Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 591
Filter
1.
Hear Res ; 447: 109024, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735179

ABSTRACT

Delayed loss of residual acoustic hearing after cochlear implantation is a common but poorly understood phenomenon due to the scarcity of relevant temporal bone tissues. Prior histopathological analysis of one case of post-implantation hearing loss suggested there were no interaural differences in hair cell or neural degeneration to explain the profound loss of low-frequency hearing on the implanted side (Quesnel et al., 2016) and attributed the threshold elevation to neo-ossification and fibrosis around the implant. Here we re-evaluated the histopathology in this case, applying immunostaining and improved microscopic techniques for differentiating surviving hair cells from supporting cells. The new analysis revealed dramatic interaural differences, with a > 80 % loss of inner hair cells in the cochlear apex on the implanted side, which can account for the post-implantation loss of residual hearing. Apical degeneration of the stria further contributed to threshold elevation on the implanted side. In contrast, spiral ganglion cell survival was reduced in the region of the electrode on the implanted side, but apical counts in the two ears were similar to that seen in age-matched unimplanted control ears. Almost none of the surviving auditory neurons retained peripheral axons throughout the basal half of the cochlea. Relevance to cochlear implant performance is discussed.


Subject(s)
Auditory Threshold , Cochlear Implantation , Cochlear Implants , Spiral Ganglion , Cochlear Implantation/instrumentation , Cochlear Implantation/adverse effects , Humans , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology , Hair Cells, Auditory, Inner/pathology , Time Factors , Cell Survival , Male , Hearing , Hearing Loss/physiopathology , Hearing Loss/pathology , Hearing Loss/surgery , Hearing Loss/etiology , Female , Hair Cells, Auditory/pathology , Aged , Nerve Degeneration , Middle Aged , Temporal Bone/pathology , Temporal Bone/surgery
2.
Sci Rep ; 14(1): 10910, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740884

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling plays a significant role in multiple biological processes, including inflammation, immunity, and cell death. However, its specific impact on the cochlea remains unclear. In this study, we aimed to investigate the effects of TGF-ß signaling suppression on auditory function and cochlear pathology in mice with kanamycin-induced ototoxicity. Kanamycin and furosemide (KM-FS) were systemically administered to 8-week-old C57/BL6 mice, followed by immediate topical application of a TGF-ß receptor inhibitor (TGF-ßRI) onto the round window membrane. Results showed significant TGF-ß receptor upregulation in spiral ganglion neurons (SGNs) after KM-FA ototoxicity, whereas expression levels in the TGF-ßRI treated group remained unchanged. Interestingly, despite no significant change in cochlear TGF-ß expression after KM-FS ototoxicity, TGF-ßRI treatment resulted in a significant decrease in TGF-ß signaling. Regarding auditory function, TGF-ßRI treatment offered no therapeutic effects on hearing thresholds and hair cell survival following KM-FS ototoxicity. However, SGN loss and macrophage infiltration were significantly increased with TGF-ßRI treatment. These results imply that inhibition of TGF-ß signaling after KM-FS ototoxicity promotes cochlear inflammation and SGN degeneration.


Subject(s)
Kanamycin , Mice, Inbred C57BL , Ototoxicity , Signal Transduction , Spiral Ganglion , Transforming Growth Factor beta , Animals , Kanamycin/toxicity , Signal Transduction/drug effects , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Transforming Growth Factor beta/metabolism , Mice , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Cochlea/metabolism , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Furosemide/pharmacology , Male
3.
Hum Mol Genet ; 33(10): 905-918, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38449065

ABSTRACT

Mutations in AIFM1, encoding for apoptosis-inducing factor (AIF), cause AUNX1, an X-linked neurologic disorder with late-onset auditory neuropathy (AN) and peripheral neuropathy. Despite significant research on AIF, there are limited animal models with the disrupted AIFM1 representing the corresponding phenotype of human AUNX1, characterized by late-onset hearing loss and impaired auditory pathways. Here, we generated an Aifm1 p.R450Q knock-in mouse model (KI) based on the human AIFM1 p.R451Q mutation. Hemizygote KI male mice exhibited progressive hearing loss from P30 onward, with greater severity at P60 and stabilization until P210. Additionally, muscle atrophy was observed at P210. These phenotypic changes were accompanied by a gradual reduction in the number of spiral ganglion neuron cells (SGNs) at P30 and ribbons at P60, which coincided with the translocation of AIF into the nucleus starting from P21 and P30, respectively. The SGNs of KI mice at P210 displayed loss of cytomembrane integrity, abnormal nuclear morphology, and dendritic and axonal demyelination. Furthermore, the inner hair cells and myelin sheath displayed abnormal mitochondrial morphology, while fibroblasts from KI mice showed impaired mitochondrial function. In conclusion, we successfully generated a mouse model recapitulating AUNX1. Our findings indicate that disruption of Aifm1 induced the nuclear translocation of AIF, resulting in the impairment in the auditory pathway.


Subject(s)
Apoptosis Inducing Factor , Cell Nucleus , Disease Models, Animal , Animals , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Mice , Humans , Cell Nucleus/metabolism , Cell Nucleus/genetics , Male , Mutation , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss/metabolism , Gene Knock-In Techniques , Protein Transport , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/metabolism
4.
Hear Res ; 442: 108935, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38113793

ABSTRACT

Sound information is transduced from mechanical vibration to electrical signals in the cochlea, conveyed to and further processed in the brain to form auditory perception. During the process, spiral ganglion neurons (SGNs) are the key cells that connect the peripheral and central auditory systems by receiving information from hair cells in the cochlea and transmitting it to neurons of the cochlear nucleus (CN). Decades of research in the cochlea greatly improved our understanding of SGN function under normal and pathological conditions, especially about the roles of different subtypes of SGNs and their peripheral synapses. However, it remains less clear how SGN central terminals or auditory nerve (AN) synapses connect to CN neurons, and ultimately how peripheral pathology links to structural alterations and functional deficits in the central auditory nervous system. This review discusses recent progress about the morphological and physiological properties of different subtypes of AN synapses and associated postsynaptic CN neurons, their changes during aging, and the potential mechanisms underlying age-related hearing loss.


Subject(s)
Cochlear Nucleus , Hearing Loss , Humans , Cochlear Nucleus/pathology , Cochlear Nerve , Neurons/pathology , Synapses/pathology , Spiral Ganglion/pathology , Cochlea/physiology
5.
Hear Res ; 439: 108895, 2023 11.
Article in English | MEDLINE | ID: mdl-37837701

ABSTRACT

The auditory nerve typically degenerates following loss of cochlear hair cells or synapses. In the case of hair cell loss neural degeneration hinders restoration of hearing through a cochlear implant, and in the case of synaptopathy suprathreshold hearing is affected, potentially degrading speech perception in noise. It has been established that neurotrophins such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) can mitigate auditory nerve degeneration. Several potential BDNF mimetics have also been investigated for neurotrophic effects in the cochlea. A recent in vitro study showed favorable effects of M3, a TrkB monoclonal antibody agonist, when compared with BDNF. In the present study we set out to examine the effect of M3 on auditory nerve preservation in vivo. Thirty-one guinea pigs were bilaterally deafened, and unilaterally treated with a single 3-µl dose of 7 mg/ml, 0.7 mg/ml M3 or vehicle-only by means of a small gelatin sponge two weeks later. During the experiment and analyses the experimenters were blinded to the three treatment groups. Four weeks after treatment, we assessed the treatment effect (1) histologically, by quantifying survival of SGCs and their peripheral processes (PPs); and (2) electrophysiologically, with two different paradigms of electrically evoked compound action potential (eCAP) recordings shown to be indicative of neural health: single-pulse stimulation with varying inter-phase gap (IPG), and pulse-train stimulation with varying inter-pulse interval. We observed a consistent and significant preservative effect of M3 on SGC survival in the lower basal turn (approximately 40% more survival than in the untreated contralateral cochlea), but also in the upper middle and lower apical turn of the cochlea. This effect was similar for the two treatment groups. Survival of PPs showed a trend similar to that of the SGCs, but was only significantly higher for the highest dose of M3. The protective effect of M3 on SGCs was not reflected in any of the eCAP measures: no statistically significant differences were observed between groups in IPG effect nor between the M3 treatment groups and the control group using the pulse-train stimulation paradigm. In short, while a clear effect of M3 was observed on SGC survival, this was not clearly translated into functional preservation.


Subject(s)
Cochlear Implants , Deafness , Guinea Pigs , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Spiral Ganglion/pathology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cochlear Nerve , Hearing , Cochlea
6.
Lab Anim ; 57(6): 631-641, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37070340

ABSTRACT

Auditory disabilities have a large impact on the human population worldwide. Research into understanding and treating hearing disabilities has increased significantly in recent years. One of the most relevant animal species in this context is the guinea pig, which has to be deafened to study several of the hearing pathologies and develop novel therapies. Applying kanamycin subcutaneously and furosemide intravenously is a long-established method in hearing research, leading to permanent hearing loss without surgical intervention at the ear. The intravenous application of furosemide requires invasive surgery in the cervical area of the animals to expose the jugular vein, since a relatively large volume (1 ml per 500 g body weight) must be injected over a period of about 2.5 min. We have established a gentler alternative by applying the furosemide by puncture of the leg veins. For this, custom-made cannula-needle devices were built to allow the vein puncture and subsequent slow injection of the furosemide. This approach was tested in 11 guinea pigs through the foreleg via the cephalic antebrachial vein and through the hind leg via the saphenous vein. Frequency-specific hearing thresholds were measured before and after the procedure to verify normal hearing and successful deafening, respectively. The novel approach of systemic deafening was successfully implemented in 10 out of 11 animals. The Vena saphena was best suited to the application. Since the animals' condition, post leg vein application, was better in comparison to animals deafened by exposure of the Vena jugularis, the postulated refinement that reduced animal stress was deemed successful.


Subject(s)
Furosemide , Hearing Loss, Sensorineural , Humans , Guinea Pigs , Animals , Furosemide/adverse effects , Kanamycin/adverse effects , Spiral Ganglion/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/pathology , Hearing , Disease Models, Animal
7.
J Neurosci Methods ; 391: 109852, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37031766

ABSTRACT

BACKGROUND: Hearing impairment is a rising public health issue, and current therapeutics fail to restore normal auditory sensation. Animal models are essential to a better understanding of the pathophysiology of deafness and developing therapeutics to restore hearing. NEW METHODS: Wild-type CBA/CaJ neonatal mice P2-5 were used in this study. Neomycin suspension (500 nl of 50 or 100 mg/ml) was micro-injected into the endolymphatic space. Cochlear morphology was examined 3 and 7 days after injection; hair cell (HC) loss, supporting cell morphology, and neurite denervation pattern were assessed with whole-mounts. At 2 and 4 weeks post-injection, the spiral ganglion neuron (SGN) density was analyzed with cryostat sections. Audiometric responses were measured with auditory brain response (ABR) at 4 weeks. RESULTS: Rapid and complete degeneration of the inner and outer HCs occurred as early as 3 days post-injection. Subsequently, time- and dose-dependent degeneration patterns were observed along the axis of the cochlear membranous labyrinth forming a flat epithelium. Likewise, the SGN histology demonstrated significant cell density reduction at 2 and 4 weeks. The ABR threshold measurements confirmed profound deafness at 4 weeks. COMPARISON WITH EXISTING METHODS: Compared to previously described local and systemic aminoglycoside injections, this method provides a reliable, robust, and rapid deafening model with a single infusion of neomycin in neonatal mice. This model also allows for investigating the effects of inner ear damage during auditory maturation. CONCLUSIONS: A single injection of neomycin into the endolymphatic space induces robust HC loss and denervation in neonatal mice.


Subject(s)
Deafness , Neomycin , Animals , Mice , Neomycin/toxicity , Animals, Newborn , Deafness/chemically induced , Mice, Inbred CBA , Cochlea , Spiral Ganglion/pathology , Evoked Potentials, Auditory, Brain Stem
8.
Neurosci Lett ; 793: 136990, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36455693

ABSTRACT

Cytomegalovirus (CMV)-induced sensorineural hearing loss (SNHL) is a worldwide epidemic. Recent studies have shown that the degree of spiral ganglion neuron (SGN) loss is correlated with hearing loss after CMV infection. We aimed to better understand the pathological mechanisms of CMV-related SGN death and to search for intervention measures. We found that both apoptosis and pyroptosis are involved in CMV-induced SGN death, which may be caused by the simultaneous activation of the p53/JNK and NLRP3/caspase-1 signaling pathways, respectively. Moreover, considering that mixed lineage kinase family (MLK1/2/3) are host restriction factors against viral infection and upstream regulators of the p53/JNK and inflammatory (including NLRP3-caspase1) signaling pathways, we further demonstrated that the MLKs inhibitor URMC-099 exhibited a protective effect against CMV-induced SGN death and hearing loss. These results indicate that MLKs signaling may be a key regulator and promising novel target for preventing apoptosis and even pyroptosis during the CMV infection of SGN cells and for treating hearing loss.


Subject(s)
Cytomegalovirus Infections , Deafness , Hearing Loss, Sensorineural , MAP Kinase Kinase Kinases , Muromegalovirus , Animals , Mice , Apoptosis , Cytomegalovirus , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/pathology , Deafness/metabolism , Deafness/pathology , Hearing Loss/metabolism , Hearing Loss/pathology , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Neurons , NLR Family, Pyrin Domain-Containing 3 Protein , Spiral Ganglion/pathology , Tumor Suppressor Protein p53 , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinase Kinase 11
9.
J Vis Exp ; (187)2022 09 28.
Article in English | MEDLINE | ID: mdl-36282698

ABSTRACT

Deafness is the most common sensory impairment, affecting approximately 5% or 430 million people worldwide as per the World Health Organization1. Aging or presbycusis is a primary cause of sensorineural hearing loss and is characterized by damage to hair cells, spiral ganglion neurons (SGNs), and the stria vascularis. These structures reside within the cochlea, which has a complex, spiral-shaped anatomy of membranous tissues suspended in fluid and surrounded by bone. These properties make it technically difficult to investigate and quantify histopathological changes. To address this need, we developed a light-sheet microscope (TSLIM) that can image and digitize the whole cochlea to facilitate the study of structure-function relationships in the inner ear. Well-aligned serial sections of the whole cochlea result in a stack of images for three-dimensional (3D) volume rendering and segmentation of individual structures for 3D visualization and quantitative analysis (i.e., length, width, surface, volume, and number). Cochleae require minimal processing steps (fixation, decalcification, dehydration, staining, and optical clearing), all of which are compatible with subsequent high-resolution imaging by scanning and transmission electron microscopy. Since all the tissues are present in the stacks, each structure can be assessed individually or relative to other structures. In addition, since imaging uses fluorescent probes, immunohistochemistry and ligand binding can be used to identify specific structures and their 3D volume or distribution within the cochlea. Here we used TSLIM to examine cochleae from aged mice to quantify the loss of hair cells and spiral ganglion neurons. In addition, advanced analyses (e.g., cluster analysis) were used to visualize local reductions of spiral ganglion neurons in Rosenthal's canal along its 3D volume. These approaches demonstrate TSLIM microscopy's ability to quantify structure-function relationships within and between cochleae.


Subject(s)
Cochlea , Fluorescent Dyes , Mice , Animals , Ligands , Cochlea/diagnostic imaging , Spiral Ganglion/diagnostic imaging , Spiral Ganglion/pathology , Microscopy, Fluorescence , Aging/pathology
10.
Hear Res ; 426: 108633, 2022 12.
Article in English | MEDLINE | ID: mdl-36288662

ABSTRACT

CHARGE syndrome is a multiple anomaly developmental disorder characterized by a variety of sensory deficits, including sensorineural hearing loss of unknown etiology. Most cases of CHARGE are caused by heterozygous pathogenic variants in CHD7, the gene encoding Chromodomain DNA-binding Protein 7 (CHD7), a chromatin remodeler important for the development of neurons and glial cells. Previous studies in the Chd7Gt/+ mouse model of CHARGE syndrome showed substantial neuron loss in the early stages of the developing inner ear that are compensated for by mid-gestation. In this study, we sought to determine if early developmental delays caused by Chd7 haploinsufficiency affect neurons, glial cells, and inner hair cell innervation in the mature cochlea. Analysis of auditory brainstem response recordings in Chd7Gt/+ adult animals showed elevated thresholds at 4 kHz and 16 kHz, but no differences in ABR Wave I peak latency or amplitude compared to wild type controls. Proportions of neurons in the Chd7Gt/+ adult spiral ganglion and densities of nerve projections from the spiral ganglion to the organ of Corti were not significantly different from wild type controls. Inner hair cell synapse formation also appeared unaffected in mature Chd7Gt/+ cochleae. However, histological analysis of adult Chd7Gt/+ cochleae revealed diminished satellite glial cells and hypermyelinated Type I spiral ganglion axons. We characterized the expression of CHD7 in developing inner ear glia and found CHD7 to be expressed during a tight window of inner ear development at the Schwann cell precursor stage at E9.5. While cochlear neurons appear to differentiate normally in the setting of Chd7 haploinsufficiency, our results suggest an important role for CHD7 in glial cells in the inner ear. This study highlights the dynamic nature of CHD7 activity during inner ear development in mice and contributes to understanding CHARGE syndrome pathology.


Subject(s)
CHARGE Syndrome , Ear, Inner , Mice , Animals , Spiral Ganglion/pathology , CHARGE Syndrome/genetics , CHARGE Syndrome/pathology , Chromatin , Ear, Inner/pathology , Neuroglia , DNA-Binding Proteins/genetics
11.
Aging Cell ; 21(10): e13708, 2022 10.
Article in English | MEDLINE | ID: mdl-36088647

ABSTRACT

Age-related hearing loss (ARHL) is the most prevalent sensory deficit in the elderly. This progressive pathology often has psychological and medical comorbidities, including social isolation, depression, and cognitive decline. Despite ARHL's enormous societal and economic impact, no therapies to prevent or slow its progression exist. Loss of synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs), a.k.a. IHC synaptopathy, is an early event in cochlear aging, preceding neuronal and hair cell loss. To determine if age-related IHC synaptopathy can be prevented, and if this impacts the time-course of ARHL, we tested the effects of cochlear overexpression of neurotrophin-3 (Ntf3) starting at middle age. We chose Ntf3 because this neurotrophin regulates the formation of IHC-SGN synapses in the neonatal period. We now show that triggering Ntf3 overexpression by IHC supporting cells starting in middle age rapidly increases the amplitude of sound-evoked neural potentials compared with age-matched controls, indicating that Ntf3 produces a positive effect on cochlear function when the pathology is minimal. Furthermore, near the end of their lifespan, Ntf3-overexpressing mice have milder ARHL, with larger sound-evoked potentials along the ascending auditory pathway and reduced IHC synaptopathy compared with age-matched controls. Our results also provide evidence that an age-related decrease in cochlear Ntf3 expression contributes to ARHL and that Ntf3 supplementation could serve as a therapeutic for this prevalent disorder. Furthermore, these findings suggest that factors that regulate synaptogenesis during development could prevent age-related synaptopathy in the brain, a process involved in several central nervous system degenerative disorders.


Subject(s)
Hair Cells, Auditory, Inner , Hearing Loss , Animals , Cochlea/pathology , Evoked Potentials, Auditory, Brain Stem/physiology , Mice , Spiral Ganglion/pathology , Synapses/pathology
12.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(4): 637-641, 2022 Jul.
Article in Chinese | MEDLINE | ID: mdl-35871734

ABSTRACT

Objective: To explore the effect of changes in the expression level of necorsis factor (NF)-κB/inducible nitric oxide synthase (iNOS) signaling pathway on hearing loss in a mouse model of sensorineural hearing loss (SNHL) induced by 3-nitropropionic acid (3-NP). Methods: The animal model was established by tympanic injection. C57BL/6 male mice were divided into three groups, 3-NP group receiving tympanic injection of 3-NP solution, 3-NP+EVP4593 group receiving tympanic injection of 3-NP solution and intraperitoneal injection of EVP4593 solution, and a control group receiving tympanic injection of phosphate buffered saline (PBS). Auditory brainstem response (ABR) was tested before and after injection. After 4 weeks, the cochlea was harvested and immunohistochemistry and qRT-PCR of NF-κB p65, RelB, iNOS, and Caspase-3 were conducted accordingly. Results: The hearing thresholds of the 3-NP group were higher than those of the control group and the 3-NP+EVP4593 group ( P<0.05), and the hearing thresholds of the 3-NP+EVP4593 group were also higher than those of the control group ( P<0.05). Immunofluorescence staining and qRT-PCR results showed that 3-NP exposure caused an increase in the expressions of NF-κB p65, RelB, and iNOS in the spiral ganglion in comparison with those of the control group ( P<0.05), and their expressions decreased with the administration of EVP4593 ( P<0.05). The expression of Caspase-3 in the spiral ganglion cells in the 3-NP group was higher than that in the control group, while in the 3-NP+EVP4593 group, it was lower than that in the 3-NP group ( P<0.05). Conclusion: This study found that, by activating the NF-κB/iNOS signaling pathway, 3-NP may cause inflammation in the spiral ganglion of the cochlear in the SNHL model mice, which may play an important role in the pathogenesis of SNHL.


Subject(s)
Hearing Loss, Sensorineural , Spiral Ganglion , Animals , Caspase 3 , Disease Models, Animal , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B , Nitric Oxide Synthase Type II , Signal Transduction , Spiral Ganglion/pathology , Spiral Ganglion/physiology
13.
Hear Res ; 418: 108458, 2022 05.
Article in English | MEDLINE | ID: mdl-35334332

ABSTRACT

Hearing loss in patients with vestibular schwannoma (VS) is commonly attributed to mechanical compression of the auditory nerve, though recent studies suggest that this retrocochlear pathology may be augmented by cochlear damage. Although VS-associated loss of inner hair cells, outer hair cells, and spiral ganglion cells has been reported, it is unclear to what extent auditory-nerve peripheral axons are damaged in VS patients. Understanding the degree of damage VSs cause to auditory nerve fibers (ANFs) is important for accurately modeling clinical outcomes of cochlear implantation, which is a therapeutic option to rehabilitate hearing in VS-affected ears. A retrospective analysis of human temporal-bone histopathology was performed on archival specimens from the Massachusetts Eye and Ear collection. Seven patients met our inclusion criteria based on the presence of sporadic, unilateral, untreated VS. Tangential sections of five cochlear regions were stained with hematoxylin and eosin, and adjacent sections were stained to visualize myelinated ANFs and efferent fibers. Following confocal microscopy, peripheral axons of ANFs within the osseous spiral lamina were quantified manually, where feasible, and with a "pixel counting" method, applicable to all sections. ANF density was substantially reduced on the VS side compared to the unaffected contralateral side. In the upper basal turn, a significant difference between the VS side and unaffected contralateral side was found using both counting methods, corresponding to the region tuned to 2000 Hz. Even spiral ganglion cells (SGCs) contralateral to VS were affected by the tumor as the majority of contralateral SGC counts were below average for age. This observation provides histological insight into the clinical observation that unilateral vestibular schwannomas pose a long-term risk of progression of hearing loss in the contralateral ear as well. Our pixel counting method for ANF quantification in the osseous spiral lamina is applicable to other pathologies involving sensorineural hearing loss. Future research is needed to classify ANFs into morphological categories, accurately predict their electrical properties, and use this knowledge to inform optimal cochlear implant programming strategies.


Subject(s)
Deafness , Hearing Loss , Neuroma, Acoustic , Humans , Cochlear Nerve/pathology , Deafness/pathology , Hearing Loss/pathology , Neuroma, Acoustic/pathology , Retrospective Studies , Spiral Ganglion/pathology , Spiral Lamina
14.
Hear Res ; 414: 108404, 2022 02.
Article in English | MEDLINE | ID: mdl-34883366

ABSTRACT

It is generally believed that the efficacy of cochlear implants is partly dependent on the condition of the stimulated neural population. Cochlear pathology is likely to affect the manner in which neurons respond to electrical stimulation, potentially resulting in differences in perception of electrical stimuli across cochlear implant recipients and across the electrode array in individual cochlear implant users. Several psychophysical and electrophysiological measures have been shown to predict cochlear health in animals and were used to assess conditions near individual stimulation sites in humans. In this study, we examined the relationship between psychophysical strength-duration functions and spiral ganglion neuron density in two groups of guinea pigs with cochlear implants who had minimally-overlapping cochlear health profiles. One group was implanted in a hearing ear (N = 10) and the other group was deafened by cochlear perfusion of neomycin, inoculated with an adeno-associated viral vector with an Ntf3-gene insert (AAV.Ntf3) and implanted (N = 14). Psychophysically measured strength-duration functions for both monopolar and tripolar electrode configurations were then compared for the two treatment groups. Results were also compared to their histological outcomes. Overall, there were considerable differences between the two treatment groups in terms of their psychophysical performance as well as the relation between their functional performance and histological data. Animals in the neomycin-deafened, neurotrophin-treated, and implanted group (NNI) exhibited steeper strength-duration function slopes; slopes were positively correlated with SGN density (steeper slopes in animals that had higher SGN densities). In comparison, the implanted hearing (IH) group had shallower slopes and there was no relation between slopes and spiral ganglion density. Across all animals, slopes were negatively correlated with ensemble spontaneous activity levels (shallower slopes with higher ensemble spontaneous activity levels). We hypothesize that differences in strength-duration function slopes between the two treatment groups were related to the condition of the inner hair cells, which generate spontaneous activity that could affect the across-fiber synchrony and/or the size of the population of neural elements responding to electrical stimulation. In addition, it is likely that spiral ganglion neuron peripheral processes were present in the IH group, which could affect membrane properties of the stimulated neurons. Results suggest that the two treatment groups exhibited distinct patterns of variation in conditions near the stimulating electrodes that altered detection thresholds. Overall, the results of this study suggest a complex relationship between psychophysical detection thresholds for cochlear implant stimulation and nerve survival in the implanted cochlea. This relationship seems to depend on the characteristics of the electrical stimulus, the electrode configuration, and other biological features of the implanted cochlea such as the condition of the inner hair cells and the peripheral processes.


Subject(s)
Cochlear Implantation , Cochlear Implants , Deafness , Animals , Cochlea/physiology , Cochlear Implantation/methods , Electric Stimulation , Guinea Pigs , Hearing/physiology , Spiral Ganglion/pathology
15.
Med Sci Monit ; 27: e933278, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34657931

ABSTRACT

BACKGROUND Sodium salicylate (SS) induces excitotoxicity of spiral ganglion neurons (SGNs) by inhibiting the response of γ-aminobutyric acid type A receptors (GABAARs). Our previous studies have shown that SS can increase the internalization of GABAARs on SGNs, which involves dopamine D1-like receptors (D1Rs) and related signaling pathways. In this study, we aimed to explore the role of D1Rs and their downstream molecule protein kinase C (PKC) in the process of SS inhibiting GABAARs. MATERIAL AND METHODS The expression of D1Rs and GABARγ2 on rat cochlear SGNs cultured in vitro was tested by immunofluorescence. Then, the SGNs were exposed to SS, D1R agonist (SKF38393), D1R antagonist (SCH23390), clathrin/dynamin-mediated endocytosis inhibitor (dynasore), and PKC inhibitor (Bisindolylmaleimide I). Western blotting and whole-cell patch clamp technique were used to assess the changes of surface and total protein of GABARγ2 and GABA-activated currents. RESULTS Immunofluorescence showed that D1 receptors (DRD1) were expressed on SGNs. Data from western blotting showed that SS promoted the internalization of cell surface GABAARs, and activating D1Rs had the same result. Inhibiting D1Rs and PKC decreased the internalization of GABAARs. Meanwhile, the phosphorylation level of GABAARγ2 S327 affected by PKC was positively correlated with the degree of internalization of GABAARs. Moreover, whole-cell patch clamp recording showed that inhibition of D1Rs or co-inhibition of D1Rs and PKC attenuated the inhibitory effect of SS on GABA-activated currents. CONCLUSIONS D1Rs mediate the GABAAR internalization induced by SS via a PKC-dependent manner and participate in the excitotoxic process of SGNs.


Subject(s)
Ototoxicity/pathology , Protein Kinase C/metabolism , Receptors, Dopamine D1/metabolism , Receptors, GABA-A/metabolism , Sodium Salicylate/adverse effects , Spiral Ganglion/pathology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines , Cells, Cultured , Disease Models, Animal , Female , Humans , Hydrazones/pharmacology , Male , Models, Animal , Neurons/drug effects , Neurons/metabolism , Ototoxicity/etiology , Patch-Clamp Techniques , Primary Cell Culture , Rats , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Spiral Ganglion/cytology , Spiral Ganglion/drug effects
16.
Curr Med Sci ; 41(4): 680-686, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34403092

ABSTRACT

OBJECTIVE: Age-related hearing loss (AHL), characterized by degeneration of cochlea structures, is the most common sensory disorder among the elderly worldwide. The calcium channel is considered to contribute to normal hearing. However, the role of the T-type voltage-activated calcium channel, Cav3.1, remains unclear in AHL. Here, we investigate the age-related change of Cav3.1 expression in the cochlea and D-gal-induced senescent HEI-OC1 cells. METHODS: Cochleae from C57BL/6 mice at 2 months and 12 months of age were assessed. Senescence in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells was induced by D-gal treatment. The immunofluorescence technique was employed to investigate the distribution of Cav3.1 in vivo and in vitro. Quantitative assessment was achieved by Western blotting and real-time PCR. RESULTS: In comparison with 2-month-old animals, 12-month old C57BL/6 mice exhibited great loss of hair cells and elevated auditory brainstem threshold. The Cav3.1 was located in hair cells, spiral ganglion cells, lateral walls, and the expression of Cav3.1 protein and mRNA decreased in the aged cochleae. D-gal-induced senescence assay confirmed the down-regulation of Cav3.1 expression in senescent HEI-OC1 cells. CONCLUSION: Our results show that age-related down-regulated expression of Cav3.1 in the cochleae is associated with AHL and may contribute to the pathogenesis of AHL.


Subject(s)
Calcium Channels, T-Type/genetics , Cochlea/metabolism , Presbycusis/genetics , Animals , Cochlea/diagnostic imaging , Cochlea/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Humans , Mice , Organ of Corti/diagnostic imaging , Organ of Corti/metabolism , Organ of Corti/pathology , Presbycusis/pathology , Spiral Ganglion/diagnostic imaging , Spiral Ganglion/metabolism , Spiral Ganglion/pathology
17.
Int J Mol Sci ; 22(12)2021 Jun 13.
Article in English | MEDLINE | ID: mdl-34199197

ABSTRACT

In the cochlea, non-sensory supporting cells are directly connected to adjacent supporting cells via gap junctions that allow the exchange of small molecules. We have previously shown that the pharmacological regulation of gap junctions alleviates cisplatin (CDDP)-induced ototoxicity in animal models. In this study, we aimed to identify specific small molecules that pass through gap junctions in the process of CDDP-induced auditory cell death and suggest new mechanisms to prevent hearing loss. We found that the cyclic adenosine monophosphate (cAMP) inducer forskolin (FSK) significantly attenuated CDDP-induced auditory cell death in vitro and ex vivo. The activation of cAMP/PKA/CREB signaling was observed in organ of Corti primary cells treated with FSK, especially in supporting cells. Co-treatment with gap junction enhancers such as all-trans retinoic acid (ATRA) and quinoline showed potentiating effects with FSK on cell survival via activation of cAMP/PKA/CREB. In vivo, the combination of FSK and ATRA was more effective for preventing ototoxicity compared to either single treatment. Our study provides the new insight that gap junction-mediated intercellular communication of cAMP may prevent CDDP-induced ototoxicity.


Subject(s)
Cell Communication , Cisplatin/adverse effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Gap Junctions/metabolism , Ototoxicity/metabolism , Signal Transduction , A549 Cells , Animals , Cell Communication/drug effects , Cell Death/drug effects , Colforsin/pharmacology , Colforsin/therapeutic use , Connexin 26/metabolism , Gap Junctions/drug effects , Hair Cells, Auditory/metabolism , HeLa Cells , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Hearing Loss/prevention & control , Humans , Mice , Protective Agents/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Spiral Ganglion/drug effects , Spiral Ganglion/pathology , Tretinoin/pharmacology , Tretinoin/therapeutic use
18.
Aging (Albany NY) ; 13(9): 12587-12606, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33909598

ABSTRACT

The aging of the population and environmental noise have contributed to high rates of presbycusis, also known as age-related hearing loss (ARHL). Because mice have a relatively short life span, murine models have not been suitable for determining the mechanism of presbycusis development and methods of diagnosis. Although the common marmoset, a non-human primate (NHP), is an ideal animal model for studying age-related diseases, its auditory spectrum has not been systematically studied. Auditory brainstem responses (ABRs) from 38 marmosets of different ages demonstrated that auditory function correlated with age. Hearing loss in geriatric common marmosets started at ultra-high frequency (>16 kHz), then extended to lower frequencies. Despite age-related deterioration of ABR threshold and amplitude in marmosets, outer hair cell (OHC) function remained stable at all ages. Spiral ganglion neurons (SGNs), which are the first auditory neurons in the auditory system, were found to degenerate distinctly in aged common marmosets, indicating that neural degeneration caused presbycusis in these animals. Similarly, age-associated ABR deterioration without loss of OHC function was observed in another NHP, rhesus monkeys. Audiometry results from these two species of NHP suggested that NHPs were ideal for studying ARHL and that neural presbycusis at high frequency may be prevalent in primates.


Subject(s)
Aging/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing/physiology , Spiral Ganglion/pathology , Animals , Hair Cells, Auditory/pathology , Macaca mulatta , Nerve Degeneration/pathology
19.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33903231

ABSTRACT

The cochlea of our auditory system is an intricate structure deeply embedded in the temporal bone. Compared with other sensory organs such as the eye, the cochlea has remained poorly accessible for investigation, for example, by imaging. This limitation also concerns the further development of technology for restoring hearing in the case of cochlear dysfunction, which requires quantitative information on spatial dimensions and the sensorineural status of the cochlea. Here, we employed X-ray phase-contrast tomography and light-sheet fluorescence microscopy and their combination for multiscale and multimodal imaging of cochlear morphology in species that serve as established animal models for auditory research. We provide a systematic reference for morphological parameters relevant for cochlear implant development for rodent and nonhuman primate models. We simulate the spread of light from the emitters of the optical implants within the reconstructed nonhuman primate cochlea, which indicates a spatially narrow optogenetic excitation of spiral ganglion neurons.


Subject(s)
Cochlea/diagnostic imaging , Cochlear Implantation , Hearing Loss, Sensorineural/therapy , Neurons/metabolism , Animals , Cochlea/pathology , Cochlear Implants , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/physiopathology , Humans , Neurons/pathology , Optogenetics , Spiral Ganglion/diagnostic imaging , Spiral Ganglion/pathology
20.
Cell Mol Neurobiol ; 41(1): 31-42, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32180095

ABSTRACT

We examined the functional and structural changes of auditory neurons (ANs) in adult mice after conductive hearing loss (CHL). Earplugs (EPs) were bilaterally inserted in male 8-week-old mice for 4 weeks [EP(+) group] and subsequently removed for 4 weeks [EP(+/-) group]. We examined the control mice [EP(-) group] with no EPs inserted at 12 weeks. The auditory brainstem response (ABR) was measured to determine the cochlear function before and after EP insertion, after EP removal, and at 4 weeks following EP removal. We examined the cochleae for hair cell (HC) and spiral ganglion neuron survival, synaptic and neural properties, and AN myelination. There was a significant elevation of the ABR threshold across all tested frequencies after EP insertion. After removing the occlusion, these threshold shifts were fully recovered. Compared with the EP(-) mice, the EP(+) mice showed a significant decrease in the ABR peak 1 amplitude and a significantly prolonged latency at all tested frequencies. There was no significant effect of auditory deprivation on the survival of HCs and ANs. Conversely, auditory deprivation caused significant damage to the synapses and myelin and a significant decrease in the AN size. Although functional changes in the ABR amplitude and latency did not fully recover in the EP(+/-) mice, almost all anatomical changes were fully recovered in the EP(+/-) mice; however, cochlear synapses only showed partial recovery. These results suggest that auditory activities are required to maintain peripheral auditory synapses and myelination in adults. The auditory deprivation model allows for assessment of the mechanisms of synaptopathy and demyelination in the auditory periphery, and synaptic and myelin regeneration in sensorineural hearing loss.


Subject(s)
Aging/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Conductive/pathology , Hearing Loss, Conductive/physiopathology , Nerve Degeneration/physiopathology , Neuronal Plasticity , Alcohol Oxidoreductases/metabolism , Animals , Co-Repressor Proteins/metabolism , Evoked Potentials, Auditory, Brain Stem , Male , Mice, Inbred C57BL , Myelin Sheath/pathology , Nerve Degeneration/pathology , Nerve Fibers/pathology , Receptors, AMPA/metabolism , Spiral Ganglion/pathology , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...