Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Chem Biol Interact ; 203(1): 154-9, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23044489

ABSTRACT

Organophosphorus nerve agents (NA), potent irreversible cholinesterase inhibitors, could induce severe seizures, status epilepticus (SE), seizure-related brain damage (SRBD) and lethality. Despite the lack of data in the case of NA, clinical evidences suggest that SE survivors could suffer from neurological/cognitive deficits and impairments such as spontaneous recurrent seizures (epilepsy) after a latent period of epileptogenesis. It is beyond doubt that an effective and quick management of the initial seizures and prevention of SRBD are critical to prevent these long-term consequences, explaining why most experimental data are focusing on the 5-40min post-exposure time frame. However, in field conditions, treatment may be delayed and with the exception of NMDA receptor antagonists, currently no drug provides protection (against lethality, seizures, SRBD and neurological consequences) when seizures are left unabated for one hour or more. Ketamine (KET) is the only NMDA antagonist licensed as an injectable drug in different countries and remains an anesthetic of choice in some difficult field conditions. In this short review paper, after a presentation of some of the key points of the pathophysiology of NA-induced SE and a quick survey of the potential therapeutic avenues in the context of delayed treatment of NA-induced SE, we will review the recent data we obtained showing that KET, in combination with atropine sulfate (AS), with or without a benzodiazepine, considerably reduces soman-induced neuroinflammation, provides neuroprotection, histologically and functionally, and also positively modify soman-induced changes in brain metabolism. Finally, we will also mention some results from safety studies including those bringing evidence that, at difference with MK-801, KET does not impair thermoregulation and even seems to reduce AS-induced heat stress. All in all, KET, in combination, appears a good candidate for the out-of-hospital treatment of severe NA-induced SE.


Subject(s)
Chemical Warfare Agents/toxicity , Ketamine/administration & dosage , Soman/toxicity , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Animals , Brain/drug effects , Brain/physiopathology , Cholinesterase Inhibitors/toxicity , Dizocilpine Maleate/administration & dosage , Guinea Pigs , Humans , Neuroprotective Agents/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Status Epilepticus/physiopathology , Time Factors
2.
J Proteome Res ; 11(7): 3782-95, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22621346

ABSTRACT

This work presents a model combining quantitative proton HRMAS NMR data and PLS-DA for neuropathology and neuroprotection evaluation. Metabolic data were also confronted to histopathological results obtained using the same experimental conditions. Soman, when not lethal, can induce status epilepticus (SE), brain damage, histological lesions, and profound cerebral metabolic disorders as revealed using (1)H HRMAS NMR. Our challenge was to evaluate delayed treatments, which could control refractory SE and avoid brain lesions. For this aim, we have built a statistical model of soman intoxication describing brain metabolite evolution during 7 days. We have then used this model to evaluate the efficiency of a combination of ketamine/atropine (KET/AS) administrated 1 and 2 h after SE induction, compared to the immediate anticonvulsant therapy midazolam/atropine sulfate (MDZ/AS). Furthermore, quantitation of HRMAS NMR data allowed us to follow individual evolution of 17 metabolites. N-Acetylaspartate, lactate, or taurine presented a long lasting disruption, while glutamine, alanine, glycerophosphocholine and myo-inositol showed disruptions for 3 days with a reversion at day 7. These changes were completely normalized by the administration of MDZ/AS. Interestingly, they were also almost completely reversed by KET/AS 1 h postsoman. This work suggests further the predictive interest of HRMAS and PLS-DA for neuropathology/neuroprotection studies and also confirms, on the metabolic aspects, the neuroprotective potentials of KET/AS combinations for the delayed treatment of soman-induced SE.


Subject(s)
Atropine/therapeutic use , Ketamine/therapeutic use , Metabolome , Neuroprotective Agents/therapeutic use , Status Epilepticus/metabolism , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Atropine/pharmacology , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Drug Therapy, Combination , Ketamine/pharmacology , Magnetic Resonance Spectroscopy , Male , Mice , Midazolam/pharmacology , Midazolam/therapeutic use , Multivariate Analysis , Neuroprotective Agents/pharmacology , Principal Component Analysis , Soman , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/pathology
3.
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
4.
Neurotoxicology ; 33(4): 789-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22155333

ABSTRACT

The Woelcke method is classically used for myelin staining. Degenerating neurons can be revealed histologically by hemalun and phloxin (H&P) where they appear "eosinophilic". In the first 24 h following soman-induced status epilepticus, we observed that the Woelcke method also revealed condensed, dark blue/black cells (W+ cells) in the gray matter of brain regions known to be sites of seizure-related brain damage, marked by the presence of eosinophilic cells. In the present study, using adjacent brain sections alternately stained with either the Woelcke or the H&P method, we show that eosinophilic cells and W+ cells are the same degenerating cells. Moreover, we show that semi-automated quantitative evaluation of W+ cells through computerized image analysis is considerably easier and faster than that of eosinophilic cells. It is therefore concluded that the Woelcke technique could be very useful, especially for quantifying acute brain cell damage following status epilepticus.


Subject(s)
Brain/pathology , Coloring Agents , Hematoxylin/analogs & derivatives , Myelin Sheath/metabolism , Neurons/pathology , Staining and Labeling/methods , Status Epilepticus/pathology , Animals , Biomarkers/metabolism , Brain/metabolism , Cell Count , Disease Models, Animal , Eosine I Bluish , Image Processing, Computer-Assisted , Male , Mice , Nerve Degeneration , Neurons/metabolism , Soman , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Time Factors
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.
Toxicology ; 277(1-3): 38-48, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20816720

ABSTRACT

The organophosphorus nerve agent soman is an irreversible cholinesterase (ChE) inhibitor that can produce long-lasting seizures and seizure-related brain damage (SRBD) in which acetylcholine and glutamate are involved. Since these neurotransmitters play a key-role in the auditory function, it was hypothesized that a hearing test may be an efficient way for detecting the central effects of soman intoxication. In the present study, distortion product otoacoustic emissions (DPOAEs), a non-invasive audiometric method, were used in rats administered with soman (70 µg/kg). Four hours post-soman, DPOAE intensities were significantly decreased. They returned to baseline one day later. The amplitude of the temporary drop of the DPOAEs was well related to the severity of the intoxication. The greatest change was recorded in the rats that survived long-lasting convulsions, i.e. those that showed the highest ChE inhibition in brain and severe encephalopathy. Furthermore, the administration, immediately after soman, of a three-drug therapy composed of atropine sulfate, HI-6 and avizafone abolished the convulsions, the transient drop of DPOAEs at 4h and the occurrence of SRBD at 28 h without modifying brain ChE inhibition. This showed that DPOAE change was not directly related to soman-induced inhibition of cerebral ChE but rather to its neuropathological consequences. The present findings strongly suggest that DPOAEs represent a promising non-invasive tool to predict SRBD occurrence in nerve agent poisoning and to control the efficacy of a neuroprotective treatment.


Subject(s)
Brain Injuries/chemically induced , Brain Injuries/pathology , Otoacoustic Emissions, Spontaneous/drug effects , Otoacoustic Emissions, Spontaneous/physiology , Soman/toxicity , Animals , Brain Injuries/prevention & control , Cochlea/drug effects , Cochlea/physiology , Forecasting , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Random Allocation , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/pathology , Seizures/prevention & control
7.
Toxicology ; 268(1-2): 78-88, 2010 Jan 31.
Article in English | MEDLINE | ID: mdl-20003915

ABSTRACT

A convulsive dose of soman induces seizure-related brain damage (SRBD), including cerebral edema (CE) and cell death. In 1993, an American study demonstrated that hypertonic mannitol (Mann) intravenously (i.v.) administered 1 min and 5h post-soman was an effective neuroprotectant in intoxicated rats. Using a similar protocol, we recently failed to reproduce this success in intoxicated mice. In the present study, also performed in mice, the persistence or the amplitude of the osmotic load was increased by reducing the time interval between two injections of Mann or by augmenting the number of injections. Mice were pre-treated with the oxime HI-6 and then intoxicated with a convulsive dose of soman (172 microg/kg). Afterward, they were administered a first i.v. bolus of Mann 20% 1 min post-challenge and a second one either 5, or 2, or 1h after. Additional animals were given either one (1 min post-soman), or two (1 min and 1h post-soman), or three (1 min, 1 and 2h post-soman) series of three injections of Mann at 5 min intervals. Non-intoxicated mice treated with Mann (same protocols as above) and intoxicated mice treated with Mann vehicle (saline) served as controls. At 24h post-intoxication, the survivors were sacrificed and their brains prepared for quantitative histological assessment of cell damage, CE, and ventricle size. Whatever the protocol, Mann had no effect on soman-induced convulsions but did provide considerable antilethal activity. Histologically, Mann did not reduce the cell damage or CE. It even showed a dose-dependent trend toward aggravation of SRBD in some regions and promoted subarachnoid hemorrhages. Conversely, in one of the treatment protocol, it reduced soman-induced enlargement of ventricle size. Although treatment with hypertonic Mann showed some benefit on mortality and ventricle size, it failed to be an effective neuroprotector in soman-intoxicated mice and even increased the detrimental impact of soman at the cerebral level. Therefore, no clear recommendation could be drawn from the present study in view of a possible clinical use of hyperosmolar treatment in the medical management of soman poisoning.


Subject(s)
Convulsants/poisoning , Mannitol/therapeutic use , Seizures/chemically induced , Soman/poisoning , Animals , Male , Mice , Osmolar Concentration , Poisoning/drug therapy
9.
Toxicology ; 253(1-3): 97-103, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18824071

ABSTRACT

PURPOSE: A convulsive dose of soman induces seizure-related brain damage (SRBD), including cerebral edema (CE) and neuronal loss. In the present study on soman-intoxicated mice, we applied diffusion-weighted magnetic resonance imaging (DW-MRI) and quantitative histology, and we measured brain water content to investigate the antiedematous and neuroprotective efficacies of two hyperosmolar treatments: mannitol (Mann) and hypertonic saline (HTS). METHODS: Mice intoxicated with soman (172 microg/kg after a protective pretreatment) were administered 1 min and 5-h post-challenge an i.v. bolus of saline, of Mann or of HTS. 1 day later, mice were examined with DW-MRI and then sacrificed for brain histology. Additional animals were intoxicated and treated similarly for the measurement of the brain water content (dry/wet weight method). RESULTS: In intoxicated controls, a significant decrease of the apparent diffusion coefficient (ADC), numerous damaged (eosinophilic) cells, high edema scores, and a significant increase in brain water content were detected 24-h post-challenge in sensitive brain structures. These soman-induced changes were not significantly modified by treatment with Mann or HTS. CONCLUSIONS: Treatment with hyperosmolar solutions did not reduce the effects of soman on ADC, on cell damage and on CE. Therefore, despite similar treatment protocols, the prominent protection by Mann that was previously demonstrated by others in poisoned rats, was not reproduced in our murine model.


Subject(s)
Brain Diseases/drug therapy , Brain Edema/drug therapy , Brain/pathology , Mannitol/therapeutic use , Neuroprotective Agents/therapeutic use , Saline Solution, Hypertonic/therapeutic use , Soman/poisoning , Animals , Body Water , Brain/drug effects , Brain Chemistry/drug effects , Brain Diseases/chemically induced , Brain Diseases/pathology , Brain Edema/chemically induced , Brain Edema/pathology , Convulsants/administration & dosage , Convulsants/poisoning , Diffusion Magnetic Resonance Imaging , Male , Mice , Statistics, Nonparametric
10.
Toxicology ; 238(2-3): 119-29, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17618030

ABSTRACT

The organophosphorus nerve agent soman is an irreversible cholinesterase (ChE) inhibitor that can produce long-lasting seizures and brain damage in which the neurotransmitters acetylcholine and glutamate are involved. These same neurotransmitters play key-roles in the auditory function. It was then assumed that exploring the hearing function may provide markers of the central events triggered by soman intoxication. In the present study, distortion product otoacoustic emissions (DPOAEs), a non-invasive audiometric method, were used to monitor cochlear functionality in rats administered with a moderate dose of soman (45 microg/kg). DPOAEs were investigated either 4h or 24h post-challenge. In parallel, the effects of soman on whole blood and brain ChE activity and on brain histology were also studied. The first main result is that DPOAE intensities were significantly decreased 4h post-soman and returned to baseline at 24h. The amplitude changes were well related to the severity of symptoms, with the greatest change being recorded in the rats that survived long-lasting convulsions. The second main result is that baseline DPOAEs recorded 8 days before soman appear to predict the severity of symptoms produced by the intoxication. Indeed, the lowest baseline DPOAEs corresponded to the occurrence of long-lasting convulsions and brain damage and to the greatest inhibition in central ChE. These results thus suggest that DPOAEs represent a promising non-invasive tool to assess and predict the central consequences of nerve agent poisoning. Further investigations will be carried out to assess the potential applications and the limits of this non-invasive method.


Subject(s)
Neurotoxicity Syndromes/etiology , Otoacoustic Emissions, Spontaneous/drug effects , Soman/toxicity , Acoustic Stimulation , Animals , Audiometry/methods , Audiometry, Evoked Response/methods , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Chemistry/drug effects , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/blood , Cholinesterase Inhibitors/toxicity , Cochlea/drug effects , Cochlea/physiopathology , Injections, Subcutaneous , Male , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/physiopathology , Rats , Rats, Wistar , Severity of Illness Index , Soman/administration & dosage , Soman/blood , Survival Analysis , Time Factors
11.
Toxicology ; 238(2-3): 166-76, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17662515

ABSTRACT

Following exposure to the organophosphorus nerve agent soman, the development of long-lasting seizures and build-up of irreversible seizure-related brain damage (SRBD) still represent a therapeutic challenge. A neuro-inflammatory reaction takes place in the brain after poisoning but its characteristics and potential role in SRBD and post-status epilepticus epileptogenesis is not well understood. In the present study we have analyzed by quantitative RT-PCR the time course of changes in mRNA levels of IL-1beta, TNFalpha, IL-6, ICAM-1 and SOCS3 in hippocampus, whole cortex and cerebellum in a mouse model of severe seizures and neuropathy up to 7 days after poisoning. Mice received an injection of the oxime HI-6 (50mg/kg) 5 min prior to the administration of a convulsive dose of soman (172 microg/kg). An important and highly significant increase of the five mRNA levels was recorded in cortex and hippocampus. In the cortex, the activation was generally detected as early as 1h post-intoxication with a peak response recorded between 6 and 24h. In the hippocampus, the gene up-regulation was delayed to 6h post-soman and the peak response observed between 24 and 48 h. After peaking, the response declined (except for ICAM in the hippocampus) but remained elevated, some of them significantly, at day 7. Interestingly, in the cerebellum, some changes were also observed but were several fold smaller. In conclusion, the present study indicates a quick neuro-inflammatory gene response that does not subside over 7 days suggesting a potential role in the neurological consequences of soman-induced status epilepticus.


Subject(s)
Gene Expression Regulation/drug effects , Inflammation/genetics , Seizures/complications , Soman/toxicity , Animals , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chemical Warfare Agents/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/etiology , Intercellular Adhesion Molecule-1/genetics , Interleukin-1beta/genetics , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Seizures/chemically induced , Soman/administration & dosage , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Time Factors , Tumor Necrosis Factors/genetics
12.
Toxicology ; 234(3): 185-93, 2007 May 20.
Article in English | MEDLINE | ID: mdl-17408839

ABSTRACT

Soman poisoning is known to induce full-blown tonic-clonic seizures, status epilepticus (SE), seizure-related brain damage (SRBD) and lethality. Previous studies in guinea-pigs have shown that racemic ketamine (KET), with atropine sulfate (AS), is very effective in preventing death, stopping seizures and protecting sensitive brain areas when given up to 1h after a supra-lethal challenge of soman. The active ketamine isomer, S(+) ketamine (S-KET), is more potent than the racemic mixture and it also induces less side-effects. To confirm the efficacy of KET and to evaluate the potential of S-KET for delayed medical treatment of soman-induced SE, we studied different S-KET dose regimens using the same paradigm used with KET. Guinea-pigs received pyridostigmine (26 microg/kg, IM) 30min before soman (62 microg/kg, 2 LD(50), IM), followed by therapy consisting of atropine methyl nitrate (AMN) (4 mg/kg, IM) 1min following soman exposure. S-KET, with AS (10mg/kg), was then administered IM at different times after the onset of seizures, starting at 1h post-soman exposure. The protective efficacy of S-KET proved to be comparable to KET against lethality and SRBD, but at doses two to three times lower. As with KET, delaying treatment by 2h post-poisoning greatly reduced efficacy. Conditions that may have led to an increased S-KET brain concentration (increased doses or number of injections, adjunct treatment with the oxime HI-6) did not prove to be beneficial. In summary, these observations confirm that ketamine, either racemic or S-KET, in association with AS and possibly other drugs, could be highly effective in the delayed treatment of severe soman intoxication.


Subject(s)
Atropine/pharmacology , Brain Damage, Chronic/chemically induced , Brain Damage, Chronic/prevention & control , Cholinesterase Inhibitors/toxicity , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Muscarinic Antagonists/pharmacology , Soman/toxicity , Status Epilepticus/chemically induced , Status Epilepticus/prevention & control , Animals , Guinea Pigs , Lethal Dose 50 , Male , Oximes , Pyridinium Compounds/pharmacology , Status Epilepticus/mortality , Stereoisomerism
13.
Toxicol Appl Pharmacol ; 220(2): 125-37, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17350063

ABSTRACT

PURPOSE: In the present study, diffusion-weighted magnetic resonance imaging (DW-MRI) and histology were used to assess cerebral edema and lesions in mice intoxicated by a convulsive dose of soman, an organophosphate compound acting as an irreversible cholinesterase inhibitor. METHODS: Three hours and 24 h after the intoxication with soman (172 microg/kg), the mice were anesthetized with an isoflurane/N(2)O mixture and their brain examined with DW-MRI. After the imaging sessions, the mice were sacrificed for histological analysis of their brain. RESULTS: A decrease in the apparent diffusion coefficient (ADC) was detected as soon as 3 h after the intoxication and was found strongly enhanced at 24 h. A correlation was obtained between the ADC change and the severity of the overall brain damage (edema and cellular degeneration): the more severe the damage, the stronger the ADC drop. Anesthesia was shown to interrupt soman-induced seizures and to attenuate edema and cell change in certain sensitive brain areas. Finally, brain water content was assessed using the traditional dry/wet weight method. A significant increase of brain water was observed following the intoxication. CONCLUSIONS: The ADC decrease observed in the present study suggests that brain edema in soman poisoning is mainly intracellular and cytotoxic. Since entry of water into the brain was also evidenced, this type of edema is certainly mixed with others (vasogenic, hydrostatic, osmotic). The present study confirms the potential of DW-MRI as a non-invasive tool for monitoring the acute neuropathological consequences (edema and neurodegeneration) of soman-induced seizures.


Subject(s)
Brain Edema/chemically induced , Brain Edema/pathology , Convulsants/poisoning , Soman/poisoning , Anesthesia , Anesthetics, Inhalation , Animals , Body Water/metabolism , Brain/pathology , Diffusion Magnetic Resonance Imaging , Electroencephalography/drug effects , Image Interpretation, Computer-Assisted , Isoflurane , Male , Mice , Nitrous Oxide
14.
Neurotoxicology ; 27(2): 201-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16309744

ABSTRACT

Soman poisoning induces long-term neuropathology characterized by the presence of damaged neurons up to 2 months after exposure in various central brain areas, especially the hippocampal CA1 layer. Rapid depletion of this layer could therefore be expected. Surprisingly, the CA1 layer remained consistently visible, suggesting delayed death of these damaged neurons, potentially accompanied by neuronal regeneration. To address this issue, mice were exposed to a convulsive dose of soman (110 microg/kg followed by 5.0mg/kg of atropine methyl nitrate (MNA) 1 min later) and brains were collected from day 1 to day 90 post-exposure. Damaged and residual healthy neurons were quantified on brain sections using hemalun-phloxin and fluorojade staining or neuronal nuclei antigen (NeuN) immunohistochemistry. On post-soman day 1, a moderate neuronal cell death was noticed in the hippocampal CA1 layer. In this area, an important and steady quantity of damaged neurons (about 48% of the whole pyramidal neurons) was detected from post-soman day 1 to day 30. Thus, throughout this period, damaged neurons seemed to survive, as confirmed by the unmodified depth of the hippocampal CA1 layer. The dramatic disappearance of the damaged neurons occurred only later during the experiment and was almost complete at day 90 after soman exposure. Interestingly, between day 30 and day 90 following poisoning, an increase in the number of residual healthy pyramidal neurons was observed. These different kinetic patterns related to the density of total, damaged and residual healthy neurons after soman poisoning demonstrate that neuronal regeneration is delayed in the hippocampal CA1 layer and is concomitant to the death of damaged neurons.


Subject(s)
Cholinesterase Inhibitors/poisoning , Hippocampus/pathology , Nerve Regeneration/physiology , Neurons/pathology , Soman/poisoning , Animals , Cell Death/physiology , Cell Nucleus/metabolism , Fluoresceins , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Models, Statistical , Organic Chemicals
15.
Toxicology ; 215(1-2): 1-24, 2005 Nov 05.
Article in English | MEDLINE | ID: mdl-16054742

ABSTRACT

The organophosphorus compound soman, an irreversible inhibitor of cholinesterases, produces seizure activity and related brain damage. Studies using various biochemical markers of programmed cell death (PCD) suggested that soman-induced cell damage in the brain was apoptotic rather than necrotic. However, it has recently become clear that not all PCD is apoptotic, and the unequivocal demonstration of apoptosis requires ultrastructural examination. Therefore, the present study was undertaken to reinvestigate the damage produced in the brains of mice sacrificed at various times within the first 24 h or at 7 days after a convulsive dose of soman. Classical histology and ultrastructural examination were performed. The immunohistochemical expression of proteins (p53, Bax) involved in PCD, DNA fragmentation (TUNEL method at light and electron microscopy levels) and the glial reaction were also explored. Our study confirms that the severity of lesions depended on the duration of convulsions and shows that cerebral changes were still occurring as late as 7 days after the onset of long-lasting convulsions. Our observations also establish that there was a large variety of ultrastructurally distinct types of cell damage, including hybrid forms between apoptosis and necrosis, but that pure apoptosis was very rare. A prominent expression of p53 and Bax proteins was detected indicating that PCD mechanisms were certainly involved in the morphologically diverse forms of cell death. Since purely apoptotic cells were very rare, these protein expressions were presumably involved either in nonapoptotic cell death mechanisms or in apoptotic mechanisms occurring in parallel with nonapoptotic ones. Moreover, evidence for DNA fragmentation by the TUNEL method was found in apoptotic but also in numerous other morphotypes of cell damage. Therefore, TUNEL-positivity and the expression of PCD-related proteins, in the absence of ultrastructural confirmation, were here shown not to provide proof of apoptosis. In soman poisoning as well as in other cerebral pathologies, premature conclusions on this question can potentially be misleading and might even lead to detrimental therapies.


Subject(s)
Apoptosis/drug effects , Brain , Chemical Warfare Agents/toxicity , Seizures/chemically induced , Soman/toxicity , Animals , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , DNA Fragmentation/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Microscopy, Electron, Transmission , Seizures/pathology , Time Factors , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
16.
Brain Res ; 1051(1-2): 164-75, 2005 Jul 27.
Article in English | MEDLINE | ID: mdl-16005443

ABSTRACT

Nerve agent poisoning is known to induce full-blown seizures, seizure-related brain damage (SRBD), and lethality. Effective and quick management of these seizures is critical. In conditions of delayed treatment, presently available measures are inadequate calling for optimization of therapeutic approaches. The effects of ketamine/atropine sulfate (KET/AS) combinations were thus assessed as potential valuable delayed therapy in soman-poisoned male guinea pigs. Animals received pyridostigmine (26 microg/kg, i.m.) 30 min before soman (62 microg/kg, i.m.) followed by therapy consisting of atropine methyl nitrate (4 mg/kg) 1 min later. KET was then administered i.m. at different times after the onset of seizures, starting at 30 min post-poisoning. KET was always injected with atropine sulfate, itself given at a dose that was unable to modify seizures (2 to 10 mg/kg). Different treatment schemes (dose and time of injection) were evaluated. Sub-anesthetic doses of KET (10 mg/kg) could prevent lethality and stop ongoing seizures only when administered 30 min after challenge. An extended delay before treatment (up to 2 h) called for an increase in KET dose (up to 60 mg/kg three times), thus reaching anesthetic levels but without the need of any ventilation support. KET proved effective in stopping seizures, highly reducing SRBD and allowing survival with a progressive loss of efficacy when treatment was delayed beyond 1 h post-challenge. Preliminary results suggest that association with the benzodiazepine midazolam (1 mg/kg) might be interesting when treatment is initiated 2 h after poisoning, i.e., when KET efficacy is dramatically reduced. All in all, these observations suggest that KET, in association with atropine sulfate and possibly other drugs, may be highly effective in the delayed treatment of severe soman intoxication.


Subject(s)
Anticonvulsants/administration & dosage , Atropine/administration & dosage , Ketamine/administration & dosage , Midazolam/administration & dosage , Status Epilepticus/drug therapy , Animals , Chemical Warfare Agents/toxicity , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Electroencephalography/drug effects , Guinea Pigs , Male , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control , Soman/toxicity , Statistics, Nonparametric , Status Epilepticus/chemically induced , Time Factors
17.
Drug Chem Toxicol ; 27(3): 213-31, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15478944

ABSTRACT

Organophosphate (OP) nerve agents are amongst the most toxic chemicals. One of them, soman, can induce severe epileptic seizures and brain damage for which therapy is incomplete. The present study shows that pretreatment with flunarizine (Flu), a voltage-dependent calcium channel blocker, when used alone, does not produce any beneficial effect against the convulsions, neuropathology and lethality induced by soman. Flu was also tested in combination with atropine sulfate and diazepam. In this case, although only some results reach statistical significance, an encouraging general trend toward an improvement of the anticonvulsant, neuroprotective and antilethal capacities of this classical anti-OP two-drug regimen is constantly observed. In the light of these findings, it seems premature to definitely reject (or recommend) Flu as a possible adjuvant medication against soman poisoning. Further studies are required to determine its real potential interest.


Subject(s)
Calcium Channel Blockers/therapeutic use , Chemical Warfare Agents/toxicity , Flunarizine/therapeutic use , Soman/toxicity , Animals , Atropine/therapeutic use , Brain/pathology , Diazepam/therapeutic use , Drug Therapy, Combination , GABA Modulators/therapeutic use , Lethal Dose 50 , Male , Muscarinic Antagonists/therapeutic use , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/prevention & control , Soman/antagonists & inhibitors
18.
Neurotoxicology ; 23(1): 1-5, 2002 May.
Article in English | MEDLINE | ID: mdl-12164543

ABSTRACT

Today, organophosphate (OP) nerve agents are still considered as potential threats in both military or terrorism situations. OP agents are potent irreversible inhibitors of central and peripheral acetylcholinesterases. Pretreatment of OP poisoning relies on the subchronic administration of the reversible acetylcholinesterase (AChE) inhibitor pyridostigmine (PYR). Since PYR does not penetrate into the brain, it does not afford protection against seizures and subsequent neuropathology induced by an OP agent such as soman. Comparatively, huperzine (HUP) is a reversible AChE inhibitor that crosses the blood-brain barrier. HUP is presently approved for human use or is in course of clinical trials for the treatment of Alzheimer's disease or myasthenia gravis. HUP is also used as supplementary drug in the USA for correction of memory impairment. Besides, HUP has also been successfully tested for pretreatment of OP poisoning. This review summarizes the therapeutical value of HUP in this field. Moreover, the modes of action of HUP underlying its efficacy against OP agents are described. Efficacy appears mainly related to both the selectivity of HUP for red cell AChE which preserves scavenger capacity of plasma butyrylcholinesterases for OP agents and to the protection conferred by HUP on cerebral AChE. Finally, recent data, showing that HUP seems to be devoid of deleterious effects in healthy subjects, are also presented. Globally, this review reinforces the therapeutical value of HUP for the optimal pretreatment of OP poisoning.


Subject(s)
Cholinesterase Inhibitors/therapeutic use , Organophosphate Poisoning , Sesquiterpenes/therapeutic use , Alkaloids , Animals , Chemical Warfare Agents/poisoning , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...