Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Pathol ; 244(2): 203-214, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29144553

RESUMO

The enzyme type 1 17ß-hydroxysteroid dehydrogenase (17ß-HSD-1), responsible for generating active 17ß-estradiol (E2) from low-active estrone (E1), is overexpressed in endometrial cancer (EC), thus implicating an increased intra-tissue generation of E2 in this estrogen-dependent condition. In this study, we explored the possibility of inhibiting 17ß-HSD-1 and impairing the generation of E2 from E1 in EC using in vitro, in vivo, and ex vivo models. We generated EC cell lines derived from the well-differentiated endometrial adenocarcinoma Ishikawa cell line and expressing levels of 17ß-HSD-1 similar to human tissues. In these cells, HPLC analysis showed that 17ß-HSD-1 activity could be blocked by a specific 17ß-HSD-1 inhibitor. In vitro, E1 administration elicited colony formation similar to E2, and this was impaired by 17ß-HSD-1 inhibition. In vivo, tumors grafted on the chicken chorioallantoic membrane (CAM) demonstrated that E1 upregulated the expression of the estrogen responsive cyclin A similar to E2, which was impaired by 17ß-HSD-1 inhibition. Neither in vitro nor in vivo effects of E1 were observed using 17ß-HSD-1-negative cells (negative control). Using a patient cohort of 52 primary ECs, we demonstrated the presence of 17ß-HSD-1 enzyme activity (ex vivo in tumor tissues, as measured by HPLC), which was inhibited by over 90% in more than 45% of ECs using the 17ß-HSD-1 inhibitor. Since drug treatment is generally indicated for metastatic/recurrent and not primary tumor, we next demonstrated the mRNA expression of the potential drug target, 17ß-HSD-1, in metastatic lesions using a second cohort of 37 EC patients. In conclusion, 17ß-HSD-1 inhibition efficiently blocks the generation of E2 from E1 using various EC models. Further preclinical investigations and 17ß-HSD-1 inhibitor development to make candidate compounds suitable for the first human studies are awaited. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Endométrio/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Embrião de Galinha , Ciclina A/metabolismo , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Estradiol/metabolismo , Estradiol/farmacologia , Estradiol Desidrogenases/genética , Estradiol Desidrogenases/metabolismo , Estrona/metabolismo , Estrona/farmacologia , Feminino , Humanos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos
2.
Steroids ; 172: 108856, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33945801

RESUMO

17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) and steroid sulfatase (STS) are involved in the synthesis of the most potent estrogen in the human body, estradiol (E2). These enzymes are known to play a pivotal role in the progression of estrogen-dependent diseases, such as breast cancer and endometriosis. Therefore, the inhibition of 17ß-HSD1 and/or STS represents a promising avenue to modulate the growth of estrogen-dependent tumors or lesions. We recently established the key role of a bromoethyl side chain added at the C3-position of a 16ß-carbamoyl-benzyl-E2 nucleus to covalently inhibit 17ß-HSD1. To extend the structure-activity relationship study to the C16ß-position of this new selective irreversible inhibitor (PBRM), we synthesized a series of analog compounds by changing the nature of the C16ß-side chain but keeping the 2-bromoethyl group at position C3. We determined their 17ß-HSD1 inhibitions in T-47D cells (transformation of E1 into E2), but we did not obtain a stronger 17ß-HSD1 inhibitor than PBRM. Compounds 16 and 17 were found to be more likely to bind to the catalytic site and showed a promising but moderate inhibitory activity with estimated IC50 values of 0.5 and 0.7 µM, respectively (about 10 times higher than PBRM). Interestingly, adding one or two sulfamate groups in the D-ring's surroundings did not significantly decrease compounds' potential to inhibit 17ß-HSD1, but clearly improved their potential to inhibit STS. These results open the door to the development of a new family of steroid derivatives with dual (17ß-HSD1 and STS) inhibiting actions.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Esteril-Sulfatase/antagonistas & inibidores , Trientina/química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Proliferação de Células , Feminino , Humanos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
3.
Mol Nutr Food Res ; 64(16): e2000289, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32640069

RESUMO

SCOPE: Urolithin A and B are gut metabolites of ellagic acid and ellagitannins associated with many beneficial effects. Evidence in vitro pointed to their potential as estrogenic modulators. However, both molecular mechanisms and biological targets involved in such activity are still poorly characterized, preventing a comprehensive understanding of their bioactivity in living organisms. This study aimed at rationally identifying novel biological targets underlying the estrogenic-modulatory activity of urolithins. METHODS AND RESULTS: The work relies on an in silico/in vitro target fishing study coupling molecular modeling with biochemical and cell-based assays. Estrogen sulfotransferase and 17ß-hydroxysteroid dehydrogenase are identified as potentially subject to inhibition by the investigated urolithins. The inhibition of the latter undergoes experimental confirmation either in a cell-free or cell-based assay, validating computational outcomes. CONCLUSIONS: The work describes target fishing as an effective tool to identify unexpected targets of food bioactives detailing the interaction at a molecular level. Specifically, it described, for the first time, 17ß-hydroxysteroid dehydrogenase as a target of urolithins and highlighted the need of further investigations to widen the understanding of urolithins as estrogen modulators in living organisms.


Assuntos
Cumarínicos/farmacologia , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Proteínas/metabolismo , 17-Hidroxiesteroide Desidrogenases/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Sistema Livre de Células , Simulação por Computador , Cumarínicos/química , Cumarínicos/metabolismo , Humanos , Ligantes , Células MCF-7 , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Sulfotransferases/química , Sulfotransferases/metabolismo
4.
Mol Nutr Food Res ; 64(10): e1900706, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32239630

RESUMO

SCOPE: Maca (Lepidium meyenii), a well-known plant from the Andean highlands of Peru, has been used widely as a nutritional supplement to increase sexual function and fecundity. However, the identity of its active ingredients and how they function remain unknown. METHODS AND RESULTS: Chemical substances in maca are identified by UPLC-Q-TOF, and the active ingredients are screened through HotMap coupled with an artificial neural network. Lepidiline A (LA), an imidazole alkaloid, is identified as the key active compound. LA affects the balance of endogenous sex hormones in mice and improves fecundity in Drosophila. Using a molecular LA probe, 17ß-hydroxysteroid dehydrogenase type 1 (HSD17B1) is revealed to be the potential target of LA using a fishing-rod strategy. It is demonstrated with experimental data that LA targets HSD17B1 to enhance the enzyme's activity and increases its bioconversion efficiency of actively formed sex hormones including estrogen to 17ß-estradiol and 4-androsten-3,7-dione to testosterone, which ultimately improves reproductive activity. CONCLUSION: LA improves the balance of endogenous sex hormones and increases fecundity by targeting HSD17B1. This underlying mechanism of action provides a useful insight into the application of maca in the regulation of dietary nutrition and healthy fertility.


Assuntos
Drosophila melanogaster/efeitos dos fármacos , Estradiol Desidrogenases/metabolismo , Fertilidade/efeitos dos fármacos , Hormônios Esteroides Gonadais/metabolismo , Lepidium/química , Alcaloides/análise , Alcaloides/química , Animais , Células CHO , Cricetulus , Drosophila melanogaster/fisiologia , Feminino , Masculino , Camundongos
5.
J Steroid Biochem Mol Biol ; 178: 167-176, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29248731

RESUMO

The development of a covalent inhibitor of 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) is a promising approach for the treatment of hormone-dependent breast cancer and endometriosis. After reporting the steroid derivative PBRM as a first potent covalent inhibitor of 17ß-HSD1 without estrogenic activity, we are now interested in studying its pharmaceutical behavior. The metabolism study in a human liver microsomal preparation showed a gradual transformation of PBRM into PBRM-O, an oxidized ketonic form of PBRM at position C17. Interestingly, PBRM-O also inhibits 17ß-HSD1 and is not estrogenic in estrogen-sensitive T-47D cells. However, when PBRM was injected subcutaneously (sc) in mice, a very small proportion of PBRM-O was measured in a 24 h-time course experiment. A pharmacokinetic study in mice revealed suitable values for half-life (T1/2 = 3.4 h), clearance (CL = 2088 mL/h kg), distribution volume (Vz = 10.3 L/kg) and absolute bioavailability (F = 65%) when PBRM was injected sc at 14.7 mg/kg. A good F value of 33% was also obtained when PBRM was given orally. A tritiated version of PBRM, 3H-PBRM, was synthesized and used for an in vivo biodistribution study that showed its gradual accumulation in various mouse tissues (peak at 6 h) followed by elimination until complete disappearance after 72 h. Elimination was found to occur in feces (93%) and urine (7%) as revealed by a mass balance experiment. PBRM was also evaluated for its toxicity in mice and it was found to be very well tolerated after weekly sc administration (30-405 mg/kg for 8 weeks) or by po administration (300-900 mg/kg for 4 weeks). Overall, these experiments represent important steps in the preclinical characterization of the pharmaceutical behavior of PBRM, as well as for its translation to clinical trials.


Assuntos
Benzamidas/química , Neoplasias da Mama/tratamento farmacológico , Endometriose/tratamento farmacológico , Inibidores Enzimáticos/farmacocinética , Estradiol Desidrogenases/antagonistas & inibidores , Animais , Apoptose , Benzamidas/farmacocinética , Benzamidas/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Proliferação de Células , Endometriose/enzimologia , Endometriose/patologia , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Eur J Med Chem ; 127: 944-957, 2017 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-27852458

RESUMO

Current endocrine therapeutics for the estrogen-dependent disease endometriosis often lead to considerable side-effects as they act by reducing estrogen action systemically. A more recent approach takes advantage of the fact that the weak estrogen estrone (E1) which is abundant in the plasma, is activated in the target cell to the highly estrogenic estradiol (E2) by 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1). 17ß-HSD1 is overexpressed in endometriosis and thus a promising target for the treatment of this disease, with the prospect of less target-associated side-effects. Potent inhibitors from the class of bicyclic substituted hydroxyphenylmethanones with sulfonamide moiety recently described by us suffered from high molecular weight and low selectivity over 17ßHSD2, the physiological adversary of 17ß-HSD1. We describe the structural optimizations leading to the discovery of (5-(3,5-dichloro-4-methoxyphenyl)thiophen-2-yl)(2,6-difluoro-3-hydroxyphenyl)methanone 20, which displayed a sub-nanomolar IC50 towards 17ß-HSD1 as well as high selectivity over the type 2 enzyme, the estrogen receptors α and ß and a range of hepatic CYP enzymes. The compound did neither show cellular toxicity, nor PXR activation nor mutagenicity in the AMES II assay. Additional favourable pharmacokinetic properties (rat) make 20 a suitable candidate for proof-of-principle studies using xenotransplanted immunodeficient rats.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Estrogênios/metabolismo , 17-Hidroxiesteroide Desidrogenases/metabolismo , Actinas/metabolismo , Animais , Técnicas de Química Sintética , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacocinética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Estudos de Viabilidade , Feminino , Humanos , Concentração Inibidora 50 , Ratos , Especificidade por Substrato
7.
Appl Biochem Biotechnol ; 182(2): 482-494, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27933483

RESUMO

Human 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1), a potential target in breast cancer prevention and therapy, was extracted from human placenta and immobilized on nonporous silica (∼5 µm) with a covalent method for the first time. The optimum initial enzyme concentration and immobilization time during the immobilization process were 0.42 mg mL-1 and 12 h, repectively. The binding was confirmed by scanning electron microscope (SEM) and infrared spectroscopy (FT-IR). It could improve the pH, thermal and storage stability compared to free enzyme. Moreover, the immobilized enzyme could be reused at least four times. A screening method based on it coupled with liquid chromatography-time-of-flight mass spectrometer (LC-TOF/MS) was established, and the half-maximal inhibitory concentration (IC 50) of apigenin for the immobilized enzyme was 291 nM. Subsequently, 10 natural products were evaluated leading to inhibition of the activity of 17ß-HSD1 at the concentration of 25 µM, and six of them inhibit the activity over 50%.


Assuntos
17-Hidroxiesteroide Desidrogenases/química , Antineoplásicos Fitogênicos/química , Apigenina/química , Enzimas Imobilizadas/química , Espectrometria de Massas , Proteínas da Gravidez/química , 17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Cromatografia Líquida , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Feminino , Glutaral/química , Humanos , Dióxido de Silício/química
8.
Cancer Lett ; 370(1): 145-52, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26497762

RESUMO

Enhancing the radiosensitivity of cancer cells is one of the most important tasks in clinical radiobiology. Endocrine therapy and radiotherapy are two cancer treatment modalities which are often given together in patients with locally-advanced breast cancer and positive hormone-receptor status. Oncostatic actions of melatonin are relevant on estrogen-dependent mammary tumors. In the present study, we wanted to evaluate the effects of the combination of ionizing radiation and melatonin on proteins involved in estrogen biosynthesis in breast cancer cells. We demonstrated a role of melatonin in mediating the sensitization of human breast cancer cells to the ionizing radiation by decreasing around 50% the activity and expression of proteins involved in the synthesis of estrogens in these cells. Thus, melatonin pretreatment before radiation reduces the amount of active estrogens at cancer cell level. Melatonin 1 nM induced a 2-fold change in p53 expression as compared to radiation alone. The regulatory action of melatonin on p53 could be a link between melatonin and its modulatory action on the sensitivity of breast cancer cells to ionizing radiation. These findings may have implications for designing clinical trials using melatonin and radiotherapy.


Assuntos
Neoplasias da Mama/radioterapia , Estrogênios/biossíntese , Melatonina/farmacologia , Tolerância a Radiação/efeitos dos fármacos , 17-Hidroxiesteroide Desidrogenases/metabolismo , Aromatase/genética , Aromatase/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células/efeitos da radiação , Feminino , Humanos , Células MCF-7 , Esteril-Sulfatase/metabolismo , Proteína Supressora de Tumor p53/genética
9.
Eur J Med Chem ; 103: 56-68, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26322835

RESUMO

17ß-Estradiol (E2), the most potent human estrogen, is known to be involved in the etiology of estrogen-dependent diseases (EDD) like breast cancer and endometriosis. 17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) catalyses the last step of E2 biosynthesis and is thus a promising target for the treatment of EDD. The previously described bicyclic substituted hydroxyphenylmethanones (BSHs) display high inhibitory potency towards human 17ß-HSD1, but marginal activity towards rodent 17ß-HSD1, precluding a proof of principle study in an animal endometriosis model. The aim of this work was to perform structural optimizations in the BSHs class to enhance inhibitory activity against rodent (mouse and rat) 17ß-HSD1 while maintaining activity against the human enzyme. The introduction of fluorine atoms on the benzoyl moiety resulted in compounds with the desired properties. Molecular docking and homology modeling were applied to elucidate the binding mode and interspecies differences in activity. Compound 33 is the most potent inhibitor of both human and rat 17ß-HSD1 up to date (IC50 = 2 nM and 97 nM, respectively).


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Fenóis/farmacologia , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Simulação de Dinâmica Molecular , Estrutura Molecular , Fenóis/síntese química , Fenóis/química , Ratos , Relação Estrutura-Atividade , Sulfonamidas/química
10.
Eur J Med Chem ; 82: 394-406, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-24929290

RESUMO

Estradiol is the most potent estrogen in humans. It is known to be involved in the development and proliferation of estrogen dependent diseases such as breast cancer and endometriosis. The last step of its biosynthesis is catalyzed by 17ß-hydroxysteroid dehydrogenase type 1 (17ß- HSD1) which consequently is a promising target for the treatment of these diseases. Recently, we reported on bicyclic substituted hydroxyphenylmethanones as potent inhibitors of 17ß-HSD1. The present study focuses on rational structural modifications in this compound class with the aim of gaining more insight into its structure-activity relationship (SAR). (4-Hydroxyphenyl)-(5-(3-hydroxyphenylsulfanyl)-thiophen-2-yl)methanone (25) was discovered as a member of a novel potent class of human 17ß-HSD1 inhibitors. Computational methods were used to elucidate its interactions with the target protein. The compound showed activity also towards the murine 17ß-HSD1 enzyme and thus is a starting point for the design of compounds suitable for evaluation in an animal disease model.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Fenóis/farmacologia , Sulfetos/química , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Camundongos , Modelos Moleculares , Estrutura Molecular , Fenóis/síntese química , Fenóis/química , Relação Estrutura-Atividade
11.
Eur J Med Chem ; 69: 201-15, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24036043

RESUMO

Under physiological conditions healthy bones are maintained by a well tightened balance between osteoclast (OCs) and osteoblast (OBs) activity. Disruption of this balance leads to osteoporosis characterized by decline in bone function and skeletal rigidity. Inhibition of 17ß-hydroxysteroid dehydrogenase type 2 (17ß-HSD2) could help maintaining the appropriate bone mass density by increasing the level of estradiol and testosterone in bone. Herein, we described the synthesis, the physicochemical properties and the biological evaluation of novel N-methylsulfonamide and retro-N-methylsulfonamide derivatives as 17ß-HSD2 inhibitors showing high potency (compound 10f, IC50 = 23 nM), with a good selectivity toward 17ß-HSD1 (the isoenzyme responsible of the reverse reaction), and a likely good in vitro ADME profile. It was also shown that the acidity of the phenolic hydroxy correlates with the inhibitory potency, suggesting pKa as a predictive parameter for the activity of this class of inhibitors.


Assuntos
Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Estradiol Desidrogenases/antagonistas & inibidores , Sulfonamidas/farmacologia , Físico-Química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Estradiol Desidrogenases/metabolismo , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA