Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Front Neurol ; 15: 1356614, 2024.
Article in English | MEDLINE | ID: mdl-38638308

ABSTRACT

Tmc1 and Tmc2 are essential pore-forming subunits of mechanosensory transduction channels localized to the tips of stereovilli in auditory and vestibular hair cells of the inner ear. To investigate expression and function of Tmc1 and Tmc2 in vestibular organs, we used quantitative polymerase chain reaction (qPCR), fluorescence in situ hybridization - hairpin chain reaction (FISH-HCR), immunostaining, FM1-43 uptake and we measured vestibular evoked potentials (VsEPs) and vestibular ocular reflexes (VORs). We found that Tmc1 and Tmc2 showed dynamic developmental changes, differences in regional expression patterns, and overall expression levels which differed between the utricle and saccule. These underlying changes contributed to unanticipated phenotypic loss of VsEPs and VORs in Tmc1 KO mice. In contrast, Tmc2 KO mice retained VsEPs despite the loss of the calcium buffering protein calretinin, a characteristic biomarker of mature striolar calyx-only afferents. Lastly, we found that neonatal Tmc1 gene replacement therapy is sufficient to restore VsEP in Tmc1 KO mice for up to six months post-injection.

2.
3.
Cells ; 11(24)2022 12 09.
Article in English | MEDLINE | ID: mdl-36552747

ABSTRACT

Peroxisome Biogenesis Disorders (PBD) and Zellweger syndrome spectrum disorders (ZSD) are rare genetic multisystem disorders that include hearing impairment and are associated with defects in peroxisome assembly, function, or both. Mutations in 13 peroxin (PEX) genes have been found to cause PBD-ZSD with ~70% of patients harboring mutations in PEX1. Limited research has focused on the impact of peroxisomal disorders on auditory function. As sensory hair cells are particularly vulnerable to metabolic changes, we hypothesize that mutations in PEX1 lead to oxidative stress affecting hair cells of the inner ear, subsequently resulting in hair cell degeneration and hearing loss. Global deletion of the Pex1 gene is neonatal lethal in mice, impairing any postnatal studies. To overcome this limitation, we created conditional knockout mice (cKO) using Gfi1Creor VGlut3Cre expressing mice crossed to floxed Pex1 mice to allow for selective deletion of Pex1 in the hair cells of the inner ear. We find that Pex1 excision in inner hair cells (IHCs) leads to progressive hearing loss associated with significant decrease in auditory brainstem responses (ABR), specifically ABR wave I amplitude, indicative of synaptic defects. Analysis of IHC synapses in cKO mice reveals a decrease in ribbon synapse volume and functional alterations in exocytosis. Concomitantly, we observe a decrease in peroxisomal number, indicative of oxidative stress imbalance. Taken together, these results suggest a critical function of Pex1 in development and maturation of IHC-spiral ganglion synapses and auditory function.


Subject(s)
Cochlea , Hair Cells, Auditory, Inner , Hearing Loss , Synapses , Animals , Mice , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Cochlea/innervation , Cochlea/metabolism , Deafness/genetics , Deafness/metabolism , Hair Cells, Auditory, Inner/metabolism , Hearing/physiology , Hearing Loss/genetics , Hearing Loss/metabolism , Mice, Knockout , Synapses/genetics , Synapses/metabolism
5.
Ear Hear ; 43(2): 582-591, 2022.
Article in English | MEDLINE | ID: mdl-34534157

ABSTRACT

OBJECTIVES: Peroxisome Biogenesis Disorders in the Zellweger Spectrum (PBD-ZSD) are autosomal recessive disorders characterized by defects in peroxisome function, biosynthesis, and/or assembly. Despite its frequent documentation, hearing loss associated with PBD-ZSD has not been extensively characterized. The purpose of this retrospective natural history study was to better characterize the hearing loss associated with PBD-ZSD and to provide additional insight into the evaluation and management of PBD-ZSD patients with hearing loss. DESIGN: Audiological data from medical records of 42 patients with PBD-ZSD or D-bifunctional protein deficiency were collected from an ongoing longitudinal retrospective natural history study. An initial dataset of 300 audiograms and/or audiometric test results from the 42 patients were used to characterize the degree of hearing loss, type of hearing loss, relationships between air and bone conduction thresholds, age-related changes in hearing loss, and benefit with amplification. RESULTS: The majority of PBD-ZSD patients in this study presented with moderately-severe to severe hearing loss and relatively slow rates of longitudinal changes in hearing sensitivity. Improvements in hearing thresholds were observed with use of hearing aid amplification. Though bone conduction data were limited, air-bone gaps and air conduction threshold fluctuations observed in several patients suggest there may be an increased occurrence of mixed hearing losses in PBD-ZSD populations. CONCLUSION: The results of this retrospective study provide insight into the hearing loss associated with PBD-ZSD, but also emphasize the need for more complete assessments of hearing loss type and middle ear function in these patients. The addition of more comprehensive datasets to the ongoing natural history study will enhance our understanding of the pathophysiology underlying PBD-ZSD and guide the development of targeted evaluation and management recommendations for patients with PBD-ZSD.


Subject(s)
Deafness , Hearing Loss , Peroxisomal Disorders , Zellweger Syndrome , Female , Humans , Male , Retrospective Studies , Zellweger Syndrome/diagnosis , Zellweger Syndrome/metabolism
7.
Mol Ther ; 28(12): 2662-2676, 2020 12 02.
Article in English | MEDLINE | ID: mdl-32818431

ABSTRACT

Usher syndrome is a syndromic form of hereditary hearing impairment that includes sensorineural hearing loss and delayed-onset retinitis pigmentosa (RP). Type 1 Usher syndrome (USH1) is characterized by congenital profound sensorineural hearing impairment and vestibular areflexia, with adolescent-onset RP. Systemic treatment with antisense oligonucleotides (ASOs) targeting the human USH1C c.216G>A splicing mutation in a knockin mouse model of USH1 restores hearing and balance. Herein, we explore the effect of delivering ASOs locally to the ear to treat hearing and vestibular dysfunction associated with Usher syndrome. Three localized delivery strategies were investigated in USH1C mice: inner ear injection, trans-tympanic membrane injection, and topical tympanic membrane application. We demonstrate, for the first time, that ASOs delivered directly to the ear correct Ush1c expression in inner ear tissue, improve cochlear hair cell transduction currents, restore vestibular afferent irregularity, spontaneous firing rate, and sensitivity to head rotation, and successfully recover hearing thresholds and balance behaviors in USH1C mice. We conclude that local delivery of ASOs to the middle and inner ear reach hair cells and can rescue both hearing and balance. These results also demonstrate the therapeutic potential of ASOs to treat hearing and balance deficits associated with Usher syndrome and other ear diseases.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Ear, Middle/drug effects , Genetic Therapy/methods , Hair Cells, Auditory/drug effects , Mutation , Oligonucleotides, Antisense/administration & dosage , Usher Syndromes/genetics , Usher Syndromes/therapy , Vestibule, Labyrinth/drug effects , Administration, Topical , Animals , Animals, Newborn , Disease Models, Animal , Female , Gene Knock-In Techniques , Hair Cells, Auditory/metabolism , Hearing/drug effects , Injections , Male , Mice , Mice, Inbred C57BL , Tympanic Membrane/drug effects , Vestibule, Labyrinth/metabolism
8.
Nat Med ; 25(7): 1123-1130, 2019 07.
Article in English | MEDLINE | ID: mdl-31270503

ABSTRACT

Since most dominant human mutations are single nucleotide substitutions1,2, we explored gene editing strategies to disrupt dominant mutations efficiently and selectively without affecting wild-type alleles. However, single nucleotide discrimination can be difficult to achieve3 because commonly used endonucleases, such as Streptococcus pyogenes Cas9 (SpCas9), can tolerate up to seven mismatches between guide RNA (gRNA) and target DNA. Furthermore, the protospacer-adjacent motif (PAM) in some Cas9 enzymes can tolerate mismatches with the target DNA3,4. To circumvent these limitations, we screened 14 Cas9/gRNA combinations for specific and efficient disruption of a nucleotide substitution that causes the dominant progressive hearing loss, DFNA36. As a model for DFNA36, we used Beethoven mice5, which harbor a point mutation in Tmc1, a gene required for hearing that encodes a pore-forming subunit of mechanosensory transduction channels in inner-ear hair cells6. We identified a PAM variant of Staphylococcus aureus Cas9 (SaCas9-KKH) that selectively and efficiently disrupted the mutant allele, but not the wild-type Tmc1/TMC1 allele, in Beethoven mice and in a DFNA36 human cell line. Adeno-associated virus (AAV)-mediated SaCas9-KKH delivery prevented deafness in Beethoven mice up to one year post injection. Analysis of current ClinVar entries revealed that ~21% of dominant human mutations could be targeted using a similar approach.


Subject(s)
Alleles , Gene Editing , Hearing Loss, Sensorineural/prevention & control , Membrane Proteins/genetics , Animals , CRISPR-Associated Protein 9/physiology , Cell Line , Cells, Cultured , Dependovirus/genetics , Disease Models, Animal , Hearing Loss, Sensorineural/genetics , Humans , Mice , Mice, Inbred C57BL
9.
PLoS One ; 13(8): e0201713, 2018.
Article in English | MEDLINE | ID: mdl-30157177

ABSTRACT

Hearing and balance depend upon the precise morphogenesis and mechanosensory function of stereocilia, the specialized structures on the apical surface of sensory hair cells in the inner ear. Previous studies of Grxcr1 mutant mice indicated a critical role for this gene in control of stereocilia dimensions during development. In this study, we analyzed expression of the paralog Grxcr2 in the mouse and evaluated auditory and vestibular function of strains carrying targeted mutations of the gene. Peak expression of Grxcr2 occurs during early postnatal development of the inner ear and GRXCR2 is localized to stereocilia in both the cochlea and in vestibular organs. Homozygous Grxcr2 deletion mutants exhibit significant hearing loss by 3 weeks of age that is associated with developmental defects in stereocilia bundle orientation and organization. Despite these bundle defects, the mechanotransduction apparatus assembles in relatively normal fashion as determined by whole cell electrophysiological evaluation and FM1-43 uptake. Although Grxcr2 mutants do not exhibit overt vestibular dysfunction, evaluation of vestibular evoked potentials revealed subtle defects of the mutants in response to linear accelerations. In addition, reduced Grxcr2 expression in a hypomorphic mutant strain is associated with progressive hearing loss and bundle defects. The stereocilia localization of GRXCR2, together with the bundle pathologies observed in the mutants, indicate that GRXCR2 plays an intrinsic role in bundle orientation, organization, and sensory function in the inner ear during development and at maturity.


Subject(s)
Cochlea/cytology , Cochlea/growth & development , Glutaredoxins/metabolism , Morphogenesis , Stereocilia/metabolism , Amino Acid Sequence , Animals , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Glutaredoxins/chemistry , Glutaredoxins/genetics , Hearing Loss/genetics , Hearing Loss/pathology , Humans , Mechanotransduction, Cellular , Mice , Models, Molecular , Mutation , Protein Conformation , Species Specificity
10.
Sci Rep ; 8(1): 12124, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30108254

ABSTRACT

Recent work has demonstrated that transmembrane channel-like 1 protein (TMC1) is an essential component of the sensory transduction complex in hair cells of the inner ear. A closely related homolog, TMC2, is expressed transiently in the neonatal mouse cochlea and can enable sensory transduction in Tmc1-null mice during the first postnatal week. Both TMC1 and TMC2 are expressed at adult stages in mouse vestibular hair cells. The extent to which TMC1 and TMC2 can substitute for each other is unknown. Several biophysical differences between TMC1 and TMC2 suggest these proteins perform similar but not identical functions. To investigate these differences, and whether TMC2 can substitute for TMC1 in mature hair cells, we generated a knock-in mouse model allowing Cre-inducible expression of Tmc2. We assayed for changes in hair cell sensory transduction and auditory and vestibular function in Tmc2 knockin mice (Tm[Tmc2]) in the presence or absence of endogenous Tmc1, Tmc2 or both. Our results show that expression of Tm[TMC2] restores sensory transduction in vestibular hair cells and transiently in cochlear hair cells in the absence of TMC1. The cellular rescue leads to recovery of balance but not auditory function. We conclude that TMC1 provides some additional necessary function, not provided by TMC2.


Subject(s)
Hair Cells, Auditory, Inner/metabolism , Hair Cells, Vestibular/metabolism , Membrane Proteins/metabolism , Postural Balance/physiology , Animals , Gene Knock-In Techniques , Hearing/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Animal , Transgenes/genetics
11.
Science ; 344(6184): 1241062, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24812404

ABSTRACT

Hearing loss is the most common sensory deficit in humans, with some estimates suggesting up to 300 million affected individuals worldwide. Both environmental and genetic factors contribute to hearing loss and can cause death of sensory cells and neurons. Because these cells do not regenerate, the damage tends to accumulate, leading to profound deafness. Several biological strategies to restore auditory function are currently under investigation. Owing to the success of cochlear implants, which offer partial recovery of auditory function for some profoundly deaf patients, potential biological therapies must extend hearing restoration to include greater auditory acuity and larger patient populations. Here, we review the latest gene, stem-cell, and molecular strategies for restoring auditory function in animal models and the prospects for translating these approaches into viable clinical therapies.


Subject(s)
Cell Engineering/methods , Genetic Therapy/methods , Hair Cells, Auditory/physiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/therapy , Mechanotransduction, Cellular , Regeneration , Stem Cell Transplantation/methods , Animals , Cochlea/cytology , Cochlea/physiology , Humans , Mice , Spiral Ganglion/cytology , Spiral Ganglion/physiology
13.
J Clin Invest ; 121(12): 4796-809, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22105175

ABSTRACT

Inner ear hair cells convert the mechanical stimuli of sound, gravity, and head movement into electrical signals. This mechanotransduction process is initiated by opening of cation channels near the tips of hair cell stereocilia. Since the identity of these ion channels is unknown, and mutations in the gene encoding transmembrane channel-like 1 (TMC1) cause hearing loss without vestibular dysfunction in both mice and humans, we investigated the contribution of Tmc1 and the closely related Tmc2 to mechanotransduction in mice. We found that Tmc1 and Tmc2 were expressed in mouse vestibular and cochlear hair cells and that GFP-tagged TMC proteins localized near stereocilia tips. Tmc2 expression was transient in early postnatal mouse cochlear hair cells but persisted in vestibular hair cells. While mice with a targeted deletion of Tmc1 (Tmc1(Δ) mice) were deaf and those with a deletion of Tmc2 (Tmc2(Δ) mice) were phenotypically normal, Tmc1(Δ)Tmc2(Δ) mice had profound vestibular dysfunction, deafness, and structurally normal hair cells that lacked all mechanotransduction activity. Expression of either exogenous TMC1 or TMC2 rescued mechanotransduction in Tmc1(Δ)Tmc2(Δ) mutant hair cells. Our results indicate that TMC1 and TMC2 are necessary for hair cell mechanotransduction and may be integral components of the mechanotransduction complex. Our data also suggest that persistent TMC2 expression in vestibular hair cells may preserve vestibular function in humans with hearing loss caused by TMC1 mutations.


Subject(s)
Deafness/genetics , Hair Cells, Auditory, Inner/physiology , Hair Cells, Vestibular/physiology , Mechanotransduction, Cellular/physiology , Membrane Proteins/physiology , Animals , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Female , Fluorescent Dyes/metabolism , Genetic Complementation Test , Gentamicins/metabolism , Hair Cells, Auditory, Inner/ultrastructure , Hair Cells, Vestibular/ultrastructure , Male , Mechanotransduction, Cellular/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , RNA, Messenger/biosynthesis , Stereocilia/physiology , Stereocilia/ultrastructure
14.
J Neurosci ; 31(34): 12241-50, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865467

ABSTRACT

The polycystic kidney disease-1 (Pkd1) gene encodes a large transmembrane protein (polycystin-1, or PC-1) that is reported to function as a fluid flow sensor in the kidney. As a member of the transient receptor potential family, PC-1 has also been hypothesized to play a role in the elusive mechanoelectrical transduction (MET) channel in inner ear hair cells. Here, we analyze two independent mouse models of PC-1, a knock-in (KI) mutant line and a hair cell-specific inducible Cre-mediated knock-out line. Both models exhibit normal MET channel function at neonatal ages despite hearing loss and ultrastructural abnormalities of sterecilia that remain properly polarized at adult ages. These findings demonstrate that PC-1 plays an essential role in stereocilia structure and maintenance but not directly in MET channel function or planar cell polarity. We also demonstrate that PC-1 is colocalized with F-actin in hair cell stereocilia in vivo, using a hemagglutinin-tagged PC-1 KI mouse model, and in renal epithelial cell microvilli in vitro. These results not only demonstrate a novel role for PC-1 in the cochlea, but also suggest insight into the development of polycystic kidney disease.


Subject(s)
Cilia/metabolism , Hair Cells, Auditory, Inner/metabolism , Mechanotransduction, Cellular/physiology , Organ of Corti/physiology , TRPP Cation Channels/physiology , Animals , Animals, Newborn , Cilia/genetics , Disease Models, Animal , Female , Gene Knock-In Techniques , Hair Cells, Auditory, Inner/cytology , HeLa Cells , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Humans , Male , Mechanotransduction, Cellular/genetics , Mice , Mice, Knockout , Mice, Transgenic , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics
15.
PLoS One ; 5(1): e8627, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20062532

ABSTRACT

The molecular composition of the hair cell transduction channel has not been identified. Here we explore the novel hypothesis that hair cell transduction channels include HCN subunits. The HCN family of ion channels includes four members, HCN1-4. They were originally identified as the molecular correlates of the hyperpolarization-activated, cyclic nucleotide gated ion channels that carry currents known as If, IQ or Ih. However, based on recent evidence it has been suggested that HCN subunits may also be components of the elusive hair cell transduction channel. To investigate this hypothesis we examined expression of mRNA that encodes HCN1-4 in sensory epithelia of the mouse inner ear, immunolocalization of HCN subunits 1, 2 and 4, uptake of the transduction channel permeable dye, FM1-43 and electrophysiological measurement of mechanotransduction current. Dye uptake and transduction current were assayed in cochlear and vestibular hair cells of wildtype mice exposed to HCN channel blockers or a dominant-negative form of HCN2 that contained a pore mutation and in mutant mice that lacked HCN1, HCN2 or both. We found robust expression of HCNs 1, 2 and 4 but little evidence that localized HCN subunits in hair bundles, the site of mechanotransduction. Although high concentrations of the HCN antagonist, ZD7288, blocked 50-70% of the transduction current, we found no reduction of transduction current in either cochlear or vestibular hair cells of HCN1- or HCN2- deficient mice relative to wild-type mice. Furthermore, mice that lacked both HCN1 and HCN2 also had normal transduction currents. Lastly, we found that mice exposed to the dominant-negative mutant form of HCN2 had normal transduction currents as well. Taken together, the evidence suggests that HCN subunits are not required for mechanotransduction in hair cells of the mouse inner ear.


Subject(s)
Ear, Inner/physiology , Hair Cells, Vestibular/physiology , Ion Channels/physiology , Mechanotransduction, Cellular , Animals , Ion Channels/genetics , Ion Channels/metabolism , Mice , RNA, Messenger/genetics
16.
J Assoc Res Otolaryngol ; 11(1): 27-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19834762

ABSTRACT

TRP genes encode a diverse family of ion channels which have been implicated in many sensory functions. Because several TRP channels have similar properties to the elusive hair cell transduction channel, recent attention has focused on TRP gene expression in the inner ear. At least four TRP genes are known to be expressed in hair cells: TRPC3, TRPV4, TRPA1, and TRPML3. However, there is little evidence supporting any of these as a component of the transduction complex. Other less well-characterized TRP channels are expressed in the inner ear, in particular, within the organ of Corti. Because of their potential role in sensory function, we investigated the developmental expression of RNA that encodes all 33 TRP subunits as well as several splice variants. We designed a quantitative PCR screen using cochlear samples acquired before, during, and after the time when mechanotransduction is acquired in sensory hair cells (embryonic day 17 to postnatal day 8). Cochleas, which included the organ of Corti, stria vascularis, and Reissner's membrane, were subdivided into four equal quadrants which allowed for regional comparison during development. Expression of RNA transcripts that encoded 33 TRP subunits plus several splice forms and beta-actin were quantified in 28 samples for a total of 1,092 individual measurements, each done in triplicate. We detected RNA that encoded all TRP channels except two: TRPC7 and TRPM8. The largest changes in RNA expression were for TRPA1 (>100-fold), TRPP3 (>50-fold), and TRPC5.2 (>20-fold) which suggested that these subunits may contribute to normal cochlear function. Furthermore, the screen revealed TRPP3 and PKD1L3 RNA expression patterns that were correlated with the acquisition of sensory transduction in outer hair cells (Lelli et al., J Neurophysiol. 101:2961-2973, 2009). Numerous spatiotemporal expression gradients were identified many of which may contribute to the normal functional development of the mouse cochlea.


Subject(s)
Cochlea/embryology , Cochlea/physiology , Gene Expression Regulation, Developmental , Transient Receptor Potential Channels/genetics , Alternative Splicing/genetics , Animals , Female , Mice , Multigene Family/genetics , Polymerase Chain Reaction , Pregnancy , TRPA1 Cation Channel , TRPC Cation Channels/genetics , TRPM Cation Channels/genetics , TRPP Cation Channels/genetics , TRPV Cation Channels/genetics
17.
J Neurosci ; 29(50): 15859-69, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016102

ABSTRACT

Morphogenesis of sensory hair cells, in particular their mechanotransduction organelle, the stereociliary bundle, requires highly organized remodeling of the actin cytoskeleton. The roles of Rho family small GTPases during this process remain unknown. Here we show that deletion of Rac1 in the otic epithelium resulted in severe defects in cochlear epithelial morphogenesis. The mutant cochlea was severely shortened with a reduced number of auditory hair cells and cellular organization of the auditory sensory epithelium was abnormal. Rac1 mutant hair cells also displayed defects in planar cell polarity and morphogenesis of the stereociliary bundle, including bundle fragmentation or deformation, and mispositioning or absence of the kinocilium. We further demonstrate that a Rac-PAK (p21-activated kinase) signaling pathway mediates kinocilium-stereocilia interactions and is required for cohesion of the stereociliary bundle. Together, these results reveal a critical function of Rac1 in morphogenesis of the auditory sensory epithelium and stereociliary bundle.


Subject(s)
Hair Cells, Auditory/enzymology , Hair Cells, Auditory/physiology , Morphogenesis/physiology , Neuropeptides/physiology , rac GTP-Binding Proteins/physiology , Animals , Animals, Newborn , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Mice , Mice, Knockout , Morphogenesis/genetics , Neuropeptides/deficiency , Neuropeptides/genetics , Organ of Corti/cytology , Organ of Corti/growth & development , Organ of Corti/physiology , Pregnancy , rac GTP-Binding Proteins/deficiency , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
18.
J Neurophysiol ; 101(6): 2961-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19339464

ABSTRACT

Inner ear hair cells are exquisite mechanosensors that transduce nanometer scale deflections of their sensory hair bundles into electrical signals. Several essential elements must be precisely assembled during development to confer the unique structure and function of the mechanotransduction apparatus. Here we investigated the functional development of the transduction complex in outer hair cells along the length of mouse cochlea acutely excised between embryonic day 17 (E17) and postnatal day 8 (P8). We charted development of the stereociliary bundle using scanning electron microscopy; FM1-43 uptake, which permeates hair cell transduction channels, mechanotransduction currents evoked by rapid hair bundle deflections, and mRNA expression of possible components of the transduction complex. We demonstrated that uptake of FM1-43 first occurred in the basal portion of the cochlea at P0 and progressed toward the apex over the subsequent week. Electrophysiological recordings obtained from 234 outer hair cells between E17 and P8 from four cochlear regions revealed a correlation between the pattern of FM1-43 uptake and the acquisition of mechanotransduction. We found a spatiotemporal gradient in the properties of transduction including onset, amplitude, operating range, time course, and extent of adaptation. We used quantitative RT-PCR to examine relative mRNA expression of several hair cell myosins and candidate tip-link molecules. We found spatiotemporal expression patterns for mRNA that encodes cadherin 23, protocadherin 15, myosins 3a, 7a, 15a, and PMCA2 that preceded the acquisition of transduction. The spatiotemporal expression patterns of myosin 1c and PMCA2 mRNA were correlated with developmental changes in several properties of mechanotransduction.


Subject(s)
Cochlea , Gene Expression Regulation, Developmental/physiology , Hair Cells, Auditory, Outer/physiology , Mechanotransduction, Cellular/physiology , Adaptation, Physiological , Animals , Animals, Newborn , Cochlea/cytology , Cochlea/embryology , Cochlea/growth & development , Embryo, Mammalian , Hair Cells, Auditory, Outer/ultrastructure , Membrane Potentials/physiology , Mice , Microscopy, Confocal/methods , Microscopy, Electron, Scanning/methods , Myosins/classification , Myosins/genetics , Myosins/metabolism , Patch-Clamp Techniques , Physical Stimulation/methods , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasma Membrane Calcium-Transporting ATPases/metabolism , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , RNA, Messenger/metabolism
19.
J Neurosci ; 27(33): 8940-51, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699675

ABSTRACT

Sensory hair cells of the inner ear express multiple physiologically defined conductances, including mechanotransduction, Ca(2+), Na(+), and several distinct K(+) conductances, all of which are critical for normal hearing and balance function. Yet, the molecular underpinnings and their specific contributions to sensory signaling in the inner ear remain obscure. We sought to identify hair-cell conductances mediated by KCNQ4, which, when mutated, causes the dominant progressive hearing loss DFNA2. We used the dominant-negative pore mutation G285S and packaged the coding sequence of KCNQ4 into adenoviral vectors. We transfected auditory and vestibular hair cells of organotypic cultures generated from the postnatal mouse inner ear. Cochlear outer hair cells and vestibular type I cells that expressed the transfection marker, green fluorescent protein, and the dominant-negative KCNQ4 construct lacked the M-like conductances that typify nontransfected control hair cells. As such, we conclude that the M-like conductances in mouse auditory and vestibular hair cells can include KCNQ4 subunits and may also include KCNQ4 coassembly partners. To examine the function of M-like conductances in hair cells, we recorded from cells transfected with mutant KCNQ4 and injected transduction current waveforms in current-clamp mode. Because the M-like conductances were active at rest, they contributed to the very low potassium-selective input resistance, which in turn hyperpolarized the resting potential and significantly attenuated the amplitude of the receptor potential. Modulation of M-like conductances may allow hair cells the ability to control the amplitude of their response to sensory stimuli.


Subject(s)
Ear, Inner/cytology , Hair Cells, Auditory, Inner/physiology , KCNQ Potassium Channels/physiology , Neural Inhibition/physiology , Animals , Animals, Newborn , Cells, Cultured , Electric Stimulation/methods , Embryo, Mammalian , Gene Expression/physiology , Gene Expression Regulation, Developmental/physiology , Genetic Vectors/physiology , Glycine/genetics , Humans , KCNQ Potassium Channels/genetics , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mutation/physiology , Neural Inhibition/genetics , Neural Inhibition/radiation effects , Organ Culture Techniques , Patch-Clamp Techniques/methods , Saccule and Utricle/embryology , Saccule and Utricle/growth & development , Saccule and Utricle/metabolism , Serine/genetics , Transfection/methods
20.
J Assoc Res Otolaryngol ; 8(1): 18-31, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17171473

ABSTRACT

The adult mammalian cochlea lacks regenerative capacity, which is the main reason for the permanence of hearing loss. Vestibular organs, in contrast, replace a small number of lost hair cells. The reason for this difference is unknown. In this work we show isolation of sphere-forming stem cells from the early postnatal organ of Corti, vestibular sensory epithelia, the spiral ganglion, and the stria vascularis. Organ of Corti and vestibular sensory epithelial stem cells give rise to cells that express multiple hair cell markers and express functional ion channels reminiscent of nascent hair cells. Spiral ganglion stem cells display features of neural stem cells and can give rise to neurons and glial cell types. We found that the ability for sphere formation in the mouse cochlea decreases about 100-fold during the second and third postnatal weeks; this decrease is substantially faster than the reduction of stem cells in vestibular organs, which maintain their stem cell population also at older ages. Coincidentally, the relative expression of developmental and progenitor cell markers in the cochlea decreases during the first 3 postnatal weeks, which is in sharp contrast to the vestibular system, where expression of progenitor cell markers remains constant or even increases during this period. Our findings indicate that the lack of regenerative capacity in the adult mammalian cochlea is either a result of an early postnatal loss of stem cells or diminishment of stem cell features of maturing cochlear cells.


Subject(s)
Hair Cells, Auditory/cytology , Saccule and Utricle/cytology , Spiral Ganglion/cytology , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers , Cell Differentiation , Electrophysiology , Female , Green Fluorescent Proteins/genetics , Hair Cells, Auditory/embryology , Hair Cells, Auditory/growth & development , Hair Cells, Auditory/physiology , Ion Channels/physiology , Mice , Mice, Transgenic , Pregnancy , Regeneration , Saccule and Utricle/embryology , Saccule and Utricle/growth & development , Saccule and Utricle/physiology , Spheroids, Cellular , Spiral Ganglion/embryology , Spiral Ganglion/growth & development , Spiral Ganglion/physiology , Stem Cells/physiology , Stria Vascularis/cytology , Stria Vascularis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...