Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
1.
Biomed Res ; 43(6): 211-221, 2022.
Article in English | MEDLINE | ID: mdl-36517023

ABSTRACT

Selective estrogen receptor modulator (SERM) binds to estrogen receptors (ERs) and acts as both an agonist or an antagonist, depending on the target tissue. Raloxifene and bazedoxifene as SERMs are currently used hormone replacement medicines for postmenopausal osteoporosis. Macrophage colony-stimulating factor (M-CSF) secreted from osteoblasts promotes osteoclastogenesis. We have previously demonstrated that transforming growth factor (TGF)-ß induces the synthesis of M-CSF via SMAD2/3, p38 mitogen-activated protein kinase (MAPK), p44/p42 MAPK and c-Jun N-terminal kinase (JNK) in osteoblast-like MC3T3-E1 cells. In the present study, we investigated whether SERM affects the M-CSF synthesis by TGF-ß in MC3T3-E1 cells. Raloxifene and bazedoxifene significantly suppressed the synthesis of M-CSF. PPT, an ERα agonist, but not ERB041, an ERß agonist, inhibited the release of M-CSF. MPP, an ERα antagonist, reversed the suppression by raloxifene of the M-CSF release. Raloxifene attenuated the TGF-ß-induced phosphorylation of JNK but not SMAD3, p42 MAPK and p38 MAPK. Bazedoxifene and PPT also inhibited the phosphorylation of JNK. Furthermore, MPP, an ERα antagonist, reversed the suppression by both raloxifene and bazedoxifene of the phosphorylation of JNK. Our results strongly indicate that raloxifene and bazedoxifene, SERMs, suppress the TGF-ß-induced synthesis of M-CSF through ERα-mediated inhibition of JNK pathway in osteoblasts.


Subject(s)
Selective Estrogen Receptor Modulators , Transforming Growth Factor beta , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , MAP Kinase Signaling System , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Raloxifene Hydrochloride/metabolism , Raloxifene Hydrochloride/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Osteoblasts/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Phosphorylation
2.
Drug Test Anal ; 14(11-12): 2002-2006, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36229870

ABSTRACT

Tamoxifen and toremifene are two selective estrogen receptor modulators (SERMs) commonly used to treat breast cancer in women. Toremifene is well-known as a triphenylethylene derivative. Carboxy toremifene is a common metabolite of toremifene and tamoxifen. Since 2005, the World Anti-Doping Agency (WADA) has banned the SERMs category during in and out of competition. These substances are in the S4 category in the WADA prohibited list as "agents with anti-oestrogenic activity." However, there is no commercially accessible carboxy toremifene reference material in the market. This research highlights the novel synthetic procedure, the development of a carboxy toremifene HPLC method, and validation, along with detailed characterization using advanced analytical techniques using 1 H NMR, HRMS, FT-IR-ATR and UV-visible spectroscopy. RP-HPLC-DAD method was developed and validated to assess the purity of carboxy toremifene. Developed reference material has shown 100% purity. Therefore, we recommend that this synthesized carboxy toremifene may be used as reference material to strengthen the WADA-accredited lab to maintain a clean sports mission during sports competitions.


Subject(s)
Selective Estrogen Receptor Modulators , Toremifene , Female , Humans , Selective Estrogen Receptor Modulators/metabolism , Spectroscopy, Fourier Transform Infrared , Tamoxifen/metabolism , Tamoxifen/therapeutic use , Quality Control
3.
Chem Biol Interact ; 365: 110091, 2022 Sep 25.
Article in English | MEDLINE | ID: mdl-35944649

ABSTRACT

Estrogen receptor α (ERα) plays a key role in the adaptive response of liver metabolism to energy demands, especially in controlling lipid metabolism. Tamoxifen (TMX), a main drug for the treatment of ER-positive breast cancer in clinical, is a selective ER modulator (SERM). However, accordingly, the long-term use of TMX would lead to nonalcoholic fatty liver (NAFLD) in clinical, which had no definite treatment up to now. Fatostatin (Fato), an inhibitor of sterol-regulatory element binding protein 2 (SREBP2), was reported as a synergistic inhibitor of ER-positive breast cancer with TMX, but the hepatic lipid regulation of this combination is still unknown. Herein, we aimed to explore the effect and mechanism of Fato against TMX-induced NAFLD. The results identified that hepatic cholesterol content increase was the main reason for TMX-induced NAFLD. It was caused by the upregulation of circulating cholesterol uptake mediated by low density lipoprotein receptor (LDLR) in liver, which resulted from the activation of SREBP2. Meanwhile, Fato could inhibit activation of SREBP2-LDLR pathway, alleviating TMX-induced hepatic cholesterol accumulation. In summary, these results provided a new insight into the mechanism of TMX-induced NAFLD. Moreover, it supported the combination of Fato and TMX for the treatment of ER-positive breast cancer to reduce the adverse effect of TMX in clinical.


Subject(s)
Breast Neoplasms , Non-alcoholic Fatty Liver Disease , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cholesterol/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Liver , Non-alcoholic Fatty Liver Disease/metabolism , Pyridines , Receptors, LDL/metabolism , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Sterol Regulatory Element Binding Protein 2/metabolism , Tamoxifen/pharmacology , Thiazoles
4.
Mol Med ; 28(1): 44, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35468719

ABSTRACT

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.


Subject(s)
Bone Neoplasms , Breast Neoplasms , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology
5.
Antimicrob Agents Chemother ; 66(3): e0194321, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35041501

ABSTRACT

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.


Subject(s)
Alphavirus Infections , Chikungunya virus , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Line , Mice , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Viral Nonstructural Proteins , Virus Replication
6.
Autophagy ; 18(10): 2323-2332, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35025696

ABSTRACT

Maintenance of bone integrity is mediated by the balanced actions of osteoblasts and osteoclasts. Because macroautophagy/autophagy regulates osteoblast mineralization, osteoclast differentiation, and their secretion from osteoclast cells, autophagy deficiency in osteoblasts or osteoclasts can disrupt this balance. However, it remains unclear whether upregulation of autophagy becomes beneficial for suppression of bone-associated diseases. In this study, we found that genetic upregulation of autophagy in osteoblasts facilitated bone formation. We generated mice in which autophagy was specifically upregulated in osteoblasts by deleting the gene encoding RUBCN/Rubicon, a negative regulator of autophagy. The rubcnflox/flox;Sp7/Osterix-Cre mice showed progressive skeletal abnormalities in femur bones. Consistent with this, RUBCN deficiency in osteoblasts resulted in elevated differentiation and mineralization, as well as an increase in the elevated expression of key transcription factors involved in osteoblast function such as Runx2 and Bglap/Osteocalcin. Furthermore, RUBCN deficiency in osteoblasts accelerated autophagic degradation of NOTCH intracellular domain (NICD) and downregulated the NOTCH signaling pathway, which negatively regulates osteoblast differentiation. Notably, osteoblast-specific deletion of RUBCN alleviated the phenotype in a mouse model of osteoporosis. We conclude that RUBCN is a key regulator of bone homeostasis. On the basis of these findings, we propose that medications targeting RUBCN or autophagic degradation of NICD could be used to treat age-related osteoporosis and bone fracture.Abbreviations: ALPL: alkaline phosphatase, liver/bone/kidney; BCIP/NBT: 5-bromo-4-chloro-3'-indolyl phosphate/nitro blue tetrazolium; BMD: bone mineral density; BV/TV: bone volume/total bone volume; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NICD: NOTCH intracellular domain; RB1CC1/FIP200: RB1-inducible coiled-coil 1; RUBCN/Rubicon: RUN domain and cysteine-rich domain containing, Beclin 1-interacting protein; SERM: selective estrogen receptor modulator; TNFRSF11B/OCIF: tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin).


Subject(s)
Osteogenesis , Osteoporosis , Alkaline Phosphatase/metabolism , Animals , Autophagy/physiology , Beclin-1/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Cysteine/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Osteoblasts/pathology , Osteocalcin/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Osteoprotegerin/metabolism , Phosphates/metabolism , Receptors, Notch , Selective Estrogen Receptor Modulators/metabolism , Sirolimus , TOR Serine-Threonine Kinases/metabolism
7.
Psychoneuroendocrinology ; 121: 104830, 2020 11.
Article in English | MEDLINE | ID: mdl-32858306

ABSTRACT

Over 20 years of accumulated evidence has shown that the major female sex hormone 17ß-estradiol can enhance cognitive functioning. However, the utility of estradiol as a therapeutic cognitive enhancer is hindered by its unwanted peripheral effects (carcinogenic). Selective estrogen receptor modulators (SERMs) avoid the unwanted effects of estradiol by acting as estrogen receptor antagonists in some tissues such as breast and uterus, but as agonists in others such as bone, and are currently used for the treatment of osteoporosis. However, understanding of their actions in the brain are limited. The third generation SERM bazedoxifene has recently been FDA approved for clinical use with an improved biosafety profile. However, whether bazedoxifene can enter the brain and enhance cognition is unknown. Using mice, the current study aimed to explore if bazedoxifene can 1) cross the blood-brain barrier, 2) rescue ovariectomy-induced hippocampal-dependent spatial memory deficit, and 3) activate neural estrogen response element (ERE)-dependent gene transcription. Using liquid chromatography-mass spectrometry (LC-MS), we firstly demonstrate that a peripheral injection of bazedoxifene can enter the brain. Secondly, we show that an acute intraperitoneal injection of bazedoxifene can rescue ovariectomy-induced spatial memory deficits. And finally, using the ERE-luciferase reporter mouse, we show in vivo that bazedoxifene can activate the ERE in the brain. The evidence shown here suggest bazedoxifene could be a viable cognitive enhancer with promising clinical applicability.


Subject(s)
Cognition/drug effects , Indoles/pharmacology , Spatial Memory/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Estradiol/pharmacology , Estrogens/metabolism , Estrogens/pharmacology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Indoles/metabolism , Mice , Mice, Inbred C57BL , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Spatial Memory/physiology
8.
Cancer Sci ; 111(8): 2718-2725, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32536008

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , F-Box Proteins/metabolism , Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Movement/genetics , Cellular Senescence/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasms/pathology , Protein Subunits/metabolism , Proteolysis , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/metabolism , Signal Transduction/genetics , Ubiquitination
9.
J Pharmacol Exp Ther ; 369(3): 389-405, 2019 06.
Article in English | MEDLINE | ID: mdl-30918069

ABSTRACT

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.


Subject(s)
Biocatalysis/drug effects , Lithocholic Acid/analogs & derivatives , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Sulfonic Acids/metabolism , Sulfotransferases/metabolism , Taurolithocholic Acid/metabolism , Cytosol/drug effects , Cytosol/metabolism , Hep G2 Cells , Humans , Kinetics , Lithocholic Acid/metabolism , Liver/cytology , Oxidation-Reduction , Selective Estrogen Receptor Modulators/metabolism , Sulfotransferases/antagonists & inhibitors
10.
EMBO Mol Med ; 11(4)2019 04.
Article in English | MEDLINE | ID: mdl-30885958

ABSTRACT

Excessive signaling through gp130, the shared receptor for the interleukin (IL)6 family of cytokines, is a common hallmark in solid malignancies and promotes their progression. Here, we established the in vivo utility of bazedoxifene, a steroid analog clinically approved for the treatment of osteoporosis, to suppress gp130-dependent tumor growth of the gastrointestinal epithelium. Bazedoxifene administration reduced gastric tumor burden in gp130Y757F mice, where tumors arise exclusively through excessive gp130/STAT3 signaling in response to the IL6 family cytokine IL11. Likewise, in mouse models of sporadic colon and intestinal cancers, which arise from oncogenic mutations in the tumor suppressor gene Apc and the associated ß-catenin/canonical WNT pathway, bazedoxifene treatment reduces tumor burden. Consistent with the proposed orthogonal tumor-promoting activity of IL11-dependent gp130/STAT3 signaling, tumors of bazedoxifene-treated Apc-mutant mice retain excessive nuclear accumulation of ß-catenin and aberrant WNT pathway activation. Likewise, bazedoxifene treatment of human colon cancer cells harboring mutant APC did not reduce aberrant canonical WNT signaling, but suppressed IL11-dependent STAT3 signaling. Our findings provide compelling proof of concept to support the repurposing of bazedoxifene for the treatment of gastrointestinal cancers in which IL11 plays a tumor-promoting role.


Subject(s)
Drug Repositioning , Gastrointestinal Neoplasms/drug therapy , Indoles/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Proliferation/drug effects , Cytokine Receptor gp130/chemistry , Cytokine Receptor gp130/metabolism , Disease Models, Animal , Female , Gastrointestinal Neoplasms/pathology , Humans , Indoles/metabolism , Indoles/pharmacology , Interleukin-11/chemistry , Interleukin-11/metabolism , Interleukin-11/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , STAT3 Transcription Factor/metabolism , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , beta Catenin/metabolism
11.
Nat Commun ; 10(1): 1033, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833575

ABSTRACT

Taxanes are a family of natural products with a broad spectrum of anticancer activity. This activity is mediated by interaction with the taxane site of beta-tubulin, leading to microtubule stabilization and cell death. Although widely used in the treatment of breast cancer and other malignancies, existing taxane-based therapies including paclitaxel and the second-generation docetaxel are currently limited by severe adverse effects and dose-limiting toxicity. To discover taxane site modulators, we employ a computational binding site similarity screen of > 14,000 drug-like pockets from PDB, revealing an unexpected similarity between the estrogen receptor and the beta-tubulin taxane binding pocket. Evaluation of nine selective estrogen receptor modulators (SERMs) via cellular and biochemical assays confirms taxane site interaction, microtubule stabilization, and cell proliferation inhibition. Our study demonstrates that SERMs can modulate microtubule assembly and raises the possibility of an estrogen receptor-independent mechanism for inhibiting cell proliferation.


Subject(s)
Antineoplastic Agents/chemistry , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/pharmacology , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/metabolism , Taxoids/chemistry , Taxoids/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/metabolism , Tubulin/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Death/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Humans , Ligands , Microtubule Proteins/drug effects , Models, Molecular , Paclitaxel/pharmacology , Tubulin/drug effects , Tumor Microenvironment
12.
Front Neuroendocrinol ; 52: 44-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30223003

ABSTRACT

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.


Subject(s)
Astrocytes/drug effects , Brain Diseases/drug therapy , Neuroprotective Agents/pharmacology , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Animals , Humans
13.
Toxicology ; 408: 80-87, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30018014

ABSTRACT

How does estrogen receptor-α bind its natural ligands - estrogens? How can other molecules mimic estrogens and elicit different estrogenic responses? The answers lie in a complex and intimate chemical biology between ligands and receptor. This delicate interaction at the ligand binding cleft signals, via conformational change, exposure of a specific new charge topography at a second site (Activation Function-2). This, in turn, attracts a regulatory protein which modulates gene expression and controls biological activity.


Subject(s)
Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/drug effects , Estrogens/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Animals , Binding Sites , Estrogen Antagonists/chemistry , Estrogen Antagonists/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Estrogens/chemistry , Estrogens/metabolism , Gene Expression Regulation/drug effects , Humans , Ligands , Models, Molecular , Molecular Mimicry , Protein Binding , Protein Conformation , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/metabolism , Structure-Activity Relationship
14.
Toxicol Appl Pharmacol ; 353: 31-42, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29906493

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Osteoclasts/drug effects , Pyrrolidines/pharmacology , Receptor, Cannabinoid, CB2/agonists , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/analogs & derivatives , Adenylyl Cyclase Inhibitors/pharmacology , Animals , Benzoxazines/pharmacology , Binding, Competitive/drug effects , Bone Marrow Cells/drug effects , CHO Cells , Cell Differentiation/drug effects , Cricetinae , Cricetulus , Drug Inverse Agonism , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Naphthalenes/pharmacology , Pyrrolidines/metabolism , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Selective Estrogen Receptor Modulators/metabolism , Tamoxifen/metabolism , Tamoxifen/pharmacology
15.
J Pharm Biomed Anal ; 156: 147-152, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29704771

ABSTRACT

Antiestrogenic compounds such as tamoxifen, toremifen and chlomifen are used illegally by athletes to minimize physical impacts such as gynecomastia resulting from the secondary effects of anabolic androgenic steroids, used to increase athletic efficiency unlawfully. The use of these compounds is banned by the World Anti-Doping Agency (WADA) and controls are made through analytical methodologies such as HPLC-MS/MS, which do not fulfil the sample throughput requirements. Moreover, compounds such as tamoxifen are also used to treat hormone receptor-positive breast cancer (ER + ).Therapeutic drug monitoring (TDM) of tamoxifen may also be clinically useful for guiding treatment decisions. An accurate determination of these drugs requires a solid phase extraction of patient serum followed by HPLC-MS/MS. In the context of an unmet need of high-throughput screening (HTS) and quantitative methods for antiestrogenic substances we have approached the development of antibodies and an immunochemical assay for the determination of these antiestrogenic compounds. The strategy applied has taken into consideration that these drugs are metabolized and excreted in urine as the corresponding 4-hydroxylated compounds. A microplate-based ELISA procedure has been developed for the analysis of these metabolites in urine with a LOD of 0.15, 0.16 and 0.63 µg/L for 4OH-tamoxifen, 4OH-toremifen and 4OH-clomifen, respectively, much lower than the MRPL established by WADA (20 µg/L).


Subject(s)
Doping in Sports/prevention & control , Drug Monitoring/methods , Selective Estrogen Receptor Modulators/urine , Testosterone Congeners/urine , Breast Neoplasms/drug therapy , Chromatography, High Pressure Liquid , Clomiphene/metabolism , Clomiphene/therapeutic use , Clomiphene/urine , Enzyme-Linked Immunosorbent Assay , Female , Healthy Volunteers , High-Throughput Screening Assays/methods , Humans , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Solid Phase Extraction , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tamoxifen/therapeutic use , Tamoxifen/urine , Tandem Mass Spectrometry , Toremifene/metabolism , Toremifene/therapeutic use , Toremifene/urine
16.
Endocrinology ; 159(5): 1972-1981, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29579190

ABSTRACT

Estrogens bind to two nuclear estrogen receptor (ER) subtypes, ERα and ERß, which are expressed in differing amounts in various tissues. The endogenous estrogen, 17ß-estradiol (E2), binds to both subtypes with nearly equal affinity and is the prototypical agonist. Selective estrogen receptor modulators (SERMs) may bind to both subtypes with equivalent affinities but have agonist activities in some tissues while having antagonist activities in others. In the present study, we demonstrate that the first reported endogenous SERM, 27-hydroxycholesterol (27-OHC), binds preferentially (>100-fold) to ERß over ERα. Furthermore, 27-OHC is not able to fully compete with E2 binding, suggesting the two may bind at different sites. We provide an allosteric ternary complex model for the simultaneous binding of 27-OHC and E2 to ERß, which accurately describes the binding data we have observed. We conclude that 27-OHC is a negative allosteric modifier of E2 binding, with an inhibitor constantof 50 nM and cooperativity factor (α) of 0.036. We also propose an in silico three-dimensional model of the simultaneous binding to guide future experiments. Further study of this unique binding model may allow for the discovery of novel ERß-selective ligands and potentially explain the lack of effectiveness of ERß-selective agonists in humans vs preclinical models.


Subject(s)
Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Hydroxycholesterols/metabolism , Allosteric Regulation , Computer Simulation , Humans , In Vitro Techniques , Models, Molecular , Selective Estrogen Receptor Modulators/metabolism
17.
Arch Pharm Res ; 41(3): 347-353, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29516347

ABSTRACT

Clomiphene citrate, a selective estrogen receptor modulator, is metabolized into its 4-hydroxylated active metabolites, primarily by CYP2D6. In this study, we investigated the effects of the most common CYP2D6 variant allele in Asians, CYP2D6*10, on the pharmacokinetics of clomiphene and its two active metabolites (4-OH-CLO and 4-OH-DE-CLO) in healthy Korean subjects. A single 50-mg oral dose of clomiphene citrate was given to 22 Korean subjects divided into three genotype groups according to CYP2D6 genotypes, CYP2D6*wt/*wt (n = 8; *wt = *1 or *2), CYP2D6*wt/*10 (n = 8) and CYP2D6*10/*10 (n = 6). Concentrations of clomiphene and its metabolites were determined using a validated HPLC-MS/MS analytical method in plasma samples collected up to 168 h after the drug intake. There was a significant difference only in the Cmax of clomiphene between three CYP2D6 genotype groups (p < 0.05). Paradoxically, the elimination half-life (t1/2) and AUC of both active metabolites were all significantly increased in the CYP2D6*10 homozygous carriers, compared with other genotype groups (all p < 0.001). The AUCinf of corrected clomiphene active moiety in CYP2D6*10/*10 subjects was 2.95- and 2.05-fold higher than that of CYP2D6*wt/*wt and *wt/*10 genotype groups, respectively (both p < 0.001). Along with the partial impacts on the biotransformation of clomiphene and its metabolites by CYP2D6 genetic polymorphism, further studies on the effects of other CYP enzymes in a multiple-dosing condition can provide more definite evidence for the inter-individual variabilities in clomiphene pharmacokinetics and/or drug response.


Subject(s)
Alleles , Clomiphene/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Selective Estrogen Receptor Modulators/pharmacokinetics , Clomiphene/metabolism , Female , Humans , Male , Selective Estrogen Receptor Modulators/metabolism , Young Adult
18.
Bioorg Med Chem Lett ; 27(11): 2590-2593, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28400239

ABSTRACT

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.


Subject(s)
Benzhydryl Compounds/chemistry , Receptors, Estrogen/metabolism , Steroids/chemistry , Benzhydryl Compounds/chemical synthesis , Benzhydryl Compounds/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Receptors, Estrogen/antagonists & inhibitors , Selective Estrogen Receptor Modulators/chemical synthesis , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/metabolism , Steroids/chemical synthesis , Steroids/metabolism , Structure-Activity Relationship
19.
J Neurochem ; 141(1): 31-36, 2017 04.
Article in English | MEDLINE | ID: mdl-28075498

ABSTRACT

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.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Plasma Membrane Transport Proteins/physiology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/physiology , Tamoxifen/metabolism , Tamoxifen/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Estrogen Antagonists/metabolism , Estrogen Antagonists/pharmacology , Humans , Mice , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology
20.
Drug Test Anal ; 9(9): 1349-1362, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28054434

ABSTRACT

Selective estrogen receptor modulators (SERMs) are chemicals that possess the anti-oestrogenic activities that are banned 'in' and 'out' of competition by the World Anti-Doping Agency (WADA) in human sports, and by the International Federation of Horseracing Authorities (IFHA) in horseracing. SERMs can be used as performance-enhancing drugs to boost the level of androgens or to compensate for the adverse effects as a result of extensive use of androgenic anabolic steroids (AASs). SERMs have indeed been abused in human sports; hence, a similar threat can be envisaged in horseracing. Numerous analytical findings attributed to the use of SERMs have been reported by WADA-accredited laboratories, including 42 cases of tamoxifen and 2 cases of toremifene in 2014. This paper describes the identification of the in vitro phase I metabolites of tamoxifen and toremifene using ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS), with an aim to identify potential screening targets for doping control in equine sports. A total of 13 and 11 in vitro metabolites have been identified for tamoxifen and toremifene, respectively, after incubation with homogenized horse liver. The more prominent in vitro biotransformation pathways include N-desmethylation, hydroxylation, and carboxylation. In addition, this is the first report of some novel metabolites for both tamoxifen and toremifene with hydroxylation occurring at the N-methyl moiety. To our knowledge, this is the first study of the phase I metabolism of tamoxifen and toremifene in horses using homogenized horse liver. Copyright © 2017 John Wiley & Sons, Ltd.


Subject(s)
Anabolic Agents/analysis , Androgens/analysis , Selective Estrogen Receptor Modulators/metabolism , Toremifene/chemistry , Anabolic Agents/chemistry , Androgens/chemistry , Animals , Chromatography, High Pressure Liquid , Horses , Humans , Hydroxylation , Selective Estrogen Receptor Modulators/chemistry , Tandem Mass Spectrometry , Toremifene/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...