Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Molecules ; 26(7)2021 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-33916785

RESUMO

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.


Assuntos
Inibidores de Catecol O-Metiltransferase/química , Inibidores de Catecol O-Metiltransferase/farmacologia , Catecol O-Metiltransferase/metabolismo , Estrogênios de Catecol/metabolismo , Masoprocol/metabolismo , Masoprocol/farmacologia , Mutagênicos/toxicidade , Sítios de Ligação , Morte Celular/efeitos dos fármacos , Dano ao DNA , Estrogênios de Catecol/química , Estrogênios de Catecol/farmacologia , Humanos , Células MCF-7 , Masoprocol/química , Metilação , Simulação de Acoplamento Molecular , Proteínas Recombinantes/metabolismo , Especificidade por Substrato/efeitos dos fármacos
2.
Int Immunopharmacol ; 86: 106712, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32585610

RESUMO

Depression has been commonly associated with type 1 diabetes (T1D) and insulin covalently modified with catecholestrogens (CEs) was found in serum of these T1D patients. This study aimed to know whether depression link to higher antibodies against estrogenized insulin in T1D. ELISA (direct binding and competition) and quantitative precipitin titration were used to detect antibodies and their affinities against estrogenized insulin in the serum of 66 depressed T1D (DT1D) patients (out of 110 T1D) and 41 control subjects. Antibodies from DT1D patients showed high binding specificity to estrogenized insulin (2-hydroestradiol-insulin; 2-OHE2-Ins) in comparison to overall T1D patients (p < 0.05) or control subjects (p < 0.001). However, T1D sera demonstrate high recognition to 2-OHE2-Ins as compared to Ins (p < 0.05) or 2-OHE2 (p < 0.001). The affinity of antibodies from DT1D and T1D patients was 1.32 × 10-7 M and 1.43 × 10-7 M, respectively. Depression linked to higher antibodies production against estrogenized insulin in T1D. Furthermore, depression in T1D generates inflammatory conditions that further increased antibodies production in T1D patients.


Assuntos
Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Depressão/imunologia , Diabetes Mellitus Tipo 1/imunologia , Estrogênios de Catecol/imunologia , Animais , Autoanticorpos/química , Autoanticorpos/isolamento & purificação , Depressão/sangue , Depressão/complicações , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/complicações , Ensaio de Imunoadsorção Enzimática , Estrogênios de Catecol/sangue , Estrogênios de Catecol/química , Feminino , Humanos , Fatores Imunológicos/sangue , Resistência à Insulina/imunologia , Masculino , Pessoa de Meia-Idade
3.
Life Sci ; 256: 117910, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32504753

RESUMO

AIMS: Insulin (Ins) covalently modified by catecholestrogens (CEs) was commonly found in diabetic patients who have developed insulin resistance. Estrogenization of insulin altered its molecular function and effect carbohydrates metabolisms in these patients. Insulin resistance is a common phenomenon in diabetes but the exact mechanism remains unknown. In this study, binding specificity and affinity of autoantibodies against estrogenized insulin (4-hydroxyestradiol-insulin; 4-OHE2-Ins) were assayed in the serum of type 1 diabetes (T1D) patients in order to explain the phenomena behind insulin resistance. MATERIALS AND METHODS: Specificity and affinity of autoantibodies from the sera of 66 T1D patients and 41 controls were analyzed by direct binding, competition ELISA and quantitative precipitin titration. Insulin was also estimated in the serum of T1D patients by ELISA. KEY FINDING: Estrogenized insulin (4-OHE2-Ins) exhibited high affinity and specificity to T1D autoantibodies in comparison to Ins (p < .05) or 4-OHE2 (p < .001). Estrogenization of insulin alters its interaction with the insulin receptor (IR). The affinity constant of 4-OHE2-Ins with the T1D autoantibodies was found to be 1.41 × 10-7 M. SIGNIFICANCE: Estrogenization of insulin by catecholestrogen makes these molecules highly antigenic and produced high-affinity autoantibodies in T1D patients. As a result, patients develop insulin resistance and presented this molecule as a potential biomarker for T1D.


Assuntos
Autoanticorpos/imunologia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Estrogênios de Catecol/química , Hipoglicemiantes/química , Insulina/química , Adulto , Autoanticorpos/metabolismo , Biomarcadores/metabolismo , Glicemia/análise , Coleta de Amostras Sanguíneas , Proposta de Concorrência , Ensaio de Imunoadsorção Enzimática , Estrogênios de Catecol/uso terapêutico , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Resistência à Insulina , Masculino , Pessoa de Meia-Idade , Receptor de Insulina/imunologia , Receptor de Insulina/metabolismo , Sensibilidade e Especificidade
4.
Anal Chem ; 91(24): 15922-15931, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31794208

RESUMO

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.


Assuntos
Estrogênios de Catecol/química , Espectrometria de Massas/métodos , Peptídeos/análise , Albumina Sérica/química , Cromatografia Líquida de Alta Pressão , Quimotripsina/metabolismo , Estrogênios de Catecol/metabolismo , Humanos , Espectrometria de Massas/normas , Nanotecnologia , Peptídeos/metabolismo , Peptídeos/normas , Padrões de Referência , Albumina Sérica/metabolismo , Tripsina/metabolismo
5.
Horm Cancer ; 10(2-3): 77-88, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30877616

RESUMO

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


Assuntos
Androgênios/efeitos adversos , Carcinogênese , Estrogênios/efeitos adversos , Neoplasias da Próstata/induzido quimicamente , Animais , Carcinoma , Adutos de DNA , Dano ao DNA , Di-Hidrotestosterona/metabolismo , Estradiol/metabolismo , Estrogênios de Catecol/química , Guanosina/análogos & derivados , Guanosina/farmacologia , Humanos , Incidência , Masculino , Próstata , Ratos , Receptores de Estrogênio/metabolismo , Testosterona/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-28666224

RESUMO

15α-Hydroxyestrogens (15α-OHEs) are metabolites of the female hormone estradiol. In this study, to discover physiological markers that can be utilized for monitoring fetal conditions and estrogen-induced cancers, we established a method for quantifying 15α-OHEs in rat urine via immunoaffinity column extraction and HPLC-electrochemical detection, and detected 15α-OHEs in urine obtained male rats treated with estradiol. Notably, the standard curves for quantification obtained using the column were linear over a range of 0.5-50ng 15α-OHEs. The accuracy of the analytical method with cleanup was 97-109% for the three kinds of 15α-OHEs examined, and the intra-assay precision of the measured values had a coefficient of variation of ≤20.6%. Therefore, the theoretical limit of quantification was 0.5ng. However, the actual measured values obtained from the urine of male rats indicated that the detection limits were 0.425, 0.103, and 0.047ng for estetrol, 15α-hydroxyestradiol, and 15α-hydroxyestrone, respectively. Our newly established method for measuring 15α-OHE concentrations in urine could facilitate characterization of the in vivo metabolic profile of 15α-OHEs in mammals under various physiological conditions, which could comprise clinical markers for monitoring human fetal health conditions in mammals.


Assuntos
Cromatografia de Afinidade/métodos , Estrogênios de Catecol/urina , Animais , Anticorpos Imobilizados/química , Anticorpos Imobilizados/metabolismo , Estrogênios de Catecol/química , Estrogênios de Catecol/metabolismo , Feminino , Limite de Detecção , Modelos Lineares , Masculino , Ratos , Reprodutibilidade dos Testes
7.
Chem Res Toxicol ; 30(7): 1448-1462, 2017 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-28616971

RESUMO

In animal models, estrogens are complete carcinogens in certain target sites. 4-Hydroxyestradiol (4-OH-E2), an endogenous metabolite of 17ß-estradiol (E2), is known to have prominent estrogenic activity plus potential genotoxicity and mutagenicity. We report here our finding that 4-OH-E2 does not induce pituitary tumors in ACI female rats, whereas E2 produces 100% pituitary tumor incidence. To probe the mechanism, we conducted a short-term animal experiment to compare the proliferative effect of 4-OH-E2 in several organs. We found that, whereas 4-OH-E2 had little ability to stimulate pituitary cell proliferation in ovariectomized female rats, it strongly stimulates cell proliferation in certain brain regions of these animals. Further, when we used in vitro cultured rat pituitary tumor cells as models, we found that 4-OH-E2 has similar efficacy as E2 in stimulating cell proliferation, but its potency is approximately 3 orders of magnitude lower than that of E2. Moreover, we found that the pituitary tumor cells have the ability to selectively metabolize 4-OH-E2 (but not E2) with ultrahigh efficiency. Additional analysis revealed that the rat pituitary expresses a membrane-bound catechol-O-methyltransferase that has an ultralow Km value (in nM range) for catechol estrogens. On the basis of these observations, it is concluded that rapid metabolic disposition of 4-OH-E2 through enzymatic O-methylation in rat anterior pituitary cells largely contributes to its apparent lack of cell proliferative and tumorigenic effects in this target site.


Assuntos
Catecol O-Metiltransferase/metabolismo , Estrogênios de Catecol/farmacologia , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Animais , Biocatálise , Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Estrogênios de Catecol/química , Feminino , Humanos , Metilação , Adeno-Hipófise/citologia , Adeno-Hipófise/enzimologia , Ratos , Ratos Endogâmicos ACI , Células Tumorais Cultivadas
8.
Sci Rep ; 6: 28804, 2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27353345

RESUMO

Proteins, covalently modified by catechol estrogens (CEs), were identified recently from the blood serum of diabetic patients and referred to as estrogenized proteins. Estrogenization of circulating insulin may occur and affect its molecular functioning. Here, the chemical reactivity of CEs towards specific amino acid residues of proteins and the structural and functional changes induced by the estrogenization of insulin were studied using cyclic voltammetry, liquid chromatography-mass spectrometry, circular dichroism spectroscopy, molecular modeling, and bioassays. Our results indicate that CEs, namely, 2- and 4-hydroxyl estrogens, were thermodynamically and kinetically more reactive than the catechol moiety. Upon co-incubation, intact insulin formed a substantial number of adducts with one or multiple CEs via covalent conjugation at its Cys 7 in the A or B chain, as well as at His10 or Lys29 in the B chain. Such conjugation was coupled with the cleavage of inter-chain disulfide linkages. Estrogenization on these sites may block the receptor-binding pockets of insulin. Insulin signaling and glucose uptake levels were lower in MCF-7 cells treated with modified insulin than in cells treated with native insulin. Taken together, our findings demonstrate that insulin molecules are susceptible to active estrogenization, and that such modification may alter the action of insulin.


Assuntos
Estrogênios de Catecol/química , Insulina/análogos & derivados , Insulina/química , Sequência de Aminoácidos , Glucose/metabolismo , Humanos , Insulina/fisiologia , Cinética , Células MCF-7 , Oxirredução , Transdução de Sinais
9.
Toxicol Sci ; 148(2): 433-42, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26342215

RESUMO

Protein adducts covalently modified by catechol estrogens (CEs), referred to as estrogenized proteins, are potential biomarkers for estrogen homeostasis or exposure to environmental toxicants. However, serum proteins endogenously modified by CEs and the modification sites remain elusive. In this study, liquid chromatography-mass spectrometry (LC-MS)-based shotgun proteomics is applied to identify site-specific protein estrogenization in human blood via a systematic approach and stringent validation. We showed CEs, namely 2- and 4-hydroxyl estrogens which are regarded as biomarkers for estrogen homeostasis, form covalent bonds with proteins, mainly via side chain Cys, Lys, or His residue. Estrogenization of purified human serum albumin (HSA) and immunoglobulin G (IgG) at specific sites was achieved by co-incubation and used as the standards to confirm the identified estrogenization in serum proteins. Based on a database search, estrogenized peptides derived from serum proteins in patient blood were identified; endogenous estrogenization of HSA and IgG-1 at multiple sites were confirmed as compared to the standards. Based on a test using Ellman's reagent, estrogenization produced stable products and irreversibly abolished the reactivity of Cys34-HSA, which is the most important antioxidant and nitric oxide carrier in blood. Given the importance of estrogen metabolism in environmental toxicology, further exploration of estrogenized proteins is warranted for biomarker discovery and/or new mechanisms in disease process.


Assuntos
Estradiol/análogos & derivados , Estrogênios de Catecol/sangue , Imunoglobulina G/metabolismo , Albumina Sérica/metabolismo , Sítios de Ligação , Cromatografia Líquida , Bases de Dados de Proteínas , Estradiol/sangue , Estradiol/química , Estrogênios de Catecol/química , Feminino , Humanos , Imunoglobulina G/química , Pessoa de Meia-Idade , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Proteômica/métodos , Albumina Sérica/química , Albumina Sérica Humana , Espectrometria de Massas em Tandem
10.
Mol Cell Endocrinol ; 400: 71-7, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25449419

RESUMO

Catechol estrogens, such as 4-hydroxyestradiol (4-OHE2), are estrogen metabolites that form DNA adducts and may induce mutations and subsequent cell transformation in mammary cells; however, little is known about their roles in endometrial carcinogenesis. Furthermore, it remains unclear whether 4-OHE2 is able to induce DNA damage on specific genes involved in carcinogenesis or a 'pro'-mutation status such as microsatellite instability (MSI). Therefore, we modified terminal transferase-dependent PCR by the application of a capillary sequencer to detect DNA damage at the single base level. Using this method, we demonstrated that 4-OHE2 directly induced DNA damage on codon 130/131 in exon 5 of PTEN, which is a mutation hot spot for PTEN in endometrial carcinoma. Whereas, both estradiol and 4-OHE2 treatment did not affect MSI status in immortalized endometrial glandular cells. 4-OHE2 might contribute to endometrial carcinogenesis by inducing PTEN mutation on codon 130/131.


Assuntos
Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Células Epiteliais/efeitos dos fármacos , Estrogênios de Catecol/farmacologia , PTEN Fosfo-Hidrolase/genética , Mutação Puntual , Sequência de Bases , Linhagem Celular Transformada , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Códon , Adutos de DNA/química , Dano ao DNA , Análise Mutacional de DNA , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Estrogênios de Catecol/química , Éxons , Feminino , Expressão Gênica , Humanos , Instabilidade de Microssatélites/efeitos dos fármacos , Dados de Sequência Molecular , PTEN Fosfo-Hidrolase/metabolismo , Reação em Cadeia da Polimerase/métodos
11.
Free Radic Biol Med ; 65: 1447-1454, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23928335

RESUMO

Homocysteine (HCys), a sulfur-containing amino acid, is formed during the metabolism of methionine. An imbalance between the rate of production and the use of HCys during methionine metabolism can result in an increase in the plasma and urinary levels of HCys. HCys has been shown to be toxic to vascular endothelial cells through several pathways. Many earlier clinical studies have revealed an association between plasma HCys and cardiovascular and other diseases. In contrast, estrogens are suggested to lower the risk of cardiovascular disease. Several studies indicate that estrogen metabolites could be responsible for cardiovascular protection. It has been demonstrated that electrophilic estrogen quinones, E1(E2)-2,3-Q and E1(E2)-3,4-Q, can alkylate DNA as well as form conjugates with glutathione. I hypothesize that estrogen quinones generated in situ by oxidative enzymes, metal ions, or molecular oxygen can interact with HCys to form conjugates. This in turn could lower the levels of toxic HCys as well as quenching the reactive estrogen quinones, resulting in cardiovascular protective effects. To test the feasibility of a protective estrogen-HCys pathway, estrogen quinones were treated with HCys. Tandem mass spectrometry analysis of the assay mixture shows the formation of estrogen-HCys conjugates. Furthermore, incubation of catechol estrogens with myeloperoxidase (MPO) in the presence of HCys resulted in the formation of respective estrogen-HCys conjugates. The identities of estrogen-HCys conjugates in MPO assay extracts were confirmed by comparing them to pure synthesized estrogen-HCys standards. I propose that through conjugation estrogens could chemically regulate HCys levels; moreover these conjugates could be used as potential biomarkers in determining health.


Assuntos
Congêneres do Estradiol/síntese química , Estrogênios de Catecol/química , Estrogênios/química , Homocisteína/química , Quinonas/química , Alquilação , Cardiotônicos/química , Doenças Cardiovasculares , DNA/química , Congêneres do Estradiol/química , Glutationa/química , Homocisteína/biossíntese , Homocisteína/sangue , Espectrometria de Massas , Oxirredução , Peroxidase/metabolismo
12.
Biochimie ; 95(2): 329-35, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23069385

RESUMO

Catecholestrogens [4-hydroxyestradiol (4-OHE(2))] have been implicated in human carcinogenesis, although the mechanism remains unestablished. In this study pUC 18 plasmid DNA was modified with 4-OHE(2) and nitric oxide (NO). The modification induced in native DNA exhibited hyperchromicity, single strand breaks, damage to restriction sites, modification of bases, decrease in Tm and change in ellipticity. Modified DNA was found to be highly immunogenic in experimental animal, eliciting high titer antibodies. Circulating cancer autoantibodies showed preferable recognition of 4-OHE(2)-NO-DNA over native form (p < 0.001) and the oxidative epitopes on the DNA isolates from cancer patients were immunochemically detected by using experimentally induced anti-4-OHE(2)-NO-DNA antibodies as a probe. Preferential recognition of 4-OHE(2)-NO-DNA by cancer autoantibodies coupled with enhanced binding of induced antibodies to DNA isolated from cancer patients is an indicative of oxidative stress induced DNA damage in cancer. Possible involvement of unique epitopes on modified DNA in cancer autoantibody induction has been suggested.


Assuntos
Anticorpos Antinucleares/imunologia , DNA de Neoplasias/imunologia , Estrogênios de Catecol/química , Neoplasias/sangue , Adulto , Anticorpos Antinucleares/química , Estudos de Casos e Controles , Dano ao DNA , DNA de Neoplasias/química , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Escherichia coli/química , Humanos , Linfócitos/química , Linfócitos/patologia , Pessoa de Meia-Idade , Neoplasias/imunologia , Neoplasias/patologia , Óxido Nítrico/química , Conformação de Ácido Nucleico/efeitos dos fármacos , Estresse Oxidativo , Plasmídeos/química
13.
Anal Bioanal Chem ; 399(1): 519-24, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21058011

RESUMO

Catechol estrogen-derived DNA adducts are formed as a result of the reaction of catechol estrogen metabolites (e.g., catechol estrogen quinones) with DNA to form depurinating adducts. Developing a new methodology for the detection of various DNA adducts is essential for medical diagnostics, and to this end, we demonstrate the applicability of on-chip capillary electrophoresis with an integrated electrochemical system for the separation and amperometric detection of various catechol estrogen-derived DNA adducts. A hybrid PDMS/glass microchip with in-channel amperometric detection interfaced with in situ palladium decoupler is utilized and presented. The influence of buffer additives along with the effect of the separation voltage on the resolving power of the microchip is discussed. Calibration plots were constructed in the range 0.4-10 µM with r(2) ≥ 0.999, and detection limits in the attomole range are reported. These results suggest that on-chip analysis is applicable for analyzing various DNA adducts as potential biomarkers for future medical diagnostics.


Assuntos
Adutos de DNA/isolamento & purificação , Eletroforese Capilar/métodos , Estrogênios de Catecol/isolamento & purificação , Técnicas Analíticas Microfluídicas/métodos , Adutos de DNA/química , Eletroquímica , Eletroforese Capilar/instrumentação , Estrogênios de Catecol/química , Humanos , Técnicas Analíticas Microfluídicas/instrumentação
14.
Chem Res Toxicol ; 23(8): 1365-73, 2010 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-20509668

RESUMO

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.


Assuntos
Dano ao DNA , Desoxiguanosina/análogos & derivados , Equilenina/análogos & derivados , Estrogênios de Catecol/metabolismo , Cavalos , Espécies Reativas de Oxigênio/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Bovinos , Linhagem Celular Tumoral , Quelantes/farmacologia , Cobre/química , Cobre/metabolismo , DNA/metabolismo , DNA de Neoplasias/metabolismo , Desoxiguanosina/metabolismo , Equilenina/química , Equilenina/metabolismo , Estrogênios de Catecol/química , Estrogênios de Catecol/farmacologia , Sequestradores de Radicais Livres/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Hidroxiestronas/química , Hidroxiestronas/metabolismo , Estrutura Molecular , NADP/química , NADP/metabolismo , Oxirredução/efeitos dos fármacos , Relação Estrutura-Atividade
15.
Molecules ; 15(4): 2347-56, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20428047

RESUMO

Benzeneboronate of catecholic carboxyl methyl esters, N-acetyldopamine, coumarin and catechol estrogens were prepared as crystalline derivatives in high yield. Related catechol compounds with extra polar functional group(s) (OH, NH2) do not form or only partially form unstable cyclic boronate derivatives.


Assuntos
Ácidos Borônicos/química , Cumarínicos/química , Dopamina/análogos & derivados , Estrogênios de Catecol/química , Cristalização , Dopamina/química , Estabilidade de Medicamentos
16.
J Steroid Biochem Mol Biol ; 113(1-2): 65-74, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095062

RESUMO

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.


Assuntos
Inibidores de Catecol O-Metiltransferase , Café/química , Estrogênios de Catecol/metabolismo , Flavonoides/farmacologia , Fenóis/farmacologia , Adulto , Biocatálise/efeitos dos fármacos , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacologia , Catecol O-Metiltransferase/química , Ácido Clorogênico/química , Ácido Clorogênico/farmacologia , Biologia Computacional , Citosol/efeitos dos fármacos , Citosol/enzimologia , Estrogênios de Catecol/química , Feminino , Flavonoides/química , Humanos , Concentração Inibidora 50 , Cinética , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Metilação/efeitos dos fármacos , Modelos Moleculares , Fenóis/química , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/química , Álcool Feniletílico/farmacologia , Placenta/citologia , Placenta/efeitos dos fármacos , Placenta/enzimologia , Polifenóis , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína
17.
J Steroid Biochem Mol Biol ; 110(1-2): 10-7, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18372170

RESUMO

High levels of endogenous estrogens are associated with increased risks of breast cancer. Estrogen levels are mainly increased by the activity of the aromatase enzyme and reduced by oxidative/conjugative metabolic pathways. In this paper, we demonstrate for the first time that catechol estrogen metabolites are potent aromatase inhibitors, thus establishing a link between aromatase activity and the processes involved in estrogen metabolism. In particular, the anti-aromatase activity of a set of natural hydroxyl and methoxyl estrogen metabolites was investigated using biochemical methods and subsequently compared with the anti-aromatase potency of estradiol and two reference aromatase inhibitors. Catechol estrogens proved to be strong inhibitors with an anti-aromatase potency two orders of magnitude higher than estradiol. A competitive inhibition mechanism was found for the most potent molecule, 2-hydroxyestradiol (2-OHE(2)) and a rational model identifying the interaction determinants of the metabolites with the enzyme is proposed based on ab initio quantum-mechanical calculations. A strong relationship between activity and electrostatic properties was found for catechol estrogens. Moreover, our results suggest that natural catechol estrogens may be involved in the control mechanisms of estrogen production.


Assuntos
Inibidores da Aromatase/farmacologia , Aromatase/metabolismo , Estrogênios de Catecol/farmacologia , Inibidores da Aromatase/química , Catálise/efeitos dos fármacos , Estradiol/química , Estradiol/farmacologia , Estrogênios/química , Estrogênios/farmacologia , Estrogênios de Catecol/química , Humanos , Cinética , Estrutura Molecular
18.
Scand J Immunol ; 64(6): 677-83, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17083625

RESUMO

Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease in which anti-double-stranded DNA antibody is a classic autoantibody that characterizes SLE. A role for oestrogens in the pathogenesis of SLE has been suspected for many years but the exact patho-aetiology remains elusive. In this study, the binding of SLE autoantibodies with native and 4-OHE(2)-NO-modified plasmid DNA were assessed. Binding specificity of antibodies was analysed by direct binding and inhibition enzyme-linked immunosorbent assay, quantitative precipitin titration and gel retardation assay. Anti-DNA IgG from SLE sera, purified on Protein A-Agarose matrix, exhibited increased recognition of 4-OHE(2)-NO-DNA than native DNA (P < 0.001). Gel retardation assay further substantiated the enhanced recognition of modified DNA by anti-DNA autoantibodies. The affinity of anti-DNA antibodies for modified polymer was found to be high as calculated by using Langmuir plot. DNA modified by 4-OHE(2)-NO presents unique neo-epitopes that might be one of the factor in antigen-driven induction of SLE autoantibodies.


Assuntos
Anticorpos Antinucleares/imunologia , Especificidade de Anticorpos , Autoanticorpos/imunologia , DNA/imunologia , Estrogênios de Catecol/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Anticorpos Antinucleares/sangue , Autoanticorpos/sangue , DNA/química , Ensaio de Desvio de Mobilidade Eletroforética , Estradiol/análogos & derivados , Estradiol/química , Estradiol/imunologia , Estrogênios de Catecol/química , Humanos
19.
Biochim Biophys Acta ; 1766(1): 63-78, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16675129

RESUMO

Exposure to estrogens is associated with increased risk of breast and other types of human cancer. Estrogens are converted to metabolites, particularly the catechol estrogen-3,4-quinones (CE-3,4-Q), that can react with DNA to form depurinating adducts. These adducts are released from DNA to generate apurinic sites. Error-prone base excision repair of this damage may lead to the mutations that can initiate breast, prostate and other types of cancer. The reaction of CE-3,4-Q with DNA forms the depurinating adducts 4-hydroxyestrone(estradiol) [4-OHE1(E2)-1-N3Ade and 4-OHE1(E2)-1-N7Gua. These two adducts constitute more than 99% of the total DNA adducts formed. Increased levels of these quinones and their reaction with DNA occur when estrogen metabolism is unbalanced. Such an imbalance is the result of overexpression of estrogen activating enzymes and/or deficient expression of the deactivating (protective) enzymes. This unbalanced metabolism has been observed in breast biopsy tissue from women with breast cancer, compared to control women. Recently, the depurinating adduct 4-OHE1(E2)-1-N3Ade has been detected in the urine of prostate cancer patients, but not in urine from healthy men. Mutagenesis by CE-3,4-Q has been approached from two different perspectives: one is mutagenic activity in the lacI reporter gene in Fisher 344 rats and the other is study of the reporter Harvey-ras gene in mouse skin and rat mammary gland. A-->G and G-->A mutations have been observed in the mammary tissue of rats implanted with the CE-3,4-Q precursor, 4-OHE2. Mutations have also been observed in the Harvey-ras gene in mouse skin and rat mammary gland within 6-12 h after treatment with E2-3,4-Q, suggesting that these mutations arise by error-prone base excision repair of the apurinic sites generated by the depurinating adducts. Treatment of MCF-10F cells, which are estrogen receptor-alpha-negative immortalized human breast epithelial cells, with E2, 4-OHE2 or 2-OHE2 induces their neoplastic transformation in vitro, even in the presence of the antiestrogen ICI-182,780. This suggests that transformation is independent of the estrogen receptor. The transformed cells exhibit specific mutations in several genes. Poorly differentiated adenocarcinomas develop when aggressively transformed MCF-10F cells are selected and injected into severe combined immune depressed (SCID) mice. These results represent the first in vitro/in vivo model of estrogen-induced carcinogenesis in human breast epithelial cells. In other studies, the development of mammary tumors in estrogen receptor-alpha knockout mice expressing the Wnt-1 oncogene (ERKO/Wnt-1) provides direct evidence that estrogens may cause breast cancer through a genotoxic, non-estrogen receptor-alpha-mediated mechanism. In summary, this evidence strongly indicates that estrogens can become endogenous tumor initiators when CE-3,4-Q react with DNA to form specific depurinating adducts. Initiated cells may be promoted by a number of processes, including hormone receptor stimulated proliferation. These results lay the groundwork for assessing risk and preventing disease.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/etiologia , Adutos de DNA/química , Estrogênios de Catecol/química , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/prevenção & controle , Suscetibilidade a Doenças , Humanos , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/prevenção & controle
20.
Mol Pharmacol ; 69(6): 2084-92, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16517757

RESUMO

The superfamily of sulfotransferase (SULT) enzymes catalyzes the sulfate conjugation of several pharmacologically important endo- and xenobiotics. SULT1A1 catalyzes the sulfation of small planar phenols such as neurotransmitters, steroid hormones, acetaminophen, and p-nitrophenol (PNP). Genetic polymorphisms in the human SULT1A1 gene define three alleles, SULT1A1*1, *2, and *3. The enzyme activities of the SULT1A1 allozymes were studied with a variety of substrates, including PNP, 17beta-estradiol, 2-methoxyestradiol, catecholestrogens, the antiestrogen 4-hydroxytamoxifen (OHT), and dietary flavonoids. Using purified recombinant SULT1A1 protein, marked differences in *1, *2, and *3 activity toward every substrate studied were noted. Substrate inhibition was observed for most substrates. In general, the trend in V(max) estimates was *1 > *3 > *2; however, V(max)/K(m) estimate trends varied with substrate. In MCF-7 cells stably expressing either SULT1A1*1 or *2, the antiestrogenic response to OHT was found to be allele-specific: the cells expressing *2 exhibited a better antiproliferative response. The intracellular stability of the *1 and *2 allozymes was examined in insect as well as mammalian cells. The SULT1A1*2 protein had a shorter half-life than the *1 protein. In addition, the *2 protein was ubiquitinated to a greater extent than *1, suggesting increased degradation via a proteasome pathway. The results of this study suggest marked differences in activity of polymorphic SULT1A1 variants, including SULT1A1*3, toward a variety of substrates. These differences are potentially critical for understanding interindividual variability in drug response and toxicity, as well as cancer risk and incidence.


Assuntos
Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Polimorfismo Genético , 2-Metoxiestradiol , Animais , Arilsulfotransferase/genética , Células Cultivadas , Estradiol/análogos & derivados , Estradiol/química , Estrogênios de Catecol/química , Humanos , Nitrofenóis/química , Fenótipo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA