Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 950
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 , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Cell Transdifferentiation/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Mice , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/cytology , Indoles , Oximes
2.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682291

ABSTRACT

The planar polarized organization of hair cells in the vestibular maculae is unique because these sensory organs contain two groups of cells with oppositely oriented stereociliary bundles that meet at a line of polarity reversal (LPR). EMX2 is a transcription factor expressed by one hair cell group that reverses the orientation of their bundles, thereby forming the LPR. We generated Emx2-CreERt2 transgenic mice for genetic lineage tracing and demonstrate Emx2 expression before hair cell specification when the nascent utricle and saccule constitute a continuous prosensory domain. Precursors labeled by Emx2-CreERt2 at this stage give rise to hair cells located along one side of the LPR in the mature utricle or saccule, indicating that this boundary is first established in the prosensory domain. Consistent with this, Emx2-CreERt2 lineage tracing in Dreher mutants, where the utricle and saccule fail to segregate, labels a continuous field of cells along one side of a fused utriculo-saccular-cochlear organ. These observations reveal that LPR positioning is pre-determined in the developing prosensory domain, and that EMX2 expression defines lineages of hair cells with oppositely oriented stereociliary bundles.


Subject(s)
Cell Lineage , Cell Polarity , Ear, Inner , Homeodomain Proteins , Mice, Transgenic , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Cell Lineage/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Cell Polarity/genetics , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology
3.
PLoS Genet ; 18(6): e1010232, 2022 06.
Article in English | MEDLINE | ID: mdl-35727824

ABSTRACT

Dync1li1, a subunit of cytoplasmic dynein 1, is reported to play important roles in intracellular retrograde transport in many tissues. However, the roles of Dync1li1 in the mammalian cochlea remain uninvestigated. Here we first studied the expression pattern of Dync1li1 in the mouse cochlea and found that Dync1li1 is highly expressed in hair cells (HCs) in both neonatal and adult mice cochlea. Next, we used Dync1li1 knockout (KO) mice to investigate its effects on hearing and found that deletion of Dync1li1 leads to early onset of progressive HC loss via apoptosis and to subsequent hearing loss. Further studies revealed that loss of Dync1li1 destabilizes dynein and alters the normal function of dynein. In addition, Dync1li1 KO results in a thinner Golgi apparatus and the accumulation of LC3+ autophagic vacuoles, which triggers HC apoptosis. We also knocked down Dync1li1 in the OC1 cells and found that the number of autophagosomes were significantly increased while the number of autolysosomes were decreased, which suggested that Dync1li1 knockdown leads to impaired transportation of autophagosomes to lysosomes and therefore the accumulation of autophagosomes results in HC apoptosis. Our findings demonstrate that Dync1li1 plays important roles in HC survival through the regulation of autophagosome transportation.


Subject(s)
Autophagosomes , Cytoplasmic Dyneins , Hair Cells, Auditory , Animals , Apoptosis/physiology , Autophagosomes/metabolism , Cochlea/cytology , Cochlea/metabolism , Cytoplasmic Dyneins/metabolism , Dyneins/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Mice
4.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119208, 2022 04.
Article in English | MEDLINE | ID: mdl-35032475

ABSTRACT

Puerarin, one of the main components of Pueraria lobata, has been reported to possess a wide range of pharmacological activities, including anti-inflammatory, antioxidative and anti-apoptotic effects. However, the role of puerarin in ototoxic drug-induced hair cell injury has not been well characterized. This study explored whether puerarin protects against cisplatin-induced hair cell damage and its potential mechanisms. The viability of puerarin-treated HEI-OC1 cells was assessed by CCK8 assay. Reactive oxygen species (ROS) was estimated with flow cytometric analysis using Cellrox Green fluorescent probe. Apoptosis-related protein levels were detected by western blot analysis. Immunostaining of the organ of Corti was performed to determine mice cochlear hair cell survival. Our results showed that puerarin improved cell viability and suppressed apoptosis in the cisplatin-damaged HEI-OC1 cells and cochlear hair cells. Mechanistic studies revealed that puerarin attenuated mitochondrial apoptosis pathway by regulating apoptotic related proteins, such as Bax and cleaved caspase-3, and attenuated ROS accumulation after cisplatin damage. Moreover, puerarin was involved in regulating the Akt pathway in HEI-OC1 cells in response to cisplatin. Our results demonstrated that puerarin administration decreased the sensitivity to apoptosis dependent on the mitochondrial apoptotic pathway by reducing ROS generation, which could be used as a new protective agent against cisplatin-induced ototoxicity.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Isoflavones/pharmacology , Mitochondria/drug effects , Animals , Caspase 3/metabolism , Cell Line , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
5.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119204, 2022 04.
Article in English | MEDLINE | ID: mdl-35026350

ABSTRACT

Cisplatin is a platinum-containing drug with ototoxicity commonly used clinically and has significant efficacy against a variety of solid tumors. One of the most important mechanisms of ototoxicity is that cisplatin induces apoptosis of hair cells. According to relevant literature, X-linked inhibitor of apoptosis protein (XIAP, anti-apoptotic protein) could inhibit the apoptotic pathway. We hypothesized that this protein might protect cochlear hair cells from cisplatin-induced injury. To figure it out, we treated cochlea of normal mice with various concentrations of cisplatin to observe the response and morphology of hair cells and determine a reasonable concentration. Next, Western Blot and quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) experiments were conducted to make an investigation about the expression of XIAP protein and mRNA. In addition, we constructed and identified XIAP overexpressing mice. Finally, we treated cochlear tissues of normal and overexpressing mice with cisplatin to investigate the cyto-protection of XIAP on hair cells, respectively. It was found that 50 µmol/L cisplatin resulted in significant loss and disorganization of hair cells, while simultaneously downregulating the protein and mRNA of XIAP. In XIAP overexpressing mice, the loss and disorganization of hair cells were significantly lessened. These results showed that XIAP can lessen cisplatin-induced hair cell loss and play a role in otoprotection.


Subject(s)
Cisplatin/pharmacology , Hair Cells, Auditory/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Down-Regulation/drug effects , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics
6.
Dev Neurobiol ; 82(1): 88-97, 2022 01.
Article in English | MEDLINE | ID: mdl-34779143

ABSTRACT

Interferon regulatory factor-7 (IRF7) is an essential regulator of both innate and adaptive immunity. It is also expressed in the otic vesicle of zebrafish embryos. However, any role for irf7 in hair cell development was uncharacterized. Does it work as a potential deaf gene to regulate hair cell development? We used whole-mount in situ hybridization (WISH) assay and morpholino-mediated gene knockdown method to investigate the role of irf7 in the development of otic vesicle hair cells during zebrafish embryogenesis. We performed RNA sequencing to gain a detailed insight into the molecules/genes which are altered upon downregulation of irf7. Compared to the wild-type siblings, knockdown of irf7 resulted in severe developmental retardation in zebrafish embryos as well as loss of neuromasts and damage to hair cells at an early stage (within 3 days post fertilization). Coinjection of zebrafish irf7 mRNA could partially rescued the defects of the morphants. atp1b2b mRNA injection can also partially rescue the phenotype induced by irf7 gene deficiency. Loss of hair cells in irf7-morphants does not result from cell apoptosis. Gene expression profiles show that, compared to wild-type, knockdown of irf7 can lead to 2053 and 2678 genes being upregulated and downregulated, respectively. Among them, 18 genes were annotated to hair cell (HC) development or posterior lateral line (PLL) development. All results suggest that irf7 plays an essential role in hair cell development in zebrafish, indicating that irf7 may be a member of deafness gene family.


Subject(s)
Hair Cells, Auditory/cytology , Interferon Regulatory Factor-7 , Zebrafish Proteins , Zebrafish , Animals , Embryonic Development , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
J Nutr Biochem ; 100: 108901, 2022 02.
Article in English | MEDLINE | ID: mdl-34748925

ABSTRACT

A nutrition deficiency is one of the various causes of hearing loss. Zinc is an essential element for cell proliferation, antioxidant reactions, and the maintenance of hearing ability. Our previous studies have reported that the auditory brainstem response (ABR) threshold is increased in mice fed with zinc-deficient diets. However, the molecular mechanism of zinc involved in auditory system remains to be elucidated. In the present study, we examined the detrimental effects of zinc deficiency on cell cycle progression in murine auditory cells (HEI-OC1). The treatment of HEI-OC1 cells with 0.5 µM TPEN (N,N,N',N'-Tetrakis (2-pyridylmethyl) ethylenediamine) for 24 h inhibited cell proliferation, accumulation of reactive oxygen species (ROS), and induction of apoptosis. The cell proliferation block was caused by a G1/S phase arrest. Supplementation of the cell growth medium with 5 µM ZnCl2 after exposure to TPEN attenuated ROS accumulation and the arrest caused by the zinc deficiency. The ABR threshold was elevated in mice fed with a zinc-deficient diet. Additionally, we observed an increased expression of p21 and decreased expression of cyclin E and pRb in the spiral ganglion (SG), the organ of Corti (OC), Limbus (L), and stria vascularis (SV) in the zinc-deficient mouse cochlea. These results indicated that zinc is an essential nutrient for proliferation via the cell cycle and that a dysregulation of the cell cycle may cause hearing loss.


Subject(s)
Cell Cycle , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Zinc/deficiency , Zinc/physiology , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Line , Cell Proliferation , Cell Survival , Chlorides/pharmacology , Cochlea/metabolism , Ethylenediamines/pharmacology , Evoked Potentials, Auditory, Brain Stem , Hearing , Homeostasis , Male , Mice , Mice, Inbred CBA , Oxidation-Reduction , Reactive Oxygen Species , Zinc Compounds/pharmacology
8.
Sci Rep ; 11(1): 23855, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903829

ABSTRACT

ATP-utilizing enzymes play key roles in hair bundles, the mechanically sensitive organelles of sensory hair cells in the inner ear. We used a fluorescent ATP analog, EDA-ATP-Cy3 (Cy3-ATP), to label ATP-binding proteins in two different preparations of unfixed hair-cell stereocilia of the mouse. In the first preparation, we lightly permeabilized dissected cochleas, then labeled them with Cy3-ATP. Hair cells and their stereocilia remained intact, and stereocilia tips in rows 1 and 2 were labeled particularly strongly with Cy3-ATP. In many cases, vanadate (Vi) traps nucleotides at the active site of myosin isoforms and presents nucleotide dissociation. Co-application with Vi enhanced the tip labeling, which is consistent with myosin isoforms being responsible. By contrast, the actin polymerization inhibitors latrunculin A and cytochalasin D had no effect, suggesting that actin turnover at stereocilia tips was not involved. Cy3-ATP labeling was substantially reduced-but did not disappear altogether-in mutant cochleas lacking MYO15A; by contrast, labeling remained robust in cochleas lacking MYO7A. In the second preparation, used to quantify Cy3-ATP labeling, we labeled vestibular stereocilia that had been adsorbed to glass, which demonstrated that tip labeling was higher in longer stereocilia. We found that tip signal was reduced by ~ 50% in Myo15ash2/sh2 stereocilia as compared to Myo15ash2/+stereocilia. These results suggest that MYO15A accounts for a substantial fraction of the Cy3-ATP tip labeling in vestibular hair cells, and so this novel preparation could be utilized to examine the control of MYO15A ATPase activity in situ.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Hair Cells, Auditory/metabolism , Indoles/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Cytochalasin D/pharmacology , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Mice , Mice, Inbred C57BL , Myosins/metabolism , Stereocilia/metabolism , Stereocilia/ultrastructure , Thiazolidines/pharmacology , Vanadates/pharmacology
9.
Neurosci Lett ; 764: 136282, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34619343

