Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Horm Cancer ; 10(2-3): 77-88, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30877616

RESUMO

Androgens are thought to cause prostate cancer, but the underlying mechanisms are unclear. Data from animal studies suggest that for androgens to cause prostate cancer, they must be aromatized to estrogen and act in concert with estrogen metabolites. We tested the hypothesis that androgen-receptor and estrogen receptor-mediated effects of androgen and estrogen are necessary, as well as genotoxicity of estrogen metabolites. NBL rats were treated with androgenic and estrogenic compounds for 16-75 weeks through slow-release silastic implants or pellets. Testosterone alone induced cancer in the prostate of 37% of rats. 5α-Dihydrotestosterone, which cannot be converted to estradiol or testosterone, did not cause a significant prostate cancer incidence (4%). Addition of estradiol to 5α-dihydrotestosterone treatment did not markedly enhance prostate cancer incidence (14%), unlike adding estradiol to testosterone treatment which induced a 100% tumor incidence. Testosterone plus estradiol treatment induced a DNA adduct detectable by 32P-postlabeling, oxidative DNA damage (8-hydroxyguanosine), and lipid peroxidation at the site within the prostate where this treatment causes cancers, preceding later cancer formation. The non-estrogenic 4-hydroxy metabolite of estradiol, when combined with testosterone, induced prostatic dysplasia within 16 weeks and, after long-term treatment, a very low incidence of prostate cancer (21%). When an estrogen that cannot be hydroxylated (2-fluoroestradiol) was added to this combined treatment with testosterone and 4-hydroxyestradiol, dysplasia frequency after 16 weeks was doubled. These results strongly support the hypothesis, but additional definitive studies are needed which may identify new targets to interfere with these mechanisms that are clinically feasible in humans.


Assuntos
Androgênios/efeitos adversos , Carcinogênese , Estrogênios/efeitos adversos , Neoplasias da Próstata/induzido quimicamente , Animais , Carcinoma , Adutos de DNA , Dano ao DNA , Di-Hidrotestosterona/metabolismo , Estradiol/metabolismo , Estrogênios de Catecol/química , Guanosina/análogos & derivados , Guanosina/farmacologia , Humanos , Incidência , Masculino , Próstata , Ratos , Receptores de Estrogênio/metabolismo , Testosterona/metabolismo
2.
ACS Omega ; 3(5): 5511-5515, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29876539

RESUMO

Arsenic trioxide (As2O3) is an environmental carcinogen and a putative endocrine disruptor. Resveratrol has been shown to reverse As2O3-induced oxidative damage. In immortalized but nontransformed estrogen receptor α-negative human breast cells (MCF10A), we observed that 25 µM resveratrol ameliorated As2O3-induced cytotoxicity. As2O3, in the presence or absence of 25 µM resveratrol, induced quinone reductase (NAD(P)H quinone dehydrogenase 1), via the induction of NFE2-related factor 2. As2O3 caused a repression of cytochrome P450 (CYP)1B1, but the addition of 25 µM resveratrol rescued the expression of cytochrome P450 1B1 and kept it at a constant level. Therefore, 25 µM resveratrol can modulate the effects of As2O3 on enzymes involved in estrogen metabolism.

3.
Int J Cancer ; 141(6): 1078-1090, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28388839

RESUMO

Endogenous estrogens become carcinogens when dangerous metabolites, the catechol estrogen quinones, are formed. In particular, the catechol estrogen-3,4-quinones can react with DNA to produce an excess of specific depurinating estrogen-DNA adducts. Loss of these adducts leaves apurinic sites in the DNA, generating subsequent cancer-initiating mutations. Unbalanced estrogen metabolism yields excessive catechol estrogen-3,4-quinones, increasing formation of depurinating estrogen-DNA adducts and the risk of initiating cancer. Evidence for this mechanism of cancer initiation comes from various types of studies. High levels of depurinating estrogen-DNA adducts have been observed in women with breast, ovarian or thyroid cancer, as well as in men with prostate cancer or non-Hodgkin lymphoma. Observation of high levels of depurinating estrogen-DNA adducts in high risk women before the presence of breast cancer indicates that adduct formation is a critical factor in breast cancer initiation. Formation of analogous depurinating dopamine-DNA adducts is hypothesized to initiate Parkinson's disease by affecting dopaminergic neurons. Two dietary supplements, N-acetylcysteine and resveratrol complement each other in reducing formation of catechol estrogen-3,4-quinones and inhibiting formation of estrogen-DNA adducts in cultured human and mouse breast epithelial cells. They also inhibit malignant transformation of these cells. In addition, formation of adducts was reduced in women who followed a Healthy Breast Protocol that includes N-acetylcysteine and resveratrol. When initiation of cancer is blocked, promotion, progression and development of the disease cannot occur. These results suggest that reducing formation of depurinating estrogen-DNA adducts can reduce the risk of developing a variety of types of human cancer.


Assuntos
Adutos de DNA/metabolismo , Estrogênios/metabolismo , Neoplasias/etiologia , Neoplasias/prevenção & controle , Doença de Parkinson/etiologia , Doença de Parkinson/prevenção & controle , Animais , Dopamina/metabolismo , Humanos , Neoplasias/metabolismo , Doença de Parkinson/metabolismo , Purinas/metabolismo
4.
J Rare Dis Res Treat ; 2(3): 22-29, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-30854528

RESUMO

Endogenous estrogens become carcinogens when excessive catechol estrogen quinone metabolites are formed. Specifically, the catechol estrogen-3,4-quinones can react with DNA to produce a large amount of specific depurinating estrogen-DNA adducts, formed at the N-3 of Ade and N-7 of Gua. Loss of these adducts leaves apurinic sites in the DNA, which can generate subsequent cancer-initiating mutations. Unbalanced estrogen metabolism yields excessive catechol estrogen-3,4-quinones, increasing formation of the depurinating estrogen-DNA adducts and the risk of initiating cancer. Evidence for this mechanism of cancer initiation comes from studies in vitro, in cell culture, in animal models and in human subjects. High levels of estrogen-DNA adducts have been observed in women with breast, ovarian or thyroid cancer, and in men with prostate cancer or non-Hodgkin lymphoma. Observation of high levels of depurinating estrogen-DNA adducts in high risk women before the presence of breast cancer indicates that adduct formation is a critical factor in breast cancer initiation. Two dietary supplements, N-acetylcysteine and resveratrol, complement each other in reducing formation of catechol estrogen-3,4-quinones and inhibiting formation of estrogen-DNA adducts in cultured human and mouse breast epithelial cells. They also inhibit malignant transformation of these epithelial cells. In addition, formation of adducts was reduced in women who followed a Healthy Breast Protocol that includes N-acetylcysteine and resveratrol. Blocking initiation of cancer prevents promotion, progression and development of the disease. These results suggest that reducing formation of depurinating estrogen-DNA adducts can reduce the risk of developing a variety of types of human cancer.

5.
Clin Transl Med ; 5(1): 12, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26979321

RESUMO

Estrogens can initiate cancer by reacting with DNA. Specific metabolites of endogenous estrogens, the catechol estrogen-3,4-quinones, react with DNA to form depurinating estrogen-DNA adducts. Loss of these adducts leaves apurinic sites in the DNA, generating mutations that can lead to the initiation of cancer. A variety of endogenous and exogenous factors can disrupt estrogen homeostasis, which is the normal balance between estrogen activating and protective enzymes. In fact, if estrogen metabolism becomes unbalanced and generates excessive catechol estrogen 3,4-quinones, formation of depurinating estrogen-DNA adducts increases and the risk of initiating cancer is greater. The levels of depurinating estrogen-DNA adducts are high in women diagnosed with breast cancer and those at high risk for the disease. High levels of depurinating estrogen-DNA adducts before the presence of breast cancer indicates that adduct formation is a critical factor in breast cancer initiation. Women with thyroid or ovarian cancer also have high levels of estrogen-DNA adducts, as do men with prostate cancer or non-Hodgkin lymphoma. Depurinating estrogen-DNA adducts are initiators of many prevalent types of human cancer. These findings and other discoveries led to the recognition that reducing the levels of estrogen-DNA adducts could prevent the initiation of human cancer. The dietary supplements N-acetylcysteine and resveratrol inhibit formation of estrogen-DNA adducts in cultured human breast cells and in women. These results suggest that the two supplements offer an approach to reducing the risk of developing various prevalent types of human cancer. Graphical abstract Major metabolic pathway in cancer initiation by estrogens.

6.
J Altern Complement Med ; 21(6): 321-6, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25974761

RESUMO

BACKGROUND/OBJECTIVE: A functional medicine approach to reduce breast cancer risk is preferable to early detection and treatment in maintaining breast health. Estrogens are implicated in breast cancer initiation through conversion to metabolites that react with DNA to form specific adducts associated with the development of breast cancer. The purpose of this study was to determine the ability of a defined clinical intervention, the AVERTi-Healthy Breast Program (AHBP), to reduce breast cancer risk conditions likely to develop into breast disease. METHODS: To obtain evidence that risk conditions in breast tissue can be reduced with a defined, multifaceted approach, this small clinical trial of 21 women measured indicators of breast health. A detailed clinical evaluation was conducted with all participants, including identification of physical symptoms, such as areas of tenderness upon palpation. Two laboratory assessments were conducted to determine the efficacy of the AHBP. First, 31 estrogen metabolites, estrogen conjugates, and depurinating estrogen-DNA adducts in urine samples taken before intervention were analyzed. The ratio of DNA adducts to metabolites and conjugates was calculated for each sample. Second, oxidative stress was analyzed by measuring the redox potential of glutathione and cysteine in blood plasma. All assessments were conducted before and after participation. RESULTS: The estrogen adduct ratio and redox potential were improved after 90 days on the AHBP. A significant mean reduction of 3.31 (p=0.03) was observed in the adduct ratio, along with a significant improvement in the redox potential of 3.80 (p=0.05). The significant change in the adduct ratio occurred in women whose oxidative stress profile also improved. CONCLUSION: These significant within-individual decreases suggest that the AHBP can reduce the risk for breast cancer in a relatively short time.


Assuntos
Neoplasias da Mama/prevenção & controle , Mama/fisiologia , Promoção da Saúde , Serviços de Saúde da Mulher , Adulto , Biomarcadores/sangue , Biomarcadores/urina , Feminino , Humanos , Pessoa de Meia-Idade , Estresse Oxidativo/fisiologia , Projetos Piloto
7.
Int J Cancer ; 134(10): 2414-23, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24170413

RESUMO

Greater exposure to estrogens is a risk factor for ovarian cancer. To investigate the role of estrogens in ovarian cancer, a spot urine sample and a saliva sample were obtained from 33 women with ovarian cancer and 34 age-matched controls. Thirty-eight estrogen metabolites, conjugates and DNA adducts were analyzed in the urine samples using ultraperformance liquid chromatography/tandem mass spectrometry, and the ratio of adducts to metabolites and conjugates was calculated for each sample. The ratio of depurinating estrogen-DNA adducts to estrogen metabolites and conjugates was significantly higher in cases compared to controls (p < 0.0001), demonstrating high specificity and sensitivity. DNA was purified from the saliva samples and analyzed for genetic polymorphisms in the genes for two estrogen-metabolizing enzymes. Women with two low-activity alleles of catechol-O-methyltransferase plus one or two high-activity alleles of cytochrome P450 1B1 had higher levels of estrogen-DNA adducts and were more likely to have ovarian cancer. These findings indicate that estrogen metabolism is unbalanced in ovarian cancer and suggest that formation of estrogen-DNA adducts plays a critical role in the initiation of ovarian cancer.


Assuntos
Adutos de DNA/urina , DNA de Neoplasias/urina , Estrogênios/urina , Idoso , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Catecol O-Metiltransferase/genética , Catecol O-Metiltransferase/metabolismo , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP1B1 , Adutos de DNA/química , Adutos de DNA/metabolismo , DNA de Neoplasias/química , DNA de Neoplasias/metabolismo , Estrogênios/química , Estrogênios/metabolismo , Feminino , Frequência do Gene , Genótipo , Humanos , Modelos Lineares , Pessoa de Meia-Idade , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/urina , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Saliva/química , Saliva/metabolismo , Espectrometria de Massas em Tandem
8.
Carcinogenesis ; 34(11): 2587-92, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23843041

RESUMO

Sulforaphane (SFN) is a potent inducer of detoxication enzymes such as NAD(P)H:quinone oxidoreductase 1 (NQO1) and glutathione-S-transferase (GST) via the Kelch-like erythroid-derived protein with CNC homology-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) signaling pathway. NQO1 reduces the carcinogenic estrogen metabolite, catechol estrogen-3,4-quinone, whereas GSTs detoxify it through conjugation with glutathione. These 3,4-quinones can react with DNA to form depurinating DNA adducts. Thus, SFN may alter estrogen metabolism and thus protect against estrogen-mediated DNA damage and carcinogenesis. Human breast epithelial MCF-10A cells were treated with either vehicle or SFN and either estradiol (E2) or its metabolite 4-hydroxyestradiol (4-OHE2). 4-Hydroxy-derived estrogen metabolites and depurinating DNA adducts formed from E2 and its interconvertable metabolite estrone (E1) were analyzed by mass spectrometry. Levels of the depurinated adducts, 4-OHE1/2-1-N3Adenine and 4-OHE1/2-1-N7Guanine, were reduced by 60% in SFN-treated cells, whereas levels of 4-OCH3E1/2 and 4-OHE1/2-glutathione conjugates increased. To constitutively enhance the expression of Nrf2-regulated genes, cells were treated with either scrambled or siKEAP1 RNA. Following E2 or 4-OHE2 treatments, levels of the adenine and guanine adducts dropped 60-70% in siKEAP1-treated cells, whereas 4-OHE1/2-glutathione conjugates increased. However, 4-OCH3E1/2 decreased 50% after siKEAP1 treatment. Thus, treatment with SFN or siKEAP1 has similar effects on reduction of depurinating estrogen-DNA adduct levels following estrogen challenge. However, these pharmacologic and genetic approaches have different effects on estrogen metabolism to O-methyl and glutathione conjugates. Activation of the Nrf2 pathway, especially elevated NQO1, may account for some but not all of the protective effects of SFN against estrogen-mediated DNA damage.


Assuntos
Anticarcinógenos/farmacologia , Mama/efeitos dos fármacos , Adutos de DNA/efeitos dos fármacos , Estrogênios/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Isotiocianatos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Apoptose , Western Blotting , Mama/citologia , Mama/metabolismo , Proliferação de Células , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Sulfóxidos , Espectrometria de Massas em Tandem
9.
Int J Cancer ; 133(11): 2642-9, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23686454

RESUMO

Well-differentiated thyroid cancer most frequently occurs in premenopausal women. Greater exposure to estrogens may be a risk factor for thyroid cancer. To investigate the role of estrogens in thyroid cancer, a spot urine sample was obtained from 40 women with thyroid cancer and 40 age-matched controls. Thirty-eight estrogen metabolites, conjugates and DNA adducts were analyzed by using ultraperformance liquid chromatography/tandem mass spectrometry and the ratio of adducts to metabolites and conjugates was calculated for each sample. The ratio of depurinating estrogen-DNA adducts to estrogen metabolites and conjugates significantly differed between cases and controls (p < 0.0001), demonstrating high specificity and sensitivity. These findings indicate that estrogen metabolism is unbalanced in thyroid cancer and suggest that formation of estrogen-DNA adducts might play a role in the initiation of thyroid cancer.


Assuntos
Adutos de DNA/urina , Estrogênios/metabolismo , Estrogênios/urina , Neoplasias da Glândula Tireoide/urina , Adulto , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/urina , Cromatografia Líquida , Feminino , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Fatores de Risco , Espectrometria de Massas em Tandem , Neoplasias da Glândula Tireoide/patologia
10.
J Steroid Biochem Mol Biol ; 132(1-2): 73-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22386952

RESUMO

This study was conducted to determine whether the ratio of estrogen-DNA adducts to their respective metabolites and conjugates in serum differed between women with early-onset breast cancer and those with average or high risk of developing breast cancer. Serum samples from women at average risk (n=63) or high risk (n=80) for breast cancer (using Gail model) and women newly diagnosed with early breast cancer (n=79) were analyzed using UPLC-MS/MS. Adduct ratios were statistically compared among the three groups, and the Area Under the Receiver Operating Characteristic Curve (AUC) was used to identify a diagnostic cut-off point. The median adduct ratio in the average-risk group was significantly lower than that of both the high-risk group and the breast cancer group (p values<0.0001), and provided good discrimination between those at average versus high risk of breast cancer (AUC=0.84, 95% CI 0.77-0.90). Sensitivity and specificity were maximized at an adduct ratio of 77. For women in the same age and BMI group, the odds of being at high risk for breast cancer was 8.03 (95% CI 3.46-18.7) times higher for those with a ratio of at least 77 compared to those with a ratio less than 77. The likelihood of being at high risk for breast cancer was significantly increased for those with a high adduct ratio relative to those with a low adduct ratio. These findings suggest that estrogen-DNA adducts deserve further study as potential biomarkers for risk of developing breast cancer.


Assuntos
Neoplasias da Mama/sangue , Adutos de DNA/sangue , Estrogênios/sangue , Adulto , Idoso , Biomarcadores/sangue , Feminino , Humanos , Pessoa de Meia-Idade , Risco
11.
Drug Discov Today Dis Mech ; 9(1-2): e55-e69, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-26246832

RESUMO

Metabolism of estrogens via the catechol estrogen pathway is characterized by a balanced set of activating and protective enzymes (homeostasis). Disruption of homeostasis, with excessive production of catechol estrogen quinones, can lead to reaction of these quinones with DNA to form depurinating estrogen-DNA adducts. Some of the mutations generated by these events can lead to initiation of breast cancer. A wealth of evidence, from studies of metabolism, mutagenicity, cell transformation and carcinogenicity, demonstrates that estrogens are genotoxic. Women at high risk for breast cancer, or diagnosed with the disease, have relatively high levels of depurinating estrogen-DNA adducts compared to normal-risk women. The dietary supplements N-acetylcysteine and resveratrol can inhibit formation of catechol estrogen quinones and their reaction with DNA to form estrogen-DNA adducts, thereby preventing initiation of breast cancer.

12.
IUBMB Life ; 63(12): 1087-93, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22045657

RESUMO

The neurotransmitter dopamine is oxidized to its quinone (DA-Q), which at neutral pH undergoes intramolecular cyclization by 1,4-Michael addition, followed by oxidation to form leukochrome, then aminochrome, and finally neuromelanin. At lower pH, the amino group of DA is partially protonated, allowing the competitive intermolecular 1,4-Michael addition with nucleophiles in DNA to form the depurinating adducts, DA-6-N3Ade and DA-6-N7Gua. Catechol estrogen-3,4-quinones react by 1,4-Michael addition to form the depurinating 4-hydroxyestrone(estradiol)-1-N3Ade [4-OHE1(E2)-1-N3Ade] and 4-OHE1(E2)-1-N7Gua adducts, which are implicated in the initiation of breast and other human cancers. The effect of pH was studied by reacting tyrosinase-activated DA with DNA and measuring the formation of depurinating adducts. The most adducts were formed at pH 4, 5, and 6, and their level was nominal at pH 7 and 8. The N3Ade adduct depurinated instantaneously, but N7Gua had a half-life of 3 H. The slow loss of the N7Gua adduct is analogous to that observed in previous studies of natural and synthetic estrogens. The antioxidants N-acetylcysteine and resveratrol efficiently blocked formation of the DA-DNA adducts. Thus, slightly acidic conditions render competitive the reaction of DA-Q with DNA to form depurinating adducts. We hypothesize that formation of these adducts could lead to mutations that initiate Parkinson's disease. If so, use of N-acetylcysteine and resveratrol as dietary supplements may prevent initiation of this disease.


Assuntos
Antioxidantes/química , Adutos de DNA/química , Adutos de DNA/genética , Dopamina/química , Monofenol Mono-Oxigenase/química , Doença de Parkinson/genética , Acetilcisteína/química , Benzoquinonas/química , Benzoquinonas/metabolismo , Adutos de DNA/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Melatonina/química , Doença de Parkinson/patologia , Resveratrol , Estilbenos/química , Ácido Tióctico/química
13.
J Steroid Biochem Mol Biol ; 127(3-5): 276-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21896331

RESUMO

Extensive evidence exists that the reaction of estrogen metabolites with DNA produces depurinating adducts that, in turn, induce mutations and cellular transformation. While it is clear that these estrogen metabolites result in a neoplastic phenotype in vitro, further evidence supporting the link between estrogen-DNA adduct formation and its role in neoplasia induction in vivo would strengthen the evidence for a genotoxic mechanism. Diethylstilbestrol (DES), an estrogen analogue known to increase the risk of breast cancer in women exposed in utero, is hypothesized to induce neoplasia through a similar genotoxic mechanism. Cultured MCF-10F human breast epithelial cells were treated with DES at varying concentrations and for various times to determine whether the addition of DES to MCF-10F cells resulted in the formation of depurinating adducts. This is the first demonstration of the formation of DES-DNA adducts in human breast cells. A dose-dependent increase in DES-DNA adducts was observed. Demonstrating that treatment of MCF-10F cells with DES, a known human carcinogen, yields depurinating adducts provides further support for the involvement of these adducts in the induction of breast neoplasia. Previous studies have demonstrated the ability of antioxidants such as resveratrol to prevent the formation of estrogen-DNA adducts, thus preventing a key carcinogenic event. In this study, when MCF-10F cells were treated with a combination of resveratrol and DES, a dose-dependent reduction in the level of DES-DNA adducts was also observed.


Assuntos
Adutos de DNA/química , Dietilestilbestrol/química , Glândulas Mamárias Humanas/efeitos dos fármacos , Estilbenos/farmacologia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Eletroquímica , Feminino , Humanos , Glândulas Mamárias Humanas/metabolismo , Resveratrol
14.
Biomarkers ; 16(5): 434-44, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21692648

RESUMO

Damage to DNA by dopamine quinone and/or catechol estrogen quinones may play a significant role in the initiation of Parkinson's disease (PD). Depurinating estrogen-DNA adducts are shed from cells and excreted in urine. The aim of this study was to discover whether higher levels of estrogen-DNA adducts are associated with PD. Forty estrogen metabolites, conjugates, and DNA adducts were analyzed in urine samples from 20 PD cases and 40 matched controls by using ultra performance liquid chromatography/tandem mass spectrometry. The levels of adducts in cases versus controls (P < 0.005) suggest that unbalanced estrogen metabolism could play a causal role in the initiation of PD.


Assuntos
Encéfalo/metabolismo , Estrogênios/metabolismo , Doença de Parkinson/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Cromatografia Líquida/métodos , Adutos de DNA/metabolismo , Estrogênios/urina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/metabolismo , Extração em Fase Sólida , Espectrometria de Massas em Tandem
15.
J Steroid Biochem Mol Biol ; 125(3-5): 169-80, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21397019

RESUMO

Among the numerous small molecules in the body, the very few aromatic ones include the estrogens and dopamine. In relation to cancer initiation, the estrogens should be considered as chemicals, not as hormones. Metabolism of estrogens is characterized by two major pathways. One is hydroxylation to form the 2- and 4-catechol estrogens, and the second is hydroxylation at the 16α position. In the catechol pathway, the metabolism involves further oxidation to semiquinones and quinones, including formation of the catechol estrogen-3,4-quinones, the major carcinogenic metabolites of estrogens. These electrophilic compounds react with DNA to form the depurinating adducts 4-OHE(1)(E(2))-1-N3Ade and 4-OHE(1)(E(2))-1-N7Gua. The apurinic sites obtained by this reaction generate the mutations that may lead to the initiation of cancer. Oxidation of catechol estrogens to their quinones is normally in homeostasis, which minimizes formation of the quinones and their reaction with DNA. When the homeostasis is disrupted, excessive amounts of catechol estrogen quinones are formed and the resulting increase in depurinating DNA adducts can lead to initiation of cancer. Substantial evidence demonstrates the mutagenicity of the estrogen metabolites and their ability to induce transformation of mouse and human breast epithelial cells, and tumors in laboratory animals. Furthermore, women at high risk for breast cancer or diagnosed with the disease, men with prostate cancer, and men with non-Hodgkin lymphoma all have relatively high levels of estrogen-DNA adducts, compared to matched control subjects. Specific antioxidants, such as N-acetylcysteine and resveratrol, can block the oxidation of catechol estrogens to their quinones and their reaction with DNA. As a result, the initiation of cancer can be prevented.


Assuntos
Estrogênios/metabolismo , Neoplasias/etiologia , Neoplasias/prevenção & controle , Animais , Neoplasias da Mama/metabolismo , Adutos de DNA/genética , Adutos de DNA/metabolismo , Estrogênios de Catecol/metabolismo , Feminino , Humanos , Masculino , Neoplasias/metabolismo
16.
Free Radic Biol Med ; 50(1): 78-85, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20934508

RESUMO

Substantial evidence suggests that catechol estrogen-3,4-quinones react with DNA to form predominantly the depurinating adducts 4-hydroxyestrone (estradiol)-1-N3Ade [4-OHE(1)(E(2))-1-N3Ade] and 4-OHE(1)(E(2))-1-N7Gua. Apurinic sites resulting from these adducts generate critical mutations that can initiate cancer. The paradigm of cancer initiation is based on an imbalance in estrogen metabolism between activating pathways that lead to estrogen-DNA adducts and deactivating pathways that lead to estrogen metabolites and conjugates. This imbalance can be improved to minimize formation of adducts by using antioxidants, such as resveratrol (Resv) and N-acetylcysteine (NAcCys). To compare the ability of Resv and NAcCys to block formation of estrogen-DNA adducts, we used the human breast epithelial cell line MCF-10F treated with 4-OHE(2). Resv and NAcCys directed the metabolism of 4-OHE(2) toward protective pathways. NAcCys reacted with the quinones and reduced the semiquinones to catechols. This pathway was also carried out by Resv. In addition, Resv induced the protective enzyme quinone reductase, which reduces E(1)(E(2))-3,4-quinones to 4-OHE(1)(E(2)). Resv was more effective at increasing the amount of 4-OCH(3)E(1)(E(2)) than NAcCys. Inhibition of estrogen-DNA adduct formation was similar at lower doses, but at higher doses Resv was about 50% more effective than NAcCys. Their combined effects were additive. Therefore, these two antioxidants provide an excellent combination to protect catechol estrogens from oxidation to catechol quinones.


Assuntos
Acetilcisteína/farmacologia , Neoplasias da Mama/prevenção & controle , Carcinoma/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , Glândulas Mamárias Humanas/efeitos dos fármacos , Estilbenos/farmacologia , Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Neoplasias da Mama/patologia , Carcinoma/patologia , Linhagem Celular , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Humanos , Glândulas Mamárias Humanas/patologia , Modelos Biológicos , Resveratrol , Estilbenos/uso terapêutico
17.
Breast Cancer Res Treat ; 126(3): 779-89, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21170676

RESUMO

Extensive data suggest that estradiol contributes to the development of breast cancer by acting as a mitogen and exerting direct genotoxic effects after enzymatic conversion to 4-hydroxyestradiol (4-OHE2) via cytochrome P450 1B1 (CYP1B1). The mammary gland, ovary, and uterus all express CYP1B1. Overexpression of this enzyme has been associated with an increased risk of breast cancer and blockade might reduce this carcinogenic effect. For this reason, we conducted systematic in vitro and in vivo studies of a CYP1B1 inhibitor, TMS (2,3',4,5'-tetramethoxystilbene). We found that TMS blocked the enzymatic conversion of radiolabeled estradiol to both 2-hydroxyestradiol (2-OHE2) and 4-OHE2, but did not inhibit Cyp1b1 message formation. In vivo studies using mass spectrometry showed that TMS inhibited formation of 2-OHE2 and 4-OHE2 and the resulting estrogen-DNA adducts. To examine its biologic actions in vivo, we investigated whether TMS could block the hyperplastic changes that occur in the developing breast of aromatase-transfected mice. We found that TMS induced a significant reduction of ductal structures in mice less than 6 months in age. In older mice, no reduction in breast morphology occurred. These latter studies uncovered unexpected estrogen agonistic actions of TMS at high doses, including a paradoxical stimulation of breast ductal structures and the endometrium. These studies suggest that the enzyme inhibitory properties of TMS, as well as the effects on developing breast, could implicate a role for TMS in breast cancer prevention, but only in low doses and on developing breast.


Assuntos
Glândulas Mamárias Animais/embriologia , Estilbenos/farmacologia , Animais , Aromatase/metabolismo , Hidrocarboneto de Aril Hidroxilases/biossíntese , Proliferação de Células , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1B1 , Estrogênios/metabolismo , Feminino , Letrozol , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Nitrilas/farmacologia , Ovariectomia/métodos , Triazóis/farmacologia
18.
IUBMB Life ; 62(10): 746-51, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20945454

RESUMO

Bisphenol A (BPA) displays weak estrogenic properties and could be a weak carcinogen by a mechanism similar to that of estrone (E(1)), estradiol (E(2)) and the synthetic estrogen diethylstilbestrol, a human carcinogen. A wide variety of scientific evidence supports the hypothesis that certain estrogen metabolites, predominantly catechol estrogen-3,4-quinones, react with DNA to cause mutations that can lead to the initiation of cancer. One of the major pathways of estrogen metabolism leads to the 4-catechol estrogens, 4-OHE(1)(E(2)), which are oxidized to their quinones, E(1)(E(2))-3,4-Q. The quinones react with DNA to form predominantly the depurinating adducts 4-OHE(1)(E(2))-1-N3Ade and 4-OHE(1)(E(2))-1-N7Gua. This process constitutes the predominant pathway in the initiation of cancer by estrogens. One pathway of BPA metabolism is hydroxylation of one of its symmetric benzene rings to form its catechol, 3-OHBPA. Subsequent oxidation to BPA-3,4-quinone would lead to reaction with DNA to form predominantly the depurinating adducts 3-OHBPA-6-N3Ade and 3-OHBPA-6-N7Gua. The resulting apurinic sites in the DNA could generate mutations in critical genes that can initiate human cancers. The catechol of BPA may also alter expression of estrogen-activating and deactivating enzymes, and/or compete with methoxylation of 4-OHE(1)(E(2)) by catechol-O-methyltransferase, thereby unbalancing the metabolism of estrogens to increase formation of E(1)(E(2))-3,4-Q and the depurinating estrogen-DNA adducts leading to cancer initiation. Thus, exposure to BPA could increase the risk of developing cancer by direct and/or indirect mechanisms. Knowledge of these mechanisms would allow us to begin to understand how BPA may act as a weak carcinogen and would be useful for regulating its use.


Assuntos
Biomarcadores Tumorais/urina , Estrogênios de Catecol/metabolismo , Estrogênios/metabolismo , Fenóis/urina , Compostos Benzidrílicos , Carcinógenos/metabolismo , Catecol O-Metiltransferase/metabolismo , DNA/genética , DNA/metabolismo , Adutos de DNA/metabolismo , Dietilestilbestrol/metabolismo , Estradiol/metabolismo , Estrogênios não Esteroides/metabolismo , Humanos , Mutação , Fenóis/metabolismo , Quinonas/metabolismo
19.
Free Radic Biol Med ; 49(3): 392-400, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20472053

RESUMO

Catechol estrogens, especially 4-hydroxylated metabolites of 17beta-estradiol (E(2)), are responsible for estrogen-induced carcinogenesis. 4-Hydroxyestradiol (4-OHE(2)), a major metabolite of E(2) formed preferentially by cytochrome P-450 1B1, is oxidized to E(2)-3,4-quinone, which can react with DNA to yield the depurinating adducts 4-OHE(2)-1-N3Ade and 4-OHE(2)-1-N7Gua. The apurinic sites generated by the loss of these depurinating adducts induce mutations that could lead to cancer initiation. In this study, we have evaluated the effects of N-acetylcysteine (NAcCys) on the metabolism of two cell lines, MCF-10F (a normal human breast epithelial cell line) and E6 (a normal mouse mammary epithelial cell line), treated with 4-OHE(2) or its reactive metabolite, E(2)-3,4-quinone. Extensive HPLC with electrochemical detection and UPLC-MS/MS analyses of the cell media demonstrated that the presence of NAcCys very efficiently shifted the estrogen metabolism toward protective methoxylation and conjugation pathways in multiple ways, whereas formation of depurinating DNA adducts was inhibited. Protection by NAcCys seems to be similar in both cell lines, irrespective of their origin (human or mouse) or the presence of estrogen receptor-alpha. This finding suggests that NAcCys, a common dietary supplement, could be used as a potential chemopreventive agent to block the initial step in the genotoxicity caused by catechol estrogen quinones.


Assuntos
Acetilcisteína/farmacologia , Adutos de DNA/metabolismo , Estrogênios de Catecol/metabolismo , Animais , Antioxidantes/farmacologia , Mama/metabolismo , Neoplasias da Mama/prevenção & controle , Linhagem Celular , Estradiol/análogos & derivados , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos
20.
Free Radic Biol Med ; 48(2): 318-24, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19909805

RESUMO

Catechol quinones of estrogens react with DNA by 1,4-Michael addition to form depurinating N3Ade and N7Gua adducts. Loss of these adducts from DNA creates apurinic sites that can generate mutations leading to cancer initiation. We compared the reactions of the catechol quinones of the leukemogenic benzene (CAT-Q) and N-acetyldopamine (NADA-Q) with 2'-deoxyguanosine (dG) or DNA. NADA was used to prevent intramolecular cyclization of dopamine quinone. Reaction of CAT-Q or NADA-Q with dG at pH 4 afforded CAT-4-N7dG or NADA-6-N7dG, which lost deoxyribose with a half-life of 3 h to form CAT-4-N7Gua or 4 h to form NADA-6-N7Gua. When CAT-Q or NADA-Q was reacted with DNA, N3Ade adducts were formed and lost from DNA instantaneously, whereas N7Gua adducts were lost over several hours. The maximum yield of adducts in the reaction of CAT-Q or NADA-Q with DNA at pH 4 to 7 was at pH 4. When tyrosinase-activated CAT or NADA was reacted with DNA at pH 5 to 8, adduct levels were much higher (10- to 15-fold), and the highest yield was at pH 5. Reaction of catechol quinones of natural and synthetic estrogens, benzene, naphthalene, and dopamine with DNA to form depurinating adducts is a common feature that may lead to initiation of cancer or neurodegenerative disease.


Assuntos
Benzeno/metabolismo , Adutos de DNA/efeitos adversos , Dopamina/metabolismo , Neoplasias/etiologia , Doenças Neurodegenerativas/etiologia , Aminobutiratos/farmacologia , Animais , Benzeno/química , Catecóis/química , Bovinos , Transformação Celular Neoplásica , Cromatografia Líquida de Alta Pressão , Adutos de DNA/química , Dopamina/análogos & derivados , Dopamina/química , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Mutagênese/efeitos dos fármacos , Mutagênese/genética , Mutagênicos/efeitos adversos , Mutagênicos/química , Naftalenos/química , Naftalenos/metabolismo , Quinonas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...