Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Chem Res Toxicol ; 23(8): 1374-83, 2010 Aug 16.
Article En | MEDLINE | ID: mdl-20540524

4-Hydroxyequilenin (4-OHEN) is a major phase I metabolite of the equine estrogens present in widely prescribed hormone replacement formulations. 4-OHEN is autoxidized to an electrophilic o-quinone that has been shown to redox cycle, generating ROS, and to covalently modify proteins and DNA and thus potentially to act as a chemical carcinogen. To establish the ability of 4-OHEN to act as a hormonal carcinogen at the estrogen receptor (ER), estrogen responsive gene expression and proliferation were studied in ER(+) breast cancer cells. Recruitment by 4-OHEN of ER to estrogen responsive elements (ERE) of DNA in MCF-7 cells was also studied and observed. 4-OHEN was a potent estrogen, with additional weak activity associated with binding to the arylhydrocarbon receptor (AhR). The potency of 4-OHEN toward classical ERalpha mediated activity was unexpected given the reported rapid autoxidation and trapping of the resultant quinone by GSH. Addition of thiols to cell cultures did not attenuate the estrogenic activity of 4-OHEN, and preformed thiol conjugates added to cell incubations only marginally reduced ERE-luciferase induction. On reaction of the 4OHEN-GSH conjugate with NADPH, 4-OHEN was observed to be regenerated at a rate dependent upon NADPH concentration, indicating that intracellular nonenzymatic and enzymatic regeneration of 4-OHEN accounts for the observed estrogenic activity of 4-OHEN. 4-OHEN is therefore capable of inducing chemical and hormonal pathways that may contribute to estrogen-dependent carcinogenesis, and trapping by cellular thiols does not provide a mechanism of termination of these pathways.


Equilenin/analogs & derivatives , Glutathione/metabolism , Animals , Cell Proliferation/drug effects , DNA, Neoplasm/drug effects , Equilenin/chemistry , Equilenin/metabolism , Equilenin/pharmacology , Glutathione/chemistry , Horses , Humans , Ligands , NADP/chemistry , NADP/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Tumor Cells, Cultured
2.
J Biol Chem ; 284(13): 8633-42, 2009 Mar 27.
Article En | MEDLINE | ID: mdl-19158089

Exposure to estrogens increases the risk of breast and endometrial cancer. It is proposed that the estrogen receptor (ER) may contribute to estrogen carcinogenesis by transduction of the hormonal signal and as a "Trojan horse" concentrating genotoxic estrogen metabolites in the nucleus to complex with DNA, enhancing DNA damage. 4-Hydroxyequilenin (4-OHEN), the major catechol metabolite of equine estrogens present in estrogen replacement formulations, autoxidizes to a redox-cycling quinone that has been shown to cause DNA damage. 4-OHEN was found to be an estrogen of nanomolar potency in cell culture using a luciferase reporter assay and, using a chromatin immunoprecipitation assay, was found to activate ERalpha binding to estrogen-responsive genes in MCF-7 cells. DNA damage was measured in cells by comparing ERalpha(+) versus ERalpha(-) cells and 4-OHEN versus menadione, a reactive oxygen species (ROS)-generating, but non-estrogenic, quinone. 4-OHEN selectively induced DNA damage in ERalpha(+) cells, whereas menadione-induced damage was not dependent on cellular ER status. The rate of 4-OHEN-induced DNA damage was significantly enhanced in ERalpha(+) cells, whereas ER status had no effect on the rate of menadione-induced damage. Imaging of ROS induced by 4-OHEN showed accumulation selective for the nucleus of ERalpha(+) cells within 5 min, whereas in ERalpha(-) or menadione-treated cells, no selectivity was observed. These data support ERalpha acting as a Trojan horse concentrating 4-OHEN in the nucleus to accelerate the rate of ROS generation and thereby amplify DNA damage. The Trojan horse mechanism may be of general importance beyond estrogen genotoxins.


Cell Nucleus/metabolism , DNA Damage/drug effects , Equilenin/analogs & derivatives , Estrogen Receptor alpha/metabolism , Estrogens, Catechol/pharmacology , Mutagens/pharmacology , Animals , Cell Line, Tumor , Equilenin/metabolism , Equilenin/pharmacology , Estrogens, Catechol/metabolism , Female , Horses , Humans , Mutagens/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species , Vitamin K 3/pharmacology , Vitamins/pharmacology , Xenopus laevis
3.
Adv Exp Med Biol ; 617: 601-7, 2008.
Article En | MEDLINE | ID: mdl-18497087

Oxidative metabolism of estrogens has been associated with genotoxicity. O-methylation of catechol estrogens is considered as a protective mechanism. 4-Methoxyequilenin (4-MeOEN) is the O-methylated product of 4-hydroxyequilenin (4-OHEN). 4-OHEN, the major catechol metabolite of the equine estrogens present in the most widely prescribed hormone replacement therapeutics, causes DNA damage via quinone formation. In this study, estrogen receptor (ERa) binding of 4-MeOEN was compared with estradiol (E2) and equilenin derivatives including 4-BrEN using computer modeling, estrogen response element (ERE)-luciferase induction in MCF-7 cells, and alkaline phosphatase (AP) induction in Ishikawa cells. 4-MeOEN induced AP and luciferase with nanomolar potency and displayed a similar profile of activity to E2. Molecular modeling indicated that MeOEN could be a ligand for ERa despite no binding being observed in the ERa competitive binding assay. Methylation of 4-OHEN may not represent a detoxification pathway, since 4-MeOEN is a full estrogen agonist with nanomolar potency.


Breast Neoplasms/drug therapy , Endometrial Neoplasms/drug therapy , Equilenin/analogs & derivatives , Transcriptional Activation/drug effects , Alkaline Phosphatase/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Computer Simulation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Equilenin/pharmacology , Estradiol/chemistry , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Horses , Humans , Luciferases/metabolism , Response Elements/genetics , Transcription, Genetic , Tumor Cells, Cultured
4.
Chem Res Toxicol ; 21(5): 1120-4, 2008 May.
Article En | MEDLINE | ID: mdl-18447394

Long-term hormone replacement therapy with equine estrogens is associated with a higher risk of breast, ovarian, and endometrial cancers. Reactive oxygen species generated through redox cycling of equine estrogen metabolites may damage cellular DNA. Such oxidative stress may be linked to the development of cancers in reproductive organs. Xeroderma pigmentosa complementation group C-knockout ( Xpc-KO) and wild-type mice were treated with equilenin (EN), and the formation of 7,8-dihydro-8-oxodeoxyguanosine (8-oxodG) was determined as a marker of typical oxidative DNA damage, using liquid chromatography electrospray tandem mass spectrometry. The level of hepatic 8-oxodG in wild-type mice treated with EN (5 or 50 mg/kg/day) was significantly increased by approximately 220% after 1 week, as compared with mice treated with vehicle. In the uterus also, the level of 8-oxodG was significantly increased by more than 150% after 2 weeks. Similar results were observed with Xpc-KO mice, indicating that Xpc does not significantly contribute to the repair of oxidative damage. Oxidative DNA damage generated by equine estrogens may be involved in equine estrogen carcinogenesis.


DNA Damage/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Estrogens/pharmacology , Horses , Animals , DNA-Binding Proteins/genetics , Equilenin/analogs & derivatives , Equilenin/chemistry , Equilenin/pharmacology , Female , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , Molecular Structure , Oxidation-Reduction
5.
Endocrinology ; 148(10): 4793-802, 2007 Oct.
Article En | MEDLINE | ID: mdl-17584965

4-Methoxyequilenin (4-MeOEN) is an O-methylated metabolite in equine estrogen metabolism. O-methylation of catechol estrogens is considered as a protective mechanism; however, comparison of the properties of 4-MeOEN with estradiol (E(2)) in human breast cancer cells showed that 4-MeOEN is a proliferative, estrogenic agent that may contribute to carcinogenesis. 4-MeOEN results from O-methylation of 4-hydroxyequilenin, a major catechol metabolite of the equine estrogens present in hormone replacement therapeutics, which causes DNA damage via quinone formation, raising the possibility of synergistic hormonal and chemical carcinogenesis. 4-MeOEN induced cell proliferation with nanomolar potency and induced estrogen response element (ERE)-mediated gene transcription of an ERE-luciferase reporter and the endogenous estrogen-responsive genes pS2 and TGF-alpha. These estrogenic actions were blocked by the antiestrogen ICI 182,780. In the standard radioligand estrogen receptor (ER) binding assay, 4-MeOEN showed very weak binding. To test for alternate ligand-ER-independent mechanisms, the possibility of aryl hydrocarbon receptor (AhR) binding and ER-AhR cross talk was examined using a xenobiotic response element-luciferase reporter and using AhR small interfering RNA silencing in the ERE-luciferase reporter assay. The results negated the possibility of AhR-mediated estrogenic activity. Comparison of gene transcription time course, ER degradation, and rapid activation of MAPK/ERK in MCF-7 cells demonstrated that the actions of 4-MeOEN mirrored those of E(2) with potency for classical and nonclassical estrogenic pathways bracketing that of E(2). Methylation of 4-OHEN may not represent a detoxification pathway because 4-MeOEN is a full, potent estrogen agonist.


Breast Neoplasms/genetics , Equilenin/analogs & derivatives , Receptors, Estrogen/metabolism , Transcriptional Activation/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Equilenin/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation , Genes, Reporter , Humans , Ligands , Luciferases/genetics , Receptors, Aryl Hydrocarbon/metabolism , Response Elements/genetics , Transcription, Genetic , Xenobiotics/metabolism
6.
Eur J Med Chem ; 41(7): 891-5, 2006 Jul.
Article En | MEDLINE | ID: mdl-16730395

Syntheses of novel heterocyclic derivatives of 18-nor-equilenin, namely, (12H-11-oxa-17-thia-15-aza-cyclopenta[a]phenanthrene-16-yl)-hydrazine (4a/b) and its fused [1,2,4]triazolo derivatives6H-5-oxa-7-thia-8,9,10a-triaza-pentaleno[4,5-a]phenanthrene (5a/b), 10-methyl-6H-5-oxa-7-thia-8,9,10a-triaza-pentaleno[4,5-a]phenanthrene (6a/b) and tetrazolo derivatives 1-substituted-6H-5-oxa-7-thia-8,9,10,10a-tetraaza-pentaleno[4,5-a]phenanthrene (7a/b) along with their antibacterial activities are reported.


Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Equilenin/chemical synthesis , Equilenin/pharmacology , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/pharmacology , Anti-Bacterial Agents/chemistry , Bacillus subtilis/drug effects , Equilenin/chemistry , Escherichia coli/drug effects , Gram-Negative Bacteria/drug effects , Heterocyclic Compounds, 4 or More Rings/chemistry , Molecular Structure
7.
Chem Biol Interact ; 161(3): 271-8, 2006 Jul 10.
Article En | MEDLINE | ID: mdl-16730688

Long-term exposure to synthetic and endogenous estrogens has been associated with the development of cancer in several tissues. One potential mechanism of estrogen carcinogenesis involves catechol formation and these catechols are further oxidized to electrophilic/redox active o-quinones, which have the potential to both initiate and promote the carcinogenic process. 4-Hydroxyequilenin (4-OHEN), a major phase I metabolite of several estrogens present in Premarin, is considerably more cytotoxic, carcinogenic, and mutagenic as compared to the catechol estrogen metabolites of endogenous estrogens. Previously, we showed that 4-OHEN autoxidized to an o-quinone and caused a variety of damage to DNA. Allowing more time between the induction of DNA damage and the entry of a damaged cell into the DNA synthetic phase of the cell cycle protects that cell from mutagenesis. Central to this response is the establishment of a G1 checkpoint. This checkpoint is mediated by the cyclin-dependent kinase inhibitor p21WAF1, a direct downstream target for transcriptional activation by p53. In this study, we investigated this signaling pathway. Surprisingly, exposure of the human MCF-10A immortalized nontransformed mammary epithelial cell line to 4-OHEN did not induce a p53-induced G1 arrest. A 24 h treatment with 4-OHEN significantly induced p53 and p21WAF1 protein expression at 10 and 20 microM, as well as significantly induced the transactivation of a p53-luciferase reporter gene at 20 microM. Significant decreases in cell proliferation were also observed with concentrations of 5 microM and higher of 4-OHEN. However, 4-OHEN did not induce a G1 checkpoint and cells with damaged DNA accumulated in the S phase. This S phase delay could be beneficial for the survival of the damaged cells which could contribute to the carcinogenic process.


DNA Damage/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Equilenin/analogs & derivatives , G1 Phase/drug effects , Mammary Glands, Human/cytology , Tumor Suppressor Protein p53/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/cytology , Equilenin/pharmacology , Humans , Phosphoserine/metabolism
8.
J Steroid Biochem Mol Biol ; 98(1): 48-55, 2006 Jan.
Article En | MEDLINE | ID: mdl-16191477

Equilenin is one of 10 kinds of estrogens that are found in pregnant mares' urine. It has been used extensively for estrogen replacement therapy in postmenopausal women. Typical inducers of the cytochrome P4501A1 (CYP1A1), such as TCDD, benzo(a)pyrene (B(a)P) and 3-methylcholanthrene, have a planar molecular structure in common and bind to the aryl hydrocarbon receptor (AhR). The structure of equilenin differs from classic estrogens by the presence of two additional double bonds in ring B of the steroid nucleus, and it is planar. This structural similarity of equilenin to the typical AhR agonist prompted us to investigate the capability of equilenin to induce CYP1A1 expression. Administration of equilenin to two mouse strains (C57BL and DBA) that exhibit different degrees of responsiveness to an Ah-receptor agonist and showed that equilenin was capable of dose-dependently increasing both the ethoxyresorufin O-deethylase activity and CYP1a proteins in both strains of mice. Equilenin also induced CYP1A1 mRNA in treated HepG2 cell lines and transcriptional activity in an XRE-directed luciferase reporter gene. Competitive binding studies using C57BL AhR indicated equilenin weakly displaced (3)H-B(a)P from AhR. Together, these data show that equilenin, an equine steroid hormone, served as an AhR ligand in the present study.


Carcinoma, Hepatocellular/drug therapy , Cytochrome P-450 CYP1A1/biosynthesis , Equilenin/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Binding, Competitive , Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP1A1/genetics , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic , Horses , Humans , Kidney/drug effects , Kidney/enzymology , Ligands , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Transcriptional Activation/drug effects
9.
Cancer Lett ; 230(1): 81-9, 2005 Dec 08.
Article En | MEDLINE | ID: mdl-16253764

Catechol O-methyltransferase (COMT) plays an important role in the inactivation of biologically active and toxic catechols. It has been shown that human soluble COMT (S-COMT) is genetically polymorphic with a wild type and at least one variant in which a valine has been substituted with a methionine at codon 108. This polymorphism has been the subject of intense molecular epidemiological studies because of the important role of COMT in the metabolism of catecholamines and catechol estrogens. Several epidemiological studies have shown that women, homozygous with the Val108Met variant, have an increased risk of developing estrogen-associated cancers. However, some other studies have shown that this COMT polymorphism is not associated with increased risk of developing cancers. These conflicting data suggest that additional COMT genetic variants might contribute to the increased risk of developing cancers. Although two new single nucleotide polymorphisms (SNP) that cause amino acid substitutions Ala22Ser and Ala52Thr have been identified recently, they have not been fully characterized. In the present study, Ala22Ser and Ala52Thr variants of human S-COMT were produced using recombinant DNA techniques, and then COMT properties were measured including enzymatic activity, thermostability, and sensitivity to inhibition mediated by 4-hydroxyequilenin (4-OHEN). The Ala22Ser variant showed lower methylation capacity and higher thermolability. In addition, this variant is sensitive to 4-OHEN mediated irreversible inhibition. Our data indicate that the Ala22Ser polymorphism might also be of functional significance and might play a role in susceptibility to estrogen-associated cancers.


Breast Neoplasms/chemically induced , Breast Neoplasms/genetics , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Equilenin/analogs & derivatives , Polymorphism, Genetic , Alanine , DNA Methylation , Equilenin/pharmacology , Escherichia coli/genetics , Estrogen Replacement Therapy/adverse effects , Female , Humans , Mutagenesis, Site-Directed , Plasmids , Serine , Temperature
10.
Mutat Res ; 550(1-2): 109-21, 2004 Jun 04.
Article En | MEDLINE | ID: mdl-15135645

Long-term exposure to synthetic and endogenous estrogens has been associated with the development of cancer in several tissues. One potential mechanism of estrogen carcinogenesis involves catechol formation and these catechols are further oxidized to electrophilic/redox active o-quinones, which have the potential to both initiate and promote the carcinogenic process. Previously we showed that 4-hydroxyequilenin (4-OHEN) autoxidized to an o-quinone and caused a variety of damage to DNA. Since these deleterious effects could contribute to gene mutations, we investigated the Chinese hamster V79 cells to ascertain the relative ability of estradiol, 4-hydroxyestradiol, 17beta-hydroxyequilenin, 4,17beta-hydroxyequilenin, estrone, 4-hydroxyestrone, equilenin, and 4-hydroxyequilenin to induce the mutation of the hypoxanthine-guanine phosphoribosyltransferase (hprt) gene. All the 4-hydroxylated catechols induced significantly more colony formations in V79 cells as compared to the parent phenols at 100nM, suggesting that the catechol estrogen metabolites are more mutagenic towards the hprt gene than estrogens. Since 4-OHEN induced the highest mutation frequency, we examined a biomarker for transformation potential of this compound in MCF-10A cells using an anchorage-independent growth assay. Although 4-OHEN induced anchorage-independent growth of these cells, the isolated clones were not able to grow as tumors in vivo when injected into nude mice. These cells were assayed for genetic changes using cDNA microarrays. Real time RT-PCR confirmation of some of the differentially expressed genes showed down-regulation of metallothionein 2A, p53, BRCA1, and c-myc. Moreover, we showed the involvement of other genes important in cell transformation and oxidative stress, strengthening the hypothesis that this mechanism plays a considerable role in 4-OHEN-induced anchorage-independent growth.


Equilenin/analogs & derivatives , Equilenin/pharmacology , Estradiol Congeners/pharmacology , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Animals , Cell Adhesion , Cell Division , Cell Line , Cell Line, Tumor , Cricetinae , DNA Mutational Analysis , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Horses , Male , Metallothionein/metabolism , Models, Chemical , Mutation , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Oxidative Stress , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
Chem Res Toxicol ; 17(4): 512-20, 2004 Apr.
Article En | MEDLINE | ID: mdl-15089093

Catechol-O-methyltransferase (COMT) plays an important role in the inactivation of biologically active and toxic catechols. It has been shown that COMT is genetically polymorphic with a wild-type and variant form where a valine has been substituted with a methionine. Several, but not all, epidemiological studies have shown that women, homozygous with the variant form, have an increased risk of developing breast cancer. Previously, we showed that 4-hydroxyequilenin (4-OHEN), a cytotoxic/genotoxic equine catechol estrogen metabolite, is both a substrate of COMT and an irreversible inhibitor of the methylation activity of COMT in vitro. To further understand the mechanism(s) of the association between the breast cancer risk and the COMT polymorphism, it was of interest to study the effect of the Val/Met polymorphism on COMT-catalyzed catechol estrogen methylation and 4-OHEN-mediated inhibition. In the present study, Michaelis-Menten analysis showed no difference between the relative ability of each form to methylate 4-OHEN. However, we found that the COMT variant form was more susceptible to 4-OHEN-mediated irreversible inactivation. Electrospray ionization mass spectrometry and SDS-gel analysis of COMT modified by 4-OHEN revealed that inhibition mechanisms include alkylation and/or oxidation of certain amino acids. In addition, site-directed mutagenesis experiments showed that Cys33 played a more important role in the variant form of COMT demonstrated by the fact that the C33A mutant of the variant form of COMT decreased its catalytic capability more dramatically as compared with that of wild type. Furthermore, thermotropic studies indicated that the variant form was more thermolabile, which suggested that the valine to methionine substitution may have changed the secondary/tertiary structure of the variant form of COMT, making it more susceptible to 4-OHEN and heat inactivation. These data suggest that 4-OHEN-mediated inhibition of the variant form of COMT in vivo might affect the detoxification efficiency of endogenous and/or exogenous catechol estrogens and play a role in the association between breast cancer risk and COMT polymorphism.


Breast Neoplasms/etiology , Breast Neoplasms/genetics , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/pharmacology , Equilenin/analogs & derivatives , Equilenin/pharmacology , Equilenin/toxicity , Catechol O-Methyltransferase Inhibitors , Estradiol Congeners , Female , Humans , Methylation , Risk Factors , Temperature
12.
J Soc Gynecol Investig ; 10(5): 302-8, 2003 Jul.
Article En | MEDLINE | ID: mdl-12853093

OBJECTIVE: In the present study, neuronal PC12 cells and hippocampal HT22 cells maintained in culture were used to test the neuroprotective effect of equine estrogens estrone, 17beta-estradiol, 17alpha-estradiol, equilin, 17beta-dihydroequilin, 17alpha-dihydroequilin, equilenin, 17beta-dihydroequilenin, 17alpha-dihydroequilenin, Delta(8)-estrone(,) and Delta(8),17beta-estradiol against glutamate toxicity. METHODS: The HT22 and PC12 cells were grown in Dulbecco modified Eagle medium supplemented with 5% horse serum, 10% fetal bovine serum, and 10 mM HEPES. The undifferentiated PC12 cells were plated on collagen-coated, 96-well plastic plates at 10,000 cells per well, and the HT22 cells were plated on uncoated 96-well plates at 2500 cells per well. Twenty-four hours after plating, various concentrations of estrogens (0.1-40 microM) and glutamate (1-10 mM) were added in a total volume of 100 microL. After 24 hours, cell viability was determined using the MTS cell proliferation assay. Results were verified in some experiments by using the lactate dehydrogenase cytotoxicity assay. RESULTS: The results indicate that cell toxicity in both cell lines was directly proportional to the concentration of glutamate. The lowest dose of glutamate that reduced cell viability by 50% under these conditions was 1.8 mM for HT22 cells and 3 mM for PC12 cells. All estrogens tested were neuroprotective against glutamate-induced cell death in a typical dose-related manner. However, these estrogens differed extensively with respect to their neuroprotective potencies. In both cell lines, the Delta(8)-ring B unsaturated estrogens were the most neuroprotective, whereas the classic estrogens 17beta-estradiol, estrone, and 17alpha-estradiol were the least potent. The order of potency was Delta(8),17beta-estradiol > Delta(8)-estrone > 17beta-dihydroequilenin > 17alpha-dihydroequilenin > equilenin > 17beta-dihydroequilin = equilin > 17alpha-dihydroequilin > 17beta-estradiol > estrone > 17alpha-estradiol in PC12 cells and Delta(8),17beta-estradiol > Delta(8)-estrone > equilenin = 17beta-dihydroequilenin > 17beta-dihydroequilin > equilin > 17alpha-dihydroequilenin > 17alpha-dihydroequilin > 17alpha-estradiol = 17beta-estradiol > estrone in HT22 cells. CONCLUSIONS: Our data indicate that the neurotoxic effects of glutamate can be inhibited differentially by various equine estrogens. The less estrogenic (uterotropic) Delta(8) estrogens were the most effective neuroprotectors, and further chemical modifications of these estrogens may provide compounds that are useful for preventing neurodegenerative diseases in both women and men.


Equilin/analogs & derivatives , Estradiol Congeners/pharmacology , Glutamic Acid/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Equilenin/pharmacology , Equilin/pharmacology , Estradiol/pharmacology , Estradiol Congeners/chemistry , Estrone/pharmacology , Hippocampus , PC12 Cells , Rats , Structure-Activity Relationship
13.
Chem Res Toxicol ; 15(7): 935-42, 2002 Jul.
Article En | MEDLINE | ID: mdl-12119004

Glutathione S-transferases (GSTs) are a family of detoxification isozymes that protect cells by conjugating GSH to a variety of toxic compounds, and they may also play a role in the regulation of both cellular proliferation and apoptosis. We have previously shown that human GST P1-1, which is the most widely distributed extrahepatic isozyme, could be inactivated by the catechol estrogen metabolite 4-hydroxyequilenin (4-OHEN) in vitro [Chang, M., Shin, Y. G., van Breemen, R. B., Blond, S. Y., and Bolton, J. L. (2001) Biochemistry 40, 4811-4820]. In the present study, we found that 4-OHEN and another catechol estrogen, 4,17beta-hydroxyequilenin (4,17beta-OHEN), significantly decreased GSH levels and the activity of GST within minutes in both estrogen receptor (ER) negative (MDA-MB-231) and ER positive (S30) human breast cancer cells. In addition, 4-OHEN caused significant decreases in GST activity in nontransformed human breast epithelial cells (MCF-10A) but not in the human hepatoma HepG2 cells, which lack GST P1-1. We also showed that GSH partially protected the inactivation of GST P1-1 by 4-OHEN in vitro, and depletion of cellular GSH enhanced the 4-OHEN-induced inhibition of GST activity. In addition, 4-OHEN GSH conjugates contributed about 27% of the inactivation of GST P1-1 by 4-OEHN in vitro. Our in vitro kinetic inhibition experiments with 4-OHEN showed that GST P1-1 had a lower K(i) value (20.8 microM) compared to glyceraldehyde-3-phosphate dehydrogenase (GAPDH, 52.4 microM), P450 reductase (PR, 77.4 microM), pyruvate kinase (PK, 159 microM), glutathione reductase (GR, 230 microM), superoxide dismutase (SOD, 448 microM), catalase (562 microM), GST M1-1 (620 microM), thioredoxin reductase (TR, 694 microM), and glutathione peroxidase (GPX, 1410 microM). In contrast to the significant inhibition of total GST activity in these human breast cancer cells, 4-OHEN only slightly inhibited the cellular GAPDH activity, and other cellular enzymes including PR, PK, GR, SOD, catalase, TR, and GPX were resistant to 4-OHEN-induced inhibition. These data suggest that GST P1-1 may be a preferred protein target for equine catechol estrogens in vivo.


Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Equilenin/analogs & derivatives , Equilenin/pharmacology , Estrogens, Catechol/pharmacology , Glutathione Transferase/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Animals , Breast Neoplasms/pathology , Glutathione S-Transferase pi , Horses , Humans , Kinetics , Substrate Specificity , Tumor Cells, Cultured
14.
J Lipid Res ; 42(11): 1789-800, 2001 Nov.
Article En | MEDLINE | ID: mdl-11714848

Estrogen replacement therapies, such as conjugated equine estrogen (CEE, Premarin), reduce the risk of coronary heart disease among postmenopausal women. In the present study, a HepG2 stable cell line (HepG2/S) that harbors a luciferase reporter gene cassette with the human apolipoprotein A-I (apoA-I) promoter region was used to examine the activity of CEE components in modulating human apoA-I promoter activity. A number of estrogens modulated apoA-I promoter activity, with equilenin (Eqn) being the most potent. Eqn produced a 3-fold increase in apoA-I promoter activity and a similar increase in apoA-I mRNA without affecting its degradation rate. Nuclear runoff assays indicated that the transcription rate of the apoA-I gene was increased 2.5-fold in Eqn-treated cells. When HepG2/S cells were exposed to Eqn, apoA-I protein secretion increased by 80%, whereas the level of secreted apoA-II remained unchanged. Transient transfection studies with human apoA-I promoter constructs derived from pGL3-luciferase reporter plasmid were used to identify the cis-acting element involved in Eqn-mediated induction. The results demonstrated that the apoA-I electrophile/antioxidant response element (EpRE/ARE) might be responsible for the increase in apoA-I promoter activity by Eqn. Cotransfection experiments using estrogen receptor (ERalpha and/or ERbeta) expression vectors have indicated that neither receptor can potentiate the Eqn-mediated induction of apoA-I promoter activity. In addition, mobility shift analysis using antibody against either ERalpha or ERbeta cannot detect the presence of these receptors in the DNA-protein complex. The data indicate that Eqn can induce the promoter activity of the human apoA-I gene, leading to an increase in apoA-I mRNA levels and apoA-I protein secretion through an ER-independent pathway involving apoA-I EpRE/ARE.


Apolipoprotein A-I/genetics , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Animals , Apolipoproteins/biosynthesis , Apolipoproteins/metabolism , Carcinoma, Hepatocellular , Equilenin/pharmacology , Estrogen Antagonists/pharmacology , Horses , Humans , Kinetics , Liver Neoplasms , Luciferases/genetics , Luciferases/metabolism , Promoter Regions, Genetic , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology , Response Elements , Transfection , Tumor Cells, Cultured
15.
Biochemistry ; 40(15): 4811-20, 2001 Apr 17.
Article En | MEDLINE | ID: mdl-11294649

The inactivation mechanism(s) of human glutathione S-transferase P1-1 (hGST P1-1) by the catechol metabolite of Premarin estrogens, 4-hydroxyequilenin (4-OHEN), was (were) studied by means of site-directed mutagenesis, electrospray ionization mass spectrometric analysis, titration of free thiol groups, kinetic studies of irreversible inhibition, and analysis of band patterns on nonreducing sodium dodecyl sulfate--polyacrylamide gel electrophoresis (SDS-PAGE). The four cysteines (Cys 14, Cys 47, Cys 101, and Cys 169 in the primary sequence) in hGST P1-1 are susceptible to electrophilic attack and/or oxidative damage leading to loss of enzymatic activity. To investigate the role of cysteine residues in the 4-OHEN-mediated inactivation of this enzyme, one or a combination of cysteine residues was replaced by alanine residues (C47A, C101A, C47A/C101A, C14A/C47A/C101A, and C47A/C101A/C169A mutants). Mutation of Cys 47 decreased the affinity for the substrate GSH but not for the cosubstrate 1-chloro-2,4-dinitrobenzene (CDNB). However, the Cys 47 mutation did not significantly affect the rate of catalysis since V(max) values of the mutants were similar or higher compared to that of wild type. Electrospray ionization mass spectrometric analyses of wild-type and mutant enzymes treated with 4-OHEN showed that a single molecule of 4-OHEN-o-quinone attached to the proteins, with the exception of the C14A/C47A/C101A mutant where no covalent adduct was detected. 4-OHEN also caused oxidative damage as demonstrated by the appearance of disulfide-bonded species on nonreducing SDS--PAGE and protection of 4-OHEN-mediated enzyme inhibition by free radical scavengers. The studies of thiol group titration and irreversible kinetic experiments indicated that the different cysteines have distinct reactivity for 4-OHEN; Cys 47 was the most reactive thiol group whereas Cys 169 was resistant to modification. These results demonstrate that hGST P1-1 is inactivated by 4-OHEN through two possible mechanisms: (1) covalent modification of cysteine residues and (2) oxidative damage leading to proteins inactivated by disulfide bond formation.


Enzyme Inhibitors/pharmacology , Equilenin/analogs & derivatives , Equilenin/pharmacology , Estrogens, Catechol/metabolism , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Amino Acid Substitution/genetics , Animals , Cysteine/genetics , Disulfides/metabolism , Dithionitrobenzoic Acid/metabolism , Electron Spin Resonance Spectroscopy , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Equilenin/metabolism , Free Radical Scavengers/pharmacology , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Histidine/genetics , Horses , Humans , Isoenzymes/genetics , Kinetics , Mutagenesis, Site-Directed , Oxidation-Reduction , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Reducing Agents/metabolism , Structure-Activity Relationship , Sulfhydryl Compounds/metabolism , Sulfhydryl Reagents/metabolism , Titrimetry
16.
Mol Cell Endocrinol ; 150(1-2): 33-7, 1999 Apr 25.
Article En | MEDLINE | ID: mdl-10411297

A large proportion of the beneficial effects that oestrogens demonstrate on the vasculature are believed to be mediated via direct effects on the vascular wall. In this study we compared a number of oestrogenic compounds isolated from pregnant mare's urine including 17beta-oestradiol and oestrone, in terms of their abilities to inhibit stimulated endothelin-1 release from normal human coronary artery endothelial cells (CAEC). We also examined their ability to stimulate expression of constitutive endothelial nitric oxide synthase (eNOS) and explored their effects on cellular angiotensin converting enzyme (ACE). All the oestrogens tested were able to inhibit serum-stimulated ET-1 release. Oestrone and 17alpha-dihydroequilenin failed to significantly affect cellular eNOS levels. 17Beta-oestradiol and oestrone significantly increased cellular ACE levels while 17beta,delta(8,9)-dehydroestradiol decreased cellular ACE. We discuss these observations in terms of their potential clinical relevance and use as a means of screening novel oestrogen-like compounds.


Endothelin-1/metabolism , Endothelium, Vascular/physiology , Estradiol/pharmacology , Estrone/pharmacology , Nitric Oxide Synthase/biosynthesis , Peptidyl-Dipeptidase A/biosynthesis , Animals , Cells, Cultured , Coronary Vessels/physiology , Equilenin/analogs & derivatives , Equilenin/pharmacology , Female , Humans , Nitric Oxide Synthase Type III , Pregnancy
17.
Chem Res Toxicol ; 12(2): 204-13, 1999 Feb.
Article En | MEDLINE | ID: mdl-10027800

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.


Equilenin/analogs & derivatives , Equilin/analogs & derivatives , Estradiol Congeners/metabolism , Quinones/metabolism , Quinones/pharmacology , Animals , Breast Neoplasms/drug therapy , Cytochrome P-450 Enzyme System/metabolism , Equilenin/metabolism , Equilenin/pharmacology , Equilin/metabolism , Equilin/pharmacology , Estradiol Congeners/pharmacology , Estrone/pharmacology , Female , Humans , Microsomes, Liver/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Stereoisomerism , Tumor Cells, Cultured
18.
Chem Res Toxicol ; 11(7): 758-65, 1998 Jul.
Article En | MEDLINE | ID: mdl-9671538

The risk factors for women developing breast and endometrium cancers are all associated with a lifetime of estrogen exposure. Estrogen replacement therapy (ERT) in particular has been correlated with a slight increased cancer risk, although the numerous benefits of ERT may negate this harmful side effect. Equilenin and equilin are equine estrogens which make up between 30% and 45% of the most widely prescribed estrogen replacement formulation, Premarin (Wyeth-Ayerst). In this study we have synthesized the catechol metabolites of equilenin [4-hydroxyequilenin (4-OHEN)] and equilin [4-hydroxyequilin (4-OHEQ)] and examined how changing unsaturation in the B ring affects the formation of o-quinone GSH conjugates and the ability of the o-quinones and/or GSH conjugates to inhibit glutathione S-transferase (GST). Interestingly, both 4-OHEN and 4-OHEQ autoxidized to o-quinones without the need of oxidative enzyme catalysis. 4-OHEN-o-quinone reacts with GSH to give two mono-GSH conjugates and one diadduct. The behavior of 4-OHEQ was found to be more complex than 4-OHEN as conjugates resulting from 4-OHEN were detected in addition to the 4-OHEQ GSH adducts. Both 4-OHEN and 4-OHEQ were found to be potent inhibitors of GST-catalyzed conjugation of GSH with 1-chloro-2,4-dinitrobenzene. In contrast, the endogenous catechol estrogens, 4-hydroxyestrone (4-OHE) and 2-hydroxyestrone (2-OHE), were without effect unless tyrosinase was present to convert the catechols to o-quinones. Scavengers of reactive oxygen species and metal chelators had no effect on GST inhibition by catechol estrogens with the exception of the catalase which protected GST activity. Kinetic studies showed that 4-OHEN was a potent irreversible inactivator of GST. Preincubation of the enzyme with 4-OHEN showed a time-dependent increase in inhibitory effect, and gel filtration did not restore GST activity confirming the irreversible nature of the enzyme inactivation. Analysis of the Kitz-Wilson plot gave a dissociation constant of the reversible enzyme-inhibitor complex (Ki = 620 microM) and a rate constant of conversion of the reversible enzyme-inhibitor complex to the irreversibly inhibited enzyme (k2 = 7.3 x 10(-)3 s-1). These data suggest that 4-OHEN is an irreversible inactivator with relatively low affinity for GST; however, once formed the 4-OHEN enzyme complex is rapidly converted to the irreversibly inhibited enzyme. The inhibition mechanism likely involves oxidation of the catechol estrogens to o-quinones and covalent modification and/or oxidation of critical amino acid residues on GST. In addition, hydrogen peroxide generated through redox cycling of the o-quinone and/or semiquinone radical and GSH could cause oxidative damage to GST.


Equilenin/analogs & derivatives , Equilin/analogs & derivatives , Estradiol Congeners/pharmacology , Glutathione Transferase/antagonists & inhibitors , Animals , Biotransformation , Catechols/metabolism , Chromatography, Gel , Chromatography, High Pressure Liquid , Dinitrochlorobenzene , Equilenin/pharmacokinetics , Equilenin/pharmacology , Equilin/pharmacokinetics , Equilin/pharmacology , Estradiol Congeners/pharmacokinetics , Glutathione/metabolism , Glutathione Transferase/metabolism , Horses , Kinetics , Oxidation-Reduction , Reactive Oxygen Species , Spectrophotometry, Ultraviolet
19.
Chem Res Toxicol ; 11(2): 94-101, 1998 Feb.
Article En | MEDLINE | ID: mdl-9511900

Premarin (Wyeth-Ayerst) is the estrogen replacement treatment of choice and continues to be one of the most widely dispensed prescriptions in the United States. In addition to endogenous estrogens, Premarin contains unsaturated estrogens including equilenin. We synthesized the catechol metabolite of equilenin, 4-hydroxyequilenin (4-OHEN), and found that the semiquinone radical of 4-OHEN reacted with 2'-deoxynucleosides generating very unusual adducts. 2'-Deoxyguanosine (dG), 2'-deoxyadenosine (dA), or 2'-deoxycytosine (dC) all gave four isomers, but no product was observed for thymidine under similar physiological conditions. The structures of these adducts were determined by electrospray mass spectrometry and NMR experiments including 1H, 13C, DQF-COSY, ROESY, HOHAHA, HMQC, and HMBC. The spectral data show that dG forms a cyclic adduct with the 4-OHEN producing 2-N1,3-N2-deoxyguanosyl-1,3-dihydroxy-5,7,9(10)-estratriene-4,17-d ione. Similarly, reaction with dA produced 1-N6,3-C2-deoxyadenosyl-2,3-dihydroxy-5,7,9(10)-estratriene-4,17-d ione, and incubations with dC resulted in 1-N3,3-N4-deoxycytosyl-2,3-dihydroxy-5,7,9(10)-estratriene-4,17-di one. We found that care needed to be taken during the isolation of the dA adducts in particular, as any exposure to acidic environments caused hydrolysis of the sugar moiety leaving alkylated adenine. In mixtures of the deoxynucleosides treated with 4-OHEN, reaction occurred primarily with dG followed by dC and dA. With DNA significant apurinic sites were produced as 4-OHEN-adenine adducts were detected in the ethanol wash prior to hydrolysis. When the DNA was hydrolyzed to deoxynucleosides and analyzed by electrospray mass spectrometry, only one isomer of 4-OHEN-dG and one isomer of 4-OHEN-dC were observed. Our data suggest that several different types of DNA lesions could be expected from 4-OHEN including apurinic sites and bulky stable adducts, in addition to the published oxidized damage to DNA caused by 4-OHEN. The production of these semiquinone radical-derived DNA adducts could play a role in the carcinogenic effects of Premarin estrogens.


DNA Adducts/biosynthesis , DNA Damage/drug effects , Equilenin/analogs & derivatives , Estrogens, Conjugated (USP)/pharmacology , Alkylation , Carcinogens/metabolism , Cytosine/analogs & derivatives , Cytosine/biosynthesis , DNA/chemistry , DNA Adducts/drug effects , Deoxyadenosines/biosynthesis , Deoxyguanosine/biosynthesis , Equilenin/pharmacology , Free Radicals/adverse effects , Humans , Oxidative Stress
20.
Carcinogenesis ; 16(10): 2571-4, 1995 Oct.
Article En | MEDLINE | ID: mdl-7586168

Free radical generation by metabolic redox cycling between catechol estrogens and their quinones and subsequent hydroxyl radical damage to DNA have been proposed to mediate estrogen-induced renal carcinogenesis in the hamster. In this study the content of 8-hydroxy-2'-deoxy-guanosine (8-OHdG), a marker product of hydroxyl radical action, was examined in DNA incubated with a liver microsomal activating system and with catechol estrogens, equilenin-3,4-quinone or with parent estrogens. Equilenin-3,4-quinone increased the formation of 8-OHdG by 50% over control levels. 4-Hydroxyestrone and 4-hydroxy-estradiol raised 8-OHdG contents significantly, to 1.61 +/- 0.79 and 1.27 +/- 0.31 8-OHdG/10(5) deoxyguanosine (dG) respectively over controls (0.68 +/- 0.25 8-OHdG/10(5) dG). The corresponding 2-hydroxylated estrogens and the parent hormones estrone, estradiol and equilenin did not affect 8-hydroxylation of guanine bases of DNA. In incubations of catechol estrogens with microsomes and cumene hydroperoxide the 4-hydroxyestrogens were oxidized to quinones more rapidly than the 2-hydroxyestrogens. Our data support a mechanism of hydroxyl radical generation from estrogens by redox cycling between 4-hydroxylated metabolites and their quinones. The rapid oxidation of 4-hydroxylated estrogens to quinones, their redox cycling and hydroxyl radical damage to DNA is consistent with the previously reported carcinogenic activities of 4-hydroxylated, but not of 2-hydroxylated, catechol estrogens.


DNA Damage , DNA/metabolism , Estrogens, Catechol/pharmacology , Liver/metabolism , Microsomes, Liver/metabolism , Quinones/metabolism , Animals , Biotransformation , Cricetinae , DNA/drug effects , Equilenin/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens, Catechol/metabolism , Estrone/pharmacology , Free Radicals/metabolism , Guanine/analogs & derivatives , Hydroxyestrones/pharmacology , Hydroxylation , Liver/drug effects , Male , Mesocricetus
...