Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 531
Filter
Add more filters

Publication year range
1.
Cell ; 163(6): 1348-59, 2015 12 03.
Article in English | MEDLINE | ID: mdl-26627734

ABSTRACT

Spontaneous electrical activity of neurons in developing sensory systems promotes their maturation and proper connectivity. In the auditory system, spontaneous activity of cochlear inner hair cells (IHCs) is initiated by the release of ATP from glia-like inner supporting cells (ISCs), facilitating maturation of central pathways before hearing onset. Here, we find that ATP stimulates purinergic autoreceptors in ISCs, triggering Cl(-) efflux and osmotic cell shrinkage by opening TMEM16A Ca(2+)-activated Cl(-) channels. Release of Cl(-) from ISCs also forces K(+) efflux, causing transient depolarization of IHCs near ATP release sites. Genetic deletion of TMEM16A markedly reduces the spontaneous activity of IHCs and spiral ganglion neurons in the developing cochlea and prevents ATP-dependent shrinkage of supporting cells. These results indicate that supporting cells in the developing cochlea have adapted a pathway used for fluid secretion in other organs to induce periodic excitation of hair cells.


Subject(s)
Ear, Inner/growth & development , Hair Cells, Auditory/cytology , Adenosine Triphosphate/metabolism , Animals , Anoctamin-1 , Chloride Channels/genetics , Chloride Channels/metabolism , Ear, Inner/cytology , Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/metabolism , Mice , Mice, Knockout , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Spiral Ganglion/cytology , Spiral Ganglion/metabolism
2.
PLoS Biol ; 22(6): e3002665, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38935589

ABSTRACT

Loss of synapses between spiral ganglion neurons and inner hair cells (IHC synaptopathy) leads to an auditory neuropathy called hidden hearing loss (HHL) characterized by normal auditory thresholds but reduced amplitude of sound-evoked auditory potentials. It has been proposed that synaptopathy and HHL result in poor performance in challenging hearing tasks despite a normal audiogram. However, this has only been tested in animals after exposure to noise or ototoxic drugs, which can cause deficits beyond synaptopathy. Furthermore, the impact of supernumerary synapses on auditory processing has not been evaluated. Here, we studied mice in which IHC synapse counts were increased or decreased by altering neurotrophin 3 (Ntf3) expression in IHC supporting cells. As we previously showed, postnatal Ntf3 knockdown or overexpression reduces or increases, respectively, IHC synapse density and suprathreshold amplitude of sound-evoked auditory potentials without changing cochlear thresholds. We now show that IHC synapse density does not influence the magnitude of the acoustic startle reflex or its prepulse inhibition. In contrast, gap-prepulse inhibition, a behavioral test for auditory temporal processing, is reduced or enhanced according to Ntf3 expression levels. These results indicate that IHC synaptopathy causes temporal processing deficits predicted in HHL. Furthermore, the improvement in temporal acuity achieved by increasing Ntf3 expression and synapse density suggests a therapeutic strategy for improving hearing in noise for individuals with synaptopathy of various etiologies.


Subject(s)
Hair Cells, Auditory, Inner , Neurotrophin 3 , Synapses , Animals , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Synapses/metabolism , Synapses/physiology , Neurotrophin 3/metabolism , Neurotrophin 3/genetics , Mice , Auditory Threshold , Evoked Potentials, Auditory/physiology , Reflex, Startle/physiology , Auditory Perception/physiology , Spiral Ganglion/metabolism , Female , Male , Hearing Loss, Hidden
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 , Disease Models, Animal , Hearing Loss , Animals , Humans , Male , Mice , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Cell Nucleus/metabolism , Cell Nucleus/genetics , Gene Knock-In Techniques , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/metabolism , Mutation , Protein Transport , Spiral Ganglion/metabolism , Spiral Ganglion/pathology
4.
EMBO Rep ; 24(9): e56702, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37477166

ABSTRACT

Cochlear inner hair cells (IHCs) form specialized ribbon synapses with spiral ganglion neurons that tirelessly transmit sound information at high rates over long time periods with extreme temporal precision. This functional specialization is essential for sound encoding and is attributed to a distinct molecular machinery with unique players or splice variants compared to conventional neuronal synapses. Among these is the active zone (AZ) scaffold protein piccolo/aczonin, which is represented by its short splice variant piccolino at cochlear and retinal ribbon synapses. While the function of piccolo at synapses of the central nervous system has been intensively investigated, the role of piccolino at IHC synapses remains unclear. In this study, we characterize the structure and function of IHC synapses in piccolo gene-trap mutant rats (Pclogt/gt ). We find a mild hearing deficit with elevated thresholds and reduced amplitudes of auditory brainstem responses. Ca2+ channel distribution and ribbon morphology are altered in apical IHCs, while their presynaptic function seems to be unchanged. We conclude that piccolino contributes to the AZ organization in IHCs and is essential for normal hearing.


Subject(s)
Hair Cells, Auditory, Inner , Neuropeptides , Rats , Animals , Hearing/physiology , Synapses/physiology , Cochlea , Spiral Ganglion/metabolism , Cytoskeletal Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 119(48): e2203935119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36409884

ABSTRACT

The afferent innervation of the cochlea is comprised of spiral ganglion neurons (SGNs), which are characterized into four subtypes (Type 1A, B, and C and Type 2). However, little is known about the factors and/or processes that determine each subtype. Here, we present a transcriptional analysis of approximately 5,500 single murine SGNs collected across four developmental time points. All four subtypes are transcriptionally identifiable prior to the onset of coordinated spontaneous activity, indicating that the initial specification process is under genetic control. Trajectory analysis indicates that SGNs initially split into two precursor types (Type 1A/2 and Type 1B/C), followed by subsequent splits to give rise to four transcriptionally distinct subtypes. Differential gene expression, pseudotime, and regulon analyses were used to identify candidate transcription factors which may regulate the subtypes specification process. These results provide insights into SGN development and comprise a transcriptional atlas of SGN maturation across the prenatal period.


Subject(s)
Neurons , Spiral Ganglion , Pregnancy , Female , Mice , Animals , Spiral Ganglion/metabolism , Neurons/metabolism , Cochlea/metabolism
6.
Hum Genet ; 143(8): 979-993, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39066985

ABSTRACT

Gasdermin E (GSDME), a member of the gasdermin protein family, is associated with post-lingual hearing loss. All GSDME pathogenic mutations lead to skipping exon 8; however, the molecular mechanisms underlying hearing loss caused by GSDME mutants remain unclear. GSDME was recently identified as one of the mediators of programmed cell death, including apoptosis and pyroptosis. Therefore, in this study, we injected mice with GSDME mutant (MT) and examined the expression levels to assess its effect on hearing impairment. We observed loss of hair cells in the organ of Corti and spiral ganglion neurons. Further, the N-terminal release from the GSDME mutant in HEI-OC1 cells caused pyroptosis, characterized by cell swelling and rupture of the plasma membrane, releasing lactate dehydrogenase and cytokines such as interleukin-1ß. We also observed that the N-terminal release from GSDME mutants could permeabilize the mitochondrial membrane, releasing cytochromes and activating the mitochondrial apoptotic pathway, thereby generating possible positive feedback on the cleavage of GSDME. Furthermore, we found that treatment with disulfiram or dimethyl fumarate might inhibit pyroptosis and apoptosis by inhibiting the release of GSDME-N from GSDME mutants. In conclusion, this study elucidated the molecular mechanism associated with hearing loss caused by GSDME gene mutations, offering novel insights for potential treatment strategies.


Subject(s)
Apoptosis , Pyroptosis , Pyroptosis/genetics , Animals , Mice , Gain of Function Mutation , Hearing Loss/genetics , Hearing Loss/pathology , Humans , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Organ of Corti/metabolism , Organ of Corti/pathology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Gasdermins
7.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791192

ABSTRACT

The synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) are the most vulnerable structures in the noise-exposed cochlea. Cochlear synaptopathy results from the disruption of these synapses following noise exposure and is considered the main cause of poor speech understanding in noisy environments, even when audiogram results are normal. Cochlear synaptopathy leads to the degeneration of SGNs if damaged IHC-SGN synapses are not promptly recovered. Oxidative stress plays a central role in the pathogenesis of cochlear synaptopathy. C-Phycocyanin (C-PC) has antioxidant and anti-inflammatory activities and is widely utilized in the food and drug industry. However, the effect of the C-PC on noise-induced cochlear damage is unknown. We first investigated the therapeutic effect of C-PC on noise-induced cochlear synaptopathy. In vitro experiments revealed that C-PC reduced the H2O2-induced generation of reactive oxygen species in HEI-OC1 auditory cells. H2O2-induced cytotoxicity in HEI-OC1 cells was reduced with C-PC treatment. After white noise exposure for 3 h at a sound pressure of 118 dB, the guinea pigs intratympanically administered 5 µg/mL C-PC exhibited greater wave I amplitudes in the auditory brainstem response, more IHC synaptic ribbons and more IHC-SGN synapses according to microscopic analysis than the saline-treated guinea pigs. Furthermore, the group treated with C-PC had less intense 4-hydroxynonenal and intercellular adhesion molecule-1 staining in the cochlea compared with the saline group. Our results suggest that C-PC improves cochlear synaptopathy by inhibiting noise-induced oxidative stress and the inflammatory response in the cochlea.


Subject(s)
Cochlea , Intercellular Adhesion Molecule-1 , Noise , Oxidative Stress , Phycocyanin , Synapses , Animals , Oxidative Stress/drug effects , Guinea Pigs , Phycocyanin/pharmacology , Phycocyanin/therapeutic use , Cochlea/metabolism , Cochlea/drug effects , Cochlea/pathology , Synapses/drug effects , Synapses/metabolism , Noise/adverse effects , Intercellular Adhesion Molecule-1/metabolism , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/pathology , Reactive Oxygen Species/metabolism , Male , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Hydrogen Peroxide/metabolism , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Antioxidants/pharmacology , Cell Line , Hearing Loss, Hidden
8.
Dev Dyn ; 252(1): 124-144, 2023 01.
Article in English | MEDLINE | ID: mdl-36284453

ABSTRACT

BACKGROUND: Proper connectivity between type I spiral ganglion neurons (SGNs) and inner hair cells (IHCs) in the cochlea is necessary for conveying sound information to the brain in mammals. Previous studies have shown that type I SGNs are heterogeneous in form, function and synaptic location on IHCs, but factors controlling their patterns of connectivity are not well understood. RESULTS: During development, cochlear supporting cells and SGNs express Semaphorin-3A (SEMA3A), a known axon guidance factor. Mice homozygous for a point mutation that attenuates normal SEMA3A repulsive activity (Sema3aK108N ) show cochleae with grossly normal patterns of IHC innervation. However, genetic sparse labeling and three-dimensional reconstructions of individual SGNs show that cochleae from Sema3aK108N mice lacked the normal synaptic distribution of type I SGNs. Additionally, Sema3aK108N cochleae show a disrupted distribution of GLUA2 postsynaptic patches around the IHCs. The addition of SEMA3A-Fc to postnatal cochleae led to increases in SGN branching, similar to the effects of inhibiting glutamate receptors. Ca2+ imaging studies show that SEMA3A-Fc decreases SGN activity. CONCLUSIONS: Contrary to the canonical view of SEMA3A as a guidance ligand, our results suggest SEMA3A may regulate SGN excitability in the cochlea, which may influence the morphology and synaptic arrangement of type I SGNs.


Subject(s)
Hair Cells, Auditory , Semaphorin-3A , Animals , Mice , Cochlea/metabolism , Neurons/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Spiral Ganglion/metabolism
9.
J Med Genet ; 59(12): 1219-1226, 2022 12.
Article in English | MEDLINE | ID: mdl-35961784

ABSTRACT

BACKGROUND: It is well established that biallelic mutations in transmembrane protease, serine 3 (TMPRSS3) cause hearing loss. Currently, there is controversy regarding the audiological outcomes after cochlear implantation (CI) for TMPRSS3-associated hearing loss. This controversy creates confusion among healthcare providers regarding the best treatment options for individuals with TMPRSS3-related hearing loss. METHODS: A literature review was performed to identify all published cases of patients with TMPRSS3-associated hearing loss who received a CI. CI outcomes of this cohort were compared with published adult CI cohorts using postoperative consonant-nucleus-consonant (CNC) word performance. TMPRSS3 expression in mouse cochlea and human auditory nerves (HAN) was determined by using hybridisation chain reaction and single-cell RNA-sequencing analysis. RESULTS: In aggregate, 27 patients (30 total CI ears) with TMPRSS3-associated hearing loss treated with CI, and 85% of patients reported favourable outcomes. Postoperative CNC word scores in patients with TMPRSS3-associated hearing loss were not significantly different than those seen in adult CI cohorts (8 studies). Robust Tmprss3 expression occurs throughout the mouse organ of Corti, the spindle and root cells of the lateral wall and faint staining within <5% of the HAN, representing type II spiral ganglion neurons. Adult HAN express negligible levels of TMPRSS3. CONCLUSION: The clinical features after CI and physiological expression of TMPRSS3 suggest against a major role of TMPRSS3 in auditory neurons.


Subject(s)
Cochlear Implantation , Deafness , Hearing Loss , Adult , Humans , Mice , Animals , Spiral Ganglion/metabolism , Serine Endopeptidases/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Deafness/genetics , Hearing Loss/genetics , Neurons/metabolism
10.
J Neurosci ; 41(13): 2930-2943, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33574178

ABSTRACT

Cochlear outer hair cells (OHCs) are known to uniquely participate in auditory processing through their electromotility, and like inner hair cells, are also capable of releasing vesicular glutamate onto spiral ganglion (SG) neurons: in this case, onto the sparse Type II SG neurons. However, unlike glutamate signaling at the inner hair cell-Type I SG neuron synapse, which is robust across a wide spectrum of sound intensities, glutamate signaling at the OHC-Type II SG neuron synapse is weaker and has been hypothesized to occur only at intense, possibly damaging sound levels. Here, we tested the ability of the OHC-Type II SG pathway to signal to the brain in response to moderate, nondamaging sound (80 dB SPL) as well as to intense sound (115 dB SPL). First, we determined the VGluTs associated with OHC signaling and then confirmed the loss of glutamatergic synaptic transmission from OHCs to Type II SG neurons in KO mice using dendritic patch-clamp recordings. Next, we generated genetic mouse lines in which vesicular glutamate release occurs selectively from OHCs, and then assessed c-Fos expression in the cochlear nucleus in response to sound. From these analyses, we show, for the first time, that glutamatergic signaling at the OHC-Type II SG neuron synapse is capable of activating cochlear nucleus neurons, even at moderate sound levels.SIGNIFICANCE STATEMENT Evidence suggests that cochlear outer hair cells (OHCs) release glutamate onto Type II spiral ganglion neurons only when exposed to loud sound, and that Type II neurons are activated by tissue damage. Knowing whether moderate level sound, without tissue damage, activates this pathway has functional implications for this fundamental auditory pathway. We first determined that OHCs rely largely on VGluT3 for synaptic glutamate release. We then used a genetically modified mouse line in which OHCs, but not inner hair cells, release vesicular glutamate to demonstrate that moderate sound exposure activates cochlear nucleus neurons via the OHC-Type II spiral ganglion pathway. Together, these data indicate that glutamate signaling at the OHC-Type II afferent synapse participates in auditory function at moderate sound levels.


Subject(s)
Acoustic Stimulation/methods , Cochlear Nucleus/metabolism , Glutamic Acid/metabolism , Hair Cells, Auditory, Outer/metabolism , Neurons/metabolism , Spiral Ganglion/metabolism , Afferent Pathways/metabolism , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Auditory Pathways/metabolism , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
Hum Mol Genet ; 29(9): 1520-1536, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32337552

ABSTRACT

Here we define a ~200 Kb genomic duplication in 2p14 as the genetic signature that segregates with postlingual progressive sensorineural autosomal dominant hearing loss (HL) in 20 affected individuals from the DFNA58 family, first reported in 2009. The duplication includes two entire genes, PLEK and CNRIP1, and the first exon of PPP3R1 (protein coding), in addition to four uncharacterized long non-coding (lnc) RNA genes and part of a novel protein-coding gene. Quantitative analysis of mRNA expression in blood samples revealed selective overexpression of CNRIP1 and of two lncRNA genes (LOC107985892 and LOC102724389) in all affected members tested, but not in unaffected ones. Qualitative analysis of mRNA expression identified also fusion transcripts involving parts of PPP3R1, CNRIP1 and an intergenic region between PLEK and CNRIP1, in the blood of all carriers of the duplication, but were heterogeneous in nature. By in situ hybridization and immunofluorescence, we showed that Cnrip1, Plek and Ppp3r1 genes are all expressed in the adult mouse cochlea including the spiral ganglion neurons, suggesting changes in expression levels of these genes in the hearing organ could underlie the DFNA58 form of deafness. Our study highlights the value of studying rare genomic events leading to HL, such as copy number variations. Further studies will be required to determine which of these genes, either coding proteins or non-coding RNAs, is or are responsible for DFNA58 HL.


Subject(s)
Blood Proteins/genetics , Calcineurin/genetics , Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Phosphoproteins/genetics , Adolescent , Adult , Animals , Calcineurin/blood , Child , Chromosome Duplication/genetics , Chromosomes, Human, Pair 2/genetics , DNA Copy Number Variations/genetics , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Genome, Human/genetics , Hearing Loss, Sensorineural/blood , Hearing Loss, Sensorineural/pathology , Heterozygote , Humans , Male , Membrane Proteins/blood , Mice , Middle Aged , Neurons/metabolism , Neurons/pathology , Phosphoproteins/blood , RNA, Messenger/blood , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Young Adult
12.
EMBO J ; 37(24)2018 12 14.
Article in English | MEDLINE | ID: mdl-30396994

ABSTRACT

Optogenetic tools, providing non-invasive control over selected cells, have the potential to revolutionize sensory prostheses for humans. Optogenetic stimulation of spiral ganglion neurons (SGNs) in the ear provides a future alternative to electrical stimulation used in cochlear implants. However, most channelrhodopsins do not support the high temporal fidelity pertinent to auditory coding because they require milliseconds to close after light-off. Here, we biophysically characterized the fast channelrhodopsin Chronos and revealed a deactivation time constant of less than a millisecond at body temperature. In order to enhance neural expression, we improved its trafficking to the plasma membrane (Chronos-ES/TS). Following efficient transduction of SGNs using early postnatal injection of the adeno-associated virus AAV-PHPB into the mouse cochlea, fiber-based optical stimulation elicited optical auditory brainstem responses (oABR) with minimal latencies of 1 ms, thresholds of 5 µJ and 100 µs per pulse, and sizable amplitudes even at 1,000 Hz of stimulation. Recordings from single SGNs demonstrated good temporal precision of light-evoked spiking. In conclusion, efficient virus-mediated expression of targeting-optimized Chronos-ES/TS achieves ultrafast optogenetic control of neurons.


Subject(s)
Channelrhodopsins/biosynthesis , Dependovirus , Gene Expression , Neurons/metabolism , Optogenetics , Spiral Ganglion/metabolism , Transduction, Genetic , Animals , Brain Stem/metabolism , Channelrhodopsins/genetics , Evoked Potentials, Auditory , HEK293 Cells , Humans , Mice , Rats , Rats, Wistar
13.
Biochem Biophys Res Commun ; 632: 69-75, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36206596

ABSTRACT

Autosomal recessive nonsyndromic auditory neuropathy is attributed to a genetic etiology. We identified a compound heterozygous missense variant, c.G736A (p.G246S) and c.C2954T (p.T985 M) in TNN of affected patients in a pedigree via candidate gene screening and exome sequencing. To determine the genetic etiology of deafness in the pedigree with a heterozygous missense variant in the gene TNN encoding tenascin-W associated with autosomal recessive nonsyndromic auditory neuropathy, the cochlear expression of tenascin-W was evaluated at mRNA and protein levels in mice, and Tnn knock out mice were generated and utilized to study the function of Tnn in the auditory system. Immunofluorescence stainings showed that tenascin-W was mainly expressed in the somatic cytoplasm of spiral ganglion neurons of mice. Homozygous Tnn knockout was lethal in mice, whereas Tnn heterozygous mice showed decreases in spiral ganglion neuron density and progressive hearing loss. We demonstrate that tenascin-W is expressed in the murine cochleae and is essential for the development of spiral ganglion neurons. An abnormal expression of tenascin-W can influence the development and function of SGNs and affect the function of the auditory system.


Subject(s)
Hearing Loss, Central , Tenascin , Animals , Mice , Hearing Loss, Central/genetics , Hearing Loss, Central/metabolism , RNA, Messenger/metabolism , Spiral Ganglion/metabolism , Tenascin/genetics , Tenascin/metabolism , Humans
14.
Stem Cells ; 39(4): 375-388, 2021 04.
Article in English | MEDLINE | ID: mdl-33378797

