Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 12573, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37537240

ABSTRACT

It has previously been shown that the zinc-finger transcription factor Gata3 has dynamic expression within the inner ear throughout embryonic development and is essential for cochlear neurosensory development. However, the temporal window for which Gata3 is required for proper formation of the cochlear neurosensory epithelia remains unclear. To investigate the role of Gata3 in cochlear neurosensory development in the late prosensory stages, we used the Sox2-creERT2 mouse line to target and conditionally delete Gata3 at E11.5, a timepoint before cells have fully committed to a neurosensory fate. While the inner ears of Sox2-creERT2: Gata3 f/f mice appear normal with no gross structural defects, the sensory cells in the organ of Corti are partially lost and disorganized in an increasing severity from base to apex. Additionally, spiral ganglion neurons display aberrant peripheral projections, including increased distances between radial bundles and disorganization upon reaching the organ of Corti. Furthermore, heterozygous Sox2-creERT2: Gata3 f/+ mice show a reduced aberrant phenotype in comparison to the homozygous mutant, supporting the hypothesis that Gata3 is not only required for proper formation at the later proneurosensory stage, but also that a specific expression level of Gata3 is required. Therefore, this study provides evidence that Gata3 plays a time-sensitive and dose-dependent role in the development of sensory and neuronal cells in late proneurosensory stages.


Subject(s)
Ear, Inner , GATA3 Transcription Factor , Animals , Mice , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Mice, Knockout , Ear, Inner/metabolism , Cochlea/metabolism , Epithelium/metabolism , Gene Expression Regulation, Developmental
2.
Res Sq ; 2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37090645

ABSTRACT

It has been previously shown that zinc-finger transcription factor Gata3 has dynamic expression within the inner ear throughout embryonic development and is essential for cochlear neurosensory development. However, the temporal window to which Gata3 is required for the formation of the cochlear neurosensory epithelia remains unclear. To investigate the role of Gata3 on cochlear neurosensory development in the late prosensory stages, we used the Sox2-cre ERT2 mouse line to target and conditionally delete Gata3 at E11.5 before the cells have fully committed to a neurosensory fate. While the inner ears of Sox2-cre ERT2 : Gata3 f/f mice appear morphologically normal, the sensory cells in the organ of Corti are partially lost and disorganized in a basal to apical gradient with the apex demonstrating the more severe phenotype. Additionally, spiral ganglion neurons display aberrant peripheral projections, such as increased distances between radial bundles and disorganization upon reaching the organ of Corti. Furthermore, heterozygous Sox2-cre ERT2 : Gata3 f/+ mice show a reduced phenotype in comparison to the homozygous mutant, supporting the concept that Gata3 is not only required for proper formation at the later proneurosensory stage, but also that a specific level of Gata3 is required. Therefore, our studies confirm that Gata3 plays a time-sensitive and dose-dependent role in the development of sensory cells in the late proneurosensory stages.

3.
Front Neurosci ; 15: 779871, 2021.
Article in English | MEDLINE | ID: mdl-35153658

ABSTRACT

During development the afferent neurons of the inner ear make precise wiring decisions in the hindbrain reflective of their topographic distribution in the periphery. This is critical for the formation of sensory maps capable of faithfully processing both auditory and vestibular input. Disorganized central projections of inner ear afferents in Fzd3 null mice indicate Wnt/PCP signaling is involved in this process and ear transplantation in Xenopus indicates that Fzd3 is necessary in the ear but not the hindbrain for proper afferent navigation. However, it remains unclear in which cell type of the inner ear Fzd3 expression is influencing the guidance of inner ear afferents to their proper synaptic targets in the hindbrain. We utilized Atoh1-cre and Neurod1-cre mouse lines to conditionally knockout Fzd3 within the mechanosensory hair cells of the organ of Corti and within the inner ear afferents, respectively. Following conditional deletion of Fzd3 within the hair cells, the central topographic distribution of inner ear afferents was maintained with no gross morphological defects. In contrast, conditional deletion of Fzd3 within inner ear afferents leads to central pathfinding defects of both cochlear and vestibular afferents. Here, we show that Fzd3 is acting in a cell autonomous manner within inner ear afferents to regulate central pathfinding within the hindbrain.

4.
Sci Rep ; 9(1): 10298, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31311957

ABSTRACT

Inner ear sensory afferent connections establish sensory maps between the inner ear hair cells and the vestibular and auditory nuclei to allow vestibular and sound information processing. While molecular guidance of sensory afferents to the periphery has been well studied, molecular guidance of central projections from the ear is only beginning to emerge. Disorganized central projections of spiral ganglion neurons in a Wnt/PCP pathway mutant, Prickle1, suggest the Wnt/PCP pathway plays a role in guiding cochlear afferents to the cochlear nuclei in the hindbrain, consistent with known expression of the Wnt receptor, Frizzled3 (Fzd3) in inner ear neurons. We therefore investigated the role of Wnt signaling in central pathfinding in Fzd3 mutant mice and Fzd3 morpholino treated frogs and found aberrant central projections of vestibular afferents in both cases. Ear transplantations from knockdown to control Xenopus showed that it is the Fzd3 expressed within the ear that mediates this guidance. Also, cochlear afferents of Fzd3 mutant mice lack the orderly topological organization observed in controls. Quantification of Fzd3 expression in spiral ganglion neurons show a gradient of expression with Fzd3 being higher in the apex than in the base. Together, these results suggest that a gradient of Fzd3 in inner ear afferents directs projections to the correct dorsoventral column within the hindbrain.


Subject(s)
Ear, Inner/metabolism , Frizzled Receptors/genetics , Rhombencephalon/metabolism , Xenopus Proteins/genetics , Animals , Frizzled Receptors/metabolism , Gene Knockdown Techniques , Mice , Mutation , Spiral Ganglion/metabolism , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Xenopus laevis
5.
Front Cell Dev Biol ; 7: 59, 2019.
Article in English | MEDLINE | ID: mdl-31069224

ABSTRACT

Central nervous system neurons become postmitotic when radial glia cells divide to form neuroblasts. Neuroblasts may migrate away from the ventricle radially along glia fibers, in various directions or even across the midline. We present four cases of unusual migration that are variably connected to either pathology or formation of new populations of neurons with new connectivities. One of the best-known cases of radial migration involves granule cells that migrate from the external granule cell layer along radial Bergman glia fibers to become mature internal granule cells. In various medulloblastoma cases this migration does not occur and transforms the external granule cell layer into a rapidly growing tumor. Among the ocular motor neurons is one unique population that undergoes a contralateral migration and uniquely innervates the superior rectus and levator palpebrae muscles. In humans, a mutation of a single gene ubiquitously expressed in all cells, induces innervation defects only in this unique motor neuron population, leading to inability to elevate eyes or upper eyelids. One of the best-known cases for longitudinal migration is the facial branchial motor (FBM) neurons and the overlapping inner ear efferent population. We describe here molecular cues that are needed for the caudal migration of FBM to segregate these motor neurons from the differently migrating inner ear efferent population. Finally, we describe unusual migration of inner ear spiral ganglion neurons that result in aberrant connections with disruption of frequency presentation. Combined, these data identify unique migratory properties of various neuronal populations that allow them to adopt new connections but also sets them up for unique pathologies.

6.
Front Cell Neurosci ; 12: 252, 2018.
Article in English | MEDLINE | ID: mdl-30135646

ABSTRACT

We review the evolution and development of organ of Corti hair cells with a focus on their molecular differences from vestibular hair cells. Such information is needed to therapeutically guide organ of Corti hair cell development in flat epithelia and generate the correct arrangement of different hair cell types, orientation of stereocilia, and the delayed loss of the kinocilium that are all essential for hearing, while avoiding driving hair cells toward a vestibular fate. Highlighting the differences from vestibular organs and defining what is known about the regulation of these differences will help focus future research directions toward successful restoration of an organ of Corti following long-term hair cell loss.

7.
Dev Biol ; 423(2): 126-137, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28159525

ABSTRACT

Vestibular hair cells of the inner ear are specialized receptors that detect mechanical stimuli from gravity and motion via the deflection of a polarized bundle of stereocilia located on their apical cell surfaces. The orientation of stereociliary bundles is coordinated between neighboring cells by core PCP proteins including the large adhesive G-protein coupled receptor Celsr1. We show that mice lacking Celsr1 have vestibular behavioral phenotypes including circling. In addition, we show that Celsr1 is asymmetrically distributed at cell boundaries between hair cells and neighboring supporting cells in the developing vestibular and auditory sensory epithelia. In the absence of Celsr1 the stereociliary bundles of vestibular hair cells are misoriented relative to their neighbors, a phenotype that is greatest in the cristae of the semicircular canals. Since horizontal semi-circular canal defects lead to circling in other mutant mouse lines, we propose that this PCP phenotype is the cellular basis of the circling behavior in Celsr1 mutants.


Subject(s)
Cell Polarity , Ear, Inner/cytology , Ear, Inner/embryology , Hair Cells, Vestibular/cytology , Receptors, G-Protein-Coupled/metabolism , Animals , Behavior, Animal , Ear, Inner/metabolism , Epithelium/metabolism , Gene Deletion , Mice, Knockout , Organ of Corti/cytology , Organ of Corti/embryology , Organ of Corti/metabolism , Phenotype , Signal Transduction , Stereocilia/metabolism
8.
Methods Mol Biol ; 1427: 243-62, 2016.
Article in English | MEDLINE | ID: mdl-27259931

ABSTRACT

The inner ear has long been at the cutting edge of tract tracing techniques that have shaped and reshaped our understanding of the ear's innervation patterns. This review provides a historical framework to understand the importance of these techniques for ear innervation and for development of tracing techniques in general; it is hoped that lessons learned will help to quickly adopt transformative novel techniques and their information and correct past beliefs based on technical limitations. The technical part of the review presents details of our protocol as developed over the last 30 years. We also include arguments as to why these recommendations work best to generate the desired outcome of distinct fiber and cell labeling, and generate reliable data for any investigation. We specifically focus on two tracing techniques, in part developed and/or championed for ear innervation analysis: the low molecular multicolor dextran amine tract tracing technique and the multicolor tract tracing technique with lipophilic dyes.


Subject(s)
Ear/innervation , Neuroanatomical Tract-Tracing Techniques/methods , Animals , Dextrans/chemistry , Ear/anatomy & histology , Fluorescent Dyes/chemistry , Mice
9.
Elife ; 52016 05 25.
Article in English | MEDLINE | ID: mdl-27223325

ABSTRACT

The neuron specific RNA-binding proteins NOVA1 and NOVA2 are highly homologous alternative splicing regulators. NOVA proteins regulate at least 700 alternative splicing events in vivo, yet relatively little is known about the biologic consequences of NOVA action and in particular about functional differences between NOVA1 and NOVA2. Transcriptome-wide searches for isoform-specific functions, using NOVA1 and NOVA2 specific HITS-CLIP and RNA-seq data from mouse cortex lacking either NOVA isoform, reveals that NOVA2 uniquely regulates alternative splicing events of a series of axon guidance related genes during cortical development. Corresponding axonal pathfinding defects were specific to NOVA2 deficiency: Nova2-/- but not Nova1-/- mice had agenesis of the corpus callosum, and axonal outgrowth defects specific to ventral motoneuron axons and efferent innervation of the cochlea. Thus we have discovered that NOVA2 uniquely regulates alternative splicing of a coordinate set of transcripts encoding key components in cortical, brainstem and spinal axon guidance/outgrowth pathways during neural differentiation, with severe functional consequences in vivo.


Subject(s)
Antigens, Neoplasm/metabolism , Axon Guidance , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental , Neurons/physiology , RNA-Binding Proteins/metabolism , RNA/metabolism , Animals , Mice , Mice, Knockout , Neuro-Oncological Ventral Antigen
10.
Neuron ; 82(2): 334-49, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24656932

ABSTRACT

The ocular motility disorder "Congenital fibrosis of the extraocular muscles type 1" (CFEOM1) results from heterozygous mutations altering the motor and third coiled-coil stalk of the anterograde kinesin, KIF21A. We demonstrate that Kif21a knockin mice harboring the most common human mutation develop CFEOM. The developing axons of the oculomotor nerve's superior division stall in the proximal nerve; the growth cones enlarge, extend excessive filopodia, and assume random trajectories. Inferior division axons reach the orbit but branch ectopically. We establish a gain-of-function mechanism and find that human motor or stalk mutations attenuate Kif21a autoinhibition, providing in vivo evidence for mammalian kinesin autoregulation. We identify Map1b as a Kif21a-interacting protein and report that Map1b⁻/⁻ mice develop CFEOM. The interaction between Kif21a and Map1b is likely to play a critical role in the pathogenesis of CFEOM1 and highlights a selective vulnerability of the developing oculomotor nerve to perturbations of the axon cytoskeleton.


Subject(s)
Axons/pathology , Eye Diseases, Hereditary/genetics , Fibrosis/genetics , Kinesins/genetics , Kinesins/metabolism , Mutation/genetics , Ocular Motility Disorders/genetics , Oculomotor Nerve/pathology , Age Factors , Animals , Animals, Newborn , Axons/ultrastructure , Cell Count , Disease Models, Animal , Embryo, Mammalian , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/physiopathology , Eye Movements/genetics , Eye Movements/physiology , Fibrosis/pathology , Fibrosis/physiopathology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/ultrastructure , Ocular Motility Disorders/pathology , Ocular Motility Disorders/physiopathology , Oculomotor Nerve/ultrastructure
11.
J Neurosci ; 33(35): 14001-16, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986237

ABSTRACT

The distinctive planar polarity of auditory hair cells is evident in the polarized organization of the stereociliary bundle. Mutations in the core planar cell polarity gene Van Gogh-like 2 (Vangl2) result in hair cells that fail to properly orient their stereociliary bundles along the mediolateral axis of the cochlea. The severity of this phenotype is graded along the length of the cochlea, similar to the hair cell differentiation gradient, suggesting that an active refinement process corrects planar polarity phenotypes in Vangl2 knock-out (KO) mice. Because Vangl2 gene deletions are lethal, Vangl2 conditional knock-outs (CKOs) were generated to test this hypothesis. When crossed with Pax2-Cre, Vangl2 is deleted from the inner ear, yielding planar polarity phenotypes similar to Vangl2 KOs at late embryonic stages except that Vangl2 CKO mice are viable and do not have craniorachischisis like Vangl2 KOs. Quantification of planar polarity deficits through postnatal development demonstrates the activity of a Vangl2-independent refinement process that rescues the planar polarity phenotype within 10 d of birth. In contrast, the Pax2-Cre;Vangl2 CKO has profound changes in the shape and distribution of outer pillar cell and Deiters' cell phalangeal processes that are not corrected during the period of planar polarity refinement. Auditory brainstem response analyses of adult mice show a 10-15 dB shift in auditory threshold, and distortion product otoacoustic emission measurements indicate that this mild hearing deficit is of cochlear origin. Together, these data demonstrate a Vangl2-independent refinement mechanism that actively reorients auditory stereociliary bundles and reveals an unexpected role of Vangl2 during supporting cell morphogenesis.


Subject(s)
Hair Cells, Auditory/cytology , Nerve Tissue Proteins/genetics , Animals , Auditory Threshold , Brain Stem/physiology , Cell Differentiation , Cochlea/cytology , Cochlea/embryology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/physiology , Hearing Loss/genetics , Mice , Mice, Knockout , Phenotype , Stereocilia/ultrastructure
12.
PLoS One ; 8(4): e62046, 2013.
Article in English | MEDLINE | ID: mdl-23614009

ABSTRACT

Hair cells of the developing mammalian inner ear are progressively defined through cell fate restriction. This process culminates in the expression of the bHLH transcription factor Atoh1, which is necessary for differentiation of hair cells, but not for their specification. Loss of several genes will disrupt ear morphogenesis or arrest of neurosensory epithelia development. We previously showed in null mutants that the loss of the transcription factor, Gata3, results specifically in the loss of all cochlear neurosensory development. Temporal expression of Gata3 is broad from the otic placode stage through the postnatal ear. It therefore remains unclear at which stage in development Gata3 exerts its effect. To better understand the stage specific effects of Gata3, we investigated the role of Gata3 in cochlear neurosensory specification and differentiation utilizing a LoxP targeted Gata3 line and two Cre lines. Foxg1(Cre)∶Gata3(f/f) mice show recombination of Gata3 around E8.5 but continue to develop a cochlear duct without differentiated hair cells and spiral ganglion neurons. qRT-PCR data show that Atoh1 was down-regulated but not absent in the duct whereas other hair cell specific genes such as Pou4f3 were completely absent. In addition, while Sox2 levels were lower in the Foxg1(Cre):Gata3(f/f) cochlea, Eya1 levels remained normal. We conclude that Eya1 is unable to fully upregulate Atoh1 or Pou4f3, and drive differentiation of hair cells without Gata3. Pax2-Cre∶Gata3(f/f) mice show a delayed recombination of Gata3 in the ear relative to Foxg1(Cre):Gata3(f/f) . These mice exhibited a cochlear duct containing patches of partially differentiated hair cells and developed only few and incorrectly projecting spiral ganglion neurons. Our conditional deletion studies reveal a major role of Gata3 in the signaling of prosensory genes and in the differentiation of cochlear neurosenory cells. We suggest that Gata3 may act in combination with Eya1, Six1, and Sox2 in cochlear prosensory gene signaling.


Subject(s)
Cochlear Duct/cytology , GATA3 Transcription Factor/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Animals , GATA3 Transcription Factor/genetics , Mice
13.
Evol Dev ; 15(1): 63-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23331918

ABSTRACT

The tetrapod auditory system transmits sound through the outer and middle ear to the organ of Corti or other sound pressure receivers of the inner ear where specialized hair cells translate vibrations of the basilar membrane into electrical potential changes that are conducted by the spiral ganglion neurons to the auditory nuclei. In other systems, notably the vertebrate limb, a detailed connection between the evolutionary variations in adaptive morphology and the underlying alterations in the genetic basis of development has been partially elucidated. In this review, we attempt to correlate evolutionary and partially characterized molecular data into a cohesive perspective of the evolution of the mammalian organ of Corti out of the tetrapod basilar papilla. We propose a stepwise, molecularly partially characterized transformation of the ancestral, vestibular developmental program of the vertebrate ear. This review provides a framework to decipher both discrete steps in development and the evolution of unique functional adaptations of the auditory system. The combined analysis of evolution and development establishes a powerful cross-correlation where conclusions derived from either approach become more meaningful in a larger context which is not possible through exclusively evolution or development centered perspectives. Selection may explain the survival of the fittest auditory system, but only developmental genetics can explain the arrival of the fittest auditory system. [Modified after (Wagner 2011)].


Subject(s)
Biological Evolution , Organ of Corti/physiology , Vertebrates/physiology , Animals , Cochlea/physiology , DNA Mutational Analysis , Developmental Biology , Ear/physiology , Evolution, Molecular , Hair Cells, Auditory, Inner/physiology , Hearing , Mice , Phylogeny , Spiral Ganglion/physiology
14.
Anat Rec (Hoboken) ; 295(11): 1760-74, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23044863

ABSTRACT

Here, we review the molecular basis of mechanosensory cell and mechanosensory organ development and evolution with an emphasis on the conservation of transcription factors and emerging data on conserved gene networks. The ear, the organ of vertebrates dedicated to the perception of sound and balance, perceives these stimuli with the use of mechanosensory cells. The developmental gene regulatory network used during mechanosensory cellular development has been conserved from ancient bilaterian cells, and modified for the extraction of specific mechanical stimuli resulting in phenotypic changes. In the vertebrate lineage, mechanosensory cells became specialized as gravistatic sensors after they became aggregated to form the ear. After this aggregation, growth, including duplication and segregation of existing neurosensory epithelia, gave rise to new epithelia and can be appreciated by comparing sensory epithelia from the inner ears of different vertebrates and their innervation by different neuronal populations. Novel directions of differentiation were apparently further expanded by incorporating unique molecular modules in newly developed sensory epithelia. For example, the saccule gave rise to the auditory epithelium and corresponding neuronal population of tetrapods, starting possibly in an aquatic environment. This novel sensory perception was followed by emergence of the central auditory nuclei and a selective cochlear nucleus projection. The data for this process is outlined and contrasted with other ideas dealing with a subset of the data.


Subject(s)
Biological Evolution , Ear/physiology , Hair Cells, Auditory/physiology , Organ of Corti/physiology , Postural Balance/physiology , Sensory Receptor Cells/physiology , Sound , Animals , Humans
15.
Adv Exp Med Biol ; 739: 173-86, 2012.
Article in English | MEDLINE | ID: mdl-22399402

ABSTRACT

We review the molecular basis of auditory development and evolution. We propose that the auditory periphery (basilar papilla, organ of Corti) evolved by transforming a newly created and redundant vestibular (gravistatic) endorgan into a sensory epithelium that could respond to sound instead of gravity. Evolution altered this new epithelia's mechanoreceptive properties through changes of hair cells, positioned the epithelium in a unique position near perilymphatic space to extract sound moving between the round and the oval window, and transformed its otolith covering into a tympanic membrane. Another important step in the evolution of an auditory system was the evolution of a unique set of "auditory neurons" that apparently evolved from vestibular neurons. Evolution of mammalian auditory (spiral ganglion) neurons coincides with GATA3 being a transcription factor found selectively in the auditory afferents. For the auditory information to be processed, the CNS required a dedicated center for auditory processing, the auditory nuclei. It is not known whether the auditory nucleus is ontogenetically related to the vestibular or electroreceptive nuclei, two sensory systems found in aquatic but not in amniotic vertebrates, or a de-novo formation of the rhombic lip in line with other novel hindbrain structures such as pontine nuclei. Like other novel hindbrain structures, the auditory nuclei express exclusively the bHLH gene Atoh1, and loss of Atoh1 results in loss of most of this nucleus in mice. Only after the basilar papilla, organ of Corti evolved could efferent neurons begin to modulate their activity. These auditory efferents most likely evolved from vestibular efferent neurons already present. The most simplistic interpretation of available data suggest that the ear, sensory neurons, auditory nucleus, and efferent neurons have been transformed by altering the developmental genetic modules necessary for their development into a novel direction conducive for sound extraction, conduction, and processing.


Subject(s)
Evolution, Molecular , Vestibule, Labyrinth/growth & development , Animals , Epithelium/metabolism , Humans , Neurons, Efferent/cytology , Sensory Receptor Cells/cytology , Vestibule, Labyrinth/cytology , Vestibule, Labyrinth/physiology
16.
PLoS One ; 7(1): e30853, 2012.
Article in English | MEDLINE | ID: mdl-22292060

ABSTRACT

In the mammalian inner ear neurosensory cell fate depends on three closely related transcription factors, Atoh1 for hair cells and Neurog1 and Neurod1 for neurons. We have previously shown that neuronal cell fate can be altered towards hair cell fate by eliminating Neurod1 mediated repression of Atoh1 expression in neurons. To test whether a similar plasticity is present in hair cell fate commitment, we have generated a knockin (KI) mouse line (Atoh1(KINeurog1)) in which Atoh1 is replaced by Neurog1. Expression of Neurog1 under Atoh1 promoter control alters the cellular gene expression pattern, differentiation and survival of hair cell precursors in both heterozygous (Atoh1(+/KINeurog1)) and homozygous (Atoh1(KINeurog1/KINeurog1)) KI mice. Homozygous KI mice develop patches of organ of Corti precursor cells that express Neurog1, Neurod1, several prosensory genes and neurotrophins. In addition, these patches of cells receive afferent and efferent processes. Some cells among these patches form multiple microvilli but no stereocilia. Importantly, Neurog1 expressing mutants differ from Atoh1 null mutants, as they have intermittent formation of organ of Corti-like patches, opposed to a complete 'flat epithelium' in the absence of Atoh1. In heterozygous KI mice co-expression of Atoh1 and Neurog1 results in change in fate and patterning of some hair cells and supporting cells in addition to the abnormal hair cell polarity in the later stages of development. This differs from haploinsufficiency of Atoh1 (Pax2cre; Atoh1(f/+)), indicating the effect of Neurog1 expression in developing hair cells. Our data suggest that Atoh1(KINeurog1) can provide some degree of functional support for survival of organ of Corti cells. In contrast to the previously demonstrated fate plasticity of neurons to differentiate as hair cells, hair cell precursors can be maintained for a limited time by Neurog1 but do not transdifferentiate as neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Organ of Corti/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Survival/genetics , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genes, Lethal/genetics , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Organ of Corti/cytology , Organ of Corti/embryology , Organ of Corti/metabolism , Organogenesis/genetics , Pregnancy
17.
PLoS One ; 7(1): e30358, 2012.
Article in English | MEDLINE | ID: mdl-22279587

ABSTRACT

Atonal homolog1 (Atoh1) is a bHLH transcription factor essential for inner ear hair cell differentiation. Targeted expression of Atoh1 at various stages in development can result in hair cell differentiation in the ear. However, the level and duration of Atoh1 expression required for proper hair cell differentiation and maintenance remain unknown. We generated an Atoh1 conditional knockout (CKO) mouse line using Tg(Atoh1-cre), in which the cre expression is driven by an Atoh1 enhancer element that is regulated by Atoh1 protein to "self-terminate" its expression. The mutant mice show transient, limited expression of Atoh1 in all hair cells in the ear. In the organ of Corti, reduction and delayed deletion of Atoh1 result in progressive loss of almost all the inner hair cells and the majority of the outer hair cells within three weeks after birth. The remaining cells express hair cell marker Myo7a and attract nerve fibers, but do not differentiate normal stereocilia bundles. Some Myo7a-positive cells persist in the cochlea into adult stages in the position of outer hair cells, flanked by a single row of pillar cells and two to three rows of disorganized Deiters cells. Gene expression analyses of Atoh1, Barhl1 and Pou4f3, genes required for survival and maturation of hair cells, reveal earlier and higher expression levels in the inner compared to the outer hair cells. Our data show that Atoh1 is crucial for hair cell mechanotransduction development, viability, and maintenance and also suggest that Atoh1 expression level and duration may play a role in inner vs. outer hair cell development. These genetically engineered Atoh1 CKO mice provide a novel model for establishing critical conditions needed to regenerate viable and functional hair cells with Atoh1 therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Hair Cells, Auditory/metabolism , Organ of Corti/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/genetics , Cochlea/embryology , Cochlea/growth & development , Cochlea/metabolism , Female , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Hair Cells, Auditory/ultrastructure , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mechanotransduction, Cellular/genetics , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Myosin VIIa , Myosins/genetics , Myosins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ of Corti/embryology , Organ of Corti/growth & development , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stereocilia/genetics , Stereocilia/metabolism , Transcription Factor Brn-3C/genetics , Transcription Factor Brn-3C/metabolism
18.
Int J Dev Biol ; 55(3): 297-303, 2011.
Article in English | MEDLINE | ID: mdl-21553382

ABSTRACT

Haploinsufficiency of Gata3 causes hypoparathyroidism, deafness and renal dysplasia (HDR) syndrome in mice and humans. Gata3 null mutation leads to early lethality around embryonic day (E)11.5, but catecholamine precursor administration can rescue Gata3 null mutants to E16.5. At E11.5, GATA3 deficiency results in the development of an empty otocyst with an endolymphatic duct. However, using rescued mice we found that some morphogenesis and neurosensory development is possible in the ear without Gata3. Extending previous studies, we find that at E16.5, Gata3 mutant inner ears can undergo partial morphogenesis and develop an endolymphatic duct, a utricular and saccular recess, and a shortened cochlear duct. In addition to the obvious morphogenic aberrations, these studies demonstrate that a subset of neurons develop and connect a fragmented sensory patch of MYO7A-positive hair cells to the vestibular nuclei of the brainstem. In situ hybridization studies reveal altered expression of several transcription factors relevant to ear development and we hypothesize that this may relate to the observed dysmorphia and restricted neurosensory development. While a cochlear duct can form, there is no concurrent cochlear neurosensory development, observations consistent with specific hearing defects encountered by HDR patients and mice with Gata3-associated expression alterations. Gata3 null mutant phenocopies the otic maldevelopment (cochlear duct formation in the absence of neurosensory development) seen in Foxg1cre mediated conditional deletion of microRNA processing enzyme, Dicer1. Finally, while GATA3 is expressed in the developing vestibulo-cochlear efferent (VCE) neurons, and its absence in the null mutants disrupts VCE projections to the ear, loss of GATA3 does not affect VCE progenitor cell migration.


Subject(s)
Ear, Inner/embryology , Ear, Inner/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Neurosecretory Systems/embryology , Neurosecretory Systems/metabolism , Animals , Catecholamines/administration & dosage , Catecholamines/pharmacology , Cell Movement , Cochlear Duct/abnormalities , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GATA3 Transcription Factor/deficiency , Hair Cells, Vestibular/cytology , Hair Cells, Vestibular/physiology , Mice , Mice, Knockout , Mutation , Myosin VIIa , Myosins/metabolism , Vestibulocochlear Nerve/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...