Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Ther ; 37(7): 3234-3245, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32440976

RESUMO

INTRODUCTION: Amenamevir (ASP2151), a herpesvirus helicase-primase inhibitor, is currently used for the treatment of herpes zoster in Japan. Amenamevir is mainly metabolized in the liver, and urinary excretion of amenamevir is approximately 10% in healthy adults. The increase of systemic exposure in non-dialysis patients with severe renal impairment was much less than that associated with nucleoside antiviral agents. The aim of this study was to evaluate the pharmacokinetics and dialyzability of a single oral dose (400 mg) of amenamevir in hemodialysis patients. METHODS: This was a single-arm, open-label, multicenter clinical pharmacology study. Nine patients aged 20-80 years with end-stage kidney disease and undergoing maintenance hemodialysis three times weekly were enrolled. Pharmacokinetics and dialyzability were investigated by serial collection of blood samples until 48 h post-dose during the study. RESULTS: The maximum plasma concentration and time to reach maximum plasma concentration during 24 h post-dose were 1585 ng/mL and 6.2 h, respectively. The area under the plasma concentration-time curve (AUC) from time zero to 24 h was 23,890 ng h/mL. The median terminal elimination half-life within 24 h before, during, and after hemodialysis was 14.7, 15.2, and 12.4 h, respectively. The AUC in hemodialysis patients was approximately double that in healthy adults. This increase in AUC was much less than that reported in nucleoside antiviral agents. The hemodialysis clearance, elimination fraction percentage, and amount of amenamevir removed were 37.8 mL/min, 28.1%, and 132.0 µg, respectively. The amount of amenamevir removed by hemodialysis was minimal. None of the hemodialysis parameters were associated with serum albumin. This study revealed no clinically relevant safety concerns. CONCLUSION: There were no clinically relevant safety concerns when 400 mg of amenamevir was administered as a single dose to hemodialysis patients without dose adjustment and/or modification of the dosing schedule. TRIAL REGISTRATION: JapicCTI-184242.


Assuntos
Antivirais/efeitos adversos , Antivirais/uso terapêutico , Herpes Zoster/tratamento farmacológico , Oxidiazóis/sangue , Oxidiazóis/farmacocinética , Oxidiazóis/uso terapêutico , Insuficiência Renal/terapia , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Diálise Renal , Adulto Jovem
2.
Invest New Drugs ; 38(4): 1067-1076, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31471863

RESUMO

Purpose BAL101553, the prodrug of the microtubule-destabilizer BAL27862, previously showed signs of antitumor activity when administered as a 2-h infusion, but its use was limited by vascular toxicity. We investigated an alternative dosing strategy aimed at improving the safety profile of BAL101553. Methods This multicenter, open-label, Phase 1 dose-escalation study used a 3 + 3 design to determine the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), pharmacokinetics, and antitumor activity of BAL101553 administered as a 48-h IV infusion on Days 1, 8, and 15 of a 28-day cycle. Patients received oral BAL101553 on Days 15-21 of cycle 2 to assess oral bioavailability. Results BAL101553 was well tolerated at doses up to ≤70 mg/m2. Three grade 3 DLTs occurred: hypotension (70 mg/m2), hyponatremia and neutropenia (both 90 mg/m2). The MTD for 48-h IV BAL101553 was 70 mg/m2. At this dose level, the AUC for BAL27862 was 8580 ng.h/mL and the Cmax was 144 ng/mL. No apparent dose-related effects on blood pressure were observed with 48-h BAL101553 IV infusion. BAL27862 oral bioavailability was >80%. Conclusions Continuous 48-h IV BAL101553 infusion achieved higher exposure of the BAL27862 active metabolite than a 2-h infusion at the RP2D and did not cause vascular toxicity. Clinicaltrials.gov registration: NCT02895360.


Assuntos
Antineoplásicos/uso terapêutico , Benzimidazóis/uso terapêutico , Neoplasias/tratamento farmacológico , Oxidiazóis/uso terapêutico , Pró-Fármacos/uso terapêutico , Administração Oral , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Benzimidazóis/efeitos adversos , Benzimidazóis/sangue , Benzimidazóis/farmacocinética , Feminino , Humanos , Infusões Intravenosas , Masculino , Dose Máxima Tolerável , Microtúbulos , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/metabolismo , Oxidiazóis/efeitos adversos , Oxidiazóis/sangue , Oxidiazóis/farmacocinética , Pró-Fármacos/efeitos adversos , Pró-Fármacos/farmacocinética , Resultado do Tratamento
3.
Clin Pharmacol Drug Dev ; 7(8): 860-870, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29870591

RESUMO

Amenamevir (formerly ASP2151) induces cytochrome P450 (CYP)2B6 and CYP3A4 and inhibits CYP2C8.  We conducted 2 studies, 1 using montelukast as a probe to assess CYP2C8 and the other bupropion to assess CYP2B6.  The montelukast study examined the effect of amenamevir on the pharmacokinetics of montelukast in 24 healthy men: each subject received montelukast 10 mg alone, followed by montelukast 10 mg with amenamevir 400 mg, or vice versa after a washout period.  In the bupropion study, 24 subjects received a single dose of 150 mg bupropion on days 1, 15, 22, and 29, and repeated once-daily doses of 400 mg amenamevir on days 6-15.  Amenamevir increased peak concentration and area under the concentration-time curve of montelukast by about 22% (ratio 121.7%, 90%CI [114.8, 129.1]; 121% [116.2, 128.4], respectively) with a similar increase in hydroxymontelukast (ratio 121.4%, 90%CI [106.4, 138.5]; 125.6 % [111.3, 141.7]).  Amenamevir reduced peak concentration and area under the concentration-time curve of bupropion by 16% (84.29%, 90%CI [78.00, 91.10]; 84.07%, 90%CI [78.85, 89.63]), with recovery after 1 week; the pharmacokinetics of the primary metabolite hydroxybupropion was unaffected.  Thus, amenamevir increased plasma concentrations of montelukast and decreased those of bupropion, but it did not do so enough to require dose adjustment of coadministered substrates of either CYP2C8 or CYP2B6.


Assuntos
Acetatos/farmacocinética , Bupropiona/farmacocinética , Citocromo P-450 CYP2B6/metabolismo , Citocromo P-450 CYP2C8/metabolismo , Oxidiazóis/farmacocinética , Quinolinas/farmacocinética , Acetatos/sangue , Adolescente , Adulto , Bupropiona/sangue , Ciclopropanos , Citocromo P-450 CYP2B6/biossíntese , Indutores do Citocromo P-450 CYP2B6/sangue , Indutores do Citocromo P-450 CYP2B6/farmacocinética , Indutores do Citocromo P-450 CYP2B6/farmacologia , Inibidores do Citocromo P-450 CYP2C8/sangue , Inibidores do Citocromo P-450 CYP2C8/farmacocinética , Inibidores do Citocromo P-450 CYP2C8/farmacologia , Interações Medicamentosas , Voluntários Saudáveis , Hepatócitos/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Oxidiazóis/sangue , Oxidiazóis/farmacologia , Quinolinas/sangue , Sulfetos , Adulto Jovem
4.
Bioorg Med Chem Lett ; 26(2): 466-471, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26687487

RESUMO

Poor solubility and cationic amphiphilic drug-likeness were liabilities identified for a lead series of S1P3-sparing, S1P1 agonists originally developed from a high-throughput screening campaign. This work describes the subsequent optimization of these leads by balancing potency, selectivity, solubility and overall molecular charge. Focused SAR studies revealed favorable structural modifications that, when combined, produced compounds with overall balanced profiles. The low brain exposure observed in rat suggests that these compounds would be best suited for the potential treatment of peripheral autoimmune disorders.


Assuntos
Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Tiadiazóis/farmacologia , Animais , Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Células Hep G2 , Humanos , Ligação de Hidrogênio , Cinética , Oxidiazóis/sangue , Oxidiazóis/síntese química , Ratos , Solubilidade , Relação Estrutura-Atividade , Tiadiazóis/sangue , Tiadiazóis/síntese química
5.
Sci Rep ; 5: 11906, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26148672

RESUMO

Fructose-based 3-acetyl-2,3-dihydro-1,3,4-oxadiazole (GLB) is a novel antitumor agent and belongs to glycosylated spiro-heterocyclic oxadiazole scaffold derivative. This research first reported a simple, specific, sensitive and stable high performance liquid chromatography-ultraviolet detector (HPLC-UV) method for the quantitative determination of GLB in plasma. In this method, the chromatographic separation was achieved with a reversed phase C18 column. The calibration curve for GLB was linear at 300 nm. The lower limit of quantification was 10 ng/mL. The precision, accuracy and stability of the method were validated adequately. This method was successfully applied to the pharmacokinetic study in rats for detection of GLB after oral administration. Moreover, the structures of parent compound GLB and its two major metabolites M1 and M2 were identified in plasma using an ultra performance liquid chromatography-electrospray ionization-quadrupole-time of flight- mass spectrometry (UPLC-ESI-QTOF-MS) method. Our results indicated that the di-hydroxylation (M1) and hydroxylation (M2) of GLB are the major metabolites. In conclusion, the present study provided valuable information on an analytical method for the determination of GLB and its metabolites in rats, can be used to support further developing of this antitumor agent.


Assuntos
Antineoplásicos/sangue , Cromatografia Líquida de Alta Pressão , Oxidiazóis/sangue , Administração Oral , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Calibragem , Cromatografia Líquida de Alta Pressão/normas , Meia-Vida , Hidroxilação , Masculino , Oxidiazóis/química , Oxidiazóis/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta
6.
Biol Psychiatry ; 78(7): 452-62, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25802079

RESUMO

BACKGROUND: Based on rodent studies, group II metabotropic glutamate receptors (mGluR2 and mGluR3) were suggested as targets for addiction treatment. However, LY379268 and other group II agonists do not discriminate between the mainly presynaptic inhibitory mGluR2 (the proposed treatment target) and mGluR3. These agonists also produce tolerance over repeated administration and are no longer considered for addiction treatment. Here, we determined the effects of AZD8529, a selective positive allosteric modulator of mGluR2, on abuse-related effects of nicotine in squirrel monkeys and rats. METHODS: We first assessed modulation of mGluR2 function by AZD8529 using functional in vitro assays in membranes prepared from a cell line expressing human mGluR2 and in primate brain slices. We then determined AZD8529 (.03-10 mg/kg, intramuscular injection) effects on intravenous nicotine self-administration and reinstatement of nicotine seeking induced by nicotine priming or nicotine-associated cues. We also determined AZD8529 effects on food self-administration in monkeys and nicotine-induced dopamine release in accumbens shell in rats. RESULTS: AZD8529 potentiated agonist-induced activation of mGluR2 in the membrane-binding assay and in primate cortex, hippocampus, and striatum. In monkeys, AZD8529 decreased nicotine self-administration at doses (.3-3 mg/kg) that did not affect food self-administration. AZD8529 also reduced nicotine priming- and cue-induced reinstatement of nicotine seeking after extinction of the drug-reinforced responding. In rats, AZD8529 decreased nicotine-induced accumbens dopamine release. CONCLUSIONS: These results provide evidence for efficacy of positive allosteric modulators of mGluR2 in nonhuman primate models of nicotine reinforcement and relapse. This drug class should be considered for nicotine addiction treatment.


Assuntos
Comportamento de Procura de Droga/efeitos dos fármacos , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Indóis/farmacologia , Oxidiazóis/farmacologia , Tabagismo/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células CHO , Cricetulus , Modelos Animais de Doenças , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Comportamento de Procura de Droga/fisiologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/sangue , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Células HEK293 , Humanos , Indóis/sangue , Masculino , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Oxidiazóis/sangue , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Saimiri , Autoadministração , Tabagismo/fisiopatologia
7.
Br J Pharmacol ; 172(7): 1739-52, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25409768

RESUMO

BACKGROUND AND PURPOSE: Catechol-O-methyltransferase (COMT) is an important target in the levodopa treatment of Parkinson's disease; however, the inhibitors available have problems, and not all patients benefit from their efficacy. Opicapone was developed to overcome those limitations. In this study, opicapone's pharmacological properties were evaluated as well as its potential cytotoxic effects. EXPERIMENTAL APPROACH: The pharmacodynamic effects of opicapone were explored by evaluating rat COMT activity and levodopa pharmacokinetics, in the periphery through microdialysis and in whole brain. The potential cytotoxicity risk of opicapone was explored in human hepatocytes by assessing cellular ATP content and mitochondrial membrane potential. KEY RESULTS: Opicapone inhibited rat peripheral COMT with ED50 values below 1.4 mg⋅kg(-1) up to 6 h post-administration. The effect was sustained over the first 8 h and by 24 h COMT had not returned to control values. A single administration of opicapone resulted in increased and sustained plasma levodopa levels with a concomitant reduction in 3-O-methyldopa from 2 h up to 24 h post-administration, while tolcapone produced significant effects only at 2 h post-administration. The effects of opicapone on brain catecholamines after levodopa administration were sustained up to 24 h post-administration. Opicapone was also the least potent compound in decreasing both the mitochondrial membrane potential and the ATP content in human primary hepatocytes after a 24 h incubation period. CONCLUSIONS AND IMPLICATIONS: Opicapone has a prolonged inhibitory effect on peripheral COMT, which extends the bioavailability of levodopa, without inducing toxicity. Thus, it exhibits some improved properties compared to the currently available COMT inhibitors.


Assuntos
Inibidores de Catecol O-Metiltransferase/farmacologia , Levodopa/farmacocinética , Oxidiazóis/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Antiparkinsonianos/farmacologia , Benzofenonas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Catecol O-Metiltransferase/metabolismo , Catecóis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Levodopa/sangue , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Modelos Biológicos , Nitrilas/farmacologia , Nitrofenóis/farmacologia , Oxidiazóis/sangue , Oxidiazóis/farmacocinética , Ratos Wistar , Tolcapona
8.
Cancer Med ; 2(5): 611-24, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24403227

RESUMO

The activation of the transcription factor hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor development, tumor progression, and resistance to chemo- and radiotherapy. In order to identify compounds targeting the HIF pathway, a small molecule library was screened using a luciferase-driven HIF-1 reporter cell line under hypoxia. The high-throughput screening led to the identification of a class of aminoalkyl-substituted compounds that inhibited hypoxia-induced HIF-1 target gene expression in human lung cancer cell lines at low nanomolar concentrations. Lead structure BAY 87-2243 was found to inhibit HIF-1α and HIF-2α protein accumulation under hypoxic conditions in non-small cell lung cancer (NSCLC) cell line H460 but had no effect on HIF-1α protein levels induced by the hypoxia mimetics desferrioxamine or cobalt chloride. BAY 87-2243 had no effect on HIF target gene expression levels in RCC4 cells lacking Von Hippel-Lindau (VHL) activity nor did the compound affect the activity of HIF prolyl hydroxylase-2. Antitumor activity of BAY 87-2243, suppression of HIF-1α protein levels, and reduction of HIF-1 target gene expression in vivo were demonstrated in a H460 xenograft model. BAY 87-2243 did not inhibit cell proliferation under standard conditions. However under glucose depletion, a condition favoring mitochondrial ATP generation as energy source, BAY 87-2243 inhibited cell proliferation in the nanomolar range. Further experiments revealed that BAY 87-2243 inhibits mitochondrial complex I activity but has no effect on complex III activity. Interference with mitochondrial function to reduce hypoxia-induced HIF-1 activity in tumors might be an interesting therapeutic approach to overcome chemo- and radiotherapy-resistance of hypoxic tumors.


Assuntos
Complexo I de Transporte de Elétrons/antagonistas & inibidores , Neoplasias Pulmonares/metabolismo , Oxidiazóis/farmacologia , Pirazóis/farmacologia , Animais , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Anidrase Carbônica IX , Anidrases Carbônicas/biossíntese , Anidrases Carbônicas/genética , Hipóxia Celular/genética , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Descoberta de Drogas/métodos , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Neoplásicos , Genes Reporter , Humanos , Fator 1 Induzível por Hipóxia/biossíntese , Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Terapia de Alvo Molecular/métodos , Oxidiazóis/administração & dosagem , Oxidiazóis/sangue , Oxidiazóis/uso terapêutico , Pirazóis/administração & dosagem , Pirazóis/sangue , Pirazóis/uso terapêutico , RNA Interferente Pequeno/genética , Bibliotecas de Moléculas Pequenas , Carga Tumoral/efeitos dos fármacos , Células Tumorais Cultivadas , Proteína Supressora de Tumor Von Hippel-Lindau/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Arch Neurol ; 69(11): 1430-40, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22892585

RESUMO

OBJECTIVE: To assess the safety, tolerability, and pharmacokinetic and pharmacodynamic effects of the -secretase inhibitor avagacestat in patients with mild to moderate Alzheimer disease (AD). DESIGN: Randomized, double-blind, placebo-controlled,24-week phase 2 study. SETTING: Global, multicenter trial. PATIENTS: A total of 209 outpatients with mild to moderate AD were randomized into the double-blind treatment phase. The median age of the patients was 75 years,58.9% were APOE ε4 carriers, and baseline measures of disease severity were similar among groups. INTERVENTION: Avagacestat, 25, 50, 100, or 125 mg daily,or placebo administered orally daily. MAIN OUTCOME MEASURES: Safety and tolerability of avagacestat. RESULTS: Discontinuation rates for the 25-mg and 50-mg doses of avagacestat were comparable with placebo but were higher in the 100-mg and 125-mg dose groups.Trends for worsening cognition, as measured by change from baseline Alzheimer Disease Assessment Scale cognitive subscale score, were observed in the 100-mg and125-mg dose groups. Treatment-emergent serious adverse events were similar across placebo and treatment groups. The most common reason for discontinuation was adverse events, predominantly gastrointestinal anddermatologic. Other adverse events occurring more frequentlyin patients undergoing treatment included reversibleglycosuria (without associated serum glucose changes), nonmelanoma skin cancer, and asymptomaticmagnetic resonance imaging findings. Exploratory cerebrospinal fluid amyloid isoforms and tau biomarker analysis demonstrated dose-dependent but not statistically significant reductions in a small subset of patients. CONCLUSIONS: Avagacestat dosed at 25 and 50 mg daily was relatively well tolerated and had low discontinuation rates. The 100-mg and 125-mg dose arms were poorly tolerated with trends for cognitive worsening. Exploratory cerebrospinal fluid biomarker substudies provide preliminary support for -secretase target engagement,but additional studies are warranted to better characterize pharmacodynamic effects at the 25- and 50-mg doses.This study establishes an acceptable safety and tolerability dose range for future avagacestat studies in AD. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00810147


Assuntos
Doença de Alzheimer/tratamento farmacológico , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/uso terapêutico , Oxidiazóis/sangue , Oxidiazóis/uso terapêutico , Sulfonamidas/sangue , Sulfonamidas/uso terapêutico , Atividades Cotidianas , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Imunoprecipitação , Cooperação Internacional , Imageamento por Ressonância Magnética , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Testes Neuropsicológicos , Avaliação de Resultados em Cuidados de Saúde , Escalas de Graduação Psiquiátrica , Fatores de Tempo , Proteínas tau/líquido cefalorraquidiano
10.
J Vet Med Sci ; 55(2): 281-5, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8513010

RESUMO

This report describes a unique response of the anion transporter in equine erythrocytes to the fluorescent substrate N-(2-aminoethyl sulfonate)-7-nitrobenz-2-oxa-3-diazole (NBD-taurine). Equine erythrocytes showed fluxes of NBD-taurine both inward and outward at rates considerably slower than those in human cells. These fluxes were completely abolished by a typical anion transport inhibitor, 4,4'-diisothiocyanostilbene-2,2'-disulfonate. Furthermore, NBD-taurine competitively inhibited the uptake of phosphate in equine red cells with an inhibition constant of phosphate that was slightly higher than the Michaelis constant of phosphate uptake. These results demonstrate that the anion transporter (band 3) in equine red cells binds NBD-taurine but does not prefer it as the substrate for transport, suggesting a structural difference between equine and human anion transporters at the polypeptide portion which is implicated in anion transport.


Assuntos
Proteínas de Transporte/sangue , Eritrócitos/metabolismo , Cavalos/sangue , Oxidiazóis/farmacologia , Taurina/análogos & derivados , Animais , Transporte Biológico , Proteínas de Transporte/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Corantes Fluorescentes , Humanos , Cinética , Oxidiazóis/sangue , Proteínas de Ligação a Fosfato , Fosfatos/sangue , Especificidade da Espécie , Taurina/sangue , Taurina/farmacologia
11.
J Cell Physiol ; 133(1): 55-63, 1987 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3312243

RESUMO

The intraerythrocytic malarial parasite Plasmodium falciparum induces permeation pathways in the plasma membrane of its host, the red blood cell. The pathways display porelike properties with selectivity toward anions and neutral molecules. They are shown here to be susceptible to chemical modification by 4,4'-diisothiocyano-2,2'-dihydrostilbene disulfonic acid (H2DIDS), an amino-reactive reagent which is impermeant to uninfected cells. At pH 7.4 the reagent affected transport only marginally while freely entering into infected cells and reacting with intracellular hemoglobin. On the other hand, at pH above 8.5, the compound blocked the pathways efficiently (IC 50 approximately equal to 50 microM, at 37 degrees C for 10 min) as judged by four criteria: (1) selective lysis of infected erythrocytes in the presence of isotonic polyols; (2) uptake of [14C] sorbitol into infected cells; (3) uptake of the fluorescent anion Nbd-taurine into infected cells under conditions in which the native anion transport systems was inhibited; and (4) labeling of intracellular hemoglobin by the permeating reagent [3H]H2DIDS. The inhibitory effect was observed only with mature forms of parasitized cells, i.e., from the trophozoite stage and onward, while the pathways of immature ring forms were refractive. However, when the probe was incorporated into the interior of hemoglobin-depleted resealed ghosts prepared from ring forms, it was found to inhibit the pore-mediated transport. On the basis of these and other studies we postulate that the H2DIDS-sensitive sites on the pathways are endofacial, thus requiring penetration of the probe (probably through the same pathway) for their inactivation. Labeling studies with the radiolabeled modifier implicate 120-Kd, 63-Kd, and/or 51-Kd polypeptides as candidates for the pore components.


Assuntos
Membrana Eritrocítica/parasitologia , Malária/sangue , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico , Ácido 4-Acetamido-4'-isotiocianatostilbeno-2,2'-dissulfônico/análogos & derivados , Ácido 4-Acetamido-4'-isotiocianatostilbeno-2,2'-dissulfônico/farmacologia , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Humanos , Oxidiazóis/sangue , Florizina/farmacologia , Plasmodium falciparum , Sorbitol/sangue , Taurina/análogos & derivados , Taurina/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA