Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
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
2.
Brain ; 139(Pt 7): 2063-81, 2016 07.
Article in English | MEDLINE | ID: mdl-27190010

ABSTRACT

Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-ß accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-ß oligomers.


Subject(s)
Alzheimer Disease/prevention & control , Gene Expression/drug effects , Hippocampus/drug effects , Memory Disorders/prevention & control , Neuronal Plasticity/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Animals , Behavior, Animal/drug effects , Cyclooxygenase Inhibitors , Disease Models, Animal , Down-Regulation , Electroencephalography , Ibuprofen , Male , Mice , Mice, Inbred C57BL , Rats , Recognition, Psychology/drug effects , Tryptophan Oxygenase/drug effects
3.
Neuroscience ; 409: 169-179, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31029729

ABSTRACT

Parkinson's disease (PD) is the most common neurodegenerative movement disorder and is characterized by the loss of neurons in the substantia nigra that project to the striatum and release dopamine (DA), which is required for normal movement. Common non-motor symptoms likely involve abnormalities with other neurotransmitters, such as serotonin, norepinephrine, acetylcholine, glycine, glutamate and gamma-aminobutyric acid (GABA). As part of a broad effort to provide better PD research tools, the Michael J. Fox Foundation for Parkinson's Research funded the generation and characterization of knockout (KO) rats for genes with PD-linked mutations, including PINK1, Parkin, DJ-1 and LRRK2. Here we extend the phenotypic characterization of these lines of KO rats to include in vivo microdialysis to measure both basal and potassium-induced release of the above neurotransmitters and their metabolites in the striatum of awake and freely moving rats at ages 4, 8 and 12 months compared to wild-type (WT) rats. We found age-dependent abnormalities in basal DA, glutamate and acetylcholine in PINK1 KO rats and age-dependent abnormalities in basal DA metabolites in Parkin and LRRK2 KO rats. Parkin KO rats had increased glycine release while DJ-1 KO rats had decreased glutamate release and increased acetylcholine release compared to WT rats. All lines except DJ-1 KO rats showed age-dependent changes in release of one or more neurotransmitters. Our data suggest these rats may be useful for studies of PD-related synaptic dysfunction and neurotransmitter dynamics as well as studies of the normal and pathogenic functions of these genes with PD-linked mutations.


Subject(s)
Acetylcholine/metabolism , Brain/metabolism , Dopamine/metabolism , Glutamic Acid/metabolism , Parkinson Disease/metabolism , Animals , Dopaminergic Neurons/metabolism , Gene Knockout Techniques , Glycine/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Male , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Rats , Serotonin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Behav Brain Res ; 186(2): 161-7, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-17868931

ABSTRACT

Increased glutamatergic activity is believed to play a significant role in the development of levodopa-induced dyskinesias (LID). LID may therefore be attenuated by a reduction in glutamatergic function. This was tested pharmacologically in MPTP monkeys by increasing the formation of kynurenic acid (KYNA), a tryptophan metabolite that inhibits glutamate release and also blocks NMDA receptors directly. KYNA synthesis was stimulated by prolonged systemic administration of the kynurenine 3-hydroxylase inhibitor Ro 61-8048. Four MPTP cynomolgus monkeys received l-dopa (LD; 100mg) with benserazide (25 mg) for one month. Progressively, all these animals developed LID. Four other MPTP monkeys received Ro 61-8048 (50mg/kg) daily 3 h before administration of LD/benserazide for one month. The addition of Ro 61-8048 reduced the development of LID but did not affect the antiparkinsonian efficacy of LD. Moreover, Ro 61-8048 administration caused sustained increases in serum kynurenine and KYNA concentrations, which reverted to basal values 24 h after the last treatment. This effect of Ro 61-8048 was less pronounced in the CSF. These results demonstrate that long-lasting elevation of KYNA levels caused by prolonged inhibition of kynurenine 3-hydroxylase is associated with a significant reduction in LID but does not compromise the benefits of chronic LD therapy.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/enzymology , Dyskinesia, Drug-Induced/etiology , Kynurenine 3-Monooxygenase/metabolism , Levodopa/adverse effects , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Drug Interactions , Dyskinesia, Drug-Induced/prevention & control , Enzyme Inhibitors/therapeutic use , Female , Kynurenic Acid/blood , Kynurenine/blood , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Macaca fascicularis , Motor Activity/drug effects , Parkinsonian Disorders/drug therapy , Quinolinic Acid/blood , Quinolinic Acid/cerebrospinal fluid , Sulfonamides/therapeutic use , Thiazoles/therapeutic use , Time Factors
5.
Nat Neurosci ; 21(7): 952-962, 2018 07.
Article in English | MEDLINE | ID: mdl-29950668

ABSTRACT

Functional neuroanatomy of Pavlovian fear has identified neuronal circuits and synapses associating conditioned stimuli with aversive events. Hebbian plasticity within these networks requires additional reinforcement to store particularly salient experiences into long-term memory. Here we have identified a circuit that reciprocally connects the ventral periaqueductal gray and dorsal raphe region with the central amygdala and that gates fear learning. We found that ventral periaqueductal gray and dorsal raphe dopaminergic (vPdRD) neurons encode a positive prediction error in response to unpredicted shocks and may reshape intra-amygdala connectivity via a dopamine-dependent form of long-term potentiation. Negative feedback from the central amygdala to vPdRD neurons might limit reinforcement to events that have not been predicted. These findings add a new module to the midbrain dopaminergic circuit architecture underlying associative reinforcement learning and identify vPdRD neurons as a critical component of Pavlovian fear conditioning. We propose that dysregulation of vPdRD neuronal activity may contribute to fear-related psychiatric disorders.


Subject(s)
Association Learning/physiology , Dopaminergic Neurons/physiology , Fear/physiology , Tegmentum Mesencephali/physiology , Animals , Behavior, Animal/physiology , Conditioning, Classical/physiology , Long-Term Potentiation/physiology , Male , Mice , Neural Pathways/physiology , Periaqueductal Gray/cytology , Periaqueductal Gray/physiology , Tegmentum Mesencephali/cytology
6.
Matters (Zur) ; 20172017.
Article in English | MEDLINE | ID: mdl-28894740

ABSTRACT

Mobility and locomotor impairments have high prevalence, morbidity, and significant mortality in older adult populations. Cerebellar functional changes have been implicated in the pathogenesis of these age-related mobility and gait deficits unrelated to stroke, Parkinson's disease, or degenerative joint disease. We thus examined total cerebellar glutamate, glutamine, GABA, glycine, dopamine, norepinephrine, tryptophan, serotonin, alanine, threonine, and asparagine content from male 2-3-month (young, n = 6) and 21-24-month-old (aged, n = 6) C57BL/6 mice. Neurotransmitter and amino acid concentrations were determined by high-performance liquid chromatography followed with mass spectroscopy. We found a significant increase in cerebellar serotonin in aged versus young mice, but otherwise no significant phenotypic differences in measured neurotransmitter concentrations. Applying current thought about cerebellar aging and cerebellar serotonergic systems, we consider how this age-related increase in cerebellar serotonin may contribute to gait ataxia.

7.
Exp Neurol ; 282: 99-118, 2016 08.
Article in English | MEDLINE | ID: mdl-27163548

ABSTRACT

Dysregulation of the kynurenine (Kyn) pathway has been associated with the progression of Huntington's disease (HD). In particular, elevated levels of the kynurenine metabolites 3-hydroxy kynurenine (3-OH-Kyn) and quinolinic acid (Quin), have been reported in the brains of HD patients as well as in rodent models of HD. The production of these metabolites is controlled by the activity of kynurenine mono-oxygenase (KMO), an enzyme which catalyzes the synthesis of 3-OH-Kyn from Kyn. In order to determine the role of KMO in the phenotype of mouse models of HD, we have developed a potent and selective KMO inhibitor termed CHDI-340246. We show that this compound, when administered orally to transgenic mouse models of HD, potently and dose-dependently modulates the Kyn pathway in peripheral tissues and in the central nervous system. The administration of CHDI-340246 leads to an inhibition of the formation of 3-OH-Kyn and Quin, and to an elevation of Kyn and Kynurenic acid (KynA) levels in brain tissues. We show that administration of CHDI-340246 or of Kyn and of KynA can restore several electrophysiological alterations in mouse models of HD, both acutely and after chronic administration. However, using a comprehensive panel of behavioral tests, we demonstrate that the chronic dosing of a selective KMO inhibitor does not significantly modify behavioral phenotypes or natural progression in mouse models of HD.


Subject(s)
Electrophysiological Phenomena/drug effects , Enzyme Inhibitors/therapeutic use , Huntington Disease/drug therapy , Huntington Disease/physiopathology , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Pyrimidines/therapeutic use , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiological Phenomena/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hippocampus/drug effects , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , In Vitro Techniques , Kynurenic Acid/metabolism , Kynurenine 3-Monooxygenase/metabolism , Male , Mice , Mice, Transgenic , Microdialysis , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Quinolinic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Trinucleotide Repeats/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
8.
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
9.
J Med Chem ; 58(3): 1159-83, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25590515

ABSTRACT

We report on the development of a series of pyrimidine carboxylic acids that are potent and selective inhibitors of kynurenine monooxygenase and competitive for kynurenine. We describe the SAR for this novel series and report on their inhibition of KMO activity in biochemical and cellular assays and their selectivity against other kynurenine pathway enzymes. We describe the optimization process that led to the identification of a program lead compound with a suitable ADME/PK profile for therapeutic development. We demonstrate that systemic inhibition of KMO in vivo with this lead compound provides pharmacodynamic evidence for modulation of kynurenine pathway metabolites both in the periphery and in the central nervous system.


Subject(s)
Enzyme Inhibitors/pharmacology , Huntington Disease/drug therapy , Kynurenine 3-Monooxygenase/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , CHO Cells , Cell Proliferation/drug effects , Cricetulus , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Huntington Disease/metabolism , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/metabolism , Mice , Models, Molecular , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
10.
PLoS Curr ; 62014 Feb 13.
Article in English | MEDLINE | ID: mdl-24558637

ABSTRACT

Huntington's disease is a neurodegenerative disorder caused by mutations in the CAG tract of huntingtin. Several studies in HD cellular and rodent systems have identified disturbances in cyclic nucleotide signaling, which might be relevant to pathogenesis and therapeutic intervention. To investigate whether selective phosphodiesterase (PDE) inhibitors can improve some aspects of disease pathogenesis in HD models, we have systematically evaluated the effects of a variety of cAMP and cGMP selective PDE inhibitors in various HD models. Here we present the lack of effect in a variety of endpoints of the PDE subtype selective inhibitor SCH-51866, a PDE1/5 inhibitor, in the R6/2 mouse model of HD, after chronic oral dosing.

11.
Schizophr Bull ; 37(6): 1147-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21036897

ABSTRACT

The levels of kynurenic acid (KYNA), an astrocyte-derived metabolite of the branched kynurenine pathway (KP) of tryptophan degradation and antagonist of α7 nicotinic acetylcholine and N-methyl-D-aspartate receptors, are elevated in the prefrontal cortex (PFC) of individuals with schizophrenia (SZ). Because endogenous KYNA modulates extracellular glutamate and acetylcholine levels in the PFC, these increases may be pathophysiologically significant. Using brain tissue from SZ patients and matched controls, we now measured the activity of several KP enzymes (kynurenine 3-monooxygenase [KMO], kynureninase, 3-hydroxyanthranilic acid dioxygenase [3-HAO], quinolinic acid phosphoribosyltransferase [QPRT], and kynurenine aminotransferase II [KAT II]) in the PFC, ie, Brodmann areas (BA) 9 and 10. Compared with controls, the activities of KMO (in BA 9 and 10) and 3-HAO (in BA 9) were significantly reduced in SZ, though there were no significant differences between patients and controls in kynureninase, QPRT, and KAT II. In the same samples, we also confirmed the increase in the tissue levels of KYNA in SZ. As examined in rats treated chronically with the antipsychotic drug risperidone, the observed biochemical changes were not secondary to medication. A persistent reduction in KMO activity may have a particular bearing on pathology because it may signify a shift of KP metabolism toward enhanced KYNA synthesis. The present results further support the hypothesis that the normalization of cortical KP metabolism may constitute an effective new treatment strategy in SZ.


Subject(s)
Kynurenine/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/metabolism , Adult , Aged , Animals , Antipsychotic Agents/pharmacology , Case-Control Studies , Female , Humans , Kynurenine 3-Monooxygenase/metabolism , Male , Metabolic Networks and Pathways/drug effects , Middle Aged , Oxidoreductases/metabolism , Pentosyltransferases/metabolism , Postmortem Changes , Rats , Rats, Sprague-Dawley , Risperidone/pharmacology , Transaminases/metabolism
12.
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
13.
Mov Disord ; 20(7): 792-802, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15954116

ABSTRACT

Homeostatic interactions between dopamine and glutamate are central to the normal physiology of the basal ganglia. This relationship is altered in Parkinsonism and in levodopa-induced dyskinesias (LID), resulting in an upregulation of corticostriatal glutamatergic function. Kynurenic acid (KYNA), a tryptophan metabolite with antagonist activity at ionotropic glutamate receptors and the capability to inhibit glutamate release presynaptically, might therefore be of therapeutic value in LID. To evaluate this hypothesis, we used a pharmacological tool, the kynurenine 3-hydroxylase inhibitor Ro 61-8048, which raises KYNA levels acutely. Ro 61-8048 was tested in MPTP cynomolgus monkeys with a stable parkinsonian syndrome and reproducible dyskinesias after each dose of levodopa. Serum and CSF concentrations of KYNA and its precursor kynurenine increased dose-dependently after Ro 61-8048 administration, alone or in combination with levodopa. Coadministration of Ro 61-8048 with levodopa produced a moderate but significant reduction in the severity of dyskinesias while maintaining the motor benefit. These results suggest that elevation of KYNA levels through inhibition of kynurenine 3-hydroxylase constitutes a promising novel approach for managing LID in Parkinson's disease.


Subject(s)
Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , Levodopa/therapeutic use , Mixed Function Oxygenases/metabolism , Parkinsonian Disorders/drug therapy , Analysis of Variance , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Kynurenic Acid/metabolism , Kynurenine 3-Monooxygenase , Macaca fascicularis , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/blood , Motor Activity/drug effects , Quinolinic Acid/blood , Quinolinic Acid/cerebrospinal fluid , Sulfonamides/blood , Sulfonamides/cerebrospinal fluid , Sulfonamides/pharmacology , Thiazoles/blood , Thiazoles/cerebrospinal fluid , Thiazoles/pharmacology , Time Factors
14.
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
SELECTION OF CITATIONS
SEARCH DETAIL