92 results on '"Peoples RW"'
Search Results
2. Synaptotoxicity of Alzheimer Beta Amyloid Can Be Explained by Its Membrane Perforating Property
- Author
-
Gendelman, HE, Sepulveda, FJ, Parodi, J, Peoples, RW, Opazo, C, Aguayo, LG, Gendelman, HE, Sepulveda, FJ, Parodi, J, Peoples, RW, Opazo, C, and Aguayo, LG
- Abstract
The mechanisms that induce Alzheimer's disease (AD) are largely unknown thereby deterring the development of disease-modifying therapies. One working hypothesis of AD is that Abeta excess disrupts membranes causing pore formation leading to alterations in ionic homeostasis. However, it is largely unknown if this also occurs in native brain neuronal membranes. Here we show that similar to other pore forming toxins, Abeta induces perforation of neuronal membranes causing an increase in membrane conductance, intracellular calcium and ethidium bromide influx. These data reveal that the target of Abeta is not another membrane protein, but that Abeta itself is the cellular target thereby explaining the failure of current therapies to interfere with the course of AD. We propose that this novel effect of Abeta could be useful for the discovery of anti AD drugs capable of blocking these "Abeta perforates". In addition, we demonstrate that peptides that block Abeta neurotoxicity also slow or prevent the membrane-perforating action of Abeta.
- Published
- 2010
3. A Single phenylalanine residue in the main intracellular loop of α1 γ-aminobutyric acid type A and glycine receptors influences their sensitivity to propofol.
- Author
-
Moraga-Cid G, Yevenes GE, Schmalzing G, Peoples RW, Aguayo LG, Moraga-Cid, Gustavo, Yevenes, Gonzalo E, Schmalzing, Günther, Peoples, Robert W, and Aguayo, Luis G
- Published
- 2011
- Full Text
- View/download PDF
4. Effect of repeated administration of soman on selected endocrine parameters and blood glucose in rats*1
- Author
-
Fletcher Hp, Spratto Gr, Akbar Wj, and Peoples Rw
- Subjects
medicine.medical_specialty ,Insulin ,medicine.medical_treatment ,Toxicology ,Glucagon ,chemistry.chemical_compound ,Epinephrine ,Endocrinology ,chemistry ,Corticosterone ,Internal medicine ,Soman ,Toxicity ,medicine ,Endocrine system ,medicine.drug ,Hormone - Abstract
The effects of repeated administration of soman on plasma glucose levels, acetylcholinesterase (AChE) activity in erythrocytes and hypothalamus, and plasma levels of corticosterone, glucagon, insulin, epinephrine, and norepinephrine were studied in male rats. Rats were given soman subcutaneously (sc), either 30 μg/kg every 24 hr for 5 or 12 days or 40 μg/kg every 24 hr for 5 days. All doses of soman markedly depressed AChE activity in the hypothalamus and completely inhibited AChE activity in erythrocytes. Soman 30 μg/kg given for 5 days did not alter plasma levels of any hormone assayed and produced few signs of intoxication. Soman 40 μg/kg given for 5 days elevated plasma levels of glucose and corticosterone and produced signs of severe cumulative intoxication. Daily administration of 30 μg/kg of soman for 12 days inhibited hypothalamic AChE activity 75%, lowered plasma insulin, and produced signs of moderate intoxication. Repeated administration of soman produced endocrine alterations only when significant signs of intoxication were evident. The absence of increases in plasma levels of catecholamines and corticosterone in rats exhibiting signs of moderate intoxication, and of catecholamines in rats exhibiting signs of severe intoxication, may indicate an impairment by soman of the normal endocrine response to stress.
- Published
- 1988
- Full Text
- View/download PDF
5. Effect of acute soman on selected endocrine parameters and blood glucose in rats*1
- Author
-
Akbar Wj, Fletcher Hp, Peoples Rw, and Spratto Gr
- Subjects
medicine.medical_specialty ,Insulin ,medicine.medical_treatment ,Toxicology ,Glucagon ,Norepinephrine (medication) ,chemistry.chemical_compound ,Epinephrine ,Endocrinology ,chemistry ,Corticosterone ,Internal medicine ,Toxicity ,Soman ,medicine ,sense organs ,Pancreatic hormone ,medicine.drug - Abstract
The effects of acute doses of soman (40, 60, or 80 micrograms/kg sc) in rats were evaluated for toxic symptoms as well as for changes in plasma levels of glucose, insulin, glucagon, corticosterone, norepinephrine, and epinephrine. The relationship between changes in these levels and depressed acetylcholinesterase activity in the hypothalamus was determined. Soman 40 micrograms/kg did not manifest significant changes in any of the parameters evaluated. However, both the 60 and 80 micrograms/kg doses of soman caused dose- and time-related increases in plasma levels of glucose, corticosterone, norepinephrine, epinephrine, and a depression of insulin. Many of these increases, as well as the severity of toxicity, appear to be inversely related to the hypothalamic acetylcholinesterase levels. The hyperglycemia following the higher doses of soman is likely due to the combined effects of elevated levels of corticosterone, catecholamines, possibly glucagon, and depressed insulin levels. Stress from the toxic effects of soman is likely partially responsible for the endocrine effects since most of the changes observed are consistent with changes in these levels that would be manifested in an animal stress model.
- Published
- 1988
- Full Text
- View/download PDF
6. Effects of ethanol on GluN1/GluN2A and GluN1/GluN2B NMDA receptor-ion channel gating kinetics.
- Author
-
Peoples RW and Ren H
- Subjects
- Humans, Ion Channel Gating, HEK293 Cells, Glutamic Acid, Receptors, N-Methyl-D-Aspartate, Ethanol pharmacology
- Abstract
Background: The N-methyl-D-aspartate receptor (NMDAR) is a major molecular target of alcohol action in the central nervous system, yet many aspects of alcohol's modulation of the activity of this ion channel remain unclear. We and others have shown that ethanol inhibition of NMDAR involves alterations in gating, especially a reduction in mean open time. However, a full description of ethanol's effects on NMDAR kinetics, including fitting them to a kinetic model, has not been reported., Methods: To determine ethanol's effects on NMDAR kinetics, we used steady-state single-channel recording in outside-out patches from HEK-293 cells transfected with recombinant GluN1/GluN2A or GluN1/GluN2B NMDAR subunits. Very low glutamate concentrations were used to isolate individual activations of the receptor., Results: In both subunit types, ethanol, at approximate whole-cell IC
50 values (156 mM, GluN2A; 150 mM, GluN2B), reduced open probability (po ) by approximately 50% and decreased mean open time without changing the frequency of opening. Open and shut time distributions exhibited two and five components, respectively; ethanol selectively decreased the time constant and relative proportion of the longer open time component. In the GluN2A subunit, ethanol increased the time constants of all but the longest shut time components, whereas in the GluN2B subunit, shut times were unchanged by ethanol. Fitting of bursts of openings (representing individual activations of the receptor) to the gating portion of a kinetic model revealed that ethanol altered two rates: the rate associated with activation of the GluN2A or GluN2B subunit, and the rate associated with the closing of the longer of the two open states., Conclusions: These results demonstrate that ethanol selectively alters individual kinetic rates and thus appears to selectively affect distinct conformational transitions involved in NMDAR gating., (© 2022 Research Society on Alcoholism.)- Published
- 2022
- Full Text
- View/download PDF
7. Modulatory Actions of the Glycine Receptor β Subunit on the Positive Allosteric Modulation of Ethanol in α2 Containing Receptors.
- Author
-
Muñoz B, Mariqueo T, Murath P, Peters C, Yevenes GE, Moraga-Cid G, Peoples RW, and Aguayo LG
- Abstract
Alpha1-containing glycine receptors (GlyRs) are major mediators of synaptic inhibition in the spinal cord and brain stem. Recent studies reported the presence of α2-containing GlyRs in other brain regions, such as nucleus accumbens and cerebral cortex. GlyR activation decreases neuronal excitability associated with sensorial information, motor control, and respiratory functions; all of which are significantly altered during ethanol intoxication. We evaluated the role of β GlyR subunits and of two basic amino acid residues, K389 and R390, located in the large intracellular loop (IL) of the α2 GlyR subunit, which are important for binding and functional modulation by Gβγ, the dimer of the trimeric G protein conformation, using HEK-293 transfected cells combined with patch clamp electrophysiology. We demonstrate a new modulatory role of the β subunit on ethanol sensitivity of α2 subunits. Specifically, we found a differential allosteric modulation in homomeric α2 GlyRs compared with the α2β heteromeric conformation. Indeed, while α2 was insensitive, α2β GlyRs were substantially potentiated by ethanol, GTP-γ-S, propofol, Zn
2+ and trichloroethanol. Furthermore, a Gβγ scavenger (ct-GRK2) selectively attenuated the effects of ethanol on recombinant α2β GlyRs. Mutations in an α2 GlyR co-expressed with the β subunit (α2AAβ) specifically blocked ethanol sensitivity, but not propofol potentiation. These results show a selective mechanism for low ethanol concentration effects on homomeric and heteromeric conformations of α2 GlyRs and provide a new mechanism for ethanol pharmacology, which is relevant to upper brain regions where α2 GlyRs are abundantly expressed., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Muñoz, Mariqueo, Murath, Peters, Yevenes, Moraga-Cid, Peoples and Aguayo.)- Published
- 2021
- Full Text
- View/download PDF
8. Positions in the N-methyl-D-aspartate Receptor GluN2C Subunit M3 and M4 Domains Regulate Alcohol Sensitivity and Receptor Kinetics.
- Author
-
Wu M, Katti P, Zhao Y, and Peoples RW
- Subjects
- HEK293 Cells, Humans, Mutagenesis, Site-Directed, Receptors, N-Methyl-D-Aspartate genetics, Central Nervous System Depressants adverse effects, Ethanol adverse effects, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors
- Abstract
Background: Alcohol alters synaptic transmission in the brain. The N-methyl-D-aspartate (NMDA) receptor (NMDAR), a subtype of glutamate-gated ion channel, is an important synaptic target of alcohol in the brain. We and others have previously identified 4 alcohol-sensitive positions in the third and fourth membrane-associated (M) domains, designated M3
1-2 and M41-2 , of the GluN1, GluN2A, and GluN2B NMDAR subunits. In the present study, we tested whether the corresponding positions in the GluN2C subunit also regulate alcohol sensitivity and ion channel gating., Methods: We performed alanine- and tryptophan-scanning mutagenesis in the GluN2C subunit followed by expression in HEK 293 cells and electrophysiological patch-clamp recording., Results: Alanine substitution at the M31 (F634) and M41-2 (M821 and M823) positions did not alter ethanol (EtOH) sensitivity, whereas substitution of alanine at the M32 position (F635) yielded nonfunctional receptors. Tryptophan substitution at the M31-2 positions did not change EtOH sensitivity, whereas tryptophan substitution at the M41 position increased, and at the M42 position decreased, EtOH sensitivity. The increased EtOH sensitivity of the tryptophan mutant at M41 is in marked contrast to previous results observed in the GluN2A and GluN2B subunits. In addition, this mutant exhibited increased desensitization, but to a much lesser extent compared to the corresponding mutations in GluN2A and GluN2B. A series of mutations at M41 altered EtOH sensitivity, glutamate potency, and desensitization. Seven amino acid substitutions (of 15 tested) at this position yielded nonfunctional receptors. Among the remaining mutants at M41 , EtOH sensitivity was not significantly correlated with hydrophobicity, molecular volume, or polarity of the substituent, or with glutamate EC50 values, but was correlated with maximal steady-state-to-peak current ratio, a measure of desensitization., Conclusions: The identity and characteristics of alcohol-sensitive positions in the GluN2C subunit differ from those previously reported for GluN2A and GluN2B subunits, despite the high homology among these subunits., (© 2019 by the Research Society on Alcoholism.)- Published
- 2019
- Full Text
- View/download PDF
9. Effect of Cholesterol on Membrane Fluidity and Association of Aβ Oligomers and Subsequent Neuronal Damage: A Double-Edged Sword.
- Author
-
Fernández-Pérez EJ, Sepúlveda FJ, Peters C, Bascuñán D, Riffo-Lepe NO, González-Sanmiguel J, Sánchez SA, Peoples RW, Vicente B, and Aguayo LG
- Abstract
Background: The beta-amyloid peptide (Aβ) involved in Alzheimer's disease (AD) has been described to associate/aggregate on the cell surface disrupting the membrane through pore formation and breakage. However, molecular determinants involved for this interaction (e.g., some physicochemical properties of the cell membrane) are largely unknown. Since cholesterol is an important molecule for membrane structure and fluidity, we examined the effect of varying cholesterol content with the association and membrane perforation by Aβ in cultured hippocampal neurons. Methods: To decrease or increase the levels of cholesterol in the membrane we used methyl-β-cyclodextrin (MβCD) and MβCD/cholesterol, respectively. We analyzed if membrane fluidity was affected using generalized polarization (GP) imaging and the fluorescent dye di-4-ANEPPDHQ. Additionally membrane association and perforation was assessed using immunocytochemistry and electrophysiological techniques, respectively. Results: The results showed that cholesterol removal decreased the macroscopic association of Aβ to neuronal membranes (fluorescent-puncta/20 μm: control = 18 ± 2 vs. MβCD = 10 ± 1, p < 0.05) and induced a facilitation of the membrane perforation by Aβ with respect to control cells (half-time for maximal charge transferred: control = 7.2 vs. MβCD = 4.4). Under this condition, we found an increase in membrane fluidity (46 ± 3.3% decrease in GP value, p < 0.001). On the contrary, increasing cholesterol levels incremented membrane rigidity (38 ± 2.7% increase in GP value, p < 0.001) and enhanced the association and clustering of Aβ (fluorescent-puncta/20 μm: control = 18 ± 2 vs. MβCD = 10 ± 1, p < 0.01), but inhibited membrane disruption. Conclusion: Our results strongly support the significance of plasma membrane organization in the toxic effects of Aβ in hippocampal neurons, since fluidity can regulate distribution and insertion of the Aβ peptide in the neuronal membrane.
- Published
- 2018
- Full Text
- View/download PDF
10. Two adjacent phenylalanines in the NMDA receptor GluN2A subunit M3 domain interactively regulate alcohol sensitivity and ion channel gating.
- Author
-
Ren H, Zhao Y, Wu M, Dwyer DS, and Peoples RW
- Subjects
- Glutamic Acid pharmacology, HEK293 Cells, Humans, Mutation, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits physiology, Receptors, N-Methyl-D-Aspartate genetics, Tryptophan genetics, Ethanol pharmacology, Ion Channel Gating drug effects, Phenylalanine chemistry, Receptors, N-Methyl-D-Aspartate chemistry, Receptors, N-Methyl-D-Aspartate physiology
- Abstract
The N-methyl-d-aspartate (NMDA) receptor is a key target of ethanol action in the central nervous system. Alcohol inhibition of NMDA receptor function involves small clusters of residues in the third and fourth membrane-associated (M) domains. Previous results from this laboratory have shown that two adjacent positions in the M3 domain, F636 and F637, can powerfully regulate alcohol sensitivity and ion channel gating. In this study, we report that these positions interact with one another in the regulation of both NMDA receptor gating and alcohol action. Using dual mutant cycle analysis, we detected interactions among various substitution mutants at these positions with respect to regulation of glutamate EC
50 , steady-state to peak current ratios (Iss :Ip ), mean open time, and ethanol IC50 . This interaction apparently involves a balancing of forces on the M3 helix, such that the disruption of function due to a substitution at one position can be reversed by a similar substitution at the other position. For example, tryptophan substitution at F636 or F637 increased or decreased channel mean open time, respectively, but tryptophan substitution at both positions did not alter open time. Interestingly, the effects of a number of mutations on receptor kinetics and ethanol sensitivity appeared to depend upon subtle structural differences, such as those between the isomeric amino acids leucine and isoleucine, as they could not be explained on the basis of sidechain molecular volume or hydrophilicity., (Copyright © 2016 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
11. Intersubunit interactions at putative sites of ethanol action in the M3 and M4 domains of the NMDA receptor GluN1 and GluN2B subunits.
- Author
-
Zhao Y, Ren H, and Peoples RW
- Subjects
- Cells, Cultured, Dose-Response Relationship, Drug, HEK293 Cells, Humans, Protein Subunits antagonists & inhibitors, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits metabolism, Receptors, N-Methyl-D-Aspartate genetics, Receptors, N-Methyl-D-Aspartate metabolism, Structure-Activity Relationship, Ethanol pharmacology, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors, Receptors, N-Methyl-D-Aspartate chemistry
- Abstract
Background and Purpose: The NMDA receptor is an important target of alcohol action in the brain. Recent studies in this laboratory have demonstrated that alcohol-sensitive positions in the intersubunit interfaces of the M3 and M4 domains of GluN1 and GluN2A subunits interact with respect to ethanol sensitivity and receptor kinetics and that alcohol-sensitive positions in the M domains of GluN2A and GluN2B subunits differ. In this study, we tested for interactions among alcohol-sensitive positions at the M domain intersubunit interfaces in GluN1/GluN2B NMDA receptors., Experimental Approach: We used whole-cell patch-clamp recording in tsA201 cells expressing tryptophan substitution mutants at ethanol-sensitive positions in the GluN1 and GluN2B NMDA receptor subunits to test for interactions among positions., Key Results: Six pairs of positions in GluN1/GluN2B significantly interacted to regulate ethanol inhibition: Gly(638) /Met(824) , Gly(638) /Leu(825) , Phe(639) /Leu(825) , Phe(639) /Gly(826) , Met(818) /Phe(637) and Val(820) /Phe(637) . Tryptophan substitution at Met(824) or Leu(825) in GluN2B did not alter ethanol sensitivity but interacted with positions in the GluN1 M3 domain to regulate ethanol action, whereas tryptophan substitution at Gly(638) , which is the cognate of an ethanol-sensitive position in GluN2A, did not alter ethanol sensitivity or interact with positions in GluN1. Two and three pairs of positions interacted to regulate glutamate steady-state and peak current EC50 , respectively, and one pair interacted with respect to macroscopic desensitization., Conclusions: Despite highly-conserved M domain sequences and similar ethanol sensitivity in the GluN2A and GluN2B subunits, the manner in which these subunits interact with the GluN1 subunit to regulate ethanol sensitivity and receptor kinetics differs., (© 2016 The British Pharmacological Society.)
- Published
- 2016
- Full Text
- View/download PDF
12. The Level of NMDA Receptor in the Membrane Modulates Amyloid-β Association and Perforation.
- Author
-
Peters C, Sepúlveda FJ, Fernández-Pérez EJ, Peoples RW, and Aguayo LG
- Subjects
- 2-Amino-5-phosphonovalerate pharmacology, Animals, Cell Membrane drug effects, Cells, Cultured, Embryo, Mammalian, Excitatory Amino Acid Agonists pharmacology, Excitatory Amino Acid Antagonists pharmacology, Female, HEK293 Cells, Hippocampus cytology, Humans, Membrane Potentials drug effects, Membrane Potentials genetics, Microtubule-Associated Proteins metabolism, N-Methylaspartate pharmacology, Pregnancy, Protein Subunits genetics, Protein Subunits metabolism, Rats, Receptors, N-Methyl-D-Aspartate genetics, Amyloid beta-Peptides pharmacology, Cell Membrane metabolism, Neurons drug effects, Neurons metabolism, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
Alzheimer's disease is a neurodegenerative disorder that affects mostly the elderly. The main histopathological markers are the senile plaques formed by amyloid-β peptide (Aβ) aggregates that can perforate the plasma membrane of cells, increasing the intracellular calcium levels and releasing synaptic vesicles that finally lead to a delayed synaptic failure. Several membrane proteins and lipids interact with Aβ affecting its toxicity in neurons. Here, we focus on NMDA receptors (NMDARs) as proteins that could be modulating the association and neurotoxic perforation induced by Aβ on the plasma membrane. In fact, our results showed that decreasing NMDARs, using enzymatic or siRNA approaches, increased the association of Aβ to the neurons. Furthermore, overexpression of NMDARs also resulted in an enhanced association between NMDA and Aβ. Functionally, the reduction in membrane NMDARs augmented the process of membrane perforation. On the other hand, overexpressing NMDARs had a protective effect because Aβ was now unable to cause membrane perforation, suggesting a complex relationship between Aβ and NMDARs. Because previous studies have recognized that Aβ oligomers are able to increase membrane permeability and produce amyloid pores, the present study supports the conclusion that NMDARs play a critical protective role on Aβ actions in hippocampal neurons. These results could explain the lack of correlation between brain Aβ burden and clinically observed dementia.
- Published
- 2016
- Full Text
- View/download PDF
13. Different sites of alcohol action in the NMDA receptor GluN2A and GluN2B subunits.
- Author
-
Zhao Y, Ren H, Dwyer DS, and Peoples RW
- Subjects
- Amino Acid Sequence, Glutamic Acid metabolism, HEK293 Cells, Humans, Membrane Potentials drug effects, Membrane Potentials physiology, Models, Molecular, Molecular Sequence Data, Mutation, Patch-Clamp Techniques, Receptors, N-Methyl-D-Aspartate genetics, Central Nervous System Depressants pharmacology, Ethanol pharmacology, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
The NMDA receptor is a major target of alcohol action in the CNS, and recent behavioral and cellular studies have pointed to the importance of the GluN2B subunit in alcohol action. We and others have previously characterized four amino acid positions in the third and fourth membrane-associated (M) domains of the NMDA receptor GluN2A subunit that influence both ion channel gating and alcohol sensitivity. In this study, we found that substitution mutations at two of the four corresponding positions in the GluN2B subunit, F637 and G826, influence ethanol sensitivity and ion channel gating. Because position 826 contains a glycine residue in the native protein, we focused our attention on GluN2B(F637). Substitution mutations at GluN2B(F637) significantly altered ethanol IC50 values, glutamate EC50 values for peak (Ip) and steady-state (Iss) current, and steady-state to peak current ratios (Iss:Ip). Changes in apparent glutamate affinity were not due to agonist trapping in desensitized states, as glutamate Iss EC50 values were not correlated with Iss:Ip values. Ethanol sensitivity was correlated with values of both Ip and Iss glutamate EC50, but not with Iss:Ip. Values of ethanol IC50, glutamate EC50, and Iss:Ip for mutants at GluN2B(F637) were highly correlated with the corresponding values for mutants at GluN2A(F636), consistent with similar functional roles of this position in both subunits. These results demonstrate that GluN2B(Phe637) regulates ethanol action and ion channel function of NMDA receptors. However, despite highly conserved M domain sequences, ethanol's actions on GluN2A and GluN2B subunits differ., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
14. Nature of the neurotoxic membrane actions of amyloid-β on hippocampal neurons in Alzheimer's disease.
- Author
-
Sepúlveda FJ, Fierro H, Fernandez E, Castillo C, Peoples RW, Opazo C, and Aguayo LG
- Subjects
- Animals, Calcium metabolism, Calcium Signaling physiology, Cell Membrane pathology, Cell Membrane ultrastructure, HEK293 Cells, Hippocampus metabolism, Humans, Membrane Potentials drug effects, Microscopy, Electron, Scanning Transmission, Patch-Clamp Techniques, Rats, Rats, Sprague-Dawley, Alzheimer Disease etiology, Amyloid beta-Peptides metabolism, Amyloid beta-Peptides toxicity, Cell Membrane drug effects, Cell Membrane metabolism, Gramicidin metabolism, Gramicidin toxicity, Hippocampus cytology, Neurons cytology
- Abstract
The mechanism by which amyloid-β (Aβ) produces brain dysfunction in patients with Alzheimer's disease is largely unknown. According to previous studies, Aβ might share perforating properties with gramicidin, a well-accepted membrane-disrupting peptide. Therefore, we hypothesize that the key steps leading to synaptotoxicity by Aβ and gramicidin involve peptide aggregation, pore formation, and calcium dysregulation. Here, we show that Aβ and gramicidin form aggregates enriched in β-sheet structures using electron microscopy, and Thioflavin and Congo Red staining techniques. Also, we found that Aβ and gramicidin display fairly similar actions in hippocampal cell membranes, i.e. inducing Ca(2+) entry and synaptoxicity characterized by the loss of synaptic proteins and a decrease in neuronal viability. These effects were not observed in a Ca(2+) free solution, indicating that both Aβ and gramicidin induce neurotoxicity by a Ca(2+)-dependent mechanism. Using combined perforated patch clamp and imaging recordings, we found that only Aβ produced a perforation that progressed from a small (Cl(-)-selective pore) to a larger perforation that allowed the entry of fluorescent molecules. Therefore, based on these results, we propose that the perforation at the plasma membrane by Aβ is a dynamic process that is critical in producing neurotoxicity similar to that found in the brains of AD patients., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
15. A novel alcohol-sensitive position in the N-methyl-D-aspartate receptor GluN2A subunit M3 domain regulates agonist affinity and ion channel gating.
- Author
-
Ren H, Zhao Y, Wu M, and Peoples RW
- Subjects
- Amino Acid Sequence, Dose-Response Relationship, Drug, Excitatory Amino Acid Agonists pharmacology, HEK293 Cells, Humans, Ion Channel Gating drug effects, Molecular Sequence Data, Mutation drug effects, Protein Binding physiology, Protein Subunits agonists, Protein Subunits physiology, Ethanol pharmacology, Excitatory Amino Acid Agonists metabolism, Ion Channel Gating physiology, Mutation physiology, Receptors, N-Methyl-D-Aspartate agonists, Receptors, N-Methyl-D-Aspartate physiology
- Abstract
Abundant evidence supports a role for N-methyl-d-aspartate (NMDA) receptor inhibition in the behavioral actions of ethanol, but the underlying molecular mechanisms have not been fully elucidated. We recently found that clusters of five positions in the third and fourth membrane-associated domains (M3 and M4) at the intersubunit interfaces form putative sites of alcohol action. In the present study, we found that one of these positions, NMDA receptor subunit, GluN2A(F636), can strongly regulate ethanol sensitivity, glutamate potency, and apparent desensitization: ethanol IC50 values, peak (Ip) and steady-state (Iss) glutamate EC50 values, and steady-state to peak current ratio (Iss:Ip) values differed significantly among the mutants tested. Changes in glutamate affinity among the various mutants were not attributable to agonist trapping due to desensitization, as glutamate peak EC50 values were correlated with values of both steady-state EC50 and Iss:Ip. The mean open times determined in selected mutants could be altered up to 4-fold but did not account for the changes in ethanol sensitivity. Ethanol sensitivity was significantly correlated with glutamate EC50 and Iss:Ip values, but the changes in ethanol IC50 among mutants at this position do not appear to be secondary to changes in ion channel kinetics. Substitution of the isomeric amino acids leucine and isoleucine had markedly different effects on ethanol sensitivity, agonist potency, and desensitization, which is consistent with a stringent structural requirement for ion channel modulation by the side chain at this position. Our results indicate that GluN2A(F636) plays an important role in both channel function and ethanol inhibition in NMDA receptors.
- Published
- 2013
- Full Text
- View/download PDF
16. Conserved extracellular cysteines differentially regulate the potentiation produced by Zn2+ in rat P2X4 receptors.
- Author
-
Li CY, Xiong KM, Wu YX, Liu YW, Chen L, Stewart RR, Peoples RW, and Yi CL
- Subjects
- Amino Acid Sequence, Animals, Conserved Sequence, Cysteine chemistry, Disulfides chemistry, Dose-Response Relationship, Drug, Female, Oocytes, Point Mutation, Rats, Receptors, Purinergic P2X4 chemistry, Receptors, Purinergic P2X4 genetics, Species Specificity, Xenopus laevis, Adenosine Triphosphate administration & dosage, Cysteine metabolism, Receptors, Purinergic P2X4 metabolism, Zinc pharmacology
- Abstract
One feature of the amino acid sequence of P2X receptors identified from mammalian species, Xenopus laevis and zebrafish is the conservation of ten cysteines in the extracellular loop. Little information is available about the role of these conserved ectodomain cysteines in the function of P2X receptors. Here, we investigated the possibility that ten conserved cysteine residues in the extracellular loop of the rat P2X4 receptor may regulate zinc potentiation of the receptor using a series of individual cysteine to alanine point mutations and functional characterization of recombinant receptors expressed in Xenopus oocytes. For the C116A, C132A, C159A, C165A, C217A and C227A mutants, 10 µM zinc did not significantly affect the current activated by an EC40 concentration of ATP. By contrast, 5 µM zinc shifted the ATP concentration-response curve to the right in a parallel manner for both the C261A and C270A mutants and the magnitudes of those shifts were similar to that of the wildtype receptor. Interestingly, for the C126A and C149A mutants, 5µM zinc potentiated ATP-activated current, but increased the maximal response to ATP by 90% and 81% respectively, without significantly changing the EC50 value of ATP. Thus, these results suggest that cysteines and disulfide bonds between cysteines are differentially involved in the potentiation of the rat P2X4 receptor by zinc., (Copyright © 2013. Published by Elsevier B.V.)
- Published
- 2013
- Full Text
- View/download PDF
17. Interactions among positions in the third and fourth membrane-associated domains at the intersubunit interface of the N-methyl-D-aspartate receptor forming sites of alcohol action.
- Author
-
Ren H, Zhao Y, Dwyer DS, and Peoples RW
- Subjects
- Amino Acid Substitution, Binding Sites, Central Nervous System Depressants metabolism, Ethanol metabolism, HEK293 Cells, Humans, Mutation, Missense, Protein Binding, Protein Structure, Tertiary, Protein Subunits genetics, Receptors, N-Methyl-D-Aspartate genetics, Central Nervous System Depressants pharmacology, Ethanol pharmacology, Protein Subunits metabolism, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
The N-methyl-D-aspartate (NMDA) glutamate receptor is a major target of ethanol in the brain. Previous studies have identified positions in the third and fourth membrane-associated (M) domains of the NMDA receptor GluN1 and GluN2A subunits that influence alcohol sensitivity. The predicted structure of the NMDA receptor, based on that of the related GluA2 subunit, indicates a close apposition of the alcohol-sensitive positions in M3 and M4 between the two subunit types. We tested the hypothesis that these positions interact to regulate receptor kinetics and ethanol sensitivity by using dual substitution mutants. In single-substitution mutants, we found that a position in both subunits adjacent to one previously identified, GluN1(Gly-638) and GluN2A(Phe-636), can strongly regulate ethanol sensitivity. Significant interactions affecting ethanol inhibition and receptor deactivation were observed at four pairs of positions in GluN1/GluN2A: Gly-638/Met-823, Phe-639/Leu-824, Met-818/Phe-636, and Leu-819/Phe-637; the latter pair also interacted with respect to desensitization. Two interactions involved a position in M4 of both subunits, GluN1(Met-818) and GluN2A(Leu-824), that does not by itself alter ethanol sensitivity, whereas a previously identified ethanol-sensitive position, GluN2A(Ala-825), did not unequivocally interact with any other position tested. These results also indicate a shift by one position of the predicted alignment of the GluN1 M4 domain. These findings have allowed for the refinement of the NMDA receptor M domain structure, demonstrate that this region can influence apparent agonist affinity, and support the existence of four sites of alcohol action on the NMDA receptor, each consisting of five amino acids at the M3-M4 domain intersubunit interfaces.
- Published
- 2012
- Full Text
- View/download PDF
18. Ethanol is a fast channel inhibitor of P2X4 receptors.
- Author
-
Ostrovskaya O, Asatryan L, Wyatt L, Popova M, Li K, Peoples RW, Alkana RL, and Davies DL
- Subjects
- Animals, Cells, Cultured, Female, HEK293 Cells, Humans, Pregnancy, Rats, Rats, Sprague-Dawley, Receptors, Purinergic P2X4 physiology, Time Factors, Ethanol pharmacology, Ion Channel Gating drug effects, Ion Channel Gating physiology, Purinergic P2X Receptor Antagonists pharmacology, Receptors, Purinergic P2X4 metabolism
- Abstract
P2X receptors (P2XRs) are ion channels gated by synaptically released ATP. The P2X4 is the most abundant P2XR subtype expressed in the central nervous system and to date is the most ethanol-sensitive. In addition, genomic findings suggest that P2X4Rs may play a role in alcohol intake/preference. However, little is known regarding how ethanol causes the inhibition of ATP-gated currents in P2X4Rs. We begin to address this issue by investigating the effects of ethanol in wild-type and mutant D331A and M336A P2X4Rs expressed in human embryonic kidney (HEK) 293 cells using whole-cell patch-clamp methods. The results suggest that residues D331 and M336 play a role in P2X4R gating and ethanol inhibits channel functioning via a mechanism different from that in other P2XRs. Key findings from the study include: 1) ethanol inhibits ATP-gated currents in a rapid manner; 2) ethanol inhibition of ATP-gated currents does not depend on voltage and ATP concentration; 3) residues 331 and 336 slow P2X4 current deactivation and regulate the inhibitory effects of ethanol; and 4) ethanol effects are similar in HEK293 cells transfected with P2X4Rs and cultured rat hippocampal neurons transduced with P2X4Rs using a recombinant lentiviral system. Overall, these findings provide key information regarding the mechanism of ethanol action on ATP-gated currents in P2X4Rs and provide new insights into the biophysical properties of P2X4Rs.
- Published
- 2011
- Full Text
- View/download PDF
19. Molecular requirements for ethanol differential allosteric modulation of glycine receptors based on selective Gbetagamma modulation.
- Author
-
Yevenes GE, Moraga-Cid G, Avila A, Guzmán L, Figueroa M, Peoples RW, and Aguayo LG
- Subjects
- Allosteric Regulation drug effects, Dose-Response Relationship, Drug, Humans, Receptors, Glycine genetics, Central Nervous System Depressants pharmacology, Ethanol pharmacology, Receptors, Glycine metabolism
- Abstract
It is now believed that the allosteric modulation produced by ethanol in glycine receptors (GlyRs) depends on alcohol binding to discrete sites within the protein structure. Thus, the differential ethanol sensitivity of diverse GlyR isoforms and mutants was explained by the presence of specific residues in putative alcohol pockets. Here, we demonstrate that ethanol sensitivity in two ligand-gated ion receptor members, the GlyR adult α(1) and embryonic α(2) subunits, can be modified through selective mutations that rescued or impaired Gβγ modulation. Even though both isoforms were able to physically interact with Gβγ, only the α(1) GlyR was functionally modulated by Gβγ and pharmacological ethanol concentrations. Remarkably, the simultaneous switching of two transmembrane and a single extracellular residue in α(2) GlyRs was enough to generate GlyRs modulated by Gβγ and low ethanol concentrations. Interestingly, although we found that these TM residues were different to those in the alcohol binding site, the extracellular residue was recently implicated in conformational changes important to generate a pre-open-activated state that precedes ion channel gating. Thus, these results support the idea that the differential ethanol sensitivity of these two GlyR isoforms rests on conformational changes in transmembrane and extracellular residues within the ion channel structure rather than in differences in alcohol binding pockets. Our results describe the molecular basis for the differential ethanol sensitivity of two ligand-gated ion receptor members based on selective Gβγ modulation and provide a new mechanistic framework for allosteric modulations of abuse drugs.
- Published
- 2010
- Full Text
- View/download PDF
20. Synaptotoxicity of Alzheimer beta amyloid can be explained by its membrane perforating property.
- Author
-
Sepulveda FJ, Parodi J, Peoples RW, Opazo C, and Aguayo LG
- Subjects
- Alzheimer Disease drug therapy, Amyloid beta-Peptides metabolism, Animals, Blotting, Western, Calcium metabolism, Cells, Cultured, Female, Humans, Immunohistochemistry, Mice, Mice, Inbred C57BL, Peptides pharmacology, Peptides therapeutic use, Pregnancy, Rats, Rats, Sprague-Dawley, Alzheimer Disease metabolism, Amyloid beta-Peptides pharmacology, Cell Membrane drug effects, Cell Membrane metabolism, Neurons drug effects, Neurons metabolism
- Abstract
The mechanisms that induce Alzheimer's disease (AD) are largely unknown thereby deterring the development of disease-modifying therapies. One working hypothesis of AD is that Abeta excess disrupts membranes causing pore formation leading to alterations in ionic homeostasis. However, it is largely unknown if this also occurs in native brain neuronal membranes. Here we show that similar to other pore forming toxins, Abeta induces perforation of neuronal membranes causing an increase in membrane conductance, intracellular calcium and ethidium bromide influx. These data reveal that the target of Abeta is not another membrane protein, but that Abeta itself is the cellular target thereby explaining the failure of current therapies to interfere with the course of AD. We propose that this novel effect of Abeta could be useful for the discovery of anti AD drugs capable of blocking these "Abeta perforates". In addition, we demonstrate that peptides that block Abeta neurotoxicity also slow or prevent the membrane-perforating action of Abeta.
- Published
- 2010
- Full Text
- View/download PDF
21. Pathologically activated neuroprotection via uncompetitive blockade of N-methyl-D-aspartate receptors with fast off-rate by novel multifunctional dimer bis(propyl)-cognitin.
- Author
-
Luo J, Li W, Zhao Y, Fu H, Ma DL, Tang J, Li C, Peoples RW, Li F, Wang Q, Huang P, Xia J, Pang Y, and Han Y
- Subjects
- Animals, Binding Sites, Binding, Competitive, Cell Line, Cell Survival drug effects, Cells, Cultured, Dimerization, Excitatory Amino Acid Agonists pharmacology, Excitatory Amino Acid Antagonists chemistry, Humans, Kinetics, Male, Membrane Potentials drug effects, Models, Molecular, Neurons metabolism, Neurons physiology, Neuroprotective Agents chemistry, Patch-Clamp Techniques, Radioligand Assay, Rats, Rats, Sprague-Dawley, Receptors, N-Methyl-D-Aspartate agonists, Receptors, N-Methyl-D-Aspartate genetics, Tacrine chemistry, Tacrine metabolism, Tacrine pharmacology, Transfection, alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid pharmacology, Excitatory Amino Acid Antagonists pharmacology, Neurons drug effects, Neuroprotective Agents pharmacology, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors, Tacrine analogs & derivatives
- Abstract
Uncompetitive N-methyl-d-aspartate (NMDA) receptor antagonists with fast off-rate (UFO) may represent promising drug candidates for various neurodegenerative disorders. In this study, we report that bis(propyl)-cognitin, a novel dimeric acetylcholinesterase inhibitor and gamma-aminobutyric acid subtype A receptor antagonist, is such an antagonist of NMDA receptors. In cultured rat hippocampal neurons, we demonstrated that bis(propyl)-cognitin voltage-dependently, selectively, and moderately inhibited NMDA-activated currents. The inhibitory effects of bis(propyl)-cognitin increased with the rise in NMDA and glycine concentrations. Kinetics analysis showed that the inhibition was of fast onset and offset with an off-rate time constant of 1.9 s. Molecular docking simulations showed moderate hydrophobic interaction between bis(propyl)-cognitin and the MK-801 binding region in the ion channel pore of the NMDA receptor. Bis(propyl)-cognitin was further found to compete with [(3)H]MK-801 with a K(i) value of 0.27 mum, and the mutation of NR1(N616R) significantly reduced its inhibitory potency. Under glutamate-mediated pathological conditions, bis(propyl)-cognitin, in contrast to bis(heptyl)-cognitin, prevented excitotoxicity with increasing effectiveness against escalating levels of glutamate and much more effectively protected against middle cerebral artery occlusion-induced brain damage than did memantine. More interestingly, under NMDA receptor-mediated physiological conditions, bis(propyl)-cognitin enhanced long-term potentiation in hippocampal slices, whereas MK-801 reduced and memantine did not alter this process. These results suggest that bis(propyl)-cognitin is a UFO antagonist of NMDA receptors with moderate affinity, which may provide a pathologically activated therapy for various neurodegenerative disorders associated with NMDA receptor dysregulation.
- Published
- 2010
- Full Text
- View/download PDF
22. Mechanism of bis(7)-tacrine inhibition of GABA-activated current in cultured rat hippocampal neurons.
- Author
-
Zhou L, Liu YW, Peoples RW, Yang M, Tian X, Ai YX, Pang YP, Li ZW, Han YF, and Li CY
- Subjects
- Analysis of Variance, Animals, Biophysics, Cells, Cultured, Dose-Response Relationship, Drug, Drug Interactions, Electric Stimulation methods, Embryo, Mammalian, Membrane Potentials drug effects, Membrane Potentials physiology, Neural Inhibition drug effects, Patch-Clamp Techniques methods, Rats, Rats, Sprague-Dawley, Tacrine pharmacology, Enzyme Inhibitors pharmacology, Hippocampus cytology, Neurons drug effects, Tacrine analogs & derivatives, gamma-Aminobutyric Acid pharmacology
- Abstract
Bis(7)-tacrine is a novel dimeric acetylcholinesterase inhibitor derived from tacrine that shows promise for the treatment of Alzheimer's disease. We have previously reported that bis(7)-tacrine inhibits GABA(A) receptors. In the present study we investigated the mechanism of bis(7)-tacrine inhibition of GABA(A) receptor function using whole-cell patch-clamp recording in cultured rat hippocampal neurons. Bis(7)-tacrine produced a gradual decline of GABA-activated current to a steady-state, but this was not an indication of use-dependence, as the gradually declining component could be eliminated by exposure to bis(7)-tacrine prior to GABA application. In addition, bis(7)-tacrine inhibition did not require the presence of agonist, and GABA-activated current recovered completely from inhibition by bis(7)-tacrine in the absence of agonist. The slow onset of inhibition by bis(7)-tacrine was not apparently due to an action at an intracellular site, as inclusion of 25 microM bis(7)-tacrine in the recording pipette did not alter inhibition by bis(7)-tacrine applied externally. Bis(7)-tacrine shifted the GABA concentration-response curve to the right in a parallel manner and the pA(2) value estimated from a Schild plot was 5.7. Bis(7)-tacrine increased the time constant of activation of GABA-gated ion channels without affecting the time constants of deactivation or desensitization. These results suggest that bis(7)-tacrine is a competitive GABA(A) receptor antagonist with slow onset and offset kinetics. The competitive inhibition of GABA receptors by bis(7)-tacrine could contribute to its ability to enhance memory.
- Published
- 2009
- Full Text
- View/download PDF
23. Conserved extracellular cysteines differentially regulate the inhibitory effect of ethanol in rat P2X4 receptors.
- Author
-
Yi CL, Liu YW, Xiong KM, Stewart RR, Peoples RW, Tian X, Zhou L, Ai YX, Li ZW, Wang QW, and Li CY
- Subjects
- Amino Acid Sequence, Animals, Conserved Sequence, Cysteine genetics, Mutation, Rats, Receptors, Purinergic P2 genetics, Receptors, Purinergic P2X4, Xenopus, Cysteine metabolism, Ethanol toxicity, Purinergic P2 Receptor Antagonists, Receptors, Purinergic P2 metabolism
- Abstract
Relatively little information is available about the molecular mechanism of ethanol inhibition of P2X receptors. Here, we investigated the possibility that 10 conserved cysteine residues in the extracellular loop of the rat P2X4 receptor may regulate ethanol inhibition of the receptor using a series of individual cysteine to alanine point mutations. Each of the mutated receptors generated robust inward current in response to ATP and the mutations produced less than a sixfold change in the ATP EC50 value. For the C116A, C126A, C149A, and C165A mutants, 100 mM ethanol did not significantly affect the current activated by an EC40 concentration of ATP. By contrast, for the C261A and C270A mutants, ethanol inhibited ATP-activated current in a competitive manner similar to that for the wild-type receptor. Interestingly, for the C132A, C159A, C217A, and C227A mutants, ethanol inhibited ATP-activated current, but decreased the maximal response to ATP by 70-75% without significantly changing the EC50 value of ATP, thus exhibiting a noncompetitive-type inhibition. The results suggest that cysteines and disulfide bonds between cysteines are differentially involved in the inhibition of the rat P2X4 receptor by ethanol.
- Published
- 2009
- Full Text
- View/download PDF
24. A selective G betagamma-linked intracellular mechanism for modulation of a ligand-gated ion channel by ethanol.
- Author
-
Yevenes GE, Moraga-Cid G, Peoples RW, Schmalzing G, and Aguayo LG
- Subjects
- Humans, Ligands, Receptors, Glycine agonists, Ethanol pharmacology, GTP-Binding Protein beta Subunits physiology, GTP-Binding Protein gamma Subunits physiology, Ion Channel Gating drug effects, Ion Channels drug effects
- Abstract
The current understanding about ethanol effects on the ligand-gated ion channel (LGIC) superfamily has been restricted to identify potential binding sites within transmembrane (TM) domains in the Cys-loop family. Here, we demonstrate a key role of the TM3-4 intracellular loop and G betagamma signaling for potentiation of glycine receptors (GlyRs) by ethanol. We discovered 2 motifs within the large intracellular loop of the GlyR alpha(1) subunit that are critical for the actions of pharmacological concentrations of ethanol. Significantly, the sites were ethanol-specific because they did not alter the sensitivity to general anesthetics, neurosteroids, or longer n-alcohols. Furthermore, G betagamma scavengers selectively attenuated the ethanol effects on recombinant and native neuronal GlyRs. These results show a selective mechanism for low-ethanol concentration effects on the GlyR and provide a mechanism on ethanol pharmacology, which may be applicable to other LGIC members. Moreover, these data provide an opportunity to develop new genetically modified animal models and novel drugs to treat alcohol-related medical concerns.
- Published
- 2008
- Full Text
- View/download PDF
25. Inhibition of NMDA-gated ion channels by bis(7)-tacrine: whole-cell and single-channel studies.
- Author
-
Liu YW, Luo JL, Ren H, Peoples RW, Ai YX, Liu LJ, Pang YP, Li ZW, Han YF, and Li CY
- Subjects
- Animals, Cells, Cultured, Dose-Response Relationship, Drug, Dose-Response Relationship, Radiation, Drug Interactions, Electric Stimulation methods, Embryo, Mammalian, Hippocampus cytology, Ion Channel Gating physiology, Membrane Potentials drug effects, Membrane Potentials physiology, Membrane Potentials radiation effects, Neurons drug effects, Neurons physiology, Patch-Clamp Techniques, Rats, Rats, Sprague-Dawley, Receptors, N-Methyl-D-Aspartate genetics, Tacrine pharmacology, Transfection, Enzyme Inhibitors pharmacology, Excitatory Amino Acid Agonists pharmacology, Ion Channel Gating drug effects, N-Methylaspartate pharmacology, Receptors, N-Methyl-D-Aspartate physiology, Tacrine analogs & derivatives
- Abstract
Bis(7)-tacrine is a novel dimeric acetylcholinesterase inhibitor derived from tacrine, and has been proposed as a promising agent to treat Alzheimer's disease. We have recently reported that bis(7)-tacrine prevents glutamate-induced neuronal apoptosis by antagonizing NMDA receptors. The purpose of this study was to characterize bis(7)-tacrine inhibition of NMDA-activated current by using patch-clamp recording techniques. In cultured rat hippocampal neurons, bis(7)-tacrine inhibited NMDA-activated whole-cell current in a concentration-dependent manner with an IC(50) of 0.66+/-0.07 microM. Bis(7)-tacrine produced a gradual decline of NMDA-activated current to a steady-state, but this was not an indication of use-dependence. Also, the slow onset of inhibition by bis(7)-tacrine was not apparently due to an action at an intracellular site. Bis(7)-tacrine, 0.5 microM, decreased the maximal response to NMDA by 40% without changing its EC(50). Bis(7)-tacrine inhibition of NMDA-activated current was not voltage-dependent, and was independent of glycine concentration. Results of single-channel experiments obtained from cells expressing NR1 and NR2A subunits revealed that bis(7)-tacrine decreased the open probability and frequency of channel opening, but did not significantly alter the mean open time or introduce rapid closures. These results suggest that bis(7)-tacrine can inhibit NMDA receptor function in a manner that is slow in onset and offset and noncompetitive with respect to both NMDA and glycine. The noncompetitive inhibition of NMDA receptors by bis(7)-tacrine could contribute to its protective effect against glutamate-induced neurotoxicity.
- Published
- 2008
- Full Text
- View/download PDF
26. Bis(7)-tacrine prevents glutamate-induced excitotoxicity more potently than memantine by selectively inhibiting NMDA receptors.
- Author
-
Liu YW, Li CY, Luo JL, Li WM, Fu HJ, Lao YZ, Liu LJ, Pang YP, Chang DC, Li ZW, Peoples RW, Ai YX, and Han YF
- Subjects
- Animals, Calcium analysis, Calcium metabolism, Cells, Cultured, Cerebral Cortex cytology, Excitatory Amino Acid Agonists pharmacology, Glutamic Acid toxicity, Kainic Acid pharmacology, Rats, Rats, Sprague-Dawley, Tacrine pharmacology, alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid pharmacology, Cerebral Cortex drug effects, Cytoprotection, Excitatory Amino Acid Antagonists pharmacology, Memantine pharmacology, Neurons drug effects, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors, Tacrine analogs & derivatives
- Abstract
We have recently reported that bis(7)-tacrine could prevent glutamate-induced neuronal apoptosis through NMDA receptors. In this study, we demonstrated that in cultured rat cortical neurons, bis(7)-tacrine (IC(50), 0.02 microM) prevented glutamate-induced excitotoxicity more substantially than memantine (IC(50), 0.7 microM). In addition, bis(7)-tacrine was more efficient than memantine in buffering the intracellular Ca(2+) triggered by glutamate. In cultured rat hippocampal neurons, bis(7)-tacrine inhibited 50 microM NMDA-activated current in a concentration-dependent manner with an IC(50) of 0.68+/-0.07 microM, which is five times more potent than that produced by memantine (IC(50), 3.41+/-0.36 microM; p<0.05). By contrast, bis(7)-tacrine, up to 5 microM, did not significantly affect the current activated by 50 microM AMPA or 50 microM kainate. These results suggest that bis(7)-tacrine is more potent than memantine against glutamate-induced neurotoxicity by selectively inhibiting NMDA-activated current.
