1
|
|
2
|
Lipina T, Weiss K, Roder J. The ampakine CX546 restores the prepulse inhibition and latent inhibition deficits in mGluR5-deficient mice. Neuropsychopharmacology 2007; 32:745-56. [PMID: 16936708 DOI: 10.1038/sj.npp.1301191] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In order to test the possible role of mGluR5 signaling in the behavioral endophenotypes of schizophrenia and other psychiatric disorders, we used genetic engineering to create mice carrying null mutations in this gene. Compared to their mGluR5(+/+) littermates, mGluR5(-/-) mice have disrupted latent inhibition (LI) as measured in a thirst-motivated conditioned emotional response procedure. Administration of the positive modulator of alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPAR), CX546, during the conditioning phase only, improved the disrupted LI in mGluR5 knockout mice and facilitated LI in control C57BL/6J mice, given extended number of conditioning trails (four conditioning stimulus-unconditioned stimulus). Prepulse inhibition (PPI) was impaired in mGluR5(-/-) mice to a level that could not be disrupted further by the antagonist of N-methyl-D-aspartate receptors - MK-801. PPI deficit of mGluR5(-/-) mice was effectively reversed by CX546, whereas aniracetam had a less pronounced effect. These data provide evidence that a potent positive AMPAR modulator can elicit antipsychotic action and represents a new approach for treatment of schizophrenia.
Collapse
MESH Headings
- Acoustic Stimulation/methods
- Analysis of Variance
- Animals
- Antidepressive Agents/pharmacology
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Conditioning, Classical/drug effects
- Dioxoles/pharmacology
- Dizocilpine Maleate/pharmacology
- Dose-Response Relationship, Drug
- Dose-Response Relationship, Radiation
- Drug Interactions
- Electroshock/adverse effects
- Excitatory Amino Acid Antagonists/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neural Inhibition/drug effects
- Neural Inhibition/genetics
- Piperidines/pharmacology
- Pyrrolidinones/pharmacology
- Receptor, Metabotropic Glutamate 5
- Receptors, Metabotropic Glutamate/deficiency
- Reflex, Startle/drug effects
- Reflex, Startle/genetics
Collapse
Affiliation(s)
- Tatiana Lipina
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
| | | | | |
Collapse
|
3
|
Gabryel B, Pudelko A, Malecki A. Erk1/2 and Akt kinases are involved in the protective effect of aniracetam in astrocytes subjected to simulated ischemia in vitro. Eur J Pharmacol 2005; 494:111-20. [PMID: 15212964 DOI: 10.1016/j.ejphar.2004.04.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2004] [Revised: 04/07/2004] [Accepted: 04/26/2004] [Indexed: 10/26/2022]
Abstract
The present study focused on the mechanism of cytoprotective effect of aniracetam on the primary rat astrocyte cultures exposed to simulated ischemia conditions in vitro. To study these mechanisms, the aniracetam-mediated modulation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3-K)/Akt kinase pathways was determined. Simulated in vitro ischemia caused death of approximately 35% of astrocytes via apoptosis and decreased cell viability about 50% at 8 h. Exposure to aniracetam at concentrations of 0.1-10 microM in these conditions significantly decreased the number of apoptotic cells. Moreover, the intensification of 3-(4,5-dimethylthazol-2-yl)-2,5-diphenyltetrazolinum bromide (MTT) conversion and the decrease of lactate dehydrogenase (LDH) release after 1 and 10 microM aniracetam treatment were observed indicating a significant increase in cell viability. When cultured astrocytes were incubated during 8 h simulated ischemia with [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene] (U0126), an extracellular regulated kinase 1 and 2 (Erk1/2) inhibitor or wortmannin, a phosphatidylinositol 3-kinase (PI3 kinase)/Akt inhibitor, the cell apoptosis was accelerated. These effects of used kinase inhibitors (both U0126 and wortmannin) were antagonized by adding 1 and 10 microM aniracetam to the culture medium. In addition, aniracetam significantly stimulated of phospho-Erk1/2 kinase and phospho-Akt expression. Maximum levels of Erk1/2 and Akt activation were observed as a result of treatment with 10 microM aniracetam. U0126 and wortmannin markedly attenuated the effects of aniracetam on expression of activated kinases. Results of the present study indicate that both Erk1/2 and PI 3-K/Akt kinase pathways are vital for cytoprotective effect of aniracetam.
Collapse
Affiliation(s)
- Bozena Gabryel
- Department of Pharmacology, Silesian Medical University, 18 Medyków Street, PL 40752 Katowice, Poland.
| | | | | |
Collapse
|
4
|
Smith AM, Wehner JM. Aniracetam improves contextual fear conditioning and increases hippocampal gamma-PKC activation in DBA/2J mice. Hippocampus 2002; 12:76-85. [PMID: 11918291 DOI: 10.1002/hipo.10008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
DBA/2J (D2) mice display poor contextual learning and have less membrane-bound hippocampal protein kinase C (PKC) compared with C57BL/6 (B6) mice. Aniracetam and oxiracetam were previously shown to improve contextual learning in D2 mice and increase PKC activity. This study investigated a possible mechanism for learning enhancement by examining the effects of aniracetam on contextual fear conditioning and activation of the y isoform of PKC (gamma-PKC) in male D2 mice. In comparison to animals treated with vehicle only (10% 2-hydroxypropyl-beta-cyclodextrin), mice treated with aniracetam (100 mg/kg) 30 min prior to fear conditioning training demonstrated significantly improved contextual learning when tested 30 min and 24 h after training. This corresponded with a significant increase in activated, membrane-bound hippocampal gamma-PKC 30 min after training. No increase in learning or gamma-PKC was found 5 min after training. These results suggest an altered time course of activation of gamma-PKC in response to treatment with aniracetam, which improves learning in D2 mice.
Collapse
Affiliation(s)
- Amy M Smith
- Institute for Behavioral Genetics and Department of Psychology, University of Colorado, Boulder 80309, USA
| | | |
Collapse
|
5
|
Gabryel B, Adamczyk J, Huzarska M, Pudełko A, Trzeciak HI. Aniracetam attenuates apoptosis of astrocytes subjected to simulated ischemia in vitro. Neurotoxicology 2002; 23:385-95. [PMID: 12387365 DOI: 10.1016/s0161-813x(02)00084-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The aim of the present study was to establish whether aniracetam is capable of protecting cultured rat astrocytes against ischemic injury. Treatment of the cultures with aniracetam (1, 10 and 100 mM) during 24 h ischemia simulated in vitro significantly decreased the number of apoptotic cells. The antiapoptotic effects of the drug were confirmed by the increase of intracellular ATP and phosphocreatine (PCr) levels and the inhibition of the caspase-3 activity. Aniracetam also attenuated cellular oxidative stress by decreased production of reactive oxygen species (ROS). These effects were associated with the decrease in levels of c-fos and c-jun mRNA in primary astrocyte cultures exposed to 24 h ischemia. When cultured astrocytes were incubated during 24 h simulated ischemia with wortmannin, a phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor or PD98059, a mitogen-activated protein (MAP)/extracellular signal regulated kinase (ERK) (MEK) inhibitor the cell apoptosis was accelerated. This effect was antagonized by adding 100 mM aniracetam to the culture medium. These findings suggest that the protective effect of aniracetam is mediated by PI 3-kinase and MEK pathways in the downstream mechanisms.
Collapse
Affiliation(s)
- Bozena Gabryel
- Department of Pharmacology, Silesian Medical University, Katowice, Poland.
| | | | | | | | | |
Collapse
|
6
|
Sekiguchi M, Yamada K, Jin J, Hachitanda M, Murata Y, Namura S, Kamichi S, Kimura I, Wada K. The AMPA receptor allosteric potentiator PEPA ameliorates post-ischemic memory impairment. Neuroreport 2001; 12:2947-50. [PMID: 11588608 DOI: 10.1097/00001756-200109170-00038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PEPA (4-[2-(Phenylsulphonylamino)ethylthio]-2,6-difluorophenoxyacetamide) is a recently developed allosteric potentiator of AMPA receptors that preferentially affects flop splice variants. We tested the effects of PEPA on ischemia-induced memory deficit in rats. Permanent unilateral occlusion of the middle cerebral artery induced severe impairment of performance of rats in the Morris water maze test. Repeated intravenous administration of PEPA (1, 3, 10 mg/kg/day for 10 days) improved test performance. In contrast, a corresponding dose of aniracetam, a representative potentiator of AMPA receptor, did not significantly improve test performance. Thus, PEPA is more effective than aniracetam in reversing impaired memory function as assessed by the Morris water maze test; and PEPA may be an effective compound for the treatment of impaired memory.
Collapse
Affiliation(s)
- M Sekiguchi
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Arai AC, Kessler M, Rogers G, Lynch G. Effects of the potent ampakine CX614 on hippocampal and recombinant AMPA receptors: interactions with cyclothiazide and GYKI 52466. Mol Pharmacol 2000; 58:802-13. [PMID: 10999951 DOI: 10.1124/mol.58.4.802] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
R,S-alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor up-modulators of the benzamide type ("ampakines") have previously been shown to enhance excitatory synaptic transmission in vivo and in vitro and AMPA receptor currents in excised patches. The present study analyzed the effects of an ampakine (CX614; 2H,3H, 6aH-pyrrolidino[2",1"-3',2']1,3-oxazino[6',5'-5,4]benz o[e]1, 4-dioxan-10-one) that belongs to a benzoxazine subgroup characterized by greater structural rigidity and higher potency. CX614 enhanced the size (amplitude and duration) of field excitatory postsynaptic potentials in hippocampal slices and autaptically evoked excitatory postsynaptic currents in neuronal cultures with EC(50) values of 20 to 40 microM. The compound blocked desensitization (EC(50) = 44 microM) and slowed deactivation of responses to glutamate by a factor of 8.4 in excised patches. Currents through homomeric, recombinant AMPA receptors were enhanced with EC(50) values that did not differ greatly across GluR1-3 flop subunits (19-37 microM) but revealed slightly lower potency at corresponding flip variants. Competition experiments using modulation of [(3)H]fluorowillardiine binding suggested that CX614 and cyclothiazide share a common binding site but cyclothiazide seems to bind to an additional site not recognized by the ampakine. CX614 did not reverse the effect of GYKI 52466 on responses to brief glutamate pulses, which indicates that they act through separate sites, a conclusion that was confirmed in binding experiments. In sum, these results extend prior evidence that ampakines are effective in enhancing synaptic responses, most likely by slowing deactivation, and that their effects are exerted through sites that are only in part shared with other modulators.
Collapse
Affiliation(s)
- A C Arai
- Department of Pharmacology, Southern Illinois University, Springfield, Illinois, 62794-9629, USA
| | | | | | | |
Collapse
|
8
|
Activation of synaptic receptors and its allosteric regulation. NEUROPHYSIOLOGY+ 2000. [DOI: 10.1007/bf02515168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
9
|
Niittykoski M, Ruotsalainen S, Haapalinna A, Larson J, Sirviö J. Activation of muscarinic M3-like receptors and beta-adrenoceptors, but not M2-like muscarinic receptors or alpha-adrenoceptors, directly modulates corticostriatal neurotransmission in vitro. Neuroscience 1999; 90:95-105. [PMID: 10188937 DOI: 10.1016/s0306-4522(98)00447-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aim of this study was to characterize the modulation of synaptic transmission in the glutamatergic corticostriatal pathway by cholinergic and adrenergic receptors. In coronal slices of mouse brain, negative-going field potentials were recorded in the dorsal striatum in response to stimulation of the overlying white matter, and their susceptibility to various pharmacological manipulations was studied. The responses were mediated by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors, since they were augmented by aniracetam (0.5-1.5 mM), a positive modulator of AMPA-type glutamate receptors, and blocked by 6-cyano-7-nitroquinoxaline-2,3-dione (> or = 10 microM), a selective antagonist of AMPA receptors. Carbachol (10 microM), a muscarinic agonist, reduced the size of responses and abolished paired-pulse depression; these effects being consistent with previous studies indicating that muscarinic activation inhibits release of glutamate in the corticostriatal pathway. Muscarinic antagonists could block the effect of carbachol. Their rank order was: 10 microM scopolamine (a non-selective muscarinic antagonist) > or = 1 microM 4-diphenylacetoxy-N-methyl-piperidine (M3/M1 antagonist)>1 microM pirenzepine (M1 antagonist)>10 microM methoctramine (M2 antagonist). McN-A-343 (1-10 microM), an M1 muscarinic agonist, was ineffective in this preparation. In contrast, isoproterenol (10-30 microM), a beta-adrenergic agonist, slightly increased the synaptic responses, but it did not affect paired-pulse depression. None of alpha-adrenergic agents (30 nM-1.0 microM dexmedetomidine, an alpha2-adrenergic agonist, 0.3 microM atipamezole, an alpha2-adrenergic antagonist or 30 microM phenylephrine, an alpha1-adrenergic agonist) influenced the size of the responses; neither did these drugs alter paired-pulse depression. These results indicate that the activation of striatal M3-like muscarinic receptors and beta-adrenoceptors, but not M2-like muscarinic receptors and alpha-adrenoceptors, modulates directly corticostriatal glutamatergic neurotransmission.
Collapse
MESH Headings
- (4-(m-Chlorophenylcarbamoyloxy)-2-butynyl)trimethylammonium Chloride/pharmacology
- 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology
- Adrenergic Fibers/drug effects
- Adrenergic Fibers/physiology
- Adrenergic alpha-Agonists/pharmacology
- Adrenergic alpha-Antagonists/pharmacology
- Adrenergic beta-Agonists/pharmacology
- Animals
- Carbachol/pharmacology
- Cerebral Cortex/drug effects
- Cerebral Cortex/physiology
- Cholinergic Fibers/drug effects
- Cholinergic Fibers/physiology
- Corpus Striatum/drug effects
- Corpus Striatum/physiology
- Diamines/pharmacology
- Excitatory Amino Acid Agonists/pharmacology
- Excitatory Postsynaptic Potentials/drug effects
- Glutamic Acid/metabolism
- Imidazoles/pharmacology
- Isoproterenol/pharmacology
- Male
- Medetomidine
- Mice
- Mice, Inbred DBA
- Muscarinic Agonists/pharmacology
- Muscarinic Antagonists/pharmacology
- Phenylephrine/pharmacology
- Piperidines/pharmacology
- Pirenzepine/pharmacology
- Pyrrolidinones/pharmacology
- Receptor, Muscarinic M2
- Receptor, Muscarinic M3
- Receptors, AMPA/drug effects
- Receptors, AMPA/physiology
- Receptors, Adrenergic, alpha/drug effects
- Receptors, Adrenergic, alpha/physiology
- Receptors, Adrenergic, beta/drug effects
- Receptors, Adrenergic, beta/physiology
- Receptors, Muscarinic/drug effects
- Receptors, Muscarinic/physiology
- Scopolamine/pharmacology
- Synaptic Transmission/drug effects
Collapse
Affiliation(s)
- M Niittykoski
- A. I. Virtanen Institute, University of Kuopio, Finland
| | | | | | | | | |
Collapse
|
10
|
Shen Y, Lu T, Yang XL. Modulation of desensitization at glutamate receptors in isolated crucian carp horizontal cells by concanavalin A, cyclothiazide, aniracetam and PEPA. Neuroscience 1999; 89:979-90. [PMID: 10199629 DOI: 10.1016/s0306-4522(98)00310-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In horizontal cells freshly dissociated from crucian carp (Carassius auratus) retina, we examined the effects of modulators of glutamate receptor desensitization, concanavalin A, cyclothiazide, aniracetam and 4-[2-(phenylsulfonylamino)ethylthio]-2,6-difluoro-phenoxyacetam ide (PEPA), on responses to rapid application of glutamate and kainate, using whole-cell voltage-clamp techniques. Incubation of concanavalin A suppressed the peak response but weakly potentiated the equilibrium response of horizontal cells to glutamate. Cyclothiazide blocked glutamate-induced desensitization in a dose-dependent manner, which resulted in a steady increase of the equilibrium current. The concentration of cyclothiazide causing a half-maximal potentiation for the equilibrium response was 85 microM. Furthermore, cyclothiazide shifted the dose-response relationship of the equilibrium current to the right, but slightly suppressed the kainate-induced sustained current. These effects of concanavalin A and cyclothiazide are consistent with the supposition that glutamate receptors of carp horizontal cells may be an alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)-preferring subtype. In order to further characterize the AMPA receptors of horizontal cells, modulation by aniracetam and PEPA of glutamate- and kainate-induced currents was studied. Aniracetam, a preferential modulator of flop variants of AMPA receptors, considerably blocked desensitization of glutamate-induced currents, but only slightly potentiated kainate-induced currents. It was further found that PEPA, a flop-preferring allosteric modulator of AMPA receptor desensitization, slightly suppressed the peak current, while it dramatically potentiated the equilibrium current induced by glutamate in a dose-dependent manner. PEPA was much potent than aniracetam at these receptors and showed the effect on glutamate-induced desensitization even at a concentration as low as 3 microM. PEPA also potentiated non-desensitizing currents induced by kainate, but with much less extent. These modulatory effects of concanavalin A, cyclothiazide, aniracetam and PEPA on AMPA receptors in carp horizontal cells were rather similar to those obtained at AMPA receptors assembled from flop variants expressed in Xenopus oocyte and HEK cell. Consequently, we speculate that the AMPA receptor on carp horizontal cells may predominantly carry the flop splice variants.
Collapse
Affiliation(s)
- Y Shen
- Shanghai Institute of Physiology and Key Laboratory of Neurobiology, Chinese Academy of Sciences
| | | | | |
Collapse
|
11
|
Affiliation(s)
- D Bleakman
- Lilly Research Laboratories, Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | |
Collapse
|
12
|
Kolta A, Lynch G, Ambros-Ingerson J. Effects of aniracetam after LTP induction are suggestive of interactions on the kinetics of the AMPA receptor channel. Brain Res 1998; 788:269-86. [PMID: 9555053 DOI: 10.1016/s0006-8993(97)01444-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The modulatory influence of aniracetam, a drug which reversibly modifies the kinetic properties of AMPA-type glutamate receptors, on synaptic responses is reported to be detectably changed by the induction of long-term potentiation (LTP). The present study used hippocampal slices to examine three issues arising from this result. First, possible contributions of inhibitory currents and postsynaptic spiking to the aniracetam/LTP interaction were investigated with infusions of GABA receptor antagonists and topical applications of tetrodotoxin. Second, tests were carried out to determine if the altered response to aniracetam is sufficiently persistent to be a plausible substrate for the extremely stable LTP effect. Third, the nature of the change responsible for the aniracetam/LTP interaction was explored with waveform analyses and a kinetic model of the AMPA receptor. The following results were obtained. LTP reduced the effect of aniracetam on the amplitude but increased its effect on the decay time constant of field EPSPs recorded under conditions in which local spiking and inhibitory responses were blocked. The LTP-induced change in the effect of aniracetam was extremely stable in that it was still evident 75 min after induction of potentiation. Finally, the waveform distortions introduced by LTP and aniracetam could be corrected by uniform stretching of the responses, suggesting that the changes introduced by each of the manipulations are unitary in nature. These distortions and the interactions between them could be reproduced in the AMPA receptor model by representing LTP as an acceleration of channel gating kinetics.
Collapse
Affiliation(s)
- A Kolta
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA
| | | | | |
Collapse
|
13
|
Yamamoto C, Sawada S, Ohno-Shosaku T. Distribution and properties of kainate receptors distinct in the CA3 region of the hippocampus of the guinea pig. Brain Res 1998; 783:227-35. [PMID: 9507146 DOI: 10.1016/s0006-8993(97)01350-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To characterize the nature of kainate (KA) receptors distinct in the CA3 region of the hippocampus, properties of depolarizations induced by pulses of KA or AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate) applied to dendrites of CA3 neurons with micropipettes were studied in thin transverse slices of the guinea pig hippocampus. KA induced depolarizations at negligible latencies only when administered to the most proximal dendritic areas. The depolarization was unaffected by tetrodotoxin or by a decrease in Ca2+ and an increase in Mg2+ concentrations. The declining slope of the KA-induced depolarization was significantly slower than that of the AMPA-induced depolarization. In comparison with the AMPA-induced depolarization, the KA-induced depolarization was much less susceptible to antagonists such as 6-cyano-7-nitroquinoxaline-2, 3-dione (CNQX) and 1-(4-aminophenyl)-4-methyl-7, 8-methylenedioxy-5H-2,3-benzodiazepine hydrochloride (GYKI52466). 6, 7,8,9-Tetrahydro-5-nitro-1H-benz[g]indole-2,3-dione-3-oxime (NS-102) and (2S,4R)-4-methylglutamate (SYM 2081) were without effects. The threshold concentration of pressure-ejected KA to induce depolarizations was about 200 nM. Excitatory postsynaptic potentials elicited by mossy fiber stimulation were more potently suppressed by CNQX than by GYKI52466. These results indicate that receptors responsible for the slow KA depolarization in the CA3 region of the hippocampus are not AMPA receptors but KA receptors. They are localized in the most proximal part of the apical dendrite and distinct from those observed in primary cultures of hippocampal neurons.
Collapse
Affiliation(s)
- C Yamamoto
- Department of Physiology, Faculty of Medicine, Kanazawa University, Kanazawa 920, Japan
| | | | | |
Collapse
|
14
|
A novel allosteric potentiator of AMPA receptors: 4--2-(phenylsulfonylamino)ethylthio--2,6-difluoro-phenoxyaceta mide. J Neurosci 1997. [PMID: 9221774 DOI: 10.1523/jneurosci.17-15-05760.1997] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We report that a novel sulfonylamino compound, 4-[2-(phenylsulfonylamino)ethylthio]-2,6-difluoro-phenoxyacetam ide (PEPA), selectively potentiates glutamate receptors of the AMPA subtype. PEPA (1-200 microM) dose dependently potentiated glutamate-evoked currents in Xenopus oocytes expressing AMPA (GluRA-GluRD), but not kainate (GluR6 and GluR6+KA2) or NMDA (zeta1 + epsilon1-epsilon4), receptor subunits. PEPA was effective at micromolar concentrations and, in contrast to the action of cyclothiazide, preferentially modulated AMPA receptor flop isoforms. At 200 microM, PEPA potentiated glutamate responses by 50-fold in oocytes expressing GluRCflop (EC50 approximately 50 microM) versus only threefold for GluRCflip; a similar preference for flop isoforms was observed for other AMPA receptor subunits. Dose-response analysis for GluRCflop revealed that 100 microM PEPA produced a sevenfold increase in AMPA receptor affinity for glutamate. PEPA produced considerably weaker potentiation of kainate-evoked than glutamate-evoked currents, suggesting modulation of the process of receptor desensitization. In human embryonic kidney 293 cells transfected with AMPA receptor subunits, PEPA either abolished or markedly slowed the rate of onset of desensitization and potentiated steady-state equilibrium currents evoked by glutamate with subunit (GluRC >/= GluRD > GluRA) and splice-variant (flop > flip) selectivity similar to that observed in oocytes. Our results show that PEPA is a novel, flop-preferring allosteric modulator of AMPA receptor desensitization at least 100 times more potent than aniracetam.
Collapse
|
15
|
Matton A, Engelborghs S, Bollengier F, Finné E, Vanhaeist L. Modulating effect of the nootropic drug, piracetam on stress- and subsequent morphine-induced prolactin secretion in male rats. Br J Pharmacol 1996; 117:502-506. [PMID: 8821540 PMCID: PMC1909305 DOI: 10.1111/j.1476-5381.1996.tb15218.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
1. The effect of the nootropic drug, piracetam on stress- and subsequent morphine-induced prolactin (PRL) secretion was investigated in vivo in male rats, by use of a stress-free blood sampling and drug administration method by means of a permanent indwelling catheter in the right jugular vein. 2. Four doses of piracetam were tested (20, 100, 200 and 400 mg kg-1), being given intraperitoneally 1 h before blood sampling; control rats received saline instead. After a first blood sample, rats were subjected to immobilization stress and received morphine, 6 mg kg-1, 90 min later. 3. Piracetam had no effect on basal plasma PRL concentration. 4. While in the non-piracetam-treated rats, stress produced a significant rise in plasma PRL concentration, in the piracetam-pretreated rats PRL peaks were attenuated, especially in the group given 100 mg kg-1 piracetam, where plasma PRL concentration was not significantly different from basal values. The dose-response relationship showed a U-shaped curve; the smallest dose had a minor inhibitory effect and the highest dose had no further effect on the PRL rise. 5. In unrestrained rats, morphine led to a significant elevation of plasma PRL concentration. After the application of immobilization stress it lost its ability to raise plasma PRL concentration in the control rats, but not in the piracetam-treated rats. This tolerance was overcome by piracetam in a significant manner but with a reversed dose-response curve; i.e. the smaller the dose of piracetam, the higher the subsequent morphine-induced PRL peak. 6. There is no simple explanation for the mechanism by which piracetam induces these contradictory effects. Interference with the excitatory amino acid system, which is also involved in opiate action, is proposed speculatively as a possible mediator of the effects of piracetam.
Collapse
Affiliation(s)
- A Matton
- Dept. of Pharmacology, Faculty of Medicine, Vrije Universiteit Brussel, Belgium
| | | | | | | | | |
Collapse
|
16
|
Fletcher EJ, Lodge D. New developments in the molecular pharmacology of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate and kainate receptors. Pharmacol Ther 1996; 70:65-89. [PMID: 8804111 DOI: 10.1016/0163-7258(96)00014-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Separation of non-N-methyl-D-aspartate subtypes of glutamate receptors, known as alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) and kainate receptors, is traced through conventional pharmacology to molecular biology. The physiology and pharmacology of recombinant receptor subtypes (GluR1-7 and KA1-2) are described. Competitive antagonists, e.g., the quinoxalinedione, 2,3-dihyroxy-6-nitro-7-sulphamoyl-benz(F)quinoxaline, and the decahydroisoquinoline, 3S,4aR,6R, 8aR-6-[2-(1(2)H-tetrazol-5-yl)ethyl]-decahydroisoquinolin e-3-carboxylate, have a broad antagonist spectrum, except that the latter is inactive on GluR6. The 2,3-benzodiazepines noncompetitively antagonise the AMPA receptor GluR1-4. Desensitisation of AMPA (GluR1-4) and kainate (GluR5-7 and KA1-2) receptors is blocked by cyclothiazide and concanavalin A, respectively. Polyamine toxins block AMPA receptors not containing GluR2 and unedited kainate receptors (GluR5-6). These data correlate well with results on native neurons characterised by techniques such as in situ hybridisation.
Collapse
Affiliation(s)
- E J Fletcher
- MRC Laboratory of Molecular Biology/Department of Zoology, Cambridge, UK
| | | |
Collapse
|
17
|
Krogsgaard-Larsen P, Ebert B, Lund TM, Bräuner-Osborne H, Sløk FA, Johansen TN, Brehm L, Madsen U. Design of excitatory amino acid receptor agonists, partial agonists and antagonists: ibotenic acid as a key lead structure. Eur J Med Chem 1996. [DOI: 10.1016/0223-5234(96)89549-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
18
|
Sharp RL, Mayne NG, Burnett JP. Cyclothiazide differentially modulates human metabotropic glutamate receptors linked to phosphoinositide hydrolysis stimulation in oocytes. Eur J Pharmacol 1994; 269:R5-7. [PMID: 7895767 DOI: 10.1016/0922-4106(94)90049-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cloned human metabotropic glutamate receptors, mGlu1 alpha and mGlu5 alpha, were functionally expressed in Xenopus oocytes. Cyclothiazide dose-dependently inhibited glutamate-stimulated human mGlu1 alpha responses (IC50 = 18 microM) in a non-competitive manner. In contrast, cyclothiazide slightly potentiated glutamate-stimulated human mGlu5 alpha responses. GYKI 52466 (1-(4-amino-phenyl)-4-methyl-7,8- methyl-endioxyl-5H-2,3-benzodiazepinehydrochloride) did not alter glutamate-stimulated human mGlu1 alpha or human mGlu5 alpha responses, either in the presence or absence of cyclothiazide. Thus, human metabotropic glutamate receptors coupled to phosphoinositide stimulation appear to contain sites sensitive to cyclothiazide but insensitive to GYKI 52466.
Collapse
Affiliation(s)
- R L Sharp
- Lilly Research Laboratories 0424, Eli Lily and Co., Lilly Corporate Center Indianapolis, IN 46285
| | | | | |
Collapse
|
19
|
Gouliaev AH, Senning A. Piracetam and other structurally related nootropics. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1994; 19:180-222. [PMID: 8061686 DOI: 10.1016/0165-0173(94)90011-6] [Citation(s) in RCA: 247] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Nearly three decades have now passed since the discovery of the piracetam-like nootropics, compounds which exhibit cognition-enhancing properties, but for which no commonly accepted mechanism of action has been established. This review covers clinical, pharmacokinetic, biochemical and behavioural results presented in the literature from 1965 through 1992 (407 references) of piracetam, oxiracetam, pramiracetam, etiracetam, nefiracetam, aniracetam and rolziracetam and their structural analogues. The piracetam-like nootropics are capable of achieving reversal of amnesia induced by, e.g., scopolamine, electroconvulsive shock and hypoxia. Protection against barbiturate intoxication is observed and some benefit in clinical studies with patients suffering from mild to moderate degrees of dementia has been demonstrated. No affinity for the alpha 1-, alpha 2-, beta-, muscarinic, 5-hydroxytryptamine-, dopamine, adenosine-A1-, mu-opiate, gamma-aminobutyric acid (GABA) (except for nefiracetam (GABAA)), benzodiazepine and glutamate receptors has been found. The racetams possess a very low toxicity and lack serious side effects. Increased turnover of different neurotransmitters has been observed as well as other biochemical findings, e.g., inhibition of enzymes such as prolylendopeptidase. So far, no generally accepted mechanism of action has, however, emerged. We believe that the effect of the racetams is due to a potentiation of already present neurotransmission and that much evidence points in the direction of a modulated ion flux by, e.g., potentiated calcium influx through non-L-type voltage-dependent calcium channels, potentiated sodium influx through alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor gated channels or voltage-dependent channels or decreases in potassium efflux. Effects on carrier mediated ion transport are also possible.
Collapse
Affiliation(s)
- A H Gouliaev
- Department of Chemistry, Aarhus University, Denmark
| | | |
Collapse
|
20
|
Chapter 6. Recent Progress in Excitatory Amino Acid Research. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 1994. [DOI: 10.1016/s0065-7743(08)60719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
21
|
Sharp RL, May PC, Mayne NG, Snyder YM, Burnett JP. Cyclothiazide potentiates agonist responses at human AMPA/kainate receptors expressed in oocytes. Eur J Pharmacol 1994; 266:R1-2. [PMID: 7511109 DOI: 10.1016/0922-4106(94)90216-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cloned human AMPA/kainate subunits were functionally expressed in Xenopus oocytes. Cyclothiazide potentiated kainate-evoked currents by 682 +/- 122% (mean +/- S.E.M., n = 5), 1396 +/- 55% (n = 4), and 690 +/- 40% (n = 14) in oocytes expressing GluR1, GluR2, and GluR1 + GluR2 receptors, respectively. AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionate)-induced currents were also potentiated by cyclothiazide. GYKI 52466 (1-(4-amino-phenyl)-4-methyl-7,8-methylendioxyl-5H-2,3-benzod++ + iazepine hydrochloride) attenuated cyclothiazide potentiation in all cases. Thus, modulatory sites for cyclothiazide and GYKI 52466 exist on individual human AMPA/kainate receptor subunits. Additionally, kainate appears to act as a desensitizing partial agonist at human GluR1 and GluR2 receptor subunits.
Collapse
Affiliation(s)
- R L Sharp
- Lilly Research Laboratories, A Division of Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285
| | | | | | | | | |
Collapse
|