Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 320
Filter
1.
Neuropharmacology ; 197: 108696, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34274351

ABSTRACT

Presynaptic kainate (KA) receptors (KARs) modulate GABA and glutamate release in the central nervous system of mammals. While some of the actions of KARs are ionotropic, metabotropic actions for these receptors have also been seen to modulate both GABA and glutamate release. In general, presynaptic KARs modulate glutamate release through their metabotropic actions in a biphasic manner, with low KA concentrations producing an increase in glutamate release and higher concentrations of KA driving weaker release of this neurotransmitter. Different molecular mechanisms are involved in this modulation of glutamate release, with a G-protein independent, Ca2+-calmodulin adenylate cyclase (AC) and protein kinase A (PKA) dependent mechanism facilitating glutamate release, and a G-protein, AC and PKA dependent mechanism mediating the decrease in neurotransmitter release. Here, we describe the events underlying the KAR modulation of glutamatergic transmission in different brain regions, addressing the possible functions of this modulation and proposing future research lines in this field. This article is part of the special Issue on 'Glutamate Receptors - Kainate receptors'.


Subject(s)
Glutamic Acid/metabolism , Receptors, Kainic Acid/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Humans , Receptors, Kainic Acid/drug effects , Receptors, Metabotropic Glutamate/drug effects , gamma-Aminobutyric Acid/metabolism
2.
Neuropharmacology ; 197: 108699, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34246686

ABSTRACT

Kainate receptors (KARs) constitute a family of ionotropic glutamate receptors (iGluRs) with distinct physiological roles in synapses and neuronal circuits. Despite structural and biophysical commonalities with the other iGluRs, AMPA receptors and NMDA receptors, their role as post-synaptic receptors involved in shaping EPSCs to transmit signals across synapses is limited to a small number of synapses. On the other hand KARs regulate presynaptic release mechanisms and control ion channels and signaling pathways through non-canonical metabotropic actions. We review how these different KAR-dependent mechanisms concur to regulate the activity and plasticity of neuronal circuits in physiological conditions of activation of KARs by endogenous glutamate (as opposed to pharmacological activation by exogenous agonists). KARs have been implicated in neurological disorders, based on genetic association and on physiopathological studies. A well described example relates to temporal lobe epilepsy for which the aberrant recruitment of KARs at recurrent mossy fiber synapses takes part in epileptogenic neuronal activity. In conclusion, KARs certainly represent an underestimated actor in the regulation of neuronal circuits, and a potential therapeutic target awaiting more selective and efficient genetic tools and/or ligands. This article is part of the special Issue on 'Glutamate Receptors - Kainate receptors'.


Subject(s)
Nerve Net/physiology , Receptors, Kainic Acid/physiology , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Humans , Nerve Net/drug effects , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/genetics , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
3.
Neurobiol Learn Mem ; 159: 16-23, 2019 03.
Article in English | MEDLINE | ID: mdl-30731234

ABSTRACT

Working memory (WM), the capacity for short-term storage and manipulation of small quantities of information, depends on fronto-parietal circuits. However, the function of the posterior parietal cortex (PPC) in WM has gone relatively understudied in rodents. Recent evidence calls into question whether the PPC is necessary for all forms of WM. Thus, the present experiment examined the role of the rat PPC in the Trial-Unique Non-matching-to-Location (TUNL) task, a touchscreen-based visuospatial WM task that relies on the rat medial prefrontal cortex (mPFC). Temporary inactivation of the PPC caused by bilateral infusions of muscimol and baclofen significantly impaired accuracy and increased the number of correction trials performed, indicating that the PPC is necessary for performance of TUNL. Additionally, we investigated the effects of blocking NMDA or non-NMDA parietal ionotropic glutamate receptors on TUNL and found that, in contrast to the prefrontal cortex, NMDA receptors in the PPC are not necessary for TUNL performance, whereas blockade of AMPA/Kainate receptors significantly impaired accuracy. These results indicate that performance of the TUNL task depends on the PPC but that NMDA receptor signaling within this brain area is not necessary for intact performance.


Subject(s)
Behavior, Animal/physiology , Memory, Short-Term/physiology , Parietal Lobe/metabolism , Psychomotor Performance/physiology , Receptors, AMPA/physiology , Receptors, Kainic Acid/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Space Perception/physiology , Visual Perception/physiology , Animals , Baclofen/pharmacology , Behavior, Animal/drug effects , GABA Agonists/pharmacology , Male , Memory, Short-Term/drug effects , Muscimol/pharmacology , Parietal Lobe/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Long-Evans , Receptors, AMPA/drug effects , Receptors, Kainic Acid/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Space Perception/drug effects , Visual Perception/drug effects
4.
Br J Anaesth ; 119(5): 1047-1054, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28981700

ABSTRACT

BACKGROUND: In addition to general anaesthetic effects, sevoflurane can also induce hyperactive behaviours during induction and recovery, which may contribute to neurotoxicity; however, the mechanism of such effects is unclear. Volatile anaesthetics including isoflurane have been found to activate the kainate (GluK2) receptor. We developed a novel mouse model and further explored the involvement of kainate (GluK2) receptors in sevoflurane-induced hyperactivity. METHODS: Maximal speed, mean speed, total movement distance and resting percentage of C57BL/6 mice were quantitatively measured using behavioural tracking software before and after sevoflurane anaesthesia. Age dependence of this model was also analysed and sevoflurane-induced hyperactivity was evaluated after intracerebral injection of the GluK2 receptor blocker NS-102. Neurones from the hippocampal CA3 region were used to undertake in vitro electrophysiological measurement of kainate currents and miniature excitatory postsynaptic potential (mEPSP). RESULTS: Sevoflurane induced significant hyperactivities in mice under sevoflurane 1% anaesthesia and during the recovery period, characterized as increased movement speed and total distance. The hyperactivity was significantly increased in young mice compared with adults (P<0.01) and pre-injection of NS-102 significantly prevented this sevoflurane-induced hyperactivity. In electrophysiological experiments, sevoflurane significantly increased the frequency of mEPSP at low concentrations and evoked kainate currents at high concentrations. CONCLUSIONS: We developed a behavioural model in mice that enabled characterization of sevoflurane-induced hyperactivity. The kainate (GluK2) receptor antagonist attenuated these sevoflurane-induced hyperactivities in vivo, suggesting that kainate receptors might be the underlying therapeutic targets for sevoflurane-induced hyperactivities in general anaesthesia.


Subject(s)
Anesthesia Recovery Period , Anesthetics, Inhalation/pharmacology , Hippocampus/drug effects , Psychomotor Agitation/etiology , Receptors, Kainic Acid/metabolism , Sevoflurane/pharmacology , Animals , Disease Models, Animal , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/genetics , GluK2 Kainate Receptor
5.
Neurosci Lett ; 643: 103-110, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28229936

ABSTRACT

Intermediate filaments (IF) can be altered under disorders such as neurodegenerative diseases. Kainic acid (KA) induce behavioral changes and histopathological alterations of the spinal cord of injected rats. Our goal was to evaluate the IF expression in neurons during this injury model. Animals were injected with KA at the C5 segment of the cervical spinal cord and euthanized at 1, 3 and 7 post injection (pi) days. Neuronal cell counting showed a significant loss of neurons at the injection site when compared with those of sham and non-operated animals. Immunohistochemistry for vimentin and neurofilament showed positive labeling of perikarya in sham and KA-injected animals since day 1 pi that lasted for the remaining experimental days. Colocalization analysis between enolase and vimentin or neurofilament confirmed a high index of colocalization in both experimental groups at day 1 pi. This index decreased in sham animals by day 3 pi whereas that of KA-injected animals remained high throughout the experiment. These results may suggest that perikarya initiate an unconventional IF expression, which may respond to the neuronal damage induced by the mechanical injury and the excitotoxic effect of KA. It seems that vimentin and neurofilament expression may be a necessary change to promote recovery of the damaged tissue.


Subject(s)
Intermediate Filaments/drug effects , Kainic Acid/pharmacology , Neurons/drug effects , Spinal Cord/metabolism , Vimentin/metabolism , Animals , Axons/metabolism , Excitatory Amino Acid Agonists/pharmacology , Intermediate Filaments/metabolism , Kainic Acid/administration & dosage , Male , Neurofilament Proteins/metabolism , Neurons/metabolism , Rats, Sprague-Dawley , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/metabolism , Spinal Cord/drug effects
6.
Neurosci Bull ; 33(3): 273-280, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28161868

ABSTRACT

Accumulating evidence has suggested resveratrol as a promising drug candidate for the treatment of epilepsy. To validate this, we tested the protective effect of resveratrol on a kainic acid (KA)-induced epilepsy model in rats and investigated the underlying mechanism. We found that acute resveratrol application partially inhibited evoked epileptiform discharges in the hippocampal CA1 region. During acute, silent and chronic phases of epilepsy, the expression of hippocampal kainate glutamate receptor (GluK2) and the GABAA receptor alpha1 subunit (GABAAR-alpha1) was up-regulated and down-regulated, respectively. Resveratrol reversed these effects and induced an antiepileptic effect. Furthermore, in the chronic phase, resveratrol treatment inhibited the KA-induced increased glutamate/GABA ratio in the hippocampus. The antiepileptic effects of resveratrol may be partially attributed to the reduction of glutamate-induced excitotoxicity and the enhancement in GABAergic inhibition.


Subject(s)
Anticonvulsants/pharmacology , CA1 Region, Hippocampal , Epilepsy, Temporal Lobe , Glutamic Acid/drug effects , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , gamma-Aminobutyric Acid/drug effects , Animals , Anticonvulsants/administration & dosage , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Disease Models, Animal , Down-Regulation , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/metabolism , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Male , Neuroprotective Agents/administration & dosage , Rats , Rats, Wistar , Receptors, GABA-A/drug effects , Receptors, Kainic Acid/drug effects , Resveratrol , Stilbenes/administration & dosage , Up-Regulation , GluK2 Kainate Receptor
7.
J Med Chem ; 59(1): 448-61, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26653877

ABSTRACT

A series of racemic aryl-substituted phenylalanines was synthesized and evaluated in vitro at recombinant rat GluA1-3, at GluK1-3, and at native AMPA receptors. The individual enantiomers of two target compounds, (RS)-2-amino-3-(3,4-dichloro-5-(5-hydroxypyridin-3-yl)phenyl)propanoic acid 37 and (RS)-2-amino-3-(3'-hydroxybiphenyl-3-yl)propanoic acid 38, were characterized. (S)-37 and (R)-38 were identified as the only biologically active isomers, both being antagonists at GluA2 receptors with Kb of 1.80 and 3.90 µM, respectively. To address this difference in enantiopharmacology, not previously seen for amino acid-based AMPA receptor antagonists, X-ray crystal structures of both eutomers in complex with the GluA2 ligand binding domain were solved. The cocrystal structures of (S)-37 and (R)-38 showed similar interactions of the amino acid parts but unexpected and different orientations and interactions of the biaromatic parts of the ligands inside the binding site, with (R)-38 having a binding mode not previously identified for amino acid-based antagonists.


Subject(s)
Excitatory Amino Acid Antagonists/chemical synthesis , Excitatory Amino Acid Antagonists/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Receptors, AMPA/metabolism , Animals , Binding Sites , Crystallography, X-Ray , In Vitro Techniques , Models, Molecular , Molecular Docking Simulation , Phenylalanine/chemical synthesis , Rats , Receptors, AMPA/drug effects , Receptors, Kainic Acid/drug effects , Recombinant Proteins , Structure-Activity Relationship , Xenopus laevis
8.
Pharmacol Res ; 101: 65-73, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26255765

ABSTRACT

Ligand-gated ion channels (LGICs) are cell surface integral proteins that mediate the fast neurotransmission in the nervous system. LGICs require auxiliary subunits for their trafficking, assembly and pharmacological modulation. Auxiliary subunits do not form functional homomeric receptors, but are reported to assemble with the principal subunits in order to modulate their pharmacological profiles. For example, nACh receptors are built at least by co-assemble of α and ß subunits, and the neuronal auxiliary subunits ß3 and α5 and muscle type ß, δ, γ, and ϵ determine the agonist affinity of these receptors. Serotonergic 5-HT3B, 5-HT3C, 5-HT3D and 5-HT3E are reported to assemble with the 5-HT3A subunit to modulate its pharmacological profile. Functional studies evaluating the role of γ2 and δ auxiliary subunits of GABAA receptors have made important advances in the understanding of the action of benzodiazepines, ethanol and neurosteroids. Glycine receptors are composed principally by α1-3 subunits and the auxiliary subunit ß determines their synaptic location and their pharmacological response to propofol and ethanol. NMDA receptors appear to be functional as heterotetrameric channels. So far, the existence of NMDA auxiliary subunits is controversial. On the other hand, Kainate receptors are modulated by NETO 1 and 2. AMPA receptors are modulated by TARPs, Shisa 9, CKAMP44, CNIH2-3 auxiliary proteins reported that controls their trafficking, conductance and gating of channels. P2X receptors are able to associate with auxiliary Pannexin-1 protein to modulate P2X7 receptors. Considering the pharmacological relevance of different LGICs auxiliary subunits in the present work we will highlight the therapeutic potential of these modulator proteins.


Subject(s)
Ligand-Gated Ion Channels/drug effects , Animals , Humans , Ion Channel Gating/drug effects , Ligand-Gated Ion Channels/chemistry , Ligand-Gated Ion Channels/metabolism , Models, Molecular , Protein Subunits , Receptors, AMPA/chemistry , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, Glutamate/chemistry , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Receptors, Glycine/chemistry , Receptors, Glycine/drug effects , Receptors, Glycine/metabolism , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Receptors, Purinergic P2X/chemistry , Receptors, Purinergic P2X/drug effects , Receptors, Purinergic P2X/metabolism , Receptors, Serotonin, 5-HT3/chemistry , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/metabolism
9.
Org Lett ; 17(16): 3972-4, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26258884

ABSTRACT

Dysiherbaine, a natural product isolated from the Marine sponge Dysidea herbacea, has been shown to be a selective agonist of non-NMDA type glutamate receptors, kainate receptors. An enantioselective synthesis of dysiherbaine is reported. Metathesis of the diene followed by conversion of the resulting alkene to the amino alcohol and addition of the amino acid provides the natural product. This synthesis differs from previous approaches to the molecule in that the functionality on the tetrahydropyran ring is installed late in the route.


Subject(s)
Alanine/analogs & derivatives , Biological Products/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Porifera/chemistry , Receptors, Kainic Acid/drug effects , Alanine/chemical synthesis , Alanine/chemistry , Alanine/pharmacology , Alkenes/chemistry , Amino Acids/chemistry , Animals , Biological Products/chemistry , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Marine Biology , Molecular Structure , Receptors, Glutamate/drug effects , Stereoisomerism
10.
J Physiol ; 592(7): 1457-77, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24396054

ABSTRACT

Postsynaptic kainate receptors mediate excitatory synaptic transmission over a broad range of temporal frequencies. In heterologous systems, the temporal responses of kainate receptors vary when different channel-forming and auxiliary subunits are co-expressed but how this variability relates to the temporal differences at central synapses is incompletely understood. The mammalian cone photoreceptor synapse provides advantages for comparing the different temporal signalling roles of kainate receptors, as cones release glutamate over a range of temporal frequencies, and three functionally distinct Off bipolar cell types receive cone signals at synapses that contain either AMPA or kainate receptors, all with different temporal properties. A disadvantage is that the different receptor subunits are not identified. We used in situ hybridization, immunocytochemistry, and pharmacology to identify the kainate receptor and auxiliary subunits in ground squirrel (Ictidomys tridecimlineatus) cb1a/b, cb2, and cb3a/b Off bipolar cell types. As expected, the types showed distinct subunit expression patterns. Kainate receptors mediated ∼80% of the synaptic response in cb3a/b cells and were heteromers of GluK1 and GluK5. Cb3a/b cells contained message for GluK1 and GluK5, and also GluK3 and the auxiliary subunit Neto1. The synaptic responses in cb1a/b cells were mediated by GluK1-containing kainate receptors that behaved differently from the receptors expressed by cb3a/b cells. AMPA receptors mediated the entire synaptic response in cb2 cells and the remaining synaptic response in cb3a/b cells. We conclude that GluK1 is the predominant kainate receptor subunit in cb1 and cb3 Off bipolar cells. Different temporal response properties may result from selective association with GluK3, GluK5, or Neto1.


Subject(s)
Receptors, Kainic Acid/metabolism , Retinal Bipolar Cells/metabolism , Synaptic Transmission , Animals , Excitatory Postsynaptic Potentials , Glutamic Acid/metabolism , Protein Subunits , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/genetics , Retinal Bipolar Cells/drug effects , Retinal Cone Photoreceptor Cells/metabolism , Sciuridae , Time Factors , GluK3 Kainate Receptor
11.
J Neurosci ; 33(22): 9536-45, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719820

ABSTRACT

Kainate receptors (KARs) are ionotropic glutamate receptors that also activate noncanonical G-protein-coupled signaling pathways to depress the slow afterhyperpolarization (sAHP). Here we show that long-term depression of KAR-mediated synaptic transmission (KAR LTD) at rat hippocampal mossy fiber synapses relieves inhibition of the sAHP by synaptic transmission. KAR LTD is induced by high-frequency mossy fiber stimulation and natural spike patterns and requires activation of adenosine A2A receptors. Natural spike patterns also cause long-term potentiation of NMDA receptor-mediated synaptic transmission that overrides the effects of KAR LTD on the cellular response to low-frequency synaptic input. However, KAR LTD is dominant at higher frequency synaptic stimulation where it decreases the cellular response by relieving inhibition of the sAHP. Thus we describe a form of glutamate receptor plasticity induced by natural spike patterns whose primary physiological function is to regulate cellular excitability.


Subject(s)
Neuronal Plasticity/physiology , Receptors, Kainic Acid/physiology , Synapses/physiology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/physiology , Data Interpretation, Statistical , Electric Stimulation , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Male , Mossy Fibers, Hippocampal/drug effects , Patch-Clamp Techniques , Pyramidal Cells/physiology , Rats , Rats, Wistar , Receptor, Adenosine A2A/physiology , Receptors, Kainic Acid/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/drug effects
12.
J Neurochem ; 126(5): 565-78, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23692284

ABSTRACT

We have investigated the mechanisms underlying the facilitatory modulation mediated by kainate receptor (KAR) activation in the cortex, using isolated nerve terminals (synaptosomes) and slice preparations. In cortical nerve terminals, kainate (KA, 100 µM) produced an increase in 4-aminopyridine (4-AP)-evoked glutamate release. In thalamocortical slices, KA (1 µM) produced an increase in the amplitude of evoked excitatory post-synaptic currents (eEPSCs) at synapses established between thalamic axon terminals from the ventrobasal nucleus onto stellate neurons of L4 of the somatosensory cortex. In both, synaptosomes and slices, the effect of KA was antagonized by 6-cyano-7-nitroquinoxaline-2,3-dione, and persisted after pre-treatment with a cocktail of antagonists of other receptors whose activation could potentially have produced facilitation of release indirectly. Mechanistically, the observed effects of KA appear to be congruent in synaptosomal and slice preparations. Thus, the facilitation by KA of synaptosomal glutamate release and thalamocortical synaptic transmission were suppressed by the inhibition of protein kinase A and occluded by the stimulation of adenylyl cyclase. Dissecting this G-protein-independent regulation further in thalamocortical slices, the KAR-mediated facilitation of synaptic transmission was found to be sensitive to the block of Ca(2+) permeant KARs by philanthotoxin. Intriguingly, the synaptic facilitation was abrogated by depletion of intracellular Ca(2+) stores by thapsigargin, or inhibition of Ca(2+) -induced Ca(2+) -release by ryanodine. Thus, the KA-mediated modulation was contingent on both Ca(2+) entry through Ca(2+) -permeable KARs and liberation of intracellular Ca(2+) stores. Finally, sensitivity to W-7 indicated that the increased cytosolic [Ca(2+) ] underpinning KAR-mediated regulation of synaptic transmission at thalamocortical synapses, requires downstream activation of calmodulin. We conclude that neocortical pre-synaptic KARs mediate the facilitation of glutamate release and synaptic transmission by a Ca(2+) -calmodulin dependent activation of an adenylyl cyclase/cAMP/protein kinase A signalling cascade, independent of G-protein involvement.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/physiology , Cerebral Cortex/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Glutamates/metabolism , Receptors, Kainic Acid/physiology , Receptors, Presynaptic/physiology , Synapses/physiology , Thalamus/physiology , Algorithms , Animals , Cerebral Cortex/drug effects , Cyclic AMP/metabolism , Data Interpretation, Statistical , Electrophysiological Phenomena , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Patch-Clamp Techniques , Receptors, Kainic Acid/drug effects , Receptors, Presynaptic/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptosomes/metabolism , Thalamus/drug effects
13.
Glycobiology ; 23(4): 412-25, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23213112

ABSTRACT

Here we report the bioactivity-guided isolation of novel galectins from the marine sponge Cinachyrella sp., collected from Iriomote Island, Japan. The lectin proteins, which we refer to as the Cinachyrella galectins (CchGs), were identified as the active principles in an aqueous sponge extract that modulated the function of mammalian ionotropic glutamate receptors. Aggregation of rabbit erythrocytes by CchGs was competed most effectively by galactosides but not mannose, a profile characteristic of members of the galectin family of oligosaccharide-binding proteins. The lectin activity was remarkably stable, with only a modest loss in hemagglutination after exposure of the protein to 100°C for 1 h, and showed little sensitivity to calcium concentration. CchG-1 and -2 appeared as 16 and 18 kDa in sodium dodecyl sulfate-polyacrylamide gel electrophoresis, respectively, whereas matrix-assisted laser desorption ionization-time-of-flight-mass spectrometry indicated broad ion clusters centered at 16,216 and 16,423, respectively. The amino acid sequences of the CchGs were deduced using a combination of Edman degradation and cDNA cloning and revealed that the proteins were distant orthologs of animal prototype galectins and that multiple isolectins comprised the CchGs. One of the isolectins was expressed as a recombinant protein and exhibited physico-chemical and biological properties comparable with those of the natural lectins. The biochemical properties of the CchGs as well as their unexpected activity on mammalian excitatory amino acid receptors suggest that further analysis of these new members of the galectin family will yield further glycobiological and neurophysiological insights.


Subject(s)
Galectins/pharmacology , Porifera/chemistry , Receptors, AMPA/drug effects , Receptors, Kainic Acid/drug effects , Action Potentials/drug effects , Amino Acid Sequence , Animals , Calcium/pharmacology , Galactosides/immunology , Galectins/chemistry , Galectins/immunology , Galectins/isolation & purification , HEK293 Cells , Hemagglutination , Humans , Male , Mannose/immunology , Mice , Molecular Sequence Data , Phylogeny , Protein Binding , Rabbits
14.
J Neurosci ; 32(15): 5186-99, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496564

ABSTRACT

Tissue-type plasminogen activator (tPA) regulates physiological processes in the brain, such as learning and memory, and plays a critical role in neuronal survival and neuroinflammation in pathological conditions. Here we demonstrate, by combining mouse in vitro and in vivo data, that tPA is an important element of the cross talk between neurons and astrocytes. The data show that tPA released by neurons is constitutively endocytosed by astrocytes via the low-density lipoprotein-related protein receptor, and is then exocytosed in a regulated manner. The exocytotic recycling of tPA by astrocytes is inhibited in the presence of extracellular glutamate. Kainate receptors of astrocytes act as sensors of extracellular glutamate and, via a signaling pathway involving protein kinase C, modulate the exocytosis of tPA. Further, by thus capturing extracellular tPA, astrocytes serve to reduce NMDA-mediated responses potentiated by tPA. Overall, this work provides the first demonstration that the neuromodulator, tPA, may also be considered as a gliotransmitter.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Glutamic Acid/pharmacology , Glutamic Acid/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Plasminogen Activator/metabolism , Albumins/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Clathrin/physiology , Dynamins/physiology , Endocytosis/drug effects , Endocytosis/physiology , Flow Cytometry , Gene Silencing , Immunohistochemistry , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Plasmids/genetics , Protein Kinase C/metabolism , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/metabolism , Receptors, LDL/metabolism , Synapsins/metabolism , Transfection , Tumor Suppressor Proteins/metabolism , alpha-Macroglobulins/metabolism
15.
Brain Res ; 1456: 1-13, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22516108

ABSTRACT

It is well known that GluK2-containing kainate receptors play essential roles in seizure and cerebral ischemia-induced neuronal death, while GluK1-containing kainate receptors could increase tonic inhibition of post-synaptic pyramidal neurons. This research investigated whether GluK1 could inhibit activation of c-Jun N-terminal kinase 3 (JNK3) signaling pathway mediated by the GluK2 in cerebral ischemia-reperfusion. The results show that GluK1 activation by (RS)-2-amino-3-(3-hydroxy-5-tert-butylisoxazol-4-yl) propanoic acid (ATPA) at 1nmol per rat could inhibit the assembly of GluK2·Postsynaptic density 95·mixed lineage kinase 3 signaling module, activation of JNK3 and its downstream signal molecules. However, the inhibition of ATPA could be prevented by GluK1 antagonist NS3763, GluK1 antisense, and GABA(A) receptor antagonist bicuculline. In addition, ATPA played a neuroprotective role against cerebral ischemia. In sum, the findings indicate that activation of GluK1 by ATPA at specific dosages may promote GABA release, which then suppresses post-synaptic GluK2-JNK3 signaling-mediated cerebral ischemic injury via GABA(A)R.


Subject(s)
Isoxazoles/pharmacology , Neuroprotective Agents/pharmacology , Propionates/pharmacology , Receptors, Kainic Acid/agonists , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Animals , Enzyme Activation/physiology , Male , Mitogen-Activated Protein Kinase 10/drug effects , Mitogen-Activated Protein Kinase 10/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/metabolism , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , GluK2 Kainate Receptor
16.
Basic Clin Pharmacol Toxicol ; 111(1): 4-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22429661

ABSTRACT

Several studies have revealed that acute ethanol inhibits the function of glutamate receptors. Glutamate receptor-mediated synaptic plasticity, such as N-methyl-D-aspartate-dependent long-term potentiation, is also inhibited by ethanol. However, the inhibition seems to be restricted to certain brain areas such as the hippocampus, amygdala and striatum. Ethanol inhibition of glutamate receptors generally requires relatively high concentrations and may therefore explain consequences of severe ethanol intoxication such as impairment of motor performance and memory. Effects of ethanol on glutamate system of developing nervous system may have a role in causing foetal alcohol syndrome. Newly found regulatory proteins of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid AMPA receptors seem to affect ethanol inhibition thus opening new lines of research.


Subject(s)
Ethanol/toxicity , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Long-Term Potentiation/drug effects , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/drug effects , N-Methylaspartate/metabolism , Neuronal Plasticity/drug effects , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/drug effects , Toxicity Tests, Acute
17.
J Pharmacol Exp Ther ; 340(3): 733-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22171094

ABSTRACT

Dystonia is a neurological disorder characterized by involuntary muscle contractions that cause twisting movements and abnormal postures. Functional imaging consistently reveals cerebellar overactivity in dystonic patients regardless of the type or etiology of the disorder. To explore mechanisms that might explain the basis for the cerebellar overactivity in dystonia, normal mice were challenged with intracerebellar application of a variety of agents that induce hyperexcitability. A nonspecific increase in cerebellar excitability, such as that produced by picrotoxin, was not associated with dystonia. Instead, glutamate receptor activation, specifically AMPA receptor activation, was necessary to evoke dystonia. AMPA receptor agonists induced dystonia, and AMPA receptor antagonists reduced the dystonia induced by glutamate receptor agonists. AMPA receptor antagonists also ameliorated the dystonia exhibited by the dystonic mouse mutant tottering, suggesting that AMPA receptors may play a role in some other genetic models of dystonia. Furthermore, AMPA receptor desensitization mediated the expression of dystonia. Preventing AMPA receptor desensitization with cyclothiazide or the nondesensitizing agonist kainic acid exacerbated the dystonic response. These results suggest the novel hypothesis that the cerebellar overactivity observed in neuroimaging studies of patients with dystonia may be an indirect reflection of abnormal glutamate signaling. In addition, these results imply that reducing AMPA receptor activation by blocking AMPA receptors and promoting AMPA receptor desensitization or negative allosteric modulators may prove to be beneficial for treating dystonia.


Subject(s)
Cerebellum/drug effects , Dystonia/chemically induced , Receptors, AMPA/drug effects , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , 4-Aminopyridine/pharmacology , Animals , Benzothiadiazines/pharmacology , Cerebellum/physiology , Dose-Response Relationship, Drug , Dystonia/drug therapy , Female , Male , Mice , Mice, Inbred C57BL , Quisqualic Acid/pharmacology , Receptors, AMPA/physiology , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/physiology
18.
Brain Res ; 1427: 10-22, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22071563

ABSTRACT

It has been known that retinal ganglion cells (RGCs) with distinct morphologies have different physiological properties. It was hypothesized that different functions of RGCs may in part result from various expressions of N-methyl-d-aspartate (NMDA), α-amino-3-hydroxyl-5-methyl-isoxazole-4-propinoic acid (AMPA), and kainic acid (KA) receptors on their dendrites. In the present study, we aimed to characterize the functional expression of AMPA and NMDA receptors of morphologically identified RGCs in the wholemount rabbit retina. The agmatine (AGB) activation assay was used to reveal functional expression of ionotropic glutamate receptors after the RGCs were targeted by injecting Neurobiotin. To examine the excitability of these glutamate receptors in an agonist specific manner, the lower concentrations of AMPA (2 µM) and NMDA (100 µM) were chosen to examine G7 (ON-OFF direction selective ganglion cells) and G11 (alpha ganglion cells) types of RGCs. We found that less than 40% of G7 type RGCs had salient AGB activation when incubated with 2 µM AMPA or 100 µM NMDA. The G11 type RGCs also showed similar activation frequencies, except that all of the OFF subtype examined had no AGB permeation under the same AMPA concentration. These results suggest that RGCs with large somata (G7 and G11 types) may express various heterogeneous functional ionotropic glutamate receptors, thus in part rendering their functional diversity.


Subject(s)
Receptors, Ionotropic Glutamate/physiology , Retinal Ganglion Cells/physiology , Synaptic Transmission/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/pharmacology , Organ Culture Techniques , Rabbits , Receptors, AMPA/drug effects , Receptors, AMPA/physiology , Receptors, Ionotropic Glutamate/biosynthesis , Receptors, Ionotropic Glutamate/genetics , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Synaptic Transmission/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
19.
Pharmacol Rep ; 63(4): 949-55, 2011.
Article in English | MEDLINE | ID: mdl-22001982

ABSTRACT

In the present study, the role of kainate (KA) receptors in hypnosis and analgesia induced by emulsified inhalation anesthetics was investigated. A mouse model of hypnosis and analgesia was established by an intraperitoneal injection of emulsified enflurane, isoflurane or sevoflurane. We intracerebroventricularly (icv) or intrathecally (it) administered KA, a KA receptor agonist to mice. The effects of the KA on the sleep time were observed using a hypnosis test, and the tail-withdrawal latency was analyzed using the tail-withdrawal test. In the hypnosis test, KA (2.5, 5 or 10 ng; icv administered) treatment had no distinctive effects on the sleep time of mice treated with emulsified inhalation anesthetics. In the tail-withdrawal test, KA (0.2, 0.4 or 0.8 ng; it administered) treatment significantly and dose-dependently decreased the tail-withdrawal latency of mice treated with emulsified anesthetics. These results suggested that KA receptors may modulate the analgesic but not hypnotic effects induced by emulsified enflurane, isoflurane or sevoflurane.


Subject(s)
Analgesics/pharmacology , Anesthetics, Inhalation/pharmacology , Kainic Acid/pharmacology , Receptors, Kainic Acid/drug effects , Analgesics/administration & dosage , Anesthetics, Inhalation/administration & dosage , Animals , Dose-Response Relationship, Drug , Emulsions , Enflurane/administration & dosage , Enflurane/pharmacology , Female , Hypnosis, Anesthetic/methods , Injections, Intraventricular , Injections, Spinal , Isoflurane/administration & dosage , Isoflurane/pharmacology , Kainic Acid/administration & dosage , Male , Methyl Ethers/administration & dosage , Methyl Ethers/pharmacology , Mice , Receptors, Kainic Acid/metabolism , Sevoflurane , Sleep/drug effects
20.
Pharmacol Biochem Behav ; 99(4): 775-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21763717

ABSTRACT

Polyamines (putrescine, spermidine and spermine) are important endogenous regulators of ion channels, such as vanilloid (TRPV1), glutamatergic (NMDA or AMPA/kainate) and acid-sensitive (ASIC) receptors. In the present study, we have investigated the possible nociceptive effect induced by polyamines and the mechanisms involved in this nociception in vivo. The subcutaneous (s.c.) injection of capsaicin (as positive control), spermine, spermidine or putrescine produced nociception with ED(50) of 0.16 (0.07-0.39)nmol/paw, 0.4 (0.2-0.7) µmol/paw, 0.3 (0.1-0.9) µmol/paw and 3.2 (0.9-11.5) µmol/paw, respectively. The antagonists of NMDA (MK801, 1 nmol/paw), AMPA/kainate (DNQX, 1 nmol/paw) or ASIC receptors (amiloride, 100 nmol/paw) failed to reduce the spermine-trigged nociception. However, the TRPV1 antagonists capsazepine or SB366791 (1 nmol/paw) reduced spermine-induced nociception, with inhibition of 81 ± 10 and 68 ± 9%, respectively. The previous desensitization with resiniferatoxin (RTX) largely reduced the spermine-induced nociception and TRPV1 expression in the sciatic nerve, with reductions of 82 ± 9% and 67 ± 11%, respectively. Furthermore, the combination of spermine (100 nmol/paw) and RTX (0.005 fmol/paw), in doses which alone were not capable of inducing nociception, produced nociceptive behaviors. Moreover, different concentrations of spermine (3-300 µM) enhanced the specific binding of [(3)H]-RTX to TRPV1 receptor. Altogether, polyamines produce spontaneous nociceptive effect through the stimulation of TRPV1, but not of ionotropic glutamate or ASIC receptors.


Subject(s)
Nociceptors/drug effects , Spermine/pharmacology , TRPV Cation Channels/drug effects , Acid Sensing Ion Channels , Animals , Biogenic Polyamines/pharmacology , Blotting, Western , Diterpenes/metabolism , Diterpenes/pharmacology , Glutamic Acid/pharmacology , Male , Mice , Nerve Fibers/drug effects , Nerve Tissue Proteins/drug effects , Pain/chemically induced , Pain/prevention & control , Pain Measurement/drug effects , Receptors, AMPA/drug effects , Receptors, Kainic Acid/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Sodium Channels/drug effects , Spermine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...