Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Hippocampus ; 22(1): 57-68, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20848601

ABSTRACT

CA3 pyramidal cells receive three main excitatory inputs: the first one is the mossy fiber input, synapsing mainly on the proximal apical dendrites. Second, entorhinal cortex cells form excitatory connections with CA3 pyramidal cells via the perforant path in the stratum lacunosum moleculare. The third input involves the ipsi-and contralateral connections, termed the associational/commissural (A/C) pathway terminating in the stratum radiatum of CA3, thus forming a feedback loop within this region. Since this excitatory recurrent synapse makes the CA3 region extremely prone to seizure development, understanding the regulation of synaptic strength of this connection is of crucial interest. Several studies suggest that kainate receptors (KAR) play a role in the regulation of synaptic strength. Our aim was to characterize the influence of KAR on A/C synaptic transmission: application of ATPA, a selective agonist of the GluK1 KAR, depressed the amplitude fEPSP without affecting the size of the fiber volley. Blockade of GABA receptors had no influence on this effect, arguing against the influence of interneuronal KARs. Pharmacological and genetic deletion studies could show that this effect was selectively due to GluK1 receptor activation. Several lines of evidence, such as PPF changes, coefficient of variance-analysis and glutamate uncaging experiments strongly argue for a presynaptic locus of suppression. This is accompanied by an ATPA-mediated reduction in Ca(2+) influx at excitatory synaptic terminals, which is most likely mediated by a G-Protein dependent mechanism, as suggested by application of pertussis toxin. Finally, analysis of miniature EPSCs in the presence and absence of extracellular Ca(2+) suggest that presynaptic KAR can also reduce transmitter release downstream and therefore independent of Ca(2+) influx.


Subject(s)
CA3 Region, Hippocampal/physiology , Calcium Signaling/physiology , Neurotransmitter Agents/metabolism , Presynaptic Terminals/physiology , Receptors, Kainic Acid/physiology , Synaptic Transmission/physiology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/metabolism , Calcium Signaling/drug effects , Mice , Mice, Knockout , Organ Culture Techniques/methods , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Kainic Acid/agonists , Receptors, Kainic Acid/deficiency , Synaptic Transmission/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
2.
Behav Brain Res ; 228(2): 406-14, 2012 Mar 17.
Article in English | MEDLINE | ID: mdl-22203159

ABSTRACT

There is a clear link between dysregulation of glutamatergic signaling and mood disorders. Genetic variants in the glutamate receptor gene GRIK4, which encodes the kainate receptor subunit GluK4, alter the susceptibility for depression, bipolar disorder and schizophrenia. Here we demonstrate that Grik4(-/-) mice have reduced anxiety and an antidepressant-like phenotype. In the elevated zero-maze, a test for anxiety and risk taking behavior, Grik4(-/-) mice spent significantly more time exploring the open areas of the maze. In anxiogenic tests of marble-burying and novelty-induced suppression of feeding, anxiety-like behavior was consistently reduced in knockout animals. In the forced swim test, a test of learned helplessness that is used to determine depression-like behavior, knockout mice demonstrated significantly less immobility suggesting that Grik4 ablation has an antidepressant-like effect. Finally, in the sucrose preference test, a test for anhedonia in rodents, Grik4(-/-) mice demonstrated increased sucrose preference. Expression of the GluK4 receptor subunit in the forebrain is restricted to the CA3 region of the hippocampus and dentate gyrus regions where KARs are known to modulate synaptic plasticity. We tested whether Grik4 ablation had effects on mossy fiber (MF) plasticity and found there to be a significant impairment in LTP likely through a loss of KAR modulation of excitability of the presynaptic MF axons. These studies demonstrate a clear anxiolytic and antidepressant phenotype associated with ablation of Grik4 and a parallel disruption in hippocampal plasticity, providing support for the importance of this receptor subunit in mood disorders.


Subject(s)
Anxiety/genetics , Depression/genetics , Receptors, Kainic Acid/deficiency , Analysis of Variance , Animals , Anxiety/physiopathology , Behavior, Animal , Depression/physiopathology , Disease Models, Animal , Electric Stimulation , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/physiology , Food Preferences/physiology , Hippocampus/physiology , In Vitro Techniques , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Maze Learning/physiology , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/physiology , Motor Activity/genetics , Patch-Clamp Techniques , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Swimming/psychology
3.
Nat Neurosci ; 14(7): 866-73, 2011 May 29.
Article in English | MEDLINE | ID: mdl-21623363

ABSTRACT

Ionotropic glutamate receptors principally mediate fast excitatory transmission in the brain. Among the three classes of ionotropic glutamate receptors, kainate receptors (KARs) have a unique brain distribution, which has been historically defined by (3)H-radiolabeled kainate binding. Compared with recombinant KARs expressed in heterologous cells, synaptic KARs exhibit characteristically slow rise-time and decay kinetics. However, the mechanisms responsible for these distinct KAR properties remain unclear. We found that both the high-affinity binding pattern in the mouse brain and the channel properties of native KARs are determined by the KAR auxiliary subunit Neto1. Through modulation of agonist binding affinity and off-kinetics of KARs, but not trafficking of KARs, Neto1 determined both the KAR high-affinity binding pattern and the distinctively slow kinetics of postsynaptic KARs. By regulating KAR excitatory postsynaptic current kinetics, Neto1 can control synaptic temporal summation, spike generation and fidelity.


Subject(s)
CA1 Region, Hippocampal/metabolism , Cerebellum/metabolism , Lipoproteins, LDL/metabolism , Membrane Proteins/metabolism , Receptors, Kainic Acid/physiology , Animals , Animals, Newborn , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Biophysics , CA1 Region, Hippocampal/cytology , Cell Line, Transformed , Cerebellum/cytology , Disks Large Homolog 4 Protein , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Excitatory Amino Acid Agonists/pharmacokinetics , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Guanylate Kinases , Humans , Immunoprecipitation , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Kainic Acid/pharmacokinetics , Kainic Acid/pharmacology , LDL-Receptor Related Proteins , Lipoproteins, LDL/deficiency , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Neurons/classification , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques/methods , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Protein Binding/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Kainic Acid/classification , Receptors, Kainic Acid/deficiency , Receptors, N-Methyl-D-Aspartate , Synaptophysin/metabolism , Transfection/methods
4.
J Neurosci ; 30(24): 8203-13, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20554871

ABSTRACT

The central nucleus of the amygdala (CeA) has been identified as a site of nociceptive processing important for sensitization induced by peripheral injury. However, the cellular signaling components underlying this function remain unknown. Here, we identify metabotropic glutamate receptor 5 (mGluR5) as an integral component of nociceptive processing in the CeA. Pharmacological activation of mGluRs with (R,S)-3,5-dihydroxyphenylglycine (DHPG) in the CeA of mice is sufficient to induce peripheral hypersensitivity in the absence of injury. DHPG-induced peripheral hypersensitivity is reduced via pharmacological blockade of mGluR5 or genetic disruption of mGluR5. Furthermore, pharmacological blockade or conditional deletion of mGluR5 in the CeA abrogates inflammation-induced hypersensitivity, demonstrating the necessity of mGluR5 in CeA-mediated pain modulation. Moreover, we demonstrate that phosphorylation of extracellular-signal regulated kinase 1/2 (ERK1/2) is downstream of mGluR5 activation in the CeA and is necessary for the full expression of peripheral inflammation-induced behavioral sensitization. Finally, we present evidence of right hemispheric lateralization of mGluR5 modulation of amygdalar nociceptive processing. We demonstrate that unilateral pharmacological activation of mGluR5 in the CeA produces distinct behavioral responses depending on whether the right or left amygdala is injected. We also demonstrate significantly higher levels of mGluR5 expression in the right amygdala compared with the left under baseline conditions, suggesting a potential mechanism for right hemispheric lateralization of amygdala function in pain processing. Together, these results establish an integral role for mGluR5 and ERK1/2 in nociceptive processing in the CeA.


Subject(s)
Amygdala/metabolism , Pain/physiopathology , Receptors, Kainic Acid/metabolism , Amygdala/drug effects , Analysis of Variance , Animals , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Formaldehyde , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/metabolism , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Pain/chemically induced , Pain/genetics , Pain Measurement/methods , Pyridines/pharmacology , Receptors, Glucocorticoid/deficiency , Receptors, Kainic Acid/deficiency
5.
J Neurosci ; 30(19): 6507-14, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463214

ABSTRACT

In the neonatal hippocampus, the activity of interneurons shapes early network bursts that are important for the establishment of neuronal connectivity. However, mechanisms controlling the firing of immature interneurons remain elusive. We now show that the spontaneous firing rate of CA3 stratum lucidum interneurons markedly decreases during early postnatal development because of changes in the properties of GluK1 (formerly known as GluR5) subunit-containing kainate receptors (KARs). In the neonate, activation of KARs by ambient glutamate exerts a tonic inhibition of the medium-duration afterhyperpolarization (mAHP) by a G-protein-dependent mechanism, permitting a high interneuronal firing rate. During development, the amplitude of the apamine-sensitive K+ currents responsible for the mAHP increases dramatically because of decoupling between KAR activation and mAHP modulation, leading to decreased interneuronal firing. The developmental shift in the KAR function and its consequences on interneuronal activity are likely to have a fundamental role in the maturation of the synchronous neuronal oscillations typical for adult hippocampal circuitry.


Subject(s)
Action Potentials/physiology , CA3 Region, Hippocampal/growth & development , CA3 Region, Hippocampal/physiology , Interneurons/physiology , Potassium/metabolism , Receptors, Kainic Acid/metabolism , Animals , Animals, Newborn , GTP-Binding Proteins/metabolism , Glutamic Acid/metabolism , In Vitro Techniques , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/physiology , Patch-Clamp Techniques , Receptors, Kainic Acid/deficiency , Receptors, Kainic Acid/genetics
6.
Neuroscience ; 169(1): 475-87, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20359526

ABSTRACT

Several groups maintain that morphine tolerance and dependence correlate with increased activity of protein kinases ERK1/2 and P38 MAPK and PKC as well as elevated levels of the neuropeptides dynorphin (DYN), substance P (sP), and calcitonin gene-related peptide (CGRP) in spinal cord dorsal horn (SCDH). They demonstrate that tolerance and dependence can be prevented, and sometimes reversed, by constitutive genetic deletion or pharmacological inhibition of these factors. Recently, we showed that mice with a constitutive deletion of the GluR5 subunit of kainate receptors (GluR5 KO) are not different from wild type (WT) littermates with respect to baseline nociceptive thresholds as well as acute morphine antinociception, morphine physical dependence and conditioned place preference. However, unlike WT, GluR5 KO mice do not develop antinociceptive tolerance following systemic morphine administration. In this report, we examined levels of these mediators in SCDH of WT and GluR5 KO mice following subcutaneous implantation of placebo or morphine pellets. Surprisingly, spinal DYN and CGRP, along with phosphorylated ERK2 (pERK2), P38 (pP38) and PKCgamma (pPKCgamma) are elevated by deletion of GluR5. Additionally, chronic systemic morphine administration increased spinal pERK2, pP38 and pPKCgamma levels in both tolerant WT and non-tolerant GluR5 KO mice. In contrast, while morphine increased spinal DYN and CGRP in WT mice, DYN remained unchanged and CGRP was reduced in GluR5 KO mice. These observations suggest that spinal ERK2, P38 and PKCgamma are likely involved in multiple adaptive responses following systemic morphine administration, whereas DYN and CGRP may contribute selectively to the development of antinociceptive tolerance.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Dynorphins/physiology , Mitogen-Activated Protein Kinase 1/physiology , Morphine/pharmacology , Narcotics/pharmacology , Pain Threshold/physiology , Pain/physiopathology , Posterior Horn Cells/metabolism , Protein Kinase C/physiology , Receptors, Kainic Acid/deficiency , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Calcitonin Gene-Related Peptide/biosynthesis , Calcitonin Gene-Related Peptide/genetics , Drug Implants , Drug Tolerance/physiology , Dynorphins/biosynthesis , Dynorphins/genetics , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/genetics , Morphine/administration & dosage , Morphine/therapeutic use , Morphine/toxicity , Morphine Dependence/physiopathology , Narcotics/administration & dosage , Narcotics/therapeutic use , Narcotics/toxicity , Pain/drug therapy , Pain Threshold/drug effects , Phosphorylation , Posterior Horn Cells/drug effects , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Protein Processing, Post-Translational , Receptors, Kainic Acid/genetics , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/genetics
7.
Biol Psychiatry ; 67(9): 864-71, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20034613

ABSTRACT

BACKGROUND: Abnormal hedonic behavior is a key feature of many psychiatric disorders. Several paradigms measure reward-seeking behavior in rodents, but each has limitations. We describe a novel approach for monitoring reward-seeking behavior in rodents: sniffing of estrus female urine by male mice, along with number of ultrasonic vocalizations (USVs) emitted during the test. METHODS: The female urine sniffing test (FUST) was designed to monitor reward-seeking activity in rodents together with tests of helplessness and sweet solution preference. USVs and dopamine release from the nucleus accumbens (NAc) were recorded. Sniffing activity was measured in 1) manipulation-naive C57BL/6J and 129S1/SVImJ mice and Wistar-Kyoto rats; 2) stressed mice; 3) two groups of mice that underwent the learned helplessness paradigm-one untreated, and one treated with the SSRI citalopram; and 4) GluR6 knockout mice, known to display lithium-responsive, mania-related behaviors. RESULTS: Males from all three strains spent significantly longer sniffing female urine than sniffing water. Males emitted USVs and showed significantly elevated NAc dopamine levels while sniffing urine. Foot-shock stress significantly reduced female urine sniffing time. Compared with mice that did not undergo the LH paradigm, LH males spent less time sniffing female urine, and citalopram treatment alleviated this reduction. Compared with their wildtype littermates, GluR6KO males sniffed female urine longer and showed enhanced saccharin preference. CONCLUSIONS: In rodents, sniffing female urine is a preferred activity accompanied by biological changes previously linked to reward-seeking activities. The FUST is sensitive to behavioral and genetic manipulation and to relevant drug treatment.


Subject(s)
Exploratory Behavior/physiology , Reward , Smell/physiology , Urine , Vocalization, Animal , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal , Citalopram/pharmacology , Dopamine/metabolism , Electroshock/methods , Female , Food Preferences/drug effects , Food Preferences/physiology , Lithium/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis/methods , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/metabolism , Rats , Rats, Inbred WKY , Receptors, Kainic Acid/deficiency , Saccharin/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Species Specificity , Sweetening Agents/pharmacology , Ultrasonics , GluK2 Kainate Receptor
8.
Neuron ; 63(6): 818-29, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19778510

ABSTRACT

Kainate receptors signal through both ionotropic and metabotropic pathways. The high-affinity subunits, GluK4 and GluK5, are unique among the five receptor subunits, as they do not form homomeric receptors but modify the properties of heteromeric assemblies. Disruption of the Grik4 gene locus resulted in a significant reduction in synaptic kainate receptor currents. Moreover, ablation of GluK4 and GluK5 caused complete loss of synaptic ionotropic kainate receptor function. The principal subunits were distributed away from postsynaptic densities and presynaptic active zones. There was also a profound alteration in the activation properties of the remaining kainate receptors. Despite this, kainate receptor-mediated inhibition of the slow afterhyperpolarization current (I(sAHP)), which is dependent on metabotropic pathways, was intact in GluK4/GluK5 knockout mice. These results uncover a previously unknown obligatory role for the high-affinity subunits for ionotropic kainate receptor function and further demonstrate that kainate receptor participation in metabotropic signaling pathways does not require their classic role as ion channels.


Subject(s)
Protein Subunits/physiology , Receptors, Kainic Acid/physiology , Signal Transduction/physiology , Animals , Biotinylation/methods , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , In Vitro Techniques , Mice , Mice, Knockout , Microscopy, Immunoelectron/methods , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques/methods , Presynaptic Terminals/metabolism , Protein Subunits/genetics , Receptors, Kainic Acid/classification , Receptors, Kainic Acid/deficiency , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure
9.
Neuron ; 63(3): 357-71, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19679075

ABSTRACT

Regulation of surface insertion and internalization of AMPA and NMDA receptors has emerged as a key mechanism for the control of synaptic strength. Regulatory elements for synaptic kainate receptors (KARs) are, however, largely undetermined. We have found that SNAP25 is critical for the synaptic removal of KARs, acting via GluK5 (i.e., KA2) subunits. SNAP25 coimmunoprecipitates with protein complexes containing PICK1, GRIP1, and GluK5 and colocalizes with GluK5 in both hippocampal neurons and transfected HEK293 cells. In hippocampal slices, purified SNAP25 antibodies and blocking peptides caused a GluK5-dependent run-up of KARs-mediated EPSC (EPSC(KAR)) recorded from CA3 pyramidal neurons when included in the patch pipette and prevented activity-dependent long-term depression of EPSC(KAR). As EPSC(KAR) LTD, SNAP25/PICK1/GluK5 interactions are dynamically regulated by PKC.


Subject(s)
Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, Kainic Acid/metabolism , Synapses/metabolism , Synaptosomal-Associated Protein 25/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line, Transformed , Cell Line, Tumor , Electric Stimulation , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Hippocampus/cytology , Hippocampus/ultrastructure , Humans , Immunoprecipitation/methods , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthalenes/pharmacology , Nerve Tissue Proteins/metabolism , Neural Pathways/physiology , Neuroblastoma , Neuronal Plasticity/drug effects , Neurons/cytology , Neurotoxins/pharmacology , Nuclear Proteins/metabolism , Patch-Clamp Techniques/methods , Protein Transport/genetics , Protein Transport/physiology , Pyridines/pharmacology , Rats , Receptors, Kainic Acid/deficiency , Synaptosomal-Associated Protein 25/genetics , Transfection , Vesicle-Associated Membrane Protein 2/metabolism
10.
J Neurosci ; 29(15): 5000-8, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19369569

ABSTRACT

Hippocampal mossy fiber (Mf) synapses are viewed as conditional detonators, assisting CA3 cells in complex network functions. By analyzing mice deficient for GluK2 (GluR6), GluK3 (GluR7) and GluK5 (KA2) genes we show that kainate receptors (KARs) play a crucial role in the control of synaptic integration and spike transmission efficacy at Mf synapses. We dissected out the role of the different KAR functions at Mf synapses and we show that presynaptic and postsynaptic KARs concur to amplify unitary Mf synaptic inputs to trigger spike discharge within a wide range of frequencies (from 1 to 50 Hz). Moreover, KARs strongly favor spike transmission in response to patterns of presynaptic activity mimicking in vivo dentate granule cell activity. By amplifying spike transmission, KARs also facilitate the induction of associative long-term potentiation in CA3. Hence the actions of KARs as amplifiers of spike transmission contribute largely to the "conditional detonator" function of Mf synapses and are likely important for spatial information processing.


Subject(s)
Action Potentials/physiology , Mossy Fibers, Hippocampal/physiology , Receptors, Kainic Acid/physiology , Synapses/physiology , Synaptic Transmission/physiology , Action Potentials/genetics , Animals , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/physiology , Receptors, Kainic Acid/deficiency , Receptors, Kainic Acid/genetics , Synapses/genetics , Synaptic Transmission/genetics
11.
J Pharmacol Exp Ther ; 328(2): 579-87, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18957577

ABSTRACT

Previous reports utilizing pharmacological antagonists implicate kainate receptor (KAR) activation in the development of morphine tolerance, dependence, conditioned place preference (CPP), and locomotor sensitization, but the role of glutamate receptor (GluR) 5-containing KAR in these effects remains unclear because of limited selectivity of the inhibitors employed. Therefore, we examined responses to systemic morphine treatment in mice expressing a constitutive deletion of GluR5 [GluR5 knockout (KO)]. Unlike wild-type (WT) littermates, GluR5 KO mice do not develop tolerance after repeated morphine administration by subcutaneous injection or via subcutaneous pellet implantation. In contrast, GluR5 KO mice do not differ from WT with respect to thermal or mechanical nociceptive thresholds, acute morphine antinociception, morphine disposition in the central nervous system (CNS), morphine physical dependence as revealed by naloxone-precipitated withdrawal or development of place preference and locomotor hyperresponsiveness after chronic morphine administration. It is surprising that continuous subcutaneous infusion of the GluR2/GluR5-preferring antagonist LY293558 [(3S,4aR,6R,8aR)-6-[2-(1(2)H-tetrazole-5-yl)ethyl]decahydroisoquinoline-3-carboxylic acid] decreased the number of naloxone-precipitated jumps to a similar extent in WT and GluR5 KO mice. We observed opioid-induced hypersensitivity in both groups during morphine withdrawal as demonstrated by equivalent reductions in thermal and mechanical thresholds; however, this hypersensitivity was not evident during continuous systemic morphine infusion. These data collectively indicate that KARs containing the GluR5 subunit contribute to the development of morphine tolerance without affecting nociceptive thresholds, morphine analgesia, or disposition in CNS of morphine and its metabolite morphine-3-glucuronide. In addition, constitutive deletion of GluR5 does not alter the morphine-induced increase in locomotor activity or the acquisition of morphine reward as measured by a CPP paradigm.


Subject(s)
Drug Tolerance/physiology , Gene Deletion , Morphine/adverse effects , Receptors, Kainic Acid/chemistry , Animals , Female , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Morphine/metabolism , Morphine/toxicity , Morphine Derivatives/metabolism , Physiological Phenomena , Receptors, Glutamate/deficiency , Receptors, Glutamate/genetics , Receptors, Glutamate/physiology , Receptors, Kainic Acid/deficiency , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/physiology
12.
Mol Psychiatry ; 13(9): 858-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18332879

ABSTRACT

The glutamate receptor 6 (GluR6 or GRIK2, one of the kainate receptors) gene resides in a genetic linkage region (6q21) associated with bipolar disorder (BPD), but its function in affective regulation is unknown. Compared with wild-type (WT) and GluR5 knockout (KO) mice, GluR6 KO mice were more active in multiple tests and super responsive to amphetamine. In a battery of specific tests, GluR6 KO mice also exhibited less anxious or more risk-taking type behavior and less despair-type manifestations, and they also had more aggressive displays. Chronic treatment with lithium, a classic antimanic mood stabilizer, reduced hyperactivity, aggressive displays and some risk-taking type behavior in GluR6 KO mice. Hippocampal and prefrontal cortical membrane levels of GluR5 and KA-2 receptors were decreased in GluR6 KO mice, and chronic lithium treatment did not affect these decreases. The membrane levels of other glutamatergic receptors were not significantly altered by GluR6 ablation or chronic lithium treatment. Together, these biochemical and behavioral results suggest a unique role for GluR6 in controlling abnormalities related to the behavioral symptoms of mania, such as hyperactivity or psychomotor agitation, aggressiveness, driven or increased goal-directed pursuits, risk taking and supersensitivity to psychostimulants. Whether GluR6 perturbation is involved in the mood elevation or thought disturbance of mania and the cyclicity of BPD are unknown. The molecular mechanism underlying the behavioral effects of lithium in GluR6 KO mice remains to be elucidated.


Subject(s)
Bipolar Disorder/metabolism , Receptors, Kainic Acid/metabolism , Analysis of Variance , Animals , Antimanic Agents/therapeutic use , Avoidance Learning/drug effects , Behavioral Symptoms , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Exploratory Behavior/drug effects , Interpersonal Relations , Lithium Carbonate/therapeutic use , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Kainic Acid/deficiency , Risk-Taking , Swimming , Time Factors , GluK2 Kainate Receptor
13.
J Neurophysiol ; 98(5): 2550-65, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17855589

ABSTRACT

In cerebellar Purkinje cells (PCs), activation of postsynaptic mGluR1 receptors inhibits parallel fiber (PF) to PC synaptic transmission by retrograde signaling. However, results were conflicting with respect to whether endocannabinoids or glutamate (Glu) is the retrograde messenger involved. Experiments in cerebellar slices from 10- to 12-day-old rats and mice confirmed that suppression of PF-excitatory postsynaptic currents (EPSCs) by mGluR1 agonists was entirely blocked by cannabinoid receptor antagonists at this early developmental stage. In contrast, suppression of PF-EPSCs by mGluR1 agonists was only partly blocked by cannabinoid receptor antagonists in 18- to 22-day-old rats, and the remaining suppression was accompanied by an increase in paired-pulse facilitation. This endocannnabinoidindependent suppression of PF-EPSCs was potentiated by the Glu uptake inhibitor D-threo-beta-benzyloxyaspartate (D-TBOA) and blocked by the desensitizing kainate (KA) receptors agonist SYM 2081, by nonsaturating concentrations of 6-cyano-7-nitroquinoxaline-2-3-dione (CNQX) [but not by GYKI 52466 hydrochloride (GYKI)] and by dialyzing PCs with guanosine 5'-[beta-thio]diphosphate (GDP-betaS). An endocannnabinoid-independent suppression of PF-EPSCs was also present in nearly mature wild-type mice but was absent in GluR6(-/-) mice. The endocannnabinoid-independent suppression of PF-EPSCs induced by mGluR1 agonists and the KA-dependent component of depolarization-induced suppression of excitation (DSE) were blocked by ryanodine acting at a presynaptic level. We conclude that retrograde release of Glu by PCs participates in mGluR1 agonist-induced suppression of PF-EPSCs at nearly mature PF-PC synapses and that Glu operates through activation of presynaptic KA receptors located on PFs and prolonged release of calcium from presynaptic internal calcium stores.


Subject(s)
Calcium/metabolism , Excitatory Amino Acid Agents/pharmacology , Feedback/drug effects , Purkinje Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , Calcium/pharmacology , Cannabinoid Receptor Modulators/agonists , Cannabinoid Receptor Modulators/antagonists & inhibitors , Cannabinoid Receptor Modulators/metabolism , Cerebellum/cytology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Feedback/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Purkinje Cells/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Kainic Acid/agonists , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/deficiency , Synapses/drug effects , Synaptic Transmission/drug effects , GluK2 Kainate Receptor
14.
Proc Natl Acad Sci U S A ; 104(29): 12181-6, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17620617

ABSTRACT

Presynaptic ionotropic glutamate receptors are emerging as key players in the regulation of synaptic transmission. Here we identify GluR7, a kainate receptor (KAR) subunit with no known function in the brain, as an essential subunit of presynaptic autoreceptors that facilitate hippocampal mossy fiber synaptic transmission. GluR7(-/-) mice display markedly reduced short- and long-term synaptic potentiation. Our data suggest that presynaptic KARs are GluR6/GluR7 heteromers that coassemble and are localized within synapses. We show that recombinant GluR6/GluR7 KARs exhibit low sensitivity to glutamate, and we provide evidence that presynaptic KARs at mossy fiber synapses are likely activated by high concentrations of glutamate. Overall, from our data, we propose a model whereby presynaptic KARs are localized in the presynaptic active zone close to release sites, display low affinity for glutamate, are likely Ca(2+)-permeable, are activated by single release events, and operate within a short time window to facilitate the subsequent release of glutamate.


Subject(s)
Autoreceptors/metabolism , Mossy Fibers, Hippocampal/metabolism , Presynaptic Terminals/metabolism , Receptors, Kainic Acid/metabolism , Animals , Cell Line , Excitatory Postsynaptic Potentials , Humans , Long-Term Potentiation , Mice , Protein Subunits/metabolism , Protein Transport , Receptors, Kainic Acid/deficiency , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism , Time Factors , GluK2 Kainate Receptor , GluK3 Kainate Receptor
15.
Dev Neurobiol ; 67(2): 146-57, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17443779

ABSTRACT

In the anterior cingulate cortex (ACC), GluR5-containing kainate receptor mediated the small portion of excitatory postsynaptic current. However, little is known about its role in modulation of neurotransmitter release in this brain region. In the present study, we address this question by using selective GluR5 agonist and antagonist, as well as GluR5(-/-) mice. Our results showed that activation of GluR5 induced action potential-dependent GABA release, which is also required for the activation of voltage-dependent calcium channel and Ca(2+) influx. The effect of GluR5 activation is selective to the GABAergic, but not glutamatergic synaptic transmission. Endogenous activation of GluR5 also enhanced GABA release to ACC pyramidal neurons and the corresponding postsynaptic tonic GABA current. Our results suggest the somatodendritic, but not presynaptic GluR5, in modulation of GABA release. The endogenous GluR5 activation and the subsequent tonic GABA current may play an inhibitory role in ACC-related brain functions.


Subject(s)
Gyrus Cinguli/cytology , Neural Inhibition/drug effects , Neural Inhibition/genetics , Receptors, Kainic Acid/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Receptors, Kainic Acid/deficiency , gamma-Aminobutyric Acid/metabolism
16.
Mol Cell Neurosci ; 34(2): 219-30, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17174564

ABSTRACT

Correct glutamate receptor localization in neurons is crucial for neurotransmission in the brain. Here we investigated the mechanisms underlying localization of kainate GluR5 receptors to dendrites in cultured hippocampal neurons. We find that the GluR5 distribution depends on association with GluR6 and KA2 subunits. The GluR5 subunit was expressed in distal dendrites only when GluR6 and KA2 subunits were present, whereas it was restricted to proximal dendrites in the absence of these subunits. The overlap between GluR5 distribution and the organization of microtubules in dendrites led us to examine whether KIF17, a microtubule motor protein expressed in distal dendrites, is involved in GluR5 localization to distal dendrites. We show here, for the first time that the microtubule motor protein KIF17 interacts with GluR6 and KA2 subunits and is required for GluR5 localization to distal dendrites, defining a novel mechanism that controls receptor localization in neurons.


Subject(s)
Dendrites/metabolism , Kinesins/metabolism , Neurons/cytology , Protein Subunits/metabolism , Receptors, Kainic Acid/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Immunohistochemistry/methods , Immunoprecipitation/methods , Mice , Mice, Knockout , Mutagenesis/physiology , Rats , Rats, Long-Evans , Receptors, Kainic Acid/deficiency , Transfection/methods
17.
J Neurosci ; 26(26): 7014-21, 2006 Jun 28.
Article in English | MEDLINE | ID: mdl-16807331

ABSTRACT

Neuronal kainate receptors are typically heteromeric complexes composed of GluR5-7 and KA1-2 subunits. Although GluR5-7 can exist as functional homomeric channels, the KA subunits cannot. KA2 is widely expressed in the CNS, and KA2/GluR6 heteromers are the most prevalent subunit composition in brain. Previous work has identified endoplasmic reticulum (ER)-retention motifs in the C terminus of KA2, which prevent surface expression of KA2 homomers. However, we find that, when these motifs are mutated, only a small fraction of KA2 is surface expressed. We now identify an additional ER retention motif in the intracellular loop region of KA2, which, when mutated together with the C-terminal motifs, significantly increases the level of KA2 surface expression. However, electrophysiological analysis of surface-expressed KA2 homomers indicates that they do not form functional ion channels. In heterologous cells, a large fraction of KA2 remains intracellular even when the trafficking motifs are mutated or when GluR6 is coexpressed. Therefore, we analyzed the trafficking of endogenous KA2 in vivo. We find that native KA2 surface expression is dramatically reduced in GluR6 knock-out mice compared with wild-type mice. In contrast, KA2 trafficking was unaffected in the GluR5 knock-out. Thus, our study demonstrates that trafficking motifs in both the intracellular loop and C terminus regulate KA2 surface expression; however, in neurons, GluR6 oligomerization is required for egress of KA2 from the ER and transport to the cell surface. The combination of these mechanisms likely prevents surface expression of nonfunctional KA2 homomers and ensures a high level of GluR6/KA2 heteromeric kainate receptors.


Subject(s)
Endoplasmic Reticulum/physiology , Intracellular Membranes/metabolism , Receptors, Kainic Acid/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Protein Transport , Receptors, Kainic Acid/deficiency , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , GluK2 Kainate Receptor
18.
J Biol Chem ; 281(22): 15475-84, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16595684

ABSTRACT

Assembly and trafficking of neurotransmitter receptors are processes contingent upon interactions between intracellular chaperone systems and discrete determinants in the receptor proteins. Kainate receptor subunits, which form ionotropic glutamate receptors with diverse roles in the central nervous system, contain a variety of trafficking determinants that promote either membrane expression or intracellular sequestration. In this report, we identify the coatomer protein complex I (COPI) vesicle coat as a critical mechanism for retention of the kainate receptor subunit KA2 in the endoplasmic reticulum. COPI subunits immunoprecipitated with KA2 subunits from both cerebellum and COS-7 cells, and beta-COP protein interacted directly with immobilized KA2 peptides containing the arginine-rich retention/retrieval determinant. Association between COPI proteins and KA2 subunits was significantly reduced upon alanine substitution of this signal in the cytoplasmic tail of KA2. Temperature-sensitive degradation of COPI complex proteins was correlated with an increase in plasma membrane localization of the homologous KA2 receptor. Assembly of heteromeric GluR6a/KA2 receptors markedly reduced association of KA2 and COPI. Finally, the reduction in COPI binding was correlated with an increased association with 14-3-3 proteins, which mediate forward trafficking of other integral signaling proteins. These interactions therefore represent a critical early checkpoint for biosynthesis of functional KARs.


Subject(s)
14-3-3 Proteins/metabolism , Coat Protein Complex I/metabolism , Receptors, Kainic Acid/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Coat Protein Complex I/chemistry , In Vitro Techniques , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Subunits , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/deficiency , Receptors, Kainic Acid/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
19.
Brain Res Mol Brain Res ; 142(1): 9-18, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16219388

ABSTRACT

Functional kainate receptors are expressed in the spinal cord substantia gelatinosa region, and their activation contributes to bi-directional regulation of excitatory synaptic transmission at primary afferent synapses with spinal cord substantia gelatinosa neurons. However, no study has reported a role(s) for kainate receptor subtypes in long-term synaptic plasticity phenomena in this region. Using gene-targeted mice lacking glutamate receptor 5 (GLU(K5)) or GLU(K6) subunit, we here show that GLU(K6) subunit, but not GLU(K5) subunit, is involved in the induction of long-term potentiation of excitatory postsynaptic potentials, evoked by two different protocols: (1) high-frequency primary afferent stimulation (100 Hz, 3 s) and (2) low-frequency spike-timing stimulation (1 Hz, 200 pulses). In addition, GLU(K6) subunit plays an important role in the expression of kainate-induced Ca2+ transients in the substantia gelatinosa. On the other hand, genetic deletion of GLU(K5) or GLU(K6) subunit does not prevent the induction of long-term depression. These results indicate that unique expression of kainate receptors subunits is important in regulating spinal synaptic plasticity and thereby processing of sensory information, including pain.


Subject(s)
Calcium/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, Kainic Acid/deficiency , Substantia Gelatinosa/cytology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Animals , Benzoates/pharmacology , Cadmium Chloride/pharmacology , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Glutamates/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , In Vitro Techniques , Kainic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/radiation effects , Neurons/drug effects , Patch-Clamp Techniques/methods , Protein Subunits/deficiency , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
20.
J Neurophysiol ; 94(3): 1805-13, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15928066

ABSTRACT

Kainate (KA) receptors are expressed widely in the CNS. However, little is known about their functional characterization, molecular identity, and role in synaptic transmission in the forebrain of adult mice. Patch-clamp recordings in genetically modified mice show that postsynaptic KA receptors contribute to fast synaptic transmission in pyramidal neurons in the anterior cingulate cortex (ACC), a forebrain region critical for higher-order cognitive brain functions such as memory and mental disorders. Single-shock stimulation could induce small KA receptor-mediated excitatory postsynaptic currents (KA EPSCs) in the presence of picrotoxin, D-2-amino-5-phosphono-pentanoic acid, and a selective AMPA receptor antagonist, GYKI 53655. KA EPSCs had a significantly slower rise time course and decay time constant compared with AMPA receptor-mediated EPSCs. High-frequency repetitive stimulation significantly facilitated the KA EPSCs. Genetic deletion of the GluR6 or GluR5 subunit significantly reduced, and GluR5 and 6 double knockout completely abolished, KA EPSCs and KA-activated currents in ACC pyramidal neurons. Our results show that KA receptors contribute to synaptic transmission in adult ACC pyramidal neurons and provide a synaptic basis for the physiology and pathology of KA receptors in ACC-related functions.


Subject(s)
Gyrus Cinguli/cytology , Gyrus Cinguli/physiology , Neurons/physiology , Receptors, Kainic Acid/physiology , Synaptic Transmission/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Benzodiazepines/pharmacology , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Gyrus Cinguli/drug effects , Gyrus Cinguli/radiation effects , In Vitro Techniques , Isoxazoles/pharmacology , Kainic Acid/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/classification , Neurons/drug effects , Neurons/radiation effects , Patch-Clamp Techniques/methods , Propionates/pharmacology , Receptors, Kainic Acid/deficiency , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects , GluK2 Kainate Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...