Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
1.
Toxicol Lett ; 354: 56-64, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34757176

ABSTRACT

The ototoxic mechanisms of cisplatin on the organ of Corti and spiral ganglion neurons have been extensively studied, while few studies have been focused on the stria vascularis (SV). Herein, we verified the functional and morphological impairment in SV induced by a single injection of cisplatin (12 mg/kg, I.P.), represented by a reduction in Endocochlear Potentials (EP) and strial atrophy, and explored underlying mechanisms. Our results revealed increased extravasation of chromatic tracers (Evans blue dye and FITC-dextran) around microvessels after cisplatin exposure. The increased vascular permeability could be attributed to changes of pericytes (PCs) and perivascular-resident macrophage-like melanocytes (PVM/Ms) in number or morphology, as well as the enhanced level of HIF-1α and downstream VEGF. This capillary leakage led to a high accumulation of cisplatin in the perivascular space in SV, and disrupted the integrity of blood-labyrinth barrier (BLB). Also, tight junction (ZO-1) loosening and Na+, K+-ATPase damage was considered to be other critical contributors of BLB breakdown, which resulted in EP drop and consequent hearing loss. This study explored the role of stria vascularis in cisplatin-induced ototoxicity in terms of BLB hyperpermeability and pointed to a novel therapeutic target for the prevention of cisplatin-related hearing loss.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cochlea/blood supply , Cochlea/drug effects , Ototoxicity/etiology , Permeability/drug effects , Stria Vascularis/drug effects , Animals , Disease Models, Animal , Male , Mice
2.
Eur J Pharmacol ; 913: 174642, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34822791

ABSTRACT

It is widely accepted that the stria vascularis (SV) in cochlea plays a critical role in the generation of endocochlear potential (EP) and the secretion of the endolymph. 17ß-estradiol (E2) is the most potent and abundant endogenous estrogen during the premenopausal period, thus, considered as the reference estrogen. This study aimd to investigate the protective effect of E2 by promoting the expression of vascular endothelial growth factor (VEGF) and thus promoting the vascular regeneration of the SV in elderly mice. After being treated with E2 either in vivo or in vitro, the hearing threshold changes of C57BL/6J elder mice continuously reduced, endothelial cell morphology improved, the number of endothelial cells (ECs) tubular nodes increased significantly, the ability of tubular formation enhanced significantly and the expression of VEGF increased. In vitro, cell model in conjunction with in vivo ovariectomized model was established to demonstrate for the first time that E2 promotes angiogenesis by promoting the secretion of VEGF through the phosphatidylinositol 3-kinase (PI3K)/AKT pathway (PI3K/AKT). In conclusion, E2 demonstrated potent angiogenesis properties with significant protection against Age-Related Hearing Loss (ARHL), which provides a new idea for the improvement of ARHL.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Estradiol/pharmacology , Hearing Loss/prevention & control , Neovascularization, Physiologic/drug effects , Stria Vascularis/drug effects , Aging/physiology , Angiogenesis Inducing Agents/therapeutic use , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Estradiol/therapeutic use , Female , Hearing Loss/physiopathology , Humans , Mice , Organ Culture Techniques , Regeneration/drug effects , Signal Transduction/drug effects , Stria Vascularis/physiology , Vascular Endothelial Growth Factor A/agonists , Vascular Endothelial Growth Factor A/metabolism
3.
Hear Res ; 390: 107935, 2020 05.
Article in English | MEDLINE | ID: mdl-32234583

ABSTRACT

In 129 Sv autosomal Alport mice, the strial capillary basement membranes (SCBMs) progressively thicken between 5 and 9 weeks of age resulting in a hypoxic microenvironment with metabolic stress and induction of pro-inflammatory cytokines and chemokines. These events occur concomitant with a drop in endocochlear potential and a susceptibility to noise-induced hearing loss under conditions that do not permanently affect age/strain-matched littermates. Here we aimed to gain an understanding of events that occur before the onset of SCBM thickening. Alport stria has normal thickness and shows levels of extracellular matrix (ECM) molecules in the SCBMs commensurate with wild-type mice. Hearing thresholds in the 3-week Alport mice do not differ from those of wild-type mice. We performed RNAseq analysis using RNA from stria vascularis isolated from 3-week Alport mice and wild type littermates. Data was processed using Ingenuity Pathway Analysis software and further distilled using manual procedures. RNAseq analysis revealed significant dysregulation of genes involved in cell adhesion, cell migration, formation of protrusions, and both actin and tubulin cytoskeletal dynamics. Overall, the data suggested changes in the cellular architecture of the stria might be apparent. To test this notion, we performed dual immunofluorescence analysis on whole mounts of the stria vascularis from these same animals stained with anti-isolectin gs-ib4 (endothelial cell marker) and anti-desmin (pericyte marker) antibodies. The results showed evidence of pericyte detachment and migration as well as the formation of membrane ruffling on pericytes in z-stacked confocal images from Alport mice compared to wild type littermates. This was confirmed by TEM analysis. Earlier work from our lab showed that endothelin A receptor blockade prevents SCBM thickening and ECM accumulation in the SCBMs. Treating cultured pericytes with endothelin-1 induced actin cytoskeletal rearrangement, increasing the ratio of filamentous to globular actin. Collectively, these findings suggest that the change in type IV collagen composition in the Alport SCBMs results in cellular insult to the pericyte compartment, activating detachment and altered cytoskeletal dynamics. These events precede SCBM thickening and hearing loss in Alport mice, and thus constitute the earliest event so far recognized in Alport strial pathology.


Subject(s)
Actin Cytoskeleton/ultrastructure , Basement Membrane/ultrastructure , Nephritis, Hereditary/pathology , Pericytes/ultrastructure , Stria Vascularis/ultrastructure , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Basement Membrane/drug effects , Basement Membrane/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Endothelin-1/pharmacology , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Male , Mice, 129 Strain , Microscopy, Confocal , Microscopy, Electron, Transmission , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Pericytes/drug effects , Pericytes/metabolism , RNA-Seq , Receptor, Endothelin A/agonists , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Signal Transduction , Stria Vascularis/drug effects , Stria Vascularis/metabolism
4.
Int. arch. otorhinolaryngol. (Impr.) ; 23(3): 267-275, July-Sept. 2019. tab, graf
Article in English | LILACS | ID: biblio-1040018

ABSTRACT

Abstract Introduction Riluzole (2-amino-6-trifluoromethoxy benzothiazole) is known as a neuroprotective, antioxidant, antiapoptotic agent. It may have beneficial effects on neuronal cell death due to cisplatin-induced ototoxicity. Objective To evaluate the effect of riluzole on cisplatin-induced ototoxicity in guinea pigs. Methods Twenty-four guinea pigs, studied in three groups, underwent auditory brainstem response evaluation using click and 8 kHz tone burst stimuli. Subsequently, 5 mg/kg of cisplatin were administered to all animals for 3 days intraperitoneally (i.p.) to induce ototoxicity. Half an hour prior to cisplatin, groups 1, 2 and 3 received 2 ml of saline i.p., 6 mg/kg of riluzole hydrochloride i.p., and 8 mg/kg of riluzole hydrochloride i.p., respectively, for 3 days. The auditory brainstem responses were repeated 24 hours after the last drug administration. The cochleae were analyzed by transmission electron microscopy (TEM). Results After drug administiration, for 8,000 Hz stimulus, group 1 had significantly higher threshold shifts when compared with groups 2 (p < 0.05) and 3 (p < 0.05), and there was no significant difference in threshold shifts between groups 2 and 3 (p > 0.05). Transmission electron microscopy findings demonstrated the protective effect of riluzole on the hair cells and the stria vascularis, especially in the group treated with 8 mg/kg of riluzole hydrochloride. Conclusion We can say that riluzolemay have a protective effect on cisplatin- induced ototoxicity. However, additional studies are needed to confirm these results and the mechanisms of action of riluzole.


Subject(s)
Animals , Male , Evoked Potentials, Auditory, Brain Stem/drug effects , Cisplatin/adverse effects , Riluzole/pharmacology , Hearing Loss, Sensorineural/chemically induced , Auditory Threshold/drug effects , Stria Vascularis/drug effects , Stria Vascularis/pathology , Cochlear Nerve/drug effects , Cochlear Nerve/pathology , Riluzole/therapeutic use , Models, Animal , Microscopy, Electron, Transmission , Guinea Pigs , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Nerve Degeneration/chemically induced
5.
ORL J Otorhinolaryngol Relat Spec ; 81(4): 202-214, 2019.
Article in English | MEDLINE | ID: mdl-31340214

ABSTRACT

Cisplatin is an anti-cancer drug that causes oxotoxic side effects such as impairment of inner ear function and hearing loss. We aimed to investigate the effects of allicin against cisplatin-induced stria vascularis damage in mice, and to clarify the mechanism underlying the protective effects of allicin against ototoxicity. Stria vascularis injury was induced in mice by intraperitoneal injection of cisplatin, which was significantly prevented by pretreatment with allicin. Allicin not only reduced cisplatin-activated expression of cleaved caspase-3 in marginal cells, PVM/Ms (perivascular resident macrophage-like melanocytes), and basal cells of the stria vascularis, but also decreased the expression of poly(ADP-ribose) polymerase-1 (PARP-1) and apoptosis-inducing factor (AIF) nuclear translocation in the stria vascularis cells. Our results demonstrate that allicin plays an effective role in protecting stria vascularis injury induced by cisplatin by inhibiting caspase-dependent, as well as caspase-independent PARP-1-AIF-mediated apoptotic pathways. Therefore, allicin may be useful in preventing cisplatin-induced ototoxicity.


Subject(s)
Apoptosis , Caspase 3/drug effects , Hearing Loss/prevention & control , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Stria Vascularis/drug effects , Sulfinic Acids/pharmacology , Animals , Antioxidants/pharmacology , Caspase 3/metabolism , Cisplatin/toxicity , Disease Models, Animal , Disulfides , Female , Hearing Loss/chemically induced , Hearing Loss/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Poly (ADP-Ribose) Polymerase-1/metabolism , Stria Vascularis/metabolism , Stria Vascularis/ultrastructure
6.
Mol Med Rep ; 20(2): 1593-1604, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31257512

ABSTRACT

The present study was designed to investigate the expression and function of transmembrane protein 16 (TMEM16A), a calcium­activated chloride channel (CaCC), in the stria vascularis (SV) of the cochlea of guinea pigs at different ages, and to understand the role of CaCCs in the pathogenesis of presbycusis (age­related hearing loss), the most common type of sensorineural hearing loss that occurs with natural aging. Guinea pigs were divided into the following groups: 2 weeks (young group), 3 months (youth group), 1 year (adult group), D­galactose intervention (D­gal group; aging model induced by subcutaneous injection of D­galactose) and T16Ainh­A01 (intraperitoneal injection of 50 µg/kg/day TMEM16A inhibitor T16Ainh­A01 for 2 weeks). Differences in the hearing of guinea pigs between the various age groups were analyzed using auditory brainstem response (ABR), and immunofluorescence staining was performed to detect TMEM16A expression in the SV and determine the distribution. Reverse transcription­quantitative PCR and western blot analyses were conducted to detect the mRNA and protein levels of TMEM16A in SV in the different age groups. Morris water maze behavior analysis demonstrated that spatial learning ability and memory were damaged in the D­gal group. Superoxide dismutase activity and malondialdehyde content assays indicated that there was oxidative stress damage in the D­gal group. The ABR thresholds gradually increased with age, and the increase in the T16Ainh­A01 group was pronounced. Immunofluorescence analysis in the cochlear SV of guinea pigs in different groups revealed that expression of TMEM16A increased with increasing age (2 weeks to 1 year); fluorescence intensity was reduced in the D­gal model of aging. As the guinea pigs continued to mature, the protein and mRNA contents of TMEM16A in the cochlea SV increased gradually, but were decreased in the D­gal group. The findings indicated that CaCCs in the cochlear SV of guinea pigs were associated with the development of hearing in guinea pigs, and that downregulation of TMEM16A may be associated with age­associated hearing loss.


Subject(s)
Aging/genetics , Anoctamin-1/genetics , Presbycusis/genetics , Stria Vascularis/metabolism , Aging/drug effects , Aging/metabolism , Animals , Anoctamin-1/antagonists & inhibitors , Anoctamin-1/metabolism , Disease Models, Animal , Female , Galactose/administration & dosage , Gene Expression Regulation , Guinea Pigs , Hearing/physiology , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Presbycusis/chemically induced , Presbycusis/metabolism , Presbycusis/physiopathology , Pyrimidines/pharmacology , Stria Vascularis/drug effects , Stria Vascularis/pathology , Thiazoles/pharmacology
7.
J Int Adv Otol ; 15(2): 229-236, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31287434

ABSTRACT

OBJECTIVES: This study aimed to evaluate the effect of quercetin on cochlear function and morphology, and its possible protective effect against acute cisplatin-induced ototoxicity in rats. MATERIALS AND METHODS: This prospective and controlled animal study was conducted on Wistar albino rats divided into four groups. Otoacoustic emission measures were performed three days after the first infiltration in Group 1 (saline), 2 (cisplatin), and 3 (quercetin). This interval was five days for Group 4 (cisplatin+quercetin). At the end of the study, the rats were decapitated with deep anesthesia, and histological changes in the cochleas were observed by light microscopy. RESULTS: Group 2 (cisplatin) revealed significant differences between the first and second measures in all frequencies. When compared to other group, the difference of the changes in Group 2 statistically significantly decreased, especially in higher frequencies. Morphologically, there were no acute changes in Group 1 and Group 3. Outer hair cell loss and the degeneration of stria vascularis and spiral ganglion were observed in both Groups 2 and 4; the damages in the latter were lesser. CONCLUSION: Quercetin does not have negative effect on cochlea, and it has protective effect on cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Antioxidants/pharmacology , Cisplatin/toxicity , Ototoxicity/prevention & control , Quercetin/pharmacology , Analysis of Variance , Animals , Female , Organ of Corti/drug effects , Organ of Corti/pathology , Ototoxicity/pathology , Rats, Wistar , Stria Vascularis/drug effects , Stria Vascularis/pathology
8.
Hear Res ; 377: 179-188, 2019 06.
Article in English | MEDLINE | ID: mdl-30954884

ABSTRACT

Can damaged or degenerated vessels be regenerated in the ear? The question is clinically important, as disruption of cochlear blood flow is seen in a wide variety of hearing disorders, including in loud sound-induced hearing loss (endothelial injury), ageing-related hearing loss (lost vascular density), and genetic hearing loss (e.g., Norrie disease: strial avascularization). Progression in cochlear blood flow (CBF) pathology can parallel progression in hair cell and hearing loss. However, neither new vessel growth in the ear, nor the role of angiogenesis in hearing, have been investigated. In this study, we used an established ex vivo tissue explant model in conjunction with a matrigel matrix model to demonstrate for the first time that new vessels can be generated by activating a vascular endothelial growth factor (VEGF-A) signal. Most intriguingly, we found that the pattern of the newly formed vessels resembles the natural 'mesh pattern' of in situ strial vessels, with both lumen and expression of tight junctions. Sphigosine-1-phosphate (S1P) in synergy with VEGF-A control new vessel size and growth. Using transgenic neural/glial antigen 2 (NG2) fluorescent reporter mice, we have furthermore discovered that the progenitors of "de novo" strial vessels are NG2-derived cells. Taken together, our data demonstrates that damaged strial microvessels can be regenerated by reprogramming NG2-derived angiogenic cells. Restoration of the functional vasculature may be critical for recovery of vascular dysfunction related hearing loss.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Antigens/metabolism , Cochlea/blood supply , Endothelial Progenitor Cells/drug effects , Neovascularization, Physiologic/drug effects , Proteoglycans/metabolism , Stria Vascularis/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Antigens/genetics , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Lysophospholipids/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Proteoglycans/genetics , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stria Vascularis/metabolism , Stria Vascularis/ultrastructure , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
9.
Biomed Res Int ; 2019: 3626032, 2019.
Article in English | MEDLINE | ID: mdl-30766882

ABSTRACT

Lead exposure causes or aggravates hearing damage to human or animal, but the detailed effects of lead exposure on auditory system including injury sites of the cochlea in mammal remain controversy. To investigate the effect of chronic lead exposure on auditory system, 40 adult guinea pigs with normal hearing were randomly divided into five groups. They were fed 2 mmol/L lead acetate in drinking water for 0, 15, 30, 60, and 90 days (n = 8), respectively. Lead concentrations in blood, cochlea, and brainstem were measured. Auditory function was measured by auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE). The morphology of cochlea and brainstem was observed, and expression of autophagy-related protein in brainstem was also assessed. The blood lead concentration reached a high level at the 15th day and kept stable, but the lead level in brainstem and cochlear tissue increased obviously at the 60th day and 90th day of lead exposure, respectively. There was no significant difference in the morphology of hair cells and stria vascularis (SV) among these five groups, but the number of spiral ganglion neuron (SGN) gradually decreased after 60 days. The differences of ABR thresholds and DPOAE amplitudes were not statistically significant among each group, but I wave latency, III latency, and I-III wave interval of ABR were delayed with the prolonging of time of lead exposure. The expressions of autophagy-related protein ATG5, ATG6, and LC3B in brainstem were increased after 30 days. These results suggest that the key target of lead toxicity was the auditory nerve conduction pathway including SGNs and brainstem, rather than cochlear hair cells and SV. Autophagy may play a very important role in lead toxicity to auditory nervous system.


Subject(s)
Cochlea/drug effects , Hearing/drug effects , Lead/adverse effects , Neurotoxicity Syndromes/physiopathology , Animals , Auditory Threshold/drug effects , Brain Stem/drug effects , Brain Stem/physiopathology , Cochlea/physiopathology , Cochlear Nerve/drug effects , Cochlear Nerve/physiopathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Guinea Pigs , Hair Cells, Auditory/drug effects , Lead/blood , Male , Neurons/drug effects , Otoacoustic Emissions, Spontaneous/drug effects , Spiral Ganglion/drug effects , Spiral Ganglion/physiopathology , Stria Vascularis/drug effects
10.
Auris Nasus Larynx ; 46(5): 790-796, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30739815

ABSTRACT

Objective The endocytosis of cationized feritin (CF) via a clathrin-mediated pathway is regulated by a signaling network. Marginal cells showed the active endocytosis of CF via a clathrin-mediated pathway. The internalization of receptors through this clathrin-mediated pathway is an important regulatory event in signal transduction. Numerous kinases are involved in endocytosis, and each endocytic route is subjected to high-order regulation by cellular signaling mechanisms. In this study, we investigated whether ROCK and MLCK signaling cascades and G-proteins regulate the endocytosis of CF in marginal cells of the stria vascularis. Methods CF was infused into the cochlear duct with pertussis toxin (PTX),Clostridium botulinum C3 toxin (BTX), guanosine(g-thio)-triphosphate (GTP-γS), ML-7, Y-27632. Endocytic activity was measured at 30 min after the start of infusion under an electron microscope. Results In marginal cells, CF was internalized via a clathrin-mediated pathway that depends on F-actin and microtubules (MT). Its processes were controlled by myosin light chain kinase (MLCK) and Rho-associated kinase (ROCK), but not affected by G-protein-coupled receptor (GPCR) or the RhoA signaling cascade. Conclusion Our previous study showed that the main endocytotic pathway of microperoxidase (MPO) did not depend on the Rho/ROCK molecular switch or actin/myosin motor system, but was mainly regulated by the RhoA signaling cascade. The present study results indicate that these signaling cascades regulating CF internalization completely differ from the cascades for MPO internalization.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Endocytosis/physiology , Ferritins/metabolism , GTP-Binding Proteins/metabolism , Myosin-Light-Chain Kinase/metabolism , Stria Vascularis/metabolism , rho-Associated Kinases/metabolism , ADP Ribose Transferases/pharmacology , Amides/pharmacology , Animals , Azepines/pharmacology , Botulinum Toxins/pharmacology , Cochlear Duct , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Guinea Pigs , Microscopy, Electron , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Myosin-Light-Chain Phosphatase/metabolism , Naphthalenes/pharmacology , Pertussis Toxin/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stria Vascularis/drug effects , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
11.
Auris Nasus Larynx ; 46(5): 703-708, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30799140

ABSTRACT

OBJECTIVE: Tsumura Suzuki Obese Diabetes (TSOD) mice exhibit early age-associated hearing loss. Histopathological analysis of these mice shows narrowing of capillaries in the stria vascularis and chronic reduction of blood flow in the cochlea. In this study, we investigated the effect of oral administration of a herbal medicine or calorie restriction on hearing in TSOD mice. METHODS: TSOD mice were divided into 4 groups: CR (calorie restriction), BF and DS (treated with the herbal medicines, Bofutsushosan and Daisaikoto, respectively), and the control group. Body weight, blood glucose levels, and auditory brainstem responses (ABRs) were measured. The cochleae were excised and evaluated histopathologically. RESULTS: Blood glucose levels were suppressed in the CR, BF, and DS groups. In addition, the elevation of ABR thresholds was inhibited in the CR, BF, and DS groups. Cochlear blood vessels remained wide in the three treatment groups compared with the control group. These results suggested that the administration of these herbal medicines improved glucose tolerance and yielded results similar to those on calorie restriction. CONCLUSION: Oral administration of 2 herbal medicines can prevent hearing function disorder in a model mouse of diabetes. The results may clarify the possibility of clinical application.


Subject(s)
Caloric Restriction , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Drugs, Chinese Herbal/pharmacology , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss/metabolism , Plant Preparations/pharmacology , Administration, Oral , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Capillaries/drug effects , Capillaries/pathology , Cochlea/blood supply , Cochlea/drug effects , Cochlea/pathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Disease Progression , Hearing Loss/etiology , Mice , Stria Vascularis/drug effects , Stria Vascularis/pathology
12.
J Cell Physiol ; 234(3): 1978-1986, 2019 03.
Article in English | MEDLINE | ID: mdl-30317595

ABSTRACT

The study of strial pericytes has gained great interest as they are pivotal for the physiology of stria vascularis. To provide an easily accessible in vitro model, here we described a growth medium-based approach to obtain and cultivate primary bovine cochlear pericytes (BCP) from the stria vascularis of explanted bovine cochleae. We obtained high-quality pericytes in 8-10 days with a > 90% purity after the second passage. Immunocytochemical analysis showed a homogeneous population of cells expressing typical pericyte markers, such as neural/glial antigen 2 (NG2), platelet-derived growth factor receptorß (PDGFRß), α-smooth muscle actin (α-SMA), and negative for the endothelial marker von Willebrand factor. When challenged with tumor necrosis factor or lipopolysaccharide, BCP changed their shape, similarly to human retinal pericytes (HRPC). The sensitivity of BCP to ototoxic drugs was evaluated by challenging with cisplatin or gentamicin for 48 hr. Compared to human retinal endothelial cells and HRPC, cell viability of BCP was significantly lower ( p < 0.05) after the treatment with gentamicin or cisplatin. These data indicate that our protocol provides a simple and reliable method to obtain highly pure strial BCP. Furthermore, BCP are suitable to assess the safety profile of molecules which supposedly exert ototoxic activity, and may represent a valid alternative to in vivo tests.


Subject(s)
Cochlea/cytology , Pericytes/cytology , Stria Vascularis/cytology , Actins/metabolism , Animals , Antigens/metabolism , Biomarkers/metabolism , Cattle , Cell Culture Techniques/methods , Cell Survival , Cisplatin/toxicity , Cochlea/drug effects , Cochlea/metabolism , Culture Media , Drug Evaluation, Preclinical/methods , Gentamicins/toxicity , In Vitro Techniques , Models, Biological , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Pericytes/drug effects , Pericytes/metabolism , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stria Vascularis/drug effects , Stria Vascularis/metabolism
13.
Cell Death Dis ; 9(8): 827, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068942

ABSTRACT

Cisplatin, a small platinum-containing molecule, is a widely used, highly effective anticancer drug. However, severe side effects have been found in cancer patients treated with cisplatin, including nephrotoxicity, neurotoxicity, and ototoxicity. These cisplatin-induced side effects can have a major impact on patient quality of life, including social development problems in pediatric patients that develop hearing loss. Previous studies have suggested that the major cause of cisplatin-induced ototoxicity is abnormal accumulation of reactive oxygen species (ROS) and oxidative stress. Alpha-lipoic acid (ALA), one of the most effective antioxidants, is known to be involved in the cellular antioxidant system and may have a protective effect on cisplatin-induced ototoxicity. However, the therapeutic effect of ALA on damaged hearing function and its detailed mechanism of action are not fully understood. This study focused on determining whether ALA has a potential as a protective and/or therapeutic agent for cisplatin-induced ototoxicity. Histological and physiological analyses were performed using cisplatin-treated mouse cochlea and HEI-OC1 culture cells in pre- and post-treatment with ALA in vitro and in vivo. We found that ALA contributes to protecting mitochondrial function by preventing ROS accumulation and inhibiting apoptotic cell death. Importantly, post-treatment with ALA consistently showed an almost equal restorative effect to pretreatment, in vitro and in vivo, supporting the possible use of ALA as a therapeutic agent for cisplatin-induced ototoxicity. This study is the first report on a strong therapeutic potential of ALA to rescue ototoxic hearing loss caused by cisplatin, and our data provide key evidence that ALA may act as a reducing agent for glutathione disulfide to increase glutathione levels on behalf of glutathione reductase. This result was consistent in both cultured cells and the mouse model, which improves the clinical value of ALA for therapy of cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Hearing Loss/prevention & control , Protective Agents/therapeutic use , Thioctic Acid/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Ear, Inner/pathology , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hearing Loss/chemically induced , Male , Mice , Protective Agents/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Spiral Ganglion/cytology , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Stria Vascularis/drug effects , Stria Vascularis/physiology , Thioctic Acid/pharmacology , bcl-2-Associated X Protein/metabolism
14.
Mol Med Rep ; 17(2): 2861-2868, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29257229

ABSTRACT

The present study was designed to investigate the electrophysiological properties of strial pericytes and the effect of aspirin on pericyte K+ channels. Pericytes were identified by determining their morphological characteristics and using pericyte­associated immunofluorescence techniques. The electrophysiological properties of strial pericytes were observed with a whole­cell patch­clamp technique. Alterations in the outward current of cochlear pericytes in the stria vascularis of guinea pigs were examined following the application of K+ channel retardants. The effects of aspirin on pericyte K+ channels were also evaluated with the whole­cell patch­clamp technique. The results demonstrated that pericytes were desmin positive, and their nuclei were large and surrounded by a small proportion of the cytoplasm. Cytoplasmic processes gradually declined in size as branches grew parallel to the capillary axis. Thus, capillaries were surrounded by tips. The electrophysiological properties of the cochlear pericytes in the stria vascularis of guinea pigs were also determined. The membrane capacitance of the pericytes was 5.9±0.3 pF, while the membrane resistance and resting potential were 2.2±0.3 GΩ and ­30.9±1.2 mV, respectively. The current densities of the pericytes (pA/pF) were 3.2±0.7, 10.6±1.0, 15.7±0.9 and 21.3±1.2 at command voltages of 0, +20, +40, and +60 mV, respectively. The K+ channels were activated when the pericytes were within the range of ­20 mV to +20 mV, particularly at 0 mV. The inhibition rates of the outward current of cochlear pericytes in the stria vascularis of the guinea pigs were determined by administering iberiotoxin (IBTX) and IBTX + 4­aminopyridine. Once the background leakage current was removed, the following inhibition rates were obtained with 3, 10, 30, 300 and 1,000 µmol/l aspirin: 20.8±4.8, 34.1±6.9, 48.2±6.7, 63.6±7.1 and 65.7±8.1%, respectively. The outward current of the cochlear pericytes in the stria vascularis was inhibited by aspirin with a half maximal inhibitory concentration of 24.5±4.5 µmol/l. The membranes of the pericytes in the stria vascularis are characterized by high­conductance calcium­activated K+ (BKCa) and voltage­dependent K+ (KV) channels. The outward current of the cochlear pericytes in the stria vascularis of guinea pigs was inhibited by aspirin in a concentration­dependent manner. In addition, BKCa and KV channels were inhibited by aspirin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antipyretics/adverse effects , Aspirin/adverse effects , Pericytes/drug effects , Potassium Channels/metabolism , Stria Vascularis/drug effects , Animals , Cells, Cultured , Guinea Pigs , Membrane Potentials/drug effects , Patch-Clamp Techniques , Pericytes/cytology , Pericytes/metabolism , Stria Vascularis/cytology , Stria Vascularis/metabolism
15.
Eur Arch Otorhinolaryngol ; 273(12): 4153-4159, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27221387

ABSTRACT

The aim of this study is to investigate the effect of rectal ozone and intratympanic ozone therapy on cisplatin-induced ototoxicity in rats. Eighteen female Wistar albino rats were included in our study. External auditory canal and tympanic membrane examinations were normal in all rats. The rats were randomly divided into three groups. Initially, all the rats were tested with distortion product otoacoustic emissions (DPOAE), and emissions were measured normally. All rats were injected with 5-mg/kg/day cisplatin for 3 days intraperitoneally. Ototoxicy had developed in all rats, as confirmed with DPOAE after 1 week. Rectal and intratympanic ozone therapy group was Group 1. No treatment was administered for the rats in Group 2 as the control group. The rats in Group 3 were treated with rectal ozone. All the rats were tested with DPOAE under general anesthesia, and all were sacrificed for pathological examination 1 week after ozone administration. Their cochleas were removed. The outer hair cell damage and stria vascularis damage were examined. In the statistical analysis conducted, a statistically significant difference between Group 1 and Group 2 was observed in all frequencies according to the DPOAE test. In addition, between Group 2 and Group 3, a statistically significant difference was observed in the DPOAE test. However, a statistically significant difference was not observed between Group 1 and Group 3 according to the DPOAE test. According to histopathological scoring, the outer hair cell damage score was statistically significantly high in Group 2 compared with Group 1. In addition, the outer hair cell damage score was also statistically significantly high in Group 2 compared with Group 3. Outer hair cell damage scores were low in Group 1 and Group 3, but there was no statistically significant difference between these groups. There was no statistically significant difference between the groups in terms of stria vascularis damage score examinations. Systemic ozone gas therapy is effective in the treatment of cell damage in cisplatin-induced ototoxicity. The intratympanic administration of ozone gas does not have any additional advantage over the rectal administration.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Hair Cells, Auditory, Outer/drug effects , Otoacoustic Emissions, Spontaneous/drug effects , Ozone/pharmacology , Animals , Cochlea/drug effects , Cochlea/pathology , Female , Hair Cells, Auditory, Outer/pathology , Random Allocation , Rats, Wistar , Stria Vascularis/drug effects , Stria Vascularis/pathology
16.
Neurosci Lett ; 619: 142-8, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26971701

ABSTRACT

Ototoxicity is one of the most important adverse effects of cisplatin chemotherapy. As a common treatment of acute sensorineural hearing loss, systemic administration of steroids was demonstrated ineffective against cisplatin-induced hearing loss (CIHL) in published studies. The current study aimed to evaluate the potential protective effect of dexamethasone (DEX) encapsulated in polyethyleneglycol-coated polylactic acid (PEG-PLA) nanoparticles (DEX-NPs) against cisplatin-induced hearing loss following systemic administration. DEX was fabricated into PEG-PLA nanoparticles using emulsion and evaporation technique as previously reported. DEX or DEX-NPs was administered intraperitoneally to guinea pigs 1h before cisplatin administration. Auditory brainstem response (ABR) threshold shifts were measured at four frequencies (4, 8, 16, and 24kHz) 1 day before and three days after cisplatin injection. Cochlear morphology was examined to evaluate inner ear injury induced by cisplatin exposure. A single dose of DEX-NPs 1h before cisplatin treatment resulted in a significant preservation of the functional and structural properties of the cochlea, which was equivalent to the effect of multidose (3 days) DEX injection. In contrast, no significant protective effect was observed by single dose injection of DEX. The results of histological examination of the cochleae were consistent with the functional measurements. In conclusion, a single dose DEX-NPs significantly attenuated cisplatin ototoxicity in guinea pigs after systemic administration at both histological and functional levels indicating the potential therapeutic benefits of these nanoparticles for enhancing the delivery of DEX in acute sensorineural hearing loss.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cochlea/drug effects , Dexamethasone/pharmacology , Hearing Loss, Sensorineural/prevention & control , Animals , Antineoplastic Agents/administration & dosage , Cochlea/pathology , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem , Guinea Pigs , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Lactic Acid , Male , Nanoparticles , Polyesters , Polyethylene Glycols , Polymers , Spiral Ganglion/drug effects , Spiral Ganglion/pathology , Stria Vascularis/drug effects , Stria Vascularis/pathology
17.
Sci Rep ; 6: 20903, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26864824

ABSTRACT

We confirmed that ATP is released from cochlear marginal cells in the stria vascular but the cell organelle in which ATP stores was not identified until now. Thus, we studied the ATP-containing cell organelles and suggest that these are lysosomes. Primary cultures of marginal cells of Sprague-Dawley rats aged 1-3 days was established. Vesicles within marginal cells stained with markers were identified under confocal laser scanning microscope and transmission electron microscope (TEM). Then ATP release from marginal cells was measured after glycyl-L-phenylalanine-ß- naphthylamide (GPN) treatment using a bioluminescent assay. Quinacrine-stained granules within marginal cells were labeled with LysoTracker, a lysosome tracer, and lysosomal-associated membrane protein 1(LAMP1), but not labeled with the mitochondrial tracer MitoTracker. Furthermore, LysoTracker-labelled puncta showed accumulation of Mant-ATP, an ATP analog. Treatment with 200 µM GPN quenched fluorescently labeled puncta after incubation with LysoTracker or quinacrine, but not MitoTracker. Quinacrine-labeled organelles observed by TEM were lysosomes, and an average 27.7 percent increase in ATP luminescence was observed in marginal cells extracellular fluid after GPN treatment. ATP-containing vesicles in cochlear marginal cells of the stria vascular from neonatal rats are likely lysosomes. ATP release from marginal cells may be via Ca(2+)-dependent lysosomal exocytosis.


Subject(s)
Adenosine Triphosphate/metabolism , Cytoplasm/metabolism , Cytoplasmic Vesicles/metabolism , Lysosomes/metabolism , Stria Vascularis/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Amines/chemistry , Amines/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Cytoplasm/drug effects , Cytoplasm/ultrastructure , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/ultrastructure , Dipeptides/pharmacology , Exocytosis , Gene Expression , Luminescent Measurements , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/drug effects , Lysosomes/ultrastructure , Microscopy, Electron, Scanning , Organic Chemicals/chemistry , Organic Chemicals/metabolism , Primary Cell Culture , Quinacrine/chemistry , Quinacrine/metabolism , Rats , Rats, Sprague-Dawley , Staining and Labeling , Stria Vascularis/cytology , Stria Vascularis/drug effects , ortho-Aminobenzoates/pharmacology
18.
Cell Tissue Res ; 362(2): 307-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26033221

ABSTRACT

Hearing loss secondary to diabetes remains under debate. In our study, we used Zucker Diabetic Fatty (ZDF) rats as an animal model of type 2 diabetes to investigate whether (1) hearing ability impairment and structural alterations of the inner ear occur in diabetes and (2) an angiotensin II receptor blocker (losartan) can protect rats from diabetic damage. Homozygous mutants were treated with a placebo or losartan and heterozygous animals served as non-diabetic controls. All animals underwent immunohistochemical and electronmicroscopical analysis. Functional testing of hearing ability was performed by click-evoked auditory brainstem responses. The present study showed significant sensorineural hearing impairment in placebo-treated diabetic rats (hearing threshold, 45.0 ± 2.1 dB SPL) compared to both non-diabetic controls (34.7 ± 4 dB SPL) and losartan-treated diabetic rats (36.1 ± 7.4 dB SPL). Concurrently, the functional decline in the placebo-treated rats was associated with significant morphological abnormalities, particularly in the intermediate cells of the stria vascularis and with strial dysfunction. These degenerative changes were indicated by the down-regulation of several pumps, ionic and cellular channels, which are involved in the cycling of K(+) and the maintenance of the endocochlear potential essential for the hearing process. Thus, the inner ear can be regarded as a target organ during hyperglycemic disorders and a metabolically induced "diabetic otopathy" may be added to angiopathy, nephropathy and neuropathy as a specific complication of diabetes mellitus. Blockade of the angiotensin II receptor can prevent this "diabetic otopathy" despite hyperglycemic serum levels.


Subject(s)
Diabetes Mellitus, Type 2/complications , Hearing Loss/drug therapy , Hyperglycemia/complications , Labyrinth Diseases/drug therapy , Losartan/pharmacology , Stria Vascularis/drug effects , Animals , Diabetes Mellitus, Experimental , Rats , Rats, Zucker
19.
PLoS One ; 9(7): e102133, 2014.
Article in English | MEDLINE | ID: mdl-25013956

ABSTRACT

Noise-induced hearing loss is at least in part due to disruption of endocochlear potential, which is maintained by various K(+) transport apparatuses including Na(+), K(+)-ATPase and gap junction-mediated intercellular communication in the lateral wall structures. In this study, we examined the changes in the ion-trafficking-related proteins in the spiral ligament fibrocytes (SLFs) following in vivo acoustic overstimulation or in vitro exposure of cultured SLFs to 4-hydroxy-2-nonenal, which is a mediator of oxidative stress. Connexin (Cx)26 and Cx30 were ubiquitously expressed throughout the spiral ligament, whereas Na(+), K(+)-ATPase α1 was predominantly detected in the stria vascularis and spiral prominence (type 2 SLFs). One-hour exposure of mice to 8 kHz octave band noise at a 110 dB sound pressure level produced an immediate and prolonged decrease in the Cx26 expression level and in Na+, K(+)-ATPase activity, as well as a delayed decrease in Cx30 expression in the SLFs. The noise-induced hearing loss and decrease in the Cx26 protein level and Na(+), K(+)-ATPase activity were abolished by a systemic treatment with a free radical-scavenging agent, 4-hydroxy-2,2,6,6-tetramethylpiperidine 1-oxyl, or with a nitric oxide synthase inhibitor, N(ω)-nitro-L-arginine methyl ester hydrochloride. In vitro exposure of SLFs in primary culture to 4-hydroxy-2-nonenal produced a decrease in the protein levels of Cx26 and Na(+), K(+)-ATPase α1, as well as Na(+), K(+)-ATPase activity, and also resulted in dysfunction of the intercellular communication between the SLFs. Taken together, our data suggest that disruption of the ion-trafficking system in the cochlear SLFs is caused by the decrease in Cxs level and Na(+), K(+)-ATPase activity, and at least in part involved in permanent hearing loss induced by intense noise. Oxidative stress-mediated products might contribute to the decrease in Cxs content and Na(+), K(+)-ATPase activity in the cochlear lateral wall structures.


Subject(s)
Aldehydes/pharmacology , Free Radical Scavengers/pharmacology , Hearing Loss, Noise-Induced/prevention & control , NG-Nitroarginine Methyl Ester/pharmacology , Piperidines/pharmacology , Spiral Ligament of Cochlea/metabolism , Aldehydes/antagonists & inhibitors , Animals , Cell Communication/drug effects , Connexin 26 , Connexin 30 , Connexins/antagonists & inhibitors , Connexins/genetics , Connexins/metabolism , Free Radicals/antagonists & inhibitors , Free Radicals/metabolism , Gene Expression Regulation , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/metabolism , Ion Transport/drug effects , Male , Mice , Mice, Transgenic , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Noise/adverse effects , Oxidative Stress/drug effects , Primary Cell Culture , Signal Transduction , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Spiral Ligament of Cochlea/drug effects , Spiral Ligament of Cochlea/pathology , Stria Vascularis/drug effects , Stria Vascularis/metabolism , Stria Vascularis/pathology
20.
Auris Nasus Larynx ; 41(3): 249-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24411828

ABSTRACT

OBJECTIVE: This study was conducted to evaluate the relationship between hearing and cochlear histopathology after arginine vasopressin administration in rats. METHODS: A total of 30 Wistar rats were injected with either 0.02 unit/g of arginine vasopressin or the same amount of isotonic saline solution. The initial auditory brain stem response threshold was recorded and additional measurements were made at 10, 30, 60, and 90 min after injection of arginine vasopressin or isotonic saline solution. The threshold for each timepoint was compared with the initial threshold. Histological quantitative assessment of endolymphatic hydrops in the cochlea was performed using light microscopy and assessment of the basal, intermediate, and marginal cells of the stria vascularis was performed with electron microscopy. RESULTS: The auditory brain stem threshold 60 min after arginine vasopressin injection increased significantly in comparison with the initial threshold (P<0.05). Although the index for endolymphatic hydrops in rats administered arginine vasopressin was not different from that in controls (P>0.05), vacuoles in the intermediate cells were increased significantly in the treated rats (P<0.01). CONCLUSION: Hearing impairment was detected without endolymphatic hydrops in rats administered arginine vasopressin. An increase of vacuoles in the intermediate cells may account for the hearing impairment induced by arginine vasopressin injection.


Subject(s)
Arginine Vasopressin/pharmacology , Cochlea/drug effects , Endolymphatic Hydrops/chemically induced , Evoked Potentials, Auditory, Brain Stem/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Auditory Threshold/drug effects , Cochlea/pathology , Cochlea/ultrastructure , Endolymphatic Hydrops/pathology , Evoked Potentials, Auditory, Brain Stem/physiology , Microscopy, Electron , Rats , Rats, Wistar , Stria Vascularis/drug effects , Stria Vascularis/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...