Open access peer-reviewed chapter

A Potential New Mechanism for Bisphenol Molecules to Initiate Breast Cancer through Alteration of Bone Morphogenetic Protein Signaling in Stem Cells and Their Microenvironment

Written By

Boris Guyot and Veronique Maguer-Satta

Submitted: 29 August 2019 Reviewed: 25 October 2019 Published: 10 December 2019

DOI: 10.5772/intechopen.90273

From the Edited Volume

Breast Cancer Biology

Edited by Dil Afroze, Bilal Rah, Shazia Ali, Faheem Shehjar, Mohd Ishaq Dar, Shailender S. Chauhan and Natasha Thakur

Chapter metrics overview

975 Chapter Downloads

View Full Metrics

Abstract

Endocrine disruptors interfere with endocrine-mediated regulations of cell or organ functions. Estrogens are one of the main hormones altered by endocrine disruptors like bisphenol A (BPA). Stem cells are active from embryogenesis to late stages of adult life. Their unique properties, such as an extended lifespan and low cycling features, render these cell privileged targets of long-term exposure to numerous factors. Therefore, stem cells are likely to be affected following exposure to endocrine disruptors. One of the major signaling pathways involved in stem cell regulation is the bone morphogenetic protein (BMP) pathway. The BMP pathway is known for its involvement in numerous physiological and pathophysiological processes. Exposure of human mammary stem cells to pollutants such as BPA initiates fundamental changes in stem cells, in particular by altering major elements of BMP signaling, such as receptor expression and localization. Lastly, BPA and its substitute bisphenol S (BPS) have similar impacts on BMP signaling despite their different ER-binding properties, supporting the hypothesis that their biological effects cannot be extrapolated only from their interaction with ERα66. We review recent discoveries in this field and discuss their implications for cancer diagnosis, prevention, and treatment, as well as their relevance for studies on endocrine disruptors.

Keywords

  • BMP
  • bisphenol
  • stem cells
  • breast cancer
  • microenvironment
  • endocrine disruptors
  • estrogen

1. Introduction

Breast cancer is the most common cancer in women and exhibits important phenotypic and genetic diversities associated with different prognoses. Breast cancer subtypes are clinically classified based on histological appearance and expression of hormone receptors such as estrogen (ER) and progesterone (PR) receptors, as well as on the amplification of the HER2 gene coding for a member of the EGF receptor family [1]. Based on these criteria, four major breast cancer subtypes have been defined: luminal A and luminal B (all ER+), HER+ (that can be either ER or ER+), and basal-like (ER) [2, 3]. The most frequent subtype encompasses ER-positive tumors that represent almost 80% of breast cancers. In these tumors, preventing ER activation via hormone therapy is efficient. This can be achieved either by using competitive antagonists of estrogens (e.g., tamoxifen), preventing its binding to and subsequent activation of ER, by using drugs blocking estrogen synthesis (antiaromatase) in postmenopausal women, or by luteinizing hormone-releasing hormone (LHRH) analogs, inhibiting release of female hormones by the ovaries [4].

Breast cancer is a multifactorial disease, and evidences of the involvement of extrinsic factors in the increase of breast cancer risk have been described, such as the environment or lifestyle. Indeed, lack of physical activity, elevated tobacco or alcohol consumption, and the use of contraceptive pills or hormone-replacement therapy (for postmenopausal women) have been shown to increase breast cancer risk [5]. Hormonal status has also been described to play a major role in breast cancer risk. It has been shown that a premature or extensive exposure to endogenous estrogens (due to an early menarche, nulliparity, late age for first full-term pregnancy, or late menopause) increases the risk of breast cancer development.

Several chemical pollutants have been classified as endocrine-disrupting chemicals (EDCs) based on the following definition: “an endocrine disruptor is an exogenous substance or mixture that alters any function(s) of hormone actions and consequently causes adverse health effects in an intact organism, or its progeny, or (sub)populations” [6, 7, 8, 9, 10, 11]. Estrogens are one of the main hormones altered by EDCs. Perturbations in estrogen functions have been identified in a wide spectrum of pathologies, including metabolic, bone, and reproductive disorders, as well as breast, endometrial, or ovarian cancers. Therefore, it is important to consider that the mammary gland is exposed throughout life not only to endogenous hormones but also to EDCs, molecules present in the environment and able to mimic these hormones.

Interest in EDCs is growing rapidly, owing notably to their extensive use in manufactured goods and their release in our environment. Among these environmental pollutants, bisphenol molecules are being increasingly studied in breast cancer due to their estrogen-mimetic properties, enabling them to activate estrogen signaling through their binding to the ER, in particular, bisphenol A (BPA) [12, 13]. Despite rising concerns about its safety [14] and progressive restrictions on its use, several million tons of BPA are still produced worldwide.

Advertisement

2. The major effects of bisphenols on BMP signaling and stem cells

2.1 BPA and breast cancer

2.1.1 BPA and estrogen signaling

BPA is an aromatic organic compound used by the plastic industry as a monomer in the synthesis of polycarbonates and epoxy resins. Polycarbonates are found in consumer plastic-like water bottles, food packaging materials, sport equipment or toys, while epoxy resins are used to coat the inside of food or beverage containers. BPA can also be found in thermal paper. BPA monomers from these compounds can be released into the environment by hydrolysis. At the structural level, BPA is a diphenyl compound with two hydroxyl groups in a “para” position rendering it highly similar to synthetic estrogen (diethylstilbestrol). This thus allows BPA to interact with various physiological receptors similar to estrogen, including ERs.

The classical genomic estrogen signaling pathway is triggered by the binding of estrogen to its α or β receptors that act as transcription factors in the nucleus. In the absence of ligands, these receptors are complexed with inhibitory molecules either in the cytoplasm or in the nucleus of the cell. Upon ligand binding, these complexes dissociate resulting in conformational changes that allow DNA binding and recruitment of cofactors to regulate expression of target genes [15]. Both ERα and ERβ are also able to initiate a nongenomic signaling pathway outside of the nucleus depending on their subcellular localization [16]. Moreover, estrogen signaling can also be mediated by other receptors, such as GPR30, EGFR, to list only a few [15, 17].

In line with the current definition of EDCs, BPA was shown to exert its activity by disrupting the estrogen signaling pathway that uses ER as a transcription factor binding to estrogen response element (ERE) sites on DNA [15]. Consequently, estrogen-mimetics (e.g., BPA) were mechanistically thought to primarily act through their binding to ERα66, the main canonical (nuclear) estrogen receptor. This nuclear receptor initiates signaling pathways at the cell membrane and transcriptional responses in the cell nucleus. BPA also upregulates the level of steroid receptor coactivators (SRC-1, SRC-3) and promotes the activity of EREs [18]. However, BPA has also been shown to bind to a number of distinct nuclear and membrane receptors, namely estrogen receptors ERα/β, androgen receptor (AR), G protein-coupled ER (GPER, GPR30), PPAR (especially PPARγ), insulin-like growth factor-1 (IGF1-R) [17, 19, 20]. BPA stimulates the release of EGFR ligands by directly targeting other molecules than ER, like ADAM17 or ADAM10 [21]. Furthermore, the impact of BPA on Ca2+ release or ERK signaling has been highlighted in the pancreas [15]. Altogether, these results indicate that BPA, in addition to its effects on the canonical estrogen pathway, is able to perturb numerous physiological processes through estrogen genomic and nongenomic signaling, as well as nonestrogen-related pathways [19, 22]. Importantly, BPA is at the origin of toxic derivatives (chlorinated bisphenols) and is also processed by cellular and biochemical mechanisms to generate a number of different BPA metabolites. All these BPA derivatives have been reported to have similar or higher toxic effects than BPA [19, 20]. In the context of the mammary gland, it is thus of utmost importance to further elucidate how BPA, its derivatives or metabolites, modulate estrogen- or nonestrogen-related signaling. This should improve our understanding of the tumorigenic potential of BPA, firstly in the luminal breast cancer subtype, and subsequently in other tumor types.

2.1.2 BPA involvement in breast cancer

Evidence gathered from studies in experimental models and human populations has already confirmed that EDCs, including BPA, contribute to increased risk of disease [23, 24]. A positive relationship between exposure to BPA and cancer development is reported in the literature [25]. However, whether BPA is actually harmful for human health remains understudied, similar to our understanding of the molecular mechanisms underlying BPA-dependent effects in cancer development.

Given the significant involvement of estrogens in both normal and pathological conditions, EDCs able to interfere with the homeostasis of the estrogen endocrine system are a potential source of several health disorders. In this context, a human population-based study detected a significant increase in serum levels of BPA and established a correlation with breast tissue density measured in mammographies [26]. This finding was attributed to the ability of BPA to increase proliferation of mammary epithelial cells from either normal or breast cancer tissues [27, 28]. Epigenetic data from human tumors or cells exposed to BPA in vitro revealed the ability of this EDC to directly induce mammary epithelial cell transformation [29].

Moreover, BPA was correlated with breast cancer patients with high risk profiles and therefore with increased disease relapse [30]. This may be due to the implication of BPA in breast cancer metastasis. This process has traditionally been associated with late stages of cancer development, though a new hypothesis on its origin has progressively emerged suggesting that it could be an inherent mark of tumor cell [31, 32]. Metastatic dissemination is a dynamic process that involves several steps: local invasion of cells from the primary tumor, intravasation leading to dissemination through the blood or lymph, extravasation to invade new tissues, implantation, and finally new tumor growth. Numerous signaling pathways and programs are activated during this process such as epithelial-to-mesenchymal transition (EMT), anoïkis, migration, and proliferation among others (for review: [33, 34]). It has been shown that ER-negative breast cancers are associated with an increased risk of developing metastases [35]. Indeed, these breast cancers express more mesenchymal markers such as vimentin and N-cadherin or EMT-transcription factors that are required for metastatic initiation. Conversely, ER-positive tumors are associated with a more differentiated luminal phenotype, expressing epithelial markers (E-cadherin, ER, FOXA1 for instance). Accordingly, a downregulation of the luminal-specific transcription factor FOXA1 is induced after BPA treatment in triple-negative tumor cell lines, leading to the induction of EMT and increasing cell motility [36]. In this study, BPA treatment was shown to activate the PI3K/AKT pathway, leading to a downregulation of epithelial genes alongside an upregulation of mesenchymal genes. Another study demonstrated that BPA promotes migration and invasion via GPER, which transduces FAK, Src, and ERK2 signaling pathway activation [37]. Promotion of GPER-induced migration by BPA or BPS occurs via different signaling pathways. Indeed, in contrast to BPA, which acts via the FAK/Scr/ERK2 pathway, it has been shown that BPS induces GPER/Hippo-YAP-dependent migration [38]. Effects of BPA, BPS, and BPF on migration and EMT properties of ER-positive tumor cell lines were compared [39]. After treatment, cells lost cell/cell contacts and acquired a fibroblast-like morphology associated with an EMT phenotype. This was further confirmed after analysis of EMT-associated protein expression showing a decrease in E-cadherin and an increase in N-cadherin. Moreover, BPA-, BPS-, and BPF-treated cells displayed a stronger migratory ability. All of these modifications were inhibited after administration of an ER antagonist, demonstrating the ER-dependent effects of these bisphenols [39].

At the mechanistic level, a large number of in vivo and in vitro studies have highlighted the ability of BPA to disrupt several key signaling pathways that are known to be involved in breast cancer [19, 40]. However, the direct involvement of BPA in breast cancer incidence is difficult to establish and remains controversial [26, 41], owing possibly to the fact that different mechanisms are depicted in either ER-positive or -negative tumors, reflecting the variety of biological effects arising from exposure to BPA [20, 28, 42]. In addition, the combinatorial effects of different pollutants encountered over a life time also complicate these studies. Hence, scientists are faced with a huge challenge in order to formally establish the transforming power of BPA, owing to the different contexts and mechanistic cascades of alterations occurring in the human breast tissue during such long-term exposure.

2.2 BPA target cells

2.2.1 Stem cells in mammary gland

Mammary gland development takes place during embryogenesis and is composed of a rudimentary ductal system blocked until puberty. Then, two master reproductive hormones are secreted, namely estrogens and progesterone. Estrogens control the growth of ducts from their distal extremity called terminal end buds (TEBs) [43, 44, 45], while progesterone is involved in lateral branch development [46, 47]. One of the major hormones involved in mammary gland development is estrogen, mostly produced by the ovaries (but also by other tissues). Estrogens, in combination with other hormones, orchestrate the growth of the ductal system and adipose tissue accumulation during puberty and at further stages of development [43, 44, 45].

In adults, the mammary gland is formed of ducts and lobules of secreting luminal epithelial cells surrounded by contractile myoepithelial cells. These epithelial cells are embedded in a stroma mainly formed of fibroblasts and adipocytes that secrete several soluble molecules regulating epithelial cell function and differentiation. Epithelial cells of the mammary gland are generated by mammary stem cells (MaSCs) and the stromal compartment by mesenchymal stem cells (MSCs) [48, 49, 50, 51]. During adulthood, the mammary gland undergoes functional and structural changes that alternate between phases of proliferation, differentiation, and apoptosis controlled by cyclic hormonal variations due to the estrous/menstrual cycle modulating the stem cell compartment [52]. However, this postpubertal mammary tree remains immature and only achieves full maturation during pregnancy and lactation. These final steps involve alveogenesis and milk production, which take place mostly under the control of progesterone and prolactin [53, 54]. Studies indicate that estrogens do not directly stimulate proliferation of ER-positive luminal cells but act via a paracrine process [55, 56]. Indeed, estrogen acts on luminal ER/PR-positive cells, leading to the cleavage and liberation of amphiregulin [57, 58], which then affects neighboring ER/PR-negative cells. These ER/PR-negative cells display characteristics of stem cells, in that, their asymmetric division is controlled by growth factors released by stromal cells [59, 60, 61, 62]. Conversely, estrogen treatment induces a deficient asymmetric division of a human MaSC cell line (MCF10F) [63]. Ovariectomized mice (or letrozole treated to inhibit endogenous estrogen synthesis and provide a normal stromal and hormonal environment for all other hormones) show a decrease in the ability of MaSCs to repopulate a mammary fat pad and to generate ductal growth and expansion without impacting the size of the MaSCs-enriched subpopulation [52]. Collectively, these studies highlight the importance of the estrogen pathway on MaSC regulation through direct and indirect effects and consequently suggest potential sensitivity of these cells to estrogen-mimetics like BPA. Furthermore, stem cells are a unique category of cells active from embryogenesis up to late stages of human adult life, and are thus more prone to be exposed to EDCs, likely altering their normal functions [64, 65, 66, 67, 68].

2.2.2 BPA, stem cells, and breast cancer

It has been shown that exposure to EDCs occurs throughout life and even during embryogenesis, at the stage of mammary gland establishment. For instance, BPA has been detected in urinary samples but also in maternal and fetal plasma, in colostrum, and in placental tissue at birth. Several studies demonstrated that a prenatal exposure to BPA induces changes in fetal mouse mammary gland, in the epithelial as well as stromal compartments, favoring fat pad maturation and increasing the mammary gland susceptibility to carcinogens [69, 70, 71]. This is accompanied by transcriptome modifications, in particular, an increase in the expression of genes belonging to the antiapoptotic family, myoepithelial differentiation, and adipogenesis, and a decrease in those involved in cell adhesion [71]. Exposure to BPA at puberty alters the function of MaSCs, leading to the appearance in the regenerated glands of early neoplastic lesions with molecular alterations similar to those detected in early neoplastic breast cancer tissues [72]. In a physiological model in which mice were treated at puberty with BPA, estrogen-dependent transcriptional events were perturbed and the number of terminal end buds was altered in a dose-dependent fashion [27]. In vitro exposure of normal human mammary epithelial cells to BPA was shown to induce their proliferation due to the secretion of autocrine growth factors and allow them to generate bigger mammospheres [73]. Treated cells displayed an increase in DNA hypermethylation of tumor suppressor genes, such as Brca1. These data support that BPA can promote early pretumoral stages corroborating findings in normal human breast epithelial cells (MCF-10F) [29, 64]. Indeed, BPA-treated human MaSC lines, such as MCF-10F, increase their expression of genes involved in DNA repair and decrease proapoptotic gene expression [74]. Chronic exposure of MCF10A cells to BPA at doses similar to those measured in contaminated water lead to major MaSC modifications affecting their stem cell properties and regulation [64]. Importantly, BPA treatment increases stem-like features by inducing the expression of ALDH1 and SOX2 genes, a human MaSCs marker and a master regulator of pluripotency in embryonic stem cells, respectively [75]. BPA also perturbs signals involved in human mammary stem cell (ERα66 negative cells) regulation, like the bone morphogenetic protein (BMP) pathway, which has been identified in their transformation [76], partly by changing BMP membrane receptor availability and priming cells to BMP signaling [64]. These data raised the hypothesis that in ER-positive tumors, under tamoxifen treatment and in a BPA-containing environment, some cells could acquire resistance to treatment by a switch in signaling pathway favoring a stem-like phenotype characterized by a decrease in treatment cytotoxicity and a modification of the stoichiometry of the type of ER (e.g., an increase in ERRγ or ERα isoform expression).

Overall, these observations strongly support that MaSCs are directly sensitive to BPA, which could be involved in their transformation and/or treatment escape [27, 72, 74].

2.3 BMP, stem cells, and cancer

2.3.1 BMP and mammary epithelial stem cells

One of the major conserved signaling pathways involved in stem cell regulation from embryogenesis up to adult stages is BMP signaling. There are 21 different soluble BMP molecules that act through serine/threonine kinase BMP receptors (BMPRs). In the context of stem cell regulation, BMP2 and BMP4 are progressively emerging as the most important BMPs. The BMP pathway is involved in numerous physiological and pathological processes [77]. BMPs control MSC regulation, such as lineage specification of adipocytes which are one of the major elements of the mammary gland microenvironment [78, 79, 80]. Alterations in BMP signaling have been implicated in metabolic disorders such as obesity in women [81, 82].

During embryogenesis in mice, BMP4 was shown to participate in the early steps of mammary gland development by regulating the dorsoventral axis establishment [83]. The BMP pathway also plays a role in mammary bud formation and outgrowth, as well as in ductal branching morphogenesis initiation. Indeed, BMP4 is expressed in both mesenchymal and epithelial cells of the mammary bud and the use of a BMP4 inhibitor leads to a decrease in bud outgrowth [84]. A link between BMPs and progesterone receptor type A involved in branching morphogenesis during postnatal mammary gland development has also been shown [85]. In addition, BMPs are also involved in the myoepithelial compartmentalization and lumen formation [85]. The knockout of a BMP extracellular antagonist, Twisted, abrogates lumen formation and disorganizes the myoepithelial layer through a decrease in SMAD1-5-8 phosphorylation and the repression of BMP targets (Msx1, Msx2, and Gata-3) [86]. In human cells, BMP2 regulates luminal epithelial cells by modulating the expression of GATA-3 and FOXA1 [76]. Finally, an in vitro study using sorted mouse mammary epithelial undifferentiated cells demonstrated the role of BMP signaling in final maturation steps such as lactogenic differentiation [87].

In healthy tissues, epithelial cells, as well as cells within the mammary gland environment (fibroblasts, adipose tissue cells, hematopoietic cells), contribute to the production of soluble BMP2 and BMP4 molecules [76], while distinct subpopulations of normal mammary epithelial cells sorted according to CD10 and EPCAM expression [88] express different elements of the BMP pathway. A role for BMP molecules in MaSC regulation was formally demonstrated by functional assay analyses following exposure of different human cell types to soluble BMP2 or BMP4 [76], and further substantiated by the use of TGF/BMP inhibitors allowing the expansion of immature epithelial basal cells [89]. Interestingly, as in the hematopoietic system [90], BMP2 and BMP4 molecules have distinct functional effects on MaSC regulation despite their strong homology. Indeed, while BMP4 modulates the compartment of MaSC and myoepithelial progenitors, BMP2 allows the commitment and proliferation of luminal progenitors [76]. However, the molecular mechanism by which BMPs interact with estrogen signaling to regulate MaSCs remains to be further deciphered.

2.3.2 BMP and breast cancer

BMP signaling is also a well-known highly complex pathway that orchestrates the development and homeostasis of adult tissues such as the neural system [91]. The importance of BMP signaling alterations in cancer stem cell features has been revealed in glioblastoma, breast cancer, and leukemia [90, 92, 93, 94, 95]. The role of BMPs, especially of BMP2 and BMP4, in breast cancer has been largely documented [96, 97]. Alterations of BMP ligand expression and signaling have been reported and shown to be clinically correlated with breast cancer progression [98, 99] and to play a major role in the development of bone metastases [99, 100, 101]. Despite the fact that BMP4 transcripts are expressed at various levels in tumor tissues or breast cancer cell lines [102], high levels of BMP4 are found in 25% of the breast cancer tumors displaying a low proliferation index but high recurrence rate [98]. BMP4 has crucial functions in promoting tumor growth arrest, migration and metastasis by mediating cell cycle arrest in G1 [102], chemokine regulation [103], and inhibition of lumen formation [104] for example. However, the biological effects of BMP4 largely depend on cell context, as they were reported to be either proliferative or antiproliferative in mammary epithelial cells according to cellular density and cooperative factors [105, 106]. The microenvironment of human primary luminal breast tumors produces abnormally high amounts of soluble BMP2 compared to healthy tissue, while higher BMPR1B levels were detected in tumor cells [76, 107]. Chronic exposure to high BMP2 concentrations was demonstrated to initiate MaSC transformation toward a luminal tumor phenotype dependent on a BMPR1B-initiated signaling cascade involved in luminal commitment of normal MaSC. This leads to a FOXA1/FOXC1 transcription factor balance switch in favor of FOXA1, simultaneously with an upregulation of GATA3 [76]. However, while an increase in soluble BMP2 in the tumor microenvironment has been shown in luminal ER-positive tumors where it is correlated with a high BMPR1B tumor expression [76], a strong decrease in BMP2 transcripts was found in ER-negative breast tumors [108]. Also, a downmodulation of the BMPR1B (Alk6) in a basal cell line (MDA-MB-231) increased cell growth in vitro [109], suggesting an antiproliferative function for BMPR1B in ER-negative tumors. Interestingly, downregulation of BMPR1A (ALK3) in MDA-MB231D (a bone metastatic clone of MDA-MB231) basal ER-negative cells inhibited their migration and bone metastatic properties [110]. Therefore, it is very likely that the BMP2/BMPR1B signal is overactivated in the context of ER-positive tumors, while being repressed in ER-negative tumors.

Some of the first steps of carcinogenesis are an increase in proliferation, evasion of apoptosis, and activation of survival signaling pathways. To achieve this, several tumor suppressor genes, like p53 or BRCA1 for instance, need to be inactivated by different mechanisms including epigenetic changes. Modulation of BMP signaling by epigenetic mechanisms [111], such as methylation of BMP-receptor promoters, has been of particular clinical interest to further stratify glioblastoma patients and propose new therapeutic strategies [92]. While different genetic alterations progressively appear following different oncogenic signals, heredity likely accounts for only 10–30% of breast cancers. Based on epidemiological studies, different factors increasing the risk of breast cancer development have been highlighted. They can be intrinsic, like mutations in BRCA1 or 2, Tp53, ATM, or also PTEN, or extrinsic, like environmental factors or lifestyle [112, 113]. In breast cancers with a genetic origin, the most commonly mutated genes are BRCA1 and BRCA2, associated with an increase in cancer risk. BRCA1 and 2 are two major regulators of double-strand breaks (DSB) DNA repair through homologous recombination (HR) and play a crucial role as tumor suppressor genes. In this context, it is interesting to note that a family member and negative regulator of P53, DNp63 has been reported to mediate activation of BMP signaling in order to govern epithelial cell plasticity, EMT, and tumorigenicity during breast cancer initiation and progression [114, 115]. DNp63 has also been identified as a repressor of BRCA1 expression exclusively in ER-positive breast cancer cells [116]. Moreover, a correlation between the BMP pathway and the P53-ATM signaling has been reported [117]. However, the importance of these different signaling crosstalks in the context of breast cancer, exposure to EDCs, and stem cell transformation need to be investigated.

2.4 BMP, estrogen, and bisphenols

2.4.1 BMP and ER crosstalk

The BMP signaling pathway is a dynamic and complex pathway, leading to the transduction of various signals depending on the nature of the BMP ligand and of the BMPR complex oligomerization induced (for review: [118, 119]). It has been shown that BMPs may interact with their receptors in two different ways [120, 121]: on the one hand, BMPs induce a BMPR complex formation called BISC (BMP-induced signaling complex), and on the other hand, a preformed BMPR complex is present before BMP fixation, known as PFC (preformed complex). These two different modes of BMP signal initiation lead to two different signaling cascades, namely the canonical SMAD-dependent pathway and the noncanonical SMAD-independent pathway [121]. SMAD-phosphorylated proteins then form a complex with SMAD4, leading to its translocation to the nucleus where it acts as a transcription factor on target genes [118, 122]. The SMAD-independent pathway does not simply encompass one signaling pathway but a multitude of downstream cascades, involving p38, Ras/ERK, and PI3K/AKT [123, 124, 125, 126]. Interestingly, SMAD1-5-8 phosphorylation is more abundant in undifferentiated murine progenitors and decreases with their differentiation until it is almost fully abrogated in the differentiated cells treated with prolactin [87]. Involvement of the BMPR1A/SMAD1-5-8 pathway in lactogenic differentiation was further confirmed by the lack of expression of a lactogenic differentiation marker (beta-casein) at the RNA and protein levels in BMPR1A knockdown mammary cell lines [87]. These data demonstrate that the BMP pathway constitutes an important regulator of the mammary gland during embryogenesis but most likely also during adulthood. However, the molecular and functional crosstalk between the BMP and estrogen signaling pathways is poorly understood. A first set of experiments describes the repression of BMP signaling by ER inhibition of BMP production through a direct interaction between SMAD1 and ER [127]. Reciprocally, a BMP2 signal was shown to upregulate the expression of ER receptors, including the induction of specific ER isoforms such as ERα36 [128, 129]. Interestingly, crosstalk between BMP4 and estrogen signaling seems to have opposite effects. Indeed, BMP4 inhibits ERα signaling by promoting receptor degradation through the proteosomal pathway, while estrogens repress BMP4 expression [130]. Similarly, estrogen represses BMP4 expression in cardiomyocytes by preventing BMP4-mediated ERβ expression and JNK activity in this system [131]. In addition, in this context, estrogen inhibition of BMP4 is independent of Smad1/5/8 activity [131]. BMP4, upon activation of its canonical pathway, represses CYP17A1 and induces the transcription of CYP19A1, involved in androgen and estrogen synthesis, respectively [132]. In a rat model of pituitary cells, estrogen stimulates the transcriptional activity of BMP4-specific SMADs through an ER-SMAD1 complex shown to stimulate prolactin production, while having no effect on the TGFβ/SMAD pathway [133]. Similarly, the inhibitory effects of estrogen signaling on the BMP pathway appear to be mediated by a direct physical interaction between ER receptors and the SMAD1 BMP signaling element in a luminal breast cancer cell line model (MCF7). The physical interaction between ERα and SMAD1 requires the DNA binding domain of ERα and this complex formation is dependent on BMP2 and estrogen [127]. Moreover, BMP signaling has also been directly identified in thyroid-lineage specification [134, 135] as well as in thyroid carcinoma [136]. Interestingly, thyroid hormone status interferes with estrogen target gene expression in breast cancer samples in menopausal women [137]. These findings highlight the need to further investigate the importance of the BMP pathway in both thyroid and estrogen signaling in a broader context of exposure to EDCs.

More recently, BMP2-mediated luminal transformation of MCF10A was shown to be accompanied by a strong activation of the estrogen signaling pathway despite the absence of ERα66 in those cells [76]. Our understanding of estrogen signaling is hindered by the existence of several isoforms generated by alternative splicing and different promoter usage [138]. Interaction of these isoforms with the BMP signaling elements has not yet being investigated but could be involved in epithelial stem cell response to BMP2. Indeed, the importance of these different ERα isoforms in mammary epithelial SC features and in the context of breast cancer is only just starting to be identified [139, 140]. These isoforms can be expressed in both ERα66-positive and -negative cells and display different subcellular localizations [141, 142]. For example, unlike ERα66, ERα36 is expressed mainly at the plasma membrane and activates estrogen nongenomic signaling by activating the ERK pathway through an interplay with the MKP3 phosphatase [143]. Interestingly, in the context of EDC research, ERα36 displays altered ligand preference and causes distinct effects compared to ERα66. For instance, the tamoxifen drug used as an estrogen antagonist in ERα66 breast cancers behaves as an estrogen agonist for ERα36 [140, 144]. Collectively, these different examples illustrate how BMP signaling through its interaction with estrogen signaling is at the crossroad of a number of fundamental physiological processes. The BMP pathway is therefore directly involved in mammary stem cell regulation and transformation, yet adverse effects of EDCs, like BPA, on the BMP pathway have not been thoroughly investigated (Figure 1).

Figure 1.

Illustration of the main findings that show a crosstalk between BMP and estrogen signaling pathways.

2.4.2 BMP and bisphenols

Works from our team and others suggest that bisphenols could act on multiple cell types of the mammary gland, and their effects may converge to provoke major dysregulations of the BMP pathway that could contribute to luminal breast cancer initiation. Indeed, we observed a major impact of BPA on the mammary microenvironment (niche) equilibrium. BPA greatly increases BMP2 production by stromal cells of the human mammary SC microenvironment reaching levels comparable to those measured in luminal breast cancer [76]. Moreover, BPA treatment leads to a decrease in estrogen and BMP15 production in oocytes delaying their maturation [145]. A decrease in BMP2 production through a direct binding of BPA to ERγ was involved in bone loss through a suppression of osteoblast differentiation reverted by inhibition of ERγ [146]. This suggests that the effects and mechanisms of BPA-induced BMP ligand production depend on the estrogen receptor expression profile and are context dependent [147]. However, the molecular mechanism by which BPA induces BMP2 production by stromal cells of the mammary gland BMP2 is not yet known. On the other hand, we have demonstrated that long-term exposure (60 days) to BPA initiates fundamental changes in human mammary stem cells themselves, in particular, by altering major BMP signaling elements such as receptor expression and localization [64]. This results in the “priming” of stem cells to exogenous activating signals of the BMP pathway and sensitizes them to be more sensitive to exogenous soluble BMP ligands. We then demonstrated for the first time that nongenotoxic alterations of both the stem cells and their niche act synergistically to initiate a transforming process mediated by the BMP signaling perturbation leading to the emergence of ER-positive tumors [76]. Interestingly, these previous studies showed that BPA impacts BMP signaling pathway members in both mammary epithelial and stromal cells that do not express ERα66. At the mechanistic level, the pathways used by BPA to induce these effects in cells remain to be deciphered, focusing notably on their reliance on other ERα isoforms or on ER-independent factors.

These questions are of great interest for understanding the effects of both BPA and estrogens since it has been reported that some cell lines respond to an estrogen signal despite their very low levels or complete absence of ER [148]. In response to accumulating evidence in favor of adverse health effects following exposure to BPA, likely mediated by its activation of ERα66, alternative bisphenols have been developed such as BPS and BPF that are considered safer due to their very low binding affinity to ERα [149, 150, 151]. However, an increasing number of studies show that these alternative bisphenol molecules are not as innocuous as anticipated, including an impact on obesity, steatosis, and reproduction [20]. In a study previously conducted in our team, assessing the impact of bisphenol on BMP2 production by stromal cells of the mammary gland, we were surprised to observe that BPA and BPS displayed very similar effects [76]. Indeed, both BPA (high affinity binding to estrogen receptors) and its substitute BPS (very weak affinity binding to estrogen receptors) induce BMP2 synthesis in the healthy breast stroma, raising concerns as to whether these bisphenols mediate their transforming effects solely through a classical ER-dependent mechanism. Since then, other studies have shown that BPS, as well as BPF, induces similar if not more potent effects than BPA [20, 152, 153]. Moreover, it was reported that BPA treatment increases aromatase expression and its activity in healthy breast fibroblasts, leading to an increase in estrogen biosynthesis and secretion. The same observations were made after treatment with BPS [154]. These results are of particular interest with regards to the important role of the microenvironment in the different steps of carcinogenesis and in the context of MaSC-driven transformation by BMP signaling. Our work thus indicates that the BMP pathway could be altered by several EDCs such as BPA and its proposed alternatives, both at the level of stem cells and their microenvironment. This suggests that early detection of increased BMP2 levels in the mammary microenvironment may constitute a reliable marker of early transformation process and could be a valuable indicator of exposure to EDCs such as bisphenols. In addition, the interplay between BMP and estrogen pathways both at the molecular and functional levels prompt us to further decipher the mechanisms underlying bisphenol- and BMP-induced transformation in mammary epithelial stem cells.

Advertisement

3. Conclusions

Different signaling pathways often engage in complex interactions synergistically mediating an appropriate cellular response. Estrogen signaling is no exception and it is likely involved in a crosstalk with the BMP pathway at multiple levels in the mammary gland. BMPs are secreted proteins active in a very large number of organs and tissues during development, adulthood, and pathogenesis [155]. Previous work suggested a close interaction between ER-mediated estrogen signaling and the BMP pathway in different cell types of the mammary gland. In a model of mammary epithelial stem cells, E2 or known EDCs like BPA or BPS were able to potentiate SMAD activation by BMP2 [64]. This was possibly due to a physical interaction between ERα isoforms and SMAD factors, such as that reported for ERα or ERβ, and could be associated with an increased risk of cell transformation by long-term exposure to BMP2. Deciphering the dysregulations of the BMP signaling pathway has been remarkably useful in identifying its importance in cancer stem cell phenotypes in the neural system [92, 93]. The role of alterations of BMP signaling to sustain cancer stem cell features has been extended by us and others in breast cancer and leukemia [90, 94, 95]. We showed that chronic exposure to high concentrations of BMP2 drives the transformation of mammary stem cells toward the luminal tumor subtype [76] through binding to its BMPR1B receptor. However, downstream mechanisms and crosstalks with estrogen signaling in those mammary stem cells remained to be understood. This is especially important in the context of several studies that demonstrated the involvement of BPA in the proliferation of either ER-positive or -negative cancer cells. In addition, BPA can trigger proliferation via nonclassical estrogen receptors, including the estrogen-related receptor gamma (ERRγ) [156, 157]. We also demonstrated that long-term exposure of human mammary stem cells (ER-negative in terms of ERα-66 expression) to pollutants such as BPA initiates fundamental changes in stem cells by altering major BMP signaling components [64], thus “priming” stem cells to exogenous BMP activation. Complementary to this effect on epithelial stem cells, we revealed an impact of BPA on the tumor microenvironment through the induction of the synthesis of high levels of BMP2 by normal fibroblasts and stromal cells reaching levels similar to those measured in breast tumors [76].

Resistance and relapse can be due to tumor adaptation or evolution. Indeed, therapies elicit a selective pressure on cells, which in turn develop resistance, notably by acquiring mutations. Resistance to tamoxifen of ER-positive tumors can be caused by a loss of ER [158], its mutation, or posttranslational modification [159] among others. It was shown that BPA is involved in chemoresistance [160] and notably in resistance to tamoxifen in ER-positive tumor cell lines [161] by decreasing tamoxifen-induced apoptosis and increasing gene expression of ERRα, which contributes to resistance to tamoxifen [162] and cell proliferation [157]. Another study demonstrated that an ERα variant could be induced by BMP2 [128] and may be involved in resistance to tamoxifen [163]. The addiction of cancer cells toward BMP signaling and the crosstalk with estrogen signaling is currently under consideration as a new therapeutic avenue for ER-positive breast cancer patients [164]. At the clinical level, targeting estrogen signaling has been decisive in improving the outcome of ER-positive breast cancer patients. At the era of immunotherapy, the analysis of the impact of bisphenols on the immune system and on tumor surveillance is crucial. This will need to be pursued to improve our understanding and implementation of antiestrogen therapies in the context of their combination with new immune treatments [165]. Overall, these data indicate that disruption of BMP signaling affects both the stem cells and their niche at different stages of the disease, which could be instrumental in the management of breast cancer.

Several studies demonstrated that BPS promotes breast cancer cell proliferation, notably through an ER-cyclin D1-CDK4/6-pRb-dependent pathway, exclusively in ER-positive breast cancer cells [38, 39, 166]. Moreover, it has also been demonstrated that BPF has the same proliferative action as BPA, BPS, or estrogen treatments on transformed ER-positive cells. Similar to BPS, this proliferative effect relies on cyclin D and E expression through ER-dependent pathways [39]. BPS, as shown for BPA, can also induce epigenetic and transcriptional changes in breast cancer cells, resulting in an increase in the expression of genes implicated in proliferation, cellular attachment as well as adhesion and migration [167]. Lastly, the bioavailability of BPS substitutes might be higher than for BPA. A recent study conducted in pigs, an ideal model for mimicking the human digestive tract, demonstrated the lower plasma clearance of BPS (3.5 lower) compared to BPA and an increased oral systemic exposure exceeding 250-fold [168]. These observations draw our attention and raise concerns about replacing BPA by BPS, as this may result in an increased internal exposure to EDCs.

To conclude, BMP signaling plays a major role in the regulation of SCs and of their microenvironment (niche), in both normal and tumor contexts. Multiple abnormalities of BMP signaling have been observed in cancer, but until recently studies had mostly focused on its role in advanced disease. However, due to the number of studies describing the importance of BMP signaling throughout breast cancer development (from initiation, progression, metastasis up to resistance), we suggest that early detection of BMP signaling alterations, such as increased levels of BMP2 and/or of BMP receptors, may constitute a reliable marker of exposure to BPA. This suggests that further investigations into alterations of the BMP pathway in the context of exposure to bisphenols should improve our understanding of associated side effects.

Advertisement

Acknowledgments

The authors thank the following funding bodies for their support of the team’s work mentioned in this review: the Association pour la Recherche contre le Cancer, La Ligue contre le Cancer, the Déchaine ton cœur association, the Région Rhône-Alpes and the Institut National du Cancer.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

The author thanks Brigitte Manship for English proofreading and critical reading.

References

  1. 1. Pourteimoor V, Mohammadi-Yeganeh S, Paryan M. Breast cancer classification and prognostication through diverse systems along with recent emerging findings in this respect; the dawn of new perspectives in the clinical applications. Tumour Biology. 2016;37(11):14479-14499
  2. 2. Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, et al. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Annals of Oncology. 2013;24(9):2206-2223
  3. 3. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(19):10869-10874
  4. 4. Lumachi F, Santeufemia DA, Basso SM. Current medical treatment of estrogen receptor-positive breast cancer. World Journal of Biological Chemistry. 2015;6(3):231-239
  5. 5. Momenimovahed Z, Salehiniya H. Epidemiological characteristics of and risk factors for breast cancer in the world. Breast Cancer (Dove Medical Press). 2019;11:151-164
  6. 6. WHO IPoCS. Global assessment of the state-of-the-science of endocrine disruptors. 2002
  7. 7. WHO/UNEP. State of the science of endocrine disrupting chemicals—2012. 2012
  8. 8. Goodson WH 3rd, Lowe L, Carpenter DO, Gilbertson M, Manaf Ali A, Lopez de Cerain Salsamendi A, et al. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: The challenge ahead. Carcinogenesis. 2015;36(Suppl 1):S254-S296
  9. 9. Kabir ER, Rahman MS, Rahman I. A review on endocrine disruptors and their possible impacts on human health. Environmental Toxicology and Pharmacology. 2015;40(1):241-258
  10. 10. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, et al. EDC-2: The endocrine society's second scientific statement on endocrine-disrupting chemicals. Endocrine Reviews. 2015;36(6):E1-E150
  11. 11. Zoeller RT, Brown TR, Doan LL, Gore AC, Skakkebaek NE, Soto AM, et al. Endocrine-disrupting chemicals and public health protection: A statement of principles from the Endocrine Society. Endocrinology. 2012;153(9):4097-4110
  12. 12. Li Y, Perera L, Coons LA, Burns KA, Tyler Ramsey J, Pelch KE, et al. Differential in vitro biological action, coregulator interactions, and molecular dynamic analysis of Bisphenol A (BPA), BPAF, and BPS ligand-ERalpha complexes. Environmental Health Perspectives. 2018;126(1):017012
  13. 13. Routledge EJ, White R, Parker MG, Sumpter JP. Differential effects of xenoestrogens on coactivator recruitment by estrogen receptor (ER) alpha and ERbeta. The Journal of Biological Chemistry. 2000;275(46):35986-35993
  14. 14. Rochester JR. Bisphenol A and human health: A review of the literature. Reproductive Toxicology. 2013;42:132-155
  15. 15. Alonso-Magdalena P, Ropero AB, Soriano S, Garcia-Arevalo M, Ripoll C, Fuentes E, et al. Bisphenol-A acts as a potent estrogen via non-classical estrogen triggered pathways. Molecular and Cellular Endocrinology. 2012;355(2):201-207
  16. 16. Hammes SR, Levin ER. Extranuclear steroid receptors: Nature and actions. Endocrine Reviews. 2007;28(7):726-741
  17. 17. MacKay H, Abizaid A. A plurality of molecular targets: The receptor ecosystem for bisphenol-A (BPA). Hormones and Behavior. 2018;101:59-67
  18. 18. Wang T, Liu B, Guan Y, Gong M, Zhang W, Pan J, et al. Melatonin inhibits the proliferation of breast cancer cells induced by bisphenol A via targeting estrogen receptor-related pathways. Thoracic Cancer. 2018;9(3):368-375
  19. 19. Murata M, Kang JH. Bisphenol A (BPA) and cell signaling pathways. Biotechnology Advances. 2018;36(1):311-327
  20. 20. Siracusa JS, Yin L, Measel E, Liang S, Yu X. Effects of bisphenol A and its analogs on reproductive health: A mini review. Reproductive Toxicology. 2018;79:96-123
  21. 21. Urriola-Munoz P, Li X, Maretzky T, McIlwain DR, Mak TW, Reyes JG, et al. The xenoestrogens biphenol-A and nonylphenol differentially regulate metalloprotease-mediated shedding of EGFR ligands. Journal of Cellular Physiology. 2018;233(3):2247-2256
  22. 22. Watson CS, Bulayeva NN, Wozniak AL, Alyea RA. Xenoestrogens are potent activators of nongenomic estrogenic responses. Steroids. 2007;72(2):124-134
  23. 23. Holmes D. Breast cancer: Increased risk with concurrent dietary and EDC exposures. Nature Reviews. Endocrinology. 2017;13(7):378
  24. 24. Hussain I, Bhan A, Ansari KI, Deb P, Bobzean SA, Perrotti LI, et al. Bisphenol-A induces expression of HOXC6, an estrogen-regulated homeobox-containing gene associated with breast cancer. Biochimica et Biophysica Acta. 2015;1849(6):697-708
  25. 25. Hafezi SA, Abdel-Rahman WM. The endocrine disruptor Bisphenol A (BPA) exerts a wide range of effects in carcinogenesis and response to therapy. Current Molecular Pharmacology. 2019;12(3):230-238;
  26. 26. Sprague BL, Trentham-Dietz A, Hedman CJ, Wang J, Hemming JD, Hampton JM, et al. Circulating serum xenoestrogens and mammographic breast density. Breast Cancer Research. 2013;15(3):R45
  27. 27. Ayyanan A, Laribi O, Schuepbach-Mallepell S, Schrick C, Gutierrez M, Tanos T, et al. Perinatal exposure to bisphenol a increases adult mammary gland progesterone response and cell number. Molecular Endocrinology. 2011;25(11):1915-1923
  28. 28. Gao H, Yang BJ, Li N, Feng LM, Shi XY, Zhao WH, et al. Bisphenol A and hormone-associated cancers: Current progress and perspectives. Medicine (Baltimore). 2015;94(1):e211
  29. 29. Fernandez SV, Russo J. Estrogen and xenoestrogens in breast cancer. Toxicologic Pathology. 2010;38(1):110-122
  30. 30. Dairkee SH, Seok J, Champion S, Sayeed A, Mindrinos M, Xiao W, et al. Bisphenol A induces a profile of tumor aggressiveness in high-risk cells from breast cancer patients. Cancer Research. 2008;68(7):2076-2080
  31. 31. Seyfried TN, Huysentruyt LC. On the origin of cancer metastasis. Critical Reviews in Oncogenesis. 2013;18(1-2):43-73
  32. 32. Weigelt B, Peterse JL, Van’t Veer LJ. Breast cancer metastasis: Markers and models. Nature Reviews. Cancer. 2005;5(8):591-602
  33. 33. Gupta GP, Massague J. Cancer metastasis: Building a framework. Cell. 2006;127(4):679-695
  34. 34. Jin X, Mu P. Targeting breast cancer metastasis. Breast Cancer (Auckl.). 2015;9(Suppl 1):23-34
  35. 35. Rangel R, Guzman-Rojas L, Kodama T, Kodama M, Newberg JY, Copeland NG, et al. Identification of new tumor suppressor genes in triple-negative breast cancer. Cancer Research. 2017;77(15): 4089-4101
  36. 36. Zhang XL, Wang HS, Liu N, Ge LC. Bisphenol A stimulates the epithelial mesenchymal transition of estrogen negative breast cancer cells via FOXA1 signals. Archives of Biochemistry and Biophysics. 2015;585:10-16
  37. 37. Castillo Sanchez R, Gomez R, Perez Salazar E. Bisphenol A induces migration through a GPER-, FAK-, Src-, and ERK2-dependent pathway in MDA-MB-231 breast cancer cells. Chemical Research in Toxicology. 2016;29(3):285-295
  38. 38. Deng Q , Jiang G, Wu Y, Li J, Liang W, Chen L, et al. GPER/hippo-YAP signal is involved in Bisphenol S induced migration of triple negative breast cancer (TNBC) cells. Journal of Hazardous Materials. 2018;355:1-9
  39. 39. Kim JY, Choi HG, Lee HM, Lee GA, Hwang KA, Choi KC. Effects of bisphenol compounds on the growth and epithelial mesenchymal transition of MCF-7 CV human breast cancer cells. Journal of Biomedical Research. 2017;31(4):358-369
  40. 40. Shafei A, Matbouly M, Mostafa E, Al Sannat S, Abdelrahman M, Lewis B, et al. Stop eating plastic, molecular signaling of bisphenol A in breast cancer. Environmental Science and Pollution Research International. 2018;25(24):23624-23630
  41. 41. Morgan M, Deoraj A, Felty Q , Roy D. Environmental estrogen-like endocrine disrupting chemicals and breast cancer. Molecular and Cellular Endocrinology. 2017;457:89-102
  42. 42. Shafei A, Ramzy MM, Hegazy AI, Husseny AK, El-Hadary UG, Taha MM, et al. The molecular mechanisms of action of the endocrine disrupting chemical bisphenol A in the development of cancer. Gene. 2018;647:235-243
  43. 43. Silberstein GB, Van Horn K, Shyamala G, Daniel CW. Essential role of endogenous estrogen in directly stimulating mammary growth demonstrated by implants containing pure antiestrogens. Endocrinology. 1994;134(1):84-90
  44. 44. Feng Y, Manka D, Wagner KU, Khan SA. Estrogen receptor-alpha expression in the mammary epithelium is required for ductal and alveolar morphogenesis in mice. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(37):14718-14723
  45. 45. Daniel CW, Silberstein GB, Strickland P. Direct action of 17 beta-estradiol on mouse mammary ducts analyzed by sustained release implants and steroid autoradiography. Cancer Research. 1987;47(22):6052-6057
  46. 46. Lydon JP, Sivaraman L, Conneely OM. A reappraisal of progesterone action in the mammary gland. Journal of Mammary Gland Biology and Neoplasia. 2000;5(3):325-338
  47. 47. Shyamala G. Progesterone signaling and mammary gland morphogenesis. Journal of Mammary Gland Biology and Neoplasia. 1999;4(1):89-104
  48. 48. Stingl J, Eirew P, Ricketson I, Shackleton M, Vaillant F, Choi D, et al. Purification and unique properties of mammary epithelial stem cells. Nature. 2006;439(7079):993-997
  49. 49. Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, Asselin-Labat ML, et al. Generation of a functional mammary gland from a single stem cell. Nature. 2006;439(7072):84-88
  50. 50. Inman JL, Robertson C, Mott JD, Bissell MJ. Mammary gland development: Cell fate specification, stem cells and the microenvironment. Development. 2015;142(6):1028-1042
  51. 51. Lloyd-Lewis B, Harris OB, Watson CJ, Davis FM. Mammary stem cells: Premise, properties, and perspectives. Trends in Cell Biology. 2017;27(8):556-567
  52. 52. Asselin-Labat ML, Vaillant F, Sheridan JM, Pal B, Wu D, Simpson ER, et al. Control of mammary stem cell function by steroid hormone signalling. Nature. 2010;465(7299):798-802
  53. 53. Brisken C. Hormonal control of alveolar development and its implications for breast carcinogenesis. Journal of Mammary Gland Biology and Neoplasia. 2002;7(1):39-48
  54. 54. Humphreys RC, Lydon JP, O’Malley BW, Rosen JM. Use of PRKO mice to study the role of progesterone in mammary gland development. Journal of Mammary Gland Biology and Neoplasia. 1997;2(4):343-354
  55. 55. Mallepell S, Krust A, Chambon P, Brisken C. Paracrine signaling through the epithelial estrogen receptor alpha is required for proliferation and morphogenesis in the mammary gland. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(7):2196-2201
  56. 56. Stingl J. Estrogen and progesterone in normal mammary gland development and in cancer. Hormones and Cancer. 2011;2(2):85-90
  57. 57. Ciarloni L, Mallepell S, Brisken C. Amphiregulin is an essential mediator of estrogen receptor alpha function in mammary gland development. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(13):5455-5460
  58. 58. LaMarca HL, Rosen JM. Estrogen regulation of mammary gland development and breast cancer: Amphiregulin takes center stage. Breast Cancer Research. 2007;9(4):304
  59. 59. Sternlicht MD, Sunnarborg SW, Kouros-Mehr H, Yu Y, Lee DC, Werb Z. Mammary ductal morphogenesis requires paracrine activation of stromal EGFR via ADAM17-dependent shedding of epithelial amphiregulin. Development. 2005;132(17):3923-3933
  60. 60. Villadsen R, Fridriksdottir AJ, Ronnov-Jessen L, Gudjonsson T, Rank F, LaBarge MA, et al. Evidence for a stem cell hierarchy in the adult human breast. Journal of Cell Biology. 2007;177(1):87-101
  61. 61. Booth BW, Smith GH. ERalpha and PR are expressed in label-retaining mammary epithelial cells that divide asymmetrically and retain their template DNA strands. Breast Cancer Research. 2006;8(4):R49
  62. 62. Booth BW, Boulanger CA, Anderson LH, Jimenez-Rojo L, Brisken C, Smith GH. Amphiregulin mediates self-renewal in an immortal mammary epithelial cell line with stem cell characteristics. Experimental Cell Research. 2010;316(3):422-432
  63. 63. Russo J, Snider K, Pereira JS, Russo IH. Estrogen induced breast cancer is the result in the disruption of the asymmetric cell division of the stem cell. Hormone Molecular Biology and Clinical Investigation. 2010;1(2):53-65
  64. 64. Clement F, Xu X, Donini CF, Clement A, Omarjee S, Delay E, et al. Long-term exposure to bisphenol A or benzo(a)pyrene alters the fate of human mammary epithelial stem cells in response to BMP2 and BMP4, by pre-activating BMP signaling. Cell Death and Differentiation. 2017;24(1):155-166
  65. 65. Kopras E, Potluri V, Bermudez ML, Williams K, Belcher S, Kasper S. Actions of endocrine-disrupting chemicals on stem/progenitor cells during development and disease. Endocrine-Related Cancer. 2014;21(2):T1-T12
  66. 66. Bateman ME, Strong AL, McLachlan JA, Burow ME, Bunnell BA. The effects of endocrine disruptors on adipogenesis and osteogenesis in mesenchymal stem cells: A review. Frontiers in Endocrinology. 2016;7:171
  67. 67. Alonso-Magdalena P, Rivera FJ, Guerrero-Bosagna C. Bisphenol-A and metabolic diseases: Epigenetic, developmental and transgenerational basis. Environmental Epigenetics. 2016;2(3):dvw022
  68. 68. Landero-Huerta DA, Vigueras-Villasenor RM, Yokoyama-Rebollar E, Arechaga-Ocampo E, Rojas-Castaneda JC, Jimenez-Trejo F, et al. Epigenetic and risk factors of testicular germ cell tumors: A brief review. Frontiers in Bioscience (Landmark Edition). 2017;22:1073-1098
  69. 69. Moral R, Wang R, Russo IH, Lamartiniere CA, Pereira J, Russo J. Effect of prenatal exposure to the endocrine disruptor bisphenol A on mammary gland morphology and gene expression signature. The Journal of Endocrinology. 2008;196(1):101-112
  70. 70. Vandenberg LN, Maffini MV, Wadia PR, Sonnenschein C, Rubin BS, Soto AM. Exposure to environmentally relevant doses of the xenoestrogen bisphenol-A alters development of the fetal mouse mammary gland. Endocrinology. 2007;148(1):116-127
  71. 71. Wadia PR, Cabaton NJ, Borrero MD, Rubin BS, Sonnenschein C, Shioda T, et al. Low-dose BPA exposure alters the mesenchymal and epithelial transcriptomes of the mouse fetal mammary gland. PLoS One. 2013;8(5):e63902
  72. 72. Wang D, Gao H, Bandyopadhyay A, Wu A, Yeh IT, Chen Y, et al. Pubertal bisphenol A exposure alters murine mammary stem cell function leading to early neoplasia in regenerated glands. Cancer Prevention Research (Philadelphia, Pa.). 2014;7(4):445-455
  73. 73. Qin XY, Fukuda T, Yang L, Zaha H, Akanuma H, Zeng Q , et al. Effects of bisphenol A exposure on the proliferation and senescence of normal human mammary epithelial cells. Cancer Biology & Therapy. 2012;13(5):296-306
  74. 74. Fernandez SV, Huang Y, Snider KE, Zhou Y, Pogash TJ, Russo J. Expression and DNA methylation changes in human breast epithelial cells after bisphenol A exposure. International Journal of Oncology. 2012;41(1):369-377
  75. 75. Lillo MA, Nichols C, Seagroves TN, Miranda-Carboni GA, Krum SA. Bisphenol A induces Sox2 in ER(+) breast cancer stem-like cells. Hormones and Cancer. 2017;8(2):90-99
  76. 76. Chapellier M, Bachelard-Cascales E, Schmidt X, Clement F, Treilleux I, Delay E, et al. Disequilibrium of BMP2 levels in the breast stem cell niche launches epithelial transformation by overamplifying BMPR1B cell response. Stem Cell Reports. 2015;4(2):239-254
  77. 77. Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, et al. Bone morphogenetic protein (BMP) signaling in development and human diseases. Genes & Diseases. 2014;1(1):87-105
  78. 78. Huang RL, Sun Y, Ho CK, Liu K, Tang QQ , Xie Y, et al. IL-6 potentiates BMP-2-induced osteogenesis and adipogenesis via two different BMPR1A-mediated pathways. Cell Death & Disease. 2018;9(2):144
  79. 79. Zhang X, Guo J, Zhou Y, Wu G. The roles of bone morphogenetic proteins and their signaling in the osteogenesis of adipose-derived stem cells. Tissue Engineering. Part B, Reviews. 2014;20(1):84-92
  80. 80. Gustafson B, Hammarstedt A, Hedjazifar S, Hoffmann JM, Svensson PA, Grimsby J, et al. BMP4 and BMP antagonists regulate human white and beige adipogenesis. Diabetes. 2015;64(5):1670-1681
  81. 81. Ribeiro S, Lopes LR, Paula Costa G, Figueiredo VP, Shrestha D, Batista AP, et al. CXCL-16, IL-17, and bone morphogenetic protein 2 (BMP-2) are associated with overweight and obesity conditions in middle-aged and elderly women. Immunity & Ageing. 2017;14:6
  82. 82. Zamani N, Brown CW. Emerging roles for the transforming growth factor-{beta} superfamily in regulating adiposity and energy expenditure. Endocrine Reviews. 2011;32(3):387-403
  83. 83. Cho KW, Kim JY, Song SJ, Farrell E, Eblaghie MC, Kim HJ, et al. Molecular interactions between Tbx3 and Bmp4 and a model for dorsoventral positioning of mammary gland development. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(45):16788-16793
  84. 84. Hens JR, Dann P, Zhang JP, Harris S, Robinson GW, Wysolmerski J. BMP4 and PTHrP interact to stimulate ductal outgrowth during embryonic mammary development and to inhibit hair follicle induction. Development. 2007;134(6):1221-1230
  85. 85. Fleming JM, Ginsburg E, Goldhar AS, Plant J, Vonderhaar BK. Progesterone receptor activates Msx2 expression by downregulating TNAP/Akp2 and activating the Bmp pathway in EpH4 mouse mammary epithelial cells. PLoS One. 2012;7(3):e34058
  86. 86. Forsman CL, Ng BC, Heinze RK, Kuo C, Sergi C, Gopalakrishnan R, et al. BMP-binding protein twisted gastrulation is required in mammary gland epithelium for normal ductal elongation and myoepithelial compartmentalization. Developmental Biology. 2013;373(1):95-106
  87. 87. Perotti C, Karayazi O, Moffat S, Shemanko CS. The bone morphogenetic protein receptor-1A pathway is required for lactogenic differentiation of mammary epithelial cells in vitro. In Vitro Cellular & Developmental Biology—Animal. 2012;48(6):377-384
  88. 88. Bachelard-Cascales E, Chapellier M, Delay E, Pochon G, Voeltzel T, Puisieux A, et al. The CD10 enzyme is a key player to identify and regulate human mammary stem cells. Stem Cells. 2010;28(6):1081-1088
  89. 89. Mou H, Vinarsky V, Tata PR, Brazauskas K, Choi SH, Crooke AK, et al. Dual SMAD signaling inhibition enables long-term expansion of diverse epithelial basal cells. Cell Stem Cell. 2016;19(2):217-231
  90. 90. Zylbersztejn F, Flores-Violante M, Voeltzel T, Nicolini FE, Lefort S, Maguer-Satta V. The BMP pathway: A unique tool to decode the origin and progression of leukemia. Experimental Hematology. 2018;61:36-44
  91. 91. Bier E, De Robertis EM. Embryo development. BMP gradients: A paradigm for morphogen-mediated developmental patterning. Science. 2015;348(6242):aaa5838
  92. 92. Lee J, Son MJ, Woolard K, Donin NM, Li A, Cheng CH, et al. Epigenetic-mediated dysfunction of the bone morphogenetic protein pathway inhibits differentiation of glioblastoma-initiating cells. Cancer Cell. 2008;13(1):69-80
  93. 93. Piccirillo SG, Reynolds BA, Zanetti N, Lamorte G, Binda E, Broggi G, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 2006;444(7120):761-765
  94. 94. Jung N, Maguer-Satta V, Guyot B. Early steps of mammary stem cells transformation by exogenous signals, effects of the bisphenols endocrine disruptors. Cancers. 2019;11(9): 1351-1368
  95. 95. Chapellier M, Maguer-Satta V. BMP2, a key to uncover luminal breast cancer origin linked to pollutant effects on epithelial stem cells niche. Molecular & Cellular Oncology. 2016;3(3):e1026527
  96. 96. Alarmo EL, Kallioniemi A. Bone morphogenetic proteins in breast cancer: Dual role in tumourigenesis? Endocrine-Related Cancer. 2010;17(2):R123-RR39
  97. 97. Thawani JP, Wang AC, Than KD, Lin CY, La Marca F, Park P. Bone morphogenetic proteins and cancer: Review of the literature. Neurosurgery. 2010;66(2):233-246
  98. 98. Alarmo EL, Huhtala H, Korhonen T, Pylkkanen L, Holli K, Kuukasjarvi T, et al. Bone morphogenetic protein 4 expression in multiple normal and tumor tissues reveals its importance beyond development. Modern Pathology: An Official Journal of the United States and Canadian Academy of Pathology, Inc. 2013;26(1):10-21
  99. 99. Zabkiewicz C, Resaul J, Hargest R, Jiang WG, Ye L. Bone morphogenetic proteins, breast cancer, and bone metastases: Striking the right balance. Endocrine-Related Cancer. 2017;24(10):R349-RR66
  100. 100. Ye L, Jiang WG. Bone morphogenetic proteins in tumour associated angiogenesis and implication in cancer therapies. Cancer Letters. 2016;380(2):586-597
  101. 101. Bellanger A, Donini CF, Vendrell JA, Lavaud J, Machuca-Gayet I, Ruel M, et al. The critical role of the ZNF217 oncogene in promoting breast cancer metastasis to the bone. The Journal of Pathology. 2017;242(1):73-89
  102. 102. Ketolainen JM, Alarmo EL, Tuominen VJ, Kallioniemi A. Parallel inhibition of cell growth and induction of cell migration and invasion in breast cancer cells by bone morphogenetic protein 4. Breast Cancer Research and Treatment. 2010;124(2):377-386
  103. 103. Owens P, Pickup MW, Novitskiy SV, Chytil A, Gorska AE, Aakre ME, et al. Disruption of bone morphogenetic protein receptor 2 (BMPR2) in mammary tumors promotes metastases through cell autonomous and paracrine mediators. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(8):2814-2819
  104. 104. Montesano R. Bone morphogenetic protein-4 abrogates lumen formation by mammary epithelial cells and promotes invasive growth. Biochemical and Biophysical Research Communications. 2007;353(3):817-822
  105. 105. Masuda H, Otsuka F, Matsumoto Y, Takano M, Miyoshi T, Inagaki K, et al. Functional interaction of fibroblast growth factor-8, bone morphogenetic protein and estrogen receptor in breast cancer cell proliferation. Molecular and Cellular Endocrinology. 2011;343(1-2):7-17
  106. 106. Montesano R, Sarkozi R, Schramek H. Bone morphogenetic protein-4 strongly potentiates growth factor-induced proliferation of mammary epithelial cells. Biochemical and Biophysical Research Communications. 2008;374(1):164-168
  107. 107. Helms MW, Packeisen J, August C, Schittek B, Boecker W, Brandt BH, et al. First evidence supporting a potential role for the BMP/SMAD pathway in the progression of oestrogen receptor-positive breast cancer. Journal of Pathology. 2005;206(3):366-376
  108. 108. Arnold SF, Tims E, McGrath BE. Identification of bone morphogenetic proteins and their receptors in human breast cancer cell lines: Importance of BMP2. Cytokine. 1999;11(12):1031-1037
  109. 109. Bokobza SM, Ye L, Kynaston HE, Mansel RE, Jiang WG. Reduced expression of BMPR-IB correlates with poor prognosis and increased proliferation of breast cancer cells. Cancer Genomics & Proteomics. 2009;6(2):101-108
  110. 110. Katsuno Y, Hanyu A, Kanda H, Ishikawa Y, Akiyama F, Iwase T, et al. Bone morphogenetic protein signaling enhances invasion and bone metastasis of breast cancer cells through Smad pathway. Oncogene. 2008;27(49):6322-6333
  111. 111. Zhang M, Wang Q , Yuan W, Yang S, Wang X, Yan JD, et al. Epigenetic regulation of bone morphogenetic protein-6 gene expression in breast cancer cells. The Journal of Steroid Biochemistry and Molecular Biology. 2007;105(1-5):91-97
  112. 112. Lal A, Ramazzotti D, Weng Z, Liu K, Ford JM, Sidow A. Comprehensive genomic characterization of breast tumors with BRCA1 and BRCA2 mutations. BMC Medical Genomics. 2019;12(1):84
  113. 113. Lima ZS, Ghadamzadeh M, Arashloo FT, Amjad G, Ebadi MR, Younesi L. Recent advances of therapeutic targets based on the molecular signature in breast cancer: Genetic mutations and implications for current treatment paradigms. Journal of Hematology & Oncology. 2019;12(1):38
  114. 114. Balboni AL, Hutchinson JA, DeCastro AJ, Cherukuri P, Liby K, Sporn MB, et al. DeltaNp63alpha-mediated activation of bone morphogenetic protein signaling governs stem cell activity and plasticity in normal and malignant mammary epithelial cells. Cancer Research. 2013;73(2):1020-1030
  115. 115. DeCastro AJ, Cherukuri P, Balboni A, DiRenzo J. DeltaNP63alpha transcriptionally activates chemokine receptor 4 (CXCR4) expression to regulate breast cancer stem cell activity and chemotaxis. Molecular Cancer Therapeutics. 2015;14(1):225-235
  116. 116. Amin R, Morita-Fujimura Y, Tawarayama H, Semba K, Chiba N, Fukumoto M, et al. DeltaNp63alpha induces quiescence and downregulates the BRCA1 pathway in estrogen receptor-positive luminal breast cancer cell line MCF7 but not in other breast cancer cell lines. Molecular Oncology. 2016;10(4):575-593
  117. 117. Chau JF, Jia D, Wang Z, Liu Z, Hu Y, Zhang X, et al. A crucial role for bone morphogenetic protein-Smad1 signalling in the DNA damage response. Nature Communications. 2012;3:836
  118. 118. Miyazono K, Maeda S, Imamura T. BMP receptor signaling: Transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine & Growth Factor Reviews. 2005;16(3):251-263
  119. 119. Yadin D, Knaus P, Mueller TD. Structural insights into BMP receptors: Specificity, activation and inhibition. Cytokine & Growth Factor Reviews. 2016;27:13-34
  120. 120. Nohe A, Hassel S, Ehrlich M, Neubauer F, Sebald W, Henis YI, et al. The mode of bone morphogenetic protein (BMP) receptor oligomerization determines different BMP-2 signaling pathways. The Journal of Biological Chemistry. 2002;277(7):5330-5338
  121. 121. Hassel S, Schmitt S, Hartung A, Roth M, Nohe A, Petersen N, et al. Initiation of Smad-dependent and Smad-independent signaling via distinct BMP-receptor complexes. The Journal of Bone and Joint Surgery. American Volume. 2003;85-A(Suppl 3):44-51
  122. 122. Miyazono K, Kamiya Y, Morikawa M. Bone morphogenetic protein receptors and signal transduction. Journal of Biochemistry. 2010;147(1):35-51
  123. 123. Gamell C, Osses N, Bartrons R, Ruckle T, Camps M, Rosa JL, et al. BMP2 induction of actin cytoskeleton reorganization and cell migration requires PI3-kinase and Cdc42 activity. Journal of Cell Science. 2008;121(Pt 23):3960-3970
  124. 124. Guicheux J, Lemonnier J, Ghayor C, Suzuki A, Palmer G, Caverzasio J. Activation of p38 mitogen-activated protein kinase and c-Jun-NH2-terminal kinase by BMP-2 and their implication in the stimulation of osteoblastic cell differentiation. Journal of Bone and Mineral Research. 2003;18(11):2060-2068
  125. 125. Hay E, Lemonnier J, Fromigue O, Marie PJ. Bone morphogenetic protein-2 promotes osteoblast apoptosis through a Smad-independent, protein kinase C-dependent signaling pathway. The Journal of Biological Chemistry. 2001;276(31):29028-29036
  126. 126. Vinals F, Lopez-Rovira T, Rosa JL, Ventura F. Inhibition of PI3K/p70 S6K and p38 MAPK cascades increases osteoblastic differentiation induced by BMP-2. FEBS Letters. 2002;510(1-2):99-104
  127. 127. Yamamoto T, Saatcioglu F, Matsuda T. Cross-talk between bone morphogenic proteins and estrogen receptor signaling. Endocrinology. 2002;143(7):2635-2642
  128. 128. Wang D, Huang P, Zhu B, Sun L, Huang Q , Wang J. Induction of estrogen receptor alpha-36 expression by bone morphogenetic protein 2 in breast cancer cell lines. Molecular Medicine Reports. 2012;6(3):591-596
  129. 129. Matsumoto Y, Otsuka F, Takano M, Mukai T, Yamanaka R, Takeda M, et al. Estrogen and glucocorticoid regulate osteoblast differentiation through the interaction of bone morphogenetic protein-2 and tumor necrosis factor-alpha in C2C12 cells. Molecular and Cellular Endocrinology. 2010;325(1-2):118-127
  130. 130. Qian SW, Liu Y, Wang J, Nie JC, Wu MY, Tang Y, et al. BMP4 cross-talks with estrogen/ERalpha signaling to regulate adiposity and glucose metabolism in females. eBioMedicine. 2016;11:91-100
  131. 131. Wang YC, Xiao XL, Li N, Yang D, Xing Y, Huo R, et al. Oestrogen inhibits BMP4-induced BMP4 expression in cardiomyocytes: A potential mechanism of oestrogen-mediated protection against cardiac hypertrophy. British Journal of Pharmacology. 2015;172(23):5586-5595
  132. 132. Liu Y, Du SY, Ding M, Dou X, Zhang FF, Wu ZY, et al. The BMP4-Smad signaling pathway regulates hyperandrogenism development in a female mouse model. The Journal of Biological Chemistry. 2017;292(28):11740-11750
  133. 133. Giacomini D, Paez-Pereda M, Stalla J, Stalla GK, Arzt E. Molecular interaction of BMP-4, TGF-beta, and estrogens in lactotrophs: Impact on the PRL promoter. Molecular Endocrinology. 2009;23(7):1102-1114
  134. 134. Serra M, Alysandratos KD, Hawkins F, McCauley KB, Jacob A, Choi J, et al. Pluripotent stem cell differentiation reveals distinct developmental pathways regulating lung- versus thyroid-lineage specification. Development. 2017;144(21):3879-3893
  135. 135. Villacorte M, Delmarcelle AS, Lernoux M, Bouquet M, Lemoine P, Bolsee J, et al. Thyroid follicle development requires Smad1/5- and endothelial cell-dependent basement membrane assembly. Development. 2016;143(11):1958-1970
  136. 136. Meng X, Zhu P, Li N, Hu J, Wang S, Pang S, et al. Expression of BMP-4 in papillary thyroid carcinoma and its correlation with tumor invasion and progression. Pathology, Research and Practice. 2017;213(4):359-363
  137. 137. Conde SJ, Luvizotto Rde A, de Sibio MT, Nogueira CR. Thyroid hormone status interferes with estrogen target gene expression in breast cancer samples in menopausal women. ISRN Endocrinology. 2014;2014:317398
  138. 138. Wang ZY, Yin L. Estrogen receptor alpha-36 (ER-alpha36): A new player in human breast cancer. Molecular and Cellular Endocrinology. 2015;418(Pt 3):193-206
  139. 139. Deng H, Zhang XT, Wang ML, Zheng HY, Liu LJ, Wang ZY. ER-alpha36-mediated rapid estrogen signaling positively regulates ER-positive breast cancer stem/progenitor cells. PLoS One. 2014;9(2):e88034
  140. 140. Wang Q , Jiang J, Ying G, Xie XQ , Zhang X, Xu W, et al. Tamoxifen enhances stemness and promotes metastasis of ERalpha36(+) breast cancer by upregulating ALDH1A1 in cancer cells. Cell Research. 2018;28(3):336-358
  141. 141. Wang Z, Zhang X, Shen P, Loggie BW, Chang Y, Deuel TF. Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66. Biochemical and Biophysical Research Communications. 2005;336(4):1023-1027
  142. 142. Lee LM, Cao J, Deng H, Chen P, Gatalica Z, Wang ZY. ER-alpha36, a novel variant of ER-alpha, is expressed in ER-positive and -negative human breast carcinomas. Anticancer Research. 2008;28(1B):479-483
  143. 143. Omarjee S, Jacquemetton J, Poulard C, Rochel N, Dejaegere A, Chebaro Y, et al. The molecular mechanisms underlying the ERalpha-36-mediated signaling in breast cancer. Oncogene. 2017;36(18):2503-2514
  144. 144. Lin SL, Yan LY, Zhang XT, Yuan J, Li M, Qiao J, et al. ER-alpha36, a variant of ER-alpha, promotes tamoxifen agonist action in endometrial cancer cells via the MAPK/ERK and PI3K/Akt pathways. PLoS One. 2010;5(2):e9013
  145. 145. Wang X, Jiang SW, Wang L, Sun Y, Xu F, He H, et al. Interfering effects of bisphenol A on in vitro growth of preantral follicles and maturation of oocyes. Clinica Chimica Acta. 2018;485:119-125
  146. 146. Watson CS, Bulayeva NN, Wozniak AL, Finnerty CC. Signaling from the membrane via membrane estrogen receptor-alpha: Estrogens, xenoestrogens, and phytoestrogens. Steroids. 2005;70(5-7):364-371
  147. 147. Thent ZC, Froemming GRA, Muid S. Bisphenol A exposure disturbs the bone metabolism: An evolving interest towards an old culprit. Life Sciences. 2018;198:1-7
  148. 148. Mei J, Hu H, McEntee M, Plummer H 3rd, Song P, Wang HC. Transformation of non-cancerous human breast epithelial cell line MCF10A by the tobacco-specific carcinogen NNK. Breast Cancer Research and Treatment. 2003;79(1):95-105
  149. 149. Dreier DA, Connors KA, Brooks BW. Comparative endpoint sensitivity of in vitro estrogen agonist assays. Regulatory Toxicology and Pharmacology. 2015;72(2):185-193
  150. 150. Huang R, Sakamuru S, Martin MT, Reif DM, Judson RS, Houck KA, et al. Profiling of the Tox21 10K compound library for agonists and antagonists of the estrogen receptor alpha signaling pathway. Scientific Reports. 2014;4:5664
  151. 151. Peyre L, Rouimi P, de Sousa G, Helies-Toussaint C, Carre B, Barcellini S, et al. Comparative study of bisphenol A and its analogue bisphenol S on human hepatic cells: A focus on their potential involvement in nonalcoholic fatty liver disease. Food and Chemical Toxicology: An International Journal Published for the British Industrial Biological Research Association. 2014;70:9-18
  152. 152. Eladak S, Grisin T, Moison D, Guerquin MJ, N’Tumba-Byn T, Pozzi-Gaudin S, et al. A new chapter in the bisphenol A story: Bisphenol S and bisphenol F are not safe alternatives to this compound. Fertility and Sterility. 2015;103(1):11-21
  153. 153. Rochester JR, Bolden AL. Bisphenol S and F: A systematic review and comparison of the hormonal activity of Bisphenol A substitutes. Environmental Health Perspectives. 2015;123(7):643-650
  154. 154. Williams GP, Darbre PD. Low-dose environmental endocrine disruptors, increase aromatase activity, estradiol biosynthesis and cell proliferation in human breast cells. Molecular and Cellular Endocrinology. 2019;486:55-64
  155. 155. Katagiri T, Watabe T. Bone morphogenetic proteins. Cold Spring Harbor Perspectives in Biology. 2016;8(6)
  156. 156. Pupo M, Pisano A, Lappano R, Santolla MF, De Francesco EM, Abonante S, et al. Bisphenol A induces gene expression changes and proliferative effects through GPER in breast cancer cells and cancer-associated fibroblasts. Environmental Health Perspectives. 2012;120(8):1177-1182
  157. 157. Song H, Zhang T, Yang P, Li M, Yang Y, Wang Y, et al. Low doses of bisphenol A stimulate the proliferation of breast cancer cells via ERK1/2/ERRgamma signals. Toxicology in Vitro. 2015;30(1 Pt B):521-528
  158. 158. Kuukasjarvi T, Kononen J, Helin H, Holli K, Isola J. Loss of estrogen receptor in recurrent breast cancer is associated with poor response to endocrine therapy. Journal of Clinical Oncology. 1996;14(9):2584-2589
  159. 159. Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM, Corbo L. Cracking the estrogen receptor's posttranslational code in breast tumors. Endocrine Reviews. 2011;32(5):597-622
  160. 160. Lapensee EW, Tuttle TR, Fox SR, Ben-Jonathan N. Bisphenol A at low nanomolar doses confers chemoresistance in estrogen receptor-alpha-positive and -negative breast cancer cells. Environmental Health Perspectives. 2009;117(2):175-180
  161. 161. Huang B, Luo N, Wu X, Xu Z, Wang X, Pan X. The modulatory role of low concentrations of bisphenol A on tamoxifen-induced proliferation and apoptosis in breast cancer cells. Environmental Science and Pollution Research International. 2019;26(3):2353-2362
  162. 162. Riggins RB, Lan JP, Zhu Y, Klimach U, Zwart A, Cavalli LR, et al. ERRgamma mediates tamoxifen resistance in novel models of invasive lobular breast cancer. Cancer Research. 2008;68(21):8908-8917
  163. 163. Gu W, Dong N, Wang P, Shi C, Yang J, Wang J. Tamoxifen resistance and metastasis of human breast cancer cells were mediated by the membrane-associated estrogen receptor ER-alpha36 signaling in vitro. Cell Biology and Toxicology. 2017;33(2):183-195
  164. 164. Shee K, Jiang A, Varn FS, Liu S, Traphagen NA, Owens P, et al. Cytokine sensitivity screening highlights BMP4 pathway signaling as a therapeutic opportunity in ER(+) breast cancer. The FASEB Journal. 2019;33(2):1644-1657
  165. 165. Welte T, Zhang XH, Rosen JM. Repurposing antiestrogens for tumor immunotherapy. Cancer Discovery. 2017;7(1):17-19
  166. 166. Lin Z, Zhang X, Zhao F, Ru S. Bisphenol S promotes the cell cycle progression and cell proliferation through ERalpha-cyclin D-CDK4/6-pRb pathway in MCF-7 breast cancer cells. Toxicology and Applied Pharmacology. 2019;366:75-82
  167. 167. Huang W, Zhao C, Zhong H, Zhang S, Xia Y, Cai Z. Bisphenol S induced epigenetic and transcriptional changes in human breast cancer cell line MCF-7. Environmental Pollution. 2019;246:697-703
  168. 168. Gayrard V, Lacroix MZ, Grandin FC, Collet SH, Mila H, Viguie C, et al. Oral systemic bioavailability of Bisphenol A and Bisphenol S in pigs. Environmental Health Perspectives. 2019;127(7):77005

Written By

Boris Guyot and Veronique Maguer-Satta

Submitted: 29 August 2019 Reviewed: 25 October 2019 Published: 10 December 2019