Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Dev Biol ; 453(2): 191-205, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31185200

ABSTRACT

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


Subject(s)
Cell Lineage , Glycogen Synthase Kinase 3 beta/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/enzymology , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Lineage/drug effects , Cell Proliferation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Female , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hair Cells, Auditory/drug effects , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/enzymology , Hair Cells, Auditory, Outer/cytology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/enzymology , Mice , Models, Biological , Protein Kinase Inhibitors/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Wnt Signaling Pathway/drug effects
2.
Am J Hum Genet ; 98(6): 1266-1270, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259055

ABSTRACT

By genetic linkage analysis in a large consanguineous Iranian family with eleven individuals affected by severe to profound congenital deafness, we were able to define a 2.8 Mb critical interval (at chromosome 1p21.2-1p21.1) for an autosomal-recessive nonsyndromic deafness locus (DFNB). Whole-exome sequencing allowed us to identify a CDC14A biallelic nonsense mutation, c.1126C>T (p.Arg376(∗)), which was present in the eight clinically affected individuals still alive. Subsequent screening of 115 unrelated individuals affected by severe or profound congenital deafness of unknown genetic cause led us to identify another CDC14A biallelic nonsense mutation, c.1015C>T (p.Arg339(∗)), in an individual originating from Mauritania. CDC14A encodes a protein tyrosine phosphatase. Immunofluorescence analysis of the protein distribution in the mouse inner ear showed a strong labeling of the hair cells' kinocilia. By using a morpholino strategy to knockdown cdc14a in zebrafish larvae, we found that the length of the kinocilia was reduced in inner-ear hair cells. Therefore, deafness caused by loss-of-function mutations in CDC14A probably arises from a morphogenetic defect of the auditory sensory cells' hair bundles, whose differentiation critically depends on the proper growth of their kinocilium.


Subject(s)
Cilia/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/etiology , Mutation/genetics , Phosphoric Monoester Hydrolases/genetics , Severity of Illness Index , Adult , Aged , Animals , Cilia/metabolism , Female , Fluorescent Antibody Technique , Hair Cells, Auditory/enzymology , Hearing Loss, Sensorineural/pathology , Humans , Larva/genetics , Larva/growth & development , Male , Mice , Middle Aged , Pedigree , Protein Tyrosine Phosphatases , Young Adult , Zebrafish/genetics , Zebrafish/growth & development
3.
Arch Toxicol ; 89(3): 415-21, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24798086

ABSTRACT

Ceramide, a sphingolipid metabolite, regulates diverse cellular processes including apoptosis, cell senescence, the cell cycle, and cellular differentiation. Exogenously administered ceramide reportedly increased cochlear hair cell death due to gentamicin-induced ototoxicity. Ceramide is mainly generated via a ceramide/sphingomyelin cycle by sphingomyelinase and sphingomyelin synthase or via de novo synthesis by serine palmitoyltransferase and ceramide synthase. This study was designed to investigate the possible involvement of neutral sphingomyelinase, sphingomyelin synthase, or serine palmitoyltransferase in hair cell death due to gentamicin. The basal turns of the organ of Corti of Sprague-Dawley rats were dissected on postnatal days 3-5. Cochlear cultures were exposed to media containing 35 µM gentamicin for 48 h to assess the effects of GW4869 (a neutral sphingomyelinase inhibitor), 2-hydroxyoleic acid (a sphingomyelin synthase activator), and myriocin (a serine palmitoyltransferase inhibitor). Hair cell loss was significantly decreased in the presence of GW4869 or 2-hydroxyoleic acid. Myriocin had no significant effects against gentamicin-induced hair cell loss. In addition, neutral sphingomyelinase was activated by gentamicin exposure. The present findings strongly suggest that the ceramide/sphingomyelin cycle plays an important role in the protection of hair cells against gentamicin-induced ototoxicity.


Subject(s)
Ceramides/biosynthesis , Gentamicins/toxicity , Hair Cells, Auditory/drug effects , Sphingomyelins/biosynthesis , Aniline Compounds/pharmacology , Animals , Animals, Newborn , Benzylidene Compounds/pharmacology , Cell Death/drug effects , Cells, Cultured , Fatty Acids, Monounsaturated/pharmacology , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Oleic Acids/pharmacology , Rats, Sprague-Dawley , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Transferases (Other Substituted Phosphate Groups)/metabolism
4.
Cell Tissue Res ; 352(3): 469-78, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584706

