Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Interact ; 259(Pt B): 187-204, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27129421

RESUMO

The antidotal treatment of organophosphates (OP) nerve agents (NA) poisoning is based on anticholinergics (e.g. atropine) combined with oxime reactivators (e.g. 2PAM) of acetylcholinesterase (AChE). This treatment is symptomatic and does not degrade the OP. New small-molecule OP scavengers were developed as bifunctional hybrids. Their molecular design was based on combining a nucleophile that directly degrades OP with a moiety that reactivates OP-inhibited AChE. The OP degrading moiety is either benzhydroxamic acid (BHA) or 4-pyridinehydroxamic acid (4PHA) coupled via (CH2)n, (n = 1 or 3) to 2PAM. Three newly synthesized oxime-hydroxamate hybrids: 2PAMPr4PHA, 2PAMMeBHA and 2,4-DiPAMMeBHA were found to detoxify sarin, cyclosarin and soman in solution at 3-10-fold faster rate than 2PAM and to reactivate OP-AChE in vitro. 2PAMPr4PHA displayed 18-fold faster reactivation than 2-PAM of cyclosarin-inhibited HuAChE (kr = 3.6 × 102 vs. 0.2 × 102 M-1min-1, respectively, 37 °C). These hybrids inhibited AChE reversibly, IC50 = 16-48 µM, thereby decreasing the inhibition rates by OPs. The LD50 (im) of 2PAMPr4PHA, 2PAMMeBHA and 2,4DiPAMMeBHA are >568, 508 and >506 µmol/kg in rats and 144, 203 and >506 µmol/kg in guinea pigs. The rate of blood ChE recovery by the hybrids administered either pre- or post-exposure to 0.8xLD50 sarin was comparable or faster than 2PAM. Antidotal efficacy of 2PAMPr4PHA, 2PAMMeBHA and 2,4DiPAMMeBHA administered with atropine, as pre-treatment to sarin in rats (im), yielded protection ratios (PR) 11.6, 11.5 and 4.7, respectively, vs. 5.5 with 2PAM. Post-treatment against various OPs in rats and guinea-pigs yielded PRs higher or similar to that of 2 PAM. Our in vivo data indicates that some hybrids may serve as efficient small molecule scavengers for mitigating the toxicity of OP NAs.


Assuntos
Agentes Neurotóxicos/toxicidade , Neurotoxinas/toxicidade , Bibliotecas de Moléculas Pequenas/farmacologia , Acetilcolinesterase/sangue , Acetilcolinesterase/metabolismo , Animais , Antídotos/farmacologia , Inibidores da Colinesterase/farmacologia , Reativadores da Colinesterase/farmacologia , Cobaias , Humanos , Ácidos Hidroxâmicos/química , Concentração Inibidora 50 , Cinética , Masculino , Organofosfatos/toxicidade , Compostos Organofosforados/toxicidade , Compostos Organotiofosforados/toxicidade , Oximas/química , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Sarina/intoxicação , Fatores de Tempo , Testes de Toxicidade Aguda
2.
Neurotoxicology ; 49: 132-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25976749

RESUMO

Sarin poisoned rats display a hyper-cholinergic activity including hypersalivation, tremors, seizures and death. Here we studied the time and dose effects of midazolam treatment following nerve agent exposure. Rats were exposed to sarin (1.2 LD50, 108 µg/kg, im), and treated 1 min later with TMB4 and atropine (TA 7.5 and 5 mg/kg, im, respectively). Midazolam was injected either at 1 min (1 mg/kg, im), or 1 h later (1 or 5 mg/kg i.m.). Cortical seizures were monitored by electrocorticogram (ECoG). At 5 weeks, rats were assessed in a water maze task, and then their brains were extracted for biochemical analysis and histological evaluation. Results revealed a time and dose dependent effects of midazolam treatment. Rats treated with TA only displayed acute signs of sarin intoxication, 29% died within 24h and the ECoG showed seizures for several hours. Animals that received midazolam within 1 min survived with only minor clinical signs but with no biochemical, behavioral, or histological sequel. Animals that lived to receive midazolam at 1h (87%) survived and the effects of the delayed administration were dose dependent. Midazolam 5 mg/kg significantly counteracted the acute signs of intoxication and the impaired behavioral performance, attenuated some of the inflammatory response with no effect on morphological damage. Midazolam 1mg/kg showed only a slight tendency to modulate the cognitive function. In addition, the delayed administration of both midazolam doses significantly attenuated ECoG compared to TA treatment only. These results suggest that following prolonged seizure, high dose midazolam is beneficial in counteracting adverse effects of sarin poisoning.


Assuntos
Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Inibidores da Colinesterase/toxicidade , Hipnóticos e Sedativos/administração & dosagem , Midazolam/administração & dosagem , Sarina/toxicidade , Análise de Variância , Animais , Lesões Encefálicas/fisiopatologia , Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Esquema de Medicação , Eletroencefalografia , Ensaio de Imunoadsorção Enzimática , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Índice de Gravidade de Doença , Fatores de Tempo
3.
Neurotoxicology ; 49: 36-44, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26001567

RESUMO

Rivastigmine, a reversible cholinesterase inhibitor, approved as a remedy in Alzheimer's disease, was suggested as pretreatment against nerve agents poisoning. We evaluated the pharmacokinetic, pharmacodynamic, physiologic, cognitive and emotional effects of repeated rivastigmine in young healthy male adults, in a double blind, placebo controlled crossover trial. Three groups completed 3 treatment periods: 0, 1.5 and 3mg twice a day, for a total of 5 intakes. Parameters monitored were: vital signs, ECG, laboratory tests, sialometry, visual accommodation, inspiratory peak flow, and cognitive function tests. Adverse reactions were mild. Peak blood levels and peak cholinesterase inhibition increased with repeated intakes, and high variability and non-linear pharmacokinetics were demonstrated. In addition, two cognitive functions were affected (perceptual speed and dynamic tracking). The complicated pharmacological profile and the high inter-personal variability limit the potential use of rivastigmine as pretreatment for war fighters and first responders.


Assuntos
Cognição/efeitos dos fármacos , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/farmacologia , Rivastigmina/sangue , Rivastigmina/farmacologia , Acetilcolinesterase/metabolismo , Adolescente , Adulto , Estudos Cross-Over , Método Duplo-Cego , Emoções/efeitos dos fármacos , Seguimentos , Voluntários Saudáveis , Humanos , Masculino , Saliva/metabolismo , Fatores de Tempo , Visão Ocular/efeitos dos fármacos , Acuidade Visual/efeitos dos fármacos , Adulto Jovem
4.
Toxicology ; 325: 115-24, 2014 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-25201353

RESUMO

Potent cholinesterase inhibitors such as sarin, induce an array of harmful effects including hypersecretion, convulsions and ultimately death. Surviving subjects demonstrate damage in specific brain regions that lead to cognitive and neurological dysfunctions. An early accumulation of acetylcholine in the synaptic clefts was suggested as the trigger of a sequence of neurochemical events such as an excessive outpour of glutamate and activation of its receptors. Indeed, alterations in NMDA and AMPA central receptors' densities were detected in brains of poisoned animals. Attempts to improve the current cholinergic-based treatment by adding potent anticonvulsants or antiglutamatergic drugs produced unsatisfactory results. In light of recent events in Syria and the probability of various scenarios of military or terrorist attacks involving organophosphate (OP) nerve agent, research should focus on finding markedly improved countermeasures. Caramiphen, an antimuscarinic drug with antiglutamatergic and GABAergic facilitating properties, was evaluated in a wide range of animals and experimental protocols against OP poisoning. Its remarkable efficacy against OP exposure was established both in prophylactic and post-exposure therapies in both small and large animals. The present review will highlight the outstanding neuroprotective effect of caramiphen as the optimal candidate for the treatment of OP-exposed subjects.


Assuntos
Antídotos/uso terapêutico , Encéfalo/efeitos dos fármacos , Substâncias para a Guerra Química/intoxicação , Inibidores da Colinesterase/intoxicação , Ciclopentanos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Intoxicação por Organofosfatos/tratamento farmacológico , Acetilcolina/metabolismo , Animais , Antídotos/efeitos adversos , Antídotos/farmacocinética , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Ciclopentanos/efeitos adversos , Ciclopentanos/farmacocinética , Humanos , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/farmacocinética , Intoxicação por Organofosfatos/etiologia , Intoxicação por Organofosfatos/metabolismo , Intoxicação por Organofosfatos/fisiopatologia
5.
Arch Toxicol ; 87(2): 347-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23052190

RESUMO

Sarin, a potent organophosphate cholinesterase inhibitor, induces an array of toxic effects including convulsions. Many antidotal treatments contain anticonvulsants to block seizure activity and the ensuing brain damage. Magnesium sulfate (MGS) is used to suppress eclamptic seizures in pregnant women with hypertension and was shown to block kainate-induced convulsions. Magnesium sulfate was evaluated herein as an anticonvulsant against sarin poisoning and its efficacy was compared with the potent anticonvulsants midazolam (MDZ) and caramiphen (CRM). Rats were exposed to a convulsant dose of sarin (96 µg/kg, im) and 1 min later treated with the oxime TMB4 and atropine to increase survival. Five minutes after initiation of convulsions, MGS, CRM, or MDZ were administered. Attenuation of tonic-clonic convulsions was observed following all these treatments. However, radio-telemetric electro-corticography (ECoG) monitoring demonstrated sustained seizure activity in MGS-injected animals while this activity was completely blocked by MDZ and CRM. This disrupted brain activity was associated with marked increase in brain translocator protein levels, a marker for brain damage, measured 1 week following exposure. Additionally, histopathological analyses of MGS-treated group showed typical sarin-induced brain injury excluding the hippocampus that was partially protected. Our results clearly show that MGS demonstrated misleading features as an anticonvulsant against sarin-induced seizures. This stems from the dissociation observed between overt convulsions and seizure activity. Thus, the presence or absence of motor convulsions may be an unreliable indicator in the assessment of clinical status and in directing adequate antidotal treatments following exposure to nerve agents in battle field or terror attacks.


Assuntos
Anticonvulsivantes/farmacologia , Antídotos/farmacologia , Substâncias para a Guerra Química/intoxicação , Sulfato de Magnésio/farmacologia , Sarina/intoxicação , Convulsões/tratamento farmacológico , Animais , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Ciclopentanos/farmacologia , Epilepsia Tônico-Clônica , Masculino , Midazolam/farmacologia , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Telemetria
6.
Toxicology ; 290(2-3): 149-55, 2011 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-21978866

RESUMO

Some organophosphorus compounds (OPs) are nerve agents that continue to concern military personnel and civilians as potential battlefield and terrorist threats. Additionally, OPs are used in agriculture where they are associated with numerous cases of intentional and accidental misuse. These toxicants induce an array of deleterious effects including respiratory distress, convulsions and ultimately death. A mechanism involving a rapid and potent inhibition of peripheral and central cholinesterases leading to a massive buildup of acetylcholine in synaptic clefts was suggested as the underlying trigger of the toxic events. Indeed, therapy comprised of an acetylcholinesterase reactivator (i.e., oxime) and a cholinergic antagonist (e.g., atropine) is the accepted major paradigm for protection. This approach yields a remarkable survival rate but fails to prevent neurological and behavioral deficits. Extensive research revealed a complex picture consisting of an early activation of several neurotransmitter systems, in which the glutamatergic plays a pivotal role., Data accumulated in recent years support the concept that multi-targeting of pathways including glutamatergic and cholinergic circuits is required for an effective treatment. Drugs that demonstrate the ability to interact with several systems (e.g., caramiphen) were found to afford a superior protection against OPs as compared to specific antimuscarinic ligands (e.g., scopolamine). Compounds that potently block muscarinic receptors, interact with the NMDA ion channel and in addition are able to modulate σ(1) sites and/or GABAergic transmission seem to represent the emerging backbone for novel antidotes against OP poisoning. Several multifunctional drugs are already used for complex diseases e.g., cancer and depression.


Assuntos
Antídotos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Intoxicação por Organofosfatos , Acetilcolina/metabolismo , Animais , Atropina/farmacologia , Antagonistas Colinérgicos/farmacologia , Ciclopentanos/farmacologia , Humanos , Antagonistas Muscarínicos/farmacologia , Intoxicação/tratamento farmacológico , Escopolamina/farmacologia
7.
Chem Biol Interact ; 187(1-3): 253-8, 2010 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-20005217

RESUMO

We have shown previously that conjugation of polyethylene glycol (PEG) chains to recombinant human acetylcholinesterase (rHuAChE) results in the extension of its residence time in the circulation of mice and monkeys [1,2]. By profiling the pharmacokinetic behavior of an array of well-defined hypolysine human mutant AChE molecules following PEGylation, we now determine that the duration of these enzyme forms in the circulation of rhesus macaques correlates with their number of appended PEG moieties, and is influenced by the actual location of the PEG chains at the molecule surface, as well. These findings, which concur with those we have previously established in mice, indicate that a common set of rules dictates the circulatory fate of PEGylated HuAChEs in rodents and non-human primates. In addition to its effect on circulatory residence, PEGylation reduces the ability of the rHuAChE bioscavenger to elicit an immune response in the heterologous mouse animal system. Thus, an inverse relationship between anti-AChE antibody production and PEG loading was observed following repeated administration of the different PEGylated hypolysine human AChEs to mice. We note however, that in rhesus macaques, the essentially homologous (human) AChE does not induce specific anti-AChE antibodies after repeated administration of high doses of the enzyme in its PEGylated form, and even in its non-PEGylated form. Taken together, these findings indicate that PEG acts by veiling enzyme-related epitopes, which would otherwise interact with host circulatory elimination pathways and immune system. The barring of such interactions by obstructive PEGs, confers the enzyme molecule with both extended circulatory residence and mitigated immunogenic properties. Further modulation by incorporation of the F338A mutation into the PEGylated hypolysine rHuAChE enzyme mold, resulted in the generation of an OP-bioscavenger that displayed reduced aging rates and could effectively protect mice against repeated exposure to CW agents. This selected 4-PEG F338A-AChE can serve as a paradigm for new generation OP-bioscavengers, specifically tailored for prophylactic treatment against toxic OP-agents.


Assuntos
Acetilcolinesterase/metabolismo , Acetilcolinesterase/farmacocinética , Biocatálise , Lisina , Mutação , Compostos Organofosforados/metabolismo , Polietilenoglicóis/química , Acetilcolinesterase/sangue , Acetilcolinesterase/genética , Animais , Antídotos/metabolismo , Antídotos/farmacocinética , Linhagem Celular , Epitopos/imunologia , Feminino , Humanos , Macaca mulatta , Masculino , Camundongos , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética
8.
Toxicol Appl Pharmacol ; 232(2): 351-8, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18680758

RESUMO

Potent cholinesterase inhibitors (e.g., soman, sarin), induce a wide range of deleterious effects including convulsions, behavioral impairments and ultimately, death. Due to the likelihood of various scenarios of military or terrorist attacks by these and other chemical weapons, research has to be aimed at finding optimal therapies. Early accumulation of acetylcholine in synaptic clefts was suggested to trigger an array of toxic events including an excessive release of glutamate, culminating in the activation of its receptors. Stimulation of the N-Methyl-D-Aspartate (NMDA) subtype of these receptors was associated with the neuronal injury that initiates organophosphate-induced brain damage. The notion of a stepwise mechanism yielded treatments based on a combination of an immediate administration of enzyme reactivators and anticholinergic drugs. This strategy dramatically increased survival rates but did not abolish convulsions and failed to prevent the ensuing cognitive dysfunction. Efforts to improve this paradigm by adding anticonvulsants or antiglutamatergic drugs with anti-epileptic characteristics produced dubious results. Under these conditions, benactyzine and caramiphen, agents with anticholinergic and antiglutamatergic properties, provided improved protection when introduced as adjunct agents to oximes, reversible cholinesterase inhibitors and/or specific antimuscarinic drugs such as atropine. In contrast, the specific antimuscarinic drug scopolamine failed to block soman-induced changes in glutamatergic and behavioral parameters even when given prophylactically. These findings along with a large number of additional reports led towards the conclusion that the therapeutic advantage of drugs such as benactyzine and caramiphen could derive from their ability to modulate central cholinergic and glutamate neurotransmission.


Assuntos
Antagonistas Colinérgicos/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Ácido Glutâmico/metabolismo , Intoxicação por Organofosfatos , Organofosfatos/antagonistas & inibidores , Animais , Antagonistas Colinérgicos/farmacologia , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Ciclopentanos/farmacologia , Ciclopentanos/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Intoxicação por Gás/tratamento farmacológico , Intoxicação por Gás/metabolismo , Intoxicação por Gás/prevenção & controle , Humanos
9.
Mol Pharmacol ; 74(3): 755-63, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18523134

RESUMO

The high reactivity of cholinesterases (ChEs) toward organophosphorus (OP) compounds has led to propose recombinant ChEs as bioscavengers of nerve agents. The bioscavenging potential of recombinant ChEs can be enhanced by conjugation of polyethylene glycol (PEG) moieties, to extend their circulatory residence. However, the ability of exogenously administered ChEs to confer long-term protection against repeated exposures to nerve agents is still limited due to the aging process, whereby organophosphate-ChE adducts undergo irreversible dealkylation, which precludes oxime-mediated reactivation of the enzyme. To generate an optimal acetylcholinesterase (AChE)-based OP bioscavenger, the F338A mutation, known to decelerate the rate of aging of AChE-OP conjugates, was incorporated into polyethylene glycol-conjugated (PEGylated) human AChE. The PEGylated F338A-AChE displayed unaltered rates of hydrolysis, inhibition, phosphylation, and reactivation and could effectively protect mice against exposure to soman (pinacolylmethyl phosphonofluoridate), sarin (O-isopropyl methylphosphonofluoridate), or O-ethyl-S-(2-isopropylaminoethyl) methylphosphonothioate (VX). Unlike PEGylated wild-type (WT)-AChE, the PEGylated F338A-AChE exhibits significantly reduced aging rates after soman inhibition and can be efficiently reactivated by the 1-[[[4(aminocarbonyl)-pyridinio]methoxy]methyl]-2(hydroxyimino)methyl]pyridinium dichloride (HI-6) oxime, both in vitro and in vivo. Accordingly, oxime administration to PEG-F338A-AChE-pretreated mice enabled them to withstand repeated soman exposure (5.4 and 4 LD(50)/dose), whereas same regime treatment of non-PEGylated F338A-AChE- or PEGylated WT-AChE-pretreated mice failed to protect against the second challenge, due to rapid clearance or irreversible aging of the latter enzymes. Thus, judicious incorporation of selected mutations into the AChE mold in conjunction with its chemical modification provides means to engineer an optimal ChE-based OP bioscavenger in terms of circulatory longevity, resistance to aging, and reduced doses required for protection, even against repeated exposures to nerve agents, such as soman.


Assuntos
Acetilcolinesterase/farmacologia , Substituição de Aminoácidos , Compostos Organofosforados/metabolismo , Polietilenoglicóis/farmacologia , Acetilcolinesterase/farmacocinética , Alanina/genética , Animais , Sítios de Ligação , Linhagem Celular , Inibidores da Colinesterase/farmacologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Hidrólise/efeitos dos fármacos , Cinética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Compostos Organofosforados/toxicidade , Fenilalanina/genética , Fosforilação/efeitos dos fármacos , Polietilenoglicóis/farmacocinética , Soman/toxicidade , Análise de Sobrevida , Fatores de Tempo
10.
Toxicol Appl Pharmacol ; 231(1): 17-23, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18485435

RESUMO

The highly toxic organophosphorous compound VX [O-ethyl-S-(isoporopylaminoethyl) methyl phosphonothiolate] undergoes an incomplete decontamination by conventional chemicals and thus evaporates from urban surfaces, e.g., pavement, long after the initial insult. As a consequence to these characteristics of VX, even the expected low levels should be examined for their potential to induce functional impairments including those associated with neuronal changes. In the present study, we developed an animal model for subchronic, low-dose VX exposure and evaluated its effects in rats. Animals were exposed to VX (2.25 microg/kg/day, 0.05 LD(50)) for three months via implanted mini osmotic pumps. The rapidly attained continuous and marked whole-blood cholinesterase inhibition (approximately 60%), fully recovered 96 h post pump removal. Under these conditions, body weight, blood count and chemistry, water maze acquisition task, sensitivity to the muscarinic agonist oxotremorine, peripheral benzodiazepine receptors density and brain morphology as demonstrated by routine histopathology, remained unchanged. However, animals treated with VX showed abnormal initial response in an Open Field test and a reduction (approximately 30%) in the expression of the exocytotic synaptobrevin/vesicle associate membrane protein (VAMP) in hippocampal neurons. These changes could not be detected one month following termination of exposure. Our findings indicate that following a subchronic, low-level exposure to the chemical warfare agent VX some important processes might be considerably impaired. Further research should be addressed towards better understanding of its potential health ramifications and in search of optimal countermeasures.


Assuntos
Química Encefálica/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Inibidores da Colinesterase/toxicidade , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/psicologia , Compostos Organotiofosforados/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Contagem de Células Sanguíneas , Análise Química do Sangue , Western Blotting , Peso Corporal/efeitos dos fármacos , Encéfalo/patologia , Inibidores da Colinesterase/administração & dosagem , Colinesterases/sangue , Colinesterases/metabolismo , Doença Crônica , Implantes de Medicamento , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Agonistas Muscarínicos/farmacologia , Síndromes Neurotóxicas/fisiopatologia , Compostos Organotiofosforados/administração & dosagem , Oxotremorina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/enzimologia
11.
Toxicol Appl Pharmacol ; 227(1): 155-62, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18320638

RESUMO

Sarin, a potent cholinesterase inhibitor, induces an array of toxic effects including convulsions and behavioral impairments. We report here on the protection provided by post-exposure antidotal treatments against a lethal dose of sarin (1.2xLD50) by scopolamine, benactyzine, trihexyphenidyl or caramiphen, administered 5, 10 or 20 min after the initiation of convulsions. A mixture of the oxime TMB4 and atropine (TA) was injected 1 min following poisoning a paradigm that may represent a scenario reminiscent of a terror incident. Surviving TA-treated rats exhibited marked tonic-clonic convulsions, weight loss, poor clinical status and abnormal cognitive performance as assessed by the Morris water maze. Additionally, a dramatic increase in the density of peripheral benzodiazepine receptors (PBRs), a faithful marker for neuronal damage, was noted. Animals treated 5 min after the development of toxic signs with benactyzine, trihexyphenidyl or caramiphen demonstrated control levels of PBR values, whereas scopolamine produced binding densities significantly above basal levels. Examined at the 10-min time point, scopolamine and trihexyphenidyl afforded no protection against brain damage and did not differ from TA-injected rats. All four drugs failed to significantly prevent the alterations when applied 20 min after onset of convulsions. Assessment of learning processes yielded similar results, where caramiphen exibited some protection at the 20-min time point. Our results show that caramiphen and benactyzine, agents with combined anticholinergic and antiglutamatergic pharmacological profiles, offer considerable shielding against sarin, even when their administration is delayed.


Assuntos
Antídotos/uso terapêutico , Benactizina/uso terapêutico , Substâncias para a Guerra Química/intoxicação , Inibidores da Colinesterase/intoxicação , Ciclopentanos/uso terapêutico , Sarina/intoxicação , Animais , Comportamento Animal/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
12.
Toxicol Appl Pharmacol ; 227(2): 265-74, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18164360

RESUMO

Freely moving rats were exposed to sarin vapor (34.2+/-0.8 microg/l) for 10 min. Mortality at 24 h was 35% and toxic sings in the surviving rats ranged from sever (prolonged convulsions) through moderate to almost no overt signs. Some of the surviving rats developed delayed, intermittent convulsions. All rats were evaluated for long-term functional deficits in comparison to air-exposed control rats. Histological analysis revealed typical cell loss at 1 week post inhalation exposure. Neuronal inflammation was demonstrated by a 20-fold increase in prostaglandin (PGE(2)) levels 24 h following exposure that markedly decreased 6 days later. An additional, delayed increase in PGE(2) was detected at 1 month and continued to increase for up to 6 months post exposure. Glial activation following neural damage was demonstrated by an elevated level of peripheral benzodiazepine receptors (PBR) seen in the brain 4 and 6 months after exposure. At the same time muscarinic receptors were unaffected. Six weeks, four and six months post exposure behavioral evaluations were performed. In the open field, sarin-exposed rats showed a significant increase in overall activity with no habituation over days. In a working memory paradigm in the water maze, these same rats showed impaired working and reference memory processes with no recovery. Our data suggest long lasting impairment of brain functions in surviving rats following a single sarin exposure. Animals that seem to fully recover from the exposure, and even animals that initially show no toxicity signs, developed some adverse neural changes with time.


Assuntos
Substâncias para a Guerra Química/toxicidade , Inibidores da Colinesterase/toxicidade , Neurônios/patologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/psicologia , Sarina/toxicidade , Administração por Inalação , Animais , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Inibidores da Colinesterase/administração & dosagem , Inibidores da Colinesterase/análise , Cognição/efeitos dos fármacos , Dinoprostona/metabolismo , Gases , Dose Letal Mediana , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória de Curto Prazo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Receptores Muscarínicos/efeitos dos fármacos , Receptores Muscarínicos/metabolismo , Sarina/administração & dosagem , Sarina/análise
13.
Arch Toxicol ; 81(5): 353-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17009047

RESUMO

The extrapolation from animal data to therapeutic effects in humans, a basic pharmacological issue, is especially critical in studies aimed to estimate the protective efficacy of drugs against nerve agent poisoning. Such efficacy can only be predicted by extrapolation of data from animal studies to humans. In pretreatment therapy against nerve agents, careful dose determination is even more crucial than in antidotal therapy, since excessive doses may lead to adverse effects or performance decrements. The common method of comparing dose per body weight, still used in some studies, may lead to erroneous extrapolation. A different approach is based on the comparison of plasma concentrations at steady state required to obtain a given pharmacodynamic endpoint. In the present study, this approach was applied to predict the prophylactic efficacy of the anticholinergic drug caramiphen in combination with pyridostigmine in man based on animal data. In two species of large animals, dogs and monkeys, similar plasma concentrations of caramiphen (in the range of 60-100 ng/ml) conferred adequate protection against exposure to a lethal-dose of sarin (1.6-1.8 LD(50)). Pharmacokinetic studies at steady state were required to achieve the correlation between caramiphen plasma concentrations and therapeutic effects. Evaluation of total plasma clearance values was instrumental in establishing desirable plasma concentrations and minimizing the number of animals used in the study. Previous data in the literature for plasma levels of caramiphen that do not lead to overt side effects in humans (70-100 ng/ml) enabled extrapolation to expected human protection. The method can be applied to other drugs and other clinical situations, in which human studies are impossible due to ethical considerations. When similar dose response curves are obtained in at least two animal models, the extrapolation to expected therapeutic effects in humans might be considered more reliable.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Intoxicação por Organofosfatos , Intoxicação/prevenção & controle , Animais , Substâncias para a Guerra Química/intoxicação , Antagonistas Colinérgicos/administração & dosagem , Antagonistas Colinérgicos/farmacocinética , Antagonistas Colinérgicos/uso terapêutico , Ciclopentanos/sangue , Ciclopentanos/farmacocinética , Ciclopentanos/uso terapêutico , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Humanos , Infusões Intravenosas , Infusões Parenterais , Dose Letal Mediana , Masculino , Taxa de Depuração Metabólica , Organofosfatos/administração & dosagem , Organofosfatos/sangue , Papio anubis , Intoxicação/sangue , Brometo de Piridostigmina/sangue , Brometo de Piridostigmina/farmacocinética , Brometo de Piridostigmina/uso terapêutico , Sarina/administração & dosagem , Sarina/intoxicação , Especificidade da Espécie , Resultado do Tratamento
14.
Toxicology ; 233(1-3): 40-6, 2007 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-17045722

RESUMO

Extensive pharmacokinetic studies in both mice and rhesus macaques, with biochemically well defined forms of native and recombinant AChEs from bovine, rhesus and human origin, allowed us to determine an hierarchical pattern by which post-translation-related factors and specific amino-acid epitopes govern the pharmacokinetic performance of the enzyme molecule. In parallel, we demonstrated that controlled conjugation of polyethylene-glycol (PEG) side-chains to lysine residues of rHuAChE also results in the generation of active enzyme with improved pharmacokinetic performance. Here, we show that equally efficient extension of circulatory residence can be achieved by specific conditions of PEGylation, regardless of the post-translation-modification state of the enzyme. The masking effect of PEGylation, which is responsible for extending circulatory lifetime, also contributes to the elimination of immunological responses following repeated administration of AChE. Finally, in vivo protection studies in mice allowed us to determine that the PEGylated AChE protects the animal from a high lethal dose (2.5 LD(50)) of soman. On a mole basis, both the recombinant AChE and its PEGylated form provide higher levels of protection against soman poisoning than the native serum-derived HuBChE. The findings that circulatory long-lived PEGylated AChE can confer superior protection to mice against OP-compound poisoning while exhibiting reduced immunogenicity, suggest that this chemically modified version of rHuAChE may serve as a highly effective bioscavenger for prophylactic treatment against OP-poisoning.


Assuntos
Acetilcolinesterase , Inibidores da Colinesterase/toxicidade , Portadores de Fármacos/química , Fármacos Neuroprotetores , Síndromes Neurotóxicas/prevenção & controle , Polietilenoglicóis/química , Soman/toxicidade , Acetilcolinesterase/efeitos adversos , Acetilcolinesterase/biossíntese , Acetilcolinesterase/química , Acetilcolinesterase/farmacocinética , Animais , Anticorpos/sangue , Linhagem Celular , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Meia-Vida , Humanos , Dose Letal Mediana , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacocinética , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacocinética , Especificidade por Substrato
15.
Mol Pharmacol ; 70(3): 1121-31, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16801396

RESUMO

Comparative protection studies in mice demonstrate that on a molar basis, recombinant human acetylcholinesterase (rHuAChE) confers higher levels of protection than native human butyrylcholinesterase (HuBChE) against organophosphate (OP) compound intoxication. For example, mice challenged with 2.5 LD50 of O-isopropyl methylphosphonofluoridate (sarin), pinacolylmethyl phosphonofluoridate (soman), and O-ethyl-S-(2-isopropylaminoethyl) methylphosphonothiolate (VX) after treatment with equimolar amounts of the two cholinesterases displayed 80, 100, and 100% survival, respectively, when pre-treatment was carried out with rHuAChE and 0, 20, and 60% survival, respectively, when pretreatment was carried out with HuBChE. Kinetic studies and active site titration analyses of the tested OP compounds with acetylcholinesterases (AChEs) and butyrylcholinesterases (BChEs) from different mammalian species demonstrate that the superior in vivo efficacy of acetyl-cholinesterases is in accordance with the higher stereoselectivity of AChE versus BChE toward the toxic enantiomers comprising the racemic mixtures of the various OP agents. In addition, we show that polyethylene glycol-conjugated (PEGy-lated) rHuAChE, which is characterized by a significantly extended circulatory residence both in mice and monkeys ( Biochem J 357: 795-802, 2001 ; Biochem J 378: 117-128, 2004 ), retains full reactivity toward OP compounds both in vitro and in vivo and provides a higher level of protection to mice against OP poisoning, compared with native serum-derived HuBChE. Indeed, PEGylated rHuAChE also confers superior prophylactic protection when administered intravenously or intramuscularly over 20 h before exposure of mice to a lethal dose of VX (1.3-1.5 LD50). These findings together with the observations that the PEGylated rHuAChE exhibits unaltered biodistribution and high bioavailability present a case for using PEGylated rHuAChE as a very efficacious bioscavenger of OP agents.


Assuntos
Acetilcolinesterase/farmacocinética , Butirilcolinesterase/sangue , Butirilcolinesterase/farmacocinética , Compostos Organofosforados/toxicidade , Polietilenoglicóis/química , Proteínas Recombinantes/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Humanos , Dose Letal Mediana , Masculino , Camundongos , Compostos Organotiofosforados/toxicidade , Sarina/toxicidade , Soman/toxicidade , Análise de Sobrevida , Distribuição Tecidual/efeitos dos fármacos
16.
Toxicol Rev ; 25(4): 231-43, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17288495

RESUMO

The traditional therapeutic treatment of organophosphate cholinesterase inhibitor (nerve agents) poisoning consists of co-treatment with an antimuscarinic (atropine) and a reactivator of inhibited acetylcholinesterase (AChE), which contains a nucleophilic oxime function. Two oximes are presently widely available for clinical use, pralidoxime and obidoxime (toxogonin), but both offer little protection against important nerve agent threats. This has highlighted the real need for the development and availability of more effective oximes for human use, a search that has been going on for up to 30 years. However, despite the demonstration of more effective and safe oximes in animal experiments, no additional oximes have been licensed for human use. HI-6, (1-[[[4(aminocarbonyl)-pyridinio]methoxy]methyl]-2(hydroxyimino)pyridinium dichloride; CAS 34433-31-3) has been studied intensively and has been proved effective in a variety of species including non-human primates and appears from clinical experience to be safe in humans. These studies have led to the fielding of HI-6 for use against nerve agents by the militaries of the Czech republic, Sweden, Canada and under certain circumstances the Organisation for the Prohibition of Chemical Weapons. Nevertheless HI-6 has not been granted a license for clinical use, must be used only under restricted guidelines and is not available for civilian use as far as is known. This article will highlight those factors relating to HI-6 that pertain to the licensing of new compounds of this type, including the mechanism of action, the clinical and pre-clinical demonstration of safety and its efficacy against a variety of nerve agents particularly in non-human primates, since no relevant human population exists. This article also contains important data on the use of HI-6 in baboons, which has not been available previously. The article also discusses the possibility of successful therapy with HI-6 against poisoning in humans relative to doses used in non-human primates and relative to its ability to reactivate inhibited human AChE.


Assuntos
Antídotos/uso terapêutico , Substâncias para a Guerra Química/intoxicação , Reativadores da Colinesterase/uso terapêutico , Intoxicação por Organofosfatos , Intoxicação/tratamento farmacológico , Compostos de Piridínio/uso terapêutico , Animais , Antídotos/farmacocinética , Reativadores da Colinesterase/farmacocinética , Feminino , Humanos , Macaca mulatta , Masculino , Compostos Organofosforados/antagonistas & inibidores , Oximas , Papio , Intoxicação/fisiopatologia , Compostos de Piridínio/farmacocinética
17.
Ann N Y Acad Sci ; 1025: 584-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15542766

RESUMO

Several drugs of abuse are known to produce an array of deleterious effects, including alterations in neuronal circuitry and, ultimately, neuronal degeneration. For instance, methamphetamine was shown to induce substantial nigrostriatal dopaminergic terminal damage, including an increase in glial fibrillary acidic protein, a marker for astrocyte proliferation. Nevertheless, there was almost no attempt to define neurodegeneration by measuring the abundance of reactive microglia. In fact, some investigators fail to differentiate between astrocytes and microglia and claim glial fibrillary acidic protein to be a marker for gliosis. To date, there are numerous methods designed to assess brain neuropathologies resulting from a wide arsenal of insults. Regardless of the cause of neuronal damage, reactive glial cells always appear at and around the site of degeneration. These cells are distinguished by the exceptional abundance of peripheral benzodiazepine receptors (PBRs; omicron3 sites), particularly as compared to surrounding neurons. Measuring the binding of specific ligands to these PBRs (for example, [3H]PK 11195) offers a unique indirect marker for reliable impairment estimation in the central nervous system. Moreover, the availability of agents such as [11C]PK 11195 paved the road to in vivo animal and human brain positron emission tomography scanning, demonstrating inflammation-like processes in several diseases. Additionally, the measurement of increased binding of PBR ligands provides a faithful indicator for the behavioral and cognitive deficits accompanying neuronal injury.


Assuntos
Doenças Neurodegenerativas/metabolismo , Receptores de GABA-A/metabolismo , Animais , Benzodiazepinonas/metabolismo , Benzodiazepinonas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Humanos , Drogas Ilícitas/metabolismo , Drogas Ilícitas/farmacologia , Tomografia por Emissão de Pósitrons/métodos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia
18.
Ann N Y Acad Sci ; 998: 533-6, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14592924

RESUMO

We have shown that mucosal administration of recombinant fragments corresponding to the human acetylcholine receptor (AChR) alpha subunit suppresses chronic ongoing experimental autoimmune myasthenia gravis (EAMG) in rats. Treated animals exhibit a Th1 to Th2/Th3 shift in their cytokine profile and downregulation of costimulatory factors. However, application of a xenogeneic recombinant fragment may have limitations when considered as a possible approach for the treatment of MG in humans. We therefore tested the potential of a syngeneic fragment and of long synthetic peptides to suppress EAMG. We found that a syngeneic fragment corresponding to the extracellular region of the rat AChR alpha subunit was as effective as the formerly described human xenogeneic fragment in suppressing ongoing EAMG. This is encouraging in view of the potential use of mucosally administered recombinant AChR fragments for the treatment of MG in humans. However, in severely affected individuals, this antigen-specific approach may need to be supported by direct modulation of cytokines and costimulatory factors known to be involved in the pathogenesis of EAMG. To test the potential of this approach, myasthenic rats were injected by antibodies either to the proinflammatory cytokine IL-18 or to the costimulatory factor CD40L. These treatments act via different mechanisms, but both lead to the alleviation of clinical symptoms even when given at the chronic phase of EAMG. We suggest that antagonists to key cytokines and/or costimulatory factors be used to augment antigen-specific treatments of myasthenia such as mucosal administration of AChR recombinant fragments.


Assuntos
Citocinas/metabolismo , Tolerância Imunológica/fisiologia , Terapia de Imunossupressão , Miastenia Gravis/terapia , Animais , Modelos Animais de Doenças , Humanos , Imunidade nas Mucosas , Interleucina-2/metabolismo , Miastenia Gravis/imunologia , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/prevenção & controle , Subunidades Proteicas , Ratos , Receptores Colinérgicos/administração & dosagem , Receptores Colinérgicos/imunologia , Proteínas Recombinantes , Células Th1/imunologia , Células Th2/imunologia
19.
Toxicol Sci ; 75(1): 108-16, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12832655

RESUMO

Soman, a powerful inhibitor of acetylcholinesterase, causes an array of toxic effects in the central nervous system including convulsions, learning and memory impairments, and, ultimately, death. We report on the protection afforded by postexposure antidotal treatments, combined with pyridostigmine (0.1 mg/kg) pretreatment, against these consequences associated with soman poisoning. Scopolamine (0.1 mg/kg) or caramiphen (10 mg/kg) were administered 5 min after soman (1.2 LD50), whereas TAB (i.e., TMB4, atropine, and benactyzine, 7.5, 3, and 1 mg/kg, respectively) was injected in rats concomitant with the development of toxic signs. Atropine (4 mg/kg) was given to the two former groups at the onset of toxic symptoms. Caramiphen and TAB completely abolished electrographic seizure activity while scopolamine treatment exhibited only partial protection. Additionally, no significant alteration in the density of peripheral benzodiazepine receptors was noted following caramiphen or TAB administration, while scopolamine application resulted in a complex outcome: a portion of the animals demonstrated no change in the number of these sites whereas the others exhibited markedly higher densities. Cognitive functions (i.e., learning and memory processes) evaluated using the Morris water maze improved considerably by the three treatments when compared to soman-injected animals; the following rank order was observed: caramiphen > TAB > scopolamine. Additionally, statistically significant correlations (r = 0.72, r = 0.73) were demonstrated between two learning parameters and [3H]Ro5-4864 binding to brain membrane. These results show that drugs with a pharmacological profile consisting of anticholinergic and antiglutamatergic properties such as caramiphen and TAB, have a substantial potential as postexposure therapies against intoxication by organophosphates.


Assuntos
Antídotos/farmacologia , Encéfalo/efeitos dos fármacos , Antagonistas Colinérgicos/farmacologia , Inibidores da Colinesterase/intoxicação , Transtornos Cognitivos/prevenção & controle , Antagonistas de Aminoácidos Excitatórios/farmacologia , Soman/intoxicação , Animais , Benzodiazepinonas/metabolismo , Encéfalo/metabolismo , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/fisiopatologia , Eletroencefalografia , Técnicas In Vitro , Ligantes , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
J Neurochem ; 84(3): 432-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12558962

RESUMO

There are numerous methods designed to monitor brain neuropathologies resulting from a wide arsenal of insults. Regardless of the cause of neuronal death, reactive glial cells always appear at and around the site of degeneration. These cells are distinguished by the exceptional abundance of peripheral benzodiazepine receptors, particularly compared with surrounding neurons. Measuring the binding of specific ligands to these peripheral benzodiazepine receptors offers a unique indirect marker for reliable damage assessment in the CNS and a faithful indicator for the accompanying cognitive deficits.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Gliose/diagnóstico , Receptores de GABA-A/metabolismo , Tomografia Computadorizada de Emissão , Animais , Autorradiografia , Benzodiazepinonas/farmacocinética , Encéfalo/patologia , Radioisótopos de Carbono , Gliose/diagnóstico por imagem , Gliose/patologia , Humanos , Isoquinolinas/farmacocinética , Ligantes , Camundongos , Valor Preditivo dos Testes , Ratos , Receptores de GABA-A/análise , Especificidade por Substrato , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...