Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 233
Filter
1.
Bioorg Med Chem ; 23(1): 66-72, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25496808

ABSTRACT

Mitochondrial uncoupling is one of the therapeutic strategies used to control energy metabolism in various metabolic diseases and in obesity. Ppc-1 (1), a prenylated quinolinecarboxylic acid isolated from cellular slime molds, shows uncoupling activity in vitro and anti-obesity activity in vivo. In this study, we synthesized Ppc-1 (1) and its derivatives, and revealed the structure-activity relationship of uncoupling activities. The triprenylated compound 18 showed mitochondrial uncoupling activity that was more potent than that of Ppc-1 (1). Compound 18 also suppressed weight gain in mice without undesired effects such as lesions on tissues. These results indicate that compound 18 could be used as a seed compound for new anti-obesity drugs.


Subject(s)
Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacology , Obesity/drug therapy , Quinolinic Acids/chemical synthesis , Quinolinic Acids/pharmacology , Animals , Anti-Obesity Agents/chemistry , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/metabolism , Prenylation , Quinolinic Acids/chemistry , Structure-Activity Relationship , Uncoupling Agents/chemical synthesis , Uncoupling Agents/chemistry , Uncoupling Agents/pharmacology
2.
Biochem Biophys Res Commun ; 404(2): 652-5, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21146500

ABSTRACT

Astrocytic glycogen metabolism sustains neuronal activity but its impact on basal glutamatergic synaptic transmission is not clear. To address this issue, we have compared the effect of glycogen breakdown inhibition on miniature excitatory postsynaptic currents (mEPSCs) in rat hippocampal pure neuronal culture (PNC) and in astrocyte-neuronal co-cultures (ANCC). Amplitudes of mEPSC in ANCC were nearly twice as large as in PNC with no difference in current kinetics. Inhibition of glycogen phosphorylase reduced mEPSC amplitude by roughly 40% in ANCC being ineffective in PNC. Altogether, these data indicate that astrocyte-neuronal interaction enhances basal mEPSCs in ANCC mainly due to astrocytic glycogen metabolism.


Subject(s)
Glutamic Acid/metabolism , Glycogen/metabolism , Hippocampus/physiology , Synaptic Transmission , Animals , Astrocytes/metabolism , Astrocytes/physiology , Coculture Techniques , Glycogen Phosphorylase/antagonists & inhibitors , Glycogen Phosphorylase/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Neurons/metabolism , Neurons/physiology , Quinolinic Acids/pharmacology , Rats , Rats, Wistar
3.
Brain Res ; 1198: 107-14, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18243165

ABSTRACT

Polyamines are aliphatic amines containing nucleophilic centers that are found in all eukaryotic cells, including brain cells. These compounds determine neuroprotection in experimental models of cerebral ischemia and neurotoxicity. In the current study we investigated the protective effects of spermine, an agonist of the polyamine binding site at the N-methyl-d-aspartate receptor, against the behavioral and neurochemical alterations induced by quinolinic acid. The unilateral intrastriatal injection of quinolinic acid (180 nmol/site into the dorsal striatum) induced stereotypical motor asymmetries, assessed by the open field and elevated body swing tests. Spermine modulated quinolinic acid-induced rotational behavior biphasically. While the previous intrastriatal administration of spermine at the dose of 0.1 nmol/site increased, the administration of spermine at the dose of 10 nmol/site reduced quinolinic acid-induced rotational behavior. Spermine (10 nmol/site) also decreased the contralateral swing behavior induced by quinolinic acid. Furthermore, the effect of 10 nmol of spermine was counteracted by the co-administration of arcaine (10 nmol), a selective antagonist of the polyamine binding site at the N-methyl-d-aspartate receptor. In addition, spermine (10 nmol) protected against quinolinic acid-induced protein carbonylation in the rat striatum, further suggesting an antioxidant role for this polyamine. These results provide evidence that the behavioral and biochemical alterations induced by quinolinic acid are attenuated or prevented by spermine through its interaction with N-methyl-d-aspartate receptor and/or its antioxidant function.


Subject(s)
Antioxidants/metabolism , Brain Chemistry/physiology , Corpus Striatum/metabolism , Oxidative Stress/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Spermine/metabolism , Animals , Antioxidants/pharmacology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Biguanides/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain Chemistry/drug effects , Corpus Striatum/drug effects , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Male , Motor Activity/drug effects , Motor Activity/physiology , Oxidative Stress/drug effects , Quinolinic Acids/antagonists & inhibitors , Quinolinic Acids/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Spermine/pharmacology
4.
J Physiol ; 586(1): 211-25, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17962329

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) display differences in their sensitivity to the channel blockers Mg(2+) and memantine that are dependent on the identity of the NR2 subunit present in the receptor-channel complex. This study used two-electrode voltage-clamp recordings from Xenopus laevis oocytes expressing recombinant NMDARs to investigate the actions of Mg(2+) and memantine at the two NMDARs displaying the largest differences in sensitivity to these blockers, namely NR1/NR2A and NR1/NR2D NMDARs. In addition, NR2A/2D chimeric subunits have been employed to examine the effects of pore-forming elements and ligand-binding domains (LBD) on the potency of the block produced by each of these inhibitors. Our results show that, as previously documented, NR2D-containing NMDARs are less sensitive to voltage-dependent Mg(2+) block than their NR2A-containing counterparts. The reduced sensitivity is determined by the M1M2M3 membrane-associated regions, as replacing these regions in NR2A subunits with those found in NR2D subunits results in a approximately 10-fold reduction in Mg(2+) potency. Intriguingly, replacing the NR2A LBD with that from NR2D subunits results in a approximately 2-fold increase in Mg(2+) potency. Moreover, when responses mediated by NR1/NR2A NMDARs are evoked by the partial agonist homoquinolinate, rather than glutamate, Mg(2+) also displays an increased potency. Memantine block of glutamate-evoked currents is most potent at NR1/NR2D NMDARs, but no differences are observed in its ability to inhibit NR2A-containing or NR2A/2D chimeric NMDARs. We suggest that the potency of block of NMDARs by Mg(2+) is influenced not only by pore-forming regions but also the LBD and the resulting conformational changes that occur following agonist binding.


Subject(s)
Dopamine Agents/pharmacology , Magnesium/pharmacology , Memantine/pharmacology , Oocytes/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Chimera , Dose-Response Relationship, Drug , Electrophysiology , Evoked Potentials/drug effects , Female , Glutamic Acid/pharmacology , Oocytes/drug effects , Patch-Clamp Techniques , Quinolinic Acids/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Xenopus laevis
5.
Toxicol Sci ; 100(1): 44-53, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17693424

ABSTRACT

Rabbit muscle glycogen phosphorylase-a (GPa) reduces arsenate (As(V)) to the more toxic arsenite (As(III)) in a glutathione (GSH)-dependent fashion. To determine whether reduction of As(V) by GPa is countered by compounds known to inhibit GP-catalyzed glycogenolysis, the effects of thiol reagents, endogenous compounds (glucose, ATP, ADP) as well as nonspecific glycogen phosphorylase inhibitors (GPIs; caffeine, quercetin, flavopiridol [FP]), and specific GPIs (1,4-dideoxy-1,4-imino-D-arabinitol [DAB], BAY U6751, CP320626) were tested on reduction of As(V) by rabbit muscle GPa in the presence of glycogen (substrate), AMP (activator), and GSH, and the As(III) formed from As(V) was quantified by high-performance liquid chromatography-hydride generation-atomic fluorescence spectrometry. The As(V)-reducing activity of GPa was moderately sensitive to thiol reagents. Glucose above 5mM and ADP or ATP at physiological levels diminished GPa-catalyzed As(V) reduction. All GPIs inhibited As(V) reduction by GPa in a concentration-dependent fashion; however, their effects were differentially affected by glucose (10mM) or AMP (200microM instead of 25microM), known modulators of the action of some GPIs on the GP-catalyzed glycogenolysis. Inhibition of As(V) reduction by DAB and quercetin was not influenced by glucose or AMP. Glucose that potentiates the inhibitory effects of caffeine, BAY U6751, and CP320626 on the glycogenolytic activity of GPa also enhanced the inhibitory effects of these GPIs on GPa-catalyzed As(V) reduction. AMP at high concentration alleviated the inhibition by BAY U6751 and CP320626 (whose antagonistic effect on GP-catalyzed glycogen breakdown is also AMP sensitive), whereas the inhibition in As(V) reduction by FP or caffeine was little affected by AMP. Thus, GPIs inhibit both the glycogenolytic and As(V)-reducing activities of GP, supporting that the latter is coupled to glycogenolysis. It was also shown that a GPa-rich extract of rat liver contained GSH-dependent As(V)-reducing activity that was inhibited by specific GPIs, suggesting that the liver-type GPa can also catalyze reduction of As(V).


Subject(s)
Arsenates/metabolism , Arsenites/metabolism , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Glycogen Phosphorylase, Liver Form/antagonists & inhibitors , Glycogen Phosphorylase, Muscle Form/antagonists & inhibitors , Glycogenolysis/drug effects , Xenobiotics/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Amides/pharmacology , Animals , Arabinose/pharmacology , Caffeine/pharmacology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Glycogen Phosphorylase, Liver Form/metabolism , Glycogen Phosphorylase, Muscle Form/metabolism , Imino Furanoses/pharmacology , Indoles/pharmacology , Male , Oxidation-Reduction , Quercetin/pharmacology , Quinolinic Acids/pharmacology , Rabbits , Rats , Rats, Wistar , Spectrometry, Fluorescence , Spectrophotometry, Atomic , Sugar Alcohols/pharmacology , Sulfhydryl Reagents/pharmacology
6.
Microbiology (Reading) ; 153(Pt 7): 2363-2372, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17600080

ABSTRACT

The evolution of bacterial pathogens from commensal organisms involves virulence gene acquisition followed by pathoadaptation to the new host, including inactivation of antivirulence loci (AVL). AVL are core ancestral genes whose expression is incompatible with the pathogenic lifestyle. Previous studies identified cadA (encoding lysine decarboxylase) as an AVL of Shigella spp. In this study, AVL of Shigella were identified by examining a phenotypic difference from its non-pathogenic ancestor, Escherichia coli. Unlike most E. coli strains, Shigella spp. are nicotinic acid auxotrophs, the pathway for the de novo synthesis of NAD being uniformly defective. In Shigella flexneri, this defect is due to alterations in the nadA and/or nadB genes encoding the enzyme complex that converts L-aspartate to quinolinate, a precursor to NAD synthesis. Quinolinate was found to inhibit invasion and cell-to-cell spread of Sh. flexneri 5a and its ability to induce polymorphonuclear neutrophil transepithelial migration. Virulence of other Shigella species was also inhibited by quinolinate. Introduction of functional nadA and nadB genes from E. coli K-12 into Sh. flexneri 5a restored its ability to synthesize quinolinate but also resulted in strong attenuation of virulence in this strain. The results define nadA and nadB as AVL in Shigella and validate the concept of pathoadaptive evolution of bacteria from commensal ancestors by inactivation of AVL. They also suggest that studies focusing on this form of bacterial evolution can identify novel inhibitors of virulence in other bacterial pathogens.


Subject(s)
NAD/biosynthesis , NAD/genetics , Quinolinic Acids/pharmacology , Shigella flexneri/pathogenicity , Virulence/genetics , Genes, Bacterial , HeLa Cells , Humans , Quinolinic Acids/metabolism , Shigella flexneri/genetics , Virulence/physiology
7.
J Neurosci ; 25(34): 7858-66, 2005 Aug 24.
Article in English | MEDLINE | ID: mdl-16120788

ABSTRACT

The NMDA ionotropic glutamate receptor is ubiquitous in mammalian central neurons. Because partial agonists bind to the same site as glutamate but induce less channel activation, these compounds provide an opportunity to probe the mechanism of activation of NMDA-type glutamate receptors. Molecular dynamics simulations and site-directed mutagenesis demonstrate that the partial agonist homoquinolinate interacts differently with binding pocket residues than glutamate. Homoquinolinate and glutamate induce distinct changes in the binding pocket, and the binding pocket exhibits significantly more motion with homoquinolinate bound than with glutamate. Patch-clamp recording demonstrates that single-channel activity induced by glutamate or by homoquinolinate has identical single-channel current amplitude and mean open-channel duration but that homoquinolinate slows activation of channel opening relative to glutamate. We hypothesize that agonist-induced conformational changes in the binding pocket control the efficacy of a subunit-specific activation step that precedes the concerted global change in the receptor-channel complex associated with ion channel opening.


Subject(s)
Glutamic Acid/analogs & derivatives , Glutamic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , Binding Sites/physiology , Cell Line , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/chemistry , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , Female , Glutamic Acid/pharmacology , Humans , Molecular Sequence Data , Protein Conformation , Quinolinic Acids/chemistry , Quinolinic Acids/metabolism , Quinolinic Acids/pharmacology , Receptors, N-Methyl-D-Aspartate/chemistry , Xenopus laevis
8.
Neuroscience ; 135(2): 463-74, 2005.
Article in English | MEDLINE | ID: mdl-16111817

ABSTRACT

Oxidative/nitrosative stress is involved in NMDA receptor-mediated excitotoxic brain damage produced by the glutamate analog quinolinic acid. The purpose of this work was to study a possible role of peroxynitrite, a reactive oxygen/nitrogen species, in the course of excitotoxic events evoked by quinolinic acid in the brain. The effects of Fe(TPPS) (5,10,15,20-tetrakis (4-sulfonatophenyl)porphyrinate iron (III)), an iron porphyrinate and putative peroxynitrite decomposition catalyst, were tested on lipid peroxidation and mitochondrial function in brain synaptic vesicles exposed to quinolinic acid, as well as on peroxynitrite formation, nitric oxide synthase and superoxide dismutase activities, lipid peroxidation, caspase-3-like activation, DNA fragmentation, and GABA levels in striatal tissue from rats lesioned by quinolinic acid. Circling behavior was also evaluated. Increasing concentrations of Fe(TPPS) reduced lipid peroxidation and mitochondrial dysfunction induced by quinolinic acid (100 microM) in synaptic vesicles in a concentration-dependent manner (10-800 microM). In addition, Fe(TPPS) (10 mg/kg, i.p.) administered 2 h before the striatal lesions, prevented the formation of peroxynitrite, the increased nitric oxide synthase activity, the decreased superoxide dismutase activity and the increased lipid peroxidation induced by quinolinic acid (240 nmol/microl) 120 min after the toxin infusion. Enhanced caspase-3-like activity and DNA fragmentation were also reduced by the porphyrinate 24 h after the injection of the excitotoxin. Circling behavior from quinolinic acid-treated rats was abolished by Fe(TPPS) six days after quinolinic acid injection, while the striatal levels of GABA, measured one day later, were partially recovered. The protective effects that Fe(TPPS) exerted on quinolinic acid-induced lipid peroxidation and mitochondrial dysfunction in synaptic vesicles suggest a primary action of the porphyrinate as an antioxidant molecule. In vivo findings suggest that the early production of peroxynitrite, altogether with the enhanced risk of superoxide anion (O2*-) and nitric oxide formation (its precursors) induced by quinolinic acid in the striatum, are attenuated by Fe(TPPS) through a recovery in the basal activities of nitric oxide synthase and superoxide dismutase. The porphyrinate-mediated reduction in DNA fragmentation simultaneous to the decrease in caspase-3-like activation from quinolinic acid-lesioned rats suggests a prevention in the risk of peroxynitrite-mediated apoptotic events during the course of excitotoxic damage in the striatum. In summary, the protective effects that Fe(TPPS) exhibited both under in vitro and in vivo conditions support an active role of peroxynitrite and its precursors in the pattern of brain damage elicited by excitotoxic events in the experimental model of Huntington's disease. The neuroprotective mechanisms of Fe(TPPS) are discussed.


Subject(s)
Brain Injuries/metabolism , Huntington Disease/prevention & control , Peroxynitrous Acid/metabolism , Porphyrins/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Analysis of Variance , Animals , Behavior, Animal , Blotting, Southern/methods , Brain Injuries/complications , Caspase 3 , Caspases/metabolism , Chromatography, High Pressure Liquid/methods , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electrophoresis, Agar Gel/methods , Huntington Disease/etiology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Mitochondria/physiology , Motor Activity/drug effects , Motor Activity/physiology , Nitric Oxide Synthase/metabolism , Quinolinic Acids/pharmacology , Rats , Rats, Wistar , Rotarod Performance Test/methods , Superoxide Dismutase/metabolism , Synaptic Vesicles/metabolism , Synaptosomes/physiology , Tyrosine/analogs & derivatives , Tyrosine/metabolism , gamma-Aminobutyric Acid/metabolism
9.
J Neurosci ; 25(9): 2285-94, 2005 Mar 02.
Article in English | MEDLINE | ID: mdl-15745954

ABSTRACT

Neurosteroids are produced de novo in neuronal and glial cells, which begin to express steroidogenic enzymes early in development. Studies suggest that neurosteroids may play important roles in neuronal circuit maturation via autocrine and/or paracrine actions. However, the mechanism of action of these agents is not fully understood. We report here that the excitatory neurosteroid pregnenolone sulfate induces a long-lasting strengthening of AMPA receptor-mediated synaptic transmission in rat hippocampal neurons during a restricted developmental period. Using the acute hippocampal slice preparation and patch-clamp electrophysiological techniques, we found that pregnenolone sulfate increases the frequency of AMPA-mediated miniature excitatory postsynaptic currents in CA1 pyramidal neurons. This effect could not be observed in slices from rats older than postnatal day 5. The mechanism of action of pregnenolone sulfate involved a short-term increase in the probability of glutamate release, and this effect is likely mediated by presynaptic NMDA receptors containing the NR2D subunit, which is transiently expressed in the hippocampus. The increase in glutamate release triggered a long-term enhancement of AMPA receptor function that requires activation of postsynaptic NMDA receptors containing NR2B subunits. Importantly, synaptic strengthening could also be triggered by postsynaptic neuron depolarization, and an anti-pregnenolone sulfate antibody scavenger blocked this effect. This finding indicates that a pregnenolone sulfate-like neurosteroid is a previously unrecognized retrograde messenger that is released in an activity-dependent manner during development.


Subject(s)
Neuronal Plasticity/drug effects , Pregnenolone/pharmacology , Presynaptic Terminals/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/drug effects , Age Factors , Animals , Animals, Newborn , Antibodies/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Chelating Agents/pharmacology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Hippocampus/cytology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Patch-Clamp Techniques/methods , Piperidines/pharmacology , Pregnenolone/immunology , Quinolinic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Synapses/physiology , Synaptic Transmission/drug effects , Tetrodotoxin/pharmacology , Time Factors
10.
Bioorg Med Chem ; 12(24): 6465-72, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15556764

ABSTRACT

Additional structural modifications of the new chemical entity, 2,8-dicyclopentyl-4-methylquinoline (DCMQ; MIC=6.25 microg/mL, M. tuberculosis H37Rv) resulted in the synthesis of four new series of the ring-substituted quinolinecarbohydrazides (series 1-4) constituting 22 analogues. All new derivatives were evaluated for in vitro antimycobacterial activities against drug-sensitive M. tuberculosis H37Rv strain. Certain ring-substituted-2-quinolinecarbohydrazide analogues described herein showed good inhibitory activity. In particular, analogues 4-(1-adamantyl)-2-quinolinecarbohydrazide (2d), 4,5-dicyclopentyl-2-quinolinecarbohydrazide (2e), 4,8-dicyclopentyl-2-quinolinecarbohydrazide (2f), and 4,5-dicyclohexyl-2-quinolinecarbohydrazide (2g) have exhibited the MIC value of 6.25 microg/mL. Further investigation of the most suitable lead prototype, 4-(1-adamantyl)-2-quinolinecarbohydrazide (2d, series 1) led to the synthesis of N2-alkyl/N2,N2-dialkyl/N2-aryl-4-(1-adamantyl)-2-quinolinecarboxamides (series 5) consisting of 13 analogues. Some of the synthesized carboxamides 7a, 7h, and 7m reported herein have exhibited excellent antimycobacterial activities in the range of 6.25-3.125 microg/mL against drug-sensitive and drug-resistant M. tuberculosis H37Rv strains.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Mycobacterium tuberculosis/drug effects , Quinolinic Acids/chemical synthesis , Amides , Anti-Bacterial Agents/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , Microbial Sensitivity Tests , Quinolinic Acids/pharmacology , Structure-Activity Relationship
11.
J Neurophysiol ; 86(4): 1644-51, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11600627

ABSTRACT

We have previously shown that presynaptic N-methyl-D-aspartate receptors (NMDARs) can facilitate glutamate release onto principal neurons in the entorhinal cortex (EC). In the present study, we have investigated the subunit composition of these presynaptic NMDARs. We recorded miniature alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated excitatory postsynaptic currents (mEPSCs), from visually identified neurons in layers II and V of the EC in vitro. In both layers, bath application of the NR2A/B subunit-selective agonist, homoquinolinic acid (HQA), resulted in a marked facilitation of mEPSC frequency. Blockade of presynaptic Ca(2+) entry through either NMDARs or voltage-gated Ca(2+) channels with Co(2+) prevented the effects of HQA, confirming that Ca(2+) entry to the terminal was required for facilitation. When the NR2B-selective antagonist, ifenprodil, was applied prior to HQA, the increase in mEPSC frequency was greatly reduced. In addition, we found that an NMDAR antagonist blocked frequency-dependent facilitation of evoked release and reduced mEPSC frequency in layer V. Thus we have demonstrated that NMDA autoreceptors in layer V of the EC bear the NR2B subunit, and that NMDARs are also present at terminals onto superficial neurons.


Subject(s)
Entorhinal Cortex/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Receptors, N-Methyl-D-Aspartate/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Autoreceptors/physiology , Calcium/metabolism , Entorhinal Cortex/cytology , Epilepsy/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Male , Piperidines/pharmacology , Quinolinic Acids/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/physiology , Tetrodotoxin/pharmacology
13.
Eur J Pharmacol ; 320(1): 87-94, 1997 Feb 05.
Article in English | MEDLINE | ID: mdl-9049607

ABSTRACT

The pharmacology of recombinant NR1a/NR2D NMDA receptors expressed in Xenopus oocytes was examined and compared to the pharmacology of NR1a/NR2A, NR1a/NR2B and NR1a/NR2C heteromers. The NR1/NR2D heteromer showed a pharmacological profile distinct from each of the other NR1/NR2 heteromers. This unique pharmacological profile was characterized by a relatively lower affinity for the agonist homoquinolinate and the antagonists 2-amino-5-phosphonopentanoate (D-AP5) and (R,E)-4-(3-phosphonoprop-2-enyl)piperazine-2-carboxylic acid (D-CPPene) but not for the antagonists (+/-)-4-(4-phenylbenzoyl) piperazine-2,3-dicarboxylic acid (PBPD) and alpha-amino-5-(phosphonomethyl)[1,1'-biphenyl]-3-propanoic acid (EAB515). NR2D-containing receptors displayed a pharmacological profile most similar to that observed for receptors containing the genetically related NR2C subunit. These findings parallel observations obtained for native NMDA receptors in the medial thalamus (presumed to contain NR2D subunits) and forebrain (presumed to contain NR2A and NR2B subunits). Thus, only compounds that discriminate between either NR2A- or NR2B-containing heteromers and NR2D-containing heteromers also discriminate between forebrain and medial thalamic NMDA receptors. While the pharmacology of the NR1a/NR2D receptor shows many parallels to the medial thalamic NMDA receptor, some differences were observed. Certain compounds which discriminate between medial thalamic and cerebellar (presumed to contain NR2C subunits) receptors (e.g., homoquinolinate, D-CPPene) did not show a similar selectivity for NR1a/NR2D receptors relative to NR1/NR2C receptors. Co-expression of NR1a, NR2B and NR2D subunits in Xenopus oocytes resulted in the formation of heteromeric complexes with unique pharmacological properties, suggesting the co-existence of these two distinct NR2 subunits in the same receptor complex.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Oocytes/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Biphenyl Compounds/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Isoquinolines/pharmacology , Piperazines/pharmacology , Propionates/pharmacology , Quinolinic Acids/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Tetrazoles/pharmacology , Transfection , Xenopus laevis
14.
Neurochem Int ; 28(4): 445-52, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8740453

ABSTRACT

Quinolinic acid is an endogenous neurotoxin with NMDA receptor agonist properties. As such it may be the etiologic agent in many diseases. In this paper the NMDA receptor agonist properties of quinolinic acid, as well as those of homoquinolinic acid, a non endogenous analogue, were investigated in Xenopus oocytes injected with 12-day-old rat cortical mRNA or with recombinant NMDA receptors. In oocytes injected with cortical mRNA, quinolinic acid was a weak NMDA receptor agonist: millimolar concentrations were necessary to induce responses that were smaller than maximal responses induced by NMDA; homoquinolinic acid and NMDA had similar affinities but different efficacies: maximal responses induced by homoquinolinic acid were larger than maximal responses induced by NMDA. Cortical mRNA, as verified by RT-PCR and restriction analysis, contains various NMDA subunits. In order to investigate if the low affinity or efficacy of quinolinic acid could be explained by receptor composition, the pharmacological properties of the putative agonists were investigated in oocytes expressing binary combinations of recombinant NMDA receptors. Quinolinic acid did not activate receptors containing NR1 + NR2C but did activate receptors containing NR1 + NR2A and NR1 + NR2B even if only at millimolar concentrations; homoquinolinic acid activated all subunit combinations but was less efficient than NMDA only in the NR1 + NR2C subunit combination. The relative efficacies of quinolinic acid and homoquinolinic acid were evaluated by comparing the maximal responses induced by these agonists with those induced by NMDA and glutamate in the same oocytes. The rank order of potency was quinolinic acid < NMDA < homoquinolinic acid < or = glutamate for the NR1 + NR2A and NR1 + NR2B combinations whereas for NR1 + NR2C it was quinolinic acid << << homoquinolinic acid < NMDA < or = glutamate. The use of quinolinic acid and homoquinolinic acid may thus help to identify endogenous receptors containing the NR2C subunit.


Subject(s)
Cerebral Cortex/metabolism , Nerve Tissue Proteins/physiology , Quinolinic Acid/pharmacology , Quinolinic Acids/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Female , Glutamic Acid/pharmacology , Glycine/pharmacology , Kainic Acid/pharmacology , Macromolecular Substances , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/drug effects , Oocytes/drug effects , Oocytes/physiology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Xenopus laevis
15.
Brain Res Bull ; 39(2): 69-73, 1996.
Article in English | MEDLINE | ID: mdl-8846115

ABSTRACT

Adult male Sprague-Dawley rats were exposed to 15 mM quinolinic acid solution or vehicle via bilateral intrastriatal dialytic administration for a period of 3 weeks. Animals were tested twice weekly for spontaneous behaviors and nocturnal activity during the 3-week dialysis period and for the 3 weeks following cessation of the dialysis period treatment. Nocturnal activity increased significantly (p < 0.005) during the first week of quinolinic acid exposure compared to vehicle exposed animals. The increase in nocturnal activity subsequently diminished to near control levels by the end of the 3-week dialysis period. During the 3-week period following cessation of dialysis, no significant differences were seen between quinolinic acid and vehicle-exposed animals. In addition, no differences were noted between quinolinic acid and vehicle-exposed animals in spontaneous behaviors either in the 3-week dialysis period or the 3-week period following cessation of dialysis. The results of this study are in agreement with other recent findings of transient nocturnal hyperactivity following striatal damage in rats. One possible explanation for the transient nature of this behavioral change is a transient effect of excitotoxicity in the striatum. During initial exposure to excitotoxins, nocturnal hypermotility could result from premorbid changes in neural function. With continued exposure, this behavioral effect may then diminish as a result of subsequent widespread striatal cell death.


Subject(s)
Corpus Striatum/physiology , Motor Activity/drug effects , Quinolinic Acids/pharmacology , Animals , Corpus Striatum/anatomy & histology , Corpus Striatum/enzymology , Dialysis , Electron Transport Complex IV/metabolism , Injections , Male , Nerve Degeneration/drug effects , Quinolinic Acids/administration & dosage , Rats , Rats, Sprague-Dawley
16.
Pharmacol Res ; 31(5): 325-9, 1995 May.
Article in English | MEDLINE | ID: mdl-7479531

ABSTRACT

The xanthomatine analogue, pyrphenoxone, which is known to diminish the incidence of cataract in animals and in man, was applied in two different in vivo models of cataract induced in rabbits by tryptophan-free dietary regimen and in rats by hypergalactosemic diet. The drug was also applied at different concentrations in an in vitro model of cataract. It was found that soluble proteins and sulphurated amino acids of the lens in all in vivo and in vitro models of cataract were higher after pyrphenoxone was applied. Furthermore, the drug treatment was followed by a dose-dependent increase in reduced glutathione content in the lens of rabbits and rats. The same was found in the in vitro model of cataract. These results suggest that pyrphenoxone may act by inducing various biochemical changes that lead to a protection of lens against oxidative processes.


Subject(s)
Eye/drug effects , Oxazines/pharmacology , Pyridines/pharmacology , Quinolinic Acids/pharmacology , Animals , Cataract/drug therapy , Diet , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glutathione/metabolism , Male , Rabbits , Rats , Rats, Sprague-Dawley
17.
Pharmacol Rev ; 45(3): 309-79, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8248282

ABSTRACT

In a little more than 10 years, the kynurenine metabolites of tryptophan have emerged from their former position as biochemical curiosities, to occupy a prominent position in research on the causes and treatment of several major CNS disorders. The pathway includes two compounds, quinolinic acid and kynurenic acid, which are remarkably specific in their pharmacological profiles: one is a selective agonist at receptors sensitive to NMDA, whereas the other is a selective antagonist at low concentrations at the strychnine-resistant glycine modulatory site associated with the NMDA receptor. It has been argued that these agents cannot be of physiological or pathological relevance because their normal extracellular concentrations, in the nanomolar range, are at least 3 orders of magnitude lower than those required to act at NMDA receptors. This is a facile argument, however, that ignores at least two possibilities. One is that both quinolinate and kynurenate may be present in very high concentrations locally at some sites in the brain that cannot be reflected in mean extracellular levels. Similar considerations apply to many neuroactive agents in the CNS. The fact that both compounds appear to be synthesised in, and thus emerge from, glial cells that are well recognised as enjoying a close physical and chemical relationship with some neurones in which the intercellular space may be severely restricted may support such a view. Certainly the realisation that NMDA receptors may not be fully saturated functionally with glycine would be consistent with the possibility that even quite low concentrations of kynurenate could maintain a partial antagonism at the glycine receptor. A second possibility is that there may be a subpopulation of NMDA receptors (or, indeed, for a quite different amino acid) that possesses a glycine modulatory site with a much lower sensitivity to glycine or higher sensitivity to kynurenate, making it more susceptible to fluctuations of endogenous kynurenine levels. Whatever the specific nature of their physiological roles, the presence of an endogenous selective agonist and antagonist acting at NMDA receptors must continue to present exciting possibilities for understanding the pathological basis of several CNS disorders as well as developing new therapeutic approaches. An imbalance in the production or removal of either of these substances would be expected to have profound implications for brain function, especially if that imbalance were present chronically.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Brain/metabolism , Kynurenic Acid/metabolism , Kynurenine/biosynthesis , Kynurenine/metabolism , Quinolinic Acids/metabolism , Animals , Brain/drug effects , Humans , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/blood , Kynurenine/physiology , Quinolinic Acids/blood , Quinolinic Acids/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
19.
J Neural Transm Gen Sect ; 94(2): 103-14, 1993.
Article in English | MEDLINE | ID: mdl-8110437

ABSTRACT

Discrete lesions in the medial or lateral subregion of the rostral caudate-putamen (CP) were induced by bilateral intracerebral injections of a low dose of quinolinic acid (30 nmol in 1 microliter/per side) in rats. Quinolinic acid lesions in the lateral CP potently reversed haloperidol-induced catalepsy (0.5 mg/kg,i.p.), while lesions in the medial CP were not effective. Spontaneous locomotor activity was not altered significantly after quinolinic acid lesions of either the medial or lateral CP. These results show that the lateral CP seems to be important for the expression of neuroleptic-induced catalepsy and thus further corroborate the concept of a functional heterogeneity of the striatum.


Subject(s)
Catalepsy/physiopathology , Caudate Nucleus/physiology , Haloperidol/antagonists & inhibitors , Putamen/physiology , Quinolinic Acids/pharmacology , Animals , Catalepsy/chemically induced , Caudate Nucleus/anatomy & histology , Caudate Nucleus/drug effects , Haloperidol/pharmacology , Male , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/physiology , Putamen/anatomy & histology , Putamen/drug effects , Rats , Rats, Sprague-Dawley
20.
Brain Res Dev Brain Res ; 67(2): 145-52, 1992 Jun 19.
Article in English | MEDLINE | ID: mdl-1380899

ABSTRACT

The developmental pharmacology of excitatory amino acid (EAA) receptors in the chick cochlear nucleus (nucleus magnocellularis, NM) was studied by means of bath application of drugs and recording of synaptically-evoked field potentials in brain slices taken from chicks aged embryonic day (E) 14 through hatching (E21). The abilities of various EAA agonists (N-methyl-D-aspartate [NMDA], kainic acid, and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid [AMPA]) to suppress postsynaptic responses by depolarization block and of EAA antagonists ((3-[RS]-2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid [CCP], dizocilpine [MK-801], 6-nitro-7-sulfamoyl-benzo(F)quinoxaline-2,3 dione [NBQX], 6-cyano-7-nitroquinoxaline-2,3-dione [CNQX] and 6,7-dinitroquinoxaline-2,3-dione [DNQX]) to suppress these responses directly were assessed quantitatively. The results support the existence of NMDA receptors in NM and suggest that the ability of these receptors to influence synaptically-evoked responses declines dramatically during the last week of embryonic life. The results similarly suggest that the non-NMDA receptors in NM undergo changes in density and/or function during a period of development when the cochlear nucleus is undergoing a variety of morphological and functional transformations.


Subject(s)
Cochlear Nerve/physiology , N-Methylaspartate/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Neurotransmitter/physiology , Synapses/physiology , Synaptic Transmission/drug effects , 6-Cyano-7-nitroquinoxaline-2,3-dione , Animals , Anticonvulsants/pharmacology , Chick Embryo , Cochlear Nerve/drug effects , Cochlear Nerve/embryology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Embryonic and Fetal Development , Evoked Potentials/drug effects , Ibotenic Acid/analogs & derivatives , Ibotenic Acid/pharmacology , Piperazines/pharmacology , Quinolinic Acid , Quinolinic Acids/pharmacology , Quinoxalines/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, Neurotransmitter/drug effects , Synapses/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...