Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 29(12): 5166-5179, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31050701

ABSTRACT

GABA signaling sustains fundamental brain functions, from nervous system development to the synchronization of population activity and synaptic plasticity. Despite these pivotal features, molecular determinants underscoring the rapid and cell-autonomous replenishment of the vesicular neurotransmitter GABA and its impact on synaptic plasticity remain elusive. Here, we show that genetic disruption of the glutamine transporter Slc38a1 in mice hampers GABA synthesis, modifies synaptic vesicle morphology in GABAergic presynapses and impairs critical period plasticity. We demonstrate that Slc38a1-mediated glutamine transport regulates vesicular GABA content, induces high-frequency membrane oscillations and shapes cortical processing and plasticity. Taken together, this work shows that Slc38a1 is not merely a transporter accumulating glutamine for metabolic purposes, but a key component regulating several neuronal functions.


Subject(s)
Amino Acid Transport System A/metabolism , Brain/physiology , GABAergic Neurons/physiology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Mice
2.
J Neurosci ; 35(7): 3016-21, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698739

ABSTRACT

To date, it has been difficult to reveal physiological Ca(2+) events occurring within the fine astrocytic processes of mature animals. The objective of the study was to explore whether neuronal activity evokes astrocytic Ca(2+) signals at glutamatergic synapses of adult mice. We stimulated the Schaffer collateral/commissural fibers in acute hippocampal slices from adult mice transduced with the genetically encoded Ca(2+) indicator GCaMP5E driven by the glial fibrillary acidic protein promoter. Two-photon imaging revealed global stimulation-evoked astrocytic Ca(2+) signals with distinct latencies, rise rates, and amplitudes in fine processes and somata. Specifically, the Ca(2+) signals in the processes were faster and of higher amplitude than those in the somata. A combination of P2 purinergic and group I/II metabotropic glutamate receptor (mGluR) antagonists reduced the amplitude of the Ca(2+) transients by 30-40% in both astrocytic compartments. Blockage of the mGluRs alone only modestly reduced the magnitude of the stimulation-evoked Ca(2+) signals in processes and failed to affect the somatic Ca(2+) response. Local application of group I or I/II mGluR agonists or adenosine triphosphate (ATP) elicited global astrocytic Ca(2+) signals that mimicked the stimulation-evoked astrocytic Ca(2+) responses. We conclude that stimulation-evoked Ca(2+) signals in astrocytic processes at CA3-CA1 synapses of adult mice (1) differ from those in astrocytic somata and (2) are modulated by glutamate and ATP.


Subject(s)
Adenosine Triphosphate/pharmacology , Astrocytes/metabolism , Calcium Signaling/drug effects , Glutamic Acid/pharmacology , Hippocampus/cytology , Synapses/drug effects , Animals , Astrocytes/drug effects , Calcium/metabolism , Calcium Signaling/physiology , Calmodulin/genetics , Calmodulin/metabolism , Dioxolanes/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenylacetates/pharmacology , Purines/pharmacology , Synapses/physiology , Synapsins/genetics , Synapsins/metabolism , Time Factors
3.
Brain Struct Funct ; 220(4): 2469-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24744149

ABSTRACT

The coupling between the water channel aquaporin-4 (AQP4) and K(+) transport has attracted much interest. In this study, we assessed the effect of Aqp4 deletion on activity-induced [K(+)]o changes in acute slices from hippocampus and corpus callosum of adult mice. We show that Aqp4 deletion has a layer-specific effect on [K(+)]o that precisely mirrors the known effect on extracellular volume dynamics. In CA1, the peak [K(+)]o in stratum radiatum during 20 Hz stimulation of Schaffer collateral/commissural fibers was significantly higher in Aqp4 (-/-) mice than in wild types, whereas no differences were observed throughout the [K(+)]o recovery phase. In stratum pyramidale and corpus callosum, neither peak [K(+)]o nor post-stimulus [K(+)]o recovery was affected by Aqp4 deletion. Our data suggest that AQP4 modulates [K(+)]o during synaptic stimulation through its effect on extracellular space volume.


Subject(s)
Aquaporin 4/deficiency , Extracellular Space/metabolism , Hippocampus/metabolism , Potassium/metabolism , Synapses/physiology , Animals , Aquaporin 4/genetics , Electric Stimulation , In Vitro Techniques , Ion-Selective Electrodes , Mice , Mice, Transgenic
4.
Nat Neurosci ; 15(8): 1153-9, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22797694

ABSTRACT

Hippocampal NMDA receptors (NMDARs) and NMDAR-dependent synaptic plasticity are widely considered crucial substrates of long-term spatial memory, although their precise role remains uncertain. Here we show that Grin1(ΔDGCA1) mice, lacking GluN1 and hence NMDARs in all dentate gyrus and dorsal CA1 principal cells, acquired the spatial reference memory water maze task as well as controls, despite impairments on the spatial reference memory radial maze task. When we ran a spatial discrimination water maze task using two visually identical beacons, Grin1(ΔDGCA1) mice were impaired at using spatial information to inhibit selecting the decoy beacon, despite knowing the platform's actual spatial location. This failure could suffice to impair radial maze performance despite spatial memory itself being normal. Thus, these hippocampal NMDARs are not essential for encoding or storing long-term, associative spatial memories. Instead, we demonstrate an important function of the hippocampus in using spatial knowledge to select between alternative responses that arise from competing or overlapping memories.


Subject(s)
Behavior, Animal/physiology , Hippocampus/physiology , Memory/physiology , Nerve Tissue Proteins/deficiency , Animals , Carrier Proteins/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Hippocampus/metabolism , Maze Learning/physiology , Mice , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate , Space Perception/physiology
5.
J Neurosci ; 32(16): 5688-703, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22514330

ABSTRACT

Alterations of learning and memory in mice with deregulated neuron-specific nuclear factor κB (NF-κB) activity support the idea that plastic changes of synaptic contacts may depend at least in part on IκB kinase (IKK)/NF-κB-related synapse-to-nucleus signaling. There is, however, little information on the molecular requirements and mechanisms regulating this IKK/NF-κB-dependent synapse development and remodeling. Here, we report that the NF-κB inducing IKK kinase complex is localized at the postsynaptic density (PSD) and activated under basal conditions in the adult mouse brain. Using different models of conditional genetic inactivation of IKK2 function in mouse principal neurons, we show that IKK/NF-κB signaling is critically involved in synapse formation and spine maturation in the adult brain. IKK/NF-κB blockade in the forebrain of mutant animals is associated with reduced levels of mature spines and postsynaptic proteins PSD95, SAP97, GluA1, AMPAR-mediated basal synaptic transmission and a spatial learning impairment. Synaptic deficits can be restored in adult animals within 1 week by IKK/NF-κB reactivation, indicating a highly dynamic IKK/NF-κB-dependent regulation process. We further identified the insulin-like growth factor 2 gene (Igf2) as a novel IKK/NF-κB target. Exogenous Igf2 was able to restore synapse density and promoted spine maturation in IKK/NF-κB signaling-deficient neurons within 24 h. This process depends on Igf2/Igf2R-mediated MEK/ERK activation. Our findings illustrate a fundamental role of IKK/NF-κB-Igf2-Igf2R signaling in synapse formation and maturation in adult mice, thus providing an intriguing link between the molecular actions of IKK/NF-κB in neurons and the memory enhancement factor Igf2.


Subject(s)
I-kappa B Kinase/metabolism , Insulin-Like Growth Factor II/metabolism , NF-kappa B/metabolism , Neurons/cytology , Signal Transduction/physiology , Synapses/physiology , Adaptor Proteins, Signal Transducing/metabolism , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Dendritic Spines , Discs Large Homolog 1 Protein , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Electrophoretic Mobility Shift Assay , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation, Developmental/genetics , Guanylate Kinases/metabolism , Hippocampus/cytology , I-kappa B Kinase/genetics , In Vitro Techniques , Insulin-Like Growth Factor II/genetics , Learning Disabilities/genetics , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Maze Learning/drug effects , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Silver Staining , Synapses/drug effects , Synapses/ultrastructure
6.
Glia ; 60(6): 867-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22419561

ABSTRACT

Little is known about the physiological roles of aquaporin-4 (AQP4) in the central nervous system. AQP4 water channels are concentrated in endfeet membranes of astrocytes but also localize to the fine astrocytic processes that abut central synapses. Based on its pattern of expression, we predicted that AQP4 could be involved in controlling water fluxes and changes in extracellular space (ECS) volume that are associated with activation of excitatory pathways. Here, we show that deletion of Aqp4 accentuated the shrinkage of the ECS that occurred in the mouse hippocampal CA1 region during activation of Schaffer collateral/commissural fibers. This effect was found in the stratum radiatum (where perisynaptic astrocytic processes abound) but not in the pyramidal cell layer (where astrocytic processes constitute but a minor volume fraction). For both genotypes the ECS shrinkage was most pronounced in the pyramidal cell layer. Our data attribute a physiological role to AQP4 and indicate that this water channel regulates extracellular volume dynamics in the mammalian brain.


Subject(s)
Aquaporin 4/deficiency , Astrocytes/physiology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/genetics , Extracellular Space/genetics , Animals , Astrocytes/ultrastructure , Biophysical Phenomena , CA1 Region, Hippocampal/ultrastructure , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Ganglionic Stimulants/pharmacology , Glial Fibrillary Acidic Protein/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Patch-Clamp Techniques , Phosphopyruvate Hydratase/metabolism , Pyramidal Cells/physiology , Quaternary Ammonium Compounds/pharmacology , Synapses/genetics , Synapses/ultrastructure
7.
Cereb Cortex ; 22(8): 1786-98, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21955919

ABSTRACT

Dopamine plays an important role in synaptic plasticity and learning and is involved in the pathogenesis of various neurological and psychiatric disorders. Here, we reveal staining of dopaminergic fibers in stratum oriens of the mouse hippocampal CA1 region, a finding that is consistent with earlier reports. Furthermore, we examined the effect of dopamine agonists on NMDAR-dependent early long-term potentiation (LTP) (40 min) during γ-aminobutyric acid (GABA)(A)-mediated blockade. LTP of the AMPA component was strongly reduced in stratum oriens but barely affected in stratum radiatum. This layer-specific effect was caused by D4 receptor activation, which augmented the inactivation of synaptic NMDAR-mediated currents (NMDA EPSCs) during LTP induction through a Ca(2+)-dependent G-protein-independent mechanism. A similar dopaminergic modulation of both NMDA EPSCs and LTP was also observed in mice constitutively lacking NR2A but was absent in mice lacking NR2B in principal forebrain neurons. Together, these experiments strongly indicate that dopaminergic modulation of early LTP in stratum oriens occurs through NMDARs containing NR2B subunits via D4Rs. Thus, a dopamine hyperfunction in stratum oriens may result in NMDAR hypofunction that could affect both normal and pathological conditions.


Subject(s)
CA1 Region, Hippocampal/physiology , Long-Term Potentiation/physiology , Receptors, Dopamine D4/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Excitatory Postsynaptic Potentials/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques
8.
Semin Cell Dev Biol ; 22(4): 400-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21827868

ABSTRACT

The synaptic vesicle-associated synapsin proteins may participate in synaptic transmission, but their exact functional role(s) here remain(s) uncertain. We here briefly describe the important characteristics of the synapsin proteins, and review recent studies on transgenic mice devoid of the gene products encoded by the synapsin I and II genes, where both neurochemical, cell biological and electrophysiological methods have been employed. We present evidence for synapsin effects on both neurotransmitter synthesis and homeostasis, as well as on synaptic vesicle development and functions. Moreover, we describe physiological analyses of excitatory glutamatergic hippocampal synapses where a novel synapsin-dependent delayed response enhancement (DRE) phase occurs, and demonstrate the postnatal developmental patterns of both frequency facilitations and DRE responses. Finally, we report synapsin I and II effects in distinct excitatory glutamatergic synapses in the hippocampus, and indicate that synapsin-dependent modulations of synaptic function may use distinct presynaptic response patterns in order to induce different classes of presynaptic plasticity.


Subject(s)
Synapsins/genetics , Synapsins/metabolism , Synaptic Transmission , Animals , Excitatory Amino Acid Agents/metabolism , Mice , Mice, Knockout , Synapses/metabolism
9.
Glia ; 59(11): 1635-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21748805

ABSTRACT

Mutations in the human Kir4.1 potassium channel gene (KCNJ10) are associated with epilepsy. Using a mouse model with glia-specific deletion of Kcnj10, we have explored the mechanistic underpinning of the epilepsy phenotype. The gene deletion was shown to delay K(+) clearance after synaptic activation in stratum radiatum of hippocampal slices. The activity-dependent changes in extracellular space volume did not differ between Kcnj10 mutant and wild-type mice, indicating that the Kcnj10 gene product Kir4.1 mediates osmotically neutral K(+) clearance. Combined, our K(+) and extracellular volume recordings indicate that compromised K(+) spatial buffering in brain underlies the epilepsy phenotype associated with human KCNJ10 mutations.


Subject(s)
Epilepsy/genetics , Potassium Channels, Inwardly Rectifying/genetics , Potassium/physiology , Animals , Aquaporin 4/genetics , Blotting, Western , Buffers , Electric Stimulation , Electrophoresis , Fluorescent Antibody Technique , Gold , Hippocampus/pathology , Humans , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Immunoelectron , Mutation/genetics , Mutation/physiology , Neuroglia/enzymology , Seizures/genetics , Seizures/physiopathology
10.
Article in English | MEDLINE | ID: mdl-21423538

ABSTRACT

Repeated release of transmitter from presynaptic elements depends on stimulus-induced Ca(2+) influx together with recruitment and priming of synaptic vesicles from different vesicle pools. We have compared three different manipulations of synaptic strength, all of which are known to increase short-term synaptic efficacy through presynaptic mechanisms, in the glutamatergic CA3-to-CA1 stratum radiatum synapse in the mouse hippocampal slice preparation. Synaptic responses elicited from the readily releasable vesicle pool during low-frequency synaptic activation (0.1 Hz) were significantly enhanced by both the adenylate cyclase activator forskolin, the priming activator ß-phorbol-12,13-dibutyrate (PDBu) and 4 mM [Ca(2+)](o') whereas during 20 Hz stimulation, the same manipulations reduced the time needed to reach the peak and increased the magnitude of the resulting frequency facilitation. In contrast, paired-pulse facilitations were unchanged in the presence of forskolin, decreased by 4 mM [Ca(2+)](o) and essentially abolished by PDBu. The subsequent delayed response enhancement (DRE) responses, elicited during continuous 20 Hz stimulations and mediated by recruited vesicles, were enhanced by forskolin, essentially unchanged by PDBu and slightly decreased by 4 mM [Ca(2+)](o·) Similar experiments done on slices devoid of the vesicle-associated synapsin I and II proteins indicated that synapsin I/II-induced enhancements of vesicle recruitment were restricted to Ca(2+)-induced frequency facilitations and forskolin-induced enhancements of the early DRE phase, whereas the proteins had minor effects during PDBu-treatment and represented constraints on late Ca(2+)-induced responses. The data indicate that in these glutamatergic synapses, the comparable enhancements of single synaptic responses induced by these biochemical mechanisms can be transformed during prolonged synaptic stimulation into highly distinct short-term plasticity patterns, which are partly dependent on synapsins I/II.

11.
Neuropharmacology ; 57(7-8): 619-26, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19698722

ABSTRACT

Although several molecular and genetic manipulations may produce hyperactive animals, hyperactivity alone is insufficient for the animal to qualify as a model of ADHD. Based on a wider range of criteria - behavioral, genetic and neurobiological - the spontaneously hypertensive rat (SHR) obtained from Charles River, Germany (SHR/NCrl) at present constitutes the best validated animal model of ADHD combined subtype (ADHD-C), and the Wistar Kyoto substrain obtained from Harlan, UK (WKY/NHsd) is its most appropriate control. Although other rat strains may behave like WKY/NHsd rats, genetic results indicate significant differences when compared to the WKY/NHsd substrain, making them less suitable controls for the SHR/NCrl. The use of WKY/NCrl, outbred Wistar, Sprague Dawley or other rat strains as controls for SHRs may produce spurious neurobiological differences. Consequently, data may be misinterpreted if insufficient care is taken in the selection of the control group. It appears likely that the use of different control strains may underlie some of the discrepancies in results and interpretations in studies involving the SHR and WKY. Finally, we argue that WKY rats obtained from Charles River, Germany (WKY/NCrl) provide a promising model for the predominantly inattentive subtype of ADHD (ADHD-PI); in this case also the WKY/NHsd substrain should be used as control.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Disease Models, Animal , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit Disorder with Hyperactivity/physiopathology , Attention Deficit Disorder with Hyperactivity/psychology , Neuronal Plasticity/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Reinforcement, Psychology , Species Specificity
12.
Cereb Cortex ; 19(3): 511-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18550596

ABSTRACT

The synapsin proteins have different roles in excitatory and inhibitory synaptic terminals. We demonstrate a differential role between types of excitatory terminals. Structural and functional aspects of the hippocampal mossy fiber (MF) synapses were studied in wild-type (WT) mice and in synapsin double-knockout mice (DKO). A severe reduction in the number of synaptic vesicles situated more than 100 nm away from the presynaptic membrane active zone was found in the synapsin DKO animals. The ultrastructural level gave concomitant reduction in F-actin immunoreactivity observed at the periactive endocytic zone of the MF terminals. Frequency facilitation was normal in synapsin DKO mice at low firing rates (approximately 0.1 Hz) but was impaired at firing rates within the physiological range (approximately 2 Hz). Synapses made by associational/commissural fibers showed comparatively small frequency facilitation at the same frequencies. Synapsin-dependent facilitation in MF synapses of WT mice was attenuated by blocking F-actin polymerization with cytochalasin B in hippocampal slices. Synapsin III, selectively seen in MF synapses, is enriched specifically in the area adjacent to the synaptic cleft. This may underlie the ability of synapsin III to promote synaptic depression, contributing to the reduced frequency facilitation observed in the absence of synapsins I and II.


Subject(s)
Actins/physiology , Mossy Fibers, Hippocampal/physiology , Synapses/physiology , Synapsins/physiology , Actins/deficiency , Animals , Excitatory Postsynaptic Potentials/physiology , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/ultrastructure , Synapses/ultrastructure , Synapsins/deficiency
13.
Neuron ; 60(5): 846-60, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19081379

ABSTRACT

Controversy revolves around the differential contribution of NR2A- and NR2B-containing NMDA receptors, which coexist in principal forebrain neurons, to synaptic plasticity and learning in the adult brain. Here, we report genetically modified mice in which the NR2B subunit is selectively ablated in principal neurons of the entire postnatal forebrain or only the hippocampus. NR2B ablation resulted in smaller NMDA receptor-mediated EPSCs with accelerated decay kinetics, as recorded in CA1 pyramidal cells. CA3-to-CA1 field LTP remained largely unaltered, although a pairing protocol revealed decreased NMDA receptor-mediated charge transfer and reduced cellular LTP. Mice lacking NR2B in the forebrain were impaired on a range of memory tasks, presenting both spatial and nonspatial phenotypes. In contrast, hippocampus-specific NR2B ablation spared hippocampus-dependent, hidden-platform water maze performance but induced a selective, short-term, spatial working memory deficit for recently visited places. Thus, both hippocampal and extra-hippocampal NR2B containing NMDA receptors critically contribute to spatial performance.


Subject(s)
Hippocampus/cytology , Maze Learning/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Analysis of Variance , Animals , Behavior, Animal , Discrimination, Psychological/physiology , Electric Stimulation/methods , Electron Transport Complex IV/metabolism , Evoked Potentials/physiology , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , In Vitro Techniques , Male , Maze Learning/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Patch-Clamp Techniques/methods , Photic Stimulation/methods , Receptors, N-Methyl-D-Aspartate/genetics , Recognition, Psychology/physiology , Space Perception/drug effects , Space Perception/physiology
14.
J Neurosci ; 27(41): 10947-56, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17928436

ABSTRACT

The AMPA receptor subunit glutamate receptor 1 (GluR1 or GluR-A) contributes to amygdala-dependent emotional learning. It remains unclear, however, to what extent different amygdala pathways depend on GluR1, or other AMPA receptor subunits, for proper synaptic transmission and plasticity, and whether GluR1-dependent long-term potentiation (LTP) is necessary for auditory and contextual fear conditioning. Here, we dissected the role of GluR1 and GluR3 (GluR-C) subunits in AMPA receptor-dependent amygdala LTP and fear conditioning using knock-out mice (GluR1-/- and GluR3-/-). We found that, whereas LTP at thalamic inputs to lateral amygdala (LA) projection neurons and at glutamatergic synapses in the basal amygdala was completely absent in GluR1-/- mice, both GluR1 and GluR3 contributed to LTP in the cortico-LA pathway. Because both auditory and contextual fear conditioning were selectively impaired in GluR1-/- but not GluR3-/- mice, we conclude that GluR1-dependent synaptic plasticity is the dominant form of LTP underlying the acquisition of auditory and contextual fear conditioning, and that plasticity in distinct amygdala pathways differentially contributes to aversive conditioning.


Subject(s)
Amygdala/physiology , Conditioning, Psychological/physiology , Fear/physiology , Long-Term Potentiation/physiology , Receptors, AMPA/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/physiology , Protein Subunits/physiology
15.
J Cell Sci ; 120(Pt 17): 3034-44, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17684059

ABSTRACT

Proline-rich tyrosine kinase 2 (PYK2) is a non-receptor tyrosine kinase expressed in many cell types and enriched in neurons. PYK2 is a cytoplasmic enzyme activated by increases in cytosolic free Ca(2+) through an unknown mechanism. We report that depolarization or electrical stimulation of hippocampal slices induced a rapid and transient nuclear accumulation of PYK2. Depolarization of cultured neurons or PC12 cells also triggered a Ca(2+)-dependent nuclear accumulation of PYK2, much more pronounced than that induced by blockade of nuclear export with leptomycin B. Src-family kinase activity, PYK2 autophosphorylation and kinase activity were not required for its nuclear translocation. Depolarization induced a slight decrease in PYK2 apparent molecular mass, compatible with a Ca(2+)-activated dephosphorylation. Pretreatment of PC12 cells with inhibitors of calcineurin (protein phosphatase 2B), cyclosporin A and FK506, prevented depolarization-induced nuclear translocation and tyrosine phosphorylation of PYK2. Transfection with dominant-negative and constitutively active calcineurin-A confirmed the role of calcineurin in the regulation of PYK2 tyrosine phosphorylation and nuclear accumulation. Our results show that depolarization independently induces nuclear translocation and tyrosine phosphorylation of PYK2, and that both responses require calcineurin activation. We suggest that PYK2 exerts some of its actions in the nucleus and that the effects of calcineurin inhibitors may involve PYK2 inhibition.


Subject(s)
Calcineurin/metabolism , Cell Nucleus/metabolism , Focal Adhesion Kinase 2/metabolism , Neurons/metabolism , Tyrosine/metabolism , Active Transport, Cell Nucleus , Animals , Calcineurin/genetics , Calcium/metabolism , Cells, Cultured , Electric Stimulation , Focal Adhesion Kinase 2/genetics , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Neurons/cytology , PC12 Cells , Phosphorylation , Rats , Rats, Sprague-Dawley
16.
J Physiol ; 583(Pt 1): 129-43, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17569738

ABSTRACT

High frequency afferent stimulation of chemical synapses often induces short-term increases in synaptic efficacy, due to increased release probability and/or increased supply of readily releasable synaptic vesicles. This may be followed by synaptic depression, often caused by vesicle depletion. We here describe an additional, novel type of delayed and transient response enhancement phase which occurred during prolonged stimulation at 5-20 Hz frequency of excitatory glutamatergic synapses in slices from the adult mouse CA1 hippocampal region. This second enhancement phase, which was most clearly defined at physiological temperatures and essentially absent at 24 degrees C, was dependent on the presence of F-actin filaments and synapsins I and/or II, and could not be ascribed to changes in presynaptic action potentials, inhibitory neurotransmission or glutamate receptor desensitization. Time course studies showed that the delayed response phase interrupted the synaptic decay 3-4 s after stimulus train initiation and continued, when examined at 5-10 Hz frequencies, for approximately 75 stimuli before decay. The novel response enhancement, probably deriving from a restricted pool of synaptic vesicles, may allow maintenance of synaptic efficacy during prolonged periods of excitatory synaptic activity.


Subject(s)
Hippocampus/physiology , Neuronal Plasticity/physiology , Presynaptic Terminals/physiology , Synaptic Vesicles/physiology , Actins/metabolism , Action Potentials/physiology , Animals , Calcium/physiology , Electric Stimulation , Mice , Mice, Knockout , Patch-Clamp Techniques , Receptors, GABA/physiology , Synapses/metabolism , Synapsins/genetics , Synapsins/metabolism , Synaptic Transmission/physiology , Temperature
17.
Neuropharmacology ; 52(1): 77-86, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16901514

ABSTRACT

Activation of NMDA receptors (NMDARs) is a requirement for persistent synaptic alterations, such as long-term potentiation of synaptic transmission (LTP). NMDARs are composed of NR1 and NR2 subunits, and NR2 subunit-dependent gating properties of NMDAR subtypes cause dramatic differences in the timing of charge transfer. These postsynaptic temporal profiles are further influenced by the frequency of synaptic activation. Here, we investigated in the CA1 region of hippocampal slices from P28 mice, whether particular NMDAR subtypes are recruited based on NR2 subunit-specific gating following different induction protocols. For high frequency afferent stimulation (HFS), we found that genetic impairment of NR2A or pharmacological block of NR2A- or NR2B-type NMDARs can reduce field LTP. In contrast, when pairing low frequency synaptic stimulation with postsynaptic depolarization (LFS pairing) in single CA1 neurons, pharmacological antagonism of either subtype modestly reduced the charge transfer during LFS pairing without reducing the LTP magnitude. These results indicate that HFS-triggered LTP is induced by more than one NMDAR subtype, whereas a single subtype is sufficient during LFS pairing. Analysis of charge transfer during LFS pairing in 13 different conditions revealed a threshold for LTP induction, which was independent of the NR2 antagonist tested. Thus, at least for LFS pairing, the amount of charge transfer, and thus Ca2+ influx, during LTP induction is a factor more critical than the participation of a particular NMDAR subtype.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Long-Term Potentiation/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Neurons/radiation effects , Patch-Clamp Techniques/methods , Receptors, N-Methyl-D-Aspartate/deficiency
18.
J Neurosci ; 26(33): 8428-40, 2006 Aug 16.
Article in English | MEDLINE | ID: mdl-16914668

ABSTRACT

We demonstrate the fundamental importance of glutamate receptor B (GluR-B) containing AMPA receptors in hippocampal function by analyzing mice with conditional GluR-B deficiency in postnatal forebrain principal neurons (GluR-B(deltaFb)). These mice are as adults sufficiently robust to permit comparative cellular, physiological, and behavioral studies. GluR-B loss induced moderate long-term changes in the hippocampus of GluR-B(deltaFb) mice. Parvalbumin-expressing interneurons in the dentate gyrus and the pyramidal cells in CA3 were decreased in number, and neurogenesis in the subgranular zone was diminished. Excitatory synaptic CA3-to-CA1 transmission was reduced, although synaptic excitability, as quantified by the lowered threshold for population spike initiation, was increased compared with control mice. These changes did not alter CA3-to-CA1 long-term potentiation (LTP), which in magnitude was similar to LTP in control mice. The altered hippocampal circuitry, however, affected spatial learning in GluR-B(deltaFb) mice. The primary source for the observed changes is most likely the AMPA receptor-mediated Ca2+ signaling that appears after GluR-B depletion, because we observed similar alterations in GluR-B(QFb) mice in which the expression of Ca2+-permeable AMPA receptors in principal neurons was induced by postnatal activation of a Q/R-site editing-deficient GluR-B allele.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Memory/physiology , Prosencephalon/metabolism , Receptors, AMPA/physiology , Space Perception/physiology , Animals , Calcium/metabolism , Cell Division , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Exploratory Behavior , Gene Silencing , Hippocampus/cytology , Maze Learning/physiology , Mice , Mice, Knockout , Neuronal Plasticity , Neurons/cytology , Neurons/metabolism , Receptors, AMPA/deficiency , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synaptic Transmission
19.
J Physiol ; 571(Pt 1): 75-82, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16322053

ABSTRACT

The effects of synapsin proteins on synaptic transmission from vesicles in the readily releasable vesicle pool have been examined by comparing excitatory synaptic transmission in hippocampal slices from mice devoid of synapsins I and II and from wild-type control animals. Application of stimulus trains at variable frequencies to the CA3-to-CA1 pyramidal cell synapse suggested that, in both genotypes, synaptic responses obtained within 2 s stimulation originated from readily releasable vesicles. Detailed analysis of the responses during this period indicated that stimulus trains at 2-20 Hz enhanced all early synaptic responses in the CA3-to-CA1 pyramidal cell synapse, but depressed all early responses in the medial perforant path-to-granule cell synapse. The synapsin-dependent part of these responses, i.e. the difference between the results obtained in the transgene and the wild-type preparations, showed that in the former synapse, the presence of synapsins I and II minimized the early responses at 2 Hz, but enhanced the early responses at 20 Hz, while in the latter synapse, the presence of synapsins I and II enhanced all responses at both stimulation frequencies. The results indicate that synapsins I and II are necessary for full expression of both enhancing and decreasing modulatory effects on synaptic transmission originating from the readily releasable vesicles in these excitatory synapses.


Subject(s)
Hippocampus/physiology , Synapses/physiology , Synapsins/physiology , Synaptic Transmission , Synaptic Vesicles/physiology , Animals , Dentate Gyrus/physiology , Electric Stimulation , Excitatory Postsynaptic Potentials , Mice , Mice, Knockout , Pyramidal Cells/physiology
20.
J Neurosci ; 25(29): 6907-10, 2005 Jul 20.
Article in English | MEDLINE | ID: mdl-16033900

ABSTRACT

NMDA receptor (NMDAR) 2A (NR2A)- and NR2B-type NMDARs coexist in synapses of CA1 pyramidal cells. Recent studies using pharmacological blockade of NMDAR subtypes proposed that the NR2A type is responsible for inducing long-term potentiation (LTP), whereas the NR2B type induces long-term depression (LTD). This contrasts with the finding in genetically modified mice that NR2B-type NMDARs induce LTP when NR2A signaling is absent or impaired, although compensatory mechanisms might have contributed to this result. We therefore assessed the contribution of the two NMDAR subtypes to LTP in mouse hippocampal slices by different induction protocols and in the presence of NMDAR antagonists, including the NR2A-type blocker NVP-AAM077, for which an optimal concentration for subtype selectivity was determined on recombinant and native NMDARs. Partial blockade of NMDA EPSCs by 40%, either by preferentially antagonizing NR2A- or NR2B-type NMDARs or by the nonselective antagonist D-AP-5, did not impair LTP, demonstrating that hippocampal LTP induction can be generated by either NMDAR subtype.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cell Line , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Humans , Kidney/cytology , Mice , Organ Culture Techniques , Piperidines/pharmacology , Pyramidal Cells/physiology , Quinoxalines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL