Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 355(1): 76-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26265321

ABSTRACT

Currently approved antidepressant drug treatment typically takes several weeks to be effective. The noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist ketamine has shown efficacy as a rapid-acting treatment of depression, but its use is associated with significant side effects. We assessed effects following blockade of the glycineB co-agonist site of the NMDA receptor, located on the GluN1 subunit, by the selective full antagonist 7-chloro-kynurenic acid (7-Cl-KYNA), delivered by systemic administration of its brain-penetrant prodrug 4-chlorokynurenine (4-Cl-KYN) in mice. Following administration of 4-Cl-KYN, 7-Cl-KYNA was promptly recovered extracellularly in hippocampal microdialysate of freely moving animals. The behavioral responses of the animals were assessed using measures of ketamine-sensitive antidepressant efficacy (including the 24-hour forced swim test, learned helplessness test, and novelty-suppressed feeding test). In these tests, distinct from fluoxetine, and similar to ketamine, 4-Cl-KYN administration resulted in rapid, dose-dependent and persistent antidepressant-like effects following a single treatment. The antidepressant effects of 4-Cl-KYN were prevented by pretreatment with glycine or the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX). 4-Cl-KYN administration was not associated with the rewarding and psychotomimetic effects of ketamine, and did not induce locomotor sensitization or stereotypic behaviors. Our results provide further support for antagonism of the glycineB site for the rapid treatment of treatment-resistant depression without the negative side effects seen with ketamine or other channel-blocking NMDA receptor antagonists.


Subject(s)
Glycine , Ketamine/pharmacology , Kynurenine/analogs & derivatives , Prodrugs/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Animals , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/psychology , Hindlimb Suspension , Ketamine/adverse effects , Kynurenine/adverse effects , Kynurenine/metabolism , Kynurenine/pharmacology , Kynurenine/therapeutic use , Male , Mice , Microdialysis , Prodrugs/adverse effects , Prodrugs/metabolism , Prodrugs/therapeutic use , Receptors, N-Methyl-D-Aspartate/metabolism , Swimming
2.
Schizophr Bull ; 40 Suppl 2: S152-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24562494

ABSTRACT

Increased brain levels of the tryptophan metabolite kynurenic acid (KYNA) have been linked to cognitive dysfunctions in schizophrenia and other psychiatric diseases. In the rat, local inhibition of kynurenine aminotransferase II (KAT II), the enzyme responsible for the neosynthesis of readily mobilizable KYNA in the brain, leads to a prompt reduction in extracellular KYNA levels, and secondarily induces an increase in extracellular glutamate, dopamine, and acetylcholine levels in several brain areas. Using microdialysis in unanesthetized, adult rats, we now show that the novel, systemically active KAT II inhibitor BFF-816, applied orally at 30 mg/kg in all experiments, mimics the effects of local enzyme inhibition. No tolerance was seen when animals were treated daily for 5 consecutive days. Behaviorally, daily injections of BFF-816 significantly decreased escape latency in the Morris water maze, indicating improved performance in spatial and contextual memory. Thus, systemically applied BFF-816 constitutes an excellent tool for studying the neurobiology of KYNA and, in particular, for investigating the mechanisms linking KAT II inhibition to changes in glutamatergic, dopaminergic, and cholinergic function in brain physiology and pathology.


Subject(s)
Brain/drug effects , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Thiazolidinediones/pharmacology , Transaminases/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Enzyme Inhibitors/administration & dosage , Heterocyclic Compounds, 3-Ring/administration & dosage , Male , Maze Learning/drug effects , Microdialysis , Rats , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Thiazolidinediones/administration & dosage
3.
J Biol Chem ; 288(51): 36554-66, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24189070

ABSTRACT

Kynurenine 3-monooxygenase (KMO), a pivotal enzyme in the kynurenine pathway (KP) of tryptophan degradation, has been suggested to play a major role in physiological and pathological events involving bioactive KP metabolites. To explore this role in greater detail, we generated mice with a targeted genetic disruption of Kmo and present here the first biochemical and neurochemical characterization of these mutant animals. Kmo(-/-) mice lacked KMO activity but showed no obvious abnormalities in the activity of four additional KP enzymes tested. As expected, Kmo(-/-) mice showed substantial reductions in the levels of its enzymatic product, 3-hydroxykynurenine, in liver, brain, and plasma. Compared with wild-type animals, the levels of the downstream metabolite quinolinic acid were also greatly decreased in liver and plasma of the mutant mice but surprisingly were only slightly reduced (by ∼20%) in the brain. The levels of three other KP metabolites: kynurenine, kynurenic acid, and anthranilic acid, were substantially, but differentially, elevated in the liver, brain, and plasma of Kmo(-/-) mice, whereas the liver and brain content of the major end product of the enzymatic cascade, NAD(+), did not differ between Kmo(-/-) and wild-type animals. When assessed by in vivo microdialysis, extracellular kynurenic acid levels were found to be significantly elevated in the brains of Kmo(-/-) mice. Taken together, these results provide further evidence that KMO plays a key regulatory role in the KP and indicate that Kmo(-/-) mice will be useful for studying tissue-specific functions of individual KP metabolites in health and disease.


Subject(s)
Brain/metabolism , Gene Deletion , Gene Knockout Techniques , Kynurenine 3-Monooxygenase/genetics , Kynurenine/metabolism , Animals , Kynurenine/analogs & derivatives , Kynurenine/blood , Kynurenine 3-Monooxygenase/metabolism , Liver/metabolism , Mice , Mice, Knockout , Organ Specificity , Tryptophan/metabolism
4.
Nat Neurosci ; 16(11): 1652-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24121737

ABSTRACT

In the reward circuitry of the brain, α-7-nicotinic acetylcholine receptors (α7nAChRs) modulate effects of Δ(9)-tetrahydrocannabinol (THC), marijuana's main psychoactive ingredient. Kynurenic acid (KYNA) is an endogenous negative allosteric modulator of α7nAChRs. Here we report that the kynurenine 3-monooxygenase (KMO) inhibitor Ro 61-8048 increases brain KYNA levels and attenuates cannabinoid-induced increases in extracellular dopamine in reward-related brain areas. In the self-administration model of drug abuse, Ro 61-8048 reduced the rewarding effects of THC and the synthetic cannabinoid WIN 55,212-2 in squirrel monkeys and rats, respectively, and it also prevented relapse to drug-seeking induced by reexposure to cannabinoids or cannabinoid-associated cues. The effects of enhancing endogenous KYNA levels with Ro 61-8048 were prevented by positive allosteric modulators of α7nAChRs. Despite a clear need, there are no medications approved for treatment of marijuana dependence. Modulation of KYNA offers a pharmacological strategy for achieving abstinence from marijuana and preventing relapse.


Subject(s)
Brain/metabolism , Kynurenic Acid/metabolism , Substance-Related Disorders , Analgesics/administration & dosage , Animals , Benzoxazines/administration & dosage , Cannabinoid Receptor Agonists/pharmacology , Conditioning, Operant/drug effects , Cues , Discrimination, Psychological/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Dronabinol/pharmacology , Drug-Seeking Behavior/drug effects , Male , Memory, Short-Term/drug effects , Morpholines/administration & dosage , Naphthalenes/administration & dosage , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Reinforcement, Psychology , Saimiri , Secondary Prevention , Self Administration , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism , Substance-Related Disorders/pathology , Sulfonamides/pharmacology , Thiazoles/pharmacology , Time Factors , Wakefulness
5.
Nat Rev Neurosci ; 13(7): 465-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22678511

ABSTRACT

The essential amino acid tryptophan is not only a precursor of serotonin but is also degraded to several other neuroactive compounds, including kynurenic acid, 3-hydroxykynurenine and quinolinic acid. The synthesis of these metabolites is regulated by an enzymatic cascade, known as the kynurenine pathway, that is tightly controlled by the immune system. Dysregulation of this pathway, resulting in hyper-or hypofunction of active metabolites, is associated with neurodegenerative and other neurological disorders, as well as with psychiatric diseases such as depression and schizophrenia. With recently developed pharmacological agents, it is now possible to restore metabolic equilibrium and envisage novel therapeutic interventions.


Subject(s)
Brain/pathology , Brain/physiology , Kynurenine/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Signal Transduction/physiology , Animals , Humans
6.
Eur J Neurosci ; 35(10): 1605-12, 2012 May.
Article in English | MEDLINE | ID: mdl-22515201

ABSTRACT

Levels of kynurenic acid (KYNA), an endogenous product of tryptophan degradation, are elevated in the brain and cerebrospinal fluid of individuals with schizophrenia (SZ). This increase has been implicated in the cognitive dysfunctions seen in the disease, as KYNA is an antagonist of the α7 nicotinic acetylcholine receptor and the N-methyl-d-aspartate receptor, both of which are critically involved in cognitive processes and in a defining neurodevelopmental period in the pathophysiology of SZ. We tested the hypothesis that early developmental increases in brain KYNA synthesis might cause biochemical and functional impairments in adulthood. To this end, we stimulated KYNA formation by adding the KYNA precursor kynurenine (100 mg/day) to the chow fed to rat dams from gestational day 15 to postnatal day 21 (PD 21). This treatment raised brain KYNA levels in the offspring by 341% on PD 2 and 210% on PD 21. Rats were then fed normal chow until adulthood (PD 56-80). In the adult animals, basal levels of extracellular KYNA, measured in the hippocampus by in vivo microdialysis, were elevated (+12%), whereas extracellular glutamate levels were significantly reduced (-13%). In separate adult animals, early kynurenine treatment was shown to impair performance in two behavioral tasks linked to hippocampal function, the passive avoidance test and the Morris water maze test. Collectively, these studies introduce a novel, naturalistic rat model of SZ, and also suggest that increases in brain KYNA during a vulnerable period in brain development may play a significant role in the pathophysiology of the disease.


Subject(s)
Cognition Disorders/chemically induced , Kynurenine/toxicity , Prenatal Exposure Delayed Effects/physiopathology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Chromatography, High Pressure Liquid , Cognition Disorders/physiopathology , Electrochemical Techniques , Female , Glutamic Acid/metabolism , Kynurenic Acid/metabolism , Kynurenine/administration & dosage , Kynurenine/analogs & derivatives , Kynurenine/blood , Kynurenine/metabolism , Male , Maze Learning/drug effects , Memory/drug effects , Microdialysis , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar , Spatial Behavior/drug effects
7.
J Neurochem ; 120(6): 1026-35, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22224417

ABSTRACT

In the mammalian brain, the α7 nicotinic and NMDA receptor antagonist kynurenic acid is synthesized by irreversible enzymatic transamination of the tryptophan metabolite l-kynurenine. d-kynurenine, too, serves as a bioprecursor of kynurenic acid in several organs including the brain, but the conversion is reportedly catalyzed through oxidative deamination by d-amino acid oxidase. Using brain and liver tissue homogenates from rats and humans, and conventional incubation conditions for kynurenine aminotransferases, we show here that kynurenic acid production from d-kynurenine, like the more efficient kynurenic acid synthesis from l-kynurenine, is blocked by the aminotransferase inhibitor amino-oxyacetic acid. In vivo, focal application of 100 µM d-kynurenine by reverse microdialysis led to a steady rise in extracellular kynurenic acid in the rat striatum, causing a 4-fold elevation after 2 h. Attesting to functional significance, this increase was accompanied by a 36% reduction in extracellular dopamine. Both of these effects were duplicated by perfusion of 2 µM l-kynurenine. Co-infusion of amino-oxyacetic acid (2 mM) significantly attenuated the in vivo effects of d-kynurenine and essentially eliminated the effects of l-kynurenine. Thus, enzymatic transamination accounts in part for kynurenic acid synthesis from d-kynurenine in the brain. These results are discussed with regard to implications for brain physiology and pathology.


Subject(s)
Brain/enzymology , Kynurenic Acid/metabolism , Kynurenine/metabolism , Liver/enzymology , Transaminases/metabolism , Animals , Area Under Curve , Brain/drug effects , Chromatography, High Pressure Liquid , Dopamine/metabolism , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Kynurenine/pharmacology , Liver/drug effects , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Time Factors
8.
Anal Biochem ; 421(2): 573-81, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22239963

ABSTRACT

We describe here a gas chromatography-tandem mass spectrometry (GC/MS/MS) method for the sensitive and concurrent determination of extracellular tryptophan and the kynurenine pathway metabolites kynurenine, 3-hydroxykynurenine (3-HK), and quinolinic acid (QUIN) in rat brain. This metabolic cascade is increasingly linked to the pathophysiology of several neurological and psychiatric diseases. Methodological refinements, including optimization of MS conditions and the addition of deuterated standards, resulted in assay linearity to the low nanomolar range. Measured in samples obtained by striatal microdialysis in vivo, basal levels of tryptophan, kynurenine, and QUIN were 415, 89, and 8 nM, respectively, but 3-HK levels were below the limit of detection (<2 nM). Systemic injection of kynurenine (100 mg/kg, i.p.) did not affect extracellular tryptophan but produced detectable levels of extracellular 3-HK (peak after 2-3 h: ~50 nM) and raised extracellular QUIN levels (peak after 2h: ~105 nM). The effect of this treatment on QUIN, but not on 3-HK, was potentiated in the N-methyl-D-aspartate (NMDA)-lesioned striatum. Our results indicate that the novel methodology, which allowed the measurement of extracellular kynurenine and 3-HK in the brain in vivo, will facilitate studies of brain kynurenines and of the interplay between peripheral and central kynurenine pathway functions under physiological and pathological conditions.


Subject(s)
Brain/metabolism , Gas Chromatography-Mass Spectrometry/methods , Kynurenine/metabolism , Tandem Mass Spectrometry/methods , Animals , Brain/pathology , Brain Chemistry , Kynurenine/analysis , Male , Microdialysis , Rats , Rats, Sprague-Dawley
9.
Psychopharmacology (Berl) ; 220(3): 627-37, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22038535

ABSTRACT

RATIONALE: Cognitive deficits represent a core symptom cluster in schizophrenia (SZ) that is predictive of outcome but not effectively treated by current antipsychotics. Thus, there is a need for validated animal models for testing potential pro-cognitive drugs. OBJECTIVE: As kynurenic acid levels are increased in prefrontal cortex (PFC) of individuals with SZ, we acutely increased brain levels of this astrocyte-derived, negative modulator of alpha7 nicotinic acetylcholine receptors (α7nAChRs) by administration of its bioprecursor kynurenine and measured the effects on extracellular kynurenic acid and glutamate levels in PFC and also performance in a set-shifting task. RESULTS: Injections of kynurenine (100 mg/kg, i.p.) increased extracellular kynurenic acid (1,500%) and decreased glutamate levels (30%) in PFC. Kynurenine also produced selective deficits in set-shifting. Saline- and kynurenine-treated rats similarly acquired the compound discrimination and intra-dimensional shift (saline, 7.0 and 6.3 trials, respectively; kynurenine, 8.0 and 6.7). Both groups required more trials to acquire the initial reversal (saline, 15.3; kynurenine, 22.2). Only kynurenine-treated rats were impaired in acquiring the extra-dimensional shift (saline, 8.2; kynurenine, 21.3). These deficits were normalized by administering the α7nAChR positive allosteric modulator galantamine (3.0 mg/kg, i.p) prior to kynurenine, as trials were comparable between galantamine + kynurenine (7.8) and controls (8.2). Bilateral local perfusion of the PFC with galantamine (5.0 µM) also attenuated kynurenine-induced deficits. CONCLUSIONS: These results validate the use of animals with elevated brain kynurenic acid levels in SZ research and support studies of drugs that normalize brain kynurenic acid levels and/or positively modulate α7nAChRs as pro-cognitive treatments for SZ.


Subject(s)
Cognition/drug effects , Galantamine/pharmacology , Kynurenic Acid/metabolism , Kynurenine/toxicity , Animals , Brain/drug effects , Brain/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Disease Models, Animal , Glutamic Acid/metabolism , Male , Nootropic Agents/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Schizophrenia/drug therapy , Schizophrenia/physiopathology , alpha7 Nicotinic Acetylcholine Receptor
10.
Neuropsychopharmacology ; 36(11): 2357-67, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21796108

ABSTRACT

Kynurenic acid (KYNA), an astrocyte-derived metabolite, antagonizes the α7 nicotinic acetylcholine receptor (α7nAChR) and, possibly, the glycine co-agonist site of the NMDA receptor at endogenous brain concentrations. As both receptors are involved in cognitive processes, KYNA elevations may aggravate, whereas reductions may improve, cognitive functions. We tested this hypothesis in rats by examining the effects of acute up- or downregulation of endogenous KYNA on extracellular glutamate in the hippocampus and on performance in the Morris water maze (MWM). Applied directly by reverse dialysis, KYNA (30-300 nM) reduced, whereas the specific kynurenine aminotransferase-II inhibitor (S)-4-(ethylsulfonyl)benzoylalanine (ESBA; 0.3-3 mM) raised, extracellular glutamate levels in the hippocampus. Co-application of KYNA (100 nM) with ESBA (1 mM) prevented the ESBA-induced glutamate increase. Comparable effects on hippocampal glutamate levels were seen after intra-cerebroventricular (i.c.v.) application of the KYNA precursor kynurenine (1 mM, 10 µl) or ESBA (10 mM, 10 µl), respectively. In separate animals, i.c.v. treatment with kynurenine impaired, whereas i.c.v. ESBA improved, performance in the MWM. I.c.v. co-application of KYNA (10 µM) eliminated the pro-cognitive effects of ESBA. Collectively, these studies show that KYNA serves as an endogenous modulator of extracellular glutamate in the hippocampus and regulates hippocampus-related cognitive function. Our results suggest that pharmacological interventions leading to acute reductions in hippocampal KYNA constitute an effective strategy for cognitive improvement. This approach might be especially useful in the treatment of cognitive deficits in neurological and psychiatric diseases that are associated with increased brain KYNA levels.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/metabolism , Kynurenic Acid/metabolism , Memory/physiology , Animals , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Hippocampus/drug effects , Kynurenic Acid/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Rats , Rats, Sprague-Dawley
11.
Cell ; 145(6): 863-74, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21640374

ABSTRACT

Metabolites in the kynurenine pathway, generated by tryptophan degradation, are thought to play an important role in neurodegenerative disorders, including Alzheimer's and Huntington's diseases. In these disorders, glutamate receptor-mediated excitotoxicity and free radical formation have been correlated with decreased levels of the neuroprotective metabolite kynurenic acid. Here, we describe the synthesis and characterization of JM6, a small-molecule prodrug inhibitor of kynurenine 3-monooxygenase (KMO). Chronic oral administration of JM6 inhibits KMO in the blood, increasing kynurenic acid levels and reducing extracellular glutamate in the brain. In a transgenic mouse model of Alzheimer's disease, JM6 prevents spatial memory deficits, anxiety-related behavior, and synaptic loss. JM6 also extends life span, prevents synaptic loss, and decreases microglial activation in a mouse model of Huntington's disease. These findings support a critical link between tryptophan metabolism in the blood and neurodegeneration, and they provide a foundation for treatment of neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Huntington Disease/drug therapy , Kynurenic Acid/analysis , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Sulfonamides/therapeutic use , Thiazoles/therapeutic use , Administration, Oral , Alzheimer Disease/physiopathology , Animals , Brain Chemistry , Disease Models, Animal , Female , Humans , Kynurenic Acid/blood , Male , Mice , Mice, Transgenic , Sulfonamides/administration & dosage , Thiazoles/administration & dosage
12.
Neuropsychopharmacology ; 35(8): 1734-42, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20336058

ABSTRACT

At endogenous brain concentrations, the astrocyte-derived metabolite kynurenic acid (KYNA) antagonizes the alpha 7 nicotinic acetylcholine receptor and, possibly, the glycine co-agonist site of the NMDA receptor. The functions of these two receptors, which are intimately involved in synaptic plasticity and cognitive processes, may, therefore, be enhanced by reductions in brain KYNA levels. This concept was tested in mice with a targeted deletion of kynurenine aminotransferase II (KAT II), a major biosynthetic enzyme of brain KYNA. At 21 days of age, KAT II knock-out mice had reduced hippocampal KYNA levels (-71%) and showed significantly increased performance in three cognitive paradigms that rely in part on the integrity of hippocampal function, namely object exploration and recognition, passive avoidance, and spatial discrimination. Moreover, compared with wild-type controls, hippocampal slices from KAT II-deficient mice showed a significant increase in the amplitude of long-term potentiation in vitro. These functional changes were accompanied by reduced extracellular KYNA (-66%) and increased extracellular glutamate (+51%) concentrations, measured by hippocampal microdialysis in vivo. Taken together, a picture emerges in which a reduction in the astrocytic formation of KYNA increases glutamatergic tone in the hippocampus and enhances cognitive abilities and synaptic plasticity. Our studies raise the prospect that interventions aimed specifically at reducing KYNA formation in the brain may constitute a promising molecular strategy for cognitive improvement in health and disease.


Subject(s)
Cognition/physiology , Glutamic Acid/metabolism , Hippocampus/physiology , Kynurenic Acid/metabolism , Long-Term Potentiation/physiology , Animals , Animals, Newborn , Avoidance Learning/physiology , Discrimination, Psychological/physiology , Exploratory Behavior/physiology , Extracellular Fluid/metabolism , Hippocampus/cytology , In Vitro Techniques , Long-Term Potentiation/genetics , Maze Learning/physiology , Mice , Mice, Knockout , Microdialysis/methods , Neuropsychological Tests , Patch-Clamp Techniques , Recognition, Psychology/physiology , Transaminases/deficiency
13.
J Mol Neurosci ; 40(1-2): 204-10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19690987

ABSTRACT

The cognitive deficits seen in schizophrenia patients are likely related to abnormal glutamatergic and cholinergic neurotransmission in the prefrontal cortex. We hypothesized that these impairments may be secondary to increased levels of the astrocyte-derived metabolite kynurenic acid (KYNA), which inhibits alpha7 nicotinic acetylcholine receptors (alpha7AChR) and may thereby reduce glutamate release. Using in vivo microdialysis in unanesthetized rats, we show here that nanomolar concentrations of KYNA, infused directly or produced in situ from its bioprecursor kynurenine, significantly decrease extracellular glutamate levels in the prefrontal cortex. This effect was prevented by the systemic administration of galantamine (3 mg/kg) but not by donepezil (2 mg/kg), indicating that KYNA blocks the allosteric potentiating site of the alpha7AChR, which recognizes galantamine but not donepezil as an agonist. In separate rats, reduction of prefrontal KYNA formation by (S)-4-ethylsulfonyl benzoylalanine, a specific inhibitor of KYNA synthesis, caused a significant elevation in extracellular glutamate levels. Jointly, our results demonstrate that fluctuations in endogenous KYNA formation bidirectionally influence cortical glutamate concentrations. These findings suggest that selective attenuation of cerebral KYNA production, by increasing glutamatergic tone, might improve cognitive function in individuals with schizophrenia.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Kynurenic Acid/metabolism , Nicotinic Antagonists/metabolism , Prefrontal Cortex/metabolism , Animals , Cholinesterase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Galantamine/pharmacology , Kynurenic Acid/pharmacology , Kynurenine/metabolism , Male , Microdialysis , Nicotinic Antagonists/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Schizophrenia/physiopathology , alpha7 Nicotinic Acetylcholine Receptor
14.
Neuron ; 57(2): 290-302, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18215625

ABSTRACT

Place-specific firing in the hippocampus is determined by path integration-based spatial representations in the grid-cell network of the medial entorhinal cortex. Output from this network is conveyed directly to CA1 of the hippocampus by projections from principal neurons in layer III, but also indirectly by axons from layer II to the dentate gyrus and CA3. The direct pathway is sufficient for spatial firing in CA1, but it is not known whether similar firing can also be supported by the input from CA3. To test this possibility, we made selective lesions in layer III of medial entorhinal cortex by local infusion of the neurotoxin gamma-acetylenic GABA. Firing fields in CA1 became larger and more dispersed after cell loss in layer III, whereas CA3 cells, which receive layer II input, still had sharp firing fields. Thus, the direct projection is necessary for precise spatial firing in the CA1 place cell population.


Subject(s)
Brain Injuries/pathology , Entorhinal Cortex/pathology , Hippocampus/pathology , Hippocampus/physiopathology , Neurons/physiology , Spatial Behavior/physiology , Action Potentials/physiology , Afferent Pathways/injuries , Afferent Pathways/physiopathology , Alkynes/pharmacology , Aminocaproates/pharmacology , Animals , Behavior, Animal , Brain Injuries/chemically induced , Brain Mapping , Entorhinal Cortex/injuries , Fluoresceins , Male , Neuroglia/pathology , Neuroglia/ultrastructure , Neurons/ultrastructure , Neurotoxins/pharmacology , Organic Chemicals/metabolism , Phytohemagglutinins/metabolism , Rats , Rats, Sprague-Dawley , Silver Staining/methods , Stilbamidines/metabolism , Vimentin/metabolism
15.
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
17.
Epilepsia ; 46(7): 1010-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16026552

ABSTRACT

PURPOSE: The tryptophan metabolite kynurenic acid (KYNA) and its synthetic derivative, 7-chlorokynurenic acid (7-Cl-KYNA), are antagonists of the glycine co-agonist ("glycine(B)") site of the N-methyl-D-aspartate (NMDA)-receptor. Both compounds have neuroprotective and anticonvulsive properties but do not readily penetrate the blood-brain barrier. However, KYNA and 7-Cl-KYNA can be formed in, and released from, astrocytes after the peripheral administration of their transportable precursors kynurenine and 4-chlorokynurenine, respectively. The present study was designed to examine these biosynthetic processes, as well as astrogliosis, in animals with spontaneously recurring seizures. METHODS: The fate and formation of KYNA and 7-Cl-KYNA was studied in vivo (microdialysis) and in vitro (tissue slices) in rats exhibiting chronic seizure activity (pilocarpine model) and in appropriate controls. Neuronal loss and gliosis in these animals were examined immunohistochemically. RESULTS: In vivo microdialysis revealed higher ambient extracellular KYNA levels and enhanced de novo formation of 7-Cl-KYNA in the entorhinal cortex and hippocampus in epileptic rats. Complementary studies in tissue slices showed increased neosynthesis of KYNA and 7-Cl-KYNA in the same two brain areas. Microscopic analysis revealed pronounced astrocytic reactions in entorhinal cortex and hippocampus in epileptic animals. CONCLUSIONS: These results demonstrate that the epileptic brain can synthesize glycine(B) receptor antagonists in situ. Astrogliosis probably accounts for their enhanced production in chronically epileptic rats. These results bode well for the use of 4-chlorokynurenine in the treatment of chronic seizure disorders.


Subject(s)
Epilepsy/metabolism , Excitatory Amino Acid Antagonists/metabolism , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Disease Models, Animal , Entorhinal Cortex/metabolism , Epilepsy/chemically induced , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Gliosis/metabolism , Hippocampus/metabolism , In Vitro Techniques , Kynurenic Acid/pharmacology , Microdialysis , Rats , Rats, Sprague-Dawley , Receptors, Glycine/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects
18.
J Neurochem ; 93(3): 762-5, 2005 May.
Article in English | MEDLINE | ID: mdl-15836634

ABSTRACT

Precise regulation of dopaminergic activity is of obvious importance for the physiology and pathology of basal ganglia. We report here that nanomolar concentrations of the astrocyte-derived neuroinhibitory metabolite kynurenic acid (KYNA) potently reduce the extracellular levels of striatal dopamine in unanesthetized rats in vivo. This effect, which is initiated by the KYNA-induced blockade of alpha7 nicotinic acetylcholine receptors, highlights the functional relevance of glia-neuron interactions in the striatum and indicates that even modest increases in the brain levels of endogenous KYNA are capable of interfering with dopaminergic neurotransmission.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Extracellular Fluid/drug effects , Kynurenic Acid/administration & dosage , Nanotechnology/methods , Animals , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Extracellular Fluid/metabolism , Male , Rats , Rats, Sprague-Dawley
19.
Neuropsychopharmacology ; 30(4): 697-704, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15496939

ABSTRACT

The content of the endogenous NMDA and alpha7 nicotinic acetylcholine receptor antagonist kynurenate (KYNA) is increased in the cerebral cortex and cerebrospinal fluid of patients with schizophrenia. In view of the very high incidence of smoking in schizophrenic individuals, a study was designed to examine the effect of acute and prolonged nicotine administration on brain KYNA levels in experimental animals. Adult male rats received subcutaneous nicotine injections twice daily for up to 10 days, and animals were routinely killed 1 h after the last injection. Neither acute treatment nor a 2-day regimen with 1 mg/kg nicotine (= 0.35 mg/kg pure base) caused changes in cerebral KYNA levels. Four- or 6 day-treatment with this dose resulted in 20-40% decreases in cerebral KYNA content. Animals treated with 1 or 10 mg/kg nicotine for 10 days showed dose-dependent, significant increases in KYNA in hippocampus, striatum, and cortex, but not in the serum. Discontinuation of nicotine treatment for 7 days restored brain KYNA to control levels. Separate animals, implanted with osmotic minipumps delivering 2 mg/kg of nicotine/day for 10 days also showed significant elevations in brain KYNA. Hippocampal microdialysis, performed in animals receiving nicotine (1 mg/kg) for 10 days, revealed a significant increase in basal extracellular KYNA levels compared to controls, whereas acute treatment with this dose produced no such change. Measurements of KYNA's bioprecursor kynurenine in brain or blood did not reveal any nicotine-induced changes. These results indicate that nicotine has a brain-specific, biphasic effect on the transamination of kynurenine to KYNA. Such nicotine-induced fluctuations in brain KYNA may cause functional changes in processes that regulate glutamatergic and cholinergic neurotransmission in the normal and diseased brain.


Subject(s)
Brain Chemistry/drug effects , Brain/drug effects , Kynurenic Acid/metabolism , Nicotine/pharmacology , Tobacco Use Disorder/etiology , Acetylcholine/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Brain Chemistry/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Administration Schedule , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Kynurenine/metabolism , Male , Rats , Rats, Sprague-Dawley , Smoking/metabolism , Tobacco Use Disorder/metabolism , Tobacco Use Disorder/physiopathology , Up-Regulation/drug effects , Up-Regulation/physiology
20.
J Neurochem ; 90(3): 621-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15255939

ABSTRACT

The naturally occurring purine nucleoside adenosine has pronounced anticonvulsant and neuroprotective properties and plays a neuromodulatory role in the CNS. Kynurenic acid (KYNA) is an astrocyte-derived, endogenous neuroinhibitory compound, which shares several of adenosine's properties. In a first attempt to examine possible interactions between these two biologically active molecules, adenosine was focally applied into the striatum of freely moving rats by reverse microdialysis, and changes in extracellular KYNA were monitored over time. A 2-h infusion of adenosine increased KYNA levels in a dose-dependent manner, with 10 mm of adenosine causing a twofold elevation within 1 h. This effect was reversible and was effectively blocked by coinfusion of the specific A1 adenosine receptor antagonist 8-cyclopentyltheophylline (100 microm). In contrast, coinfusion of adenosine with MSX-3 (100 microm), an A2A receptor antagonist, did not affect the adenosine-induced increase in KYNA levels. Local striatal perfusion with the A1 receptor agonist N6-cyclopentyladenosine (100 microm) mimicked the effect of adenosine, whereas perfusion with the A2A receptor agonist CGS-21680 (100 microm) was ineffective. Finally, we tested the effect of adenosine (10 mm) on extracellular KYNA in striata that had been injected with quinolinate (60 nmol/1 microL) 7 days earlier. In this neuron-depleted tissue, perfusion with adenosine failed to affect extracellular KYNA levels. These data demonstrate that adenosine is capable of raising extracellular KYNA in the rat striatum by interacting with postsynaptic neuronal A1 receptors. This mechanism may result in a synergism between the neurobiological effects of adenosine and KYNA.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/metabolism , Kynurenic Acid/metabolism , Neostriatum/metabolism , Neurons/metabolism , Receptor, Adenosine A1/metabolism , Theophylline/analogs & derivatives , Adenosine/administration & dosage , Adenosine/pharmacology , Animals , Drug Administration Routes , Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Infusions, Parenteral , Kynurenic Acid/analysis , Male , Microdialysis , Neostriatum/cytology , Neostriatum/drug effects , Neurons/drug effects , Phenethylamines/pharmacology , Quinolinic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/drug effects , Theophylline/pharmacology , Wakefulness
SELECTION OF CITATIONS
SEARCH DETAIL
...