Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 12(20): 19834-19851, 2020 10 24.
Article in English | MEDLINE | ID: mdl-33099273

ABSTRACT

Foxg1 plays important roles in regeneration of hair cell (HC) in the cochlea of neonatal mouse. Here, we used Sox9-CreER to knock down Foxg1 in supporting cells (SCs) in the utricle in order to investigate the role of Foxg1 in HC regeneration in the utricle. We found Sox9 an ideal marker of utricle SCs and bred Sox9CreER/+Foxg1loxp/loxp mice to conditionally knock down Foxg1 in utricular SCs. Conditional knockdown (cKD) of Foxg1 in SCs at postnatal day one (P01) led to increased number of HCs at P08. These regenerated HCs had normal characteristics, and could survive to at least P30. Lineage tracing showed that a significant portion of newly regenerated HCs originated from SCs in Foxg1 cKD mice compared to the mice subjected to the same treatment, which suggested SCs trans-differentiate into HCs in the Foxg1 cKD mouse utricle. After neomycin treatment in vitro, more HCs were observed in Foxg1 cKD mice utricle compared to the control group. Together, these results suggest that Foxg1 cKD in utricular SCs may promote HC regeneration by inducing trans-differentiation of SCs. This research therefore provides theoretical basis for the effects of Foxg1 in trans-differentiation of SCs and regeneration of HCs in the mouse utricle.


Subject(s)
Cell Transdifferentiation , Forkhead Transcription Factors/deficiency , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/metabolism , Nerve Tissue Proteins/deficiency , SOX9 Transcription Factor/metabolism , Saccule and Utricle/metabolism , Animals , Animals, Newborn , Cell Lineage , Cell Proliferation , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , Male , Mice, Knockout , Neomycin/toxicity , Nerve Tissue Proteins/genetics , Ototoxicity , Phenotype , SOX9 Transcription Factor/genetics , Saccule and Utricle/drug effects , Saccule and Utricle/pathology , Signal Transduction
2.
Sci Rep ; 10(1): 6740, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317718

ABSTRACT

To protect the audiosensory organ from tissue damage from the immune system, the inner ear is separated from the circulating immune system by the blood-labyrinth barrier, which was previously considered an immune-privileged site. Recent studies have shown that macrophages are distributed in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis; however, the direct pathogen defence mechanism used by audiosensory receptor hair cells (HCs) has remained obscure. Here, we show that HCs are protected from pathogens by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs). In isolated murine cochlear sensory epithelium, we established Theiler's murine encephalomyelitis virus, which infected the SCs and GERCs, but very few HCs. The virus-infected SCs produced interferon (IFN)-α/ß, and the viruses efficiently infected the HCs in the IFN-α/ß receptor-null sensory epithelium. Interestingly, the virus-infected SCs and GERCs expressed macrophage marker proteins and were eliminated from the cell layer by cell detachment. Moreover, lipopolysaccharide induced phagocytosis of the SCs without cell detachment, and the SCs phagocytosed the bacteria. These results reveal that SCs function as macrophage-like cells, protect adjacent HCs from pathogens, and provide a novel anti-infection inner ear immune system.


Subject(s)
Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Outer/physiology , Labyrinth Supporting Cells/immunology , Macrophages/immunology , Spiral Ganglion/physiology , Stria Vascularis/physiology , Animals , Animals, Newborn , Escherichia coli/immunology , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Outer/cytology , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interferon-beta/biosynthesis , Interferon-beta/immunology , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/virology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/virology , Mice , Mice, Inbred ICR , Organ Culture Techniques , Phagocytosis/drug effects , Saccharomyces cerevisiae/immunology , Spiral Ganglion/cytology , Stria Vascularis/cytology , Theilovirus/growth & development , Theilovirus/pathogenicity
3.
Hear Res ; 385: 107839, 2020 01.
Article in English | MEDLINE | ID: mdl-31760261

ABSTRACT

Hair cells in the auditory organ of the vertebrate inner ear are the sensory receptors that convert acoustic stimuli into electrical signals that are conveyed along the auditory nerve to the brainstem. Hair cells are highly susceptible to ototoxic drugs, infection, and acoustic trauma, which can cause cellular degeneration. In mammals, hair cells that are lost after damage are not replaced, leading to permanent hearing impairments. By contrast, supporting cells in birds and other non-mammalian vertebrates regenerate hair cells after damage, which restores hearing function. The cellular mechanisms that regulate hair cell regeneration are not well understood. We investigated the role of vascular endothelial growth factor (VEGF) during regeneration of auditory hair cells in chickens after ototoxic injury. Using RNA-Seq, immunolabeling, and in situ hybridization, we found that VEGFA, VEGFC, VEGFR1, VEGFR2, and VEGFR3 were expressed in the auditory epithelium, with VEGFA expressed in hair cells and VEGFR1 and VEGFR2 expressed in supporting cells. Using organotypic cultures of the chicken cochlear duct, we found that blocking VEGF receptor activity during hair cell injury reduced supporting cell proliferation as well as the numbers of regenerated hair cells. By contrast, addition of recombinant human VEGFA to organ cultures caused an increase in both supporting cell division and hair cell regeneration. VEGF's effects on supporting cells were preserved in isolated supporting cell cultures, indicating that VEGF can act directly upon supporting cells. These observations demonstrate a heretofore uncharacterized function for VEGF signaling as a critical positive regulator of hair cell regeneration in the avian inner ear.


Subject(s)
Avian Proteins/metabolism , Cell Proliferation , Hair Cells, Auditory, Inner/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Regeneration , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis , Avian Proteins/genetics , Cell Proliferation/drug effects , Cells, Cultured , Chickens , Gene Expression Regulation , Hair Cells, Auditory, Inner/drug effects , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/pathology , Mechanotransduction, Cellular , Regeneration/drug effects , Time Factors , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
4.
Hear Res ; 386: 107860, 2020 02.
Article in English | MEDLINE | ID: mdl-31869657

ABSTRACT

Purinergic receptors protect the cochlea during high-intensity stimulation by providing a parallel shunt pathway through non-sensory neighboring epithelial cells for cation absorption. So far, there is no direct functional evidence for the presence and type/subunit of purinergic receptors in the utricle of the vestibular labyrinth. The goal of the present study was to investigate which purinergic receptors are expressed and carry cation-absorption currents in the utricular transitional cells and macula. Purinergic agonists induced cation-absorption currents with a potency order of ATP > bzATP = αßmeATP â‰« ADP = UTP = UDP. ATP and bzATP are full agonists, whereas αßmeATP is a partial agonist. ATP-induced currents were partially inhibited by 100 µM suramin, 10 µM pyridoxal-phosphate-6-azo-(benzene-2,4-disulfonic acid (PPADS), or 5 µM 5-(3-bromophenyl)-1,3-dihydro-2H-benzofuro[3,2-e]-1, 4-diazepin-2-one (5-BDBD), and almost completely blocked by 100 µM Gd3+ or by a combination of 10 µM PPADS and 5 µM 5-BDBD. Expression of the P2RX2 and P2RX4 receptor was detected by immunocytochemistry in transitional cells and macular supporting cells. This is the first study to demonstrate that ATP induces cation currents carried by a combination of P2RX2 and P2RX4 in utricular transitional and macular epithelial cells, and supporting the hypothesis that purinergic receptors protect utricular hair cells during elevated stimulus intensity levels.


Subject(s)
Adenosine Triphosphate/metabolism , Labyrinth Supporting Cells/metabolism , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X4/metabolism , Saccule and Utricle/metabolism , Animals , Drug Partial Agonism , Labyrinth Supporting Cells/drug effects , Membrane Potentials , Mice, Inbred C57BL , Mice, Transgenic , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X2/drug effects , Receptors, Purinergic P2X4/drug effects , Saccule and Utricle/cytology , Saccule and Utricle/drug effects , Signal Transduction , Sulfate Transporters/genetics , Sulfate Transporters/metabolism
5.
Hear Res ; 371: 75-86, 2019 01.
Article in English | MEDLINE | ID: mdl-30504093

ABSTRACT

Ca2+ is an important intracellular messenger and regulator in both physiological and pathophysiological mechanisms in the hearing organ. Investigation of cellular Ca2+ homeostasis in the mature cochlea is hampered by the special anatomy and high vulnerability of the organ. A quick, straightforward and reliable Ca2+ imaging method with high spatial and temporal resolution in the mature organ of Corti is missing. Cell cultures or isolated cells do not preserve the special microenvironment and intercellular communication, while cochlear explants are excised from only a restricted portion of the organ of Corti and usually from neonatal pre-hearing murines. The hemicochlea, prepared from hearing mice allows tonotopic experimental approach on the radial perspective in the basal, middle and apical turns of the organ. We used the preparation recently for functional imaging in supporting cells of the organ of Corti after bulk loading of the Ca2+ indicator. However, bulk loading takes long time, is variable and non-selective, and causes the accumulation of the indicator in the extracellular space. In this study we show the improved labeling of supporting cells of the organ of Corti by targeted single-cell electroporation in mature mouse hemicochlea. Single-cell electroporation proved to be a reliable way of reducing the duration and variability of loading and allowed subcellular Ca2+ imaging by increasing the signal-to-noise ratio, while cell viability was retained during the experiments. We demonstrated the applicability of the method by measuring the effect of purinergic, TRPA1, TRPV1 and ACh receptor stimulation on intracellular Ca2+ concentration at the cellular and subcellular level. In agreement with previous results, ATP evoked reversible and repeatable Ca2+ transients in Deiters', Hensen's and Claudius' cells. TRPA1 and TRPV1 stimulation by AITC and capsaicin, respectively, failed to induce any Ca2+ response in the supporting cells, except in a single Hensen's cell in which AITC evoked transients with smaller amplitude. AITC also caused the displacement of the tissue. Carbachol, agonist of ACh receptors induced Ca2+ transients in about a third of Deiters' and fifth of Hensen's cells. Here we have presented a fast and cell-specific indicator loading method allowing subcellular functional Ca2+ imaging in supporting cells of the organ of Corti in the mature hemicochlea preparation, thus providing a straightforward tool for deciphering the poorly understood regulation of Ca2+ homeostasis in these cells.


Subject(s)
Calcium/metabolism , Cochlea/cytology , Cochlea/metabolism , Adenosine Triphosphate/metabolism , Aniline Compounds/administration & dosage , Animals , Calcium Chelating Agents/administration & dosage , Calcium Signaling/drug effects , Carbachol/administration & dosage , Cochlea/drug effects , Electroporation/methods , Fluoresceins/administration & dosage , Fluorescent Dyes/administration & dosage , Fura-2/administration & dosage , In Vitro Techniques , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Mice , Mice, Inbred BALB C , Organ of Corti/cytology , Organ of Corti/drug effects , Organ of Corti/metabolism , Receptors, Cholinergic/metabolism , Single-Cell Analysis/methods , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
6.
Hear Res ; 373: 10-22, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30578960

ABSTRACT

In mammals, the cochlear sensory epithelium becomes quiescent early during development. After the first postnatal week, there is no cell replacement or proliferation, and severe damage leads to permanent deafness. Supporting cells' trans-differentiation has been suggested as a way to regenerate cochlear hair cells after damage. However, they are also needed for proper functionality. Cdkn1b (p27Kip1) participates in the cochlear terminal mitosis state achieved during development. Its expression is maintained in adult supporting cells and its postnatal deletion has induced cochlear proliferation in vitro and in vivo. Therefore, its manipulation has been proposed as a feasible way to induce proliferation of supporting cells after birth. Nevertheless, the literature is scarce regarding feasible methods to directly decrease p27Kip1 in the clinical domain. The effects of p27Kip1 knockdown using viral vectors are not completely elucidated and no pharmacological approaches to decrease p27Kip1 in the cochlea have been tested in vivo before. This study explores the ability of p27Kip1 messenger knockdown and pharmacological transcriptional inhibition to induce proliferation of supporting cells in the P0 neonatal rat cochlea in vivo. Respectively, lentiviral vectors transducing shRNA against p27Kip1 were administered into the scala media or Alsterpaullone 2-Cyanoethyl into the round window niche. Cell markers and gene expression were assessed through immunostaining and qRT-PCR. Despite both methods significantly decreasing p27Kip1 expression in vivo, signs of toxicity in the organ of Corti were not found; however, relevant proliferation was not found either. Finally, cochlear damage was added to increase the response in vitro, achieving only a mild to moderate proliferation induction. We conclude that our approaches were not able to stimulate the recall of supporting cell proliferation despite significantly decreased p27Kip1 levels in vivo. Considering the evaluation of the cochlea at a very responsive stage, we propose that the level of isolated modification of p27Kip1 expression in living mammals achievable through these approaches is insufficient to induce proliferation of supporting cells. Future proliferation induction experiments in the cochlea should study other methods and genes.


Subject(s)
Cell Proliferation , Cochlea/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Labyrinth Supporting Cells/metabolism , Animals , Animals, Newborn , Benzazepines/pharmacology , Cell Proliferation/drug effects , Cochlea/drug effects , Cochlea/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Down-Regulation , Indoles/pharmacology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Signal Transduction , Tissue Culture Techniques
7.
Cell Stress Chaperones ; 23(6): 1311-1317, 2018 11.
Article in English | MEDLINE | ID: mdl-30196524

ABSTRACT

Heat shock protein 60 (HSP60) is a highly conserved chaperone molecule that plays important roles in mediating some physiological and pathological functions. However, researchers have not yet determined whether HSP60 is expressed in the mammalian cochlea. This study constitutes the first investigation of the expression of HSP60 in the postnatal rat cochlea. We also examined the expression of HSP60 in rats with drug-induced hearing loss. Auditory thresholds were assessed by monitoring the auditory brainstem response (ABR) prior to and after drug injection. Expression levels of the HSP60 gene (Hsp60) and HSP60 protein in the rat cochlea were detected by quantitative real-time polymerase chain reaction and Western blotting, respectively. The distribution of HSP60 in the rat cochlea was further examined by immunofluorescence staining. We have demonstrated that HSP60 was expressed in the postnatal rat cochlea in an age-dependent and cell-specific manner. In addition, after drug exposure, the average hearing threshold of rats in the experimental group was significantly higher than that in the control group, with increased HSP60 expression level in response to kanamycin and furosemide treatments. HSP60 expression was observed in the supporting cells (SCs) within the organ of Corti in both the uninjured and the injured cochlea, but it was undetectable in the mechanosensory hair cells (HCs) and spiral ganglion neurons. Therefore, our research suggests that HSP60 may play an important role in auditory function.


Subject(s)
Chaperonin 60/metabolism , Cochlea/metabolism , Hearing Loss/metabolism , Mitochondrial Proteins/metabolism , Animals , Auditory Threshold/drug effects , Chaperonin 60/genetics , Cochlea/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Furosemide/pharmacology , Hair Cells, Auditory/drug effects , Hearing Loss/chemically induced , Kanamycin/pharmacology , Labyrinth Supporting Cells/drug effects , Mitochondrial Proteins/genetics , Rats , Rats, Sprague-Dawley , Spiral Ganglion/drug effects , Transcriptional Activation , Up-Regulation
8.
Hear Res ; 364: 1-11, 2018 07.
Article in English | MEDLINE | ID: mdl-29754876

ABSTRACT

Permanent hearing loss is often a result of damage to cochlear hair cells, which mammals are unable to regenerate. Non-mammalian vertebrates such as birds replace damaged hair cells and restore hearing function, but mechanisms controlling regeneration are not understood. The secreted protein bone morphogenetic protein 4 (BMP4) regulates inner ear morphogenesis and hair cell development. To investigate mechanisms controlling hair cell regeneration in birds, we examined expression and function of BMP4 in the auditory epithelia (basilar papillae) of chickens of either sex after hair cell destruction by ototoxic antibiotics. In mature basilar papillae, BMP4 mRNA is highly expressed in hair cells, but not in hair cell progenitors (supporting cells). Supporting cells transcribe genes encoding receptors for BMP4 (BMPR1A, BMPR1B, and BMPR2) and effectors of BMP4 signaling (ID transcription factors). Following hair cell destruction, BMP4 transcripts are lost from the sensory epithelium. Using organotypic cultures, we demonstrate that treatments with BMP4 during hair cell destruction prevent supporting cells from upregulating expression of the pro-hair cell transcription factor ATOH1, entering the cell cycle, and fully transdifferentiating into hair cells, but they do not induce cell death. By contrast, noggin, a BMP4 inhibitor, increases numbers of regenerated hair cells. These findings demonstrate that BMP4 antagonizes hair cell regeneration in the chicken basilar papilla, at least in part by preventing accumulation of ATOH1 in hair cell precursors.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Proliferation/drug effects , Hair Cells, Auditory/drug effects , Labyrinth Supporting Cells/drug effects , Regeneration/drug effects , Animals , Anti-Bacterial Agents/toxicity , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Carrier Proteins/pharmacology , Cell Communication/drug effects , Cell Transdifferentiation , Chickens , Female , Gentamicins/toxicity , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/pathology , Male , Signal Transduction/drug effects , Tissue Culture Techniques
9.
Hear Res ; 364: 129-141, 2018 07.
Article in English | MEDLINE | ID: mdl-29563067

ABSTRACT

Paraquat (PQ), one of the most widely used herbicides, is extremely dangerous because it generates the highly toxic superoxide radical. When paraquat was applied to cochlear organotypic cultures, it not only damaged the outer hair cells (OHCs) and inner hair cells (IHCs), but also caused dislocation of the hair cell rows. We hypothesized that the dislocation arose from damage to the support cells (SCs) that anchors hair cells within the epithelium. To test this hypothesis, rat postnatal cochlear cultures were treated with PQ. Shortly after PQ treatment, the rows of OHCs separated from one another and migrated radially away from IHCs suggesting loss of cell-cell adhesion that hold the hair cells in proper alignment. Hair cells dislocation was associated with extensive loss of SCs in the organ of Corti, loss of tympanic border cells (TBCs) beneath the basilar membrane, the early appearance of superoxide staining and caspase-8 labeling in SCs below the OHCs and disintegration of E-cadherin and ß-catenin in the organ of Corti. Damage to the TBCs and SCs occurred prior to loss of OHC or IHC loss suggesting a form of detachment-induced apoptosis referred to as anoikis.


Subject(s)
Anoikis/drug effects , Cochlea/drug effects , Hair Cells, Auditory/drug effects , Herbicides/toxicity , Labyrinth Supporting Cells/drug effects , Paraquat/toxicity , Animals , Animals, Newborn , Cadherins/metabolism , Caspase 8/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cochlea/metabolism , Cochlea/pathology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Superoxides/metabolism , Time Factors , Tissue Culture Techniques , beta Catenin/metabolism
10.
Int J Pediatr Otorhinolaryngol ; 92: 61-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28012535

ABSTRACT

OBJECTIVE: Cisplatin is commonly used to treat solid tumors. However, permanent hearing loss is a major side effect of cisplatin chemotherapy and often results in dose reduction of the cisplatin chemotherapy. Peanut sprouts show cytoprotective properties owing to their antioxidant activities. This study was designed to investigate the effect of peanut sprout extract (PSE) on cisplatin-induced ototoxicity in an auditory cell line, HEI-OC1 cells. METHODS: Cells were exposed to cisplatin for 24 h, with or without pre-treatment with PSE, cell viability was examined using the MTT assay. Apoptotic cells were identified by double staining with Hoechst 33258 and propidium iodide. Western blot analysis was performed to examine apoptotic proteins including C-PARP and C-caspase, anti-apoptotic protein Bcl-2, and Nrf2 redox system activation. Mitochondrial reactive oxygen species (ROS) were investigated to examine whether PSE could scavenge cisplatin-induced ROS. Real-time PCR analyses were performed to investigate the mRNA levels of antioxidant enzymes including NQO1, HO-1, GPx2, Gclc, and catalase. RESULTS: The cisplatin-treated group showed reduced cell viability, increased apoptotic properties and markers, and increased ROS levels. PSE pre-treatment before cisplatin exposure significantly increased cell viability and reduced apoptotic properties and ROS production. These effects resulted from the up-regulation of antioxidant genes, including NQO1, HO-1, GPx2, Gclc, and catalase through Akt phosphorylation and Nrf2 activation. CONCLUSION: Our results demonstrate that PSE protects from cisplatin-induced cytotoxicity by activating the antioxidant effects via the Akt/Nrf-2 pathway in this auditory cell line, and indicate that PSE may provide novel treatment to prevent cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Arachis , Cisplatin/toxicity , Hair Cells, Auditory/drug effects , Labyrinth Supporting Cells/drug effects , Plant Extracts/pharmacology , Seedlings , Animals , Blotting, Western , Caspases/drug effects , Caspases/metabolism , Catalase/drug effects , Catalase/genetics , Cell Line , Cell Survival/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/genetics , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , In Vitro Techniques , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction/drug effects , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
11.
Free Radic Biol Med ; 101: 211-225, 2016 12.
Article in English | MEDLINE | ID: mdl-27769922

ABSTRACT

Experimental and human investigations have raised the level of concern about the potential ototoxicity of organic solvents and their interaction with noise. The main objective of this study was to characterize the effects of the combined noise and styrene exposure on hearing focusing on the mechanism of damage on the sensorineural cells and supporting cells of the organ of Corti and neurons of the ganglion of Corti. The impact of single and combined exposures on hearing was evaluated by auditory functional testing and histological analyses of cochlear specimens. The mechanism of damage was studied by analyzing superoxide anion and lipid peroxidation expression and by computational analyses of immunofluorescence data to evaluate and compare the oxidative stress pattern in outer hair cells versus the supporting epithelial cells of the organ of Corti. The oxidative stress hypothesis was further analyzed by evaluating the protective effect of a Coenzyme Q10 analogue, the water soluble Qter, molecule known to have protective antioxidant properties against noise induced hearing loss and by the analysis of the expression of the endogenous defense enzymes. This study provides evidence of a reciprocal noise-styrene synergism based on a redox imbalance mechanism affecting, although with a different intensity of damage, the outer hair cell (OHC) sensory epithelium. Moreover, these two damaging agents address preferentially different cochlear targets: noise mainly the sensory epithelium, styrene the supporting epithelial cells. Namely, the increase pattern of lipid peroxidation in the organ of Corti matched the cell damage distribution, involving predominantly OHC layer in noise exposed cochleae and both OHC and Deiters' cell layers in the styrene or combined exposed cochleae. The antioxidant treatment reduced the lipid peroxidation increase, potentiated the endogenous antioxidant defense system at OHC level in both exposures but it failed to ameliorate the oxidative imbalance and cell death of Deiters' cells in the styrene and combined exposures. Current antioxidant therapeutic approaches to preventing sensory loss focus on hair cells alone. It remains to be seen whether targeting supporting cells, in addition to hair cells, might be an effective approach to protecting exposed subjects.


Subject(s)
Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Outer/drug effects , Hearing Loss, Noise-Induced/metabolism , Labyrinth Supporting Cells/drug effects , Noise/adverse effects , Styrene/toxicity , Animals , Antioxidants/pharmacology , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Noise-Induced/prevention & control , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/pathology , Lipid Peroxidation/drug effects , Male , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Wistar , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
12.
Hear Res ; 332: 17-28, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26688175

ABSTRACT

Hair cells in posthatch chickens regenerate spontaneously through mitosis or the transdifferentiation of supporting cells in response to antibiotic injury. However, how embryonic chicken cochleae respond to antibiotic treatment remains unknown. This study is the first to indicate that unlike hair cells in posthatch chickens, the auditory epithelium was free from antibiotic injury (25-250 mg gentamicin/kg) in embryonic chickens, although FITC-conjugated gentamicin actually reached embryonic hair cells. Next, we examined and counted the cells and performed labeling for BrdU, Sox2, Atoh1/Math1, PV or p27(kip1) (triple or double labeling) in the injured cochlea ducts after gentamicin treatment at 2 h (h), 15 h, 24 h, 2 days (d), 3 d and 7 d after BrdU treatment in posthatch chickens. Our results indicated that following gentamicin administration, proliferating cells (BrdU+) were labeled for Atoh1/Math1 in the damaged areas 3d after gentamicin administration, whereas hair cells (PV+) renewed through mitosis (BrdU+) or direct transdifferentiation (BrdU-) were evident only after 5 d of gentamicin administration. In addition, Sox2 expression was up-regulated in triggered supporting cells at an early stage of regeneration, but stopped at the advent of mature hair cells. Our study also indicated that p27(kip1) was expressed in both hair cells and supporting cells but was down-regulated in a subgroup of the supporting cells that gave rise to hair cells. These data and the obtained dynamic changes of the cells labeled for BrdU, Sox2, Atoh1/Math1, PV or p27(kip1) are useful for understanding supporting cell behaviors and their fate specification during hair cell regeneration.


Subject(s)
Anti-Bacterial Agents/toxicity , Cell Lineage/drug effects , Cell Transdifferentiation/drug effects , Cochlear Duct/drug effects , Gentamicins/toxicity , Hair Cells, Auditory/drug effects , Labyrinth Supporting Cells/drug effects , Regeneration/drug effects , Age Factors , Animals , Animals, Newborn , Biomarkers/metabolism , Chick Embryo , Chickens , Cochlear Duct/embryology , Cochlear Duct/metabolism , Cochlear Duct/pathology , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/pathology , Mitosis/drug effects , Time Factors
13.
Cell Death Dis ; 6: e1605, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25611380

ABSTRACT

Reactive oxygen species (ROS) accumulation are involved in noise- and ototoxic drug-induced hair cell loss, which is the major cause of hearing loss. Bmi1 is a member of the Polycomb protein family and has been reported to regulate mitochondrial function and ROS level in thymocytes and neurons. In this study, we reported the expression of Bmi1 in mouse cochlea and investigated the role of Bmi1 in hair cell survival. Bmi1 expressed in hair cells and supporting cells in mouse cochlea. Bmi1(-/-) mice displayed severe hearing loss and patched outer hair cell loss from postnatal day 22. Ototoxic drug-induced hair cells loss dramatically increased in Bmi1(-/-) mice compared with that in wild-type controls both in vivo and in vitro, indicating Bmi1(-/-) hair cells were significantly more sensitive to ototoxic drug-induced damage. Cleaved caspase-3 and TUNEL staining demonstrated that apoptosis was involved in the increased hair cell loss of Bmi1(-/-) mice. Aminophenyl fluorescein and MitoSOX Red staining showed the level of free radicals and mitochondrial ROS increased in Bmi1(-/-) hair cells due to the aggravated disequilibrium of antioxidant-prooxidant balance. Furthermore, the antioxidant N-acetylcysteine rescued Bmi1(-/-) hair cells from neomycin injury both in vitro and in vivo, suggesting that ROS accumulation was mainly responsible for the increased aminoglycosides sensitivity in Bmi1(-/-) hair cells. Our findings demonstrate that Bmi1 has an important role in hair cell survival by controlling redox balance and ROS level, thus suggesting that Bmi1 may work as a new therapeutic target for the prevention of hair cell death.


Subject(s)
Hair Cells, Auditory/pathology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , DNA Damage , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hearing Loss/chemically induced , Hearing Loss/pathology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Neomycin/adverse effects , Oxidants/metabolism , Oxidation-Reduction/drug effects , Polycomb Repressive Complex 1/deficiency , Proto-Oncogene Proteins/deficiency , Reactive Oxygen Species/metabolism
14.
Hear Res ; 289(1-2): 74-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22543087

ABSTRACT

Loss of hair cells in humans leads to irreversible hearing deficits, since auditory hair cells are not replaced. In contrast, hair cells are regenerated in the auditory epithelium of mature birds after damage by non-sensory supporting cells that transdifferentiate into hair cells by mitotic and/or non-mitotic mechanisms. Factors controlling these processes are poorly understood. The basic helix-loop-helix transcription factor ATOH1 is both necessary and sufficient for developmental hair cell differentiation, but it is unclear if it plays the same role in the mitotic and non-mitotic pathways in hair cell regeneration. We examined Atoh1 expression and function during hair cell regeneration in chickens. Atoh1 transcripts were increased in many supporting cells in the damaged auditory epithelium shortly after ototoxin administration and later became restricted to differentiating hair cells. Fate-mapping in vitro using an Atoh1 enhancer reporter demonstrated that only 56% of the supporting cells that spontaneously upregulate Atoh1 enhancer activity after damage acquired the hair cell fate. Inhibition of notch signaling using a gamma secretase antagonist stimulated an increase in Atoh1 reporter activity and induced a higher proportion of supporting cells with Atoh1 activity (73%) to differentiate as hair cells. Forced overexpression of Atoh1 in supporting cells triggered 66% of them to acquire the hair cell fate and nearly tripled their likelihood of cell cycle entry. These findings demonstrate that Atoh1 is broadly upregulated in supporting cells after damage, but a substantial proportion of supporting cells with Atoh1 activation fails to acquire hair cell features, in part due to gamma secretase-dependent activities.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/metabolism , Regeneration , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle , Cell Transdifferentiation , Chickens , Dipeptides/pharmacology , Electroporation , Enhancer Elements, Genetic , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Gene Transfer Techniques , Genes, Reporter , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , Organ Culture Techniques , RNA, Messenger/metabolism , Receptors, Notch/metabolism , Regeneration/drug effects , Streptomycin/toxicity , Time Factors , Transcriptional Activation
15.
Hear Res ; 289(1-2): 27-39, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22575790

ABSTRACT

The avian basilar papilla (BP) is a likely homolog of the auditory sensory epithelium of the mammalian cochlea, the organ of Corti. During mammalian development Fibroblast growth factor receptor-3 (Fgfr3) is known to regulate the differentiation of auditory mechanosensory hair cells (HCs) and supporting cells (SCs), both of which are required for sound detection. Fgfr3 is expressed in developing progenitor cells (PCs) and SCs of both the BP and the organ of Corti; however its role in BP development is unknown. Here we utilized an in vitro whole organ embryonic culture system to examine the role of Fgf signaling in the developing avian cochlea. SU5402 (an antagonist of Fgf signaling) was applied to developing BP cultures at different stages to assay the role of Fgf signaling during HC formation. Similar to the observed effects of inhibition of Fgfr3 in the mammalian cochlea, Fgfr inhibition in the developing BP increased the number of HCs that formed. This increase was not associated with increased proliferation, suggesting that inhibition of the Fgf pathway leads to the direct conversion of PCs or supporting cells into HCs, a process known as transdifferentiation. This also implies that Fgf signaling is required to prevent the conversion of PCs and SCs into HCs. The ability of Fgf signaling to inhibit transdifferentiation suggests that its down-regulation may be essential for the initial steps of HC formation, as well as for the maintenance of SC phenotypes.


Subject(s)
Cell Transdifferentiation , Fibroblast Growth Factors/metabolism , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/metabolism , Organ of Corti/metabolism , Signal Transduction , Animals , Cell Proliferation , Cell Transdifferentiation/drug effects , Chick Embryo , Hair Cells, Auditory/drug effects , Labyrinth Supporting Cells/drug effects , Organ Culture Techniques , Organ of Corti/drug effects , Organ of Corti/embryology , Phenotype , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects
16.
PLoS One ; 6(8): e23861, 2011.
Article in English | MEDLINE | ID: mdl-21909368

ABSTRACT

When inner ear hair cells die, humans and other mammals experience permanent hearing and balance deficits, but non-mammalian vertebrates quickly recover these senses after epithelial supporting cells give rise to replacement hair cells. A postnatal decline in cellular plasticity appears to limit regeneration in mammalian balance organs, where declining proliferation responses are correlated with decreased spreading of supporting cells on artificial and native substrates. By culturing balance epithelia on substrates that differed in flexibility, we assessed spreading effects independent of age, showing a strong correlation between shape change and supporting cell proliferation. Then we made excision wounds in utricles cultured from young and old chickens and mice and compared quantified levels of spreading and proliferation. In utricles from young mice, and both young and old chickens, wounds re-epithelialized in <24 hours, while those in utricles from mature mice took three times longer. More cells changed shape in the fastest healing wounds, which accounted for some differences in the levels of proliferation, but inter-species and age-related differences in shape-sensitive restriction points, i.e., the cellular thresholds for shape changes that promote S-phase, were evident and may be particularly influential in the responses to hair cell losses in vivo.


Subject(s)
Chickens/anatomy & histology , Ear/pathology , Regeneration/physiology , Acoustic Maculae/drug effects , Acoustic Maculae/pathology , Acoustic Maculae/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Collagen/pharmacology , Drug Combinations , Ear/physiology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , Laminin/pharmacology , Mice , Proteoglycans/pharmacology , Regeneration/drug effects , S Phase/drug effects , Wound Healing/drug effects
17.
Hear Res ; 282(1-2): 236-42, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21810458

ABSTRACT

OBJECTIVE: Lateral line system of the zebrafish is a useful model for study of hair cell toxicity and regeneration. We found that low molecular weight fucoidan (LMWF) stimulated the regeneration of mechanosensory hair cells after neomycin-induced cell death in zebrafish lateral line. The aims of this study were to quantify the regenerative effects of LMWF and determine their relationship to the Notch and FGF signaling pathways. METHODS: Wild-type zebrafish and three different transgenic zebrafish lines (Pou4f3::GFP, scm1::GFP, and ET20::GFP) were used. At 4.5-6 days post-fertilization, lateral line hair cells of larvae were eliminated using neomycin (500 µM). Larvae were then treated with LMWF. Neuromasts were observed using confocal microscopy. Stereocilia morphology was observed using scanning electron microscopy, and the location and status of regeneration was assessed using 5-bromo-2-deoxyuridine (BrdU) incorporation. RESULTS: Hair cells damaged by neomycin treatment regenerated faster in wild-type and Pou4f3::GFP larvae treated with LMWF (50 µg/ml) than in untreated controls. LMWF also enhanced the regeneration of supporting cells in scm1::GFP and ET20::GFP larvae. Increased numbers of BrdU-labeled cells were found after LMWF treatment in neuromast regions corresponding to internal and peripheral supporting cells. The effect of LMWF was mimicked by the Notch signaling inhibitor N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butyl ester (DAPT), but the effects of LMWF and DAPT were not additive. CONCLUSION: LMWF enhances the regeneration of hair cells damaged by neomycin. The mechanism may involve the Notch signaling pathway. LMWF shows promise as a therapeutic agent for hearing and balance disorders.


Subject(s)
Aminoglycosides/toxicity , Cell Proliferation/drug effects , Hair Cells, Auditory/drug effects , Lateral Line System/drug effects , Mechanotransduction, Cellular/drug effects , Neomycin/toxicity , Polysaccharides/pharmacology , Regeneration/drug effects , Animals , Animals, Genetically Modified , Cell Death/drug effects , Dose-Response Relationship, Drug , Fibroblast Growth Factors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , Lateral Line System/metabolism , Lateral Line System/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Scanning , Receptors, Notch/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology
18.
J Neurosci ; 30(9): 3473-81, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20203207

ABSTRACT

Cisplatin is a chemotherapeutic agent that is widely used in the treatment of solid tumors. Ototoxicity is a common side effect of cisplatin therapy and often leads to permanent hearing loss. The sensory organs of the avian ear are able to regenerate hair cells after aminoglycoside ototoxicity. This regenerative response is mediated by supporting cells, which serve as precursors to replacement hair cells. Given the antimitotic properties of cisplatin, we examined whether the avian ear was also capable of regeneration after cisplatin ototoxicity. Using cell and organ cultures of the chick cochlea and utricle, we found that cisplatin treatment caused apoptosis of both auditory and vestibular hair cells. Hair cell death in the cochlea occurred in a unique pattern, progressing from the low-frequency (distal) region toward the high-frequency (proximal) region. We also found that cisplatin caused a dose-dependent reduction in the proliferation of cultured supporting cells as well as increased apoptosis in those cells. As a result, we observed no recovery of hair cells after ototoxic injury caused by cisplatin. Finally, we explored the potential for nonmitotic hair cell recovery via activation of Notch pathway signaling. Treatment with the gamma-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester failed to promote the direct transdifferentiation of supporting cells into hair cells in cisplatin-treated utricles. Taken together, our data show that cisplatin treatment causes maintained changes to inner ear supporting cells and severely impairs the ability of the avian ear to regenerate either via proliferation or by direct transdifferentiation.


Subject(s)
Cisplatin/toxicity , Ear, Inner/drug effects , Nerve Degeneration/chemically induced , Nerve Regeneration/drug effects , Neurotoxins/toxicity , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Chick Embryo , Deafness/chemically induced , Deafness/pathology , Deafness/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Ear, Inner/pathology , Ear, Inner/physiopathology , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Hair Cells, Vestibular/drug effects , Hair Cells, Vestibular/pathology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Regeneration/physiology , Organ Culture Techniques , Pitch Perception/drug effects , Pitch Perception/physiology , Receptors, Notch/drug effects , Receptors, Notch/metabolism
19.
Neuroscience ; 166(4): 1185-93, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20060033

ABSTRACT

Adenovirus vectors (AdVs) are efficient tools for gene therapy in many tissues. Several studies have demonstrated successful transgene transduction with AdVs in the inner ear of rodents [Kawamoto K, Ishimoto SI, Minoda R, Brough DE, Raphael Y (2003) J Neurosci 23:4395-4400]. However, toxicity of AdVs [Morral N, O'Neal WK, Rice K, Leland MM, Piedra PA, Aguilar-Cordova E, Carey KD, Beaudet AL, Langston C (2002) Hum Gene Ther 13:143-154.] or lack of tropism to important cell types such as hair cells [Shou J, Zheng JL, Gao WQ (2003) Mol Cell Neurosci 23:169-179] appears to limit their experimental and potential clinical utility. Histone deacetylase inhibitors (HDIs) are known to enhance AdV-mediated transgene expression in various organs [Dion LD, Goldsmith KT, Tang DC, Engler JA, Yoshida M, Garver RI Jr (1997) Virology 231:201-209], but their effects in the inner ear have not been documented. We investigated the ability of one HDI, trichostatin A (TSA), to enhance AdV-mediated transgene expression in inner ear tissue. We cultured neonatal rat macular and cochlear explants, and transduced them with an AdV encoding green fluorescent protein (Ad-GFP) under the control of a constitutive promoter for 24 h. In the absence of TSA, GFP expression was limited, and very few hair cells were transduced. TSA did not enhance transduction when applied at the onset of Ad-GFP transduction. However, administration of TSA during or just after Ad-GFP application increased GFP expression in supporting cells approximately fourfold. Moreover, vestibular hair cell transduction was enhanced approximately sixfold, and that of inner hair cells by more than 17-fold. These results suggest that TSA increases AdV-mediated transgene expression in the inner ear, including the successful transduction of hair cells. HDIs, some of which are currently under clinical trials (Sandor et al., 2002), could be useful tools in overcoming current limitations of gene therapy in the inner ear using Ad-GFP.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Hair Cells, Auditory/metabolism , Hearing Loss, Sensorineural/therapy , Histone Deacetylase Inhibitors/pharmacology , Transduction, Genetic/methods , Adenoviridae/genetics , Animals , Animals, Newborn , Cells, Cultured , Drug Delivery Systems/methods , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hair Cells, Auditory/drug effects , Hair Cells, Vestibular/drug effects , Hair Cells, Vestibular/metabolism , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/physiopathology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Nerve Growth Factors/pharmacology , Nerve Growth Factors/therapeutic use , Organ Culture Techniques , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Transgenes/genetics
20.
Neurochem Int ; 56(3): 487-94, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20026213

ABSTRACT

We previously reported that treatment of the rat cochlea with a mitochondrial toxin, 3-nitropropionic acid (3-NP), causes temporary to permanent hearing loss depending on the amount of the drug. Furthermore, apoptosis of cochlear lateral wall fibrocytes, which are important for maintaining the endolymph, is a predominant pathological feature in this animal model. 3-NP is known to induce oxidative stress as well as neuronal apoptosis. C/EBP homologous protein gene (chop) is one of the marker genes induced during endoplasmic reticulum (ER) stress, and is also considered to be involved in apoptosis. To elucidate the molecular mechanism of cochlear fibrocyte apoptosis induced by 3-NP, we studied spatiotemporal expression of C/EBP homologous protein (CHOP) and other signaling molecules related to ER stress as well as the appearance of apoptotic cells in the cochlear lateral wall after 3-NP treatment. Quantitative real-time PCR revealed that chop and activating transcription factor 4 gene (atf-4) showed marked increase within 6h, whereas expression of other ER stress-responsive genes such as grp78 and grp94 did not change. Immunohistochemistry showed that 3-NP treatment caused up-regulation of CHOP, especially in type II and type IV fibrocytes, followed by the appearance of terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling (TUNEL)-positive apoptotic cells in the same confined area. Thus, apoptosis of lateral wall fibrocytes induced by 3-NP is likely to be mediated by induction of CHOP. These results contribute clarification of pathological mechanism of cochlear fibrocytes and may lead to development of novel therapeutic strategy for hearing loss.


Subject(s)
Hearing Loss, Sensorineural/metabolism , Labyrinth Supporting Cells/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Organ of Corti/metabolism , Transcription Factor CHOP/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Acute Disease , Animals , Apoptosis/drug effects , Apoptosis/physiology , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/physiopathology , In Situ Nick-End Labeling , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/pathology , Male , Mitochondria/drug effects , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/physiopathology , Neurotoxins/toxicity , Nitro Compounds/toxicity , Organ of Corti/drug effects , Organ of Corti/physiopathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Propionates/toxicity , Rats , Rats, Sprague-Dawley , Transcription Factor CHOP/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...