ABSTRACT

Several studies have demonstrated a link between diabetes and the dysfunction of the inner ear. Few studies, however, have reported the signalling mechanisms involved in metabolic control in human inner ear cells. Knowledge of the expression and role of the insulin receptor and downstream signalling components in the inner ear is sparce. Our immunohistochemistry approach has shown that the insulin receptor, insulin receptor substrate 1 (IRS1), protein kinase B (PKB) and insulin-sensitive glucose transporter (GLUT4) are expressed in the sensory epithelium of the human saccule, which also exhibits expression of a calcium-sensitive cAMP/cGMP phosphodiesterase 1C (PDE1C) and the vasopressin type 2 receptor. IRS1 and PDE1C are selectively expressed in sensory epithelial hair cells, whereas the other components are expressed in sensory epithelial supporting cells or in both cell types, as judged from co-expression or non-co-expression with glial fibrillary acidic protein, a marker for supporting cells. Furthermore, IRS1 appears to be localized in association with sensory nerves, whereas GLUT4 is expressed in the peri-nuclear area of stromal cells, as is the case for aquaporin 2. Thus, the insulin receptor, insulin signalling components and selected cAMP signalling components are expressed in the human saccule. In addition to well-known mechanisms of diabetes complications, such as neuropathy and vascular lesions, the expression of these proteins in the saccule could have a role in the observed link between diabetes and balance/hearing disorders.


Subject(s)
Epithelium/metabolism , Insulin/metabolism , Saccule and Utricle/metabolism , Sensation , Signal Transduction , Aquaporin 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Glucose Transporter Type 4/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/enzymology , Humans , Insulin Receptor Substrate Proteins/metabolism , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Receptors, Vasopressin/metabolism , Saccule and Utricle/cytology , Stromal Cells/cytology , Stromal Cells/metabolism
5.
Neuron ; 53(3): 371-86, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17270734

ABSTRACT

When stimulated strongly, a hair cell's mechanically sensitive hair bundle may consume ATP too rapidly for replenishment by diffusion. To provide a broad view of the bundle's protein complement, including those proteins participating in energy metabolism, we used shotgun mass spectrometry methods to identify proteins of purified chicken vestibular bundles. In addition to cytoskeletal proteins, proteins involved in Ca(2+) regulation, and stress-response proteins, many of the most abundant bundle proteins that were identified by mass spectrometry were involved in ATP synthesis. After beta-actin, the cytosolic brain isoform of creatine kinase was the next most abundant bundle protein; at approximately 0.5 mM, creatine kinase is capable of maintaining high ATP levels despite 1 mM/s ATP consumption by the plasma-membrane Ca(2+)-ATPase. Consistent with this critical role in hair bundle function, the creatine kinase circuit is essential for high-sensitivity hearing as demonstrated by hearing loss in creatine kinase knockout mice.


Subject(s)
Adenosine Triphosphate/metabolism , Chickens/physiology , Creatine Kinase/metabolism , Hair Cells, Auditory/metabolism , Animals , Brain/enzymology , Creatine Kinase/genetics , Cytosol/enzymology , Ear, Inner/enzymology , Ear, Inner/metabolism , Energy Metabolism/physiology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Hair Cells, Auditory/enzymology , Hearing/physiology , Immunohistochemistry , Isoenzymes/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/metabolism , Postural Balance/physiology , Rana catesbeiana , Saccule and Utricle/cytology , Saccule and Utricle/enzymology , Saccule and Utricle/metabolism , Signal Transduction/physiology
6.
Biochem Biophys Res Commun ; 415(1): 94-8, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22020098

ABSTRACT

Expression of antioxidant enzymes is regulated by transcription factor NF-E2-related factor (Nrf2) and induced by oxidative stress. Reactive oxygen species contribute to the formation of several types of cochlear injuries, including age-related hearing loss and gentamicin ototoxicity. In this study, we examined the roles of Nrf2 in age-related hearing loss and gentamicin ototoxicity by measuring auditory brainstem response thresholds in Nrf2-knockout mice. Although Nrf2-knockout mice maintained normal auditory thresholds at 3 months of age, their hearing ability was significantly more impaired than that of age-matched wild-type mice at 6 and 11 months of age. Additionally, the numbers of hair cells and spiral ganglion cells were remarkably reduced in Nrf2-knockout mice at 11 months of age. To examine the importance of Nrf2 in protecting against gentamicin-induced ototoxicity, 3-day-old mouse organ of Corti explants were cultured with gentamicin. Hair cell loss caused by gentamicin treatment was enhanced in the Nrf2-deficient tissues. Furthermore, the expressions of some Nrf2-target genes were activated by gentamicin treatment in wild-type mice but not in Nrf2-knockout mice. The present findings indicate that Nrf2 protects the inner ear against age-related hearing injuries and gentamicin ototoxicity by up-regulating antioxidant enzymes and detoxifying proteins.


