Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Biotech Histochem ; 92(7): 536-544, 2017.
Article in English | MEDLINE | ID: mdl-28925723

ABSTRACT

Age-related hearing loss (ARHL) is characterized by an age-dependent decline of auditory function characterized by with loss of sensory hair cells, spiral ganglion neurons, and stria vascularis (SV) cells in the cochlea of the inner ear. Aging and age-related diseases result from accumulated oxidative damage caused by reactive oxygen species (ROS) generated by mitochondria. The isocitrate dehydrogenase (IDH) family includes three enzymes in human cells: IDH1, IDH2, and IDH3. Although all three enzymes catalyze the same enzymatic reaction, that is, oxidative decarboxylation of isocitrate to produce α-ketoglutarate, each IDH enzyme has unique features. We identified and characterized IDH expression in the cochlea and vestibule of the murine inner ear. We examined the mRNA expression levels of Idh family members in the cochlea and vestibule using reverse transcription-PCR (RT-PCR) and detected expression of IDH family members in both tissues. We also used immunohistochemistry to localize IDH family members within the cochlea and vestibule of the adult mouse inner ear. IDH1 was detected throughout the cochlea. IDH2 was expressed specifically in the hair cells, spiral ganglion, and stria vascularis. IDH3α was found in the cell bodies of neurons of the spiral ganglion, the stria vascularis, and in types II, IV, and V cells of the spiral ligament in a pattern that resembled the location of the Na+, K+-ATPase ion channel. We postulate that the IDH family participates in transporting K+ ions in the cochlea. In the vestibule, all IDH family members were detected in both hair cells and the vestibular ganglion. We hypothesize that IDH1, IDH2, and IDH3 function to protect proteins in the inner ear from oxidative stress during K+ recycling.


Subject(s)
Ear, Inner , Isocitrate Dehydrogenase/metabolism , Animals , Ear, Inner/enzymology , Ear, Inner/metabolism , Female , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction
2.
Eur Arch Otorhinolaryngol ; 273(10): 3053-61, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26831119

ABSTRACT

To investigate the effect of transtympanic betamethasone administration on hearing function with histologic correlation, rats were divided into three transtympanic treatment groups: isotonic saline (group I, n = 10), gentamicin (group II, n = 10) and betamethasone (group III, n = 10). Distortion product otoacoustic emission thresholds were compared on day 10. Also histological effects on cellular apoptosis in both the inner and outer hair cells in organ of Corti and spiral ganglion neurons were evaluated. Distortion product otoacoustic emission thresholds were comparable (p > 0.05) between group I and group III in all measurements. Distortion product otoacoustic emission thresholds of group II were significantly elevated in all measurements when compared with group I (p < 0.05) and group III (p < 0.05). In the Terminal deoxynucleotidyl transferase dUTP Nick End Labelling (TUNEL), Caspase-3, Caspase-8 and Caspase-9 staining method the amount of apoptotic cells in group II were significantly elevated in all measurements compared with group I (p < 0.05). In the TUNEL staining method the amount of apoptotic cells in Group III were significantly elevated compared with group I in both the organ of Corti and spiral ganglion neurons (p < 0.05). The overall histological results revealed that the severity of cellular apoptosis caused by betamethasone was somewhere between isotonic saline and gentamicin. Transtympanic betamethasone does not affect inner ear function as measured by distortion product otoacoustic emission responses, but some increase in cellular apoptosis in the organ of Corti and spiral ganglion neurons was observed. These findings suggest that transtympanic betamethasone may have mild ototoxic effects. Further studies are needed to obtain precise results for transtympanic application of betamethasone.


Subject(s)
Betamethasone/administration & dosage , Ear, Inner/drug effects , Glucocorticoids/administration & dosage , Otoacoustic Emissions, Spontaneous/drug effects , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Cochlea/drug effects , Cochlea/enzymology , Ear, Inner/enzymology , Ear, Inner/physiology , Gentamicins/pharmacology , Hair Cells, Auditory, Outer/drug effects , In Situ Nick-End Labeling , Male , Organ of Corti/drug effects , Otoacoustic Emissions, Spontaneous/physiology , Rats , Rats, Wistar , Sensory Thresholds/drug effects , Sensory Thresholds/physiology , Spiral Ganglion/drug effects , Tympanic Membrane
3.
J Neurosci ; 36(4): 1347-61, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26818521

ABSTRACT

Mechanosensory hair cells (HCs) residing in the inner ear are critical for hearing and balance. Precise coordination of proliferation, sensory specification, and differentiation during development is essential to ensure the correct patterning of HCs in the cochlear and vestibular epithelium. Recent studies have revealed that FGF20 signaling is vital for proper HC differentiation. However, the mechanisms by which FGF20 signaling promotes HC differentiation remain unknown. Here, we show that mitogen-activated protein 3 kinase 4 (MEKK4) expression is highly regulated during inner ear development and is critical to normal cytoarchitecture and function. Mice homozygous for a kinase-inactive MEKK4 mutation exhibit significant hearing loss. Lack of MEKK4 activity in vivo also leads to a significant reduction in the number of cochlear and vestibular HCs, suggesting that MEKK4 activity is essential for overall development of HCs within the inner ear. Furthermore, we show that loss of FGF20 signaling in vivo inhibits MEKK4 activity, whereas gain of Fgf20 function stimulates MEKK4 expression, suggesting that Fgf20 modulates MEKK4 activity to regulate cellular differentiation. Finally, we demonstrate, for the first time, that MEKK4 acts as a critical node to integrate FGF20-FGFR1 signaling responses to specifically influence HC development and that FGFR1 signaling through activation of MEKK4 is necessary for outer hair cell differentiation. Collectively, this study provides compelling evidence of an essential role for MEKK4 in inner ear morphogenesis and identifies the requirement of MEKK4 expression in regulating the specific response of FGFR1 during HC development and FGF20/FGFR1 signaling activated MEKK4 for normal sensory cell differentiation. SIGNIFICANCE STATEMENT: Sensory hair cells (HCs) are the mechanoreceptors within the inner ear responsible for our sense of hearing. HCs are formed before birth, and mammals lack the ability to restore the sensory deficits associated with their loss. In this study, we show, for the first time, that MEKK4 signaling is essential for the development of normal cytoarchitecture and hearing function as MEKK4 signaling-deficient mice exhibit a significant reduction of HCs and a hearing loss. We also identify MEKK4 as a critical hub kinase for FGF20-FGFR1 signaling to induce HC differentiation in the mammalian cochlea. These results reveal a new paradigm in the regulation of HC differentiation and provide significant new insights into the mechanism of Fgf signaling governing HC formation.


Subject(s)
Ear, Inner , Gene Expression Regulation, Developmental/physiology , MAP Kinase Kinase Kinase 4/metabolism , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Ear, Inner/cytology , Ear, Inner/enzymology , Ear, Inner/growth & development , Embryo, Mammalian , Evoked Potentials, Auditory, Brain Stem/genetics , Female , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory, Inner/physiology , MAP Kinase Kinase Kinase 4/genetics , Male , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Pregnancy , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics , Spiral Ganglion/cytology , Tubulin/metabolism
4.
Acta Otolaryngol ; 134(4): 331-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24460154

ABSTRACT

CONCLUSION: It is suggested that SIRT1 and 3, and probably SIRT4 and 5, play an important role in the neuroprotection of the inner ear. SIRT2 may be related to neuroprotection and myelin sheath formation, while SIRT6 seems to have a significant role in maintaining the energy balance by metabolic regulation. OBJECTIVE: To analyze the expression of sirtuins (SIRT1-7) in the normal mouse inner ear. METHODS: CBA/J mice were used for this study. The localization of SIRT1-7 in the inner ear, i.e. cochlea, vestibular end organs, and endolymphatic sac, was investigated using real-time PCR and immunohistochemistry. RESULTS: We found high levels of mRNA of all seven sirtuins in the inner ear. In the immunohistochemical study, SIRT1-7 were abundant in many inner ear structures, i.e. stria vascularis, inner and outer hair cells, spiral ganglion cells, vestibular sensory and ganglion cells, vestibular dark and transitional cells, and the endolymphatic sac.


Subject(s)
Ear, Inner/enzymology , Gene Expression Regulation , RNA, Messenger/genetics , Sirtuins/genetics , Animals , Ear, Inner/cytology , Immunohistochemistry , Mice , Mice, Inbred CBA , Real-Time Polymerase Chain Reaction , Sirtuins/biosynthesis , Tomography, Optical Coherence
5.
J Control Release ; 166(3): 268-76, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23313113

ABSTRACT

OBJECTIVE: We have recently developed a novel inner ear drug delivery system using chitosan glycerophosphate (CGP) hydrogel loaded with drugs commonly used for treatment of inner ear diseases, significantly improving the drugs' sustained delivery. The goal of this study is to evaluate the effectiveness of chitosanase as a "switch off" mechanism for this drug delivery system when side effects and potential ototoxicities appear during treatment. To evaluate this effect, we tested gentamicin (GENT) in the inner ear following CGP delivery with/without regulation. METHODS: Purified chitosanase was obtained and used for regulating the CGP delivery system. In vitro studies were performed to evaluate the effect of the interaction between chitosanase and CGP-hydrogel loaded with GENT or Texas Red-labeled GENT (GTTR). In vivo studies were performed using our mouse model to investigate the regulatory effect of chitosanase application on the delivery of GENT to the inner ear. To assess the potential drug rerouting regulatory effect of chitosanase the GTTR fluorescence intensity was evaluated at the round window niche (RWN) and the Eustachian tube (ET). To further characterize this regulatory effect, GENT concentration in the perilymph of the inner ear was analyzed by chromatographic tandem mass spectrometry (LC-MS/MS), and the uptake in the inner ear cells was measured using fluorescence microscopy following CGP delivery with/without chitosanase application. RESULTS: The chitosanase effectively digested the CGP-hydrogel, quickly releasing GENT and GTTR from the system in vitro. When reacted with GENT alone chitosanase did not produce any reducing sugars and did not affect GENT's antimicrobial activity. In vivo GTTR was effectively rerouted from the RWN to the ET, limiting its uptake in inner ear hair cells. Concurrent with these findings, GENT concentration in the inner ear perilymph was significantly decreased after chitosanase application. CONCLUSION: Our study findings suggest that, for the first time, sustained and controlled inner ear drug delivery can be successfully regulated enhancing its translation potential for clinical application. The use of chitosanase to digest the CGP-hydrogel results in the rerouting of the loaded drug away from the RWN, effectively downregulating its delivery to the inner ear. This important modification to our drug delivery system has the ability to deliver therapy to the inner ear until desired effect is achieved and to stop this process when side effects or treatment-related ototoxicities start to occur, providing a novel and salient approach for safe and effective delivery to the inner ear.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Drug Delivery Systems/methods , Ear, Inner/metabolism , Gentamicins/administration & dosage , Animals , Anti-Bacterial Agents/pharmacokinetics , Blotting, Western , Chitosan/analogs & derivatives , Chitosan/chemistry , Delayed-Action Preparations , Drug Carriers/chemistry , Ear, Inner/enzymology , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Female , Gentamicins/pharmacokinetics , Glycerophosphates/chemistry , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Hydrogels , Labyrinth Diseases/drug therapy , Male , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Models, Biological , Plasmids , Staphylococcus aureus/drug effects , Tissue Distribution
6.
Dis Model Mech ; 6(2): 434-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23065636

ABSTRACT

Mutations in the ATP6V0A4 gene lead to autosomal recessive distal renal tubular acidosis in patients, who often show sensorineural hearing impairment. A first Atp6v0a4 knockout mouse model that recapitulates the loss of H(+)-ATPase function seen in humans has been generated and recently reported (Norgett et al., 2012). Here, we present the first detailed analysis of the structure and function of the auditory system in Atp6v0a4(-/-) knockout mice. Measurements of the auditory brainstem response (ABR) showed significantly elevated thresholds in homozygous mutant mice, which indicate severe hearing impairment. Heterozygote thresholds were normal. Analysis of paint-filled inner ears and sections from E16.5 embryos revealed a marked expansion of cochlear and endolymphatic ducts in Atp6v0a4(-/-) mice. A regulatory link between Atp6v0a4, Foxi1 and Pds has been reported and we found that the endolymphatic sac of Atp6v0a4(-/-) mice expresses both Foxi1 and Pds, which suggests a downstream position of Atp6v0a4. These mutants also showed a lack of endocochlear potential, suggesting a functional defect of the stria vascularis on the lateral wall of the cochlear duct. However, the main K(+) channels involved in the generation of endocochlear potential, Kcnj10 and Kcnq1, are strongly expressed in Atp6v0a4(-/-) mice. Our results lead to a better understanding of the role of this proton pump in hearing function.


Subject(s)
Ear, Inner/enzymology , Ear, Inner/pathology , Endolymph/enzymology , Hearing Loss/enzymology , Hearing Loss/pathology , Protein Subunits/deficiency , Proton-Translocating ATPases/deficiency , Animals , Animals, Newborn , Anion Transport Proteins/metabolism , Ear, Inner/physiopathology , Endolymphatic Sac/pathology , Endolymphatic Sac/physiopathology , Epithelium/metabolism , Epithelium/pathology , Evoked Potentials, Auditory , Forkhead Transcription Factors/metabolism , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/ultrastructure , Hearing Loss/physiopathology , Humans , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Mutation/genetics , Phenotype , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/metabolism , Proton-Translocating ATPases/metabolism , Stria Vascularis/metabolism , Stria Vascularis/pathology , Sulfate Transporters , Vacuolar Proton-Translocating ATPases
7.
Dev Dyn ; 242(3): 269-80, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23233153

ABSTRACT

BACKGROUND: Carbonic anhydrases (CAs), which catalyze CO(2) hydration to bicarbonate and protons, have been suggested to regulate potassium homeostasis and endocochlear potential in the mammalian cochlea. Sixteen mammalian CA isozymes are currently known. To understand the specific roles of CA isozymes in the inner ear, a systematic survey was conducted to reveal temporal and spatial expression patterns of all 16 CA isozymes during inner ear development. RESULTS: Our quantitative reverse transcriptase-polymerase chain reaction results showed that different tissues express unique combinations of CA isozymes. During inner ear development, transcripts of four cytosolic isozymes (Car1, Car2, Car3, and Car13), two membrane-bound isozymes (Car12 and Car14), and two CA-related proteins (Car8 and Car11) were expressed at higher levels than other isozymes. Spatial expression patterns of these isozymes within developing inner ears were determined by in situ hybridization. Each isozyme showed a unique expression pattern during development. For example, Car12 and Car13 expression closely overlapped with Pendrin, an anion exchanger, while Car2 overlapped with Na-K-ATPase in type II and IV otic fibrocytes, suggesting functional relationships in the inner ear. CONCLUSIONS: The temporal and spatial expression patterns of each CA isozyme suggest unique and differential roles in inner ear development and function.


Subject(s)
Carbonic Anhydrases/biosynthesis , Ear, Inner/embryology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , RNA, Messenger/biosynthesis , Animals , Anion Transport Proteins/biosynthesis , Ear, Inner/cytology , Ear, Inner/enzymology , Gene Expression Profiling , Isoenzymes/biosynthesis , Mice , Sulfate Transporters
8.
Int J Exp Pathol ; 93(6): 450-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23136997

ABSTRACT

Mutations in phosphoribosyl pyrophosphate synthetase 1 (PRPS1) are associated with a spectrum of non-syndromic to syndromic hearing loss. PRPS1 transcript levels have been shown to be regulated by the microRNA-376 genes. The long primary RNA transcript of the miR-376 RNA cluster members undergo extensive and simultaneous A → I editing at one or both of two specific sites (+4 and +44) in particular human and mouse tissues. The PRPS1 gene, which contains target sites for the edited version of miR-376a-5p within its 3'UTR, has been shown to be repressed in a tissue-specific manner. To investigate whether the transcription of Prps1 is regulated by miR-376 cluster members in the mouse inner ear, we first quantified the expression of the mature miR-376 RNAs by quantitative real-time-PCR. The spatio-temporal patterns of miR-376 expression were assessed by in situ hybridization. Finally, we examined whether A →I editing of pri-miR-376 RNAs occurs in mouse inner ear by direct sequencing. Our data showed that the miR-376a-3p, b-3p, c-3p are present in mouse embryonic inner ears and intensive expression of miR-376a-3p/b-3p was detected in the sensory epithelia and ganglia of both auditory and vestibular portions of the inner ear. In adult inner ear, the expression of miR-376a-3p/b-3p is restricted within ganglion neurons of auditory and vestibular systems as well as the cells in the stria vascularis. Only unedited pri-miR-376 RNAs were detected in the cochlea suggesting that the activity of PRPS1 in the inner ear may not be regulated through the editing of miR-376 cluster.


Subject(s)
Ear, Inner/enzymology , MicroRNAs/genetics , Ribose-Phosphate Pyrophosphokinase/genetics , Animals , Cochlea/embryology , Cochlea/enzymology , Ear, Inner/embryology , Female , Gene Expression Regulation , Hair Cells, Auditory, Inner/enzymology , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Neurons/enzymology , Real-Time Polymerase Chain Reaction , Vestibule, Labyrinth/embryology , Vestibule, Labyrinth/enzymology
9.
Cell Mol Life Sci ; 69(14): 2429-34, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22562580

ABSTRACT

Cisplatin is a widely used chemotherapeutic agent that causes significant hearing loss. Previous studies have shown that cisplatin exposure is associated with increase in reactive oxygen species (ROS) in the cochlea. The inner ear expresses a unique isoform of NADPH oxidase, NOX3. This enzyme may be the primary source of ROS generation in the cochlea. The knockdown of NOX3 by pretreatment with siRNA prevented cisplatin ototoxicity, as demonstrated by preservation of hearing thresholds and inner ear sensory cells. Trans-tympanic NOX3 siRNA reduced the expression of NOX3 and biomarkers of cochlear damage, including transient receptor vanilloid 1 (TRPV1) channel and kidney injury molecule-1 (KIM-1) in cochlear tissues. In addition, siRNA against NOX3 reduced apoptosis as demonstrated by TUNEL staining, and prevented the increased expression of Bax and abrogated the decrease in Bcl2 expression following cisplatin administration. Trans-tympanic administration of siRNA directed against NOX3 may provide a useful method of attenuating cisplatin ototoxicity. In this paper, we review recent publications dealing with the role of NOX3 in ototoxicity and the effects of siRNA against cisplatin-induced hearing loss.


Subject(s)
Hearing Loss/enzymology , Membrane Proteins/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , RNA, Small Interfering/metabolism , Cisplatin/toxicity , Ear, Inner/enzymology , Hearing Loss/chemically induced , Hearing Loss/pathology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Paraquat/toxicity , RNA Interference
10.
Clin Genet ; 82(1): 56-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21534946

ABSTRACT

Mutations in the TMPRSS3 gene are known to cause autosomal recessive non-syndromic hearing impairment (ARNSHI). After undergoing a genome scan, 10 consanguineous Pakistani families with ARNSHI were found to have significant or suggestive evidence of linkage to the TMPRSS3 region. In order to elucidate if the TMPRSS3 gene is responsible for ARNSHI in these families, the gene was sequenced using DNA samples from these families. Six TMPRSS3 variants were found to cosegregate in 10 families. None of these variants were detected in 500 control chromosomes. Four novel variants, three of which are missense [c.310G>A (p.Glu104Lys), c.767C>T (p.Ala256Val) and c.1273T>C (p.Cys425Arg)] and one nonsense [c.310G>T (p.Glu104Stop)], were identified. The pathogenicity of novel missense variants was investigated through bioinformatics analyses. Additionally, the previously reported deletion c.208delC (p.His70ThrfsX19) was identified in one family and the known mutation c.1219T>C (p.Cys407Arg) was found in five families, which makes c.1219T>C (p.Cys407Arg) as the most common TMPRSS3 mutation within the Pakistani population. Identification of these novel variants lends support to the importance of elements within the low-density lipoprotein receptor A (LDLRA) and serine protease domains in structural stability, ligand binding and proteolytic activity for proper TMPRSS3 function within the inner ear.


Subject(s)
Ear, Inner/pathology , Hearing Loss/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Serine Endopeptidases/genetics , Case-Control Studies , Chromosomes, Human, Pair 21 , Consanguinity , Ear, Inner/enzymology , Exons , Female , Genes, Recessive , Genetic Linkage , Genetic Loci , Hearing Loss/enzymology , Hearing Loss/pathology , Humans , Male , Models, Molecular , Mutation , Pedigree , Phenotype , Protein Structure, Tertiary
11.
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
12.
BMC Neurosci ; 12: 102, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21988729

ABSTRACT

BACKGROUND: Caspase-3 is one of the most downstream enzymes activated in the apoptotic pathway. In caspase-3 deficient mice, loss of cochlear hair cells and spiral ganglion cells coincide closely with hearing loss. In contrast with the auditory system, details of the vestibular phenotype have not been characterized. Here we report the vestibular phenotype and inner ear anatomy in the caspase-3 deficient (Casp3(-/-)) mouse strain. RESULTS: Average ABR thresholds of Casp3(-/-) mice were significantly elevated (P < 0.05) compared to Casp3(+/-) mice and Casp3(+/+) mice at 3 months of age. In DPOAE testing, distortion product 2F1-F2 was significantly decreased (P < 0.05) in Casp3(-/-) mice, whereas Casp3(+/-) and Casp3(+/+) mice showed normal and comparable values to each other. Casp3(-/-) mice were hyperactive and exhibited circling behavior when excited. In lateral canal VOR testing, Casp3(-/-) mice had minimal response to any of the stimuli tested, whereas Casp3(+/-) mice had an intermediate response compared to Casp3(+/+) mice. Inner ear anatomical and histological analysis revealed gross hypomorphism of the vestibular organs, in which the main site was the anterior semicircular canal. Hair cell numbers in the anterior- and lateral crista, and utricle were significantly smaller in Casp3(-/-) mice whereas the Casp3(+/-) and Casp3(+/+) mice had normal hair cell numbers. CONCLUSIONS: These results indicate that caspase-3 is essential for correct functioning of the cochlea as well as normal development and function of the vestibule.


Subject(s)
Caspase 3/deficiency , Ear, Inner/enzymology , Ear, Inner/physiopathology , Vestibular Diseases/enzymology , Vestibular Diseases/physiopathology , Animals , Behavior, Animal/physiology , Caspase 3/genetics , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Vestibular Diseases/genetics , Vestibule, Labyrinth/enzymology , Vestibule, Labyrinth/metabolism , Vestibule, Labyrinth/physiopathology
13.
Hear Res ; 282(1-2): 184-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21875659

ABSTRACT

Heterozygous mutations in the gene encoding chromodomain-DNA-binding-protein 7 (CHD7) cause CHARGE syndrome, a multiple anomaly condition which includes vestibular dysfunction and hearing loss. Mice with heterozygous Chd7 mutations exhibit semicircular canal dysgenesis and abnormal inner ear neurogenesis, and are an excellent model of CHARGE syndrome. Here we characterized Chd7 expression in mature middle and inner ears, analyzed morphological features of mutant ears and tested whether Chd7 mutant mice have altered responses to noise exposure and correlated those responses to inner and middle ear structure. We found that Chd7 is highly expressed in mature inner and outer hair cells, spiral ganglion neurons, vestibular sensory epithelia and middle ear ossicles. There were no obvious defects in individual hair cell morphology by prestin immunostaining or scanning electron microscopy, and cochlear innervation appeared normal in Chd7(Gt)(/+) mice. Hearing thresholds by auditory brainstem response (ABR) testing were elevated at 4 and 16 kHz in Chd7(Gt)(/+) mice, and there were reduced distortion product otoacoustic emissions (DPOAE). Exposure of Chd7(Gt)(/+) mice to broadband noise resulted in variable degrees of hair cell loss which inversely correlated with severity of stapedial defects. The degrees of hair cell loss and threshold shifts after noise exposure were more severe in wild type mice than in mutants. Together, these data indicate that Chd7(Gt)(/+) mice have combined conductive and sensorineural hearing loss, correlating with changes in both middle and inner ears.


Subject(s)
CHARGE Syndrome/enzymology , DNA-Binding Proteins/metabolism , Ear, Inner/enzymology , Ear, Middle/enzymology , Hearing Loss, Conductive/enzymology , Hearing Loss, Sensorineural/enzymology , Acoustic Stimulation , Age Factors , Animals , Auditory Threshold , CHARGE Syndrome/genetics , CHARGE Syndrome/pathology , CHARGE Syndrome/physiopathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Ear, Inner/abnormalities , Ear, Inner/physiopathology , Ear, Inner/ultrastructure , Ear, Middle/abnormalities , Ear, Middle/physiopathology , Ear, Middle/ultrastructure , Evoked Potentials, Auditory, Brain Stem , Female , Genes, Reporter , Hearing Loss, Conductive/genetics , Hearing Loss, Conductive/pathology , Hearing Loss, Conductive/physiopathology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/physiopathology , Immunohistochemistry , Male , Mice , Mice, 129 Strain , Mice, Transgenic , Microscopy, Electron, Scanning , Molecular Motor Proteins/metabolism , Mutation , Noise , Otoacoustic Emissions, Spontaneous , Promoter Regions, Genetic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
14.
PLoS One ; 6(1): e16547, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21304972

ABSTRACT

BACKGROUND: The endothelial-blood/tissue barrier is critical for maintaining tissue homeostasis. The ear harbors a unique endothelial-blood/tissue barrier which we term "blood-labyrinth-barrier". This barrier is critical for maintaining inner ear homeostasis. Disruption of the blood-labyrinth-barrier is closely associated with a number of hearing disorders. Many proteins of the blood-brain-barrier and blood-retinal-barrier have been identified, leading to significant advances in understanding their tissue specific functions. In contrast, capillaries in the ear are small in volume and anatomically complex. This presents a challenge for protein analysis studies, which has resulted in limited knowledge of the molecular and functional components of the blood-labyrinth-barrier. In this study, we developed a novel method for isolation of the stria vascularis capillary from CBA/CaJ mouse cochlea and provided the first database of protein components in the blood-labyrinth barrier as well as evidence that the interaction of Na(+)/K(+)-ATPase α1 (ATP1A1) with protein kinase C eta (PKCη) and occludin is one of the mechanisms of loud sound-induced vascular permeability increase. METHODOLOGY/PRINCIPAL FINDINGS: Using a mass-spectrometry, shotgun-proteomics approach combined with a novel "sandwich-dissociation" method, more than 600 proteins from isolated stria vascularis capillaries were identified from adult CBA/CaJ mouse cochlea. The ion transporter ATP1A1 was the most abundant protein in the blood-labyrinth barrier. Pharmacological inhibition of ATP1A1 activity resulted in hyperphosphorylation of tight junction proteins such as occludin which increased the blood-labyrinth-barrier permeability. PKCη directly interacted with ATP1A1 and was an essential mediator of ATP1A1-initiated occludin phosphorylation. Moreover, this identified signaling pathway was involved in the breakdown of the blood-labyrinth-barrier resulting from loud sound trauma. CONCLUSIONS/SIGNIFICANCE: The results presented here provide a novel method for capillary isolation from the inner ear and the first database on protein components in the blood-labyrinth-barrier. Additionally, we found that ATP1A1 interaction with PKCη and occludin was involved in the integrity of the blood-labyrinth-barrier.


Subject(s)
Ear, Inner/enzymology , Protein Kinase C/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Capillaries , Databases, Protein , Ear, Inner/blood supply , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Occludin , Proteomics/methods , Sodium-Potassium-Exchanging ATPase/metabolism
15.
Am J Hum Genet ; 88(1): 19-29, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21185009

ABSTRACT

The DFNB74 locus for autosomal-recessive, nonsyndromic deafness segregating in three families was previously mapped to a 5.36 Mb interval on chromosome 12q14.2-q15. Subsequently, we ascertained five additional consanguineous families in which deafness segregated with markers at this locus and refined the critical interval to 2.31 Mb. We then sequenced the protein-coding exons of 18 genes in this interval. The affected individuals of six apparently unrelated families were homozygous for the same transversion (c.265T>G) in MSRB3, which encodes a zinc-containing methionine sulfoxide reductase B3. c.265T>G results in a substitution of glycine for cysteine (p.Cys89Gly), and this substitution cosegregates with deafness in the six DFNB74 families. This cysteine residue of MSRB3 is conserved in orthologs from yeast to humans and is involved in binding structural zinc. In vitro, p.Cys89Gly abolished zinc binding and MSRB3 enzymatic activity, indicating that p.Cys89Gly is a loss-of-function allele. The affected individuals in two other families were homozygous for a transition mutation (c.55T>C), which results in a nonsense mutation (p.Arg19X) in alternatively spliced exon 3, encoding a mitochondrial localization signal. This finding suggests that DFNB74 deafness is due to a mitochondrial dysfunction. In a cohort of 1,040 individuals (aged 53-67 years) of European ancestry, we found no association between 17 tagSNPs for MSRB3 and age-related hearing loss. Mouse Msrb3 is expressed widely. In the inner ear, it is found in the sensory epithelium of the organ of Corti and vestibular end organs as well as in cells of the spiral ganglion. Taken together, MSRB3-catalyzed reduction of methionine sulfoxides to methionine is essential for hearing.


Subject(s)
Deafness/enzymology , Deafness/genetics , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Aged , Animals , Base Sequence , Binding Sites/genetics , Carrier Proteins/genetics , Cohort Studies , Ear, Inner/enzymology , Exons/genetics , Female , Genes, Recessive , Genetic Linkage , Genetic Loci , Hearing Loss/genetics , Homozygote , Humans , Male , Methionine Sulfoxide Reductases , Mice , Middle Aged , Molecular Sequence Data , Mutation , Polymorphism, Single Nucleotide , White People/genetics
16.
Mamm Genome ; 22(3-4): 156-69, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21161235

ABSTRACT

The inner ear consists of the cochlea (the organ of hearing) and the vestibular system (the organs of balance). Within the vestibular system, linear acceleration and gravity are detected by the saccule and utricle. Resting above the neurosensory epithelia of these organs are otoconia, minute proteinaceous and crystalline (calcite) inertial masses that shift under the physical forces imparted by linear movements and gravity. It is the transduction and sensation of these movements and their integration with vision and proprioceptive inputs that contribute to the sensation of balance. It has been proposed that a reactive oxygen species- (ROS-) generating NADPH oxidase comprising the gene products of the Nox3, Noxo1, and Cyba genes plays a critical and constructive role in the process of inner-ear development, specifically, the deposition of otoconia. Inactivation in mouse of any of the NADPH oxidase components encoded by the Nox3, Noxo1, or Cyba gene results in the complete congenital absence of otoconia and profound vestibular dysfunction. Here we describe our use of PCR, reverse transcription-PCR (RT-PCR), and rapid amplification of cDNA ends (RACE) with traditional and high-throughput (HTP) sequencing technologies to extend and complete the molecular characterization of an allelic series of seven mutations in the Nox3 gene. Collectively, the mutation spectrum includes an endogenous retrovirus insertion, two missense mutations, a splice donor mutation, a splice acceptor mutation, premature translational termination, and a small duplication. Together, these alleles provide tools to investigate the mechanisms of otoconial deposition over development, throughout aging, and in various disease states.


Subject(s)
Ear, Inner/enzymology , Mice/genetics , Mutation , NADPH Oxidases/genetics , Alleles , Animals , Base Sequence , DNA Mutational Analysis , Ear, Inner/growth & development , Mice/metabolism , Mice, Inbred C57BL , Mice, Inbred Strains , Molecular Sequence Data , NADPH Oxidases/metabolism
17.
Acta Otolaryngol ; 131(3): 242-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21189050

ABSTRACT

CONCLUSIONS: Cyclo-oxygenase-2 (COX-2) enzyme would not appear to be constitutively expressed in human perilymph while it is always induced in the perilymph of patients affected by sensorineural hearing loss (SNHL). The COX-2 isoform may be involved in hearing loss and, therefore, pathological states of the inner ear should possibly be further analyzed to clarify the clinical relevance of prostaglandin and selective COX-2 antagonist therapy. OBJECTIVES: Perilymph samples from a group of patients with bilateral SNHL and another with conductive hearing loss were collected to evaluate the presence of the COX-2 enzyme. The possible correlation between different causes of deafness and the expression of COX-2 in the human ear was studied. METHODS: A prospective clinical study of 14 patients with severe or profound hearing loss who underwent cochlear implant surgery and 4 patients with conductive hearing loss who underwent stapes surgery was carried out. Western blot analysis of perilymph samples was performed with monoclonal anti-human COX-2 antibody. RESULTS: COX-2 enzyme was detected in all patients affected by SNHL and was absent in all those with conductive hearing loss due to otosclerosis.


Subject(s)
Cyclooxygenase 2/metabolism , Hearing Loss, Conductive/enzymology , Hearing Loss, Sensorineural/enzymology , Perilymph/enzymology , Adolescent , Adult , Aged , Blotting, Western , Child, Preschool , Ear, Inner/enzymology , Female , Humans , Male , Middle Aged , Prospective Studies , Young Adult
18.
Development ; 137(18): 3139-50, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20736290

ABSTRACT

Inner ear neurogenesis is positively regulated by the pro-neural bHLH transcription factors Ngn1 and NeuroD, but the factors that act upstream of this regulation are not well understood. Recent evidence in mouse and Drosophila suggests that neural development depends on proper chromatin remodeling, both for maintenance of neural stem cells and for proper neuronal differentiation. Here, we show that CHD7, an ATP-dependent chromatin remodeling enzyme mutated in human CHARGE syndrome, is necessary for proliferation of inner ear neuroblasts and inner ear morphogenesis. Conditional deletion of Chd7 in the developing otocyst using Foxg1-Cre resulted in cochlear hypoplasia and complete absence of the semicircular canals and cristae. Conditional knockout and null otocysts also had reductions in vestibulo-cochlear ganglion size and neuron number in combination with reduced expression of Ngn1, Otx2 and Fgf10, concurrent with expansion of the neural fate suppressor Tbx1 and reduced cellular proliferation. Heterozygosity for Chd7 mutations had no major effects on expression of otic patterning genes or on cell survival, but resulted in decreased proliferation within the neurogenic domain. These data indicate that epigenetic regulation of gene expression by CHD7 must be tightly coordinated for proper development of inner ear neuroblasts.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Ear, Inner/embryology , Ear, Inner/enzymology , Gene Expression Regulation, Developmental , Neurogenesis , Animals , Cell Proliferation , DNA Helicases/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Ear, Inner/cytology , Ear, Inner/innervation , Epigenesis, Genetic , Female , Humans , Male , Mice , Mice, Knockout
19.
Brain Res ; 1355: 16-30, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20633544

ABSTRACT

Mouse UBPy (mUBPy) belongs to the family of ubiquitin-specific processing proteases (UBPs). In this study we have investigated the expression of mUBPy in the brain and sensory organs of mouse at different embryonic stages (E9, E11, E13, E15, E17, E19) and during the postnatal stages P0, P1, P2, P4 and P5 using Western blot and immunohistochemistry. mUBPy-immunoreactive cell bodies first appeared at stage E11 in several brain regions, particularly in the walls surrounding the vesicles and the ventricles. Subsequently, at stage E13, new mUBPy-positive cells appeared in the corpus striatum, the caudate nucleus, the thalamus, the epithalamus, the hypothalamus and the pons. At E15 the mUBPy pattern was very similar to that observed at E13, whereas at stage E17 mUBPy-immunoreactivity significantly decreased and a high number of mUBPy-immunoreactive cells was found only to line the third ventricle and within the mantle layer of the fourth ventricle. At E19 and P0, no mUBPy-immunoreactive element was found in the brain. At the postnatal stages P2 and P5, mUBPy-positive cells were detected in all subdivisions of the brain, with high concentrations in several cortex regions. Double labeling with the mUBPy antiserum and antisera against specific cell markers showed that the enzyme is expressed both in neurons and astrocytes. Outside the brain, mUBPy was detected, from stage E11, in the eye, within the lens and the cornea, in the inner ear, at the level of the cochlear and vestibular systems and in the olfactory epithelium. The spatio-temporal expression of mUBPy suggests that the enzyme may be involved in neuroregulatory processes during embryogenesis.


Subject(s)
Brain/enzymology , Ear, Inner/enzymology , Endopeptidases/biosynthesis , Endosomal Sorting Complexes Required for Transport/biosynthesis , Eye/enzymology , Olfactory Mucosa/enzymology , Ubiquitin Thiolesterase/biosynthesis , Animals , Brain/embryology , Brain/growth & development , Ear, Inner/embryology , Ear, Inner/growth & development , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Eye/embryology , Eye/growth & development , Female , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Male , Mice , Olfactory Mucosa/embryology , Olfactory Mucosa/growth & development , Ubiquitin Thiolesterase/genetics
20.
Am J Physiol Cell Physiol ; 296(3): C544-57, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19144862

ABSTRACT

Reissner's membrane epithelium forms much of the barrier that produces and sustains the large ionic differences between cochlear endolymph and perilymph. We have reported that Reissner's membrane contributes to normal cochlear function by absorbing Na(+) from endolymph via amiloride-sensitive channels in gerbil inner ear. We used mouse Reissner's membrane to 1) identify candidate genes involved in the Na(+) transport pathway, 2) determine whether their level of expression was regulated by the synthetic glucocorticoid dexamethasone, and 3) obtain functional evidence for the physiological importance of these genes. Transcripts were present for alpha-, beta-, and gamma-subunits of epithelial Na(+) channel (ENaC); corticosteroid receptors GR (glucocorticoid receptor) and MR (mineralocorticoid receptor); GR agonist regulator 11beta-hydroxysteroid dehydrogenase (HSD) type 1 (11beta-HSD1); Na(+) transport control components SGK1, Nedd4-2, and WNKs; and K(+) channels and Na(+)-K(+)-ATPase. Expression of the MR agonist regulator 11beta-HSD2 was not detected. Dexamethasone upregulated transcripts for alpha- and beta-subunits of ENaC ( approximately 6- and approximately 3-fold), KCNK1 ( approximately 3-fold), 11beta-HSD1 ( approximately 2-fold), SGK1 ( approximately 2-fold), and WNK4 ( approximately 3-fold). Transepithelial currents from the apical to the basolateral side of Reissner's membrane were sensitive to amiloride (IC(50) approximately 0.7 muM) and benzamil (IC(50) approximately 0.1 muM), but not EIPA (IC(50) approximately 34 muM); amiloride-blocked transepithelial current was not immediately changed by forskolin/IBMX. Currents were reduced by ouabain, lowered bath Na(+) concentration (from 150 to 120 mM), and K(+) channel blockers (XE-991, Ba(2+), and acidification from pH 7.4 to 6.5). Dexamethasone-stimulated current and gene expression were reduced by mifepristone, but not spironolactone. These molecular, pharmacological, and functional observations are consistent with Na(+) absorption by mouse Reissner's membrane, which is mediated by apical ENaC and/or other amiloride-sensitive channels, basolateral Na(+)-K(+)-ATPase, and K(+)-permeable channels and is under the control of glucocorticoids. These results provide an understanding and a molecular definition of an important transport function of Reissner's membrane epithelium in the homeostasis of cochlear endolymph.


Subject(s)
Dexamethasone/pharmacology , Ear, Inner/metabolism , Endolymph/metabolism , Epithelial Cells/drug effects , Epithelial Sodium Channels/drug effects , Glucocorticoids/pharmacology , Sodium/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Amiloride/pharmacology , Animals , Cyclic AMP/metabolism , Ear, Inner/drug effects , Ear, Inner/enzymology , Endosomal Sorting Complexes Required for Transport , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Gene Expression Profiling/methods , Hormone Antagonists/pharmacology , Immediate-Early Proteins/metabolism , Membrane Potentials , Mice , Mice, Inbred C57BL , Nedd4 Ubiquitin Protein Ligases , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Sodium Channel Blockers/pharmacology , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Time Factors , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...