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










Publication year range
1.
Elife ; 112022 12 14.
Article in English | MEDLINE | ID: mdl-36515985

ABSTRACT

Upon trauma, the adult murine peripheral nervous system (PNS) displays a remarkable degree of spontaneous anatomical and functional regeneration. To explore extrinsic mechanisms of neural repair, we carried out single-cell analysis of naïve mouse sciatic nerve, peripheral blood mononuclear cells, and crushed sciatic nerves at 1 day, 3 days, and 7 days following injury. During the first week, monocytes and macrophages (Mo/Mac) rapidly accumulate in the injured nerve and undergo extensive metabolic reprogramming. Proinflammatory Mo/Mac with a high glycolytic flux dominate the early injury response and rapidly give way to inflammation resolving Mac, programmed toward oxidative phosphorylation. Nerve crush injury causes partial leakiness of the blood-nerve barrier, proliferation of endoneurial and perineurial stromal cells, and entry of opsonizing serum proteins. Micro-dissection of the nerve injury site and distal nerve, followed by single-cell RNA-sequencing, identified distinct immune compartments, triggered by mechanical nerve wounding and Wallerian degeneration, respectively. This finding was independently confirmed with Sarm1-/- mice, in which Wallerian degeneration is greatly delayed. Experiments with chimeric mice showed that wildtype immune cells readily enter the injury site in Sarm1-/- mice, but are sparse in the distal nerve, except for Mo. We used CellChat to explore intercellular communications in the naïve and injured PNS and report on hundreds of ligand-receptor interactions. Our longitudinal analysis represents a new resource for neural tissue regeneration, reveals location- specific immune microenvironments, and reports on large intercellular communication networks. To facilitate mining of scRNAseq datasets, we generated the injured sciatic nerve atlas (iSNAT): https://cdb-rshiny.med.umich.edu/Giger_iSNAT/.


Subject(s)
Peripheral Nerve Injuries , Wallerian Degeneration , Mice , Animals , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Leukocytes, Mononuclear , Sciatic Nerve/metabolism , Nerve Degeneration , Nerve Crush , Peripheral Nerve Injuries/metabolism , Nerve Regeneration , Cytoskeletal Proteins/metabolism , Armadillo Domain Proteins/metabolism
2.
Hear Res ; 424: 108601, 2022 10.
Article in English | MEDLINE | ID: mdl-36126618

ABSTRACT

Many factors contribute to hearing loss commonly found in older adults. There can be natural aging of cellular elements, hearing loss previously induced by environmental factors such as noise or ototoxic drugs as well as genetic and epigenetic influences. Even when noise overstimulation does not immediately cause permanent hearing loss it has recently been shown to increase later age-related hearing loss (ARHL). The present study further investigated this condition in the UMHET4 mouse model by comparing a small arms fire (SAF)-like impulse noise exposure that has the greatest immediate effect in more apical cochlear regions to a broadband noise (BBN) exposure that has the greatest immediate effect in more basal cochlear regions. Both noise exposures were given at levels that only induced temporary auditory brainstem response (ABR) threshold shifts (TS). Mice were noise exposed at 5 months of age followed by ABR assessment at 6, 12, 18, 21, and 24 months of age. Mice that received the SAF-like impulse noise had accelerated age-related TS at 4 kHz that appeared at 12 months of age (significantly increased compared to no-noise controls). This increased TS at 4 kHz continued at 18 and 21 months but was no longer significantly greater at 24 months of age. The SAF-like impulse noise also induced a significantly greater mean TS at 48 kHz, first appearing at 18 months of age and continuing to be significantly greater than controls at 21 and 24 months. The BBN induced a different pace and pattern of enhanced age-related ABR TS. The mean TS for the BBN group first became significantly greater than controls at 18 months of age and only at 48 kHz. It remained significantly greater than controls at 21 months but was no longer significantly greater at 24 months of age. Results, therefore, show different influences on ARHL for the two different noise exposure conditions. Noise-induced enhancement appears to provide more an acceleration than overall total increase in ARHL.


Subject(s)
Hearing Loss, Noise-Induced , Presbycusis , Animals , Auditory Threshold/physiology , Cochlea , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Noise-Induced/genetics , Mice , Noise/adverse effects , Presbycusis/genetics
3.
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
4.
Front Cell Neurosci ; 15: 658972, 2021.
Article in English | MEDLINE | ID: mdl-33897373

ABSTRACT

Our previous study demonstrated rapamycin added to diet at 4 months of age had significantly less age-related outer hair cell loss in the basal half of the cochlea at 22 months of age compared to mice without rapamycin. The present study tested adding rapamycin to diet later in life, at 14 months of age, and added a longitudinal assessment of auditory brain stem response (ABR). The present study used UMHET4 mice, a 4 way cross in which all grandparental strains lack the Cdh23753A allele that predisposes to early onset, progressive hearing loss. UMHET4 mice typically have normal hearing until 16-17 months, then exhibit threshold shifts at low frequencies/apical cochlea and later in more basal high frequency regions. ABR thresholds at 4, 12, 24, and 48 kHz were assessed at 12, 18, and 24 months of age and compared to baseline ABR thresholds acquired at 5 months of age to determine threshold shifts (TS). There was no TS at 12 months of age at any frequency tested. At 18 months of age mice with rapamycin added to diet at 14 months had a significantly lower mean TS at 4 and 12 kHz compared to mice on control diet with no significant difference at 24 and 48 kHz. At 24 months of age, the mean 4 kHz TS in rapamycin diet group was no longer significantly lower than the control diet group, while the 12 kHz mean remained significantly lower. Mean TS at 24 and 48 kHz in the rapamycin diet group became significantly lower than in the control diet group at 24 months. Hair cell counts at 24 months showed large loss in the apical half of most rapamycin and control diet mice cochleae with no significant difference between groups. There was only mild outer hair cell loss in the basal half of rapamycin and control diet mice cochleae with no significant difference between groups. The results show that a later life addition of rapamycin can decrease age-related hearing loss in the mouse model, however, it also suggests that this decrease is a delay/deceleration rather than a complete prevention.

5.
Dev Neurobiol ; 81(5): 546-567, 2021 07.
Article in English | MEDLINE | ID: mdl-33561889

ABSTRACT

The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.


Subject(s)
Auditory Pathways , Cochlea , Acoustic Stimulation , Animals , Auditory Pathways/physiology , Axons , Cochlea/physiology , Humans , Mammals , Neuroglia
6.
Article in English | MEDLINE | ID: mdl-30617057

ABSTRACT

Hidden hearing loss (HHL), a recently described auditory disorder, has been proposed to affect auditory neural processing and hearing acuity in subjects with normal audiometric thresholds, particularly in noisy environments. In contrast to central auditory processing disorders, HHL is caused by defects in the cochlea, the peripheral auditory organ. Noise exposure, aging, ototoxic drugs, and peripheral neuropathies are some of the known risk factors for HHL. Our knowledge of the causes and mechanisms of HHL are based primarily on animal models. However, recent clinical studies have also shed light on the etiology and prevalence of this cochlear disorder and how it may affect auditory perception in humans. Here, we review the current knowledge regarding the causes and cellular mechanisms of HHL, summarize information on available noninvasive tests for differential diagnosis, and discuss potential therapeutic approaches for treatment of HHL.


Subject(s)
Hearing Loss/etiology , Hearing Loss/physiopathology , Hearing Loss/therapy , Animals , Cochlea/physiopathology , Cochlear Nerve/physiopathology , Diagnosis, Differential , Disease Models, Animal , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Inner/physiology , Humans
7.
Sci Rep ; 9(1): 15362, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31653916

ABSTRACT

Noise exposures causing only transient threshold shifts can destroy auditory-nerve synapses without damaging hair cells. Here, we asked whether virally mediated neurotrophin3 (NT3) overexpression can repair this damage. CBA/CaJ mice at 6 wks were injected unilaterally with adeno-associated virus (AAV) containing either NT3 or GFP genes, via the posterior semicircular canal, 3 wks prior to, or 5 hrs after, noise exposure. Controls included exposed animals receiving vehicle only, and unexposed animals receiving virus. Thresholds were measured 2 wks post-exposure, just before cochleas were harvested for histological analysis. In separate virus-injected animals, unexposed cochleas were extracted for qRT-PCR. The GFP reporter showed that inner hair cells (IHCs) were transfected throughout the cochlea, and outer hair cells mainly in the apex. qRT-PCR showed 4- to 10-fold overexpression of NT3 from 1-21 days post-injection, and 1.7-fold overexpression at 40 days. AAV-NT3 delivered prior to noise exposure produced a dose-dependent reduction of synaptopathy, with nearly complete rescue at some cochlear locations. In unexposed ears, NT3 overexpression did not affect thresholds, however GFP overexpression caused IHC loss. In exposed ears, NT3 overexpression increased permanent threshold shifts. Thus, although NT3 overexpression can minimize noise-induced synaptic damage, the forced overexpression may be harmful to hair cells themselves during cochlear overstimulation.


Subject(s)
Cochlea/pathology , Dependovirus/metabolism , Neurotrophin 3/metabolism , Noise , Synapses/pathology , Animals , Auditory Threshold , Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem , Green Fluorescent Proteins/metabolism , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Neurotrophin 3/genetics , Otoacoustic Emissions, Spontaneous , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synapses/metabolism
8.
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
9.
Otolaryngol Head Neck Surg ; 156(6): 1114-1118, 2017 06.
Article in English | MEDLINE | ID: mdl-28566047

ABSTRACT

Experience with procurement of research funding and grantsmanship is an essential skill and one that is rarely taught in a manner that adequately prepares trainees for the magnitude of this professional requirement. The aims of the program described in this article are (1) to provide a mentored experience in grantsmanship through designing and concisely outlining an individual research study and (2) to supplement extramural funding mechanisms for clinical trainees to produce meaningful and substantive clinical and/or basic science research. A total of $10,000 of departmental chair discretionary funds is allocated for resident research annually. The first 2 cycles have successfully allocated the allotted funding through a competitive, scored grant evaluation process. Awardees have already produced meaningful data that have been nationally presented, submitted for publication, and integrated into an National Institutes of Health grant submission. The feasibility of implementing an intramural competitive resident research grant may have broad application within varied training environments.


Subject(s)
Biomedical Research/economics , Competitive Behavior , Otolaryngology/education , Research Support as Topic , Education, Medical, Graduate , Humans , Internship and Residency , National Institutes of Health (U.S.) , Professional Competence , United States
10.
J Assoc Res Otolaryngol ; 16(6): 695-712, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26463873

ABSTRACT

SLC44A2 (solute carrier 44a2), also known as CTL2 (choline transporter-like protein 2), is expressed in many supporting cell types in the cochlea and is implicated in hair cell survival and antibody-induced hearing loss. In mice with the mixed C57BL/6-129 background, homozygous deletion of Slc44a2 exons 3­10 (Slc44a2(Δ/Δ)resulted in high-frequency hearing loss and hair cell death. To reduce effects associated with age-related hearing loss (ARHL) in these strains, mice carrying the Slc44a2Δ allele were backcrossed to the ARHL-resistant FVB/NJ strain and evaluated after backcross seven(N7) (99 % FVB). Slc44a2(Δ/Δ) mice produced abnormally spliced Slc44a2 transcripts that contain a frame shift and premature stop codons. Neither full-length SLC44A2 nor a putative truncated protein could be detected in Slc44a2(Δ/Δ) mice, suggesting a likely null allele. Auditory brain stem responses (ABRs) of mice carrying the Slc44a2Δ allele on an FVB/NJ genetic background were tested longitudinally between the ages of 2 and 10 months. By 6 months of age,Slc44a2(Δ/Δ) mice exhibited hearing loss at 32 kHz,but at 12 and 24 kHz had sound thresholds similar to those of wild-type Slc44a2(+/+) and heterozygous +/Slc44a2Δ mice. After 6 months of age, Slc44a2(Δ/Δ) mutants exhibited progressive hearing loss at all frequencies and +/Slc44a2(Δ) mice exhibited moderate threshold elevations at high frequency. Histologic evaluation of Slc44a2(Δ/Δ) mice revealed extensive hair cell and spiral ganglion cell loss, especially in the basal turn of the cochlea. We conclude that Slc44a2 function is required for long-term hair cell survival and maintenance of hearing.


Subject(s)
Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/genetics , Membrane Transport Proteins/genetics , Spiral Ganglion/pathology , Amino Acid Sequence , Animals , Female , Gene Deletion , Hearing Loss, Sensorineural/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data
11.
Hum Mutat ; 35(5): 618-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24619944

ABSTRACT

More than 360 million humans are affected with some degree of hearing loss, either early or later in life. A genetic cause for the disorder is present in a majority of the cases. We mapped a locus (DFNB101) for hearing loss in humans to chromosome 5q in a consanguineous Pakistani family. Exome sequencing revealed an insertion mutation in GRXCR2 as the cause of moderate-to-severe and likely progressive hearing loss in the affected individuals of the family. The frameshift mutation is predicted to affect a conserved, cysteine-rich region of GRXCR2, and to result in an abnormal extension of the C-terminus. Functional studies by cell transfections demonstrated that the mutant protein is unstable and mislocalized relative to wild-type GRXCR2, consistent with a loss-of-function mutation. Targeted disruption of Grxcr2 is concurrently reported to cause hearing loss in mice. The structural abnormalities in this animal model suggest a role for GRXCR2 in the development of stereocilia bundles, specialized structures on the apical surface of sensory cells in the cochlea that are critical for sound detection. Our results indicate that GRXCR2 should be considered in differential genetic diagnosis for individuals with early onset, moderate-to-severe and progressive hearing loss.


Subject(s)
Frameshift Mutation , Glutaredoxins/genetics , Hearing Loss/genetics , Animals , Exome , Genes, Recessive , Hearing Loss/diagnosis , Hearing Loss/pathology , Humans , Mice , Mutation , Pedigree
12.
Neurobiol Aging ; 33(8): 1842.e15-29, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22305187

ABSTRACT

A genetically heterogeneous population of mice was tested for hearing at 8, 18, and 22 months by auditory brainstem response (ABR), and genotyped at 128 markers to identify loci that modulate late life hearing loss. Half of the test mice were exposed to noise for 2 hours at age 20 months. Polymorphisms affecting hearing at 18 months were noted on chromosomes 2, 3, 7, 10, and 15. Most of these loci had effects only on responses to 48 kHz stimuli, but a subset also influenced the auditory brainstem response at lower frequencies. Loci on chromosomes 4, 10, 12, and 14 had significant effects on hearing at 22 months in noise-exposed mice, and loci on chromosomes 10 and 11 had effects on mice not exposed to noise. Outer hair cell loss was modulated by polymorphisms on chromosomes 10, 11, 12, 17, and 19. Resistance to age-related hearing loss is thus modulated by a set of genetic effects, some age-specific, some frequency specific, some dependent on prior exposure to noise, and some of which compromise survival of cochlear hair cells.


Subject(s)
Aging/genetics , Alleles , Gene Frequency/genetics , Hearing Loss/genetics , Polymorphism, Single Nucleotide/genetics , Animals , Mice
13.
J Assoc Res Otolaryngol ; 13(1): 29-37, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21932106

ABSTRACT

Diverse cellular and environmental stresses can activate the heat shock response, an evolutionarily conserved mechanism to protect proteins from denaturation. Stressors activate heat shock transcription factor 1 (HSF1), which binds to heat shock elements in the genes for heat shock proteins, leading to rapid induction of these important molecular chaperones. Both heat and noise stress are known to activate the heat shock response in the cochlea and protect it from subsequent noise trauma. However, the contribution of HSF1 to induction of heat shock proteins following noise trauma has not been investigated at the molecular level. We evaluated the role of HSF1 in the cochlea following noise stress by examining induction of heat shock proteins in Hsf1 ( +/- ) control and Hsf1 ( -/- ) mice. Heat stress rapidly induced expression of Hsp25, Hsp47, Hsp70.1, Hsp70.3, Hsp84, Hsp86, and Hsp110 in the cochleae of wild-type and Hsf1 ( +/- ) mice, but not in Hsf1 ( -/- ) mice, confirming the essential role of HSF1 in mediating the heat shock response. Exposure to broadband noise (2-20 kHz) at 106 dB SPL for 2 h produced partial hearing loss. Maximal induction of heat shock proteins occurred 4 h after the noise. In comparison to heat stress, noise stress resulted in lower induced levels of Hsp25, Hsp70.1, Hsp70.3, Hsp86, and Hsp110 in Hsf1 ( +/- ) mice. Induction of these heat shock proteins was attenuated, but not completely eliminated, in Hsf1 ( -/- ) mice. These same noise exposure conditions induced genes for several immediate early transcription factors and maximum induction occurred earlier than for heat shock proteins. Thus, additional signaling pathways and transcriptional regulators that are activated by noise probably contribute to induction of heat shock proteins in the cochlea.


Subject(s)
Cochlea/physiology , DNA-Binding Proteins/genetics , Fever/genetics , Fever/physiopathology , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/physiopathology , Transcription Factors/genetics , Animals , DNA-Binding Proteins/metabolism , Female , Fever/metabolism , Gene Expression/physiology , Genes, Immediate-Early/physiology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Hearing Loss, Noise-Induced/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Mutant Strains , Molecular Chaperones , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Noise/adverse effects , Signal Transduction/physiology , Stress, Physiological/physiology , Transcription Factors/metabolism
14.
Am J Hum Genet ; 86(2): 148-60, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20137774

ABSTRACT

Recessive mutations at the mouse pirouette (pi) locus result in hearing loss and vestibular dysfunction due to neuroepithelial defects in the inner ear. Using a positional cloning strategy, we have identified mutations in the gene Grxcr1 (glutaredoxin cysteine-rich 1) in five independent allelic strains of pirouette mice. We also provide sequence data of GRXCR1 from humans with profound hearing loss suggesting that pirouette is a model for studying the mechanism of nonsyndromic deafness DFNB25. Grxcr1 encodes a 290 amino acid protein that contains a region of similarity to glutaredoxin proteins and a cysteine-rich region at its C terminus. Grxcr1 is expressed in sensory epithelia of the inner ear, and its encoded protein is localized along the length of stereocilia, the actin-filament-rich mechanosensory structures at the apical surface of auditory and vestibular hair cells. The precise architecture of hair cell stereocilia is essential for normal hearing. Loss of function of Grxcr1 in homozygous pirouette mice results in abnormally thin and slightly shortened stereocilia. When overexpressed in transfected cells, GRXCR1 localizes along the length of actin-filament-rich structures at the dorsal-apical surface and induces structures with greater actin filament content and/or increased lengths in a subset of cells. Our results suggest that deafness in pirouette mutants is associated with loss of GRXCR1 function in modulating actin cytoskeletal architecture in the developing stereocilia of sensory hair cells.


Subject(s)
Ear, Inner/physiopathology , Genetic Loci/genetics , Glutaredoxins/genetics , Mutation/genetics , Actin Cytoskeleton , Alleles , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Conserved Sequence , DNA Mutational Analysis , Evolution, Molecular , Female , Gene Expression Regulation , Glutaredoxins/chemistry , Hearing Loss/genetics , Hearing Loss/physiopathology , Humans , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Pedigree , Protein Structure, Tertiary , Protein Transport
15.
Dev Dyn ; 237(4): 941-52, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18330929

ABSTRACT

The inner ear is a complex organ containing sensory tissue, including hair cells, the development of which is not well understood. Our long-term goal is to discover genes critical for the correct formation and function of the inner ear and its sensory tissue. A novel gene, transmembrane inner ear (Tmie), was found to cause hearing-related disorders when defective in mice and humans. A homologous tmie gene in zebrafish was cloned and its expression characterized between 24 and 51 hours post-fertilization. Embryos injected with morpholinos (MO) directed against tmie exhibited circling swimming behavior (approximately 37%), phenocopying mice with Tmie mutations; semicircular canal formation was disrupted, hair cell numbers were reduced, and maturation of electrically active lateral line neuromasts was delayed. As in the mouse, tmie appears to be required for inner ear development and function in the zebrafish and for hair cell maturation in the vestibular and lateral line systems as well.


Subject(s)
Ear, Inner/embryology , Ear, Inner/physiology , Lateral Line System/embryology , Lateral Line System/physiology , Membrane Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish , Amino Acid Sequence , Animals , Behavior, Animal/physiology , Ear, Inner/anatomy & histology , Female , Gene Expression Regulation, Developmental , Humans , Lateral Line System/anatomy & histology , Male , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Morphogenesis , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Sequence Alignment , Swimming/physiology , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish/physiology , Zebrafish Proteins/metabolism
16.
J Assoc Res Otolaryngol ; 7(3): 317-28, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16794912

ABSTRACT

The vertebrate cochlea is a complex organ optimized for sound transduction. Auditory hair cells, with their precisely arranged stereocilia bundles, transduce sound waves to electrical signals that are transmitted to the brain. Mutations in the unconventional myosin XV cause deafness in both human DFNB3 families and in shaker 2 (sh2) mice as a result of defects in stereocilia. In these mutant mice, hair cells have relatively normal spatial organization of stereocilia bundles but lack the graded, stair-step organization. We used sh2 mice as an experimental model to investigate the molecular consequences of the sh2 mutation in the Myo15 gene. Gene expression profiling with Affymetrix GeneChips in deaf homozygous (sh2/sh2) mice at 3 weeks and 3 months of age, and in age-matched, normal-hearing heterozygotes (+/sh2) identified only a few genes whose expression was affected by genotype, but a large number with age-associated changes in expression in both normal mice and sh2/sh2 homozygotes. Microarray data analyzed using Robust Multiarray Average identified Aim1, Dbi, and Tm4sf3 as genes with increased expression in sh2/sh2 homozygotes. These increases were confirmed by quantitative reverse transcription-polymerase chain reaction. Genes exhibiting altered expression with age encoded collagens and proteins involved in collagen maturation, extracellular matrix, and bone mineralization. These results identified potential cellular pathways associated with myosin XV defects, and age-associated molecular events that are likely to be involved in maturation of the cochlea and auditory function.


Subject(s)
Aging/metabolism , Gene Expression Regulation , Hair Cells, Auditory/metabolism , Mutation , Myosins/biosynthesis , Aging/genetics , Aging/pathology , Animals , Calcinosis/genetics , Calcinosis/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Hair Cells, Auditory/pathology , Hearing Loss/genetics , Hearing Loss/metabolism , Hearing Loss/pathology , Humans , Mice , Mice, Mutant Strains , Myosins/genetics , Oligonucleotide Array Sequence Analysis/methods , POU Domain Factors/genetics
17.
Audiol Neurootol ; 9(5): 303-14, 2004.
Article in English | MEDLINE | ID: mdl-15347914

ABSTRACT

The mouse mutant 'pirouette' (pi) exhibits profound hearing loss and vestibular defects due to inheritance of a recessive mutation on chromosome 5. Dysfunction has been correlated with defects during maturation of sensory cells in the inner ear. As an initial step in characterizing pirouette at the genetic level, we have localized the candidate interval to a small region on central chromosome 5 by analysis of a congenic strain of pirouette mice. This region exhibits conserved synteny with human chromosome 4 and suggests that pirouette may be a genetic model of the human nonsyndromic deafness disorder DFNB25, which has been localized to 4p15.3-q12. In addition to the original spontaneous pirouette strain, we have identified and characterized 2 additional mouse strains with allelic mutations at the same locus. Analysis of the morphology in each of the 3 pirouette alleles indicated very similar early postnatal alterations in maturation of stereocilia and suggests that the gene affected in pirouette normally plays a role in building or maintaining these structures that are critical for sensory mechanotransduction.


Subject(s)
Deafness/genetics , Mutagenesis, Insertional , Mutation , Transgenes , Actins/analysis , Alleles , Animals , Cell Line , Evoked Potentials, Auditory, Brain Stem/genetics , Genotype , Hair Cells, Auditory/chemistry , Hair Cells, Auditory/ultrastructure , Humans , Immunohistochemistry , Membrane Glycoproteins , Mice , Mice, Mutant Strains , Microfilament Proteins/analysis , Microscopy, Electron, Scanning , Phosphoproteins/analysis
18.
Hum Mol Genet ; 12(21): 2797-805, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12966030

ABSTRACT

The unconventional myosin genes Myo15, Myo6 and Myo7a are essential for hearing in both humans and mice. Despite the expression of each gene in multiple organs, mutations result in identifiable phenotypes only in auditory or ocular sensory organs. The pirouette (pi) mouse also exhibits deafness and an inner ear pathology resembling that of Myo15 mutant mice and thus may be functionally related to Myo15. In order to investigate possible interactions between Myo15 and Myo6, Myo7a, and the gene affected in pirouette, we crossed Myo15(sh2/sh2) mice to the three other mutant mouse strains. Hearing in doubly heterozygous mice was similar to age-matched singly heterozygous animals, indicating that partial deficiency for both Myo15 and one of these other deafness genes does not reduce hearing. Viable double mutants were obtained from each cross, indicating that potential overlapping functions between these genes in other organs are not essential for viability. All critical cell types of the cochlear sensory epithelium were present in double mutant mice and cochlear stereocilia exhibited a superimposition of single mutant phenotypes. These data suggest that the function of Myo15 is distinct from that of Myo6, Myo7a or pi in development and/or maintenance of stereocilia.


Subject(s)
Cochlea/pathology , Cochlear Diseases/genetics , Myosin Heavy Chains/genetics , Myosins/genetics , Animals , Cochlea/metabolism , Cochlear Diseases/metabolism , Dyneins , Female , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/ultrastructure , Hearing/genetics , Hearing/physiology , Male , Mice , Mice, Mutant Strains , Microscopy, Electron, Scanning , Mutation , Myosin Heavy Chains/metabolism , Myosin VIIa , Myosins/metabolism
19.
Hum Mol Genet ; 11(16): 1887-98, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12140191

ABSTRACT

The recessive mutation at the mouse spinner (sr) locus results in hearing loss and vestibular dysfunction due to neuroepithelial defects in the inner ear. Using a positional cloning strategy, we have identified the mutant locus responsible for this pathology. The affected gene (Tmie) lies within a 40 kb deletion in the original sr allele. In a newly identified allele, Tmie contains a nonsense mutation expected to truncate the C-terminal end of its product. The 153 amino acid protein encoded by the gene shows no similarity to other known proteins, and is predicted to contain a signal peptide and at least one transmembrane domain. Tmie transcripts were identified in several tissues, including the cochlea. Loss of function of Tmie results in postnatal alterations of sensory hair cells in the cochlea, including defects in stereocilia, the apical projections of hair cells that are important in mechanotransduction of sound. These morphological defects are associated with a profound failure to develop normal auditory function. Consistent with a conserved role for this gene in the cochlea, the genetic mapping data presented here support human TMIE as the gene affected at DFNB6, a non-syndromic hearing loss locus. The spinner mutant is thus a valuable model for insight into mechanisms of human deafness and development of sensory cell function.


Subject(s)
Hearing Loss/genetics , Membrane Proteins/genetics , Mutation , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary/genetics , Disease Models, Animal , Female , Hair Cells, Auditory/abnormalities , Hair Cells, Auditory/ultrastructure , Hearing Loss/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron, Scanning , Molecular Sequence Data , Sequence Deletion , Sequence Homology, Nucleic Acid
20.
Am J Hum Genet ; 71(3): 632-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12145746

ABSTRACT

We have identified five different homozygous recessive mutations in a novel gene, TMIE (transmembrane inner ear expressed gene), in affected members of consanguineous families segregating severe-to-profound prelingual deafness, consistent with linkage to DFNB6. The mutations include an insertion, a deletion, and three missense mutations, and they indicate that loss of function of TMIE causes hearing loss in humans. TMIE encodes a protein with 156 amino acids and exhibits no significant nucleotide or deduced amino acid sequence similarity to any other gene.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Deafness/genetics , Genetic Linkage/genetics , Membrane Proteins/genetics , Mutation/genetics , Amino Acid Sequence , Base Sequence , Consanguinity , Female , Genes, Recessive/genetics , Haplotypes/genetics , Homozygote , Humans , Male , Membrane Proteins/chemistry , Molecular Sequence Data , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...