Subject(s)
Aging , Anti-Bacterial Agents/adverse effects , Ear, Inner/enzymology , Gentamicins/adverse effects , Hearing Loss/chemically induced , Hearing Loss/genetics , NF-E2-Related Factor 2/physiology , Animals , Ear, Inner/drug effects , Gene Expression Regulation, Developmental , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/enzymology , Heme Oxygenase-1/genetics , Mice , Mice, Knockout , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , Response Elements , Spiral Ganglion/drug effects , Spiral Ganglion/enzymology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
7.
BMC Neurosci ; 12: 114, 2011 Nov 14.
Article in English | MEDLINE | ID: mdl-22082490

ABSTRACT

BACKGROUND: Inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, known as statins, are commonly used as cholesterol-lowering drugs. During the past decade, evidence has emerged that statins also have neuroprotective effects. Research in the retina has shown that simvastatin, a commonly used statin, increases Akt phosphorylation in vivo, indicating that the PI3K/Akt pathway contributes to the protective effects achieved. While research about neuroprotective effects have been conducted in several systems, the effects of statins on the inner ear are largely unknown. RESULTS: We evaluated whether the 3-hydroxy-3-methylglutaryl-coenzyme A reductase is present within the rat cochlea and whether simvastatin is able to protect auditory hair cells from gentamicin-induced apoptotic cell death in a in vitro mouse model. Furthermore, we evaluated whether simvastatin increases Akt phosphorylation in the organ of Corti. We detected 3-hydroxy-3-methylglutaryl-coenzyme A reductase mRNA in organ of Corti, spiral ganglion, and stria vascularis by reverse transcriptase-polymerase chain reaction (RT-PCR). Moreover, we observed a dose-dependent and significant reduction of hair cell loss in organs of Corti treated with simvastatin in addition to gentamicin, as compared to samples treated with gentamicin alone. The protective effect of simvastatin was reversed by addition of mevalonate, a downstream metabolite blocked by simvastatin, demonstrating the specificity of protection. Finally, Western blotting showed an increase in organ of Corti Akt phosphorylation after simvastatin treatment in vitro. CONCLUSION: These results suggest a neuroprotective effect of statins in the inner ear, mediated by reduced 3-hydroxy-3-methylglutaryl-coenzyme A reductase metabolism and Akt activation.


Subject(s)
Gentamicins/toxicity , Hair Cells, Auditory/drug effects , Hearing Loss, Sensorineural/drug therapy , Neuroprotective Agents/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Simvastatin/toxicity , Animals , Animals, Newborn , Disease Models, Animal , Gentamicins/antagonists & inhibitors , Hair Cells, Auditory/enzymology , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/physiopathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Mice , Mice, Transgenic , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/physiology , Rats , Rats, Wistar , Signal Transduction/physiology
8.
Nat Cell Biol ; 5(5): 422-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12717441

ABSTRACT

Maintenance of the post-mitotic state in the post-natal mammalian brain is an active process that requires the cyclin-dependent kinase inhibitors (CKIs) p19Ink4d (Ink4d) and p27Kip1 (Kip1). In animals with targeted deletions of both Ink4d and Kip1, terminally differentiated, post-mitotic neurons are observed to re-enter the cell cycle, divide and undergo apoptosis. However, when either Ink4d or Kip1 alone are deleted, the post-mitotic state is maintained, suggesting a redundant role for these genes in mature neurons. In the organ of Corti--the auditory sensory epithelium of mammals--sensory hair cells and supporting cells become post-mitotic during embryogenesis and remain quiescent for the life of the animal. When lost as a result of environmental insult or genetic abnormality, hair cells do not regenerate, and this loss is a common cause of deafness in humans. Here, we report that targeted deletion of Ink4d alone is sufficient to disrupt the maintenance of the post-mitotic state of sensory hair cells in post-natal mice. In Ink4d-/- animals, hair cells are observed to aberrantly re-enter the cell cycle and subsequently undergo apoptosis, resulting in progressive hearing loss. Our results identify a novel mechanism underlying a non-syndromic form of progressive hearing loss in mice.


Subject(s)
Apoptosis/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Hair Cells, Auditory/enzymology , Hearing Loss/enzymology , Hearing Loss/genetics , Nerve Regeneration/genetics , Tumor Suppressor Proteins/deficiency , Animals , Caspase 3 , Caspases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Division/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p19 , Cyclin-Dependent Kinase Inhibitor p27 , Dyneins , Fetus , Fluorescent Antibody Technique , Hair Cells, Auditory/ultrastructure , Homeostasis/genetics , Mice , Mice, Knockout , Myosin VIIa , Myosins/metabolism , Tumor Suppressor Proteins/genetics
9.
Mol Cell Biochem ; 353(1-2): 291-303, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21503676

ABSTRACT

In the present study, we aim to explore whether the caspase-3-dependent pathway is involved in the apoptotic cell death that occurs in the hair cells (HCs) of guinea pig cochlea following a salicylate treatment. Guinea pigs received sodium salicylate (Na-SA), at a dose of 200 mg·kg(-1)·d(-1) i.p., as a vehicle for 5 consecutive days. In some experiments, N-benzyloxycarbonyl-Asp-Glu-Val-Asp-fluoromethylketone (zDEVD-FMK), a specific apoptosis inhibitor, was directly applied into the cochlea via the round window niche (RWN) prior to salicylate treatment for determination of caspase-3 activation. Alterations in auditory function were evaluated with auditory brainstem responses (ABR) thresholds. Caspase-3 activity was determined by measuring the proteolytic cleavage product of caspase-3 (N-terminated peptide substrate). DNA fragmentation within the nuclei was examined with a terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) method. Ultrastructure variation in the target cell was assessed by electron microscopy (EM). Salicylate treatment initiated an obvious elevation in ABR thresholds with a maximum average shift of 60 dB sound pressure level (SPL), and caused significant apoptosis in both inner (IHCs) and outer (OHCs) hair cells resulted from an evident increasing in immunoreactivity to caspase-3 protease. Transmission electron microscopy (TEM) displayed chromatin condensation and nucleus margination accompanied by cell body shrinkage in the OHCs, but not in the IHCs. Scanning electron microscopy (SEM) showed breakdown, fusion, and loss in the stereociliary bundles at the apex of OHCs rather than IHCs. zDEVD-FMK pretreatment prior to salicylate injection substantially attenuated an expression of the apoptotic protease and protected HCs against apoptotic death, followed by a moderate relief in the thresholds of ABR, an alleviation in the submicroscopic structure was also identified. In particular, disorientation and insertion in the hair bundles at the apex of OHCs was exhibited though no classic apoptotic change found. The above changes were either prevented or significantly attenuated by zDEVD-FMK. These findings indicate that salicylate could damage cochlear hair cells via inducing apoptosis associated with caspase-3 activation.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Hair Cells, Auditory/drug effects , Oligopeptides/pharmacology , Salicylates/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Auditory Threshold/drug effects , Caspase 3/metabolism , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation/drug effects , Guinea Pigs , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/ultrastructure , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/enzymology , Hair Cells, Auditory, Inner/ultrastructure , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/enzymology , Hair Cells, Auditory, Outer/ultrastructure , Immunohistochemistry , In Situ Nick-End Labeling , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Signal Transduction/drug effects
10.
Toxicol Lett ; 349: 115-123, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34089817

ABSTRACT

Cisplatin, the most widely used platinum-based anticancer drug, often causes progressive and irreversible sensorineural hearing loss in cancer patients. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. Nicotinamide adenine dinucleotide (NAD+), a co-substrate for the sirtuin family and PARPs, has emerged as a potent therapeutic molecular target in various diseases. In our investigates, we observed that NAD+ level was changed in the cochlear explants of mice treated with cisplatin. Supplementation of a specific inhibitor (TES-1025) of α-amino-ß-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), a rate-limiting enzyme of NAD+de novo synthesis pathway, promoted SIRT1 activity, increased mtDNA contents and enhanced AMPK expression, thus significantly reducing hair cells loss and deformation. The protection was blocked by EX527, a specific SIRT1 inhibitor. Meanwhile, the use of NMN, a precursor of NAD+ salvage synthesis pathway, had shown beneficial effect on hair cell under cisplatin administration, effectively suppressing PARP1. In vivo experiments confirmed the hair cell protection of NAD+ modulators in cisplatin treated mice and zebrafish. In conclusion, we demonstrated that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway could both prevent ototoxicity of cisplatin. These results suggested that direct modulation of cellular NAD+ levels could be a promising therapeutic approach for protection of hearing from cisplatin-induced ototoxicity.


Subject(s)
Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/drug effects , Hearing Loss/prevention & control , Hearing/drug effects , NAD/biosynthesis , Ototoxicity/prevention & control , Sirtuin 1/metabolism , Animals , Animals, Genetically Modified , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/metabolism , Cisplatin , Disease Models, Animal , Enzyme Activation , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/pathology , Hearing Loss/chemically induced , Hearing Loss/enzymology , Hearing Loss/physiopathology , Lateral Line System/drug effects , Lateral Line System/enzymology , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Ototoxicity/enzymology , Ototoxicity/etiology , Ototoxicity/physiopathology , Zebrafish
11.
J Neurosci ; 29(50): 15859-69, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016102

ABSTRACT

Morphogenesis of sensory hair cells, in particular their mechanotransduction organelle, the stereociliary bundle, requires highly organized remodeling of the actin cytoskeleton. The roles of Rho family small GTPases during this process remain unknown. Here we show that deletion of Rac1 in the otic epithelium resulted in severe defects in cochlear epithelial morphogenesis. The mutant cochlea was severely shortened with a reduced number of auditory hair cells and cellular organization of the auditory sensory epithelium was abnormal. Rac1 mutant hair cells also displayed defects in planar cell polarity and morphogenesis of the stereociliary bundle, including bundle fragmentation or deformation, and mispositioning or absence of the kinocilium. We further demonstrate that a Rac-PAK (p21-activated kinase) signaling pathway mediates kinocilium-stereocilia interactions and is required for cohesion of the stereociliary bundle. Together, these results reveal a critical function of Rac1 in morphogenesis of the auditory sensory epithelium and stereociliary bundle.


Subject(s)
Hair Cells, Auditory/enzymology , Hair Cells, Auditory/physiology , Morphogenesis/physiology , Neuropeptides/physiology , rac GTP-Binding Proteins/physiology , Animals , Animals, Newborn , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Mice , Mice, Knockout , Morphogenesis/genetics , Neuropeptides/deficiency , Neuropeptides/genetics , Organ of Corti/cytology , Organ of Corti/growth & development , Organ of Corti/physiology , Pregnancy , rac GTP-Binding Proteins/deficiency , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
12.
Toxicol Lett ; 313: 11-18, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31220555

ABSTRACT

Previous study reported that either selective GSK-3ß inhibitor or up-regulating autophagy can alleviate cisplatin-induced ototoxicity. Other studies indicate that the activity of GSK-3ß is closely associated with the autophagy level. The purpose of this study is to primarily explore the role of autophagy in the alleviation effect of GSK-3ß inhibition on cisplatin-induced ototoxicity in vivo and in vitro. We observed the autophagy changes induced by GSK-3ß inhibitor in outer hair cells (OHCs) in a cisplatin-induced ototoxicity rat model. In addition, autophagy inhibitor 3-MA was used in vitro experiments to observe the influence of autophagy inhibition on the cell protection effect due to GSK-3ß inactivation. The relationship among autophagy, GSK-3ß and cell damage were inferred. Negative regulation of GSK-3ß significantly enhanced autophagy and alleviated cisplatin-induced hearing loss, OHC death in vivo and apoptosis in vitro. The autophagy inhibitor 3-MA inverted the protective effect of negative regulation of GSK-3ß. These results indicated that enhancing autophagy may be a key downstream effect of GSK-3ß inhibition in the alleviation of cisplatin-induced ototoxicity both in vivo and in vitro.


Subject(s)
Autophagy/drug effects , Cisplatin , Ear Diseases/drug therapy , Glycogen Synthase Kinase 3 beta/metabolism , Hair Cells, Auditory/drug effects , Lithium Chloride/pharmacology , Animals , Auditory Fatigue/drug effects , Cell Line , Disease Models, Animal , Down-Regulation , Ear Diseases/chemically induced , Ear Diseases/enzymology , Ear Diseases/pathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/pathology , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects
13.
Neuron ; 11(4): 581-94, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8398149

ABSTRACT

By adapting to sustained stimuli, hair cells of the internal ear maintain their optimal sensitivity to minute displacements. Biophysical experiments have suggested that adaptation is mediated by a molecular motor, most likely a member of the myosin family. To provide direct evidence for the presence of myosin isozymes in hair bundles, we used photoaffinity labeling with vanadate-trapped uridine and adenine nucleotides to identify proteins of 120, 160, and 230 kd in a preparation of hair bundles purified from the bullfrog's sacculus. The photoaffinity labeling properties of these proteins, particularly the 120 kd protein, resembled those of other well-characterized myosins. A 120 kd hair-bundle protein was also recognized by a monoclonal antibody directed against a vertebrate myosin I isozyme. Immunofluorescence microscopy localized this protein near the beveled top edge of the hair bundle, the site of mechanoelectrical transduction and adaptation.


Subject(s)
Cilia/chemistry , Hair Cells, Auditory/chemistry , Myosins/isolation & purification , Adenosine Triphosphate/metabolism , Animals , Antibodies, Monoclonal , Autoradiography , Calcium/pharmacology , Cilia/enzymology , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Hair Cells, Auditory/enzymology , Isoenzymes/analysis , Isoenzymes/isolation & purification , Molecular Weight , Myosins/analysis , Phosphorus Radioisotopes , Rana catesbeiana , Uridine Triphosphate/metabolism , Vanadates/pharmacology
14.
Neuron ; 17(3): 523-33, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8816715

ABSTRACT

To ensure optimal sensitivity for mechanoelectrical transduction, hair cells adapt to prolonged stimuli using active motors. Adaptation motors are thought to employ myosin molecules as their force-producing components. We find that beryllium fluoride, vanadate, and sulfate, phosphate analogs that inhibit the ATPase activity of myosin, inhibit adaptation by abolishing motor force production. Phosphate analogs interact with a 120-kDa bundle protein, most likely myosin 1 beta, in a manner that coincides with their effects on adaptation. Features of transduction following inhibition of motor force production suggest that the gating and extent springs of the hair cell orient in parallel at rest and that the negative limit of adaptation arises when force in the stretched extent spring matches the force output of the adaptation motor.


Subject(s)
Adaptation, Physiological/physiology , Hair Cells, Auditory/enzymology , Phosphates/physiology , Adaptation, Physiological/drug effects , Beryllium/pharmacology , Electrophysiology , Fluorides/pharmacology , Hair Cells, Auditory/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Microdialysis , Models, Biological , Myosins/antagonists & inhibitors , Myosins/chemistry , Photochemistry , Signal Transduction/physiology , Sulfates/pharmacology , Vanadates/pharmacology
15.
Apoptosis ; 13(9): 1184-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18670884

ABSTRACT

Cisplatin, a chemotherapeutic drug that is widely used to treat various cancers, promotes ototoxicity at higher doses. In this study, the effect of epicatechin (EC) on cisplatin-induced hair cell death was investigated in a cochlear organ of Corti-derived cell line, HEI-OC1, and in vivo in zebrafish. Cisplatin promoted apoptosis and altered mitochondrial membrane potential (MMP) in HEI-OC1 cells. EC inhibited cisplatin-induced apoptosis and intracellular reactive oxygen species (ROS) generation. Labeling of zebrafish lateral line hair cells by the fluorescent dye YO-PRO1 was lost upon exposure to cisplatin, and EC protected against this cisplatin-induced loss of labeling in a dose-dependent manner. Scanning and transmission electron micrographs showed that treatment with EC protected against cisplatin-induced loss of kinocilium and stereocilia in zebrafish neuromasts. These results suggest that EC prevents cisplatin-induced ototoxicity by blocking ROS generation and by preventing changes in MMP.


Subject(s)
Catechin/pharmacology , Cisplatin/pharmacology , Cytoprotection/drug effects , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Animals , Caspase 3/metabolism , Catechin/chemistry , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cisplatin/antagonists & inhibitors , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/ultrastructure , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Zebrafish
16.
J Neurotrauma ; 25(1): 72-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18355160

ABSTRACT

Noise exposure is known to induce cell death signaling in the cochlea. Since c-Jun N-terminal kinase (JNK) signaling is known to induce both cell survival and apoptosis, the present study focused on early changes (within 24 h) after impulse noise exposure, inquiring whether cell death is always related to phosphorylation of JNK in the inner ear. Anesthetized adult albino rats were exposed to a single impulse noise exposure (194 kPa) and sacrificed 3 or 24 h later. Paraffin-embedded sections were examined for positive staining of phosphorylated JNK and the presence of cells with fragmented DNA (TUNEL staining). Positive TUNEL staining was observed at the spiral limbus and in the stria vascularis at 24 h following impulse noise exposure, but no correlation with JNK activation was found at these locations. In the hearing organ (organ of Corti) and in the lateral wall, TUNEL-reactive cells were observed at 24 h following trauma. This was preceded by p-JNK staining at 3 h, indicating JNK-activated cell death in these regions. Finally, p-JNK reactivity was observed in the spiral ganglion with no correlation to TUNEL staining within the time frame of this study. These results suggest that JNK activation following impulse noise exposure may not always be related to cell death, and conversely, some cells may die through JNK-independent signaling.


Subject(s)
Cochlea/enzymology , Hair Cells, Auditory/enzymology , Hearing Loss, Noise-Induced/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Noise/adverse effects , Acoustic Stimulation , Animals , Biomarkers/metabolism , Cell Death/physiology , Cochlea/pathology , Cochlea/physiopathology , DNA Fragmentation , Enzyme Activation/physiology , Female , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/physiopathology , In Situ Nick-End Labeling , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Spiral Ganglion/enzymology , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology , Time Factors
17.
Hear Res ; 240(1-2): 1-11, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18487027

ABSTRACT

Aminoglycoside antibiotics induce caspase-dependent apoptotic death in cochlear hair cells. Apoptosis, a regulated form of cell death, can be induced by many stressors, which activate signaling pathways that result in the controlled dismantling of the affected cell. The caspase family of proteases is activated in the apoptotic signaling pathway and is responsible for cellular destruction. The initiator caspase-9 and the effector caspase-3 are both activated in chick cochlear hair cells following aminoglycoside exposure. We have analyzed caspase activation in the avian cochlea during gentamicin-induced hair cell death to compare two different methods of caspase detection: caspase antibodies and CaspaTag kits. Caspase antibodies bind to the cleaved activated form of caspase-9 or caspase-3 in specific locations in fixed tissue. CaspaTag is a fluorescent inhibitor that binds to a reactive cysteine residue on the large subunit of the caspase heterodimer in unfixed tissue. To induce cochlear hair cell loss, 1-2 week-old chickens received a single injection of gentamicin (300 mg/kg). Chicks were sacrificed 24, 30, 42, 48, 72, or 96 h after injection. Cochleae were dissected and labeled for activated caspase-9 or caspase-3 using either caspase-directed antibodies or CaspaTag kits. Ears were co-labeled with either phalloidin or myosin VI to visualize hair cells and to determine the progression of cochlear damage. The timing of caspase activation was similar for both assays; however, caspase-9 and caspase-3 antibodies labeled only those cells currently undergoing apoptotic cell death. Conversely, CaspaTag-labeled all the cells that have undergone apoptotic cell death and ejection from the sensory epithelium, in addition to those that are currently in the cell death process. This makes CaspaTag ideal for showing an overall pattern or level of cell death over a period of time, while caspase antibodies provide a snapshot of cell death at a specific time point.


Subject(s)
Anti-Bacterial Agents/toxicity , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cochlea/drug effects , Fluorescent Antibody Technique, Indirect , Gentamicins/toxicity , Reagent Kits, Diagnostic , Animals , Caspase Inhibitors , Chickens , Cochlea/enzymology , Cochlea/pathology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Fluorescent Dyes/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/enzymology , Hair Cells, Auditory/pathology , Microscopy, Fluorescence , Time Factors
18.
Hear Res ; 241(1-2): 7-17, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18534795

ABSTRACT

Using video, fluorescence and confocal microscopy, quantitative analysis and modeling, we investigated intracellular processes mediating the calcium/calmodulin (Ca(2+)/CaM)-dependent slow motility in hair cells dissociated from the rostral region of amphibian papilla, one of the two auditory organs in frogs. The time course of shape changes in these hair cells during the period of pretreatment with several specific inhibitors, as well as their response to the calcium ionophore, ionomycin, were recorded and compared. These cells respond to ionomycin with a tri-phasic shape change: an initial phase of iso-volumetric length decrease; a period of concurrent shortening and swelling; and the final phase of increase in both length and volume. We found that both the myosin light chain kinase inhibitor, ML-7, and antagonists of the multifunctional Ca(2+)/CaM-dependent kinases, KN-62 and KN-93, inhibit the iso-volumetric shortening phase of the response to ionomycin. The type 1 protein phosphatase inhibitors, calyculin A and okadaic acid induce minor shortening on their own, but do not significantly alter phase 1 response. However, they appear to counter effects of the inhibitors of Ca(2+)/CaM-dependent kinases. We hypothesize that an active actomyosin-based process mediates the iso-volumetric shortening in the frog rostral amphibian papillar hair cells.


Subject(s)
Cell Movement , Cell Shape , Hair Cells, Auditory/metabolism , Myosin Light Chains/metabolism , Organ of Corti/metabolism , Actomyosin/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Movement/drug effects , Cell Shape/drug effects , Cell Size , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/enzymology , Ionophores/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Video , Models, Biological , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Myosin-Light-Chain Phosphatase/metabolism , Organ of Corti/cytology , Organ of Corti/drug effects , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Rana pipiens , Signal Processing, Computer-Assisted , Time Factors
19.
Int J Mol Med ; 39(4): 960-968, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28290603

ABSTRACT

Fenofibrate, an activator of peroxisome proliferator-activated receptors (PPARs), has been shown to protect the kidneys and brain cells from oxidative stress; however, its role in preventing hearing loss has not been reported to date, at least to the best of our knowledge. In this study, we demonstrated the protective effects of fenofibrate against gentamicin (GM)-induced ototoxicity. We found that the auditory brainstem response threshold which was increased by GM was significantly reduced by pre-treatment with fenofibrate in rats. In cochlear explants, the disruption of hair cell layers by GM was also markedly attenuated by pre-treatment with fenofibrate. In addition, fenofibrate almost completely abolished GM-induced reactive oxygen species generation, which seemed to be mediated at least in part by the restoration of the expression of PPAR­α­dependent antioxidant enzymes, including catalase and superoxide dismutase (SOD)-1. Of note, fenofibrate markedly increased the expression of heme oxygenase-1 (HO-1) which was also induced to a certain degree by GM alone. The induced expression of HO-1 by fenofibrate appeared to be essential for mediating the protective effects of fenofibrate, as the inhibition of HO-1 activity significantly diminished the protective effects of fenofibrate against the GM-mediated death of sensory hair cells in cochlea explant culture, as well as in zebrafish neuromasts. These results suggest that fenofibrate protects sensory hair cells from GM-induced toxicity by upregulating PPAR­α-dependent antioxidant enzymes, including HO-1. Our results provide insight into the preventive therapy for hearing loss caused by aminoglycoside antibiotics.


Subject(s)
Antioxidants/metabolism , Catalase/metabolism , Fenofibrate/pharmacology , Gentamicins/adverse effects , Hair Cells, Auditory/enzymology , Heme Oxygenase (Decyclizing)/metabolism , Superoxide Dismutase-1/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Death , Enzyme Activation/drug effects , Female , Gentamicins/pharmacology , Hair Cells, Auditory/pathology , Male , PPAR alpha/metabolism , Rats , Rats, Sprague-Dawley
20.
J Neurosci ; 23(27): 9208-19, 2003 Oct 08.
Article in English | MEDLINE | ID: mdl-14534255

ABSTRACT

A screen for protein tyrosine phosphatases (PTPs) expressed in the chick inner ear yielded a high proportion of clones encoding an avian ortholog of protein tyrosine phosphatase receptor Q (Ptprq), a receptor-like PTP. Ptprq was first identified as a transcript upregulated in rat kidney in response to glomerular nephritis and has recently been shown to be active against inositol phospholipids. An antibody to the intracellular domain of Ptprq, anti-Ptprq, stains hair bundles in mice and chicks. In the chick ear, the distribution of Ptprq is almost identical to that of the 275 kDa hair-cell antigen (HCA), a component of hair-bundle shaft connectors recognized by a monoclonal antibody (mAb) that stains inner-ear hair bundles and kidney glomeruli. Furthermore, anti-Ptprq immunoblots a 275 kDa polypeptide immunoprecipitated by the anti-HCA mAb from the avian inner ear, indicating that the HCA and Ptprq are likely to be the same molecule. In two transgenic mouse strains with different mutations in Ptprq, anti-Ptprq immunoreactivity cannot be detected in the ear. Shaft connectors are absent from mutant vestibular hair bundles, but the stereocilia forming the hair bundle are not splayed, indicating that shaft connectors are not necessary to hold the stereocilia together; however, the mice show rapid postnatal deterioration in cochlear hair-bundle structure, associated with smaller than normal transducer currents with otherwise normal adaptation properties, a progressive loss of basal-coil cochlear hair cells, and deafness. These results reveal that Ptprq is required for formation of the shaft connectors of the hair bundle, the normal maturation of cochlear hair bundles, and the long-term survival of high-frequency auditory hair cells.


Subject(s)
Cochlea/enzymology , Hair Cells, Auditory/enzymology , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Antigens, Surface/metabolism , Cell Separation , Cell Survival/genetics , Chickens , Cilia/enzymology , Cilia/ultrastructure , Cochlea/cytology , Cochlea/embryology , Deafness/genetics , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Hair Cells, Auditory/ultrastructure , Mechanotransduction, Cellular/physiology , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Patch-Clamp Techniques , Phalloidine , Phosphoric Monoester Hydrolases/genetics , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3
SELECTION OF CITATIONS
SEARCH DETAIL