Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(37): e2207433119, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36074819

RESUMO

A cardinal feature of the auditory pathway is frequency selectivity, represented in a tonotopic map from the cochlea to the cortex. The molecular determinants of the auditory frequency map are unknown. Here, we discovered that the transcription factor ISL1 regulates the molecular and cellular features of auditory neurons, including the formation of the spiral ganglion and peripheral and central processes that shape the tonotopic representation of the auditory map. We selectively knocked out Isl1 in auditory neurons using Neurod1Cre strategies. In the absence of Isl1, spiral ganglion neurons migrate into the central cochlea and beyond, and the cochlear wiring is profoundly reduced and disrupted. The central axons of Isl1 mutants lose their topographic projections and segregation at the cochlear nucleus. Transcriptome analysis of spiral ganglion neurons shows that Isl1 regulates neurogenesis, axonogenesis, migration, neurotransmission-related machinery, and synaptic communication patterns. We show that peripheral disorganization in the cochlea affects the physiological properties of hearing in the midbrain and auditory behavior. Surprisingly, auditory processing features are preserved despite the significant hearing impairment, revealing central auditory pathway resilience and plasticity in Isl1 mutant mice. Mutant mice have a reduced acoustic startle reflex, altered prepulse inhibition, and characteristics of compensatory neural hyperactivity centrally. Our findings show that ISL1 is one of the obligatory factors required to sculpt auditory structural and functional tonotopic maps. Still, upon Isl1 deletion, the ensuing central plasticity of the auditory pathway does not suffice to overcome developmentally induced peripheral dysfunction of the cochlea.


Assuntos
Vias Auditivas , Núcleo Coclear , Células Ciliadas Auditivas , Proteínas com Homeodomínio LIM , Neurogênese , Gânglio Espiral da Cóclea , Fatores de Transcrição , Animais , Vias Auditivas/embriologia , Cóclea/embriologia , Cóclea/inervação , Núcleo Coclear/embriologia , Células Ciliadas Auditivas/fisiologia , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/fisiologia , Camundongos , Neurogênese/genética , Gânglio Espiral da Cóclea/enzimologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
2.
Science ; 371(6525)2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33414193

RESUMO

The ability to perceive and interact with the world depends on a diverse array of neural circuits specialized for carrying out specific computations. Each circuit is assembled using a relatively limited number of molecules and common developmental steps, from cell fate specification to activity-dependent synaptic refinement. Given this shared toolkit, how do individual circuits acquire their characteristic properties? We explore this question by comparing development of the circuitry for seeing and hearing, highlighting a few examples where differences in each system's sensory demands necessitate different developmental strategies.


Assuntos
Vias Auditivas/embriologia , Núcleo Coclear/embriologia , Neurogênese , Retina/embriologia , Vias Visuais/embriologia , Animais , Audição/fisiologia , Camundongos , Células Receptoras Sensoriais/ultraestrutura , Sinapses/ultraestrutura , Visão Ocular/fisiologia
3.
Int J Mol Sci ; 22(1)2020 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-33374462

RESUMO

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


Assuntos
Células Ciliadas Auditivas/fisiologia , Neurônios/fisiologia , Animais , Sequência de Bases , Tronco Encefálico , Cóclea/embriologia , Cóclea/fisiologia , Núcleo Coclear/embriologia , Núcleo Coclear/fisiologia , Orelha Interna/embriologia , Orelha Interna/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Mutação , Neurogênese , Medicina Regenerativa/métodos , Gânglio Espiral da Cóclea/embriologia , Gânglio Espiral da Cóclea/fisiologia
4.
Hear Res ; 382: 107784, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31522073

RESUMO

Morphological studies in developing brain determine critical periods of proliferation, neurogenesis, gliogenesis, and apoptosis. During these periods both intrinsic and extrinsic pathological factors can hamper development. These time points are not available for the human cochlear nucleus (CN). We have used design-based stereology and determined that 18-22 weeks of gestation (WG) are critical in the development of the human CN. Twenty-three fetuses and seven postnatal brainstems were processed for cresyl violet (CV) staining and immunoexpression of NeuN (neurons), GFAP (astrocytes), Ki-67 (proliferation) and TUNEL (apoptosis) and 3-D reconstruction. The volume of CN, total number of neurons selected profiles and the volume of neurons and their nuclei were estimated. Data were grouped (G) into: G1:18-20 WG, G2: 21-24 WG, G3: 25-28 WG and G4 >29 WG. The dimensions of morphologically identified neurons were also measured. The CN primordium was first identifiable at 10WG. Definitive DCN (Dorsal cochlear nucleus) and VCN (ventral cochlear nucleus) were identifiable at 16 WG. There was a sudden growth spurt in total volume of CN, number of neurons and astrocytes from 18 WG. We also observed an increase in proliferation and apoptosis after 22 WG. The number of neurons identifiable by CV was significantly lower than that by NeuN-immunostaining till 25 WG (p = 0.020), after which, both methods were equivalent. Eight morphological types of neurons were identifiable by 26 WG and could be resolved into four clusters by volume and diameter. The CN changed orientation from small, flat and horizontal at 10-16 WG to larger and oblique from 18WG onwards. Prevention of exposure to noxious factors at 18-22 WG may be important in preventing congenital deafness.


Assuntos
Astrócitos , Núcleo Coclear/crescimento & desenvolvimento , Neurônios , Fatores Etários , Antígenos Nucleares/análise , Apoptose , Astrócitos/química , Benzoxazinas/química , Proliferação de Células , Pré-Escolar , Núcleo Coclear/química , Núcleo Coclear/embriologia , Corantes/química , Idade Gestacional , Proteína Glial Fibrilar Ácida/análise , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Lactente , Recém-Nascido , Antígeno Ki-67/análise , Proteínas do Tecido Nervoso/análise , Neurogênese , Neurônios/química , Coloração e Rotulagem
5.
Elife ; 72018 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-30566077

RESUMO

Vestibular function was established early in vertebrates and has remained, for the most part, unchanged. In contrast, each group of tetrapods underwent independent evolutionary processes to solve the problem of hearing on land, resulting in a remarkable mixture of conserved, divergent and convergent features that define extant auditory systems. The vestibuloacoustic nuclei of the hindbrain develop from a highly conserved ground plan and provide an ideal framework on which to address the participation of developmental processes to the evolution of neuronal circuits. We employed an electroporation strategy to unravel the contribution of two dorsoventral and four axial lineages to the development of the chick hindbrain vestibular and auditory nuclei. We compare the chick developmental map with recently established genetic fate-maps of the developing mouse hindbrain. Overall, we find considerable conservation of developmental origin for the vestibular nuclei. In contrast, a comparative analysis of the developmental origin of hindbrain auditory structures echoes the complex evolutionary history of the auditory system. In particular, we find that the developmental origin of the chick auditory interaural time difference circuit supports its emergence from an ancient vestibular network, unrelated to the analogous mammalian counterpart.


Assuntos
Tronco Encefálico/embriologia , Núcleo Coclear/embriologia , Núcleos Vestibulares/embriologia , Vestíbulo do Labirinto/embriologia , Animais , Vias Auditivas/citologia , Vias Auditivas/embriologia , Vias Auditivas/metabolismo , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Embrião de Galinha , Galinhas , Núcleo Coclear/citologia , Núcleo Coclear/metabolismo , Eletroporação , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Rombencéfalo/citologia , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Especificidade da Espécie , Núcleos Vestibulares/citologia , Núcleos Vestibulares/metabolismo , Vestíbulo do Labirinto/citologia , Vestíbulo do Labirinto/metabolismo
6.
J Neurosci ; 38(29): 6445-6460, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-29950504

RESUMO

A global loss of the fragile X mental retardation protein (FMRP; encoded by the Fmr1 gene) leads to sensory dysfunction and intellectual disabilities. One underlying mechanism of these phenotypes is structural and functional deficits in synapses. Here, we determined the autonomous function of postsynaptic FMRP in circuit formation, synaptogenesis, and synaptic maturation. In normal cochlea nucleus, presynaptic auditory axons form large axosomatic endbulb synapses on cell bodies of postsynaptic bushy neurons. In ovo electroporation of drug-inducible Fmr1-shRNA constructs produced a mosaicism of FMRP expression in chicken (either sex) bushy neurons, leading to reduced FMRP levels in transfected, but not neighboring nontransfected, neurons. Structural analyses revealed that postsynaptic FMRP reduction led to smaller size and abnormal morphology of individual presynaptic endbulbs at both early and later developmental stages. We further examined whether FMRP reduction affects dendritic development, as a potential mechanism underlying defective endbulb formation. Normally, chicken bushy neurons grow extensive dendrites at early stages and retract these dendrites when endbulbs begin to form. Neurons transfected with Fmr1 shRNA exhibited a remarkable delay in branch retraction, failing to provide necessary somatic surface for timely formation and growth of large endbulbs. Patch-clamp recording verified functional consequences of dendritic and synaptic deficits on neurotransmission, showing smaller amplitudes and slower kinetics of spontaneous and evoked EPSCs. Together, these data demonstrate that proper levels of postsynaptic FMRP are required for timely maturation of somatodendritic morphology, a delay of which may affect synaptogenesis and thus contribute to long-lasting deficits of excitatory synapses.SIGNIFICANCE STATEMENT Fragile X mental retardation protein (FMRP) regulates a large variety of neuronal activities. A global loss of FMRP affects neural circuit development and synaptic function, leading to fragile X syndrome (FXS). Using temporally and spatially controlled genetic manipulations, this study provides the first in vivo report that autonomous FMRP regulates multiple stages of dendritic development, and that selective reduction of postsynaptic FMRP leads to abnormal development of excitatory presynaptic terminals and compromised neurotransmission. These observations demonstrate secondary influence of developmentally transient deficits in neuronal morphology and connectivity to the development of long-lasting synaptic pathology in FXS.


Assuntos
Núcleo Coclear/embriologia , Núcleo Coclear/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurogênese/fisiologia , Sinapses/fisiologia , Animais , Embrião de Galinha , Feminino , Masculino , Neurônios/fisiologia , Transmissão Sináptica/fisiologia
7.
J Neurosci ; 38(12): 2967-2980, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29439165

RESUMO

Tonotopic differentiation is fundamental for signal processing in the auditory system. However, when and how this differentiation arises remain elusive. We addressed this issue using electrophysiology and immunohistochemistry in nucleus magnocellularis of chickens of both sexes, which is known to differ in the expression of Kv1.1 channels depending on characteristic frequency (CF). Just after hearing onset (embryonic day 12-14), Kv1 current gradually increased to a slightly larger extent in neurons with higher CF, causing a tonotopic difference of Kv1 current before hatch. However, after hatch, a much larger increase of Kv1 current occurred, particularly in higher-CF neurons, due to an augmentation of Kv1.1 expression at the plasma membrane. This later change in expression led to the large tonotopic difference of Kv1 current characteristic of mature animals. Attenuation of auditory input by inducing conductive or sensorineural hearing loss around hatch suppressed the differentiation in a level-dependent manner. Moreover, elevation of auditory input during embryonic periods could not reproduce the differentiation, suggesting that the capacity of neurons to drive Kv1.1 expression via auditory input develops in a cell-specific manner, thus underlying the frequency-specific expression of the channel within the nucleus. The results indicated that the tonotopic differentiation of Kv1.1 in nucleus magnocellularis is partially determined before hatch, but largely driven by afferent input after hatch. Our results highlight the importance of neuronal capacity for sound to drive ion channel expression as well as the level of auditory experience in the frequency tuning of brainstem auditory circuits.SIGNIFICANCE STATEMENT Tuning-frequency-specific expression of ion channels is a prerequisite for auditory system function, but its underlying mechanisms remain unclear. Here, we revealed in avian cochlear nucleus that the expression of Kv1.1 became more dependent on auditory input at a late period of maturation in neurons tuned to higher-frequency sound, leading to frequency-specific Kv1.1 expression. Attenuation of auditory input during this period suppressed the differentiation in a level-dependent manner, whereas elevation of input in earlier periods could not reproduce the differentiation. Thus, the capacity of neurons to drive Kv1.1 expression via auditory input develops in a cell-specific manner and directs differentiation, highlighting the importance of neuronal character as well as the level of input in the frequency tuning of auditory circuits.


Assuntos
Percepção Auditiva/fisiologia , Núcleo Coclear/metabolismo , Canal de Potássio Kv1.1/biossíntese , Neurogênese/fisiologia , Estimulação Acústica , Animais , Vias Auditivas/metabolismo , Embrião de Galinha , Galinhas , Núcleo Coclear/embriologia , Núcleo Coclear/crescimento & desenvolvimento , Feminino , Audição/fisiologia , Masculino
8.
J Chem Neuroanat ; 93: 38-47, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-28341180

RESUMO

Auditory impulses perceived by the hair cells of the organ of corti are relayed in the cochlear nucleus, the first relay station in the brainstem, by the cochlear nerve. The human foetus is well known to respond to sound during the last trimester of gestation. On the contrary, studies conducted in rat, cat and mouse have shown that these mammals have an immature auditory system at the time of birth. There are very few reports available regarding the morphological and functional maturation of the cochlear nucleus in human. Although the human cochlear nucleus neurons attain adult morphological characters by mid-gestation, there are hardly any studies discussing the functional maturation of the cochlear nucleus. Hence the present study was aimed at observing the morphological as well as functional maturation of the human foetal cochlear nuclei at various gestational ages. Morphological maturation was observed qualitatively while stereological estimation of the volume of well defined ventral cochlear nucleus (VCN) was calculated by the Cavalieri principle; neuronal count and density was estimated by dissector principle. The functional maturation was assessed by observing the expression of synaptophysin, a synaptic marker, at different gestational ages and by the presence of parvalbumin, a calcium binding functional neuronal marker by immunohistochemistry. Neurons showed coarse Nissl's substance and well developed cell processes and gradual increase in cell size by the 24th-30th gestational week. Synaptophysin labeling in the complete cochlear nucleus was observed at 20 weeks of gestation. Adult pattern of synaptophysin labeling was observed finally at37weeks of gestation. Earliest presence of parvalbumin expression was detected at 16 weeks of gestation and a distinct adult pattern was seen at 37 weeks of gestation. This study concluded that morphological and functional maturation of the human cochlear nuclei occurs simultaneously during mid-gestation which represents the critical period of development and continues up to term.


Assuntos
Núcleo Coclear/anatomia & histologia , Núcleo Coclear/embriologia , Adulto , Vias Auditivas , Contagem de Células , Tamanho Celular , Núcleo Coclear/metabolismo , Feminino , Idade Gestacional , Humanos , Imageamento Tridimensional , Imuno-Histoquímica , Masculino , Neurônios/ultraestrutura , Corpos de Nissl/ultraestrutura , Parvalbuminas/metabolismo , Gravidez , Sinaptofisina/metabolismo
9.
eNeuro ; 4(2)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28413822

RESUMO

Topography in the avian cochlear nucleus magnocellularis (NM) is represented as gradually increasing characteristic frequency (CF) along the caudolateral-to-rostromedial axis. In this study, we characterized the organization and cell biophysics of the caudolateral NM (NMc) in chickens (Gallus gallus). Examination of cellular and dendritic architecture first revealed that NMc contains small neurons and extensive dendritic processes, in contrast to adendritic, large neurons located more rostromedially. Individual dye-filling study further demonstrated that NMc is divided into two subregions, with NMc2 neurons having larger and more complex dendritic fields than NMc1. Axonal tract tracing studies confirmed that NMc1 and NMc2 neurons receive afferent inputs from the auditory nerve and the superior olivary nucleus, similar to the adendritic NM. However, the auditory axons synapse with NMc neurons via small bouton-like terminals, unlike the large end bulb synapses on adendritic NM neurons. Immunocytochemistry demonstrated that most NMc2 neurons express cholecystokinin but not calretinin, distinct from NMc1 and adendritic NM neurons that are cholecystokinin negative and mostly calretinin positive. Finally, whole-cell current clamp recordings revealed that NMc neurons require significantly lower threshold current for action potential generation than adendritic NM neurons. Moreover, in contrast to adendritic NM neurons that generate a single-onset action potential, NMc neurons generate multiple action potentials to suprathreshold sustained depolarization. Taken together, our data indicate that NMc contains multiple neuron types that are structurally, connectively, molecularly, and physiologically different from traditionally defined NM neurons, emphasizing specialized neural properties for processing low-frequency sounds.


Assuntos
Vias Auditivas/fisiologia , Núcleo Coclear/citologia , Neurônios/citologia , Neurônios/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Animais Recém-Nascidos , Calbindina 2/metabolismo , Embrião de Galinha , Galinhas , Colecistocinina/metabolismo , Núcleo Coclear/embriologia , Núcleo Coclear/crescimento & desenvolvimento , Dendritos/fisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Antagonistas GABAérgicos/farmacologia , Imageamento Tridimensional , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Picrotoxina/farmacologia
10.
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
11.
Brain Behav Evol ; 88(3-4): 161-176, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27866201

RESUMO

The neurons in the mammalian and avian auditory hindbrain nuclei share a number of significant morphological and physiological properties for fast, secure and precise neurotransmission, such as giant synapses, voltage-gated K+ channels and fast AMPA receptors. Based on the independent evolution of the middle ear in these two vertebrate lineages, on different embryonic origins of the nuclei and on marked differences on the circuit level, these similarities are assumed to reflect convergent evolution. Independent acquisition of similar phenotypes can be produced by divergent evolution of genetic mechanisms or by similar molecular mechanisms. The distinction between these two possibilities requires knowledge of the gene regulatory networks (GRNs) that orchestrate the development of auditory hindbrain structures. We therefore compared the expression pattern of GRN components, both transcription factors (TFs) and noncoding RNA, during terminal differentiation of the auditory hindbrain structures in mouse and chicken when neurons acquire their final morphological and electrophysiological properties. In general, we observed broad expression of these genes in the mouse auditory cochlear nucleus complex and the superior olivary complex at both postnatal day 4 (P4) and at P25, and for the chicken at the equivalent developmental stages, i.e. embryonic day 13 (E13) and at P14-P17. Our data are in agreement with a model based on similar molecular mechanisms underlying terminal differentiation and maintenance of neuronal cell identity in the auditory hindbrain of different vertebrate lineages. This conservation might reflect developmental constraints arising from the tagmatic organization of rhombomeres and the evolutionarily highly conserved GRNs operating in these structures.


Assuntos
Vias Auditivas , Evolução Biológica , Galinhas/genética , Núcleo Coclear , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Camundongos/genética , Rombencéfalo , Complexo Olivar Superior , Animais , Vias Auditivas/embriologia , Vias Auditivas/metabolismo , Embrião de Galinha , Núcleo Coclear/embriologia , Núcleo Coclear/metabolismo , Feminino , Masculino , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Complexo Olivar Superior/embriologia , Complexo Olivar Superior/metabolismo
12.
J Neurosci ; 36(32): 8500-15, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27511020

RESUMO

UNLABELLED: In the auditory system, sounds are processed in parallel frequency-tuned circuits, beginning in the cochlea. Auditory nerve fibers reflect this tonotopy and encode temporal properties of acoustic stimuli by "locking" discharges to a particular stimulus phase. However, physiological constraints on phase-locking depend on stimulus frequency. Interestingly, low characteristic frequency (LCF) neurons in the cochlear nucleus improve phase-locking precision relative to their auditory nerve inputs. This is proposed to arise through synaptic integration, but the postsynaptic membrane's selectivity for varying levels of synaptic convergence is poorly understood. The chick cochlear nucleus, nucleus magnocellularis (NM), exhibits tonotopic distribution of both input and membrane properties. LCF neurons receive many small inputs and have low input thresholds, whereas high characteristic frequency (HCF) neurons receive few, large synapses and require larger currents to spike. NM therefore presents an opportunity to study how small membrane variations interact with a systematic topographic gradient of synaptic inputs. We investigated membrane input selectivity and observed that HCF neurons preferentially select faster input than their LCF counterparts, and that this preference is tolerant of changes to membrane voltage. We then used computational models to probe which properties are crucial to phase-locking. The model predicted that the optimal arrangement of synaptic and membrane properties for phase-locking is specific to stimulus frequency and that the tonotopic distribution of input number and membrane excitability in NM closely tracks a stimulus-defined optimum. These findings were then confirmed physiologically with dynamic-clamp simulations of inputs to NM neurons. SIGNIFICANCE STATEMENT: One way that neurons represent temporal information is by phase-locking, which is discharging in response to a particular phase of the stimulus waveform. In the auditory system, central neurons are optimized to retain or improve phase-locking precision compared with input from the auditory nerve. However, the difficulty of this computation varies systematically with stimulus frequency. We examined properties that contribute to temporal processing both physiologically and in a computational model. Neurons processing low-frequency input benefit from integration of many weak inputs, whereas those processing higher frequencies progressively lose precision by integration of multiple inputs. Here, we reveal general features of input-output optimization that apply to all neurons that process time varying input.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Coclear/citologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Modelos Neurológicos , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Vias Auditivas/fisiologia , Embrião de Galinha , Núcleo Coclear/embriologia , Núcleo Coclear/crescimento & desenvolvimento , Simulação por Computador , Estimulação Elétrica , Técnicas In Vitro , Técnicas de Patch-Clamp
13.
Dev Biol ; 414(2): 149-60, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27151208

RESUMO

Auditory information is initially processed in the cochlear nuclei before being relayed to the brain. The cochlear nuclei are subdivided into dorsal, anterior ventral, and posterior ventral domains, each containing several subtypes of neurons that are thought to play discrete roles in the processing of sound. However, the ontogeny of these neurons is poorly understood, and this gap in knowledge hampers efforts to understand the basic neural circuitry of this nucleus. Here, we reveal that Bhlhb5 is expressed in both excitatory (unipolar brush cells) and inhibitory neurons (cartwheel cells) of the DCN during development. To gain genetic access to Bhlhb5-expressing neurons in the DCN, we generated a Bhlhb5::flpo knockin allele. Using an intersectional genetic strategy, we labeled cartwheel cells, thereby providing proof of concept that subpopulations of Bhlhb5-expressing neurons can be genetically targeted. Moreover, fate-mapping experiments using this allele revealed that Bhlhb5 is required for the proper development of the DCN, since mice lacking Bhlhb5 showed a dramatically diminished number of neurons, including unipolar brush and cartwheel cells. Intriguingly, the Bhlhb5::flpo allele also genetically labels numerous other regions of the nervous system that process sensory input, including the dorsal horn, the retina, and the nucleus of the lateral olfactory tract, hinting at a more general role for Bhlhb5 in the development of neurons that mediate sensory integration.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Núcleo Coclear/crescimento & desenvolvimento , Células Receptoras Sensoriais/metabolismo , Alelos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Contagem de Células , Linhagem da Célula , Núcleo Coclear/embriologia , Núcleo Coclear/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Proteínas Luminescentes/análise , Camundongos , Camundongos Knockout , Bulbo Olfatório/metabolismo , Fator de Transcrição PAX6/metabolismo , Células do Corno Posterior/metabolismo , Retina/metabolismo
14.
Neural Dev ; 10: 27, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26666565

RESUMO

BACKGROUND: In the auditory brainstem, ventral cochlear nucleus (VCN) axons project to the contralateral, but not ipsilateral, medial nucleus of trapezoid body (MNTB), terminating in the calyx of Held. Dorsal VCN neurons, representing high frequencies, synapse with medial MNTB neurons, while low frequency-coding ventral VCN neurons synapse with lateral MNTB neurons, reflecting tonotopic organization. The mechanisms that ensure strictly contralateral targeting and topographic ordering are incompletely understood. Here we examined the roles of ephrin-A signaling in both types of targeting. RESULTS: Ephrin-A2 and ephrin-A5 are expressed in VCN cells during late embryonic and early postnatal development. At these ages ephrin-A2 is expressed in axons surrounding MNTB and ephrin-A5 is expressed in MNTB principal neurons. Ephrin-A2/A5 double knockout mice displayed axon targeting errors in which VCN axons project to MNTB on both sides of the brainstem, where they terminate in calyceal endings. Ephrin-A2 and ephrin-A5 single knockout mice showed a similar phenotype. In contrast to effects on contralateral targeting, ephrin-A2/A5 double knockout mice showed no defects in formation of tonotopically ordered projections from VCN to MNTB. CONCLUSIONS: These findings demonstrate that distinct mechanisms regulate targeting of VCN axons to the contralateral MNTB and targeting to appropriate tonotopic locations. Ephrin-A signaling plays a similar role to ephrin-B signaling in the VCN-MNTB pathway, where both classes normally prevent formation of calyceal projections to ipsilateral MNTB. These classes may rely in part on common signaling pathways.


Assuntos
Axônios/fisiologia , Padronização Corporal/fisiologia , Núcleo Coclear/embriologia , Efrina-A2/metabolismo , Efrina-A5/metabolismo , Neurogênese/fisiologia , Animais , Vias Auditivas/citologia , Núcleo Coclear/citologia , Imunofluorescência , Lateralidade Funcional , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Eur J Neurosci ; 41(11): 1416-29, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25903469

RESUMO

In the avian nucleus magnocellularis (NM) endbulb of Held giant synapses develop from temporary bouton terminals. The molecular regulation of this process is not well understood. Furthermore, it is unknown how the postsynaptic specialization of the endbulb synapses develops. We therefore analysed expression of the postsynaptic scaffold protein PSD-95 during the transition from bouton-to-endbulb synapses. PSD-95 has been implicated in the regulation of the strength of glutamatergic synapses and could accordingly be of functional relevance for giant synapse formation. PSD-95 protein was expressed at synaptic sites in embryonic chicken auditory brainstem and upregulated between embryonic days (E)12 and E16. We applied immunofluorescence staining and confocal microscopy to quantify pre-and postsynaptic protein signals during bouton-to-endbulb transition. Giant terminal formation progressed along the tonotopic axis in NM, but was absent in low-frequency NM. We found a tonotopic gradient of postsynaptic PSD-95 signals in NM. Furthermore, PSD-95 immunosignals showed the greatest increase between E12 and E15, temporally preceding the bouton-to-endbulb transition. We then applied whole-cell electrophysiology to measure synaptic currents elicited by synaptic terminals during bouton-to-endbulb transition. With progressing endbulb formation postsynaptic currents rose more rapidly and synapses were less susceptible to short-term depression, but currents were not different in amplitude or decay-time constant. We conclude that development of presynaptic specializations follows postsynaptic development and speculate that the early PSD-95 increase could play a functional role in endbulb formation.


Assuntos
Proteínas Aviárias/metabolismo , Núcleo Coclear/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Terminações Pré-Sinápticas/fisiologia , Animais , Galinhas , Núcleo Coclear/embriologia , Núcleo Coclear/metabolismo , Potenciais Pós-Sinápticos Excitadores , Terminações Pré-Sinápticas/metabolismo
16.
Cell Mol Life Sci ; 72(3): 519-535, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25332098

RESUMO

Development and evolution of auditory hindbrain nuclei are two major unsolved issues in hearing research. Recent characterization of transgenic mice identified the rhombomeric origins of mammalian auditory nuclei and unraveled genes involved in their formation. Here, we provide an overview on these data by assembling them into rhombomere-specific gene regulatory networks (GRNs), as they underlie developmental and evolutionary processes. To explore evolutionary mechanisms, we compare the GRNs operating in the mammalian auditory hindbrain with data available from the inner ear and other vertebrate groups. Finally, we propose that the availability of genomic sequences from all major vertebrate taxa and novel genetic techniques for non-model organisms provide an unprecedented opportunity to investigate development and evolution of the auditory hindbrain by comparative molecular approaches. The dissection of the molecular mechanisms leading to auditory structures will also provide an important framework for auditory processing disorders, a clinical problem difficult to tackle so far. These data will, therefore, foster basic and clinical hearing research alike.


Assuntos
Percepção Auditiva/fisiologia , Evolução Biológica , Núcleo Coclear/embriologia , Redes Reguladoras de Genes/fisiologia , Audição/fisiologia , Colículos Inferiores/embriologia , Complexo Olivar Superior/embriologia , Animais , Núcleo Coclear/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Colículos Inferiores/metabolismo , Camundongos , Modelos Biológicos , Especificidade da Espécie , Complexo Olivar Superior/metabolismo , Tretinoína/metabolismo
17.
J Neurosci ; 34(9): 3443-53, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24573300

RESUMO

The axon initial segment (AIS) is the site of spike initiation in neurons. Previous studies revealed that spatial distribution of the AIS varies greatly among neurons to meet their specific needs. However, when and how this differentiation arises is unknown. Neurons in the avian nucleus laminaris (NL) are binaural coincidence detectors for sound localization and show differentiation in the distribution of the AIS, with shorter length and a more distal position from the soma with an increase in tuning frequency. We studied these characteristics of the AIS in NL neurons of the chicken during development and found that the AIS differentiates in its distribution after initial formation, and this is driven by activity-dependent and activity-independent mechanisms that differentially regulate distal and proximal boundaries of the AIS. Before hearing onset, the ankyrinG-positive AIS existed at a wide stretch of proximal axon regardless of tuning frequency, but Na+ channels were only partially distributed within the AIS. Shortly after hearing onset, Na+ channels accumulated along the entire AIS, which started shortening and relocating distally to a larger extent in neurons with higher tuning frequencies. Ablation of inner ears abolished the shortening of the AIS without affecting the position of its proximal boundary, indicating that both distal and proximal AIS boundaries are disassembled during development, and the former is dependent on afferent activity. Thus, interaction of these activity-dependent and activity-independent mechanisms determines the cell-specific distribution of the AIS in NL neurons and plays a critical role in establishing the function of sound localization circuit.


Assuntos
Vias Auditivas , Axônios/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/citologia , Potenciais de Ação/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Anquirinas/metabolismo , Vias Auditivas/embriologia , Vias Auditivas/crescimento & desenvolvimento , Vias Auditivas/metabolismo , Embrião de Galinha , Galinhas , Núcleo Coclear/citologia , Núcleo Coclear/embriologia , Núcleo Coclear/crescimento & desenvolvimento , Simulação por Computador , Feminino , Técnicas In Vitro , Masculino , Modelos Neurológicos , Glicoproteína Associada a Mielina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Fosfopiruvato Hidratase/metabolismo
18.
J Comp Neurol ; 520(6): 1262-77, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22020566

RESUMO

Nucleus laminaris (NL) neurons in the avian auditory brainstem are coincidence detectors necessary for the computation of interaural time differences used in sound localization. In addition to their excitatory inputs from nucleus magnocellularis, NL neurons receive inhibitory inputs from the superior olivary nucleus (SON) that greatly improve coincidence detection in mature animals. The mechanisms that establish mature distributions of inhibitory inputs to NL are not known. We used the vesicular GABA transporter (VGAT) as a marker for inhibitory presynaptic terminals to study the development of inhibitory inputs to NL between embryonic day 9 (E9) and E17. VGAT immunofluorescent puncta were first seen sparsely in NL at E9. The density of VGAT puncta increased with development, first within the ventral NL neuropil region and subsequently throughout both the ventral and dorsal dendritic neuropil, with significantly fewer terminals in the cell body region. A large increase in density occurred between E13­15 and E16­17, at a developmental stage when astrocytes that express glial fibrillary acidic protein (GFAP) become mature. We cultured E13 brainstem slices together with astrocyte-conditioned medium (ACM) obtained from E16 brainstems and found that ACM, but not control medium, increased the density of VGAT puncta. This increase was similar to that observed during normal development. Astrocyte-secreted factors interact with the terminal ends of SON axons to increase the number of GABAergic terminals. These data suggest that factors secreted from GFAP-positive astrocytes promote maturation of inhibitory pathways in the auditory brainstem.


Assuntos
Astrócitos/metabolismo , Tronco Encefálico/embriologia , Núcleo Coclear/embriologia , Fatores de Crescimento Neural/metabolismo , Inibição Neural/fisiologia , Terminações Pré-Sinápticas/metabolismo , Animais , Astrócitos/citologia , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Embrião de Galinha , Galinhas , Núcleo Coclear/citologia , Núcleo Coclear/metabolismo , Técnicas de Cultura de Órgãos/métodos , Transmissão Sináptica/fisiologia
19.
Dev Neurobiol ; 72(9): 1243-55, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22021100

RESUMO

Precision in auditory brainstem connectivity underlies sound localization. Cochlear activity is transmitted to the ventral cochlear nucleus (VCN) in the mammalian brainstem via the auditory nerve. VCN globular bushy cells project to the contralateral medial nucleus of the trapezoid body (MNTB), where specialized axons terminals, the calyces of Held, encapsulate MNTB principal neurons. The VCN-MNTB pathway is an essential component of the circuitry used to compute interaural intensity differences that are used for localizing sounds. When input from one ear is removed during early postnatal development, auditory brainstem circuitry displays robust anatomical plasticity. The molecular mechanisms that control the development of auditory brainstem circuitry and the developmental plasticity of these pathways are poorly understood. In this study we examined the role of EphB signaling in the development of the VCN-MNTB projection and in the reorganization of this pathway after unilateral deafferentation. We found that EphB2 and EphB3 reverse signaling are critical for the normal development of the projection from VCN to MNTB, but that successful circuit assembly most likely relies upon the coordinated function of many EphB proteins. We have also found that ephrin-B reverse signaling repels induced projections to the ipsilateral MNTB after unilateral deafferentation, suggesting that similar mechanisms regulate these two processes.


Assuntos
Vias Auditivas/embriologia , Vias Auditivas/crescimento & desenvolvimento , Tronco Encefálico/embriologia , Tronco Encefálico/crescimento & desenvolvimento , Receptor EphB2/fisiologia , Receptor EphB3/fisiologia , Vias Aferentes/fisiologia , Animais , Vias Auditivas/citologia , Tronco Encefálico/citologia , Núcleo Coclear/citologia , Núcleo Coclear/embriologia , Núcleo Coclear/crescimento & desenvolvimento , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Neurogênese/fisiologia , Receptor EphB2/deficiência , Receptor EphB2/genética , Receptor EphB3/deficiência , Receptor EphB3/genética , Transdução de Sinais/fisiologia
20.
Hear Res ; 276(1-2): 70-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21093567

RESUMO

Sound localization requires precise and specialized neural circuitry. A prominent and well-studied specialization is found in the mammalian auditory brainstem. Globular bushy cells of the ventral cochlear nucleus (VCN) project contralaterally to neurons of the medial nucleus of the trapezoid body (MNTB), where their large axons terminate on cell bodies of MNTB principal neurons, forming the calyces of Held. The VCN-MNTB pathway is necessary for the accurate computation of interaural intensity and time differences; MNTB neurons provide inhibitory input to the lateral superior olive, which compares levels of excitation from the ipsilateral ear to levels of tonotopically matched inhibition from the contralateral ear, and to the medial superior olive, where precise inhibition from MNTB neurons tunes the delays of binaural excitation. Here we review the morphological and physiological aspects of the development of the VCN-MNTB pathway and its calyceal termination, along with potential mechanisms that give rise to its precision. During embryonic development, VCN axons grow towards the midline, cross the midline into the region of the presumptive MNTB and then form collateral branches that will terminate in calyces of Held. In rodents, immature calyces of Held appear in MNTB during the first few days of postnatal life. These calyces mature morphologically and physiologically over the next three postnatal weeks, enabling fast, high fidelity transmission in the VCN-MNTB pathway.


Assuntos
Vias Auditivas/crescimento & desenvolvimento , Nervo Coclear/crescimento & desenvolvimento , Núcleo Coclear/crescimento & desenvolvimento , Ponte/crescimento & desenvolvimento , Envelhecimento/fisiologia , Animais , Vias Auditivas/embriologia , Vias Auditivas/fisiologia , Axônios/fisiologia , Nervo Coclear/embriologia , Nervo Coclear/fisiologia , Núcleo Coclear/embriologia , Núcleo Coclear/fisiologia , Desenvolvimento Embrionário , Humanos , Ponte/embriologia , Ponte/fisiologia , Transmissão Sináptica , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...