Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
J Physiol ; 594(13): 3683-703, 2016 07 01.
Article En | MEDLINE | ID: mdl-27104476

KEY POINTS: Lateral superior olive (LSO) principal neurons receive AMPA receptor (AMPAR) - and NMDA receptor (NMDAR)-mediated EPSCs and glycinergic IPSCs. Both EPSCs and IPSCs have slow kinetics in prehearing animals, which during developmental maturation accelerate to sub-millisecond decay time-constants. This correlates with a change in glutamate and glycine receptor subunit composition quantified via mRNA levels. The NMDAR-EPSCs accelerate over development to achieve decay time-constants of 2.5 ms. This is the fastest NMDAR-mediated EPSC reported. Acoustic trauma (AT, loud sounds) slow AMPAR-EPSC decay times, increasing GluA1 and decreasing GluA4 mRNA. Modelling of interaural intensity difference suggests that the increased EPSC duration after AT shifts interaural level difference to the right and compensates for hearing loss. Two months after AT the EPSC decay times recovered to control values. Synaptic transmission in the LSO matures by postnatal day 20, with EPSCs and IPSCs having fast kinetics. AT changes the AMPAR subunits expressed and slows the EPSC time-course at synapses in the central auditory system. ABSTRACT: Damaging levels of sound (acoustic trauma, AT) diminish peripheral synapses, but what is the impact on the central auditory pathway? Developmental maturation of synaptic function and hearing were characterized in the mouse lateral superior olive (LSO) from postnatal day 7 (P7) to P96 using voltage-clamp and auditory brainstem responses. IPSCs and EPSCs show rapid acceleration during development, so that decay kinetics converge to similar sub-millisecond time-constants (τ, 0.87 ± 0.11 and 0.77 ± 0.08 ms, respectively) in adult mice. This correlated with LSO mRNA levels for glycinergic and glutamatergic ionotropic receptor subunits, confirming a switch from Glyα2 to Glyα1 for IPSCs and increased expression of GluA3 and GluA4 subunits for EPSCs. The NMDA receptor (NMDAR)-EPSC decay τ accelerated from >40 ms in prehearing animals to 2.6 ± 0.4 ms in adults, as GluN2C expression increased. In vivo induction of AT at around P20 disrupted IPSC and EPSC integration in the LSO, so that 1 week later the AMPA receptor (AMPAR)-EPSC decay was slowed and mRNA for GluA1 increased while GluA4 decreased. In contrast, GlyR IPSC and NMDAR-EPSC decay times were unchanged. Computational modelling confirmed that matched IPSC and EPSC kinetics are required to generate mature interaural level difference functions, and that longer-lasting EPSCs compensate to maintain binaural function with raised auditory thresholds after AT. We conclude that LSO excitatory and inhibitory synaptic drive matures to identical time-courses, that AT changes synaptic AMPARs by expression of subunits with slow kinetics (which recover over 2 months) and that loud sounds reversibly modify excitatory synapses in the brain, changing synaptic function for several weeks after exposure.


Acoustic Stimulation , Brain Stem/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Receptors, AMPA/physiology , Animals , Excitatory Postsynaptic Potentials , Female , Inhibitory Postsynaptic Potentials , Male , Mice, Inbred CBA , Protein Subunits/physiology
2.
J Physiol ; 593(7): 1685-700, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25605440

Hyperpolarization-activated non-specific cation-permeable channels (HCN) mediate I(H) currents, which are modulated by cGMP and cAMP and by nitric oxide (NO) signalling. Channel properties depend upon subunit composition (HCN1-4 and accessory subunits) as demonstrated in expression systems, but physiological relevance requires investigation in native neurons with intact intracellular signalling. Here we use the superior olivary complex (SOC), which exhibits a distinctive pattern of HCN1 and HCN2 expression, to investigate NO modulation of the respective I(H) currents, and compare properties in wild-type and HCN1 knockout mice. The medial nucleus of the trapezoid body (MNTB) expresses HCN2 subunits exclusively, and sends inhibitory projections to the medial and lateral superior olives (MSO, LSO) and the superior paraolivary nucleus (SPN). In contrast to the MNTB, these target nuclei possess an I(H) with fast kinetics, and they express HCN1 subunits. NO is generated in the SOC following synaptic activity and here we show that NO selectively suppresses HCN1, while enhancing IH mediated by HCN2 subunits. NO hyperpolarizes the half-activation of HCN1-mediated currents and slows the kinetics of native IH currents in the MSO, LSO and SPN. This modulation was independent of cGMP and absent in transgenic mice lacking HCN1. Independently, NO signalling depolarizes the half-activation of HCN2-mediated I(H) currents in a cGMP-dependent manner. Thus, NO selectively suppresses fast HCN1-mediated I(H) and facilitates a slow HCN2-mediated I(H) , so generating a spectrum of modulation, dependent on the local expression of HCN1 and/or HCN2.


Brain Stem/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Nitric Oxide/pharmacology , Potassium Channels/physiology , Animals , Brain Stem/metabolism , Female , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , In Vitro Techniques , Male , Membrane Potentials , Mice, Inbred CBA , Mice, Knockout , Neurons/metabolism , Nitric Oxide Synthase Type I/metabolism , Potassium Channels/genetics
3.
Hear Res ; 321: 45-51, 2015 Mar.
Article En | MEDLINE | ID: mdl-25602577

Kv1.1 subunits of low voltage-activated (Kv) potassium channels are encoded by the Kcna1 gene and crucially determine the synaptic integration window to control the number and temporal precision of action potentials in the auditory brainstem of mammals and birds. Prior electrophysiological studies showed that auditory signaling is compromised in monaural as well as in binaural neurons of the auditory brainstem in Kv1.1 knockout mice (Kcna1(-/-)). Here we examine the behavioral effects of Kcna1 deletion on sensory tasks dependent on either binaural processing (detecting the movement of a sound source across the azimuth), monaural processing (detecting a gap in noise), as well as binaural summation of the acoustic startle reflex (ASR). Hearing thresholds measured by auditory brainstem responses (ABR) do not differ between genotypes, but our data show a much stronger performance of wild type mice (+/+) in each test during binaural hearing which was lost by temporarily inducing a unilateral hearing loss (through short term blocking of one ear) thus remarkably, leaving no significant difference between binaural and monaural hearing in Kcna1(-/-) mice. These data suggest that the behavioral effect of Kv1.1 deletion is primarily to impede binaural integration and thus to mimic monaural hearing.


Auditory Perception , Behavior, Animal , Hearing Disorders/metabolism , Hearing , Kv1.1 Potassium Channel/deficiency , Acoustic Stimulation , Animals , Auditory Pathways/physiopathology , Auditory Perception/genetics , Auditory Threshold , Evoked Potentials, Auditory, Brain Stem , Female , Genotype , Hearing/genetics , Hearing Disorders/genetics , Hearing Disorders/physiopathology , Kv1.1 Potassium Channel/genetics , Male , Mice, Inbred C3H , Mice, Knockout , Phenotype , Psychoacoustics , Reflex, Startle , Signal Detection, Psychological , Sound Localization , Time Factors
4.
Article En | MEDLINE | ID: mdl-24987336

Glycinergic inhibition plays a central role in the auditory brainstem circuitries involved in sound localization and in the encoding of temporal action potential firing patterns. Modulation of this inhibition has the potential to fine-tune information processing in these networks. Here we show that nitric oxide (NO) signaling in the auditory brainstem (where activity-dependent generation of NO is documented) modulates the strength of inhibition by changing the chloride equilibrium potential. Recent evidence demonstrates that large inhibitory postsynaptic currents (IPSCs) in neurons of the superior paraolivary nucleus (SPN) are enhanced by a very low intracellular chloride concentration, generated by the neuronal potassium chloride co-transporter (KCC2) expressed in the postsynaptic neurons. Our data show that modulation by NO caused a 15 mV depolarizing shift of the IPSC reversal potential, reducing the strength of inhibition in SPN neurons, without changing the threshold for action potential firing. Regulating inhibitory strength, through cGMP-dependent changes in the efficacy of KCC2 in the target neuron provides a postsynaptic mechanism for rapidly controlling the inhibitory drive, without altering the timing or pattern of the afferent spike train. Therefore, this NO-mediated suppression of KCC2 can modulate inhibition in one target nucleus (SPN), without influencing inhibitory strength of other target nuclei (MSO, LSO) even though they are each receiving collaterals from the same afferent nucleus (a projection from the medial nucleus of the trapezoid body, MNTB).


Cyclic GMP/metabolism , Inhibitory Postsynaptic Potentials/physiology , Medulla Oblongata/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Symporters/metabolism , Action Potentials/physiology , Animals , Auditory Pathways/physiology , Gerbillinae , Mice , K Cl- Cotransporters
5.
J Neurosci ; 33(38): 15044-9, 2013 Sep 18.
Article En | MEDLINE | ID: mdl-24048834

The medial nucleus of the trapezoid body (MNTB) in the superior olivary complex (SOC) is an inhibitory hub considered critical for binaural sound localization. We show that genetic ablation of MNTB neurons in mice only subtly affects this ability by prolonging the minimum time required to detect shifts in sound location. Furthermore, glycinergic innervation of the SOC is maintained without an MNTB, consistent with the existence of parallel inhibitory inputs. These findings redefine the role of MNTB in sound localization and suggest that the inhibitory network is more complex than previously thought.


Glycine/metabolism , Neural Inhibition/physiology , Olivary Nucleus/cytology , Olivary Nucleus/physiology , Sound Localization/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Acoustic Stimulation , Animals , Animals, Newborn , Auditory Pathways/physiology , Early Growth Response Protein 2/genetics , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality , Glycine Plasma Membrane Transport Proteins/metabolism , Homeodomain Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/genetics , Patch-Clamp Techniques , Sound Localization/drug effects , Strychnine/pharmacology , Valine/analogs & derivatives , Valine/pharmacology
6.
J Neurosci ; 33(21): 9113-21, 2013 May 22.
Article En | MEDLINE | ID: mdl-23699522

The central auditory brainstem provides an efferent projection known as the medial olivocochlear (MOC) system, which regulates the cochlear amplifier and mediates protection on exposure to loud sound. It arises from neurons of the ventral nucleus of the trapezoid body (VNTB), so control of neuronal excitability in this pathway has profound effects on hearing. The VNTB and the medial nucleus of the trapezoid body are the only sites of expression for the Kv2.2 voltage-gated potassium channel in the auditory brainstem, consistent with a specialized function of these channels. In the absence of unambiguous antagonists, we used recombinant and transgenic methods to examine how Kv2.2 contributes to MOC efferent function. Viral gene transfer of dominant-negative Kv2.2 in wild-type mice suppressed outward K(+) currents, increasing action potential (AP) half-width and reducing repetitive firing. Similarly, VNTB neurons from Kv2.2 knock-out mice (Kv2.2KO) also showed increased AP duration. Control experiments established that Kv2.2 was not expressed in the cochlea, so any changes in auditory function in the Kv2.2KO mouse must be of central origin. Further, in vivo recordings of auditory brainstem responses revealed that these Kv2.2KO mice were more susceptible to noise-induced hearing loss. We conclude that Kv2.2 regulates neuronal excitability in these brainstem nuclei by maintaining short APs and enhancing high-frequency firing. This safeguards efferent MOC firing during high-intensity sounds and is crucial in the mediation of protection after auditory overexposure.


Auditory Pathways/physiology , Cochlea/physiology , Hearing Loss/prevention & control , Noise/adverse effects , Olivary Nucleus/physiology , Shab Potassium Channels/physiology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Cell Line, Tumor , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hearing Loss/etiology , In Vitro Techniques , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Mutation/genetics , Neuroblastoma/pathology , Patch-Clamp Techniques , Shab Potassium Channels/deficiency , Shaw Potassium Channels/metabolism , Transfection
7.
J Neurosci ; 32(41): 14058-63, 2012 Oct 10.
Article En | MEDLINE | ID: mdl-23055473

Synaptic transmission is highly dependent on recent activity and can lead to depression or facilitation of synaptic strength. This phenomenon is called "short-term synaptic plasticity" and is shown at all synapses. While much work has been done to understand the mechanisms of short-term changes in the state of synapses, short-term plasticity is often thought of as a mechanistic consequence of the design of a synapse. This review will attempt to go beyond this view and discuss how, on one hand, complex neuronal activity affects the short-term state of synapses, but also how these dynamic changes in synaptic strength affect information processing in return.


Mental Processes/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cellular Senescence/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Humans , Time Factors
8.
Article En | MEDLINE | ID: mdl-22969707

The inferior colliculus (IC) is a midbrain nucleus that exhibits sensitivity to differences in interaural time and intensity (ITDs and IIDs) and integrates information from the auditory brainstem to provide an unambiguous representation of sound location across the azimuth. Further upstream, in the lateral superior olive (LSO), absence of low-threshold potassium currents in Kcna1(-/-) mice interfered with response onset timing and restricted IID-sensitivity to the hemifield of the excitatory ear. Assuming the IID-sensitivity in the IC to be at least partly inherited from LSO neurons, the IC IID-encoding was compared between wild-type (Kcna1(+/+)) and Kcna1(-/-) mice. We asked whether the effect observed in the Kcna1(-/-) LSO is (1) simply propagated into the IC, (2) is enhanced and amplified or, (3) alternatively, is compensated and so no longer detectable. Our results show that general IC response properties as well as the distribution of IID-functions were comparable in Kcna1(-/-) and Kcna1(+/+) mice. In agreement with the literature IC neurons exhibited a higher level-invariance of IID-sensitivity compared to LSO neurons. However, manipulating the timing between the inputs of the two ears caused significantly larger shifts of IID-sensitivity in Kcna1(-/-) mice, whereas in the wild-type IC the IID functions were stable and less sensitive to changes of the temporal relationship between the binaural inputs. We conclude that the IC not only inherits IID-sensitivity from the LSO, but that the convergence with other, non-olivary inputs in the wild-type IC acts to quality-control, consolidate, and stabilize IID representation; this necessary integration of inputs is impaired in the absence of the low-threshold potassium currents mediated by Kv1.1.

9.
PLoS One ; 7(1): e29965, 2012.
Article En | MEDLINE | ID: mdl-22253838

Multiple parallel auditory pathways ascend from the cochlear nucleus. It is generally accepted that the origin of these pathways are distinct groups of neurons differing in their anatomical and physiological properties. In extracellular in vivo recordings these neurons are typically classified on the basis of their peri-stimulus time histogram. In the present study we reconsider the question of classification of neurons in the anteroventral cochlear nucleus (AVCN) by taking a wider range of response properties into account. The study aims at a better understanding of the AVCN's functional organization and its significance as the source of different ascending auditory pathways. The analyses were based on 223 neurons recorded in the AVCN of the Mongolian gerbil. The range of analysed parameters encompassed spontaneous activity, frequency coding, sound level coding, as well as temporal coding. In order to categorize the unit sample without any presumptions as to the relevance of certain response parameters, hierarchical cluster analysis and additional principal component analysis were employed which both allow a classification on the basis of a multitude of parameters simultaneously. Even with the presently considered wider range of parameters, high number of neurons and more advanced analytical methods, no clear boundaries emerged which would separate the neurons based on their physiology. At the current resolution of the analysis, we therefore conclude that the AVCN units more likely constitute a multi-dimensional continuum with different physiological characteristics manifested at different poles. However, more complex stimuli could be useful to uncover physiological differences in future studies.


Anterior Thalamic Nuclei/cytology , Cell Differentiation , Cochlear Nucleus/cytology , Neurons/cytology , Acoustic Stimulation , Action Potentials/physiology , Animals , Cluster Analysis , Gerbillinae , Principal Component Analysis
10.
Neuron ; 71(5): 911-25, 2011 Sep 08.
Article En | MEDLINE | ID: mdl-21903083

Offset responses upon termination of a stimulus are crucial for perceptual grouping and gap detection. These gaps are key features of vocal communication, but an ionic mechanism capable of generating fast offsets from auditory stimuli has proven elusive. Offset firing arises in the brainstem superior paraolivary nucleus (SPN), which receives powerful inhibition during sound and converts this into precise action potential (AP) firing upon sound termination. Whole-cell patch recording in vitro showed that offset firing was triggered by IPSPs rather than EPSPs. We show that AP firing can emerge from inhibition through integration of large IPSPs, driven by an extremely negative chloride reversal potential (E(Cl)), combined with a large hyperpolarization-activated nonspecific cationic current (I(H)), with a secondary contribution from a T-type calcium conductance (I(TCa)). On activation by the IPSP, I(H) potently accelerates the membrane time constant, so when the sound ceases, a rapid repolarization triggers multiple offset APs that match onset timing accuracy.


Action Potentials/physiology , Neurons/physiology , Reaction Time/physiology , Acoustic Stimulation/methods , Action Potentials/drug effects , Animals , Animals, Newborn , Auditory Pathways/physiology , Biophysics , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Chlorides/metabolism , Computer Simulation , Cyclic Nucleotide-Gated Cation Channels/deficiency , Electric Stimulation , Functional Laterality , Furosemide/pharmacology , Gene Expression Regulation/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , In Vitro Techniques , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Mibefradil/pharmacology , Mice , Mice, Inbred CBA , Mice, Knockout , Models, Neurological , Neurons/drug effects , Olivary Nucleus/cytology , Patch-Clamp Techniques/methods , Potassium Channels/deficiency , Psychoacoustics , Pyrimidines/pharmacology , Reaction Time/drug effects , Reaction Time/genetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Stilbamidines/metabolism , Symporters/metabolism , Synaptic Potentials/drug effects , Synaptic Potentials/physiology , K Cl- Cotransporters
11.
Neuron ; 71(2): 291-305, 2011 Jul 28.
Article En | MEDLINE | ID: mdl-21791288

Activity-dependent changes in synaptic strength are well established as mediating long-term plasticity underlying learning and memory, but modulation of target neuron excitability could complement changes in synaptic strength and regulate network activity. It is thought that homeostatic mechanisms match intrinsic excitability to the incoming synaptic drive, but evidence for involvement of voltage-gated conductances is sparse. Here, we show that glutamatergic synaptic activity modulates target neuron excitability and switches the basis of action potential repolarization from Kv3 to Kv2 potassium channel dominance, thereby adjusting neuronal signaling between low and high activity states, respectively. This nitric oxide-mediated signaling dramatically increases Kv2 currents in both the auditory brain stem and hippocampus (>3-fold) transforming synaptic integration and information transmission but with only modest changes in action potential waveform. We conclude that nitric oxide is a homeostatic regulator, tuning neuronal excitability to the recent history of excitatory synaptic inputs over intervals of minutes to hours.


Action Potentials/physiology , Neurons/metabolism , Nitric Oxide/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Animals, Newborn , Biophysics , Brain Stem/cytology , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Hippocampus/cytology , Hydrazines/pharmacology , In Vitro Techniques , Indoles/pharmacology , Mice , Mice, Inbred CBA , Mice, Knockout , Nitric Oxide/deficiency , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Potassium Channel Blockers/pharmacology , RNA, Messenger/metabolism , Shab Potassium Channels/deficiency , Shab Potassium Channels/metabolism , Shaw Potassium Channels/deficiency , Shaw Potassium Channels/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tetraethylammonium/pharmacology , Transfection
12.
Hear Res ; 279(1-2): 22-31, 2011 Sep.
Article En | MEDLINE | ID: mdl-21397677

The aim of this review is to consider the various forms and functions of transmission across the calyx of Held/MNTB synapse and how its modulation might contribute to auditory processing. The calyx of Held synapse is the largest synapse in the mammalian brain which uses the conventional excitatory synaptic transmitter, glutamate. It is sometimes portrayed as the 'ultimate' in synaptic signalling: it is a synaptic relay in which a single axon forms one synaptic terminal onto one specific target neuron. Questions that are often raised are: "Why does such a large and secure synapse need any form of modulation? Surely it is built simply to guarantee firing an action potential in the target neuron? If this synapse is so secure, why is a synapse needed at all?" Investigating these questions explains some general limitations of transmission at synapses and provides insight into the ionic basis of neuronal function by bringing together in vivo and in vitro approaches. We will start by defining the firing behaviour of MNTB neurons in vitro (in response to synaptic stimulation or current injection) and in vivo (in response to sound) and examining the reasons for different types of firing under the two conditions. Then we will consider some of the mechanisms by which transmission can be regulated. We will finish by discussing the following hypothesis: modulation and adaptation of presynaptic and postsynaptic conductances at the calyx of Held relay synapse are aimed at maximising the security of sound onset encoding while providing secondary information on frequency spectrum, harmonic envelope and duration of sound throughout the later part of the response.


Synapses/physiology , Synaptic Transmission , Animals , Auditory Pathways , Central Nervous System/physiology , Glutamic Acid/metabolism , Hearing , Humans , Ion Channels/metabolism , Models, Biological , Neurons/metabolism , Potassium Channels, Voltage-Gated/metabolism , Receptors, AMPA/metabolism , Sound , Synapses/metabolism
13.
J Physiol ; 589(Pt 5): 1143-57, 2011 Mar 01.
Article En | MEDLINE | ID: mdl-21224222

Voltage-gated potassium (Kv) channels containing Kv1.1 subunits are strongly expressed in neurons that fire temporally precise action potentials (APs). In the auditory system, AP timing is used to localize sound sources by integrating interaural differences in time (ITD) and intensity (IID) using sound arriving at both cochleae. In mammals, the first nucleus to encode IIDs is the lateral superior olive (LSO), which integrates excitation from the ipsilateral ventral cochlear nucleus and contralateral inhibition mediated via the medial nucleus of the trapezoid body. Previously we reported that neurons in this pathway show reduced firing rates, longer latencies and increased jitter in Kv1.1 knockout (Kcna1−/−) mice. Here, we investigate whether these differences have direct impact on IID processing by LSO neurons. Single-unit recordings were made from LSO neurons of wild-type (Kcna1+/+) and from Kcna1−/− mice. IID functions were measured to evaluate genotype-specific changes in integrating excitatory and inhibitory inputs. In Kcna1+/+ mice, IID sensitivity ranged from +27 dB (excitatory ear more intense) to −20 dB (inhibitory ear more intense), thus covering the physiologically relevant range of IIDs. However, the distribution of IID functions in Kcna1−/− mice was skewed towards positive IIDs, favouring ipsilateral sound positions. Our computational model revealed that the reduced performance of IID encoding in the LSO of Kcna1−/− mice is mainly caused by a decrease in temporal fidelity along the inhibitory pathway. These results imply a fundamental role for Kv1.1 in temporal integration of excitation and inhibition during sound source localization.


Auditory Pathways/physiology , Kv1.1 Potassium Channel/metabolism , Neurons/physiology , Olivary Nucleus/physiology , Sound Localization/physiology , Acoustic Stimulation , Action Potentials/physiology , Animals , Electrophysiology , Immunohistochemistry , Kv1.1 Potassium Channel/genetics , Mice , Mice, Knockout , Models, Neurological
14.
Eur J Neurosci ; 31(9): 1574-81, 2010 May.
Article En | MEDLINE | ID: mdl-20525070

Extracellular signals from the endbulb of Held-spherical bushy cell (SBC) synapse exhibit up to three component waves ('P', 'A' and 'B'). Signals lacking the third component (B) are frequently observed but as the origin of each of the components is uncertain, interpretation of this lack of B has been controversial: is it a failure to release transmitter or a failure to generate or propagate an action potential? Our aim was to determine the origin of each component. We combined single- and multiunit in vitro methods in Mongolian gerbils and Wistar rats and used pharmacological tools to modulate glutamate receptors or voltage-gated sodium channels. Simultaneous extra- and intracellular recordings from single SBCs demonstrated a presynaptic origin of the P-component, consistent with data obtained with multielectrode array recordings of local field potentials. The later components (A and B) correspond to the excitatory postsynaptic potential (EPSP) and action potential of the SBC, respectively. These results allow a clear interpretation of in vivo extracellular signals. We conclude that action potential failures occurring at the endbulb-SBC synaptic junction largely reflect failures of the EPSP to trigger an action potential and not failures of synaptic transmission. The data provide the basis for future investigation of convergence of excitatory and inhibitory inputs in modulating transmission at a fully functional neuronal system using physiological stimulation.


Extracellular Space/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Synapses/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Brain/drug effects , Brain/physiology , Cochlea/drug effects , Cochlea/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Extracellular Space/drug effects , Gerbillinae , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microelectrodes , Neurons/drug effects , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Wistar , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Sodium Channels/metabolism , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
15.
Hear Res ; 268(1-2): 234-49, 2010 Sep 01.
Article En | MEDLINE | ID: mdl-20561574

The phase of low-frequency sinusoids is encoded in phase-coupled discharges of spherical bushy cells (SBCs) of the anteroventral cochlear nucleus and transmitted to the medial superior olive, where binaural input-coincidence is used for processing of sound source localization. SBCs are innervated by auditory nerve fibers through large, excitatory synapses (endbulbs of Held) and by inhibitory inputs, which effectively reduce SBC discharge rates. Here we monitor presynaptic potentials of endbulb-terminals and postsynaptic spikes of SBCs in extracellular single unit recordings in vivo. We compare postsynaptic phase-coupling of SBCs and their presynaptic immediate auditory nerve input. In all but one SBC discharge rates at the characteristic frequency were reduced pre-to-postsynaptically and phase-coupling accuracy was increased in one-third of them. We investigated the contribution of systemic inhibition on spike timing in SBCs by iontophoretic application of glycine- and GABA-receptor antagonists (strychnine, bicuculline). Discharge rate increased in one-third of the units during antagonist application, which was accompanied by a deterioration of phase-coupling accuracy in half of those units. These results suggest that the phase-coupling accuracy is improved in a subpopulation of SBCs during transmission from the auditory nerve to the SBCs by reduction of spike rates.


Auditory Pathways/physiology , Cochlear Nerve/physiology , Cochlear Nucleus/physiology , Gerbillinae/physiology , Sound Localization , Synaptic Transmission , Acoustic Stimulation , Animals , Auditory Pathways/drug effects , Bicuculline/administration & dosage , Cochlear Nerve/drug effects , Cochlear Nucleus/cytology , Cochlear Nucleus/drug effects , GABA-A Receptor Antagonists/administration & dosage , Glycine Agents/administration & dosage , Iontophoresis , Strychnine/administration & dosage , Synaptic Potentials , Synaptic Transmission/drug effects , Time Factors
16.
J Neurosci ; 29(30): 9510-20, 2009 Jul 29.
Article En | MEDLINE | ID: mdl-19641114

The calyx of Held synapse in the medial nucleus of the trapezoid body of the auditory brainstem has become an established in vitro model to study the development of fast glutamatergic transmission in the mammalian brain. However, we still lack in vivo data at this synapse on the maturation of spontaneous and sound-evoked discharge activity before and during the early phase of acoustically evoked signal processing (i.e., before and after hearing onset). Here we report in vivo single-unit recordings in mice from postnatal day 8 (P8) to P28 with a specific focus on developmental changes around hearing onset (P12). Data were obtained from two mouse strains commonly used in brain slice recordings: CBA/J and C57BL/6J. Spontaneous discharge rates progressively increased from P8 to P13, initially showing bursting patterns and large coefficients of variation (CVs), which changed to more continuous and random discharge activity accompanied by gradual decrease of CV around hearing onset. From P12 on, sound-evoked activity yielded phasic-tonic discharge patterns with discharge rates increasing up to P28. Response thresholds and shapes of tuning curves were adult-like by P14. A gradual shortening in response latencies was observed up to P18. The three-dimensional tonotopic organization of the medial nucleus of the trapezoid body yielded a high-to-low frequency gradient along the mediolateral and dorsoventral but not in the rostrocaudal axes. These data emphasize that models of signal transmission at the calyx of Held based on in vitro data have to take developmental changes in firing rates and response latencies up to the fourth postnatal week into account.


Evoked Potentials, Auditory, Brain Stem , Neurons/physiology , Olivary Nucleus/growth & development , Olivary Nucleus/physiology , Acoustic Stimulation , Action Potentials , Aging/physiology , Analysis of Variance , Animals , Animals, Newborn , Auditory Perception/physiology , Auditory Threshold/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Microelectrodes , Olivary Nucleus/anatomy & histology , Reaction Time , Synapses/physiology , Synaptic Transmission/physiology , Time Factors
17.
J Neurosci ; 29(25): 7991-8004, 2009 Jun 24.
Article En | MEDLINE | ID: mdl-19553439

Complexins (CPXs I-IV) presumably act as regulators of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex, but their function in the intact mammalian nervous system is not well established. Here, we explored the role of CPXs in the mouse auditory system. Hearing was impaired in CPX I knock-out mice but normal in knock-out mice for CPXs II, III, IV, and III/IV as measured by auditory brainstem responses. Complexins were not detectable in cochlear hair cells but CPX I was expressed in spiral ganglion neurons (SGNs) that give rise to the auditory nerve. Ca(2+)-dependent exocytosis of inner hair cells and sound encoding by SGNs were unaffected in CPX I knock-out mice. In the absence of CPX I, the resting release probability in the endbulb of Held synapses of the auditory nerve fibers with bushy cells in the cochlear nucleus was reduced. As predicted by computational modeling, bushy cells had decreased spike rates at sound onset as well as longer and more variable first spike latencies explaining the abnormal auditory brainstem responses. In addition, we found synaptic transmission to outlast the stimulus at many endbulb of Held synapses in vitro and in vivo, suggesting impaired synchronization of release to stimulus offset. Although sound encoding in the cochlea proceeds in the absence of complexins, CPX I is required for faithful processing of sound onset and offset in the cochlear nucleus.


Auditory Pathways/physiology , Cochlear Nucleus/metabolism , Hearing/physiology , Nerve Tissue Proteins/deficiency , Neurons/metabolism , Synaptic Transmission/physiology , Adaptor Proteins, Vesicular Transport , Animals , Cochlear Nerve/physiology , Hair Cells, Auditory, Inner/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Synapses/metabolism
18.
Neuron ; 60(4): 642-56, 2008 Nov 26.
Article En | MEDLINE | ID: mdl-19038221

Neuronal nitric oxide synthase (nNOS) is broadly expressed in the brain and associated with synaptic plasticity through NMDAR-mediated calcium influx. However, its physiological activation and the mechanisms by which nitric oxide (NO) influences synaptic transmission have proved elusive. Here, we exploit the unique input-specificity of the calyx of Held to characterize NO modulation at this glutamatergic synapse in the auditory pathway. NO is generated in an activity-dependent manner by MNTB principal neurons receiving a calyceal synaptic input. It acts in the target neuron and adjacent inactive neurons to modulate excitability and synaptic efficacy, inhibiting postsynaptic Kv3 potassium currents (via phosphorylation), reducing EPSCs and so increasing action potential duration and reducing transmission fidelity. We conclude that NO serves as a volume transmitter and slow dynamic modulator, integrating spontaneous and evoked neuronal firing, thereby providing an index of global activity and regulating information transmission across a population of active and inactive neurons.


Auditory Pathways/metabolism , Glutamic Acid/metabolism , Nitrergic Neurons/metabolism , Nitric Oxide/metabolism , Pons/metabolism , Synaptic Transmission/physiology , Action Potentials/physiology , Animals , Auditory Pathways/cytology , Excitatory Postsynaptic Potentials/physiology , Mice , Mice, Inbred CBA , Nitric Oxide Synthase Type I/metabolism , Organ Culture Techniques , Pons/cytology , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Shaw Potassium Channels/metabolism , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure
19.
Eur J Neurosci ; 27(12): 3191-204, 2008 Jun.
Article En | MEDLINE | ID: mdl-18598262

The neuronal representation of acoustic amplitude modulations is an important prerequisite for understanding the processing of natural sounds. We investigated this representation in the medial nucleus of the trapezoid body (MNTB) of the Mongolian gerbil using sinusoidal amplitude modulations (SAM). Depending on the SAM's carrier frequency (f(C)) MNTB cells either increase or decrease their discharge rates, indicating underlying excitatory and inhibitory/suppressive mechanisms. As natural sounds typically are composed of multiple spectral components we investigated how stimuli containing two spectral components are represented in the MNTB, especially when they have opposing effects on the discharge rate. Three conditions were compared: SAM stimuli (1) with rate-increasing f(C), (2) with rate-increasing f(C) and an additional unmodulated rate-decreasing pure tone, and (3) with rate-decreasing f(C) and an unmodulated, rate-increasing pure tone. We found that responses under all three conditions showed comparable strength of phase-locking. Adding a rate-decreasing tone to a rate-increasing SAM increased phase-locking for modulation frequencies (f(AM)) of < or = 600 Hz. A comparison of two possible coding strategies--phase-locking vs. envelope reproduction--indicates that both strategies are realized to different degrees depending on the f(AM). We measured latencies for following modulations in rate-increasing and rate-decreasing SAMs using a modified reverse correlation approach. Although latencies varied between 2.5 and 5 ms between cells, a decrease in rate consistently followed an increase in rate with a delay of about 0.2 ms in each cell. These results suggest a temporally precise representation of rate-increasing and rate-decreasing stimuli at the level of the MNTB during dynamic stimulation.


Auditory Pathways/physiology , Models, Neurological , Neural Inhibition/physiology , Neurons/physiology , Olivary Nucleus/physiology , Acoustic Stimulation , Action Potentials/physiology , Animals , Auditory Pathways/cytology , Excitatory Postsynaptic Potentials/physiology , Gerbillinae , Inhibitory Postsynaptic Potentials/physiology , Olivary Nucleus/cytology , Reaction Time/physiology
20.
J Neurosci ; 23(27): 9199-207, 2003 Oct 08.
Article En | MEDLINE | ID: mdl-14534254

The voltage-gated potassium (Kv) channel subunit Kv1.1, encoded by the Kcna1 gene, is expressed strongly in the ventral cochlear nucleus (VCN) and the medial nucleus of the trapezoid body (MNTB) of the auditory pathway. To examine the contribution of the Kv1.1 subunit to the processing of auditory information, in vivo single-unit recordings were made from VCN neurons (bushy cells), axonal endings of bushy cells at MNTB cells (calyces of Held), and MNTB neurons of Kcna1-null (-/-) mice and littermate control (+/+) mice. Thresholds and spontaneous firing rates of VCN and MNTB neurons were not different between genotypes. At higher sound intensities, however, evoked firing rates of VCN and MNTB neurons were significantly lower in -/- mice than +/+ mice. The SD of the first-spike latency (jitter) was increased in VCN neurons, calyces, and MNTB neurons of -/- mice compared with +/+ controls. Comparison along the ascending pathway suggests that the increased jitter found in -/- MNTB responses arises mostly in the axons of VCN bushy cells and/or their calyceal terminals rather than in the MNTB neurons themselves. At high rates of sinusoidal amplitude modulations, -/- MNTB neurons maintained high vector strength values but discharged on significantly fewer cycles of the amplitude-modulated stimulus than +/+ MNTB neurons. These results indicate that in Kcna1-null mice the absence of the Kv1.1 subunit results in a loss of temporal fidelity (increased jitter) and the failure to follow high-frequency amplitude-modulated sound stimulation in vivo.


Auditory Pathways/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/deficiency , Reaction Time/physiology , Acoustic Stimulation/methods , Action Potentials/physiology , Animals , Auditory Threshold , Cochlear Nucleus/physiology , Electrodes, Implanted , Electrophysiology , Kv1.1 Potassium Channel , Mice , Mice, Knockout , Potassium Channels/genetics , Reaction Time/genetics , Time Factors
...