Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727276

ABSTRACT

In mammals, hearing loss is irreversible due to the lack of the regenerative capacity of the auditory epithelium. However, stem/progenitor cells in mammalian cochleae may be a therapeutic target for hearing regeneration. The ubiquitin proteasome system plays an important role in cochlear development and maintenance. In this study, we investigated the role of ubiquitin C-terminal hydrolase L1 (UCHL1) in the process of the transdifferentiation of auditory supporting cells (SCs) into hair cells (HCs). The expression of UCHL1 gradually decreased as HCs developed and was restricted to inner pillar cells and third-row Deiters' cells between P2 and P7, suggesting that UCHL1-expressing cells are similar to the cells with Lgr5-positive progenitors. UCHL1 expression was decreased even under conditions in which supernumerary HCs were generated with a γ-secretase inhibitor and Wnt agonist. Moreover, the inhibition of UCHL1 by LDN-57444 led to an increase in HC numbers. Mechanistically, LDN-57444 increased mTOR complex 1 activity and allowed SCs to transdifferentiate into HCs. The suppression of UCHL1 induces the transdifferentiation of auditory SCs and progenitors into HCs by regulating the mTOR pathway.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , Signal Transduction , TOR Serine-Threonine Kinases , Ubiquitin Thiolesterase , Animals , Cell Transdifferentiation/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Indoles , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/cytology , Oximes , TOR Serine-Threonine Kinases/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Rats
2.
Cell Rep ; 35(3): 109016, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882317

ABSTRACT

The mammalian cochlea cannot regenerate functional hair cells (HCs) spontaneously. Atoh1 overexpression as well as other strategies are unable to generate functional HCs. Here, we simultaneously upregulated the expression of Gfi1, Pou4f3, and Atoh1 in postnatal cochlear supporting cells (SCs) in vivo, which efficiently converted SCs into HCs. The newly regenerated HCs expressed HC markers Myo7a, Calbindin, Parvalbumin, and Ctbp2 and were innervated by neurites. Importantly, many new HCs expressed the mature and terminal marker Prestin or vesicular glutamate transporter 3 (vGlut3), depending on the subtypes of the source SCs. Finally, our patch-clamp analysis showed that the new HCs in the medial region acquired a large K+ current, fired spikes transiently, and exhibited signature refinement of ribbon synapse functions, in close resemblance to native wild-type inner HCs. We demonstrated that co-upregulating Gfi1, Pou4f3, and Atoh1 enhances the efficiency of HC generation and promotes the functional maturation of new HCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , Hair Cells, Auditory/metabolism , Homeodomain Proteins/genetics , Labyrinth Supporting Cells/metabolism , Organogenesis/genetics , Transcription Factor Brn-3C/genetics , Transcription Factors/genetics , Action Potentials/physiology , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calbindins/genetics , Calbindins/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Homeodomain Proteins/metabolism , Ion Transport , Labyrinth Supporting Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Myosin VIIa/genetics , Myosin VIIa/metabolism , Neurites/metabolism , Neurites/ultrastructure , Parvalbumins/genetics , Parvalbumins/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Signal Transduction , Transcription Factor Brn-3C/metabolism , Transcription Factors/metabolism
3.
Cell Rep ; 34(12): 108900, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761346

ABSTRACT

In contrast to mammals, birds recover naturally from acquired hearing loss, which makes them an ideal model for inner ear regeneration research. Here, we present a validated single-cell RNA sequencing resource of the avian cochlea. We describe specific markers for three distinct types of sensory hair cells, including a previously unknown subgroup, which we call superior tall hair cells. We identify markers for the supporting cells associated with tall hair cells, which represent the facultative stem cells of the avian inner ear. Likewise, we present markers for supporting cells that are located below the short cochlear hair cells. We further infer spatial expression gradients of hair cell genes along the tonotopic axis of the cochlea. This resource advances neurobiology, comparative biology, and regenerative medicine by providing a basis for comparative studies with non-regenerating mammalian cochleae and for longitudinal studies of the regenerating avian cochlea.


Subject(s)
Chickens/anatomy & histology , Cochlea/cytology , Animals , Biomarkers/metabolism , Epithelium/physiology , Gene Expression Profiling , Gene Expression Regulation , Hair Cells, Auditory, Inner/cytology , Labyrinth Supporting Cells/cytology , RNA-Seq , Reproducibility of Results , Single-Cell Analysis
4.
Sci Rep ; 10(1): 6740, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32317718

ABSTRACT

To protect the audiosensory organ from tissue damage from the immune system, the inner ear is separated from the circulating immune system by the blood-labyrinth barrier, which was previously considered an immune-privileged site. Recent studies have shown that macrophages are distributed in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis; however, the direct pathogen defence mechanism used by audiosensory receptor hair cells (HCs) has remained obscure. Here, we show that HCs are protected from pathogens by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs). In isolated murine cochlear sensory epithelium, we established Theiler's murine encephalomyelitis virus, which infected the SCs and GERCs, but very few HCs. The virus-infected SCs produced interferon (IFN)-α/ß, and the viruses efficiently infected the HCs in the IFN-α/ß receptor-null sensory epithelium. Interestingly, the virus-infected SCs and GERCs expressed macrophage marker proteins and were eliminated from the cell layer by cell detachment. Moreover, lipopolysaccharide induced phagocytosis of the SCs without cell detachment, and the SCs phagocytosed the bacteria. These results reveal that SCs function as macrophage-like cells, protect adjacent HCs from pathogens, and provide a novel anti-infection inner ear immune system.


Subject(s)
Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Outer/physiology , Labyrinth Supporting Cells/immunology , Macrophages/immunology , Spiral Ganglion/physiology , Stria Vascularis/physiology , Animals , Animals, Newborn , Escherichia coli/immunology , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Outer/cytology , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interferon-beta/biosynthesis , Interferon-beta/immunology , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/virology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/virology , Mice , Mice, Inbred ICR , Organ Culture Techniques , Phagocytosis/drug effects , Saccharomyces cerevisiae/immunology , Spiral Ganglion/cytology , Stria Vascularis/cytology , Theilovirus/growth & development , Theilovirus/pathogenicity
5.
Cell Mol Life Sci ; 77(7): 1401-1419, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31485717

ABSTRACT

Foxg1 is one of the forkhead box genes that are involved in morphogenesis, cell fate determination, and proliferation, and Foxg1 was previously reported to be required for morphogenesis of the mammalian inner ear. However, Foxg1 knock-out mice die at birth, and thus the role of Foxg1 in regulating hair cell (HC) regeneration after birth remains unclear. Here we used Sox2CreER/+ Foxg1loxp/loxp mice and Lgr5-EGFPCreER/+ Foxg1loxp/loxp mice to conditionally knock down Foxg1 specifically in Sox2+ SCs and Lgr5+ progenitors, respectively, in neonatal mice. We found that Foxg1 conditional knockdown (cKD) in Sox2+ SCs and Lgr5+ progenitors at postnatal day (P)1 both led to large numbers of extra HCs, especially extra inner HCs (IHCs) at P7, and these extra IHCs with normal hair bundles and synapses could survive at least to P30. The EdU assay failed to detect any EdU+ SCs, while the SC number was significantly decreased in Foxg1 cKD mice, and lineage tracing data showed that much more tdTomato+ HCs originated from Sox2+ SCs in Foxg1 cKD mice compared to the control mice. Moreover, the sphere-forming assay showed that Foxg1 cKD in Lgr5+ progenitors did not significantly change their sphere-forming ability. All these results suggest that Foxg1 cKD promotes HC regeneration and leads to large numbers of extra HCs probably by inducing direct trans-differentiation of SCs and progenitors to HCs. Real-time qPCR showed that cell cycle and Notch signaling pathways were significantly down-regulated in Foxg1 cKD mice cochlear SCs. Together, this study provides new evidence for the role of Foxg1 in regulating HC regeneration from SCs and progenitors in the neonatal mouse cochlea.


Subject(s)
Cell Transdifferentiation , Cochlea/cytology , Forkhead Transcription Factors/deficiency , Hair Cells, Auditory/cytology , Labyrinth Supporting Cells/cytology , Nerve Tissue Proteins/deficiency , Animals , Animals, Newborn , Cell Count , Cell Lineage , Cell Proliferation , Cell Survival , Cochlea/innervation , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Hair Cells, Auditory/ultrastructure , Labyrinth Supporting Cells/ultrastructure , Mechanotransduction, Cellular , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Synapses/metabolism
6.
Article in English | MEDLINE | ID: mdl-30181352

ABSTRACT

Macrophages are present in most somatic tissues, where they detect and attack invading pathogens. Macrophages also participate in many nonimmune functions, particularly those related to tissue maintenance and injury response. The sensory organs of the inner ear contain resident populations of macrophages, and additional macrophages enter the ear after acoustic trauma or ototoxicity. As expected, such macrophages participate in the clearance of cellular debris. However, otic macrophages can also influence the long-term survival of both hair cells and afferent neurons after injury. The signals that recruit macrophages into the injured ear, as well as the precise contributions of macrophages to inner ear pathology, remain to be determined.


Subject(s)
Ear, Inner/pathology , Hair Cells, Auditory/pathology , Macrophages/pathology , Animals , Apoptosis , Ear, Inner/cytology , Hair Cells, Auditory/cytology , Humans , Labyrinth Supporting Cells/cytology , Macrophages/cytology , Mammals , Models, Animal , Phagocytosis
7.
Hear Res ; 371: 75-86, 2019 01.
Article in English | MEDLINE | ID: mdl-30504093

ABSTRACT

Ca2+ is an important intracellular messenger and regulator in both physiological and pathophysiological mechanisms in the hearing organ. Investigation of cellular Ca2+ homeostasis in the mature cochlea is hampered by the special anatomy and high vulnerability of the organ. A quick, straightforward and reliable Ca2+ imaging method with high spatial and temporal resolution in the mature organ of Corti is missing. Cell cultures or isolated cells do not preserve the special microenvironment and intercellular communication, while cochlear explants are excised from only a restricted portion of the organ of Corti and usually from neonatal pre-hearing murines. The hemicochlea, prepared from hearing mice allows tonotopic experimental approach on the radial perspective in the basal, middle and apical turns of the organ. We used the preparation recently for functional imaging in supporting cells of the organ of Corti after bulk loading of the Ca2+ indicator. However, bulk loading takes long time, is variable and non-selective, and causes the accumulation of the indicator in the extracellular space. In this study we show the improved labeling of supporting cells of the organ of Corti by targeted single-cell electroporation in mature mouse hemicochlea. Single-cell electroporation proved to be a reliable way of reducing the duration and variability of loading and allowed subcellular Ca2+ imaging by increasing the signal-to-noise ratio, while cell viability was retained during the experiments. We demonstrated the applicability of the method by measuring the effect of purinergic, TRPA1, TRPV1 and ACh receptor stimulation on intracellular Ca2+ concentration at the cellular and subcellular level. In agreement with previous results, ATP evoked reversible and repeatable Ca2+ transients in Deiters', Hensen's and Claudius' cells. TRPA1 and TRPV1 stimulation by AITC and capsaicin, respectively, failed to induce any Ca2+ response in the supporting cells, except in a single Hensen's cell in which AITC evoked transients with smaller amplitude. AITC also caused the displacement of the tissue. Carbachol, agonist of ACh receptors induced Ca2+ transients in about a third of Deiters' and fifth of Hensen's cells. Here we have presented a fast and cell-specific indicator loading method allowing subcellular functional Ca2+ imaging in supporting cells of the organ of Corti in the mature hemicochlea preparation, thus providing a straightforward tool for deciphering the poorly understood regulation of Ca2+ homeostasis in these cells.


Subject(s)
Calcium/metabolism , Cochlea/cytology , Cochlea/metabolism , Adenosine Triphosphate/metabolism , Aniline Compounds/administration & dosage , Animals , Calcium Chelating Agents/administration & dosage , Calcium Signaling/drug effects , Carbachol/administration & dosage , Cochlea/drug effects , Electroporation/methods , Fluoresceins/administration & dosage , Fluorescent Dyes/administration & dosage , Fura-2/administration & dosage , In Vitro Techniques , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/drug effects , Labyrinth Supporting Cells/metabolism , Mice , Mice, Inbred BALB C , Organ of Corti/cytology , Organ of Corti/drug effects , Organ of Corti/metabolism , Receptors, Cholinergic/metabolism , Single-Cell Analysis/methods , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
8.
Development ; 145(23)2018 11 27.
Article in English | MEDLINE | ID: mdl-30389848

ABSTRACT

Lack of sensory hair cell (HC) regeneration in mammalian adults is a major contributor to hearing loss. In contrast, the neonatal mouse cochlea retains a transient capacity for regeneration, and forced Wnt activation in neonatal stages promotes supporting cell (SC) proliferation and induction of ectopic HCs. We currently know little about the temporal pattern and underlying mechanism of this age-dependent regenerative response. Using an in vitro model, we show that Wnt activation promotes SC proliferation following birth, but prior to postnatal day (P) 5. This age-dependent decline in proliferation occurs despite evidence that the Wnt pathway is postnatally active and can be further enhanced by Wnt stimulators. Using an in vivo mouse model and RNA sequencing, we show that proliferation in the early neonatal cochlea is correlated with a unique transcriptional response that diminishes with age. Furthermore, we find that augmenting Wnt signaling through the neonatal stages extends the window for HC induction in response to Notch signaling inhibition. Our results suggest that the downstream transcriptional response to Wnt activation, in part, underlies the regenerative capacity of the mammalian cochlea.


Subject(s)
Cochlea/physiology , Mammals/physiology , Regeneration/genetics , Transcription, Genetic , Wnt Signaling Pathway/genetics , Animals , Animals, Newborn , Cell Proliferation , Cell Transdifferentiation , Embryo, Mammalian/cytology , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/metabolism , Male , Mice , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , SOXB1 Transcription Factors/metabolism , TCF Transcription Factors/metabolism , beta Catenin/metabolism
9.
Nat Commun ; 8: 15046, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28492243

ABSTRACT

The adult mammalian cochlear sensory epithelium houses two major types of cells, mechanosensory hair cells and underlying supporting cells, and lacks regenerative capacity. Recent evidence indicates that a subset of supporting cells can spontaneously regenerate hair cells after ablation only within the first week postparturition. Here in vivo clonal analysis of mouse inner ear cells during development demonstrates clonal relationship between hair and supporting cells in sensory organs. We report the identification in mouse of a previously unknown population of multipotent stem/progenitor cells that are capable of not only contributing to the hair and supporting cells but also to other cell types, including glia, in cochlea undergoing development, maturation and repair in response to damage. These multipotent progenitors originate from Eya1-expressing otic progenitors. Our findings also provide evidence for detectable regenerative potential in the postnatal cochlea beyond 1 week of age.


Subject(s)
Hair Cells, Auditory/cytology , Hearing/physiology , Labyrinth Supporting Cells/cytology , Multipotent Stem Cells/cytology , Neuroglia/cytology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Differentiation , Embryo, Mammalian , Female , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Cells, Auditory/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Labyrinth Supporting Cells/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Multipotent Stem Cells/metabolism , Myosin VIIa , Myosins/genetics , Myosins/metabolism , Neuroglia/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Red Fluorescent Protein
10.
Semin Cell Dev Biol ; 65: 47-59, 2017 05.
Article in English | MEDLINE | ID: mdl-27686400

ABSTRACT

The vertebrate inner ear is a precision sensory organ, acting as both a microphone to receive sound and an accelerometer to detect gravity and motion. It consists of a series of interlinked, fluid-filled chambers containing patches of sensory epithelia, each with a specialised function. The ear contains many different differentiated cell types with distinct morphologies, from the flask-shaped hair cells found in thickened sensory epithelium, to the thin squamous cells that contribute to non-sensory structures, such as the semicircular canal ducts. Nearly all cell types of the inner ear, including the afferent neurons that innervate it, are derived from the otic placode, a region of cranial ectoderm that develops adjacent to the embryonic hindbrain. As the ear develops, the otic epithelia grow, fold, fuse and rearrange to form the complex three-dimensional shape of the membranous labyrinth. Much of our current understanding of the processes of inner ear morphogenesis comes from genetic and pharmacological manipulations of the developing ear in mouse, chicken and zebrafish embryos. These traditional approaches are now being supplemented with exciting new techniques-including force measurements and light-sheet microscopy-that are helping to elucidate the mechanisms that generate this intricate organ system.


Subject(s)
Cell Lineage/genetics , Ectoderm/cytology , Epithelial Cells/cytology , Hair Cells, Auditory/cytology , Labyrinth Supporting Cells/cytology , Organogenesis/genetics , Animals , Cell Differentiation , Cell Movement , Chick Embryo , Ectoderm/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/metabolism , Mice , Species Specificity , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish
11.
Sci Rep ; 6: 19484, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26786414

ABSTRACT

The auditory sensory epithelium, composed of mechano-sensory hair cells (HCs) and highly specialized glial-like supporting cells (SCs), is critical for our ability to detect sound. SCs provide structural and functional support to HCs and play an essential role in cochlear development, homeostasis and repair. Despite their importance, however, surprisingly little is known about the molecular mechanisms guiding SC differentiation. Here, we provide evidence that in addition to its well-characterized inhibitory function, canonical Notch signaling plays a positive, instructive role in the differentiation of SCs. Using γ-secretase inhibitor DAPT to acutely block canonical Notch signaling, we identified a cohort of Notch-regulated SC-specific genes, with diverse functions in cell signaling, cell differentiation, neuronal innervation and synaptogenesis. We validated the newly identified Notch-regulated genes in vivo using genetic gain (Emx2(Cre/+); Rosa26(N1ICD/+)) and loss-of-function approaches (Emx2(Cre/+); Rosa26(DnMAML1/+)). Furthermore, we demonstrate that Notch over-activation in the differentiating murine cochlea (Emx2(Cre/+); Rosa26(N1ICD/+)) actively promotes a SC-specific gene expression program. Finally, we show that outer SCs -so called Deiters' cells are selectively lost by prolonged reduction (Emx2(Cre/+); Rosa26(DnMAML1/+/+)) or abolishment of canonical Notch signaling (Fgfr3-iCreER; Rbpj(-/Δ)), indicating a critical role for Notch signaling in Deiters' cell development.


Subject(s)
Labyrinth Supporting Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Count , Cell Death , Cell Differentiation/genetics , Cochlea/cytology , Cochlea/embryology , Cochlea/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Labyrinth Supporting Cells/cytology , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity/genetics , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cell ; 163(6): 1348-59, 2015 12 03.
Article in English | MEDLINE | ID: mdl-26627734

ABSTRACT

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


Subject(s)
Ear, Inner/growth & development , Hair Cells, Auditory/cytology , Adenosine Triphosphate/metabolism , Animals , Anoctamin-1 , Chloride Channels/genetics , Chloride Channels/metabolism , Ear, Inner/cytology , Ear, Inner/metabolism , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/metabolism , Mice , Mice, Knockout , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Spiral Ganglion/cytology , Spiral Ganglion/metabolism
13.
Cell Rep ; 13(1): 31-42, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26387953

ABSTRACT

The organ of Corti, the auditory organ of the mammalian inner ear, contains sensory hair cells and supporting cells that arise from a common sensory progenitor. The molecular bases allowing the specification of these progenitors remain elusive. In the present study, by combining microarray analyses with conditional deletion of Dicer in the developing inner ear, we identified that miR-124 controls cell fate in the developing organ of Corti. By targeting secreted frizzled-related protein 4 (Sfrp4) and Sfrp5, two inhibitors of the Wnt pathway, we showed that miR-124 controls the ß-catenin-dependent and also the PCP-related non-canonical Wnt pathways that contribute to HC differentiation and polarization in the organ of Corti. Thus, our work emphasizes the importance of miR-124 as an epigenetic safeguard that fine-tunes the expression of genes critical for cell patterning during cochlear differentiation.


Subject(s)
Hair Cells, Auditory/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Labyrinth Supporting Cells/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , beta Catenin/genetics , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Cell Differentiation/genetics , Cell Polarity , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Embryo, Mammalian , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Labyrinth Supporting Cells/cytology , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Organogenesis/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonuclease III/deficiency , Ribonuclease III/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
14.
Proc Natl Acad Sci U S A ; 112(29): E3864-73, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26139524

ABSTRACT

Proper tissue development requires strict coordination of proliferation, growth, and differentiation. Strict coordination is particularly important for the auditory sensory epithelium, where deviations from the normal spatial and temporal pattern of auditory progenitor cell (prosensory cell) proliferation and differentiation result in abnormal cellular organization and, thus, auditory dysfunction. The molecular mechanisms involved in the timing and coordination of auditory prosensory proliferation and differentiation are poorly understood. Here we identify the RNA-binding protein LIN28B as a critical regulator of developmental timing in the murine cochlea. We show that Lin28b and its opposing let-7 miRNAs are differentially expressed in the auditory sensory lineage, with Lin28b being highly expressed in undifferentiated prosensory cells and let-7 miRNAs being highly expressed in their progeny-hair cells (HCs) and supporting cells (SCs). Using recently developed transgenic mouse models for LIN28B and let-7g, we demonstrate that prolonged LIN28B expression delays prosensory cell cycle withdrawal and differentiation, resulting in HC and SC patterning and maturation defects. Surprisingly, let-7g overexpression, although capable of inducing premature prosensory cell cycle exit, failed to induce premature HC differentiation, suggesting that LIN28B's functional role in the timing of differentiation uses let-7 independent mechanisms. Finally, we demonstrate that overexpression of LIN28B or let-7g can significantly alter the postnatal production of HCs in response to Notch inhibition; LIN28B has a positive effect on HC production, whereas let-7 antagonizes this process. Together, these results implicate a key role for the LIN28B/let-7 axis in regulating postnatal SC plasticity.


Subject(s)
Cochlea/embryology , Cochlea/metabolism , DNA-Binding Proteins/metabolism , Embryonic Development , Mammals/embryology , Mammals/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Lineage , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Epithelium/embryology , Epithelium/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Time Factors
16.
Aging (Albany NY) ; 6(6): 496-510, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25063730

ABSTRACT

Supporting cells (SCs) of the cochlear (auditory) and vestibular (balance) organs hold promise as a platform for therapeutic regeneration of the sensory hair cells. Prior data have shown proliferative restrictions of adult SCs forced to re-enter the cell cycle. By comparing juvenile and adult SCs in explant cultures, we have here studied how proliferative restrictions are linked with DNA damage signaling. Cyclin D1 overexpression, used to stimulate cell cycle re-entry, triggered higher proliferative activity of juvenile SCs. Phosphorylated form of histone H2AX (γH2AX) and p53 binding protein 1 (53BP1) were induced in a foci-like pattern in SCs of both ages as an indication of DNA double-strand break formation and activated DNA damage response. Compared to juvenile SCs, γH2AX and the repair protein Rad51 were resolved with slower kinetics in adult SCs, accompanied by increased apoptosis. Consistent with thein vitro data, in a Rb mutant mouse model in vivo, cell cycle re-entry of SCs was associated with γH2AX foci induction. In contrast to cell cycle reactivation, pharmacological stimulation of SC-to-hair-cell transdifferentiation in vitro did not trigger γH2AX. Thus, DNA damage and its prolonged resolution are critical barriers in the efforts to stimulate proliferation of the adult inner ear SCs.


Subject(s)
Adult Stem Cells/cytology , Cell Proliferation/physiology , DNA Damage/physiology , Labyrinth Supporting Cells/cytology , Signal Transduction/physiology , Animals , Mice , Organ Culture Techniques
17.
Cell Mol Neurobiol ; 34(7): 1011-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24935409

ABSTRACT

Matrix metalloproteinases (MMPs) play an important role in modeling of the extracellular matrix. There is increasing evidence that these proteases are important in neurite elongation and axonal guidance during development in the central nervous system and retina. Moreover, they are also expressed after acute injury and can be the key mediators of pathogenesis. However, the role of MMPs in the inner ear is largely unknown. Our group recently demonstrated that general inhibition of MMPs resulted in auditory hair cell loss in vitro. In the present study, we investigated the role of MMPs in inner ear spiral ganglion neuron (SGN) survival, neuritogenesis and neurite extension by blocking MMPs known to be involved in axonal guidance, neurite elongation, and apoptosis in other neuronal systems. Spiral ganglion (SG) explants from 5-day-old Wistar rats were treated with different concentrations of the general MMP inhibitor GM6001, a specific MMP-2 inhibitor, and a specific MMP-9 inhibitor, in vitro. The general inhibitor of MMPs and the specific inhibition of MMP-2 significantly reduced both the number of neurites that extended from SG explants, as well as the length of individual neurites. However, neither the general inhibitor of MMPs nor the specific inhibition of MMP-2 influenced SGN survival. Inhibition of MMP-9 had no influence on SGNs. The data suggest that MMPs, and more specifically MMP-2, influence the growth of developing afferent neurites in the mammalian inner ear in vivo.


Subject(s)
Ear, Inner/cytology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Neurons/enzymology , Spiral Ganglion/enzymology , Animals , Cell Count , Cell Survival/drug effects , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/enzymology , Matrix Metalloproteinase 9/genetics , Neurites/drug effects , Neurites/enzymology , Neurons/cytology , Neurons/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Spiral Ganglion/cytology , Spiral Ganglion/drug effects
18.
Semin Cell Dev Biol ; 24(5): 507-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23665151

ABSTRACT

Cell cycle exit and acquirement of a postmitotic state is essential for the proper development of organs. In the present review, we examine the role of the cell cycle control in the sensory epithelia of the mammalian inner ear. We describe the roles of the core cell cycle regulators in the proliferation of prosensory cells and in the initiation and maintenance of terminal mitosis of the sensory epithelia. We also discuss how other intracellular signalling may influence the cell cycle. Finally, we address the question of whether manipulations of the cell cycle may have the potential to create replacement cells for the damaged inner sensory epithelia.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle/genetics , Hair Cells, Auditory/physiology , Labyrinth Supporting Cells/physiology , Receptors, Notch/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Humans , Labyrinth Supporting Cells/cytology , Morphogenesis/physiology , Receptors, Notch/metabolism , Regeneration , Signal Transduction
19.
Semin Cell Dev Biol ; 24(5): 470-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23578865

ABSTRACT

In the inner ear, Notch signaling has been proposed to specify the sensory regions, as well as regulate the differentiation of hair cells and supporting cell within those regions. In addition, Notch plays an important role in otic neurogenesis, by determining which cells differentiate as neurons, sensory cells and non-sensory cells. Here, I review the evidence for the complex and myriad roles Notch participates in during inner ear development. A particular challenge for those studying ear development and Notch is to decipher how activation of a single pathway can lead to different outcomes within the ear, which may include changes in the intrinsic properties of the cell, Notch modulation, and potential non-canonical pathways.


Subject(s)
Calcium-Binding Proteins/genetics , Hair Cells, Auditory/physiology , Intercellular Signaling Peptides and Proteins/genetics , Labyrinth Supporting Cells/physiology , Membrane Proteins/genetics , Neurogenesis/physiology , Receptors, Notch/genetics , Sensory Receptor Cells/physiology , Signal Transduction/genetics , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Labyrinth Supporting Cells/cytology , Membrane Proteins/metabolism , Mutation , Receptors, Notch/metabolism , Sensory Receptor Cells/cytology , Serrate-Jagged Proteins
20.
Semin Cell Dev Biol ; 24(5): 448-59, 2013 May.
Article in English | MEDLINE | ID: mdl-23545368

ABSTRACT

Sensory epithelia of the inner ear contain two major cell types: hair cells and supporting cells. It has been clear for a long time that hair cells play critical roles in mechanoreception and synaptic transmission. In contrast, until recently the more abundant supporting cells were viewed as serving primarily structural and homeostatic functions. In this review, we discuss the growing information about the roles that supporting cells play in the development, function and maintenance of the inner ear, their activities in pathological states, their potential for hair cell regeneration, and the mechanisms underlying these processes.


Subject(s)
Hair Cells, Auditory/physiology , Labyrinth Supporting Cells/physiology , Sensory Receptor Cells/physiology , Transcription Factors/genetics , Animals , Cell Polarity , Cell Transdifferentiation , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Humans , Labyrinth Supporting Cells/cytology , Mechanotransduction, Cellular , Morphogenesis , Mutation , Regeneration , Sensory Receptor Cells/cytology , Synapses/physiology , Synaptic Transmission , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...