Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Cell Rep ; 43(7): 114427, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38986610

ABSTRACT

Kainate (KA)-type glutamate receptors (KARs) are implicated in various neuropsychiatric and neurological disorders through their ionotropic and metabotropic actions. However, compared to AMPA- and NMDA-type receptor functions, many aspects of KAR biology remain incompletely understood. Our study demonstrates an important role of KARs in organizing climbing fiber (CF)-Purkinje cell (PC) synapses and synaptic plasticity in the cerebellum, independently of their ion channel or metabotropic functions. The amino-terminal domain (ATD) of the GluK4 KAR subunit binds to C1ql1, provided by CFs, and associates with Bai3, an adhesion-type G protein-coupled receptor expressed in PC dendrites. Mice lacking GluK4 exhibit no KAR-mediated responses, reduced C1ql1 and Bai3 levels, and fewer CF-PC synapses, along with impaired long-term depression and oculomotor learning. Remarkably, introduction of the ATD of GluK4 significantly improves all these phenotypes. These findings demonstrate that KARs act as synaptic scaffolds, orchestrating synapses by forming a KAR-C1ql1-Bai3 complex in the cerebellum.


Subject(s)
Cerebellum , Neuronal Plasticity , Purkinje Cells , Receptors, Kainic Acid , Synapses , Animals , Synapses/metabolism , Receptors, Kainic Acid/metabolism , Neuronal Plasticity/physiology , Cerebellum/metabolism , Mice , Purkinje Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Humans
2.
Nat Commun ; 10(1): 4991, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31676751

ABSTRACT

Overinhibition is assumed one of the main causes of cognitive deficits (e.g. memory impairment) in mouse models of Down syndrome (DS). Yet the mechanisms that drive such exaggerated synaptic inhibition and their behavioral effects remain unclear. Here we report the existence of bidirectional alterations to the synaptic inhibition on CA1 pyramidal cells in the Ts2Cje mouse model of DS which are associated to impaired spatial memory. Furthermore, we identify triplication of the kainate receptor (KAR) encoding gene Grik1 as the cause of these phenotypes. Normalization of Grik1 dosage in Ts2Cje mice specifically restored spatial memory and reversed the bidirectional alterations to CA1 inhibition, but not the changes in synaptic plasticity or the other behavioral modifications observed. We propose that modified information gating caused by disturbed inhibitory tone rather than generalized overinhibition underlies some of the characteristic cognitive deficits in DS.


Subject(s)
CA1 Region, Hippocampal/physiology , Dendrites/physiology , Down Syndrome/physiopathology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Spatial Memory/physiology , Animals , CA1 Region, Hippocampal/cytology , Disease Models, Animal , Down Syndrome/pathology , Female , Humans , Male , Mice
3.
ACS Chem Neurosci ; 10(11): 4685-4695, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31622082

ABSTRACT

Selective pharmacological tool compounds are invaluable for understanding the functions of the various ionotropic glutamate receptor subtypes. For the kainate receptors, these compounds are few. Here we have synthesized nine novel quinoxaline-2,3-diones with substitutions in the 7-position to investigate the structure-activity relationship at kainate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Compound 11 exhibited the highest binding affinity across GluK1-3 while having selectivity toward kainate vs AMPA receptors. Compound 11 potently inhibited glutamate evoked currents at homomeric GluK1 and GluK3 receptors in HEK293 cells with Kb values of 65 and 39 nM, respectively. The binding mode of 11 in the ligand binding domain of GluK1 was investigated by X-ray crystallography, revealing that 11 stabilizes the receptor in an open conformation, consistent with its demonstrated antagonism. Furthermore, 11 was tested for analgesic effects in the mouse tail flick test where it significantly increased tail flick latency at doses where 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]-quinoxaline-7-sulfonamide (NBQX) was ineffective.


Subject(s)
Analgesics/metabolism , Crystallography, X-Ray/methods , Excitatory Amino Acid Antagonists/metabolism , Quinoxalines/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Analgesics/chemistry , Analgesics/pharmacology , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Humans , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Protein Structure, Secondary , Protein Structure, Tertiary , Quinoxalines/chemistry , Quinoxalines/pharmacology , Rats , Receptors, Kainic Acid/chemistry , Structure-Activity Relationship
4.
Cell Rep ; 23(13): 3827-3838, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29949767

ABSTRACT

Altered glutamatergic neurotransmission is thought to contribute to mental disorders and neurodegenerative diseases. Copy-number variation in genes associated with glutamatergic synapses represents a source of genetic variability, possibly underlying neurological and mental disease susceptibility. The GRIK4 gene encodes a high-affinity kainate receptor subunit of essentially unknown function, although de novo duplication of the 11q23.3-q24.1 locus to which it maps has been detected in autism and other disorders. To determine how changes in the dose of Grik4 affect synaptic activity, we studied mice overexpressing this gene in the forebrain. A mild gain in Grik4 enhances synaptic transmission, causing a persistent imbalance in inhibitory and excitatory activity and disturbing the circuits responsible for the main amygdala outputs. These changes in glutamatergic activity reverse when Grik4 levels are normalized; thus, they may account for the behavioral abnormalities in disorders like autism or schizophrenia.


Subject(s)
Basolateral Nuclear Complex/metabolism , Receptors, Kainic Acid/genetics , Animals , Basolateral Nuclear Complex/pathology , Behavior, Animal , Excitatory Postsynaptic Potentials/drug effects , Female , Gene Dosage , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Glutamate/metabolism , Receptors, Kainic Acid/metabolism , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology
5.
J Neurosci ; 35(40): 13619-28, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26446216

ABSTRACT

The understanding of brain diseases requires the identification of the molecular, synaptic, and cellular disruptions underpinning the behavioral features that define the disease. The importance of genes related to synaptic function in brain disease has been implied in studies describing de novo germline mutations and copy number variants. Indeed, de novo copy number variations (deletion or duplication of a chromosomal region) of synaptic genes have been recently implicated as risk factors for mental retardation or autism. Among these genes is GRIK4, a gene coding for a glutamate receptor subunit of the kainate type. Here we show that mice overexpressing grik4 in the forebrain displayed social impairment, enhanced anxiety, and depressive states, accompanied by altered synaptic transmission, showing more efficient information transfer through the hippocampal trisynaptic circuit. Together, these data indicate that a single gene variation in the glutamatergic system results in behavioral symptomatology consistent with autism spectrum disorders as well as in alterations in synaptic function in regions involved in social activity. Autistic features of these mice represent powerful tools for improving diagnosis and testing of specific treatments targeting abnormalities in glutamatergic signaling related to autism spectrum disorders. SIGNIFICANCE STATEMENT: A genetic overlap exists between autism spectrum disorders (ASD), currently thought to represent a continuum of the same disorder with varying degrees of severity, and other neurodevelopmental and neuropsychiatric endophenotypes. We show that the duplication of a single gene coding for a high-affinity kainate receptor subunit (i.e., grik4) in a limited area of the brain recapitulates behavioral endophenotypes seen in humans diagnosed with autism (anhedonia, depression, anxiety, and altered social interaction), including some humans with GRIK4 duplications. Therefore, it should be possible to use mice overexpressing grik4 to directly address circuit dysfunctions associated with ASDs and test specific treatments of autism-related behaviors.


Subject(s)
Autism Spectrum Disorder/genetics , Hippocampus/cytology , Mutation/genetics , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , Synaptic Transmission/genetics , Animals , Animals, Newborn , Autism Spectrum Disorder/physiopathology , Cell Line, Transformed , Dark Adaptation/genetics , Disease Models, Animal , Disks Large Homolog 4 Protein , Exploratory Behavior/physiology , Food Preferences , Guanylate Kinases/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Interpersonal Relations , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Sucrose/administration & dosage , Swimming/physiology
6.
Nat Neurosci ; 15(8): 1134-43, 2012 Jul 08.
Article in English | MEDLINE | ID: mdl-22772332

ABSTRACT

Developing axons must control their growth rate to follow the appropriate pathways and establish specific connections. However, the regulatory mechanisms involved remain elusive. By combining live imaging with transplantation studies in mice, we found that spontaneous calcium activity in the thalamocortical system and the growth rate of thalamocortical axons were developmentally and intrinsically regulated. Indeed, the spontaneous activity of thalamic neurons governed axon growth and extension through the cortex in vivo. This activity-dependent modulation of growth was mediated by transcriptional regulation of Robo1 through an NF-κB binding site. Disruption of either the Robo1 or Slit1 genes accelerated the progression of thalamocortical axons in vivo, and interfering with Robo1 signaling restored normal axon growth in electrically silent neurons. Thus, modifications to spontaneous calcium activity encode a switch in the axon outgrowth program that allows the establishment of specific neuronal connections through the transcriptional regulation of Slit1 and Robo1 signaling.


Subject(s)
Axons/physiology , Calcium Signaling/genetics , Cerebral Cortex/physiology , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Thalamus/physiology , Animals , Axons/pathology , Calcium/metabolism , Cerebral Cortex/growth & development , Female , Gene Expression Regulation, Developmental/genetics , Mice , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Thalamus/growth & development , Roundabout Proteins
7.
Nature ; 464(7293): 1376-80, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20393464

ABSTRACT

Schizophrenia is a complex disorder that interferes with the function of several brain systems required for cognition and normal social behaviour. Although the most notable clinical aspects of the disease only become apparent during late adolescence or early adulthood, many lines of evidence suggest that schizophrenia is a neurodevelopmental disorder with a strong genetic component. Several independent studies have identified neuregulin 1 (NRG1) and its receptor ERBB4 as important risk genes for schizophrenia, although their precise role in the disease process remains unknown. Here we show that Nrg1 and ErbB4 signalling controls the development of inhibitory circuitries in the mammalian cerebral cortex by cell-autonomously regulating the connectivity of specific GABA (gamma-aminobutyric acid)-containing interneurons. In contrast to the prevalent view, which supports a role for these genes in the formation and function of excitatory synapses between pyramidal cells, we found that ErbB4 expression in the mouse neocortex and hippocampus is largely confined to certain classes of interneurons. In particular, ErbB4 is expressed by many parvalbumin-expressing chandelier and basket cells, where it localizes to axon terminals and postsynaptic densities receiving glutamatergic input. Gain- and loss-of-function experiments, both in vitro and in vivo, demonstrate that ErbB4 cell-autonomously promotes the formation of axo-axonic inhibitory synapses over pyramidal cells, and that this function is probably mediated by Nrg1. In addition, ErbB4 expression in GABA-containing interneurons regulates the formation of excitatory synapses onto the dendrites of these cells. By contrast, ErbB4 is dispensable for excitatory transmission between pyramidal neurons. Altogether, our results indicate that Nrg1 and ErbB4 signalling is required for the wiring of GABA-mediated circuits in the postnatal cortex, providing a new perspective to the involvement of these genes in the aetiology of schizophrenia.


Subject(s)
Cerebral Cortex/metabolism , ErbB Receptors/metabolism , Interneurons/metabolism , Neural Pathways/physiology , Neuregulin-1/metabolism , Signal Transduction , gamma-Aminobutyric Acid/metabolism , Animals , Cell Differentiation , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Dendrites/metabolism , Embryo, Mammalian , ErbB Receptors/deficiency , ErbB Receptors/genetics , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Female , In Vitro Techniques , Mice , Neural Inhibition/genetics , Neural Inhibition/physiology , Pyramidal Cells/metabolism , Receptor, ErbB-4 , Schizophrenia/genetics , Schizophrenia/metabolism , Synapses/metabolism
8.
J Neurosci ; 30(1): 242-54, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053906

ABSTRACT

Protein kinase C (PKC) enhances NMDA receptor (NMDAR)-mediated currents and promotes NMDAR delivery to the cell surface via SNARE-dependent exocytosis. Although the mechanisms of PKC potentiation are established, the molecular target of PKC is unclear. Here we show that synaptosomal-associated protein of 25 kDa (SNAP-25), a SNARE protein, is functionally relevant to PKC-dependent NMDAR insertion, and identify serine residue-187 as the molecular target of PKC phosphorylation. Constitutively active PKC delivered via the patch pipette potentiated NMDA (but not AMPA) whole-cell currents in hippocampal neurons. Expression of RNAi targeting SNAP-25 or mutant SNAP-25(S187A) and/or acute disruption of the SNARE complex by treatment with BoNT A, BoNT B or SNAP-25 C-terminal blocking peptide abolished NMDAR potentiation. A SNAP-25 peptide and function-blocking antibody suppressed PKC potentiation of NMDA EPSCs at mossy fiber-CA3 synapses. These findings identify SNAP-25 as the target of PKC phosphorylation critical to PKC-dependent incorporation of synaptic NMDARs and document a postsynaptic action of this major SNARE protein relevant to synaptic plasticity.


Subject(s)
Gene Targeting , Protein Kinase C/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptosomal-Associated Protein 25/metabolism , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Female , Humans , Mice , Mice, Mutant Strains , Molecular Sequence Data , Phosphorylation , Protein Binding/physiology , Protein Kinase C/genetics , Protein Transport/physiology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Synaptosomal-Associated Protein 25/genetics , Xenopus laevis
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.
Mol Cell Neurosci ; 37(2): 323-34, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18065236

ABSTRACT

Glutamate receptors of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type play an important role in synaptic plasticity and contribute to cell death under excitotoxic conditions. AMPA receptors form heterotetramers of four homologous subunits (GluR1-4), which exist in two functionally different isoforms, flip and flop, generated by alternative splicing. We identified transcripts for alternatively spliced isoforms of AMPA receptor subunits which lack both the flip and the flop exons, in hippocampal and retinal cultures. These transcripts originate AMPA receptor subunits lacking the flip/flop cassette, the fourth transmembrane domain and the intracellular C-terminus. Truncated GluR1 associates with full-length GluR1 and exerts a dominant negative effect, giving rise to non-functional receptors. Moreover, truncated GluR1 reaches the cell surface, but is not efficiently targeted to the synapse. Hippocampal neuronal transfection with truncated GluR1 resulted in a significant reduction in apoptotic neuronal death triggered by toxic concentrations of glutamate. Furthermore, mRNA coding for the truncated subunits is consistently detected in some regions of the brain in epileptic rats and in hippocampal neurons submitted to toxic concentrations of glutamate. The existence of truncated AMPA receptor subunits may constitute an intrinsic neuroprotective mechanism.


Subject(s)
Alternative Splicing/genetics , Cytoprotection/genetics , Protein Isoforms/genetics , RNA, Messenger/genetics , Receptors, AMPA/genetics , Synaptic Transmission/genetics , Animals , Cell Death/genetics , Cell Line , Cells, Cultured , Chick Embryo , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Mutation/genetics , Protein Isoforms/chemistry , Protein Structure, Tertiary/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Rats , Rats, Wistar , Receptors, AMPA/chemistry
11.
Neuron ; 48(2): 279-88, 2005 Oct 20.
Article in English | MEDLINE | ID: mdl-16242408

ABSTRACT

AMPA-type glutamate receptors are specifically inhibited by the noncompetitive antagonists GYKI-53655 and CP-465,022, which act through sites and mechanisms that are not understood. Using receptor mutagenesis, we found that these antagonists bind at the interface between the S1 and S2 glutamate binding core and channel transmembrane domains, specifically interacting with S1-M1 and S2-M4 linkers, thereby disrupting the transduction of agonist binding into channel opening. We also found that the antagonists' affinity is higher for agonist-unbound receptors than for activated nondesensitized receptors, further depending on the level of S1 and S2 domain closure. These results provide evidence for substantial conformational changes in the S1-M1 and S2-M4 linkers following agonist binding and channel opening, offering a conceptual frame to account for noncompetitive antagonism of AMPA receptors.


Subject(s)
Benzodiazepines/pharmacology , Membrane Potentials/physiology , Quinazolines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/physiology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Blotting, Western/methods , Brain/drug effects , Cell Line , Cloning, Molecular/methods , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Membrane Potentials/drug effects , Models, Neurological , Mutagenesis/physiology , Patch-Clamp Techniques/methods , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Quinazolines/pharmacokinetics , Rats , Receptors, AMPA/chemistry , Transfection/methods , Tritium/pharmacokinetics , Xenopus laevis
12.
J Neurosci ; 24(41): 8986-93, 2004 Oct 13.
Article in English | MEDLINE | ID: mdl-15483117

ABSTRACT

Although some physiological functions of kainate receptors (KARs) still remain unclear, recent advances have highlighted a role in synaptic physiology. In hippocampal slices, kainate depresses GABA-mediated synaptic inhibition and increases the firing rate of interneurons. However, the sensitivity to agonists of these responses differs, suggesting that the presynaptic and somatic KARs have a distinct molecular composition. Hippocampal interneurons express several distinct KAR subunits that can assemble into heteromeric receptors with a variety of pharmacological properties and that, in principle, could fulfill different roles. To address which receptor types mediate each of the effects of kainate in interneurons, we used new compounds and mice deficient for specific KAR subunits. In a recombinant assay, 5-carboxyl-2,4-di-benzamido-benzoic acid (NS3763) acted exclusively on homomeric glutamate receptor subunit 5 (GluR5), whereas 3S,4aR,6S,8aR-6-((4-carboxyphenyl)methyl) 1,2,3,4,4a,5,6,7,8,8a-decahydroisoquinoline-3-carboxylic acid (LY382884) antagonized homomeric GluR5 and any heteromeric combination containing GluR5 subunits. In hippocampal slices, LY382884, but not NS3763, was able to prevent kainate-induced depression of evoked IPSC. In contrast, neither prevented the concomitant increase in spontaneous IPSC frequency. The selectivity of these compounds was seen additionally in knock-out mice, such that they were inactive in GluR5-/- mice but completely effective in GluR6-/- mice. Our data indicate that in wild-type mice, CA1 interneurons express heteromeric GluR6 -KA2 receptors in their somatic compartments and GluR5-GluR6 or GluR5-KA2 at presynaptic terminals. However, functional compensation appears to take place in the null mutants, a new pharmacological profile emerging more compatible with the activity of homomeric receptors in both compartments: GluR5 in GluR6-/- mice and GluR6 in GluR5-/- mice.


Subject(s)
Hippocampus/metabolism , Interneurons/metabolism , Receptors, Kainic Acid/metabolism , Animals , Benzamides/pharmacology , Benzoates/pharmacology , Cell Line , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , Humans , Interneurons/drug effects , Isoquinolines/pharmacology , Kidney/cytology , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Protein Subunits/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/genetics , Transfection , GluK2 Kainate Receptor
13.
J Neurosci ; 23(25): 8641-8, 2003 Sep 24.
Article in English | MEDLINE | ID: mdl-14507963

ABSTRACT

Ionotropic glutamate receptors of the kainate and AMPA subtypes share a number of structural features, both topographical and in terms of stoichiometry. In addition, AMPA and kainate receptors share similar pharmacological and biophysical properties in that they are activated by common agonists and display rapid activation and desensitization characteristics. However, we show here that in contrast to AMPA receptor-mediated responses (native or recombinant GluR3 receptor), the response of native and recombinant (GluR6) kainate receptors to glutamate was drastically reduced in the absence of extracellular Na+ (i.e., when replaced by Cs+). Removal of Na+ increases the rate of desensitization, indicating that external Na+ modulates channel gating. Whereas the size of the substituting cation is important in mimicking the action of Na+ (Li+>K+>Cs+), modulation was voltage independent. These results indicate the existence of different gating mechanisms for AMPA and kainate receptors. By using chimeric AMPA-kainate receptors derived from GluR3 and GluR6, we have identified a key residue in the S2 segment of GluR6 (M770) that is largely responsible for the sensitivity of the receptor to external Na+. Thus, these results show the existence of a specific kainate receptor gating mechanism that requires external Na+ to be operative.


Subject(s)
Extracellular Space/metabolism , Ion Channel Gating/physiology , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , Sodium/physiology , Allosteric Regulation/physiology , Animals , Cells, Cultured , Humans , Ion Channel Gating/drug effects , Kainic Acid/pharmacology , Kidney/cytology , Kidney/metabolism , Models, Molecular , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary/physiology , Receptor, Metabotropic Glutamate 5 , Receptors, Kainic Acid/drug effects , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium/metabolism , Sodium/pharmacology , Structure-Activity Relationship , Transfection , Xenopus
14.
Neuron ; 39(3): 543-53, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12895426

ABSTRACT

The potent neurotoxin kainate activates ion channel-forming receptors. However, it can also activate a G protein-coupled signaling pathway to inhibit transmitter release in central neurons. It remains unclear whether the same receptor complex is involved in both signaling activities. Here we show that in a population of dorsal root ganglion cells, exposure to kainate elicits a G protein-dependent increase in intracellular Ca2+. Furthermore, in these cells a brief exposure to kainate inhibited the K+-induced Ca2+ increase, a process that was sensitive to the G protein inhibitor Pertussis toxin and inhibitors of protein kinase C. This metabotropic action did not require ion channel activity and was not observed in neurons prepared from mice deficient for the ion channel-forming subunit GluR5. These results indicate that GluR5, an ion channel-forming subunit, signals through a second messenger cascade, inhibiting voltage-dependent Ca2+ channels. Thus, such a system represents a noncanonical signaling route of ion channel-forming receptors.


Subject(s)
Receptors, Kainic Acid/physiology , Signal Transduction/physiology , Animals , Calcium Channels/physiology , Calcium Signaling/physiology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Wistar , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL