Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Dev Dyn ; 252(1): 124-144, 2023 01.
Article in English | MEDLINE | ID: mdl-36284453

ABSTRACT

BACKGROUND: Proper connectivity between type I spiral ganglion neurons (SGNs) and inner hair cells (IHCs) in the cochlea is necessary for conveying sound information to the brain in mammals. Previous studies have shown that type I SGNs are heterogeneous in form, function and synaptic location on IHCs, but factors controlling their patterns of connectivity are not well understood. RESULTS: During development, cochlear supporting cells and SGNs express Semaphorin-3A (SEMA3A), a known axon guidance factor. Mice homozygous for a point mutation that attenuates normal SEMA3A repulsive activity (Sema3aK108N ) show cochleae with grossly normal patterns of IHC innervation. However, genetic sparse labeling and three-dimensional reconstructions of individual SGNs show that cochleae from Sema3aK108N mice lacked the normal synaptic distribution of type I SGNs. Additionally, Sema3aK108N cochleae show a disrupted distribution of GLUA2 postsynaptic patches around the IHCs. The addition of SEMA3A-Fc to postnatal cochleae led to increases in SGN branching, similar to the effects of inhibiting glutamate receptors. Ca2+ imaging studies show that SEMA3A-Fc decreases SGN activity. CONCLUSIONS: Contrary to the canonical view of SEMA3A as a guidance ligand, our results suggest SEMA3A may regulate SGN excitability in the cochlea, which may influence the morphology and synaptic arrangement of type I SGNs.


Subject(s)
Hair Cells, Auditory , Semaphorin-3A , Animals , Mice , Cochlea/metabolism , Neurons/metabolism , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Spiral Ganglion/metabolism
2.
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
3.
J Neurosci ; 39(33): 6425-6438, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31209173

ABSTRACT

During nervous system development, axons often undergo elaborate changes in branching patterns before circuits have achieved their mature patterns of innervation. In the auditory system, type I spiral ganglion neurons (SGNs) project their peripheral axons into the cochlear epithelium and then undergo a process of branch refinement before forming synapses with sensory hair cells. Here, we report that Semaphorin-5B (Sema5B) acts as an important mediator of this process. During cochlear development in mouse, immature hair cells express Sema5B, whereas the SGNs express both PlexinA1 and PlexinA3, which are known Sema5B receptors. In these studies, genetic sparse labeling and three-dimensional reconstruction techniques were leveraged to determine the morphologies of individual type I SGNs after manipulations of Sema5B signaling. Treating cultured mouse cochleae with Sema5B-Fc (to activate Plexin-As) led to type I SGNs with less numerous, but longer terminal branches. Conversely, cochleae from Sema5b knock-out mice showed type I SGNs with more numerous, but shorter terminal branches. In addition, conditional loss of Plxna1 in SGNs (using Bhlhb5Cre) led to increased type I SGN branching, suggesting that PlexinA1 normally responds to Sema5B in this process. In these studies, mice of either sex were used. The data presented here suggest that Sema5B-PlexinA1 signaling limits SGN terminal branch numbers without causing axonal repulsion, which is a role that distinguishes Sema5B from other Semaphorins in cochlear development.SIGNIFICANCE STATEMENT The sensorineural components of the cochlea include hair cells, which respond mechanically to sound waves, and afferent spiral ganglion neurons (SGNs), which respond to glutamate released by hair cells and transmit auditory information into the CNS. An important component of synapse formation in the cochlea is a process of SGN "debranching" whereby SGNs lose extraneous branches before developing unramified bouton endings that contact the hair cells. In this work, we have found that the transmembrane ligand Semaphorin-5B and its receptor PlexinA1 regulate the debranching process. The results in this report provide new knowledge regarding the molecular control of cochlear afferent innervation.


Subject(s)
Neurogenesis/physiology , Neurons/metabolism , Semaphorins/metabolism , Spiral Ganglion/embryology , Animals , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Spiral Ganglion/metabolism
4.
Semin Cell Dev Biol ; 65: 80-87, 2017 05.
Article in English | MEDLINE | ID: mdl-27760385

ABSTRACT

In hearing, mechanically sensitive hair cells (HCs) in the cochlea release glutamate onto spiral ganglion neurons (SGNs) to relay auditory information to the central nervous system (CNS). There are two main SGN subtypes, which differ in morphology, number, synaptic targets, innervation patterns and firing properties. About 90-95% of SGNs are the type I SGNs, which make a single bouton connection with inner hair cells (IHCs) and have been well described in the canonical auditory pathway for sound detection. However, less attention has been given to the type II SGNs, which exclusively innervate outer hair cells (OHCs). In this review, we emphasize recent advances in the molecular mechanisms that control how type II SGNs develop and form connections with OHCs, and exciting new insights into the function of type II SGNs.


Subject(s)
Hair Cells, Auditory/metabolism , Hearing/physiology , Mechanotransduction, Cellular , Sensory Receptor Cells/metabolism , Spiral Ganglion/metabolism , Animals , Cell Differentiation , Cell Lineage/physiology , Ephrin-A5/genetics , Ephrin-A5/metabolism , Gene Expression Regulation, Developmental , Glutamic Acid/metabolism , Hair Cells, Auditory/cytology , Humans , Mammals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sensory Receptor Cells/cytology , Spiral Ganglion/cytology
5.
Synapse ; 73(5): e22087, 2019 05.
Article in English | MEDLINE | ID: mdl-30592086

ABSTRACT

In mammals, hair cells and spiral ganglion neurons (SGNs) in the cochlea together are sophisticated "sensorineural" structures that transduce auditory information from the outside world into the brain. Hair cells and SGNs are joined by glutamatergic ribbon-type synapses composed of a molecular machinery rivaling in complexity the mechanoelectric transduction components found at the apical side of the hair cell. The cochlear hair cell ribbon synapse has received much attention lately because of recent and important findings related to its damage (sometimes termed "synaptopathy") as a result of noise overexposure. During development, ribbon synapses between type I SGNs and inner hair cells form in the time window between birth and hearing onset and is a process coordinated with type I SGN myelination, spontaneous activity, synaptic pruning, and innervation by efferents. In this review, we highlight new findings regarding the diversity of type I SGNs and inner hair cell synapses, and the molecular mechanisms of selective hair cell targeting. Also discussed are cell adhesion molecules and protein constituents of the ribbon synapse, and how these factors participate in ribbon synapse formation. We also note interesting new insights into the morphological development of type II SGNs, and the potential for cochlear macrophages as important players in protecting SGNs. We also address recent studies demonstrating that the structural and physiological profiles of the type I SGNs do not reach full maturity until weeks after hearing onset, suggesting a protracted development that is likely modulated by activity.


Subject(s)
Neurogenesis , Spiral Ganglion/growth & development , Synapses/physiology , Animals , Hair Cells, Auditory/cytology , Hair Cells, Auditory/physiology , Humans , Spiral Ganglion/cytology , Spiral Ganglion/physiology , Synapses/ultrastructure
6.
BMC Biol ; 16(1): 105, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30253757

ABSTRACT

During hearing in mammals, "sensorineural" inner hair cells convert sound wave-generated mechanical input into electrical activity, resulting in glutamate release onto type I spiral ganglion neurons (SGNs) at specialized synapses known as "ribbon synapses". New findings published here in BMC Biology by Sonntag and colleagues indicate a role for the proteoglycan Brevican in forming perineurounal net (PNN) baskets at these synapses and controlling the spatial distribution of presynaptic voltage-gated calcium channels that regulate glutamate release. These findings may provide insight into the mechanism by which individual ribbon synapses within a single hair cell can function in an independent manner to facilitate hearing within a broad dynamic range.


Subject(s)
Brevican , Calcium , Animals , Extracellular Matrix , Hair , Synapses
7.
Dev Dyn ; 247(7): 934-950, 2018 07.
Article in English | MEDLINE | ID: mdl-29536590

ABSTRACT

BACKGROUND: In the cochlea, auditory development depends on precise patterns of innervation by afferent and efferent nerve fibers, as well as a stereotyped arrangement of hair and supporting cells. Neuronal cell adhesion molecule (NrCAM) is a homophilic cell adhesion molecule that controls diverse aspects of nervous system development, but the function of NrCAM in cochlear development is not well understood. RESULTS: Throughout cochlear innervation, NrCAM is detectable on spiral ganglion neuron (SGN) afferent and olivocochlear efferent fibers, and on the membranes of developing hair and supporting cells. Neonatal Nrcam-null cochleae show errors in type II SGN fasciculation, reduced efferent innervation, and defects in the stereotyped packing of hair and supporting cells. Nrcam loss also leads to dramatic changes in the profiles of presynaptic afferent and efferent synaptic markers at the time of hearing onset. Despite these numerous developmental defects, Nrcam-null adults do not show defects in auditory acuity, and by postnatal day 21, the developmental deficits in ribbon synapse distribution and sensory domain structure appear to have been corrected. CONCLUSIONS: NrCAM is expressed by several neural and sensory epithelial subtypes within the developing cochlea, and the loss of Nrcam confers numerous, but nonpermanent, developmental defects in innervation and sensory domain patterning. Developmental Dynamics 247:934-950, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Body Patterning/physiology , Cell Adhesion Molecules, Neuronal/physiology , Cell Adhesion Molecules/metabolism , Cochlea/innervation , Sensory Receptor Cells/chemistry , Animals , Axon Guidance , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/physiology , Cochlea/cytology , Cochlea/growth & development , Hair Cells, Auditory , Mice , Spiral Ganglion
8.
Dev Dyn ; 246(12): 1047-1056, 2017 12.
Article in English | MEDLINE | ID: mdl-28929539

ABSTRACT

BACKGROUND: During murine kidney development, new cortical blood vessels form and pattern in cycles that coincide with cycles of collecting duct branching and the accompanying splitting of the cap mesenchyme (nephron progenitor cell populations that "cap" collecting duct ends). At no point in the patterning cycle do blood vessels enter the cap mesenchyme. We hypothesized that the exclusion of blood vessels from the cap mesenchyme may be controlled, at least in part, by an anti-angiogenic signal expressed by the cap mesenchyme cells. RESULTS: We show that semaphorin-3f (Sema3f), a known anti-angiogenic factor, is expressed in cap mesenchymal cells and its receptor, neuropilin-2 (Nrp2), is expressed by newly forming blood vessels in the cortex of the developing kidney. We hypothesized that Sema3f/Nrp2 signaling excludes vessels from the cap mesenchyme. Genetic ablation of Sema3f and of Nrp2, however, failed to result in vessels invading the cap mesenchyme. CONCLUSIONS: Despite complementary expression patterns, our data suggest that Sema3f and Nrp2 are dispensable for the exclusion of vessels from the cap mesenchyme during kidney development. These results should provoke additional experiments to ascertain the biological significance of Sema3f/Nrp2 expression in the developing kidney. Developmental Dynamics 246:1047-1056, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Kidney , Membrane Proteins/biosynthesis , Mesoderm , Models, Biological , Neovascularization, Physiologic/physiology , Nerve Tissue Proteins/biosynthesis , Neuropilin-2/biosynthesis , Animals , Kidney/blood supply , Kidney/embryology , Membrane Proteins/genetics , Mesoderm/blood supply , Mesoderm/embryology , Mice , Nerve Tissue Proteins/genetics , Neuropilin-2/genetics
9.
Semin Cell Dev Biol ; 24(5): 460-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23660234

ABSTRACT

In mammals, auditory information is processed by the hair cells (HCs) located in the cochlea and then rapidly transmitted to the CNS via a specialized cluster of bipolar afferent connections known as the spiral ganglion neurons (SGNs). Although many anatomical aspects of SGNs are well described, the molecular and cellular mechanisms underlying their genesis, how they are precisely arranged along the cochlear duct, and the guidance mechanisms that promote the innervation of their hair cell targets are only now being understood. Building upon foundational studies of neurogenesis and neurotrophins, we review here new concepts and technologies that are helping to enrich our understanding of the development of the nervous system within the inner ear.


Subject(s)
Cochlear Duct/physiology , Hair Cells, Auditory/physiology , Nerve Growth Factors/genetics , Neurogenesis/physiology , Sensory Receptor Cells/physiology , Spiral Ganglion/physiology , Animals , Cell Movement , Cochlear Duct/cytology , Cochlear Duct/growth & development , Cochlear Duct/innervation , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mechanotransduction, Cellular , Nerve Growth Factors/metabolism , Sensory Receptor Cells/cytology , Spiral Ganglion/cytology , Spiral Ganglion/growth & development , Spiral Ganglion/innervation , Synapses/physiology , Synaptic Transmission
10.
Dev Biol ; 376(1): 86-98, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23318633

ABSTRACT

The organ of Corti, located within the mammalian cochlea, contains a precise mosaic of hair cells (HC) and supporting cells (SC), the patterning of which is critical for auditory function. Progenitors of HCs and SCs are found in the same post-mitotic region of the cochlear duct during early stages of cochlear development, and both HCs and SCs are absent in mice lacking the transcription factor Atoh1. Based on existing data, Atoh1 is thought to be the earliest determinant of HC fate, and to have a cell-autonomous role in HC differentiation, but the lineage of Atoh1-positive cells within the cochlear duct remains unclear. To address this issue, we used an inducible Atoh1(Cre⁎PR) allele to permanently mark Atoh1-expressing cells at different developmental time points. We found that up to 30% of cells from the Atoh1-lineage develop as SCs, and that the number of Atoh1-positive SCs decreases both spatially and temporally in a pattern consistent with ongoing commitment. Modulation of Notch signaling, necessary for formation of the HC-SC mosaic, changes the percentage of cells from the Atoh1-lineage that develop as either HCs or SCs. The HC-SC ratio is also affected by morphogenesis of the cochlea, as inhibiting the outgrowth of the cochlear duct increases the number of Atoh1-lineage cells that develop as SCs. Our results demonstrate that the Atoh1-lineage is established early in cochlear development, but also show that expression of Atoh1 does not absolutely result in commitment to a HC fate.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cochlea/embryology , Gene Expression Regulation, Developmental/physiology , Hair Cells, Auditory/cytology , Labyrinth Supporting Cells/cytology , Age Factors , Analysis of Variance , Animals , Cochlea/cytology , Gene Expression Profiling , Hair Cells, Auditory/metabolism , Immunohistochemistry , Labyrinth Supporting Cells/metabolism , Mice , Receptors, Notch/metabolism , Signal Transduction/physiology
11.
G3 (Bethesda) ; 14(5)2024 05 07.
Article in English | MEDLINE | ID: mdl-38577978

ABSTRACT

Genetic variability significantly contributes to individual differences in skeletal muscle mass; however, the specific genes involved in that process remain elusive. In this study, we examined the role of positional candidates, Rps6ka6 and Pou3f4, of a chromosome X locus, implicated in muscle mass variability in CFW laboratory mice. Histology of hindlimb muscles was studied in CFW male mice carrying the muscle "increasing" allele C (n = 15) or "decreasing" allele T (n = 15) at the peak marker of the locus, rs31308852, and in the Pou3f4y/- and their wild-type male littermates. To study the role of the Rps6ka6 gene, we deleted exon 7 (Rps6ka6-ΔE7) using clustered regularly interspaced palindromic repeats-Cas9 based method in H2Kb myogenic cells creating a severely truncated RSK4 protein. We then tested whether that mutation affected myoblast proliferation, migration, and/or differentiation. The extensor digitorum longus muscle was 7% larger (P < 0.0001) due to 10% more muscle fibers (P = 0.0176) in the carriers of the "increasing" compared with the "decreasing" CFW allele. The number of fibers was reduced by 15% (P = 0.0268) in the slow-twitch soleus but not in the fast-twitch extensor digitorum longus (P = 0.2947) of Pou3f4y/- mice. The proliferation and migration did not differ between the Rps6ka6-ΔE7 and wild-type H2Kb myoblasts. However, indices of differentiation (myosin expression, P < 0.0001; size of myosin-expressing cells, P < 0.0001; and fusion index, P = 0.0013) were significantly reduced in Rps6ka6-ΔE7 cells. This study suggests that the effect of the X chromosome locus on muscle fiber numbers in the fast-twitch extensor digitorum longus is mediated by the Rps6ka6 gene, whereas the Pou3f4 gene affects fiber number in slow-twitch soleus.


Subject(s)
Muscle, Skeletal , POU Domain Factors , Ribosomal Protein S6 Kinases, 90-kDa , Animals , Male , Mice , Alleles , Cell Differentiation , Cell Movement , Cell Proliferation , Genetic Loci , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , POU Domain Factors/metabolism
12.
iScience ; 26(10): 107769, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37720106

ABSTRACT

The cochlea consists of diverse cellular populations working in harmony to convert mechanical stimuli into electrical signals for the perception of sound. Otic mesenchyme cells (OMCs), often considered a homogeneous cell type, are essential for normal cochlear development and hearing. Despite being the most numerous cell type in the developing cochlea, OMCs are poorly understood. OMCs are known to differentiate into spatially and functionally distinct cell types, including fibrocytes of the lateral wall and spiral limbus, modiolar osteoblasts, and specialized tympanic border cells of the basilar membrane. Here, we show that OMCs are transcriptionally and functionally heterogeneous and can be divided into four distinct populations that spatially correspond to OMC-derived cochlear structures. We also show that this heterogeneity and complexity of OMCs commences during early phases of cochlear development. Finally, we describe the cell-cell communication network of the developing cochlea, inferring a major role for OMC in outgoing signaling.

13.
J Neurosci ; 29(11): 3404-18, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295147

ABSTRACT

Reverse signaling via glycosylphosphatidylinositol (GPI)-linked Ephrins may help control cell proliferation and outgrowth within the nervous system, but the mechanisms underlying this process remain poorly understood. In the embryonic enteric nervous system (ENS) of the moth Manduca sexta, migratory neurons forming the enteric plexus (EP cells) express a single Ephrin ligand (GPI-linked MsEphrin), whereas adjacent midline cells that are inhibitory to migration express the cognate receptor (MsEph). Knocking down MsEph receptor expression in cultured embryos with antisense morpholino oligonucleotides allowed the EP cells to cross the midline inappropriately, consistent with the model that reverse signaling via MsEphrin mediates a repulsive response in the ENS. Src family kinases have been implicated in reverse signaling by type-A Ephrins in other contexts, and MsEphrin colocalizes with activated forms of endogenous Src in the leading processes of the EP cells. Pharmacological inhibition of Src within the developing ENS induced aberrant midline crossovers, similar to the effect of blocking MsEphrin reverse signaling. Hyperstimulating MsEphrin reverse signaling with MsEph-Fc fusion proteins induced the rapid activation of endogenous Src specifically within the EP cells, as assayed by Western blots of single embryonic gut explants and by whole-mount immunostaining of cultured embryos. In longer cultures, treatment with MsEph-Fc caused a global inhibition of EP cell migration and outgrowth, an effect that was prevented by inhibiting Src activation. These results support the model that MsEphrin reverse signaling induces the Src-dependent retraction of EP cell processes away from the enteric midline, thereby helping to confine the neurons to their appropriate pathways.


Subject(s)
Cell Movement/physiology , Ephrins/physiology , Glycosylphosphatidylinositols/physiology , Insect Proteins/physiology , Manduca/physiology , Neurons/physiology , src-Family Kinases/physiology , Animals , Humans , Insect Proteins/genetics , Neurons/cytology , Receptors, Eph Family/physiology , Signal Transduction/physiology
14.
eNeuro ; 7(4)2020.
Article in English | MEDLINE | ID: mdl-32675174

ABSTRACT

The mammalian cochlea undergoes a highly dynamic process of growth and innervation during development. This process includes spiral ganglion neuron (SGN) branch refinement, a process whereby Type I SGNs undergo a phase of "debranching" before forming unramified synaptic contacts with inner hair cells. Using Sox2CreERT2 and R26RtdTomato as a strategy to genetically label individual SGNs in mice of both sexes, we report on both a time course of SGN branch refinement and a role for P2rx3 in this process. P2rx3 is an ionotropic ATP receptor that was recently implicated in outer hair cell spontaneous activity and Type II SGN synapse development (Ceriani et al., 2019), but its function in Type I SGN development is unknown. Here, we demonstrate that P2rx3 is expressed by Type I SGNs and hair cells during developmental periods that coincide with SGN branching refinement. P2rx3 null mice show SGNs with more complex branching patterns on their peripheral synaptic terminals and near their cell bodies around the time of birth. Loss of P2rx3 does not appear to confer general changes in axon outgrowth or hair cell formation, and alterations in branching complexity appear to mostly recover by postnatal day (P)6. However, when we examined the distribution of Type I SGN subtypes using antibodies that bind Calb2, Calb1, and Pou4f1, we found that P2rx3 null mice showed an increased proportion of SGNs that express Calb2. These data suggest P2rx3 may be necessary for normal Type I SGN differentiation in addition to serving a role in branch refinement.


Subject(s)
Neurons , Spiral Ganglion , Animals , Cochlea , Female , Hair Cells, Auditory, Inner , Male , Mice , Receptors, Purinergic , Receptors, Purinergic P2X3 , Transcription Factor Brn-3A
15.
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
16.
J Neurosci ; 28(15): 3846-60, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18400884

ABSTRACT

We have investigated whether reverse signaling via a glycosyl-phosphatidylinositol (GPI)-linked ephrin controls the behavior of migratory neurons in vivo. During the formation of the enteric nervous system (ENS) in the moth Manduca, approximately 300 neurons [enteric plexus (EP) cells] migrate onto the midgut via bilaterally paired muscle bands but avoid adjacent midline regions. As they migrate, the EP cells express a single ephrin ligand (MsEphrin; a GPI-linked ligand), whereas the midline cells express the corresponding Eph receptor (MsEph). Blocking endogenous MsEphrin-MsEph receptor interactions in cultured embryos resulted in aberrant midline crossing by the neurons and their processes. In contrast, activating endogenous MsEphrin on the EP cells with dimeric MsEph-Fc constructs inhibited their migration and outgrowth, supporting a role for MsEphrin-dependent reverse signaling in this system. In short-term cultures, blocking endogenous MsEph receptors allowed filopodia from the growth cones of the neurons to invade the midline, whereas activating neuronal MsEphrin led to filopodial retraction. MsEphrin-dependent signaling may therefore guide the migratory enteric neurons by restricting the orientation of their leading processes. Knocking down MsEphrin expression in the EP cells with morpholino antisense oligonucleotides also induced aberrant midline crossing, consistent with the effects of blocking endogenous MsEphrin-MsEph interactions. Unexpectedly, this treatment enhanced the overall extent of migration, indicating that MsEphrin-dependent signaling may also modulate the general motility of the EP cells. These results demonstrate that MsEphrin-MsEph receptor interactions normally prevent midline crossing by migratory neurons within the developing ENS, an effect that is most likely mediated by reverse signaling through this GPI-linked ephrin ligand.


Subject(s)
Cell Movement/physiology , Ephrins/metabolism , Glycosylphosphatidylinositols/metabolism , Manduca/embryology , Neurons/physiology , Signal Transduction/physiology , Animals , Axons/physiology , Embryo, Nonmammalian/physiology , Embryonic Development/physiology , Enteric Nervous System/cytology , Enteric Nervous System/embryology , Ephrins/antagonists & inhibitors , Ephrins/genetics , Growth Cones/physiology , Ligands , Oligonucleotides, Antisense/pharmacology , Pseudopodia/physiology , Receptors, Eph Family/physiology
17.
J Comp Neurol ; 502(2): 175-91, 2007 May 10.
Article in English | MEDLINE | ID: mdl-17348007

ABSTRACT

Eph receptor tyrosine kinases and their ephrin ligands participate in the control of neuronal growth and migration in a variety of contexts, but the mechanisms by which they guide neuronal motility are still incompletely understood. By using the enteric nervous system (ENS) of the tobacco hornworm Manduca sexta as a model system, we have explored whether Manduca ephrin (MsEphrin; a GPI-linked ligand) and its Eph receptor (MsEph) might regulate the migration and outgrowth of enteric neurons. During formation of the Manduca ENS, an identified set of approximately 300 neurons (EP cells) populates the enteric plexus of the midgut by migrating along a specific set of muscle bands forming on the gut, but the neurons strictly avoid adjacent interband regions. By determining the mRNA and protein expression patterns for MsEphrin and the MsEph receptor and by examining their endogenous binding patterns within the ENS, we have demonstrated that the ligand and its receptor are distributed in a complementary manner: MsEphrin is expressed exclusively by the migratory EP cells, whereas the MsEph receptor is expressed by midline interband cells that are normally inhibitory to migration. Notably, MsEphrin could be detected on the filopodial processes of the EP cells that extended up to but not across the midline cells expressing the MsEph receptor. These results suggest a model whereby MsEphrin-dependent signaling regulates the response of migrating neurons to a midline inhibitory boundary, defined by the expression of MsEph receptors in the developing ENS.


Subject(s)
Cell Movement/physiology , Enteric Nervous System/cytology , Ephrins/metabolism , Gene Expression/physiology , Manduca/anatomy & histology , Neurons/physiology , Receptor, EphA1/metabolism , Animals , Biological Evolution , Embryo, Nonmammalian , Microscopy, Electron/methods , Neurons/ultrastructure , RNA, Messenger/metabolism , Receptor, EphA1/genetics
18.
Elife ; 42015 Aug 24.
Article in English | MEDLINE | ID: mdl-26302206

ABSTRACT

Auditory function is dependent on the formation of specific innervation patterns between mechanosensory hair cells (HCs) and afferent spiral ganglion neurons (SGNs). In particular, type I SGNs must precisely connect with inner HCs (IHCs) while avoiding connections with nearby outer HCs (OHCs). The factors that mediate these patterning events are largely unknown. Using sparse-labeling and time-lapse imaging, we visualized for the first time the behaviors of developing SGNs including active retraction of processes from OHCs, suggesting that some type I SGNs contact OHCs before forming synapses with IHCs. In addition, we demonstrate that expression of Semaphorin-3F in the OHC region inhibits type I SGN process extension by activating Neuropilin-2 receptors expressed on SGNs. These results suggest a model in which cochlear innervation patterns by type I SGNs are determined, at least in part, through a Semaphorin-3F-mediated inhibitory signal that impedes processes from extending beyond the IHC region.


Subject(s)
Cell Communication , Cochlea/embryology , Hair Cells, Auditory, Inner/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Neuropilin-2/metabolism , Spiral Ganglion/physiology , Animals , Cells, Cultured , Immunohistochemistry , Mice , Organ Culture Techniques , Time-Lapse Imaging
19.
PLoS One ; 9(10): e109043, 2014.
Article in English | MEDLINE | ID: mdl-25299585

ABSTRACT

The temporal bone encases conductive and sensorineural elements of the ear. Mutations of POU3F4 are associated with unique temporal bone abnormalities and X-linked mixed deafness (DFNX2/DFN3). However, the target genes and developmental processes controlled by POU3F4 transcription factor activity have remained largely uncharacterized. Ephrin-B2 (Efnb2) is a signaling molecule with well-documented effects on cell adhesion, proliferation, and migration. Our analyses of targeted mouse mutants revealed that Efnb2 loss-of-function phenocopies temporal bone abnormalities of Pou3f4 hemizygous null neonates: qualitatively identical malformations of the stapes, styloid process, internal auditory canal, and cochlear capsule were present in both mutants. Using failed/insufficient separation of the stapes and styloid process as a quantitative trait, we found that single gene Efnb2 loss-of-function and compound Pou3f4/Efnb2 loss-of-function caused a more severe phenotype than single gene Pou3f4 loss-of-function. Pou3f4 and Efnb2 gene expression domains overlapped at the site of impending stapes-styloid process separation and at subcapsular mesenchyme surrounding the cochlea; at both these sites, Efnb2 expression was attenuated in Pou3f4 hemizygous null mutants relative to control. Results of immunoprecipitation experiments using chromatin isolated from nascent middle ear mesenchyme supported the hypothesis of a physical association between Pou3f4 and specific non-coding sequence of Efnb2. We propose that Efnb2 is a target of Pou3f4 transcription factor activity and an effector of mesenchymal patterning during temporal bone development.


Subject(s)
Bone Development/physiology , Nerve Tissue Proteins/metabolism , POU Domain Factors/metabolism , Temporal Bone/metabolism , Temporal Bone/physiology , Animals , Cochlea/metabolism , Cochlea/physiology , Ear, Inner/metabolism , Ear, Inner/physiology , Ear, Middle/metabolism , Ear, Middle/physiology , Ephrin-B2/genetics , Ephrin-B2/metabolism , Female , Mice , Mice, Inbred C57BL , Mutation/genetics , Nerve Tissue Proteins/genetics , POU Domain Factors/genetics
20.
Neuron ; 73(1): 49-63, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22243746

ABSTRACT

Peripheral axons from auditory spiral ganglion neurons (SGNs) form an elaborate series of radially and spirally oriented projections that interpret complex aspects of the auditory environment. However, the developmental processes that shape these axon tracts are largely unknown. Radial bundles are comprised of dense SGN fascicles that project through otic mesenchyme to form synapses within the cochlea. Here, we show that radial bundle fasciculation and synapse formation are disrupted when Pou3f4 (DFNX2) is deleted from otic mesenchyme. Further, we demonstrate that Pou3f4 binds to and directly regulates expression of Epha4, Epha4⁻/⁻ mice present similar SGN defects, and exogenous EphA4 promotes SGN fasciculation in the absence of Pou3f4. Finally, Efnb2 deletion in SGNs leads to similar fasciculation defects, suggesting that ephrin-B2/EphA4 interactions are critical during this process. These results indicate a model whereby Pou3f4 in the otic mesenchyme establishes an Eph/ephrin-mediated fasciculation signal that promotes inner radial bundle formation.


Subject(s)
Axons/physiology , Mesoderm/cytology , Nerve Tissue Proteins/metabolism , Neurons/cytology , POU Domain Factors/metabolism , Signal Transduction/physiology , Spiral Ganglion/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Coculture Techniques , Embryo, Mammalian , Ephrin-B2/deficiency , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , POU Domain Factors/genetics , Pregnancy , Signal Transduction/genetics , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL