Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
FEBS Open Bio ; 11(4): 1084-1092, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33565718

RESUMO

P-glycoprotein (Pgp) detoxifies cells by exporting hundreds of chemically dissimilar hydrophobic and amphipathic compounds and is implicated in multidrug resistance (MDR) in the treatment of cancers. Photoaffinity labeling of plasma membrane vesicles of MDR CHO B30 cells with the anthracycline [125 I]-iodomycin, subsequent sequential cleavage with BNPS-skatol and endoproteinase Lys-C, and the Edman sequencing of the purified photoaffinity-labeled peptide identified the lysine residue at position 268 in the hamster Pgp primary sequence as the major photobinding site of iodomycin in CHO B30 cells. Lysine 268 is located adjacent to the cytosolic terminus of transmembrane 5. According to thermodynamic and kinetic analyses, this location should present the equilibrium binding site of ATP-free Pgp for daunomycin and iodomycin in B30 cells.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/química , Sítios de Ligação , Daunorrubicina/análogos & derivados , Lisina/química , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Daunorrubicina/química , Daunorrubicina/metabolismo , Humanos , Radioisótopos do Iodo/química , Radioisótopos do Iodo/metabolismo , Lisina/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/farmacologia , Peptídeos/química , Ligação Proteica , Relação Estrutura-Atividade
2.
Mol Cell Endocrinol ; 301(1-2): 216-24, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-19014997

RESUMO

Lowering local estradiol concentration by inhibition of the estradiol-synthesizing enzyme 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) has been proposed as a promising new therapeutic option to treat estrogen-dependent diseases like endometriosis and breast cancer. Based on a molecular modelling approach we designed and synthesized novel C15-substituted estrone derivatives. Subsequent biological evaluation revealed that potent inhibitors of human 17beta-HSD1 can be identified in this compound class. The best, compound 21, inhibited recombinant human 17beta-HSD1 with an IC50 of 10nM and had no effect on the activity of recombinant human 17beta-hydroxysteroid dehydrogenase type 2 (17beta-HSD2), the enzyme catalyzing estradiol inactivation. These properties were retained in a cell-based enzyme activity assays. In spite of the estrogen backbone compound 21 did not show estrogen receptor mediated effects in vitro or in vivo. In conclusion, estrone C15 derivative compound 21 can be regarded as a promising lead compound for further development as a 17beta-HSD1 inhibitor.


Assuntos
Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Estrona/análogos & derivados , Estrona/farmacologia , Animais , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Estradiol/química , Estrona/química , Humanos , Modelos Moleculares
3.
Mol Cell Endocrinol ; 301(1-2): 59-64, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-18762229

RESUMO

Endometriosis is a very common disease in pre-menopausal women, where defective metabolism of steroid hormones plays an important role in its development and promotion. In the present study, we have examined the expression of 11 estrogen and progesterone metabolizing enzymes and their corresponding receptors in samples of ovarian endometriomas and control endometrium. Expression analysis revealed significant up-regulation of enzymes involved in estradiol formation (aromatase, sulfatase and all reductive 17beta-hydroxysteroid dehydrogenases) and in progesterone inactivation (AKR1C1 and AKR1C3). Among the estrogen and progesterone receptors, ERalpha was down-regulated, ERbeta was up-regulated, and there was no significant difference in expression of progesterone receptors A and B (PRAB). Our data indicate that several enzymes of estrogen and progesterone metabolism are aberrantly expressed in endometriosis, which can lead to increased local levels of mitogenic estradiol and decreased levels of protective progesterone. Changes in estrogen receptor expression suggest that estradiol may also act via non-estrogen receptor-mediated pathways, while expression of progesterone receptors still needs further investigation.


Assuntos
Endometriose/metabolismo , Estrogênios/metabolismo , Ovário/metabolismo , Ovário/patologia , Progesterona/metabolismo , 17-Hidroxiesteroide Desidrogenases/metabolismo , 20-Hidroxiesteroide Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/metabolismo , Adulto , Membro C3 da Família 1 de alfa-Ceto Redutase , Aromatase/metabolismo , Endometriose/enzimologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Hidroxiprostaglandina Desidrogenases/metabolismo , Imuno-Histoquímica , Pessoa de Meia-Idade , Ovário/enzimologia , Receptores de Progesterona/metabolismo , Sulfotransferases
4.
Mol Cell Endocrinol ; 301(1-2): 205-11, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-18950679

RESUMO

17beta-Hydroxysteroid dehydrogenase type 1 (17beta-HSD1) catalyzes the transformation of estrone (E1) into the most potent estrogen, estradiol (E2), which stimulates cell proliferation and decreases apoptosis. 17beta-HSD1 is often strongly overexpressed in estrogen-dependent diseases (like breast cancer and endometriosis). Thus, this over expressed enzyme is a promising novel target for the development of selective inhibitors, which could be used as drugs for the treatment of these diseases. Using a structure- and ligand-based approach, a pharmacophore model was proposed and a new class of non-steroidal inhibitors of 17beta-HSD1 was designed. Enzyme inhibition was evaluated in vitro using the human enzyme. After identification of the 6-(3'-hydroxyphenyl)-2-naphthol scaffold 1, the potency of this class of inhibitors was further improved by substitution of the 1-position of the naphthalene ring by a phenyl group (compound 18, IC(50)=20nM). Compound 18 also showed a good selectivity toward 17beta-HSD2 and the estrogen receptors alpha and beta.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Modelos Moleculares , Naftalenos/química , Naftalenos/farmacologia , Humanos , Concentração Inibidora 50 , Relação Estrutura-Atividade
5.
Mol Cell Endocrinol ; 301(1-2): 158-62, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-19026716

RESUMO

Hydroxysteroid (17beta) dehydrogenase 1 (HSD17B1) catalyzes the reaction between the low active 17-ketosteroids and the highly active 17beta-hydroxysteroids. In the present study, we have generated transgenic (TG) mice expressing human (h) HSD17B1 under mouse mammary tumor virus (MMTV) promoter (MMTV-hHSD17B1TG mice). The MMTV-hHSD17B1TG mice were used to characterize HSD17B1 enzyme activity and properties of HSD17B1 inhibitor in vivo. Expression of the transgene was detected by enzyme activity and RT-PCR analysis. Increased HSD17B1 activity in the TG mice was detected in vivo by applying estrone as a substrate via an intravenous injection. The developed enzyme activity measurement was then applied to analyze the efficacy of HSD17B1 inhibitor in vivo. The results indicated that the MMTV-hHSD17B1TG mouse model is a valuable novel tool to test human HSD17B1 inhibition by various compounds in vivo. With the potent hHSD17B1 inhibitor compound tested, at highest an 85% and 33% inhibition of the enzyme activity in males and in females, respectively, was observed.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Animais , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Transgênicos , Modelos Animais , Regiões Promotoras Genéticas/genética , Transgenes
6.
J Med Chem ; 51(7): 2158-69, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18324762

RESUMO

Human 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) catalyzes the reduction of the weak estrogen estrone (E1) to the highly potent estradiol (E2). This reaction takes place in the target cell where the estrogenic effect is exerted via the estrogen receptor (ER). Estrogens, especially E2, are known to stimulate the proliferation of hormone-dependent diseases. 17beta-HSD1 is overexpressed in many breast tumors. Thus, it is an attractive target for the treatment of these diseases. Ligand- and structure-based drug design led to the discovery of novel, selective, and potent inhibitors of 17beta-HSD1. Phenyl-substituted bicyclic moieties were synthesized as mimics of the steroidal substrate. Computational methods were used to obtain insight into their interactions with the protein. Compound 5 turned out to be a highly potent inhibitor of 17beta-HSD1 showing good selectivity (17beta-HSD2, ERalpha and beta), medium cell permeation, reasonable metabolic stability (rat hepatic microsomes), and little inhibition of hepatic CYP enzymes.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Estrogênios/metabolismo , Naftalenos/síntese química , Naftalenos/farmacologia , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/enzimologia , Quinolinas/síntese química , Quinolinas/farmacologia , Animais , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Sítios de Ligação , Células CACO-2 , Simulação por Computador , Desenho de Fármacos , Inibidores Enzimáticos/química , Humanos , Ligação de Hidrogênio , Fígado/enzimologia , Masculino , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Estrutura Molecular , Naftalenos/química , Quinolinas/química , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Relação Estrutura-Atividade
7.
Mol Cell Biol ; 25(9): 3704-14, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831475

RESUMO

The transcriptional regulator Yin Yang 1 (YY1) controls many aspects of cell behavior and is essential for development. We analyzed the fate of YY1 during apoptosis and studied the functional consequences. We observed that this factor is rapidly translocated into the cell nucleus in response to various apoptotic stimuli, including activation of Fas, stimulation by tumor necrosis factor, and staurosporine and etoposide treatment. Furthermore, YY1 is cleaved by caspases in vitro and in vivo at two distinct sites, IATD(12)G and DDSD(119)G, resulting in the deletion of the first 119 amino acids early in the apoptotic process. This activity generates an N-terminally truncated YY1 fragment (YY1Delta119) that has lost its transactivation domain but retains its DNA binding domain. Indeed, YY1Delta119 is no longer able to stimulate gene transcription but interacts with DNA. YY1Delta119 but not the wild-type protein or the caspase-resistant mutant YY1D12A/D119A enhances Fas-induced apoptosis, suggesting that YY1 is involved in a positive feedback loop during apoptosis. Our findings provide evidence for a new mode of regulation of YY1 and define a novel aspect of the involvement of YY1 in the apoptotic process.


Assuntos
Apoptose/fisiologia , Caspases/fisiologia , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Diferenciação Celular , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/fisiologia , Proliferação de Células , Proteínas de Ligação a DNA/análise , Fatores de Ligação de DNA Eritroide Específicos , Humanos , Estrutura Terciária de Proteína , Fatores de Transcrição/análise , Transcrição Gênica , Fator de Transcrição YY1
8.
Endocrinology ; 147(11): 5333-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16916945

RESUMO

Hydroxysteroid (17-beta) dehydrogenase 1 (HSD17B1) catalyzes the conversion between estrone (E1) and estradiol (E2). The reaction is reversible in vitro, but the data in cultured cells suggest that E2 production is the predominant reaction in physiological conditions. However, the hypothesis has not been verified in vivo. In the present study, estrogen-dependent MCF-7 human breast cancer cells were stably transfected with an expression plasmid for human HSD17B1. The enzyme efficiently converted E1 to E2 and enhanced the estrogen-dependent growth of cultured MCF-7 cells in the presence of hormonally less active E1. The HSD17B1-expressing cells also formed estrogen-dependent tumors in immunodeficient nude mice. After treating the mice with an appropriate dose of the substrate (E1, 0.1 micromol/kg x d), a marked difference in tumor growth was observed between nontransfected and HSD17B1-transfected MCF-7 cells, mean tumor weights at the end of E1 treatment being 23.2 and 130.4 mg, respectively. Furthermore, estrogen-dependent growth of the HSD17B1-expressing xenografts in the presence of E1 was markedly inhibited by administering 5 micromol/kg x d of a specific HSD17B1 inhibitor. After a 4-wk treatment, the tumor size was reduced by 59.8% as compared with the nontreated tumors, whereas the uterine growth of the mice was not affected by the HSD17B1 inhibitor used. This was in line with the induction of apoptosis of the tumors. The results evidently show that estrogenic response for E1 is enhanced by the local action of HSD17B1 in vivo, and thus, the enzyme is a potential target for pharmacological inhibition of estrogen action.


Assuntos
Estradiol Desidrogenases/fisiologia , Estrogênios/farmacologia , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Hormônio-Dependentes/enzimologia , Animais , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Receptor alfa de Estrogênio/fisiologia , Estrogênios/metabolismo , Feminino , Humanos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/patologia , Transplante Heterólogo
9.
Mol Cell Endocrinol ; 248(1-2): 192-8, 2006 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-16413669

RESUMO

The estradiol-synthesizing enzyme 17beta-hydroxysteroid dehydrogenase type 1 (17betaHSD1) is mainly responsible for the conversion of estrone (E1) to the potent estrogen estradiol (E2). It is a key player to control tissue levels of E2 and is therefore an attractive target in estradiol-dependent diseases like breast cancer or endometriosis. We selected a unique non-steroidal pyrimidinone core to start a lead optimization program. We optimized this core by modulation of R1-R6. Its binding mode at the substrate-binding site of 17betaHSD1 is complex and difficult to predict. Nevertheless, some basic structure-activity relationships could be identified. In vitro, the most active pyrimidinone derivative showed effective inhibition of recombinant human 17betaHSD1 at nanomolar concentrations. In intact cells overexpressing the human enzyme, IC50 values in the lower micromolar range were determined. Furthermore, the pyrimidinone proved its use in vivo by significantly reducing 17betaHSD1-dependent tumor growth in a new nude mouse model.


Assuntos
Desenho Assistido por Computador , Desenho de Fármacos , Inibidores Enzimáticos/química , Estradiol Desidrogenases/antagonistas & inibidores , Pirimidinonas/química , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Conformação Proteica , Relação Estrutura-Atividade
10.
J Appl Physiol (1985) ; 95(3): 1145-52, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12909600

RESUMO

Pulmonary surfactant prevents alveolar collapse and contributes to airway patency by reducing surface tension. Although alveolar surfactant, consisting mainly of phospholipids (PL) together with neutral lipids and surfactant-specific proteins, originates from type II pneumocytes, the contribution of airway epithelia to the PL fraction of conductive airway surfactant is still debated. We, therefore, analyzed the composition, synthesis, and release of phosphatidylcholine (PC) molecular species as the main surfactant PL of the rat trachea compared with the lung. Analyses of individual PC molecular species with HPLC and electrospray ionization mass spectrometry revealed that the rat trachea contained and synthesized much more palmitoyloleoyl-PC, palmitoyllinoleoyl-PC, and palmitoylarachidonoyl-PC, together with increased amounts of alkylacyl-PC, and less surfactant-specific species such as dipalmitoyl-PC than the lung. Organ cultures with [methyl-3H]choline as precursor of PC revealed that, in the trachea, synthesized PC was retained in the tissue, rather than secreted. [Methyl-3H]choline-labeled dipalmitoyl-PC was a negligible component in the trachea, and, in contrast to the lungs, palmitoyloleoyl-PC was enriched in tracheal secretions. We conclude that the surfactant fraction in the airways does not originate from the airways but is produced in the alveolar space and transported upward.


Assuntos
Colina/análogos & derivados , Pulmão/metabolismo , Fosfatidilcolinas/metabolismo , Surfactantes Pulmonares/metabolismo , Traqueia/metabolismo , Animais , Colina/metabolismo , Cromatografia Líquida de Alta Pressão , Técnicas In Vitro , Cinética , Pulmão/ultraestrutura , Microscopia Eletrônica , Alvéolos Pulmonares/metabolismo , Ratos , Ratos Sprague-Dawley , Mucosa Respiratória/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Traqueia/ultraestrutura
11.
Anticancer Res ; 24(3a): 1433-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15274306

RESUMO

Estradiol (E2) is one of the main factors which control the growth and evolution of breast cancer. Consequently, to block the formation of E2 inside cancer cells has been an important target in recent years. Breast cancer cells possess all the enzymatic systems (e.g. sulfatase, aromatase, 17beta-hydroxysteroid dehydrogenase [17beta-HSD]) involved in the conversion of estrogen precursors into E2. Sulfotransferase, which converts estrogen to its sulfate, is also present in this tumoral tissue. Duphaston is a synthetic progestogen with properties similar to the natural progesterone. In the present study we examined the effect of Duphaston and its 20-dihydro-metabolite on the sulfatase and 17beta-HSD activities in MCF-7 and T-47D breast cancer cells. The cells were incubated with estrone sulfate (E1S) (5x10(-9)M) in the absence or presence of Duphaston or its 20-dihydro-metabolite (5x10(-5) to 5x10(-9)M) for 24h at 37 degrees C. In another series of experiments, estrone (E1) (5x10(-9)M) was incubated with T-47D cells in the absence or presence of the two progestogens (5x10(-5) to 5x10(-9)M) for 24h at 37 degrees C. E1S, E1 and E2 were characterized by thin layer chromatography and quantified using the corresponding standard. Duphaston and its 20-dihydro-metabolite, at concentrations of 5x10(-7) and 5x10(-5)M, inhibited the conversion of E1S to E2 by 14% and 63%, 65% and 74%, respectively, in MCF-7 cells; the values were 15% and 48% and 31% and 51%, respectively, in T-47D cells. In another series of experiments it was observed that, after 24-h incubation, E1 (5x10(-9)M) was converted in a great proportion to E2 in the T-47D cells and that this transformation was significantly inhibited by Duphaston and its 20-dihydro-metabolite. The IC50 value, corresponding to 50% of the inhibition in the conversion of 1 to E2, was 9x10(-6)M for 20-dihydro-metabolite in this cell line. It was concluded that the progestogen Duphaston and its 20-dihydro-metabolite are potent inhibitory agents on sulfatase and 17beta-HSD activities in breast cancer cells. Duphaston is a progestogen with properties similar to the endogenous progesterone. The data open interesting perspectives to study the biological responses of these progestogens in clinical trials of patients with breast cancer.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Didrogesterona/análogos & derivados , Didrogesterona/farmacologia , Estrona/análogos & derivados , Sulfatases/antagonistas & inibidores , 17-Hidroxiesteroide Desidrogenases/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Estradiol/biossíntese , Estrona/metabolismo , Humanos , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/enzimologia , Sulfatases/metabolismo
12.
Steroids ; 78(1): 69-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23127813

RESUMO

An expanding body of evidence indicates the possible role of estrane derivatives as useful anticancer agents. The aim of this study was to describe the cytotoxic effects of 63 newly synthetized estrone-16-oxime ethers on human cancer cell lines (cervix carcinoma HeLa, breast carcinoma MCF7 and skin epidermoid carcinoma A431), studied by means of the MTT assay. Four of the most promising compounds were selected for participation in additional experiments in order to characterize the mechanism of action, including cell cycle analysis, morphological study and the 5-bromo-2'-deoxyuridine incorporation assay. The cancer selectivity was tested on a noncancerous fibroblast cell line (MRC-5). Since apoptosis and cell cycle disturbance were observed, caspase-3 activities were further assayed for the two most effective agents. These estrone-16-oxime analogs activated caspase-3 and changed the mRNA level expression of endogenous factors regulating the G1-S phase transition (retinoblastoma protein, CDK4 and p16). The repression of retinoblastoma protein was reinforced at a protein level too. These experimental data lead to the conclusion that estrone-16-oxime ethers may be regarded as potential starting structures for the design of novel anticancer agents.


Assuntos
Antineoplásicos/farmacologia , Estrona/análogos & derivados , Estrona/farmacologia , Oximas/farmacologia , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Forma Celular , Sobrevivência Celular/efeitos dos fármacos , Replicação do DNA , Ensaios de Seleção de Medicamentos Antitumorais , Estrona/síntese química , Éteres/síntese química , Éteres/farmacologia , Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Concentração Inibidora 50 , Células MCF-7 , Oximas/síntese química
13.
J Steroid Biochem Mol Biol ; 125(3-5): 231-42, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21397694

RESUMO

In the search for novel biomarkers of endometriosis, we selected 152 genes from the GeneLogic database based on results of genome-wide expression analysis of ovarian endometriosis, plus 20 genes related to estrogen metabolism and action. We then performed low-density array analysis of these 172 genes on 11 ovarian endometriosis samples and 9 control endometrium samples. Principal component analysis of the gene expression levels showed clear separation between the endometriosis and control groups. We identified 78 genes as differentially expressed. Based on Ingenuity pathway analysis, these differentially expressed genes were arranged into groups according to biological function. These analyses revealed that 32 differentially expressed genes are estrogen related, 23 of which have not been reported previously in connection with endometriosis. Functional annotation showed that 25 and 22 genes are associated with the biological terms "secreted" and "extracellular region", respectively. Differential expression of 4 out of 5 genes related to estrogen metabolism and action (ESR1, ESR2, PGR and BGN) was also confirmed by immunohistochemistry. Our study thus reveals differential expression of several genes that have not previously been associated with endometriosis and that encode potential novel biomarkers and drug targets.


Assuntos
Biomarcadores/análise , Endometriose/genética , Endometriose/metabolismo , Adulto , Aromatase/genética , Aromatase/metabolismo , Biglicano/genética , Biglicano/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Adulto Jovem
14.
PLoS One ; 5(6): e10969, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20544026

RESUMO

Steroid-related cancers can be treated by inhibitors of steroid metabolism. In searching for new inhibitors of human 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD 1) for the treatment of breast cancer or endometriosis, novel substances based on 15-substituted estrone were validated. We checked the specificity for different 17beta-HSD types and species. Compounds were tested for specificity in vitro not only towards recombinant human 17beta-HSD types 1, 2, 4, 5 and 7 but also against 17beta-HSD 1 of several other species including marmoset, pig, mouse, and rat. The latter are used in the processes of pharmacophore screening. We present the quantification of inhibitor preferences between human and animal models. Profound differences in the susceptibility to inhibition of steroid conversion among all 17beta-HSDs analyzed were observed. Especially, the rodent 17beta-HSDs 1 were significantly less sensitive to inhibition compared to the human ortholog, while the most similar inhibition pattern to the human 17beta-HSD 1 was obtained with the marmoset enzyme. Molecular docking experiments predicted estrone as the most potent inhibitor. The best performing compound in enzymatic assays was also highly ranked by docking scoring for the human enzyme. However, species-specific prediction of inhibitor performance by molecular docking was not possible. We show that experiments with good candidate compounds would out-select them in the rodent model during preclinical optimization steps. Potentially active human-relevant drugs, therefore, would no longer be further developed. Activity and efficacy screens in heterologous species systems must be evaluated with caution.


Assuntos
Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Animais , Avaliação Pré-Clínica de Medicamentos , Estradiol Desidrogenases/metabolismo , Humanos , Especificidade da Espécie , Especificidade por Substrato
15.
J Steroid Biochem Mol Biol ; 117(4-5): 93-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19631742

RESUMO

The enzyme 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) has become an important drug target for breast cancer because it catalyzes the interconversion of estrone to the biologically more potent estradiol which also plays a crucial role in the etiology of breast cancer. Patients with an increased expression of the 17beta-HSD1 gene have a significantly worse outcome than patients without. Inhibitors for 17beta-HSD1 are therefore included in therapy development. Here we have studied binding of 17beta-HSD1 to substrates and a number of inhibitors using NMR spectroscopy. Ligand observed NMR spectra show a strong pH dependence for the phytoestrogens luteolin and apigenin but not for the natural ligands estradiol and estrone. Moreover, NMR competition experiments show that the phytoestrogens do not replace the estrogens despite their similar inhibition levels in the in vitro assay. These results strongly support an additional 17beta-HSD1 binding site for phytoestrogens which is neither the substrate nor the co-factor binding site. Docking experiments suggest the dimer interface as a possible location. An additional binding site for the phytoestrogens may open new opportunities for the design of inhibitors, not only for 17beta-HSD1, but also for other family members of the short chain dehydrogenases.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Fitoestrógenos/metabolismo , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/genética , Sequência de Bases , Sítios de Ligação , Primers do DNA , Concentração de Íons de Hidrogênio , Ligantes , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular
16.
J Med Chem ; 51(15): 4685-98, 2008 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-18630892

RESUMO

17beta-Estradiol (E2) is implicated in the genesis and the development of estrogen-dependent diseases. Its concentration is mainly regulated by 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1), which catalyzes the reduction of the weak estrogen estrone (E1) to the highly potent E2. This enzyme is thus an important target for the treatment of hormone-dependent diseases. Thirty-seven novel substituted 6-phenyl-2-naphthols were synthesized and evaluated for 17beta-HSD1 inhibition, selectivity toward 17beta-HSD2 and the estrogen receptors (ERs) alpha and beta, and pharmacokinetic properties. SAR studies revealed that the compounds most likely bind according to binding mode B to the active site, i.e., the 6-phenyl moiety mimicking the steroidal A-ring. While substitution at the phenyl ring decreased activity, introduction of substituents at the naphthol moiety led to highly active compounds, especially in position 1. The 1-phenyl compound 32 showed a very high inhibitory activity for 17beta-HSD1 (IC50 = 20 nM) and good selectivity (17beta-HSD2 and ERs) and pharmacokinetic properties after peroral application.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Naftóis/síntese química , Naftóis/farmacologia , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Inibidores Enzimáticos/química , Humanos , Masculino , Modelos Moleculares , Estrutura Molecular , Naftóis/química , Ratos , Ratos Wistar , Esteroides/química , Esteroides/farmacologia , Relação Estrutura-Atividade
17.
Fertil Steril ; 88(4 Suppl): 1029-38, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17316633

RESUMO

OBJECTIVE: To investigate the regulation of estrogen-converting enzymes in human ectopic endometrial tissue. DESIGN: Animal study. SETTING: Academic medical center. ANIMAL(S): Sixty female nude mice with implanted human endometrial tissue. PATIENT(S): Twenty-two premenopausal women undergoing endometrial biopsy or hysterectomy. INTERVENTION(S): Human endometrial tissue was implanted into the peritoneal cavity of nude mice, and the effect of therapeutic drugs on transcription of steroid receptors and estrogen-converting enzymes was analyzed. MAIN OUTCOME MEASURE(S): Transcript levels of steroid hormone receptors, 17beta-hydroxysteroid dehydrogenase type 1 and 2, aromatase, and steroid sulfatase as well as proliferation rate were analyzed in the human ectopic endometrial tissue. RESULT(S): Steroid receptors and estrogen-converting enzymes were expressed in the ectopic human endometrial fragments. Application of medroxyprogesterone acetate, dydrogesterone, danazol, and the aromatase inhibitor finrozole significantly inhibited aromatase transcription. In addition, danazol caused a significant decrease in transcription of steroid sulfatase, and finrozole, of 17beta-hydroxysteroid dehydrogenase type 1 in parallel to a decrease in proliferation rate in the ectopic human endometrial tissue. CONCLUSION(S): Pharmacological regulation of transcription of estrogen-converting enzymes in human endometrium cultured in nude mice may help to develop new therapeutic concepts based on local regulation of estrogen metabolism in endometriosis.


Assuntos
Coristoma/enzimologia , Endométrio/enzimologia , Estrogênios/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Animais , Coristoma/genética , Endometriose/enzimologia , Endometriose/genética , Estrogênios/genética , Feminino , Humanos , Camundongos , Camundongos Nus
18.
Planta ; 226(1): 225-34, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17245569

RESUMO

A plant lectin was isolated from barley (Hordeum vulgare) coleoptiles using acidic extraction and different chromatographic methods. Sequencing of more than 50% of the protein sequence by Edman degradation confirmed a full-length cDNA clone. The subsequently identified open reading frame encodes for a 15 kDa protein which could be found in the soluble fraction of barley coleoptiles. This protein exhibited specificity towards mannose sugar and is therefore, accordingly named as Horcolin (Hordeum vulgare coleoptile lectin). Database searches performed with the Horcolin protein sequence revealed a sequence and structure homology to the lectin family of jacalin-related lectins. Together with its affinity towards mannose, Horcolin is now identified as a new member of the mannose specific subgroup of jacalin-related lectins in monocot species. Horcolin shares a high amino acid homology to the highly light-inducible protein HL#2 and, in addition to two methyl jasmonic acid-inducible proteins of 32.6 and 32.7 kDa where the jasmonic acid-inducible proteins are examples of bitopic chimerolectins containing a dirigent and jacalin-related domain. Immunoblot analysis with a cross-reactive anti-HL#2 antibody in combination with Northern blot analysis of the Horcolin cDNA revealed tissue specific expression of Horcolin in the coleoptiles. The function of Horcolin is discussed in the context of its particular expression in coleoptiles and is then compared to other lectins, which apparently share a related response to biotic or abiotic stress factors.


Assuntos
Hordeum/química , Lectinas de Plantas/genética , Lectinas de Plantas/isolamento & purificação , Sequência de Aminoácidos , Clonagem Molecular , Bases de Dados de Proteínas , Regulação da Expressão Gênica de Plantas , Hordeum/genética , Dados de Sequência Molecular , Lectinas de Plantas/química
19.
Gynecol Endocrinol ; 23(2): 105-11, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17454161

RESUMO

Endometriosis is defined as the presence of endometrial glands and stroma within extrauterine sites, and it is well known that endometriosis is an estrogen-dependent disease. The defective formation and metabolism of steroid hormones is responsible for the promotion and development of endometriosis. In the present study we examined the mRNA levels of six enzymes that are involved in the metabolism of estrogen and progesterone--aromatase, 17beta-hydroxysteroid dehydrogenase (17beta-HSD) types 1, 2 and 7, sulfatase and sulfotransferase--and of the steroid receptors--estrogen receptors alpha and beta (ERalpha, ERbeta) and progesterone receptors A and B (PRAB)--implicated in human ovarian endometriosis. We analyzed 16 samples of ovarian endometriosis and 9 of normal endometrium. The real-time polymerase chain reaction analyses revealed that six of the nine genes investigated are differentially regulated. Aromatase, 17beta-HSD types 1 and 7, sulfatase and ERbeta were statistically significantly upregulated, while ERalpha was significantly downregulated, in the endometriosis group compared with the control group. There were no significant differences in 17beta-HSD type 2, sulfotransferase and PRAB gene expression. Our results indicate that, in addition to the previously reported upregulation of aromatase, upregulation of 17beta-HSD types 1 and 7 and sulfatase can also increase the local estradiol concentration. This could thus be responsible for the estrogen-dependent growth of endometriotic tissue. Surprisingly ERalpha was downregulated.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Endometriose/metabolismo , Receptor alfa de Estrogênio/metabolismo , Perfilação da Expressão Gênica , Doenças Ovarianas/metabolismo , Esteril-Sulfatase/metabolismo , 17-Hidroxiesteroide Desidrogenases/genética , Adulto , Aromatase/genética , Aromatase/metabolismo , Regulação para Baixo/genética , Endometriose/enzimologia , Endometriose/genética , Estradiol/sangue , Receptor alfa de Estrogênio/genética , Feminino , Humanos , Doenças Ovarianas/enzimologia , Doenças Ovarianas/genética , Esteril-Sulfatase/genética , Regulação para Cima/genética
20.
Eur J Biochem ; 269(4): 1278-86, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11856362

RESUMO

Previous biochemical studies have indicated that the membrane-bound thyrotropin-releasing hormone (TRH)-degrading enzyme (TRH-DE) from brain and liver and the serum TRH-DE are derived from the same gene. These studies also suggested that the serum enzyme is of liver origin. The present study was undertaken to verify these hypotheses. In different species, a close relationship between the activities of the serum enzyme and the particulate liver enzyme was noticed. The activity of the serum enzyme decreased when rats were treated with thioacetamide, a known hepatotoxin. With hepatocytes cultured in a sandwich configuration, release of the TRH-DE into the culture medium could also be demonstrated. The trypsin-solubilized particulate liver TRH-DE and the serum TRH-DE were purified to electrophoretic homogeneity. Both enzymes and the brain TRH-DE were recognized by a monoclonal antibody generated with the purified brain enzyme as antigen. Lectin blot analysis indicated that the serum enzyme and the liver enzyme are glycoproteins containing a sugar structure of the complex type, whereas the brain enzyme exhibits an oligomannose/hybrid glycostructure. A molecular mass of 97 000 Da could be estimated for all three enzymes after deglycosylation and SDS/PAGE followed by Western blotting. Fragment analysis of the serum TRH-DE revealed that the peptide sequences correspond to the cDNA deduced amino-acid sequences of the membrane-bound brain TRH-DE, whereby two peptides were identified that are encoded by exon 1. These data strongly support the hypothesis that the TRH-DEs are all derived from the same gene, whereby the serum enzyme is generated by proteolytic cleavage of the particulate liver enzyme.


Assuntos
Aminopeptidases/isolamento & purificação , Proteínas Sanguíneas/isolamento & purificação , Fígado/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Sequência de Aminoácidos , Aminopeptidases/metabolismo , Animais , Proteínas Sanguíneas/metabolismo , Bovinos , Células Cultivadas , Glicoproteínas/isolamento & purificação , Glicoproteínas/metabolismo , Hepatócitos/citologia , Fígado/efeitos dos fármacos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Ácido Pirrolidonocarboxílico/análogos & derivados , Ratos , Análise de Sequência de Proteína , Suínos , Tioacetamida/farmacologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA