Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Mol Psychiatry ; 17(1): 71-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21606927

ABSTRACT

Genetic mutations of SHANK3 have been reported in patients with intellectual disability, autism spectrum disorder (ASD) and schizophrenia. At the synapse, Shank3/ProSAP2 is a scaffolding protein that connects glutamate receptors to the actin cytoskeleton via a chain of intermediary elements. Although genetic studies have repeatedly confirmed the association of SHANK3 mutations with susceptibility to psychiatric disorders, very little is known about the neuronal consequences of these mutations. Here, we report the functional effects of two de novo mutations (STOP and Q321R) and two inherited variations (R12C and R300C) identified in patients with ASD. We show that Shank3 is located at the tip of actin filaments and enhances its polymerization. Shank3 also participates in growth cone motility in developing neurons. The truncating mutation (STOP) strongly affects the development and morphology of dendritic spines, reduces synaptic transmission in mature neurons and also inhibits the effect of Shank3 on growth cone motility. The de novo mutation in the ankyrin domain (Q321R) modifies the roles of Shank3 in spine induction and morphology, and actin accumulation in spines and affects growth cone motility. Finally, the two inherited mutations (R12C and R300C) have intermediate effects on spine density and synaptic transmission. Therefore, although inherited by healthy parents, the functional effects of these mutations strongly suggest that they could represent risk factors for ASD. Altogether, these data provide new insights into the synaptic alterations caused by SHANK3 mutations in humans and provide a robust cellular readout for the development of knowledge-based therapies.


Subject(s)
Actins/metabolism , Carrier Proteins/genetics , Dendrites/ultrastructure , Dendritic Spines/genetics , Mutation/genetics , Neurons/cytology , Animals , Autistic Disorder/genetics , Cell Line, Transformed/cytology , Cells, Cultured , Chlorocebus aethiops , Dendrites/genetics , Dendritic Spines/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , Microscopy, Confocal , Nerve Tissue Proteins , Transfection , Tubulin/metabolism
2.
Neuron ; 8(4): 653-62, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1314618

ABSTRACT

We studied the effects of nitric oxide (NO)-producing agents on N-methyl-D-aspartate (NMDA) receptor activation in cultured neurons. 3-Morpholino-sydnonimine (SIN-1) blocked both NMDA-induced currents and the associated increase in intracellular Ca2+. The actions of SIN-1 were reversible and suppressed by hemoglobin. A degraded SIN-1 solution that did not release NO was unable to block NMDA receptors. This showed that the SIN-1 effects were due to NO and not to another breakdown product. Similar results were obtained with 1-nitrosopyrrolidine (an NO-containing drug) and with NO released from NaNO2. Pretreatment with hemoglobin potentiated NMDA-induced effects, demonstrating that endogenous NO modulates NMDA receptors. Since NMDA receptor activation induces NO synthesis, these results suggest a feedback inhibition of NMDA receptors by NO under physiological condition.


Subject(s)
Nitric Oxide/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Calcium/metabolism , Cyclic GMP/biosynthesis , In Vitro Techniques , Mice , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , N-Methylaspartate/pharmacology , N-Nitrosopyrrolidine/pharmacology , Nitroso Compounds/pharmacology , Sodium Nitrite/pharmacology
3.
Trends Neurosci ; 23(2): 80-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10652549

ABSTRACT

Metabotropic glutamate receptors (mGluRs) can increase intracellular Ca2+ concentration via Ins(1,4,5)P3- and ryanodine-sensitive Ca2+ stores in neurons. Both types of store are coupled functionally to Ca2+-permeable channels found in the plasma membrane. The mGluR-mediated increase in intracellular Ca2+ concentration can activate Ca2+-sensitive K+ channels and Ca2+-dependent nonselective cationic channels. These mGluR-mediated effects often result from mobilization of Ca2+ from ryanodine-sensitive, rather than Ins(1,4, 5)P3-sensitive, Ca2+ stores, suggesting that close functional interactions exist between mGluRs, intracellular Ca2+ stores and Ca2+-sensitive ion channels in the membrane.


Subject(s)
Calcium/metabolism , Ion Channels/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Calcium Channels/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Homer Scaffolding Proteins , Humans , Neuronal Plasticity , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/metabolism , Potassium Channels/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
4.
J Neurosci ; 20(21): 7896-904, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11050109

ABSTRACT

Although presynaptic localization of mGluR7 is well established, the mechanism by which the receptor may control Ca(2+) channels in neurons is still unknown. We show here that cultured cerebellar granule cells express native metabotropic glutamate receptor type 7 (mGluR7) in neuritic processes, whereas transfected mGluR7 was also expressed in cell bodies. This allowed us to study the effect of the transfected receptor on somatic Ca(2+) channels. In transfected neurons, mGuR7 selectively inhibited P/Q-type Ca(2+) channels. The effect was mimicked by GTPgammaS and blocked by pertussis toxin (PTX) or a selective antibody raised against the G-protein alphao subunit, indicating the involvement of a G(o)-like protein. The mGuR7 effect did not display the characteristics of a direct interaction between G-protein betagamma subunits and the alpha1A Ca(2+) channel subunit, but was abolished by quenching betagamma subunits with specific intracellular peptides. Intracellular dialysis of G-protein betagamma subunits did not mimic the action of mGluR7, suggesting that both G-protein betagamma and alphao subunits were required to mediate the effect. Inhibition of phospholipase C (PLC) blocked the inhibitory action of mGluR7, suggesting that a coincident activation of PLC by the G-protein betagamma with alphao subunits was required. The Ca(2+) chelator BAPTA, as well as inhibition of either the inositol trisphosphate (IP(3)) receptor or protein kinase C (PKC) abolished the mGluR7 effect. Moreover, activation of native mGluR7 induced a PTX-dependent IP(3) formation. These results indicated that IP(3)-mediated intracellular Ca(2+) release was required for PKC-dependent inhibition of the Ca(2+) channels. Possible control of synaptic transmission by the present mechanisms is discussed.


Subject(s)
Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Type C Phospholipases/metabolism , Animals , Antibodies/pharmacology , Barium/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Heterotrimeric GTP-Binding Proteins/metabolism , Intracellular Fluid/metabolism , Mice , Neurites/metabolism , Neurons/cytology , Patch-Clamp Techniques , Pertussis Toxin , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/drug effects , Transfection , Virulence Factors, Bordetella/pharmacology
5.
J Neurosci ; 20(23): 8710-6, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11102477

ABSTRACT

The physiological actions of neurotransmitter receptors are intimately linked to their proper neuronal compartment localization. Here we studied the effect of the metabotropic glutamate receptor (mGluR)-interacting proteins, Homer1a, b, and c, in the targeting of mGluR5 in neurons. We found that mGluR5 was exclusively localized in cell bodies when transfected alone in cultured cerebellar granule cells. In contrast, mGluR5 was found also in dendrites when coexpressed with Homer1b or Homer1c, and in both dendrites and axons when cotransfected with Homer1a. In dendrites, cotransfected mGluR5 and Homer1b/c formed clusters that colocalized with the synaptic marker synaptophysin. Interestingly when transfected alone, the Homer proteins were also translocated to neurites but did not form such clusters. Depolarization of the neurons with a mixture of ionotropic glutamate receptor agonists, NMDA and kainate, or potassium channel blockers, tetraethylammonium and 4-aminopyridine, induced transient expression of endogenous Homer1a and persistent neuritic localization of transfected mGluR5 even long after degradation of Homer1a. These results suggest that Homer1a/b/c proteins are involved in the targeting of mGluR5 to dendritic synaptic sites and/or axons and that this effect can be regulated by neuronal activity. Because the activity-dependent effect of endogenous Homer1a was also long-lasting, the axonal targeting of mGluR5 by this protein is likely to play an important role in synaptic plasticity.


Subject(s)
Axons/metabolism , Carrier Proteins/metabolism , Dendrites/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Excitatory Amino Acid Agonists/pharmacology , Homer Scaffolding Proteins , Humans , Kainic Acid/metabolism , Kainic Acid/pharmacology , Mice , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Neurites/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Neuropeptides/genetics , Neuropeptides/pharmacology , Potassium Channel Blockers , Protein Binding/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptophysin/metabolism , Transfection
6.
Free Radic Biol Med ; 7(1): 3-8, 1989.
Article in English | MEDLINE | ID: mdl-2546869

ABSTRACT

The action of a reactive oxygen intermediate, that is, hydrogen peroxide (H2O2) on modulation of synaptic transmission was examined in the hippocampal brain slice preparation. Microinjection of H2O2 into the apical dendritic region of the CA1 pyramidal cells produced no change in either the pattern or amplitude of paired pulse facilitation compared to saline injection (control). Long term potentiation (LTP), induced by high frequency stimulation of homosynaptic inputs, however, was blocked by microinjection of H2O2 into the dendritic tree. LTP was seen in only 2 out of 10 slices investigated when treated with H2O2 while LTP was seen in 4 out of 5 slices when saline injected. The results suggest that a reactive oxygen intermediate can selectively modify synaptic mechanisms in the hippocampus.


Subject(s)
Hippocampus/drug effects , Hydrogen Peroxide/pharmacology , Synapses/drug effects , Synaptic Transmission/drug effects , Action Potentials/drug effects , Animals , Corpus Striatum/physiology , Dendrites/drug effects , Dendrites/physiology , Electric Stimulation , Hippocampus/physiology , In Vitro Techniques , Microinjections , Neural Pathways/physiology , Rats , Rats, Inbred Strains , Synapses/physiology
7.
FEBS Lett ; 250(2): 519-22, 1989 Jul 03.
Article in English | MEDLINE | ID: mdl-2473923

ABSTRACT

Charybdotoxin, a short scorpion venom neurotoxin, which was thought to be specific for the blockade of Ca2+-activated K+ channels also blocks a class of voltage-sensitive K+ channels that are known to be the target of other peptide neurotoxins from snake and bee venoms such as dendrotoxin and MCD peptide. Charybdotoxin also inhibits 125I-dendrotoxin and 125I-MCD peptide binding to their receptors. All these effects are observed with an IC50 of about 30 nM.


Subject(s)
Elapid Venoms/antagonists & inhibitors , Neurotoxins/toxicity , Potassium Channels/drug effects , Scorpion Venoms/toxicity , Animals , Cells, Cultured , Charybdotoxin , Electrophysiology , Rats , Synaptosomes/metabolism
8.
Eur J Neurosci ; 3(8): 778-789, 1991.
Article in English | MEDLINE | ID: mdl-12106464

ABSTRACT

Trans-1-amino-cyclopentyl-1,3-dicarboxylic acid (trans-ACPD), a specific agonist of the glutamate phosphoinositide-coupled receptor (Qp receptor), increased the amplitude of the outward K+ current recorded in the whole-cell configuration of the patch-clamp technique in mouse cultured cerebellar granule cells. This effect was abolished by buffering internal Ca2+ with BAPTA [1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid]. Activation of a large-conductance K+ channel was observed when trans-ACPD or quisqualic acid (QA), another Qp receptor agonist, was applied outside the cell-attached patch pipettes. No activation was observed with alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), a specific agonist of ionotropic non-N-methyl-d-aspartate (non-NMDA) receptors. The effects of trans-ACPD or QA were potentiated in the presence of external Ca2+. The channel was also directly activated by both micromolar concentrations of internal Ca2+ and membrane depolarization. Its unitary conductance was 100 - 115 pS under asymmetrical K+ and 195 - 235 pS under high symmetrical K+ conditions. In the absence of agonist, the channel was blocked by 1 mM external tetraethylammonium. This is the first description of a large conductance Ca2+-activated K+ channel in cultured cerebellar granule cells. It possesses properties similar to those of the so-called 'big K+ channel' described in other preparations. Our cell-attached experiments demonstrated an indirect coupling between Qp receptors and this channel. The most likely hypothesis is that the second messenger system inositol 1,4,5-triphosphate (IP3)-Ca2+ was involved in the coupling process. This hypothesis was further strengthened by our whole-cell experiments. On the basis of the voltage- and Ca2+-sensitivities of the studied channel, we estimated an increase of 350 to 570 nM in internal Ca2+ concentration when Qp receptors were stimulated by 100 microM trans-ACPD. Under physiological conditions, stimulation of Qp receptors by the endogenous neurotransmitter should lead to similar K+ channel activation and therefore would tend to reduce the efficacy of ionotropic glutamate synaptic receptor stimulation responsible for cell excitation.

9.
Neuropharmacology ; 27(1): 57-65, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3281053

ABSTRACT

In a previous study, it was shown that helium pressure depressed excitatory synaptic transmission mediated by the Schaffer-commissural afferents and increased the intrinsic excitability of pyramidal cells, in the CA1 region of hippocampal slices in the rat. In the present study, the neurochemical bases of these changes was investigated. Various excitatory amino acids were studied under normal and up to 80 atm of helium. At normal pressure, the amino acids tested induced a decrease in the field excitatory postsynaptic potential (EPSP) and antidromic field potential of CA1 pyramidal cells. These changes probably resulted from the well known depolarizing effect of the compounds. Quisqualate is supposed to activate the synaptic receptors of the pathway tested. Since the effect of this amino acid and other agonists were not significantly affected by helium pressure, it is suggested that the depressed hippocampal synaptic potentials under pressure did not result from reduced sensitivity of synaptic receptors. On the other hand, helium pressure enhanced the action of N-methyl-D-aspartate (NMDA) and depressed the GABA-mediated inhibition of CA1 pyramidal cells. Given that the excitability of these neurones is modulated by NMDA-related events and GABA inhibition, these results indicate that both neurochemical systems were probably involved in the helium pressure-induced hyperexcitability of the cells studied.


Subject(s)
Amino Acids/pharmacology , Atmospheric Pressure , Helium/toxicity , Hippocampus/drug effects , Neural Inhibition/drug effects , gamma-Aminobutyric Acid/physiology , Action Potentials/drug effects , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , High Pressure Neurological Syndrome/physiopathology , In Vitro Techniques , Magnesium/pharmacology , N-Methylaspartate , Rats , Rats, Inbred Strains
10.
Neuropharmacology ; 39(12): 2244-54, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10974308

ABSTRACT

Imidazolines have been shown to be neuroprotective in focal and global ischemia in the rat. However, their mechanism of action is still unclear. We have studied the neuroprotective effects of imidazolines against NMDA-induced neuronal death and hypoxic insult in cerebellar and striatal neuronal cultures. All of the imidazolines tested decreased the NMDA-mediated neurotoxicity in a non-competitive manner. Antazoline was the most effective (IC(50) of 5 microM, maximal neuroprotection reaching 90% at 100 microM). The neuroprotective effects were still present when the imidazolines were applied during the post-insult period. Antazoline, idazoxan and guanabenz also showed neuroprotective effects against hypoxia-induced neuronal death (neuroprotection reaching 95% for antazoline at 100 microM). Antazoline was still active if applied during the reoxygenation period (15% neuroprotection). To determine the mechanism of the neuroprotective effects, the possible interaction of imidazolines with NMDA receptors was studied. Imidazolines dose-dependently and non-competitively inhibited NMDA currents. As found for the neuroprotective effects, antazoline was the most effective imidazoline, with an IC(50) of 4 microM and a maximal inhibition of 90% at 100 microM. This blockade was rapid, reversible and voltage-dependent. We compared these effects to those of the classical non-competitive antagonist of NMDA channels, MK-801. In contrast to imidazolines, blockade of the NMDA current by MK-801 was voltage-independent and reversible only at positive potentials. When co-applied with MK-801, antazoline prevented the long lasting blockade of the NMDA current by MK-801. These results are consistent with the existence of overlapping binding sites for these drugs on the NMDA receptor channel. They indicate that imidazolines exert a strong neuroprotective effect against excitotoxicity and hypoxia in cerebellar and striatal primary neuronal cultures by inhibiting NMDA receptors. Since these effects were non-competitive, imidazolines appear to be interesting new drugs with therapeutic potential.


Subject(s)
Imidazoles/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cell Count , Cell Death/drug effects , Cell Hypoxia/drug effects , Cells, Cultured , Dizocilpine Maleate/pharmacology , Electrophysiology , Excitatory Amino Acid Agonists/toxicity , Excitatory Amino Acid Antagonists/pharmacology , Mice , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/toxicity , Patch-Clamp Techniques
11.
Neuropharmacology ; 34(8): 929-37, 1995 Aug.
Article in English | MEDLINE | ID: mdl-8532174

ABSTRACT

We investigated the mechanisms by which metabotropic glutamate receptors (mGluRs) modulate specific Ca2+ channels in cerebellar granule cells. A large fraction of the current in granule cells is carried by L- and Q-type Ca2+ channels (about 26% each), whereas N- and P-type contribute proportionally less to the global current (9 and 15%, respectively). l-Aminocyclopentane-dicarboxylate (t-ACPD), (2S,3S,4S)-alpha-(carboxycyclopropyl)-glycine (L-CCGI) and (S)-4-carboxy-3-hydroxyphenylglycine [(S)-4C3HPG], but not L(+)-2-amino-4-phosphonobutyrate (L-AP4) reduced the Ca2+ current amplitude. The t-ACPD-induced inhibition was fully antagonized by (+/-)-methyl-4-carboxyphenylglycine [(+/-)-MCPG] and blocked by pertussis toxin (PTX). These results are consistent with inhibitory response mediated by mGluR2/R3. The use of specific Ca2+ channel blockers provided evidence that mGluR2/R3 inhibited both L- and N-type Ca2+ currents. In PTX-treated cells, Glu or t-ACPD, but not L-CCGI or L-AP4, increased the Ca2+ current. Consistent with the activation of mGluR1, the antagonists (+)-MCPG and (S)-4C3HPG prevented the facilitation of Ca2+ current produced by t-ACPD. The mGluR1-activated facilitation was completely blocked by nimodipine, indicating that L-type Ca2+ currents were selectively potentiated.


Subject(s)
Calcium Channels/metabolism , Cerebellum/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cerebellum/cytology , Cerebellum/drug effects , Culture Techniques , Electrophysiology , Male , Mice , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Pertussis Toxin , Receptors, Metabotropic Glutamate/agonists , Virulence Factors, Bordetella/pharmacology
12.
Neuropharmacology ; 33(11): 1267-74, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7532813

ABSTRACT

In striatal neurones in culture, N-methyl-D-aspartate-(NMDA), kainate-(Kai) and K(+)-dependent cGMP production is entirely mediated via nitric oxide (NO). Low concentrations of lavendustin-A (< or = 0.3 microM), a highly specific tyrosine kinase inhibitor, reduced irreversibly and in a time-dependent manner NMDA-stimulated cGMP production. After a preincubation period of 20 min with lavendustin-A (0.3 microM), the inhibition of NMDA-induced cGMP production was equal to 56 +/- 8% (n = 6). After the same preincubation period, the IC50 of the lavendustin-A blockade was 30 +/- 15 nM. Genistein, another tyrosine kinase inhibitor also inhibited NMDA-dependent cGMP production with high potencies (< or = 3 microM). Whatever the tyrosine kinase inhibitor tested, the basal cGMP production remained unaffected. Kai-, K(+)-, and ionomycin-induced cGMP production was also inhibited by lavendustin-A, and genistein. In contrast, tyrosine kinase inhibitors were unable to block NO donor-induced cGMP production. Using patch clamp experiments, we have also found that lavendustin-A (0.3-1 microM), the most potent tyrosine kinase inhibitor used, (a) did not reduce the NMDA receptor-mediated current, (b) only slighly affected Kai receptor-mediated current (16.4 +/- 3.4% inhibition) and (c) had a marked effect on voltage-sensitive Ca2+ channel- (VSCC) mediated currents (44.4 +/- 4.9% inhibition). A reduction in VSCC activity certainly explains the inhibition of K(+)-, Kai- and possibly part of the NMDA-induced cGMP production.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Neurons/metabolism , Nitric Oxide/biosynthesis , Protein-Tyrosine Kinases/antagonists & inhibitors , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Amino Acid Oxidoreductases/biosynthesis , Animals , Calcium Channels/drug effects , Calcium Channels/metabolism , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Cyclic GMP/biosynthesis , Dizocilpine Maleate/pharmacology , Genistein , Isoflavones/pharmacology , Mice , N-Methylaspartate/pharmacology , Neostriatum/drug effects , Neostriatum/metabolism , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/enzymology , Nitric Oxide Synthase , Patch-Clamp Techniques , Phenols/pharmacology
13.
Neuropharmacology ; 38(6): 793-803, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10465683

ABSTRACT

We describe a method to transfer cDNA into neuronal primary cultures with a commercialised cationic lipid, Transfast. Cultures were transfected at a rate of about 5% with green fluorescent protein (GFP) cDNA. Comparing Transfast to other transfection reagents, we found this compound to be the most efficient. GFP-transfected mouse cerebellar granule cells displayed normal whole-cell voltage-sensitive and unitary big K+ channel currents. We also used this transfection method with success to transfer GFP cDNA into primary cultures of striatum and colliculus. Transfast was then used to cotransfect cultured cerebellar cells with GFP cDNA, in conjunction with cDNA coding for the metabotropic glutamate receptor type 5 (mGlu5 receptor). Ninety percent of the cells expressing GFP also expressed mGlu5 receptor. Though neurones were best transfected one day after plating, they still expressed both GFP and mGlu5 receptor proteins 2 weeks after plating, i.e. after full differentiation. A functional test of the expressed mGlu5 receptor was thus performed in GFP-transfected neurones. Stimulation of mGlu5 receptor induced single big K+ channel activity, as it was the case for the native mGlu1 receptor. This indicated that the transfected mGlu5 receptor plasmid was functionally expressed and that both mGlu1 and mGlu5 receptors may share common coupling mechanisms to big K+ channels in neurones.


Subject(s)
Cerebellum/physiology , DNA/genetics , Gene Transfer Techniques , Neurons/physiology , Plasmids/genetics , Receptors, Metabotropic Glutamate/physiology , Animals , Calcium Channels/physiology , Cells, Cultured , Cerebellum/cytology , Mice , Patch-Clamp Techniques , Potassium Channels/physiology
14.
J Med Chem ; 40(19): 3119-29, 1997 Sep 12.
Article in English | MEDLINE | ID: mdl-9301676

ABSTRACT

The four stereoisomers of 1-aminocyclopentane-1,3,4-tricarboxylic acid {ACPT-I (18) and -II (19), (3R, 4R)-III [(-)-20], and (3S,4S)-III [(+)-20]} have been synthesized and evaluated for their effects at glutamate receptors subtypes. ACPTs are ACPD analogues in which a third carboxylic group has been added at position 4 in the cyclopentane ring. None of the ACPT isomers showed a significant effect on ionotropic NMDA, KA, and AMPA receptors. On the other hand, ACPT-II (19) was found to be a general competitive antagonist for metabotropic receptors (mGluRs) and exhibited a similar affinity for mGluR1a (KB = 115 +/- 2 microM), mGluR2 (KB = 88 +/- 21 microM), and mGluR4a (KB = 77 +/- 9 microM), the representative members of group I, II and III mGluRs, respectively. Two other isomers, ACPT-I (18) and (+)-(3S,4S)-ACPT-III [(+)-20], were potent agonists at the group III receptor mGluR4a (EC50 = 7.2 +/- 2.3 and 8.8 +/- 3.2 microM) and competitive antagonists with low affinity for mGluR1a and mGluR2 (KB > 300 microM). Finally, (-)-(3R,4R)-ACPT-III [(-)-20] was a competitive antagonist with poor but significant affinity for mGluR4a (KB = 220 microM). These results demonstrate that the addition of a third carboxylic group to ACPD can change its activity (from agonist to antagonist) and either increase or decrease its selectivity and/or affinity for the various mGluR subtypes.


Subject(s)
Cyclopentanes/chemical synthesis , GABA Agonists/chemical synthesis , GABA Antagonists/chemical synthesis , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Tricarboxylic Acids/chemical synthesis , Animals , Binding, Competitive , Cell Line , Cells, Cultured , Cerebellum/drug effects , Cerebellum/physiology , Cyclopentanes/chemistry , Cyclopentanes/pharmacology , GABA Agonists/chemistry , GABA Agonists/pharmacology , GABA Antagonists/chemistry , GABA Antagonists/pharmacology , Humans , Indicators and Reagents , Inositol/metabolism , Inositol Phosphates/metabolism , Kinetics , Mice , Molecular Conformation , Molecular Structure , Neurons/drug effects , Neurons/physiology , Receptors, Metabotropic Glutamate/classification , Receptors, Metabotropic Glutamate/metabolism , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship , Transfection , Tricarboxylic Acids/chemistry , Tricarboxylic Acids/pharmacology
15.
Neuroscience ; 92(1): 1-6, 1999.
Article in English | MEDLINE | ID: mdl-10392825

ABSTRACT

Recent reports have outlined that cerebellar long-term depression requires the activation of subtype 1 metabotropic glutamate receptors, since long-term depression is impaired in subtype 1 metabotropic glutamate receptor (mGluR1) knockout mice. In order to better define the role of mGluR1-activated signal transduction pathways, we attempted to rescue cerebellar long-term depression in mGluR1 knockout mice by direct activation of subsequent intracellular cascades. The present results demonstrate that the inositol-1,4,5-trisphosphate signal transduction pathway remains functional in mGluR1 knockout mice, that calcium release from internal stores evoked by the combined photolytic release of inositol- 1,4,5-trisphosphate/pairing protocol is sufficient to rescue long-term depression in these mutants, and that this long-term depression is sensitive to a protein kinase C inhibitor. Therefore, our results provide compelling evidence that the impairment of long-term depression observed in mGluR1 knockout mice is not a consequence of developmental abnormalities, but is directly due to mGluR1 gene inactivation.


Subject(s)
Cerebellum/physiology , Inositol 1,4,5-Trisphosphate/physiology , Long-Term Potentiation/physiology , Receptors, Metabotropic Glutamate/deficiency , Animals , Calcium/physiology , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/physiology , Inositol 1,4,5-Trisphosphate/analogs & derivatives , Inositol 1,4,5-Trisphosphate/metabolism , Long-Term Potentiation/drug effects , Mice , Mice, Knockout/genetics , Photolysis , Protein Kinase C/antagonists & inhibitors , Purkinje Cells/metabolism , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/physiology
16.
Br J Pharmacol ; 105(4): 973-9, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1324059

ABSTRACT

1. The aim of the present study was to examine the effect of 5-hydroxytryptamine (5-HT) on K+ current in primary culture of mouse colliculi neurones and to identify the 5-HT receptor subtype that could be involved in this effect. 2. The voltage-activated K+ current of the neurones was partially blocked by 8-bromo adenosine 3':5'-cyclic monophosphate (8-bromo-cyclic AMP). This effect was mimicked by 5-HT and the action of 5-HT could be antagonized by H7, a non specific protein kinase inhibitor, and by PKI, the specific cyclic AMP-dependent protein kinase blocker. 3. A similar cyclic AMP-dependent blockade of the K+ current was found with renzapride (BRL 24,924) and other 5-HT4 receptor agonists such as cisapride, BIMU 8, zacopride and 5-methoxytryptamine (5-MeOT). ICS 205,930, the classical 5-HT4 receptor blocker, could not be used in this study because it inhibited the studied K+ current by itself. However, the novel 5-HT4 receptor antagonist, DAU 6285 blocked the effects of 5-HT and renzapride on the K+ current. 4. The current was insensitive to the 5-HT1 and 5-HT3 receptor agonists (8-hydroxy-2-(di-n-propylamino) tetralin, RU 24,969, carboxamidotryptamine, 2-CH3-5-HT) as well as to 5-HT1, 5-HT2 and 5-HT3 antagonists (methiothepin, ketanserin, ondansetron [GR 38,032]). Moreover, these antagonists did not affect the actions of the tested 5-HT4 receptor agonists. 5. The present results show that part of the voltage-activated K+ current in mouse colliculi neurones is cyclic AMP-sensitive and the blockade of the current by 5-HT involves the 5-HT4 receptor subtype.The putative implication of 5-HT4 receptors in neuronal plasticity, via a blockade of K+ channels, is discussed.


Subject(s)
Potassium/metabolism , Protein Kinases/metabolism , Receptors, Serotonin/metabolism , Superior Colliculi/metabolism , Animals , Cells, Cultured , Cyclic AMP/pharmacology , Enzyme Activation/drug effects , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Receptors, Serotonin/classification , Receptors, Serotonin/drug effects , Serotonin/pharmacology , Superior Colliculi/drug effects
17.
J Chem Neuroanat ; 10(3-4): 231-40, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8811428

ABSTRACT

Nitric oxide is an endogenous molecule that plays a role of second messenger in the central and peripheral nervous system. A major action of this molecule is to control ionic channel activity. Because of technical difficulties to use nitric oxide as a gaseous compound, nitric oxide donors are often utilized under controlled experimental conditions. Here we will review the advantages and limitations in using these compounds. Nitric oxide can affect ionic channels through direct interactions or through the production of cGMP. We will describe an example of direct action of nitric oxide on glutamate-gated channels. We will also review indirect actions of nitric oxide on various potassium and calcium channels. Finally, we will discuss the complex physiological consequences of the action of nitric oxide on these ionic channels.


Subject(s)
Ion Channel Gating/physiology , Ion Channels/physiology , Nitric Oxide/physiology , Receptors, Glutamate/physiology , Animals , Male , Rats
18.
Neurochem Int ; 6(3): 325-32, 1984.
Article in English | MEDLINE | ID: mdl-20488052

ABSTRACT

The release of l-[(3)H]cysteine sulfinic acid, l-[(3)H]glutamatic acid and [(3)H]GABA from preloaded slices of various rat brain regions in response to either 30 mM K(+) or veratrin was investigated. All these aminoacids were released by both depolarizing agents, which did not produce any changes in the spontaneous efflux of [(3)H]lysine. The K(+) stimulated cysteine sulfinate release from superfused slices was found partly Ca(2+)-dependent in the subiculum, and mainly Ca(2+)-independent in the hippocampus whereas the K(+)-elicited glutamate release was partly Ca(2+)-dependent in both regions. The veratrine-induced release of both cysteine sulfinate and glutamate was blocked by verapamil in a dose-dependent way, although a small verapamil concentration independent release remained. The release pattern of both amino acids was heterogeneous, but roughly correlated among brain regions, except in the subiculum and hypothalamus. These findings demonstrate the releasability of both substances from various brain regions and suggest that those releases occur from different pools, being probably mainly of neuronal origin. They give further evidence that cysteine sulfinate as well as glutamate may serve a neurotransmitter role in the CNS.

19.
Neuroreport ; 6(7): 1037-40, 1995 May 09.
Article in English | MEDLINE | ID: mdl-7632890

ABSTRACT

We investigated the mode of action of argiotoxin636 on isolated N-methyl-D-aspartate (NMDA) receptor channels in cultured cerebellar granule cells. We found that the toxin blocks NMDA channels by decreasing their opening probability and by inducing a flickering activity, in a voltage-dependent manner. Our results indicate that argiotoxin636 acts as an open-channel blocker and might therefore be a useful tool for studying the structure of glutamate-gated channels.


Subject(s)
Cerebellum/metabolism , Phenylacetates/pharmacology , Polyamines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spider Venoms/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Indoleacetic Acids , Ion Channel Gating/drug effects , Kinetics , Mice , Patch-Clamp Techniques
20.
Neuroreport ; 3(1): 77-80, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1535233

ABSTRACT

The effects of a nitric oxide (NO) donor, sodium nitroprusside (SNP), on N-methyl-D-aspartate (NMDA) receptors were assessed by optical measurements of intracellular calcium concentration ([Ca2+]i) and patch-clamp techniques in cultured central neurons. SNP selectively blocked NMDA-mediated currents and increases in [Ca2+]i. SNP inhibited the binding of [3H]-CGS 19755. The blockade of NMDA responses by SNP was prevented by CPP or APV which are selective competitive NMDA receptor antagonists. These effects were not necessarily mediated by NO, since they were mimicked by ferrocyanide ions, the NO companion photolysis product of SNP.


Subject(s)
Ferrocyanides/metabolism , Nitroprusside/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Calcium/metabolism , Female , Mice , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Pipecolic Acids/pharmacology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL