Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Anesthesiology ; 140(3): 463-482, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38118175

ABSTRACT

BACKGROUND: Carriers of mutations in the mitochondrial electron transport chain are at increased risk of anesthetic-induced neurotoxicity. To investigate the neurotoxicity mechanism and to test preconditioning as a protective strategy, this study used a Drosophila melanogaster model of Leigh syndrome. Model flies carried a mutation in ND23 (ND2360114) that encodes a mitochondrial electron transport chain complex I subunit. This study investigated why ND2360114 mutants become susceptible to lethal, oxygen-modulated neurotoxicity within 24 h of exposure to isoflurane but not sevoflurane. METHODS: This study used transcriptomics and quantitative real-time reverse transcription polymerase chain reaction to identify genes that are differentially expressed in ND2360114 but not wild-type fly heads at 30 min after exposure to high- versus low-toxicity conditions. This study also subjected ND2360114 flies to diverse stressors before isoflurane exposure to test whether isoflurane toxicity could be diminished by preconditioning. RESULTS: The ND2360114 mutation had a greater effect on isoflurane- than sevoflurane-mediated changes in gene expression. Isoflurane and sevoflurane did not affect expression of heat shock protein (Hsp) genes (Hsp22, Hsp27, and Hsp68) in wild-type flies, but isoflurane substantially increased expression of these genes in ND2360114 mutant flies. Furthermore, isoflurane and sevoflurane induced expression of oxidative (GstD1 and GstD2) and xenobiotic (Cyp6a8 and Cyp6a14) stress genes to a similar extent in wild-type flies, but the effect of isoflurane was largely reduced in ND2360114 flies. In addition, activating stress response pathways by pre-exposure to anesthetics, heat shock, hyperoxia, hypoxia, or oxidative stress did not suppress isoflurane-induced toxicity in ND2360114 mutant flies. CONCLUSIONS: Mutation of a mitochondrial electron transport chain complex I subunit generates differential effects of isoflurane and sevoflurane on gene expression that may underlie their differential effects on neurotoxicity. Additionally, the mutation produces resistance to preconditioning by stresses that protect the brain in other contexts. Therefore, complex I activity modifies molecular and physiologic effects of anesthetics in an anesthetic-specific manner.


Subject(s)
Anesthetics, Inhalation , Isoflurane , Methyl Ethers , Animals , Isoflurane/toxicity , Sevoflurane/pharmacology , Anesthetics, Inhalation/toxicity , Drosophila melanogaster/genetics , Oxidative Stress , Electron Transport Complex I/genetics , Methyl Ethers/pharmacology
2.
Anesthesiology ; 138(6): 585-586, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37158651

Subject(s)
Anesthesia , Anesthesiology
3.
Exp Biol Med (Maywood) ; 248(7): 545-552, 2023 04.
Article in English | MEDLINE | ID: mdl-37208922

ABSTRACT

One of the unsolved mysteries of medicine is how do volatile anesthetics (VAs) cause a patient to reversibly lose consciousness. In addition, identifying mechanisms for the collateral effects of VAs, including anesthetic-induced neurotoxicity (AiN) and anesthetic preconditioning (AP), has proven challenging. Multiple classes of molecules (lipids, proteins, and water) have been considered as potential VA targets, but recently proteins have received the most attention. Studies targeting neuronal receptors or ion channels had limited success in identifying the critical targets of VAs mediating either the phenotype of "anesthesia" or their collateral effects. Recent studies in both nematodes and fruit flies may provide a paradigm shift by suggesting that mitochondria may harbor the upstream molecular switch activating both primary and collateral effects. The disruption of a specific step of electron transfer within the mitochondrion causes hypersensitivity to VAs, from nematodes to Drosophila and to humans, while also modulating the sensitivity to collateral effects. The downstream effects from mitochondrial inhibition are potentially legion, but inhibition of presynaptic neurotransmitter cycling appears to be specifically sensitive to the mitochondrial effects. These findings are perhaps of even broader interest since two recent reports indicate that mitochondrial damage may well underlie neurotoxic and neuroprotective effects of VAs in the central nervous system (CNS). It is, therefore, important to understand how anesthetics interact with mitochondria to affect CNS function, not just for the desired facets of general anesthesia but also for significant collateral effects, both harmful and beneficial. A tantalizing possibility exists that both the primary (anesthesia) and secondary (AiN, AP) mechanisms may at least partially overlap in the mitochondrial electron transport chain (ETC).


Subject(s)
Anesthetics, Inhalation , Anesthetics , Humans , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/metabolism , Anesthetics/pharmacology , Mitochondria/metabolism , Central Nervous System
4.
J Vis Exp ; (192)2023 02 24.
Article in English | MEDLINE | ID: mdl-36912551

ABSTRACT

Volatile general anesthetics (VGAs) are used worldwide on millions of people of all ages and medical conditions. High concentrations of VGAs (hundreds of micromolar to low millimolar) are necessary to achieve a profound and unphysiological suppression of brain function presenting as "anesthesia" to the observer. The full spectrum of the collateral effects triggered by such high concentrations of lipophilic agents is not known, but interactions with the immune-inflammatory system have been noted, although their biological significance is not understood. To investigate the biological effects of VGAs in animals, we developed a system termed the serial anesthesia array (SAA) to exploit the experimental advantages offered by the fruit fly (Drosophila melanogaster). The SAA consists of eight chambers arranged in series and connected to a common inflow. Some parts are available in the lab, and others can be easily fabricated or purchased. A vaporizer, which is necessary for the calibrated administration of VGAs, is the only commercially manufactured component. VGAs constitute only a small percentage of the atmosphere flowing through the SAA during operation, as the bulk (typically over 95%) is carrier gas; the default carrier is air. However, oxygen and any other gases can be investigated. The SAA's principal advantage over prior systems is that it allows the simultaneous exposure of multiple cohorts of flies to exactly titrable doses of VGAs. Identical concentrations of VGAs are achieved within minutes in all the chambers, thus providing indistinguishable experimental conditions. Each chamber can contain from a single fly to hundreds of flies. For example, the SAA can simultaneously examine eight different genotypes or four genotypes with different biological variables (e.g., male vs. female, old vs. young). We have used the SAA to investigate the pharmacodynamics of VGAs and their pharmacogenetic interactions in two experimental fly models associated with neuroinflammation-mitochondrial mutants and traumatic brain injury (TBI).


Subject(s)
Anesthesia , Brain Injuries, Traumatic , Animals , Male , Female , Drosophila melanogaster , Drosophila
5.
Int J Mol Sci ; 24(3)2023 Jan 17.
Article in English | MEDLINE | ID: mdl-36768163

ABSTRACT

The mitochondrial electron transport chain (mETC) contains molecular targets of volatile general anesthetics (VGAs), which places carriers of mutations at risk for anesthetic complications. The ND-2360114 and mt:ND2del1 lines of fruit flies (Drosophila melanogaster) that carry mutations in core subunits of Complex I of the mETC replicate numerous characteristics of Leigh syndrome (LS) caused by orthologous mutations in mammals and serve as models of LS. ND-2360114 flies are behaviorally hypersensitive to volatile anesthetic ethers and develop an age- and oxygen-dependent anesthetic-induced neurotoxicity (AiN) phenotype after exposure to isoflurane but not to the related anesthetic sevoflurane. The goal of this paper was to investigate whether the alkane volatile anesthetic halothane and other mutations in Complex I and in Complexes II-V of the mETC cause AiN. We found that (i) ND-2360114 and mt:ND2del1 were susceptible to toxicity from halothane; (ii) in wild-type flies, halothane was toxic under anoxic conditions; (iii) alleles of accessory subunits of Complex I predisposed to AiN; and (iv) mutations in Complexes II-V did not result in an AiN phenotype. We conclude that AiN is neither limited to ether anesthetics nor exclusive to mutations in core subunits of Complex I.


Subject(s)
Anesthetics, Inhalation , Anesthetics , Isoflurane , Animals , Drosophila melanogaster/genetics , Halothane/pharmacology , Anesthetics, Inhalation/pharmacology , Ether , Electrons , Isoflurane/pharmacology , Mutation , Drosophila , Ethers , Electron Transport Complex I/genetics , Ethyl Ethers , Mammals
6.
J Pharmacol Exp Ther ; 381(3): 229-235, 2022 06.
Article in English | MEDLINE | ID: mdl-35347062

ABSTRACT

We tested the hypothesis that obesity influences the pharmacodynamics of volatile general anesthetics (VGAs) by comparing effects of anesthetic exposure on mortality from traumatic brain injury (TBI) in lean and obese Drosophila melanogaster We induced TBI with a high-impact trauma device. Starvation-selection over multiple generations resulted in an obese phenotype (SS flies). Fed flies served as lean controls (FC flies). Adult (1-8-day-old) SS and FC flies were exposed to equianesthetic doses of isoflurane or sevoflurane either before or after TBI. The principal outcome was percent mortality 24 hours after injury, expressed as the Mortality Index at 24 hours (MI24). TBI resulted in a lower MI24 in FC than in SS flies [21 (2.35) and 57.8 (2.14), respectively n = 12, P = 0.0001]. Pre-exposure to isoflurane or sevoflurane preconditioned FC flies to TBI, reducing the risk of death to 0.53 (0.25 to 1.13) and 0.82 (0.43 to 1.58), respectively, but had no preconditioning effect in SS flies. Postexposure to isoflurane or sevoflurane increased the risk of death in SS flies, but only postexposure to isoflurane increased the risk in FC flies [1.39 (0.81 to 2.38)]. Thus, obesity affects the pharmacodynamics of VGAs, thwarting the preconditioning effect of isoflurane and sevoflurane in TBI. SIGNIFICANCE STATEMENT: Inadvertent preconditioning in models of traumatic brain injury (TBI) is a recognized confounder. The findings in a fruit fly (Drosophila melanogaster) model of closed-head TBI indicate that anesthetic pharmacodynamics are profoundly affected by obesity. Specifically, obesity thwarts the brain-protective effect of anesthetic preconditioning. This finding is important for experimental studies of TBI and supports the versatility of the fruit fly as a model for the exploration of anesthetic pharmacodynamics in a wide parameter space.


Subject(s)
Anesthetics, Inhalation , Brain Injuries, Traumatic , Isoflurane , Anesthetics, Inhalation/pharmacology , Animals , Drosophila , Drosophila melanogaster , Isoflurane/pharmacology , Obesity , Sevoflurane/pharmacology
8.
Anesthesiology ; 134(6): 835-837, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33909884
9.
Int J Mol Sci ; 21(24)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333797

ABSTRACT

Background: Mice carrying the GABAA receptor ß3(N265M) point mutation, which renders receptors incorporating ß3-subunits insensitive to many general anesthetics, have been used experimentally to link modulation of different receptor subtypes to distinct behavioral endpoints. Remarkably, however, the effect of the mutation on the susceptibility to modulation by isoflurane (a standard reference agent for inhalational vapors) has never been tested directly. Therefore, we compared the modulation by isoflurane of expressed α5ß3(N265M)γ2L receptors with their wild type counterparts. Methods: Using whole-cell electrophysiological recording and rapid solution exchange techniques, we tested the effects of isoflurane at concentrations ranging from 80 µM to 320 µM on currents activated by 1 µM GABA. We measured drug modulation of wild-type α5ß3γ2L GABAA receptors and their counterparts harboring the ß3(N265M) mutation. Results: Currents elicited by GABA were enhanced two- to four-fold by isoflurane, in a concentration-dependent manner. Under the same conditions, receptors incorporating the ß3(N265M) mutation were enhanced by approximately 1.5- to two-fold; i.e., modulation by isoflurane was attenuated by approximately one-half. Direct activation by isoflurane was also present in mutant receptors but also attenuated. Conclusions: In contrast to the complete insensitivity of ß3(N265M) mutant receptors to etomidate and propofol, the mutation has only a partial effect on receptor modulation by isoflurane. Therefore, the persistence of isoflurane effects in mutant mice does not exclude a possible contribution of ß3-GABAA receptors.


Subject(s)
Anesthetics, Inhalation/pharmacology , Isoflurane/pharmacology , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/pharmacology , Animals , Etomidate/pharmacology , HEK293 Cells , Humans , Mice, Transgenic , Patch-Clamp Techniques , Point Mutation , Propofol/pharmacology , Receptors, GABA-A/physiology , Recombinant Proteins
10.
Int J Mol Sci ; 21(18)2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32967238

ABSTRACT

Following traumatic brain injury (TBI), the time window during which secondary injuries develop provides a window for therapeutic interventions. During this time, many TBI victims undergo exposure to hyperoxia and anesthetics. We investigated the effects of genetic background on the interaction of oxygen and volatile general anesthetics with brain pathophysiology after closed-head TBI in the fruit fly Drosophila melanogaster. To test whether sevoflurane shares genetic risk factors for mortality with isoflurane and whether locomotion is affected similarly to mortality, we used a device that generates acceleration-deceleration forces to induce TBI in ten inbred fly lines. After TBI, we exposed flies to hyperoxia alone or in combination with isoflurane or sevoflurane and quantified mortality and locomotion 24 and 48 h after TBI. Modulation of TBI-induced mortality and locomotor impairment by hyperoxia with or without anesthetics varied among fly strains and among combinations of agents. Resistance to increased mortality from hyperoxic isoflurane predicted resistance to increased mortality from hyperoxic sevoflurane but did not predict the degree of locomotion impairment under any condition. These findings are important because they demonstrate that, in the context of TBI, genetic background determines the latent toxic potentials of oxygen and anesthetics.


Subject(s)
Anesthetics, Inhalation/pharmacology , Genetic Background , Head Injuries, Closed , Hyperoxia , Isoflurane/pharmacology , Sevoflurane/pharmacology , Animals , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Drosophila melanogaster , Head Injuries, Closed/drug therapy , Head Injuries, Closed/genetics , Head Injuries, Closed/metabolism , Head Injuries, Closed/pathology , Humans , Hyperoxia/drug therapy , Hyperoxia/genetics , Hyperoxia/metabolism , Hyperoxia/pathology , Oxygen Consumption/drug effects
11.
Anesthesiology ; 133(4): 839-851, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32773682

ABSTRACT

BACKGROUND: General anesthetics influence mitochondrial homeostasis, placing individuals with mitochondrial disorders and possibly carriers of recessive mitochondrial mutations at increased risk of perioperative complications. In Drosophila, mutations in the ND23 subunit of complex I of the mitochondrial electron transport chain-analogous to mammalian NDUFS8-replicate key characteristics of Leigh syndrome, an inherited mitochondrial disorder. The authors used the ND23 mutant for testing the hypothesis that anesthetics have toxic potential in carriers of mitochondrial mutations. METHODS: The authors exposed wild-type flies and ND23 mutant flies to behaviorally equivalent doses of isoflurane or sevoflurane in 5%, 21%, or 75% oxygen. The authors used percent mortality (mean ± SD, n ≥ 3) at 24 h after exposure as a readout of toxicity and changes in gene expression to investigate toxicity mechanisms. RESULTS: Exposure of 10- to 13-day-old male ND23 flies to isoflurane in 5%, 21%, or 75% oxygen resulted in 16.0 ± 14.9% (n = 10), 48.2 ± 16.1% (n = 9), and 99.2 ± 2.0% (n = 10) mortality, respectively. Comparable mortality was observed in females. In contrast, under the same conditions, mortality was less than 5% for all male and female groups exposed to sevoflurane, except 10- to 13-day-old male ND23 flies with 9.6 ± 8.9% (n = 16) mortality. The mortality of 10- to 13-day-old ND23 flies exposed to isoflurane was rescued by neuron- or glia-specific expression of wild-type ND23. Isoflurane and sevoflurane differentially affected expression of antioxidant genes in 10- to 13-day-old ND23 flies. ND23 flies had elevated mortality from paraquat-induced oxidative stress compared with wild-type flies. The mortality of heterozygous ND23 flies exposed to isoflurane in 75% oxygen increased with age, resulting in 54.0 ± 19.6% (n = 4) mortality at 33 to 39 days old, and the percent mortality varied in different genetic backgrounds. CONCLUSIONS: Mutations in the mitochondrial complex I subunit ND23 increase susceptibility to isoflurane-induced toxicity and to oxidative stress in Drosophila. Asymptomatic flies that carry ND23 mutations are sensitized to hyperoxic isoflurane toxicity by age and genetic background.


Subject(s)
Anesthetics, Inhalation/toxicity , Electron Transport Complex I/genetics , Isoflurane/toxicity , Mitochondria/genetics , Mutation/genetics , Aging/drug effects , Aging/genetics , Aging/pathology , Animals , Animals, Genetically Modified , Drosophila , Male , Mitochondria/drug effects , Mitochondria/pathology , Mutation/drug effects , Sevoflurane/toxicity
12.
Br J Anaesth ; 125(1): 77-86, 2020 07.
Article in English | MEDLINE | ID: mdl-32466842

ABSTRACT

BACKGROUND: General anaesthetics interact with the pathophysiological mechanisms of traumatic brain injury (TBI). We used a Drosophila melanogaster (fruit fly) model to test the hypothesis that ageing and genetic background modulate the effect of anaesthetics and hyperoxia on TBI-induced mortality in the context of blunt trauma. METHODS: We exposed flies to isoflurane or sevoflurane under normoxic or hyperoxic conditions and TBI, and subsequently quantified the effect on mortality 24 h after injury. To determine the effect of age on anaesthetic-induced mortality, we analysed flies at 1-8 and 43-50 days old. To determine the effect of genetic background, we performed a genome-wide association study (GWAS) analysis on a collection of young inbred, fully sequenced lines. RESULTS: Exposure to anaesthetics and hyperoxia differentially affected mortality in young and old flies. Pre-exposure of young but not old flies to anaesthetics reduced mortality. Post-exposure selectively increased mortality. For old but not young flies, hyperoxia enhanced the effect on mortality of post-exposure to isoflurane but not to sevoflurane. Post-exposure to isoflurane in hyperoxia increased the mortality of young fly lines in the Drosophila Genetic Reference Panel collection to different extents. GWAS analysis of these data identified single nucleotide polymorphisms in genes involved in cell water regulation and oxygen sensing as being associated with the post-exposure effect on mortality. CONCLUSIONS: Ageing and genetic background influence the effects of volatile general anaesthetics and hyperoxia on mortality in the context of traumatic brain injury. Polymorphisms in specific genes are identified as potential causes of ageing and genetic effects.


Subject(s)
Aging/physiology , Anesthetics, Inhalation/pharmacology , Brain Injuries, Traumatic/physiopathology , Brain/physiopathology , Drosophila melanogaster , Genetic Background , Animals , Brain/drug effects , Brain Injuries, Traumatic/genetics , Disease Models, Animal , Genome-Wide Association Study/methods , Hyperoxia/physiopathology , Isoflurane/pharmacology , Polymorphism, Genetic/genetics , Sevoflurane/pharmacology , Wounds, Nonpenetrating/physiopathology
14.
Anesth Analg ; 127(5): e92-e93, 2018 11.
Article in English | MEDLINE | ID: mdl-30234541
15.
Anesth Analg ; 127(5): e85, 2018 11.
Article in English | MEDLINE | ID: mdl-30222654
16.
Anesth Analg ; 126(6): 1979-1986, 2018 06.
Article in English | MEDLINE | ID: mdl-29596093

ABSTRACT

BACKGROUND: Exposure to anesthetics is common in the majority of early survivors of life-threatening injuries. Whether and to what degree general anesthetics influence outcomes from major trauma is unknown. Potential confounding effects of general anesthetics on outcome measures are usually disregarded. We hypothesized that exposure to isoflurane or sevoflurane modulates the outcome from blunt trauma with traumatic brain injury (bTBI). METHODS: We tested the hypothesis in a novel model of bTBI implemented in Drosophila melanogaster. Fruit flies of the standard laboratory strain w were cultured under standard conditions. We titrated the severity of bTBI to a mortality index at 24 hours (MI24) of approximately 20% under control conditions. We administered standard doses of isoflurane and sevoflurane before, before and during, or after bTBI and measured the resulting MI24. We report the MI24 as mean ± standard deviation. RESULTS: Isoflurane or sevoflurane administered for 2 hours before bTBI reduced the MI24 from 22.3 ± 2.6 to 10.4 ± 1.8 (P < 10, n = 12) and from 19.3 ± 0.9 to 8.9 ± 1.1 (P < .0001, n = 8), respectively. In contrast, administration of isoflurane after bTBI increased the MI24 from 18.5% ± 4.3% to 25.3% ± 9.1% (P = .0026, n = 22), while sevoflurane had no effect (22.4 ± 7.1 and 21.5 ± 5.8, n = 22). CONCLUSIONS: In a whole animal model of bTBI, general anesthetics were not indifferent with respect to early mortality. Therefore, collateral effects of general anesthetics should be considered in the interpretation of results obtained in vertebrate trauma models. Invertebrate model organisms can serve as a productive platform to interrogate anesthetic targets that mediate collateral effects and to inform trauma research in higher organisms about the potential impact of anesthetics on outcomes.


Subject(s)
Anesthetics, Inhalation/toxicity , Brain Injuries, Traumatic/mortality , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Wounds, Nonpenetrating/mortality , Wounds, Nonpenetrating/pathology , Anesthetics, Inhalation/administration & dosage , Animals , Brain Injuries, Traumatic/chemically induced , Drosophila melanogaster , Female , Male , Mortality/trends , Wounds, Nonpenetrating/chemically induced
17.
Sci Rep ; 8(1): 2348, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402974

ABSTRACT

Genetic variability affects the response to numerous xenobiotics but its role in the clinically-observed irregular responses to general anesthetics remains uncertain. To investigate the pharmacogenetics of volatile general anesthetics (VGAs), we developed a Serial Anesthesia Array apparatus to expose multiple Drosophila melanogaster samples to VGAs and behavioral assays to determine pharmacokinetic and pharmacodynamic properties of VGAs. We studied the VGAs isoflurane and sevoflurane in four wild type strains from the Drosophila Genetic Reference Panel, two commonly used laboratory strains (Canton S and w 1118 ), and a mutant in Complex I of the mitochondrial electron transport chain (ND23 60114 ). In all seven strains, isoflurane was more potent than sevoflurane, as predicted by their relative lipid solubilities, and emergence from isoflurane was slower than from sevoflurane, reproducing cardinal pharmacokinetic and pharmacodynamic properties in mammals. In addition, ND23 60114 flies were more sensitive to both agents, as observed in worms, mice, and humans carrying Complex I mutations. Moreover, we found substantial variability among the fly strains both in absolute and in relative pharmacokinetic and pharmacodynamic profiles of isoflurane and sevoflurane. These data indicate that naturally occurring genetic variations measurably influence cardinal pharmacologic properties of VGAs and that flies can be used to identify relevant genetic variations.


Subject(s)
Anesthetics, Inhalation/pharmacokinetics , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Isoflurane/pharmacokinetics , Sevoflurane/pharmacokinetics , Animals , Female , Genetic Variation , Kinetics , Male , Mitochondria/genetics
18.
Anesth Analg ; 119(6): 1277-84, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25211390

ABSTRACT

BACKGROUND: Modulation of γ-aminobutyric acid type A receptors (GABAARs) by general anesthetics may contribute to their ability to produce amnesia. Receptors containing α5 subunits, which mediate tonic and slow synaptic inhibition, are co-localized with ß3 and γ2 subunits in dendritic layers of the hippocampus and are sensitive to low (amnestic) concentrations of anesthetics. Because α5 and ß3 subunits influence performance in hippocampus-dependent learning tasks in the presence and absence of general anesthetics, and the experimental inhaled drug 1,2-dichlorohexafluorocyclobutane (F6) impairs hippocampus-dependent learning, we hypothesized that F6 would modulate receptors that incorporate α5 and ß3 subunits. We hypothesized further that the ß3(N265M) mutation, which controls receptor modulation by general anesthetics, would similarly influence modulation by F6. METHODS: Using whole-cell electrophysiologic recording techniques, we tested the effects of F6 at concentrations ranging from 4 to 16 µM on receptors expressed in human embryonic kidney 293 cells. We measured drug modulation of wild-type α5ß3 and α5ß3γ2L GABAARs and receptors harboring the ß3(N265M) mutation. We also tested the effects of F6 on α1ß2γ2L receptors, which were reported previously to be insensitive to this drug when expressed in Xenopus oocytes. RESULTS: F6 enhanced the responses of wild-type α5ß3γ2L but not α1ß2γ2L receptors to low concentrations of GABA in a concentration-dependent manner. Receptors that incorporated the mutant ß3(N265M) subunit were insensitive to F6. When applied together with a high concentration of GABA, F6 blocked currents through α5ß3 but not α5ß3γ2L receptors. F6 did not alter deactivation of α5ß3γ2L receptors after brief high- concentration pulses of GABA. CONCLUSIONS: The nonimmobilizer F6 modulates GABAARs in a manner that depends on subunit composition and mode of receptor activation by GABA, supporting a possible role for α5-containing receptors in suppression of learning and memory by F6. Furthermore, common structural requirements indicate that similar molecular mechanisms may be responsible for the enhancing effects of F6 and conventional general anesthetics.


Subject(s)
Anesthetics/pharmacology , Chlorofluorocarbons/pharmacology , Cyclobutanes/pharmacology , GABA Modulators/pharmacology , Mutation , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , gamma-Aminobutyric Acid/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Kinetics , Membrane Potentials , Receptors, GABA-A/metabolism
19.
Anesth Analg ; 119(2): 311-320, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25046786

ABSTRACT

Within two decades of the discovery of anesthesia, the physicochemical concept of colloid and the biological concept of protoplasm had emerged. Fusion of these concepts into a theoretical framework, which has been largely forgotten decades ago, promised to uncover fundamental biological truths and determined research into anesthetic mechanisms for a century after "Ether Day." Observations of optical changes in unstained tissue were condensed into a theory of anesthesia by coagulation of protoplasm in the 1870s. The underlying hypotheses, conformational changes of proteins within the protoplasm cause all behavioral effects of anesthesia, continued to be pursued well into the 20th century. The goal was to explain anesthesia using physical chemistry within a fundamental cell biological framework. This large body of work, swept aside during the decades of lipid membrane hegemony, has remained in obscurity even after proteins in excitable membranes became firmly established as mediators of the immediate anesthetic effects. This article is a reminder of the prolonged interdisciplinary research effort dedicated to "protoplasmic theories" at a time when attention is increasingly directed toward examining the nature of (un)consciousness well as noncanonical consequences of anesthetic exposure that are not easily accounted for within conventional pharmacological concepts.


Subject(s)
Anesthesia/history , Anesthetics/history , Biomedical Research/history , Cytoplasm , Membrane Proteins/history , Anesthetics/chemistry , Anesthetics/pharmacology , Animals , Colloids , Cytoplasm/drug effects , Cytoplasm/metabolism , Flocculation , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Models, Biological , Protein Denaturation
20.
Anesthesiology ; 118(4): 986-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23511527
SELECTION OF CITATIONS
SEARCH DETAIL