Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurophysiol ; 125(6): 2461-2479, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33949873

RESUMO

Spiral ganglion neurons (SGNs) form single synapses on inner hair cells (IHCs), transforming sound-induced IHC receptor potentials into trains of action potentials. SGN neurons are classified by spontaneous firing rates as well as their threshold response to sound intensity levels. We investigated the hypothesis that synaptic specializations underlie mouse SGN response properties and vary with pillar versus modiloar synapse location around the hair cell. Depolarizing hair cells with 40 mM K+ increased the rate of postsynaptic responses. Pillar synapses matured later than modiolar synapses. Excitatory postsynaptic current (EPSC) amplitude, area, and number of underlying events per EPSC were similar between synapse locations at steady state. However, modiolar synapses produced larger monophasic EPSCs when EPSC rates were low and EPSCs became more multiphasic and smaller in amplitude when rates were higher, while pillar synapses produced more monophasic and larger EPSCs when the release rates were higher. We propose that pillar and modiolar synapses have different operating points. Our data provide insight into underlying mechanisms regulating EPSC generation.NEW & NOTEWORTHY Data presented here provide the first direct functional evidence of late synaptic maturation of the hair cell- spiral ganglion neuron synapse, where pillar synapses mature after postnatal day 20. Data identify a presynaptic difference in release during stimulation. This difference may in part drive afferent firing properties.


Assuntos
Cóclea/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Neurônios/fisiologia , Gânglio Espiral da Cóclea/fisiologia , Sinapses/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gânglio Espiral da Cóclea/crescimento & desenvolvimento
2.
Neural Plast ; 2020: 9387560, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33123191

RESUMO

Autophagy and apoptosis have a complex interplay in the early embryo development. The development of spiral ganglion neurons (SGNs) in addition to Corti's organ in the mammalian cochlea remains crucial in the first two-week postnatal period. To investigate the roles of apoptosis and autophagy in the development of SGNs, light microscopy was used to observe the morphological changes of SGNs. The number of SGNs was decreased from P1 to P7 and plateaued from P10 to P14. Immunohistochemistry results revealed positive expression of cleaved-caspase3, bcl-2, microtubule-associated protein light chain 3-II (LC3-II), Beclin1, and sequestosome 1 (SQSTM1/P62) in SGNs. The apoptotic bodies and autophagosomes and autolysosomes were also identified by transmission electron microscopy at P1 and P7. Real-time PCR and western blotting results revealed that the apoptotic activity peaked at P7 and the autophagy activity was gradually upregulated along with the development. Taken together, our results for the first time showed that autophagy and apoptosis in SGNs play distinct roles during specific developmental phases in a time-dependent manner.


Assuntos
Apoptose , Proteínas Relacionadas à Autofagia/metabolismo , Neurônios/metabolismo , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/metabolismo , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Ratos Sprague-Dawley
3.
J Comp Neurol ; 528(12): 1967-1985, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31994726

RESUMO

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.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Fatores do Domínio POU/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Animais , Sobrevivência Celular , Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento
4.
J Neurosci ; 39(41): 8013-8023, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31462532

RESUMO

Type II spiral ganglion neurons provide afferent innervation to outer hair cells of the cochlea and are proposed to have nociceptive functions important for auditory function and homeostasis. These neurons are anatomically distinct from other classes of spiral ganglion neurons because they extend a peripheral axon beyond the inner hair cells that subsequently makes a distinct 90 degree turn toward the cochlear base. As a result, patterns of outer hair cell innervation are coordinated with the tonotopic organization of the cochlea. Previously, it was shown that peripheral axon turning is directed by a nonautonomous function of the core planar cell polarity (PCP) protein VANGL2. We demonstrate using mice of either sex that Fzd3 and Fzd6 similarly regulate axon turning, are functionally redundant with each other, and that Fzd3 genetically interacts with Vangl2 to guide this process. FZD3 and FZD6 proteins are asymmetrically distributed along the basolateral wall of cochlear-supporting cells, and are required to promote or maintain the asymmetric distribution of VANGL2 and CELSR1. These data indicate that intact PCP complexes formed between cochlear-supporting cells are required for the nonautonomous regulation of axon pathfinding. Consistent with this, in the absence of PCP signaling, peripheral axons turn randomly and often project toward the cochlear apex. Additional analyses of Porcn mutants in which WNT secretion is reduced suggest that noncanonical WNT signaling establishes or maintains PCP signaling in this context. A deeper understanding of these mechanisms is necessary for repairing auditory circuits following acoustic trauma or promoting cochlear reinnervation during regeneration-based deafness therapies.SIGNIFICANCE STATEMENT Planar cell polarity (PCP) signaling has emerged as a complementary mechanism to classical axon guidance in regulating axon track formation, axon outgrowth, and neuronal polarization. The core PCP proteins are also required for auditory circuit assembly, and coordinate hair cell innervation with the tonotopic organization of the cochlea. This is a non-cell-autonomous mechanism that requires the formation of PCP protein complexes between cochlear-supporting cells located along the trajectory of growth cone navigation. These findings are significant because they demonstrate how the fidelity of auditory circuit formation is ensured during development, and provide a mechanism by which PCP proteins may regulate axon outgrowth and guidance in the CNS.


Assuntos
Cóclea/inervação , Receptores Frizzled/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Gânglio Espiral da Cóclea/citologia , Aciltransferases/genética , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Polaridade Celular , Cóclea/crescimento & desenvolvimento , Feminino , Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Externas , Masculino , Proteínas de Membrana/genética , Camundongos , Mutação/genética , Órgão Espiral/crescimento & desenvolvimento , Órgão Espiral/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia
5.
Synapse ; 73(5): e22087, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30592086

RESUMO

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.


Assuntos
Neurogênese , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Sinapses/fisiologia , Animais , Células Ciliadas Auditivas/citologia , Células Ciliadas Auditivas/fisiologia , Humanos , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/fisiologia , Sinapses/ultraestrutura
6.
Neuron ; 99(3): 511-524.e5, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30077356

RESUMO

Neurons in the developing auditory system exhibit spontaneous bursts of activity before hearing onset. How this intrinsically generated activity influences development remains uncertain, because few mechanistic studies have been performed in vivo. We show using macroscopic calcium imaging in unanesthetized mice that neurons responsible for processing similar frequencies of sound exhibit highly synchronized activity throughout the auditory system during this critical phase of development. Spontaneous activity normally requires synaptic excitation of spiral ganglion neurons (SGNs). Unexpectedly, tonotopic spontaneous activity was preserved in a mouse model of deafness in which glutamate release from hair cells is abolished. SGNs in these mice exhibited enhanced excitability, enabling direct neuronal excitation by supporting cell-induced potassium transients. These results indicate that homeostatic mechanisms maintain spontaneous activity in the pre-hearing period, with significant implications for both circuit development and therapeutic approaches aimed at treating congenital forms of deafness arising through mutations in key sensory transduction components.


Assuntos
Córtex Auditivo/crescimento & desenvolvimento , Vias Auditivas/crescimento & desenvolvimento , Audição/fisiologia , Homeostase/fisiologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Estimulação Acústica/métodos , Animais , Córtex Auditivo/química , Vias Auditivas/química , Cóclea/química , Cóclea/crescimento & desenvolvimento , Feminino , Células Ciliadas Auditivas/química , Células Ciliadas Auditivas/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Distribuição Aleatória , Gânglio Espiral da Cóclea/química
7.
Wiley Interdiscip Rev Dev Biol ; 7(6): e324, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29944783

RESUMO

Developing sensory systems must coordinate the growth of neural circuitry spanning from receptors in the peripheral nervous system (PNS) to multilayered networks within the central nervous system (CNS). This breadth presents particular challenges, as nascent processes must navigate across the CNS-PNS boundary and coalesce into a tightly intermingled wiring pattern, thereby enabling reliable integration from the PNS to the CNS and back. In the auditory system, feedforward spiral ganglion neurons (SGNs) from the periphery collect sound information via tonotopically organized connections in the cochlea and transmit this information to the brainstem for processing via the VIII cranial nerve. In turn, feedback olivocochlear neurons (OCNs) housed in the auditory brainstem send projections into the periphery, also through the VIII nerve. OCNs are motor neuron-like efferent cells that influence auditory processing within the cochlea and protect against noise damage in adult animals. These aligned feedforward and feedback systems develop in parallel, with SGN central axons reaching the developing auditory brainstem around the same time that the OCN axons extend out toward the developing inner ear. Recent findings have begun to unravel the genetic and molecular mechanisms that guide OCN development, from their origins in a generic pool of motor neuron precursors to their specialized roles as modulators of cochlear activity. One recurrent theme is the importance of efferent-afferent interactions, as afferent SGNs guide OCNs to their final locations within the sensory epithelium, and efferent OCNs shape the activity of the developing auditory system. This article is categorized under: Nervous System Development > Vertebrates: Regional Development.


Assuntos
Vias Auditivas/metabolismo , Tronco Encefálico/metabolismo , Cóclea/metabolismo , Nervos Cranianos/metabolismo , Vias Eferentes/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Animais , Vias Auditivas/citologia , Vias Auditivas/crescimento & desenvolvimento , Tronco Encefálico/citologia , Tronco Encefálico/crescimento & desenvolvimento , Cóclea/citologia , Cóclea/crescimento & desenvolvimento , Cóclea/inervação , Nervos Cranianos/citologia , Nervos Cranianos/crescimento & desenvolvimento , Vias Eferentes/citologia , Vias Eferentes/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Morfogênese/genética , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neurônios Aferentes/citologia , Neurônios Aferentes/metabolismo , Neurônios Eferentes/citologia , Neurônios Eferentes/metabolismo , Transdução de Sinais , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Otol Neurotol ; 39(1): 119-126, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29227456

RESUMO

HYPOTHESIS: Microtopographical patterns generated by photopolymerization of methacrylate polymer systems will direct growth of neurites from adult neurons, including spiral ganglion neurons (SGNs). BACKGROUND: Cochlear implants (CIs) provide hearing perception to patients with severe to profound hearing loss. However, their ability to encode complex auditory stimuli is limited due, in part, to poor spatial resolution caused by spread of the electrical currents in the inner ear. Directing the regrowth of SGN peripheral processes towards stimulating electrodes could help reduce current spread and improve spatial resolution provided by the CI. Previous work has demonstrated that micro- and nano-scale patterned surfaces precisely guide the growth of neurites from a variety of neonatal neurons including SGNs. Here, we sought to determine the extent to which adult neurons likewise respond to these topographical surface features. METHODS: Photopolymerization was used to fabricate methacrylate polymer substrates with micropatterned surfaces of varying amplitudes and periodicities. Dissociated adult dorsal root ganglion neurons (DRGNs) and SGNs were cultured on these surfaces and the alignment of the neurite processes to the micropatterns was determined. RESULTS: Neurites from both adult DRGNs and SGNs significantly aligned to the patterned surfaces similar to their neonatal counterparts. Further DRGN and SGN neurite alignment increased as the amplitude of the microfeatures increased. Decreased pattern periodicity also improved neurite alignment. CONCLUSION: Microscale surface topographic features direct the growth of adult SGN neurites. Topographical features could prove useful for guiding growth of SGN peripheral axons towards a CI electrode array.


Assuntos
Implantes Cocleares , Regeneração Tecidual Guiada/métodos , Regeneração Nervosa , Neuritos , Animais , Células Cultivadas , Gânglios Espinais/crescimento & desenvolvimento , Polímeros , Gânglio Espiral da Cóclea/crescimento & desenvolvimento
9.
Front Neural Circuits ; 11: 25, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28450830

RESUMO

We investigate the importance of the degree of peripheral or central target differentiation for mouse auditory afferent navigation to the organ of Corti and auditory nuclei in three different mouse models: first, a mouse in which the differentiation of hair cells, but not central auditory nuclei neurons is compromised (Atoh1-cre; Atoh1f/f ); second, a mouse in which hair cell defects are combined with a delayed defect in central auditory nuclei neurons (Pax2-cre; Atoh1f/f ), and third, a mouse in which both hair cells and central auditory nuclei are absent (Atoh1-/-). Our results show that neither differentiated peripheral nor the central target cells of inner ear afferents are needed (hair cells, cochlear nucleus neurons) for segregation of vestibular and cochlear afferents within the hindbrain and some degree of base to apex segregation of cochlear afferents. These data suggest that inner ear spiral ganglion neuron processes may predominantly rely on temporally and spatially distinct molecular cues in the region of the targets rather than interaction with differentiated target cells for a crude topological organization. These developmental data imply that auditory neuron navigation properties may have evolved before auditory nuclei.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Diferenciação Celular/genética , Células Ciliadas Auditivas/fisiologia , Malformações do Sistema Nervoso/patologia , Fator de Transcrição PAX2/deficiência , Rombencéfalo/patologia , Gânglio Espiral da Cóclea , Animais , Animais Recém-Nascidos , Vias Auditivas/embriologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Núcleo Coclear/citologia , Núcleo Coclear/embriologia , Núcleo Coclear/crescimento & desenvolvimento , Embrião de Mamíferos , Camundongos , Camundongos Knockout , Malformações do Sistema Nervoso/genética , Fator de Transcrição PAX2/genética , Gânglio Espiral da Cóclea/embriologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/patologia , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
10.
Artigo em Chinês | MEDLINE | ID: mdl-29798427

RESUMO

Neurotrophin-3 (NT-3) attracted increasing attention about NTFs researches in recent years. But the mechanism of promoting the development of neurons and neurite extension is not clear. Recombinant human NT-3 or NT-3 gene is commonly used in the treatment of peripheral and central neurons system damage. When rats are born, the cochlear is not mature yet. It is a suitable experimental animal for studying the morphological and functional development of the peripheral auditory pathway. It was found that NT-3 could promote the survival, growth, division and extension of the cochlear neurons in rats. To make clear the role of NT-3 in the development of spiral ganglion in the rat cochlear will be of significance for the treatment of nervous hearing loss by NT-3 in the future.


Assuntos
Neurotrofina 3/fisiologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Animais , Cóclea , Humanos , Fatores de Crescimento Neural , Neurônios , Ratos , Proteínas Recombinantes
11.
Colloids Surf B Biointerfaces ; 149: 105-114, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27736723

RESUMO

Cochlear implants (CI) allow for hearing rehabilitation in patients with sensorineural hearing loss or deafness. Restricted CI performance results from the spatial gap between spiral ganglion neurons and the CI, causing current spread that limits spatially restricted stimulation and impairs frequency resolution. This may be substantially improved by guiding peripheral processes of spiral ganglion neurons towards and onto the CI electrode contacts. An injectable, peptide-based hydrogel was developed which may provide a permissive scaffold to facilitate neurite growth towards the CI. To test hydrogel capacity to attract spiral ganglion neurites, neurite outgrowth was quantified in an in vitro model using a custom-designed hydrogel scaffold and PuraMatrix®. Neurite attachment to native hydrogels is poor, but significantly improved by incorporation of brain-derived neurotrophic factor (BDNF), covalent coupling of the bioactive laminin epitope IKVAV and the incorporation a full length laminin to hydrogel scaffolds. Incorporation of full length laminin protein into a novel custom-designed biofunctionalized hydrogel (IKVAV-GGG-SIINFEKL) allows for neurite outgrowth into the hydrogel scaffold. The study demonstrates that peptide-based hydrogels can be specifically biofunctionalized to provide a permissive scaffold to attract neurite outgrowth from spiral ganglion neurons. Such biomaterials appear suitable to bridge the spatial gap between neurons and the CI.


Assuntos
Hidrogéis/farmacologia , Neuritos/efeitos dos fármacos , Crescimento Neuronal/efeitos dos fármacos , Peptídeos/farmacologia , Gânglio Espiral da Cóclea/efeitos dos fármacos , Alicerces Teciduais , Sequência de Aminoácidos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Cóclea/efeitos dos fármacos , Cóclea/fisiologia , Cóclea/ultraestrutura , Implantes Cocleares , Feminino , Hidrogéis/química , Laminina/metabolismo , Laminina/farmacologia , Masculino , Camundongos , Neuritos/fisiologia , Neuritos/ultraestrutura , Crescimento Neuronal/fisiologia , Peptídeos/química , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/fisiologia , Gânglio Espiral da Cóclea/ultraestrutura , Técnicas de Cultura de Tecidos
12.
Mol Med Rep ; 13(5): 4357-64, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27052602

RESUMO

Chemotactic cytokines (chemokines) are a highly conserved class of secreted signaling molecules that are important in various cellular processes. CXC chemokine ligand 12 (CXCL12) and its receptor, CXC chemokine receptor 4 (CXCR4) have been previously reported to be crucial for the establishment of neural networks in different neuronal systems. However, it is unclear whether the CXCL12/CXCR4 signaling pathway regulates the development of the cochlea. The current study investigated the effects of the CXCL12/CXCR4 signaling pathway on cochlear development in neonatal mice. The expression levels of CXCL12 and CXCR4 were detected using immunofluorescence, reverse transcription­quantitative polymerase chain reaction and western blot analysis demonstrating that CXCL12 and CXCR4 expression were significantly increased during cochlear development in neonatal mice. Treatment of spiral ganglion neurons with CXCL12 significantly decreased the protein expression levels of caspase­3 and cleaved caspase­3, indicating that CXCL12/CXCR4 signaling increased cell survival of spiral ganglion neurons. Furthermore, CXCL12 treatment significantly increased the number and length of neurites extending from spiral ganglion neurons. By contrast, the in vitro effects of CXCL12 were significantly abrogated by AMD100, a CXCR4 antagonist. Additionally, inhibiting CXCL12/CXCR4 signaling in neonatal mice significantly reduced the cell number and altered the morphology of spiral ganglion neurons in vivo. Thus, the present study indicates that the CXCL12/CXCR4 signaling pathway is important during the development of cochleae in neonatal mice.


Assuntos
Quimiocina CXCL12/biossíntese , Regulação da Expressão Gênica/fisiologia , Neuritos/metabolismo , Receptores CXCR4/biossíntese , Transdução de Sinais/fisiologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Animais , Caspase 3/biossíntese , Camundongos , Gânglio Espiral da Cóclea/citologia
13.
Sci Rep ; 6: 23799, 2016 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-27040640

RESUMO

Proper structural organization of spiral ganglion (SG) innervation is crucial for normal hearing function. However, molecular mechanisms underlying the developmental formation of this precise organization remain not well understood. Here, we report in the developing mouse cochlea that deleted in colorectal cancer (Dcc) contributes to the proper organization of spiral ganglion neurons (SGNs) within the Rosenthal's canal and of SGN projections toward both the peripheral and central auditory targets. In Dcc mutant embryos, mispositioning of SGNs occurred along the peripheral auditory pathway with misrouted afferent fibers and reduced synaptic contacts with hair cells. The central auditory pathway simultaneously exhibited similar defective phenotypes as in the periphery with abnormal exit of SGNs from the Rosenthal's canal towards central nuclei. Furthermore, the axons of SGNs ascending into the cochlear nucleus had disrupted bifurcation patterns. Thus, Dcc is necessary for establishing the proper spatial organization of SGNs and their fibers in both peripheral and central auditory pathways, through controlling axon targeting and cell migration. Our results suggest that Dcc plays an important role in the developmental formation of peripheral and central auditory circuits, and its mutation may contribute to sensorineural hearing loss.


Assuntos
Vias Auditivas/anormalidades , Cóclea/anormalidades , Mutação , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/genética , Animais , Vias Auditivas/embriologia , Vias Auditivas/metabolismo , Cóclea/embriologia , Cóclea/metabolismo , Receptor DCC , Desenvolvimento Embrionário , Perda Auditiva Neurossensorial/genética , Camundongos , Neurônios/fisiologia , Receptores de Superfície Celular/metabolismo , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/metabolismo , Proteínas Supressoras de Tumor/metabolismo
14.
Neuroscience ; 325: 50-62, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27012610

RESUMO

Neural activity during early development is known to alter innervation pathways in the central and peripheral nervous systems. We sought to examine how reduced sound-induced sensory activity in the cochlea affected the consolidation of glutamatergic synapses between inner hair cells (IHC) and the primary auditory neurons as these synapses play a primary role in transmitting sound information to the brain. A unilateral conductive hearing loss was induced prior to the onset of sound-mediated stimulation of the sensory hair cells, by rupturing the tympanic membrane and dislocating the auditory ossicles in the left ear of P11 mice. Auditory brainstem responses at P15 and P21 showed a 40-50-dB increase in thresholds for frequencies 8-32kHz in the dislocated ear relative to the control ear. Immunohistochemistry and confocal microscopy were subsequently used to examine the effect of this attenuation of sound stimulation on the expression of RIBEYE, which comprises the presynaptic ribbons, Shank-1, a postsynaptic scaffolding protein, and the GluA2/3 and 4 subunits of postsynaptic AMPA receptors. Our results show that dislocation did not alter the number of pre- or postsynaptic protein puncta. However, dislocation did increase the size of RIBEYE, GluA4, GluA2/3 and Shank-1 puncta, with postsynaptic changes preceding presynaptic changes. Our data suggest that a reduction in sound stimulation during auditory development induces plasticity in the molecular make-up of IHC glutamatergic synapses, but does not affect the number of these synapses. Up-regulation of synaptic proteins with sound attenuation may facilitate a compensatory increase in synaptic transmission due to the reduced sensory stimulation of the IHC.


Assuntos
Cóclea/crescimento & desenvolvimento , Células Ciliadas Auditivas Internas/metabolismo , Terminações Pré-Sinápticas/metabolismo , Receptores de AMPA/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Estimulação Acústica , Oxirredutases do Álcool , Animais , Vias Auditivas/crescimento & desenvolvimento , Vias Auditivas/metabolismo , Proteínas Correpressoras , Proteínas de Ligação a DNA/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Células Ciliadas Auditivas Internas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Privação Sensorial , Gânglio Espiral da Cóclea/crescimento & desenvolvimento
15.
Neuroscience ; 319: 46-58, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26812032

RESUMO

Sensorineural hearing loss, as a consequence of acoustic trauma, aging, genetic defects or ototoxic drugs, is highly associated with irreversible damage of cochlear hair cells (HCs) and secondary degeneration of spiral ganglion (SG) cells. Cochlear implants (CIs), which bypass the lost HC function by direct electrical stimulation of the remaining auditory neurons, offer an effective therapy option. Several studies imply that components of the extracellular matrix (ECM) have a great impact on the adhesion and growth of spiral ganglion neurons (SGNs) during development. Based on these findings, ECM proteins might act as bioactive CI substrates to optimize the electrode-nerve interface and to improve efficacy of these implants. In the present study, we focused on the ECM glycoproteins Tenascin-C (TN-C), Laminin (LN), and Fibronectin (FN), which show a prominent expression along the growth route of SGNs and the niche around HCs during murine postnatal development in vivo. We compared their influence on adhesion, neurite length, and neurite number of SGNs in vitro. Moreover, we studied the expression of the chondroitin sulfate proteoglycan (CSPG) dermatan sulfate-dependent proteoglycan-1 (DSD-1-PG), an interaction partner of TN-C. In sum, our in vitro data suggest that TN-C acts as an anti-adhesive and inhibitory factor for the growth of SGNs. The DSD-1 carbohydrate epitope is specifically localized to HC stereocilia and SG fibers. Interestingly, TN-C and the DSD-1-PG exhibit a mutually exclusive expression pattern, with the exception of a very restricted region beneath the habenula perforata, where SG neurites grow through the basilar membrane (BM) toward the HCs. The complementary expression of TN-C, LN, FN, and the DSD-1 epitope suggests that TN-C may act as an important boundary formation molecule in the developing postnatal mouse inner ear, which makes it a promising candidate to regulate neurite outgrowth in the light of CIs.


Assuntos
Neurogênese/fisiologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Tenascina/metabolismo , Animais , Células Cultivadas , Matriz Extracelular/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos BALB C , Neuritos
16.
Dev Neurobiol ; 76(4): 452-69, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26178595

RESUMO

During the development of periphery auditory circuitry, spiral ganglion neurons (SGNs) form a spatially precise pattern of innervation of cochlear hair cells (HCs), which is an essential structural foundation for central auditory processing. However, molecular mechanisms underlying the developmental formation of this precise innervation pattern remain not well understood. Here, we specifically examined the involvement of Eph family members in cochlear development. By performing RNA-sequencing for different types of cochlear cell, in situ hybridization, and immunohistochemistry, we found that EphA7 was strongly expressed in a large subset of SGNs. In EphA7 deletion mice, there was a reduction in the number of inner radial bundles originating from SGNs and projecting to HCs as well as in the number of ribbon synapses on inner hair cells (IHCs), as compared with wild-type or heterozygous mutant mice, attributable to fewer type I afferent fibers. The overall activity of the auditory nerve in EphA7 deletion mice was also reduced, although there was no significant change in the hearing intensity threshold. In vitro analysis further suggested that the reduced innervation of HCs by SGNs could be attributed to a role of EphA7 in regulating outgrowth of SGN neurites as knocking down EphA7 in SGNs resulted in diminished SGN fibers. In addition, suppressing the activity of ERK1/2, a potential downstream target of EphA7 signaling, either with specific inhibitors in cultured explants or by knocking out Prkg1, also resulted in reduced SGN fibers. Together, our results suggest that EphA7 plays an important role in the developmental formation of cochlear innervation pattern through controlling SGN fiber ontogeny. Such regulation may contribute to the salience level of auditory signals presented to the central auditory system.


Assuntos
Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Neurônios/metabolismo , Receptor EphA7/metabolismo , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Cóclea/citologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Perfilação da Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Neuritos/metabolismo , Neurônios/citologia , Receptor EphA7/genética , Gânglio Espiral da Cóclea/citologia , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
17.
Hear Res ; 330(Pt B): 157-69, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26231304

RESUMO

Sound signal is detected by sensory hair cells located in the cochlear region of the inner ear, and transmitted to the central nervous system by the spiral ganglion neurons (SGNs). These bipolar neurons develop long peripheral processes to connect hair cells, forming ribbon synapses, specialised for the precision and speed required to process auditory information. The establishment of a complex innervation pattern relies on specific signals, intrinsic to SGNs or provided by neighbouring cells, which are tightly controlled in time and space. In this paper, we review recent advances about stepwise development of afferent auditory neuronal circuitries, from neuron specification within the early otic vesicle to definitive synaptic connections with target cells. We especially focus on the cellular and molecular developmental changes involved in fibre outgrowth and extension to the sensory epithelium, specific afferent targeting to hair cells, and synaptic pruning.


Assuntos
Cóclea/inervação , Nervo Coclear/fisiologia , Células Ciliadas Auditivas/fisiologia , Audição , Neurogênese , Transmissão Sináptica , Animais , Vias Auditivas/fisiologia , Percepção Auditiva , Cóclea/crescimento & desenvolvimento , Nervo Coclear/crescimento & desenvolvimento , Nervo Coclear/metabolismo , Células Ciliadas Auditivas/metabolismo , Humanos , Morfogênese , Proteínas do Tecido Nervoso/metabolismo , Gânglio Espiral da Cóclea/crescimento & desenvolvimento
18.
Eur J Histochem ; 59(2): 2486, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-26150157

RESUMO

Inositol 1, 4, 5-trisphosphate receptor (IP3R) has been established to be essential for hearing. However, the expression of IP3R in the cochlea in the period of auditory development remains unknown. We investigated the expression of IP3R in the developing rat cochlea using immunohistochemistry and real-time reverse transcription polymerase chain reaction (RT-PCR). We observed its presence in the developing rat cochlea, and changes in IP3R protein expressions from the early post-natal period to adult. At birth (post-natal day 0, P0), IP3R expression was only found in Hensen's cell. IP3R immunoreactivity first appeared in the sensory hair cells in the organ of Corti at P2. This localization was confirmed by means of double-labeling experiments with Myosin VIIA, a marker for cochlear hair cells. Colocalization of IP3R and Myosin VIIA from P2 to the second post-natal week suggested early expression of IP3R in developing inner and outer hair cells. Claudius' cells near the spiral ligament were labelled for IP3R from P8 onwards. Transient IP3R expression was observed in the stria vascularis in early post-natal rat from P4 to P8. Spiral ganglion neurons also exhibited weaker IP3R fluorescence signals during post-natal development. The results of RT-PCR demonstrated that all three IP3R isoforms (IP3R1, IP3R2, and IP3R3) were present in rat cochlea during four different developmental stages of cochlea, from P0 to P28. Present immunohistochemical evidence for both change and maintenance of expression of IP3R during post-natal development of the rat cochlea indicated the possible involvement of IP3R-mediated calcium signaling in cochlear development.


Assuntos
Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Animais , Western Blotting , Sinalização do Cálcio/fisiologia , Feminino , Células Ciliadas Auditivas Internas/metabolismo , Imuno-Histoquímica , Masculino , Microscopia Confocal , Miosina VIIa , Miosinas/metabolismo , Órgão Espiral/crescimento & desenvolvimento , Órgão Espiral/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/metabolismo , Estria Vascular/metabolismo
19.
J Neurosci ; 35(20): 7878-91, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25995473

RESUMO

During nervous system development, critical periods are usually defined as early periods during which manipulations dramatically change neuronal structure or function, whereas the same manipulations in mature animals have little or no effect on the same property. Neurons in the ventral cochlear nucleus (CN) are dependent on excitatory afferent input for survival during a critical period of development. Cochlear removal in young mammals and birds results in rapid death of target neurons in the CN. Cochlear removal in older animals results in little or no neuron death. However, the extent to which hair-cell-specific afferent activity prevents neuronal death in the neonatal brain is unknown. We further explore this phenomenon using a new mouse model that allows temporal control of cochlear hair cell deletion. Hair cells express the human diphtheria toxin (DT) receptor behind the Pou4f3 promoter. Injections of DT resulted in nearly complete loss of organ of Corti hair cells within 1 week of injection regardless of the age of injection. Injection of DT did not influence surrounding supporting cells directly in the sensory epithelium or spiral ganglion neurons (SGNs). Loss of hair cells in neonates resulted in rapid and profound neuronal loss in the ventral CN, but not when hair cells were eliminated at a more mature age. In addition, normal survival of SGNs was dependent on hair cell integrity early in development and less so in mature animals. This defines a previously undocumented critical period for SGN survival.


Assuntos
Núcleo Coclear/crescimento & desenvolvimento , Células Ciliadas Auditivas/citologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Animais , Morte Celular , Núcleo Coclear/citologia , Núcleo Coclear/fisiologia , Toxina Diftérica/farmacologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Audição , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/fisiologia
20.
Neurosci Lett ; 584: 265-9, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25451728

RESUMO

The molecular mechanisms controlling the proliferation and differentiation of spiral ganglion cells (SGCs) in the inner ear are still largely unknown. TIS21 is a transcriptional cofactor that shows antiproliferative, antiapoptotic, and prodifferentiative effects on neural progenitor cells. To investigate the function of TIS21 during SGC development, we analyzed SGC neurogenesis from embryonic day 13.5 (E13.5) to postnatal day 4 (P4) in Tis21-GFP knock-in mice, in which the protein-encoding exon of the Tis21 gene was replaced by EGFP. Through E13.5 to P4, we found fewer SGCs in homozygous Tis21-GFP knock-in mice than in wild-type mice. Our results suggest that TIS21 is required for development of SGCs. Deleting Tis21 may affect progenitor cells or neuroblasts at the beginning of cochlear-vestibular ganglion formation and would consequently lead to a decrease in the number of SGCs.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Neurogênese , Gânglio Espiral da Cóclea/embriologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Proteínas Supressoras de Tumor/metabolismo , Animais , Animais Recém-Nascidos , Contagem de Células , Técnicas de Introdução de Genes , Homozigoto , Proteínas Imediatamente Precoces/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gânglio Espiral da Cóclea/citologia , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...