Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Oncol ; 23(5): 1407-12, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14532983

RESUMO

Black cohosh is an increasingly popular alternative to estrogen replacement therapy for the relief of menopausal symptoms, primarily hot flushes. However, an important consideration for long-term therapy is potential toxicity and carcinogenicity. Therefore, we undertook a study to assess the estrogenic activity of black cohosh to examine its safety for those with, or at high risk of developing, breast cancer. Several assays were utilized as listed: RNAse protection assays, which ascertain the regulation of the expression of E2-responsive genes; estrogen-responsive-element (ERE)-luciferase, which determines modulation of the ER function by transactivation of the ERE; the Ishikawa cell system, which has an E2-regulated endogenous alkaline phosphatase; and colony formation of ER-expressing breast cancer cells, which indicates possible progression of early stage breast cancer into a more aggressive state. Black cohosh extracts did not demonstrate estrogenic activity in any of these assay systems. This is an encouraging step in the assessment of the safety of black cohosh for treatment of menopausal hot flushes.


Assuntos
Neoplasias da Mama/patologia , Cimicifuga/metabolismo , Estrogênios/metabolismo , Menopausa/efeitos dos fármacos , Extratos Vegetais , Fosfatase Alcalina/metabolismo , Neoplasias da Mama/tratamento farmacológico , Adesão Celular , Divisão Celular , Linhagem Celular Tumoral , Humanos , RNA Mensageiro/metabolismo , Receptores de Estrogênio/metabolismo , Ribonucleases/metabolismo
2.
Chem Res Toxicol ; 14(1): 82-90, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11170511

RESUMO

Estrogen replacement therapy has been correlated with an increased risk of developing hormone-dependent cancers. 4-Hydroxyequilenin (4-OHEN) is a catechol metabolite of equilenin and equilin which are components of the estrogen replacement formulation marketed under the name of Premarin (Wyeth-Ayerst). Previously, we showed that 4-OHEN autoxidizes to potent cytotoxic quinoids which can consume reducing equivalents and molecular oxygen, and cause a variety of DNA lesions, including formation of bulky stable adducts, apurinic sites, and oxidation of the phosphate-sugar backbone and purine/pyrimidine bases [Bolton, J. L., Pisha, E., Zhang, F., and Qiu, S. (1998) Chem. Res. Toxicol. 11, 1113-1127]. All of these deleterious effects could contribute to the cytotoxic/genotoxic effects of equine estrogens in vivo. In the study presented here, we studied the oxidative and carcinogenic potential of 4-OHEN and the catechol metabolite of the endogenous estrogen, 4-hydroxyestrone (4-OHE), in the JB6 clone 41 5a and C3H 10T(1/2) murine fibroblast cells. The relative ability of 4-OHEN and 4-OHE to induce oxidative stress was measured in these cells by oxidative cleavage of 2',7'-dichlorodiacylfluorosceindiacetate to dichlorofluoroscein. 4-OHEN (1 microM) displayed an increase in the level of reactive oxygen species comparable to that observed with 100 microM H(2)O(2). In contrast, 4-OHE demonstrated antioxidant capabilities in the 5-50 microM range. With both cell lines, we assessed single-strand DNA cleavage using the comet assay and the formation of oxidized DNA bases, such as 8-oxodeoxyguanosine, utilizing the Trevigen Fpg comet assay. 4-OHEN caused single-strand breaks and oxidized bases in a dose-dependent manner in both cell lines, whereas 4-OHE did not induce DNA damage. Since oxidative stress has been implicated in cellular transformation, we used the JB6 clone 41 5a anchorage independence assay to ascertain the relative ability of 4-OHEN and 4-OHE to act as tumor promoters. 4-OHEN caused a slight but significant increase in the extent of cellular transformation at the 100 nM dose; however, in the presence of NADH, which catalyzes redox cycling of 4-OHEN, the transformation ability of 4-OHEN was dramatically increased. 4-OHE did not induce transformation of the JB6 clone 41 5a in the 0.1-10 microM range. The initiation, promotion, and complete carcinogenic transformation potentials of both metabolites were measured in the C3H 10T(1/2) cells. 4-OHEN demonstrated activity in all stages of transformation at doses of 10 nM to 1 microM, whereas 4-OHE only demonstrated promotional capabilities at the 10 microM dose. These data suggest that oxidative stress could be partially responsible for the carcinogenic effects caused by 4-OHEN and that 4-OHEN is a more potent transforming agent than 4-OHE in vitro.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Equilenina/análogos & derivados , Equilenina/toxicidade , Congêneres do Estradiol/toxicidade , Animais , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Linhagem Celular , Dano ao DNA , Células Epidérmicas , Epiderme/efeitos dos fármacos , Equilenina/metabolismo , Congêneres do Estradiol/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Cavalos , Hidroxiestronas/metabolismo , Hidroxiestronas/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Espécies Reativas de Oxigênio/metabolismo
3.
Chem Res Toxicol ; 13(5): 342-50, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10813650

RESUMO

Estrogen replacement therapy has been correlated with an increased risk of developing breast or endometrial cancer. 4-Hydroxyequilenin (4-OHEN) is a catechol metabolite of equilenin which is a minor component of the estrogen replacement formulation marketed under the name of Premarin (Wyeth-Ayerst). Previously, we showed that 4-OHEN autoxidizes to quinoids which can consume reducing equivalents and molecular oxygen, are potent cytotoxins, and cause a variety of damage to DNA, including formation of bulky stable adducts, apurinic sites, and oxidation of the phosphate-sugar backbone and purine/pyrimidine bases [Bolton, J. L., Pisha, E., Zhang, F., and Qiu, S. (1998) Chem. Res. Toxicol. 11, 1113-1127]. All of these deleterious effects could contribute to the cytotoxic and genotoxic effects of equilenin in vivo. In the study presented here, we examined the relative toxicity of 4-OHEN in estrogen receptor (ER) positive cells (MCF-7 and S30) compared to that in breast cancer cells without the estrogen receptor (MDA-MB-231). The data showed that 4-OHEN was 4-fold more toxic to MCF-7 cells (LC(50) = 6.0 +/- 0. 2 microM) and 6-fold more toxic to S30 cells (LC(50) = 4.0 +/- 0.1 microM) than to MDA-MB-231 cells (LC(50) = 24 +/- 0.3 microM). Using the single-cell gel electrophoresis assay (comet assay) to assess DNA damage, we found that 4-OHEN causes concentration-dependent DNA single-strand cleavage in all three cell lines, and this effect could be enhanced by agents which catalyze redox cycling (NADH) or deplete cellular GSH (diethyl maleate). In addition, the ER(+) cell lines (MCF-7 and S30) were considerably more sensitive to induction of DNA damage by 4-OHEN than the ER(-) cells (MDA-MB-231). 4-OHEN also caused a concentration-dependent increase in the amount of mutagenic lesion 8-oxo-dG in the S30 cells as determined by LC/MS-MS. Cell morphology assays showed that 4-OHEN induces apoptosis in these cell lines. As observed with the toxicity assay and the comet assay, the ER(+) cells were more sensitive to induction of apoptosis by 4-OHEN than MDA-MB-231 cells. Finally, the endogenous catechol estrogen metabolite 4-hydroxyestrone (4-OHE) was considerably less effective at inducing DNA damage and apoptosis in breast cancer cell lines than 4-OHEN. Our data suggest that the cytotoxic effects of 4-OHEN may be related to its ability to induce DNA damage and apoptosis in hormone sensitive cells in vivo, and these effects may be potentiated by the estrogen receptor.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Dano ao DNA/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Equilenina/análogos & derivados , Congêneres do Estradiol/toxicidade , Neoplasias da Mama/tratamento farmacológico , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Fragmentação do DNA , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Equilenina/metabolismo , Equilenina/toxicidade , Congêneres do Estradiol/metabolismo , Estrogênios de Catecol/toxicidade , Feminino , Humanos , Hidroxiestronas/toxicidade , Células Tumorais Cultivadas
4.
Chem Res Toxicol ; 12(2): 204-13, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10027800

RESUMO

The risk factors for women developing breast and endometrial cancers are all associated with a lifetime of estrogen exposure. Estrogen replacement therapy in particular has been correlated with a slight increased cancer risk. Previously, we showed that equilenin, a minor component of Premarin (Wyeth-Ayerst), was metabolized to highly cytotoxic quinoids which caused oxidative stress and alkylation of DNA in vitro [Bolton, J. L., Pisha, E., Zhang, F., and Qiu, S. (1998) Chem. Res. Toxicol. 11, 1113-1127]. In this study, we have compared the chemistry of the major catechol metabolite of equilin (4-hydroxyequilin), which is found in several estrogen replacement formulations, to the equilenin catechol (4-hydroxyequilenin). Unlike endogenous catechol estrogens, both equilin and equilenin were primarily converted by rat liver microsomes to 4-hydroxylated rather than 2-hydroxylated o-quinone GSH conjugates. With equilin, a small amount of 2-hydroxyequilin GSH quinoids were detected (4-hydroxyequilin:2-hydroxyequilin ratio of 6:1); however, no peaks corresponding to 2-hydroxyequilenin were observed in incubations with equilenin. These data suggest that unsaturation in the B ring alters the regiochemistry of P450-catalyzed hydroxylation from primarily 2-hydroxylation for endogenous estrogens to 4-hydroxylation for equine estrogens. 4-Hydroxyequilenin-o-quinone reacts with GSH to give two mono-GSH conjugates and one di-adduct. The behavior of 4-hydroxyequilin was found to be more complex than 4-hydroxyequilenin as conjugates resulting from 4-hydroxyequilenin were detected in addition to the 4-hydroxyequilin-GSH adducts. The mechanism of decomposition of 4-hydroxyequilin likely involves isomerization to a quinone methide which readily aromatizes to 4-hydroxyequilenin followed by autoxidation to 4-hydroxyequilenin-o-quinone. Similar results were obtained with 2-hydroxyequilin, although, in contrast to 4-hydroxyequilenin, 2-hydroxyequilenin does not autoxidize and the reaction stops at the catechol. Since 4-hydroxyequilin is converted to 4-hydroxyequilenin and 4-hydroxyequilenin-o-quinone, similar effects were observed for this equine catechol, including consumption of NAD(P)H likely by the 4-hydroxyequilenin-o-quinone, depletion of molecular oxygen by 4-hydroxyequilenin or its semiquinone radical, and alkylation of deoxynucleosides and DNA by 4-hydroxyequilenin quinoids. Finally, preliminary studies conducted with the human breast tumor cell line MCF-7 demonstrated that the cytotoxic effects of the catechol estrogens from estrone, equilin, and 2-hydroxyequilenin were similar, whereas 4-hydroxyequilenin was a much more potent cytotoxin ( approximately 30-fold). These results suggest that the catechol metabolites of equine estrogens have the ability to cause alkylation/redox damage in vivo primarily through formation of 4-hydroxyequilenin quinoids.


Assuntos
Equilenina/análogos & derivados , Equilina/análogos & derivados , Congêneres do Estradiol/metabolismo , Quinonas/metabolismo , Quinonas/farmacologia , Animais , Neoplasias da Mama/tratamento farmacológico , Sistema Enzimático do Citocromo P-450/metabolismo , Equilenina/metabolismo , Equilenina/farmacologia , Equilina/metabolismo , Equilina/farmacologia , Congêneres do Estradiol/farmacologia , Estrona/farmacologia , Feminino , Humanos , Microssomos Hepáticos/metabolismo , Oxirredução , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Células Tumorais Cultivadas
6.
J Nat Prod ; 61(10): 1257-62, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9784163

RESUMO

Four novel steroidal alkaloids, (+)-(20S)-20-(dimethylamino)-3-(3'alpha-isopropyl)-lactam-5alpha-+ ++preg n-2-en-4-one (1), (+)-(20S)-20-(dimethylamino)-16alpha-hydroxy-3-(3'alpha-isopropyl) -la ctam-5alpha-pregn-2-en-4-one (2), (+)-(20S)-3-(benzoylamino)-20-(dimethylamino)-5alpha-pregn-2-en-++ +4beta -yl acetate (3), and (+)-(20S)-2alpha-hydroxy-20-(dimethylamino)-3beta-phthalimido-5 alpha- pregnan-4beta-yl acetate (4), as well as five known compounds, (-)-pachyaximine A (5), (+)-spiropachysine (6), (+)-axillaridine A (7), (+)-epipachysamine D (8), and (+)-pachysamine B (9), were isolated from Pachysandra procumbens, using a bioassay-guided fractionation based on inhibition of 3H-tamoxifen binding at the antiestrogen binding site (AEBS). Compounds 1-7 and 9 demonstrated significant activity as AEBS-inhibitory agents, and compounds 3, 5 and 9 were found to potentiate significantly the antiestrogenic effect mediated by tamoxifen in cultured Ishikawa cells. The structure elucidation of compounds 1-4 was carried out by spectral data interpretation.


Assuntos
Alcaloides/isolamento & purificação , Antagonistas de Estrogênios/metabolismo , Fitosteróis/isolamento & purificação , Plantas/química , Alcaloides/química , Alcaloides/farmacologia , Animais , Sítios de Ligação , Linhagem Celular , Feminino , Espectroscopia de Ressonância Magnética , Fitosteróis/química , Fitosteróis/farmacologia , Ratos , Ratos Sprague-Dawley , Tamoxifeno/antagonistas & inibidores , Tamoxifeno/farmacologia
7.
Bioorg Med Chem Lett ; 8(13): 1707-12, 1998 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-9873420

RESUMO

Betulinic acid has been modified at C-3, C-20, and C-28 positions and the toxicity of the derivatives has been evaluated against cultured human melanoma (MEL-2) and human epidermoid carcinoma of the mouth (KB) cell lines. This preliminary investigation demonstrates that simple modifications of the parent structure of betulinic acid can produce potentially important derivatives, which may be developed as antitumor drugs.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Melanoma/patologia , Triterpenos/síntese química , Triterpenos/farmacologia , Antineoplásicos/química , Carcinoma de Células Escamosas/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Ligação de Hidrogênio , Neoplasias Bucais/patologia , Triterpenos Pentacíclicos , Triterpenos/química , Ácido Betulínico
8.
Chem Biol Interact ; 106(2): 133-48, 1997 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-9366899

RESUMO

Catechols are widespread in the environment, especially as constituents of edible plants. A number of these catechols may undergo oxidative metabolism to electrophilic o-quinones (3,5-cyclohexadien-1,2-dione) by oxidative enzymes such as cytochrome P450 and peroxidases. Alkylation of cellular nucleophiles by these intermediates and the formation of reactive oxygen species, especially through redox cycling of o-quinones, could contribute to the cytotoxic properties of the parent catechols. In contrast, isomerization of the o-quinones to electrophilic quinone methides (4-methylene-2,5-cyclohexadien-1-one, QM) could cause cellular damage primarily through alkylation. In this investigation, we treated human melanoma cells with two groups of catechols. These cells have high levels of tyrosinase required to oxidize catechols to quinoids. For catechols which are oxidized to o-quinones that cannot isomerize to quinone methides or form unstable quinone methides, plots of the cytotoxicity data (ED50) versus the reactivity of the o-quinones gave an excellent linear correlation; decreasing o-quinone reactivity led to a decrease in the cytotoxic potency of the catechol. In contrast, catechols which are metabolized by the o-quinone/p-quinone methide bioactivation pathway were equally cytotoxic but showed no correlation between the reactivity of the o-quinones and the cytotoxic potency of the catechols. The most likely explanation for this effect is a change in cytotoxic mechanism from o-quinone-mediated inhibition of cell growth to a bioactivation pathway based on both o-quinone and p-QM formation. These results substantiate the conclusion that the involvement of the o-quinone/ QM pathway in catechol toxicity depends on a combination between the rate of enzymatic formation of the o-quinone, the rate of isomerization to the more electrophilic QM, and the chemical reactivity of the quinoids.


Assuntos
Catecóis/farmacologia , Melanócitos/efeitos dos fármacos , Melanoma/patologia , Quinonas/metabolismo , Biotransformação , Catecóis/síntese química , Catecóis/metabolismo , Morte Celular/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Isomerismo , Cinética , Espectroscopia de Ressonância Magnética , Melanócitos/citologia , Melanócitos/metabolismo , Melanoma/metabolismo , Modelos Químicos , Estrutura Molecular , Monofenol Mono-Oxigenase/metabolismo , Oxirredução , Oxirredutases/metabolismo , Quinonas/química , Quinonas/farmacologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas
9.
Carcinogenesis ; 18(5): 1093-101, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9163701

RESUMO

There is a clear association between excessive exposure to estrogens and the development of cancer in several tissues including breast and endometrium. The risk factors for women developing these cancers are all associated with longer estrogen exposure, as may be facilitated by early menses, late menopause and long-term estrogen replacement therapy. Equilenin (1,3,5(10),6,8-estrapentaen-3-ol-17-one) or its 17-hydroxylated analogs make up 15% of the most widely prescribed estrogen replacement formulation, Premarin, and yet there is very little information on the human metabolism of these estrogens. In this study, we synthesized the catechol metabolite of equilenin, 4-hydroxyequilenin, and examined how aromatization of the B ring affects the formation and reactivity of the o-quinone (3,5-cyclohexadien-1,2-dione). 4-Hydroxyequilenin-o-quinone is much more redox-active and longer-lived than the endogenous catechol estrone-o-quinones, which suggests that the mechanism(s) of toxicity of the former could be quite different. Interestingly, the rate of reduction of the 4-hydroxyequilenin-o-quinone is increased at least 13-fold in the presence of NAD(P)H:quinone oxidoreductase (DT-diaphorase). Once NADH is consumed however, the catechol auto-oxidized rapidly to the o-quinone. NADH consumption was accompanied by dicumarol-sensitive oxygen uptake both with the purified enzyme and with cytosol from human melanoma cells with high levels of DT-diaphorase activity. P450 reductase and rat liver microsomes also catalyzed NADPH consumption and oxygen uptake. 4-Hydroxyestrone-o-quinone was also rapidly reduced by NAD(P)H; however, this o-quinone does not auto-oxidize and once the o-quinone is reduced the reaction terminates. Including oxidative enzymes in the incubation completes the redox couple and 4-hydroxyestrone-o-quinone behaves like 4-hydroxyequilenin-o-quinone. These data suggest that reduction of estrogen-o-quinones may not result in detoxification. Instead this could represent a cytotoxic mechanism involving consumption of reducing equivalents (NADH/NADPH) as well as formation of superoxide and other reactive oxygen species leading to oxidative stress. Finally, we have compared the cytotoxicity of 4-hydroxyequilenin with that of the estrone catechols in human melanoma cells. 4-Hydroxyequilenin is 5-fold more toxic in these cells compared with 4-hydroxyestrone (ED50 = 7.8 versus 38 microM, respectively) suggesting that formation of the longer-lived redox-active 4-hydroxyequilenin-o-quinone was responsible for the cytotoxic differences. These results substantiate the conclusion that the involvement of quinoids in catechol estrogen toxicity depends on a combination of the rate of formation of the o-quinone, the lifetime of the o-quinone, and the electrophilic/redox reactivity of the quinoids.


Assuntos
Equilenina/análogos & derivados , Estrogênios de Catecol/toxicidade , Animais , Equilenina/química , Equilenina/metabolismo , Humanos , Cinética , Melanoma/metabolismo , Microssomos Hepáticos/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADP/metabolismo , Oxirredução , Consumo de Oxigênio , Quinonas/metabolismo , Ratos , Células Tumorais Cultivadas
10.
Nat Med ; 1(10): 1046-51, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7489361

RESUMO

As a result of bioassay-guided fractionation, betulinic acid, a pentacyclic triterpene, was identified as a melanoma-specific cytotoxic agent. In follow-up studies conducted with athymic mice carrying human melanomas, tumour growth was completely inhibited without toxicity. As judged by a variety of cellular responses, antitumour activity was mediated by the induction of apoptosis. Betulinic acid is inexpensive and available in abundant supply from common natural sources, notably the bark of white birch trees. The compound is currently undergoing preclinical development for the treatment or prevention of malignant melanoma.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Dano ao DNA , Inibidores do Crescimento/farmacologia , Melanoma/patologia , Triterpenos/farmacologia , Animais , Apoptose , Guanidinas/farmacologia , Humanos , Neoplasias Hepáticas/patologia , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Triterpenos Pentacíclicos , Putrescina/farmacologia , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Ácido Betulínico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA