1,337
Views
8
CrossRef citations to date
0
Altmetric
Original Research Paper

Rapid, nongenomic signaling effects of several xenoestrogens involved in early- vs. late-stage prostate cancer cell proliferation

&
Article: e995003 | Received 11 Jun 2014, Accepted 26 Nov 2014, Published online: 25 Mar 2015

Abstract

Xenoestrogens (XEs) are exogenous mimics capable of binding to estrogen receptors (ERs), competing with/disrupting the actions of physiological estrogens, and promoting tumor growth in the prostate and other endocrine tissues. Humans are exposed to numerous XEs including environmental contaminants such as plastics monomer bisphenol A (BPA), and dietary phytoestrogens such as coumestrol and genistein from soy, and resveratrol, highest in red grapes. There is growing interest in the ability of phytoestrogens to prevent or treat tumors. We previously reported that multiple cellular mechanisms influence the number of prostate cancer cells after estradiol or diethylstilbestrol treatment. We now examine the effect of these XEs on signaling mechanisms that alter the number of LAPC-4 (androgen-dependent) and PC-3 (androgen-independent) cells at environment- and diet-relevant concentrations. Coumestrol and genistein both increased the number of LAPC-4 and PC-3 cells dramatically. Rapid alterations of phospho- and total-cyclin D1 levels most closely correlated with the XE-induced changes in cell numbers. Sustained activation (phosphorylation) of the extracellular signal-regulated kinases 1 and 2 as a prelude to generation of reactive oxygen species also partially contributed to the XE's effects on cell numbers. Early-stage cells expressed higher levels of all 3 ERs (including those in membranes) than did late-stage cells; ER subtypes were variably involved in the signaling responses. Taken together, these results show that each XE can elicit its own signature constellation of signaling responses, highlighting the importance of managing exposures to both environmental and dietary XEs for existing prostate tumors. These mechanisms may offer new cellular targets for therapy.

Introduction

Prostate cancers are well-known for their initial androgen responsiveness, which diminishes with the progression of disease stage. While the corresponding decrease in androgen receptor (AR) levels that accompanies this decline in responses has been well documented,Citation1 little is known about the relationship of tumor progression with the estrogen receptor (ER) types that might be involved, such as those that are thought to mediate the therapeutic effects of the pharmaceutical estrogen diethylstilbestrol (DES). There are many types of xenoestrogens (XEs) – exogenous estrogen-like compounds that bind to ER ligand binding pockets.Citation2-4 In normal or cancer cells, they imitate, compete with, or disrupt the actions of physiological estrogens.Citation5,6 Some XEs are known to promote tumor development in many tissues by stimulating inappropriate endocrine responses via ERs, promoting angiogenesis, increasing DNA adducts, or altering the epigenome.Citation7-12 Actions of XEs via ERs have also been shown to cause the proliferation of established endocrine tumors or tumor cell lines of many types, including those from brain, breast, kidney, lung, pancreas, prostate, and testis.Citation13-20

Alternatively, some XEs, especially dietary compounds, have been credited with preventing tumors in some of these tissues,Citation21–24 highlighting a broad range of XE response profiles. Genistein is a phytoestrogen found in soy products, fava beans, and some coffee bean preparationsCitation25 that can cause cell cycle arrest and growth inhibition at concentrations within the ranges achieved by the diets of some cultures (10−8 M to 10−6 M), via the down regulation of cyclin B.Citation26,27 Coumestrol (found in red clover, alfalfa sprouts, and also some soy products) can kill breast and colon cancer cells by producing reactive oxygen species (ROS).Citation28 Resveratrol, found in grapes, can decrease cell numbers by increasing intracellular calcium levels, disrupting G1/S progression, and stimulating apoptosis.Citation29-33

XEs also include environmental contaminants from the manufacture, use, or leachates of consumer products (e.g. plastics, chlorinated pesticides, alkylphenol surfactants). Bisphenol A (BPA) is a component of many plastic products (such as water bottles, food containers, receipt paper, and the inner coatings of food cans Citation34) and leaches from these products more readily with heat or acidity. As a result, BPA is a common environmental and human/animal contaminant Citation35 that has been shown to alter cell proliferation,Citation36 cell signaling through the activation of mitogen-activated protein kinases (MAPKs)Citation36-38 and intracellular calcium levels,Citation39,40 prostate cancer cell migration,Citation41 and increase susceptibility to certain diseases.Citation7,42

Epidemiological studies generally support an association between diets high in phytoestrogens and low cancer incidence.Citation43–45 African-Americans have a higher incidence of prostate cancer,Citation46 and dietary differences are being investigated as a possible factor in tumor development and progression within that population. East Asians consume high amounts of phytoestrogens, and their incidence of many types of cancer, including prostate cancer, is much less.Citation45 Asian diets contain high levels of soy ingredients, with the best-known active estrogenic components being daidzein, genistein, and coumestrol.Citation47 Genetics may also play a role in the sensitivity of various cancer-relevant mechanisms to estrogens, including the ability to metabolize dietary phytoestrogens to more active compounds,Citation48 though this can also be due to the type of gut microbiome present.Citation49

Until recently, most studies on XEs were focused on the gene expression (genomic) consequences of exposures.Citation50-52 Even at high concentrations, XEs generally elicited only low levels of transcriptional responses, so these compounds were thus labeled as weak estrogens.Citation50,53 However, XEs can also initiate non-genomic responses, so classifying them as weak without taking these more rapid cellular responses into consideration may be misleading.Citation15,20,54-58 Some endogenous estrogen metabolites such as estriol were formerly labeled as weak because of their limited ability to activate specific transcription, but have recently been found to have profound effects on disease expression.Citation59,60 Estriol, like XEs can have quite potent effects on nongenomic responses.Citation61 Because of this belief that XEs were weak, many past studies did not evaluate estrogens at environmentally relevant low doses (reviewed inCitation62). Dose responses to estrogens are typically non-monotonic and therefore must be assessed over a wide and detailed range of concentrations (reaching down to the femtomolar to nanomolar range) to predict their ability to act, especially at relevant environmental and dietary levels.Citation9,20,61,63

We have previously shown that XEs can rapidly activate cellular signaling pathways in tumor cells of other tissues (pituitary, breast, adrenal), and when in combination with them can modify the actions of physiological estrogens.Citation37,38,55,64-66 MAPKs can be rapidly activated or deactivated by XEs, leading to alterations in such functional end points as proliferation, apoptosis, and prolactin release.Citation20,67 Moreover, these MAPK phosphorylations [such as those for the extracellular signal regulated kinases (ERKs), c-Jun N-terminal kinases (JNKs), and p38 kinase] often occur at low physiological estrogen and XE concentrations.Citation20,37,68

We also recently demonstrated that estradiol (E2) and diethylstilbestrol (DES) can rapidly stimulate or deactivate ERKs in LAPC-4 and PC-3 prostate cancer cells,Citation69 and when sustained, cause ROS generation, contributing to a decrease in viable cells.Citation69,70 In addition, estrogen-induced rapid phosphorylation of cyclin D1 led to its subsequent prompt degradation, which in turn was correlated to the ability of E2 and DES to inhibit growth of these cells.Citation69 We will now investigate if some XEs also alter the viability of prostate cancer tumor cells via these mechanisms. Elucidating how these XEs function in early- vs. late-stage prostate tumor cells could lead to selective advice for patients about diet and exposure to environmental estrogens.

Materials and Methods

Cell lines and hormones

We chose cell lines representing the 2 main types of prostate cancers – androgen-dependent vs. androgen-independent. LAPC-4 androgen-dependent prostate cancer cells (passages 45–50)Citation71 were maintained to sub-confluence in phenol red-free Iscove's Modified Dulbecco's Medium (IMDM; MediaTech, Manassas, VA) with 10% fetal bovine serum (FBS, Atlanta Biologicals, Lawrenceville, GA), 4 mM L-glutamine (Sigma-Aldrich, St. Louis, MO), and 10−9 M dihydrotestosterone (Sigma-Aldrich). PC-3 androgen-independent prostate cancer cells (passages 18–23)Citation72 were maintained by growth in phenol red-free RPMI 1640 (Sigma-Aldrich) with 10% FBS and 2 mM L-glutamine. Both cell lines were propagated at 37°C in 5% CO2. BPA, coumestrol, genistein, and resveratrol (all from Sigma-Aldrich) were dissolved in ethanol to a stock concentration of 10 mM before serial dilution into IMDM or RPMI 1640 at concentrations ranging from 10−14M to 10−6M (and a final EtOH concentration of 0.0001%).

MTT cell viability assay

Cells were plated at 5,000 cells/well in poly-D-lysine-coated (BD Biosciences, Bedford, MA) 96-well assay plates, (Corning, Tewksbury, MA), and then allowed to attach overnight. The next day, XE treatments were added in 100 μL of medium with 1% 4x charcoal-stripped FBS. Extensive charcoal stripping of serum was done to remove and thus minimize the effect of any steroid hormones already present; these conditions were previously optimized to demonstrate effects of steroids and mimics on cell proliferation for these cell lines. After three days, treatments were removed and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT, Sigma-Aldrich) was added for 1 h. Cells were then lysed and the signal read at 590 nm in a Wallac 1420 plate reader (Perkin Elmer, Waltham, MA).

Plate immunoassays

Phosphorylated proteins were recognized by antibodies (Abs) specific for these post-translationally modified epitopes: pERK1/2 (Thr202/Tyr204) and phospho-cyclin D1 (Thr286) (both from Cell Signaling, Danvers, MA). Changes in total cyclin D1 levels were measured by using an Ab to cyclin D1 recognizing both modified and unmodified protein cyclin D1 (Cell Signaling Cat. No. 2922). ER Abs used included ERα (MC-20, Santa Cruz Biotechnology, Dallas, TX); ERβ (clone 9.88, Sigma-Aldrich); and GPR30 (Cat. No. NLS4271, Novus Biologicals, Littleton, CO). Membrane and total ER levels were measured by controlling for permeabilization of the cell membrane. A plate immunoassay developed in our labCitation73 and used in many of our past studies was recently adaptedCitation74 for use with the BIOMEK FXP workstation (Beckman Coulter, Brea, CA) to automate the majority of the plate assay's liquid handling, decreasing experimental variability and increasing experimental output.

Prostate cancer cells were plated at 10,000 cells/well in 96-well assay plates, allowed to attach overnight, and given 100 μL of medium with 1% 4x charcoal-stripped FBS for 48 h. Cells were then treated with XEs for up to 60 min on the workstation, followed by fixation (2% paraformaldehyde, 1% gluteraldehyde) ± permeabilization (0.15 M sucrose, 0.5% Nonidet P-40; to access internal vs. extracellular epitopes). The primary Ab to the phosphorylated epitopes was then added and incubated with the cells overnight. The next day, biotinylated anti-mouse/anti-rabbit IgG secondary Ab (Vector Labs, Burlingame, CA) was added for 1 h. Next, cells were incubated for 1 h with avidin-biotinylated conjugated alkaline phosphatase (ABC-AP, Vector Labs), then for 30 min with para-nitrophenylphosphate substrate (Thermo Scientific, Rockford, IL), allowing the yellow color of the para-nitrophenyl product to accumulate. Plates were read at 405 nm in a Wallac 1420 plate reader. Readings were then normalized to cell number, estimated by the crystal violet dye (Sigma Aldrich) assay as described previously.Citation73

Subtype-selective ER antagonist assays

To further determine ER subtype involvement in altering ROS formation or cyclin D1 phosphorylation, the following ER antagonists were used at their receptor-selective concentrations: for ERα, 10−7M MPP; for ERβ, 10−6M PHTPP; and for GPR30, 10−6M G15 (all from Tocris Bioscience, Minneapolis, MN). All were dissolved in ethanol to a stock concentration of 10 mM, then serially diluted into culture medium. Final ethanol concentrations were 0.0001%, which was used as vehicle control for all studies. Cells were incubated with antagonists for 30 min before XE treatments.

ROS assays

Cells were plated at 10,000 cells/well in a 96-well assay plate, then allowed to attach overnight. Cells were then treated with 100 μL of medium containing 1% 4x charcoal-stripped FBS for 48 h. 2’,7’-Dichlorodihydrofluorescein diacetate (DCDHF, Enzo Life Sciences, Farmingdale, NY; 15 μM) was loaded into cells for 1 h, and XE treatments were then administered for 15 min. Hydrogen peroxide (Fisher Scientific, Pittsburg, PA) and ethanol (0.0001%) were used as positive and negative controls, respectively. E2 (1 nM) was a positive control for previously determined estrogenic responses.Citation69 Dichlorofluorescein production, formed as a result of ROS/DCDHF interaction, was measured at an excitation of 485 nm, and an emission of 538 nm in a SpectraMax M3 Multi-Mode Microplate Reader (Molecular Devices).

Statistics

All experiments were conducted a minimum of 3 times. One-way analysis of variance was conducted for all experiments except ER quantification, which was analyzed using a Student's t-test. A Holm-Sidak post hoc test was used to measure the significance of each treatment versus the vehicle control. Significance was set at P < 0.05, unless otherwise stated.

Results and Discussion

XE effects on the number of viable cells

XEs at environment- or diet-relevant concentrations caused some increases in the numbers of LAPC-4 and PC-3 prostate cancer cells, observed here after 3 d of exposure in media containing 1% charcoal-stripped serum (). Coumestrol increased viable cell numbers at all but the lowest concentration assessed (10−14M) in both cell lines (by >200% in LAPC-4 cells, >400% in PC-3 cells). There was a strikingly different response to genistein between cell lines representing different tumor stages; genistein did not affect LAPC-4 cells, while it strongly stimulated the growth of PC-3 cells (by ∼4-fold at concentrations from 0.1 nM to 1 μM, all levels achievable by some diets). BPA caused a small stimulatory effect in LAPC-4 cells, at concentrations >10−12M (maximal increases of ∼40–50%), but had no effect on PC-3 cells. Resveratrol showed minimal, but significant stimulation compared to vehicle in both cell lines at most concentrations tested. For comparison to other estrogens assessed in our other studiesCitation69 (see insets), even at physiological (10−10–10−8M) concentrations E2 instead caused a significant decrease in cell viability for both cell lines by ∼20–30%. The actions of these dietary estrogens in causing prostate cancer cell growth are perhaps unexpected, given the epidemiological evidence that cultures with diets rich in genistein and coumestrol show decreased levels of prostate cancer.Citation75,76 However, some other studies have also shown a prostate cell proliferation effect by these phytoestrogens.Citation77–79

Figure 1. Cell number after 3 d of XE treatment. LAPC-4 and PC-3 prostate cancer cells were treated with XEs and viable cells were measured by the MTT assay. In all figures throughout the manuscript white symbols denote LAPC-4 cells and black symbols PC-3 cells. *denotes significance from vehicle (V) controls at P < 0.05, and shaded horizontal bars represent the response to V ± SEM. In this and other graphs, where error bars are not visible, they were within the size of the symbol. Dietary or environmentally relevant concentration ranges are shown by the solid horizontal bars below the graphs for each XE. The insets show cell numbers after 3 d of E2 treatment, for comparison (and see Citation69).

Figure 1. Cell number after 3 d of XE treatment. LAPC-4 and PC-3 prostate cancer cells were treated with XEs and viable cells were measured by the MTT assay. In all figures throughout the manuscript white symbols denote LAPC-4 cells and black symbols PC-3 cells. *denotes significance from vehicle (V) controls at P < 0.05, and shaded horizontal bars represent the response to V ± SEM. In this and other graphs, where error bars are not visible, they were within the size of the symbol. Dietary or environmentally relevant concentration ranges are shown by the solid horizontal bars below the graphs for each XE. The insets show cell numbers after 3 d of E2 treatment, for comparison (and see Citation69).

Subcellular location and levels of ERs

We next asked which types of ERs were present in LAPC-4 and PC-3 cell lines that could mediate these changes in viable cell number. We had previously noted a variable dependence of rapid responses on all 3 ER subtypes (determined by using selective antagonists) in our studies on E2 vs. DES treatment of these cells.Citation69 Plasma membrane versions of estrogen receptors (mERs) are thought to mediate rapid signaling involved in cell number changes in other cell types [reviewed inCitation64,80,81], so we examined the subcellular location of these receptors here. Using our plate immunoassay ± cell permeabilization with detergent (), we observed that late-stage tumor cells (panel B) had much lower expression of ERs than did early-stage cells (panel A; note the ∼fold6- vertical scale difference between panels A and B), a frequent finding among steroid receptors in endocrine cancers of multiple types,Citation82-84 although this evaluation for mERs in prostate tumor cells is novel. We also saw that membrane receptor populations were much lower than total (and thus intracellular) receptor forms in early-stage cells; the levels of mERs α and β were about 20% and 24% of their total receptor populations, respectively, as we have seen previously for the proportion of membrane versions of these receptors in other tumor cell types.Citation85-87 Although much lower, we detected significant levels of all 3 ER types in PC-3 cells. In these late-stage tumor cells the membrane receptor population was a much larger percentage of the total receptor numbers, perhaps in keeping with their more undifferentiated state, as we have seen with membrane glucocorticoid receptors in human lymphoma cells compared to normal circulating lymphocytes.Citation88 ERβ predominated in LAPC-4 cells (and to a lesser extent in PC-3 cells), as expected based on the literature regarding the dominance of this receptor type in normal prostate tissues and the early-stage tumors that arise from them.Citation89,90 However, there were also significant levels of ERα and GPR30, suggesting that they might also play a role in mediating estrogenic mechanisms. Interestingly, we found that the sizable amount of GPR30 was largely intracellular in LAPC-4 cells. GPR30 has been identified in other prostate cancer studies, but the subcellular location was not elucidated.Citation91-93 The subcellular location of GPR30 in other tissues and their cancers has been a point of contention; different groups have demonstrated this receptor form as either primarily in the plasma membrane or in the endoplasmic reticulum.Citation94,95

Figure 2. ER subtype (α, β, and GPR30) levels (total vs. membrane) in LAPC-4 and PC-3 prostate cancer cells. The negative control samples used no primary antibody (Ab) for any of the ER subtypes, as indicated by the first bar and the shaded bar extending horizontally across the graph (average ±SEM ). *denotes significance from controls at P < 0.05.

Figure 2. ER subtype (α, β, and GPR30) levels (total vs. membrane) in LAPC-4 and PC-3 prostate cancer cells. The negative control samples used no primary antibody (Ab) for any of the ER subtypes, as indicated by the first bar and the shaded bar extending horizontally across the graph (average ±SEM ). *denotes significance from controls at P < 0.05.

Phospho-ERK

Our next goal was to identify pathways and mechanisms responsible for any changes in numbers of viable cells, and ERK phosphorylation is one mechanism that has traditionally been associated with cell proliferation.Citation70 We selected an effective and environment- or diet-relevant concentration for each XE studied (10−9M BPA, 10−7M coumestrol, 10−7M genistein, and 10−8M resveratrol; see for relevant ranges), and measured their ability to elicit ERK phosphorylation in both cell lines over 60 min (). We observed activation for all compounds except genistein, but found that a sustained (60 min) pERK response did not predict a XE's positive influence on cell number, as has been a long-held association.Citation96,97 The most striking result was the difference between cell line-specific responses after resveratrol treatment, which caused a strong ERK deactivation (40%) in LAPC-4 cells, while it had slightly increased the number of viable cells (). In PC-3 cells resveratrol rapidly activated and sustained pERK (at 60 min), and caused modest cell proliferation. BPA and coumestrol rapidly stimulated ERK phosphorylation in both cell lines (), all with sustained levels at 60 min, but had no proliferation effects in late-stage cells. Genistein rapidly though modestly deactivated ERK in both cell lines, but substantially increased viable PC-3 cell numbers. Only the activation of ERK by coumestrol in both cell types correlated with its ability to cause these cells to proliferate. Therefore, these XEs elicited unique patterns of ERK activation/deactivation, which could contribute to cell survival, cell death, or proliferation,Citation70,96,98 but clearly more mechanisms needed to be considered.

Figure 3. Phospho-ERK (pERK) levels in LAPC-4 and PC-3 cells after XE treatments. LAPC-4 and PC-3 cells were treated with 10–9M BPA, 10−7M coumestrol, 10−7M genistein, and 10−8M resveratrol. pERK was measured up to 60 min via the plate immunoassay. *denotes significance from vehicle (shown at time 0) controls at P < 0.05, and horizontal shaded bars represent the response to vehicle ± SEM.

Figure 3. Phospho-ERK (pERK) levels in LAPC-4 and PC-3 cells after XE treatments. LAPC-4 and PC-3 cells were treated with 10–9M BPA, 10−7M coumestrol, 10−7M genistein, and 10−8M resveratrol. pERK was measured up to 60 min via the plate immunoassay. *denotes significance from vehicle (shown at time 0) controls at P < 0.05, and horizontal shaded bars represent the response to vehicle ± SEM.

ROS generation

Others have recently associated sustained ERK activations with ROS generation leading to cell killing,Citation70 and we recently extended this to the ability of a physiological estrogen (E2) to induce cell death in early-stage prostate cancer cells.Citation69 Therefore, we examined if XEs (at optimal concentrations for such ERK responses) could be linked to any ROS elevations (measured at the peak time of 15 min, time course not shown). The positive controls for ROS generation, including both H2O2 and E2, caused robust ROS generation (), and as we saw previously, cell death.Citation69 Most XE treatments generated significant ROS levels regardless of whether they had caused sustained ERK activation (), though BPA and genistein did so only in one cell line each. These ROS increases were all modest compared to those caused by E2. Therefore, ROS elevation due to sustained ERK activation was not considered to be a primary mechanistic determinant of viable cell number in these studies. Perhaps higher levels of ROS need to be generated to kill this cell type.

Figure 4. ROS levels after treatment with 10−10M E2, 10−6MH2O2, 10−9MBPA, 10−7M coumestrol, 10−7M genistein, and 10−8M resveratrol, ± ER subtype-selective antagonists. Antagonists (Antag) were 10−7M MMP for ERα; 10−6M PHTPP for ERβ; and 10−6M G15 for GPR30. ROS levels were measured after 15 min of each XE treatment (the optimal response time). *denotes significance from vehicle (V) controls at P < 0.05, while # denotes significance from paired XE treatment values (P < 0.05). ERα inhibition was significantly different vs. resveratrol alone in PC-3 cells ($) at P < 0.09. The shaded horizontal bars represent the response to vehicle (V) ± SEM.

Figure 4. ROS levels after treatment with 10−10M E2, 10−6MH2O2, 10−9MBPA, 10−7M coumestrol, 10−7M genistein, and 10−8M resveratrol, ± ER subtype-selective antagonists. Antagonists (Antag) were 10−7M MMP for ERα; 10−6M PHTPP for ERβ; and 10−6M G15 for GPR30. ROS levels were measured after 15 min of each XE treatment (the optimal response time). *denotes significance from vehicle (V) controls at P < 0.05, while # denotes significance from paired XE treatment values (P < 0.05). ERα inhibition was significantly different vs. resveratrol alone in PC-3 cells ($) at P < 0.09. The shaded horizontal bars represent the response to vehicle (V) ± SEM.

Therefore, these MAPK, and potentially linked ROS responses, did not individually predict cell proliferation vs. cell killing effects. Possibly a more traditional route of ROS generation not involving ERKs was involved in these cells. Estrogens have been shown to damage DNA,Citation99 which can also cause ROS generation.Citation100,101 The ability of these XEs to induce ROS was different from that of E2,Citation69 further highlighting the imperfect mimicry of physiological estrogens by XEs. Overall, the pERK and ROS responses to XE treatments in both cell lines do not appear to be lone driving mechanisms that elicit changes in cell numbers. Therefore, we have to consider the combined contribution of these responses to an overall balance of competing mechanisms (see below).

We observed previously that E2 required different ER subtypes to elicit ROS responses in LAPC-4 vs. PC-3 cells.Citation69 Here XEs also demonstrated unique ER-use signatures for this response (). In LAPC-4 (early-stage) cells, BPA caused these modest ROS increases independent of any known ERs, and genistein did not raise ROS levels (). The increases due to coumestrol required ERα and GPR30, while resveratrol required only ERα. A somewhat different profile was evident in PC-3 (late-stage) cells, where coumestrol and resveratrol both increased ROS via ERβ, while genistein stimulation of ROS did not require any known ERs, and BPA did not cause a response (). Clearly, the regulation of this pathway via ERs became quite different as cell types progressed to a less differentiated state with far lower receptor numbers (). The involvement of more than one ER subtype in some XE-generated responses also suggests the participation of multiple pathways.

Total and phosphorylated cyclin D1

Control of cyclin D1 levelsCitation102,103 is a mechanism we previously identified as being a significant contributor to E2- or DES-evoked declines in prostate cancer cell survival.Citation69 Both of these estrogens caused rapid cyclin D1 phospho-activation leading to swift degradation of this cell-cycle protein in early-stage cells, while this mechanism only operated for E2 in late stage cells. Lower cyclin D1 levels was reflected also in lower cell numbers. Similar responses to single effective concentrations of XEs (changes in phosphorylation of cyclin D1; changes in total cyclin levels) are shown in . Of the 4 XEs studied here, the 3 phytoestrogens (coumestrol, genistein, and resveratrol) all affected cyclin D1 phosphorylation levels, though in distinctly different directions, and differently for early- vs. late-stage cells (). In each case where cyclin D1 was phosphorylated, the corresponding expected rapid decline in total cyclin D1 levels occurred (panels C and D). Interestingly, we observed significant declines in total cyclin D1 as early as 5 min after genistein or resveratrol treatment (∼10%), but the largest decreases for most compounds were seen after 4 h. Coumestrol caused cyclin dephosphorylation, resulting in cyclin D1 level increases, correlating very well with its ability to increase cell numbers. Resveratrol signaling significantly phosphorylated cyclin D1 in both cell lines, driving total cyclin D1 levels down, yet while eliciting very small increases in cell proliferation, a less perfect correlation. Genistein was the only compound that caused opposing effects on these mechanisms in the 2 cell lines. In LAPC-4 cells, it increased phosphorylated cyclin D1, causing its degradation, but that did not correlate with measured changes in viable cell numbers. However, in PC-3 cells, genistein depressed cyclin phosphorylation, allowing increases in cyclin levels and correlating with a strikingly robust cell proliferative response. BPA did not affect cyclin D1 phosphorylation in either cell line, nor did it change levels of total cyclin D1, in keeping with its minimal effects on cell numbers. While these correlations generally go in the expected direction, they do not entirely predict the degree of the functional (cell number-changing) responses. Therefore, we ultimately considered all of these mechanisms together (see summation in Fig. 6), to see if other mechanisms in some cases modified this dominant response to cyclin D1 changes (see Conclusions). Others have also shown that phytoestrogens can affect other cell-cycle protein levels,Citation26,27,31,32,104 which in turn affected the number of cells.

Figure 5. Cyclin D1 phosphorylation and degradation by XEs, and inhibition by ER-selective antagonists. Cyclin phosphorylation was measured at 1–60 min, and total cyclin D1 levels over 16 h of XE treatment. For 5A and 5B, LAPC-4 and PC-3 cells were pretreated with antagonists (Antag) for each of the 3 ER subtypes: α (MPP), β (PHTPP), and GPR30 (G15,) and then treated with 10−9M BPA, 10−7M coumestrol, 10−7M genistein, 10−8M resveratrol, 10−10M E2 or 10−6M DES. Shaded horizontal bars represent V ± SEM. * denotes significance compared to vehicle (V) at P < 0.05. # denotes significance from paired XE treatment responses at P < 0.05. For 5C and 5D, LAPC-4 and PC-3 cells were treated with each XE for the times indicated and total cyclin D1 levels were measured with a plate immunoassay.

Figure 5. Cyclin D1 phosphorylation and degradation by XEs, and inhibition by ER-selective antagonists. Cyclin phosphorylation was measured at 1–60 min, and total cyclin D1 levels over 16 h of XE treatment. For 5A and 5B, LAPC-4 and PC-3 cells were pretreated with antagonists (Antag) for each of the 3 ER subtypes: α (MPP), β (PHTPP), and GPR30 (G15,) and then treated with 10−9M BPA, 10−7M coumestrol, 10−7M genistein, 10−8M resveratrol, 10−10M E2 or 10−6M DES. Shaded horizontal bars represent V ± SEM. * denotes significance compared to vehicle (V) at P < 0.05. # denotes significance from paired XE treatment responses at P < 0.05. For 5C and 5D, LAPC-4 and PC-3 cells were treated with each XE for the times indicated and total cyclin D1 levels were measured with a plate immunoassay.

Figure 6. Summary of XE responses for mechanisms that affect the number of viable LAPC-4 vs. PC-3 cells. Estradiol is shown for comparison, summarizing the data from our previous publication Citation69. Mechanisms in red text contribute to decreases in viable cell numbers, while mechanisms in green text increase the number of viable cells. Gray text indicates mechanisms that did not make any contribution to changes in cell numbers. These mechanistic contributions are summed in the red and green numbers in the upper right-hand corner of each box. Δ = change

Figure 6. Summary of XE responses for mechanisms that affect the number of viable LAPC-4 vs. PC-3 cells. Estradiol is shown for comparison, summarizing the data from our previous publication Citation69. Mechanisms in red text contribute to decreases in viable cell numbers, while mechanisms in green text increase the number of viable cells. Gray text indicates mechanisms that did not make any contribution to changes in cell numbers. These mechanistic contributions are summed in the red and green numbers in the upper right-hand corner of each box. Δ = change

Other signaling pathways that we have not examined here may also contribute to the net change in cyclin D1 phosphorylation and consequent decline in total protein levels. Glycogen synthase kinase 3β, which is regulated by the phosphoinositide 3-kinase/protein kinase B pathway (PI3K/Akt), has also been shown to phosphorylate cyclin D1 on Thr286, as well as regulate the protein's subcellular location in mouse fibroblasts.Citation105 However, the exact role of GSK-3β/PI3K/Akt in driving phosphorylation of cyclin D1 has been debated, as Guo et al., found that the activity of those pathways did not change (during the relevant S-phase), nor decrease cyclin D1 protein levels in mouse or human fibroblasts.Citation106 In addition, inhibition of GSK-3β in MCF-7 breast cancer cells did not completely disrupt cyclin D1 protein degradation.Citation107 Other pathways that can contribute to cyclin D1 degradation include p38, which we previously studied for activation by DES and E2 Citation69 and did not include here because it was activated for all cell types and treatments (and therefore may be permissive, but not directly causative). Cyclin D1 phosphorylation through p-p38 seems to be especially prevalent in response to the damage to DNA caused by environmental agents which require rapid cellular responses to prevent propagation of genomic mistakes.Citation108,109 The Mirk/Dyrk1b kinase, active during G0/G1, has also been shown to regulate cyclin D1 protein levels through phosphorylation at Thr288.Citation110 Therefore, multiple pathways can cause phosphorylation of cyclin D1, but they may influence degradation of cyclin D1 to varying degrees, and may be tissue-selective.

We next examined which ER subtypes (α, β, or GPR30) might be involved in the cyclin D1 phosphorylations, again using selective antagonists for each receptor subtype. For genistein, either the ERβ or GPR30 antagonist reversed the cyclin D1 phosphorylation in LAPC-4 cells, but only the ERβ antagonist decreased p-cyclin D1 levels in PC-3 cells. Coumestrol apparently did not utilize any of the known ER subtypes in LAPC-4 cells to decrease p-cyclin D1 levels, but ERα was required in PC-3 cells. Coumestrol's lack of dependence on any known receptor subtype in LAPC-4 cells is surprising, given the plentiful expression of all of these receptors in that cell line. It is possible that the low p-cyclin levels and thus larger errors in the measurement, caused by coumestrol treatment made antagonist reversals difficult to detect. Resveratrol's induction of p-cyclin D1 levels in both cell lines showed a dependence on ERs α and GPR30 in LAPC-4 cells, and on ERα in PC-3 cells. Therefore, each XE showed a dependence on a different ER subtype or subtype combination in the 2 cell lines. These dependencies are consistent with what has been previously shown about receptor subtype binding preferences for these XE compounds.Citation6 For example, resveratrol has a higher binding preference for ERα than for ERβ,Citation2 while coumestrol and genistein are strong ERβ agonists,Citation3 but are still capable of binding to ERα.Citation6,111,112 The predominance of ERβ in these prostate cell lines may influence their responses to these XEs that affect cell number. Mostly genomic pathways have been examined in the past, such as the ability to activate ER reporter constructs, with differences between cell types for different XEs.Citation113-115 Few comparisons for nongenomic responses are available, though we previously observed different MAPK activation patterns and mostly positive proliferative responses to E2 and various XEs in GH3/B6/F10 pituitary cellsCitation116 that have high mERα and low mERβ levels.

Also consistent with our results are known XE effects that do not involve these ERs. An example is the well-known direct inhibitory effect of genistein on tyrosine kinases.Citation117-119 In other instances, small lipophilic compounds like these can intercalate into cell membranes and as “border lipids” influence the actions of proteins embedded in them. Lipophilic estrogens can change membrane fluidity,Citation120,121 especially when they are present at relatively high concentrations (as is true for most effective phytoestrogen concentrations resulting from dietary exposures). Because changes in cell numbers are best observed after 3 d, the nuclear-localized receptors forms involved in slower transcriptional regulationCitation122,123 may also be relevant to these effects, which we did not examine in our studies of these more novel rapid actions.

Another possible contributor to tumor cell behavior in prostate cell lines is the tumor-suppressor p53.Citation124 It is mutated in LAPC-4 cells, and not present in PC-3 cells, and therefore unlikely to drive estrogen-mediated prostate tumor viability in our present studies. We also chose cell models for our studies that do not present the added complication of mutant ARs (such as in LnCaP cellsCitation125) to which estrogens can more readily bind and elicit effects, especially at high concentrations. LAPC-4 cells have wild-type ARs and PC-3 cells do not express ARs.Citation124

Conclusions

The estrogen-induced mechanism that dominated our effective predictions of cell growth behavior in these studies was the rapid phosphorylation of cyclin D1, followed shortly thereafter by its degradation. This mechanism largely predicted the primary response to each XE in terms of cell number changes (except in the case of resveratrol). However, no single mechanism entirely predicted the degree of these XE-induced changes, so we also examined the balance of the effects of other pathways (summarized in Fig 6). In this figure, each of the mechanisms examined in these studies for each cell type due to each XE is summarized in colored text: red denotes actions causing decreases in viable cells, while green represents mechanisms that increase cell numbers, and gray indicates no effect. These mechanistic contributions are summed in the red and green numbers in the upper right-hand corner of each box. The modest effects of these XEs are in contrast to the strong cell growth inhibitory/cell killing effects we saw previously with E2; these previous conclusions Citation69 are shown in the first column of the table for the sake of comparison. E2 engaged a total of 7 signaling responses in LAPC-4 cells and PC-3 cells – though in this table we list only the 4 that were examined here that gave the best predictions of therapeutic responses (decreases in cell numbers) to compare with XEs. Coumestrol is a robust stimulator of cell growth in both early- and late-stage prostate cancer cells, matching its strong positive effects on cyclin D1 status and levels. The ability to activate sustained ERK, and through it to increase ROS levels, did not decrease the cyclin D1-driven outcome. Genistein's different effects on early- vs. late-stage cells could also be largely explained by the cyclin D1 changes. Resveratrol had a very small effect in both cell lines, though the altered cyclin D1 levels and the ROS generation should have predicted a large decrease in cell viability, which was not observed. BPA showed the smallest changes in these responses that we examined, corresponding to only minimal growth stimulation, in only LAPC-4 cells.

Once again, mechanistic responses (phosphorylations, cyclin level changes, and ROS generation) to these varied estrogens were documented to be very rapid, supporting the notion that important tumor-altering effects can occur, or at least be initiated, via nongenomic signaling mechanisms. This was supported by our demonstration that membrane versions of these receptors are present in these tumor cells. Our present and recent Citation20,37,38,40,54,55,64,67,126 studies continue to support conclusions about the ability of XEs to imperfectly mimic physiological and pharmaceutical estrogens, as well as their unique patterns of mechanism engagement and ER requirements.Citation20,37,38,54,55,64,67,87,126,127 Because of these differences, it is important to consider the potential effects of each XE individually at its typical culturally- or environmentally-relevant levels, to determine what exposure advice these mechanistic studies may point to. Given that some of these compounds can have profound stimulatory effects on prostate cancer cell numbers (particularly the soy-related phytoestrogens coumestrol and genistein, and especially in late-stage tumor cells), it may be prudent to advise such patients against consuming foods that contain these phytoestrogens. On a lower priority level, resveratrol and BPA exposures may warrant similar warnings (Fig 6). Because BPA and genistein had different effects on the number of viable cancer cells, depending on prostate cancer stage, patients having recurring or long-term tumors may need different exposure advice. Because cell growth-promoting mechanisms receive stimulation via different ER subtypes depending upon the compound, it may be prudent to recommend blocking of these effects via all of these receptors. Alternatively, testing of a patient's individual tumor receptor profiles may allow for tailoring of therapies with antagonists for individual receptor subtypes.

Our initial hypothesis was that some of these alternative dietary estrogens might fulfill the hoped for anti-tumor signaling and cell growth effects. It appears that this is not the case, and the best estrogen to mediate tumor cell killing effects is E2, profiled in more detail for these mechanisms (and others) in our previous report.Citation69 Taken together, these findings should have profound implications for dietary recommendations for prostate cancer patients, as well for as the development of ER-specific treatments to shrink tumors or slow tumor progression.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Huang CK, Luo J, Lee SO, Chang C. Androgen receptor differential roles in stem/progenitor cells including prostate, embryonic, stromal, and hematopoietic lineages. Stem Cells 2014; 32(9):2299-308; PMID:24740898
  • Bowers JL, Tyulmenkov VV, Jernigan SC, Klinge CM. Resveratrol acts as a mixed agonist/antagonist for estrogen receptors alpha and beta. Endocrinology 2000; 141:3657-67; PMID:11014220
  • McCarty MF. Isoflavones made simple – Genistein's agonist activity for the beta-type estrogen receptor mediates their health benefits. Medi Hypotheses 2006; 66:1093-114; PMID:16513288; http://dx.doi.org/10.1016/j.mehy.2004.11.046
  • Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 1997; 138:863-70; PMID:9048584
  • Chandsawangbhuwana C, Baker ME. 3D models of human ERα and ERβ complexed with coumestrol. Steroids 2014; 80:37-43; PMID:24315835; http://dx.doi.org/10.1016/j.steroids.2013.11.019
  • Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van der Burg B, Gustafsson JA. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 1998; 139:4252-63; PMID:9751507
  • Prins GS, Hu WY, Shi GB, Hu DP, Majumdar S, Li G, Huang K, Nelles JL, Ho SM, Walker CL, et al. Bisphenol A promotes human prostate stem-progenitor cell self-renewal and increases in vivo carcinogenesis in human prostate epithelium. Endocrinology 2014; 155:805-17; PMID:24424067; http://dx.doi.org/10.1210/en.2013-1955
  • Richter C, Birnbaum L, Farabollini F, Newbold R, Rubin B, Talsness C, Vandenbergh J, Walser-Kuntz D, vom Saal F. In vivo effects of bisphenol A in laboratory rodent studies. Reprod Toxicol 2007; 24:199-224; PMID:17683900; http://dx.doi.org/10.1016/j.reprotox.2007.06.004
  • Vandenberg L, Colborn T, Hayes T, Heindel J, Jacobs D, Lee D, Shioda T, Soto A, Vom Saal F, Welshons W, et al. Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocr Rev 2012; 33:378-455; PMID:22419778; http://dx.doi.org/10.1210/er.2011-1050
  • Bharathi SP, Raj HM, Jain S, Banerjee BD, Ahmed T, Arora VK. Role of pesticides in the induction of tumor angiogenesis. Anticancer Res 2013; 33:231-40; PMID:23267150
  • De Flora S, Micale RT, La Maestra S, Izzotti A, D'Agostini F, Camoirano A, Davoli SA, Troglio MG, Rizzi F, Davalli P, et al. Upregulation of clusterin in prostate and DNA damage in spermatozoa from bisphenol A-treated rats and formation of DNA adducts in cultured human prostatic cells. Toxicol Sci 2011; 122:45-51; PMID:21536718; http://dx.doi.org/10.1093/toxsci/kfr096
  • Weinhouse C, Anderson OS, Bergin IL, Vandenbergh DJ, Gyekis JP, Dingman MA, Yang J, Dolinoy DC. Dose-dependent incidence of hepatic tumors in adult mice following perinatal exposure to bisphenol A. Environ Health Perspect 2014; 122:485-91; PMID:24487385
  • Giannandrea F, Paoli D, Figà-Talamanca I, Lombardo F, Lenzi A, Gandini L. Effect of endogenous and exogenous hormones on testicular cancer: the epidemiological evidence. Int J Dev Biol 2013; 57:255-63; PMID:23784836; http://dx.doi.org/10.1387/ijdb.130015fg
  • Cotter KA, Yershov A, Novillo A, Callard GV. Multiple structurally distinct ERα mRNA variants in zebrafish are differentially expressed by tissue type, stage of development and estrogen exposure. Gen Comp Endocrinol 2013; 194:217-29; PMID:24090614; http://dx.doi.org/10.1016/j.ygcen.2013.09.014
  • Wong RLY, Walker CL. Molecular Pathways: Environmental estrogens activate nongenomic signaling to developmentally reprogram the epigenome. Clin Cancer Res 2013; 19:3732-7; PMID:23549878; http://dx.doi.org/10.1158/1078-0432.CCR-13-0021
  • Vilahur N, Molina-Molina JM, Bustamante M, Murcia M, Arrebola JP, Ballester F, Mendez MA, Garcia-Esteban R, Guxens M, Marina LS, et al. Male specific association between xenoestrogen levels in placenta and birthweight. Environ Int 2013; 51:174-81; PMID:23262415; http://dx.doi.org/10.1016/j.envint.2012.10.004
  • Ibarluzea Jm J, Fernandez MF, Santa-Marina L, Olea-Serrano MF, Rivas AM, Aurrekoetxea JJ, Exposito J, Lorenzo M, Torne P, Villalobos M, et al. Breast cancer risk and the combined effect of environmental estrogens. Cancer Causes Control 2004; 15:591-600; PMID:15280638; http://dx.doi.org/10.1023/B:CACO.0000036167.51236.86
  • Wetherill YB, Petre CE, Monk KR, Puga A, Knudsen KE. The xenoestrogen bisphenol A induces inappropriate androgen receptor activation and mitogenesis in prostatic adenocarcinoma cells. Mol Cancer Ther 2002; 1:515-24; PMID:12479269
  • Rice S, Whitehead SA. Phytoestrogens and breast cancer–promoters or protectors? Endocr Relat Cancer 2006; 13:995-1015; PMID:17158751; http://dx.doi.org/10.1677/erc.1.01159
  • Jeng Y-J, Watson C. Proliferative and anti-proliferative effects of dietary levels of phytoestrogens in rat pituitary GH3/B6/F10 cells - the involvement of rapidly activated kinases and caspases. BMC Cancer 2009; 9:1-17; PMID:19118499; http://dx.doi.org/10.1186/1471-2407-9-334
  • Zhang Y, Li Q, Zhou D, Chen H. Genistein, a soya isoflavone, prevents azoxymethane-induced up-regulation of WNT/beta-catenin signalling and reduces colon pre-neoplasia in rats. Br J Nutr 2013; 109:33-42; PMID:22716201; http://dx.doi.org/10.1017/S0007114512000876
  • Schleipen B, Hertrampf T, Fritzemeier KH, Kluxen FM, Lorenz A, Molzberger A, Velders M, Diel P. ERbeta-specific agonists and genistein inhibit proliferation and induce apoptosis in the large and small intestine. Carcinogenesis 2011; 32:1675-83; PMID:21856997; http://dx.doi.org/10.1093/carcin/bgr188
  • Chen J, Zeng J, Xin M, Huang W, Chen X. Formononetin induces cell cycle arrest of human breast cancer cells via IGF1/PI3K/Akt pathways in vitro and in vivo. Horm Metab Res 2011; 43:681-6; PMID:21932171; http://dx.doi.org/10.1055/s-0031-1275703
  • Shanmugam MK, Kannaiyan R, Sethi G. Targeting cell signaling and apoptotic pathways by dietary agents: role in the prevention and treatment of cancer. Nutr Cancer 2011; 63:161-73; PMID:21294053; http://dx.doi.org/10.1080/01635581.2011.523502
  • Alves RC, Almeida IM, Casal S, Oliveira MB. Isoflavones in coffee: influence of species, roast degree, and brewing method. J Agric Food Chem 2010; 58:3002-7; PMID:20131840; http://dx.doi.org/10.1021/jf9039205
  • Adjakly M, Ngollo M, Boiteux JP, Bignon YJ, Guy L, Bernard-Gallon D. Genistein and daidzein: different molecular effects on prostate cancer. Anticancer Res 2013; 33:39-44; PMID:23267126
  • Shenouda NS, Zhou C, Browning JD, Ansell PJ, Sakla MS, Lubahn DB, Macdonald RS. Phytoestrogens in common herbs regulate prostate cancer cell growth in vitro. Nutr Cancer 2004; 49:200-8; PMID:15489213; http://dx.doi.org/10.1207/s15327914nc4902_12
  • Lee YH, Yuk HJ, Park KH, Bae YS. Coumestrol induces senescence through protein kinase CKII inhibition-mediated reactive oxygen species production in human breast cancer and colon cancer cells. Food Chem 2013; 141:381-8; PMID:23768371; http://dx.doi.org/10.1016/j.foodchem.2013.03.053
  • Bommareddy A, Eggleston W, Prelewicz S, Antal A, Witczak Z, McCune DF, Vanwert AL. Chemoprevention of prostate cancer by major dietary phytochemicals. Anticancer Res 2013; 33:4163-74; PMID:24122979
  • Chang HT, Chou CT, Chen IL, Liang WZ, Kuo DH, Huang JK, Shieh P, Jan CR. Mechanisms of resveratrol-induced changes in [Ca(2+)]i and cell viability in PC3 human prostate cancer cells. J Recept Signal Transduct Res 2013; 33:298-303; PMID:23898810; http://dx.doi.org/10.3109/10799893.2013.822886
  • Wang TT, Schoene NW, Kim YS, Mizuno CS, Rimando AM. Differential effects of resveratrol and its naturally occurring methylether analogs on cell cycle and apoptosis in human androgen-responsive LNCaP cancer cells. Mol Nutr Food Res 2010; 54:335-44; PMID:20077416; http://dx.doi.org/10.1002/mnfr.200900143
  • Benitez DA, Pozo-Guisado E, Alvarez-Barrientos A, Fernandez-Salguero PM, Castellon EA. Mechanisms involved in resveratrol-induced apoptosis and cell cycle arrest in prostate cancer-derived cell lines. J Androl 2007; 28:282-93; PMID:17050787; http://dx.doi.org/10.2164/jandrol.106.000968
  • Scarlatti F, Sala G, Ricci C, Maioli C, Milani F, Minella M, Botturi M, Ghidoni R. Resveratrol sensitization of DU145 prostate cancer cells to ionizing radiation is associated to ceramide increase. Cancer Lett 2007; 253:124-30; PMID:17321671; http://dx.doi.org/10.1016/j.canlet.2007.01.014
  • Vandenberg L, Chahoud I, Heindel J, Padmanabhan V, Paumgartten F, Schoenfelder G. Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A. Environ Health Perspect 2010; 118:1055-70; PMID:20338858; http://dx.doi.org/10.1289/ehp.0901716
  • Vandenberg LN, Hauser R, Marcus M, Olea N, Welshons WV. Human exposure to bisphenol A (BPA). Reproduct Toxicol 2007; 24:139-77; PMID:17825522; http://dx.doi.org/10.1016/j.reprotox.2007.07.010
  • Ge LC, Chen ZJ, Liu HY, Zhang KS, Liu H, Huang HB, Zhang G, Wong CK, Giesy JP, Du J, et al. Involvement of activating ERK1/2 through G protein coupled receptor 30 and estrogen receptor alpha/beta in low doses of bisphenol A promoting growth of Sertoli TM4 cells. Toxicol Lett 2014; 226:81-9; PMID:24495410; http://dx.doi.org/10.1016/j.toxlet.2014.01.035
  • Vinas R, Watson CS. Mixtures of xenoestrogens disrupt estradiol-induced non-genomic signaling and downstream functions in pituitary cells. Environ Health 2013; 12:26; PMID:23530988; http://dx.doi.org/10.1186/1476-069X-12-26
  • Bulayeva NN, Watson CS. Xenoestrogen-induced ERK-1 and ERK-2 activation via multiple membrane-initiated signaling pathways. Environ Health Perspect 2004; 112:1481-7; PMID:15531431; http://dx.doi.org/10.1289/ehp.7175
  • Wang W, Wang J, Wang Q, Wu W, Huan F, Xiao H. Bisphenol A modulates calcium currents and intracellular calcium concentration in rat dorsal root ganglion neurons. J Membr Biol 2013; 246:391-7; PMID:23575985; http://dx.doi.org/10.1007/s00232-013-9545-8
  • Wozniak AL, Bulayeva NN, Watson CS. Xenoestrogens at picomolar to nanomolar concentrations trigger membrane estrogen receptor-alpha-mediated Ca2+ fluxes and prolactin release in GH3/B6 pituitary tumor cells. Environ Health Perspect 2005; 113:431-9; PMID:15811834; http://dx.doi.org/10.1289/ehp.7505
  • Derouiche S, Warnier M, Mariot P, Gosset P, Mauroy B, Bonnal JL, Slomianny C, Delcourt P, Prevarskaya N, Roudbaraki M. Bisphenol A stimulates human prostate cancer cell migration remodelling of calcium signalling. Springerplus 2013; 2:54; PMID:23450760; http://dx.doi.org/10.1186/2193-1801-2-54
  • Midoro-Horiuti T, Tiwari R, Watson CS, Goldblum RM. Maternal bisphenol a exposure promotes the development of experimental asthma in mouse pups. Environ Health Perspect 2010; 118:273-7; PMID:20123615; http://dx.doi.org/10.1289/ehp.0901259
  • Lamartiniere CA. Protection against breast cancer with genistein: a component of soy. Am J Clin Nutr 2000; 71:1705S-7S; discussion 8S-9S; PMID:10837323
  • Magee PJ, Rowland IR. Phyto-oestrogens, their mechanism of action: current evidence for a role in breast and prostate cancer. Br J Nutr 2004; 91:513-31; PMID:15035679; http://dx.doi.org/10.1079/BJN20031075
  • Adlercreutz H. Phytoestrogens: epidemiology and a possible role in cancer protection. Environ Health Perspect 1995; 103(Suppl 7):103-12; PMID:8593855; http://dx.doi.org/10.1289/ehp.95103s7103
  • Abd Elmageed ZY, Moroz K, Srivastav SK, Fang Z, Crawford BE, Moparty K, Thomas R, Abdel-Mageed AB. High circulating estrogens and selective expression of ERbeta in prostate tumors of Americans: implications for racial disparity of prostate cancer. Carcinogenesis 2013; 34:2017-23; PMID:23658372; http://dx.doi.org/10.1093/carcin/bgt156
  • Kunisue T, Tanabe S, Isobe T, Aldous KM, Kannan K. Profiles of phytoestrogens in human urine from several Asian countries. J Agric Food Chem 2010; 58:9838-46; PMID:20707345; http://dx.doi.org/10.1021/jf102253j
  • Vergne S, Sauvant P, Lamothe V, Chantre P, Asselineau J, Perez P, Durand M, Moore N, Bennetau-Pelissero C. Influence of ethnic origin (Asian v. Caucasian) and background diet on the bioavailability of dietary isoflavones. Br J Nutr 2009; 102:1642-53; PMID:19622188; http://dx.doi.org/10.1017/S0007114509990833
  • Renouf M, Hendrich S. Bacteroides uniformis is a putative bacterial species associated with the degradation of the isoflavone genistein in human feces. J Nutr 2011; 141:1120-6; PMID:21525249; http://dx.doi.org/10.3945/jn.111.140988
  • Gaido KW, Leonard LS, Lovell S, Gould JC, Babai D, Portier CJ, McDonnell DP. Evaluation of chemicals with endocrine modulating activity in a yeast-based steroid hormone receptor gene transcription assay. Toxicol Appl Pharmacol 1997; 143:205-12; PMID:9073609; http://dx.doi.org/10.1006/taap.1996.8069
  • Singleton DW, Feng Y, Chen Y, Busch SJ, Lee AV, Puga A, Khan SA. Bisphenol-A and estradiol exert novel gene regulation in human MCF-7 derived breast cancer cells. Mol Cell Endocrinol 2004; 221:47-55; PMID:15223131; http://dx.doi.org/10.1016/j.mce.2004.04.010
  • Sheeler CQ, Dudley MW, Khan SA. Environmental estrogens induce transcriptionally active estrogen receptor dimers in yeast: activity potentiated by the coactivator RIP140. Environ Health Perspect 2000; 108:97-103; PMID:10656848; http://dx.doi.org/10.1289/ehp.0010897
  • Gutendorf B, Westendorf J. Comparison of an array of in vitro assays for the assessment of the estrogenic potential of natural and synthetic estrogens, phytoestrogens and xenoestrogens. Toxicology 2001; 166:79-89; PMID:11518614; http://dx.doi.org/10.1016/S0300-483X(01)00437-1
  • Kochukov M, Jeng Y-J, Watson C. Alkylphenol xenoestrogens with varying carbon chain lengths differentially and potently activate signaling and functional responses in GH3/B6/F10 somatomammotropes. Environ Health Perspect 2009; 117:723-30; PMID:19479013; http://dx.doi.org/10.1289/ehp.0800182
  • Vinas R, Watson C. Bisphenol s disrupts estradiol-induced nongenomic signaling in a rat pituitary cell line: effects on cell functions. Environ Health Perspect 2013; 121:352-8; PMID:23458715; http://dx.doi.org/10.1289/ehp.1205826
  • Watson CS, Alyea RA, Jeng YJ, Kochukov MY. Nongenomic actions of low concentration estrogens and xenoestrogens on multiple tissues. Mol Cell Endocrinol 2007; 274:1-7; PMID:17601655; http://dx.doi.org/10.1016/j.mce.2007.05.011
  • Zsarnovszky A, Le HH, Wang HS, Belcher SM. Ontogeny of rapid estrogen-mediated extracellular signal-regulated kinase signaling in the rat cerebellar cortex: potent nongenomic agonist and endocrine disrupting activity of the xenoestrogen bisphenol A. Endocrinology 2005; 146:5388-96; PMID:16123166; http://dx.doi.org/10.1210/en.2005-0565
  • Nadal A, Ropero AB, Laribi O, Maillet M, Fuentes E, Soria B. Nongenomic actions of estrogens and xenoestrogens by binding at a plasma membrane receptor unrelated to estrogen receptor alpha and estrogen receptor beta. Proc Natl Acad Sci U S A 2000; 97:11603-8; PMID:11027358; http://dx.doi.org/10.1073/pnas.97.21.11603
  • Sicotte NL, Liva SM, Klutch R, Pfeiffer P, Bouvier S, Odesa S, Wu TC, Voskuhl RR. Treatment of multiple sclerosis with the pregnancy hormone estriol. Ann Neurol 2002; 52:421-8; PMID:12325070; http://dx.doi.org/10.1002/ana.10301
  • Voskuhl R, Wang H, Jackson Wu T, Sicotte N, Bates A, Beaver G, Corboy J, Cross A, Dhib-Jalbut S, Ford C, et al. A combination trial of estriol plus glatimer acetate in relapsing-remitting multiple sclerosis. Abstract from the American Academy of Neurology 2014 Annual Meeting 2014.
  • Watson C, Jeng Y-J, Kochukov M. Nongenomic actions of estradiol compared with estrone and estriol in pituitary tumor cell signaling and proliferation. FASEB J 2008; 22:3328-36; PMID:18541692; http://dx.doi.org/10.1096/fj.08-107672
  • vom Saal FS, Welshons WV. Large effects from small exposures. II. The importance of positive controls in low-dose research on bisphenol A. Environ Res 2006; 100:50-76; PMID:16256977; http://dx.doi.org/10.1016/j.envres.2005.09.001
  • Zhong L, Xiang X, Lu W, Zhou P, Wang L. Interference of xenoestrogen o,p′-DDT on the action of endogenous estrogens at environmentally realistic concentrations. Bull Environ Contam Toxicol 2013; 90:591-5; PMID:23478948; http://dx.doi.org/10.1007/s00128-013-0976-9
  • Jeng Y-J, Kochukov M, Watson C. Membrane estrogen receptor-alpha-mediated nongenomic actions of phytoestrogens in GH3/B6/F10 pituitary tumor cells. J Mol Signal 2009; 4:1-11; PMID:19320971; http://dx.doi.org/10.1186/1750-2187-4-2
  • Watson CS, Bulayeva NN, Wozniak AL, Alyea RA. Xenoestrogens are potent activators of nongenomic estrogenic responses. Steroids 2007; 72:124-34; PMID:17174995; http://dx.doi.org/10.1016/j.steroids.2006.11.002
  • Watson CS, Hu G, Paulucci-Holthauzen AA. Rapid actions of xenoestrogens disrupt normal estrogenic signaling. Steroids 2013; PMID:24269739
  • Jeng Y, Kochukov M, Watson C. Combinations of physiologic estrogens with xenoestrogens alter calcium and kinase responses, prolactin release, and membrane estrogen receptor trafficking in rat pituitary cells. Environ Health 2010; 9:61; PMID:20950447; http://dx.doi.org/10.1186/1476-069X-9-61
  • Watson CS, Bulayeva NN, Wozniak AL, Finnerty CC. Signaling from the membrane via membrane estrogen receptor-α: Estrogens, xenoestrogens, and phytoestrogens. Steroids 2005; 70:364-71; PMID:15862819; http://dx.doi.org/10.1016/j.steroids.2005.03.002
  • Koong LY, Watson CS. Direct estradiol and diethylstilbestrol actions on early- versus late-stage prostate cancer cells http://dx.doi.org/10.1002/pros.22875. Prostate 2014; 74(16):1589-603; PMID: 25213831; http://dx.doi.org/10.1002/pros.22875
  • Cagnol S, Chambard JC. ERK and cell death: Mechanisms of ERK-induced cell death - apoptosis, autophagy and senescence. FEBS J 2010; 277:2-21; PMID:19843174; http://dx.doi.org/10.1111/j.1742-4658.2009.07366.x
  • Klein KA, Reiter RE, Redula J, Moradi H, Zhu XL, Brothman AR, Lamb DJ, Marcelli M, Belldegrun A, Witte ON, et al. Progression of metastatic human prostate cancer to androgen independence in immunodeficient SCID mice. Nat Med 1997; 3:402-8; PMID:9095173; http://dx.doi.org/10.1038/nm0497-402
  • Kaighn ME, Narayan KS, Ohnuki Y, Lechner JF, Jones LW. Establishment and characterization of a human prostatic carcinoma cell line (PC-3). Invest Urol 1979; 17:16-23; PMID:447482
  • Bulayeva NN, Gametchu B, Watson CS. Quantitative measurement of estrogen-induced ERK 1 and 2 activation via multiple membrane-initiated signaling pathways. Steroids 2004; 69:181-92; PMID:15072920; http://dx.doi.org/10.1016/j.steroids.2003.12.003
  • Viñas R, Goldblum RM, Watson CS. Rapid estrogenic signaling activities of the modified (chlorinated, sulfonated, and glucuronidated) endocrine disruptor bisphenol A. Endocrine Disruptors 2013; 1:0-9.
  • Gallagher RP, Kutynec CL. Diet, micronutrients and prostate cancer: a review of the evidence. Can J Urol 1997; 4:22-7; PMID:12735830
  • Hedelin M, Balter KA, Chang ET, Bellocco R, Klint A, Johansson JE, Wiklund F, Thellenberg-Karlsson C, Adami HO, Gronberg H. Dietary intake of phytoestrogens, estrogen receptor-beta polymorphisms and the risk of prostate cancer. Prostate 2006; 66:1512-20; PMID:16921512; http://dx.doi.org/10.1002/pros.20487
  • Nakamura H, Wang Y, Kurita T, Adomat H, Cunha GR. Genistein increases epidermal growth factor receptor signaling and promotes tumor progression in advanced human prostate cancer. PLoS One 2011; 6:e20034; PMID:21603581; http://dx.doi.org/10.1371/journal.pone.0020034
  • El Touny LH, Banerjee PP. Identification of a biphasic role for genistein in the regulation of prostate cancer growth and metastasis. Cancer Res 2009; 69:3695-703; PMID:19351854; http://dx.doi.org/10.1158/0008-5472.CAN-08-2958
  • Wang X, Clubbs EA, Bomser JA. Genistein modulates prostate epithelial cell proliferation via estrogen- and extracellular signal-regulated kinase-dependent pathways. J Nutr Biochem 2006; 17:204-10; PMID:16198100; http://dx.doi.org/10.1016/j.jnutbio.2005.07.005
  • Watson C, Gametchu B. Membrane-initiated steroid actions and the proteins that mediate them. Proc Soc Exp Biol Med 1999; 220:9-19; PMID:9893163; http://dx.doi.org/10.3181/00379727-220-44338
  • Zivadinovic D, Gametchu B, Watson CS. Membrane estrogen receptor-alpha levels in MCF-7 breast cancer cells predict cAMP and proliferation responses. Breast Cancer Res 2005; 7:R101-12; PMID:15642158; http://dx.doi.org/10.1186/bcr958
  • Hess KR, Pusztai L, Buzdar AU, Hortobagyi GN. Estrogen receptors and distinct patterns of breast cancer relapse. Breast Cancer Res Treat 2003; 78:105-18; PMID:12611463; http://dx.doi.org/10.1023/A:1022166517963
  • Bianchini G, Pusztai L, Karn T, Iwamoto T, Rody A, Kelly C, Muller V, Schmidt S, Qi Y, Holtrich U, et al. Proliferation and estrogen signaling can distinguish patients at risk for early versus late relapse among estrogen receptor positive breast cancers. Breast Cancer Res 2013; 15:R86; PMID:24060333
  • Signoretti S, Loda M. Estrogen receptor beta in prostate cancer: brake pedal or accelerator? Am J Pathol 2001; 159:13-6; PMID:11438447; http://dx.doi.org/10.1016/S0002-9440(10)61666-5
  • Campbell CH, Watson CS. A comparison of membrane vs. intracellular estrogen receptor-alpha in GH(3)/B6 pituitary tumor cells using a quantitative plate immunoassay. Steroids 2001; 66:727-36; PMID:11522334; http://dx.doi.org/10.1016/S0039-128X(01)00106-4
  • Watson CS, Campbell CH, Gametchu B. Membrane oestrogen receptors on rat pituitary tumour cells: immuno-identification and responses to oestradiol and xenoestrogens. Exp Physiol 1999; 84:1013-22; PMID:10564698; http://dx.doi.org/10.1111/j.1469-445X.1999.01903.x
  • Alyea R, Laurence S, Kim S, Katzenellenbogen B, Katzenellenbogen J, Watson C. The roles of membrane estrogen receptor subtypes in modulating dopamine transporters in PC-12 cells. J Neurochem 2008; 106:1525-33; PMID:18489713; http://dx.doi.org/10.1111/j.1471-4159.2008.05491.x
  • Gametchu B, Watson CS. Plasma membrane-associated glucocorticoid hormone receptor in human leukemic patients: clinical implications. In: Glucocorticoid Receptor Structure and Leukemic Cell Responses. B Gametchu, ed. Austin: R.G. Landes Company, 1995:163-76.
  • Hartman J, Ström A, Gustafsson J-Å. Current concepts and significance of estrogen receptor β in prostate cancer. Steroids 2012; 77:1262-6; PMID:22824289; http://dx.doi.org/10.1016/j.steroids.2012.07.002
  • Lai JS, Brown LG, True LD, Hawley SJ, Etzioni RB, Higano CS, Ho SM, Vessella RL, Corey E. Metastases of prostate cancer express estrogen receptor-beta. Urology 2004; 64:814-20; PMID:15491740; http://dx.doi.org/10.1016/j.urology.2004.05.036
  • Chan QK, Lam HM, Ng CF, Lee AY, Chan ES, Ng HK, Ho SM, Lau KM. Activation of GPR30 inhibits the growth of prostate cancer cells through sustained activation of Erk1/2, c-jun/c-fos-dependent upregulation of p21, and induction of G(2) cell-cycle arrest. Cell Death Differ 2010; 17:1511-23; PMID:20203690; http://dx.doi.org/10.1038/cdd.2010.20
  • Comeglio P, Morelli A, Cellai I, Vignozzi L, Sarchielli E, Filippi S, Maneschi E, Corcetto F, Corno C, Gacci M, et al. Opposite effects of tamoxifen on metabolic syndrome-induced bladder and prostate alterations: a role for GPR30/GPER? Prostate 2014; 74:10-28; PMID:24037776; http://dx.doi.org/10.1002/pros.22723
  • Zhang Z, Duan L, Du X, Ma H, Park I, Lee C, Zhang J, Shi J. The proliferative effect of estradiol on human prostate stromal cells is mediated through activation of ERK. Prostate 2008; 68:508-16; PMID:18213633; http://dx.doi.org/10.1002/pros.20722
  • Revankar CM, Mitchell HD, Field AS, Burai R, Corona C, Ramesh C, Sklar LA, Arterburn JB, Prossnitz ER. Synthetic estrogen derivatives demonstrate the functionality of intracellular GPR30. ACS Chem Biol 2007; 2:536-44; PMID:17655271; http://dx.doi.org/10.1021/cb700072n
  • Yu X, Filardo EJ, Shaikh ZA. The membrane estrogen receptor GPR30 mediates cadmium-induced proliferation of breast cancer cells. Toxicol Appl Pharmacol 2010; 245:83-90; PMID:20153348; http://dx.doi.org/10.1016/j.taap.2010.02.005
  • Deschenes-Simard X, Kottakis F, Meloche S, Ferbeyre G. ERKs in cancer: friends or foes? Cancer Res 2014; 74:412-9; PMID:24408923; http://dx.doi.org/10.1158/0008-5472.CAN-13-2381
  • Roskoski Jr R. ERK1/2 MAP kinases: Structure, function, and regulation. Pharmacol Res 2012; 66:105-43; PMID:22569528; http://dx.doi.org/10.1016/j.phrs.2012.04.005
  • Chang F, Steelman LS, Shelton JG, Lee JT, Navolanic PM, Blalock WL, Franklin R, McCubrey JA. Regulation of cell cycle progression and apoptosis by Ras/Raf/MEK/ERK pathway. Int J Oncol 2003; 22:469-80; PMID:12579299
  • Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D. Estrogens as endogenous genotoxic agents–DNA adducts and mutations. J Natl Cancer Inst Monogr 2000; (27):75-93; PMID:10963621; http://dx.doi.org/10.1093/oxfordjournals.jncimonographs.a024247
  • Yuan L, Dietrich AK, Nardulli AM. 17beta-Estradiol alters oxidative stress response protein expression and oxidative damage in the uterus. Mol Cell Endocrinol 2014; 382:218-26; PMID:24103313; http://dx.doi.org/10.1016/j.mce.2013.09.023
  • Spencer WA, Vadhanam MV, Jeyabalan J, Gupta RC. Oxidative DNA damage following microsome/Cu(II)-mediated activation of the estrogens, 17beta-estradiol, equilenin, and equilin: role of reactive oxygen species. Chem Res Toxicol 2012; 25:305-14; PMID:22126130; http://dx.doi.org/10.1021/tx200356v
  • Alao JP. The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention. Mol Cancer 2007; 6:1-16; PMID:17199893; http://dx.doi.org/10.1186/1476-4598-6-24
  • Terada Y, Nakashima O, Inoshita S, Kuwahara M, Sasaki S, Marumo F. Mitogen-activated protein kinase cascade and transcription factors: the opposite role of MKK3/6-p38K and MKK1-MAPK. Nephrol Dial Transplant 1999; 14:45-7; PMID:10048449; http://dx.doi.org/10.1093/ndt/14.suppl_1.45
  • Zheng J, Li H, Zhu H, Xiao X, Ma Y. Genistein inhibits estradiol- and environmental endocrine disruptor-induced growth effects on neuroblastoma cells. Oncol Lett 2013; 5:1583-6; PMID:23761822
  • Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev 1998; 12:3499-511; PMID:9832503; http://dx.doi.org/10.1101/gad.12.22.3499
  • Yang K, Guo Y, Stacey WC, Harwalkar J, Fretthold J, Hitomi M, Stacey DW. Glycogen synthase kinase 3 has a limited role in cell cycle regulation of cyclin D1 levels. BMC Cell Biol 2006; 7:33; PMID:16942622; http://dx.doi.org/10.1186/1471-2121-7-33
  • Alao JP, Gamble SC, Stavropoulou AV, Pomeranz KM, Lam EW, Coombes RC, Vigushin DM. The cyclin D1 proto-oncogene is sequestered in the cytoplasm of mammalian cancer cell lines. Mol Cancer 2006; 5:7; PMID:16503970; http://dx.doi.org/10.1186/1476-4598-5-7
  • Casanovas O, Miro F, Estanyol JM, Itarte E, Agell N, Bachs O. Osmotic stress regulates the stability of cyclin D1 in a p38SAPK2-dependent manner. J Biol Chem 2000; 275:35091-7; PMID:10952989; http://dx.doi.org/10.1074/jbc.M006324200
  • Mikhailov A, Shinohara M, Rieder CL. Topoisomerase II and histone deacetylase inhibitors delay the G2/M transition by triggering the p38 MAPK checkpoint pathway. J Cell Biol 2004; 166:517-26; PMID:15302851; http://dx.doi.org/10.1083/jcb.200405167
  • Zou Y, Ewton DZ, Deng X, Mercer SE, Friedman E. Mirk/dyrk1B kinase destabilizes cyclin D1 by phosphorylation at threonine 288. J Biol Chem 2004; 279:27790-8; PMID:15075324; http://dx.doi.org/10.1074/jbc.M403042200
  • Pettersson K, Delaunay F, Gustafsson JA. Estrogen receptor beta acts as a dominant regulator of estrogen signaling. Oncogene 2000; 19:4970-8; PMID:11042684; http://dx.doi.org/10.1038/sj.onc.1203828
  • van Lipzig MM, ter Laak AM, Jongejan A, Vermeulen NP, Wamelink M, Geerke D, Meerman JH. Prediction of ligand binding affinity and orientation of xenoestrogens to the estrogen receptor by molecular dynamics simulations and the linear interaction energy method. J Med Chem 2004; 47:1018-30; PMID:14761204; http://dx.doi.org/10.1021/jm0309607
  • Harris DM, Besselink E, Henning SM, Go VLW, Heber D. Phytoestrogens induce differential estrogen receptor alpha- or beta-mediated responses in transfected breast cancer cells. Exp Biol Med 2005; 230:558-68; PMID:16118406
  • Schreihofer DA. Transcriptional regulation by phytoestrogens in neuronal cell lines. Mol Cell Endocrinol 2005; 231:13-22; PMID:15713532; http://dx.doi.org/10.1016/j.mce.2004.12.006
  • Matthews JB, Twomey K, Zacharewski TR. In vitro and in vivo interactions of bisphenol A and its metabolite, bisphenol A glucuronide, with estrogen receptors alpha and beta. Chem Res Toxicol 2001; 14:149-57; PMID:11258963
  • Jeng YJ, Watson CS. Proliferative and anti-proliferative effects of dietary levels of phytoestrogens in rat pituitary GH3/B6/F10 cells - the involvement of rapidly activated kinases and caspases. BMC Cancer 2009; 9:334; PMID:19765307; http://dx.doi.org/10.1186/1471-2407-9-334
  • Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, Shibuya M, Fukami Y. Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem 1987; 262:5592-5; PMID:3106339
  • Yan GR, Xiao CL, He GW, Yin XF, Chen NP, Cao Y, He QY. Global phosphoproteomic effects of natural tyrosine kinase inhibitor, genistein, on signaling pathways. Proteomics 2010; 10:976-86; PMID:20049867
  • Daibata M, Mellinghoff I, Takagi S, Humphreys RE, Sairenji T. Effect of genistein, a tyrosine kinase inhibitor, on latent EBV activation induced by cross-linkage of membrane IgG in Akata B cells. J Immunol 1991; 147:292-7; PMID:1711079
  • Golden GA, Mason RP, Tulenko TN, Zubenko GS, Rubin RT. Rapid and opposite effects of cortisol and estradiol on human erythrocyte Na+,K+-ATPase activity: relationship to steroid intercalation into the cell membrane. Life Sci 1999; 65:1247-55; PMID:10503940; http://dx.doi.org/10.1016/S0024-3205(99)00360-4
  • Whiting KP, Restall CJ, Brain PF. Steroid hormone-induced effects on membrane fluidity and their potential roles in non-genomic mechanisms. Life Sci 2000; 67:743-57; PMID:10968404; http://dx.doi.org/10.1016/S0024-3205(00)00669-X
  • Routledge EJ, White R, Parker MG, Sumpter JP. Differential effects of xenoestrogens on coactivator recruitment by estrogen receptor (ER) alpha and ERbeta. J Biol Chem 2000; 275:35986-93; PMID:10964929; http://dx.doi.org/10.1074/jbc.M006777200
  • Leclercq G, Jacquot Y. Interactions of isoflavones and other plant derived estrogens with estrogen receptors for prevention and treatment of breast cancer-considerations concerning related efficacy and safety. J Steroid Biochem Mol Biol 2014; 139:237-44; PMID:23274118; http://dx.doi.org/10.1016/j.jsbmb.2012.12.010
  • van Bokhoven A, Varella-Garcia M, Korch C, Johannes WU, Smith EE, Miller HL, Nordeen SK, Miller GJ, Lucia MS. Molecular characterization of human prostate carcinoma cell lines. Prostate 2003; 57:205-25; PMID:14518029; http://dx.doi.org/10.1002/pros.10290
  • Maggiolini M, Vivacqua A, Carpino A, Bonofiglio D, Fasanella G, Salerno M, Picard D, Ando S. The mutant androgen receptor T877A mediates the proliferative but not the cytotoxic dose-dependent effects of genistein and quercetin on human LNCaP prostate cancer cells. Mol Pharmacol 2002; 62:1027-35; PMID:12391264; http://dx.doi.org/10.1124/mol.62.5.1027
  • Jeng Y, Watson C. Combinations of physiologic estrogens with xenoestrogens alter ERK phosphorylation profiles in rat pituitary cells. Environ Health Perspect 2011; 119:104-12; PMID:20870566; http://dx.doi.org/10.1289/ehp.1002512
  • Watson CS, Jeng Y-J, Guptarak J. Endocrine disruption via estrogen receptors that participate in nongenomic signaling pathways. J Steroid Biochem Mol Biol 2011; 127:44-50; PMID:21300151; http://dx.doi.org/10.1016/j.jsbmb.2011.01.015