Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Curr Issues Mol Biol ; 45(9): 7336-7351, 2023 Sep 09.
Article in English | MEDLINE | ID: mdl-37754248

ABSTRACT

To identify effective treatment modalities for breast cancer with acquired resistance, we first compared the responsiveness of estrogen receptor-positive breast cancer MCF-7 cells and long-term estrogen-deprived (LTED) cells (a cell model of endocrine therapy-resistant breast cancer) derived from MCF-7 cells to G-1 and 2-methoxyestradiol (2-MeO-E2), which are microtubule-destabilizing agents and agonists of the G protein-coupled estrogen receptor 1 (GPER1). The expression of GPER1 in LTED cells was low (~0.44-fold), and LTED cells displayed approximately 1.5-fold faster proliferation than MCF-7 cells. Although G-1 induced comparable antiproliferative effects on both MCF-7 and LTED cells (IC50 values of >10 µM), 2-MeO-E2 exerted antiproliferative effects selective for LTED cells with an IC50 value of 0.93 µM (vs. 6.79 µM for MCF-7 cells) and induced G2/M cell cycle arrest. Moreover, we detected higher amounts of ß-tubulin proteins in LTED cells than in MCF-7 cells. Among the ß-tubulin (TUBB) isotype genes, the highest expression of TUBB2B (~3.2-fold) was detected in LTED cells compared to that in MCF-7 cells. Additionally, siTUBB2B restores 2-MeO-E2-mediated inhibition of LTED cell proliferation. Other microtubule-targeting agents, i.e., paclitaxel, nocodazole, and colchicine, were not selective for LTED cells. Therefore, 2-MeO-E2 can be an antiproliferative agent to suppress LTED cell proliferation.

2.
Biol Pharm Bull ; 46(7): 946-954, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37183024

ABSTRACT

Allergic contact dermatitis (ACD) is a common skin disorder caused by contact with allergens. The optimal treatment for ACD is to avoid contact with allergens. However, in some cases, avoiding exposure is not possible when the allergens are unknown. Therefore, establishing treatment methods other than allergen avoidance is important. We previously reported that the continuous administration of methionine, an essential amino acid, in a mouse model of atopic dermatitis alleviated its symptoms. In the present study, we investigated the effect of methionine on a mouse model of ACD caused by 1-fluoro-2,4-dinitrobenzene (DNFB). Differences in the effect of methionine were observed in DNFB-induced ACD model mice based on the mouse strain used. This difference was attributed to the suppression of hepatic dimethylglycine (DMG) production, which is associated with the suppression of hepatic betaine-homocysteine methyltransferase (Bhmt) expression by ACD. Although we did not reveal the mechanism underlying DMG suppression, our study suggests the presence of interactions between the liver and skin in dermatitis, such as the regulation of hepatic metabolic enzyme expression in dermatitis and the alleviation of dermatitis symptoms by the hepatic metabolism status of DMG.


Subject(s)
Dermatitis, Allergic Contact , Methionine , Mice , Animals , Dinitrofluorobenzene/toxicity , Dermatitis, Allergic Contact/drug therapy , Allergens , Racemethionine
3.
Curr Issues Mol Biol ; 44(9): 3849-3858, 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36135176

ABSTRACT

Experimental evidence accumulated by our research group and others strongly suggests that (-)-xanthatin, a xanthanolide sesquiterpene lactone, exhibits anti-proliferative effects on human breast cancer cells (in vitro) as well as anti-tumor effects in experimental animals (in vivo). In cancer biology, it is now critically important for anti-cancer agents to selectively target cancer stem cells (CSCs) in order to overcome cancer therapeutic resistance and recurrence. However, it has not yet been established whether (-)-xanthatin abrogates the formation of breast CSCs. In the present study, we utilized chemically synthesized pure (-)-xanthatin and a culture system to obtain mammospheres from human breast cancer MCF-7 cells, which are a CSC-enriched population. We herein demonstrated for the first time that (-)-xanthatin exhibited the ability to kill mammospheres, similar to salinomycin, an established selective killer of CSCs. A possible anti-proliferative mechanism toward mammospheres by (-)-xanthatin is discussed.

4.
Arch Biochem Biophys ; 731: 109428, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36228705

ABSTRACT

Cannabidiolic acid (CBDA) can activate peroxisome proliferator-activated receptor-α (PPARα) and PPARγ. Whether CBDA can activate PPARß/δ has not been examined sufficiently to date. Since previous studies showed that triple-negative breast cancer cells respond to activation of PPARß/δ, the present study examined the effect of CBDA in MDA-MB-231 cells and compared the activities of CBDA with known PPARß/δ agonists/antagonists. Expression of the PPARß/δ target genes angiopoietin-like 4 (ANGPTL4) and adipocyte differentiation-related protein (ADRP) was increased by CBDA. Interestingly, ligand activation of PPARß/δ with GW501516 caused an increase in expression of both ANGPTL4 and ADRP, but the magnitude of this effect was markedly increased when co-treated with CBDA. Specificity of these effects were confirmed by showing that CBDA-induced expression of ANGPTL4 and ADRP is mitigated in the presence of either a PPARß/δ antagonist or an inverse agonist. Results from these studies suggest that CBDA can synergize with PPARß/δ and might interact with endogenous agonists that modulate PPARß/δ function.


Subject(s)
Cannabinoids , PPAR delta , PPAR-beta , PPAR-beta/genetics , PPAR-beta/metabolism , PPAR delta/genetics , PPAR delta/metabolism , PPAR alpha
5.
Biol Pharm Bull ; 44(10): 1524-1529, 2021.
Article in English | MEDLINE | ID: mdl-34602561

ABSTRACT

Bisphenol A (BPA) has been shown to induce the activation of nuclear estrogen receptor α/ß (ERα/ß) in both in vitro and in vivo settings. We originally obtained a 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), a possible active metabolite of BPA, strongly activating the ERs-mediated transcription in MCF-7 cells with an EC50 of 2.8 nM (i.e., BPA's EC50 = 519 nM). Environmental estrogens can also target G protein-coupled estrogen receptor 1 (GPER1), a membrane-type ER. However, the effects of BPA/MBP on GPER1, have not yet been fully resolved. In this study, we used MCF-7, a ERα/ERß/GPER1-positive human breast cancer cell line, as a model to investigate the effects of the exposure to BPA or MBP. Our results revealed that at concentrations below 1 nM MBP, but not BPA, downregulates the expression of GPER1 mRNA via upregulated ERß, and the MCF-7 cells pre-treated with MBP display resistance to GPER1 agonist G-1-mediated anti-proliferative effects. Because GPER1 can act as a tumor suppressor in several types of cancer including breast cancer, the importance of MBP-mediated decrease in GPER1 expression in breast cancer cells is discussed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cyclopentanes/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Phenols/pharmacology , Quinolines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclopentanes/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Phenols/therapeutic use , Quinolines/therapeutic use , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
6.
Biochem Biophys Res Commun ; 531(2): 215-222, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32798015

ABSTRACT

The functional role of fatty acid 2-hydroxylase (FA2H) is controversial in the field of cancer biology due to the dual role of FA2H, particularly related to its interaction with triple-negative breast cancer (TNBC). A previous biochemical- and clinical-focused study suggested that FA2H could dampen TNBC aggressiveness. However, another epidemiological study demonstrated that FA2H expression is associated with shorter disease-free survival in TNBC cases. We reported that FA2H is a peroxisome proliferator-activated receptor α (PPARα)-regulated gene in human breast cancer MDA-MB-231 cells, in vitro experimental models for TNBC analysis. PPARα activation by its ligand reportedly results in an aggressive MDA-MB-231 cell phenotype, as well as estrogen receptor α (ERα)-positive MCF-7 cells. The results of this study show that i) MDA-MB-231 cells express very low levels of FA2H compared to the MCF-7 cells, reflecting a low basal-level PPARα-driven transcriptional activity compared to the MCF-7 cells, and ii) the increased FA2H expression stimulates the MDA-MB-231 and MCF-7 breast cancer cell migration without affecting proliferation. Taken together, our findings indicate that FA2H might be a breast cancer cell migration stimulator, independently of the ERα expression status.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Mixed Function Oxygenases/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Humans , Mixed Function Oxygenases/genetics , Mutation/genetics
7.
Mol Pharmacol ; 95(3): 260-268, 2019 03.
Article in English | MEDLINE | ID: mdl-30552153

ABSTRACT

Bisphenol A (BPA), recognized as an endocrine disruptor, is thought to exert its activity through a mechanism involving the activation of estrogen receptors (ERs) α/ß However, a major problem is that very high concentrations of BPA are required (i.e., those in excess of environmental levels) for effective activation of ERα/ß-mediated transcriptional activities in vitro, despite the BPA-induced estrogenic effects observed in vivo. To elucidate the causal reasons, we successfully identified a BPA metabolite, 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), which exhibits highly potent estrogenic activity both in vivo and in vitro. We have focused on the biologic relationship between breast tumor promotion and MBP/BPA, because BPA is considered to be a human carcinogen owing to its breast tumor-promoting properties. In general, humans are exposed to many endocrine disruptors, including BPA. In the present study, we used the ERα/ß-positive human breast cancer cell line MCF-7 as an experimental model to investigate the effects of repeated exposure to BPA/MBP at concentrations found in the environment on the expression of ERα/ß and to determine the particular ER subtype involved. We demonstrated that repeated exposure to MBP, but not to BPA, significantly downregulated ERα protein expression and stimulated the proliferation of MCF-7 cells through the activation of ERß-mediated signaling.


Subject(s)
Benzhydryl Compounds/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast/drug effects , Cell Proliferation/drug effects , Estrogen Receptor beta/metabolism , Phenols/pharmacology , Breast/metabolism , Cell Line, Tumor , Central Nervous System Stimulants/pharmacology , Down-Regulation/drug effects , Endocrine Disruptors/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Female , Humans , MCF-7 Cells , Signal Transduction/drug effects
8.
Arch Biochem Biophys ; 662: 219-225, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30553767

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are a family of ligand-activated nuclear transcription factors, with three characterized subtypes: PPARα, PPARß/δ, and PPARγ. The biological correlation between the two PPAR subtypes PPARα and γ and carcinogenesis is well-characterized; however, substantially less is known about the biological functions of PPARß/δ. PPARß/δ has been reported to repress transcription when PPARß/δ and PPARα or PPARγ are simultaneously expressed in some cells, and MDA-MB-231 cells express functional levels of PPARß/δ. We have previously reported that Δ9-tetrahydrocannabinol (Δ9-THC), a major cannabinoid component of the drug-type cannabis plant, can stimulate the expression of fatty acid 2-hydroxylase (FA2H) via upregulation of PPARα expression in human breast cancer MDA-MB-231 cells. Although the possibility of an inhibitory interaction between PPARα and PPARß/δ has not been demonstrated in MDA-MB-231 cells, we reasoned if this interaction were to exist, Δ9-THC should make PPARα free to achieve FA2H induction. Here, we show that a PPARß/δ-mediated suppression of PPARα function, but not of PPARγ, exists in MDA-MB-231 cells and Δ9-THC causes FA2H induction via mechanisms underlying the cancellation of PPARß/δ-mediated inhibition of PPARα, in addition to the upregulation of PPARα.


Subject(s)
Dronabinol/pharmacology , Mixed Function Oxygenases/genetics , PPAR alpha/biosynthesis , PPAR delta/metabolism , PPAR-beta/metabolism , Up-Regulation/drug effects , Cell Line, Tumor , Humans , PPAR delta/genetics , PPAR-beta/genetics , Sulfones/pharmacology , Thiophenes/pharmacology , Transcription, Genetic/drug effects
9.
Biol Pharm Bull ; 40(8): 1192-1198, 2017.
Article in English | MEDLINE | ID: mdl-28769000

ABSTRACT

Thiazolidinediones (TZDs) are known as peroxisome proliferator-activated receptor γ (PPARγ) activators, and are used in the treatment of diabetes. Although the usefulness of TZDs has been demonstrated, some of their side effects are becoming an obstacle to their clinical applicability; edema is known to be evoked by the "structural characteristics" of TZD, but not by the PPARγ activation. Thus, novel therapeutic modalities (i.e., non-TZD-type PPARγ activators) having different structures to those of TZDs are desired. We previously identified bongkrekic acid (BKA) as a PPARγ activator using the human breast cancer MCF-7 cell line as a model system. In the present study, we newly synthesized BKA analogs and examined the usefulness of BKA and its analogs as PPARγ activators in differentiated adipocyte cells. Among the chemicals investigated, one of the BKA analogs (BKA-#2) strongly stimulated PPARγ and the differentiation of 3T3-L1 cells similar to pioglitazone, a positive control. Furthermore, BKA-#2 reduced the size of lipid droplets in the mature adipocyte cells. The possible modulation mechanism by BKA-#2 is discussed.


Subject(s)
Bongkrekic Acid/analogs & derivatives , Bongkrekic Acid/pharmacology , PPAR gamma/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Lipid Droplets/drug effects , Mice
10.
Biol Pharm Bull ; 40(11): 1909-1916, 2017.
Article in English | MEDLINE | ID: mdl-29093337

ABSTRACT

Bisphenols are endocrine disruptors that are widely found in the environment. Accumulating experimental evidence suggests an adverse interaction between bisphenols and estrogen signaling. Most studies have performed experiments that focused on estrogen receptor (ER) engagement by bisphenols. Therefore, the effects of bisphenols on the expression of ERα (ESR1) and ERß (ESR2) remain largely unknown. In the present study, we examined the effects of four bisphenols: bisphenol A (BPA), bisphenol B (BPB), bisphenol S (BPS), and bisphenol AF (BPAF), on estrogen signaling in two human breast cancer cell lines (MCF-7 and SK-BR-3). Among these bisphenols, BPAF up-regulated the expression of ERß, and this was coupled with the abrogation of estrogen response element (ERE)-mediated transcriptional activities as well as the down-regulation of Cdc2 expression in MCF-7 cells, without influencing the expression of ERα. BPAF functioned as an agonist of ERα at lower concentrations (nanomolar order), but did not exhibit any modulatory action on ERα transiently expressed in SK-BR-3 cells in the presence or absence of 17ß-estradiol (E2) at higher concentrations (micromolar order). The introduction of ERß cDNA resulted in greater reductions in MCF-7 cell viability than with BPAF alone. Since ERß is a suppressive molecule of ERα function, these results provide rational evidence for BPAF functioning as an anti-estrogenic compound via the induction of ERß at higher concentrations.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Phenols/pharmacology , Signal Transduction/drug effects , CDC2 Protein Kinase/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Endocrine Disruptors/pharmacology , Estradiol/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Response Elements/drug effects , Transcription, Genetic/drug effects , Up-Regulation
11.
Biol Pharm Bull ; 44(11): 1593, 2021.
Article in English | MEDLINE | ID: mdl-34719636
12.
Drug Metab Dispos ; 42(2): 229-38, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24255116

ABSTRACT

Functional protein-protein interactions between UDP-glucuronosyltransferase (UGT)1A isoforms and cytochrome P450 (CYP)3A4 were studied. To this end, UGT1A-catalyzed glucuronidation was assayed in Sf-9 cells that simultaneously expressed UGT and CYP3A4. In the kinetics of UGT1A6-catalyzed glucuronidation of serotonin, both Michaelis constant (Km) and maximal velocity (Vmax) were increased by CYP3A4. When CYP3A4 was coexpressed with either UGT1A1 or 1A7, the Vmax for the glucuronidation of the irinotecan metabolite (SN-38) was significantly increased. S50 and Km both which are the substrate concentration giving 0.5 Vmax were little affected by simultaneous expression of CYP3A4. This study also examined the catalytic properties of the allelic variants of UGT1A1 and 1A7 and their effects on the interaction with CYP3A4. Although the UGT1A1-catalyzing activity of 4-methylumbelliferone glucuronidation was reduced in its variant, UGT1A1*6, the coexpression of CYP3A4 restored the impaired function to a level comparable with the wild type. Similarly, simultaneous expression of CYP3A4 increased the Vmax of UGT1A7*1 (wild type) and *2 (N129K and R131K), whereas the same was not observed in UGT1A7*3 (N129K, R131K, and W208R). In the kinetics involving different concentrations of UDP-glucuronic acid (UDP-GlcUA), the Km for UDP-GlcUA was significantly higher for UGT1A7*2 and *3 than *1. The Km of UGT1A7*1 and *3 was increased by CYP3A4, whereas *2 did not exhibit any such change. These results suggest that (1) CYP3A4 changes the catalytic function of the UGT1A subfamily in a UGT isoform-specific manner and (2) nonsynonymous mutations in UGT1A7*3 reduce not only the ability of UGT to use UDP-GlcUA but also CYP3A4-mediated enhancement of catalytic activity, whereas CYP3A4 is able to restore the UGT1A1*6 function.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Biotransformation , Camptothecin/analogs & derivatives , Camptothecin/metabolism , Catalysis , Cytochrome P-450 CYP3A/genetics , Glucuronosyltransferase/genetics , Humans , Hymecromone/metabolism , Isoenzymes , Kinetics , Mutation , Protein Interaction Mapping , Serotonin/metabolism , Sf9 Cells , Substrate Specificity , Transfection
13.
Biol Pharm Bull ; 37(9): 1435-8, 2014.
Article in English | MEDLINE | ID: mdl-25177025

ABSTRACT

Δ(9)-Tetrahydrocannabinol (Δ(9)-THC), a biologically active constituent of marijuana, possesses a wide variety of pharmacological and toxicological effects (e.g., analgesia, hypotension, reduction of inflammation, and anti-cancer effects). Among Δ(9)-THC's biological activities, its recognized anti-estrogenic activity has been the subject of investigations. Since Δ(9)-THC is used as both a drug of abuse (marijuana) and as a preventive therapeutic to treat pain and nausea in cancer patients undergoing chemotherapy in the United States and other countries (synthesized Δ(9)-THC; dronabinol), it is important to investigate the mechanistic basis underlying the anti-estrogenic activity of Δ(9)-THC. Since Δ(9)-THC has "no" binding potential for estrogen receptor α (ERα) which can be activated by estrogen (E2), the question of how Δ(9)-THC exerts its inhibitory effect on ERα is not resolved. We have recently reported that ERß, a second type of ER, is involved in the Δ(9)-THC abrogation of E2/ERα-mediated transcriptional activity. Here we discuss the possible mechanism(s) of the Δ(9)-THC-mediated disruption of E2/ERα signaling by presenting our recent findings as well.


Subject(s)
Dronabinol/pharmacology , Endocrine Disruptors/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Animals , Estradiol/metabolism , Humans , Signal Transduction , Up-Regulation
14.
Biol Pharm Bull ; 37(2): 331-4, 2014.
Article in English | MEDLINE | ID: mdl-24492731

ABSTRACT

Few studies have examined xanthocidin, a biotic isolated from Streptomyces xanthocidicus in 1966, because its supply is limited. Based on its chemical structure, xanthocidin has the potential to become a lead compound in the production of agrochemicals and anti-cancer drugs; however, it is unstable under both basic and acidic conditions. We recently established the total synthesis of xanthocidin using the FeCl3-mediated Nazarov reaction, and obtained two stable derivatives (#1 and #2). The results of the present study demonstrated that these derivatives exhibited the inhibitory activity of topoisomerase IIα, known as a molecular target for cancer chemotherapy, and this was attributed to the respective exo-methylene ketone group without DNA intercalation. The results obtained also suggest that these derivatives may have value as lead compounds in the synthesis of topoisomerase IIα inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Biological Products/pharmacology , Cyclopentanes/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Streptomyces/chemistry , Topoisomerase II Inhibitors/pharmacology , Antigens, Neoplasm , Antineoplastic Agents/chemical synthesis , DNA Topoisomerases, Type II , Humans , Molecular Structure , Topoisomerase II Inhibitors/chemical synthesis
15.
Chem Res Toxicol ; 26(7): 1073-9, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23718638

ABSTRACT

Δ(9)-Tetrahydrocannabinol (Δ(9)-THC) has been reported as possessing antiestrogenic activity, although the mechanisms underlying these effects are poorly delineated. In this study, we used the estrogen receptor α (ERα)-positive human breast cancer cell line, MCF-7, as an experimental model and showed that Δ(9)-THC exposures markedly suppresses 17ß-estradiol (E2)- induced MCF-7 cell proliferation. We demonstrate that these effects result from Δ(9)-THC's ability to inhibit E2-liganded ERα activation. Mechanistically, the data obtained from biochemical analyses revealed that (i) Δ(9)-THC up-regulates ERß, a repressor of ERα, inhibiting the expression of E2/ERα-regulated genes that promote cell growth and that (ii) Δ(9)-THC induction of ERß modulates E2/ERα signaling in the absence of direct interaction with the E2 ligand binding site. Therefore, the data presented support the concept that Δ(9)-THC's antiestrogenic activities are mediated by the ERß disruption of E2/ERα signaling.


Subject(s)
Dronabinol/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Dronabinol/chemistry , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/biosynthesis , Humans , Ligands , MCF-7 Cells , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Forensic Toxicol ; 41(2): 287-293, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36583834

ABSTRACT

PURPOSE: The effects of extended Δ9-tetrahydrocannabinol (Δ9-THC) exposure on estrogen receptor-positive human breast cancer MCF-7 cells have been investigated; however, the effects of Δ9-THC exposure for a shorter duration remain unclear. In this study, we sought to study whether Δ9-THC stimulates the migration of MCF-7 cells under both estrogenic and estrogen-deprived conditions over a short period (approximately 6 h). METHODS: MCF-7 cells were treated with Δ9-THC under estrogenic or estrogen-deprived conditions, and cell migration was subsequently analyzed. RESULTS: Δ9-THC-stimulated migration of MCF-7 cells 6 h after exposure was only observed in the estrogen-deprived condition. However, Δ9-THC-mediated migration was counteracted under estrogenic conditions without affecting cell proliferation and estrogen receptor expression during this period. CONCLUSIONS: Δ9-THC can stimulate MCF-7 cell migration under estrogen-deprived conditions; however, there is an interfering interaction between Δ9-THC and the estrogenic milieu that influences the migration of MCF-7 cells.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Dronabinol/pharmacology , Receptors, Estrogen/genetics , MCF-7 Cells , Estrogens/pharmacology , Estrone , Cell Movement
17.
Toxicol Lett ; 378: 31-38, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36863540

ABSTRACT

It was previously identified that there may be an active metabolite of bisphenol A (BPA), 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP). An in vitro system was developed to detect MBP toxicity to the Michigan Cancer Foundation-7 (MCF-7) cells that had been repeatedly exposed to a low dose of the metabolite. MBP profoundly activated estrogen receptor (ER)-dependent transcription as a ligand, with an EC50 of 2.8 nM. Women are continuously exposed to numerous estrogenic environmental chemicals; but their susceptibility to these chemicals may be significantly altered after menopause. Long-term estrogen-deprived (LTED) cells, which display ligand-independent ER activation, are a postmenopausal breast cancer model derived from MCF-7 cells. In this study, we investigated the estrogenic effects of MBP on LTED cells in a repeated exposure in vitro model. The results suggest that i) nanomolar levels of MBP reciprocally disrupt the balanced expression of ERα and ERß proteins, leading to the dominant expression of ERß, ii) MBP stimulates ERs-mediated transcription without acting as an ERß ligand, and iii) MBP utilizes mitogen-activated protein kinase and phosphatidylinositol-3 kinase signaling to evoke its estrogenic action. Moreover, the repeated exposure strategy was effective for detecting low-dose estrogenic-like effects caused by MBP in LTED cells.


Subject(s)
Breast Neoplasms , Receptors, Estrogen , Humans , Female , Receptors, Estrogen/genetics , Estradiol/toxicity , MCF-7 Cells , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Ligands , Estrogens , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism
18.
Genes Cells ; 16(12): 1200-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22093184

ABSTRACT

A repressor composed of homodimeric subunits, as is often found in bacteria, possesses two effector-binding sites per molecule, enabling sophisticated regulation by the cooperative binding of two effector molecules. Positive cooperativity generates a narrower region of effector concentration for switching, but little is known about the role of negative cooperativity. d-camphor, an inducer for Pseudomonas putida cytochrome P450cam hydroxylase operon (camDCAB), binds to the homodimeric cam repressor (CamR). Here, we report solid evidence that the complex of CamR and an operator DNA is not dissociated by the first binding of d-camphor but, at a higher concentration, is dissociated by the second binding. d-camphor thus binds to the CamR in two steps with negative cooperativity, yielding two distinct dissociation constants of K(d1 ) =( ) 0.064 ± 0.030 and K(d2 ) =( ) 14 ± 0.3 µm, as well as the Hill coefficient of 0.56 ± 0.05 (<1). The first binding guarantees the high specificity of the inducer by the high affinity, although the second binding turns on the gene expression at a 200-fold higher concentration, a more suitable switching point for the catabolism of d-camphor.


Subject(s)
Bacterial Proteins/metabolism , Camphor 5-Monooxygenase/metabolism , Multiprotein Complexes/metabolism , Pseudomonas putida/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Bacterial Proteins/genetics , Binding Sites , Camphor/metabolism , Camphor/pharmacology , Camphor 5-Monooxygenase/genetics , Cloning, Molecular , DNA, Bacterial/metabolism , Electrophoretic Mobility Shift Assay , Escherichia coli , Fluorometry , Gene Expression Regulation, Bacterial/drug effects , Kinetics , Multiprotein Complexes/genetics , Operon , Plasmids , Protein Binding/drug effects , Pseudomonas putida/drug effects , Pseudomonas putida/genetics , Recombinant Proteins/genetics , Repressor Proteins/genetics , Transformation, Bacterial
19.
J Toxicol Sci ; 47(4): 159-168, 2022.
Article in English | MEDLINE | ID: mdl-35370244

ABSTRACT

Detailed in vitro studies on the effects of perfluorooctanoic acid (PFOA) have demonstrated that activation of peroxisome proliferator-activated receptor α (PPARα) is a key process by which PFOA affects the malignancy of estrogen receptor α (ERα)-positive breast cancer cells. However, there is very little information on the PPARα-regulated genes responsible for the effects of PFOA in ERα-negative breast cancer cell malignancy. We recently demonstrated that fatty acid 2-hydroxylase (FA2H) stimulates the migration of ERα-negative human MDA-MB-231 cells, and PPARα is a key factor for the induction of FA2H in these cells. However, evidence for the relationship between PFOA exposure and PPARα-FA2H axis-driven migration has not been obtained. Here we analyzed the effects of PFOA on PPARα transcription and FA2H expression in relation to MDA-MB-231 cell migration. We found that simultaneously with stimulated migration, PFOA upregulated FA2H and activated the transcription of PPARα. FA2H-selective siRNA, but not siRNA control, clearly dampened PFOA-mediated cell migration. There is an inhibitory interaction between PPARα and PPARß/δ (i.e., PPARß/δ can suppress PPARα-mediated transcription) in MDA-MB-231 cells, but even in the presence of PPARß/δ expression, PFOA appeared to free PPARα to upregulate FA2H. Collectively, our findings show that i) PFOA activates PPARα-mediated transcription, ii) PFOA stimulates migration dependent on FA2H expression, and iii) mechanistically, PFOA relieves PPARß/δ suppression of PPARα activity to upregulate FA2H in MDA-MB-231 cells.


Subject(s)
Receptors, Estrogen , Triple Negative Breast Neoplasms , Caprylates/toxicity , Cell Movement , Fluorocarbons , Humans , Mixed Function Oxygenases/genetics
20.
Chem Res Toxicol ; 24(6): 855-65, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21568272

ABSTRACT

exo-Methylene lactone group-containing compounds, such as (--)-xanthatin, are present in a large variety of biologically active natural products, including extracts of Xanthium strumarium (Cocklebur). These substances are reported to possess diverse functional activities, exhibiting anti-inflammatory, antimalarial, and anticancer potential. In this study, we synthesized six structurally related xanthanolides containing exo-methylene lactone moieties, including (--)-xanthatin and (+)-8-epi-xanthatin, and examined the effects of these chemically defined substances on the highly aggressive and farnesyltransferase inhibitor (FTI)-resistant MDA-MB-231 cancer cell line. The results obtained demonstrate that (--)-xanthatin was a highly effective inhibitor of MDA-MB-231 cell growth, inducing caspase-independent cell death, and that these effects were independent of FTase inhibition. Further, our results show that among the GADD45 isoforms, GADD45γ was selectively induced by (--)-xanthatin and that GADD45γ-primed JNK and p38 signaling pathways are, at least in part, involved in mediating the growth inhibition and potential anticancer activities of this agent. Given that GADD45γ is becoming increasingly recognized for its tumor suppressor function, the results presented here suggest the novel possibility that (--)-xanthatin may have therapeutic value as a selective inducer of GADD45γ in human cancer cells, in particular in FTI-resistant aggressive breast cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Furans/chemistry , Furans/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Xanthium/chemistry , Antineoplastic Agents, Phytogenic/chemical synthesis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type I/metabolism , Female , Furans/chemical synthesis , Gene Expression Regulation, Neoplastic/drug effects , Heme Oxygenase-1/genetics , Humans , Interleukin-18/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , GADD45 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL