Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Sci Rep ; 10(1): 5877, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245997

ABSTRACT

Human spiral ganglion (HSG) cell bodies located in the bony cochlea depend on a rich vascular supply to maintain excitability. These neurons are targeted by cochlear implantation (CI) to treat deafness, and their viability is critical to ensure successful clinical outcomes. The blood supply of the HSG is difficult to study due to its helical structure and encasement in hard bone. The objective of this study was to present the first three-dimensional (3D) reconstruction and analysis of the HSG blood supply using synchrotron radiation phase-contrast imaging (SR-PCI) in combination with histological analyses of archival human cochlear sections. Twenty-six human temporal bones underwent SR-PCI. Data were processed using volume-rendering software, and a representative three-dimensional (3D) model was created to allow visualization of the vascular anatomy. Histologic analysis was used to verify the segmentations. Results revealed that the HSG is supplied by radial vascular twigs which are separate from the rest of the inner ear and encased in bone. Unlike with most organs, the arteries and veins in the human cochlea do not follow the same conduits. There is a dual venous outflow and a modiolar arterial supply. This organization may explain why the HSG may endure even in cases of advanced cochlear pathology.


Subject(s)
Imaging, Three-Dimensional/methods , Microscopy, Phase-Contrast/methods , Spiral Ganglion/blood supply , Synchrotrons , Adult , Cochlea/anatomy & histology , Cochlea/diagnostic imaging , Cochlea/ultrastructure , Humans , Spiral Ganglion/anatomy & histology , Spiral Ganglion/diagnostic imaging , Spiral Ganglion/ultrastructure , Veins/anatomy & histology , Veins/diagnostic imaging , Veins/ultrastructure
2.
Hear Res ; 390: 107928, 2020 05.
Article in English | MEDLINE | ID: mdl-32143110

ABSTRACT

Severe damage to the organ of Corti leads to degeneration of the spiral ganglion cells (SGCs) which form the auditory nerve. This degeneration starts at the level of synaptic connection of the peripheral processes (PPs) of SGCs with the cochlear hair cells. It is generally thought that from this point SGC degeneration progresses in a retrograde fashion: PPs degenerate first, followed by the SGC soma with a delay of several weeks to many months. Evidence for this course of events, both in animals and in humans, is not unambiguous, while this knowledge is important since the presence or absence of the different neural elements may greatly influence the response to electrical stimulation with a cochlear implant (CI). We therefore aimed to provide a comprehensive account of the course of SGC degeneration in the guinea pig cochlea after ototoxic treatment. Histological analysis of eighteen healthy and thirty-three deafened cochleas showed that the degeneration of SGCs and their peripheral processes was simultaneous rather than sequential. As the site of excitation for electrical stimulation with a CI may depend on the course of degeneration of the various neural elements, this finding is relevant both for understanding the electrophysiological mechanisms behind cochlear implantation and for recent efforts to induce PP resprouting for improved electrode-neural interface. Since excitation of the PPs is often thought to result in (secondary) longer-latency activity, we tested the hypothesis that having relatively many PPs produces a larger N2 peak in the electrically evoked compound action potential (eCAP); the present findings however do not support this theory. The course of the degeneration process may vary among species, and may depend on the cause of deafness, but the present findings at least indicate that gradual retrograde degeneration of the auditory nerve is not an elemental process following severe damage to the organ of Corti.


Subject(s)
Deafness/pathology , Hair Cells, Auditory/ultrastructure , Nerve Degeneration , Spiral Ganglion/ultrastructure , Animals , Auditory Threshold , Deafness/chemically induced , Deafness/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Furosemide , Guinea Pigs , Kanamycin , Ototoxicity , Spiral Ganglion/physiopathology
3.
Biosci Rep ; 39(2)2019 02 28.
Article in English | MEDLINE | ID: mdl-30626727

ABSTRACT

Aminoglycoside antibiotics-induced hearing loss is a common sensorineural impairment. Spiral ganglion neurons (SGNs) are first-order neurons of the auditory pathway and are critical for the maintenance of normal hearing. In the present study, we investigated the time-course of morphological changes and the degeneration process of spiral ganglion cells (SGCs) following chronic kanamycin-induced deafness and determined whether the endoplasmic reticulum (ER) stress was involved in the degeneration of SGNs. We detected density changes in SGCs and the expressions of Bip, inositol requirement 1 (IRE1)α, activating transcription factor-6α, p-PERK, p-eIF2α, CHOP, and caspase-12 at each time point after kanamycin treatment. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was also performed. The number of SGC deletions reached ∼50% at the 70th day after kanamycin administration and the ER of most SGCs were dilated. The expression of p-PERK, p-eIF2α, p-IRE1α, Bip, caspase-12, and Chop was significantly unregulated after kanamycin treatment. The number of SGCs that were positive for both TUNEL and caspase-12 increased from day 7 to 28. Taken together, these data demonstrate that ER stress was involved in kanamycin-induced apoptosis of SGNs. Kanamycin-induced SGN apoptosis is mediated, at least in part, by ER stress-induced upregulation of CHOP and caspase-12.


Subject(s)
Deafness/pathology , Endoplasmic Reticulum Stress , Kanamycin/adverse effects , Neurons/pathology , Spiral Ganglion/pathology , Animals , Apoptosis/drug effects , Caspase 12/metabolism , Deafness/chemically induced , Deafness/physiopathology , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Heat-Shock Proteins/metabolism , Male , Multienzyme Complexes/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/ultrastructure , Transcription Factor CHOP/metabolism , eIF-2 Kinase/metabolism
4.
Sci Rep ; 8(1): 7446, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748613

ABSTRACT

This study investigated micro-patterned, high-density complementary metal-oxide-semiconductor (CMOS) electrode array to be used as biologically permissive environment for organization, guidance and electrical stimulation of spiral ganglion neurons (SGN). SGNs extracted and isolated from cochleae of P5-P7 rat pups and adult guinea pigs were cultured 1, 4 and 7 days in vitro on glass coverslips (control) and CMOS electrode array. The cultures were analyzed visually and immunohistochemically for SGN presence, outgrowth, neurite alignment, neurite length, neurite asymmetry as well as the contact of a neuronal soma and neurites with the micro-electrodes. Our findings indicate that topographical environment of CMOS chip with micro-patterned pillars enhanced growth, survival, morphology, neural orientation and alignment of SGNs in vitro compared to control. Smaller spacing (0.8-1.6 µm) between protruding pillars on CMOS led SGNs to develop structured and guided neurites oriented along three topographical axes separated by 60°. We found morphological basis for positioning of the micro-electrodes on the chip that was appropriate for direct contact of SGNs with them. This configuration allowed CMOS electrode array to electrically stimulate the SGN whose responses were observed with live Fluo 4 calcium imaging.


Subject(s)
Cell Culture Techniques/instrumentation , Electric Stimulation/instrumentation , Lab-On-A-Chip Devices , Neurons/cytology , Semiconductors , Spiral Ganglion/cytology , Animals , Axon Guidance , Cells, Cultured , Electrodes , Equipment Design , Guinea Pigs , Metals/chemistry , Neurites/metabolism , Neurites/ultrastructure , Neurogenesis , Neurons/metabolism , Neurons/ultrastructure , Oxides/chemistry , Rats , Spiral Ganglion/metabolism , Spiral Ganglion/ultrastructure
5.
Front Immunol ; 9: 223, 2018.
Article in English | MEDLINE | ID: mdl-29487598

ABSTRACT

The human inner ear, which is segregated by a blood/labyrinth barrier, contains resident macrophages [CD163, ionized calcium-binding adaptor molecule 1 (IBA1)-, and CD68-positive cells] within the connective tissue, neurons, and supporting cells. In the lateral wall of the cochlea, these cells frequently lie close to blood vessels as perivascular macrophages. Macrophages are also shown to be recruited from blood-borne monocytes to damaged and dying hair cells induced by noise, ototoxic drugs, aging, and diphtheria toxin-induced hair cell degeneration. Precise monitoring may be crucial to avoid self-targeting. Macrophage biology has recently shown that populations of resident tissue macrophages may be fundamentally different from circulating macrophages. We removed uniquely preserved human cochleae during surgery for treating petroclival meningioma compressing the brain stem, after ethical consent. Molecular and cellular characterization using immunofluorescence with antibodies against IBA1, TUJ1, CX3CL1, and type IV collagen, and super-resolution structured illumination microscopy (SR-SIM) were made together with transmission electron microscopy. The super-resolution microscopy disclosed remarkable phenotypic variants of IBA1 cells closely associated with the spiral ganglion cells. Monitoring cells adhered to neurons with "synapse-like" specializations and protrusions. Active macrophages migrated occasionally nearby damaged hair cells. Results suggest that the human auditory nerve is under the surveillance and possible neurotrophic stimulation of a well-developed resident macrophage system. It may be alleviated by the non-myelinated nerve soma partly explaining why, in contrary to most mammals, the human's auditory nerve is conserved following deafferentiation. It makes cochlear implantation possible, for the advantage of the profoundly deaf. The IBA1 cells may serve additional purposes such as immune modulation, waste disposal, and nerve regeneration. Their role in future stem cell-based therapy needs further exploration.


Subject(s)
Cochlea/immunology , DNA-Binding Proteins/immunology , Macrophages/immunology , Spiral Ganglion/immunology , Aged , Calcium-Binding Proteins , Cell Movement/immunology , Cochlea/cytology , Cochlea/transplantation , Cochlea/ultrastructure , Cochlear Implantation , DNA-Binding Proteins/metabolism , Deafness/surgery , Female , Hair Cells, Auditory/immunology , Hair Cells, Auditory/ultrastructure , Humans , Immunohistochemistry/methods , Macrophages/metabolism , Male , Microfilament Proteins , Microscopy, Electron, Transmission , Middle Aged , Spiral Ganglion/cytology , Spiral Ganglion/ultrastructure
6.
Sci Rep ; 7(1): 8638, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819108

ABSTRACT

Mammalian Sperm Associated Antigen 6 (SPAG6) is the orthologue of Chlamydomonas PF16, a protein localized in the axoneme central apparatus. Recent studies showed that Spag6 has a role in brain neuronal proliferation and differentiation. The mammalian spiral ganglion neurons (SGNs) are specialzed bipolar neurons in the inner ear. However, the role of SPAG6 in SGN has not been elucidated. Therefore, We hypothesized that a Spag6 knockout would affect the development and function of SGNs. We utilized Spag6-deficient mice and SGN explants to define the role of SPAG6. On postnatal day 30 (P30) mutant mice had lower SGN density compared to their wild-type littermates, and more apoptosis was evident in the mutants. Increased Bax expression, a disturbed distribution of cytochrome c, and cleaved caspase-3 positive staining indicated that increased apoptosis involved a mitochondrial pathway. Transmission electron microscopy revealed abnormalities in the ultrastructure of mutant SGNs as early as P7. In vitro, lack of SPAG6 affected the growth of neurites and growth cones. Additionally, SPAG6 deficiency decreased synapse density in SGN explants. Finally, Spag6 mutant SGNs were more sensitive to the microtubule stabilizing agent, paclitaxel. These findings suggest that Spag6 plays a crucial role in SGN development and function.


Subject(s)
Apoptosis/genetics , Microtubule Proteins/genetics , Mutation , Neurons/drug effects , Neurons/metabolism , Paclitaxel/pharmacology , Spiral Ganglion/cytology , Spiral Ganglion/metabolism , Animals , Cell Line , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Mice , Mice, Knockout , Microscopy, Confocal , Neurons/ultrastructure , Spiral Ganglion/ultrastructure
7.
Sci Rep ; 7(1): 7500, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790422

ABSTRACT

The human inner ear has an intricate spiral shape often compared to shells of mollusks, particularly to the nautilus shell. It has inspired many functional hearing theories. The reasons for this complex geometry remain unresolved. We digitized 138 human cochleae at microscopic resolution and observed an astonishing interindividual variability in the shape. A 3D analytical cochlear model was developed that fits the analyzed data with high precision. The cochlear geometry neither matched a proposed function, namely sound focusing similar to a whispering gallery, nor did it have the form of a nautilus. Instead, the innate cochlear blueprint and its actual ontogenetic variants were determined by spatial constraints and resulted from an efficient packing of the cochlear duct within the petrous bone. The analytical model predicts well the individual 3D cochlear geometry from few clinical measures and represents a clinical tool for an individualized approach to neurosensory restoration with cochlear implants.


Subject(s)
Cochlear Duct/anatomy & histology , Models, Statistical , Petrous Bone/anatomy & histology , Spiral Ganglion/anatomy & histology , Spiral Lamina/anatomy & histology , Spiral Ligament of Cochlea/anatomy & histology , Animal Shells/anatomy & histology , Animal Shells/ultrastructure , Animals , Autopsy , Biological Variation, Individual , Cochlear Duct/physiology , Cochlear Duct/ultrastructure , Hearing/physiology , Humans , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/methods , Models, Anatomic , Nautilus/anatomy & histology , Nautilus/ultrastructure , Petrous Bone/physiology , Spiral Ganglion/physiology , Spiral Ganglion/ultrastructure , Spiral Lamina/physiology , Spiral Lamina/ultrastructure , Spiral Ligament of Cochlea/physiology , Spiral Ligament of Cochlea/ultrastructure
8.
J Assoc Res Otolaryngol ; 18(6): 751-769, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28717876

ABSTRACT

Sensorineural hearing loss is commonly caused by damage to cochlear sensory hair cells. Coinciding with hair cell degeneration, the peripheral fibres of type I spiral ganglion neurons (SGNs) that normally form synaptic connections with the inner hair cell gradually degenerate. We examined the time course of these degenerative changes in type I SGNs and their satellite Schwann cells at the ultrastructural level in guinea pigs at 2, 6, and 12 weeks following aminoglycoside-induced hearing loss. Degeneration of the peripheral fibres occurred prior to the degeneration of the type I SGN soma and was characterised by shrinkage of the fibre followed by retraction of the axoplasm, often leaving a normal myelin lumen devoid of axoplasmic content. A statistically significant reduction in the cross-sectional area of peripheral fibres was evident as early as 2 weeks following deafening (p < 0.001, ANOVA). This was followed by a decrease in type I SGN density within Rosenthal's canal that was statistically significant 6 weeks following deafening (p < 0.001, ANOVA). At any time point examined, few type I SGN soma were observed undergoing degeneration, implying that once initiated, soma degeneration was rapid. While there was a significant reduction in soma area as well as changes to the morphology of the soma, the ultrastructure of surviving type I SGN soma appeared relatively normal over the 12-week period following deafening. Satellite Schwann cells exhibited greater survival traits than their type I SGN; however, on loss of neural contact, they reverted to a non-myelinating phenotype, exhibiting an astrocyte-like morphology with the formation of processes that appeared to be searching for new neural targets. In 6- and 12-week deafened cochlea, we observed cellular interaction between Schwann cell processes and residual SGNs that distorted the morphology of the SGN soma. Understanding the response of SGNs, Schwann cells, and the complex relationship between them following aminoglycoside deafening is important if we are to develop effective therapeutic techniques designed to rescue SGNs.


Subject(s)
Deafness/pathology , Organ of Corti/ultrastructure , Schwann Cells/ultrastructure , Spiral Ganglion/ultrastructure , Aminoglycosides , Animals , Deafness/chemically induced , Guinea Pigs
9.
Neurotoxicology ; 62: 151-161, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28655499

ABSTRACT

Carbon disulfide (CS2) is used in industry; it has been shown to have neurotoxic effects, causing central and distal axonopathies.However, it is not considered cochleotoxic as it does not affect hair cells in the organ of Corti, and the only auditory effects reported in the literature were confined to the low-frequency region. No reports on the effects of combined exposure to low-frequency noise and CS2 have been published to date. This article focuses on the effects on rat hearing of combined exposure to noise with increasing concentrations of CS2 (0, 63,250, and 500ppm, 6h per day, 5 days per week, for 4 weeks). The noise used was a low-frequency noise ranging from 0.5 to 2kHz at an intensity of 106dB SPL. Auditory function was tested using distortion product oto-acoustic emissions, which mainly reflects the cochlear performances. Exposure to noise alone caused an auditory deficit in a frequency area ranging from 3.6 to 6 kHz. The damaged area was approximately one octave (6kHz) above the highest frequency of the exposure noise (2.8kHz); it was a little wider than expected based on the noise spectrum.Consequently, since maximum hearing sensitivity is located around 8kHz in rats, low-frequency noise exposure can affect the cochlear regions detecting mid-range frequencies. Co-exposure to CS2 (250-ppm and over) and noise increased the extent of the damaged frequency window since a significant auditory deficit was measured at 9.6kHz in these conditions.Moreover, the significance at 9.6kHz increased with the solvent concentrations. Histological data showed that neither hair cells nor ganglion cells were damaged by CS2. This discrepancy between functional and histological data is discussed. Like most aromatic solvents, carbon disulfide should be considered as a key parameter in hearing conservation régulations.


Subject(s)
Carbon Disulfide/toxicity , Hearing/drug effects , Hearing/radiation effects , Noise/adverse effects , Acoustic Stimulation , Analysis of Variance , Animals , Carbon Disulfide/blood , Dose-Response Relationship, Radiation , Female , Hearing Tests , Microscopy, Atomic Force , Myosins/metabolism , Organ of Corti/drug effects , Organ of Corti/metabolism , Organ of Corti/radiation effects , Organ of Corti/ultrastructure , Rats , Rats, Wistar , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/radiation effects , Spiral Ganglion/ultrastructure , Thiazolidines/urine , Time Factors
10.
J Histochem Cytochem ; 65(3): 173-184, 2017 03.
Article in English | MEDLINE | ID: mdl-28056182

ABSTRACT

In response to toxic stressors, cancer cells defend themselves by mobilizing one or more epidermal growth factor receptor (EGFR) cascades that employ xeroderma pigmentosum-A (XPA) to repair damaged genes. Recent experiments discovered that neurons within the auditory nerve exhibit basal levels of EGFR+XPA co-expression. This finding implied that auditory neurons in particular or neurons in general have the capacity to mobilize an EGFR+XPA defense. Therefore, the current study tested the hypothesis that noise stress would alter the expression pattern of EGFR/XPA within the auditory nerve. Design-based stereology was used to quantify the proportion of neurons that expressed EGFR, XPA, and EGFR+XPA with and without noise stress. The results revealed an intricate neuronal response that is suggestive of alterations to both co-expression and individual expression of EGFR and XPA. In both the apical and middle cochlear coils, the noise stress depleted EGFR+XPA expression. Furthermore, there was a reduction in the proportion of neurons that expressed XPA-alone in the middle coils. However, the noise stress caused a significant increase in the proportion of neurons that expressed EGFR-alone in the middle coils. The basal cochlear coils failed to mobilize a significant response to the noise stress. These results suggest that EGFR and XPA might be part of the molecular defense repertoire of the auditory nerve.


Subject(s)
Cochlear Nerve/physiology , Cochlear Nerve/ultrastructure , ErbB Receptors/analysis , Noise , Stress, Physiological , Xeroderma Pigmentosum Group A Protein/analysis , Animals , Cochlear Nerve/chemistry , ErbB Receptors/metabolism , Immunohistochemistry/methods , Male , Neurons/chemistry , Neurons/metabolism , Neurons/ultrastructure , Rats, Long-Evans , Spiral Ganglion/chemistry , Spiral Ganglion/physiology , Spiral Ganglion/ultrastructure , Xeroderma Pigmentosum Group A Protein/metabolism
11.
Colloids Surf B Biointerfaces ; 149: 105-114, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27736723

ABSTRACT

Cochlear implants (CI) allow for hearing rehabilitation in patients with sensorineural hearing loss or deafness. Restricted CI performance results from the spatial gap between spiral ganglion neurons and the CI, causing current spread that limits spatially restricted stimulation and impairs frequency resolution. This may be substantially improved by guiding peripheral processes of spiral ganglion neurons towards and onto the CI electrode contacts. An injectable, peptide-based hydrogel was developed which may provide a permissive scaffold to facilitate neurite growth towards the CI. To test hydrogel capacity to attract spiral ganglion neurites, neurite outgrowth was quantified in an in vitro model using a custom-designed hydrogel scaffold and PuraMatrix®. Neurite attachment to native hydrogels is poor, but significantly improved by incorporation of brain-derived neurotrophic factor (BDNF), covalent coupling of the bioactive laminin epitope IKVAV and the incorporation a full length laminin to hydrogel scaffolds. Incorporation of full length laminin protein into a novel custom-designed biofunctionalized hydrogel (IKVAV-GGG-SIINFEKL) allows for neurite outgrowth into the hydrogel scaffold. The study demonstrates that peptide-based hydrogels can be specifically biofunctionalized to provide a permissive scaffold to attract neurite outgrowth from spiral ganglion neurons. Such biomaterials appear suitable to bridge the spatial gap between neurons and the CI.


Subject(s)
Hydrogels/pharmacology , Neurites/drug effects , Neuronal Outgrowth/drug effects , Peptides/pharmacology , Spiral Ganglion/drug effects , Tissue Scaffolds , Amino Acid Sequence , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cochlea/drug effects , Cochlea/physiology , Cochlea/ultrastructure , Cochlear Implants , Female , Hydrogels/chemistry , Laminin/metabolism , Laminin/pharmacology , Male , Mice , Neurites/physiology , Neurites/ultrastructure , Neuronal Outgrowth/physiology , Peptides/chemistry , Spiral Ganglion/growth & development , Spiral Ganglion/physiology , Spiral Ganglion/ultrastructure , Tissue Culture Techniques
12.
Nutrients ; 8(8)2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27483303

ABSTRACT

We evaluated the role of iron deficiency (ID) without anemia on hearing function and cochlear pathophysiology of young rats before and after noise exposure. We used rats at developmental stages as an animal model to induce ID without anemia by dietary iron restriction. We have established this dietary restriction model in the rat that should enable us to study the effects of iron deficiency in the absence of severe anemia on hearing and ribbon synapses. Hearing function was measured on Postnatal Day (PND) 21 after induction of ID using auditory brainstem response (ABR). Then, the young rats were exposed to loud noise on PND 21. After noise exposure, hearing function was again measured. We observed the morphology of ribbon synapses, hair cells and spiral ganglion cells (SGCs), and assessed the expression of myosin VIIa, vesicular glutamate transporter 3 and prestin in the cochlea. ID without anemia did not elevate ABR threshold shifts, but reduced ABR wave I peak amplitude of young rats. At 70, 80, and 90 dB SPL, amplitudes of wave I (3.11 ± 0.96 µV, 3.52 ± 1.31 µV, and 4.37 ± 1.08 µV, respectively) in pups from the ID group were decreased compared to the control (5.92 ± 1.67 µV, 6.53 ± 1.70 µV, and 6.90 ± 1.76 µV, respectively) (p < 0.05). Moreover, ID without anemia did not impair the morphology hair cells and SGCs, but decreased the number of ribbon synapses. Before noise exposure, the mean number of ribbon synapses per inner hair cell (IHC) was significantly lower in the ID group (8.44 ± 1.21) compared to that seen in the control (13.08 ± 1.36) (p < 0.05). In addition, the numbers of ribbon synapses per IHC of young rats in the control (ID group) were 6.61 ± 1.59, 3.07 ± 0.83, 5.85 ± 1.63 and 12.25 ± 1.97 (3.75 ± 1.45, 2.03 ± 1.08, 3.81 ± 1.70 and 4.01 ± 1.65) at 1, 4, 7 and 14 days after noise exposure, respectively. Moreover, ABR thresholds at 4 and 8 kHz in young rats from the ID group were significantly elevated at 7 and 14 days after noise exposure compared to control (p < 0.05). The average number of young rat SGCs from the ID group were significantly decreased in the basal turn of the cochlea compared to the control (p < 0.05). Therefore, ID without anemia delayed the recovery from noise-induced hearing loss and ribbon synapses damage, increased SGCs loss, and upregulated prestin after noise exposure. Thus, the cochleae in rat pups with ID without anemia were potentially susceptible to loud noise exposure, and this deficit may be attributed to the reduction of ribbon synapses and SGCs.


Subject(s)
Anemia, Iron-Deficiency/physiopathology , Cochlea/physiopathology , Cochlear Nerve/physiopathology , Disease Models, Animal , Hearing Loss, Noise-Induced/etiology , Nutritional Status , Anemia, Iron-Deficiency/diet therapy , Anemia, Iron-Deficiency/metabolism , Anemia, Iron-Deficiency/pathology , Animals , Auditory Cortex/metabolism , Auditory Cortex/physiopathology , Auditory Cortex/ultrastructure , Auditory Threshold/radiation effects , Brain Stem/metabolism , Brain Stem/physiopathology , Brain Stem/ultrastructure , Cochlea/innervation , Cochlea/metabolism , Cochlea/ultrastructure , Cochlear Nerve/metabolism , Cochlear Nerve/radiation effects , Cochlear Nerve/ultrastructure , Disease Susceptibility , Female , Gene Expression Regulation, Developmental/radiation effects , Hearing Loss, Noise-Induced/prevention & control , Iron, Dietary/therapeutic use , Male , Microscopy, Electron, Scanning , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Noise/adverse effects , Random Allocation , Rats, Sprague-Dawley , Spiral Ganglion/metabolism , Spiral Ganglion/physiopathology , Spiral Ganglion/ultrastructure , Weaning
13.
Neurotox Res ; 29(4): 594-604, 2016 May.
Article in English | MEDLINE | ID: mdl-26913517

ABSTRACT

Mild maternal iron deficiency anemia (IDA) adversely affects the development of cochlear hair cells of the young offspring, but the mechanisms underlying the association are incompletely understood. The aim of this study was to evaluate whether mild maternal IDA in guinea pigs could interrupt inner hair cell (IHC) ribbon synapse density and outer hair cell motility of the offspring. Here, we established a dietary restriction model that allows us to study quantitative changes in the number of IHC ribbon synapses and hearing impairment in response to mild maternal IDA in young guinea pig. The offspring were weaned on postnatal day (PND) 9 and then were given the iron-sufficient diet. On PND 24, pups were examined the hearing function by auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) measurements. Then, the cochleae were harvested for assessment of the number of IHC ribbon synapses by immunofluorescence, the morphology of cochlear hair cells, and spiral ganglion cells (SGCs) by scanning electron microscope and hematoxylin-eosin staining, the location, and expression of vesicular glutamate transporter (VGLUT) 3, myosin VIIa, and prestin by immunofluorescence and blotting. Here, we show that mild maternal IDA in guinea pigs induced elevated ABR threshold shifts, declined DPOAE level shifts, and reduced the number of ribbon synapses, impaired the morphology of cochlear hair cells and SGCs in offsprings. In addition, downregulation of VGLUT3 and myosin VIIa, and upregulation of prestin were observed in the cochlea of offsprings from mild maternal IDA in guinea pigs. These data indicate that mild maternal IDA in guinea pigs induced hearing impairment in offsprings, and this deficit may be attributed to the reduction of ribbon synapse density and dysregulation of VGLUT3, myosin VIIa, and prestin.


Subject(s)
Anemia, Iron-Deficiency/complications , Hearing Loss/etiology , Hearing Loss/pathology , Myosins/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Synapses/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Acoustic Stimulation , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Guinea Pigs , Hair Cells, Auditory, Inner/cytology , Male , Myosin VIIa , Otoacoustic Emissions, Spontaneous , Pregnancy , Psychoacoustics , Spiral Ganglion/cytology , Spiral Ganglion/ultrastructure , Synapses/ultrastructure , Up-Regulation/physiology
14.
Otolaryngol Head Neck Surg ; 154(1): 157-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26378184

ABSTRACT

OBJECTIVE: Profile count method for estimating cell number in sectioned tissue applies a correction factor for double count (resulting from transection during sectioning) of count units selected to represent the cell. For human spiral ganglion cell counts, we attempted to address apparent confusion between published correction factors for nucleus and nucleolus count units that are identical despite the role of count unit diameter in a commonly used correction factor formula. STUDY DESIGN: We examined a portion of human cochlea to empirically derive correction factors for the 2 count units, using 3-dimensional reconstruction software to identify double counts. SETTING: The Neurotology and House Histological Temporal Bone Laboratory at University of California at Los Angeles. SUBJECTS AND METHODS: Using a fully sectioned and stained human temporal bone, we identified and generated digital images of sections of the modiolar region of the lower first turn of cochlea, identified count units with a light microscope, labeled them on corresponding digital sections, and used 3-dimensional reconstruction software to identify double-counted count units. RESULTS: For 25 consecutive sections, we determined that double-count correction factors for nucleus count unit (0.91) and nucleolus count unit (0.92) matched the published factors. We discovered that nuclei and, therefore, spiral ganglion cells were undercounted by 6.3% when using nucleolus count units. CONCLUSION: We determined that correction factors for count units must include an element for undercounting spiral ganglion cells as well as the double-count element. We recommend a correction factor of 0.91 for the nucleus count unit and 0.98 for the nucleolus count unit when using 20-µm sections.


Subject(s)
Cell Nucleus Structures , Spiral Ganglion/cytology , Cell Count , Humans , Middle Aged , Spiral Ganglion/ultrastructure
15.
Acta Otolaryngol ; 135(11): 1093-102, 2015.
Article in English | MEDLINE | ID: mdl-26139555

ABSTRACT

CONCLUSION: Noise exposure can cause a decline in cochlear ribbon synapses and result in consequent hearing loss. The reduction of synaptic puncta appears reversible and may contribute to hearing restoration in mice after noise exposure. OBJECTIVE: To detect whether noise induced reversible changes of cochlear ribbon synapses contribute to temporary hearing loss in C57BL/6J mice. METHODS: The mice were assigned randomly to five groups and exposed to white noise at 110 dB SPL for 2 h except the control group. ABR thresholds were acquired before noise exposure (control), immediately following exposure (Day 0), or on Days 4, 7, or 14 after noise exposure. Light microscopy, scanning emission microscopy, and whole mounts examination was utilized to study whether there is morphology change of outer hair cells (OHC), inner hair cells (IHC), or spiral ganglion cells (SGN) due to the 110 dB white noise. Moreover, experimental approaches, including immunostaining and confocal microcopy, were used to detect whether ribbon synapses were the primary targets of noise-induced temporary hearing loss. RESULT: Exposure to 110 dB white noise for 2 h induced TTS in mice, with the maximal ABR threshold elevations seen on the 4(th) day after noise exposure. There were no significant morphological changes in the cochlea. Paralleled changes of pre-synaptic ribbons in both the number and post-synaptic density (PSDs) during this noise exposure were detected. The number of pre-synaptic ribbon, post-synaptic density (PSDs), and co-localized puncta correlated with the shifts of ABR thresholds. Moreover, a complete recovery of ABR thresholds and synaptic puncta was seen on the 14(th) day after the noise stimulations.


Subject(s)
Auditory Threshold/physiology , Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem/physiology , Hair Cells, Auditory, Inner/ultrastructure , Hair Cells, Auditory, Outer/ultrastructure , Hearing Loss, Noise-Induced/pathology , Acoustic Stimulation/adverse effects , Animals , Cochlea/metabolism , Cochlea/ultrastructure , Disease Models, Animal , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Outer/metabolism , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/physiopathology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Scanning , Neuronal Plasticity , Spiral Ganglion/metabolism , Spiral Ganglion/ultrastructure , Synapses
16.
Neurosci Lett ; 599: 38-42, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26002079

ABSTRACT

Treatment of partial hearing loss with the combined electrical and acoustical stimulation (EAS) aims at restoring the hearing while preserving the residual hearing. The aim of present study was to establish an in vitro system to study the effects of an electrical field on the auditory hair cells and spiral ganglion cells. Cochlear tissues containing the organ of Corti, spiral limbus and spiral ganglion neurons were dissected from post-natal Wistar rats (p3-p5) and cultured in the micro-channels. Electric current was homogenously applied on the apical, medial and basal parts of explants. Biphasic rectangular pulses were applied continuously over a period of 30 h or 42 h and the explants were fixed and stained to visualize the hair cells and neurites. Application of electrical field for 30 h has not induced significant changes in the number of inner or outer hair cells when compared to the control. However, after 42 h of electric stimulation, the number of hair cells decreased significantly by about 30%. The medial and basal fragments were particularly affected. The number of neurites has not been influenced but significant neuritic beading, consistent with neurodegeneration, was observed after 42 h of electric stimulation. Although performed with immature auditory tissues, our findings hint at the possibility of particular electric current inducing damage or loss of auditory hair cells, which should be considered when designing EAS electrodes.


Subject(s)
Electric Stimulation Therapy/adverse effects , Organ of Corti/cytology , Animals , Cell Count , Electric Stimulation/adverse effects , Hair Cells, Auditory/cytology , Rats, Wistar , Spiral Ganglion/ultrastructure , Tissue Culture Techniques
17.
J Neurosci Res ; 93(6): 848-58, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25648717

ABSTRACT

Damaging effects on the cochlea of high-intensity acoustic overexposures have been extensively documented, but only few works have focused on the danger of moderate noise levels. Using scanning and transmission electron microscopy, we explored the noise-induced neuroepithelial changes that occur in the cochlea of rats subjected to moderate intensities, 70 and 85 dB SPL, for an extended period of time (6 hr/day over 3 months). Although the full quota of outer and inner sensory hair cells remained present, we detected discrete abnormalities, likely resulting from metabolic impairment, in both types of hair cell within the basal region of the cochlea. In contrast, important noise-dependent losses of spiral ganglion neurons had occurred. In addition, we found cytoplasmic accumulations of lipofuscin-like aggregates in most of the surviving cochlear neurons. These results strongly suggest that noise levels comparable to those of certain working environments, with sufficient exposure duration, pose a severe risk to the cochlea. Moreover, our data support the notion that long-duration exposure to moderate noise is a causative factor of presbycusis.


Subject(s)
Noise/adverse effects , Vestibulocochlear Nerve Diseases/etiology , Animals , Cell Count , Cochlea/pathology , Cochlea/ultrastructure , Disease Models, Animal , Hair Cells, Auditory/pathology , Hair Cells, Auditory/ultrastructure , Microscopy, Electron , Psychoacoustics , Rats , Rats, Wistar , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Spiral Ganglion/pathology , Spiral Ganglion/ultrastructure , Time Factors , Vestibulocochlear Nerve Diseases/pathology
18.
J Neurosci ; 35(7): 3263-75, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698761

ABSTRACT

Saposin B (Sap B) is an essential activator protein for arylsulfatase A in the hydrolysis of sulfatide, a lipid component of myelin. To study Sap B's role in hearing and balance, a Sap B-deficient (B(-/-)) mouse was evaluated. At both light and electron microscopy (EM) levels, inclusion body accumulation was seen in satellite cells surrounding spiral ganglion (SG) neurons from postnatal month 1 onward, progressing into large vacuoles preceding satellite cell degeneration, and followed by SG degeneration. EM also revealed reduced or absent myelin sheaths in SG neurons from postnatal month 8 onwards. Hearing loss was initially seen at postnatal month 6 and progressed thereafter for frequency-specific stimuli, whereas click responses became abnormal from postnatal month 13 onward. The progressive hearing loss correlated with the accumulation of inclusion bodies in the satellite cells and their subsequent degeneration. Outer hair cell numbers and efferent function measures (distortion product otoacoustic emissions and contralateral suppression) were normal in the B(-/-) mice throughout this period. Alcian blue staining of SGs demonstrated that these inclusion bodies corresponded to sulfatide accumulation. In contrast, changes in the vestibular system were much milder, but caused severe physiologic deficits. These results demonstrate that loss of Sap B function leads to progressive sulfatide accumulation in satellite cells surrounding the SG neurons, leading to satellite cell degeneration and subsequent SG degeneration with a resultant loss of hearing. Relative sparing of the efferent auditory and vestibular neurons suggests that alternate glycosphingolipid metabolic pathways predominate in these other systems.


Subject(s)
Hearing Disorders/etiology , Leukodystrophy, Metachromatic/complications , Leukodystrophy, Metachromatic/genetics , Nerve Degeneration/etiology , Saposins/deficiency , Satellite Cells, Perineuronal/pathology , Spiral Ganglion/pathology , Acoustic Stimulation , Animals , Cell Death/genetics , Cochlea/metabolism , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Functional Laterality , Hearing Tests , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Otoacoustic Emissions, Spontaneous/genetics , Saposins/genetics , Spiral Ganglion/ultrastructure , Swimming/psychology
19.
PLoS One ; 9(9): e108146, 2014.
Article in English | MEDLINE | ID: mdl-25268135

ABSTRACT

Hair cells and spiral ganglion neurons of the mammalian auditory system do not regenerate, and their loss leads to irreversible hearing loss. Aminoglycosides induce auditory hair cell death in vitro, and evidence suggests that phosphatidylinositol-3-kinase/Akt signaling opposes gentamicin toxicity via its downstream target, the protein kinase Akt. We previously demonstrated that somatostatin-a peptide with hormone/neurotransmitter properties-can protect hair cells from gentamicin-induced hair cell death in vitro, and that somatostatin receptors are expressed in the mammalian inner ear. However, it remains unknown how this protective effect is mediated. In the present study, we show a highly significant protective effect of octreotide (a drug that mimics and is more potent than somatostatin) on gentamicin-induced hair cell death, and increased Akt phosphorylation in octreotide-treated organ of Corti explants in vitro. Moreover, we demonstrate that somatostatin receptor-1 knockout mice overexpress somatostatin receptor-2 in the organ of Corti, and are less susceptible to gentamicin-induced hair cell loss than wild-type or somatostatin-1/somatostatin-2 double-knockout mice. Finally, we show that octreotide affects auditory hair cells, enhances spiral ganglion neurite number, and decreases spiral ganglion neurite length.


Subject(s)
Hair Cells, Auditory/metabolism , Hearing Loss/genetics , Receptors, Somatostatin/genetics , Spiral Ganglion/metabolism , Animals , Cell Death/drug effects , Gene Expression Regulation , Gentamicins , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Hearing Loss/chemically induced , Hearing Loss/physiopathology , Hearing Loss/prevention & control , Mice , Mice, Knockout , Neurites/drug effects , Neurites/metabolism , Neurites/ultrastructure , Octreotide/pharmacology , Organ Culture Techniques , Phosphorylation/drug effects , Protective Agents/pharmacology , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Somatostatin/deficiency , Signal Transduction , Spiral Ganglion/drug effects , Spiral Ganglion/ultrastructure
20.
Hear Res ; 309: 103-12, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333928

ABSTRACT

The spiral ganglion, which is primarily composed of spiral ganglion neurons and satellite glial cells, transmits auditory information from sensory hair cells to the central nervous system. Atrial natriuretic peptide (ANP), acting through specific receptors, is a regulatory peptide required for a variety of cardiac, neuronal and glial functions. Although previous studies have provided direct evidence for the presence of ANP and its functional receptors (NPR-A and NPR-C) in the inner ear, their presence within the cochlear spiral ganglion and their regulatory roles during auditory neurotransmission and development is not known. Here we investigated the expression patterns and levels of ANP and its receptors within the cochlear spiral ganglion of the postnatal rat using immunofluorescence and immunoelectron microscopy techniques, reverse transcription-polymerase chain reaction and Western blot analysis. We have demonstrated that ANP and its receptors colocalize in both subtypes of spiral ganglion neurons and in perineuronal satellite glial cells. Furthermore, we have analyzed differential expression levels associated with both mRNA and protein of ANP and its receptors within the rat spiral ganglion during postnatal development. Collectively, our research provides direct evidence for the presence and synthesis of ANP and its receptors in both neuronal and non-neuronal cells within the cochlear spiral ganglion, suggesting possible roles for ANP in modulating neuronal and glial functions, as well as neuron-satellite glial cell communication, within the spiral ganglion during auditory neurotransmission and development.


Subject(s)
Atrial Natriuretic Factor/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Spiral Ganglion/metabolism , Age Factors , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Gene Expression Regulation , Neuroglia/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Atrial Natriuretic Factor/genetics , Spiral Ganglion/cytology , Spiral Ganglion/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...