Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Environmental Toxicology and Pharmacology 51 (2017) 56–70

Contents lists available at ScienceDirect

Environmental Toxicology and Pharmacology


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

Human exposure to endocrine disrupting chemicals: effects on the


male and female reproductive systems
Stavros Sifakis a , Vasilis P. Androutsopoulos b,∗ , Aristeidis M. Tsatsakis c ,
Demetrios A. Spandidos b
a
Department of Obstetrics and Gynecology, University of Crete, Medical School, Heraklion, GR 71003, Greece
b
Department of Clinical Virology, University of Crete, Medical School, Heraklion, GR 71003, Greece
c
Department of Toxicology, University of Crete, Medical School, Heraklion, GR 71003, Greece

a r t i c l e i n f o a b s t r a c t

Article history: Endocrine disrupting chemicals (EDCs) comprise a group of chemical compounds that have been exam-
Received 20 December 2016 ined extensively due to the potential harmful effects in the health of human populations. During the past
Received in revised form 22 February 2017 decades, particular focus has been given to the harmful effects of EDCs to the reproductive system. The
Accepted 26 February 2017
estimation of human exposure to EDCs can be broadly categorized into occupational and environmental
Available online 6 March 2017
exposure, and has been a major challenge due to the structural diversity of the chemicals that are derived
by many different sources at doses below the limit of detection used by conventional methodologies. Ani-
Keywords:
mal and in vitro studies have supported the conclusion that endocrine disrupting chemicals affect the
Endocrine disrupting chemicals
Reproductive system
hormone dependent pathways responsible for male and female gonadal development, either through
Exposure direct interaction with hormone receptors or via epigenetic and cell–cycle regulatory modes of action. In
Fertility human populations, the majority of the studies point towards an association between exposure to EDCs
Cancer and male and/or female reproduction system disorders, such as infertility, endometriosis, breast cancer,
Mechanism testicular cancer, poor sperm quality and/or function. Despite promising discoveries, a causal relation-
ship between the reproductive disorders and exposure to specific toxicants is yet to be established, due
to the complexity of the clinical protocols used, the degree of occupational or environmental exposure,
the determination of the variables measured and the sample size of the subjects examined. Future stud-
ies should focus on a uniform system of examining human populations with regard to the exposure to
specific EDCs and the direct effect on the reproductive system.
© 2017 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
2. EDCs and pesticide occupational and environmental exposure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3. Endocrine disrupting mechanism of action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
3.1. Enzyme and receptor-mediated mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
3.2. Epigenetic mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.3. Alternative mechanisms of action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.4. Metabolism of EDCs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
4. EDCs and female reproductive system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
5. EDCs and male reproductive system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
5.1. Semen quality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
5.2. Testicular cancer and cryptorchidism. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .64
5.3. EDCs modes of action on the male reproductive system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
5.4. EDCs, obesity, and male fertility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67

∗ Corresponding author.
E-mail address: androuts@uoc.gr (V.P. Androutsopoulos).

http://dx.doi.org/10.1016/j.etap.2017.02.024
1382-6689/© 2017 Elsevier B.V. All rights reserved.
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 57

6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67

1. Introduction

Endocrine disrupting chemicals (EDCs) are exogenous chem-


ical entities or mixtures of compounds that interfere with any
aspect of hormone action responsible for the maintenance of
homeostasis and the regulation of developmental processes. The
research conducted in the field of EDCs has increased consider-
ably over the last two decades, due to the potentially harmful
effects to the human body and the increasing knowledge in the
areas of developmental biology and environmental toxicology. It
is well known that compounds with endocrine disrupting mech-
anism of action can seriously affect human reproduction and
several studies have demonstrated a considerable decrease in fer-
tility biomarkers, notably sperm counts, in human populations
that have been exposed to EDCs (Safe, 2013; Slutsky et al., 1999;
Perry et al., 2007; Jouannet et al., 2001). The toxic effects of EDCs
have resulted in the restriction of their use in countries where
evidence of extensive exposure is ample (Knez, 2013). In some
westernized countries, the use of certain EDCs, such as polychlo-
rinated biphenyls or polybrominated diphenyl ethers has been
banned. However, in some cases the human exposure to EDCs is Fig. 1. Chemical structures of the main endocrine disrupting agents investigated in
inevitable, when such chemicals are used in the occupation or this study.
are widely dispersed across the environment. A specific example
includes the chemical class of pesticides. A pesticide is “any sub-
stance or a combination of substances used to prevent or eradicate 2. EDCs and pesticide occupational and environmental
unwanted insects, including vectors of diseases in human-beings exposure
and animals, weeds or fungi, in order to enhance food produc-
tion and aid production processing, storage, transport or marketing Organophosphate (OPs) pesticides are one of the most widely
of the food and agricultural commodities” (Sifakis et al., 2011; used class of pesticides for agricultural purposes (Koureas et al.,
Mehrpour et al., 2014; Abdollahi et al., 2004). Pesticides can act 2014). They are metabolized by xenobiotic metabolizing enzymes,
as EDCs, depending on their structure and their precise mecha- notably the cytochrome P450 (CYP) and the Paraoxonase (PON)
nism of action. Examples of pesticides with endocrine disrupting families of enzymes and are therefore not persistent in the envi-
properties include the organochlorines and the organophospho- ronment (Androutsopoulos et al., 2013). The exposure to OPs
rus pesticides. The exposure of human populations to pesticides is is assessed by the detection of their corresponding secondary
a matter of major concern, irrespective of their structural diver- metabolites notably the dialkyl phosphates in biological matrices
sity. It has been estimated that approximately 750,000 individuals such as urine (Androutsopoulos et al., 2013; Sokoloff et al., 2016;
are exposed annually to new forms of pesticides in the USA (Perry, Omoike et al., 2015). The exposure of humans to OPs is either
2008). The fate of pesticide chemicals depends on their metabolism through the occupation or the environment. Occupational expo-
by xenobiotic detoxification enzymes and their degradation to sure depends on a variety of factors, such as personal protective
smaller molecular weight compounds that may persist in the equipment (gloves, boots, and glasses), precaution practice (safety
environment. clothes and routine washing of hands), the type of job that deter-
The input of pesticides and EDCs on the reproductive system mines the frequency and extent of exposure (farmers, loaders, and
has been well defined. The adverse effects of the latter compounds applicators), specific training in the safe use and handling of pes-
as regards the health of the reproductive system include infertility, ticides and legislation (directives and laws to protect employees
cancer, cryptorchidism, decreased semen quality and hypospadias. from occupational toxins, periodic health surveillance) (Mehrpour
The major determinants of the toxic effects caused by pesticides et al., 2014). Dermal exposure is considered to be the main route for
and EDCs on the human reproductive system include the dose, occupational exposure and is defined as the transfer of the pesticide
the frequency of exposure, the route of exposure and the geno- from the surface of the foliage to the skin of the worker (Kapka-
typic characteristics of the exposed subjects (Hernández et al., Skrzypczak et al., 2011). The general quantitative levels for the
2013). In this review, a comprehensive synopsis of EDCs on the dermal exposure are difficult to estimate, due to the wide variabil-
human reproductive system is presented, with particular focus on ity in exposure parameters, such as the surface area exposed and
the class of pesticide chemicals and the commonly encountered the frequency of contact, although it has been shown that expo-
endocrine disrupting molecule Bisphenol A (BPA). The aspects of sure can occur in the presence of protective clothing, as the latter
endocrine disruption, with regard to reproductive disorders are could be the source of additional exposure (Jurewicz et al., 2009;
thoroughly addressed by evidence derived from in vitro, in vivo and Poet and McDougal, 2002). The activity of plasma cholinesterase
epidemiological studies. Information regarding EDCs and/or pes- has been used as a marker of chronic exposure to OP components
ticide exposure is also presented. The chemical structures of the (De Silva et al., 2006).
most common EDCs that are investigated in this study are outlined The second route of exposure to pesticides and EDCs involves
in Fig. 1. the environment. The factors that influence the extent of exposure
58 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

are namely, the type of chemical, the route of exposure, the timing response that is termed an agonistic effect, and/or a lack of hor-
and duration, the age, the gender and the genetic predisposition monal response that is termed an antagonistic action. Agonistic
(Mehrpour et al., 2014). For example, the US environmental pro- effects of MTX, an organochlorine pesticide used as an insecti-
tection agency (EPA) reference dose of the EDC BPA that is used as cide that was indented to replace DDT, have been reported for
a synthetic of plastic and epoxy resins is 50 ␮g/kg/day, although the estrogen receptor subtypes ER␣ and ER␤, whereas an oppo-
the actual daily human exposure may be much higher (Taylor site response was noted for the androgen receptor (Gaido et al.,
et al., 2011; Tzatzarakis et al., 2016). As regards organochlorine- 2000; Waters et al., 2001; Mrema et al., 2013). A similar anti-
type compounds this class includes environmental pollutants with androgenic effect has been noted for the environmental polluting
EDC-type of activity and organochlorine pesticides that are either chemical tetrachlorodibenzo-p-dioxin (TCDD) that has been shown
in use or have been banned for several years and are present in the to be an inhibitor or antagonist of hormone synthesis (Gore et al.,
environment due to the lack of metabolism and biodegradation. 2015). An additional example is the insecticide endosulfan that
Organochlorine compounds such as DDT and dioxins are not metab- has demonstrated estrogenic activity and has been shown to cause
olized by the human body and accumulate for a long period of time. ovarian regression in in vitro and in vivo models (Senthilkumaran,
In addition, such compounds appear to be a lot more persistent 2015). Endosulfan competes with estradiol (E2) for binding to
in the environment compared to organophosphorus compounds. the estrogen receptor, yet with lower affinity (Lemaire et al.,
Risk assessment studies have shown that risk quotient estimates 2006). This in turn affects gene expression of sex specific genes.
for heptachlor epoxide, endrin, dieldrin, DDT, and endosulfan pos- In contrast to the ovarian-mediated biological effects, endosulfan
sessed potential risk for aquatic organisms based on the observed caused the decrease of several male reproductive system associated
mean concentrations in the water sediments Clarias gariepinus genes such as testis-related transcription factors (Sox9a and wt1)
and Tilapia zilli from the Owan river (Ogbeide et al., 2015). The (Rajakumar et al., 2012), whereas BPA has been classified as a pro-
estimated average daily intake for the aforementioned organochlo- totypical non-steroidal estrogen receptor that possesses binding
rine pesticides was above their respective acceptable average daily affinity for the ER␣ and ER␤ receptors (Welshons et al., 2003).
intake (Ogbeide et al., 2015). The organochlorine exposure of DDT In addition to the hormone-related receptors, EDCs act on
and sumDDT-compounds and hexachlorobenzene was found to be enzymes involved in steroidogenesis and the metabolism of hor-
elevated in fish samples on St. Lawrence Island that was previously mones. For example phthalates, which constitute a particular class
used as a defense site, indicating that contamination of pesticides of pesticides, exert anti-androgenic effects by inhibiting the syn-
in the food chain is possible and that it can have a major impact thesis of testosterone in Leydig cells, as a result of direct CYP17
of exposure in human populations (Byrne et al., 2015). In contrast inhibition (Foster, 2005). Furthermore, EDCs have been shown to
to the aforementioned observations, the contamination of soil by inhibit the activity of 5-␣ reductase, which is one of the most impor-
DDT, DDD and DDE in the Xiangfen County in China was not an tant enzymes involved in the production of dihydrotestosterone
issue of concern as regards the long-term health impact for adults from testosterone and thus in the regulation of the masculin-
and children (Ma et al., 2016). This is possibly due to the low con- ization of the external gentalia and the prostate (Fisher, 2004).
centration of the organochlorine compounds that was estimated to Thiophosphates, a class of organophosphorous pesticides, inhibit
427.81 ng/g and is unlikely to affect significantly the health of the P450 enzymes namely, CYP3A4 and CYP1A2 that are involved in
exposed population (Ma et al., 2016). Thus, the exact contamina- the metabolism of estrone and testosterone in the liver (Usmani
tion of the environment and the consequent exposure of the human et al., 2003, Usmani et al., 2006).
populations to DDT-related compounds depend on the regional and In addition, EDCs have been shown to affect hormone receptor
geographical burden and require extensive risk-assessment and expression in animal models. Bisphenol A (BPA) caused dysregula-
monitoring studies regarding their bioaccumulation and environ- tion of steroid receptors and co-regulators in the rat testes, whereas
mental degradation. in utero exposure of Sprague–Dawley rats to diethylhexyl phthalate
The detection of pesticide exposure in several biological matri- (DEHP) resulted in increased mineralocorticoid receptor mRNA and
ces has been employed as a method to identify potential cases protein expression in adult interstitial Leydig cells (Skinner et al.,
of reproductive system disorders. The research work conducted 2011; Martinez–Arguelles et al., 2009).
by our group has been pioneering in the determination of expo- It is important to highlight that EDCs exhibit multiple hormone-
sure assessment of pesticides to human populations. The detection binding activities irrespective of binding to hormonal receptors. For
of Dialkyl phosphates (DAPs) and pyrethroid pesticides has been example, dichlorodiphenyltrichloroethane (DDT) is an agonist for
documented in meconium, maternal hair, infant hair, cord blood, the oestrogen receptor, whereas one of the metabolites of DDT is an
amniotic fluid and reproductive tissue samples of pregnant women anti-androgen (Kelce et al., 1995). BPA is a thyroid hormone antago-
and normal volunteers by HPLC/MS methodologies (Tsatsakis nist in addition to its estrogenic and androgenic activity (Moriyama
et al., 2009; Tsatsakis, 2008; Tsatsakis, 2006; Relakis et al., 1999; et al., 2002; Wetherill et al., 2005).
Koutroulakis et al., 2014; Koureas et al., 2012). In addition BPA As regards the effects of EDCs on the thyroid function, sev-
was detected in the hair samples of 69 Greek volunteers using the eral studies have indicated that EDCs interfere with TH and TSH
aforementioned assays (Tzatzarakis et al., 2015). signaling via a majority of pathways that result in alteration of
deiodinase activity, inhibition of TH excretion and/or metabolism,
blockage of iodine uptake by thyroid cells, competitive inhibi-
3. Endocrine disrupting mechanism of action tion of the thyroid transport protein TTR and antagonism of
complexes that originate from the thyroid hormone responsive
3.1. Enzyme and receptor-mediated mechanisms elements (TREs) (Noyes et al., 2013; Viluksela et al., 2004; Butt
and Stapleton, 2013; Nishimura et al., 2002; Meerts et al., 2002;
EDCs exert their toxicity by interfering with the normal Hamers et al., 2011). The structural similarity of TH with spe-
hormonal homeostatic mechanisms that promote growth and cific TH-EDCs namely, brominated flame retardants, hydroxylated
development of tissues. The classical action with respect to the polychlorinated biphenyls (PCBs) metabolites, and dioxin (PCDD)
reproductive system involves interference of EDCs with hormone results in binding with the high affinity TH transport protein TTR
binding to the corresponding receptor, notably the androgen recep- and consequently inhibition of T4-TTR binding (Grimm et al., 2013).
tor (AR) or the estrogen receptor (ER). Following binding to a The effects of EDCs are extended beyond the disruption of nor-
receptor the EDC can trigger two types of responses: a hormonal mal TH levels to the alteration of the lipid profile and glucose
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 59

homeostasis of the exposed individuals, since TH regulates lipid and et al., 1997), whereas when the animals were exposed during adult-
glucose metabolism. For example, BPA has been shown to cause hood to the same interval, no such pattern of methylation was
hyperlipidemia and increase abdominal adipose tissue in mice observed. The exact etiology was attributed to hypomethylation
(Miyawaki et al., 2007). Finally, certain TH-EDCs further interfere of the exon-4 of the fos gene that was discovered in subsequent
with the actions of hormones that act via nuclear receptors such as studies (Li et al., 2003). DES is further associated with hypermeth-
sex hormones (estrogen, progesterone, and androgen) and/or inter- ylation in the promoter and intron regions of the Hoxa-10 gene
act with their corresponding nuclear receptors (ER, PR, and AR). that in turn induces anterior transformations of the reproductive
At the molecular level, EDCs can affect the expression of tract (Block et al., 2000). Importantly, the interference of EDCs to
steroid and sex hormone related enzymes by inducing their corre- the epigenetic regulation of several genes has been associated with
sponding transcription, via binding to nuclear receptors. Notably, additional cell signaling pathways that act upstream, including the
organochlorine pesticides and dioxins have been documented to ER and the PI3 K/Akt pathways (Bredfeldt et al., 2010; Falck and
bind with considerable potency to the aryl hydrocarbon recep- Forsberg, 1996).
tor (AhR) that induces the expression of CYP1 genes that in turn The epigenetic changes in the ovary have been documented
metabolizes estradiol (E2) to hydroxylated derivatives (Park et al., for the organochlorine pesticide methoxychlor (MTX). The use of
2017). TCDD can further interfere with ovarian steroidogenesis and bisulfite-sequencing PCR and methylation-specific PCR has demon-
previous studies have shown that it can decrease progesterone, strated that MTX caused hypermethylation in multiple CpGs in
androstenedione, testosterone and E2 levels in vitro and in vivo the ER␤ promoter sequences in a study conducted by Zama and
(Karman et al., 2012a,b; Chen et al., 2009). The organochlorine pes- Uzumcu in 2009 (Zama and Uzumcu, 2009). The extent of DNA
ticide MTX can inhibit ovarian steroidogenesis and consequently methylation in the promoter regions appears to be age-dependent,
the production of E2, testosterone and androstenedione by the as the aforementioned genes reveal a different methylation pattern
downregulation of the expression of CYP19␣1, CYP17␣1, CYP11␣1 in neonatal models of ovaries following exposure to EDCs (MTX,
and 17␤-hydroxysteroid dehydrogenase enzymes (Basavarajappa DES) (Zama and Uzumcu, 2009; Tang et al., 2008). With regard
et al., 2011). A similar anti-estrogenic effect has been noted for the to the gene targets that are methylated by MTX, genome-wide
MTX metabolite 2,2-bis(p-hydroxyphenyl)-1,1, 1-trichloroethane methylation analyses have indicated that the majority of candi-
(HPTE) that can reduce the FSH-stimulated synthesis of proges- date genomic regions include transcription factors and ribosomal
terone and estrogen by decreasing the expression of CYP11␣1 and proteins. Specifically targets such as PTEN and IGF have been iden-
CYP19␣1 (Zachow and Uzumcu, 2006). tified as critical signaling pathways that are hypermethylated by
The induction of endogenous-compound-metabolizing and/or MTX (Zama and Uzumcu, 2009).
xenobiotic-metabolizing enzymes has been reported to occur by
several EDCs and affects their metabolic fate in the human body. For 3.3. Alternative mechanisms of action
example, the induction of the AhR and the E2 metabolizing enzyme
CYP1B1 has been documented to occur for the EDC class of phtha- Several other modes of action of EDCs as regards the disruption
lates (Zachow and Uzumcu, 2006). As in the cases of MTX and TCDD of the normal reproductive system development have been pro-
phthalates decrease the levels of E2 and DEHP in vivo as a result of posed that are not directly related to the regulation of hormonal
an additional downregulation of CYP19a1 expression (Gupta et al., function. For example, mono-(2ethyl-hexyl-phthalate) (MEHP) has
2010; Hannon et al., 2015). been shown to affect ovarian follicle health by inducing the gen-
eration of reactive oxygen species and by increasing the level of
3.2. Epigenetic mechanisms oxidative stress, while concomitantly disrupting the expression of
the enzymes superoxide dismutase 1 (SOD1) and glutathione per-
The epigenetic effects of EDCs refer to heritable changes in gene oxidase (GPX) (Wang et al., 2012). The disruption of the normal cell
function in the absence of DNA sequence alterations. Notably, epi- cycle control has also been documented for the phthalate dibutyl
genetic effects are mediated by transcription factors that repress phthalate (DBP) that inhibits the expression of cyclin E1, cyclin A2
or enhance the transcription of specific genes. The main epige- and cyclin B1 and results in cell cycle arrest and subsequent inhibi-
netic mechanisms include DNA methylation, post-translational tion of antral follicle growth (Craig et al., 2013). In addition, BPA was
modifications of histone proteins (histone acetylation and histone shown to induce apoptosis, via a caspase 3-mediated mechanism
deacetylation) and non-coding RNA (Jirtle and Skinner, 2007). DNA in female rat follicles and to regress the luteum development that
methylation usually leads to a reduction of gene expression, since is considered as the main cause of infertility (Lee et al., 2013). Fig. 2
it affects the binding of transcription factors to the DNA. Post- represents a summary of the modes of EDCs action with regard to
translational modifications of the histone proteins at specific amino the adverse effects caused in the reproductive system.
acid residues, such as lysine may alter the structure and function
of chromatin (Turner, 2009). 3.4. Metabolism of EDCs
Generally it is accepted that acetylation of histones results
in the activation of transcription as a result of the relaxation of The in vitro metabolism of EDCs is considered a major factor
chromatin, whereas deacetylation results in the silencing of genes that contributes to their corresponding adverse effects. Gener-
and transcriptional repression. Non-coding RNAs are transcripts of ally organochlorine compounds such as dioxin and DDT are not
sequences that do not code for proteins but regulate the expression metabolized readily due to the extensive number of chlorine
of genes in a cis and trans manner (Chang et al., 2006). They are atoms in their structure that remain unconjugated. Consequently,
involved in specific functions such as X-chromosome inactivation, such compounds exhibit a considerably long half-life in the
genomic imprinting and developmental patterning and differenti- human body that contributes to the continuous stimulation of
ation. the adverse effects (Androutsopoulos et al., 2013). Organochlo-
Studies in laboratory animals have demonstrated that EDCs such rine compounds including environmental pollutants and pesticides
as diethylstilbestrol (DES) can activate the expression of immedi- can induce the expression of phase I and II metabolizing enzymes
ately early genes in neonatal development, such as c-fos, c-jun, notably CYP1A1, CYP1B1, UGT1A6 and NQQ1 via the Aryl hydro-
c-myc and lactoferrin that are upregulated in childhood (Nelson carbon receptor (AhR) (Androutsopoulos et al., 2009). This in
et al., 1994). This effect was accompanied by hypomethylation of turn facilitates metabolism of polycyclic aromatic hydrocarbons,
the promoter region of the lactoferrin gene in the adult uterus (Li nitrosamines and various environmental chemicals to biologically
60 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

Fig. 2. The molecular mechanism of action of EDCs with regard to the disorders of the reproductive system. (A) Enzyme inhibition and enzyme up/down–regulation. Pesticides,
such as methoxychlor can down–regulate the expression of steroid metabolizing enzymes via antagonism with the estrogen receptor (ER). Environmental pollutants, such as
TCDD can induce the expression of steroid and carcinogen–metabolizing enzymes via the Aryl hydrocarbon receptor (AhR). (B) The epigenetic effects of EDCs. Methoxychlor
has been shown to hypermethylate the promoter of the ER␤ receptor and IGF and PTEN sequences. (C) The effects of EDCs on the antral follicle cell cycle. BPA has been reported
to down–regulate the expression of cyclin E1, cyclin A2 and cyclin B1 and results in cell cycle arrest and consequently apoptosis via a caspase–3 mediated mechanism.

active metabolites (Androutsopoulos et al., 2009). With regard to the corresponding oxon metabolite that is in turn hydrolyzed by
to the EDC methoxychlor, it has been shown that the active PON1 to DAP (Androutsopoulos et al., 2013). The metabolites of OPs
metabolite hydroxyphenyltrichloroethane exerts approximately retain a part of the activity of the parent compound, although the
100-fold greater estrogenic activity and potency in the target majority of the studies in human populations have focused notably
organs of animals exposed to methoxychlor compared with the on the detection of OPs metabolites rather than the evaluation of
parent compound (Aoyama and Chapin, 2014). Methoxychlor their corresponding biological activity.
mainly undergoes demethylation reactions and hydroxylation by
cytochrome P450, such as CYP3A4, CYP3A5 and CYP2B6. In contrast 4. EDCs and female reproductive system
to MTX, BPA has been shown to be notably metabolized via sulfa-
tion and glucuronidation reactions to the metabolites Bisphenol The main biological adverse effects of EDCs with regard to the
A-4-ulfate and Bisphenol A-4D-glucuronide, respectively that do development of the reproductive system are attributed to folliculo-
not possess activity on endocrine receptors (Gramec Skledar et al., genesis. The primordial follicles evolve to primary, pre-antral and
2016). Hydroxylation reactions have further been documented for antral follicles. EDCs, such as BPA, MTX, TCDD, and phthalates can
BPA by phase I enzymes. The metabolite 5-hydroxybisphenol A interfere with the development of the aforementioned types of fol-
is considered to be less potent than the parent compound (Elsby licles (Fig. 3A). Toxicity caused to the antral follicles by EDCs can
et al., 2001). The metabolism of phthalates follows a similar path- lead to infertility. BPA has been associated with female fertility
way to that of BPA with glucuronidation reactions demonstrating problems, polycystic ovary syndrome and endometriosis, whereas
a key process in the detoxification of the compound activity, in women undergoing fertility treatments BPA levels have been
whereas organophosphorus pesticides are notably metabolized to associated with decreased antral follicle counts and a reduction
the common metabolite dialkyl phosphate (DAP) via the enzyme in the number of oocytes (Kandaraki et al., 2011; Caserta et al.,
Paraoxonase 1 (PON1). An initial desulfuration reaction occurs by 2014; Souter et al., 2013). The results reported in human popu-
cytochrome P450 enzymes namely, CYP2B6, CYP2C19 and CYP3A4 lations have been verified by experimental animal studies, where
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 61

Fig. 3. The pathophysiological and biochemical mechanism of action of EDCs with regard to the disorders of the reproductive system. (A) The effects of EDCs on folliculogenesis.
The growth of primordial follicles to antral follicles is affected by exposure to EDCs. The antral follicle is responsible for the ovulation and the production of sex steroid
hormones. The exposure to EDCs can lead to infertility and impaired production of steroid hormones. (B) The life stages of the individuals exposed to EDCs are a major
factor that determines the extent of the adverse effects. Notably severe health disorders are initiated at the early stages of life (infancy, childhood) that manifest following
a considerably period of time (adulthood) to specific symptoms and diseases. (C) Biochemical modes of EDCs action with regard to infertility and adverse effects on the
female reproductive system. Inhibition of aromatase by EDCs leads to reduced plasma E2 levels, impaired oocyte development and consequently infertility. (D) Inhibition
of 5␣–reductase by EDCs interferes with the metabolism of testosterone to dihydrotestosterone (DHT) in the male reproductive system and in turn with the binding and
activation of the androgen receptor(AR)-DHT complex, resulting in impairment in the transcription of the corresponding AR target genes.

the latter compound was shown to reduce the primordial follicle coworkers used a very high concentration range of 1–500 mg/kg of
pool and lead to premature ovarian failure (Wang et al., 2014). methoxychlor (MTX) in postnatal female rats in order to demon-
Similarly, MTX inhibits folliculogenesis in vivo by increasing the strate reduction in ovarian weight by 50, 100, and 500 mg/kg of
expression of the anti-Müllerian hormone in the pre-antral and MTX treatment (Uzumcu et al., 2006). In contrast to the aforemen-
early antral follicles (Uzumcu et al., 2006), whereas TCDD pos- tioned studies, Wang et al., reported doses of exposure of 0.5, 20,
sesses anti-proliferative effects on the rat ovary and can interfere and 50 ␮g/kg of BPA in newborn FVB mice in order to demonstrated
with ovarian steroidogenesis (Heimler et al., 1998; Karman et al., fertility reduction as an adverse effect of BPA (Wang et al., 2014).
2012a,b). An increase in the atretic follicles and a decrease in the Although the doses used in the latter study are similar with the
primary and secondary follicle numbers have been noted in rats exposure to BPA noted in human populations (2.78 ng/ml in urine,
following 600 mg/kg of exposure to the phthalate DHEP for 60 con- 1.6 ␮g/L in serum), the majority of the studies conducted in humans
secutive days (Xu et al., 2010). comprised adult and not newborn subjects. Taken together, the
Although the toxic effects of EDCs in female reproduction in findings derived by the aforementioned studies suggested that the
experimental animals are apparent, in human populations this experimental dose of exposure and the selection of the target group
association is not directly related to causality, as some studies have under investigation were major determinants of the “adversity” of
indicated positive associations and others no associations of EDC EDCs.
exposure and female reproductive system disorders (Rochester, In addition, research in humans and EDCs is less in quantity,
2013; Ehrlich et al., 2012a,b; Yang et al., 2009; Philippat et al., compared to animal and in vitro studies (Rochester, 2013). For
2012). This apparent contradiction occurs mainly due to the sever- example BPA has been studied in humans with regard to the
ity of exposure that determines the actual dose of daily intake. It adverse health outcomes on the reproductive system by various
is a lot more difficult to determine the environmental exposure research groups during the period of 2002 and 2012 and it was
to EDCs and the dose of human exposure, compared to laboratory found that higher BPA exposure, as demonstrated by higher BPA
investigations where the conditions and notably the dose treat- concentration in plasma or urine, was associated with poorer ovar-
ment are adequately controlled. Consequently, the differences that ian response, reduced number of mature oocytes, reduced number
are noted among studies with regard to the biological effects of of fertilized oocytes, lower peak E2 in response to hyperstimulation
EDCs are notably attributed to the different experimental condi- with human chorionic gonadotrophin (hCG), decreased probability
tions and study protocols that define the dose of exposure. Although of fertilization, higher implantation failure and infertility (Ehrlich
the mechanism of EDCs action can be adequately demonstrated et al., 2012a,b; Mok-Lin et al., 2010; Bloom et al., 2011; Fujimoto
in animal models, the dose of exposure used is frequently super- et al., 2011; Ehrlich et al., 2012b; Caserta et al., 2013) (Table 1).
ficial compared with the actual exposure that occurs in human As regards the concentration of the sex hormone prolactin, it was
populations. For example, mineralocorticoid receptor dysfunction found that it was significantly higher in BPA exposed women vs.
was demonstrated in rats, following exposure to DEHP at a dose non-exposed women in an occupational case control study of 155
range of 100 to 950 mg from gestational day 14 to gestational subjects (Hao et al., 2011;), whereas BPA exposure has been further
day 19 (Martinez–Arguelles et al., 2009). This dose is consider- linked to polycystic ovary syndrome as documented by a cross-
ably greater than the actual exposure of humans to DEHP, whereas sectional study in 171 women (Kandaraki et al., 2011).
the life stage of animal exposure corresponds to early childhood In terms of breast cancer development and BPA, non-significant
in humans, which will in theory deteriorate the adverse effects associations have been reported by the studies of Yang et al. and
caused compared with the later stages of life. Similarly, Uzumcu and Aschengrau et al. (Yang et al., 2009; Aschengrau et al., 1998), while
62 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

Table 1
A summary of the studies that have assessed the associations between the exposure of human populations to pesticides and EDC agents and the incidence of disorders
affecting the female reproductive system. (CS: cross sectional study; CC: case-control study; CHS: Cohort study).

Authors, Year Chemical N Results Variables Study Type


Kandaraki et al., BPA 171 (100 PCOS, 71 Normal) Higher BPA levels in PCOS, Hormonal parameters, BMI CS
2011 association of BPA with
androgens
Caserta et al., 2013 BPA, phthalates 61 women Higher BPA and receptor Urinary BPA, EDC levels in CS
levels in infertile group serum, oestrogen and
nuclear receptors
Souter et al., 2013 BPA 209 women on fertility Higher BPA levels Urinary BPA, Antral Follicle CHS
treatment associated with lower AFC count (AFC), FSH
Ehrlich et al., BPA 147 women with IVF cycles Negative association Urinary BPA, oocyte CHS
2012a,b between urinary BPA and maturation, fertilization,
serum peak E2 and oocyte embryo quality
yield
Ehrlich et al., BPA 137 women Positive association Urinary BPA, ␤-hCG, CHS
2012a,b between urinary BPA and implantation failure
implantation failure
Mok-Lin et al., 2010 BPA 84 women Urinary BPA is inversely Urinary BPA, E2 levels, CHS
correlatedwith serum peak number of oocytes
E2
Bloom et al., 2011 BPA 44 women with IVF Serum BPA is associated Serum BPA, E2 levels, CHS
with reduced E2 levels number of oocytes
Fujimoto et al., BPA 58 infertile females, 37 Inverse association Serum BPA, fertilization CHS
2011 male partners between BPA and
fertilization
Yang et al., 2009 BPA 167 women with breast No association with BPA Blood BPA, incidence of CC
cancer, 152 controls and breast cancer breast cancer
Aschengrau et al., BPA, BHA, benzyl 261 women with breast Association with PCBs, Levels of EDCs and CC
1998 phthalate, nonylphenol cancer, 753 controls 4-octylphenol and breast pesticides and incidence of
cancer breast cancer
Philippat et al., BPA, phenols, 288 mother-newborn pairs Positive/Negative Levels of EDC and CC
2012 phthalates (72 cases, 216 controls) association of pesticides, birth weight,
benzophenone-3/2,5 DCP head circumference, birth
with male birth weight, length
respectively
Sugiura–Ogasawara BPA 77 women (45 cases with Serum BPA is associated Serum BPA, ANA, NK cells, CC
et al., 2005 miscarriages, 32 controls) with recurrent miscarriage prolactin, progesterone,
TSH, fT4
Buck Louis et al., BPA, phthalates 626 women mECPP, mEHHP, mEOHP, BPA, 14 phthalate CHS
2013 mEHP, mBP and mCMHP metabolites,
were associated with endometriosis, age, BMI,
endometriosis creatinine
Raanan et al., 2015 DAP, DE, DM 359 mothers and children Early life exposure (1–7 Urinary DAP, DE, DM and CHS
years) to DAP, DE, DM is metabolites, respiratory
associated with respiratory symptoms in childhood
symptoms
Mahalingajah et al., HCB, DDT, DDE 720 women with IVF Serum HCB is associated Serum HCB, DDT, DDE and CHS
2012 with failed implantation metabolites, fertilization

the latter chemical has been linked with developmental and birth populations examined. A case control study that was published by
weight abnormalities of infants, premature delivery and miscar- Caserta and colleagues revealed that serum BPA was detected in a
riages (Sugiura–Ogasawara et al., 2005; Philippat et al., 2012). The significantly higher number of women, compared with controls in
results of the aforementioned studies are indicative of the severe a sample size of 61 participants, whereas the presence of specific
adversity of BPA on the female reproductive system. In contrast to phthalate metabolites and not BPA in urine were associated with
the observations by Sugiura-Ogasawa et al., Cantorinine et al., and the diagnosis of endometriosis in a study that comprised 14 clini-
Phillipant and coworkers, the major disadvantage encountered by cal centers in the US. (Caserta et al., 2013; Buck Louis et al., 2013).
such studies was the small number of subjects that were analyzed. Overall, the data in the literature support that there is some evi-
With the exception of the study conducted by Aschengrau et al., the dence that BPA may contribute to infertility in humans (Rochester,
aforementioned studies comprised a sample size of less than 200 2013).
participants. This indicates that more work is required to confirm Similar effects with those noted in the case of BPA in humans
these findings. As regards infertility, the studies were not directly have been reported for a variety of other EDCs. For exam-
related to the general population, since infertile couples and/or cou- ple epidemiological evidence has supported the conclusion that
ples undergoing IVF appeared more sensitive to BPA based on the there is a strong association between developmental exposure to
endpoints: beta-hCG, embryo quality and decreased probability of organochlorine pesticides and subsequent female fertility prob-
fertilization (Bloom et al., 2011; Chen et al., 2013; Ehrlich et al., lems (Zama and Uzumcu, 2009; Zama and Uzumcu, 2010). Although
2012a,b). However, approximately 1/3 of these women had infer- phthalates have been extensively studied for their effects in male
tility diagnoses due to the female factor, 1/3 due to the male factor reproduction, there is accumulating evidence that they exert addi-
and 1/3 had unexplained infertility (Bloom et al., 2011; Chen et al., tional adverse effects in the female reproductive system. The
2013; Ehrlich et al., 2012a,b; Ehrlich et al., 2012b; Mok-Lin et al., most commonly studied phthalates are di-(2-ethyl-hexyl) phtha-
2010; Fujimoto et al., 2011). This suggests that neither male, nor late and the metabolite mono-(2-ethyl-hexyl) phthalate (MEHP)
female infertility can alone explain the sensitivity to BPA in the (Craig et al., 2011). The levels of phthalates have been associated
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 63

