Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Anesth Analg ; 118(6): 1284-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24413549

ABSTRACT

BACKGROUND: Volatile anesthetics cause widespread apoptosis in the developing brain. Carbon monoxide (CO) has antiapoptotic properties, and exhaled endogenous CO is commonly rebreathed during low-flow anesthesia in infants and children, resulting in subclinical CO exposure. Thus, we aimed to determine whether CO could limit isoflurane-induced apoptosis in the developing brain. METHODS: Seven-day-old male CD-1 mouse pups underwent 1-hour exposure to 0 (air), 5, or 100 ppm CO in air with or without isoflurane (2%). We assessed carboxyhemoglobin levels, cytochrome c peroxidase activity, and cytochrome c release from forebrain mitochondria after exposure and quantified the number of activated caspase-3 positive cells and TUNEL positive nuclei in neocortex, hippocampus, and hypothalamus/thalamus. RESULTS: Carboxyhemoglobin levels approximated those expected in humans after a similar time-weighted CO exposure. Isoflurane significantly increased cytochrome c peroxidase activity, cytochrome c release, the number of activated caspase-3 cells, and TUNEL positive nuclei in the forebrain of air-exposed mice. CO, however, abrogated isoflurane-induced cytochrome c peroxidase activation and cytochrome c release from forebrain mitochondria and decreased the number of activated caspase-3 positive cells and TUNEL positive nuclei after simultaneous exposure with isoflurane. CONCLUSIONS: Taken together, the data indicate that CO can limit apoptosis after isoflurane exposure via inhibition of cytochrome c peroxidase depending on concentration. Although it is unknown whether CO directly inhibited isoflurane-induced apoptosis, it is possible that low-flow anesthesia designed to target rebreathing of specific concentrations of CO may be a desired strategy to develop in the future in an effort to prevent anesthesia-induced neurotoxicity in infants and children.


Subject(s)
Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/toxicity , Apoptosis/drug effects , Brain/drug effects , Carbon Monoxide/pharmacology , Isoflurane/antagonists & inhibitors , Isoflurane/toxicity , Animals , Animals, Newborn , Carboxyhemoglobin/metabolism , Caspase 3/metabolism , Cytochrome-c Peroxidase/antagonists & inhibitors , Cytochrome-c Peroxidase/metabolism , Cytochromes c/metabolism , Female , Heme/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Pregnancy
2.
Br J Anaesth ; 110 Suppl 1: i73-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23535829

ABSTRACT

BACKGROUND: Animal studies have shown that memory deficits in the early post-anaesthetic period can be prevented by pre-treatment with an inverse agonist that preferentially inhibits α5 subunit-containing γ-aminobutyric acid type A (α5GABA(A)) receptors. The goal of this in vitro study was to determine whether inverse agonists that inhibit α5GABA(A) receptors reduce anaesthetic potentiation of GABAA receptor activity. METHODS: Cultures of hippocampal neurones were prepared from Swiss white mice, wild-type mice (genetic background C57BL/6J and Sv129Ev) and α5GABA(A)receptor null mutant (Gabra5-/-) mice. Whole-cell voltage clamp techniques were used to study the effects of the α5GABA(A) receptor-preferring inverse agonists L-655,708 and MRK-016 on anaesthetic potentiation of GABA-evoked currents. RESULTS: L-655,708 (50 nM) reduced sevoflurane potentiation of GABA-evoked current in wild-type neurones but not Gabra5-/- neurones, and produced a rightward shift in the sevoflurane concentration-response plot [sevoflurane EC50: 1.9 (0.1) mM; sevoflurane+L-655,708 EC(50): 2.4 (0.2) mM, P<0.05]. Similarly, L-655,708 (50 nM) reduced isoflurane potentiation of GABA-evoked current [isoflurane: 4.0 (0.6) pA pF(-1); isoflurane+L-655,708: 3.1 (0.5) pA pF(-1), P<0.01]. MRK-016 also reduced sevoflurane and isoflurane enhancement of GABA-evoked current [sevoflurane: 1.5 (0.1) pA pF(-1); sevoflurane+MRK-016 (10 nM): 1.2 (0.1) pA pF(-1), P<0.05; isoflurane: 3.5 (0.3) pA pF(-1); isoflurane+MRK-016 (1 nM): 2.9 (0.2) pA pF(-1), P<0.05]. CONCLUSIONS: L-655,708 and MRK-016 reduced the potentiation by inhaled anaesthetics of GABAA receptor activated by a low concentration of GABA. Future studies are required to determine whether this effect contributes to the memory preserving properties of inverse agonists after anaesthesia.


Subject(s)
Anesthetics, Inhalation/pharmacology , Neurons/drug effects , Receptors, GABA-A/drug effects , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Evoked Potentials/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Imidazoles/administration & dosage , Imidazoles/pharmacology , Isoflurane/antagonists & inhibitors , Isoflurane/pharmacology , Isoxazoles/administration & dosage , Isoxazoles/pharmacology , Methyl Ethers/administration & dosage , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Patch-Clamp Techniques , Receptors, GABA-A/deficiency , Receptors, GABA-A/physiology , Sevoflurane , Triazines/administration & dosage , Triazines/pharmacology
3.
Br J Anaesth ; 110 Suppl 1: i82-91, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23604542

ABSTRACT

BACKGROUND: Isoflurane can increase pro-inflammatory cytokine interleukin (IL)-6 levels. However, the up-stream mechanism remains unknown. Nuclear factor-kappa B (NF-κB) promotes the generation of pro-inflammatory cytokines. We examined the effects of isoflurane and sevoflurane on the NF-κB signalling pathway and its association with IL-6 levels in cultured cells. METHODS: H4 human neuroglioma cells (H4 cells), and mouse primary neurones and microglia were treated with 2% isoflurane or 4.1% sevoflurane for 6 h, for analysis of IL-6 and NF-κB. Pyrrolidine dithiocarbamate (an NF-κB inhibitor) or 2-deoxy-d-glucose (2-DG) (an inhibitor of glucose glycolysis) was applied 1 h before anaesthetic treatment. RESULTS: Isoflurane or sevoflurane treatment increased the levels of IL-6 [isoflurane: 410% (54); sevoflurane: 290% (24)], the nuclear levels of NF-κB [isoflurane: 170% (36); sevoflurane: 320% (30)], and the transcription activity of NF-κB in H4 cells. Moreover, isoflurane enhanced the transcription activity of NF-κB in mouse microglia, but not primary neurones. Finally, pyrrolidine dithiocarbamate and 2-DG attenuated isoflurane-induced increases in IL-6 and NF-κB, and the transcription activity of NF-κB. CONCLUSIONS: These studies in H4 cells suggest that the NF-κB signalling pathway could contribute to isoflurane or sevoflurane-induced neuroinflammation. This could lead to the targeted intervention of anaesthetic-induced neuroinflammation.


Subject(s)
Anesthetics, Inhalation/pharmacology , Inflammation Mediators/metabolism , Interleukin-6/biosynthesis , Microglia/drug effects , NF-kappa B/physiology , Neurons/drug effects , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Antimetabolites/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Deoxyglucose/pharmacology , Glioma , Humans , Isoflurane/antagonists & inhibitors , Isoflurane/pharmacology , Methyl Ethers/pharmacology , Mice , Microglia/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , Neurons/metabolism , Proline/analogs & derivatives , Proline/pharmacology , Sevoflurane , Signal Transduction/drug effects , Signal Transduction/physiology , Thiocarbamates/pharmacology , Transcription, Genetic/drug effects , Tumor Cells, Cultured
4.
Anesth Analg ; 113(5): 1152-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21918167

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.


Subject(s)
Anesthetics, Inhalation/pharmacology , Excitatory Amino Acid Agonists/toxicity , Hippocampus/drug effects , Isoflurane/pharmacology , Pyramidal Cells/drug effects , gamma-Aminobutyric Acid/toxicity , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Blotting, Western , Calcium/metabolism , Calcium Channels/drug effects , Calcium Signaling/drug effects , Caspase 3/biosynthesis , Cells, Cultured , Dose-Response Relationship, Drug , Hippocampus/cytology , Isoflurane/antagonists & inhibitors , Patch-Clamp Techniques , Pyramidal Cells/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/drug effects
5.
Anesthesiology ; 112(3): 567-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124973

ABSTRACT

BACKGROUND: We tested the hypothesis that in newborn rats, sevoflurane may cause seizures, neurotoxicity, and impairment in synaptic plasticity-effects that may be diminished by the Na-K-2Cl cotransporter 1 inhibitor, bumetanide. METHODS: Electroencephalography, activated caspase-3, and hippocampal long-term potentiation were measured in rats exposed to 2.1% sevoflurane for 0.5-6 h at postnatal days 4-17 (P4-P17). RESULTS: Arterial blood gas samples drawn at a sevoflurane concentration of 2.1% showed no evidence of either hypoxia or hypoventilation in spontaneously breathing rats. Higher doses of sevoflurane (e.g., 2.9%) caused respiratory depression. During anesthesia maintenance, the electroencephalography exhibited distinctive episodes of epileptic seizures in 40% of P4-P8 rats. Such seizure-like activity was not detected during anesthesia maintenance in P10-P17 rats. Emergence from 3 h of anesthesia with sevoflurane resulted in tonic/clonic seizures in some P10-P17 rats but not in P4-P8 rats. Bumetanide (5 micromol/kg, intraperitoneally) significantly decreased seizures in P4-P9 rats but did not affect the emergence seizures in P10-P17 rats. Anesthesia of P4 rats with sevoflurane for 6 h caused a significant increase in activated caspase-3 and impairment of long-term potentiation induction measured at 1 and 14-17 days after exposure to sevoflurane, respectively. Pretreatment of P4 rats with bumetanide nearly abolished the increase in activated caspase-3 but did not alleviate impairment of long-term potentiation. CONCLUSION: These results support the possibility that excitatory output of sevoflurane-potentiated gamma-aminobutyric acid type A/glycine systems may contribute to epileptogenic and neurotoxic effects in early postnatal rats.


Subject(s)
Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/toxicity , Bumetanide/therapeutic use , Diuretics/therapeutic use , Epilepsy/chemically induced , Epilepsy/prevention & control , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/toxicity , Neurotoxicity Syndromes/prevention & control , Anesthesia, Inhalation , Animals , Animals, Newborn , Blotting, Western , Caspase 3/metabolism , Electroencephalography/drug effects , Electrophysiology , Enzyme Activation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Sevoflurane
6.
Anesthesiology ; 112(3): 614-22, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124979

ABSTRACT

BACKGROUND: The general anesthetic gas xenon is neuroprotective and is undergoing clinical trials as a treatment for ischemic brain injury. A small number of molecular targets for xenon have been identified, the N-methyl-D-aspartate (NMDA) receptor, the two-pore-domain potassium channel TREK-1, and the adenosine triphosphate-sensitive potassium channel (KATP). However, which of these targets are relevant to acute xenon neuroprotection is not known. Xenon inhibits NMDA receptors by competing with glycine at the glycine-binding site. We test the hypothesis that inhibition of the NMDA receptor at the glycine site underlies xenon neuroprotection against hypoxia-ischemia. METHODS: We use an in vitro model of hypoxia-ischemia to investigate the mechanism of xenon neuroprotection. Organotypic hippocampal brain slices from mice are subjected to oxygen-glucose deprivation, and injury is quantified by propidium iodide fluorescence. RESULTS: We show that 50% atm xenon is neuroprotective against hypoxia-ischemia when applied immediately after injury or after a delay of 3 h after injury. To validate our method, we show that neuroprotection by gavestinel is abolished when glycine is added, confirming that NMDA receptor glycine site antagonism underlies gavestinel neuroprotection. We then show that adding glycine abolishes the neuroprotective effect of xenon, consistent with competitive inhibition at the NMDA receptor glycine site mediating xenon neuroprotection. CONCLUSIONS: We show that xenon neuroprotection against hypoxia- ischemia can be reversed by increasing the glycine concentration. This is consistent with competitive inhibition by xenon at the NMDA receptor glycine site, playing a significant role in xenon neuroprotection. This finding may have important implications for xenon's clinical use as an anesthetic and neuroprotectant.


Subject(s)
Anesthetics, Inhalation/pharmacology , Hypoxia-Ischemia, Brain/prevention & control , Neuroprotective Agents , Receptors, Glycine/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Xenon/pharmacology , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Binding, Competitive/drug effects , Coloring Agents , Excitatory Amino Acid Antagonists/pharmacology , Glucose/deficiency , Glycine/pharmacology , Glycine Agents/pharmacology , Hippocampus/pathology , Hyperbaric Oxygenation , Hypoxia-Ischemia, Brain/pathology , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Neurons/pathology , Neuroprotective Agents/antagonists & inhibitors , Organ Culture Techniques , Propidium , Xenon/antagonists & inhibitors
8.
Anesth Analg ; 109(2): 641-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19608842

ABSTRACT

BACKGROUND: The mechanism and site of action within the spinal cord by which volatile anesthetics produce immobility are not well understood. Little work has been done directly comparing anesthetic effects on neurons with specific functional characteristics that mediate transfer of nociceptive information within the spinal cord. METHODS: Adult male rats were anesthetized and prepared for extracellular single-unit recordings from the lumbar dorsal horn. Nociceptive-specific (NS) and wide dynamic range (WDR) neurons were identified and noxious heat-evoked neuronal spike rates evaluated at 0.8 and 1.2 anesthetic minimum alveolar anesthetic concentration (MAC) halothane or isoflurane. In another group, noxious heat-evoked responses from NS neurons were evaluated at 0.8, 1.2 MAC halothane, and 1.2 MAC halothane plus IV naloxone (0.1 mg/kg). RESULTS: Increasing halothane from 0.8 to 1.2 MAC reduced the heat-evoked neuronal responses of NS neurons (n = 9) from 827 +/- 122 (mean +/- se) to 343 +/- 48 spikes/min (P < 0.05) but not WDR neurons (n = 9), 617 +/- 79 to 547 +/- 78 spikes/min. Increasing isoflurane from 0.8 to 1.2 MAC reduced the heat-evoked neuronal response of NS neurons (n = 9) from 890 +/- 339 to 188 +/- 97 spikes/min (P < 0.05) but did not alter the response of WDR neurons (n = 9) in which evoked spike rate went from 576 +/- 132 to 601 +/- 119 spikes/min. In a separate group, the response of NS neurons went from 282 +/- 60 to 74 +/- 32 spikes/min (P < 0.05) when halothane was increased from 0.8 to 1.2 MAC. IV administration of naloxone increased the heat-evoked response to 155 +/- 46 spikes/min (P < 0.05). CONCLUSIONS: NS but not WDR neurons in the lumbar dorsal horn are depressed by peri-MAC increases of halothane and isoflurane. This depression, at least with halothane, can be partially reversed by the opioid antagonist naloxone. Given that opioid receptors are not likely involved in the mechanisms by which volatile anesthetics produce immobility, this suggests that, although the neuronal depression is of substantial magnitude and occurs concurrent to the production of immobility, it may not play a major role in the production of this anesthetic end point.


Subject(s)
Anesthetics, Inhalation/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Nociceptors/drug effects , Posterior Horn Cells/drug effects , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Depression, Chemical , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiology , Halothane/pharmacology , Hot Temperature , Isoflurane/pharmacology , Male , Microelectrodes , Rats , Rats, Sprague-Dawley
9.
J Anesth ; 23(2): 301-3, 2009.
Article in English | MEDLINE | ID: mdl-19444578

ABSTRACT

The mechanism of the analgesic effect of nitrous oxide (N(2)O) has not been completely clarified. Although we have reported that the analgesic effect of N(2)O was significantly decreased in nociceptin-orphanin FQ (N/OFQ) receptor (NOP)-deficient mice, the effect of nociceptin receptor antagonists on N(2)O-induced analgesia has not been reported. In this investigation, we examined the effect of the NOP antagonist JTC-801 on N(2)O-induced analgesia in 129Sv mice by the writhing test and tail flick test, and demonstrated that the analgesic effect of N(2)O was suppressed by the intraperitoneal administration of JTC-801.


Subject(s)
Aminoquinolines/pharmacology , Anesthetics, Inhalation/antagonists & inhibitors , Benzamides/pharmacology , Narcotic Antagonists , Nitrous Oxide/antagonists & inhibitors , Acetic Acid , Anesthetics, Inhalation/pharmacology , Animals , Male , Mice , Nitrous Oxide/pharmacology , Pain Measurement/drug effects , Receptors, Opioid , Nociceptin Receptor
10.
Anaesthesia ; 63(6): 583-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18279487

ABSTRACT

Aminophylline is usually used during anaesthesia to treat bronchospasm but recent findings suggest that it can also be used to shorten recovery time after general anaesthesia. However, it is unclear whether aminophylline shows similar properties during a steady-state phase of deep surgical anaesthesia. We therefore wanted to test the hypothesis that the administration of aminophylline leads to an increase in bispectral index as a surrogate parameter suggesting a lighter plane of anaesthesia. The study was designed as a double-blind, randomised, controlled trial with two main groups (aminophylline and placebo) and two subgroups (sevoflurane and propofol). We studied 60 patients. The injection of aminophylline 3 mg x kg(-1) was associated with significant increases in bispectral index up to 10 min after its injection, while heart rate and blood pressure did not change. It appears that aminophylline has the ability to partially antagonise the sedative effects of general anaesthetics.


Subject(s)
Aminophylline/pharmacology , Anesthesia, Inhalation , Anesthesia, Intravenous , Electroencephalography/drug effects , Adult , Aged , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Intravenous/antagonists & inhibitors , Blood Pressure/drug effects , Bronchodilator Agents/pharmacology , Double-Blind Method , Female , Heart Rate/drug effects , Humans , Male , Middle Aged , Monitoring, Intraoperative/methods
12.
Genetics ; 161(1): 109-19, 2002 May.
Article in English | MEDLINE | ID: mdl-12019227

ABSTRACT

Volatile anesthetics (VAs) disrupt nervous system function by an ill-defined mechanism with no known specific antagonists. During the course of characterizing the response of the nematode C. elegans to VAs, we discovered that a C. elegans pheromone antagonizes the VA halothane. Acute exposure to pheromone rendered wild-type C. elegans resistant to clinical concentrations of halothane, increasing the EC(50) from 0.43 +/- 0.03 to 0.90 +/- 0.02. C. elegans mutants that disrupt the function of sensory neurons required for the action of the previously characterized dauer pheromone blocked pheromone-induced resistance (Pir) to halothane. Pheromone preparations from loss-of-function mutants of daf-22, a gene required for dauer pheromone production, lacked the halothane-resistance activity, suggesting that dauer and Pir pheromone are identical. However, the pathways for pheromone's effects on dauer formation and VA action were not identical. Not all mutations that alter dauer formation affected the Pir phenotype. Further, mutations in genes not known to be involved in dauer formation completely blocked Pir, including those altering signaling through the G proteins Goalpha and Gqalpha. A model in which sensory neurons transduce the pheromone activity through antagonistic Go and Gq pathways, modulating VA action against neurotransmitter release machinery, is proposed.


Subject(s)
Caenorhabditis elegans/metabolism , Halothane/antagonists & inhibitors , Pheromones/metabolism , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/metabolism , Animals , Caenorhabditis elegans/genetics , Drug Resistance/genetics , GTP-Binding Protein alpha Subunits, Gi-Go , Halothane/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Larva/metabolism , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Signal Transduction
13.
Biochem Pharmacol ; 50(9): 1373-7, 1995 Oct 26.
Article in English | MEDLINE | ID: mdl-7503786

ABSTRACT

The influence of spermidine, a polyamine agonist, on volatile anesthetic inhibition of N-methyl-D-aspartate (NMDA) receptor activation, as indicated by glutamate stimulation of [3H]MK-801 ([3H](5D,10S)-(+)-methyl-10,11-dihydro-5H- dibenzo[a,d]cyclohepten-5,10-imine) binding, was studied in rat brain. Spermidine reserved the inhibition caused by four volatile anesthetics (enflurane, halothane, methoxyflurane and chloroform) at the same concentrations (EC50 approximately 3 microM) at which it potentiated glutamate opening of the NMDA ion channel. The anesthetics had no effect on the direct stimulation of channel opening by spermidine, which occurred at concentrations of spermidine greater than 30 microM in the absence of receptor agonist. In these actions, spermidine closely resembled the allosteric co-agonist glycine. The present results suggest that anesthetic action on NMDA receptors involves a set of sites on the channel complex that is distinct from the recognition sites for glutamate, glycine, and channel blockers, and are consistent with the idea that blockade of NMDA receptors contributes to the development of the anesthetic state.


Subject(s)
Anesthetics, Inhalation/pharmacology , Brain/ultrastructure , Dizocilpine Maleate/metabolism , Excitatory Amino Acid Antagonists/metabolism , Glutamic Acid/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Spermidine/pharmacology , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Depression, Chemical , Drug Interactions , Male , Rats , Rats, Wistar , Signal Transduction/drug effects , Stimulation, Chemical , Tritium
14.
Eur J Pharmacol ; 503(1-3): 49-53, 2004 Oct 25.
Article in English | MEDLINE | ID: mdl-15496295

ABSTRACT

The activation of intracerebral corticotropin releasing factor (CRF) system is involved in nitrous oxide analgesia. We evaluated the effect of general anesthetics on nitrous-oxide-induced CRF activation and antinociception. Male Sprague-Dawley rats inhaled isoflurane (0%, 0.6%, 1.0% and 1.5%) or were administered with intravenous propofol (0, 0.1 and 0.2 mg/kg/min), with or without 75% nitrous oxide inhalation, for 90 min. The brain was fixed with fixative, and brain sections, including the paraventricular nucleus of the hypothalamus, were double immunostained with c-Fos and CRF antibodies to assess the activation of CRF-containing neurons. In other groups of rats, the effect of propofol on nitrous oxide antinociception was evaluated with tail flick latency tests. Both inhaled isoflurane and intravenous propofol inhibited nitrous-oxide-induced activation of CRF neurons, suggesting that these general anesthetics may inhibit one of the analgesic mechanisms of nitrous oxide. Indeed, propofol inhibited the antinociceptive action of nitrous oxide, as evaluated with tail flick latencies (TFL).


Subject(s)
Anesthetics, General/pharmacology , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/pharmacology , Corticotropin-Releasing Hormone/physiology , Neurons/drug effects , Nitrous Oxide/antagonists & inhibitors , Nitrous Oxide/pharmacology , Animals , Biomarkers , Brain Chemistry/drug effects , Immunohistochemistry , Isoflurane/pharmacology , Male , Pain Measurement/drug effects , Propofol/pharmacology , Proto-Oncogene Proteins c-fos/chemistry , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects
15.
Naunyn Schmiedebergs Arch Pharmacol ; 360(4): 376-81, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10551274

ABSTRACT

Administration of 5-HT2 receptor agonists induced malignant hyperthermia (MH) in susceptible pigs. Furthermore, the 5-HT2 receptor antagonist ritanserin prevented 5-HT-induced porcine MH. It has been shown that 5-HT2 receptor agonists induce marked contractures in skeletal muscle specimens from MH susceptible (MHS) but not in specimens from normal patients. The purpose of this study was to investigate the effects of ritanserin on halothane-induced contractures in muscle specimens from MHS patients. Twenty-five patients aged 8-56 years (29.5+/-13.6) classified as MHS by the in vitro contracture test (IVCT) with halothane and caffeine according to the protocol of the European MH Group participated in this study. Muscle specimens were pretreated with ritanserin 10 micromol/l (n= 14), 20 micromol/l (n=14) and 100 micromol/l (n=12) for 10 min and subsequently halothane was added incrementally (0.11-0.22-0.44 mmol/l) to the tissue bath as described in the European MH protocol. The results of the halothane contracture test were used as control. Following administration of halothane, muscle contractures reached a maximum of 16.9+/-4.2 mN. Ritanserin led to a significant inhibition of halothane-induced contractures in MHS muscles. Following pretreatment with ritanserin, halothane-induced contracture maximum was significantly smaller with 7.5+/-3.1 mN after 10 micromol/l ritanserin, 4.9+/-1.5 mN after 20 micromol/l ritanserin and 0.5+/- 0.2 mN after 100 micromol/l ritanserin than without pretreatment. Administration of ritanserin induced at all concentrations a decrease in muscle twitch height. Increase in muscle twitch following halothane was reduced in a concentration-dependent manner by ritanserin. The presented findings indicate that 5-HT might be involved in the mechanisms of halothane-induced MH in humans. Further studies have to determine the pathophysiological role of the 5-HT system in MH, and whether ritanserin could be an alternative for treatment or prevention of halothane-induced MH.


Subject(s)
Malignant Hyperthermia/pathology , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Ritanserin/pharmacology , Serotonin Antagonists/pharmacology , Adolescent , Adult , Anesthetics, Inhalation/antagonists & inhibitors , Caffeine/pharmacology , Child , Dose-Response Relationship, Drug , Drug Interactions , Halothane/antagonists & inhibitors , Humans , Malignant Hyperthermia/etiology , Middle Aged , Time Factors
16.
Nucl Med Commun ; 23(1): 53-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11748438

ABSTRACT

Halothane and its metabolites cause liver damage by decreasing liver blood flow and generating free-radical species. Catechin suppresses lipid peroxidation and increases enzyme activity, therefore it seems to be capable of protecting liver parenchyma against the direct toxic effect of halothane. The aim of this study was to investigate the role of hepatobiliary scintigraphy in detecting liver damage after halothane anaesthesia and the protective effect of catechin in comparison with histo-chemical analysis. Thirty rabbits, divided into three groups (A, controls; B, halothane; and C, catechin+halothane), were investigated. In group A no anaesthesia was administered. Group B only received halothane, while group C was pretreated with catechin and halothane anaesthesia was administered for 2 h. Dynamic scintigrams were taken for 60 min after injecting 99mTc-mebrofenin, and the time of peak uptake (TPU) and the time for half of the activity to clear from the liver (T(1/2)) were calculated. Rabbits were killed, and malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT) levels were measured in hepatic tissue. The TPU and T(1/2) values of group A is significantly lower than in groups B and C (P<0.0002 and P<0.0002, respectively, for TPU; and P<0.0002 and P<0.0003, respectively, for T(1/2)). The TPU and T(1/2) values of group B were significantly higher than in group C (P<0.0003 and P<0.0003, respectively). The hepatic MDA level of group A was significantly lower than in groups B and C (P<0.0002 and P<0.0002, respectively). SOD, GSH-Px and CAT levels of group A were significantly higher than in groups B and C (P<0.0002, P<0.0001 and P<0.003, respectively, for group A vs group B; and P<0.0005, P<0.0002 and P<0.03, respectively, for group A vs group C). The MDA level of group B was significantly higher than that in group C (P<0.0002). SOD, GSH-Px and CAT levels of group B were significantly lower than in group C (P<0.0002, P<0.0002 and P<0.003, respectively). According to these results, we suggest that catechin protects liver parenchyma against the toxic effect of halothane and its metabolites, and that, compared to invasive histo-chemical analysis, hepatobiliary scintigraphy is a useful and alternative non-invasive method for detecting the protective effect of catechin on liver parenchyma after halothane anaesthesia.


Subject(s)
Anesthetics, Inhalation/toxicity , Bile Ducts/diagnostic imaging , Catechin/therapeutic use , Chemical and Drug Induced Liver Injury/diagnostic imaging , Chemical and Drug Induced Liver Injury/prevention & control , Halothane/toxicity , Liver/diagnostic imaging , Liver/pathology , Anesthesia , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Chemical and Drug Induced Liver Injury/enzymology , Half-Life , Halothane/antagonists & inhibitors , Histocytochemistry , Liver/enzymology , Rabbits , Radionuclide Imaging
17.
Masui ; 47(8): 944-54, 1998 Aug.
Article in Japanese | MEDLINE | ID: mdl-9753959

ABSTRACT

In the present study, effects of midazolam, thiopental sodium, propofol, and nitrous oxide upon SEP in a clinically used dose were investigated on 24 male volunteers. In addition, antagonistic actions of flumazenil and naloxone against effects of midazolam and nitrous oxide, respectively, on SEP were studied. Midazolam had no effect on latencies of N 20 and P 25, but increased latency of P 45 and attenuated P 100 amplitude. Flumazenil reversed these effects of midazolam of P 45 latency and P 100 amplitude to their control values. While thiopental sodium and propofol suppressed P 100 amplitude, they had no effect on N 20, P 25, P 45 latencies. Nitrous oxide elongated latencies of N 20, P 25, P 45 and decreased P 100 amplitude. Naloxone reversed the effects of nitrous oxide on N 20 and P 25 latencies without affecting increased P 45 latency and attenuated P 100 amplitude. These results suggest that midazolam might have an analgesic action of suppressing cortical sensory neurons, whereas thiopental sodium and propofol have no effect on neurons in the primary sensory cortex. The finding that naloxone antagonized the increased latencies of N 20 and P 25 by nitrous oxide could be explained by the analgesic action of nitrous oxide that could be mediated by opioid receptors. The results also indicate that electrical activities of the cortical neurons in the associated area are more susceptible to psychotropic agents than those in the primary sensory cortex. The effects of anesthetics on SEP appear to reflect their characteristics of functioning mechanisms on cortical neurons. Analysis of SEP is, therefore, useful for the assessment of the mechanism and the acting site of anesthetics in the sensory cortex.


Subject(s)
Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Evoked Potentials, Somatosensory/drug effects , Median Nerve/physiology , Adult , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Intravenous/antagonists & inhibitors , Electric Stimulation , Flumazenil/pharmacology , Humans , Male , Midazolam/antagonists & inhibitors , Midazolam/pharmacology , Naloxone/pharmacology , Nitrous Oxide/antagonists & inhibitors , Nitrous Oxide/pharmacology , Propofol/pharmacology , Reaction Time/drug effects , Thiopental/pharmacology
18.
Ann Fr Anesth Reanim ; 32(10): e135-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24011619

ABSTRACT

BACKGROUND AND PURPOSE: Volatile anesthetic isoflurane contributes to postoperative cognitive dysfunction and inhibition of long-term potentiation (LTP), a synaptic model of learning and memory, but the mechanisms are uncertain. Central neuronal α4ß2 subtype nicotinic acetylcholine receptors (nAChRs) are involved in the induction of LTP in the hippocampus. Isoflurane inhibits α4ß2 nAChRs at concentrations lower than those used for anesthesia. Therefore, we hypothesized that isoflurane-inhibited LTP induction of hippocampal CA1 neurons via α4ß2 nAChRs subtype inhibition. METHODS: Transverse hippocampal slices (400µm thick) were obtained from male rats (6-8 weeks old). Population spikes were evoked using extracellular electrodes by electrical stimulation of the Schaffer collateral-commissural pathway of rat hippocampal slices. LTP was induced using high frequency stimulation (HFS; 100Hz, 1s). Clinically relevant concentrations (0.125-0.5mM) of isoflurane with or without nicotine (nAChRs agonist), mecamylamine (nAChRs antagonist), 3-[2(S)-2-azetidinylmethoxy] pyridine (A85380) and epibatidine (α4ß2 nAChRs agonist), dihydro ß erythroidine (DHßE) (α4ß2 nAChRs antagonist) were added to the perfusion solution 20min before HFS to test their effects on LTP by HFS respectively. RESULTS: A brief HFS induced stable LTP in rat hippocampal slices, but LTP was significantly inhibited in the presence of isoflurane at concentrations of 0.125-0.5mM. The inhibitive effect of isoflurane on LTP was not only reversible and could be prevented by nAChRs agonist nicotine and α4ß2 nAChRs agonist A85380 and epibatidine, but also mimicked and potentiated by nAChRs antagonist mecamylamine and α4ß2 nAChRs antagonist DHßE. CONCLUSIONS: Inhibition of α4ß2 nAChRs subtype of hippocampus participates in isoflurane-mediated LTP inhibition.


Subject(s)
Anesthetics, Inhalation/pharmacology , CA1 Region, Hippocampal/drug effects , Isoflurane/pharmacology , Long-Term Potentiation/drug effects , Muscarinic Agonists/pharmacology , Neurons/drug effects , Receptors, Nicotinic/drug effects , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Azetidines/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CA1 Region, Hippocampal/cytology , Dihydro-beta-Erythroidine/pharmacology , Drug Interactions , Electric Stimulation , In Vitro Techniques , Isoflurane/antagonists & inhibitors , Mecamylamine/pharmacology , Neural Pathways/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
19.
Curr Alzheimer Res ; 9(3): 334-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22272607

ABSTRACT

There is an open controversy about the role of surgery and anesthesia in the pathogenesis of Alzheimer's disease (AD). Clinical studies have shown a high prevalence of these procedures in subjects with AD but the interpretation of these studies is difficult because of the co-existence of multiple variables. Experimental studies in vitro and in vivo have shown that small molecular weight volatile anesthetics enhance amyloidogenesis in vitro and produce behavioral deficits and brain lesions similar to those found in patients with AD. We examined the effect of co-treatment with trehalose on isoflurane-induced amyloidogenesis in mice. WT and APP(swe) mice, of 11 months of age, were exposed to 1% isoflurane, 3 times, for 1.5 hours each time and sacrificed 24 hours after their last exposure to isoflurane. The right hemi-brain was used for histological analysis and the contra-lateral hemi-brain used for biochemical studies. In this study, we have shown that repetitive exposure to isoflurane in pre-symptomatic mature APP(swe) mice increases apoptosis in hippocampus and cerebral cortex, enhances astrogliosis and the expression of GFAP and that these effects are prevented by co-treatment with trehalose, a disaccharide with known effects as enhancer of autophagy. We have also confirmed that in our model the co-treatment with trehalose increases the expression of autophagic markers as well as the expression of chaperones. Cotreatment with trehalose reduces the levels of ß amyloid peptide aggregates, tau plaques and levels of phospho-tau. Our study, therefore, provides new therapeutic avenues that could help to prevent the putative pro-amyloidogenic properties of small volatile anesthetics.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Isoflurane/antagonists & inhibitors , Isoflurane/toxicity , Plaque, Amyloid/drug therapy , Trehalose/pharmacology , Alzheimer Disease/physiopathology , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/toxicity , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/pharmacology , Plaque, Amyloid/pathology , Treatment Outcome
20.
Neuroscience ; 205: 167-77, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22244976

ABSTRACT

The present study was designed to evaluate the possible neuroprotective effects of metabotropic glutamate receptor (mGluR7) allosteric agonist N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) on developmental sevoflurane neurotoxicity. To achieve the objective, hippocampal cultures (7 DIV, 7 day in vitro) were treated with different doses of L-(+)-2-amino-4-phosphonobutyric acid (L-AP4, an agonist of group III mGluRs), (RS)-α-Methylserine-O-phosphate (MSOP, an antagonist of group III mGluRs), AMN082 or cis-2-[[(3,5-dichlorophenyl)amino]carbonyl]cyclohexanecarboxylic acid (VU0155041, an agonist of mGluR4) before exposed to sevoflurane. Cell apoptosis were determined by flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick-end-labeling (TUNEL)-staining. For in vivo study, rat pups (7 PND, 7 postnatal day) were injected with AMN082, L-AP4 or saline before sevoflurane exposure. Extracellular signal-regulated kinase 1 and 2 (ERK1/2), c-Jun N-terminal kinase (JNK), p38, caspase-3, Bcl-2, and Bax were detected by Western blot. The locomotor activity and cognitive functions were evaluated by open-field test and Morris water maze (MWM), respectively. We found that L-AP4 prevented sevoflurane-induced cell apoptosis, but MSOP promoted. Specially, application of AMN082 contributed to the relief of sevoflurane-induced apoptosis in vitro, whereas VU0155041 did not. In addition, sevoflurane treatment led to a decrease of Bcl-2 and an increase of caspase-3 and Bax, which were mitigated by AMNO82 in vivo. Moreover, we showed that sevoflurane treatment resulted in a remarkable suppression of phospho-ERK1/2, which was restored by AMN082. Application of U0126 (an inhibitor of MEK) abolished the neuroprotective effects of AMN082 on sevoflurane neurotoxicity both in vitro and in vivo. In addition, sevoflurane exposure also led to an increase of phospho-JNK, but SP600125 (an inhibitor of JNK) did not attenuate sevoflurane-induced apoptosis. The total and phosphorylated p38 remained unchanged in sevoflurane-treated rat pups. Finally, AMN082 improved the learning and memory defects caused by postnatal sevoflurane exposure without alternations in emotion or locomotor activity. These preliminary data indicate that AMN082 may protect immature brain against sevoflurane neurotoxicity, and the ERK1/2 MAP kinase signaling is likely to be involved. Further studies are needed to fully assess the neuroprotective role of mGluR7 agonist AMN082 in developmental anesthetic neurotoxicity.


Subject(s)
Benzhydryl Compounds/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/toxicity , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Receptors, Metabotropic Glutamate/agonists , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/toxicity , Animals , Animals, Newborn , Benzhydryl Compounds/therapeutic use , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/physiology , Sevoflurane
SELECTION OF CITATIONS
SEARCH DETAIL