Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
Add more filters

Publication year range
1.
Molecules ; 26(7)2021 Apr 03.
Article in English | MEDLINE | ID: mdl-33916785

ABSTRACT

Nordihydroguaiaretic acid (NDGA) is a major lignan metabolite found in Larrea spp., which are widely used in South America to treat various diseases. In breast tissue, estradiol is metabolized to the catechol estrogens such as 4-hydroxyestradiol (4-OHE2), which have been proposed to be cancer initiators potentially involved in mammary carcinogenesis. Catechol-O-methyltransferase (COMT) catalyzes the O-methylation of catechol estrogens to their less toxic methoxy derivatives, such as 4-O-methylestradiol (4-MeOE2). The present study investigated the novel biological activities of NDGA in relation to COMT and the effects of COMT inhibition by NDGA on 4-OHE2-induced cyto- and genotoxicity in MCF-7 human breast cancer cells. Two methoxylated metabolites of NDGA, 3-O-methylNDGA (3-MNDGA) and 4-O-methyl NDGA (4-MNDGA), were identified in the reaction mixture containing human recombinant COMT, NDGA, and cofactors. Km values for the COMT-catalyzed metabolism of NDGA were 2.6 µM and 2.2 µM for 3-MNDGA and 4-MNDGA, respectively. The COMT-catalyzed methylation of 4-OHE2 was inhibited by NDGA at an IC50 of 22.4 µM in a mixed-type mode of inhibition by double reciprocal plot analysis. Molecular docking studies predicted that NDGA would adopt a stable conformation at the COMT active site, mainly owing to the hydrogen bond network. NDGA is likely both a substrate for and an inhibitor of COMT. Comet and apurinic/apyrimidinic site quantitation assays, cell death, and apoptosis in MCF-7 cells showed that NDGA decreased COMT-mediated formation of 4-MeOE2 and increased 4-OHE2-induced DNA damage and cytotoxicity. Thus, NDGA has the potential to reduce COMT activity in mammary tissues and prevent the inactivation of mutagenic estradiol metabolites, thereby increasing catechol estrogen-induced genotoxicities.


Subject(s)
Catechol O-Methyltransferase Inhibitors/chemistry , Catechol O-Methyltransferase Inhibitors/pharmacology , Catechol O-Methyltransferase/metabolism , Estrogens, Catechol/metabolism , Masoprocol/metabolism , Masoprocol/pharmacology , Mutagens/toxicity , Binding Sites , Cell Death/drug effects , DNA Damage , Estrogens, Catechol/chemistry , Estrogens, Catechol/pharmacology , Humans , MCF-7 Cells , Masoprocol/chemistry , Methylation , Molecular Docking Simulation , Recombinant Proteins/metabolism , Substrate Specificity/drug effects
2.
Anal Chem ; 91(24): 15922-15931, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31794208

ABSTRACT

Abundant blood proteins adducted by active electrophiles are excellent markers to predict the risk of electrophile-induced toxicity. However, detecting endogenously adducted proteins by bottom-up selective (or parallel) reaction monitoring (SRM/PRM) is challenging because of the high variability in sample preparation and detection as well as low adduction levels. Here, we reported a new approach in developing PRM methods by combining intact protein measurement with standard additions to target optimal conditions for detecting catechol estrogens (CEs)-adducted human serum albumin (HSA). Blood serum was added with multiple amounts of CEs to obtain serum standards. Intact protein measurement revealed two linear ranges of adduction levels (adducted-CE/HSA): 0.34-0.42 (R2 > 0.94) and 0.81-8.54 (R2 > 0.96) against the amount of added CEs, respectively. Six adduction sites were identified by trypsin (K20, C34, K73, K281, H338, K378) or chymotrypsin (K20, C34, K378) digestion. PRM methods targeting all adducted/nonadducted peptide pairs based on chymotrypsin or trypsin digestion were developed, and the data were compared with those obtained by intact protein measurement. Correlation plots indicated that chymotrypsin-PRM leads to poor sensitivity and largely underestimated protein adduction levels. Trypsin-PRM leads to sensitive and highly correlated (R2 > 0.91) protein adduction levels with a detection limit below the endogenous level and relative standard deviation <25%. As a proof of concept, clinical serum samples were examined by trypsin-PRM, and a slightly higher adduction level was observed for the obesity group when compared with the healthy group. This is the first report on determining adduction levels of blood proteins for long-term exposure to CEs. The standard addition approach can be generally applied to protein adductomics with resolvable mass increments by intact protein measurement to accelerate the development of bottom-up methods close to the inherent limit.


Subject(s)
Estrogens, Catechol/chemistry , Mass Spectrometry/methods , Peptides/analysis , Serum Albumin/chemistry , Chromatography, High Pressure Liquid , Chymotrypsin/metabolism , Estrogens, Catechol/metabolism , Humans , Mass Spectrometry/standards , Nanotechnology , Peptides/metabolism , Peptides/standards , Reference Standards , Serum Albumin/metabolism , Trypsin/metabolism
3.
Drug Metab Dispos ; 47(8): 919-927, 2019 08.
Article in English | MEDLINE | ID: mdl-31147316

ABSTRACT

The cytochrome P450 (P450) 1 family is an important phase I enzyme involved in carcinogen activation. Nitidine chloride (NC) is a pharmacologically active alkaloid with polyaromatic hydrocarbon found in the roots of Zanthoxylum nitidum (Roxb.) DC, a traditional medicinal herb widely used in China. We examined the inhibitory effects of NC on CYP1A1, 1B1, and 1A2. NC significantly inhibited CYP1A1- and 1B1-catalyzed ethoxyresorufin O-deethylation activity (IC50 = 0.28 ± 0.06 and 0.32 ± 0.02 µM, respectively) in a concentration-dependent manner, but only showed slight inhibition of CYP1A2 activity (IC50 > 50 µM). Kinetic analysis revealed that NC competitively inhibited CYP1B1 with a K i value of 0.47 ± 0.05 µM, whereas NC caused a mixed type of inhibition on CYP1A1 with K i and K I values of 0.14 ± 0.04 and 0.19 ± 0.09 µM, respectively. The observed enzyme inhibition neither required NADPH nor revealed time dependency. Molecular docking manifested the generation of strong hydrogen-bonding interactions of Ser116 in CYP1A1 and Ser127 in CYP1B1 with methoxy moiety of NC. Additionally, NC-induced alteration of estradiol (E2) metabolism was also investigated in the present study. Hydroxyestradiols, including 2-hydroxyestradiol [(2-OHE2) nontoxic] and 4-hydroxyestradiol [(4-OHE2) genotoxic] generated in recombinant enzyme incubation systems and cultured MCF-7 cells were analyzed, and NC was found to preferentially inhibit the nontoxic 2-hydroxylation activity of E2 mediated by CYP1A1. In conclusion, NC was a mixed type inhibitor of CYP1A1 and a competitive inhibitor of CYP1B1. The remarkable inhibition on E2 2-hydroxylation might increase the risk of 4-OHE2-induced genotoxicity. SIGNIFICANCE STATEMENT: CYP1 enzymes catalyze oxidative metabolism of a variety of compounds and are known to play a crucial role in the development of cancer. CYP1A1 and CYP1A2 are responsible for hydroxylation of estradiol (E2) at the C-2 position, resulting in the formation of 2-OHE2, which is proposed to be a detoxification pathway. However, CYP1B1-mediated hydroxylation of E2 at the C-4 position has been suggested to be a tumor initiator. The present study found that nitidine chloride is a mixed type inhibitor of CYP1A1 and a competitive inhibitor of CYP1B1. NC preferentially inhibited the nontoxic E2 2-hydroxylation pathway mediated by CYP1A1, which might increase the risk of 4-OHE2-induced genotoxicity and cause severe drug-drug interactions.


Subject(s)
Benzophenanthridines/pharmacology , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Estradiol/metabolism , Metabolic Networks and Pathways/drug effects , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1B1/metabolism , Drug Interactions , Drugs, Chinese Herbal , Enzyme Assays , Estrogens, Catechol/metabolism , Estrogens, Catechol/toxicity , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Docking Simulation , Mutagenicity Tests , Plant Roots/chemistry , Protein Binding , Recombinant Proteins/metabolism , Serine/metabolism , Zanthoxylum/chemistry
4.
Phytother Res ; 33(4): 1033-1043, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30680817

ABSTRACT

Human cytochrome P450 1B1 (CYP1B1)-mediated formation of 4-hydroxyestradiol (4-OHE2) from 17ß-estradiol plays an important role in the progression of human breast cancer, while the biotransformation of 17ß-estradiol to 2-hydroxyestradiol mediated by cytochrome P450 1A1 (CYP1A1) is considered as a less harmful pathway. In this study, inhibitory effects of flavonoids baicalein and oroxylin A, a metabolite of baicalein in human body, on CYP1A1 and 1B1 activities were investigated in vitro. The inhibition intensities of baicalein and oroxylin A towards CYP1B1 were greater than towards CYP1A1 with a mixed mechanism. In addition, oroxylin A showed a stronger inhibitory effect than baicalein towards the CYP1B1-mediated 17ß-estradiol 4-hydroxylation, with the IC50 values of 0.0146 and 2.27 µM, respectively. Docking studies elucidated that oroxylin A had a stronger binding affinity than baicalein for CYP1B1. In MCF-7 cells, compared with baicalein-treated groups, oroxylin A with lower doses decreased and increased the formation of 4-OHE2 and 2-hydroxyestradiol, respectively, with a preferential induction of mRNA of CYP1A1 over CYP1B1. In conclusion, this study demonstrated that oroxylin A showed a stronger inhibitory effect than baicalein on CYP1B1-mediated 4-OHE2 formation in MCF-7 cells, providing crucial implications for their possibly preventive/therapeutic potential against breast cancer via inhibition of CYP1B1, particularly of oroxylin A.


Subject(s)
Carcinogenesis/drug effects , Carcinogenesis/genetics , Cytochrome P-450 CYP1B1/genetics , Estradiol/analogs & derivatives , Estrogens, Catechol/metabolism , Estrogens, Catechol/toxicity , Flavanones/pharmacology , Flavonoids/pharmacology , Breast Neoplasms/chemically induced , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis/chemically induced , Carcinogens/metabolism , Carcinogens/toxicity , Down-Regulation/drug effects , Down-Regulation/genetics , Estradiol/metabolism , Female , Flavanones/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells
5.
Drug Metab Rev ; 49(1): 56-83, 2017 02.
Article in English | MEDLINE | ID: mdl-27826992

ABSTRACT

Catechol-O-methyltransferase, COMT, is an important phase II enzyme catalyzing the transfer of a methyl-group from S-adenosylmethionine to a catechol-containing substrate molecule. A genetic variant Val158Met in the COMT gene leads to a several-fold decrease in the enzymatic activity giving rise to the accumulation of potentially carcinogenic endogenous catechol estrogens and their reactive intermediates and increasing thus the risk of tumorigenesis. However, numerous association studies between the COMT genotype and susceptibility to various malignancies have shown inconsistent and controversial findings indicating that additional gene-gene and gene-environment interactions might be crucial in modulating the physiological role of the COMT. In this review article, the important contribution of dietary catechol-containing flavonoids to modification of the relationships between the COMT genotype and cancer risk is discussed. Whereas, the diverse anticancer activities of common phytochemicals, such as green tea polyphenols, quercetin, fisetin or luteolin, can be markedly changed (both decreased or increased) by the COMT-mediated O-methylation of these exogenous substrates, flavonoids can also behave as potent inhibitors of the COMT enzyme slowing detoxification of endogenous catechol estrogens. Such a many-featured functioning of the COMT and its complex regulation by several different genetic and environmental factors, including plant-based food ingredients, emphasizes the necessity to further stratify the association studies between the COMT genotype and tumor risk by consumption of catechol-containing dietary flavonoids. Currently, it can be only speculated that some of the possible associations might be masked by the regular intake of specific food polyphenols, taking effect in certain communities or populations.


Subject(s)
Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Catechols/metabolism , Estrogens, Catechol/metabolism , Genetic Predisposition to Disease/genetics , Neoplasms/genetics , Catechol O-Methyltransferase Inhibitors/pharmacology , Diet , Estrogens, Catechol/adverse effects , Flavonoids/pharmacology , Humans , Polymorphism, Single Nucleotide/genetics
6.
Orv Hetil ; 158(24): 929-937, 2017 Jun.
Article in Hungarian | MEDLINE | ID: mdl-28604076

ABSTRACT

Considerable knowledge has been gathered on the physiological role of estrogens. However, fairly little information is available on the role of compounds produced in the breakdown process of estrone and estradiol wich may play a role in various diseases associated with estrogen impact. To date, approximately 15 extragonadal estrogen-related compounds have been identified. These metabolites may exert protective, or, instead, pro-inflammatory and/or pro-oncogenic activity in a tissue-specific manner. Systemic and local estrogen metabolite levels are not necesserily correlated, which may promote the diagnostic significance of the locally produced estrogen metabolites in the future. The aim of the present study is a bibliographic review of the extragonadal metabolome in peripheral tissues, and to highlight the role of the peripheral tissue homeostasis of estrogens as well as the non-hormonal biological activity and clinical significance of the estrogen metabolome. Orv Hetil. 2017; 158(24): 929-937.


Subject(s)
Estrogens/metabolism , Receptors, Estrogen/metabolism , Estradiol/metabolism , Estrogens/physiology , Estrogens, Catechol/metabolism , Estrone/metabolism , Humans
7.
Nutr Cancer ; 67(7): 1201-7, 2015.
Article in English | MEDLINE | ID: mdl-26381237

ABSTRACT

Spices are used worldwide, particularly in the Asian and Middle Eastern countries, and considered protective against degenerative diseases, including cancer. Here, we report the efficacy of aqueous and non-aqueous extracts of 11 Apiaceae spices for free radical-scavenging activity and to inhibit cytochrome P450s in two separate reactions involving: 1) 4-hydroxy-17ß-estradiol (4E2), DNA, and CuCl2 and 2) 17ß-estradiol, rat liver microsomes, cofactors, DNA and CuCl2. Oxidative DNA adducts resulting from redox cycling of 4E2 were analyzed by (32)P-postlabeling. Aqueous (5 mg/ml) and non-aqueous extracts (6 mg/ml) substantially inhibited (83-98%) formation of DNA adducts in the microsomal reaction. However, in nonmicrosomal reaction, only aqueous extracts showed the inhibitory activity (83-96%). Adduct inhibition was also observed at five-fold lower concentrations of aqueous extracts of cumin (60%) and caraway (90%), and 10-fold lower concentrations of carrot seeds (76%) and ajowan (90%). These results suggests the presence of 2 groups of phytochemicals: polar compounds that have free radical-scavenging activity and lipophilic compounds that selectively inhibit P450 activity associated with estrogen metabolism. Because most of these Apiaceae spices are used widely with no known toxicity, the phytochemicals from the Apiaceae spices used in foods may be potentially protective against estrogen-mediated breast cancer.


Subject(s)
Antioxidants/pharmacology , Apiaceae/chemistry , Plant Extracts/pharmacology , Spices , Animals , Cytochrome P-450 Enzyme Inhibitors/pharmacology , DNA Adducts , DNA Damage/drug effects , Estradiol/metabolism , Estrogens, Catechol/metabolism , Estrogens, Catechol/pharmacokinetics , Free Radical Scavengers/pharmacology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Plant Extracts/chemistry , Rats , Testis/drug effects , Testis/metabolism
8.
Mol Biol Rep ; 40(4): 3315-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23283740

ABSTRACT

CYP1B1 encodes an estrogen enzyme that oxidizes 17ß-estradiol to 4-hydroxyestradiol. The evidence demonstrates there may be a relationship between CYP1B1 and thyroid function. To date, no study has evaluated if genetic polymorphisms that regulate concentrations of serum FT3 and FT4 contribute to Polycyctic Ovary Syndrome (PCOS). To identify polymorphisms in the CYP1B1 locus associated with PCOS, we genotyped three common polymorphisms across the CYP1B1 locus in 226 patients. A test for association of common variants with susceptibility to PCOS was conducted in a large cohort of 609 subjects. The functional polymorphism CYP1B1 L432V (rs1056836) is associated with serum T4 (P = 0.003), serum FT3 (P < 0.001) and serum FT4 concentrations (P < 0.001). Our study provides the first evidence that genetic variants in CYP1B1 can be associated with serum T4, FT4 and FT3 levels in PCOS. These findings imply novel pathophysiological links between the CYP1B1 locus and thyroid function in PCOS.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Genetic Association Studies , Polycystic Ovary Syndrome/genetics , Thyroxine/genetics , Triiodothyronine/genetics , Adult , Cytochrome P-450 CYP1B1 , Estrogens, Catechol/genetics , Estrogens, Catechol/metabolism , Female , Genetic Predisposition to Disease , Humans , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/pathology , Polymorphism, Single Nucleotide , Thyroid Function Tests , Thyroxine/blood , Triiodothyronine/blood
9.
Gen Comp Endocrinol ; 177(1): 128-42, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22429727

ABSTRACT

Seasonal, periovulatory and 2-hydroxyestradiol-17ß (2-OHE(2))-induced changes on ovarian prostaglandin (PG) E(2) and F(2α) were investigated under in vivo or in vitro in the female catfish Heteropneustes fossilis. Both PGE(2) and PGF(2α) increased significantly during ovarian recrudescence with the peak levels in spawning phase. The PGs showed periovulatory changes with the peak levels at 16 h after the hCG treatment. Incubation of postvitellogenic ovary fragments with estradiol-17ß (E(2)), 2-OHE(2) or 2-methoxyE(2) produced concentration-dependent increases in PG levels; 2-OHE(2) was more effective. In order to identify the receptor mechanism involved in the 2-OHE(2)-induced PG stimulation, the ovarian pieces were incubated with phentolamine (an α-adrenergic antagonist), propranolol (a ß-adrenergic antagonist) or tamoxifen (an estrogen receptor blocker) alone or in combination with 2-OHE(2). The incubation of the tissues with the receptor blockers alone did not produce any significant effect on basal PG levels. However, co- and pre-incubation of the tissues with the blockers resulted in inhibition of the stimulatory effect of 2-OHE(2) on the PGs. Phentolamine was more effective than propranolol. The signal transduction pathway(s) involved in the 2-OHE(2)-induced PG secretion was investigated. The incubation of the ovarian pieces with 3-isobutyl-1-methylxanthine (IBMX, a phosphodiesterase inhibitor), chelerythrine (a protein kinase C inhibitor) and PD098059 (a mitogen-activated protein kinase inhibitor) significantly lowered the basal secretion of PGF(2α) and PGE(2). In contrast, H89 (a protein kinase A inhibitor) increased the basal secretion of PGs at 1 and 5 µM concentration and decreased it at 10 µM concentration. The co- or pre-incubation with IBMX, H89, chelerythrine and PD098059 significantly inhibited the stimulatory effect of 2-OHE(2) on PGF(2α) and PGE(2) levels. The inhibition was higher in the pre-incubation groups. Chelerythrine was the most effective followed by PD098059, IBMX and H89. The results suggest that 2-OHE(2) may employ both adrenergic and estrogen receptors, or a novel receptor mechanism having properties of both adrenergic and estrogen receptors.


Subject(s)
Estrogens, Catechol/metabolism , Ovary/metabolism , Prostaglandins/metabolism , Animals , Catfishes/metabolism , Female , Receptors, Adrenergic/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/physiology
10.
Carcinogenesis ; 32(8): 1285-93, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21665890

ABSTRACT

Estrogens are major risk factors for the development of breast cancer; they can be metabolized to catechols, which are further oxidized to DNA-reactive quinones and semiquinones (SQs). These metabolites are mutagenic and may contribute to the carcinogenic activity of estrogens. Redox cycling of the SQs and subsequent generation of reactive oxygen species (ROS) is also an important mechanism leading to DNA damage. The SQs of exogenous estrogens have been shown to redox cycle, however, redox cycling and the generation of ROS by endogenous estrogens has never been characterized. In the present studies, we determined whether the catechol metabolites of endogenous estrogens, including 2-hydroxyestradiol, 4-hydroxyestradiol, 4-hydroxyestrone and 2-hydroxyestriol, can redox cycle in breast epithelial cells. These catechol estrogens, but not estradiol, estrone, estriol or 2-methoxyestradiol, were found to redox cycle and generate hydrogen peroxide (H(2)O(2)) and hydroxyl radicals in lysates of three different breast epithelial cell lines: MCF-7, MDA-MB-231 and MCF-10A. The generation of ROS required reduced nicotinamide adenine dinucleotide phosphate as a reducing equivalent and was inhibited by diphenyleneiodonium, a flavoenzyme inhibitor, indicating that redox cycling is mediated by flavin-containing oxidoreductases. Using extracellular microsensors, catechol estrogen metabolites stimulated the release of H(2)O(2) by adherent cells, indicating that redox cycling occurs in viable intact cells. Taken together, these data demonstrate that catechol metabolites of endogenous estrogens undergo redox cycling in breast epithelial cells, resulting in ROS production. Depending on the localized concentrations of catechol estrogens and enzymes that mediate redox cycling, this may be an important mechanism contributing to the development of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast/metabolism , Epithelial Cells/metabolism , Estrogens, Catechol/metabolism , Reactive Oxygen Species/metabolism , Breast/cytology , Cells, Cultured , Female , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction
11.
IUBMB Life ; 62(10): 746-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20945454

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/urine , Estrogens, Catechol/metabolism , Estrogens/metabolism , Phenols/urine , Benzhydryl Compounds , Carcinogens/metabolism , Catechol O-Methyltransferase/metabolism , DNA/genetics , DNA/metabolism , DNA Adducts/metabolism , Diethylstilbestrol/metabolism , Estradiol/metabolism , Estrogens, Non-Steroidal/metabolism , Humans , Mutation , Phenols/metabolism , Quinones/metabolism
12.
Drug Metab Dispos ; 38(10): 1892-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20606002

ABSTRACT

The human catechol-O-methyltransferase (COMT) is a polymorphic enzyme that catalyzes the O-methylation of catechol estrogens. Recent animal studies showed that placental COMT is involved in the development of placentas and embryos, probably via the formation of 2-methoxyestradiol. In this study, we analyzed a total of 36 human term placentas to determine their cytosolic COMT activity for the O-methylation of catechol estrogens as well as their sensitivity to inhibition by heat and dietary compounds. Large variations (up to 4-fold) in the COMT activity for the formation of methoxyestrogens were noted with different human placental samples. The cytosolic COMTs in different human placentas also displayed considerable differences in their sensitivity to heat inactivation. This differential sensitivity was not associated with the overall catalytic activity for the O-methylation of catechol estrogen substrates. It was observed that there was a positive correlation (r = 0.760) between the sensitivity of the human placental COMT to heat inactivation and its sensitivity to inhibition by (-)-epigallocatechin-3-gallate (a well known tea polyphenol with COMT-inhibiting activity) but an inverse correlation (r = 0.544) between heat inactivation and inhibition by quercetin (another dietary COMT inhibitor). The differences in inhibition by these two dietary compounds are due to different mechanisms of COMT inhibition involved.


Subject(s)
Catechol O-Methyltransferase/metabolism , Diet , Enzyme Inhibitors/pharmacology , Estrogens, Catechol/pharmacokinetics , Flavonoids/pharmacology , Hot Temperature , Phenols/pharmacology , Placenta/enzymology , Adult , Catechol O-Methyltransferase Inhibitors , Cytosol/enzymology , Enzyme Inhibitors/administration & dosage , Estrogens, Catechol/metabolism , Female , Flavonoids/administration & dosage , Humans , In Vitro Techniques , Methylation , Phenols/administration & dosage , Polyphenols , Pregnancy
13.
Chem Res Toxicol ; 23(8): 1365-73, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20509668

ABSTRACT

Metabolic activation of estrogens to catechols and further oxidation to highly reactive o-quinones generates DNA damage including apurinic/apyrimidinic (AP) sites. 4-Hydroxyequilenin (4-OHEN) is the major catechol metabolite of equine estrogens present in estrogen replacement formulations, known to cause DNA strand breaks, oxidized bases, and stable and depurinating adducts. However, the direct formation of AP sites by 4-OHEN has not been characterized. In the present study, the induction of AP sites in vitro by 4-OHEN and the endogenous catechol estrogen metabolite, 4-hydroxyestrone (4-OHE), was examined by an aldehyde reactive probe assay. Both 4-OHEN and 4-OHE can significantly enhance the levels of AP sites in calf thymus DNA in the presence of the redox cycling agents, copper ion and NADPH. The B-ring unsaturated catechol 4-OHEN induced AP sites without added copper, whereas 4-OHE required copper. AP sites were also generated much more rapidly by 4-OHEN. For both catechol estrogens, the levels of AP sites correlated linearly with 8-oxo-dG levels, implying that depuriniation resulted from reactive oxygen species (ROS) rather than depurination of estrogen-DNA adducts. ROS modulators such as catalase, which scavenges hydrogen peroxide and a Cu(I) chelator, blocked the formation of AP sites. In MCF-7 breast cancer cells, 4-OHEN significantly enhanced the formation of AP sites with added NADH. In contrast, no significant induction of AP sites was detected in 4-OHE-treated cells. The greater redox activity of the equine catechol estrogen produces rapid oxidative DNA damage via ROS, which is enhanced by redox cycling agents and interestingly by NADPH-dependent quinone oxidoreductase.


Subject(s)
DNA Damage , Deoxyguanosine/analogs & derivatives , Equilenin/analogs & derivatives , Estrogens, Catechol/metabolism , Horses , Reactive Oxygen Species/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cattle , Cell Line, Tumor , Chelating Agents/pharmacology , Copper/chemistry , Copper/metabolism , DNA/metabolism , DNA, Neoplasm/metabolism , Deoxyguanosine/metabolism , Equilenin/chemistry , Equilenin/metabolism , Estrogens, Catechol/chemistry , Estrogens, Catechol/pharmacology , Free Radical Scavengers/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Hydroxyestrones/chemistry , Hydroxyestrones/metabolism , Molecular Structure , NADP/chemistry , NADP/metabolism , Oxidation-Reduction/drug effects , Structure-Activity Relationship
14.
Front Neuroendocrinol ; 29(4): 463-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18275991

ABSTRACT

Estradiol is a potent preventative against neurodegenerative disease, in part, by activating antioxidant defense systems scavenging reactive oxygen species, limiting mitochondrial protein damage, improving electron transport chain activity and reducing mitochondrial DNA damage. Estradiol also increases the activity of complex IV of the electron transport chain, improving mitochondrial respiration and ATP production under normal and stressful conditions. However, the high oxidative cellular environment present during neurodegeneration makes estradiol a poor agent for treatment of existing disease. Oxidative stress stimulates the production of the hydroperoxide-dependent hydroxylation of estradiol to the catecholestrogen metabolites, which can undergo reactive oxygen species producing redox cycling, setting up a self-generating toxic cascade offsetting any antioxidant/antiapoptotic effects generated by the parent estradiol. Additional disease-induced factors can further perpetuate this cycle. For example dysregulation of the catecholamine system could alter catechol-O-methyltransferase-catalyzed methylation, preventing removal of redox cycling catecholestrogens from the system enhancing pro-oxidant effects of estradiol.


Subject(s)
Estradiol/metabolism , Neurodegenerative Diseases/metabolism , Oxidative Stress , Aging/physiology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Antioxidants/metabolism , Calcium/metabolism , Estrogens, Catechol/metabolism , Humans , Mitochondria/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/metabolism , Oxidants/metabolism , Oxidation-Reduction , Receptors, Estrogen/metabolism , tau Proteins/metabolism
15.
Biochem Biophys Res Commun ; 382(4): 651-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19289110

ABSTRACT

Mammary gland-distributed and ER-bound UDP-glucuronosyltransferase (UGT)-2B7 metabolizes genotoxic catechol-estrogens (CE) associated with breast cancer initiation. Although UGT2B7 has 3 PKC- and 2 tyrosine kinase (TK)-sites, its inhibition by genistein, herbimycin-A and PP2 with parallel losses in phospho-tyrosine and phospho-Y438-2B7 content indicated it requires tyrosine phosphorylation, unlike required PKC phosphorylation of UGT1A isozymes. 2B7 mutants at PKC-sites had essentially normal activity, while its TK-sites mutants, Y236F- and Y438F-2B7, were essentially inactive. Overexpression of regular or active Src, but not dominant-negative Src, in 2B7-transfected COS-1 cells increased 2B7 activity and phospho-Y438-2B7 by 50%. Co-localization of 2B7 and regular SrcTK in COS-1 cells that was dissociated by pretreatment with Src-specific PP2-inhibitor provided strong evidence Src supports 2B7 activity. Consistent with these findings, evidence indicates an appropriate set of ER proteins with Src-homology binding-domains, including 2B7 and well-known multi-functional Src-engaged AKAP12 scaffold, supports Src-dependent phosphorylation of CE-metabolizing 2B7 enabling it to function as a tumor suppressor.


Subject(s)
Breast Neoplasms/enzymology , Estrogens, Catechol/metabolism , Glucuronosyltransferase/metabolism , src-Family Kinases/metabolism , Animals , Benzoquinones/pharmacology , COS Cells , Chlorocebus aethiops , DNA Damage , Genistein/pharmacology , Glucuronosyltransferase/genetics , Humans , Lactams, Macrocyclic/pharmacology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Estrogen/metabolism , Rifabutin/analogs & derivatives , Transfection , src-Family Kinases/antagonists & inhibitors
16.
J Steroid Biochem Mol Biol ; 113(1-2): 65-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19095062

ABSTRACT

In the present study, we investigated the inhibitory effect of three catechol-containing coffee polyphenols, chlorogenic acid, caffeic acid and caffeic acid phenethyl ester (CAPE), on the O-methylation of 2- and 4-hydroxyestradiol (2-OH-E(2) and 4-OH-E(2), respectively) catalyzed by the cytosolic catechol-O-methyltransferase (COMT) isolated from human liver and placenta. When human liver COMT was used as the enzyme, chlorogenic acid and caffeic acid each inhibited the O-methylation of 2-OH-E(2) in a concentration-dependent manner, with IC(50) values of 1.3-1.4 and 6.3-12.5 microM, respectively, and they also inhibited the O-methylation of 4-OH-E(2), with IC(50) values of 0.7-0.8 and 1.3-3.1 microM, respectively. Similar inhibition pattern was seen with human placental COMT preparation. CAPE had a comparable effect as caffeic acid for inhibiting the O-methylation of 2-OH-E(2), but it exerted a weaker inhibition of the O-methylation of 4-OH-E(2). Enzyme kinetic analyses showed that chlorogenic acid and caffeic acid inhibited the human liver and placental COMT-mediated O-methylation of catechol estrogens with a mixed mechanism of inhibition (competitive plus noncompetitive). Computational molecular modeling analysis showed that chlorogenic acid and caffeic acid can bind to human soluble COMT at the active site in a similar manner as the catechol estrogen substrates. Moreover, the binding energy values of these two coffee polyphenols are lower than that of catechol estrogens, which means that coffee polyphenols have higher binding affinity for the enzyme than the natural substrates. This computational finding agreed perfectly with our biochemical data.


Subject(s)
Catechol O-Methyltransferase Inhibitors , Coffee/chemistry , Estrogens, Catechol/metabolism , Flavonoids/pharmacology , Phenols/pharmacology , Adult , Biocatalysis/drug effects , Caffeic Acids/chemistry , Caffeic Acids/pharmacology , Catechol O-Methyltransferase/chemistry , Chlorogenic Acid/chemistry , Chlorogenic Acid/pharmacology , Computational Biology , Cytosol/drug effects , Cytosol/enzymology , Estrogens, Catechol/chemistry , Female , Flavonoids/chemistry , Humans , Inhibitory Concentration 50 , Kinetics , Liver/cytology , Liver/drug effects , Liver/enzymology , Methylation/drug effects , Models, Molecular , Phenols/chemistry , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/pharmacology , Placenta/cytology , Placenta/drug effects , Placenta/enzymology , Polyphenols , Protein Binding/drug effects , Protein Structure, Secondary
17.
Cancer Res ; 67(2): 812-7, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234793

ABSTRACT

The oxidative metabolism of estrogens has been implicated in the development of breast cancer; yet, relatively little is known about the mechanism by which estrogens cause DNA damage and thereby initiate mammary carcinogenesis. To determine how the metabolism of the parent hormone 17beta-estradiol (E2) leads to the formation of DNA adducts, we used the recombinant, purified phase I enzyme, cytochrome P450 1B1 (CYP1B1), which is expressed in breast tissue, to oxidize E2 in the presence of 2'-deoxyguanosine or 2'-deoxyadenosine. We used both gas and liquid chromatography with tandem mass spectrometry to measure E2, the 2- and 4-catechol estrogens (2-OHE2, 4-OHE2), and the depurinating adducts 4-OHE(2)-1(alpha,beta)-N7-guanine (4-OHE2-N7-Gua) and 4-OHE(2)-1(alpha,beta)-N3-adenine (4-OHE2-N3-Ade). CYP1B1 oxidized E2 to the catechol 4-OHE2 and the labile quinone 4-hydroxyestradiol-quinone to produce 4-OHE2-N7-Gua and 4-OHE2-N3-Ade in a time- and concentration-dependent manner. Because the reactive quinones were produced as part of the CYP1B1-mediated oxidation reaction, the adduct formation followed Michaelis-Menten kinetics. Under the conditions of the assay, the 4-OHE2-N7-Gua adduct (Km, 4.6+/-0.7 micromol/L; kcat, 45+/-1.6/h) was produced 1.5 times more efficiently than the 4-OHE2-N3-Ade adduct (Km, 4.6+/-1.0 micromol/L; kcat, 30+/-1.5/h). The production of adducts was two to three orders of magnitude lower than the 4-OHE2 production. The results present direct proof of CYP1B1-mediated, E2-induced adduct formation and provide the experimental basis for future studies of estrogen carcinogenesis.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Deoxyadenosines/metabolism , Deoxyguanosine/metabolism , Estradiol/metabolism , Cytochrome P-450 CYP1B1 , DNA Adducts/metabolism , Estrogens, Catechol/metabolism , Kinetics , Recombinant Proteins/metabolism
18.
Carcinogenesis ; 29(2): 363-70, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18192686

ABSTRACT

The tissue concentrations of the female sex hormone 17beta-estradiol (E2) and its reactive catechol metabolites such as 4-hydroxyestradiol (4-HO-E2) play important roles in hormonal carcinogenesis. They are influenced by the activity of local enzymes involved in the metabolic activation and inactivation of E2. In the mammary gland, catechol estrogens are predominately inactivated by catechol-O-methyltransferase (COMT). Food supplements containing the soy isoflavones genistein and daidzein are consumed because they are believed to protect from breast cancer; however, this proposed benefit is controversial. The aim of the present study was to investigate the influence of soy isoflavones on the gene expression and activity of COMT in cultured human mammary adenocarcinoma MCF-7 cells. Levels of COMT messenger RNA (mRNA) were determined by reverse transcription/competitive polymerase chain reaction and COMT activity was determined by high-performance liquid chromatography analysis of the methylation products of both the model substrate quercetin and the physiological relevant substrate 4-HO-E2. Our study demonstrates for the first time that soy isoflavones at hormonally active concentrations cause a significant reduction of both COMT mRNA levels and COMT activity as well as of the methylation of 4-HO-E2. Experiments using the estrogen receptor (ER) antagonist ICI 182,780 support a role of the ER in the isoflavone-induced down-regulation of COMT expression. Thus, this study not only demonstrates that hormonally active concentrations of soy isoflavones inhibit the detoxification of catechols in this human breast cancer cell line but also implies that diet might influence COMT activity to a greater extent than heretofore recognized.


Subject(s)
Breast Neoplasms/drug therapy , Catechol O-Methyltransferase/metabolism , Estradiol/analogs & derivatives , Gene Expression Regulation, Neoplastic , Glycine max/metabolism , Cell Line, Tumor , Cell Proliferation , Diet , Estradiol/metabolism , Estradiol/pharmacology , Estrogens, Catechol/metabolism , Female , Fulvestrant , Humans , Models, Biological , Models, Chemical , Receptors, Estrogen/metabolism
19.
Int J Mol Med ; 22(6): 769-79, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020775

ABSTRACT

Human catechol-O-methyltransferase (COMT, EC 2.1.1.6) catalyzes the transfer of the methyl group to a variety of endogenous and exogenous catechol substrates using S-adenosyl-L-methionine as the methyl donor. This enzymatic O-methylation plays an important role in the inactivation of biologically-active and toxic catechols. A number of studies in recent years have sought to characterize the polymorphism of human COMT, and also to determine the catalytic activity of polymorphic enzymes. We report here the identification of a new haplotype of the human COMT gene with triplet point mutations, which encodes the D51G/S60F/K162R mutant of the soluble COMT and the D101G/S110F/K212R mutant of the membrane-bound COMT. Kinetic analysis showed that these new COMT variants had essentially the same kinetic characteristics and catalytic activity as the wild-type COMTs for the O-methylation of 2-hydroxyestradiol and 4-hydroxyestradiol in vitro, but they have asignificantly reduced thermostability at 37 degrees C. In addition, the mutant enzymes have different binding affinities for S-adenosyl-L-methionine compared with the wild-type COMTs. In agreement with our biochemical observations, molecular modeling studies also showed that the variant human COMT proteins shared nearly the same overall structures as the wild-type proteins. The binding energy values of the mutant COMTs in complex with catechol estrogen substrates were similar to those of the wild-type COMTs bound with the same substrates.


Subject(s)
Catechol O-Methyltransferase/chemistry , Catechol O-Methyltransferase/genetics , Point Mutation , Catechol O-Methyltransferase/metabolism , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogens, Catechol/metabolism , Haplotypes , Humans , Kinetics , Models, Molecular , Polymorphism, Genetic , Protein Stability , Sequence Alignment
20.
Cancer Epidemiol Biomarkers Prev ; 16(3): 481-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17372243

ABSTRACT

Polymorphisms within genes responsible for estrogen catabolism could alter cellular levels of genotoxic 4-hydroxylated catechol estrogens and antiangiogenic 2-methoxyestradiol, thus influencing risk of developing ovarian cancer. We carried out a population-based case-control study of 310 epithelial ovarian cancer cases and 585 controls in African-American and Caucasian women ages 35 to 54 years from Seattle, Atlanta, and Detroit metropolitan areas. Subjects were interviewed and genotyped for CYP1A1 m1, m2, m3, and m4; CYP1B1 Arg(48)Gly, Ala(119)Ser, Val(432)Leu, and Asn(453)Ser; COMT Val(158)Met; UGT1A1 A(TA)nTAA; and SULT1A1 Arg(213)His polymorphisms. Unconditional logistic regression was used to calculate odds ratios (OR). Haplotypes were inferred and analyzed using models based on expectation-maximization with progressive ligation and Bayesian coalescence theory. CYP1B1 Leu(432) carriers were at increased risk of ovarian cancer, with an adjusted OR of 1.5 (95% confidence interval, 1.1-2.3) compared with Val(432) homozygotes. The most common CYP1B1 haplotype was Arg(48)-Ala(119)-Val(432)-Asn(453). All other haplotypes with frequencies >5% contained the Leu(432) allele. In diplotype analyses, relative to women homozygous for Arg(48)-Ala(119)-Val(432)-Asn(453), women with diplotypes containing at least one Leu(432) allele had adjusted ORs ranging from 1.3 to 2.2. Among women homozygous for COMT Met(158), carriers of CYP1B1 Leu(432) had a 2.6-fold increase in risk relative to CYP1B1 Val(432) homozygotes (95% confidence interval, 1.1-5.9). This latter result is opposite in direction from a similar analysis conducted by other investigators in a different study population. No association of ovarian cancer risk was observed with any of the other polymorphisms examined, either alone or in combination.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Estrogens, Catechol/genetics , Estrogens, Catechol/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polymorphism, Genetic , Adult , Black or African American/genetics , Alleles , Bayes Theorem , Case-Control Studies , Estrogens/genetics , Estrogens/metabolism , Estrogens, Catechol/biosynthesis , Female , Genotype , Haplotypes , Humans , Logistic Models , Middle Aged , Ovarian Neoplasms/epidemiology , Population Surveillance , Registries , Risk Factors , SEER Program , United States/epidemiology , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL