You are on page 1of 15

Biomedicine & Pharmacotherapy 155 (2022) 113730

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Molecular consequences of the exposure to toxic substances for the


endocrine system of females
Alicja Kowalczyk a, *, Marcjanna Wrzecińska b, Ewa Czerniawska-Piątkowska b,
José Pedro Araújo c, Przemysław Cwynar a
a
Department of Environmental Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, Chełmońskiego 38C, Wrocław, Poland
b
Department of Ruminant Science, West Pomeranian University of Technology, Klemensa Janickiego 29, 71-270 Szczecin, Poland
c
Mountain Research Centre (CIMO), Instituto Politécnico de Viana do Castelo, Rua D. Mendo Afonso, 147, Refóios do Lima, 4990-706 Ponte de Lima, Portugal

A R T I C L E I N F O A B S T R A C T

Keywords: Endocrine-disrupting chemicals (EDCs) are common in the environment and in everyday products such as cos­
Reproduction metics, plastic food packaging, and medicines. These substances are toxic in small doses (even in the order of
Endocrine-disrupting chemicals micrograms) and enter the body through the skin, digestive or respiratory system. Numerous studies confirm the
Toxic substances
negative impact of EDCs on living organisms. They disrupt endocrine functions, contributing to the development
Female fertility
Hormones
of neoplastic and neurological diseases, as well as problems with the circulatory system and reproduction. EDCs
Molecular changes affect humans and animals by modulating epigenetic processes that can lead to disturbances in gene expression
or failure and even death. They also affect steroid hormones by binding to their receptors as well as interfering
with synthesis and secretion of hormones. Prenatal exposure may be related to the impact of EDCs on offspring,

Abbreviations: 1-NP, 1-nitropyrene; 4-CP, 4-cumylphenol; ACTH, adrenocorticotropic hormone; ADI, acceptable daily intake; AMH, Anti-Mullerian Hormone;
AOP, advanced oxidation processes; AR, androgen receptor; Bax, pro-apoptotic protein; Bcl-2, anti-apoptotic protein; BBP, butyl benzyl phthalate; BPAF, bisphenol
AF; BFR, brominated flame retardants; BHC, benzene hexachloride; BPA, bisphenol A; BPC, bisphenol C; BPE, bisphenol E; BPS, biphenyl S; CCA, clear cell
adenocarcinoma; CCM, catalytic ceramic membrane; CEC, contaminants of emerging concern; COCs, cumulus-oocyte complexes; CRH, a corticotropin-releasing
hormone; CTB, cytotrophoblast cells; DBP, di-butylphthalate; DCHP, di-cyclohexylphthalate; DDD, dichlorodiphenyldichloroethane; DDT, dichlorodiphenyltri­
chloroethane; DEHP, di(2-Ethylhexyl)phthalate, diethylhexyl phthalate; DES, diethylstilbestrol; E1, estrone; E2, estradiol; E3, estriol; EE2, 17α-ethinylestradiol; EGF,
epidermal growth factor; EGFR, epidermal growth factor receptor; EM, early menopause; ERE, estrogen-responsive element; ERRγ, estrogen related receptor γ; ERα,
estrogen receptor α; ERβ, estrogen receptor β; EZH2, enhancer of zeste homolog 2; FGF, fibroblast growth factor; FO, forward osmosis; FSH, follicle-stimulating
hormone; GH, growth hormone; GHRN, growth hormone-releasing hormone; GnRH, gonadotropin-releasing hormone; GPER, G protein-coupled estrogen receptor 1;
HAND2, antiproliferative factor; HBB, hexabromobiphenyl; HBCD, hexabromocyclododecane; HFRs, halogenated flame retardants; Hg, mercury; HPA axis, hypo­
thalamus-pituitary gland- adrenal axis; HPG, hypothalamic-pituitary gland-gonadal; HPT, hypothalamus-pituitary gland- thyroid axis; HPTE, hydroxychlor; HSP,
heat shock protein; HSP70, heat shock proteins 70; HSP90, heat shock protein 90; IGF1, IFG2 – insulin-like growth factors 1, 2; LH, luteinizing hormone; LIF,
leukemia inhibitory factor; LOAEL, lowest observed adverse effect level; MAPK, MAP kinase; MBP, monobutyl phthalate; MBzP, mono-benzyl phthalate; MCHP,
mono-cyclohexyl phthalate; MCOMHP, mono (6-COOH-2-methylheptyl) phthalate; MECPP, mono(2-ethyl-5-carboxypentyl) phthalate; MEHHP, mono (2-ethyl-5-
hydroxyhexyl) phthalate; MeHP, mono-2-ethylhexyl phthalate; MEOHP, mono (2-ethyl-5-oxohexyl) phthalate; MEP, mono-ethyl phthalate; MiBP, mono-isobutyl
phthalate; MiNP, mono-isononyl phthalate; miRNA, microRNA; MLL1, mixed-lineage leukaemia 1; MMP, mono-methyl phthalate; MMP, phthalate metabolites mono-
methyl phthalate; MOP, mono-octyl phthalate; MXC, methoxychlor; ncRNA, non-coding RNA; NCX1, Na+/Ca2+ exchanger 1; NOEL, no-observed-effect level; NR,
nuclear receptor; o-PP, ortho-phenylphenol; OCP, organochlorine pesticides; PAC/UF, Activated Carbon with Ultrafiltration; PACs, polycyclic aromatic compounds;
PAEs, phthalates esters; PAHs, polycyclic aromatic hydrocarbons; PBBs, polybrominated biphenyls; PBDEs, polybrominated diphenyl ethers; PC, polycarbonate;
PCBs, polychlorinated biphenyls; PCDDs, polychlorinated dibenzodioxins; PCDFs, polychlorinated dibenzofurans; PCOS, polycystic ovary syndrome; PFAS, Per- and
polyfluoroalkyl substances; PFOA, perfluoroctanoic acid; PFOS, perfluorooctanesulfonic acid; PI3K, phosphoinositide 3-kinase; PMAC1, plasma membrane Ca2+-
ATPase 1; PMS, peroxymonosulfate; POF, premature ovarian failure; POI, primary ovarian insufficiency; POPs, Persistent organic pollutants; PR, progesterone re­
ceptor; PRC2, polycomb 2 repressive complex; PTM, post-translational histone modifications; ROS, reactive oxygen species; SGK1, serine-threonine protein kinase;
SOD, superoxide dismutase; T3, triiodothyronine; T4, thyroxine; TAM, tamoxifen; TBBPA, tetrabromobisphenol A; TCDD, 2,3,7,8-tetrachlorodibenzodioxin; TCS,
triclosan; TRH, thyrotropin-releasing hormone; TRPV5, TRPV6 – transient receptor potential cation channel subfamily V (TRPV) member 5 and 6; TSH, thyroid-
stimulating hormone.
* Correspondence to: Chełmońskiego 38C, 51-630 Wrocław, Poland.
E-mail addresses: alicja.kowalczyk@upwr.edu.pl (A. Kowalczyk), marcjanna.wrzecinska@zut.edu.pl (M. Wrzecińska), ewa.czerniawska-piatkowska@zut.edu.pl
(E. Czerniawska-Piątkowska), pedropi@esa.ipvc.pt (J.P. Araújo), przemyslaw.cwynar@upwr.edu.pl (P. Cwynar).

https://doi.org/10.1016/j.biopha.2022.113730
Received 2 August 2022; Received in revised form 5 September 2022; Accepted 19 September 2022
Available online 21 September 2022
0753-3322/© 2022 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

resulting in effects of these substances on the ovaries and leading to the reduction of fertility through distur­
bances in the function of steroid receptors or problems with steroidogenesis and gametogenesis. Current liter­
ature indicates the need to continue research on the effects of EDCs on the female reproductive system. The aim
of this review was to identify the effects of endocrine-disrupting chemicals on the female reproductive system
and their genetic effects based on recent literature.

1. Background TAM, which were administered for 6 weeks. After exposure, male and
female reproductive organs were dissected and the effect of TAMs on
1.1. Endocrine-disrupting chemicals (EDCs) them was determined. From the second week of exposure, the snails
were shown to lay fewer eggs than those of the control group, and the
Endocrine-disrupting chemicals (EDCs) are a group of chemical males were less active in mating behavior. The fewest eggs were laid by
compounds that are considered pollutants [1,2]. These substances snails in the group in which 600 μg/L of TAM was administered (33),
include polycyclic aromatic hydrocarbons (PHAs), polychlorinated bi­ and the most by snails from the control (270). The snails from the
phenyls (PCBs), pesticides, flame retardants, phthalates, phenols, and control group were characterized by the highest fertility, and in the
toxic metals [3–6]. Among these factors, one of the best studied and experimental groups, the fertility decreased drastically with the con­
known substance influencing the endocrine system is bisphenol A (BPA) centration of TAM. It was shown that the content of estradiol and pro­
[7]. The exposure to mentioned substances occurs through the respira­ gesterone in females dropped drastically to the lowest dose of the
tory and digestive systems or dermal contact [8]. Most of them are preparation from the level for E2 ( ~500 pg/g to ~20 pg/g), and with
highly lipophilic and can accumulate in the adipose tissue [2,9,10]. progesterone (~1100 pg/g to ~0 pg/g) [17]. Livestock is exposed to
Therefore, the accumulation of these substances is frequent and more­ EDCs through the consumption of water or feed, and the accumulation
over, they have a long half-life [11]. It should be noted that BPA has of these substances impairs reproductive performance by disturbing the
approximately 6 h of an average half-life in human tissues, for phtha­ hormonal balance. In ruminants, rumen poisoning is observed due to
lates, the half-life was reported usually as several hours (up to around toxic substances released from plastic [18]. Moreover, the organochlo­
30 h), for the 2,3,7,8-tetrachlorodibenzodioxin (TCDD) this time is rine compounds, such as DDT, can accumulate in products that reach in
oscillating in a period of 5–10 years, and for polybrominated diphenyl fats, such as ruminant’s meat and milk [5]. Ho et al. [2] indicate that
ethers (PBDEs) it is 2–16 years, but for PCBs, it is already a half-life of there are almost 1500 EDCs, and this number may increase by intro­
one to 35 years [12,13]. It has been proven, that EDCs, such as plasti­ ducing new substances to the market.
cizers, pesticides, and persistent organic pollutants (POPs), can get into In current literature, the authors highlight the influence of these
the surface water, as well as the groundwater with industrial and do­ compounds on hormonal signaling and health pathways, fertility, and
mestic wastewaters and with agricultural drains [14]. In the surface and homeostasis of the organism [3–6,9,11]. Exposure to these chemicals
groundwater, the EDCs can pose a threat to water intake points and has long-term effects [5]. It has been proven, that exposure to EDCs
sources of drinking water [13]. In research conducted by Čelić et al. [15] causes health issues such as brain, liver, and lung damage, carcinogen­
the content of selected endocrine-disrupting chemicals was analyzed in esis, and reproductive and endocrine disorders [4].
90 samples taken from 30 different locations, including untreated urban The aim of this review was to identify the effects of endocrine-
and industrial wastewater, water from the Danube, fountains, and water disrupting chemicals on the female reproductive system and their ge­
networks in Serbia. It was shown, that in terms of the concertation of netic effects based on recent literature.
urban sewage, estrogens and their sulphate metabolites were the highest
(average concentrations: estradiol (E2): 7.2 ng/L, estrone (E1): 5.9 ng/L,
1.2. Classification of EDCs
estriol (E3): 4.9 ng/L, E1–3S: 4.4 ng/L, E3–3S: 6.6 ng/L), while BPA
concertation was 9.1 ng/L. Moreover, the most frequently detected EDCs
Endocrine-disrupting chemicals are widespread due to their
in drinking water was bisphenol A (up to 57 % of the frequency), up to
numerous and frequent use throughout the world, so they become a
70 % in surface waters, and up to 84 % in wastewater. Detection of
global issue [1,19]. They can be found in food, textiles, electronics,
estrogens in sewage was up to 30 % for E2, 84 % for E1, and 34 % for E3,
plastics, medical devices, cosmetics, etc. [20]. These substances are
while in surface waters the detection rate reached 60 % for E1 and 27 %
divided into two groups depending on the origin – natural EDCs and
for E3. No estrogens were found in the drinking water. In this research, a
anthropogenic EDCs. Anthropogenic endocrine-disrupting chemicals,
risk assessment was carried out, with E1 and E2 showing the highest risk
also known as synthetic EDCs, have sources of human activity, and on
(18 and 26, respectively), and BPA representing the risk in only one
the other hand natural EDCs can come from plants and animals, such as
sample (RQ 14) in an industrialized area. In the case of estrogenic ac­
animal excrement [1,5]. Table 1 (Table 1) presents sources of selected
tivity, the total estrogenic activity exceeded the threshold of 1 ng/L of
endocrine-disrupting chemicals.
estradiol. The authors suggested that aquatic organisms may be exposed
to the EDCs in the case of chronic exposure [15]. It has been suggested
1.2.1. Persistent organic pollutants (POP)
that EDCs contribute to the death of certain animal species by increasing
These substances are persistent in the environment and highly toxic.
sterility, such as in alligators, and sex change in fish and crustaceans.
Among them, there are polychlorinated industrial chemicals, poly­
Exposure of frogs to organochlorine pesticides, especially herbicides and
brominated biphenyls (PBBs), aldrin, heptachlor, dichlorodiphenyltri­
fungicides, results in hermaphrodism, and male gonadal dysfunction is
chloroethane (DDT), and polycyclic aromatic hydrocarbons (PAH).
observed in alligators. Moreover, reproductive and hormonal disorders
These substances were banned by the Stockholm Convention in 2004
caused by exposure to PCBs and PBDEs have been detected in seals and
[13,26].
whales [16]. In studies conducted on freshwater snails, golden apple
PCBs are aromatic compounds that have been widely used in agri­
snail Pomacea canaliculata (Lamarck) (Caenogastropoda: Ampullariidae)
culture and industry, including for capacitors, paints, plastics, and
investigated the effect of tamoxifen (TAM) on the snail’s hormonal
pesticides. Despite the limitations in the use of PCBs, they are still pre­
balance. TAM is used in the treatment of breast cancer and infertility,
sent in the environment. PCB has neurotoxic, carcinogenic, hepatotoxic,
and its concentration in surface waters is a maximum of 212 ng/L. In the
nephrotoxic, and cytotoxic effects [13,29].
study, the snails were divided into groups according to the TAM doses –
PBDEs are polyhalogenated aromatic compounds used as flame re­
150, 300, and 600 μg/L, respectively, and the control group without
tardants. They are mainly found in textiles, plastics, and electronics

2
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

Table 1 [32].
Sources of selected EDCs.
EDCs Source References 1.2.2. Non-persistant EDCs
The BPA and phthalates are a group of plasticizers that are used in
Dichlorodiphenyltrichloroethane agricultural practices [5,8,21,
(DDT), (pesticides, insecticides, 22] plastic products to obtain shape and flexibility. As opposed to the POPs,
dichlorodiphenyldichloroethane fungicides) non-persistant EDCs are not lipophilic and do not bioaccumulate in the
(DDD), aldrin, dieldrin, endrin, body [29]. BPA is used to produce epoxy resins, polycarbonates, poly­
chlordane, benzene hexachloride sulfones, or as an additive to plasticizers. It is present in electronics,
(BHC), toxaphene
Bisphenol A (BPA), styrene, packaging (food-storage [23–25]
plastic packaging, and ophthalmic lenses. Phthalates are a group of
polycarbonate (PC), phthalates esters materials, plastics), multifunctional industrial chemicals used as plasticizers, e.g. for the
(PAEs), butyl benzyl phthalate (BBP), thermal receipt paper production of polyvinyl chloride, personal care products, solvents, or as
di-butylphthalate (DBP), toxic metals carriers for insecticidal preparations [33].
Brominated flame retardants (BFR) [8,13,25,
polybrominated diphenyl ethers 26]
(PBDEs), bisphenol A, 1.3. Degradation of EDCs
tetrabromobisphenol A (TBBPA)
Triclosan (TCS), chlorine, chlorine medical care (drugs, [1,16,20, The contaminants of emerging concern (CECs) are a category of
dioxide, chloramine, parabens, 17α- biocides, disinfectants, 26,27]
chemicals that have been detected in the water cycle and include in­
ethinylestradiol (EE2), prednisone, personal care products)
fluoxetine, codeine, benzophenones and pharmaceuticals and
dustrial chemicals, endocrine-disrupting chemicals, and persistent
personal care products organic pollutants [16]. The CECs are not easily removed from the water
(PPCPs), sunscreens through physical processes during the treatment of water, such as
Ortho-phenylphenol (o-PP), textile industry, PVC [13,19] filtration, adsorption, coagulation, or precipitation [5,16]. Chemical
polybrominated diphenyl ethers, plastics
methods including advanced oxidation processes (AOP), electro­
phthalates
Polycyclic aromatic hydrocarbons combustion of coal, oil, [28] chemical oxidation, ozonation, and photocatalysis may have a higher
(PAHs), toxic metals, sulfate, nitrate foods, tobacco, wood, EDCs removal efficiency than physical methods but are characterized by
fumes the formation of EDC byproducts [5]. The currently used technologies,
Per- and polyfluoroalkyl substances domestic products, fire- [26] such as chemical coagulation, precipitation, adsorption, and activated
(PFAS), perfluorooctanesulfonic acid fighting foams, surface
(PFOS), perfluorooctanoic acid coating
sludge, for water purification, reduce the concentrations of these sub­
(PFOA) stances, not their complete removal [16]. For this reason, innovative
Alkyphenols, nonylphenols, octylphenol detergents, pesticides, [26] methods to remove EDCs from wared are fooled. A promising approach
industrial surfactant may be carbon adsorption, membrane technology, or nanostructured
Persistent organic pollutants (POPs), industry process, forest [25]
photocatalysts [5]. A study by Chen et al. [34] used a nganese (III)
polychlorinated dibenzodioxins fires, volcanic eruptions
(PCDDs), polychlorinated oxide-based catalytic ceramic membrane (CCM) to activate perox­
dibenzofurans (PCDFs), ymonosulfate (PMS) to degrade various EDCs from drinking water,
polybrominated diphenyl ethers including bisphenol A, bisphenol S, bisphenol F, bisphenol B, bisphenol
(PBDEs) Z, bisphenol D, bisphenol E, bisphenol AP, 17α-ethinylestradiol. These
Polycyclic aromatic chemicals (PACs), cigarettes, internal [25]
cenzopyrene, pyrene, anthracene combustion,
membranes showed the potential to efficiently degrade EDCs in the
miscellaneous burning mg/L to ng/L concentration range. Mentioned studies have achieved
over 90 % efficiency in removing EDC from water, except bisphenol S
(efficiency of about 20 %) and bisphenol AF (about 70 %) [34]. Table 2
[26]. In the European Union, the ban on the use of PBDE was introduced shows the methods of EDC degradation technologies and their
in 2004 and 2008 [13]. effectiveness.
The organochlorine compounds (OCs) have been produced for
various purposes and belong to the group of persistent organic pollut­ 2. Mechanism of EDCs action
ants. They accumulate in organisms. These agents have been widely
used in agriculture as organochlorine pesticides (OCP), insecticides, or The endocrine system consists of glands that secrete hormones and
rodenticides. Among them, dichlorodiphenyltrichloroethane (DDT), regulate hormonal balance [28,40]. The hypothalamus, pituitary, pineal
mirex, dielrin, chlorate, heptachlor, endrin, aldrin, lindane, and endo­ gland, thyroid gland, parathyroid glands, pancreas, and gonads are part
sulfan are distinguished [26]. They were banned by Stockholm of the endocrine system [13,28,40]. Hormones act in genomic and
Convention in 2001. However, the organochlorine compounds are still non-genomic signaling [28]. Steroid hormones are lipophilic, and they
present in the environment (mainly in soil and water) [30]. act through genomic activity [41]. In the genomic mechanism, steroid
The brominated flame retardants (BFRs) include polybrominated hormones are bound to the nuclear receptors (NR), which are located in
diphenyl esters, brominated flame retardants (BFR), hex­ the cytoplasm, and the receptor itself undergoes conformational changes
abromobiphenyl (HBB), and hexabromocyclododecane (HBCD). PBDEs and translocation to the nucleus, where it participates in the regulation
are polyhalogenated aromatic compounds used as flame retardants [26]. of transcription In turn, in a non-genomic mechanism, gene expression is
They are mainly found in textiles, plastics, and electronics. In the Eu­ modulated by intracellular signaling pathways, and mainly hydrophilic
ropean Union, the ban on the use of PBDE was introduced in 2004 and hormones (e.g. catecholamine) bind to cell surface receptors [28].
2008 [13]. BFRs are used in a variety of products such as furniture, Nuclear receptors are a group of proteins that are transcription fac­
textiles, carpets, electronics molding, automotive components, building tors regulated by ligands, mainly estrogen and progesterone [13,42]. By
materials, insulators, etc. to improve their fire resistance. Moreover, sequencing the human genome, 48 nuclear receptors were revealed.
these substances are common in the environment and show the ability to Among the NRs, estrogen (ER), progesterone (PR), and androgen re­
accumulate [31]. ceptors (AR) are distinguished [41]. There are estrogen receptor α
Dioxins and furans are divided into groups that include poly­ (ERα), estrogen receptor β (ERβ), and the membrane receptor coupled
chlorinated dibenzo-ρ-dioxins (PCDDs), polychlorinated dibenzofurans with the G protein (GPER, previously termed as a G protein-coupled
(PCDFs), and dioxin-like polychlorinated biphenyls (dl-PCB). TCDD is receptor 30, GPR30), which participates in the regulation of
the most potent dioxin of PCDD. The main sources of dioxins are vol­ non-genomic mechanisms [6,10,13,43,44]. The ERα and ERβ are re­
canic eruptions, waste incineration, including tobacco, and industry ceptors for the natural estrogen 17β-estradiol, that are involved in the

3
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

Table 2
EDCs degradation methods and their effectiveness.
Method Type of EDCs Effectiveness References

Adsorption Activated carbon BPA 100 % [5]


Activated Carbon with Ultrafiltration 17α-ethynylestradiol 80 % [35]
(PAC/ UF)
Nano catalyst ZnO nanoparticles BPA 97 % [36]
TiO2 nanosized Phenol 78 % [37]
Advanced UV BPA 48 % [38]
oxidative Ozonation cyclodien pesticides, dichlorodiphenyltrichloroethanes (DDTs), phthalates, poly- > 80 % [39]
processes aromatic hydrocarbons (PAHs) polychlorinated biphenyls (PCBs) ~80 %
di-cyclohexylphthalate (DCHP) ~40 %
naphthalene, phenanthrene, di-n-butylphthalate (DBP)
Membrane Forward osmosis (FO) Triclosan 79 % [35]
Catalytic ceramic membrane (CCM) to bisphenol A, bisphenol F, bisphenol B, bisphenol Z, bisphenol D, bisphenol E, > 90 % [34]
activate peroxymonosulfate (PMS) bishenol AP, 17α-ethinylestradiol ~20 %
bisphenol S ~70 %
bisphenol AF

growth and maintenance of a variety of tissues such as the mammary shaped with a local minimum or maximum, respectively, or even
gland, uterus, and bone [45]. The main targets of EDCs are the ERα and biphasic with two minima or maxima [51]. A feature of EDCs is the
ERβ, the androgen receptor, which is present in the nucleus (ER) or ability to create NMDRCs [52]. Non-monotonic dose-response curves
cytoplasm (AR). Binding to EDCs results in conformational changes that were noted for EDCs such as pesticides, polychlorinated biphenyls, di­
result in the dissociation of the chaperone proteins and the formation of oxins, bisphenol A, and phthalates [53]. NMDRCs have been demon­
binding to specific response elements (ERs) in DNA, which has an effect strated for various EDCs in a variety of biological systems, including cell
on the modulation of gene transcription [45]. Recently, the culture, whole organ culture, laboratory animals, and human pop­
estrogen-related receptor γ (ERRγ) has been reported as a target of EDCs. ulations [52,54]. The mechanism of NMDRC production, which is well
The ERRγ is part of the ERR family of receptors, on which ERRα and known, is cytotoxicity, whereby increasing doses of substances induce a
ERRβ are also distinguished [45,46]. The estrogen-related receptor γ is toxic effect. There is also evidence that high doses of substances are able
an orphan nuclear receptor that plays an important role in the meta­ to inhibit cell proliferation, which has the opposite effect on cytotox­
bolism of mitochondria (mainly lipids, and pyruvate), and acts as a icity. Another mechanism of NMDRC is the influence of hormone con­
constitutive activator of transcription. BPA was found to bind strongly to centration on the number of receptors. If the number of receptors that
ERRγ. The action of BPA on this receptor is to incorporate BPA into the arise does not coincide with the number of degrading receptors, a
ligand-binding pocket by ERRγ, and the structure of bisphenol A with decrease in the response associated with an increase in hormone con­
two phenol-hydroxyl groups is the basis for the formation of hydrogen centrations is observed [52]. One of the EDCs capable of producing
bonds with the receptor. 17 different BPA derivatives have been found to NMDRC is bisphenol A [55]. It is a widely researched compound with
be able to bind to the estrogen-related receptor γ [47]. The ERRγ activity high toxicological priority. In the article, Vandenberg et al. [52]
can be modulated by phenols, phytoestrogens, and pesticides. The collected 109 in vitro BPA studies published during 2007–2013. Studies
bisphenol E (BPE) acts on the transcriptional activity of ERRγ, while the with single doses and those that did not provide information on the
diethylstilbestrol (DES) is an inverse agonist of ERRγ, that reduces the shape of the dose-response curve and did not show an effect of BPA were
action of the estrogen-related receptor γ. In the research on the stable excluded from the analyzes. Of the analyzed experiments, NMDRC was
cell line (HG5LN), the EC50 (half maximal effective concertation) was obtained in 23.6 % (59) of the studies and at least one endpoint in over
established for over 30 environmental substances, which were ligands to 30 % of the analyzes [52].
ERRγ. It was determined, that BPA is a fairy weak agonist of estrogen Endocrine-disrupting chemicals act on the endocrine system in
receptors (EC50 approximately 0.5 µM), however, another biphenol – different ways. They can inhibit hormones binding to receptors or pre­
bisphenol C (BPC), hydroxychlor (HPTE) show EC50 values at the level vent hormone signaling [56]. These substances can also block or activate
of 30 nM and 50 nM [46]. The steroid hormone receptors are associated natural hormone synthesis and degradation of natural hormones [16].
with the heat shock proteins (HSP) – mainly HSP70 and HSP90, which These compounds have been shown to adversely affect the endocrine
regulate their functions. Activation of GPER occurs by binding to the system even at low doses (for example: in Danio rerio, the toxicity of BPA
estrogen receptor, which forms the complex, dimerizes, and then the for the development of fish is at the level of 50 μM [57]) by acting on
chaperones are disconnected. Stimulation of adenylate cyclase occurs, various receptors, including steroid sex hormone receptors, such as es­
and the release of membrane-bound epidermal growth factor (EGF) trogen receptors, androgen receptors, and thyroid hormone receptors
occurs. The complex then translocates to the nucleus and binds to a [56,58]. Table 3 shows the acceptable daily intake (ADI), no observed
specific estrogen-responsive element (ERE) that is located in the pro­ effect levels (NOEL), and the lowest observed adverse effect level
moter region of the target gene. In the cell nucleus, the estrogen-ER (LOAEL) of selected EDCs in terms of their influence on reproduction.
complex is involved in the transcription of target genes [13,48,49]. As an endocrine-disrupting chemical, bisphenol A, may act mainly
Deletions in GPER cause insulin resistance and hypertension. GPER1 through estrogen receptors [58,66,67]. BPA is an estrogen antagonist
agonists have been shown to activate insulin release from the pancreas and exhibits non-nuclear activation of the signal mediated by the ERα.
and vasodilation. Moreover, GPER agonists prevent inflammation. EDCs BPA is also antagonistic to estradiol and causes ERβ signal limitation
have been shown to bind to GPER via an affinity for estrogens, which with its nuclear receptor [6,68]. BPA has been shown to induce estro­
adversely affects physiology. By activating GPER, phthalates contribute genic activity and interfere with the endocrine system [6,42]. In addi­
to the development of breast and cervical cancer [48,50]. tion, it induces DNA damage and cell damage, e.g., lymphocytes.
Non-monotonic dose-response curves (NMDRCs) have been devel­ Normal estrogen signaling is necessary for the maintenance of immune
oped in research into the treatment of cells, animal, and human models function. Moreover, lymphocytes have steroidogenic abilities and ex­
with EDCs. It is a plot of the biological response as a function of a specific press ERα and ERβ. Therefore, it is believed that BPA may have a
EDC, the slope of which changes at some point in the dose-response negative effect on the immune functions and affect the activity of lym­
curve. Such dose-response curves can be U-shaped or inverted U- phocytes [69]. BPA reduces the activity of superoxide dismutase (SOD),

4
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

Table 3
ADI, NOEL, LOEAL of selected EDCs.
EDCs Doses

Acceptable Daily Intake (ADI) No Observed Effect Level (NOEL) Lowest Observed Adverse Effect Level (LOAEL) Species References

E2 0.05 mg/kg bw/day 10 mg/kg bw/day adult human [59]


methyl- and ethylparaben 10 mg/kg bw/day 1000 mg/kg bw/day adult human [60]
Parabens 0.3 mg/kg bw/day 1000 mg/kg bw/day 100 mg/kg bw/day for rats infants [60,61]
BBP 0.5 mg/kg bw/day 50 mg/kg bw per day 250 mg/kg bw/day rats [62,63]
BPA 4–5 μg/kg bw/d 5 mg/kg bw/day 120 mg/kg bw/day mice [64]
DBP 50 µg/kg bw/day 50 mg/kg bw/day 2 mg/kg bw/day rats [62]
DEHP 0.05 mg/kg bw/day 5 mg/kg bw/day 3 mg/kg bw/day rats [65]

resulting in an increase in reactive oxygen species (ROS) in cells, which Epigenetic changes caused by exposure to EDCs have a direct impact on
can contribute to DNA damage [6,42]. the emergence of diseases and dysfunctions [75]. Epigenetic mecha­
nisms include DNA methylation, post-translational histone modifica­
tions (PTM), chromatin modification, and non-coding RNAs (ncRNA)
2.1. Other key characteristics of endocrine-disrupting chemicals that regulate gene expression and are involved in homeostasis and cell
development [76]. In the case of DNA methylation within the promoter
La Merrill et al. [70]point to other key features of endocrine dis­ region, transcription is lost. It has been shown that these changes may
rupting chemicals. EDCs are capable of interacting with or directly affect subsequent generations [74]. During DNA methylation, modifi­
activating hormone receptors, which may have negative health conse­ cation of the cytosine residue in CpG dinucleotides occurs, resulting in
quences. These substances are capable of antagonizing hormone re­ difficulties in gene transcription and consequently in its silencing [48,
ceptors, which is associated with inhibiting or blocking the action of 58,77]. Disturbances in methylation can lead to retarded growth and
endogenous hormones, acting as their antagonists. EDCs can modulate development of organism [48]. In turn, modifications of chromatin
the expression of the hormone receptor (s) by degrading them or through changes of histone proteins in the DNA structure may disturb
reducing their expression. These substances can alter the signal trans­ the reading of the transcription regulation. Long non-coding RNA shows
duction in hormone-responsive cells, modifying the progressive intra­ the ability to interact and modify chromatin, which causes its further
cellular changes, which may then weaken or intensify the effects of modification [58]. Another example of an epigenetic mechanism is
hormones. In addition, hormones can act by modifying epigenetic pro­ imprinting, which is the methylation of genes or histones of an allele
cesses, and EDCs can disrupt the effects of hormones and their ability to inherited from a parent [7,75].
induce epigenetic changes [54,70]. Endocrine-disrupting chemicals can Accurate hormone signaling is a key factor that influences the
also modulate hormone synthesis by, for example, limiting the uptake of development of the body and its reproductive capacity [78]. There is a
compounds necessary for the hormone synthesis process. These sub­ growing body of research pointing to the effects of EDCs on reproductive
stances can alter the transport of hormones across cell membranes pathologies [79]. Disorders caused by EDCs are associated with poly­
through selective and passive transport processes or alter the distribu­ cyclic ovarian syndromes, uterine fibrosis, endometriosis, miscarriages,
tion of hormones or circulating hormone levels by altering the breast, and prostate cancer [2,5,10,58]. Exposure to EDCs through skin
bioavailability of the hormones. Another feature of EDCs is changing the contact, inhalation, and gastrointestinal tract increases the risk of breast
rate of inactivation, degradation, or removal of hormones, which mod­ cancer [80]. The mammary gland is susceptible to substances that
ulates their concentration, as well as changes their metabolism in the disrupt the endocrine system. Bisphenol A is reported to be a key sub­
body. A final characteristic of EDCs to be reported is the ability to alter stance that interferes with the development of the mammary gland and
the total number or position of cells in hormone-producing or induces the neoplastic process [81]. In rats, the effects of BPA on the
hormone-responsive tissues by interfering with or promoting cell dif­ mammary gland were analyzed [82]. A group of 10 rats was adminis­
ferentiation, proliferation, or apoptosis [54,70]. The key characteristics tered a dose of 5 mg/kg BPA for 8 weeks, and then the mammary glands
of endocrine-disrupting chemicals are shown in the Fig. 1 (Fig. 1). were dissected, the control was group of 10 animals without treatment
and 10 rats, were administered 1 ml of corn oil for 8 weeks. BPA was
3. Endocrine aspects of EDCs shown to induce structural changes in the mammary gland of rats - the
number and size of ducts and acini increased, and epithelial nuclei
It has been proven that EDCs can disrupt the action of steroid hor­ showed darker color and different shape compared to control. Moreover,
mones, such as estrogens, and androgens, and can lead to disorders in it was shown that the content of collagen fibers increased within the
the reproduction [28]. The hypothalamus and the anterior pituitary connective tissue stroma, and infiltration of inflammatory cells was
gland play a crucial role in regulating the secretion of luteinizing hor­ observed. This study showed that the substance could induce the
mone (LH) and follicle-stimulating hormone (FSH), which are involved development of breast cancer mainly by modulating the cell prolifera­
in modulating the function of the ovaries, uterus, and the secretion of tion process [82]. The effects of BPA and 4-cumylphenol (4-CP) on the
reproductive hormones in females [29]. Puberty is an important factor viability of the MCF-7 human breast cancer cell line in vitro were
for achieving a normal reproductive performance [71]. According to investigated. The results of the cell viability assay showed that both test
Franssen et al. [9] late puberty in females, metabolic disorders, as well substances in the range from 10-9 to 10-5 M stimulated cell proliferation
as impaired fertility, may result from prenatal and postnatal exposure to in a non-monotonic dose-response manner. At the same time, the cell
EDCs. The process of puberty is influenced by the action of the cycle progressed from the G0/G1 phase to the S and G2/M phase, and
hypothalamus-pituitary gland-gonadal (HPG) axis, and disturbances the expression levels of ERα mRNA and the anti-apoptotic protein
within this axis translate into the puberty [9,71]. Moreover, (Bcl-2) increased. At high doses (10-4 M), the substances showed an
endocrine-disrupting chemicals prolong puberty [72,73]. antiproliferative effect in MCF-7 cells by inducing cell apoptosis and
EDCs are able to induce diseases and fertility disorders due to their blocking the cell cycle in the G0/G1 phase, the levels of Bcl-2 mRNA
genotoxic activity, as well as by modulating epigenetic processes in expression decreased, while mRNA of pro-apoptotic protein (Bax)
which steroid hormones, such as sex hormones, are involved [48]. This increased [83]. Peremiquel-Trillas et al. [84] conducted a population
leads to changes in DNA, which may directly lead to death or mutation study involving 1513 cases of breast cancer patients who were
[42]. These changes may be passed on to the next generations [42,74].

5
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

Fig. 1. Key characteristics of EDCs (


Source: [54,70]).

professionally exposed to alkylphenol compounds. It was found that a slightly increased risk of breast cancer (OR = 1.23; 95 % CI =
27.2 % (412 cases) had occupational exposure to alkylphenol com­ 1.01–1.48) compared to the non-exposed person. The highest associa­
pounds, and people with occupational exposure to these compounds tion was found in subjects with frequent exposure and high exposure
showed a greater risk of breast cancer than patients not exposed to these intensity (OR = 1.25; 95 % CI = 1.01–1.55). The results suggest that
compounds. People professionally exposed to these compounds showed there is an association between the risk of breast cancer and alkylphenol

6
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

compounds [84]. Moreover, it has been shown that women in Greenland the hypothalamus and pituitary gland [28,68,90]. Among
who had high levels of PFOS and PFOA, and PFAS in their blood serum endocrine-disrupting chemicals, polychlorinated biphenyls (PCBs) are
were associated with an increased risk of breast cancer. PFOS is asso­ considered to be the compounds that have the greatest impact on the
ciated with tumors with the presence of estrogen receptors [80]. In development of the brain as well as its functions [94].
another study, the presence of phthalates in the urine of women was Endocrine-disrupting chemicals can act by genomic or non-genomic
correlated with the risk of breast cancer, and out of 140 modifications of the pathways of nuclear receptors, non-steroidal re­
phthalate-responsive genes, 105 genes were obtained that mutated in ceptors, and ion channels, and may be associated with abnormal growth,
the tissues of the breast tumor. Phthalates induce mutations in genes inflammation, and epigenetic control [58,68]. Graceli et al. [68]
involved in the biosynthesis of steroid hormones. Among the phthalates distinguished bisphenol A, phthalates, polychlorinated biphenyls, pol­
tested, DBP (Odds Ratio = 1.5, 95 % Confidence Interval; 1.06–2.11, ybrominated diphenyl ethers, polybrominated biphenyls, dichlor­
p = 0.002) and DEHP (Odds ratio = 2.97, 95 % Confidence Interval; odiphenyltrichloroethane, and atrazine as the EDCs with the greatest
1.18–7.47, p = 0.005) showed the highest affinity for breast cancer influence on the functioning of the hypothalamus and pituitary gland.
[85]. The reproductive system is particularly sensitive to hormonal ab­ These toxic substances have been proven to inhibit steroidogenesis and
normalities during development, as it reacts to disturbances in gene interfere with the activity of TSH and FSH receptors [68]. Moreover,
expression that lead to changes in tissues and structure within this sys­ BPA has been shown to influence the release of kisspeptin neuropeptides
tem [58]. EDCs cause disturbances in the oestrus cycle length and cause from the hypothalamus and the secretion of GnRH, leading to a change
germ cell nest breakdown [86]. In females, EDCs have been detected in in the endocrine function of the hypothalamus [95]. As was noticed by
follicular fluid, amniotic fluid, milk, and blood serum [79,87]. It has Pinilla et al. [82], the GnRH neurons are considered a major hierarchical
been proven, that bisphenol A retards the action of ovaries, uterus, element of this system, and GnRH operates as the final output signal for
steroidogenesis, or event disrupts follicular formation [73]. Further­ the hypothalamic regulation of the downstream elements of the HPG
more, the negative influence of BPA is indicated on the level of estro­ axis. Adequate pulsatile secretion of GnRH is mandatory for proper
gens, the quality of oocytes, the formation of blastocysts, and attainment and maintenance of reproductive function [82].
implantation failure is indicated [73,88]. In addition, there is also a
direct negative effect of BPA on uterine morphology, endometrial 3.2. Ovary
health, and the number and growth of ovarian follicles [88].
Van Duursen et al. [89] report in their article that in the case of fe­ The life of the female reproductive lifespan is determined by the
male reproduction there is still a lack of reports and studies showing the amount of the primordial follicle pool, which is established in the pre­
effects of exposure to EDCs. natal life [78]. EDCs negatively affect the processes that take place in the
ovary, such as primary follicles, follicle formation, follicle growth, and
3.1. Hypothalamus activity, or steroidogenic activity. In this way, endocrine-disrupting
chemicals contribute to the onset of temporary or permanent infer­
The hypothalamus is involved in the process of feeding, and main­ tility in females [58,73]. EDCs contribute to the induction of ovarian cell
taining body weight, fluid balance, blood pressure, and body tempera­ apoptosis [86]. In the research conducted by Shi et al. [73], the effect of
ture [90]. The main role in the mammalian endocrine system is played BPA, BPE, and BPS on follicular development was examined. The ex­
by the hypothalamic and pituitary glands [68]. These crucial centers of amination was carried out on pregnant mice, which were divided into
the endocrine system are involved in maintaining homeostasis, and groups – the control group, and tested groups based on doses of EDCs –
modulating the body’s functions responsible for metabolism, repro­ 0.5, 20, or 50 μg/kg/day doses of either BPA, or BPE, or BPS. The ani­
duction, growth, lactation, or water balance. Moreover, the mals were treated with the appropriate doses on the day 11 of gestation.
hypothalamic-pituitary gland-thyroid (HPT) axis, the During the research, the influence of prenatal exposure to BPA, BPE, and
hypothalamic-pituitary gland-gonadal (HPG) axis, and the BPS on the ovaries was analyzed. Prenatal exposure to BPA and BPE (0.5
hypothalamic-pituitary gland-adrenal (HPA) axis are distinguished [28, and 50 μg/kg/day) and BPS (0.5, 20 μg/kg/day) resulted in significantly
68,90,91]. more remain in germ cells nest compared to the control group. More­
In the HPT axis, the hypothalamus is responsible for the secretion of over, BPA (0.5 μg/kg/day) and all doses of BPE and BPS exposure
the thyrotropin-releasing hormone (TRH), which signals the release of significantly (P < ).01) reduced primary follicles almost doubled from
the thyroid-stimulating hormone (TSH) from the pituitary gland. [91]. 11 % for the control group to about 5 % for the test groups. In all study
In turn, TSH participates in the regulation of thyroid metabolism and the groups administered bisphenols, a smaller number of primary follicles
stimulation of the thyroxine (T4) and triiodothyronine (T3) hormones was obtained, and in the case of secondary follicles, only BPA in any
influencing metabolism [68]. The HPA axis is involved in the secretion doses induced a significantly (P < 0.01) lower number of follicles from
of corticotropin-releasing hormone (CRH), which activates the adreno­ 2.5 % in the control group to about 0.8 for 0.5 µg BPA /kg/day and 50 µg
corticotropic hormone (ACTH) from the pituitary gland, which acts on BPA/kg/day and about 0.5 % for 20 μg/kg/day BPA [73]. The influence
adrenal glands to stimulate cortisol production. The adrenal glands also of pesticides on the low weight of the ovaries of impaired ovaries and
secrete catecholamines, epinephrine, and norepinephrine, to maintain pesticides on the rate of follicular arthritis have been described [96].
homeostasis and protect against stress [90]. ACTH is also involved in the EDCs, and in particular phthalates, have been shown to reduce the pri­
secretion of aldosterone, influencing the water balance [68]. In turn, on mary antral follicles [78].
the hypothalamus-pituitary gland-gonadal axis, the arcuate nucleus of Aging of the ovaries is a natural process that leads to a decrease in the
the hypothalamus secretes growth hormone-releasing hormone (GHRN) quality and quantity of oocytes in the follicles. This process reduces
peptides and gonadotropin-releasing hormone (GnRH) that stimulate reproductive and endocrine capacity [97]. Ovarian aging can be influ­
the secretion of growth hormone (GH) and gonadotropins. GH stimu­ enced by genetic, autoimmune, and also environmental factors, such as
lates body growth, and LH and FSH stimulate gametogenesis as well as endocrine-disrupting chemicals [98,99]. Exposure to EDCs has been
the synthesis of sex hormones [68]. LH stimulates progesterone and FSH shown to affect ovarian dysfunction and aging, leading to infertility or
stimulates follicular growth by binding to G protein-coupled receptors premature ovarian failure (POF). The mechanisms of action of EDCs may
on granular cells and stimulating aromatase expression. Aromatase is an be the induction of epigenetic changes, hormonal disorders, or inter­
enzyme produced by granular cells [28,92]. The HPG axis is involved in action with hormone receptors [99].
steroidogenesis and folliculogenesis. Exposure to BPA can lead to dys­ Exposure to EDCs can also lead to early menopause (EM), polycystic
functions on this axis and can lead to disorders in the fertility [93]. ovary syndrome (PCOS), and primary ovarian insufficiency (POI), or
Reports are confirming the influence of EDCs on the functioning of even can reduce fertility and disrupt oogenesis [78,96]. The research

7
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

carried out on a group of 30 women diagnosed with primary ovarian pathology or even endometrial cancer [10,105]. These hormones are
insufficiency was tested for the analysis of serum phthalates [100]. The also required to stimulate endometrial proliferation. Moreover, dieth­
mean concentration of monobutyl phthalate (MBP) in the serum was ylstilbestrol and 17α-ethinylestradiol (EE2) act like estrogen via estro­
statistically significantly higher in the POI group than in the control gen receptors and are associated with the induction of epigenetic
group (8.45 ± 4.2 vs. 5.0 ± 3.47 ng/ml, p < 0.001). Other concentra­ changes in the uterus [44,58]. In the presence of estrogen, uterine cells
tions of phthalate metabolites in serum, such as mono-ethyl phthalate produce fibroblast growth factor (FGF), which activates endometrial
(MEP), mono- (2ethylhexyl) phthalate (MeHP), mono-benzyl phthalate FGF receptor signaling and results in endometrial cells. In turn, in the
(MBzP), mono-methyl phthalate (MMP), mono-buthyl phthalate (MBP) presence of progesterone, the expression of the antiproliferative factor
and BPA concentrations were higher in patients with POI, but the au­ (HAND2) occurs which inhibits FGF expression [105]. The uncontrolled
thors found no significant differences. Among the examined EDCs, the proliferation of the uterine lining leads to the risk of endometrial cancer
most important was MBP, the authors of which indicate the main factor [106]. Moreover, exposure to EDCs may be associated with the risk of
in the development of POI [100]. In the research conducted by Hu et al. developing endometrial cancer (EC) [106]. Leung et al. [107] in a study
[87] in women with polycystic ovarian syndrome, higher levels of BPA in rats showed that exposure to 25 and 250 μg/kg BW/d) BPA affected
were found in the follicular fluid and blood serum of patients compared the oestrus cycle as the well as uterine physiology, including the growth
to the control group of healthy women. In addition, neonatal exposure of of endometrial cancer. Polycyclic aromatic compounds (PACs) such as
rats to BPA is reported to influence the development of PCOS in the adult benzopyrene and halogenated flame retardants (HFRs) have also been
life of animals and disturbances in the ovarian and vaginal morphology shown to contribute to uterine carcinogenesis [106].
such as cyst formation [87,93]. Prenatal exposure to BPA and TCS is The uterus is very sensitive to the effects of estrogen, therefore es­
associated with a decrease in follicle stimulating hormone concentration trogenic compounds such as BPA may have an adverse effect on the
and disturbed estrogen protein expression in ovarian granule cells, functioning of the uterus as well as on pregnancy [105]. Studies con­
which leads to a reduction in female fertility [86,93]. ducted in mice have shown that bisphenol A causes decreased lutei­
The di (2-Ethylhexyl) phthalate (DEHP; diethylhexyl phthalate) is an nizing hormone as well as abnormal oestrus cycle and reduced fertility.
endocrine disruptor that affects the functioning of the ovaries. In a study A study by Neff et al. [105] found that chronic exposure to low doses of
by Brehm et al. [101] in pregnant mice treated with DEHP: BPA (60 µg/kg/d) resulted in abnormal endometrial proliferation.
20 µg/kg/day, 200 µg/kg/day, 500 mg/kg/day and 750 mg/kg/day, Moreover, BPA has been shown to increase FGF production, leading to
the effect of DEHP on 3 generations (F1, F2, F3) was analyzed. Exposure activation of the FGF receptor in the uterus of non-pregnant animals. At
to DEHP has been shown to have long-term health effects on posterity the same time, a decrease in HAND2 expression was obtained, which
(F2 and F3). Additionally, DEHP accelerates folliculogenesis and re­ corresponded to an increase in FGF production. The authors concluded
duces the number of follicles, leading to premature reproductive aging that such pathway activation is the cause of uterine hyperplasia [105].
[101,102]. In turn, exposure to DEHP and phthalates induces cyst for­ In addition, disturbances in the action of progesterone and estradiol
mation in the ovaries. The dose of 200 µg/kg/day decreased the body contribute to the development of uterine fibroids [108]. Leiomyomas
weight of the F3 mice. Prenatal exposure to DEHP resulted in a decrease have their pathogenesis in EDC-induced female hormone imbalance
in ovarian weight in the studied generations, and a higher uterine [109]. The research conducted by Zhang et al. [109] investigated the
weight was recorded in the F1 generation, which may have been due to effect of exposure to phthalates, toxic metals, and equol on the occur­
an increase in estradiol levels [101]. rence of leiomyomas and endometriosis in women aged 20–54 years.
Methoxychlor (MXC) is an organochlorine pesticide that disrupts The authors obtained the influence of the studied endocrine-disrupting
hormonal balance [102]. MXC causes decreased ovarian mass, ovulation chemicals on the occurrence of fibroids, but no effect on endometri­
disorders, ovarian follicle atresia, as well as granule cell apoptosis, and osis. Mercury (Hg) and equol had the greatest effect on fibroids [109].
increased production of hydrogen peroxide, leading to DNA damage Changes in the uterine endometrium are epigenetically regulated
[103]. The studies were carried out on 96 mice approximately 4 weeks [10,44,76]. Xiong et al. [10] proved that BPA alters histone modification
of age [103]. The animals were divided into 12 groups (N = 8) and during endometrial decision-making, leading to impaired processes and
administered orally for 30 days to the MXC study groups and sesame oil failure of embryo implantation. In their studies, the authors examined
to the control groups. Four study groups were supplemented with MXC the effect of bisphenol A on MLL1 (mixed-lineage leukemia 1) and EZH2
32 mg/kg/day, and four were treated with 64 mg/kg/day. After 30 days (enhancer of zeste homolog 2) in human endometrium stromal cells in
of exposure to MXC, the animals were euthanized, and then the ovaries vitro. MLL1 and EZH2 are histone methylation transferases and play an
were analyzed, and the two study groups with the test doses and MXC important role in endometrial proliferation and development [10].
were supplemented for 60 days. Both doses of MXC significantly MLL1 is a histone methyltransferase that can trimethylate histone H3.
increased the number of primary and pre-antral follicles compared to the BPA was shown to inhibit the expression of MLL1 at the concentrations
controls, and the dose of 64 mg/kg/day (10.4 ± 3.6) almost doubled the of 10 and 100 nM [10]. Furthermore, 10 nM and 10 µM BPA have an
number of atretic follicles compared to the controls (4.9 ± 3.5). More­ influence on the morphology of endometrial cells during decision
over, there were no changes in serum anti-Müllerian hormone (AMH) making. EZH2, together with the catalytic subunit of the polycomb 2
levels in the study groups after exposure to MCX. In contrast, after 60 repressive complex (PRC2), participates in the suppression of the gene
days of dosing with 64 mg/kg/day of MXC, treatment with MXC by the methylation of histone H3 in lysine 27. Lowering the level of
significantly increased AMH levels (~ 1300 pg/ml) compared to the EZH2 leads to a reduction in the work of DNA damage in the uterine
control (~ 900 pg/ml AMH). Serum anti-Müllerian hormone is recog­ muscle, which is involved in the neoplasm [10,44,76]. Furthermore, in
nized as a marker of the quantitative aspect of the ovarian reserve [103]. the studies conducted, the expression of the serine-threonine protein
In prenatal exposure studies in mice, it has been shown that MXC con­ kinase (SGK1) protein was found to be reduced by the estrogen receptor
tributes to irregular oestrus cycles, lowering progesterone levels, as well in endometrial cells, leading to implantation failure in the mouse model
as stillbirths, and induction of oxidative stress causing damage to genetic [10]. SGK1 transmits a growth factor signal from phosphoinositide 3-ki­
material, leading to abnormalities in oocyte structure of oocytes [104]. nase (PI3K) during implantation in response to embryonic serine pro­
teases [110]. Exposure to very small amounts of bisphenol A (<1 nmol)
3.3. Uterus has been shown to increase the production of reactive oxygen species
(ROS), damaging genetic material, and the expression of inflammatory
The key hormones for maintaining the health of the uterus and the factors in the uterus [10].
reproductive cycle are estrogen and progesterone. In the case of disor­ Endometriosis is an estrogen-dependent endocrine disorder charac­
ders between these two hormones, there is a loss of pregnancy, uterine terized by endometrial hyperplasia with pain, bleeding, and infertility as

8
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

common complications [48,111]. This condition is associated with an influence of E2 in the livers of one-year-old female offspring (F1) was
increased risk of developing epithelial ovarian cancer. Environmental also studied. The animals were divided into groups according to the
factors such as endocrine-disrupting chemicals are believed to doses of E2 – the first group took the acceptable daily intake (ADI) of
contribute to the development of the endometriosis [111–113]. 0.05 μg/kg body weight/day, and the second group get a dose similar to
Bisphenol A, polychlorinated biphenyls, dioxins, organochlorine pesti­ the level of no observed effects level (NOEL) equal to 10 μg/kg body
cides, and phthalate esters are associated with the appearance of weight/day, the last one – high dose of 1000 μg/kg body weight/day,
endometriosis, but also primary and ovarian failure or miscarriage [48, and the control group which was not supplemented with E2. During the
112,114]. In the studies conducted by Ao et al. [115] on women who analysis of gene expression, as well as the analysis of DNA methylation
had miscarried before the 20th week of pregnancy, the effect of EDCs on changes, including in the liver, and endometrium of sows, induction of
miscarriage was examined. During the study, urine samples were tissue-specific changes in the expression of cell cycle regulatory genes (e.
collected and tested for BPA, bisphenol AF (BPAF), and BPS. In all tested g., DCKN2D, CDC42EP4, GADD45A, MGMT, CCN1, etc.), tumor sup­
samples, bisphenols were detected at the level of 78–90 %, of which BPA pressor genes (e.g. HIC1, BGN, PSAT1, etc.), two genes specific for
was the most frequently detected (87 %). Studies have shown an asso­ methylation - DNMT3a, MeCP2 in the studied pigs compared to the
ciation between these bisphenol analogues and the risk of unexplained control group. The highest increase of CDKN2D expression in the
miscarriage [115]. DEHP, which is one of the phthalate esters, has a endometrium was shown for groups of animals with NOEL and HIGH
much higher concentration in the plasma and urine of women suffering doses (9.0, 15.2 times increased). In turn, exposure to NOEL and HIGH
from endometriosis compared to healthy women [116]. In the study doses of E2 induced hypomethylation in the liver tissues of sows and in
conducted by Nazir et al. [116] on 50 women aged 20–40 struggling blastocysts. In the F1 generation, higher DNA hypomethylation was
with endometriosis, it was shown that sick women had a higher con­ found in the liver than in other tissues [121].
centration of phthalate esters compared to healthy women. The authors Endocrine-disrupting chemicals such as, for example, phthalates
found an association between DEHP exposure and endometriosis [116]. have the ability to cross the fetal-placental barrier and interfere with
placental development and embryonic growth [7,75]. Toxic metals,
3.4. Pregnancy bisphenol A, polycyclic aromatic hydrocarbons, and phthalates present
in the blood of pregnant women negatively affect the birth weight of
Exposure to EDCs during critical stages of development, such as children [122]. There are associated phthalates with fluctuations in the
embryogenesis, and implantation, can modulate the immune response, concentrations of steroid hormones like estradiol and estriol during
and promote inflammation that during pregnancy leads to preterm pregnancy [122]. During the research, the prenatal exposure to the birth
birth, pre-eclampsia, or reduced fetal growth [7,48]. weight of the newborn was assessed [123]. The study included 1857
During early pregnancy, a series of events occur that begin with pregnant women from whom urine and blood samples were collected
conception, then the blastocyst develops and implants in the uterus, and analyzed for EDCs – OC, phenols, metals, PFAS. During the tests,
which gradually becomes susceptible to the implantation process [75, BPA (88 %) and PCB (74 %) were most often detected in the tested
117]. Uterine receptivity, i.e. the ability to accept the embryo, occurs in samples. It was found that exposure to mixtures of OC and metals was
the middle luteal phase, during which progesterone and estrogen are associated with lower birth weight, with trans-nanochlor and lead
crucial, and then decisive takes place [118]. During which an increase in having the greatest impact, reducing the mean birth weight by 38 g (95
progesterone levels is observed, endometrial cell differentiation is %) and 39 g (95 %), respectively. In contrast, PFAS, phenols, and
induced, and the uterus becomes susceptible to embryo implantation. phthalates were not associated with birth weight [123]. In turn, expo­
Estrogen and progesterone modulate the production of cytokines, sure to organochlorine pesticides may be associated with decreased
growth factors, and prostaglandins that are important for the implan­ fertility, including the inability to become pregnant [124].
tation [117,118]. Another key factor is calcium, which is also involved EDCs have been suggested to cause preterm birth that occurs before
in fertilization. These factors contribute to the preparation of the 37 weeks gestation and is associated with health complications and in­
endometrium for the implantation [118]. During implantation, molec­ fant mortality [125]. Exposure to phthalates and parabens has been
ular events are also crucial [117,118]. Any disruption of genes encoding shown to increase the risk of preterm birth, as shown in studies on
factors and calcium channels (cationic channel of the transient receptor pregnant women. During the research, the impact of using hair oils
potential cation channel subfamily V (TRPV) member 5 and 6 (TRPV5 containing parabens and phthalates was analyzed on the time of birth
and TRPV6), or mucin 1 (MUC1), leukemia inhibitory factor (LIF), ho­ [125]. Phthalates alter the inflammatory pathways associated with
meobox A10 (HOXA10), needed for implantation, resulting in infertility preterm birth, while parabens disrupt the signaling of estrogen and
and implantation disorder [117,118]. Homeobox 10 and LIF are markers progesterone receptors. Of the 154 tested pregnant women, 7 % gave
of embryo implantation that are involved in endometrial decisionuali­ birth prematurely and used the oils before [125]. In addition, these
zation and receptivity [119]. Mucin 1, on the other hand, is a trans­ substances endanger embryos and fetuses during pregnancy, limiting
membrane proteoglycan that regulates uterine perceptiveness and their development [3].
enables implantation [117]. TRPV is involved in the exchange of cal­
cium ions [118]. Exposure to endosulfan, an organochlorine pesticide, 3.4.1. Embryo
has been shown to result in dysregulation and expression of HIXA10 and During embryonic development, which is critical for genome pro­
has been associated with implantation failure [119]. In turn, studies in gramming, environmental changes that occur may result in changes in
mice showed that 2 mg/kg of body weight 1-nitropyrene (1-NP) gene expression or cell function, affecting cell metabolism [7]. During
inhibited the leukemia inhibitory factor (LIF) pathway, as well as embryogenesis, cell differentiation is controlled by growth factors as
reduced the expression of estrogen and progesterone receptors, thus well as hormones, and this can be interfered with by EDCs [7]. The
interfering with endometrial proliferation and implantation capacity success of embryo development depends on the previous stages of
[120]. It has been proven that abnormal expression of RPV5, TRPV6, oogenesis and therefore depends on the quality of the oocytes [126].
PMCA1, and NCX1 genes caused by exposure to EDCs negatively affects Early developmental exposure to these substances is of key importance
calcium ion transport and interferes with implantation, and may even as embryos and fetuses are very sensitive to toxins and changes in
lead to apoptosis and necrosis [118]. Plasma membrane Ca2+-ATPase 1 endocrine management [7]. BPA has been detected in the amniotic fluid,
(PMCA1) and Na+/Ca2+ exchanger 1 (NCX1) participate in the flow of newborn blood, and placenta. Low-dose (nM) BPA has been found to
calcium ions [118]. have a similar affinity to 17β-estradiol in stimulating a cellular response.
In the studies on pregnant sow and 10 day blastocyst, the effect of There is a relationship between high levels of BPA and phthalates in the
17β-estradiol was analyzed [121]. Furthermore, the intergenerational urine and preterm birth [7].

9
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

In the research conducted by Zhou et al. [127] in rat embryos, it was hormone receptors in the placenta influences the susceptibility of the
found that EDCs to which mothers were exposed exhibited an effect of structure to hormonal disorders. BPA has the ability to methylate as well
increasing the thickness of the interventricular septum. Moreover, the as apoptosis of placental cells [131]. Exposure to polycyclic aromatic
exposure of females during pregnancy to endocrine-disrupting chem­ hydrocarbons and polybrominated diphenyl ethers has been shown to
icals results in low birth weight of the offspring as well as a risk of modify the IGF2 content in the placenta, which disrupts the balance
premature birth [3]. In addition, exposure to endocrine disruptors of the between the developing fetus and the mother [75,129]. In studies in
embryo may be associated with birth defects (teratogenic), impaired mice, IGF2 overexpression has been found to lead to placental and fetal
fertility during sexual maturity, and an increased risk of neurological hypertrophy [132]. EDCs are able to alter imprinting in the placenta of
and cardiovascular diseases [106,128]. The embryo exposed to EDCs mice, resulting in altered placental function in mice [75]. Moreover,
shows reduced development and growth. In turn, studies on chickens epigenetic changes caused by EDCs contribute to the occurrence of ab­
proved that the exposure to 220 ppm of BPA in chicken embryos normalities and neoplasms in the placenta [75]. Epidermal growth
increased their mortality by 60 %, and the remaining embryos were factor (EGF) and epidermal growth factor receptor (EGFR) are important
characterized by malformations or deformation of the bones and skull to placental physiology, which stimulates placental growth and func­
[128]. tion, including proliferation, and EGFR is a marker of proliferation in
In studies on bovine embryos, 0.05 mg/ml of biphenyl A and trophoblasts. Hypermethylation of EGF and EGFR following contact
biphenyl S (BPS) were tested for the oocyte maturation process and with endocrine-disrupting chemicals has been shown to lead to com­
further for in vitro embryonic development [126]. For the study, oocytes plications such as pre-eclampsia in pregnant females. The mechanism of
were aspirated post-mortem from cows’ ovaries to the nutrient medium action of endocrine-disrupting chemicals on DNA methylation distur­
and then matured. Mature cumulus-oocyte complexes (COCs) were bance is not yet known [75]. In a study conducted by Shi et al. [73],
subjected to IVF, after fertilization, the blastocysts were retained at the authors analyzed the influence of biphenyls in 0.5, 20, or 50 µg/kg/day
2–4 cell stage, 8–16 cell stage, and the morula stage. 0.05 mg/ml BPA doses in pregnant mice. It has been proven, that prenatal exposure to
and 0.05 mg/ml BPS were tested on these groups [126]. Granular cells EDCs, particularly biphenyls, results in impaired estrous cycles, a
secrete Anti-Mullerian Hormone (AMH), which mediates communica­ reduction in follicle count, and fertility in adult mice [73]. Moreover,
tion with the oocyte and the maturation process. Incubation of oocytes biphenyl stimulates germ cell breakdown in developing ovaries [73].
with 0.05 mg/ml BPA and BPS during oocyte maturation decreased the Imprinting is important in the regulation of the placental tran­
amount of AMH and inhibited the growth and development of cow scriptome, where its loss results in an imbalance between the embryo
embryos as well as fragmentation of their DNA due to bisphenol [126]. and the mother, and also results in a lower weight of the placenta [7,75].
Organochlorine pesticides and atrazine have a strong influence on microRNAs (miRNAs) play a role in the epigenetic control of gene
blastocyst formation and development, and MXC may directly influence expression by acting post-transcriptionally inhibiting gene expression
genetic abnormalities during embryo development [96]. [7]. These short, non-coding miRNAs are important in the proliferation,
embryonic development, embryonic development, and apoptosis of cells
3.4.2. Fetus whose synthesis is influenced by estrogen signaling. Therefore, BPA can
Fetal growth depends on fetal factors, endocrine regulation, and interfere with miRNA expression [7].
interaction between the placenta, mother, and fetus [7129,130]. Exposure of the fetus to diethylstilbestrol (DES) results in the
Essential hormones in fetal growth are insulin-like growth factors (IGF1, development of pathology in adulthood. It has been proven that the risk
IGF2), insulin, thyroid hormones, growth hormone, leptin, progester­ of developing clear cell adenocarcinoma (CCA) of the vagina and uterus,
one, estrogen, and cortisol [122,129]. Disturbances in the content of the development of endometriosis, premature menopause, and
these compounds result in disorders in the fetal growth [129]. abnormal gametogenesis, is also associated with unfavorable indicators
During pregnancy, hormonal disorders lead to difficulties in the of conception and pregnancy, and in men, it resulted in testicular hy­
maintenance and development of pregnancy, and even to its loss [18, perplasia and cryptorchidism [48,78,128]. DES was widely prescribed
131]. EDCs are transmitted to the fetus via the placenta [7]. The fetus is to pregnant women in the 1970 s as a preventive measure for miscar­
protected at an early stage from exogenous estrogens by riage [48]. There are also reports that exposure of prenatal to EDCs
alpha-fetoprotein, which is an estrogen-binding plasma protein. It is results in DNA methylation of genes encoding steroid hormone re­
noted that some endocrine disruptors may have a low affinity for this ceptors, which affects subsequent reproductive disorders [113].
protein and thus be unprotected. The fetus will then be exposed to these The effect of EDCs on female reproduction is shown in Fig. 2 (Fig. 2).
substances. EDCs, through their receptor affinity, are able to bind to
hormone receptors and thus be transferred to the fetus. Moreover, 4. Conclusions
endocrine-disrupting chemicals negatively affect cell differentiation,
and any defects in the tissues and their genome can be passed on to the Endocrine-disrupting chemicals adversely affect various body sys­
offspring [58]. However, the fetus is very sensitive to toxic substances, tems. In turn, in the reproductive system, they contribute, among others,
and its exposure negatively affects subsequent reproductive abilities, but to premature aging of the ovaries, dysfunction, and pathology of the
also cognitive and behavioral disorders, such as autism, attention deficit ovaries and uterus, endometriosis, polycystic ovary syndrome, prema­
hyperactivity disorder, or cardiovascular diseases, including hyperten­ ture births, or epigenetic changes in DNA methylation disturbing cell
sion, and coronary artery disease [7,18,130,131]. proliferation and even their apoptosis. The substances are common in
EDCs are toxic to the placenta, disrupting its functioning, and this the environment and show the ability to accumulate. In females, they
translates directly into the development of the fetus [18]. EDCs can were found in the follicular fluid, milk, and plasma. There is a need to
negatively affect birth weight, and placental weight [131]. The use of further investigate the effects of EDCs on the female reproductive sys­
personal care products containing phthalates, parabens, and biphenyls tem. Despite the health risks presented, especially for female repro­
has been shown to affect the development of the fetus and placenta ductive health, the use of EDCs is increasing every day. Long-term
through the presence of these substances in the circulatory system exposure to these substances of animals, humans, and the environment
[7131]. The placenta, as an organ necessary for the development of the is not yet fully understood. More research is needed to understand the
fetus, is involved in the transport between the mother and the fetus. The impact of EDCs on health and the environment and to develop methods
placenta of fetal origin develops from the blastocyst trophectoderm, for the disposal of EDCs. Moreover, there is a need to develop technol­
which is made up of stem cells (cytotrophoblast cells – CTB). EDCs have ogies for the production of products without the addition of EDCs to
been shown to limit the differentiation of CTB cells through epigenetic reduce human exposure to these substances.
changes or changes in gene regulation. Also, the expression of steroid

10
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

Fig. 2. The effect of ECDs on female reproduction [1–3,5–7,14,58,71,72].

Funding conceptualization and writing—original draft preparation; Alicja


Kowalczyk and Marcjanna Wrzecińska writing the final version of the
The APC/BPC is financed/co-financed by Wrocław University of manuscript. All authors have read and agreed to the published version of
Environmental and Life Sciences. the manuscript.

Ethics approval and consent to participate Coflict of interest statement

Not applicable. No potential conflict of interest was reported by the authors.

Consent for publication Data Availability

Not applicable. No data was used for the research described in the article.

Availability of data and material Acknowledgements

Not applicable. Not applicable.

CRediT authorship contribution statement References

All authors have made an important contribution to the creation of [1] C. Su, Y. Cui, D. Liu, H. Zhang, Y. Baninla, Endocrine disrupting compounds,
pharmaceuticals and personal care products in the aquatic environment of china:
this paper. Alicja Kowalczyk, Marcjanna Wrzecińska, Ewa Czerniawska- which chemicals are the prioritized ones? Sci. Total Environ. 720 (2020), 137652
Piątkowska, Jose Pedro Araujo and Przemysław Cwynar https://doi.org/10.1016/j.scitotenv.2020.137652.

11
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

[2] V. Ho, L. Pelland-St-Pierre, S. Gravel, M.F. Bouchard, M.-A. Verner, F. Labrèche, phenol pollutants, Environ. Pollut. 278 (2021), 116853, https://doi.org/
Endocrine disruptors: challenges and future directions in epidemiologic research, 10.1016/j.envpol.2021.116853.
Environ. Res. 204 (2022), 111969, https://doi.org/10.1016/j. [24] H.-T. Ong, H. Samsudin, H. Soto-Valdez, Migration of endocrine-disrupting
envres.2021.111969. chemicals into food from plastic packaging materials: an overview of chemical
[3] Y. Wu, J. Wang, Y. Wei, J. Chen, L. Kang, C. Long, S. Wu, L. Shen, G. Wei, risk assessment, techniques to monitor migration, and international regulations,
Maternal exposure to endocrine disrupting chemicals (EDCs) and preterm birth: a Crit. Rev. Food Sci. Nutr. 62 (2022) 957–979, https://doi.org/10.1080/
systematic review, meta-analysis, and meta-regression analysis, Environ. Pollut. 10408398.2020.1830747.
292 (2022), 118264, https://doi.org/10.1016/j.envpol.2021.118264. [25] A.Z. Aris, Z.A. Mohd Hir, M.R. Razak, Metal-organic frameworks (mofs) for the
[4] M. Al Sharabati, R. Abokwiek, A. Al-Othman, M. Tawalbeh, C. Karaman, adsorptive removal of selected endocrine disrupting compounds (EDCs) from
Y. Orooji, F. Karimi, Biodegradable polymers and their nano-composites for the aqueous solution: a review, Appl. Mater. Today 21 (2020), 100796, https://doi.
removal of endocrine-disrupting chemicals (EDCs) from wastewater: a review, org/10.1016/j.apmt.2020.100796.
Environ. Res. 202 (2021), 111694, https://doi.org/10.1016/j. [26] C.D. Metcalfe, S. Bayen, M. Desrosiers, G. Muñoz, S. Sauvé, V. Yargeau, An
envres.2021.111694. introduction to the sources, fate, occurrence and effects of endocrine disrupting
[5] A. Ismanto, T. Hadibarata, R.A. Kristanti, L. Maslukah, N. Safinatunnajah, chemicals released into the environment, Environ. Res. 207 (2022), 112658,
W. Kusumastuti, Endocrine disrupting chemicals (EDCs) in environmental https://doi.org/10.1016/j.envres.2021.112658.
matrices: occurrence, fate, health impact, physio-chemical and bioremediation [27] F.D.L. Leusch, P.A. Neale, F. Busetti, M. Card, A. Humpage, J.D. Orbell, H.
technology, Environ. Pollut. 302 (2022), 119061, https://doi.org/10.1016/j. F. Ridgway, M.B. Stewart, J.P. van de Merwe, B.I. Escher, Transformation of
envpol.2022.119061. endocrine disrupting chemicals, pharmaceutical and personal care products
[6] R. Meli, A. Monnolo, C. Annunziata, C. Pirozzi, M.C. Ferrante, Oxidative stress during drinking water disinfection, Sci. Total Environ. 657 (2019) 1480–1490,
and BPA toxicity: an antioxidant approach for male and female reproductive https://doi.org/10.1016/j.scitotenv.2018.12.106.
dysfunction, Antioxidants 9 (2020) 405, https://doi.org/10.3390/ [28] E.C. Plunk, S.M. Richards, Endocrine-disrupting air pollutants and their effects on
antiox9050405. the hypothalamus-pituitary-gonadal axis, IJMS 21 (2020) 9191, https://doi.org/
[7] S. Basak, M.K. Das, A.K. Duttaroy, Plastics derived endocrine-disrupting 10.3390/ijms21239191.
compounds and their effects on early development, Birth Defects Res. 112 (2020) [29] B. Yilmaz, H. Terekeci, S. Sandal, F. Kelestimur, Endocrine disrupting chemicals:
1308–1325, https://doi.org/10.1002/bdr2.1741. exposure, effects on human health, mechanism of action, models for testing and
[8] V. Padmanabhan, W. Song, M. Puttabyatappa, Praegnatio perturbatio—impact of strategies for prevention, Rev. Endocr. Metab. Disord. 21 (2020) 127–147,
endocrine-disrupting chemicals, Endocr. Rev. 42 (2021) 295–353, https://doi. https://doi.org/10.1007/s11154-019-09521-z.
org/10.1210/endrev/bnaa035. [30] E. Junqué, S. Garcia, M.Á. Martínez, J. Rovira, M. Schuhmacher, J.O. Grimalt,
[9] D. Franssen, T. Svingen, D. Lopez Rodriguez, M. Van Duursen, J. Boberg, A.- Changes of organochlorine compound concentrations in maternal serum during
S. Parent, A putative adverse outcome pathway network for disrupted female pregnancy and comparison to serum cord blood composition, Environ. Res. 182
pubertal onset to improve testing and regulation of endocrine disrupting (2020), 108994, https://doi.org/10.1016/j.envres.2019.108994.
chemicals, Neuroendocrinology 112 (2022) 101–114, https://doi.org/10.1159/ [31] P. Xiong, X. Yan, Q. Zhu, G. Qu, J. Shi, C. Liao, G. Jiang, A review of
000515478. environmental occurrence, fate, and toxicity of novel brominated flame
[10] Y. Xiong, X. Wen, H. Liu, M. Zhang, Y. Zhang, Bisphenol a affects endometrial retardants, Environ. Sci. Technol. 53 (2019) 13551–13569, https://doi.org/
stromal cells decidualization, involvement of epigenetic regulation, J. Steroid 10.1021/acs.est.9b03159.
Biochem. Mol. Biol. 200 (2020), 105640, https://doi.org/10.1016/j. [32] S.K. Kirkok, J.K. Kibet, T.K. Kinyanjui, F.I. Okanga, A review of persistent organic
jsbmb.2020.105640. pollutants: dioxins, furans, and their associated nitrogenated analogues, SN Appl.
[11] R. Lauretta, A. Sansone, M. Sansone, F. Romanelli, M. Appetecchia, Endocrine Sci. 2 (2020) 1729, https://doi.org/10.1007/s42452-020-03551-y.
disrupting chemicals: effects on endocrine glands, Front. Endocrinol. 10 (2019) [33] R.J. Wright, R. Bosch, M.I. Gibson, J.A. Christie-Oleza, Plasticizer degradation by
178, https://doi.org/10.3389/fendo.2019.00178. marine bacterial isolates: a proteogenomic and metabolomic characterization,
[12] E. Diamanti-Kandarakis, J.-P. Bourguignon, L.C. Giudice, R. Hauser, G.S. Prins, A. Environ. Sci. Technol. 54 (2020) 2244–2256, https://doi.org/10.1021/acs.
M. Soto, R.T. Zoeller, A.C. Gore, Endocrine-disrupting chemicals: an endocrine est.9b05228.
society scientific statement, Endocr. Rev. 30 (2009) 293–342, https://doi.org/ [34] L. Chen, T. Maqbool, G. Nazir, C. Hou, Y. Yang, J. Guo, X. Zhang, Developing the
10.1210/er.2009-0002. large-area manganese-based catalytic ceramic membrane for peroxymonosulfate
[13] T. Encarnação, A.A. Pais, M.G. Campos, H.D. Burrows, Endocrine disrupting activation: applications in degradation of endocrine disrupting compounds in
chemicals: impact on human health, wildlife and the environment, Sci. Prog. 102 drinking water, J. Membr. Sci. 655 (2022), 120602, https://doi.org/10.1016/j.
(2019) 3–42, https://doi.org/10.1177/0036850419826802. memsci.2022.120602.
[14] G.M. Calderón-Moreno, J. Vergara-Sánchez, H. Saldarriaga-Noreña, M.L. García- [35] M.F.M.A. Zamri, R. Bahru, F. Suja’, A.H. Shamsuddin, S.K. Pramanik, I.M.
Betancourt, M.L. Domínguez-Patiño, G.E. Moeller-Chávez, J.G. Ronderos-Lara, M. R. Fattah, Treatment strategies for enhancing the removal of endocrine-disrupting
I. Arias-Montoya, I.J. Montoya-Balbas, M.A. Murillo-Tovar, Occurrence and risk chemicals in water and wastewater systems, J. Water Process Eng. 41 (2021),
assessment of steroidal hormones and phenolic endocrine disrupting compounds 102017, https://doi.org/10.1016/j.jwpe.2021.102017.
in surface water in Cuautla River, Mexico, Water 11 (2019) 2628, https://doi. [36] M. Alikarami, R. Darvishi Cheshmeh Soltani, A. Khataee, An innovative
org/10.3390/w11122628. combination of electrochemical and photocatalytic processes for decontamination
[15] M. Čelić, B.D. Škrbić, S. Insa, J. Živančev, M. Gros, M. Petrović, Occurrence and of bisphenol a endocrine disruptor form aquatic phase: insight into mechanism,
assessment of environmental risks of endocrine disrupting compounds in enhancers and bio-toxicity assay, Sep. Purif. Technol. 220 (2019) 42–51, https://
drinking, surface and wastewaters in Serbia, Environ. Pollut. 262 (2020), 114344, doi.org/10.1016/j.seppur.2019.03.056.
https://doi.org/10.1016/j.envpol.2020.114344. [37] R.B. González-González, L. Parra-Arroyo, R. Parra-Saldívar, R.A. Ramirez-
[16] C. Pironti, M. Ricciardi, A. Proto, P.M. Bianco, L. Montano, O. Motta, Endocrine- Mendoza, H.M.N. Iqbal, Nanomaterial-based catalysts for the degradation of
disrupting compounds: an overview on their occurrence in the aquatic endocrine-disrupting chemicals – a way forward to environmental remediation,
environment and human exposure, Water 13 (2021) 1347, https://doi.org/ Mater. Lett. 308 (2022), 131217, https://doi.org/10.1016/j.matlet.2021.131217.
10.3390/w13101347. [38] C.G. Moreira, M.H. Moreira, V.M.O.C. Silva, H.G. Santos, D.M. Bila, F.V. Fonseca,
[17] J. Xue, L. Zhao, D. Fan, H. Feng, Z. Li, Effects of environmental-related Treatment of bisphenol A (BPA) in water using UV/H2O2 and reverse osmosis
concentration of tamoxifen on hormonal balance and reproduction of the Golden (RO) membranes: assessment of estrogenic activity and membrane adsorption,
Apple Snail, Pomacea canaliculata (Caenogastropoda: Ampullariidae), Water Air Water Sci. Technol. 80 (2019) 2169–2178, https://doi.org/10.2166/
Soil Pollut. 232 (2021) 517, https://doi.org/10.1007/s11270-021-05472-x. wst.2020.024.
[18] C. Yang, G. Song, W. Lim, A mechanism for the effect of endocrine disrupting [39] T. Olmez-Hanci, S. Dogruel, A.D.A. Emek, C.E. Yılmazer, S. Çınar, O. Kiraz,
chemicals on placentation, Chemosphere 231 (2019) 326–336, https://doi.org/ E. Citil, A.K. Orhon, E. Siltu, S.M. Gucver, et al., Performance of ozone and
10.1016/j.chemosphere.2019.05.133. peroxone on the removal of endocrine disrupting chemicals (EDCs) coupled with
[19] E.K. Radwan, M.B.M. Ibrahim, A. Adel, M. Farouk, The occurrence and risk cost analysis, Water Sci. Technol. 82 (2020) 640–650, https://doi.org/10.2166/
assessment of phenolic endocrine-disrupting chemicals in Egypt’s drinking and wst.2020.339.
source water, Environ. Sci. Pollut. Res 27 (2020) 1776–1788, https://doi.org/ [40] N. Garcia-Reyero, The clandestine organs of the endocrine system, Gen. Comp.
10.1007/s11356-019-06887-0. Endocrinol. 257 (2018) 264–271, https://doi.org/10.1016/j.ygcen.2017.08.017.
[20] M. Milanović, L. Đurić, N. Milošević, N. Milić, Comprehensive insight into [41] S.R. Wilkenfeld, C. Lin, D.E. Frigo, Communication between genomic and non-
triclosan—from widespread occurrence to health outcomes, Environ. Sci. Pollut. genomic signaling events coordinate steroid hormone actions, Steroids 133
Res (2021), https://doi.org/10.1007/s11356-021-17273-0. (2018) 2–7, https://doi.org/10.1016/j.steroids.2017.11.005.
[21] Í.F.S. Gonçalves, T.M. Souza, L.R. Vieira, F.C. Marchi, A.P. Nascimento, D. [42] M.D. Lakshmanan, K. Shaheer, Endocrine disrupting chemicals may deregulate
F. Farias, Toxicity testing of pesticides in zebrafish—a systematic review on DNA repair through estrogen receptor mediated seizing of CBP/P300 acetylase,
chemicals and associated toxicological endPoints, Environ. Sci. Pollut. Res 27 J. Endocrinol. Invest 43 (2020) 1189–1196, https://doi.org/10.1007/s40618-
(2020) 10185–10204, https://doi.org/10.1007/s11356-020-07902-5. 020-01241-5.
[22] L. Rani, K. Thapa, N. Kanojia, N. Sharma, S. Singh, A.S. Grewal, A.L. Srivastav, [43] Z.-R. Tang, R. Zhang, Z.-X. Lian, S.-L. Deng, K. Yu, Estrogen-receptor expression
J. Kaushal, An extensive review on the consequences of chemical pesticides on and function in female reproductive disease, Cells 8 (2019) 1123, https://doi.
human health and environment, J. Clean. Prod. 283 (2021), 124657, https://doi. org/10.3390/cells8101123.
org/10.1016/j.jclepro.2020.124657. [44] C.S. Rosenfeld, P.S. Cooke, Endocrine disruption through membrane estrogen
[23] P. Astrahan, L. Korzen, M. Khanin, Y. Sharoni, Á. Israel, Seaweeds fast EDC receptors and novel pathways leading to rapid toxicological and epigenetic
bioremediation: supporting evidence of EE2 and BPA degradation by the red effects, J. Steroid Biochem. Mol. Biol. 187 (2019) 106–117, https://doi.org/
seaweed gracilaria sp., and a proposed model for the remedy of marine-borne 10.1016/j.jsbmb.2018.11.007.

12
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

[45] L. Toporova, P. Balaguer, Nuclear receptors are the major targets of endocrine chemical exposure in the mammalian hypothalamic-pituitary axis, Mol. Cell.
disrupting chemicals, Mol. Cell. Endocrinol. 502 (2020), 110665, https://doi. Endocrinol. 518 (2020), 110997, https://doi.org/10.1016/j.mce.2020.110997.
org/10.1016/j.mce.2019.110665. [69] P. Di Pietro, R. D’Auria, A. Viggiano, E. Ciaglia, R. Meccariello, R.D. Russo, A.
[46] E. Thouennon, V. Delfosse, R. Bailly, P. Blanc, A. Boulahtouf, M. Grimaldi, A. Puca, C. Vecchione, S.L. Nori, A. Santoro, Bisphenol A Induces DNA damage in
A. Barducci, W. Bourguet, P. Balaguer, Insights into the activation mechanism of cells exerting immune surveillance functions at peripheral and central Level,
human estrogen-related receptor γ by environmental endocrine disruptors, Cell. Chemosphere 254 (2020), 126819, https://doi.org/10.1016/j.
Mol. Life Sci. 76 (2019) 4769–4781, https://doi.org/10.1007/s00018-019- chemosphere.2020.126819.
03129-x. [70] M.A. La Merrill, L.N. Vandenberg, M.T. Smith, W. Goodson, P. Browne, H.
[47] A. Matsushima, T. Teramoto, Y. Kakuta, Crystal structure of endocrine-disrupting B. Patisaul, K.Z. Guyton, A. Kortenkamp, V.J. Cogliano, T.J. Woodruff, et al.,
chemical bisphenol a and estrogen-related receptor γ, J. Biochem. 171 (2022) Consensus on the key characteristics of endocrine-disrupting chemicals as a basis
23–25, https://doi.org/10.1093/jb/mvab145. for hazard identification, Nat. Rev. Endocrinol. 16 (2020) 45–57, https://doi.org/
[48] J.M. Hall, C.W. Greco, Perturbation of nuclear hormone receptors by endocrine 10.1038/s41574-019-0273-8.
disrupting chemicals: mechanisms and pathological consequences of exposure, [71] L. Lucaccioni, V. Trevisani, L. Marrozzini, N. Bertoncelli, B. Predieri, L. Lugli,
Cells 9 (2019) 13, https://doi.org/10.3390/cells9010013. A. Berardi, L. Iughetti, Endocrine-disrupting chemicals and their effects during
[49] H. Hua, H. Zhang, Q. Kong, Y. Jiang, Mechanisms for estrogen receptor female puberty: a review of current evidence, IJMS 21 (2020) 2078, https://doi.
expression in human cancer, Exp. Hematol. Oncol. 7 (2018) 24, https://doi.org/ org/10.3390/ijms21062078.
10.1186/s40164-018-0116-7. [72] D. López-Rodríguez, C.F. Aylwin, V. Delli, E. Sevrin, M. Campanile, M. Martin,
[50] W. Yang, W. Tan, J. Zheng, B. Zhang, H. Li, X. Li, MEHP promotes the D. Franssen, A. Gérard, S. Blacher, E. Tirelli, et al., Multi- and transgenerational
proliferation of cervical cancer via GPER mediated activation of Akt, Eur. J. outcomes of an exposure to a mixture of endocrine-disrupting chemicals (EDCs)
Pharmacol. 824 (2018) 11–16, https://doi.org/10.1016/j.ejphar.2018.01.040. on puberty and maternal behavior in the female rat, Environ. Health Perspect.
[51] N. Pupinyo, C. D’Costa, A. Heiskanen, W. Laiwattanapaisal, J. Emnéus, 129 (2021), 087003, https://doi.org/10.1289/EHP8795.
Impedance-based E-screen cell biosensor for the real-time screening of [73] M. Shi, N. Sekulovski, J.A. MacLean, A. Whorton, K. Hayashi, Prenatal exposure
xenoestrogenic compounds, ACS EST Water 2 (2022) 446–456, https://doi.org/ to bisphenol a analogues on female reproductive functions in mice, Toxicol. Sci.
10.1021/acsestwater.1c00383. 168 (2019) 561–571, https://doi.org/10.1093/toxsci/kfz014.
[52] L.N. Vandenberg, Non-monotonic dose responses in studies of endocrine [74] B. Robaire, G. Delbes, J.A. Head, V.L. Marlatt, C.J. Martyniuk, S. Reynaud, V.
disrupting chemicals: bisphenol A as a case study, dose-response.1, Dose- L. Trudeau, J.A. Mennigen, A cross-species comparative approach to assessing
Response 12 (2014), https://doi.org/10.2203/dose-response.13-020. multi- and transgenerational effects of endocrine disrupting chemicals, Environ.
Vandenberg. Res. 204 (2022), 112063, https://doi.org/10.1016/j.envres.2021.112063.
[53] EFSA Scientific Committee, S. More, D. Benford, S. Hougaard Bennekou, [75] N.M. Grindler, L. Vanderlinden, R. Karthikraj, K. Kannan, S. Teal, A.J. Polotsky,
V. Bampidis, C. Bragard, T. Halldorsson, A. Hernandez-Jerez, K. Koutsoumanis, T.L. Powell, I.V. Yang, T. Jansson, Exposure to phthalate, an endocrine disrupting
C. Lambré, et al., Opinion on the impact of non-monotonic dose responses on chemical, alters the first trimester placental methylome and transcriptome in
EFSA′ s human health risk assessments, EFS2 (2021) 19, https://doi.org/10.2903/ women, Sci. Rep. 8 (2018) 6086, https://doi.org/10.1038/s41598-018-24505-w.
j.efsa.2021.6877. [76] A.M. Retis-Resendiz, I.N. González-García, M. León-Juárez, I. Camacho-Arroyo,
[54] L.N. Vandenberg, Endocrine disrupting chemicals: strategies to protect present M. Cerbón, E.R. Vázquez-Martínez, The role of epigenetic mechanisms in the
and future generations, Expert Rev. Endocrinol. Metab. 16 (2021) 135–146, regulation of gene expression in the cyclical endometrium, Clin. Epigenet 13
https://doi.org/10.1080/17446651.2021.1917991. (2021) 116, https://doi.org/10.1186/s13148-021-01103-8.
[55] M.A. Badding, L. Barraj, A.L. Williams, C. Scrafford, R. Reiss, CLARITY-BPA core [77] E.C. Plunk, S.M. Richards, Epigenetic modifications due to environment, ageing,
study: analysis for non-monotonic dose-responses and biological relevance, Food nutrition, and endocrine disrupting chemicals and their effects on the endocrine
Chem. Toxicol. 131 (2019), 110554, https://doi.org/10.1016/j.fct.2019.06.001. system, Int. J. Endocrinol. 2020 (2020) 1–11, https://doi.org/10.1155/2020/
[56] H. Tan, X. Wang, H. Hong, E. Benfenati, J.P. Giesy, G.C. Gini, R. Kusko, X. Zhang, 9251980.
H. Yu, W. Shi, Structures of endocrine-disrupting chemicals determine binding to [78] V.L. Marlatt, S. Bayen, D. Castaneda-Cortès, G. Delbès, P. Grigorova, V.
and activation of the estrogen receptor α and androgen receptor, Environ. Sci. S. Langlois, C.J. Martyniuk, C.D. Metcalfe, L. Parent, A. Rwigemera, et al.,
Technol. 54 (2020) 11424–11433, https://doi.org/10.1021/acs.est.0c02639. Impacts of endocrine disrupting chemicals on reproduction in wildlife and
[57] D. Di Paola, F. Capparucci, G. Lanteri, M. Cordaro, R. Crupi, R. Siracusa, humans, Environ. Res. 208 (2022), 112584, https://doi.org/10.1016/j.
R. D’Amico, R. Fusco, D. Impellizzeri, S. Cuzzocrea, et al., Combined toxicity of envres.2021.112584.
xenobiotics bisphenol a and heavy metals on zebrafish embryos (Danio rerio), [79] J. Parker, C. O’Brien, F.L. Gersh, Developmental origins and transgenerational
Toxics 9 (2021) 344, https://doi.org/10.3390/toxics9120344. inheritance of polycystic ovary syndrome, Aust. N. Z. J. Obstet. Gynaecol. 61
[58] S. Rattan, J.A. Flaws, The epigenetic impacts of endocrine disruptors on female (2021) 922–926, https://doi.org/10.1111/ajo.13420.
reproduction across generations, Biol. Reprod. 101 (2019) 635–644, https://doi. [80] A. Santaliz Casiano, A. Lee, D. Teteh, Z. Madak Erdogan, L. Treviño, Endocrine-
org/10.1093/biolre/ioz081. disrupting chemicals and breast cancer: disparities in exposure and importance of
[59] V.L. Flöter, G. Galateanu, R.W. Fürst, D. Seidlová-Wuttke, W. Wuttke, E. Möstl, T. research inclusivity, Endocrinology 163 (2022) bqac034, https://doi.org/
B. Hildebrandt, S.E. Ulbrich, Sex-specific effects of low-dose gestational estradiol- 10.1210/endocr/bqac034.
17β exposure on bone development in porcine offspring, Toxicology 366–367 [81] J. Winkler, P. Liu, K. Phong, J.H. Hinrichs, N. Ataii, K. Williams, E. Hadler-Olsen,
(2016) 60–67, https://doi.org/10.1016/j.tox.2016.07.012. S. Samson, Z.J. Gartner, S. Fisher, et al., Bisphenol A replacement chemicals, BPF
[60] J. Boberg, C. Taxvig, S. Christiansen, U. Hass, Possible endocrine disrupting and BPS, induce protumorigenic changes in human mammary gland organoid
effects of parabens and their metabolites, Reprod. Toxicol. 30 (2010) 301–312, morphology and proteome, Proc. Natl. Acad. Sci. U. S. A 119 (2022),
https://doi.org/10.1016/j.reprotox.2010.03.011. e2115308119, https://doi.org/10.1073/pnas.2115308119.
[61] Cressey, P. HEALTH RISK ASSESSMENT: PARABENS IN PERSONAL CARE [82] M.A.A. Ibrahim, R.H. Elbakry, N.A. Bayomy, Effect of bisphenol A on
PRODUCTS; Science for Communities, 2021. morphology, apoptosis and proliferation in the resting mammary gland of the
[62] EFSA Panel on Food Contact Materials, Enzymes and Processing Aids (CEP); adult albino rat, Int. J. Exp. Pathl. 97 (2016) 27–36, https://doi.org/10.1111/
Silano, V.; Barat Baviera, J.M.; Bolognesi, C.; Chesson, A.; Cocconcelli, P.S.; iep.12164.
Crebelli, R.; Gott, D.M.; Grob, K.; Lampi, E.; et al. Update of the Risk Assessment [83] X. Wang, N. Luo, Z. Xu, X. Zheng, B. Huang, X. Pan, The estrogenic proliferative
of Di-butylphthalate (DBP), Butyl-benzyl-phthalate (BBP), Bis(2–ethylhexyl) effects of two alkylphenols and a preliminary mechanism exploration in MCF-7
Phthalate (DEHP), Di-isononylphthalate (DINP) and Di-isodecylphthalate (DIDP) breast cancer cells, Environ. Toxicol. 35 (2020) 628–638, https://doi.org/
for Use in Food Contact Materials; 2019; 10.1002/tox.22898.
[63] EFSA Opinion of the Scientific Panel on Food Additives, Flavourings, Processing [84] P. Peremiquel-Trillas, Y. Benavente, M. Martín-Bustamante, D. Casabonne,
Aids and Materials in Contact with Food (AFC) on a Request from the Commission B. Pérez-Gómez, I. Gómez-Acebo, A. Oliete-Canela, M. Diéguez-Rodríguez,
Related to Butylbenzylphthalate (BBP) for Use in Food Contact Materials I. Tusquets, P. Amiano, et al., Alkylphenolic compounds and risk of breast and
Question N◦ EFSA-Q-2003–190 2005. prostate cancer in the MCC-spain study, Environ. Int. 122 (2019) 389–399,
[64] EFSA Opinion of the Scientific Panel on Food Additives, Flavourings, Processing https://doi.org/10.1016/j.envint.2018.12.007.
Aids and Materials in Contact with Food (AFC) Related to 2,2-BIS(4-HYDROXY­ [85] A. Mukherjee Das, A. Gogia, M. Garg, A. Elaiyaraja, P. Arambam, S. Mathur,
PHENYL)PROPANE; The EFSA Journal (2006) 428, 1 of 75; 2006; R. Babu-Rajendran, S.V.S. Deo, L. Kumar, B.C. Das, et al., Urinary concentration
[65] EFSA Opinion of the Scientific Panel on Food Additives, Flavourings, Processing of endocrine-disrupting phthalates and breast cancer risk in indian women: a
Aids and Materials in Contact with Food (AFC) on a Request from the Commission case-control study with a focus on mutations in phthalate-responsive genes,
Related to Bis(2-Ethylhexyl)Phthalate (DEHP) for Use in Food Contact Materials; Cancer Epidemiol. 79 (2022), 102188, https://doi.org/10.1016/j.
The EFSA Journal (2005) 243, 1–20; 2005; canep.2022.102188.
[66] R.D. Björvang, P. Damdimopoulou, Persistent environmental endocrine- [86] Y.J. Cho, J.H. Yun, S.J. Kim, H.Y. Kwon, Nonpersistent endocrine disrupting
disrupting chemicals in ovarian follicular fluid and in vitro fertilization treatment chemicals and reproductive health of women, Obstet. Gynecol. Sci. 63 (2020) 1,
outcome in women, Upsala J. Med. Sci. 125 (2020) 85–94, https://doi.org/ https://doi.org/10.5468/ogs.2020.63.1.1.
10.1080/03009734.2020.1727073. [87] Y. Hu, S. Wen, D. Yuan, L. Peng, R. Zeng, Z. Yang, Q. Liu, L. Xu, D. Kang, The
[67] M. Iwamoto, T. Masuya, M. Hosose, K. Tagawa, T. Ishibashi, K. Suyama, T. Nose, association between the environmental endocrine disruptor bisphenol a and
E. Yoshihara, M. Downes, R.M. Evans, et al., Bisphenol A derivatives act as novel polycystic ovary syndrome: a systematic review and meta-analysis, Gynecol.
coactivator-binding inhibitors for estrogen receptor β, J. Biol. Chem. 297 (2021), Endocrinol. 34 (2018) 370–377, https://doi.org/10.1080/
101173 https://doi.org/10.1016/j.jbc.2021.101173. 09513590.2017.1405931.
[68] J.B. Graceli, R.S. Dettogni, E. Merlo, O. Niño, C.S. da Costa, J.F. Zanol, E.A. Ríos [88] M.P. Green, A.J. Harvey, B.J. Finger, G.A. Tarulli, Endocrine disrupting
Morris, L. Miranda-Alves, A.C. Denicol, The impact of endocrine-disrupting chemicals: impacts on human fertility and fecundity during the peri-conception

13
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

period, Environ. Res. 194 (2021), 110694, https://doi.org/10.1016/j. [110] M.S. Salker, J.H. Steel, Z. Hosseinzadeh, J. Nautiyal, Z. Webster, Y. Singh,
envres.2020.110694. S. Brucker, F. Lang, J.J. Brosens, Activation of SGK1 in endometrial epithelial
[89] M.B.M. van Duursen, J. Boberg, S. Christiansen, L. Connolly, P. Damdimopoulou, cells in response to PI3K/AKT inhibition impairs embryo implantation, Cell
P. Filis, P.A. Fowler, B.M. Gadella, J. Holte, K. Jääger, et al., Safeguarding female Physiol. Biochem 39 (2016) 2077–2087, https://doi.org/10.1159/000447903.
reproductive health against endocrine disrupting chemicals-The FREIA Project, [111] D. Sirohi, R. Al Ramadhani, L.D. Knibbs, Environmental exposures to endocrine
IJMS 21 (2020) 3215, https://doi.org/10.3390/ijms21093215. disrupting chemicals (EDCs) and their role in endometriosis: a systematic
[90] J.A. Hinds, E.R. Sanchez, The role of the hypothalamus–pituitary–adrenal (HPA) literature review, Rev. Environ. Health 36 (2021) 101–115, https://doi.org/
axis in test-induced anxiety: assessments, physiological responses, and molecular 10.1515/reveh-2020-0046.
details, Stresses 2 (2022) 146–155, https://doi.org/10.3390/stresses2010011. [112] X. Wen, Y. Xiong, X. Qu, L. Jin, C. Zhou, M. Zhang, Y. Zhang, The risk of
[91] M.W. Asmat, M.H. Ramzan, F. Ramzan, Exogenous neurokinin b administration endometriosis after exposure to endocrine-disrupting chemicals: a meta-analysis
may have a strong effect on negative feedback loop of hypothalamic pituitary of 30 epidemiology studies, Gynecol. Endocrinol. 35 (2019) 645–650, https://doi.
thyroid axis, Int J. Pept. Res Ther. 26 (2020) 1259–1267, https://doi.org/ org/10.1080/09513590.2019.1590546.
10.1007/s10989-019-09925-0. [113] K. Wieczorek, D. Szczęsna, J. Jurewicz, Environmental exposure to non-persistent
[92] A. Sen, H.M. Hoffmann, Role of core circadian clock genes in hormone release endocrine disrupting chemicals and endometriosis: a systematic review, IJERPH
and target tissue sensitivity in the reproductive axis, Mol. Cell. Endocrinol. 501 19 (2022) 5608, https://doi.org/10.3390/ijerph19095608.
(2020), 110655, https://doi.org/10.1016/j.mce.2019.110655. [114] M.A. Moreira Fernandez, Z.L. Cardeal, M.M. Carneiro, L.C. André, Study of
[93] P. Ananthasubramanian, S. Ananth, S. Kumaraguru, S. Barathi, W. Santosh, possible association between endometriosis and phthalate and bisphenol A by
R. Vasantharekha, Associated effects of endocrine disrupting chemicals (edcs) on biomarkers analysis, J. Pharm. Biomed. Anal. 172 (2019) 238–242, https://doi.
neuroendocrine axes and neurotransmitter profile in polycystic ovarian syndrome org/10.1016/j.jpba.2019.04.048.
condition, Proc. Zool. Soc. 74 (2021) 378–386, https://doi.org/10.1007/s12595- [115] J. Ao, X. Huo, J. Zhang, Y. Mao, G. Li, J. Ye, Y. Shi, F. Jin, S. Bao, J. Zhang,
021-00411-4. Environmental exposure to bisphenol analogues and unexplained recurrent
[94] M.P. Reilly, M.N. Kunkel, L.M. Thompson, A. Zentay, C.D. Weeks, D. Crews, L. miscarriage: a case-control study, Environ. Res. 204 (2022), 112293, https://doi.
K. Cormack, A.C. Gore, Effects of endocrine-disrupting chemicals on org/10.1016/j.envres.2021.112293.
hypothalamic oxytocin and vasopressin systems, J. Exp. Zool. A Ecol. Integr. [116] S. Nazir, Z. Usman, M. Imran, K. Lone, G. Ahmad, Women diagnosed with
Physiol. 337 (2022) 75–87, https://doi.org/10.1002/jez.2475. endometriosis show high serum levels of diethyl hexyl phthalate, J. Hum. Reprod.
[95] Z. Sheng, C. Wang, F. Ren, Y. Liu, B. Zhu, Molecular mechanism of endocrine- Sci. 11 (2018) 131, https://doi.org/10.4103/jhrs.JHRS_137_17.
disruptive effects induced by bisphenol A: the role of transmembrane G-protein [117] D. Caserta, F. Costanzi, M.P. De Marco, L. Di Benedetto, E. Matteucci, C. Assorgi,
estrogen receptor 1 and Integrin Аvβ3, J. Environ. Sci. 75 (2019) 1–13, https:// M.C. Pacilli, A.R. Besharat, F. Bellati, I. Ruscito, Effects of endocrine-disrupting
doi.org/10.1016/j.jes.2018.05.002. chemicals on endometrial receptivity and embryo implantation: a systematic
[96] R. Cabry, P. Merviel, A. Madkour, E. Lefranc, F. Scheffler, R. Desailloud, V. Bach, review of 34 mouse model studies, IJERPH 18 (2021) 6840, https://doi.org/
M. Benkhalifa, The impact of endocrine disruptor chemicals on oocyte/embryo 10.3390/ijerph18136840.
and clinical outcomes in IVF, Endocr. Connect. 9 (2020) R134–R142, https://doi. [118] D.N. Tran, E.-M. Jung, C. Ahn, J.-H. Lee, Y.-M. Yoo, E.-B. Jeung, Effects of
org/10.1530/EC-20-0135. bisphenol A and 4-tert-octylphenol on embryo implantation failure in mouse,
[97] J.L. Chiang, P. Shukla, K. Pagidas, N.S. Ahmed, S. Karri, D.D. Gunn, W.W. Hurd, IJERPH 15 (2018) 1614, https://doi.org/10.3390/ijerph15081614.
K.K. Singh, Mitochondria in ovarian aging and reproductive longevity, Ageing [119] M.M. Milesi, V. Lorenz, J. Varayoud, Aberrant hoxa10 gene methylation as a
Res. Rev. 63 (2020), 101168, https://doi.org/10.1016/j.arr.2020.101168. mechanism for endosulfan-induced implantation failures in rats, Mol. Cell.
[98] R. Battaglia, P. Musumeci, M. Ragusa, D. Barbagallo, M. Scalia, M. Zimbone, J. Endocrinol. 547 (2022), 111576, https://doi.org/10.1016/j.mce.2022.111576.
M. Lo Faro, P. Borzì, P. Scollo, M. Purrello, et al., Ovarian aging increases small [120] Y. Liang, Q. Shuai, Y. Wang, S. Jin, Z. Feng, B. Chen, T. Liang, Z. Liu, H. Zhao,
extracellular vesicle CD81+ release in human follicular fluid and influences Z. Chen, et al., 1-Nitropyrene exposure impairs embryo implantation through
MiRNA profiles, Aging 12 (2020) 12324–12341, https://doi.org/10.18632/ disrupting endometrial receptivity genes expression and producing excessive
aging.103441. ROS, Ecotoxicol. Environ. Saf. 227 (2021), 112939, https://doi.org/10.1016/j.
[99] T. Ding, W. Yan, T. Zhou, W. Shen, T. Wang, M. Li, S. Zhou, M. Wu, J. Dai, ecoenv.2021.112939.
K. Huang, et al., Endocrine disrupting chemicals impact on ovarian aging: [121] V.A. van der Weijden, V.L. Flöter, S.E. Ulbrich, Gestational oral low-dose
evidence from epidemiological and experimental evidence, Environ. Pollut. 305 estradiol-17β induces altered DNA methylation of CDKN2D and PSAT1 in
(2022), 119269, https://doi.org/10.1016/j.envpol.2022.119269. embryos and adult offspring, Sci. Rep. 8 (2018) 7494, https://doi.org/10.1038/
[100] Ş. Özel, A. Tokmak, O. Aykut, A. Aktulay, N. Hançerlioğulları, Y. Engin Ustun, s41598-018-25831-9.
Serum levels of phthalates and bisphenol-A in patients with primary ovarian [122] M. Banker, M. Puttabyatappa, P. O’Day, J.M. Goodrich, A.S. Kelley, S.E. Domino,
insufficiency, Gynecol. Endocrinol. 35 (2019) 364–367, https://doi.org/10.1080/ Y.R. Smith, D.C. Dolinoy, P.X.K. Song, R.J. Auchus, et al., Association of maternal-
09513590.2018.1534951. neonatal steroids with early pregnancy endocrine disrupting chemicals and
[101] E. Brehm, S. Rattan, L. Gao, J.A. Flaws, Prenatal exposure to Di(2-Ethylhexyl) pregnancy outcomes, J. Clin. Endocrinol. Metab. 106 (2021) 665–687, https://
phthalate causes long-term transgenerational effects on female reproduction in doi.org/10.1210/clinem/dgaa909.
mice, Endocrinology 159 (2018) 795–809, https://doi.org/10.1210/en.2017- [123] J.M.Y. Hu, T.E. Arbuckle, P. Janssen, B.P. Lanphear, L.H. Zhuang, J.M. Braun,
03004. A. Chen, L.C. McCandless, Prenatal exposure to endocrine disrupting chemical
[102] M.D. Patel, A. Lateef, H. Das, A.S. Patel, A.G. Patel, A.B. Joshi, Effect of age, sex mixtures and infant birth weight: a Bayesian analysis using kernel machine
and physiological stages on hematological indices of banni buffalo (Bubalus regression, Environ. Res. 195 (2021), 110749, https://doi.org/10.1016/j.
bubalis, Vet. World 9 (2016) 38–42, https://doi.org/10.14202/ envres.2021.110749.
vetworld.2016.38-42. [124] B. Vessa, B. Perlman, P.G. McGovern, S.S. Morelli, Endocrine disruptors and
[103] L.V. Tannenbaum, L. Gao, J.A. Flaws, Ovarian follicle resilience in mice orally female fertility: a review of pesticide and plasticizer effects,
dosed with methoxychlor: are reproductive impacts possible in mammals as S2666334122000356, FS Rep. (2022), https://doi.org/10.1016/j.
ecological receptors, Ecol. Indic. 106 (2019), 105502, https://doi.org/10.1016/j. xfre.2022.04.003.
ecolind.2019.105502. [125] E.V. Preston, V. Fruh, M.R. Quinn, M.R. Hacker, B.J. Wylie, K. O’Brien,
[104] Y. Liu, Y.-L. Wang, M.-H. Chen, Z. Zhang, B.-H. Xu, R. Liu, L. Xu, S.-W. He, F.-P. Li, S. Mahalingaiah, T. James-Todd, Endocrine disrupting chemical-associated hair
Z.-Q. Qi, et al., Methoxychlor exposure induces oxidative stress and affects mouse product use during pregnancy and gestational age at delivery: a pilot study,
oocyte meiotic maturation: M ETHOXYCHLOR A FFECTS M OUSE O OCYTE M Environ. Health 20 (2021) 86, https://doi.org/10.1186/s12940-021-00772-5.
ATURATION, Mol. Reprod. Dev. 83 (2016) 768–779, https://doi.org/10.1002/ [126] A.C. Saleh, R. Sabry, G.F. Mastromonaco, L.A. Favetta, BPA and BPS affect the
mrd.22683. expression of anti-mullerian hormone (AMH) and its receptor during bovine
[105] A.M. Neff, S.C. Blanco, J.A. Flaws, I.C. Bagchi, M.K. Bagchi, Chronic exposure of oocyte maturation and early embryo development, Reprod. Biol. Endocrinol. 19
mice to bisphenol-A alters uterine fibroblast growth factor signaling and leads to (2021) 119, https://doi.org/10.1186/s12958-021-00773-6.
aberrant epithelial proliferation, Endocrinology 160 (2019) 1234–1246, https:// [127] R. Zhou, W. Cheng, Y. Feng, W. Wang, F. Liang, F. Luo, S. Yang, Y. Wang,
doi.org/10.1210/en.2018-00872. Combined effects of BPA and PFOS on fetal cardiac development: in vitro and in
[106] D. Caserta, M.P. De Marco, A.R. Besharat, F. Costanzi, Endocrine disruptors and vivo experiments, Environ. Toxicol. Pharmacol. 80 (2020), 103434, https://doi.
endometrial cancer: molecular mechanisms of action and clinical implications, a org/10.1016/j.etap.2020.103434.
systematic review, IJMS 23 (2022) 2956, https://doi.org/10.3390/ [128] S. Ghimire, X. Zhang, J. Zhang, C. Wu, Use of chicken embryo model in toxicity
ijms23062956. studies of endocrine-disrupting chemicals and nanoparticles, Chem. Res. Toxicol.
[107] Y.-K. Leung, J. Biesiada, V. Govindarajah, J. Ying, A. Kendler, M. Medvedovic, S.- 35 (2022) 550–568, https://doi.org/10.1021/acs.chemrestox.1c00399.
M. Ho, Low-dose bisphenol A in a rat model of endometrial cancer: A CLARITY- [129] M.E. Street, S. Bernasconi, Endocrine-disrupting chemicals in human fetal growth,
BPA Study, Environ. Health Perspect. 128 (2020), 127005, https://doi.org/ IJMS 21 (2020) 1430, https://doi.org/10.3390/ijms21041430.
10.1289/EHP6875. [130] C. Viguié, E. Chaillou, V. Gayrard, N. Picard-Hagen, P.A. Fowler, Toward a better
[108] M.V. Bariani, R. Rangaswamy, H. Siblini, Q. Yang, A. Al-Hendy, A.R. Zota, The understanding of the effects of endocrine disrupting compounds on health:
role of endocrine-disrupting chemicals in uterine fibroid pathogenesis, Curr. human-relevant case studies from sheep models, Mol. Cell. Endocrinol. 505
Opin. Endocrinol., Diabetes Obes. 27 (2020) 380–387, https://doi.org/10.1097/ (2020), 110711, https://doi.org/10.1016/j.mce.2020.110711.
MED.0000000000000578. [131] J. Gingrich, E. Ticiani, A. Veiga-Lopez, Placenta disrupted: endocrine disrupting
[109] Y. Zhang, Y. Lu, H. Ma, Q. Xu, X. Wu, Combined exposure to multiple endocrine chemicals and pregnancy, Trends Endocrinol. Metab. 31 (2020) 508–524, https://
disruptors and uterine leiomyomata and endometriosis in US women, Front. doi.org/10.1016/j.tem.2020.03.003.
Endocrinol. 12 (2021), 726876, https://doi.org/10.3389/fendo.2021.726876. [132] W. Song, M. Puttabyatappa, L. Zeng, D. Vazquez, S. Pennathur, V. Padmanabhan,
Developmental programming: prenatal bisphenol A treatment disrupts mediators

14
A. Kowalczyk et al. Biomedicine & Pharmacotherapy 155 (2022) 113730

of placental function in sheep, Chemosphere 243 (2020), 125301, https://doi.


org/10.1016/j.chemosphere.2019.125301.

15

You might also like