Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7: 42216, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181574

ABSTRACT

p53 is a key component of a signaling network that protects cells against various stresses. As excess p53 is detrimental to cells, its levels are tightly controlled by several mechanisms. The E3 ubiquitin ligase Mdm2 is a major negative regulator of p53. The significance of balanced p53 levels in normal tissues, at different stages of lifetime, is poorly understood. We have studied in vivo how the disruption of Mdm2/p53 interaction affects the early-embryonic otic progenitor cells and their descendants, the auditory supporting cells and hair cells. We found that p53 accumulation, as a consequence of Mdm2 abrogation, is lethal to both proliferative progenitors and non-proliferating, differentiating cells. The sensitivity of postmitotic supporting cells to excess p53 decreases along maturation, suggesting that maturation-related mechanisms limit p53's transcriptional activity towards pro-apoptotic factors. We have also investigated in vitro whether p53 restricts supporting cell's regenerative capacity. Unlike in several other regenerative cellular models, p53 inactivation did not alter supporting cell's proliferative quiescence nor transdifferentiation capacity. Altogether, the postmitotic status of developing hair cells and supporting cells does not confer protection against the detrimental effects of p53 upregulation. These findings might be linked to auditory disturbances observed in developmental syndromes with inappropriate p53 upregulation.


Subject(s)
Ear, Inner/embryology , Ear, Inner/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Aging/metabolism , Animals , Animals, Newborn , Apoptosis , Cell Cycle , Cell Survival , Cell Transdifferentiation , Cochlea/embryology , Cochlea/pathology , Integrases/metabolism , Mice , Morphogenesis , Protein Binding , Stem Cells/metabolism , Up-Regulation
2.
Acta Otolaryngol ; 128(10): 1110-5, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18607951

ABSTRACT

CONCLUSION: The average recovery of hearing and cessation of tinnitus was significantly better after hyperbaric oxygen therapy (HBOT) than after normobaric oxygen therapy (NBOT). HBOT can be valuable adjuvant therapy for patients with acute acoustic trauma (AAT). OBJECTIVES: AAT was one of the early indications for the use of HBOT. The rationale of administering oxygen to patients with AAT is based on experimental studies showing that noise exposure results in cochlear hypoxia, which could be compensated by HBOT. The aim of this study was to investigate the efficacy of HBOT in patients with AAT. PATIENTS AND METHODS: We compared the recovery from hearing impairment and tinnitus in 60 ears treated with HBOT with 60 ears treated with NBOT. The HBOT was given daily for 1-8 days. There were no significant differences in clinical or audiological data between HBOT and NBOT groups. RESULTS: The average recovery of hearing both at high and speech frequencies was significantly better and tinnitus persisted less commonly after the HBOT than after the NBOT. Normal hearing at the end of the follow-up period was regained in 42 ears in the HBOT group and in 24 ears in the NBOT group (p<0.01).


Subject(s)
Hearing Loss, Noise-Induced/therapy , Hyperbaric Oxygenation , Recovery of Function , Tinnitus/therapy , Acute Disease , Adolescent , Adult , Audiometry, Pure-Tone , Case-Control Studies , Humans , Male , Retrospective Studies , Young Adult
3.
Am J Hum Genet ; 69(4): 673-84, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11524702

ABSTRACT

Usher syndrome type 3 (USH3) is an autosomal recessive disorder characterized by progressive hearing loss, severe retinal degeneration, and variably present vestibular dysfunction, assigned to 3q21-q25. Here, we report on the positional cloning of the USH3 gene. By haplotype and linkage-disequilibrium analyses in Finnish carriers of a putative founder mutation, the critical region was narrowed to 250 kb, of which we sequenced, assembled, and annotated 207 kb. Two novel genes-NOPAR and UCRP-and one previously identified gene-H963-were excluded as USH3, on the basis of mutational analysis. USH3, the candidate gene that we identified, encodes a 120-amino-acid protein. Fifty-two Finnish patients were homozygous for a termination mutation, Y100X; patients in two Finnish families were compound heterozygous for Y100X and for a missense mutation, M44K, whereas patients in an Italian family were homozygous for a 3-bp deletion leading to an amino acid deletion and substitution. USH3 has two predicted transmembrane domains, and it shows no homology to known genes. As revealed by northern blotting and reverse-transcriptase PCR, it is expressed in many tissues, including the retina.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 3/genetics , Deafness/genetics , Genetic Linkage/genetics , Membrane Proteins/genetics , Mutation/genetics , Retinal Degeneration/genetics , Base Sequence , Cloning, Molecular , Contig Mapping , Expressed Sequence Tags , Female , Finland , Founder Effect , Gene Expression Profiling , Haplotypes/genetics , Humans , Linkage Disequilibrium/genetics , Male , Membrane Proteins/chemistry , Molecular Sequence Data , Pedigree , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Messenger/analysis , RNA, Messenger/genetics , Retina/metabolism , Syndrome
4.
Article in English | MEDLINE | ID: mdl-11054017

ABSTRACT

We have studied the morphological and cellular changes in the cochlear nucleus (CN) after cochlear nerve degeneration and whether these changes can be prevented by rescuing the primary cochlear neurons from degeneration with local glial cell line derived neurotrophic factor (GDNF) treatment. Degeneration of spiral ganglion neurons was seen to lead to a reduction of the volume of the anteroventral cochlear nucleus (AVCN); the size of the cell nuclei in the AVCN also was reduced. No differences were observed in cell density. After intrascalar GDNF treatment the volume of the AVCN was significantly larger when compared to the untreated side, and the size of the cell nuclei in the AVCN was significantly larger on the treated side. After degeneration of spiral ganglion neurons, an increased number of apoptotic cell nuclei were seen in ipsilateral CN and superior olivary complex. This increase was significantly smaller after intrascalar GDNF treatment. Degeneration of primary cochlear neurons seems to lead to an increase in the number of CN neurons undergoing apoptotic cell death. This can be prevented partially by rescuing primary cochlear neurons from degeneration with local GDNF treatment.


Subject(s)
Apoptosis/drug effects , Cochlear Nucleus/injuries , Nerve Growth Factors , Nerve Tissue Proteins/therapeutic use , Neuroprotective Agents/therapeutic use , Organ of Corti/injuries , Animals , Auditory Cortex , Brain Stem/pathology , Cochlear Nucleus/pathology , Glial Cell Line-Derived Neurotrophic Factor , Guinea Pigs , Hearing Loss, Noise-Induced/prevention & control , Humans , Nerve Tissue Proteins/pharmacology , Neurons/pathology , Neuroprotective Agents/pharmacology , Noise/adverse effects , Trauma Severity Indices , Vestibulocochlear Nerve Diseases/prevention & control
5.
J Neurosci ; 20(16): 6125-34, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934262

ABSTRACT

Interactions between FGF10 and the IIIb isoform of FGFR-2 appear to be crucial for the induction and growth of several organs, particularly those that involve budding morphogenesis. We determined their expression patterns in the inner ear and analyzed the inner ear phenotype of mice specifically deleted for the IIIb isoform of FGFR-2. FGF10 and FGFR-2(IIIb) mRNAs showed distinct, largely nonoverlapping expression patterns in the undifferentiated otic epithelium. Subsequently, FGF10 mRNA became confined to the presumptive cochlear and vestibular sensory epithelia and to the neuronal precursors and neurons. FGFR-2(IIIb) mRNA was expressed in the nonsensory epithelium of the otocyst that gives rise to structures such as the endolymphatic and semicircular ducts. These data suggest that in contrast to mesenchymal-epithelial-based FGF10 signaling demonstrated for other organs, the inner ear seems to depend on paracrine signals that operate within the epithelium. Expression of FGF10 mRNA partly overlapped with FGF3 mRNA in the sensory regions, suggesting that they may form parallel signaling pathways within the otic epithelium. In addition, hindbrain-derived FGF3 might regulate otocyst morphogenesis through FGFR-2(IIIb). Targeted deletion of FGFR-2(IIIb) resulted in severe dysgenesis of the cochleovestibular membraneous labyrinth, caused by a failure in morphogenesis at the otocyst stage. In addition to the nonsensory epithelium, sensory patches and the cochleovestibular ganglion remained at a rudimentary stage. Our findings provide genetic evidence that signaling by FGFR-2(IIIb) is critical for the morphological development of the inner ear.


Subject(s)
Ear, Inner/embryology , Fibroblast Growth Factors/genetics , Protein Isoforms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction/physiology , Animals , Cochlea/cytology , Cochlea/embryology , Cochlea/metabolism , Ear, Inner/cytology , Ear, Inner/metabolism , Embryo, Mammalian , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 3 , Mice , Mice, Knockout , Mice, Transgenic , Mutation/physiology , Phenotype , Protein Isoforms/deficiency , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/deficiency , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/deficiency
6.
J Neurosci ; 20(1): 43-50, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10627579

ABSTRACT

We have studied the mechanisms of auditory hair cell death after insults in vitro and in vivo. We show DNA fragmentation of hair cell nuclei after ototoxic drug and intense noise trauma. By using phospho-specific c-Jun-N-terminal kinase (JNK) and c-Jun antibodies in immunohistochemistry, we show that the JNK pathway, associated with stress, injury, and apoptosis, is activated in hair cells after trauma. CEP-1347, a derivative of the indolocarbazole K252a, is a small molecule that has been shown to attenuate neurodegeneration by blocking the activation of JNK (). Subcutaneously delivered CEP-1347 attenuated noise-induced hearing loss. The protective effect was demonstrated by functional tests, which showed less hearing threshold shift in CEP-1347-treated than in nontreated guinea pigs, and by morphometric methods showing less hair cell death in CEP-1347-treated cochleas. In organotypic cochlear cultures, CEP-1347 prevented neomycin-induced hair cell death. In addition to hair cells, CEP-1347 promoted survival of dissociated cochlear neurons. These results suggest that therapeutic intervention in the JNK signaling cascade, possibly by using CEP-1347, may offer opportunities to treat inner ear injuries.


Subject(s)
Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/cytology , Hearing Loss, Noise-Induced/drug therapy , Indoles/pharmacology , Neurons, Afferent/cytology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Aminoglycosides/toxicity , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/physiology , Hearing Loss, Noise-Induced/chemically induced , Hearing Loss, Noise-Induced/pathology , Neomycin/toxicity , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Noise/adverse effects , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Cell Tissue Res ; 295(3): 369-82, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022958

ABSTRACT

Analyses of single and double mutants of members of the neurotrophin family and their receptors are reviewed. These data demonstrate that the two neurotrophins, brain-derived neurotrophic factor (BDNF) and neurotrophin 3 (NT-3), and their high-affinity receptors trkB and trkC, are the sole support for the developing afferent innervation of the ear. Neurotrophins are first expressed in the otocyst around the time afferent sensory neurons become postmitotic. They are crucial for the survival of certain topologically distinct populations of sensory neurons. BDNF supports all sensory neurons to the semicircular canals, most sensory neurons to the saccule and utricle, and many sensory neurons to the apex and middle turn of the cochlea. In contrast, NT-3 supports few sensory neurons to the utricle and saccule, all sensory neurons to the basal turn of the cochlea and most sensory neurons to the middle and apical turn. Some topologically restricted effects reflect the pattern of neurotrophin distribution as revealed by in situ hybridization (e.g., loss of all innervation to the semicircular canal sensory epithelia in BDNF or trkB mutants). However, other topologically restricted effects cannot be explained on the basis of current knowledge of neurotrophin or neurotrophin receptor distribution. Data on mutants also support the notion that BDNF may play a role in neonatal plastic reorganization of the pattern of innervation in the ear and possibly the brainstem. In contrast, data obtained thus far on the ability of neurotrophins to rescue adult sensory neuron after insults to cochlear hair cells are less compelling. The ear is a model system to test the interactions of the two neurotrophins, BDNF and NT-3, with their two high-affinity receptors, trkB and trkC.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Ear/innervation , Nerve Growth Factors/physiology , Adult , Animals , Ear/embryology , Ganglia/physiology , Glial Cell Line-Derived Neurotrophic Factor , Hair Cells, Auditory , Humans , Nerve Tissue Proteins/physiology , Neuronal Plasticity , Neurons, Afferent/physiology , Neurotrophin 3
8.
Nature ; 397(6716): 251-5, 1999 Jan 21.
Article in English | MEDLINE | ID: mdl-9930699

ABSTRACT

GABA (gamma-aminobutyric acid) is the main inhibitory transmitter in the adult brain, and it exerts its fast hyperpolarizing effect through activation of anion (predominantly Cl-)-permeant GABA(A) receptors. However, during early neuronal development, GABA(A)-receptor-mediated responses are often depolarizing, which may be a key factor in the control of several Ca2+-dependent developmental phenomena, including neuronal proliferation, migration and targeting. To date, however, the molecular mechanism underlying this shift in neuronal electrophysiological phenotype is unknown. Here we show that, in pyramidal neurons of the rat hippocampus, the ontogenetic change in GABA(A)-mediated responses from depolarizing to hyperpolarizing is coupled to a developmental induction of the expression of the neuronal (Cl-)-extruding K+/Cl- co-transporter, KCC2. Antisense oligonucleotide inhibition of KCC2 expression produces a marked positive shift in the reversal potential of GABAA responses in functionally mature hippocampal pyramidal neurons. These data support the conclusion that KCC2 is the main Cl- extruder to promote fast hyperpolarizing postsynaptic inhibition in the brain.


Subject(s)
Carrier Proteins/physiology , Pyramidal Cells/physiology , Symporters , gamma-Aminobutyric Acid/physiology , Animals , Blotting, Southern , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Differentiation , Chlorides/metabolism , Electrophysiology , GABA Agonists/pharmacology , Gene Expression Regulation, Developmental , Guinea Pigs , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/metabolism , In Vitro Techniques , Muscimol/pharmacology , Potassium/metabolism , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , K Cl- Cotransporters
9.
Ann N Y Acad Sci ; 884: 292-304, 1999 Nov 28.
Article in English | MEDLINE | ID: mdl-10842601

ABSTRACT

Many of the neurotrophic factors promote the survival of developing peripheral sensory neurons, and they might be useful as therapeutic agents in the adult neuronal systems. During development, neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF) mRNAs are expressed in the auditory sensory epithelium, which composes the peripheral target field of the cochlear (spiral) neurons. NT-3 mRNA is also expressed in the inner hair cells of the mature organ of Corti. mRNAs encoding their signal-transducing receptors, TrkC and TrkB, respectively, are expressed in the cochlear neurons. In addition to neurotrophins, which seem to have an important role during development, another neurotrophic factor, glial cell-line-derived neurotrophic factor (GDNF), seems to be involved in the maintenance of postnatal auditory neurons. In the present work, the cellular distribution of neurotrophins and GDNF in the developing and adult rat inner ear are compared. The effects of recombinant neurotrophins and GDNF on dissociated cochlear neurons in vitro are also compared. Recently, NT-3 and BDNF were used in vivo as therapeutic agents to protect guinea pig cochlear neurons from aminoglycoside-induced degeneration and GDNF from noise-induced degeneration. These data demonstrate that NT-3, BDNF, and GDNF might be potential candidates for prevention of degeneration of the auditory nerve in man.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cochlea/metabolism , Nerve Growth Factors , Nerve Tissue Proteins/metabolism , Neurotrophin 3/metabolism , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cochlea/drug effects , Cochlea/embryology , Glial Cell Line-Derived Neurotrophic Factor , Nerve Tissue Proteins/pharmacology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neurotrophin 3/pharmacology , Rats , Rats, Sprague-Dawley
10.
Ann N Y Acad Sci ; 884: 305-11, 1999 Nov 28.
Article in English | MEDLINE | ID: mdl-10842602

ABSTRACT

Trauma and loss of cochlear inner hair cells causes a series of events that result first in the retraction of the peripheral processes of the auditory nerve, scar formation in the organ of Corti, and over the course of weeks to months (depending on the species) the loss of auditory nerve cell bodies (spiral ganglion cells). Neurotrophic factors play an important role in the mature nervous system as survival factors for maintenance and protection and also can play a role in regrowth. Studies in the cochlea now show that application of exogenous neurotrophic factors can enhance survival of spiral ganglion cells after deafness and induce regrowth of peripheral processes, perhaps by replacing lost endogenous factors. Combinations of factors may be most effective for achieving greatest survival and regrowth. Our studies find that brain-derived neurotrophic factor (BDNF) and glial-line-derived neurotrophic factor (GDNF) are very effective at enhancing spiral ganglion cell survival following deafness from ototoxic drugs or noise. It has also been found that BDNF plus fibroblast growth factor (FGF) is very effective at inducing process regrowth. Electrical stimulation also acts to enhance spiral ganglion cell survival, and the combination of electrical stimulation and neurotrophic factors could prove a most effective intervention.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/pharmacology , Spiral Ganglion/drug effects , Aminoglycosides , Animals , Anti-Bacterial Agents/adverse effects , Brain-Derived Neurotrophic Factor/therapeutic use , Deafness/chemically induced , Deafness/drug therapy , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Glial Cell Line-Derived Neurotrophic Factor , Nerve Growth Factors/therapeutic use , Nerve Tissue Proteins/therapeutic use , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Spiral Ganglion/physiology
11.
Hear Res ; 124(1-2): 17-26, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9822899

ABSTRACT

For patients with profound hearing loss, cochlear implants have become the treatment of choice. These devices provide auditory information through direct electrical stimulation of the auditory nerve. Prosthesis function depends on survival and electrical excitability of the cochlear neurons. Degeneration of the auditory nerve occurs after lesions of its peripheral target field (organ of Corti), specifically, including loss of inner hair cells (IHCs). There is now evidence that local treatment of the cochlea with neurotrophins may enhance survival of auditory neurons after aminoglycoside-induced deafness. Glial cell line-derived neurotrophic factor (GDNF) has recently been shown to be an important survival factor in other regions of the nervous system. By in situ hybridization, we now show that IHCs of the neonatal and mature rat cochlea synthesize GDNF and that GDNF-receptor alpha, but not c-Ret, is expressed in the rat spiral ganglion. We also show that GDNF is a potent survival-promoting factor for rat cochlear neurons in vitro. Finally, we examined GDNF efficacy to enhance cochlear-nerve survival after IHC lesions in vivo. We found that chronic intracochlear infusion of GDNF greatly enhances survival of guinea pig cochlear neurons after noise-induced IHC lesions. Our results demonstrate that GDNF is likely to be an endogeneous survival factor in the normal mammalian cochlea and it could have application as a pharmacological treatment to prevent secondary auditory nerve degeneration following organ of Corti damage.


Subject(s)
Drosophila Proteins , Glial Fibrillary Acidic Protein/physiology , Nerve Degeneration/physiopathology , Neurons, Afferent/physiology , Noise , Vestibulocochlear Nerve Injuries , Vestibulocochlear Nerve/physiopathology , Animals , Cell Survival , Glial Cell Line-Derived Neurotrophic Factor Receptors , Glial Fibrillary Acidic Protein/genetics , In Situ Hybridization , Nerve Degeneration/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Vestibulocochlear Nerve/pathology
12.
J Neurobiol ; 33(7): 1019-33, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9407020

ABSTRACT

We studied the expression of neurotrophins and their Trk receptors in the chicken cochlea. Based on in situ hybridization, brain-derived neurotrophic factor (BDNF) is the major neurotrophin there, in contrast to the mammalian cochlea, where neurotrophin-3 (NT-3) predominates. NT-3 mRNA labeling was weak and found only during a short time period in the early cochleas. During embryogenesis, BDNF mRNA was first seen in early differentiating hair cells. Afferent cochlear neurons expressed trkB mRNA from the early stages of gangliogenesis onward. In accordance, in vitro, BDNF promoted survival of dissociated neurons and stimulated neuritogenesis from ganglionic explants. High levels of BDNF mRNA in hair cells and trkB mRNA in cochlear neurons persisted in the mature cochlea. In addition, mRNA for the truncated TrkB receptor was expressed in nonneuronal cells, specifically in supporting cells, located adjacent to the site of BDNF synthesis and nerve endings. Following acoustic trauma, regenerated hair cells acquired BDNF mRNA expression at early stages of differentiation. Truncated trkB mRNA was lost from supporting cells that regenerated into hair cells. High levels of BDNF mRNA persisted in surviving hair cells and trkB mRNA in cochlear neurons after noise exposure. These results suggest that in the avian cochlea, peripheral target-derived BDNF contributes to the onset and maintenance of hearing function by supporting neuronal survival and regulating the (re)innervation process. Truncated TrkB receptors may regulate the BDNF concentration available to neurites, and they might have an important role during reinnervation.


Subject(s)
Cochlea/metabolism , Nerve Growth Factors/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Regeneration/physiology , Animals , Basilar Membrane/metabolism , Brain-Derived Neurotrophic Factor/biosynthesis , Chick Embryo , Chickens , Cochlea/embryology , Cochlea/growth & development , Neurites/physiology , Neurons/cytology , RNA, Messenger/biosynthesis , Receptor, Ciliary Neurotrophic Factor , Receptor, trkC
13.
Acta Otolaryngol Suppl ; 529: 59-62, 1997.
Article in English | MEDLINE | ID: mdl-9288269

ABSTRACT

In audiological "toughening" or "conditioning" phenomenon prior exposure to moderate noise reduces the extent of hearing deterioration caused by the subsequent exposure to traumatic test noise known to cause inner ear damage. "Toughening" has been demonstrated in many mammalian laboratory animals such as guinea pig and chinchilla but not in rat or mouse. Our aim was to study the occurrence of this phenomenon in the rat. Ninety-one white male Wistar rats were divided into four groups: unexposed control group (U, n = 10), "conditioning" only (C, n = 32), "conditioning" plus test noise (C + T, n = 36) and test noise only (T, n = 13). Groups C and C + T were "conditioned" for 10 hours with 4.0 kHz OBN between 55 and 95 dB sound pressure levels (SPLs). After 10 hours rest groups C + T and T were exposed to the same noise at 105 dB SPL for 13 hours. The hearing thresholds were determined by auditory brainstem response audiometry (ABR) either immediately after or 3 weeks after the exposures. After that the animals were sacrificed. The cochleas were removed and perilymphatically fixed and further processed for quantitative cytocochleograms. Both the temporary (TTS) and the permanent threshold shifts (PTS) were smaller in animals which had been "conditioned" prior exposure to traumatic noise. Yet only 95 dB SPL "conditioning" gave statistically significant difference (p < 0.05) in PTS. From our results we conclude that "conditioning" effect seems to be present also in the rat. However to confirm this, further experiments are needed. The mechanisms behind "conditioning" are still unknown and also to clarify them, further efforts are needed.


Subject(s)
Auditory Threshold/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Noise-Induced/prevention & control , Acoustic Stimulation/methods , Animals , Audiometry, Evoked Response , Hearing Loss, Noise-Induced/physiopathology , Male , Rats , Rats, Wistar , Time Factors
14.
Proc Natl Acad Sci U S A ; 92(20): 9269-73, 1995 Sep 26.
Article in English | MEDLINE | ID: mdl-7568115

ABSTRACT

Here we show that the mature cochlear neurons are a rich source of acidic fibroblast growth factor (aFGF), which is expressed in the neuronal circuitry consisting of afferent and efferent innervation. The site of action of neuronal aFGF is likely to reside in the organ of Corti, where one of the four known FGF receptor (FGFR) tyrosine kinases--namely, FGFR-3 mRNA--is expressed. Following acoustic overstimulation, known to cause damage to the organ of Corti, a rapid up-regulation of FGFR-3 is evident in this sensory epithelium, at both mRNA and protein levels. The present results provide in vivo evidence for aFGF being a sensory neuron-derived, anterogradely transported factor that may exert trophic effects on a peripheral target tissue. In this sensory system, aFGF, rather than being a neurotrophic factor, seems to promote maintenance of the integrity of the organ of Corti. In addition, aFGF, released from the traumatized nerve endings, may be one of the first signals initiating protective recovery and repair processes following damaging auditory stimuli.


Subject(s)
Fibroblast Growth Factor 1/physiology , Gene Expression , Neurons/physiology , Organ of Corti/physiology , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/physiology , Animals , Antibodies , Fibroblast Growth Factor 1/analysis , Fibroblast Growth Factor 1/immunology , Humans , Immunohistochemistry , In Situ Hybridization , Organ of Corti/cytology , RNA Probes , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/biosynthesis , Recombinant Proteins/immunology , Up-Regulation
15.
Eur J Neurosci ; 7(5): 863-74, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7542125

ABSTRACT

Employing complementary technical approaches, we have studied the expression of acidic fibroblast growth factor (aFGF) and FGF receptors in rat dorsal root ganglia. The results clearly showed that within spinal nerves aFGF and two high-affinity FGF receptors, FGFR-1 and FGFR-2, were prominently expressed in neurons, while expression in Schwann cells was undetectable. FGFR-3 and FGFR-4 were not expressed in dorsal root ganglia. Acidic FGF mRNA was detected in the majority of dorsal root ganglion neurons, including all size classes: FGFR-1 and FGFR-2 transcripts were only detected in subpopulations of mainly large and medium size neurons. In subcellular fractionation studies on dorsal root ganglion and spinal root tissue, aFGF was recovered in the soluble fraction and was thus not tightly associated with neuronal membranes. During development FGFR-1 and FGFR-2 mRNAs were found to be present at all stages examined (embryonic days 15-21 and postnatal days 1-120). Acidic FGF mRNA and protein were first detected at embryonic day 18, and their expression then increased progressively up to postnatal levels. In cultures of dorsal root ganglion neurons derived from day 15 embryos, aFGF expression was first detected 3 days after plating. The resulting neuron cultures continued to express aFGF in a Schwann cell-independent manner. In combination, these results indicate that aFGF expression in dorsal root ganglia is initiated and maintained in postmitotic neurons. Furthermore, the data suggest that the physiological function of aFGF in the peripheral nervous system is connected to processes specific to the mature sensory (and motor) system, such as the maintenance and survival of peripheral nerve neurons.


Subject(s)
Fibroblast Growth Factor 1/biosynthesis , Ganglia, Spinal/metabolism , Neurons/metabolism , Receptors, Fibroblast Growth Factor/biosynthesis , Animals , Antibodies , Blotting, Northern , Blotting, Western , Cells, Cultured , Fibroblast Growth Factor 1/genetics , Gene Expression , Immunohistochemistry , In Situ Hybridization , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Fibroblast Growth Factor/genetics
16.
J Comp Neurol ; 349(4): 596-602, 1994 Nov 22.
Article in English | MEDLINE | ID: mdl-7860790

ABSTRACT

To understand the possible role of retinoic acid during inner ear development and cellular regeneration, we have examined the expression pattern of two intracellular retinoid-binding proteins, the cellular retinol- and retinoic acid-binding proteins of type I in the developing and mature rat inner ear. Expression of cellular retinol-binding protein type I was seen in the supporting cells of the organ of Corti and vestibular organs as soon as the first signs of differentiation of the adjacent hair cells were seen. In the developing organ of Corti, the expression pattern followed the basal-to-apical coil differentiation gradient. After the 1st postnatal week, detectable expression of cellular retinol-binding protein type I disappeared from the organ of Corti, but persisted in the supporting cells of vestibular organs throughout life. Expression of cellular retinoic acid-binding protein type I was not found in the inner ear sensory epithelia. Cellular retinol-binding protein type I has previously been shown to act as a substrate carrier in the synthesis of retinoic acid from its precursor, retinol. Our data suggest that retinoic acid is synthesized in the developing sensory epithelium of the cochlear and vestibular organs and that a concentration gradient formed by retinoic acid may have a role in differentiation of the cochlear sensory epithelium. Furthermore, retinoic acid may have a role in damage-induced hair cell regeneration in the developing and mature vestibular organs as well as in the developing auditory organ. The absence of cellular retinol-binding protein type I from the supporting cells of the mature organ of Corti may be associated with the inability of this organ to regenerate hair cells after damage.


Subject(s)
Cochlea/embryology , Cochlea/metabolism , Fetus/metabolism , Retinol-Binding Proteins/metabolism , Sensation/physiology , Vestibule, Labyrinth/metabolism , Aging/metabolism , Animals , Animals, Newborn , Blotting, Western , Cochlea/cytology , Embryonic and Fetal Development , Epithelium/metabolism , Fetus/physiology , Hair Cells, Auditory/physiology , Immunohistochemistry , Nerve Regeneration , Rats , Receptors, Retinoic Acid/metabolism , Retinol-Binding Proteins, Cellular , Vestibule, Labyrinth/cytology
17.
Hear Res ; 75(1-2): 131-44, 1994 May.
Article in English | MEDLINE | ID: mdl-8071140

ABSTRACT

We show that trkB and trkC mRNAs, encoding the high-affinity receptor tyrosine kinases for brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), respectively, as well as low-affinity nerve growth factor receptor (p75LNGFR) mRNA are expressed in the cochleovestibular ganglion (CVG) before and during innervation of the target fields. Correspondingly, from preinnervation stages onward, BDNF and NT-3, but neither nerve growth factor (NGF) nor neurotrophin-4 (NT-4) mRNAs are expressed in the sensory epithelium of the otic vesicle, the peripheral target field of CVG neurons. No neurotrophin transcripts were detected by in situ hybridization in the medullary central targets. In explant cultures, neuritogenesis from both the cochlear and vestibular part of the CVG was promoted by BDNF, while NT-3 evoked neurites mainly from the cochlear neurons. Also NT-4 stimulated neurite outgrowth from the CVG in vitro. In dissociated neuron-enriched cultures, NT-3 and BDNF promoted survival of overlapping subsets of CVG neurons and, correspondingly, results from in situ hybridization showed that both trkC and trkB mRNAs were expressed in most neurons of this ganglion. The negligible effect of NGF seen in the bioassays agrees well with the expression of only a few trkA transcripts, encoding the high-affinity receptor for NGF, in the CVG. Based on the spatiotemporal expression patterns and biological effects in vitro, peripherally-synthesized BDNF and NT-3 regulate the survival of CVG neurons as well as the establishment of neuron-target cell contacts in the early-developing inner ear. In addition, the expression of trkB mRNA, more specifically its truncated form, and trkC as well as p75LNGFR mRNAs in distinct non-neuronal structures indicates novel roles for these molecules during development.


Subject(s)
Ear, Inner/innervation , Ganglia/metabolism , Nerve Growth Factors/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Animals , Brain-Derived Neurotrophic Factor , Cell Division , Cell Survival , Cells, Cultured , Cochlear Nerve/embryology , Cochlear Nerve/metabolism , Culture Techniques , Ear, Inner/embryology , Ear, Inner/metabolism , Ganglia/embryology , Hair Cells, Auditory, Outer/cytology , Hair Cells, Auditory, Outer/physiology , Mice , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Nerve Tissue Proteins/physiology , Neurons/cytology , Neurotrophin 3 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor/physiology , Vestibular Nerve/embryology , Vestibular Nerve/metabolism
18.
J Cell Biol ; 122(5): 1053-65, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8354693

ABSTRACT

We examined the expression of the neurotrophins (NTFs) and their receptor mRNAs in the rat trigeminal ganglion and the first branchial arch before and at the time of maxillary nerve growth. The maxillary nerve appears first at embryonic day (E)10 and reaches the epithelium of the first branchial arch at E12, as revealed by anti-L1 immunohistochemistry. In situ hybridization demonstrates, that at E10-E11, neurotrophin-3 (NT-3) mRNA is expressed mainly in the mesenchyme, but neurotrophin-4 (NT-4) mRNA in the epithelium of the first branchial arch. NGF and brain-derived neurotrophic factor (BDNF) mRNAs start to be expressed in the distal part of the first brachial arch shortly before its innervation by the maxillary nerve. Trigeminal ganglia strongly express the mRNA of trkA at E10 and thereafter. The expression of mRNAs for low-affinity neurotrophin receptor (LANR), trkB, and trkC in trigeminal ganglia is weak at E10, but increases by E11-E12. NT-3, NT-4, and more prominently BDNF, induce neurite outgrowth from explant cultures of the E10 trigeminal ganglia but no neurites are induced by NGF, despite the expression of trkA. By E12, the neuritogenic potency of NGF also appears. The expression of NT-3 and NT-4 and their receptors in the trigeminal system prior to target field innervation suggests that these NTFs have also other functions than being the target-derived trophic factors.


Subject(s)
Maxillary Nerve/growth & development , Nerve Growth Factors/analysis , Receptors, Nerve Growth Factor/analysis , Trigeminal Ganglion/chemistry , Animals , Brain-Derived Neurotrophic Factor , Culture Techniques , Embryo, Mammalian/chemistry , Embryo, Mammalian/innervation , Embryonic and Fetal Development , Female , In Situ Hybridization , Male , Maxillary Nerve/chemistry , Maxillary Nerve/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurotrophin 3 , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/physiology , Trigeminal Ganglion/physiology , Trigeminal Ganglion/ultrastructure
19.
Hear Res ; 65(1-2): 69-78, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8080462

ABSTRACT

In situ hybridization was used to study the expression of mRNAs of nerve growth factor (NGF), brain-derived neutrophic factor (BDNF), neurotrophin-3 (NT-3), neurotrophin-5 (NT-5) and the components of their high-affinity receptors in the early postnatal and adult rat inner ears. NGF or NT-5 transcripts were not detected in the inner ear neuroepithelium or in the innervating neurons. NT-3 mRNA was intensely expressed over the one-week-old and adult inner hair cells (IHCs) but in the outer hair cells (OHCs) and vestibular maculae only during the early postnatal period. BDNF mRNA was expressed in the IHCs and OHCs of the early postnatal cochlea but not in the adult organ of Corti. High levels of BDNF transcripts were observed in the sensory epithelia of all vestibular end organs. mRNAs of low affinity NGF receptor, trkB and trkC, but not of trk, were expressed in the spiral and vestibular ganglia. In addition, the non-catalytic form of trkB mRNA localized to the sensory epithelia of maculae utriculi and sacculi. The present results show that of the neurotrophins examined, NT-3 is the predominant neurotrophin in the adult organ of Corti and BDNF is that in vestibular organs. The expression patterns of NT-3 and BDNF mRNAs suggest that these neurotrophins may participate in the maintenance of mature cochleovestibular neurons and they may be involved in the survival response of injured neurons.


Subject(s)
Cochlea/metabolism , Ear, Inner/metabolism , Nerve Growth Factors/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Cell Surface/biosynthesis , Animals , Brain-Derived Neurotrophic Factor , Hair Cells, Auditory/metabolism , In Situ Hybridization , Nerve Growth Factors/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurotrophin 3 , Organ of Corti/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics
20.
Acta Otolaryngol Suppl ; 503: 121-6, 1993.
Article in English | MEDLINE | ID: mdl-8470477

ABSTRACT

Neurons of the spiral and vestibular ganglia of the adult rat were labelled with a panel of monoclonal and polyclonal antibodies which were raised against different subunits of neurofilament proteins (NFPs). Thirteen antibodies labelled intensely the perikarya of a distinct group of somata in both the spiral and vestibular ganglia while the majority of somata were unreactive or showed a weaker reaction. The distinction between intensely immunostained and other cell somata was more conspicuous in the spiral than in the vestibular ganglia. Intensely stained somata formed a subpopulation comprising about 9% (range from 7.2 to 11.1%) in the spiral and about 32% (range from 21.2 to 36.4%) in the vestibular ganglia of the total ganglion cell population. Antibodies against different subunits of NFPs seemed always to stain the same somata. In morphometric analysis the mean diameters of intensely labelled spiral ganglion cells were clearly smaller (9.9 microns) than those of the slightly reactive cells (11.9 microns). In Scarpa's ganglia the intensely reactive cells were larger in size (mean diameter 21.4 microns) than the slightly reactive cells (mean diameter 14.7 microns). In the spiral ganglia, the intensely labelled group of neurons seems to correspond to the morphologically distinct type II cells which may also functionally differ from type I cells. In Scarpa's ganglia, the intensely stained subgroup of somata may correspond to the large neurons innervating central regions of cristae and maculae. Their functional significance remains to be elucidated.


Subject(s)
Neurofilament Proteins/analysis , Spiral Ganglion/anatomy & histology , Vestibular Nerve/anatomy & histology , Animals , Fluorescent Antibody Technique , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...