ArticlePDF AvailableLiterature Review

A structural view of nuclear hormone receptor: Endocrine disruptor interactions

Authors:

Abstract and Figures

Endocrine-disrupting chemicals (EDCs) represent a broad class of exogenous substances that cause adverse effects in the endocrine system by interfering with hormone biosynthesis, metabolism, or action. The molecular mechanisms of EDCs involve different pathways including interactions with nuclear hormone receptors (NHRs) which are primary targets of a large variety of environmental contaminants. Here, based on the crystal structures currently available in the Protein Data Bank, we review recent studies showing the many ways in which EDCs interact with NHRs and impact their signaling pathways. Like the estrogenic chemical diethylstilbestrol, some EDCs mimic the natural hormones through conserved protein-ligand contacts, while others, such as organotins, employ radically different binding mechanisms. Such structure-based knowledge, in addition to providing a better understanding of EDC activities, can be used to predict the endocrine-disrupting potential of environmental pollutants and may have applications in drug discovery.
Content may be subject to copyright.
REVIEW
A structural view of nuclear hormone receptor: endocrine
disruptor interactions
Albane le Maire William Bourguet
Patrick Balaguer
Received: 14 October 2009 / Revised: 3 December 2009 / Accepted: 22 December 2009
ÓBirkha
¨user Verlag, Basel/Switzerland 2010
Abstract Endocrine-disrupting chemicals (EDCs) repre-
sent a broad class of exogenous substances that cause
adverse effects in the endocrine system by interfering with
hormone biosynthesis, metabolism, or action. The molec-
ular mechanisms of EDCs involve different pathways
including interactions with nuclear hormone receptors
(NHRs) which are primary targets of a large variety of
environmental contaminants. Here, based on the crystal
structures currently available in the Protein Data Bank, we
review recent studies showing the many ways in which
EDCs interact with NHRs and impact their signaling
pathways. Like the estrogenic chemical diethylstilbestrol,
some EDCs mimic the natural hormones through conserved
protein–ligand contacts, while others, such as organotins,
employ radically different binding mechanisms. Such
structure-based knowledge, in addition to providing a
better understanding of EDC activities, can be used to
predict the endocrine-disrupting potential of environmental
pollutants and may have applications in drug discovery.
Keywords Nuclear hormone receptors
Endocrine-disrupting chemicals Environmental
pollutants Crystal structures Hormone
Introduction
Endocrine-disrupting chemicals (EDCs) are exogenous
substances that interfere with the function of hormonal
systems and produce a range of developmental, reproduc-
tive, neurological, immune, or metabolic diseases in
humans and wildlife [1,2]. Many EDCs are man-made
chemicals produced by industry and released into the
environment, for example phthalate or bisphenol A (BPA)
plasticizers, organotins, pesticides, dioxins, polychlorinated
biphenyls, flame retardants, or alkylphenols. Some naturally
occurring EDCs can also be found in plants or fungi, such as
the so-called phytoestrogens: genistein, daidzein, or the
mycoestrogen zearalenone. The sources of exposure to
EDCs are diverse and vary widely around the world. There
are several historical examples of toxic pills or contami-
nation that show a direct causal relationship between a
unique chemical and the manifestation of an endocrine or
reproductive dysfunction (see below). However, these types
of single exposure are not representative of more common
persistent exposures to a broad mix of chemicals and con-
taminants. Industrialized and agricultural areas are typically
characterized by contamination from a wide range of
chemicals that may seep into soil and groundwater. These
complex mixtures enter the food chain and accumulate in
animals and humans. Exposure occurs through drinking
A. le Maire W. Bourguet (&)
INSERM, U554, Centre de Biochimie Structurale,
34090 Montpellier, France
e-mail: bourguet@cbs.cnrs.fr
A. le Maire W. Bourguet
CNRS, UMR5048, Universite
´s Montpellier 1 & 2,
34090 Montpellier, France
P. Balaguer (&)
Institut de Recherche en Cance
´rologie de Montpellier (IRCM),
34298 Montpellier, France
e-mail: patrick.balaguer@valdorel.fnclcc.fr
P. Balaguer
INSERM, U896, 34298 Montpellier, France
P. Balaguer
Universite
´Montpellier 1, 34298 Montpellier, France
P. Balaguer
CRLC Val d’Aurelle Paul Lamarque, 34298 Montpellier, France
Cell. Mol. Life Sci.
DOI 10.1007/s00018-009-0249-2 Cellular and Molecular Life Sciences
water, breathing contaminated air, ingesting food, or con-
tacting contaminated soil. People working with pesticides,
fungicides, and industrial chemicals are particularly
exposed to these toxic substances and thus have a high risk
for developing reproductive or endocrine abnormalities.
EDCs can affect the endocrine systems of an organism in a
wide variety of ways. These include mimicking natural
hormones, antagonizing their action or modifying their
synthesis, metabolism, and transport. Moreover, these
substances can act via multiple pathways including mem-
brane receptors, the aryl hydrocarbon receptor, or the
enzymatic machineries involved in hormone biosynthesis/
metabolism. However, most of the reported harmful effects
of EDCs are attributed to their interference with hormonal
signaling mediated by nuclear hormone receptors (NHRs)
[36]. The first example of endocrine disruption was pro-
vided by the pharmaceutical diethylstilbestrol (DES, Fig. 1)
which was used to prevent miscarriage in women with high
risk pregnancies. In the 1970s, prenatal exposure to DES
was linked with the development of vaginal cancer in
so-called DES-daughters and DES-toxic effects were sub-
sequently attributed to the interaction of this compound
with estrogen receptors (ERs) which are members of the
NHR family [7]. Thus, most of the subsequent studies have
focused on NHRs involved in reproductive processes, in
particular ERs and the androgen receptor (AR). More
recently, studies have shown that the activity of the preg-
nane X receptor (PXR), the constitutive androstane receptor
(CAR), the estrogen related receptors (ERRs), the thyroid
hormone receptors (TRs), the retinoid X receptors (RXRs),
or the peroxisome proliferator-activated receptors (PPARs)
can also be affected by EDCs. Based on the conservation of
structural and functional NHR features and on the large
structural and chemical diversity of compounds found in the
environment, one can predict that all members of the NHR
family are potential targets of EDCs.
OH
OH
OH
OH
O
OH
OH OOH N
N
N
NH2
N
N
N
NH2
OH
Estrogen receptors (ERs)
E2 DES GEN PhIP 4-0H-PhIP
Estrogen related receptors (ERRs)
OH
OH
OH
OH
BPA
Retinoid X receptors (RXRs)
OH O
9-cis RA TBT
Sn
Cl
Sn
Cl
O
O
OH
Pregnane X receptor (PXR)
MA
O
OOH
O
OH
O
O
OH
N
N
OH OH
OH
NH
N
OH
OH
O
O
O
O
O
O
O
O
O
OH
OH
O
ZEN
O
O
OH
OH
OH
α-Zearalenol
Cl
Cl
Cl
Cl Cl
Cl
O
Cl
Cl
Cl
Cl
Chlordecone
DES TPT
Hyperforin Colupulone Rifampicin
Fig. 1 Chemical structures of representative EDCs discussed in
this review. E2 17b-estradiol, DES diethylstilbestrol, GEN geni-
stein, PhIP 2-amino-1-methyl-6-phenylimidazo [4-5-b] pyridine,
4-OH-PhIP 4-hydroxy-2-amino-1-methyl-6-phenylimidazo [4-5-b]
pyridine, ZEN zearalenone, BPA bisphenol A, 9-cis RA 9-cis retinoic
acid, TBT tributyltin, TPT triphenyltin, MA methoprene acid
A. le Maire et al.
Human NHRs (Table 1) are a family of 48 transcription
factors, many of which have been shown to be activated by
ligands. NHRs regulate cognate gene networks involved in
key physiological functions such as cell growth and dif-
ferentiation, development, homeostasis, or metabolism
[8,9]. As a consequence, dysfunctional NHR signaling
(i.e., receptor mutation or inappropriate exposure to envi-
ronmental pollutants) often leads to proliferative,
reproductive, and metabolic diseases, including hormonal
cancers, infertility, obesity, or diabetes. NHRs are modular
proteins composed of several domains (Fig. 2a), most
notably an N-terminal domain which harbors a ligand-
independent activation function (AF-1), a central DNA
binding domain (DBD), and a C-terminal ligand binding
domain (LBD) hosting a ligand-dependent transcriptional
activation function (AF-2) [8]. In the absence of the cog-
nate ligand, some NHRs are located in the nucleus, bind to
the DNA response elements of their target genes, and
recruit corepressors, while others are located in the cyto-
plasm in an inactive complex with chaperones. Ligand
binding induces major structural alterations of the receptor
LBDs leading to (1) destabilization of corepressor or
Table 1 The 48 human nuclear receptors
NHR Subtypes M, D, H Ligands
a
NR1A1, A2 TR a,bH Thyroid hormones
NR1B1, B2, B3 RAR a,b,cH Retinoic acid
NR1C1, C2, C3 PPAR a,b,cH Fatty acids, leukotriene B4 (a), fibrates (a),
prostaglandin J2 (c), thiazolidinediones (c)
NR1D1, D2 Rev-erb a,bM, D Heme
NR1F1, F2, F3 ROR a,b,cM Cholesterol (a), cholesteryl sulfate (a), retinoic
acid (b), Orphan (c)
NR1H3, H2 LXR a,bH Oxysterols, T0901317, GW3965
NR1H4, NR1H5 FXR a,bH Bile acids (a), fexaramine (a), lanosterol (b)
NR1I1 VDR H Vitamin D, 1, 25-dihydroxyvitamin D3
NR1I2 PXR H Xenobiotics, 16a-cyanopregnenolone
NR1I3 CAR H Xenobiotics
NR2A1, A2 HNF4 a,cD Orphans
NR2B1, B2, B3 RXR a,b,cD Retinoic acids
NR2C1, C2 TR 2, 4 D, H Orphans
NR2E2 TLL M, D Orphan
NR2E3 PNR M, D Orphan
NR2, F1, F2 COUP-TF I, II D, H Orphans
NR2F6 EAR-2 M Orphan
NR3A1, A2 ER a,bD Estradiol-17b, tamoxifen (a), raloxifene (a),
various synthetic compounds (b)
NR3B1, B2, B3 ERR a,b,cM, D Orphan (a), DES (b,c), 4-OH tamoxifen (b,c)
NR3C1 GR D Cortisol, dexamethasone, RU486
NR3C2 MR D Aldosterone, spirolactone
NR3C3 PR D Progesterone, medroxyprogesterone acetate,
RU486
NR3C2 AR D Testosterone, flutamide
NR4A1 NGFI-B M, D, H Orphan
NR4A2 NURR1 M, H Orphan
NR4A3 NOR1 M Orphan
NR5A1 SF1 M Orphan
NR5A2 LRH-1 Orphan
NR6A1 GCNF D Orphan
NR0B1 DAX-1 Orphan
NR0B2 SHP H Orphan
Adapted from [9]
MMonomer, Ddimer, HRXR-heterodimer
a
For ligand specific of a subtype, the concerned subtype is indicated in parentheses
Structural basis of endocrine disruption
chaperone interfaces, (2) exposure of nuclear localization
signals to allow nuclear translocation and DNA binding of
cytoplasmic receptors, and (3) recruitment of coactivators
triggering gene transcription through chromatin remodeling
and activation of the general transcription machinery.
The crystal structures of many NHR LBDs have been
determined, revealing a conserved core of 12 a-helices
(H1–H12) and a short two-stranded antiparallel b-sheet
(s1 and s2) arranged into a three-layered sandwich fold
(Fig. 2b). This arrangement generates a mostly hydropho-
bic cavity in the lower half of the domain which can
accommodate the cognate ligand. In all hormone-bound
LBD structures, the ligand binding pocket (LBP) is sealed
by helix H12. This conformation is specifically induced by
the binding of hormones or synthetic agonists and is
referred to as the ‘active conformation’ because it allows
the dissociation of corepressors and favors the recruitment
of transcriptional coactivators [1012]. It is noteworthy
that this conformational state can also be achieved by some
constitutively active orphan receptors for which no natural
ligand has been identified. In this active-form, helices H3,
H4, and H12 define a hydrophobic binding groove for short
LxxLL helical motifs (L stands for leucine and x for any
amino acid) found within coactivators (Fig. 2b). In contrast
to agonist binding, interaction with antagonists prevents the
correct positioning of helix H12, thus avoiding association
with the LxxLL motifs of coactivators [1012].
Several receptors including ERs, RXRa, ERRa, ERRc,
and PXR have been crystallized in complex with various
EDCs. This review provides an overview of the biological
function of these receptors, of their associated environ-
mental disruptors, and of the mechanisms by which some
of these substances bind to NHRs and alter their signaling
pathways at concentrations within the micro- to nanomolar
ranges.
Estrogen receptors
Estrogen receptors and their environmental
ligands (xenoestrogens)
Estrogen receptors (ERaand ERb) are receptors for the sex
hormone, 17b-estradiol (E2, Fig. 1), which play important
roles in the growth and maintenance of a diverse range of
tissues such as the mammary gland, uterus, bone, or the
cardiovascular system. Both ERs are widely distributed
throughout the body, displaying distinct but overlapping
expression patterns in a variety of tissues [13]. ERais
expressed primarily in the uterus, liver, kidney, and heart,
whereas ERbis expressed primarily in the ovary, prostate,
lung, gastrointestinal tract, bladder, and hematopoietic and
central nervous systems [14]. However, ERaand ERbare
coexpressed in a number of tissues including the mammary
gland, thyroid, adrenal, bone, and some regions of the
brain. Although ERaand ERbshare similar mechanisms of
action, several differences in the transcriptional abilities of
each receptor as well as distinct phenotypes between gene-
null animals have been identified, suggesting that these
receptors may regulate distinct cellular pathways [13,15].
Interestingly, when ERs are coexpressed, ERbexhibits an
inhibitory action on ERa-mediated gene expression [16,
17], such that ERbhas been shown to antagonize several
ERa-mediated effects including fat reduction and cellular
proliferation in breast, uterus, or prostate [1820]. Breast
cancer is one of the leading forms of cancer in the western
world, and it is well documented that the mitogenic actions
of E2 are critical in the etiology and progression of this
pathology. Accordingly, current therapies of breast cancers
are primarily based on estrogen antagonists that interact
with ERaand completely or partially shut down the cor-
responding hormone-responsive pathway. Because both
ERs are expressed in the normal breast, it is pertinent to ask
whether EDCs with different selectivity for ERs present
Fig. 2 Schematic illustration of the structural and functional orga-
nization of NHRs. aNHRs contain a well-conserved DNA binding
domain (DBD), a moderately conserved ligand binding domain
(LBD), and a highly divergent N-terminal A/B region. Two transcrip-
tional activation functions have been described in these receptors: a
‘constitutively active’ AF-1 in region A/B and an AF-2 which
corresponds to a coactivator binding surface formed by helices H3,
H4, and H12 of the LBD whose completion requires the presence of
the hormone. bOverall structure of NHR LBDs exemplified by the
ERaLBD homodimer (PDB code 1GWR) in complex with estradiol
(stick representation) and a coactivator fragment (CoA). H1–H12 and
s1, s2 denote a-helices and b-strands, respectively
A. le Maire et al.
differential effects on breast cancer proliferation. The
LBDs of ERaand ERbshare a high degree of homology in
their primary amino acid sequence and are very similar in
their tertiary architecture [11,21]. It is therefore not sur-
prising that most compounds tested so far bind to ERaand
ERbwith similar affinities or have similar potencies in
activation of estrogen response element (ERE)-mediated
reporter gene expression [22]. However, some ERa-or
ERb-selective ligands have been identified [23].
High affinity xenoestrogens
For historical reasons, most studies on endocrine disruption
have focused on sex hormone receptors, thus leading to the
discovery of a large panel of substances interfering with
ER signaling. Indeed, the group of molecules identified as
ER endocrine disruptors is highly heterogenous and
includes high affinity ligands (Kd values between 10 pM
and 1 nM), which are pharmaceutical agents contained in
contraceptive pills, human estrogens (E2, estriol, and
estrone), and some natural compounds such as zearalenone
(ZEN) and its metabolites; when available, affinities and
activities of the compounds are reported in Table 2. The
most notorious pharmaceutical EDC is DES (Fig. 1), an
orally active synthetic nonsteroidal estrogen that, due to its
teratogenic effects, was banned in 1971. In utero exposure
to DES provoked abnormal cervical, uterine, and oviduct
anatomy [24], vaginal adenocarcinoma [25], infertility, and
ectopic pregnancy [26]. Ethynylestradiol which is present
in contraceptive pills is commonly found in environmental
samples such as the water of sewage treatment plants or
rivers [2729]. In spite of its structural differences with E2,
ZEN (Fig. 1) is one of the most active endocrine disruptors
[30]. ZEN is a mycoestrogen with non-estrogenic chemical
structure which is produced by several species of Fusarium
fungi, and it widely contaminates agricultural products [31]
such that ZEN exists in almost all agricultural crops [32].
After consumption, ZEN is metabolized to provide two
major stereo isomers, a- and b-zearalenol [33,34], the
a-metabolite displaying the highest estrogenic activity [30].
Medium/low affinity xenoestrogens
Many other environmental compounds interact with ERs
with medium to low affinity (Kd values between 1 nM and
10 lM). Phytoestrogens are plant-derived substances that
have estrogenic activity [22]. They are classified according
to their chemical structure: isoflavones, flavones, flava-
nones, coumestans, stilbenes, and lignans. The most widely
studied are the isoflavones which are present at high con-
centrations in soy products and red clover [35]. In recent
years, efforts to implement healthier eating habits have
resulted in an increased consumption of soy products and,
hence, increased exposure to phytoestrogens. Genistein
(GEN, Fig. 1) is the principal phytoestrogen in soy and has
a wide range of biological actions. It has a structural simi-
larity with E2 and is an agonist for both ERs, albeit with a
marked preference for ERb[22,36]. The traditionally low
breast cancer incidence rates in Asia are commonly asso-
ciated with the high dietary intake of phytoestrogens, but
the role of phytoestrogens as anticancer or chemopreventive
agents remains under investigation. Other estrogenic
Table 2 EC
50
s of ligands for human ERa,ERb, RXRa, and PXR as reported
ERaERbRXRaPXR
E2 0.02–0.05 nM [24,47] 0.07–0.2 nM [24,47]ND 10lM[98]
DES 0.2 nM [47] 0.4 nM [47]ND10lM[98]
GEN 38 nM [24,47] 6–9 nM [24,47]ND NA[98]
PhIP 30 nM [37]NA[37]NDND
BPA 4 lM[40]5lM[40]2lM
a
[124][10 lM[98]
ZEN 1 nM
b
1nM
b
ND 10 lM[98]
a-Zearalenol 0.1 nM
b
0.1 nM
b
ND 1 lM[98]
Chlordecone 12 lM[39] Antagonist [39]ND ND
TBT ND ND 3 nM [120]ND
Hyperforin ND ND ND 0.11 lM[90]
Rifampicin ND ND ND 0.7 lM[90]
All values are reported as EC
50
s in stably or transiently transfected human cell lines
ND No relevant EC
50
for the ligand/receptor interaction has been reported to our knowledge, NA the ligand is inactive, Antagonist the ligand acts
as an antagonist
a
BPA EC
50
determined by a RXR yeast two-hybrid assay
b
Balaguer P., unpublished results
Structural basis of endocrine disruption
substances are present in foodstuffs, including the
heterocyclic amine 2-amino-1-methyl-6-phenylimidazo
[4-5-b]pyridine (PhIP) contained in cooked meat. PhIP
(Fig. 1) which induces tumors of the breast and prostate in
mice and which has been characterized accordingly as a
compound with significant estrogenic activity inducing
transcription of E2-regulated genes [37,38]. Certain
insecticides and herbicides such as dichloro-diphenyl-
tricholoethane (DDT), methoxychlor, hexachlorocyclo-
hexanes, and related compounds are also suspected to act as
environmental estrogenic chemicals. Chemically stable and
strongly lipophilic, organochlorine compounds tend to
accumulate in lipid-rich tissues and induce endocrine dis-
ruption at exposure levels measured in the environment.
Interestingly, some pesticides such as chlordecone (Fig. 1)
or methoxychlor display ERaagonistic, but ERbantago-
nistic, activities [39], thus increasing the risk of cancer
development. Finally, many industrial compounds display
estrogenic activity. These include BPA (Fig. 1), alkyl-
phenols, and benzophenones. BPA, a monomer of
polycarbonate plastics, is one of the highest-volume
chemicals in commerce. Polycarbonates are used in many
consumer products, including food and water containers,
baby bottles, medical tubing, or epoxy resin, and small
amounts of BPA can migrate from polymers to food or
water, especially when heated. Interestingly, BPA displays
estrogen-like activities at nanomolar doses, but the mech-
anism by which it exerts its biological actions remains
enigmatic. Although BPA binds ERs, its binding affinity is
several orders of magnitude lower than that of E2 [22,40],
and it has been suggested that BPA could also act through
binding to membrane ERs, G-protein coupled receptor 30
[41], or ERRc[42]. Alkylphenols are degradation deriva-
tives of alkylphenols ethoxylates which are widely used
surfactants and detergents in domestic and industrial prod-
ucts. Alkylphenols are present at significant levels in
samples of every environmental compartment examined,
including fish muscle tissue [43], and are also generally
ubiquitous in food. Benzophenones used in topical sun-
screen preparations have also been shown to activate ERs
with a stronger affinity for ERb[44,45].
Structural basis of xenoestrogen action
Natural estrogens
In the ERs, the hormone binding pockets are lined with 20
mostly hydrophobic residues that interact with the steroid
scaffold [46,47]. A few polar residues located at the two
ends of the pockets form hydrogen bonds with the polar
groups at the 3- and 17-positions of estrogens (Fig. 3a).
Selectivity of hormone binding derives from both the shape
of the hydrophobic portion of the pocket and the presence
of receptor-specific hydrogen-bond networks. In contrast to
other steroids which possess a 3-keto group, estrogens have
a 3-hydroxyl group. Thus, part of the specificity of E2 for
ERs is supported by a network of water-mediated hydrogen
bonds involving a hydrogen-bond acceptor in ERs (E353 in
ERa) while a hydrogen-bond donor (glutamine residue) is
present in other steroid receptors. At the other end of the
ligand, the 17-hydroxyl group of E2 is hydrogen-bonded to
H524 (ERa).
Xenoestrogens in food: the example of GEN and PhIP
As noted above, GEN (Fig. 1) is an isoflavonoid phytoes-
trogen which binds to both ER isotypes with a preference
for ERb[22,48]. Although it is a non-steroidal compound,
GEN possesses a diphenolic structure that shares structural
similarity with the steroidal core of estrogens. Conse-
quently, structures of GEN-bound ERaand ERbreveal that
it binds to both ERs nearly identically and in a manner
reminiscent of that observed for the natural hormone [46,
4951]. The phenol ring of GEN mimics the hydroxyl
group of the A-ring of E2 and interacts with residues E353,
R394, and a highly ordered water molecule, whereas the
isoflavone portion superimposes on the C and D rings of E2
and makes a hydrogen bond with H524 (Fig. 3b). Because
there are only two conservative residue substitutions in the
hormone binding pockets of the two receptor subtypes
(ERaL384/ERbM336 and ERaM421/ERbI373), the
explanation of the 40-fold ERbselectivity of GEN could
not be convincingly explained by visual inspection of
crystal structures. Overall, it appears that ligand selectivity
arises from a combination of subtle differences involving
sequence differences outside the binding pocket that alter
the shape of the cavity [52] and proximal ligand–amino
acid contacts [49]. Using quantum chemical calculations,
Manas et al. [49] demonstrated that, although conservative,
the two substitutions are capable of contributing signifi-
cantly to the observed ERbselectivity of GEN on the basis
of differential electronic interactions mediated by the sul-
fur-containing side chain of methionines relative to purely
aliphatic side chains of leucine and isoleucine residues
[49]. GEN-bound ERs have been crystallized both in the
presence and absence of a coactivator fragment [49,50].
Interestingly, the activation helix H12 was observed in two
different orientations in the two complexes (Fig. 3c). In the
presence of the coactivator fragment, H12 adopts the
canonical active conformation, whereas it is found in the
antagonist-like conformation in the binary ER-GEN com-
plex. This inactive conformation results from the
unwinding of helix H11 and the consecutive lengthening of
the loop connecting H11 and H12. These structural data
account for the observation that GEN acts as a partial AF-2
A. le Maire et al.
agonist, antagonizing E2 but displaying a residual agonist
activity in some cells [48] (Balaguer et al., unpublished
data). As demonstrated for other partial AF-2 agonists [53]
and in contrast to pure agonists, GEN appears unable to
induce the receptor active conformation on its own, and the
presence of a coactivator is required to fully stabilize this
conformational state. Thus, the overall activity of partial
AF-2 agonists like GEN is determined by the concentration
of coregulators which may vary between cell types or
tissues [54].
Similarly, the mutagenic compound PhIP and one of its
metabolites 4-OH-PhIP have been crystallized with ERa
[38]. PhIP is a heterocyclic amine with no hydroxyl groups
(Fig. 1) and thus differs significantly from classical ER
ligands. The phenyl ring occupies a position similar to that
adopted by the A- and B-rings of E2 but does not form the
typical hydrogen bonds with R394 and E353 (Fig. 3d). The
remaining portion of the ligand mimics the C- and D-rings
of E2. Except for the NH
2
moiety which interacts favorably
with H524, the hydrogen-bonding potential of the other
Fig. 3 Structural determinants of ligand recognition by ERs. aClose-
up view of the E2 binding pocket in ERa(PDB code 1GWR). ERa
residues involved in hormone binding as well as the E2 molecule are
colored in yellow.bSuperposition of ERaand ERbLBPs in complex
with E2 (yellow, PDB code 1GWR) and GEN (blue, PDB code
1QKM), respectively. Residues involved in E2 and GEN binding are
colored in yellow and blue, respectively. cClose-up view of the
positions of helix H12 in two GEN-ERbcomplexes in the presence
(blue, PDB code 1X7J) and absence (green, PDB code 1QKM) of a
coactivator fragment. dSuperposition of ERaLBPs in complex with
PhIP (yellow, PDB code 2QXM) and 4-OH-PhIP (pink, PDB code
2QSE). Residues involved in PhIP and 4-OH-PhIP binding are
colored in yellow and pink, respectively. eSuperposition of ERa
LBPs in complex with PhIP (blue, PDB code 2QXM) and E2 (yellow,
PDB code 1GWR), respectively. fSuperposition of ERband ERa
LBPs in complex with GEN (blue, PDB code 1X7J) and DES (green,
PDB code 3ERD), respectively. Residues involved in GEN and DES
binding are colored in blue and green, respectively. WWater
molecule. Helix and residue numbers are indicated
Structural basis of endocrine disruption
nitrogen atoms of the heterocyclic ring is not satisfied by
interaction with the receptor LBP. Compared with the ERa-
E2 complex, the structure with PhIP shows a substantial
shift in the last three turns of helix H11 against which helix
H12 docks in the active conformation (Fig. 3e). This loss
of stabilizing contacts between the two secondary struc-
tural elements likely renders helix H12 more dynamic in
solution [38]. Together, these structural data explain the
functional properties (affinity and activity) of PhIP. Inter-
estingly, the PhIP metabolite 4-OH-PhIP differs from its
parent compound by a hydroxyl group which reproduces
the interactions made by the A-ring hydroxyl of E2. This
structural observation leads to the prediction that 4-OH-
PhIP should display a better affinity towards ER and also a
higher estrogenic activity than its parent compound.
Pharmaceuticals: the example of DES
In contrast to the partial agonists described above, the
synthetic nonsteroidal DES (Fig. 1) functions as a full
agonist. The interaction of DES with ERa[47] resembles
that of GEN or E2 with the two conserved hydroxyl groups
involved in hydrogen bonds with E353, R394, and a water
molecule on one side and H524 on the other side (Fig. 3f).
Nevertheless, although the DES B ring occupies the same
position as the GEN C ring, the phenolic A ring and the
ethyl groups of DES do not superimpose the isoflavone ring
of GEN, as a rotation angle of roughly 50°is observed
between the aromatic rings (Fig. 3f). Thus, DES forms
additional non-polar contacts with the LBD. In particular,
contacts with A350 (H3), L384 (H5), F404 (S1), or L428
(H7) may account for the higher affinity of DES for the
receptor [14], whereas specific interactions made with
W383 (H4) or L540 (H12) may help stabilize the active
conformation and explain the full agonistic profile of this
compound.
Estrogen-related receptor c
Estrogen-related receptors and their environmental
ligands
ERRcis a member of the ERR subfamily of orphan
receptors, which are closely related to ERs [55]. The ERR
family includes three members, ERRa, ERRb, and ERRc.
The three receptors are very similar, with 90% sequence
identity in the DBD and more than 60% in the LBD. ERRa
is highly expressed in muscle, heart, bone, and adipose
tissue, as well as in the central nervous system [56]. ERRc
is expressed in a tissue-restricted manner, for example very
strongly in the mammalian brain during development, and
then in the brain, lung, and many others tissues during
adulthood. In terms of structure, ERRs are very close to
ERs. Sequence alignment reveals a 60% identity in the
DBD regions and a moderate similarity (\35%) of the
LBDs, consistent with the incapacity of ERRs to bind E2
[57]. Nevertheless, it has been demonstrated that ERRs can
interfere with estrogen signaling [55]. Indeed, ERs and
ERRs recognize the same DNA binding elements, share
common target genes and are coexpressed in many tissues
[58,59]. Recent publications report that ERRamay rep-
resent a biomarker of poor prognosis in breast and ovarian
cancers suggesting an involvement of the receptor in cell
proliferation. In contrast, exogenous overexpression of
ERRband ERRcin prostate cancer cell line results in
inhibition of proliferation [60]. Furthermore, treatment
with an ERRb/cagonist has been shown to promote this
antiproliferative effect, consistent with ERRcbeing a
favorable prognosis factor [61]. Additionally, several lines
of evidence suggest that ERRs play a central role in reg-
ulating energy metabolism. ERRs are expressed in tissues
associated with lipid metabolism and high energy demands,
their transcriptional activity is highly dependent on the
presence of coregulators implicated in the control of met-
abolic programs, genetic studies in mice reveal that their
presence is essential for the generation of energy and
related tissue-specific functions, and functional genomics/
proteomics studies have associated ERRs with the control
of vast metabolic gene networks, in particular those
involved in mitochondrial biogenesis and function [62].
The rise in the incidence of metabolic syndromes correlates
with the rise in the use and distribution of industrial
chemicals that may play a role in generation of obesity
[63], suggesting that EDCs and ERRs may be linked to this
epidemic crisis.
To date, the ERRs have not been shown to interact with
any physiologically relevant small molecules, suggesting
that these receptors manifest constitutive activity [64,65],
and, indeed, crystallographic analyses of ERRs indicated
that these receptors adopt the transcriptionally active con-
formation in the absence of any ligand [65]. The
compounds screened for activity on the ERRs were known
endocrine disruptors with estrogen-like activity. Two
organochlorine pesticides, toxaphene and chlordane, were
found to act as weak antagonists of ERRa[66]. DES
inhibits the constitutive activity of all three ERRs [67,68].
The phytoestrogen kaempferol is an ERRaand cantagonist
[69]. In contrast to the numerous reported ERR antagonists,
few agonists of ERR have been identified. However, phy-
toestrogens (GEN, daidzein) function as non-selective and
low affinity ERR agonists [70]. Finally BPA, bisphenol E
(BPE), and others phenols were recently reported to bind
with high affinity to ERRc[42,64].
A. le Maire et al.
Structural basis of BPA- and DES-ERR interactions
Structure of the unliganded ERRc
The crystal structure of unliganded ERRcLBD [65,71]
superimposes well with that of the E2-bound ERaLBD,
revealing that the receptor can adopt a transcriptionally
active conformation in the absence of ligand (Fig. 4a). This
conformation allows an interaction with LxxLL motifs of
coactivators via a charge clamp comprised of K284 (H3)
and E452 (H12). The structure reveals that ERRcexhibits a
relatively small putative LBP (Fig. 4a) delimited by 22
amino acids and measuring 280 A
˚
3
in volume compared
with 480 A
˚
3
for ERa[71]. This observation may explain
why most of the exogenous compounds interacting with
ERRs act as antagonists rather than agonists (see the
description of the DES-bound structure bellow). The empty
ERRcpocket is formed by hydrophobic and a few polar
(Y326, N346) or charged (E275, R316) residues. The
substitution of several residues between ERaand ERRc
LBPs [L525/F435 (H11), G521/A431 (H11), and L540/
F450 (H12)] leads to the decrease of the cavity volume for
ERRcthat precludes E2 binding to the orphan receptor.
Nevertheless, it was predicted that ERRccould bind small
phenol-containing ligands because the two charged resi-
dues which form a hydrogen bonding network anchoring
the 3-OH group of the A-ring of estradiol in ERa(E353 in
H3 and R394 in H5) are conserved in ERRc(E275 and
R316).
Structure of BPA-bound ERRc
Although a natural ligand remains to be found for ERRc,
several synthetic ligands have been identified for this
receptor [42,64]. All display a conserved phenol ring such
as BPA, a small symmetric molecule with two phenol rings
and two methyl groups linked to a central sp
3
carbon atom
(Fig. 1). The structure of ERRcin complex with BPA
(Fig. 4b) has been solved, revealing an overall protein
conformation indistinguishable from that of the unliganded
receptor [7274]. A close look at the LBP shows that, as
previously anticipated, one of the two phenol-hydroxyl
groups of BPA forms hydrogen bonds with residues E275
(H3) and R316 (H5) while the second is hydrogen bonded
to N346 in H7. A hydrogen bond between Y326 and N346
holds N346 in position to interact with the second phenol
group of BPA. Interestingly, this asparagine residue is not
conserved in ERRaand b, thus accounting for the specific
ERRc-BPA interaction. Moreover, comparison of ERRc
and ERRaLBPs reveals a much smaller cavity for the latter
[75]. The replacement of two alanine residues (A272 and
A431) in ERRcby a phenylalanine (F328) and a valine
(V491) in ERRaaccounts for this size reduction and fur-
ther explains why BPA does not bind to the latter. BPA
binding provokes only minimal LBP rearrangements.
E275, which appears disordered in the unliganded struc-
ture, adopts a unique conformation to make a hydrogen
bond with one OH group of BPA, and the side chain of
L345 moves away from the pocket upon BPA binding to
open up the cavity and make room for the second phenol
group of BPA. In summary, ERRcpossesses a LBP to
which BPA can bind with high affinity and specificity
while preserving the constitutively active conformation of
the receptor. The limited impact of BPA binding on
receptor structure is consistent with the fact that this ligand
does not enhance or disrupt coactivator binding and thus
appears as a functionally silent ligand [72,73]. Thus, the
classical mechanism of NHR activation involving the
Fig. 4 Structural determinants of ligand recognition by ERRs.
aOverall structure of unliganded ERRcLBD (PDB code 1KV6) in
cartoon representation. The unoccupied LBP is highlighted in black.
bSuperposition of ERRcLBPs in absence of ligand (yellow, PDB
code 1KV6) and in complex with BPA (green, PDB code 2E2R).
Residues involved in BPA binding are colored in green and
equivalent residues in the unliganded receptor are colored in yellow.
cSuperposition of ERRcLBP in absence of ligand (yellow, PDB code
1KV6) and of the DES-bound form (blue, PDB code 1SP9). Residues
involved in DES binding are colored in blue and equivalent residues
in the unliganded receptor are colored in yellow
Structural basis of endocrine disruption
re-localization and stabilization of helix H12 in the active
conformation does not explain how ERRccould mediate
the estrogenic effects of BPA. Rather, thermal stability
studies revealed that BPA binding leads to global ther-
modynamic stabilization of ERRcLBD, a phenomenon
which could increase steady state levels of the receptor and
impact both its cellular half-life and biological activity
[7173].
Structure of DES-bound ERRc
In contrast to BPA, DES deactivates ERRs and acts as a so-
called inverse agonist by disrupting the basal interaction
between ERRs and coactivators [68]. The crystal structure
of the DES-bound ERRc(Fig. 4c) shows that DES-medi-
ated inverse agonism is based on the rearrangement of the
side chain of F435 (H11), which upon ligand binding flips
out and sterically interferes with H12, thus displacing it
from its agonist position [76]. Interestingly, the recently
reported crystal structure of ERRcLBD in complex with
the synthetic agonist GSK4716 [71] revealed an unex-
pected rearrangement of the phenol binding residues E275
and R316, which allows access to an additional pocket of
390 A
˚
3
resulting in the formation of a single combined
pocket of 610 A
˚
3
. This structure reveals that ERRccan
accommodate larger ligands than previously anticipated.
Pregnane X receptor
Pregnane X receptor and its environmental ligands
PXR, also known as steroid and xenobiotic receptor (SXR)
and pregnane activated receptor (PAR), is activated by
xenobiotics and acts as a master regulator of phase I to III
of drug metabolism. PXR is involved in the biosynthesis,
distribution, and metabolism of steroids, bile acids, and
xenobiotics [77]. Activated PXR binds to gene promoters
as a heterodimer with RXR and induces the expression of
target genes such as CYP3A. This receptor plays a prom-
inent role as protector of the endocrine system from
chemical perturbation by sensing increases in the concen-
tration of a multitude of EDCs and inducing detoxification
pathways to prevent other NHRs from interactions with
these chemicals. Literature has described PXR activation as
‘Jekyll and Hyde’ or ‘ying and yang’ [78,79] to illustrate
both its beneficial and prejudicial effects. In fact, activation
of PXR can be positive, as it accelerates the detoxification
process and consequently the elimination of xenobiotics. In
contrast, the premature metabolism of active compounds
such as hormones or drugs means that target responses will
not be activated and can lead to harmful effects or adverse
interactions. The metabolism of inactive compounds can
also lead to the synthesis of active metabolites, such as the
transformation of methoxychlor by CYP2C11, a PXR-
induced enzyme, into phenolic estrogenic compounds [80].
It has also been observed that coregulatory proteins work in
concert with ligands to stabilize PXR LBD such that the
ligand-induced transcriptional response would depend on
which coregulator binds to PXR LBD [81]. This observa-
tion suggests that promoter context (i.e., the combination of
coregulators associated with a target gene promoter) is an
important parameter in determining the transcriptional
activity of PXR. Support for this notion comes from greater
PXR activation by pregnenolone on a CYP3A4 PXR
response element (PXRE) than on a PXRE from the mul-
tidrug resistance protein 1 (MDR1) gene which correlates
with recruitment of the coactivators SRC-1 and not AIB-1
[82]. Overall, it is difficult to conclude whether the xeno-
biotic activation of PXR is predominantly negative or
positive, but clearly PXR plays an essential role in endo-
crine disruption. Unlike most NHRs that tend to be
specialized to bind few ligands with structural homologies,
PXR is able to bind a large number of structurally diverse
ligands with a wide range of affinities [8387]. PXR binds
a multitude of drugs such as the antibiotic rifampicin
[8891], the anti-cancer taxol [92,93], the anti-cholesterol
SR12813 [91,94], the barbituric phenobarbital [90], the St
John’s Wort anti-depressor hyperforine (Fig. 1)[95,96],
and many more, recently reviewed in [97]. Furthermore,
numerous studies have focused on its ability to bind
environmental compounds, such as pesticides [91], natural
and synthetic estrogens and alkylphenols [87,98100],
polychlorinated biphenyls [98,101], brominated flame
retardants [102], or antimicrobial triclosan [98].
Structural basis of PXR-xenobiotics interactions
By virtue of this low ligand selectivity and its dual
response characteristics, PXR differs from the receptors
previously discussed. Contrary to ER and RXR, which
possess high affinity-specific physiological ligands or ERR
which has no known natural ligand, PXR is activated by a
variety of structurally distinct endogenous and exogenous
compounds. Crystallographic studies have revealed several
unique characteristics of PXR that account for its promis-
cuous ligand binding properties. First, PXR possesses a
large LBP which can accommodate compounds with larger
volumes than that of classical NHR ligands (823 Da for the
macrocyclic rifampicin vs 272 for E2). Second, several
loops clustering at the bottom of the LBD confer a high
plasticity allowing the receptor LBP to adopt different
shapes according to the bound ligands. Three of these
flexible elements are found in a PXR-specific sequence of
approximately 60 residues inserted between helices H1 and
H3. This segment folds as a two-stranded antiparallel
A. le Maire et al.
b-sheet (s1 and s1’, residues 210–228), a disordered loop
(177–197), and two flexible loops (198–210 and 229–235)
that appear to be characterized by either rather high ther-
mal B factors (indicating structural mobility) in the
unliganded and hyperforin-bound PXR [85,94] or com-
plete disorder in the structures with rifampicin [103] and
colupulone [104] (Fig. 5a, b). Another region adjacent to
the PXR LBP with high structural dynamics resides
between s4 and H7 (residues 309–321). Thus, it appears
that PXR displays a flexible portion of its LBP that allows
the receptor to modify its shape and volume to bind ligands
via an induced-fit mechanism. For example, the binding
cavity of unbound PXR is 1,294 A
˚
3
in volume but expands
to 1,544 A
˚
3
in the hyperforin-PXR complex. Third, the
unliganded PXR LBP contains 28 residues including eight
polar amino acids with the potential to form hydrogen
bonds with ligands. Among them, three polar (S247, Q285,
and H407) and three hydrophobic (M243, W299, and
F420) residues are consistently involved in ligand binding.
The remaining contacting residues depend on the size and
the chemical nature of the bound ligands. For example,
rifampicin contacts 18 residues while hyperforin interacts
with 13 amino acid side chains. Rifampicin which is 40%
larger than hyperforin and occupies regions not filled by
the latter makes specific interactions with residues V211,
L239, L308, and R410 (Fig. 5c, d). Interestingly, PXR is
also able to bind smaller endogenous molecules such as
steroidal hormones. The recently reported crystal structure
of PXR LBD in complex with estradiol reveals that E2 fills
only a very small part of the LBP, leaving 1,000 A
˚
3
unoccupied [87]. Moreover, the structure shows that the
position of the bound ligand as well as the key stabilizing
interactions differ significantly from that found in the
E2-bound ER complex structure. Notably, E2 in PXR binds
closely adjacent to H12 in an orientation nearly perpen-
dicular to that observed in the ER complex, and key
E2-contacting residues in ER (E353, R394, and H524 in
ERa) are replaced in PXR such that the 3-OH group on the
A-ring forms an hydrogen bond with S247, whereas the 17
OH-group on the D-ring is hydrogen bonded to R410 and
Fig. 5 Structural determinants of ligand recognition by PXR.
aClose-up view of the flexible elements found specifically in PXR.
They are inserted between helices H1 and H3 and comprise two-
stranded antiparallel b-sheet (s1 and s10), a disordered loop and two
flexible loops. These elements are visible in the structure of
unliganded PXR (green, PDB code 1ILG) whereas they are too
disordered to be visible in the structure of PXR bound to rifampicin
(purple, PDB code 1SKX). bThe structure of PXR LBD in complex
with hyperforin (PDB code 1M13) is rendered by thermal displace-
ment parameters (B factor) ranging from blue (low) to red (high).
cClose-up view of PXR LBP in complex with rifampicin (PDB code
1SKX). PXR residues involved in rifampicin binding are colored in
purple and rifampicin is shown in yellow.dClose-up view of PXR
LBP in complex with hyperforin (PDB code 1M13). PXR residues
involved in hyperforin binding are colored in green and hyperforin is
shown in yellow
Structural basis of endocrine disruption
S208 (not shown). Globally, the capacity of PXR to
accommodate a variety of chemical scaffolds critically
involves the deformability of its cavity together with the
unique distribution of hydrophobic, polar, and charged
residues over the LBP. CAR is another receptor that
responds to several endo- and xenobiotics [105,106].
However, the smaller and less conformable LBP of CAR is
most probably the reason why this receptor recognizes
fewer EDCs and plays a lesser role than PXR as a xeno-
sensor. Together, these structural observations contribute to
the better understanding of how PXR can detect structur-
ally and chemically different compounds. However, they
also point to the fact that, due to the high plasticity of PXR
LBP, modeling of the interaction between this receptor and
EDCs in view of computational prediction of receptor–
ligand interaction might be difficult.
Retinoid X receptors
Retinoid X receptors and their environmental ligands
RXRa,b, and coccupy a particular position in the NHR
superfamily, as they are the common heterodimerization
partners for one-third of the 48 family members (Table 1).
Consequently, RXRs are involved in the control of multiple
signaling pathways in both ligand-dependent and -inde-
pendent manners [107]. RXRs form three different types of
dimers: RXR homodimer, permissive heterodimers, and
non-permissive heterodimers. RXR permissive heterodi-
mers (e.g., RXR-PPAR, RXR-LXR, or RXR-PXR) are
activated upon ligand binding to RXR even in the absence
of the partner receptor ligand, whereas non-permissive
heterodimers (e.g., RXR-RAR, RXR-VDR, or RXR-TR)
cannot be activated by the RXR ligand alone, and RXR
serves as a silent partner in absence of partner ligand.
Differential corepressor interaction accounts for the diverse
activation profiles of permissive and non-permissive het-
erodimers [108]. However, in both cases, RXR ligands and
ligands of the partner receptors can act synergistically to
activate heterodimers [109,110]. This regulatory control of
nuclear signaling pathways by multiple RXR heterodimers
allows environmental RXR ligands to potentially trigger a
multitude of adverse effects on human health.
RXRs are activated by 9-cis retinoic acid (9-cis RA;
Fig. 1) as well as docosahexaenoic acid [111,112]. In
addition, synthetic RXR ligands, referred to as rexinoids,
are already used or are being developed for cancer therapy
and treatment of metabolic diseases [113]. On the other
hand, RXR can also be activated by environmental exo-
genous chemicals, including some organotin compounds
(Fig. 1), which in so doing act as endocrine disruptors
[114116]. The RXRa/PPARcheterodimer was reported to
play a major role in mediating the deleterious effects of
organotins which are ubiquitously present throughout the
environment due to their widespread use since the 1960s in
many industrial and agricultural processes [117]. Impor-
tantly, these studies demonstrated that the two biocides,
tributyltin (TBT; Fig. 1) and triphenyltin (TPT; Fig. 1), are
able to activate this heterodimer at nanomolar concentra-
tions thereby inducing various toxicities ranging from
adipogenesis in mammals [118,119] to the development of
male reproductive organs in female gastropods [120,121].
Interestingly, a recent study revealed that in fact TBT
activates all three RXR/PPARa,c,dheterodimers essen-
tially through its interaction with RXR [122]. Another
example of an EDC that modulates RXR function is
methoprene which is an insect growth regulator in
domestic and agricultural use as a pesticide. At least one
metabolite of methoprene, methoprene acid (MA, Fig. 1),
directly binds to and activates mammalian RXRs [123].
Finally, BPA and other phenols (4-tert-octylphenol,
nonylphenol mixture) showed high induction of RXR
directly or after being metabolized [124].
Structural basis of xenorexinoids action
MA binds to and activates the three subtypes of RXR with
a lower affinity than 9-cis RA [123]. In contrast, TPT and
TBT, two of the most active organotin compounds, bind to
and activate RXRs as efficiently as 9-cis RA (Table 1).
Structures of the complexes between RXR and TBT [122],
TPT (described in this review) and MA [125], were
obtained, and their comparison with that of the 9-cis RA-
bound RXR [126] allowed the description of the binding
mode of these EDCs and the explanation of their different
binding affinities. In RXR, 9-cis RA is buried in an
essentially hydrophobic pocket formed by residues located
on helices H3, H5, H7, and H11, and the b-turn (Fig. 6a).
RXR features an L-shaped pocket which requires a sharp
bend or a twist of the polyene side chain in the retinoid
skeleton to allow binding. The pocket is sealed by R316
which forms an ionic interaction with the carboxylate
group of 9-cis RA on one side and by helices H7, H11 and
H12 on the other side. Thus, the restrictive structural,
chemical, and conformational features of the RXR LBP are
perfectly adapted to accommodate the 9-cis isomer and not
the elongated all-trans isomer of retinoic acid. Interest-
ingly, the pesticide MA displays molecular characteristics
similar to 9-cis RA which allow interaction with RXR,
namely a conformable aliphatic chain and a carboxylate
moiety. In the crystal structure [125], it was observed that,
contrary to 9-cis RA, MA does not interact with H12
directly, an observation that could account for the poor
agonist-driven H12 recruitment. MA adopts an L-shaped
conformation to fit the RXR LBP by two consecutive 90°
A. le Maire et al.
bond rotations, and thereby mimics the sharp cis-bend of
9-cis RA (Fig. 6b). In the same manner as 9-cis RA, the
carboxylate is anchored by an ionic interaction with R387
from helix H5. The methoxy iso-butyl moiety is smaller
than the b-ionone ring of 9-cis RA, and therefore makes
fewer hydrophobic contacts with the receptor. These dif-
ferences in interactions with the receptor most likely
account for the lower affinity observed for MA in com-
parison with 9-cis RA. In contrast to MA, the two
organotin compounds, TBT or TPT, interact strongly with
RXRs and act as full RXR agonists whereas they neither
structurally nor chemically resemble 9-cis RA (Fig. 1).
Organotins form a group of more than 250 tin compounds
containing a variety of mono-, di-, tri-, or tetra-substituted
organic groups and, contrary to classical rexinoids (RXR-
specific ligands), they do not contain an acidic head group
for interaction with R316. The previously reported struc-
ture of RXR in complex with TBT [122], and the crystal
structure of TPT-bound RXR presented here, reveal that, as
compared with 9-cis RA, the two organotins occupy only a
small part of RXR LBP (Fig. 6c). Moreover, the structures
show that the high affinity of the organotins for RXR
derives mainly from the formation of a covalent bond
between the tin atom of organotins and the sulfur atom of a
cysteine residue (C432) in helix H11. The remaining
interactions involve van der Waals contacts between
almost all organotin carbon atoms and RXR residues.
Interestingly, although TBT and TPT interact with only a
subset of binding pocket residues, they are engaged in
enough essential contacts to efficiently stabilize RXRa
in its active conformation. The particular position of C432
in helix H11 likely accounts for the high agonist activity of
TBT and TPT. Indeed, several studies have pointed out the
importance of this secondary structural element which
should adopt a helical conformation for full receptor acti-
vation [38,127,128]. The presence of a cysteine residue at
this particular H11 position is unique to RXRs and may
allow for the stabilization of the helical conformation of
H11 and of the active receptor form by TBT and TPT.
Other NHRs including PPARcand the glucocorticoid
receptor (GR) have been identified as organotin targets
[114,118,119,129]. PPARcand GR contain cysteine
residues at different positions which could help to anchor
the organotins in the LBPs. However, these cysteine resi-
dues could fix the tin compounds in regions of the LBPs
which do not allow efficient stabilization of the active
receptor conformation. Accordingly, organotins were
shown to act as partial agonist or antagonist for PPARc
[122] and GR [129], respectively, indicating that tin-con-
taining compounds could use the specific Sn-S interaction
to modulate the transcriptional activity of a number of
NHRs, the functional outcome being dictated by the
structure of the organotin and the position of the anchoring
cysteine in the LBP.
Concluding comments
Deregulation of NHR-mediated transcription accounts for
the deleterious effects of many EDCs. Thus, characteriza-
tion of the harmful interaction between receptors and
environmental compounds, both at the structural and
functional levels, as well as the development of robust in
vivo, in vitro, and in silico screening methods, are impor-
tant for assessment of the toxic potential of large numbers
of chemicals [5,130132]. In this context, computer-aided
technologies which allow activity prediction of endocrine
disruptors and environmental risk assessment have been
developed. Computational tools very often include the use
of quantitative structure–activity relationship (QSAR)
methods in which the chemical structure of a compound is
quantitatively correlated with its biological activity through
Fig. 6 Structural determinants of ligand recognition by RXRs.
aClose-up view of the 9-cis-RA binding pocket of RXRa(PDB
code 1FBY). RXR residues involved in 9-cis-RA binding are colored
in yellow and 9-cis-RA is shown in orange.bSuperposition of RXRa
and RXRbLBPs in complex with 9-cis-RA (orange, PDB code
1FBY) and methoprene acid (violet, PDB code 1UHL), respectively.
Residues involved in 9-cis-RA and methoprene acid binding are
colored in yellow and pink, respectively. cSuperposition of RXRa
LBPs in complex with 9-cis-RA (orange, PDB code 1FBY) and TPT
(blue, PDB code 3KWY). Residues involved in 9-cis-RA and TPT
binding are colored in yellow and blue, respectively
Structural basis of endocrine disruption
a number of molecular descriptors, including the molecular
weight and parameters, to account for hydrophobicity,
topology, or electronic properties [133137]. The growing
number of crystal structures of NHR LBDs in the presence
of various ligands greatly facilitates the understanding of
ligand binding and receptor modulation. This structural
information can also be used with modeling and docking
tools to predict the interaction of EDCs with NHRs
[138142]. However, several levels of structural com-
plexity limit the current application of these computational
approaches.
The examples reviewed in the present article indicate
that EDCs can be roughly divided into three classes
depending on their structural and chemical proximity with
endogenous hormones. The first class contains all EDCs
mimicking the natural hormones through conserved pro-
tein–ligand contacts. Representative examples for ERs and
RXRs are DES, GEN, and MA (Fig. 1) which display most
of the structural and chemical requirements for interaction
with their respective receptors in a hormone-like fashion.
Other EDCs such as a-zearalenol (Fig. 1) most probably
fall into this category [143]. The second class contains
compounds displaying only a fraction of the anchoring
chemical groups exhibited by the endogenous hormone as
exemplified by PhIP and its metabolite 4-OH-PhIP [144].
Lastly, the third class corresponds to chemicals which
display molecular properties distinct from the endogenous
ligands but that still bind to NHRs by employing radically
different binding mechanisms. The best experimentally
validated example of this concept is the case of TBT and
TPT. Although they do not possess the classical features of
rexinoids, namely a carboxylic head and a long aliphatic
tail, these organotin compounds bind RXRs with high
affinity and are able to efficiently activate these receptors
through the establishment of a covalent link with a cysteine
residue of the LBP. Similarly, due to a chemical structure
unrelated to that of E2, the pesticide chlordecone (Fig. 1)
should exert its estrogenic activity through ER via a non-
estrogen-like binding mechanism.
Computational methods can be effective in recognizing
the putative endocrine activity of compounds of the two-
first classes of EDCs. As an example, ERRccan be bound
by a high variety of phenols [42,69], and modeling and
docking tools can provide valuable help in selecting ERRc
binding compounds from this large family of environ-
mental contaminants [145,146]. In contrast, prediction of
the endocrine disruptive activity of the last category of
compounds will likely be much less straightforward.
Another difficult case is PXR which is activated by a
variety of structurally distinct endogenous and exogenous
compounds. PXR ligands can be categorized into potency
groups, weak, moderate, and strong [83,84,87,91,99],
and modeling and docking tools could be used to predict
the affinity of new chemicals for PXR. However, due to the
structural flexibility of its LBP, predicting the responses of
PXR to environmental chemicals remains a difficult task.
One of the major challenges of computational methods
could be the prediction of all the potential target receptors
for a given chemical compound. Nevertheless, the example
provided by the structures of E2 bound to PXR and ER
revealing that the two receptors bind the same hormone in
remarkably distinct manners suggests that one cannot
easily take advantage of the information provided by the
structure of a given receptor/ligand complex to predict the
binding mode of the same ligand to another receptor.
Finally, compounds that target so-called orphan receptors
might also be difficult to identify owing to the significant
and hardly predictable conformational changes generally
associated with ligand binding.
To date, only a few EDC-bound NHRs have been
crystallized as compared with the 140,000 synthetic
chemicals used in consumer products. Thus, it appears that
efforts in elucidating the mechanisms of NHR/EDC inter-
actions by crystallography and other structural methods
must be pursued in order to deal with difficult cases and to
increase our knowledge of the structural mechanisms and
molecular interactions used by different receptors and a
wide range of structurally and chemically diverse com-
pounds. As exemplified by organotin compounds, such
studies can also reveal unforeseen binding modes and
provide guidelines for the rational design of novel NHR
modulators. Together with the improvement of computa-
tional methods, this mounting structural data should
increase the effectiveness of in silico screening strategies.
As the European Union’s 2006 Registration, Evaluation,
Authorization, and Restriction of Chemicals (REACH)
regulation aims to assess the toxicity of more than 100,000
synthetic chemicals, there is a strong demand for such
computational tools which would allow reduction of the
cost of the evaluation as well as animal lives [147].
Acknowledgments This work was supported by funds from the
INSERM, CNRS, Universite
´Montpellier 1 & 2, the French National
Research Agency (ANR-07-PCVI-0001-01 to W.B.), the Agence
Franc¸aise de Se
´curite
´Sanitaire de l’Environnement et du Travail
(AFSSET, RD-2005-007 to P.B.) and the European Union Commis-
sion (CASCADE FOOD-CT-2004-506319 to P.B.). We thank
Catherine Teyssier, Pierre Germain, and Catherine A Royer for crit-
ical reading of the manuscript.
References
1. Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, Hauser
R, Prins GS, Soto AM, Zoeller RT, Gore AC (2009) Endocrine-
disrupting chemicals: an Endocrine Society scientific statement.
Endocr Rev 30:293–342
2. Hotchkiss AK, Rider CV, Blystone CR, Wilson VS, Hartig PC,
Ankley GT, Foster PM, Gray CL, Gray LE (2008) Fifteen years
A. le Maire et al.
after ‘Wingspread’’: environmental endocrine disrupters and
human and wildlife health: where we are today and where we
need to go. Toxicol Sci 105:235–259
3. Swedenborg E, Ruegg J, Makela S, Pongratz I (2009) Endocrine
disruptive chemicals: mechanisms of action and involvement in
metabolic disorders. J Mol Endocrinol 43:1–10
4. Tabb MM, Blumberg B (2006) New modes of action for
endocrine-disrupting chemicals. Mol Endocrinol 20:475–482
5. Janosek J, Hilscherova K, Blaha L, Holoubek I (2006) Envi-
ronmental xenobiotics and nuclear receptors: interactions,
effects and in vitro assessment. Toxicol In Vitro 20:18–37
6. Toppari J (2008) Environmental endocrine disrupters. Sex Dev
2:260–267
7. Herbst AL, Ulfelder H, Poskanzer DC (1971) Adenocarcinoma
of the vagina: association of maternal stilbestrol therapy with
tumor appearance in young women. N Engl J Med 284:878–881
8. Gronemeyer H, Gustafsson JA, Laudet V (2004) Principles for
modulation of the nuclear receptor superfamily. Nat Rev Drug
Discov 3:950–964
9. Germain P, Staels B, Dacquet C, Spedding M, Laudet V (2006)
Overview of nomenclature of nuclear receptors. Pharmacol Rev
58:685–704
10. Bourguet W, Germain P, Gronemeyer H (2000) Nuclear
receptor ligand-binding domains: three-dimensional structures,
molecular interactions and pharmacological implications.
Trends Pharmacol Sci 21:381–388
11. Pike AC (2006) Lessons learnt from structural studies of
the oestrogen receptor. Best Pract Res Clin Endocrinol Metab
20:1–14
12. Renaud JP, Moras D (2000) Structural studies on nuclear
receptors. Cell Mol Life Sci 57:1748–1769
13. Couse JF, Korach KS (1999) Estrogen receptor null mice: what
have we learned and where will they lead us? Endocr Rev
20:358–417
14. Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J,
Nilsson S, Gustafsson JA (1997) Comparison of the ligand
binding specificity and transcript tissue distribution of estrogen
receptors alpha and beta. Endocrinology 138:863–870
15. Curtis SW, Washburn T, Sewall C, DiAugustine R, Lindzey J,
Couse JF, Korach KS (1996) Physiological coupling of growth
factor and steroid receptor signaling pathways: estrogen receptor
knockout mice lack estrogen-like response to epidermal growth
factor. Proc Natl Acad Sci USA 93:12626–12630
16. Liu MM, Albanese C, Anderson CM, Hilty K, Webb P, Uht RM,
Price RH Jr, Pestell RG, Kushner PJ (2002) Opposing action of
estrogen receptors alpha and beta on cyclin D1 gene expression.
J Biol Chem 277:24353–24360
17. Pettersson K, Delaunay F, Gustafsson JA (2000) Estrogen
receptor beta acts as a dominant regulator of estrogen signaling.
Oncogene 19:4970–4978
18. Weihua Z, Saji S, Makinen S, Cheng G, Jensen EV, Warner M,
Gustafsson JA (2000) Estrogen receptor (ER) beta, a modulator
of ERalpha in the uterus. Proc Natl Acad Sci USA 97:5936–
5941
19. Lindberg MK, Moverare S, Skrtic S, Gao H, Dahlman-Wright
K, Gustafsson JA, Ohlsson C (2003) Estrogen receptor (ER)-
beta reduces ERalpha-regulated gene transcription, supporting a
‘ying yang’ relationship between ERalpha and ERbeta in mice.
Mol Endocrinol 17:203–208
20. Ogawa S, Eng V, Taylor J, Lubahn DB, Korach KS, Pfaff DW
(1998) Roles of estrogen receptor-alpha gene expression in
reproduction-related behaviors in female mice. Endocrinology
139:5070–5081
21. Nahoum V, Bourguet W (2007) Androgen and estrogen recep-
tors: potential of crystallography in the fight against cancer. Int J
Biochem Cell Biol 39:1280–1287
22. Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van
der Saag PT, van der Burg B, Gustafsson JA (1998) Interaction
of estrogenic chemicals and phytoestrogens with estrogen
receptor beta. Endocrinology 139:4252–4263
23. Veeneman GH (2005) Non-steroidal subtype selective estro-
gens. Curr Med Chem 12:1077–1136
24. Kaufman RH, Korhonen MO, Strama T, Adam E, Kaplan A
(1982) Development of clear cell adenocarcinoma in
DES-exposed offspring under observation. Obstet Gynecol
59:68S–72S
25. Herbst AL (1981) The current status of the DES-exposed pop-
ulation. Obstet Gynecol Annu 10:267–278
26. Newbold R (1995) Cellular and molecular effects of develop-
mental exposure to diethylstilbestrol: implications for other
environmental estrogens. Environ Health Perspect 103(Suppl
7):7–83
27. Cargouet M, Perdiz D, Mouatassim-Souali A, Tamisier-Karolak
S, Levi Y (2004) Assessment of river contamination by estro-
genic compounds in Paris area (France). Sci Total Environ
324:55–66
28. Ribeiro C, Tiritan ME, Rocha E, Rocha MJ (2009) Seasonal and
spatial distribution of several endocrine-disrupting compounds
in the Douro River Estuary, Portugal. Arch Environ Contam
Toxicol 56:1–11
29. Ying GG, Kookana RS, Kumar A, Mortimer M (2009) Occur-
rence and implications of estrogens and xenoestrogens in
sewage effluents and receiving waters from South East
Queensland. Sci Total Environ 407:5147–5155
30. Takemura H, Shim JY, Sayama K, Tsubura A, Zhu BT, Shimoi
K (2007) Characterization of the estrogenic activities of zea-
ralenone and zeranol in vivo and in vitro. J Steroid Biochem
Mol Biol 103:170–177
31. Trenholm HL, Warner RM, Fitzpatrick DW (1984) Rapid,
sensitive liquid chromatographic method for determination of
zearalenone and alpha- and beta-zearalenol in wheat. J Assoc
Off Anal Chem 67:968–972
32. Veldman A, Borggreve GJ, Mulders EJ, van de Lagemaat D
(1992) Occurrence of the mycotoxins ochratoxin A, zearalenone
and deoxynivalenol in feed components. Food Addit Contam
9:647–655
33. Kiessling KH, Pettersson H (1978) Metabolism of zearalenone
in rat liver. Acta Pharmacol Toxicol (Copenh) 43:285–290
34. Kuiper-Goodman T, Scott PM, Watanabe H (1987) Risk
assessment of the mycotoxin zearalenone. Regul Toxicol Phar-
macol 7:253–306
35. Messina M, Kucuk O, Lampe JW (2006) An overview of the
health effects of isoflavones with an emphasis on prostate cancer
risk and prostate-specific antigen levels. J AOAC Int 89:1121–
1134
36. Escande A, Pillon A, Servant N, Cravedi JP, Larrea F, Muhn P,
Nicolas JC, Cavailles V, Balaguer P (2006) Evaluation of ligand
selectivity using reporter cell lines stably expressing estrogen
receptor alpha or beta. Biochem Pharmacol 71:1459–1469
37. Lauber SN, Ali S, Gooderham NJ (2004) The cooked food
derived carcinogen 2-amino-1-methyl-6-phenylimidazo[4, 5-b]
pyridine is a potent oestrogen: a mechanistic basis for its tissue-
specific carcinogenicity. Carcinogenesis 25:2509–2517
38. Nettles KW, Bruning JB, Gil G, Nowak J, Sharma SK, Hahm
JB, Kulp K, Hochberg RB, Zhou H, Katzenellenbogen JA,
Katzenellenbogen BS, Kim Y, Joachmiak A, Greene GL (2008)
NFkappaB selectivity of estrogen receptor ligands revealed by
comparative crystallographic analyses. Nat Chem Biol 4:241–
247
39. Lemaire G, Mnif W, Mauvais P, Balaguer P, Rahmani R (2006)
Activation of alpha- and beta-estrogen receptors by persistent
pesticides in reporter cell lines. Life Sci 79:1160–1169
Structural basis of endocrine disruption
40. Paris F, Balaguer P, Terouanne B, Servant N, Lacoste C,
Cravedi JP, Nicolas JC, Sultan C (2002) Phenylphenols,
biphenols, bisphenol-A and 4-tert-octylphenol exhibit alpha and
beta estrogen activities and antiandrogen activity in reporter cell
lines. Mol Cell Endocrinol 193:43–49
41. Bouskine A, Nebout M, Brucker-Davis F, Benahmed M,
Fenichel P (2009) Low doses of bisphenol A promote human
seminoma cell proliferation by activating PKA and PKG via a
membrane G-protein-coupled estrogen receptor. Environ Health
Perspect 117:1053–1058
42. Okada H, Tokunaga T, Liu X, Takayanagi S, Matsushima A,
Shimohigashi Y (2008) Direct evidence revealing structural
elements essential for the high binding ability of bisphenol A
to human estrogen-related receptor-gamma. Environ Health
Perspect 116:32–38
43. Ying GG, Williams B, Kookana R (2002) Environmental fate of
alkylphenols and alkylphenol ethoxylates: a review. Environ Int
28:215–226
44. Kunz PY, Galicia HF, Fent K (2006) Comparison of in vitro and
in vivo estrogenic activity of UV filters in fish. Toxicol Sci
90:349–361
45. Molina-Molina JM, Escande A, Pillon A, Gomez E, Pakdel F,
Cavailles V, Olea N, Ait-Aissa S, Balaguer P (2008) Profiling of
benzophenone derivatives using fish and human estrogen
receptor-specific in vitro bioassays. Toxicol Appl Pharmacol
232:384–395
46. Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, Bonn T,
Engstrom O, Ohman L, Greene GL, Gustafsson JA, Carlquist M
(1997) Molecular basis of agonism and antagonism in the
oestrogen receptor. Nature 389:753–758
47. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard
DA, Greene GL (1998) The structural basis of estrogen receptor/
coactivator recognition and the antagonism of this interaction by
tamoxifen. Cell 95:927–937
48. Barkhem T, Carlsson B, Nilsson Y, Enmark E, Gustafsson J,
Nilsson S (1998) Differential response of estrogen receptor
alpha and estrogen receptor beta to partial estrogen agonists/
antagonists. Mol Pharmacol 54:105–112
49. Manas ES, Xu ZB, Unwalla RJ, Somers WS (2004) Under-
standing the selectivity of genistein for human estrogen
receptor-beta using X-ray crystallography and computational
methods. Structure 12:2197–2207
50. Pike AC, Brzozowski AM, Hubbard RE, Bonn T, Thorsell AG,
Engstrom O, Ljunggren J, Gustafsson JA, Carlquist M (1999)
Structure of the ligand-binding domain of oestrogen receptor
beta in the presence of a partial agonist and a full antagonist.
EMBO J 18:4608–4618
51. Vivat V, Gofflo D, Garcia T, Wurtz JM, Bourguet W, Philibert
D, Gronemeyer H (1997) Sequences in the ligand-binding
domains of the human androgen and progesterone receptors
which determine their distinct ligand identities. J Mol Endo-
crinol 18:147–160
52. Nettles KW, Sun J, Radek JT, Sheng S, Rodriguez AL,
Katzenellenbogen JA, Katzenellenbogen BS, Greene GL (2004)
Allosteric control of ligand selectivity between estrogen recep-
tors alpha and beta: implications for other nuclear receptors. Mol
Cell 13:317–327
53. Nahoum V, Perez E, Germain P, Rodriguez-Barrios F, Manzo F,
Kammerer S, Lemaire G, Hirsch O, Royer CA, Gronemeyer H,
de Lera AR, Bourguet W (2007) Modulators of the structural
dynamics of the retinoid X receptor to reveal receptor function.
Proc Natl Acad Sci USA 104:17323–17328
54. Smith CL, O’Malley BW (2004) Coregulator function: a key to
understanding tissue specificity of selective receptor modulators.
Endocr Rev 25:45–71
55. Giguere V (2002) To ERR in the estrogen pathway. Trends
Endocrinol Metab 13:220–225
56. Bonnelye E, Vanacker JM, Dittmar T, Begue A, Desbiens X,
Denhardt DT, Aubin JE, Laudet V, Fournier B (1997) The
ERR-1 orphan receptor is a transcriptional activator expressed
during bone development. Mol Endocrinol 11:905–916
57. Horard B, Vanacker JM (2003) Estrogen receptor-related
receptors: orphan receptors desperately seeking a ligand. J Mol
Endocrinol 31:349–357
58. Vanacker JM, Pettersson K, Gustafsson JA, Laudet V (1999)
Transcriptional targets shared by estrogen receptor- related
receptors (ERRs) and estrogen receptor (ER) alpha, but not by
ERbeta. EMBO J 18:4270–4279
59. Lu D, Kiriyama Y, Lee KY, Giguere V (2001) Transcriptional
regulation of the estrogen-inducible pS2 breast cancer marker
gene by the ERR family of orphan nuclear receptors. Cancer Res
61:6755–6761
60. Yu S, Wang X, Ng CF, Chen S, Chan FL (2007) ERRgamma
suppresses cell proliferation and tumor growth of androgen-
sensitive and androgen-insensitive prostate cancer cells and its
implication as a therapeutic target for prostate cancer. Cancer
Res 67:4904–4914
61. Ariazi EA, Jordan VC (2006) Estrogen-related receptors as
emerging targets in cancer and metabolic disorders. Curr Top
Med Chem 6:203–215
62. Giguere V (2008) Transcriptional control of energy homeostasis
by the estrogen-related receptors. Endocr Rev 29:677–696
63. Baillie-Hamilton PF (2002) Chemical toxins: a hypothesis to
explain the global obesity epidemic. J Altern Complement Med
8:185–192
64. Li J, Ma M, Wang Z (2009). In vitro profiling of endocrine
disrupting effects of phenols. Toxicol In Vitro (in press)
65. Greschik H, Wurtz JM, Sanglier S, Bourguet W, van Dorsselaer
A, Moras D, Renaud JP (2002) Structural and functional evi-
dence for ligand-independent transcriptional activation by the
estrogen-related receptor 3. Mol Cell 9:303–313
66. Yang C, Chen S (1999) Two organochlorine pesticides, toxa-
phene and chlordane, are antagonists for estrogen-related
receptor alpha-1 orphan receptor. Cancer Res 59:4519–4524
67. Coward P, Lee D, Hull MV, Lehmann JM (2001) 4-Hydroxy-
tamoxifen binds to and deactivates the estrogen-related receptor
gamma. Proc Natl Acad Sci USA 98:8880–8884
68. Tremblay GB, Kunath T, Bergeron D, Lapointe L, Champigny
C, Bader JA, Rossant J, Giguere V (2001) Diethylstilbestrol
regulates trophoblast stem cell differentiation as a ligand of
orphan nuclear receptor ERR beta. Genes Dev 15:833–838
69. Wang J, Fang F, Huang Z, Wang Y, Wong C (2009) Kaempferol
is an estrogen-related receptor alpha and gamma inverse agonist.
FEBS Lett 583:643–647
70. Suetsugi M, Su L, Karlsberg K, Yuan YC, Chen S (2003)
Flavone and isoflavone phytoestrogens are agonists of estrogen-
related receptors. Mol Cancer Res 1:981–991
71. Wang L, Zuercher WJ, Consler TG, Lambert MH, Miller AB,
Orband-Miller LA, McKee DD, Willson TM, Nolte RT (2006)
X-ray crystal structures of the estrogen-related receptor-gamma
ligand binding domain in three functional states reveal the
molecular basis of small molecule regulation. J Biol Chem
281:37773–37781
72. Abad MC, Askari H, O’Neill J, Klinger AL, Milligan C,
Lewandowski F, Springer B, Spurlino J, Rentzeperis D (2008)
Structural determination of estrogen-related receptor gamma in
the presence of phenol derivative compounds. J Steroid Bio-
chem Mol Biol 108:44–54
73. Matsushima A, Kakuta Y, Teramoto T, Koshiba T, Liu X,
Okada H, Tokunaga T, Kawabata S, Kimura M, Shimohigashi Y
A. le Maire et al.
(2007) Structural evidence for endocrine disruptor bisphenol A
binding to human nuclear receptor ERR gamma. J Biochem
142:517–524
74. Matsushima A, Teramoto T, Okada H, Liu X, Tokunaga T,
Kakuta Y, Shimohigashi Y (2008) ERRgamma tethers strongly
bisphenol A and 4-alpha-cumylphenol in an induced-fit manner.
Biochem Biophys Res Commun 373:408–413
75. Kallen J, Schlaeppi JM, Bitsch F, Filipuzzi I, Schilb A, Riou V,
Graham A, Strauss A, Geiser M, Fournier B (2004) Evidence for
ligand-independent transcriptional activation of the human
estrogen-related receptor alpha (ERRalpha): crystal structure of
ERRalpha ligand binding domain in complex with peroxisome
proliferator-activated receptor coactivator-1alpha. J Biol Chem
279:49330–49337
76. Greschik H, Flaig R, Renaud JP, Moras D (2004) Structural
basis for the deactivation of the estrogen-related receptor
gamma by diethylstilbestrol or 4-hydroxytamoxifen and deter-
minants of selectivity. J Biol Chem 279:33639–33646
77. Orans J, Teotico DG, Redinbo MR (2005) The nuclear xeno-
biotic receptor pregnane X receptor: recent insights and new
challenges. Mol Endocrinol 19:2891–2900
78. Biswas A, Mani S, Redinbo MR, Krasowski MD, Li H, Ekins S
(2009) Elucidating the ‘Jekyll and Hyde’ nature of PXR: the
case for discovering antagonists or allosteric antagonists. Pharm
Res 26:1807–1815
79. Moreau A, Vilarem MJ, Maurel P, Pascussi JM (2008) Xeno-
receptors CAR and PXR activation and consequences on lipid
metabolism, glucose homeostasis, and inflammatory response.
Mol Pharm 5:35–41
80. Mikamo E, Harada S, Nishikawa J, Nishihara T (2003) Endo-
crine disruptors induce cytochrome P450 by affecting
transcriptional regulation via pregnane X receptor. Toxicol Appl
Pharmacol 193:66–72
81. Watkins RE, Davis-Searles PR, Lambert MH, Redinbo MR
(2003) Coactivator binding promotes the specific interaction
between ligand and the pregnane X receptor. J Mol Biol
331:815–828
82. Masuyama H, Suwaki N, Tateishi Y, Nakatsukasa H, Segawa T,
Hiramatsu Y (2005) The pregnane X receptor regulates gene
expression in a ligand- and promoter-selective fashion. Mol
Endocrinol 19:1170–1180
83. Benod C, Subra G, Nahoum V, Mallavialle A, Guichou JF, Milhau
J, Roble
´s S, Bourguet W, Pascussi JM, Balaguer P, Chavanieu A
(2008) N-1H-Benzimidazol-5-ylbenzenesulfonamide derivatives
as potent hPXR agonists. Bioorg Med Chem 16:3537–3549
84. Lemaire G, Benod C, Nahoum V, Pillon A, Boussioux AM,
Guichou JF, Subra G, Pascussi JM, Bourguet W, Chavanieu A,
Balaguer P (2007) Discovery of a highly active ligand of human
pregnane x receptor: a case study from pharmacophore model-
ing and virtual screening to ‘in vivo’’ biological activity. Mol
Pharmacol 72:572–581
85. Watkins RE, Wisely GB, Moore LB, Collins JL, Lambert MH,
Williams SP, Willson TM, Kliewer SA, Redinbo MR (2001)
The human nuclear xenobiotic receptor PXR: structural deter-
minants of directed promiscuity. Science 292:2329–2333
86. Xue Y, Chao E, Zuercher WJ, Willson TM, Collins JL, Redinbo
MR (2007) Crystal structure of the PXR-T1317 complex pro-
vides a scaffold to examine the potential for receptor
antagonism. Bioorg Med Chem 15:2156–2166
87. Xue Y, Moore LB, Orans J, Peng L, Bencharit S, Kliewer SA,
Redinbo MR (2007) Crystal structure of the pregnane X
receptor-estradiol complex provides insights into endobiotic
recognition. Mol Endocrinol 21:1028–1038
88. Bertilsson G, Berkenstam A, Blomquist P (2001) Functionally
conserved xenobiotic responsive enhancer in cytochrome P450
3A7. Biochem Biophys Res Commun 280:139–144
89. Blumberg B, Sabbagh W Jr, Juguilon H, Bolado J Jr, van Meter
CM, Ong ES, Evans RM (1998) SXR, a novel steroid and
xenobiotic-sensing nuclear receptor. Genes Dev 12:3195–3205
90. Lehmann JM, McKee DD, Watson MA, Willson TM, Moore JT,
Kliewer SA (1998) The human orphan nuclear receptor PXR is
activated by compounds that regulate CYP3A4 gene expression
and cause drug interactions. J Clin Invest 102:1016–1023
91. Lemaire G, Mnif W, Pascussi JM, Pillon A, Rabenoelina F,
Fenet H, Gomez E, Casellas C, Nicolas JC, Cavailles V,
Duchesne MJ, Balaguer P (2006) Identification of new human
pregnane X receptor ligands among pesticides using a stable
reporter cell system. Toxicol Sci 91:501–509
92. Jones SA, Moore LB, Shenk JL, Wisely GB, Hamilton GA,
McKee DD, Tomkinson NC, LeCluyse EL, Lambert MH,
Willson TM, Kliewer SA, Moore JT (2000) The pregnane X
receptor: a promiscuous xenobiotic receptor that has diverged
during evolution. Mol Endocrinol 14:27–39
93. Synold TW, Dussault I, Forman BM (2001) The orphan nuclear
receptor SXR coordinately regulates drug metabolism and
efflux. Nat Med 7:584–590
94. Watkins RE, Maglich JM, Moore LB, Wisely GB, Noble SM,
Davis-Searles PR, Lambert MH, Kliewer SA, Redinbo MR
(2003) 2.1 A
˚crystal structure of human PXR in complex with
the St. John’s wort compound hyperforin. Biochemistry
42:1430–1438
95. Moore JT, Kliewer SA (2000) Use of the nuclear receptor PXR
to predict drug interactions. Toxicology 153:1–10
96. Wentworth JM, Agostini M, Love J, Schwabe JW, Chatterjee
VK (2000) St John’s wort, a herbal antidepressant, activates the
steroid X receptor. J Endocrinol 166:R11–R16
97. di Masi A, Marinis ED, Ascenzi P, Marino M (2009). Nuclear
receptors CAR and PXR: molecular, functional, and biomedical
aspects. Mol Aspects Med (in press)
98. Jacobs MN, Nolan GT, Hood SR (2005) Lignans, bacteriocides
and organochlorine compounds activate the human pregnane X
receptor (PXR). Toxicol Appl Pharmacol 209:123–133
99. Mnif W, Pascussi JM, Pillon A, Escande A, Bartegi A, Nicolas
JC, Cavailles V, Duchesne MJ, Balaguer P (2007) Estrogens and
antiestrogens activate hPXR. Toxicol Lett 170:19–29
100. Kinani S, Bouchonnet S, Creusot N, Bourcier S, Balaguer P,
Porcher JM, Ait-Aissa S (2010) Multiple endocrine activities in
sediments from small rivers in North of France assessed by the
combined use of in vitro bioassays and chemical analysis.
Environ Pollut 158:74–83
101. Tabb MM, Kholodovych V, Grun F, Zhou C, Welsh WJ,
Blumberg B (2004) Highly chlorinated PCBs inhibit the human
xenobiotic response mediated by the steroid and xenobiotic
receptor (SXR). Environ Health Perspect 112:163–169
102. Pacyniak EK, Cheng X, Cunningham ML, Crofton K, Klaassen
CD, Guo GL (2007) The flame retardants, polybrominated
diphenyl ethers, are pregnane X receptor activators. Toxicol Sci
97:94–102
103. Chrencik JE, Orans J, Moore LB, Xue Y, Peng L, Collins JL,
Wisely GB, Lambert MH, Kliewer SA, Redinbo MR (2005)
Structural disorder in the complex of human pregnane X
receptor and the macrolide antibiotic rifampicin. Mol Endocri-
nol 19:1125–1134
104. Teotico DG, Bischof JJ, Peng L, Kliewer SA, Redinbo MR
(2008) Structural basis of human pregnane X receptor activation
by the hops constituent colupulone. Mol Pharmacol 74:1512–
1520
105. Kretschmer XC, Baldwin WS (2005) CAR and PXR: xenosen-
sors of endocrine disrupters? Chem Biol Interact 155:111–128
106. di Masi A, Marinis ED, Ascenzi P, Marino M (2009) Nuclear
receptors CAR and PXR: molecular, functional, and biomedical
aspects. Mol Aspects Med 30:297–343
Structural basis of endocrine disruption
107. Mangelsdorf DJ, Evans RM (1995) The RXR heterodimers and
orphan receptors. Cell 83:841–850
108. Germain P, Iyer J, Zechel C, Gronemeyer H (2002) Co-regulator
recruitment and the mechanism of retinoic acid receptor syn-
ergy. Nature 415:187–192
109. Chen JY, Clifford J, Zusi C, Starrett J, Tortolani D, Ostrowski J,
Reczek PR, Chambon P, Gronemeyer H (1996) Two distinct
actions of retinoid-receptor ligands. Nature 382:819–822
110. Botling J, Castro DS, Oberg F, Nilsson K, Perlmann T (1997)
Retinoic acid receptor/retinoid X receptor heterodimers can be
activated through both subunits providing a basis for synergistic
transactivation and cellular differentiation. J Biol Chem
272:9443–9449
111. Heyman RA, Mangelsdorf DJ, Dyck JA, Stein RB, Eichele G,
Evans RM, Thaller C (1992) 9-cis retinoic acid is a high affinity
ligand for the retinoid X receptor. Cell 68:397–406
112. de Urquiza AM, Liu S, Sjoberg M, Zetterstrom RH, Griffiths W,
Sjovall J, Perlmann T (2000) Docosahexaenoic acid, a ligand for
the retinoid X receptor in mouse brain. Science 290:2140–2144
113. de Lera AR, Bourguet W, Altucci L, Gronemeyer H (2007)
Design of selective nuclear receptor modulators: RAR and RXR
as a case study. Nat Rev Drug Discov 6:811–820
114. Nakanishi T (2008) Endocrine disruption induced by organotin
compounds; organotins function as a powerful agonist for
nuclear receptors rather than an aromatase inhibitor. J Toxicol
Sci 33:269–276
115. Grun F, Blumberg B (2006) Environmental obesogens:
organotins and endocrine disruption via nuclear receptor sig-
naling. Endocrinology 147:S50–S55
116. Grun F, Blumberg B (2009) Endocrine disrupters as obesogens.
Mol Cell Endocrinol 304:19–29
117. Antizar-Ladislao B (2008) Environmental levels, toxicity and
human exposure to tributyltin (TBT)-contaminated marine
environment: a review. Environ Int 34:292–308
118. Grun F, Watanabe H, Zamanian Z, Maeda L, Arima K, Cubacha
R, Gardiner DM, Kanno J, Iguchi T, Blumberg B (2006)
Endocrine-disrupting organotin compounds are potent inducers
of adipogenesis in vertebrates. Mol Endocrinol 20:2141–2155
119. Kanayama T, Kobayashi N, Mamiya S, Nakanishi T, Nishikawa
J (2005) Organotin compounds promote adipocyte differentia-
tion as agonists of the peroxisome proliferator-activated receptor
gamma/retinoid X receptor pathway. Mol Pharmacol 67:766–
774
120. Castro LF, Lima D, Machado A, Melo C, Hiromori Y, Nishik-
awa J, Nakanishi T, Reis-Henriques MA, Santos MM (2007)
Imposex induction is mediated through the Retinoid X Receptor
signalling pathway in the neogastropod Nucella lapillus. Aquat
Toxicol 85:57–66
121. Nishikawa J, Mamiya S, Kanayama T, Nishikawa T, Shiraishi F,
Horiguchi T (2004) Involvement of the retinoid X receptor in
the development of imposex caused by organotins in gastropods.
Environ Sci Technol 38:6271–6276
122. le Maire A, Grimaldi M, Roecklin D, Dagnino S, Vivat-Hannah
V, Balaguer P, Bourguet W (2009) Activation of RXR-PPAR
heterodimers by organotin environmental endocrine disruptors.
EMBO Rep 10:367–373
123. Harmon MA, Boehm MF, Heyman RA, Mangelsdorf DJ (1995)
Activation of mammalian retinoid X receptors by the insect
growth regulator methoprene. Proc Natl Acad Sci USA
92:6157–6160
124. Li J, Ma M, Wang Z (2008) A two-hybrid yeast assay to
quantify the effects of xenobiotics on retinoid X receptor-med-
iated gene expression. Toxicol Lett 176:198–206
125. Svensson S, Ostberg T, Jacobsson M, Norstrom C, Stefansson
K, Hallen D, Johansson IC, Zachrisson K, Ogg D, Jendeberg L
(2003) Crystal structure of the heterodimeric complex of
LXRalpha and RXRbeta ligand-binding domains in a fully
agonistic conformation. EMBO J 22:4625–4633
126. Egea PF, Mitschler A, Rochel N, Ruff M, Chambon P, Moras D
(2000) Crystal structure of the human RXRalpha ligand-binding
domain bound to its natural ligand: 9-cis retinoic acid. EMBO J
19:2592–2601
127. Pogenberg V, Guichou JF, Vivat-Hannah V, Kammerer S, Perez
E, Germain P, de Lera AR, Gronemeyer H, Royer CA, Bourguet
W (2005) Characterization of the interaction between retinoic
acid receptor/retinoid X receptor (RAR/RXR) heterodimers and
transcriptional coactivators through structural and fluorescence
anisotropy studies. J Biol Chem 280:1625–1633
128. Nettles KW, Bruning JB, Gil G, O’Neill EE, Nowak J, Guo Y,
Kim Y, DeSombre ER, Dilis R, Hanson RN, Joachimiak A,
Greene GL (2007) Structural plasticity in the oestrogen receptor
ligand-binding domain. EMBO Rep 8:563–568
129. Gumy C, Chandsawangbhuwana C, Dzyakanchuk AA,
Kratschmar DV, Baker ME, Odermatt A (2008) Dibutyltin
disrupts glucocorticoid receptor function and impairs gluco-
corticoid-induced suppression of cytokine production. PLoS
One 3:e3545
130. Witorsch RJ (2002) Endocrine disruptors: can biological effects
and environmental risks be predicted? Regul Toxicol Pharmacol
36:118–130
131. de Fur PL (2004) Use and role of invertebrate models in
endocrine disruptor research and testing. ILAR J 45:484–493
132. Roncaglioni A, Benfenati E (2008) In silico-aided prediction of
biological properties of chemicals: oestrogen receptor-mediated
effects. Chem Soc Rev 37:441–450
133. Waller CL, Juma BW, Gray LE Jr, Kelce WR (1996) Three-
dimensional quantitative structure–activity relationships for
androgen receptor ligands. Toxicol Appl Pharmacol 137:219–
227
134. Jacobs MN (2004) In silico tools to aid risk assessment of
endocrine disrupting chemicals. Toxicology 205:43–53
135. Devillers J, Marchand-Geneste N, Carpy A, Porcher JM (2006)
SAR and QSAR modeling of endocrine disruptors. SAR QSAR
Environ Res 17:393–412
136. Fang H, Tong W, Branham WS, Moland CL, Dial SL, Hong H,
Xie Q, Perkins R, Owens W, Sheehan DM (2003) Study of 202
natural, synthetic, and environmental chemicals for binding to
the androgen receptor. Chem Res Toxicol 16:1338–1358
137. Ekins S, Chang C, Mani S, Krasowski MD, Reschly EJ, Iyer M,
Kholodovych V, Ai N, Welsh WJ, Sinz M, Swaan PW, Patel R,
Bachmann K (2007) Human pregnane X receptor antagonists
and agonists define molecular requirements for different binding
sites. Mol Pharmacol 72:592–603
138. D’Ursi P, Salvi E, Fossa P, Milanesi L, Rovida E (2005)
Modelling the interaction of steroid receptors with endocrine
disrupting chemicals. BMC Bioinformatics 6(Suppl 4):S10
139. Celik L, Davey J, Lund D, Schiott B (2008) Exploring inter-
actions of endocrine-disrupting compounds with different
conformations of the human estrogen receptor alpha ligand
binding domain: a molecular docking study. Chem Res Toxicol
21:2195–2206
140. Marchand-Geneste N, Cazaunau M, Carpy AJ, Laguerre M,
Porcher JM, Devillers J (2006) Homology model of the rainbow
trout estrogen receptor (rtERalpha) and docking of endocrine
disrupting chemicals (EDCs). SAR QSAR Environ Res 17:93–
105
141. Lill MA, Winiger F, Vedani A, Ernst B (2005) Impact of
induced fit on ligand binding to the androgen receptor: a mul-
tidimensional QSAR study to predict endocrine-disrupting
effects of environmental chemicals. J Med Chem 48:5666–5674
142. Sivanesan D, Rajnarayanan RV, Doherty J, Pattabiraman N
(2005) In-silico screening using flexible ligand binding pockets:
A. le Maire et al.
a molecular dynamics-based approach. J Comput Aided Mol
Des 19:213–228
143. Zavodszky MI, Lei M, Thorpe MF, Day AR, Kuhn LA (2004)
Modeling correlated main-chain motions in proteins for flexible
molecular recognition. Proteins 57:243–261
144. Bennion BJ, Cosman M, Lightstone FC, Knize MG, Mont-
gomery JL, Bennett LM, Felton JS, Kulp KS (2005) PhIP
carcinogenicity in breast cancer: computational and experi-
mental evidence for competitive interactions with human
estrogen receptor. Chem Res Toxicol 18:1528–1536
145. Kim Y, Koh M, Kim DK, Choi HS, Park SB (2009) Efficient
discovery of selective small molecule agonists of estrogen-
related receptor gamma using combinatorial approach. J Comb
Chem 11:928–937
146. Nose T, Shimohigashi Y (2008) A docking modelling rationally
predicts strong binding of bisphenol A to estrogen-related
receptor gamma. Protein Pept Lett 15:290–296
147. Hartung T, Rovida C (2009) Chemical regulators have over-
reached. Nature 460:1080–1081
Structural basis of endocrine disruption
... The prevalence of receptor activity across the extracts and migrates reflects the ability of PXR to bind structurally diverse compounds. 32 Further, it shows that plastic FCAs not only contain, but also leach PXR agonists, suggesting a potential human and environmental exposure to PXR activating chemicals. ...
Preprint
Full-text available
Plastics constitute a vast array of substances, with over 16 000 known plastic chemicals, including intentionally and non-intentionally added substances. Toxicity and thousands of chemicals are extractable from plastics; however, the extent to which toxicity and chemicals migrate from everyday plastic products remains poorly understood. This study aims to characterize the endocrine and metabolism disrupting activity, as well as chemical composition of migrates from plastic food contact articles (FCAs) from four countries as significant sources of human exposure. Additionally, strategies for prioritization of chemicals were explored. Fourteen plastic FCAs covering seven polymer types with high global market shares were migrated into water and a water-ethanol mixture as food simulants according to European regulations. The migrates were analyzed using reporter gene assays for nuclear receptors relevant to human health and non-target chemical analysis to characterize the chemical composition of the migrates. All FCA migrates interfered with at least two nuclear receptors, predominantly targeting pregnane X receptor. Moreover, peroxisome proliferator receptor gamma was broadly activated by the migrates, though mostly with lower potencies, while estrogenic and antiandrogenic activities were more selectively induced by specific FCAs. Fewer chemicals and less toxicity migrated into water compared to the water-ethanol mixture. The latter exhibited similar toxicity and number of chemicals as methanol extracts of the same FCAs. Novel strategies were employed to address the chemical complexity of FCAs and narrow down the list of potential active chemicals. By comparing the composition of multiple leachates of one sample and using stepwise partial least squares regressions, we successfully reduced the chemical complexity, pinpointed potential endocrine disruptors such as triphenyl phosphate and prioritized chemicals for further identification efforts. This study demonstrates the migration of endocrine and metabolism disrupting chemicals from plastic FCAs into food simulants, rendering a migration of these compounds into food and beverages probable. Abstract Figure
... PXR plays a key role in cellular detoxification and can bind structurally diverse chemicals due to a large ligand-binding pocket with several loops in the ligand-binding domain. 40,41 PXR has important cellular functions beyond its role as xenobiotic sensor, such as energy homeostasis and inflammation. 42−44 A drug-induced dysregulation of PXR is associated with adverse health effects, including hypercholesterolemia and cardiovascular disease. ...
Article
Plastics are complex chemical mixtures of polymers and various intentionally and nonintentionally added substances. Despite the well-established links between certain plastic chemicals (bisphenols and phthalates) and adverse health effects, the composition and toxicity of real-world mixtures of plastic chemicals are not well understood. To assess both, we analyzed the chemicals from 36 plastic food contact articles from five countries using nontarget high-resolution mass spectrometry and reporter-gene assays for four nuclear receptors that represent key components of the endocrine and metabolic system. We found that chemicals activating the pregnane X receptor (PXR), peroxisome proliferator receptor γ (PPARγ), estrogen receptor α (ERα), and inhibiting the androgen receptor (AR) are prevalent in plastic packaging. We detected up to 9936 chemical features in a single product and found that each product had a rather unique chemical fingerprint. To tackle this chemical complexity, we used stepwise partial least-squares regressions and prioritized and tentatively identified the chemical features associated with receptor activity. Our findings demonstrate that most plastic food packaging contains endocrine- and metabolism-disrupting chemicals. Since samples with fewer chemical features induce less toxicity, chemical simplification is key to producing safer plastic packaging.
... Several RXR crystal structures are available in the protein database [16]. Two of them were co-crystallized with triorganotin ligands, triphenyltin hydride (PDB entry code 3KWY [17]) and tributyltin hydride (PDB entry code 3E94 [18]). The structure 3KWY was used because of its structural similarity of its co-crystallized ligand to triphenyltin isoselenocyanate. ...
Article
Full-text available
Several commercially available triorganotin compounds were previously found to function as agonist ligands for nuclear retinoid X receptor (RXR) molecules. Triphenyltin isoselenocyanate (TPT-NCSe), a novel selenium atom containing a derivative of triorganotin origin, was found to represent a new cognate bioactive ligand for RXRs. TPT-NCSe displayed a concentration- and time-dependent decrease in the cell viability in both human breast carcinoma MCF-7 (estrogen receptor positive) and MDA‑MB‑231 (triple negative) cell lines. Reactive oxygen species levels generated in response to TPT-NCSe were significantly higher in both carcinoma cell lines treated with TPT-NCSe when compared to mock-treated samples. Treatment with 500 nM TPT-NCSe caused a decrease in SOD1 and increased SOD2 mRNA in MCF-7 cells. The levels of SOD2 mRNA were more increased following the treatment with TPT-NCSe along with 1 μM all-trans retinoic acid (AtRA) in MCF-7 cells. An increased superoxide dismutase SOD1 and SOD2 mRNA levels were also detected in combination treatment of 500 nM TPT-NCSe and 1 μM AtRA in TPT-NCSe-treated MDA-MB-231 cells. The data have also shown that TPT-NCSe induces apoptosis via a caspase cascade triggered by the mitochondrial apoptotic pathway. TPT-NCSe modulates the expression levels of apoptosis‑related proteins, Annexin A5, Bcl‑2 and BAX family proteins, and finally, it enhances the expression levels of its cognate nuclear receptor subtypes RXRalpha and RXRbeta.
... It is also possible that the environmental chemical is associated with the outcome by way of a nonlinear relationship. Once in the body, some chemicals mimic naturally occurring hormones, prostaglandins, and/or growth factors (le Maire et al., 2010). To evaluate this, we performed mediation analysis using the semi-parametric shape-restricted regression analysis developed in this paper. ...
Article
Full-text available
Often linear regression is used to perform mediation analysis. However, in many instances, the underlying relationships may not be linear, as in the case of placental-fetal hormones and fetal development. Although, the exact functional form of the relationship may be unknown, one may hypothesize the general shape of the relationship. For these reasons, we develop a novel shape-restricted inference-based methodology for conducting mediation analysis. This work is motivated by an application in fetal endocrinology where researchers are interested in understanding the effects of pesticide application on birth weight, with human chorionic gonadotropin (hCG) as the mediator. We assume a practically plausible set of nonlinear effects of hCG on the birth weight and a linear relationship between pesticide exposure and hCG, with both exposure-outcome and exposure-mediator models being linear in the confounding factors. Using the proposed methodology on a population-level prenatal screening program data, with hCG as the mediator, we discovered that, while the natural direct effects suggest a positive association between pesticide application and birth weight, the natural indirect effects were negative.
... PXR plays a key role in cellular detoxification and can bind structurally diverse chemicals due to a large ligand binding pocket with several loops in the ligand binding domain. 40,41 PXR has important cellular functions beyond its role as xenobiotic sensor, such as energy homeostasis and inflammation. [42][43][44] A drug-induced dysregulation of PXR is associated with adverse health effects, including hypercholesterolemia and cardiovascular disease. ...
Preprint
Full-text available
Plastics are complex chemical mixtures of polymers and various intentionally and non-intentionally added substances. Despite the well-established links between certain plastic chemicals (bisphenols, phthalates) and adverse health effects, the composition and toxicity of real-world mixtures of plastic chemicals is not well understood. To assess both, we analyzed the chemicals from 36 plastic food contact articles from five countries using nontarget high-resolution mass spectrometry and reporter gene assays for four nuclear receptors that represent key components of the endocrine and metabolic system. We found that chemicals activating the pregnane X receptor (PXR), peroxisome proliferator receptor gamma (PPARγ), estrogen receptor alpha (ERα), and inhibiting the androgen receptor (AR) are prevalent in plastic packaging. We detected up to 9936 chemical features in a single product and found that each product has a rather unique chemical fingerprint. To tackle this chemical complexity, we used stepwise partial least squares regressions and prioritized and tentatively identified the chemical features associated with receptor activity. Our findings demonstrate that most plastic food packaging contains endocrine and metabolism disrupting chemicals. Since samples with fewer chemical features induced less toxicity, chemical simplification is key to produce safer plastic packaging.
... In the former, NRs are major players, as they are prime targets of EDCs (Fig. 3B). Indeed, many endocrine-disrupting mechanisms have been reported as NRs-mediated [20,44,55,57]. The development of imposex by exposure to organotins has been associated to the retinoid X receptor (RXR) signaling pathway [58][59][60][61][62]; the lipid homeostasis perturbation by obesogens has been linked to the PPARγ signaling pathways [3,[63][64][65][66], as well as to the modulation of other NRs (FXR, liver X receptor [LXR], ecdysone receptor [EcR]) by some EDCs [9]; and the interaction of xenoestrogens with ERs was associated with abnormal reproductive development [1,55]. ...
Article
Plastics constitute a vast array of substances, with over 16 000 known plastic chemicals, including intentionally and non-intentionally added substances. Thousands of chemicals, including toxic ones, are extractable from plastics, however, the extent to which these compounds migrate from everyday products into food or water remains poorly understood. This study aims to characterize the endocrine and metabolism disrupting activity, as well as the chemical composition of migrates from plastic food contact articles (FCAs) from four countries as significant sources of human exposure. Fourteen plastic FCAs covering seven polymer types with high global market shares were migrated into water and a water–ethanol mixture as food simulants according to European regulations. The migrates were analyzed using reporter gene assays for nuclear receptors relevant to human health and non-target chemical analysis to characterize the chemical composition. Chemicals migrating from each FCA interfered with at least two nuclear receptors, predominantly targeting pregnane X receptor (24/28 migrates). Moreover, peroxisome proliferator receptor gamma was activated by 19 out of 28 migrates, though mostly with lower potencies. Estrogenic and antiandrogenic activity was detected in eight and seven migrates, respectively. Fewer chemicals and less toxicity migrated into water compared to the water–ethanol mixture. However, 73 % of the 15 430 extractable chemical features also transferred into food simulants, and the water–ethanol migrates exhibited a similar toxicity prevalence compared to methanol extracts. The chemical complexity differed largely between FCAs, with 8 to 10 631 chemical features migrating into food simulants. Using stepwise partial least squares regressions, we successfully narrowed down the list of potential active chemicals, identified known endocrine disrupting chemicals, such as triphenyl phosphate, and prioritized chemical features for further identification. This study demonstrates the migration of endocrine and metabolism disrupting chemicals from plastic FCAs into food simulants, rendering a migration of these compounds into food and beverages probable.
Article
Endocrine-disrupting chemicals (EDCs) are chemicals that disrupt the normal functioning of endocrine system hormones, leading to a range of adverse health effects in humans and wildlife. Exposure to EDCs is ubiquitous and occurs through contaminated food and water, air, consumer products, and transfer from parents to offspring. Effective regulation has been challenging due to a limited understanding of EDCs’ complex and nonlinear dose-response relationships, as well as difficulty in attributing specific health effects to individual EDC exposures in real-world scenarios. Current EDC policies face limitations in terms of the diversity and complexity of EDCs, the lack of comprehensive testing requirements, and the need for more robust regulatory frameworks that consider cumulative and mixture effects of EDCs. Understanding these aspects is crucial for developing effective and evidence-based EDC policies that can safeguard public health and the environment.
Article
Full-text available
The receptor for 9-cis-retinoic acid, retinoid X receptor (RXR), forms heterodimers with several nuclear receptors, including the receptor for all-trans-retinoic acid, RAR. Previous studies have shown that retinoic acid receptor can be activated in RAR/RXR heterodimers, whereas RXR is believed to be a silent co-factor. In this report we show that efficient growth arrest and differentiation of the human monocytic cell line U-937 require activation of both RAR and RXR. Also, we demonstrate that the allosteric inhibition of RXR is not obligatory and that RXR can be activated in the RAR/RXR heterodimer in the presence of RAR ligands. Remarkably, RXR inhibition by RAR can also be relieved by an RAR antagonist. Moreover, the dose response of RXR agonists differ between RXR homodimers and RAR/RXR heterodimers, indicating that these complexes are pharmacologically distinct. Finally, the AF2 activation domain of both subunits contribute to activation even if only one of the receptors is associated with ligand. Our data emphasize the importance of signaling through both subunits of a heterodimer in the physiological response to retinoids and show that the activity of RXR is dependent on both the identity and the ligand binding state of its partner.
Article
Full-text available
The role of gene expression of the estrogen receptor-a form (ERa) in the regulation of female reproductive behavior was investigated in estrogen receptor knockout (ERKO) mice, deficient specifically for the ERa, but not the ERb, gene. Estrogen- or estrogen- plus progesterone- treated gonadectomized ERKO mice did not show any lordosis re- sponse. Detailed behavioral analysis revealed that ERKO females were also deficient in sexual behavioral interactions preceding the lordosis response. They were extremely rejective toward attempted mounts by stud male mice, which could not show any intromissions. During resident-intruder aggression tests, gonadally intact ERKO females were more aggressive toward female intruder mice than wild- type (WT) mice. Gonadectomy did not influence the levels of aggres- sive behavior, and their genotype differences when mice were tested both before and after gonadectomy. However, when mice were tested after gonadectomy for the first time, very few ERKO mice showed aggression. In contrast to aggression, male-type sexual behavior shown by resident mice toward female intruder mice during aggres- sion tests was not different between ERKO and WT mice and was completely abolished after gonadectomy of the resident mice. Finally, it was also found that ERKO females showed greatly reduced levels of parental behavior toward newborn pups placed in their home cage. These changes in parental behavior were not influenced by gonad- ectomy. ERKO females retrieved significantly fewer numbers of pups with longer latencies compared with wild-type (WT) or heterozygous (HZ) littermates when they were tested as gonadally intact or 20 - 65 days after gonadectomy. In addition, during parental behavior tests, a significantly higher percentage of ERKO mice exhibited infanticide compared with WT and HZ mice, which rarely showed infanticide. Taken together, these findings suggest that ERa gene expression plays a key role in female mice, not only for sexual behavior but also for other interrelated behaviors, such as parental and aggressive behaviors. In addition, persistence of genotype differences in parental and aggressive behavior after gonadectomy indicates that ERa acti- vation during neural developmental processes may also be involved in the regulation of these behaviors. (Endocrinology 139: 5070 -5081, 1998)
Article
Full-text available
Many of the effects of estrogens on the uterus are mediated by ERα, the predominant ER in the mature organ. Because of the poor reproductive capacity of ERβ knockout (BERKO) female mice (small litter size, multiple-resorbed fetuses), the role of uterine ERβ was explored. In the immature uterus, ERα and ERβ are expressed at comparable levels in the epithelium and stroma, and 17β-estradiol (E2) treatment decreases ERβ in the stroma. The immature uterus of untreated BERKO mice exhibits elevated levels of progesterone receptor (PR) and the proliferation-associated protein, Ki-67. It also exhibits exaggerated responsiveness to E2, as indicated by enlargement of the lumen, increase in volume and protein content of uterine secretion, induction of the luminal epithelial secretory protein, complement C3, and its regulatory cytokine IL-1β, and induction of vascular endothelial growth factor and insulin-like growth factor 1 but not its receptor. As expected, E2 increased PR in the stroma and decreased it in the luminal epithelium of wild-type mice. In the BERKO uterus, E2 induced PR in the stroma but did not down-regulate it in the epithelium. Increased cell proliferation and exaggerated response to E2 in BERKO suggest that ERβ plays a role in modulation of the effects of ERα and in addition (or as a consequence of this) has an antiproliferative function in the immature uterus.
Article
Full-text available
Oestrogens are involved in the growth, development and homeostasis of a number of tissues1. The physiological effects of these steroids are mediated by a ligand-inducible nuclear transcription factor, the oestrogen receptor (ER)2. Hormone binding to the ligand-binding domain (LBD) of the ER initiates a series of molecular events culminating in the activation or repression of target genes. Transcriptional regulation arises from the direct interaction of the ER with components of the cellular transcription machinery3,4. Here we report the crystal structures of the LBD of ER in complex with the endogenous oestrogen, 17-oestradiol, and the selective antagonist raloxifene5, at resolutions of 3.1 and 2.6 Å, respectively. The structures provide a molecular basis for the distinctive pharmacophore of the ER and its catholic binding properties. Agonist and antagonist bind at the same site within the core of the LBD but demonstrate different binding modes. In addition, each class of ligand induces a distinct conformation in the transactivation domain of the LBD, providing structural evidence of the mechanism of antagonism.
Article
Many of the effects of estrogens on the uterus are mediated by ER alpha, the predominant ER in the mature organ. Because of the poor reproductive capacity of ER beta knockout (BERKO) female mice (small litter size, multiple-resorbed fetuses), the role of uterine ER beta was explored. In the immature uterus, ER alpha and ER beta are expressed at comparable levels in the epithelium and stroma, and 17 beta-estradiol (E-2) treatment decreases ER beta in the stroma. The immature uterus of untreated BERKO mice exhibits elevated levels of progesterone receptor (PR) and the proliferation-associated protein, Ki-67. It also exhibits exaggerated responsiveness to E-2, as indicated by enlargement of the lumen, increase in volume and protein content of uterine secretion, induction of the luminal epithelial secretory protein, complement C3, and its regulatory cytokine IL-1 beta, and induction of vascular endothelial growth factor and insulin-like growth factor 1 but not its receptor. As expected, E-2 increased PR in the stroma and decreased it in the luminal epithelium of wild-type mice. In the BERKO uterus, E-2 induced PR in the stroma but did not down-regulate it in the epithelium, Increased cell proliferation and exaggerated response to E-2 in BERKO suggest that ER beta plays a role in modulation of the effects of ER alpha and in addition (or as a consequence of this) has an antiproliferative function in the immature uterus.
Article
The rat estrogen receptor (ER) exists as two subtypes, ERα and ERβ, which differ in the C-terminal ligand binding domain and in the N-terminal transactivation domain. In this study we investigated the messenger RNA expression of both ER subtypes in rat tissues by RT-PCR and compared the ligand binding specificity of the ER subtypes. Saturation ligand binding analysis of in vitro synthesized human ERα and rat ERβ protein revealed a single binding component for 16α-iodo-17β-estradiol with high affinity[ dissociation constant (Kd) = 0.1 nm for ERα protein and 0.4 nm for ERβ protein]. Most estrogenic substances or estrogenic antagonists compete with 16α-[125I]iodo-17β-estradiol for binding to both ER subtypes in a very similar preference and degree; that is, diethylstilbestrol > hexestrol > dienestrol > 4-OH-tamoxifen > 17β-estradiol > coumestrol, ICI-164384 > estrone, 17α-estradiol > nafoxidine, moxestrol > clomifene > estriol, 4-OH-estradiol > tamoxifen, 2-OH-estradiol, 5-androstene-3β,17β-diol, genistein for the ERα protein and dienestrol > 4-OH-tamoxifen > diethylstilbestrol > hexestrol > coumestrol, ICI-164384 > 17β-estradiol > estrone, genistein > estriol > nafoxidine, 5-androstene-3β,17β-diol > 17α-estradiol, clomifene, 2-OH-estradiol > 4-OH-estradiol, tamoxifen, moxestrol for the ERβ protein. The rat tissue distribution and/or the relative level of ERα and ERβ expression seems to be quite different, i.e. moderate to high expression in uterus, testis, pituitary, ovary, kidney, epididymis, and adrenal for ERα and prostate, ovary, lung, bladder, brain, uterus, and testis for ERβ. The described differences between the ER subtypes in relative ligand binding affinity and tissue distribution could contribute to the selective action of ER agonists and antagonists in different tissues.
Article
Nuclear receptors (NRs) are ligand-activated transcription factors sharing a common evolutionary history and having similar sequence features at the protein level. Selective ligand(s) for some NRs is not known, therefore these NRs have been named “orphan receptors”. Whenever ligands have been recognized for any of the orphan receptor, it has been categorized and grouped as “adopted” orphan receptor. This group includes the constitutive androstane receptor (CAR) and the pregnane X receptor (PXR). They function as sensors of toxic byproducts derived from endogenous metabolites and of exogenous chemicals, in order to enhance their elimination. This unique function of CAR and PXR sets them apart from the steroid hormone receptors. The broad response profile has established that CAR and PXR are xenobiotic sensors that coordinately regulate xenobiotic clearance in the liver and intestine via induction of genes involved in drug and xenobiotic metabolism. In the past few years, research has revealed new and mostly unsuspected roles for CAR and PXR in modulating hormone, lipid, and energy homeostasis as well as cancer and liver steatosis. The purpose of this review is to highlight the structural and molecular bases of CAR and PXR impact on human health, providing information on mechanisms through which diet, chemical exposure, and environment ultimately impact health and disease.