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










Publication year range
1.
Environ Pollut ; 239: 281-288, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29660500

ABSTRACT

In this study, we determined the concentration of equine estrogens, such as equilin (Eq) and equilenin (Eqn), in the river water collected from nine research stations in Hokkaido, Japan. The LC-MS/MS analysis revealed that Eq concentrations were 2.7 ±â€¯6.7, 0.22 ±â€¯0.12, and 1.2 ±â€¯0.64 ng/L in Sep 2015, Feb 2016, and Jul 2016, respectively. Eqn had concentration levels similar to those of Eq. Comparison of the concentrations at nine research stations showed that seasonal variation was observed in the detected Eq and Eqn concentration levels. This study was the first to show the occurrences and seasonal variation of Eq and Eqn in the river water of Japan. We further investigated the reproductive and transgenerational effects of Eq in Japanese medaka (Oryzias latipes) exposed to 10, 100, and 1000 ng/L for 21 days and assessed the transcriptional profiles of the estrogen-responsive genes in the livers of both sexes. The reproduction assay demonstrated that 1000 ng/L of Eq adversely affected the reproduction (i.e. fecundity) in the F0 generation and that the hatching of F1 generation fertilized eggs was reduced in the 100 and 1000 ng/L treatment groups. Our qRT-PCR assay revealed that the mRNA expression levels of hepatic vitellogenin 1 and 2, choriogenin L and H, and estrogen receptor α were significantly up-regulated in males exposed to 100 and/or 1000 ng/L of Eq. In contrast, the transcriptional levels of several genes, such as pregnane X receptor and cytochrome P450 3A, were down-regulated in the livers of males after the 21-d exposure. These results suggest that Eq has endocrine-disrupting potential such as reproductive and transgenerational effects by the modulation of hepatic estrogen-responsive genes expression on medaka.


Subject(s)
Endocrine Disruptors/analysis , Environmental Monitoring , Equilenin/analysis , Equilin/analysis , Oryzias/physiology , Water Pollutants, Chemical/analysis , Animals , Climate , Endocrine Disruptors/metabolism , Endocrine System/drug effects , Equilenin/metabolism , Equilin/metabolism , Estrogen Receptor alpha , Estrogens/metabolism , Female , Fertility/drug effects , Fresh Water , Gene Expression , Horses , Japan , Liver/metabolism , Male , Oryzias/metabolism , Pregnane X Receptor , Receptors, Steroid , Reproduction/drug effects , Rivers , Seasons , Vitellogenins/metabolism , Water Pollutants, Chemical/metabolism
2.
Mol Cells ; 38(5): 409-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25947291

ABSTRACT

Low-barrier hydrogen bonds (LBHBs) have been proposed to have important influences on the enormous reaction rate increases achieved by many enzymes. Δ(5)-3-ketosteroid isomerase (KSI) catalyzes the allylic isomerization of Δ(5)-3-ketosteroid to its conjugated Δ(4)-isomers at a rate that approaches the diffusion limit. Tyr14, a catalytic residue of KSI, has been hypothesized to form an LBHB with the oxyanion of a dienolate steroid intermediate generated during the catalysis. The unusual chemical shift of a proton at 16.8 ppm in the nuclear magnetic resonance spectrum has been attributed to an LBHB between Tyr14 Oη and C3-O of equilenin, an intermediate analogue, in the active site of D38N KSI. This shift in the spectrum was not observed in Y30F/Y55F/D38N and Y30F/Y55F/Y115F/D38N mutant KSIs when each mutant was complexed with equilenin, suggesting that Tyr14 could not form LBHB with the intermediate analogue in these mutant KSIs. The crystal structure of Y30F/Y55F/Y115F/D38N-equilenin complex revealed that the distance between Tyr14 Oη and C3-O of the bound steroid was within a direct hydrogen bond. The conversion of LBHB to an ordinary hydrogen bond in the mutant KSI reduced the binding affinity for the steroid inhibitors by a factor of 8.1-11. In addition, the absence of LBHB reduced the catalytic activity by only a factor of 1.7-2. These results suggest that the amount of stabilization energy of the reaction intermediate provided by LBHB is small compared with that provided by an ordinary hydrogen bond in KSI.


Subject(s)
Equilenin/metabolism , Pseudomonas putida/enzymology , Steroid Isomerases/chemistry , Steroid Isomerases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Equilenin/chemistry , Hydrogen Bonding , Models, Molecular , Mutation , Protein Binding , Proton Magnetic Resonance Spectroscopy , Pseudomonas putida/genetics , Steroid Isomerases/metabolism , Substrate Specificity
3.
J Appl Toxicol ; 35(9): 1040-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25611945

ABSTRACT

Although several previous studies have demonstrated the presence of equine estrogens in the aquatic environment, limited data are currently available on the endocrine-disrupting potentials in fish and the risks they pose to aquatic organisms. To investigate the interactions of major equine estrogens equilin (Eq) and equilenin (Eqn), as well as their metabolites 17α-dihydroequilin, 17ß-dihydroequilin, 17α-dihydroequilenin and 17ß-dihydroequilenin, with the estrogen receptor α (ERα) of medaka (Oryzias latipes), a three-dimensional model of the ligand-binding domain (LBD) of ERα was built in silico, and docking simulations were performed. The docking simulation analysis indicated that the interaction of 17ß-dihydroequilenin with the ERα LBD is the most potent, followed by those of 17α-dihydroequilin and 17ß-dihydroequilin, whereas those of Eq and Eqn were least potent. We further analyzed gene expression profiles in the livers of male medaka exposed to Eq and Eqn. A DNA microarray representing 6000 genes revealed that 24-h exposure to Eq and Eqn (100 ng/L) upregulated the expression of 6 and 34 genes in the livers of males, respectively. Genes upregulated by Eq included the estrogenic biomarker genes vitellogenins and choriogenins, suggesting the estrogenic potential of Eq. In contrast, Eqn exposure upregulated several cancer-related genes, such as mediator complex subunit 16 and RAS oncogene family members, suggesting a carcinogenic potential for Eqn. These results suggest that equine estrogens may have not only endocrine-disrupting potentials via the ERα signaling pathway but also carcinogenic potency in male medaka.


Subject(s)
Endocrine Disruptors/toxicity , Equilenin/toxicity , Equilin/toxicity , Liver/drug effects , Oryzias/metabolism , Water Pollutants, Chemical/toxicity , Animals , Endocrine Disruptors/metabolism , Equilenin/metabolism , Equilin/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Ligands , Liver/metabolism , Male , Molecular Docking Simulation , Oligonucleotide Array Sequence Analysis , Protein Binding , Transcriptome/drug effects , Water Pollutants, Chemical/metabolism
4.
Chem Biol Interact ; 196(1-2): 1-10, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22290292

ABSTRACT

o-Quinone forming estrogens and selective estrogen receptor modulators (SERMs) have been associated with carcinogenesis. LY2066948, a novel SERM in development by Eli Lilly for the treatment of uterine fibroids and myomas, has structural similarity to the equine estrogen equilenin present in hormone replacement formulations; both contain a naphthol group susceptible to oxidative metabolism to o-quinones. LY2066948 was synthesized and assayed for antiestrogenic activity, and in cell culture was confirmed to be a more potent antiestrogen than the prototypical SERM, 4-hydroxytamoxifen. Oxidation of LY2066948 with 2-iodoxybenzoic acid gave an o-quinone (t(1/2)=3.9 ± 0.1h) which like 4-hydroxyequilenin-o-quinone (t(1/2)=2.5 ± 0.2 h) was observed to be exceptionally long-lived with the potential to cause cytotoxicity and/or genotoxicity. In model reactions with tyrosinase, the catechol metabolites of LY2066948 and equilenin were products; interestingly, in the presence of ascorbate to inhibit autoxidation, these catechols were formed quantitatively. Tyrosinase incubations in the presence of GSH gave the expected GSH conjugates resulting from trapping of the o-quinones, which were characterized by LC-MS/MS. Incubations of LY2066948 or equilenin with rat liver microsomes also gave detectable o-quinone trapped GSH conjugates; however, as observed with other SERMs, oxidative metabolism of LY2066948 mainly occurred on the amino side chain to yield the N-dealkylated metabolite. CYP1B1 is believed to be responsible for extra-hepatic generation of genotoxic estrogen quinones and o-quinone GSH conjugates were detected in equilenin incubations. However, in corresponding incubations with CYP1B1 supersomes, no o-quinone GSH conjugates of LY2066948 were detected. These studies suggest that although the naphthol group is susceptible to oxidative metabolism to long-lived o-quinones, the formation of these quinones by cytochrome P450 can be attenuated by the chemistry of the remainder of the molecule as in the case of LY2066948.


Subject(s)
Equilenin/analogs & derivatives , Naphthalenes/chemistry , Piperidines/chemistry , Quinones/chemistry , Selective Estrogen Receptor Modulators/chemistry , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytochrome P-450 CYP1B1 , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Equilenin/chemistry , Equilenin/metabolism , Female , Half-Life , Inhibitory Concentration 50 , Kinetics , Magnetic Resonance Spectroscopy , Microsomes, Liver , Naphthalenes/metabolism , Naphthalenes/pharmacology , Oxidation-Reduction , Piperidines/metabolism , Piperidines/pharmacology , Quinones/metabolism , Quinones/pharmacology , Rats , Rats, Sprague-Dawley , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Tandem Mass Spectrometry
5.
Chem Res Toxicol ; 24(12): 2153-66, 2011 Dec 19.
Article in English | MEDLINE | ID: mdl-21910479

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are suspect human lung carcinogens and can be metabolically activated to remote quinones, for example, benzo[a]pyrene-1,6-dione (B[a]P-1,6-dione) and B[a]P-3,6-dione by the action of either P450 monooxygenase or peroxidases, and to non-K region o-quinones, for example B[a]P-7,8-dione, by the action of aldo keto reductases (AKRs). B[a]P-7,8-dione also structurally resembles 4-hydroxyequilenin o-quinone. These three classes of quinones can redox cycle, generate reactive oxygen species (ROS), and produce the mutagenic lesion 8-oxo-dGuo and may contribute to PAH- and estrogen-induced carcinogenesis. We compared the ability of a complete panel of human recombinant AKRs to catalyze the reduction of PAH o-quinones in the phenanthrene, chrysene, pyrene, and anthracene series. The specific activities for NADPH-dependent quinone reduction were often 100-1000 times greater than the ability of the same AKR isoform to oxidize the cognate PAH-trans-dihydrodiol. However, the AKR with the highest quinone reductase activity for a particular PAH o-quinone was not always identical to the AKR isoform with the highest dihydrodiol dehydrogenase activity for the respective PAH-trans-dihydrodiol. Discrete AKRs also catalyzed the reduction of B[a]P-1,6-dione, B[a]P-3,6-dione, and 4-hydroxyequilenin o-quinone. Concurrent measurements of oxygen consumption, superoxide anion, and hydrogen peroxide formation established that ROS were produced as a result of the redox cycling. When compared with human recombinant NAD(P)H:quinone oxidoreductase (NQO1) and carbonyl reductases (CBR1 and CBR3), NQO1 was a superior catalyst of these reactions followed by AKRs and last CBR1 and CBR3. In A549 cells, two-electron reduction of PAH o-quinones causes intracellular ROS formation. ROS formation was unaffected by the addition of dicumarol, suggesting that NQO1 is not responsible for the two-electron reduction observed and does not offer protection against ROS formation from PAH o-quinones.


Subject(s)
Alcohol Oxidoreductases/metabolism , Equilenin/analogs & derivatives , NAD(P)H Dehydrogenase (Quinone)/metabolism , Polycyclic Aromatic Hydrocarbons/metabolism , Quinones/metabolism , Alcohol Oxidoreductases/genetics , Aldehyde Reductase , Aldo-Keto Reductases , Benzopyrenes/chemistry , Benzopyrenes/toxicity , Biocatalysis , Cell Line, Tumor , Equilenin/chemistry , Equilenin/metabolism , Equilenin/toxicity , Humans , Isomerism , NAD(P)H Dehydrogenase (Quinone)/genetics , Oxidation-Reduction/drug effects , Polycyclic Aromatic Hydrocarbons/chemistry , Polycyclic Aromatic Hydrocarbons/toxicity , Quinones/chemistry , Quinones/toxicity , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
7.
Chem Res Toxicol ; 23(8): 1374-83, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20540524

ABSTRACT

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.


Subject(s)
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
8.
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
9.
J Am Chem Soc ; 132(18): 6474-80, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20397697

ABSTRACT

A light-activated reaction analog has been developed to mimic the catalytic reaction cycle of Delta(5)-3-ketosteroid isomerase to probe the functionally relevant protein solvation response to the catalytic charge transfer. Delta(5)-3-ketosteroid isomerase from Pseudomonas putida catalyzes a C-H bond cleavage and formation through an enolate intermediate. Conversion of the ketone substrate to the enolate intermediate is simulated by a photoacid bound to the active site oxyanion hole. In the ground state, the photoacid electrostatically resembles the enolate intermediate while the low pK(a) excited state resembles the ketone starting material. Time-resolved fluorescence experiments with photoacids coumarin 183 and equilenin show the active site of Delta(5)-3-ketosteroid isomerase to be largely unperturbed by the light-activated reaction. The small solvation response for the photoacid at the active site as compared with a simple solvent suggests the active site does not significantly change its electrostatic environment during the catalytic cycle. Instead, the reaction takes place in an electrostatically preorganized environment.


Subject(s)
Catalytic Domain , Isomerases/chemistry , Isomerases/metabolism , Ketosteroids/metabolism , Solvents/chemistry , Biocatalysis , Coumarins/metabolism , Equilenin/metabolism , Light , Models, Molecular , Pseudomonas putida/enzymology , Spectrometry, Fluorescence
10.
Nucleic Acids Res ; 38(12): e133, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20406772

ABSTRACT

Estrogen-DNA adducts are potential biomarkers for assessing the risk and development of estrogen-associated cancers. 4-Hydroxyequilenin (4-OHEN) and 4-hydroxyequilin (4-OHEQ), the metabolites of equine estrogens present in common hormone replacement therapy (HRT) formulations, are capable of producing bulky 4-OHEN-DNA adducts. Although the formation of 4-OHEN-DNA adducts has been reported, their quantitative detection in mammalian cells has not been done. To quantify such DNA adducts, we generated a novel monoclonal antibody (4OHEN-1) specific for 4-OHEN-DNA adducts. The primary epitope recognized is one type of stereoisomers of 4-OHEN-dA adducts and of 4-OHEN-dC adducts in DNA. An immunoassay with 4OHEN-1 revealed a linear dose-response between known amounts of 4-OHEN-DNA adducts and the antibody binding to those adducts, with a detection limit of approximately five adducts/10(8) bases in 1 microg DNA sample. In human breast cancer cells, the quantitative immunoassay revealed that 4-OHEN produces five times more 4-OHEN-DNA adducts than does 4-OHEQ. Moreover, in a mouse model for HRT, oral administration of Premarin increased the levels of 4-OHEN-DNA adducts in various tissues, including the uterus and ovaries, in a time-dependent manner. Thus, we succeeded in establishing a novel immunoassay for quantitative detection of 4-OHEN-DNA adducts in mammalian cells.


Subject(s)
Antibodies, Monoclonal/immunology , DNA Adducts/immunology , Enzyme-Linked Immunosorbent Assay , Aging , Animals , Antibody Specificity , Cell Line, Tumor , DNA Adducts/analysis , DNA Adducts/chemistry , Equilenin/analogs & derivatives , Equilenin/chemistry , Equilenin/metabolism , Equilin/analogs & derivatives , Equilin/chemistry , Equilin/metabolism , Estrogens, Conjugated (USP)/administration & dosage , Female , Humans , Mice , Mice, Inbred BALB C
11.
Proc Natl Acad Sci U S A ; 107(5): 1960-5, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080683

ABSTRACT

The catalytic importance of enzyme active-site interactions is frequently assessed by mutating specific residues and measuring the resulting rate reductions. This approach has been used in bacterial ketosteroid isomerase to probe the energetic importance of active-site hydrogen bonds donated to the dienolate reaction intermediate. The conservative Tyr16Phe mutation impairs catalysis by 10(5)-fold, far larger than the effects of hydrogen bond mutations in other enzymes. However, the less-conservative Tyr16Ser mutation, which also perturbs the Tyr16 hydrogen bond, results in a less-severe 10(2)-fold rate reduction. To understand the paradoxical effects of these mutations and clarify the energetic importance of the Tyr16 hydrogen bond, we have determined the 1.6-A resolution x-ray structure of the intermediate analogue, equilenin, bound to the Tyr16Ser mutant and measured the rate effects of mutating Tyr16 to Ser, Thr, Ala, and Gly. The nearly identical 200-fold rate reductions of these mutations, together with the 6.4-A distance observed between the Ser16 hydroxyl and equilenin oxygens in the x-ray structure, strongly suggest that the more moderate rate effect of this mutant is not due to maintenance of a hydrogen bond from Ser at position 16. These results, additional spectroscopic observations, and prior structural studies suggest that the Tyr16Phe mutation results in unfavorable interactions with the dienolate intermediate beyond loss of a hydrogen bond, thereby exaggerating the apparent energetic benefit of the Tyr16 hydrogen bond relative to the solution reaction. These results underscore the complex energetics of hydrogen bonding interactions and site-directed mutagenesis experiments.


Subject(s)
Steroid Isomerases/chemistry , Steroid Isomerases/genetics , Amino Acid Substitution , Catalytic Domain/genetics , Comamonas testosteroni/enzymology , Comamonas testosteroni/genetics , Crystallography, X-Ray , Equilenin/chemistry , Equilenin/metabolism , Hydrogen Bonding , Ketosteroids/chemistry , Ketosteroids/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Steroid Isomerases/metabolism
12.
J Comput Biol ; 16(11): 1577-91, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19958084

ABSTRACT

The functional prediction of proteins is one of the most challenging problems in modern biology. An established computational technique involves the identification of three-dimensional local similarities in proteins. In this article, we present a novel method to quickly identify promising binding sites. Our aim is to efficiently detect putative binding sites without explicitly aligning them. Using the theory of Spherical Harmonics, a candidate binding site is modeled as a Binding Ball. The Binding Ball signature, offered by the Spherical Fourier coefficients, can be efficiently used for a fast detection of putative regions. Our contribution includes the Binding Ball modeling and the definition of a scoring function that does not require aligning candidate regions. Our scoring function can be computed efficiently using a property of Spherical Fourier transform (SFT) that avoids the evaluation of all alignments. Experiments on different ligands show good discrimination power when searching for known binding sites. Moreover, we prove that this method can save up to 40% in time compared with traditional approaches.


Subject(s)
Fourier Analysis , Proteins/chemistry , Adenosine Triphosphate/metabolism , Binding Sites , Databases, Protein , Equilenin/metabolism , Ligands , Models, Molecular , Time Factors
14.
Biochemistry ; 48(30): 7098-109, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19527068

ABSTRACT

The equine estrogens equilin (EQ) and equilenin (EN) are the active components in the widely prescribed hormone replacement therapy formulation Premarin. Metabolic activation of EQ and EN generates the catechol 4-hydroxyequilenin (4-OHEN) that autoxidizes to the reactive o-quinone form in aerated aqueous solutions. The o-quinones react predominantly with C, and to a lesser extent with A and G, to form premutagenic cyclic covalent DNA adducts in vitro and in vivo. To obtain insights into the structural properties of these biologically important DNA lesions, we have synthesized site-specifically modified oligonucleotides containing the stereoisomeric 1'S,2'R,3'R-4-OHEN-C3 and 1'R,2'S,3'S-4-OHEN-C4 adducts derived from the reaction of 4-OHEN with the C in the oligonucleotide 5'-GGTAGCGATGG in aqueous solution. A combined NMR and computational approach was utilized to determine the conformational characteristics of the two major 4-OHEN-C3 and 4-OHEN-C4 stereoisomeric adducts formed in this oligonucleotide hybridized with its complementary strand. In both cases, the modified C adopts an anti glycosidic bond conformation; the equilenin distal ring protrudes into the minor groove while its two proximal hydroxyl groups are exposed on the major groove side of the DNA duplex. The bulky 4-OHEN-C adduct distorts the duplex within the central GC*G portion, but Watson-Crick pairing is maintained adjacent to C* in both stereoisomeric adducts. For the 4-OHEN-C3 adduct, the equilenin rings are oriented toward the 5'-end of the modified strand, while in 4-OHEN-C4 the equilenin is 3'-directed. Correspondingly, the distortions of the double-helical structures are more pronounced on the 5'- or the 3'-side of the lesion, respectively. These differences in stereoisomeric adduct conformations may play a role in the processing of these lesions in cellular environments.


