Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Exp Gerontol ; 48(11): 1173-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23871848

ABSTRACT

We report here beneficial effects of life-long dietary restriction on the progression of age-associated cochlear degeneration in female Sprague-Dawley rats. Thirty-month old rats on a 70% dietary restriction were compared to ad libitum fed age-matched rats, and three-month old adult rats. As expected, aged dietary restricted rats displayed about 20% higher survival rate than age-matched rats fed ad libitum. This difference was reflected also in the auditory system. In the dietary restricted group, 73% of the subjects had preserved auditory reflexes (Preyer), and only modest degeneration of the stria vascularis of the inner ear was observed. In contrast, aged ad libitum fed animals, of which only 15% had detectable Preyer reflexes, showed a marked thinning, cellular degeneration and loss of cell processes in the stria vascularis. The extent of loss of sensory hair cells (~24%) was similar in both the aged groups, and neither group showed a significant reduction in the number of spiral ganglion neurons across adult life-span. The observations thus demonstrate that dietary restriction delays age-related degradation of the auditory system. The results provide further insights into the mechanisms of strial presbycusis.


Subject(s)
Aging/pathology , Aging/physiology , Caloric Restriction , Stria Vascularis/pathology , Stria Vascularis/physiology , Animals , Disease Models, Animal , Female , Microscopy, Electron, Transmission , Neurons/pathology , Presbycusis/pathology , Presbycusis/physiopathology , Presbycusis/prevention & control , Rats , Rats, Sprague-Dawley , Reflex, Acoustic/physiology , Spiral Ganglion/innervation , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology
2.
Semin Cell Dev Biol ; 24(5): 460-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23660234

ABSTRACT

In mammals, auditory information is processed by the hair cells (HCs) located in the cochlea and then rapidly transmitted to the CNS via a specialized cluster of bipolar afferent connections known as the spiral ganglion neurons (SGNs). Although many anatomical aspects of SGNs are well described, the molecular and cellular mechanisms underlying their genesis, how they are precisely arranged along the cochlear duct, and the guidance mechanisms that promote the innervation of their hair cell targets are only now being understood. Building upon foundational studies of neurogenesis and neurotrophins, we review here new concepts and technologies that are helping to enrich our understanding of the development of the nervous system within the inner ear.


Subject(s)
Cochlear Duct/physiology , Hair Cells, Auditory/physiology , Nerve Growth Factors/genetics , Neurogenesis/physiology , Sensory Receptor Cells/physiology , Spiral Ganglion/physiology , Animals , Cell Movement , Cochlear Duct/cytology , Cochlear Duct/growth & development , Cochlear Duct/innervation , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mechanotransduction, Cellular , Nerve Growth Factors/metabolism , Sensory Receptor Cells/cytology , Spiral Ganglion/cytology , Spiral Ganglion/growth & development , Spiral Ganglion/innervation , Synapses/physiology , Synaptic Transmission
3.
Nat Commun ; 4: 1438, 2013.
Article in English | MEDLINE | ID: mdl-23385583

ABSTRACT

Hearing requires an optimal afferent innervation of sensory hair cells by spiral ganglion neurons in the cochlea. Here we report that complementary expression of ephrin-A5 in hair cells and EphA4 receptor among spiral ganglion neuron populations controls the targeting of type I and type II afferent fibres to inner and outer hair cells, respectively. In the absence of ephrin-A5 or EphA4 forward signalling, a subset of type I projections aberrantly overshoot the inner hair cell layer and invade the outer hair cell area. Lack of type I afferent synapses impairs neurotransmission from inner hair cells to the auditory nerve. By contrast, radial shift of type I projections coincides with a gain of presynaptic ribbons that could enhance the afferent signalling from outer hair cells. Ephexin-1, cofilin and myosin light chain kinase act downstream of EphA4 to induce type I spiral ganglion neuron growth cone collapse. Our findings constitute the first identification of an Eph/ephrin-mediated mutual repulsion mechanism responsible for specific sorting of auditory projections in the cochlea.


Subject(s)
Afferent Pathways/metabolism , Ephrin-A5/metabolism , Hair Cells, Auditory, Inner/metabolism , Receptor, EphA4/metabolism , Signal Transduction , Animals , Auditory Threshold , Ephrin-A5/deficiency , Ephrin-A5/genetics , Gene Expression Regulation , Growth Cones/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Models, Biological , Receptor, EphA4/genetics , Spiral Ganglion/cytology , Spiral Ganglion/innervation , Spiral Ganglion/metabolism , Synaptic Transmission
4.
PLoS One ; 7(11): e49550, 2012.
Article in English | MEDLINE | ID: mdl-23185359

ABSTRACT

Noise-exposure at levels low enough to avoid a permanent threshold shift has been found to cause a massive, delayed degeneration of spiral ganglion neurons (SGNs) in mouse cochleae. Damage to the afferent innervation was initiated by a loss of synaptic ribbons, which is largely irreversible in mice. A similar delayed loss of SGNs has been found in guinea pig cochleae, but at a reduced level, suggesting a cross-species difference in SGN sensitivity to noise. Ribbon synapse damage occurs "silently" in that it does not affect hearing thresholds as conventionally measured, and the functional consequence of this damage is not clear. In the present study, we further explored the effect of noise on cochlear afferent innervation in guinea pigs by focusing on the dynamic changes in ribbon counts over time, and resultant changes in temporal processing. It was found that (1) contrary to reports in mice, the initial loss of ribbons largely recovered within a month after the noise exposure, although a significant amount of residual damage existed; (2) while the response threshold fully recovered in a month, the temporal processing continued to be deteriorated during this period.


Subject(s)
Spiral Ganglion/innervation , Spiral Ganglion/physiopathology , Action Potentials , Animals , Auditory Threshold , Cochlea/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Guinea Pigs , Hearing , Male , Mice , Microscopy, Confocal/methods , Neurons/drug effects , Noise , Synapses , Time Factors
5.
Hear Res ; 278(1-2): 1, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21723932
6.
Hear Res ; 282(1-2): 289-302, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21763410

ABSTRACT

It has been demonstrated that INS can be utilized to stimulate spiral ganglion cells in the cochlea. Although neural stimulation can be achieved without direct contact of the radiation source and the tissue, the presence of fluids or bone between the target structure and the radiation source may lead to absorption or scattering of the radiation, which may limit the efficacy of INS. The present study demonstrates the neural structures in the radiation beam path that can be stimulated. Histological reconstructions and microCT of guinea pig cochleae stimulated with an infrared laser suggest that the orientation of the beam from the optical fiber determined the site of stimulation in the cochlea. Best frequencies of the INS-evoked neural responses obtained from the central nucleus of the inferior colliculus matched the histological sites in the spiral ganglion.


Subject(s)
Cochlea/innervation , Cochlea/radiation effects , Inferior Colliculi/radiation effects , Infrared Rays , Lasers, Semiconductor , Spiral Ganglion/innervation , Spiral Ganglion/radiation effects , Acoustic Stimulation , Animals , Cochlea/diagnostic imaging , Evoked Potentials, Auditory, Brain Stem , Female , Guinea Pigs , Inferior Colliculi/physiology , Male , Scattering, Radiation , Time Factors , X-Ray Microtomography
7.
Hear Res ; 278(1-2): 106-12, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21640179

ABSTRACT

Loss of spiral ganglion neurons is a major cause of age-related hearing loss (presbycusis). Despite being the third most prevalent condition afflicting elderly persons, there are no known medications to prevent presbycusis. Because calcium signaling has long been implicated in age-related neuronal death, we investigated T-type calcium channels. This family is comprised of three members (Ca(v)3.1, Ca(v)3.2, and Ca(v)3.3), based on their respective main pore-forming alpha subunits: α1G, α1H, and α1I. In the present study, we report a significant delay of age-related loss of cochlear function and preservation of spiral ganglion neurons in α1H null and heterozygous mice, clearly demonstrating an important role for Ca(v)3.2 in age-related neuronal loss. Furthermore, we show that anticonvulsant drugs from a family of T-type calcium channel blockers can significantly preserve spiral ganglion neurons during aging. To our knowledge, this is the first report of drugs capable of diminishing age-related loss of spiral ganglion neurons.


Subject(s)
Anticonvulsants/pharmacology , Calcium Channels, T-Type/metabolism , Presbycusis/metabolism , Presbycusis/prevention & control , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Aging/drug effects , Aging/metabolism , Aging/pathology , Animals , Base Sequence , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/deficiency , Calcium Channels, T-Type/genetics , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/pathology , Mice , Mice, Congenic , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Presbycusis/pathology , RNA/genetics , RNA/metabolism , Spiral Ganglion/innervation , Spiral Ganglion/pathology
8.
Hear Res ; 278(1-2): 96-105, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21616131

ABSTRACT

Significant advances in the functional outcomes achieved with cochlear implantation will likely require tissue-engineering approaches to improve the neural prosthesis interface. One strategy is to direct spiral ganglion neuron (SGN) axon growth in a highly organized fashion to approximate or contact stimulating electrodes. Here we assessed the ability of micropatterns induced by photopolymerization in methacrylate (MA) polymer systems to direct cultured neonatal rat SGN neurite growth and alignment of SG Schwann cells (SGSCs). SGN survival and neurite length were comparable among various polymer compositions. Remarkably, there was no significant difference in SGN survival or neurite length between laminin and non-laminin coated MA polymer substrates, suggesting high biocompatibility with SG tissue. Micropatterning with photopolymerization generated microchannels with a ridge periodicity of 50 µm and channel depths of 0.6-1.0 µm. SGN neurites grew within the grooves of the microchannels. These topographies strongly induced alignment of dissociated SGN neurites and SGSCs to parallel the pattern. By contrast, fibroblasts failed to align with the micropattern suggesting cell specific responses to topographical cues. SGN neurites extending from explants turned to parallel the pattern as they encountered the microchannels. The extent of turning was significantly correlated with angle at which the neurite initially encountered the pattern. These results indicate that SGN neurites respond to microtopographical features and that these features can be used to direct neurite growth in a highly organized fashion.


Subject(s)
Schwann Cells/cytology , Spiral Ganglion/cytology , Tissue Engineering/methods , Tissue Scaffolds , Animals , Biocompatible Materials , Cell Survival , Cells, Cultured , Cochlear Implants , Deafness/pathology , Deafness/surgery , Neurites/ultrastructure , Polymethacrylic Acids , Rats , Spiral Ganglion/innervation , Surface Properties
9.
Hear Res ; 278(1-2): 69-76, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21557994

ABSTRACT

A cochlear implant can restore hearing function by electrically exciting spiral ganglion neurons (SGNs) in the deaf cochlea. However, following deafness SGNs undergo progressive degeneration ultimately leading to their death. One significant cause of SGN degeneration is the loss of neurotrophic support that is normally provided by cells within the organ of Corti (OC). The administration of exogenous neurotrophins (NTs) can protect SGNs from degeneration but the effects are short-lived once the source of NTs has been exhausted. NT gene therapy, whereby cells within the cochlea are transfected with genes enabling them to produce NTs, is one strategy for providing a cellular source of NTs that may provide long-term support for SGNs. As the SGNs normally innervate sensory cells within the OC, targeting residual OC cells for gene therapy in the deaf cochlea may provide a source of NTs for SGN protection and targeted regrowth of their peripheral fibers. However, the continual degeneration of the OC over extended periods of deafness may deplete the cellular targets for NT gene therapy and hence limit the effectiveness of this method in preventing SGN loss. This study examined the effects of deafness duration on the efficacy of NT gene therapy in preventing SGN loss in guinea pigs that were systemically deafened with aminoglycosides. Adenoviral vectors containing green fluorescent protein (GFP) with or without genes for Brain Derived Neurotrophic Factor (BDNF) and Neurotrophin-3 (NT3) were injected into the scala media (SM) compartment of cochleae that had been deafened for one, four or eight weeks prior to the viral injection. The results showed that viral transfection of cells within the SM was still possible even after severe degeneration of the OC. Supporting cells (pillar and Deiters' cells), cells within the stria vascularis, the spiral ligament, endosteal cells lining the scala compartments and interdental cells in the spiral limbus were transfected. However, the level of transfection was remarkably lower following longer durations of deafness. There was a significant increase in SGN survival in the entire basal turn for cochleae that received NT gene therapy compared to the untreated contralateral control cochleae for the one week deaf group. In the four week deaf group significant SGN survival was observed in the lower basal turn only. There was no increase in SGN survival for the eight week deaf group in any cochlear region. These findings indicated that the efficacy of NT gene therapy diminished with increasing durations of deafness leading to reduced benefits in terms of SGN protection. Clinically, there remains a window of opportunity in which NT gene therapy can provide ongoing trophic support for SGNs.


Subject(s)
Deafness/therapy , Genetic Therapy/methods , Nerve Growth Factors/genetics , Spiral Ganglion/pathology , Adenoviridae/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/therapeutic use , Cell Count , Cell Survival , Deafness/pathology , Deafness/physiopathology , Female , Gene Expression , Genes, Reporter , Genetic Vectors , Guinea Pigs , Male , Nerve Growth Factors/therapeutic use , Neurons/pathology , Neurotrophin 3/genetics , Neurotrophin 3/therapeutic use , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Spiral Ganglion/innervation , Spiral Ganglion/physiopathology , Time Factors
10.
Hear Res ; 281(1-2): 56-64, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21596129

ABSTRACT

Following the onset of sensorineural hearing loss, degeneration of mechanosensitive hair cells and spiral ganglion cells (SGCs) in humans and animals occurs to variable degrees, with a trend for greater neural degeneration with greater duration of deafness. Emergence of the cochlear implant prosthesis has provided much needed aid to many hearing impaired patients and has become a well-recognized therapy worldwide. However, ongoing peripheral nerve fiber regression and subsequent degeneration of SGC bodies can reduce the neural targets of cochlear implant stimulation and diminish its function. There is increasing interest in bio-engineering approaches that aim to enhance cochlear implant efficacy by preventing SGC body degeneration and/or regenerating peripheral nerve fibers into the deaf sensory epithelium. We review the advancements in maintaining and regenerating nerves in damaged animal cochleae, with an emphasis on the therapeutic capacity of neurotrophic factors delivered to the inner ear after an insult. Additionally, we summarize the histological process of neuronal degeneration in the inner ear and describe different animal models that have been employed to study this mechanism. Research on enhancing the biological infrastructure of the deafened cochlea in order to improve cochlear implant efficacy is of immediate clinical importance.


Subject(s)
Cochlea/innervation , Hearing Loss, Sensorineural/physiopathology , Nerve Regeneration , Animals , Cochlea/injuries , Cochlea/pathology , Cochlear Implantation/instrumentation , Cochlear Implants , Correction of Hearing Impairment , Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/pathology , Hearing Loss, Sensorineural/rehabilitation , Humans , Models, Animal , Nerve Growth Factors/pharmacology , Persons With Hearing Impairments/rehabilitation , Spiral Ganglion/innervation , Spiral Ganglion/pathology
11.
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
12.
Hear Res ; 278(1-2): 2-20, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21530629

ABSTRACT

In mammals, the initial bridge between the physical world of sound and perception of that sound is established by neurons of the spiral ganglion. The cell bodies of these neurons give rise to peripheral processes that contact acoustic receptors in the organ of Corti, and the central processes collect together to form the auditory nerve that projects into the brain. In order to better understand hearing at this initial stage, we need to know the following about spiral ganglion neurons: (1) their cell biology including cytoplasmic, cytoskeletal, and membrane properties, (2) their peripheral and central connections including synaptic structure; (3) the nature of their neural signaling; and (4) their capacity for plasticity and rehabilitation. In this report, we will update the progress on these topics and indicate important issues still awaiting resolution.


Subject(s)
Spiral Ganglion/physiology , Animals , Auditory Cortex/physiology , Auditory Pathways/physiology , Cochlear Nerve/physiology , Cytoskeleton/physiology , Hair Cells, Auditory/physiology , Humans , Nerve Growth Factors/physiology , Nerve Regeneration , Neuronal Plasticity , Neurons/physiology , Spiral Ganglion/cytology , Spiral Ganglion/innervation
13.
Hear Res ; 278(1-2): 21-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21414397

ABSTRACT

The bipolar spiral ganglion neurons apparently delaminate from the growing cochlear duct and migrate to Rosenthal's canal. They project radial fibers to innervate the organ of Corti (type I neurons to inner hair cells, type II neurons to outer hair cells) and also project tonotopically to the cochlear nuclei. The early differentiation of these neurons requires transcription factors to regulate migration, pathfinding and survival. Neurog1 null mice lack formation of neurons. Neurod1 null mice show massive neuronal death combined with aberrant central and peripheral projections. Prox1 protein is necessary for proper type II neuron process navigation, which is also affected by the neurotrophins Bdnf and Ntf3. Neurotrophin null mutants show specific patterns of neuronal loss along the cochlea but remaining neurons compensate by expanding their target area. All neurotrophin mutants have reduced radial fiber growth proportional to the degree of loss of neurotrophin alleles. This suggests a simple dose response effect of neurotrophin concentration. Keeping overall concentration constant, but misexpressing one neurotrophin under regulatory control of another one results in exuberant fiber growth not only of vestibular fibers to the cochlea but also of spiral ganglion neurons to outer hair cells suggesting different effectiveness of neurotrophins for spiral ganglion neurite growth. Finally, we report here for the first time that losing all neurons in double null mutants affects extension of the cochlear duct and leads to formation of extra rows of outer hair cells in the apex, possibly by disrupting the interaction of the spiral ganglion with the elongating cochlea.


Subject(s)
Hair Cells, Auditory/cytology , Organ of Corti/cytology , Spiral Ganglion/innervation , Animals , Auditory Pathways/cytology , Auditory Pathways/physiology , Hair Cells, Auditory/physiology , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology , Organ of Corti/physiology , Spiral Ganglion/physiology
14.
Hear Res ; 278(1-2): 34-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21420476

ABSTRACT

Thin-sheet laser imaging microscopy (TSLIM) was used to serially section five whole cochleas from 4-wk-old CBA/JCr mice. Three-dimensional reconstructions of Rosenthal's canal (RC) were produced in order to measure canal length and volume, to generate orthogonal cross sections for area measurements, and to determine spiral ganglion neuron (SGN) number. RC length averaged 2.0 mm ± 0.04 (SEM) as measured along the centroid of the canal compared to an average basilar membrane (BM) length of 5.9 ± 0.05 (SEM). RC volume averaged 0.036 mm(3) ± 0.009 (SEM). Significant increases in the radial area of RC were observed at the base (13%), middle (62%), and apex (90%) of its length. The total number of spiral ganglion neurons (SGNs) in RC in each of the five animals averaged 8626 ± 96 (SEM). SGN number increased at the expanded regions of RC. Increased area and cell number at the base and apex are likely related to extensions of the organ of Corti past the length of RC in these areas. The increase in area and cell number in the middle of the RC appears to be related to the most sensitive frequency region of the organ of Corti. Volume imaging or tomography of the cochlea as provided by TSLIM has the potential to be an efficient and accurate semi-automated method for the quantitative assessment of the number of SGNs and hair cells of the organ of Corti.


Subject(s)
Spiral Ganglion/anatomy & histology , Animals , Cell Count , Female , Imaging, Three-Dimensional , Mice , Mice, Inbred CBA , Microscopy, Confocal , Models, Anatomic , Models, Neurological , Neurons/cytology , Spiral Ganglion/cytology , Spiral Ganglion/innervation
15.
Hear Res ; 278(1-2): 43-51, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21329751

ABSTRACT

Accurate counting of neurons in the cochlea has a significant impact on the interpretation of research and clinically relevant data. However, reports of numbers of neurons in the spiral ganglion are widely variable across studies, even within the same species. We suggest that the implementation of a more standardized, unbiased counting method will improve the consistency and accuracy of neuron counts and will impact scientific interpretations. To test this view, we compared, in different ways, the numbers of neurons in the spiral ganglia of developing gerbils, previously reported to decrease by 22-27% between birth and age 7 days. Cochleae from gerbils, aged newborn, 7 days, 20 days, 1.5 years and 2.5 years were embedded in Araldite and serially sectioned at 5 µm. A computer based stereological method was used to unambiguously count every neuron in serial sections, either throughout the entire cochlea, or in a 100-µm segment of the cochlea. No significant changes in neuron numbers during cochlear maturation were found. We demonstrate that in methods using sampling of sections, the identity of the starting section and the interval between sections impacts the variability of the estimate of neuron numbers. In addition, we show that packing density differs between the newborn and seven-day old animals. The data demonstrate that variability in counting methods and the comparison of non-uniform samples can lead to neuron number estimates that show differences where none exist. We propose that a standardized counting protocol be implemented across studies and suggest possible approaches to different types of comparisons between neurons of spiral ganglia from different sources.


Subject(s)
Cochlea/innervation , Gerbillinae/anatomy & histology , Neurons/cytology , Animals , Animals, Newborn , Cell Count , Cochlea/cytology , Cochlea/growth & development , Female , Gerbillinae/growth & development , Histological Techniques , Male , Spiral Ganglion/cytology , Spiral Ganglion/growth & development , Spiral Ganglion/innervation
16.
Hear Res ; 278(1-2): 52-68, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21281707

ABSTRACT

As with other elements of the peripheral auditory system, spiral ganglion neurons display specializations that vary as a function of location along the tonotopic axis. Previous work has shown that voltage-gated K(+) channels and synaptic proteins show graded changes in their density that confers rapid responsiveness to neurons in the high frequency, basal region of the cochlea and slower, more maintained responsiveness to neurons in the low frequency, apical region of the cochlea. In order to understand how voltage-gated calcium channels (VGCCs) may contribute to these diverse phenotypes, we identified the VGCC α-subunits expressed in the ganglion, investigated aspects of Ca(2+)-dependent neuronal firing patterns, and mapped the intracellular and intercellular distributions of seven VGCC α-subunits in the spiral ganglion in vitro. Initial experiments with qRT-PCR showed that eight of the ten known VGCC α-subunits were expressed in the ganglion and electrophysiological analysis revealed firing patterns that were consistent with the presence of both LVA and HVA Ca(2+) channels. Moreover, we were able to study seven of the α-subunits with immunocytochemistry, and we found that all were present in spiral ganglion neurons, three of which were neuron-specific (Ca(V)1.3, Ca(V)2.2, and Ca(V)3.3). Further characterization of neuron-specific α-subunits showed that Ca(V)1.3 and Ca(V)3.3 were tonotopically-distributed, whereas Ca(V)2.2 was uniformly distributed in apical and basal neurons. Multiple VGCC α-subunits were also immunolocalized to Schwann cells, having distinct intracellular localizations, and, significantly, appearing to distinguish putative compact (Ca(V)2.3, Ca(V)3.1) from loose (Ca(V)1.2) myelin. Electrophysiological evaluation of spiral ganglion neurons in the presence of TEA revealed Ca(2+) plateau potentials with slopes that varied proportionately with the cochlear region from which neurons were isolated. Because afterhyperpolarizations were minimal or absent under these conditions, we hypothesize that differential density and/or kinetics of one or more of the VGCC α-subunits could account for observed tonotopic differences. These experiments have set the stage for defining the clear multiplicity of functional control in neurons and Schwann cells of the spiral ganglion.


Subject(s)
Spiral Ganglion/metabolism , Action Potentials , Animals , Base Sequence , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , DNA Primers/genetics , Electrophysiological Phenomena , Immunohistochemistry , Mice , Mice, Inbred CBA , Neurons/metabolism , Protein Subunits , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/metabolism , Spiral Ganglion/cytology , Spiral Ganglion/innervation
17.
Gene Ther ; 18(6): 560-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21228883

ABSTRACT

Apoptosis is responsible for cochlear cell death induced by noise. Here, we show that transgenic (TG) mice that overexpress X-linked inhibitor of apoptosis protein (XIAP) under control of the ubiquitin promoter display reduced hearing loss and cochlear damage induced by acoustic overstimulation (125 dB sound pressure level, 6 h) compared with wild-type (WT) littermates. Hearing status was evaluated using the auditory brainstem response (ABR), whereas cochlear damage was assessed by counts of surviving hair cells (HCs) and spiral ganglion neurons (SGNs) as well as their fibers to HCs. Significantly smaller threshold shifts were found for TG mice than WT littermates. Correspondingly, the TG mice also showed a reduced loss of HCs, SGNs and their fibers to HCs. HC loss was limited to the basal end of the cochlea that detects high frequency sound. In contrast, the ABRs demonstrated a loss of hearing sensitivity across the entire frequency range tested (2-32 kHz) indicating that the hearing loss could not be fully attributed to HC loss alone. The TG mice displayed superior hearing sensitivity over this whole range, suggesting that XIAP overexpression reduces noise-induced hearing loss not only by protecting HCs but also other components of the cochlea.


Subject(s)
Hearing Loss, Noise-Induced/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , Animals , Auditory Threshold , Cochlea/injuries , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Spiral Ganglion/innervation , Ubiquitin/genetics
18.
Dev Neurobiol ; 67(13): 1721-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17600813

ABSTRACT

Extracellular matrix (ECM) molecules have been shown to function as cues for neurite guidance in various populations of neurons. Here we show that laminin (LN) and fibronectin (FN) presented in stripe micro-patterns can provide guidance cues to neonatal (P5) inner ear spiral ganglion (SG) neurites. The response to both ECM molecules was dose-dependent. In a LN versus poly-L-lysine (PLL) assay, neurites were more often observed on PLL at low coating concentrations (5 and 10 microg/mL), while they were more often on LN at a high concentration (80 microg/mL). In a FN versus PLL assay, neurites were more often on PLL than on FN stripes at high coating concentrations (40 and 80 microg/mL). In a direct competition between LN and FN, neurites were observed on LN significantly more often than on FN at both 10 and 40 microg/mL. The data suggest a preference by SG neurites for LN at high concentrations, as well as avoidance of both LN at low and FN at high concentrations. The results also support a potential model for neurite guidance in the developing inner ear in vivo. LN, in the SG and osseus spiral lamina may promote SG dendrite growth toward the organ of Corti. Within the organ of Corti, lower concentrations of LN may slow neurite growth, with FN beneath each row of hair cells providing a stop or avoidance signal. This could allow growth cone filopodia increased time to sample their cellular targets, or direct the fibers upward toward the hair cells.


Subject(s)
Fibronectins/metabolism , Laminin/metabolism , Neurites/metabolism , Spiral Ganglion/innervation , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Immunohistochemistry , In Vitro Techniques , Rats , Rats, Sprague-Dawley , Spiral Ganglion/cytology , Spiral Ganglion/metabolism
19.
Hear Res ; 40(1-2): 93-109, 1989 Jun 15.
Article in English | MEDLINE | ID: mdl-2768087

ABSTRACT

Cochlear efferent and afferent units were recorded from the spiral ganglion in anesthetized guinea pigs. Efferent units were identified by their regular discharge patterns and their long response latencies to tone bursts. In some cases these physiological criteria were confirmed by anatomical tracing of single axons labeled with horseradish peroxidase. Labeled efferent axons traveled in the vestibular nerve root and the intraganglionic spiral bundle, and crossed the tunnel to innervate up to 61 outer hair cells over longitudinal distances of 0.15 to 2.1 mm. Efferent units were subdivided into classes by their excitatory responses to monaural tone bursts. Fifty-seven percent responded only to ipsilateral-ear sound, 28% only to contralateral-ear sound, and 15% to sound in either ear. Tuning curves from efferent units were generally similar in shape to afferent units, often having equally low thresholds and equally high Q10s. Efferent Q10s were somewhat lower from preparations anesthetized with a Urethane/Innovar combination than from preparations anesthetized with a Nembutal/Innovar combination. Efferent units with spontaneous activity were uncommon at the start of the recording sessions but were more frequently encountered later in the experiments. Spontaneous activity could often be suppressed by tonal stimuli, even sometimes to the non-excitatory ear of Ipsi and Contra units.


Subject(s)
Cochlea/innervation , Olivary Nucleus/physiology , Spiral Ganglion/innervation , Acoustic Stimulation , Action Potentials/drug effects , Anesthetics/pharmacology , Animals , Cochlea/physiology , Efferent Pathways/physiology , Guinea Pigs , Olivary Nucleus/cytology , Reaction Time/physiology , Spiral Ganglion/physiology
20.
Hear Res ; 34(1): 63-8, 1988 Jul 01.
Article in English | MEDLINE | ID: mdl-3403386

ABSTRACT

Aspartate aminotransferase and glutaminase immunoreactive labeling of the auditory nerve has previously been reported. In the present study, the development of these immunoreactivities was examined in the auditory nerve of the rat, at ages ranging from 17 days gestation to four postnatal weeks. Cells and processes were examined in the cochlea, and fibers and terminals in the cochlear nucleus. In the cochlea, immunoreactive labeling with antisera to both enzymes was first seen at 20 gestational days, in spiral ganglion cells. It was not until two postnatal weeks, however, that this immunoreactive labeling was first seen in primary afferent terminals around spherical cells in the anteroventral cochlear nucleus. This correlates with the establishment of mature synaptic connections and function.


Subject(s)
Aging/metabolism , Aspartate Aminotransferases/metabolism , Cochlea/innervation , Glutaminase/metabolism , Spiral Ganglion/innervation , Vestibulocochlear Nerve/enzymology , Animals , Cochlear Nerve/embryology , Cochlear Nerve/enzymology , Cochlear Nerve/growth & development , Immunohistochemistry , Rats , Rhombencephalon/embryology , Rhombencephalon/enzymology , Rhombencephalon/growth & development , Spiral Ganglion/embryology , Spiral Ganglion/growth & development , Vestibulocochlear Nerve/embryology , Vestibulocochlear Nerve/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...