Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 333
Filter
1.
Sci Rep ; 14(1): 17576, 2024 Jul 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
2.
Sci Rep ; 14(1): 13376, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38862572

ABSTRACT

In individuals with hearing loss, protection of residual hearing is essential following cochlear implantation to facilitate acoustic and electric hearing. Hearing preservation requires slow insertion, atraumatic electrode and delivery of the optimal quantity of a pharmacological agent. Several studies have reported variable hearing outcomes with osmotic pump-mediated steroid delivery. New drugs, such as sialyllactose (SL) which have anti-inflammatory effect in many body parts, can prevent tissue overgrowth. In the present study, the positive effects of the pharmacological agent SL against insults were evaluated in vitro using HEI-OC1 cells. An animal model to simulate the damage due to electrode insertion during cochlear implantation was used. SL was delivered using osmotic pumps to prevent loss of the residual hearing in this animal model. Hearing deterioration, tissue fibrosis and ossification were confirmed in this animal model. Increased gene expressions of inflammatory cytokines were identified in the cochleae following dummy electrode insertion. Following the administration of SL, insertion led to a decrease in hearing threshold shifts, tissue reactions, and inflammatory markers. These results emphasize the possible role of SL in hearing preservation and improve our understanding of the mechanism underlying hearing loss after cochlear implantation.


Subject(s)
Cochlear Implantation , Hearing Loss , Lactose , Animals , Lactose/analogs & derivatives , Lactose/pharmacology , Hearing Loss/prevention & control , Hearing Loss/drug therapy , Hearing/drug effects , Cochlea/drug effects , Cochlea/metabolism , Mice , Disease Models, Animal , Cell Line , Cytokines/metabolism , Male , Sialic Acids
3.
Eur J Med Chem ; 271: 116404, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38631262

ABSTRACT

Hearing loss (HL) is a health burden that seriously affects the quality of life of cancer patients receiving platinum-based chemotherapy, and few FDA-approved treatment specifically targets this condition. The main mechanisms that contribute to cisplatin-induced hearing loss are oxidative stress and subsequent cell death, including ferroptosis revealed by us as a new mechanism recently. In this study, we employed the frontier molecular orbital (FMO) theory approach as a convenient prediction method for the glutathione peroxidase (GPx)-like activity of isoselenazolones and discovered new isoselenazolones with great GPx-like activity. Notably, compound 19 exhibited significant protective effects against cisplatin-induced hair cell (HC) damage in vitro and in vivo and effectively reverses cisplatin-induced hearing loss through oral administration. Further investigations revealed that this compound effectively alleviated hair cell oxidative stress, apoptosis and ferroptosis. This research highlights the potential of GPx mimics as a therapeutic strategy against cisplatin-induced hearing loss. The application of quantum chemistry (QC) calculations in the study of GPx mimics sheds light on the development of new, innovative treatments for hearing loss.


Subject(s)
Cisplatin , Glutathione Peroxidase , Hearing Loss , Cisplatin/pharmacology , Glutathione Peroxidase/metabolism , Animals , Hearing Loss/drug therapy , Hearing Loss/chemically induced , Humans , Quantum Theory , Molecular Structure , Mice , Structure-Activity Relationship , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Oxidative Stress/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Discovery , Dose-Response Relationship, Drug , Apoptosis/drug effects
4.
Adv Healthc Mater ; 13(16): e2303149, 2024 06.
Article in English | MEDLINE | ID: mdl-38514042

ABSTRACT

Inner ear delivery requires safe and effective drug delivery vehicles incorporating high-viscosity formulations with permeation enhancers. This study designs novel thermoresponsive-smart polymer-bile acid and cyclodextrin-based nanogels for inner ear delivery. Nanogels are examined for their rheological and physical properties. The biocompatibility studies will be assessed on auditory and macrophage cell lines by investigating the impact of nanogels on cellular viability, mitochondrial respiration, glycolysis, intracellular oxidative stress, inflammatory profile, and macrophage polarization. Novel ther nanogels based on bile acid and beta-cyclodextrin show preserved porous nanogels' inner structure, exhibit non-Newtonian, shear-thinning fluid behavior, have fast gelation at 37 °C and minimal albumin adsorption on the surface. The nanogels have minimal impact on cellular viability, mitochondrial respiration, glycolysis, intracellular oxidative stress, and inflammatory profile of the auditory cell line House Ear Institute-Organ of Corti 1 after 24 h incubation. Nanogel exposure of 24 h to macrophage cell line RAW264.7 leads to decreased viability, mitochondrial dysfunction, and increased intracellular ROS and inflammatory cytokines. However, polarization changes from M2 anti-inflammatory to M1 pro-inflammatory macrophages are minimal, and inflammatory products of RAW264.7 macrophages do not overly disrupt the survivability of HEI-OC1 cells. Based on these results, thermoresponsive bile acid and cyclodextrin nanogels can be potential drug delivery vehicles for inner ear drug delivery.


Subject(s)
Hearing Loss , Nanogels , Animals , Mice , RAW 264.7 Cells , Hearing Loss/drug therapy , Nanogels/chemistry , Bile Acids and Salts/chemistry , Cell Survival/drug effects , Cyclodextrins/chemistry , Polyethylene Glycols/chemistry , Drug Delivery Systems/methods , Macrophages/metabolism , Macrophages/drug effects , Cell Line , Polyethyleneimine
5.
Colloids Surf B Biointerfaces ; 237: 113855, 2024 May.
Article in English | MEDLINE | ID: mdl-38513298

ABSTRACT

Local drug delivery has been exploited recently to treat hearing loss, as this method can both bypass the blood-labyrinth barrier and provide sustained drug release. Combined drug microcrystals (MCs) offer additional advantages for sensorineural hearing loss treatment via intratympanic (IT) injection due to their shape effect and combination strategy. In this study, to endow viscous effects of hydrogels, nonspherical dexamethasone (DEX) and lipoic acid (LA) MCs were incorporated into silk fibroin (SF) hydrogels, which were subsequently administered to the tympanic cavity to investigate their pharmaceutical properties. First, we prepared DEX and LA MCs by a traditional precipitation technique followed by SF hydrogel incorporation (SF+DEX+LA). After characterization of the physicochemical features, including morphology, rheology, and dissolution, both a suspension of combined DEX and LA MCs (DEX+LA) and SF+DEX+LA were administered to guinea pigs by IT injection, after which the pharmacokinetics, biodegradation and biocompatibility were evaluated. To our surprise, compared to the DEX+LA group, the pharmacokinetics of the SF+DEX+LA hydrogel group did not improve significantly, which may be ascribed to their nonspherical shape and deposition effects of the drugs MCs. The cochlear tissue in each group displayed good morphology, with no obvious inflammatory reactions. This combined MC suspension has the clear advantages of no vehicle, easy scale-up preparation, and good biocompatibility and outcomes, which paves the way for practical treatment of hearing loss via local drug delivery.


Subject(s)
Ear, Inner , Fibroins , Hearing Loss , Thioctic Acid , Animals , Guinea Pigs , Hydrogels/chemistry , Thioctic Acid/pharmacology , Dexamethasone , Silk/metabolism , Ear, Inner/metabolism , Hearing Loss/drug therapy , Hearing Loss/metabolism , Fibroins/pharmacology
6.
Calcif Tissue Int ; 114(3): 255-266, 2024 03.
Article in English | MEDLINE | ID: mdl-38226986

ABSTRACT

X-linked hypophosphatemia (XLH) is the most common monogenetic cause of chronic hypophosphatemia, characterized by rickets and osteomalacia. Disease manifestations and treatment of XLH patients in the Netherlands are currently unknown. Characteristics of XLH patients participating in the Dutch observational registry for genetic hypophosphatemia and acquired renal phosphate wasting were analyzed. Eighty XLH patients, including 29 children, were included. Genetic testing, performed in 78.8% of patients, showed a PHEX mutation in 96.8%. Median (range) Z-score for height was - 2.5 (- 5.5; 1.0) in adults and - 1.4 (- 3.7; 1.0) in children. Many patients were overweight or obese: 64.3% of adults and 37.0% of children. All children received XLH-related medication e.g., active vitamin D, phosphate supplementation or burosumab, while 8 adults used no medication. Lower age at start of XLH-related treatment was associated with higher height at inclusion. Hearing loss was reported in 6.9% of children and 31.4% of adults. Knee deformities were observed in 75.0% of all patients and osteoarthritis in 51.0% of adult patients. Nephrocalcinosis was observed in 62.1% of children and 33.3% of adults. Earlier start of XLH-related treatment was associated with higher risk of nephrocalcinosis and detection at younger age. Hyperparathyroidism longer than six months was reported in 37.9% of children and 35.3% of adults. This nationwide study confirms the high prevalence of adiposity, hearing loss, bone deformities, osteoarthritis, nephrocalcinosis and hyperparathyroidism in Dutch XLH patients. Early start of XLH-related treatment appears to be beneficial for longitudinal growth but may increase development of nephrocalcinosis.


Subject(s)
Familial Hypophosphatemic Rickets , Hearing Loss , Hyperparathyroidism , Hypophosphatemia , Nephrocalcinosis , Osteoarthritis , Child , Adult , Humans , Familial Hypophosphatemic Rickets/complications , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/diagnosis , Nephrocalcinosis/genetics , Nephrocalcinosis/complications , Fibroblast Growth Factors/genetics , Hypophosphatemia/epidemiology , Hypophosphatemia/genetics , Phosphates , Hyperparathyroidism/complications , Obesity/complications , Hearing Loss/complications , Hearing Loss/drug therapy
7.
J Pharm Pharmacol ; 76(4): 295-306, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38206827

ABSTRACT

OBJECTIVES: Biochemical alterations due to diabetes development and progress are complex and diabetes-associated injury to various tissues has been well reported. Nevertheless, a close investigation of the literature demonstrates limited coverage regarding these biochemical and molecular alterations within the inner ear and their impact on the vestibulocochlear environment. A closer look at these may reveal pharmacological targets that could alleviate the severity of disease in patients. KEY FINDINGS: Tight control of glucose levels within the highly metabolic inner ear structures is crucial for their physiology and function. Impaired glucose homeostasis is well known to occur in vestibulocochlear malfunctioning. Moreover, the involvement of insulin signalling, and glucose transporters were recently confirmed in vestibulocochlear structures and are believed to play a crucial role in auditory and vestibular functions. CONCLUSION: Oxidative overload, glucolipotoxicity, perturbed blood rheology, endothelial dysfunction, compromised microvascular supply, and neurotoxicity are reported in many diabetic complications such as nephropathy, retinopathy, and diabetic neuropathy and are incriminated in the disruption of blood labyrinth barrier as well as vestibulocochlear neuritis. Dysfunctional insulin signalling was recently reported in the Organ of Corti. Insulin resistance in the inner ear niche warrants further studies to verify and uncover new pharmacological targets to manage this debilitating condition better.


Subject(s)
Diabetes Mellitus , Ear, Inner , Hearing Loss , Insulins , Humans , Ear, Inner/metabolism , Hearing Loss/drug therapy , Hearing Loss/etiology , Hearing Loss/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Insulins/metabolism , Glucose/metabolism
8.
Sci Rep ; 14(1): 183, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167873

ABSTRACT

The increased risk of hearing loss with macrolides remains controversial. We aimed to systematically review and meta-analyze data on the clinical risk of hearing loss, tinnitus, and ototoxicity following macrolide use. A systematic search was conducted across PubMed, MEDLINE, Cochrane, and Embase databases from database inception to May 2023. Medical Subject Heading (MeSH) terms and text keywords were utilized, without any language restrictions. In addition to the electronic databases, two authors manually and independently searched for relevant studies in the US and European clinical trial registries and Google Scholar. Studies that involved (1) patients who had hearing loss, tinnitus, or ototoxicity after macrolide use, (2) intervention of use of macrolides such as azithromycin, clarithromycin, erythromycin, fidaxomicin, roxithromycin, spiramycin, and/or telithromycin, (3) comparisons with specified placebos or other antibiotics, (4) outcomes measured as odds ratio (OR), relative risk (RR), hazard ratio (HR), and mean difference for ototoxicity symptoms using randomized control trial (RCT)s and observational studies (case-control, cross-section, and cohort studies) were included. Data extraction was performed independently by two extractors, and a crosscheck was performed to identify any errors. ORs along with their corresponding 95% confidence intervals (CIs) were estimated using random-effects models. The Preferred Reporting Items for Systematic Reviews and Meta-analyses reporting guidelines for RCTs and Meta-Analysis of Observational Studies in Epidemiology guidelines for observational studies were followed. We assessed the hearing loss risk after macrolide use versus controls (placebos and other antibiotics). Based on data from 13 studies including 1,142,021 patients (n = 267,546 for macrolide and n = 875,089 for controls), the overall pooled OR was 1.25 (95% CI 1.07-1.47). In subgroup analysis by study design, the ORs were 1.37 (95% CI 1.08-1.73) for RCTs and 1.33 (95% CI 1.24-1.43) for case-control studies, indicating that RCT and case-control study designs showed a statistically significant higher risk of hearing loss. The group with underlying diseases such as multiple infectious etiologies (OR, 1.16 [95% CI 0.96-1.41]) had a statistically significant lower risk than the group without (OR, 1.53 [95% CI 1.38-1.70] P = .013). The findings from this systematic review and meta-analysis suggest that macrolide antibiotics increase the risk of hearing loss and that healthcare professionals should carefully consider this factor while prescribing macrolides.


Subject(s)
Deafness , Hearing Loss , Ototoxicity , Tinnitus , Humans , Macrolides/adverse effects , Tinnitus/drug therapy , Ototoxicity/drug therapy , Anti-Bacterial Agents/adverse effects , Hearing Loss/chemically induced , Hearing Loss/epidemiology , Hearing Loss/drug therapy
9.
Curr Drug Targets ; 25(3): 158-170, 2024.
Article in English | MEDLINE | ID: mdl-38192136

ABSTRACT

Bile acids play important roles in the human body, and changes in their pool can be used as markers for various liver pathologies. In addition to their functional effects in modulating inflammatory responses and cellular survivability, the unconjugated or conjugated, secondary, or primary nature of bile acids accounts for their various ligand effects. The common hydrophilic bile acids have been used successfully as local treatment to resolve drug-induced cell damage or to ameliorate hearing loss. From various literature references, bile acids show concentration and tissue-dependent effects. Some hydrophobic bile acids act as ligands modulating vitamin D receptors, muscarinic receptors, and calcium-activated potassium channels, important proteins in the inner ear system. Currently, there are limited resources investigating the therapeutic effects of bile acid on hearing loss and little to no information on detecting bile acids in the remote ear system, let alone baseline bile acid levels and their prevalence in healthy and disease conditions. This review presents both hydrophilic and hydrophobic human bile acids and their tissue-specific effects in modulating cellular integrity, thus considering the possible effects and extended therapeutic applicability of bile acids to the inner ear tissue.


Subject(s)
Bile Acids and Salts , Hearing Loss , Animals , Humans , Bile Acids and Salts/metabolism , Bile Acids and Salts/therapeutic use , Ear, Inner/drug effects , Ear, Inner/metabolism , Hearing/drug effects , Hearing Loss/drug therapy , Hydrophobic and Hydrophilic Interactions , Ligands , Receptors, Calcitriol/metabolism , Receptors, Muscarinic/metabolism
10.
Arthritis Rheumatol ; 76(3): 444-454, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37738164

ABSTRACT

OBJECTIVE: Cryopyrin-associated periodic syndromes (CAPS), also known as NLRP3-associated autoinflammatory diseases, are a spectrum of rare autoinflammatory diseases caused by gain-of-function variants in the NLRP3 gene, resulting in inflammasome hyperactivation and dysregulated release of interleukin-1ß (IL-1ß). Many patients with CAPS develop progressive sensorineural hearing loss (SNHL) because of cochlear autoinflammation, which may be the sole manifestation in rare cases. This study was undertaken to establish the suspected diagnosis of CAPS in a family presenting with autosomal-dominant progressive/acute SNHL and a novel missense variant in the NLRP3 gene of unknown significance (NM_001079821.3:c.1784G>A p.Ser595Asn). METHODS: We conducted an ex vivo functional assessment of the NLRP3 inflammasome in heterozygous individuals (n = 10) and healthy family members (n = 5). RESULTS: The assay revealed hyperactivation of the inflammasome among heterozygous individuals, supporting the hypothesis that this missense variant is a pathogenic gain-of-function variant. Administration of IL-1 receptor antagonist resulted in a substantial clinical improvement among pediatric patients, who exhibited near resolution of hearing impairment within 1 to 3 months of treatment. CONCLUSION: Our findings highlight the crucial role of early diagnosis and treatment with an anti-IL-1 agent in reversing cochlear damage. Furthermore, our results suggest that high- and ultrahigh-frequency ranges need to be included in the auditory assessment to enable early detection of subclinical SNHL. Finally, incorporating functional inflammasome assessment as part of the clinical evaluation could establish the diagnosis in inconclusive cases.


Subject(s)
Cryopyrin-Associated Periodic Syndromes , Hearing Loss , Child , Humans , Cryopyrin-Associated Periodic Syndromes/drug therapy , Cryopyrin-Associated Periodic Syndromes/genetics , Family , Hearing Loss/drug therapy , Hearing Loss/genetics , Hearing Loss/complications , Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
11.
Clin Otolaryngol ; 49(1): 1-15, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37818931

ABSTRACT

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


Subject(s)
Antineoplastic Agents , Hearing Loss , Neoplasms , Ototoxicity , Adult , Humans , Cisplatin/therapeutic use , Antineoplastic Agents/therapeutic use , Carboplatin/adverse effects , Ototoxicity/etiology , Ototoxicity/prevention & control , Ototoxicity/drug therapy , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Hearing Loss/drug therapy , Neoplasms/drug therapy , Neoplasms/chemically induced , Adrenal Cortex Hormones/therapeutic use , Randomized Controlled Trials as Topic
12.
J Am Soc Nephrol ; 35(1): 22-40, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37962623

ABSTRACT

SIGNIFICANCE STATEMENT: To combat both untoward effects of nephrotoxicity and ototoxicity in cisplatin-treated patients, two potential therapeutic oral anticancer drugs AZD5438 and dabrafenib, a phase-2 clinical trial protein kinase CDK2 inhibitor and an US Food and Drug Administration-approved drug BRAF inhibitor, respectively, were tested in an established mouse AKI model. Both drugs have previously been shown to protect significantly against cisplatin-induced hearing loss in mice. Each drug ameliorated cisplatin-induced increases in the serum biomarkers BUN, creatinine, and neutrophil gelatinase-associated lipocalin. Drugs also improved renal histopathology and inflammation, mitigated cell death by pyroptosis and necroptosis, and significantly enhanced overall survival of cisplatin-treated mice. BACKGROUND: Cisplatin is an effective chemotherapy agent for a wide variety of solid tumors, but its use is dose-limited by serious side effects, including AKI and hearing loss. There are no US Food and Drug Administration-approved drugs to treat both side effects. Recently, two anticancer oral drugs, AZD5438 and dabrafenib, were identified as protective against cisplatin-induced hearing loss in mice. We hypothesize that similar cell stress and death pathways are activated in kidney and inner ear cells when exposed to cisplatin and tested whether these drugs alleviate cisplatin-induced AKI. METHODS: The HK-2 cell line and adult FVB mice were used to measure the protection from cisplatin-induced cell death and AKI by these drugs. Serum markers of kidney injury, BUN, creatinine, and neutrophil gelatinase-associated lipocalin as well as histology of kidneys were analyzed. The levels of markers of kidney cell death, including necroptosis and pyroptosis, pERK, and proliferating cell nuclear antigen, were also examined by Western blotting and immunofluorescence. In addition, CDK2 knockout (KO) mice were used to confirm AZD5438 protective effect is through CDK2 inhibition. RESULTS: The drugs reduced cisplatin-induced cell death in the HK-2 cell line and attenuated cisplatin-induced AKI in mice. The drugs reduced serum kidney injury markers, inhibited cell death, and reduced the levels of pERK and proliferating cell nuclear antigen, all of which correlated with prolonged animal survival. CDK2 KO mice were resistant to cisplatin-induced AKI, and AZD5438 conferred no additional protection in the KO mice. CONCLUSIONS: Cisplatin-induced damage to the inner ear and kidneys shares similar cellular beneficial responses to AZD5438 and dabrafenib, highlighting the potential therapeutic use of these agents to treat both cisplatin-mediated kidney damage and hearing loss.


Subject(s)
Acute Kidney Injury , Antineoplastic Agents , Hearing Loss , Humans , Mice , Animals , Cisplatin/toxicity , Lipocalin-2 , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Proliferating Cell Nuclear Antigen/therapeutic use , Creatinine , Drug Repositioning , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Antineoplastic Agents/toxicity , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Mice, Inbred Strains , Mice, Knockout , Apoptosis
13.
Laryngoscope ; 134(3): 1457-1463, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37589298

ABSTRACT

OBJECTIVE: Determine whether combination therapy with ganciclovir (GCV) and a Quercetin-P188 solution improves hearing outcomes in a murine cytomegalovirus (CMV) model. METHODS: BALB/c mice were infected with murine CMV on postnatal day 3 (p3). Quercetin was solubilized in saline using P188 (QP188). Treatment groups received either GCV, QP188, GCV and QP188, or P188 delivery vehicle BID at 12-hour intervals via intraperitoneal injection. All treatment groups were treated for 14 days starting at p3. Uninfected controls were treated with the combined regimen, saline or P188 delivery vehicle. Auditory thresholds were assessed using distortion product otoacoustic emission (DPOAE) and auditory brainstem response (ABR) testing at 4, 6, and 8 weeks of age. Temporal bones from separate CMV-infected groups were harvested at p10, and viral load was determined by quantitative polymerase chain reaction. RESULTS: CMV-infected mice receiving combination therapy GCV+QP188 demonstrated statistically significant lower ABR (p < 0.001) and DPOAE thresholds (p < 0.001) compared with mice treated with GCV monotherapy, QP188 monotherapy, and P188 delivery vehicle at 4, 6, and 8 weeks of age. GCV+QP188 combination therapy, GCV monotherapy, and QP188 monotherapy resulted in a nonsignificant reduction in mean viral titers compared to P188 monotherapy (p = 0.08). CONCLUSION: Combining GCV with the excipients quercetin and P188 effectively ameliorated CMV-induced sensorineural hearing loss in a murine model. This result may be partially explained by a reduction in viral titers in mouse temporal bones that correlate with in vitro studies demonstrating additive antiviral effect in cell culture. LEVEL OF EVIDENCE: NA Laryngoscope, 134:1457-1463, 2024.


Subject(s)
Cytomegalovirus Infections , Deafness , Hearing Loss , Animals , Mice , Ganciclovir/pharmacology , Ganciclovir/therapeutic use , Cytomegalovirus , Quercetin/pharmacology , Quercetin/therapeutic use , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/drug therapy , Hearing Loss/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
14.
Yakugaku Zasshi ; 143(11): 977-981, 2023.
Article in Japanese | MEDLINE | ID: mdl-37914344

ABSTRACT

Pharmacists are required to provide and collect medication information based on patients' health literacy and communication abilities to provide effective pharmaceutical care. Due to a lack of understanding of hearing loss and awareness of the inconvenience that patients with hearing loss face, appropriate actions for effective communication in medication education are not fully implemented. An e-learning system consisting of two courses, a learning course and an evaluation course, has been developed. The learning course explains hearing loss and appropriate actions in medication education and investigates pharmacists' recognition of medication education, while the evaluation course assesses the implementation of necessary actions in medication education and changes in pharmacists' recognition of medication education with patients compared to before the e-learning. From February to September 2022, 41 pharmacists completed the learning course, with 22 advancing to the evaluation course. Prior to learning, they had difficulty in communicating with patients with hearing loss. However, after the learning course, their confidence in medication education improved with a better understanding of hearing loss and practice of appropriate actions. They also felt that pharmacists who were unfamiliar with the system should understand hearing loss and take actions tailored to patients' hearing loss. Further examination may be needed of the effects of the e-learning on patients with hearing loss.


Subject(s)
Computer-Assisted Instruction , Deafness , Hearing Loss , Humans , Pharmacists , Learning , Hearing Loss/drug therapy
15.
Sci Rep ; 13(1): 18957, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919361

