Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chem Biol Interact ; 239: 146-52, 2015 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-26163455

RESUMO

The antioxidant flavonoid 7-mono-O-(ß-hydroxyethyl)-rutoside (monoHER) effectively protects against doxorubicin-induced cardiotoxicity in mice. Doxorubicin is a very effective anticancer drug. The clinical use of doxorubicin is limited by severe cardiotoxicity. Free radicals, i.e., hydroxyl and superoxide radicals play a crucial role in this toxicity. In this study the involvement of the major metabolite of monoHER, 4'-O-methylmonoHER (methylmonoHER) in the protective effect of monoHER is studied. MethylmonoHER displayed antioxidant activity i.e., TEAC, hydroxyl and superoxide radical scavenging activity; nevertheless monoHER appeared to be superior compared to methylmonoHER. As a result of scavenging, flavonoids are oxidized and display reactivity towards thiols. Oxidized methylmonoHER, is far less thiol reactive towards creatine kinase than monoHER, which indicates that methylmonoHER is less toxic towards thiol containing enzymes. The thiol-reactivity of oxidized methylmonoHER was also negligible towards KEAP1 compared to monoHER. These results indicate that methylmonoHER hardly protects against radical damage via scavenging or via activating the NRF2 defense system. Also in HUVECs, methylmonoHER provided far less protection against oxidative stress (EC50>100µM) than monoHER which was a very potent protector (EC50=80nM). The results indicate that the contribution of methylmonoHER to the protection against doxorubicin-induced cardiotoxicity by monoHER is relatively low.


Assuntos
Antioxidantes/farmacologia , Hidroxietilrutosídeo/análogos & derivados , Estresse Oxidativo/efeitos dos fármacos , Rutina/análogos & derivados , Antioxidantes/metabolismo , Creatina Quinase/metabolismo , Doxorrubicina/efeitos adversos , Sequestradores de Radicais Livres/farmacologia , Glutationa/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Hidroxietilrutosídeo/metabolismo , Hidroxietilrutosídeo/farmacologia , Radical Hidroxila/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Rutina/química , Rutina/farmacologia , Superóxidos/metabolismo
2.
Biochem Biophys Res Commun ; 456(1): 179-82, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25462563

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease. An evidence-based pharmacological treatment for NAFLD is still lacking, but flavonoids have shown therapeutic potential. The present study was designed to investigate the effect of the flavonoid monoHER on the onset of NAFLD in Ldlr(-/-) mice on a high-fat and high-cholesterol diet. The focus was put on the effect on oxidative stress as well as the adaptive response. Wild type mice served as a control and the effect of monoHER was compared to that of a placebo. In the Ldlr(-/-) group, monoHER provided only a mild protection against oxidative stress. In the placebo Ldlr(-/-) group an adaptive response elicited by the NRF2 antioxidant defense system was observed, evidenced by a higher HO-1 and Gpx3 gene expression, as well as an increased redox status, evidenced by the higher GSH/GSSG ratio. In the monoHER treated Ldlr(-/-) group both the adaptive response as well as the increase in redox status tended to be higher, although this did not reach significance on a group level. Unexpectedly, a strong within animal relationship was found that links a high adaptive response to a low redox status in the monoHER Ldlr(-/-) group. This correlation was absent in the placebo and wild type group. The concept that emerges is that a thiol-reactive oxidation product of monoHER, formed during oxidative stress, selectively induces the NRF2 pathway and enforces the endogenous antioxidant shield, to provide protection against NAFLD.


Assuntos
Flavonoides/farmacologia , Hidroxietilrutosídeo/análogos & derivados , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Receptores de LDL/genética , Animais , Antioxidantes/farmacologia , Feminino , Regulação da Expressão Gênica , Glutationa Peroxidase/metabolismo , Heme Oxigenase-1/metabolismo , Hidroxietilrutosídeo/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Oxirredução , Espécies Reativas de Oxigênio
3.
Int J Mol Sci ; 15(5): 7475-84, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24786288

RESUMO

Antioxidants act as intermediates by picking up the high unselective reactivity of radicals and transferring it to other molecules. In this process the reactivity is reduced and becomes selective. This channeling of the reactivity can cause selective toxicity. The antioxidant quercetin is known to channel the reactivity towards thiol groups. The present study compares the thiol reactivity of quercetin with that of 4'O-methylquercetin (tamarixetin) towards creatine kinase (CK), a vital protein that contains a critical thiol moiety. Our results showed that oxidized quercetin and oxidized tamarixetin both adduct CK, which then loses its enzymatic function. Ascorbate, an important representative of the antioxidant network, is able to prevent adduction to and thus the inhibition of the enzyme by tamarixetin but not by quercetin. Apparently, tamarixetin is less thiol toxic than quercetin, because--rather than adduction to CK--tamarixetin quinone prefers to pass reactivity to the antioxidant network, i.e., to ascorbate. The findings exemplify that radical scavenging flavonoids pick up the reactivity of radicals and act as a pivot in directing the way the reactivity is channeled. A mere minor structural difference of only one methyl moiety between quercetin and tamarixetin appears to have a high impact on the selective, thiol toxicity.


Assuntos
Antioxidantes/toxicidade , Creatina Quinase/antagonistas & inibidores , Creatina Quinase/metabolismo , Dissacarídeos/toxicidade , Quercetina/análogos & derivados , Quercetina/toxicidade , Compostos de Sulfidrila/metabolismo , Antioxidantes/química , Dissacarídeos/química , Modelos Moleculares , Oxirredução , Quercetina/química
4.
Toxicol In Vitro ; 28(4): 538-43, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24412621

RESUMO

The flavonoid 7-mono-O-(ß-hydroxyethyl)-rutoside (monoHER) is an effective protector against doxorubicin induced toxicity which has been related to the antioxidant activity of monoHER. The present study examines the potential relevance of the direct scavenging activity of the flavonoid. The potency of the direct antioxidant effect was confirmed by its instantaneous protection against intracellular oxidative stress in human umbilical vein endothelial cells at therapeutically achievable concentrations (EC50=60 nM) underpinning the involvement of a direct scavenging activity. This direct effect of monoHER is substantiated by (i) its site specific scavenging effect, i.e. on a molecular level monoHER is positioned at the location of radical formation, (ii) its position in the antioxidant network, i.e. on a biochemical level oxidized monoHER quickly reacts with ascorbate or glutathione, (iii) its location in the vascular system, i.e. on a cellular level monoHER is localized in the endothelial and smooth muscle cells in the vascular wall. It is concluded that the flavonoid monoHER can display a physiologically important direct antioxidant effect.


Assuntos
Antioxidantes/farmacologia , Células Endoteliais/efeitos dos fármacos , Rutina/análogos & derivados , Rutina/farmacologia , Animais , Artérias Carótidas/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Humanos , Hidroxietilrutosídeo/análogos & derivados , Hidroxietilrutosídeo/farmacologia , Radical Hidroxila , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo
5.
Free Radic Biol Med ; 51(11): 2118-23, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21982895

RESUMO

During the scavenging of free radicals flavonoids are oxidized to electrophilic quinones. Glutathione (GSH) can trap these quinones, thereby forming GSH-flavonoid adducts. The aim of this study was to compare the stability of the GSH-flavonoid adduct of 7-mono-O-(ß-hydroxyethyl)rutoside (monoHER) with that of quercetin. It was found that GSH-quercetin reacts with the thiol N-acetyl-L-cysteine (NAC) to form NAC-quercetin, whereas GSH-monoHER does not react with NAC. In addition, the adduct of the monoHER quinone with the dithiol dithiothreitol (DTT) is relatively stable, whereas the DTT-quercetin adduct is readily converted into quercetin and DTT disulfide. These differences in reactivity of the thiol-flavonoid adducts demonstrate that GSH-monoHER is much more stable than GSH-quercetin. This difference in reactivity was corroborated by molecular quantum chemical calculations. Thus, although both flavonoid quinones are rapidly scavenged by GSH, the advantage of monoHER is that it forms a stable conjugate with GSH, thereby preventing a possible spread of toxicity. These findings demonstrate that even structurally comparable flavonoids behave differently, which will be reflected in the biological effects of these flavonoids.


Assuntos
Glutationa/química , Hidroxietilrutosídeo/análogos & derivados , Quercetina/química , Hidroxietilrutosídeo/química , Estrutura Molecular , Compostos de Sulfidrila/química
6.
Drug Metab Dispos ; 39(5): 750-6, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21266594

RESUMO

The clinical use of the anticancer drug doxorubicin is limited by severe cardiotoxicity. In mice, the semisynthetic antioxidant flavonoid 7-mono-O-(ß-hydroxyethyl)-rutoside (monoHER) has been successfully used as a protector against doxorubicin-induced cardiotoxicity. However, most monoHER has already been cleared from the body at the time that doxorubicin concentrations are still high. This result suggests that not only the parent compound monoHER itself but also monoHER metabolites could be responsible for the observed cardioprotective effects in mice. Therefore, in the present study, we investigated the metabolism of monoHER in mice. Mice were administered 500 mg/kg monoHER intraperitoneally. At different time points after monoHER administration, bile was collected and analyzed for the presence of monoHER metabolites. The formed metabolites were identified by liquid chromatography-diode array detection-time of flight-mass spectrometry. Thirteen different metabolites were identified. The observed routes of monoHER metabolism are methylation, glucuronidation, oxidation of its hydroxyethyl group, GSH conjugation, and hydrolysis of its disaccharide. In line with other flavonoids, methylated monoHER and the monoHER glucosides are expected to have relatively high cellular uptake and low clearance from the body. Therefore, these metabolites might contribute to the observed protection of monoHER against doxorubicin-induced cardiotoxicity.


Assuntos
Antioxidantes/química , Antioxidantes/metabolismo , Bile/metabolismo , Cardiotônicos/metabolismo , Hidroxietilrutosídeo/análogos & derivados , Animais , Antioxidantes/análise , Antioxidantes/farmacologia , Cardiotônicos/química , Cardiotônicos/farmacologia , Doxorrubicina/toxicidade , Glucuronídeos/química , Glucuronídeos/metabolismo , Glutationa/química , Glutationa/metabolismo , Hidroxietilrutosídeo/química , Hidroxietilrutosídeo/metabolismo , Hidroxietilrutosídeo/farmacologia , Masculino , Metilação , Camundongos , Camundongos Endogâmicos BALB C
7.
PLoS One ; 5(11): e13880, 2010 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-21079733

RESUMO

Antioxidants can scavenge highly reactive radicals. As a result the antioxidants are converted into oxidation products that might cause damage to vital cellular components. To prevent this damage, the human body possesses an intricate network of antioxidants that pass over the reactivity from one antioxidant to another in a controlled way. The aim of the present study was to investigate how the semi-synthetic flavonoid 7-mono-O-(ß-hydroxyethyl)-rutoside (monoHER), a potential protective agent against doxorubicin-induced cardiotoxicity, fits into this antioxidant network. This position was compared with that of the well-known flavonoid quercetin. The present study shows that the oxidation products of both monoHER and quercetin are reactive towards thiol groups of both GSH and proteins. However, in human blood plasma, oxidized quercetin easily reacts with protein thiols, whereas oxidized monoHER does not react with plasma protein thiols. Our results indicate that this can be explained by the presence of ascorbate in plasma; ascorbate is able to reduce oxidized monoHER to the parent compound monoHER before oxidized monoHER can react with thiols. This is a major difference with oxidized quercetin that preferentially reacts with thiols rather than ascorbate. The difference in selectivity between monoHER and quercetin originates from an intrinsic difference in the chemical nature of their oxidation products, which was corroborated by molecular quantum chemical calculations. These findings point towards an essential difference between structurally closely related flavonoids in their interplay with the endogenous antioxidant network. The advantage of monoHER is that it can safely channel the reactivity of radicals into the antioxidant network where the reactivity is completely neutralized.


Assuntos
Antioxidantes/metabolismo , Flavonoides/metabolismo , Hidroxietilrutosídeo/análogos & derivados , Quercetina/metabolismo , Antioxidantes/química , Antioxidantes/farmacologia , Ácido Ascórbico/química , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Flavonoides/química , Flavonoides/farmacologia , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Hidroxietilrutosídeo/química , Hidroxietilrutosídeo/metabolismo , Hidroxietilrutosídeo/farmacologia , Modelos Químicos , Estrutura Molecular , Oxidantes/química , Oxidantes/metabolismo , Oxirredução/efeitos dos fármacos , Quercetina/química , Quercetina/farmacologia
8.
Free Radic Biol Med ; 46(12): 1567-73, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19272444

RESUMO

Flavonoids protect against oxidative stress by scavenging free radicals. During this protection flavonoids are oxidized. The oxidized flavonoids formed are often reactive. Consequently, protection by flavonoids can result in the formation of toxic products. In this study the oxidation of 7-mono-O-(beta-hydroxyethyl)rutoside (monoHER), which is a constituent of the registered drug Venoruton, was studied in the absence and presence of glutathione (GSH). MonoHER was oxidized by horseradish peroxidase/H(2)O(2). Spectrophotometric and HPLC analysis showed that in the presence of GSH, a monoHER-GSH conjugate was formed, which was identified as 2'-glutathionyl monohydroxyethylrutoside by mass spectrometric analysis and (1)H NMR. Preferential formation of this glutathione adduct in the B ring at C2' was confirmed by molecular quantum chemical calculations. This conjugate was also detected in the bile fluid of a healthy volunteer after iv administration of monoHER, demonstrating its formation in vivo. These results indicate that in the process of offering protection against free radicals, monoHER is converted into an oxidation product that is reactive toward thiols. The formation of this thiol-reactive oxidation product is potentially harmful. Thus, the supposed beneficial effect of monoHER as an antioxidant may be accompanied by the formation of products with an electrophilic, toxic potential.


Assuntos
Glutationa/química , Hidroxietilrutosídeo/análogos & derivados , Adulto , Bile/química , Líquidos Corporais/química , Simulação por Computador , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Peróxido de Hidrogênio/química , Hidroxietilrutosídeo/administração & dosagem , Hidroxietilrutosídeo/análise , Hidroxietilrutosídeo/química , Infusões Intravenosas , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Modelos Químicos , Oxirredução , Teoria Quântica , Valores de Referência , Extração em Fase Sólida , Espectrofotometria Ultravioleta , Fatores de Tempo
9.
Cardiovasc Toxicol ; 7(2): 154-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17652822

RESUMO

Cardiotoxic side-effects of doxorubicin limit the clinical use of this anti-cancer agent. Iron chelators have been studied as protectors against doxorubicin-induced cardiotoxicity. These iron chelators do not provide optimal protection and have certain drawbacks. We therefore looked for new protectors and decided that these new compounds should combine iron chelating and antioxidant activity. Flavonoids appeared to possess those combined iron chelating and antioxidant properties. Quantum chemical evaluation of radical stabilization and determination of physico-chemical properties of a series of flavonoids brought our attention to the semi-synthetic flavonoid 7-monohydroxyetylrutoside (monoHER). Both in vitro (using an electrically paced mouse left atrium model) and in vivo (using a mouse ECG telemetry model) experiments corroborated the protective effect of monoHER. MonoHER also showed anti-inflammatory properties. A subsequent clinical phase I study showed that an i.v. dose of 1,500mg/m2 is a feasible and safe dose to be evaluated in a phase II study to investigate the protective properties of monoHER against doxorubicin-induced cardiotoxicity in cancer patients.


Assuntos
Antraciclinas/antagonistas & inibidores , Antibióticos Antineoplásicos/antagonistas & inibidores , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Cardiopatias/induzido quimicamente , Cardiopatias/prevenção & controle , Animais , Antraciclinas/efeitos adversos , Antraciclinas/toxicidade , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/toxicidade , Cardiotônicos/farmacologia , Ensaios Clínicos como Assunto , Ensaios Clínicos Fase I como Assunto , Doxorrubicina/efeitos adversos , Doxorrubicina/toxicidade , Eletrocardiografia/efeitos dos fármacos , Humanos , Ferro/fisiologia , Camundongos , Superóxidos/metabolismo
10.
Biochim Biophys Acta ; 1772(9): 1065-74, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17572073

RESUMO

Anthracycline antibiotics (e.g. doxorubicin and daunorubicin) are among the most effective and widely used anticancer drugs. Unfortunately, their clinical use is limited by the dose-dependent cardiotoxicity. Flavonoids represent a potentially attractive class of compounds to mitigate the anthracycline cardiotoxicity due to their iron-chelating, antioxidant and carbonyl reductase-inhibitory effects. The relative contribution of various characteristics of the flavonoids to their cardioprotective activity is, however, not known. A series of ten flavonoids including quercetin, quercitrin, 7-monohydroxyethylrutoside (monoHER) and seven original synthetic compounds were employed to examine the relationships between their inhibitory effects on carbonyl reduction, iron-chelation and antioxidant properties with respect to their protective potential against doxorubicin-induced cardiotoxicity. Cardioprotection was investigated in the neonatal rat ventricular cardiomyocytes whereas the H9c2 cardiomyoblast cells were used for cytotoxicity testing. Iron chelation was examined via the calcein assay and antioxidant effects and site-specific scavenging were quantified by means of inhibition of lipid peroxidation and hydroxyl radical scavenging activity, respectively. Inhibition of carbonyl reductases was assessed in cytosol from human liver. None of the flavonoids tested had better cardioprotective action than the reference cardioprotector, monoHER. However, a newly synthesized quaternary ammonium analog with comparable cardioprotective effects has been identified. No direct correlation between the iron-chelating and/or antioxidant effect and cardioprotective potential has been found. A major role of carbonyl reductase inhibition seems unlikely, as the best two cardioprotectors of the series are only weak reductase inhibitors.


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Antioxidantes/farmacologia , Cardiotônicos/farmacologia , Doxorrubicina/efeitos adversos , Flavonoides/farmacologia , Quelantes de Ferro/farmacologia , Adolescente , Adulto , Animais , Animais Recém-Nascidos , Antibióticos Antineoplásicos/efeitos adversos , Células Cultivadas , Feminino , Coração/efeitos dos fármacos , Humanos , Ferro/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Ratos , Ratos Wistar
11.
Front Biosci ; 12: 2013-9, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17127439

RESUMO

Alpha-tocopheryl succinate (TOS) is a well-known potent and selective apoptotic agent. This apoptotic activity has been ascribed to its detergent-like property which is also shared by the structurally related compound, alpha-tocopheryl phosphate (TOP). TOP meets the structural requirements that have been described for the apoptotic activity of TO esters, i.e. the combination of three structural, one functional, one signalling and one hydrophobic domain. In this study, we have investigated the effect of TOP on the osteosarcoma cell line MG-63 using TOS as a reference compound. As compared with TOS, TOP showed a higher proliferative and apoptosis inducing activity on the MG-63 cancer cell line. The cytotoxic effect of TOP and TOS seems to be due to the effect of the intact compounds, since only a minor conversion into alpha-tocopheryl (TO) could be detected. EPR experiments showed that TOS and TOP reduced membrane fluidity, whereas TO had no effect. In addition, induction of erythrocyte hemolysis by TOP depended on the pH. These results suggest that the detergent-like activity of these compounds might be involved in their biological effect. Due to the potent biological activities, TOP might be clinically useful.


Assuntos
Apoptose , Inibidores do Crescimento/toxicidade , alfa-Tocoferol/análogos & derivados , Linhagem Celular Tumoral , Inibidores do Crescimento/química , Hemólise/efeitos dos fármacos , Humanos , Fluidez de Membrana/efeitos dos fármacos , Tocoferóis , Vitamina E/análogos & derivados , Vitamina E/química , Vitamina E/toxicidade , alfa-Tocoferol/química , alfa-Tocoferol/toxicidade
12.
Cancer Chemother Pharmacol ; 60(4): 509-14, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17177067

RESUMO

Doxorubicin (DOX) is a potent antitumor agent for different types of cancer, but the cumulative, dose-related cardiotoxicity limits its clinical use. The incidence of abnormal cardiac function after treatment with DOX appears to increase with time. Therefore, late cardiotoxicity is-especially in young surviving patients-a major concern. The aim of this study was to evaluate in mice whether the semisynthetic flavonoid 7-monohydroxyethylrutoside (monoHER) also protected against DOX-induced cardiotoxicity after a long period of follow-up. Four groups of 6 Balb/c mice were treated weekly during 6 weeks with saline, DOX alone (4 mg/kg i.v.), DOX preceded by monoHER (500 mg/kg i.p.), or DOX preceded by monoHER followed by long-term weekly monoHER injections during the observation period of 6 months. Half of the mice treated with DOX only developed DOX-induced heart failure and died within 6 months of observation. Two mice co-treated with monoHER showed weight loss and shortness of breath, whereas one mouse was found dead in its cage known with weight loss. The group receiving DOX plus long-term repeated doses of monoHER started to lose weight. Five out of six mice in this group developed shortness of breath and died before the end of the study with symptoms of cardiac failure induced by DOX. Statistical comparison of the histological heart damage between the different experimental groups was not possible, because the animals died at different time-points in the observation period and DOX-induced cardiotoxicity progressed with time. Nevertheless, it was clear that the initial cardioprotective effect of monoHER was not prolonged during the half-year observation period. It was even suggested that addition of repeated doses of monoHER tended to aggravate DOX-induced cardiotoxicity. It cannot be excluded that the dose and frequency of monoHER administration is crucial in obtaining an optimal antioxidant activity without a pro-oxidant activity of monoHER.


Assuntos
Doxorrubicina/efeitos adversos , Cardiopatias/prevenção & controle , Hidroxietilrutosídeo/análogos & derivados , Animais , Comportamento Animal/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Cardiopatias/patologia , Hidroxietilrutosídeo/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/ultraestrutura , Tempo , Redução de Peso/efeitos dos fármacos
13.
Cancer Chemother Pharmacol ; 58(5): 699-702, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16565833

RESUMO

PURPOSE: Despite its well-known cardiotoxicity, the anthracyclin doxorubicin (DOX) continues to be an effective and widely used chemotherapeutic agent. DOX-induced cardiac damage presumably results from the formation of free radicals by DOX. Reactive oxygen species particularly affect the cardiac myocytes because these cells seem to have a relatively poor antioxidant defense system. The semisynthetic flavonoid monohydroxyethylrutoside (monoHER) showed cardioprotection against DOX-induced cardiotoxicity through its radical scavenging and iron chelating properties. Because of the relatively short final half-life of monoHER (about 30 min), it is expected that the time interval between monoHER and DOX might be of influence on the cardioprotective effect of monoHER. Therefore, the aim of the present study was to investigate this possible effect. METHODS: Six groups of 6 BALB/c mice were treated with saline, DOX alone or DOX (4 mg/kg i.v.) preceded by monoHER (500 mg/kg i.p.) with an interval of 10, 30, 60 or 120 min. After a 6-week treatment period and additional observation for 2 weeks, the mice were sacrificed. Their cardiac tissues were processed for light microscopy, after which cardiomyocyte damage was evaluated according to Billingham (in Cancer Treat Rep 62(6):865-872, 1978). Microscopic evaluation revealed that treatment with DOX alone induced significant cardiac damage in comparison to the saline control group (P<0.001). RESULTS: The number of damaged cardiomyocytes was 9.6-fold (95% CI 4.4-21.0) higher in mice treated with DOX alone than that in animals of the control group. The ratio of aberrant cardiomyocytes in mice treated with DOX preceded by monoHER and those in mice treated with saline ranged from 1.6 to 2.8 (mean 2.2, 95% CI 1.2-4.1, P=0.019). The mean protective effect by adding monoHER before DOX led to a significant 4.4-fold reduction (P<0.001, 95% CI 2.3-8.2) of abnormal cardiomyocytes. This protective effect did not depend on the time interval between monoHER and DOX administration (P=0.345). CONCLUSION: The results indicate that in an outpatient clinical setting monoHER may be administered shortly before DOX.


Assuntos
Cardiomiopatias/prevenção & controle , Doxorrubicina/administração & dosagem , Hidroxietilrutosídeo/administração & dosagem , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/patologia , Relação Dose-Resposta a Droga , Doxorrubicina/toxicidade , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Vídeo/métodos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fatores de Tempo , Testes de Toxicidade Aguda/métodos , Vacúolos/efeitos dos fármacos , Vacúolos/patologia
14.
Int J Oncol ; 28(1): 237-44, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16328001

RESUMO

Gemcitabine (dFdC) can increase the sensitivity of both cisplatin (CDDP)-sensitive and -resistant cell lines. It has been postulated that both formation and repair of platinum-(Pt)-DNA adducts are related to these effects. Therefore, we investigated the effects of dFdC on the formation and repair of Pt-DNA adducts in the human ovarian cancer cell line, A2780, and its CDDP- or dFdC-resistant variants, ADDP and AG6000, which have a different expression of various repair enzymes. Cells were exposed for 1 h to CDDP alone or combined with dFdC in IC50 concentrations, followed by a 1-h exposure to thiourea and, subsequently, by a drug-free period of 1, 3 or 23 h (i.e. 2, 4 or 24 h after CDPP +/- dFdC removal). Pt-DNA adducts were quantified with 32P-post-labeling. The gene expression of the repair enzymes, XPA and XRCC1, was the same in all 3 cell lines but ERCC1, ERCC3 and XPC were 2-6 times higher in AG6000 compared to A2780 cells. In contrast, both ERCC1 and ERCC3 were 10- and 1.5-fold lower in ADDP cells compared to A2780. The mismatch enzyme, MLH1, was lower in ADDP cells. At equally toxic CDDP concentrations, all cell lines formed comparable peak levels of total Pt-DNA adducts (36-48 fmol/microg DNA). However, the time at which peak levels were reached showed large variation. The repair of the adducts was very efficient in the resistant cell lines whereas, in A2780 cells, plateau levels were retained until 24 h after CDDP exposure. In A2780 cells, dFdC shifted the adduct peaks from 4 h to directly after CDDP exposure and increased peak levels by >3.9-fold. dFdC also enhanced the repair of adducts by >1.7-fold and increased the Pt-GG:Pt-AG ratio compared to CDDP alone by >1.4-fold. Overall, dFdC decreased the area under the Pt-DNA adduct-time curve (AUA0-25 h) in A2780 cells by 2.7-fold. In ADDP cells, dFdC shifted the adduct peaks from 2 to 4 h and increased them by >2.2-fold. dFdC also increased the Pt-GG:Pt-AG ratio during the repair process by 1.4-fold. Overall, dFdC increased the AUA0-25 h in ADDP cells by 1.7-fold. In AG6000 cells, dFdC increased the Pt-GG:Pt-AG ratio by 1.6-fold directly after exposure but did not clearly affect the AUA0-25 h. In conclusion, dFdC can affect both Pt-DNA adduct formation and repair, depending on the initial sensitivity of the cells.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Adutos de DNA , Reparo do DNA/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Neoplasias Ovarianas/patologia , Cisplatino/farmacologia , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Células Tumorais Cultivadas , Gencitabina
15.
Haematologica ; 89(11): 1287-98, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15531450

RESUMO

BACKGROUND AND OBJECTIVES: Oxidant damage is an important contributor to the premature destruction of erythrocytes and anemia in thalassemias. To assess the extent of oxidant damage of circulating erythrocytes and the effects of antioxidant therapy on erythrocyte characteristics and anemia, we used a mouse model of human beta-thalassemia intermedia (b1/b2 deletion). DESIGN AND METHODS: Several parameters indicative of oxidant damage were measured at baseline and following administration of the semi-synthetic flavonoid antioxidant, 7-monohydroxyethylrutoside (monoHER), to beta-thalassemic mice at a dose of either 500 mg/kg i.p. once a day (n=6) or 250 mg/kg i.p. twice a day (n=6) for 21 days. RESULTS: Significant erythrocyte oxidant damage at baseline was indicated by: (i) dehydration, reduced cell K content, and up-regulated K-Cl co-transport; (ii) marked membrane externalization of phosphatidylserine; (iii) reduced plasma and membrane content of vitamin E; and (iv) increased membrane bound IgG. MonoHER treatment increased erythrocyte K content, and markedly improved all cellular indicators of oxidant stress and of lipid membrane peroxidation. While anemia did not improve, monoHER therapy reduced reticulocyte counts, improved survival of a fraction of red cells, and reduced ineffective erythropoiesis with decreased total bilirubin, lactate dehydrogenase and plasma iron. INTERPRETATION AND CONCLUSIONS: Antioxidant therapy reverses several indicators of oxidant damage in vivo. These promising antioxidant effects of monoHER should be investigated further.


Assuntos
Antioxidantes/uso terapêutico , Eritrócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Talassemia beta/sangue , Talassemia beta/tratamento farmacológico , Animais , Anexinas/sangue , Antioxidantes/metabolismo , Cloretos/sangue , Modelos Animais de Doenças , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Feminino , Hidroxietilrutosídeo/análogos & derivados , Hidroxietilrutosídeo/sangue , Hidroxietilrutosídeo/uso terapêutico , Imunoglobulina G/sangue , Transporte de Íons , Masculino , Camundongos , Potássio/sangue , Vitamina E/sangue , Talassemia beta/patologia
16.
Biochim Biophys Acta ; 1683(1-3): 16-21, 2004 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15238215

RESUMO

The antioxidant activities of RRR-vitamin E (VE), all-rac-vitamin E (all-rac-VE), trolox, RRR-vitamin E acetate (VEA), all-rac-vitamin E phosphate (VEP) and RRR-vitamin E succinate (VES) were compared. In this study, the rank order in the inhibition of lipid peroxidation (LPO) of VE and its derivatives was trolox>VE approximately all-rac-VE>VEA>VES. VE and trolox inhibited LPO in non-heated and heated rat liver microsomes. It has generally been accepted that this is due to scavenging of free radicals by these antioxidants, and during this protection the antioxidants are oxidized. VEA and VES have to be converted into VE by esterases to obtain antioxidant activity against LPO. VEP, however, had a potent antioxidant effect of its own without conversion to VE. In contrast to VE, VEP is not consumed during this protection. Of the compounds tested, VEP is the most potent in induction of hemolysis of erythrocytes. EPR experiments using the spin label 16-doxylstearic acid showed that VEP reduces membrane fluidity, in contrast to VE. This indicates that VEP acts as a detergent and forms a barrier that might inhibit the transfer of radicals from one polyunsaturated fatty acid to another. This new mechanism may form the basis for a new class of antioxidants.


Assuntos
Antioxidantes/farmacologia , Membrana Celular/fisiologia , Bicamadas Lipídicas , Peroxidação de Lipídeos/efeitos dos fármacos , Vitamina E/análogos & derivados , Animais , Eritrócitos/química , Eritrócitos/metabolismo , Esterases/metabolismo , Hemólise , Humanos , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Oxirredução , Ratos , Ratos Endogâmicos Lew , Vitamina E/farmacologia
17.
Biochem Pharmacol ; 68(3): 493-502, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15242815

RESUMO

Disodium 2,2'-dithio-bis-ethane sulfonate (BNP7787) is under investigation as a potential new chemoprotector against cisplatin-induced nephrotoxicity. The selective protection of BNP7787 appears to arise from the preferential uptake of the drug in the kidneys, where BNP7787 would undergo intracellular conversion into mesna (2-mercapto ethane sulfonate), which in turn can prevent cisplatin induced toxicities. In the present study, we have investigated whether the reduction of BNP7787 into the reactive compound mesna is restricted to the kidney or whether it can also occur in other organs, cells and physiological compartments, including the cytosolic fraction of the renal cortex, plasma, red blood cells (RBCs), liver and small intestine from rats and several tumors (OVCAR-3, MRI-H-207 and WARD). We also determined whether the endogenous thiols glutathione (GSH) and cysteine and the enzyme systems glutaredoxin and thioredoxin, which are all present in the kidney, can be involved in the BNP7787 reduction. UV detection and micro-HPLC with dual electrochemical detection were used to analyze the various incubation mixtures. Our observations are that, in contrast to plasma, a very large reductive conversion of BNP7787 to mesna was measured in RBC lysate. Intact RBCs, however, did not take up BNP7787. Although BNP7787 could be reduced in cytosol of liver and several tumors, this reduction will not be relevant in vivo, since these tissues do not take up large amounts of BNP7787. Kidney cortex cytosol was, similar to the small intestine cytosol, able to substantially reduce BNP7787 to mesna. The ability to reduce BNP7787 in the presence of the endogenous thiols GSH and cysteine, the glutaredoxin system as well as the thioredoxin system, could at least in part explain the high BNP7787 reductive activity of the kidney cortex cytosol. In conclusion, the high reduction of BNP7787 into mesna in the kidney as well as our earlier observation that the distribution of BNP7787 and mesna was mainly restricted to rat kidney are strong arguments in favor of selective protection of the kidney by BNP7787.


Assuntos
Cisplatino/efeitos adversos , Mesna/análogos & derivados , Mesna/farmacocinética , Substâncias Protetoras/farmacocinética , Animais , Antineoplásicos/efeitos adversos , Cisteína/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Eletroquímica , Enzimas/metabolismo , Eritrócitos/metabolismo , Glutationa/metabolismo , Humanos , Mesna/sangue , Mesna/metabolismo , Mesna/farmacologia , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Distribuição Tecidual
18.
Toxicol Appl Pharmacol ; 194(2): 180-8, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14736498

RESUMO

Production of superoxide radicals from doxorubicin is widely accepted to be the cause of the cardiotoxicity induced by this antitumor agent. Pretreatment with superoxide dismutase could improve the therapeutic application. Aim of the present study was to determine whether lecithinized superoxide dismutase (PC-SOD) can serve as a cardioprotective drug during doxorubicin treatment. The protective potential of PC-SOD on doxorubicin-induced cardiotoxicity was investigated in BALB/c mice. The possible influence of PC-SOD on the antitumor activity of doxorubicin was investigated in vitro as well as in vivo. Mice were treated intravenously with doxorubicin (4 mg x kg(-1)) or doxorubicin and PC-SOD (5000, 20000 or 80000 U x kg(-1)) weekly x 6 and appropriate controls were included. Cardiotoxicity was monitored for 8 weeks by ECG measurement. The influence of PC-SOD on the antitumor activity of doxorubicin was evaluated in three human malignant cell lines. Nude mice bearing OVCAR-3 human ovarian cancer xenografts were treated intravenously with doxorubicin (8 mg x kg(-1)) alone or preceded by PC-SOD 20000 or 80000 U x kg(-1) weekly x 2 and appropriate controls were included. PC-SOD prevented doxorubicin-induced cardiotoxicity already at 5000 U x kg(-1) whereas 20000 and 80000 U x kg(-1) were equally protective. No toxicity was observed in mice treated with PC-SOD. PC-SOD did not interfere with the antiproliferative effects of doxorubicin in vitro. In vivo, PC-SOD had no negative effect on the inhibition of xenograft growth induced by doxorubicin. It can be concluded that PC-SOD protects the heart, but not the tumor against doxorubicin. These data suggest that PC-SOD may be a suitable cardioprotector during doxorubicin treatment.


Assuntos
Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Miocárdio/enzimologia , Miocárdio/patologia , Fosfatidilcolinas/farmacologia , Superóxido Dismutase/farmacologia , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/prevenção & controle , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/toxicidade , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/prevenção & controle , Fosfatidilcolinas/uso terapêutico , Superóxido Dismutase/uso terapêutico
19.
J Biol Chem ; 279(11): 9693-7, 2004 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-14701855

RESUMO

The ability of the sulfur-containing compounds glutathione (GSH), glutathione disulphide (GSSG), S-methylglutathione (GSMe), lipoic acid (LA), and dihydrolipoic acid (DHLA) to protect against hypochlorous acid (HOCl)-mediated damage and peroxynitrite (ONOOH)-induced damage has been compared. Protective activity was assessed in competition assays by monitoring several detectors, i.e. dihydrorhodamine-123 (DHR-123) oxidation, alpha(1)-antiproteinase (alpha(1)-AP) inactivation, and glutathione S-transferase P1-1 (GST-P1-1) inactivation. In addition, nitration of tyrosine was measured to assess protection of the sulfur-containing compounds against ONOOH. For protection against HOCl, the efficacy of the antioxidant was controlled by the ratio of the reaction rates of the antioxidant and the detector molecule with the oxidant. The rank order of the activity of the antioxidants (GSH > DHLA approximately LA approximately GSMe > GSSG) appeared to be independent of the detector used. However, the rank order of the antioxidants against ONOOH-induced damage is strongly dependent on the detector. LA was 40 times less active than GSH in the inhibition of ONOOH-induced DHR-123 oxidation, whereas LA was 20 times more active than GSH in preventing the inhibition of GST-P1-1 by ONOOH. This points to different molecular mechanisms of ONOOH damage to DHR-123 compared with ONOOH damage to GST-P1-1. LA is a poor antioxidant in protecting against the form of ONOOH damage involved in DHR-123 oxidation. In the form of ONOOH toxicity involved in GST-P1-1 inhibition, LA is the most potent sulfur-containing antioxidant in our series. It is proposed that an intermediate product in which both sulfur atoms of LA have reacted is involved in the reaction of ONOOH with LA. The high potency of LA to protect GST-P1-1 against ONOOH might be of therapeutic interest.


Assuntos
Glutationa/metabolismo , Ácido Hipocloroso/metabolismo , Ácido Peroxinitroso/metabolismo , Ácido Tióctico/análogos & derivados , Ácido Tióctico/metabolismo , Ácido Tióctico/farmacologia , Tirosina/análogos & derivados , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Relação Dose-Resposta a Droga , Glutationa S-Transferase pi , Glutationa Transferase/metabolismo , Concentração Inibidora 50 , Isoenzimas/metabolismo , Modelos Biológicos , Modelos Químicos , Óxido Nítrico/química , Oxigênio/metabolismo , Potássio/química , Ligação Proteica , Rodaminas/metabolismo , Enxofre/metabolismo , Tirosina/química , Tirosina/metabolismo
20.
FEBS Lett ; 555(3): 601-5, 2003 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-14675781

RESUMO

The presumed protective effect of folic acid on the pathogenesis of cardiovascular, hematological and neurological diseases and cancer has been associated with the antioxidant activity of folic acid. Peroxynitrite (PON) scavenging activity and inhibition of lipid peroxidation (LPO) of the physiological forms of folate and of structurally related compounds were tested. It was found that the fully reduced forms of folate, i.e. tetrahydrofolate (THF) and 5-methyltetrahydrofolate (5-MTHF), had the most prominent antioxidant activity. It appeared that their protection against LPO is less pronounced than their PON scavenging activity. The antioxidant activity of these forms of folic acid resides in the pterin core, the antioxidant pharmacophore is 4-hydroxy-2,5,6-triaminopyrimidine. It is suggested that an electron donating effect of the 5-amino group is of major importance for the antioxidant activity of 4-hydroxy-2,5,6-triaminopyrimidine. A similar electron donating effect is probably important for the antioxidant activity of THF and 5-MTHF.


Assuntos
Antioxidantes/química , Antioxidantes/farmacologia , Tetra-Hidrofolatos/química , Tetra-Hidrofolatos/farmacologia , Animais , Benzotiazóis , Ácidos Graxos/farmacologia , Ácido Fólico/metabolismo , Ácido Fólico/farmacologia , Sequestradores de Radicais Livres/farmacologia , Concentração Inibidora 50 , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ácido Peroxinitroso/antagonistas & inibidores , Pterinas/química , Pterinas/farmacologia , Ratos , Ratos Endogâmicos Lew , Ácidos Sulfônicos/análise , Ácidos Sulfônicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...