Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 960
Filter
1.
PLoS One ; 19(7): e0305742, 2024.
Article in English | MEDLINE | ID: mdl-39028743

ABSTRACT

In vivo gene delivery to tissues using adeno-associated vector (AAVs) has revolutionized the field of gene therapy. Yet, while sensorineural hearing loss is one of the most common sensory disorders worldwide, gene therapy applied to the human inner ear is still in its infancy. Recent advances in the development recombinant AAVs have significantly improved their cell tropism and transduction efficiency across diverse inner ear cell types to a level that renders this tool valuable for conditionally manipulating gene expression in the context of developmental biology studies of the mouse inner ear. Here, we describe a protocol for in utero micro-injection of AAVs into the embryonic inner ear, using the AAV-PHP.eB and AAV-DJ serotypes that respectively target the sensory hair cells and the supporting cells of the auditory sensory epithelium. We also aimed to standardize procedures for imaging acquisition and image analysis to foster research reproducibility and allow accurate comparisons between studies. We find that AAV-PHP.eB and AAV-DJ provide efficient and reliable tools for conditional gene expression targeting cochlear sensory and supporting cells in the mouse inner ear, from late embryonic stages on.


Subject(s)
Dependovirus , Ear, Inner , Gene Transfer Techniques , Genetic Vectors , Animals , Dependovirus/genetics , Mice , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Female , Transduction, Genetic/methods , Pregnancy , Genetic Therapy/methods , Humans
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 , Transcription Factors , Animals , Mice , Cell Lineage/genetics , Cell Polarity/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice, Transgenic , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Transcription Factors/metabolism , Transcription Factors/genetics
3.
Proc Natl Acad Sci U S A ; 119(28): e2118938119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35867760

ABSTRACT

The vertebrate inner ear arises from a pool of progenitors with the potential to contribute to all the sense organs and cranial ganglia in the head. Here, we explore the molecular mechanisms that control ear specification from these precursors. Using a multiomics approach combined with loss-of-function experiments, we identify a core transcriptional circuit that imparts ear identity, along with a genome-wide characterization of noncoding elements that integrate this information. This analysis places the transcription factor Sox8 at the top of the ear determination network. Introducing Sox8 into the cranial ectoderm not only converts non-ear cells into ear progenitors but also activates the cellular programs for ear morphogenesis and neurogenesis. Thus, Sox8 has the unique ability to remodel transcriptional networks in the cranial ectoderm toward ear identity.


Subject(s)
Ear, Inner , Ectoderm , Gene Expression Regulation, Developmental , SOXE Transcription Factors , Animals , Ear, Inner/embryology , Ectoderm/embryology , SOXE Transcription Factors/physiology , Skull , Vertebrates/embryology
4.
PLoS Comput Biol ; 17(11): e1009063, 2021 11.
Article in English | MEDLINE | ID: mdl-34723957

ABSTRACT

A common feature of morphogenesis is the formation of three-dimensional structures from the folding of two-dimensional epithelial sheets, aided by cell shape changes at the cellular-level. Changes in cell shape must be studied in the context of cell-polarised biomechanical processes within the epithelial sheet. In epithelia with highly curved surfaces, finding single-cell alignment along a biological axis can be difficult to automate in silico. We present 'Origami', a MATLAB-based image analysis pipeline to compute direction-variant cell shape features along the epithelial apico-basal axis. Our automated method accurately computed direction vectors denoting the apico-basal axis in regions with opposing curvature in synthetic epithelia and fluorescence images of zebrafish embryos. As proof of concept, we identified different cell shape signatures in the developing zebrafish inner ear, where the epithelium deforms in opposite orientations to form different structures. Origami is designed to be user-friendly and is generally applicable to fluorescence images of curved epithelia.


Subject(s)
Cell Shape/physiology , Image Processing, Computer-Assisted/statistics & numerical data , Models, Biological , Animals , Biomechanical Phenomena , Cell Polarity , Computational Biology , Computer Simulation , Ear, Inner/embryology , Epithelium/embryology , Imaging, Three-Dimensional , Microscopy, Fluorescence , Morphogenesis , Proof of Concept Study , Software , Zebrafish/embryology
5.
Development ; 148(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34414417

ABSTRACT

Branchio-oto-renal syndrome (BOR) is a disorder characterized by hearing loss, and craniofacial and/or renal defects. Variants in the transcription factor Six1 and its co-factor Eya1, both of which are required for otic development, are linked to BOR. We previously identified Sobp as a potential Six1 co-factor, and SOBP variants in mouse and humans cause otic phenotypes; therefore, we asked whether Sobp interacts with Six1 and thereby may contribute to BOR. Co-immunoprecipitation and immunofluorescence experiments demonstrate that Sobp binds to and colocalizes with Six1 in the cell nucleus. Luciferase assays show that Sobp interferes with the transcriptional activation of Six1+Eya1 target genes. Experiments in Xenopus embryos that either knock down or increase expression of Sobp show that it is required for formation of ectodermal domains at neural plate stages. In addition, altering Sobp levels disrupts otic vesicle development and causes craniofacial cartilage defects. Expression of Xenopus Sobp containing the human variant disrupts the pre-placodal ectoderm similar to full-length Sobp, but other changes are distinct. These results indicate that Sobp modifies Six1 function and is required for vertebrate craniofacial development, and identify Sobp as a potential candidate gene for BOR.


Subject(s)
Bone Development , Homeodomain Proteins/metabolism , Metalloproteins/metabolism , Nuclear Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Branchio-Oto-Renal Syndrome/embryology , Branchio-Oto-Renal Syndrome/genetics , Cell Nucleus/metabolism , Ear, Inner/embryology , Ear, Inner/metabolism , Ectoderm/embryology , Ectoderm/metabolism , Gene Expression , Homeodomain Proteins/genetics , Larva/growth & development , Metalloproteins/genetics , Neural Crest/embryology , Neural Crest/metabolism , Nuclear Proteins/genetics , Protein Binding , Protein Tyrosine Phosphatases/metabolism , Transcriptional Activation , Xenopus Proteins/genetics , Xenopus laevis
6.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204426

ABSTRACT

The last decade has witnessed the identification of several families affected by hereditary non-syndromic hearing loss (NSHL) caused by mutations in the SMPX gene and the loss of function has been suggested as the underlying mechanism. In the attempt to confirm this hypothesis we generated an Smpx-deficient zebrafish model, pointing out its crucial role in proper inner ear development. Indeed, a marked decrease in the number of kinocilia together with structural alterations of the stereocilia and the kinocilium itself in the hair cells of the inner ear were observed. We also report the impairment of the mechanotransduction by the hair cells, making SMPX a potential key player in the construction of the machinery necessary for sound detection. This wealth of evidence provides the first possible explanation for hearing loss in SMPX-mutated patients. Additionally, we observed a clear muscular phenotype consisting of the defective organization and functioning of muscle fibers, strongly suggesting a potential role for the protein in the development of muscle fibers. This piece of evidence highlights the need for more in-depth analyses in search for possible correlations between SMPX mutations and muscular disorders in humans, thus potentially turning this non-syndromic hearing loss-associated gene into the genetic cause of dysfunctions characterized by more than one symptom, making SMPX a novel syndromic gene.


Subject(s)
Ear, Inner/embryology , Ear, Inner/metabolism , Muscle Proteins/deficiency , Muscles/embryology , Muscles/metabolism , Zebrafish/embryology , Zebrafish/genetics , Animals , Embryonic Development , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Hair Cells, Auditory/metabolism , Mechanotransduction, Cellular/genetics , Muscle Development/genetics , Organogenesis/genetics , Phenotype , Protein Transport
7.
Development ; 148(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33795231

ABSTRACT

All epithelial components of the inner ear, including sensory hair cells and innervating afferent neurons, arise by patterning and differentiation of epithelial progenitors residing in a simple sphere, the otocyst. Here, we identify the transcriptional repressors TBX2 and TBX3 as novel regulators of these processes in the mouse. Ablation of Tbx2 from the otocyst led to cochlear hypoplasia, whereas loss of Tbx3 was associated with vestibular malformations. The loss of function of both genes (Tbx2/3cDKO) prevented inner ear morphogenesis at midgestation, resulting in indiscernible cochlear and vestibular structures at birth. Morphogenetic impairment occurred concomitantly with increased apoptosis in ventral and lateral regions of Tbx2/3cDKO otocysts around E10.5. Expression analyses revealed partly disturbed regionalisation, and a posterior-ventral expansion of the neurogenic domain in Tbx2/3cDKO otocysts at this stage. We provide evidence that repression of FGF signalling by TBX2 is important to restrict neurogenesis to the anterior-ventral otocyst and implicate another T-box factor, TBX1, as a crucial mediator in this regulatory network.


Subject(s)
Apoptosis , Ear, Inner/embryology , Gene Expression Regulation, Developmental , Organogenesis , Signal Transduction , T-Box Domain Proteins/biosynthesis , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Mice , Mice, Knockout , T-Box Domain Proteins/genetics
8.
Elife ; 102021 03 11.
Article in English | MEDLINE | ID: mdl-33704062

ABSTRACT

The auditory and vestibular organs of the inner ear and the neurons that innervate them originate from Sox2-positive and Notch-active neurosensory domains specified at early stages of otic development. Sox2 is initially present throughout the otic placode and otocyst, and then it becomes progressively restricted to a ventro-medial domain. Using gain- and loss-of-function approaches in the chicken otocyst, we show that these early changes in Sox2 expression are regulated in a dose-dependent manner by Wnt/beta-catenin signalling. Both high and very low levels of Wnt activity repress Sox2 and neurosensory competence. However, intermediate levels allow the maintenance of Sox2 expression and sensory organ formation. We propose that a dorso-ventral (high-to-low) gradient and wave of Wnt activity initiated at the dorsal rim of the otic placode progressively restricts Sox2 and Notch activity to the ventral half of the otocyst, thereby positioning the neurosensory competent domains in the inner ear.


Subject(s)
Avian Proteins/genetics , Chickens/genetics , Ear, Inner/embryology , Gene Expression Regulation , SOXB1 Transcription Factors/genetics , Wnt Signaling Pathway , Animals , Avian Proteins/metabolism , Chick Embryo/embryology , Chickens/metabolism , Ear, Inner/metabolism , Gain of Function Mutation , Loss of Function Mutation , SOXB1 Transcription Factors/metabolism
9.
Dev Dyn ; 250(10): 1450-1462, 2021 10.
Article in English | MEDLINE | ID: mdl-33715274

ABSTRACT

BACKGROUND: Eya2 expression during mouse development has been studied by in situ hybridization and it has been shown to be involved skeletal muscle development and limb formation. Here, we generated Eya2 knockout (Eya2- ) and a lacZ knockin reporter (Eya2lacZ ) mice and performed a detailed expression analysis for Eya2lacZ at different developmental stages to trace Eya2lacZ -positive cells in Eya2-null mice. We describe that Eya2 is not only expressed in cranial sensory and dorsal root ganglia, retina and olfactory epithelium, and somites as previously reported, but also Eya2 is specifically detected in other organs during mouse development. RESULTS: We found that Eya2 is expressed in ocular and trochlear motor neurons. In the inner ear, Eya2lacZ is specifically expressed in differentiating hair cells in both vestibular and cochlear sensory epithelia of the inner ear and Eya2-/- or Eya2lacZ/lacZ mice displayed mild hearing loss. Furthermore, we detected Eya2 expression during both salivary gland and thymus development and Eya2-null mice had a smaller thymus. CONCLUSIONS: As Eya2 is coexpressed with other members of the Eya family genes, these results together highlight that Eya2 as a potential regulator may act synergistically with other Eya genes to regulate the differentiation of the inner ear sensory hair cells and the formation of the salivary gland and thymus.


Subject(s)
Ear, Inner/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Hearing Loss/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Cell Differentiation/physiology , Ear, Inner/embryology , Hearing Loss/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Protein Tyrosine Phosphatases/genetics
10.
Viruses ; 13(1)2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33419104

ABSTRACT

Hearing loss, one of the most prevalent chronic health conditions, affects around half a billion people worldwide, including 34 million children. The World Health Organization estimates that the prevalence of disabling hearing loss will increase to over 900 million people by 2050. Many cases of congenital hearing loss are triggered by viral infections during different stages of pregnancy. However, the molecular mechanisms by which viruses induce hearing loss are not sufficiently explored, especially cases that are of embryonic origins. The present review first describes the cellular and molecular characteristics of the auditory system development at early stages of embryogenesis. These developmental hallmarks, which initiate upon axial specification of the otic placode as the primary root of the inner ear morphogenesis, involve the stage-specific regulation of several molecules and pathways, such as retinoic acid signaling, Sonic hedgehog, and Wnt. Different RNA and DNA viruses contributing to congenital and acquired hearing loss are then discussed in terms of their potential effects on the expression of molecules that control the formation of the auditory and vestibular compartments following otic vesicle differentiation. Among these viruses, cytomegalovirus and herpes simplex virus appear to have the most effect upon initial molecular determinants of inner ear development. Moreover, of the molecules governing the inner ear development at initial stages, SOX2, FGFR3, and CDKN1B are more affected by viruses causing either congenital or acquired hearing loss. Abnormalities in the function or expression of these molecules influence processes like cochlear development and production of inner ear hair and supporting cells. Nevertheless, because most of such virus-host interactions were studied in unrelated tissues, further validations are needed to confirm whether these viruses can mediate the same effects in physiologically relevant models simulating otic vesicle specification and growth.


Subject(s)
Cytomegalovirus/isolation & purification , Ear, Inner/embryology , Ear, Inner/virology , Hearing Loss/virology , Simplexvirus/isolation & purification , Animals , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytomegalovirus/pathogenicity , Hearing Loss/congenital , Humans , Receptor, Fibroblast Growth Factor, Type 3/genetics , SOXB1 Transcription Factors/genetics , Signal Transduction , Simplexvirus/pathogenicity
11.
Mol Med Rep ; 23(3)2021 03.
Article in English | MEDLINE | ID: mdl-33398372

ABSTRACT

Brain 4 (Brn4) is a transcription factor belonging to the POU3 family, and it is important for the embryonic development of the neural tube, inner ear and pancreas. In addition, it serves a crucial role in neural stem cell differentiation and reprogramming. The present review aimed to summarize the chromosomal location, species homology, protein molecular structure and tissue distribution of Brn4, in addition to its biological processes, with the aim of providing a reference of its structure and function for further studies, and its potential use as a gene therapy target.


Subject(s)
Ear, Inner/embryology , Embryonic Development , Neural Tube/embryology , POU Domain Factors , Pancreas/embryology , Animals , Humans , POU Domain Factors/genetics , POU Domain Factors/metabolism
12.
Dev Biol ; 471: 65-75, 2021 03.
Article in English | MEDLINE | ID: mdl-33316259

ABSTRACT

The function of the inner ear depends on the maintenance of high concentrations of K+ ions. The slow-inactivating delayed rectifier Kv2.1/KCNB1 channel works in the inner ear in mammals. The kcnb1 gene is expressed in the otic vesicle of developing zebrafish, suggesting its role in development of the inner ear. In the present study, we found that a Kcnb1 loss-of-function mutation affected development of the inner ear at multiple levels, including otic vesicle expansion, otolith formation, and the proliferation and differentiation of mechanosensory cells. This resulted in defects of kinocilia and stereocilia and abnormal function of the inner ear detected by behavioral assays. The quantitative transcriptional analysis of 75 genes demonstrated that the kcnb1 mutation affected the transcription of genes that are involved in K+ metabolism, cell proliferation, cilia development, and intracellular protein trafficking. These results demonstrate a role for Kv2.1/Kcnb1 channels in development of the inner ear in zebrafish.


Subject(s)
Cell Proliferation , Ear, Inner/embryology , Mechanotransduction, Cellular , Potassium Channels, Voltage-Gated/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cilia/genetics , Cilia/metabolism , Loss of Function Mutation , Potassium Channels, Voltage-Gated/genetics , Protein Transport/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
13.
Int J Dev Biol ; 65(4-5-6): 357-364, 2021.
Article in English | MEDLINE | ID: mdl-32930350

ABSTRACT

Cell differentiation, proliferation, and morphogenesis are generally driven by instructive signals that are sent and interpreted by adjacent tissues, a process known as induction. Cell recruitment is a particular case of induction in which differentiated cells produce a signal that drives adjacent cells to differentiate into the same type as the inducers. Once recruited, these new cells may become inducers to continue the recruitment process, closing a feed-forward loop that propagates the growth of a specific cell-type population. So far, little attention has been given to cell recruitment as a developmental mechanism. Here, we review the components of cell recruitment and discuss its contribution to development in three different examples: the Drosophila wing, the vertebrate inner ear, and the mammalian thyroid gland. Finally, we posit some open questions about the role of cell recruitment in organ patterning and growth.


Subject(s)
Drosophila , Mammals , Morphogenesis , Vertebrates , Animals , Drosophila/embryology , Ear, Inner/embryology , Gene Expression Regulation, Developmental , Mammals/embryology , Thyroid Gland/embryology , Vertebrates/embryology , Wings, Animal/embryology
14.
Dev Biol ; 469: 160-171, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33131705

ABSTRACT

The inner ear comprises four epithelial domains: the cochlea, vestibule, semicircular canals, and endolymphatic duct/sac. These structures are segregated at embryonic day 13.5 (E13.5). However, these four anatomical structures remain undefined at E10.5. Here, we aimed to identify lineage-specific genes in the early developing inner ear using published data obtained from single-cell RNA-sequencing (scRNA-seq) of embryonic mice. We downloaded 5000 single-cell transcriptome data, named 'auditory epithelial trajectory', from the Mouse Organogenesis Cell Atlas. The dataset was supposed to include otic epithelial cells at E9.5-13.5. We projected the 5000 â€‹cells onto a two-dimensional space encoding the transcriptional state and visualised the pattern of otic epithelial cell differentiation. We identified 15 clusters, which were annotated as one of the four components of the inner ear epithelium using known genes that characterise the four different tissues. Additionally, we classified 15 clusters into sub-regions of the four inner ear components. By comparing transcriptomes between these 15 clusters, we identified several candidates of lineage-specific genes. Characterising these new candidate genes will help future studies about inner ear development.


Subject(s)
Ear, Inner/embryology , Ear, Inner/metabolism , Animals , Cell Differentiation/genetics , Cochlea/metabolism , Computer Simulation , Ear, Inner/cytology , Embryo, Mammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , In Situ Hybridization , Mice , Mice, Inbred ICR , RNA, Messenger/metabolism , RNA-Seq , Single-Cell Analysis , Vestibule, Labyrinth/metabolism
15.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374462

ABSTRACT

This review provides an up-to-date source of information on the primary auditory neurons or spiral ganglion neurons in the cochlea. These neurons transmit auditory information in the form of electric signals from sensory hair cells to the first auditory nuclei of the brain stem, the cochlear nuclei. Congenital and acquired neurosensory hearing loss affects millions of people worldwide. An increasing body of evidence suggest that the primary auditory neurons degenerate due to noise exposure and aging more readily than sensory cells, and thus, auditory neurons are a primary target for regenerative therapy. A better understanding of the development and function of these neurons is the ultimate goal for long-term maintenance, regeneration, and stem cell replacement therapy. In this review, we provide an overview of the key molecular factors responsible for the function and neurogenesis of the primary auditory neurons, as well as a brief introduction to stem cell research focused on the replacement and generation of auditory neurons.


Subject(s)
Hair Cells, Auditory/physiology , Neurons/physiology , Animals , Base Sequence , Brain Stem , Cochlea/embryology , Cochlea/physiology , Cochlear Nucleus/embryology , Cochlear Nucleus/physiology , Ear, Inner/embryology , Ear, Inner/physiology , Evoked Potentials, Auditory, Brain Stem , Hearing Loss, Sensorineural/physiopathology , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mutation , Neurogenesis , Regenerative Medicine/methods , Spiral Ganglion/embryology , Spiral Ganglion/physiology
16.
Genetics ; 216(4): 1153-1185, 2020 12.
Article in English | MEDLINE | ID: mdl-33077489

ABSTRACT

Transcription factors that contain a homeodomain DNA-binding domain have crucial functions in most aspects of cellular function and embryonic development in both animals and plants. Hmx proteins are a subfamily of NK homeodomain-containing proteins that have fundamental roles in development of sensory structures such as the eye and the ear. However, Hmx functions in spinal cord development have not been analyzed. Here, we show that zebrafish (Danio rerio) hmx2 and hmx3a are coexpressed in spinal dI2 and V1 interneurons, whereas hmx3b, hmx1, and hmx4 are not expressed in spinal cord. Using mutational analyses, we demonstrate that, in addition to its previously reported role in ear development, hmx3a is required for correct specification of a subset of spinal interneuron neurotransmitter phenotypes, as well as correct lateral line progression and survival to adulthood. Surprisingly, despite similar expression patterns of hmx2 and hmx3a during embryonic development, zebrafish hmx2 mutants are viable and have no obviously abnormal phenotypes in sensory structures or neurons that require hmx3a In addition, embryos homozygous for deletions of both hmx2 and hmx3a have identical phenotypes to severe hmx3a single mutants. However, mutating hmx2 in hypomorphic hmx3a mutants that usually develop normally, results in abnormal ear and lateral line phenotypes. This suggests that while hmx2 cannot compensate for loss of hmx3a, it does function in these developmental processes, although to a much lesser extent than hmx3a More surprisingly, our mutational analyses suggest that Hmx3a may not require its homeodomain DNA-binding domain for its roles in viability or embryonic development.


Subject(s)
Ear, Inner/metabolism , Lateral Line System/metabolism , Spinal Cord/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Animals , Binding Sites , Ear, Inner/embryology , Interneurons/metabolism , Lateral Line System/embryology , Neurogenesis , Spinal Cord/embryology , Transcription Factors/chemistry , Transcription Factors/genetics , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
17.
Dev Dyn ; 249(7): 867-883, 2020 07.
Article in English | MEDLINE | ID: mdl-32384225

ABSTRACT

BACKGROUND: Sensorineural hearing loss is an understudied consequence of congenital Zika syndrome, and balance disorders are essentially unreported to date. Also lacking is information about the susceptibility and the pathogenesis of the developing inner ear following Zika virus (ZIKV) exposure. To address this, ZIKV was delivered directly into the otic cup/otocyst of chicken embryos and infection of inner ear tissues was evaluated using immunohistochemistry. RESULTS: After injections on embryonic days 2 to 5, ZIKV infection was observed in 90% of the samples harvested 2 to 8 days later; however, the degree of infection was highly variable across individuals. ZIKV was detected in all regions of the inner ear, associated ganglia, and in the surrounding periotic mesenchyme. Detection of virus peaked earlier in the ganglion and vestibular compartments, and later in the cochlea. ZIKV infection increased cell death robustly in the auditory ganglion, and modestly in the auditory sensory organ. Macrophage accumulation was found to overlap with dense viral infection in some tissues. Additionally, dysmorphogenesis of the semicircular canals and ganglion was observed for a subset of injection conditions. CONCLUSIONS: This article presents evidence of direct ZIKV infection of developing inner ear epithelium and shows previously unknown inner ear dysmorphogenesis phenotypes.


Subject(s)
Ear, Inner/embryology , Ear, Inner/virology , Hearing Loss, Sensorineural/embryology , Zika Virus Infection/virology , Zika Virus/metabolism , Animals , Cell Death , Chick Embryo , Chickens , Cochlea , Ear, Inner/metabolism , Epithelium/metabolism , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Macrophages/metabolism , Phenotype , Semicircular Canals/embryology , Semicircular Canals/metabolism , Time Factors , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
18.
Mol Brain ; 13(1): 29, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32127020

ABSTRACT

Tsukushi (TSK)-a small, secreted, leucine-rich-repeat proteoglycan-interacts with and regulates essential cellular signaling cascades. However, its functions in the mouse inner ear are unknown. In this study, measurement of auditory brainstem responses, fluorescence microscopy, and scanning electron microscopy revealed that TSK deficiency in mice resulted in the formation of abnormal stereocilia in the inner hair cells and hearing loss but not in the loss of these cells. TSK accumulated in nonprosensory regions during early embryonic stages and in both nonprosensory and prosensory regions in late embryonic stages. In adult mice, TSK was localized in the organ of Corti, spiral ganglion cells, and the stria vascularis. Moreover, loss of TSK caused dynamic changes in the expression of key genes that drive the differentiation of the inner hair cells in prosensory regions. Finally, our results revealed that TSK interacted with Sox2 and BMP4 to control stereocilia formation in the inner hair cells. Hence, TSK appears to be an essential component of the molecular pathways that regulate inner ear development.


Subject(s)
Ear, Inner/embryology , Ear, Inner/metabolism , Proteoglycans/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Hair Cells, Auditory, Inner/metabolism , Hearing , Ligaments/metabolism , Mice, Knockout , Proteoglycans/deficiency , Proteoglycans/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Spiral Ganglion/metabolism , Stereocilia/metabolism
19.
Biomolecules ; 10(3)2020 02 28.
Article in English | MEDLINE | ID: mdl-32121147

ABSTRACT

The vertebrate inner ear is responsible for detecting sound, gravity, and head motion. These mechanical forces are detected by mechanosensitive hair cells, arranged in a series of sensory patches in the vestibular and cochlear regions of the ear. Hair cells form synapses with neurons of the VIIIth cranial ganglion, which convey sound and balance information to the brain. They are surrounded by supporting cells, which nourish and protect the hair cells, and which can serve as a source of stem cells to regenerate hair cells after damage in non-mammalian vertebrates. The Notch signaling pathway plays many roles in the development of the inner ear, from the earliest formation of future inner ear ectoderm on the side of the embryonic head, to regulating the production of supporting cells, hair cells, and the neurons that innervate them. Notch signaling is re-deployed in non-mammalian vertebrates during hair cell regeneration, and attempts have been made to manipulate the Notch pathway to promote hair cell regeneration in mammals. In this review, we summarize the different modes of Notch signaling in inner ear development and regeneration, and describe how they interact with other signaling pathways to orchestrate the fine-grained cellular patterns of the ear.


Subject(s)
Ear, Inner/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Ear, Inner/embryology , Hair Cells, Auditory/physiology , Humans , Regeneration
20.
Dev Biol ; 462(1): 74-84, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32147304

ABSTRACT

The five vestibular organs of the inner ear derive from patches of prosensory cells that express the transcription factor SOX2 and the Notch ligand JAG1. Previous work suggests that JAG1-mediated Notch signaling is both necessary and sufficient for prosensory formation and that the separation of developing prosensory patches is regulated by LMX1a, which antagonizes Notch signaling. We used an inner ear-specific deletion of the Rbpjκ gene in which Notch signaling is progressively lost from the inner ear to show that Notch signaling, is continuously required for the maintenance of prosensory fate. Loss of Notch signaling in prosensory patches causes them to shrink and ultimately disappear. We show this loss of prosensory fate is not due to cell death, but rather to the conversion of prosensory tissue into non-sensory tissue that expresses LMX1a. Notch signaling is therefore likely to stabilize, rather than induce prosensory fate.


Subject(s)
Ear, Inner/embryology , Jagged-1 Protein/metabolism , Receptors, Notch/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation , Ear, Inner/metabolism , Gene Expression Regulation, Developmental/genetics , Hair Cells, Auditory, Inner/cytology , Jagged-1 Protein/genetics , LIM-Homeodomain Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred ICR , Organogenesis/physiology , Receptors, Notch/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL