Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Steroid Biochem Mol Biol ; 159: 31-40, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26907965

RESUMEN

Mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) are descended from a common ancestral corticoid receptor. The basis for specificities of human MR for aldosterone and human GR for glucocorticoids, such as cortisol, bearing 17α-hydroxyl-groups, is incompletely understood. Differences in MR at S843 and L848 and GR at the corresponding P637 and Q642 have been proposed as important in their different responses to glucocorticoids with 17α-hydroxyl-groups. We investigated the impact of these residues on binding affinity (Ki) and transcriptional activation (EC50) of mutants MR-S843P, MR-L848Q and MR-S843P/L848Q and mutants GR-P637S, GR-Q642L and GR-P637S/Q642L in the presence of different corticosteroids. Aldosterone, cortisol and corticosterone had similar affinities for wild-type MR and all mutants, while dexamethasone had increased affinity for the three mutants. However, transactivation of MR-S843P and MR-S843P/L848Q by all four steroids was significantly lower than for wild-type MR. In contrast, transactivation of MR-L848Q tended to be 3-fold higher for cortisol and corticosterone and increased 7-fold for dexamethasone, indicating that MR-L848Q has an increased response to glucocorticoids, while retaining a strong response to aldosterone. Compared to wild-type GR, GR-P637S and GR-Q642L had increased affinities and significantly increased transcriptional activity with aldosterone and corticosterone, and GR-P637S had similar transcriptional activity with cortisol and dexamethasone, while GR-Q642L and GR-P637S/Q642L had a significant decrease in transcriptional activity with cortisol and dexamethasone. 3D-models of these MR and GR mutants revealed that dexamethasone and aldosterone, respectively, fit nicely into the steroid-binding pocket, consistent with the affinity of dexamethasone for MR mutants and aldosterone for GR mutants.


Asunto(s)
Aldosterona/fisiología , Hidrocortisona/fisiología , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Activación Transcripcional , Sustitución de Aminoácidos , Sitios de Unión , Secuencia Conservada , Dexametasona , Glutamina/química , Células HEK293 , Humanos , Enlace de Hidrógeno , Leucina/química , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Prolina/química , Unión Proteica , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/química , Receptores de Mineralocorticoides/genética , Serina/química , Homología Estructural de Proteína
2.
Mol Inform ; 33(1): 15-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27485195

RESUMEN

11ß-Hydroxysteroid dehydrogenases (11ß-HSD) control the intracellular concentrations of glucocorticoids: 11ß-HSD1 converts the inactive cortisone to the active cortisol, and 11ß-HSD2 is responsible for the opposite reaction. Inhibition of 11ß-HSD1 is beneficial in the treatment of metabolic syndrome, whereas 11ß-HSD2 inhibition leads to hypertension. Therefore, 11ß-HSD1 inhibitors should be selective over 11ß-HSD2. To support drug discovery and toxicological studies, we have previously reported pharmacophore models for 11ß-HSD1 and 2 inhibition. These models represent the common chemical features of 11ß-HSD inhibitors, which were used as virtual screening filter. Since new inhibitors are constantly discovered, the quality of the pharmacophore models has to be evaluated in order to maintain a good predictive power. In this study, we report a systematic evaluation and refinement of our pharmacophore model collection. We employed our models for virtual screening, especially focusing on the 11ß-HSD2 inhibition. In total, 42 compounds were biologically evaluated and among these we discovered 17 11ß-HSD inhibitors that decreased the residual enzyme activity to 50% or less at the concentration of 20 µM. The experimental 11ß-HSD1 and 2 readouts from these compounds were used for further model refinement. Evaluation metrics were applied for a quantitative comparison of the old and newly generated models which resulted in a set of improved pharmacophore models offering reliable in silico tools for the identification of novel and selective 11ß-HSD inhibitors.

3.
Int J Cell Biol ; 2013: 769536, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24348564

RESUMEN

17 ß -Hydroxysteroid dehydrogenase type 1 (17 ß -HSD1) catalyzes the conversion of estrone to the potent estrogen estradiol. 17 ß -HSD1 is highly expressed in breast and ovary tissues and represents a prognostic marker for the tumor progression and survival of patients with breast cancer and other estrogen-dependent tumors. Therefore, the enzyme is considered a promising drug target against estrogen-dependent cancers. For the development of novel inhibitors, an improved understanding of the structure-function relationships is essential. In the present study, we examined the role of a cysteine residue, Cys(10), in the Rossmann-fold NADPH binding region, for 17 ß -HSD1 function and tested the sensitivity towards sulfhydryl modifying chemicals. 3D structure modeling revealed important interactions of Cys(10) with residues involved in the stabilization of amino acids of the NADPH binding pocket. Analysis of enzyme activity revealed that 17 ß -HSD1 was irreversibly inhibited by the sulfhydryl modifying agents N-ethylmaleimide (NEM) and dithiocarbamates. Preincubation with increasing concentrations of NADPH protected 17 ß -HSD1 from inhibition by these chemicals. Cys(10)Ser mutant 17 ß -HSD1 was partially protected from inhibition by NEM and dithiocarbamates, emphasizing the importance of Cys(10) in the cofactor binding region. Substitution of Cys(10) with serine resulted in a decreased protein half-life, without significantly altering kinetic properties. Despite the fact that Cys(10) on 17 ß -HSD1 seems to have limited potential as a target for new enzyme inhibitors, the present study provides new insight into the structure-function relationships of this enzyme.

4.
Endocrinology ; 154(1): 205-13, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23183177

RESUMEN

Recent studies proposed a functional coupling between 17ß-hydroxysteroid dehydrogenase 3 (17ß-HSD3)-dependent testosterone formation and 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1)-mediated interconversion of glucocorticoids through competition for the luminal pyridine nucleotide pool. To test this hypothesis, we used human embryonic kidney-293 cells transfected with 17ß-HSD3 and/or 11ß-HSD1, in the absence or presence of hexose-6-phosphate dehydrogenase that generates reduced nicotinamide adenine dinucleotide phosphate (NADPH) in the endoplasmic reticulum and determined enzyme activities. As an endogenous cell model, mouse MA-10 Leydig cells were used. 17ß-HSD3-dependent reduction of Δ4-androstene-3,17-dione was affected by neither coexpression with 11ß-HSD1 nor overexpression or knockdown of hexose-6-phosphate dehydrogenase. In contrast, knockdown of glucose-6-phosphate dehydrogenase decreased 17ß-HSD3 activity, indicating dependence on cytoplasmic NADPH. Upon selective permeabilization of the plasma membrane by digitonin, 17ß-HSD3 but not 11ß-HSD1 was detected by antibodies against C-terminal epitope tags, suggesting a cytoplasmic orientation of 17ß-HSD3. The cytoplasmic orientation was confirmed using proteinase K digestion of microsomal preparations and by analysis of glycosylation of wild-type 17ß-HSD3 and chimera in which the N-terminal anchor sequences between 17ß-HSD3 and 11ß-HSD1 were exchanged. In conclusion, the results demonstrate a cytoplasmic orientation of 17ß-HSD3 and dependence on glucose-6-phosphate dehydrogenase-generated NADPH, explaining the lack of a direct functional coupling with the luminal 11ß-HSD1-mediated glucocorticoid metabolism.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Deshidrogenasas de Carbohidratos/metabolismo , Citoplasma/metabolismo , Microsomas/enzimología , NADP/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Línea Celular , Humanos , Masculino , Ratones , Modelos Biológicos , Unión Proteica
5.
J Neuroinflammation ; 9: 260, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23190711

RESUMEN

BACKGROUND: Microglia, the resident macrophage-like cells in the brain, regulate innate immune responses in the CNS to protect neurons. However, excessive activation of microglia contributes to neurodegenerative diseases. Corticosteroids are potent modulators of inflammation and mediate their effects by binding to mineralocorticoid receptors (MR) and glucocorticoid receptors (GR). Here, the coordinated activities of GR and MR on the modulation of the nuclear factor-κB (NF-κB) pathway in murine BV-2 microglial cells were studied. METHODS: BV-2 cells were treated with different corticosteroids in the presence or absence of MR and GR antagonists. The impact of the glucocorticoid-activating enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) was determined by incubating cells with 11-dehydrocorticosterone, with or without selective inhibitors. Expression of interleukin-6 (IL-6), tumor necrosis factor receptor 2 (TNFR2), and 11ß-HSD1 mRNA was analyzed by RT-PCR and IL-6 protein expression by ELISA. NF-κB activation and translocation upon treatment with various corticosteroids were visualized by western blotting, immunofluorescence microscopy, and translocation assays. RESULTS: GR and MR differentially regulate NF-κB activation and neuroinflammatory parameters in BV-2 cells. By converting inactive 11-dehydrocorticosterone to active corticosterone, 11ß-HSD1 essentially modulates the coordinated action of GR and MR. Biphasic effects were observed for 11-dehydrocorticosterone and corticosterone, with an MR-dependent potentiation of IL-6 and tumor necrosis factor-α (TNF-α) expression and NF-κB activation at low/moderate concentrations and a GR-dependent suppression at high concentrations. The respective effects were confirmed using the MR ligand aldosterone and the antagonist spironolactone as well as the GR ligand dexamethasone and the antagonist RU-486. NF-κB activation could be blocked by spironolactone and the inhibitor of NF-κB translocation Cay-10512. Moreover, an increased expression of TNFR2 was observed upon treatment with 11-dehydrocorticosterone and aldosterone, which was reversed by 11ß-HSD1 inhibitors and/or spironolactone and Cay-10512. CONCLUSIONS: A tightly coordinated GR and MR activity regulates the NF-κB pathway and the control of inflammatory mediators in microglia cells. The balance of GR and MR activity is locally modulated by the action of 11ß-HSD1, which is upregulated by pro-inflammatory mediators and may represent an important feedback mechanism involved in resolution of inflammation.


Asunto(s)
Citocinas/metabolismo , Microglía/metabolismo , FN-kappa B/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/fisiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Corticoesteroides/farmacología , Análisis de Varianza , Animales , Línea Celular Transformada , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citocinas/genética , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Microglía/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
6.
PLoS One ; 7(10): e46958, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23056542

RESUMEN

BACKGROUND: Impaired corticosteroid action caused by genetic and environmental influence, including exposure to hazardous xenobiotics, contributes to the development and progression of metabolic diseases, cardiovascular complications and immune disorders. Novel strategies are thus needed for identifying xenobiotics that interfere with corticosteroid homeostasis. 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2) and mineralocorticoid receptors (MR) are major regulators of corticosteroid action. 11ß-HSD2 converts the active glucocorticoid cortisol to the inactive cortisone and protects MR from activation by glucocorticoids. 11ß-HSD2 has also an essential role in the placenta to protect the fetus from high maternal cortisol concentrations. METHODS AND PRINCIPAL FINDINGS: We employed a previously constructed 3D-structural library of chemicals with proven and suspected endocrine disrupting effects for virtual screening using a chemical feature-based 11ß-HSD pharmacophore. We tested several in silico predicted chemicals in a 11ß-HSD2 bioassay. The identified antibiotic lasalocid and the silane-coupling agent AB110873 were found to concentration-dependently inhibit 11ß-HSD2. Moreover, the silane AB110873 was shown to activate MR and stimulate mitochondrial ROS generation and the production of the proinflammatory cytokine interleukin-6 (IL-6). Finally, we constructed a MR pharmacophore, which successfully identified the silane AB110873. CONCLUSIONS: Screening of virtual chemical structure libraries can facilitate the identification of xenobiotics inhibiting 11ß-HSD2 and/or activating MR. Lasalocid and AB110873 belong to new classes of 11ß-HSD2 inhibitors. The silane AB110873 represents to the best of our knowledge the first industrial chemical shown to activate MR. Furthermore, the MR pharmacophore can now be used for future screening purposes.


Asunto(s)
Corticoesteroides/metabolismo , Disruptores Endocrinos/farmacología , Xenobióticos/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Bioensayo , Células COS , Chlorocebus aethiops , Bases de Datos Farmacéuticas , Evaluación Preclínica de Medicamentos , Disruptores Endocrinos/química , Disruptores Endocrinos/metabolismo , Disruptores Endocrinos/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Interleucina-6/metabolismo , Microglía/citología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Simulación del Acoplamiento Molecular , Conformación Proteica , Receptores de Mineralocorticoides/metabolismo , Silanos/química , Superóxidos/metabolismo , Interfaz Usuario-Computador , Xenobióticos/química , Xenobióticos/metabolismo , Xenobióticos/toxicidad
7.
J Steroid Biochem Mol Biol ; 119(1-2): 1-13, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20100573

RESUMEN

The primary function of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) is to catalyze the conversion of inactive to active glucocorticoid hormones and to modulate local glucocorticoid-dependent gene expression. Thereby 11beta-HSD1 plays a key role in the regulation of metabolic functions and in the adaptation of the organism to energy requiring situations. Importantly, elevated 11beta-HSD1 activity has been associated with metabolic disorders, and recent investigations with rodent models of obesity and type 2 diabetes provided evidence for beneficial effects of 11beta-HSD1 inhibitors, making this enzyme a promising therapeutic target. Several earlier and recent studies, mainly performed in vitro, revealed a relatively broad substrate spectrum of 11beta-HSD1 and suggested that this enzyme has additional functions in the metabolism of some neurosteroids (7-oxy- and 11-oxyandrogens and -progestins) and 7-oxysterols, as well as in the detoxification of various xenobiotics that contain reactive carbonyl groups. While there are many studies on the effect of inhibitors on cortisone reduction and circulating glucocorticoid levels and on the transcriptional regulation of 11beta-HSD1 in obesity and diabetes, only few address the so-called alternative functions of this enzyme. We review recent progress on the biochemical characterization of 11beta-HSD1, with a focus on cofactor and substrate specificity and on possible alternative functions of this enzyme.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/fisiología , Glucocorticoides/efectos adversos , Glucocorticoides/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , Secuencia de Aminoácidos , Animales , Deshidrogenasas de Carbohidratos/metabolismo , Glucocorticoides/farmacocinética , Humanos , Modelos Biológicos , NADP/metabolismo , Conformación Proteica , Especificidad por Sustrato , Toxicología
8.
Biochem Pharmacol ; 79(8): 1189-99, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20005209

RESUMEN

The prevalence of male reproductive disorders and testicular cancer is steadily increasing. Because the exposure to chemicals disrupting natural hormone action has been associated with these diseases, it is important to identify endocrine disrupting chemicals (EDCs) and their targets of action. Here, a 3D-structural database that can be applied for virtual screening approaches to facilitate the identification of EDCs was constructed. The database was screened using pharmacophores of 17beta-hydroxysteroid dehydrogenase type 3 (17beta-HSD3), which catalyzes the last step of testosterone synthesis in testicular Leydig cells and plays an essential role during male sexual development. Among other chemicals, benzophenone (BP) UV-filters were predicted as potential 17beta-HSD3 inhibitors. Biological analyses revealed (2,4-dihydroxyphenyl)-phenylmethanone (also known as benzophenone-1, BP-1) as an inhibitor of human 17beta-HSD3 (IC(50) 1.05microM). BP-1 also efficiently blocked conversion of androstenedione to testosterone by mouse and rat 17beta-HSD3 in whole-organ enzyme assays. Moreover, BP-1 antagonized the testosterone-dependent activation of androgen receptors (IC(50) 5.7microM), suggesting synergistic anti-androgenic effects of BP-1 by preventing testosterone formation and blocking receptor activation. In addition, analyses of several commonly used UV-filters on estrogen- and androgen-metabolizing 17beta-HSD enzymes revealed 3-benzylidene camphor (3-BC) and 4-methylbenzylidene camphor (4-MBC) as low micromolar 17beta-HSD2 inhibitors. In conclusion, screening of virtual chemical structure libraries can facilitate the identification of compounds interfering with hormone action. The potential disruption of 17beta-HSD enzyme function by the UV-filters BP-1, 3-BC and 4-MBC requires further investigation and should be considered for safety assessment of these chemicals.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Benzofenonas/farmacología , Disruptores Endocrinos/farmacología , Inhibidores Enzimáticos/farmacología , Antagonistas de Receptores Androgénicos , Animales , Bases de Datos Factuales , Masculino , Ratones , Ratas , Receptores Androgénicos , Relación Estructura-Actividad , Testículo/metabolismo , Testosterona/biosíntesis
9.
Mol Cell Endocrinol ; 301(1-2): 117-22, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19010388

RESUMEN

Intracellular glucocorticoid reactivation is catalyzed by 11beta-hydroxysteroid dehydrogenase 1 (11beta-HSD1), which functions predominantly as a reductase in cells expressing hexose-6-phosphate dehydrogenase (H6PDH). We recently showed that the ratios of cortisone to cortisol and 7-keto- to 7-hydroxy-neurosteroids are regulated by 11beta-HSD1 and very much depend on coexpression with H6PDH, providing cosubstrate NADPH. Here, we investigated the impact of H6PDH on the modulation of 11beta-HSD1-dependent interconversion of cortisone and cortisol by inhibitors and alternative substrates. Using HEK-293 cells expressing 11beta-HSD1 or coexpressing 11beta-HSD1 and H6PDH, we observed significant differences of 11beta-HSD1 inhibition by natural and pharmaceutical compounds as well as endogenous hormone metabolites. Furthermore, we show potent and dose-dependent inhibition of 11beta-HSD1 by 7-keto-DHEA in differentiated human THP-1 macrophages and in HEK-293 cells overexpressing 11beta-HSD1 with or without H6PDH. In contrast, 7-ketocholesterol (7-KC) did not inhibit 11beta-HSD1 in HEK-293 cells, even in the presence of H6PDH, but inhibited 11beta-HSD1 reductase activity in differentiated THP-1 macrophages (IC(50) 8.1+/-0.9microM). 7-Keto-DHEA but not 7-KC inhibited 11beta-HSD1 in HEK-293 cell lysates. In conclusion, cellular factors such as H6PDH can significantly modulate the effect of inhibitors and alternative 7-oxygenated substrates on intracellular glucocorticoid availability.


Asunto(s)
Deshidrogenasas de Carbohidratos/metabolismo , Inhibidores Enzimáticos/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Extractos Celulares , Línea Celular , Corticosterona/química , Corticosterona/metabolismo , Deshidroepiandrosterona/metabolismo , Humanos , Cetocolesteroles/química , Cetocolesteroles/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Modelos Moleculares , Especificidad por Sustrato/efectos de los fármacos
10.
Mol Cell Endocrinol ; 301(1-2): 137-41, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18778749

RESUMEN

To assess the impact of the NADPH/NADP(+) ratio and the influence of extracellular glucose on 11beta-hydroxysteroid dehydrogenase 1 (11beta-HSD1) activity, we applied microsomal preparations and intact HEK-293 cells expressing 11beta-HSD1 in the presence or absence of hexose-6-phosphate dehydrogenase (H6PDH). A NADPH/NADP(+) ratio of ten or higher was required for efficient microsomal 11beta-HSD1 reductase activity. Measurements in intact cells suggested that the ER-luminal NADPH concentration is highly sensitive to fluctuating extracellular glucose levels. Lowering glucose in the culture medium dose-dependently decreased 11beta-HSD1 reductase activity and diminished the cortisol/cortisone ratio measured after 24h of incubation. Coexpression with H6PDH potentiated 11beta-HSD1 reductase activity at high glucose. This effect was significantly decreased at low glucose, with concomitantly increased 11beta-HSD1 dehydrogenase activity. In contrast, 11beta-HSD1 reductase activity in H4IIE liver cells and in 3T3-L1 adipocytes was less sensitive to changes in the medium. 11beta-HSD1 dehydrogenase activity was observed in H4IIE cells only at subphysiological glucose levels, indicating a highly efficient supply of substrate for H6PDH and NADPH generation in the ER-lumen. Our results suggest significant cell type-specific differences in ER-luminal NADPH generation that might allow a fine-tuned regulation of glucocorticoid action.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Glucosa/farmacología , NADP/metabolismo , Animales , Glucocorticoides/metabolismo , Humanos , Ratones , Ratas , Receptores de Glucocorticoides/metabolismo , Activación Transcripcional/efectos de los fármacos
11.
J Med Chem ; 51(14): 4188-99, 2008 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-18533708

RESUMEN

17Beta-hydroxysteroid dehydrogenase type 1 (17beta-HSD1) plays a pivotal role in the local synthesis of the most potent estrogen estradiol. Its expression is a prognostic marker for the outcome of patients with breast cancer and inhibition of 17beta-HSD1 is currently under consideration for breast cancer prevention and treatment. We aimed to identify nonsteroidal 17beta-HSD1 inhibitor scaffolds by virtual screening with pharmacophore models built from crystal structures containing steroidal compounds. The most promising model was validated by comparing predicted and experimentally determined inhibitory activities of several flavonoids. Subsequently, a virtual library of nonsteroidal compounds was screened against the 3D pharmacophore. Analysis of 14 selected compounds yielded four that inhibited the activity of human 17beta-HSD1 (IC 50 below 50 microM). Specificity assessment of identified 17beta-HSD1 inhibitors emphasized the importance of including related short-chain dehydrogenase/reductase (SDR) members to analyze off-target effects. Compound 29 displayed at least 10-fold selectivity over the related SDR enzymes tested.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Modelos Químicos , Catálisis , Línea Celular , Evaluación Preclínica de Medicamentos , Flavonoides/química , Flavonoides/farmacología , Humanos , Bibliotecas de Moléculas Pequeñas
12.
Biochim Biophys Acta ; 1783(8): 1536-43, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18381077

RESUMEN

Hexose-6-phosphate dehydrogenase (H6PDH) has been shown to stimulate 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1)-dependent local regeneration of active glucocorticoids. Here, we show that coexpression with H6PDH results in a dramatic shift from 11beta-HSD1 oxidase to reductase activity without affecting the activity of the endoplasmic reticular enzyme 17beta-HSD2. Immunoprecipitation experiments revealed coprecipitation of H6PDH with 11beta-HSD1 but not with the related enzymes 11beta-HSD2 and 17beta-HSD2, suggesting a specific interaction between H6PDH and 11beta-HSD1. The use of the 11beta-HSD1/11beta-HSD2 chimera indicates that the N-terminal 39 residues of 11beta-HSD1 are sufficient for interaction with H6PDH. An important role of the N-terminus was indicated further by the significantly stronger interaction of 11beta-HSD1 mutant Y18-21A with H6PDH compared to wild-type 11beta-HSD1. The protein-protein interaction and the involvement of the N-terminus of 11beta-HSD1 were confirmed by Far-Western blotting. Finally, fluorescence resonance energy transfer (FRET) measurements of HEK-293 cells expressing fluorescently labeled proteins provided evidence for an interaction between 11beta-HSD1 and H6PDH in intact cells. Thus, using three different methods, we provide strong evidence that the functional coupling between 11beta-HSD1 and H6PDH involves a direct physical interaction of the two proteins.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Deshidrogenasas de Carbohidratos/metabolismo , Retículo Endoplásmico/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , Far-Western Blotting , Deshidrogenasas de Carbohidratos/análisis , Deshidrogenasas de Carbohidratos/aislamiento & purificación , Línea Celular , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunoprecipitación , NADP/metabolismo , Dominios y Motivos de Interacción de Proteínas
13.
PLoS One ; 2(6): e561, 2007 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-17593962

RESUMEN

BACKGROUND: The role of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) in the regulation of energy metabolism and immune system by locally reactivating glucocorticoids has been extensively studied. Experiments determining initial rates of enzyme activity revealed that 11beta-HSD1 can catalyze both the reductase and the dehydrogenase reaction in cell lysates, whereas it predominantly catalyzes the reduction of cortisone to cortisol in intact cells that also express hexose-6-phosphate dehydrogenase (H6PDH), which provides cofactor NADPH. Besides its role in glucocorticoid metabolism, there is evidence that 11beta-HSD1 is involved in the metabolism of 7-keto- and 7-hydroxy-steroids; however the impact of H6PDH on this alternative function of 11beta-HSD1 has not been assessed. METHODOLOGY: We investigated the 11beta-HSD1-dependent metabolism of the neurosteroids 7-keto-, 7alpha-hydroxy- and 7beta-hydroxy-dehydroepiandrosterone (DHEA) and 7-keto- and 7beta-hydroxy-pregnenolone, respectively, in the absence or presence of H6PDH in intact cells. 3D-structural modeling was applied to study the binding of ligands in 11beta-HSD1. PRINCIPAL FINDINGS: We demonstrated that 11beta-HSD1 functions in a reversible way and efficiently catalyzed the interconversion of these 7-keto- and 7-hydroxy-neurosteroids in intact cells. In the presence of H6PDH, 11beta-HSD1 predominantly converted 7-keto-DHEA and 7-ketopregnenolone into their corresponding 7beta-hydroxy metabolites, indicating a role for H6PDH and 11beta-HSD1 in the local generation of 7beta-hydroxy-neurosteroids. 3D-structural modeling offered an explanation for the preferred formation of 7beta-hydroxy-neurosteroids. CONCLUSIONS: Our results from experiments determining the steady state concentrations of glucocorticoids or 7-oxygenated neurosteroids suggested that the equilibrium between cortisone and cortisol and between 7-keto- and 7-hydroxy-neurosteroids is regulated by 11beta-HSD1 and greatly depends on the coexpression with H6PDH. Thus, the impact of H6PDH on 11beta-HSD1 activity has to be considered for understanding both glucocorticoid and neurosteroid action in different tissues.


Asunto(s)
Deshidrogenasas de Carbohidratos/metabolismo , Deshidroepiandrosterona/metabolismo , Neurotransmisores/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Células Cultivadas , Cortisona/metabolismo , Deshidroepiandrosterona/química , Humanos , Hidrocortisona/metabolismo , Riñón/citología , Riñón/metabolismo , Modelos Moleculares , Neurotransmisores/química , Especificidad por Sustrato
14.
J Am Soc Nephrol ; 18(4): 1262-70, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17314322

RESUMEN

Apparent mineralocorticoid excess (AME) is a severe form of hypertension that is caused by impaired activity of 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2), which converts biologically active cortisol into inactive cortisone. Mutations in HSD11B2 result in cortisol-induced activation of mineralocorticoid receptors and cause hypertension with hypokalemia, metabolic alkalosis, and suppressed circulating renin and aldosterone concentrations. This study uncovered the first patient with AME who was described in the literature, identified the genetic defect in HSD11B2, and provided evidence for a novel mechanism of reduced 11beta-HSD2 activity. This study identified a cluster of amino acids (335 to 339) in the C-terminus of 11beta-HSD2 that are essential for protein stability. The cluster includes Tyr(338), which is mutated in the index patient, and Arg(335) and Arg(337), previously reported to be mutated in hypertensive patients. It was found that wild-type 11beta-HSD2 is a relatively stable enzyme with a half-life of 21 h, whereas that of Tyr(338)His and Arg(337)His was 3 and 4 h, respectively. Enzymatic activity of Tyr(338)His was partially retained at 26 degrees C or in the presence of the chemical chaperones glycerol and dexamethasone, indicating thermodynamic instability and misfolding. The results provide evidence that the degradation of both misfolded mutant Tyr(338)His and wild-type 11beta-HSD2 occurs through the proteasome pathway. Therefore, impaired 11beta-HSD2 protein stability rather than reduced gene expression or loss of catalytic activity seems to be responsible for the development of hypertension in some individuals with AME.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/química , Hipertensión/etiología , Síndrome de Exceso Aparente de Mineralocorticoides/etiología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Cortisona/sangre , Estabilidad de Enzimas , Humanos , Hidrocortisona/sangre , Mutación , Pliegue de Proteína , Termodinámica
15.
FEBS Lett ; 580(17): 4081-5, 2006 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-16814782

RESUMEN

Recent epidemiological studies demonstrated a beneficial effect of coffee consumption for the prevention of type 2 diabetes, however, the underlying mechanisms remained unknown. We demonstrate that coffee extract, corresponding to an Italian Espresso, inhibits recombinant and endogenous 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) activity. The inhibitory component is heat-stable with considerable polarity. Coffee extract blocked 11beta-HSD1-dependent cortisol formation, prevented the subsequent nuclear translocation of the glucocorticoid receptor and abolished glucocorticoid-induced expression of the key gluconeogenic enzyme phosphoenolpyruvate carboxykinase. We suggest that at least part of the anti-diabetic effects of coffee consumption is due to inhibition of 11beta-HSD1-dependent glucocorticoid reactivation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Coffea , Café , Diabetes Mellitus Tipo 2/enzimología , Extractos Vegetales/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Carboxiliasas/biosíntesis , Línea Celular , Núcleo Celular/metabolismo , Coffea/química , Café/química , Diabetes Mellitus Tipo 2/prevención & control , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucocorticoides/metabolismo , Gluconeogénesis/efectos de los fármacos , Humanos , Hidrocortisona/biosíntesis , Extractos Vegetales/química , Receptores de Glucocorticoides/metabolismo
16.
J Med Chem ; 49(12): 3454-66, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16759088

RESUMEN

11beta-Hydroxysteroid dehydrogenase (11beta-HSD) enzymes catalyze the conversion of biologically inactive 11-ketosteroids into their active 11beta-hydroxy derivatives and vice versa. Inhibition of 11beta-HSD1 has considerable therapeutic potential for glucocorticoid-associated diseases including obesity, diabetes, wound healing, and muscle atrophy. Because inhibition of related enzymes such as 11beta-HSD2 and 17beta-HSDs causes sodium retention and hypertension or interferes with sex steroid hormone metabolism, respectively, highly selective 11beta-HSD1 inhibitors are required for successful therapy. Here, we employed the software package Catalyst to develop ligand-based multifeature pharmacophore models for 11beta-HSD1 inhibitors. Virtual screening experiments and subsequent in vitro evaluation of promising hits revealed several selective inhibitors. Efficient inhibition of recombinant human 11beta-HSD1 in intact transfected cells as well as endogenous enzyme in mouse 3T3-L1 adipocytes and C2C12 myotubes was demonstrated for compound 27, which was able to block subsequent cortisol-dependent activation of glucocorticoid receptors with only minor direct effects on the receptor itself. Our results suggest that inhibitor-based pharmacophore models for 11beta-HSD1 in combination with suitable cell-based activity assays, including such for related enzymes, can be used for the identification of selective and potent inhibitors.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , Inhibidores Enzimáticos/química , Modelos Moleculares , Animales , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Bases de Datos Factuales , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/toxicidad , Humanos , Hidrocortisona/fisiología , Ligandos , Ratones , Conformación Proteica , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/genética , Programas Informáticos , Activación Transcripcional/efectos de los fármacos , Transfección
17.
Mol Cell Endocrinol ; 248(1-2): 15-23, 2006 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-16412558

RESUMEN

11Beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) is essential for the local activation of glucocorticoid receptors (GR). Unlike unliganded cytoplasmic GR, 11beta-HSD1 is an endoplasmic reticulum (ER)-membrane protein with lumenal orientation. Cortisone might gain direct access to 11beta-HSD1 by free diffusion across membranes, indirectly via intracellular binding proteins or, alternatively, by insertion into membranes. Membranous cortisol, formed by 11beta-HSD1 at the ER-lumenal side, might then activate cytoplasmic GR or bind to ER-lumenal secretory proteins. Compartmentalization of 11beta-HSD1 is important for its regulation by hexose-6-phosphate dehydrogenase (H6PDH), which regenerates cofactor NADPH in the ER lumen and stimulates oxoreductase activity. ER-lumenal orientation of 11beta-HSD1 is also essential for the metabolism of the alternative substrate 7-ketocholesterol (7KC), a major cholesterol oxidation product found in atherosclerotic plaques and taken up from processed cholesterol-rich food. An 11beta-HSD1 mutant adopting cytoplasmic orientation efficiently catalyzed the oxoreduction of cortisone but not 7KC, indicating access to cortisone from both sides of the ER-membrane but to 7KC only from the lumenal side. These aspects may be relevant for understanding the physiological role of 11beta-HSD1 and for developing therapeutic interventions to control glucocorticoid reactivation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/fisiología , Membrana Celular/efectos de los fármacos , Retículo Endoplásmico/enzimología , Glucocorticoides/farmacología , Cetocolesteroles/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Secuencia de Aminoácidos , Deshidrogenasas de Carbohidratos/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/enzimología , Glucocorticoides/metabolismo , Humanos , Datos de Secuencia Molecular
18.
Environ Health Perspect ; 113(11): 1600-6, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16263518

RESUMEN

Organotins, important environmental pollutants widely used in agricultural and industrial applications, accumulate in the food chain and induce imposex in several marine species as well as neurotoxic and immunotoxic effects in higher animals. Reduced birth weight and thymus involution, observed upon exposure to organotins, can also be caused by excessive glucocorticoid levels. We now demonstrate that organotins efficiently inhibit 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2), converting active 11beta-hydroxyglucocorticoids into inactive 11-ketoglucocorticoids, but not 11beta-HSD1, which catalyzes the reverse reaction. Di- and tributyltin as well as di- and triphenyltin inhibited recombinant and endogenous 11beta-HSD2 in lysates and intact cells with IC50 values between 500 nM and 3 microM. Dithiothreitol protected 11beta-HSD2 from organotin-dependent inhibition, indicating that organotins act by binding to one or more cysteines. Mutational analysis and 3-D structural modeling revealed several important interactions of cysteines in 11beta-HSD2. Cys90, Cys228, and Cys264 were essential for enzymatic stability and catalytic activity, suggesting that disruption of such interactions by organotins leads to inhibition of 11beta-HSD2. Enhanced glucocorticoid concentrations due to disruption of 11beta-HSD2 function may contribute to the observed organotin-dependent toxicity in some glucocorticoid-sensitive tissues such as thymus and placenta.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/antagonistas & inhibidores , Glucocorticoides/metabolismo , Compuestos Orgánicos de Estaño/toxicidad , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Línea Celular , Ditiotreitol/farmacología , Humanos , Mutagénesis Sitio-Dirigida , Transfección
19.
Bioinformatics ; 21(3): 418-20, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15374875

RESUMEN

UNLABELLED: We have created PhenomicDB, a multi-species genotype/phenotype database by merging public genotype/phenotype data from a wide range of model organisms and Homo sapiens. Until now these data were available in distinct organism-specific databases (e.g. WormBase, OMIM, FlyBase and MGI). We compiled this wealth of data into a single integrated resource by coarse-grained semantic mapping of the phenotypic data fields, by including common gene indices (NCBI Gene), and by the use of associated orthology relationships. With its use-case-oriented user interface, PhenomicDB allows scientists to compare and browse known phenotypes for a given gene or a set of genes from different organisms simultaneously. AVAILABILITY: PhenomicDB has been implemented at Schering AG as described below. A PhenomicDB implementation differing in some technical details has been set up for the public at Metalife AG http://www.phenomicDB.de SUPPLEMENTARY INFORMATION: database model, semantic mapping table.


Asunto(s)
Bases de Datos Genéticas , Documentación/métodos , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Difusión de la Información/métodos , Almacenamiento y Recuperación de la Información/métodos , Animales , Genotipo , Humanos , Fenotipo , Especificidad de la Especie
20.
FEBS Lett ; 571(1-3): 129-33, 2004 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-15280030

RESUMEN

The impact of hexose-6-phosphate dehydrogenase (H6PDH) on 11beta-hydroxysteroid dehydrogenase (11beta-HSD) type 1 activity was investigated upon coexpression in HEK-293 cells. Confocal microscopy analysis indicated colocalisation of both enzymes at the lumenal side of the endoplasmic reticulum (ER) membrane. Functional analysis in intact cells revealed fivefold stimulation of 11beta-HSD1 oxoreductase activity and sixfold decrease of dehydrogenase activity upon coexpression with H6PDH, without changing kinetic parameters in cell lysates. Thus, H6PDH directly determines the reaction direction of 11beta-HSD1 in intact cells as an oxoreductase without changing intrinsic catalytic properties of 11beta-HSD1 by regenerating NADPH in the ER-lumen.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasas/metabolismo , Deshidrogenasas de Carbohidratos/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasas/genética , Regiones no Traducidas 5'/genética , Adipocitos/enzimología , Secuencia de Bases , Deshidrogenasas de Carbohidratos/genética , Línea Celular , Clonación Molecular , Cortisona/metabolismo , Humanos , Hidrocortisona/metabolismo , Riñón , Cinética , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...