1
|
Ozdemi̇r C, Isik B, Koca G, Inan MA. Effects of mid‑gestational sevoflurane and magnesium sulfate on maternal oxidative stress, inflammation and fetal brain histopathology. Exp Ther Med 2024; 28:286. [PMID: 38827470 PMCID: PMC11140313 DOI: 10.3892/etm.2024.12574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 03/08/2024] [Indexed: 06/04/2024] Open
Abstract
Models of inflammation, oxidative stress, hyperoxia and hypoxia have demonstrated that magnesium sulfate (MgSO4), a commonly used drug in obstetrics, has neuroprotective potential. In the present study, the effects of MgSO4 treatment on inflammation, oxidative stress and fetal brain histopathology were evaluated in an experimental rat model following sevoflurane (Sv) exposure during the mid-gestational period. Rats were randomly divided into groups: C (control; no injections or anesthesia), Sv (exposure to 2.5% Sv for 2 h), MgSO4 (administered 270 mg/kg MgSO4 intraperitoneally) and Sv + MgSO4 (Sv administered 30 min after MgSO4 injection). Inflammatory and oxidative stress markers were measured in the serum and neurotoxicity was investigated histopathologically in fetal brain tissue. Short-term mid-gestational exposure to a 1.1 minimum alveolar concentration of Sv did not significantly increase the levels of any of the measured biochemical markers, except for TNF-α. Histopathological evaluations demonstrated no findings suggestive of pathological apoptosis, neuroinflammation or oxidative stress-induced cell damage. MgSO4 injection prior to anesthesia caused no significant differences in biochemical or histopathological marker levels compared to the C and Sv groups. The present study indicated that short-term exposure to Sv could potentially be considered a harmless external stimulus to the fetal brain.
Collapse
Affiliation(s)
- Cagri Ozdemi̇r
- Department of Anesthesiology and Reanimation, Mamak State Hospital, 06270 Ankara, Turkey
| | - Berrin Isik
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, 06560 Ankara, Turkey
| | - Gulce Koca
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, 06560 Ankara, Turkey
| | - Mehmet Arda Inan
- Department of Medical Pathology, Faculty of Medicine, Gazi University, 06560 Ankara, Turkey
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Zhang Y, Sun Q, Fan A, Dong G. Isoflurane triggers the acute cognitive impairment of aged rats by damaging hippocampal neurons via the NR2B/CaMKII/CREB pathway. Behav Brain Res 2021; 405:113202. [PMID: 33636236 DOI: 10.1016/j.bbr.2021.113202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 11/18/2022]
Abstract
Isoflurane was responsible for acute neuronal impairment, but its potential molecular mechanisms in damaging hippocampal neurons had not been clearly understood. This study aimed to explore the underlying mechanism of how isoflurane affected the cognitive function of aged rats by damaging the hippocampal neurons. Acute cognitive impairment was found in aged Wistar rats via Morris water maze test and Y-maze test after isoflurane anesthesia in a dose-dependent manner compared with the control group in vivo. Isoflurane also decreased the viabilities and strengthened the apoptotic potential of hippocampal neurons by damaging the mitochondria in a time-dependent manner compared with the control group which was reported by MTT, immunofluorescent assay, flow cytometry and western blot assay in vitro. Isoflurane jeopardized hippocampal neurons by directly inactivating the NR2B/CaMKII/CREB pathway and its harmful effects could be ameliorated by adding CaMKII activator CdCl2. These findings provided evidence that the cognitive ability of aged rats was injured by isoflurane exposure and isoflurane also inhibited the viability and enhanced the apoptosis of hippocampal neurons by damaging the mitochondria through inhibition of the NR2B/CaMKII/CREB pathway and its harmful roles could be partially ameliorated by CdCl2. Our study demonstrated that isoflurane could cause acute neuronal damage and we provided fresh insights that contributed to the safe use of anesthetic agents and the prevention of PND in elderly people.
Collapse
Affiliation(s)
- Yuangui Zhang
- Department of Anesthesiology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang City, Shandong Province, 261000, China
| | - Qingqing Sun
- Department of Anesthesiology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang City, Shandong Province, 261000, China
| | - Aixia Fan
- Department of Anesthesiology, Xintai People's Hospital, No. 1329, Xinfu Road, Xintai City, Shandong Province, 271200, China
| | - Guimin Dong
- Department of Anesthesiology, Weifang People's Hospital, No. 151, Guangwen Street, Kuiwen District, Weifang City, Shandong Province, 261000, China.
| |
Collapse
|
5
|
Iqbal F, Thompson AJ, Riaz S, Pehar M, Rice T, Syed NI. Anesthetics: from modes of action to unconsciousness and neurotoxicity. J Neurophysiol 2019; 122:760-787. [PMID: 31242059 DOI: 10.1152/jn.00210.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern anesthetic compounds and advanced monitoring tools have revolutionized the field of medicine, allowing for complex surgical procedures to occur safely and effectively. Faster induction times and quicker recovery periods of current anesthetic agents have also helped reduce health care costs significantly. Moreover, extensive research has allowed for a better understanding of anesthetic modes of action, thus facilitating the development of more effective and safer compounds. Notwithstanding the realization that anesthetics are a prerequisite to all surgical procedures, evidence is emerging to support the notion that exposure of the developing brain to certain anesthetics may impact future brain development and function. Whereas the data in support of this postulate from human studies is equivocal, the vast majority of animal research strongly suggests that anesthetics are indeed cytotoxic at multiple brain structure and function levels. In this review, we first highlight various modes of anesthetic action and then debate the evidence of harm from both basic science and clinical studies perspectives. We present evidence from animal and human studies vis-à-vis the possible detrimental effects of anesthetic agents on both the young developing and the elderly aging brain while discussing potential ways to mitigate these effects. We hope that this review will, on the one hand, invoke debate vis-à-vis the evidence of anesthetic harm in young children and the elderly, and on the other hand, incentivize the search for better and less toxic anesthetic compounds.
Collapse
Affiliation(s)
- Fahad Iqbal
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew J Thompson
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Saba Riaz
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marcus Pehar
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Rice
- Department of Anesthesiology, Perioperative and Pain Medicine, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Naweed I Syed
- Vi Riddell Pain Program, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
6
|
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
|
7
|
Brosnan RJ, Pham TL. Anesthetic-sensitive ion channel modulation is associated with a molar water solubility cut-off. BMC Pharmacol Toxicol 2018; 19:57. [PMID: 30217234 PMCID: PMC6137927 DOI: 10.1186/s40360-018-0244-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/22/2018] [Indexed: 12/31/2022] Open
Abstract
Background NMDA receptor modulation by hydrocarbons is associated with a molar water solubility cut-off. Low-affinity phenolic modulation of GABAA receptors is also associated with a cut-off, but at much lower molar solubility values. We hypothesized that other anesthetic-sensitive ion channels exhibit distinct cut-off effects associated with hydrocarbon molar water solubility, and that cut-off values are comparatively similar between related receptors than phylogenetically distant ones. Methods Glycine or GABAA receptors or TREK-1, TRESK, Nav1.2, or Nav1.4 channels were expressed separately in frog oocytes. Two electrode voltage clamp techniques were used to study current responses in the presence and absence of hydrocarbon series from eight functional groups with progressively increasing size at saturated aqueous concentrations. Null response (cut-off) was defined by current measurements that were statistically indistinguishable between baseline and hydrocarbon exposure. Results Ion channels exhibited cut-off effects associated with hydrocarbon molar water solubility in the following order of decreasing solubility: Nav1.2 ≈ Nav1.4 ≳ TRESK ≈ TREK-1 > GABAA >> glycine. Previously measured solubility cut-off values for NMDA receptors were intermediate between those for Nav1.4 and TRESK. Conclusions Water solubility cut-off responses were present for all anesthetic-sensitive ion channels; distinct cut-off effects may exist for all cell surface receptors that are sensitive to volatile anesthetics. Suggested is the presence of amphipathic receptor sites normally occupied by water molecules that have dissociation constants inversely related to the cut-off solubility value. Poorly soluble hydrocarbons unable to reach concentrations sufficient to out-compete water for binding site access fail to modulate the receptor.
Collapse
Affiliation(s)
- Robert J Brosnan
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| | - Trung L Pham
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
8
|
Uchida T, Shimada K, Tanabe R, Kubota T, Ito D, Yamazaki K, Gohara K. Xenon pressure dependence on the synchronized burst inhibition of rat cortical neuronal network cultured on multi-electrode arrays. IBRO Rep 2018; 3:45-54. [PMID: 30135941 PMCID: PMC6084913 DOI: 10.1016/j.ibror.2017.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/04/2017] [Accepted: 09/01/2017] [Indexed: 01/11/2023] Open
Abstract
Mature rat cortical neuronal networks cultured on multi-electrode arrays (MEAs) are known to show spontaneous synchronized bursts accompanied by independent single spikes. The spontaneous synchronized bursts can be inhibited by Xe gas. In this study, we adjust the Xe gas pressure to control the amount of Xe in a neuron-cultured MEA medium. We show that the synchronized bursts cease completely within several minutes by applying Xe gas at partial pressures above 0.3 MPa. After depressurizing and purging with fresh air, the synchronized bursts recover to their original frequency. Thus, we confirmed that Xe acts as a network-activity inhibitor of the cultured neuronal network on MEAs. But below 0.3 MPa, the synchronized bursts are inhibited only partially, depending on the Xe partial pressure. Based on the partial-pressure influence on the change of the neuronal network activities, we find the critical concentration of Xe for the inhibition effect to be approximately 9.5 mM, a value above which more than 90% of the synchronized burst activity is inhibited. Further systematic observations with Xe-air mixed gases show that pressurized air with a small amount of Xe suppresses the inhibition of synchronized bursts, suggesting an air component that can accelerate the synchronized bursts.
Collapse
Affiliation(s)
- Tsutomu Uchida
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| | - Koichiro Shimada
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| | - Ryutaro Tanabe
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| | - Tatsuya Kubota
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| | - Daisuke Ito
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| | - Kenji Yamazaki
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| | - Kazutoshi Gohara
- Division of Applied Physics, Faculty of Engineering, Hokkaido University, N13 W8 Kita-ku, Sapporo, Hokkaido 062-8628, Japan
| |
Collapse
|
9
|
Zhang X, Shen F, Xu D, Zhao X. A lasting effect of postnatal sevoflurane anesthesia on the composition of NMDA receptor subunits in rat prefrontal cortex. Int J Dev Neurosci 2016; 54:62-69. [PMID: 27025552 DOI: 10.1016/j.ijdevneu.2016.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/21/2016] [Accepted: 01/21/2016] [Indexed: 01/28/2023] Open
Abstract
Sevoflurane is widely used in pediatric anesthesia and studies have shown that it is capable of inducing neurodegeneration and subsequent cognitive disorders in the developing brain. However, the evidence that anesthetics are toxic to the human brain is insufficient. N-Methyl-d-aspartate (NMDA) receptors, critical for learning and memory, display expression changes with age and can be modulated by inhalation anesthetics. Generally, NMDA receptor (NR) type 1 is expressed at birth, peaks around the third postnatal week, and then declines slightly to adult levels. NR2Bs slowly decrease and NR2As gradually increase during postnatal development. These developmental switches of NMDA receptor subunits composition mark the transition from immature to adult neural processing and allow for the final maturation of associative learning abilities. In this study, we aimed to evaluate the effect of repeated sevoflurane anesthesia on NMDA receptor subunits composition in the developing rat brain and related behavioral disorders. Six-day-old male Sprague Dawley rats were randomly allocated into either a control group (group con) or a sevoflurane group (group sevo). Group sevo inhaled 2.1% sevoflurane carried by 70% oxygen for 2h each day from postnatal day (PND) 6 to PND 8. The same procedure, without applying the sevoflurane, was executed in group con. The membrane protein expression of NR1, NR2A and NR2B in the prefrontal cortex (PFC) and hippocampus was assessed at the end of the three days of anesthesia and at PND 21. An open field test was carried out to assess spontaneous locomotion on PNDs 21, 28 and 35. Y maze performance was used to assess attention and working memory on PND 28. Sevoflurane induced upregulation of NR1 and NR2B in the PFC at the end of anesthesia. On PND 21, NR1 and NR2B receptors were significantly increased whereas NR2A receptors were significantly decreased in the PFC in group sevo. Sevoflurane-treated rats showed hyper-locomotion and impairment of working memory in the behavior tests. These results indicate that repeated sevoflurane anesthesia at early stage of life can induce a long lasting effect of interfering with NMDA receptor subunits composition in rat PFC. These changes may contribute to the effects of sevoflurane on neuronal development and subsequent neurobehavioral disorders.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- International Peace Maternity & Child Health Hospital, Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, China
| | - Fengyan Shen
- Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Daojie Xu
- Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Xuan Zhao
- Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China.
| |
Collapse
|
10
|
Wang H, Xu Z, Wu A, Dong Y, Zhang Y, Yue Y, Xie Z. 2-deoxy-D-glucose enhances anesthetic effects in mice. Anesth Analg 2015; 120:312-9. [PMID: 25390277 DOI: 10.1213/ane.0000000000000520] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND The mechanisms of general anesthesia by volatile drugs remain largely unknown. Mitochondrial dysfunction and reduction in energy levels have been suggested to be associated with general anesthesia status. 2-Deoxy-D-glucose (2-DG), an analog of glucose, inhibits hexokinase and reduces cellular levels of adenosine triphosphate (ATP). 3-Nitropropionic acid is another compound which can deplete ATP levels. In contrast, idebenone and L-carnitine could rescue deficits of energy. We therefore sought to determine whether 2-DG and/or 3-nitropropionic acid can enhance the anesthetic effects of isoflurane, and whether idebenone and L-carnitine can reverse the actions of 2-DG. METHODS C57BL/6J mice (8 months old) received different concentrations of isoflurane with and without the treatments of 2-DG, 3-nitropropionic acid, idebenone, and L-carnitine. Isoflurane-induced loss of righting reflex (LORR) was determined in the mice. ATP levels in H4 human neuroglioma cells were assessed after these treatments. Finally, 31P-magnetic resonance spectroscopy was used to determine the effects of isoflurane on brain ATP levels in the mice. RESULTS 2-DG enhanced isoflurane-induced LORR (P = 0.002, N = 15). 3-Nitropropionic acid also enhanced the anesthetic effects of isoflurane (P = 0.005, N = 15). Idebenone (idebenone + saline versus idebenone + 2-DG: P = 0.165, N = 15), but not L-carnitine (L-carnitine + saline versus L-carnitine + 2-DG: P < 0.0001, N = 15), inhibited the effects of 2-DG on enhancing isoflurane-induced LORR in the mice, as evidenced by 2-DG not enhancing isoflurane-induced LORR in the mice pretreated with idebenone. Idebenone (idebenone + saline versus idebenone + 2-DG: P = 0.177, N = 6), but not L-carnitine (L-carnitine + saline versus L-carnitine + 2-DG: P = 0.029, N = 6), also mitigated the effects of 2-DG on reducing ATP levels in cells, as evidenced by 2-DG not decreasing ATP levels in the cells pretreated with idebenone. Finally, isoflurane decreased ATP levels in both cultured cells and mouse brains (β-ATP: P = 0.003, N = 10; β-ATP/phosphocreatine: P = 0.006, N = 10; β-ATP/inorganic phosphate: P = 0.001, N = 10). CONCLUSIONS These results from our pilot studies have established a system and generated a hypothesis that 2-DG enhances anesthetic effects via reducing energy levels. These findings should promote further studies to investigate anesthesia mechanisms.
Collapse
Affiliation(s)
- Hui Wang
- From the *Department of Anesthesia, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China; †Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts; and ‡Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
11
|
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]
|
12
|
Quintana A, Morgan PG, Kruse SE, Palmiter RD, Sedensky MM. Altered anesthetic sensitivity of mice lacking Ndufs4, a subunit of mitochondrial complex I. PLoS One 2012; 7:e42904. [PMID: 22912761 PMCID: PMC3422219 DOI: 10.1371/journal.pone.0042904] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/12/2012] [Indexed: 02/07/2023] Open
Abstract
Anesthetics are in routine use, yet the mechanisms underlying their function are incompletely understood. Studies in vitro demonstrate that both GABA(A) and NMDA receptors are modulated by anesthetics, but whole animal models have not supported the role of these receptors as sole effectors of general anesthesia. Findings in C. elegans and in children reveal that defects in mitochondrial complex I can cause hypersensitivity to volatile anesthetics. Here, we tested a knockout (KO) mouse with reduced complex I function due to inactivation of the Ndufs4 gene, which encodes one of the subunits of complex I. We tested these KO mice with two volatile and two non-volatile anesthetics. KO and wild-type (WT) mice were anesthetized with isoflurane, halothane, propofol or ketamine at post-natal (PN) days 23 to 27, and tested for loss of response to tail clamp (isoflurane and halothane) or loss of righting reflex (propofol and ketamine). KO mice were 2.5 - to 3-fold more sensitive to isoflurane and halothane than WT mice. KO mice were 2-fold more sensitive to propofol but resistant to ketamine. These changes in anesthetic sensitivity are the largest recorded in a mammal.
Collapse
Affiliation(s)
- Albert Quintana
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Philip G. Morgan
- Department of Anesthesiology and Pain Medicine, University of Washington and Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington, United States of America
- * E-mail:
| | - Shane E. Kruse
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Richard D. Palmiter
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Margaret M. Sedensky
- Department of Anesthesiology and Pain Medicine, University of Washington and Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington, United States of America
| |
Collapse
|
13
|
Brosnan RJ, Thiesen R. Increased NMDA receptor inhibition at an increased Sevoflurane MAC. BMC Anesthesiol 2012; 12:9. [PMID: 22672766 PMCID: PMC3439310 DOI: 10.1186/1471-2253-12-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 06/06/2012] [Indexed: 11/10/2022] Open
Abstract
Background Sevoflurane potently enhances glycine receptor currents and more modestly decreases NMDA receptor currents, each of which may contribute to immobility. This modest NMDA receptor antagonism by sevoflurane at a minimum alveolar concentration (MAC) could be reciprocally related to large potentiation of other inhibitory ion channels. If so, then reduced glycine receptor potency should increase NMDA receptor antagonism by sevoflurane at MAC. Methods Indwelling lumbar subarachnoid catheters were surgically placed in 14 anesthetized rats. Rats were anesthetized with sevoflurane the next day, and a pre-infusion sevoflurane MAC was measured in duplicate using a tail clamp method. Artificial CSF (aCSF) containing either 0 or 4 mg/mL strychnine was then infused intrathecally at 4 μL/min, and the post-infusion baseline sevoflurane MAC was measured. Finally, aCSF containing strychnine (either 0 or 4 mg/mL) plus 0.4 mg/mL dizocilpine (MK-801) was administered intrathecally at 4 μL/min, and the post-dizocilpine sevoflurane MAC was measured. Results Pre-infusion sevoflurane MAC was 2.26%. Intrathecal aCSF alone did not affect MAC, but intrathecal strychnine significantly increased sevoflurane requirement. Addition of dizocilpine significantly decreased MAC in all rats, but this decrease was two times larger in rats without intrathecal strychnine compared to rats with intrathecal strychnine, a statistically significant (P < 0.005) difference that is consistent with increased NMDA receptor antagonism by sevoflurane in rats receiving strychnine. Conclusions Glycine receptor antagonism increases NMDA receptor antagonism by sevoflurane at MAC. The magnitude of anesthetic effects on a given ion channel may therefore depend on the magnitude of its effects on other receptors that modulate neuronal excitability.
Collapse
Affiliation(s)
- Robert J Brosnan
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| | | |
Collapse
|
14
|
Mawhinney LJ, de Rivero Vaccari JP, Alonso OF, Jimenez CA, Furones C, Moreno WJ, Lewis MC, Dietrich WD, Bramlett HM. Isoflurane/nitrous oxide anesthesia induces increases in NMDA receptor subunit NR2B protein expression in the aged rat brain. Brain Res 2011; 1431:23-34. [PMID: 22137658 DOI: 10.1016/j.brainres.2011.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 10/13/2011] [Accepted: 11/02/2011] [Indexed: 10/15/2022]
Abstract
Postoperative cognitive dysfunction, POCD, afflicts a large number of elderly surgical patients following surgery with general anesthesia. Mechanisms of POCD remain unclear. N-methyl-D-aspartate (NMDA) receptors, critical in learning and memory, that display protein expression changes with age are modulated by inhalation anesthetics. The aim of this study was to identify protein expression changes in NMDA receptor subunits and downstream signaling pathways in aged rats that demonstrated anesthesia-induced spatial learning impairments. Three-month-old and 18-month-old male Fischer 344 rats were randomly assigned to receive 1.8% isoflurane/70% nitrous oxide (N(2)O) anesthesia for 4h or no anesthesia. Spatial learning was assessed at 2weeks and 3months post-anesthesia in Morris water maze. Hippocampal and cortical protein lysates of 18-month-old rats were immunoblotted for activated caspase 3, NMDA receptor subunits, and extracellular-signal regulated kinase (ERK) 1/2. In a separate experiment, Ro 25-6981 (0.5mg/kg dose) was administered by I.P. injection before anesthesia to 18-month-old rats. Immunoblotting of NR2B was performed on hippocampal protein lysates. At 3months post-anesthesia, rats treated with anesthesia at 18-months-old demonstrated spatial learning impairment corresponding to acute and long-term increases in NR2B protein expression and a reduction in phospho-ERK1/2 in the hippocampus and cortex. Ro 25-6981 pretreatment attenuated the increase in acute NR2B protein expression. Our findings suggest a role for disruption of NMDA receptor mediated signaling pathways in the hippocampus and cortex of rats treated with isoflurane/ N(2)O anesthesia at 18-months-old, leading to spatial learning deficits in these animals. A potential therapeutic intervention for anesthesia associated cognitive deficits is discussed.
Collapse
Affiliation(s)
- Lana J Mawhinney
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Zhao YL, Xiang Q, Shi QY, Li SY, Tan L, Wang JT, Jin XG, Luo AL. GABAergic excitotoxicity injury of the immature hippocampal pyramidal neurons' exposure to isoflurane. Anesth Analg 2011; 113:1152-60. [PMID: 21918167 DOI: 10.1213/ane.0b013e318230b3fd] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Certain anesthetics exhibit neurotoxicity in the brains of immature but not mature animals. γ-Aminobutyric acid (GABA), the primary inhibitory neurotransmitter in the adult brain, is excitatory on immature neurons via its action at the GABAA receptor, depolarizing the membrane potential and inducing a cytosolic Ca2+ increase ([Ca2+]i), because of a reversed transmembrane chloride gradient. Recent experimental data from several rodent studies have demonstrated that exposure to isoflurane during an initial phase causes neuronal excitotoxicity and apoptosis. GABAA receptor-mediated synaptic voltage-dependent calcium channels' (VDCCs) overactivation and Ca2+ influx are involved in these neural changes. METHODS We monitored [Ca2+]i using Fluo-4 AM fluorescence imaging. Using whole-cell patch clamp techniques, IVDCC (voltage-dependent calcium channel currents) were recorded from primary cultures of rat hippocampal neurons (5-day culture) exposed to isoflurane. To further investigate the neurotoxicity of high cytosolic-free calcium after isoflurane in a dose- and time-dependent manner, the possibility of increased caspase-3 levels was evaluated by Western blot and quantitative real-time polymerase chain reaction. Statistical significance was assessed using the Student t test or 1-way analysis of variance followed by the Tukey post hoc test. RESULTS Under control conditions, isoflurane enhanced the GABA-induced [Ca2+]i increase in a dose-dependent manner. Dantrolene and nicardipine markedly inhibited this enhancement mediated by isoflurane. Moreover, in Ca2+-free media, pretreatment with isoflurane did not show any influence on the caffeine-induced increase of [Ca2+]i. Similarly, using whole-cell recording, isoflurane increased the peak amplitude of IVDCC in the cultured neurons from rat hippocampus by depolarization pulses. Isoflurane (0.25, 0.5, 0.75, and 1 minimum alveolar concentration [MAC]) potentiated IVDCC peak current amplitude by 109.11%±9.03%, 120.56%±11.46%, 141.33%±13.87%, and 146.78%±15.87%, respectively. To analyze variation in protein levels, the effect of treatments with isoflurane on caspase-3 activity was dose- and time-dependent, reaching a maximal caspase-3 activity after exposure to 1 MAC for 6 hours (P<0.001). However, in the mRNA levels, hippocampal caspase-3 mRNA levels began to be significantly increased in isoflurane-treated developing rat hippocampal neurons after 6 hours of exposure to 0.25 MAC isoflurane (P<0.001). CONCLUSIONS Isoflurane-mediated enhancement of GABA-triggered [Ca2+]i release results from membrane depolarization with subsequent activation of VDCCs and further Ca2+-induced Ca2+ release from the ryanodine-sensitizing Ca2+ store. An increase in [Ca2+]i, caused by activation of the GABAA receptor and opening of VDCCs, is necessary for isoflurane-induced calcium overload of immature rat hippocampal neurons, which may be involved in the mechanism of an isoflurane-induced neurotoxic effect in the developing rodent brain.
Collapse
Affiliation(s)
- Y L Zhao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Rd., Wuhan, Hubei, 430030, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Petrenko AB, Yamakura T, Kohno T, Sakimura K, Baba H. Reduced immobilizing properties of isoflurane and nitrous oxide in mutant mice lacking the N-methyl-D-aspartate receptor GluR(epsilon)1 subunit are caused by the secondary effects of gene knockout. Anesth Analg 2010; 110:461-5. [PMID: 19933527 DOI: 10.1213/ane.0b013e3181c76e73] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Until recently, the N-methyl-D-aspartate (NMDA) receptor was considered to possibly mediate the immobility produced by inhaled anesthetics such as isoflurane and nitrous oxide. However, new evidence suggests that the role of this receptor in abolition of the movement response may be less important than previously thought. To provide further evidence supporting or challenging this view, we examined the anesthetic potencies of isoflurane and nitrous oxide in genetically modified animals with established NMDA receptor dysfunction caused by GluRepsilon1 subunit knockout. METHODS The immobilizing properties of inhaled anesthetics in mice quantitated by the minimum alveolar anesthetic concentration (MAC) were evaluated using the classic tail clamp method. RESULTS Compared with wild-type controls, NMDA receptor GluRepsilon1 subunit knockout mice displayed larger isoflurane MAC values indicating a resistance to the immobilizing action of isoflurane. Knockout mice were previously shown to have enhanced monoaminergic tone as a result of genetic manipulation, and this increase in MAC could be abolished in our experiments by pretreatment with the serotonin 5-hydroxytryptamine type 2A receptor antagonist ketanserin or with the dopamine D2 receptor antagonist droperidol at doses that did not affect MAC values in wild-type animals. Mutant mice also displayed resistance to the isoflurane MAC-sparing effect of nitrous oxide, but this resistance was similarly abolished by ketanserin and droperidol. Thus, resistance to the immobilizing action of inhaled anesthetics in knockout mice seems to be secondary to increased monoaminergic activation after knockout rather than a direct result of impaired NMDA receptor function. CONCLUSIONS Our results confirm recent findings indicating no critical contribution of NMDA receptors to the immobility induced by isoflurane and nitrous oxide. In addition, they demonstrate the ability of changes secondary to genetic manipulation to affect the results obtained in global knockout studies.
Collapse
Affiliation(s)
- Andrey B Petrenko
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi, Niigata 951-8510, Japan.
| | | | | | | | | |
Collapse
|
18
|
Abstract
BACKGROUND Sevoflurane is often presented as a near-perfect anaesthetic. After 10 years in the operating room, new uses are emerging outside. OBJECTIVE To remind readers of the principal characteristics of sevoflurane, to affirm its usefulness for day-case anaesthesia and to consider the recent new uses. METHODS The discussion of the physical properties, pharmacokinetics, metabolism, mechanisms of action and clinical effects is based on classic, essential papers. Recent literature concerning emerging utilizations of sevoflurane was analysed. RESULTS Sevoflurane presents many benefits with minimum inconvenience. It allows rapid inhalation induction, maintenance and rapid recovery. It has little toxicity and its haemodynamic and respiratory depressive effects are moderate and well tolerated. It is already widely use for sedation for magnetic resonance imaging in children. Its use in paediatric or adult intensive care could improve the management of pain and sedation.
Collapse
Affiliation(s)
- Fabrice Michel
- Faculty of Medicine, Paediatric and Neonatal Intensive Care Unit, University of the Mediterranean, North University Hospital, Marseille Cedex 20, France.
| | | |
Collapse
|
19
|
Isoflurane suppresses stress-enhanced fear learning in a rodent model of post-traumatic stress disorder. Anesthesiology 2009; 110:487-95. [PMID: 19212264 DOI: 10.1097/aln.0b013e3181974f3e] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND A minority of patients who experience awareness and/or pain during surgery subsequently develop post-traumatic stress disorder. In a rodent model of post-traumatic stress disorder, stress-enhanced fear learning (SEFL), rats are preexposed to a stressor of 15 foot shocks. Subsequent exposure to a single foot shock produces an enhanced fear response. This effect is akin to sensitized reactions shown by some post-traumatic stress disorder patients to cues previously associated with the traumatic event. METHODS The authors studied the effect of isoflurane and nitrous oxide on SEFL. Rats were exposed to the inhaled anesthetic during or after a 15-foot shock stressor. Then, rats were given a single foot shock in a different environment. Their fear response was quantified in response to the 15-foot shock and single-foot shock environments. SEFL longevity was tested by placing a 90-day period between the 15 foot shocks and the single foot shock. In addition, the intensity of the foot shock was increased to evaluate treatment effectiveness. RESULTS Increasing isoflurane concentrations decreased SEFL when given during, but not after, the stressor. At 0.40 minimum alveolar concentration (MAC), isoflurane given during the stressor blocked SEFL 90 days later. A threefold increase in the stressor intensity increased the isoflurane concentration required to block SEFL to no more than 0.67 MAC. As with isoflurane, nitrous oxide suppressed SEFL at a similar MAC fraction. CONCLUSIONS These results suggest that sufficient concentrations (perhaps 0.67 MAC or less) of an inhaled anesthetic may prevent SEFL.
Collapse
|
20
|
Sewell JC, Raines DE, Eger EI, Laster MJ, Sear JW. A comparison of the molecular bases for N-methyl-D-aspartate-receptor inhibition versus immobilizing activities of volatile aromatic anesthetics. Anesth Analg 2009; 108:168-75. [PMID: 19095845 DOI: 10.1213/ane.0b013e31818de158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Aromatic anesthetics exhibit a wide range of N-methyl-d-aspartate (NMDA) receptor inhibitory potencies and immobilizing activities. We sought to characterize the molecular basis of NMDA receptor inhibition using comparative molecular field analysis (CoMFA), and compare the results to those from an equivalent model for immobilizing activity. METHODS Published potency data for 14 compounds were supplemented with new values for 2 additional agents. The anesthetics were divided into a training set (n = 12) used to formulate the activity models and a test set (n = 4) used to independently assess the models' predictive capability. The anesthetic structures were geometry optimized using ab initio quantum mechanics and aligned by field-fit minimization to provide the best correlation between the steric and electrostatic fields of the molecules and one or more lead structures. Orientations that yielded CoMFA models with the greatest predictive capability (assessed by leave-one-out cross-validation) were retained. RESULTS The final CoMFA model for the inhibition of NR1/NR2B NMDA receptors explained 99.3% of the variance in the observed activities of the 12 training set agents (F(2,)(9) = 661.5, P < 0.0001). The model effectively predicted inhibitory potency for the training set (cross-validated r(2)(CV) = 0.944) and 4 excluded test set compounds (predictive r(2)(Pred) = 0.966). The equivalent model for immobility in response to noxious stimuli explained 98.0% of the variance in the observed activities for the training set (F(2,)(9) = 219.2, P < 0.0001) and exhibited adequate predictive capability for both the training set (r(2)(CV) = 0.872) and test set (r(2)(Pred) = 0.926) agents. Comparison of pharmacophoric maps showed that several key steric and electrostatic regions were common to both activity models, but differences were observed in the relative importance of these key regions with respect to the two aspects of anesthetic activity. CONCLUSIONS The similarities in the pharmacophoric maps are consistent with NMDA receptors contributing part of the immobilizing activity of volatile aromatic anesthetics.
Collapse
Affiliation(s)
- Jason C Sewell
- Nuffield Department of Anesthetics, University of Oxford, The John Radcliffe Hospital, Headington, Oxford, UK
| | | | | | | | | |
Collapse
|
21
|
Eger EI, Raines DE, Shafer SL, Hemmings HC, Sonner JM. Is a new paradigm needed to explain how inhaled anesthetics produce immobility? Anesth Analg 2008; 107:832-48. [PMID: 18713892 DOI: 10.1213/ane.0b013e318182aedb] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A paradox arises from present information concerning the mechanism(s) by which inhaled anesthetics produce immobility in the face of noxious stimulation. Several findings, such as additivity, suggest a common site at which inhaled anesthetics act to produce immobility. However, two decades of focused investigation have not identified a ligand- or voltage-gated channel that alone is sufficient to mediate immobility. Indeed, most putative targets provide minimal or no mediation. For example, opioid, 5-HT3, gamma-aminobutyric acid type A and glutamate receptors, and potassium and calcium channels appear to be irrelevant or play only minor roles. Furthermore, no combination of actions on ligand- or voltage-gated channels seems sufficient. A few plausible targets (e.g., sodium channels) merit further study, but there remains the possibility that immobilization results from a nonspecific mechanism.
Collapse
Affiliation(s)
- Edmond I Eger
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California 94143-0464, USA.
| | | | | | | | | |
Collapse
|
22
|
Eger EI, Tang M, Liao M, Laster MJ, Solt K, Flood P, Jenkins A, Raines D, Hendrickx JF, Shafer SL, Yasumasa T, Sonner JM. Inhaled anesthetics do not combine to produce synergistic effects regarding minimum alveolar anesthetic concentration in rats. Anesth Analg 2008; 107:479-85. [PMID: 18633026 DOI: 10.1213/01.ane.0000295805.70887.65] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND We hypothesized that pairs of inhaled anesthetics having divergent potencies [one acting weakly at minimum alveolar anesthetic concentration (MAC); one acting strongly at MAC] on specific receptors/channels might act synergistically, and that such deviations from additivity would support the notion that anesthetics act on multiple sites to produce anesthesia. METHODS Accordingly, we studied the additivity of MAC for 11 anesthetic pairs divergently (one weakly, one strongly) affecting a specific receptor/channel at MAC. By "divergently," we usually meant that at MAC the more strongly acting anesthetic enhanced or blocked the in vitro receptor or channel at least twice (and usually more) as much as did the weakly acting anesthetic. The receptors/channels included: TREK-1 and TASK-3 potassium channels; and gamma-aminobutyric acid type A, glycine, N-methyl-D-aspartic acid, and acetylcholine receptors. We also studied the additivity of cyclopropane-benzene because the N-methyl-D-aspartic acid blocker MK-801 had divergent effects on the MACs of these anesthetics. We also studied four pairs that included nitrous oxide because nitrous oxide had been reported to produce infraadditivity (antagonism) when combined with isoflurane. RESULTS All combinations produced a result within 10% of that which would be predicted by additivity except for the combination of isoflurane with nitrous oxide where infraadditivity was found. CONCLUSIONS Such results are consistent with the notion that inhaled anesthetics act on a single site to produce immobility in the face of noxious stimulation.
Collapse
Affiliation(s)
- Edmond I Eger
- Department of Anesthesia, S-455, University of California, San Francisco, CA 94143-0464, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yao A, Kim J, Atherley R, Jinks SL, Carstens E, Shargh S, Sulger A, Antognini JF. The effects of aromatic anesthetics on dorsal horn neuronal responses to noxious stimulation. Anesth Analg 2008; 106:1759-64. [PMID: 18499606 DOI: 10.1213/ane.0b013e3181732ee3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Gamma-aminobutyric acid type A receptor potentiation and/or N-methyl-d-aspartate (NMDA) receptor inhibition might explain the anesthetic properties of fluorinated aromatic compounds. We hypothesized that depression of dorsal horn neuronal responses to noxious stimulation would correlate with the magnitude of effect of benzene (BNZ), o-difluorobenzene, and hexafluorobenzene (HFB) on NMDA receptors. METHODS Rats were anesthetized with desflurane. A T13-L1 laminectomy allowed extracellular recording of neuronal activity from the lumbar spinal cord. After discontinuing desflurane administration, MAC for each aromatic anesthetic was determined. A 5-s noxious mechanical stimulus was then applied to the hindpaw receptive field of nociceptive dorsal horn neurons, and single-neuron responses were recorded at 0.8 and 1.2 MAC. These responses were also recorded in decerebrate rats receiving BNZ and HFB at 0-1.2 MAC. RESULTS In intact rats, depression of responses of dorsal horn neurons to noxious stimulation by peri-MAC increases in BZN, o-difluorobenzene, and HFB correlated directly with their in vitro capacity to block NMDA receptors. In decerebrate rats, 1.2 MAC BNZ depressed nociceptive responses by 60%, with a further percentage decrease continuing from 0.8 to 1.2 MAC approximately equal to that found in intact rats. In decerebrate rats, HFB caused a progressive dose-related decrease in MAC (maximum 25%), but in intact rats, an increase from 0.8 to 1.2 neuronal response caused an (insignificant) increase in neuronal response. CONCLUSIONS The findings in intact rats suggest that NMDA blockade contributes to the depression of dorsal horn neurons to nociceptive stimulation by fluorinated aromatic anesthetics. These results, combined with the additional findings in decerebrate rats, suggest that supraspinal effects (perhaps on gamma-aminobutyric acid type A receptors) may have a supraspinal facilitatory effect on nociception for HFB.
Collapse
Affiliation(s)
- Aubrey Yao
- Department of Anesthesiology and Pain Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Isoflurane-induced caspase-3 activation is dependent on cytosolic calcium and can be attenuated by memantine. J Neurosci 2008; 28:4551-60. [PMID: 18434534 DOI: 10.1523/jneurosci.5694-07.2008] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Increasing evidence indicates that caspase activation and apoptosis are associated with a variety of neurodegenerative disorders, including Alzheimer's disease. We reported that anesthetic isoflurane can induce apoptosis, alter processing of the amyloid precursor protein (APP), and increase amyloid-beta protein (Abeta) generation. However, the mechanism by which isoflurane induces apoptosis is primarily unknown. We therefore set out to assess effects of extracellular calcium concentration on isoflurane-induced caspase-3 activation in H4 human neuroglioma cells stably transfected to express human full-length APP (H4-APP cells). In addition, we tested effects of RNA interference (RNAi) silencing of IP(3) receptor, NMDA receptor, and endoplasmic reticulum (ER) calcium pump, sacro-/ER calcium ATPase (SERCA1). Finally, we examined the effects of the NMDA receptor partial antagonist, memantine, in H4-APP cells and brain tissue of naive mice. EDTA (10 mM), BAPTA (10 microM), and RNAi silencing of IP(3) receptor, NMDA receptor, or SERCA1 attenuated caspase-3 activation. Memantine (4 microM) inhibited isoflurane-induced elevations in cytosolic calcium levels and attenuated isoflurane-induced caspase-3 activation, apoptosis, and cell viability. Memantine (20 mg/kg, i.p.) reduced isoflurane-induced caspase-3 activation in brain tissue of naive mice. These results suggest that disruption of calcium homeostasis underlies isoflurane-induced caspase activation and apoptosis. We also show for the first time that the NMDA receptor partial antagonist, memantine, can prevent isoflurane-induced caspase-3 activation and apoptosis in vivo and in vitro. These findings, indicating that isoflurane-induced caspase activation and apoptosis are dependent on cytosolic calcium levels, should facilitate the provision of safer anesthesia care, especially for Alzheimer's disease and elderly patients.
Collapse
|
25
|
Irifune M, Katayama S, Takarada T, Shimizu Y, Endo C, Takata T, Morita K, Dohi T, Sato T, Kawahara M. MK-801 enhances gabaculine-induced loss of the righting reflex in mice, but not immobility. Can J Anaesth 2008; 54:998-1005. [PMID: 18056209 DOI: 10.1007/bf03016634] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE gamma-Aminobutyric acid (GABA) and N-methyl-D-aspartate (NMDA) receptors are important targets for anesthetic action at the in vitro cellular level. Gabaculine is a GABA-trans-aminase inhibitor that increases endogenous GABA in the brain, and enhances GABA activity. We have recently shown that unconsciousness is associated with the enhanced GABA activity due to gabaculine, but that immobility is not. MK-801 is a selective NMDA channel blocker. In this study, we examined behaviourally whether gabaculine in combination with MK-801 could produce these components of the general anesthetic state. We further compared the effect of MK-801 with ketamine, another NMDA channel blocker. METHODS All drugs were administered intraperitoneally to adult male ddY mice. To assess the general anesthetic components, two endpoints were used. One was loss of the righting reflex (LORR; as a measure of unconsciousness) and the other was loss of movement in response to tail-clamp stimulation (as a measure of immobility). RESULTS Large doses of MK-801 alone (10-50 mg.kg(-1)) induced neither LORR nor immobility in response to noxious stimulation. However, even a small dose (0.2 mgxkg(-1)) significantly enhanced gabaculine-induced LORR (P < 0.05), although gabaculine in combination with MK-801 (0.2-10 mgxkg(-1)) produced no immobility. However, gabaculine plus a subanesthetic dose of ketamine (30 mgxkg(-1)), which acts on NMDA, opioid and nicotinic acetylcholine receptors and neuronal Na(+) channels, suppressed the pain response, but did not achieve a full effect. Ketamine alone dose-dependently produced both LORR and immobility. CONCLUSION These findings suggest that gabaculine-induced LORR is modulated by blocking NMDA receptors, but that immobility is not mediated through GABA or NMDA receptors.
Collapse
Affiliation(s)
- Masahiro Irifune
- Department of Dental Anesthesiology, Division of Clinical Medical Science, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734- 8553, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kim J, Yao A, Atherley R, Carstens E, Jinks SL, Antognini JF. Neurons in the ventral spinal cord are more depressed by isoflurane, halothane, and propofol than are neurons in the dorsal spinal cord. Anesth Analg 2007; 105:1020-6, table of contents. [PMID: 17898382 PMCID: PMC2693417 DOI: 10.1213/01.ane.0000280483.17854.56] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Volatile anesthetics act primarily in the spinal cord to produce immobility but their exact site of action is unclear. Between 0.8 and 1.2 minimum alveolar anesthetic concentration (MAC), isoflurane does not depress neurons in the dorsal horn, suggesting that it acts at a more ventral site within the spinal cord such as in premotor interneurons and motoneurons. We hypothesized that isoflurane, halothane, and propofol would exert a greater depressant effect on nociceptive responses of ventral horn neurons when compared with dorsal horn neurons. METHODS Rats were anesthetized with isoflurane or halothane and responses of dorsal (<1200 microm deep) and ventral (>1200 microm) lumbar neurons to noxious mechanical stimulation of the hindpaw were determined at 0.8 and 1.2 MAC. In a third group anesthetized with isoflurane at 0.8 MAC, we administered 5 mg/kg propofol while recording responses from dorsal horn or ventral horn neurons. RESULTS Dorsal horn neuronal responses were not significantly affected when either isoflurane or halothane was increased from 0.8 to 1.2 MAC; propofol also had no significant effect. On the other hand, with increased isoflurane or halothane concentration, responses of ventral horn neurons were depressed by 60% and 45%, respectively. Propofol profoundly depressed (>90%) ventral horn neurons. CONCLUSIONS These data suggest that, in the peri-MAC range, isoflurane, halothane, and propofol have little or no effect on neuronal responses to noxious mechanical stimulation in the spinal dorsal horn but depress such responses in the ventral horn. Immobility produced in the 0.8-1.2 MAC range by these anesthetics appears to result from a depressant action in the ventral horn.
Collapse
Affiliation(s)
- JongBun Kim
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California
- Department of Anesthesia and Pain Medicine, Catholic University of Korea, Seoul, Korea
| | - Aubrey Yao
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California
| | - Richard Atherley
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California
| | - Earl Carstens
- Section of Neurobiology, Physiology and Behavior, University of California, Davis, California
| | - Steven L. Jinks
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California
| | - Joseph F. Antognini
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California
- Section of Neurobiology, Physiology and Behavior, University of California, Davis, California
| |
Collapse
|
27
|
Irifune M, Kikuchi N, Saida T, Takarada T, Shimizu Y, Endo C, Morita K, Dohi T, Sato T, Kawahara M. Riluzole, a Glutamate Release Inhibitor, Induces Loss of Righting Reflex, Antinociception, and Immobility in Response to Noxious Stimulation in Mice. Anesth Analg 2007; 104:1415-21, table of contents. [PMID: 17513634 DOI: 10.1213/01.ane.0000263267.04198.36] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The general anesthetic state comprises behavioral and perceptual components, including amnesia, unconsciousness, analgesia, and immobility. In vitro, glutamatergic excitatory neurons are important targets for anesthetic action at the cellular and microcircuits levels. Riluzole (2-amino-6-[trifluoromethoxy]benzothiazole) is a neuroprotective drug that inhibits glutamate release from nerve terminals in the central nervous system. Here, we examined in vivo the ability of riluzole to produce components of the general anesthetic state through a selective blockade of glutamatergic neurotransmission. METHODS Riluzole was administered intraperitoneally in adult male ddY mice. To assess the general anesthetic components, three end-points were used: 1) loss of righting reflex (LORR; as a measure of unconsciousness), 2) loss of movement in response to noxious stimulation (as a measure of immobility), and 3) loss of nociceptive response (as a measure of analgesia). RESULTS The intraperitoneal administration of riluzole induced LORR in a dose-dependent fashion with a 50% effective dose value of 27.4 (23.3-32.2; 95% confidence limits) mg/kg. The behavioral and microdialysis studies revealed that time-course changes in impairment and LORR induced by riluzole corresponded with decreased glutamate levels in the mouse brain. This suggests that riluzole-induced LORR (unconsciousness) could result, at least in part, from its ability to decrease brain glutamate concentrations. Riluzole dose-dependently produced not only LORR, but also loss of movement in response to painful stimulation (immobility), and loss of nociceptive response (analgesia) with 50% effective dose values of 43.0 (37.1-49.9), and 10.0 (7.4-13.5) mg/kg, respectively. These three dose-response curves were parallel, suggesting that the behavioral effects of riluzole may be mediated through a common site of action. CONCLUSIONS These findings suggest that riluzole-induced LORR, immobility, and antinociception appear to be associated with its ability to inhibit glutamatergic neurotransmission in the central nervous system.
Collapse
Affiliation(s)
- Masahiro Irifune
- Department of Dental Anesthesiology, Division of Clinical Medical Science, Programs for Applied Biomedicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
BACKGROUND Previous studies have found that the local anesthetic/sodium channel blocker lidocaine decreased MAC by maximum amounts approximately equal to the decreases produced by dizocilpine (MK-801), a N-methyl-d-aspartate (NMDA) receptor antagonist. Blockade of sodium channels by inhaled anesthetics has been suggested as a possible cause for impairment of transmission through NMDA receptors. We postulated that the net effect of lidocaine and MK-801 on MAC would be the same, albeit by affecting NMDA neurotransmission at different points. METHODS We measured the effect of various lidocaine infusions on the MAC of cyclopropane, halothane, isoflurane, and o-difluorobenzene in rats. We also measured the effect of concurrent lidocaine-MK-801 infusion on the MAC of isoflurane and o-difluorobenzene. RESULTS Our data contradicted our predictions. (a) We found no limit to the effect of lidocaine infusion, in some cases finding that lidocaine, alone, produced immobility; (b) lidocaine infusion did not decrease the MAC of o-difluorobenzene differently from the MAC of other inhaled anesthetics; and (c) the addition of MK-801 equally affected the decrease in MAC produced by lidocaine infusion for isoflurane versus o-difluorobenzene. CONCLUSION Lidocaine does not primarily decrease MAC by decreasing the release of glutamate from nerve terminals.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California 94143-0464, USA
| | | | | | | | | |
Collapse
|
29
|
Kim J, Atherley R, Werner DF, Homanics GE, Carstens E, Antognini JF. Isoflurane depression of spinal nociceptive processing and minimum alveolar anesthetic concentration are not attenuated in mice expressing isoflurane resistant gamma-aminobutyric acid type-A receptors. Neurosci Lett 2007; 420:209-12. [PMID: 17543455 PMCID: PMC3045261 DOI: 10.1016/j.neulet.2007.04.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Revised: 04/24/2007] [Accepted: 04/24/2007] [Indexed: 01/04/2023]
Abstract
Anesthetics produce immobility and depress spinal nociceptive processing, but the exact sites and mechanisms of anesthetic action are unknown. The gamma-aminobutyric acid type-A (GABAA) receptor is thought to be important to anesthetic action. We studied knock-in mice that had mutations in the alpha1 subunit of the GABAA receptor that imparts resistance to isoflurane in in vitro systems. We determined the isoflurane minimum alveolar concentration (MAC) that produces immobility in 50% of subjects and responses of lumbar neurons (single-unit recordings) to noxious stimulation (5 s pinch) of the hindpaw. Isoflurane MAC did not differ between wild-type (1.1+/-0.1%) and knock-in (1.1+/-0.1%) mice. Isoflurane depressed neuronal responses to noxious stimulation (60 s period during and after pinch) similarly in both wild-type and knock-in mice (555+/-133 and 636+/-106 impulses/min, respectively, at 0.8 MAC and 374+/-81 and 409+/-85 impulses/min at 1.2 MAC). We conclude that isoflurane enhancement of alpha1-containing GABAA receptors is not required to produce immobility or depress spinal nociceptive processing.
Collapse
Affiliation(s)
- JongBun Kim
- Department of Anesthesiology, The Catholic University of Korea, Seoul, Korea
| | - Richard Atherley
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, California 95616 United States
| | - David F. Werner
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Gregg E. Homanics
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Earl Carstens
- Section of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, California 95616 United States
| | - Joseph F. Antognini
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, California 95616 United States
- Section of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, California 95616 United States
- Correspondence to: Joseph F. Antognini, M.D., Department of Anesthesiology and Pain Medicine, University of California, Davis, TB-170, Davis, California 95616, FAX 530-752-7807,
| |
Collapse
|
30
|
Antognini JF, Raines DE, Solt K, Barter LS, Atherley RJ, Bravo E, Laster MJ, Jankowska K, Eger EI. Hexafluorobenzene acts in the spinal cord, whereas o-difluorobenzene acts in both brain and spinal cord, to produce immobility. Anesth Analg 2007; 104:822-8. [PMID: 17377088 DOI: 10.1213/01.ane.0000255226.63909.32] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Previous work demonstrated that isoflurane and halothane act on the spinal cord rather than on the brain to produce immobility in the face of noxious stimulation. These anesthetics share many effects on specific receptors, and thus do not test the broad applicability of the mediation of immobility by the cord. We sought to test such an applicability by determining whether the cord mediated the immobilizing effects of two aromatic anesthetics that differ greatly in their ability to block N-methyl-d-aspartate receptors. METHODS We investigated the actions of hexafluorobenzene (HFB) and o-difluorobenzene (ODFB) using an intact goat model that allowed selective delivery of anesthetics to the brain. Because our results suggested a significant cerebral effect of ODFB, in other goats we administered halothane 0.5% to the brain, while determining the ODFB concentration delivered to the body (the cord) required for immobility. We chose halothane because the present and previous studies found that cerebral halothane concentrations alone required for producing immobility far exceeded those required in the cord. We also applied the above techniques to another benzene-containing anesthetic, propofol. RESULTS Prebypass minimum alveolar concentration (MAC) for HFB was 0.82% +/- 0.14% (mean +/- sd); increased to 2.04% +/- 0.8% (P < 0.01) during selective delivery to the cranial circulation; and returned to 0.79% +/- 0.28% postbypass. Corresponding values for ODFB were 0.46% +/- 0.07%, 0.63% +/- 0.12% (P < 0.05), and 0.44% +/- 0.10%. ODFB MAC was 0.32% +/- 0.17% during selective halothane delivery to brain. But when ODFB was administered to the whole body, MAC was 0.37% +/- 0.05%, (NS). Like HFB, the halothane requirement increased threefold when delivered only to the head. In four of five animals, propofol requirements increased by 240%, but in one animal propofol requirements decreased, and the overall change was not statistically significant. CONCLUSIONS These data suggest that HFB, like halothane, produces immobility, predominantly by a spinal cord action, and that HFB differs from ODFB with respect to brain versus spinal sites of action. Nonetheless, although ODFB can produce immobility via a cerebral action, it also can do this via an independent action in the spinal cord. Thus, our results continue to support the spinal cord as the primary site at which inhaled anesthetics, and perhaps propofol, produce immobility.
Collapse
Affiliation(s)
- Joseph F Antognini
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California 95616, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Antognini JF, Atherley RJ, Dutton RC, Laster MJ, Eger EI, Carstens E. The excitatory and inhibitory effects of nitrous oxide on spinal neuronal responses to noxious stimulation. Anesth Analg 2007; 104:829-35. [PMID: 17377089 DOI: 10.1213/01.ane.0000255696.11833.24] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Because of the logistical obstacles to measurement under hyperbaric conditions, the effect of nitrous oxide (N2O) alone on spinal neuronal responses has not been tested. We hypothesized that, like other inhaled anesthetics, N2O would depress spinal neuronal responses to noxious stimulation. METHODS The lumbar spinal cord was exposed in rats anesthetized with isoflurane. Mechanically ventilated rats were placed into a hyperbaric chamber and needle electrodes were inserted into the hindpaws. Isoflurane administration was discontinued and anesthesia converted to N2O by pressurizing the chamber with N2O. A microelectrode was inserted into the lumbar cord using computer-controlled motors and a hydraulic microdrive. Neuronal responses to electrical stimulation of the hindpaw were sought at 1.5, 2, and 2.5 atm N2O (0.8-1.3 minimum alveolar concentration). RESULTS Increasing N2O partial pressures variably affected neuronal responses to a 2 s 100-Hz electrical stimulus. Neuronal depth and neuronal response were correlated, with superficial neurons tending to be facilitated, while deeper neurons were depressed; (overall responses were 1331 +/- 408, 1594 +/- 383, and 1578 +/- 500 impulses/min at 1.5, 2, and 2.5 atm N2O, respectively; mean, standard error). N2O did not affect neuronal responses to a repetitive "windup" stimulus. Infusion of the N-methyl-d-aspartate blocker MK-801 into separate rats increased the neuronal response to the 100-Hz stimulus (from 781 +/- 216 to 1352 +/- 269 impulses/min, P < 0.05). CONCLUSIONS N2O facilitated superficial spinal neuronal responses to noxious stimulation while depressing deeper neurons. These results suggest that anesthetic partial pressures of N2O have divergent effects on spinal neuronal responses to noxious stimulation, the specific responses depending on the depth of the spinal neurons.
Collapse
Affiliation(s)
- Joseph F Antognini
- Department of Anesthesiology and Pain Medicine, University of California, Davis, California 95616, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Eger EI. Does the N-Methyl-d-Aspartate Receptor Contribute to the Effects of Isoflurane, Sevoflurane, and Ethanol? Anesth Analg 2007. [DOI: 10.1213/01.ane.0000251919.66115.3d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
33
|
Sato Y, Seo N, Kobayashi E. Does the N-Methyl-d-Aspartate Receptor Contribute to the Effects of Isoflurane, Sevoflurane, and Ethanol? Anesth Analg 2007; 104:219-20; author reply 220. [PMID: 17179285 DOI: 10.1213/01.ane.0000251916.92662.57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Ng KP, Antognini JF. Isoflurane and Propofol Have Similar Effects on Spinal Neuronal Windup at Concentrations that Block Movement. Anesth Analg 2006; 103:1453-8. [PMID: 17122222 DOI: 10.1213/01.ane.0000247732.33602.f5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND We investigated the actions of isoflurane and propofol on neuronal windup in the spinal cord of intact rats. We hypothesized that propofol would depress windup more than isoflurane. METHODS In a cross-over design, rats received 0.8 and 1.2 minimum alveolar concentration (MAC) isoflurane and 0.8 and 1.2 ED50 (effective dose(50%)) of propofol, as recordings were made from single units in the lumbar cord (n = 13). Electrical stimuli were applied (20 stimuli at 0.1, 1, and 3 Hz). Neuronal responses were analyzed for those occurring in the C-fiber range (100-400 ms after each stimulus), combined C-fiber and afterdischarge range (100-1000 ms) and the 100-333 ms range for the 3 Hz stimuli. Absolute windup was also calculated (the sum of action potentials for 20 stimuli - 20 x response to the first stimulus). RESULTS At 1 Hz, total action potentials (mean, standard error) summed across the 20 stimuli (100-1000 ms range) were 571 +/- 106 and 742 +/- 214 for isoflurane (at 0.8 and 1.2 MAC) and 586 +/- 148 and 641 +/- 143 for propofol (at 0.8 and 1.2 ED50), respectively (P = NS); corresponding values for the 0.1 Hz stimuli were 345 +/- 104, 370 +/- 108, 430 +/- 86, and 403 +/- 106 (P = NS), and for the 3 Hz stimuli (100-333 ms range) were 266 +/- 66, 333 +/- 76, 343 +/- 85, and 252 +/- 72 (P = NS). Absolute windup in the 100-1000 ms range was greater for 1.2 MAC isoflurane at 1 Hz (445 +/- 82, P < 0.01), when compared with absolute windup at 0.8 MAC isoflurane and 0.8 and 1.2 ED50 propofol (232 +/- 31, 88 +/- 65, and 210 +/- 41, respectively). CONCLUSIONS These data suggest that isoflurane and propofol have similar effects on neuronal windup in the spinal cord, although there was enhanced absolute windup at 1.2 MAC isoflurane for the 1 Hz stimulus.
Collapse
Affiliation(s)
- Kevin P Ng
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, California 95616, USA
| | | |
Collapse
|
35
|
Abstract
BACKGROUND Depletion of central nervous system catecholamines, including dopamine, can decrease MAC (the minimum alveolar concentration of an inhaled anesthetic required to suppress movement in response to a noxious stimulus in 50% of test subjects); release of central nervous system catecholamines, including dopamine, can increase MAC; and increased free dopamine concentrations in the striatum can decrease MAC. Such findings suggest that dopamine receptors might mediate part of the capacity of inhaled anesthetics to provide immobility in the face of noxious stimulation. METHODS We measured the effect of blockade of D2 dopamine-mediated transmission with 0.3 mg/kg or 3.0 mg/kg droperidol on the MAC of cyclopropane, desflurane, halothane, isoflurane, or sevoflurane in rats, and the effect of 3.0 mg/kg droperidol on the dose or concentration of etomidate (an anesthetic known to act principally by enhancing the response of gamma-aminobutyric acid(A) receptors to gamma-aminobutyric acid) required to suppress movement in response to noxious stimulation. RESULTS Blockade of D2 dopamine-mediated transmission with droperidol does not decrease the MAC of cyclopropane, desflurane, halothane, isoflurane, or sevoflurane or its equivalent for etomidate in rats. CONCLUSIONS These data, plus data from studies by others about D1 dopamine receptors, indicate that dopamine receptors do not mediate the immobility produced by inhaled anesthetics.
Collapse
Affiliation(s)
- Yasumasa Tanifuji
- Department of Anesthesia, University of California, San Francisco, CA 94143-0464, USA
| | | | | | | | | | | |
Collapse
|
36
|
Antognini JF, Atherley RJ, Laster MJ, Carstens E, Dutton RC, Eger EI. A method for recording single unit activity in lumbar spinal cord in rats anesthetized with nitrous oxide in a hyperbaric chamber. J Neurosci Methods 2006; 160:215-22. [PMID: 17045342 PMCID: PMC2693129 DOI: 10.1016/j.jneumeth.2006.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 08/31/2006] [Accepted: 09/05/2006] [Indexed: 11/24/2022]
Abstract
The limited potency of nitrous oxide mandates the use of a hyperbaric chamber to produce anesthesia. Use of a hyperbaric chamber complicates anesthetic delivery, ventilation, and electrophysiological recording. We constructed a hyperbaric acrylic-aluminum chamber allowing recording of single unit activity in spinal cord of rats anesthetized only with N(2)O. Large aluminum plates secured to each other by rods that span the length of the chamber close each end of the chamber. The 122 cm long, 33 cm wide chamber housed ventilator, intravenous infusion pumps, recording headstage, including hydraulic microdrive and stepper motors (controlled by external computers). Electrical pass-throughs in the plates permitted electrical current or signals to enter or leave the chamber. In rats anesthetized only with N(2)O we recorded extracellular action potentials with a high signal-to-noise ratio. We also recorded electroencephalographic activity. This technique is well-suited to study actions of weak anesthetics such as N(2)O and Xe at working pressures of 4-5 atm or greater. The safety of such pressures depends on the wall thickness and chamber diameter.
Collapse
Affiliation(s)
- Joseph F Antognini
- Department of Anesthesiology and Pain Medicine, University of California, Davis, TB-170, Davis, CA 95616, USA.
| | | | | | | | | | | |
Collapse
|