Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Interact ; 259(Pt B): 182-186, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27000540

RESUMO

The atypical butyrylcholinesterase (aBuChE) from Oryzias latipes shares approximately 65% sequence similarity to both acetylcholinesterase and butyrylcholinesterase and was studied for its capacity to spontaneously reactivate following inhibition by organophosphorus nerve agents. Like other cholinesterases, aBuChE was inhibited by all G- and V-type nerve agents. Interestingly, aBuChE was able to undergo spontaneous reactivation after inhibition with VR (t1/2 = 5.5 ± 0.2 h). Mass spectrometry of aBuChE after VR inhibition confirmed the presence of a covalently bound adduct of the size expected for non-aged VR on the peptide containing the active site serine. To understand the effect of substrate volume on rates of reactivation, the capacity of aBuChE to bind and spontaneously reactivate after inhibition with five V-agent analogues was examined. No appreciable reactivation was detected for enzyme inhibited by V2 (VX with O-isopropyl on retained group), V4 (VX with N-diethyl leaving group termination), or V5 (VX with N-dimethyl leaving group termination). Minimal reactivation was detected with V1 (VX with O-propyl on retained group). Conversely, spontaneous reactivation was observed when aBuChE was inhibited by V3 (VX with O-isobutyl on retained group; t1/2 = 6.3 ± 0.4 h). The data suggest that the ability of aBuChE to spontaneously reactivate after inhibition by V-agent analogues is related to the structure of the retained group. These results provide structural information that may shed light on the design of improved small molecule reactivators of nerve agent-inhibited acetylcholinesterase or butyrylcholinesterase, and further suggest that re-engineering the active site of a cholinesterase could result in enzymes with clinically relevant rates of nerve agent hydrolysis.


Assuntos
Butirilcolinesterase/química , Compostos Organotiofosforados/química , Animais , Butirilcolinesterase/genética , Butirilcolinesterase/metabolismo , Domínio Catalítico , Meia-Vida , Cinética , Larva/metabolismo , Espectrometria de Massas , Mariposas/crescimento & desenvolvimento , Mariposas/metabolismo , Compostos Organotiofosforados/metabolismo , Oryzias/metabolismo , Ligação Proteica , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação
2.
Bioorg Med Chem Lett ; 24(7): 1711-4, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24630558

RESUMO

The goal of this research was to identify structurally novel, non-quaternarypyridinium reactivators of GF (cyclosarin)-inhibited hAChE that possess the capacity to mediate in vitro reactivation of GF-inhibited human acetylcholinesterase (hAChE). New compounds were designed, synthesized and assessed in GF-inhibited hAChE assays. Structure activity relationships for AChE binding and reactivation of GF-inhibited hAChE were developed. Lead compounds from two different chemical series, represented by compounds 17 and 38, displayed proficient in vitro reactivation of GF-inhibited hAChE, while also possessing low inhibition of native enzyme.


Assuntos
Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Desenho de Fármacos , Compostos Organofosforados/farmacologia , Inibidores da Colinesterase/síntese química , Inibidores da Colinesterase/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Compostos Organofosforados/síntese química , Compostos Organofosforados/química , Relação Estrutura-Atividade
3.
Chem Biol Interact ; 203(1): 72-6, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-22982773

RESUMO

Administration of oxime therapy is currently the standard approach used to reverse the acute toxicity of organophosphorus (OP) compounds, which is usually attributed to OP inhibition of acetylcholinesterase (AChE). Rate constants for reactivation of OP-inhibited AChE by even the best oximes, such as HI-6 and obidoxime, can vary >100-fold between OP-AChE conjugates that are easily reactivated and those that are difficult to reactivate. To gain a better understanding of this oxime specificity problem for future design of improved reactivators, we conducted a QSAR analysis for oxime reactivation of AChE inhibited by OP agents and their analogues. Our objective was to identify common mechanism(s) among OP-AChE conjugates of phosphates, phosphonates and phosphoramidates that result in resistance to oxime reactivation. Our evaluation of oxime reactivation of AChE inhibited by a sarin analogue, O-methyl isopropylphosphonofluoridate, or a cyclosarin analogue, O-methyl cyclohexylphosphonofluoridate, indicated that AChE inhibited by these analogues was at least 70-fold more difficult to reactivate than AChE inhibited by sarin or cyclosarin. In addition, AChE inhibited by an analogue of tabun (i.e., O-ethyl isopropylphosphonofluoridate) was nearly as resistant to reactivation as tabun-inhibited AChE. QSAR analysis of oxime reactivation of AChE inhibited by these OP compounds and others suggested that the presence of both a large substituent (i.e., ≥ the size of dimethylamine) and an alkoxy substituent in the structure of OP compounds is the common feature that results in resistance to oxime reactivation of OP-AChE conjugates whether the OP is a phosphate, phosphonate or phosphoramidate.


Assuntos
Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/toxicidade , Reativadores da Colinesterase/farmacologia , Compostos Organofosforados/toxicidade , Oximas/farmacologia , Inibidores da Colinesterase/química , Proteínas Ligadas por GPI/metabolismo , Humanos , Cinética , Cloreto de Obidoxima/química , Cloreto de Obidoxima/farmacologia , Compostos Organofosforados/química , Oximas/química , Compostos de Piridínio/química , Compostos de Piridínio/farmacologia , Relação Quantitativa Estrutura-Atividade , Proteínas Recombinantes/metabolismo , Sarina/análogos & derivados , Sarina/química , Sarina/toxicidade
4.
Cell Mol Neurobiol ; 31(6): 909-20, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21538119

RESUMO

Electrophysiological and ultrastructural studies were performed on phrenic nerve-hemidiaphragm preparations isolated from wild-type and acetylcholinesterase (AChE) knockout (KO) mice to determine the compensatory mechanisms manifested by the neuromuscular junction to excess acetylcholine (ACh). The diaphragm was selected since it is the primary muscle of respiration, and it must adapt to allow for survival of the organism in the absence of AChE. Nerve-elicited muscle contractions, miniature endplate potentials (MEPPs) and evoked endplate potentials (EPPs) were recorded by conventional electrophysiological techniques from phrenic nerve-hemidiaphragm preparations isolated from 1.5- to 2-month-old wild-type (AChE(+/+)) or AChE KO (AChE(-/-)) mice. These recordings were chosen to provide a comprehensive assessment of functional alterations of the diaphragm muscle resulting from the absence of AChE. Tension measurements from AChE(-/-) mice revealed that the amplitude of twitch tensions was potentiated, but tetanic tensions underwent a use-dependent decline at frequencies below 70 Hz and above 100 Hz. MEPPs recorded from hemidiaphragms of AChE(-/-) mice showed a reduction in frequency and a prolongation in decay (37%) but no change in amplitude compared to values observed in age-matched wild-type littermates. In contrast, MEPPs recorded from hemidiaphragms of wild-type mice that were exposed for 30 min to the selective AChE inhibitor 5-bis(4-allyldimethyl-ammoniumphenyl)pentane-3-one (BW284C51) exhibited a pronounced increase in amplitude (42%) and a more marked prolongation in decay (76%). The difference between MEPP amplitudes and decays in AChE(-/-) hemidiaphragms and in wild-type hemidiaphragms treated with BW284C51 represents effective adaptation by the former to a high ACh environment. Electron microscopic examination revealed that diaphragm muscles of AChE(-/-) mice had smaller nerve terminals and diminished pre- and post-synaptic surface contacts relative to neuromuscular junctions of AChE(+/+) mice. The morphological changes are suggested to account, in part, for the ability of muscle from AChE(-/-) mice to function in the complete absence of AChE.


Assuntos
Acetilcolinesterase/deficiência , Acetilcolinesterase/metabolismo , Colina/metabolismo , Sinapses/fisiologia , Sinapses/ultraestrutura , Potenciais de Ação/efeitos dos fármacos , Animais , Conotoxinas/farmacologia , Diafragma/efeitos dos fármacos , Diafragma/inervação , Diafragma/fisiologia , Potenciais Evocados/efeitos dos fármacos , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Knockout , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Tono Muscular/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
5.
Toxicol Appl Pharmacol ; 231(2): 157-64, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18508103

RESUMO

A structure-activity analysis was used to evaluate the variation in oxime efficacy of 2-PAM, obidoxime, HI-6 and ICD585 against nerve agents. In vivo oxime protection and in vitro oxime reactivation were used as indicators of oxime efficacy against VX, sarin, VR and cyclosarin. Analysis of in vivo oxime protection was conducted with oxime protective ratios (PR) from guinea pigs receiving oxime and atropine therapy after sc administration of nerve agent. Analysis of in vitro reactivation was conducted with second-order rate contants (k(r2)) for oxime reactivation of agent-inhibited acetylcholinesterase (AChE) from guinea pig erythrocytes. In vivo oxime PR and in vitro k(r2) decreased as the volume of the alkylmethylphosphonate moiety of nerve agents increased from VX to cyclosarin. This effect was greater with 2-PAM and obidoxime (>14-fold decrease in PR) than with HI-6 and ICD585 (<3.7-fold decrease in PR). The decrease in oxime PR and k(r2) as the volume of the agent moiety conjugated to AChE increased was consistent with a steric hindrance mechanism. Linear regression of log (PR-1) against log (k(r2)[oxime dose]) produced two offset parallel regression lines that delineated a significant difference between the coupling of oxime reactivation and oxime protection for HI-6 and ICD585 compared to 2-PAM and obidoxime. HI-6 and ICD585 appeared to be 6.8-fold more effective than 2-PAM and obidoxime at coupling oxime reactivation to oxime protection, which suggested that the isonicotinamide group that is common to both of these oximes, but absent from 2-PAM and obidoxime, is important for oxime efficacy.


Assuntos
Inibidores da Colinesterase/toxicidade , Reativadores da Colinesterase/farmacologia , Compostos Organofosforados/toxicidade , Oximas/farmacologia , Acetilcolinesterase/efeitos dos fármacos , Acetilcolinesterase/metabolismo , Animais , Atropina/farmacologia , Substâncias para a Guerra Química/toxicidade , Reativadores da Colinesterase/química , Eritrócitos/enzimologia , Cobaias , Modelos Lineares , Masculino , Cloreto de Obidoxima/farmacologia , Compostos Organotiofosforados/toxicidade , Oximas/química , Compostos de Pralidoxima/farmacologia , Compostos de Piridínio/farmacologia , Sarina/toxicidade , Relação Estrutura-Atividade
6.
J Anal Toxicol ; 32(1): 86-91, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18269799

RESUMO

The organophosphorus (OP) nerve agent soman (GD) contains two chiral centers (a carbon and a phosphorus atom), resulting in four stereoisomers (C+P+, C-P+, C+P-, and C-P-); the P- isomers exhibit a mammalian toxicity that is approximately 1000-fold greater than that of the P+ isomers. The capacity to assess the binding or hydrolysis of each of the four stereoisomers is an important tool in the development of enzymes with the potential to protect against GD intoxication. Using a gas chromatography-mass spectrometry-based approach, we have examined the capacity of plasma-derived human serum albumin, plasma-purified human butyrylcholinesterase, goat milk-derived recombinant human butyrylcholinesterase, and recombinant human paraoxonase 1 to interact with each of the four stereoisomers of GD in vitro at pH 7.4 and 25 degrees C. Under these experimental conditions, the butyrylcholinesterase samples were found to bind GD with a relative preference for the more toxic stereoisomers (C-P- > C+P- > C-P+ > C+P+), while human serum albumin and paraoxonase 1 interacted with GD with a relative preference for the less toxic isomers (C-P+/C+P+ > C+P-/C-P-). The results indicate that these human proteins exhibit distinct stereoselective interactions with GD. The approach described presents a means to rapidly assess substrate stereospecificity, supporting future efforts to develop more effective OP bioscavenger proteins.


Assuntos
Proteínas Sanguíneas/química , Cromatografia Gasosa-Espectrometria de Massas/métodos , Soman/química , Arildialquilfosfatase/química , Arildialquilfosfatase/metabolismo , Proteínas Sanguíneas/metabolismo , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/metabolismo , Catálise , Substâncias para a Guerra Química/química , Substâncias para a Guerra Química/metabolismo , Inibidores da Colinesterase/química , Inibidores da Colinesterase/metabolismo , Humanos , Cinética , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Albumina Sérica/química , Albumina Sérica/metabolismo , Soman/metabolismo , Estereoisomerismo
7.
FEBS J ; 274(5): 1183-91, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17286579

RESUMO

Human serum paraoxonase 1 (HuPON1; EC 3.1.8.1) is a calcium-dependent six-fold beta-propeller enzyme that has been shown to hydrolyze an array of substrates, including organophosphorus (OP) chemical warfare nerve agents. Although recent efforts utilizing site-directed mutagenesis have demonstrated specific residues (such as Phe222 and His115) to be important in determining the specificity of OP substrate binding and hydrolysis, little effort has focused on the substrate stereospecificity of the enzyme; different stereoisomers of OPs can differ in their toxicity by several orders of magnitude. For example, the C+/-P- isomers of the chemical warfare agent soman (GD) are known to be more toxic by three orders of magnitude. In this study, the catalytic activity of HuPON1 towards each of the four chiral isomers of GD was measured simultaneously via chiral GC/MS. The catalytic efficiency (k(cat)/K(m)) of the wild-type enzyme for the various stereoisomers was determined by a simultaneous solution of hydrolysis kinetics for each isomer. Derived k(cat)/K(m) values ranged from 625 to 4130 mm(-1).min(-1), with isomers being hydrolyzed in the order of preference C+P+ > C-P+ > C+P- > C-P-. The results indicate that HuPON1 hydrolysis of GD is stereoselective; substrate stereospecificity should be considered in future efforts to enhance the OPase activity of this and other candidate bioscavenger enzymes.


Assuntos
Arildialquilfosfatase/sangue , Arildialquilfosfatase/química , Substâncias para a Guerra Química/metabolismo , Soman/metabolismo , Arildialquilfosfatase/genética , Arildialquilfosfatase/metabolismo , Arildialquilfosfatase/fisiologia , Catálise , Substâncias para a Guerra Química/análise , Substâncias para a Guerra Química/química , Substâncias para a Guerra Química/toxicidade , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Cinética , Soman/análise , Soman/química , Soman/toxicidade , Estereoisomerismo , Especificidade por Substrato
8.
Toxicology ; 233(1-3): 31-9, 2007 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-17188793

RESUMO

Currently fielded treatments for nerve agent intoxication promote survival, but do not afford complete protection against either nerve agent-induced motor and cognitive deficits or neuronal pathology. The use of human plasma-derived butyrylcholinesterase (HuBuChE) to neutralize the toxic effects of nerve agents in vivo has been shown to both aid survival and protect against decreased cognitive function after nerve agent exposure. Recently, a commercially produced recombinant form of human butyrylcholinesterase (r-HuBuChE; PharmAthene Inc.) expressed in the milk of transgenic goats has become available. This material is biochemically similar to plasma-derived HuBuChE in in vitro assays. The pharmacokinetic characteristics of a polyethylene glycol coated (pegylated) form of r-HuBuChE were determined in guinea pigs; the enzyme was rapidly bioavailable with a half-life (t(1/2)) and pharmacokinetic profile that resembled that of plasma-derived huBuChE. Guinea pigs were injected with 140mg/kg (i.m.) of pegylated r-HuBuChE 18h prior to exposure (sc) to 5.5xLD(50) VX or soman. VX and soman were administered in a series of three injections of 1.5xLD(50), 2.0xLD(50), and 2.0xLD(50), respectively, with injections separated by 2h. Pretreatment with pegylated r-HuBuChE provided 100% survival against multiple lethal doses of VX and soman. Guinea pigs displayed no signs of nerve agent toxicity following exposure. Assessments of motor activity, coordination, and acquisition of spatial memory were performed for 2 weeks following nerve agent exposure. There were no measurable decreases in motor or cognitive function during this period. In contrast, animals receiving 1.5xLD(50) challenges of soman or VX and treated with standard atropine, 2-PAM, and diazepam therapy showed 50 and 100% survival, respectively, but exhibited marked decrements in motor function and, in the case of GD, impaired spatial memory acquisition. The advances in this field have resulted in the decision to select both the plasma-derived and the recombinant form of BuChE for advanced development and transition to clinical trials. Efforts have now been expanded to identify a catalytic protein capable of not only binding, but also rapidly hydrolyzing the standard threat nerve agents. Recent work has focused on paraoxonase-1 (PON1), a naturally occurring human serum enzyme with the capacity to catalyze the hydrolysis of nerve agents, albeit too slowly to afford dramatic protection. Using rational design, several amino acids involved in substrate binding have been identified and site-directed mutations have revealed that residue H115 plays an important role in binding. In addition, the stereospecificity of PON1 for the catalytic hydrolysis of soman has been examined. The enzyme exhibits a slight stereospecificity for the C+P+ isomer of soman, which is due more to preferential binding than to selective hydrolysis of this isomer. The results suggest that it may be possible to engineer a mutant form of PON1 with enhanced activity and stereospecificity for the most toxic nerve agent isoforms.


Assuntos
Butirilcolinesterase , Inibidores da Colinesterase/toxicidade , Fármacos Neuroprotetores , Síndromes Neurotóxicas/prevenção & controle , Compostos Organoplatínicos/toxicidade , Animais , Butirilcolinesterase/química , Butirilcolinesterase/farmacologia , Butirilcolinesterase/uso terapêutico , Catálise , Inibidores da Colinesterase/química , Humanos , Dose Letal Mediana , Modelos Moleculares , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Compostos Organoplatínicos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Estereoisomerismo
9.
Arch Toxicol ; 80(11): 756-60, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16770629

RESUMO

The hypothesis that acetylcholinesterase (AChE) inhibition is the mechanism of toxicity of organophosphorus (OP) compounds was examined by mathematically modeling the in vivo lethal effects of OP compounds and determining the amount of variation in OP toxicity that is explained by AChE inhibition. Mortality dose-response curves for several OP compounds (i.e., VX, soman, cyclosarin, sarin, tabun, diisopropylfluorophosphate and paraoxon) exhibited steep probit slopes (> 9.6) in guinea pigs. Steep probit slopes were also observed when the mortality dose-response curves for soman were examined in mice, rats, rabbits and non-human primates. The consistently steep probit slopes of the dose-response curves for highly toxic OP compounds suggested that these compounds have a single specific mechanism of toxicity regardless of the OP compound or the species in which it was tested. Regression analysis indicated that 93% of the 3,280-fold variation in the median lethal doses (i.e., LD(50)) of OP compounds in rats was explained by the variation in their in vitro rate constants for inhibition of AChE. Conversely, 91% of the 23-fold variation in the ability of the oximes pralidoxime and obidoxime to protect against the toxicity of OP compounds in guinea pigs was explained by the variation in the in vitro ability of oximes to reactivate OP-inhibited AChE. The best explanation for this variety of observations was that the primary mechanism of in vivo toxicity for highly toxic OP compounds is the inhibition of AChE, and the residual unexplained variation in OP toxicity that might be explained by other mechanisms represents < 10% of the total variation in OP toxicity.


Assuntos
Inibidores da Colinesterase/toxicidade , Compostos Organofosforados/toxicidade , Animais , Reativadores da Colinesterase/farmacologia , Cobaias , Dose Letal Mediana , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Biológicos , Cloreto de Obidoxima/farmacologia , Compostos de Pralidoxima/farmacologia , Coelhos , Ratos , Ratos Sprague-Dawley
10.
Arch Toxicol ; 80(11): 719-31, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16718492

RESUMO

A physiologically based pharmacokinetic (PB/PK) model has been developed in advanced computer simulation language (ACSL) to describe blood and tissue concentration-time profiles of the C(+/-)P(-) stereoisomers of soman after inhalation, subcutaneous and intravenous exposures at low (0.8-1.0 x LD(50)), medium (2-3 x LD(50)) and high (6 x LD(50)) levels of soman challenge in three species (rat, guinea pig, marmoset). Allometric formulae were used to compute the compartment volumes, blood flow rates, tidal volume and respiratory rate based upon total animal weight. Blood/tissue partition coefficients for soman, initial carboxylesterase and acetylcholinesterase levels and the rate constants for interactions between soman and these enzymes were species-dependent and were obtained from in vitro measurements reported in the literature. The model incorporated arterial and venous blood, lung, kidney, liver, richly perfused, poorly perfused and fat tissue compartments as well as subcutaneous and nasal exposure site compartments. First-order absorption from linearly filled soman deposits into metabolizing exposure site compartments was employed to model subcutaneous and inhalation exposures. The model was validated by comparing the predicted and observed values for C(+/-)P(-)-soman in arterial blood at various times following exposure and by regression analysis. Sensitivity analysis was used to determine the effects of perturbations in the model parameters on the time-course of arterial C(-)P(-)-soman concentrations for different exposure routes. In our evaluation of 28 datasets, predicted values were generally within 95% confidence limits of the observed values, and regression coefficients comparing predicted and observed data were greater than 0.85 for 95% of the intravenous and subcutaneous datasets and 25% of the inhalation datasets. We conclude that the model predicts the soman toxicokinetics for doses >or=1 x LD(50) for intravenous and subcutaneous exposures and inhalation exposures of 8 min or less sufficiently well to allow its use in the modeling of bioscavenger protection.


Assuntos
Substâncias para a Guerra Química/farmacocinética , Inibidores da Colinesterase/farmacocinética , Modelos Biológicos , Soman/farmacocinética , Administração por Inalação , Animais , Callithrix , Inibidores da Colinesterase/sangue , Simulação por Computador , Cobaias , Injeções Intravenosas , Injeções Subcutâneas , Masculino , Ratos , Ratos Wistar , Soman/sangue
11.
Environ Toxicol Pharmacol ; 17(2): 87-94, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21782718

RESUMO

The proinflammatory cytokine human interleukin-6 (hIL-6) plays an important role in the early and late courses of inflammation, trauma, and wound healing caused by sulfur mustard (HD). Previously, we demonstrated that hIL-6 might be involved in the early event of structural changes of the signal transducer glycoprotein, which indirectly initiates the cascade of events, such as skin irritation and blister formation observed in the pathophysiology of HD injury. In this present work, we focus on the neutralization effect of IL-6 antibodies with regard to the modulation of hIL-6 secretion. Levels of secreted cytokine hIL-6 in normal human epidermal keratinocytes (NHEK) stimulated with HD (10(-4)M) and incubated for 24h at 37°C were determined by enzyme immunoassay, protein immunocytologic assay and reverse-transcriptase-polymerase chain reaction (RT-PCR). The ratio of HD-treated NHEK to constitutive non-stimulated NHEK controls (S/C) on the induction of hIL-6 is reported. S/C was four-fold higher than non-stimulated NHEK controls as determined by ELISA. By using a more sensitive immunocytologic assay, Luminex(100)™, the increment was verified. hIL-6 levels in NHEK stimulated with HD were 21±11ng/mL as measured by Luminex(100)™. The messenger RNA expression of the cytokine (hIL-6) gene was analyzed semiquantitatively. RT-PCR demonstrated that HD induced an increase in the transcription of hIL-6 gene. Selective immunosuppression, using IL-6 neutralizing antibodies, led to a reduction of such expression of HD-induced transcription of hIL-6 in human keratinocytes. The neutralization by pre-incubating NHEK with monoclonal anti-IL6 antibodies decreased hIL-6 secretion by 76%±1.8 ((*)P<0.05).

12.
J Neurosci Methods ; 128(1-2): 53-65, 2003 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-12948548

RESUMO

Many researchers are familiar with the spreadsheet capabilities of Microsoft Excel, but have never explored using customized VISUAL BASIC FOR APPLICATIONS (VBA) macros embedded in the program. At the United States Army Medical Research Institute of Chemical Defense (USAMRICD), the implementation of VBA program code to carry out repetitive operations has resulted in a tremendous savings in both the time and manpower required to reliably capture, analyze, and plot data from research protocols. A set of "template" workbooks was developed and is used to organize data from different types of studies. At the heart of the concept is a "setup information" worksheet onto which the user enters information about the study (i.e. the number of subjects, groups, graphs). Clicking a control button on this worksheet launches the VBA code that creates worksheets for each subject, group and chart specified. A "pairmatch" function allows the user to create groups either randomly or by pair matching based on user-specified variables. Controls are provided that run VBA program code to parse subject data files into the proper worksheets and perform group averages. Charts are updated automatically whenever group averages change so that the researcher always has an up-to-date plot available. The ability to reformat the captured data onto a "statistical output" worksheet allows data to be imported easily to statistical software packages. The concept is being used for several different types of studies at USAMRICD and has significantly reduced not only amount of time spent on data management, but also the number of data entry errors. Readers interested in acquiring an electronic copy of the startle workbook example, which contains the complete VBA code, should send the request to the authors at reseco@erols.com or maurice.sipos@us.army.mil.


Assuntos
Estimulação Acústica/métodos , Sistemas de Gerenciamento de Base de Dados/normas , Reflexo de Sobressalto/fisiologia , Software , Animais , Substâncias para a Guerra Química/farmacologia , Sistemas de Gerenciamento de Base de Dados/estatística & dados numéricos , Cobaias , Reflexo de Sobressalto/efeitos dos fármacos , Software/estatística & dados numéricos
13.
Math Biosci ; 181(2): 133-43, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12445758

RESUMO

The ability of certain organophosphorus (OP) compounds to inhibit acetylcholinesterase (AChE) has made them useful for industrial (insecticides) and military (nerve agents) purposes. We have previously published a single compartment mathematical model of the interactions between OP nerve agents and the enzymes affected by these agents. That model, which could be used to predict the LD50 of seven nerve agents in rats, has been extended to include the protective actions of stoichiometric and catalytic OP-scavenger enzymes (delivered as pretreatments) so that protective ratios attributable to the scavengers may be predicted. Prediction of expected human protection from in vitro rate constant and initial enzyme level measurements is the ultimate goal for this work. The enhanced model predicts the LD50 from rate constants of the OP agent's binding reactions with AChE, carboxylesterase (CaE) and a stoichiometric scavenger (S); a first-order OP elimination rate (including a contribution due to a catalytic scavenger); and whole body estimates of AChE, CaE and S. The ratio of the scavenger-treated LD50 estimate to the scavenger-free LD50 estimate provided a theoretical expression describing the scavenger's contributions to the protective ratio. Published in vivo protective ratios for two stoichiometric scavengers (fetal bovine serum AChE and human utyrylcholinesterase) against challenge by several OP agents in mice were compared with ratios predicted by the model. A linear regression analysis of in vivo protective ratios in mice versus the ratios predicted by the model from the in vitro measurements resulted in an R(2) value of 0.902. The catalytic scavenger portion of the theory could not be validated due to a lack of published data. We conclude that the one-compartment model can be used to make reasonable estimates of the protective ratio attributable to stoichiometric scavengers, but can make no conclusions regarding the ability of the model to predict catalytic scavenger protection ratios.


Assuntos
Antídotos/uso terapêutico , Inibidores da Colinesterase/intoxicação , Colinesterases/uso terapêutico , Modelos Biológicos , Intoxicação por Organofosfatos , Animais , Antídotos/metabolismo , Catálise , Inibidores da Colinesterase/farmacocinética , Colinesterases/metabolismo , Humanos , Cinética , Dose Letal Mediana , Modelos Lineares , Camundongos , Compostos Organofosforados/antagonistas & inibidores , Compostos Organofosforados/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...