post-traumatic administration of UCMSC exosomes significantly improved hearing loss and rescued the loss of cochlear hair cells in mice receiving chronic cisplatin injection. Neuropathological gene panel analyses further revealed the UCMSC exosomes treatment led to beneficial changes in the expression levels of many genes in the cochlear tissues of cisplatin-injected mice. In conclusion, UCMSC exosomes exerted protective effects in treating ototoxicity-induced hearing loss by promoting tissue remodeling and repair.


Subject(s)
Cochlear Diseases/etiology , Cochlear Diseases/therapy , Exosomes/metabolism , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/cytology , Animals , Antineoplastic Agents/adverse effects , Biological Therapy , Biomarkers , Cisplatin/adverse effects , Cochlear Diseases/pathology , Disease Models, Animal , Exosomes/transplantation , Hair Cells, Auditory, Outer/pathology , Hearing Loss/etiology , Hearing Loss/metabolism , Hearing Loss/therapy , Immunophenotyping , Mice , MicroRNAs/genetics , Proteomics/methods , Treatment Outcome
5.
Cell Death Dis ; 12(7): 682, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234110

ABSTRACT

The prevalence of noise-induced hearing loss (NIHL) continues to increase, with limited therapies available for individuals with cochlear damage. We have previously established that the transcription factor FOXO3 is necessary to preserve outer hair cells (OHCs) and hearing thresholds up to two weeks following mild noise exposure in mice. The mechanisms by which FOXO3 preserves cochlear cells and function are unknown. In this study, we analyzed the immediate effects of mild noise exposure on wild-type, Foxo3 heterozygous (Foxo3+/-), and Foxo3 knock-out (Foxo3-/-) mice to better understand FOXO3's role(s) in the mammalian cochlea. We used confocal and multiphoton microscopy to examine well-characterized components of noise-induced damage including calcium regulators, oxidative stress, necrosis, and caspase-dependent and caspase-independent apoptosis. Lower immunoreactivity of the calcium buffer Oncomodulin in Foxo3-/- OHCs correlated with cell loss beginning 4 h post-noise exposure. Using immunohistochemistry, we identified parthanatos as the cell death pathway for OHCs. Oxidative stress response pathways were not significantly altered in FOXO3's absence. We used RNA sequencing to identify and RT-qPCR to confirm differentially expressed genes. We further investigated a gene downregulated in the unexposed Foxo3-/- mice that may contribute to OHC noise susceptibility. Glycerophosphodiester phosphodiesterase domain containing 3 (GDPD3), a possible endogenous source of lysophosphatidic acid (LPA), has not previously been described in the cochlea. As LPA reduces OHC loss after severe noise exposure, we treated noise-exposed Foxo3-/- mice with exogenous LPA. LPA treatment delayed immediate damage to OHCs but was insufficient to ultimately prevent their death or prevent hearing loss. These results suggest that FOXO3 acts prior to acoustic insult to maintain cochlear resilience, possibly through sustaining endogenous LPA levels.


Subject(s)
Forkhead Box Protein O3/deficiency , Hair Cells, Auditory, Outer/metabolism , Hearing Loss, Noise-Induced/metabolism , Animals , Cell Death , Disease Models, Animal , Female , Forkhead Box Protein O3/genetics , Gene Expression Regulation , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hearing , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/pathology , Homozygote , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Male , Mice, Knockout , Noise , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Time Factors
6.
Neurotox Res ; 39(4): 1227-1237, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33900547

ABSTRACT

Paraquat, a superoxide generator, can damage the cochlea causing an ototoxic hearing loss. The purpose of the study was to determine if deletion of Bak, a pro-apoptotic gene, would reduce paraquat ototoxicity or if deletion of Sirt3, which delays age-related hearing loss under caloric restriction, would increase paraquat ototoxicity. We tested these two hypotheses by treating postnatal day 3 cochlear cultures from Bak±, Bak-/-, Sirt3±, Sirt3-/-, and WT mice with paraquat and compared the results to a standard rat model of paraquat ototoxicity. Paraquat damaged nerve fibers and dose-dependently destroyed rat outer hair cells (OHCs) and inner hair cells (IHCs). Rat hair cell loss began in the base of the cochlea with a 10 µM dose and as the dose increased from 50 to 500 µM, the hair cell loss increased near the base of the cochlea and spread toward the apex of the cochlea. Rat OHC losses were consistently greater than IHC losses. Unexpectedly, in all mouse genotypes, paraquat-induced hair cell lesions were maximal near the apex of the cochlea and minimal near the base. This unusual damage gradient is opposite to that seen in paraquat-treated rats and in mice and rats treated with other ototoxic drugs. However, paraquat always induced greater OHC loss than IHC loss in all mouse strains. Contrary to our hypothesis, Bak deficient mice were more vulnerable to paraquat ototoxicity than WT mice (Bak-/- > Bak± > WT), suggesting that Bak plays a protective role against hair cell stress. Also, contrary to expectation, Sirt3-deficient mice did not differ significantly from WT mice, possibly due to the fact that Sirt3 was not experimentally upregulated in Sirt3-expressing mice prior to paraquat treatment. Our results show for the first time a gradient of ototoxic damage in mice that is greater in the apex than the base of the cochlea.


Subject(s)
Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Outer/drug effects , Herbicides/toxicity , Paraquat/toxicity , Sirtuin 3/deficiency , bcl-2 Homologous Antagonist-Killer Protein/deficiency , Animals , Animals, Newborn , Cells, Cultured , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , Dose-Response Relationship, Drug , Female , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Sirtuin 3/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics
7.
ACS Appl Mater Interfaces ; 13(6): 7102-7114, 2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33528239

ABSTRACT

Overproduction of reactive oxygen species (ROS) and inflammation are two key pathogeneses of noise-induced hearing loss (NIHL), which leads to outer hair cell (OHC) damage and hearing loss. In this work, we successfully developed ROS-responsive nanoparticles as berberine (BBR) carriers (PL-PPS/BBR) for OHC-targeted therapy of NIHL: Prestin-targeting peptide 2 (PrTP2)-modified nanoparticles (PL-PPS/BBR), which effectively accumulated in OHC areas, and poly(propylene sulfide)120 (PPS120), which scavenged ROS and converted to poly(propylene sulfoxide)120 in a ROS environment to disintegrate and provoke the rapid release of BBR with anti-inflammatory and antioxidant effects. In this study, satisfactory anti-inflammatory and antioxidant effects of PL-PPS/BBR were confirmed. Immunofluorescence and scanning electron microscopy (SEM) images showed that PL-PPS/BBR effectively accumulated in OHCs and protected the morphological integrity of OHCs. The auditory brainstem response (ABR) results demonstrated that PL-PPS/BBR significantly improved hearing in NIHL guinea pigs after noise exposure. This work suggested that PL-PPS/BBR may be a new potential treatment for noise-associated injury with clinical application.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Berberine/pharmacology , Hair Cells, Auditory, Outer/drug effects , Hearing Loss, Noise-Induced/drug therapy , Reactive Oxygen Species/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antioxidants/chemistry , Berberine/chemistry , Disease Models, Animal , Drug Carriers/chemistry , Evoked Potentials, Auditory, Brain Stem/drug effects , Guinea Pigs , Molecular Structure , Nanoparticles/chemistry , Particle Size , Reactive Oxygen Species/metabolism , Surface Properties
8.
Arch Toxicol ; 95(3): 1003-1021, 2021 03.
Article in English | MEDLINE | ID: mdl-33495873

ABSTRACT

The peripheral auditory and vestibular systems rely on sensorineural structures that are vulnerable to ototoxic agents that cause hearing loss and/or equilibrium deficits. Although attention has focused on hair cell loss as the primary pathology underlying ototoxicity, evidence from the peripheral vestibular system indicates that hair cell loss during chronic exposure is preceded by synaptic uncoupling from the neurons and is potentially reversible. To determine if synaptic pathology also occurs in the peripheral auditory system, we examined the extent, time course, and reversibility of functional and morphological alterations in cochleae from mice exposed to 3,3'-iminodipropionitrile (IDPN) in drinking water for 2, 4 or 6 weeks. Functionally, IDPN exposure caused progressive high- to low-frequency hearing loss assessed by measurement of auditory brainstem response wave I absolute thresholds and amplitudes. The extent of hearing loss scaled with the magnitude of vestibular dysfunction assessed behaviorally. Morphologically, IDPN exposure caused progressive loss of outer hair cells (OHCs) and synapses between the inner hair cells (IHCs) and primary auditory neurons. In contrast, IHCs were spared from ototoxic damage. Importantly, hearing loss consistent with cochlear synaptopathy preceded loss of OHCs and synapses and, moreover, recovered if IDPN exposure was stopped before morphological pathology occurred. Our observations suggest that synaptic uncoupling, perhaps as an early phase of cochlear synaptopathy, also occurs in the peripheral auditory system in response to IDPN exposure. These findings identify novel mechanisms that contribute to the earliest stages of hearing loss in response to ototoxic agents and possibly other forms of acquired hearing loss.


Subject(s)
Cochlea/drug effects , Hearing Loss/chemically induced , Nitriles/toxicity , Ototoxicity/etiology , Animals , Cochlea/pathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hearing Loss/physiopathology , Male , Mice , Mice, 129 Strain , Nitriles/administration & dosage , Ototoxicity/physiopathology , Synapses/drug effects , Synapses/pathology , Time Factors
9.
J Cyst Fibros ; 20(2): 271-277, 2021 03.
Article in English | MEDLINE | ID: mdl-32147183

ABSTRACT

BACKGROUND: Cystic fibrosis patients are often adminstered tobramycin to treat pulmonary infections. Unfortunately, a common side effect is hearing loss, which can fluctuate. Ebselen has known anti-inflammatory properties and could reduce the incidence and severity of tobramycin-induced hearing loss. METHODS: In vitro: neonatal cochlear cultures were treated with tobramycin or cotreated with tobramycin and ebselen for 3 days. In vivo: adult mice were injected with tobramycin or tobramycin and ebselen for 14 days. ABRs were collected in a repeated measures design until 56 days after treatments. ABR threshold shifts were analyzed and a novel cochleotoxic criteria applied to determine the incidence of ototoxicity. Cochlear immunohistology was analyzed for IHC and OHC loss. RESULTS: Tobramycin leads to significant IHC and OHC loss in cochlear explant cultures. Ebselen co-treatment at 1:20 concentrations resulted in significant otoprotection. Tobramycin leads to significant ABR threshold shifts that are ameliorated by ebselen co-treatment. Hearing loss did not correlate with significant IHC or OHC loss. CONCLUSIONS: This mouse model of tobramycin-induced ototoxicity is clinically relevant in that it results in an incidence and severity of hearing loss recently documented in clinic. The in vitro experiments show that tobramycin kills hair cells and that ebselen co-treatment can attenuate this ototoxicity. The in vivo model shows tobramycin-induced hearing loss is ameliorated by ebselen co-treatment, but this is not explained by concomitant hair cell loss. These preclinical data support the testing of ebselen in CF patients receiving tobramycin treatment.


Subject(s)
Isoindoles/pharmacology , Organoselenium Compounds/pharmacology , Ototoxicity/prevention & control , Tobramycin/toxicity , Animals , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory, Outer/drug effects , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Mice
10.
Commun Biol ; 3(1): 792, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33361775

ABSTRACT

The stereocilia of the inner ear sensory cells contain the actin-binding protein radixin, encoded by RDX. Radixin is important for hearing but remains functionally obscure. To determine how radixin influences hearing sensitivity, we used a custom rapid imaging technique to visualize stereocilia motion while measuring electrical potential amplitudes during acoustic stimulation. Radixin inhibition decreased sound-evoked electrical potentials. Other functional measures, including electrically induced sensory cell motility and sound-evoked stereocilia deflections, showed a minor amplitude increase. These unique functional alterations demonstrate radixin as necessary for conversion of sound into electrical signals at acoustic rates. We identified patients with RDX variants with normal hearing at birth who showed rapidly deteriorating hearing during the first months of life. This may be overlooked by newborn hearing screening and explained by multiple disturbances in postnatal sensory cells. We conclude radixin is necessary for ensuring normal conversion of sound to electrical signals in the inner ear.


Subject(s)
Cytoskeletal Proteins/metabolism , Hair Cells, Auditory, Outer/metabolism , Membrane Proteins/metabolism , Stereocilia/metabolism , Acoustic Stimulation , Alleles , Animals , Arsenicals/pharmacology , Child, Preschool , Cytoskeletal Proteins/genetics , Disease Models, Animal , Female , Fluorescent Antibody Technique , Gene Expression , Genetic Variation , Genotype , Guinea Pigs , Hair Cells, Auditory, Outer/drug effects , Hearing Loss/diagnosis , Hearing Loss/genetics , Humans , Mechanotransduction, Cellular/genetics , Membrane Proteins/genetics , Models, Biological , Pedigree , Stereocilia/drug effects
11.
Int J Mol Sci ; 21(18)2020 Sep 13.
Article in English | MEDLINE | ID: mdl-32933159

ABSTRACT

The administration of immune checkpoint inhibitors (ICIs) often leads to immune-related adverse events. However, their effect on auditory function is largely unexplored. Thorough preclinical studies have not been published yet, only sporadic cases and pharmacovigilance reports suggest their significance. Here we investigated the effect of anti-PD-1 antibody treatment (4 weeks, intraperitoneally, 200 µg/mouse, 3 times/week) on hearing function and cochlear morphology in C57BL/6J mice. ICI treatment did not influence the hearing thresholds in click or tone burst stimuli at 4-32 kHz frequencies measured by auditory brainstem response. The number and morphology of spiral ganglion neurons were unaltered in all cochlear turns. The apical-middle turns (<32 kHz) showed preservation of the inner and outer hair cells (OHCs), whilst ICI treatment mitigated the age-related loss of OHCs in the basal turn (>32 kHz). The number of Iba1-positive macrophages has also increased moderately in this high frequency region. We conclude that a 4-week long ICI treatment does not affect functional and morphological integrity of the inner ear in the most relevant hearing range (4-32 kHz; apical-middle turns), but a noticeable preservation of OHCs and an increase in macrophage activity appeared in the >32 kHz basal part of the cochlea.


Subject(s)
Antibodies, Monoclonal/pharmacology , Auditory Threshold/drug effects , Cochlea/drug effects , Hair Cells, Auditory, Outer/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing , Immune Checkpoint Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Spiral Ganglion/drug effects
12.
Neurotox Res ; 38(3): 808-823, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32607920

ABSTRACT

2-Hydroxypropyl-ß-cyclodextrin (HPßCD), a cholesterol chelator, is being used to treat diseases associated with abnormal cholesterol metabolism such as Niemann-Pick C1 (NPC1). However, the high doses of HPßCD needed to slow disease progression may cause hearing loss. Previous studies in mice have suggested that HPßCD ototoxicity results from selective outer hair cell (OHC) damage. However, it is unclear if HPßCD causes the same type of damage or is more or less toxic to other species such as rats, which are widely used in toxicity research. To address these issues, rats were given a subcutaneous injection of HPßCD between 500 and 4000 mg/kg. Distortion product otoacoustic emissions (DPOAE), the cochlear summating potential (SP), and compound action potential (CAP) were used to assess cochlear function followed by quantitative analysis of OHC and inner hair cell (IHC) loss. The 3000- and 4000-mg/kg doses abolished DPOAE and greatly reduced SP and CAP amplitudes. These functional deficits were associated with nearly complete loss of OHC as well as ~ 80% IHC loss over the basal two thirds of the cochlea. The 2000-mg/kg dose abolished DPOAE and significantly reduced SP and CAP amplitudes at the high frequencies. These deficits were linked to OHC and IHC losses in the high-frequency region of the cochlea. Little or no damage occurred with 500 or 1000 mg/kg of HPßCD. The HPßCD-induced functional and structural deficits in rats occurred suddenly, involved damage to both IHC and OHC, and were more severe than those reported in mice.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Outer/drug effects , Hearing Loss/chemically induced , Animals , Cochlea/drug effects , Otoacoustic Emissions, Spontaneous/drug effects , Ototoxicity/drug therapy , Rats, Sprague-Dawley
13.
Sci Rep ; 10(1): 6222, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32277153

ABSTRACT

Outer hair cell (OHC) nonlinear capacitance (NLC) represents voltage sensor charge movements of prestin (SLC26a5), the protein responsible for OHC electromotility. Previous measures of NLC frequency response have employed methods which did not assess the influence of dielectric loss (sensor charge movements out of phase with voltage) that may occur, and such loss conceivably may influence prestin's frequency dependent activity. Here we evaluate prestin's complex capacitance out to 30 kHz and find that prestin's frequency response determined using this approach coincides with all previous estimates. We also show that membrane tension has no effect on prestin's frequency response, despite substantial shifts in its voltage operating range, indicating that prestin transition rate alterations do not account for the shifts. The magnitude roll-off of prestin activity across frequency surpasses the reductions of NLC caused by salicylate treatments that are known to abolish cochlear amplification. Such roll-off likely limits the effectiveness of prestin in contributing to cochlear amplification at the very high acoustic frequencies processed by some mammals.


Subject(s)
Electric Capacitance , Hair Cells, Auditory, Outer/metabolism , Membrane Potentials/physiology , Proteins/metabolism , Animals , Cells, Cultured , Guinea Pigs , Hair Cells, Auditory, Outer/cytology , Hair Cells, Auditory, Outer/drug effects , Membrane Potentials/drug effects , Patch-Clamp Techniques , Primary Cell Culture , Salicylates/pharmacology
14.
Hear Res ; 389: 107905, 2020 04.
Article in English | MEDLINE | ID: mdl-32062294

ABSTRACT

Cisplatin is used to treat a variety of solid tumors in both children and adults. However, cisplatin has serious side-effects, some of which may permanently affect patients' quality of life following treatment, such as ototoxicity. There is currently no FDA-approved therapy for the prevention or treatment of cisplatin-induced hearing loss. Herein we examine the potential for statins to prevent cisplatin-induced ototoxicity. Statins, a class of drugs commonly used to prevent or manage hypercholesterolemia, have been of clinical utility for decades with dependable outcomes and reliable safety profiles in humans. Statins are known to be protective in animal models of noise-induced and age-related hearing loss. Moreover, studies have demonstrated an additive benefit of statins in cancer treatment. In the current study, lovastatin reduces cisplatin-induced hearing loss in adult mice. Lovastatin-mediated protection was significantly greater among female than male mice, and the dose of lovastatin required for protection was different between the sexes. Taken together our data indicate that lovastatin reduces cisplatin-induced hearing loss in mice and suggest that concurrent statin and cisplatin therapy may represent a feasible clinical strategy for reducing cisplatin-induced ototoxicity that should be explored for future clinical use.


Subject(s)
Anticholesteremic Agents/pharmacology , Cochlea/drug effects , Hearing Loss/prevention & control , Hearing/drug effects , Lovastatin/pharmacology , Animals , Auditory Threshold/drug effects , Cisplatin , Cochlea/metabolism , Cochlea/pathology , Cochlea/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hearing Loss/chemically induced , Hearing Loss/metabolism , Hearing Loss/physiopathology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred CBA , Ototoxicity
15.
Braz. j. otorhinolaryngol. (Impr.) ; 86(1): 30-37, Jan.-Feb. 2020. tab, graf
Article in English | LILACS | ID: biblio-1089360

ABSTRACT

Abstract Introduction Ototoxicity is a health problem appearing after powerful treatments in serious health conditions. It is sometimes inevitable when treatment of the serious disease is required. Cisplatin is an antineoplastic agent which was investigated previously to reveal increased nitrogen and reactive oxygen radicals that damages hair cells, resulting in ototoxicity. N-acetylcysteine, previously shown to decrease ototoxicity caused by different agents, is known to be a powerful in vitro antioxidant. Probably N-acetylcysteine, in addition to its antioxidant effect, blocks a cascade where reactive oxygen species result in apoptosis in the cochlea. Objectives The possible preventive effect of N-acetylcysteine in cisplatin ototoxicity was studied with auditory brain stem responses, otoacoustic emissions, and histopathological investigation of the cochlea in a scanning electron microscopy. Methods This study was conducted on 21 Wistar Albino rats in four groups. 1 mL/kg/day three times in total intraperitoneal (i.p.) Saline (n = 5), 500 mg/kg/day i.p. three times in total N-acetylcysteine (n = 5), i.p. 15 mg/kg cisplatin alone (single dose) (n = 5) and i.p. 15 mg/kg cisplatin plus 500 mg/kg/day N-acetylcysteine (n = 6) were administered. The rats were anesthetized to study the hearing tests before and after the experiment. The rats were sacrificed to investigate the cochleas by scanning electron microscopy. Results Auditory brain stem responses and otoacoustic emissions values were attenuated in the cisplatin group. The group that received N-acetylcysteine in addition to cisplatin had better auditory brain stem responses thresholds and otoacoustic emissions. The samples obtained from the cisplatin group showed surface irregularities, degeneration areas, and total or partial severe stereocilia losses. The changes were milder in the cisplatin + N-acetylcysteine group. Conclusion Cisplatin ototoxicity can be detected by auditory brain stem responses and otoacoustic emissions testing in rats. N-acetylcysteine may protect the cochlear cells from histopathological changes. We concluded that N-acetylcysteine given 4 h after cisplatin injection has a potential otoprotective effect against cisplatin ototoxicity. which suggests it could be used in clinical trials.


Resumo Introdução A ototoxicidade é um problema que pode ocorrer após certos tipos de tratamentos para condições graves de saúde. Às vezes é inevitável quando o tratamento da doença é necessário. A cisplatina é um agente antineoplásico cujo uso em pesquisas anteriores demonstrou aumentar os radicais livres de nitrogênio e espécies reativas de oxigênio que danificam as células ciliadas e resultam em ototoxicidade. Por outro lado, a N-acetilcisteína, que já demonstrou diminuir a ototoxicidade causada por diferentes agentes, é conhecida por ser um potente antioxidante in vitro. Provavelmente a N-acetilcisteína, além de seu efeito antioxidante, bloqueia uma cascata onde espécies reativas de oxigênio resultam em apoptose na cóclea. Objetivos Estudar o possível efeito preventivo da N-acetilcisteína na ototoxicidade por cisplatina por meio de potencial evocado auditivo de tronco encefálico, emissões otoacústicas e investigação histopatológica da cóclea por microscopia eletrônica de varredura. Método Este estudo foi realizado em 21 ratos albinos Wistar, separados em quatro grupos. Foram administrados: 1 mL/kg/dia intraperitoneal (i.p.) de solução salina (n = 5), três vezes no total; 500 mg/kg/dia i.p. de N-acetilcisteína (n = 5), três vezes no total; 15 mg/kg i.p. (dose única) somente de cisplatina (n = 5) e 15 mg/kg i.p. de cisplatina e 500 mg/kg/dia i.p. de N-acetilcisteína (n = 6). Os ratos foram anestesiados para estudo dos testes auditivos antes e depois do experimento. Os ratos foram sacrificados para investigação da cóclea por microscopia eletrônica de varredura. Resultados Os potenciais evocados auditivos de tronco encefálico e os valores das emissões otoacústicas estavam atenuados no grupo cisplatina. O grupo que recebeu N-acetilcisteína além da cisplatina apresentou melhores limiares de respostas auditivas do tronco encefálico e emissões otoacústicas. As amostras obtidas do grupo cisplatina apresentaram irregularidades de superfície, áreas de degeneração, com perdas graves totais ou parciais de estereocílios. As alterações foram mais leves no grupo cisplatina + N-acetilcisteína. Conclusão A ototoxicidade por cisplatina pode ser detectada por meio de potenciais evocados auditivos de tronco encefálico e pelo teste de emissões otoacústicas em ratos. A N-acetilcisteína pode proteger as células cocleares contra alterações histopatológicas. Concluímos que a N-acetilcisteína administrada 4 horas após a injeção de cisplatina tem potencial efeito otoprotetor contra a ototoxicidade por cisplatina e pode ser utilizada em ensaios clínicos.


Subject(s)
Animals , Male , Acetylcysteine/administration & dosage , Cisplatin/adverse effects , Protective Agents/administration & dosage , Ototoxicity/etiology , Antineoplastic Agents/adverse effects , Antioxidants/administration & dosage , Acetylcysteine/pharmacology , Microscopy, Electron, Scanning , Evoked Potentials, Auditory, Brain Stem , Rats, Wistar , Cochlea/pathology , Apoptosis , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Protective Agents/pharmacology , Disease Models, Animal , Stereocilia/drug effects , Stereocilia/pathology , Ototoxicity/prevention & control , Hearing Tests , Antioxidants/pharmacology
16.
Braz J Otorhinolaryngol ; 86(1): 30-37, 2020.
Article in English | MEDLINE | ID: mdl-30268784

ABSTRACT

INTRODUCTION: Ototoxicity is a health problem appearing after powerful treatments in serious health conditions. It is sometimes inevitable when treatment of the serious disease is required. Cisplatin is an antineoplastic agent which was investigated previously to reveal increased nitrogen and reactive oxygen radicals that damages hair cells, resulting in ototoxicity. N-acetylcysteine, previously shown to decrease ototoxicity caused by different agents, is known to be a powerful in vitro antioxidant. Probably N-acetylcysteine, in addition to its antioxidant effect, blocks a cascade where reactive oxygen species result in apoptosis in the cochlea. OBJECTIVES: The possible preventive effect of N-acetylcysteine in cisplatin ototoxicity was studied with auditory brain stem responses, otoacoustic emissions, and histopathological investigation of the cochlea in a scanning electron microscopy. METHODS: This study was conducted on 21 Wistar Albino rats in four groups. 1mL/kg/day three times in total intraperitoneal (i.p.) Saline (n=5), 500mg/kg/day i.p. three times in total N-acetylcysteine (n=5), i.p. 15mg/kg cisplatin alone (single dose) (n=5) and i.p. 15mg/kg cisplatin plus 500mg/kg/day N-acetylcysteine (n=6) were administered. The rats were anesthetized to study the hearing tests before and after the experiment. The rats were sacrificed to investigate the cochleas by scanning electron microscopy. RESULTS: Auditory brain stem responses and otoacoustic emissions values were attenuated in the cisplatin group. The group that received N-acetylcysteine in addition to cisplatin had better auditory brain stem responses thresholds and otoacoustic emissions. The samples obtained from the cisplatin group showed surface irregularities, degeneration areas, and total or partial severe stereocilia losses. The changes were milder in the cisplatin+N-acetylcysteine group. CONCLUSION: Cisplatin ototoxicity can be detected by auditory brain stem responses and otoacoustic emissions testing in rats. N-acetylcysteine may protect the cochlear cells from histopathological changes. We concluded that N-acetylcysteine given 4h after cisplatin injection has a potential otoprotective effect against cisplatin ototoxicity. which suggests it could be used in clinical trials.


Subject(s)
Acetylcysteine/administration & dosage , Antineoplastic Agents/adverse effects , Antioxidants/administration & dosage , Cisplatin/adverse effects , Ototoxicity/etiology , Protective Agents/administration & dosage , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis , Cochlea/drug effects , Cochlea/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hearing Tests , Male , Microscopy, Electron, Scanning , Ototoxicity/prevention & control , Protective Agents/pharmacology , Rats, Wistar , Signal-To-Noise Ratio , Stereocilia/drug effects , Stereocilia/pathology
17.
Dev Biol ; 453(2): 191-205, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31185200

ABSTRACT

The development of asymmetric patterns along biologically relevant axes is a hallmark of many vertebrate organs or structures. One example is the sensory epithelium of the mammalian auditory system. Two distinct types of mechanosensory hair cells (inner and outer) and at least six types of associated supporting cells are precisely and asymmetrically arrayed along the radial (medial-lateral) axis of the cochlear spiral. Immunolabeling of developing cochleae indicates differential expression of Glycogen synthase kinase 3ß (GSK3ß) along the same axis. To determine whether GSK3ß plays a role in specification of cell fates along the medial-lateral axis, GSK3 activity was blocked pharmacologically in cochlear explants. Results indicate significant changes in both the number of hair cells and in the specification of hair cell phenotypes. The overall number of inner hair cells increased as a result of both a shift in the medial boundary between sensory and non-sensory regions of the cochlea and a change in the specification of inner and outer hair cell phenotypes. Previous studies have inhibited GSK3 as a method to examine effects of canonical Wnt signaling. However, quantification of changes in Wnt pathway target genes in GSK3-inhibited cochleae, and treatment with more specific Wnt agonists, indicated that the Wnt pathway is not activated. Instead, expression of Bmp4 in a population of GSK3ß-expressing cells was shown to be down-regulated. Finally, addition of BMP4 to GSK3-inhibited cochleae achieved a partial rescue of the hair cell phenotype. These results demonstrate a role for GSK3ß in the specification of cellular identities along the medial-lateral axis of the cochlea and provide evidence for a positive role for GSK3ß in the expression of Bmp4.


Subject(s)
Cell Lineage , Glycogen Synthase Kinase 3 beta/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/enzymology , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Lineage/drug effects , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Female , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hair Cells, Auditory/drug effects , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/enzymology , Hair Cells, Auditory, Outer/cytology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/enzymology , Mice , Models, Biological , Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Wnt Signaling Pathway/drug effects
18.
Hear Res ; 375: 66-74, 2019 04.
Article in English | MEDLINE | ID: mdl-30827780

ABSTRACT

Cisplatin-induced ototoxicity results in significant, permanent hearing loss in pediatric and adult cancer survivors. Elucidating the mechanisms underlying cisplatin-induced hearing loss as well as the development of therapies to reduce and/or reverse cisplatin ototoxicity have been impeded by suboptimal animal models. Clinically, cisplatin is most commonly administered in multi-dose, multi-cycle protocols. However, many animal studies are conducted using single injections of high-dose cisplatin, which is not reflective of clinical cisplatin administration protocols. Significant limitations of both high-dose, single-injection protocols and previous multi-dose protocols in rodent models include high mortality rates and relatively small changes in hearing sensitivity. These limitations restrict assessment of both long-term changes in hearing sensitivity and effects of potential protective therapies. Here, we present a detailed method for an optimized mouse model of cisplatin ototoxicity that utilizes a multi-cycle administration protocol that better approximates the type and degree of hearing loss observed clinically. This protocol results in significant hearing loss with very low mortality. This mouse model of cisplatin ototoxicity provides a platform for examining mechanisms of cisplatin-induced hearing loss as well as developing therapies to protect the hearing of cancer patients receiving cisplatin therapy.


Subject(s)
Cisplatin/toxicity , Ototoxicity/etiology , Animals , Auditory Threshold/drug effects , Cisplatin/administration & dosage , Disease Models, Animal , Disease Progression , Drug Administration Schedule , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Humans , Male , Mice , Mice, Inbred CBA , Otoacoustic Emissions, Spontaneous/drug effects , Ototoxicity/pathology , Ototoxicity/physiopathology
19.
Biol Pharm Bull ; 42(1): 73-80, 2019.
Article in English | MEDLINE | ID: mdl-30606991

ABSTRACT

It is well-known that outer hair cell (OHC) loss occurs in the cochlea of animal models of permanent hearing loss induced by intense noise exposure. Our earlier studies demonstrated the production of hydroxynonenal and peroxynitrite, as well as the disruption of gap junction-mediated intercellular communication (GJIC), in the cochlear spiral ligament prior to noise-induced sudden hearing loss. The goal of the present study was to evaluate the mechanism underlying cochlear OHC loss after sudden hearing loss induced by intense noise exposure. In organ of Corti explant cultures from mice, no significant OHC loss was observed after in vitro exposure to 4-hydroxynonenal (a product of lipid peroxidation), H2O2, SIN-1 (peroxynitrite generator), and carbenoxolone (a gap junction inhibitor). Interestingly, in vivo intracochlear carbenoxolone injection through the posterior semicircular canal caused marked OHC and hearing loss, as well as the disruption of gap junction-mediated intercellular communication in the cochlear spiral ligament. However, no significant OHC loss was observed in vivo in animals treated with 4-hydroxynonenal and SIN-1. Taken together, our data suggest that disruption of GJIC in the cochlear lateral wall structures is an important cause of cochlear OHC loss in models of hearing loss, including those induced by noise.


Subject(s)
Acoustic Stimulation/adverse effects , Cell Communication/physiology , Gap Junctions/metabolism , Hair Cells, Auditory, Outer/metabolism , Hearing Loss, Noise-Induced/metabolism , Spiral Ligament of Cochlea/metabolism , Aldehydes/toxicity , Animals , Cell Communication/drug effects , Cochlea/drug effects , Cochlea/metabolism , Gap Junctions/drug effects , Hair Cells, Auditory, Outer/drug effects , Hearing Loss, Noise-Induced/chemically induced , Hearing Loss, Noise-Induced/etiology , Hydrogen Peroxide/toxicity , Male , Mice , Organ Culture Techniques , Spiral Ligament of Cochlea/drug effects
20.
Otol Neurotol ; 40(2): 254-263, 2019 02.
Article in English | MEDLINE | ID: mdl-30570608

ABSTRACT

HYPOTHESIS: SENS-401 (R-azasetron besylate) is effective against severe acoustic trauma-induced hearing loss. BACKGROUND: SENS-401 has calcineurin inhibiting properties and attenuates cisplatin-induced hearing loss in a rat model. Cisplatin-induced and acoustic trauma-induced hearing loss share common apoptotic pathways. METHODS: The dose-response relationship of SENS-401 (6.6 mg/kg BID, 13.2 mg/kg BID, 26.4 mg/kg QD) and treatment time-window (13.2 mg/kg BID starting 24, 72, and 96 h posttrauma) versus placebo for 28 days were evaluated in a male rat model of severe acoustic trauma-induced hearing loss (120 dB SPL, 2 h) using auditory brainstem response (ABR) and distortion product otoacoustic emissions (DPOAE) measures followed by cochlear outer hair cell (OHC) counting with myosin-VIIa immunolabeling. RESULTS: All SENS-401 doses improved ABR threshold shift and recovery, reaching statistical significance (p < 0.05) for ABR threshold recoveries after 28-days treatment. DPOAE amplitude loss and recovery improved markedly for 13.2 mg/kg BID SENS-401, reaching significance after 14 days (p < 0.05). Significant improvements in ABR threshold shifts/recovery and DPOAE amplitude loss occurred with up to 96-hours delay in initiating SENS-401 (p < 0.05), and in DPOAE amplitude recovery with up to 72-hours delay (p < 0.05). Significantly more surviving OHCs were present after SENS-401 treatment compared with placebo after 24 to 96-hours delay posttrauma, with up to 5.3-fold more cells in the basal cochlea turn. CONCLUSIONS: In vivo data support the otoprotective potential of twice daily oral SENS-401. Improvements in hearing loss recovery make SENS-401 a promising clinical candidate for acoustic trauma-induced hearing loss, including when treatment is not initiated immediately.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Sensorineural/drug therapy , Oxazines/pharmacology , Acoustic Stimulation/adverse effects , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cisplatin/toxicity , Hair Cells, Auditory, Outer/drug effects , Hearing Loss, Sensorineural/chemically induced , Male , Otoacoustic Emissions, Spontaneous/drug effects , Oxazines/administration & dosage , Oxazines/therapeutic use , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...