Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Trends Hear ; 192015 Dec 01.
Article in English | MEDLINE | ID: mdl-26631107

ABSTRACT

Combined electric and acoustic stimulation has proven to be an effective strategy to improve hearing in some cochlear implant users. We describe an acoustic microactuator to directly deliver stimuli to the perilymph in the scala tympani. The 800 µm by 800 µm actuator has a silicon diaphragm driven by a piezoelectric thin film (e.g., lead-zirconium-titanium oxide or PZT). This device could also be used as a component of a bimodal acoustic-electric electrode array. In the current study, we established a guinea pig model to test the actuator for its ability to deliver auditory signals to the cochlea in vivo. The actuator was placed through the round window of the cochlea. Auditory brainstem response (ABR) thresholds, peak latencies, and amplitude growth were calculated for an ear canal speaker versus the intracochlear actuator for tone burst stimuli at 4, 8, 16, and 24 kHz. An ABR was obtained after removal of the probe to assess loss of hearing related to the procedure. In some animals, the temporal bone was harvested for histologic analysis of cochlear damage. We show that the device is capable of stimulating ABRs in vivo with latencies and growth functions comparable to stimulation in the ear canal. Further experiments will be necessary to evaluate the efficiency and safety of this modality in long-term auditory stimulation and its ability to be integrated with conventional cochlear implant arrays.


Subject(s)
Acoustic Stimulation/instrumentation , Acoustic Stimulation/methods , Cochlear Implants , Evoked Potentials, Auditory, Brain Stem/physiology , Animals , Auditory Threshold/physiology , Cochlear Implantation/methods , Disease Models, Animal , Female , Guinea Pigs , Piezosurgery/methods , Prosthesis Design , Random Allocation , Sensitivity and Specificity
2.
J Neurosci ; 35(20): 7878-91, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995473

ABSTRACT

During nervous system development, critical periods are usually defined as early periods during which manipulations dramatically change neuronal structure or function, whereas the same manipulations in mature animals have little or no effect on the same property. Neurons in the ventral cochlear nucleus (CN) are dependent on excitatory afferent input for survival during a critical period of development. Cochlear removal in young mammals and birds results in rapid death of target neurons in the CN. Cochlear removal in older animals results in little or no neuron death. However, the extent to which hair-cell-specific afferent activity prevents neuronal death in the neonatal brain is unknown. We further explore this phenomenon using a new mouse model that allows temporal control of cochlear hair cell deletion. Hair cells express the human diphtheria toxin (DT) receptor behind the Pou4f3 promoter. Injections of DT resulted in nearly complete loss of organ of Corti hair cells within 1 week of injection regardless of the age of injection. Injection of DT did not influence surrounding supporting cells directly in the sensory epithelium or spiral ganglion neurons (SGNs). Loss of hair cells in neonates resulted in rapid and profound neuronal loss in the ventral CN, but not when hair cells were eliminated at a more mature age. In addition, normal survival of SGNs was dependent on hair cell integrity early in development and less so in mature animals. This defines a previously undocumented critical period for SGN survival.


Subject(s)
Cochlear Nucleus/growth & development , Hair Cells, Auditory/cytology , Spiral Ganglion/growth & development , Animals , Cell Death , Cochlear Nucleus/cytology , Cochlear Nucleus/physiology , Diphtheria Toxin/pharmacology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hearing , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Spiral Ganglion/cytology , Spiral Ganglion/physiology
3.
Hear Res ; 297: 91-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23178236

ABSTRACT

Auditory hair cells transduce sound vibrations into membrane potential changes, ultimately leading to changes in neuronal firing and sound perception. This review provides an overview of the characteristics and repair capabilities of traumatized auditory sensory epithelium in the adult vertebrate ear. Injured mammalian auditory epithelium repairs itself by forming permanent scars but is unable to regenerate replacement hair cells. In contrast, injured non-mammalian vertebrate ear generates replacement hair cells to restore hearing functions. Non-sensory support cells within the auditory epithelium play key roles in the repair processes.


Subject(s)
Ear, Inner/physiology , Epithelium/pathology , Hair Cells, Auditory/physiology , Nerve Regeneration/physiology , Regeneration/physiology , Wounds and Injuries , Animals , Brain-Derived Neurotrophic Factor/metabolism , Ear, Inner/physiopathology , Hearing , Hearing Loss, Noise-Induced/physiopathology , Humans , Membrane Potentials , Organ of Corti/physiopathology , Spiral Ganglion/pathology , Stem Cell Transplantation/methods , Vertebrates/physiology
4.
J Neurosci ; 31(43): 15329-39, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22031879

ABSTRACT

The capacity of adult mammals to regenerate sensory hair cells is not well defined. To explore early steps in this process, we examined reactivation of a transiently expressed developmental gene, Atoh1, in adult mouse utricles after neomycin-induced hair cell death in culture. Using an adenoviral reporter for Atoh1 enhancer, we found that Atoh1 transcription is activated in some hair cell progenitors (supporting cells) 3 d after neomycin treatment. By 18 d after neomycin, the number of cells with Atoh1 transcriptional activity increased significantly, but few cells acquired hair cell features (i.e., accumulated ATOH1 or myosin VIIa protein or developed stereocilia). Treatment with DAPT, an inhibitor of γ-secretase, reduced notch pathway activity, enhanced Atoh1 transcriptional activity, and dramatically increased the number of Atoh1-expressing cells with hair cell features, but only in the striolar/juxtastriolar region. Similar effects were seen with TAPI-1, an inhibitor of another enzyme required for notch activity (TACE). Division of supporting cells was rare in any control or DAPT-treated utricles. This study shows that mature mammals have a natural capacity to initiate vestibular hair cell regeneration and suggests that regional notch activity is a significant inhibitor of direct transdifferentiation of supporting cells into hair cells following damage.


Subject(s)
Nerve Regeneration/physiology , Neural Inhibition/physiology , Receptors, Notch/metabolism , Saccule and Utricle/cytology , ADAM Proteins/pharmacology , ADAM17 Protein , Amyloid Precursor Protein Secretases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Calbindins , Calmodulin/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Dipeptides/pharmacology , Green Fluorescent Proteins/genetics , Hair Cells, Auditory/drug effects , Hydroxamic Acids/pharmacology , Indoles , Mice , Neomycin/toxicity , Nerve Regeneration/drug effects , Neural Inhibition/drug effects , Organ Culture Techniques , Protein Synthesis Inhibitors/toxicity , S100 Calcium Binding Protein G/metabolism , Saccule and Utricle/injuries , Time Factors , Transduction, Genetic/methods , Ventricular Myosins/metabolism
5.
Cell Cycle ; 10(8): 1237-48, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21403466

ABSTRACT

Cell cycle inhibitors, such as the cyclin-dependent kinase (Cdk) inhibitor proteins and retinoblastoma (Rb) family members, control exit from the cell cycle during the development of a variety of terminally differentiated tissues. It is unclear whether sustained expression of these proteins is required to prevent cell cycle re-entry in quiescent and terminally differentiated cells. The organ of Corti (cochlear sensory epithelium) and pars intermedia (intermediate lobe of the pituitary) are two tissues that share the characteristic of ongoing cell division in mice lacking either the p27(Kip1) Cdk inhibitor, Ink4 proteins, or Rb. Here, we use tamoxifen-inducible mouse models to delete p27(Kip1) in postnatal animals and show this is sufficient to induce proliferation in both the organ of Corti and pars intermedia. Thus, these tissues remain sensitive to the presence of p27(Kip1) even after their developmental exit from the cell cycle. The neonatal cochlea displayed heightened sensitivity to changes in p27(Kip1) expression, with a proliferative response higher than that of constitutive null mice. In adults, the proliferative response was reduced but was accompanied by increased cell survival. In contrast, re-establishment of normal p27(Kip1) expression in animals with established pituitary tumors, in an inducible "knock-on" model, led to cessation of pituitary tumor growth, indicating the cells had maintained their susceptibility to p27-mediated growth suppression. Although restoration of p27(Kip1) did not induce apoptosis, it did lead to resolution of pathological features and normalization of gene expression. Our data underscore the importance of p27(Kip1) expression in the maintenance of cellular quiescence and terminal differentiation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/metabolism , Neoplasms, Experimental/metabolism , Organ of Corti/metabolism , Pituitary Gland, Intermediate/metabolism , Animals , Animals, Newborn , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Survival , Chickens , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cyclins/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Organ of Corti/cytology , Organ of Corti/embryology , Organogenesis , Pituitary Gland, Intermediate/embryology , Pituitary Gland, Intermediate/pathology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tamoxifen/pharmacology
6.
Dev Neurobiol ; 70(4): 253-67, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20095043

ABSTRACT

Usher syndrome is the leading cause of combined deaf-blindness, but the molecular mechanisms underlying the auditory and visual impairment are poorly understood. Usher I is characterized by profound congenital hearing loss, vestibular dysfunction, and progressive retinitis pigmentosa beginning in early adolescence. Using the c.216G>A cryptic splice site mutation in Exon 3 of the USH1C gene found in Acadian Usher I patients in Louisiana, we constructed the first mouse model that develops both deafness and retinal degeneration. The same truncated mRNA transcript found in Usher 1C patients is found in the cochleae and retinas of these knock-in mice. Absent auditory-evoked brainstem responses indicated that the mutant mice are deaf at 1 month of age. Cochlear histology showed disorganized hair cell rows, abnormal bundles, and loss of both inner and outer hair cells in the middle turns and at the base. Retinal dysfunction as evident by an abnormal electroretinogram was seen as early as 1 month of age, with progressive loss of rod photoreceptors between 6 and 12 months of age. This knock-in mouse reproduces the dual sensory loss of human Usher I, providing a novel resource to study the disease mechanism and the development of therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Deafness/physiopathology , Disease Models, Animal , Retinal Degeneration/physiopathology , Usher Syndromes/physiopathology , Adaptor Proteins, Signal Transducing/metabolism , Aging , Animals , Cell Cycle Proteins , Cochlea/pathology , Cochlea/physiopathology , Cochlea/ultrastructure , Cytoskeletal Proteins , Deafness/pathology , Electroretinography , Evoked Potentials, Auditory, Brain Stem , Exons , Gene Knock-In Techniques , Hair Cells, Auditory/pathology , Hair Cells, Auditory/physiology , Louisiana , Mice , Mice, Transgenic , Mutation, Missense , RNA Splice Sites , RNA, Messenger/metabolism , Retina/pathology , Retina/physiopathology , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/physiology , Usher Syndromes/pathology
7.
J Neurosci ; 30(2): 478-90, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071511

ABSTRACT

Humans and other mammals are highly susceptible to permanent hearing and balance deficits due to an inability to regenerate sensory hair cells lost to inner ear trauma. In contrast, nonmammalian vertebrates, such as birds, robustly regenerate replacement hair cells and restore hearing and balance functions to near-normal levels. There is considerable interest in understanding the cellular mechanisms responsible for this difference in regenerative capacity. Here we report on involvement of the TGFbeta superfamily type II activin receptors, Acvr2a and Acvr2b, in regulating proliferation in mature avian auditory sensory epithelium. Cultured, posthatch avian auditory sensory epithelium treated with Acvr2a and Acvr2b inhibitors shows decreased proliferation of support cells, the cell type that gives rise to new hair cells. Conversely, addition of activin A, an Acvr2a/b ligand, potentiates support cell proliferation. Neither treatment (inhibitor or ligand) affected hair cell survival, suggesting a specific effect of Acvr2a/b signaling on support cell mitogenicity. Using immunocytochemistry, Acvr2a, Acvr2b, and downstream Smad effector proteins were differentially localized in avian and mammalian auditory sensory epithelia. Collectively, these data suggest that signaling through Acvr2a/b promotes support cell proliferation in mature avian auditory sensory epithelium and that this signaling pathway may be incomplete, or actively blocked, in the adult mammalian ear.


Subject(s)
Activins/pharmacology , Cell Proliferation/drug effects , Cochlear Duct/cytology , Epithelium/metabolism , Gene Expression Regulation, Developmental/physiology , Activin Receptors, Type II/metabolism , Activins/genetics , Activins/metabolism , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Count/methods , Chick Embryo , Chickens , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelium/anatomy & histology , Gene Expression Regulation, Developmental/drug effects , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/metabolism , Mice , Organ Culture Techniques , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Smad Proteins/metabolism
8.
J Assoc Res Otolaryngol ; 10(4): 525-44, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19644644

ABSTRACT

Significant sensory hair cell loss leads to irreversible hearing and balance deficits in humans and other mammals. Future therapeutic strategies to repair damaged mammalian auditory epithelium may involve inserting stem cells into the damaged epithelium, inducing non-sensory cells remaining in the epithelium to transdifferentiate into replacement hair cells via gene therapy, or applying growth factors. Little is currently known regarding the status and characteristics of the non-sensory cells that remain in the deafened auditory epithelium, yet this information is integral to the development of therapeutic treatments. A single high-dose injection of the aminoglycoside kanamycin coupled with a single injection of the loop diuretic furosemide was used to kill hair cells in adult mice, and the mice were examined 1 year after the drug insult. Outer hair cells are lost throughout the entire length of the cochlea and less than a third of the inner hair cells remain in the apical turn. Over 20% and 55% of apical organ of Corti support cells and spiral ganglion cells are lost, respectively. We examined the expression of several known support cell markers to investigate for possible support cell dedifferentiation in the damaged ears. The support cell markers investigated included the microtubule protein acetylated tubulin, the transcription factor Sox2, and the Notch signaling ligand Jagged1. Non-sensory epithelial cells remaining in the organ of Corti retain acetylated tubulin, Sox2 and Jagged1 expression, even when the epithelium has a monolayer-like appearance. These results suggest a lack of marked SC dedifferentiation in these aged and badly damaged ears.


Subject(s)
Deafness/pathology , Labyrinth Supporting Cells/cytology , Aging/pathology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/biosynthesis , Cell Differentiation , Deafness/chemically induced , Deafness/metabolism , Diuretics/administration & dosage , Diuretics/adverse effects , Furosemide/administration & dosage , Furosemide/adverse effects , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/biosynthesis , Jagged-1 Protein , Kanamycin/administration & dosage , Kanamycin/adverse effects , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Membrane Proteins/analysis , Membrane Proteins/biosynthesis , Mice , Protein Synthesis Inhibitors/administration & dosage , Protein Synthesis Inhibitors/adverse effects , SOXB1 Transcription Factors/analysis , SOXB1 Transcription Factors/biosynthesis , Serrate-Jagged Proteins , Spiral Ganglion/drug effects , Spiral Ganglion/pathology , Tubulin/analysis , Tubulin/biosynthesis
9.
Hear Res ; 252(1-2): 61-70, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19450430

ABSTRACT

Estrogen signaling in auditory and vestibular sensory epithelia is a newly emerging focus propelled by the role of estrogen signaling in many other proliferative systems. Understanding the pathways with which estrogen interacts can provide a means to identify how estrogen may modulate proliferative signaling in inner ear sensory epithelia. Reviewed herein are two signaling families, EGF and TGFbeta. Both pathways are involved in regulating proliferation of supporting cells in mature vestibular sensory epithelia and have well characterized interactions with estrogen signaling in other systems. It is becoming increasingly clear that elucidating the complexity of signaling in regeneration will be necessary for development of therapeutics that can initiate regeneration and prevent progression to a pathogenic state.


Subject(s)
Ear, Inner/physiology , Estrogens/physiology , Regeneration/physiology , Animals , Cell Proliferation , Embryonic Stem Cells/physiology , Epidermal Growth Factor/physiology , ErbB Receptors/physiology , Female , Humans , Male , Models, Biological , Receptors, Estrogen/physiology , Signal Transduction , Transforming Growth Factor alpha/physiology , Transforming Growth Factor beta/physiology
10.
J Assoc Res Otolaryngol ; 9(1): 65-89, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18157569

ABSTRACT

Inner ear hair cells detect environmental signals associated with hearing, balance, and body orientation. In humans and other mammals, significant hair cell loss leads to irreversible hearing and balance deficits, whereas hair cell loss in nonmammalian vertebrates is repaired by the spontaneous generation of replacement hair cells. Research in mammalian hair cell regeneration is hampered by the lack of in vivo damage models for the adult mouse inner ear and the paucity of cell-type-specific markers for non-sensory cells within the sensory receptor epithelia. The present study delineates a protocol to drug damage the adult mouse auditory epithelium (organ of Corti) in situ and uses this protocol to investigate Sox2 and Jagged1 expression in damaged inner ear sensory epithelia. In other tissues, the transcription factor Sox2 and a ligand member of the Notch signaling pathway, Jagged1, are involved in regenerative processes. Both are involved in early inner ear development and are expressed in developing support cells, but little is known about their expressions in the adult. We describe a nonsurgical technique for inducing hair cell damage in adult mouse organ of Corti by a single high-dose injection of the aminoglycoside kanamycin followed by a single injection of the loop diuretic furosemide. This drug combination causes the rapid death of outer hair cells throughout the cochlea. Using immunocytochemical techniques, Sox2 is shown to be expressed specifically in support cells in normal adult mouse inner ear and is not affected by drug damage. Sox2 is absent from auditory hair cells, but is expressed in a subset of vestibular hair cells. Double-labeling experiments with Sox2 and calbindin suggest Sox2-positive hair cells are Type II. Jagged1 is also expressed in support cells in the adult ear and is not affected by drug damage. Sox2 and Jagged1 may be involved in the maintenance of support cells in adult mouse inner ear.


Subject(s)
Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Organ of Corti/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Animals, Newborn , Anti-Bacterial Agents/toxicity , Biomarkers/metabolism , Chickens , Cochlear Diseases/chemically induced , Disease Models, Animal , Diuretics/toxicity , Furosemide/toxicity , Jagged-1 Protein , Kanamycin/toxicity , Mice , Mice, Inbred CBA , Organ of Corti/drug effects , Serrate-Jagged Proteins , Time Factors
11.
Gene Expr Patterns ; 7(7): 798-807, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17604700

ABSTRACT

A cascade of transcription factors is believed to regulate the coordinate differentiation of primordial inner ear cells into the subtypes of hair cells and supporting cells. While candidate genes involved in this process have been identified, the temporal and spatial patterns of expression of many of these have not been carefully described during the extended period of inner ear development and functional maturation. We systematically examined the expression of two such transcription factors, LHX3 and SOX2, from the time of hair cell terminal mitoses into adulthood. We show that LHX3 is expressed specifically in auditory and vestibular hair cells soon after terminal mitoses and persists into the adult in vestibular hair cells. While SOX2 expression is widespread in the inner ear sensory epithelia prior to hair cell differentiation, it has a unique pattern of expression in the mature auditory and vestibular organs.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Ear, Inner/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/physiology , Trans-Activators/biosynthesis , Trans-Activators/physiology , Animals , Cell Differentiation , Deafness , Gene Expression Profiling , LIM-Homeodomain Proteins , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Models, Genetic , Protein Structure, Tertiary , SOXB1 Transcription Factors , Time Factors , Tissue Distribution , Transcription Factors
12.
J Comp Neurol ; 496(2): 172-86, 2006 May 10.
Article in English | MEDLINE | ID: mdl-16538679

ABSTRACT

We carried out an analysis of the expression of Prox1, a homeo-domain transcription factor, during mouse inner ear development with particular emphasis on the auditory system. Prox1 is expressed in the otocyst beginning at embryonic day (E)11, in the developing vestibular sensory patches. Expression is down regulated in maturing (myosin VIIA immunoreactive) vestibular hair cells and subsequently in the underlying support cell layer by E16.5. In the auditory sensory epithelium, Prox1 is initially expressed at embryonic day 14.5 in a narrow stripe of cells at the base of the cochlea. This stripe encompasses the full thickness of the sensory epithelium, including developing hair cells and support cells. Over the next several days the stripe of expression extends to the apex, and as the sensory epithelium differentiates Prox1 becomes restricted to a subset of support cells. Double labeling for Prox1 and cell-type-specific markers revealed that the outer hair cells transiently express Prox1. After E18, Prox1 protein is no longer detectable in hair cells, but it continues to be expressed in support cells for the rest of embryogenesis and into the second postnatal week. During this time, Prox1 is not expressed in all support cell types in the organ of Corti, but is restricted to developing Deiters' and pillar cells. The expression is maintained in these cells into the second week of postnatal life, at which time Prox1 is dynamically down regulated. These studies form a baseline from which we can analyze the role of Prox1 in vertebrate sensory development.


Subject(s)
Cochlea/metabolism , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cochlea/embryology , Cochlea/growth & development , Embryo, Mammalian , Epithelium/embryology , Epithelium/growth & development , Epithelium/metabolism , Female , Gene Expression/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Mice , Mice, Transgenic , Models, Biological , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , S100 Proteins/metabolism , Tumor Suppressor Proteins , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
13.
J Assoc Res Otolaryngol ; 4(3): 422-43, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14690060

ABSTRACT

In humans, hair cell loss often leads to hearing and balance impairments. Hair cell replacement is vigorous and spontaneous in avians and nonmammalian vertebrates. In mammals, in contrast, it occurs at a very low rate, or not at all, presumably because of a very low level of supporting cell proliferation following injury. Heregulin (HRG), a member of the epidermal growth factor (EGF) family of growth factors, is reported to be a potent mitogen for neonatal rat vestibular sensory epithelium, but its effects in adults are unknown. We report here that HRG-alpha stimulates cell proliferation in organotypic cultures of neonatal, but not adult, mouse utricular sensory epithelia. Our findings support the idea that the proliferative capabilities of the adult mammalian vestibular sensory epithelia differ significantly from that seen in neonatal animals. Immunohistochemistry reveals that HRG-binding receptors (erbBs 2-4) and erbB1 are widely expressed in vestibular and auditory sensory epithelia in neonatal and adult mouse inner ear. The distribution of erbBs in the neonatal and adult mouse ear is consistent with the EGF receptor/ligand family regulating diverse cellular processes in the inner ear, including cell proliferation and differentiation.


Subject(s)
ErbB Receptors/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Neuregulin-1/pharmacology , Age Factors , Animals , Animals, Newborn , Antibodies , Cell Division/drug effects , ErbB Receptors/immunology , Hair Cells, Auditory/drug effects , Mice , Mitogens/pharmacology , Organ Culture Techniques , Organ of Corti/cytology , Organ of Corti/physiology , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Receptor, ErbB-4 , Regeneration/drug effects , Saccule and Utricle/cytology , Saccule and Utricle/physiology , Vestibule, Labyrinth/cytology , Vestibule, Labyrinth/physiology
14.
J Comp Neurol ; 463(2): 177-95, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12815755

ABSTRACT

Ototoxic drugs stimulate cell proliferation in adult rat vestibular sensory epithelia, as does the infusion of transforming growth factor alpha (TGFalpha) plus insulin. We sought to determine whether new hair cells can be regenerated by means of a mitotic pathway. Previously, studies have shown that the nuclei of some newly generated cells are located in the lumenal half of the sensory epithelium, suggesting that some may be newly generated sensory hair cells. The aim of this study was to examine the ultrastructural characteristics of newly proliferated cells after TGFalpha stimulation and/or aminoglycoside damage in the utricular sensory epithelium of the adult rat. The cell proliferation marker tritiated-thymidine was infused, with or without TGFalpha plus insulin, into the inner ears of normal or aminoglycoside-damaged rats for 3 or 7 days by means of osmotic pumps. Autoradiographic techniques and light microscopy were used to identify cells synthesizing DNA. Sections with labeled cells were re-embedded, processed for transmission electron microscopy, and the ultrastructural characteristics of the labeled cells were examined. The following five classes of tritiated-thymidine labeled cells were identified in the sensory epithelium: (1) labeled cells with synaptic specializations that appeared to be newly generated hair cells, (2) labeled supporting cells, (3) labeled leukocytes, (4) labeled cells that we have classified as "active cells" in that they are relatively nondescript but contain massive numbers of polyribosomes, and (5) labeled degenerating hair cells. These findings suggest that new hair cells can be generated in situ by means of a mitotic mechanism in the vestibular sensory epithelium of adult mammals.


Subject(s)
Hair Cells, Vestibular/chemistry , Hair Cells, Vestibular/ultrastructure , Thymidine/metabolism , Acoustic Maculae/chemistry , Acoustic Maculae/ultrastructure , Animals , Male , Rats , Rats, Sprague-Dawley , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...