Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 453
1.
Brain Res ; 1771: 147660, 2021 11 15.
Article En | MEDLINE | ID: mdl-34529964

Since we found that inhibition of cyclooxygenase-2 (COX-2) with concomitant application of a metabotropic glutamate receptor subtype 5 (mGluR5) antagonist (MTEP) down-regulates mGluR7 in the hippocampus (HC) and changes behavior of mice, our team decided to investigate the mechanism responsible for the observed changes. The amino acid glutamate (Glu) is a major excitatory neurotransmitter in the brain. Glu uptake is regulated by excitatory amino acid transporters (EAAT). There are five transporters with documented expression in neurons and glia in the central nervous system (CNS). EAATs, maintain the correct transmission of the Glu signal and prevent its toxic accumulation by removing Glu from the synapse. It has been documented that the toxic level of Glu is one of the main causes of mental and cognitive abnormalities. Given the above mechanisms involved in the functioning of the Glu synapse, we hypothesized modification of Glu uptake, involving EAATs as the cause of the observed changes. This study investigated the level of selected EAATs in the HC after chronic treatment with mGluR5 antagonist MTEP, NS398, and their combination using Western blot. Concomitant MTEP treatment with NS398 or a single administration of the above causes changes in LTP and modulation of EAAT levels in mouse HC. As EAATs are cellular markers of oxidative stress mechanisms, the E. coli lipopolysaccharide (LPS) challenge was performed. The modified Barnes maze test (MBM) revealed alterations in the mouse spatial learning abilities. This study reports an interaction between the mGluR5 and COX-2 in the HC, with EAAT1 and EAAT3 involvement.


Cyclooxygenase 2/physiology , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 3/biosynthesis , Hippocampus/metabolism , Oxidative Stress , Receptors, Kainic Acid/physiology , Animals , Cyclooxygenase 2 Inhibitors/pharmacology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 3/genetics , Lipopolysaccharides/pharmacology , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Receptors, Kainic Acid/antagonists & inhibitors , Spatial Learning/drug effects
2.
Neuropharmacology ; 199: 108783, 2021 11 01.
Article En | MEDLINE | ID: mdl-34509497

Alcohol use disorder (AUD) is a neuropsychiatric condition affecting millions of people worldwide. Topiramate (TPM) is an antiepileptic drug that has been shown to reduce ethanol drinking in humans. However, TPM is associated with a variety of adverse effects due to its interaction with many receptor systems and intracellular pathways. GluK1-containing kainate receptors (GluK1*KARs) are non-selectively inhibited by TPM, and genetic association studies suggest that this receptor system could be targeted to reduce drinking in AUD patients. We examined the efficacy of LY466195, a selective inhibitor of GluK1*KAR, in reducing ethanol consumption in the intermittent two-bottle choice paradigm in mice. The effect of LY466195 on various ethanol-related phenotypes was investigated by quantification of alcohol intake, physical signs of withdrawal, conditioned place preference (CPP) and in vivo microdialysis in the nucleus accumbens. Selective GluK1*KAR inhibition reduced ethanol intake and preference in a dose-dependent manner. LY466195 treatment attenuated the physical manifestations of ethanol withdrawal and influenced the rewarding properties of ethanol. Interestingly, LY466195 injection also normalized changes in dopamine levels in response to acute ethanol in ethanol-dependent mice, but had no effect in ethanol-naïve mice, suggesting ethanol state-dependent effects. The data point to GluK1*KARs as an attractive pharmacological target for the treatment of AUD.


Alcohol Drinking/drug therapy , Alcoholism/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Isoquinolines/pharmacology , Receptors, Kainic Acid/antagonists & inhibitors , Reward , Substance Withdrawal Syndrome/drug therapy , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Isoquinolines/administration & dosage , Mice , Receptors, Ionotropic Glutamate
3.
Cell Death Dis ; 11(11): 963, 2020 11 10.
Article En | MEDLINE | ID: mdl-33173027

Mutations in the PARK2 gene encoding the protein parkin cause autosomal recessive juvenile Parkinsonism (ARJP), a neurodegenerative disease characterized by dysfunction and death of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Since a neuroprotective therapy for ARJP does not exist, research efforts aimed at discovering targets for neuroprotection are critically needed. A previous study demonstrated that loss of parkin function or expression of parkin mutants associated with ARJP causes an accumulation of glutamate kainate receptors (KARs) in human brain tissues and an increase of KAR-mediated currents in neurons in vitro. Based on the hypothesis that such KAR hyperactivation may contribute to the death of nigral DA neurons, we investigated the effect of KAR antagonism on the DA neuron dysfunction and death that occur in the parkinQ311X mouse, a model of human parkin-induced toxicity. We found that early accumulation of KARs occurs in the DA neurons of the parkinQ311X mouse, and that chronic administration of the KAR antagonist UBP310 prevents DA neuron loss. This neuroprotective effect is associated with the rescue of the abnormal firing rate of nigral DA neurons and downregulation of GluK2, the key KAR subunit. This study provides novel evidence of a causal role of glutamate KARs in the DA neuron dysfunction and loss occurring in a mouse model of human parkin-induced toxicity. Our results support KAR as a potential target in the development of neuroprotective therapy for ARJP.


Alanine/analogs & derivatives , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Thymine/analogs & derivatives , Alanine/pharmacology , Animals , Disease Models, Animal , Dopaminergic Neurons/pathology , Down-Regulation , Female , Male , Mice , Mice, Inbred C57BL , Mutation , Parkinson Disease/genetics , Parkinson Disease/pathology , Receptors, Kainic Acid/metabolism , Thymine/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , GluK2 Kainate Receptor
4.
Epilepsy Res ; 167: 106452, 2020 11.
Article En | MEDLINE | ID: mdl-32911258

OBJECTIVE: The purpose of the current analysis was to investigate the direct inhibitory effects of perampanel and other anti-seizure medications (ASMs) on the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartic acid (NMDA), and kainate glutamate receptor subtypes using electrophysiological assessments. METHODS: AMPA receptor subunit-expressing cell lines (hGluA1-4, including two kinds of Q/R RNA-editing variants of hGluA2), NMDA receptor-expressing cells (hNR1/hNR2B), and kainate receptor-expressing cells (hGluK2) were developed in house. The effects of perampanel, and other ASMs including topiramate, phenobarbital, lamotrigine, gabapentin, carbamazepine, valproate, levetiracetam, and lacosamide, on AMPA, NMDA, and kainate receptors were evaluated by automated patch-clamp technique. In the same way, 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline (NBQX) and GYKI 52466 were evaluated as reference compounds of AMPA receptor antagonists. For the AMPA receptor functional assay, AMPA currents were elicited by AMPA in the presence of cyclothiazide. NMDA with glycine was used as a stimulant for the NMDA receptor assays, while glutamate was used for the kainate receptor assays. The mean 50 % inhibitory concentration (IC50) values were determined based on sigmoidal-curve fitting using GraphPad Prism software. RESULTS: Perampanel inhibited functions of hGluA1-4, but did not inhibit hNR1/hNR2B and hGluK2 up to 25 µM, the maximum soluble concentration. The IC50 values were 660 nM for hGluA1, 780 nM for hGluA2(R), 1200 nM for hGluA2(Q), 1200 nM for hGluA3, and 1800 nM for hGluA4. NBQX and GYKI 52466 also inhibited the function of all AMPA receptor subunits, but did not inhibit hNR1/hNR2B and hGluK2. The IC50 values for NBQX were 880 nM for hGluA1, 290 nM for hGluA2(R), 310 nM for hGluA2(Q), 330 nM for hGluA3, and 630 nM for hGluA4. For GYKI 52466, IC50 values were 25,000 nM for hGluA1, 30,000 nM for hGluA2(R), 42,000 nM for hGluA2(Q), 28,000 nM for hGluA3, and 53,000 nM for hGluA4. Phenobarbital inhibited hGluA2(R) at an IC50 value of 730,000 nM. The majority of other ASMs evaluated in this study did not show a direct inhibitory effect on almost any of the glutamate receptor functions examined up to 1 M. However, lamotrigine and carbamazepine inhibited hNR1/hNR2B function at IC50 values of 930,000 and 1,000,000 nM, respectively. SIGNIFICANCE: Only a few ASMs evaluated in this study showed direct interaction with ionotropic glutamate receptors. Perampanel is the only ASM that had a potent inhibitory effect on all AMPA receptor subtypes, but did not inhibit NMDA or kainate receptor subunits; while phenobarbital inhibited GluA2(R), and carbamazepine and lamotrigine inhibited the NMDA receptor at high concentration ranges.


Nitriles/pharmacology , Pyridones/pharmacology , Receptors, AMPA/drug effects , Receptors, Ionotropic Glutamate/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , Animals , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , N-Methylaspartate/pharmacology , Receptors, AMPA/metabolism , Receptors, Ionotropic Glutamate/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
5.
J Biol Chem ; 295(19): 6280-6288, 2020 05 08.
Article En | MEDLINE | ID: mdl-32161119

Kainate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors are two major, closely related receptor subtypes in the glutamate ion channel family. Excessive activities of these receptors have been implicated in a number of central nervous system diseases. Designing potent and selective antagonists of these receptors, especially of kainate receptors, is useful for developing potential treatment strategies for these neurological diseases. Here, we report on two RNA aptamers designed to individually inhibit kainate and AMPA receptors. To improve the biostability of these aptamers, we also chemically modified these aptamers by substituting their 2'-OH group with 2'-fluorine. These 2'-fluoro aptamers, FB9s-b and FB9s-r, were markedly resistant to RNase-catalyzed degradation, with a half-life of ∼5 days in rat cerebrospinal fluid or serum-containing medium. Furthermore, FB9s-r blocked AMPA receptor activity. Aptamer FB9s-b selectively inhibited GluK1 and GluK2 kainate receptor subunits, and also GluK1/GluK5 and GluK2/GluK5 heteromeric kainate receptors with equal potency. This inhibitory profile makes FB9s-b a powerful template for developing tool molecules and drug candidates for treatment of neurological diseases involving excessive activities of the GluK1 and GluK2 subunits.


Aptamers, Nucleotide/metabolism , Receptors, Kainic Acid/metabolism , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Base Sequence , Fluorine/chemistry , HEK293 Cells , Humans , Receptors, Kainic Acid/antagonists & inhibitors , Substrate Specificity , Transcription, Genetic , GluK2 Kainate Receptor
6.
Toxicology ; 432: 152381, 2020 02 28.
Article En | MEDLINE | ID: mdl-31981724

Chronic glutamate excitotoxicity has been thought to be involved in numerous neurodegenerative disorders. A small but significant loss of membrane cholesterol has been reported following a short stimulation of ionotropic glutamate receptors (iGluRs). We investigated the alteration of brain cholesterol following chronic glutamate treatment. The alteration of cholesterol levels was evaluated in the hippocampus from the adult rats that received the subcutaneous injection with monosodium l-glutamate at 1, 3, 5, and 7 days of age. The regulation of CYP46A1, LXRα, and ApoE levels were assayed following subtoxic glutamate treatment in SH-SY5Y cells as well as HT-22 cells lacking iGluRs. The ratio of 24S-hydroxycholesterol to cholesterol was elevated in the adult rats exposed to monosodium l-glutamate before the weaning age, compared to the control. The blockers of NMDA receptor (MK801) and mGluR5 (MPEP) attenuated the glutamate-induced loss of cholesterol and elevation of 24S-hydroxycholesterol level in SH-SY5Y cells. The induction of the mRNA levels of CYP46A1, LXRα, and ApoE by glutamate was observed in both SH-SY5Y cells and HT-22 cells; additionally, MK801 and MPEP attenuated the increases in these genes in SH-SY5Y cells. The increase in the binding of LXRα proteins with ApoE promoter following glutamate treatment was attenuated by MK801. The luciferase assay indicated the binding of CREB protein with CYP46A1 promoter, and the glutamate-induced CREB expression was inhibited by MK801. The results suggest that glutamate, the major excitatory neurotransmitter, may affect the metabolism and redistribution of cholesterol in the neuronal cells via its specific receptors during chronic exposure.


Apolipoproteins E/biosynthesis , Brain Chemistry/drug effects , Cholesterol 24-Hydroxylase/biosynthesis , Cholesterol/metabolism , Sodium Glutamate/pharmacology , Animals , Cell Line , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Homeostasis , Liver X Receptors/antagonists & inhibitors , Maze Learning/drug effects , Rats , Rats, Wistar , Receptors, Glutamate/drug effects , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Up-Regulation/drug effects
7.
Neurobiol Dis ; 133: 104406, 2020 01.
Article En | MEDLINE | ID: mdl-30798006

One of the devastating effects of acute exposure to organophosphates, like nerve agents, is the induction of severe and prolonged status epilepticus (SE), which can cause death, or brain damage if death is prevented. Seizures after exposure are initiated by muscarinic receptor hyperstimulation-after inhibition of acetylcholinesterase by the organophosphorus agent and subsequent elevation of acetylcholine-but they are reinforced and sustained by glutamatergic hyperexcitation, which is the primary cause of brain damage. Diazepam is the FDA-approved anticonvulsant for the treatment of nerve agent-induced SE, and its replacement by midazolam is currently under consideration. However, clinical data derived from the treatment of SE of any etiology, as well as studies on the control of nerve agent-induced SE in animal models, have indicated that diazepam and midazolam control seizures only temporarily, their antiseizure efficacy is reduced as the latency of treatment from the onset of SE increases, and their neuroprotective efficacy is limited or absent. Here, we review data on the discovery of a novel anticonvulsant and neuroprotectant, LY293558, an AMPA/GluK1 receptor antagonist. Treatment of soman-exposed immature, young-adult, and aged rats with LY293558, terminates SE with limited recurrence of seizures, significantly protects from brain damage, and prevents long-term behavioral deficits, even when LY293558 is administered 1 h post-exposure. More beneficial effects and complete neuroprotection is obtained when LY293558 administration is combined with caramiphen, which antagonizes NMDA receptors. Further efficacy studies may bring the LY293558 + caramiphen combination therapy on the pathway to approval for human use.


Anticonvulsants/pharmacology , Isoquinolines/pharmacology , Neuroprotective Agents/pharmacology , Organophosphate Poisoning , Receptors, AMPA/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors , Tetrazoles/pharmacology , Animals , Cholinesterase Inhibitors/toxicity , Humans , Nerve Agents/toxicity , Organophosphate Poisoning/drug therapy , Rats , Soman/toxicity , Status Epilepticus/chemically induced
8.
ACS Chem Neurosci ; 10(11): 4685-4695, 2019 11 20.
Article En | MEDLINE | ID: mdl-31622082

Selective pharmacological tool compounds are invaluable for understanding the functions of the various ionotropic glutamate receptor subtypes. For the kainate receptors, these compounds are few. Here we have synthesized nine novel quinoxaline-2,3-diones with substitutions in the 7-position to investigate the structure-activity relationship at kainate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Compound 11 exhibited the highest binding affinity across GluK1-3 while having selectivity toward kainate vs AMPA receptors. Compound 11 potently inhibited glutamate evoked currents at homomeric GluK1 and GluK3 receptors in HEK293 cells with Kb values of 65 and 39 nM, respectively. The binding mode of 11 in the ligand binding domain of GluK1 was investigated by X-ray crystallography, revealing that 11 stabilizes the receptor in an open conformation, consistent with its demonstrated antagonism. Furthermore, 11 was tested for analgesic effects in the mouse tail flick test where it significantly increased tail flick latency at doses where 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]-quinoxaline-7-sulfonamide (NBQX) was ineffective.


Analgesics/metabolism , Crystallography, X-Ray/methods , Excitatory Amino Acid Antagonists/metabolism , Quinoxalines/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Analgesics/chemistry , Analgesics/pharmacology , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Humans , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Protein Structure, Secondary , Protein Structure, Tertiary , Quinoxalines/chemistry , Quinoxalines/pharmacology , Rats , Receptors, Kainic Acid/chemistry , Structure-Activity Relationship
9.
Neuropharmacology ; 160: 107690, 2019 12 01.
Article En | MEDLINE | ID: mdl-31271770

Pain is among the most common symptoms in cancer and approximately 90% of patients experience end-stage cancer pain. The management of cancer pain is challenging due to the significant side effects associated with opioids, and novel therapeutic approaches are needed. MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores joined by a 22-atom spacer. MMG22 exhibited extraordinary analgesia following intrathecal administration in a mouse model of bone cancer pain. Here, we assessed the effectiveness of systemic administration of MMG22 in reducing cancer pain and evaluated whether MMG22 displays side effects associated with opioids. Fibrosarcoma cells were injected into and around the calcaneus bone in C3H mice. Mechanical hyperalgesia was defined as an increase in the paw withdrawal frequencies (PWFs) evoked by application of a von Frey monofilament (3.9 mN bending force) applied to the plantar surface of the hind paw Subcutaneous (s.c.), intramuscular (i.m.), and oral (p.o.) administration of MMG22 produced robust dose-dependent antihyperalgesia, whose ED50 was orders of magnitude lower than morphine. Moreover, the ED50 for MMG22 decreased with disease progression. Importantly, s.c. administration of MMG22 did not produce acute (24 h) or long-term (9 days) tolerance, was not rewarding (conditioned place preference test), and did not produce naloxone-induced precipitated withdrawal or alter motor function. A possible mechanism of action of MMG22 is discussed in terms of inhibition of spinal NMDAR via antagonism of its co-receptor, mGluR5, and concomitant activation of neuronal MOR. We suggest that MMG22 may be a powerful alternative to traditional opioids for managing cancer pain. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Cancer Pain/drug therapy , Cancer Pain/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Disease Models, Animal , Drug Administration Routes , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Hyperalgesia/drug therapy , Ligands , Male , Mice , Mice, Inbred C3H , Morphine/therapeutic use , Receptors, Kainic Acid/administration & dosage , Receptors, Opioid, mu/administration & dosage
10.
ACS Chem Neurosci ; 10(3): 1841-1853, 2019 03 20.
Article En | MEDLINE | ID: mdl-30620174

Among the ionotropic glutamate receptors, the physiological role of kainate receptors is less well understood. Although ligands with selectivity toward the kainate receptor subtype GluK1 are available, tool compounds with selectivity at the remaining kainate receptor subtypes are sparse. Here, we have synthesized a series of quinoxaline-2,3-diones with substitutions in the N1-, 6-, and 7-position to investigate the structure-activity relationship (SAR) at GluK1-3 and GluK5. Pharmacological characterization at native and recombinant kainate and AMPA receptors revealed that compound 37 had a GluK3-binding affinity ( Ki) of 0.142 µM and 8-fold preference for GluK3 over GluK1. Despite lower binding affinity of 22 at GluK3 ( Ki = 2.91 µM), its preference for GluK3 over GluK1 and GluK2 was >30-fold. Compound 37 was crystallized with the GluK1 ligand-binding domain to understand the SAR. The X-ray structure showed that 37 stabilized the protein in an open conformation, consistent with an antagonist binding mode.


Quinoxalines/pharmacology , Receptors, AMPA/metabolism , Receptors, Kainic Acid/metabolism , Structure-Activity Relationship , Animals , Models, Molecular , Protein Domains/physiology , Receptors, Kainic Acid/antagonists & inhibitors
11.
Sci Rep ; 8(1): 7035, 2018 05 04.
Article En | MEDLINE | ID: mdl-29728616

Kainate receptors (KARs) are glutamate receptors with peak expression during late embryonic and early postnatal periods. Altered KAR-mediated neurotransmission and subunit expression are observed in several brain disorders, including epilepsy. Here, we examined the role of KARs in regulating seizures in neonatal C57BL/6 mice exposed to a hypoxic insult. We found that knockout of the GluK2 subunit, or blockade of KARs by UBP310 reduced seizure susceptibility during the period of reoxygenation. Following the hypoxic insult, we observed an increase in excitatory neurotransmission in hippocampal CA3 pyramidal cells, which was blocked by treatment with UBP310 prior to hypoxia. Similarly, we observed increased excitatory neurotransmission in CA3 pyramidal cells in an in vitro hippocampal slice model of hypoxic-ischemia. This increase was absent in slices from GluK2-/- mice and in slices treated with UBP310, suggesting that KARs regulate, at least in part, excitatory synaptic neurotransmission following in vivo hypoxia in neonatal mice. Data from these hypoxia models demonstrate that KARs, specifically those containing the GluK2 subunit, contribute to alterations in excitatory neurotransmission and seizure susceptibility, particularly during the reoxygenation period, in neonatal mice. Therapies targeting KARs may prove successful in treatment of neonates affected by hypoxic seizures.


Hypoxia/metabolism , Receptors, Kainic Acid/metabolism , Seizures/etiology , Seizures/metabolism , Alanine/analogs & derivatives , Alanine/pharmacology , Animals , Animals, Newborn , Disease Models, Animal , Disease Susceptibility , Electroencephalography , Hippocampus/metabolism , Hippocampus/physiopathology , Hypoxia/genetics , Mice , Mice, Knockout , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/genetics , Seizures/drug therapy , Seizures/physiopathology , Synaptic Potentials , Thymine/analogs & derivatives , Thymine/pharmacology
12.
Bull Exp Biol Med ; 164(4): 434-438, 2018 Mar.
Article En | MEDLINE | ID: mdl-29500804

We performed screening of nootropic properties of 10 new derivatives of quinolizidine alkaloid (-)-cytisine. Compounds with ß-endo stereochemistry were more active than α-endo-isomers. Under stress conditions (3aR,4S,8S,12R,12aS,12bR)-10-methyl-2-phenyloctahydro-1H-4,12a-etheno-8,12-methanopyrrolo[3',4':3,4]pyrido[1,2-a] [1,5]diazocine-1,3,5(4H)-trione enhanced memory and had a positive effect on cognitive functions of rats. According to molecular docking data, the nootropic activity of the compound can be associated with its affinity for the glutamate-binding subunits GluK1 and GluR2 of the kainate and AMPA receptor, respectively.


Alkaloids/pharmacology , Nootropic Agents/pharmacology , Receptors, AMPA/chemistry , Receptors, Kainic Acid/chemistry , Alkaloids/chemical synthesis , Animals , Avoidance Learning/drug effects , Azocines/chemical synthesis , Azocines/pharmacology , Binding Sites , Female , Gene Expression , Male , Mice , Molecular Docking Simulation , Nootropic Agents/chemical synthesis , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Quinolizines/chemical synthesis , Quinolizines/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Receptors, Kainic Acid/agonists , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Structure-Activity Relationship , Toxicity Tests, Acute
13.
J Med Chem ; 61(5): 2124-2130, 2018 03 08.
Article En | MEDLINE | ID: mdl-29451794

Starting from 1-4 and 7 structural templates, analogues based on bioisosteric replacements (5a-c vs 1, 2 and 6 vs 7) were synthesized for completing the SAR analysis. Interesting binding properties at GluA2, GluK1, and GluK3 receptors were discovered. The requirements for GluK3 interaction were elucidated by determining the X-ray structures of the GluK3-LBD with 2 and 5c and by computational studies. Antinociceptive potential was demonstrated for GluK1 partial agonist 3 and antagonist 7 (2 mg/kg ip).


Receptors, Kainic Acid/chemistry , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/analogs & derivatives , Analgesics/chemistry , Animals , Crystallography, X-Ray , Ligands , Protein Binding , Receptors, AMPA , Receptors, Kainic Acid/agonists , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Structure-Activity Relationship , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/chemistry , GluK3 Kainate Receptor
14.
ACS Chem Neurosci ; 8(11): 2437-2445, 2017 11 15.
Article En | MEDLINE | ID: mdl-28872832

Glutamate ion channels have three subtypes, that is, α-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA), kainate, and N-methyl-d-aspartate (NMDA) receptors. Excessive activity of these receptor subtypes either individually or collectively is involved in various neurological disorders. RNA aptamers as antagonists of these receptors are potential therapeutics. For developing aptamer therapeutics, the RNA aptamers must be chemically modified to become ribonuclease-resistant or stable in biological fluids. Using systematic evolution of ligands by exponential enrichment (SELEX) and a chemically modified library, prepared enzymatically (i.e., the library contains RNAs with 2'-fluoro modified nucleoside triphosphates or ATPs, CTPs and UTPs, but regular GTPs), we have isolated an aptamer. The short aptamer (69 nucleotides) FN1040s selectively inhibits the GluA1 and GluA2Qflip AMPA receptor subunits, whereas the full-length aptamer (101 nucleotides) FN1040 additionally inhibits GluK1, but not GluK2, kainate receptor, and GluN1a/2A and GluN1a/2B, the two major native NMDA receptors. The two aptamers show similar potency (2-4 µM) and are stable with a half-life of at least 2 days in serum-containing medium or cerebrospinal fluid. Therefore, these two aptamers are amenable for in vivo use.


Aptamers, Nucleotide/chemistry , Excitatory Amino Acid Antagonists/chemistry , Receptors, AMPA/antagonists & inhibitors , Adenosine/analogs & derivatives , Animals , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/pharmacology , Binding, Competitive , Cattle , Cerebrospinal Fluid , Cytidine/analogs & derivatives , Drug Design , Drug Evaluation, Preclinical , Excitatory Amino Acid Antagonists/chemical synthesis , Excitatory Amino Acid Antagonists/pharmacology , Gene Library , HEK293 Cells , Humans , Models, Molecular , Nucleic Acid Conformation , Patch-Clamp Techniques , Rats , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , SELEX Aptamer Technique , Serum , Substrate Specificity , Uridine/analogs & derivatives
15.
Neurosci Lett ; 655: 137-142, 2017 Aug 10.
Article En | MEDLINE | ID: mdl-28673833

Fast ripples (FR) are high frequency oscillations (250-600Hz) that have been associated with epilepsy. FR are assumed to be generated in small areas of the hippocampus (1mm3) that contain pathologically interconnected glutamate pyramidal cell clusters. Additionally, a relation between glutamate neurotransmission and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/kainite (AMPA/KA) and metabotropic mGluR5 receptors is well established. Therefore, we hypothesized that antagonism of these glutamate receptors would decrease FR activity. For this propose, we induced status epilepticus with a kainic acid injection in the posterior right hippocampus and performed intracranial EEG recordings to detect and evaluate the presence of FR 15days after the injection. The glutamate AMPA/KA receptor antagonist CNQX (10mg/kg) and the mGluR5 antagonist MPEP (20mg/kg) were administered intraperitoneally, and the effects of the drugs were evaluated for a period of three hours after their administration. The results show a decrease in the number of FR in the first hour after drug administration in both cases (CNQX, p=0.0125; MPEP, p=0.0132) and a return to basal values in the third hour of the experiment, but not significant differences in the number of oscillations per event of FR, and the frequency and duration of each event of FR. We therefore conclude that blockade of AMPA/KA and mGluR5 receptors transiently decreases the generation of FR; however, the mechanisms by which this effect is achieved are to be further analyzed in future experiments.


6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Kainic Acid/pharmacology , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptors, AMPA/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors , Animals , Electroencephalography , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Rats, Wistar , Seizures/physiopathology
16.
J Biol Chem ; 292(18): 7338-7347, 2017 05 05.
Article En | MEDLINE | ID: mdl-28325839

AMPA and kainate receptors, along with NMDA receptors, represent different subtypes of glutamate ion channels. AMPA and kainate receptors share a high degree of sequence and structural similarities, and excessive activity of these receptors has been implicated in neurological diseases such as epilepsy. Therefore, blocking detrimental activity of both receptor types could be therapeutically beneficial. Here, we report the use of an in vitro evolution approach involving systematic evolution of ligands by exponential enrichment with a single AMPA receptor target (i.e. GluA1/2R) to isolate RNA aptamers that can potentially inhibit both AMPA and kainate receptors. A full-length or 101-nucleotide (nt) aptamer selectively inhibited GluA1/2R with a KI of ∼5 µm, along with GluA1 and GluA2 AMPA receptor subunits. Of note, its shorter version (55 nt) inhibited both AMPA and kainate receptors. In particular, this shorter aptamer blocked equally potently the activity of both the GluK1 and GluK2 kainate receptors. Using homologous binding and whole-cell recording assays, we found that an RNA aptamer most likely binds to the receptor's regulatory site and inhibits it noncompetitively. Our results suggest the potential of using a single receptor target to develop RNA aptamers with dual activity for effectively blocking both AMPA and kainate receptors.


Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Receptors, AMPA/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors , Animals , Cell Line , Rats , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/metabolism , GluK2 Kainate Receptor
17.
Neuropharmacology ; 112(Pt A): 16-28, 2017 01.
Article En | MEDLINE | ID: mdl-27236079

Kainate receptors belong to the family of ionotropic glutamate receptors. These receptors assemble from five subunits (GluK1-5) into tetrameric ion channels. Kainate receptors are located at both pre- and postsynaptic membranes in the central nervous system where they contribute to excitatory synaptic transmission and modulate network excitability by regulating neurotransmitter release. Dysfunction of kainate receptors has been implicated in several neurological disorders such as epilepsy, schizophrenia and depression. Here we provide a review on the current understanding of kainate receptor structure and how they bind agonists, antagonists and ions. The first structure of the ligand-binding domain of the GluK1 subunit was reported in 2005, seven years after publication of the crystal structure of a soluble construct of the ligand-binding domain of the AMPA-type subunit GluA2. Today, a full-length structure has been determined of GluK2 by cryo electron microscopy to 7.6 Å resolution as well as 84 high-resolution crystal structures of N-terminal domains and ligand-binding domains, including agonist and antagonist bound structures, modulatory ions and mutations. However, there are still many unanswered questions and challenges in front of us. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.


Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Excitatory Amino Acid Agents/pharmacology , Humans , Ligands , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Receptors, Kainic Acid/agonists , Receptors, Kainic Acid/antagonists & inhibitors
18.
Reprod Fertil Dev ; 29(6): 1231-1238, 2017 Jun.
Article En | MEDLINE | ID: mdl-27225229

Vitamin D is a versatile signalling molecule that plays a critical role in calcium homeostasis. There are several studies showing the genomic action of vitamin D in the control of reproduction; however, the quick non-genomic action of vitamin D at the hypothalamic level is not well understood. Therefore, to investigate the effect of vitamin D on juvenile gonadotrophin-releasing hormone (GnRH) neurons, excitatory neurotransmitter receptor agonists N-methyl-D-aspartate (NMDA, 30µM) and kainate (10µM) were applied in the absence or in the presence of vitamin D3 (VitaD3, 10nM). The NMDA-mediated responses were decreased by VitaD3 in the absence and in the presence of tetrodotoxin (TTX), a sodium-channel blocker, with the mean relative inward current being 0.56±0.07 and 0.66±0.07 (P<0.05), respectively. In addition, VitaD3 induced a decrease in the frequency of gamma-aminobutyric acid mediated (GABAergic) spontaneous postsynaptic currents and spontaneous postsynaptic currents induced by NMDA application with a mean relative frequency of 0.595±0.07 and 0.56±0.09, respectively. Further, VitaD3 decreased the kainate-induced inward currents in the absence and in the presence of TTX with a relative inward current of 0.64±0.06 and 0.68±0.06, respectively (P<0.05). These results suggest that VitaD3 has a non-genomic action and partially inhibits the NMDA and kainate receptor-mediated actions of GnRH neurons, suggesting that VitaD3 may regulate the hypothalamic-pituitary-gonadal (HPG) axis at the time of pubertal development.


Cholecalciferol/metabolism , Gonadotropin-Releasing Hormone/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/metabolism , Preoptic Area/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Epigenesis, Genetic , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Kainic Acid/metabolism , Male , Mice, Transgenic , N-Methylaspartate/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Preoptic Area/cytology , Preoptic Area/drug effects , Receptors, Kainic Acid/agonists , Receptors, Kainic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Sodium Channel Blockers/pharmacology , Synaptic Potentials/drug effects
19.
Cell Mol Neurobiol ; 37(2): 315-328, 2017 Mar.
Article En | MEDLINE | ID: mdl-27053349

Glutamate receptors in mesolimbic areas such as the nucleus accumbens, ventral tegmental area, prefrontal cortex (PFC), and hippocampus (HIP) are a component of the mechanisms of drug-induced reward and can modulate the firing pattern of dopaminergic neurons in the reward system. In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Therefore, in the present study, we investigated the changes in phosphorylated CREB (p-CREB) and c-fos induction within the nucleus accumbens (NAc), HIP, and PFC after intracerebroventricular (ICV) administration of different doses of CNQX or vehicle during extinction period or reinstatement of morphine-induced CPP. In all groups, the CPP procedure was done; afterward, the conditioning scores were recorded by Ethovision software. After behavioral test recording, we dissected out the NAc, HIP, and PFC regions and measured the p-CREB/CREB ratio and c-fos level by Western blot analysis. Our results showed that administration of CNQX significantly shortened the extinction of morphine CPP. Besides, ICV microinjection of CNQX following extinction period decreased the reinstatement of morphine CPP in extinguished rats. In molecular section, in treatment group, all mentioned factors were dose-dependently decreased in comparison with vehicle group (DMSO) after ICV microinjection of different doses of CNQX but not in pre-extinction microinjection. These findings suggested that antagonism of AMPA receptor decreased p-CREB/CREB ratio and c-fos level in the PFC, NAc, and HIP. Modulation of the drug memory reconsolidation may be useful for faster extinction of drug-induced reward and attenuation of drug-seeking behavior.


Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Morphine/administration & dosage , Receptors, AMPA/metabolism , Receptors, Kainic Acid/metabolism , Animals , Brain/drug effects , Brain/metabolism , Conditioning, Psychological/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Extinction, Psychological/drug effects , Infusions, Intraventricular , Male , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors
20.
J Med Chem ; 60(1): 441-457, 2017 01 12.
Article En | MEDLINE | ID: mdl-28005385

Ionotropic glutamate receptor antagonists are valuable tool compounds for studies of neurological pathways in the central nervous system. On the basis of rational ligand design, a new class of selective antagonists, represented by (2S,4R)-4-(2-carboxyphenoxy)pyrrolidine-2-carboxylic acid (1b), for cloned homomeric kainic acid receptors subtype 1 (GluK1) was attained (Ki = 4 µM). In a functional assay, 1b displayed full antagonist activity with IC50 = 6 ± 2 µM. A crystal structure was obtained of 1b when bound in the ligand binding domain of GluK1. A domain opening of 13-14° was seen compared to the structure with glutamate, consistent with 1b being an antagonist. A structure-activity relationship study showed that the chemical nature of the tethering atom (C, O, or S) linking the pyrrolidine ring and the phenyl ring plays a key role in the receptor selectivity profile and that substituents on the phenyl ring are well accommodated by the GluK1 receptor.


Proline/antagonists & inhibitors , Pyrrolidines/pharmacology , Receptors, Ionotropic Glutamate/antagonists & inhibitors , Receptors, Kainic Acid/antagonists & inhibitors , Crystallography, X-Ray , Drug Design , Inhibitory Concentration 50 , Pyrrolidines/chemistry , Structure-Activity Relationship
...