ABSTRACT

Evaluation of hearing loss patients using clinical audiometry has been unable to give a definitive cellular or molecular diagnosis, hampering the development of treatments of sensorineural hearing loss. However, biopsy of inner ear tissue without losing residual hearing function for pathologic diagnosis is extremely challenging. In a clinical setting, perilymph can be accessed, potentially allowing the development of fluid based diagnostic tests. Recent approaches to improving inner ear diagnostics have been focusing on the evaluation of the proteomic or miRNA profiles of perilymph. Inspired by recent characterization and classification of many neurodegenerative diseases using exosomes which not only are produced in locally in diseased tissue but are transported beyond the blood brain barrier, we demonstrate the isolation of human inner ear specific exosomes using a novel ultrasensitive immunomagnetic nano pom-poms capture-release approach. Using perilymph samples harvested from surgical procedures, we were able to isolate exosomes from sensorineural hearing loss patients in only 2-5 µL of perilymph. By isolating sensory hair cell derived exosomes through their expression level of myosin VIIa, we for the first-time sample material from hair cells in the living human inner ear. This work sets up the first demonstration of immunomagnetic capture-release nano pom-pom isolated exosomes for liquid biopsy diagnosis of sensorineural hearing loss. With the ability to isolate exosomes derived from different cell types for molecular characterization, this method also can be developed for analyzing exosomal biomarkers from more accessible patient tissue fluids such as plasma.


Subject(s)
Exosomes/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/diagnosis , Perilymph/cytology , Adult , Audiometry , Cell Fractionation , Feasibility Studies , Female , Hair Cells, Auditory/cytology , Hearing Loss, Sensorineural/pathology , Humans , Immunomagnetic Separation , Liquid Biopsy/methods , Male , Middle Aged
10.
Stem Cell Reports ; 16(9): 2257-2273, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525385

ABSTRACT

Hair cell degeneration is a major cause of sensorineural hearing loss. Hair cells in mammalian cochlea do not spontaneously regenerate, posing a great challenge for restoration of hearing. Here, we establish a robust, high-throughput cochlear organoid platform that facilitates 3D expansion of cochlear progenitor cells and differentiation of hair cells in a temporally regulated manner. High-throughput screening of the FDA-approved drug library identified regorafenib, a VEGFR inhibitor, as a potent small molecule for hair cell differentiation. Regorafenib also promotes reprogramming and maturation of hair cells in both normal and neomycin-damaged cochlear explants. Mechanistically, inhibition of VEGFR suppresses TGFB1 expression via the MEK pathway and TGFB1 downregulation directly mediates the effect of regorafenib on hair cell reprogramming. Our study not only demonstrates the power of a cochlear organoid platform in high-throughput analyses of hair cell physiology but also highlights VEGFR-MEK-TGFB1 signaling crosstalk as a potential target for hair cell regeneration and hearing restoration.


Subject(s)
Cellular Reprogramming , Cochlea/metabolism , High-Throughput Screening Assays , Mitogen-Activated Protein Kinase Kinases/metabolism , Organoids/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Culture Techniques, Three Dimensional/methods , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Cochlea/cytology , Drug Discovery/methods , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Mice , Mice, Transgenic , Organoids/cytology , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
11.
Chaos ; 31(7): 073142, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34340330

ABSTRACT

Coupled hair cells of the auditory and vestibular systems perform the crucial task of converting the energy of sound waves and ground-borne vibrations into ionic currents. We mechanically couple groups of living, active hair cells with artificial membranes, thus mimicking in vitro the coupled dynamical system. We identify chimera states and frequency clustering in the dynamics of these coupled nonlinear, autonomous oscillators. We find that these dynamical states can be reproduced by our numerical model with heterogeneity of the parameters. Furthermore, we find that this model is most sensitive to external signals when poised at the onset of synchronization, where chimera and cluster states are likely to form. We, therefore, propose that the partial synchronization in our experimental system is a manifestation of a system poised at the verge of synchronization with optimal sensitivity.


