Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neurotoxicology ; 50: 10-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26205086

ABSTRACT

Exposure to organophosphorus (OP) compounds, such as pesticides and the chemical warfare agents (soman and sarin), respectively represents a major health problem and a threat for civilian and military communities. OP poisoning may induce seizures, status epilepticus and even brain lesions if untreated. We recently proved that a combination of atropine sulfate and ketamine, a glutamatergic antagonist, was effective as an anticonvulsant and neuroprotectant in mice and guinea-pigs exposed to soman. Since OP exposure may also occur in conditions of heat strain due to climate, wearing of protective gears or physical exercise, we previously demonstrated that ketamine/atropine association may be used in a hot environment without detrimental effects. In the present study, we assess soman toxicity and evaluate the effects of the ketamine/atropine combination on soman toxicity in a warm thermoneutral environment. Male Wistar rats, exposed to 31°C (easily reached under protective equipments), were intoxicated by soman and treated with an anesthetic dose of ketamine combined with atropine sulfate. Body core temperature and spontaneous locomotor activity were continuously monitored using telemetry. At the end of the warm exposure, blood chemistry and brain mRNA expression of some specific genes were measured. In soman-intoxicated animals, metabolic and genic modifications were related to convulsions rather than to soman intoxication by itself. In the warm environment, ketamine/atropine combination did not produce any side-effect on the assessed variables. Furthermore, the ketamine/atropine combination exhibited beneficial therapeutic effects on soman-intoxicated rats such as a limitation of convulsion-induced hyperthermia and of the increase in some blood chemistry markers.


Subject(s)
Anticonvulsants/therapeutic use , Atropine/therapeutic use , Chemical Warfare Agents/toxicity , Epilepsy/chemically induced , Epilepsy/drug therapy , Ketamine/therapeutic use , Soman/toxicity , Animals , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Cholinesterases/blood , Corticosterone/blood , Disease Models, Animal , Drug Therapy, Combination , Fever/chemically induced , Fever/drug therapy , Gene Expression Regulation/drug effects , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Time Factors
2.
Eur J Pharmacol ; 691(1-3): 77-85, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22796677

ABSTRACT

Exposure to organophosphorus compounds, either pesticides or chemical warfare agents such as soman or sarin, represents a major health problem. Organophosphorus poisoning may induce seizures, status epilepticus and even brain lesions if untreated. Ketamine, an antagonist of glutamatergic receptors, was recently proved to be effective in combination with atropine sulfate as an anticonvulsant and neuroprotectant in mice and guinea pigs exposed to soman. Organophosphorus exposure may also occur in conditions of contemporary heat exposure. Since both MK-801, a more potent glutamatergic antagonist than ketamine, and atropine sulfate are detrimental for thermoregulation, we evaluated the pathophysiological consequences of ketamine/atropine combinations in a hot environment. Male wistar rats were exposed to 38°C ambient temperature and treated with atropine sulfate and/or ketamine (anesthetic and subanesthetic doses). The abdominal temperature and spontaneous locomotor activity were continuously monitored using telemetry. At the end of heat exposure, blood chemistry and the mRNA expression of some specific genes in the brain were assessed. Unlike MK-801, ketamine did not induce any deleterious effect on thermoregulation in rats. Conversely, atropine sulfate led to heatstroke and depressed the heat-induced blood corticosterone increase. Furthermore, it induced a dramatic increase in Hsp70 and c-Fos mRNA levels and a decrease in IκBα mRNA expression, both suggesting brain aggression. When combined with the anesthetic dose of ketamine, some of the atropine-induced metabolic disturbances were modified. In conclusion, ketamine can be used in hot environment and may even limit some of the biological alterations induced by atropine sulfate in these conditions.


Subject(s)
Body Temperature Regulation/drug effects , Hot Temperature , Ketamine/adverse effects , Animals , Atropine/pharmacology , Behavior, Animal/drug effects , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Brain/physiology , Drug Interactions , Gene Expression Regulation/drug effects , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
3.
Toxicol Lett ; 210(1): 71-7, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22306367

ABSTRACT

The link between cell death and increased cyclooxygenases-2 (COX-2) activity has not been clearly established. In this study, we examined whether COX-2 activation contributed to the mechanism of neurotoxicity produced by an organophosphorous nerve agent in cultured rat cortical neurons. Exposure of neuronal cells to the nerve agent, VX resulted in an increase in COX enzyme activity in the culture media. A concentration dependent increase in the activity levels of COX-2 enzyme was observed while there was little to no effect on COX-1. In addition, COX-2 mRNA and protein levels increased several hours post-VX exposure. Pre-treatment of the cortical cells with the COX-2 selective inhibitor, NS 398 resulted in a decrease in both the enzyme activity and prostaglandin (PGE(2) and PGF(2α)) release, as well as in a reduction in cell death. These findings indicate that the increase in COX-2 activity may contribute to the mechanism of VX-induced neurotoxicity in cultured rat cortical neuron.


Subject(s)
Cell Death/physiology , Cerebral Cortex/drug effects , Cyclooxygenase 2/physiology , Neurons/enzymology , Animals , Blotting, Western , Cerebral Cortex/cytology , Cholinesterase Inhibitors/pharmacology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/metabolism , Dinoprost/analysis , Dinoprostone/analysis , Enzyme Activation/drug effects , Neurons/chemistry , Neurons/drug effects , Organothiophosphorus Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
4.
Toxicol Appl Pharmacol ; 259(2): 195-209, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22245128

ABSTRACT

Epileptic seizures and status epilepticus (SE) induced by the poisoning with organophosphorus nerve agents (OP), like soman, are accompanied by neuroinflammation whose role in seizure-related brain damage (SRBD) is not clear. Antagonists of the NMDA glutamate ionotropic receptors are currently among the few compounds able to arrest seizures and provide neuroprotection even during refractory status epilepticus (RSE). Racemic ketamine (KET), in combination with atropine sulfate (AS), was previously shown to counteract seizures and SRBD in soman-poisoned guinea-pigs. In a mouse model of severe soman-induced SE, we assessed the potentials of KET/AS combinations as a treatment for SE/RSE-induced SRBD and neuroinflammation. When starting 30min after soman challenge, a protocol involving six injections of a sub-anesthetic dose of KET (25mg/kg) was evaluated on body weight loss, brain damage, and neuroinflammation whereas during RSE, anesthetic protocols were considered (KET 100mg/kg). After confirming that during RSE, KET injection was to be repeated despite some iatrogenic deaths, we used these proof-of-concept protocols to study the changes in mRNA and related protein contents of some inflammatory cytokines, chemokines and adhesion molecules in cortex and hippocampus 48h post-challenge. In both cases, the KET/AS combinations showed important neuroprotective effects, suppressed neutrophil granulocyte infiltration and partially suppressed glial activation. KET/AS could also reduce the increase in mRNA and related pro-inflammatory proteins provoked by the poisoning. In conclusion, the present study confirms that KET/AS treatment has a strong potential for SE/RSE management following OP poisoning. The mechanisms involved in the reduction of central neuroinflammation remain to be studied.


Subject(s)
Atropine/pharmacology , Chemical Warfare Agents/toxicity , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Muscarinic Antagonists/pharmacology , Soman/toxicity , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Animals , Area Under Curve , Brain/cytology , Brain/drug effects , Brain/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Male , Mice , Neuroglia/immunology , Neutrophils/immunology , Polymerase Chain Reaction , RNA, Messenger/chemistry , RNA, Messenger/genetics , Random Allocation , Status Epilepticus/immunology
5.
Epilepsia ; 52(12): 2315-25, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21955106

ABSTRACT

PURPOSE: Neuroinflammation appears as a prominent feature of the mesiotemporal lobe epilepsy syndrome (MTLE) that is observed in human patients and animal models. However, the precise temporal relationship of its development during epileptogenesis remains to be determined. The aim of the present study was to investigate (1) the time course and spatial distribution of neuronal death associated with seizure development, (2) the time course of microglia and astrocyte activation, and (3) the kinetics of induction of mRNAs from neuroinflammatory-related proteins during the emergence of recurrent seizures. METHODS: Experimental MTLE was induced by the unilateral intrahippocampal injection of kainate in C57BL/6 adult mice. Microglial and astrocytic changes in both ipsilateral and contralateral hippocampi were examined by respectively analyzing griffonia simplicifolia (GSA) lectin staining and glial fibrillary acidic protein (GFAP) immunoreactivity. Changes in mRNA levels of selected genes of cytokine and cytokine regulatory proteins (interleukin-1ß, IL-1ß; interleukin-1 receptor antagonist, IL-1Ra; suppressor of cytokine signaling 3, SOCS3) and enzymes of the eicosanoid pathway (group IVA cytosolic phospholipase A2, cPLA(2)-α; cycloxygenase-2, COX-2) were studied by reverse transcription-quantitative real time polymerase chain reaction. KEY FINDINGS: Our data show an immediate cell death occurring in the kainate-injected hippocampus during the initial status epilepticus (SE). A rapid increase of activated lectin-positive cells and GFAP-immunoreactivity was subsequently detected in the ipsilateral hippocampus. In the same structure, Il-1ß, IL-1Ra, and COX-2 mRNA were specifically increased during SE and epileptogenesis with a different time course. Conversely, the expression of SOCS3 mRNA, a surrogate marker of interleukin signaling, was mainly increased in the contralateral hippocampus after SE. SIGNIFICANCE: Our data show that specific neuroinflammatory pathways are activated in a time- and structure-dependent manner with putative distinct roles in epileptogenesis.


Subject(s)
Cytokines/metabolism , Epilepsy, Temporal Lobe/complications , Gene Expression Regulation/physiology , Inflammation/etiology , Seizures/etiology , Animals , Cell Death/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/genetics , Disease Models, Animal , Eicosanoids/genetics , Eicosanoids/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/metabolism , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , Plant Lectins , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors
6.
J Neurosci Res ; 88(5): 1000-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19937810

ABSTRACT

Reference genes are often used to normalize expression of data from real-time quantitative reverse transcription-polymerase chain reaction (RT-qPCR), and only a validation of their stability during a given experimental paradigm leads to reliable interpretations. The present study was thus designed to validate potential reference genes in a mouse model of mesiotemporal lobe epilepsy (MTLE) with focal seizures after unilateral intrahippocampal injection of kainate (KA). Ipsilateral and contralateral hippocampi were removed during nonconvulsive status epilepticus (5 hr), epileptogenesis (7 days), and the chronic period of recurrent focal seizures (21 days). Naive animals were equally studied. The stability of eight potential reference genes (hypoxanthine phosphoribosyltransferase, Hprt1; peptidylprolyl isomerase A, Ppia; TATA box binding protein, Tbp; beta-actin, Actb; acidic ribosomal phosphoprotein P0, Arbp; glyceraldehyde-3-phosphate dehydrogenase, Gapdh; ribosomal RNA 18S, 18S rRNA; and glucuronidase beta, Gusb) were determined using geNorm and NormFinder software. The first five (Hprt1, Ppia, Tbp, Actb, and Arbp) were found to be stable across the different phases of the disease and appeared adequate for normalizing RT-qPCR data in this model. This was in contrast to the other three (18S rRNA, Gapdh, and Gusb), which showed unstable expressions and should be avoided. The analysis of KA-induced changes in the expression of glial fibrillary acidic protein (Gfap) gene resulted in various relative expressions or even a completely different pattern when unstable reference genes were used. These results highlight the absolute need to validate the reference genes for a correct interpretation of mRNA quantification.


Subject(s)
Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Computational Biology/methods , Disease Models, Animal , Epilepsies, Partial/genetics , Epilepsies, Partial/metabolism , Epilepsies, Partial/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Genomic Instability/genetics , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Molecular Biology/methods , RNA, Messenger/analysis , Reference Standards , Software , Status Epilepticus/genetics , Status Epilepticus/metabolism , Status Epilepticus/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL