Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.092
Filter
1.
Pharmacol Res Perspect ; 12(4): e1204, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969959

ABSTRACT

Reversible axonal swelling and brainstem auditory evoked potential (BAEP) changes were observed in standard chronic (9-month) toxicology studies in dogs treated with ritlecitinib, an oral Janus kinase 3/tyrosine kinase expressed in hepatocellular carcinoma family kinase inhibitor, at exposures higher than the approved 50-mg human dose. To evaluate the clinical relevance of the dog toxicity finding, this phase 2a, double-blind study assessed BAEP changes and intraepidermal nerve fiber (IENF) histology in adults with alopecia areata treated with ritlecitinib. Patients were randomized to receive oral ritlecitinib 50 mg once daily (QD) with a 4-week loading dose of 200 mg QD or placebo for 9 months (placebo-controlled phase); they then entered the active-therapy extension and received ritlecitinib 50 mg QD (with a 4-week loading dose of 200 mg in patients switching from placebo). Among the 71 patients, no notable mean differences in change from baseline (CFB) in Waves I-V interwave latency (primary outcome) or Wave V amplitude on BAEP at a stimulus intensity of 80 dB nHL were observed in the ritlecitinib or placebo group at Month 9, with no notable differences in interwave latency or Wave V amplitude between groups. The CFB in mean or median IENF density and in percentage of IENFs with axonal swellings was minimal and similar between groups at Month 9. Ritlecitinib treatment was also not associated with an imbalanced incidence of neurological and audiological adverse events. These results provide evidence that the BAEP and axonal swelling finding in dogs are not clinically relevant in humans.


Subject(s)
Alopecia Areata , Evoked Potentials, Auditory, Brain Stem , Nerve Fibers , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Alopecia Areata/drug therapy , Alopecia Areata/pathology , Double-Blind Method , Evoked Potentials, Auditory, Brain Stem/drug effects , Nerve Fibers/drug effects , Nerve Fibers/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Animals , Dogs
2.
Sci Rep ; 14(1): 17576, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39079962

ABSTRACT

Hearing impairment in patients with chronic kidney disease (CKD), can affect the quality of life. At present, hearing dysfunction does not have an approved pharmacologic therapy. This study aimed to investigate the protective effects and possible mechanisms of curcumin as a therapeutic agent on hearing impairment in patients with chronic kidney disease. We conducted a randomized controlled trial of 40 chronic kidney disease patients not on dialysis with hearing impairment. Participants were randomly divided into two groups. One group received curcumin daily and the other received a placebo for 12 weeks. The interval between III and V waves, latency of wave V, auditory brain stem response (ABR) threshold, speech reception threshold (SRT), and speech discrimination score (SDS) were evaluated and analyzed before and after the intervention. After treatment, in the curcumin group, III-V waves interval and the latency of wave V were significantly reduced (P value < 0.0001), also ABR threshold was demonstrated a significant improvement (P value < 0.0001). In the trial group, the SDS was increased (P = 0.001) and the SRT was attenuated (P < 0.0001). We had either significant deterioration due to the course of the disease or insignificant changes in the placebo group. Daily administration of curcumin, can significantly improve hearing impairment in CKD patients. Accordingly, curcumin should be considered as a therapeutic option for treating hearing impairment in patients with chronic kidney disease.


Subject(s)
Auditory Threshold , Curcumin , Renal Insufficiency, Chronic , Humans , Curcumin/therapeutic use , Male , Female , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/physiopathology , Middle Aged , Double-Blind Method , Auditory Threshold/drug effects , Aged , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss/drug therapy , Adult , Treatment Outcome
3.
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
4.
Int J Pediatr Otorhinolaryngol ; 181: 111981, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749259

ABSTRACT

OBJECTIVES: Obtaining perfect immobility or sleep in children undergoing ABR auditory brainstem response) testing can be challenging. We examined the effectiveness and safety of intranasal dexmedetomidine for sedation of children undergoing ABR testing. MATERIAL AND METHODS: We included prospectively all patients aged from 1 to 15 years for whom sedation for ABR testing was required, between July 2018 and November 2021. We administered an initial dose of 2.5 µg/kg intranasal dexmedetomidine with a repeat dose of 1 µg/kg if needed 30 min later. Collected data included success rate of sedation, sedation onset and recovery times and incidence of side effects. RESULTS: ABR testing was undertaken successfully in 57 of the 59 patients, giving a total success rate of 96,6 %. (95 % confidence interval 88.5 %-99.1 %). The median time to onset of sleep was 32 ± 18.3 min. The median duration of sedation recovery time was 48 ± 24.7 min. We recorded the adverse effects. Thirty-one patients experienced bradycardia and 28 patients experienced hypotension, all of which resolved without intervention. CONCLUSION: Intranasal dexmedetomidine is an effective, safe, simple of use and noninvasive method for sedation in children. It could have a major role in auditory brainstem response testing, specially in the case of non-cooperative children. REGISTRATION NUMBER OF THE TRIAL: NCT03530371.


Subject(s)
Administration, Intranasal , Dexmedetomidine , Evoked Potentials, Auditory, Brain Stem , Hypnotics and Sedatives , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Conscious Sedation/methods , Dexmedetomidine/administration & dosage , Evoked Potentials, Auditory, Brain Stem/drug effects , Hypnotics and Sedatives/administration & dosage , Prospective Studies
5.
Toxicol In Vitro ; 99: 105852, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38789064

ABSTRACT

Cisplatin is an effective chemotherapeutic agent; however, ototoxicity is one of its negative effects that greatly limits the use of cisplatin in clinical settings. Previous research has shown that the most important process cisplatin damage to inner ear cells, such as hair cells (HCs), is the excessive production and accumulation of ROS. Schisandrin B (SchB), is a low-toxicity, inexpensive, naturally occurring antioxidant with a variety of pharmacological effects. Therefore, the potential antioxidant effects of SchB may be useful for cisplatin ototoxicity treatment. In this study, the effects of SchB on cochlear hair cell viability, ROS levels, and expression of apoptosis-related molecules were evaluated by CCK-8, immunofluorescence, flow cytometry, and qRT-PCR, as well as auditory brainstem response (ABR) and dysmorphic product otoacoustic emission (DPOAE) tests to assess the effects on inner ear function. The results showed that SchB treatment increased cell survival, prevented apoptosis, and reduced cisplatin-induced ROS formation. SchB treatment reduced the loss of cochlear HCs caused by cisplatin in exosome culture. In addition, SchB treatment attenuated cisplatin-induced hearing loss and HC loss in mice. This study demonstrates the ability of SchB to inhibit cochlear hair cell apoptosis and ROS generation and shows its potential therapeutic effect on cisplatin ototoxicity.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Survival , Cisplatin , Cyclooctanes , Hair Cells, Auditory, Inner , Lignans , Oxidative Stress , Polycyclic Compounds , Reactive Oxygen Species , Cisplatin/toxicity , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Polycyclic Compounds/toxicity , Animals , Apoptosis/drug effects , Lignans/pharmacology , Oxidative Stress/drug effects , Antineoplastic Agents/toxicity , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Hair Cells, Auditory, Inner/drug effects , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Antioxidants/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Male , Ototoxicity/prevention & control
6.
Hear Res ; 446: 109004, 2024 May.
Article in English | MEDLINE | ID: mdl-38608332

ABSTRACT

The naturally occurring amino acid, l-ergothioneine (EGT), has immense potential as a therapeutic, having shown promise in the treatment of other disease models, including neurological disorders. EGT is naturally uptaken into cells via its specific receptor, OCTN1, to be utilized by cells as an antioxidant and anti-inflammatory. In our current study, EGT was administered over a period of 6 months to 25-26-month-old CBA/CaJ mice as a possible treatment for age-related hearing loss (ARHL), since presbycusis has been linked to higher levels of cochlear oxidative stress, apoptosis, and chronic inflammation. Results from the current study indicate that EGT can prevent aging declines of some key features of ARHL. However, we found a distinct sex difference for the response to the treatments, for hearing - Auditory Brainstem Responses (ABRs) and Distortion Product Otoacoustic Emissions (DPOAEs). Males exhibited lower threshold declines in both low dose (LD) and high dose (HD) test groups throughout the testing period and did not display some of the characteristic aging declines in hearing seen in Control animals. In contrast, female mice did not show any therapeutic effects with either treatment dose. Further confirming this sex difference, EGT levels in whole blood sampling throughout the testing period showed greater uptake of EGT in males compared to females. Additionally, RT-PCR results from three tissue types of the inner ear confirmed EGT activity in the cochlea in both males and females. Males and females exhibited significant differences in biomarkers related to apoptosis (Cas-3), inflammation (TNF-a), oxidative stress (SOD2), and mitochondrial health (PGC1a).These changes were more prominent in males as compared to females, especially in stria vascularis tissue. Taken together, these findings suggest that EGT has the potential to be a naturally derived therapeutic for slowing down the progression of ARHL, and possibly other neurodegenerative diseases. EGT, while effective in the treatment of some features of presbycusis in aging males, could also be modified into a general prophylaxis for other age-related disorders where treatment protocols would include eating a larger proportion of EGT-rich foods or supplements. Lastly, the sex difference discovered here, needs further investigation to see if therapeutic conditions can be developed where aging females show better responsiveness to EGT.


Subject(s)
Aging , Antioxidants , Cochlea , Disease Models, Animal , Disease Progression , Ergothioneine , Evoked Potentials, Auditory, Brain Stem , Mice, Inbred CBA , Oxidative Stress , Presbycusis , Animals , Ergothioneine/pharmacology , Female , Evoked Potentials, Auditory, Brain Stem/drug effects , Male , Presbycusis/physiopathology , Presbycusis/pathology , Presbycusis/drug therapy , Presbycusis/metabolism , Presbycusis/prevention & control , Oxidative Stress/drug effects , Aging/drug effects , Aging/pathology , Antioxidants/pharmacology , Sex Factors , Cochlea/drug effects , Cochlea/metabolism , Cochlea/physiopathology , Cochlea/pathology , Age Factors , Apoptosis/drug effects , Otoacoustic Emissions, Spontaneous/drug effects , Superoxide Dismutase/metabolism , Auditory Threshold/drug effects , Hearing/drug effects , Mice , Anti-Inflammatory Agents/pharmacology
7.
Otolaryngol Head Neck Surg ; 171(2): 530-537, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38545636

ABSTRACT

OBJECTIVE: The objective was to determine the effects of older age on hearing preservation after cochlear implantation (CI), and whether steroids improve hearing preservation in older animals. We hypothesized greater hearing preservation would be observed in (1) young animals compared to older animals and (2) older animals receiving steroids compared to no steroids. The secondary objective was to assess levels of fibrosis utilizing optical coherence tomography (OCT). STUDY DESIGN: Experimental Animal Study. SETTING: Laboratory. METHODS: Three groups of guinea pigs: young (YCI; 8.5 ± 0.5 weeks; n = 10), old (OCI; 19.1 ± 1.0 months; n = 9) and old + steroids (OCI+S; 19.1 ± 1.0 months; n = 9) underwent CI. The OCI+S group received a steroid taper over 7 days starting 2 days before surgery to 4 days after. Auditory brainstem response (ABR) measurements were performed preoperatively and postoperatively. OCT imaging was performed to assess cochleae for extent of fibrotic tissue growth in the scala tympani. RESULTS: The YCI group had significantly better hearing preservation as measured by smaller increases in ABR thresholds [mean shift: 2.79 ± 0.66] compared to the OCI group [mean shift = 12.44 ± 5.6]. The OCI+S group had significantly better hearing preservation [2.66 ± 1.50] compared to the OCI group. No significant differences was seen in fibrosis across groups. CONCLUSIONS: Young animals and older animals that received steroids had better hearing after CI than older animals not given steroids, but hearing preservation was not correlated with the level of fibrosis assessed using OCT. This work is the first to investigate differences in hearing preservation by age in an animal model, and supports the protective effects of steroids on hearing preservation in older individuals.


Subject(s)
Aging , Cochlea , Cochlear Implantation , Tomography, Optical Coherence , Animals , Guinea Pigs , Cochlea/drug effects , Aging/physiology , Evoked Potentials, Auditory, Brain Stem/drug effects , Glucocorticoids/pharmacology , Disease Models, Animal , Fibrosis , Age Factors
8.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884516

ABSTRACT

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


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Kanamycin/toxicity , Ototoxicity/drug therapy , PPAR gamma/metabolism , Telmisartan/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Animals , Anti-Bacterial Agents/toxicity , Antihypertensive Agents/pharmacology , Auditory Threshold/drug effects , Cochlea/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Heme Oxygenase (Decyclizing)/genetics , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , PPAR gamma/genetics , Rats , Rats, Sprague-Dawley
9.
Pak J Pharm Sci ; 34(3): 843-854, 2021 May.
Article in English | MEDLINE | ID: mdl-34602405

ABSTRACT

The aim of research is to unveil the mechanisms of the beneficial effects of XYD on PCIV in a rabbit model. 40 New Zealand white rabbits were randomly divided into 5 groups,including normal control group (NC), model control group (MC), low-dose of XYD group (LXYD), high-dose of XYD group (HXYD) and Yang-Xue-Qin-Nao group (YXQN). PCIV rabbit model was established by feeding high-fat diet companied with paravertebral sclerotherapy and rotation exercise. The general observation, step-down test, rheoencephalogram, blood tests, histopathological detection and the plasma concentration of the effective component of XYD were investigated. After pharmacological intervening, the step-down time, REG, PL, IPL, blood viscosity, the levels of blood lipids, CRGP were significantly improved. Moreover, the vertebral artery showed the reduced stenosis of arterial lumen and less proliferation of fibrous tissue in the arterial wall in the LXYD, HXYD and YXQN group. Based on the LC-MS detection, the blood concentrations of puerarin in the LXYD and HXYD group were significantly increased after pharmacological intervening. XYD could ameliorate the symptoms of vertigo, Qi-deficiency and blood stasis in PCIV rabbits via effectively regulating the levels of blood lipids and vasoactive substances, decreasing blood viscosity, increasing CBF and protecting vestibular function.


Subject(s)
Behavior, Animal/drug effects , Drugs, Chinese Herbal/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Vertebral Artery/drug effects , Vertebrobasilar Insufficiency/physiopathology , Vertigo/physiopathology , Vestibular Nuclei/drug effects , Animals , Disease Models, Animal , Hemorheology , Lipid Metabolism/drug effects , Medicine, Chinese Traditional , Rabbits , Vertebral Artery/pathology , Vertebral Artery/ultrastructure , Vestibular Nuclei/pathology , Vestibular Nuclei/ultrastructure
10.
J Neurophysiol ; 126(5): 1660-1669, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34644166

ABSTRACT

Studies of in vivo neuronal responses to auditory inputs in the superior olive complex (SOC) are usually done under anesthesia. However, little attention has been paid to the effect of anesthesia itself on response properties. Here, we assessed the effect of anesthesia depth under ketamine-xylazine anesthetics on auditory evoked response properties of lateral SOC neurons. Anesthesia depth was tracked by monitoring EEG spectral peak frequencies. An increase in anesthesia depth led to a decrease of spontaneous discharge activities and an elevated response threshold. The temporal responses to suprathreshold tones were also affected, with adapted responses reduced but peak responses unaffected. Deepening the anesthesia depth also increased first spike latency. However, spike jitter was not affected. Auditory brainstem responses to clicks confirmed that ketamine-xylazine anesthesia depth affects auditory neuronal activities and the effect on spike rate and spike timing persists through the auditory pathway. We concluded from those observations that ketamine-xylazine affects lateral SOC response properties depending on the anesthesia depth.NEW & NOTEWORTHY We studied how the depth of ketamine-xylazine anesthesia altered response properties of lateral superior olive complex neurons, and auditory brainstem evoked responses. Our results provide direct evidence that anesthesia depth affects auditory neuronal responses and reinforce the notion that both the anesthetics and the anesthesia depth should be considered when interpreting/comparing in vivo neuronal recordings.


Subject(s)
Anesthesia , Anesthetics, General/pharmacology , Auditory Perception/drug effects , Ketamine/pharmacology , Superior Olivary Complex/drug effects , Xylazine/pharmacology , Animals , Electroencephalography/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Gerbillinae , Male
11.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576224

ABSTRACT

Delivery of substances into the inner ear via local routes is increasingly being used in clinical treatment. Studies have focused on methods to increase permeability through the round window membrane (RWM) and enhance drug diffusion into the inner ear. However, the clinical applications of those methods have been unclear and few studies have investigated the efficacy of methods in an inner ear injury model. Here, we employed the medium chain fatty acid caprate, a biologically safe, clinically applicable substance, to modulate tight junctions of the RWM. Intratympanic treatment of sodium caprate (SC) induced transient, but wider, gaps in intercellular spaces of the RWM epithelial layer and enhanced the perilymph and cochlear concentrations/uptake of dexamethasone. Importantly, dexamethasone co-administered with SC led to significantly more rapid recovery from noise-induced hearing loss at 4 and 8 kHz, compared with the dexamethasone-only group. Taken together, our data indicate that junctional modulation of the RWM by SC enhances dexamethasone uptake into the inner ear, thereby hastening the recovery of hearing sensitivity after noise trauma.


Subject(s)
Dexamethasone/pharmacokinetics , Ear, Inner/drug effects , Hearing Loss, Noise-Induced/drug therapy , Round Window, Ear/drug effects , Animals , Cochlea/drug effects , Decanoic Acids/pharmacology , Dexamethasone/administration & dosage , Diffusion , Drug Delivery Systems/methods , Evoked Potentials, Auditory, Brain Stem/drug effects , Fatty Acids/chemistry , Hearing , Male , Microscopy, Electron, Transmission , Perilymph/drug effects , Permeability , Rats
12.
Mar Drugs ; 19(8)2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34436282

ABSTRACT

One of the well-known causes of hearing loss is noise. Approximately 31.1% of Americans between the ages of 20 and 69 years (61.1 million people) have high-frequency hearing loss associated with noise exposure. In addition, recurrent noise exposure can accelerate age-related hearing loss. Phlorofucofuroeckol A (PFF-A) and dieckol, polyphenols extracted from the brown alga Ecklonia cava, are potent antioxidant agents. In this study, we investigated the effect of PFF-A and dieckol on the consequences of noise exposure in mice. In 1,1-diphenyl-2-picrylhydrazyl assay, dieckol and PFF-A both showed significant radical-scavenging activity. The mice were exposed to 115 dB SPL of noise one single time for 2 h. Auditory brainstem response(ABR) threshold shifts 4 h after 4 kHz noise exposure in mice that received dieckol were significantly lower than those in the saline with noise group. The high-PFF-A group showed a lower threshold shift at click and 16 kHz 1 day after noise exposure than the control group. The high-PFF-A group also showed higher hair cell survival than in the control at 3 days after exposure in the apical turn. These results suggest that noise-induced hair cell damage in cochlear and the ABR threshold shift can be alleviated by dieckol and PFF-A in the mouse. Derivatives of these compounds may be applied to individuals who are inevitably exposed to noise, contributing to the prevention of noise-induced hearing loss with a low probability of adverse effects.


Subject(s)
Antioxidants/therapeutic use , Benzofurans/therapeutic use , Dioxins/therapeutic use , Hearing Loss, Noise-Induced/drug therapy , Kelp , Plant Extracts/therapeutic use , Animals , Antioxidants/pharmacology , Aquatic Organisms , Benzofurans/pharmacology , Cochlea/drug effects , Dioxins/pharmacology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory/drug effects , Male , Mice , Mice, Inbred C57BL , Phytotherapy , Plant Extracts/pharmacology
13.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34360766

ABSTRACT

Age-related hearing loss (ARHL) is the most common sensory disorder among older people, and yet, the treatment options are limited to medical devices such as hearing aids and cochlear implants. The high prevalence of ARHL mandates the development of treatment strategies that can prevent or rescue age-related cochlear degeneration. In this study, we investigated a novel pharmacological strategy based on inhibition of the adenosine A2A receptor (A2AR) in middle aged C57BL/6 mice prone to early onset ARHL. C57BL/6J mice were treated with weekly istradefylline (A2AR antagonist; 1 mg/kg) injections from 6 to 12 months of age. Auditory function was assessed using auditory brainstem responses (ABR) to tone pips (4-32 kHz). ABR thresholds and suprathreshold responses (wave I amplitudes and latencies) were evaluated at 6, 9, and 12 months of age. Functional outcomes were correlated with quantitative histological assessments of sensory hair cells. Cognitive function was assessed using the Morris water maze and the novel object recognition test, and the zero maze test was used to assess anxiety-like behaviour. Weekly injections of istradefylline attenuated ABR threshold shifts by approximately 20 dB at mid to high frequencies (16-32 kHz) but did not improve ABR suprathreshold responses. Istradefylline treatment improved hair cell survival in a turn-dependent manner, whilst the cognitive function was unaffected by istradefylline treatment. This study presents the first evidence for the rescue potential of istradefylline in ARHL and highlights the role of A2AR in development of age-related cochlear degeneration.


Subject(s)
Aging , Auditory Threshold/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Presbycusis , Purines/pharmacology , Animals , Male , Mice , Presbycusis/drug therapy , Presbycusis/pathology , Presbycusis/physiopathology
14.
Int J Immunopathol Pharmacol ; 35: 20587384211034086, 2021.
Article in English | MEDLINE | ID: mdl-34344210

ABSTRACT

INTRODUCTION: Cruciferous vegetables are a rich source of sulforaphane (SFN), which acts as a natural HDAC inhibitor (HDACi). Our previous study found that HDACi could restore histone acetyltransferase/histone deacetylase (HAT/HDAC) balance in the cochlea and attenuate gentamicin-induced hearing loss in guinea pigs. Here, we investigated the protective effect of SFN on cisplatin-induced hearing loss (CIHL). METHODS: Thirty rats were randomly divided into 3 equal groups: the control group, cisplatin group, and SFN+cisplatin group. Rats were injected with SFN (30 mg/kg once a day) and cisplatin (7 mg/kg twice a day) for 7 days to investigate the protective role of SFN on CIHL. We observed auditory brainstem response (ABR) threshold shifts and immunostained cochlear basilar membranes of rats. For in vitro experiments, we treated HEI-OC1 cells and rat cochlear organotypic cultures with SFN (5, 10, and 15 µM) and cisplatin (10 µM). Immunofluorescence, cell viability, and protein analysis were performed to further analyze the protective mechanism of SFN on CIHL. RESULTS: SFN (30 mg/kg once a day) decreased cisplatin (7 mg/kg twice a day)-induced ABR threshold shifts and outer hair cell loss. CCK-8 assay showed that cisplatin (10 µM) reduced the viability of HEI-OC1 cells to 42%, and SFN had a dose-dependent protective effect. In cochlear organotypic cultures, we found that SFN (10 and 15 µM) increased cisplatin (10 µM)-induced myosin 7a+ cell count and restored ciliary morphology. SFN (5, 10, and 15 µM) reversed the cisplatin (10 µM)-induced increase in HDAC2, -4, and -5 and SFN (15 µM) reversed the cisplatin (10 µM)-induced decrease in H3-Ack9 [acetyl-histone H3 (Lys9)] protein expression in HEI-OC1 cells. Neither cisplatin nor cisplatin combined with SFN affected the expression of HDAC7, or HDAC9. CONCLUSION: SFN prevented disruption of the HAT/HDAC balance, protecting against CIHL in rats.


Subject(s)
Antineoplastic Agents , Cisplatin , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Isothiocyanates/therapeutic use , Sulfoxides/therapeutic use , Animals , Cell Count , Cilia/pathology , Cochlea/pathology , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory, Outer/pathology , Histone Deacetylases/biosynthesis , Histone Deacetylases/drug effects , Histone Deacetylases/genetics , Rats , Rats, Wistar
15.
Drug Des Devel Ther ; 15: 3443-3450, 2021.
Article in English | MEDLINE | ID: mdl-34413631

ABSTRACT

OBJECTIVE: To investigate the hearing protection outcomes of different drug-eluting analog electrode arrays implanted into guinea pig cochleae. METHODS: Sixty guinea pigs were randomly divided into a negative control group and five experimental groups implanted separately with blank (drug carrier), dexamethasone (DXM), aracytine (Ara-C), Ara-C+DXM, and nicotinamide adenine dinucleotide (NAD+) eluting analog electrode arrays. Micro CT was used to supervise the surgical procedure. Auditory brainstem response (ABR) thresholds of the guinea pigs were measured and analyzed. RESULTS AND CONCLUSIONS: Compared with the negative control, all other groups showed a significant increase in ABR threshold (p<0.001) after surgery. Among them, there was no obvious difference between the blank (0 vs 90 days: 59.70±10.57 vs 64.60±9.47 dB SPL) and the NAD+ group (0 vs 90 days: 59.90±9.87 vs 64.70±8.65 dB SPL). On the other hand, the ABR thresholds in the DXM (0 days: 58.10±10.73 dB SPL; 90 days: 51.70±9.07 dB SPL) and the Ara-C group (0 days: 59.00±10.05 dB SPL; 90 days: 51.60±8.48 dB SPL) decreased significantly compared with the former two groups (p<0.001). However, the Ara-C+DXM group showed no further benefit (p>0.05). In addition, a significantly higher survival rate of spiral ganglion neurons in cochleae was observed in the Ara-C and/or DXM groups.


Subject(s)
Cochlear Implants , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss/prevention & control , Animals , Cytarabine/administration & dosage , Dexamethasone/administration & dosage , Electrodes, Implanted , Female , Guinea Pigs , NAD/administration & dosage , Polymers/chemistry
16.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070066

ABSTRACT

Megalin has been proposed as an endocytic receptor for aminoglycosides as well as estrogen and androgen. We aimed to investigate the otoprotective effects of antiandrogens (flutamide, FM) on kanamycin (KM)-induced hearing loss in rats. Rats were divided into four groups. The KM group was administered KM (20 mg/kg/day) for 5 days, while the FM group received FM (15 mg/kg/day) for 10 days. In the KM + FM group, KM and FM (15 mg/kg/day) were simultaneously injected for 5 days and then FM was injected for 5 days. Auditory brainstem responses were measured. Western blotting and/or quantitative reverse transcriptase-polymerase chain reaction were performed for megalin, cytochrome P450 1A1 (Cyp1a1), Cyp1b1, metallothionein 1A (MT1A), MT2A, tumor necrosis factor (TNF)-α, caspase 3, and cleaved caspase 3. The FM + KM group showed attenuated auditory thresholds when compared with the KM group at 4, 8, 16, and 32 kHz (all p < 0.05). The KM + FM group showed lower megalin and Cyp1b1 levels than the KM group (all p < 0.05). The KM + FM group revealed lower MT1A, TNFα, and caspase 3 protein levels, compared with those in the KM group (all p < 0.05). Androgen receptor inhibition protects against cochlear injuries in KM-induced hearing loss rats by attenuating megalin expression, revealing anti-inflammatory and anti-apoptotic effects.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Hearing Loss, Sensorineural/prevention & control , Animals , Anti-Bacterial Agents/toxicity , Auditory Threshold/drug effects , Cochlea/drug effects , Cochlea/pathology , Cochlea/physiopathology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Evoked Potentials, Auditory, Brain Stem/drug effects , Flutamide/pharmacology , Gene Expression/drug effects , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/physiopathology , Kanamycin/toxicity , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Metallothionein/genetics , Metallothionein/metabolism , Protective Agents/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
17.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799684

ABSTRACT

Age-related hearing loss (ARHL), a sensorineural hearing loss of multifactorial origin, increases its prevalence in aging societies. Besides hearing aids and cochlear implants, there is no FDA approved efficient pharmacotherapy to either cure or prevent ARHL. We hypothesized that selegiline, an antiparkinsonian drug, could be a promising candidate for the treatment due to its complex neuroprotective, antioxidant, antiapoptotic, and dopaminergic neurotransmission enhancing effects. We monitored by repeated Auditory Brainstem Response (ABR) measurements the effect of chronic per os selegiline administration on the hearing function in BALB/c and DBA/2J mice, which strains exhibit moderate and rapid progressive high frequency hearing loss, respectively. The treatments were started at 1 month of age and lasted until almost a year and 5 months of age, respectively. In BALB/c mice, 4 mg/kg selegiline significantly mitigated the progression of ARHL at higher frequencies. Used in a wide dose range (0.15-45 mg/kg), selegiline had no effect in DBA/2J mice. Our results suggest that selegiline can partially preserve the hearing in certain forms of ARHL by alleviating its development. It might also be otoprotective in other mammals or humans.


Subject(s)
Aging/physiology , Disease Models, Animal , Hearing Loss, Sensorineural/drug therapy , Selegiline/pharmacology , Administration, Oral , Animals , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Auditory Threshold/drug effects , Auditory Threshold/physiology , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Sensorineural/physiopathology , Humans , Male , Mice, Inbred BALB C , Mice, Inbred DBA , Protective Agents/administration & dosage , Protective Agents/pharmacology , Selegiline/administration & dosage , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
18.
Front Neural Circuits ; 15: 597401, 2021.
Article in English | MEDLINE | ID: mdl-33679335

ABSTRACT

Systemic nicotine enhances neural processing in primary auditory cortex (A1) as determined using tone-evoked, current-source density (CSD) measurements. For example, nicotine enhances the characteristic frequency (CF)-evoked current sink in layer 4 of A1, increasing amplitude and decreasing latency. However, since presenting auditory stimuli within a stream of stimuli increases the complexity of response dynamics, we sought to determine the effects of nicotine on CSD responses to trains of CF stimuli (one-second trains at 2-40 Hz; each train repeated 25 times). CSD recordings were obtained using a 16-channel multiprobe inserted in A1 of urethane/xylazine-anesthetized mice, and analysis focused on two current sinks in the middle (layer 4) and deep (layers 5/6) layers. CF trains produced adaptation of the layer 4 response that was weak at 2 Hz, stronger at 5-10 Hz and complete at 20-40 Hz. In contrast, the layer 5/6 current sink exhibited less adaptation at 2-10 Hz, and simultaneously recorded auditory brainstem responses (ABRs) showed no adaptation even at 40 Hz. Systemic nicotine (2.1 mg/kg) enhanced layer 4 responses throughout the one-second stimulus train at rates ≤10 Hz. Nicotine enhanced both response amplitude within each train and the consistency of response timing across 25 trials. Nicotine did not alter the degree of adaptation over one-second trials, but its effect to increase amplitudes revealed a novel, slower form of adaptation that developed over multiple trials. Nicotine did not affect responses that were fully adapted (20-40 Hz trains), nor did nicotine affect any aspect of the layer 5/6 current sink or ABRs. The overall effect of nicotine in layer 4 was to enhance all responses within each train, to emphasize earlier trials across multiple trials, and to improve the consistency of timing across all trials. These effects may improve processing of complex acoustic streams, including speech, that contain information in the 2-10 Hz range.


Subject(s)
Auditory Cortex/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory/drug effects , Nicotine/pharmacology , Acoustic Stimulation/methods , Acoustics , Animals , Auditory Cortex/physiology , Evoked Potentials, Auditory/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Male , Mice , Time Factors
19.
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
20.
Hear Res ; 404: 108204, 2021 05.
Article in English | MEDLINE | ID: mdl-33618164

ABSTRACT

Various methods have been tested and deployed clinically to identify and minimize cisplatin ototoxicity. Upon early identification of hearing loss, one of the possible approaches to reducing future ototoxicity is to increase the gaps or breaks between cycles or doses of cisplatin. However, recent findings about the retention of cisplatin in the cochlea and the potential for its long-term ototoxic effects call into question whether such an approach is effective in reducing hearing loss. The current study was undertaken to determine whether increasing the rest intervals between cycles of cisplatin altered the resulting ototoxicity. CBA/CaJ mice were exposed to a cumulative dose of 48 mg/kg cisplatin delivered in three cycles of 16 mg/kg (4 mg/kg per day for 4 consecutive days). The cycles were separated by either 10, 17, or 87 days to determine if the inter-cycle rest intervals affected resulting ototoxicity. Ototoxicity was measured using auditory brainstem response threshold shifts and hair cell losses. Results indicated that longer intervals between cycles of cisplatin led to lower threshold shifts and outer hair cell lesions. The results support the principle that 'slowing down' cisplatin dosing by increasing rest intervals between doses can reduce the ototoxic side effect. Further testing is needed to optimize the timing and to determine the impact of longer inter-cycle intervals on cisplatin's anti-tumor efficacy.


Subject(s)
Hearing Loss , Ototoxicity , Animals , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Mice , Mice, Inbred CBA
SELECTION OF CITATIONS
SEARCH DETAIL