Subject(s)
Cytidine/chemistry , DNA Adducts/chemistry , Equilenin/analogs & derivatives , Equilin/chemistry , Estradiol Congeners/chemistry , Nucleic Acid Conformation , Oligonucleotides/chemistry , Animals , Base Sequence , Cytidine/metabolism , DNA Damage , Equilenin/chemistry , Equilenin/metabolism , Equilin/metabolism , Horses , Humans , Molecular Conformation , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Oligonucleotides/genetics , Oligonucleotides/metabolism , Stereoisomerism
15.
J Biol Chem ; 284(13): 8633-42, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19158089

ABSTRACT

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.


Subject(s)
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
16.
Biochemistry ; 44(44): 14565-76, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16262256

ABSTRACT

The drug Premarin is the most widely used formula for hormone replacement therapy. However, long-term exposure to estrogens from the Premarin drug increases the risk of breast cancer. Equilin and equilenin, major components of Premarin, are predominantly metabolized to 4-hydroxyequilenin (4-OHEN). The quinoids produced by 4-OHEN oxidation react with dG, dA, and dC to form unusual stable cyclic bulky adducts, with four stereoisomers identified for each base adduct. The 4-OHEN-dC adducts are most predominant. They are mutagenic in vitro and have been found in human tumor tissue. We have carried out molecular modeling and molecular dynamics simulations to investigate structures and thermodynamics of the four 4-OHEN-dC stereoisomeric adducts in DNA duplexes. Our results show that the structure of each stereoisomer adduct in duplex DNA is specifically governed by its unique stereochemistry. The bulky adducts, with an obstructed Watson-Crick edge and an equilenin ring system near perpendicular to the damaged cytosine, are located in the B-DNA major or minor groove, with the modified cytosine in the syn or anti conformation, respectively. The DNA duplex structures are distorted, in terms of Watson-Crick pairing at and near the lesion, stacking interactions, and groove dimensions. Stereochemistry determines the orientation of the equilenin rings with respect to the 5'- to 3'-direction of the modified strand, as well as the positioning of the equilenin moiety's methyl and hydroxyl groups for each stereoisomer. The unusual structures and the stereochemical effects underlie their biological processing as miscoding DNA lesions whose mutagenic properties may contribute to breast cancer.


Subject(s)
Cytosine/chemistry , DNA Adducts , Equilenin/chemistry , Nucleic Acid Conformation , Animals , Equilenin/metabolism , Horses , Humans , Hydrogen Bonding , Models, Molecular , Molecular Conformation , Molecular Structure , Thermodynamics
17.
Chem Res Toxicol ; 18(11): 1737-47, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16300383

ABSTRACT

Equilenin, an important component of a widely prescribed hormone replacement formulation for postmenopausal women, is metabolized by mammalian P450 enzymes to the catechol 4-hydroxyequilenin (4-OHEN). The oxidized o-quinone derivative of 4-OHEN is known to form cyclic covalent adducts with DNA [Bolton, J. (1998) Chem. Res. Toxicol. 11, 1113] in vitro and in vivo. The characteristics of 4-OHEN-DNA adduct formation were investigated with the oligonucleotides 5'-d(CCATCGCTACC) (I), its complementary strand 5'-d(GGTAGCGATGG) (II), one rich in C and the other in G, and the duplexes I.II. The identities of the modified bases were elucidated in terms of four stereoisomeric 4-OHEN-2'-deoxynucleoside standards described earlier [Shen et al. (2001) Chem. Res. Toxicol. 11, 94; Embrechts et al. J. Mass Spectrom. 36, 317). The reactions of 4-OHEN with C are favored overwhelmingly in both single-stranded I and II with no guanine adducts observed in either case, and only minor proportions of A adducts were detected in sequence II. However, guanine adducts are observed in oligonucleotides that contain only G and unreactive T residues. The relative levels of cyclic covalent adducts observed in single-stranded I, II, and duplex I.II are approximately 54:21:5, with only the end C groups in I modified in the I.II duplex. When 4-OHEN is reacted with calf thymus DNA, the reaction yield of cyclic adducts is more than approximately 10(3)-fold lower than in I. The cyclic 4-OHEN adducts lead to a pronounced thermal destabilization of duplexes I.II. Overall, cyclic adduct formation is markedly dependent on the sequence context and secondary structure of the DNA. The latter effect is attributed to the poor accessibilities of 4-OHEN to the reactive nucleotide Watson-Crick hydrogen-bonding interface in the interior of the duplex. In the single-stranded oligonucleotides I and II, the strikingly different selectivities of adduct formation are attributed to the formation of noncovalent preassociation complexes that favor reaction geometries with C, rather than with A or G. Finally, the levels of several typical biomarkers of oxidative DNA damage (including 8-oxo-2'-deoxyguanosine) are formed in I in aqueous solutions with a yield at least 10 times smaller than the yield of cyclic 4-OHEN-dC adducts under identical reaction conditions.


Subject(s)
DNA Adducts/metabolism , DNA/chemistry , Equilenin/analogs & derivatives , Base Sequence , Chromatography, High Pressure Liquid , Circular Dichroism , DNA/metabolism , DNA Damage , Equilenin/chemistry , Equilenin/metabolism , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oxidation-Reduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Chem Biol Interact ; 152(2-3): 151-63, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15840388

ABSTRACT

Catechol O-methyltransferase (COMT) plays an important role in the inactivation of biologically active and toxic catechols. This enzyme is genetically polymorphic with a wild type and a variant form. Numerous epidemiological studies have shown that the variant form is associated with an increased risk of developing estrogen-associated cancers and a wide spectrum of mental disorders. There are seven cysteine residues in human S-COMT, all of which exist as free thiols and are susceptible to electrophilic attack and/or oxidative damage leading to enzyme inactivation. Here, the seven cysteine residues were systematically replaced by alanine residues by means of site-directed mutagenesis. The native forms and cysteine/alanine mutants were assayed for enzymatic activity, thermal stability, methylation regioselectivity, and reactivity of cysteine residues to thiol reagent. Our data showed that although there is only one encoding base difference between these two COMT forms, this difference might induce structural changes in the local area surrounding some cysteine residues, which might further contribute to the different roles they might play in enzymatic activity, and to the different susceptibility to enzyme inactivation.


Subject(s)
Catechol O-Methyltransferase/metabolism , Cysteine/metabolism , Equilenin/analogs & derivatives , Methionine/metabolism , Valine/metabolism , Catechol O-Methyltransferase/chemistry , Catechol O-Methyltransferase/genetics , Cysteine/genetics , Equilenin/metabolism , Humans , Kinetics , Methionine/genetics , Mutagenesis, Site-Directed , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Valine/genetics
19.
FEBS J ; 272(8): 1999-2011, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15819891

ABSTRACT

A structural motif called the small exterior hydrophobic cluster (SEHC) has been proposed to explain the stabilizing effect mediated by solvent-exposed hydrophobic residues; however, little is known about its biological roles. Unusually, in Delta(5)-3-ketosteroid isomerase from Pseudomonas putida biotype B (KSI-PI) Trp92 is exposed to solvent on the protein surface, forming a SEHC with the side-chains of Leu125 and Val127. In order to identify the role of the SEHC in KSI-PI, mutants of those amino acids associated with the SEHC were prepared. The W92A, L125A/V127A, and W92A/L125A/V127A mutations largely decreased the conformational stability, while the L125F/V127F mutation slightly increased the stability, indicating that hydrophobic packing by the SEHC is important in maintaining stability. The crystal structure of W92A revealed that the decreased stability caused by the removal of the bulky side-chain of Trp92 could be attributed to the destabilization of the surface hydrophobic layer consisting of a solvent-exposed beta-sheet. Consistent with the structural data, the binding affinities for three different steroids showed that the surface hydrophobic layer stabilized by SEHC is required for KSI-PI to efficiently recognize hydrophobic steroids. Unfolding kinetics based on analysis of the Phi(U) value also indicated that the SEHC in the native state was resistant to the unfolding process, despite its solvent-exposed site. Taken together, our results demonstrate that the SEHC plays a key role in the structural integrity that is needed for KSI-PI to stabilize the hydrophobic surface conformation and thereby contributes both to the overall conformational stability and to the binding of hydrophobic steroids in water solution.


Subject(s)
Pseudomonas putida/classification , Pseudomonas putida/enzymology , Steroid Isomerases/chemistry , Steroid Isomerases/metabolism , Steroids/metabolism , Amino Acid Motifs , Catalysis , Enzyme Stability , Equilenin/metabolism , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Mutation/genetics , Nandrolone/metabolism , Protein Binding , Protein Conformation , Protein Denaturation , Protein Folding , Pseudomonas putida/genetics , Steroid Isomerases/genetics
20.
Biochem J ; 375(Pt 2): 297-305, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12852789

ABSTRACT

KSI (ketosteroid isomerase) from Comamonas testosteroni is a homodimeric enzyme that catalyses the allylic isomerization of Delta5-3-ketosteroids to their conjugated Delta4-isomers at a reaction rate equivalent to the diffusion-controlled limit. Based on the structural analysis of KSI at a high resolution, the conserved cis-Pro39 residue was proposed to be involved in the proper positioning of Asp38, a critical catalytic residue, since the residue was found not only to be structurally associated with Asp38, but also to confer a structural rigidity on the local active-site geometry consisting of Asp38, Pro39, Val40, Gly41 and Ser42 at the flexible loop between b-strands B1 and B2. In order to investigate the structural role of the conserved cis-Pro39 residue near the active site of KSI, Pro39 was replaced with alanine or glycine. The free energy of activation for the P39A and P39G mutants increased by 10.5 and 16.7 kJ/mol (2.5 and 4.0 kcal/mol) respectively, while DG(U)H2O (the free-energy change for unfolding in the absence of urea at 25.00+/-0.02 degrees C) decreased by 31.0 and 35.6 kJ/mol (7.4 and 8.5 kcal/mol) respectively, compared with the wild-type enzyme. The crystal structure of the P39A mutant in complex with d-equilenin [d-1,3,5(10),6,8-estrapentaen-3-ol-17-one], a reaction intermediate analogue, determined at 2.3 A (0.23 nm) resolution revealed that the P39A mutation significantly disrupted the proper orientations of both d-equilenin and Asp38, as well as the local active-site geometry near Asp38, which resulted in substantial decreases in the activity and stability of KSI. Upon binding 1-anilinonaphthalene-8-sulphonic acid, the fluorescence intensities of the P39A and P39G mutants were increased drastically, with maximum wavelengths blue-shifted upon binding, indicating that the mutations might alter the hydrophobic active site of KSI. Taken together, our results demonstrate that the conserved cis-Pro39 residue plays a crucial role in the proper positioning of the critical catalytic base Asp38 and in the structural integrity of the active site in KSI.


Subject(s)
Aspartic Acid/chemistry , Comamonas testosteroni/enzymology , Proline/chemistry , Steroid Isomerases/chemistry , Androstenedione/chemistry , Androstenedione/metabolism , Aspartic Acid/genetics , Aspartic Acid/metabolism , Binding Sites/genetics , Catalysis , Catalytic Domain , Conserved Sequence/genetics , Crystallography, X-Ray , Enzyme Stability , Equilenin/chemistry , Equilenin/metabolism , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Mutation , Proline/genetics , Proline/metabolism , Protein Binding , Protein Folding , Protein Structure, Tertiary , Steroid Isomerases/genetics , Steroid Isomerases/metabolism , Structure-Activity Relationship , Substrate Specificity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...