Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 66(3): e0194321, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35041501

RESUMO

Alphaviruses cause animal or human diseases that are characterized by febrile illness, debilitating arthralgia, or encephalitis. Selective estrogen receptor modulators (SERMs), a class of FDA-approved drugs, have been shown to possess antiviral activities against multiple viruses, including hepatitis C virus, Ebola virus, dengue virus, and vesicular stomatitis virus. Here, we evaluated three SERM compounds, namely, 4-hydroxytamoxifen, tamoxifen, and clomifene, for plausible antiviral properties against two medically important alphaviruses, chikungunya virus (CHIKV) and Sindbis virus (SINV). In cell culture settings, these SERMs displayed potent activity against CHIKV and SINV at nontoxic concentrations with 50% effective concentration (EC50) values ranging between 400 nM and 3.9 µM. Further studies indicated that these compounds inhibit a postentry step of the alphavirus life cycle, while enzymatic assays involving purified recombinant proteins confirmed that these SERMs target the enzymatic activity of nonstructural protein 1 (nsP1), the capping enzyme of alphaviruses. Finally, tamoxifen treatment restrained CHIKV growth in the infected mice and diminished musculoskeletal pathologies. Combining biochemical analyses, cell culture-based studies, and in vivo analyses, we strongly argue that SERM compounds, or their derivatives, may provide for attractive therapeutic options against alphaviruses.


Assuntos
Infecções por Alphavirus , Vírus Chikungunya , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Linhagem Celular , Camundongos , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Proteínas não Estruturais Virais , Replicação Viral
2.
Mol Med ; 28(1): 44, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35468719

RESUMO

BACKGROUND: Menopausal hormone therapy (MHT) is recommended for only five years to treat vasomotor symptoms and vulvovaginal atrophy because of safety concerns with long-term treatment. We investigated the ability of 2',3',4'-trihydroxychalcone (2',3',4'-THC) to modulate estrogen receptor (ER)-mediated responses in order to find drug candidates that could potentially prevent the adverse effects of long-term MHT treatment. METHODS: Transfection assays, real time-polymerase chain reaction, and microarrays were used to evaluate the effects of 2',3',4'-THC on gene regulation. Radioligand binding studies were used to determine if 2',3',4'-THC binds to ERα. Cell proliferation was examined in MCF-7 breast cancer cells by using growth curves and flow cytometry. Western blots were used to determine if 2',3',4'-THC alters the E2 activation of the MAPK pathway and degradation of ERα. Chromatin immunoprecipitation was used to measure ERα binding to genes. RESULTS: The 2',3',4'-THC/E2 combination produced a synergistic activation with ERα on reporter and endogenous genes in human U2OS osteosarcoma cells. Microarrays identified 824 genes that we termed reprogrammed genes because they were not regulated in U2OS-ERα cells unless they were treated with 2',3',4'-THC and E2 at the same time. 2',3',4'-THC blocked the proliferation of MCF-7 cells by preventing the E2-induced activation of MAPK and c-MYC transcription. The antiproliferative mechanism of 2',3',4'-THC differs from selective estrogen receptor modulators (SERMs) because 2',3',4'-THC did not bind to the E2 binding site in ERα like SERMs. CONCLUSION: Our study suggests that 2',3',4'-THC may represent a new class of ERα modulators that do not act as a direct agonists or antagonists. We consider 2',3',4'-THC to be a reprogramming compound, since it alters the activity of ERα on gene regulation and cell proliferation without competing with E2 for binding to ERα. The addition of a reprogramming drug to estrogens in MHT may offer a new strategy to overcome the adverse proliferative effects of estrogen in MHT by reprogramming ERα as opposed to an antagonist mechanism that involves blocking the binding of estrogen to ERα.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Feminino , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Estradiol/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia
3.
Cancer Sci ; 111(8): 2718-2725, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32536008

RESUMO

Ubiquitin-dependent protein degradation has been implicated in the control of various cellular processes such as cell cycle control, transcriptional regulation, DNA damage repair, and apoptosis, many of which are involved in the initiation, progression, metastasis, and drug resistance of cancers. E3 ubiquitin ligases are known to be the second most prevalent cancer-related functional gene family next to protein kinases. Of these, FBXO22, an F-box receptor subunit of SCF E3 ligase, has recently been proposed to play a critical role in multiple aspects related to cancer development and therapy response. Firstly, FBXO22 is a key regulator of senescence induction through ubiquitylation of p53 for degradation. FBXO22 also acts as a molecular switch for the antagonistic and agonistic actions of selective estrogen receptor modulators (SERM) and determines the sensitivity of breast cancer to SERM by ubiquitylating KDM4B complexed with unliganded or SERMs-bound estrogen receptor (ER). Furthermore, FBXO22 binds to Bach1, a pro-metastatic transcription factor, suppressing Bach1-driven metastasis of lung adenocarcinoma, and loss of FBXO22 facilitates metastasis. These findings, as well as other reports, unveiled strikingly important roles of FBXO22 in cancer development and therapeutic strategy. In this review, we summarize recent findings of how FBXO22 regulates major cancer suppression pathways.


Assuntos
Epigênese Genética , Proteínas F-Box/metabolismo , Neoplasias/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Movimento Celular/genética , Senescência Celular/genética , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Neoplasias/patologia , Subunidades Proteicas/metabolismo , Proteólise , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Transdução de Sinais/genética , Ubiquitinação
4.
Front Neuroendocrinol ; 52: 44-64, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30223003

RESUMO

Synthetic selective modulators of the estrogen receptors (SERMs) have shown to protect neurons and glial cells against toxic insults. Among the most relevant beneficial effects attributed to these compounds are the regulation of inflammation, attenuation of astrogliosis and microglial activation, prevention of excitotoxicity and as a consequence the reduction of neuronal cell death. Under pathological conditions, the mechanism of action of the SERMs involves the activation of estrogen receptors (ERs) and G protein-coupled receptor for estrogens (GRP30). These receptors trigger neuroprotective responses such as increasing the expression of antioxidants and the activation of kinase-mediated survival signaling pathways. Despite the advances in the knowledge of the pathways activated by the SERMs, their mechanism of action is still not entirely clear, and there are several controversies. In this review, we focused on the molecular pathways activated by SERMs in brain cells, mainly astrocytes, as a response to treatment with raloxifene and tamoxifen.


Assuntos
Astrócitos/efeitos dos fármacos , Encefalopatias/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Cloridrato de Raloxifeno/farmacologia , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Animais , Humanos
5.
J Pharmacol Exp Ther ; 369(3): 389-405, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30918069

RESUMO

Lithocholic acid (LCA) is a bile acid associated with adverse effects, including cholestasis, and it exists in vivo mainly as conjugates known as glyco-LCA (GLCA) and tauro-LCA (TLCA). Tamoxifen has been linked to the development of cholestasis, and it inhibits sulfotransferase 2A1 (SULT2A1)-catalyzed dehydroepiandrosterone (DHEA) sulfonation. The present study was done to characterize the sulfonation of LCA, GLCA, and TLCA and to investigate whether triphenylethylene (clomifene, tamoxifen, toremifene, ospemifene, droloxifene), benzothiophene (raloxifene, arzoxifene), tetrahydronaphthalene (lasofoxifene, nafoxidine), indole (bazedoxifene), and benzopyran (acolbifene) classes of selective estrogen receptor modulator (SERM) inhibit LCA, GLCA, and TLCA sulfonation. Human recombinant SULT2A1, but not SULT2B1b or SULT1E1, catalyzed LCA, GLCA, and TLCA sulfonation, whereas each of these enzymes catalyzed DHEA sulfonation. LCA, GLCA, and TLCA sulfonation is catalyzed by human liver cytosol, and SULT2A1 followed the substrate inhibition model with comparable apparent K m values (≤1 µM). Each of the SERMs inhibited LCA, GLCA, and TLCA sulfonation with varying potency and mode of enzyme inhibition. The potency and extent of inhibition of LCA sulfonation were attenuated or increased by structural modifications to toremifene, bazedoxifene, and lasofoxifene. The inhibitory effect of raloxifene, bazedoxifene, and acolbifene on LCA sulfonation was also observed in HepG2 human hepatocellular carcinoma cells. Overall, among the SERMs investigated, bazedoxifene and raloxifene were the most effective inhibitors of LCA, GLCA, and TLCA sulfonation. These findings provide insight into the structural features of specific SERMs that contribute to their inhibition of SULT2A1-catalyzed LCA sulfonation. Inhibition of LCA, GLCA, and TLCA detoxification by a SERM may provide a biochemical basis for adverse effects associated with a SERM.


Assuntos
Biocatálise/efeitos dos fármacos , Ácido Litocólico/análogos & derivados , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Ácidos Sulfônicos/metabolismo , Sulfotransferases/metabolismo , Ácido Taurolitocólico/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Células Hep G2 , Humanos , Cinética , Ácido Litocólico/metabolismo , Fígado/citologia , Oxirredução , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Sulfotransferases/antagonistas & inibidores
6.
Toxicol Appl Pharmacol ; 353: 31-42, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29906493

RESUMO

Selective estrogen receptor modulators (SERMs) target estrogen receptors (ERs) to treat breast cancer and osteoporosis. Several SERMs exhibit anti-cancer activity not related to ERs. To discover novel anti-cancer drugs acting via ER-independent mechanisms, derivatives of the SERM tamoxifen, known as the "ridaifen" compounds, have been developed that exhibit reduced or no ER affinity, while maintaining cytotoxicity. Tamoxifen and other SERMs bind to cannabinoid receptors with moderate affinity. Therefore, ER-independent effects of SERMs might be mediated via cannabinoid receptors. This study determined whether RID-B, a first generation ridaifen compound, exhibits affinity and/or activity at CB1 and/or CB2 cannabinoid receptors. RID-B binds with high affinity (Ki = 43.7 nM) and 17-fold selectivity to CB2 over CB1 receptors. RID-B acts as an inverse agonist at CB2 receptors, modulating G-protein and adenylyl cyclase activity with potency values predicted by CB2 affinity. Characteristic of an antagonist, RID-B co-incubation produces a parallel-rightward shift in the concentration-effect curve of CB2 agonist WIN-55,212-2 to inhibit adenylyl cyclase activity. CB2 inverse agonists are reported to exhibit anti-inflammatory and anti-ostoeclastogenic effects. In LPS-activated macrophages, RID-B exhibits anti-inflammatory effects by reducing levels of nitric oxide (NO), IL-6 and IL-1α, but not TNFα. Only reduction of NO concentration by RID-B is mediated by cannabinoid receptors. RID-B also exhibits pronounced anti-osteoclastogenic effects, reducing the number of osteoclasts differentiating from primary bone marrow macrophages in a cannabinoid receptor-dependent manner. In summary, the tamoxifen derivative RID-B, developed with reduced affinity for ERs, is a high affinity selective CB2 inverse agonist with anti-inflammatory and anti-osteoclastogenic properties.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Osteoclastos/efeitos dos fármacos , Pirrolidinas/farmacologia , Receptor CB2 de Canabinoide/agonistas , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/análogos & derivados , Inibidores de Adenilil Ciclases/farmacologia , Animais , Benzoxazinas/farmacologia , Ligação Competitiva/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Células CHO , Diferenciação Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Agonismo Inverso de Drogas , Camundongos , Camundongos Endogâmicos C57BL , Morfolinas/farmacologia , Naftalenos/farmacologia , Pirrolidinas/metabolismo , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia
7.
J Neurochem ; 141(1): 31-36, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28075498

RESUMO

As one of the primary mechanisms by which dopamine signaling is regulated, the dopamine transporter (DAT) is an attractive pharmacological target for the treatment of diseases based in dopaminergic dysfunction. In this work we demonstrate for the first time that the commonly prescribed breast cancer therapeutic tamoxifen and its major metabolites, 4-hydroxytamoxifen and endoxifen, inhibit DAT function. Tamoxifen inhibits [3 H]dopamine uptake into human DAT (hDAT)-N2A cells via an uncompetitive or mixed mechanism. Endoxifen, an active metabolite of tamoxifen, asymmetrically inhibits DAT function in hDAT-N2A cells, showing a preference for the inhibition of amphetamine-stimulated dopamine efflux as compared to dopamine uptake. Importantly, we demonstrate that the effects of tamoxifen and its metabolites on the DAT occur independently of its activity as selective estrogen receptor modulators. This work suggests that tamoxifen is inhibiting DAT function through a previously unidentified mechanism.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina/antagonistas & inibidores , Proteínas da Membrana Plasmática de Transporte de Dopamina/fisiologia , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/fisiologia , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Antagonistas de Estrogênios/metabolismo , Antagonistas de Estrogênios/farmacologia , Humanos , Camundongos , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia
8.
Bioorg Med Chem Lett ; 27(11): 2590-2593, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28400239

RESUMO

Steroids are important components of cell membranes and are involved in several physiological functions. A diphenylmethane (DPM) skeleton has recently been suggested to act as a mimetic of the steroid skeleton. However, difficulties are associated with efficiently introducing different substituents between two phenyl rings of the DPM skeleton, and, thus, further structural development based on the DPM skeleton has been limited. We herein developed an efficient synthetic method for introducing different substituents into two phenyl rings of the DPM skeleton. We also synthesized DPM-based estrogen receptor (ER) modulators using our synthetic method and evaluated their ER transcriptional activities.


Assuntos
Compostos Benzidrílicos/química , Receptores de Estrogênio/metabolismo , Esteroides/química , Compostos Benzidrílicos/síntese química , Compostos Benzidrílicos/metabolismo , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Células HEK293 , Humanos , Concentração Inibidora 50 , Receptores de Estrogênio/antagonistas & inibidores , Moduladores Seletivos de Receptor Estrogênico/síntese química , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Esteroides/síntese química , Esteroides/metabolismo , Relação Estrutura-Atividade
9.
Environ Toxicol ; 31(12): 1834-1850, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26303313

RESUMO

Tamoxifen is an endocrine-active pharmaceutical (EAP) that is used world-wide. Because tamoxifen is a ubiquitous pharmaceutical and interacts with estrogen receptors, a case study was conducted with this compound to (1) determine effects on reproductive endpoints in a nontarget species (i.e., a fish), (2) compare biologically-active metabolites across species, (3) assess whether in vitro assays predict in vivo results, and (4) investigate metabolomic profiles in tamoxifen-treated fish to better understand the biological mechanisms of tamoxifen toxicity. In reproductive assays, tamoxifen exposure caused a significant reduction in egg production and significantly increased ovarian aromatase activity in spawning adult cunner fish (Tautogolabrus adspersus). In plasma from tamoxifen-exposed cunner, the predominant metabolite was 4-hydroxytamoxifen, while in rats it was N-desmethyltamoxifen. Because 4-hydroxytamoxifen is a more biologically active metabolite than N-desmethyltamoxifen, this difference could result in a different level of risk for the two species. The results of in vitro assays with fish hepatic microsomes to assess tamoxifen metabolism did not match in vivo results, indicating probable differences in excretion of tamoxifen metabolites in fish compared with rats. For the first time, a complete in vitro characterization of the metabolism of tamoxifen using fish microsomes is presented. Furthermore, a metabolomic investigation of cunner gonad extracts demonstrates that tamoxifen alters the biochemical profile in this nontarget species. Understanding the consequence of tamoxifen exposure in nontarget species, and assessing the discrepancies between sex- and species-mediated endpoints, is a step toward understanding how to accurately assess the risks posed by EAPs, such as tamoxifen, in the aquatic environment. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1834-1850, 2016.


Assuntos
Perciformes/fisiologia , Moduladores Seletivos de Receptor Estrogênico/toxicidade , Tamoxifeno/toxicidade , Poluentes Químicos da Água/toxicidade , Animais , Bioensaio , Ovos , Exposição Ambiental , Feminino , Microssomos Hepáticos/metabolismo , Reprodução/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo
10.
Clin Calcium ; 26(11): 1571-1581, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-27777390

RESUMO

Selective estrogen receptor modulators(SERMs), or estrogen receptor agonists/antagonists(ERAAs), are a class of compounds that function as estrogen receptor(ER)agonists or antagonists tissue-dependently. Two types of SERMs are currently used in Japan to treat osteoporosis, raloxifene(RLX:Evista)and bazedoxifene(BZA:Viviant). This article describes the pharmacokinetics of these two drugs.


Assuntos
Moduladores Seletivos de Receptor Estrogênico/farmacocinética , Administração Oral , Envelhecimento , Feminino , Humanos , Nefropatias , Masculino , Osteoporose/tratamento farmacológico , Pós-Menopausa , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Caracteres Sexuais
11.
J Cell Physiol ; 230(5): 1104-14, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25292157

RESUMO

Estrogen receptor (ER) activation has complex effects on bone cells, and loss of circulating estradiol adversely affects skeletal status in women. Hormone replacement therapy effectively circumvents bone loss after menopause, but enhances disease risk in other tissues. Here we show that prostaglandin E2 (PGE2) augments the activity of an osteoblast-derived selective ER modulator, ObSERM. The stimulatory effect of PGE2 is replicated in part by either the PG receptor EP3 agonist 17-phenyl trinor PGE2 or by the PG receptor FP agonist PGF2α⋅ Whereas activation of the various PG receptors induces multiple downstream signals, the response to PGE2 was mimicked by activators of protein kinase C, and suppressed by inhibition of protein kinase C but not by inhibition of protein kinase A. Moreover, inhibition of nitric oxide synthesis and activation of the PTH and Wnt pathways increases ObSERM activity. Our studies therefore reveal that ObSERM activity is controlled in distinct ways and revise our understanding of ER activation within bone by agents or events associated with PG expression. They also predict ways to sustain or improve bone formation, fracture repair, and surgical healing without adding the risk of disease in other tissues where ER activation also has important biological functions.


Assuntos
Dinoprostona/farmacologia , Osteoblastos/metabolismo , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , Receptores de Prostaglandina/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
12.
Drug Metab Dispos ; 43(9): 1353-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26070840

RESUMO

Women at high risk of developing breast cancer are prescribed selective estrogen response modulators, including raloxifene, as chemoprevention. Patients often seek complementary and alternative treatment modalities, including herbal products, to supplement prescribed medications. Milk thistle preparations, including silibinin and silymarin, are top-selling herbal products that may be consumed by women taking raloxifene, which undergoes extensive first-pass glucuronidation in the intestine. Key constituents in milk thistle, flavonolignans, were previously shown to be potent inhibitors of intestinal UDP-glucuronosyl transferases (UGTs), with IC50s ≤ 10 µM. Taken together, milk thistle preparations may perpetrate unwanted interactions with raloxifene. The objective of this work was to evaluate the inhibitory effects of individual milk thistle constituents on the intestinal glucuronidation of raloxifene using human intestinal microsomes and human embryonic kidney cell lysates overexpressing UGT1A1, UGT1A8, and UGT1A10, isoforms highly expressed in the intestine that are critical to raloxifene clearance. The flavonolignans silybin A and silybin B were potent inhibitors of both raloxifene 4'- and 6-glucuronidation in all enzyme systems. The Kis (human intestinal microsomes, 27-66 µM; UGT1A1, 3.2-8.3 µM; UGT1A8, 19-73 µM; and UGT1A10, 65-120 µM) encompassed reported intestinal tissue concentrations (20-310 µM), prompting prediction of clinical interaction risk using a mechanistic static model. Silibinin and silymarin were predicted to increase raloxifene systemic exposure by 4- to 5-fold, indicating high interaction risk that merits further evaluation. This systematic investigation of the potential interaction between a widely used herbal product and chemopreventive agent underscores the importance of understanding natural product-drug interactions in the context of cancer prevention.


Assuntos
Glucuronídeos/metabolismo , Mucosa Intestinal/metabolismo , Cloridrato de Raloxifeno/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Silybum marianum/química , Neoplasias da Mama/prevenção & controle , Interações Medicamentosas , Feminino , Humanos , Cloridrato de Raloxifeno/uso terapêutico , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico
13.
Drug Metab Dispos ; 43(6): 812-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25834030

RESUMO

Inhibition of drug metabolizing enzymes is a major mechanism in drug-drug interactions (DDIs). A number of cases of DDIs via inhibition of UDP-glucuronosyltranseferases (UGTs) have been reported, although the changes in pharmacokinetics are relatively small in comparison with drugs that are metabolized by cytochrome P450s. Most of the past studies have investigated hepatic UGTs, although recent studies have revealed a significant contribution of UGTs in the small intestine to drug clearance. To evaluate potential DDIs caused by inhibition of intestinal UGTs, we assessed inhibitory effects of 578 compounds, including drugs, xenobiotics, and endobiotics, on human UGT1A8 and UGT1A10, which are major contributors to intestinal glucuronidation. We identified 29 inhibitors by monitoring raloxifene glucuronidation with recombinant UGTs. All of the inhibitors potently inhibited UGT1A1 activity, as well. We found that zafirlukast is a potent general inhibitor of UGT1As and a moderate inhibitor of UGT2Bs because it monitors 4-methylumbelliferone glucuronidation by recombinant UGTs. However, zafirlukast did not potently inhibit diclofenac glucuronidation, suggesting that the inhibitory effects might be substrate specific. Inhibitory effects of zafirlukast on some UGT substrates were further investigated in human liver and human small intestine microsomes in order to evaluate potential DDIs. The R values (the ratios of intrinsic clearance with and without an inhibitor) revealed that zafirlukast has potential to cause clinical DDIs in the small intestine. Although we could not identify specific UGT1A8 and UGT1A10 inhibitors, zafirlukast was identified as a general inhibitor for UGTs in vitro. The present study suggests that the inhibition of UGT in the small intestine would be an underlying mechanism for DDIs.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Intestino Delgado/efeitos dos fármacos , Antagonistas de Leucotrienos/farmacologia , Desintoxicação Metabólica Fase II , Microssomos/efeitos dos fármacos , Compostos de Tosil/farmacologia , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Inibidores Enzimáticos/efeitos adversos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Indóis , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Intestino Delgado/enzimologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Antagonistas de Leucotrienos/efeitos adversos , Antagonistas de Leucotrienos/metabolismo , Microssomos/enzimologia , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Fenilcarbamatos , Cloridrato de Raloxifeno/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Bibliotecas de Moléculas Pequenas , Especificidade por Substrato , Sulfonamidas , Compostos de Tosil/efeitos adversos , Compostos de Tosil/metabolismo
14.
J Bone Miner Metab ; 33(2): 161-72, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24633538

RESUMO

Bazedoxifene (BZA) is a novel selective estrogen receptor modulator in clinical development for the prevention and treatment of postmenopausal osteoporosis. This preclinical study evaluated the efficacy and safety of BZA in preventing ovariectomy (OVX)-induced bone loss in aged cynomolgus monkeys. Animals (18 per group) underwent OVX and were administered BZA (0.2, 0.5, 1, 5, or 25 mg/kg/day) or vehicle, or were sham-operated and administered vehicle, by daily oral gavage for 18 months. Biochemical markers of bone turnover were assessed at 6, 12, and 18 months, along with bone densitometry using dual energy X-ray absorptiometry and peripheral quantitative computed tomography. Animals were killed after 18 months. Uterine and pituitary weights were determined, and histomorphometric and biomechanical measurements were performed. OVX vehicle controls showed increases in bone turnover associated with cancellous and cortical bone osteopenia (in vivo), and slight decreases (not statistically significant) in biomechanical strength parameters at the lumbar spine and femoral neck. BZA partially preserved cortical and cancellous bone mass by preventing the OVX-induced increases in bone turnover. Although the response was often similar among BZA-treated groups, the strongest efficacy was generally seen at 25 mg/kg/day. Treatment with BZA did not adversely affect measures of bone strength and was well tolerated; there was no evidence of uterotrophic activity, mammary tissue was unaffected, and there were no adverse effects on plasma lipids. Treatment of ovariectomized animals with BZA partially prevented changes in bone remodeling that correlated with increases in bone mineral density, while maintaining bone strength and a favorable safety profile.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Indóis/farmacologia , Osteoporose Pós-Menopausa/tratamento farmacológico , Absorciometria de Fóton/métodos , Animais , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Humanos , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/metabolismo , Macaca fascicularis , Osteoporose Pós-Menopausa/metabolismo , Ovariectomia/métodos , Moduladores Seletivos de Receptor Estrogênico/metabolismo
15.
Climacteric ; 17 Suppl 2: 60-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25320023

RESUMO

Despite increased survivorship among patients, breast cancer remains the most common cancer among women and is the second leading cause of cancer death in women. The magnitude of this problem provides a strong impetus for new chemopreventative strategies and/or lifestyle changes that reduce cancer incidence. It is of significance, therefore, that several studies positively correlate obesity to the development of breast cancer. Importantly, obesity is also highly associated with elevated cholesterol, and cholesterol itself is a risk factor for breast cancer. Furthermore, patients taking statins demonstrate a lower breast cancer incidence and decreased recurrence. The recent observation that 27-hydroxycholesterol (27HC) is produced in a stoichiometric manner from cholesterol, together with our recent demonstration that it exerts partial agonist activity on both the estrogen and liver X receptors, suggested a potential mechanistic link between hyper-cholesterolemia and breast cancer incidence. Using genetic and pharmacological approaches, we have recently shown that elevation of circulating 27HC significantly increases tumor growth and metastasis in murine models of breast cancer. Further, we have demonstrated in appropriate animal models that the impact of high-fat diet on tumor pathogenesis can be mitigated by statins or by small molecule inhibitors of CYP27A1. These findings suggest that pharmacological or dietary modifications that lower total cholesterol, and by inference 27HC, are likely to reduce the impact of obesity/metabolic syndrome on breast cancer incidence.


Assuntos
Neoplasias da Mama/etiologia , Colesterol na Dieta/toxicidade , Receptores de Estrogênio/metabolismo , Animais , Comunicação Autócrina/imunologia , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/patologia , Colesterol/sangue , Colesterol na Dieta/sangue , Modelos Animais de Doenças , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Hidroxicolesteróis/sangue , Hidroxicolesteróis/síntese química , Hiperlipidemias/complicações , Hiperlipidemias/tratamento farmacológico , Incidência , Recidiva Local de Neoplasia , Obesidade/complicações , Comunicação Parácrina/imunologia , Fatores de Risco , Moduladores Seletivos de Receptor Estrogênico/metabolismo
16.
Int J Cancer ; 132(6): 1475-85, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22915089

RESUMO

We investigated the in vitro metabolism and estrogenic and antiestrogenic activity of toremifene (TOR), tamoxifen (TAM) and their metabolites to better understand the potential effects of cytochrome P-450 2D6 (CYP2D6) status on the activity of these drugs in women with breast cancer. The plasma concentrations of TOR and its N-desmethyl (NDM) and 4-hydroxy (4-OH) metabolites during steady-state dosing with TOR were also determined. Unlike TOR, TAM and its NDM metabolite were extensively oxidized to 4-OH TAM and 4-OH-NDM TAM by CYP2D6, and the rate of metabolism was affected by CYP2D6 status. 4-OH-NDM TOR concentrations were not measurable at steady state in plasma of subjects taking 80 mg of TOR. Molecular modeling provided insight into the lack of 4-hydroxylation of TOR by CYP2D6. The 4-OH and 4-OH-NDM metabolites of TOR and TAM bound to estrogen receptor (ER) subtypes with fourfold to 30-fold greater affinity were 35- to 187-fold more efficient at antagonizing ER transactivation and had antiestrogenic potency that was up to 360-fold greater than their parent drugs. Our findings suggest that variations in CYP2D6 metabolic capacity may cause significant differences in plasma concentrations of active TAM metabolites (i.e., 4-OH TAM and 4-OH-NDM TAM) and contribute to variable pharmacologic activity. Unlike TAM, the clinical benefits in subjects taking TOR to treat metastatic breast cancer would not likely be subject to allelic variation in CYP2D6 status or affected by coadministration of CYP2D6-inhibiting medications.


Assuntos
Citocromo P-450 CYP2D6/fisiologia , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Toremifeno/metabolismo , Adulto , Humanos , Masculino , Oxirredução , Relação Estrutura-Atividade , Tamoxifeno/análogos & derivados , Tamoxifeno/sangue
17.
J Mammary Gland Biol Neoplasia ; 17(2): 147-53, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22588661

RESUMO

Estrogen Receptor (ER) is a nuclear receptor that mediates the actions of estrogen and tamoxifen. ER is expressed in a major fraction of human breast cancers. Recently, genomic maps for estrogen- and tamoxifen-ER have been published. Interestingly, estrogen and tamoxifen induce similar genomic interactions and both ligands have been shown to use co-operating factors. The interactions of these co-operating factors within ER regions have impact both on ER-DNA interactions and gene expression regulated by estrogen and tamoxifen. Moreover, the study of chromatin changes induced by these factors has also provided significant insight into our understanding of ER transcriptional regulation. This methods review describes some functional genomic methods to study the influence of both ER ligands and ER co-operating factors. The analysis of protein-DNA interactions and chromatin changes can be explored by using classical and novel methods such as Chromatin Immunoprecipitation (ChIP) or Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE). This review also explores the properties of each of these methods and the advantages of combining them with high throughput sequencing.


Assuntos
Neoplasias da Mama/metabolismo , Genômica/métodos , Glândulas Mamárias Humanas/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Antineoplásicos Hormonais/metabolismo , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Reagentes de Ligações Cruzadas/química , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Glândulas Mamárias Humanas/efeitos dos fármacos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Receptores de Estrogênio/química , Receptores de Estrogênio/genética , Elementos Reguladores de Transcrição/efeitos dos fármacos , Elementos de Resposta/efeitos dos fármacos , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia
18.
Drug Metab Dispos ; 40(2): 389-96, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22096084

RESUMO

Tamoxifen, an antiestrogen used in the prevention and treatment of breast cancer, is extensively metabolized by cytochrome P450 enzymes. Its biotransformation to α-hydroxytamoxifen (α-OHT), which may be genotoxic, and to N-desmethyltamoxifen (N-DMT), which is partially hydroxylated to 4-hydroxy-N-DMT (endoxifen), a potent antiestrogen, is mediated by CYP3A enzymes. However, the potential contribution of CYP3A5 and the impact of its low-expression variants on the formation of these metabolites are not clear. Therefore, we assessed the contributions of CYP3A4 and CYP3A5 and examined the impact of CYP3A5 genotypes on the formation of α-OHT and N-DMT, by using recombinant CYP3A4 and CYP3A5 and human liver microsomes (HLM) genotyped for CYP3A5 variants. We observed that the catalytic efficiency [intrinsic clearance (CL(int))] for α-OHT formation with recombinant CYP3A4 was 5-fold higher than that with recombinant CYP3A5 (0.81 versus 0.16 nl · min⁻¹ · pmol cytochrome P450⁻¹). There was no significant difference in CL(int) values between the three CYP3A5-genotyped HLM (*1/*1, *1/*3, and *3/*3). For N-DMT formation, the CL(int) with recombinant CYP3A4 was only 1.7-fold higher, relative to that with recombinant CYP3A5. In addition, the CL(int) for N-DMT formation by HLM with CYP3A5*3/*3 alleles was approximately 3-fold lower than that for HLM expressing CYP3A5*1/*1. Regression analyses of tamoxifen metabolism with respect to testosterone 6ß-hydroxylation facilitated assessment of CYP3A5 contributions to the formation of the two metabolites. The CYP3A5 contributions to α-OHT formation were negligible, whereas the contributions to N-DMT formation ranged from 51 to 61%. Our findings suggest that polymorphic CYP3A5 expression may affect the formation of N-DMT but not that of α-OHT.


Assuntos
Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Microssomos Hepáticos/enzimologia , Polimorfismo Genético , Tamoxifeno/análogos & derivados , Alelos , Antineoplásicos Hormonais/metabolismo , Humanos , Hidroxilação , Cinética , Proteínas Recombinantes/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Especificidade por Substrato , Tamoxifeno/metabolismo
19.
Drug Metab Dispos ; 40(6): 1210-5, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22434874

RESUMO

Toremifene (TOR) is a selective estrogen receptor modulator used in adjuvant therapy for breast cancer and in clinical trials for prostate cancer prevention. The chemical structure of TOR differs from that of tamoxifen (TAM) by the presence of a chlorine atom in the ethyl side chain, resulting in a more favorable toxicity spectrum with TOR. In addition, some patients who fail on TAM therapy benefit from high-dose TOR therapy. Several studies have indicated that functional genetic variants in the TAM metabolic pathway influence response to therapy, but pharmacogenomic studies of patients treated with TOR are lacking. In this study, we examined individual variability in sulfation of 4-hydroxy TOR (4-OH TOR) (the active metabolite of TOR) in human liver cytosols from 104 subjects and found approximately 30-fold variation in activity. 4-OH TOR sulfation was significantly correlated (r = 0.98, P < 0.0001) with ß-naphthol sulfation (diagnostic for SULT1A1) but not with 17ß estradiol sulfation, a diagnostic substrate for SULT1E1(r = 0.09, P = 0.34). Examination of recombinant sulfotransferases (SULTs) revealed that SULT1A1 and SULT1E1 catalyzed 4-OH TOR sulfation, with apparent Km values of 2.6 and 6.4 µM and Vmax values of 8.5 and 5.5 nmol x min(-1) x mg protein(-1), respectively. 4-OH TOR sulfation was inhibited by 2,6-dichloro-4-nitrophenol (IC50 = 2.34 ± 0.19 µM), a specific inhibitor of SULT1A1. There was also a significant association between SULT1A1 genotypes and copy number and 4-OH TOR sulfation in human liver cytosols. These results indicate that variability in sulfation could contribute to response to TOR in the treatment of breast and prostate cancer.


Assuntos
Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Variação Genética/fisiologia , Farmacogenética , Tamoxifeno/análogos & derivados , Toremifeno/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Feminino , Humanos , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Farmacogenética/métodos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Adulto Jovem
20.
Chem Res Toxicol ; 25(7): 1472-83, 2012 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-22642258

RESUMO

The bioactivation of both endogenous and equine estrogens to electrophilic quinoid metabolites has been postulated as a contributing factor in carcinogenic initiation and/or promotion in hormone sensitive tissues. Bearing structural resemblance to estrogens, extensive studies have shown that many selective estrogen receptor modulators (SERMs) are subject to similar bioactivation pathways. Lasofoxifene (LAS), a third generation SERM which has completed phase III clinical trials for the prevention and treatment of osteoporosis, is currently approved in the European Union for this indication. Previously, Prakash et al. (Drug Metab. Dispos. (2008) 36, 1218-1226) reported that similar to estradiol, two catechol regioisomers of LAS are formed as primary oxidative metabolites, accounting for roughly half of the total LAS metabolism. However, the potential for further oxidation of these catechols to electrophilic o-quinones has not been reported. In the present study, LAS was synthesized and its oxidative metabolism investigated in vitro under various conditions. Incubation of LAS with tyrosinase, human liver microsomes, or rat liver microsomes in the presence of GSH as a trapping reagent resulted in the formation of two mono-GSH and two di-GSH catechol conjugates which were characterized by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Similar conjugates were also detected in incubations with P450 3A4, P450 2D6, and P450 1B1 supersomes. Interestingly, these conjugates were also detected as major metabolites when compared to competing detoxification pathways such as glucuronidation and methylation. The 7-hydroxylasofoxifene (7-OHLAS) catechol regioisomer was also synthesized and oxidized either chemically or enzymatically to an o-quinone that was shown to form depurinating adducts with DNA. Collectively, these data show that analogous to estrogens, LAS is oxidized to catechols and o-quinones which could potentially contribute to in vivo toxicity for this SERM.


Assuntos
Estradiol/metabolismo , Pirrolidinas/metabolismo , Quinonas/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tetra-Hidronaftalenos/metabolismo , Animais , Catecóis/química , Catecóis/metabolismo , Bovinos , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/metabolismo , DNA/química , Estradiol/química , Glutationa/metabolismo , Humanos , Microssomos Hepáticos/metabolismo , Monofenol Mono-Oxigenase/metabolismo , Oxirredução , Pirrolidinas/química , Quinonas/química , Ratos , Moduladores Seletivos de Receptor Estrogênico/química , Estereoisomerismo , Espectrometria de Massas em Tandem , Tetra-Hidronaftalenos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA