Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Physiol Pharmacol ; 65(4): 531-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25179085

ABSTRACT

Orexin A (OA) is an endogenous peptide regulating awakefulness, known to reduce anaesthesia in animals, but on cellular level its mechanisms to reverse anaesthetics are unknown. Primary cortical cell cultures from newborn rat brains are used and live cell light microscopy is performed to measure 1) neurite retraction after propofol, thiopental, barbituric acid and ketamine exposure and 2) the effect of OA application either before or after anaesthetics. Cytoskeletal reorganization is evaluated with fluorescence microscopy, protein changes are detected with Western blots and mass spectrometry is used to identify proteins after treatment with anaesthetics and/or OA. Adult rats are anaesthesized with propofol, and the cytoskeletal morphology is studied. Orexin A reverses and inhibits neurite retraction and actin ring formation induced by propofol and thiopental. No effect on retraction or actin rings was seen for ketamine (not active on gamma-aminobutiric acid A (GABA(A)) receptors), the non-anaesthetic barbituric acid, OA or solvents used. OA increases the tyrosine phosphorylation of a 50 kDa protein, identified as vimentin. Propofol induces an immediate granular appearance of vimentin, which OAreverses to a smooth distribution. Cytoskeletal morphology changes are also induced by propofol in vivo. All OA effects are blocked with an orexin receptor1 (OX1) antagonist. We conclude that OA reverses the GABAA receptor mediated cellular effects of both propofol and thiopental in rat brain cells. The morphologic changes of actin and vimentin caused by propofol and thiopental, and the subsequent reversal by OA, deepens our understanding of the mechanisms of anaesthesia.


Subject(s)
Anesthetics, Intravenous/pharmacology , Hypnotics and Sedatives/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Neurons/drug effects , Neuropeptides/pharmacology , Propofol/pharmacology , Thiopental/pharmacology , Actins/metabolism , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Male , Neurons/metabolism , Orexins , Rats, Sprague-Dawley , Tyrosine/metabolism , Vimentin/metabolism
2.
J Physiol Pharmacol ; 62(1): 119-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21451217

ABSTRACT

Neuronal intracellular transport is performed by motor proteins, which deliver vesicles, organelles and proteins along cytoskeletal tracks inside the neuron. We have previously shown that the anesthetic propofol causes dose- and time-dependent, reversible retraction of neuronal neurites. We hypothesize that propofol alters the vesicular transport of cortical neurons due to this neurite retraction. Primary cultures of co-cultivated rat cortical neurons and glial cells were exposed to either 2 µM propofol, control medium or the lipid vehicle, in time-response experiments. Reversibility was tested by washing propofol off the cells. The role of the GABA(A) receptor (GABA(A)R) was assessed with the GABA(A)R antagonist gabazine. Vesicles were tracked using differential interference contrast video microscopy. Propofol caused a retrograde movement in 83.4±5.2% (mean±S.E.M.) of vesicles, which accelerated over the observed time course (0.025±0.012 µm.s⁻¹). In control medium, vesicles moved predominantly anterograde (84.6±11.1%) with lower velocity (0.011±0.004 µm.s⁻¹). Cells exposed to the lipid vehicle showed the same dynamic characteristics as cells in control medium. The propofol-induced effect on vesicle transport was reversible and blocked by the GABA(A)R antagonist gabazine in low concentration. Our results show that propofol causes a reversible, accelerating vesicle movement toward the neuronal cell body that is mediated via synaptic GABA(A)R. We have previously reported that propofol initiates neurite retraction, and we propose that propofol causes vesicle movement by retrograde flow of cytoplasm from the narrowed neurite.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Neurons/drug effects , Neurons/metabolism , Propofol/pharmacology , Transport Vesicles/drug effects , Animals , Cells, Cultured , Cerebral Cortex/cytology , Molecular Motor Proteins/metabolism , Neurites/metabolism , Neuroglia/metabolism , Neurons/cytology , Pyridazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism
3.
J Physiol Pharmacol ; 59(3): 617-32, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18953102

ABSTRACT

The anaesthetic propofol interacts with the GABA(A) receptor, but its cellular signalling pathways are not fully understood. Propofol causes reorganisation of the actin cytoskeleton into ring structures in neurons. Is this reorganisation a specific effect of propofol as apposed to GABA, and which cellular pathways are involved? We used fluorescence-marked actin in cultured rat neurons to evaluate the percentage of actin rings caused by propofol or GABA in combination with rho, rho kinase (ROK), PI3-kinase or tyrosine kinase inhibitors, with or without the presence of extracellular calcium. Confocal microscopy was performed on propofol-stimulated cells and changes in actin between cellular compartments were studied with Western blot. Propofol (3 microg x ml-1), but not GABA (5 microM), caused transcellular actin ring formation, that was dependent on influx of extracellular calcium and blocked by rho, ROK, PI3-kinase or tyrosine kinase inhibitors. Propofol uses rho/ROK to translocate actin from the cytoskeleton to the membrane and its actin ring formation is dependent on an interaction site close to the GABA site on the GABA(A) receptor. GABA does not cause actin rings, implying that this is a specific effect of propofol.


Subject(s)
Cytoskeleton/drug effects , Hypnotics and Sedatives/pharmacology , Neurons/drug effects , Propofol/pharmacology , Receptors, GABA-A/drug effects , Signal Transduction/drug effects , Actins/chemistry , Actins/metabolism , Animals , Blotting, Western , Cells, Cultured , Enzyme Inhibitors/pharmacology , Indicators and Reagents , Microscopy, Confocal , Microscopy, Fluorescence , Nerve Tissue Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
4.
Br J Anaesth ; 101(3): 374-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18587139

ABSTRACT

BACKGROUND: The mechanism by which anaesthetic agents produce general anaesthesia is not yet fully understood. Retraction of neurites is an important function of individual neurones and neural plexuses during normal and pathological conditions, and it has been shown that such a retraction pathway exists in developing and mature neurones. We hypothesized that propofol decreases neuronal activity by causing retraction of neuronal neurites. METHODS: Primary cultures of rat cortical neurones were exposed in concentration- and time-response experiments to 0.02, 0.2, 2, and 20 microM propofol or lipid vehicle. Neurones were pretreated with the GABA(A) receptor (GABA(A)R) antagonist, bicuculline, the myosin II ATPase activity inhibitor, blebbistatin, and the F-actin stabilizing agent, phalloidin, followed by administration of propofol (20 microM). Changes in neurite retraction were evaluated using time-lapse light microscopy. RESULTS: Propofol caused a concentration- and time-dependent reversible retraction of cultured cortical neurone neurites. Bicuculline, blebbistatin, and phalloidin completely inhibited propofol-induced neurite retraction. Images of retracted neurites were characterized by a retraction bulb and a thin trailing membrane remnant. CONCLUSIONS: Cultured cortical rat neurones retract their neurites after exposure to propofol in a concentration- and time-dependent manner. This retraction is GABA(A)R mediated, reversible, and dependent on actin and myosin II. Furthermore, the concentrations and times to full retraction and recovery correspond to those observed during propofol anaesthesia.


Subject(s)
Anesthetics, Intravenous/pharmacology , Neurites/drug effects , Propofol/pharmacology , Anesthetics, Intravenous/antagonists & inhibitors , Animals , Bicuculline/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , GABA Antagonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neurites/ultrastructure , Phalloidine/pharmacology , Propofol/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
5.
Acta Anaesthesiol Scand ; 47(2): 157-64, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12631044

ABSTRACT

BACKGROUND: Propofol is known to interact with the gamma-aminobutyric acidA (GABA(A)) receptor, however, activating the receptor alone is not sufficient for producing anaesthesia. METHODS: To compare propofol and GABA, their interaction with the GABAA receptor beta subunit and actin were studied in three cellular fractions of cultured rat neurons using Western blot technique. RESULTS: Propofol tyrosine phosphorylated the GABA(A) receptor beta2 (MW 54 and 56 kDa) and beta3 (MW 57 kDa) subtypes. The increase was shown in both the cytoskeleton (beta2(54) and beta2(56) subtypes) and the cell membrane (beta2(54) and beta3 subtypes). Concurrently the 56 kDa beta2 subtype was reduced in the cytosol. Propofol, but not GABA, also tyrosine phosphorylated actin in the cell membrane and cytoskeletal fraction. Without extracellular calcium available, the amount of actin decreased in the cytoskeleton, but tyrosine phosphorylation was unchanged. GABA caused increased tyrosine phosphorylation of beta2(56) and beta3 subtypes in the membrane and both beta2 subtypes in the cytoskeleton but no cytosolic tyrosine phosphorylation. CONCLUSION: The difference between propofol and GABA at the GABA(A) receptor was shown to take place in the membrane, where the beta2(54) was increased by propofol and instead the beta2(56) subtype was increased by GABA. Only propofol also tyrosine phosphorylated actin in the cell membrane and cytoskeletal fraction. This interaction between the GABAA receptor and actin might explain the difference between anaesthesia and physiological neuronal inhibition.


Subject(s)
Actins/drug effects , Anesthesia , Anesthetics, Intravenous/pharmacology , Propofol/pharmacology , Receptors, GABA-A/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sleep/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , Blotting, Western , Cell Membrane/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Cytosol/drug effects , Indicators and Reagents , Neurons/drug effects , Neurons/ultrastructure , Phosphorylation , Precipitin Tests , Protein Subunits , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/ultrastructure , Tyrosine/metabolism
6.
Acta Physiol Scand ; 175(3): 227-35, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12100362

ABSTRACT

Propofol, an intravenous anaesthetic, has been shown to interact with the beta-subunit of the gamma-amino butyric acid(A) (GABA(A)) receptor and also to cause changes in [Ca2+]i. The GABA(A) receptor, a suggested target for anaesthetics, is known to be regulated by kinases. We have investigated if tyrosine kinase is involved in the intracellular signal system used by propofol to cause anaesthesia. We used primary cell cultured neurones from newborn rats, pre-incubated with or without a tyrosine kinase inhibitor before propofol stimulation. The effect of propofol on tyrosine phosphorylation and changes in [Ca2+]i were investigated. Propofol (3 microg mL(-1), 16.8 microM) increased intracellular calcium levels by 122 +/- 34% (mean +/- SEM) when applied to neurones in calcium free medium. This rise in [Ca2+]i was lowered by 68% when the cells were pre-incubated with the tyrosine kinase inhibitor herbimycin A before exposure to propofol (P < 0.05). Propofol caused an increase (33 +/- 10%) in tyrosine phosphorylation, with maximum at 120 s, of the beta-subunit of the GABA(A)-receptor. This tyrosine phosphorylation was decreased after pre-treatment with herbimycin A (44 +/- 7%, P < 0.05), and was not affected by the absence of exogenous calcium in the medium. Tyrosine kinase participates in the propofol signalling system by inducing the release of calcium from intracellular stores and by modulating the beta-subunit of the GABA(A)-receptor.


Subject(s)
Calcium/metabolism , Neurons/drug effects , Neurons/metabolism , Propofol/pharmacology , Protein-Tyrosine Kinases/metabolism , Receptors, GABA-A/metabolism , Animals , Benzoquinones , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Lactams, Macrocyclic , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinones/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Reference Values , Rifabutin/analogs & derivatives
7.
Br J Anaesth ; 83(3): 467-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10655921

ABSTRACT

Propofol depresses both cerebral oxygen consumption and glucose utilization. We tested the hypothesis that these well described effects on brain metabolism are manifest by a reduction in neuronal acid production in vitro. The rate of extracellular acidification in primary cell cultures of rat cortical neurones was measured using a novel instrument (silicon microphysiometer) after stimulation with propofol 0.3, 3 and 30 micrograms ml-1. Intralipid 10% served as a control. Propofol 3 micrograms ml-1 caused a mean decrease of 1.51 (SEM 0.71)% in baseline acidification rate, which was significantly greater than that produced by 0.3 microgram ml-1 or Intralipid alone (P < 0.05). The reduction after stimulation with propofol 30 micrograms ml-1 was 4.68 (0.35)% of baseline rates and this in turn was significantly greater than that elicited by propofol 3 or 0.3 microgram ml-1, or Intralipid (P < 0.001). We have confirmed the depressant effect of propofol on cerebral metabolism and established that propofol inhibits neuronal acid excretion in vitro.


Subject(s)
Anesthetics, Intravenous/pharmacology , Cerebral Cortex/drug effects , Propofol/pharmacology , Animals , Cell Culture Techniques , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Emulsions , Extracellular Space/metabolism , Fat Emulsions, Intravenous/pharmacology , Hydrogen-Ion Concentration/drug effects , Phospholipids , Potentiometry , Rats , Rats, Sprague-Dawley , Soybean Oil
8.
Br J Anaesth ; 79(1): 78-83, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9301393

ABSTRACT

We have examined the effects of the volatile general anaesthetic agent, halothane, on K+ and carbachol stimulated [3H]noradrenaline release and associated increases in intracellular Ca2+ in a cultured human neuroblastoma cell line, SH-SY5Y. K+ (but not carbachol) stimulated [3H]noradrenaline release, and the increase in intracellular Ca2+ concentration was entirely extracellular Ca2+ dependent. Halothane produced a dose-dependent reduction in K+ evoked release of [3H]noradrenaline with significant inhibition (17%) occurring from 1.26 atm%. Basal and carbachol evoked release were unaffected. Halothane also produced a dose-dependent reduction in K+ evoked increases (measured at the peak) in intracellular Ca2+ with significant inhibition (29%) occurring from 0.88 atm%. K+ plateau, basal and carbachol evoked increases in intracellular Ca2+ were unaffected. These data suggest that halothane reduced Ca2+ entry through voltage-sensitive Ca2+ channels and implicate this important class of ion channel in the mechanism of anaesthesia.


Subject(s)
Anesthetics, Inhalation/pharmacology , Calcium/metabolism , Halothane/pharmacology , Neurons/drug effects , Norepinephrine/metabolism , Carbachol/pharmacology , Dose-Response Relationship, Drug , Humans , Neuroblastoma , Neurons/physiology , Potassium/antagonists & inhibitors , Potassium/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...