Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Hippocampus ; 27(12): 1217-1223, 2017 12.
Article in English | MEDLINE | ID: mdl-28881444

ABSTRACT

The perirhinal cortex (PRh) is a key region downstream of auditory cortex (ACx) that processes familiarity linked mnemonic signaling. In gerbils, ACx-driven EPSPs recorded in PRh neurons are largely shunted by GABAergic inhibition (Kotak et al., 2015, Frontiers in Neural Circuits, 9). To determine whether inhibitory shunting prevents the induction of excitatory long-term potentiation (e-LTP), we stimulated ACx-recipient PRh in a brain slice preparation using theta burst stimulation (TBS). Under control conditions, without GABA blockers, the majority of PRh neurons exhibited long-term depression. A very low concentration of bicuculline increased EPSP amplitude, but under this condition TBS did not significantly increase e-LTP induction. Since PRh synaptic inhibition included a GABAB receptor-mediated component, we added a GABAB receptor antagonist. When both GABAA and GABAB receptors were blocked, TBS reliably induced e-LTP in a majority of PRh neurons. We conclude that GABAergic transmission is a vital mechanism regulating e-LTP induction in the PRh, and may be associated with auditory learning.


Subject(s)
Long-Term Potentiation/physiology , Neural Inhibition/physiology , Perirhinal Cortex/metabolism , Receptors, GABA/metabolism , Animals , Bicuculline/pharmacology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABA Antagonists/pharmacology , Gerbillinae , Long-Term Potentiation/drug effects , Microelectrodes , Neural Inhibition/drug effects , Neurons/drug effects , Neurons/metabolism , Perirhinal Cortex/drug effects , Tissue Culture Techniques
4.
Cell Rep ; 19(12): 2462-2468, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636935

ABSTRACT

Corticostriatal circuits play a fundamental role in regulating many behaviors, and their dysfunction is associated with many neurological disorders. In contrast, sensory disorders, like hearing loss (HL), are commonly linked with processing deficits at or below the level of the auditory cortex (ACx). However, HL can be accompanied by non-sensory deficits, such as learning delays, suggesting the involvement of regions downstream of ACx. Here, we show that transient developmental HL differentially affected the ACx and its downstream target, the sensory striatum. Following HL, both juvenile ACx layer 5 and striatal neurons displayed an excitatory-inhibitory imbalance and lower firing rates. After hearing was restored, adult ACx neurons recovered balanced excitatory-inhibitory synaptic gain and control-like firing rates, but striatal neuron synapses and firing properties did not recover. Thus, a brief period of abnormal cortical activity may induce cellular impairments that persist into adulthood and contribute to neurological disorders that are striatal in origin.


Subject(s)
Auditory Cortex/growth & development , Animals , Auditory Cortex/cytology , Auditory Pathways , Corpus Striatum/physiology , Excitatory Postsynaptic Potentials , Female , Gerbillinae , Male , Sensory Deprivation/physiology
5.
Nat Neurosci ; 19(12): 1743-1749, 2016 12.
Article in English | MEDLINE | ID: mdl-27798629

ABSTRACT

A fundamental impediment to understanding the brain is the availability of inexpensive and robust methods for targeting and manipulating specific neuronal populations. The need to overcome this barrier is pressing because there are considerable anatomical, physiological, cognitive and behavioral differences between mice and higher mammalian species in which it is difficult to specifically target and manipulate genetically defined functional cell types. In particular, it is unclear the degree to which insights from mouse models can shed light on the neural mechanisms that mediate cognitive functions in higher species, including humans. Here we describe a novel recombinant adeno-associated virus that restricts gene expression to GABAergic interneurons within the telencephalon. We demonstrate that the viral expression is specific and robust, allowing for morphological visualization, activity monitoring and functional manipulation of interneurons in both mice and non-genetically tractable species, thus opening the possibility to study GABAergic function in virtually any vertebrate species.


Subject(s)
Brain/virology , Dependovirus/isolation & purification , GABAergic Neurons/virology , Interneurons/physiology , Vertebrates/virology , Animals , Behavior, Animal , Brain/metabolism , Cells, Cultured , Dependovirus/genetics , Female , GABAergic Neurons/pathology , Genetic Vectors/genetics , Mice, Inbred C57BL
6.
Nat Commun ; 7: 10416, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26786281

ABSTRACT

Sensory systems influence one another during development and deprivation can lead to cross-modal plasticity. As auditory function begins before vision, we investigate the effect of manipulating visual experience during auditory cortex critical periods (CPs) by assessing the influence of early, normal and delayed eyelid opening on hearing loss-induced changes to membrane and inhibitory synaptic properties. Early eyelid opening closes the auditory cortex CPs precociously and dark rearing prevents this effect. In contrast, delayed eyelid opening extends the auditory cortex CPs by several additional days. The CP for recovery from hearing loss is also closed prematurely by early eyelid opening and extended by delayed eyelid opening. Furthermore, when coupled with transient hearing loss that animals normally fully recover from, very early visual experience leads to inhibitory deficits that persist into adulthood. Finally, we demonstrate a functional projection from the visual to auditory cortex that could mediate these effects.


Subject(s)
Auditory Cortex/physiology , Auditory Cortex/physiopathology , Visual Cortex/physiopathology , Animals , Female , Gerbillinae , Hearing/physiology , Hearing Loss/physiopathology , Humans , Male , Neuronal Plasticity/physiology , Sensory Deprivation/physiology , Vision, Ocular/physiology , Visual Cortex/physiology
7.
Article in English | MEDLINE | ID: mdl-26321918

ABSTRACT

The representation of acoustic cues involves regions downstream from the auditory cortex (ACx). One such area, the perirhinal cortex (PRh), processes sensory signals containing mnemonic information. Therefore, our goal was to assess whether PRh receives auditory inputs from the auditory thalamus (MG) and ACx in an auditory thalamocortical brain slice preparation and characterize these afferent-driven synaptic properties. When the MG or ACx was electrically stimulated, synaptic responses were recorded from the PRh neurons. Blockade of type A gamma-aminobutyric acid (GABA-A) receptors dramatically increased the amplitude of evoked excitatory potentials. Stimulation of the MG or ACx also evoked calcium transients in most PRh neurons. Separately, when fluoro ruby was injected in ACx in vivo, anterogradely labeled axons and terminals were observed in the PRh. Collectively, these data show that the PRh integrates auditory information from the MG and ACx and that auditory driven inhibition dominates the postsynaptic responses in a non-sensory cortical region downstream from the ACx.


Subject(s)
Auditory Cortex/cytology , Auditory Cortex/physiology , Auditory Pathways/physiology , Neurons/physiology , Synapses/physiology , Animals , Auditory Pathways/drug effects , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Calcium/metabolism , Dextrans/metabolism , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , GABA-A Receptor Antagonists/pharmacology , Gerbillinae , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Quinoxalines/pharmacology , Rhodamines/metabolism , Statistics, Nonparametric , Synapses/drug effects , Thalamus/physiology
8.
J Neurosci ; 35(16): 6318-25, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904785

ABSTRACT

Auditory learning is associated with an enhanced representation of acoustic cues in primary auditory cortex, and modulation of inhibitory strength is causally involved in learning. If this inhibitory plasticity is associated with task learning and improvement, its expression should emerge and persist until task proficiency is achieved. We tested this idea by measuring changes to cortical inhibitory synaptic transmission as adult gerbils progressed through the process of associative learning and perceptual improvement. Using either of two procedures, aversive or appetitive conditioning, animals were trained to detect amplitude-modulated noise and then tested daily. Following each training session, a thalamocortical brain slice was generated, and inhibitory synaptic properties were recorded from layer 2/3 pyramidal neurons. Initial associative learning was accompanied by a profound reduction in the amplitude of spontaneous IPSCs (sIPSCs). However, sIPSC amplitude returned to control levels when animals reached asymptotic behavioral performance. In contrast, paired-pulse ratios decreased in trained animals as well as in control animals that experienced unpaired conditioned and unconditioned stimuli. This latter observation suggests that inhibitory release properties are modified during behavioral conditioning, even when an association between the sound and reinforcement cannot occur. These results suggest that associative learning is accompanied by a reduction of postsynaptic inhibitory strength that persists for several days during learning and perceptual improvement.


Subject(s)
Association Learning/physiology , Auditory Cortex/physiology , Neural Inhibition/physiology , Animals , Auditory Perception/physiology , Conditioning, Classical/physiology , Gerbillinae , Inhibitory Postsynaptic Potentials/physiology , Male , Pyramidal Cells/physiology , Synaptic Transmission/physiology
9.
Cereb Cortex ; 25(8): 2083-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-24554724

ABSTRACT

Sensory deprivation can induce profound changes to central processing during developmental critical periods (CPs), and the recovery of normal function is maximal if the sensory input is restored during these epochs. Therefore, we asked whether mild and transient hearing loss (HL) during discrete CPs could induce changes to cortical cellular physiology. Electrical and inhibitory synaptic properties were obtained from auditory cortex pyramidal neurons using whole-cell recordings after bilateral earplug insertion or following earplug removal. Varying the age of HL onset revealed brief CPs of vulnerability for membrane and firing properties, as well as, inhibitory synaptic currents. These CPs closed 1 week after ear canal opening on postnatal day (P) 18. To examine whether the cellular properties could recover from HL, earplugs were removed prior to (P17) or after (P23), the closure of these CPs. The earlier age of hearing restoration led to greater recovery of cellular function, but firing rate remained disrupted. When earplugs were removed after the closure of these CPs, several changes persisted into adulthood. Therefore, long-lasting cellular deficits that emerge from transient deprivation during a CP may contribute to delayed acquisition of auditory skills in children who experience temporary HL.


Subject(s)
Auditory Cortex/growth & development , Auditory Cortex/physiopathology , Hearing Loss/physiopathology , Pyramidal Cells/physiology , Acoustic Stimulation , Action Potentials/physiology , Age of Onset , Animals , Disease Models, Animal , Ear Protective Devices , Gerbillinae , Hearing Tests , Neural Inhibition/physiology , Neural Pathways/physiopathology , Patch-Clamp Techniques , Recovery of Function/physiology , Sensory Deprivation/physiology , Thalamus/growth & development , Thalamus/physiopathology , Tissue Culture Techniques
10.
Article in English | MEDLINE | ID: mdl-24994969

ABSTRACT

Principal neurons of the lateral superior olivary nucleus (LSO) respond selectively to interaural level differences (ILD). To perform this computation, LSO neurons integrate excitatory synaptic drive from the ipsilateral ear with inhibitory synaptic drive from the contralateral ear via the medial nucleus of the trapezoid body (MNTB). Previous research demonstrated that inhibitory terminals from the MNTB to the LSO are eliminated during development. Furthermore, MNTB synapses display an activity- and age-dependent long-term depression (iLTD) that may contribute to inhibitory synapse elimination. However, inhibitory synapses that are stabilized become stronger. Here, we asked whether MNTB synapses displayed activity-dependent strengthening. Whole-cell recordings were obtained from LSO neurons in a gerbil brain slice before and after hearing onset. The inhibitory MNTB afferents were stimulated at a low rate, similar to spontaneous discharge rates observed in vivo. The MNTB-evoked inhibitory responses were strengthened by 40-300% when synaptic activity was coupled with postsynaptic membrane depolarization, exogenous glutamate application, or activation of ipsilateral excitatory synaptic inputs. This inhibitory long-term potentiation (iLTP) was associated with increased spontaneous inhibitory postsynaptic current (IPSC) amplitude and frequency. One hour after iLTP induction, IPSCs could not be de-potentiated by the MNTB stimulation pattern that induces iLTD in control slices. iLTP could only be induced after hearing onset (>P12), and was blocked in the presence of a GABAB receptor antagonist. Together, these results suggest a developmental period during which the induction of iLTP depends on the conjoint activation of GABAB receptors and postsynaptic depolarization. We propose that iLTP may support stabilization of un-pruned MNTB connections and contribute to the emergence of ILD processing in the mature LSO.


Subject(s)
Auditory Pathways/physiology , Inhibitory Postsynaptic Potentials/physiology , Long-Term Potentiation/physiology , Neurons/physiology , Olivary Nucleus/physiology , Age Factors , Animals , Auditory Pathways/growth & development , Gerbillinae , Olivary Nucleus/growth & development , Patch-Clamp Techniques , Synapses/physiology
11.
J Neurophysiol ; 112(4): 802-13, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24848460

ABSTRACT

Behavioral and neural findings demonstrate that animals can locate low-frequency sounds along the azimuth by detecting microsecond interaural time differences (ITDs). Information about ITDs is also available in the amplitude modulations (i.e., envelope) of high-frequency sounds. Since medial superior olivary (MSO) neurons encode low-frequency ITDs, we asked whether they employ a similar mechanism to process envelope ITDs with high-frequency carriers, and the effectiveness of this mechanism compared with the process of low-frequency sound. We developed a novel hybrid in vitro dynamic-clamp approach, which enabled us to mimic synaptic input to brain-slice neurons in response to virtual sound and to create conditions that cannot be achieved naturally but are useful for testing our hypotheses. For each simulated ear, a virtual sound, computer generated, was used as input to a computational auditory-nerve model. Model spike times were converted into synaptic input for MSO neurons, and ITD tuning curves were derived for several virtual-sound conditions: low-frequency pure tones, high-frequency tones modulated with two types of envelope, and speech sequences. Computational models were used to verify the physiological findings and explain the biophysical mechanism underlying the observed ITD coding. Both recordings and simulations indicate that MSO neurons are sensitive to ITDs carried by spectrotemporally complex virtual sounds, including speech tokens. Our findings strongly suggest that MSO neurons can encode ITDs across a broad-frequency spectrum using an input-slope-based coincidence-detection mechanism. Our data also provide an explanation at the cellular level for human localization performance involving high-frequency sound described by previous investigators.


Subject(s)
Evoked Potentials, Auditory , Models, Neurological , Sound Localization , Animals , Cochlear Nerve/physiology , Gerbillinae , Humans , Neurons/physiology , Speech Perception , Superior Olivary Complex/cytology , Superior Olivary Complex/physiology
12.
J Neurophysiol ; 110(4): 999-1008, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23719211

ABSTRACT

Sensory deprivation, such as developmental hearing loss, leads to an adjustment of synaptic and membrane properties throughout the central nervous system. These changes are thought to compensate for diminished sound-evoked activity. This model predicts that compensatory changes should be synergistic with one another along each functional pathway. To test this idea, we examined the excitatory thalamic drive to two types of cortical inhibitory interneurons that display differential effects in response to developmental hearing loss. The inhibitory synapses made by fast-spiking (FS) cells are weakened by hearing loss, whereas those made by low threshold-spiking (LTS) cells remain strong but display greater short-term depression (Takesian et al. 2010). Whole-cell recordings were made from FS or LTS interneurons in a thalamocortical brain slice, and medial geniculate (MG)-evoked postsynaptic potentials were analyzed. Following hearing loss, MG-evoked net excitatory potentials were smaller than normal at FS cells but larger than normal at LTS cells. Furthermore, MG-evoked excitatory potentials displayed less short-term depression at FS cells and greater short-term depression at LTS cells. Thus deprivation-induced adjustments of excitatory synapses onto inhibitory interneurons are cell-type specific and parallel the changes made by the inhibitory afferents.


Subject(s)
Auditory Cortex/physiopathology , GABAergic Neurons/physiology , Geniculate Bodies/physiopathology , Hearing Loss/physiopathology , Neuronal Plasticity , Animals , Auditory Cortex/growth & development , GABAergic Neurons/classification , Geniculate Bodies/growth & development , Gerbillinae , Neural Inhibition , Neural Pathways , Synaptic Potentials
13.
PLoS One ; 8(1): e53438, 2013.
Article in English | MEDLINE | ID: mdl-23326429

ABSTRACT

Inhibitory synapse dysfunction may contribute to many developmental brain disorders, including the secondary consequences of sensory deprivation. In fact, developmental hearing loss leads to a profound reduction in the strength of inhibitory postsynaptic currents (IPSCs) in the auditory cortex, and this deficit persists into adulthood. This finding is consistent with the general theory that the emergence of mature synaptic properties requires activity during development. Therefore, we tested the prediction that inhibitory strength can be restored following developmental hearing loss by boosting GABAergic transmission in vivo. Conductive or sensorineural hearing loss was induced surgically in gerbils prior to hearing onset and GABA agonists were then administered for one week. IPSCs were subsequently recorded from pyramidal neurons in a thalamocortical brain slice preparation. Administration of either a GABA(A) receptor a1 subunit specific agonist (zolpidem), or a selective GABA reuptake inhibitor (SGRI), rescued IPSC amplitude in hearing loss animals. Furthermore, this restoration persisted in adults, long after drug treatment ended. In contrast, a GABA(B) receptor agonist baclofen did not restore inhibitory strength. IPSCs could also be restored when SGRI administration began 3 weeks after sensory deprivation. Together, these results demonstrate long-lasting restoration of cortical inhibitory strength in the absence of normal experience. This suggests that in vivo GABA(A) receptor activation is sufficient to promote maturation, and this principle may extend to other developmental disorders associated with diminished inhibitory function.


Subject(s)
Gerbillinae/physiology , Hearing Loss/physiopathology , Neural Inhibition , Synapses/pathology , Animals , Baclofen/pharmacology , Baclofen/therapeutic use , Cochlea/drug effects , Cochlea/physiopathology , GABA Uptake Inhibitors/pharmacology , GABA Uptake Inhibitors/therapeutic use , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , Hearing Loss/drug therapy , Hearing Loss/pathology , Inhibitory Postsynaptic Potentials/drug effects , Neural Inhibition/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, GABA-A/metabolism , Synapses/drug effects , Zolpidem
14.
J Neurophysiol ; 107(3): 937-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22090457

ABSTRACT

The developmental plasticity of excitatory synapses is well established, particularly as a function of age. If similar principles apply to inhibitory synapses, then we would expect manipulations during juvenile development to produce a greater effect and experience-dependent changes to persist into adulthood. In this study, we first characterized the maturation of cortical inhibitory synapse function from just before the onset of hearing through adulthood. We then examined the long-term effects of developmental conductive hearing loss (CHL). Whole cell recordings from gerbil thalamocortical brain slices revealed a significant decrease in the decay time of inhibitory currents during the first 3 mo of normal development. When assessed in adults, developmental CHL led to an enduring decrease of inhibitory synaptic strength, whereas the maturation of synaptic decay time was only delayed. Early CHL also depressed the maximum discharge rate of fast-spiking, but not low-threshold-spiking, inhibitory interneurons. We then asked whether adult onset CHL had a similar effect, but neither inhibitory current amplitude nor decay time was altered. Thus inhibitory synapse function displays a protracted development during which deficits can be induced by juvenile, but not adult, hearing loss. These long-lasting changes to inhibitory function may contribute to the auditory processing deficits associated with early hearing loss.


Subject(s)
Auditory Cortex/physiopathology , Auditory Perception/physiology , Hearing Loss, Conductive/physiopathology , Inhibitory Postsynaptic Potentials/physiology , Synapses/physiology , Age Factors , Animals , Auditory Cortex/growth & development , Auditory Pathways/physiopathology , Female , Gerbillinae , Interneurons/physiology , Male , Patch-Clamp Techniques
15.
Hear Res ; 279(1-2): 140-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21463668

ABSTRACT

Functional inhibitory synapses form in auditory cortex well before the onset of normal hearing. However, their properties change dramatically during normal development, and many of these maturational events are delayed by hearing loss. Here, we review recent findings on the developmental plasticity of inhibitory synapse strength, kinetics, and GABAA receptor localization in auditory cortex. Although hearing loss generally leads to a reduction of inhibitory strength, this depends on the type of presynaptic interneuron. Furthermore, plasticity of inhibitory synapses also depends on the postsynaptic target. Hearing loss leads reduced GABAA receptor localization to the membrane of excitatory, but not inhibitory neurons. A reduction in normal activity in development can also affect the use-dependent plasticity of inhibitory synapses. Even moderate hearing loss can disrupt inhibitory short- and long-term synaptic plasticity. Thus, the cortex did not compensate for the loss of inhibition in the brainstem, but rather exacerbated the response to hearing loss by further reducing inhibitory drive. Together, these results demonstrate that inhibitory synapses are exceptionally dynamic during development, and deafness-induced perturbation of inhibitory properties may have a profound impact on auditory processing.


Subject(s)
Neuronal Plasticity/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Auditory Cortex/embryology , Auditory Cortex/physiology , Auditory Pathways , Deafness/pathology , Hearing Loss , Humans , Kinetics , Mice , Models, Biological , Neural Inhibition/physiology , Neurons/physiology , Receptors, GABA-A/metabolism
16.
PLoS Biol ; 8(6): e1000406, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20613857

ABSTRACT

Low-frequency sound localization depends on the neural computation of interaural time differences (ITD) and relies on neurons in the auditory brain stem that integrate synaptic inputs delivered by the ipsi- and contralateral auditory pathways that start at the two ears. The first auditory neurons that respond selectively to ITD are found in the medial superior olivary nucleus (MSO). We identified a new mechanism for ITD coding using a brain slice preparation that preserves the binaural inputs to the MSO. There was an internal latency difference for the two excitatory pathways that would, if left uncompensated, position the ITD response function too far outside the physiological range to be useful for estimating ITD. We demonstrate, and support using a biophysically based computational model, that a bilateral asymmetry in excitatory post-synaptic potential (EPSP) slopes provides a robust compensatory delay mechanism due to differential activation of low threshold potassium conductance on these inputs and permits MSO neurons to encode physiological ITDs. We suggest, more generally, that the dependence of spike probability on rate of depolarization, as in these auditory neurons, provides a mechanism for temporal order discrimination between EPSPs.


Subject(s)
Auditory Pathways , Sound , Synapses/physiology , Animals , Excitatory Postsynaptic Potentials , Gerbillinae , In Vitro Techniques
17.
J Neurosci ; 30(7): 2716-27, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20164356

ABSTRACT

Short-term changes in synaptic gain support information processing throughout the CNS, yet we know little about the developmental regulation of such plasticity. Here we report that auditory experience is necessary for the normal maturation of synaptic inhibitory short-term plasticity (iSTP) in the auditory cortex, and that presynaptic GABA(B) receptors regulate this development. Moderate or severe hearing loss was induced in gerbils, and iSTP was characterized by measuring inhibitory synaptic current amplitudes in response to repetitive stimuli. We reveal a profound developmental shift of iSTP from depressing to facilitating after the onset of hearing. Even moderate hearing loss prevented this shift. This iSTP change was mediated by a specific class of inhibitory interneurons, the low-threshold spiking cells. Further, using paired recordings, we reveal that presynaptic GABA(B) receptors at interneuron-pyramidal connections regulate iSTP in an experience-dependent manner. This novel synaptic mechanism may support the emergence of mature temporal processing in the auditory cortex.


Subject(s)
Neural Inhibition/physiology , Neuronal Plasticity/physiology , Presynaptic Terminals/physiology , Pyramidal Cells/physiology , Receptors, GABA-B/metabolism , Age Factors , Animals , Animals, Newborn , Auditory Cortex/cytology , Auditory Cortex/growth & development , Auditory Pathways/physiology , Baclofen/pharmacology , Biophysics , Disease Models, Animal , Electric Stimulation/methods , GABA Agonists/pharmacology , GABA-B Receptor Antagonists , Gerbillinae , Hearing Loss, Conductive/pathology , Hearing Loss, Conductive/physiopathology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/physiology , Morpholines/pharmacology , Neural Inhibition/drug effects , Patch-Clamp Techniques/methods , Presynaptic Terminals/drug effects , Pyramidal Cells/drug effects
18.
J Neurosci ; 30(1): 331-41, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053914

ABSTRACT

Long-term synaptic plasticity is a putative mechanism for learning in adults. However, there is little understanding of how synaptic plasticity mechanisms develop or whether their maturation depends on experience. Since inhibitory synapses are particularly malleable to sensory stimulation, long-lasting potentiation of inhibitory synapses was characterized in auditory thalamocortical slices. Intracortical high-frequency electrical stimulation led to a 67% increase in inhibitory synaptic currents. In the absence of stimulation, inhibitory potentiation was induced by a brief exposure to exogenous brain-derived neurotrophic factor (BDNF). BDNF exposure occluded any additional potentiation by high-frequency afferent stimulation, suggesting that BDNF signaling is sufficient to account for inhibitory potentiation. Moreover, inhibitory potentiation was reduced significantly by extracellular application of a BDNF scavenger or by intracellular blockade of BDNF receptor [tropomyosin-related kinase B (TrkB)] signaling. In contrast, glutamatergic or GABAergic antagonists did not prevent the induction of inhibitory potentiation. Since BDNF and TrkB expression are influenced strongly by activity, we predicted that inhibitory potentiation would be diminished by manipulations that decrease central auditory activity, such as hearing loss. Two forms of hearing loss were examined: conductive hearing loss in which the cochleae are not damaged or sensorineural hearing loss in which both cochleae are removed. Both forms of hearing loss were found to reduce significantly the magnitude of inhibitory potentiation. These data indicate that early experience is necessary for the normal development of BDNF-mediated long-lasting inhibitory potentiation, which may be associated with perceptual deficits at later ages.


Subject(s)
Auditory Cortex/physiology , Auditory Perception/physiology , Hearing/physiology , Long-Term Synaptic Depression/physiology , Neural Inhibition/physiology , Acoustic Stimulation/methods , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/physiology , Gerbillinae , Neuronal Plasticity/physiology
19.
Future Neurol ; 4(3): 331-349, 2009 May 01.
Article in English | MEDLINE | ID: mdl-20161214

ABSTRACT

Hearing loss during development leads to central deficits that persist even after the restoration of peripheral function. One key class of deficits is due to changes in central inhibitory synapses, which play a fundamental role in all aspects of auditory processing. This review focuses on the anatomical and physiological alterations of inhibitory connections at several regions within the central auditory pathway following hearing loss. Such aberrant inhibitory synaptic function may be linked to deficits in encoding binaural and spectral cues. Understanding the cellular changes that occur at inhibitory synapses following hearing loss may provide specific loci that can be targeted to improve function.

20.
Cereb Cortex ; 18(12): 2855-67, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18403398

ABSTRACT

We have shown previously that auditory experience regulates the maturation of excitatory synapses in the auditory cortex (ACx). In this study, we used electron microscopic immunocytochemistry to determine whether the heightened excitability of the ACx following neonatal sensorineural hearing loss (SNHL) also involves pre- or postsynaptic alterations of GABAergic synapses. SNHL was induced in gerbils just prior to the onset of hearing (postnatal day 10). At P17, the gamma-aminobutyri acid type A (GABA(A)) receptor's beta2/3-subunit (GABA(A)beta2/3) clusters residing at plasma membranes in layers 2/3 of ACx was reduced significantly in size (P < 0.05) and number (P < 0.005), whereas the overall number of immunoreactive puncta (intracellular + plasmalemmal) remained unchanged. The reduction of GABA(A)beta2/3 was observed along perikaryal plasma membranes of excitatory neurons but not of GABAergic interneurons. This cell-specific change can contribute to the enhanced excitability of SNHL ACx. Presynaptically, GABAergic axon terminals were significantly larger but less numerous and contained 47% greater density of glutamic acid decarboxylase immunoreactivity (P < 0.05). This suggests that GABA synthesis may be upregulated by a retrograde signal arising from lowered levels of postsynaptic GABA(A)R. Thus, both, the pre- and postsynaptic sides of inhibitory synapses that form upon pyramidal neurons of the ACx are regulated by neonatal auditory experience.


Subject(s)
Auditory Cortex/physiopathology , Hearing Loss, Sensorineural/physiopathology , Receptors, GABA-A/physiology , Synapses/physiology , Animals , Auditory Cortex/pathology , Auditory Cortex/ultrastructure , Axons/pathology , Axons/ultrastructure , Dendrites/pathology , Dendrites/ultrastructure , Gerbillinae , Glutamate Decarboxylase/metabolism , Hearing Loss, Sensorineural/enzymology , Hearing Loss, Sensorineural/pathology , Neurons/enzymology , Neurons/physiology , Reference Values , Synapses/ultrastructure , Thalamus/pathology , Thalamus/physiopathology , Thalamus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...