1
|
Khalid A, Farhat N. Structural Insight on Interaction of NMDA receptor with fentanyl, ketamine and Isoflurane: A Computational Study to Unravel Mode of Binding. Cell Biochem Biophys 2024:10.1007/s12013-024-01499-z. [PMID: 39231847 DOI: 10.1007/s12013-024-01499-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 09/06/2024]
Abstract
NMDA receptors are considered targets for many anesthetics if they are modulated by the drugs at clinically relevant concentrations. Volatile anesthetics like isoflurane and ketamine interact with NMDA receptors, inhibiting channel activation and thus blocking NMDA neurotransmission at clinically relevant concentrations. The mode of binding of commonly used drugs like ketamine, isoflurane, and fentanyl is poorly understood. We used molecular docking, molecular dynamics simulations, and DFT calculation of these drugs against the NMDA receptor. Using well-defined computational methods, we identified that these drugs have high docking scores and significant interaction with receptors. These drugs bind to the substrate-binding pocket and form a remarkable number of interactions. We have found that these interactions are stable and have low HOMO-LUMO energy gaps. This study provides enough evidences of strong and stable interaction between drugs and NMDA receptor.
Collapse
Affiliation(s)
- Atif Khalid
- Department of Anaesthesiology, Veeranga AvantiBai Lodhi Autonomous State Medical College, Etah, India
| | - Nabeela Farhat
- School of Energy Science and Engineering, IIT Guwahati, Guwahati, India.
| |
Collapse
|
2
|
Song XJ, Hu JJ. Neurobiological basis of emergence from anesthesia. Trends Neurosci 2024; 47:355-366. [PMID: 38490858 DOI: 10.1016/j.tins.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/25/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024]
Abstract
The suppression of consciousness by anesthetics and the emergence of the brain from anesthesia are complex and elusive processes. Anesthetics may exert their inhibitory effects by binding to specific protein targets or through membrane-mediated targets, disrupting neural activity and the integrity and function of neural circuits responsible for signal transmission and conscious perception/subjective experience. Emergence from anesthesia was generally thought to depend on the elimination of the anesthetic from the body. Recently, studies have suggested that emergence from anesthesia is a dynamic and active process that can be partially controlled and is independent of the specific molecular targets of anesthetics. This article summarizes the fundamentals of anesthetics' actions in the brain and the mechanisms of emergence from anesthesia that have been recently revealed in animal studies.
Collapse
Affiliation(s)
- Xue-Jun Song
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, China.
| | - Jiang-Jian Hu
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, China
| |
Collapse
|
3
|
McGuigan S, Marie DJ, O'Bryan LJ, Flores FJ, Evered L, Silbert B, Scott DA. The cellular mechanisms associated with the anesthetic and neuroprotective properties of xenon: a systematic review of the preclinical literature. Front Neurosci 2023; 17:1225191. [PMID: 37521706 PMCID: PMC10380949 DOI: 10.3389/fnins.2023.1225191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Xenon exhibits significant neuroprotection against a wide range of neurological insults in animal models. However, clinical evidence that xenon improves outcomes in human studies of neurological injury remains elusive. Previous reviews of xenon's method of action have not been performed in a systematic manner. The aim of this review is to provide a comprehensive summary of the evidence underlying the cellular interactions responsible for two phenomena associated with xenon administration: anesthesia and neuroprotection. Methods A systematic review of the preclinical literature was carried out according to the PRISMA guidelines and a review protocol was registered with PROSPERO. The review included both in vitro models of the central nervous system and mammalian in vivo studies. The search was performed on 27th May 2022 in the following databases: Ovid Medline, Ovid Embase, Ovid Emcare, APA PsycInfo, and Web of Science. A risk of bias assessment was performed utilizing the Office of Health Assessment and Translation tool. Given the heterogeneity of the outcome data, a narrative synthesis was performed. Results The review identified 69 articles describing 638 individual experiments in which a hypothesis was tested regarding the interaction of xenon with cellular targets including: membrane bound proteins, intracellular signaling cascades and transcription factors. Xenon has both common and subtype specific interactions with ionotropic glutamate receptors. Xenon also influences the release of inhibitory neurotransmitters and influences multiple other ligand gated and non-ligand gated membrane bound proteins. The review identified several intracellular signaling pathways and gene transcription factors that are influenced by xenon administration and might contribute to anesthesia and neuroprotection. Discussion The nature of xenon NMDA receptor antagonism, and its range of additional cellular targets, distinguishes it from other NMDA antagonists such as ketamine and nitrous oxide. This is reflected in the distinct behavioral and electrophysiological characteristics of xenon. Xenon influences multiple overlapping cellular processes, both at the cell membrane and within the cell, that promote cell survival. It is hoped that identification of the underlying cellular targets of xenon might aid the development of potential therapeutics for neurological injury and improve the clinical utilization of xenon. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier: 336871.
Collapse
Affiliation(s)
- Steven McGuigan
- Department of Anesthesia and Acute Pain Medicine, St. Vincent's Hospital, Melbourne, VIC, Australia
- Department of Critical Care, University of Melbourne, Melbourne, VIC, Australia
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Boston, MA, United States
| | - Daniel J. Marie
- Department of Anesthesia and Acute Pain Medicine, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Liam J. O'Bryan
- Department of Anesthesia and Acute Pain Medicine, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Francisco J. Flores
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Boston, MA, United States
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lisbeth Evered
- Department of Anesthesia and Acute Pain Medicine, St. Vincent's Hospital, Melbourne, VIC, Australia
- Department of Critical Care, University of Melbourne, Melbourne, VIC, Australia
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
| | - Brendan Silbert
- Department of Anesthesia and Acute Pain Medicine, St. Vincent's Hospital, Melbourne, VIC, Australia
- Department of Critical Care, University of Melbourne, Melbourne, VIC, Australia
| | - David A. Scott
- Department of Anesthesia and Acute Pain Medicine, St. Vincent's Hospital, Melbourne, VIC, Australia
- Department of Critical Care, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
4
|
Kotani N, Jang IS, Nakamura M, Nonaka K, Nagami H, Akaike N. Depression of Synaptic N-methyl-D-Aspartate Responses by Xenon and Nitrous Oxide. J Pharmacol Exp Ther 2023; 384:187-196. [PMID: 36272733 DOI: 10.1124/jpet.122.001346] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/12/2022] [Accepted: 10/14/2022] [Indexed: 12/13/2022] Open
Abstract
In "synapse bouton preparation" of rat hippocampal CA3 neurons, we examined how Xe and N2O modulate N-methyl-D-aspartate (NMDA) receptor-mediated spontaneous and evoked excitatory post-synaptic currents (sEPSCNMDA and eEPSCNMDA). This preparation is a mechanically isolated single neuron attached with nerve endings (boutons) preserving normal physiologic function and promoting the exact evaluation of sEPSCNMDA and eEPSCNMDA responses without influence of extrasynaptic, glial, and other neuronal tonic currents. These sEPSCs and eEPSCs are elicited by spontaneous glutamate release from many homologous glutamatergic boutons and by focal paired-pulse electric stimulation of a single bouton, respectively. The s/eEPSCAMPA/KA and s/eEPSCNMDA were isolated pharmacologically by their specific antagonists. Thus, independent contributions of pre- and postsynaptic responses could also be quantified. All kinetic properties of s/eEPSCAMPA/KA and s/eEPSCNMDA were detected clearly. The s/eEPSCNMDA showed smaller amplitude and slower rise and 1/e decay time constant (τ Decay) than s/eEPSCAMPA/KA Xe (70%) and N2O (70%) significantly decreased the frequency and amplitude without altering the τ Decay of sEPSCNMDA They also decreased the amplitude but increased the Rf and PPR without altering the τ Decay of the eEPSCNMDA These data show clearly that "synapse bouton preparation" can be an accurate model for evaluating s/eEPSCNMDA Such inhibitory effects of gas anesthetics are primarily due to presynaptic mechanisms. Present results may explain partially the powerful analgesic effects of Xe and N2O. SIGNIFICANCE STATEMENT: We could record pharmacologically isolated NMDA receptor-mediated spontaneous and (action potential-evoked) excitatory postsynaptic currents (sEPSCNMDA and eEPSCNMDA) and clearly detect all kinetic parameters of sEPSCNMDA and eEPSCNMDA at synaptic levels by using "synapse bouton preparation" of rat hippocampal CA3 neurons. We found that Xe and N2O clearly suppressed both sEPSCNMDA and eEPSCNMDA. Different from previous studies, present results suggest that Xe and N2O predominantly inhibit the NMDA responses by presynaptic mechanisms.
Collapse
Affiliation(s)
- Naoki Kotani
- Kitamoto Hospital, Saitama, Japan (N.K., N.A.); Kyungpook National University, Daegu, Republic of Korea (I.S.J., M.N.); Kumamoto Health Science University, Kumamoto, Japan (K.N.), and Kumamoto Kinoh Hospital, Kumamoto, Japan (H.N., N.A.)
| | - Il-Sung Jang
- Kitamoto Hospital, Saitama, Japan (N.K., N.A.); Kyungpook National University, Daegu, Republic of Korea (I.S.J., M.N.); Kumamoto Health Science University, Kumamoto, Japan (K.N.), and Kumamoto Kinoh Hospital, Kumamoto, Japan (H.N., N.A.)
| | - Michiko Nakamura
- Kitamoto Hospital, Saitama, Japan (N.K., N.A.); Kyungpook National University, Daegu, Republic of Korea (I.S.J., M.N.); Kumamoto Health Science University, Kumamoto, Japan (K.N.), and Kumamoto Kinoh Hospital, Kumamoto, Japan (H.N., N.A.)
| | - Kiku Nonaka
- Kitamoto Hospital, Saitama, Japan (N.K., N.A.); Kyungpook National University, Daegu, Republic of Korea (I.S.J., M.N.); Kumamoto Health Science University, Kumamoto, Japan (K.N.), and Kumamoto Kinoh Hospital, Kumamoto, Japan (H.N., N.A.)
| | - Hideaki Nagami
- Kitamoto Hospital, Saitama, Japan (N.K., N.A.); Kyungpook National University, Daegu, Republic of Korea (I.S.J., M.N.); Kumamoto Health Science University, Kumamoto, Japan (K.N.), and Kumamoto Kinoh Hospital, Kumamoto, Japan (H.N., N.A.)
| | - Norio Akaike
- Kitamoto Hospital, Saitama, Japan (N.K., N.A.); Kyungpook National University, Daegu, Republic of Korea (I.S.J., M.N.); Kumamoto Health Science University, Kumamoto, Japan (K.N.), and Kumamoto Kinoh Hospital, Kumamoto, Japan (H.N., N.A.)
| |
Collapse
|
5
|
Zhang M, Chen Y, Liu J, Yang Y, Wang R, Zhang D, Zhu T. Development of NMDA receptors contributes to the enhancement of electroencephalogram oscillations under volatile anesthetics in rats. Front Neural Circuits 2022; 16:1065374. [PMID: 36589861 PMCID: PMC9797678 DOI: 10.3389/fncir.2022.1065374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background Volatile anesthetics including sevoflurane and isoflurane enhance oscillations of cortical electroencephalogram (EEG), partly by their modulations on glutamate-mediated excitatory synaptic transmission. Expression of NMDA receptors is increased during neonatal development. However, how the development of NMDA receptors influences EEG under volatile anesthesia remains unclear. Methods Expressions of NMDA receptor subtypes (NR1, NR2A, and NR2B) during neonatal development were measured by Western blotting. MAC (minimal alveolar concentration) of isoflurane and sevoflurane that inducing loss of righting reflex (LORR) and no response to tail-clamp (immobility) were measured to verify the effect of NR1 expression on anesthetic potency during neonatal development. Cortical electroencephalogram recording was used to examine the influence of NR1 expression on the power density of EEG. Results The expressions of GluNR1, GluNR2A and GluNR2B receptors were gradually increased during neonatal development in cortex, hippocampus and thalamus of rats. Knockdown of NR1 enhanced the sedative potency of volatile anesthetics but not on immobility potency in postnatal day 14 (P14)-P17 rats. For cortical EEG, along with the increased concentration of volatile anesthetics, cortical slow-delta oscillations of P5 rats were inhibited, theta and alpha oscillations were not changed significantly; while these oscillations were enhanced until high anesthetic concentrations in P21 rats. Knockdown of NR1 in forebrain suppressed the enhancement of cortical EEG oscillations in P21 rats. Conclusion The development of NMDA receptors may contribute to the enhancement of cortical EEG oscillations under volatile anesthetics.
Collapse
Affiliation(s)
- Mingyue Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yali Chen
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yaoxin Yang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Rurong Wang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,*Correspondence: Donghang Zhang,
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Tao Zhu,
| |
Collapse
|
6
|
Laha K, Zhu M, Gemperline E, Rau V, Li L, Fanselow MS, Lennertz R, Pearce RA. CPP impairs contextual learning at concentrations below those that block pyramidal neuron NMDARs and LTP in the CA1 region of the hippocampus. Neuropharmacology 2022; 202:108846. [PMID: 34687710 PMCID: PMC8627488 DOI: 10.1016/j.neuropharm.2021.108846] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/29/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Drugs that block N-methyl-d-aspartate receptors (NMDARs) suppress hippocampus-dependent memory formation; they also block long-term potentiation (LTP), a cellular model of learning and memory. However, the fractional block that is required to achieve these effects is unknown. Here, we measured the dose-dependent suppression of contextual memory in vivo by systemic administration of the competitive antagonist (R,S)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP); in parallel, we measured the concentration-dependent block by CPP of NMDAR-mediated synapses and LTP of excitatory synapses in hippocampal brain slices in vitro. We found that the dose of CPP that suppresses contextual memory in vivo (EC50 = 2.3 mg/kg) corresponds to a free concentration of 53 nM. Surprisingly, applying this concentration of CPP to hippocampal brain slices had no effect on the NMDAR component of evoked field excitatory postsynaptic potentials (fEPSPNMDA), or on LTP. Rather, the IC50 for blocking the fEPSPNMDA was 434 nM, and for blocking LTP was 361 nM - both nearly an order of magnitude higher. We conclude that memory impairment produced by systemically administered CPP is not due primarily to its blockade of NMDARs on hippocampal pyramidal neurons. Rather, systemic CPP suppresses memory formation by actions elsewhere in the memory-encoding circuitry.
Collapse
Affiliation(s)
- Kurt Laha
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Mengwen Zhu
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Erin Gemperline
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| | - Vinuta Rau
- Department of Anesthesiology, University of California-San Francisco, San Francisco, CA, USA.
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
| | - Michael S Fanselow
- Departments of Psychology and Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Richard Lennertz
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Robert A Pearce
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
7
|
Platholi J, Hemmings HC. Effects of general anesthetics on synaptic transmission and plasticity. Curr Neuropharmacol 2021; 20:27-54. [PMID: 34344292 PMCID: PMC9199550 DOI: 10.2174/1570159x19666210803105232] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
General anesthetics depress excitatory and/or enhance inhibitory synaptic transmission principally by modulating the function of glutamatergic or GABAergic synapses, respectively, with relative anesthetic agent-specific mechanisms. Synaptic signaling proteins, including ligand- and voltage-gated ion channels, are targeted by general anesthetics to modulate various synaptic mechanisms, including presynaptic neurotransmitter release, postsynaptic receptor signaling, and dendritic spine dynamics to produce their characteristic acute neurophysiological effects. As synaptic structure and plasticity mediate higher-order functions such as learning and memory, long-term synaptic dysfunction following anesthesia may lead to undesirable neurocognitive consequences depending on the specific anesthetic agent and the vulnerability of the population. Here we review the cellular and molecular mechanisms of transient and persistent general anesthetic alterations of synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Jimcy Platholi
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| | - Hugh C Hemmings
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| |
Collapse
|
8
|
Chen MH, Fang C, Wu NY, Xia YH, Zeng YJ, Ouyang W. Genetic variation of rs12918566 affects GRIN2A expression and is associated with spontaneous movement response during sevoflurane anesthesia induction. Brain Behav 2021; 11:e02165. [PMID: 34291608 PMCID: PMC8413822 DOI: 10.1002/brb3.2165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/02/2021] [Accepted: 04/11/2021] [Indexed: 11/09/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors mediate excitatory neurotransmission in the nervous system and are preferentially inhibited by general anesthetics such as sevoflurane. Spontaneous movement is a common complication during sevoflurane anesthesia induction and seriously affects operations. In this study, we investigated the relationship between NMDA polymorphisms and spontaneous movement during sevoflurane induction. This prospective clinical study enrolled 393 patients undergoing sevoflurane anesthesia as part of their surgical routine. In the GRIN1, GRIN2A, and GRIN2B genes, 13 polymorphisms that form a heteromeric complex as part of the NMDA receptor were selected using Haploview and genotyped using matrix-assisted laser desorption ionization-time of flight mass spectrometry MassARRAY. Both RNAfold and Genotype-Tissue Expression portals were used to identify gene expression profiles. Our data showed that 35.8% of subjects exhibited spontaneous movement. The GRIN2A rs12918566 polymorphism was associated with spontaneous movement during sevoflurane induction. A logistic regression analysis of additive, dominant, and recessive models indicated a significant association (odds ratio [OR] (95% confidence limit [CI]): 0.58 (0.42-0.80), p = .00086; OR (95% CI): 0.51 (0.31-0.84), p = .0075, and OR (95% CI): 0.47 (0.27-0.81), p = .0060, respectively). After false discovery rate (FDR) correction, the additive model was still significant with a PFDR =0.010. Bioinformatics demonstrated that the rs12918566 genomic variation affected GRIN2A expression in brain tissue. We also revealed that GRIN2A rs12918566 was significantly associated with spontaneous movement during sevoflurane induction. We believe the NMDA receptor plays an important role in regulating the anesthetic effects of sevoflurane.
Collapse
Affiliation(s)
- Ming-Hua Chen
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Chao Fang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China.,Postdoctoral Research Station of Clinical Medicine, Third Xiangya Hospital of Central South University, Changsha, China
| | - Na-Yiyuan Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu-Hao Xia
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - You-Jie Zeng
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Wen Ouyang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
9
|
Petzold GC, Dreier JP. Spreading depolarization evoked by endothelin-1 is inhibited by octanol but not by carbenoxolone. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
10
|
The escalation in ethanol consumption following chronic intermittent ethanol exposure is blunted in mice expressing ethanol-resistant GluN1 or GluN2A NMDA receptor subunits. Psychopharmacology (Berl) 2021; 238:271-279. [PMID: 33052417 PMCID: PMC7796987 DOI: 10.1007/s00213-020-05680-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
N-Methyl-D-aspartate receptors (NMDARs) are glutamate-gated ion channels essential for glutamatergic transmission and plasticity. NMDARs are inhibited by acute ethanol and undergo brain region-specific adaptations after chronic alcohol exposure. In previous studies, we reported that knock-in mice expressing ethanol-insensitive GluN1 or GluN2A NMDAR subunits display altered behavioral responses to acute ethanol and genotype-dependent changes in drinking using protocols that do not produce dependence. A key unanswered question is whether the intrinsic ethanol sensitivity of NMDARs also plays a role in determining behavioral adaptations that accompany the development of dependence. To test this, we exposed mice to repeated cycles of chronic intermittent ethanol (CIE) vapor known to produce a robust escalation in ethanol consumption and preference. As expected, wild-type mice showed a significant increase from baseline in ethanol consumption and preference after each of the four weekly CIE cycles. In contrast, ethanol consumption in male GluN2A(A825W) mice was unchanged following cycles 1, 2, and 4 of CIE with a modest increase appearing after cycle 3. Wild-type and GluN2A(A825W) female mice did not show a clear or consistent escalation in ethanol consumption or preference following CIE treatment. In male GluN1(F639A) mice, the increase in ethanol consumption observed with their wild-type littermates was delayed until later cycles of exposure. These results suggest that the acute ethanol sensitivity of NMDARs especially those containing the GluN2A subunit may be a critical factor in the escalation of ethanol intake in alcohol dependence.
Collapse
|
11
|
Zhou W, Guan Z. Ion Channels in Anesthesia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:401-413. [DOI: 10.1007/978-981-16-4254-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
12
|
Yuki K, Hou L, Shibamura-Fujiogi M, Koutsogiannaki S, Soriano SG. Mechanistic consideration of the effect of perioperative volatile anesthetics on phagocytes. Clin Immunol 2021; 222:108635. [PMID: 33217544 PMCID: PMC7856197 DOI: 10.1016/j.clim.2020.108635] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/12/2020] [Accepted: 11/14/2020] [Indexed: 02/06/2023]
Abstract
A growing literature has shown that volatile anesthetics are promiscuous molecules targeting multiple molecules, some of which are critical for immunological functions. We focused on studies that delineated target molecules of volatile anesthetics on immune cells and summarized the effects of volatile anesthetics on immune functions. We also presented the perspectives of studying volatile anesthetics-mediated immunomodulation.
Collapse
Affiliation(s)
- Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| | - Lifei Hou
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| | - Miho Shibamura-Fujiogi
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| | - Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| | - Sulpicio G Soriano
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Chen M, Ouyang W, Xia Y, Zeng Y, Wang S, Duan K, Fang C. Association between well‐characterized gene polymorphisms and the hypnosis response caused by sevoflurane‐induced anaesthesia. J Clin Pharm Ther 2020; 45:1442-1451. [PMID: 33016519 DOI: 10.1111/jcpt.13275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/28/2020] [Accepted: 09/07/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Ming‐Hua Chen
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
- Hunan Key Laboratory of Brain Homeostasis Third Xiangya Hospital Central South University Changsha China
| | - Wen Ouyang
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
- Hunan Key Laboratory of Brain Homeostasis Third Xiangya Hospital Central South University Changsha China
| | - Yu‐Hao Xia
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
| | - You‐Jie Zeng
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
| | - Sai‐Ying Wang
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
| | - Kai‐Ming Duan
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
| | - Chao Fang
- Department of Anesthesiology Third Xiangya Hospital Central South University Changsha China
- Hunan Key Laboratory of Brain Homeostasis Third Xiangya Hospital Central South University Changsha China
- Postdoctoral Research Workstation of Clinical Medicine Third Xiangya Hospital Central South University Changsha China
| |
Collapse
|
14
|
Cross-Talk between P2X and NMDA Receptors. Int J Mol Sci 2020; 21:ijms21197187. [PMID: 33003406 PMCID: PMC7582700 DOI: 10.3390/ijms21197187] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 02/01/2023] Open
Abstract
Purinergic P2X receptors (P2X) are ATP-gated ion channels widely expressed in the CNS. While the direct contribution of P2X to synaptic transmission is uncertain, P2X reportedly affect N-methyl-D-aspartate receptor (NMDAR) activity, which has given rise to competing theories on the role of P2X in the modulation of synapses. However, P2X have also been shown to participate in receptor cross-talk: an interaction where one receptor (e.g., P2X2) directly influences the activity of another (e.g., nicotinic, 5-HT3 or GABA receptors). In this study, we tested for interactions between P2X2 or P2X4 and NMDARs. Using two-electrode voltage-clamp electrophysiology experiments in Xenopus laevis oocytes, we demonstrate that both P2X2 and P2X4 interact with NMDARs in an inhibited manner. When investigating the molecular domains responsible for this phenomenon, we found that the P2X2 c-terminus (CT) could interfere with both P2X2 and P2X4 interactions with NMDARs. We also report that 11 distal CT residues on the P2X4 facilitate the P2X4–NMDAR interaction, and that a peptide consisting of these P2X4 CT residues (11C) can disrupt the interaction between NMDARs and P2X2 or P2X4. Collectively, these results provide new evidence for the modulatory nature of P2X2 and P2X4, suggesting they might play a more nuanced role in the CNS.
Collapse
|
15
|
Kubota H, Akaike H, Okamitsu N, Jang IS, Nonaka K, Kotani N, Akaike N. Xenon modulates the GABA and glutamate responses at genuine synaptic levels in rat spinal neurons. Brain Res Bull 2020; 157:51-60. [PMID: 31987927 DOI: 10.1016/j.brainresbull.2020.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 01/11/2023]
Abstract
Effects of xenon (Xe) on whole-cell currents induced by glutamate (Glu), its three ionotropic subtypes, and GABA, as well as on the fast synaptic glutamatergic and GABAergic transmissions, were studied in the mechanically dissociated "synapse bouton preparation" of rat spinal sacral dorsal commissural nucleus (SDCN) neurons. This technique evaluates pure single or multi-synapse responses from native functional nerve endings and enables us to quantify how Xe influences pre- and postsynaptic transmissions accurately. Effects of Xe on glutamate (Glu)-, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-, kainate (KA)- and N-methyl-d-aspartate (NMDA)- and GABAA receptor-mediated whole-cell currents were investigated by the conventional whole-cell patch configuration. Excitatory and inhibitory postsynaptic currents (EPSCs and IPSCs) were measured as spontaneous (s) and evoked (e) EPSCs and IPSCs. Evoked synaptic currents were elicited by paired-pulse focal electric stimulation. Xe decreased Glu, AMPA, KA, and NMDA receptor-mediated whole-cell currents but did not change GABAA receptor-mediated whole-cell currents. Xe decreased the frequency and amplitude but did not affect the 1/e decay time of the glutamatergic sEPSCs. Xe decreased the frequency without affecting the amplitude and 1/e decay time of GABAergic sIPSCs. Xe decreased the amplitude and increased the failure rate (Rf) and paired-pulse ratio (PPR) without altering the 1/e decay time of both eEPSC and eIPSC, suggesting that Xe acts on the presynaptic side of the synapse. The presynaptic inhibition was greater in eEPSCs than in eIPSCs. We conclude that Xe decreases glutamatergic and GABAergic spontaneous and evoked transmissions at the presynaptic level. The glutamatergic presynaptic responses are the main target of anesthesia-induced neuronal responses. In contrast, GABAergic responses minimally contribute to Xe anesthesia.
Collapse
Affiliation(s)
- Hisahiko Kubota
- Department of Pharmacology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hironari Akaike
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Nobuharu Okamitsu
- Department of Electrics and Computer Engineering, Faculty of Engineering, Hiroshima Institute of Technology, 2-1-1 Miyake, Saeki-ku, Hiroshima, 731-5193, Japan
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea
| | - Kiku Nonaka
- Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kita-ku, Kumamoto, 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama, 343-0821, Japan
| | - Norio Akaike
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama, 343-0821, Japan; Research Division for Clinical Pharmacology, Medical Corporation, Juryo Group, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kita-ku, Kumamoto, 860-8518, Japan.
| |
Collapse
|
16
|
Hao X, Ou M, Zhang D, Zhao W, Yang Y, Liu J, Yang H, Zhu T, Li Y, Zhou C. The Effects of General Anesthetics on Synaptic Transmission. Curr Neuropharmacol 2020; 18:936-965. [PMID: 32106800 PMCID: PMC7709148 DOI: 10.2174/1570159x18666200227125854] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
General anesthetics are a class of drugs that target the central nervous system and are widely used for various medical procedures. General anesthetics produce many behavioral changes required for clinical intervention, including amnesia, hypnosis, analgesia, and immobility; while they may also induce side effects like respiration and cardiovascular depressions. Understanding the mechanism of general anesthesia is essential for the development of selective general anesthetics which can preserve wanted pharmacological actions and exclude the side effects and underlying neural toxicities. However, the exact mechanism of how general anesthetics work is still elusive. Various molecular targets have been identified as specific targets for general anesthetics. Among these molecular targets, ion channels are the most principal category, including ligand-gated ionotropic receptors like γ-aminobutyric acid, glutamate and acetylcholine receptors, voltage-gated ion channels like voltage-gated sodium channel, calcium channel and potassium channels, and some second massager coupled channels. For neural functions of the central nervous system, synaptic transmission is the main procedure for which information is transmitted between neurons through brain regions, and intact synaptic function is fundamentally important for almost all the nervous functions, including consciousness, memory, and cognition. Therefore, it is important to understand the effects of general anesthetics on synaptic transmission via modulations of specific ion channels and relevant molecular targets, which can lead to the development of safer general anesthetics with selective actions. The present review will summarize the effects of various general anesthetics on synaptic transmissions and plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yu Li
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| | - Cheng Zhou
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| |
Collapse
|
17
|
Nonaka K, Kotani N, Akaike H, Shin MC, Yamaga T, Nagami H, Akaike N. Xenon modulates synaptic transmission to rat hippocampal CA3 neurons at both pre- and postsynaptic sites. J Physiol 2019; 597:5915-5933. [PMID: 31598974 DOI: 10.1113/jp278762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/07/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Xenon (Xe) non-competitively inhibited whole-cell excitatory glutamatergic current (IGlu ) and whole-cell currents gated by ionotropic glutamate receptors (IAMPA , IKA , INMDA ), but had no effect on inhibitory GABAergic whole-cell current (IGABA ). Xe decreased only the frequency of glutamatergic spontaneous and miniature excitatory postsynaptic currents and GABAergic spontaneous inhibitory postsynaptic currents without changing the amplitude or decay times of these synaptic responses. Xe decreased the amplitude of both the action potential-evoked excitatory and the action potential-evoked inhibitory postsynaptic currents (eEPSCs and eIPSCs, respectively) via a presynaptic inhibition in transmitter release. We conclude that the main site of action of Xe is presynaptic in both excitatory and inhibitory synapses, and that the Xe inhibition is much greater for eEPSCs than for eIPSCs. ABSTRACT To clarify how xenon (Xe) modulates excitatory and inhibitory whole-cell and synaptic responses, we conducted an electrophysiological experiment using the 'synapse bouton preparation' dissociated mechanically from the rat hippocampal CA3 region. This technique can evaluate pure single- or multi-synapse responses and enabled us to accurately quantify how Xe influences pre- and postsynaptic aspects of synaptic transmission. Xe inhibited whole-cell glutamatergic current (IGlu ) and whole-cell currents gated by the three subtypes of glutamate receptor (IAMPA , IKA and INMDA ). Inhibition of these ionotropic currents occurred in a concentration-dependent, non-competitive and voltage-independent manner. Xe markedly depressed the slow steady current component of IAMPA almost without altering the fast phasic IAMPA component non-desensitized by cyclothiazide. It decreased current frequency without affecting the amplitude and current kinetics of glutamatergic spontaneous excitatory postsynaptic currents and miniature excitatory postsynaptic currents. It decreased the amplitude, increasing the failure rate (Rf) and paired-pulse rate (PPR) without altering the current kinetics of glutamatergic action potential-evoked excitatory postsynaptic currents. Thus, Xe has a clear presynaptic effect on excitatory synaptic transmission. Xe did not alter the GABA-induced whole-cell current (IGABA ). It decreased the frequency of GABAergic spontaneous inhibitory postsynaptic currents without changing the amplitude and current kinetics. It decreased the amplitude and increased the PPR and Rf of the GABAergic action potential-evoked inhibitory postsynaptic currents without altering the current kinetics. Thus, Xe acts exclusively at presynaptic sites at the GABAergic synapse. In conclusion, our data indicate that a presynaptic decrease of excitatory transmission is likely to be the major mechanism by which Xe induces anaesthesia, with little contribution of effects on GABAergic synapses.
Collapse
Affiliation(s)
- Kiku Nonaka
- Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kita-ku, Kumamoto, 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama, 343-0821, Japan
| | - Hironari Akaike
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Min-Chul Shin
- Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kita-ku, Kumamoto, 861-5598, Japan
| | - Toshitaka Yamaga
- Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kita-ku, Kumamoto, 861-5598, Japan
| | - Hideaki Nagami
- Research Division for Clinical Pharmacology, Medical Corporation, Juryo Group, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kita-ku, Kumamoto, 860-8518, Japan
| | - Norio Akaike
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama, 343-0821, Japan.,Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.,Research Division for Clinical Pharmacology, Medical Corporation, Juryo Group, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kita-ku, Kumamoto, 860-8518, Japan
| |
Collapse
|
18
|
Abstract
Inhalants are a loosely organized category of abused compounds defined entirely by their common route of administration. Inhalants include volatile solvents, fuels, volatile anesthetics, gasses, and liquefied refrigerants, among others. They are ubiquitous in modern society as ingredients in a wide variety of household, commercial, and medical products. Persons of all ages abuse inhalants but the highest prevalence of abuse is in younger adolescents. Although inhalants have been shown to act upon a host of neurotransmitter receptors, the stimulus effects of the few inhalants which have been trained or tested in drug discrimination procedures suggest that their discriminative stimulus properties are mediated by a few key neurotransmitter receptor systems. Abused volatile solvent inhalants have stimulus effects that are similar to a select group of GABAA positive modulators comprised of benzodiazepines and barbiturates. In contrast the stimulus effects of nitrous oxide gas appear to be at least partially mediated by uncompetitive antagonism of NMDA receptors. Finally, volatile anesthetic inhalants have stimulus effects in common with both GABAA positive modulators as well as competitive NMDA antagonists. In addition to a review of the pharmacology underlying the stimulus effects of inhalants, the chapter also discusses the scientific value of utilizing drug discrimination as a means of functionally grouping inhalants according to their abuse-related pharmacological properties.
Collapse
Affiliation(s)
- Keith L Shelton
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Room 746, P.O. Box 980613, Richmond, VA, 23298-0613, USA.
| |
Collapse
|
19
|
McGinnity CJ, Årstad E, Beck K, Brooks DJ, Coles JP, Duncan JS, Galovic M, Hinz R, Hirani E, Howes OD, Jones PA, Koepp MJ, Luo F, Riaño Barros DA, Singh N, Trigg W, Hammers A. Comment on " In Vivo [ 18F]GE-179 Brain Signal Does Not Show NMDA-Specific Modulation with Drug Challenges in Rodents and Nonhuman Primates". ACS Chem Neurosci 2019; 10:768-772. [PMID: 30346706 DOI: 10.1021/acschemneuro.8b00246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Schoenberger and colleagues ( Schoenberger et al. ( 2018 ) ACS Chem. Neurosci. 9 , 298 - 305 ) recently reported attempts to demonstrate specific binding of the positron emission tomography (PET) radiotracer, [18F]GE-179, to NMDA receptors in both rats and Rhesus macaques. GE-179 did not work as expected in animal models; however, we disagree with the authors' conclusion that "the [18F]GE-179 signal seems to be largely nonspecific". It is extremely challenging to demonstrate specific binding for the use-dependent NMDA receptor intrachannel ligands such as [18F]GE-179 in animals via traditional blocking, due to its low availability of target sites ( Bmax'). Schoenberger and colleagues anesthetized rats and Rhesus monkeys using isoflurane, which has an inhibitory effect on NMDA receptor function and thus would be expected to further reduce the Bmax'. The extent of glutamate release achieved in the provocation experiments is uncertain, as is whether a significant increase in NMDA receptor channel opening can be expected under anesthesia. Prior data suggest that the uptake of disubstituted arylguanidine-based ligands such as GE-179 can be reduced by phencyclidine binding site antagonists, if injection is performed in the absence of ketamine and isoflurane anesthesia, e.g., with GE-179's antecedent, CNS 5161 ( Biegon et al. ( 2007 ) Synapse 61 , 577 - 586 ), and with GMOM ( van der Doef et al. ( 2016 ) J. Cereb. Blood Flow Metab. 36 , 1111 - 1121 ). However, the extent of nonspecific uptake remains uncertain.
Collapse
Affiliation(s)
- Colm J. McGinnity
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, United Kingdom
- King’s
College London & Guy’s and St Thomas’ PET Centre,
St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Erik Årstad
- Institute of Nuclear Medicine and Department of Chemistry, University College London, London NW1 2BU, United Kingdom
| | - Katherine Beck
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, United Kingdom
| | - David J. Brooks
- Department of Nuclear Medicine, Aarhus University, Aarhus 8200, Denmark
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE1 7RU, United Kingdom
| | - Jonathan P. Coles
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - John S. Duncan
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- Epilepsy Society, Gerrards Cross SL9 0RJ, United Kingdom
| | - Marian Galovic
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- Epilepsy Society, Gerrards Cross SL9 0RJ, United Kingdom
- Department of Neurology, Kantonsspital St Gallen, 9007 St. Gallen, Switzerland
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester M20 3LJ, United Kingdom
| | - Ella Hirani
- GE Healthcare Ltd, Amersham HP7 9LL, United Kingdom
| | - Oliver D. Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, United Kingdom
| | | | - Matthias J. Koepp
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- Epilepsy Society, Gerrards Cross SL9 0RJ, United Kingdom
| | - Feng Luo
- GE Healthcare Ltd, Amersham HP7 9LL, United Kingdom
| | - Daniela A. Riaño Barros
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Beckenham, London BR3 3BX, United Kingdom
| | - Nisha Singh
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, United Kingdom
| | | | - Alexander Hammers
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, United Kingdom
- King’s
College London & Guy’s and St Thomas’ PET Centre,
St Thomas’ Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
20
|
Pflanz NC, Daszkowski AW, James KA, Mihic SJ. Ketone body modulation of ligand-gated ion channels. Neuropharmacology 2018; 148:21-30. [PMID: 30562540 DOI: 10.1016/j.neuropharm.2018.12.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/27/2018] [Accepted: 12/10/2018] [Indexed: 01/01/2023]
Abstract
Ketogenesis is a metabolic process wherein ketone bodies are produced from the breakdown of fatty acids. In humans, fatty acid catabolism results in the production of acetyl-CoA which can then be used to synthesize three ketone bodies: acetoacetate, acetone, and β-hydroxybutyrate. Ketogenesis occurs at a higher rate in situations of low blood glucose, such as during fasting, heavy alcohol consumption, and in situations of low insulin, as well as in individuals who follow a 'ketogenic diet' consisting of low carbohydrate and high fat intake. This diet has various therapeutic indications, including reduction of seizure likelihood in epileptic patients and alcohol withdrawal syndrome. However, the mechanisms underlying these therapeutic benefits are still unclear, with studies suggesting various mechanisms such as a shift in energy production in the brain, effects on neurotransmitter production, or effects on various protein targets. Two-electrode voltage clamp electrophysiology in Xenopus laevis oocytes was used to investigate the actions of ketone bodies on three ionotropic receptors: GABAA, glycine, and NMDA receptors. While physiologically-relevant concentrations of acetone have little effect on inhibitory GABA or glycine receptors, β-hydroxybutyrate inhibits the effects of agonists of these receptors at concentrations achieved in vivo. Additionally, both acetone and β-hydroxybutyrate act as inhibitors of glutamate at the excitatory NMDA receptor. Due to the role of hyperexcitability in the pathogenesis of epilepsy and alcohol withdrawal, the inhibitory actions of acetone and β-hydroxybutyrate at NMDA receptors may underlie the therapeutic benefit of a ketogenic diet for these disorders.
Collapse
Affiliation(s)
- Natasha C Pflanz
- Department of Neuroscience, Division of Pharmacology and Toxicology, Waggoner Center for Alcohol and Addiction Research, Institutes for Neuroscience and Cell and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anna W Daszkowski
- Department of Neuroscience, Division of Pharmacology and Toxicology, Waggoner Center for Alcohol and Addiction Research, Institutes for Neuroscience and Cell and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Keith A James
- Department of Neuroscience, Division of Pharmacology and Toxicology, Waggoner Center for Alcohol and Addiction Research, Institutes for Neuroscience and Cell and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - S John Mihic
- Department of Neuroscience, Division of Pharmacology and Toxicology, Waggoner Center for Alcohol and Addiction Research, Institutes for Neuroscience and Cell and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
21
|
Das RK, Walsh K, Hannaford J, Lazzarino AI, Kamboj SK. Nitrous oxide may interfere with the reconsolidation of drinking memories in hazardous drinkers in a prediction-error-dependent manner. Eur Neuropsychopharmacol 2018; 28:828-840. [PMID: 29887289 DOI: 10.1016/j.euroneuro.2018.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/29/2018] [Accepted: 05/17/2018] [Indexed: 12/16/2022]
Abstract
Weakening drinking-related reward memories by blocking their reconsolidation is a potential novel strategy for treating alcohol use disorders. However, few viable pharmacological options exist for reconsolidation interference in humans. We therefore examined whether the NMDA receptor antagonising gas, Nitrous Oxide (N2O) could reduce drinking by preventing the post-retrieval restabilisation of alcohol memories in a group of hazardous drinkers. Critically, we focussed on whether prediction error (PE; a key determinant of reconsolidation) was experienced at retrieval. Sixty hazardous drinkers were randomised to one of three groups that retrieved alcohol memories either with negative PE (Retrieval + PE), no PE (Retrieval no PE) or non-alcohol memory retrieval with PE (No-retrieval +PE). All participants then inhaled 50% N2O for 30 min. The primary outcome was change in beer consumption and alcohol cue-driven urge to drink from the week preceding manipulation (baseline) to the week following manipulation (test). The manipulation did not affect drinking following the intended retrieval+/- PE conditions However, a manipulation check, using a measure of subjective surprise, revealed that the group-level manipulation did not achieve the intended differences in PE at retrieval. Assessment of outcomes according to whether alcohol-relevant PE was actually experienced at retrieval, showed N2O produced reductions in drinking in a retrieval and PE-dependent fashion. These preliminary findings highlight the importance of directly testing assumptions about memory reactivation procedures in reconsolidation research and suggest that N2O should be further investigated as a potential reconsolidation-blocking agent.
Collapse
Affiliation(s)
- R K Das
- Clinical Psychopharmacology Unit, University College London, 1-19 Torrington Place, London WC1E 7HB, United Kingdom; Educational Psychology, Clinical, Educational and Health Psychology, University College London, 26 Bedford Way, London WC1H 0AP, United Kingdom.
| | - K Walsh
- Clinical Psychopharmacology Unit, University College London, 1-19 Torrington Place, London WC1E 7HB, United Kingdom
| | - J Hannaford
- Clinical Psychopharmacology Unit, University College London, 1-19 Torrington Place, London WC1E 7HB, United Kingdom
| | - A I Lazzarino
- Department of Epidemiology and Public Health, UCL, 1-19 Torrington Place, London WC1E 7HB, United Kingdom
| | - S K Kamboj
- Clinical Psychopharmacology Unit, University College London, 1-19 Torrington Place, London WC1E 7HB, United Kingdom
| |
Collapse
|
22
|
Salabert AS, Mora-Ramirez E, Beaurain M, Alonso M, Fontan C, Tahar HB, Boizeau ML, Tafani M, Bardiès M, Payoux P. Evaluation of [ 18F]FNM biodistribution and dosimetry based on whole-body PET imaging of rats. Nucl Med Biol 2017; 59:1-8. [PMID: 29413751 DOI: 10.1016/j.nucmedbio.2017.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/05/2017] [Accepted: 12/17/2017] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The aim of this work was to study the biodistribution, metabolism and radiation dosimetry of rats injected with [18F]FNM using PET/CT images. This novel radiotracer targeting NMDA receptor has potential for investigation for neurological and psychiatric diseases. METHODS Free fraction and stability in fresh human plasma were determined in vitro. PET/CT was performed on anesthetized rats. Organs were identified and 3D volumes of interest (VOIs) were manually drawn on the CT in the center of each organ. Time activity curves (TACs) were created with these VOIs, enabling the calculation of residence times. To confirm these values, ex vivo measurements of organs were performed. Plasma and urine were also collected to study in vivo metabolism. Data was extrapolated to humans, effective doses were estimated using ICRP-60 and ICRP-89 dosimetric models and absorbed doses were estimated using OLINDA/EXM V1.0 and OLINDA/EXM V2.0 (which use weighting factors from ICRP-103 to do the calculations). RESULTS The [18F]FNM was stable in human plasma and the diffusible free fraction was 53%. As with memantine, this tracer is poorly metabolized in vivo. Ex vivo distributions validated PET/CT data as well as demonstrating a decrease of radiotracer uptake in the brain due to anesthesia. Total effective dose was around 6.11 μSv/MBq and 4.65 μSv/MBq for female and male human dosimetric models, respectively. CONCLUSIONS This study shows that the presented compound exhibits stability in plasma and plasma protein binding very similar to memantine. Its dosimetry shows that it is suitable for use in humans due to a low total effective dose compared to other PET radiotracers.
Collapse
Affiliation(s)
- Anne-Sophie Salabert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France; University Hospital, Radiopharmacy Unit, Toulouse, France.
| | - Erick Mora-Ramirez
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France; Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France; Universidad de Costa Rica, CICANUM-Escuela de Física, San José, Costa Rica.
| | - Marie Beaurain
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France; University Hospital, Radiopharmacy Unit, Toulouse, France.
| | - Mathieu Alonso
- University Hospital, Radiopharmacy Unit, Toulouse, France.
| | | | - Hafid Belhadj Tahar
- Research and Expertise Group, French Association for the Promotion of Medical Research (AFPREMED), Toulouse, France.
| | - Marie Laure Boizeau
- Advanced Technology Institute in Life Sciences (ITAV), Centre National de la Recherche Scientifique-Université Paul Sabatier Toulouse III (CNRS-UPS), Université Paul Sabatier Toulouse III (UPS), Université de ToulouseToulouse, France.
| | - Mathieu Tafani
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France; University Hospital, Radiopharmacy Unit, Toulouse, France.
| | - Manuel Bardiès
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France; Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.
| | - Pierre Payoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France; University Hospital, Nuclear Medicine Unit, Toulouse, France.
| |
Collapse
|
23
|
Molecular Mechanisms of Anesthetic Neurotoxicity: A Review of the Current Literature. J Neurosurg Anesthesiol 2017; 28:361-372. [PMID: 27564556 DOI: 10.1097/ana.0000000000000348] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Data from epidemiologic studies and animal models have raised a concern that exposure to anesthetic agents during early postnatal life may cause lasting impairments in cognitive function. It is hypothesized that this is due to disruptions in brain development, but the mechanism underlying this toxic effect remains unknown. Ongoing research, particularly in rodents, has begun to address this question. In this review we examine currently postulated molecular mechanisms of anesthetic toxicity in the developing brain, including effects on cell death pathways, growth factor signaling systems, NMDA and GABA receptors, mitochondria, and epigenetic factors. The level of evidence for each putative mechanism is critically evaluated, and we attempt to draw connections between them where it is possible to do so. Although there are many promising avenues of research, at this time no consensus can be reached as to a definitive mechanism of injury.
Collapse
|
24
|
Davis BM, Brenton J, Davis S, Shamsher E, Sisa C, Grgic L, Cordeiro MF. Assessing anesthetic activity through modulation of the membrane dipole potential. J Lipid Res 2017; 58:1962-1976. [PMID: 28818873 PMCID: PMC5625120 DOI: 10.1194/jlr.m073932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 08/09/2017] [Indexed: 12/21/2022] Open
Abstract
There is great individual variation in response to general anesthetics (GAs) leading to difficulties in optimal dosing and sometimes even accidental awareness during general anesthesia (AAGA). AAGA is a rare, but potentially devastating, complication affecting between 0.1% and 2% of patients undergoing surgery. The development of novel personalized screening techniques to accurately predict a patient’s response to GAs and the risk of AAGA remains an unmet clinical need. In the present study, we demonstrate the principle of using a fluorescent reporter of the membrane dipole potential, di-8-ANEPPs, as a novel method to monitor anesthetic activity using a well-described inducer/noninducer pair. The membrane dipole potential has previously been suggested to contribute a novel mechanism of anesthetic action. We show that the fluorescence ratio of di-8-ANEPPs changed in response to physiological concentrations of the anesthetic, 1-chloro-1,2,2-trifluorocyclobutane (F3), but not the structurally similar noninducer, 1,2-dichlorohexafluorocyclobutane (F6), to artificial membranes and in vitro retinal cell systems. Modulation of the membrane dipole provides an explanation to overcome the limitations associated with the alternative membrane-mediated mechanisms of GA action. Furthermore, by combining this technique with noninvasive retinal imaging technologies, we propose that this technique could provide a novel and noninvasive technique to monitor GA susceptibility and identify patients at risk of AAGA.
Collapse
Affiliation(s)
| | - Jonathan Brenton
- University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Sterenn Davis
- University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Ehtesham Shamsher
- University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Claudia Sisa
- University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Ljuban Grgic
- University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - M Francesca Cordeiro
- University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom .,Western Eye Hospital, Imperial College Healthcare National Health Service Trust, and Imperial College Ophthalmic Research Group, Imperial College London, London NW1 5QH, United Kingdom
| |
Collapse
|
25
|
Yuki K, Eckenhoff RG. Mechanisms of the Immunological Effects of Volatile Anesthetics: A Review. Anesth Analg 2017; 123:326-35. [PMID: 27308954 DOI: 10.1213/ane.0000000000001403] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Volatile anesthetics (VAs) have been in clinical use for a very long time. Their mechanism of action is yet to be fully delineated, but multiple ion channels have been reported as targets for VAs (canonical VA targets). It is increasingly recognized that VAs also manifest effects outside the central nervous system, including on immune cells. However, the literature related to how VAs affect the behavior of immune cells is very limited, but it is of interest that some canonical VA targets are reportedly expressed in immune cells. Here, we review the current literature and describe canonical VA targets expressed in leukocytes and their known roles. In addition, we introduce adhesion molecules called β2 integrins as noncanonical VA targets in leukocytes. Finally, we propose a model for how VAs affect the function of neutrophils, macrophages, and natural killer cells via concerted effects on multiple targets as examples.
Collapse
Affiliation(s)
- Koichi Yuki
- From the *Department of Anesthesiology, Perioperative and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, Massachusetts; †Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts; and ‡Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
26
|
Krystal JH, Petrakis IL, O’Malley S, Krishnan-Sarin S, Pearlson G, Yoon G. NMDA Glutamate Receptor Antagonism and the Heritable Risk for Alcoholism: New Insights from a Study of Nitrous Oxide. Int J Neuropsychopharmacol 2017; 20:351-353. [PMID: 28158462 PMCID: PMC5409033 DOI: 10.1093/ijnp/pyw118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/21/2016] [Accepted: 01/16/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- John H Krystal
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Connecticut (Drs Krystal, Petrakis, O’Malley, Krishnan-Sarin, Pearlson, and Yoon); Psychiatry Services, VA Connecticut Healthcare System, West Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Behavioral Health Services, Yale-New Haven Hospital, New Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Olin Center for Neuropsychiatry Research, Institute of Living, Hartford, Connecticut (Dr Pearlson)
| | - Ismene L Petrakis
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Connecticut (Drs Krystal, Petrakis, O’Malley, Krishnan-Sarin, Pearlson, and Yoon); Psychiatry Services, VA Connecticut Healthcare System, West Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Behavioral Health Services, Yale-New Haven Hospital, New Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Olin Center for Neuropsychiatry Research, Institute of Living, Hartford, Connecticut (Dr Pearlson)
| | - Stephanie O’Malley
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Connecticut (Drs Krystal, Petrakis, O’Malley, Krishnan-Sarin, Pearlson, and Yoon); Psychiatry Services, VA Connecticut Healthcare System, West Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Behavioral Health Services, Yale-New Haven Hospital, New Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Olin Center for Neuropsychiatry Research, Institute of Living, Hartford, Connecticut (Dr Pearlson)
| | - Suchitra Krishnan-Sarin
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Connecticut (Drs Krystal, Petrakis, O’Malley, Krishnan-Sarin, Pearlson, and Yoon); Psychiatry Services, VA Connecticut Healthcare System, West Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Behavioral Health Services, Yale-New Haven Hospital, New Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Olin Center for Neuropsychiatry Research, Institute of Living, Hartford, Connecticut (Dr Pearlson)
| | - Godfrey Pearlson
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Connecticut (Drs Krystal, Petrakis, O’Malley, Krishnan-Sarin, Pearlson, and Yoon); Psychiatry Services, VA Connecticut Healthcare System, West Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Behavioral Health Services, Yale-New Haven Hospital, New Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Olin Center for Neuropsychiatry Research, Institute of Living, Hartford, Connecticut (Dr Pearlson)
| | - Gihyun Yoon
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Connecticut (Drs Krystal, Petrakis, O’Malley, Krishnan-Sarin, Pearlson, and Yoon); Psychiatry Services, VA Connecticut Healthcare System, West Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Behavioral Health Services, Yale-New Haven Hospital, New Haven, Connecticut (Drs Krystal, Petrakis, and Yoon); Olin Center for Neuropsychiatry Research, Institute of Living, Hartford, Connecticut (Dr Pearlson)
| |
Collapse
|
27
|
Sun Y, Zhan L, Cheng X, Zhang L, Hu J, Gao Z. The Regulation of GluN2A by Endogenous and Exogenous Regulators in the Central Nervous System. Cell Mol Neurobiol 2017; 37:389-403. [PMID: 27255970 DOI: 10.1007/s10571-016-0388-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/25/2016] [Indexed: 12/25/2022]
Abstract
The NMDA receptor is the most widely studied ionotropic glutamate receptor, and it is central to many physiological and pathophysiological processes in the central nervous system. GluN2A is one of the two main types of GluN2 NMDA receptor subunits in the forebrain. The proper activity of GluN2A is important to brain function, as the abnormal regulation of GluN2A may induce some neuropsychiatric disorders. This review will examine the regulation of GluN2A by endogenous and exogenous regulators in the central nervous system.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Liying Zhan
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
| | - Xiaokun Cheng
- North China Pharmaceutical Group New Drug Research and Development Co., Ltd, Shijiazhuang, 050015, People's Republic of China
| | - Linan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jie Hu
- School of Nursing, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China.
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
28
|
Clinical concentrations of chemically diverse general anesthetics minimally affect lipid bilayer properties. Proc Natl Acad Sci U S A 2017; 114:3109-3114. [PMID: 28265069 DOI: 10.1073/pnas.1611717114] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
General anesthetics have revolutionized medicine by facilitating invasive procedures, and have thus become essential drugs. However, detailed understanding of their molecular mechanisms remains elusive. A mechanism proposed over a century ago involving unspecified interactions with the lipid bilayer known as the unitary lipid-based hypothesis of anesthetic action, has been challenged by evidence for direct anesthetic interactions with a range of proteins, including transmembrane ion channels. Anesthetic concentrations in the membrane are high (10-100 mM), however, and there is no experimental evidence ruling out a role for the lipid bilayer in their ion channel effects. A recent hypothesis proposes that anesthetic-induced changes in ion channel function result from changes in bilayer lateral pressure that arise from partitioning of anesthetics into the bilayer. We examined the effects of a broad range of chemically diverse general anesthetics and related nonanesthetics on lipid bilayer properties using an established fluorescence assay that senses drug-induced changes in lipid bilayer properties. None of the compounds tested altered bilayer properties sufficiently to produce meaningful changes in ion channel function at clinically relevant concentrations. Even supra-anesthetic concentrations caused minimal bilayer effects, although much higher (toxic) concentrations of certain anesthetic agents did alter lipid bilayer properties. We conclude that general anesthetics have minimal effects on bilayer properties at clinically relevant concentrations, indicating that anesthetic effects on ion channel function are not bilayer-mediated but rather involve direct protein interactions.
Collapse
|
29
|
Hambrecht-Wiedbusch VS, Li D, Mashour GA. Paradoxical Emergence: Administration of Subanesthetic Ketamine during Isoflurane Anesthesia Induces Burst Suppression but Accelerates Recovery. Anesthesiology 2017; 126:482-494. [PMID: 28099246 PMCID: PMC5309196 DOI: 10.1097/aln.0000000000001512] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Promoting arousal by manipulating certain brain regions and/or neurotransmitters has been a recent research focus, with the goal of trying to improve recovery from general anesthesia. The current study tested the hypothesis that a single subanesthetic dose of ketamine during isoflurane anesthesia would increase cholinergic tone in the prefrontal cortex and accelerate recovery. METHODS Adult male rats were implanted with electroencephalography electrodes (frontal, parietal, and occipital cortex) and a microdialysis guide cannula targeted for the prefrontal cortex. After establishing general anesthesia with isoflurane, animals were randomly assigned to receive a saline control or ketamine injection. When isoflurane was discontinued nearly 90 min after drug or saline administration, recovery from anesthesia was measured by experimenters and blinded observers. During the entire experiment, electrophysiologic signals were recorded and acetylcholine was quantified by high-performance liquid chromatography with electrochemical detection. RESULTS A single dose of subanesthetic ketamine caused an initial 125% increase in burst suppression ratio (last isoflurane sample: 37.48 ± 24.11% vs. isoflurane after ketamine injection: 84.36 ± 8.95%; P < 0.0001), but also a significant 44% reduction in emergence time (saline: 877 ± 335 s vs. ketamine: 494 ± 108 s; P = 0.0005; n = 10 per treatment). Furthermore, ketamine caused a significant 317% increase in cortical acetylcholine release (mean after ketamine injection: 0.18 ± 0.16 pmol vs. ketamine recovery: 0.75 ± 0.41 pmol; P = 0.0002) after isoflurane anesthesia was discontinued. CONCLUSIONS Administration of subanesthetic doses of ketamine during isoflurane anesthesia increases anesthetic depth but-paradoxically-accelerates the recovery of consciousness, possibly through cholinergic mechanisms.
Collapse
Affiliation(s)
- Viviane S. Hambrecht-Wiedbusch
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA
- Center of Consciousness Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Duan Li
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA
- Center of Consciousness Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - George A. Mashour
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA
- Center of Consciousness Science, University of Michigan, Ann Arbor, MI 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
30
|
Brosnan RJ, Fukushima FB, Pham TL. Anesthetic synergy between two n-alkanes. Vet Anaesth Analg 2017; 44:577-588. [PMID: 28583773 DOI: 10.1016/j.vaa.2016.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/28/2016] [Accepted: 07/03/2016] [Indexed: 11/17/2022]
Abstract
OBJECTIVE N-butane and n-pentane can both produce general anesthesia. Both compounds potentiate γ-aminobutyric acid type A (GABAA) receptor function, but only butane inhibits N-methyl-d-aspartate (NMDA) receptors. It was hypothesized that butane and pentane would exhibit anesthetic synergy due to their different actions on ligand-gated ion channels. STUDY DESIGN Prospective experimental study. ANIMALS A total of four Xenopus laevis frogs and 43 Sprague-Dawley rats. METHODS Alkane concentrations for all studies were determined via gas chromatography. Using a Xenopus oocyte expression model, standard two-electrode voltage clamp techniques were used to measure NMDA and GABAA receptor responses in vitro as a function of butane and pentane concentrations relevant to anesthesia. The minimum alveolar concentrations (MAC) of butane and pentane were measured separately in rats, and then pentane MAC was measured during coadministration of 0.25, 0.50 or 0.75 times MAC of butane. An isobole with 95% confidence intervals was constructed using regression analysis. A sum of butane and pentane that was statistically less than the lower-end confidence bound isobole indicated a synergistic interaction. RESULTS Both butane and pentane dose-dependently potentiated GABAA receptor currents over the study concentration range. Butane dose-dependently inhibited NMDA receptor currents, but pentane did not modulate NMDA receptors. Butane and pentane MAC in rats was 39.4±0.7 and 13.7±0.4 %, respectively. A small but significant (p<0.03) synergistic anesthetic effect with pentane was observed during administration of either 0.50 or 0.75×MAC butane. CONCLUSIONS Butane and pentane show synergistic anesthetic effects in vivo consistent with their different in vitro receptor effects. CLINICAL RELEVANCE Findings support the relevance of NMDA receptors in mediating anesthetic actions for some, but not all, inhaled agents.
Collapse
Affiliation(s)
- Robert J Brosnan
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA.
| | - Fabíola B Fukushima
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Trung L Pham
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| |
Collapse
|
31
|
Smothers CT, Woodward JJ. Differential effects of TM4 tryptophan mutations on inhibition of N-methyl-d-aspartate receptors by ethanol and toluene. Alcohol 2016; 56:15-19. [PMID: 27814790 DOI: 10.1016/j.alcohol.2016.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/23/2016] [Accepted: 10/01/2016] [Indexed: 10/20/2022]
Abstract
The voluntary use and abuse of alcohol and inhalants is a recognized health problem throughout the world. Previous studies have shown that these agents affect brain function in a variety of ways including direct inhibition of key ion channels that regulate neuronal excitability. Among these, the N-methyl-d-aspartate (NMDA) receptor is particularly important given its key role in glutamatergic synaptic transmission, neuronal plasticity and learning and memory. Previous studies from this laboratory and others have identified key residues within transmembrane (TM) domains of the NMDA receptor that appear to regulate its sensitivity to alcohol and anesthetics. In this study, we extend these findings and examine the role of a TM4 residue in modulating sensitivity of recombinant NMDA receptors to ethanol and toluene. HEK293 cells were transfected with GluN1-1a and either wild-type or tryptophan-substituted GluN2(A-D) subunits and whole-cell currents were recorded using patch-clamp electrophysiology in the absence or presence of ethanol or toluene. Both ethanol (100 mM) and toluene (1 or 3 mM) reversibly inhibited glutamate-activated currents from wild-type NMDARs with GluN2B containing receptors showing heightened sensitivity to either agent. Substitution of tryptophan (W) at positions 825, 826, 823 or 850 in the TM4 domain of GluN2A, GluN2B, GluN2C or GluN2D subunits; respectively, significantly reduced the degree of inhibition by ethanol. In contrast, toluene inhibition of glutamate-activated currents in cells expressing the TM4-W mutants was not different from that of the wild-type controls. These data suggest that despite similarities in their action on NMDARs, ethanol and toluene may act at different sites to reduce ion flux through NMDA receptors.
Collapse
|
32
|
Exploring the Effects on Lipid Bilayer Induced by Noble Gases via Molecular Dynamics Simulations. Sci Rep 2015; 5:17235. [PMID: 26601882 PMCID: PMC4658558 DOI: 10.1038/srep17235] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/27/2015] [Indexed: 12/30/2022] Open
Abstract
Noble gases seem to have no significant effect on the anesthetic targets due to their simple, spherical shape. However, xenon has strong narcotic efficacy and can be used clinically, while other noble gases cannot. The mechanism remains unclear. Here, we performed molecular dynamics simulations on phospholipid bilayers with four kinds of noble gases to elucidate the difference of their effects on the membrane. Our results showed that the sequence of effects on membrane exerted by noble gases from weak to strong was Ne, Ar, Kr and Xe, the same order as their relative narcotic potencies as well as their lipid/water partition percentages. Compared with the other three kinds of noble gases, more xenon molecules were distributed between the lipid tails and headgroups, resulting in membrane’s lateral expansion and lipid tail disorder. It may contribute to xenon’s strong anesthetic potency. The results are well consistent with the membrane mediated mechanism of general anesthesia.
Collapse
|
33
|
Xu M, Smothers CT, Woodward JJ. Cysteine substitution of transmembrane domain amino acids alters the ethanol inhibition of GluN1/GluN2A N-methyl-D-aspartate receptors. J Pharmacol Exp Ther 2015; 353:91-101. [PMID: 25635140 DOI: 10.1124/jpet.114.222034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
N-Methyl-d-aspartate receptors (NMDARs) are inhibited by behaviorally relevant concentrations of ethanol, and residues within transmembrane (TM) domains of NMDARs, including TM3 GluN1 phenylalanine 639 (F639), regulate this sensitivity. In the present study, we used cysteine (C) mutagenesis to determine whether there are additional residues within nearby TM domains that regulate ethanol inhibition on NMDARs. GluN1(F639C)/GluN2A receptors were less inhibited by ethanol than wild-type receptors, and inhibition was restored to wild-type levels following treatment with ethanol-like methanethiosulfonate reagents. Molecular modeling identified six residues in the GluN1 TM1 domain (valine V566; serine S569) and the GluN2A TM4 domain (methionine, M817; V820, F821, and leucine, L824) that were in close vicinity to the TM3 F639 residue, and these were individually mutated to cysteine and tested for ethanol inhibition and receptor function. The F639C-induced decrease in ethanol inhibition was blunted by coexpression of GluN1 TM1 mutants V566C and S569C, and statistically significant interactions were observed for ethanol inhibition among V566C, F639C, and GluN2A TM4 mutants V820C and F821C and S569C, F639C, and GluN2A TM4 mutants F821C and L824C. Ethanol inhibition was also reduced when either GluN1 TM1 mutant V566C or S569C was combined with GluN2A V820C, suggesting a novel TM1:TM4 intrasubunit site of action for ethanol. Cysteines substituted at TM3 and TM4 sites previously suggested to interact with ethanol had less dramatic effects on ethanol inhibition. Overall, the results from these studies suggest that interactions among TM1, TM3, and TM4 amino acids in NMDARs are important determinants of ethanol action at these receptors.
Collapse
Affiliation(s)
- Minfu Xu
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - C Thetford Smothers
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
34
|
Brosnan RJ, Pham TL. Hydrocarbon molar water solubility predicts NMDA vs. GABAA receptor modulation. BMC Pharmacol Toxicol 2014; 15:62. [PMID: 25410726 PMCID: PMC4247779 DOI: 10.1186/2050-6511-15-62] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 11/13/2014] [Indexed: 12/02/2022] Open
Abstract
Background Many anesthetics modulate 3-transmembrane (such as NMDA) and 4-transmembrane (such as GABAA) receptors. Clinical and experimental anesthetics exhibiting receptor family specificity often have low water solubility. We hypothesized that the molar water solubility of a hydrocarbon could be used to predict receptor modulation in vitro. Methods GABAA (α1β2γ2s) or NMDA (NR1/NR2A) receptors were expressed in oocytes and studied using standard two-electrode voltage clamp techniques. Hydrocarbons from 14 different organic functional groups were studied at saturated concentrations, and compounds within each group differed only by the carbon number at the ω-position or within a saturated ring. An effect on GABAA or NMDA receptors was defined as a 10% or greater reversible current change from baseline that was statistically different from zero. Results Hydrocarbon moieties potentiated GABAA and inhibited NMDA receptor currents with at least some members from each functional group modulating both receptor types. A water solubility cut-off for NMDA receptors occurred at 1.1 mM with a 95% CI = 0.45 to 2.8 mM. NMDA receptor cut-off effects were not well correlated with hydrocarbon chain length or molecular volume. No cut-off was observed for GABAA receptors within the solubility range of hydrocarbons studied. Conclusions Hydrocarbon modulation of NMDA receptor function exhibits a molar water solubility cut-off. Differences between unrelated receptor cut-off values suggest that the number, affinity, or efficacy of protein-hydrocarbon interactions at these sites likely differ.
Collapse
Affiliation(s)
- Robert J Brosnan
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | | |
Collapse
|
35
|
Richardson KJ, Shelton KL. N-methyl-D-aspartate receptor channel blocker-like discriminative stimulus effects of nitrous oxide gas. J Pharmacol Exp Ther 2014; 352:156-65. [PMID: 25368340 DOI: 10.1124/jpet.114.218057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Nitrous oxide (N2O) gas is a widely used anesthetic adjunct in dentistry and medicine that is also commonly abused. Studies have shown that N2O alters the function of the N-methyl-d-aspartate (NMDA), GABAA, opioid, and serotonin receptors among others. However, the receptors systems underlying the abuse-related central nervous system effects of N2O are unclear. The present study explores the receptor systems responsible for producing the discriminative stimulus effects of N2O. B6SJLF1/J male mice trained to discriminate 10 minutes of exposure to 60% N2O + 40% oxygen versus 100% oxygen served as subjects. Both the high-affinity NMDA receptor channel blocker (+)-MK-801 maleate [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate] and the low-affinity blocker memantine partially mimicked the stimulus effects of N2O. Neither the competitive NMDA antagonist, CGS-19755 (cis-4-[phosphomethyl]-piperidine-2-carboxylic acid), nor the NMDA glycine-site antagonist, L701-324 [7-chloro-4-hydroxy-3-(3-phenoxy)phenyl-2(1H)-quinolinone], produced N2O-like stimulus effects. A range of GABAA agonists and positive modulators, including midazolam, pentobarbital, muscimol, and gaboxadol (4,5,6,7-tetrahydroisoxazolo[4,5-c]pyridine-3-ol), all failed to produce N2O-like stimulus effects. The μ-, κ-, and δ-opioid agonists, as well as 5-hydroxytryptamine (serotonin) 1B/2C (5-HT1B/2C) and 5-HT1A agonists, also failed to produce N2O-like stimulus effects. Ethanol partially substituted for N2O. Both (+)-MK-801 and ethanol but not midazolam pretreatment also significantly enhanced the discriminative stimulus effects of N2O. Our results support the hypothesis that the discriminative stimulus effects of N2O are at least partially mediated by NMDA antagonist effects similar to those produced by channel blockers. However, as none of the drugs tested fully mimicked the stimulus effects of N2O, other mechanisms may also be involved.
Collapse
Affiliation(s)
- Kellianne J Richardson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Keith L Shelton
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
36
|
Song YK, Lee C, Seo DH, Park SN, Moon SY, Park CH. Interaction between postoperative shivering and hyperalgesia caused by high-dose remifentanil. Korean J Anesthesiol 2014; 66:44-51. [PMID: 24567813 PMCID: PMC3927001 DOI: 10.4097/kjae.2014.66.1.44] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/14/2013] [Accepted: 09/03/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High-dose remifentanil-based anesthesia is associated with opioid-induced hyperalgesia (OIH) and postanesthetic shivering (PAS). These effects can be prevented by N-methyl-d-aspartate (NMDA) receptor antagonists. This study aimed to investigate correlations between OIH and PAS caused by high-dose remifentanil and the effects of low-dose ketamine on OIH and PAS. METHODS Seventy-five patients scheduled for single-port laparoscopic gynecologic surgery were randomly allocated into three groups, each of which received intraoperative remifentanil: group L at 0.1 µg/kg/min; group H at 0.3 µg/kg/min; and group HK at 0.3 µg/kg/min plus 0.25 mg/kg ketamine just before incision, followed by a continuous infusion of 5 µg/kg/min ketamine until skin closure. RESULTS PAS, postoperative tactile pain threshold, and the extent of hyperalgesia in group H were significantly different (P < 0.05) than in the other two groups. PAS was significantly correlated with OIH, including mechanically evoked pain such as postoperative tactile pain threshold (r = -0.529, P = 0.01) (r = -0.458, P = 0.021) and the extent of hyperalgesia (r = 0.537, P = 0.002) (r = 0.384, P = 0.031), respectively, in group H and group HK. Notably, both groups were treated with high-dose remifentanil. Tympanic membrane temperature, time to first postoperative analgesic requirement, postoperative pain scores, analgesic consumption, and cumulative patient-controlled analgesia volume containing morphine were comparable in all three groups. CONCLUSIONS OIH, including the enhanced perception of pain, and PAS were both associated with high-dose remifentanil, were significantly correlated and were attenuated by a low dose of ketamine. This suggests that a common mechanism in part mediated through activation of the central glutamatergic system (e.g., NMDA receptors), underlies the two effects caused by high doses of remifentanil.
Collapse
Affiliation(s)
- Yoon-Kang Song
- Department of Anesthesiology and Pain Medicine, Wonkwang University College of Medicine, Iksan, Korea
| | - Cheol Lee
- Department of Anesthesiology and Pain Medicine, Wonkwang University College of Medicine, Iksan, Korea
| | - Dong-Hyuk Seo
- Department of Anesthesiology and Pain Medicine, Wonkwang University College of Medicine, Iksan, Korea
| | - Seong-Nam Park
- Department of Obstetrics and Gynecology, Wonkwang University College of Medicine, Iksan, Korea
| | - Seo-Young Moon
- Department of Anesthesiology and Pain Medicine, Wonkwang University Sanbon Hospital, Gunpo, Korea
| | - Chang-Hyun Park
- Department of Anesthesiology and Pain Medicine, Presbyterian Medical Center, Jeonju, Korea
| |
Collapse
|
37
|
Petrenko AB, Yamakura T, Sakimura K, Baba H. Defining the role of NMDA receptors in anesthesia: Are we there yet? Eur J Pharmacol 2014; 723:29-37. [DOI: 10.1016/j.ejphar.2013.11.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 11/05/2013] [Accepted: 11/24/2013] [Indexed: 12/26/2022]
|
38
|
Beckley JT, Woodward JJ. Volatile solvents as drugs of abuse: focus on the cortico-mesolimbic circuitry. Neuropsychopharmacology 2013; 38:2555-67. [PMID: 23954847 PMCID: PMC3828545 DOI: 10.1038/npp.2013.206] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/12/2013] [Accepted: 08/13/2013] [Indexed: 12/15/2022]
Abstract
Volatile solvents such as those found in fuels, paints, and thinners are found throughout the world and are used in a variety of industrial applications. However, these compounds are also often intentionally inhaled at high concentrations to produce intoxication. While solvent use has been recognized as a potential drug problem for many years, research on the sites and mechanisms of action of these compounds lags behind that of other drugs of abuse. In this review, we first discuss the epidemiology of voluntary solvent use throughout the world and then consider what is known about their basic pharmacology and how this may explain their use as drugs of abuse. We next present data from preclinical and clinical studies indicating that these substances induce common addiction sequelae such as dependence, withdrawal, and cognitive impairments. We describe how toluene, the most commonly studied psychoactive volatile solvent, alters synaptic transmission in key brain circuits such as the mesolimbic dopamine system and medial prefrontal cortex (mPFC) that are thought to underlie addiction pathology. Finally, we make the case that activity in mPFC circuits is a critical regulator of the mesolimbic dopamine system's ability to respond to volatile solvents like toluene. Overall, this review provides evidence that volatile solvents have high abuse liability because of their selective effects on critical nodes of the addiction neurocircuitry, and underscores the need for more research into how these compounds induce adaptations in neural circuits that underlie addiction pathology.
Collapse
Affiliation(s)
- Jacob T Beckley
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA,Center for Drug and Alcohol Programs, Department of Psychiatry/Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - John J Woodward
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA,Center for Drug and Alcohol Programs, Department of Psychiatry/Neurosciences, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosciences, Medical University of South Carolina, IOP 4 North, 67 President Street, MSC 861, Charleston, SC 29425, USA, Tel: +(843) 792 5225, Fax: +(843) 792 7353, E-mail:
| |
Collapse
|
39
|
den Hartog CR, Beckley JT, Smothers TC, Lench DH, Holseberg ZL, Fedarovich H, Gilstrap MJ, Homanics GE, Woodward JJ. Alterations in ethanol-induced behaviors and consumption in knock-in mice expressing ethanol-resistant NMDA receptors. PLoS One 2013; 8:e80541. [PMID: 24244696 PMCID: PMC3828265 DOI: 10.1371/journal.pone.0080541] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/14/2013] [Indexed: 01/03/2023] Open
Abstract
Ethanol's action on the brain likely reflects altered function of key ion channels such as glutamatergic N-methyl-D-aspartate receptors (NMDARs). In this study, we determined how expression of a mutant GluN1 subunit (F639A) that reduces ethanol inhibition of NMDARs affects ethanol-induced behaviors in mice. Mice homozygous for the F639A allele died prematurely while heterozygous knock-in mice grew and bred normally. Ethanol (44 mM; ∼0.2 g/dl) significantly inhibited NMDA-mediated EPSCs in wild-type mice but had little effect on responses in knock-in mice. Knock-in mice had normal expression of GluN1 and GluN2B protein across different brain regions and a small reduction in levels of GluN2A in medial prefrontal cortex. Ethanol (0.75-2.0 g/kg; i.p.) increased locomotor activity in wild-type mice but had no effect on knock-in mice while MK-801 enhanced activity to the same extent in both groups. Ethanol (2.0 g/kg) reduced rotarod performance equally in both groups but knock-in mice recovered faster following a higher dose (2.5 g/kg). In the elevated zero maze, knock-in mice had a blunted anxiolytic response to ethanol (1.25 g/kg) as compared to wild-type animals. No differences were noted between wild-type and knock-in mice for ethanol-induced loss of righting reflex, sleep time, hypothermia or ethanol metabolism. Knock-in mice consumed less ethanol than wild-type mice during daily limited-access sessions but drank more in an intermittent 24 h access paradigm with no change in taste reactivity or conditioned taste aversion. Overall, these data support the hypothesis that NMDA receptors are important in regulating a specific constellation of effects following exposure to ethanol.
Collapse
Affiliation(s)
- Carolina R. den Hartog
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jacob T. Beckley
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Thetford C. Smothers
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Daniel H. Lench
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Zack L. Holseberg
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Hleb Fedarovich
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Meghin J. Gilstrap
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Gregg E. Homanics
- Departments of Anesthesiology and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - John J. Woodward
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
40
|
Israel Y, Rivera-Meza M, Karahanian E, Quintanilla ME, Tampier L, Morales P, Herrera-Marschitz M. Gene specific modifications unravel ethanol and acetaldehyde actions. Front Behav Neurosci 2013; 7:80. [PMID: 23847486 PMCID: PMC3703538 DOI: 10.3389/fnbeh.2013.00080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 06/18/2013] [Indexed: 12/20/2022] Open
Abstract
Ethanol is metabolized into acetaldehyde mainly by the action of alcohol dehydrogenase in the liver, while mainly by the action of catalase in the brain. Aldehyde dehydrogenase-2 metabolizes acetaldehyde into acetate in both organs. Gene specific modifications reviewed here show that an increased liver generation of acetaldehyde (by transduction of a gene coding for a high-activity liver alcohol dehydrogenase ADH1*B2) leads to increased blood acetaldehyde levels and aversion to ethanol in animals. Similarly aversive is an increased acetaldehyde level resulting from the inhibition of liver aldehyde dehydrogenase-2 (ALDH2) synthesis (by an antisense coding gene against aldh2 mRNA). The situation is diametrically different when acetaldehyde is generated in the brain. When the brain ventral tegmental area (VTA) is endowed with an increased ability to generate acetaldehyde (by transfection of liver rADH) the reinforcing effects of ethanol are increased, while a highly specific inhibition of catalase synthesis (by transduction of a shRNA anti catalase mRNA) virtually abolishes the reinforcing effects of ethanol as seen by a complete abolition of ethanol intake in rats bred for generations as high ethanol drinkers. Data shows two divergent effects of increases in acetaldehyde generation: aversive in the periphery but reinforcing in the brain.
Collapse
Affiliation(s)
- Yedy Israel
- Faculty of Medicine, Molecular and Clinical Pharmacology Program, University of Chile Santiago, Chile ; Department of Pharmacological and Toxicological Chemistry, University of Chile Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
41
|
Booker RD, Sum AK. Biophysical changes induced by xenon on phospholipid bilayers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1347-56. [PMID: 23376329 DOI: 10.1016/j.bbamem.2013.01.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 12/28/2012] [Accepted: 01/22/2013] [Indexed: 12/20/2022]
Abstract
Structural and dynamic changes in cell membrane properties induced by xenon, a volatile anesthetic molecule, may affect the function of membrane-mediated proteins, providing a hypothesis for the mechanism of general anesthetic action. Here, we use molecular dynamics simulation and differential scanning calorimetry to examine the biophysical and thermodynamic effects of xenon on model lipid membranes. Our results indicate that xenon atoms preferentially localize in the hydrophobic core of the lipid bilayer, inducing substantial increases in the area per lipid and bilayer thickness. Xenon depresses the membrane gel-liquid crystalline phase transition temperature, increasing membrane fluidity and lipid head group spacing, while inducing net local ordering effects in a small region of the lipid carbon tails and modulating the bilayer lateral pressure profile. Our results are consistent with a role for nonspecific, lipid bilayer-mediated mechanisms in producing xenon's general anesthetic action.
Collapse
Affiliation(s)
- Ryan D Booker
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO 80401, USA
| | | |
Collapse
|
42
|
Lee C, Lee HW, Kim JN. Effect of oral pregabalin on opioid-induced hyperalgesia in patients undergoing laparo-endoscopic single-site urologic surgery. Korean J Anesthesiol 2013; 64:19-24. [PMID: 23372881 PMCID: PMC3558643 DOI: 10.4097/kjae.2013.64.1.19] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/07/2012] [Accepted: 08/10/2012] [Indexed: 02/08/2023] Open
Abstract
Background Pregabalin is an antiepileptic drug that is effective for treating postoperative pain, neuropathic pain, anxiety, and hemodynamic instability. The aim of this study was to investigate the effect of a single preoperative dose of pregabalin in patients with opioid-induced hyperalgesia (OIH). Methods Ninety ASA I-II patients undergoing laparoendoscopic single-site urologic surgery were randomly assigned to one of the following three groups that received either pregabalin or placebo 1 h before anesthesia and an intraoperative remifentanil infusion. Group plL received placebo and 0.05 µg/kg/min remifentanil, group plH received placebo and 0.3 µg/kg/min remifentanil, and group prH received 300 mg pregabalin plus 0.3 µg/kg/min remifentanil. The primary endpoint was pain intensity upon movement 1, 6, 12, and 24 h after surgery. Secondary endpoints were the area of hyperalgesia and mechanical hyperalgesia threshold 24 h after surgery, time to first postoperative analgesic requirement, and cumulative postoperative volume of morphine administered via a patient-controlled analgesia (PCA) pump over 24 h. Results The time to first postoperative analgesic requirement in group plH was significantly shorter than that in group plL. The injected PCA volume was significantly greater in group plH than that in the other two groups. Postoperative pain intensity in group plH was significantly greater than that in the other two groups at 6, 12, and 24 h after surgery. The mechanical hyperalgesia threshold and the area of hyperalgesia around the surgical incision 24 h after surgery in group plH differed significantly from those in the other two groups, which were not significantly different. Adverse effects were comparable among groups. Conclusions High-dose remifentanil induced hyperalgesia, including increased pain intensity, increased area of hyperalgesia, and decreased mechanical hyperalgesia threshold. These effects were attenuated by oral administration of a single preoperative dose of pregabalin (300 mg) in patients undergoing laparo-endoscopic single-site urologic surgery.
Collapse
Affiliation(s)
- Cheol Lee
- Department of Anesthesiology and Pain Medicine, Wonkwang University College of Medicine, Iksan, Korea
| | | | | |
Collapse
|
43
|
Takeda A, Okita A, Kaneko K, Nagura T, Iwase N, Sekine S, Kakinuma T, Noguchi M, Hatakeyama K. Hypnotic effect of volatile anesthetics is mediated by PKC-γ dynamics. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 118:307-310. [PMID: 23564155 DOI: 10.1007/978-3-7091-1434-6_60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
BACKGROUND Although protein kinase C-γ (PKC-γ) is a target for the effects of volatile anesthetics, the molecular mechanisms of the kinase function remain unclear. We examined the effects of different types of anesthetics on PKC-γ knockout mice, and investigated the dynamics of the kinase in mouse brain. METHODS We measured the required number of times for loss of righting reflex (rtfLORR) after administration of isoflurane, sevoflurane, and propofol on PKC-γ knockout mice and compared with those of wild-type mice. We also used immunoblotting to investigate the intracellular distribution of PKC-γ and phosphorylated PKC-γ (p-PKC-γ) in brain of wild-type mice anesthetized by these anesthetics. RESULTS Isoflurane and sevoflurane significantly prolonged the rtfLORRs in PKC-γ knockout mice compared with those in wild-type mice, while no significant difference was observed between knockout and wild-type mice treated with propofol. Examination of the cellular fractions showed that PKC-γ was significantly decreased, whereas p-PKC-γ was significantly increased in the synaptic membrane fraction (P2). There was no significant change in the supernatant fraction (S). In propofol-treated mice, PKC-γ and p-PKC-γ showed no significant changes in P2 or S. CONCLUSION Our results provide new evidence to support the possibility of the involvement of PKC-γ in the actions of volatile anesthetics.
Collapse
Affiliation(s)
- Akiko Takeda
- Department of Anesthesiology, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Anesthetic protection of neurons injured by hypothermia and rewarming: roles of intracellular Ca2+ and excitotoxicity. Anesthesiology 2012; 117:280-92. [PMID: 22728782 DOI: 10.1097/aln.0b013e318260a7b9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Mild hypothermia is neuroprotective after cerebral ischemia but surgery involving profound hypothermia (PH, temperature less than 18°C) is associated with neurologic complications. Rewarming (RW) from PH injures hippocampal neurons by glutamate excitotoxicity, N-methyl-D-aspartate receptors, and intracellular calcium. Because neurons are protected from hypoxia-ischemia by anesthetic agents that inhibit N-methyl-D-aspartic acid receptors, we tested whether anesthetics protect neurons from damage caused by PH/RW. METHODS Organotypic cultures of rat hippocampus were used to model PH/RW injury, with hypothermia at 4°C followed by RW to 37°C and assessment of cell death 1 or 24 h later. Cell death and intracellular Ca were assessed with fluorescent dye imaging and histology. Anesthetic agents were present in the culture media during PH and RW or only RW. RESULTS Injury to hippocampal CA1, CA3, and dentate neurons after PH and RW involved cell swelling, cell rupture, and adenosine triphosphate (ATP) loss; this injury was similar for 4 through 10 h of PH. Isoflurane (1% and 2%), sevoflurane (3%) and xenon (60%) reduced cell loss but propofol (3 μM) and pentobarbital (100 μM) did not. Isoflurane protection involved reduction in N-methyl-D-aspartate receptor-mediated Ca influx during RW but did not involve γ-amino butyric acid receptors or KATP channels. However, cell death increased over the next day. CONCLUSION Anesthetic protection of neurons rewarmed from 4°C involves suppression of N-methyl-D-aspartate receptor-mediated Ca overload in neurons undergoing ATP loss and excitotoxicity. Unlike during hypoxia/ischemia, anesthetic agents acting predominantly on γ-aminobutyric acid receptors do not protect against PH/RW. The durability of anesthetic protection against cold injury may be limited.
Collapse
|
45
|
Yamamoto E, Akimoto T, Shimizu H, Hirano Y, Yasui M, Yasuoka K. Diffusive nature of xenon anesthetic changes properties of a lipid bilayer: molecular dynamics simulations. J Phys Chem B 2012; 116:8989-95. [PMID: 22715916 DOI: 10.1021/jp303330c] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Effects of general anesthesia can be controllable by the ambient pressure. We perform molecular dynamics simulations for a 1-palmitoyl-2-oleoyl phosphatidylethanolamine lipid bilayer with or without xenon molecules by changing the pressure to elucidate the mechanism of the pressure reversal of general anesthesia. According to the diffusive nature of xenon molecules in the lipid bilayer, a decrease in the orientational order of the lipid tails, an increase in the area and volume per lipid molecule, and an increase in the diffusivity of lipid molecules are observed. We show that the properties of the lipid bilayer with xenon molecules at high pressure come close to those without xenon molecules at 0.1 MPa. Furthermore, we find that xenon molecules are concentrated in the middle of the lipid bilayer at high pressures by the pushing effect and that the diffusivity of xenon molecules is suppressed. These results suggest that the pressure reversal originates from a jamming and suppression of the diffusivity of xenon molecules in lipid bilayers.
Collapse
Affiliation(s)
- Eiji Yamamoto
- Department of Mechanical Engineering, Keio University, 3-4-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Isoflurane enhances both fast and slow synaptic inhibition in the hippocampus at amnestic concentrations. Anesthesiology 2012; 116:816-23. [PMID: 22343472 DOI: 10.1097/aln.0b013e31824be0e3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Inhibition mediated by γ-aminobutyric acid type A (GABA A) receptors has long been considered an important target for a variety of general anesthetics. In the hippocampus, two types of phasic GABA A receptor-mediated inhibition coexist: GABA A,fast, which is expressed primarily at peri-somatic sites, and GABAA,slow, which is expressed primarily in the dendrites. Their spatial segregation suggests distinct functions: GABA A,slow may control plasticity of dendritic synapses, whereas GABA A,fast controls action potential initiation at the soma. We examined modulation of GABA A,fast and GABA A,slow inhibition by isoflurane at amnesic concentrations, and compared it with modulation by behaviorally equivalent doses of the GABA A receptor-selective drug etomidate. METHODS Whole cell recordings were obtained from pyramidal cells in organotypic hippocampal cultures prepared from C57BL/6 × 129/SvJ F1 hybrid mice. GABA A receptor-mediated currents were isolated using glutamate receptor antagonists. GABAA,slow currents were evoked by electrical stimulation in the stratum lacunosum-moleculare. Miniature GABA A,fast currents were recorded in the presence of tetrodotoxin. RESULTS 100 μM isoflurane (approximately EC50,amnesia) slowed fast- and slow-inhibitory postsynaptic current decay by approximately 25%. Higher concentrations, up to 400 μM, produced proportionally greater effects without altering current amplitudes. The effects on GABA A,slow were approximately one-half those produced by equi-amnesic concentrations of etomidate. CONCLUSIONS Isoflurane enhances both types of phasic GABA A receptor-mediated inhibition to similar degrees at amnesic concentrations. This pattern differs from etomidate, which at low concentrations selectively enhances slow inhibition. These effects of isoflurane are sufficiently large that they may contribute substantially to its suppression of hippocampal learning and memory.
Collapse
|
47
|
Antagonism of NMDA receptors by butanesulfonyl-homospermine guanidine and neuroprotective effects in in vitro and in vivo. Neurosci Lett 2011; 506:251-5. [PMID: 22119002 DOI: 10.1016/j.neulet.2011.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/14/2011] [Accepted: 11/10/2011] [Indexed: 12/20/2022]
Abstract
The polyamine derivative BsHSPMG (butanesulfonyl-homospermine with guanidine group) was found to inhibit macroscopic currents strongly at heteromeric N-methyl-D-aspartate (NMDA) receptors (NR1/NR2A and NR1/NR2B) and Ca(2+)-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (homomeric glutamate receptor 1) receptors expressed in Xenopus laevis oocytes on voltage-clamp recording. The IC(50) values of BsHSPMG for NR1/NR2A, NR1/NR2B, NR1/NR2C, and NR1/NR2D receptors were 0.016, 0.021, 5.4, and 9.0 μM, respectively. BsHSPMG inhibited the activity of NR1/NR2A and NR1/NR2B receptors more strongly and did it for those of NR1/NR2C and NR1/NR2D receptors more weakly than a therapeutic drug of Alzheimer's disease, memantine. The inhibition by BsHSPMG was voltage-dependent, since it was prominent at -100 mV compared to that at -20 mV. Mutations including NR1 N616Q, E621Q, N650A, L655A, T807C, NR2B W559L, M562S, W607L, N616Q, and V620E, among others, reduced the inhibition by BsHSPMG, suggesting that BsHSPMG penetrates the channel pore of NMDA receptors deeply. The toxicity of BsHSPMG in neuroblastoma SH-SY5Y cells was much weaker than that of memantine. The effect of BsHSPMG was measured on the focal cerebral ischemia induced by occlusion (1 h) of the middle cerebral artery in mice. BsHSPMG applied before or after occlusion greatly reduced the volume of infarct in mice. These findings demonstrate that BsHSPMG penetrates the NMDA channel pore and exhibits neuroprotective effects against excitatory toxicity in mice.
Collapse
|
48
|
Xu M, Smothers CT, Trudell J, Woodward JJ. Ethanol inhibition of constitutively open N-methyl-D-aspartate receptors. J Pharmacol Exp Ther 2011; 340:218-26. [PMID: 22005043 DOI: 10.1124/jpet.111.187179] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
N-Methyl-D-aspartate (NMDA) receptors gate a slow and calcium-rich component of the postsynaptic glutamate response. Like all ionotropic glutamate receptors, NMDA subunits contain a highly conserved motif (SYTANLAAF) in the transmembrane (TM) 3 domain that is critically involved in channel gating. Mutation of an alanine in this domain (A7; underlined above) results in constitutively open receptors that show reduced sensitivity to several allosteric modulators. In this study, we examined the effects of ethanol, a substance that inhibits NMDA currents via an unknown mechanism, on tonically active NMDA receptors expressed in human embryonic kidney 293 cells. Ethanol (100 mM) inhibited currents from GluN1(A7R)/GluN2A and GluN1(A7R)/GluN2B receptors by approximately 50%, whereas those from GluN1/GluN2B(A7R) receptors were reduced by less than 10%. In cysteine-substituted GluN1 and GluN2 A7 mutants, estimated ethanol IC₅₀ values for agonist-gated currents were 101, 117, 103, and 69 mM for GluN1(A7C)/GluN2A, GluN1(A7C)/GluN2B, GluN1/GluN2A(A7C), and GluN1/GluN2B(A7C) receptors, respectively. After exposure to the thiol-modifying reagent 2-(trimethylammonium)ethyl methanethiosulfonate (MTSET), A7C mutants showed robust agonist-independent currents and reduced sensitivity to ethanol (IC₅₀ values of 371, 256, 715, and 958 mM, respectively, as above). In contrast, cysteine modification of the ligand-binding domain resulted in constitutively open receptors that showed robust ethanol inhibition. Ethanol inhibition of MTSET-treated GluN1(A7C) receptors was further reduced by TM3/TM4 mutations previously shown to reduce ethanol sensitivity of agonist-gated receptors. Overall, these results show that ethanol affects NMDA receptor function at a site distal from agonist binding and appears to exert greater effects via perturbation of GluN2 subunits.
Collapse
Affiliation(s)
- Minfu Xu
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
49
|
Lee C, Song YK, Jeong HM, Park SN. The effects of magnesium sulfate infiltration on perioperative opioid consumption and opioid-induced hyperalgesia in patients undergoing robot-assisted laparoscopic prostatectomy with remifentanil-based anesthesia. Korean J Anesthesiol 2011; 61:244-50. [PMID: 22025948 PMCID: PMC3198187 DOI: 10.4097/kjae.2011.61.3.244] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 03/14/2011] [Accepted: 03/14/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Opioids not only exert an antinociceptive effect, but also modulate central N-methyl-D-aspartate (NMDA) receptors, resulting in hyperalgesia and acute opioid tolerance. This study was aimed to investigate the effect of the NMDA receptor antagonist, magnesium in preventing remifentanil-induced hyperalgesia. METHODS For this study, 75 patients scheduled for robot-assisted laparoscopic prostatectomy were randomly allocated into three groups of patients whose incision sites were infiltrated: Group M, with 25% magnesium sulfate 80 mg/kg; Group S, with the same volume of saline under remifentanil-based anesthesia, and Group D, with the same volume of saline under desflurane based anesthesia. All three groups were infiltrated into incision sites after pneumoperitoneum. Intraoperative evaluation included mean remifentanil dose, and postoperative evaluation included pain severity at time intervals of 30 min, 6, 12, 24 and 36 hours, time to first postoperative analgesic requirement, and analgesic dosage required during 24 hours. RESULTS Mean remifentanil doses during the intraoperative periods in group M were significantly lower than those in group S (P < 0.001). The time to first postoperative analgesic requirement in postoperative period in groups M and D was significantly longer than that in group S (P < 0.001). Visual analog scale scores for pain in groups M and D were significantly lower than those in group S for 12 hours after operation. CONCLUSIONS A relatively high dose and continuous infusion of remifentanil were associated with opioid induced hyperalgesia. Wound infiltration with magnesium sulfate decreased opioid consumption and reduces opioid induced hyperalgesia.
Collapse
Affiliation(s)
- Cheol Lee
- Department of Anesthesiology and Pain Medicine, Wonkwang University School of Medicine, Iksan, Korea
| | | | | | | |
Collapse
|
50
|
Brosnan RJ. GABA(A) receptor antagonism increases NMDA receptor inhibition by isoflurane at a minimum alveolar concentration. Vet Anaesth Analg 2011; 38:231-9. [PMID: 21492389 DOI: 10.1111/j.1467-2995.2011.00605.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE At the minimum alveolar concentration (MAC), isoflurane potentiates GABA(A) receptor currents and inhibits NMDA receptor currents, and these actions may be important for producing anesthesia. However, isoflurane modulates GABA(A) receptors more potently than NMDA receptors. The objective of this study was to test whether isoflurane would function as a more potent NMDA receptor antagonist if its efficacy at GABA(A) receptors was decreased. STUDY DESIGN Prospective experimental study. ANIMALS Fourteen 10-week-old male Sprague-Dawley rats weighing 269 ± 12 g. METHODS Indwelling lumbar subarachnoid catheters were surgically placed in isoflurane-anesthetized rats. Two days later, the rats were anesthetized with isoflurane, and artificial CSF containing either 0 or 1 mg kg(-1) picrotoxin, a GABA(A) receptor antagonist, was infused intrathecally at 1 μL minute(-1). The baseline isoflurane MAC was then determined using a standard tail clamp technique. MK801 (dizocilpine), an NMDA receptor antagonist, was then administered intravenously at 0.5 mg kg(-1). Isoflurane MAC was re-measured. RESULTS Picrotoxin increased isoflurane MAC by 16% compared to controls. MK801 significantly decreased isoflurane MAC by 0.72% of an atmosphere in controls versus 0.47% of an atmosphere in rats receiving intrathecal picrotoxin. CONCLUSIONS AND CLINICAL RELEVANCE A smaller MK801 MAC-sparing effect in the picrotoxin group is consistent with greater NMDA antagonism by isoflurane in these animals, since it suggests that fewer NMDA receptors are available upon which MK801 could act to decrease isoflurane MAC. Decreasing isoflurane GABA(A) potentiation increases isoflurane NMDA antagonism at MAC. Hence, the magnitude of an anesthetic effect on a given channel or receptor at MAC may depend upon effects at other receptors.
Collapse
Affiliation(s)
- Robert J Brosnan
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| |
Collapse
|