Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Recept Signal Transduct Res ; 41(2): 123-137, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32705921

RESUMEN

Aromatase and steroidal sulfatase (STS) are steroidogenic enzyme that increases the concentration of estrogens in circulation, a primary factor leading to breast cancer. At molecular level, 87% of STS is expressed and an inhibitor targeting STS could decrease the level of estrogens. In an attempt to identify the chemical structural requirement targeting placental STS inhibition, 26 compounds with pIC50 ranging from 4.61 to 9.46 were subjected to computational studies including Quantitative Structural-Activity Relationship (QSAR), MolecularDocking followed by Density Functional Theory (DFT) studies. A robust and predictable model were developed with good R2 (0.834) and cross-validated correlation coefficient value Q2 LOO (0.786) explaining the relationship quantitatively. The regression graphs suggests that the STS inhibition was greatly dependent on the electro topological state of an atom, sum of the atom type E-state (SdssC), maximum E-states for strong hydrogen bond acceptors (maxHBa) and basic group count descriptor (BCUTp-1h). Furthermore, docking results showed favorable interactions of sulfamate analogs with catalytically important amino acid residues such as LEU74, VAL101, and VAL486. The interactions of the best active compound 3j when compared with standard Irosustat show similar binding energies. DFT studies further confirm the presence of HOMO orbital centered on chromenone ring further highlighting its importance for receptor ligand hydrophobic interaction. The study reveals that substitution of thio in chromenone nucleus and introduction of adamantyl substitution at second position are favorable in inhibiting the enzyme STS.


Asunto(s)
Inhibidores Enzimáticos/química , Antagonistas de Estrógenos/química , Relación Estructura-Actividad Cuantitativa , Esteril-Sulfatasa/ultraestructura , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Inhibidores Enzimáticos/uso terapéutico , Antagonistas de Estrógenos/uso terapéutico , Estrógenos/metabolismo , Femenino , Humanos , Enlace de Hidrógeno/efectos de los fármacos , Simulación del Acoplamiento Molecular , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/química , Esteril-Sulfatasa/genética
2.
J Enzyme Inhib Med Chem ; 35(1): 1163-1184, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32363947

RESUMEN

The purpose of this review article is to provide an overview of recent achievements in the synthesis of novel steroid sulphatase (STS) inhibitors. STS is a crucial enzyme in the biosynthesis of active hormones (including oestrogens and androgens) and, therefore, represents an extremely attractive molecular target for the development of hormone-dependent cancer therapies. The inhibition of STS may effectively reduce the availability of active hormones for cancer cells, causing a positive therapeutic effect. Herein, we report examples of novel STS inhibitors based on steroidal and nonsteroidal cores that contain various functional groups (e.g. sulphamate and phosphorus moieties) and halogen atoms, which may potentially be used in therapies for hormone-dependent cancers. The presented work also includes examples of multitargeting agents with STS inhibitory activities. Furthermore, the fundamental discoveries in the development of the most promising drug candidates exhibiting STS inhibitory activities are highlighted.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo
3.
Drug Dev Res ; 80(6): 857-866, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31301181

RESUMEN

In the present work, we described convenient methods for the synthesis of N-thiophosphorylated 3-(4-aminophenyl)-coumarin-7-O-sulfamates as steroid sulfatase (STS) inhibitors. To design the structures of the potential STS inhibitors, molecular modeling techniques were used. A computational docking method was used to determine the binding modes of the synthesized inhibitors as well as to identify potential interactions between specified functional groups on the inhibitors and the amino acid residues present in the active site of the enzyme. The inhibitory activities of the synthesized compounds were tested in an enzymatic assay with STS isolated from a human placenta. Within the set of newly synthesized compounds, 9e demonstrated the highest inhibitory activity in the enzymatic assay with an IC50 value of 0.201 µM (the IC50 value of 667-COUMATE in the same test was 0.062 µM). Furthermore, we tried to verify if the obtained STS inhibitors are able to pass through the cellular membrane effectively in cell line experiments. In the course of our study, we determined the STS activity in the MCF-7 cell line after incubation in the presence of the inhibitors (at 100 nM concentration). For this evaluation, we included newly synthesized compounds 9a-g and their N-phosphorylated analogs 6a-h, whose synthesis has been previously described. We found that the lowest STS activities were measured in the presence of N-phosphorylated derivatives 6e (0.1% of STS activity) and 6f (0.2% of STS activity). The measured STS activity in the presence of 667-COUMATE (used as a reference) was 0.1%. Moreover, at concentrations up to 1 µM, the most active compounds (6e, 6f, 9b, and 9e) did not exert any toxic effects on zebrafish embryos.


Asunto(s)
Cumarinas/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Cumarinas/química , Embrión no Mamífero , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo , Sulfonamidas/química , Pez Cebra
4.
Comput Biol Chem ; 68: 71-77, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28257906

RESUMEN

At least 19 sulfatase genes have been reported on the human genome, including four arylsulfatase (ARS) genes (ARSD; ARSE; ARSF; ARSH) and a sterylsulfatase (STS) gene located together on the X-chromosome. Bioinformatic analyses of mammalian genomes were undertaken using known human STS and ARS amino acid sequences to study the evolution of these genes and proteins encoded on eutherian and marsupial genomes. Several domain regions and key residues were conserved including signal peptides, active site residues, metal (Ca2+) and substrate binding sequences, transmembranes and N-glycosylation sites. Phylogenetic analyses describe the relationships and potential origins of these genes during mammalian evolution. Primate ARSH enzymes lacked signal peptide sequences which may influence their biological functions. CpG117 and CpG92 were detected within the 5' region of the human STS and ARSD genes, respectively, and miR-205 within the 3'-UTR for the human STS gene, using bioinformatic methods A proposal is described for a primordial invertebrate STS-like gene serving as an ancestor for unequal cross over events generating the gene complex on the eutherian mammalian X-chromosome.


Asunto(s)
Arilsulfatasas/química , Arilsulfatasas/genética , Evolución Molecular , Genes Ligados a X/genética , Esteril-Sulfatasa/química , Esteril-Sulfatasa/genética , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Alineación de Secuencia , Strongylocentrotus purpuratus/química , Strongylocentrotus purpuratus/genética
5.
Eur J Med Chem ; 119: 169-82, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27155470

RESUMEN

Steroid sulfatase (STS), the enzyme which converts inactive sulfated steroid precursors into active hormones, is a promising therapeutic target for the treatment of estrogen-sensitive breast cancer. We report herein the synthesis and in vitro study of dual-action STS inhibitors with selective estrogen-receptor modulator (SERM) effects. A library of tetrahydroisoquinoline-N-substituted derivatives (phenolic compounds) was synthesized by solid-phase chemistry and tested on estrogen-sensitive breast cancer T-47D cells. Three phenolic compounds devoid of estrogenic activity and toxicity emerged from this screening. Their sulfamate analogs were then synthesized, tested in STS-transfected HEK-293 cells, and found to be potent inhibitors of the enzyme (IC50 of 3.9, 8.9, and 16.6 nM). When tested in T-47D cells they showed no estrogenic activity and produced a moderate antiestrogenic activity. The compounds were further tested on osteoblast-like Saos-2 cells and found to significantly stimulate their proliferation as well as their alkaline phosphatase activity, thus suggesting a SERM activity. These results are supported by molecular docking experiments.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Ácidos Sulfónicos/farmacología , Fosfatasa Alcalina/metabolismo , Aminas/química , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica , Moduladores Selectivos de los Receptores de Estrógeno/química , Moduladores Selectivos de los Receptores de Estrógeno/metabolismo , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo , Ácidos Sulfónicos/química , Ácidos Sulfónicos/metabolismo
7.
Drug Dev Res ; 76(8): 450-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26415657

RESUMEN

A series of phosphate and thiophosphate flavone derivatives were synthesized and biologically evaluated in vitro for inhibition of steroid sulfatase (STS) activity. The described synthesis includes the straightforward preparation of 7-hydroxy-2-phenyl-4H-chromen-4-one 3a, 2-(4-fluorophenyl)-7-hydroxy-4H-chromen-4-one 3b, 7-hydroxy-2-(4-(trifluoromethyl)phenyl)-4H-chromen-4-one 3c, 7-hydroxy-2-(p-tolyl)-4H-chromen-4-one 3d modified with different phosphate or thiophosphate moieties. The inhibitory properties of the synthesized compounds were tested against human placenta STS. Some of the novel STS inhibitors had good activities against STS. In particular, the bis-(4-oxo-2-(p-tolyl)-4H-chromen-7-yl) hydrogenthiophosphate, 6i had the most potent inhibitory effect with an IC50 value of 3.25 µM as compared to an IC50 value of 8.50 µM for the 2-(4-trifluoromethylphenyl)-chromen-4-one-7-O-sulfamate used as a reference.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Flavonas/síntesis química , Flavonas/farmacología , Fosfatos/síntesis química , Fosfatos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Sitios de Unión , Inhibidores Enzimáticos/química , Femenino , Flavonas/química , Humanos , Simulación del Acoplamiento Molecular , Fosfatos/química , Placenta/enzimología , Embarazo , Esteril-Sulfatasa/química , Esteril-Sulfatasa/aislamiento & purificación
8.
J Med Chem ; 58(19): 7634-58, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25992880

RESUMEN

In 1994, following work from this laboratory, it was reported that estrone-3-O-sulfamate irreversibly inhibits a new potential hormone-dependent cancer target steroid sulfatase (STS). Subsequent drug discovery projects were initiated to develop the core aryl O-sulfamate pharmacophore that, over some 20 years, have led to steroidal and nonsteroidal drugs in numerous preclinical and clinical trials, with promising results in oncology and women's health, including endometriosis. Drugs have been designed to inhibit STS, e.g., Irosustat, as innovative dual-targeting aromatase-steroid sulfatase inhibitors (DASIs) and as multitargeting agents for hormone-independent tumors, such as the steroidal STX140 and nonsteroidal counterparts, acting inter alia through microtubule disruption. The aryl sulfamate pharmacophore is highly versatile, operating via three distinct mechanisms of action, and imbues attractive pharmaceutical properties. This Perspective gives a personal view of the work leading both to the therapeutic concepts and these drugs, their current status, and how they might develop in the future.


Asunto(s)
Inhibidores de la Aromatasa/química , Inhibidores de la Aromatasa/farmacología , Descubrimiento de Drogas , Esteril-Sulfatasa/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Endometriosis/tratamiento farmacológico , Estrona/análogos & derivados , Estrona/farmacología , Femenino , Humanos , Masculino , Terapia Molecular Dirigida/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Esteril-Sulfatasa/química , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
9.
J Steroid Biochem Mol Biol ; 153: 160-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25843211

RESUMEN

Estrogen sulfamate derivatives were the first irreversible active-site-directed inhibitors of steroid sulfatase (STS), an emerging drug target for endocrine therapy of hormone dependent diseases that catalyzes inter alia the hydrolysis of estrone sulfate to estrone. In recent years this has stimulated clinical investigation of the estradiol derivative both as an oral prodrug and its currently ongoing exploration in endometriosis. 2-Substituted steroid sulfamate derivatives show considerable potential as multi-targeting agents for hormone-independent disease, but are also potent STS inhibitors. The steroidal template has spawned nonsteroidal STS inhibitors one of which, Irosustat, has been evaluated clinically in breast cancer, endometrial cancer and prostate cancer and there is potential for innovative dual-targeting approaches. This review surveys the role of estrogen sulfamates, their analogues and current status.


Asunto(s)
Cumarinas/farmacología , Inhibidores Enzimáticos/farmacología , Estradiol/análogos & derivados , Estrógenos/farmacología , Estrona/análogos & derivados , Esteril-Sulfatasa/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Cumarinas/química , Cumarinas/uso terapéutico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Estradiol/química , Estradiol/farmacología , Estradiol/uso terapéutico , Estrógenos/análogos & derivados , Estrógenos/uso terapéutico , Estrona/química , Estrona/farmacología , Estrona/uso terapéutico , Humanos , Modelos Moleculares , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo , Sulfonamidas/química , Sulfonamidas/uso terapéutico
10.
Drug Dev Res ; 76(2): 94-104, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25845343

RESUMEN

In the present work, we report convenient methods for the synthesis and biological evaluation of phosphate and thiophosphate biphenyl derivatives exhibiting steroid sulfatase (STS) activity. The described synthesis is based on straightforward preparation of biphenyl-4-ol and 4'-hydroxy-biphenyl-4-carboxylic acid ethyl ester modified with various phosphate or thiophosphate moieties. The inhibitory effects of these compounds were tested on STS isolated from human placenta and led to two compounds of interest, 5a and 5d with IC50 values of 28.0 and 22.1 µM, respectively and that had interesting new binding modes in the STS active site.


Asunto(s)
Compuestos de Bifenilo/síntesis química , Inhibidores Enzimáticos/síntesis química , Placenta/metabolismo , Esteril-Sulfatasa/química , Sitios de Unión , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Simulación del Acoplamiento Molecular , Embarazo , Esteril-Sulfatasa/antagonistas & inhibidores , Relación Estructura-Actividad
11.
J Steroid Biochem Mol Biol ; 137: 27-49, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23291110

RESUMEN

Many enzymes catalyse reactions that have an oestrogen as a substrate and/or a product. The reactions catalysed include aromatisation, oxidation, reduction, sulfonation, desulfonation, hydroxylation and methoxylation. The enzymes that catalyse these reactions must all recognise and bind oestrogen but, despite this, they have diverse structures. This review looks at each of these enzymes in turn, describing the structure and discussing the mechanism of the catalysed reaction. Since oestrogen has a role in many disease states inhibition of the enzymes of oestrogen metabolism may have an impact on the state or progression of the disease and inhibitors of these enzymes are briefly discussed. This article is part of a Special Issue entitled 'CSR 2013'.


Asunto(s)
Estrógenos/metabolismo , 17-Hidroxiesteroide Deshidrogenasas/química , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Secuencia de Aminoácidos , Animales , Aromatasa/química , Aromatasa/metabolismo , Biocatálisis , Catecol O-Metiltransferasa/química , Catecol O-Metiltransferasa/metabolismo , Estrógenos/química , Humanos , Hidroxilación , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Homología de Secuencia de Aminoácido , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo , Sulfotransferasas/química , Sulfotransferasas/metabolismo
12.
Mol Cancer Ther ; 7(8): 2435-44, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723489

RESUMEN

An improved steroid sulfatase inhibitor was prepared by replacing the N-propyl group of the second-generation steroid-like inhibitor (2) with a N-3,3,3-trifluoropropyl group to give (10). This compound is 5-fold more potent in vitro, completely inhibits rat liver steroid sulfatase activity after a single oral dose of 0.5 mg/kg, and exhibits a significantly longer duration of inhibition over (2). These biological properties are attributed to the increased lipophilicity and metabolic stability of (10) rendered by its trifluoropropyl group and also the potential H-bonding between its fluorine atom(s) and Arg(98) in the active site of human steroid sulfatase. Like other sulfamates, (10) is expected to be sequestered, and transported by, erythrocytes in vivo because it inhibits human carbonic anhydrase II (hCAII) potently (IC(50), 3 nmol/L). A congener (4), which possesses a N-(pyridin-3-ylmethyl) substituent, is even more active (IC(50), 0.1 nmol/L). To rationalize this, the hCAII-(4) adduct, obtained by cocrystallization, reveals not only the sulfamate group and the backbone of (4) interacting with the catalytic site and the associated hydrophobic pocket, respectively, but also the potential H-bonding between the N-(pyridin-3-ylmethyl) group and Nepsilon(2) of Gln(136). Like (2), both (10) and its phenolic precursor (9) are non-estrogenic using a uterine weight gain assay. In summary, a highly potent, long-acting, and nonestrogenic steroid sulfatase inhibitor was designed with hCAII inhibitory properties that should positively influence in vivo behavior. Compound (10) and other related inhibitors of this structural class further expand the armory of steroid sulfatase inhibitors against hormone-dependent breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Flúor/química , Esteril-Sulfatasa/antagonistas & inhibidores , Animales , Cromatografía Liquida , Cristalografía por Rayos X , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Proteica , Ratas , Ratas Wistar , Espectrometría de Masa por Ionización de Electrospray , Esteril-Sulfatasa/química
13.
Mol Cell Endocrinol ; 283(1-2): 76-82, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18180093

RESUMEN

Steroid sulphatase (STS) catalyses the formation of active steroids from inactive steroid sulphates. High levels of intra-tumoural STS mRNA are associated with a poor prognosis in post-menopausal patients with oestrogen receptor positive breast cancer. In this study, analysis of the mutated STS protein showed that N- and C-terminal truncated STS constructs are inactive. Histidine 136, located inside the active site, is crucial for STS activity whereas proline 212, which allows the protein turn into the membrane, is not. Mutations in glycosylation sites asparagine 47 and 259 decreased STS activity while asparagine 333 and 459 mutations did not affect it. However, immunoblot studies revealed that all four N-linked sites are glycosylated to some extent. In addition, a polyclonal antibody raised in rabbits against human STS was developed and characterised. These data increase our knowledge of the STS enzyme structure and may help design new STS inhibitors.


Asunto(s)
Mutagénesis Sitio-Dirigida , Mutación Puntual/genética , Esteril-Sulfatasa/genética , Esteril-Sulfatasa/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Glicosilación , Humanos , Sueros Inmunes , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Esteril-Sulfatasa/química , Esteril-Sulfatasa/inmunología
14.
Cell Mol Life Sci ; 64(15): 2013-22, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17558559

RESUMEN

The sulfatase family of enzymes catalyzes hydrolysis of sulfate ester bonds of a wide variety of substrates. Seventeen genes have been identified in this class of sulfatases, many of which are associated with genetic disorders leading to reduction or loss of function of the corresponding enzymes. Amino acid sequence homology suggests that the enzymes have similar overall folds, mechanisms of action, and bivalent metal ion-binding sites. A catalytic cysteine residue, strictly conserved in prokaryotic and eukaryotic sulfatases, is post-translationally modified into a formylglycine. Hydroxylation of the formylglycine residue by a water molecule forming the activated hydroxylformylglycine (a formylglycine hydrate or a gem-diol) is a necessary step for the enzyme's sulfatase activity. Crystal structures of three human sulfatases, arylsulfatases A and B(ARSA and ARSB), and estrone/dehydroepiandrosterone sulfatase or steroid sulfatase (STS), also known as arylsulfatase C, have been determined. While ARSA and ARSB are water-soluble enzymes, STS has a hydrophobic domain and is an integral membrane protein of the endoplasmic reticulum. In this article, we compare and contrast sulfatase structures and revisit the proposed catalytic mechanism in light of available structural and functional data. Examination of the STS active site reveals substrate-specific interactions previously identified as the estrogen-recognition motif. Because of the proximity of the catalytic cleft of STS to the membrane surface, the lipid bilayer has a critical role in the constitution of the active site, unlike other sulfatases.


Asunto(s)
Sulfatasas/química , Sulfatasas/metabolismo , Aminoácidos/química , Neoplasias de la Mama/enzimología , Catálisis , Dominio Catalítico , Cerebrósido Sulfatasa/química , Cerebrósido Sulfatasa/metabolismo , Cristalografía por Rayos X , Femenino , Humanos , Técnicas In Vitro , Masculino , Modelos Moleculares , N-Acetilgalactosamina-4-Sulfatasa/química , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Embarazo , Neoplasias de la Próstata/enzimología , Estructura Terciaria de Proteína , Electricidad Estática , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo
15.
J Med Chem ; 50(15): 3540-60, 2007 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-17580845

RESUMEN

By introducting the steroid sulfatase inhibitory pharmacophore into aromatase inhibitor 1 (YM511), two series of single agent dual aromatase-sulfatase inhibitors (DASIs) were generated. The best DASIs in vitro (JEG-3 cells) are 5, (IC50(aromatase) = 0.82 nM; IC50(sulfatase) = 39 nM), and 14, (IC50(aromatase) = 0.77 nM; IC50(sulfatase) = 590 nM). X-ray crystallography of 5, and docking studies of selected compounds into an aromatase homology model and the steroid sulfatase crystal structure are presented. Both 5 and 14 inhibit aromatase and sulfatase in PMSG pretreated adult female Wistar rats potently 3 h after a single oral 10 mg/kg dose. Almost complete dual inhibition is observed for 5 but the levels were reduced to 85% (aromatase) and 72% (sulfatase) after 24 h. DASI 5 did not inhibit aldosterone synthesis. The development of a potent and selective DASI should allow the therapeutic potential of dual aromatase-sulfatase inhibition in hormone-dependent breast cancer to be assessed.


Asunto(s)
Inhibidores de la Aromatasa/síntesis química , Modelos Moleculares , Esteril-Sulfatasa/antagonistas & inhibidores , Sulfonamidas/síntesis química , Triazoles/síntesis química , Animales , Inhibidores de la Aromatasa/química , Inhibidores de la Aromatasa/farmacología , Línea Celular Tumoral , Cristalografía por Rayos X , Femenino , Humanos , Conformación Proteica , Ratas , Ratas Wistar , Esteril-Sulfatasa/química , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología , Triazoles/química , Triazoles/farmacología
16.
J Steroid Biochem Mol Biol ; 99(1): 67-75, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16621525

RESUMEN

A form of steroid sulphate sulphohydrolase (EC 3.1.6.2) hydrolysing the dehydroepiandrosterone sulphate (DHEAS-ase) was purified from human placenta microsomes. During the purification procedure the DHEAS-ase was separated from the oestrone sulphate sulphohydrolase (OS-ase). The purified DHEAS-ase revealed specific activity of 1520 nmolxmin-1xmgprotein-1 and exhibited optimal activity at pH 8.4. The Km value was established to be 3.3+/-0.07x10(-5) M. The pI value was around 8.7. The molecular weight estimated by gel filtration was 7.4 kDa. The purified DHEAS-ase was not sensitive to the common sulphohydrolase inhibitors, such as phosphate, sulphate and sulphide ions, but was inhibited by several phosphohydrolase inhibitors (ammonium molybdate, vanadium oxide(V), zinc acetate). Steroids effected inhibition or activation of the purified enzyme. The data concerning substances reacting with -SH groups suggest that in the physiological conditions DHEAS-ase is controlled by the redox status of the cell.


Asunto(s)
Microsomas/enzimología , Placenta/enzimología , Proteínas Gestacionales/química , Proteínas Gestacionales/aislamiento & purificación , Esteril-Sulfatasa/química , Esteril-Sulfatasa/aislamiento & purificación , Inhibidores Enzimáticos/química , Femenino , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Embarazo , Proteínas Gestacionales/fisiología , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/fisiología
17.
Biochemistry ; 44(18): 6858-66, 2005 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15865431

RESUMEN

Carbonic anhydrase (CA) catalyzes the reversible hydration of carbon dioxide to hydrogen carbonate. The role of CA in maintaining pH balance has made it an attractive drug target for the treatment of cancer, and it has recently been implicated in the delivery of sulfamate-containing drugs. With the acceptance of steroid sulfatase as a target for hormone-dependent cancer, novel dual aromatase-steroid sulfatase inhibitors (DASIs) containing a sulfamate group are now being developed. In this study, we show that CA II is potently inhibited by several members of this class of inhibitor. The structures of CA II complexed with 4-[(4-O-sulfamoylbenzyl)(4-cyanophenyl)amino]-4H-[1,2,4]triazole (K(D) = 84 +/- 5 nM) and 4-[(3-bromo-4-O-sulfamoylbenzyl)(4-cyanophenyl)amino]-4H-[1,2,4]triazole (K(D) = 454 +/- 29 nM) are reported to 2.02 and 1.76 A, respectively. Both inhibitors ligate to the active site zinc(II) atom via their sulfamate nitrogen, while the rest of the molecule is contained within the hydrophobic binding pocket. Key protein residues include Val-121, Phe-131, Val-135, Val-143, Leu-141, Leu-198, Pro-202, and Leu-204. Despite being structurally similar, the two ligands experience different types of binding particularly in the sulfamate-containing aromatic ring and the opposite geometric arrangement of the triazole and cyanophenyl groups around the configurationally invertible central nitrogen atom. Small changes in inhibitor structure can cause large changes in binding to CA II, and this underlines the importance of structure-based drug design with this enzyme and other isoforms relevant to potential anticancer therapy. Moreover, these results underpin the idea that binding to erythrocyte CA II may be a general method of stabilizing and delivering sulfamate-based drugs in vivo.


Asunto(s)
Inhibidores de la Aromatasa/química , Anhidrasa Carbónica II/química , Estrona/análogos & derivados , Estrona/química , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/metabolismo , Inhibidores de la Aromatasa/metabolismo , Sitios de Unión , Anhidrasa Carbónica II/antagonistas & inhibidores , Anhidrasa Carbónica II/metabolismo , Cristalización , Cristalografía por Rayos X , Dimetilsulfóxido/química , Estabilidad de Medicamentos , Estabilidad de Enzimas , Estrona/metabolismo , Humanos , Ligandos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solventes , Esteril-Sulfatasa/química
18.
Endocr Rev ; 26(2): 171-202, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15561802

RESUMEN

Steroid sulfatase (STS) is responsible for the hydrolysis of aryl and alkyl steroid sulfates and therefore has a pivotal role in regulating the formation of biologically active steroids. The enzyme is widely distributed throughout the body, and its action is implicated in physiological processes and pathological conditions. The crystal structure of the enzyme has been resolved, but relatively little is known about what regulates its expression or activity. Research into the control and inhibition of this enzyme has been stimulated by its important role in supporting the growth of hormone-dependent tumors of the breast and prostate. STS is responsible for the hydrolysis of estrone sulfate and dehydroepiandrosterone sulfate to estrone and dehydroepiandrosterone, respectively, both of which can be converted to steroids with estrogenic properties (i.e., estradiol and androstenediol) that can stimulate tumor growth. STS expression is increased in breast tumors and has prognostic significance. The role of STS in supporting tumor growth prompted the development of potent STS inhibitors. Several steroidal and nonsteroidal STS inhibitors are now available, with the irreversible type of inhibitor having a phenol sulfamate ester as its active pharmacophore. One such inhibitor, 667 COUMATE, has now entered a phase I trial in postmenopausal women with breast cancer. The skin is also an important site of STS activity, and deficiency of this enzyme is associated with X-linked ichthyosis. STS may also be involved in regulating part of the immune response and some aspects of cognitive function. The development of potent STS inhibitors will allow investigation of the role of this enzyme in physiological and pathological processes.


Asunto(s)
Estrona/análogos & derivados , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Ensayos Clínicos Fase I como Asunto , Cumarinas/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Estrona/farmacología , Estrona/uso terapéutico , Femenino , Humanos , Modelos Moleculares , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/enzimología , Esteril-Sulfatasa/química , Esteril-Sulfatasa/genética , Sulfonamidas/farmacología , Ácidos Sulfónicos
19.
Methods Enzymol ; 400: 273-93, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16399355

RESUMEN

The sulfatase family of enzymes catalyzes the hydrolysis of sulfate ester bonds of a wide variety of substrates. Nine human sulfatase proteins and their genes have been identified, many of which are associated with genetic disorders leading to reduction or loss of function of the corresponding enzyme. A catalytic cysteine residue, strictly conserved in prokaryotic and eukaryotic sulfatases, is modified posttranslationally into a formylglycine. Hydroxylation of the formylglycine residue by a water molecule forming the activated hydroxylformylglycine (a formylglycine hydrate or a gem-diol) is a necessary step for sulfatase activity of the enzyme. Crystal structures of three human sulfatases, arylsulfatases A and B (ARSA and ARSB) and C, also known as steroid sulfatase or estrone/dehydroepiandrosterone sulfatase (ES), have been determined. In addition, the crystal structure of a homologous bacterial arylsulfatase from Pseudomonas aeruginosa (PAS) is also available. While ARSA, ARSB, and PAS are water-soluble enzymes, ES has a hydrophobic domain and is presumed to be bound to the endoplasmic reticulum membrane. This chapter compares and contrasts four sulfatase structures and revisits the proposed catalytic mechanism in light of available structural and functional data. Examination of the ES active site reveals substrate-specific interactions previously identified in another steroidogenic enzyme. Possible influence of the lipid bilayer in substrate capture and recognition by ES is described. Finally, mapping the genetic mutations into the ES structure provides an explanation for the loss of enzyme function in X-linked ichthyosis.


Asunto(s)
Sulfatasas/química , Sulfatasas/clasificación , Animales , Arilsulfatasas/química , Arilsulfatasas/genética , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Humanos , Ictiosis Ligada al Cromosoma X/enzimología , Ictiosis Ligada al Cromosoma X/genética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia , Esteril-Sulfatasa/química , Esteril-Sulfatasa/genética , Especificidad por Sustrato , Sulfatasas/genética
20.
Biochim Biophys Acta ; 1739(1): 1-4, 2004 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-15607112

RESUMEN

X-linked ichthyosis is an inherited genetic disorder of the skin that results from steroid sulfatase (STS) deficiency. Seven critical point mutations have been previously reported for the STS gene, six leading to amino acid substitutions and one to a premature termination of the polypeptide chain. The three-dimensional structure of the full-length human enzyme has been recently determined. Amino acid substitutions due to point mutations in X-linked ichthyosis are mapped onto the three-dimensional structure of human STS. In each case, the substitution appears to cause disruption of the active site architecture or to interfere with the enzyme's putative membrane-associating motifs crucial to the integrity of the catalytic cleft, thereby providing an explanation for the loss of STS activity.


Asunto(s)
Ictiosis Ligada al Cromosoma X/genética , Mutación Puntual , Esteril-Sulfatasa/genética , Esteril-Sulfatasa/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Humanos , Modelos Moleculares , Conformación Proteica , Esteril-Sulfatasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA