Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 18: 1414484, 2024.
Article in English | MEDLINE | ID: mdl-38962512

ABSTRACT

Acetylcholine (ACh) is a prevalent neurotransmitter throughout the nervous system. In the brain, ACh is widely regarded as a potent neuromodulator. In neurons, ACh signals are conferred through a variety of receptors that influence a broad range of neurophysiological phenomena such as transmitter release or membrane excitability. In sensory circuitry, ACh modifies neural responses to stimuli and coordinates the activity of neurons across multiple levels of processing. These factors enable individual neurons or entire circuits to rapidly adapt to the dynamics of complex sensory stimuli, underscoring an essential role for ACh in sensory processing. In the auditory system, histological evidence shows that acetylcholine receptors (AChRs) are expressed at virtually every level of the ascending auditory pathway. Despite its apparent ubiquity in auditory circuitry, investigation of the roles of this cholinergic network has been mainly focused on the inner ear or forebrain structures, while less attention has been directed at regions between the cochlear nuclei and midbrain. In this review, we highlight what is known about cholinergic function throughout the auditory system from the ear to the cortex, but with a particular emphasis on brainstem and midbrain auditory centers. We will focus on receptor expression, mechanisms of modulation, and the functional implications of ACh for sound processing, with the broad goal of providing an overview of a newly emerging view of impactful cholinergic modulation throughout the auditory pathway.

2.
J Neurosci ; 44(8)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383485

ABSTRACT

The medial nucleus of the trapezoid body (MNTB) has been intensively investigated as a primary source of inhibition in brainstem auditory circuitry. MNTB-derived inhibition plays a critical role in the computation of sound location, as temporal features of sounds are precisely conveyed through the calyx of Held/MNTB synapse. In adult gerbils, cholinergic signaling influences sound-evoked responses of MNTB neurons via nicotinic acetylcholine receptors (nAChRs; Zhang et al., 2021) establishing a modulatory role for cholinergic input to this nucleus. However, the cellular mechanisms through which acetylcholine (ACh) mediates this modulation in the MNTB remain obscure. To investigate these mechanisms, we used whole-cell current and voltage-clamp recordings to examine cholinergic physiology in MNTB neurons from Mongolian gerbils (Meriones unguiculatus) of both sexes. Membrane excitability was assessed in brain slices, in pre-hearing (postnatal days 9-13) and post-hearing onset (P18-20) MNTB neurons during bath application of agonists and antagonists of nicotinic (nAChRs) and muscarinic receptors (mAChRs). Muscarinic activation induced a potent increase in excitability most prominently prior to hearing onset with nAChR modulation emerging at later time points. Pharmacological manipulations further demonstrated that the voltage-gated K+ channel KCNQ (Kv7) is the downstream effector of mAChR activation that impacts excitability early in development. Cholinergic modulation of Kv7 reduces outward K+ conductance and depolarizes resting membrane potential. Immunolabeling revealed expression of Kv7 channels as well as mAChRs containing M1 and M3 subunits. Together, our results suggest that mAChR modulation is prominent but transient in the developing MNTB and that cholinergic modulation functions to shape auditory circuit development.


Subject(s)
Receptors, Nicotinic , Trapezoid Body , Animals , Female , Male , Trapezoid Body/physiology , Gerbillinae , Synaptic Transmission/physiology , Neurons/physiology , Receptors, Nicotinic/metabolism , Cholinergic Agents , Auditory Pathways/physiology
4.
Front Neural Circuits ; 15: 715369, 2021.
Article in English | MEDLINE | ID: mdl-34335196

ABSTRACT

The superior olivary complex (SOC) is a major computation center in the brainstem auditory system. Despite previous reports of high expression levels of cholinergic receptors in the SOC, few studies have addressed the functional role of acetylcholine in the region. The source of the cholinergic innervation is unknown for all but one of the nuclei of the SOC, limiting our understanding of cholinergic modulation. The medial nucleus of the trapezoid body, a key inhibitory link in monaural and binaural circuits, receives cholinergic input from other SOC nuclei and also from the pontomesencephalic tegmentum. Here, we investigate whether these same regions are sources of cholinergic input to other SOC nuclei. We also investigate whether individual cholinergic cells can send collateral projections bilaterally (i.e., into both SOCs), as has been shown at other levels of the subcortical auditory system. We injected retrograde tract tracers into the SOC in gerbils, then identified retrogradely-labeled cells that were also immunolabeled for choline acetyltransferase, a marker for cholinergic cells. We found that both the SOC and the pontomesencephalic tegmentum (PMT) send cholinergic projections into the SOC, and these projections appear to innervate all major SOC nuclei. We also observed a small cholinergic projection into the SOC from the lateral paragigantocellular nucleus of the reticular formation. These various sources likely serve different functions; e.g., the PMT has been associated with things such as arousal and sensory gating whereas the SOC may provide feedback more closely tuned to specific auditory stimuli. Further, individual cholinergic neurons in each of these regions can send branching projections into both SOCs. Such projections present an opportunity for cholinergic modulation to be coordinated across the auditory brainstem.


Subject(s)
Acoustic Stimulation/methods , Auditory Pathways/physiology , Cholinergic Neurons/physiology , Superior Olivary Complex/physiology , Animals , Auditory Pathways/chemistry , Auditory Pathways/enzymology , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/chemistry , Cholinergic Neurons/enzymology , Female , Gerbillinae , Male , Olivary Nucleus/chemistry , Olivary Nucleus/enzymology , Olivary Nucleus/physiology , Superior Olivary Complex/chemistry , Superior Olivary Complex/enzymology
5.
J Neurosci ; 41(4): 674-688, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33268542

ABSTRACT

The medial nucleus of trapezoid body (MNTB) is a major source of inhibition in auditory brainstem circuitry. The MNTB projects well-timed inhibitory output to principal sound-localization nuclei in the superior olive (SOC) as well as other computationally important centers. Acoustic information is conveyed to MNTB neurons through a single calyx of Held excitatory synapse arising from the cochlear nucleus. The encoding efficacy of this large synapse depends on its activity rate, which is primarily determined by sound intensity and stimulus frequency. However, MNTB activity rate is additionally influenced by inhibition and possibly neuromodulatory inputs, albeit their functional role is unclear. Happe and Morley (2004) discovered prominent expression of α7 nAChRs in rat SOC, suggesting possible engagement of ACh-mediated modulation of neural activity in the MNTB. However, the existence and nature of this putative modulation have never been physiologically demonstrated. We probed nicotinic cholinergic influences on acoustic responses of MNTB neurons from adult gerbils (Meriones unguiculatus) of either sex. We recorded tone-evoked MNTB single-neuron activity in vivo using extracellular single-unit recording. Piggyback multibarrel electrodes enabled pharmacological manipulation of nAChRs by reversibly applying antagonists to two receptor types, α7 and α4ß2. We observed that tone-evoked responses are dependent on ACh modulation by both nAChR subtypes. Spontaneous activity was not affected by antagonist application. Functionally, we demonstrate that ACh contributes to sustaining high discharge rates and enhances signal encoding efficacy. Additionally, we report anatomic evidence revealing novel cholinergic projections to MNTB arising from pontine and superior olivary nuclei.SIGNIFICANCE STATEMENT This study is the first to physiologically probe how acetylcholine, a pervasive neuromodulator in the brain, influences the encoding of acoustic information by the medial nucleus of trapezoid body, the most prominent source of inhibition in brainstem sound-localization circuitry. We demonstrate that this cholinergic input enhances neural discrimination of tones from noise stimuli, which may contribute to processing important acoustic signals, such as speech. Additionally, we describe novel anatomic projections providing cholinergic input to the MNTB. Together, these findings shed new light on the contribution of neuromodulation to fundamental computational processes in auditory brainstem circuitry and to a more holistic understanding of modulatory influences in sensory processing.


Subject(s)
Acoustic Stimulation , Parasympathetic Nervous System/physiology , Trapezoid Body/physiology , Acetylcholine/physiology , Animals , Auditory Pathways/physiology , Female , Gerbillinae , Male , Neurons/physiology , Olivary Nucleus/physiology , Pons/physiology , Receptors, Nicotinic/physiology , Sound , alpha7 Nicotinic Acetylcholine Receptor/physiology
6.
J Neurophysiol ; 116(6): 2676-2688, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27655966

ABSTRACT

In mammals with good low-frequency hearing, the medial superior olive (MSO) computes sound location by comparing differences in the arrival time of a sound at each ear, called interaural time disparities (ITDs). Low-frequency sounds are not reflected by the head, and therefore level differences and spectral cues are minimal or absent, leaving ITDs as the only cue for sound localization. Although mammals with high-frequency hearing and small heads (e.g., bats, mice) barely experience ITDs, the MSO is still present in these animals. Yet, aside from studies in specialized bats, in which the MSO appears to serve functions other than ITD processing, it has not been studied in small mammals that do not hear low frequencies. Here we describe neurons in the mouse brain stem that share prominent anatomical, morphological, and physiological properties with the MSO in species known to use ITDs for sound localization. However, these neurons also deviate in some important aspects from the typical MSO, including a less refined arrangement of cell bodies, dendrites, and synaptic inputs. In vitro, the vast majority of neurons exhibited a single, onset action potential in response to suprathreshold depolarization. This spiking pattern is typical of MSO neurons in other species and is generated from a complement of Kv1, Kv3, and IH currents. In vivo, mouse MSO neurons show bilateral excitatory and inhibitory tuning as well as an improvement in temporal acuity of spiking during bilateral acoustic stimulation. The combination of classical MSO features like those observed in gerbils with more unique features similar to those observed in bats and opossums make the mouse MSO an interesting model for exploiting genetic tools to test hypotheses about the molecular mechanisms and evolution of ITD processing.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Superior Olivary Complex/cytology , Superior Olivary Complex/metabolism , Acoustic Stimulation , Animals , Animals, Newborn , Auditory Pathways/physiology , Choline O-Acetyltransferase/metabolism , Electric Stimulation , Glycine Plasma Membrane Transport Proteins/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Models, Neurological , Neurons/metabolism , Patch-Clamp Techniques , Phosphopyruvate Hydratase/metabolism , Psychoacoustics , Stilbamidines/pharmacokinetics , Vesicular Glutamate Transport Protein 1/metabolism
7.
J Neurosci ; 36(32): 8500-15, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27511020

ABSTRACT

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.


Subject(s)
Action Potentials/physiology , Cochlear Nucleus/cytology , Excitatory Postsynaptic Potentials/physiology , Models, Neurological , Neurons/physiology , Synaptic Transmission/physiology , Analysis of Variance , Animals , Animals, Newborn , Auditory Pathways/physiology , Chick Embryo , Cochlear Nucleus/embryology , Cochlear Nucleus/growth & development , Computer Simulation , Electric Stimulation , In Vitro Techniques , Patch-Clamp Techniques
8.
Article in English | MEDLINE | ID: mdl-24672432

ABSTRACT

For all neurons, a proper balance of synaptic excitation and inhibition is crucial to effect computational precision. Achievement of this balance is remarkable when one considers factors that modulate synaptic strength operate on multiple overlapping time scales and affect both pre- and postsynaptic elements. Recent studies have shown that inhibitory transmitters, glycine and GABA, are co-released in auditory nuclei involved in the computation of interaural time disparities (ITDs), a cue used to process sound source location. The co-release expressed at these synapses is heavily activity dependent, and generally occurs when input rates are high. This circuitry, in both birds and mammals, relies on inhibitory input to maintain the temporal precision necessary for ITD encoding. Studies of co-release in other brain regions suggest that GABA and glycine receptors (GlyRs) interact via cross-suppressive modulation of receptor conductance. We performed in vitro whole-cell recordings in several nuclei of the chicken brainstem auditory circuit to assess whether this cross-suppressive phenomenon was evident in the avian brainstem. We evaluated the effect of pressure-puff applied glycine on synaptically evoked inhibitory currents in nucleus magnocellularis (NM) and the superior olivary nucleus (SON). Glycine pre-application reduced the amplitude of inhibitory postsynaptic currents (IPSCs) evoked during a 100 Hz train stimulus in both nuclei. This apparent glycinergic modulation was blocked in the presence of strychnine. Further experiments showed that this modulation did not depend on postsynaptic biochemical interactions such as phosphatase activity, or direct interactions between GABA and GlyR proteins. Rather, voltage clamp experiments in which we manipulated Cl(-) flux during agonist application suggest that activation of one receptor will modulate the conductance of the other via local changes in Cl(-) ion concentration within microdomains of the postsynaptic membrane.


Subject(s)
Auditory Pathways/drug effects , Glycine/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Neural Inhibition/drug effects , Neurons/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Chickens , Female , Male , Olivary Nucleus/drug effects , Patch-Clamp Techniques , Receptors, Glycine/metabolism , Synaptic Transmission/drug effects
9.
J Neurosci ; 34(4): 1314-24, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24453322

ABSTRACT

In the auditory system, sounds are processed in parallel frequency-tuned circuits, beginning in the cochlea. Activity of auditory nerve fibers reflects this frequency-specific topographic pattern, known as tonotopy, and imparts frequency tuning onto their postsynaptic target neurons in the cochlear nucleus. In birds, cochlear nucleus magnocellularis (NM) neurons encode the temporal properties of acoustic stimuli by "locking" discharges to a particular phase of the input signal. Physiological specializations exist in gradients corresponding to the tonotopic axis in NM that reflect the characteristic frequency (CF) of their auditory nerve fiber inputs. One feature of NM neurons that has not been investigated across the tonotopic axis is short-term synaptic plasticity. NM offers a rather homogeneous population of neurons with a distinct topographical distribution of synaptic properties that is ideal for the investigation of specialized synaptic plasticity. Here we demonstrate for the first time that short-term synaptic depression (STD) is expressed topographically, where unitary high CF synapses are more robust with repeated stimulation. Correspondingly, high CF synapses drive spiking more reliably than their low CF counterparts. We show that postsynaptic AMPA receptor desensitization does not contribute to the observed difference in STD. Further, rate of recovery from depression, a presynaptic property, does not differ tonotopically. Rather, we show that another presynaptic feature, readily releasable pool (RRP) size, is tonotopically distributed and inversely correlated with vesicle release probability. Mathematical model results demonstrate that these properties of vesicle dynamics are sufficient to explain the observed tonotopic distribution of STD.


Subject(s)
Auditory Perception/physiology , Cochlear Nucleus/physiology , Long-Term Synaptic Depression/physiology , Models, Neurological , Synaptic Transmission/physiology , Animals , Chick Embryo , Chickens , Excitatory Postsynaptic Potentials , Female , Male , Models, Theoretical , Patch-Clamp Techniques
10.
J Neurophysiol ; 111(3): 565-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24198323

ABSTRACT

Localization of low-frequency acoustic stimuli is processed in dedicated neural pathways where coincidence-detecting neurons compare the arrival time of sound stimuli at the two ears, or interaural time disparity (ITD). ITDs occur in the submillisecond range, and vertebrates have evolved specialized excitatory and inhibitory circuitry to compute these differences. Glycinergic inhibition is a computationally significant and prominent component of the mammalian ITD pathway. However, evidence for glycinergic transmission is limited in birds, where GABAergic inhibition has been thought to be the dominant or exclusive inhibitory transmitter. Indeed, previous work showed that GABA antagonists completely eliminate inhibition in avian nuclei specialized for processing temporal features of sound, nucleus magnocellularis (NM) and nucleus laminaris (NL). However, more recent work shows that glycine is coexpressed with GABA in synaptic terminals apposed to neurons in both nuclei (Coleman WL, Fischl MJ, Weimann SR, Burger RM. J Neurophysiol 105: 2405-2420, 2011; Kuo SP, Bradley LA, Trussell LO. J Neurosci 29: 9625-9634, 2009). Here we show complementary evidence of functional glycine receptor (GlyR) expression in NM and NL. Additionally, we show that glycinergic input can be evoked under particular stimulus conditions. Stimulation at high but physiologically relevant rates evokes a slowly emerging glycinergic response in NM and NL that builds over the course of the stimulus. Glycinergic response magnitude was stimulus rate dependent, representing 18% and 7% of the total inhibitory current in NM and NL, respectively, at the end of the 50-pulse, 200-Hz stimulus. Finally, we show that the glycinergic component is functionally relevant, as its elimination reduced inhibition of discharges evoked by current injection into NM neurons.


Subject(s)
Auditory Pathways/metabolism , Basal Nucleus of Meynert/metabolism , Glycine/metabolism , Inhibitory Postsynaptic Potentials , Receptors, Glycine/metabolism , Sound Localization , Animals , Auditory Pathways/physiology , Basal Nucleus of Meynert/physiology , Chickens , Evoked Potentials, Auditory , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Receptors, Glycine/genetics
11.
Front Neural Circuits ; 8: 145, 2014.
Article in English | MEDLINE | ID: mdl-25565972

ABSTRACT

Spherical bushy cells (SBCs) in the anteroventral cochlear nucleus respond to acoustic stimulation with discharges that precisely encode the phase of low-frequency sound. The accuracy of spiking is crucial for sound localization and speech perception. Compared to the auditory nerve input, temporal precision of SBC spiking is improved through the engagement of acoustically evoked inhibition. Recently, the inhibition was shown to be less precise than previously understood. It shifts from predominantly glycinergic to synergistic GABA/glycine transmission in an activity-dependent manner. Concurrently, the inhibition attains a tonic character through temporal summation. The present study provides a comprehensive understanding of the mechanisms underlying this slow inhibitory input. We performed whole-cell voltage clamp recordings on SBCs from juvenile Mongolian gerbils and recorded evoked inhibitory postsynaptic currents (IPSCs) at physiological rates. The data reveal activity-dependent IPSC kinetics, i.e., the decay is slowed with increased input rates or recruitment. Lowering the release probability yielded faster decay kinetics of the single- and short train-IPSCs at 100 Hz, suggesting that transmitter quantity plays an important role in controlling the decay. Slow transmitter clearance from the synaptic cleft caused prolonged receptor binding and, in the case of glycine, spillover to nearby synapses. The GABAergic component prolonged the decay by contributing to the asynchronous vesicle release depending on the input rate. Hence, the different factors controlling the amount of transmitters in the synapse jointly slow the inhibition during physiologically relevant activity. Taken together, the slow time course is predominantly determined by the receptor kinetics and transmitter clearance during short stimuli, whereas long duration or high frequency stimulation additionally engage asynchronous release to prolong IPSCs.


Subject(s)
Cochlear Nucleus/physiology , Inhibitory Postsynaptic Potentials/physiology , Synaptic Transmission/physiology , Animals , Cochlear Nucleus/drug effects , Electric Stimulation/methods , Gerbillinae , Glycine/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Patch-Clamp Techniques , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/drug effects , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
13.
J Physiol ; 590(13): 3047-66, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22473782

ABSTRACT

Interaural time disparities (ITDs) are the primary cues for localisation of low-frequency sound stimuli. ITDs are computed by coincidence-detecting neurones in the medial superior olive (MSO) in mammals. Several previous studies suggest that control of synaptic gain is essential for maintaining ITD selectivity as stimulus intensity increases. Using acute brain slices from postnatal day 7 to 24 (P7­P24) Mongolian gerbils, we confirm that activation of GABAB receptors reduces the amplitude of excitatory and inhibitory synaptic currents to the MSO and, moreover, show that the decay kinetics of IPSCs are slowed in mature animals. During repetitive stimuli, activation of GABAB receptors reduced the amount of depression observed, while PSC suppression and the slowed kinetics were maintained. Additionally, we used physiological and modelling approaches to test the potential impact of GABAB activation on ITD encoding in MSO neurones. Current clamp recordings from MSO neurones were made while pharmacologically isolated excitatory inputs were bilaterally stimulated using pulse trains that simulate ITDs in vitro. MSO neurones showed strong selectivity for bilateral delays. Application of both GABAB agonists and antagonists demonstrate that GABAB modulation of synaptic input can sharpen ITD selectivity. We confirmed and extended these results in a computational model that allowed for independent manipulation of each GABAB-dependent effect. Modelling suggests that modulation of both amplitude and kinetics of synaptic inputs by GABAB receptors can improve precision of ITD computation. Our studies suggest that in vivo modulation of synaptic input by GABAB receptors may act to preserve ITD selectivity across various stimulus conditions.


Subject(s)
Olivary Nucleus/physiology , Receptors, GABA-B/physiology , Sound Localization/physiology , Acoustic Stimulation , Animals , Excitatory Postsynaptic Potentials , Gerbillinae , In Vitro Techniques , Inhibitory Postsynaptic Potentials , Models, Biological
14.
PLoS One ; 7(2): e31364, 2012.
Article in English | MEDLINE | ID: mdl-22347467

ABSTRACT

Connexins (Cx) are the subunits of gap junctions, membraneous protein channels that permit the exchange of small molecules between adjacent cells. Cx43 is required for cell proliferation in the zebrafish caudal fin. Previously, we found that a Cx43-like connexin, cx40.8, is co-expressed with cx43 in the population of proliferating cells during fin regeneration. Here we demonstrate that Cx40.8 exhibits novel differential subcellular localization in vivo, depending on the growth status of the fin. During fin ontogeny, Cx40.8 is found at the plasma membrane, but Cx40.8 is retained in the Golgi apparatus during regeneration. We next identified a 30 amino acid domain of Cx40.8 responsible for its dynamic localization. One possible explanation for the differential localization is that Cx40.8 contributes to the regulation of Cx43 in vivo, perhaps modifying channel activity during ontogenetic growth. However, we find that the voltage-gating properties of Cx40.8 are similar to Cx43. Together our findings reveal that Cx40.8 exhibits differential subcellular localization in vivo, dependent on a discrete domain in its carboxy terminus. We suggest that the dynamic localization of Cx40.8 differentially influences Cx43-dependent cell proliferation during ontogeny and regeneration.


Subject(s)
Animal Fins/chemistry , Animal Fins/physiology , Connexins/metabolism , Regeneration , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Proliferation , Connexin 43 , Gap Junction alpha-5 Protein
15.
J Comp Neurol ; 520(7): 1493-508, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22102107

ABSTRACT

Topographic maps are salient features of neuronal organization in sensory systems. Inhibitory components of neuronal circuitry are often embedded within this organization, making them difficult to isolate experimentally. The auditory system provides opportunities to study the topographic organization of inhibitory long-range projection nuclei, such as the superior olivary nucleus (SON). We analyzed the topographic organization of response features of neurons in the SON of chickens. Quantitative methods were developed to assess and communicate this organization. These analyses led to three main conclusions: 1) sound frequency is linearly arranged from dorsal (low frequencies) to ventral (high frequencies) in SON; 2) this tonotopic organization is less precise than the organization of the excitatory nuclei in the chicken auditory brainstem; and 3) neurons with different response patterns to pure tone stimuli are interspersed throughout the SON and show similar tonotopic organizations. This work provides a predictive model to determine the optimal stimulus frequency for a neuron from its spatial location in the SON.


Subject(s)
Auditory Perception/physiology , Brain Mapping , Neurons/physiology , Olivary Nucleus/physiology , Acoustic Stimulation , Animals , Auditory Pathways/physiology , Chickens , Electrophysiology , Image Processing, Computer-Assisted , Imaging, Three-Dimensional
16.
J Neurophysiol ; 106(1): 4-14, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525367

ABSTRACT

Interaural time differences (ITDs) are the primary cue animals, including humans, use to localize low-frequency sounds. In vertebrate auditory systems, dedicated ITD processing neural circuitry performs an exacting task, the discrimination of microsecond differences in stimulus arrival time at the two ears by coincidence-detecting neurons. These neurons modulate responses over their entire dynamic range to sounds differing in ITD by mere hundreds of microseconds. The well-understood function of this circuitry in birds has provided a fruitful system to investigate how inhibition contributes to neural computation at the synaptic, cellular, and systems level. Our recent studies in the chicken have made significant progress in bringing together many of these findings to provide a cohesive picture of inhibitory function.


Subject(s)
Auditory Pathways/physiology , Birds/physiology , Feedback, Physiological/physiology , Sound Localization/physiology , Animals , Brain Stem/anatomy & histology , Brain Stem/physiology , Humans , Neurons/physiology
17.
J Neurosci ; 30(36): 12075-83, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826670

ABSTRACT

GABAergic modulation of activity in avian cochlear nucleus neurons has been studied extensively in vitro. However, how this modulation actually influences processing in vivo is not known. We investigated responses of chicken nucleus magnocellularis (NM) neurons to sound while pharmacologically manipulating the inhibitory input from the superior olivary nucleus (SON). SON receives excitatory inputs from nucleus angularis (NA) and nucleus laminaris (NL), and provides GABAergic inputs to NM, NA, NL, and putatively to the contralateral SON. Results from single-unit extracellular recordings from 2 to 4 weeks posthatch chickens show that firing rates of auditory nerve fibers increased monotonically with sound intensity, while that of NM neurons saturated or even decreased at moderate or loud sound levels. Blocking GABAergic input with local application of TTX into the SON induced an increase in firing rate of ipsilateral NM, while that of the contralateral NM decreased at high sound levels. Moreover, local application of bicuculline to NM also increased the firing rate of NM neurons at high sound levels, reduced phase locking, and broadened the frequency-tuning properties of NM neurons. Following application of DNQX, clear evidence of inhibition was observed. Furthermore, the inhibition was tuned to a broader frequency range than the excitatory response areas. We conclude that GABAergic inhibition from SON has at least three physiological influences on the activity of NM neurons: it regulates the firing activity of NM units in a sound-level-dependent manner; it improves phase selectivity; and it sharpens frequency tuning of NM neuronal responses.


Subject(s)
Action Potentials/physiology , Cochlear Nucleus/cytology , Neural Inhibition/physiology , Neurons/cytology , gamma-Aminobutyric Acid/metabolism , Acoustic Stimulation/methods , Action Potentials/drug effects , Analysis of Variance , Animals , Animals, Newborn , Bicuculline/pharmacology , Chickens/anatomy & histology , Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality , GABA Antagonists/pharmacology , Neural Inhibition/drug effects , Neurons/classification , Neurons/drug effects , Psychoacoustics , Quinoxalines/pharmacology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Time Factors
18.
J Neurosci ; 27(8): 2112-23, 2007 Feb 21.
Article in English | MEDLINE | ID: mdl-17314306

ABSTRACT

Mature nucleus magnocellularis (NM) neurons, the avian homolog of bushy cells of the mammalian anteroventral cochlear nucleus, maintain high [Cl-]i and depolarize in response to GABA. Depolarizing GABAergic postsynaptic potentials (GPSPs) activate both the synaptic conductance and large outward currents, which, when coupled together, inhibit spikes via shunting and spike threshold accommodation. We studied the maturation of the synaptic and voltage-dependent components of inhibition in embryonic NM neurons using whole-cell and gramicidin-perforated patch-clamp techniques to measure Cl- reversal potential, GABAergic synaptic responses, and voltage-dependent outward currents. We found that GABA enhanced excitability in immature NM neurons, undergoing a switch to inhibitory between embryonic day 14 (E14) and E18. Low-voltage-activated Kv1-type (dendrotoxin-I sensitive) K+ currents increased in amplitude between E14 and E18, whereas Cl- reversal potential and synaptic conductances remained relatively stable during this period. GABA was rendered inhibitory because of this increase in low-voltage activated outward currents. GPSPs summed with other inputs to increase spike probability at E14. GPSPs shunted spikes at E18, but blocking Kv1 channels transformed this inhibition to excitation, similar to E14 neurons. Subthreshold depolarizing current steps, designed to activate outward currents similar to depolarizing GPSPs, enhanced excitability at E14 but inhibited spiking in E18 neurons. Blocking Kv1 channels reversed this effect, rendering current steps excitatory. We present the novel finding that the developmental transition of GABAergic processing from increasing neuronal excitability to inhibiting spiking can depend on changes in the expression of voltage-gated channels rather than on a change in Cl- reversal potential.


Subject(s)
Brain Stem/embryology , Embryonic Development/physiology , Neural Inhibition/physiology , Shaker Superfamily of Potassium Channels/physiology , gamma-Aminobutyric Acid/physiology , Action Potentials , Animals , Chick Embryo , Electric Conductivity , Electric Stimulation , Embryo, Mammalian/physiology , Embryo, Nonmammalian , Evoked Potentials , In Vitro Techniques , Membrane Potentials , Neurons/physiology , Patch-Clamp Techniques , Receptors, GABA-A/physiology , Synaptic Transmission
19.
J Comp Neurol ; 489(1): 11-22, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15977167

ABSTRACT

Nucleus magnocellularis (NM), nucleus angularis (NA), and nucleus laminaris (NL), second- and third-order auditory neurons in the avian brainstem, receive GABAergic input primarily from the superior olivary nucleus (SON). Previous studies have demonstrated that both GABA(A) and GABA(B) receptors (GABA(B)Rs) influence physiological properties of NM neurons. We characterized the distribution of GABA(B)R expression in these nuclei during development and after deafferentation of the excitatory auditory nerve (nVIII) inputs. We used a polyclonal antibody raised against rat GABA(B)Rs in the auditory brainstem during developmental periods that are thought to precede and include synaptogenesis of GABAergic inputs. As early as embryonic day (E)14, dense labeling is observed in NA, NM, NL, and SON. At earlier ages immunoreactivity is present in somas as diffuse staining with few puncta. By E21, when the structure and function of the auditory nuclei are known to be mature, GABA(B) immunoreactivity is characterized by dense punctate labeling in NM, NL, and a subset of NA neurons, but label is sparse in the SON. Removal of the cochlea and nVIII neurons in posthatch chicks resulted in only a small decrease in immunoreactivity after survival times of 14 or 28 days, suggesting that a major proportion of GABA(B)Rs may be expressed postsynaptically or on GABAergic terminals. We confirmed this interpretation with immunogold TEM, where expression at postsynaptic membrane sites is clearly observed. The characterization of GABA(B)R distribution enriches our understanding of the full complement of inhibitory influences on central auditory processing in this well-studied neuronal circuit.


Subject(s)
Brain Stem/metabolism , Chickens/physiology , Neurons, Afferent/physiology , Receptors, GABA-B/biosynthesis , Animals , Basal Nucleus of Meynert/cytology , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/ultrastructure , Blotting, Western , Brain Stem/growth & development , Brain Stem/ultrastructure , Chick Embryo , Cochlear Nerve/growth & development , Cochlear Nerve/metabolism , Cochlear Nerve/ultrastructure , Denervation , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Immunoelectron , Tissue Embedding
20.
J Neurophysiol ; 93(3): 1429-38, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15483063

ABSTRACT

Neurons of nucleus magnocellularis (NM), a division of avian cochlear nucleus that performs precise temporal encoding, receive glutamatergic excitatory input solely from the eighth nerve and GABAergic inhibitory input primarily from the ipsilateral superior olivary nucleus. GABA activates both ligand-gated Cl- channels [GABA(A) receptors (GABA(A)Rs)] and G protein-coupled receptors (GABA(B) receptors). The net effect of GABA(A)R-mediated input to NM is inhibitory, although depolarizing. Several studies have shown that this shunting, inhibitory GABAergic input can evoke action potentials in postsynaptic NM neurons, which could interfere with their temporal encoding. While this GABA-mediated firing is limited by a low-voltage-activated K+ conductance, we have found evidence for a second mechanism. We investigated modulation of GABA(A)R-mediated responses by GABA(B)Rs using whole cell recording techniques. Bath-applied baclofen, a GABA(B)R agonist, produced dose-dependent suppression of evoked inhibitory postsynaptic currents (eIPSCs). This suppression was blocked by CGP52432, a potent and selective GABA(B)R antagonist. Baclofen reduced the frequency but not the amplitude of miniature IPSCs (mIPSCs) and did not affect postsynaptic currents elicited by puff application of a specific GABA(A)R agonist muscimol, suggesting a presynaptic mechanism for the GABA(B)R-mediated modulation. Firing of NM neurons by synaptic stimulation of GABAergic inputs to NM was eliminated by baclofen. However, endogenous GABA(B)R activity in the presynaptic inhibitory terminals was not observed. We propose that presynaptic GABA(B)Rs function as autoreceptors, regulating synaptic strength of GABA(A)R-mediated inhibition, and prevent NM neurons from generating firing during activation of the inhibitory inputs.


Subject(s)
Cochlear Nucleus/cytology , Neural Inhibition/physiology , Neurons/physiology , Receptors, GABA-A/metabolism , Receptors, GABA-B/physiology , Analysis of Variance , Animals , Baclofen/pharmacology , Benzylamines/pharmacology , Calcium/pharmacology , Chick Embryo , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , GABA-B Receptor Agonists , GABA-B Receptor Antagonists , In Vitro Techniques , Magnesium/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Neural Inhibition/drug effects , Neural Inhibition/radiation effects , Neurons/drug effects , Neurons/radiation effects , Patch-Clamp Techniques/methods , Phosphinic Acids/pharmacology , Reaction Time/drug effects , Reaction Time/radiation effects , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...