Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Acta Biochim Pol ; 58(2): 193-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21666889

RESUMEN

A conjugate of pyridine-4-aldoxime and atropine (ATR-4-OX) was synthesized and its antidotal efficiency was tested in vitro on tabun- or paraoxon-inhibited acetylcholinesterase (AChE) of human erythrocytes as well as in vivo using soman-, tabun- or paraoxon-poisoned mice. Its genotoxic profile was assessed on human lymphocytes in vitro and was found acceptable for further research. ATR-4-OX showed very weak antidotal activity, inadequate for soman or tabun poisoning. Conversely, it was effective against paraoxon poisoning both in vitro and in vivo. All animals treated with 5 % or 25 % LD(50) doses of the new oxime survived after administration of 10.0 or 16.0 LD(50) doses of paraoxon, respectively. Based on the persistence of toxicity symptoms in mice, the atropine moiety had questionable effects in attenuating such symptoms. It appears that ATR-4-OX has a therapeutic effect related to the reactivation of phosphylated AChE, but not to receptor antagonization.


Asunto(s)
Antídotos/farmacología , Derivados de Atropina/farmacología , Inhibidores de la Colinesterasa/envenenamiento , Reactivadores de la Colinesterasa/farmacología , Intoxicación por Organofosfatos , Paraoxon/envenenamiento , Compuestos de Pralidoxima/farmacología , Soman/envenenamiento , Acetilcolinesterasa , Adulto , Animales , Derivados de Atropina/síntesis química , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Pruebas de Enzimas , Eritrocitos/efectos de los fármacos , Eritrocitos/enzimología , Humanos , Linfocitos/efectos de los fármacos , Masculino , Ratones , Organofosfatos , Compuestos de Pralidoxima/síntesis química
2.
Crit Care Med ; 39(4): 803-11, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21242797

RESUMEN

OBJECTIVES: Pralidoxime is an organic cation used as an antidote in addition to atropine to treat organophosphate poisoning. Pralidoxime is rapidly eliminated by the renal route and thus has limited action. The objectives of this work were as follows. 1) Study the role of organic cation transporters in the renal secretion of pralidoxime using organic cation transporter substrates (tetraethylammonium) and knockout mice (Oct1/2⁻/⁻; Oct3⁻/⁻). 2) Assess whether sustained high plasma concentrations increase pralidoxime antidotal activity toward paraoxon-induced respiratory toxicity. SETTING: INSERM U705, Faculté de Pharmacie, Université Paris Descartes, 4 Avenue de l'Observatoire, 75006 Paris, France. SUBJECTS: Rodents: Knockout mice (Oct1/2⁻/⁻; Oct3⁻/⁻) and Sprague-Dawley rats. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: In rats, the renal clearance of pralidoxime was 3.6-fold higher than the creatinine clearance. Pretreatment with tetraethylammonium (75 mg/kg) in rats or deficiencies in organic cation transporters 1 and 2 in mice (Oct1/2⁻/⁻) resulted in a significant increase in plasma pralidoxime concentrations. Lack of Oct3 did not alter plasma pralidoxime concentrations. The antidotal activity of pralidoxime (50 mg/kg intramuscularly) was longer and with greater effect, resulting in a return to normal values when administered to rats pretreated with tetraethylammonium. CONCLUSIONS: Pralidoxime is secreted in rats and mice by renal Oct1 and/or Oct2 but not by Oct3. Modulation of organic cation transporter activity increased the plasma pralidoxime concentrations and the antidotal effect of pralidoxime with sustained return within the normal range of respiratory variables in paraoxon-poisoned rats. These results suggest a promising approach in an animal model toward the increase in efficiency of pralidoxime. However, further studies are needed before these results are extended to human poisoning.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/efectos de los fármacos , Antídotos/uso terapéutico , Compuestos Organotiofosforados/envenenamiento , Compuestos de Pralidoxima/uso terapéutico , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/fisiología , Animales , Antídotos/farmacocinética , Insecticidas/envenenamiento , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Paraoxon/envenenamiento , Pletismografía Total , Compuestos de Pralidoxima/agonistas , Compuestos de Pralidoxima/farmacocinética , Compuestos de Amonio Cuaternario/farmacología , Ratas , Ratas Sprague-Dawley
3.
Toxicol Sci ; 116(2): 660-72, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20498006

RESUMEN

Empirical studies suggest that the antidotal effect of pralidoxime depends on plasma concentrations with therapeutic effects associated with concentrations above 4 mg/l. The purpose of this study was to determine the pharmacokinetic-toxicodynamic (PK-TD) relationships for the antidotal effect of pralidoxime on paraoxon-induced toxicity in rats. Diethylparaoxon inactivation of whole-blood cholinesterase activity was studied both in vitro and in male Sprague-Dawley rats. Toxin-induced respiratory effects were measured via whole-body plethysmography in control and pralidoxime-treated animals (50 mg/kg im injection). In the in vitro analysis, cholinesterase reactivation by pralidoxime in blood-poisoned diethylparaoxon (10nM) was proportional to the logarithm of drug concentrations. A mechanism-based TD model was developed, which well described the inhibition of cholinesterases by diethylparaoxon and reactivation with pralidoxime. The in vitro pralidoxime EC(50) was estimated to be 4.67 mg/l. Animals exposed to diethylparaoxon exhibited a decrease in respiratory rate and an increase in expiratory time, and pralidoxime treatment resulted in a rapid complete but transient (< 30 min) correction in respiratory toxicity. In contrast, there was a fast and total reactivation of blood cholinesterase activity over the 210-min study period. The in vitro TD model was extended to capture the time-course of in vivo pralidoxime antidotal effects, which explained the complex relationship between drug exposure and pharmacological response profile. This study provides insights into the role of oxime-rescue of paraoxon-induced toxicity, and the final PK-TD model might prove useful in optimizing the design and development of such therapy.


Asunto(s)
Antídotos/farmacología , Insecticidas/envenenamiento , Paraoxon/envenenamiento , Compuestos de Pralidoxima/farmacología , Respiración/efectos de los fármacos , Animales , Colinesterasas/sangre , Leucocitos/enzimología , Masculino , Modelos Biológicos , Compuestos de Pralidoxima/farmacocinética , Ratas , Ratas Sprague-Dawley
4.
Toxicology ; 272(1-3): 46-51, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20385200

RESUMEN

The therapy of organophosphorus compound (OP) poisoning is still a challenge to clinical toxicologists. To alleviate peripheral respiratory failure oximes, e.g. obidoxime and pralidoxime, are used to reactivate inhibited acetylcholinesterase (AChE) with the intention to restore the disturbed neuromuscular function. In severe human OP poisoning the persistence of poison may counteract effective reactivation by oximes. Therefore, the study was designed to investigate the effect of the clinically used oximes obidoxime, pralidoxime and the experimental compounds HI 6 and HLö 7 in the presence of different paraoxon concentrations. The mouse phrenic nerve-diaphragm preparation was used as a functional model. After washout of paraoxon remarkably low concentrations of obidoxime or HLö 7 were sufficient for restoration of paraoxon-impaired muscle force. In the presence of paraoxon, obidoxime was the most effective oxime and therapeutically used concentrations (10-20microM) were able to restore muscle function even in the presence of 1microM paraoxon. HLö 7 was less effective, but superior to HI 6 and pralidoxime. Generally, a reactivation of AChE to about 30-40% of normal was sufficient for restoration of muscle force. Thus, the data presented strongly support the administration of appropriately dosed oximes, preferably obidoxime, in paraoxon-poisoned patients to restore paraoxon-impaired muscle force.


Asunto(s)
Acetilcolinesterasa/metabolismo , Diafragma/metabolismo , Oximas/uso terapéutico , Paraoxon/envenenamiento , Fenómenos Físicos , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos , Cloruro de Obidoxima/uso terapéutico , Compuestos de Pralidoxima/uso terapéutico , Compuestos de Piridinio/uso terapéutico
5.
Toxicol Lett ; 189(1): 48-56, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19406220

RESUMEN

We recently showed in a rat model of dichromate-induced acute renal failure (ARF) that the elimination but not the distribution of pralidoxime was altered resulting in sustained plasma pralidoxime concentrations. The aim of this study was to compare the efficiency of pralidoxime in normal and acute renal failure rats against paraoxon-induced respiratory toxicity. Ventilation at rest was assessed using whole-body plethysmography after subcutaneous administration of either saline or paraoxon (50% of the LD(50)), in the control and ARF rats. Thirty minutes after administration of paraoxon, either saline or 50mg/kg of pralidoxime was administered intramuscularly. ARF had no significant effects on the ventilation at rest. The effects of paraoxon on respiration were not significantly different in the control and ARF group. Paraoxon increased the total time (T(TOT)), expiratory time (T(E)) and tidal volume (V(T)), and decreased the respiratory frequency (f). In paraoxon-poisoned rats with normal renal function, pralidoxime had a significant but transient effect regarding the T(TOT) and V(T) (p<0.05). In the ARF group, the same dose of pralidoxime significantly decreased the T(TOT), T(E), and V(T) and increased f during 90 min (p<0.01). In conclusion, pralidoxime had partial and transient effects towards paraoxon-induced respiratory toxicity in control rats; and a complete and sustained correction in ARF rats.


Asunto(s)
Lesión Renal Aguda/sangre , Antídotos/uso terapéutico , Insecticidas/envenenamiento , Paraoxon/envenenamiento , Compuestos de Pralidoxima/uso terapéutico , Trastornos Respiratorios/prevención & control , Animales , Antídotos/farmacocinética , Antídotos/farmacología , Modelos Animales de Enfermedad , Dosificación Letal Mediana , Masculino , Pletismografía Total , Compuestos de Pralidoxima/sangre , Compuestos de Pralidoxima/farmacología , Ratas , Ratas Sprague-Dawley , Respiración/efectos de los fármacos , Trastornos Respiratorios/sangre , Trastornos Respiratorios/inducido químicamente
6.
Biopharm Drug Dispos ; 28(3): 145-50, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17315239

RESUMEN

Immediate administration of antidotal treatment is crucial in severe organophosphate (OP) poisoning and the use of an open intravenous (i.v.) line might also be required. The state of casualties might prevent getting access to their veins. The bone injection gun (BIG) was established as a simple method for introducing an intraosseous (i.o.) line and could be applied while wearing a protective suit. The present study followed the pharmacokinetics of the anticonvulsive drug midazolam after i.o. administration in pigs compared with i.v. and the common intramuscular (i.m.) administration. A new method for monitoring midazolam concentrations in plasma was developed. Plasma concentrations following both i.v. and i.o. administrations peaked at 2 min post injection and only at 10 min following the i.m. route. In an antidotal treatment study against paraoxone poisoning, the anticonvulsive effect of midazolam appeared immediately following i.o. administration, while it took 5-10 min to exhibit a similar effect following i.m. administration. This study indicates that the use of i.o. administration after OP poisoning might provide the necessary fast response for rapid termination of convulsions. The BIG might offer a convenient method for treating casualties in the chemical arena by teams wearing full protective gear.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Antídotos/uso terapéutico , Midazolam/uso terapéutico , Intoxicación por Organofosfatos , Paraoxon/envenenamiento , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/farmacocinética , Antídotos/administración & dosificación , Antídotos/farmacocinética , Disponibilidad Biológica , Relación Dosis-Respuesta a Droga , Infusiones Intraóseas/instrumentación , Inyecciones Intramusculares/instrumentación , Inyecciones Intravenosas , Midazolam/administración & dosificación , Midazolam/farmacocinética , Intoxicación/tratamiento farmacológico , Convulsiones/inducido químicamente , Porcinos
7.
Toxicology ; 233(1-3): 209-13, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17250944

RESUMEN

Organophosphorus (OP) pesticides or nerve agents cause severe intoxication by inhibition of acetylcholinesterase, finally resulting in death due to respiratory failure. The phrenic nerve diaphragm preparation is considered as the classic model to investigate the effect of OP intoxications and oxime treatment at the neuromuscular junction. However, this preparation is unsuitable for larger species or for muscle strips from biopsies where no nerve is available for stimulation. An alternative technique is the indirect field stimulation of muscles containing intramuscular nerve branches only. The proposed method by Wolthuis et al. [Wolthuis, O.L., Vanwersch, R.A.P., Van Der Wiel, H.J., 1981. The efficacy of some bis-pyridinium oximes as antidotes to soman in isolated muscles of several species including man. Eur. J. Pharmacol. 70, 355-369] was modified and experimentally reevaluated in isolated mouse diaphragms. To confirm that electrical field stimulation technique induced muscle contraction only via the neuromuscular endplate the nicotinic antagonists pancuronium or d-tubocurarine (1microM) were given. In the presence of a nicotinic antagonist hardly any contraction was blocked after indirect field stimulation technique with very short pulses (5micros, <0.6A), in contrast to direct muscle stimulation (broader pulse width, or higher amplitude >0.6A). During paraoxon circumfusion (20min, 1micromol/l) muscle force generation by indirect stimulation was almost completely blocked. Restoration of paralyzed muscle function to 80% of initial values could be achieved after paraoxon wash out (20min) and circumfusion with obidoxime (1micromol/l, 20min). This data correspond quite well to data shown earlier when using conventional nerve stimulation techniques.


Asunto(s)
Contracción Muscular/efectos de los fármacos , Oximas/farmacología , Paraoxon/envenenamiento , Animales , Diafragma/efectos de los fármacos , Diafragma/inervación , Estimulación Eléctrica , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos , Bloqueantes Neuromusculares/farmacología , Pancuronio/farmacología , Tubocurarina/farmacología
8.
Toxicol Appl Pharmacol ; 219(2-3): 235-40, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17056080

RESUMEN

Recently, the FDA approved the medical use of oral pyridostigmine as prophylactic treatment of possible nerve agent exposure: the concept is to block the cholinesterase transitorily using the carbamate (pyridostigmine) in order to deny access to the active site of the enzyme to the irreversible inhibitor (nerve agent) on subsequent exposure. We have shown previously that tiapride is in vitro a weak inhibitor of acetylcholinesterase and that in rats administration of tiapride before the organophosphate paraoxon significantly decreases mortality. The purpose of the present study was to compare tiapride- and pyridostigmine-based pretreatment strategies, either alone or in combination with pralidoxime reactivation, by using a prospective, non-blinded study in a rat model of acute high-dose paraoxon exposure. Groups 1-6 received 1 microMol paraoxon (approximately LD75) groups 2-6 received in addition: G(2)50 microMol tiapride 30 min before paraoxonG(3)50 microMol tiapride 30 min before paraoxon and 50 microMol pralidoxime 1 min after paraoxon G41 microMol pyridostigmine 30 min before paraoxon G(5)1 microMol pyridostigmine 30 min before paraoxon and 50 microMol pralidoxime 1 min after paraoxon G(6)50 microMol pralidoxime 1 min after paraoxon. Mortality data were compared using Kaplan-Meier plots and logrank tests. Mortality is statistically significantly influenced by all treatment strategies. Tiapride pretreatment followed by pralidoxime treatment (G3) is aux par with pyridostigmine pretreatment followed by pralidoxime treatment (G5). Tiapride pretreatment only (G2) is inferior to pyridostigmine pretreatment only (G4). The best results are achieved with pyridostigmine pretreatment only or pralidoxime treatment only (G4 and G6).


Asunto(s)
Inhibidores de la Colinesterasa/uso terapéutico , Paraoxon/envenenamiento , Bromuro de Piridostigmina/uso terapéutico , Clorhidrato de Tiapamilo/uso terapéutico , Enfermedad Aguda , Animales , Unión Competitiva , Inhibidores de la Colinesterasa/administración & dosificación , Inhibidores de la Colinesterasa/farmacología , Colinesterasas/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Paraoxon/farmacocinética , Intoxicación/enzimología , Intoxicación/prevención & control , Bromuro de Piridostigmina/administración & dosificación , Bromuro de Piridostigmina/farmacología , Ratas , Ratas Wistar , Clorhidrato de Tiapamilo/administración & dosificación , Clorhidrato de Tiapamilo/farmacología
9.
Mol Cell Biochem ; 285(1-2): 79-86, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16479322

RESUMEN

INTRODUCTION: Accidental and suicidal exposures to organophosphorus compounds (OPC) are frequent. The inhibition of esterases by OPC leads to an endogenous ACh poisoning. Recently, the FDA approved, based on animal experiments, for military combat medical use oral pyridostigmine (PSTG) for pre-exposure treatment of soman; the concept is to block the cholinesterase reversibly using the carbamate pyridostigmine in order to deny access to the active site of the enzyme to the irreversible inhibitor (OPC) on subsequent exposure. We have shown previously that tiapride (TIA) is in vitro a weak inhibitor of AChE. We also have shown recently that in rats coadministration of TIA with the organophosphate paraoxon significantly decreases mortality without having an impact on red blood cell cholinesterase (RBC-AChE) activity. PURPOSE OF THE STUDY: To establish in a prospective, non-blinded study in a rat model of acute high dose OPC (paraoxon; POX) exposure the ideal point in time for TIA pre-treatment administration and to correlate it with measured TIA plasma levels. MATERIAL AND METHODS: There were six groups of rats in each cycle of the experiment and each group contained six rats. The procedure was repeated twelve times (cycles) (n = 72 for each arm; half male and half female). All substances were applied ip. All groups (1-6) received 1 microMol POX ( approximately LD(75)); groups 1-5 also received 50 microMol TIA at different points in time. Group 1 (G(1)): TIA 120 min before POX Group 2 (G(2)): TIA 90 min before POX, Group 3 (G(3)): TIA 60 min before POX, Group 4 (G(4)): TIA 30 min before POX, Group 5 (G(5)): TIA & POX simultaneously, Group 6 (G(6)): POX only. The animals were monitored for 48 hours and mortality/survival times were recorded at 30 min, 1, 2, 3, 4, 24 and 48 h. AChE activities were determined at 30 min, 24 and 48 h in surviving animals. Statistical analysis was performed on the mortality data, cumulative survival times and enzyme activity data. Mortality data was compared using Kaplan-Meier plots. Cumulative survival times and enzyme activites were compared using the Mann-Whitney rank order test. No Bonferroni correction for multiple comparisons was applied and an alpha < or= 0.05 was considered significant. RESULTS: Mortality is statistically significantly reduced by TIA pre-treatment at all points-in-time. Highest protection is achieved if TIA is given 90 to 0 min before OPC exposure. The reduction in mortality is not correlated to TIA plasma levels (C (max) approximately 120 min post ip-administration). TIA pre-treatment is not affecting AChE activity regardless of the timing of administration. CONCLUSION: The lack of correlation between TIA plasma levels and degree of mortality reduction as well as the lack of protective effect on enzyme activity seem to indicate that the site of action of TIA is not the blood. While our hypothesis that TIA would protect AChE in a pyridostigmine-like manner (via protection of the enzyme) could not be confirmed, the reduction in mortality with TIA pre-treatment is nevertheless of potential interest.


Asunto(s)
Esquema de Medicación , Paraoxon/envenenamiento , Clorhidrato de Tiapamilo/administración & dosificación , Animales , Colinesterasas/metabolismo , Eritrocitos/enzimología , Femenino , Masculino , Estudios Prospectivos , Ratas , Ratas Wistar , Análisis de Supervivencia , Clorhidrato de Tiapamilo/sangre , Clorhidrato de Tiapamilo/farmacología , Factores de Tiempo
10.
Toxicol Sci ; 77(2): 258-62, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12857941

RESUMEN

Prophylactic and therapeutic efficacy against organophosphorus (OP) intoxication by pralidoxime (2-PAM) and atropine were studied and compared with sterically stabilized long-circulating liposomes encapsulating recombinant organophosphorus hydrolase (OPH), either alone or in various specific combinations, in paraoxon poisoning. Prophylactic and therapeutic properties of atropine and 2-PAM are diminished when they are used alone. However, their prophylactic effects are enhanced when they are used in combination. Present studies indicate that sterically stabilized liposomes (SL) encapsulating recombinant OPH (SL-OPH) alone can provide much better therapeutic and prophylactic protection than the classic 2-PAM + atropine combination. This protection was even more dramatic when SL-OPH was employed in combination with 2-PAM and/or atropine: the magnitude of prophylactic antidotal protection was an astounding 1022 LD(50) [920 mg/kg (LD(50) of paraoxon with antagonists)/ 0.95 mg/kg (LD(50) of control paraoxon)], and the therapeutic antidotal protection was 156 LD(50) [140 mg/kg (LD(50) of paraoxon with antagonists)/0.9 mg/kg (LD(50) of control paraoxon)]. The current study firmly establishes the value of using liposome encapsulating OPH.


Asunto(s)
Arildialquilfosfatasa/administración & dosificación , Atropina/farmacología , Atropina/uso terapéutico , Insecticidas/envenenamiento , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/prevención & control , Paraoxon/envenenamiento , Compuestos de Pralidoxima/farmacología , Compuestos de Pralidoxima/uso terapéutico , Animales , Antídotos/administración & dosificación , Antídotos/farmacología , Antídotos/uso terapéutico , Reactivadores de la Colinesterasa/farmacología , Reactivadores de la Colinesterasa/uso terapéutico , Combinación de Medicamentos , Dosificación Letal Mediana , Liposomas , Masculino , Ratones , Ratones Endogámicos BALB C , Antagonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/uso terapéutico
11.
Acad Emerg Med ; 10(3): 286-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12615598

RESUMEN

OBJECTIVE: Organophosphates are used as pesticides, herbicides, and chemical warfare agents. Treatment of organophosphate poisoning is with intravenous atropine and pralidoxime in addition to supportive care. This study determined the efficacy of oral agents in preventing death from organophosphate poisoning. METHODS: The organophosphate paraoxon (8 mg/kg) was used in a murine model with lethality at four and 24 hours as an end point. For oral treatment, 15 male Balbc mice were given either atropine sulfate (4 mg/kg), or a combination of atropine sulfate (4 mg/kg) with pralidoxime (100 mg/kg), by oral gavage. A control group of 22 mice received water by oral gavage. Chi-square analysis was used to compare results in the different groups. RESULTS: Of the control group, six of 22 survived to four hours after paraoxon exposure. Of the exposed animals treated with oral atropine, eight of 15 survived to four hours. Of the exposed animals treated with a combination of atropine and pralidoxime, 13 of 15 survived to four hours. All animals surviving to four hours survived to 24 hours. The increased survival of animals in the atropine group relative to the control group was not significant (p = 0.09). Survival was significant in the group treated with atropine and pralidoxime relative to atropine alone (p = 0.02) and to the control group (p = 0.0002). All treated mice surviving at four hours were alive at 24 hours. CONCLUSIONS: Both oral atropine and a combination of oral atropine and pralidoxime improved survival, and combination therapy achieved statistical significance. Generalization of this result to other organophosphate pesticides, other doses of paraoxon, and other species cannot be made without further investigations.


Asunto(s)
Antídotos/uso terapéutico , Atropina/uso terapéutico , Insecticidas/envenenamiento , Antagonistas Muscarínicos/uso terapéutico , Paraoxon/envenenamiento , Parasimpatolíticos/uso terapéutico , Compuestos de Pralidoxima/uso terapéutico , Animales , Masculino , Ratones , Ratones Endogámicos BALB C
12.
Pharmacogenetics ; 13(2): 81-8, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12563177

RESUMEN

OBJECTIVES: Human serum paraoxonase (PON1) hydrolyses organophosphate pesticides (OPs) entering the blood circulation and tissue fluid thus limiting toxicity. The PON1 coding region has two polymorphisms involving the amino acids at position 55 (Lt<--M) and 192 (Qt<--R), giving rise to isoenzymes which differ in their catalytic rate for the hydrolysis of OPs. We therefore hypothesized that individuals inheriting low activity isoforms of PON1 would be more liable to report symptoms of OP toxicity. METHODS: We have therefore investigated the relationship between PON1 genetic polymorphisms and PON1 activity in farmers reporting chronic ill health which they attributed to OP exposure whilst sheep dipping (cases) and farmers who carried out similar activities, but remained well (controls). Diazoxon, paraoxon and phenylacetate were used as substrates for PON1. Diazoxon is the active metabolite of diazinon, the sheep dip most commonly used in the UK. RESULTS: Cases were found to be more likely to have the R192 allele ( 0.01) and to have the L55 allele ( 0.05) than the controls. This combination of R and L genotypes was associated with lower PON1 activity towards diazoxon in both cases and controls. Farmers in the lowest quintile for the rate of serum diazoxon hydrolysis had a greater risk of being a case i.e. of reporting ill health (odds ratio 2.47 (95% CI 1.35-2.82)), than the other four quintiles of diazoxon hydrolysis. The rate of serum hydrolysis of paraoxon was greatest in cases and controls with the R/L haplotype (both 0.001). CONCLUSIONS: The farmers reporting chronic ill health due to organophosphate exposure have a higher proportion of the PON1-192R polymorphism associated with lower rates of diazoxon hydrolysis and lower rates of diazoxon hydrolysis than the controls and that their ill health may be explained by a lower ability to detoxify diazoxon.


Asunto(s)
Esterasas/genética , Predisposición Genética a la Enfermedad , Insecticidas/envenenamiento , Enfermedades Profesionales/genética , Polimorfismo Genético/genética , Animales , Arildialquilfosfatasa , Biomarcadores , Estudios de Casos y Controles , Esterasas/sangre , Femenino , Frecuencia de los Genes , Humanos , Hígado/enzimología , Pruebas de Función Hepática , Masculino , Persona de Mediana Edad , Enfermedades Profesionales/enzimología , Intoxicación por Organofosfatos , Compuestos Organofosforados , Paraoxon/envenenamiento , Fenilacetatos/envenenamiento , Factores de Riesgo , Ovinos
13.
J Toxicol Clin Toxicol ; 39(1): 27-31, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11327223

RESUMEN

INTRODUCTION: Intoxications with organophosphorous compounds, especially paraoxon, are frequent. Organophosphorous compounds inhibit serine hydrolases such as acetylcholine, butyrilcholine, and carboxyl esterases although acetylcholine and butyrylcholine are too sensitive to paraoxon to be useful markers of severity. They cannot show a dose-dependent inhibition during an acute organophosphorous compounds exposure because maximal enzyme inhibition is reached at very low organophosphorous compounds concentrations. PURPOSE: To determine in vitro the dose-effect relationship between the activity of the paraoxon-sensitive phenylvalerate hydrolase, a member of the carboxvl esterases family, and the paraoxon dose, and to assess its utility as a putatively less sensitive enzyme marker to monitor the severity of an acute paraoxon intoxication. MATERIALS AND METHODS: Phenylvalerate hydrolase and butyrylcholine activities were determined in serum of nine healthy human volunteers before and after addition of different concentrations of paraoxon. The determination of phenyl-valerate hydrolase activity was carried out using a modification of the method described by Johnson. A commercially available kit was used to measure butyrylcholine activity. RESULTS: Paraoxon inhibits phenyl-valerate hydrolase activity at concentrations above 10 M. Maximal inhibition (approximately 50% of baseline) is achieved at concentrations above 2.5 x 10(-7) M. The IC50 value of paraoxon for phenyl-valerate hydrolase is 34+/-2 nM. The uninhibited phenyl-valerate hydrolase activity is due to paraoxon-resistent isoforms. Paraoxon begins inhibiting butyrylcholine activity at concentrations above 10(-9) M. At concentrations above 5 x 10(-5) M, no butyrylcholine activity is measulrable. The IC50 value of paraoxon for butyrylcholine is 150+/-23 nM. CONCLUSION: The paraoxon-sensitive subunit of phenyl-valerate hydrolase shows dose-dependent inhibition when exposed to paraoxon in vitro, but it is even more sensitive than butyrylcholine to paraoxon inhibition. Determinations of phenyl-valerate hydrolase activity to assess the severity of an acute organophosphorous compounds poisoning cannot be recommended, but phenyl-valerate hydrolase may have utility in worker surveillance.


Asunto(s)
Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Colina/análogos & derivados , Colina/antagonistas & inhibidores , Insecticidas/envenenamiento , Paraoxon/envenenamiento , Biomarcadores , Hidrolasas de Éster Carboxílico/sangre , Colina/sangre , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino
15.
Morfologiia ; 118(4): 66-9, 2000.
Artículo en Ruso | MEDLINE | ID: mdl-12629810

RESUMEN

Poisoning with phosphacol causes dose--dependent decrease of blood flow speed in small vessels of mesoappendix. Administration of LD50 of phosphacole results in blood stagnation, simultaneous blood pressure fall, which leads to death of part of the animals. Electron microscopic study revealed the presence of acetyl and buthyryl cholinesterase in endotheliocytes of mesoappendicular capillaries, the activity of which was completely suppressed by administration of LD50 of phosphacol. 0,0-dimethyl-0 (2,2-dichlorvinyl) phosphate LD10 caused the damage of endotheliocyte surface. It was suggested that endothelial cholino-receptors that are activated through the rise of redundant acetyl-choline level in blood on the background of cholinesterase inhibition participate in the mechanism of pathological reactions described. Such variant of toxic effect was characterized as distant.


Asunto(s)
Acetilcolina/fisiología , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Inhibidores de la Colinesterasa/envenenamiento , Paraoxon/envenenamiento , Acetilcolinesterasa/metabolismo , Animales , Apéndice/irrigación sanguínea , Apéndice/efectos de los fármacos , Apéndice/ultraestructura , Presión Sanguínea/efectos de los fármacos , Butirilcolinesterasa/metabolismo , Capilares/efectos de los fármacos , Capilares/ultraestructura , Diclorvos/toxicidad , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/ultraestructura , Dosificación Letal Mediana , Ratas , Ratas Endogámicas , Factores de Tiempo
16.
Toxicol Appl Pharmacol ; 156(1): 56-63, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10101099

RESUMEN

This investigation effort is focused on increasing organophosphate (OP) degradation by phosphotriesterase to antagonize OP intoxication. For these studies, sterically stabilized liposomes encapsulating recombinant phosphotriesterase were employed. This enzyme was obtained from Flavobacterium sp. and was expressed in Escherichia coli. It has a broad substrate specificity, which includes parathion, paraoxon, soman, sarin, diisopropylfluorophosphate, and other organophosphorous compounds. Paraoxon is rapidly hydrolyzed by phosphotriesterase to the less toxic 4-nitrophenol and diethylphosphate. This enzyme was isolated and purified over 1600-fold and subsequently encapsulated within sterically stabilized liposomes (SL). The properties of this encapsulated phosphotriesterase were investigated. When these liposomes containing phosphotriesterase were incubated with paraoxon, it readily degraded the paraoxon. Hydrolysis of paraoxon did not occur when these sterically stabilized liposomes contained no phosphotriesterase. These sterically stabilized liposomes (SL) containing phosphotriesterases (SL)* were employed as a carrier model to antagonize the toxic effects of paraoxon by hydrolyzing it to the less toxic 4-nitrophenol and diethylphosphate. This enzyme-SL complex (SL)* was administered intravenously to mice either alone or in combination with pralidoxime (2-PAM) and/or atropine intraperitoneally. These results indicate that this carrier model system provides a striking enhanced protective effects against the lethal effects of paraoxon. Moreover when these carrier liposomes were administered with 2-PAM and/or atropine, a dramatic enhanced protection was observed.


Asunto(s)
Esterasas/administración & dosificación , Insecticidas/envenenamiento , Paraoxon/envenenamiento , Animales , Arildialquilfosfatasa , Portadores de Fármacos , Punto Isoeléctrico , Liposomas , Masculino , Ratones , Ratones Endogámicos BALB C , Paraoxon/antagonistas & inhibidores , Compuestos de Pralidoxima/farmacología , Proteínas Recombinantes/administración & dosificación
17.
Biotechnol Bioeng ; 62(6): 659-65, 1999 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-10068213

RESUMEN

Current events across the globe necessitate rapid technological advances to combat the epidemic of nerve agent chemical weapons. Biocatalysis has emerged as a viable tool in the detoxification of organophosphorus neurotoxins, such as the chemical weapons VX and sarin. Efficient detoxification of contaminated equipment, machinery, and soils are of principal concern. This study describes the incorporation of a biocatalyst (organophosphorus hydrolase, E.C. 3.1.8.1) into conventional formulations of fire fighting foam. The capacity of fire fighting foams to decrease volatilization of contained contaminants, increase surface wettability, and control the rate of enzyme delivery to large areas makes them useful vehicles for enzyme application at surfaces. The performance of enzyme containing foams has been shown to be not only reproducible but also predictable. An empirical model provides reasonable estimations for the amounts of achievable surface decontamination as a function of the important parameters of the system. Theoretical modeling illustrates that the enzyme-containing foam is capable of extracting agent from the surface and is catalytically active at the foam-surface interface and throughout the foam itself. Biocatalytic foam has proven to be an effective, "environmentally friendly" means of surface and soil decontamination.


Asunto(s)
Sustancias para la Guerra Química/farmacocinética , Esterasas/metabolismo , Neurotoxinas/farmacocinética , Paraoxon/farmacocinética , Arildialquilfosfatasa , Catálisis , Sustancias para la Guerra Química/química , Escherichia coli/enzimología , Esterasas/administración & dosificación , Esterasas/química , Incendios , Hidrólisis , Inactivación Metabólica , Neurotoxinas/química , Paraoxon/química , Paraoxon/envenenamiento , Contaminantes del Suelo/análisis , Contaminantes del Suelo/farmacocinética , Propiedades de Superficie , Volatilización
18.
Neurotoxicology ; 19(6): 833-8, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9863772

RESUMEN

Acute organophosphorus anticholinesterase poisoning induces a necrotizing end-plate myopathy in rats and patients. Acetylcholine (ACh) excess leads to prolonged synaptic currents and increased influx of cations including calcium through the postsynaptic ACh receptor channels with prolonged muscle membrane depolarization, excess calcium influx into the sarcoplasm, and ultimately muscle fiber necrosis. Quinoline derivatives such as quinidine induce or worsen pre- and postsynaptic disorders of neuromuscular transmission in humans, and are beneficial in patients suffering from a rare congenital myasthenic syndrome called the slow channel congenital myasthenic syndrome. These drugs correct the prolonged opening times of the mutated acetylcholine receptor channels in this myasthenic syndrome. We treated paraoxon-poisoned rats with 4 x 10 or 4 x 50 mg/kg of quinidine and assessed the severity of the necrotizing myopathy in gastrocnemius and diaphragm muscle biopsies. Fasciculations were decreased and the necrotizing myopathy was prevented in most treated rats, with absence of necrotic muscle fibers in most animals in the high-dose group. Survival was not different from untreated poisoned animals. A number of physiological mechanisms, including blocking of presynaptic voltage-gated sodium or calcium channels or inhibition of the postsynaptic ACh receptors channels may have contributed to the attenuation of the myonecrosis. The optimal dose and the drug of choice amongst the clinically available quinoline derivatives remains to be determined.


Asunto(s)
Inhibidores de la Colinesterasa/envenenamiento , Insecticidas/envenenamiento , Enfermedades Musculares/prevención & control , Paraoxon/envenenamiento , Quinidina/uso terapéutico , Animales , Biopsia , Insecticidas/antagonistas & inhibidores , Masculino , Fibras Musculares Esqueléticas/patología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/patología , Necrosis , Paraoxon/antagonistas & inhibidores , Ratas , Ratas Wistar
19.
Toxicol Appl Pharmacol ; 152(1): 184-92, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9772214

RESUMEN

We investigated the failure of 2-PAM to protect honey bees against poisoning with paraoxon. The protective effect of the oxime 2-PAM against inhibition of acetylcholinesterase (AChE) by paraoxon was estimated in vitro and in vivo and was correlated with the mortality of paraoxon-treated bees. In vitro, 2-PAM protected 90% of AChE activity in the presence of paraoxon and reactivated more than 90% of inhibited AChE. Minor soluble and major membrane-bound forms of bee AChE presented about similar extents of reactivation, but the first order rate constant of reactivation (kobs) of the soluble form is threefold higher than that of the membrane-bound form. However, this difference did not significantly influence the reactivation kinetics of total AChE; the constant kobs of the membrane-bound form reflected that of total AChE. The linear kinetic profile of total AChE reactivation supported the conclusion that there was a single population of reactivatable species. The bimolecular rate constant of reactivation (kr), the dephosphorylation rate constant (k2), and the dissociation constant (Kd) were 646 M-1.min-1, 0.84 min-1 and 1. 30 mM, respectively. In vivo, administration of 2-PAM, after paraoxon exposure, induced a complete protection of AChE activity, but did not elicit any significant effect on mortality in paraoxon-treated bees. The inefficiency of 2-PAM to antagonize paraoxon-induced mortality was not changed by the administration of 2-PAM in pretreatment-therapy and in therapy treatments. These results indicated that the mortality of paraoxon-poisoned honey bees was not due to a lack of AChE reactivation.


Asunto(s)
Acetilcolinesterasa/metabolismo , Abejas/efectos de los fármacos , Inhibidores de la Colinesterasa/envenenamiento , Reactivadores de la Colinesterasa/farmacología , Insecticidas/envenenamiento , Paraoxon/envenenamiento , Compuestos de Pralidoxima/farmacología , Animales , Abejas/enzimología , Relación Dosis-Respuesta a Droga , Intoxicación/mortalidad , Tasa de Supervivencia
20.
J Appl Toxicol ; 18(4): 293-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9719430

RESUMEN

A therapeutic regimen was established to keep blood pressure, heart rate and haematocrit within the normal range during high-dose paraoxon (PX) exposure (ca. 150 x LD50) in mini pigs in order to achieve survival. Previous experiments showed that mini pigs exposed to high-dose PX died shortly after PX infusion due to hypertension, tachycardia and increased haematocrit if no antihypertensive and fluid therapy was initiated. Therefore, antihypertensive and fluid therapy with magnesium (MgSO4) and Ringer's solution was established to keep the blood pressure, heart rate and haematocrit within a pre-established normal range. Anaesthesized mini pigs received intravenously PX (54 mg kg(-1) body wt.) dissolved in alcohol. The control group received alcohol in corresponding amounts. When the blood pressure and heart rate increased, MgSO4 was given intravenously until measured values reached the normal range. When the haematocrit increased, fluids were given intravenously until the haematocrit reached the normal range. The measured values in the PX group were compared with the measured values of the control group using the 'rank order test'. As intended, no statistically significant differences between blood pressure, heart rate or haematocrit were found after therapy, but the PX group required statistically significantly more MgSO4 and fluids than the control group to keep the blood pressure, heart rate and haematocrit within the normal range. We assume that the increased need of antihypertensive therapy is due to a phaeochromocytoma-like pattern caused by an excessive release of catecholamines from the adrenal medulla, which is under sympathotonic control and activated by acetylcholine. Paraoxon is known to cause endogenous acetylcholine poisoning. The high fluid requirements in the PX group are most probably caused by extravasation of fluids due to the damage inflicted on biological membranes by organophosphorus compounds. An activation of secretory glands probably also contributes to the increase in haematocrit through consumption of fluids. In conclusion, the survival of mini pigs exposed to high-dose PX can be achieved by tight control of blood pressure, heart rate and haematocrit using MgSO4 and fluids.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Intoxicación por Organofosfatos , Paraoxon/envenenamiento , Pruebas de Toxicidad , Animales , Antihipertensivos/uso terapéutico , Colinesterasas/sangre , Fluidoterapia , Hematócrito , Hipertensión/prevención & control , Inyecciones Intravenosas , Soluciones Isotónicas/uso terapéutico , Sulfato de Magnesio/uso terapéutico , Compuestos Organofosforados/administración & dosificación , Paraoxon/administración & dosificación , Distribución Aleatoria , Solución de Ringer , Porcinos , Porcinos Enanos , Taquicardia/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...