Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Dev Biol ; 435(2): 185-197, 2018 03 15.
Article En | MEDLINE | ID: mdl-29410165

Advancements in fetal intervention procedures have led to increases in the number of pregnant women undergoing general anesthesia during the second trimester-a period characterized by extensive proliferation of fetal neural stem cells (NSCs). However, few studies have investigated the effects of mid-gestational sevoflurane exposure on fetal NSC proliferation or postnatal learning and memory function. In the present study, pregnant rats were randomly assigned to a control group (C group), a low sevoflurane concentration group (2%; L group), a high sevoflurane concentration group (3.5%; H group), a high sevoflurane concentration plus lithium chloride group (H + Li group), and a lithium chloride group (Li group) at gestational day 14. Rats received different concentrations of sevoflurane anesthesia for 2 h. The offspring rats were weaned at 28 days for behavioral testing (i.e., Morris Water Maze [MWM]), and fetal brains or postnatal hippocampal tissues were harvested for immunofluorescence staining, real-time PCR, and Western blotting analyses in order to determine the effect of sevoflurane exposure on NSC proliferation and the Wnt/ß-catenin signaling pathway. Our results indicated that maternal exposure to 3.5% sevoflurane (H group) during the mid-gestational period impaired the performance of offspring rats in the MWM test, reduced NSC proliferation, and increased protein levels of fetal glycogen synthase kinase-3 beta (GSK-3ß). Such treatment also decreased levels of ß-catenin protein, CD44 RNA, and Cyclin D1 RNA relative to those observed in the C group. However, these effects were transiently attenuated by treatment with lithium chloride. Conversely, maternal exposure to 2% sevoflurane (L group) did not influence NSC proliferation or the Wnt signaling pathway. Our results suggest that sevoflurane exposure during the second trimester inhibits fetal NSC proliferation via the Wnt/ß-catenin pathway and impairs postnatal learning and memory function in a dose-dependent manner.


Anesthetics, Inhalation/toxicity , Fetus/drug effects , Learning Disabilities/chemically induced , Memory Disorders/chemically induced , Methyl Ethers/toxicity , Neural Stem Cells/drug effects , Prenatal Exposure Delayed Effects , Anesthetics, Inhalation/administration & dosage , Animals , Cell Division/drug effects , Cyclin D1/biosynthesis , Dose-Response Relationship, Drug , Female , Gestational Age , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/metabolism , Hyaluronan Receptors/biosynthesis , Lithium Chloride/therapeutic use , Maze Learning/drug effects , Methyl Ethers/administration & dosage , Methyl Ethers/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Pregnancy , Rats , Rats, Sprague-Dawley , Sevoflurane , Spatial Behavior/drug effects , Wnt Signaling Pathway/drug effects
2.
Mol Med Rep ; 16(5): 7829-7834, 2017 Nov.
Article En | MEDLINE | ID: mdl-28944861

General anesthesia in patients with or at risk for neuronal injury remains challenging due to the neurotoxic effects of volatile anesthetics. One inhalation anesthetic, sevoflurane, induces neuronal damage, including neuroapoptosis, and learning and memory impairment. Panax Notoginseng Saponins (PNS) is the active ingredient of Sanqui and has been reported to exert neuroprotective effects. In the current study, the protective effect of PNS on sevoflurane­induced nerve cell injury was explored. Cell proliferation was significantly reduced in a dose­dependent manner following stimulation with sevoflurane. Furthermore, cell apoptosis and the protein expression of caspase­3, caspase­9 and Bax were significantly increased, while the expression of Bcl­2 was decreased in the sevoflurane group compared with normal control. Furthermore, the protein level of Bace­1, APP and Aß were elevated in the sevoflurane group compared with the control group. By contrast, PNS treatment significantly reduced the neurotoxicity induced by sevoflurane. Additionally, sevoflurane reduced activation of the AKT signaling pathway, which was activated by PNS treatment. In conclusion, the results suggested that PNS attenuates sevoflurane­induced neurotoxicity through by stimulating cell proliferation and inhibiting cell apoptosis. These effects were mediated, at least in part, by activating the AKT signaling pathway.


Anesthetics, Inhalation/toxicity , Methyl Ethers/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/pharmacology , Panax notoginseng/chemistry , Saponins/pharmacology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Proliferation , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression Regulation/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Methyl Ethers/toxicity , Mice , Mice, Inbred BALB C , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/isolation & purification , Plant Extracts/chemistry , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Saponins/isolation & purification , Sevoflurane , Signal Transduction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
3.
Neurotox Res ; 31(2): 218-229, 2017 02.
Article En | MEDLINE | ID: mdl-27682474

Neonatal exposure to volatile anesthetics causes apoptotic neurodegeneration in the developing brain, possibly leading to neurocognitive deficits in adulthood. Endoplasmic reticulum (ER) stress might be associated with sevoflurane (sevo)-induced neuroapoptosis. However, the signaling pathway regulating sevo-induced neuroapoptosis is not understood. We investigated the effects of neonatal sevo exposure on ER signaling pathway activation. Seven-day-old mouse pups were divided into control (C) and sevo (S; 3 % sevo exposure, 6 h) groups. ER stress marker [protein kinase RNA-like ER kinase (PERK), eukaryotic translation initiation factor 2α (eIF2α), activating transcription factor 4 (ATF4), CHOP, and caspase-12] levels were determined by western blotting. To understand the role of eIF2α in sevo-induced ER stress and caspase-3 activation, pups were pretreated with an eIF2α dephosphorylation inhibitor, salubrinal, and a potent and selective inhibitor of PERK, GSK2656157, before sevo exposure, and the effects on ER stress signaling and neuroapoptosis were examined. We investigated whether neonatal exposure to sevo increased ß-site APP-cleaving enzyme 1 (BACE-1) expression. Neonatal sevo exposure elevated caspase-3 activation. ER stress signaling was activated, along with increased PERK and eIF2α phosphorylation, and upregulation of proapoptotic proteins (ATF4 and CHOP) in the cerebral cortex of the developing brain. Pretreatment with salubrinal augmented sevo-induced eIF2α phosphorylation, which inhibited ER stress-mediated ATF4 and caspase-3 activation. Inhibition of PERK phosphorylation due to GSK2656157 pretreatment reduced the sevo-induced increase in eIF2α phosphorylation. Sevo increased BACE-1 expression, which was attenuated by GSK2656157 and salubrinal pretreatment. Our data suggested that neonatal sevo exposure-induced neuroapoptosis is mediated via the PERK-eIF2α-ATF4-CHOP axis of the ER stress signaling pathway. Modulation of eIF2α phosphorylation may play a key role in sevo-induced neurotoxicity in the developing brain.


Amyloid Precursor Protein Secretases/biosynthesis , Apoptosis/drug effects , Aspartic Acid Endopeptidases/biosynthesis , Cerebral Cortex/metabolism , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Methyl Ethers/adverse effects , Activating Transcription Factor 4/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Caspase 3/metabolism , Cinnamates/pharmacology , Indoles/pharmacology , Methyl Ethers/antagonists & inhibitors , Mice , Phosphorylation/drug effects , Sevoflurane , Signal Transduction/drug effects , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transcription Factor CHOP/metabolism
4.
BMC Anesthesiol ; 15: 25, 2015.
Article En | MEDLINE | ID: mdl-25774088

BACKGROUND: At present, sevoflurane inhalation anesthesia used on infants is well-known. But long-time exposure to inhalation anesthetic could cause neurologic disorder, especially nerve degeneration in infant and developing brain. The central nervous system degeneration of infants could affect the memory and cognitive function. γ-Aminobutyric acid (GABA) is a known inhibitory neurotransmitter in central nervous system. Inhalation anesthetic sevoflurane may activate GABAA receptor to inhibit central nervous system, leading to apoptosis of neural degeneration, cognitive dysfunction in the critical period of brain development. METHODS: Neural stem cells were derived from Wistar embryos, cultured in vitro. Third generation of neural stem cells were randomly divided into four groups according to cultured suspension: Sevoflurane group (Group S), GABAA receptor antagonists, Bicuculline group (Group B), Sevoflurane + GABAA receptor antagonists, Bicuculline group (Group S + B), dimethyl sulphoxide (DMSO) group (Group D). Group B and Group D did not receive sevoflurane preconditioning. Group S and Group S + B were pretreated with 1 minimum alveolar concentration (MAC) sevoflurane for 0 h, 3 h, 6 h, and 12 h. Group S + B and Group B were pretreated with bicuculline (10 uM). Group D was treated with DMSO (10 uL/mL). After treatments above, all groups were cultured for 48 h. Then we measured the cells viability by Cell Counting Kit (CCK-8) assay, cytotoxicity by Lactate Dehydrogenase (LDH) assay, apoptosis ratio with Annexin V/propidium iodide (PI) staining by flow cytometry, and the expression of GABAAR, anti-apoptotic protein Bcl-2, pro-apoptotic protein Bax and Caspase-3 by western blotting. RESULTS: After exposing to sevoflurane for 0 h, 3 h, 6 h, and 12 h with 1MAC, we found that cell viability obviously decreased and cytotoxicity increased in time-dependent way. And Annexin V/PI staining indicated increased apoptosis ratio by flow cytometry. The protein level of GABAA receptor, pro-apoptotic protein Bax and apoptosis protein Caspase-3 increased; while anti-apoptotic protein Bcl-2 decreased. And bicuculline could reverse all detrimental results caused by sevoflurane. CONCLUSION: Sevoflurane can inhibit the central nervous system by activating GABAA, resulting in apoptosis of neural stem cells, thus leading to the NSCs degeneration.


Apoptosis/drug effects , Methyl Ethers/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Animals , Bicuculline/pharmacology , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Methyl Ethers/antagonists & inhibitors , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Receptors, GABA-A/metabolism , Sevoflurane , bcl-2-Associated X Protein/metabolism
5.
Br J Anaesth ; 113(6): 1032-8, 2014 Dec.
Article En | MEDLINE | ID: mdl-25086587

BACKGROUND: Our previous reports demonstrated that genetic deletion of µ-opioid receptor has no influence on the anaesthetic and antinociceptive effects of nitrous oxide (N2O) in mice, and that an antagonist selective for κ-opioid receptor (KOP), but not that selective for δ-opioid receptor, suppresses the antinociceptive effect of N2O. However, it is not known whether genetic deletion of KOP affects the N2O actions. METHODS: We measured the minimum alveolar concentration (MAC) of volatile anaesthetics in the absence and presence of N2O. The antinociceptive action of N2O was tested by an acetic acid-writhing test and a hot-plate test. The number of c-Fos-immunopositive cells in sections from the lumbar spinal cord was counted to test whether the descending inhibitory system participates in the pharmacological action of N2O. The hypnotic action of N2O was assessed by measuring the N2O-induced decrease in the EC50 for loss of the righting reflex (EC50-LORR) of sevoflurane. RESULTS: Sevoflurane MAC was not significantly reduced by N2O and its antinociceptive action was almost completely abolished in KOP-knockout (KO) mice. The N2O-induced increase in c-Fos-immunopositive cells in laminae III-IV of the lumbar spinal cord was significant in wild-type (WT), but not in KOP-KO mice. In contrast, sevoflurane EC50-LORR was similarly reduced by N2O in WT and KOP-KO mice. CONCLUSIONS: Our study suggests that N2O demonstrates its antinociceptive action and reduces sevoflurane MAC in mice through KOP activation, whereas its hypnotic potency is not dependent on KOP activation.


Analgesics, Non-Narcotic/pharmacology , Nitrous Oxide/pharmacology , Receptors, Opioid, kappa/drug effects , Anesthetics, Inhalation/pharmacology , Animals , Isoflurane/pharmacology , Lumbar Vertebrae , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Pain/physiopathology , Pain/prevention & control , Pain Measurement/methods , Pain Threshold/drug effects , Pain Threshold/physiology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Opioid, kappa/deficiency , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/physiology , Sevoflurane , Spinal Cord/drug effects , Spinal Cord/metabolism
6.
Paediatr Anaesth ; 24(7): 749-59, 2014 Jul.
Article En | MEDLINE | ID: mdl-24725211

INTRODUCTION: Recent data on newborn animals exposed to anesthetics have raised safety concerns regarding anesthesia practices in young children. Indeed, studies on rodents have demonstrated a widespread increase in brain apoptosis shortly after exposure to sevoflurane, followed by long-term neurologic impairment. In this context, we aimed to evaluate the protective effect of rh-EPO, a potent neuroprotective agent, in rat pups exposed to sevoflurane. MATERIAL AND METHODS: At postnatal day 7, 75 rat pups were allocated into three groups: SEVO + EPO (n = 27) exposed to sevoflurane 2 vol% (0.5 MAC) for 6 h in an air/O2 mixture (60/40) + 5000 UI.kg(-1) rh-EPO IP; SEVO (n = 27) exposed to sevoflurane + vehicle IP; and CONTROL (n = 21) exposed to the mixture without sevoflurane + vehicle IP. Three days after anesthesia (D10), apoptosis was quantified on brain extract with TUNEL method and caspase 3. NGF and BDNF expression was determined by Western blotting. Rats reaching adulthood were evaluated in terms of exploration capacities (object exploration duration) together with spatial and object learning (water maze and novel object test). RESULTS: Sevoflurane exposure impaired normal behavior in adult rats by reducing the exploratory capacities during the novel object test and impaired both spatial and object learning capacities in adult rats (water maze, ratio time to find platform 3rd trial/1st trial: 1.1 ± 0.2 vs 0.4 ± 0.1; n = 9, SEVO vs CONTROL; P = 0.01). Rh-EPO reduced sevoflurane-induced behavior and learning abnormalities in adult rats (water maze, ratio time to find platform 3rd trial/1st trial: 0.3 ± 0.1 vs 1.1 ± 0.2; n = 9, SEVO + EPO vs SEVO; P = 0.01). Three days after anesthesia, rh-EPO prevented sevoflurane-induced brain apoptosis (5 ± 3 vs 35 ± 6 apoptotic cells·mm(-2) ; n = 6, SEVO + EPO vs SEVO; P = 0.01) and elevation of caspase three level and significantly increased the brain expression of BDNF and NGF (n = 6, SEVO + EPO vs SEVO; P = 0.01). CONCLUSION: Six hours of sevoflurane anesthesia in newborn rats induces significant long-term cognitive impairment. A single administration of rh-EPO immediately after postnatal exposure to sevoflurane reduces both early activation of apoptotic phenomenon and late onset of neurologic disorders.


Anesthetics, Inhalation/toxicity , Erythropoietin/therapeutic use , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/toxicity , Nervous System Diseases/chemically induced , Nervous System Diseases/prevention & control , Animals , Animals, Newborn , Apoptosis/drug effects , Blood Gas Analysis , Brain/pathology , Cognition Disorders/chemically induced , Cognition Disorders/prevention & control , Epoetin Alfa , Maze Learning/drug effects , Memory/drug effects , Nervous System Diseases/psychology , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Recombinant Proteins/therapeutic use , Sevoflurane
7.
Br J Anaesth ; 110 Suppl 1: i73-81, 2013 Jun.
Article En | MEDLINE | ID: mdl-23535829

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.


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
8.
Neuroscience ; 205: 167-77, 2012 Mar 15.
Article En | MEDLINE | ID: mdl-22244976

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.


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
9.
J Cardiovasc Pharmacol Ther ; 16(1): 72-8, 2011 Mar.
Article En | MEDLINE | ID: mdl-20938040

PURPOSE: It is known that selective cyclooxygenase 2(COX-2) inhibitors increase mortality in patients with previous myocardial infarction, and it has been suggested that COX-2 plays an important role in cardioprotection against ischemia. The current study was carried out to determine whether COX-2 is involved in the mechanisms of sevoflurane- and olprinone-induced early-phase preconditioning (E-PreC) and postconditioning (PostC) in rat hearts. METHODS: Male Sprague-Dawley rats were anesthetized with sodium pentobarbital. After opening the chest, all rats underwent 30-minute occlusion of left anterior descending coronary artery followed by 2-hour reperfusion, and the infarct size was measured after the reperfusion. The rats were randomly assigned to groups with pre- and postischemic exposure to sevoflurane and administration of olprinone with or without a selective COX-2 inhibitor, NS-398. RESULTS: The infarct size in the control group was 42% ± 6% of the area at risk. Infarct size was significantly reduced by pre- and postischemic administration of sevoflurane (16% ± 7% and 17% ± 6%, respectively), as well as by olprinone (14% ± 4% and 15% ± 10%, respectively). NS-398 prevented the protective effects of both pre- and postischemic exposure to sevoflurane (35% ± 8% and 42% ± 10%, respectively), whereas the protective effect of both pre- and postischemic administration of olprinone was not influenced by NS-398 (12% ± 5% and 19% ± 7%, respectively). CONCLUSIONS: Cyclooxygenase 2 could be a critical mediator of sevoflurane-induced but not olprinone-induced E-PreC or PostC in rat hearts.


Cardiotonic Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Methyl Ethers/pharmacology , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Animals , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/therapeutic use , Coronary Vessels/drug effects , Heart/drug effects , Imidazoles/pharmacology , Imidazoles/therapeutic use , Ischemic Postconditioning/methods , Ischemic Preconditioning, Myocardial/methods , Male , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/therapeutic use , Myocardial Infarction/physiopathology , Myocardium/metabolism , Nitrobenzenes/pharmacology , Pyridones/pharmacology , Pyridones/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Sevoflurane , Sulfonamides/pharmacology
10.
J Zhejiang Univ Sci B ; 11(9): 661-72, 2010 Sep.
Article En | MEDLINE | ID: mdl-20803770

Sevoflurane postconditioning reduces myocardial infarct size. The objective of this study was to examine the role of the phosphatidylinositol-3-kinase (PI3K)/Akt pathway in anesthetic postconditioning and to determine whether PI3K/Akt signaling modulates the expression of pro- and antiapoptotic proteins in sevoflurane postconditioning. Isolated and perfused rat hearts were prepared first, and then randomly assigned to the following groups: Sham-operation (Sham), ischemia/reperfusion (Con), sevoflurane postconditioning (SPC), Sham plus 100 nmol/L wortmannin (Sham+Wort), Con+Wort, SPC+Wort, and Con+dimethylsulphoxide (DMSO). Sevoflurane postconditioning was induced by administration of sevoflurane (2.5%, v/v) for 10 min from the onset of reperfusion. Left ventricular developed pressure (LVDP), left ventricular end-diastolic pressure (LVEDP), maximum increase in rate of LVDP (+dP/dt), maximum decrease in rate of LVDP (-dP/dt), heart rate (HR), and coronary flow (CF) were measured at baseline, R30 min (30 min of reperfusion), R60 min, R90 min, and R120 min. Creatine kinase (CK) and lactate dehydrogenase (LDH) were measured after 5 min and 10 min reperfusion. Infarct size was determined by triphenyltetrazolium chloride staining at the end of reperfusion. Total Akt and phosphorylated Akt (phospho-Akt), Bax, Bcl-2, Bad, and phospho-Bad were determined by Western blot analysis. Analysis of variance (ANOVA) and Student-Newman-Keuls' test were used to investigate the significance of differences between groups. The LVDP, + or - dP/dt, and CF were higher and LVEDP was lower in the SPC group than in the Con group at all points of reperfusion (P<0.05). The SPC group had significantly reduced CK and LDH release and decreased infarct size compared with the Con group [(22.9 + or - 8)% vs. (42.4 + or - 9.4)%, respectively; P<0.05]. The SPC group also had increased the expression of phospho-Akt, Bcl-2, and phospho-Bad, and decreased the expression of Bax. Wortmannin abolished the cardioprotection of sevoflurane postconditioning. Sevoflurane postconditioning may protect the isolated rat heart. Activation of PI3K and modulation of the expression of pro- and antiapoptotic proteins may play an important role in sevoflurane-induced myocardial protection.


Anesthetics, Inhalation/pharmacology , Apoptosis/physiology , Ischemic Postconditioning/methods , Methyl Ethers/pharmacology , Myocardial Reperfusion Injury/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Androstadienes/pharmacology , Anesthetics, Inhalation/antagonists & inhibitors , Anesthetics, Inhalation/therapeutic use , Animals , Apoptosis/drug effects , Blood Pressure/physiology , Blotting, Western , Creatine Kinase/analysis , Heart Rate/physiology , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Ischemic Postconditioning/standards , L-Lactate Dehydrogenase/analysis , Male , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/therapeutic use , Myocardial Reperfusion Injury/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Sevoflurane , Signal Transduction/physiology , Ventricular Pressure/physiology , Wortmannin
11.
Anesthesiology ; 112(3): 567-75, 2010 Mar.
Article En | MEDLINE | ID: mdl-20124973

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.


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
12.
Appl Microbiol Biotechnol ; 77(6): 1359-65, 2008 Jan.
Article En | MEDLINE | ID: mdl-18043916

The filamentous fungus Graphium sp. (ATCC 58400) co-metabolically oxidizes the gasoline oxygenate methyl tertiary butyl ether (MTBE) after growth on gaseous n-alkanes. In this study, the enzymology and regulation of MTBE oxidation by propane-grown mycelia of Graphium sp. were further investigated and defined. The trends observed during MTBE oxidation closely resembled those described for propane-grown cells of the bacterium Mycobacterium vaccae JOB5. Propane-grown mycelia initially oxidized the majority ( approximately 95%) of MTBE to tertiary butyl formate (TBF), and this ester was biotically hydrolyzed to tertiary butyl alcohol (TBA). However, unlike M. vaccae JOB5, our results collectively suggest that propane-grown mycelia only have a limited capacity to degrade TBA. None of the products of MTBE exerted a physiologically relevant regulatory effect on the rate of MTBE or propane oxidation, and no significant effect of TBA was observed on the rate of TBF hydrolysis. Together, these results suggest that the regulatory effects of MTBE oxidation intermediates proposed for MTBE-degrading organisms such as Mycobacterium austroafricanum are not universally relevant mechanisms for MTBE-degrading organisms. The results of this study are discussed in terms of their impact on our understanding of the diversity of aerobic MTBE-degrading organisms and pathways and enzymes involved in these processes.


Ascomycota/metabolism , Methyl Ethers/antagonists & inhibitors , Methyl Ethers/metabolism , Biodegradation, Environmental , Mycelium/metabolism , Oxidation-Reduction , Propane/metabolism , tert-Butyl Alcohol/metabolism
13.
Acta Anaesthesiol Scand ; 52(1): 111-6, 2008 Jan.
Article En | MEDLINE | ID: mdl-17996001

BACKGROUND: Post-ischemic administration of volatile anesthetics activates ischemia-reperfusion injury salvage process and decreases myocardial damage. However, the mechanisms underlying anesthetic post-conditioning and effects of lidocaine on it remain unclear. Here we report the cardioprotection of sevoflurane-induced post-conditioning and the effects of lidocaine on it. METHODS: Isolated perfused rat hearts were exposed to 40 min of ischemia followed by 1 h of reperfusion. Volatile anesthetic post-conditioning was induced by 15 min of 3 vol% sevourane (1.5 minimum alveolar concentration) administered at the onset of reperfusion. In some experiments, lidocaine was coadministered with sevoflurane in different concentrations (2, 10 and 20 microg/ml). Infarct size was determined by dividing the total necrotic area of the left ventricle by the total left ventricular slice area (percent necrotic area). RESULTS: Sevoflurane-induced post-conditioning signicantly improved post-ischemia functional recovery and decreased infarct size (47.3+/-5.6% in unprotected hearts vs. 18.6+/-3.1% in anesthetic post-conditioning, P<0.05). Sevourane-mediated cardioprotection was abolished by 20 microg/ml lidocaine. CONCLUSIONS: Sevourane-induced post-conditioning effectively protected myocardium against reperfusion damage and its cytoprotection was reversed by 20 microg/ml lidocaine.


Cardiotonic Agents/antagonists & inhibitors , Heart/drug effects , Lidocaine/toxicity , Methyl Ethers/antagonists & inhibitors , Myocardial Reperfusion Injury/prevention & control , Animals , Biomarkers , Cardiotonic Agents/pharmacology , Heart Ventricles/pathology , In Vitro Techniques , KATP Channels/drug effects , KATP Channels/physiology , L-Lactate Dehydrogenase/analysis , Lidocaine/pharmacology , Male , Methyl Ethers/pharmacology , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Necrosis , Rats , Rats, Sprague-Dawley , Sevoflurane
14.
Anesthesiology ; 107(4): 616-20, 2007 Oct.
Article En | MEDLINE | ID: mdl-17893458

BACKGROUND: The authors hypothesized that intramuscular sevoflurane injection allows diagnostic differentiation between malignant hyperthermia-susceptible (MHS) and -nonsusceptible (MHN) pigs by measurement of intramuscular lactate and carbon dioxide partial pressure (PCO2), and that dantrolene reduces the sevoflurane-induced PCO2 increase. METHODS: With approval of the local animal care committee, microdialysis probes with attached microtubing for sevoflurane injection were placed in the adductor muscles of nine MHS and six MHN pigs, and PCO2 probes with microtubing were positioned in the triceps muscle of eight MHS and six MHN pigs. After equilibration, sevoflurane boluses at different concentrations and a sevoflurane-dantrolene bolus were injected synchronously. Lactate, pyruvate, and glucose as well as PCO2 were measured spectrophotometrically, and the rate of PCO2 increase was calculated. RESULTS: Intramuscular sevoflurane injection increased local lactate and PCO2 dose dependently, and significantly higher in MHS than in MHN pigs. Measurement of the rate of PCO2 increase allowed a distinct differentiation between single MHS and MHN pigs. No significant increase in PCO2 was found with sevoflurane and dantrolene. CONCLUSIONS: Local sevoflurane induces a hypermetabolic reaction measured by PCO2 and lactate increases. The reduced PCO2 increase in MHS after sevoflurane and dantrolene injection is likely to be a result of the sevoflurane-mediated calcium release and its antagonism by dantrolene. Sevoflurane may be useful for a less invasive diagnostic test for malignant hyperthermia in humans.


Anesthetics, Inhalation , Hypertension, Malignant/diagnosis , Methyl Ethers , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/antagonists & inhibitors , Animals , Blood Glucose/metabolism , Carbon Dioxide/blood , Dantrolene/pharmacology , Dose-Response Relationship, Drug , Hypertension, Malignant/physiopathology , Injections, Intramuscular , Lactic Acid/blood , Methyl Ethers/administration & dosage , Methyl Ethers/antagonists & inhibitors , Microdialysis , Muscle Relaxants, Central/pharmacology , Pyruvic Acid/blood , Sevoflurane , Swine
15.
J Anesth ; 21(3): 361-6, 2007.
Article En | MEDLINE | ID: mdl-17680189

PURPOSE: For anesthetic management of cesarean sections, regardless of the use of regional or general anesthesia, it is crucial to achieve sufficient uterine contraction immediately following the delivery of an infant in order to reduce excessive bleeding. No previous study has investigated the ability of alprostadil, a synthesized prostaglandin, to inhibit myometrial relaxation induced by volatile anesthetics. The aim of the present study was to investigate the inhibitory effects of alprostadil on sevoflurane-induced myometrial relaxation using myometrial strips isolated from pregnant rats. METHODS: Myometrial strips were isolated from Sprague-Dawley rats (300-400 g) in the late stage of gestation (19-21 days). The time course of changes in spontaneous myometrium contraction was studied in the presence and absence of sevoflurane. Additionally, alprostadil was titrated at three different concentrations during continuous introduction of sevoflurane 2%, and myometrium contraction was studied. As an index of contraction, the area under the contraction curve was used, and data were analyzed by repeated measure one-way analysis of variance. RESULTS: We have shown a significant decrease in myometrium contraction as a result of the use of sevoflurane (2%). Additionally, alprostadil has been shown to inhibit myometrial relaxation induced by sevoflurane in a dose-dependent manner. The areas under the contraction curve were 87%, 87%, 129%, and 172% of the baseline value for the control and at low, medium, and high concentrations of alprostadil, respectively. CONCLUSION: The ability of alprostadil to inhibit myometrial relaxation induced by sevoflurane suggests that the use of alprostadil during general anesthesia for cesarean section may be advantageous for the reduction of postpartum bleeding.


Alprostadil/pharmacology , Anesthetics, Inhalation/antagonists & inhibitors , Methyl Ethers/antagonists & inhibitors , Muscle Relaxation/drug effects , Myometrium/drug effects , Vasodilator Agents/pharmacology , Animals , Area Under Curve , Dose-Response Relationship, Drug , Female , Muscle Relaxation/physiology , Myometrium/physiology , Polymerase Chain Reaction , Pregnancy , Rats , Rats, Sprague-Dawley , Sevoflurane , Uterine Contraction/drug effects
16.
Br J Anaesth ; 91(4): 583-6, 2003 Oct.
Article En | MEDLINE | ID: mdl-14504163

BACKGROUND: Physostigmine, a centrally acting anticholinesterase, antagonizes the hypnotic effect of propofol, as shown by the return of consciousness (response to commands) or wakefulness (spontaneous eye-opening without response to commands) and by recovery of auditory evoked potentials (40 Hz auditory steady-state response (ASSR)) and the bispectral index (BIS). We measured the effects of physostigmine on the hypnotic effect of inhaled volatile anaesthetics, using sevoflurane as the representative agent. METHODS: Eight healthy volunteers received sevoflurane adjusted to produce loss of consciousness. Physostigmine (plus glycopyrrolate) was given while the end-tidal concentration of sevoflurane was kept constant. RESULTS: Loss of consciousness was accompanied by a significant (P<0.02) decrease in ASSR amplitude (to 21% of awake value) and BIS (to 70% of awake value). Five subjects had return of consciousness or wakefulness after physostigmine. The others showed no behavioural change. Physostigmine caused a significant increase of the mean ASSR amplitude from 0.11 (SD 0.04) to 0.17 (0.06) microV (P<0.05). The BIS also increased, from 66 (12) to 74 (12), but the difference was not significant. CONCLUSIONS: Physostigmine can antagonize, at least partially, the hypnotic effect of sevoflurane and changes in arousal after physostigmine are shown by ASSR measurements. However, the antagonism is not as clear or reliable as with propofol.


Anesthesia, Inhalation/methods , Anesthetics, Inhalation/antagonists & inhibitors , Cholinesterase Inhibitors/pharmacology , Evoked Potentials, Auditory/drug effects , Methyl Ethers/antagonists & inhibitors , Physostigmine/pharmacology , Adolescent , Adult , Humans , Sevoflurane , Wakefulness/drug effects
17.
Anesthesiology ; 97(3): 725-31, 2002 Sep.
Article En | MEDLINE | ID: mdl-12218541

BACKGROUND: Sevoflurane enhances coronary collateral blood flow independent of adenosine triphosphate-regulated potassium channels. The authors tested the hypothesis that this volatile anesthetic increases coronary collateral blood flow by either opening calcium-activated potassium channels or by directly stimulating nitric oxide synthesis in the canine coronary collateral circulation. METHODS: Twelve weeks after left anterior descending coronary artery ameroid constrictor implantation, barbiturate-anesthetized dogs (n = 22) were instrumented for measurement of hemodynamics and retrograde coronary flow. Dogs received sevoflurane ([0.5 and 1.0 minimum alveolar concentration [MAC]) during intracoronary infusions of drug vehicle (0.9% saline), the calcium-activated potassium channel antagonist iberiotoxin (13 microg/min), or the nitric oxide synthase inhibitor -nitro-l-arginine methyl ester (l-NAME, 300 microg/min). Retrograde coronary collateral blood flow was measured under baseline conditions, during and after administration of sevoflurane, and during intracoronary infusion of bradykinin. Data are mean +/- SEM. RESULTS: Sevoflurane increased (* < 0.05) retrograde coronary collateral blood flow (from 65 +/- 11 during control to 67 +/- 12* and 71 +/- 12* ml/min during 0.5 and 1.0 MAC, respectively). Iberiotoxin but not l-NAME attenuated these sevoflurane-induced increases in retrograde flow (6 +/- 1*, 7 +/- 2*, and 3 +/- 2 ml/min during vehicle, l-NAME, and iberiotoxin, respectively). After discontinuation of sevoflurane, retrograde flow returned to baseline values in each group. Bradykinin increased retrograde flow in vehicle- (63 +/- 12 to 69 +/- 12* ml/min) but not in iberiotoxin- (61 +/- 7 to 62 +/- 5 ml/min) or l-NAME-treated dogs (64 +/- 11 to 63 +/- 10 ml/min). CONCLUSIONS: The results demonstrate that sevoflurane increases coronary collateral blood flow, in part, through activation of calcium-activated potassium channels. This action occurs independent of nitric oxide synthesis.


Anesthetics, Inhalation/pharmacology , Collateral Circulation/drug effects , Coronary Circulation/drug effects , Methyl Ethers/pharmacology , Potassium Channels, Calcium-Activated/physiology , Animals , Bradykinin/pharmacology , Constriction, Pathologic/physiopathology , Dogs , Hemodynamics/drug effects , In Vitro Techniques , Methyl Ethers/antagonists & inhibitors , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Potassium Channels, Calcium-Activated/drug effects , Sevoflurane
18.
Anesth Analg ; 94(3): 569-72; table of contents, 2002 Mar.
Article En | MEDLINE | ID: mdl-11867377

UNLABELLED: In this double-blinded study, we investigated the effect of physostigmine on sevoflurane anesthesia and recovery. Forty female patients scheduled for breast biopsy were randomly assigned to receive either physostigmine 2 mg IV or an equal volume of normal saline after skin closure. Anesthesia was induced and maintained with sevoflurane in oxygen. After skin closure, a steady state of 0.6% inspired and end-tidal sevoflurane concentration was obtained, heart rate, blood pressure, and Bispectral index (BIS) baseline values were recorded, and physostigmine or normal saline was administered. Hemodynamics and BIS values were also recorded 5, 8, and 10 min after treatments. Anesthesia was discontinued, and orientation, sedation, sitting ability, and "picking up matches" scores were recorded immediately after extubation and 15 and 30 min later. No differences were found between the two groups in BIS (69, 70, 70, and 71 in the Physostigmine group versus 70, 74, 75, and 76 in the Control group) or blood pressure. Only heart rate was increased 8 min after physostigmine (P < 0.05 versus the control). Scores assessing early recovery were similar in the two groups at all time points. We conclude that physostigmine does not change BIS or enhance recovery after sevoflurane anesthesia. IMPLICATIONS: This double-blinded, randomized study investigated the impact of physostigmine of BIS values during 0.6% sevoflurane anesthesia as well as in the postoperative recovery, when sevoflurane is administered as a sole anesthetic. Physostigmine has no effect on BIS values or on the tests assessing recovery.


Anesthesia , Anesthetics, Inhalation/antagonists & inhibitors , Cholinesterase Inhibitors/pharmacology , Electroencephalography/drug effects , Methyl Ethers/antagonists & inhibitors , Physostigmine/pharmacology , Adult , Double-Blind Method , Humans , Prospective Studies , Sevoflurane
19.
Appl Environ Microbiol ; 67(12): 5601-7, 2001 Dec.
Article En | MEDLINE | ID: mdl-11722912

Biodegradation of methyl tert-butyl ether (MTBE) by the hydrogen-oxidizing bacterium Hydrogenophaga flava ENV735 was evaluated. ENV735 grew slowly on MTBE or tert-butyl alcohol (TBA) as sole sources of carbon and energy, but growth on these substrates was greatly enhanced by the addition of a small amount of yeast extract. The addition of H(2) did not enhance or diminish MTBE degradation by the strain, and MTBE was only poorly degraded or not degraded by type strains of Hydrogenophaga or hydrogen-oxidizing enrichment cultures, respectively. MTBE degradation activity was constitutively expressed in ENV735 and was not greatly affected by formaldehyde, carbon monoxide, allyl thiourea, or acetylene. MTBE degradation was inhibited by 1-amino benzotriazole and butadiene monoepoxide. TBA degradation was inducible by TBA and was inhibited by formaldehyde at concentrations of >0.24 mM and by acetylene but not by the other inhibitors tested. These results demonstrate that separate, independently regulated genes encode MTBE and TBA metabolism in ENV735.


Betaproteobacteria/growth & development , Betaproteobacteria/metabolism , Methyl Ethers/metabolism , Betaproteobacteria/classification , Betaproteobacteria/isolation & purification , Biodegradation, Environmental , Culture Media , Hydrogen , Methyl Ethers/antagonists & inhibitors , Oxidation-Reduction , tert-Butyl Alcohol/metabolism
20.
Neoplasma ; 40(3): 167-72, 1993.
Article En | MEDLINE | ID: mdl-8394517

The inhibition of intercellular gap junctional communication (IGJC) by alkyl ethers (ethylene glycol, monomethyl ether, polyethylene glycol 1,000 and polyethylene glycol 6000) was examined using V79 Chinese hamster cells in vitro. Ethylene glycol and monomethyl ether inhibited IGJC very strongly, whilst the other agents inhibited IGJC only insignificantly. When the cells were treated with the combination of two agents, ethylene glycol and monomethyl ether, a significant increase in the inhibition of IGJC occurred. This was probably the result of potentiation rather than an addition effect. The effect of ethylene glycol was antagonized by dibutyryl cyclic adenosine monophosphate (DbcAMP). This effect was most intensive when the cells were treated with both agents at the same time and, in other experimental combinations, the effect was lower but also significant. Caffeine did not influence IGJC either in combination with DbcAMP or by itself.


Bucladesine/toxicity , Cell Communication/drug effects , Ethylene Glycols/toxicity , Intercellular Junctions/drug effects , Methyl Ethers/toxicity , Animals , Bucladesine/antagonists & inhibitors , Bucladesine/pharmacology , Caffeine/pharmacology , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Drug Interactions , Ethylene Glycol , Ethylene Glycols/antagonists & inhibitors , Intercellular Junctions/physiology , Methyl Ethers/antagonists & inhibitors , Polyethylene Glycols/toxicity , Time Factors
...