Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Nanobiotechnology ; 21(1): 256, 2023 Aug 07.
Article in English | MEDLINE | ID: mdl-37550745

ABSTRACT

BACKGROUND: Organophosphate (OP)-induced delayed neurological damage is attributed to permanent neuropathological lesions caused by irreversible OP-neurocyte interactions, without potent brain-targeted etiological antidotes to date. The development of alternative therapies to achieve intracerebral OP detoxification is urgently needed. METHODS: We designed a brain-targeted nanoreactor by integrating enzyme immobilization and biomimetic membrane camouflaging protocols with careful characterization, and then examined its blood-brain barrier (BBB) permeability both in vitro and in vivo. Subsequently, the oxidative stress parameters, neuroinflammatory factors, apoptotic proteins and histopathological changes were measured and neurobehavioral tests were performed. RESULTS: The well-characterized nanoreactors exerted favourable BBB penetration capability both in vitro and in vivo, significantly inhibiting OP-induced intracerebral damage. At the cellular and tissue levels, nanoreactors obviously blocked oxidative stress, cellular apoptosis, inflammatory reactions and brain histopathological damage. Furthermore, nanoreactors radically prevented the occurrence of OP-induced delayed cognitive deficits and psychiatric abnormality. CONCLUSION: The nanoreactors significantly prevented the development of OP-induced delayed neurological damage, suggesting a potential brain-targeted etiological strategy to attenuate OP-related delayed neurological and neurobehavioral disorders.


Subject(s)
Organophosphate Poisoning , Organophosphates , Humans , Organophosphates/metabolism , Organophosphate Poisoning/metabolism , Organophosphate Poisoning/pathology , Brain/metabolism , Antidotes/metabolism , Nanotechnology
2.
Int J Mol Sci ; 23(15)2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35897817

ABSTRACT

Organophosphate (OP) compounds include highly toxic chemicals widely used both as pesticides and as warfare nerve agents. Existing countermeasures are lifesaving, but do not alleviate all long-term neurological sequelae, making OP poisoning a public health concern worldwide and the search for fully efficient antidotes an urgent need. OPs cause irreversible acetylcholinesterase (AChE) inhibition, inducing the so-called cholinergic syndrome characterized by peripheral manifestations and seizures associated with permanent psychomotor deficits. Besides immediate neurotoxicity, recent data have also identified neuroinflammation and microglia activation as two processes that likely play an important, albeit poorly understood, role in the physiopathology of OP intoxication and its long-term consequences. To gain insight into the response of microglia to OP poisoning, we used a previously described model of diisopropylfluorophosphate (DFP) intoxication of zebrafish larvae. This model reproduces almost all the defects seen in poisoned humans and preclinical models, including AChE inhibition, neuronal epileptiform hyperexcitation, and increased neuronal death. Here, we investigated in vivo the consequences of acute DFP exposure on microglia morphology and behaviour, and on the expression of a set of pro- and anti-inflammatory cytokines. We also used a genetic method of microglial ablation to evaluate the role in the OP-induced neuropathology. We first showed that DFP intoxication rapidly induced deep microglial phenotypic remodelling resembling that seen in M1-type activated macrophages and characterized by an amoeboid morphology, reduced branching, and increased mobility. DFP intoxication also caused massive expression of genes encoding pro-inflammatory cytokines Il1ß, Tnfα, Il8, and to a lesser extent, immuno-modulatory cytokine Il4, suggesting complex microglial reprogramming that included neuroinflammatory activities. Finally, microglia-depleted larvae were instrumental in showing that microglia were major actors in DFP-induced neuroinflammation and, more importantly, that OP-induced neuronal hyperactivation was markedly reduced in larvae fully devoid of microglia. DFP poisoning rapidly triggered massive microglia-mediated neuroinflammation, probably as a result of DFP-induced neuronal hyperexcitation, which in turn further exacerbated neuronal activation. Microglia are thus a relevant therapeutic target, and identifying substances reducing microglial activation could add efficacy to existing OP antidote cocktails.


Subject(s)
Isoflurophate , Organophosphate Poisoning , Acetylcholinesterase/metabolism , Animals , Antidotes , Brain/metabolism , Cholinesterase Inhibitors/pharmacology , Cytokines/metabolism , Humans , Isoflurophate/metabolism , Isoflurophate/toxicity , Microglia/metabolism , Neuroinflammatory Diseases , Organophosphate Poisoning/drug therapy , Organophosphate Poisoning/etiology , Organophosphate Poisoning/metabolism , Organophosphates/metabolism , Rats , Rats, Sprague-Dawley , Zebrafish/metabolism
3.
Toxicol Appl Pharmacol ; 416: 115466, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33631229

ABSTRACT

Soman, an organophosphorus (OP) compound, disrupts nervous system function through inactivation of acetylcholinesterase (AChE), the enzyme that breaks down acetylcholine at synapses. Left untreated, a state of prolonged seizure activity (status epilepticus, SE) is induced, causing widespread neuronal damage and associated cognitive and behavioral impairments. Previous research demonstrated that therapeutic stimulation of A1 adenosine receptors (A1ARs) can prevent or terminate soman-induced seizure. This study examined the ability of three potent A1AR agonists to provide neuroprotection and, ultimately, prevent observable cognitive and behavioral deficits following exposure to soman. Sprague Dawley rats were challenged with a seizure-inducing dose of soman (1.2 x LD50) and treated 1 min later with one of the following A1AR agonists: (6)-Cyclopentyladenosine (CPA), 2-Chloro-N6-cyclopentyladenosine (CCPA) or N-bicyclo(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (cdENBA). An active avoidance shuttle box task was used to evaluate locomotor responses to aversive stimuli at 3, 7 and 14 days post-exposure. Animals treated with CPA, CCPA or cdENBA demonstrated a higher number of avoidance responses and a faster reaction to the aversive stimulus than the soman/saline control group across all three sessions. Findings suggest that A1AR agonism is a promising neuroprotective countermeasure, capable of preventing the long-term deficits in learning and memory that are characteristic of soman intoxication.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Anticonvulsants/pharmacology , Brain/drug effects , Neuroprotective Agents/pharmacology , Organophosphate Poisoning/prevention & control , Receptor, Adenosine A1/drug effects , Seizures/prevention & control , Soman , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Deoxyadenosines/pharmacology , Disease Models, Animal , Male , Organophosphate Poisoning/etiology , Organophosphate Poisoning/metabolism , Organophosphate Poisoning/physiopathology , Rats, Sprague-Dawley , Receptor, Adenosine A1/metabolism , Seizures/chemically induced , Seizures/metabolism , Seizures/pathology
4.
Int J Legal Med ; 135(4): 1437-1447, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33987742

ABSTRACT

Organophosphorus pesticides (OPS) are widely used in the world, and many poisoning cases were caused by them. Phorate intoxication is especially common in China. However, there are currently few methods for discriminating phorate poisoning death from phorate exposure after death and interpretation of false-positive results due to the lack of effective biomarkers. In this study, we investigated the metabonomics of rat plasma at different dose levels of acute phorate intoxication using ultra-performance liquid chromatography quadrupole-time of flight mass spectrometry (UPLC-Q-TOF-MS) analysis. A total of 11 endogenous metabolites were significantly changed in the groups exposed to phorate at LD50 level and three times of LD50 (3LD50) level compared with the control group, which could be potential biomarkers of acute phorate intoxication. Plasma metabonomics analysis showed that diethylthiophosphate (DETP) could be a useful biomarker of acute phorate intoxication. The levels of uric acid, acylcarnitine, succinate, gluconic acid, and phosphatidylcholine (PC) (36:2) were increased, while pyruvate level was decreased in all groups exposed to phorate. The levels of ceramides (Cer) (d 18:0/16:0), palmitic acid, and lysophosphatidylcholine (lysoPC) (18:1) were only changed after 3LD50 dosage. The results of this study indicate that the dose-dependent relationship exists between metabolomic profile change and toxicities associated with apoptosis, fatty acid metabolism disorder, energy metabolism disorder especially tricarboxylic acid (TCA) cycle, as well as liver, kidney, and nervous system functions after acute exposure of phorate. This study shows that metabonomics is a useful tool in identifying biomarkers for the forensic toxicology study of phorate poisoning.


Subject(s)
Metabolome , Metabolomics , Organophosphate Poisoning/blood , Organophosphate Poisoning/metabolism , Phorate/blood , Phorate/metabolism , Animals , Biomarkers/blood , Biomarkers/metabolism , Chromatography, High Pressure Liquid , Disease Models, Animal , Lethal Dose 50 , Mass Spectrometry , Rats
5.
Neurobiol Dis ; 133: 104431, 2020 01.
Article in English | MEDLINE | ID: mdl-30905768

ABSTRACT

Current medical countermeasures for organophosphate (OP)-induced status epilepticus (SE) are not effective in preventing long-term morbidity and there is an urgent need for improved therapies. Rat models of acute intoxication with the OP, diisopropylfluorophosphate (DFP), are increasingly being used to evaluate therapeutic candidates for efficacy in mitigating the long-term neurologic effects associated with OP-induced SE. Many of these therapeutic candidates target neuroinflammation and oxidative stress because of their implication in the pathogenesis of persistent neurologic deficits associated with OP-induced SE. Critical to these efforts is the rigorous characterization of the rat DFP model with respect to outcomes associated with acute OP intoxication in humans, which include long-term electroencephalographic, neurobehavioral, and neuropathologic effects, and their temporal relationship to neuroinflammation and oxidative stress. To address these needs, we examined a range of outcomes at later times post-exposure than have previously been reported for this model. Adult male Sprague-Dawley rats were given pyridostigmine bromide (0.1 mg/kg, im) 30 min prior to administration of DFP (4 mg/kg, sc), which was immediately followed by atropine sulfate (2 mg/kg, im) and pralidoxime (25 mg/kg, im). This exposure paradigm triggered robust electroencephalographic and behavioral seizures that rapidly progressed to SE lasting several hours in 90% of exposed animals. Animals that survived DFP-induced SE (~70%) exhibited spontaneous recurrent seizures and hyperreactive responses to tactile stimuli over the first 2 months post-exposure. Performance in the elevated plus maze, open field, and Pavlovian fear conditioning tests indicated that acute DFP intoxication reduced anxiety-like behavior and impaired learning and memory at 1 and 2 months post-exposure in the absence of effects on general locomotor behavior. Immunohistochemical analyses revealed significantly increased expression of biomarkers of reactive astrogliosis, microglial activation and oxidative stress in multiple brain regions at 1 and 2 months post-DFP, although there was significant spatiotemporal heterogeneity across these endpoints. Collectively, these data largely support the relevance of the rat model of acute DFP intoxication as a model for acute OP intoxication in the human, and support the hypothesis that neuroinflammation and/or oxidative stress represent potential therapeutic targets for mitigating the long-term neurologic sequelae of acute OP intoxication.


Subject(s)
Brain , Disease Models, Animal , Isoflurophate/toxicity , Neurotoxicity Syndromes , Oxidative Stress/drug effects , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Organophosphate Poisoning/metabolism , Organophosphate Poisoning/pathology , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced
6.
Annu Rev Pharmacol Toxicol ; 57: 309-327, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28061690

ABSTRACT

Originally, organophosphorus (OP) toxicology consisted of acetylcholinesterase inhibition by insecticides and chemical threat agents acting as phosphorylating agents for serine in the catalytic triad, but this is no longer the case. Other serine hydrolases can be secondary OP targets, depending on the OP structure, and include neuropathy target esterase, lipases, and endocannabinoid hydrolases. The major OP herbicides are glyphosate and glufosinate, which act in plants but not animals to block aromatic amino acid and glutamine biosynthesis, respectively, with safety for crops conferred by their expression of herbicide-tolerant targets and detoxifying enzymes from bacteria. OP fungicides, pharmaceuticals including calcium retention agents, industrial chemicals, and cytochrome P450 inhibitors act by multiple noncholinergic mechanisms, often with high potency and specificity. One type of OP-containing fire retardant forms a highly toxic bicyclophosphate γ-aminobutyric acid receptor antagonist upon combustion. Some OPs are teratogenic, mutagenic, or carcinogenic by known mechanisms that can be avoided as researchers expand knowledge of OP chemistry and toxicology for future developments in bioregulation.


Subject(s)
Insecticides/toxicity , Occupational Exposure/prevention & control , Organophosphate Poisoning/prevention & control , Xenobiotics/toxicity , Animals , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/therapeutic use , Herbicides/chemistry , Herbicides/metabolism , Herbicides/toxicity , Humans , Insecticides/chemistry , Insecticides/metabolism , Organophosphate Poisoning/metabolism , Xenobiotics/chemistry , Xenobiotics/metabolism
7.
Anal Bioanal Chem ; 410(6): 1735-1748, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29313079

ABSTRACT

There is a growing interest in biological models to investigate the effect of neurotransmitter dysregulation on the structure and function of the central nervous system (CNS) at different stages of development. Zebrafish, a vertebrate model increasingly used in neurobiology and neurotoxicology, shares the common neurotransmitter systems with mammals, including glutamate, GABA, glycine, dopamine, norepinephrine, epinephrine, serotonin, acetylcholine, and histamine. In this study, we have evaluated the performance of liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the multiresidue determination of neurotransmitters and related metabolites. In a first step, ionization conditions were tested in positive electrospray mode and optimum fragmentation patterns were determined to optimize two selected reaction monitoring (SRM) transitions. Chromatographic conditions were optimized considering the chemical structure and chromatographic behavior of the analyzed compounds. The best performance was obtained with a Synergy Polar-RP column, which allowed the separation of the 38 compounds in 30 min. In addition, the performance of LC-MS/MS was studied in terms of linearity, sensitivity, intra- and inter-day precision, and overall robustness. The developed analytical method was able to quantify 27 of these neurochemicals in zebrafish chemical models for mild (P1), moderate (P2), and severe (P3) acute organophosphorus poisoning (OPP). The results show a general depression of synaptic-related neurochemicals, including the excitatory and inhibitory amino acids, as well as altered phospholipid metabolism, with specific neurochemical profiles associated to the different grades of severity. These results confirmed that the developed analytical method is a new tool for neurotoxicology research using the zebrafish model.


Subject(s)
Chromatography, High Pressure Liquid/methods , Organophosphate Poisoning/diagnosis , Tandem Mass Spectrometry/methods , Zebrafish , Acetylcholine/analysis , Acetylcholine/metabolism , Animals , Disease Models, Animal , Dopamine/analysis , Dopamine/metabolism , Epinephrine/analysis , Epinephrine/metabolism , Glutamic Acid/analysis , Glutamic Acid/metabolism , Glycine/analysis , Glycine/metabolism , Histamine/analysis , Histamine/metabolism , Humans , Neurotransmitter Agents/analysis , Neurotransmitter Agents/metabolism , Norepinephrine/analysis , Norepinephrine/metabolism , Organophosphate Poisoning/metabolism , Serotonin/analysis , Serotonin/metabolism , Zebrafish/metabolism , gamma-Aminobutyric Acid/analysis , gamma-Aminobutyric Acid/metabolism
8.
Toxicol Ind Health ; 34(2): 110-118, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29415640

ABSTRACT

Malathion can be ingested, inhaled, or absorbed through the skin, but acute toxicity is maximized when administered orally. Intravenous lipid emulsion (ILE) treatment is used as a new therapeutic method in cases of systemic toxicity caused by some lipid soluble agents. This study aimed to examine the potential treatment effect of ILE on rat lung tissue in a toxicokinetic model of malathion exposure. Twenty-one adult Wistar albino rats were randomly divided into three equal groups. The groups were organized as group I (control), group II (malathion), and group III (malathion + ILE treatment). Malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were evaluated in lung tissues. Immunohistochemical and Western blot analyses were performed to determine the bax, bcl-2, and caspase-3 expression levels. Tissue GSH-Px and SOD activities were decreased and MDA levels were increased in the malathion group. ILE administration increased GSH-Px and SOD activity and decreased MDA levels compared to the malathion group. Furthermore, expression of bax, bcl-2, and caspase-3 significantly increased in the malathion group, and ILE infusion reduced these expression levels. The present study revealed that acute oral malathion administration increased oxidative stress and apoptosis in the lung tissue of rats. ILE infusion prevented oxidative stress and decreased the deleterious effects of malathion. Taken together, the findings of our study suggest that lipid emulsion infusion has treatment efficacy on malathion-induced lung toxicity.


Subject(s)
Apoptosis/drug effects , Fat Emulsions, Intravenous/therapeutic use , Insecticides/toxicity , Lung/drug effects , Malathion/toxicity , Organophosphate Poisoning/therapy , Oxidative Stress/drug effects , Administration, Oral , Animals , Apoptosis Regulatory Proteins/metabolism , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/toxicity , Immunohistochemistry , Insecticides/administration & dosage , Lipid Peroxidation/drug effects , Lung/metabolism , Lung/pathology , Malathion/administration & dosage , Malondialdehyde/metabolism , Organophosphate Poisoning/etiology , Organophosphate Poisoning/metabolism , Organophosphate Poisoning/pathology , Oxidoreductases/metabolism , Random Allocation , Rats , Rats, Wistar , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Toxicokinetics
9.
Toxicol Ind Health ; 34(2): 69-82, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29141517

ABSTRACT

In this study, we aimed to study the possible preventive effect of docosahexaenoic acid (DHA), a dietary omega-3 fatty acid, on toxicity caused by chlorpyrifos (CPF). Six groups of Sprague Dawley rats (200-250 g) consisting of equal numbers of males and females (n = 8) were assigned to study. The rats were orally given for 5 days. The control group was administered pure olive oil, which was the vehicle for CPF. The CPF challenge groups were administered oral physiological saline, pure olive oil, or DHA (50, 100 and 400 mg/kg dosages) for 5 days. The animals were weighed on the sixth day and then administered CPF (279 mg/kg, subcutaneously). The rats were weighed again 24 h following CPF administration. The body temperatures and locomotor activities of the rats were also measured. Blood samples, brain and liver tissues were collected for biochemical, histopathological and immunohistochemical examinations. A comparison with the control group demonstrated that CPF administration increased malondialdehyde (MDA) levels in blood, brain and liver, while it reduced catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx) concentrations ( p < 0.05-0.001). Advanced oxidation protein products (AOPPs) increased only in the brain ( p < 0.001). DHA reduced these changes in MDA and AOPP values ( p < 0.05-0.001), while it increased CAT, SOD and GPx concentrations ( p < 0.05-0.001). Similarly, DHA prevented the decreases in body weight, body temperature and locomotor activities caused by CPF at 100 mg/kg and 400 mg/kg dosages ( p < 0.05-0.001). Similar to the physiological and biochemical changes, the histopathological damage scores, which increased with CPF ( p < 0.05-0.01), decreased at all three dosages of DHA ( p < 0.05-0.01). Our findings suggest that DHA, by supporting the antioxidant mechanism, reduces toxicity caused by CPF.


Subject(s)
Antioxidants/therapeutic use , Chlorpyrifos/toxicity , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Insecticides/toxicity , Organophosphate Poisoning/prevention & control , Oxidative Stress/drug effects , Animals , Antioxidants/administration & dosage , Behavior, Animal/drug effects , Biomarkers/blood , Biomarkers/metabolism , Body Temperature Regulation/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Chlorpyrifos/administration & dosage , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/toxicity , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/therapeutic use , Fatty Acids, Omega-3/administration & dosage , Female , Injections, Subcutaneous , Insecticides/administration & dosage , Liver/drug effects , Liver/metabolism , Liver/pathology , Locomotion/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Organophosphate Poisoning/blood , Organophosphate Poisoning/metabolism , Organophosphate Poisoning/pathology , Rats, Sprague-Dawley , Weight Loss/drug effects
10.
J Assoc Physicians India ; 66(8): 18-22, 2018 Aug.
Article in English | MEDLINE | ID: mdl-31324078

ABSTRACT

BACKGROUND: Organophosphorus insecticides (OPI) are one of the most extensively used classes of insecticides. Huge scientific body of evidence suggests that OPI exposure is a major toxicological threat that may affect human and animal health because of their various toxicities such as neurotoxicity, endocrine toxicity, immunotoxicity, reproductive toxicity, genotoxicity and ability to induce organ damage, alterations in cellular oxidative balance and disrupt glucose homeostasis. Mortality among organophosphorous (OP) poisoning patients despite advancements in its management is of concern.Of the various contributing factors,extremes and fluctuation in the glycemic status is a well documented parameter affecting the outcomes in critical illness although studies with respect to OP poisoning are deficient. All varieties of glycemic changes from hypoglycemia to hyperglycemia and ketoacidosis in OP poisoning along with other toxicological effects are reported,studies corroborating these findings are only few. The present endeavor was undertaken to study various glycemic changes in acute OP poisoning and it's bearing on clinical severity and clinical outcome. AIMS AND OBJECTIVES: 1. To assess the glycemic status by estimating random blood glucose level at the time of admission in cases of acute organophosphorous poisoning 2. To assess severity of the poisoning with various poisoning scales(PSS and POP) and level of serum pseudocholinesterase. 3. To correlate the documented blood glucose level with the severity and clinical outcome. METHOD: A prospective analytical study of 100 patients with diagnosed acute poisoning, above the age of 18 years, non diabetic, with no history of mixed poisoning or condition affecting blood glucose levels and fulfilling the inclusion and exclusion criterias was done over a period of one year. The glycemic status at the time of presentation was documented and the patients were grouped into hypoglycemics, euglycemics and hyperglycemics and the same was correlated with the severity and clinical outcome using descriptive statistics, association and test of significance using MedCalc. RESULTS: A prospective analytical study of 100 patients of acute organophosphate poisoning was done and on the basis of blood glucose levels at the time of presentation were further categorised into hypoglycemics (37%), euglycemics (52%) hyperglycaemic (11%). The outcome in terms of mortality was 59.45%,9.6% and 63.63% in the respective groups. The ventilator requirements among the three groups were 94.59%,53.84% and 100% respectively. Chisquare test to study the association between the presentation Random Blood Glucose (RBG) and the established Peradeniya Organophosphorous Poisoning Scale (POP) (Table 1) and Poisoning Severity Scale (PSS) (Table 2) revealed the study to be statistically significant (p value= 0.001)indicating both the extremes of glycemic status are associated with higher clinical severity and poorer outcomes. CONCLUSION: We conclude that the glycemic status at the time of presentation in acute organophosphate poisoning patients is a simple, cheap, reliable marker in guiding the clinical severity and outcome when considered with clinical severity scores and S.ChE in a resource limited country like India.


Subject(s)
Hyperglycemia , Insecticides/poisoning , Organophosphate Poisoning/diagnosis , Humans , India , Organophosphate Poisoning/metabolism , Prospective Studies
11.
J Neurochem ; 142 Suppl 2: 26-40, 2017 08.
Article in English | MEDLINE | ID: mdl-28542985

ABSTRACT

Organophosphorus agents (OPs) irreversibly inhibit acetylcholinesterase (AChE) causing a major cholinergic syndrome. The medical counter-measures of OP poisoning have not evolved for the last 30 years with carbamates for pretreatment, pyridinium oximes-based AChE reactivators, antimuscarinic drugs and neuroprotective benzodiazepines for post-exposure treatment. These drugs ensure protection of peripheral nervous system and mitigate acute effects of OP lethal doses. However, they have significant limitations. Pyridostigmine and oximes do not protect/reactivate central AChE. Oximes poorly reactivate AChE inhibited by phosphoramidates. In addition, current neuroprotectants do not protect the central nervous system shortly after the onset of seizures when brain damage becomes irreversible. New therapeutic approaches for pre- and post-exposure treatments involve detoxification of OP molecules before they reach their molecular targets by administrating catalytic bioscavengers, among them phosphotriesterases are the most promising. Novel generation of broad spectrum reactivators are designed for crossing the blood-brain barrier and reactivate central AChE. This is an article for the special issue XVth International Symposium on Cholinergic Mechanisms.


Subject(s)
Acetylcholinesterase/metabolism , Blood-Brain Barrier/drug effects , Cholinesterase Inhibitors/pharmacology , Cholinesterase Reactivators/metabolism , Neuroprotective Agents/pharmacology , Organophosphate Poisoning/drug therapy , Animals , Blood-Brain Barrier/metabolism , Cholinesterase Reactivators/pharmacology , Humans , Organophosphate Poisoning/metabolism
12.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1858-1866, 2017 07.
Article in English | MEDLINE | ID: mdl-27262357

ABSTRACT

In the previous study, we demonstrated that dichlorvos induces oxidative stress in dopaminergic neuronal cells and subsequent caspase activation mediates apoptosis. In the present study, we evaluated the effect and mechanism of dichlorvos induced oxidative stress on cell cycle activation in NGF-differentiated PC12 cells. Dichlorvos exposure resulted in oxidative DNA damage along with activation of cell cycle machinery in differentiated PC12 cells. Dichlorvos exposed cells exhibited an increased expression of p53, cyclin-D1, pRb and decreased expression of p21suggesting a re-entry of differentiated cells into the cell cycle. Cell cycle analysis of dichlorvos exposed cells revealed a reduction of cells in the G0/G1 phase of the cell cycle (25%), and a concomitant increase of cells in S phase (30%) and G2/M phase (43.3%) compared to control PC12 cells. Further, immunoblotting of cytochrome c, Bax, Bcl-2 and cleaved caspase-3 revealed that dichlorvos induces a caspase-dependent cell death in PC12 cells. These results suggest that Dichlorvos exposure has the potential to generate oxidative stress which evokes activation of cell cycle machinery leading to apoptotic cell death via cytochrome c release from mitochondria and subsequent caspase-3 activation in differentiated PC12 cells.


Subject(s)
Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dichlorvos/adverse effects , Dopaminergic Neurons/metabolism , Neurodegenerative Diseases/metabolism , Organophosphate Poisoning/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dichlorvos/pharmacology , Dopaminergic Neurons/pathology , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Organophosphate Poisoning/genetics , Organophosphate Poisoning/pathology , Oxidative Stress/drug effects , PC12 Cells , Rats
13.
Biochim Biophys Acta ; 1852(4): 651-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25486268

ABSTRACT

Disorders affecting the presynaptic, synaptic, and postsynaptic portions of the neuromuscular junction arise from various mechanisms in children and adults, including acquired autoimmune or toxic processes as well as genetic mutations. Disorders include autoimmune myasthenia gravis associated with acetylcholine receptor, muscle specific kinase or Lrp4 antibodies, Lambert-Eaton myasthenic syndrome, nerve terminal hyperexcitability syndromes, Guillain Barré syndrome, botulism, organophosphate poisoning and a number of congenital myasthenic syndromes. This review focuses on the various molecular and pathophysiological mechanisms of these disorders, characterization of which has been crucial to the development of treatment strategies specific for each pathogenic mechanism. In the future, further understanding of the underlying processes may lead to more effective and targeted therapies of these disorders. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Subject(s)
Botulism , Guillain-Barre Syndrome , Lambert-Eaton Myasthenic Syndrome , Myasthenia Gravis , Organophosphate Poisoning , Adolescent , Adult , Autoantibodies/immunology , Autoantibodies/metabolism , Botulism/genetics , Botulism/immunology , Botulism/metabolism , Botulism/pathology , Child , Child, Preschool , Guillain-Barre Syndrome/genetics , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/metabolism , Guillain-Barre Syndrome/pathology , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/immunology , LDL-Receptor Related Proteins/metabolism , Lambert-Eaton Myasthenic Syndrome/genetics , Lambert-Eaton Myasthenic Syndrome/immunology , Lambert-Eaton Myasthenic Syndrome/metabolism , Lambert-Eaton Myasthenic Syndrome/pathology , Myasthenia Gravis/genetics , Myasthenia Gravis/immunology , Myasthenia Gravis/metabolism , Myasthenia Gravis/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/immunology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Organophosphate Poisoning/genetics , Organophosphate Poisoning/immunology , Organophosphate Poisoning/metabolism , Organophosphate Poisoning/pathology , Receptors, Cholinergic/genetics , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism
14.
Arch Toxicol ; 90(12): 2831-2859, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27582056

ABSTRACT

Irreversible inhibition of acetylcholinesterase (AChE) by organophosphates leads to many failures in living organism and ultimately in death. Organophosphorus compounds developed as nerve agents such as tabun, sarin, soman, VX and others belong to the most toxic chemical warfare agents and are one of the biggest threats to the modern civilization. Moreover, misuse of nerve agents together with organophosphorus pesticides (e.g. malathion, paraoxon, chlorpyrifos, etc.) which are annually implicated in millions of intoxications and hundreds of thousand deaths reminds us of insufficient protection against these compounds. Basic treatments for these intoxications are based on immediate administration of atropine and acetylcholinesterase reactivators which are currently represented by mono- or bis-pyridinium aldoximes. However, these antidotes are not sufficient to ensure 100 % treatment efficacy even they are administered immediately after intoxication, and in general, they possess several drawbacks. Herein, we have reviewed new efforts leading to the development of novel reactivators and proposition of new promising strategies to design novel and effective antidotes. Structure-activity relationships and biological activities of recently proposed acetylcholinesterase reactivators are discussed and summarized. Among further modifications of known oximes, the main attention has been paid to dual binding site ligands of AChE as the current mainstream strategy. We have also discussed new chemical entities as potential replacement of oxime functional group.


Subject(s)
Acetylcholinesterase/chemistry , Antidotes/pharmacology , Cholinesterase Reactivators/pharmacology , Drug Design , Organophosphate Poisoning/drug therapy , Organophosphorus Compounds/antagonists & inhibitors , Pesticides/antagonists & inhibitors , Acetylcholinesterase/metabolism , Animals , Antidotes/chemistry , Antidotes/therapeutic use , Binding Sites , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/toxicity , Cholinesterase Reactivators/chemistry , Cholinesterase Reactivators/therapeutic use , Humans , Ligands , Molecular Conformation , Molecular Structure , Nerve Agents/chemistry , Nerve Agents/toxicity , Organophosphate Poisoning/etiology , Organophosphate Poisoning/metabolism , Organophosphorus Compounds/toxicity , Pesticides/toxicity , Protein Conformation , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacology , Structure-Activity Relationship
15.
Bioconjug Chem ; 26(8): 1753-8, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26121420

ABSTRACT

Acetylcholinesterase (AChE) is the physiological target of organophosphate nerve agent compounds. Currently, the development of a formulation for prophylactic administration of cholinesterases as bioscavengers in established risk situations of exposure to nerve agents is the incentive for many efforts. While cholinesterase bioscavengers were found to be highly effective in conferring protection against nerve agent exposure in animal models, their therapeutic use is complicated by short circulatory residence time. To create a bioscavenger with prolonged plasma half-life, compatible with biotechnological production and purification, a chimeric recombinant molecule of HuAChE coupled to the Fc region of human IgG1 was designed. The novel fusion protein, expressed in cultured cells under optimized conditions, maintains its full enzymatic activity, at levels similar to those of the recombinant AChE enzyme. Thus, this novel fusion product retained its binding affinity toward BW284c5 and propidium, and its bioscavenging reactivity toward the organophosphate-AChE inhibitors sarin and VX. Furthermore, when administered to mice, AChE-Fc exhibits exceptional circulatory residence longevity (MRT of 6000 min), superior to any other known cholinesterase-based recombinant bioscavengers. Owing to its optimized pharmacokinetic performance, high reactivity toward nerve agents, and ease of production, AChE-Fc emerges as a promising next-generation organophosphate bioscavenger.


Subject(s)
Acetylcholinesterase/pharmacokinetics , Immunoglobulin Fc Fragments/chemistry , Organophosphate Poisoning/drug therapy , Organophosphorus Compounds/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Acetylcholinesterase/chemistry , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Half-Life , Humans , Immunoglobulin Fc Fragments/metabolism , Mice , Mice, Inbred ICR , Organophosphate Poisoning/metabolism , Recombinant Fusion Proteins/chemistry , Tissue Distribution
16.
Altern Ther Health Med ; 21(3): 54-62, 2015.
Article in English | MEDLINE | ID: mdl-26026145

ABSTRACT

Detoxification is a vital cellular task that, if lacking, can lead to early morbidity and mortality. The process of detoxification involves the mobilization, biotransformation, and elimination of toxicants of exogenous and endogenous origin. This article discusses the phase I and phase II detoxification and biotransformation pathways and promotes using food to support these highly complex processes. The author identifies the comprehensive elimination diet as a useful therapeutic tool for clinicians and patients to use to achieve detoxification. Using this diet, the patient removes the most common allergenic foods and beverages from the diet and replaces them with nonallergenic choices for a period of 4 wk, gradually adding back the eliminated foods and observing their effects. Another effective clinical tool that the author discusses is the detox-focused core food plan, which identifies the variety of foods required to supply key nutrients that can maximize the effectiveness of detoxification. Finally, the author provides a case study in which these tools were used to help a patient suffering from major, debilitating illnesses that resulted from exposure to malathion, including severe vomiting and diarrhea, headaches, night sweats, severe arthralgias and myalgias, episcleritis, and shortness of breath. The article details the interventions used and the clinical results (ie, successful resolution of most issues after 3 mo).


Subject(s)
Diet Therapy , Inactivation, Metabolic , Liver/metabolism , Biotransformation , Female , Humans , Liver/enzymology , Malathion/pharmacokinetics , Middle Aged , Models, Biological , Organophosphate Poisoning/metabolism
17.
Acta Medica (Hradec Kralove) ; 58(4): 128-30, 2015.
Article in English | MEDLINE | ID: mdl-26960825

ABSTRACT

BACKGROUND: The method of continual determination of the rat blood cholinesterase activity was developed to study the changes of the blood cholinesterases following different intervetions. AIMS: The aim of this study is registration of cholinesterase activity in the rat blood and its changes to demonstrate detoxification capacity of rats to inactivate sarin or VX in vivo. METHODS: The groups of female rats were premedicated (ketamine and xylazine) and cannulated to a. femoralis. Continual blood sampling (0.02 ml/min) and monitoring of the circulating blood cholinesterase activity were performed. Normal activity was monitored 1-2 min and then the nerve agent was administered i.m. (2×LD50). Using different time intervals of the leg compression and relaxation following the agent injection, cholinesterase activity was monitored and according to the inhibition obtained, detoxification capacity was assessed. RESULTS: Administration of sarin to the leg, then 1 and 5 min compression and 20 min later relaxation showed that further inhibition in the blood was not observed. On the other hand, VX was able to inhibit blood cholinesterases after this intervention. CONCLUSIONS: The results demonstrated that sarin can be naturally detoxified on the contrary to VX. Described method can be used as model for other studies dealing with changes of cholinesterases in the blood following different factors.


Subject(s)
Cholinesterase Inhibitors/pharmacokinetics , Cholinesterases/metabolism , Organophosphate Poisoning/metabolism , Organothiophosphorus Compounds/pharmacokinetics , Sarin/pharmacokinetics , Animals , Cholinesterase Inhibitors/toxicity , Female , Inactivation, Metabolic , Organothiophosphorus Compounds/toxicity , Rats , Rats, Wistar , Sarin/toxicity
18.
Am J Emerg Med ; 32(7): 693-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24856742

ABSTRACT

Organophosphate poisoning is a serious clinical entity and considerable morbidity and mortality. Several factors have been identified to predict outcomes of organophosphate poisoning. Organophosphates are lipophilic and therefore predicted to have a large volume of distribution and to rapidly distribute into tissue and fat. Thus, toxic effects of organophosphate would be expected to last longer in obese patients. We investigated the relationship between obesity and clinical course in 112 acute organophosphate-poisoned patients from an initial medical record review of 234 patients. One hundred twenty-two patients were excluded: 6 were children, 14 had an uncertain history of exposure and of uncertain agent, 10 were transferred to another hospital, 67 were discharged from the emergency department because their toxicity was mild, 21 had carbamate poisoning, and 4 did not have height or weight checked. Clinical features, body mass index, Glasgow Coma Scale, laboratory findings, serum cholinesterase activity, electrocardiogram finding, management, and outcomes were examined. The lipid solubility of the implicated organophosphate was characterized by its octanol/water coefficient. Forty of 112 patients were obese. Obese patients who were poisoned by high lipophilicity organophosphate compounds had a need for longer use of mechanical ventilation, intensive care unit care, and total length of admission. Body mass index can provide a guide to physicians in predicting clinical course and management in organophosphate-poisoned patients.


Subject(s)
Atropine/therapeutic use , Cholinesterase Reactivators/therapeutic use , Muscarinic Antagonists/therapeutic use , Obesity/metabolism , Organophosphate Poisoning/therapy , Organophosphates/metabolism , Pralidoxime Compounds/therapeutic use , Adult , Aged , Body Mass Index , Cohort Studies , Female , Glasgow Coma Scale , Humans , Intensive Care Units , Length of Stay , Male , Middle Aged , Obesity/complications , Organophosphate Poisoning/complications , Organophosphate Poisoning/metabolism , Prognosis , Respiration, Artificial , Retrospective Studies , Severity of Illness Index , Solubility
19.
Int J Toxicol ; 33(4): 307-318, 2014 07.
Article in English | MEDLINE | ID: mdl-24985121

ABSTRACT

The activities of different xenobiotic-metabolizing enzymes in liver subcellular fractions from Wistar rats exposed to a glyphosate (GLP)-based herbicide (Roundup full II) were evaluated in this work. Exposure to the herbicide triggered protective mechanisms against oxidative stress (increased glutathione peroxidase activity and total glutathione levels). Liver microsomes from both male and female rats exposed to the herbicide had lower (45%-54%, P < 0.01) hepatic cytochrome P450 (CYP) levels compared to their respective control animals. In female rats, the hepatic 7-ethoxycoumarin O-deethylase (a general CYP-dependent enzyme activity) was 57% higher (P < 0.05) in herbicide-exposed compared to control animals. Conversely, this enzyme activity was 58% lower (P < 0.05) in male rats receiving the herbicide. Lower (P < 0.05) 7-ethoxyresorufin O-deethlyase (EROD, CYP1A1/2 dependent) and oleandomycin triacetate (TAO) N-demethylase (CYP3A dependent) enzyme activities were observed in liver microsomes from exposed male rats. Conversely, in females receiving the herbicide, EROD increased (123%-168%, P < 0.05), whereas TAO N-demethylase did not change. A higher (158%-179%, P < 0.01) benzyloxyresorufin O-debenzylase (a CYP2B-dependent enzyme activity) activity was only observed in herbicide-exposed female rats. In herbicide-exposed rats, the hepatic S-oxidation of methimazole (flavin monooxygenase dependent) was 49% to 62% lower (P < 0.001), whereas the carbonyl reduction of menadione (a cytosolic carbonyl reductase-dependent activity) was higher (P < 0.05). Exposure to the herbicide had no effects on enzymatic activities dependent on carboxylesterases, glutathione transferases, and uridinediphospho-glucuronosyltransferases. This research demonstrated certain biochemical modifications after exposure to a GLP-based herbicide. Such modifications may affect the metabolic fate of different endobiotic and xenobiotic substances. The pharmacotoxicological significance of these findings remains to be clarified.


Subject(s)
Glycine/analogs & derivatives , Herbicides/toxicity , Liver/drug effects , Organophosphate Poisoning/enzymology , Oxidative Stress/drug effects , Water Pollutants, Chemical/toxicity , Xenobiotics/metabolism , 7-Alkoxycoumarin O-Dealkylase/antagonists & inhibitors , 7-Alkoxycoumarin O-Dealkylase/chemistry , 7-Alkoxycoumarin O-Dealkylase/metabolism , Animals , Carbonyl Reductase (NADPH)/chemistry , Carbonyl Reductase (NADPH)/metabolism , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/chemistry , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/chemistry , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2B1/chemistry , Cytochrome P-450 CYP2B1/metabolism , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Female , Glycine/administration & dosage , Glycine/toxicity , Herbicides/administration & dosage , Liver/enzymology , Liver/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Organophosphate Poisoning/metabolism , Oxygenases/antagonists & inhibitors , Oxygenases/metabolism , Random Allocation , Rats, Wistar , Sex Characteristics , Water Pollutants, Chemical/administration & dosage , Glyphosate
SELECTION OF CITATIONS
SEARCH DETAIL