Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 82: 11-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24607890

ABSTRACT

The tryptophan metabolite kynurenic acid (KYNA) is an endogenous antagonist of the α7 nicotinic acetylcholine receptor (α7nAChR) and, at higher concentrations, inhibits ionotropic glutamate receptors. Increases in KYNA levels are seen in brain and cerebrospinal fluid in individuals with schizophrenia (SZ) and may be causally related to cognitive deficits in SZ and other psychiatric diseases. As dysfunction of circuits involving GABAergic neurons in the prefrontal cortex (PFC) likely plays a role in the cognitive impairments seen in these disorders, we examined the effects of KYNA on extracellular GABA in this brain area. Applied to awake rats for 2 h by reverse dialysis, KYNA concentration-dependently and reversibly reduced extracellular GABA levels, with 300 nM KYNA causing a nadir of ∼45% of baseline concentrations. This effect was not duplicated by reverse dialysis of the selective glycineB receptor antagonist 7-Cl-KYNA (100 nM) or the AMPA/kainate receptor antagonist CNQX (100 µM), and was prevented by co-application of galantamine (5 µM), a positive allosteric modulator of the α7nAChR. Conversely, inhibition of endogenous KYNA formation by reverse dialysis of (S)-4-(ethylsulfonyl)benzoylalanine (ESBA; 5 mM) reversibly increased GABA levels in the PFC, reaching a peak of ∼160% of baseline concentrations. Co-infusion of 30 nM KYNA neutralized this effect. Taken together, these results demonstrate a role for endogenous KYNA in the bi-directional control of GABAergic neurotransmission in the PFC. Pharmacological manipulation of KYNA may therefore be useful in the treatment of GABAergic impairments in SZ and other brain disorders involving the PFC.


Subject(s)
Extracellular Space/metabolism , Kynurenic Acid/metabolism , Prefrontal Cortex/metabolism , gamma-Aminobutyric Acid/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Central Nervous System Agents/pharmacology , Cholinergic Agents/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Space/drug effects , Galantamine/pharmacology , Glutamic Acid/metabolism , Kynurenic Acid/antagonists & inhibitors , Male , Microdialysis , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Receptors, Glycine/antagonists & inhibitors , Receptors, Glycine/metabolism , Schiff Bases/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
2.
J Psychiatry Neurosci ; 38(5): 306-16, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23611177

ABSTRACT

BACKGROUND: Glutamate N-methyl-D-aspartate (NMDA) receptor antagonists exert fast-acting antidepressant effects, providing a promising way to develop a new classification of antidepressant that targets the glutamatergic system. In the present study, we examined the potential antidepressant action of 7-chlorokynurenic acid (7-CTKA), a glycine recognition site NMDA receptor antagonist, in a series of behavioural models of depression and determined the molecular mechanisms that underlie the behavioural actions of 7-CTKA. METHODS: We administered the forced swim test, novelty-suppressed feeding test, learned helplessness paradigm and chronic mild stress (CMS) paradigm in male rats to evaluate the possible rapid antidepressant-like actions of 7-CTKA. In addition, we assessed phospho-glycogen synthase kinase-3ß (p-GSK3ß) level, mammalian target of rapamycin (mTOR) function, and postsynaptic protein expression in the medial prefrontal cortex (mPFC) and hippocampus. RESULTS: Acute 7-CTKA administration produced rapid antidepressant-like actions in several behavioural tests. It increased p-GSK3ß, enhanced mTOR function and increased postsynaptic protein levels in the mPFC. Activation of GSK3ß by LY294002 completely blocked the antidepressant-like effects of 7-CTKA. Moreover, 7-CTKA did not produce rewarding properties or abuse potential. LIMITATIONS: It is possible that 7-CTKA modulates glutamatergic transmission, thereby causing enduring alterations of GSK3ß and mTOR signalling, although we did not provide direct evidence to support this possibility. Thus, the therapeutic involvement of synaptic adaptions engaged by 7-CTKA requires further study. CONCLUSION: Our findings demonstrate that acute 7-CTKA administration produced rapid antidepressant-like effects, indicating that the behavioural response to 7-CTKA is mediated by GSK3ß and mTOR signalling function in the mPFC.


Subject(s)
Antidepressive Agents/pharmacology , Kynurenic Acid/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Choice Behavior/drug effects , Chromones/administration & dosage , Chromones/pharmacology , Dose-Response Relationship, Drug , Enzyme Activators/pharmacology , Feeding Behavior/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Helplessness, Learned , Hippocampus/drug effects , Hippocampus/metabolism , Immobility Response, Tonic/drug effects , Kynurenic Acid/antagonists & inhibitors , Kynurenic Acid/pharmacology , Male , Microinjections , Morpholines/administration & dosage , Morpholines/pharmacology , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Stress, Psychological/psychology , TOR Serine-Threonine Kinases/metabolism
3.
Neuropharmacology ; 70: 254-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23422298

ABSTRACT

Kynurenic acid (KYNA), one of the main product of the kynurenine pathway originating from tryptophan, is considered to be neuroprotective. Dysregulation of KYNA activity is thought to be involved in neurodegenerative diseases, the physiopathology of which evokes excitotoxicity, oxidative stress and/or protein aggregation. The neuroprotective effect of KYNA is generally attributed to its antagonistic action on NMDA receptors. However, this single target action appears insufficient to support KYNA beneficial effects against complex neurodegenerative processes including neuroinflammation, ß-amyloid peptide (Aß) toxicity and apoptosis. Novel insights are therefore required to elucidate KYNA neuroprotective mechanisms. Here, we combined cellular, biochemical, molecular and pharmacological approaches to demonstrate that low micromolar concentrations of KYNA strongly induce neprilysin (NEP) gene expression, protein level and enzymatic activity increase in human neuroblastoma SH-SY5Y cells. Furthermore, our studies revealed that KYNA exerts a protective effect on SH-SY5Y cells by increasing their viability through a mechanism independent from NMDA receptors. Interestingly, KYNA also induced NEP activity and neuroprotection in mouse cortical neuron cultures the viability of which was more promoted than SH-SY5Y cell survival under KYNA treatment. KYNA-evoked neuroprotection disappeared in the presence of thiorphan, an inhibitor of NEP activity. NEP is a well characterized metallopeptidase whose deregulation leads to cerebral Aß accumulation and neuronal death in Alzheimer's disease. Therefore, our results suggest that a part of the neuroprotective role of KYNA may depend on its ability to induce the expression and/or activity of the amyloid-degrading enzyme NEP in nerve cells.


Subject(s)
Kynurenic Acid/pharmacology , Neprilysin/biosynthesis , Neurons/physiology , Neuroprotective Agents/pharmacology , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction , Gene Expression/drug effects , Humans , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/antagonists & inhibitors , Mice , Neprilysin/antagonists & inhibitors , Neprilysin/metabolism , Neuroprotective Agents/metabolism , Primary Cell Culture , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Thiorphan/pharmacology
4.
Article in English | MEDLINE | ID: mdl-23178230

ABSTRACT

Antipsychotics, which act predominantly as dopamine D2 receptor antagonists, have several shortcomings. The exact pathophysiological mechanism leading to dopaminergic dysfunction in schizophrenia is still unclear, but inflammation has been postulated to be a key player in the pathophysiology of the disorder. A dysfunction in activation of the type 1 immune response seems to be associated with an imbalance in tryptophan/kynurenine metabolism; the degrading enzymes involved in this metabolism are regulated by cytokines. Kynurenic acid (KYNA), an N-methyl-d-aspartate antagonist, was found to be increased in critical regions of the central nervous system (CNS) in schizophrenia, resulting in reduced glutamatergic neurotransmission. The differential activation of microglial cells and astrocytes as functional carriers of the immune system in the CNS may also contribute to this imbalance. The immunological effects of many existing antipsychotics, however, rebalance in part the immune imbalance and overproduction of KYNA. The immunological imbalance results in an inflammatory state combined with increased prostaglandin E(2) production and increased cyclo-oxygenase-2 (COX-2) expression. Growing evidence from clinical studies with COX-2 inhibitors points to favorable effects of anti-inflammatory therapy in schizophrenia, in particular in an early stage of the disorder. Further options for immunomodulating therapies in schizophrenia will be discussed.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Schizophrenia/drug therapy , Schizophrenia/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Controlled Clinical Trials as Topic/methods , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/physiology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/prevention & control , Kynurenic Acid/antagonists & inhibitors , Kynurenic Acid/metabolism , Schizophrenia/metabolism , Treatment Outcome
5.
J Neurol Sci ; 323(1-2): 1-8, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22939820

ABSTRACT

The kynurenine pathway (KP), the primary route of tryptophan degradation in mammalian cells, consists of many metabolites including kynurenic acid (KYNA), quinolinic acid (QUIN), 3-hydroxykynurenine (3-HK) and picolinic acid (PIC). The former two are neuroactive, while the latter two are molecules with pro-oxidants and antioxidants properties. These agents are considered to be involved in aging and numerous neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). Several studies have demonstrated that altered kynurenine metabolism plays an important role in the pathogenesis of this group of diseases. The important metabolites and key enzymes show significant importance in those disorders. Both analogs of the neuroprotective metabolites and small molecule enzyme inhibitors preventing the formation of neurotoxic compounds may have potential therapeutic significance. In this review we discuss the mechanistic and therapeutic considerations of KP in aging and the main neurodegenerative diseases and review the updated knowledge in this therapeutic field.


Subject(s)
Kynurenine/metabolism , Molecular Targeted Therapy , Neurodegenerative Diseases/metabolism , Aging/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Animals , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Huntington Disease/drug therapy , Huntington Disease/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/metabolism , Kynurenic Acid/antagonists & inhibitors , Kynurenic Acid/metabolism , Kynurenine/analogs & derivatives , Kynurenine/pharmacology , Kynurenine/therapeutic use , Microglia/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/prevention & control , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxins/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/prevention & control , Quinolinic Acid/antagonists & inhibitors , Quinolinic Acid/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Tryptophan/metabolism
6.
Cent Nerv Syst Agents Med Chem ; 9(1): 32-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-20021336

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult onset, progressive and fatal motor neuron degenerative disease [1]. The aetiology of ALS is currently unknown, though strongly suggested to be multifactorial. Recently, the kynurenine pathway (KP) has emerged as a potential contributing factor [2]. The KP is a major route for the metabolism of tryptophan, generating neuroactive intermediates in the process. These catabolites include the excitotoxic N-methyl-D-aspartate (NMDA) receptor agonist, quinolinic acid (QUIN) [3] and the neuroprotective NMDA receptor antagonist, kynurenic acid (KYNA) [4,5]. These catabolites appear to play a key role in the communication between the nervous and immune systems, and also in modulating cell proliferation and tissue function [6]. As the cause of ALS is still unknown, there is presently no efficient treatment for it. Currently, Riluzole is the drug of choice but its effect is relatively modest [7]. Targeting the KP, hence, could offer a new therapeutic option to improve ALS treatment [8]. Several drugs that block the KP are already under investigation by our laboratory and others, some of which are in or about to enter clinical trials for other diseases. For example, the KP inhibitors, Teriflunomide (Sanofi-Aventis) and Laquinimod (Teva Neuroscience). Recently, a KP inhibitor has also reached the Japan market as an immunomodulative drug [9]: Tranilast/Rizaben (Angiogen Ltd.) is an anthranilic acid derivative [8]. Finally, the 8-hydroxyquinolinine metal attenuating compounds, Clioquinol and PBT2, interestingly have close structural similarity with KYNA and QUIN. Such drugs would open a new and important therapeutic door for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/pathology , Functional Laterality/physiology , Kynurenic Acid/antagonists & inhibitors , Kynurenine/antagonists & inhibitors , Motor Neurons/drug effects , Quinolinic Acid/metabolism , Riluzole/therapeutic use , Tryptophan/metabolism , Adult , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Brain/drug effects , D-Aspartic Acid/metabolism , Humans , Japan , Kynurenic Acid/metabolism , Kynurenine/metabolism , Motor Neurons/physiology , N-Methylaspartate/metabolism , Signal Transduction/drug effects , Tryptophan/antagonists & inhibitors
7.
Eur J Neurosci ; 29(3): 529-38, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187269

ABSTRACT

We tested the hypothesis that fluctuations in the levels of kynurenic acid (KYNA), an endogenous antagonist of the alpha7 nicotinic acetylcholine (ACh) receptor, modulate extracellular ACh levels in the medial prefrontal cortex in rats. Decreases in cortical KYNA levels were achieved by local perfusion of S-ESBA, a selective inhibitor of the astrocytic enzyme kynurenine aminotransferase II (KAT II), which catalyses the formation of KYNA from its precursor L-kynurenine. At 5 mm, S-ESBA caused a 30% reduction in extracellular KYNA levels, which was accompanied by a two-threefold increase in basal cortical ACh levels. Co-perfusion of KYNA in the endogenous range (100 nm), which by itself tended to reduce basal ACh levels, blocked the ability of S-ESBA to raise extracellular ACh levels. KYNA perfusion (100 nm) also prevented the evoked ACh release caused by d-amphetamine (2.0 mg/kg). This effect was duplicated by the systemic administration of kynurenine (50 mg/kg), which resulted in a significant increase in cortical KYNA formation. Jointly, these data indicate that astrocytes, by producing and releasing KYNA, have the ability to modulate cortical cholinergic neurotransmission under both basal and stimulated conditions. As cortical KYNA levels are elevated in individuals with schizophrenia, and in light of the established role of cortical ACh in executive functions, our findings suggest that drugs capable of attenuating the production of KYNA may be of benefit in the treatment of cognitive deficits in schizophrenia.


Subject(s)
Acetylcholine/metabolism , Astrocytes/metabolism , Cerebral Cortex/metabolism , Kynurenic Acid/metabolism , Presynaptic Terminals/metabolism , Amphetamine/pharmacology , Animals , Astrocytes/drug effects , Central Nervous System Stimulants/pharmacology , Cerebral Cortex/drug effects , Enzyme Inhibitors/pharmacology , Kynurenic Acid/antagonists & inhibitors , Male , Nicotinic Antagonists/metabolism , Nicotinic Antagonists/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Presynaptic Terminals/drug effects , Rats , Rats, Wistar , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Schizophrenia/physiopathology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Transaminases/antagonists & inhibitors , Transaminases/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , alpha7 Nicotinic Acetylcholine Receptor
8.
Neuroscience ; 159(1): 196-203, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19138730

ABSTRACT

Fluctuations in the endogenous levels of kynurenic acid (KYNA), a potent alpha7 nicotinic and NMDA receptor antagonist, affect extracellular dopamine (DA) concentrations in the rat brain. Moreover, reductions in KYNA levels increase the vulnerability of striatal neurons to NMDA receptor-mediated excitotoxic insults. We now assessed the role of a key KYNA-synthesizing enzyme, kynurenine aminotransferase II (KAT II), in these processes in the rodent striatum, using KAT II KO mice-which have reduced KYNA levels-and the selective KAT II inhibitor (S)-4-(ethylsulfonyl)benzoylalanine (S-ESBA) as tools. S-ESBA (applied by reverse dialysis) raised extracellular DA levels in the striatum of KYNA-deficient mice threefold and caused a much larger, 15-fold increase in wild-type mice. In the rat striatum, S-ESBA produced a 35% reduction in extracellular KYNA, which was accompanied by a 270% increase in extracellular DA. The latter effect was abolished by co-infusion of 100 nM KYNA. Intrastriatal S-ESBA pre-treatment augmented the size of a striatal quinolinate lesion by 370%, and this potentiation was prevented by co-infusion of KYNA. In separate animals, acute inhibition of KAT II reduced the de novo synthesis of KYNA during an early excitotoxic insult without enhancing the formation of the related neurotoxic metabolites 3-hydroxykynurenine and quinolinate. Taken together, these results provide further support for the concept that KAT II is a critical determinant of functionally relevant KYNA fluctuations in the rodent striatum.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Extracellular Fluid/metabolism , Kynurenic Acid/antagonists & inhibitors , Kynurenic Acid/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Animals, Newborn , Corpus Striatum/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Extracellular Fluid/drug effects , Functional Laterality , Kynurenic Acid/pharmacology , Kynurenine/analogs & derivatives , Kynurenine/pharmacology , Mice , Mice, Knockout , Microdialysis/methods , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Norbornanes/pharmacology , Quinolinic Acid/toxicity , Transaminases/deficiency , Tritium/metabolism
9.
Pharmacol Rep ; 59(3): 268-73, 2007.
Article in English | MEDLINE | ID: mdl-17652826

ABSTRACT

We have evaluated the effect of diabetes-mimicking conditions on the inhibition of kynurenic acid (KYNA) production exerted by mitochondrial toxins: 3-nitropropionic acid (3-NPA) and aminooxyacetic acid (AOAA), by endogenous agonists of glutamate receptors: L-glutamate and L-cysteine sulfinate, and by a risk factor of atherosclerosis, D,L-homocysteine. Hyperglycemia (30 mM; 2 h) itself did not influence KYNA synthesis in brain cortical slices. However, it significantly enhanced the inhibitory effects of 3-NPA, AOAA and D,L-homocysteine, but not of L-glutamate and L-cysteine sulfinate, on KYNA production. Their IC(50) values were lowered from 5.8 (4.5-7.4) to 3.7 (3.1-4.5) mM (p < 0.01), from 11.6 (8.6-15.5) to 7.1 (4.9-10.3) microM (p < 0.05), and from 4.5 (3.5-5.8) to 2.4 (1.8-3.2) mM (p < 0.01), respectively. The obtained data suggest that during hyperglycemia, the mitochondrial impairment and high levels of D,L-homocysteine evoke stronger inhibition of KYNAsynthesis what may further exacerbate brain dysfunction and play a role in central complications of diabetes.


Subject(s)
Aminooxyacetic Acid/toxicity , Brain/metabolism , Homocysteine/toxicity , Hyperglycemia/metabolism , Kynurenic Acid/metabolism , Mitochondria/drug effects , Nitro Compounds/toxicity , Propionates/toxicity , Animals , Brain/drug effects , Glutamic Acid/toxicity , Kynurenic Acid/antagonists & inhibitors , Male , Rats , Rats, Wistar
10.
J Pharmacol Exp Ther ; 322(1): 48-58, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17446300

ABSTRACT

Galantamine, a drug used to treat Alzheimer's disease, is a nicotinic allosteric potentiating ligand, and kynurenic acid (KYNA), a neuroactive metabolite of the kynurenine pathway, is an endogenous noncompetitive inhibitor of alpha7* nicotinic receptors (nAChRs) [the asterisk next to the nAChR subunit is intended to indicate that the exact subunit composition of the receptor is not known (Pharmacol Rev 51:397-401, 1999)]. Here, possible interactions between KYNA and galantamine at alpha7* nAChRs were examined in vitro and in vivo. In the presence of tetrodotoxin (TTX), approximately 85% of cultured hippocampal neurons responded to choline (0.3-30 mM) with alpha7* nAChR-subserved whole-cell (type IA) currents. In the absence of TTX and in the presence of glutamate receptor antagonists, choline triggered inhibitory postsynaptic currents (IPSCs) by activating alpha7* nAChRs on GABAergic neurons synapsing onto the neurons under study. Galantamine (1-10 microM) potentiated, whereas KYNA (10 nM-1 mM) inhibited, choline-triggered responses. Galantamine (1 microM), applied before KYNA, shifted to the right the concentration-response relationship for KYNA to inhibit type IA currents, increasing the IC(50) of KYNA from 13.9 +/- 8.3 to 271 +/- 131 microM. Galantamine, applied before or after KYNA, antagonized inhibition of choline-triggered IPSCs by KYNA. Local infusion of KYNA (100 nM) in the rat striatum reduced extracellular dopamine levels in vivo. This effect resulted from alpha7* nAChR inhibition and was blocked by coapplied galantamine (1-5 microM). It is concluded that galantamine competitively antagonizes the actions of KYNA on alpha7* nAChRs. Reducing alpha7* nAChR inhibition by endogenous KYNA may be an important determinant of the effectiveness of galantamine in neurological and psychiatric disorders associated with decreased alpha7* nAChR activity in the brain.


Subject(s)
Galantamine/pharmacology , Kynurenic Acid/antagonists & inhibitors , Receptors, Nicotinic/drug effects , Allosteric Regulation , Alzheimer Disease/drug therapy , Animals , Atropine/pharmacology , Cells, Cultured , Choline/pharmacology , Corpus Striatum/drug effects , Dopamine/metabolism , Male , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor
11.
Neurochem Res ; 30(8): 963-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16258845

ABSTRACT

In the present study we demonstrate for the first time that both kynurenine aminotransferase (KAT) isoforms I and II are present in the permanent immature rat oligodendrocytes cell line (OLN-93). Moreover, we provide evidence that OLN-93 cells are able to synthesize kynurenic acid (KYNA) from exogenously added L-kynurenine and we characterize its regulation by extrinsic factors. KYNA production in OLN-93 cells was depressed in the presence of aminotransferase inhibitor, aminooxyacetic acid and was not affected by depolarizing agents such as 50 mM K+ and 4-aminopyridine. Glutamate agonists, L-glutamate and D,L-homocysteine significantly decreased KYNA production. Selective agonist of ionotropic glutamate receptors Amino-2,3-dihydro-5-methyl-3-oxo-4-isoxazolepropionic acid (AMPA) lowered KYNA production in OLN-93 cell line, whereas N-methyl-D-aspartate (NMDA) had no influence on KYNA production. Furthermore, KYNA synthesis in OLN-93 cells was decreased in a concentration-dependent manner by amino acids transported by L-system, L-leucine, L-cysteine and L-tryptophan. The role of KYNA synthesis in oligodendrocytes needs further investigation.


Subject(s)
Isoenzymes/metabolism , Kynurenic Acid/metabolism , Oligodendroglia/enzymology , Transaminases/metabolism , Amino Acids/metabolism , Animals , Cell Line , Immunohistochemistry , Kynurenic Acid/antagonists & inhibitors , Oligodendroglia/metabolism , Rats
12.
J Biomol Struct Dyn ; 22(4): 399-410, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15588104

ABSTRACT

N-Methyl-D-Aspartate (NMDA) receptors are the ligand gated as well as voltage sensitive ionotropic glutamate receptors, widely distributed in the vertebrate central nervous system and they play critical role in the pathogenesis of schizophrenia. Molecular dynamics simulations have been carried out on high resolution crystal structure of NR1 subunit of NMDA receptor ligand binding core (S1S2) in four different conformations. We have investigated consequence of D481N/K483Q double mutation of NR1 subunit from simulation results of (a) glycine bound form (WG), (b) unbound (closed-apo) form (WOG), (c) a double mutated form (DM), and (d) the antagonist (5,7-dichlorokynuric acid) bound form (DCKA). The MD simulations and simulated annealing for 4ns show a distinct conformation for the double mutated conformation that neither follows the antagonist nor apo conformation. There are two distinct sites, loop1 and loop2 where the double mutated structure in its glycine bound form shows significant RMSD deviations as compared to the wild-type. The interactions of glycine with the receptor remain theoretically unchanged in the double mutated structure and there is no detachment of S1S2 domains. The results suggest that separation of S1 and S2 domains may not be essential for channel inactivation. Therefore, it is hypothesized that hypoactivation of NMDA receptor channels may arise out of the conformational changes at non-conserved Loop1 and Loop2 regions observed in the mutated structure. The Loop1 and Loop2 regions responsible for inter-subunit interactions in a functional NMDA receptor, may therefore, render the ligand bound form defunct. This may account for behavioral anomalies due to receptor inactivation seen in grin1 mutated mice.


Subject(s)
Computer Simulation , Glycine/metabolism , N-Methylaspartate/chemistry , N-Methylaspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Amino Acid Sequence , Binding Sites/genetics , Crystallography, X-Ray , Glycine/chemistry , Hydrogen Bonding , Kynurenic Acid/antagonists & inhibitors , Kynurenic Acid/metabolism , Models, Molecular , Molecular Sequence Data , N-Methylaspartate/genetics , Point Mutation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Water/chemistry
13.
FEBS Lett ; 577(3): 381-5, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15556614

ABSTRACT

Kynurenine aminotransferase (KAT) catalyzes the formation of kynurenic acid (KYNA), the natural antagonist of ionotropic glutamate receptors. This study tests potential substrates and assesses the effects of amino acids and keto acids on the activity of mosquito KAT. Various keto acids, when simultaneously present in the same reaction mixture, display a combined effect on KAT catalyzed KYNA production. Moreover, methionine and glutamine show inhibitory effects on KAT activity, while cysteine functions as either an antagonist or an inhibitor depending on the concentration. Therefore, the overall level of keto acids and cysteine might modulate the KYNA synthesis. Results from this study will be useful in the study of KAT regulation in other animals.


Subject(s)
Culicidae/metabolism , Cysteine/pharmacology , Keto Acids/pharmacology , Kynurenic Acid/antagonists & inhibitors , Transaminases/metabolism , Animals , Baculoviridae/genetics , Catalysis , Cells, Cultured , Culicidae/cytology , Dose-Response Relationship, Drug , Kinetics , Kynurenic Acid/metabolism , Substrate Specificity
14.
Biol Psychiatry ; 56(4): 255-60, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15312813

ABSTRACT

BACKGROUND: Recent studies show that endogenous levels of kynurenic acid (KYNA) are increased in the cerebrospinal fluid of schizophrenic patients. Prepulse inhibition (PPI) of the acoustic startle reflex is an operational measure of sensorimotor gating that is reduced in neuropsychiatric disorders, such as schizophrenia. Previous studies show that administration of N-methyl-D-aspartate (NMDA) receptor antagonists, such as phencyclidine or MK-801, leads to deficits in sensorimotor gating that mimic those observed in schizophrenic patients. METHODS: The present study examined the effects of the endogenous NMDA receptor antagonist KYNA on startle and PPI in rats. Elevation of endogenous brain levels of KYNA was achieved through intraperitoneal (IP) administration of kynurenine (100 mg/kg), the precursor of KYNA, or by intravenous administration of PNU 156561A (10 mg/kg). RESULTS: A fourfold increase in brain KYNA levels, as induced by kynurenine or PNU 156561A, significantly reduced PPI. There were no differences in startle magnitudes between control rats and drug-treated rats. The disruption of PPI was restored by administration of the antipsychotic drugs haloperidol (.2 mg/kg, IP) or clozapine (7.5 mg/kg, IP). CONCLUSIONS: The present results suggest that brain KYNA serves as an endogenous modulator of PPI and are consistent with the hypothesis that KYNA contributes to the pathophysiology of schizophrenia.


Subject(s)
Brain/metabolism , Kynurenic Acid/metabolism , Neural Inhibition/drug effects , Reflex, Startle/drug effects , Acoustic Stimulation/methods , Animals , Antipsychotic Agents/pharmacology , Brain/drug effects , Brain/radiation effects , Brain Chemistry/drug effects , Butyrates/pharmacology , Clozapine/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Radiation , Drug Interactions , Haloperidol/pharmacology , Kynurenic Acid/antagonists & inhibitors , Kynurenine/pharmacology , Male , Neural Inhibition/physiology , Probenecid/pharmacology , Rats
15.
Naunyn Schmiedebergs Arch Pharmacol ; 369(3): 300-4, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14770276

ABSTRACT

Kynurenic acid (KYNA) is a broad-spectrum antagonist at all subtypes of ionotropic glutamate receptors, but is preferentially active at the strychnine-insensitive glycine allosteric site of the N-methyl-D-aspartate (NMDA) receptor and is also a non-competitive antagonist at the alpha7 nicotinic receptor. KYNA occurs in the CNS, urine, serum and amniotic fluid. Whilst it possesses anticonvulsant and neuroprotective properties in the brain, its role in the periphery, however, is unknown. In this study we demonstrated the presence of kynurenine aminotransferase (KAT) I and II in the cytoplasm of bovine aortic endothelial cells (BAEC). BAEC incubated in the presence of the KYNA precursor L-kynurenine synthesized KYNA concentration- and time-dependently. KYNA production was inhibited by the aminotransferase inhibitor aminooxyacetic acid but was not affected by a depolarising concentration of K(+) or by 4-aminopyridine. The glutamate agonists L-aspartate and L-glutamate depressed KYNA production significantly. The selective ionotropic glutamate receptor agonists alpha-amino-2,3-dihydro-5-methyl-3-oxo-4-isoxazolepropionic acid (AMPA) and NMDA were ineffective in this respect. D,L-Homocysteine and L-homocysteine sulphinic acid lowered KYNA production in BAEC. Further investigations are needed to assess the role and importance of KYNA in vessels and peripheral tissues.


Subject(s)
Aorta/cytology , Endothelium, Vascular/drug effects , Homocysteine/pharmacology , Kynurenic Acid/metabolism , Animals , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Kynurenic Acid/antagonists & inhibitors , Male , Transaminases/metabolism
16.
Neurosci Lett ; 346(1-2): 97-100, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12850557

ABSTRACT

In the present study the effect of endogenous sulphur-containing amino acids, L-cysteine sulphinate, L-cysteate, L-homocysteine sulphinate and L-homocysteate, on the production of glutamate receptor antagonist, kynurenic acid (KYNA), was evaluated. The experiments comprised the measurements of (a). KYNA synthesis in rat cortical slices and (b). the activity of KYNA biosynthetic enzymes, kynurenine aminotransferases (KATs). All studied compounds reduced KYNA production and inhibited the activity of KAT I and/or KAT II, thus acting most probably intracellularly. L-Cysteine sulphinate in very low, micromolar concentrations selectively affected the activity of KAT II, the enzyme catalyzing approximately 75% of KYNA synthesis in the brain. L-Cysteine sulphinate potency was higher than other studied sulphur-containing amino acids, than L-aspartate, L-glutamate, or any other known KAT II inhibitor. Thus, L-cysteine sulphinate might act as a modulator of KYNA formation in the brain.


Subject(s)
Cerebral Cortex/drug effects , Cysteine/pharmacology , Enzyme Inhibitors/pharmacology , Kynurenic Acid/antagonists & inhibitors , Transaminases/antagonists & inhibitors , Amino Acids, Sulfur/pharmacology , Animals , Cerebral Cortex/enzymology , Cysteine/analogs & derivatives , Dose-Response Relationship, Drug , In Vitro Techniques , Kynurenic Acid/metabolism , Male , Rats , Rats, Wistar , Transaminases/biosynthesis
17.
Neuropsychopharmacology ; 28(10): 1770-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12865892

ABSTRACT

The mode of action by which the atypical antipsychotic drug clozapine exerts its superior efficacy to ameliorate both positive and negative symptoms is still unknown. In the present in vivo electrophysiological study, we investigate the effects of haloperidol (a typical antipsychotic drug) and clozapine on ventral tegmental area (VTA) dopamine (DA) neurons in a situation of hyperdopaminergic activity in order to mimic tentatively a condition similar to that seen in schizophrenia. Increased DA transmission was induced by elevating endogenous levels of the N-methyl-D-aspartate receptor and alpha7(*) nicotinic receptor antagonist kynurenic acid (KYNA; by means of PNU 156561A, 40 mg /kg, i.v.). In control rats, i.v. administered haloperidol (0.05-0.8 mg/kg) or clozapine (1.25-10 mg/kg) was associated with increased firing rate and burst firing activity of VTA DA neurons. However, in rats displaying hyperdopaminergia (induced by elevated levels of KYNA), the effects of clozapine on VTA DA neurons were converted into pure inhibitory responses, including decrease in burst firing activity. In contrast, haloperidol still produced an excitatory action on VTA DA neurons in rats with elevated levels of endogenous brain KYNA. The results of the present study suggest that clozapine facilitates or inhibits VTA DA neurotransmission, depending on brain concentration of KYNA. Such an effect of clozapine may be related to its unique effect in also ameliorating negative symptoms of schizophrenia.


Subject(s)
Clozapine/pharmacology , Dopamine/metabolism , GABA Antagonists/pharmacology , Kynurenic Acid/metabolism , Neurons/drug effects , Ventral Tegmental Area/drug effects , Action Potentials/drug effects , Animals , Butyrates/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Haloperidol/pharmacology , Kynurenic Acid/antagonists & inhibitors , Male , Neural Inhibition/drug effects , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology
18.
Psychopharmacology (Berl) ; 166(4): 360-5, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12601505

ABSTRACT

RATIONALE: Histamine plays an important role in modulating acquisition and retention in learning and memory process in experimental animals. OBJECTIVES: We examined the effects of polyamine and histamine on the N-methyl- d-aspartate (NMDA) receptor glycine site antagonist 7-chlorokynurenic acid-induced spatial memory deficits in radial maze performance in rats. METHOD: Effects of histamine (0.5 or 1 nmol/site intracerebroventricularly), spermidine (1 nmol/site, intracerebroventricularly) and spermine (1 nmol/site, intracerebroventricularly) on spatial memory deficit in 9-week-old-male Wistar rats were observed. Both reference and working memory errors occurred in radial maze performance in rats, following intracerebroventricular injection of 7-chlorokynurenic acid (10 nmol/site). RESULTS: Spermidine (1 nmol/site, intracerebroventricularly) or spermine (1 nmol/site, intracerebroventricularly) antagonized 7-chlorokynurenic acid-induced deficits on working memory but not on reference memory errors. Intracerebroventricular histamine (0.5 or 1 nmol/site) or thioperamide (100 nmol/site) also ameliorated 7-chlorokynurenic acid-induced working memory deficits. To determine whether the effects of histamine involve histamine receptors, the effects of some methylhistamines were examined. The effects of R-alpha-methylhistamine on radial maze performance were mimicked by histamine. N(alpha)-methylhistamine had no effect on 7-chlorokynurenic acid-induced memory deficits, whereas 1-methylhistamine, but not 3-methylhistamine reversed 7-chlorokynurenic acid-induced working memory deficits. CONCLUSION: These results suggest that the amelioration of 7-chlorokynurenic acid-induced working memory deficits by histamine may involve a direct action of histamine at the polyamine sites on NMDA receptors.


Subject(s)
Excitatory Amino Acid Antagonists/toxicity , Histamine/pharmacology , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/toxicity , Memory Disorders/chemically induced , Polyamines/pharmacology , Animals , Kynurenic Acid/antagonists & inhibitors , Male , Methylhistamines/pharmacology , Motor Activity/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spermidine/pharmacology , Spermine/pharmacology
19.
Pol J Pharmacol ; 55(5): 741-6, 2003.
Article in English | MEDLINE | ID: mdl-14704470

ABSTRACT

The present studies sought to determine the concentrations of endogenous kynurenic acid (KYNA) and to measure the activity of kynurenine aminotransferases (KAT) I and II in the discrete brain regions of 3- and 6-month old WAG/Rij rats, a genetic model of absence epilepsy. Analogues experiments were performed using age-matched ACI rats, which served as a non-epileptic control. The age-dependent increase in KYNA concentration in the frontal cortex of WAG/Rij rats was considerably reduced in comparison to what was found in ACI rats. Consequently, the concentration of KYNA in the frontal cortex of epileptic rats was significantly lower than in non-epileptic controls. There were no such strain differences in other brain regions. The activities of KAT I and II also showed age-dependent increase with an exception for KAT II in the frontal cortex. Our data suggest that selective deficits of endogenous KYNA may account for increased excitability in the frontal cortex, which in turn may lead to the development of spontaneous spike-wave discharges in WAG/Rij rats.


Subject(s)
Epilepsy, Absence/genetics , Frontal Lobe/chemistry , Kynurenic Acid/chemistry , Kynurenic Acid/metabolism , Rats, Inbred Strains/genetics , Aging/physiology , Animals , Brain Chemistry/genetics , Corpus Striatum/chemistry , Disease Models, Animal , Electroencephalography , Frontal Lobe/enzymology , Frontal Lobe/physiopathology , Kynurenic Acid/antagonists & inhibitors , Kynurenine , Rats , Thalamic Nuclei/chemistry , Thalamic Nuclei/enzymology , Transaminases/chemistry , Transaminases/metabolism
20.
Neurosci Lett ; 330(1): 49-52, 2002 Sep 13.
Article in English | MEDLINE | ID: mdl-12213632

ABSTRACT

The aim of the present study was to evaluate the effect of mitochondrial inhibitors, 1-methyl-4-phenylpyridinium (MPP(+)) and 3-nitropropionic acid (3-NPA), on the brain production of endogenous glutamate antagonist, kynurenic acid (KYNA). MPP(+) and 3-NPA dose-dependently impaired the synthesis of KYNA in rat cortical slices. Enzymatic studies revealed that MPP(+) inhibits in a concentration-dependent manner the activity of kynurenine aminotransferase II (KAT II), but not the activity of kynurenine aminotransferase I (KAT I). 3-NPA impaired the activity of both enzymes, KAT I and KAT II. Thus, MPP(+)- and 3-NPA-evoked neurotoxicity may be at least partially associated with the depletion of KYNA.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Cerebral Cortex/drug effects , Kynurenic Acid/metabolism , Propionates/pharmacology , Transaminases/biosynthesis , Animals , Cerebral Cortex/enzymology , Kynurenic Acid/antagonists & inhibitors , Male , Mitochondria/drug effects , Mitochondria/enzymology , Nitro Compounds , Rats , Rats, Wistar , Transaminases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...