Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Hear Res ; 345: 43-51, 2017 03.
Article in English | MEDLINE | ID: mdl-28034618

ABSTRACT

Our previous studies have shown that the stimulation of A1 adenosine receptors in the inner ear can mitigate the loss of sensory hair cells and hearing loss caused by exposure to traumatic noise. Here, we focus on the role of adenosine receptors (AR) in the development of noise-induced neural injury in the cochlea using A1AR and A2AAR null mice (A1AR-/- and A2AAR-/-). Wildtype (WT) and AR deficient mice were exposed to octave band noise (8-16 kHz, 100 dB SPL) for 2 h to induce cochlear injury and hearing loss. Auditory thresholds and input/output functions were assessed using auditory brainstem responses (ABR) before and two weeks post-exposure. The loss of outer hair cells (OHC), afferent synapses and spiral ganglion neurons (SGN) were assessed by quantitative histology. A1AR-/- mice (6-8 weeks old) displayed a high frequency hearing loss (ABR threshold shift and reduced ABR wave I and II amplitudes). This hearing loss was further aggravated by acute noise exposure and exceeded the hearing loss in the WT and A2AAR-/- mice. All mice experienced the loss of OHC, synaptic ribbons and SGN after noise exposure, but the loss of SGN was significantly higher in A1AR-/- mice than in the A2AAR-/- and WT genotypes. The A2AAR-/- demonstrated better preservation of OHC and afferent synapses and the minimal loss of SGN after noise exposure. The findings suggest that the loss of A1AR expression results in an increased susceptibility to cochlear neural injury and hearing loss, whilst absence of A2AAR increases cochlear resistance to acoustic trauma.


Subject(s)
Cochlea/metabolism , Hearing Loss, Noise-Induced/metabolism , Hearing , Noise/adverse effects , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Auditory Threshold , Cochlea/injuries , Cochlea/pathology , Cochlea/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Genetic Predisposition to Disease , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Noise-Induced/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protective Factors , Receptor, Adenosine A1/deficiency , Receptor, Adenosine A1/genetics , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Risk Factors , Spiral Ganglion/injuries , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Synapses/metabolism , Synapses/pathology , Time Factors
2.
Acta Otolaryngol ; 136(11): 1115-1120, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27228317

ABSTRACT

CONCLUSIONS: Transplantation of OECs into the cochlea may protect and increase the survival of SGCs. OBJECTIVE: To investigate the protective effect of the transplantation of olfactory ensheathing cells (OECs) on injured spiral ganglion cells (SGCs) in rats. METHODS: OECs were transplanted into the cochlea in rats with SGCs that were injured by kanamycin sulfate (KM). An equal volume of D-Hanks was injected into the cochlea of control rats. Auditory brainstem responses (ABRs) were recorded from the rats in both groups to monitor changes in hearing thresholds. Immunofluorescence was employed to examine the density and morphology of SGCs to assess the ototoxic condition of the cochlea. RESULTS: There was no significant difference in the ABR threshold at each frequency between the control and experimental groups. Notably, in the experimental group, a number of Hoechst 3334-labeled nuclei were detected from the apex to the basal turn of the cochlea, demonstrating that the OECs were successfully transplanted and survived in the cochlea. In the experimental group, most of the SGCs were tightly arranged, and the nuclear membrane, chromatin, and nucleolus were all clear. The SGCs in the control group were loosely arranged, and only a few normal SGCs were observed in this group.


Subject(s)
Hearing Loss/therapy , Neuroglia/transplantation , Spiral Ganglion/injuries , Animals , Auditory Threshold , Hearing Loss/pathology , Random Allocation , Rats, Sprague-Dawley , Spiral Ganglion/pathology
3.
Neurosci Lett ; 578: 111-6, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24993301

ABSTRACT

Spiral ganglion neuron (SGN) injury is a generally accepted precursor of auditory neuropathy. Receptor-interacting protein 3 (RIP3) has been reported as an important necroptosis pathway mediator that can be blocked by necrostatin-1 (Nec-1). In our study, we sought to identify whether necroptosis participated in SGN injury. Ouabain was applied to establish an SGN injury model. We measured the auditory brain-stem response (ABR) threshold shift as an indicator of the auditory conditions. Positive ß3-tubulin immunofluorescence staining indicated the surviving SGNs. RIP3 expression was evaluated using immunofluorescence, quantitative real-time polymerase chain reaction and western blot. SGN injury promoted an increase in RIP3 expression that could be suppressed by application of the necroptosis inhibitor Nec-1. A decreased ABR threshold shift and increased SGN density were observed when Nec-1 was administered with apoptosis inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD). These results demonstrated that necroptosis is an indispensable pathway separately from apoptosis leading to SGN death pathway, in which RIP3 plays an important role.


Subject(s)
Neurons/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Spiral Ganglion/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Death/physiology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Imidazoles/pharmacology , Indoles/pharmacology , Neurons/drug effects , Oligopeptides/pharmacology , Ouabain/toxicity , Rats , Rats, Sprague-Dawley , Spiral Ganglion/drug effects , Spiral Ganglion/injuries
4.
Stem Cells Dev ; 23(5): 502-14, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24172073

ABSTRACT

A loss of sensory hair cells or spiral ganglion neurons from the inner ear causes deafness, affecting millions of people. Currently, there is no effective therapy to repair the inner ear sensory structures in humans. Cochlear implantation can restore input, but only if auditory neurons remain intact. Efforts to develop stem cell-based treatments for deafness have demonstrated progress, most notably utilizing embryonic-derived cells. In an effort to bypass limitations of embryonic or induced pluripotent stem cells that may impede the translation to clinical applications, we sought to utilize an alternative cell source. Here, we show that adult human mesenchymal-like stem cells (MSCs) obtained from nasal tissue can repair spiral ganglion loss in experimentally lesioned cochlear cultures from neonatal rats. Stem cells engraft into gentamicin-lesioned organotypic cultures and orchestrate the restoration of the spiral ganglion neuronal population, involving both direct neuronal differentiation and secondary effects on endogenous cells. As a physiologic assay, nasal MSC-derived cells engrafted into lesioned spiral ganglia demonstrate responses to infrared laser stimulus that are consistent with those typical of excitable cells. The addition of a pharmacologic activator of the canonical Wnt/ß-catenin pathway concurrent with stem cell treatment promoted robust neuronal differentiation. The availability of an effective adult autologous cell source for inner ear tissue repair should contribute to efforts to translate cell-based strategies to the clinic.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Nerve Regeneration , Adult , Animals , Cochlea/growth & development , Cochlea/injuries , Cochlea/pathology , Ear, Inner/growth & development , Ear, Inner/pathology , Humans , Neurons/pathology , Rats , Spiral Ganglion/growth & development , Spiral Ganglion/injuries , Spiral Ganglion/pathology , Wnt Signaling Pathway/genetics
5.
PLoS One ; 8(7): e67618, 2013.
Article in English | MEDLINE | ID: mdl-23840874

ABSTRACT

Given the frequent use of improvised explosive devices (IEDs) around the world, the study of traumatic blast injuries is of increasing interest. The ear is the most common organ affected by blast injury because it is the body's most sensitive pressure transducer. We fabricated a blast chamber to re-create blast profiles similar to that of IEDs and used it to develop a reproducible mouse model to study blast-induced hearing loss. The tympanic membrane was perforated in all mice after blast exposure and found to heal spontaneously. Micro-computed tomography demonstrated no evidence for middle ear or otic capsule injuries; however, the healed tympanic membrane was thickened. Auditory brainstem response and distortion product otoacoustic emission threshold shifts were found to be correlated with blast intensity. As well, these threshold shifts were larger than those found in control mice that underwent surgical perforation of their tympanic membranes, indicating cochlear trauma. Histological studies one week and three months after the blast demonstrated no disruption or damage to the intra-cochlear membranes. However, there was loss of outer hair cells (OHCs) within the basal turn of the cochlea and decreased spiral ganglion neurons (SGNs) and afferent nerve synapses. Using our mouse model that recapitulates human IED exposure, our results identify that the mechanisms underlying blast-induced hearing loss does not include gross membranous rupture as is commonly believed. Instead, there is both OHC and SGN loss that produce auditory dysfunction.


Subject(s)
Blast Injuries/complications , Ear/injuries , Ear/pathology , Hearing Loss/etiology , Hearing Loss/pathology , Animals , Cochlea/injuries , Cochlea/pathology , Female , Mice , Spiral Ganglion/injuries , Spiral Ganglion/pathology , Tympanic Membrane/injuries , Tympanic Membrane/pathology
6.
Neurosci Lett ; 534: 101-6, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23219799

ABSTRACT

Neural stem cell (NSC) transplantation into the cochlea is widely used for the treatment of spiral ganglion neuron (SGN) degenerative disease and injury in the animal models, but the migration of the transplanted NSCs to the injury region is difficult and the mechanism is still unclear. In this study, we aimed to validate whether the SGN-degenerated cochlear microenvironment plays a role in the NSC migration and investigated whether stromal cell-derived factor-1 (SDF-1) was involved in the NSCs migration. Using a rat SGN degeneration model, we demonstrated that the transplanted NSCs are more likely to migrate to the injury region during the early post-injury (EPI) than the late post-injury (LPI) stage and the control cochlea. We found that the expressions of SDF-1 increased transiently after SGN degeneration. Additionally, we showed that the NSCs express CXCR4, a receptor for SDF-1. We observed that the region to which the transplanted NSC localized coincides with the region where the SDF-1 is highly expressed following the degeneration of SGNs. Finally, we observed that the increased SDF-1 is derived from the Schwann cells in the SGN-degenerated model. These results suggest that SDF-1, which is derived from cochlear Schwann cells and up-regulated in the early injury microenvironment, plays a beneficial role in the NSC migration to the injury region. Optimizing SDF-1 expression in the host microenvironment or increasing the CXCR4 expression of the donor stem cells may improve the migration efficiency of transplanted cells toward the injury region in the cochlea.


Subject(s)
Chemokine CXCL12/metabolism , Nerve Degeneration/pathology , Neural Stem Cells/cytology , Neurons/pathology , Spiral Ganglion/pathology , Animals , Cell Movement , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/metabolism , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Olfactory Bulb/cytology , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism , Schwann Cells/metabolism , Spiral Ganglion/injuries , Spiral Ganglion/metabolism
7.
Am J Otolaryngol ; 32(1): 8-12, 2011.
Article in English | MEDLINE | ID: mdl-20022668

ABSTRACT

OBJECTIVE: To study whether adenovirus-mediated human ß-nerve growth factor (Ad-hNGFß) gene has any protective effect on rat cochlear spiral ganglion after blast exposure. METHODS: Deafness was induced by blast exposure (172.0 dB) in 20 healthy rats. Seven days after blast exposure, Ad-hNGFß was infused into the perilymphatic space of 10 animals as the hNGFß/blast group, and artificial perilymph fluid (APF) was infused into the perilymphatic space of 10 animals as the APF/blast control group. An additional control group consisted of 10 healthy rats which received Ad-hNGFß target gene with no blast exposure (hNGFß/control group). Auditory functions were monitored by thresholds of auditory brain stem responses (ABR). At weeks 1, 4, and 8 postoperatively, the animals were killed, and the cochleae were removed for immunohistochemical, hematoxylin and eosin staining study. RESULTS: The ABR threshold shifts in the hNGFß/blast group were significantly smaller than that of APF/blast control group. There were no significant differences of the ABR values between before and after operation in the hNGFß/control group. Expression of Ad-hNGFß protein was detected in each turn of the cochlea in the first week, with almost equal intensity in all turns. In the fourth week, the reactive intensity decreased. In the eighth week, no reaction was detectable. The results of hematoxylin and eosin stain showed that the number of spiral ganglions in the hNGFß/blast group was significantly greater than that of the APF/blast control group in the 4th week (P < .01). CONCLUSION: Adenovirus-mediated human ß-nerve growth factor can be expressed at a high level and for a relatively long period in the blast impaired cochlea, suggesting that Ad-hNGFß has a protective effect on rat cochlear spiral ganglion cells after blast exposure.


Subject(s)
Blast Injuries , Deafness/prevention & control , Nerve Growth Factor/pharmacology , Spiral Ganglion/injuries , Adenoviridae , Animals , Deafness/etiology , Evoked Potentials, Auditory , Gene Transfer Techniques , Genetic Therapy , Humans , Immunohistochemistry , Nerve Growth Factor/administration & dosage , Perilymph , Random Allocation , Rats
8.
J Assoc Res Otolaryngol ; 10(1): 111-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18958527

ABSTRACT

Lesions of spiral ganglion cells, representing a restricted sector of the auditory nerve array, produce immediate changes in the frequency tuning of inferior colliculus (IC) neurons. There is a loss of excitation at the lesion frequencies, yet responses to adjacent frequencies remain intact and new regions of activity appear. This leads to immediate changes in tuning and in tonotopic progression. Similar effects are seen after different methods of peripheral damage and in auditory neurons in other nuclei. The mechanisms that underlie these postlesion changes are unknown, but the acute effects seen in IC strongly suggest the "unmasking" of latent inputs by the removal of inhibition. In this study, we explore computational models of single neurons with a convergence of excitatory and inhibitory inputs from a range of characteristic frequencies (CFs), which can simulate the narrow prelesion tuning of IC neurons, and account for the changes in CF tuning after a lesion. The models can reproduce the data if inputs are aligned relative to one another in a precise order along the dendrites of model IC neurons. Frequency tuning in these neurons approximates that seen physiologically. Removal of inputs representing a narrow range of frequencies leads to unmasking of previously subthreshold excitatory inputs, which causes changes in CF. Conversely, if all of the inputs converge at the same point on the cell body, receptive fields are broad and unmasking rarely results in CF changes. However, if the inhibition is tonic with no stimulus-driven component, then unmasking can still produce changes in CF.


Subject(s)
Inferior Colliculi/physiology , Models, Neurological , Spiral Ganglion/injuries , Action Potentials/physiology , Animals , Dendrites/physiology , Guinea Pigs , Models, Theoretical , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Neurons/physiology
9.
J Neurosci Res ; 83(7): 1323-30, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16511870

ABSTRACT

After left unilateral cochlear ablation (UCA) in young adult guinea pigs, the appearance of plasticities in auditory pathways suggested altered gene expression and modified phenotypic behaviors of auditory neurons. Because phosphorylated cyclic-AMP response element-binding protein (CREB-P) is a transcription factor that binds to certain genes to facilitate their expression, CREB-P levels were measured after UCA and correlated with postablation plasticities. After UCA, Western blotting was employed to quantify CREB-P levels and illustrate CREB levels in the anteroventral (AVCN), posteroventral (PVCN), and dorsal (DCN) cochlear nucleus; the lateral (LSO) and medial superior olive (MSO); the medial nucleus of the trapezoid body (MNTB); and the central nucleus of the inferior colliculus (ICc) for up to 145 days. We also quantified the levels of several protein synthesis regulators and synaptic markers in the AVCN at 60 days. Sucrose-based extraction buffer improved CREB-P recovery. CREB-P levels became depressed at 3 and 7 postablation days, except in the PVCN, where they were elevated at 7 days, and in the ICc, where they were elevated at both times. At 60 days, CREB-P levels in all the nuclei were elevated. In the AVCN, levels of the protein synthesis regulators and synaptic markers were also elevated at 60 days. By 145 days, CREB-P levels again declined, except in the AVCN, where elevations persisted and increased on the ablated side, and in the ICc, where CREB-P elevations remained. The changes in CREB-P levels coincided with several plasticities in glutamatergic and glycinergic transmitter release and receptor activities, and alterations in neurotrophic support, that developed after UCA. These findings suggest that UCA altered CREB-P levels, which in turn might have contributed to plasticities that appear after UCA.


Subject(s)
Auditory Pathways/metabolism , Brain Stem/metabolism , Cochlea/injuries , Cyclic AMP Response Element-Binding Protein/metabolism , Neuronal Plasticity/physiology , Spiral Ganglion/injuries , Animals , Biomarkers/metabolism , Cochlea/physiopathology , Cochlear Nucleus/metabolism , Denervation , Disease Models, Animal , Gene Expression Regulation/physiology , Guinea Pigs , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/physiopathology , Inferior Colliculi/metabolism , Nerve Tissue Proteins/metabolism , Olivary Nucleus/metabolism , Presynaptic Terminals/metabolism , Receptors, Glutamate/metabolism , Receptors, Glycine/metabolism , Spiral Ganglion/physiopathology , Synapses/metabolism , Synaptic Transmission/physiology , Up-Regulation/physiology
10.
Ann Otol Rhinol Laryngol ; 110(9): 883-91, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11558767

ABSTRACT

The insertion of an intrascalar electrode array during cochlear implantation causes immediate damage to the inner ear and may result in delayed onset of additional damage that may interfere with neuronal stimulation. To date, there have been reports on fewer than 50 temporal bone specimens from patients who had undergone implantation during life. The majority of these were single-channel implants, whereas the majority of implants inserted today are multichannel systems. This report presents the histopathologic findings in temporal bones from 8 individuals who in life had undergone multichannel cochlear implantation, with particular attention to the type and location of trauma and to long-term changes within the cochlea. The effect of these changes on spiral ganglion cell counts and the correlation between speech comprehension and spiral ganglion cell counts were calculated. In 4 of the 8 cases, the opposite, unimplanted ear was available for comparison. In 3 of the 4 cases, there was no significant difference between the spiral ganglion cell counts on the implanted and unimplanted sides. In addition, in this series of 8 cases, there was an apparent negative correlation between residual spiral ganglion cell count and hearing performance during life as measured by single-syllable word recognition. This finding suggests that abnormalities in the central auditory pathways are at least as important as spiral ganglion cell loss in limiting the performance of implant users.


Subject(s)
Cochlear Implants , Aged , Aged, 80 and over , Cadaver , Cell Count , Cochlear Duct/injuries , Cochlear Implants/adverse effects , Deafness/physiopathology , Deafness/surgery , Female , Hearing , Humans , Male , Middle Aged , Osteogenesis , Postoperative Period , Speech Perception , Spiral Ganglion/injuries , Stria Vascularis , Wounds and Injuries/etiology , Wounds and Injuries/pathology
11.
Brain Res ; 905(1-2): 152-60, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11423090

ABSTRACT

Post-traumatic invasion of macrophages into the cochlear nerve of the rat and measurement of how their invasion was modified by the administration of methylprednisolone were investigated for the first time by using a reproducible and quantifiable experimental model of cochlear nerve injury. Two weeks after precise cochlear nerve compression, a massive invasion of ED1 immunostained macrophages was observed at the compressed portion of the cochlear nerve, and this invasion of macrophages was markedly reduced in the rats to which methylprednisolone had been administered during the pre- and post-compression period. Concomitantly, the residual number of spiral ganglion cells was found to be greater in the compression+methylprednisolone group than in the control compression group. The tissue loss observed in the lesion epicenter was also significantly less in the compression+methylprednisolone group than in the control compression group. The results of our present study demonstrated the effectiveness of methylprednisolone treatment to ameliorate trauma induced cochlear nerve degeneration in the acute phase. However, these results may reflect the sum effects of methylprednisolone on macrophages, including both its beneficial effect by inhibiting the negative aspects of macrophages through attenuating macrophage recruitment to the lesion site, and at the same time an undesirable effect by sacrificing the positive aspects of macrophage function. Moreover, one reservation should be added that the protective effects of steroid to injured cochlear nerve may have operated via a pathway not related to macrophage function. Besides macrophages, various cells and factors participate in the process of CNS injury, and their effects may potentially work either positively or negatively with respect to CNS protection and regeneration at each particular time during the on-going process of CNS injury. Therefore, future investigation in CNS injury should be directed toward understanding such complex mechanisms involved in this process.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cochlear Nerve/drug effects , Cochlear Nerve/injuries , Macrophage Activation/drug effects , Macrophages/drug effects , Methylprednisolone/pharmacology , Vestibulocochlear Nerve Diseases/drug therapy , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cochlear Nerve/pathology , Disease Models, Animal , Macrophage Activation/physiology , Macrophages/metabolism , Macrophages/pathology , Male , Nerve Crush , Neural Conduction/drug effects , Neural Conduction/physiology , Rats , Rats, Sprague-Dawley , Retrograde Degeneration/drug therapy , Retrograde Degeneration/pathology , Retrograde Degeneration/physiopathology , Spiral Ganglion/drug effects , Spiral Ganglion/injuries , Spiral Ganglion/pathology , Vestibulocochlear Nerve Diseases/pathology , Vestibulocochlear Nerve Diseases/physiopathology
12.
Hear Res ; 151(1-2): 125-132, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124459

ABSTRACT

We investigated whether methylprednisolone sodium succinate can ameliorate cochlear nerve degeneration following compression injury on the cerebellopontine angle portion of the cochlear nerve, using a quantitative animal experimental model that we have developed recently. In this model, cochlear nerve degeneration after compression could be quantitatively evaluated, while cochlear ischemia induced by the compression carefully maintained below the critical limit that causes irreversible damage to the cochlea. Eleven rats were treated with methylprednisolone during the pre- and post-compression period. Two weeks after compression, the numbers of SGC were compared between the rats that received the compression without and with methylprednisolone treatment. Methylprednisolone treatment improved the survival of SGC following cochlear nerve injury statistically highly significantly in the basal turn where the traumatic stress had been less than in the other cochlear turns in our experimental setting. Although it was not statistically significant, greater survival was also observed in the other cochlear turns. The results of this experimental study indicated that at least a portion of injured cochlear nerve had been potentially treatable, and that methylprednisolone might prevent such cochlear neurons from entering into the vicious process of irreversible damaging process.


Subject(s)
Cochlear Nerve/drug effects , Cochlear Nerve/injuries , Methylprednisolone/pharmacology , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Animals , Cell Count , Cochlear Nerve/pathology , Cochlear Nerve/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory , Male , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Rats , Rats, Sprague-Dawley , Spiral Ganglion/drug effects , Spiral Ganglion/injuries , Spiral Ganglion/pathology
13.
Hear Res ; 147(1-2): 200-20, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10962186

ABSTRACT

Many studies have reported plastic changes in central auditory frequency organization after chronic cochlear lesions. These studies employed mechanical, acoustic or drug-induced disruptions of restricted regions of the organ of Corti that permanently alter its tuning and sensitivity and require an extended recovery period before central effects can be measured. In this study, mechanical lesions were made to 1 mm sectors of the spiral ganglion (SG). These lesions remove a restricted portion of the cochlear output, but leave the organ of Corti and basilar membrane intact. Multiunit mapping assessed the pre- and post-lesion tonotopic organization of the inferior colliculus (IC). Immediately after SG lesions, IC neurons previously tuned to the lesion frequencies became less sensitive to those frequencies but more sensitive to lesion edge frequencies, resulting in a shift in their characteristic frequencies (CFs). Notches in the excitatory response areas at frequencies corresponding to the lesion frequencies and expansion of spatial tuning curves were also observed. CFs of neurons tuned to unlesioned frequencies were unchanged. These results suggest that 'plastic' changes similar to those observed after long survival times in previous studies require little or no experience and occur within minutes to hours following the lesion.


Subject(s)
Inferior Colliculi/physiopathology , Spiral Ganglion/injuries , Spiral Ganglion/physiopathology , Action Potentials , Animals , Auditory Perception/physiology , Cats , Deafness/pathology , Deafness/physiopathology , Inferior Colliculi/pathology , Neuronal Plasticity , Neurons/pathology , Neurons/physiology , Spiral Ganglion/pathology
14.
J Neurocytol ; 28(10-11): 939-54, 1999.
Article in English | MEDLINE | ID: mdl-10900096

ABSTRACT

Laser beam ablation of spiral ganglion neurons was performed in seven organotypic cultures of the newborn mouse cochlea between 5 and 8 days in vitro, with a recovery period of from 18 hours to 3 days. Direct somatic injury (laser or mechanical) inflicted on hair cells does not necessarily cause their death; many of them survive, repair damage and re-establish their neurosensory connections. By contrast, laser irradiation and ablation of their afferent spiral ganglion neurons causes a most spectacular degeneration of sensory cells within 18-48 hours after the insult. Ultrastructurally, the degenerated hair cells-characteristically the inner hair cells-display "dark-cell vacuolar degeneration" that combines the signs of apoptotic death (the peripheral condensation of nuclear chromatin and nuclear pyknosis) with signs of cell edema, vacuolization and necrosis. The ultimate condensation of the cytoplasm gives the dead cells a jet black appearance. The irradiated spiral ganglion neurons die displaying similar pathological characteristics. The extent and locus of inner hair cell degeneration correspond to that of ablated spiral ganglion neurons: ultimately the ablation of one neuron causes degeneration of a single inner hair cell within the closest radial segment of the afferent innervation. The elimination of spiral ganglion neurons by mechanical means does not affect hair cell survival. It is inferred that the laser pulse acts as a stimulus depolarizing the neuronal membrane of the spiral ganglion neurons and their radial fibers and causing the excitotoxic death of their synaptic sensory cells through excessive stimulation of the glutamatergic receptors. Reciprocal pre-and postsynaptic synapses between the afferent dendrites and inner hair cells in culture could possibly serve as entryways of the stimulus. The pathogenesis of this apparent transsynaptically-induced apoptotic death of inner hair cells will be further examined in culture.


Subject(s)
Apoptosis , Hair Cells, Auditory, Inner/pathology , Neurons, Afferent/ultrastructure , Spiral Ganglion/injuries , Spiral Ganglion/ultrastructure , Animals , Cell Survival/radiation effects , Diffusion Chambers, Culture , Hair Cells, Auditory, Inner/ultrastructure , Lasers , Light , Mice , Mice, Inbred ICR , Organ Culture Techniques , Organ of Corti/cytology
15.
Rev Laryngol Otol Rhinol (Bord) ; 119(5): 317-22, 1998.
Article in English | MEDLINE | ID: mdl-10089801

ABSTRACT

On the basis of data reported in the literature, the authors have attempted to define the relationship between the functional results of cochlear implants and possible traumatic damage caused by the insertion of electrodes and their support into the cochlear bony walls. These findings show that traumatic conditions result in functional damage only when they involve the body of Corti's ganglion cells or the central part of their axon, whereas functional results are not influenced by traumatic damage to the peripheral part of the axon. Traumatic damage sustained by other non-nervous structures and the inevitable fibrosis and subsequent bone metaplasia processes which occur when a foreign body penetrates a living organism also appear to be unimportant.


Subject(s)
Cochlea/injuries , Cochlear Implants , Electrodes, Implanted , Postoperative Complications/etiology , Axons/pathology , Cochlea/pathology , Equipment Failure Analysis , Foreign-Body Reaction/pathology , Humans , Organ of Corti/injuries , Organ of Corti/pathology , Postoperative Complications/pathology , Spiral Ganglion/injuries , Spiral Ganglion/pathology
16.
Acta Otolaryngol ; 113(4): 498-501, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8379305

ABSTRACT

Afferent auditory neurons are essential for the transmission of auditory information from Corti's organ to the central auditory pathway. Auditory neurons are very sensitive to acute insult and have a limited ability to regenerate injured neuronal processes. Therefore, these neurons appear to be a limiting factor in restoration of hearing function following an injury to the peripheral auditory receptor. In a previous study nerve growth factor (NGF) was shown to stimulate neurite repair but not survival of injured auditory neurons. In this study, we have demonstrated a neuritogenesis promoting effect of naftidrofuryl in an vitro model for injury to adult auditory neurons, i.e. dissociated cell cultures of adult rat spiral ganglia. Conversely, naftidrofuryl did not have any demonstrable survival promoting effect on these in vitro preparations of injured auditory neurons. The potential uses of this drug as a therapeutic agent in acute diseases of the inner ear are discussed in the light of these observations.


Subject(s)
Nafronyl/pharmacology , Nerve Regeneration/drug effects , Neurites/drug effects , Spiral Ganglion/drug effects , Spiral Ganglion/injuries , Animals , Cell Count , Cell Survival , Culture Media , Culture Techniques , Fibroblast Growth Factor 2 , Laminin , Neurites/physiology , Neurons/drug effects , Neurons/physiology , Peptides , Rats , Rats, Wistar , Spiral Ganglion/physiopathology
17.
Hear Res ; 58(2): 166-74, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1568938

ABSTRACT

Trimethyltin (TMT) is a potent ototoxicant which acutely disrupts generation of the action potential evoked by a broad range of tone frequencies and subsequently produces selective high frequency impairment and outer hair cell (OHC) damage in the extreme basal turn of the cochlea. We investigated the development of TMT ototoxicity in the guinea pig 6-48 h following treatment using the compound action potential (CAP), cochlear microphonic (CM), endocochlear potential (EP) and light and electron microscopic examinations. At all time intervals studied, TMT reduced CAP sensitivity and CM amplitude. The effect was relatively broad across test frequencies at 6 h and subsequently became restricted to higher frequencies. No disruption of the EP was observed between 6 and 24 h following TMT. OHC pathology in the basal turn of the cochlea 12 h following TMT consisted of vacuolization in the supranuclear region and disruption of the cuticular plate; some mitochondria exhibited dark inclusions. Type 1 spiral ganglion cells appeared swollen at 24 h with separation of myelin from the cell bodies. No pathological changes were observed in the inner hair cells (IHC). The present data identify the OHC as targets responsible for the loss of CM sensitivity after TMT as the EP was unaffected. These data suggest that CAP and CM recovery at low and middle frequencies following acute TMT administration is accompanied by recovery of neurotransmission at the IHC or Type 1 SGC level and OHC recovery at apical regions of the cochlea.


Subject(s)
Cochlea/drug effects , Trimethyltin Compounds/toxicity , Action Potentials/drug effects , Animals , Cochlea/injuries , Cochlea/physiopathology , Cochlear Microphonic Potentials/drug effects , Guinea Pigs , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/injuries , Hair Cells, Auditory/ultrastructure , Male , Microscopy, Electron , Spiral Ganglion/drug effects , Spiral Ganglion/injuries , Spiral Ganglion/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...