ABSTRACT

Hearing loss is the most widely spread sensory disorder in our society. In the majority of cases, it is caused by the loss or malfunctioning of cells in the cochlea: the mechanosensory hair cells, which act as primary sound receptors, and the connecting auditory neurons of the spiral ganglion, which relay the signal to upper brain centers. In contrast to other vertebrates, where damage to the hearing organ can be repaired through the activity of resident cells, acting as tissue progenitors, in mammals, sensory cell damage or loss is irreversible. The understanding of gene and cellular functions, through analysis of different animal models, has helped to identify causes of disease and possible targets for hearing restoration. Translation of these findings to novel therapeutics is, however, hindered by the lack of cellular assays, based on human sensory cells, to evaluate the conservation of molecular pathways across species and the efficacy of novel therapeutic strategies. In the last decade, stem cell technologies enabled to generate human sensory cell types in vitro, providing novel tools to study human inner ear biology, model disease, and validate therapeutics. This review focuses specifically on two technologies: directed differentiation of pluripotent stem cells and direct reprogramming of somatic cell types to sensory hair cells and neurons. Recent development in the field are discussed as well as how these tools could be implemented to become routinely adopted experimental models for hearing research.


Subject(s)
Cell Differentiation/genetics , Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Hair Cells, Auditory/cytology , Hearing Loss/therapy , Spiral Ganglion/cytology , Animals , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Hair Cells, Auditory/metabolism , Hearing/physiology , Hearing Loss/genetics , Hearing Loss/metabolism , Hearing Loss/pathology , Humans , Mechanotransduction, Cellular , Organoids/cytology , Organoids/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Spiral Ganglion/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Proc Natl Acad Sci U S A ; 116(18): 9084-9093, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30975754

ABSTRACT

Encoding the wide range of audible sounds in the mammalian cochlea is collectively achieved by functionally diverse type I spiral ganglion neurons (SGNs) at each tonotopic position. The firing of each SGN is thought to be driven by an individual active zone (AZ) of a given inner hair cell (IHC). These AZs present distinct properties according to their position within the IHC, to some extent forming a gradient between the modiolar and the pillar IHC side. In this study, we investigated whether signaling involved in planar polarity at the apical surface can influence position-dependent AZ properties at the IHC base. Specifically, we tested the role of Gαi proteins and their binding partner LGN/Gpsm2 implicated in cytoskeleton polarization and hair cell (HC) orientation along the epithelial plane. Using high and superresolution immunofluorescence microscopy as well as patch-clamp combined with confocal Ca2+ imaging we analyzed IHCs in which Gαi signaling was blocked by Cre-induced expression of the pertussis toxin catalytic subunit (PTXa). PTXa-expressing IHCs exhibited larger CaV1.3 Ca2+-channel clusters and consequently greater Ca2+ influx at the whole-cell and single-synapse levels, which also showed a hyperpolarized shift of activation. Moreover, PTXa expression collapsed the modiolar-pillar gradients of ribbon size and maximal synaptic Ca2+ influx. Finally, genetic deletion of Gαi3 and LGN/Gpsm2 also disrupted the modiolar-pillar gradient of ribbon size. We propose a role for Gαi proteins and LGN in regulating the position-dependent AZ properties in IHCs and suggest that this signaling pathway contributes to setting up the diverse firing properties of SGNs.


Subject(s)
Cell Polarity/physiology , Hair Cells, Auditory, Inner/metabolism , Synapses/metabolism , Animals , Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Cochlea/metabolism , Female , Hair Cells, Auditory/metabolism , Hair Cells, Auditory, Inner/physiology , Hair Cells, Vestibular/metabolism , Hearing/physiology , Immunohistochemistry/instrumentation , Male , Mice , Mice, Knockout , Patch-Clamp Techniques/methods , Sound , Spiral Ganglion/metabolism , Synapses/physiology
16.
Cell Tissue Res ; 386(2): 239-247, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34155579

ABSTRACT

Ischemia-reperfusion injury (I/R)-induced inflammatory process can mediate cochlea damage-related hearing loss; whether cochlear spiral ganglion progenitor cell-derived exosomes (CSGPC-exos) play a protective role by carrying functional microRNAs into recipient cells is unknown. Different doses of CSGPC-exos (0.1 µg/µl, 0.2 µg/µl, 0.5 µg/µl, 1.0 µg/µl) were administrated into the cochleae of the I/R group induced with 30-min occlusion of the bilateral vertebral arteries and sham surgery group. The speech-evoked auditory brainstem response (ABR) test was utilized to estimate the auditory threshold shift. The relative expression of proinflammatory cytokines was detected with RT-PCR and Western blot, while parvalbumin and caspase-3 expression were detected by immunofluorescence staining in the cochleae. The relative expression of microRNAs (miR-21-5p, miR-26a-5p, and miR-181a-5p) was detected in the cochleae. I/R significantly up-regulated ABR threshold shift and promoted hair cell apoptosis indicated by parvalbumin and caspase-3 staining, while CSGPC-exos (0.5 µg/µl, 1.0 µg/µl) could diminish such damages with downregulated proinflammatory factors (IL-6, IL-1ß, TNF-α, and Cox-2) and upregulated anti-inflammatory miRNAs (miR-21-5p, miR-26a-5p, and miR-181a-5p) expression in the cochleae. CSGPC-exos could protect cochleae damage from I/R, probably via inhibiting the inflammatory process.


Subject(s)
Cochlea/pathology , Exosomes/transplantation , Reperfusion Injury/therapy , Spiral Ganglion/cytology , Stem Cells/cytology , Animals , Cells, Cultured , Female , Mice, Inbred C57BL , MicroRNAs/analysis , Reperfusion Injury/pathology , Spiral Ganglion/metabolism , Stem Cells/metabolism
17.
FASEB J ; 34(3): 4764-4782, 2020 03.
Article in English | MEDLINE | ID: mdl-32027432

ABSTRACT

Damage or degeneration of inner ear spiral ganglion neurons (SGNs) causes hearing impairment. Previous in vitro studies indicate that cochlear glial cells can be reprogrammed into SGNs, however, it remains unknown whether this can occur in vivo. Here, we show that neonatal glial cells can be converted, in vivo, into SGNs (defined as new SGNs) by simultaneous induction of Neurog1 (Ngn1) and Neurod1. New SGNs express SGN markers, Tuj1, Map2, Prox1, Mafb and Gata3, and reduce glial cell marker Sox10 and Scn7a. The heterogeneity within new SGNs is illustrated by immunostaining and transcriptomic assays. Transcriptomes analysis indicates that well reprogrammed SGNs are similar to type I SGNs. In addition, reprogramming efficiency is positively correlated with the dosage of Ngn1 and Neurod1, but declined with aging. Taken together, our in vivo data demonstrates the plasticity of cochlear neonatal glial cells and the capacity of Ngn1 and Neurod1 to reprogram glial cells into SGNs. Looking ahead, we expect that combination of Neurog1 and Neurod1 along with other factors will further boost the percentage of fully converted (Mafb+/Gata3+) new SGNs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Spiral Ganglion/cytology , Spiral Ganglion/metabolism , Animals , Base Sequence , Fluorescent Antibody Technique , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Tubulin/genetics , Tubulin/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
Cell Biol Int ; 45(12): 2490-2498, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34435719

ABSTRACT

Age-related hearing loss (ARHL) is the most prevalent sensory deficit in the elderly, but its mechanism remains unclear. Scaffold protein prohibitin 2 (PHB2) has been widely involved in aging and neurodegeneration. However, the role of PHB2 in ARHL is undeciphered to date. To investigate the expression pattern and the role of PHB2 in ARHL, we used C57BL/6 mice and HEI-OC1 cell line as models. In our study, we have found PHB2 exists in the cochlea and is expressed in hair cells, spiral ganglion neurons, and HEI-OC1 cells. In mice with ARHL, mitophagy is reduced and correspondingly the expression level of PHB2 is decreased. Moreover, after H2 O2 treatment the mitophagy is activated and the PHB2 expression is increased. These findings indicate that PHB2 may exert an important role in ARHL through mitophagy. Findings from this study will be helpful for elucidating the mechanism underlying the ARHL and for providing a new target for ARHL treatment.


Subject(s)
Aging/metabolism , Cochlea/metabolism , Hearing Loss/metabolism , Prohibitins/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitophagy/physiology , Neurons/metabolism , Presbycusis/metabolism , Spiral Ganglion/metabolism
19.
J Neurosci ; 39(33): 6425-6438, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31209173

ABSTRACT

During nervous system development, axons often undergo elaborate changes in branching patterns before circuits have achieved their mature patterns of innervation. In the auditory system, type I spiral ganglion neurons (SGNs) project their peripheral axons into the cochlear epithelium and then undergo a process of branch refinement before forming synapses with sensory hair cells. Here, we report that Semaphorin-5B (Sema5B) acts as an important mediator of this process. During cochlear development in mouse, immature hair cells express Sema5B, whereas the SGNs express both PlexinA1 and PlexinA3, which are known Sema5B receptors. In these studies, genetic sparse labeling and three-dimensional reconstruction techniques were leveraged to determine the morphologies of individual type I SGNs after manipulations of Sema5B signaling. Treating cultured mouse cochleae with Sema5B-Fc (to activate Plexin-As) led to type I SGNs with less numerous, but longer terminal branches. Conversely, cochleae from Sema5b knock-out mice showed type I SGNs with more numerous, but shorter terminal branches. In addition, conditional loss of Plxna1 in SGNs (using Bhlhb5Cre) led to increased type I SGN branching, suggesting that PlexinA1 normally responds to Sema5B in this process. In these studies, mice of either sex were used. The data presented here suggest that Sema5B-PlexinA1 signaling limits SGN terminal branch numbers without causing axonal repulsion, which is a role that distinguishes Sema5B from other Semaphorins in cochlear development.SIGNIFICANCE STATEMENT The sensorineural components of the cochlea include hair cells, which respond mechanically to sound waves, and afferent spiral ganglion neurons (SGNs), which respond to glutamate released by hair cells and transmit auditory information into the CNS. An important component of synapse formation in the cochlea is a process of SGN "debranching" whereby SGNs lose extraneous branches before developing unramified bouton endings that contact the hair cells. In this work, we have found that the transmembrane ligand Semaphorin-5B and its receptor PlexinA1 regulate the debranching process. The results in this report provide new knowledge regarding the molecular control of cochlear afferent innervation.


Subject(s)
Neurogenesis/physiology , Neurons/metabolism , Semaphorins/metabolism , Spiral Ganglion/embryology , Animals , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Spiral Ganglion/metabolism
20.
J Neurosci ; 39(27): 5284-5298, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31085606

ABSTRACT

Acoustic signals are relayed from the ear to the brain via spiral ganglion neurons (SGNs) that receive auditory information from the cochlear inner hair cells (IHCs) and transmit that information to the cochlear nucleus of the brainstem. Physiologically distinct classes of SGNs have been characterized by their spontaneous firing rate and responses to sound and those physiological distinctions are thought to correspond to stereotyped synaptic positions on the IHC. More recently, single-cell profiling has identified multiple groups of SGNs based on transcriptional profiling; however, correlations between any of these groups and distinct neuronal physiology have not been determined. In this study, we show that expression of the POU (Pit-Oct-Unc) transcription factor Pou4f1 in type I SGNs in mice of both sexes correlates with a synaptic location on the modiolar side of IHCs. Conditional deletion of Pou4f1 in SGNs beginning in mice at embryonic day 13 rescues the early path-finding and apoptotic phenotypes reported for germline deletion of Pou4f1, resulting in a phenotypically normal development of SGN patterning. However, conditional deletion of Pou4f1 in SGNs alters the activation of Ca2+ channels in IHCs primarily by increasing their voltage sensitivity. Moreover, the modiolar to pillar gradient of active zone Ca2+ influx strength is eliminated. These results demonstrate that a subset of modiolar-targeted SGNs retain expression of Pou4f1 beyond the onset of hearing and suggest that this transcription factor plays an instructive role in presynaptic Ca2+ signaling in IHCs.SIGNIFICANCE STATEMENT Physiologically distinct classes of type I spiral ganglion neurons (SGNs) are necessary to encode sound intensities spanning the audible range. Although anatomical studies have demonstrated structural correlates for some physiologically defined classes of type I SGNs, an understanding of the molecular pathways that specify each type is only now emerging. Here, we demonstrate that expression of the transcription factor Pou4f1 corresponds to a distinct subgroup of type I SGNs that synapse on the modiolar side of inner hair cells. The conditional deletion of Pou4f1 after SGN formation does not disrupt ganglion size or morphology, change the distribution of IHC synaptic locations, or affect the creation of synapses, but it does influence the voltage dependence and strength of Ca2+ influx at presynaptic active zones in inner hair cells.


Subject(s)
Calcium Signaling , Hearing/physiology , Neurons/metabolism , Presynaptic Terminals/metabolism , Spiral Ganglion/metabolism , Transcription Factor Brn-3A/metabolism , Animals , Evoked Potentials, Auditory, Brain Stem , Female , Hair Cells, Auditory, Inner , Male , Mice, Inbred C57BL , Spiral Ganglion/cytology
SELECTION OF CITATIONS
SEARCH DETAIL