Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pharmazie ; 62(1): 60-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17294816

RESUMO

This study was conducted to investigate possible mechanisms underlying the gastroprotective effect of nicorandil on experimentally-induced gastric lesions in rats. The rats were randomly assigned to vehicle (saline or tween 80), nicorandil (2 mg/kg), glibenclamide (2 mg/kg), nicorandil plus glibenclamide- and cimetidine (50 mg/kg)-pretreated groups, in addition to the non-stressed control group, to demonstrate whether the KATP channel opening activity contributed to nicorandil's gastroprotection. Gastric lesions were induced by water immersion-restraint stress (WIRS) and ulcer indices were determined. Gastric juice parameters (pH, free and total acid output, and pepsin and mucin concentrations) were determined for each group. Another group of rats was divided into control, saline-pretreated and nicorandil (2 mg/kg)-pretreated subgroups. The rats were subjected to 5 h of WIRS and the stomachs were used for determination of gastric mucosal levels of lipid peroxides, histamine, prostaglandin E2 (PGE2) and total nitrites. Nicorandil displayed significant protection against gastric lesions formation. Glibenclamide, when administered concomitantly with nicorandil, abolished its protective effects. Nicorandil significantly reduced gastric acid secretion and pepsin concentration, but upon co-administration with glibenclamide, these effects were blocked. Additionally, nicorandil significantly reduced gastric mucosal lipid peroxides and total nitrites back to near normal levels and significantly increased gastric mucosal PGE2, but did not alter significantly histamine levels. The results confirm a gastroprotective effect for nicorandil, the mechanism of which comprises KATP channel opening, free radical scavenging, PGE2 elevation, decrease of proteolytic activity and acid output and prevention of the detrimental increase of nitric oxide during WIRS, probably, by inhibiting iNOS activity.


Assuntos
Antiulcerosos , Nicorandil/farmacologia , Úlcera Gástrica/prevenção & controle , Animais , Dinoprostona/metabolismo , Ácido Gástrico/metabolismo , Suco Gástrico/química , Mucosa Gástrica/patologia , Histamina/metabolismo , Concentração de Íons de Hidrogênio , Imersão , Canais KATP , Ligadura , Peróxidos Lipídicos/metabolismo , Masculino , Mucinas/análise , Nitritos/metabolismo , Pepsina A/análise , Canais de Potássio Corretores do Fluxo de Internalização/agonistas , Piloro/fisiologia , Ratos , Ratos Sprague-Dawley , Restrição Física , Úlcera Gástrica/etiologia , Úlcera Gástrica/patologia , Estresse Psicológico/complicações
2.
Pharmacology ; 79(2): 76-85, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17159379

RESUMO

AIM: This study investigated possible mechanisms underlying the gastroprotective effect of nicorandil on experimentally-induced gastric lesions in rats. METHODS: Rats were randomly assigned to vehicle-, nicorandil (10 mg/kg)-, glibenclamide (6 mg/kg)-, nicorandil + glibenclamide- and cimetidine-pretreated groups, in addition to non-stressed control group, to demonstrate whether the K(ATP )channel opening contributed to nicorandil's gastroprotection. Lesions were induced by water immersion-restraint stress (WIRS) and ulcer indices were determined. Gastric juice parameters (pH, acid output, pepsin and mucin concentrations) were determined. Another set of rats was divided into control, saline-pretreated and nicorandil (10 mg/kg)-pretreated groups. Rats underwent WIRS and their stomachs were used for determination of gastric mucosal lipid peroxides, histamine, PGE(2), and total nitrites levels. RESULTS: Nicorandil displayed significant protection against gastric lesions formation, abolished by concomitant administration of glibenclamide. Nicorandil significantly reduced gastric acid and pepsin secretion, but upon coadministration with glibenclamide, these effects were blocked. Additionally, nicorandil significantly reduced gastric mucosal lipid peroxides and total nitrites, but did not affect PGE(2) and histamine levels. CONCLUSION: Results confirm a gastroprotective effect for nicorandil, the mechanism of which comprises K(ATP) channel opening, free radical scavenging, decrease of pepsin and acid secretion and prevention of the detrimental rise in nitric oxide during WIRS.


Assuntos
Antiulcerosos/farmacologia , Nicorandil/farmacologia , Canais de Potássio/agonistas , Úlcera Gástrica/prevenção & controle , Animais , Antiulcerosos/uso terapêutico , Cimetidina/farmacologia , Cimetidina/uso terapêutico , Quimioterapia Combinada , Suco Gástrico/química , Glibureto/farmacologia , Glibureto/uso terapêutico , Concentração de Íons de Hidrogênio , Imersão , Masculino , Nicorandil/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Úlcera Gástrica/etiologia , Estresse Psicológico/complicações
3.
Cancer Chemother Pharmacol ; 46(3): 235-40, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11021741

RESUMO

PURPOSE: The purpose of this investigation was to study the effects of combining oral 5-(phenylselenenyl)acyclouridine (PSAU) with 2',3',5'-tri-O-acetyluridine (TAU) on the levels of plasma uridine in mice. PSAU is a new lipophilic and potent inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. PSAU has 100% oral bioavailability and is a powerful enhancer of the bioavailability of oral uridine. TAU is a prodrug of uridine and a far superior source of uridine than uridine itself. METHODS: Oral TAU was administered to mice alone or with PSAU. The plasma levels of uridine and its catabolites as well as PSAU were measured using HPLC and pharmacokinetic analysis was performed. RESULTS: Oral administration of 2000 mg/kg TAU increased plasma uridine by over 250-fold with an area under the curve (AUC) of 754 micromol x h/l. Coadministration of PSAU at 30 and 120 mg/kg with TAU further improved the bioavailability of plasma uridine resulting from the administration of TAU alone by 1.7- and 3.9-fold, respectively, and reduced the Cmax and AUC of plasma uracil. CONCLUSION: The exceptional effectiveness of PSAU plus TAU in elevating and sustaining a high plasma uridine concentration could be useful in the management of medical disorders that are remedied by administration of uridine, as well as the rescue or protection from host toxicities of various chemotherapeutic pyrimidine analogues.


Assuntos
Inibidores Enzimáticos/farmacologia , Compostos Organosselênicos/farmacologia , Pró-Fármacos/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia , Uridina/sangue , Uridina/farmacologia , Acetatos , Animais , Disponibilidade Biológica , Sinergismo Farmacológico , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Feminino , Camundongos , Compostos Organosselênicos/sangue , Compostos Organosselênicos/farmacocinética , Pró-Fármacos/farmacocinética , Uracila/sangue , Uracila/farmacocinética , Uridina/análogos & derivados , Uridina/farmacocinética , Uridina Fosforilase/antagonistas & inibidores
4.
Biochem Pharmacol ; 60(5): 687-92, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10927027

RESUMO

The effect of co-administration of 5-(phenylselenenyl)acyclouridine (PSAU), a new uridine phosphorylase (UrdPase, EC 2.4.2.3) inhibitor, on the efficacy of 5-fluoro-2'-deoxyuridine (FdUrd) was tested against murine colon C26-10 tumor xenografts. In contrast to our previous results with human tumors, co-administration of PSAU with FdUrd decreased instead of increasing the efficacy of FdUrd against tumor growth. However, co-administration of PSAU with FdUrd (300 mg/kg/day) protected the mice completely from the 83% mortality induced by the same dose of FdUrd alone. Enzyme studies indicated that UrdPase in colon C26-10 tumors is responsible for the catabolism of FdUrd to 5-fluorouracil (FUra), as colon C26-10 tumors do not have thymidine phosphorylase (dThdPase, EC 2.4.2.4). In contrast, colon C26-10 tumors had extraordinarily high UrdPase activity (300 micromol/min/mg protein), which was at least 200-fold higher than the highest UrdPase activity in any of the human xenografts we tested previously. Furthermore, the activities of UrdPase and orotate phosphoribosyltransferase (OPRTase, EC 2.4.2.10) were 192- and 2-fold higher, respectively, while that of dihydrouracil dehydrogenase (EC 1.3.1.2) was 1000-fold lower in the tumor than in the host liver. It is suggested that FdUrd exerts its anticancer effects against colon C26-10 tumors mainly through the catabolism of FdUrd to FUra by UrdPase, which then could be anabolized to 5-fluorouridine 5'-monophosphate (FUMP) by OPRTase and ultimately to other toxic 5-fluorouridine nucleotides, hence inducing the observed FdUrd toxic effects. Co-administration of PSAU with FdUrd inhibited UrdPase and the catabolism of FdUrd to FUra. This would result in the observed reduction of the antitumor efficacy of FdUrd. In addition, the increase in plasma uridine concentration induced by PSAU as well as the catabolism of FUra by the high dihydrouracil dehydrogenase activity in the liver also may have circumvented any residual FUra toxic effects against the host. These results clearly demonstrate that the anticancer efficacy of the combination of UrdPase inhibitors and FdUrd is not general and is dependent largely on the type of tumor under treatment and the mode of FdUrd metabolism in these tumors.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Floxuridina/farmacologia , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Interações Medicamentosas , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Transplante Heterólogo , Células Tumorais Cultivadas
5.
Biochem Pharmacol ; 60(3): 427-31, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10856438

RESUMO

Administration of 200 mg/kg of 5-fluorouracil (FUra) to mice bearing human colon carcinoma DLD-1 xenografts resulted in 100% mortality. Oral administration of 2000 mg/kg of 2',3',5'-tri-O-acetyluridine (TAU), a prodrug of uridine, in combination with 120 mg/kg of 5-(benzyloxybenzyl)barbituric acid acyclonucleoside (BBBA), the most potent known inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2. 3), 2 hr after the administration of the same dose of FUra completely protected the mice (100% survival) from the toxicity of FUra. This combination also reduced tumor weight by 67% compared with 46% achieved by the maximum tolerated dose (50 mg/kg) of FUra alone. Similarly, administration of BBBA plus TAU 1 hr before or 4 hr after the administration of FUra reduced the tumor weight by 53 and 37%, respectively. However, these schedules were less effective in protecting the host from the toxicity of FUra than when the treatment was carried out at 2 hr after FUra administration. TAU alone did not protect from FUra host toxicity. The efficiency of the BBBA plus TAU combination in rescuing from FUra host toxicities is attributed to the exceptional effectiveness of this combination in raising and maintaining higher plasma uridine concentrations than those achieved by TAU alone or by equimolar doses of uridine (Ashour et al., Biochem Pharmacol 51: 1601-1612, 1996). The present results suggest that the BBBA plus TAU combination can provide a better substitute for the massive doses of uridine required to achieve the high levels of uridine necessary to rescue or protect from FUra host toxicities without the toxic side-effects associated with such doses of uridine. The combination of TAU plus BBBA may also allow the escalation of FUra doses for better chemotherapeutic efficacy. Alternatively, the combination may be used as a rescue regimen in the occasional cases where cancer patients receive a lethal overdose of FUra.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Barbitúricos/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoruracila/toxicidade , Uridina Fosforilase/antagonistas & inibidores , Uridina/análogos & derivados , Acetatos , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Barbitúricos/uso terapêutico , Interações Medicamentosas , Feminino , Fluoruracila/uso terapêutico , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Pró-Fármacos/farmacologia , Células Tumorais Cultivadas , Uridina/farmacologia
6.
Cancer Chemother Pharmacol ; 45(5): 351-61, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10803917

RESUMO

PURPOSE: The purpose of this investigation was to evaluate the efficacy of oral 5-(phenylselenenyl)-acyclouridine (PSAU) in increasing endogenous plasma uridine concentration as well as its ability to improve the bioavailability of oral uridine. PSAU is a new potent and specific inhibitor of uridine phosphorylase (Urd-Pase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. This compound was designed as a lipophilic inhibitor in order to facilitate its access to the liver and intestine, the main organs involved in uridine catabolism. METHODS: Oral PSAU was administered orally to mice alone or with uridine. The plasma levels of PSAU as well as uridine and its catabolites were measured using high-performance liquid chromatography and pharmacokinetic analysis was performed. RESULTS: PSAU has an oral bioavailability of 100% and no PSAU metabolites were detected. PSAU has no apparent toxicity at high doses. Oral administration of PSAU at 30 and 120 mg/kg increased baseline concentration of endogenous plasma uridine (2.6 +/- 0.7 microM) by 3.2- and 8.7-fold, respectively, and remained three- and six-fold higher, respectively, than the controls for over 8 h. PSAU, however, did not alter the concentration of endogenous plasma uracil. Co-administration of PSAU with uridine elevated the concentration of plasma uridine over that resulting from the administration of either alone, and reduced the peak plasma concentration (C(max)) and area under the curve (AUC) of plasma uracil. Co-administration of PSAU at 30 mg/kg and 120 mg/kg improved the low bioavailability of oral uridine (7.7%) administered at 1,320 mg/kg by 4.8- and 4.2-fold, respectively, and reduced the AUC of plasma uracil from 1,421 to 787 micromol/h x l and 273 micromol/h x l, respectively. Similar results were observed when PSAU was co-administered with lower doses of uridine. Oral PSAU at 30 mg/kg and 120 mg/kg improved the bioavailability of oral 330 mg/kg uridine by 5.2- and 8.9-fold, and that of oral 660 mg/kg uridine by 6.4- and 9.0-fold, respectively. However, the reduction in the AUC values of plasma uracil was less dramatic than that seen when the high dose of 1,320 mg/kg uridine was used. CONCLUSION: The effectiveness of the PSAU plus uridine combination in elevating and sustaining high plasma uridine concentration may be useful to rescue or protect from host toxicity of various chemotherapeutic pyrimidine analogs as well as in the management of medical disorders that are remedied by administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Uridina Fosforilase/antagonistas & inibidores , Uridina/sangue , Animais , Disponibilidade Biológica , Feminino , Camundongos , Compostos Organosselênicos/farmacocinética , Uracila/farmacocinética , Uracila/farmacologia
7.
Biochem Pharmacol ; 51(12): 1601-11, 1996 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-8687475

RESUMO

5-(m-Benzyloxybenzyl)barbituric acid acyclonucleoside (BBBA), the most potent inhibitor known of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism, and 2',3',5'-tri-O-acetyluridine (TAU), a prodrug of uridine, were used to investigate the possibility of improving the bioavailability of oral uridine in mice. Oral BBBA administered at 30, 60, 120, and 240 mg/kg increased the concentration of plasma uridine (2.6 +/- 0.7 microM) by 3.2-, 4.6-, 5.4-, and 7.2-fold, respectively. After administration of 120 and 240 mg/kg BBBA, plasma uridine concentration remained 3- and 6-fold, respectively, higher than the plasma concentration at zero time (C0) for over 8 hr. On the other hand, BBBA did not change the concentration of plasma uracil. TAU was far more superior than uridine in improving the bioavailability of plasma uridine. The relative bioavailability of plasma uridine released from oral TAU (53%) was 7-fold higher than that (7.7%) obtained by oral uridine. Oral TAU at 460, 1000, and 2000 mg/kg achieved area under the curve (AUC) values of plasma uridine of 82, 288, and 754 mumol.hr/L, respectively. Coadministration of BBBA with uridine or TAU further improved the bioavailability of plasma uridine resulting from the administration of either alone and reduced the Cmax and AUC of plasma uracil. Coadministration of BBBA at 30, 60, and 120 mg/kg improved the relative bioavailability of uridine released from 2000 mg/kg TAU (53%) by 1.7-, 2.7-, and 3.9-fold, respectively, while coadministration of the same doses of BBBA with an equimolar dose of uridine (1320 mg/kg) increased the relative bioavailability of oral uridine (7.7%) by 4.1-, 5.3-, and 7.8-fold, respectively. Moreover, the AUC and Cmax of plasma uridine after BBBA (120 mg/kg) coadministration with TAU were 3.5- and 11.5-fold, respectively, higher than those obtained from coadministration of BBBA with an equimolar dose of uridine. The exceptional effectiveness of the BBBA plus TAU combination in elevating and sustaining high plasma uridine concentration can be useful in the management of medical disorders that are remedied by administration of uridine as well as to rescue or protect from host-toxicities of various chemotherapeutic pyrimidine analogues.


Assuntos
Barbitúricos/metabolismo , Barbitúricos/farmacologia , Pró-Fármacos/farmacologia , Uridina/urina , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos , Fatores de Tempo , Uridina/farmacocinética
8.
Cancer Res ; 55(5): 1092-8, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7866994

RESUMO

5-(Benzyloxybenzyl)barbituric acid acyclonucleoside (BBBA) was recently synthesized as a potent and specific inhibitor of uridine phosphorylase (EC 2.4.2.3), the enzyme responsible for the catabolism of 5-fluoro-2'-deoxyuridine (FdUrd) in many types of tumors that are deficient or have little thymidine phosphorylase (EC 2.4.2.4) activity. The effect of BBBA on modulating the antitumor efficacy of FdUrd was evaluated in vitro, against the human colon carcinomas DLD-1 and HCT-15 grown in culture, and in vivo, against DLD-1 grown as xenografts in anti-thymocyte serum immunosuppressed mice. The concentrations of FdUrd that produced 50% growth inhibition after a 3-h exposure were 88 and 340 nM for HCT-15 and DLD-1, respectively. BBBA alone, at all concentrations tested, had no significant effect on the growth of DLD-1 and HCT-15 in culture. However, BBBA at 5, 10, 20, and 40 nM potentiated (P < 0.05) the cytotoxicity of FdUrd (340 nM; 3 h) against DLD-1 in culture by 20, 33, 55, and 63%, respectively. Similarly, BBBA at 10 and 20 nM potentiated the cytotoxicity of FdUrd (88 nM; 3 h) against HCT-15 in culture by 37 and 45%, respectively. In soft agar, BBBA (10 nM) also enhanced the cytocidal effect of FdUrd (10 and 32 nM) against DLD-1 by 41 and 55%, respectively, and against HCT-15 by 6 and 31%, respectively. Increasing BBBA dose to 20 nM enhanced further the FdUrd (10 and 32 nM) cytotoxicity against DLD-1 by 76 and 77%, respectively, and HCT-15 by 31 and 48%, respectively. BBBA also potentiated the chemotherapeutic efficacy of FdUrd in anti-thymocyte serum immunosuppressed mice bearing DLD-1 xenografts with no apparent host toxicity. At a low tumor burden (2.5 x 10(6) cells/mouse), 2 days treatment with FdUrd alone (50 mg/kg/day x 2) did not result in significant reduction in tumor volume. Coadministration of BBBA at 5 and 10 mg/kg/day x 2 did not potentiate the efficacy of FdUrd over that achieved by FdUrd alone, but it significantly reduced the tumor volume by 27 and 32%, respectively, when compared with untreated controls. FdUrd alone at 150 mg/kg/day x 2 reduced the tumor volume by 29%. This reduction in tumor volume was enhanced 1.8-fold by coadministration of BBBA (10 mg/kg/day x 2). At a higher tumor burden (5 x 10(6) cells/mouse) and 4 days treatment, BBBA at 10 and 30 mg/kg/day x 4 reduced further the tumor volume produced by FdUrd alone (200 mg/kg/day x 4) by 1.2- and 1.4-fold, respectively.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Barbitúricos/farmacologia , Floxuridina/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Sinergismo Farmacológico , Feminino , Floxuridina/metabolismo , Humanos , Terapia de Imunossupressão/métodos , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...