Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 606
Filter
1.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39120083

ABSTRACT

In multicellular tissues, the size and shape of cells are intricately linked with their physiological functions. In the vertebrate auditory organ, the neurosensory epithelium develops as a mosaic of sensory hair cells (HCs), and their glial-like supporting cells, which have distinct morphologies and functional properties at different frequency positions along its tonotopic long axis. In the chick cochlea, the basilar papilla (BP), proximal (high-frequency) HCs, are larger than their distal (low-frequency) counterparts, a morphological feature essential for sound perception. Mitochondrial dynamics, which constitute the equilibrium between fusion and fission, regulate differentiation and functional refinement across a variety of cell types. We investigate this as a potential mechanism for regulating the shape of developing HCs. Using live imaging in intact BP explants, we identify distinct remodelling of mitochondrial networks in proximal compared with distal HCs. Manipulating mitochondrial dynamics in developing HCs alters their normal morphology along the proximal-distal (tonotopic) axis. Inhibition of the mitochondrial fusion machinery decreased proximal HC surface area, whereas promotion of fusion increased the distal HC surface area. We identify mitochondrial dynamics as a key regulator of HC morphology in developing inner ear epithelia.


Subject(s)
Cochlea , Hair Cells, Auditory , Mitochondria , Mitochondrial Dynamics , Animals , Cochlea/embryology , Cochlea/cytology , Cochlea/growth & development , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Mitochondria/metabolism , Chick Embryo , Cell Shape , Chickens , Cell Differentiation
2.
Proc Natl Acad Sci U S A ; 121(35): e2405217121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39172791

ABSTRACT

Intercellular signaling mediated by evolutionarily conserved planar cell polarity (PCP) proteins aligns cell polarity along the tissue plane and drives polarized cell behaviors during tissue morphogenesis. Accumulating evidence indicates that the vertebrate PCP pathway is regulated by noncanonical, ß-catenin-independent Wnt signaling; however, the signaling components and mechanisms are incompletely understood. In the mouse hearing organ, both PCP and noncanonical Wnt (ncWnt) signaling are required in the developing auditory sensory epithelium to control cochlear duct elongation and planar polarity of resident sensory hair cells (HCs), including the shape and orientation of the stereociliary hair bundle essential for sound detection. We have recently discovered a Wnt/G-protein/PI3K pathway that coordinates HC planar polarity and intercellular PCP signaling. Here, we identify Wnt7b as a ncWnt ligand acting in concert with Wnt5a to promote tissue elongation in diverse developmental processes. In the cochlea, Wnt5a and Wnt7b are redundantly required for cochlear duct coiling and elongation, HC planar polarity, and asymmetric localization of core PCP proteins Fzd6 and Dvl2. Mechanistically, Wnt5a/Wnt7b-mediated ncWnt signaling promotes membrane recruitment of Daple, a nonreceptor guanine nucleotide exchange factor for Gαi, and activates PI3K/AKT and ERK signaling, which promote asymmetric Fzd6 localization. Thus, ncWnt and PCP signaling pathways have distinct mutant phenotypes and signaling components, suggesting that they act as separate, parallel pathways with nonoverlapping functions in cochlear morphogenesis. NcWnt signaling drives tissue elongation and reinforces intercellular PCP signaling by regulating the trafficking of PCP-specific Frizzled receptors.


Subject(s)
Cell Polarity , Wnt Proteins , Wnt Signaling Pathway , Wnt-5a Protein , Animals , Cell Polarity/physiology , Wnt Proteins/metabolism , Wnt Proteins/genetics , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Mice , Wnt Signaling Pathway/physiology , Cochlea/metabolism , Cochlea/cytology , Cochlea/growth & development , Hair Cells, Auditory/metabolism , Frizzled Receptors/metabolism , Frizzled Receptors/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Morphogenesis
3.
Dev Dyn ; 253(8): 771-780, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38264972

ABSTRACT

The sensory epithelium of the cochlea, the organ of Corti, has complex cytoarchitecture consisting of mechanosensory hair cells intercalated by epithelial support cells. The support cells provide important trophic and structural support to the hair cells. Thus, the support cells must be stiff yet compliant enough to withstand and modulate vibrations to the hair cells. Once the sensory cells are properly patterned, the support cells undergo significant remodeling from a simple epithelium into a structurally rigid epithelium with fluid-filled spaces in the murine cochlea. Cell adhesion molecules such as cadherins are necessary for sorting and connecting cells in an intact epithelium. To create the fluid-filled spaces, cell adhesion properties of adjoining cell membranes between cells must change to allow the formation of spaces within an epithelium. However, the dynamic localization of cadherins has not been properly analyzed as these spaces are formed. There are three cadherins that are reported to be expressed during the first postnatal week of development when the tunnel of Corti forms in the cochlea. In this study, we characterize the dynamic localization of cadherins that are associated with cytoskeletal remodeling at the contacting membranes of the inner and outer pillar cells flanking the tunnel of Corti.


Subject(s)
Cadherins , Cochlea , Animals , Cadherins/metabolism , Mice , Epithelium/metabolism , Cochlea/metabolism , Cochlea/growth & development , Cochlea/cytology , Organ of Corti/metabolism , Organ of Corti/cytology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Cell Adhesion/physiology
5.
ACS Chem Neurosci ; 14(10): 1896-1904, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37146126

ABSTRACT

Cochlear calcium (Ca2+) waves are vital regulators of the cochlear development and establishment of hearing function. Inner supporting cells are believed to be the main region generating Ca2+ waves that work as internal stimuli to coordinate the development of hair cells and the mapping of neurons in the cochlea. However, Ca2+ waves in interdental cells (IDCs) that connect to inner supporting cells and spiral ganglion neurons are rarely observed and poorly understood. Herein, we reported the mechanism of IDC Ca2+ wave formation and propagation by developing a single-cell Ca2+ excitation technology, which can easily be accomplished using a two-photon microscope for simultaneous microscopy and femtosecond laser Ca2+ excitation in any target individual cell in fresh cochlear tissues. We demonstrated that the store-operated Ca2+ channels in IDCs are responsible for Ca2+ wave formation in these cells. The specific architecture of the IDCs determines the propagation of Ca2+ waves. Our results provide the mechanism of Ca2+ formation in IDCs and a controllable, precise, and noninvasive technology to excite local Ca2+ waves in the cochlea, with good potential for research on cochlear Ca2+ and hearing functions.


Subject(s)
Calcium Signaling , Cochlea , Intracellular Calcium-Sensing Proteins , Single-Cell Analysis , Cochlea/cytology , Cochlea/growth & development , Intracellular Calcium-Sensing Proteins/physiology , Single-Cell Analysis/methods , Microscopy, Fluorescence, Multiphoton , Animals , Mice , Mice, Inbred C57BL
6.
J Histochem Cytochem ; 70(8): 583-596, 2022 08.
Article in English | MEDLINE | ID: mdl-35975307

ABSTRACT

Herein, we aimed to use double-labeling immunofluorescence to describe the expression pattern of Calbindin-D28K (CaBP28K) in the mouse cochlea from late embryonic (E) stages to the adulthood. CaBP28K was expressed in the inner hair cells (IHCs) and the greater epithelial ridge (GER) at E17. In addition, its expression was observed in the interdental cells. On postnatal day 1 (P1), CaBP28K immunoreactivity was observed in the IHCs and outer hair cells (OHCs) and was also specifically expressed in the nucleus and the cytoplasm of spiral ganglion neurons (SGNs). At P8, CaBP28K labeling disappeared from the interdental cells, and the CaBP28K-positive domain within the GER shifted from the entire cytoplasm to only the apical and basal regions. At P14, CaBP28K immunoreactivity was lost from the GER; however, its expression in the IHCs and OHCs, as well as the SGNs, persisted into adulthood. The identification of CaBP28K in the hair cells (HCs) and cuticular plates, as well as SGNs, was confirmed by its colocalization with several markers for Sox2, Myosin VIIa, Phalloidin, and Tuj1. We also detected colocalization with calmodulin in the cytoplasm of both HCs and SGNs. Western blot revealed an increase in CaBP28K postnatal expression in the mouse cochlea.


Subject(s)
Calbindin 1/genetics , Cochlea/growth & development , Neurons , Spiral Ganglion , Animals , Calbindin 1/analysis , Calbindin 1/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Neurons/metabolism
7.
Protein Pept Lett ; 29(7): 567-573, 2022.
Article in English | MEDLINE | ID: mdl-35546749

ABSTRACT

Protein tyrosine phosphatase receptor-type Q (PTPRQ), a member of the type III tyrosine phosphatase receptor (R3 PTPR) family, is composed of three domains, including 18 extracellular fibronectin type III (FN3) repeats, a transmembrane helix, and a cytoplasmic phosphotyrosine phosphatase (PTP) domain. PTPRQ was initially identified as a transcript upregulated in glomerular mesangial cells in a rat model of glomerulonephritis. Subsequently, studies found that PTPRQ has phosphotyrosine phosphatase and phosphatidylinositol phosphatase activities and can regulate cell proliferation, apoptosis, differentiation, and survival. Further in vivo studies showed that PTPRQ is necessary for the maturation of cochlear hair bundles and is considered a potential gene for deafness. In the recent two decades, 21 mutations in PTPRQ have been linked to autosomal recessive hearing loss (DFNB84) and autosomal dominant hearing loss (DFNA73). Recent mutations, deletions, and amplifications of PTPRQ have been observed in many types of cancers, which indicate that PTPRQ might play an essential role in the development of many cancers. In this review, we briefly describe PTPRQ structure and enzyme activity and focus on the correlation between PTPRQ and human disease. A profound understanding of PTPRQ could be helpful in the identification of new therapeutic targets to treat associated diseases.


Subject(s)
Cochlea/metabolism , Hearing Loss , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Animals , Cochlea/growth & development , Fibronectins , Hearing Loss/genetics , Humans , Phosphatidylinositols , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 3/chemistry
8.
J Chem Neuroanat ; 118: 102023, 2021 12.
Article in English | MEDLINE | ID: mdl-34481914

ABSTRACT

Spontaneous bursting activity is already generated in the cochlea before hearing onset and represents an important condition of the functional and anatomical organization of auditory brainstem nuclei. In the present study, cochlea ablation induced changes were characterized in auditory brainstem nuclei indirectly innervated by auditory nerve fibers before hearing onset. In Meriones unguiculatus immunohistochemical labeling of calbindin-D28k (CB) and synaptophysin (SYN) were performed. The influence of cochlea-ablation on CB or SYN was analyzed by considering their differential immunoreaction during development. During the normal postnatal development, CB was first detected in somata of the medial nucleus of the trapezoid body (MNTB) at postnatal day (P)4. The immunoreaction increased gradually in parallel to the appearance of CB-immunoreactive terminal fields in distinct superior olivary complex (SOC) nuclei. Cochlear removal at P5 or P9 in animals with 24 and 48 h survival times resulted in an increase in somatic CB-labeling in the lesioned MNTB including terminal fields compared to the non-lesioned MNTB. SYN-immunolabeling was first detected at P0 and began to strongly encircle the MNTB neurons at P4. A further progression was observed with age. Cochlear ablation resulted in a significant reduction of SYN-labeled MNTB areas of P5-cochlea-ablated gerbils after 48 h post-lesion. In P9 cochlea-ablated gerbils, a redistribution of SYN-positive terminals was seen after 24 and 48 h. Taken together, the destruction of cochlea differentially influences CB- and SYN-labeling in the MNTB, which should be considered in association with different critical periods before hearing onset.


Subject(s)
Auditory Pathways/growth & development , Calbindins/metabolism , Cochlea/physiology , Hearing/physiology , Synaptophysin/metabolism , Trapezoid Body/growth & development , Aging/physiology , Animals , Auditory Pathways/drug effects , Cochlea/growth & development , Cochlear Nucleus , Gerbillinae , Immunohistochemistry , Neurons/physiology , Olivary Nucleus/growth & development , Presynaptic Terminals/physiology , Trapezoid Body/drug effects
9.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34436572

ABSTRACT

Type 2 deiodinase (Dio2) amplifies levels of 3,5,3'-L-triiodothyronine (T3), the active form of thyroid hormone, and is essential for cochlear maturation and auditory development. However, cellular routes for endocrine signaling in the compartmentalized, anatomically complex cochlea are little understood. Dio2 generates T3 from thyroxine (T4), a more abundant thyroid hormone precursor in the circulation, and is dramatically induced in the cochlea before the onset of hearing. The evidence implies that specific Dio2-expressing cell types critically mediate T3 signaling but these cell types are poorly defined because Dio2 is expressed transiently at low levels. Here, using a Dio2CreERt2 knockin that activates a fluorescent reporter, we define Dio2-expressing cochlear cell types at high resolution in male or female mice. Dio2-positive cells were detected in vascularized supporting tissues but not in avascular internal epithelia, indicating segregation of T3-generating and T3-responding tissues. In the spiral ligament and spiral limbus, Dio2-positive fibrocytes clustered around vascular networks that convey T4 into cochlear tissues. In the otic capsule, Dio2-positive osteoblasts localized at cartilage surfaces as the bony labyrinth matures. We corroborated the identities of Dio2-positive lineages by RNA-sequencing of individual cells. The results suggest a previously unrecognized role for fibrocytes in mediating hormonal signaling. We discuss a model whereby fibrocytes mediate paracrine-like control of T3 signaling to the organ of Corti and epithelial target tissues.


Subject(s)
Cochlea/metabolism , Fibroblasts/metabolism , Iodide Peroxidase/genetics , Osteoblasts/metabolism , Animals , Animals, Newborn , Cell Lineage/genetics , Cell Tracking/methods , Cochlea/growth & development , Female , Fibroblasts/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Knock-In Techniques , Genes, Reporter , Integrases/genetics , Iodide Peroxidase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/physiology , Single-Cell Analysis , Iodothyronine Deiodinase Type II
10.
Neural Plast ; 2021: 5511010, 2021.
Article in English | MEDLINE | ID: mdl-34306061

ABSTRACT

As a general sensory disorder, hearing loss was a major concern worldwide. Autophagy is a common cellular reaction to stress that degrades cytoplasmic waste through the lysosome pathway. Autophagy not only plays major roles in maintaining intracellular homeostasis but is also involved in the development and pathogenesis of many diseases. In the auditory system, several studies revealed the link between autophagy and hearing protection. In this review, we aimed to establish the correlation between autophagy and hair cells (HCs) from the aspects of ototoxic drugs, aging, and acoustic trauma and discussed whether autophagy could serve as a potential measure in the protection of HCs.


Subject(s)
Autophagy , Hearing Loss, Sensorineural/prevention & control , Aging/genetics , Aging/physiology , Animals , Autophagy/drug effects , Cisplatin/toxicity , Cochlea/blood supply , Cochlea/growth & development , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Humans , Insulin-Like Growth Factor I/physiology , Ischemia/physiopathology , Mice , Mice, Knockout , MicroRNAs/genetics , Oxidative Stress , Resveratrol/therapeutic use , Sleep Deprivation/complications
11.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34061174

ABSTRACT

During embryonic development, the otic epithelium and surrounding periotic mesenchymal cells originate from distinct lineages and coordinate to form the mammalian cochlea. Epithelial sensory precursors within the cochlear duct first undergo terminal mitosis before differentiating into sensory and non-sensory cells. In parallel, periotic mesenchymal cells differentiate to shape the lateral wall, modiolus and pericochlear spaces. Previously, Wnt activation was shown to promote proliferation and differentiation of both otic epithelial and mesenchymal cells. Here, we fate-mapped Wnt-responsive epithelial and mesenchymal cells in mice and found that Wnt activation resulted in opposing cell fates. In the post-mitotic cochlear epithelium, Wnt activation via ß-catenin stabilization induced clusters of proliferative cells that dedifferentiated and lost epithelial characteristics. In contrast, Wnt-activated periotic mesenchyme formed ectopic pericochlear spaces and cell clusters showing a loss of mesenchymal and gain of epithelial features. Finally, clonal analyses via multi-colored fate-mapping showed that Wnt-activated epithelial cells proliferated and formed clonal colonies, whereas Wnt-activated mesenchymal cells assembled as aggregates of mitotically quiescent cells. Together, we show that Wnt activation drives transition between epithelial and mesenchymal states in a cell type-dependent manner.


Subject(s)
Cochlea/embryology , Epithelium/metabolism , Mesenchymal Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Cell Dedifferentiation , Cell Differentiation , Cell Proliferation , Cochlea/cytology , Cochlea/growth & development , Mesoderm/metabolism , Mice , Mice, Transgenic , Wnt Proteins , beta Catenin/metabolism
12.
Curr Med Sci ; 41(1): 153-157, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33582920

ABSTRACT

K+ cycling in the cochlea is critical to maintain hearing. Many sodium-potassium pumps are proved to participate in K+ cycling, such as Na/K-ATPase. The α2-Na/K-ATPase is an important isoform of Na/K-ATPase. The expression of α2-Na/K-ATPase in the cochlea is not clear. In this study, we used C57BL/6 mice as a model of presbycusis and implemented immunohistochemistry staining and quantitative real time-PCR, and the α2-Na/K-ATPase expression pattern was confirmed in the inner ear. It was found α2-Na/K-ATPase was expressed widely in cochlea and its mRNA and protein expression was gradually reduced with aging (4-, 14-, 26- and 48-weeks old mice). We suspected that, the down-regulation of α2-Na/K-ATPase expression might be associated with the remodeling of K+ cycling, degeneration of morphological structure and decrease of hearing function in aging C57 mice. In conclusion, we speculated that the reduction of α2-Na/K-ATPase might play an important role in the pathogenesis of age-related hearing loss.


Subject(s)
Aging/metabolism , Cochlea/metabolism , Hearing Loss, Sensorineural/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Aging/pathology , Animals , Cochlea/growth & development , Hearing Loss, Sensorineural/genetics , Mice , Mice, Inbred C57BL , Sodium-Potassium-Exchanging ATPase/metabolism
13.
FEBS J ; 288(1): 325-353, 2021 01.
Article in English | MEDLINE | ID: mdl-32323465

ABSTRACT

Cochlear development is a complex process with precise spatiotemporal patterns. A detailed understanding of this process is important for studies of congenital hearing loss and regenerative medicine. However, much of our understanding of cochlear development is based on rodent models. Animal models that bridge the gap between humans and rodents are needed. In this study, we investigated the development of hearing organs in a small New World monkey species, the common marmoset (Callithrix jacchus). We describe the general stages of cochlear development in comparison with those of humans and mice. Moreover, we examined more than 25 proteins involved in cochlear development and found that expression patterns were generally conserved between rodents and primates. However, several proteins involved in supporting cell processes and neuronal development exhibited interspecific expression differences. Human fetal samples for studies of primate-specific cochlear development are extremely rare, especially for late developmental stages. Our results support the use of the common marmoset as an effective alternative for analyses of primate cochlear development.


Subject(s)
Callithrix/genetics , Cochlea/metabolism , Gene Expression Regulation, Developmental , Models, Animal , Organogenesis/genetics , Animals , Aquaporin 4/genetics , Aquaporin 4/metabolism , Calbindin 1/genetics , Calbindin 1/metabolism , Callithrix/embryology , Callithrix/growth & development , Callithrix/metabolism , Cochlea/anatomy & histology , Cochlea/cytology , Cochlea/growth & development , Conserved Sequence , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Embryo, Mammalian , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Myosin VIIa/genetics , Myosin VIIa/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Peripherins/genetics , Peripherins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Species Specificity , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism , Tubulin/genetics , Tubulin/metabolism
14.
J Neurosci ; 41(4): 594-612, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33303678

ABSTRACT

Spontaneous bursts of electrical activity in the developing auditory system arise within the cochlea before hearing onset and propagate through future sound-processing circuits of the brain to promote maturation of auditory neurons. Studies in isolated cochleae revealed that this intrinsically generated activity is initiated by ATP release from inner supporting cells (ISCs), resulting in activation of purinergic autoreceptors, K+ efflux, and subsequent depolarization of inner hair cells. However, it is unknown when this activity emerges or whether different mechanisms induce activity during distinct stages of development. Here we show that spontaneous electrical activity in mouse cochlea from both sexes emerges within ISCs during the late embryonic period, preceding the onset of spontaneous correlated activity in inner hair cells and spiral ganglion neurons, which begins at birth and follows a base to apex developmental gradient. At all developmental ages, pharmacological inhibition of P2Y1 purinergic receptors dramatically reduced spontaneous activity in these three cell types. Moreover, in vivo imaging within the inferior colliculus revealed that auditory neurons within future isofrequency zones exhibit coordinated neural activity at birth. The frequency of these discrete bursts increased progressively during the postnatal prehearing period yet remained dependent on P2RY1. Analysis of mice with disrupted cholinergic signaling in the cochlea indicate that this efferent input modulates, rather than initiates, spontaneous activity before hearing onset. Thus, the auditory system uses a consistent mechanism involving ATP release from ISCs and activation of P2RY1 autoreceptors to elicit coordinated excitation of neurons that will process similar frequencies of sound.SIGNIFICANCE STATEMENT In developing sensory systems, groups of neurons that will process information from similar sensory space exhibit highly correlated electrical activity that is critical for proper maturation and circuit refinement. Defining the period when this activity is present, the mechanisms responsible and the features of this activity are crucial for understanding how spontaneous activity influences circuit development. We show that, from birth to hearing onset, the auditory system relies on a consistent mechanism to elicit correlate firing of neurons that will process similar frequencies of sound. Targeted disruption of this activity will increase our understanding of how these early circuits mature and may provide insight into processes responsible for developmental disorders of the auditory system.


Subject(s)
Auditory Pathways/growth & development , Auditory Pathways/physiology , Receptors, Purinergic/physiology , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/physiology , Cochlea/growth & development , Cochlea/physiology , Female , Hair Cells, Auditory/physiology , Hair Cells, Auditory, Inner/physiology , Inferior Colliculi/physiology , Labyrinth Supporting Cells/physiology , Male , Mice , Parasympathetic Nervous System/drug effects , Parasympathetic Nervous System/physiology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/physiology , Retina/physiology , Spiral Ganglion/physiology
15.
J Neurosci Res ; 99(2): 699-728, 2021 02.
Article in English | MEDLINE | ID: mdl-33181864

ABSTRACT

Neuronal diversity in the cochlea is largely determined by ion channels. Among voltage-gated channels, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels open with hyperpolarization and depolarize the cell until the resting membrane potential. The functions for hearing are not well elucidated and knowledge about localization is controversial. We created a detailed map of subcellular location and co-expression of all four HCN subunits across different mammalian species including CBA/J, C57Bl/6N, Ly5.1 mice, guinea pigs, cats, and human subjects. We correlated age-related hearing deterioration in CBA/J and C57Bl/6N with expression levels of HCN1, -2, and -4 in individual auditory neurons from the same cohort. Spatiotemporal expression during murine postnatal development exposed HCN2 and HCN4 involvement in a critical phase of hair cell innervation. The huge diversity of subunit composition, but lack of relevant heteromeric pairing along the perisomatic membrane and axon initial segments, highlighted an active role for auditory neurons. Neuron clusters were found to be the hot spots of HCN1, -2, and -4 immunostaining. HCN channels were also located in afferent and efferent fibers of the sensory epithelium. Age-related changes on HCN subtype expression were not uniform among mice and could not be directly correlated with audiometric data. The oldest mice groups revealed HCN channel up- or downregulation, depending on the mouse strain. The unexpected involvement of HCN channels in outer hair cell function where HCN3 overlaps prestin location emphasized the importance for auditory function. A better understanding may open up new possibilities to tune neuronal responses evoked through electrical stimulation by cochlear implants.


Subject(s)
Aging/metabolism , Cochlea/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Neurons/metabolism , Potassium Channels/physiology , Animals , Cats , Cochlea/growth & development , Evoked Potentials, Auditory, Brain Stem , Female , Gene Expression Regulation , Guinea Pigs , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/biosynthesis , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Neurons/ultrastructure , Potassium Channels/biosynthesis , Potassium Channels/genetics , Subcellular Fractions/metabolism
16.
J Neurosci ; 40(49): 9401-9413, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33127852

ABSTRACT

During cochlear development, the Notch ligand JAGGED 1 (JAG1) plays an important role in the specification of the prosensory region, which gives rise to sound-sensing hair cells and neighboring supporting cells (SCs). While JAG1's expression is maintained in SCs through adulthood, the function of JAG1 in SC development is unknown. Here, we demonstrate that JAG1 is essential for the formation and maintenance of Hensen's cells, a highly specialized SC subtype located at the edge of the auditory epithelium. Using Sox2CreERT2/+::Jag1loxP/loxP mice of both genders, we show that Jag1 deletion at the onset of differentiation, at embryonic day 14.5, disrupted Hensen's cell formation. Similar loss of Hensen's cells was observed when Jag1 was deleted after Hensen's cell formation at postnatal day (P) 0/P1 and fate-mapping analysis revealed that in the absence of Jag1, some Hensen's cells die, but others convert into neighboring Claudius cells. In support of a role for JAG1 in cell survival, genes involved in mitochondrial function and protein synthesis were downregulated in the sensory epithelium of P0 cochlea lacking Jag1 Finally, using Fgfr3-iCreERT2 ::Jag1loxP/loxP mice to delete Jag1 at P0, we observed a similar loss of Hensen's cells and found that adult Jag1 mutant mice have hearing deficits at the low-frequency range.SIGNIFICANCE STATEMENT Hensen's cells play an essential role in the development and homeostasis of the cochlea. Defects in the biophysical or functional properties of Hensen's cells have been linked to auditory dysfunction and hearing loss. Despite their importance, surprisingly little is known about the molecular mechanisms that guide their development. Morphologic and fate-mapping analyses in our study revealed that, in the absence of the Notch ligand JAGGED1, Hensen's cells died or converted into Claudius cells, which are specialized epithelium-like cells outside the sensory epithelium. Confirming a link between JAGGED1 and cell survival, transcriptional profiling showed that JAGGED1 maintains genes critical for mitochondrial function and tissue homeostasis. Finally, auditory phenotyping revealed that JAGGED1's function in supporting cells is necessary for low-frequency hearing.


Subject(s)
Cochlea/metabolism , Jagged-1 Protein/metabolism , Labyrinth Supporting Cells/physiology , Animals , Cell Survival , Cochlea/cytology , Cochlea/growth & development , Down-Regulation , Evoked Potentials, Auditory, Brain Stem , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Jagged-1 Protein/genetics , Male , Mice , Mice, Knockout , Pregnancy , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
17.
Development ; 147(12)2020 06 22.
Article in English | MEDLINE | ID: mdl-32571852

ABSTRACT

The cochlea, a coiled structure located in the ventral region of the inner ear, acts as the primary structure for the perception of sound. Along the length of the cochlear spiral is the organ of Corti, a highly derived and rigorously patterned sensory epithelium that acts to convert auditory stimuli into neural impulses. The development of the organ of Corti requires a series of inductive events that specify unique cellular characteristics and axial identities along its three major axes. Here, we review recent studies of the cellular and molecular processes regulating several aspects of cochlear development, such as axial patterning, cochlear outgrowth and cellular differentiation. We highlight how the precise coordination of multiple signaling pathways is required for the successful formation of a complete organ of Corti.


Subject(s)
Cochlea/growth & development , Animals , Auditory Perception , Cell Differentiation , Cochlea/anatomy & histology , Cochlea/metabolism , Hair Cells, Auditory/metabolism , Mitosis , Organ of Corti/anatomy & histology , Organ of Corti/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction
18.
Nat Commun ; 11(1): 2389, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32404924

ABSTRACT

Mammalian hearing requires the development of the organ of Corti, a sensory epithelium comprising unique cell types. The limited number of each of these cell types, combined with their close proximity, has prevented characterization of individual cell types and/or their developmental progression. To examine cochlear development more closely, we transcriptionally profile approximately 30,000 isolated mouse cochlear cells collected at four developmental time points. Here we report on the analysis of those cells including the identification of both known and unknown cell types. Trajectory analysis for OHCs indicates four phases of gene expression while fate mapping of progenitor cells suggests that OHCs and their surrounding supporting cells arise from a distinct (lateral) progenitor pool. Tgfßr1 is identified as being expressed in lateral progenitor cells and a Tgfßr1 antagonist inhibits OHC development. These results provide insights regarding cochlear development and demonstrate the potential value and application of this data set.


Subject(s)
Cochlea/cytology , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Outer/cytology , Hair Cells, Auditory/cytology , Organ of Corti/cytology , Animals , Cells, Cultured , Cochlea/embryology , Cochlea/growth & development , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Outer/metabolism , Mice , Organ of Corti/embryology , Organ of Corti/growth & development , Single-Cell Analysis/methods , Time Factors
19.
Int J Mol Sci ; 21(4)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32070057

ABSTRACT

In mammals Homer1, Homer2 and Homer3 constitute a family of scaffolding proteins with key roles in Ca2+ signaling and Ca2+ transport. In rodents, Homer proteins and mRNAs have been shown to be expressed in various postnatal tissues and to be enriched in brain. However, whether the Homers are expressed in developing tissues is hitherto largely unknown. In this work, we used immunohistochemistry and in situ hybridization to analyze the expression patterns of Homer1, Homer2 and Homer3 in developing cephalic structures. Our study revealed that the three Homer proteins and their encoding genes are expressed in a wide range of developing tissues and organs, including the brain, tooth, eye, cochlea, salivary glands, olfactory and respiratory mucosae, bone and taste buds. We show that although overall the three Homers exhibit overlapping distribution patterns, the proteins localize at distinct subcellular domains in several cell types, that in both undifferentiated and differentiated cells Homer proteins are concentrated in puncta and that the vascular endothelium is enriched with Homer3 mRNA and protein. Our findings suggest that Homer proteins may have differential and overlapping functions and are expected to be of value for future research aiming at deciphering the roles of Homer proteins during embryonic development.


Subject(s)
Brain/metabolism , Homer Scaffolding Proteins/genetics , Animals , Brain/growth & development , Cell Differentiation/genetics , Cochlea/growth & development , Cochlea/metabolism , Eye/growth & development , Eye/metabolism , Gene Expression Regulation, Developmental/genetics , Mice , Salivary Glands/growth & development , Salivary Glands/metabolism , Signal Transduction/genetics , Tooth/growth & development , Tooth/metabolism
20.
J Comp Neurol ; 528(12): 1967-1985, 2020 08.
Article in English | MEDLINE | ID: mdl-31994726

ABSTRACT

During inner ear development, primary auditory neurons named spiral ganglion neurons (SGNs) are surrounded by otic mesenchyme cells, which express the transcription factor Pou3f4. Mutations in Pou3f4 are associated with DFNX2, the most common form of X-linked deafness and typically include developmental malformations of the middle ear and inner ear. It is known that interactions between Pou3f4-expressing mesenchyme cells and SGNs are important for proper axon bundling during development. However, Pou3f4 continues to be expressed through later phases of development, and potential interactions between Pou3f4 and SGNs during this period had not been explored. To address this, we documented Pou3f4 protein expression in the early postnatal mouse cochlea and compared SGNs in Pou3f4 knockout mice and littermate controls. In Pou3f4y/- mice, SGN density begins to decline by the end of the first postnatal week, with approximately 25% of SGNs ultimately lost. This period of SGN loss in Pou3f4y/- cochleae coincides with significant elevations in SGN apoptosis. Interestingly, this period also coincides with the presence of a transient population of Pou3f4-expressing cells around and within the spiral ganglion. To determine if Pou3f4 is normally required for SGN peripheral axon extension into the sensory domain, we used a genetic sparse labeling approach to track SGNs and found no differences compared with controls. We also found that Pou3f4 loss did not lead to changes in the proportions of Type I SGN subtypes. Overall, these data suggest that otic mesenchyme cells may play a role in maintaining SGN populations during the early postnatal period.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , POU Domain Factors/metabolism , Spiral Ganglion/metabolism , Animals , Cell Survival , Cochlea/cytology , Cochlea/growth & development , Cochlea/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Spiral Ganglion/cytology , Spiral Ganglion/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL