Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Steroid Biochem Mol Biol ; 206: 105790, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33246154

RESUMO

17ß-Hydroxysteroid dehydrogenase type 2 (17ß-HSD2) catalyzes the conversion of highly active estrogens and androgens into their less active forms using NAD+ as cofactor. Substrate and cofactor specificities of 17ß-HSD2 have been reported and potent 17ß-HSD2 inhibitors have been discovered in a ligand-based approach. However, the molecular basis and the amino acids involved in the enzymatic functionality are poorly understood, as no crystal structure of the membrane-associated 17ß-HSD2 exists. The functional properties of only few amino acids are known. The lack of topological information impedes structure-based drug design studies and limits the design of biochemical experiments. The aim of this work was the determination of the 17ß-HSD2 topology. For this, the first homology model of 17ß-HSD2 in complex with NAD+ and 17ß-estradiol was built, using a multi-fragment "patchwork" approach. To confirm the quality of the model, fifteen selected amino acids were exchanged one by one using site directed mutagenesis. The mutants' functional behavior demonstrated that the generated model was of very good quality and allowed the identification of several key amino acids involved in either ligand or internal structure stabilization. The final model is an optimal basis for further experiments like, for example, lead optimization.


Assuntos
Estradiol Desidrogenases/genética , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade , Aminoácidos/genética , Catálise , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/química , Estradiol Desidrogenases/ultraestrutura , Humanos , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular
2.
J Hazard Mater ; 385: 121616, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-31780289

RESUMO

The efficient bioremediation of estrogen contamination in complex environments is of great concern. Here the strain Stenotrophomonas maltophilia SJTH1 was found with great and stable estrogen-degradation efficiency even under stress environments. The strain could utilize 17ß-estradiol (E2) as a carbon source and degrade 90% of 10 mg/L E2 in a week; estrone (E1) was the first degrading intermediate of E2. Notably, diverse pH conditions (3.0-11.0) and supplements of 4% salinity, 6.25 mg/L of heavy metal (Cd2+ or Cu2+), or 1 CMC of surfactant (Tween 80/ Triton X-100) had little effect on its cell growth and estrogen degradation. The addition of low concentrations of copper and Tween 80 even promoted its E2 degradation. Bioaugmentation of strain SJTH1 into solid clay soil achieved over 80% removal of E2 contamination (10 mg/kg) within two weeks. Further, the whole genome sequence of S. maltophilia SJTH1 was obtained, and a series of potential genes participating in stress-tolerance and estrogen-degradation were predicted. Four dehydrogenases similar to 17ß-hydroxysteroid dehydrogenases (17ß-HSDs) were found to be induced by E2, and the four heterogenous-expressed enzymes could oxidize E2 into E1 efficiently. This work could promote bioremediation appliance potential with microorganisms and biodegradation mechanism study of estrogens in complex real environments.


Assuntos
Proteínas de Bactérias/isolamento & purificação , Estradiol Desidrogenases/isolamento & purificação , Estradiol/metabolismo , Stenotrophomonas maltophilia/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biodegradação Ambiental , Estradiol Desidrogenases/química , Estradiol Desidrogenases/genética , Cinética , Octoxinol/farmacologia , Oxirredução , Polissorbatos/farmacologia , Alinhamento de Sequência , Stenotrophomonas maltophilia/efeitos dos fármacos , Stenotrophomonas maltophilia/enzimologia , Stenotrophomonas maltophilia/genética , Tensoativos/farmacologia
3.
Eur J Med Chem ; 178: 93-107, 2019 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-31176098

RESUMO

Estrogens are the major female sex steroid hormones, estradiol (E2) being the most potent form in humans. Disturbing the balance between E2 and its weakly active oxidized form estrone (E1) leads to diverse types of estrogen-dependent diseases such as endometriosis or osteoporosis. 17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) catalyzes the biosynthesis of E2 by reduction of E1 while the type 2 enzyme catalyzes the reverse reaction. Thus, 17ß-HSD1 and 17ß-HSD2 are attractive targets for treatment of estrogen-dependent diseases. Recently, we reported the first proof-of-principle study of a 17ß-HSD2 inhibitor in a bone fracture mouse model, using subcutaneous administration. In the present study, our aim was to improve the in vitro ADME profile of the most potent 17ß-HSD1 and 17ß-HSD2 inhibitors described so far. The optimized compounds show strong and selective inhibition of both the human enzymes and their murine orthologs. In addition, they display good metabolic stability in human liver microsomes (S9 fraction), low in vitro cytotoxicity as well as better aqueous solubility and physicochemical properties compared to the lead compounds. These achievements make the compounds eligible for testing in preclinical in vivo animal model studies on the effects of inhibition of 17ß-HSD1 and 17ß-HSD2.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Inibidores Enzimáticos/farmacocinética , Estradiol Desidrogenases/antagonistas & inibidores , Fenóis/farmacocinética , Tiofenos/farmacocinética , Animais , Sítios de Ligação , Desenho de Fármacos , Estabilidade de Medicamentos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Estradiol Desidrogenases/química , Estradiol Desidrogenases/metabolismo , Células HEK293 , Humanos , Camundongos , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Estrutura Molecular , Fenóis/síntese química , Fenóis/química , Fenóis/metabolismo , Ligação Proteica , Solubilidade , Relação Estrutura-Atividade , Tiofenos/síntese química , Tiofenos/química , Tiofenos/metabolismo
4.
Gene ; 588(1): 54-61, 2016 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-27102893

RESUMO

At the late 1940s, 17ß-HSD1 was discovered as the first member of the 17ß-HSD family with its gene cloned. The three-dimensional structure of human 17ß-HSD1 is the first example of any human steroid converting enzyme. The human enzyme's structure and biological function have thus been studied extensively in the last two decades. In humans, the enzyme is expressed in placenta, ovary, endometrium and breast. The high activity of estrogen activation provides the basis of 17ß-HSD1's implication in estrogen-dependent diseases, such as breast cancer, endometriosis and non-small cell lung carcinomas. Its dual function in estrogen activation and androgen inactivation has been revealed in molecular and breast cancer cell levels, significantly stimulating the proliferation of such cells. The enzyme's overexpression in breast cancer was demonstrated by clinical samples. Inhibition of human 17ß-HSD1 led to xenograft tumor shrinkage. Unfortunately, through decades of studies, there is still no drug using the enzyme's inhibitors available. This is due to the difficulty to get rid of the estrogenic activity of its inhibitors, which are mostly estrogen analogues. New non-steroid inhibitors for the enzyme provide new hope for non-estrogenic inhibitors of the enzyme.


Assuntos
Estradiol Desidrogenases/química , Estradiol Desidrogenases/metabolismo , Estrogênios/metabolismo , Androgênios/genética , Androgênios/metabolismo , Animais , Estradiol Desidrogenases/antagonistas & inibidores , Estradiol Desidrogenases/genética , Estrogênios/genética , Humanos , Mutação , Neoplasias/metabolismo , Especificidade de Órgãos
5.
Future Med Chem ; 7(11): 1431-56, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26230882

RESUMO

Low estradiol level in postmenopausal women is implicated in osteoporosis, which occurs because of the high bone resorption rate. Estrogen formation is controlled by 17-ß hydroxysteroid dehydrogenase 17-ß HSD enzymes, where 17-ß HSD type 1 contributes in the formation of estradiol, while type 2 catalyzes its catabolism. Inhibiting 17-ß HSD2 can help in increasing estradiol concentration. Several promising 17-ß HSD2 inhibitors that can act at low nanomolar range have been identified. However, there are some specific challenges associated with the application of these compounds. Our review provides an up-to-date summary of the current status and recent progress in the production of 17-ß HSD2 inhibitors as well as the future challenges in their clinical application.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Estradiol Desidrogenases/antagonistas & inibidores , Estradiol Desidrogenases/metabolismo , Osteoporose/tratamento farmacológico , Osteoporose/enzimologia , Animais , Descoberta de Drogas/métodos , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/química , Humanos , Modelos Moleculares , Osteoporose/patologia
6.
J Mol Cell Biol ; 7(6): 568-79, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25966904

RESUMO

17ß-hydroxysteroid dehydrogenase (17ß-HSD) type 1 is known as a critical target to block the final step of estrogen production in estrogen-dependent breast cancer. Recent confirmation of the role of dyhydroxytestosterone (DHT) in counteracting estrogen-induced cell growth prompted us to study the reductive 17ß-HSD type 7 (17ß-HSD7), which activates estrone while markedly inactivating DHT. The role of DHT in breast cancer cell proliferation is demonstrated by its independent suppression of cell growth in the presence of a physiological concentration of estradiol (E2). Moreover, an integral analysis of a large number of clinical samples in Oncomine datasets demonstrated the overexpression of 17ß-HSD7 in breast carcinoma. Inhibition of 17ß-HSD7 in breast cancer cells resulted in a lower level of E2 and a higher level of DHT, successively induced regulation of cyclinD1, p21, Bcl-2, and Bik, consequently arrested cell cycle in the G(0)/G(1) phase, and triggered apoptosis and auto-downregulation feedback of the enzyme. Such inhibition led to significant shrinkage of xenograft tumors with decreased cancer cell density and reduced 17ß-HSD7 expression. Decreased plasma E2 and elevated plasma DHT levels were also found. Thus, the dual functional 17ß-HSD7 is proposed as a novel target for estrogen-dependent breast cancer by regulating the balance of E2 and DHT. This demonstrates a conceptual advance on the general belief that the major role of this enzyme is in cholesterol metabolism.


Assuntos
17-Hidroxiesteroide Desidrogenases/metabolismo , Androgênios/metabolismo , Neoplasias da Mama/enzimologia , Di-Hidrotestosterona/metabolismo , Estradiol/metabolismo , Estrogênios/metabolismo , 17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 17-Hidroxiesteroide Desidrogenases/genética , Androgênios/sangue , Androgênios/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias da Mama/metabolismo , Pontos de Checagem do Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Colesterol/metabolismo , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Di-Hidrotestosterona/sangue , Di-Hidrotestosterona/farmacologia , Estradiol/sangue , Estradiol Desidrogenases/química , Estrogênios/sangue , Estrona/metabolismo , Feminino , Fase G1 , Humanos , Células MCF-7 , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais , Enzimas Multifuncionais/antagonistas & inibidores , Enzimas Multifuncionais/química , Enzimas Multifuncionais/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fase de Repouso do Ciclo Celular
7.
J Med Chem ; 54(21): 7547-57, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21972996

RESUMO

Estrogen deficiency in postmenopausal women or elderly men is often associated with the skeletal disease osteoporosis. The supplementation of estradiol (E2) in osteoporotic patients is known to prevent bone fracture but cannot be administered because of adverse effect. As 17ß-hydroxysteroid dehydrogenase type 2 (17ß-HSD2) oxidizes E2 to its inactive form estrone (E1) and has been found in osteoblastic cells, it is an attractive target for the treatment of osteoporosis. Twenty-one novel, naphthalene-derived compounds have been synthesized and evaluated for their 17ß-HSD2 inhibition and their selectivity toward 17ß-HSD1 and the estrogen receptors (ERs) α and ß. Compound 19 turned out to be the most potent and selective inhibitor of 17ß-HSD2 in cell-free assays and had a very good cellular activity in MDA-MB-231 cells, expressing naturally 17ß-HSD2. It also showed marked inhibition of the E1-formation by the rat and mouse orthologous enzymes and strong inhibition of monkey 17ß-HSD2. It is thus an appropriate candidate to be further evaluated in a disease-oriented model.


Assuntos
Estradiol Desidrogenases/antagonistas & inibidores , Naftóis/síntese química , Fenóis/síntese química , 17-Hidroxiesteroide Desidrogenases , Animais , Ligação Competitiva , Callithrix , Linhagem Celular Tumoral , Estradiol Desidrogenases/química , Feminino , Humanos , Técnicas In Vitro , Camundongos , Microssomos Hepáticos/enzimologia , Naftóis/química , Naftóis/farmacologia , Fenóis/química , Fenóis/farmacologia , Placenta/enzimologia , Gravidez , Ensaio Radioligante , Ratos , Receptores de Estrogênio/metabolismo , Relação Estrutura-Atividade
8.
PLoS One ; 6(8): e22990, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21857977

RESUMO

17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) catalyzes the reduction of estrone to estradiol, which is the most potent estrogen in humans. Inhibition of 17ß-HSD1 and thereby reducing the intracellular estradiol concentration is thus a promising approach for the treatment of estrogen dependent diseases. In the past, several steroidal and non-steroidal inhibitors of 17ß-HSD1 have been described but so far there is no cocrystal structure of the latter in complex with 17ß-HSD1. However, a distinct knowledge of active site topologies and protein-ligand interactions is a prerequisite for structure-based drug design and optimization. An elegant strategy to enhance this knowledge is to compare inhibition values obtained for one compound toward ortholog proteins from various species, which are highly conserved in sequence and differ only in few residues. In this study the inhibitory potencies of selected members of different non-steroidal inhibitor classes toward marmoset 17ß-HSD1 were determined and the data were compared with the values obtained for the human enzyme. A species specific inhibition profile was observed in the class of the (hydroxyphenyl)naphthols. Using a combination of computational methods, including homology modelling, molecular docking, MD simulation, and binding energy calculation, a reasonable model of the three-dimensional structure of marmoset 17ß-HSD1 was developed and inhibition data were rationalized on the structural basis. In marmoset 17ß-HSD1, residues 190 to 196 form a small α-helix, which induces conformational changes compared to the human enzyme. The docking poses suggest these conformational changes as determinants for species specificity and energy decomposition analysis highlighted the outstanding role of Asn152 as interaction partner for inhibitor binding. In summary, this strategy of comparing the biological activities of inhibitors toward highly conserved ortholog proteins might be an alternative to laborious x-ray or site-directed mutagenesis experiments in certain cases. Additionally, it facilitates inhibitor design and optimization by offering new information on protein-ligand interactions.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Relação Estrutura-Atividade , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Sítios de Ligação/genética , Callithrix , Simulação por Computador , Cristalografia por Raios X , Estradiol Desidrogenases/química , Estradiol Desidrogenases/metabolismo , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Especificidade por Substrato
9.
J Med Chem ; 53(22): 8176-86, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20977238

RESUMO

Estradiol (E2), the most important estrogen in humans, is involved in the initiation and progression of estrogen-dependent diseases such as breast cancer and endometriosis. Its local production in the target cell is regulated by 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1), which catalyzes E2-formation by reduction of the weak estrogen estrone (E1). Because the enzyme is expressed in the diseased tissues, inhibition of 17ß-HSD1 is considered as a promising therapy for the treatment of estrogen-dependent diseases. For the development of novel inhibitors, a structure- and ligand-based design strategy was applied, resulting in bicyclic substituted hydroxyphenylmethanones. In vitro testing revealed high inhibitory potencies toward human placental 17ß-HSD1. Compounds were further evaluated with regard to selectivity (17ß-HSD2, estrogen receptors ERα and ERß), intracellular activity (T47D cells), and metabolic stability. The most promising compounds, 14 and 15, showed IC(50) values in the low nanomolar range in the cell-free and cellular assays (8-27 nM), more than 30-fold selectivity toward 17ß-HSD2 and no affinity toward the ERs. The data obtained make these inhibitors interesting candidates for further preclinical evaluation.


Assuntos
Estradiol Desidrogenases/antagonistas & inibidores , Fenóis/síntese química , Tiazóis/síntese química , Tiofenos/síntese química , Linhagem Celular Tumoral , Desenho de Fármacos , Estabilidade de Medicamentos , Estradiol Desidrogenases/química , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Técnicas In Vitro , Microssomos Hepáticos/metabolismo , Fenóis/química , Fenóis/farmacologia , Placenta/metabolismo , Gravidez , Relação Estrutura-Atividade , Tiazóis/química , Tiazóis/farmacologia , Tiofenos/química , Tiofenos/farmacologia
10.
ChemMedChem ; 3(9): 1371-6, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18576452

RESUMO

Knowledge about the orientation of ligands or inhibitors bound to a protein is vital for the development of new drugs. It was recently shown that solvent accessibility epitopes for protein ligands can be mapped by transferring magnetization from water molecules to the ligand to derive the ligand orientation. This is based on the fact that NMR signals of ligands arising from magnetization transferred from solvent molecules via the protein have a different sign from those arising from direct magnetization transfer from bulk water. Herein we critically evaluate the applicability of solvent accessibility mapping to derive binding orientations for ligands of two dehydrogenases (AKR1C3 and HSD17beta1) with very different binding pockets, including complexes in which the ligand is buried more deeply inside the protein. We also evaluate the possibility of using co-solvents, such as DMSO, for magnetization transfer.


Assuntos
3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Dimetil Sulfóxido/química , Epitopos/química , Estradiol Desidrogenases/antagonistas & inibidores , Flavonoides/farmacologia , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Indometacina/farmacologia , 3-Hidroxiesteroide Desidrogenases/química , Membro C3 da Família 1 de alfa-Ceto Redutase , Sítios de Ligação/efeitos dos fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/química , Flavonoides/química , Humanos , Hidroxiprostaglandina Desidrogenases/química , Indometacina/química , Ligantes , Espectroscopia de Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética/normas , Magnetismo , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Padrões de Referência , Solventes/química , Propriedades de Superfície , Água/química
11.
Arch Biochem Biophys ; 467(1): 76-86, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17888864

RESUMO

In this study, we characterized rat and mouse aldo-keto reductases (AKR1C16 and AKR1C13, respectively) with 92% sequence identity. The recombinant enzymes oxidized non-steroidal alcohols using NAD+ as the preferred coenzyme, and showed low 3alpha/17beta/20alpha-hydroxysteroid dehydrogenase (HSD) activities. The substrate specificity differs from that of rat NAD+-dependent 3alpha-HSD (AKR1C17) that shares 95% sequence identity with AKR1C16. To elucidate the residues determining the substrate specificity of the enzymes, we performed site-directed mutagenesis of Tyr24, Asp128 and Phe129 of AKR1C16 with the corresponding residues (Ser, Tyr and Leu, respectively) of AKR1C17. The double mutation (Asp128/Tyr-Phe129/Leu) had few effects on the substrate specificity, while the Tyr24/Ser mutant showed only 3alpha-HSD activity, and the triple mutation of the three residues produced an enzyme that had almost the same properties as AKR1C17. The importance of the residue 24 for substrate recognition was verified by the mutagenesis of Ser24/Tyr of AKR1C17 which resulted in a decrease in 3alpha-HSD activity and appearance of 17beta- and 20alpha-HSD activities. AKR1C16 is also 92% identical with rat NAD+-dependent 17beta-HSD (AKR1C24), which possesses Tyr24. The replacement of Asp128, Phe129 and Ser137 of AKR1C16 with the corresponding residues (Glu, Ser and Phe, respectively) of AKR1C24 increased the catalytic efficiency for 17beta- and 20alpha-hydroxysteroids.


Assuntos
Estradiol Desidrogenases/química , Hidroxiesteroide Desidrogenases/fisiologia , Álcoois/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Estradiol Desidrogenases/genética , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ratos , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Especificidade por Substrato , Distribuição Tecidual , Tirosina/química
12.
Breast Cancer Res Treat ; 106(1): 57-64, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17260097

RESUMO

In situ synthesis of oestrogens is of great importance in the development and progression of breast cancer. 17beta-hydroxysteroid dehydrogenase (17HSD) type 2 catalyses oxidation from oestradiol to oestrone, and thereby protects the breast epithelial cells from oestradiol. Low expression of 17HSD type 2 has been associated with decreased survival in breast cancer, but no studies have investigated the mechanism behind the low expression. The 17HSD type 2 gene (HSD17B2) was screened for mutations with Single Stranded Conformation Polymorphism (SSCP)-DNA sequencing in 59 sporadic breast cancer cases, 19 hereditary breast cancer cases and seven breast cancer cell lines. DNA samples from 226 healthy individuals were used to identify if changes were previously unknown polymorphisms. No mutation was detected and therefore mutations in HSD17B2 do not explain why some breast tumours exhibit low 17HSD type 2 expression. However, a previously unknown polymorphism was found in exon four (Met226Val). Using molecular modelling, we found that the substituted residue is located at the outer part of the steroid binding site, probably causing minor alterations in the substrate binding. We further studied if the polymorphism contributes to breast cancer susceptibility in a larger material, but did not find an increased risk in the group of 317 sporadic breast cancer patients, 188 breast cancer patients with two close relatives with breast cancer or 122 hereditary breast cancer patients, compared to the healthy control group. We suggest that the detected polymorphism does not contribute to a higher risk of developing breast cancer.


Assuntos
Neoplasias da Mama/genética , Estradiol Desidrogenases/genética , Éxons , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Polimorfismo Conformacional de Fita Simples , Idoso , Estudos de Casos e Controles , Domínio Catalítico , Linhagem Celular Tumoral , Análise Mutacional de DNA , Estradiol Desidrogenases/química , Feminino , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Conformação Proteica , Medição de Risco , Fatores de Risco
13.
Genomics ; 88(6): 820-830, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16860536

RESUMO

Human short-chain dehydrogenases/reductases with dual retinol/sterol substrate specificity (RODH-like enzymes) are thought to contribute to the oxidation of retinol for retinoic acid biosynthesis and to the metabolism of androgenic and neuroactive 3alpha-hydroxysteroids. Here, we investigated the phylogeny and orthology of these proteins to understand better their origins and physiological roles. Phylogenetic and genomic analysis showed that two proteins (11-cis-RDH and RDHL) are highly conserved, and their orthologs can be identified in the lower taxa, such as amphibians and fish. Two other proteins (RODH-4 and 3alpha-HSD) are significantly less conserved. Orthologs for 3alpha-HSD are present in all mammals analyzed, whereas orthologs for RODH-4 can be identified in some mammalian species but not in others due to species-specific gene duplications. Understanding the evolution and divergence of RODH-like enzymes in various vertebrate species should facilitate further investigation of their in vivo functions using animal models.


Assuntos
Genômica , Hidroxiesteroides/metabolismo , Oxirredutases/genética , Filogenia , Vitamina A/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/química , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , 3-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Álcool Desidrogenase/química , Álcool Desidrogenase/genética , Álcool Desidrogenase/metabolismo , Oxirredutases do Álcool/química , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Sequência de Aminoácidos , Animais , Estradiol Desidrogenases/química , Estradiol Desidrogenases/genética , Estradiol Desidrogenases/metabolismo , Humanos , Dados de Sequência Molecular , Oxirredutases/química , Oxirredutases/metabolismo , Especificidade por Substrato
14.
Mol Cell Endocrinol ; 248(1-2): 38-46, 2006 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-16480815

RESUMO

17Beta-hydroxysteroid dehydrogenases/ketosteroid reductases (17beta-HSDs/KSRs) catalyze the last step of sex steroid synthesis or the first step of their degradation, and are thus critical for many physiological processes. The multispecificity demonstrated by 17beta-HSDs is important for steroid metabolism in gonadal and peripheral tissues, and is a consequence of the architecture of their binding and catalytic sites. Structurally, most of the family members are short chain dehydrogenase-reductases (SDRs) except the type 5 enzyme, which is an aldo-keto reductase (AKR). 17Beta-HSD type 1, a representative of the SDR family, has been studied extensively since the 1950s. However, its structure was not determined until the 1990s. It has always been considered as estrogen specific, in accord with the narrow binding tunnel that has been structurally determined and has been found to be complementary to estrogens. A recent study revealed that, in spite of the enzyme's narrow binding tunnel, the pseudo-symmetry of C19 steroids leads to its alternative binding, resulting in the multispecificity of the enzyme. Expressed in ovary, breast and placenta, the enzyme catalyzes the formation of another estrogen A-diol from DHEA in addition to the biosynthesis of estradiol; it also inactivates the most active androgen DHT by both 17beta-hydroxysteroid oxidation and 3-ketosteroid reduction. Type 5 17beta-HSD (AKR1C3) differs significantly from the type 1 enzyme by possessing a spacious and flexible steroid-binding site. This is estimated to be about 960 or 470 A3 in ternary complex with testosterone or 4-dione, respectively, whereas the binding site volume of 17beta-HSD1 is only about 340 A3. This characteristic of the 17beta-HSD5 binding site permits the docking of various steroids in different orientations, which encompasses a wider range of activities from 20alpha-, 17beta- and 3alpha-HSD/KSR to prostaglandin 11-ketoreductase. The in vitro activities of the enzyme are significantly lower than the type 1 enzyme. In the ternary complex with testosterone, the steroid C3-C17 position is quasi-reversed as compared to the complex with 4-dione. The multi-specificity contributes significantly to steroid metabolism in peripheral tissues, due to the high levels of 17beta-HSD5 mRNA in both breast and prostate tissues.


Assuntos
17-Hidroxiesteroide Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/química , Estradiol Desidrogenases/química , Hidroxiprostaglandina Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Estradiol Desidrogenases/metabolismo , Humanos , Hidroxiprostaglandina Desidrogenases/metabolismo , Conformação Proteica , Esteroides/metabolismo , Especificidade por Substrato , Distribuição Tecidual
15.
Mol Cell Endocrinol ; 248(1-2): 208-13, 2006 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-16412556

RESUMO

Estrogens, especially estradiol, have been shown to stimulate the proliferation of hormone-dependent types of breast cancer cells. 17Beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) enzyme catalyses the synthesis of the active female estrogen, estradiol and is thus an attractive target for structure-based ligand design for the prevention and control of breast tumour growth. In this study, the active site of 17beta-HSD1 has been reviewed, and three crystal structure complexes (estradiol/NADP+, equilin/NADP+, dehydroepiandrosterone) of 17beta-HSD1 have been selected to be analysed for de novo ligand design. The boundary surface, hydrophobic interactions and hydrogen bonding sites in the ligand binding domain for each ligand complex were analysed to create a comprehensive image of the active site.


Assuntos
Desidroepiandrosterona/química , Equilina/química , Estradiol Desidrogenases/química , Estradiol/química , Software , Sítios de Ligação , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Ligantes , Estrutura Terciária de Proteína
16.
Mol Cell Endocrinol ; 248(1-2): 236-8, 2006 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-16337735

RESUMO

Three series of steroid derivatives, enones 1, enols 2 and saturated alcohols 3, were easily synthesized from estrone according to a sequence of three reactions: an aldol condensation with an aromatic aldehyde (R(a-g)CHO) to afford 1, the carbonyl reduction of 1 to obtain the enol 2, and the double bond reduction of 2 to give 3 with the R(a-g) group 16beta-oriented. All compounds were tested as inhibitors of type 1 17beta-HSD. The inhibitory potency increases in the following order 1<2<3, suggesting that the presence of a flexible 16beta-methylene group allows a better positioning of the aryl moiety. With an IC50 of 0.8 microM, the 16beta-benzyl-E2 (3a) is the best inhibitor in this series.


Assuntos
Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Estradiol/análogos & derivados , Estrona/análogos & derivados , Álcoois/síntese química , Álcoois/química , Álcoois/farmacologia , Cicloexanonas/síntese química , Cicloexanonas/química , Cicloexanonas/farmacologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Estradiol/química , Estradiol Desidrogenases/química , Estrona/química , Humanos , Conformação Proteica
17.
FASEB J ; 17(10): 1334-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12759331

RESUMO

Steroid hormones play an essential role in a wide range of physiological and pathological processes, such as growth, metabolism, aging, and hormone-sensitive cancers. Estrogens are no exception and influence growth, differentiation, and functioning of many target tissues, such as the mammary gland, uterus, hypothalamus, pituitary, bone, and liver. Although very similar in structure, each steroid class (i.e., estrogens, androgens, progestins, mineral corticoids, or glucocorticoids) is responsible for distinct physiological processes. To permit specific biological responses for a given steroid class, specific proteins are responsible for steroid bioactivation, action, and inactivation, yet they have low or no affinity to other classes. Estrogens make no exception and possess their own set of related proteins. To understand the molecular basis underlying estrogen recognition from other steroids, structural features of estrogen-specific proteins were analyzed along with their ability to discriminate between steroid hormones belonging to different classes. Hence, the study of all estrogen-specific proteins for which an atomic structure has been determined demonstrated that a common steroid-binding pocket architecture is shared by these proteins. This architecture is composed of the following elements: i) a glutamate residue acting as a proton acceptor coupled with a proton donor that interact with the steroid O3; ii) a proton donor (His or Ser) that interacts with O17; iii) a highly conserved sandwich-like structure providing steric hindrance and preventing C19 steroid from binding; and iv) several amino acid residues interacting with the C18. As these different estrogen-specific proteins are not related in overall sequence, the inference is that the steroid binding site in these proteins has originated by convergent evolution.


Assuntos
Estrogênios/metabolismo , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Androgênios/metabolismo , Anticorpos/química , Anticorpos/metabolismo , Sítios de Ligação , Estradiol Desidrogenases/química , Estradiol Desidrogenases/metabolismo , Receptor alfa de Estrogênio , Estrogênios/imunologia , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica , Receptores de Estrogênio/química , Receptores de Estrogênio/metabolismo , Sulfotransferases/química , Sulfotransferases/metabolismo
18.
Proc Natl Acad Sci U S A ; 96(3): 840-5, 1999 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-9927655

RESUMO

Excess 17beta-estradiol (E2), the most potent of human estrogens, is known to act as a stimulus for the growth of breast tumors. Human estrogenic 17beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1), which catalyzes the reduction of inactive estrone (E1) to the active 17beta-estradiol in breast tissues, is a key enzyme responsible for elevated levels of E2 in breast tumor tissues. We present here the structure of the ternary complex of 17beta-HSD1 with the cofactor NADP+ and 3-hydroxyestra-1,3,5,7-tetraen-17-one (equilin), an equine estrogen used in estrogen replacement therapy. The ternary complex has been crystallized with a homodimer, the active form of the enzyme, in the asymmetric unit. Structural and kinetic data presented here show that the 17beta-HSD1-catalyzed reduction of E1 to E2 in vitro is specifically inhibited by equilin. The crystal structure determined at 3.0-A resolution reveals that the equilin molecule is bound at the active site in a mode similar to the binding of substrate. The orientation of the 17-keto group with respect to the nicotinamide ring of NADP+ and catalytic residues Tyr-155 and Ser-142 is different from that of E2 in the 17beta-HSD1-E2 complex. The ligand and substrate-entry loop densities are well defined in one subunit. The substrate-entry loop adopts a closed conformation in this subunit. The result demonstrates that binding of equilin at the active site of 17beta-HSD1 is the basis for inhibition of E1-to-E2 reduction by this equine estrogen in vitro. One possible outcome of estrogen replacement therapy in vivo could be reduction of E2 levels in breast tissues and hence the reduced risk of estrogen-dependent breast cancer.


Assuntos
Equilina/metabolismo , Estradiol Desidrogenases/química , Estradiol Desidrogenases/metabolismo , NADP/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Dimerização , Equilina/química , Humanos , Análise dos Mínimos Quadrados , Substâncias Macromoleculares , Modelos Moleculares , Dados de Sequência Molecular , NADP/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Spodoptera , Transfecção
19.
J Steroid Biochem Mol Biol ; 70(4-6): 229-35, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10622412

RESUMO

Human estrogenic 17beta-hydroxysteroid dehydrogenase (17beta-HSD1) catalyzes the synthesis of 17beta-estradiol (E2) from estrone, in the ovary and peripheral tissues. While the structures of 17beta-HSD1 alone and in complex with E2 have been determined (D. Ghosh, V. Pletnev, D.-W. Zhu, Z. Wawrzak, W.-L. Duax, W. Pangborn, F. Labrie, S.-X. Lin, Structure of human 17beta-hydroxysteroid dehydrogenase at 2.20 A resolution, Structure 3 (1995) 503-513), no structures of inhibitor/enzyme complex, either modeled or from crystallography, have been reported before the submission of the present paper. The best available inhibitors are among the 'dual-site inhibitors', blocking estrogenic 17beta-HSD and the estrogen receptor. These compounds belong to a family of estradiol analogues having an halogen atom at the 16alpha position and an extended alkyl-amide chain at the 7alpha position (C. Labrie, G. Martel, J.M. Dufour, G. Levesque, Y. Merand, F. Labrie, Novel compounds inhibit estrogen formation and action, Cancer Res. 52 (1992) 610-615). We now report the crystallization of this enzyme/inhibitor complex. The complex of the best available dual-site inhibitor, EM-139, with 17beta-HSD1 has been crystallized using both cocrystallization and soaking methods. Crystals are isomorphous to the native crystals grown in the presence of 0.06% beta-octyl-glucoside and polyethyleneglycol 4000, with a monoclinic space group C2. Data at 1.8 A have been collected from a synchrotron source. Even though the size of the inhibitor is greater than that of the substrate, our preliminary X-ray-diffraction study shows that EM-139 fits into the active site in a position similar to that of estrogen. The availability of such structural data will help design more potent inhibitors of estrogenic 17beta-HSD.


Assuntos
Estradiol Desidrogenases/química , Apoenzimas/química , Cristalização , Cristalografia por Raios X , Estradiol/análogos & derivados , Estradiol/química , Estradiol/metabolismo , Estradiol/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Estradiol Desidrogenases/metabolismo , Antagonistas de Estrogênios/química , Antagonistas de Estrogênios/farmacologia , Estrona/metabolismo , Feminino , Humanos , Ovário/enzimologia
20.
Biochem J ; 330 ( Pt 3): 1361-8, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9494107

RESUMO

After our previous report on the cloning of two cDNA species in guinea pig, both encoding the same hepatic 79 kDa multifunctional protein 1 (MFP-1) [Caira, Cherkaoui-Malki, Hoefler and Latruffe (1996) FEBS Lett. 378, 57-60], here we report the cloning of a cDNA encoding a second multifunctional peroxisomal protein (MFP-2) in guinea-pig liver. This 2356 nt cDNA encodes a protein of 735 residues (79.7 kDa) whose sequence shows 83% identity with rat MFP-2 [Dieuaide-Noubhani, Novikov, Baumgart, Vanhooren, Fransen, Goethals, Vandekerckhove, Van Veldhoven and Mannaerts (1996) Eur. J. Biochem. 240, 660-666]. In parallel, we studied the effect of ciprofibrate, a hypolipaemic agent also known as peroxisome proliferator in rodent, on the expression of MFP-1 and MFP-2 (2.6 kb) in rats and guinea pigs. By Northern blotting analysis we demonstrated that three MFP-1-related mRNA species are expressed in the guinea-pig liver. The expression of two of them (3.5 and 2.6 kb) is slightly increased by ciprofibrate, whereas the 3.0 kb MFP-1 mRNA is, unlike the rat one, strongly down-regulated in guinea pigs treated with ciprofibrate. In a similar way, the hepatic expression of the guinea-pig 2.6 kb MFP-2 mRNA is also down-regulated in guinea pigs treated with ciprofibrate. These results demonstrate (1) that in contrast with the unique 3.0 kb MFP-1 rat mRNA, at least three hepatic MFP-1-related mRNA species are co-expressed in guinea pig; and (2) that, opposed to the accepted idea of non-responsiveness of the guinea pig to ciprofibrate, this drug affects MFP-1 and MFP-2 gene expression in this species. Also, the mRNA species for acyl-CoA oxidase and thiolase, two other enzymes of the peroxisomal beta-oxidation pathway that are induced severalfold in responsive species are down-regulated in guinea pig. This paper is the first, to our knowledge, reporting the down-regulation of the expression of genes encoding enzymes involved in the peroxisomal beta-oxidation of fatty acids (MFP-1) and bile acid synthesis (MFP-2) in mammals.


Assuntos
Ácido Clofíbrico/análogos & derivados , Estradiol Desidrogenases/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/metabolismo , Microcorpos/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Ácido Clofíbrico/farmacologia , Clonagem Molecular , DNA Complementar , Enoil-CoA Hidratase/metabolismo , Estradiol Desidrogenases/química , Ácidos Fíbricos , Cobaias , Hipolipemiantes/farmacologia , Fígado/efeitos dos fármacos , Masculino , Microcorpos/efeitos dos fármacos , Microcorpos/enzimologia , Dados de Sequência Molecular , Oxirredutases/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA