Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Proc Natl Acad Sci U S A ; 119(37): e2207433119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36074819

ABSTRACT

A cardinal feature of the auditory pathway is frequency selectivity, represented in a tonotopic map from the cochlea to the cortex. The molecular determinants of the auditory frequency map are unknown. Here, we discovered that the transcription factor ISL1 regulates the molecular and cellular features of auditory neurons, including the formation of the spiral ganglion and peripheral and central processes that shape the tonotopic representation of the auditory map. We selectively knocked out Isl1 in auditory neurons using Neurod1Cre strategies. In the absence of Isl1, spiral ganglion neurons migrate into the central cochlea and beyond, and the cochlear wiring is profoundly reduced and disrupted. The central axons of Isl1 mutants lose their topographic projections and segregation at the cochlear nucleus. Transcriptome analysis of spiral ganglion neurons shows that Isl1 regulates neurogenesis, axonogenesis, migration, neurotransmission-related machinery, and synaptic communication patterns. We show that peripheral disorganization in the cochlea affects the physiological properties of hearing in the midbrain and auditory behavior. Surprisingly, auditory processing features are preserved despite the significant hearing impairment, revealing central auditory pathway resilience and plasticity in Isl1 mutant mice. Mutant mice have a reduced acoustic startle reflex, altered prepulse inhibition, and characteristics of compensatory neural hyperactivity centrally. Our findings show that ISL1 is one of the obligatory factors required to sculpt auditory structural and functional tonotopic maps. Still, upon Isl1 deletion, the ensuing central plasticity of the auditory pathway does not suffice to overcome developmentally induced peripheral dysfunction of the cochlea.


Subject(s)
Auditory Pathways , Cochlear Nucleus , Hair Cells, Auditory , LIM-Homeodomain Proteins , Neurogenesis , Spiral Ganglion , Transcription Factors , Animals , Auditory Pathways/embryology , Cochlea/embryology , Cochlea/innervation , Cochlear Nucleus/embryology , Hair Cells, Auditory/physiology , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/physiology , Mice , Neurogenesis/genetics , Spiral Ganglion/enzymology , Transcription Factors/genetics , Transcription Factors/physiology
2.
Biomed Res Int ; 2015: 925890, 2015.
Article in English | MEDLINE | ID: mdl-25918725

ABSTRACT

Rapamycin is an antifungal agent with immunosuppressive properties. Rapamycin inhibits the mammalian target of rapamycin (mTOR) by blocking the mTOR complex 1 (mTORC1). mTOR is an atypical serine/threonine protein kinase, which controls cell growth, cell proliferation, and cell metabolism. However, less is known about the mTOR pathway in the inner ear. First, we evaluated whether or not the two mTOR complexes (mTORC1 and mTORC2, resp.) are present in the mammalian cochlea. Next, tissue explants of 5-day-old rats were treated with increasing concentrations of rapamycin to explore the effects of rapamycin on auditory hair cells and spiral ganglion neurons. Auditory hair cell survival, spiral ganglion neuron number, length of neurites, and neuronal survival were analyzed in vitro. Our data indicates that both mTOR complexes are expressed in the mammalian cochlea. We observed that inhibition of mTOR by rapamycin results in a dose dependent damage of auditory hair cells. Moreover, spiral ganglion neurite number and length of neurites were significantly decreased in all concentrations used compared to control in a dose dependent manner. Our data indicate that the mTOR may play a role in the survival of hair cells and modulates spiral ganglion neuronal outgrowth and neurite formation.


Subject(s)
Hair Cells, Auditory/enzymology , Neurites/enzymology , Sirolimus/adverse effects , Spiral Ganglion/enzymology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Survival/drug effects , Hair Cells, Auditory/pathology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Neurites/pathology , Rats , Rats, Wistar , Sirolimus/pharmacology , Spiral Ganglion/pathology , TOR Serine-Threonine Kinases/metabolism
3.
PLoS One ; 10(3): e0121599, 2015.
Article in English | MEDLINE | ID: mdl-25811375

ABSTRACT

The kinase Akt is a key downstream mediator of the phosphoinositide-3-kinase signaling pathway and participates in a variety of cellular processes. Akt comprises three isoforms each encoded by a separate gene. There is evidence to indicate that Akt is involved in the survival and protection of auditory hair cells in vitro. However, little is known about the physiological role of Akt in the inner ear-especially in the intact animal. To elucidate this issue, we first analyzed the mRNA expression of the three Akt isoforms in the inner ear of C57/BL6 mice by real-time PCR. Next, we tested the susceptibility to gentamicin-induced auditory hair cell loss in isoform-specific Akt knockout mice compared to wild-types (C57/BL6) in vitro. To analyze the effect of gene deletion in vivo, hearing and cochlear microanatomy were evaluated in Akt isoform knockout animals. In this study, we found that all three Akt isoforms are expressed in the cochlea. Our results further indicate that Akt2 and Akt3 enhance hair cell resistance to ototoxicity, while Akt1 does not. Finally, we determined that untreated Akt1 and Akt2/Akt3 double knockout mice display significant hearing loss, indicating a role for these isoforms in normal hearing. Taken together, our results indicate that each of the Akt isoforms plays a distinct role in the mammalian inner ear.


Subject(s)
Hair Cells, Auditory/cytology , Hair Cells, Auditory/enzymology , Hearing/physiology , Mammals/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Animals, Newborn , Cell Survival , Disease Susceptibility , Evoked Potentials, Auditory, Brain Stem , Gene Expression Regulation, Enzymologic , Gentamicins , Isoenzymes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spiral Ganglion/enzymology , Stria Vascularis/enzymology
4.
Cell Mol Neurobiol ; 34(7): 1011-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24935409

ABSTRACT

Matrix metalloproteinases (MMPs) play an important role in modeling of the extracellular matrix. There is increasing evidence that these proteases are important in neurite elongation and axonal guidance during development in the central nervous system and retina. Moreover, they are also expressed after acute injury and can be the key mediators of pathogenesis. However, the role of MMPs in the inner ear is largely unknown. Our group recently demonstrated that general inhibition of MMPs resulted in auditory hair cell loss in vitro. In the present study, we investigated the role of MMPs in inner ear spiral ganglion neuron (SGN) survival, neuritogenesis and neurite extension by blocking MMPs known to be involved in axonal guidance, neurite elongation, and apoptosis in other neuronal systems. Spiral ganglion (SG) explants from 5-day-old Wistar rats were treated with different concentrations of the general MMP inhibitor GM6001, a specific MMP-2 inhibitor, and a specific MMP-9 inhibitor, in vitro. The general inhibitor of MMPs and the specific inhibition of MMP-2 significantly reduced both the number of neurites that extended from SG explants, as well as the length of individual neurites. However, neither the general inhibitor of MMPs nor the specific inhibition of MMP-2 influenced SGN survival. Inhibition of MMP-9 had no influence on SGNs. The data suggest that MMPs, and more specifically MMP-2, influence the growth of developing afferent neurites in the mammalian inner ear in vivo.


Subject(s)
Ear, Inner/cytology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Neurons/enzymology , Spiral Ganglion/enzymology , Animals , Cell Count , Cell Survival/drug effects , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/enzymology , Matrix Metalloproteinase 9/genetics , Neurites/drug effects , Neurites/enzymology , Neurons/cytology , Neurons/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Spiral Ganglion/cytology , Spiral Ganglion/drug effects
5.
Kulak Burun Bogaz Ihtis Derg ; 23(2): 104-11, 2013.
Article in English | MEDLINE | ID: mdl-23611324

ABSTRACT

OBJECTIVES: This study aims to investigate the effect of exogenous glucocorticoid exposure in the prenatal period on hearing and to evaluate the effectiveness of caffeic acid phenethyl ester (CAPE), an antioxidant, on the prevention of the inner ear injury. MATERIALS AND METHODS: Dexamethasone was given to half of twelve Sprague-Dawley pregnant rats and the distilled water was given to the remaining half. The real subjects were obtained by born of the offsprings. When the all subjects were two months of age, they were exposed to 110 dB noise during four hours as a stressor effect. These subjects were divided into three groups. Group 1: subjects to whose mothers were given distilled water; Group 2: subjects to whose mothers were given dexamethasone; Group 3: subjects to whose mothers were given dexamethasone and CAPE. RESULTS: While there was no statistical significance in hearing thresholds which exposed and not exposed to exogenous dexamethasone before noise exposure (p>0.05) between the groups, the elevation of hearing thresholds of subjects which exposed to exogenous dexamethasone was statistically significant after noise exposure (p<0.05). CONCLUSION: Prenatally exposure to exogenous glucocorticoids may cause the inner ear susceptible to the effect of noise, and CAPE is effective to prevent the possible damage.


Subject(s)
Caffeic Acids/administration & dosage , Dexamethasone/adverse effects , Ear, Inner/drug effects , Glucocorticoids/adverse effects , Hearing/drug effects , Phenylethyl Alcohol/analogs & derivatives , Prenatal Exposure Delayed Effects/chemically induced , Animals , Auditory Threshold/drug effects , Caspases/analysis , Dexamethasone/administration & dosage , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Glucocorticoids/administration & dosage , Immunohistochemistry , Noise/adverse effects , Organ of Corti/enzymology , Phenylethyl Alcohol/administration & dosage , Pregnancy , Rats , Rats, Sprague-Dawley , Spiral Ganglion/enzymology , Stress, Physiological , Stria Vascularis/enzymology
6.
Anat Rec (Hoboken) ; 296(2): 326-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23165776

ABSTRACT

Mitochondrial degeneration in the inner ear is likely a contributing factor in age-related hearing loss and other otopathologies such as Meniere's disease. Most mitochondrial proteins are synthesized in the cytosol and imported through the mitochondrial membranes by translocators. The translocase of the outer membrane (Tom) is the universal entry gate for all proteins that are imported into mitochondria. Altered function of the translocator could alter protein transport into the mitochondria, and disrupt function. In this study, we determined the immunolocalization of Tom20, a major mitochondrial protein import receptor, in microdissected human cochlea frozen sections obtained from postmortem autopsy and celloidin-embedded archival specimens. We used affinity purified rabbit polyclonal antibodies against Tom20. We also determined the Tom20 immunolocalization in the mouse inner ear. In the human and mouse cochlea, Tom20 was ubiquitously distributed in the organ of Corti, allowing well-delineated visualization of inner and outer hair cells. Tom20 immunoreactivity localized in the cytoplasm of spiral ganglia neurons. In the inner ear of aged subjects with Meniere's disease, there was decreased expression of Tom20. These results suggest that Tom20 can be used in the inner ear as a marker for mitochondrial protein import.


Subject(s)
Cochlea/enzymology , Immunohistochemistry , Membrane Transport Proteins/analysis , Meniere Disease/enzymology , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Receptors, Cell Surface/analysis , Adolescent , Aged , Aged, 80 and over , Animals , Biomarkers/analysis , Child , Female , Fixatives , Formaldehyde , Frozen Sections , Hair Cells, Auditory/enzymology , Humans , Male , Mice , Mice, Inbred CBA , Microdissection , Middle Aged , Mitochondrial Precursor Protein Import Complex Proteins , Rats , Spiral Ganglion/enzymology , Tissue Fixation/methods
7.
Hear Res ; 283(1-2): 33-44, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22154930

ABSTRACT

Regrowth of peripheral spiral ganglion neuron (SGN) fibers is a primary objective in efforts to improve cochlear implant outcomes and to potentially reinnervate regenerated hair cells. Cyclic adenosine monophosphate (cAMP) regulates neurite growth and guidance via activation of protein kinase A (PKA) and Exchange Protein directly Activated by Cylic AMP (Epac). Here we explored the effects of cAMP signaling on SGN neurite length in vitro. We find that the cAMP analog, cpt-cAMP, exerts a biphasic effect on neurite length; increasing length at lower concentrations and reducing length at higher concentrations. This biphasic response occurs in cultures plated on laminin, fibronectin, or tenascin C suggesting that it is not substrate dependent. cpt-cAMP also reduces SGN neurite branching. The Epac-specific agonist, 8-pCPT-2'-O-Me-cAMP, does not alter SGN neurite length. Constitutively active PKA isoforms strongly inhibit SGN neurite length similar to higher levels of cAMP. Chronic membrane depolarization activates PKA in SGNs and also inhibits SGN neurite length. However, inhibition of PKA fails to rescue neurite length in depolarized cultures implying that activation of PKA is not necessary for the inhibition of SGN neurite length by chronic depolarization. Expression of constitutively active phosphatidylinositol 3-kinase, but not c-Jun N-terminal kinase, isoforms partially rescues SGN neurite length in the presence of activated PKA. Taken together, these results suggest that activation of cAMP/PKA represents a potential strategy to enhance SGN fiber elongation following deafness; however such therapies will likely require careful titration so as to promote rather than inhibit nerve fiber regeneration.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Nerve Regeneration , Neurons/enzymology , Spiral Ganglion/enzymology , Animals , Animals, Newborn , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Activators/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potentials , Nerve Regeneration/drug effects , Neurites/enzymology , Neurons/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Recombinant Fusion Proteins/metabolism , Signal Transduction , Spiral Ganglion/drug effects , Thionucleotides/pharmacology , Tissue Culture Techniques , Transfection
8.
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
9.
Hear Res ; 278(1-2): 77-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21554942

ABSTRACT

Jun N-terminal kinase (JNK) is a multifunctional protein kinase crucial for neuronal apoptosis as well as neurite growth. We have previously shown that JNK activity is correlated with spiral ganglion neuron (SGN) apoptosis following hair cell loss in rats (Alam et al., 2007) implying that JNK inhibition may have therapeutic potential to protect SGNs in deaf individuals. Here we investigated the role of JNK in neurite outgrowth from cultured neonatal rat and mouse SGNs. We show that JNK is required for initial growth of neurites and for continued extension of already established neurites. The effect of JNK inhibition on neurite growth is rapid and is also rapidly reversible after washout of the inhibitor. Using phosphoJNK immunoreactivity as an indicator, we show that JNK is activated in growth cones within 30 min after transfer to medium lacking neurotrophic stimuli (5 K medium) but activation in the nucleus and soma requires hours. By transfecting epitope-tagged JNK1, JNK2, or JNK3 isoforms into SGNs, we found that all are present in the nucleus and cytoplasm and that there is no preferential redistribution to the nucleus after transfer to 5 K medium. Cotransfection of dominant-negative (dn) JNK1 and JNK2 into SGNs reduced neurite growth, although transfection of dnJNK1 or dnJNK2 alone had no significant effect. SGNs cultured from JNK3(-/-) mice showed reduced neurite growth that was further reduced by transfection of dnJNK1 and dnJNK2. This indicates that all three JNK isoforms promote SGN neurite growth although there may be functional redundancy between JNK1 and JNK2.


Subject(s)
MAP Kinase Signaling System , Neurites/enzymology , Neurites/ultrastructure , Spiral Ganglion/enzymology , Spiral Ganglion/innervation , Animals , Cells, Cultured , Enzyme Activation , Kinetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/deficiency , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Neurons/enzymology , Neurons/ultrastructure , Phosphorylation , Rats , Spiral Ganglion/ultrastructure , Subcellular Fractions/enzymology , Transfection
10.
Proc Natl Acad Sci U S A ; 107(29): 13051-6, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20616061

ABSTRACT

A significantly increased risk for dominant sensorineural deafness in patients who have Hirschsprung disease (HSCR) caused by endothelin receptor type B and SOX10 has been reported. Despite the fact that c-RET is the most frequent causal gene of HSCR, it has not been determined whether impairments of c-Ret and c-RET cause congenital deafness in mice and humans. Here, we show that impaired phosphorylation of c-Ret at tyrosine 1062 causes HSCR-linked syndromic congenital deafness in c-Ret knockin (KI) mice. The deafness involves neurodegeneration of spiral ganglion neurons (SGNs) with not only impaired phosphorylation of Akt and NF-kappaB but decreased expression of calbindin D28k in inner ears. The congenital deafness involving neurodegeneration of SGNs in c-Ret KI mice was rescued by introducing constitutively activated RET. Taken together with our results for three patients with congenital deafness with c-RET-mediated severe HSCR, our results indicate that c-Ret and c-RET are a deafness-related molecule in mice and humans.


Subject(s)
Hearing Loss/complications , Hearing Loss/enzymology , Hirschsprung Disease/complications , Hirschsprung Disease/enzymology , Proto-Oncogene Proteins c-ret/metabolism , Amino Acid Substitution/genetics , Animals , Deafness/complications , Deafness/enzymology , Enzyme Activation , Gene Knock-In Techniques , Hearing Loss/congenital , Immunohistochemistry , Mice , Mutation/genetics , NF-kappa B/metabolism , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/pathology , Neurons/ultrastructure , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/genetics , Spiral Ganglion/enzymology , Spiral Ganglion/pathology , Spiral Ganglion/ultrastructure
11.
Auris Nasus Larynx ; 37(3): 274-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19900774

ABSTRACT

OBJECTIVE: Fabry disease is characterized by genetic alpha-galactosidase A deficiency, resulting in accumulation of glycolipids (GL-3) and tissue damage. Hearing loss is also common and attributed to GL-3 accumulation in the inner ear. The only reported histological studies dealt with murine and human specimens. Accordingly, histopathological studies of the cochlea were performed on an alpha-galactosidase A deficient murine model of Fabry disease, using C57BL6/J mice as the controls. METHODS: The hearing ability was evaluated using the ABR threshold, while cochlear specimens were observed light microscopically and ultrathin temporal bone sections by TEM. RESULTS: HE staining showed no accumulation of GL-3 or abnormal cochlear morphology in the alpha-galactosidase A deficient mice, but toluidine blue staining and TEM revealed GL-3 accumulation in the stria vascularis and kidney. No GL-3 accumulation was detected in the C57BL6/J controls by either HE staining or TEM. The alpha-galactosidase A deficient mice and the controls showed no clear differences in the ABR threshold (hearing acuity), but for older animals the threshold was higher in the C57BL6/J controls. CONCLUSION: In summary, although the alpha-galactosidase A deficient mice showed no clear hearing loss, GL-3 accumulation was demonstrated in the cochlea.


Subject(s)
Cochlea/enzymology , Cochlea/pathology , Fabry Disease/enzymology , Fabry Disease/metabolism , Fabry Disease/pathology , Animals , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/physiology , Fabry Disease/genetics , Hearing Loss, Sensorineural/diagnosis , Mice , Mice, Inbred C57BL , Spiral Ganglion/enzymology , Spiral Ganglion/pathology
12.
Hear Res ; 253(1-2): 116-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19376215

ABSTRACT

Manganese superoxide dismutase 2 (SOD2) is a key metabolic anti-oxidant enzyme for detoxifying free radicals inside mitochondria. This study documents a gradient in expression of SOD2 by spiral ganglion cells in basal versus apical turn of cochlea that is consistent with differential vulnerability of high frequency hearing to free radical damage. Immunohistochemical methods were used to identify distribution of SOD2 in temporal bone sections from mice, rats, macaques, and humans. In mice and rats, both the proportion of SOD2 immunopositive type 1 spiral ganglion cells and the intensity of immunoreactivity were elevated near cochlear apex. In macaques and humans, the proportion of SO2 immunopositive spiral ganglion cells was equal across cochlear turn, but the intensity of immunoreactivity remained highest for ganglion cells near cochlear apex. Strong SOD2 immunoreactivity was also observed in human type 1 spiral ganglion cells. The average area density of SOD2 immunoreactivity in ganglion cells for each species and cochlear turn showed an allometric relationship with body weight, which is consistent with a conserved basal metabolic characteristic. These findings suggest that spiral ganglion cell responses to ROS exposure may vary along cochlear spiral with lower response capacity at cochlear base contributing to cumulative susceptibility to high frequency hearing loss.


Subject(s)
Spiral Ganglion/enzymology , Superoxide Dismutase/metabolism , Animals , Child, Preschool , Female , Humans , Immunohistochemistry , Macaca , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/metabolism , Models, Biological , Rats , Rats, Long-Evans , Rats, Wistar , Species Specificity , Spiral Ganglion/cytology , Spiral Ganglion/metabolism , Superoxides/metabolism , Tissue Distribution
13.
J Assoc Res Otolaryngol ; 10(1): 37-49, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19082858

ABSTRACT

Processing of sound in the cochlea involves both afferent and efferent innervation. The Na,K-ATPase (NKA) is essential for cells that maintain hyperpolarized membrane potentials and sodium and potassium concentration gradients. Heterogeneity of NKA subunit expression is one mechanism that tailors physiology to particular cellular demands. Therefore, to provide insight into molecular differences that distinguish the various innervation pathways in the cochlea, we performed a variety of double labeling experiments with antibodies against three of the alpha isoforms of the NKA (NKA alpha 1-3) and markers identifying particular subsets of neurons or supporting cells in whole mount preparations of the organ of Corti and spiral ganglion. We found that the NKA alpha 3 is abundantly expressed within the membranes of the spiral ganglion somata, the type I afferent terminals contacting the inner hair cells, and the medial efferent terminals contacting the outer hair cells. We also found expression of the NKA alpha 1 in the supporting cells that neighbor the inner hair cells and express the glutamate transporter GLAST. These findings suggest that both the NKA alpha 1 and NKA alpha 3 are poised to play an essential role in the regulation of the type I afferent synapses, the medial efferent synapses, and also glutamate transport from the afferent-inner hair cell synapse.


Subject(s)
Organ of Corti/enzymology , Sodium-Potassium-Exchanging ATPase/biosynthesis , Spiral Ganglion/enzymology , Animals , Biological Transport, Active/physiology , Excitatory Amino Acid Transporter 1/biosynthesis , Glutamic Acid/metabolism , Hair Cells, Auditory, Inner/enzymology , Immunohistochemistry , Isoenzymes/biosynthesis , Labyrinth Supporting Cells/enzymology , Neurons, Afferent/enzymology , Neurons, Efferent/enzymology , Organ of Corti/cytology , Presynaptic Terminals/enzymology , Rats , Spiral Ganglion/cytology , Synapses/physiology
14.
Article in Chinese | MEDLINE | ID: mdl-18717313

ABSTRACT

OBJECTIVE: To study the differences of regulation of sodium salicylate on the auditory brain stem response (ABR) threshold and expression of glutamic acid decarboxylase (GAD) protein in spiral ganglion of juvenile and adult guinea pigs. METHODS: Fourty juvenile guinea pigs which were born just four days and fourty adult guinea pigs which were born thirty days were selected. They were divided four groups (group A; group B; group C; group D). ABR threshold was detected before administration, after administration for 15 days and after administration stopped for 30 days. The protein expression of GAD were measured after administration for 15 days and after administration stopped for 30 days by the method of immunohistochemistry. RESULTS: ABR threshold of juvenile sodium salicylate groups (group C) was increased remarkably than that of before administration and the control after administration for 15 days (P < 0.001). ABR threshold of group C was returned to the level of that of before administration and after administration stopped for 30 days. ABR threshold of adult sodium salicylate groups (group D) was increased remarkably than that of before administration and the control after administration for 15 days (P < 0.001). ABR threshold of group D was kept the high level after administration stopped for 30 days. The protein expression of GAD of sodium salicylate groups (group C and D) was decreased than that of the control after administration for 15 days. The protein expression of group C was more visible regression than that of group D (t = 4.7, P < 0.001). The protein expression of group C was returned the level of before administration after administration stopped for 30 days, but the protein expression of group D was kept the high level. CONCLUSIONS: The results suggest that sodium salicylate can regulate differently ABR threshold and expression of GAD protein in spiral ganglion of juvenile and adult guinea pigs. The effects of sodium salicylate on ABR threshold and expression of GAD protein in spiral ganglion of juvenile pigs are more noticeable than that of adult guinea pigs, but these changes are easier to return the normal than that of adult guinea pigs.


Subject(s)
Auditory Threshold/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Glutamate Decarboxylase/metabolism , Sodium Salicylate/pharmacology , Spiral Ganglion/drug effects , Animals , Evoked Potentials, Auditory, Brain Stem/physiology , Guinea Pigs , Spiral Ganglion/enzymology
15.
J Neurotrauma ; 25(1): 72-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18355160

ABSTRACT

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


Subject(s)
Cochlea/enzymology , Hair Cells, Auditory/enzymology , Hearing Loss, Noise-Induced/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Noise/adverse effects , Acoustic Stimulation , Animals , Biomarkers/metabolism , Cell Death/physiology , Cochlea/pathology , Cochlea/physiopathology , DNA Fragmentation , Enzyme Activation/physiology , Female , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/physiopathology , In Situ Nick-End Labeling , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Spiral Ganglion/enzymology , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology , Time Factors
16.
Hear Res ; 235(1-2): 8-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17980526

ABSTRACT

Isolating cells from the cochlea to perform molecular biology assessment presents a challenge, because it is not possible to dissect pure cell pools by conventional methods. Thus, we set out to demonstrate that laser microdissection and pressure catapulting (LMPC) is superior to conventional manual cochlea dissection for this purpose. Spiral ganglions (SG) were isolated from neonatal rat cochleae by manual dissection and LMPC. Also, modioli were manually dissected. Total RNA was isolated from all three cell pools. In order to demonstrate contamination of the dissected cell pool, we determined the expression of type II iodothyronine deiodinase (D2), claudin 11 (Cld-11), neurofilament light chain (NF-L) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) transcripts by RT-PCR. The results showed that LMPC is not only a suitable method for selectively dissecting cochlear tissues, but in addition the molecular markers confirmed pure spiral ganglion cell pools without indication for any contamination by other cells. This indicates that LMPC is capable of providing a pure SG cell pool in contrast to conventional manual dissection. Therefore, LMPC presents a new technique for cochlear tissue separation improving the validity of molecular biological studies of the inner ear.


Subject(s)
Cell Separation/methods , Cochlea/chemistry , Lasers , Microdissection , RNA, Messenger/analysis , Spiral Ganglion/chemistry , Animals , Animals, Newborn , Claudins , Cochlea/cytology , Cochlea/enzymology , Frozen Sections , Glyceraldehyde-3-Phosphate Dehydrogenases/analysis , Iodide Peroxidase/analysis , Nerve Tissue Proteins/analysis , Neurofilament Proteins/analysis , Phenotype , Pressure , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Spiral Ganglion/enzymology , Iodothyronine Deiodinase Type II
17.
Hear Res ; 209(1-2): 76-85, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16055286

ABSTRACT

Mice, in which the genetics can be manipulated and the life span is relatively short, enable evaluation of the effects of specific gene expression on cochlear degeneration over time. Antioxidant enzymes such as Cu/Zn superoxide dismutase (SOD1) protect cells from toxic, reactive oxygen species and may be involved in age-related degeneration. The effects of SOD1 deletion and over-expression on the cochlea were examined in Sod1-null mice, Sod1 transgenic mice and in age- and genetics-matched controls. Auditory brainstem responses (ABR) were measured and cochleae were histologically examined. The absence of SOD1 resulted in hearing loss at an earlier age than in wildtype or heterozygous mice. The cochleae of the null mice had severe spiral ganglion cell degeneration at 7-9 months of age. The stria vascularis in the aged, null mice was thinner than in the heterozygous or wildtype mice. Over-expression of SOD1 did not protect against hearing loss except at 24 months of age. In conclusion, SOD1 seems important for survival of cochlear neurons and the stria vascularis, however even half the amount is sufficient and an over abundance does not provide much protection from age-related hearing loss.


Subject(s)
Aging/metabolism , Hearing Loss/enzymology , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Aging/pathology , Analysis of Variance , Animals , Brain Stem/physiology , Cochlea/enzymology , Cochlea/metabolism , Cochlea/pathology , Female , Gene Expression , Hearing Loss/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Organ of Corti/enzymology , Organ of Corti/metabolism , Organ of Corti/pathology , Presbycusis/physiopathology , Spiral Ganglion/enzymology , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Stria Vascularis/enzymology , Stria Vascularis/metabolism , Stria Vascularis/pathology , Superoxide Dismutase-1
18.
Hear Res ; 202(1-2): 21-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15811695

ABSTRACT

The aim of this study was to investigate the involvement of oxidative stress and apoptosis in an animal model of Meniere's disease. Endolymphatic hydrops (ELH) is generally accepted as the decisive histological characteristic of Meniere's disease. Closure of the endolymphatic duct (Kimura's method) was used to induce endolymphatic hydrops in guinea pigs. Sham-operated animals served as controls. After 4 weeks the animals operated showed a significant elevation of the hearing thresholds as measured by audiometric brainstem responses (ABR) pre- and postoperatively. Immediately after the second ABR measurement, the animals were sacrificed for further immunohistological examinations of the inner ear with specific antibodies to active caspase-3 (cas-3) as a marker for apoptosis and antibodies to 8-isoprostane (8-iso) and nitrotyrosine (NT) as indicators of oxidative stress. Compared with the sham-operated controls, hydropic cochleae showed strong immunostaining for both oxidative stress markers in spiral ganglion cells, in the blood-vessels and fibrocytes of the lateral wall, as well as in supporting cells of the organ of Corti. Activation of cas-3 in spiral ganglion cells and the lateral wall was found exclusively in hydropic cochleae. Our findings suggest that oxidative stress is involved in the development of endolymphatic hydrops and may lead to cellular damage which induces apoptosis by activation of cas-3. Apoptotic cell death might contribute to the sensorineural hearing loss found in later stages of Meniere's disease.


Subject(s)
Caspases/metabolism , Cochlea/metabolism , Cochlear Diseases/metabolism , Dinoprost/analogs & derivatives , Endolymphatic Hydrops/metabolism , Oxidative Stress , Tyrosine/analogs & derivatives , Animals , Apoptosis , Audiometry , Caspase 3 , Cochlea/enzymology , Cochlea/physiopathology , Cochlear Diseases/enzymology , Cochlear Diseases/pathology , Cochlear Diseases/physiopathology , Dinoprost/metabolism , Disease Models, Animal , Endolymphatic Hydrops/enzymology , Endolymphatic Hydrops/pathology , Endolymphatic Hydrops/physiopathology , Enzyme Activation , Evoked Potentials, Auditory, Brain Stem , Guinea Pigs , Immunohistochemistry/methods , Meniere Disease/metabolism , Spiral Ganglion/enzymology , Spiral Ganglion/pathology , Staining and Labeling , Tyrosine/metabolism
19.
Acta Otolaryngol Suppl ; (554): 12-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15513504

ABSTRACT

Boettcher cells lie on the basilar membrane beneath Claudius cells. The cells are considered supporting cells for the organ of Corti, and present only in the lower turn of the cochlea, which responds to high-frequency sound. Boettcher cells interdigitate with each other, and project microvilli into the intercellular space. Their structural specialization suggests that Boettcher cells may play a significant role in the function of the cochlea. Nitric oxide synthase (NOS) has previously been detected in substructures of the cochlea. In the cochlea, it is believed that nitric oxide plays an important role in neurotransmission, blood flow regulation, and induction of cytotoxicity under pathological conditions. Findings concerning detection of NOS on Boettcher cells are rare. We demonstrated here the localization of NOS on Boettcher cells of the rat by immunohistochemistry using polyclonal antibody to NOS. On observation with the light microscope using DAB staining, positive immunostaining to NOS was observed in Boettcher cells. In immunoelectron micrographs, NOS was detected abundantly in the cytoplasm of the interdigitations. This suggests that the interdigitations may play significant roles by using NOS. It follows from this that the nitric oxide (NO) on Boettcher cells may influences neighboring Boettcher cells. The ultrastructure of Boettcher cells suggests that they may be active cells, which perform both secretory and absorptive functions.


Subject(s)
Cochlea/cytology , Cochlea/enzymology , Nitric Oxide Synthase/metabolism , Animals , Basilar Membrane/cytology , Basilar Membrane/enzymology , Cochlea/ultrastructure , Free Radical Scavengers/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/enzymology , Male , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/physiology , Nitric Oxide/physiology , Nitric Oxide Synthase/immunology , Organ of Corti/cytology , Organ of Corti/enzymology , Rats , Spiral Ganglion/cytology , Spiral Ganglion/enzymology , Stria Vascularis/cytology , Stria Vascularis/enzymology , Vasoconstriction/physiology
20.
ORL J Otorhinolaryngol Relat Spec ; 66(6): 297-301, 2004.
Article in English | MEDLINE | ID: mdl-15668527

ABSTRACT

Prostaglandins have been used in experimental models and clinical studies for the therapy of sudden hearing loss and tinnitus with conflicting results. However, little is known about the rate-limiting enzymes of prostaglandin synthesis in the inner ear, the generally constitutively expressed cyclooxygenase 1 (COX-1) and the distress-inducible cyclooxygenase 2 (COX-2). To extend our knowledge concerning the physiological expression and localization of these two enzymes, immunohistochemical stainings of the guinea pig cochlea were performed. Light microscopical analysis revealed a homogenous distribution of COX-1 within nearly all cell types of the organ of Corti, but no COX-1 expression in the cuticular plates of pillar cells. COX-2 was found to be expressed in all cell types, with much stronger expression in Hensen cells, neighboring Deiters cells and cuticular plates of outer hair cells. Both COX-1 and COX-2 immunoreactions were also found in the spiral ganglion. We conclude that both COX subtypes are expressed in the guinea pig cochlea under physiological conditions. The prominent expression of the distress-inducible COX-2 isoform in cell types under mechanical stress during noise reception might support the hypothesis of a cytoprotective function of COX products in hearing and in cellular stress situations like intense noise exposure.


Subject(s)
Immunohistochemistry/methods , Organ of Corti/enzymology , Prostaglandin-Endoperoxide Synthases/analysis , Spiral Ganglion/enzymology , Animals , Cyclooxygenase 1 , Cyclooxygenase 2 , Disease Models, Animal , Guinea Pigs , Hearing Loss, Sudden/drug therapy , Hearing Loss, Sudden/enzymology , Organ of Corti/drug effects , Prostaglandins, Synthetic/metabolism , Prostaglandins, Synthetic/therapeutic use , Spiral Ganglion/drug effects , Tinnitus/drug therapy , Tinnitus/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...