ABSTRACT

Studies have reported that cytokines and their related signaling pathways play a role in inner ear diseases. In clinical practice, approximately 50% of pediatric cancer patients experience irreversible hearing loss after cisplatin treatment. However, currently, there is a lack of systematic research on the causal relationship between circulating cytokines and cisplatin-induced hearing loss in children. Genetic variant data for 41 circulating cytokines were obtained from a meta-analysis of genome-wide association studies (GWAS) among 8293 individuals of Finnish descent. The GWAS data for Cisplatin-induced hearing loss in children were derived from a multicenter cohort of European pediatric cancer patients and survivors (N = 390), including both cases with hearing loss after cisplatin chemotherapy and controls without hearing loss. Multiple methods were employed for bidirectional Mendelian randomization (MR) estimation. Bonferroni correction was applied to adjust the original P-values, followed by a series of sensitivity analyses. In the directional Mendelian randomization (MR) analysis, it was found that IL-17 was significantly associated with a reduced risk of Cisplatin-induced hearing loss in children (OR: 0.18, CI: 0.06-0.48, P < 0.001, FDR = 0.041). In the reverse MR analysis, there were some nominal causal relationships of Cisplatin-induced hearing loss in children with certain cytokines [M-CSF: (OR: 1.04, CI: 1.01-1.08, P = 0.010, FDR = 0.41); IL-2RA: (OR: 1.03, CI: 1.00-1.05, P = 0.044, FDR = 0.447); MIP-1ß: (OR: 1.02, CI: 1.00-1.04, P = 0.041, FDR = 0.447)]. leave-one-out analysis demonstrated that only M-CSF exhibited stability. These findings reveal a causal relationship between IL-17 and cisplatin-induced hearing loss in children. Further research is needed to determine the potential protective mechanisms of IL-17 in cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents , Deafness , Hearing Loss , Neoplasms , Humans , Child , Cisplatin/therapeutic use , Antineoplastic Agents/therapeutic use , Macrophage Colony-Stimulating Factor , Genome-Wide Association Study , Mendelian Randomization Analysis , Interleukin-17/genetics , Hearing Loss/chemically induced , Hearing Loss/genetics , Hearing Loss/drug therapy , Neoplasms/drug therapy , Cytokines/therapeutic use , Multicenter Studies as Topic
16.
JCI Insight ; 8(24)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-37934596

ABSTRACT

The widely used chemotherapy cisplatin causes permanent hearing loss in 40%-60% of patients with cancer. One drug, sodium thiosulfate, is approved by the FDA for use in pediatric patients with localized solid tumors for preventing cisplatin-induced hearing loss, but more drugs are desperately needed. Here, we tested dabrafenib, an FDA-approved BRAF kinase inhibitor and anticancer drug, in a clinically relevant multidose cisplatin mouse model. The protective effects of dabrafenib, given orally twice daily with cisplatin, were determined by functional hearing tests and cochlear outer hair cell counts. Toxicity of the drug cotreatment was evaluated, and levels of phosphorylated ERK were measured. A dabrafenib dose of 3 mg/kg BW, twice daily, in mice, was determined to be the minimum effective dose, and it is equivalent to one-tenth of the daily FDA-approved dose for human cancer treatment. The levels of hearing protection acquired, 20-25 dB at the 3 frequencies tested, in both female and male mice, persisted for 4 months after completion of treatments. Moreover, dabrafenib exhibited a good in vivo therapeutic index (> 25), protected hearing in 2 mouse strains, and diminished cisplatin-induced weight loss. This study demonstrates that dabrafenib is a promising candidate drug for protection from cisplatin-induced hearing loss.


Subject(s)
Antineoplastic Agents , Deafness , Hearing Loss , Neoplasms , Humans , Male , Female , Child , Mice , Animals , Cisplatin/toxicity , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Hearing Loss/drug therapy , Antineoplastic Agents/toxicity , Imidazoles/pharmacology , Imidazoles/therapeutic use , Neoplasms/drug therapy
17.
Int J Mol Sci ; 24(22)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38003734

ABSTRACT

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


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Deafness , Hearing Loss , Osteosarcoma , Ototoxicity , Humans , Child , Cisplatin/adverse effects , Antineoplastic Agents/adverse effects , Ototoxicity/etiology , Ototoxicity/drug therapy , Hearing Loss/drug therapy , Osteosarcoma/drug therapy , Bone Neoplasms/drug therapy , Inflammation/drug therapy
18.
Expert Opin Investig Drugs ; 32(8): 685-692, 2023.
Article in English | MEDLINE | ID: mdl-37695693

ABSTRACT

INTRODUCTION: : Hearing loss has a high prevalence, with aging, noise exposure, ototoxic drug therapies, and genetic mutations being some of the leading causes of hearing loss. Health conditions such as cardiovascular disease and diabetes are associated with hearing loss, perhaps due to shared vascular pathology in the ear and in other tissues. AREAS COVERED: : Issues in the design of preclinical research preclude the ability to make comparisons regarding the relative efficacy of different drugs of interest for possible hearing loss prevention or hearing restoration. This has not slowed the advancement of candidate therapeutics into human clinical testing. There is a robust pipeline with drugs that have different mechanisms of action providing diverse candidate therapies and opportunities for combination therapies to be considered. EXPERT OPINION: : Much of the preclinical research literature lacks standard study design elements such as dose response testing, and lack of standardization of test protocols significantly limits conclusions regarding relative efficacy. Nonetheless, the many positive results to date have supported translation of preclinical efforts into clinical trials assessing potential human benefits. Approval of the first hearing loss prevention therapeutic is a major success, providing a pathway for other drugs to follow.


Subject(s)
Hearing Loss, Sensorineural , Hearing Loss , Humans , Hearing Loss/drug therapy , Hearing Loss/prevention & control , Hearing Loss/complications , Hearing Loss, Sensorineural/drug therapy , Hearing Loss, Sensorineural/etiology
19.
Cell Death Dis ; 14(9): 605, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37704645

ABSTRACT

Hearing loss impacts the quality of life and affects communication resulting in social isolation and reduced well-being. Despite its impact on society and economy, no therapies for age-related hearing loss are available so far. Loss of mechanosensory hair cells of the cochlea is a common event of hearing loss in humans. Studies performed in birds demonstrating that they can be replaced following the proliferation and transdifferentiation of supporting cells, strongly pointed out on HCs regeneration as the main focus of research aimed at hearing regeneration. Neurotrophins are growth factors involved in neuronal survival, development, differentiation, and plasticity. NGF has been involved in the interplay between auditory receptors and efferent innervation in the cochlea during development. During embryo development, both NGF and its receptors are highly expressed in the inner ears. It has been reported that NGF is implicated in the differentiation of auditory gangliar and hair cells. Thus, it has been proposed that NGF administration can decrease neuronal damage and prevent hearing loss. The main obstacle to the development of hearing impairment therapy is that efficient means of delivery for selected drugs to the cochlea are missing. Herein, in this study NGF was administered by the intranasal route. The first part of the study was focused on a biodistribution study, which showed the effective delivery in the cochlea; while the second part was focused on analyzing the potential therapeutic effect of NGF in senescence-accelerated prone strain 8 mice. Interestingly, intranasal administration of NGF resulted protective in counteracting hearing impairment in SAMP8 mice, ameliorating hearing performances (analyzed by auditory brainstem responses and distortion product otoacoustic emission) and hair cells morphology (analyzed by microscopy analysis). The results obtained were encouraging indicating that the neurotrophin NGF was efficiently delivered to the inner ear and that it was effective in counteracting hearing loss.


Subject(s)
Deafness , Hearing Loss , Humans , Animals , Mice , Aged , Administration, Intranasal , Nerve Growth Factor/pharmacology , Quality of Life , Tissue Distribution , Hearing Loss/drug therapy
20.
Yakugaku Zasshi ; 143(8): 673-682, 2023.
Article in Japanese | MEDLINE | ID: mdl-37532576

ABSTRACT

To improve medication education for patients with hearing loss, pharmacists must better understand hearing loss and provide and collect medication information based on patients' health literacy and communication abilities. However, no systematic educational e-learning systems for hearing loss are currently available. Therefore, an e-learning system based on instructional design, microlearning principles, and multimedia teaching materials was developed. The e-learning system used Moodle, an open-source e-learning system, and included two courses: one for self-directed learning by watching videos and answering quizzes, and another for evaluating medication education after learning. A study was conducted on 84 pharmacists and 36 pharmacy students who took the learning course to investigate the factors that hinder their understanding and progress of self-directed learning. Although they fully understood the content by watching videos, students with no experience in medication education required an explanation to understand how to communicate with patients. As the learning course was self-directed and related to communication lectures, all students completed it; however, the completion rate for pharmacists was approximately 50%. The following factors could have slowed pharmacists' e-learning progress: difficulty accessing the learning course through system login and long-duration content, such as answers to free descriptions. This survey found that this e-learning method can be used for self-directed learning about medication education for patients with hearing loss. Further improvement of the e-learning system is necessary so that recognizing the need to understand hearing loss and take appropriate actions for patients with hearing loss in medication education can lead to self-directed learning among pharmacists.


Subject(s)
Deafness , Hearing Loss , Students, Pharmacy , Humans , Learning , Hearing Loss/drug therapy , Health Education
SELECTION OF CITATIONS
SEARCH DETAIL