Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

12 R. Kiyama, Y.

Wada-Kiyama / Environment International 83 (2015) 11–40

4. Evaluation of estrogen action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27


4.1. Assays for detecting estrogen action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.1.1. Ligand-binding assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.1.2. Reporter-gene assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.1.3. Yeast two-hybrid assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
4.1.4. Transcription assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
4.1.5. Protein assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
4.1.6. Cell assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
4.1.7. Animal test . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
4.1.8. Signaling pathway analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
4.2. Pathway-based risk assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
5. Conclusion and future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31

1. Introduction hormones in the body responsible for the maintenance of homeostasis


and the regulation of developmental processes” (Kavlock et al., 1996).
Estrogen is a female hormone that plays a role in menstrual and During our research for characterization of estrogenic chemicals, we
estrous reproductive cycles. The role of estrogen is thus complex and found that quite a number of chemicals have been reported to be estro-
includes involvement in the physiology of reproductive organs and genic. They were analyzed by a number of different assays and evaluat-
tissues (e.g., breast, ovary and endometrium), lipid metabolism, protein ed by different criteria to judge that they are estrogenic. Furthermore,
synthesis, behavior (e.g., lordosis behavior) and diseases (e.g., cancer their mechanisms of action seem to be quite complicated. As we failed
and neurodegenerative/cardiovascular diseases) (Deroo and Korach, to find a comprehensive list of estrogenic chemicals in literatures, we
2006; Gillies and McArthur, 2010). While three major estrogens, examined potentially estrogenic chemicals one by one to find why
estrone, estradiol and estriol, are known, there are a number of estro- they were reported to be estrogenic.
genic chemicals of natural and industrial origins, as mostly character-
ized by comparing them with the major estrogens. Furthermore, 2. Estrogenic chemicals
estrogenic chemicals are those that are not only directly responsible
for activating or inhibiting estrogen action, but also those indirectly The mechanism of estrogenic signaling at the cellular level is sum-
modulating its action; thus, a variety of chemicals should be considered marized in Fig. 1 (see also Kiyama and Zhu, 2014; Kiyama et al., 2014).
to understand its action. Estrogenic chemicals are thus considered as a Estrogenic signal networks can be categorized into intracellular and ex-
type of important chemicals acting as an endocrine disruptor, which is tracellular types. The genomic pathway, which involves transcription of
defined as “an exogenous agent that interferes with the production, re- target genes, and the non-genomic pathway, which rapidly transduces
lease, transport, metabolism, binding, action or elimination of natural signals mediated by membrane-bound estrogen receptors (ERs) and/

Intracellular Network Extracellular Network


Genomic Pathway Non-genomic Pathway Autocrine/Paracrine
Signaling
Hormone/
Estrogen Estrogen
GF/Cytokine

Membrane Other
ER Receptor
Crosstalk
Bypassing
Signaling
Signaling Signaling Signaling-pathway
Protein
Proteins Protein Analysis (S)
Yeast Two-
hybrid Assay (Y)

Ligand-binding Pol II
Estrogen
Assay (L)
Co-regulator
Nuclear Transcription Protein Function
ER

ERE Target Gene


Protein Cell Assay (C)
Assay (P)
Animal Test (A)
Reporter-gene Transcription
Assay (R) Assay (T)

Fig. 1. Signaling pathways, crosstalk and autocrine/paracrine/homeostatic signaling networks of estrogenic chemicals.
R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40 13

or other receptors through crosstalk and/or bypassing, belong to the in assay systems, which were reported for the chemicals summarized
former, while the pathways of autocrine and/or paracrine signaling, below.
which involve other hormones, growth factors and cytokines, belong
to the latter. The list of chemicals showing contradictory results in estrogenicity.
Phlorizin (Wang et al., 2010), quercetin/naringenin (Marino et al., 2012), naringin
The genomic pathway, or the classical pathway, is initiated by (Guo et al., 2011), epigallocatechin 3-gallate (Kuruto-Niwa et al., 2000), glabridin
the binding of chemicals (or ligands) with the nuclear ERs, ERα (Tamir et al., 2000), enterodiol/enterolactone (Feng et al., 2008),
and ERβ, and the ligand-bound ERs act as transcription factors to 3-methylcholanthrene (Swedenborg et al., 2008), abietic acid/botulin/β-sitosterol
up-regulate or down-regulate the transcription of target genes or (Mellanen et al., 1996), methoxychlor (Lemaire et al., 2006), bifenthrin/permethrin
(Brander et al., 2012), raloxifene (Lee et al., 2011), tamoxifen (Jiang et al., 1995;
estrogen-responsive genes, in transcription machinery containing RNA
Obrero et al., 2002), PCBs (Zhang et al., 2014a) and BDE 47 (Karpeta et al., 2014).
polymerase II and co-regulators of transcription, such as RIP-140,
p300/CBP and SRC-1 (Parker, 1998; Moggs and Orphanides, 2001). On
the other hand, estrogen binds to the membrane or cytoplasmic recep- Biphasic activity, where estrogenic activity was observed at low
tors and stimulates signaling proteins through the non-genomic path- concentrations and anti-estrogenic activity at high concentrations, has
way, which occurs rapidly, sometimes just minutes after stimulation, been reported for phytoestrogens (Wang, 2002). Enterolactone, a lignin,
and involves a number of signaling pathways. This type of signaling stimulates DNA synthesis in MCF-7 cells at 10–50 μM, but inhibits it at
starts by the binding of ligands to membrane/cytoplasmic ERs, and higher concentrations. Enterolactone was shown to be a selective ER ac-
several ERs, including G-protein-coupled estrogen receptor 1 (GPER), tivator through its interaction with ER (Penttinen et al., 2007). SERMs
have been identified. However, several new receptors and variants are the chemicals that show differential (agonist or antagonist) effects
of known receptors were identified, such as ER-X and ER-α36 (see on different tissues or cells, where the effects are based on the modula-
Section 3), increasing the complexity of estrogenic signaling. Estrogen- tion of receptor functions by differential interactions between the
related receptors also mediate estrogenic signaling. Meanwhile, recent receptor and the ligand, which would be beneficial for the treatment of
studies have revealed that intracellular networks cooperate with various hormone-responsive cancer and osteoporosis (Maximov et al., 2013).
types of autocrine and/or paracrine signaling, and thus cells in different Such effects are likely to occur at downstream pathways, such as in the
tissues or locations are also involved in the extracellular networks case of raloxifene (Lee et al., 2011) and tamoxifen (Jiang et al., 1995;
(see Section 3). Obrero et al., 2002), where ER-dependent and -independent modula-
One of the reasons why a number of new signaling pathways were tions of signaling were observed.
identified recently is the availability of a variety of new technologies
to detect estrogenic chemicals (Fig. 1); they revealed the novel charac- 2.1.1. Phenolic estrogens
teristics of chemicals, such as biphasic and metabolic effects detected by Phenolic estrogens are categorized by the number of skeletal
cell proliferation assay, the activity of selective estrogen-receptor mod- carbons (Table 1; Vermerris and Nicholson, 2008). While phenol
ulators (SERMs) detected by cell assay and new estrogen-responsive itself is not estrogenic, simple modifications, such as chlorination
genes detected by DNA microarray assay (see Section 4). Thus, estro- (pentachlorophenol), nitration (nitrophenol) and alkylation
genic chemicals include chemicals or their metabolites that have not (nonylphenol), confer estrogenicity on the derivatives. The derivatives
only estrogenic effects but also anti-estrogenic ones, and they transduce of phenolic acid and phenolic aldehyde, such as gallic acid, parabens,
signals to estrogen signaling through crosstalk, bypassing and extracel- benzaldehydes, syringic acid and ellagic acid, show estrogenicity. More
lular networks. complex phenolics and their derivatives, including phenylethanoids
(acteoside and martynoside), tyrosine derivatives (thyroid hormone),
phenylacetic acid, naphthoquinones, bisphenols, benzophenones and
2.1. Structure-based categorization of estrogenic chemicals phenylpropanoids, also show estrogenicity. Phenylpropanoids are a
group of chemicals synthesized by plants from phenylalanine, and
To understand the action of estrogenic chemicals at the molecular include hydroxycinnamic acids (caffeic acid and ferulic acid),
level, especially at the cell signaling level, we categorized estrogenic phenylpropenes (anethole and eugenol), coumarins and related
chemicals based on several characteristics. First, the estrogenic chemicals (auraptene, bergapten, daphnetin, esculetin, icariin and
chemicals categorized by structure are summarized in Table 1. A icaritin), stilbenes/stilbenoids, anthraquinones, chalcones/chalconoids
number of estrogenic chemicals are phenolics, and phenolic and flavones/flavonoids (anthoxanthins, flavanones, flavanonols, flavans,
estrogens include simple phenols, phenolic acids/phenolic alde- anthocyanidines, isoflavones, isoflavanes, isoflavenes, coumestans,
hydes, acetophenones, tyrosine derivatives, phenylacetic acids, pterocarpans and flavonolignans).
hydroxycinnamic acids, phenylpropenes, coumarins/isocoumarins/ PAHs are known to show estrogenicity, which is partly explained by
chromones, naphthoquinones, bisphenols, benzophenones, stil- the presence of hydroxylated phenolic rings (3,9-benz[a]anthracene
benes/stilbenoids, anthraquinones, chalcones/chalconoids, fla- diols and cinanthrenol A; Table 2) or by the introduction of a hydroxyl
vones/flavonoids, lignans/neolignans, diarylheptanoids, flavolans group after hydroxylation or metabolization within a cell.
and hydroxylated polycyclic aromatic hydrocarbons (PAHs). On the
other hand, chemicals without a phenolic ring can also show estrogenic 2.1.2. Non-phenolic estrogens
activity, which include anilines, carboranes, indoles, metalloestrogens, A number of non-phenolics have been identified as estrogenic
perfluorinated compounds, phthalates, PAHs and terpenes/terpenoids chemicals, which include anilines, carboranes (boron estrogens), indoles,
(monoterpenes, sesquiterpenes, diterpenes, triterpenes, tetraterpenes, metalloestrogens, perfluorinated compounds, phthalates, PAHs and ter-
sterols, steroids, saponins and meroterpenes). penes/terpenoids (Table 1). Anilines are aromatic amines and used as
While estrogenic or anti-estrogenic activity can be detected by vari- precursors to polyurethane, synthetic dyes and other industrial
ous methods (see Section 4), they have limitations in their sensitivity chemicals. Because of similarities in their structure and biological activity,
(e.g., concentration-dependent activity), distinguishing agonists/ anilines were expected to show estrogenicity (Hamblen et al., 2003).
antagonists and specificity (e.g., cell/tissue types and signaling path- Carboranes are a group of chemicals composed of boron, carbon and hy-
ways). Thus, the same estrogenic chemicals can show contradictory drogen atoms in polyhedral forms, and have advantages for designing
results (listed in Tables 1 and 2), such as differences in estrogenic and new ER agonists/antagonists and SERMs due to their unique three-
anti-estrogenic, agonistic and antagonistic, and ligand/pathway- dimensional structure and superior synthetic flexibility (Armstrong and
dependent and -independent responses, differences in increased and Valliant, 2007). Indoles consist of a benzene ring fused to a pyrrole ring.
decreased gene expression or enzymatic activity, as well as differences Some indoles show estrogenicity without having a phenolic hydroxyl
18 R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40

Table 1 (continued)

Estrogenic chemical Signaling pathway Reference (assaya)

Saponin
Ginsenoside Rg1 (estrogenic) ER Gao et al., 2014 (P, R, T)
Ginsenoside Rg3 ER/PI3K/Akt/AMPK/NO/cardioprotection Hien et al., 2010 (S)
Ginsenoside Rh2 (estrogenic) ER Kovalchuk et al., 2006 (Y)
Ginsenoside Re ER/NO/K channel/vasodilation Nakaya et al., 2007 (S)
Gypenoside XVII ER/PI3K/Akt/Nrf2/ARE/autophagy Meng et al., 2014 (S)
Protopanaxadiol, protopanaxatriol ERβ/GR/NO/cardioprotection Leung et al., 2009 (S)
Soysaponin I (estrogenic) ER Ali et al., 2009 (R)
Withaferin A ERα/MAPK/p53/apoptosis Zhang et al., 2011 (S)
Meroterpene
Bakuchiol (estrogenic) ER Lim et al., 2011 (C, L, R)

Estrogenic chemicals categorized by structure are shown along with their effects, such as estrogenic and anti-estrogenic.
a
Abbreviations for assays are: animal test (A), cell-proliferation assay (C), ligand-binding assay (L), proten assay (e.g., Western blotting and immunoassay) (P), reporter-gene assay (R),
signaling pathway analysis (S), transcription assay (e.g., RT-PCR and DNA microarray assay) (T) and yeast two-hybrid assay (Y).
b
Phenolics are categorized by the number of skeletal carbons according to Vermerris and Nicholson (2008). AhR: arylhydrocarbon receptor; AMPK: AMP-activated protein kinase;
CaMKII: Ca2+/calmodulin-dependent protein kinase; EGFR: epidermal growth factor receptor; ER: estrogen receptor; ERK: extracellular signal-regulated kinase; FOXO: Forkhead box O;
GPCR: G-protein-coupled receptor; GPER: G-protein-coupled estrogen receptor 1; GR: glucocorticoid receptor; HIF-1α: hypoxia-inducible factor 1; IGF-1R: insulin-like growth factor 1
receptor; IL: interleukin; IRS-1: insulin receptor substrate-1; JNK: c-Jun N-terminal kinase; LXR: liver X receptor; MAPK: mitogen-activated protein kinase; mER: membrane estrogen
receptor; MT: melatonin receptor; NO: nitrogen oxide; PARP: poly(ADP-ribose) polymerase; PDGF: platelet-derived growth factor; PI3K: phosphoinositide 3-kinase; PKA: protein kinase
A; PPAR: peroxisome proliferator-activated receptor; PR: progesterone receptor; RARα: retinoic acid receptor α; SERM: selective estrogen receptor modulator; TGF-β: transforming
growth factor β; TNF-α: tumor necrosis factor α; TrkA receptor: neurotrophic tyrosine kinase receptor type 1.

group (Table 1). Metalloestrogens are inorganic metal ions that bind to 2007) and to be important for the development of SERMs (Baker,
and activate ERs, and are classified into groups of metal/metalloid anions 2013), while B-ring, which is saturated in the above compounds but un-
and bivalent cationic metals (Byrne et al., 2013). Estrogenic activity has saturated in equilin and equilenin, is associated with the affinity for ERs
been reported for arsenic, antimony, barium, cadmium, calcium, chromi- and the differential actions between ERα and ERβ (Bhavnani et al.,
um, cobalt, copper, lead, lithium, mercury, nickel, selenium, tin, vanadate 2008; Bhavnani and Stanczyk, 2014).
and zinc (Table 1). Perfluorinated compounds, such as fluorotelomer al-
cohols and perfluorohexane/perfluorooctane sulfonates, are 2.2. Function-based categorization of estrogenic chemicals
fluorocarbon-based oligomers, synthesized by telomerization. Volatile
fluorotelomer alcohols form perfluorinated carboxylic acids, such as Estrogenic chemicals can be categorized by applications, usage and ef-
perfluorooctanoic acid (PFOA) and perfluorononanoic acid (PFNA), by fects (Table 2): They are related to various types of food additives/
biodegradation, which persist in the environment and are found in the dietary supplements, pesticides (antimicrobials/essential oils, biocides,
blood of humans and wildlife. They have been shown to be estrogenic. disinfectants/sanitizers, fungicides, herbicides, insecticides, miticides, re-
Phthalate esters are esters of phthalic acid and alcohols, and widely pellents, rodenticides and pheromones), pharmacological estrogens
used as plasticizers. Various phthalate esters, such as bis(2-ethylhexyl) (mycoestrogens, nonsteroidals, pharmaceuticals, pure ER antagonists,
phthalate, butyl benzyl phthalate, dibutyl phthalate, diethyl phthalate, SERMs and steroidals), plasticizers (phthalate/trimellitate/salicylate
diisobutyl phthalate, diisononyl phthalate and di(n-butyl) phthalate, esters and organophosphates) and pollutants (heavy metals or
were found to show estrogenicity by various technologies (Table 1). Fi- metalloestrogens, persistent organic pollutants or POPs, pesticide resi-
nally, terpenes/terpenoids are a group of chemicals having isoprene dues, pharmaceutical pollutants and PAHs). Note that these categories
units, (C5H8)n, and their derivatives, such as mono-, sesqui-, di-, tri- are arbitrary, so many estrogenic chemicals may belong to more than
and tetra-terpenes, as well as a diverse group of meroterpenes/sterols/ one category.
steroids/saponins. Some of them are estrogenic (Table 1).
2.2.1. Food additives/dietary supplements
2.1.3. Chemical basis for estrogen action The categories of food additives and dietary supplements contain a
The structural basis of estrogenicity has been examined by compar- number of chemicals with structural diversity (Table 2). The categories
ing the activity of chemicals before and after substituting chemical of active components, antioxidants/preservatives, color additives,
groups. Anstead & Kym examined benz[a]anthracene and its analogs cosmetics, emulsifiers/thickeners, flavor enhancers and vitamins consist
by molecular modeling, and found that a polycyclic planar ring system of phenolics and chemicals having a phenolic ring (e.g., coumestrol,
mimicking the steroid AB-ring is an important feature for the binding gallate, hydroxyanisole and tetrahydrocannabinol), unsaturated hydro-
affinity for ERs and carcinogenicity (Anstead and Kym, 1995). Routledge carbons (e.g., carotenes, linoleic acid and lycopene), azo compounds
& Sumpter examined the estrogenicity of alkylphenols with different (e.g., tartrazine and sunset yellow), polysaccharides/sugar derivatives
structural features (differences in size, branching and position) by a (e.g., carrageenan, cyclodextrins and guanylate), saponins (ginsenosides
yeast reporter gene assay, and found that alkylphenols substituted at and notoginsenosides), and vitamins and their metabolites (e.g., niacin
the 4-position with 6- to 8-carbon-containing, tertiary alkyl groups and tocopherols). The chemicals in these categories have roles in various
exhibited higher activities (Routledge and Sumpter, 1997). By examin- biological effects: osteoporosis (caffeine; Zhou et al., 2009), apoptosis
ing 120 aromatic chemicals, Schultz et al. found the importance of the (geniposide; Li et al., 2014b), cardioprotection (nectandrin B; Hien et al.,
hydroxyl groups at positions 3 (C3–OH; A-ring) and 17 (C17–OH; 2011), neuroprotection (notoginsenoside R1; Meng et al., 2014), vascular
D-ring) of 17β-estradiol and hydrophobicity of its B- and C-rings relaxation (notoginsenoside Ft1; Shen et al., 2014), cell proliferation (vita-
(Schultz et al., 2002). Hamblen et al. further examined the importance min D; Gilad et al., 2005) and inflammation (niacin; Santolla et al., 2014).
of C3–OH by substituting it (4-substituted phenols) with the amino
group (4-substituted anilines) and found that 4-substituted anilines 2.2.2. Pesticides
showed activities at least 105 times lower than that of 17β-estradiol Pesticides are substances intended for preventing, destroying,
(Hamblen et al., 2003). C3–OH, which is present in 17β-estradiol, repelling or mitigating any pest, and include avicides, bactericides, disin-
17α-estradiol and 17α-ethinylestradiol but absent in mestranol and fectants (antimicrobials), fungicides, herbicides, insecticides, miticides,
quinestrol, is suggested to confer anti-apoptotic behavior (Mann et al., molluscicides, nematicides, predacides, piscicides, repellents,
22 R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40

Table 2 (continued)

Estrogenic chemical Signaling pathway Reference (assaya)

Pesticide residue (see pesticide shown above)


Pharmaceutical pollutant (see pharmacological estrogen
shown above)
Polycyclic aromatic hydrocarbon (see Table 1)

Estrogenic chemicals categorized by applications, usages and effects are shown along with their effects, such as estrogenic and anti-estrogenic. Characterization of estrogenicity, such as
estrogenic/anti-estrogenic and agonist/antagonist, is based on the description in each reference. Only a representative category for each chemical is shown.
a
Abbreviations for assays are: animal test (A), cell-proliferation assay (C), ligand-binding assay (L), proten assay (e.g., Western blotting and immunoassay) (P), reporter-gene assay (R),
signaling pathway analysis (S), transcription assay (e.g., RT-PCR and DNA microarray assay) (T) and yeast two-hybrid assay (Y). AhR: arylhydrocarbon receptor; BHC: benzene hexachlo-
ride; CAR: constitutive androstane receptor; DDD: dichlorodiphenyldichloroethane; DDE: dichlorodiphenyldichloroethylene; DDT: dichlorodiphenyltrichloroethane; E2: estradiol; ER: es-
trogen receptor; ERK: extracellular signal-regulated kinase; GPER: G-protein-coupled estrogen receptor 1; HCH: hexachlorocyclohexane; IGF-1R: insulin-like growth factor 1 receptor;
IRS-1: insulin receptor substrate-1; MAPK: mitogen-activated protein kinase; mER: membrane estrogen receptor; NO: nitrogen oxide; PBDE: polybrominated diphenyl ether; PCB:
polychlorinated biphenyl; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A; PKC: protein kinase C; PPAR: peroxisome proliferator-activated receptor; RAR: retinoic acid receptor;
ROS: reactive oxygen species; SERM: selective estrogen receptor modulator; SXR: steroid and xenobiotic receptor; TCDD: 2,3,7,8-tetrachlorodibenzo-p-dioxin; TGF-β: transforming
growth factor β; TNF-α: tumor necrosis factor α; VDR: vitamin D receptor; VEGF: vascular endothelial growth factor.

rodenticides, defoliants, desiccants and growth regulators (Randall gene activation in different cells (Ruh et al., 1995; Gould et al., 1998;
et al., 2013). The pesticides within a particular class may share a com- Gaido et al., 1999; Yoon et al., 2001; Safe et al., 2001).
mon mode of action due to their structural similarity, and several chem-
ical groups, such as carbamates, organochlorides, organometallic 2.2.4. Plasticizers
compounds, organophosphates, pyrethroids and sulfonylureas, have Plasticizers are used to increase the plasticity or fluidity of mainly
been used. Many of them show estrogenic activity (Table 2), such as plastics, and some dicarboxylic and tricarboxylic esters, such as phthalate
those summarized below. and trimellitate esters, salicylate esters, such as benzyl salicylate (a tissue
conditioner), and organophosphates, such as tricresyl phosphate and
The list of pesticides showing estrogenic activity. triphenyl phosphate, used as plasticizers, show estrogenicity (Table 2).
Aldicarb, bendiocarb, carbaryl, methiocarb, methomyl, oxamyl, pirimicarb and Phthalates and trimellitates are di- or tri-carboxylic acid derivatives of
propoxur (carbamates); aldorin, atrazine, chlordane, chlordecon, chloroxylenol, benzene, respectively, and they do not have a phenolic hydroxyl group.
DDT, dichlorophenyls, dicofol, dieldrin, endosulfan, endrin, fenarimol, fenvalerate,
Estrogenicity is increased when the mass of participating alcohols in es-
hexachlorobenzene, hexachlorocyclohexanes, hexachlorophene, methoxychlor,
nitrofen, procymidone, propiconazole, terbuthylazine, toxaphene, triadimefon, ters becomes large (Parveen et al., 2008). Meanwhile, organophosphates
triclopyr, triclosan and vinclozolin (organochlorides); tributyltin (an organometallic are phosphoric acid esters of alcohols and used widely for solvents,
compound); chlorpyrifos, diazinon, fenthion, malathion, parathion and quinalphos pesticides and plasticizers, but they may cause adverse neuropsychiatric
(organophosphates); eugenol, nitrophenol and 4-phenylphenol (phenolics);
and neurological effects, such as on the neurobehavioral development of
bifenthrin, cyfluthrin, cyhalothrin, cypermethrin, deltamethrin, etofenprox,
permethrin, sumithrin and tetramethrin (pyrethroids); and sulfonylureas.
fetuses and children, by inhibiting acetylcholinesterase in nerve cells
(Chen, 2012). Tricresyl phosphate and triphenyl phosphate are esters of
cresol or phenol, respectively, and while there is no phenolic ring, they
Estrogenic pesticides bind to ERs and affect the downstream signaling
interact with the ER and show anti-proliferative effects through the
pathways, such as the ER/ERK pathway for cell proliferation (tributyltin,
NF-κB pathway (Sabatucci et al., 2006).
hydroxytyrosol, oleuropein, diedrin, procymidone and methoxychlor),
the ER/ERK/Akt/P70S6K pathway for cardioprotection (brefeldin A), the
2.2.5. Pollutants
ER/IGF-1 pathway for cell proliferation (hexachlorobenzene), the mem-
Pollutants can be tentatively categorized into heavy metals and
brane ER/K channel pathway for insulin secretion (sulfonylurea), the
organic compounds, although any compounds found in the environ-
ER/cyclin D1/Ras/Bax pathway for apoptosis (methoxychlor) and the
ment can be considered as pollutants. The organic pollutants include
ryanodine receptor/ER/PKCε/PKA/Ca2+ pathway in the sweat gland
POPs (Wang and Needham, 2007), pesticide residues, pharmaceutical
(ryanodine). Note that the same chemical may act as an estrogenic or
pollutants and PAHs, such as the chemicals derived from industrial
an anti-estrogenic depending on the assay used (e.g., bifenthrin, chlorpyr-
products, including pesticides, pharmaceuticals, plastics and volatile or-
ifos and permethrin) or the receptors involved (such as methoxychlor)
ganic compounds (e.g., those in paint and coatings), and their deriva-
(see Table 2).
tives, and most of the categories include estrogenic chemicals
(Table 2). Estrogenic chemicals of industrial origins are those listed in
2.2.3. Pharmacological estrogens the categories of pesticides, pharmacological estrogens and plasticizers
Pharmacological estrogens can be categorized into the groups of (see above), while those of other industrial origins or their byproducts
mycoestrogens, nonsteroidals, pharmaceuticals, pure ER antagonists, are DDT, dioxins/dioxin-like compounds (Aroclors, PCBs and TCDD),
SERMs and steroidals (Pazaiti et al., 2012). Mycoestrogens are estrogens hexachlorocyclohexanes (HCHs) and PBDEs/PCDEs.
produced by fungi, and include zearalenone, which is widely contami-
nated in agricultural products as a mycotoxin. Zearalenone and its de- 3. Molecular mechanisms of estrogenic signaling
rivatives interact with ERα and ERβ in a manner similar to estrogen
and thus have been used as anabolic agents for animals and also pro- The mechanism of estrogenic signaling can be divided into pathways
posed for hormonal replacement therapy in postmenopausal women and networks (Fig. 1). The pathways that direct the signal to specific
or as oral contraceptives (Pazaiti et al., 2012). A number of pharmaceu- functional outcomes, such as apoptosis, carcinogenesis, cell growth/pro-
ticals, such as those used for the replacement or addition of estrogen liferation, differentiation/development and inflammation, are mostly
(diethylstilbestrol, pure ER agonists, SERMs and steroids) or for other initiated by the binding of estrogen or estrogenic chemicals to ERs.
purposes (acetaminophen, aspirin, bezafibrate, ephedrine, fenofibrate, ERs can be classified into the groups of nuclear and membrane recep-
gemfibrozil and tocotrienol), are estrogenic (Table 2). Note that a num- tors, which transduce signals through various pathways (Table 3). Spe-
ber of chemicals other than those listed in Table 2 have been reported to cific signaling pathways, such as PI3K, MAPK/ERK and NF-κB signaling
be SERMs, which include those categorized as synthetic estrogens and (Ribeiro and Freiman, 2014), can be found in the pathways for different
phytoestrogens, such as bisphenol A, methoxychlor, naringenin, outcomes as functional modules, where specific signaling pathways and
nonylphenol, octylphenol and resveratrol, based on the differences in functional outcomes can be selected by selecting specific effectors.
24 R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40

Table 3 (continued)

Receptor signaling pathway Reference

Other (continued)
Melatonin/α1/β-adrenoceptor/ER/cAMP/pinealocytes Hernández-Díaz et al., 2001
Methyl-amoorain + DMBA/ER/Wnt/β-catenin/carcinogenesis Mandal et al., 2013
Protopanaxadiol, protopanaxatriol/ERβ/GR/NO/cardioprotection Leung et al., 2009
Resveratrol/ER/TGF-β2/Smad/lung epithelial cells Suenaga et al., 2008
Ryanodine/ryanodine receptor/ER/PKCε/PKA/Ca2+/sweat gland Muchekehu and Harvey, 2008
Tamoxifen/EGFR/HER2/MAPK/Akt/ER/proliferation Osborne et al., 2005 (review)
Thyroid hormone/TR/ERα/proteolysis Alarid et al., 2003
Thyroid hormone/TR/integrin receptor/ERα/proliferation Meng et al., 2011
Autocrine/paracrine signaling
Autocrine signaling
E2/ER/Akt/IGF-1R/InsR/breast cancer Fox et al., 2013
E2/ER/ERBB4/PR/cell growth Zhu et al., 2006
E2/ER/IGF/IGF-1R/breast cancer Hamelers et al., 2003
E2/ER/IGF-1R/MAPK/cyclin D1, cyclin E/cell growth Kashima et al., 2009
E2/ER/MAPK/PDGF/PDGFR/proliferation Barbarisi et al., 2001
E2/ER/PDGF/PDGFR/ERK/proliferation Finlay et al., 2003
Exemestane/ER/amphiregulin/EGFR/MAPK/proliferation Wang et al., 2008a
Wnt/EGFR/ERK/ER/proliferation (crosstalk) Schlange et al., 2007
Paracrine signaling
Adiponectin/APNR/ERβ/differentiation Rahal and Simmen, 2011
E2/ER/Factor X/coagulation factor VII/Thrombin/development Jazin et al., 1990
E2/ER/FGF/FGFR/Tbx3/carcinogenesis Fillmore et al., 2010
E2/ER/TGF-β1/TGF-βR/Smad2/thyroid Gantus et al., 2011
E2/ER/Wnt/β-catenin/AXIN2/proliferation Ono et al., 2013
E2/ERα/JNK/cardioprotection Mahmoodzadeh et al., 2014
MCP-1/CCRs/PI3K/Akt/mTOR/ERα/proliferation (ligand-independent) Riverso et al., 2014
Autocrine/paracrine signaling
Androgen/AR/E2/ERα, ERβ/spermatogenesis Pearl et al., 2011
E2/ER/ANF/ANF receptor/cGMP/anti-hypertrophy Babiker et al., 2004
E2/ER/IGF-1/IGF-1R/osteoblasts Kassem et al., 1998
E2/ER/NF-κB/cytokines/cytokine receptors/inflammation, etc. Härkönen and Väänänen, 2006 (review)
E2/ER/STAT3/IL-6/IL6R/apoptosis Wang et al., 2001
E2/ERα/CXCL12/CXCR4/MAPK/proliferation Hall and Korach, 2003
E2/ERβ/CXCL12/CXCR4/MAPK/proliferation Sauvé et al., 2009
17α-E2/ER-X/MAPK/ERK/PI3K/Akt/brain Toran-Allerand et al., 2005

Estrogen-signaling pathways are categorized by the types of receptors and the ways of signaling. AhR: arylhydrocarbon receptor; ANF: atrial natriuretic factor; APNR: adiponectin receptor;
AR: androgen receptor; CAR: constitutive androstane receptor; CCRs: CC chemokine receptors; DDE: dichlorodiphenyldichloroethylene; DMBA: 7,12-dimethylbenz(a)anthracene; E2: es-
tradiol; ER: estrogen receptor; ERK: extracellular signal-regulated kinase; ERR: estrogen-related receptor; FOXO: Forkhead box O; GPER: G-protein-coupled estrogen receptor 1; GR: glu-
cocorticoid receptor; IGF: insulin-like growth factor; IGF-1R: IGF-1 receptor; IL-1: interleukin 1; IL6R: interleukin 6 receptor; InsR: insulin receptor; IRS-1: insulin receptor substrate-1;
JNK: c-Jun N-terminal kinase; LXR: liver X receptor; MAPK: mitogen-activated protein kinase; mER: membrane estrogen receptor; mTOR: mammalian target of rapamycin; NO: nitrogen
oxide; PDGF: platelet-derived growth factor; PDGFR: PDGF receptor; TrkA receptor: neurotrophic tyrosine kinase receptor type 1; PI3K: phosphoinositide 3-kinase; PKA: protein kinase A;
PKC: protein kinase C; PR: progesterone receptor; RAR: retinoic acid receptor; SERM: selective estrogen receptor modulator; TCDD: 2,3,7,8-tetrachlorodibenzo-p-dioxin; TGF-β:
transforming growth factor β; TGFBRs: TGF-β receptors; TR: thyroid hormone receptor; VDR; vitamin D receptor; VEGF: vascular endothelial growth factor; VEGFR-1:VEGF receptor 1.

Signaling pathways can influence each other not only through signaling 14q23.2–q23.3 on human chromosomes, respectively. These receptors
pathways by crosstalk (exchanging signals between different receptors have similar molecular sizes (595 amino acids for ERα and 530 amino
and/or pathways) and/or bypassing (signaling uni-directionally from acids for ERβ) and share a common structural architecture composed
one pathway to another) at the intracellular level, but also through the se- of three functional domains: the A/B domain at the N-terminal is in-
cretion of hormones or growth factors to transduce signals to different volved in transcriptional activation of estrogen-responsive genes (AF-
cells or tissues, which may result in completely different types of func- 1); the C domain contains a two-zinc-finger structure responsible for re-
tional outcome. Feedback regulation of such signaling networks through ceptor dimerization and DNA binding; and the E/F domain at the C-
crosstalk/bypassing and autocrine/paracrine networks would make sig- terminal mediates ligand binding, receptor dimerization, nuclear trans-
naling more complex, which may also contribute to pathways other location and transactivation of target gene expression (AF-2) in associ-
than estrogen signaling and eventually result in forming a homeostatic ation with co-regulators (Nilsson et al., 2001). While no ligand binds to
network. the A/B domain and thus AF-1 is ligand-independent, ligands bind to the
E/F domain and thus AF-2 is affected by ligands. Furthermore, ESR1 and
3.1. Receptors ESR2 are known to show splice variants, which add variation in terms of
cell/tissue-specific expression, ligand-binding specificity, cell localiza-
Estrogenic chemicals first bind to ERs to initiate cell signaling. There tion and cell functions (Taylor et al., 2010). Transactivation of target
are three major ERs, in which ERα and ERβ are nuclear ERs and the G- genes is mediated by the binding of ERs with estrogen-responsive
protein-coupled estrogen receptor 1 (GPER) is a membrane ER. In addi- genes with specific sequence motifs, such as GGTCACAGTGACC (Klein-
tion, recent studies have revealed the presence of other ERs, such as the Hitpass et al., 1986), often with the help of other transcription factors,
estrogen-related receptors (ERRs), variants of ERα or ERβ (ER-X and such as Sp1 and AP1 (Safe, 2001).
ER-α36) and uncharacterized ERs. These receptors share some common The estrogenic chemicals that transduce signals through ERα and/or
features, although there are differences in structure, signaling pathways, ERβ are summarized in Tables 1 and 2. While they usually cannot distin-
cell specificity and functional outcomes. guish between ERα and ERβ, some show a preference or exclusivity for
one of these (Escande et al., 2006). The difference in transactivation ac-
3.1.1. Nuclear receptors tivity of various compounds between ERα and ERβ was investigated,
There are two major nuclear ERs, ERα and ERβ, which are encoded where some phytoestrogens showed significant preferences in the
by different genes, ESR1 located at 6q25.1 and ESR2 located at binding affinity (Kuiper et al., 1998). Such differences in estrogenic
R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40 25

action between ERα and ERβ have been investigated by using selective with other signaling pathways, such as GPER/TGF-β1 crosstalk in
antagonists against each receptor, such as 4,4ʹ,4″-(4-propyl-[1H]- fulvestrant-induced cell migration (Li et al., 2014a) or icariin-induced
pyrazole-1,3,5-triyl)trisphenol (PPT) for ERα (Stauffer et al., 2000) expression of type IV collagen (Li et al., 2013a), GPER/IL-1β crosstalk
and 2,3-bis (4-hydroxyphenyl)-propionitrile (DPN) for ERβ (Meyers in genistein-induced inflammatory response (Luo et al., 2012), and
et al., 2001) (Table 3). GPER/EGFR crosstalk in equol-induced NO synthesis (Rowlands et al.,
Recently, another nuclear receptor family, estrogen-related receptor 2011), icariin- or icaritin-induced cell proliferation (Ma et al., 2014) or
(ERRs), was found to be partly involved in estrogen signaling. ERRs niacin-induced inflammatory response (Santolla et al., 2014). GPER is
(ERRα, ERRβ and ERRγ) act as ligand-dependent transcription factors, known to inhibit serotonin 1A receptor selectively (Xu et al., 2009;
although no natural ligand has been identified. ERRs regulate the McAllister et al., 2012; Akama et al., 2013) and to accelerate the efficacy
genes that are distinct from ER-regulated genes and are associated of its inhibitor fluoxetine (Li et al., 2013c) in neuronal cells, so it is a
with physiological functions, such as negative regulation of osteoblast target to treat mood disorders.
differentiation (Gallet and Vanacker, 2010). Estrogenic signaling medi-
ated by ERRs has been reported for biochanin A, daidzein, genistein
and resveratrol (estrogenic), as well as apigenin, chlordane, diethylstil- 3.1.3. Other membrane receptors
bestrol, kaempferol and toxaphene (Table 3). Crosstalk between ER and Several groups of researchers found that cell signaling is initiated by
ERRs was reported for resveratrol-induced functional modulation of the unidentified or unspecified membrane receptors (Soltysik and Czekaj,
respiratory chain (Lopes Costa et al., 2014). 2013). Some of them are likely to be variants of ERα or ERβ, while
the others are likely to be factors other than ERα, ERβ, ER-X or GPER
3.1.2. Membrane receptors (Nethrapalli et al., 2005). Endogenous estrogens (E1, E2 and E3) and
The presence of membrane ERs (mERs) was evidenced by rapid, thus estrogenic chemicals, such as DDE, 17α-E2, geniposide, naringenin
non-genomic, mechanisms of estrogen signaling, which were observed (anti-estrogenic), raloxifene, retinoic acid, sulfonylurea and tamoxifen,
as rapid alterations in the levels of cAMP, cGMP and Ca2+, and in the bind to mER and transduce signals through pathways, such as ERK,
activity of enzymes (Soltysik and Czekaj, 2013). There are several NF-κB and PI3K/MAPK signaling, to contribute to apoptosis, cell prolifer-
mERs, which are classified by their structure: canonical ERα and ERβ ation, differentiation and other cell functions (Table 3). Crosstalk was
translocated to the membrane after modification, variants of ERα and implicated between mER and other receptors, such as natriuretic pep-
ERβ (e.g., ER-X and ER-α36), GPER and less characterized mERs. Canon- tide receptor (Stratton et al., 2010), RAR (Kauss et al., 2008), and IGFR
ical ERα and ERβ bind to caveolin-1 after being palmitoylated, which are and EGFR in signalosomes (Soltysik and Czekaj, 2013) or K channels
translocated to the membrane and anchored there as a dimer (Soltysik (Nadal et al., 1998).
and Czekaj, 2013). They mediate rapid signaling for hypothalamic func-
tion (Micevych and Kelly, 2012). 3.2. Cell functions and signaling pathways
ER-X is a 62–63 kDa membrane protein likely to be derived from
ERα as a splicing variant (Soltysik and Czekaj, 2013). ER-X is activated There are a number of signaling pathways linked to estrogen and
with 17α-E2, and associated with the function of the brain and the estrogenic chemicals (a comprehensive list is shown in Kiyama et al.,
uterus through MAPK and ERK signaling (Table 3). ER-X could be 2014), which are associated with specific cell functions, such as chroma-
involved in the E2-induced inhibition of Ca2+ influx and contraction in tin/epigenesis, apoptosis, autophagy, cellular metabolism, translational
murine cardiomyocytes derived from ERα/ERβ-knockout mice (Ullrich control, cell cycle/DNA damage/cytoskeletal formation, immunology/
et al., 2008). inflammation response, neurological diseases and development/
ER-α36 is a splice variant of ERα lacking both AF-1 and AF-2 but differentiation. Here, we discuss further the role of estrogenic chemicals
retaining the DNA-binding domain and the partial ligand-binding do- in some cell functions: apoptosis, carcinogenesis, cell growth and prolif-
main, and is predominantly localized at the membrane. ER-α36 inhibits eration, differentiation/development and inflammation.
wild-type ERα (ER-α66) and ERβ in a dominant-negative manner and
thus contributes to the resistance of breast cancer to endocrine therapy
(Rao et al., 2011). ER-α36 mediates estrogenic signaling through the 3.2.1. Apoptosis
MAPK/ERK pathway in breast cancer cells and c-Src-mediated signaling Estrogen is known to stimulate growth and inhibit apoptosis
in gastric cancer cells (Table 3). ER-α36 is involved in testosterone- (see Section 3.2.3). However, recent progress has revealed that estrogen
initiated carcinogenesis by transducing estrogenic signals after testos- can induce apoptosis through several pathways, such as Fas/FasL, mito-
terone is converted to estrogen (Lin et al., 2009). chondrial, NF-κB and PI3K/Akt pathways (Lewis-Wambi and Jordan,
GPER, previously known as G-protein-coupled receptor 30 (GPR30), 2009). Fas/FasL plays a key role in the extrinsic pathway, a type of
is a G-protein-coupled receptor (GPCR) encoded by the GPER gene lo- signaling of external origin, while the mitochondrial pathway is induced
cated at chromosome 7p22.3 with high expression in the hypothala- by various stresses and involves p53, Bax and other pro-apoptotic
mus, pituitary gland, adrenal medulla, renal pelvis and ovary (Hazell proteins. Estrogenic chemicals are also related to either the inhibition
et al., 2009; Soltysik and Czekaj, 2013). GPER is a 7-membrane-spanning or the induction of apoptosis (Tables 1 and 2): Aroclor 1254, caffeic
protein with high affinity for E2 and other chemicals such as aldoste- acid phenethyl ester, epigallocatechin 3-gallate, formononetin, 25-
rone; thus, the binding of estrogen to GPER induces rapid non- hydroxycholesterol, irigenin, mestranol, naringenin, plumbagin, quer-
genomic signaling. Other than the cell membrane, GPER has been cetin, quinestrol, tamoxifen, tectorigenin, withaferin A and wogonin
reported to be located at the endoplasmic reticulum, Golgi apparatus (pro-apoptotic); and equilenin, equilin, geniposide, ginsenoside Rb1,
and even in the nucleus (Soltysik and Czekaj, 2013). GPER can be methoxychlor, raloxifene and triclosan (anti-apoptotic). Furthermore,
activated directly by the binding of ligands other than estrogen, such estrogenic chemicals transduce signals through specific signaling path-
as bisphenol A, daidzein, dieldrin, diethylstilbestrol, equol, fulvestrant, ways: Aroclor 1254 (ERα/cyclin D1/Bcl-2/caspase-3), epigallocatechin
genistein, icariin, icaritin, kepone, niacin, nonylphenol, quercetin, ralox- 3-gallate (ER/MAPK/Akt/caspase-3), formononetin (ER/Ras/MAPK/
ifene, resveratrol and zearalenone as agonists to the receptor (Table 3). Bax/Bcl-2), naringenin (ERα, ERβ/MAPK/caspase-3), plumbagin (ER/
Kajta et al. used two tetrahydro-3H-cyclopenta[c]quinoline analogs, NF-κB), quercetin (ERβ/caspase-3), withaferin A (ERα/MAPK/p53)
G15, a selective GPER antagonist, and G1, a selective GPER agonist, to and wogonin (ER/PARP/Bax) (pro-apoptotic); and E2 (ER/STAT3/IL-6/
study the neuroprotective effect of daidzein (Kajta et al., 2013). GPER IL6R), geniposide (mER/PI3K/MAPK), ginsenoside Rb1 (ER/ERK/Akt),
is involved in the signaling pathways mediated by other receptors, methoxychlor and triclosan (ER/cyclin D1/Ras/Bax) and raloxifene
such as serotonin/serotonin 1A receptor (Li et al., 2013c), and crosstalk (ER/TNF-α/ERK/caspase-3) (anti-apoptotic).
26 R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40

3.2.2. Carcinogenesis signaling (Meng et al., 2011) and vitamin D in VDR/ER signaling (Gilad
Estrogen plays significant roles in the development of breast and et al., 2005; Swami et al., 2013), while the signals induced by estrogen
other cancers, and blockade of estrogen action with antagonists reduces affect other signaling pathways through autocrine/paracrine networks
the risk (Villa, 2008; Yue et al., 2013). The estrogenic chemicals (includ- involving the secretion of hormones and growth factors, such as
ing those that modulate estrogenic signaling) that are associated with ER/CXCL12/CXCR4 (Hall and Korach, 2003; Sauvé et al., 2009), ER/
carcinogenesis or cancer functions are as follows: cadmium, di(n-butyl) EGFR (Osborne et al., 2005), ER/ERBB4 (Zhu et al., 2006), ER/IGF-1R
phthalate, parabens and testosterone (positive effects); and genistein, (Kashima et al., 2009), ER/PDGFR (Barbarisi et al., 2001; Finlay
β-naphthoflavone, phenylacetate and tocopherols (negative effects) et al., 2003) and Wnt/β-catenin signaling (Ono et al., 2013). There
(Tables 1 and 2). The signaling pathways involved in estrogen-related are cases of ligand-independent activation of ER, where combina-
carcinogenesis or cancer functions are: cadmium (ER/ERK), di(n-butyl) tions of signal inducers and receptors, such as ethanol/adenosine re-
phthalate (ER/TGF-β), E2 (ER/Akt/IGF-1R/InsR, ER/IGF/IGF-1R, ERα/IRS- ceptor (Etique et al., 2009) and MCP-1/CCRs (Riverso et al., 2014),
1/IGF-1R/PI3K/Akt and ER-α36/MAPK/ERK), methyl-amoorain + DMBA affect ERα signaling in the absence of its ligands for the induction
(ER/Wnt/β-catenin), testosterone (ER-α36/ERK/Akt) and tocopherols of cell proliferation. On the other hand, exemestane, an aromatase
(E2/ER/PPARγ/Akt). Crosstalk of signaling in breast cancer has been re- inhibitor, modulates estrogen synthesis and affects ER and EGFR syn-
ported for the signaling pathways between estrogen and AhR (β-naph- ergistically by autocrine signaling for cell proliferation (Wang et al.,
thoflavone: Wang et al., 2014), IGF-1 (genistein; Hwang et al., 2013), 2008a). Note that silent estrogens, a type of estrogenic chemicals
TGF-β (Band and Laiho, 2011), PR (parabens; Vo et al., 2011) or PPARγ without explicit cell proliferation activity, were observed for some
(di(n-butyl) phthalate; Lee et al., 2009) (Table 3). The cancer-related chemicals including brefeldin A, revealed by DNA microarray assay
signaling induced by estrogen or estrogenic chemicals can be modulated (Dong et al., 2013a; Kiyama and Zhu, 2014). The presence of such a
by other chemicals, such as testosterone (Lin et al., 2009), tocopherols type of estrogenic chemicals suggests that signaling pathways related
(Lee et al., 2009) and TGF-β (Lee et al., 2014). to estrogen action, such as cell growth, can be separated or differentially
manifested not only by SERMs but also by different types of chemicals,
and such a new type of chemicals could be explored to modulate estro-
3.2.3. Cell growth and proliferation gen action at the cell signaling level.
Cell growth and differentiation are key steps for the differentiation
and development of cells, tissues and organs and thus include a number 3.2.4. Differentiation/development
of cellular events, such as cell division, cell adhesion, cell movement, cell Estrogen plays critical roles in the sexual differentiation and devel-
survival and cellular response to a variety of stimuli and stresses. Estro- opment of the brain, breast, prostate and other tissues and organs
gen promotes the growth and proliferation of various types of cell for (Ohtani-Kaneko, 2006; McPherson et al., 2008; Simões and Vivanco,
various cellular events, and thus, cell growth and proliferation have 2011). Estrogenic chemicals associated with cell differentiation and de-
been examined for various estrogenic chemicals and the signaling path- velopment are adiponectin, estradiolquinone, genistein, medicarpin,
ways related to them (Tables 2 and 3). Estrogenic chemicals associated resveratrol and tamoxifen (estrogenic or synergistic), and androstenol
with cell growth and differentiation are summarized below. and retinoic acid (anti-estrogenic or inhibitory), and the associated
signaling pathways are ER/NO/cGMP (resveratrol), ERα/MAPK/NF-κB/
The list of estrogenic chemicals associated with cell growth and differentiation. AP-1 (genistein), ERα/Wnt3A/β-catenin (tamoxifen) and mER/MAPK
Benzo[a]pyrene, bisphenol A, p,p'-DDE, o,p'-DDT, dieldrin, equilin, equol, estrone (retinoic acid) (Tables 2 and 3). Crosstalk was observed for the ER
(E1), estradiol (E2), estriol (E3), 17α-ethinylestradiol, 17β-E2 valerate, ferulic acid, signaling with CAR (androstenol; Min et al., 2002), LTD4 receptor
genistein, β-HCH, hexachlorobenzene, icariin, icaritin, methoxychlor, puerarin,
(estradiolquinone; Dietsch et al., 1996) and APNR (adiponectin; Rahal
procymidone, quercetin and tributyltin (estrogenic); and aloe-emodin, baicalein,
bergapten, calycosin, emodin, formononetin, celastrol, curcumin, daphnetin, and Simmen, 2011).
esculetin, epigallocatechin gallate + curcumin, fulvestrant, hydroxytyrosol,
indole-3-carbinol, luteolin, naringenin, oleuropein, quercetin, secoisolariciresinol 3.2.5. Inflammation
and tamoxifen (anti-estrogenic). Estrogen has anti-inflammatory and vasoprotective effects when
it is administered, and several signaling pathways, such as the TNF-α/
Isoliquiritigenin and piceatannol show dose-dependent estrogenic NF-κB/JNK pathway, have been suggested to mediate its effects (Xing
and anti-estrogenic effects (Maggiolini et al., 2002; Vo et al., 2010). et al., 2009). Estrogenic chemicals affect the inflammatory response
The major signaling pathways identified for estrogenic chemicals are positively (daidzein and genistein) or negatively (baicalein, genistein,
ER/Akt (benzo[a]pyrene, bisphenol A, dieldrin, ferulic acid, luteolin 4-hydroxyphenyl sulfonamides, niacin, p-n-nonylphenol, p-n-
and puerarin), ER/ERK (baicalein, benzo[a]pyrene, p,p'-DDE, o,p'-DDT, octylphenol, oroxylin-A and resveratrol), or modulate the signaling in-
dieldrin, E1, E2, E3, equol, ferulic acid, β-HCH, hydroxytyrosol, methoxy- duced by estrogen (parthenolide, an NF-κB inhibitor) (Tables 1 and
chlor, naringenin, oleuropein, puerarin, quercetin and tributyltin), ER/ 2). The signaling pathways involved are ER/NF-κB (4-hydroxyphenyl
MAPK (17α-ethinylestradiol, 17β-E2 valerate, naringenin, piceatannol, sulfonamides), ER/NF-κB/NO (p-n-nonylphenol and p-n-octylphenol),
procymidone and tributyltin) and GPER/ERK (dieldrin, genistein and ER/NF-ĸB/NO/TNF-α (baicalein), ER/TNF-α/IL-1β/IL-6/NO (oroxylin-
quercetin). There are cases where signals induced by estrogenic A), ERα/TNF-α/NO (daidzein and genistein), GPER/EGFR/ERK (niacin)
chemicals affect other signaling pathways: bisphenol A, calycosin, and GPER/IL-1β/MAPK (genistein). The anti-inflammatory effects of
formononetin, hexachlorobenzene, indole-3-carbinol, luteolin and ta- an SERM, raloxifene, are ER-independent (Lee et al., 2011), suggesting
moxifen for IGF-1 signaling; equilin for NMDA signaling; icariin, icaritin the presence of a new anti-inflammatory target. Resveratrol shows
and secoisolariciresinol for EGF signaling; celastrol and piceatannol for pathway-selective ER signaling, where it activates the inflammatory
progesterone signaling; epigallocatechin gallate + curcumin for VEGF pathway but not the cell proliferation pathway, presumably by alter-
signaling; and fulvestrant for Wnt/β-catenin signaling. Furthermore, ing the way of recruiting the coregulators associated with ERα
estrogenic signaling can be modulated by chemicals in other signaling (Nwachukwu et al., 2014).
pathways by crosstalk, such as bergapten in TGF-βR/ERα (Panno et al.,
2012), farnesol in farnesoid × receptor/ER signaling (Journe et al., 3.3. Signaling networks
2008), melatonin in MT1/ERα signaling (Kiefer et al., 2005), progester-
one and progestin in PR/ER signaling (Migliaccio et al., 1998; Vallejo Estrogen receptors or estrogenic signaling can crosstalk with other
et al., 2005; Ballaré et al., 2006), retinoic acid in RARα/ERα signaling receptors or signaling, either (1) by modulating estrogenic signaling
(Ombra et al., 2013), 3,5,3′-triiodothyronine in integrin receptor/ERα (mostly induced with E2) by signals induced with other chemicals or
R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40 27

(2) by the interaction between different receptors or pathways. The for- Autocrine/paracrine signaling was reported for receptors, such as
mer case may include crosstalk through the genomic pathway, such as ANF receptor, APNR, AR, CCRs, CXCR4, EGFR (ERBB4), FGFR, IGF-1R/
between ERα and AhR (Safe and Wormke, 2003). The latter case may InsR, IL6R, PDGFR and TGF-βR, where the hormones and growth factors
include (2–1) direct interaction between different receptors or path- released are ANF, adiponectin, androgen, MCP-1, CXCL12 (SDF-1), EGF
ways, (2–2) one-way crosstalk, or bypassing, where signaling is uni- (amphiregulin), FGF, IGF-1/insulin, IL-6, PDGF and TGF-β1, respectively.
directional because one of the pathways involved is abortive due to The signaling pathways involved are ERK, JNK, MAPK, NF-κB, PI3K/Akt/
the lack of appropriate effectors or signal mediators, and (2–3) para- mTOR and Wnt/β-catenin signaling, which are related to cell growth
crine and autocrine signaling, where other hormones or growth factors and proliferation in breast and other cancer cells, and differentiation
are involved in the signaling. Note that, in some cases, ligands are not and other functions (Table 3).
significant or required to transduce signaling from one signaling path- In paracrine signaling, hormones and growth factors are secreted
way to another, where activation/inhibition of signaling is done by from a cell type and transported to another cell type. For example,
direct interactions between receptors, or the first signaling results in adiponectin secreted from adipocytes is transported to epithelial cells,
the production or the activation of the second receptor. and binding of adiponectin to its receptor and synergistic activation of
ERβ with genistein facilitate epithelial cell differentiation (Rahal and
Simmen, 2011). MCP-1 secreted from normal epithelial cells binds to
3.3.1. Crosstalk/bypassing
its receptor on breast cancer cells and activates their cell division with
Crosstalk of ER or estrogenic signaling pathways with other recep-
the help of activation of ERα through PI3K/Akt/mTOR signaling
tors or receptor-mediated signaling pathways has been reported for
(Riverso et al., 2014). Estrogen-induced activation of a procoagulant/co-
receptors, such as AhR, EGFR, HER2 and IGF-1R, and signaling pathways,
agulation factor further activates prothrombin to form thrombin, which
such as MAPK/ERK, c-Src, PI3K/Akt, NF-κB and NOTCH (Safe and
is a paracrine factor that acts to develop the uterus (Jazin et al., 1990).
Wormke, 2003; Ribeiro and Freiman, 2014). These pathways mostly
Wnt proteins secreted from mature myometrial cells in response to es-
promote the growth of cancer cells, so understanding them is essential
trogen and progesterone induce myometrial stem cells to develop
for cancer treatment. Furthermore, signals induced by the binding
leiomyomas in the uterus (Ono et al., 2013). Estrogen-induced secretion
of ligands (as agonists), such as biochanin A, formononetin, 3-
of growth factors and hormones from cardiomyocytes to other cells in-
methylcholanthrene, β-naphthoflavone and TCDD, to AhR are mediated
duces neovascularization and attenuates fibrosis (Mahmoodzadeh et al.,
by crosstalk between ER and AhR to contribute to various cell functions
2014). The role of 17α-E2 as an autocrine/paracrine factor was implicat-
through pathways such as PI3K/AKT and MAPK/ERK signaling, or
ed in brain functions and therapeutic use in association with its pre-
through transcription or proteasome-mediated degradation of ERα
ferred receptor ER-X (Toran-Allerand et al., 2005). Finally, since
(Table 3). Biochanin A and formononetin are estrogenic chemicals, but
estrogen itself is an important member of the autocrine/paracrine fac-
could act as AhR agonists and thus be anti-estrogenic through the
tors, the modulation of estrogen signaling is crucial to develop SERMs
crosstalk (Medjakovic and Jungbauer, 2008). AhR signaling is bypassed
for the prevention and treatment of postmenopausal degenerative and
by ER signaling (or crosstalk between ER and AhR is uni-directional) be-
neoplastic diseases (Härkönen and Väänänen, 2006).
cause inhibitory effects were observed only for β-naphthoflavone,
which inhibits ER signaling, but not for 17α-ethinylestradiol, which
3.3.3. Homeostatic networks
inhibits AhR signaling (Gräns et al., 2010).
Extracellular networks adopt a homeostatic nature when they have
Crosstalk has been reported between ER and PR for ER ligands
a system of feedback regulation in autocrine/paracrine signaling.
(genistein and piceatannol) or PR ligands (progesterone and progestin)
While such networks play important physiological and developmental
(Table 3), or between ER and AR in a ligand-independent manner
roles, such as that in estrogen regulation of estrous cyclicity (Yeo and
(Migliaccio et al., 2006). While ERβ and PR form a complex and crosstalk
Herbison, 2014), ovulation (Wintermantel et al., 2006) and spermato-
with each other (Vallejo et al., 2005), crosstalk between ER and growth
genesis (Chimento et al., 2014), too much positive or negative feedback
factors could down-regulate PR expression (Thakkar and Mehta, 2011).
regulation would cause the development and progression of cancer and
Crosstalk between ER and IGF-1R induced with estrogen or
other diseases. For example, positive feedback activation in an autocrine
estrogenic/anti-estrogenic chemicals, such as calycosin, formononetin,
loop between CXCR4/CXCL12 and ERα/ERβ signaling pathways pro-
genistein, ginsenoside Rg1, indole-3-carbinol and resveratrol, involves
motes the ER-dependent development of breast cancer (Sauvé et al.,
pathways, such as IRS-1 and PI3K/Akt signaling, and cell functions,
2009; Boudot et al., 2011).
such as cell proliferation and neuroprotection (Table 3).
The feedback regulation involving estrogen can be categorized by the
Crosstalk between ER and other receptors, such as α1/β-
role of ER: (1) modulation of signaling proteins or transcription factors for
adrenoceptor, CAR, GR, HER2, integrin receptor, LXRβ, natriuretic
growth factor and other signaling-related genes to increase/decrease the
peptide receptor, PPARγ, RAR, ryanodine receptor, TGF-βR, TR and
activity of signal mediators (e.g., Sauvé et al., 2009), (2) transcriptional
VEGFR-1, was also reported along with estrogen or estrogenic chemicals
modulation of ER genes to increase/decrease (mostly increase) the
(lycopene, resveratrol, tamoxifen and tocopherols), or ligands for recep-
amount, and thus the sensitivity, of ERs (e.g., Wintermantel et al., 2006)
tors, such as androstenol/CAR, cholesterol/LXRβ, epigallocatechin
and (3) modulation (mostly increasing) of estrogen synthesis by regulat-
gallate/EGFR, ethanol/adenosine receptor, hesperetin/TrkA receptor,
ing aromatase activity (e.g., Wang et al., 2008a). Examples of feedback
isoproterenol/β-adrenoreceptor, protopanaxadiol and protopanaxatriol/
regulation have been reported for estrogen, although other estrogenic
GR, ryanodine/ryanodine receptor, thyroid hormone/TR and vitamin
chemicals could also be involved, and such regulation of estrogen signal-
D/VDR and Wnt/Fz (Table 3).
ing could be used as targets for developing tumor-targeting drugs (Renoir
et al., 2013).
3.3.2. Autocrine/paracrine signaling
Since cells communicate with other cells in the neighborhood or 4. Evaluation of estrogen action
in distant locations (by paracrine or endocrine signaling) or with them-
selves (by autocrine signaling) through hormones, growth factors, Estrogen action is evaluated first by the structural and functional
cytokines, ions and other mediators, it is important to examine such com- characteristics of chemicals (Tables 1 and 2), whose functional out-
munications to understand the action of estrogen under physiological comes are interpreted partly by the signaling pathways that they
conditions. The signals induced by estrogen or estrogenic chemicals mod- induce (see Section 3). When these collections of information
ulate signaling pathways for other hormones and growth factors, which are used to predict the effect of estrogenic chemicals, we need
are secreted and transported, and finally affect the same cell or other cells. to understand the details of the technologies, especially their
28 R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40

advantages/disadvantages and the limitations of the methods used phenanthrenes, 2-phenylindoles, 2-phenyl-isoindole-1,3-dione, phthalates,
quercetin, triazine, umbelliferone and zeranol (by means of competitive binding of
for the evaluation. Comparison of in vitro assays including their ad-
ERs against 3H–E2); caffeic acid phenethyl ester (by means of recombinant hERα);
vantages and limitations was already discussed (Mueller, 2004; auraptene, 3,9-benz[a]anthracene diols, carboranes, coumestrol, DDT, eugenol, glycinol,
Kiyama and Zhu, 2014). Here, we summarize the methods used juglone, naringenin, naringin, PBDEs, psoralidin, phenanthrenes and tocotrienol
for the evaluation of estrogen action. (by means of molecular modeling); and arctigenin, bisphenol A, hexachlorophene,
liquiritigenin and menadione (by means of fluorescence polarization).

4.1. Assays for detecting estrogen action


4.1.2. Reporter-gene assay
Estrogenic activity can be detected by various methods, such as In a reporter-gene assay, the receptor, upon binding to ligands, binds
ligand-binding assay, reporter-gene assay, yeast two-hybrid assay, tran- to a specific response element on DNA and induces transcription of the
scription assay, protein assay, cell assay, animal tests and signaling downstream reporter gene. There are variations of this assay, such as in
pathway analysis, which are based on the molecular and cellular mech- terms of the reporters: β-galactosidase (lacZ), chloramphenicol acetyl-
anisms of estrogen action (Fig. 1). The assays used and the pathways transferase (CAT), luciferase and green fluorescent protein (GFP); cul-
identified for various estrogenic chemicals are summarized in Tables 1 ture systems; receptor/reporter gene constructs; and the protocols for
and 2. Note that signaling pathway analysis is categorized separately the transfection of reporter constructs into host cells. There are two
from other assays because it usually consists of the assays categorized types of culture system, dependent on the status of the expression of en-
in other methods, and focuses on the identification of signaling path- dogenous ERs. MVLN cells were developed from human breast cancer
ways and functional outcomes rather than the evaluation of MCF-7 cells constitutively expressing ERs by introducing a reporter
estrogenicity alone. Here, we briefly discuss the assays used to detect gene expressing firefly luciferase under the control of the estrogen-
and characterize the estrogenic activity of chemicals. responsive element derived from the 5′-flanking region of the Xenopus
vitellogenin A2 gene (Pons et al., 1990). New cell lines for luciferase-
4.1.1. Ligand-binding assay based reporter-gene assay have been developed to detect steroid hor-
Ligand-binding assay is a well-established method to detect mones effectively: TGRM-Luc cells for glucocorticoids, TM-Luc cells for
receptor–ligand interactions. An extract from hormonally respon- progestagens, TARN-Luc cells for androgens and MMV-Luc cells for es-
sive tissues, such as the uterus and liver (Song et al., 1999; trogens (Willemsen et al., 2004). They were used to assess estrogenic
Tollefsen and Nilsen, 2008), an extract from cultured cells expressing and androgenic endocrine disruptors in sport supplements (Plotan
ERs (Olsen et al., 2003) or a recombinant receptor protein (Goodin et al., 2012). More recently, a panel of mammalian reporter cell-line-
et al., 2002) is used as a source of ERs. The assay quantifies the ability based CALUX (Chemically Activated LUciferase eXpression) bioassays
of a test chemical to compete with 3H-labeled 17β-E2 in binding to was developed to detect chemicals with androgen, estrogen, progester-
ERs, and the result is often expressed as the concentration showing one and glucocorticoids (Sonneveld et al., 2005). The ER-CALUX bioas-
50% inhibition, or IC50. The result is also expressed as a relative bind- say utilizes human osteosarcoma (U2-OS) cells, which have been
ing affinity, the ratio of IC50 between the test and unlabeled E 2. In stably transformed with specific estrogen-response elements linked to
place of radioactive ligands, assays using non-radioactive ligands a luciferase reporter gene, and evaluates estrogenicity by quantifying
have been developed, such as the fluorescence polarization method luciferase protein.
(Jung et al., 2004; Hashimoto et al., 2001; Mersereau et al., 2008) When cells do not express endogenous receptors, it is necessary to
and the competitive enzyme immunoassay (Nishizuka et al., 2004). transfect two different plasmids constitutively expressing either a re-
While such assays are easy and fast to perform, they cannot distin- ceptor or a reporter gene. Yeast cells are frequently used in this type
guish between agonists and antagonists. Recently, McLachlan et al. of assay because less time is required to detect the response with signif-
developed a biosensor that can distinguish agonists and antagonists, icant reproducibility. In the yeast estrogen screen (YES) assay, yeast
where two halves of a split Venus fluorescent protein fused to either cells (Saccharomyces cerevisiae) were stably transfected with DNA
end of the ERα ligand-binding domain produce a fluorescent signal of two plasmid constructs containing the human ER gene and the
after conformational changes induced by binding estrogenic estrogen-response element-linked lacZ, and β-galactosidase activity
chemicals to the ligand-binding domain (McLachlan et al., 2011). was detected colorimetrically using chromogenic substrates, such as
The quantitative structure–activity relationship (QSAR) method has o-nitrophenyl-β-D-galactoside (ONPG) (Arnold et al., 1996). However,
been developed on the basis of the molecular dynamic structure of the the colorimetric assays take several days to perform and require cell
compound and used to predict estrogenic activity and to explore the lysis. These problems were solved by using fluorescence-based re-
interaction between ERs and ligands (Li and Gramatica, 2010). To obtain porters, such as firefly luciferase (Leskinen et al., 2005) or GFP (Bovee
more detailed information about ER–ligand interactions at the atomic et al., 2004). However, in yeast-based assays, the discrimination be-
level, computational molecular modeling is a powerful tool. Li et al. tween agonists and antagonists is not effective, and the differences in
carried out a molecular modeling study combining molecular docking, membrane permeability, transport proteins and signal transduction
molecular dynamics stimulation and binding free energy calculation to pathways may cause differences in the response to chemicals between
examine the interaction between hydroxylated polychlorinated yeast cells and humans. Reporter-gene assay was used to characterize
biphenyls (HO-PCBs) and ERs, and found that several amino acid the estrogenicity of the chemicals (Tables 1 and 2), which are summa-
residues formed hydrophobic and hydrogen–bond interactions with rized below.
the ligand (Li et al., 2013d). Ligand-binding assay was used to character-
ize the estrogenicity of the chemicals (Tables 1 and 2), which are
The list of estrogenic chemicals characterized by reporter-gene assay.
summarized below. Acetaminophen, alkylanilines, caffeic acid phenethyl ester, chlordecone, epigallo-
catechin 3-gallate, fisetin, galangin, isocoumarins, luteolin, menadione, naringenin,
naringin, parabens, pentachlorophenol and 3-phenoxybenzaldehyde (by means of
β-galactosidase-based reporters); acteoside, anthraquinone, apigenin, auraptene,
The list of estrogenic chemicals characterized by ligand-binding assay. benz[a]acridine, benzanthrone, benzo[a]pyrene, biochanin A, butylated
Acetaminophen, acetylsalicylic acid, aldicarb, 3-alkyl naphthalenes, alkylphenols, hydroxyanisole, cajanin, carboranes, β-carotene, daidzein, dieldrin, ephedrine,
anethole, benzo[a]pyrene, bisphenol A, boric acid, chlordecone, citral, epigallocatechin 3-gallate, formononetin, genistein, ginsenoside Rg1, glycinol,
dichlorophenyls, 9,10-dimethyl-m-carborane, diphenylamines, enterodiol, glyceollins, glyphosate, β-HCH, hexachlorophene, hydroxychalcones, insecticides,
enterolactone, epigallocatechin 3-gallate, equilenin, geraniol, glabridin, glycitein, γ-linolenic acid, liquiritigenin, luteolin, kaempferol, malathion, martynoside,
β-HCH, indole-3-carbinol, isocoumarins, isorhamnetin, kaempferide, kaempferol, metalloestrogens, naringenin, naringin, PBDEs, PCBs, perfluorinated compounds,
metalloestrogens, 3-(4-methylbenzylidene) camphor, myricetin, nerol, PBDEs, phenanthrenes, 2-phenylindoles, phthalates, propyl gallate, psoralidin,
R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40 29

rhodoeosein, sesamin/sesamolin/sesamol, tartrazine, tricresyl phosphate and design could be combined by the Generally Applicable Gene-set Enrich-
zearalenone (by means of luciferase-based reporters); indole-3-carbinol and
ment (GAGE) method and used to predict estrogen signaling pathways
metalloestrogens (by means of CAT-based reporter genes); and bifenthrin,
chloroxylenol, insecticides and taxifolin (by means of ER-CALUX). (Luo et al., 2009). Recently, DNA microarrays have been developed for
non-mammalian genes and used for evaluation of estrogenic chemicals
(Baker et al., 2013; Hao et al., 2013). Transcription assay was used to
4.1.3. Yeast two-hybrid assay characterize the estrogenicity of the chemicals (Tables 1 and 2), which
Yeast two-hybrid assay was originally designed to detect protein– are summarized below.
protein interaction in vivo and to identify the genes encoding the
interacting proteins using transcriptional activators such as yeast The list of estrogenic chemicals characterized by transcription assay.
GAL4 protein. To evaluate estrogenic activity, two plasmids are used: a Apigenin, arsenic, benzo[a]pyrene, biochanin A, curcumin, cyanidin-3-glycoside,
plasmid carrying a gene encoding a fusion protein containing the daphnetin, DDT, dieldrin, epigallocatechin 3-gallate, esculetin, fenarimol,
GAL4 DNA-binding domain (GAL4DBD) and the human ERα/β ligand- fluorotelomer alcohols, flutolanil, ginsenoside Rg1, glycinol, hydroxymatairesinol,
insecticides, isorhamnetin, kaempferol, liquiritigenin, lycopene, myricetin,
binding domain (hERα/β LBD) and another plasmid containing a
naringenin, naringin, irisolidone, parabens, perfluorobutanesulfonic acid,
cDNA fragment encoding the ER-binding domain of a co-regulator perfluorooctyl iodide, psoralidin, quercetin, sesamin/sesamolin/sesamol, silibinin,
with a region encoding GAL4 transactivation domain (GAL4AD) fused tartrazine, TCDD, tectorigenin, tocotrienol, zearalenone and zeranol (by RT-PCR);
to the lacZ gene. Yeast cells are transformed with DNA of both plasmids, curcumin, genistein, juglone, phloretin and phthalates (by means of DNA microarray
and the transformants are selected by growth on a medium lacking leu- assay); and β-carotene, indole-3-carbinol, insecticides, lycopene and SERMs
(by Northern blotting).
cine and tryptophan. Test chemicals are added to the yeast cells to eval-
uate β-galactosidase activity. Since hERα/β LBD and the co-regulator
interact with each other only in the presence of a ligand, the expression 4.1.5. Protein assay
of the lacZ gene is dependent on the ligand binding. The assay revealed Various analytical matrices for qualitative and quantitative screen-
that the ER also binds to nuclear receptor co-regulators, such as RIP140, ing of endocrine disruptors have been developed from large-scale phys-
SRC-1, TIF1 and TIF2, in response to chemicals according to their estro- icochemical methods, such as mass spectrometry (MS), to bench-scale
genic activity, and demonstrated that the interaction between steroid approaches, such as Western blotting and immunohistochemistry. In
hormone receptors and co-regulators can be a useful tool for identifying order to detect protein quantitatively, radio-, enzyme- and fluoro-
chemicals that interact with the receptors (Nishikawa et al., 1999). The based immunoassays (IAs), ELISA, liquid chromatography (LC), gas
entire hERα in combination with the nuclear receptor-binding domain chromatography (GC), MS and combined MS with immunoassay
of co-regulators, SRC-1 or TIF2, resulted in a higher response to estrogen (MSIA) are available. It should be noted that a single or several protein
than GAL4DBD-hERαLBD (Lee et al., 2006). This suggests that the syner- markers are often used in these assays, whereas sets of protein markers,
gistic action of AF-1 and AF-2 of hERα mediates the recruitment of such as those used for expressional profiling in transcription assays, are
co-regulators, such as SRC-1, and also that a ligand-dependent direct in- rarely used, mainly due to the difficulty in preparing tens or hundreds of
teraction between the B domain in AF-1 and C-terminal domain can sta- antibodies or peptides at low cost and in predicting estrogen action by
bilize the cooperative interaction between ERα and co-regulators proteomic profiling. Thus, ER proteins and markers, such as APOA1
(Métivier et al., 2001). The yeast two-hybrid assay was compared with (ApoA-1), CCDN1 (cyclin D1), PGR (PR), TFF1 (pS2) and vitellogenin,
a commercial enzyme-linked immunosorbent assay (ELISA) kit to mea- are often detected alone by protein assay. Protein assay was used to
sure estrogenic activity or total estrogen concentrations, respectively, in characterize the estrogenicity of the chemicals (Tables 1 and 2), which
natural and waste waters (Allinson et al., 2011). While the two assays are summarized below.
showed a very good correlation and the ELISA kit is a reasonably rapid
tool for preliminary screening of water samples, the yeast two-hybrid
The list of estrogenic chemicals characterized by protein assay.
assay may not be accurate enough to represent mammalian or human
Acetaminophen, biochanin A, caffeic acid phenethyl ester, β-carotene, coumestrol,
systems, and ELISA may not be able to detect unknown compounds. curcumin, daphnetin, epigallocatechin 3-gallate, esculetin, flutolanil, genistein,
Yeast two-hybrid assay was used to characterize the estrogenicity of glyphosate, insecticides, irisolidone, metalloestrogens, myricetin, naringin, parabens,
acetaminophen, bisphenols, n-butyl benzyl phthalate, β-cyclodextrin, PCB3, tartrazine, tectorigenin, tocotrienol and zearalenone (by Western blotting);
bisphenol A, β-cyclodextrin, genistein, insecticides, nothofagin, oxybenzone, SERMs,
fenthion sulfoxide, hexachlorophene, 2-hydroxyanthraquinone, mena-
silibinin and triclopyr (by immunoassay); myricetin (by ELISA); and epigallocatechin
dione, naringenin, naringin, PCBs and phlorizin (Tables 1 and 2). 3-gallate, ginsenoside Rg1 and psoralidin (by ChIP assay).

4.1.4. Transcription assay


Analysis of the transcripts of genes, or transcriptomic analysis, is one 4.1.6. Cell assay
of the most effective approaches to predict the toxicity of chemicals, and Cell growth and proliferation can be detected by a variety of
various transcription assays, such as DNA microarray assay, Northern methods: visualization of cell sizes and shapes by microscopy after ap-
blotting, RNA-seq, RT-PCR and the serial analysis of gene expression propriate staining, counting live cell numbers by the dye exclusion
(SAGE), have been developed (Kiyama and Zhu, 2014). These assays method using trypan blue or by cytometry, such as fluorescence-
quantify the expression of ER genes or specific marker genes, such as activated cell sorting (FACS), distinguishing dead/viable cells, cell
APOA1 (ApoA-1), BRCA2, CCDN1 (cyclin D1), PGR and TFF1 (pS2) (by types, cell differentiation and biomarkers, and detecting metabolic
Northern blotting and RT-PCR), or sets of genes responding to estrogen or redox activity by colorimetric (MTT) or fluorimetric (resazurin)
or estrogen-responsive genes (by DNA microarray assay, RNA-seq and methods. The formazan-based MTT assay is a colorimetric assay for
SAGE). DNA microarray-based evaluation of endocrine disruptors has assessing cell viability. MTT, a yellow tetrazole derivative, is reduced to
been discussed previously (Tanji and Kiyama, 2004; Kiyama and Zhu, purple formazan by NAD(P)H-dependent oxidoreductase. In contrast,
2014). One of the advantages for DNA microarray assay over other tran- resazurin-based assays, such as alamarBlue assay (Desaulniers et al.,
scription assays is that it can give gene-expression profiles of the 1998), exhibit greater sensitivity than cell counting, microfluorometric
chemicals examined, which could be used to compare the effects of DNA determination assay and even MTT assay. Other colorimetric
chemicals by comparing the genes related to various cell functions. chemicals, such as MTS (a tetrazolium) and sulforhodamine B (SRB),
Comparative evaluation of DNA microarray, RT-PCR and real-time have been used in cell assays.
RT-PCR showed DNA microarray assay has the advantage over the Cell lines derived from breast cancer have been used in cell assays.
others in effectiveness and quickness in diagnosis (Sultankulova et al., Estrogenic breast cancer cell lines, such as MCF-7, T-47D and MDA-
2014). Gene expression datasets of different size and experimental MB-231 cells, are commonly used to examine the effects of chemicals
30 R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40

on cell functions, such as cell viability, cell proliferation, cell cycle, apo- animals. Animal tests were used to characterize the estrogenicity of
ptosis and cell migration. For the determination of estrogenic activity, the chemicals (Tables 1 and 2), which are summarized below.
cell proliferation assays are widely used. The E-screen assay developed
by Soto et al. (1995) is based on the growth promotion of MCF-7 cells The list of estrogenic chemicals characterized by animal tests.
in the presence of estrogen, and is widely used as a screening tool for es- Acetaminophen, benzo[a]pyrene, boric acid, caffeic acid phenethyl ester,
trogenic chemicals (Grünfeld and Bonefeld-Jorgensen, 2004; Maras daphnetin, 2,4-dihydroxybenzophenone, esculetin, ephedrine, epigallocatechin
3-gallate, estrone 3-carboranylmethyl ether, fenarimol, fluoxetine, ginsenoside Rg1,
et al., 2006; Resende et al., 2013). Cell assay was used to characterize
glabridin, glycitein, insecticides, naringenin, naringin, 4-nitrophenol, PCBs,
the estrogenicity of the chemicals (Tables 1 and 2), which are summa- 2-phenylindoles, phthalates and zeranol (by rodent uterotrophic assay); pinosylvin
rized below. and β-sitosterol (by rainbow trout feeding assay); 3-(4-methylbenzylidene) camphor
and perfluorobutanesulfonic acid (by using Xenopus); and DDT, oxybenzone,
perfluorooctyl iodide and phthalates (by using medaka).
The list of estrogenic chemicals characterized by cell assay.
Alachlor, anethole, arsenic, boric acid, citral, eugenol, geraniol, glyphosate,
licochalcone A, metalloestrogens, nerol, nitrophenols, PCB118, 4-phenylphenol,
phthalates and soysaponin I (by YES assay); 4-aryl-coumarin dimer, biochanin A, 4.1.8. Signaling pathway analysis
2,6-dihydroxyanthraquinone, epigallocatechin 3-gallate, formononetin, Signaling pathway analysis consists of a variety of methods and
glyceollins, 6-hydroxymusizin, insecticides, juglone, naringenin, naringin, protocols, which have variations depending on the signaling pathways,
pharmaceuticals, phlorizin, pinosylvin, propelargonidins, silibinin and β-sitosterol signaling networks and functional outcomes. Various types of cell
(by MTT assay); apigenin, epigallocatechin 3-gallate and naringin (by MTS assay);
signaling induced by estrogenic chemicals are summarized in Tables 1
benzyl salicylate, conazoles, cyanidin-3-glycoside, dieldrin and SERMs (by SRB
assay); cajanin (by alamarBlue assay); arsenic (by trypan blue assay); coumestrol to 3, and the signaling pathways related to apoptosis, carcinogenesis,
and dieldrin (by using CellTiter); β-carotene, glyphosate, indole-3-carbinol, cell growth/differentiation, differentiation/development and inflamma-
lycopene, metalloestrogens and phthalates (by using cell counters); bezafibrate, tion are discussed in Section 3. Signaling pathway analysis was used to
bisphenol A, daphnetin, esculetin, fenofibrate, fisetin, fluorotelomer alcohols,
characterize the estrogenicity of the chemicals (Tables 1 and 2), which
flutolanil, galangin, gemfibrozil, insecticides, irisolidone, isorhamnetin luteolin,
kaempferol, metalloestrogens, PCBs, quercetin, semicarbazide and tectorigenin
are summarized below.
(by E-screen assay); and β-carotene, epigallocatechin 3-gallate,
hydroxymatairesinol, juglone and lycopene (by flow cytometry). The list of estrogenic chemicals characterized by signaling pathway analysis.
Aroclor 1254, epigallocatechin 3-gallate, formononetin, geniposide, ginsenoside
Rb1, methoxychlor/triclosan, naringenin, plumbagin, quercetin, raloxifene,
4.1.7. Animal test withaferin A and wogonin (for apoptosis); cadmium, di(n-butyl) phthalate,
Animal tests have been used for basic research, such as genetics, methyl-amoorain + DMBA, testosterone and tocopherols (for carcinogenesis);
baicalein, benzo[a]pyrene, bisphenol A, p,p′-DDE, o,p′-DDT, equol,
developmental biology and behavioral studies, as well as applied
17α-ethinylestradiol, 17β-E2 valerate, dieldrin, ferulic acid, genistein, β-HCH,
research, such as biomedical research, xenotransplantation, drug testing hydroxytyrosol, luteolin, methoxychlor, naringenin, oleuropein, piceatannol,
and toxicology tests. Some of the abnormality in animals used for procymidone, puerarin, quercetin and tributyltin (for cell growth and proliferation);
aminal tests, such as ovotestis, was found originally by the observation adiponectin, androstenol, estradiolquinone, genistein, resveratrol, retinoic acid and
of wild animals, such as alligators and fish. In vivo biomarkers, such as tamoxifen (for differentiation/development); and baicalein, daidzein genistein,
4-hydroxyphenyl sulfonamides, niacin, p-n-nonylphenol, p-n-octylphenol and
vitellogenin and aromatase, have been used as physiological indicators
oroxylin-A (for inflammation).
to estimate efficiently the impact of estrogenic chemicals, although
careful interpretateion is needed (Nadzialek et al., 2011). Meanwhile,
animal tests for toxicology testing have been conducted by pharmaceu- 4.2. Pathway-based risk assessment
tical companies and animal testing facilities, and one million animals are
used every year in Europe in toxicology tests (Commission of the Replacing animal tests with in vitro assays has been accelerated in
European Communities, 2007), to examine industrial products, such basic science as well as in the industrial technology field. In 2007, the
as those listed in Table 2. Test results indicate acute, subchronic or US National Research Council (NRC) released a report “Toxicity Testing
chronic effects or the damage to specific organs, such as eye and skin in the 21st Century: A Vision and a Strategy” (National Research Council,
irritancy, mutagenicity, carcinogenicity, teratogenicity and reproductive 2007), in which they called for a transformation of toxicity testing from
problems. The results are expressed using variables such as LD50, the animal tests to a system based on in vitro assays where perturbations in
concentration of chemicals at which 50% of the test animals are killed. key toxicity pathways in response to chemicals can be evaluated.
Animal tests have several problems: they cost several million dollars Current and future toxicity testing can be categorized into four options:
per substance, take three or four years to complete, can potentially over- status quo animal testing as Option I, animal testing partly assisted by in
estimate the risks, especially giving false-positive results, and show var- silico and in vitro screens as Option II, minimum in vivo testing largely
iability in the results between the effect of high doses of chemicals in assisted by in vitro screens as Option III and complete in vitro (in silico)
animals and the effects of low doses in large numbers of humans, as testing without animal tests as Option IV. While this report had a pro-
well as in the rationale of how to use data on one species to predict found impact for environmental scientists, toxicologists and pharmacol-
the risk in another. ogists as well as scientists and researchers in other fields, no practical
Animal tests are useful to screen endocrine disruptors to evaluate protocol has ever been established. Although a number of toxicity path-
adverse endocrine-mediated reproductive and/or developmental ways have already been identified, most are still partial and do not have
effects in multi-generation reproduction studies. In general, the use of sufficient annotations for their use for risk assessment, such as that in
living organisms allows the variation of assays based on a wide range human exposure to chemicals. Although prototype protocols for risk
of species with different end-points. The rodent uterotrophic bioassay assessment were examined for quercetin in DNA-damage-related sig-
is the most widely used in vivo screening assay, based on the response naling (Adeleye et al., 2015) and benzo[a]pyrene and dioxin in AhR sig-
of the estrogen-sensitive uterus. Animals are injected with the test naling (National Research Council, US, 2010), other pathways for these
substances and uterotrophic activities are estimated by the uterine chemicals (such as those shown in Tables 1 and 2) are not included, indi-
weight (Zacharewski et al., 1998; Tamir et al., 2000; Dang et al., 2007; cating the difficulty of such an approach.
Jiménez-Orozco et al., 2011). Similarly, Hershberger assay has been What is important to use pathway-based risk assessment is not how
used to identify potential (anti-)androgenic compounds (Yamada well such a strategy can mimic animal testing, but how to shift from
et al., 2000) and frog embryo teratogenesis assay (FETAX) to detect outcome-based assessment, where only the result of assays or the fate
developmental toxicants in the environment (Fort et al., 2001). These of animals is evaluated, to mechanism-based assessment, where the
animal tests are time-consuming and most require the use of live process occurring in cells is evaluated before outcomes have emerged.
R. Kiyama, Y. Wada-Kiyama / Environment International 83 (2015) 11–40 31

This paradigm shift requires reliable assays, which are based on the pre- References
dictability of the risk and are as valuable as the fate of animals. However,
as mentioned above, the variability of signaling pathways is so limited at Abdelrahim, M., Ariazi, E., Kim, K., Khan, S., Barhoumi, R., Burghardt, R., Liu, S., Hill, D.,
Finnell, R., Wlodarczyk, B., Jordan, V.C., Safe, S., 2006. 3-Methylcholanthrene and
this moment that the variability of chemicals is restricted to limited other aryl hydrocarbon receptor agonists directly activate estrogen receptor
numbers of categories, so the characteristics of each chemical cannot be alpha. Cancer Res. 66 (4), 2459–2467.
properly evaluated. Interesting and suggestive approaches, however, can Adeleye, Y., Andersen, M., Clewell, R., Davies, M., Dent, M., Edwards, S., Fowler, P.,
Malcomber, S., Nicol, B., Scott, A., Scott, S., Sun, B., Westmoreland, C., White, A.,
be found in cancer genomics, where pathway-based pharmacogenomics Zhang, Q., Carmichael, P.L., 2015. Implementing Toxicity Testing in the 21st Century
has been a key strategy for developing personalized cancer medicine (TT21C): making safety decisions using toxicity pathways, and progress in a proto-
(Chin et al., 2011), and in the US National Cancer Institute (NCI) 60 type risk assessment. Toxicology 332, 102–111.
Ahmad, A., Banerjee, S., Wang, Z., Kong, D., Sarkar, F.H., 2008. Plumbagin-induced apopto-
human tumor cell line anticancer drug screen (NCI60), where more sis of human breast cancer cells is mediated by inactivation of NF-kappaB and Bcl-2.
than 100,000 chemicals were examined for screening candidate drugs J. Cell. Biochem. 105 (6), 1461–1471.
using 60 human tumor cell lines (Shoemaker, 2006). The characteristics Ahmed, S., Valen, E., Sandelin, A., Matthews, J., 2009. Dioxin increases the interaction
between aryl hydrocarbon receptor and estrogen receptor alpha at human promoters.
of each chemical could be evaluated to give sufficient predictability, Toxicol. Sci. 111 (2), 254–266.
which is also crucial for personalized medicine. Akama, K.T., Thompson, L.I., Milner, T.A., McEwen, B.S., 2013. Post-synaptic density-
95 (PSD-95) binding capacity of G-protein-coupled receptor 30 (GPR30), an es-
trogen receptor that can be identified in hippocampal dendritic spines. J. Biol.
Chem. 288 (9), 6438–6450.
5. Conclusion and future prospects Alarid, E.T., Preisler-Mashek, M.T., Solodin, N.M., 2003. Thyroid hormone is an inhibitor
of estrogen-induced degradation of estrogen receptor-alpha protein: estrogen-
Here, we summarized estrogenic chemicals, which are categorized by dependent proteolysis is not essential for receptor transactivation function in the
pituitary. Endocrinology 144 (8), 3469–3476.
structure (Table 1) or by characteristics other than structure (Table 2) Ali, Z., Khan, S.I., Khan, I.A., 2009. Soyasaponin Bh, a triterpene saponin containing a
(Section 2). Estrogenic activity is understood by the mechanism-based unique hemiacetal-functional five-membered ring from Glycine max (soybeans).
characterization of estrogenic chemicals as summarized in Fig. 1, so we Planta Med. 75 (4), 371–374.
Ali, I., Damdimopoulou, P., Stenius, U., Adamsson, A., Mäkelä, S.I., Åkesson, A., Berglund, M.,
characterized the signaling pathways in which each estrogenic chemical Håkansson, H., Halldin, K., 2012. Cadmium-induced effects on cellular signaling path-
is involved and the methods by which each estrogenic chemical is ways in the liver of transgenic estrogen reporter mice. Toxicol. Sci. 127 (1), 66–75.
assayed (Section 3). Signaling pathways are further characterized by Alimirah, F., Peng, X., Yuan, L., Mehta, R.R., von Knethen, A., Choubey, D., Mehta, R.G.,
2012. Crosstalk between the peroxisome proliferator-activated receptor γ (PPARγ)
the receptors and the networks involving other receptors and signaling and the vitamin D receptor (VDR) in human breast cancer cells: PPARγ binds to
pathways as well as the pathways involving autocrine/paracrine signal- VDR and inhibits 1α,25-dihydroxyvitamin D3 mediated transactivation. Exp. Cell
ing and homeostatic networks (Table 3). We found here that a number Res. 318 (19), 2490–2497.
Allinson, M., Shiraishi, F., Allinson, G., 2011. A comparison of recombinant receptor-
of chemicals with quite diversified structures, phenolics and non-
reporter gene bioassays and a total estrogen enzyme linked immunosorbent assay
phenolics of natural or industrial origin, show estrogenicity, and a variety for the rapid screening of estrogenic activity in natural and waste waters. Bull.
of signaling pathways and networks, owing not only to the differential Environ. Contam. Toxicol. 86 (5), 461–464.
Andersen, H.R., Vinggaard, A.M., Rasmussen, T.H., Gjermandsen, I.M., Bonefeld-Jørgensen,
roles of ERs but also to the complexity of pathways, effectors and cells/
E.C., 2002. Effects of currently used pesticides in assays for estrogenicity,
tissues, are associated with estrogenicity. While this kind of diversity androgenicity, and aromatase activity in vitro. Toxicol. Appl. Pharmacol. 179 (1),
and complexity would be an obstacle to understand clearly the effect of 1–12.
estrogenic chemicals and to establish the methodology for risk assess- Andersen, H.R., Bonefeld-Jørgensen, E.C., Nielsen, F., Jarfeldt, K., Jayatissa, M.N., Vinggaard,
A.M., 2006. Estrogenic effects in vitro and in vivo of the fungicide fenarimol. Toxicol.
ment, pathway-based risk assessment would give superiority as a risk as- Lett. 163 (2), 142–152.
sessment technology, once established, because sensitivity would be Anita, Y., Radifar, M., Kardono, L.B., Hanafi, M., Istyastono, E.P., 2012. Structure-based de-
higher due to multiple monitoring points in signaling pathways, sign of eugenol analogs as potential estrogen receptor antagonists. Bioinformation 8
(19), 901–906.
which are then integrated into a single or a few outcomes, such as Anstead, G.M., Kym, P.R., 1995. Benz[a]anthracene diols: predicted carcinogenicity and
those detected by animal assay, cell-proliferation assay, ligand- structure–estrogen receptor binding affinity relationships. Steroids 60 (5), 383–394.
binding assay, protein assay, reporter-gene assay and signaling path- Arbo, M.D., Franco, M.T., Larentis, E.R., Garcia, S.C., Sebben, V.C., Leal, M.B., Dallegrave, E.,
Limberger, R.P., 2009. Screening for in vivo (anti)estrogenic activity of ephedrine
way analysis (see Section 4). Furthermore, complex systems such as and p-synephrine and their natural sources Ephedra sinica Stapf. (Ephedraceae) and
in animal tests are not required, providing less expensive, quick, high- Citrus aurantium L. (Rutaceae) in rats. Arch. Toxicol. 83 (1), 95–99.
throughput and highly sensitive assays. Ariazi, E.A., Jordan, V.C., 2006. Estrogen-related receptors as emerging targets in cancer
and metabolic disorders. Curr. Top. Med. Chem. 6 (3), 203–215.
However, we still need to understand more about signaling path- Armstrong, A.F., Valliant, J.F., 2007. The bioinorganic and medicinal chemistry of
ways to utilize the information obtained by the analysis for risk assess- carboranes: from new drug discovery to molecular imaging and therapy. Dalton
ment and to refine the methodology based on the pathways, which Trans. 38, 4240–4251.
Arnold, S.F., Robinson, M.K., Notides, A.C., Guillette Jr., L.J., McLachlan, J.A., 1996. A yeast es-
need to be reasonably developed in order to replace and reduce animal
trogen screen for examining the relative exposure of cells to natural and xenoestrogens.
tests. First, the current information about genes is quite large, although Environ. Health Perspect. 104 (5), 544–548.
it is mostly descriptive and explanatory, and lacks predictability. Mean- Arulmozhiraja, S., Shiraishi, F., Okumura, T., Iida, M., Takigami, H., Edmonds, J.S., Morita, M.,
while, pathway-based risk assessment could be used as a partial 2005. Structural requirements for the interaction of 91 hydroxylated polychlorinated
biphenyls with estrogen and thyroid hormone receptors. Toxicol. Sci. 84 (1), 49–62.
replacement of or as a preliminary screening step for animal tests, Ashok, B.T., Chen, Y., Liu, X., Bradlow, H.L., Mittelman, A., Tiwari, R.K., 2001. Abrogation
such as uterotrophic assay and life-cycle assay, to detect alterations at of estrogen-mediated cellular and biochemical effects by indole-3-carbinol. Nutr.
early stages before using the uterus and other tissues/organs. The com- Cancer 41 (1–2), 180–187.
Axon, A., May, F.E., Gaughan, L.E., Williams, F.M., Blain, P.G., Wright, M.C., 2012.
binations of chemicals, biological outcomes and assay systems would Tartrazine and sunset yellow are xenoestrogens in a new screening assay to iden-
provide quite a large number of variations to distinguish predicted tify modulators of human oestrogen receptor transcriptional activity. Toxicology
outcomes of potential risks. 298 (1–3), 40–51.
Babiker, F.A., De Windt, L.J., van Eickels, M., Thijssen, V., Bronsaer, R.J., Grohé, C., van
Bilsen, M., Doevendans, P.A., 2004. 17beta-estradiol antagonizes cardiomyocyte hy-
pertrophy by autocrine/paracrine stimulation of a guanylyl cyclase A receptor-cyclic
Acknowledgments guanosine monophosphate-dependent protein kinase pathway. Circulation 109 (2),
269–276.
Bachmeier, B.E., Mirisola, V., Romeo, F., Generoso, L., Esposito, A., Dell'eva, R., Blengio, F.,
This research was supported partly by a Knowledge Cluster Initiative Killian, P.H., Albini, A., Pfeffer, U., 2010. Reference profile correlation reveals
program and a Grant-in-aid for Basic Areas from the Ministry of Educa- estrogen-like trancriptional activity of Curcumin. Cell. Physiol. Biochem. 26 (3),
471–482.
tion, Culture, Sports, Science and Technology of Japan, and by the Guide-
Baker, M.E., 2013. What are the physiological estrogens? Steroids 78 (3), 337–340.
line Program for Medical Device Development from the Ministry of Baker, M.E., Vidal-Dorsch, D.E., Ribecco, C., Sprague, L.J., Angert, M., Lekmine, N., Ludka, C.,
Economy, Trade and Industry of Japan. Martella, A., Ricciardelli, E., Bay, S.M., Gully, J.R., Kelley, K.M., Schlenk, D., Carnevali, O.,

You might also like