Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
J Biochem Mol Toxicol ; 32(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-28960787

ABSTRACT

The objective of the present study was to characterize the role of novel resveratrol (Res) analogs: 4-(E)-{(4-hydroxyphenylimino)-methylbenzene, 1, 2-diol} (HPIMBD) and 4-(E)-{(p-tolylimino)-methylbenzene-1,2-diol} (TIMBD) as potent antioxidants against breast cancer. Non-neoplastic breast epithelial cell lines MCF-10A and MCF-10F were treated with 17ß-estradiol (E2), Res, HPIMBD, and TIMBD for up to 72 h. mRNA and protein levels of antioxidant genes, superoxide dismutase 3 (SOD3) and N-quinoneoxidoreductase-1 (NQO1) and transcription factors, nuclear factor erythroid 2-related factor (Nrf) 1, 2 and 3 were quantified after the above treatments. Generation of reactive oxygen species (ROS) was measured by CM-H2-DCFDA and oxidative-DNA damage was determined by measuring 8-hydroxy-2-deoxyguanosine (8-OHdG). HPIMBD and TIMBD scavenged cellular ROS production, attenuated oxidative DNA damage, increased mRNA and protein expression levels of SOD3 and NQO1 and activated Nrf signaling pathway. Our studies demonstrate that HPIMBD and TIMBD have the potential as novel antioxidants to prevent development of breast cancer.


Subject(s)
Anticarcinogenic Agents/metabolism , Antioxidants/metabolism , Breast Neoplasms/prevention & control , Breast/metabolism , Catechols/metabolism , Schiff Bases/metabolism , Stilbenes/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Anticarcinogenic Agents/adverse effects , Antioxidants/adverse effects , Breast/cytology , Breast/pathology , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catechols/adverse effects , Cell Line , Cell Proliferation , Cell Survival , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Dietary Supplements/adverse effects , Enzyme Induction , Estradiol/adverse effects , Female , Humans , NAD(P)H Dehydrogenase (Quinone)/chemistry , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Resveratrol , Schiff Bases/adverse effects , Signal Transduction , Stilbenes/adverse effects , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
2.
Oncotarget ; 7(32): 51747-51762, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27351134

ABSTRACT

We have recently shown that 4-(E)-{(4-hydroxyphenylimino)-methylbenzene, 1,2-diol} (HPIMBD) and 4-(E)-{(p-tolylimino)-methylbenzene-1,2-diol} (TIMBD), novel analogs of resveratrol (Res), selectively inhibited the proliferation of breast cancer cells. In the current study, we tested HPIMBD and TIMBD individually in combination with tamoxifen (Tam) for inhibition of growth of breast cancer cells. Tamoxifen was first tested on non-neoplastic breast epithelial cell lines and its dose that does not inhibit their growth was determined. A combination of this low dose of Tam with either of the Res analogs HPIMBD or TIMBD, resulted in synergistic inhibition of proliferation of breast cancer cells. Both estrogen receptor (ER)-positive and negative breast cancer cell lines responded to the combination. The combination resulted in a substantial decrease in IC50 values of Res analogs in all breast cancer cell lines tested. Mechanistic studies showed a synergistic increase in apoptosis and autophagy genes (beclin-1 and LC3BII/I) with the combination in ER-negative MDA-MB-231 cells. In ER-positive MCF-7 and T47D cells, the mechanism of synergy was found to be inhibition of expression of ERα and oncogene c-Myc. The combination treatment had a synergistic effect in inhibiting the colony forming and spheroid forming ability of cancer cells. Taken together, our findings indicate that a combination of Tam and Res analogs HPIMBD or TIMBD represents a novel approach to enhancing the use of Tam in therapy for breast cancers. Considering the urgent need for novel therapeutic strategies to treat ER-negative breast cancers and overcoming resistance in ER-positive cancers, this combinatorial approach is worthy of continued investigation.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Catechols/pharmacology , Cell Proliferation/drug effects , Schiff Bases/pharmacology , Stilbenes/pharmacology , Tamoxifen/pharmacology , Apoptosis/drug effects , Cells, Cultured , Drug Synergism , Female , Humans , MCF-7 Cells , Resveratrol , Stilbenes/chemistry
3.
Toxicol Appl Pharmacol ; 301: 1-13, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26970359

ABSTRACT

Breast cancer is a public health concern worldwide. Prolonged exposure to estrogens has been implicated in the development of breast neoplasms. Epidemiologic and experimental evidence suggest a chemopreventive role of phytoestrogens in breast cancers. Resveratrol, a naturally occurring phytoestrogen, has been shown to have potent anti-cancer properties. However, poor efficacy and bioavailability have prevented the use of resveratrol in clinics. In order to address these problems, we have synthesized a combinatorial library of azaresveratrol analogs and tested them for their ability to inhibit the proliferation of breast cancer cells. We have recently shown that 4-(E)-{(p-tolylimino)-methylbenzene-1,2-diol} (TIMBD), has better anti-cancer properties than resveratrol and any other resveratrol analog we have synthesized so far. The objective of this study was to investigate the regulation of estrogen receptors (ERs) α and ß by TIMBD in breast cancer cell lines. We demonstrate that TIMBD significantly induces the mRNA and protein expression levels of ERß and inhibits that of ERα. TIMBD inhibits mRNA and protein expression levels of oncogene c-Myc, and cell cycle protein cyclin D1, which are important regulators of cellular proliferation. TIMBD significantly induces protein expression levels of tumor suppressor genes p53 and p21 in MCF-7 cells. TIMBD inhibits c-Myc in an ERß-dependent fashion in MCF-10A and ERß1-transfected MDA-MB-231 cells, suggesting regulation of ERs as an important upstream mechanism of this analog. ERß plays a partial role in inhibition of proliferation by TIMBD while ERα overexpression does not significantly affect TIMBD's inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Stilbenes/pharmacology , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Humans , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Resveratrol
4.
J Steroid Biochem Mol Biol ; 144 Pt B: 500-12, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25242450

ABSTRACT

Breast cancer is the second leading cause of death among women in the United States. Estrogens have been implicated as major risk factors in the development of breast neoplasms. Recent epidemiologic studies have suggested a protective role of phytoestrogens in prevention of breast and other cancers. Resveratrol, a naturally occurring phytoestrogen found notably in red grapes, berries and peanuts, has been shown to possess potent anti-cancer properties. However, the poor efficacy of resveratrol has prevented its use in a clinical setting. In order to improve the efficacy of resveratrol, we have synthesized a small combinatorial library of azaresveratrol analogs and tested them for their ability to inhibit the growth of breast cancer cell lines. We have recently shown that one of the synthesized analogs, 4-(E)-{(4-hydroxyphenylimino)-methylbenzene,1,2-diol} (HPIMBD), has better anti-cancer properties than resveratrol. The objective of this study was to investigate the differential regulation of estrogen receptors (ERs) α and ß as a potential mechanism of inhibition of breast cancer by HPIMBD. Estrogen receptors α and ß have been shown to have opposing roles in cellular proliferation. Estrogen receptor α mediates the proliferative responses of estrogens while ERß plays an anti-proliferative and pro-apoptotic role. We demonstrate that HPIMBD significantly induces the expression of ERß and inhibits the expression of ERα. HPIMBD also inhibits the protein expression levels of oncogene c-Myc and cell cycle protein cyclin D1, genes downstream to ERα and important regulators of cell cycle, and cellular proliferation. HPIMBD significantly induces protein expression levels of tumor suppressors p53 and p21 in MCF-7 cells. Additionally, HPIMBD inhibits c-Myc in an ERß-dependent fashion in MCF-10A and ERß1-transfected MDA-MB-231 cells, suggesting regulation of ERs as an important upstream mechanism of this novel compound. Molecular docking studies confirm higher affinity for binding of HPIMBD in the ERß cavity. Thus, HPIMBD, a novel azaresveratrol analog may inhibit the proliferation of breast cancer cells by differentially modulating the expressions of ERs α and ß.


Subject(s)
Antineoplastic Agents/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Schiff Bases/pharmacology , para-Aminobenzoates/pharmacology , Catechols , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Humans , Molecular Docking Simulation , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Resveratrol , Stilbenes
5.
J Biochem Mol Toxicol ; 28(12): 529-38, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25130429

ABSTRACT

The objective of the present study was to characterize the role of resveratrol (Res) and vitamin C (VC) in prevention of estrogen-induced breast cancer through regulation of cap "n"collar (CNC) b-zip transcription factors. Human breast epithelial cell line MCF-10A was treated with 17ß-estradiol (E2) and VC or Res with or without E2. mRNA and protein expression levels of CNC b-zip transcription factors nuclear factor erythroid 2-related factor 1 (Nrf1), nuclear factor erythroid 2 related factor 2 (Nrf2), nuclear factor erythroid 2 related factor 3 (Nrf3), and Nrf2-regulated antioxidant enzymes superoxide dismutase 3 (SOD3) and NAD(P)H: quinone oxidoreductase 1 (NQO1) were quantified. The treatment with E2 suppressed, whereas VC and Res prevented E2-mediated decrease in the expression levels of SOD3, NQO1, Nrf2 mRNA, and protein in MCF-10A cells. The treatment with E2, Res, or VC significantly increased mRNA and protein expression levels of Nrf1. 17ß-Estradiol treatment significantly increased but VC or Res decreased Nrf3 mRNA and protein expression levels. Our studies demonstrate that estrogen-induced breast cancer might be prevented through upregulation of antioxidant enzymes via Nrf-dependent pathways.


Subject(s)
Antioxidants/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms , Estradiol/adverse effects , Estrogens/adverse effects , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Neoplasm Proteins/metabolism , Superoxide Dismutase/biosynthesis , Breast Neoplasms/chemically induced , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Line, Tumor , Estradiol/pharmacology , Estrogens/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans
6.
Carcinogenesis ; 35(8): 1872-80, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24894866

ABSTRACT

The importance of estrogens in the etiology of breast cancer is widely recognized. Estrogen-induced oxidative stress has been implicated in this carcinogenic process. Resveratrol (Res), a natural antioxidant phytoestrogen has chemopreventive effects against a variety of illnesses including cancer. The objective of the present study was to characterize the mechanism(s) of Res-mediated protection against estrogen-induced breast carcinogenesis. Female August Copenhagen Irish rats were treated with 17ß-estradiol (E2), Res and Res + E2 for 8 months. Cotreatment of rats with Res and E2 inhibited E2-mediated proliferative changes in mammary tissues and significantly increased tumor latency and reduced E2-induced breast tumor development. Resveratrol treatment alone or in combination with E2 significantly upregulated expression of nuclear factor erythroid 2-related factor 2 (NRF2) in mammary tissues. Expression of NRF2-regulated antioxidant genes NQO1, SOD3 and OGG1 that are involved in protection against oxidative DNA damage was increased in Res- and Res + E2-treated mammary tissues. Resveratrol also prevented E2-mediated inhibition of detoxification genes AOX1 and FMO1. Inhibition of E2-mediated alterations in NRF2 promoter methylation and expression of NRF2 targeting miR-93 after Res treatment indicated Res-mediated epigenetic regulation of NRF2 during E2-induced breast carcinogenesis. Resveratrol treatment also induced apoptosis and inhibited E2-mediated increase in DNA damage in mammary tissues. Increased apoptosis and decreased DNA damage, cell migration, colony and mammosphere formation in Res- and Res + E2-treated MCF-10A cells suggested a protective role of Res against E2-induced mammary carcinogenesis. Small-interfering RNA-mediated silencing of NRF2 inhibited Res-mediated preventive effects on the colony and mammosphere formation. Taken together, these results suggest that Res inhibits E2-induced breast carcinogenesis via induction of NRF2-mediated protective pathways.


Subject(s)
Cell Transformation, Neoplastic/pathology , Estrogens/toxicity , Mammary Neoplasms, Experimental/prevention & control , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , Antioxidants , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Female , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats, Inbred ACI , Rats, Inbred Strains , Real-Time Polymerase Chain Reaction , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
7.
BMC Cancer ; 13: 253, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23697596

ABSTRACT

BACKGROUND: Estrogen metabolism-mediated oxidative stress is suggested to play an important role in estrogen-induced breast carcinogenesis. We have earlier demonstrated that antioxidants, vitamin C (Vit C) and butylated hydroxyanisole (BHA) inhibit 17ß-estradiol (E2)-mediated oxidative stress and oxidative DNA damage, and breast carcinogenesis in female August Copenhagen Irish (ACI) rats. The objective of the present study was to characterize the mechanism by which above antioxidants prevent DNA damage during breast carcinogenesis. METHODS: Female ACI rats were treated with E2; Vit C; Vit C + E2; BHA; and BHA + E2 for up to 240 days. mRNA and protein levels of a DNA repair enzyme 8-Oxoguanine DNA glycosylase (OGG1) and a transcription factor NRF2 were quantified in the mammary and mammary tumor tissues of rats after treatment with E2 and compared with that of rats treated with antioxidants either alone or in combination with E2. RESULTS: The expression of OGG1 was suppressed in mammary tissues and in mammary tumors of rats treated with E2. Expression of NRF2 was also significantly suppressed in E2-treated mammary tissues and in mammary tumors. Vitamin C or BHA treatment prevented E2-mediated decrease in OGG1 and NRF2 levels in the mammary tissues. Chromatin immunoprecipitation analysis confirmed that antioxidant-mediated induction of OGG1 was through increased direct binding of NRF2 to the promoter region of OGG1. Studies using silencer RNA confirmed the role of OGG1 in inhibition of oxidative DNA damage. CONCLUSIONS: Our studies suggest that antioxidants Vit C and BHA provide protection against oxidative DNA damage and E2-induced mammary carcinogenesis, at least in part, through NRF2-mediated induction of OGG1.


Subject(s)
Antioxidants/pharmacology , DNA Damage , DNA Glycosylases/biosynthesis , Estrogens/toxicity , Mammary Neoplasms, Experimental/metabolism , NF-E2-Related Factor 2/biosynthesis , 8-Hydroxy-2'-Deoxyguanosine , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Damage/physiology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Deoxyguanosine/biosynthesis , Female , Humans , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , RNA Interference , Rats , Rats, Inbred ACI , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Up-Regulation
8.
Carcinogenesis ; 34(5): 1165-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23492819

ABSTRACT

MicroRNAs (miRNA) are small non-coding RNAs that regulate the expression of approximately 60% of all human genes and play important roles in disease processes. Recent studies have demonstrated a link between dysregulated expression of miRNAs and breast carcinogenesis. Long-term estrogen exposure is implicated in development of human breast cancers, yet underlying mechanisms remain elusive. We have recently demonstrated that antioxidant vitamin C (vit C) prevents estrogen-induced breast tumor development. In this study, we investigated the role of vit C in the regulation of microRNA-93 (miR-93) and its target gene(s) in a rat model of mammary carcinogenesis. Female August Copenhagen Irish (ACI) rats were treated with vit C in the presence or absence of 17ß-estradiol (E2) for 8 months. We demonstrate an increased expression of the miR-93 in E2-treated mammary tissues and in human breast cell lines and vit C treatment reverted E2-mediated increase in miR-93 levels. MiRNA target prediction programs suggest one of the target genes of miR-93 to be nuclear factor erythroid 2-related factor 2 (NRF2). In contrast with miR-93 expression, NRF2 protein expression was significantly decreased in E2-treated mammary tissues, mammary tumors, and in breast cancer cell lines, and its expression was significantly increased after vit C treatment. Ectopic expression of miR-93 decreased protein expression of NRF2 and NRF2-regulated genes. Furthermore, miR-93 decreased apoptosis, increased colony formation, mammosphere formation, cell migration and DNA damage in breast epithelial cells, whereas silencing of miR-93 in these cells inhibited these carcinogenic processes. Taken together, our findings suggest an oncogenic potential of miR-93 during E2-induced breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , MicroRNAs/genetics , NF-E2-Related Factor 2/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Ascorbic Acid/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Transformation, Neoplastic/metabolism , DNA Damage , Estradiol/pharmacology , Estrogens/adverse effects , Female , Humans , NF-E2-Related Factor 2/metabolism , Rats , Rats, Inbred ACI , Rats, Sprague-Dawley , Up-Regulation/drug effects
9.
Carcinogenesis ; 33(1): 156-63, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072621

ABSTRACT

Exact mechanisms underlying the initiation and progression of estrogen-related cancers are not clear. Literature, evidence and our studies strongly support the role of estrogen metabolism-mediated oxidative stress in estrogen-induced breast carcinogenesis. We have recently demonstrated that antioxidants vitamin C and butylated hydroxyanisole (BHA) or estrogen metabolism inhibitor α-naphthoflavone (ANF) inhibit 17ß-estradiol (E2)-induced mammary tumorigenesis in female ACI rats. The objective of the current study was to identify the mechanism of antioxidant-mediated protection against E2-induced DNA damage and mammary tumorigenesis. Female ACI rats were treated with E2 in the presence or absence of vitamin C or BHA or ANF for up to 240 days. Nuclear factor erythroid 2-related factor 2 (NRF2) and NAD(P)H-quinone oxidoreductase 1 (NQO1) were suppressed in E2-exposed mammary tissue and in mammary tumors after treatment of rats with E2 for 240 days. This suppression was overcome by co-treatment of rats with E2 and vitamin C or BHA. Time course studies indicate that NQO1 levels tend to increase after 4 months of E2 treatment but decrease on chronic exposure to E2 for 8 months. Vitamin C and BHA significantly increased NQO1 levels after 120 days. 8-Hydroxydeoxyguanosine (8-OHdG) levels were higher in E2-exposed mammary tissue and in mammary tumors compared with age-matched controls. Vitamin C or BHA treatment significantly decreased E2-mediated increase in 8-OHdG levels in the mammary tissue. In vitro studies using silencer RNA confirmed the role of NQO1 in prevention of oxidative DNA damage. Our studies further demonstrate that NQO1 upregulation by antioxidants is mediated through NRF2.


Subject(s)
Antioxidants/pharmacology , DNA Damage , Estradiol/toxicity , Mammary Neoplasms, Experimental/prevention & control , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Animals , Ascorbic Acid/pharmacology , Benzoflavones/pharmacology , Butylated Hydroxyanisole/pharmacology , Female , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/physiology , Organ Specificity , Oxidation-Reduction , Rats , Rats, Inbred ACI
10.
PLoS One ; 6(9): e25125, 2011.
Article in English | MEDLINE | ID: mdl-21966433

ABSTRACT

Epidemiological and experimental evidences strongly support the role of estrogens in breast tumor development. Both estrogen receptor (ER)-dependent and ER-independent mechanisms are implicated in estrogen-induced breast carcinogenesis. Tamoxifen, a selective estrogen receptor modulator is widely used as chemoprotectant in human breast cancer. It binds to ERs and interferes with normal binding of estrogen to ERs. In the present study, we examined the effect of long-term tamoxifen treatment in the prevention of estrogen-induced breast cancer. Female ACI rats were treated with 17ß-estradiol (E2), tamoxifen or with a combination of E2 and tamoxifen for eight months. Tissue levels of oxidative stress markers 8-iso-Prostane F(2α) (8-isoPGF(2α)), superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase, and oxidative DNA damage marker 8-hydroxydeoxyguanosine (8-OHdG) were quantified in the mammary tissues of all the treatment groups and compared with age-matched controls. Levels of tamoxifen metabolizing enzymes cytochrome P450s as well as estrogen responsive genes were also quantified. At necropsy, breast tumors were detected in 44% of rats co-treated with tamoxifen+E2. No tumors were detected in the sham or tamoxifen only treatment groups whereas in the E2 only treatment group, the tumor incidence was 82%. Co-treatment with tamoxifen decreased GPx and catalase levels; did not completely inhibit E2-mediated oxidative DNA damage and estrogen-responsive genes monoamine oxygenase B1 (MaoB1) and cell death inducing DFF45 like effector C (Cidec) but differentially affected the levels of tamoxifen metabolizing enzymes. In summary, our studies suggest that although tamoxifen treatment inhibits estrogen-induced breast tumor development and increases the latency of tumor development, it does not completely abrogate breast tumor development in a rat model of estrogen-induced breast cancer. The inability of tamoxifen to completely inhibit E2-induced breast carcinogenesis may be because of increased estrogen-mediated oxidant burden.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogens/adverse effects , Oxidative Stress/drug effects , Tamoxifen/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Animals , Breast Neoplasms/chemically induced , Catalase/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Estradiol/adverse effects , Female , Glutathione Peroxidase/metabolism , Isoprostanes/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Rats , Superoxide Dismutase/metabolism
11.
Toxicol Appl Pharmacol ; 247(2): 83-90, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20600213

ABSTRACT

Phytoestrogens are plant compounds that structurally mimic the endogenous estrogen 17beta-estradiol (E(2)). Despite intense investigation, the net effect of phytoestrogen exposure on the breast remains unclear. The objective of the current study was to examine the effects of quercetin on E(2)-induced breast cancer in vivo. Female ACI rats were given quercetin (2.5 g/kg food) for 8 months. Animals were monitored weekly for palpable tumors, and at the end of the experiment, rats were euthanized, breast tumor and different tissues excised so that they could be examined for histopathologic changes, estrogen metabolic activity and oxidant stress. Quercetin alone did not induce mammary tumors in female ACI rats. However, in rats implanted with E(2) pellets, co-exposure to quercetin did not protect rats from E(2)-induced breast tumor development with 100% of the animals developing breast tumors within 8 months of treatment. No changes in serum quercetin levels were observed in quercetin and quercetin+E(2)-treated groups at the end of the experiment. Tumor latency was significantly decreased among rats from the quercetin+E(2) group relative to those in the E(2) group. Catechol-O-methyltransferase (COMT) activity was significantly downregulated in quercetin-exposed mammary tissue. Analysis of 8-isoprostane F(2alpha) (8-iso-PGF(2alpha)) levels as a marker of oxidant stress showed that quercetin did not decrease E(2)-induced oxidant stress. These results indicate that quercetin (2.5 g/kg food) does not confer protection against breast cancer, does not inhibit E(2)-induced oxidant stress and may exacerbate breast carcinogenesis in E(2)-treated ACI rats. Inhibition of COMT activity by quercetin may expose breast cells chronically to E(2) and catechol estrogens. This would permit longer exposure times to the carcinogenic metabolites of E(2) and chronic exposure to oxidant stress as a result of metabolic redox cycling to estrogen metabolites, and thus quercetin may exacerbate E(2)-induced breast tumors in female ACI rats.


Subject(s)
Estrogens/toxicity , Mammary Neoplasms, Experimental/chemically induced , Phytoestrogens/toxicity , Quercetin/toxicity , Animals , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase Inhibitors , Dinoprost/analogs & derivatives , Dinoprost/biosynthesis , Drug Synergism , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Phytoestrogens/administration & dosage , Quercetin/administration & dosage , Quercetin/metabolism , Rats , Rats, Inbred ACI
SELECTION OF CITATIONS
SEARCH DETAIL