Subject(s)
Ear, Inner , Hair Cells, Auditory , Cluster Analysis , Ear, Inner/cytology , Hair Cells, Auditory/cytology , Membranes, Artificial
12.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34266958

ABSTRACT

During embryonic development, hierarchical cascades of transcription factors interact with lineage-specific chromatin structures to control the sequential steps in the differentiation of specialized cell types. While examples of transcription factor cascades have been well documented, the mechanisms underlying developmental changes in accessibility of cell type-specific enhancers remain poorly understood. Here, we show that the transcriptional "master regulator" ATOH1-which is necessary for the differentiation of two distinct mechanoreceptor cell types, hair cells in the inner ear and Merkel cells of the epidermis-is unable to access much of its target enhancer network in the progenitor populations of either cell type when it first appears, imposing a block to further differentiation. This block is overcome by a feed-forward mechanism in which ATOH1 first stimulates expression of POU4F3, which subsequently acts as a pioneer factor to provide access to closed ATOH1 enhancers, allowing hair cell and Merkel cell differentiation to proceed. Our analysis also indicates the presence of both shared and divergent ATOH1/POU4F3-dependent enhancer networks in hair cells and Merkel cells. These cells share a deep developmental lineage relationship, deriving from their common epidermal origin, and suggesting that this feed-forward mechanism preceded the evolutionary divergence of these very different mechanoreceptive cell types.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Mechanoreceptors/metabolism , Transcription Factor Brn-3C/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cochlea/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Hair Cells, Auditory/cytology , Homeodomain Proteins/genetics , Humans , Merkel Cells/metabolism , Mice , Transcription Factor Brn-3C/genetics
13.
Cell Rep ; 36(2): 109358, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260939

ABSTRACT

The utricle is a vestibular sensory organ that requires mechanosensitive hair cells to detect linear acceleration. In neonatal mice, new hair cells are derived from non-sensory supporting cells, yet cell type diversity and mechanisms of cell addition remain poorly characterized. Here, we perform computational analyses on single-cell transcriptomes to categorize cell types and resolve 14 individual sensory and non-sensory subtypes. Along the periphery of the sensory epithelium, we uncover distinct groups of transitional epithelial cells, marked by Islr, Cnmd, and Enpep expression. By reconstructing de novo trajectories and gene dynamics, we show that as the utricle expands, Islr+ transitional epithelial cells exhibit a dynamic and proliferative phase to generate new supporting cells, followed by coordinated differentiation into hair cells. Taken together, our study reveals a sequential and coordinated process by which non-sensory epithelial cells contribute to growth of the postnatal mouse sensory epithelium.


Subject(s)
Ear, Inner/cytology , Sensation/genetics , Single-Cell Analysis , Transcriptome/genetics , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , Epithelial Cells/cytology , Hair Cells, Auditory/cytology , Mice , Reproducibility of Results , Saccule and Utricle/cytology , Transcription, Genetic
14.
Sci Rep ; 11(1): 13893, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230535

ABSTRACT

Several studies have shown how different cell lines can influence the differentiation of stem cells through co-culture systems. The House Ear Institute-Organ of Corti 1 (HEI-OC1) is considered an important cell line for in vitro auditory research. However, it is unknown if HEI-OC1 cells can promote the differentiation of embryonic stem cells (ESCs). In this study, we investigated whether co-culture of ESCs with HEI-OC1 cells promotes differentiation. To this end, we developed a co-culture system of mouse ESCs with HEI-OC1 cells. Dissociated or embryonic bodies (EBs) of ESCs were introduced to a conditioned and inactivated confluent layer of HEI-OC1 cells for 14 days. The dissociated ESCs coalesced into an EB-like form that was smaller than the co-cultured EBs. Contact co-culture generated cells expressing several otic progenitor markers as well as hair cell specific markers. ESCs and EBs were also cultured in non-contact setup but using conditioned medium from HEI-OC1 cells, indicating that soluble factors alone could have a similar effect. The ESCs did not form into aggregates but were still Myo7a-positive, while the EBs degenerated. However, in the fully differentiated EBs, evidence to prove mature differentiation of inner ear hair cell was still rudimentary. Nevertheless, these results suggest that cellular interactions between ESCs and HEI-OC1 cells may both stimulate ESC differentiation.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Hair Cells, Auditory/cytology , Animals , Biomarkers/metabolism , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Epithelium/metabolism , Gene Expression Regulation/drug effects , Mice , Myosin VIIa/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism
15.
Cell Tissue Res ; 386(2): 321-333, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34319434

ABSTRACT

Human otic organoids generated from pluripotent stem cells (PSCs) provide a promising platform for modeling, drug testing, and cell-based therapies of inner ear diseases. However, providing the appropriate niche that resembles inner ear development and its vasculature to generate otic organoids is less conspicuous. Here, we devised a strategy to enhance maturation of otic progenitor cells toward human hair cell-like cells (HCLCs) by assembling three-dimensional (3D) otic organoids that contain human PSC-derived otic cells, endothelial cells, and mesenchymal stem cells (MSCs). Heterotopic implantation of otic organoids, designated as grafted otic organoids (GOs), in ex ovo chick embryo chorioallantoic membrane (CAM) stimulated maturation of the HCLCs. Functional analysis revealed the presence of voltage-gated potassium currents without detectable sodium currents in these cells in the GOs. Our results demonstrated that implantation of 3D heterotypic cell mixtures of otic organoids improved maturation of human HCLCs. This GO-derived HCLCs could be an attractive source for drug discovery and other biomedical applications.


Subject(s)
Hair Cells, Auditory/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Chick Embryo , Ear, Inner/cytology , Humans
16.
Dev Cell ; 56(17): 2471-2485.e5, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34331868

ABSTRACT

Adult mammalian tissues such as heart, brain, retina, and the sensory structures of the inner ear do not effectively regenerate, although a latent capacity for regeneration exists at embryonic and perinatal times. We explored the epigenetic basis for this latent regenerative potential in the mouse inner ear and its rapid loss during maturation. In perinatal supporting cells, whose fate is maintained by Notch-mediated lateral inhibition, the hair cell enhancer network is epigenetically primed (H3K4me1) but silenced (active H3K27 de-acetylation and trimethylation). Blocking Notch signaling during the perinatal period of plasticity rapidly eliminates epigenetic silencing and allows supporting cells to transdifferentiate into hair cells. Importantly, H3K4me1 priming of the hair cell enhancers in supporting cells is removed during the first post-natal week, coinciding with the loss of transdifferentiation potential. We hypothesize that enhancer decommissioning during cochlear maturation contributes to the failure of hair cell regeneration in the mature organ of Corti.


Subject(s)
Cell Differentiation/physiology , Hair Cells, Auditory/metabolism , Receptors, Notch/metabolism , Regeneration/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Transdifferentiation/genetics , Cell Transdifferentiation/physiology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Hair Cells, Auditory/cytology , Mice, Transgenic , Regulatory Sequences, Nucleic Acid/genetics
17.
Neural Plast ; 2021: 9950533, 2021.
Article in English | MEDLINE | ID: mdl-34122536

ABSTRACT

As part of the inner ear, the vestibular system is responsible for sense of balance, which consists of three semicircular canals, the utricle, and the saccule. Increasing evidence has indicated that the noncanonical Wnt/PCP signaling pathway plays a significant role in the development of the polarity of the inner ear. However, the role of canonical Wnt signaling in the polarity of the vestibule is still not completely clear. In this study, we found that canonical Wnt pathway-related genes are expressed in the early stage of development of the utricle and change dynamically. We conditionally knocked out ß-catenin, a canonical Wnt signaling core protein, and found that the cilia orientation of hair cells was disordered with reduced number of hair cells in the utricle. Moreover, regulating the canonical Wnt pathway (Licl and IWP2) in vitro also affected hair cell polarity and indicated that Axin2 may be important in this process. In conclusion, our results not only confirm that the regulation of canonical Wnt signaling affects the number of hair cells in the utricle but also provide evidence for its role in polarity development.


Subject(s)
Hair Cells, Auditory/physiology , Saccule and Utricle/cytology , Wnt Signaling Pathway/physiology , Animals , Axin Protein/analysis , Cell Polarity , Female , Gene Knockout Techniques , Hair Cells, Auditory/cytology , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Saccule and Utricle/embryology , Saccule and Utricle/physiology , beta Catenin/deficiency , beta Catenin/physiology
18.
Nat Commun ; 12(1): 2861, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001891

ABSTRACT

Hair cells detect sound, head position or water movements when their mechanosensory hair bundle is deflected. Each hair bundle has an asymmetric architecture that restricts stimulus detection to a single axis. Coordinated hair cell orientations within sensory epithelia further tune stimulus detection at the organ level. Here, we identify GPR156, an orphan GPCR of unknown function, as a critical regulator of hair cell orientation. We demonstrate that the transcription factor EMX2 polarizes GPR156 distribution, enabling it to signal through Gαi and trigger a 180° reversal in hair cell orientation. GPR156-Gαi mediated reversal is essential to establish hair cells with mirror-image orientations in mouse otolith organs in the vestibular system and in zebrafish lateral line. Remarkably, GPR156-Gαi also instructs hair cell reversal in the auditory epithelium, despite a lack of mirror-image organization. Overall, our work demonstrates that conserved GPR156-Gαi signaling is integral to the framework that builds directional responses into mechanosensory epithelia.


Subject(s)
Epithelium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism , Animals , Cell Polarity/genetics , Female , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Hair Cells, Auditory/cytology , Homeodomain Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal/methods , Receptors, G-Protein-Coupled/genetics , Transcription Factors/genetics , Zebrafish/metabolism
19.
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
20.
Aging (Albany NY) ; 13(8): 11678-11695, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33882456

ABSTRACT

Piccolo is a presynaptic protein with high conservation among different species, and the expression of Piccolo is extensive in vertebrates. Recently, a small fragment of Piccolo (Piccolino), arising due to the incomplete splicing of intron 5/6, was found to be present in the synapses of retinas and cochleae. However, the comprehensive function of Piccolo in the retina and cochlea remains unclear. In this study, we generated Piccolo knockout mice using CRISPR-Cas9 technology to explore the function of Piccolo. Unexpectedly, whereas no abnormalities were found in the cochlear hair cells of the mutant mice, significant differences were found in the retinas, in which two layers (the outer nuclear layer and the outer plexiform layer) were absent. Additionally, the amplitudes of electroretinograms were significantly reduced and pigmentation was observed in the fundoscopy of the mutant mouse retinas. The expression levels of Bassoon, a homolog of Piccolo, as well as synapse-associated proteins CtBP1, CtBP2, Kif3A, and Rim1 were down-regulated. The numbers of ribbon synapses in the retinas of the mutant mice were also reduced. Altogether, the phenotype of Piccolo-/- mice resembled the symptoms of retinitis pigmentosa (RP) in humans, suggesting Piccolo might be a candidate gene of RP and indicates Piccolo knockout mice are a good model for elucidating the molecular mechanisms of RP.


Subject(s)
Cytoskeletal Proteins/metabolism , Hair Cells, Auditory/metabolism , Neuropeptides/metabolism , Retina/pathology , Retinitis Pigmentosa/genetics , Animals , Cytoskeletal Proteins/genetics , Disease Models, Animal , Female , Hair Cells, Auditory/cytology , Humans , Introns/genetics , Male , Mice , Mice, Knockout , Neuropeptides/genetics , RNA Splicing , Retina/cytology , Retinitis Pigmentosa/pathology , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...