- Published
- 2008
- Full Text
- View/download PDF
27. Functional interactions of alcohol-sensitive sites in the N-methyl-D-aspartate receptor M3 and M4 domains.
- Author
-
Ren H, Salous AK, Paul JM, Lamb KA, Dwyer DS, and Peoples RW
- Subjects
- Cell Line, Computer Simulation, Electrophysiology, Ethanol chemistry, Ethanol pharmacology, Glutamic Acid metabolism, Humans, Models, Molecular, Mutation genetics, Patch-Clamp Techniques, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Structure, Tertiary, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits metabolism, Receptors, N-Methyl-D-Aspartate genetics, Sensitivity and Specificity, Ethanol metabolism, Receptors, N-Methyl-D-Aspartate chemistry, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
The N-methyl-D-aspartate receptor is an important mediator of the behavioral effects of ethanol in the central nervous system. Previous studies have demonstrated sites in the third and fourth membrane-associated (M) domains of the N-methyl-D-aspartate receptor NR2A subunit that influence alcohol sensitivity and ion channel gating. We investigated whether two of these sites, Phe-637 in M3 and Met-823 in M4, interactively regulate the ethanol sensitivity of the receptor by testing dual substitution mutants at these positions. A majority of the mutations decreased steady-state glutamate EC(50) values and maximal steady-state to peak current ratios (I(ss)/I(p)), whereas only two mutations altered peak glutamate EC(50) values. Steady-state glutamate EC(50) values were correlated with maximal glutamate I(ss)/I(p) values, suggesting that changes in glutamate potency were attributable to changes in desensitization. In addition, there was a significant interaction between the substituents at positions 637 and 823 with respect to glutamate potency and desensitization. IC(50) values for ethanol among the mutants varied over the approximate range 100-325 mm. The sites in M3 and M4 significantly interacted in regulating ethanol sensitivity, although this was apparently dependent upon the presence of methionine in position 823. Molecular dynamics simulations of the NR2A subunit revealed possible binding sites for ethanol near both positions in the M domains. Consistent with this finding, the sum of the molecular volumes of the substituents at the two positions was not correlated with ethanol IC(50) values. Thus, there is a functional interaction between Phe-637 and Met-823 with respect to glutamate potency, desensitization, and ethanol sensitivity, but the two positions do not appear to form a unitary site of alcohol action.
- Published
- 2008
- Full Text
- View/download PDF
28. The 5-HT3B subunit confers spontaneous channel opening and altered ligand properties of the 5-HT3 receptor.
- Author
-
Hu XQ and Peoples RW
- Subjects
- Cell Line, Cell Physiological Phenomena, DNA, Complementary metabolism, Humans, Indoles pharmacology, Kinetics, Ligands, Models, Biological, Models, Chemical, Patch-Clamp Techniques, Protein Binding, Protein Conformation, Transfection, Receptors, Serotonin, 5-HT3 chemistry, Receptors, Serotonin, 5-HT3 metabolism
- Abstract
Current receptor theory suggests that there is an equilibrium between the inactive (R) and active (R*) conformations of ligand-gated ion channels and G protein-coupled receptors. The actions of ligands in both receptor types could be appropriately explained by this two-state model. Ligands such as agonists and antagonists affect receptor function by stabilizing one or both conformations. The 5-HT3 receptor is a member of the Cys-loop ligand-gated ion channel superfamily participating in synaptic transmission. Here we show that co-expression of the 5-HT3A and 5-HT3B receptor subunits in the human embryonic kidney (HEK) 293 cells results in a receptor that displays a low level of constitutive (or agonist-independent) activity. Furthermore, we also demonstrate that the properties of ligands can be modified by receptor composition. Whereas the 5-hydroxytryptamine (5-HT) analog 5-methoxyindole is a partial agonist at the 5-HT3A receptor, it becomes a "protean agonist" (functioning as an agonist and an inverse agonist at the same receptor) at the 5-HT3AB receptor (after the Greek god Proteus, who was able to change his shape and appearance at will). In addition, the 5-HT analog 5-hydroxyindole is a positive allosteric modulator for the liganded active (AR*) conformation of the 5-HT3A and 5-HT3AB receptors and a negative allosteric modulator for the spontaneously active (R*) conformation of the 5-HT3AB receptor, suggesting that the spontaneously active (R*) and liganded active (AR*) conformations are differentially modulated by 5-hydroxyindole. Thus, the incorporation of the 5-HT3B subunit leads to spontaneous channel opening and altered ligand properties.
- Published
- 2008
- Full Text
- View/download PDF
29. Arginine 246 of the pretransmembrane domain 1 region alters 2,2,2-trichloroethanol action in the 5-hydroxytryptamine3A receptor.
- Author
-
Hu XQ and Peoples RW
- Subjects
- Allosteric Regulation drug effects, Allosteric Regulation physiology, Amino Acid Sequence drug effects, Amino Acid Sequence genetics, Animals, Arginine chemistry, Cell Line, Ethylene Chlorohydrin pharmacology, Humans, Mice, Molecular Sequence Data, Point Mutation drug effects, Protein Structure, Tertiary drug effects, Protein Structure, Tertiary genetics, Receptors, Serotonin, 5-HT3 chemistry, Serotonin 5-HT3 Receptor Agonists, Arginine genetics, Ethylene Chlorohydrin analogs & derivatives, Point Mutation genetics, Receptors, Serotonin, 5-HT3 genetics
- Abstract
Ligand-gated ion channels participate in synaptic transmission, and they are involved in neurotransmitter release. The functions of the channels are regulated by a variety of modulators. The interaction of 2,2,2-trichloroethanol, the active hypnotic metabolite of chloral hydrate, with the 5-hydroxytryptamine (5-HT) (serotonin) type 3 receptor results in a positive allosteric modulation. We have demonstrated previously that arginine 246 (R246) located in the pretransmembrane domain 1 is critical for coupling agonist binding to gating. In this study, we examined the role of R246 in the action of trichloroethanol with a combination of mutagenesis and whole-cell patch-clamp techniques. The R246A mutation converted the partial agonist dopamine into a full agonist at the 5-HT(3A) receptor, and it facilitated activation of the mutant receptor by dopamine, suggesting an enhanced gating process due to the mutation. The positive modulation of the 5-HT(3A) receptor by trichloroethanol was dramatically reduced by the R246A mutation. Trichloroethanol had little agonist activity in the wild-type receptor (<1% of maximal 5-HT response). However, the R246A mutation significantly increased the direct activation of the receptor by trichloroethanol in the absence of agonist ( approximately 10% of maximal 5-HT response). The current activated by trichloroethanol could be blocked by the competitive 5-HT(3) receptor antagonist tropanyl 3,5-dichlorobenzoate (MDL 72222), and it had a similar reversal potential to those of current activated by 5-HT. In addition, predesensitization of the mutant receptor by trichloroethanol prevented 5-HT from activating the receptor. These data suggest that R246 is a crucial site for mediating the actions of both agonists and modulators.
- Published
- 2008
- Full Text
- View/download PDF
30. Modulation of 5-HT3 receptor desensitization by the light chain of microtubule-associated protein 1B expressed in HEK 293 cells.
- Author
-
Sun H, Hu XQ, Emerit MB, Schoenebeck JC, Kimmel CE, Peoples RW, Miko A, and Zhang L
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Apoptosis Regulatory Proteins, Cell Line, Cell Membrane metabolism, Cells, Cultured, Female, Hippocampus cytology, Humans, Kidney cytology, Kidney embryology, Microtubules drug effects, Microtubules metabolism, Neurons cytology, Neurons metabolism, Nocodazole pharmacology, Oligonucleotides, Antisense pharmacology, Rats, Rats, Sprague-Dawley, Receptors, Serotonin, 5-HT3 drug effects, Tubulin Modulators pharmacology, Kidney metabolism, Microtubule-Associated Proteins metabolism, Nerve Tissue Proteins metabolism, Receptors, Serotonin, 5-HT3 metabolism
- Abstract
Regulation of ligand-gated ion channel (LGIC) function and trafficking by cytoskeleton proteins has been the topic of recent research. Here, we report that the light chain (LC1) of microtubule-associated protein 1B (MAP1B) specifically interacted with the 5-HT(3A) receptor, a predominant serotonin-gated ion channel in the brain. LC1 and 5-HT(3A) receptors were colocalized in central neurons and in HEK 293 cells expressing 5-HT(3A) receptors. LC1 reduced the steady-state density of 5-HT(3A) receptors at the membrane surface of HEK 293 cells and significantly accelerated receptor desensitization time constants from 3.8 +/- 0.3 s to 0.8 +/- 0.1 s. However, LC1 did not significantly alter agonist binding affinity and single-channel conductance of 5-HT(3A) receptors. On the other hand, application of specific LC1 antisense oligonucleotides and nocodazole, a microtubule disruptor, significantly prolonged the desensitization time of the recombinant and native neuronal 5-HT(3) receptors by 3- to 6-fold. This kinetic change induced by nocodazole was completely rescued by addition of LC1 but not GABA(A) receptor-associated protein (GABARAP), suggesting that LC1 can specifically interact with 5-HT(3A) receptors. These observations suggest that the LC1-5-HT(3A) receptor interaction contributes to a mechanism that regulates receptor desensitization kinetics. Such dynamic regulation may play a role in reshaping the efficacy of 5-HT(3) receptor-mediated synaptic transmission.
- Published
- 2008
- Full Text
- View/download PDF
31. Mutations at F637 in the NMDA receptor NR2A subunit M3 domain influence agonist potency, ion channel gating and alcohol action.
- Author
-
Ren H, Salous AK, Paul JM, Lipsky RH, and Peoples RW
- Subjects
- Analysis of Variance, Cell Line, Dose-Response Relationship, Drug, Electrophysiology, Ethanol administration & dosage, Glutamic Acid administration & dosage, Glutamic Acid pharmacology, Humans, Inhibitory Concentration 50, Mutation, Ethanol pharmacology, Ion Channel Gating physiology, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
Background and Purpose: NMDA receptors are important molecular targets of ethanol action in the CNS. Previous studies have identified a site in membrane-associated domain 3 (M3) of the NR1 subunit and two sites in M4 of the NR2A subunit that influence alcohol action; the sites in NR2A M4 also regulate ion channel gating. The purpose of this study was to determine whether mutations at the site in the NR2A subunit corresponding to the NR1 M3 site influence alcohol action and ion channel gating., Experimental Approach: We investigated the effects of mutations at phenylalanine (F) 637 of the NR2A subunit using whole-cell and single-channel patch-clamp electrophysiological recording in transiently-transfected HEK 293 cells., Key Results: Mutations at F637 in the NR2A subunit altered peak and steady-state glutamate EC(50) values, maximal steady-state to peak current ratios (I(ss):I(p)), mean open time, and ethanol IC(50) values. Differences in glutamate potency among the mutants were not due to changes in desensitization. Ethanol IC(50) values were significantly correlated with glutamate EC(50) values, but not with maximal I(ss):I(p) or mean open time. Ethanol IC(50) values were linearly and inversely related to molecular volume of the substituent., Conclusions and Implications: These results demonstrate that NR2A(F637) influences NMDA receptor affinity, ion channel gating, and ethanol sensitivity. The changes in NMDA receptor affinity are likely to be the result of altered ion channel gating. In contrast to the cognate site in the NR1 subunit, the action of ethanol does not appear to involve occupation of a critical volume at NR2A(F637).
- Published
- 2007
- Full Text
- View/download PDF
32. Beta-N-methylamino-L-alanine enhances neurotoxicity through multiple mechanisms.
- Author
-
Lobner D, Piana PM, Salous AK, and Peoples RW
- Subjects
- Animals, Brain metabolism, Brain physiopathology, Cells, Cultured, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Cerebral Cortex pathology, Cyanobacteria Toxins, Hazardous Substances toxicity, Mice, Nerve Degeneration metabolism, Nerve Degeneration physiopathology, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases physiopathology, Neurons drug effects, Neurons metabolism, Neurons pathology, Neurotoxins toxicity, Oxidative Stress physiology, Patch-Clamp Techniques, Receptor, Metabotropic Glutamate 5, Receptors, Metabotropic Glutamate drug effects, Receptors, Metabotropic Glutamate metabolism, Receptors, N-Methyl-D-Aspartate drug effects, Receptors, N-Methyl-D-Aspartate metabolism, Amino Acids, Diamino toxicity, Brain drug effects, Excitatory Amino Acid Agonists toxicity, Nerve Degeneration chemically induced, Neurodegenerative Diseases chemically induced, Oxidative Stress drug effects
- Abstract
The idea that the environmental toxin beta-N-methylamino-l-alanine (BMAA) is involved in neurodegenerative diseases on Guam has risen and fallen over the years. The theory has gained greater interest with recent reports that BMAA is biomagnified, is widely distributed around the planet, and is present in the brains of Alzheimer's patients in Canada. We provide two important new findings. First, we show that BMAA at concentrations as low as 10 muM can potentiate neuronal injury induced by other insults. This is the first evidence that BMAA at concentrations below the mM range can enhance death of cortical neurons and illustrates potential synergistic effects of environmental toxins with underlying neurological conditions. Second, we show that the mechanism of BMAA toxicity is threefold: it is an agonist for NMDA and mGluR5 receptors, and induces oxidative stress. The results provide further support for the hypothesis that BMAA plays a role in neurodegenerative diseases.
- Published
- 2007
- Full Text
- View/download PDF
33. An interaction involving an arginine residue in the cytoplasmic domain of the 5-HT3A receptor contributes to receptor desensitization mechanism.
- Author
-
Hu XQ, Sun H, Peoples RW, Hong R, and Zhang L
- Subjects
- Amino Acids, Animals, Cytoplasm, Electrophysiology, Ion Channel Gating, Kinetics, Mice, Mutation, Protein Structure, Tertiary, Receptors, Serotonin, 5-HT3 genetics, Static Electricity, Xenopus laevis, Arginine, Receptors, Serotonin, 5-HT3 physiology
- Abstract
A large cytoplasmic domain accounts for approximately one-third of the entire protein of one superfamily of ligand-gated membrane ion channels, which includes nicotinic acetylcholine (nACh), gamma-aminobutyric acid type A (GABA(A)), serotonin type 3 (5-HT3), and glycine receptors. Desensitization is one functional feature shared by these receptors. Because most molecular studies of receptor desensitization have focused on the agonist binding and channel pore domains, relatively little is known about the role of the large cytoplasmic domain (LCD) in this process. To address this issue, we sequentially deleted segments of the LCD of the 5-HT3A receptor and examined the function of the mutant receptors. Deletion of a small segment that contains three amino acid residues (425-427) significantly slowed the desensitization kinetics of the 5-HT3A receptor. Both deletion and point mutation of arginine 427 altered desensitization kinetics in a manner similar to that of the (425-427) deletion without significantly changing the apparent agonist affinity. The extent of receptor desensitization was positively correlated with the polarity of the amino acid residue at 427: the desensitization accelerates with increasing polarity. Whereas the R427L mutation produced the slowest desensitization, it did not significantly alter single channel conductance of 5-HT3A receptor. Thus, the arginine 427 residue in the LCD contributes to 5-HT3A receptor desensitization, possibly through forming an electrostatic interaction with its neighboring residues. Because the polarity of the amino acid residue at 427 is highly conserved, such a desensitization mechanism may occur in other members of the Cys-loop family of ligand-gated ion channels.
- Published
- 2006
- Full Text
- View/download PDF
34. Role of extracellular histidines in agonist sensitivity of the rat P2X4 receptor.
- Author
-
Xiong K, Stewart RR, Hu XQ, Werby E, Peoples RW, Weight FF, and Li C
- Subjects
- Adenosine Triphosphate pharmacology, Animals, Cell Line, Female, Humans, In Vitro Techniques, Mutation, Oocytes physiology, Patch-Clamp Techniques, Rats, Receptors, Purinergic P2 genetics, Receptors, Purinergic P2X4, Xenopus laevis, Histidine physiology, Purinergic P2 Receptor Agonists
- Abstract
Relatively little information is available about the relationship between the molecular structure of each of the seven subtypes of P2X receptors and their function. Here, we investigated the possible function of three histidine residues in the extracellular loop of rat P2X(4) receptors. Mutation of histidine 241 to alanine (H241A) in the rat P2X(4) receptor decreased the EC(50) value of the ATP concentration-response curve from 8.4 to 0.7 microM. In contrast, the histidine mutation H140A or H286A slightly increased the EC(50) value. Maximal current responses were significantly larger in oocytes expressing rat H241A-mutated receptors compared to those expressing wildtype, H140A or H286A receptors. In addition, significantly less receptor protein was detected in H241A-expressing oocytes than in oocytes expressing wildtype, H140A or H286A receptors. Moreover, ATP-activated current in H241A-expressing cells activated faster than in wildtype receptor-expressing cells. The increased maximal current amplitude, the decrease in protein expression and the more rapid activation kinetics suggest that the H241A mutation facilitates opening of the receptor-channel (gating).
- Published
- 2004
- Full Text
- View/download PDF
35. Sites in the fourth membrane-associated domain regulate alcohol sensitivity of the NMDA receptor.
- Author
-
Honse Y, Ren H, Lipsky RH, and Peoples RW
- Subjects
- Amino Acid Sequence, Amino Acid Substitution genetics, Cell Line, Cell Membrane genetics, Dose-Response Relationship, Drug, Humans, Molecular Sequence Data, Protein Structure, Tertiary genetics, Protein Subunits genetics, Protein Subunits metabolism, Receptors, N-Methyl-D-Aspartate genetics, Cell Membrane physiology, Ethanol pharmacology, Receptors, N-Methyl-D-Aspartate chemistry, Receptors, N-Methyl-D-Aspartate physiology
- Abstract
N-methyl-D-aspartate (NMDA) receptors are important target sites of alcohol action in the central nervous system. Alcohol inhibits NMDA receptor current by an action on ion channel gating, apparently through a direct action on a region of the NMDA receptor accessible from the extracellular environment. Our previous studies have revealed an important role for a methionine residue (Met823) in membrane-associated domain 4 (M4) of the NR2A subunit in channel gating as well as alcohol sensitivity of the NMDA receptor. The role of sites in M4 of the NMDA receptor NR2A subunit adjacent to Met823 was investigated using tryptophan-scanning mutagenesis and electrophysiological recording. Receptors containing NR1 and NR2A(V820W) or NR2A(M817W) mutant subunits expressed in HEK 293 cells were not functional. The mutation Ala826Trp modified apparent desensitization, and the mutations Ala825Trp and Ala826Trp changed the mean open time of the channel as determined by fluctuation analysis. In addition, the mutations Tyr822Trp and Ala825Trp significantly altered the concentration-response curves for ethanol inhibition. The changes in mean open time did not appear to be able to account for the observed differences in ethanol sensitivity. These results indicate that this region in M4 of the NR2A subunit may be involved in the action of alcohol.
- Published
- 2004
- Full Text
- View/download PDF
36. A site of alcohol action in the fourth membrane-associated domain of the N-methyl-D-aspartate receptor.
- Author
-
Ren H, Honse Y, and Peoples RW
- Subjects
- Cell Line, Cell Membrane drug effects, Humans, Mutation, Receptors, N-Methyl-D-Aspartate genetics, Receptors, N-Methyl-D-Aspartate metabolism, Ethanol pharmacology, Receptors, N-Methyl-D-Aspartate drug effects
- Abstract
The N-methyl-d-aspartate (NMDA) subtype of ionotropic glutamate receptor is an important mediator of the behavioral effects of ethanol in the central nervous system. Although ethanol is known to inhibit NMDA receptors by influencing ion-channel gating, its molecular site of action and the mechanism underlying this effect have not been established. We have previously identified a conserved methionine residue in the fourth membrane-associated domain of the NMDA receptor NR2A subunit (Met823) that influences desensitization and gating of the ion channel. Here we report that this residue plays an important role in mediating the effect of ethanol on the NMDA receptor. Ethanol IC50 values among functional substitution mutants at this position varied over the range approximately 130-225 mm. There was a weak correlation between ethanol IC50 and mean open time of NR2A(Met823) mutants that was dependent on inclusion of the value for the tryptophan mutant. In the absence of this value, there was no trend toward a correlation among the remaining mutants. Desensitization appeared to influence the action of ethanol, because ethanol IC50 of the mutants was correlated with the steadystate to peak current ratio. With the exception of tryptophan, ethanol sensitivity was significantly related to the molecular volume and hydrophobicity of the substituent. The relation between ethanol sensitivity and the molecular volume and hydrophobicity at this position suggests that this residue interacts with or forms part of a site of ethanol action and that the presence of a tryptophan residue in this site disrupts its ability to interact with ethanol.
- Published
- 2003
- Full Text
- View/download PDF
37. Ethanol sensitivity of recombinant homomeric and heteromeric AMPA receptor subunits expressed in Xenopus oocytes.
- Author
-
Akinshola BE, Yasuda RP, Peoples RW, and Taylor RE
- Subjects
- Animals, Dose-Response Relationship, Drug, Female, Gene Expression Regulation physiology, Kainic Acid pharmacology, Oocytes, Protein Subunits agonists, Protein Subunits biosynthesis, Receptors, AMPA agonists, Receptors, AMPA biosynthesis, Recombinant Proteins agonists, Recombinant Proteins biosynthesis, Recombinant Proteins genetics, Xenopus laevis, Ethanol pharmacology, Gene Expression Regulation drug effects, Protein Subunits genetics, Protein Subunits metabolism, Receptors, AMPA genetics, Receptors, AMPA metabolism
- Abstract
Background: Ethanol is known to acutely inhibit AMPA receptor function, and sensitivity of AMPA receptors to ethanol is dependent on subunit composition in vivo and in vitro. A commonly used in vitro expression system for studying recombinant receptor subunits is the Xenopus laevis oocyte and two-electrode voltage-clamp electrophysiological recording. To date, ethanol sensitivity of injected receptor subunit complementary RNA (cRNA) has not been shown to be correlated with the actual expression of receptor subunits in oocytes. In this study, we compared ethanol sensitivity of homomeric and heteromeric AMPA receptor subunits microinjected into Xenopus oocytes and confirmed subunit expression in oocytes by immunoblot., Methods: cRNAs coding for the "flop" type AMPA GluR1 or GluR3 (homomeric), GluR2/GluR3 (heteromeric combination), and GluR1/2/3 (heteromeric combination) were microinjected in equimolar amounts of 16 to 20 ng into oocytes, which were studied for their sensitivity to ethanol. Oocytes injected with cRNA for homomeric or heteromeric subunit combinations were homogenized and the expressed subunits quantified with anti-GluR1, anti-GluR2, and anti-GluR2/3 antibodies., Results: Ethanol concentrations of 10 to 500 mM consistently inhibited currents activated in oocytes by 200 microM kainic acid. The expressed homomeric GluR1 receptor and heteromeric GluR1/2/3 receptor combination currents showed similar sensitivity to ethanol inhibition with half-maximal inhibition values of 170 +/- 12 mM and 176 +/- 8 mM, respectively. The expressed homomeric GluR3 receptor and heteromeric GluR2/3 receptor combination currents were differentially sensitive to ethanol inhibition with respective IC50 values of 238 +/- 9 mM and 338 +/- 16 mM., Conclusion: The expressed homomeric and heteromeric "flop" type AMPA receptors were differentially sensitive to ethanol, which may in part explain differential ethanol sensitivity in native neurons.
- Published
- 2003
- Full Text
- View/download PDF
38. Dynorphin A inhibits NMDA receptors through a pH-dependent mechanism.
- Author
-
Kanemitsu Y, Hosoi M, Zhu PJ, Weight FF, Peoples RW, McLaughlin JS, and Zhang L
- Subjects
- Allosteric Regulation drug effects, Allosteric Regulation physiology, Animals, Cell Line, Dynorphins pharmacology, Extracellular Fluid drug effects, Extracellular Fluid metabolism, Female, Hippocampus drug effects, Hippocampus metabolism, Humans, Hydrogen-Ion Concentration, Membrane Potentials drug effects, Membrane Potentials physiology, Neural Inhibition drug effects, Neurons drug effects, Oocytes, Organ Culture Techniques, Oxidation-Reduction drug effects, Patch-Clamp Techniques, Protein Structure, Tertiary physiology, Rats, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors, Synaptic Transmission drug effects, Xenopus laevis, Dynorphins metabolism, Neural Inhibition physiology, Neurons metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Synaptic Transmission physiology
- Abstract
Dynorphin A (DynA), an endogenous agonist of kappa-opioid receptors, has also been reported to directly interact with the NMDA receptor. DynA inhibition of NMDA receptor function has been suggested to be involved in its neuroprotective action during ischemic and acidic conditions. However, the effect of external pH on DynA inhibition of the NMDA receptor has not been reported. Here, we show that DynA inhibition of the NMDA receptor is dependent on extracellular pH over the range of pH 6.7-8.3, and the inhibition by 10 microM DynA increases at low pH by three- to four-fold in hippocampal neurons and in Xenopus oocytes expressing NR1-1a/2B subunits. Molecular studies showed that the interacting site for DynA on the NMDA receptor is distinct from that of proton or redox sites. Peptide mapping demonstrated important contributions of positively charged residues and specific structural organization of the peptide to the potency of DynA inhibition. Thus, DynA inhibits NMDA receptors through an allosteric mechanism, which is pH dependent and involves the specific structural features of the peptide.
- Published
- 2003
- Full Text
- View/download PDF
39. Modulation of glycine-activated ion channel function by G-protein betagamma subunits.
- Author
-
Yevenes GE, Peoples RW, Tapia JC, Parodi J, Soto X, Olate J, and Aguayo LG
- Subjects
- Animals, Cells, Cultured, Chloride Channels physiology, Electric Conductivity, Electrophysiology, GTP-Binding Proteins physiology, Humans, Mice, Mice, Inbred C57BL, Neurons metabolism, Peptides pharmacology, Receptors, Glycine drug effects, Receptors, Glycine metabolism, Receptors, Glycine physiology, Spinal Cord cytology, Spinal Cord metabolism, Glycine pharmacology, Heterotrimeric GTP-Binding Proteins pharmacology, Ion Channels drug effects, Ion Channels metabolism
- Abstract
Glycine receptors (GlyRs), together with GABA(A) and nicotinic acetylcholine (ACh) receptors, form part of the ligand-activated ion channel superfamily and regulate the excitability of the mammalian brain stem and spinal cord. Here we report that the ability of the neurotransmitter glycine to gate recombinant and native ionotropic GlyRs is modulated by the G protein betagamma dimer (Gbetagamma). We found that the amplitude of the glycine-activated Cl- current was enhanced after application of purified Gbetagamma or after activation of a G protein-coupled receptor. Overexpression of three distinct G protein alpha subunits (Galpha), as well as the Gbetagamma scavenger peptide ct-GRK2, significantly blunted the effect of G protein activation. Single-channel recordings from isolated membrane patches showed that Gbetagamma increased the GlyR open probability (nP(o)). Our results indicate that this interaction of Gbetagamma with GlyRs regulates both motor and sensory functions in the central nervous system.
- Published
- 2003
- Full Text
- View/download PDF
40. A site in the fourth membrane-associated domain of the N-methyl-D-aspartate receptor regulates desensitization and ion channel gating.
- Author
-
Ren H, Honse Y, Karp BJ, Lipsky RH, and Peoples RW
- Subjects
- Amino Acid Sequence, Cell Line, Excitatory Amino Acid Agonists metabolism, Glycine metabolism, Humans, Methionine metabolism, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, N-Methylaspartate metabolism, Patch-Clamp Techniques, Protein Structure, Secondary, Protein Structure, Tertiary, Receptors, N-Methyl-D-Aspartate genetics, Regression Analysis, Sequence Alignment, Ion Channel Gating physiology, Receptors, N-Methyl-D-Aspartate chemistry, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
The N-methyl-d-aspartate (NMDA) receptor has four membrane-associated domains, three of which are membrane-spanning (M1, M3, and M4) and one of which is a re-entrant pore loop (M2). The M1-M3 domains have been demonstrated to influence the function of the ion channel, but a similar role for the M4 domain has not been reported. We have identified a methionine residue (Met(823)) in the M4 domain of the NR2A subunit that regulates desensitization and ion channel gating. A tryptophan substitution at this site did not alter the EC(50) for glycine or the peak NMDA EC(50) but decreased the steady-state NMDA EC(50) and markedly increased apparent desensitization, mean open time, and peak current density. Results of rapid solution exchange experiments revealed that changes in microscopic desensitization rates and closing rates could account for the changes in macroscopic desensitization, steady-state NMDA EC(50), and current density. Other amino acid substitutions at this site could increase or decrease the rate of desensitization and mean open time of the ion channel. Both mean open time and desensitization were dependent primarily upon the hydrophobic character of the amino acid at the position. These results demonstrate an important role for hydrophobic interactions at Met(823) in regulation of NMDA receptor function.
- Published
- 2003
- Full Text
- View/download PDF
41. GABA(A) receptors as molecular sites of ethanol action. Direct or indirect actions?
- Author
-
Aguayo LG, Peoples RW, Yeh HH, and Yevenes GE
- Subjects
- Animals, Binding Sites, Brain cytology, Brain metabolism, Calcium physiology, Central Nervous System Depressants metabolism, Electrophysiology, Ethanol metabolism, Hippocampus cytology, Hippocampus drug effects, Hippocampus metabolism, Humans, Neurons metabolism, Protein Subunits, Receptors, GABA-A metabolism, Signal Transduction physiology, Brain drug effects, Central Nervous System Depressants pharmacology, Ethanol pharmacology, Receptors, GABA-A drug effects
- Abstract
Despite the fact that ethanol is one of the most widely used psychoactive agents, the mechanisms and sites of action by which it modifies brain functions are only now being elucidated. Studies over the last decade have shown that ethanol can specifically alter the function of several ligand-activated ion channels including N-methyl-D-aspartate (NMDA), serotonin (5-HT(3)), glycine and GABA(A) receptors. After several years of extensive research in this field, the resolution of what, where and how ethanol modifies GABA(A) receptors continues to be controversial. For example, after demonstrating that ethanol was able to alter Cl(-) flux in synaptoneurosomes and cultured neurons, several electrophysiological studies were unable to show enhancement of the GABA(A) receptor current in single neurons. The lack of positive results with low ethanol concentrations was interpreted as being due to receptor heterogeneity and differences in intracellular modulation by protein kinases and calcium. The existence of high receptor heterogeneity with respect to ethanol sensitivity has been supported by studies done in a variety of cell types which showed that ethanol potentiated some, but not other neurons. Adding to this complexity, it was shown that while some hippocampal GABA(A) receptors can be affected by ethanol concentrations between 1 and 100 mM, others are only sensitive to concentrations above 200 mM. The curve of the relationship between low ethanol concentrations and current enhancement suggests a high degree of complexity in the molecular interaction because of its steepness and "inverted" U shape. Similarly, the effects of ethanol on GABA(A) receptors seems much more complex than those of benzodiazepines, barbiturates and neurosteroids. The major problem encountered in advancing understanding of the mechanism of ethanol action in native neuronal receptors has been the large variability detected in ethanol sensitivity. For example, several studies have shown that only some groups of neurons are sensitive to pharmacologically relevant concentrations of ethanol (1-100 mM). This receptor sensitivity variability has not been resolved using recombinant expression systems. For example, studies performed in recombinant receptors, although important for elucidating molecular requirements, have shown that they are less sensitive to ethanol suggesting that neuronal substrates are important for ethanol actions. In this review, we discuss the possibility that ethanol's action on the GABA(A) receptor may not be due solely to a direct interaction with the receptor protein, but that its effects could also be modulated by intracellular regulation, and that this latter effect is the more physiologically relevant one. Data in cortical and hippocampal neurons suggest that ethanol action on the receptor is labile, and that it also depends on repetitive stimulation and neuron integrity. In addition, the action of ethanol can be modified by activation of protein kinases and neuronal development. Finally, we discuss that the best approach for studying the interaction between the receptor and ethanol is through the combined use of recombinant receptors and overexpression in neurons.
- Published
- 2002
- Full Text
- View/download PDF
42. Inhibition of N-methyl-D-aspartate receptors by straight-chain diols: implications for the mechanism of the alcohol cutoff effect.
- Author
-
Peoples RW and Ren H
- Subjects
- Animals, CHO Cells, Cells, Cultured, Cricetinae, Electrophysiology, Hexanes, Humans, Receptors, N-Methyl-D-Aspartate genetics, Receptors, N-Methyl-D-Aspartate metabolism, Receptors, N-Methyl-D-Aspartate physiology, Transfection, Glycols pharmacology, Hexanols pharmacology, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors
- Abstract
n-Alkanol inhibition of N-methyl-D-aspartate (NMDA) receptors exhibits a "cutoff" effect: alcohols with up to eight to nine carbon atoms inhibit the receptor, whereas larger alcohols do not. This phenomenon was originally proposed to result from size exclusion; i.e., alcohols above the cutoff are too large to bind to an amphiphilic site on the receptor. In the present study, 1,Omega-diols with 3 to 14 carbon atoms inhibited NMDA-activated current in Chinese hamster ovary and human embryonic kidney 293 cells transiently expressing NR1 and NR2B NMDA receptor subunits. Results of fluctuation analysis experiments were consistent with a similar mechanism of inhibition of NMDA-activated current by alcohols and diols. The average change in apparent energy of binding of the diols caused by addition of a methylene group was 2.1 kJ/mol, which is consistent with an important role of hydrophobic interactions. Because 1,Omega-diols with 9 to 14 carbons inhibited NMDA-activated current, despite having molecular volumes exceeding that at the cutoff point for 1-alkanols, a size exclusion mechanism seems inadequate to explain the cutoff effect. A disparity in hydrophobicity values at the cutoff for alcohols and diols, however, revealed that hydrophobicity could also not entirely explain the cutoff phenomenon. From these results, it seems that the cutoff effect on NMDA receptors results primarily from the inability of long-chain alcohols to achieve adequate concentrations at their site of action due to low aqueous solubility, although other factors may also contribute to the effect.
- Published
- 2002
- Full Text
- View/download PDF
43. Alcohols inhibit N-methyl-D-aspartate receptors via a site exposed to the extracellular environment.
- Author
-
Peoples RW and Stewart RR
- Subjects
- Animals, Binding Sites, CHO Cells, Cricetinae, Dose-Response Relationship, Drug, Electrophysiology, Extracellular Space, Glycine pharmacology, Green Fluorescent Proteins, Luminescent Proteins genetics, Luminescent Proteins metabolism, Membrane Potentials drug effects, Mutation, N-Methylaspartate pharmacology, Pentanols metabolism, Pentanols pharmacology, Receptors, N-Methyl-D-Aspartate genetics, Receptors, N-Methyl-D-Aspartate physiology, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Transfection, Ethanol pharmacology, Receptors, N-Methyl-D-Aspartate antagonists & inhibitors
- Abstract
N-Methyl-D-aspartate (NMDA) receptors are important CNS target sites of alcohols, but the site and mechanism of action of alcohols on NMDA receptors remains unclear. In CHO-K1 cells transfected with NR1/NR2B NMDA receptor subunits, ethanol inhibited NMDA-activated current with an IC(50) of 138 mM. Truncation of the intracellular C-terminal domain of the NR1 subunit (NR1T) did not alter ethanol sensitivity when combined with the NR2B subunit, but a similar truncation of the NR2B subunit (NR2BT) slightly enhanced ethanol sensitivity of receptors formed from coexpression with either NR1 or NR1T subunits. 1-Pentanol applied externally inhibited NMDA receptors with an IC(50) of 9.9 mM, but intracellular application of 1-pentanol (25 mM) did not alter NMDA receptor inhibition by externally applied ethanol or 1-pentanol. In addition, the amplitude of NMDA-activated current did not decrease during the time required for 1-pentanol (25 mM) to diffuse throughout the cytoplasm. Ethanol did not inhibit NMDA receptors when bath-applied in cell-attached patches or when applied to the cytoplasmic face of inside-out membrane patches. These results appear to be best explained by an action of alcohols on the NMDA receptor-channel protein, at a site located in a domain exposed to, or only accessible from, the extracellular environment.
- Published
- 2000
- Full Text
- View/download PDF
44. Alcohol action on membrane ion channels gated by extracellular ATP (P2X receptors).
- Author
-
Weight FF, Li C, and Peoples RW
- Subjects
- Animals, Cell Membrane drug effects, Receptors, Purinergic P2 physiology, Adenosine Triphosphate physiology, Ethanol pharmacology, Ion Channel Gating drug effects, Ion Channels drug effects, Receptors, Purinergic P2 drug effects
- Abstract
Extracellular adenosine 5'-triphosphate (ATP) has been reported to produce excitatory actions in the nervous system, such as excitatory postsynaptic potentials or currents in both central and peripheral neurons, via activation of a class of ATP-gated membrane ion channels designated P2X receptors. This article reviews studies of alcohol effects on these receptor-channels. Ethanol has been found to inhibit ATP-gated ion channel function by shifting the agonist concentration-response curve to the right in a parallel manner, increasing the EC50 without affecting Emax of this curve. To distinguish whether this inhibition involves competitive antagonism of agonist action or a decrease in the affinity of the agonist binding site, the kinetics of activation and deactivation of agonist-activated current were studied. Ethanol was found to decrease the time-constant of deactivation of ATP-gated ion channels without affecting the time-constant of activation, indicating that ethanol inhibits the function of these receptors by an allosteric decrease in the affinity of the agonist binding site. The inhibition of ATP-gated ion channel function by a number of alcohols was found to exhibit a distinct cutoff effect that appeared to be related to the molecular volume of the alcohols. For alcohols with a molecular volume of < or = 42.2 ml/mol, potency for inhibiting ATP-activated current was correlated with lipid solubility (order of potency: 1-propanol = trifluoroethanol > monochloroethanol > ethanol > methanol). However, despite increased lipid solubility, alcohols with a molecular volume of > or = 46.1 ml/mol (1-butanol, 1-pentanol, trichloroethanol, and dichloroethanol) were without effect on the ATP-activated current. This cutoff effect has been interpreted as evidence that alcohols inhibit the function of ATP-gated ion channels by interacting with a hydrophobic pocket of circumscribed dimensions on the receptor protein. To evaluate the localization of this presumed alcohol binding site, the effect of the intracellular application of ethanol was studied on the inhibition of ATP-activated current by extracellularly applied ethanol. The intracellular application of 100 mM ethanol did not affect the inhibition of current by 100 mM extracellular ethanol, suggesting that the alcohol inhibition of ATP-gated ion channel function involves the extracellular domain of the receptor. Finally, recent studies suggest that the alcohol sensitivity of ATP-gated channels may be regulated by physiological mechanisms.
- Published
- 1999
- Full Text
- View/download PDF
45. Differential modulation by copper and zinc of P2X2 and P2X4 receptor function.
- Author
-
Xiong K, Peoples RW, Montgomery JP, Chiang Y, Stewart RR, Weight FF, and Li C
- Subjects
- Adenosine Triphosphate pharmacology, Animals, Dose-Response Relationship, Drug, Electric Conductivity, Female, Oocytes, Osmolar Concentration, Patch-Clamp Techniques, Receptors, Purinergic P2 physiology, Receptors, Purinergic P2X2, Receptors, Purinergic P2X4, Xenopus laevis, Copper pharmacology, Receptors, Purinergic P2 drug effects, Zinc pharmacology
- Abstract
Differential Modulation by Copper and Zinc of P2X2 and P2X4 Receptor Function. The modulation by Cu2+ and Zn2+ of P2X2 and P2X4 receptors expressed in Xenopus oocytes was studied with the two-electrode, voltage-clamp technique. In oocytes expressing P2X2 receptors, both Cu2+ and Zn2+, in the concentration range 1-130 microM, reversibly potentiated current activated by submaximal concentrations of ATP. The Cu2+ and Zn2+ concentrations that produced 50% of maximal potentiation (EC50) of current activated by 50 microM ATP were 16.3 +/- 0.9 (SE) microM and 19.6 +/- 1.5 microM, respectively. Cu2+ and Zn2+ potentiation of ATP-activated current was independent of membrane potential between -80 and +20 mV and did not involve a shift in the reversal potential of the current. Like Zn2+, Cu2+ increased the apparent affinity of the receptor for ATP, as evidenced by a parallel shift of the ATP concentration-response curve to the left. However, Cu2+ did not enhance ATP-activated current in the presence of a maximally effective concentration of Zn2+, suggesting a common site or mechanism of action of Cu2+ and Zn2+ on P2X2 receptors. For the P2X4 receptor, Zn2+, from 0.5 to 20 microM enhanced current activated by 5 microM ATP with an EC50 value of 2.4 +/- 0.2 microM. Zn2+ shifted the ATP concentration-response curve to the left in a parallel manner, and potentiation by Zn2+ was voltage independent. By contrast, Cu2+ in a similar concentration range did not affect ATP-activated current in oocytes expressing P2X4 receptors, and Cu2+ did not alter the potentiation of ATP-activated current produced by Zn2+. The results suggest that Cu2+ and Zn2+ differentially modulate the function of P2X2 and P2X4 receptors, perhaps because of differences in a shared site of action on both subunits or the absence of a site for Cu2+ action on the P2X4 receptor.
- Published
- 1999
- Full Text
- View/download PDF
46. Distinct ATP-activated currents in different types of neurons dissociated from rat dorsal root ganglion.
- Author
-
Li C, Peoples RW, Lanthorn TH, Li ZW, and Weight FF
- Subjects
- Adenosine Triphosphate analogs & derivatives, Adenosine Triphosphate physiology, Animals, Capsaicin pharmacology, Cell Size, Ganglia, Spinal cytology, In Vitro Techniques, Male, Membrane Potentials drug effects, Membrane Potentials physiology, Nerve Fibers ultrastructure, Neurons classification, Neurons cytology, Patch-Clamp Techniques, Rats, Rats, Sprague-Dawley, Thionucleotides pharmacology, Adenosine Triphosphate pharmacology, Ganglia, Spinal physiology, Neurons physiology
- Abstract
Rat dorsal root ganglion neurons can be classified into at least three distinct groups based on cell size, afferent fiber diameter, electrophysiological properties, sensitivity to vanilloid agonists such as capsaicin, and function. In the present study, ATP-activated current in these neurons was characterized using whole-cell patch-clamp recording. Small diameter (<30 microm) cells had high capsaicin sensitivity, high affinity for ATP, and rapidly desensitizing ATP-activated current. Medium diameter (30-50 microm) cells had no capsaicin sensitivity, lower affinity for ATP and slowly desensitizing ATP-activated current. Large diameter (>50 microm) cells were insensitive to both capsaicin and ATP. These findings suggest that distinct types of ATP receptor-ion channels are expressed in different types of dorsal root ganglion neurons, and may contribute to the functional differences among these types of neurons.
- Published
- 1999
- Full Text
- View/download PDF
47. Differential alcohol modulation of GABA(A) and NMDA receptors.
- Author
-
Peoples RW and Weight FF
- Subjects
- Animals, Hippocampus cytology, Hippocampus metabolism, Linear Models, Mice, Neurons metabolism, Patch-Clamp Techniques, Rats, Solubility, Water chemistry, Ethanol pharmacology, Hippocampus drug effects, Neurons drug effects, Receptors, GABA-A drug effects, Receptors, N-Methyl-D-Aspartate drug effects
- Abstract
NMDA and GABA(A) receptors are believed to be important CNS targets of alcohol action. In mouse hippocampal neurons, n-alcohols from ethanol to dodecanol enhanced GABA-activated ion current, whereas higher alcohols had no effect. Alcohols below pentanol affected NMDA receptors more potently than GABA(A) receptors. Increasing alcohol carbon chain length produced a greater average change in apparent binding energy and potency for modulation of GABA(A) than of NMDA receptor-channels, with the result that alcohols above pentanol affected GABA(A) receptors more potently than NMDA receptors. The anesthetic potency of n-alcohols in rats more closely reflected NMDA receptor modulatory potency for lower alcohols and GABA(A) receptor modulatory potency for higher alcohols. The results suggest that there may be fundamental differences in the sites through which alcohols affect NMDA and GABA(A) receptor function.
- Published
- 1999
- Full Text
- View/download PDF
48. NMDA receptor pharmacology and analysis of patch-clamp recordings.
- Author
-
Wright JM and Peoples RW
- Subjects
- Animals, Patch-Clamp Techniques, Receptors, N-Methyl-D-Aspartate physiology
- Published
- 1999
- Full Text
- View/download PDF
49. Inhibition of excitatory amino acid-activated currents by trichloroethanol and trifluoroethanol in mouse hippocampal neurones.
- Author
-
Peoples RW and Weight FF
- Subjects
- Animals, Cells, Cultured, Ethylene Chlorohydrin pharmacology, Hippocampus cytology, Hippocampus physiology, Mice, Neurons drug effects, Neurons physiology, Ethylene Chlorohydrin analogs & derivatives, Hippocampus drug effects, N-Methylaspartate pharmacology, Trifluoroethanol pharmacology
- Abstract
1. The effects of the active metabolite of chloral derivative sedative-hypnotic agents, 2,2,2-trichloroethanol (trichloroethanol), and its analog 2,2,2-trifluoroethanol (trifluoroethanol), were studied on ion current activated by the excitatory amino acids N-methyl-D-aspartate (NMDA) and kainate in mouse hippocampal neurones in culture using whole-cell patch-clamp recording. 2. Both trichloroethanol and trifluoroethanol inhibited excitatory amino acid-activated currents in a concentration-dependent manner. Trichloroethanol inhibited NMDA- and kainate-activated currents with IC50 values of 6.4 and 12 mM, respectively, while trifluoroethanol inhibited NMDA- and kainate-activated currents with IC50 values of 28 and 35 mM, respectively. 3. Both trichloroethanol and trifluoroethanol appeared to be able to inhibit excitatory amino acid-activated currents by 100 per cent. 4.Concentration-response analysis of NMDA- and kainate-activated current revealed that trichloroethanol decreased the maximal response to both agonists without significantly affecting their EC50 values. 5. Both trichloroethanol and trifluoroethanol inhibited excitatory amino acid-activated currents more potently than did ethanol. The inhibitory potency of trichloroethanol and trifluoroethanol appears to be associated with their increased hydrophobicity. 6. The observation that trichloroethanol inhibits excitatory amino acid-activated currents at anaesthetic concentrations suggests that inhibition of excitatory amino acid receptors may contribute to the CNS depressant effects of chloral derivative sedative-hypnotic agents.
- Published
- 1998
- Full Text
- View/download PDF
50. Inhibition of NMDA-gated ion channels by the P2 purinoceptor antagonists suramin and reactive blue 2 in mouse hippocampal neurones.
- Author
-
Peoples RW and Li C
- Subjects
- Animals, Cells, Cultured, Glycine pharmacology, Hippocampus embryology, Hippocampus metabolism, Mice, Neurons, Afferent drug effects, Neurons, Afferent metabolism, Patch-Clamp Techniques, Pyridoxal Phosphate analogs & derivatives, Pyridoxal Phosphate pharmacology, Excitatory Amino Acid Agonists pharmacology, Hippocampus drug effects, Ion Channels antagonists & inhibitors, N-Methylaspartate pharmacology, Protein Synthesis Inhibitors pharmacology, Purinergic P2 Receptor Antagonists, Suramin pharmacology, Triazines pharmacology
- Abstract
1. The action of suramin and reactive blue 2 on N-methyl-D-aspartate (NMDA)-activated ion current was studied in mouse hippocampal neurones in culture by use of whole-cell patch-clamp recording. 2. Suramin and reactive blue 2 inhibited steady-state current activated by 25 microM NMDA with IC50 values of 68 and 11 microM, respectively. 3. Reactive blue 2 produced a gradual decline of NMDA-activated current to a steady-state, but this slow onset was not an indication of use-dependence, as it could be eliminated by exposure to reactive blue 2 before NMDA application. In addition, NMDA-activated current recovered completely from inhibition by reactive blue 2 in the absence of agonist. 4. The slow onset of inhibition by reactive blue 2 was not apparently due to an action at an intracellular site, as inclusion of 250 microM reactive blue 2 in the recording pipette did not alter inhibition by 25 microM reactive blue 2 applied externally. 5. Reactive blue 2 and suramin inhibited NMDA-gated channels in a voltage-independent manner. 6. Reactive blue 2, 25 microM, decreased the maximal response to NMDA from 1441 to 598 pA without changing its EC50. In contrast, 75 microM suramin increased the EC50 for NMDA from 13 to 35 microM, and decreased the maximal response to NMDA from 1822 to 1498 pA. Schild analysis of suramin inhibition of NMDA-activated current yielded a nonlinear plot. 7. Both agents decreased the maximal response to glycine without altering its EC50. 8. Suramin and reactive blue 2 appear to inhibit NMDA receptor-channels in a manner that is noncompetitive with respect to both NMDA and glycine. However, inhibition by suramin differed from that by reactive blue 2, in that suramin significantly increased the EC50 of NMDA.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.