with significantly higher risk of implantation failure in women et al., 2012). The latter adverse effects along with the decrease in
undergoing infertility treatments (Hauser et al., 2016). As regards the quality of semen share similar pathophysiological patterns and
DDT and dioxin-type compounds, the evidence in humans is lesser, are important risk factors for the development of the male fertility
since these molecules have been banned for more than 30 years disorders.
worldwide. The levels of HCB, DDT and DDE were examined in
the serum of 720 women undergoing in vitro fertilization (IVF) and 5.1. Semen quality
the HCB concentrations were associated with failed implantation
compared with DDE and DDT, where no significant difference was The quality of semen has been investigated as an important vari-
noted (Mahalingajah et al., 2012). The results of the clinical studies able for the determination of the adverse effects caused by EDCs.
conducted with regard to the association of EDCs with the female However, initial studies that were conducted in early 1980s were
reproductive system are summarized in Table 1. observational and did not aim at investigating the contribution of
Accumulating evidence suggests a potential association with EDC exposure on the developmental defects, with regard to the
OPs and the disruption of normal female reproductive functions. male reproductive system. The initial hypothesis was to explore
Raanan and coworkers reported higher prenatal DAP concen- the cause of a deteriorating semen quality in specific human popu-
trations with the occurrence of respiratory symptoms in early lations. A meta-analysis conducted by Carlsen (Carlsen et al., 1992)
childhood during the period of 0.5–5 years of age (Raanan et al., demonstrated a significant decline in mean sperm concentrations
2015). A similar study in China that comprised 249 pregnant from 113 to 66 million/ml. During that period, additional confound-
women indicated that higher levels of DAPs and their metabolites ing factors, such as the period of abstinence, the methodological
in the urine of the women was the predominant risk factor for aspects of semen analysis, the age and the fertility were of con-
neonatal neurobehavioral development (Zhang et al., 2014). With sideration regarding the decrease in semen quality, although they
regard to the association of the female reproductive dysfunction were not proven sufficient for a definitive conclusion (Jouannet
and infertility with the exposure to OPs, the majority of the data in et al., 2001). In addition, this trend could have been the outcome
the literature has been derived by in vivo and experimental studies. of geographical variation. The follow-up study that was carried out
Evidence regarding the impairment of fetal growth and develop- by Swan and coworkers confirmed that the afore mentioned find-
ment caused by prenatal exposure to OPs in humans has been ings were not the result of bias, as the confounding factors were
associated with the activity of the paraoxonase 1 enzyme (PON1) accordingly adjusted and an overall decline in the sperm concen-
in the fetus and during early childhood. This process increases the tration was noted annually at 1.5 and 3.1% of the population in the
vulnerability of the fetus and child to OP poisoning and suggests USA and Europe, respectively (Swan et al., 1997). The meta-analysis
that the exposure to OPs has a greater impact on the fetal and that was conducted by Swan did however confirm that sperm qual-
infant growth compared with the respective exposure of the adults ity trends could be affected by the variation in the geographical
to the same concentration of pesticides (Fig. 3B) (Peiris–John and regions (Swan et al., 2000). Furthermore, studies that were con-
Wickremasinghe, 2008). ducted in North Europe and particularly Denmark have provided
Overall, the data reported in the literature in human popula- supportive evidence that semen quality can be largely affected by
tions are suggestive of an association between EDCs and disorders the location of the human population that is examined (Jørgensen
in the female reproductive system. Several discrepancies between et al., 2001; Jørgensen et al., 2002). Danish men were reported
studies are attributed to the complexity of the protocols, the degree to have the lowest semen quality, followed by Norwegian, Scot-
of occupational or environmental exposure and the sample size of tish and French, whereas the Finnish population had the greatest
the subjects examined. The timing of exposure is of great impor- (Jørgensen et al., 2001; Jørgensen et al., 2002). In contrast to the
tance since the same dosage exerts diverse effects in individuals of early observations by Jorgensen and colleagues in 2001, subsequent
different age. Furthermore, the exposure during critical develop- studies that were conducted in Finland revealed a discouraging
mental phases (intrauterine life, neonatal period, and childhood) temporal decrease in semen quality in men from the general pop-
is considered to affect the reproductive system greater compared ulation in the period between 1998 and 2006 (Jørgensen et al.,
with the adulthood (Fig. 3B). The effects noted may be the result of 2011; Jørgensen et al., 2012). The data derived by the studies of
synergism and/or antagonism, since exposure is determined by a Jorgensen and similar researchers suggest that a large proportion
mixture of toxicants. Furthermore, the results may appear imme- of young men from the general population (approximately 43%)
diately, and/or they may manifest later in life and in the following may exhibit suboptimal sperm counts (less than 40 million/ml)
generations, with different susceptibility, depending on the genetic (Andersson et al., 2008; Jørgensen et al., 2012; Jørgensen et al.,
polymorphisms of the exposed individuals. With regard to the 2011). The aforementioned epidemiological studies suggest that
mechanism involved, the contribution of aromatase inhibition by the different geographical locations of the human population res-
EDCs to the development of impaired oocytes is one of the main idency, cannot be the sole contributing factor for the decrease in
factors responsible for infertility (Fig. 3C). the sperm count. Thus, alternative environmental factors have to
be considered for the increased incidence of poor semen quality in
specific human populations of North Europe.
5. EDCs and male reproductive system Recent studies that aimed at investigating the association of
the exposure to pesticides and the sperm quality have corrobo-
The adverse effects caused by EDCs in the male reproductive sys- rated the findings by Jørgensen and colleagues. The occupational
tem have been a matter of increasing alert during the last decades. exposure to pesticides increases the risk of morphological abnor-
Although several studies from the early 1940s have proposed the malities in the sperm of farm workers that includes a decline in
decline in semen quality as the main adverse effect caused by EDCs sperm count and a decreased percentage of viable sperms. OP pes-
on the male reproductive system, the diverging results from the ticides such as parathion and methyl parathion can decrease the
different study designs were not able to suggest a definitive conclu- concentration of the sperm by damaging the seminiferous epithe-
sion regarding the dose, the magnitude of exposure and the type of lium, while it has been suggested that pesticide exposure affects sex
chemical that contributed to the occurrence of the adverse effects. accessory glands that may also reduce the seminal volume (Perry
Similar trends of adverse effects on the male reproductive system et al., 2011; Yucra et al., 2006). The exposure to pesticides reduces
have been reported, such as the incidence of cryptorchidism and the seminal volume, increases the seminal pH and increases the
testicular cancer that may be related to EDC exposure (Nordkap abnormal sperm head morphology (Yucra et al., 2006). In addition,
64 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

Lifeng et al. demonstrated that sperm motility could be affected The urinary levels of BPA have been associated with lower sperm
by a limited number of pyrethroid pesticides, such as fenvalerate quality parameters (Li et al., 2011) in men that were occupationally
(Lifeng et al., 2006). Specific adverse effects on the male repro- exposed to this chemical. Of note is that the median urine con-
ductive system have been reported to occur by organochlorine centration estimated was approximately 70 times lower than the
pesticides, such as endosulfan and DDT due to the disruption of the accepted daily intake according to the USEPA, whereas the control
hypothalamic–pituitary testes axis and the direct interaction with group exhibited exposure doses of approximately 2,000 times less
the sex steroid receptors in the target tissue (Mehrpour et al., 2014). than the accepted daily intake. This finding highlighted that BPA can
As regards organophosphate pesticides, the exposure of male farm- cause harmful effects at doses much lower than the USEPA accepted
ers from three different communities in Malaysia to malathion daily intake (Li et al., 2011). BPA was further shown to be associ-
and/or paraquat resulted in significantly lower values of sperm ated with lower sperm count; sperm morphology, sperm motion
concentration, pH, mean volumes motility of sperm cells, com- and DNA damage (Meeker et al., 2010). As regards hormonal levels,
pared with the non-exposed group (Hossain et al., 2010). The results BPA was associated with higher FSH, lower inhibin B and a lower
have been supported by experimental animal studies where both oestradiol: testosterone ratio (Meeker et al., 2010). BPA was further
malathion and parathion were shown to lower the body weights, associated with a decreased free androgen-index and higher testos-
the reproductive organ weights and the sperm counts of adult rats terone in men in a study that comprised 375 fertile men (Mendiola
(Narayana et al., 2006; Geng et al., 2015). The mechanism of action et al., 2010).
was attributed to the induction of spermatogenic apoptosis via the Similar data have been published for PCBs, which is a struc-
modulation of the expression of the proteins Bax and Bcl-2 and turally related class of environmental pollutants. In the study
the concomitant reduction in the levels of the reproductive hor- conducted by Petersen and colleagues, 266 fertile men were exam-
mones LH, FSH and testosterone (Geng et al., 2015). The indirect ined with regard to the exposure to PCBs, hormone concentration
modes of pesticide action involve the interference with the neu- and semen quality (Petersen et al., 2015). The ratio of testos-
roendocrine control at the testicular level that results in the altered terone/oestradiol and the levels of FSH and sex hormone binding
production and release of testosterone, the excess production of globulin (SHBG) were positively correlated with serum PCB lev-
free radicals and the induction of oxidative stress (Talamanca et al., els, whereas no association was noted between the latter and
2001; Abdollahi et al., 2004). A direct interaction with regard to the semen quality variables, such as semen volume, sperm con-
the modulation of the testosterone/oestradiol hormonal concen- centration, total sperm count, sperm morphology and period of
tration has been noted for the class of polychlorinated biphenyls in abstinence (Petersen et al., 2015). The results of the clinical stud-
266 fertile men, indicating that structurally similar compounds to ies conducted with regard to the association of EDCs with the male
the class of organochlorine pesticides that possess endocrine dis- reproductive system are summarized in Table 2.
rupting activity can affect the male reproductive system (Petersen
et al., 2015). 5.2. Testicular cancer and cryptorchidism
The adverse effects of phthalates on sperm quality were con-
firmed by ex vivo studies, where spermatozoa were incubated in The incidence of testicular cancer has notably been investigated
vitro and exposed with high concentrations of phthalates (Pant in cohorts located in Denmark and Norway, where it was shown
et al., 2011). It was reported that the sperm motility was decreased that approximately 1% of men will develop testicular cancer during
and that cytotoxicity was caused at long-term exposures (> 3 days) their life (Skakkebaek et al., 2007). It is important to note that during
of human semen samples to the metabolite DEHP (Pant et al., 2011). the period between 1998 and 2002 the incidence of testicular can-
In parallel DHEP has been shown to inhibit testosterone production, cer in men in Norway was approximately 5-fold higher compared
when cultured in vitro with explants derived from human testes with the corresponding incidence in Spain (Chia et al., 2010; Llanes
(Desdoits–Lethimonier et al., 2012). et al., 2008). This finding suggests that the incidence may be higher
Studies that were conducted in human populations corrobo- in developed rather than developing countries. Such differences
rated the in vitro findings and suggested that exposure to phthalate were attributed initially to the heterogeneity of the populations
metabolites is correlated with lower motility of spermatozoa in examined, with regard to the ethnic origin and the immigration
men from subfertile couples (Duty et al., 2003). The DNA damage frequency from countries with a high morbidity rate of this can-
induced in spermatozoa, the sperm motility and the morphology cer (Nordkap et al., 2012). The prevalence of cryptorchidism has
of the spermatozoa were weakly associated with the exposure to received a considerable increase in Denmark during the period
phthalate (Hauser et al., 2007; Duty et al., 2004; Hauser et al., of 1997–2001 (Boisen et al., 2004), whereas a lower incidence
2006; Liu et al., 2012), whereas with regard to the disruption of the was observed in England and Finland, respectively (Boisen et al.,
hormonal function, an inverse association between MEHP metabo- 2004; Boisen et al., 2005). Such geographic trends were followed
lite increase and testosterone and oestradiol levels was reported by specialized studies where it was shown that maternal-lifestyle
(Meeker et al., 2009). factors, such as the increased intake of pain killers, notably para-
In addition to the phthalates, the human exposure to the EDC cetamol and ibuprofen might be associated with cryptorchidism in
BPA affects the concentration of testosterone in plasma in men the offsprings (Jensen et al., 2007; Thorup et al., 2006; Stoorgaard
and women, whereas the hormones acting upstream such as FSH et al., 2003). It has been suggested that the exposure to environmen-
are downregulated, possibly due to the severity of the endocrine tal chemicals, such as phthalates that results in poor semen quality
disrupting action. Mendiola and colleagues investigated the expo- may coincide with the development of testicular cancer, cryptor-
sure of 360 fertile men to BPA and highlighted that the levels of chidism and hypospadias (Nordkap et al., 2012). The “so called”
the compound were inversely correlated with the concentration of testicular dysgenesis syndrome hypothesis is based on the combi-
indices of androgens, such as testosterone (Mendiola et al., 2010). In nation of clinical observations with the aforementioned 3 disorders
contrast to the findings by Mendiola et al., non-significant trends of the male reproductive system. Particularly the link between tes-
between urinary BPA levels and oestradiol/testosterone index as ticular cancer and impaired semen quality has been proposed as
well as lower sperm counts and DNA damage, were reported by an explanation for the aberrant differentiation of the gonads and
Meeker and co-workers, suggesting the lack of sufficient infor- the subsequent formation of the Leydig cells that produces the
mation in the literature to support conclusive evidence regarding hormone testosterone and results in the initiation of the masculin-
strong associations between the aforementioned 2 parameters ization of the male fetus (Sharpe, 2006; Sharpe et al., 2003). Despite
(Meeker et al., 2010). such promising observations, the exact contribution of pesticide
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 65

Table 2
A summary of the studies that have assessed the associations between the exposure of human populations to pesticides and EDC agents and the incidence of disorders
affecting the male reproductive system. (CS: cross sectional study; CC: case-control study; CHS: Cohort study; L: longitudinal study; PS: prospective study).

Authors, Year Chemical N Results Variables Study Type


Carlsen et al., 1992 N/A 14,947 men A genuine decline in semen Mean sperm density and L
quality is observed over the mean seminal volume
last 50 years in USA, Europe
and Australia
Swan et al., 1997 N/A 14,947 men Decline in sperm density in Mean sperm density L
USA and Europe
Swan et al., 2000 N/A 20,833 men Decline in sperm density Mean sperm density L
1.5%/year and 3%/year in USA
and Europe/Australia
respectively
Jørgensen et al., N/A 1082 fertile men Danish men had the lowest Sperm concentration and CS
2001 sperm concentrations, morphology
followed by French and
Scottish men. Finnish men had
the highest sperm counts
Jørgensen et al., N/A 968 men Finish and Estonian Sperm concentration CS
2002 populations had higher sperm
concentrations than
Norwegian and Dannish
Jørgensen et al., N/A 858 men (sperm Decline in sperm Sperm concentration, PS–CS
2011 parameters), 5974 men concentrations/counts in Finish sperm counts and
(testicular cancer registry) men over the years 1978–1987 incidence of testicular
with a simultaneous increase cancer
in testicular cancer 1954–2008
Jørgensen et al., N/A 4867 men Increase in sperm Sperm concentration, PS–CS
2012 concentration and/or counts in sperm counts, semen
Danish men over the years volume, sperm
1996–2010 morphology
Perry et al., 2011 OPs 94 cases and 95 controls Odds of being a case were Sperm concentration, CC
greater in men with higher sperm motility, urine DMP
urinary concentrations of and DAP concentrations
dimethylphosphate (DMP)
Yucra et al., 2006 OPs 31 sprayers and 80 controls Pesticide sprayers had Sperm concentration, CC
significantly reduced seminal sperm motility, semen
volume, percentage of motility, volume, sperm
percentage of sperm with morphology, LH, T levels
normal morphology, serum LH
and T levels, increased time of
liquefaction, seminal pH,
percentage of immature sperm
morphology,
Lifeng et al., 2006 Fenvalerate, 32 cases, 46 internal Sperm motion parameters; fenvalerate, toluene, CC
toluene, xylene controls, 22 external sperm progression and beat xylene concentrations in
controls cross frequency in the work place, dermal
exposure group were contamination of
decreased significantly fenvalerate, semen
compared with the internal qualities
and the external control
groups. Opposite effect for
abnormality rate of viscidity
and coagulation
Hossain et al., 2010 Paraquat, 152 farmers (62 exposed) The mean values of volume, Volume, pH, sperm CS
malathion pH, sperm concentration, concentration, motility,
motility, and WBC count were morphology and WBC
significantly less in the count were examined and
exposed group. Greater risk of recorded
abnormal semen parameters
was noted in the exposed
group
Petersen et al., PCBs 266 fertile men positive association between PCB exposure, semen CS
2015 serum–PCB and quality and reproductive
testosterone/estradiol ratio, hormones
elevated PCB exposure was
associated with increased
serum concentrations of SHBG,
no association between serum
PCB concentration and semen
quality variables
Pant et al., 2011 Phthalates 180 men, 65 correlation between phthalate Semen analysis, phthalate CC
asthenospermic, 65 ester diethylhexyl phthalate, ester concentration, in vitro
oligoasthenospermic, 50 di-n-butyl phthalate (DEHP cell viability
fertile and DBP) and sperm motility
both in vitro and in vivo
conditions
66 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

Table 2 (Continued)

Duty et al., 2003 Phthalates 168 men from subfertile Dose–response relations Urinary phthalate CC
couples between tertiles of mono-butyl metabolites, semen
phthalates and tertiles of analysis, sperm
mono-benzyl phthalate and concentration, sperm
sperm motility and sperm motility and morphology
concentration, respectively
Liu et al., 2012 Phthalates 150 men Borderline–significant Urinary levels of MMP, CHS
dose–response relationship MEP, MBP, MBzP, MEHP,
between MBP and sperm MEOHP, semen volume,
concentration, significant sperm concentration,
positive correlation between motility and sperm motion
MEP and straight–line velocity parameters
of sperm motion
Hauser et al., 2007 Phthalates 379 men from an infertility Monoethyl phthalate (MEP), Urinary concentrations of CS
clinic Mono-(2-ethylhexyl) phthalate phthalate metabolites,
(MEHP) were associated with sperm DNA damage
increased DNA damage measurements (comet
assay)
Hauser et al., 2006 Phthalates 463 male partners of Dose–response relationships of Urinary concentrations of CHS
subfertile couples MBP with low sperm phthalates, semen
concentration and motility parameters
Meeker et al., 2009 Phthalates 425 men from an infertility MEHP was inversely associated Urinary concentrations of CS
clinic with testosterone, estradiol, MEHP and other phthalate
and free androgen index, the monoester metabolites,
ratio of testosterone to serum levels of
estradiol was positively testosterone, estradiol,
associated with MEHP SHBG, FSH, LH, inhibin B,
prolactin
Mendiola et al., BPA 375 fertile men no significant associations FSH, LH, testosterone, CS
2010 between semen parameter and inhibin B, estradiol, SHBG,
urinary BPA concentration, androgen index
significant inverse association
between urinary BPA
concentration and FAI levels,
SHBG and the FAI/LH ratio
Meeker et al., 2010 BPA 190 men from an infertility Urinary BPA concentration was urinary BPA CS
clinic associated with declines in concentrations, semen
sperm concentration, motility, quality, and sperm DNA
and morphology of along with damage
a 10% increase in sperm DNA
damage
Li et al., 2011 BPA 218 men increasing urine BPA level was Semen parameters CHS
statistically significantly
associated with decreased
sperm concentration,
decreased total sperm count,
decreased sperm vitality
decreased sperm motility, no
association of urine BPA level
with semen volume or
abnormal sperm morphology
Boisen et al., 2004 1068 Danish boys, 1494 Prevalence of cryptorchidism Testicular position, L
Finnish boys, 5798 Finnish at birth was 9.0% in Denmark demographics,
newborns and 2.4% in Finland. The rate in confounding factors
Denmark was significantly
higher than that reported 40
years ago.

and EDC exposure to the development of testicular cancer has not Wolffian ducts in order to form the epididymis, seminal vesi-
been fully elucidated to date, although a combination of genetic cles, and vas deferens (Sweeney et al., 2015). The inhibition of
background and environmental factors have been demonstrated to the enzymes 5␣-reductase and aromatase by EDCs is one of the
account for the reduced sperm counts and the formation of the dis- main mechanisms responsible for the adverse effects noted, as
ease. In addition, the latter effects may not be recognized until 2–3 5␣-reductase is required for the conversion of the androgens
decades following fetal exposure to the corresponding chemical to testosterone, whereas aromatase catalyzes the metabolism of
compounds. This highlights the limitations of the clinical stud- androgens to estrogen (Fig. 3C, D) (Sweeney et al., 2015). The modu-
ies and the importance of the human population epidemiological lation of the expression levels of the two estrogen receptor isoforms
assessment studies (Publicover et al., 2007; Nordkap et al., 2012). namely, ER␣ and ER␤ is an additional mode of action of certain
EDCs. The expression of ER␤ is maintained during morphogene-
5.3. EDCs modes of action on the male reproductive system sis and cellular differentiation in humans and is decreased in early
puberty. The expression of the receptor is increased in response to
The effects of EDCs on the male reproductive system are notably elevated levels of androgens in plasma that may be associated with
attributed to the interactions of these chemicals with the normal benign prostate hyperplasia and/or prostate cancer. It has been
production of steroid hormones that are responsible for the ini- shown that early life exposure of neonatal rats to BPA enhances the
tiation of prostate development and the masculinization of the prostate susceptibility to estrogen-induced hyperplasia (Ho et al.,
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 67

2006). This effect is possibly due to the androgenic action of BPA, are not clear, although several factors have been proposed to affect
although epigenetic etiologies with regard to genes involved in the overall outcome of the study results as mentioned previously.
prostate cellular proliferation have been proposed (Sweeney et al., With regard to the male reproductive system, the contribution of
2015; Ho et al., 2006). geographical and seasonal variation of the semen parameters must
be considered. The majority of the epidemiological studies that
have examined chemical exposure and the associated semen qual-
5.4. EDCs, obesity, and male fertility
ity deterioration are cross-sectional. The measurement of exposure
and the possible health impairment are conducted simultaneously
The interaction of EDCs with male fertility is further associated
in the cross-sectional design that may result in skewed findings.
with the obesity status of the individuals exposed. It has been sug-
Consequently, a longitudinal design is required in studies of semen
gested that the biological effects of certain EDCs are prolonged in
quality. Despite contradictory observations the overall conclu-
obese individuals, due to the greater retention of the substances in
sion points toward a positive association between exposure to
the subcutaneous fat mass (Cardoso et al., 2017). Fat soluble tox-
EDCs and reproductive system disorders such as female infertility,
icants, such as PCBs and dioxin-type compounds can be stored in
miscarriage, endometriosis, cancer, semen quality, and testicular
the subcutaneous adipose tissue, resulting in an exacerbation of
dysgenesis, although the most critical parameter that is subject
the endocrine disrupting effects. The bioaccumulation of obeso-
to consideration is the frequency and dose of exposure. Future
gens is greater in obese individuals compared with lean subjects,
studies should focus on a uniform system of the investigation of
due to a larger fat mass that reduces the clearance rate of the tox-
human populations with regard to the exposure to specific EDCs
icants over time (Jung et al., 1997). Mechanistically, EDCs such as
and the direct effect on the reproductive system. In addition, the
BPA and phthalates may promote insulin resistance accounting for
use of advanced molecular biology techniques that are employed to
reduced gonadotrophin secretion at the hypothalamic–pituitary
evaluate DNA damage in spermatozoa namely, single cell gel elec-
level (Cardoso et al., 2017). Furthermore, permanent exposure to
trophoresis assay (COMET), terminal transferase dUTP Nick End
EDCs stored in adipose tissue (obesogens), may increase aromatase
Labelling (TUNEL), sperm chromatin structure assay (SCSA) and
activity that in turn increases E2 levels. The increased produc-
in situ nick translation (ISNT) as well as and sperm aneuploidy
tion of E2 can compromise downstream signaling events of the
must be included. Finally, scientists should adopt a methodologi-
Hypothalamic–pituitary axis and consequently alter the spermato-
cal strategy for the implementation of stricter rules and guidelines
genic process (Cardoso et al., 2017). Animal studies have shown that
on the safety of the use of chemicals and their effects on human
exposure to DDT induced transgenerational actions that promoted
reproduction.
obesity. The next generations of the gestating female rats that were
exposed to 50 mg/kg/day of DDT, at gestational days of 8–14, devel-
oped obesity and contained sperm epimutations (Skinner et al., Acknowledgments
2013). With regard to human populations there are few epidemi-
ological studies that have focused on the effects of obesogens in This work did not receive funding from a grant application.
sperm quality and consequently it is not possible to draw definite
conclusions on the exact interaction of the aforementioned com-
pounds with the molecular mechanisms involved in the production References
and function of human sperm (Cardoso et al., 2017).
Abdollahi, M., Ranjbar, A., Shadnia, S., Nikfar, S., Rezaie, A., 2004. Pesticides and
oxidative stress: a review. Med. Sci. Monit. 10, RA141–RA147.
Andersson, A.M., Jørgensen, N., Main, K.M., Toppari, J., Rajpert–De Meyts, E., Leffers,
6. Conclusion H., Juul, A., Jensen, T.K., Shakkebaek, N.E., 2008. Adverse trends in male
reproductive health: we may have reached a crucial “tipping point”. Int. J.
Endocrine disrupting chemicals comprise a class of compounds Androl. 31, 74–80.
Androutsopoulos, V.P., Tsatsakis, A.M., Spandidos, D.A., 2009. Cytochrome P450
that can potentially cause harmful effects to the male and female
CYP1A1: wider roles in cancer progression or prevention. BMC Cancer 9, 187.
reproductive systems. There has been significant progress in the Androutsopoulos, V.P., Hernandez, A.F., Liesivuori, J., Tsatsakis, A.M., 2013. A
identification of the biological action of EDCs by in vivo and in vitro mechanistic overview of health associated effects of low levels of
organochlorine and organophosphorous pesticides. Toxicology 307, 89–94.
experimental studies. In addition to the classical action of EDCs
Aoyama, H., Chapin, R.E., 2014. Reproductive toxicities of methoxychlor based on
that includes the agonism and/or antagonism with hormone and estrogenic properties of the compound and its estrogenic metabolite,
nuclear receptors, the significant scientific advances in the field hydroxyphenyltrichloroethane. Vitam. Horm. 94, 193–210.
of molecular biology have demonstrated that endocrine disrup- Aschengrau, A., Coogan, P.F., Quinn, M., Cashins, L.J., 1998. Occupational exposure
to estrogenic chemicals and the occurrence of breast cancer: an exploratory
tion by several compounds can interfere with the cell cycle, the analysis. Am. J. Ind. Med. 34, 6–14.
apoptotic machinery and the epigenetic regulation of the target Basavarajappa, M.S., Craig, Z.R., Hernandez-Ochoa, I., Paulose, T., Leslie, T.C., Flaws,
cells. In contrast to the experimental evidence, data derived from J.A., 2011. Methoxychlor reduces estradiol levels by altering steroidogenesis
and metabolism in mouse antral follicles in vitro. Toxicol. Appl. Pharmacol.
clinical and epidemiological studies have been somewhat contro- 253, 161–169.
versial as to what extent EDCs can affect the male and female Block, K., Kardana, A., Igarashi, P., Taylor, H.S., 2000. In utero diethylstilbestrol
reproductive systems. This discrepancy among clinical studies is (DES) exposure alters Hox gene expression in the developing mullerian
system. FASEB J. 14, 1101–1108.
attributed notably to the complexity of the clinical protocols used, Bloom, M.S., Kim, D., Vom Saal, F.S., Taylor, J.A., Cheng, G., Lamb, J.D., Fujimoto, V.Y.,
the degree of occupational or environmental exposure, the selec- 2011. Bisphenol A exposure reduces the estradiol response to gonadotropin
tion of the target group under investigation, the determination of stimulation during in vitro fertilization. Fertil. Steril. 96, 672–677.
Boisen, K.A., Chellakooty, M., Schmidt, I.M., Kai, C.M., Damgaard, I.N., Suomi, A.M.,
the variables measured and the sample size of the subjects exam-
Toppari, J., Skakkebaek, N.E., Main, K.M., 2005. Hypospadias in a cohort of 1072
ined. The findings can be broadly categorized into three different Danish newborn boys: prevalence and relationship to placental weight,
classes: 1) studies that have shown strong evidence of associa- anthropometrical measurements at birth, and reproductive hormone levels at
three months of age. J. Clin. Endocrinol. Metab. 90, 4041–4046.
tion between the adverse effects caused by EDCs and the extent
Boisen, K.A., Kaleva, M., Main, K.M., Virtanen, H.E., Haavisto, A.M., Schmidt, I.M.,
of exposure, 2) studies that have shown evidence of some associ- Chellakooty, M., Damgaard, I.N., Mau, C., Reunanen, M., Skakkebaek, N.E.,
ation between the adverse effects caused by EDCs and the extent Toppari, J., 2004. Difference in prevalence of congenital cryptorchidism in
of exposure and 3) studies that have shown no evidence of associ- infants between two Nordic countries. Lancet 363, 1264–1269.
Bredfeldt, T.G., Greathouse, K.L., Safe, S.H., Hung, M.C., Bedford, M.T., Walker, C.L.,
ation between the adverse effects caused by EDCs and the extent 2010. Xenoetsrogen-induced regulation of EZH2 and histone methylation via
of exposure. The explanations for such contradictory conclusions estrogen receptor signalling to PI3 K/AKT. Mol. Endocrinol. 24, 993–1006.
68 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

Buck Louis, G.M., Peterson, C.M., Chen, Z., Croughan, M., Sundaram, R., Stanford, J., alpha and beta, and androgen receptor: structure-activity studies. Mol.
Varner, M.W., Kennedy, A., Giudice, L., Fujimoto, V.Y., Sun, L., Wang, L., Guo, Y., Pharmacol. 58, 852–858.
Kannan, K., 2013. Bisphenol A and phthalates and endometriosis: the Geng, X., Shao, H., Zhang, Z., Ng, J.C., Peng, C., 2015. Malathion–induced testicular
endometriosis: Natural history, diagnosis and outcomes study. Fertil. Steril. toxicity is associated with spermatogenic apoptosis and alterations in
100, 162–169. testicular enzymes and hormone levels in male Wistar rats. Environ. Toxicol.
Butt, C.M., Stapleton, H.M., 2013. Inhibition of thyroid hormone sulfotransferase Pharmacol. 39, 659–667.
activity by brominated flame retardants and halogenated phenolics. Chem. Gore, A.C., Chappell, V.A., Fenton, S.E., Flaws, J.A., Nadal, A., Prins, G.S., Toppari, J.,
Res. Toxicol. 26, 1692–1702. Zoeller, R.T., 2015. Executive summary to EDC-2: The endocrine society’s
Byrne, S., Miller, P., Waghiyi, V., Buck, C.L., von Hippel, F.A., Carpenter, D.O., 2015. second scientific statement on endocrine-disrupting chemicals. Endocr. Rev.
Persistent organochlorine pesticide exposure related to a formerly used 36, 593–602.
defence site on St. Lawrence Island, Alaska: data from sentinel fish and human Gramec Skledar, D., Peterlin, Mašič, L., 2016. Bisphenol A and its analogs: do their
sera. J. Toxicol. Environ. Health A. 78, 976–992. metabolites have endocrine activity? Environ. Toxicol. Pharmacol. 47, 182–199.
Cardoso, A.M., Alves, M.G., Mathur, P.P., Oliveira, P.F., Cavaco, J.E., Rato, L., 2017. Grimm, F.A., Lehmler, H.-J., He, X., Robertson, L.W., Duffel, M.W., 2013. Sulfated
Obesogens and male fertility. Obes. Rev. 18, 109–125. metabolites of polychlorinated biphenyls are high-affinity ligands for the
Carlsen, E., Giwercman, A., Keiding, N., Skakkebaek, N.E., 1992. Evidence for thyroid hormone transport protein transthyretin. Environ. Health Perspect.
decreasing quality of semen during past 50 years. BMJ 305, 609–613. 121, 657–662.
Caserta, D., Bordi, G., Ciardo, F., Marci, R., La Rocca, C., Tait, S., Bergamasco, B., Gupta, R.K., Singh, J.M., Leslie, T.C., Meachum, S., Flaws, J.A., Yao, H.H., 2010.
Stecca, L., Mantovani, A., Guerranti, C., Fanello, E.L., Perra, G., Borghini, F., Di-(2-ethylhexyl) phthalate and mono-(2-ethylhexyl) phthalate inhibit growth
Focardi, S.E., Moscarini, M., 2013. The influence of endocrine disruptors in a and reduce estradiol levels of antral follicles in vitro. Toxicol. Appl. Pharmacol.
selected population of infertile women. Gynecol. Endocrinol. 29, 444–447. 242, 224–230.
Caserta, D., Di Segni, N., Mallozi, M., Giovanale, V., Mantovani, A., Marci, R., Hamers, T., Kamstra, J.H., Cenijn, P.H., Pencikova, K., Palkova, L., Simeckova, P.,
Moscarini, M., 2014. Bisphenol A and the female reproductive tract: an et al., 2011. In vitro toxicity profiling of ultrapure non-dioxin-like
overview of recent laboratory evidence and epidemiological studies. Reprod. polychlorinated biphenyl congeners and their relative toxic contribution to
Biol. Endocrinol. 12, 37. PCB mixtures in humans. Toxicol. Sci. 121, 88–100.
Chang, S.C., Tucker, T., Thorogood, N.P., Brown, C.J., 2006. Mechanisms of Hannon, P.R., Brannick, K.E., Wang, W., Gupta, R.K., Flaws, J.A., 2015.
X-chromosome inactivation. Front. Biosci. 11, 852–866. Di(2-ethylhexyl) phthalate inhibits antral follicle growth, induces atresia, and
Chen, M., Tang, R., Fu, G., Xu, B., Zhu, P., Qiao, S., Chen, X., Xu, B., Qin, Y., Lu, C., Hang, inhibits steroid hormone production in cultured mouse antral follicles. Toxicol.
B., Xia, Y., Wang, X., 2013. Association of exposure to phenols and idiopathic Appl. Pharmacol. 284, 42–53.
male fertility. J. Hazard. Material. 250-251, 115–121. Hao, J., Wang, J., Zhao, W., Ding, L., Gao, E., Yuan, W., 2011. Effect of bisphenol A
Chen, X., Ma, X.M., Ma, S.W., Goenraads, P.J., Zhang, C.M., Liu, J., Zhao, L.J., Sun, M., exposure on sex hormone level in occupational women. Wei Sheng Yan Jiu 40,
Tang, N.J., 2009. Proteomic analysis of the rat ovary following chronic low-dose 312–314.
exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). J. Toxicol. Environ. Hauser, R., Gaskins, A.J., Souter, I., Smith, K.W., Dodge, L.E., Ehrlich, S., Meeker, J.D.,
Health A 72, 717–726. Calafat, A.M., Williams, P.L., EARTH Study Team, 2016. Urinary phthalate
Chia, V.M., Quraishi, S.M., Devesa, S.S., Purdue, M.P., Cook, M.B., McGlynn, K.A., metabolite concentrations and reproductive outcomes among women
2010. Interventional trends in the incidence of testicular cancer, 1973–2002. undergoing in vitro fertilization: results from the EARTH study. Environ.
Cancer Epidemiol. Biomarkers Prev. 19, 1151–1159. Health Perspect. 124, 831–839.
Craig, Z.R., Hannon, P.R., Wang, W., Ziv-Gal, A., Flaws, J.A., 2013. Di-n-butyl Hauser, R., Meeker, J.D., Duty, S., Silva, M.J., Calafat, A.M., 2006. Altered semen
phthalate disrupts the expression of genes involved in cell cycle and apoptotic quality in relation to urinary concentrations of phthalate monoester and
pathways in mouse ovarian antral follicles. Biol. Reprod. 88, 23. oxidative metabolites. Epidemiology 17, 682–691.
Craig, Z.R., Wang, W., Flaws, J.A., 2011. Endocrine-disrupting chemicals in ovarian Hauser, R., Meeker, J.D., Singh, N.P., Silva, M.J., Ryan, L., Duty, S., Calafat, A.M., 2007.
function: effects on steroidogenesis, metabolism and nuclear receptor DNA damage in human sperm is related to urinary levels of phthalate
signaling. Reproduction 142, 633–646. monoester and oxidative metabolites. Hum. Reprod. 22, 688–695.
Desdoits–Lethimonier, C., Albert, O., Le Bizec, B., Perdu, E., Zalko, D., Courant, F., Heimler, I., Trewin, A.L., Chaffin, C.L., Rawlins, R.G., Hutz, R.J., 1998. Modulation of
Lesné, L., Guillé, F., Dejucq-Rainsford, N., Jégou, B., 2012. Human testis ovarian follicle maturation and effects on apoptotic cell death in Holtzman rats
steroidogenesis is inhibited by phthalates. Hum. Reprod. 27, 1451–1459. exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in utero and
De Silva, H.J., Samarawickrema, N.A., Wickremasinghe, A.R., 2006. Toxicity due to lactationally. Reprod. Toxicol. 12, 69–73.
organophosphorous compounds: what about chronic exposure? Trans. R. Soc. Hernández, A.F., Parrón, T., Tsatsakis, A.M., Requena, M., Alarcón, R.,
Trop. Med. Hyg. 100, 803–806. López–Guarnido, O., 2013. Toxic effects of pesticide mixtures at a molecular
Duty, S.M., Silva, M.J., Barr, D.B., Brock, J.W., Ryan, L., Chen, Z., Herrick, R.F., level: their relevance to human health. Toxicology 307, 136–145.
Christiani, D.C., Hauser, R., 2003. Phthalate exposure and human semen Hossain, F., Ali, O., D’Souza, U.J., Naing, D.K., 2010. Effects of pesticide use on semen
parameters. Epidemiology 14, 269–277. quality among farmers in rural areas of Sabah, Malaysia. J. Occup. Health 52,
Duty, S.M., Calafat, A.M., Silva, M.J., Brock, J.W., Ryan, L., Chen, Z., Overstreet, J., 353–360.
Hauser, R., 2004. The relationship between environmental exposure to Ho, S.M., Tang, W.Y., Belmonte de Frausto, J., Prins, G.S., 2006. Developmental
phthalates and computer-aided sperm analysis motion parameters. J. Androl. exposure to estradiol and bisphenol A increases susceptibility to prostate
25, 293–302. carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant.
Ehrlich, S., Williams, P.L., Missmer, S.A., Flaws, J.A., Berry, K.F., Calafat, A.M., Ye, X., Cancer Res. 66, 5624–5632.
Petrozza, J.C., Wright, D., Hauser, R., 2012a. Urinary bisphenol A concentrations Jensen, M.S., Toft, G., Thulstrup, A.M., Bonde, J.P., Olsen, J., 2007. Cryptorchidism
and implantation failure among women undergoing in vitro fertilization. according to maternal gestational smoking. Epidemiology 18, 220–225.
Environ. Health Perspect. 120, 978–983. Jirtle, R.L., Skinner, M.K., 2007. Environmental epigenomics and disease
Ehrlich, S., Williams, P.L., Missmer, S.A., Flaws, J.A., Ye, X., Calafat, A.M., Petrozza, susceptibility. Nat. Rev. Genet. 8, 253–262.
J.C., Wright, D., Hauser, R., 2012b. Urinary bisphenol A concentrations and early Jørgensen, N., Andersen, A.G., Eustache, F., Irvine, D.S., Suominen, J., Petersen, J.H.,
reproductive health outcomes among women undergoing IVF. Hum. Reprod. Andersen, A.N., Auger, J., Cawood, E.H., Horte, A., Jensen, T.K., Jouannet, P.,
27, 3583–3592. Keiding, P., Vierula, M., Toppari, J., Skakkebaek, N.E., 2001. Regional differences
Elsby, R., Maggs, J.L., Ashby, J., Park, B.K., 2001. Comparison of the modulatory in semen quality in Europe. Hum. Reprod. 16, 1012–1019.
effects of human and rat liver microsomal metabolism on the estrogenicity of Jørgensen, N., Carlsen, E., Nermoen, I., Punab, M., Suominen, J., Andersen, A.G.,
bisphenol A: implications for extrapolation to humans. J. Pharmacol. Exp. Ther. Andersson, A.M., Haugen, T.B., Horte, A., Jensen, T.K., Magnus, Ø., Petersen, J.H.,
297, 103–113. Vierula, M., Toppari, J., Skakkebaek, N.E., 2002. East–west gradient in semen
Falck, L., Forsberg, J.G., 1996. Immunohistochemical studies on the expression and quality in the Nordic–Baltic area: a study of men from the general population
estrogen dependency of EGF and its receptor and C-fos proto-oncogene in the in Denmark, Norway, Estonia and Finland. Hum. Reprod. 17, 2199–
uterus and vagina of normal and neonatally estrogen-treated mice. Anat. Rec. 2208.
245, 459–471. Jørgensen, N., Joensen, U.N., Jensen, T.K., Jensen, M.B., Almstrup, K., Olesen, I.A.,
Figà–Talamanca, I., Traina, M.E., Urbani, E., 2001. Occupational exposures to Juul, A., Andersson, A.M., Carlsen, E., Petersen, J.H., Toppari, J., Skakkebaek, N.E.,
metals, solvents and pesticides: recent evidence on male reproductive effects 2012. Human semen quality in the new millennium: a prospective
and biological markers. Occup. Med. (Lond.) 51, 174–188. cross–sectional population based study of 4867 men. BMJ Open 2 (pii),
Fisher, J.S., 2004. Environmental anti-androgens and male reproductive health: e000990.
focus on phthalates and testicular dysgenesis syndrome. Reproduction 127, Jørgensen, N., Vierula, M., Jacobsen, R., Pukkala, E., Perheentupa, A., Virtanen, H.E.,
305–315. Skakkebaek, N.E., Toppari, J., 2011. Recent adverse trends in semen quality and
Foster, P.M., 2005. Mode of action: impaired fetal leydig cell function—effects on testis cancer incidence among Finnish men. Int. J. Androl. 34, e37–e48.
male reproductive development produced by certain phthalate esters. Crit. Jouannet, P., Wang, C., Eustache, F., Kold–Jensen, T., Auger, J., 2001. Semen quality
Rev. Toxicol. 35, 713–719. and male reproductive health: the controversy about human sperm
Fujimoto, V.Y., Kim, D., vom Saal, F.S., Lamb, J.D., Taylor, J.A., Bloom, M.S., 2011. concentration decline. APMIS 109, 333–344.
Serum unconjugated bisphenol A concentrations in women may adversely Jung, D., Becher, H., Edler, L., Flesch–Janys, D., Gurn, P., Konietzko, J., et al., 1997.
influence oocyte quality during in vitro fertilization. Fertil. Steril. 95, Elimination of beta–hexachlorocyclohexane in occupationally exposed
1816–1819. persons. J. Toxicol. Environ. Health 51, 23–34.
Gaido, K.W., Maness, S.C., McDonnell, D.P., Dehal, S.S., Kupfer, D., Safe, S., 2000. Jurewicz, J., Hanke, W., Sobala, W., Ligocka, D., 2009. Assesment of the dermal
Interaction of methoxychlor and related compounds with estrogen receptor exposure to azoxystrobin among women tending cucumbers in selected polish
S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70 69

greenhouses after restricted entry intervals expired—the role of the protective Mehrpour, O., Karrari, P., Zamani, N., Tsatsakis, A.M., Abdollahi, M., 2014.
gloves. Int. J. Occup. Med. Environ. Health 22, 261–267. Occupational exposure to pesticides and consequences on male semen and
Kandaraki, E., Chatzigeorgiou, A., Livadas, S., Palioura, E., Economou, F., Koutsilieris, fertility: a review. Toxicol. Lett. 230, 146–156.
M., Palimeri, S., Panidis, D., Diamanti-Kandarakis, E., 2011. Endocrine Mendiola, J., Jørgensen, N., Andersson, A.M., Calafat, A.M., Ye, X., Redmon, J.B.,
disruptors and polycystic ovary syndrome (PCOS): elevated serum levels of Drobnis, E.Z., Wang, C., Sparks, A., Thurston, S.W., Liu, F., Swan, S.H., 2010. Are
bisphenol A in women with PCOS. J. Clin. Endocrinol. Metab. 96, E480–E484. environmental levels of bisphenol A associated with reproductive function in
Kapka-Skrzypczak, L., Cyranka, M., Skrzypczak, M., Kruszewski, M., 2011. fertile men? Environ. Health Perspect. 118, 1286–1291.
Biomonitoring and biomarkers of organophosphate pesticides exposure-state Miyawaki, J., Sakayama, K., Kato, H., Yamamoto, H., Masuno, H., 2007. Perinatal and
of the art. Ann. Agricult. Environ. Med. 18, 294–303. postnatal exposure to bisphenol A increases adipose tissue mass and serum
Karman, B.N., Basavarajappa, M.S., Craig, Z.R., Flaws, J.A., 2012a. cholesterol level in mice. J. Atheroscler. Thromb. 14, 245–252.
2,3,7,8-Tetrachlorodibenzo-p-dioxin activates the aryl hydrocarbon receptor Mok-Lin, E., Ehrlich, S., Williams, P.L., Petrozza, J., Wright, D.L., Calafat, A.M., Ye, X.,
and alters sex steroid hormone secretion without affecting growth of mouse Hauser, R., 2010. Urinary bisphenol A concentrations and ovarian response
antral follicles in vitro. Toxicol. Appl. Pharmacol. 261, 88–96. among women undergoing IVF. Int. J. Androl. 33, 385–393.
Karman, B.N., Basavarajappa, M.S., Hannon, P., Flaws, J.A., 2012b. Dioxin exposure Moriyama, K., Tagami, T., Akamizu, T., Usui, T., Saijo, M., Kanamoto, N., et al., 2002.
reduces the steroidogenic capacity of mouse antral follicles mainly at the level Thyroid hormone action is disrupted by bisphenol A as an antagonist. J. Clin.
of HSD17B1 without altering atresia. Toxicol. Appl. Pharmacol. 264, 1–12. Endocrinol. Metab. 87, 5185–5190.
Kelce, W.R., Stone, C.R., Laws, S.C., Gray, L.E., Kemppainen, J.A., Wilson, E.M., 1995. Mrema, E.J., Rubino, F.M., Brambilla, G., Moretto, A., Tsatsakis, A.M., Colosio, C.,
Persistent DDT metabolite p,p –DDE is a potent androgen receptor antagonist. 2013. Persistent organochlorinated pesticides and mechanisms of their
Nature 375, 581–585. toxicity. Toxicology 307, 74–88.
Knez, 2013. Endocrine-disrupting chemicals and male reproductive health. Narayana, K., Prashanthi, N., Nayanatara, A., Kumar, H.H., Abhilash, K., Bairy, K.L.,
Reprod. Biomed. Online 26, 440–448. 2006. Neonatal methyl parathion exposure affects the growth and functions of
Koureas, M., Tsakalof, A., Tsatsakis, A., Hadjichristodoulou, C., 2012. Systematic the male reproductive system in the adult rat. Folia Morphol. (Warsz) 65,
review of biomonitoring studies to determine the association between 26–33.
exposure to organophosphorus and pyrethroid insecticides and human health Nelson, K.G., Sakai, Y., Eitzman, B., Steed, T., McLachlan, J., 1994. Exposure to
outcomes. Toxicol. Lett. 210, 155–168. diethylstilbesterol during a critical developmental period of the mouse
Koureas, M., Tsakalof, A., Tzatzarakis, M., Vakonaki, E., Tsatsakis, A., reproductive tract leads to persistent induction of two estrogen-regulated
Hadjichristodoulou, C., 2014. Biomonitoring of organophosphate exposure of genes. Cell Growth Differ. 5, 595–606.
pesticide sprayers and comparison of exposure levels with other population Nishimura, N., Miyabara, Y., Sato, M., Yonemoto, J., Tohyama, C., 2002.
groups in Thessaly (Greece). Occup. Environ. Med. 71, 126–133. Immunohistochemical localization of thyroid stimulating hormone induced by
Koutroulakis, D., Sifakis, S., Tzatzarakis, M.N., Alegakis, A.K., Theodoropoulou, E., a low oral dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin in female
Kavvalakis, M.P., Kappou, D., Tsatsakis, A.M., 2014. Dialkyl phosphates in Sprague-Dawley rats. Toxicology 171, 73–82.
amniotic fluid as a biomarker of fetal exposure to organophosphates in Crete, Nordkap, L., Joensen, U.N., Blomberg Jensen, M., Jørgensen, N., 2012. Regional
Greece; association with fetal growth. Reprod. Toxicol. 46, 98–105. differences and temporal trends in male reproductive health disorders: semen
Lee, S.G., Kim, J.Y., Chung, J.Y., Kim, Y.J., Park, J.E., Oh, S., Yoon, Y.D., Yoo, K.S., Yoo, quality may be a sensitive marker of environmental exposures. Mol. Cell
Y.H., Kim, J.M., 2013. Bisphenol A exposure during adulthood causes Endocrinol. 355, 221–230.
augmentation of follicular atresia and luteal regression by decreasing Noyes, P.D., Lema, S.C., Macaulay, L.J., Douglas, N.K., Stapleton, H.M., 2013. Low
17␤-estradiol synthesis via downregulation of aromatase in rat ovary. Environ. level exposure to the flame retardant BDE-209 reduces thyroid hormone levels
Health Perspect. 121, 663–669. and disrupts thyroid signaling in fathead minnows. Environ. Sci. Technol. 47,
Lemaire, G., Mnif, W., Mauvais, P., Balaguer, P., Rahmani, R., 2006. Activation of 10012–10021.
alpha- and beta-estrogen receptors by persistent pesticides in reporter cell Ogbeide, O., Tongo, I., Ezemonye, L., 2015. Risk assesment of agricultural pesticides
lines. Life Sci. 79, 1160–1169. in water, sediment, and fish from Owan river, Edo State, Nigeria. Environ.
Li, D.K., Zhou, Z., Miao, M., He, Y., Wang, J., Ferber, J., Herrinton, L.J., Gao, E., Yuan, Monit. Assess. 187, 654.
W., 2011. Urine bisphenol–A (BPA) level in relation to semen quality. Fertil. Omoike, O.E., Lewis, R.C., Meeker, J.D., 2015. Associations between urinary
Steril. 95, 625–630. biomarkers of exposure to organophosphate insecticides and serum
Li, S., Hansman, R., Newbold, R., Davis, B., McLachlan, J.A., Barrett, J.C., 2003. reproductive hormones in men from NHANES 1999–2002. Reprod. Toxicol. 53,
Neonatal diethylstilbestrol exposure induces persistent elevation of c-fos 99–104.
expression and hypomehtylation in its exon-4 in mouse uterus. Mol. Carcinog. Pant, N., Pant, A., Shukla, M., Mathur, N., Gupta, Y., Saxena, D., 2011. Environmental
38, 78–84. and experimental exposure of phthalate esters: the toxicological consequence
Li, S., Washburn, K.A., Moore, R., Uno, T., Teng, C., Newbold, R.R., McLachlan, J.A., on human sperm. Hum. Exp. Toxicol. 30, 507–514.
Negishi, M., 1997. Developmental exposure to diethylstilbestrol elicits Park, S.A., Lee, M.H., Na, H.K., Surh, Y.J., 2017. 4-hydroxyestradiol induces
demethylation of estrogen-responsive lactoferrin gene in mouse uterus. mammary epithelial cell transformation through Nrf2-mediated heme
Cancer Res. 57, 4356–4359. oxygenase-1 overexpression. Oncotarget 8, 164–178.
Lifeng, T., Shoulin, W., Junmin, J., Xuezhao, S., Yannan, L., Qianli, W., Longsheng, C., Peiris–John, R.J., Wickremasinghe, R., 2008. Impact of low-level exposure to
2006. Effects of fenvalerate exposure on semen quality among occupational organophosphates on human reproduction and survival. Trans. R. Soc. Trop.
workers. Contraception 73, 92–96. Med. Hyg. 102, 239–245.
Liu, L., Bao, H., Liu, F., Zhang, J., Shen, H., 2012. Phthalates exposure of Chinese Perry, M.J., Venners, S.A., Barr, D.B., Xu, X., 2007. Environmental pyrethroid and
reproductive age couples and its effect on male semen quality, a primary organophosphorous insecticide exposures and sperm concentration. Reprod.
study. Environ. Int. 42, 78–83. Toxicol. 23, 113–118.
Llanes Gonzalez, L., Lujan Galan, M., Rodriguez Garcia, N., Garcia Tello, A., Perry, M.J., Venners, S.A., Chen, X., Liu, X., Tang, G., Xing, H., Barr, D.B., Xu, X., 2011.
Berenguer Sanchez, A., 2008. Trends in the incidence of testicular germ cell Organophosphorous pesticide exposures and sperm quality. Reprod. Toxicol.
cancer in 300.000 inhabitants Spanish population (1991–2005). Actas Urol. 31, 75–79.
Esp. 32, 691–695. Perry, M.J., 2008. Effects of environmental and occupational pesticide exposure on
Ma, J., Pan, L.B., Yang, X.Y., Liu, X.L., Tao, S.Y., Zhao, L., Qin, X.P., Sun, Z.J., Hou, H., human sperm: a systematic review. Hum. Reprod. Update 14, 233–242.
Zhou, Y.Z., 2016. DDT, DDD, and DDE in soil of Xiangfen County, China: Petersen, M.S., Halling, J., Weihe, P., Jensen, T.K., Grandjean, P., Nielsen, F.,
residues, sources, spatial distribution, and health risks. Chemosphere 163, Jørgensen, N., 2015. Spermatogenic capacity in fertile men with elevated
578–583. exposure to polychlorinated biphenyls. Environ. Res. 138, 345–351.
Mahalingajah, S., Missmer, S.A., Maity, A., Williams, P.L., Meeker, J.D., Berry, K., Philippat, C., Mortamais, M., Chevrier, C., Petit, C., Calafat, A.M., Ye, X., Silva, M.J.,
Ehrlich, S., Perry, M.J., Cramer, D.W., Hauser, R., 2012. Association of Brambilla, C., Pin, I., Charles, M.A., Cordier, S., Slama, R., 2012. Exposure to
hexachlorobenzene (HCB), dichlorodiphenyltrichloroethane (DDT), and phthalates and phenols during pregnancy and offspring size at birth. Environ.
dichlorodiphenyltrichloroethylene (DDE) with in vitro fertilization outcomes. Health Perspect. 120, 464–470.
Environ. Health Perspect. 120, 316–320. Poet, T.S., McDougal, J.N., 2002. Skin absorption and human risk assessment. Chem.
Martinez–Arguelles, D.B., Culty, M., Zirkin, B.R., Papadopoulos, V., 2009. In utero Biol. Interact. 140, 217–227.
exposure to di–(2–ethylhexyl) phthalate decreases mineralocorticoid receptor Publicover, S., Harper, C.V., Barratt, C., 2007. [Ca2+]i signalling in sperm–making
expression in the adult testis. Endocrinology 150, 5575–5585. the most of what you ‘ve got. Nat. Cell Biol. 9, 235–242.
Meeker, J.D., Calafat, A.M., Hauser, R., 2009. Urinary metabolites of Raanan, R., Harley, K.G., Balmes, J.R., Bradman, A., Lipsett, M., Eskenazi, B., 2015.
di(2–ethylhexyl) phthalate are associated with decreased steroid hormone Early-life exposure to organophosphate pesticides and pediatric respiratory
levels in adult men. J. Androl. 30, 287–297. symptoms in the CHAMACOS cohort. Environ. Health Perspect. 123, 179–185.
Meeker, J.D., Ehrlich, S., Toth, T.L., Wright, D.L., Calafat, A.M., Trisini, A.T., Ye, X., Rajakumar, A., Singh, R., Chakrabarty, S., Murugananthkumar, R., Laldinsangi, C.,
Hauser, R., 2010. Semen quality and sperm DNA damage in relation to urinary Prathibha, Y., Sudhakumari, C.C., Dutta-Gupta, A., Senthilkumaran, B., 2012.
bisphenol A among men from an infertility clinic. Reprod. Toxicol. 30, Endosulfan and flutamide impair testicular development in the juvenile Asian
532–539. catfish Clarias batrachus. Aquat. Toxicol. 110-111, 123–132.
Meerts, I.A., Assink, Y., Cenijn, P.H., Van Den Berg, J.H., Weijers, B.M., Bergman, A., Relakis, K., Sifakis, S., Froudarakis, G., Papadopoulou, E., Michalodimitrakis, E.N.,
et al., 2002. Placental transfer of a hydroxylated polychlorinated biphenyl and Koumantakis, E., Tsatsakis, A.M., 1999. Disposition of pesticides and toxicants
effects on fetal and maternal thyroid hormone homeostasis in the rat. Toxicol. in the human reproductive system in cases of acute poisoning. Clin. Exp.
Sci. 68, 361–371. Obstet. Gynecol. 26, 207–210.
70 S. Sifakis et al. / Environmental Toxicology and Pharmacology 51 (2017) 56–70

Rochester, J.R., 2013. Bisphenol A and human health: a review of the literature. cord blood and meconium analysis to detect fetal exposure to environmental
Reprod. Toxicol. 42, 132–155. pesticides” by Ostrea et al. Environ. Res. 106, 277–283, Environ. Res. 109
Safe, S., 2013. Endocrine disruptors and falling sperm counts: lessons learned or (2009) 352–353.
not! Asian J. Androl. 15, 191–194. Tsatsakis, A.M., 2006. ‘Maternal hair–an appropriate matrix for detecting maternal
Senthilkumaran, B., 2015. Pesticide- and sex steroid analogue-induced endocrine exposure to pesticides during pregnancy’. Environ. Res. 102, 365.
disruption differentially targets hypothalamo-hypophyseal-gonadal system Turner, B.M., 2009. Epigenetic responses to environmental change and their
during gametogenesis in teleosts- a review. Gen. Comp. Endocrinol. 219, evolutionary implications. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 364,
136–142. 3403–3418.
Sharpe, R.M., McKinnell, C., Kivlin, C., Fisher, J.S., 2003. Proliferation and functional Tzatzarakis, M.N., Karzi, V., Vakonaki, E., Goumenou, M., Kavvalakis, M., Stivaktakis,
maturation of Sertoli cells, and their relevance to disorders of testis function in P., et al., 2016. Bisphenol A in soft drinks and canned foods and data evaluation.
adulthood. Reproduction 125, 769–784. Food Addit. Contam. Part B Surveill. 11, 1–6.
Sharpe, R.M., 2006. Pathways of endocrine disruption during male sexual Tzatzarakis, M.N., Vakonaki, E., Kavvalakis, M.P., Barmpas, M., Kokkinakis, E.N.,
differentiation and masculinization. Best Pract. Res. Clin. Endocrinol. Metab. Xenos, K., Tsatsakis, A.M., 2015. Biomonitoring of bisphenol A in hair of Greek
20, 91–110. population. Chemosphere 118, 336–341.
Sifakis, S., Tsatsakis, A., Mparmpas, M., Soldin, O.P., 2011. Pesticide exposure and Usmani, K.A., Rose, R.L., Hodgson, E., 2003. Inhibition and activation of the human
health related issues in male and female reproductive system. INTECH. liver and human cytochrome P450 3A4 metabolism of testosterone by
Skakkebaek, N.E., Rajpert–De Meyts, E., Jørgensen, N., Main, K.M., Leffers, H., deployment–related chemicals. Drug Metab. Dispos. 31, 384–391.
Andersson, A.M., Juul, A., Jensen, T.K., Toppari, J., 2007. Testicular cancer trends Usmani, K.A., Cho, T.M., Rose, R.L., Hodgson, E., 2006. Inhibition of the human liver
as ‘whistle blowers’ of testicular developmental problems in populations. Int. J. microsomal and human cytochrome P450 1A2 and 3A4 metabolism of
Androl. 30, 198–204. estradiol by deployement–related and other chemicals. Drug Metab. Dispos.
Slutsky, M., Levin, J.L., Levy, B.S., 1999. Azoospermia and oligospermia among a 34, 1606–1614.
large cohort of DBCP applicators in 12 countries. Int. J. Occup. Environ. Health Uzumcu, M., Kuhn, P.E., Marano, J.E., Armenti, A.E., Passantino, L., 2006. Early
5, 116–122. postnatal methoxychlor exposure inhibits folliculogenesis and stimulates
Sokoloff, K., Fraser, W., Arbuckle, T.E., Fisher, M., Gaudreau, E., LeBlanc, A., Morisset, anti-Mullerian hormone production in the rat ovary. J. Endocrinol. 191,
A.S., Bouchard, M.F., 2016. Determinants of urinary concentrations of dialkyl 549–558.
phosphates among pregnant women in Canada–resutls from the MIREC study. Viluksela, M., Raasmaja, A., Lebofsky, M., Stahl, B.U., Rozman, K.K., 2004.
Environ. Int. 94, 133–140. Tissue-specific effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the
Souter, I., Smith, K.W., Dimitriadis, I., Ehrlich, S., Williams, P.L., Calafat, A.M., activity of 5 -deiodinases I and II in rats. Toxicol. Lett. 147, 133–142.
Hauser, R., 2013. The association of bisphenol-A urinary concentrations with Wang, W., Craig, Z.R., Basavarajappa, M.S., Hafner, K.S., Flaws, J.A., 2012.
antral follicle counts and other measures of ovarian reserve in women Mono-(2-ethylhexyl) phthalate induces oxidative stress and inhibits growth of
undergoing infertility treatments. Reprod. Toxicol. 42, 224–231. mouse ovarian antral follicles. Biol. Reprod. 87, 152.
Stoorgaard, L., Bonde, J.P., Ernst, E., Spano, M., Andersen, C.Y., Frydenberg, M., Wang, W., Hafner, K.S., Flaws, J.A., 2014. In utero bisphenol A exposure disrupts
Olsen, J., 2003. Does smoking during pregnancy affect sons’ sperm counts? germ cell nest breakdown and reduces fertility with age in the mouse. Toxicol.
Epidemiology 14, 278–286. Appl. Pharmacol. 276, 157–164.
Skinner, M.K., Manikham, M., Guerrero–Bosagna, C., 2011. Epigenetic Waters, K.M., Safe, S., Gaido, K.W., 2001. Differential gene expression in response
transgenerational actions of endocrine disruptors. Reprod. Toxicol. 31, to methoxychlor and estradiol through ERalpha, ERbeta and AR in
337–343. reproductive tissues of female mice. Toxicol. Sci. 63, 47–56.
Skinner, M.K., Manikkam, M., Tracey, R., Guerrero–Bosagna, C., Haque, M., Nilsson, Welshons, W.V., Thayer, K.A., Judy, B.M., Taylor, J.A., Curran, E.M., vom Saal, F.S.,
E.E., 2013. Ancestral dichlorodiphenyltrichloroethane (DDT) exposure 2003. Large effects from small exposures I. Mechanisms for
promotes epigenetic transgenerational inheritance of obesity. BMC Med. 11, endocrine-disrupting chemicals with estrogenic activity. Environ. Health
228. Perspect. 111, 994–1006.
Sugiura–Ogasawara, M., Ozaki, Y., Sonta, S., Makino, T., Suzumori, K., 2005. Wetherill, Y.B., Fisher, N.L., Staubach, A., Danielsen, M., de Vere White, R.W.,
Exposure to bisphenol A is associated with recurrent miscarriage. Hum. Knudsken, K.E., 2005. Xenoestrogen action in prostate cancer: pleiotropic
Reprod. 20, 2325–2329. effects dependent on androgen receptor status. Cancer Res. 65, 54–65.
Swan, S.H., Elkin, E.P., Fenster, L., 1997. Have sperm densities declined? a Xu, C., Chen, J.A., Qiu, Z., Zhao, Q., Luo, J., Yang, L., Zeng, H., Huang, Y., Zhang, L., Cao,
reanalysis of global trend data. Environ. Health Perspect. 105, 1228–1232. J., Shu, W., 2010. Ovotoxicity and PPAR-mediated aromatase downregulation
Swan, S.H., Elkin, E.P., Fenster, L., 2000. The question of declining sperm density in female Sprague-Dawley rats following combined oral exposure to
revisited: an analysis of 101 studies published 1934–1996. Environ. Health benzo[a]pyrene and di-(2-ethylhexyl) phthalate. Toxicol. Lett. 199, 323–332.
Perspect. 108, 961–966. Yang, M., Ryu, J.H., Jeon, R., Kang, D., Yoo, K.Y., 2009. Effects of bisphenol A on
Sweeney, M.F., Hasan, N., Soto, A.M., Sonnenschein, C., 2015. Environmental breast cancer and its risk factors. Arch. Toxicol. 83, 281–285.
endocrine disruptors: effects on the human male reproductive system. Rev. Yucra, S., Rubio, J., Gasco, M., Gonzales, C., Steenland, K., Gonzales, G.F., 2006.
Endocr. Metab. Disord. 16, 341–357. Semen quality and reproductive sex hormone levels in Peruvian pesticide
Tang, W.Y., Newbold, R., Mardilovich, K., Jefferson, W., Cheng, R.Y., Medvedovic, M., sprayers. Int J. Occup. Environ. Health 12, 355–361.
Ho, S.M., 2008. Persistent hypomethylation in the promoter of nucleosomal Zachow, R., Uzumcu, M., 2006. The methoxychlor metabolite,
binding protein 1 (Nsbp1) correlates with overexpression of Nsbp1 in mouse 2,2-bis-(p-hydroxyphenyl)-1,1,1-trichloroethane, inhibits steroidogenesis in
uteri neonatally exposed to diethylstilbestrol or genistein. Endocrinology 149, rat ovarian granulosa cells in vitro. Reprod. Toxicol. 22, 659–665.
5922–5931. Zama, A.M., Uzumcu, M., 2010. Epigenetic effects of endocrine-disrupting
Taylor, J.A., Richter, C.A., Ruhlen, R.L., vom Saal, F.S., 2011. Estrogenic chemicals on female reproduction: an ovarian perspective. Front.
environmental chemicals and drugs: mechanisms for effects on the developing Neuroendocrinol. 31, 420–439.
male urogenital system. J. Steroid Biochem. Mol. Biol. 127, 83–95. Zama, A.M., Uzumcu, M., 2009. Fetal and neonatal exposure to the endocrine
Thorup, J., Cortes, D., Petersen, B.L., 2006. The incidence of bilateral cryptorchidism disruptor methoxychlor causes epigenetic alterations in adult ovarian genes.
is increased and the fertility potential is reduced in sons born to mothers who Endocrinology 150, 4681–4691.
have smoked during pregnancy. J. Urol. 176, 734–737. Zhang, Y., Han, S., Liang, D., Shi, X., Wang, F., Liu, W., Zhang, L., Chen, L., Gu, Y., Tian,
Tsatsakis, A.M., Tzatzarakis, M.N., Koutroulakis, D., Toutoudaki, M., Sifakis, S., 2009. Y., 2014. Prenatal exposure to organophosphate pesticides and
Dialkyl phosphates in meconium as a biomarker of prenatal exposure to neurobehavioral development of neonates: a birth cohort study in Shenyang.
organophosphate pesticides: a study on pregnant women of rural areas in China. PLoS ONE 9, e88491.
Crete, Greece. Xenobiotica 39, 364–373.
Tsatsakis, A.M., 2008. Commentary on the levels of pesticide exposure in general
and rural population based on the article titled “A comparison of infant hair,

You might also like