Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Nov 2.
Published in final edited form as: Crit Rev Food Sci Nutr. 2017 Nov 2;57(16):3384–3404. doi: 10.1080/10408398.2015.1126547

Risks and benefits related to alimentary exposure to xenoestrogens

Ilaria Paterni a, Carlotta Granchi a, Filippo Minutolo a,b,*
PMCID: PMC6104637  NIHMSID: NIHMS1501865  PMID: 26744831

Abstract

Xenoestrogens are widely diffused in the environment and in food, thus a large portions of human population is worldwide exposed to them. Among alimentary xenoestrogens, phytoestrogens (PhyEs) are increasingly being consumed because of their potential health benefits, although there are also important risks associated to their ingestion. Furthermore, other xenoestrogens that may be present in food are represented by other chemicals possessing estrogenic activities, that are commonly defined as endocrine disrupting chemicals (EDCs). EDCs pose a serious health concern since they may cause a wide range of health problems, starting from pre-birth till adult lifelong exposure. We herein provide an overview of the main classes of xenoestrogens, which are classified on the basis of their origin, their structures and their occurrence in the food chain. Furthermore, their either beneficial or toxic effects on human health are discussed in this review.

1. Introduction

Xenoestrogens are estrogen mimicking compounds which are not produced by our body. These compounds are structurally or functionally related to human sex hormone 17β-estradiol (E2) and bind the estrogen receptors (ERs) with various degrees of affinity and selectivity. Due to their interaction with the ERs, xenoestrogens cause several effects on our organism, which sometimes may be beneficial for our health, whereas in most cases these effects are detrimental. Our exposure to xenoestrogens is mostly related to food ingestion, because these compounds may be present in the aliments, as well as in packaging materials or food contaminants (Figure 1).

Figure 1.

Figure 1

Human exposure to alimentary sources of xenoestrogens.

Xenoestrogens may be of either synthetic or natural origin (Figure 2). The most abundant class of natural xenoestrogens is represented by phytoestrogens (PhyEs). PhyEs are plant-derived compounds often exerting a wide range of biological activities, including estrogenic activity, which are found in a variety of foods, most notably soy. Among natural xenoestrogens there are also mycoestrogens, which are substances produced by fungi. Synthetic xenoestrogens are instead molecules produced by chemical synthesis, which are widely used in industrial products, comprising also pharmaceutical estrogens. These synthetic compounds can interfere with the endocrine system and, therefore, cause several health problems. In fact, they are classified as endocrine disrupting chemicals (EDCs). EDCs are mostly man-made compounds, found in various materials such as pesticides, metals, additives or contaminants in food and personal care products. EDCs are defined as compounds that "interfere with the synthesis, secretion, transport, binding, action, or elimination of natural hormones in the body that are responsible for the development, behavior, fertility and maintenance of homeostasis" (Crisp et al., 1988).

Figure 2.

Figure 2

Classification of xenoestrogens.

According to the World Health Organization (WHO), EDCs are suspected to be associated with altered reproductive function in males and females, increased incidence of breast cancer, abnormal growth patterns and neurodevelopmental delays in children, as well as changes in immune function (http://www.who.int/ceh/risks/cehemerging2/en). Human exposure to EDCs can occur via ingestion of food, dust and water, inhalation of gases and particles in the air and through the skin. EDCs can also be transferred from pregnant women to developing fetus or to children through the placenta or breast milk, respectively. It is important to note that also PhyEs may act as EDCs.

Considering that man-made chemicals, comprising estrogenic compounds, have become a part of everyday life, in 2012 the WHO developed the document "State of the Science of Endocrine Disrupting Chemicals-2012", in collaboration with the United Nations Environment Programme (UNEP), in which the key points of this important field are summarized (WHO/UNEP, 2013). It is now widely known that most humans are continually exposed to these potentially dangerous substances. This is proven by the increasing incidence of many endocrine related disorders, comprising genital malformation, neurobehavioral disorders and the increased global rates of endocrine-related cancers.

This review reports on the most recent information about food-related xenoestrogens, with a particular focus on: 1) the potential benefits/risks related to PhyEs intake; 2) the risks linked to exposure to estrogenic EDCs (Tables 1 and 2).

Table 1.

Natural xenoestrogens: Biological activities and sources.

Natural
Xenoestrogens
Compounds Activities1 Sources2

Phytoestrogens Lignans3 Lariciresinol Cardioprotective (clinical trial) berries, seeds, grain, nuts, fruits and cruciferous vegetables
Matairesinol
Pinoresinol Reduced risk breast cancer (in vitro)
Secoisolariciresinol

Flavonoids4 3-(2',4'-Dihydroxybenzyl)-5,7-dihydroxy-6-methylchroman-4-one In vitro estrogenic activity (ERβ) various4

3-(4'-Dihydroxybenzyl)-5,7-dihydroxy-6-methylchroman-4-one In vitro estrogenic activity (ERβ) various4

3-O-Methylquercetin In vitro estrogenic activity (ERβ) Liriope platyphylla aerial part

6-(1,1-Dimethylallyl)genistein In vitro estrogenic activity Derived from genistein metabolism in plants

6-(1,1-Dimethylallyl)naringenin In vitro estrogenic activity Derived from naringenin metabolism in plants

6-C-Methylquercetin-3-methyl ether In vitro estrogenic activity (ERβ) Liriope platyphylla aerial part

7-Hydroxy-6,4'-dimethoxyisoflavone In vitro estrogenic activity Butea superba Roxb.

8-Prenylgenistein In vitro estrogenic activity Derived from genistein metabolism in plants

8-Prenylnaringenin In vitro estrogenic activity Derived from naringenin metabolism in plants

Apigenin Protective role in prostate cancer cells (in vitro) Parsley and mint
Protective role in colorectal cancer cells (in vitro)

Calycosin Inhibition proliferation MCF7 cells (Akt) (in vitro) various4

Daidzein Neuroprotective (in vitro) Soy
Protective role in prostate cancer cells (in vivo)

Daidzin In vitro estrogenic activity Pueraria Mirifica leaves

Genistein Neuroprotective (in vivo) Soy
Protective in AD (in vivo)
Protective in PD (in vivo)
Reduce symptoms of multiple sclerosis (in vivo)
Protective role in colorectal cancer (in vivo)
Protective role in prostate cancer (in vivo)
Osteoporosis (in vivo)
Increased breast cancer risk (in vitro)

Genistin In vitro estrogenic activity Pueraria Mirifica leaves

Glabridin In vitro estrogenic/antiestrogenic activity Glycyrrhiza glabra root

Kaempferol In vitro estrogenic activity (ERβ)

Kwakhurin In vitro estrogenic activity Pueraria Mirifica tuberous root

Liquiritigenin Antinociceptive activity (in vivo) Glycyrrhiza uralensis root

Naringenin Protective role in colorectal cancer (in vitro) Rhizoma drynarie
Protective in AD (in vivo)

Naringin In vitro estrogenic activity Rhizoma drynarie

Puerarin Protective in PD (in vitro) Pueraria Mirifica leaves

Quercetin Activate ERα in MCF7 cells (in vitro) Mango fruit, lovage and dill
Protective role in endometrial cancer (in vivo)

Scutellarin Protective in AD (in vivo) Soy, Erigeron bevicarpus, Zingiber officinalis and Amomum subulatum rox

S-Equol5 Protective role in prostate cancer (in vivo) Milk thistle
Osteoporosis (in vivo)
Menopausal symptoms (clinical trial)

Sylimarin Protective role in colorectal cancer (in vivo) various4

Chalcones Isoliquiritigenin Increased breast cancer risk (in vitro) Glycyrrhiza glabra

Other structure Bakuchiol In vitro estrogenic activity Psoralea corylifolia seeds

β-Thujaplicin Reduced breast cancer risk (in vitro) Chamaecyparis octusa

Coumestrol In vitro estrogenic activity (ERβ) various4

Ferutinin Osteoporosis (in vivo) Ferula hermonsis root

Glyceollin I Neuroprotective (in vivo) Soybeans (cultured with fungi)
Reduced breast and ovarian cancer (in vivo)6

Glyceollin II Neuroprotective (in vivo)
Reduced breast cancer risk (in vivo)6

Glyceollin III Neuroprotective (in vivo)
Reduced breast cancer risk (in vivo)6

Mangiferin ERα activation in MCF7 (in vitro) Mango fruit

Medicarpin Osteoporosis (in vivo)

Norathyniol ERβ activation in MCF7 cells (in vitro) Mango fruit

Oleocanthal In vitro estrogenic/antiestrogenic activity Olive oil

Pterostilbene Neuroprotective (in vitro) Grapes and berries

Mycoestrogens Zearalenone False pregnancy (in vivo) Contaminant in corn, oats, wheat, rice (produced by Fusarium species)
Decreased fertility (in vivo)
Negatively affects male reproductive system (in vitro)

Drugs of abuse Δ9-Tetrahydrocannabinol Affects ERα/ERβ ratio (in vitro) and reproductive behavior. Marijuana
1

The activity is mediated by interaction with ERs.

2

For details, see USDA database. The origin is reported when specified in the reference paper.

3

Metabolized to active enterolignans (enterolactone and enterodiol).

4

Generally found in: soybean, plants, berries, wine, seeds, grains, nuts and legumes.

5

Metabolized from daidzein by bacterial flora in the intestine.

6

Tested as a mixture of Glyceollin I, II and III.

Table 2.

Synthetic xenoestrogens: Biological activities and sources.

Synthetic Xenoestrogens Activities Sources

4-tert-Octylphenol Negatively affects pregnancy and development Generally found in plastic and, consequently, as contaminants in foodstuff, fruits and vegetables.

Benzylbutylphtalate Negatively affects pregnancy and development

Bisphenol A (BPA) Correlated with hormone related cancer
Alteration neuroendocrine system
Altered development observed in aquatic species
Negatively affects pregnancy and development

Bisphenol A glucuronide Generates adipogenesis (in vivo)

Bisphenol S Cardiotoxicity

Di-(2-ethylhexyl) phthalate Negatively affects pregnancy and development

Dibutyl phtalate Negatively affects pregnancy and development

Terephtalic acid In vitro estrogenic effect

Tetrachlorobisphenol A More toxic than BPA, Potentially carginogenic

Ethynil estradiol Reduced fertility and fecundity Pharmaceuticals (found in aquatic environment)
Behavior changes in aquatic species

Bytylparaben Personal care products1

Ethylparaben In vitro estrogenic activity

Methylparaben Increased breast cancer risk

Butylated hydroxyanisole In vitro estrogenic/antiestrogenic activity Food preservatives

Benzophenone-2 In vitro estrogenic activity UV Filters in cosmetic and topical sunscreens1

4-Methylbenzylidene Camphor Highly diffused in aquatic environment

Polychlorinated biphenyls Neurological and hormonal diseases Used as coolant, plasticizers and pesticides and found in several food supplies

Chlordecone Associated with hormone related cancer Pesticides (fruits and vegetables)

Dichlorodiphenyldichloroethylene

Dichlorodiphenyltrichloroethane Negatively affects pregnancy and development

Hydroxyphenyltrichloroethane

Methoxychlor

TCDD Carcinogenicity, hepatotoxicity, immunotoxicity reproductive and developmental toxicity Environmental contaminant
1

Direct contact to human and indirect exposure through aquatic environments.

2. Natural xenoestrogens

2.1 Phytoestrogens (PhyEs): general features

Phytoestrogens (PhyEs) are natural compounds structurally and/or functionally correlated with E2. Interest in the potential role of PhyEs in preventing or treating diseases is demonstrated by the increasing number of papers that investigate the amounts of various PhyEs in food and plants. Most PhyEs are estrogen receptor ligands with a preference for the beta subtype (see, for example, Genistein β/α = 20, Daidzein β/α = 7, S-Equol β/α = 32, Coumestrol β/α = 7, Naringenin β/α = 11, Apigenin β/α = 20) (Minutolo et al., 2011; Paterni et al., 2014). Considering their chemical structures, PhyEs can be divided into different classes: lignans (Figure 3), flavonoids (flavones, flavonols, flavanones, isoflavones, isoflavandiols, homoisoflavones) and chalcones (Figure 4) (Sirotkin and Harrat, 2014). However the structures of PhyEs span a wide range of chemical classes and, therefore, some of them cannot be ascribed to any of the previously mentioned classes (Figure 5).

Figure 3.

Figure 3

Chemical structures of lignans and their metabolites enterolignans.

Figure 4.

Figure 4

Chemical structures of flavonoids and chalcones.

Figure 5.

Figure 5

PhyEs with different chemical structures.

Lignans are polyphenolic components of plant cell walls and are found in berries, seeds, grains, nuts, fruits and cruciferous vegetables. They derive from phenylalanine via dimerization of substituted cinnamic alcohols. Secoisolaricinesol, matairesinol, lariciresinol and pinoresinol are four examples of lignans (Figure 3). Once ingested, they are metabolized by intestinal bacteria to enterolignans, such as enterodiol and enterolactone (Figure 3), which exert an estrogenic activity, albeit their effect is considerably weaker than that of estradiol (Rowland et al., 2003) .

Flavonoids, which represent the main class of PhyEs, can be found in berries, wine, grains, nuts, soybean, legumes, etc (Figure 4). Their content in selected foods are reported in several accessible databases (United States Department of Agriculture, 2008; United States Department of Agriculture, 2011). Flavonoids are often conjugated with glucose or other carbohydrate moieties in their inactive form, whereas their bioactive form is represented by the un-conjugated compound (aglycone). Chalcones, which are flavonoid-related compounds lacking the heterocyclic C ring of flavonoids, can also be found in several edible plants and fruits (Figure 4). One of the most important source of estrogenic flavonoids is soy, containing relevant amounts of isoflavones genistein and daidzein, and the great interest for this nutrient is demonstrated by the increased number of soy-based products. Plants have always constituted an important source of flavonoids exerting estrogenic activity, as demonstrated by the high isoflavonoids content of Pueraria Mirifica leaves, in particular of puerarin, daidzin, genistin, daidzein, and genistein (Jungsukcharoen et al., 2014). Flavonoids obtained by the ethanolic extract of Liriope platyphylla aerial parts comprised about 17 already known PhyEs. All these compounds were evaluated for their estrogenic activity using a reporter-gene assay system, and two flavones, 3-O-methylquercetin and 6-C-methylquercetin-3-methyl ether, resulted the compounds with the most potent dose-dependent estrogenic activity. Moreover, these two compounds, together with another flavone, kaempferol, showed a high preferential affinity for the ERβ subtype in binding assays. Furthermore, two homoisoflavones, 3-(4'-hydroxybenzyl)-5,7-dihydroxy-6-methylchroman-4-one and 3-(2',4'-dihydroxy-benzyl)-5,7-dihydroxy-6-methylchroman-4-one (Figure 4), in which the hydroxyphenyl group in position 2 of isoflavones is replaced by an hydroxybenzyl group in position 3 of the chromanone scaffold, displayed preferential binding affinities for ERβ (Tsai et al., 2015). Butea superba Roxb., bearing to the Legouminosae family, is commonly found in deciduous Thai forest and it is used by Thai indigenous for promoting male potency, becoming a traditional medicine used to promote male vigor. A recent study evaluated the behavior of six previously isolated components of this plant and, in addition, tested 23 different tuberous ethanolic samples, which were collected from 23 different provinces in Thailand, for their estrogenic and antiestrogenic activity. Among the six single components tested, 7-hydroxy-6,4'-dimethoxyisoflavone resulted the most potent estrogenic component, with a relative potency of 15% on ERα and of 5% on ERβ. Then, the screening of the 23 tuberous ethanolic samples of Butea superba Roxb. revealed that practically all of them displayed a certain estrogenic activity, with a slight preference for the beta receptor subtype (Cherdshewasart et al., 2010).

Recently, some PhyEs extracted from mango fruit, such as mangiferin and quercetin, were found to selectively activate ERα in breast cancer MCF-7 cells, whereas norathyriol, which is a metabolite of mangiferin, activates also ERβ (Wilkinson et al., 2015). Oleocanthal, a bioactive compound found in olive oil, showed to bind to both estrogen receptors, inducing either agonistic or antagonistic effects depending on the cell line. This differential modulatory action could in part explain its wide range of pharmacological activities, such as anti-inflammatory, antiproliferative, and neuroprotective properties (Keiler et al., 2015).

Plants also produce second metabolites starting from some of the above mentioned PhyEs. One of the main metabolic reaction in plants is prenylation, which produces second metabolites of genistein, such as 8-prenylgenistein (8PG) and 6-(1,1-dimethylallyl)genistein (6DMAG), and of naringenin, such as and 8-prenylnaringenin (8PN) and 6-(1,1-dimethylallyl)naringenin (6DMAN). The activity of these compounds was evaluated in three functionally different estrogen receptor assays (YEAST assay, MVLN luciferase assay and Alkaline phosphatase activity of Ishikawa cells). Naringenin metabolites 6DMAN and 8PN showed an enhanced estrogenic activity compared to that of naringenin, whereas genistein derivatives 6DMAG and 8PG generally displayed a weaker estrogenic potency than that of genistein (Kretzschamar et al., 2010). In another study, kwakhurin, another prenylated PhyEs isolated from the tuberous root of Pueraria Mirifica, showed a moderated estrogenic activity, comparable to that of daidzein (Chansakaow et al., 2000). However, is important to note that the introduction of a prenyl bulky group in the structure of these PhyEs metabolites may also change their functional properties in terms of agonistic/antagonistic activities. For example glabridin, an isoflavandiol found in the root extract of licorice (Glycyrrhiza glabra) shows a phytoSERM profile, since it behaves as an antagonist or agonist, depending on the cell lines utilized in the functional assays (Simons et al., 2012).

Genomic analyses revealed that most PhyEs exhibit a wide variety of patterns of gene regulation, which may be either similar or distinct from each other and from that of E2. For example, the expression patterns of genistein and S-equol clustered together quite closely and were distinctly different from that of liquiritigenin, whose profile is the most different from that of E2 (Gong et al., 2014).

The content of flavonoids PhyEs in 16 different types of bean, which are commonly present in the daily diet of several countries, was analysed. High levels of genistein, daidzein and quercetin were found at different concentrations depending on the species (de Lima et al, 2014). Some common culinary herbs constitute excellent sources of flavonoids, such as apigenin (in parsley and mint) or quercetin (in lovage and dill) (Justesen et al., 2001). In spite of the fact that natural rice generally produces low levels of PhyEs, new studies led to the production a transgenic rice expressing high levels of flavonoids (in particular genistein, naringenin kaempferol and apigenin), thanks to the introduction of their biosynthetic genes (Ogo et al, 2013).

Many components of traditional Chinese medicine (TCM) are based on their estrogenic activities. In fact plants that are commonly used in TCM contain a wide variety of PhyEs. In particular, the TCM botanical formulation MF101 contains bakuchiol, a meroterpene phenol extracted from the seeds of Psoralea corylifolia, naringin and its active metabolite naringenin, two compounds extracted from Rhizoma drynariae (Gu Sui Bu), and flavanone liquiritigenin, the most active estrogenic compound from licorice root (Glycyrrhizae uralensis) (Li et al., 2015). In addition, a recent study demonstrated the estrogenic activity of some classical components of TCM, which proved to sustain bone growth and maturation without affecting other estrogen-dependent tissues (Tiosano et al., 2014). Finally, a terpenoid PhyEs, ferutinin, which was first extracted from Ferula hermonis root (Abourashed et al., 2001) was found to possess beneficial estrogenic effects on bone loss (Palumbo et al., 2012).

It is worth nothing that increasing efforts have been done in recent years in order to improve the efficiency of the methods used to detect PhyEs in food and plants (Wang et al., 2014; Li et al., 2013). Furthermore the amount of PhyEs is significantly affected by the manufacturing processes. For example, TPC (total phenolic content) and TFC (total flavonoids content) increased after fermentation of the plant-derived materials, and, for example, this is a popular processing method utilized to improve the texture and nutritional properties of soybean products. Even though fermentation could cause the breakdown of some healthful substances during heat treatment, soaking, and ripening, it is still a cost-effective method that can produce higher amounts of antioxidant and nutritious substances, such as the aglycone-forms of isoflavones (Xu et al., 2015).

2.2 Therapeutic applications of PhyEs as nutraceutics

Currently, there is a great debate about the use of PhyEs as useful preventive or therapeutic compounds in several diseases, such as menopausal symptoms, osteoporosis, cardiovascular pathologies, and different types of cancer, as shown by the increasing number of studies and clinical trials concerning their uses, although contrasting results have so far been obtained. Moreover, it is important to mention that over-consumption of PhyEs and their derivatives might produce endocrine-related side effects, especially on the uterus (Barals et al., 2014).

It should be noticed that, while several papers describe the potential benefits of PhyEs, there is a general lack of information about their side effects (Patisaul and Jefferson, 2010). In particular, the genotoxicity of several PhyEs needs to be carefully considered. Practically, all PhyEs (or their metabolites) exhibit pro-apoptotic effects in some cell systems, but this aspect is far from being well understood. Furthermore, the variable concentrations of extracts can provoke different responses in assays. In fact, food extracts are often found to be devoid of genotoxic effects in in vitro assays, despite the clearly established genotoxic activity of their isolated pure phytoestrogenic compounds (Stopper et al., 2005). Nevertheless, the beneficial actions of alimentary PhyEs generally outrun their risks, and the next section illustrates their various potential therapeutic applications.

2.2.1 Beneficial effects on the Central Nervous System (CNS)

Neuroprotective role of PhyEs mediated by ERβ activation was investigated in several studies. Daidzein proved to be neuroprotective by inhibiting the pro-apoptotic and neurotoxic effects caused by glutamate treatment in hippocampal cells. This effect seems to be dependent on ERβ activation, since it was reversed when cells were pre-treated with a selective ERβ antagonist, while the administration of a selective ERα antagonist did not influence the anti-apoptotic activity of daidzein. The membrane associated estrogen receptor GPR30 is also involved in the neuroprotective action of daidzein (Kajta et al., 2013). Another positive role of ERβ in neuroprotection was highlighted in a study where genistein was evaluated in a model of global cerebral ischemia in gerbilis. Administration of genistein proved to be neuroprotective, to reverse ischemia-induced memory impairment, to decrease malondialdehyde overproduction in the brain and to fully promote the survival of pyramidal cells in the CA1 hippocampal subfield. Interestingly, these effects are fully prevented by the administration of an ERβ antagonist 30 minutes before carotid occlusion (Donzelli et al., 2010). Furthermore, genistein and daidzein proved to be neuroprotective in stroke-like injury in vitro in an ER-dependent manner (Schereihofer et al., 2009). Dietary soy and soy isoflavones were neuroprotective in experimental cerebral ischemia. Ovariectomised Sprague-Dawley mice were divided into two groups, one fed with a soy-free diet and another one with a diet containing high levels of soy PhyEs for 5 weeks. Soy intake was associated with a reduced inflammatory response in the cerebral cortex after transient middle cerebral artery occlusion (Shambayati et al., 2014). Among PhyEs, pterostilbene (PTER), a dimethylated analogue of resveratrol found in grapes and berries, exhibits a neuroprotective role in H2O2-treated SY5Y cells, mainly through ERα stimulation, which presumably involves a positive regulation of anti-apoptosis Bcl-2 expression and the activation of the MAPK/ERK and PI3K/AKT signaling pathways (Song et al., 2015).

Antinociceptive activity of PhyEs liquiritigenin was assessed in a rat model of persistent neuropatic pain. Intraperitoneal administration of liquiritigenin alleviated in a dose-dependent manner mechanical, thermal and cold hyperalgesia. Moreover, antinociceptive tolerance was not observed after daily administration (Chen et al., 2014).

PhyEs are useful also in the treatment of depression, as demonstrated by a study comprising 40 women. These patients were divided into 4 groups and treated respectively with fluoxetine (10 mg), soybean (100 mg), sertraline (50 mg), and sertraline (50 mg) plus soybean (100 mg). Soybean showed an antidepressant effect per se, and the association of soybean and antidepressants proved to be synergistic (Estrella et al., 2014).

Great interest is currently focused on the use of PhyEs in the prevention and treatment of Alzheimer’s disease (AD), which is characterized by a neuronal dysfunction and by the presence of amyloid plaques in the brain. Many evidences highlight that the intake of PhyEs may prevent the AD. Moreover the increased frequency of the disease in post-menopausal women suggests a role for estrogens in development of AD. ERs regulate the expression of insulin-degrading enzyme (IDE), which plays a significant role in the catalysis of the beta amyloid protein. Among ERs, ERβ seems to have the most important role. In fact, hippocampal neurons treated with an ERβ agonist showed an increase in the IDE expression, which was completely abolished by the treatment with an ERs antagonist, whereas treatment with an ERα agonist did not produce any effect (Zhao et al., 2011). Intrahippocampal Aβ(1–40)-injected rats exhibited a lower spontaneous alternation score in the Y-maze spontaneous alternation test, an impaired retention and recall capability in the passive avoidance test, and fewer correct choices and more errors in the RAM task. Genistein significantly improved most of these parameters. However, when co-administered with fulvestrant, a potent ERs unselective antagonist, these beneficial results were reversed, thus demonstrating the involvement of the estrogenic pathway in the cognitive enhancement action of genistein (Bagheri et al., 2011). A phyto-β-SERM formulation composed of equal parts of soy-derived PhyEs (genistein, daidzein, and equol), displaying a 83-fold binding selectivity for ERβ over ERα, was found to ameliorate health, prolonged survival, improved spatial recognition memory, and attenuated amyloid-β deposition and plaque formation in the brain in a female triple transgenic mouse model of AD (Zhao et al., 2013). Scutellarin, the major flavone present in Herba Erigerontis, a Chinese medicine derived from Erigeron breviscapus (Zhu et al., 2009), Zingiber officinalis and Amomum subulatum Roxb. extracts (Kumal et al., 2015) proved to be active against AD symptoms. Genistein displayed a significant efficacy also against Parkinson disease (PD) (Baluchnejadmojarad et al., 2009). Another PhyEs showing efficacy against PD is puerarin, the major bioactive isoflavone isolated from the root of the Pueraria lobata (Willd.) Ohwi, which is well known as Gegen (Chinese name) in TCM. Puerarin has multiple pharmacological activities, but its protective effect against PD seemed to due to an antagonistic effect on estrogen-response element (ERE)-reporter transcription via binding to ERβ (Zhou et al., 2014). Some PhyEs showed efficacy also in reducing the symptoms of multiple sclerosis. Experimental allergic encephalomyelitis (EAE) is an autoimmune disease validated as an animal model of multiple sclerosis, genistein reduced EAE severity in mice only if administration started in the early phases of the disease (Jahromi et al., 2014). Despite these positive results, observational studies and randomised controlled trials in humans resulted in inconclusive findings about the effects of PhyEs on the cognitive functions of elderly people, and further studies are required (Soni et al., 2014).

2.2.2 Breast Cancer

One of the most important issues concerning the impact of PhyEs in human health is their ability to either increase or reduce the risk of developing breast cancer. The results so far obtained from preclinical studies are still controversial. Daidzein stimulates the growth of breast cancer cells and potentiates estrogen-induced cell proliferation in the uterus (Gaete et al., 2011). However, high concentrations of genistein inhibits the growth of breast cancer cells regardless of their ER status. On the other hand, low concentrations of genistein stimulate ER-positive breast tumors and abrogate tamoxifen-mediated growth inhibition in ER-positive breast cancer. Recent studies also demonstrated non-estrogenic anticancer effect of isoflavones (e.g., reactivation of epigenetically silenced tumor suppressor genes) (Kwon, 2014).

Chalcone isoliquiritigenin, a component of licorice extracts, behaves as an ER agonist in MCF7 cells, although its behavior was controversial and seemed to be dependent on the doses and on the exposure times. For example, high doses (> 10 µM) induce a dramatic drop in cell numbers, whereas long term exposure to low doses (< 1 µM) promotes cells growth through ERα activation (Maggiolini et al., 2002).

Some studies suggested a protective role of PhyEs against the development of breast cancer when their ingestion started prepubertally and it was associated with their ability to alter the morphology of the mammary gland during pubertal breast growth. PhyEs exposure during adolescence modulated the adult mammary gland sensitivity to E2 and lifelong exposure resulted in dose-dependent differential effects on proliferation, gene expression, and DNA methylation in rat mammary glands. Further studies will be necessary to clarify their effective role, considering that response varies from different race and ethnicity (Blei et al., 2015). The preventive role of PhyEs if assumed during adolescence was also supported by the result of a study showing that intake of PhyEs during adolescence among Ontario women was associated with a reduced risk of developing breast cancer (Anderson et al., 2013). Intake of lignans was also associated with a reduced risk of developing breast cancer (Abarzua et al., 2012), moreover enterodiol and enterolactone, alone or in combination with tamoxifen, seemed to reduce human breast cancer cell adhesion, invasion and migration in vitro (Chen and Thopmson, 2003). However, the estrogenic behavior of enterolignans is not fully clear. It was hypothesized that they may act as antiestrogens on estrogen receptor-positive breast cancer and, therefore, be beneficial in breast cancer prevention strategies (Bergman et al., 2007).

β-Thujaplicin, one of the major constituents in Chamaecyparis obtusa, exerts various health beneficial effects. A recent study demonstrated that it significantly suppresses the proliferation of MCF7 and T47D luminal subtype breast cancer cells by arresting the cell cycle transition from the G1 to the S phase. Interestingly, β-thujaplicin down-regulated ERα expression, which led to cell growth inhibition. These results suggest that β-thujaplicin might be considered as a potent agent that regulates the hormone sensitive mammary tumorigenesis (Ko et al., 2015).

Glyceollins are a family of soy-derived isoflavonoid phtyoalexins, whose production is increased in soybeans in response to various stress signals, such as fungal infection. The three major components of glyceollins identified in soybeans were Glyceollin I, Glyceollin II and Glyceollin III. Initially this glyceollins mixture was evaluated for its anticancer activity in a xenograft model of MCF-7 and BG-1 tumor formation in immunocompromised female ovariectomized nu/nu mice, showing an important antiestrogenic activity in vivo with a significantly suppressed estrogen-stimulated tumor growth of MCF-7 and BG-1 cells in ovariectomized female nude mice, and no agonistic effect in the uterus (Salvo et al., 2006). Then, these three components were purified and investigated separately: the active component of this mixture resulted to be glyceollin I, with an IC50 value of 1.68 µM against ERα, whereas the IC50 values of glyceollins II and III were 6.57 and >10 µM, respectively. Thus, this study demonstrated that glyceollin I acts as a potent ERs antagonist (Zimmerman et al., 2006). Moreover, further studies attributed the antiestrogenic activity of glyceollin I to the natural (−)-glyceollin I enantiomer, which was compared to those of the racemate and of the unnatural (+) enantiomer (Payton-Stewart et al., 2010). Furthermore, Glyceollin I proved to reduce E2-stimulated proliferation of MCF7 cells and E2-stimulated breast and ovarian cancer in vitro, and in vivo reduced E2-stimulated breast and ovarian cancer and E2-induced uterus growth, in an ERs dependent manner (Salvo et al., 2012). Although glyceollins seem to act as ER antagonists, in another recent study from a different research group, glyceollin I, II and III were tested as a mixture and they were found to behave as agonists with a preferential stimulation of ERβ (Kim et al., 2010). Glyceollins seem to be active also in central nervous system where they display neuroprotective effects (Bamji et al., 2015).

Finally, it is important to consider that PhyEs can exert their anticancer activity also by interfering with targets that are different from the ERs; for example, genistein and calycosin, inhibit the growth of MCF7 cells through their regulation of Akt signaling pathways (Chen et al., 2015; Chen et al., 2014).

Overall, despite the large number of studies reported during the past few years in this area, both in vitro and in vivo studies are still controversial, and postmenopausal women with high risk of developing breast cancer should avoid PhyEs intake, due to their possible capacity to stimulate proliferation in sensible tissue (Helle et al., 2014).

2.2.3 Endometriosis and endometrial cancer

Endometriosis is a hormone-related disease in which ERs are involved. PhyEs quercetin was used in an endometriosis rat model and it proved to modify the levels of ERs and progesterone receptor (PR) in hypothalamus, pituitary and endometrium, thus inhibiting estrogen and progesterone binding to their receptors. Quercetin regulates ERs and PR both in the reproductive system and in the CNS, leading to a decrease of serum estradiol. This caused the deficiency of hormone supplement in ectopic endometrium and reduced the height of endometrial epithelial layer and the number of ectopic endometrial glands, eventually leading to the atrophy of ectopic endometrial epithelial (Cao et al., 2014). A protective role of PhyEs in endometrial cancer is supported by the results of a clinical trial, where a multiethnic cohort study evaluated endometrial cancer risk in postmenopausal women after soy intake. This study showed that the risk of developing endometrial cancers was decreased in postmenopausal women with the higher intake of soy (in particular daidzein and genistein) (Ollberding et al., 2012).

2.2.4 Prostate cancer

PhyEs intake seems to be correlated also with a reduced risk of prostate cancer (PCa) in Asian populations. Genistein, daidzein and equol, an isoflavandiol metabolized from daidzein by bacterial flora in the intestines and prevalently found as S-enantiomer in humans, inhibit prostatic carcinogenesis in some animal models. ERα expression was upregulated in the epithelial cells in high-grade prostatic intraepithelial neoplasia (HGPIN), and most likely mediated the effects of estradiol in promoting prostate carcinogenesis as demonstrated in animal models. This role of ERα was further supported by the ability of the ERα antagonist toremifene to reduce the incidence of PCa in men with HGPIN. Inhibition of PCa epithelial cell growth is one of the most recognized protective roles of soy isoflavones in PCa. Moreover, genistein reduced the expression of ERα in two animal models. Furthermore, most PhyEs are ERβ-selective ligands and this could be useful in the chemoprevention of prostate cancer, where ERβ seems to play an anti-proliferative and proapoptotic role. In spite of these evidences, it is also important to note that other mechanisms, including antioxidant defense, DNA repair, inhibition of angiogenesis and metastasis, potentiation of radio- and chemotherapeutic agents, might contribute to the soy-mediated cancer prevention (Mahmoud et al., 2014).

A protective role of ERβ against prostate cancer was also associated to ingestion of apigenin, a dietary flavonoid with proteasome-inhibitory activity. Apigenin induced apoptosis in prostate cancer cells (PC3), without affecting the mitochondrial membrane potential of the cells, showing specifically chymotripsin-like activity of proteasome with no activity on tripsine and caspase-like activity. This was reflected by the low toxicity against normal prostate cancer cells (RWPE-1). This activity influenced the expression of both ERs in PC3 cells. In fact, ERβ is usually downregulated in prostate cancer. Apigenin increased in a dose-dependent manner the expression of ERβ and downregulated the expression of ERα. Apigenin displayed a very interesting pharmacological profile and can now be considered as a lead compound for targeting prostate cancer (Singh et al., 2014). Rat with testosterone-induced prostatic hyperplasia were treated with Pueraria mirifica (PM) extract, which contains daidzenin and genistein, and a reduction of the increase in the prostate/body weight ratio was found after treatment, showing a significant inhibition of benign prostatic hyperplasia (Masrudin and Mohamad, 2014). These two compounds were found also to be efficient in reversing DNA methylation in prostate cancer cell lines, such as PC3, DU145 and LNCaP. Interestingly, genistein and daidzein induced a demethylation of all the promoter regions studied, with the exception of those that were unmethylated in control cells, and this action was found to be probably mediated by ERβ (Adjakly et al., 2012). Recently, beverage Haelan 951, containing genistein and daidzein, displayed some promising effects on the viability, growth and apoptosis of different human pancreatic cancer cell lines (Rothe et al., 2015).

2.2.5 Colorectal cancer

A blend of ERβ agonists and lignin (Eviendep®, CM&D Pharma Limited, United Kingdom) was used in alimentary supplementation in familiar adenomatous polyposis (FAP) patients with ileal pouch-anal anastomosis (IPAA) for three months (5 mg each day at breakfast and dinner). FAP is a disease with autosomal dominant inheritance, patients are exposed to a high risk of developing duodenal adenomas. This study demonstrated that supplementation with Eviendep® for a short period (90 d) resulted effective in reducing polyps number by 32% and size by 51%. Moreover, different PhyEs contained in Eviendep® are in a lower dose than the effective dose needed for a single component to obtain the same effect, showing a synergistic and/or potentiating effect that is desirable for a long-term exposure (Calabrese et al., 2013). This result supports the protective role of ERβ in colorectal carcinogenesis. Genistein also showed a protective role against colorectal cancer in vivo. In fact, ovariectomized (OVX) female Wistar rats treated for three weeks with genistein showed a reduced proliferation in ileal and colonic mucosa cells, with an increased rate of apoptosis in the small intestine and colon. These effects were completely antagonized by ERs antagonist fulvestrant (Schleipen et al., 2011). In mice implanted with human colorectal cancer tumors, orally administered genistein did not inhibit tumor growth, but it did inhibit distant metastasis formation, and displayed no toxicity (Xiao et al., 2014). Apigenin and naringenin showed to be effective in the reduction of ER-mediated cell growth in non-malignant young adult mouse colonocytes (YAMC) and, interestingly, when administered in combination, they showed a strong enhanced activity with significantly lower concentration compared to the single administration (Yang et al., 2014). Sylimarin, a less common PhyEs extracted from milk thistle (Silybum marianum), showed protective properties against colorectal cancer. In fact, a reduction in the progression of colorectal cancer was observed in male mice fed with a sylimarin-enriched diet, together with an increase level of ERβ expression in the intestinal mucosa (Barone et al., 2010).

2.2.6 Osteoporosis

Increasing evidences underline that estrogen receptors, and in particular ERα, might play a crucial role in bone homeostasis. This is supported by the fact that many postmenopausal women are affected by this pathology, in which marked bone resorption is observed (Nilsson et al., 2011). The main strategy used in the treatment of osteoporosis is estrogen replacement therapy (ERT) (Sexton, 2001), but in recent years patients turned their attention to PhyEs composition, due to the fact that a long-term therapy, such as ERT, may induce a series of side effects related to the stimulation of ERα (Harris et al., 2002). Bakuchiol is a meroterpene phenol extracted from the seeds of Psoralea corylifolia, used to treat osteoporosis. This estrogenic component activates both ER subtypes, with a stronger binding affinity for ERα than for ERβ (Xin et al., 2010). Also medicarpin, a phytoalexin found in dietary legumes and structurally related to isoflavones, demonstrated to stimulate osteoblast differentiation and mineralization, by increasing ERβ expression in osteoblast. Medicarpin acts as an ER agonist in the bone, but, differently from ERT, it does not promote uterus growth. Administration of medicarpin to female Sprague–Dawley rats for 30 days resulted in an increased formation of osteoprogenitor cells in the bone marrow and osteoid formation (mineralization surface, mineral apposition/bone formation rates). Moreover, thanks to its excellent bioavailability, it may be considered as a very interesting option in osteoporosis treatment (Bhargavan et al., 2012). Another PhyEs that proved to inhibit bone loss in ovariectomized mice and rats is equol. Interestingly, all the positive effects of equol were abolished by an estrogen receptor antagonist (ICI182780), showing the implication of ERs in its effects (Wang et al., 2014). Genistein also proved to be potentially useful in the treatment and prevention of osteoporosis, since it partially reversed the detrimental effect of intermittent hypoxia on bone mass and bone strength in vivo (Song et al., 2014). Oral administration of ferutinin, a plant-derived ester of a sesquiterpenic alcohol, which acts as an ERα agonist (IC50 = 33.1 nM) and an agonist/antagonist for ERβ (IC50 = 180.5 nM), was able to improve bone regeneration of critical-size bone defects in vivo (Zavatti et al., 2015). Finally, a recent study revealed that in southern Chinese postmenopausal women the PhyEs dietary intake influences the bone mineral density, depending on the different ERα gene polymorfisms (Luo et al., 2015).

2.2.7 Menopause symptoms

There is a great debate about the use of PhyEs in menopause. In fact, up to now only few studies were carried out in order to evaluate the benefits and the potential side effects of PhyEs in postmenopausal women. For example, PhyEs were evaluated for the treatment of vasomotor symptom (VMS). A recent study evaluated the association of self-reported vasomotor symptom frequency with race/ethnicity among a diverse midlife US population and explored menopause symptom differences by dietary soy isoflavone (genistein + daidzein) consumption. This study confirmed that VMS expression varied by race/ethnicity. In US population it is difficult to evaluate whether these differences could be related to dietary soy intake, due to the extremely low soy intake. Interestingly, headache and pain were more common in American Indian women rather than in Asian Indian women, so VMS may also be related to the different perception of pain depending on different environments (Reed et al., 2013). Among PhyES, S-Equol was found to be effective in treating symptoms related to menopause such as vasomotor symptoms and osteoporosis (Jackson et al., 2011). S-Equol was used in a randomised, double-blind, placebo-controlled trial in Japanese women aged between 40 and 59 years. Significant improvements were observed in mood-related symptoms in equol-treated women (Ishiwata et al., 2009).

A nice review by Bedell et al. analysed the pros and cons of the use of PhyEs in menopause. The key point was that PhyEs were quite a new therapy in the treatment of menopausal symptoms and sometimes different studies produced contrasting results. The main evidence was that all the studies so far analyzed evidenced that PhyEs intake was devoid of serious adverse effects, although the real beneficial effects were far from be well-established (Bedell et al., 2014).

2.2.8 Cardiovascular diseases

Strong scientific evidence suggests that estrogens might play an important role in cardioprotection and cardiovascular diseases. The mechanisms involved in the cardioprotective role of estrogens are currently under investigation, and there is strong evidence supporting the involvement of both ERα and ERβ (Murphy, 2011). PhyEs have been considered for many years as important food components that are useful in preventing cardiovascular diseases, with favorable clinically effects on the low-density lipoprotein (LDL), which is considered to be one of the main responsible factors for the cardiovascular risk (Wroblewski et al., 2010). The analysis of several cohorts studies, which evaluated the association between flavonoids intake and incidence or mortality from cardiovascular disease, revealed that small amounts of flavonoid-based PhyEs may lower the risk of coronary heart disease mortality (Petersin et al., 2012). The association between urinary PhyEs levels and cardiovascular mortality risk was also evaluated, and it was found that enterolignan urinary concentrations were associated with a reduced risk of death from cardiovascular disease, whereas an opposite association was found for urinary concentration of total isoflavones (Reger et al., 2015). These results are consistent with previously published results, which confirmed that enterolignan intake was correlated with elevated high-density lipoprotein (HDL) levels and reduced triglyceride concentrations in US adults (Penelalco et al., 2012). However, it is important to note that that PhyEs metabolism could change among different race and ethnicity, hence these results should be interpreted with caution. The American Heart Association (AHA) does not recommend the use of isoflavone PhyEs supplements in food or pills for the prevention of cardiovascular diseases, since direct cardiovascular health benefits of PhyEs seems to be only marginal, whereas some possible undesired side effects may take place (Sacks et al., 2006).

2.2.9 Other diseases

Daidzein and genistein exerted a variety of effects on catecholamine signaling, including catecholamine synthesis, secretion, and uptake in the adrenal medulla. These findings may provide new insight into the pharmacological potentials of plant flavonoids on the catecholamine system. In particular, daidezein stimulated catecholamine synthesis through plasma membrane estrogen receptors in cultured bovine adrenal medullary cells, which were used as a model of central and peripheral sympathetic neurons (Yanagihara et al., 2014). Genistein proved to be effective against pulmonary hypertension (PH). PH was induced in male rats by using monocrotaline. In this model, genistein-based therapy resulted in significant improvements in lung and heart functions, moreover it reversed PH-induced pulmonary vascular remodeling in vivo and inhibited human pulmonary artery smooth muscle cell proliferation in vitro through ERβ (Matori et al., 2012). Interestingly PhyEs intake can also alter the metabolic profile as demonstrated in rainbow trouts (Oncorhynchus mykiss), where genistein and daidzein seem to produce estrogenic effects in liver and to affect growth performance and nutrient partitioning (Cleveland and Manor, 2015).

2.2.10 Interactions with other drugs

Use of soy supplements is increasing among patients, due to their perception that soy PhyEs may have positive effects on their health. It is important to mention that PhyEs can interfere with other drugs and, therefore, alter their therapeutic efficacy. A recent paper reports that PhyEs dramatically increased the cardiotoxicity in male mice receiving at the same time a chemotherapeutic tyrosine kinase inhibitor, such as Sunitinib. This combination produced an augmented lethality in vivo and an increased toxicity in isolated cardiomyocytes, when compared to the administration of Sunitinib alone (Harvey and Lainwand, 2015). Genistein intake was correlated with an enhanced multidrug chemoresistance in hepatocellular carcinoma (Rigalli et al., 2015). Pharmacokinetic interactions of genistein and imatinib were evaluated in rats and genistein seems to decrease imatinib plasma levels probably through effects on cytochrome CYP3A4; however the clinical significance of the pharmacokinetic interaction between imatinib and genistein still needs to be confirmed through further studies (Wang et al., 2015). Therefore, considering the increasing interest and consumption of PhyEs, further studies about PhyEs-drugs interactions are urgently needed, since it is now proved that PhyEs intake can alter the efficacy and the safety of several drugs.

2.3 Mycoestrogens: zearalenone

Food can be contaminated by naturally-occurring mycoestrogens, which are estrogens produced by fungi. One of the most widely diffused mycotoxin is zearalenone (Figure 6), a resorcyclic acid lactone produced by Fusarium species, in particular Fusarium graminearum and Fusarium culmorum, which is frequently found as contaminant in corn, oats, wheat, and rice. Zearalenone is a potent ERs agonist with an IC50 in the nanonomolar range (ERα-RBA = 4.3%, ERβ-RBA = 6.0%). Thus, zearalenone enters the food chain and represents a serious risk for both animals and humans (Takemura et al., 2007). Several adverse effects were associated to zearalenone exposure in animals. For example, cases of ovarian atrophy, prolonged estrus, persistent corpora lutea, false pregnancy, decreased fertility, stillbirth, implantation failure and delivery of weak piglets were observed in sexually mature female pigs (Malekinejad et al., 2005). Moreover, zearalenone was found to negatively affect the reproductive system of males, mostly by decreasing sperm counts and viability (Filannino et al., 2011). However, zearalenone is rapidly metabolized, in particular its glucuronidation represents an important and fast reaction of detoxification (Frizzel et al., 2015).

Figure 6.

Figure 6

Chemical structures of other natural xenoestrogens.

2.4 Drugs of abuse: Δ9-tetrahydrocannabinol

Several compounds, both of natural and synthetic origin, were found to bind the ERs as off-targets, thus representing an important problem due to the possible side effects related to their probable behavior as endocrine disruptors. An example is represented by Δ9-tetrahydrocannabinol (Δ9-THC, Figure 6), which is a biologically active component found in Marijuana, a widespread recreational drug of abuse and a recently recognized efficient therapeutic agent in support to cancer patients. Δ9-THC exhibited a variety of pharmacological and toxicological effects, such as analgesia, hypotension, reduction of inflammation and anticancer effects. However Δ9-THC is also well known for its psychotropic effects. Recently Δ9-THC was recognized to produce endocrine disrupting effects, in particular on reproductive behavior, by altering the ERα/ERβ ratio in cells, via an up-regulation of ERβ (Takeda, 2014). These evidences are important and, if confirmed, Δ9-THC should be considered as an established estrogenic EDC.

3. Synthetic xenoestrogens: Endocrine Disrupting Chemicals (EDCs)

The human organism is exposed to several substances, especially from the environment, which could cause severe adverse effects and pathologies, by acting as endocrine disrupting compounds. Among these substances one of the major class is represented by synthetic xenoestrogen (Figure 7). These compounds are able to bind to the ERs by means of various types of protein-ligand interactions, which affect their different affinity and selectivity. A recent correlative analysis study revealed that these chemicals, displaying binding affinities ranging from subnanomolar to micromolar values, act in a subtype-dependent manner as full agonists or partial agonists/antagonists, by using different combinations of the activation functions 1 and 2 of ERα and ERβ (Delfosse et al., 2015).

Figure 7.

Figure 7

Chemical structures of synthetic xenoestrogens.

Most of the synthetic xenoestrogens that may be found in the environment and, therefore, in food, are structurally related to natural estrogens and possess estrogenic activities. Among them there are, for example, synthetic hormones (such as ethynylestradiol EE2, present in birth control pills), compounds commonly used in personal care products due to their antibacterial properties (such as parabens), and pesticides. One of the most dangerous class is represented by bisphenols, synthetic compounds that are used in the production of certain plastics and resins (Delfosse et al., 2015). Several bisphenols were found to be estrogenic EDCs by binding to both receptor subtypes (Stossi et al., 2014). One of the most widely diffused and dangerous synthetic xenoestrogen is bisphenol A (BPA), which is widely produced and largely diffused in the environment and in foodstuff, such as plastic bottles and thermal paper. The increasing interest in studying BPA actions is due to the fact that BPA exposure may be associated to the development of several types of hormone-related cancers (Gao et al., 2015) and to the alteration of the neuroendocrine system, which could affect brain development and produce physiological/behavioral effects (Rebuli et al., 2014). To avoid these problems, nowadays BPA has been replaced by other bispenols, such as bisphenol S (BPS). However, also this derivative shows some cardiotoxicity in female rat hearts, just like BPA (Gao et al., 2015). In addition, BPA metabolites formation should be seriously taken into due consideration. In fact, BPA is rapidly converted in vivo to BPA-glucuronide (BPA-G), which constitutes its predominant metabolite. BPA-G has long been believed to be biologically inactive, because it lacks estrogenic activity. Nevertheless, it was recently shown that BPA-G induces adipogenesis, and this effect was not observed in the presence of ERs-antagonist fulvestrant, thus suggesting that BPA-G-generated adipogenesis follows an estrogenic pathway (Boucher et al., 2015). Halogenated bisphenols A (H-BPAs), such as tetrachlorobisphenol A (TCBPA), which are widely used in industrial production of several materials, were found to be present in various environmental matrices and detected in human serum and breast milk. Furthermore, they showed to be more potent toxicants than BPA itself (Song et al., 2014). Synthetic xenoestrogens are generally pervasive and widely dispersed in the environment and may bio-accumulate. In fact, these chemicals typically end up in either stormwater or wastewater, with a potentially serious impact on aquatic life and wildlife. This is a very important problem related to food contamination, including also fruits and vegetables, since vegetable fields are principally irrigated by reclaimed water, which often contains considerable amounts of these EDCs (Sidhu et al., 2015). Fruits and vegetable represent also one of the main human exposure sources to organochlorine pesticides, dangerous compounds related with increased risk of developing several hormonally-related cancers (Lerro et al., 2015). The increasing interest in the determination of these chemicals is due to consolidated evidences underlining their role in the pathogenesis of several diseases such as cancer, obesity (Yang et al., 2015), diabetes (Chevalier and Fenichel, 2015) and reproductive pathologies (Kumar and Holt, 2015). EDCs exposure is particularly dangerous during childhood. In fact it was estimated that EDCs exposures in the EU substantially contributed to the development of obesity and diabetes, with a moderate probability of €18 billion costs per year. These results further emphasize the need to control EDCs exposures and diffusion in the environment (Legler et al., 2015). Moreover, nearly €15 billion annual costs in the EU were associated with male reproductive disorders and diseases due to EDCs exposure. These estimates represented only a few EDCs for which there were sufficient epidemiological studies and those with the highest probability of causation (Hauser et al., 2015). However, one of the major costs/year was attributed to neurobehavioral deficits and disease derived from EDCs exposure, with a high probability of >€150 billion costs/year (Bellanger et al., 2015).

Several diseases, such as those affecting female and male reproductive systems, neurodevelopment, metabolic disorders and thyroid, were confirmed to be correlated to EDCs exposure by the WHO in 2012 (WHO/UNEP, 2013). Moreover, it nowadays evident that human fertility rates are declining all over the world, especially in industrialized countries. It is worthwhile mentioning that exposures to these substances during pregnancy may be really dangerous for the health of the fetus and produce negative effects during the growth of the child (WHO/UNEP, 2013). In addition, it is important to note that EDCs do not interact only with ERs, but also with a large panel of other nuclear receptors comprising the androgen receptor, the estrogen-related receptors and the retinoid X receptors (Grimaldi et al., 2015), therefore some of their effects on human health might not be necessarily due to the activation of estrogenic pathways.

Hormones were also found to be added illegally in some categories of food. For example, in recent years several emerging healthcare anti-aging products indicated to regulate blood pressure and blood glucose, contain substances, such as estradiol and 17α-ethinylestradiol, that are illegally used to improve the potency of these products (He et al., 2014).

Finally, 2,3,7,8-tetrachloro-p-dioxin (TCDD) is one of the most relevant member of the general class od dioxins, which are persistent environmental contaminants. TCDD is produced as a side-product during chemical synthesis, especially of some herbicides, or as a byproduct of combustion of some organic compounds. TCDD is able to produce estrogenic effects in various tissues, since it binds to the aryl hydrocarbon receptor (AhR), which then interacts with the ERs and elicits an AhR-mediated estrogenic action (Boverhof et al. 2006).

An important aspect related to monitoring EDCs exposure levels in humans is the reliability of the analysis of samples and the identification and quantification of EDCs. In fact samples are often constituted by complicated matrices, where initially the compound of interest must be extracted from, then isolated, identified and quantified. Several analytical methods are nowadays available to detect estrogenic EDCs, depending on the nature of the sample to analyze (milk, meat, miscellaneous matrices, fish, packaging materials, etc) in a cost-effective way, and increasing efforts in finding more sensible and accurate analytical methods are currently underway (Adamusova et al., 2014; Capriotti et al., 2013; Wang et al., 2008; Kudlaka et al., 2015; Rotroff et al. 2014).

3.1 EDCs in foodstuff

EDCs are widely diffused in food, as shown by the high number of studies on different food samples which confirm their presence in them. For example, BPA was detected in all vegetable and fruit samples including pumpkin, sweet potato, citrus, and apple from local markets (Lu et al., 2013). Fruits and vegetables can be exposed to synthetic xenoestrogens during the irrigations process, in which plants are irrigated with wastewater effluents and freshwater and treated with several types of biosolids. All these factors influence their content of estrogenic compounds. Biosolids had a minor effect on hormones content, whereas irrigation water, which contained significant amounts of hormones, had the most substantial effect for example on the EDCs content in lettuce (Shargil et al., 2015). In a separate study samples of food and milk infant formula were found to possess estrogenic activity, showing that estrogenic compounds were omnipresent in foodstuff and were not limited to soy-based food (Behr et al., 2011). Xenoestrogenic compounds may also be found among food preservatives. For example, butylated hydroxyanisole (BHA), an antioxidant consisting of a mixture of two regioisomeric organic compounds, 2-tert-butyl-4-hydroxyanisole and 3-tert-butyl-4-hydroxyanisole, is used as a food additive (E320). Contrasting results have so far been reported, since a study described BHA as an ERs agonist (Ter Veld et al., 2006), whereas another one considered it as an anti-estrogenic compound, which was able to decrease uterine weight in immature female rats (Kang et al., 2006).

Synthetic xenoestrogens are also present in food packages and can pass from package to food. A recent study analyzed food contact material samples for their (anti)-estrogenic and (anti)-androgenic activity. Estrogenic activity was detected in 7 of 10 of bottle water samples and in 44% of food packaging samples (Mertl et al., 2014).

BPA is widely present in thermal paper receipts. Nevertheless, a study analyzing BPA urine content in volunteers who normally handle BPA-containing thermal paper receipts (either professionally or as consumers) seems to indicate that this type of exposure has a limited effect on systemic BPA levels (Porras et al., 2014). In addition to BPA, also terephthalic acid (TPA), a major chemical precursor of polyethylene terephthalate (PET) containers that are used for food storage and beverages, was found to increase the ERα:ERβ ratio in human breast cells, suggesting an estrogenic effect. Moreover, in addition to its carcinogenic potential, TPA also promoted resistance to tamoxifen induced apoptosis (Luciani-Torres et al., 2015). These chemicals were found also in animal foodstuff, with most samples showing significant estrogenic activities and high BPA contents (Wang et al., 2014).

3.2 Synthetic xenoestrogens and personal-care products

Xenoestrogenic EDCs are broadly diffused in personal care products which are daily used by people. A class of compounds of interest, due to their possible estrogenic activity, comprises parabens, which are alkyl esters of p-hydroxybenzoic acid. Parabens are extensively used in cosmetics, pharmaceuticals, personal care products and also in food products due to their antimicrobial activity. In addition to food ingestion, exposure to parabens may occur through skin, lips, eyes, oral mucosa, nails and hair. Due to their efficient antibacterial and fungicidal properties, stability at different pH values and temperatures and low cost, there is a widespread chronic human exposure to parabens. Therefore, these compounds have been considered as endocrine disruptors to be controlled for their possible adverse effects. Parabens are considered as weak estrogenic agents and their estrogenic activity is considerably lower than that of 17β-estradiol, but it is not negligible, as demonstrated by several studies which reported their estrogenic activity in cells (Karpuzoglu et al., 2013). Rats and rabbits exposed to parabens (such as methyl, ethyl or butyl parabens) showed that these chemicals were taken up in considerable amounts, but then they were rapidly hydrolyzed and excreted. Nevertheless, these in vivo experiments underlined that daily sperm production and serum testosterone levels were negatively affected by parabens, which also produced uterotrophic effects in ovariectomised rats. Almost all the currently known parabens resulted positive in estrogenic assays. Estrogenic potency of these derivatives increases with the alkyl chain length. Moreover, long chain parabens also seem to be more toxic. It is important to note that parabens have also been used in baby and children products, thus representing a serious risk for their health and development (Bobjer et al., 2010). ERα expression was found to be increased in MCF7 cancer cells that had been exposed to parabens, thus confirming that these compounds actively contribute to breast cancer progression. Interestingly, parabens action was prevented by ERs blocker ICI182780 in MCF7 cells but not in non-cancerous breast cells, suggesting a different mechanism of action for these two cell lines (Wrobel and Gregoraszczuk, 2014).

Among personal care products, some estrogenic compounds are used as UV filters in cosmetics, topical sunscreens in lotions, and hair sprays to protect skin and hair from UV irradiation. For example, 4-methylbenzylidene camphor (4-MBC) is a widely used UV absorbing substance used in sunscreen products. It was demonstrated that 4-MBC binds to ERs, although its affinity is not extremely high (Klann et al., 2005). Nevertheless, 4-MBC is potentially dangerous for two reasons: 1) its estrogenic activity can directly be harmful to humans who abundantly use products where it is contained; 2) its dispersion in water and its consequent bioaccumulation in aquatic species poses a serious environmental concern. In fact, a high bioaccumulation of UV filters was demonstrated in seawater samples from beaches in Gran Canaria Island (Rodriguez Sanchez et al., 2015), and high levels of 4-MBT were found in fish from Iberian river basins (Gago-Ferrero et al., 2015). Another widely diffused UV filter is benzophenone 2 (BP-2). Here again, high levels of this compound were found in aquatic species. It is important to note that BP-2 exposure in fish produced estrogenic effects through ERα on vitellogenin induction, secondary sex characteristics, gonadal development and reproduction (Weisbrod et al., 2007).

3.3 EDCs in the environment and effects on wildlife and aquatic species

Considering the wide range of diffusion of estrogenic EDCs in the environment in the past few decades, it is not surprising that in recent years many researchers turned their attention to the study of the effects of these compounds in animal models. Aquatic habitat is highly contaminated by EDCs. These chemicals were detected in almost all aquatic sources tested and, most importantly, in many cases their contents were above bioactive concentrations. The effect of BPA exposure in aquatic species consisted in activation of the ERs, increase of brain aromatase activity, disruption of gametogenesis in both males and females, altered development (neuronal, cardiac, germ cells, and sexual differentiation) and changes in sex ratios after embryonic exposure (Bhandari et al., 2014). It should be noted that the effects of exposure to these chemicals may vary from species to species (Segner et al., 2013), and this could be partially explained by considering the differences in the receptor ligand binding domains and, consequently, in the different interaction of xenoestrogens with the ERs of each species (Miyagawa et al., 2014).

A recent study reported the presence of several bisphenol (BP) derivatives detected in different samples of sewage sludge collected from wastewater treatment plants in China. Estrogenic activity was detected by using a bioluminescence yeast estrogen screen (BLYES) assay and all the collected samples showed significant estrogenic activities. When the same assays were repeated in the presence of ICI182780, a potent ERs antagonist, a marked reduction in the bioluminescence was observed, indicating that BPs induced bioluminescence by activating the ERs. This study underlined that BPs are widely diffused in China representing an emerging problem about the presence of estrogenic contaminants that are potentially dangerous for human health (Ruan et al., 2015).

Pharmaceutical estrogens, such as ethynylestradiol (EE2), which is used in contraceptive formulation, currently represents an emerging environmental problem (Khan et al., 2014). The affinity of EE2 for the human ERs is considerably higher than that of estradiol. Moreover, due to its nonpolar and hydrophobic character, it is resistant to biodegradation and it is nowadays wildly diffused in the environment. The effects observed in exposed organisms were determined by the analysis of male and female fish plasma vitellogenin, an estrogen-inducible yolk precursor protein which is normally produced only in mature female fish. The results indicated progressively and time-dependent increased amounts of this protein, accelerated proportions of intersex fish, decreased eggs and sperm production, reduced gamete quality, completed feminization of male fish, reduced fertility and fecundity and also behavior changes. Although aquatic species in early life seemed to be more sensible to EE2 exposure, EE2 also proved to be extremely toxic to adult aquatic organism. Furthermore, vertebrates, including mammals, proved to be sensible to EE2 exposure, in particular during their postnatal development (Aris et al., 2014).

Dioxins, in particular TCDD, are persistent environmental contaminants with EDCs properties, which significantly affect wildlife. In addition to their direct interference with the endocrine system, which are mostly related to reproduction and pregnancy, they cause a wide variety of other toxic effects, such as carcinogenicity, hepatotoxicity, and immunosuppression (Radenkova-Saeva J. 2009). These effects are particularly dangerous because these compounds are very resistant to metabolism and biodegradation.

It should be noticed that aquatic species, such as fish, amphibians, aquatic reptiles and mammals, are most affected by EDCs exposures and their population is declining in contaminated areas. This phenomenon may be associated to the negative effects of EDC on fertility. However similar effects have also been observed in terrestrial species. This has a profound impact on humans, since we are at the top of the food chain and our eating habits may expose us to risky levels of EDCs.

3.4 Effects of EDCs on human health

Disruption of ER signaling by EDCs has been demonstrated to produce adverse effects on human health and, in particular, on fertility, pregnancy and fetus development. Hyperplasia of endometrial glands, dystocia, prolapse of the uterus and infertility were observed in sheep and in other ruminants species exposed to forages containing estrogenic contaminants (Bazer et al., 2014). One of the main class of compounds affecting pregnancy is represented by organochlorine pesticides. The effect of exposure to methoxychlor (MXC), a weak synthetic estrogen used as a pesticide with estrogenic, anti-estrogenic and anti-androgenic activities, was evaluated in male fetal and neonatal rats. In this study, MCX exposure during embryonic and postnatal development reduced body weight and bone size; moreover cortical porosity resulted negatively affected compared to control. MXC might act as an ERβ antagonist, thus inhibiting the proper mineralization of the developing skeleton, however significant differences may be observed in different species (Fagnant et al., 2014). In addition, it should be considered that the major metabolite of MXC, hydroxyphenyltrichloroethane (HPTE), is even more potent than MCX as an EDC (Uzumcu et al., 2006).

Another dangerous substance widely used as pesticide in last decades is dichlorodiphenyltrichloroethane (DDT), it was showed that stimulation of membrane estrogen receptor seemed to play an important role in the propagation of DDT-induced apoptosis during the early stages of neural development (Kajta et al., 2014). Among other chlorinated pesticides, dichlorodiphenyldichloroethylene (DDE) proved to negatively affect pregnancy. In fact the presence of DDE and other organochlorine pesticides in maternal blood and placental tissue represents a very dangerous prenatal exposure hazard for fetuses, due to chronic bioaccumulation and poor elimination, with possible deleterious effect on pregnancy and fetus health (Tyagu et al., 2015). Chlordecone, an organochlorine insecticide which has been widely used in the French West Indies until 1993, resulted in a long-term soil and water contamination. Chlordecone exposure through contaminated food during critical periods of development (gestation and early infancy) was found to negatively affect infant growth (Coste et al., 2015).

Another dangerous class of pesticides are the polichlorinated biphenyls (PCBs), industrial chemicals historically used as pesticides, coolants or heat transfer agents in electrical transformers, which showed estrogenic or anti-estrogenic activities (Zhang et al., 2014). Due to their elevated toxicity, PCBs use was banned in 1979. However, due to their widespread use, these compounds are still present in several food supplies (fish, dairy, hamburger, and poultry being the most contaminated) and in our bodies. In particular, PCBs exposure could cause long-term health problems, such as neurological and hormonal diseases, especially for those who were exposed in utero (Crinnion, 2011).

4-tert-Octylphenol, another EDCs extensively used in industry and widely diffused in the environment, displayed adverse effects on fat metabolism in pregnant rats, by inhibiting fat deposition via an interference with the regulation of lipogenic genes in the liver and adipose tissue. This alteration might affect the nutrition balance during pregnancy and can cause metabolism-related diseases, however the exact role of the ERs in these alterations was not investigated in more details (Kim et al., 2015).

BPA fetal exposure was studied in mice and it seemed to advance puberty onset, disturb estrous cyclicity, increase body weight gain, and cause fertility problems, which became more severe with age. Therefore, it is possible that BPA prenatal exposure in humans may affect the development and function of the uterus, which lead to early and late pregnancy losses (Wang et al., 2014). In addition, BPA exposure during early pregnancy affects placenta morphology and angiogenesis in mice. Embryonic structures (head, forelimb) also showed some alterations, potentially related to unbalanced embryonic nutrition and/or to modulation of genes involved in embryo development. These findings support the fact that placenta is directly involved in the effects of BPA, even at environmentally relevant dose levels (Tait et al., 2015). Moreover, the first emerging studies evaluating the effects of BPA exposure in several generations of mice underlined that some reproductive dysfunctions in female mice can be transgenerational in nature (Ziv-Gal et al., 2015).

Fetal exposure to BPA was evaluated by analyzing the correlation between placental BPA concentration, infant birth weight and other maternal and infant parameters in the southeastern USA. All placenta samples contained quantifiable amounts of BPA and fetal growth was found to be affected by this EDC. This evidence was found to be particularly alarming, since BPA detoxification is lower in fetus liver than in that of adults (Troisi et al., 2014). It should be considered that the effects of BPA exposure during pregnancy and early life may also appear only in adulthood. Perinatal exposure to BPA and to other EDCs can influence the behavior in adulthood, often leading to parental and social behavior problems. For example, rodents exposed to synthetic xenoestrogens (BPA and phtalates) during perinatal period showed compromised social behavior (Rosenfeld, 2015). Additional studies evaluated the correlation between prenatal BPA exposure and the behavior of children in school age. The negative effects linked to this exposure seem to be gender-dependent, with boys being more affected than girls. In particular, children exposed to BPA during pregnancy showed altered behavior with depression problems, somatic complications and oppositional/defiant behavior at age 6–10 (Evans, 2014). More recently, BPA exposure in early life was correlated with an altered susceptibility to tumor formation in particular sites, such as prostate and breast. This could be due to an epigenetic reprogramming of these tissues depending on the activation of a non genomic response by BPA and other EDCs (Trevino et al., 2015). BPA exposure during fetal life is currently being considered as a time bomb for prostate cancer. In fact, during embryonal development, prostate stem cells mainly have an epithelial structure and some of them persist during adulthood. It was observed that when these cells were exposed to estrogen-like endocrine disruptors, they become more prone to abnormal growth, leading to cancer (Lobaccaro and Trousson, 2014). BPA exposure was also evaluated in prepubertal and adult rats, alone or in combination with PhyEs genistein. Rats assumed BPA, genistein or a combination of both through lactation. Then cells apoptosis, proliferation and protein expression were investigated, with a particular focus on mammary gland modifications. BPA exposure increased cell proliferation in mammary gland of adult PND50 rats, a process associated with an increased risk of chemically-induced cancer development. On the other hand, genistein exposure stimulated cell proliferation in prepubertal rats, a process that correlates with mammary gland maturation and chemoprevention. Interestingly, the combination of both of them produces the same effect as that of genistein alone, which induces mammary gland maturation, thus suggesting mammary chemoprevention (Wang et al., 2014).

In addition to bisphenols, another chemical class of substances that are used as plasticizers and frequently found in several food products is represented by phthalate esters, which can exert adverse effects on human reproduction. Among them, dibutyl phtalate (DBP) is one of the most commonly diffused. DBP perinatal exposure significantly increases estradiol and decreased testosterone serum levels in immature offspring rats. It also up-regulates the aromatase level and down-regulated the expression of ERβ, which may cause neurotoxicity in immature offspring (Li et al., 2014). However, the interaction of DBP with the ERs is not completely understood yet (Rouiller-Fabre et al., 2015). In utero exposure to another plasticizer, butyl benzyl phthalate, induced a delayed pubertal onset and modified the morphology of the mammary gland. In addition, modifications in gene expressions that are associated with an increased susceptibility to carcinogenesis were observed (Moral et al., 2011). Interestingly, the metabolites levels of another phthalate, di-(2-ethylhexyl)phthalate, in urine proved to be related with obesity and insulin resistance in humans (Smerieri et al., 2015).

However, EDCs may also cause other types of pathologies, which are not strictly related to reproduction, such as hepatotoxicity, immunosuppression, type 2 diabetes, and cancer (Soto et al. 2010).

4. Conclusion

The wide diffusion of xenoestrogens in the environment and the high probability of exposure to them, especially in the food chain, make this class of compounds a urgent concern to be taken care of. PhyEs intake has rapidly increased in recent years, due to the growing number of marketed soy-based products, and supported by several evidences that underline the important role of PhyEs enriched food in health prevention. People generally prefer natural products to classical therapeutic treatments, perceiving PhyEs as safer and deprived of significant side effects, which are instead typical of standard therapies. Nevertheless, this general conviction misjudges the problems that may be encountered with PhyEs intake, since there are contrasting data about these products and, in particular, no clear results deriving from clinical trials are currently available. Moreover, the correlation of PhyEs efficacy with the race and ethnicity (Asian vs European) contributes to the questionable use of PhyEs. PhyEs may actually represent an important class of alternative therapeutic agents in several pathologies, however further information and studies are necessary to have a better understanding of the real benefits compared to the possible problems related to their use.

In general, EDCs’ influence in biological systems represents a global problem and interdisciplinary studies are increasing in order to evaluate the environmental fate, endocrine potential, metabolic pathways and toxic effects related to EDCs exposure. Human exposure to EDCs is inevitable and people are daily exposed to these chemicals through both the consumption of contaminated food and the use of industrial products containing them, most often in chronic low doses. The long-term exposure may adversely affect the endocrine system, leading to a negative impact on health and, unfortunately, this is far to be exactly established. Many sources of EDCs are still not known, because of insufficient reporting and information on chemicals in products, materials and goods and this lack of information worsens the present situation. EDCs have been widely found in materials used by infants and children, such as baby bottles, as well as in food and breast milk. This is a major problem since young people generally have higher exposure to chemicals due to their higher metabolic rates, higher amounts of ingested food, drink and air per body weight, when compared to those in adults, as well as higher hand-to-mouth and object-to-mouth activities. Moreover, it has been shown that exposure to EDCs in pre-birth or early life may sometimes manifest only later in life or even in future generations, making risk evaluations very difficult. Therefore, more scientific evidence is badly needed to identify the effects of EDCs in humans.

Acknowledgments

Support from the University of Pisa (Intramural funding) and from the National Institutes of Health (R01GM098453) are gratefully acknowledged.

References

  1. Abarzua S, Serikava T, Szewczyk M, Richter D, Piechulla B, Briese V. Antiproliferative activity of lignans against the breast carcinoma cell lines MCF 7 and BT 20. Arch. Gynecol. Obstet. 2012;285:1145–1151. doi: 10.1007/s00404-011-2120-6. [DOI] [PubMed] [Google Scholar]
  2. Abourashed EA, Galal AM, El-Feraly FS, Khan IA. Separation and quantification of the major daucane esters of Ferula hermonis by HPLC. Planta Med. 2001;67(7):681–682. doi: 10.1055/s-2001-17354. [DOI] [PubMed] [Google Scholar]
  3. Adamusova H, Bosakova Z, Coufal P, Pacakova V. Analysis of estrogens and estrogen mimics in edicble matrices- A Review. J. Sep. Sci. 2014;37:885–905. doi: 10.1002/jssc.201301234. [DOI] [PubMed] [Google Scholar]
  4. Adjakly M, Ngollo M, Lebert A, Dagdemir A, Penault-Llorca F, Boiteux JP, Bignon YJ, Guy L, Bernard-Gallon D. Comparative Effects of Soy Phytoestrogens and 17β-Estradiol on DNA Methylation of a Panel of 24 Genes in Prostate Cancer Cell Lines. Nutr. Cancer. 2012;66:474–482. doi: 10.1080/01635581.2014.884236. [DOI] [PubMed] [Google Scholar]
  5. Anderson LN, Cotterchio M, Boucher BA, Kreiger N. Phytoestrogen intake from foods, during adolescence and adulthood, and risk of breast cancer by estrogen and progesterone receptor tumor subgroup among Ontario women. Int. J. Cancer. 2013;132:1683–1692. doi: 10.1002/ijc.27788. [DOI] [PubMed] [Google Scholar]
  6. Aris AZ, Shamsuddin AS, Praveena SM. Occurrence of 17α-ethynylestradiol (EE2) in the environment and effect on exposed biota: a review. Environment International. 2014;69:104–119. doi: 10.1016/j.envint.2014.04.011. [DOI] [PubMed] [Google Scholar]
  7. Bagheri M, Joghataei M, Mohseni S, Roghani M. Genistein ameliorates learning and memory deficits in amyloid β(1–40) rat model. Neurobiol. Learn. Mem. 2011;95:270–276. doi: 10.1016/j.nlm.2010.12.001. [DOI] [PubMed] [Google Scholar]
  8. Baluchnejadmojarad T, Roghani M, Nadoushan MRJ, Bagheri M. Neuroprotective Effect of Genistein in 6-Hydroxydopamine Hemi parkinsonian Rat Model. Phytother. Res. 2009;23:132–135. doi: 10.1002/ptr.2564. [DOI] [PubMed] [Google Scholar]
  9. Bamji SF, Page RB, Patel D, Sanders A, Alvarez AR, Gambrell C, Naik K, Raghavan AM, Burow ME, Boue ME, Klinge CM, Ivanova M, Corbitt C. Soy glyceollins regulate transcript abundance in the female mouse brain. Funct. Integr. Genomics. 2015;15:549–561. doi: 10.1007/s10142-015-0442-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Barals N, Ozer S, Karabulut G. The estrogenic effects of apigenin, phloretin and myricetin based on uterotrophic assay in immature Wistar albino rats. Toxicol. Lett. 2014;226:35–42. doi: 10.1016/j.toxlet.2014.01.030. [DOI] [PubMed] [Google Scholar]
  11. Barone M, Tanzi S, Lofano K, Scavo MP, Pricci M, Demarinis L, Papagni S, Guido R, Maiorano E, Ingravallo G, Comelli MC, Francavilla A, Di Leo A. Dietary-induced ERβ upregulation counteracts intestinal neoplasia development in intact male ApcMin/+ mice. Carcinogenesis. 2010;31:269–274. doi: 10.1093/carcin/bgp275. [DOI] [PubMed] [Google Scholar]
  12. Bazer FW, Wu G, Johnson GA, Wang X. Environmental factors affecting pregnancy: Endocrine disrupters, nutrients and metabolic pathways. Mol. Cell. Endocrinol. 2014;398:53–68. doi: 10.1016/j.mce.2014.09.007. [DOI] [PubMed] [Google Scholar]
  13. Bedell S, Nachtigall M, Naftolin F. The pros and cons of plant estrogens for menopause. J. Steroid Biochem. Mol. Biol. 2014;139:225–236. doi: 10.1016/j.jsbmb.2012.12.004. [DOI] [PubMed] [Google Scholar]
  14. Behr M, Oehlmann J, Wagner M. Estrogens in the daily diet: In vitro analysis indicates that estrogenic activity is omnipresent in foodstuff and infant formula. Food Chem. Toxicol. 2011;49:2681–2688. doi: 10.1016/j.fct.2011.07.039. [DOI] [PubMed] [Google Scholar]
  15. Bellanger M, Demeneix B, Grandjean P, Zoeller RT, Trasande L. Neurobehavioral deficits, diseases, and associated costs of exposure to endocrine-disrupting chemicals in the European union. J. Clin. Endocrinol. Metab. 2015;100:1256–1266. doi: 10.1210/jc.2014-4323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bergman Jungestrom M, Thompson LU, Dabrosin T. Flaxseed and its lignans inhibit estradiol-induced growth, angiogenesis, and secretion of vascular endothelial growth factor in human breast cancer xenografts in vivo. Clin. Cancer. Res. 2007;13:1061–1067. doi: 10.1158/1078-0432.CCR-06-1651. [DOI] [PubMed] [Google Scholar]
  17. Bhandari RK, Deem SL, Holliday DK, Jandegian CM, Kassotis CD, Nagel SC, Tillitt DE, vom Saal FS, Rosenfeld CS. Effects of the environmental estrogenic contaminants bisphenol A and 17α-ethinyl estradiol on sexual development and adult behaviors in aquatic wildlife species. Gen. Comp. Endocrinol. 2014;214:195–219. doi: 10.1016/j.ygcen.2014.09.014. [DOI] [PubMed] [Google Scholar]
  18. Bhargavan B, Singh D, Gautam AK, Mishra JS, Kumar A, Goel A, Dixit M, Pandey R, Manickavasagam L, Dwivedi SD, Chakravarti B, Jain JK, Ramachandran R, Maurya R, Trivedi A, Chattopadhyay N, Sanyal S. Medicarpin, a legume phytoalexin, stimulates osteoblastdifferentiation and promotes peak bone mass achievement in rats: evidence for estrogen receptor β-mediated osteogenic action of medicarpin. Journal of Nutritional Biochemistry. 2012;23:27–38. doi: 10.1016/j.jnutbio.2010.11.002. [DOI] [PubMed] [Google Scholar]
  19. Blei T, Soukup ST, Schmalbach K, Pudenz M, Moller FJ, Egert B, Wortz N, Kurrat A, Muller D, Vollmer G, Gerhauser C, Lehmann L, Kulling SE, Diel P. Dose-dependent effects of isoflavone exposure during early lifetime on the rat mammary gland: Studies on estrogen sensitivity, isoflavone metabolism, and DNA methylation. Mol. Nutr. Food Res. 2015;59:270–283. doi: 10.1002/mnfr.201400480. [DOI] [PubMed] [Google Scholar]
  20. Bobjer J, Taxvig C, Christiansen S, Hass U. Possible endocrine disrupting effects of parabens and their metabolites. Reprod. Toxicol. 2010;30:301–312. doi: 10.1016/j.reprotox.2010.03.011. [DOI] [PubMed] [Google Scholar]
  21. Boucher JA, Boudreau A, Ahmed S, Atlas E. In Vitro Effects of Bisphenol A β-D-Glucuronide (BPA-G) on Adipogenesis in Human and Murine Preadipocytes. Environ. Health. Perspect. 2015 doi: 10.1289/ehp.1409143. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Boverhof DR, Kwekel JC, Humes DG, Burgoon LD, Zacharewski TR. Dioxin Induces an Estrogen-Like, Estrogen Receptor-Dependent Gene Expression Response in the Murine Uterus. Mol. Pharmacol. 2006;69:1599–1606. doi: 10.1124/mol.105.019638. [DOI] [PubMed] [Google Scholar]
  23. Calabrese C, Praticò C, Calafiore A, Coscia M, Gentilini L, Poggioli G, Gionchetti P, Campieri M, Rizzello F. Eviendep® reduces number and size of duodenal polyps in familial adenomatous polyposis patients with ileal pouchanal anastomosis. World. J. Gastroenterol. 2013;19:5671–5677. doi: 10.3748/wjg.v19.i34.5671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cao W, Zhuang M, Yang Y, Xie S, Cui J, Cao L, Zhang T, Zhu Y. Preliminary Study of Quercetin Affecting the Hypothalamic-Pituitary-Gonadal Axis on Rat Endometriosis Model. Evid.-based Complement. Altern. Med. 2014 doi: 10.1155/2014/781684. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Capriotti AL, Cavalier C, Colapicchioni V, Piovesana S, Samperi R, Lagana A. Analytical strategies based on chromatography–mass spectrometry for the determination of estrogen-mimicking compounds in food. Journal of Chromatography A. 2013;1313:62–77. doi: 10.1016/j.chroma.2013.06.054. [DOI] [PubMed] [Google Scholar]
  26. Chansakaow S, Ishikawa T, Sekine K, Okada M, Higuchi Y, Kudo M, Chaichantipyuth C. Isoflavonoids from Pueraria mirifica and their Estrogenic Activity. Planta Med. 2000;66:572–575. doi: 10.1055/s-2000-8603. [DOI] [PubMed] [Google Scholar]
  27. Chen J, Thopmson LU. Lignans and tamoxifen, alone or in combination, reduce human breast cancer cell adhesion, invasion and migration in vitro. Breast Cancer Res. Treat. 2003;80:163–70. doi: 10.1023/A:1024513815374. [DOI] [PubMed] [Google Scholar]
  28. Chen J, Hou R, Zhang X, Ye Y, Wang Y, Tian J. Calycosin Suppresses Breast Cancer Cell Growth via ERβ-Dependent Regulation of IGF-1R, p38 MAPK and PI3K/Akt Pathways. PLoS One. 2014;9:e91245. doi: 10.1371/journal.pone.0091245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chen L, Chen W, Qian X, Fang Y, Zhu N. Liquiritigenin alleviates mechanical and cold hyperalgesia in a rat neuropathic pain model. Sci. Rep. 2014;4:5676. doi: 10.1038/srep05676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chen J, Lin C, Yong W, Ye Y, Huang Z. Calycosin and Genistein Induce Apoptosis by Inactivation of HOTAIR/p-Akt Signaling Pathway in Human Breast Cancer MCF-7 Cells. Cell Physiol. Biochem. 2015;35:722–728. doi: 10.1159/000369732. [DOI] [PubMed] [Google Scholar]
  31. Cherdshewasart W, Mahapanichkul T, Boonchird C. Estrogenic and anti-estrogenic activities of the Thai traditional herb. Butea superba Roxb. Biosci. Biotechnol. Biochem. 2010;74:2176–2182. doi: 10.1271/bbb.100159. [DOI] [PubMed] [Google Scholar]
  32. Chevalier N, Fenichel P. Endocrine disruptors: New players in the pathophysiology of type 2 diabetes? Diabetes. Metab. 2015;14:107–115. doi: 10.1016/j.diabet.2014.09.005. [DOI] [PubMed] [Google Scholar]
  33. Cleveland BM, Manor ML. Effects of phytoestrogens on growth-related and lipogenic genes in rainbow trout (Oncorhynchus mykiss) Comp. Biochem. Physiol. C. Toxicol. Pharmacol. 2015;170:28–37. doi: 10.1016/j.cbpc.2015.02.001. [DOI] [PubMed] [Google Scholar]
  34. Coste N, Pelé F, Comets E, Rouget F, Monfort C, Bodeau-Livinec F, Linganiza EM, Bataille H, Kadhel P, Multigner L, Cordier S. Perinatal exposure to chlordecone and infant growth. Environ. Res. 2015;142:123–134. doi: 10.1016/j.envres.2015.06.023. [DOI] [PubMed] [Google Scholar]
  35. Crinnion WJ. Polychlorinated biphenyls: persistent pollutants with immunological, neurological, and endocrinological consequences. Altern. Med. Rev. 2011;16:5–13. [PubMed] [Google Scholar]
  36. Crisp TM, Clegg ED, Cooper RL, Wood WP, Anderson DG, Baetcke KP, Hoffmann JL, Morrow MS, Rodier DJ, Schaeffer JE, Touart LW, Zeeman MG, Patel YM. Environmental endocrine disruption: an effects assessment and analysis. Environ. Health. Perspect. 1998;106(Suppl 1):11–56. doi: 10.1289/ehp.98106s111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. de Lima PF, Colombo CA, Chiorato AF, Yamaguchi LF, Kato MJ, Carbonel SAM. Occurrence of Isoflavonoids in Brazilian Common Bean Germplasm (Phaseolus vulgaris L.) J. Agric. Food Chem. 2014;62:9699–9704. doi: 10.1021/jf5033312. [DOI] [PubMed] [Google Scholar]
  38. Delfosse V, Grimaldi M, Cvailles V, Balaguer P, Bourguet W. Structural and Functional Profiling of Environmental Ligands for Estrogen Receptors. Environ. Health Perspect. 2014;122:1306–1313. doi: 10.1289/ehp.1408453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Delfosse V, Le Maire A, Balaguer P, Bourget W. A structural perspective on nuclear receptors as targets of environmental compounds. Acta Pharmacol. Sin. 2015;36:88–101. doi: 10.1038/aps.2014.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Donzelli A, Braida D, Finardi A, Capurro V, Valsecchi AE, Colleoni M, Sala M. Neuroprotective Effects of Genistein in Mongolian Gerbils: Estrogen Receptor–β Involvement. J. Pharmacol. Sci. 2010;114:158–167. doi: 10.1254/jphs.10164fp. [DOI] [PubMed] [Google Scholar]
  41. Estrella SEN, Landa AI, Lafuente JV, Gargiulo PA. Effects of antidepressants and soybean association in depressive menopausal women. Acta Pol. Pharm. 2014;71:323–327. [PubMed] [Google Scholar]
  42. Evans SF, Kobrosly RW, Barrett ES, Thurston SW, Calafat AM, Weiss B, Stahlhut R, Yolton K, Swan SH. Prenatal bisphenol A exposure and maternally reported behavior in boys and girls. Neurotoxicology. 2014;45:91–99. doi: 10.1016/j.neuro.2014.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Fagnant HS, Uzumzu M, Buckendahl P, Dunn MG, Shupper P, Shapes SA. Fetal and Neonatal Exposure to the Endocrine Disruptor, Methoxychlor, Reduces Lean Body Mass and Bone Mineral Density and Increases Cortical Porosity. Calcif. Tissue. Int. 2014;95:521–529. doi: 10.1007/s00223-014-9916-x. [DOI] [PubMed] [Google Scholar]
  44. Filannino A, Stout TAE, Gadella BM, Sostaric E, Pizzi F, Colenbrander B, Dell'Aquila ME, Minervini F. Dose-response effects of estrogenic mycotoxins (zearalenone, alpha- and beta-zearalenol) on motility, hyperactivation and the acrosome reaction of stallion sperm. Reprod. Biol. Endocrin. 2011;9:134. doi: 10.1186/1477-7827-9-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Frizzel C, Uhlig S, Miels CO, Verhaegen S, Elliot CT, Eriksen GS, Sorlie M, Ropstad E, Connolly L. Biotransformation of zearalenone and zearalenols to their major 4 glucuronide metabolites reduces estrogenic activity. Toxicol. Vitro. 2015;29:575–581. doi: 10.1016/j.tiv.2015.01.006. [DOI] [PubMed] [Google Scholar]
  46. Gaete L, Tchernitchin AN, Bustamante R, Villena J, Lemus I, Gidekel M, Cabrera G, Astroga P. Daidzein–Estrogen Interaction in the Rat Uterus and Its Effect on Human Breast Cancer Cell Growth. J. Med. Food. 2011;15:1081–1090. doi: 10.1089/jmf.2011.0322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gago-Ferrero P, Diaz-Cruz MS, Barcelo D. UV filters bioaccumulation in fish from Iberian river basins. Sci. Total Environ. 2015;518–519:518–525. doi: 10.1016/j.scitotenv.2015.03.026. [DOI] [PubMed] [Google Scholar]
  48. Gao H, Yang B, Li N, Feng L, Shi X, Zhao W, Liu S. Bisphenol A and Hormone-Associated Cancers: Current .Progress and Perspectives. Medicine. 2015;94:e22. doi: 10.1097/MD.0000000000000211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Gao X, Ma J, Chen Y, Wang H. Rapid Responses and Mechanism of Action for Low-Dose Bisphenol S on ex Vivo Rat Hearts and Isolated Myocytes: Evidence of Female-Specific Proarrhythmic Effects. Environ. Health Perspect. 2015;123:571–578. doi: 10.1289/ehp.1408679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gong P, Madak-Erdogan Z, Li J, Cheng J, Greenlief CM, Helferich W, Katzenellenbogen JA, Katzenellenbogen BS. Transcriptomic analysis identifies gene networks regulated by estrogen receptor α (ERα) and ERβ that control distinct effects of different botanical estrogens. Nucl. Recept. Signal. 2014;12:e001. doi: 10.1621/nrs.12001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Grimaldi M, Boulahtouf A, Delfosse V, Thouennon E, Bourguet W, Balaguer P. Reporter cell lines for the characterization of the interactions between human nuclear receptors and endocrine disruptors. Front. Endocrinol. 2015;6:62. doi: 10.3389/fendo.2015.00062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Harris HA, Katzenellenbogen JA, Katzenellenbogen BS. Characterization of the Biological Roles of the Estrogen Receptors, ERα and ERβ, in Estrogen Target Tissues in Vivo through the Use of an ERα-Selective Ligand. Endocrinology. 2002;143:4172–4177. doi: 10.1210/en.2002-220403. [DOI] [PubMed] [Google Scholar]
  53. Harvey PA, Leinwand LA. Dietary phytoestrogens present in soy dramatically increase cardiotoxicity in male mice receiving a chemotherapeutic tyrosine kinase inhibitor. Mol. Cell. Endocrinol. 2015;399:330–335. doi: 10.1016/j.mce.2014.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hauser L, Skakkebaek NE, Hass U, Toppari J, Juul A, Andersson AM, Kortenkamp A, Heindel JJ, Trasande L. Male reproductive disorders, diseases, and costs of exposure to endocrine-disrupting chemicals in the European union. J. Clin. Endocrinol. Metab. 2015;100:1267–1277. doi: 10.1210/jc.2014-4325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. He X, Xi C, Tang B, Wang G, Chen D, Peng T, Mu Z. Simultaneous determination of 30 hormones illegally added to anti-ageing functional foods using UPLC-MS/MS coupled with SPE clean-up. Food Addit. Contam. Part A-Chem. 2014;31:1625–1638. doi: 10.1080/19440049.2014.953602. [DOI] [PubMed] [Google Scholar]
  56. Helle J, Kraker K, Bader MI, Keiler AM, Zierau O, Vollmer G, Welsh J, Kretzschmar G. Assessment of the proliferative capacity of the flavanones 8-prenylnaringenin, 6-(1.1 dimethylallyl)naringenin and naringenin in MCF-7 cells and the rat mammary gland. Mol. Cell. Endocrinol. 2014;392:125–135. doi: 10.1016/j.mce.2014.05.014. [DOI] [PubMed] [Google Scholar]
  57. http://www.who.int/ceh/risks/cehemerging2/en/ Last accessed 05/22/2015
  58. Ishiwata N, Melby MK, Mizuno S, Watanabe S. New equol supplement for relieving menopausal symptoms: Randomized, placebo-controlled trial of Japanese women. Menopause. 2009;16:141–148. doi: 10.1097/gme.0b013e31818379fa. [DOI] [PubMed] [Google Scholar]
  59. Jackson RL, Greiwe JS, Schwen RJ. Emerging evidence of the health benefits of S-equol, an estrogen receptor β agonist. Nutrition Rev. 2011;69:432–448. doi: 10.1111/j.1753-4887.2011.00400.x. [DOI] [PubMed] [Google Scholar]
  60. Jahromi SR, Arrefhosseini SR, Ghaemi A, Alizadeh A, Sabetghadam F, Togha M. Effect of oral genistein administration in early and late phases of allergic encephalomyelitis. Iran. J. Basic Med. Sci. 2014;17:509–515. [PMC free article] [PubMed] [Google Scholar]
  61. Jungsukcharoen J, Dhiani BA, Cherdshewasart W, Vinayavekhin N, Sangvanich P, Boonchird C. Pueraria mirifica leaves, an alternative potential isoflavonoid source. Biosci. Biotechnol. Biochem. 2014;78:917–926. doi: 10.1080/09168451.2014.910091. [DOI] [PubMed] [Google Scholar]
  62. Justesen U, Knuthsen P. Composition of flavonoids in fresh herbs and calculation of flavonoid intake by use of herbs in traditional Danish dishes. Food Chem. 2001;73:245–250. [Google Scholar]
  63. Kajta M, Litwa E, Wnuk A, Lason W, Zelek-Molik A, Nalepa I, Grzegorzewska-Hiczwa M, Tokarski K, Golas A, Guzik E, Grochowalski A, Szychowski KA, Wojtowicz AK. Isomer-nonspecific action of dichlorodiphenyltrichloroethane on aryl hydrocarbon receptor and G-protein-coupled receptor 30 intracellular signaling in apoptotic neuronal cells. Mol. Cell. Endocrinol. 2014;392:90–105. doi: 10.1016/j.mce.2014.05.008. [DOI] [PubMed] [Google Scholar]
  64. Kajta M, Rzemieniec J, Litwa E, Lason W, Lenartowicz M, Krzeptowski W, Wojtowicz AK. The key involvement of estrogen receptor β and G-protein-coupled receptor 30 in the neuroprotective ation of daidzein. Neuroscience. 2013;238:345–360. doi: 10.1016/j.neuroscience.2013.02.005. [DOI] [PubMed] [Google Scholar]
  65. Kang GK, Jeon SH, Cho JH, Kim DG, Park JM, Cho MH. Evaluation of estrogenic and androgenic activity of butylated hydroxyanisole in immature female and castrated rats. Toxicology. 2006;213:147–156. doi: 10.1016/j.tox.2005.05.027. [DOI] [PubMed] [Google Scholar]
  66. Karpuzoglu E, Holladay SD, Gogal RM. Parabens: Potential impact of Low-Affinity Estrogen receptor Binding chemicals on Human health. J. Toxicol. Env. Health B Crit. Rev. 2013;16:321–335. doi: 10.1080/10937404.2013.809252. [DOI] [PubMed] [Google Scholar]
  67. Keiler AM, Djiogue S, Ehrhardt T, Zierau O, Skaltsounis L, Halabalaki M, Vollmer G. Oleocanthal Modulates Estradiol-Induced Gene Expression Involving Estrogen Receptor α. Planta Med. 2015;81:1263–1269. doi: 10.1055/s-0035-1546194. [DOI] [PubMed] [Google Scholar]
  68. Khan U, Nicell JA. Contraceptive Options and Their Associated Estrogenic Environmental Loads: Relationships and Trade-Offs. PLoS One. 2014;9:e92630. doi: 10.1371/journal.pone.0092630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kim HJ, Suh H-J, Kim JH, Kangm SC, Park S, Lee CH, Kim J-S. Estrogenic Activity of Glyceollins Isolated from Soybean Elicited with Aspergillus sojae. J. Med. Food. 2010;13:382–390. doi: 10.1089/jmf.2009.1281. [DOI] [PubMed] [Google Scholar]
  70. Kim J, Kang EJ, Park MN, Kim JE, Kim SC, Jeung EB, Lee GS, Hwang DY, An BS. The adverse effect of 4-tert-octylphenol on fat metabolism in pregnant rats via regulation of lipogenic proteins. Environ. Toxicol. Pharmacol. 2015;40:284–291. doi: 10.1016/j.etap.2015.06.020. [DOI] [PubMed] [Google Scholar]
  71. Klann A, Levy G, Lutz I, Muller C, Kloas W, Hildebrandt JP. Estrogen-like effects of ultraviolet screen 3-(4-methylbenzylidene)- camphor (Eusolex 6300) on cell proliferation and gene induction in mammalian and amphibian cells. Environ. Res. 2005;97:274–281. doi: 10.1016/j.envres.2004.07.004. [DOI] [PubMed] [Google Scholar]
  72. Ko J, Bao C, Park HC, Kim M, Choi HK, Kim YS, Lee HJ. β-Thujaplicin modulates estrogen receptor signaling and inhibits proliferation of human breast cancer cells. Biosci. Biotechnol. Biochem. 2015;10:1–7. doi: 10.1080/09168451.2015.1008978. [DOI] [PubMed] [Google Scholar]
  73. Kretzschamar G, Zierau O, Wober J, Tiscer S, Metz P, Vollmer G. Prenylation has a compound specific effect on the estrogenicity of naringenin and genistein. J. Steroid Biochem. Mol. Biol. 2010;118:1–6. doi: 10.1016/j.jsbmb.2009.08.005. [DOI] [PubMed] [Google Scholar]
  74. Kudłaka B, Szczepańskaa N, Owczareka K, Mazerskab Z, Namieśnika J. Revision of Biological Methods for Determination of EDC Presence and Their Endocrine Potential. Crit. Rev. Anal. Chem. 2015;45:191–200. doi: 10.1080/10408347.2014.904731. [DOI] [PubMed] [Google Scholar]
  75. Kumal A, Goyal R, Kumar S, Jain S, Jain N, Kumar P. Estrogenic and Anti-Alzheimer’s studies of Zingiber officinalis as well as Amomum subulatum Roxb.: the success story of dry techniques. Med. Chem. Res. 2015;24:1089–1097. [Google Scholar]
  76. Kumar E, Holt WV. Impacts of endocrine disrupting chemicals on reproduction in wildlife. Adv. Exp. Med. Biol. 2015;753:55–70. doi: 10.1007/978-1-4939-0820-2_4. [DOI] [PubMed] [Google Scholar]
  77. Kwon Y. Effect of soy isoflavones on the growth of human breast tumors: findings from preclinical studies. Food Sci. Nutr. 2014;2:613–622. doi: 10.1002/fsn3.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Legler J, Fletcher T, Govarts E, Porta M, Blumberg B, Heindel JJ, Trasande L. Obesity, Diabetes, and Associated Costs of Exposure to Endocrine-Disrupting Chemicals in the European Union. J. Clin. Endocrinol. Metab. 2015;100:1278–1288. doi: 10.1210/jc.2014-4326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Lerro CC, Koutros S, Andreotti G, Fruesen MC, Alavanja MC, Blair A, Hoppin JA, Sandler DP, Lubin JH, Ma X, Zhang Y, Beane Freeman LE. Organophosphate insecticide use and cancer incidence among spouses of pesticide applicators in the Agricultural Health Study. Occup. Environ. Med. 2015;72:736–744. doi: 10.1136/oemed-2014-102798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Li J, Zou M, Liu X, Yun C, Qi X, Zhang X. Determination of estrogens in animal-derived food by matrix solid-phase dispersion extraction coupled with ultra performance liquid chromatography-quadruple time of flight mass spectrometry. Anal. Methods. 2013;5:1004–1009. [Google Scholar]
  81. Li L, Bonnetton F, Chen XY, Laudet V. Botanical compounds and their regulation of nuclear receptor action: The case of traditional Chinese medicine. Mol. Cell. Endocrinol. 2015;401:221–237. doi: 10.1016/j.mce.2014.10.028. [DOI] [PubMed] [Google Scholar]
  82. Li X, Jiang L, Cheng L, Chen H. Dibutyl phthalate-induced neurotoxicity in the brain of immature and mature rat offspring. Brain Dev. 2014;36:653–660. doi: 10.1016/j.braindev.2013.09.002. [DOI] [PubMed] [Google Scholar]
  83. Lobaccaro JA, Trousson A. Environmental Estrogen Exposure During Fetal Life: A Time Bomb for Prostate Cancer. Endocrinology. 2014;155:656–658. doi: 10.1210/en.2014-1057. [DOI] [PubMed] [Google Scholar]
  84. Lu J, Wu J, Stoffella PJ, Wilson PC. Analysis of Bisphenol A, Nonylphenol, and Natural Estrogens in Vegetables and Fruits Using Gas Chromatography–Tandem Mass Spectrometry. J. Agric. Food Chem. 2013;61:84–89. doi: 10.1021/jf304971k. [DOI] [PubMed] [Google Scholar]
  85. Luciani-Torres MG, Moore DH, Goodson WH, Dairkee SH. Exposure to the polyester PET precursor—terephthalic acid induces and perpetuates DNA damage-harboring non-malignant human breast cells. Carcinogenesis. 2015;36:168–176. doi: 10.1093/carcin/bgu234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Luo D, Liu Y, Zhou Y, Chen Z, Yang L, Xu Q, Xu H, Kuang H, Huang Q, He M, Peng W. Association between dietary phytoestrogen intake and bone mineral density varied with estrogen receptor alpha gene polymorphisms in southern Chinese postmenopausal women. Food Funct. 2015;6:1977–1983. doi: 10.1039/c5fo00295h. [DOI] [PubMed] [Google Scholar]
  87. Maggiolini M, Statti G, Vivacqua A, Gabriele S, Rago V, Loizzo M, Menichini F, Amdò S. Estrogenic and antiproliferative activities of isoliquiritigenin in MCF7 breast cancer cells. J. Steroid Biochem. Mol. Biol. 2002;82:315–322. doi: 10.1016/s0960-0760(02)00230-3. [DOI] [PubMed] [Google Scholar]
  88. Mahmoud AM, Yang W, Bosland MC. Soy isoflavones and prostate cancer: A review of molecular mechanisms. J. Steroid Biochem. Mol. Biol. 2014;140:116–132. doi: 10.1016/j.jsbmb.2013.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Malekinejad H, Maas-Bakker R, Fink-Gremmels J. Enzyme kinetics of zearalenone biotransformation: pH and cofactor effects. Arch. Toxicol. 2005;79:547–553. doi: 10.1007/s00204-005-0664-6. [DOI] [PubMed] [Google Scholar]
  90. Masrudin SS, Mohamad J. Preventive effect of Pueraria mirifica on testosteroneinduced prostatic hyperplasia in Sprague Dawley rats. Andrologia. 2014;47:1153–1159. doi: 10.1111/and.12396. [DOI] [PubMed] [Google Scholar]
  91. Matori H, Umar S, Nadadur RD, Sharma S, Partow-Navis R, Afkhami M, Amjedi M, Eghbali M. Genistein, a Soy Phytoestrogen, Reverses Severe Pulmonary Hypertension and Prevents Right Heart Failure in Rats. Hypertension. 2012;60:425–430. doi: 10.1161/HYPERTENSIONAHA.112.191445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Mertl J, Kirchnawy C, Osorio V, Grininger A, Richter A, Bergmair J, Pyerin M, Washuttl M, Tacker M. Characterization of Estrogen and Androgen Activity of Food Contact Materials by Different In Vitro Bioassays (YES, YAS, ERa and AR CALUX) and Chromatographic Analysis (GC-MS, HPLC-MS) PLoS One. 2014;9:e100952. doi: 10.1371/journal.pone.0100952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Minutolo F, Macchia M, Katzenellenbogen BS, Katzenellenbogen JA. Estrogen receptor β ligands: recent advances and biomedical applications. Med. Res. Rev. 2011;31:364–442. doi: 10.1002/med.20186. [DOI] [PubMed] [Google Scholar]
  94. Miyagawa S, Lange A, Hirakawa I, Tohyama S, Ogino Y, Mizutani T, Kaami Y, Kusano T, Ihara M, Tanaka H, Tazarazako N, Ohta Y, Katsu Y, Tyler CR, Iguchi T. Differing Species Responsiveness of Estrogenic Contaminants in Fish Is Conferred by the Ligand Binding Domain of the Estrogen Receptor. Environ. Sci. Technol. 2014;48:5254–5263. doi: 10.1021/es5002659. [DOI] [PubMed] [Google Scholar]
  95. Moral R, Santucci-Pereira J, Wang R, Russo IH, Lamartiniere CA, Russo J. In utero exposure to butyl benzyl phthalate induces modifications in the morphology and the gene expression profile of the mammary gland: an experimental study in rats. Environ. Health. 2011;10:5. doi: 10.1186/1476-069X-10-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Murphy E. Estrogen signaling and cardiovascular disease. Circ. Res. 2011;109:687–696. doi: 10.1161/CIRCRESAHA.110.236687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Nilsson S, Gustafsson J-Å. Estrogen Receptors: Therapies Targeted to Receptor Subtypes. Clin. Pharmacol. Ther. 2011;89:44–55. doi: 10.1038/clpt.2010.226. [DOI] [PubMed] [Google Scholar]
  98. Ogo Y, Ozawa K, Ishimaru T, Murayama T, Takaiwa F. Transgenic rice seed synthesizing diverse flavonoids at high levels: a new platform for flavonoid production with associated health benefits. Plant Biotechnol. J. 2013;11:734–746. doi: 10.1111/pbi.12064. [DOI] [PubMed] [Google Scholar]
  99. Ollberding NJ, Lim U, Wilkens LR, Setiawan VW, Shvetsov YB, Henderson BE, Kolonel LN, Goodman MT. Legume, Soy, Tofu, and Isoflavone Intake and Endometrial Cancer Risk in Postmenopausal Women in the Multiethnic Cohort Study. J. Natl. Cancer Inst. 2012;104:67–76. doi: 10.1093/jnci/djr475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Palumbo C, Cavani F, Bertoni L, Ferretti M. Role of Phytoestrogen Ferutinin in Preventing/Recovering Bone Loss: Results from Experimental Ovariectomized Rat Models, Osteoporosis. In: Yannis Dionyssiotis., editor. InTech. 2012. Available from: http://www.intechopen.com/books/osteoporosis/role-of-phytoestrogen-ferutinin-in preventing recovering-bone-loss-results-from-experimental-ovariec. [DOI]
  101. Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors alpha (ERα) and beta (ERβ): subtype-selective ligands and clinical potential. Steroids. 2014;90:13–29. doi: 10.1016/j.steroids.2014.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Patisaul HB, Jefferson W. The pros and cons of phytoestrogens. Frontiers in Neuroendrocrinology. 2010;31:400–419. doi: 10.1016/j.yfrne.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Payton-Stewart F, Khupse RS, Bouè SM, Elliot S, Zimmermann MC, Skripnikov EV, Ashe H, Tilgman SL, Beckman BS, Cleveland TE, McLachlan JA, Bhatnagar D, Wiese TE, Erhardt P, Burow ME. Glyceollin I enantiomers distinctly regulate ER-mediated gene expression. Steroids. 2010;75:870–878. doi: 10.1016/j.steroids.2010.05.007. [DOI] [PubMed] [Google Scholar]
  104. Penalvo JL, Lopez-Romero P. Urinary enterolignan concentrations are positively associated with serum HDL cholesterol and negatively associated with serum triglycerides in U.S. adults. J. Nutr. 2012;142:751–756. doi: 10.3945/jn.111.150516. [DOI] [PubMed] [Google Scholar]
  105. Petersin JJ, Dwyer JT, Jacque PF, McCullough ML. Associations between flavonoids and cardiovascular disease incidence or mortality in European and US populations. Nutr. Rev. 2012;70:491–508. doi: 10.1111/j.1753-4887.2012.00508.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Porras SP, Heinala M, Santonen T. Bisphenol A exposure via thermal paper receipts. Toxicol. Lett. 2014;230:413–420. doi: 10.1016/j.toxlet.2014.08.020. [DOI] [PubMed] [Google Scholar]
  107. Rebuli ME, Cao J, Sluzas E, Barry Delcos E, Camacho L, Lewis SM, Vanlandingham MM, Patisaul HB. Investigation of the Effects of Subchronic Low Dose Oral Exposure to Bisphenol A (BPA) and Ethinyl Estradiol (EE) on Estrogen Receptor Expression in the Juvenile and Adult Female Rat Hypothalamus. Toxicol. Sci. 2014;140:190–203. doi: 10.1093/toxsci/kfu074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Reed SD, Lampe JV, Qu C, Gundersen G, Fuller S, Copeland WK, Newton KM. Self-reported menopausal symptoms in a racially diverse population and soy food consumption. Maturitas. 2013;75:152–158. doi: 10.1016/j.maturitas.2013.03.003. [DOI] [PubMed] [Google Scholar]
  109. Reger MK, Zollinger TW, Liu Z, Jones J, Zhang J. Urinary phytoestrogens and cancer, cardiovascular, and all cause mortality in the continuous National Health and Nutrition Examination Survey. Eur. J. Nutr. 2015 doi: 10.1007/s00394-015-0917-y. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  110. Rigalli JP, Ciriaci N, Arias A, Ceballos MP, Villenueva SSM, Luquita MG, Mottino AD, Ghanem CI, Catania VA, Ruiz ML. Regulation of Multidrug Resistance Proteins by Genistein in a Hepatocarcinoma Cell Line: Impact on Sorafenib Cytotoxicity. PLoS One. 2015;10:e0119502. doi: 10.1371/journal.pone.0119502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Sánchez Rodriguez A, Sanz MR, Betancort Rodriguez JR. Occurrence of eight UV filters in beaches of Gran Canaria (Canary Islands). An approach to environmental risk assessment. Chemosphere. 2015;131:85–90. doi: 10.1016/j.chemosphere.2015.02.054. [DOI] [PubMed] [Google Scholar]
  112. Radenkova-Saeva J. Immunotoxicity of dioxins. J. Clin. Med. 2009;2:8–13. [Google Scholar]
  113. Rosenfeld CS. Bisphenol A and phthalate endocrine disruption of parental and social behaviors. Front. Neurosci. 2015;9:1–15. doi: 10.3389/fnins.2015.00057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Rothe J, Wakileh M, Dreibiger K, Weber H. The flavonoid beverage Haelan 951 induces growth arrest and apoptosis in pancreatic carcinoma cell lines in vitro. BMC Complement. Altern. Med. 2015;15:212. doi: 10.1186/s12906-015-0734-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Rotroff DM, Martin MT, Dix DJ, Filer DL, Houck KA, Knudsen TB, Sipes NS, Reif DM, Xia M, Huang R, Judson RS. Predictive Endocrine Testing in the 21st Century Using in Vitro Assays of Estrogen Receptor Signaling Responses. Environ. Sci. Technol. 2014;48:8706–8716. doi: 10.1021/es502676e. [DOI] [PubMed] [Google Scholar]
  116. Rouiller-Fabre V, Guerquin MJ, N'Tumba-Byn T, Muczynski V, Moison D, Tourpin S, Messiaen S, Haber R, Livera G. Nuclear receptors and endocrine disruptors in fetal and neonatal testes: a gapped landscape. Front. Endocrinol. (Lausanne) 2015;6:58. doi: 10.3389/fendo.2015.00058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Rowland I, Faughnan M, Wahala K, Williamson G, Cassidy A. Bioavailability of phyto-oestrogens. Br. J. Nutr. 2003;89:S45–58. doi: 10.1079/BJN2002796. [DOI] [PubMed] [Google Scholar]
  118. Ruan T, Liang D, Song S, Song M, Wang H, Jiang G. Evaluation of the in vitro estrogenicity of emerging bisphenol analogs and their respective estrogenic contributions in municipal sewage sludge in China. Chemosphere. 2015;124:150–155. doi: 10.1016/j.chemosphere.2014.12.017. [DOI] [PubMed] [Google Scholar]
  119. Sacks FM, Lichtenstein A, Van Horn L, Harris W, Kris-Etherton P, Winston M, for the AHA Nutrition Committee Soy Protein, Isoflavones, and Cardiovascular Health A Summary of a Statement for Professionals From the American Heart Association Nutrition Committee. Circulation. 2006;113:1034–1044. doi: 10.1161/CIRCULATIONAHA.106.171052. [DOI] [PubMed] [Google Scholar]
  120. Salvo VA, Boué SM, Fonseca JP, Elliott S, Corbitt C, Collins-Burow BM, Curiel TJ, Srivastav SK, Shih BY, Carter-Wientjes C, Wood CE, Erhardt PW, Beckaman BS, McLachlan JA, Cleveland TE, Burow ME. Antiestrogenic Glyceollins Suppress Human Breast and Ovarian Carcinoma Tumorigenesis. Clin. Cancer Res. 2006;12:7159–7164. doi: 10.1158/1078-0432.CCR-06-1426. [DOI] [PubMed] [Google Scholar]
  121. Schereihofer DA, Redmond L. Soy phytoestrogens are neuroprotective against stroke-like injury in vitro. Neuroscience. 2009;158:602–609. doi: 10.1016/j.neuroscience.2008.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Schleipen B, Hertrampfk T, Fritzmeier KH, Kluxen FM, Lorenze A, Molzberger A, Velders M, Diel P. ERβ specific agonists and genistein inhibit proliferation and induce apoptosis in the large and small intestine. Carcinogenesis. 2011;32:1675–1683. doi: 10.1093/carcin/bgr188. [DOI] [PubMed] [Google Scholar]
  123. Segner H, Casanova-Nakayama A, Kase R, Tyler C-R. Impact of environmental estrogens on fish considering the diversity of estrogen signaling. Gen. Comp. Endocrinol. 2013;191:190–201. doi: 10.1016/j.ygcen.2013.05.015. [DOI] [PubMed] [Google Scholar]
  124. Sexton MJ, Gherman RB. Selective estrogen receptor modulators: the ideal estrogen replacement. Prim. Care Update OB Gyns. 2001;8:25–30. doi: 10.1016/s1068-607x(00)00066-4. [DOI] [PubMed] [Google Scholar]
  125. Shambayati M, Patel M, Ma Y, Cunningham RL, Schreihofer DA. Central inflammatory response to experimental stroke is inhibited by a neuroprotective dose of dietary soy. Brain Research. 2014;1953:76–82. doi: 10.1016/j.brainres.2014.09.042. [DOI] [PubMed] [Google Scholar]
  126. Shargil D, Gerstl Z, Fine P, Nitsan I, Kurtzman D. Impact of biosolids and wastewater effluent application to agricultural land on steroidal hormone content in lettuce plants. Sci. Total. Environ. 2015;505:357–366. doi: 10.1016/j.scitotenv.2014.09.100. [DOI] [PubMed] [Google Scholar]
  127. Sidhu HS, Wilson PC, O'Connor GA. Endocrine-Disrupting Compounds in Reclaimed Water and Residential Ponds and Exposure Potential for Dislodgeable Residues in Turf Irrigated With Reclaimed Water. Arch. Environ. Contam. Toxicol. 2015;69:81–88. doi: 10.1007/s00244-015-0147-6. [DOI] [PubMed] [Google Scholar]
  128. Simons R, Gruppen H, Bovee TFH, Verbruggen MA, Vincken J-P. Prenylated isoflavonoids from plants as selective estrogen receptor modulators (phytoSERMs) Food Funct. 2012;3:810–827. doi: 10.1039/c2fo10290k. [DOI] [PubMed] [Google Scholar]
  129. Singh V, Sharma V, Verma V, Pandey D, Yadav SK, Maikhuri JP, Gupta G. Apigenin manipulates the ubiquitin–proteasome system to rescue estrogen receptor-β from degradation and induce apoptosis in prostate cancer cells. Eur. J. Nutr. 2014;54:1255–1267. doi: 10.1007/s00394-014-0803-z. [DOI] [PubMed] [Google Scholar]
  130. Sirotkin AV, Harrat AH. Phytoestrogens and their effects. Eur. J. Pharmacol. 2014;741:230–236. doi: 10.1016/j.ejphar.2014.07.057. [DOI] [PubMed] [Google Scholar]
  131. Smerieri A, Testa C, Lazzeroni P, Nuti F, Grossi E, Cesari S, Montanini L, Latini G, Bernasconi S, Papini AM, Street ME. Di-(2-Ethylhexyl) Phthalate Metabolites in Urine Show Age-Related Changes and Associations with Adiposity and Parameters of Insulin Sensitivity in Childhood. PLoS One. 2015;10:e0117831. doi: 10.1371/journal.pone.0117831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Song L, Liang X, Zhou Y. Estrogen-mimicking isoflavone genistein prevents bone loss in a rat model of obstructive sleep apnea-hypopnea syndrome. Int. J. Clin. Exp. Pathol. 2014;7:1687–1694. [PMC free article] [PubMed] [Google Scholar]
  133. Song M, Liang D, Liang Y, Chen M, Wang F, Wang H, Jiang G. Assessing developmental toxicity and estrogenic activity of halogenated bisphenol A on zebrafish (Danio rerio) Chemosphere. 2014;112:275–281. doi: 10.1016/j.chemosphere.2014.04.084. [DOI] [PubMed] [Google Scholar]
  134. Song Z, Han S, Pan X, Gong Y, Wang M. Pterostilbene mediates neuroprotection against oxidative toxicity via oestrogen receptor α signalling pathways. J. Pharm. Pharmacol. 2015;67:720–730. doi: 10.1111/jphp.12360. [DOI] [PubMed] [Google Scholar]
  135. Soni M, Rahardji TBW, Soekardi R, Sulistyowati Y, Lestariningsih, Yesufu-Udechuko A, Irsan A, Hogervorst E. Phytoestrogens and cognitive function: a review. Maturitas. 2014;77:209–220. doi: 10.1016/j.maturitas.2013.12.010. [DOI] [PubMed] [Google Scholar]
  136. Soto AM, Sonnenschein C. Environmental causes of cancer: endocrine disruptors as carcinogens. Nat. Rev. Endocrinol. 2010;6:363–370. doi: 10.1038/nrendo.2010.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Stopper H, Schmitt E, Kobras K. Genotoxicity of phytoestrogens. Mutation Research. 2005;574:139–155. doi: 10.1016/j.mrfmmm.2005.01.029. [DOI] [PubMed] [Google Scholar]
  138. Stossi F, Bolt MJ, Aschcroft FJ, Lamerdin JE, Melnick JS, Powell RT, Dandekar RD, Mancini MG, Walker CL, Westwick JK, Mancini MA. Defining Estrogenic Mechanisms of Bisphenol A Analogs through High Throughput Microscopy-Based Contextual Assays. Chem. Biol. 2014;21:743–753. doi: 10.1016/j.chembiol.2014.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Tait S, Tassinari R, Maranghi F, Mantovani A. Bisphenol A affects placental layers morphology and angiogenesis during early pregnancy phase in mice. J. Appl. Toxicol. 2015;35:1278–1291. doi: 10.1002/jat.3176. [DOI] [PubMed] [Google Scholar]
  140. Takeda S. Δ9-Tetrahydrocannabinol Targeting Estrogen Receptor Signaling: The Possible Mechanism of Action Coupled with Endocrine Disruption. Biol. Pharm. Bull. 2014;37:1435–1438. doi: 10.1248/bpb.b14-00226. [DOI] [PubMed] [Google Scholar]
  141. Takemura H, Shim JY, Sayama K, Tsubura A, Zhu BT, Shimoi K. Characterization of the estrogenic activities of zearalenone and zeranol in vivo and in vitro. J. Steroid. Biochem. Mol. Biol. 2007;103:170–177. doi: 10.1016/j.jsbmb.2006.08.008. [DOI] [PubMed] [Google Scholar]
  142. Ter Veld MGR, Schouten B, Louisse J, Van Es DS, Vander Saag PT, Rietjens MCM, Murk AJ. Estrogenic Potency of Food-Packaging-Associated Plasticizers and Antioxidants As Detected in ERα and ERβ Reporter Gene Cell Lines. J. Agric. Food Chem. 2006;54:4407–4416. doi: 10.1021/jf052864f. [DOI] [PubMed] [Google Scholar]
  143. Tiosano D, Paris F, Grimaldi M, Georgescu V, Servant N, Hochberg Z, Balaguer P, Sultan C. Evidence of ERalpha and ERbeta selectivity and partial estrogen agonism in traditional Chinese medicine. Reprod. Biol.Endocrinol. 2014;12:97. doi: 10.1186/1477-7827-12-97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Trevino LS, Wang Q, Walker CL. Hypothesis: Activation of rapid signaling by environmental estrogens and epigenetic reprogramming in breast cancer. Reprod. Toxicol. 2015;54:136–140. doi: 10.1016/j.reprotox.2014.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Troisi J, Mikelson C, Richards S, Symes S, Adair D, Zullo F, Guida M. Placental concentrations of bisphenol A and birth weight from births in the Southeastern U.S. Placenta. 2014;35:947–952. doi: 10.1016/j.placenta.2014.08.091. [DOI] [PubMed] [Google Scholar]
  146. Tsai Y, Hsu C, El-Shazly ME, Chiang S, Wu C, Wu C, Lai W, Yen M, Wu Y, Chang F. Phytochemicals and Estrogen-Receptor Agonists from the Aerial Parts of Liriope platyphylla. Molecules. 2015;20:6844–6855. doi: 10.3390/molecules20046844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Tyagu V, Garg N, Mustafa MD, Banerjee BD, Guleria K. Organochlorine pesticide levels in maternal blood and placental tissue with reference to preterm birth: a recent trend in North Indian population. Environ. Monit. Assess. 2015;187:471. doi: 10.1007/s10661-015-4369-x. [DOI] [PubMed] [Google Scholar]
  148. United States Department of Agriculture (Agricultural Research Service). Database for the Isoflavone content of selected foods. Release 2.0. 2008. [Google Scholar]
  149. United States Department of Agriculture (Agricultural Research Service). Database for the Flavone content of selected foods. Release 3. 2011. [Google Scholar]
  150. Uzumcu M, Kuhn PE, Marano JE, Armenti AE, Passantino L. Early postnatal methoxychlor exposure inhibits folliculogenesis and stimulates anti-Mullerian hormone production in the rat ovary. J. Endocrinol. 2006;191:549–558. doi: 10.1677/joe.1.06592. [DOI] [PubMed] [Google Scholar]
  151. Wang J, Jenkins S, Lamartiniere CA. Cell proliferation and apoptosis in rat mammary glands following combinational exposure to bisphenol A and genistein. BMC Cancer. 2014;14:379. doi: 10.1186/1471-2407-14-379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Wang J, Xia Y, Xiao Y, Ying C, Long J, Zhang H, Chen X, Mao C, Li X, Wang L, Xu S. Assessment of estrogen disrupting potency in animal foodstuffs of China by combined biological and chemical analyses. J. Environ. Sci. 2014;26:2131–2137. doi: 10.1016/j.jes.2014.08.013. [DOI] [PubMed] [Google Scholar]
  153. Wang J, Xu J, Shu FR, Chen K, Min MT. Equol promotes rat osteoblast proliferation and differentiation through activating estrogen receptor. Genet. Mol. Res. 2014;13:5055–5063. doi: 10.4238/2014.July.4.21. [DOI] [PubMed] [Google Scholar]
  154. Wang P, Xiao Y, Liu W, Wang J, Yang Y. Vortex-assisted hollow fibre liquid-phase microextraction technique combined with high performance liquid chromatography-diode array detection for the determination of oestrogens in milk samples. Food Chem. 2014;172:385–390. doi: 10.1016/j.foodchem.2014.09.092. [DOI] [PubMed] [Google Scholar]
  155. Wang S, Huang W, Fang G, Zhang Y, Qiao H. Analysis of steroidal estrogen residues in food and environmental samples. Intern. J. Environ. Anal. Chem. 2008;88:1–25. [Google Scholar]
  156. Wang W, Hafner KS, Flaws JA. In utero bisphenol A exposure disrupts germ cell nest breakdown and reduces fertility with age in the mouse. Toxicol. Appl. Pharmacol. 2014;276:157–164. doi: 10.1016/j.taap.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Wang Z, Wang L, Xia M, Sun W, Huang C, Cui X, Hu G, Lian Q, Wang Z. Pharmacokinetics Interaction between Imatinib and Genistein in Rats. Biomed. Res. Int. 2015 doi: 10.1155/2015/368976. Article ID 368976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Weisbrod CJ, Kunz PY, Zenker AF, Fent K. Effects of the UV filter benzophenone-2 on reproduction in fish. Toxicol. Appl. Pharmacol. 2007;225:255–66. doi: 10.1016/j.taap.2007.08.004. [DOI] [PubMed] [Google Scholar]
  159. WHO/UNEP. State of the science of endocrine disrupting chemicals - 2012. An assessment of the state of the science of endocrine disruptors prepared by a group of experts for the United Nations Environment Programme (UNEP) and WHO. 2013. [Google Scholar]
  160. Wilkinson AS, Taing M, Pierson JT, Lin C, Dietzgen RG, Shaw N, Gidley MJ, Monteith GR, Roberts-Thomson SJ. Estrogen modulation properties of mangiferin and quercetin and the mangiferin metabolite norathyriol. Food Funct. 2015;6:1847–1854. doi: 10.1039/c5fo00133a. [DOI] [PubMed] [Google Scholar]
  161. Wrobel AM, Gregoraszczuk EL. Actions of methyl-, propyl- and butylparaben on estrogen receptor-α and -β and the progesterone receptor in MCF-7 cancer cells and non-cancerous MCF-10A cells. Toxicol. Lett. 2014;230:375–381. doi: 10.1016/j.toxlet.2014.08.012. [DOI] [PubMed] [Google Scholar]
  162. Wroblewski Lissin L, Cooke JP. Phytoestrogens and Cardiovascular Health. J. Am. Coll. Cardiol. 2000;35:1403–1410. doi: 10.1016/s0735-1097(00)00590-8. [DOI] [PubMed] [Google Scholar]
  163. Xiao X, Liu Z, Wang R, Wang J, Zhang S, Cai X, Wu K, Bergan RC, Xu L, Fan D. Genistein suppresses FLT4 and inhibits human colorectal cancer metastasis. Oncotarget. 2014;6:3225–3239. doi: 10.18632/oncotarget.3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Xin D, Wang H, Yang J, Su YF, Fan GW, Wang YF, Zhu Y, Gao XM. Phytoestrogens from Psoralea corylifolia reveal estrogen receptor-subtype selectivity. Phytomedicine. 2010;17:126–131. doi: 10.1016/j.phymed.2009.05.015. [DOI] [PubMed] [Google Scholar]
  165. Xu L, Du B, Xu B. A systematic, comparative study on the beneficial health components and antioxidant activities of commercially fermented soy products marketed in China. Food Chem. 2015;174:202–213. doi: 10.1016/j.foodchem.2014.11.014. [DOI] [PubMed] [Google Scholar]
  166. Yanagihara N, Zhang H, Toyohira Y, Takahashi K, Ueno K, Tsutsui M, Takahashi K. New Insights Into the Pharmacological Potential of Plant Flavonoids in the Catecholamine System. J. Pharmacol. Sci. 2014;124:123–128. doi: 10.1254/jphs.13r17cp. [DOI] [PubMed] [Google Scholar]
  167. Yang L, Allred KF, Dykes L, Allred CD, Awika JM. Enhanced action of apigenin and naringenin combination on estrogenreceptor activation in non-malignant colonocytes: Implications on sorghumderived phytoestrogens. Food Funct. 2014;6:749–755. doi: 10.1039/c4fo00300d. [DOI] [PubMed] [Google Scholar]
  168. Yang O, Kim HL, Weon J, Seo YR. Endocrine-disrupting Chemicals: Review of Toxicological Mechanisms Using Molecular Pathway Analysis. J. Cancer. Prev. 2015;20:12–24. doi: 10.15430/JCP.2015.20.1.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Zavatti M, Bertoni L, Maraldi T, Resca E, Beretti F, Fuida M, La Sala GB, De Pol A. Critical-size bone defect repair using amniotic fluid stem cell/collagen constructs: effect of oral ferutinin treatment in rats. Life Sci. 2015;121:174–183. doi: 10.1016/j.lfs.2014.10.020. [DOI] [PubMed] [Google Scholar]
  170. Zhang Q, Lu M, Wang C, Du J, Zhou P, Zhao M. Characterization of estrogen receptor α activities in polychlorinated biphenyls by in vitro dual-luciferase reporter gene assay. Environ. Pollut. 2014;189:169–175. doi: 10.1016/j.envpol.2014.03.001. [DOI] [PubMed] [Google Scholar]
  171. Zhao L, Mao Z, Chen S, Schneider LS, Brinton RD. Early Intervention with an Estrogen Receptor β-Selective Phytoestrogenic Formulation Prolongs Survival, Improves Spatial Recognition Memory, and Slows Progression of Amyloid Pathology in a Female Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. 2013;37:403–419. doi: 10.3233/JAD-122341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Zhao L, Yao J, Mao Z, Chen S, Wang Y, Brinton RD. 17β-Estradiol regulates insulin-degrading enzyme expression via an ERβ/PI3-K pathway in hippocampus: relevance to Alzheimer’s prevention. Neurobiol. Aging. 2011;32:1949–1963. doi: 10.1016/j.neurobiolaging.2009.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Zhou Y, Zhang H, Peng C. Puerarin: A Review of Pharmacological Effects. Phytother. Res. 2014;28:961–975. doi: 10.1002/ptr.5083. [DOI] [PubMed] [Google Scholar]
  174. Zhu JTT, Choi RCY, Li J, Xie JQH, Bi CWC, Cheung AWH, Dong TTX, Jiang ZY, Chen JJ, Tsim KWK. Estrogenic and Neuroprotective Properties of Scutellarin from Erigeron breviscapus: A Drug against Postmenopausal Symptoms and Alzheimer’s Disease. Planta Med. 2009;75:1489–1493. doi: 10.1055/s-0029-1185776. [DOI] [PubMed] [Google Scholar]
  175. Zimmerman MC, Tilghman SL, Bouè SM, Salco VA, Elliot S, Williams KY, Skripnikova EV, Ashe H, Payton-Stewart F, Vanhoy-Rhodes L, Fonseca JP, Corbitt C, Collins-Burow BM, Howell MH, Lacey M, Shih BY, Carter-Wientjes C, Cleveland TE, McLachlan JA, Wiese TE, Beckman BS, Wiese TE, Beckman BS, Burow ME. Glyceollin I, a Novel Antiestrogenic Phytoalexin Isolated from Activated Soy. J. Pharmacol. Exp. Ther. 2006;332:35–45. doi: 10.1124/jpet.109.160382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Ziv-Gal A, Wang W, Zhou C, Jodi A, Flaws JA. The effects of in utero bisphenol A exposure on reproductive capacity in several generations of mice. Toxicology and Applied Pharmacology. 2015;3:354–362. doi: 10.1016/j.taap.2015.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES