Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 522
Filter
1.
J Hypertens ; 40(12): 2394-2405, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36189462

ABSTRACT

OBJECTIVE: Cyclooxygenase (COX) is critical in regulating cardiovascular function, but its role involved in the central control of blood pressure (BP) is uncovered. The tonic glutamatergic inputs to the rostral ventrolateral medulla (RVLM) are enhanced in hypertension. Here, the present study was designed to investigate the effect and mechanism of central COX on tonic glutamatergic inputs to the RVLM and BP regulation. METHODS: Wistar-Kyoto (WKY) rats and spontaneous hypertensive rats (SHRs) received RVLM microinjection of adeno-associated viral vectors to promote or inhibit the COX2 expression were subjected to subsequent experiments. Glutamate level and glutaminase expression were detected by ELISA and western blot, respectively. The function of tonic glutamatergic inputs was assessed by BP response to microinjection of the glutamate receptor antagonist into the RVLM. PC12 cells were used to detect the underlying signal pathway. RESULTS: The RVLM COX2 expression and prostaglandin E2 level were significant higher in SHRs than in WKY rats. Overexpression of COX2 in the RVLM produced an increase in basal BP, RVLM glutamate level, and glutaminase expression in WKY rats, while they were significantly reduced by interfering with COX2 expression in SHRs. Microinjections of the glutamate receptor antagonist into the RVLM produced a significant BP decrease in WKY rats with COX2 overexpression pretreatment. Furthermore, the increased levels of BP, glutamate content, and glutaminase activity in the RVLM evoked by central infusion of angiotensin II were attenuated in COX2 knockout mice. It was also found that prostaglandin E2 increased supernatant glutamate level and phosphorylation of signal transducer and activator of transcription 3 in PC12 cells. CONCLUSION: Our findings suggest that upregulated COX2 expression enhances the tonically active glutamatergic inputs to the RVLM, which is associated with cardiovascular regulation in hypertension.


Subject(s)
Cyclooxygenase 2 , Glutaminase , Hypertension , Animals , Mice , Rats , Blood Pressure/physiology , Dinoprostone/metabolism , Excitatory Amino Acid Antagonists/metabolism , Glutamic Acid/metabolism , Glutaminase/metabolism , Medulla Oblongata , Rats, Inbred SHR , Rats, Inbred WKY
2.
Biochem Pharmacol ; 197: 114892, 2022 03.
Article in English | MEDLINE | ID: mdl-34968492

ABSTRACT

Treating major depression is a medical need that remains unmet by monoaminergic therapeutic strategies that commonly fail to achieve symptom remission. A breakthrough in the treatment of depression was the discovery that the anesthetic (R,S)-ketamine (ketamine), when administered at sub-anesthetic doses, elicits rapid (sometimes within hours) antidepressant effects in humans that are otherwise resistant to monoaminergic-acting therapies. While this finding was revolutionary and led to the FDA approval of (S)-ketamine (esketamine) for use in adults with treatment-resistant depression and suicidal ideation, the mechanisms underlying how ketamine or esketamine elicit their effects are still under active investigation. An emerging view is that metabolism of ketamine may be a crucial step in its mechanism of action, as several metabolites of ketamine have neuroactive effects of their own and may be leveraged as therapeutics. For example, (2R,6R)-hydroxynorketamine (HNK), is readily observed in humans following ketamine treatment and has shown therapeutic potential in preclinical tests of antidepressant efficacy and synaptic potentiation while being devoid of the negative adverse effects of ketamine, including its dissociative properties and abuse potential. We discuss preclinical and clinical studies pertaining to how ketamine and its metabolites produce antidepressant effects. Specifically, we explore effects on glutamate neurotransmission through N-methyl D-aspartate receptors (NMDARs) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), synaptic structural changes via brain derived neurotrophic factor (BDNF) signaling, interactions with opioid receptors, and the enhancement of serotonin, norepinephrine, and dopamine signaling. Strategic targeting of these mechanisms may result in novel rapid-acting antidepressants with fewer undesirable side effects compared to ketamine.


Subject(s)
Antidepressive Agents/metabolism , Antidepressive Agents/therapeutic use , Depression/drug therapy , Depression/metabolism , Ketamine/metabolism , Ketamine/therapeutic use , Animals , Brain-Derived Neurotrophic Factor/metabolism , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/therapeutic use , Humans , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Folia Morphol (Warsz) ; 81(2): 379-386, 2022.
Article in English | MEDLINE | ID: mdl-33778937

ABSTRACT

BACKGROUND: Nesfatin-1 is a newly identified satiety peptide that has regulatory effects on food intake and glucose metabolism, and is located in the hypothalamic nuclei, including the supraoptic nucleus (SON). In this study, we have investigated the hypothesis that nesfatin-1 neurons are activated by refeeding and intraperitoneal glucose injection and that the glutamatergic system has regulatory influences on nesfatin-1 neurons in the SON. MATERIALS AND METHODS: The first set of experiments analysed activation of nesfatin-1 neurons after refeeding as a physiological stimulus and the effectiveness of the glutamatergic system on this physiological stimulation. The subjects were randomly divided into three groups: fasting group, refeeding group and antagonist (CNQX + refeeding) group. The second set of experiments analysed activation of nesfatin-1 neurons by glucose injection as a metabolic stimulus and the effectiveness of the glutamatergic system on this metabolic stimulation. The subjects were randomly divided into three groups: saline group, glucose group and antagonist (CNQX + glucose) group. RESULTS: Refeeding significantly increased the number of activated nesfatin-1 neurons by approximately 66%, and intraperitoneal glucose injection activated these neurons by about 55%, compared to the fasting and saline controls. The injections of glutamate antagonist (CNQX) greatly decreased the number of activated nesfatin-1 neurons. CONCLUSIONS: This study suggested that nesfatin-1 neurons were activated by peripheral and/or metabolic signals and that this effect was mediated through the glutamatergic system.


Subject(s)
Excitatory Amino Acid Antagonists , Glucose , 6-Cyano-7-nitroquinoxaline-2,3-dione/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Eating/physiology , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Glucose/metabolism , Glucose/pharmacology , Humans , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nucleobindins
4.
Neurosci Res ; 176: 57-65, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34656645

ABSTRACT

The immature brain is highly sensitive to disturbances in the functioning of N-methyl-d-aspartate (NMDA) receptor in rodents, and blockade of the receptor during postnatal brain development period causes schizophrenia-like behavior in adulthood. During the postnatal period, NR2A- and NR2B-containing NMDA receptors are highly expressed, and these two subunits show different expression patterns in the brain. However, the functions of these two NMDA receptors are unknown. In this study, we treated rats with an NR2A-preferring NMDA receptor antagonist (PEAQX, 10 mg/kg), an NR2B-selective NMDA receptor antagonist (ifenprodil, 7.5 mg/kg), or a nonselective blocker of the NMDA receptor (MK-801, 0.4 mg/kg) during the neonatal period. Rats neonatally treated with MK-801 or PEAQX showed spatial working memory deficits in the Y-maze test. PEAQX-treated rats also showed greater reactivity to acoustic stimuli and hypersensitivity to acute MK-801 challenge. However, ifenprodil treatment did not cause any detectable behavioral changes. These results suggest that the NR2A-containing NMDA receptor is indispensable for proper brain development in rats, and functional disturbances in this subunit impair hippocampus-dependent spatial working memory in adulthood.


Subject(s)
Excitatory Amino Acid Antagonists , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/metabolism , Memory Disorders/chemically induced , Memory, Short-Term , Rats
5.
J Pharmacol Exp Ther ; 379(1): 41-52, 2021 10.
Article in English | MEDLINE | ID: mdl-34493631

ABSTRACT

We describe a clinical candidate molecule from a new series of glutamate N-methyl-d-aspartate receptor subunit 2B-selective inhibitors that shows enhanced inhibition at extracellular acidic pH values relative to physiologic pH. This property should render these compounds more effective inhibitors of N-methyl-d-aspartate receptors at synapses responding to a high frequency of action potentials, since glutamate-containing vesicles are acidic within their lumen. In addition, acidification of penumbral regions around ischemic tissue should also enhance selective drug action for improved neuroprotection. The aryl piperazine we describe here shows strong neuroprotective actions with minimal side effects in preclinical studies. The clinical candidate molecule NP10679 has high oral bioavailability with good brain penetration and is suitable for both intravenous and oral dosing for therapeutic use in humans. SIGNIFICANCE STATEMENT: This study identifies a new series of glutamate N-methyl-d-aspartate (NMDA) receptor subunit 2B-selective negative allosteric modulators with properties appropriate for clinical advancement. The compounds are more potent at acidic pH, associated with ischemic tissue, and this property should increase the therapeutic safety of this class by improving efficacy in affected tissue while sparing NMDA receptor block in healthy brain.


Subject(s)
Brain/drug effects , Brain/metabolism , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Acids , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , Female , Hydrogen-Ion Concentration , Male , Mice , Mice, Inbred C57BL , Xenopus laevis
6.
Cell Rep ; 36(9): 109648, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469715

ABSTRACT

Metabotropic glutamate receptors (mGluRs) are dimeric G-protein-coupled receptors activated by the main excitatory neurotransmitter, L-glutamate. mGluR activation by agonists binding in the venus flytrap domain is regulated by positive (PAM) or negative (NAM) allosteric modulators binding to the 7-transmembrane domain (7TM). We report the cryo-electron microscopy structures of fully inactive and intermediate-active conformations of mGlu5 receptor bound to an antagonist and a NAM or an agonist and a PAM, respectively, as well as the crystal structure of the 7TM bound to a photoswitchable NAM. The agonist induces a large movement between the subunits, bringing the 7TMs together and stabilizing a 7TM conformation structurally similar to the inactive state. Using functional approaches, we demonstrate that the PAM stabilizes a 7TM active conformation independent of the conformational changes induced by agonists, representing an alternative mode of mGlu activation. These findings provide a structural basis for different mGluR activation modes.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Signal Transduction/drug effects , Cryoelectron Microscopy , Crystallography, X-Ray , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Antagonists/metabolism , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Subunits , Receptor, Metabotropic Glutamate 5/metabolism , Receptor, Metabotropic Glutamate 5/ultrastructure , Structure-Activity Relationship
7.
J Appl Toxicol ; 41(12): 2083-2089, 2021 12.
Article in English | MEDLINE | ID: mdl-34002392

ABSTRACT

Ketamine is an N-methyl-D-aspartate (NMDA) receptor antagonist. Used as an anesthetic, potential neurotoxic and cardiotoxic effects of ketamine in animal models have been reported. The underlying mechanisms of ketamine-induced toxicity are not clear. The zebrafish is an ideal model for toxicity assays because of its predictive capability in chemical testing, which compares well with that of mammalian models. To gain insight into potential mechanisms of ketamine effects, we performed real-time quantitative polymerase chain reaction-based gene expression array analyses. Gene expression analysis was conducted for multiple genes (a total of 84) related to 10 major signaling pathways including the transforming growth factor ß (TGFß), Wingless and Int-1 (Wnt), nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), Janus kinase/signal transducers and activators of transcription (JAK/STAT), p53, Notch, Hedgehog, peroxisome proliferator-activated receptor (PPAR), oxidative stress, and hypoxia pathways. Our results show that ketamine altered the expression of specific genes related to hypoxia, p53, Wnt, Notch, TGFß, PPAR, and oxidative stress pathways. Thus, we can further focus on these specific pathways to elucidate the mechanisms by which ketamine elicits a toxic response.


Subject(s)
Anesthetics, Dissociative/toxicity , Gene Expression/drug effects , Ketamine/toxicity , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Zebrafish/genetics , Animals , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Excitatory Amino Acid Antagonists/metabolism , Signal Transduction , Zebrafish/embryology
8.
Pharmacol Biochem Behav ; 203: 173154, 2021 04.
Article in English | MEDLINE | ID: mdl-33609599

ABSTRACT

Interest in developing NMDA receptor antagonists with reduced side-effects for neurological and psychiatric disorders has been re-energized by the recent introduction of esketamine into clinical practice for treatment-resistant depression. Structural analogs of dextromethorphan bind with low affinity to the NMDA receptor ion channel, have functional effects in vivo, and generally display a lower propensity for side-effects than that of ketamine and other higher affinity antagonists. As such, the aim of the present study was to determine whether a series of N-substituted-3-alkoxy-substituted dextromethorphan analogs produce their anticonvulsant effects through NMDA receptor blockade. Compounds were studied against NMDA-induced seizures in rats. Compounds were administered intracerebroventricularly in order to mitigate confounds of drug metabolism that arise from systemic administration. Comparison of the anticonvulsant potencies to their affinities for NMDA, σ1, and σ2 binding sites were made in order to evaluate the contribution of these receptors to anticonvulsant efficacy. The potencies to block convulsions were positively associated with their affinities to bind to the NMDA receptor ion channel ([3H]-TCP binding) (r = 0.71, p < 0.05) but not to σ1 receptors ([3H]-SKF 10047 binding) (r = -0.31, p = 0.46) or to σ2 receptors ([3H]-DTG binding) (p = -0.38, p = 0.36). This is the first report demonstrating that these dextromethorphan analogs are functional NMDA receptor antagonists in vivo. Given their potential therapeutic utility and favorable side-effect profiles, such low affinity NMDA receptor antagonists could be considered for further development in neurological (e.g., anticonvulsant) and psychiatric (e.g., antidepressant) disorders.


Subject(s)
Anticonvulsants/administration & dosage , Dextromethorphan/analogs & derivatives , Dextromethorphan/administration & dosage , Dextrorphan/administration & dosage , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Antagonists/administration & dosage , N-Methylaspartate/adverse effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/chemically induced , Seizures/drug therapy , Alcohols/chemistry , Animals , Anticonvulsants/metabolism , Binding Sites , Dextromethorphan/metabolism , Dextrorphan/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/metabolism , Infusions, Intraventricular , Ligands , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/metabolism , Treatment Outcome , Sigma-1 Receptor
9.
FEBS J ; 288(3): 995-1007, 2021 02.
Article in English | MEDLINE | ID: mdl-32543078

ABSTRACT

Ionotropic glutamate receptors are ligand-gated ion channels governing neurotransmission in the central nervous system. Three major types of antagonists are known for the AMPA-type receptor GluA2: competitive, noncompetitive (i.e., negative allosteric modulators; NAMs) used for treatment of epilepsy, and uncompetitive antagonists. We here report a 4.65 Å resolution X-ray structure of GluA2, revealing that four molecules of the competitive antagonist ZK200775 and four molecules of the NAM GYKI53655 are capable of binding at the same time. Using negative stain electron microscopy, we show that GYKI53655 alone or ZK200775/GYKI53655 in combination predominantly results in compact receptor forms. The agonist AMPA provides a mixed population of compact and bulgy shapes of GluA2 not impacted by addition of GYKI53655. Taken together, this suggests that the two different mechanisms of antagonism that lead to channel closure are independent and that the distribution between bulgy and compact receptors primarily depends on the ligand bound in the glutamate binding site. DATABASE: The atomic coordinates and structure factors from the crystal structure determination have been deposited in the Protein Data Bank under accession code https://doi.org/10.2210/pdb6RUQ/pdb. The electron microscopy 3D reconstruction volumes have been deposited in EMDB (EMD-4875: Apo; EMD-4920: ZK200775/GYKI53655; EMD-4921: AMPA compact; EMD-4922: AMPA/GYKI53655 bulgy; EMD-4923: GYKI53655; EMD-4924: AMPA bulgy; EMD-4925: AMPA/GYKI53655 compact).


Subject(s)
Benzodiazepines/metabolism , Excitatory Amino Acid Antagonists/metabolism , Organophosphonates/metabolism , Quinoxalines/metabolism , Receptors, AMPA/metabolism , Recombinant Proteins/metabolism , Allosteric Regulation , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Crystallography, X-Ray , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Organophosphonates/chemistry , Organophosphonates/pharmacology , Protein Binding , Protein Domains , Quinoxalines/chemistry , Quinoxalines/pharmacology , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/genetics , Recombinant Proteins/chemistry , Sf9 Cells , Spodoptera
10.
Pharmacol Res ; 161: 105128, 2020 11.
Article in English | MEDLINE | ID: mdl-32805354

ABSTRACT

Cannabidiol (CBD) is a major phytocannabinoid in Cannabis sativa. CBD is being increasingly reported as a clinical treatment for neurological diseases. Febrile seizure is one of the most common diseases in children with limited therapeutic options. We investigated possible therapeutic effects of CBD on febrile seizures and the underlying mechanism. Use of a hyperthermia-induced seizures model revealed that CBD significantly prolonged seizure latency and reduced the severity of thermally-induced seizures. Hippocampal neuronal excitability was significantly decreased by CBD. Further, CBD significantly reduced the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) mediated evoked excitatory postsynaptic currents (eEPSCs) and the amplitude and frequency of miniature EPSCs (mEPSCs). Furthermore, CBD significantly accelerated deactivation in GluA1 and GluA2 subunits. Interestingly, CBD slowed receptor recovery from desensitization of GluA1, but not GluA2. These effects on kinetics were even more prominent when AMPAR was co-expressed with γ-8, the high expression isoform 8 of transmembrane AMPAR regulated protein (TARPγ8) in the hippocampus. The inhibitory effects of CBD on AMPAR depended on its interaction with the distal N-terminal domain of GluA1/GluA2. CBD inhibited AMPAR activity and reduced hippocampal neuronal excitability, thereby improving the symptoms of febrile seizure in mice. The putative binding site of CBD in the N-terminal domain of GluA1/GluA2 may be a drug target for allosteric gating modulation of AMPAR.


Subject(s)
Anticonvulsants/pharmacology , Brain Waves/drug effects , CA1 Region, Hippocampal/drug effects , Cannabidiol/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hyperthermia/complications , Receptors, AMPA/antagonists & inhibitors , Seizures, Febrile/prevention & control , Animals , Anticonvulsants/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Cannabidiol/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/metabolism , Excitatory Postsynaptic Potentials/drug effects , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Kinetics , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/drug effects , Models, Molecular , Protein Binding , Reaction Time/drug effects , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Seizures, Febrile/etiology , Seizures, Febrile/metabolism , Seizures, Febrile/physiopathology
11.
Neurochem Int ; 138: 104752, 2020 09.
Article in English | MEDLINE | ID: mdl-32445659

ABSTRACT

Kynurenic acid (KYNA) is one of the most significant metabolite of the kynurenine pathway both in terms of functional and potential therapeutic value. It is an N-methyl-D-aspartate (NMDA) receptor antagonist, but it can also activate the G-protein coupled receptor 35 (GPR35), which shares several structural and functional properties with cannabinoid receptors. Previously our group demonstrated that systemic chronic KYNA treatment altered opioid receptor G-protein activity. Opioid receptors also overlap in many features with cannabinoid receptors. Thus, our aim was to examine the direct in vitro and systemic, chronic in vivo effect of KYNA on type 1 cannabinoid receptor (CB1R) binding and G-protein activity. Based on competition and [35S]GTPγS G-protein binding assays in rat brain, KYNA alone did not show significant binding towards the CB1R, nor did it alter CB1R ligand binding and agonist activity in vitro. When rats were chronically treated with KYNA (single daily, i.p., 128 mg/kg for 9 days), the KYNA plasma and cerebrospinal fluid levels significantly increased compared to vehicle treated group. Furthermore, in G-protein binding assays, in the whole brain the amount of G-proteins in basal and in maximum activity coupled to the CB1R also increased due to the treatment. At the same time, the overall stimulatory properties of the receptor remained unaltered in vehicle and KYNA treated samples. Similar observations were made in rat hippocampus, but not in the cortex and brainstem. In saturation binding assays the density of CB1Rs in rat whole brain and hippocampus were also significantly enhanced after the same treatment, without significantly affecting ligand binding affinity. Thus, KYNA indirectly and brain region specifically increases the abundance of functional CB1Rs, without modifying the overall binding and activity of the receptor. Supposedly, this can be a compensatory mechanism on the part of the endocannabinoid system induced by the long-term KYNA exposure.


Subject(s)
Brain/drug effects , Brain/metabolism , Kynurenic Acid/administration & dosage , Kynurenic Acid/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Benzoxazines/metabolism , Benzoxazines/pharmacology , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Male , Morpholines/metabolism , Morpholines/pharmacology , Naphthalenes/metabolism , Naphthalenes/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
12.
Neuropharmacology ; 166: 107947, 2020 04.
Article in English | MEDLINE | ID: mdl-31926944

ABSTRACT

Ketamine, an NMDA receptor antagonist and fast acting antidepressant, produces a rapid burst of glutamate in the ventral medial prefrontal cortex (mPFC). Preclinical studies have demonstrated that pyramidal cell activity in the vmPFC is necessary for the rapid antidepressant response to ketamine in rodents. We sought to characterize the effects of ketamine and its stereoisomers (R and S), as well as a metabolite, (2R,6R)-hydroxynorketamine (HNK), on vmPFC activity using a genetically encoded calcium indicator (GCaMP6f). Ratiometric fiber photometry was utilized to monitor GCaMP6f fluorescence in pyramidal cells of mouse vmPFC prior to and immediately following administration of compounds. GCaMP6f signal was assessed to determine correspondance of activity between compounds. We observed dose dependent effects with (R,S)-ketamine (3-100 mg/kg), with the greatest effects on GCaMP6f activity at 30 mg/kg and lasting up to 20 min. (S)-ketamine (15 mg/kg), which has high affinity for the NMDA receptor channel produced similar effects to (R,S)-ketamine, but compounds with low NMDA receptor affinity, including (R)-ketamine (15 mg/kg) and (2R,6R)-HNK (30 mg/kg) had little or no effect on GCaMP6f activity. The initial response to administration of (R,S)-ketamine as well as (S)-ketamine is characterized by a brief period of robust GCaMP6f activation, consistent with increased activity of vmPFC pyramidal neurons. Because (2R,6R)-HNK and (R)-ketamine are reported to have antidepressant activity in rodent models the current results indicate that different initiating mechanisms lead to similar brain adaptive consequences that underlie the rapid antidepressant responses.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/analogs & derivatives , Ketamine/pharmacology , Prefrontal Cortex/metabolism , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/metabolism , Ketamine/chemistry , Ketamine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Photometry/methods , Prefrontal Cortex/drug effects , Stereoisomerism
13.
Molecules ; 25(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979301

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disease caused by a CAG expansion mutation in the huntingtin gene. As a result, intranuclear inclusions of mutant huntingtin protein are formed, which damage striatal medium spiny neurons (MSNs). A review of Positron Emission Tomography (PET) studies relating to HD was performed, including clinical and preclinical data. PET is a powerful tool for visualisation of the HD pathology by non-invasive imaging of specific radiopharmaceuticals, which provide a detailed molecular snapshot of complex mechanistic pathways within the brain. Nowadays, radiochemists are equipped with an impressive arsenal of radioligands to accurately recognise particular receptors of interest. These include key biomarkers of HD: adenosine, cannabinoid, dopaminergic and glutamateric receptors, microglial activation, phosphodiesterase 10 A and synaptic vesicle proteins. This review aims to provide a radiochemical picture of the recent developments in the field of HD PET, with significant attention devoted to radiosynthetic routes towards the tracers relevant to this disease.


Subject(s)
Biomarkers/metabolism , Brain/diagnostic imaging , Huntington Disease/diagnostic imaging , Positron-Emission Tomography , Radiopharmaceuticals , Brain/pathology , Cannabinoid Receptor Agonists/metabolism , Carbon Radioisotopes/chemistry , Dopamine Antagonists/chemical synthesis , Dopamine Antagonists/chemistry , Dopamine Antagonists/metabolism , Excitatory Amino Acid Antagonists/chemical synthesis , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/metabolism , Fluorine Radioisotopes/chemistry , GABA Antagonists/chemical synthesis , GABA Antagonists/chemistry , GABA Antagonists/metabolism , Humans , Huntington Disease/pathology , Microglia/metabolism , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/metabolism , Purinergic P1 Receptor Antagonists/chemical synthesis , Purinergic P1 Receptor Antagonists/chemistry , Purinergic P1 Receptor Antagonists/metabolism , Radiopharmaceuticals/chemistry
14.
J Neurochem ; 153(5): 549-566, 2020 06.
Article in English | MEDLINE | ID: mdl-31821563

ABSTRACT

N-methyl-D-aspartate (NMDA) receptor channels are activated by glutamate (or NMDA) and glycine. The channels also undergo desensitization, which denotes decreased channel availability, after prolonged exposure to the activating ligands. Glycine apparently has a paradoxical negative effect on desensitization, as the increase in ambient glycine in concentrations required for channel activation would increase sustained NMDA receptor currents. We hypothesized that this classical "glycine-dependent desensitization" could be glycine-dependent activation in essence. By performing electrophysiological recordings and biophysical analyses with rat brain NMDA receptors heterogeneously expressed in Xenopus laevis oocytes, we characterized that the channel opened by "only" NMDA (in nominally glycine-free condition probably with the inevitable nanomolar glycine) would undergo a novel form of deactivation rather than desensitization, and is thus fully available for subsequent activation. Moreover, external tetrapentylammonium ions (TPentA), tetrabutylammonium ions, and tetrapropylammonium ions (TPA, in higher concentrations) block the pore and prohibit channel desensitization with a simple "foot-in-the-door" hindrance effect. TpentA and TPA have the same voltage dependence but show different flow dependence in binding affinity, revealing a common binding site at an electrical distance of ~0.7 from the outside yet differential involvement of the flux-coupling region in the external pore mouth. The smaller tetraethylammonium ion and the larger tetrahexylammonium and tetraheptylammonium ions may block the channel but could not affect desensitization. We conclude that NMDA receptor desensitization requires concomitant binding of both glycine and glutamate, and thus movement of both GluN1 and GluN2 subunits. Desensitization gate itself embodies a highly restricted pore reduction with a physical distance of ~4 Å from the charged nitrogen atom of bound tetraalkylammonium ions, and is located very close to the activation gate in the bundle-crossing region in the external pore vestibule.


Subject(s)
Glutamic Acid/metabolism , Glycine/metabolism , Ion Channel Gating/physiology , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Female , Glutamic Acid/pharmacology , Glycine/pharmacology , Ion Channel Gating/drug effects , Ligands , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Protein Binding/drug effects , Protein Binding/physiology , Rats , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Xenopus laevis
15.
ACS Chem Neurosci ; 10(11): 4685-4695, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31622082

ABSTRACT

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.


Subject(s)
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
16.
PLoS Pathog ; 15(1): e1007570, 2019 01.
Article in English | MEDLINE | ID: mdl-30695069

ABSTRACT

Glutamate-gated chloride channel receptors (GluClRs) mediate inhibitory neurotransmission at invertebrate synapses and are primary targets of parasites that impact drastically on agriculture and human health. Ivermectin (IVM) is a broad-spectrum pesticide that binds and potentiates GluClR activity. Resistance to IVM is a major economic and health concern, but the molecular and synaptic mechanisms of resistance are ill-defined. Here we focus on GluClRs of the agricultural endoparasite, Haemonchus contortus. We demonstrate that IVM potentiates inhibitory input by inducing a tonic current that plateaus over 15 minutes and by enhancing post-synaptic current peak amplitude and decay times. We further demonstrate that IVM greatly enhances the active durations of single receptors. These effects are greatly attenuated when endogenous IVM-insensitive subunits are incorporated into GluClRs, suggesting a mechanism of IVM resistance that does not affect glutamate sensitivity. We discovered functional groups of IVM that contribute to tuning its potency at different isoforms and show that the dominant mode of access of IVM is via the cell membrane to the receptor.


Subject(s)
Chloride Channels/metabolism , Haemonchus/drug effects , Ivermectin/pharmacology , Animals , Chloride Channels/antagonists & inhibitors , Excitatory Amino Acid Antagonists/metabolism , Glutamic Acid/pharmacology , HEK293 Cells , Haemonchus/metabolism , Humans , Inhibitory Postsynaptic Potentials/drug effects , Patch-Clamp Techniques/methods , Receptors, Glutamate/metabolism , Xenopus laevis/embryology
17.
Addict Biol ; 24(4): 590-603, 2019 07.
Article in English | MEDLINE | ID: mdl-29667304

ABSTRACT

Recurrent relapse is a major problem in treating opiate addiction. Pavlovian conditioning plays a role in recurrent relapse whereby exposure to cues learned during drug intake can precipitate relapse to drug taking. α7 nicotinic acetylcholine receptors (nAChRs) have been implicated in attentional aspects of cognition and mechanisms of learning and memory. In this study we have investigated the role of α7 nAChRs in morphine-conditioned place preference (morphine-CPP). CPP provides a model of associative learning that is pertinent to associative aspects of drug dependence. The α7 nAChR antagonist methyllycaconitine (MLA; 4 mg/kg s.c.) had no effect on the acquisition, maintenance, reconsolidation or extinction of morphine-CPP but selectively attenuated morphine-primed reinstatement of CPP, in both mice and rats. Reinstatement of morphine-CPP in mice was accompanied by a selective increase in [3 H]-AMPA binding (but not in [3 H]-MK801 binding) in the ventral hippocampus that was prevented by prior treatment with MLA. Administration of MLA (6.7 µg) directly into the ventral hippocampus of rats prior to a systemic priming dose of morphine abolished reinstatement of morphine-CPP, whereas MLA delivered into the dorsal hippocampus or prefrontal cortex was without effect. These results suggest that α7 nAChRs in the ventral hippocampus play a specific role in the retrieval of associative drug memories following a period of extinction, making them potential targets for the prevention of relapse.


Subject(s)
Aconitine/analogs & derivatives , Analgesics, Opioid , Conditioning, Classical/drug effects , Extinction, Psychological/drug effects , Hippocampus/drug effects , Morphine , Nicotinic Antagonists/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Aconitine/pharmacology , Animals , Dizocilpine Maleate/metabolism , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Antagonists/metabolism , Hippocampus/metabolism , Mice , Opioid-Related Disorders , Rats , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Recurrence , Tritium , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
18.
Pharmacol Rep ; 70(6): 1089-1096, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30308459

ABSTRACT

BACKGROUND: Kynurenic acid (KYNA) is a biologically active metabolite of tryptophan exerting action on several receptors located in the brain and periphery. KYNA can be synthesized endogenously or supplied in the diet. It was documented that KYNA is present in various types of food. However, its presence in beverages was not yet investigated. Here, we measured content of KYNA in tea and coffee as well as analyzed distribution and fate of intragastrically administered labelled KYNA in mice. METHODS: 16 and 13 studied samples of tea and coffee, respectively were of commercial origin. Tea and coffee infusions were prepared according to the producers' guidelines. KYNA content in beverages was measured by means of HPLC detection. Adult male mice were used for analysis of fate of intragastrically administered labelled KYNA and collected samples were analyzed using liquid scintillation counter. RESULTS: KYNA was identified in all studied beverages. Amounts of KYNA found in various types of beverages differed significantly. The highest content of KYNA in tea and coffee was 8.7 µg/100 ml and 0.63 µg/100 ml, respectively. It was found that KYNA administered intragastrically as a liquid is absorbed from the digestive system and readily excreted in urine. The atypical kinetics of KYNA distribution were found in intestinal content of cecum, where it appeared later and persisted longer than in other tissues. CONCLUSIONS: Our data show that tea and coffee intake may contribute to KYNA content in the human organism. The distribution pattern of KYNA delivered as a liquid suggests that it either directly affects digestive system's functioning and intestinal microbiome composition, or participates in the whole body pool of KYNA.


Subject(s)
Coffee/metabolism , Kynurenic Acid/administration & dosage , Kynurenic Acid/metabolism , Liver/metabolism , Spleen/metabolism , Tea/metabolism , Animals , Beverages , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Liver/drug effects , Male , Mice , Spleen/drug effects , Tissue Distribution/drug effects , Tissue Distribution/physiology
19.
Neuron ; 99(5): 956-968.e4, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30122377

ABSTRACT

AMPA receptors mediate fast excitatory neurotransmission and are critical for CNS development and function. Calcium-permeable subsets of AMPA receptors are strongly implicated in acute and chronic neurological disorders. However, despite the clinical importance, the therapeutic landscape for specifically targeting them, and not the calcium-impermeable AMPA receptors, remains largely undeveloped. To address this problem, we used cryo-electron microscopy and electrophysiology to investigate the mechanisms by which small-molecule blockers selectively inhibit ion channel conductance in calcium-permeable AMPA receptors. We determined the structures of calcium-permeable GluA2 AMPA receptor complexes with the auxiliary subunit stargazin bound to channel blockers, including the orb weaver spider toxin AgTx-636, the spider toxin analog NASPM, and the adamantane derivative IEM-1460. Our structures provide insights into the architecture of the blocker binding site and the mechanism of trapping, which are critical for development of small molecules that specifically target calcium-permeable AMPA receptors.


Subject(s)
Calcium/metabolism , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Animals , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Receptors, AMPA/antagonists & inhibitors , Spider Venoms/chemistry , Spider Venoms/metabolism , Spider Venoms/pharmacology
20.
Biochem Pharmacol ; 155: 356-365, 2018 09.
Article in English | MEDLINE | ID: mdl-30028996

ABSTRACT

While many orthosteric ligands have been developed for the mGlu2 receptor, little is known about their target binding kinetics and how these relate to those of the endogenous agonist glutamate. Here, the kinetic rate constants, i.e. kon and koff, of glutamate were determined for the first time followed by those of the synthetic agonist LY354740 and antagonist LY341495. To increase the understanding of the binding mechanism and impact of allosteric modulation thereon, kinetic experiments were repeated in the presence of allosteric modulators. Functional assays were performed to further study the interplay between the orthosteric and allosteric binding sites, including an impedance-based morphology assay. We found that dissociation rate constants of orthosteric mGlu2 ligands were all within a small 6-fold range, whereas association rate constants were ranging over more than three orders of magnitude and correlated to both affinity and potency. The latter showed that target engagement of orthosteric mGlu2 ligands is kon-driven in vitro. Moreover, only the off-rates of the two agonists were decreased by a positive allosteric modulator (PAM), thereby increasing their affinity. Interestingly, a PAM increased the duration of a glutamate-induced cellular response. A negative allosteric modulator (NAM) increased both on- and off-rate of glutamate without changing its affinity, while it did not affect these parameters for LY354740, indicating probe-dependency. In conclusion, we found that affinity- or potency-based orthosteric ligand optimization primarily results in ligands with high kon values. Moreover, positive allosteric modulators alter the binding kinetics of orthosteric agonists mainly by decreasing koff, which we were able to correlate to a lengthened cellular response. Together, this study shows the importance of studying binding kinetics in early drug discovery, as this may provide important insights towards improved efficacy in vivo.


Subject(s)
Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Antagonists/metabolism , Glutamic Acid/metabolism , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Amino Acids/metabolism , Amino Acids/pharmacology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Bridged Bicyclo Compounds/metabolism , Bridged Bicyclo Compounds/pharmacology , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , Ligands , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Xanthenes/metabolism , Xanthenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...