Open access peer-reviewed chapter

G-Protein Coupled Hormone Receptors of the Hypothalamic-Pituitary-Gonadal Axis are Targets of Endocrine Disrupting Chemicals

Written By

Valentine Suteau, Patrice Rodien and Mathilde Munier

Submitted: 23 November 2020 Reviewed: 27 January 2021 Published: 28 April 2021

DOI: 10.5772/intechopen.96240

From the Edited Volume

Hot Topics in Endocrinology and Metabolism

Edited by Hassan Massoud Heshmati

Chapter metrics overview

440 Chapter Downloads

View Full Metrics

Abstract

Endocrine-disrupting chemicals have received significant concern, since they ubiquitously persist in the environment and are able to induce adverse effects on health, and more particularly on reproductive function. Most of the studies focused on nuclear hormone receptors as mediators of sex steroid hormones signaling. However, there are increasing evidences that peptides hormones of the Hypothalamo-Pituitary-Gonadal axis are targets of endocrine-disrupting chemicals (as Gonadotropin-Releasing Hormone, Follicle-Stimulating Hormone, Luteinizing Hormone…). The majority of these hormones act on G protein-coupled membrane receptors. This review summarizes the effects of endocrine-disrupting chemicals on homeostasis of peptides hormone of Hypothalamo-Pituitary-Gonadal axis and on their G protein-coupled membrane receptors signaling revealed by experimental, clinical, and epidemiological studies in human.

Keywords

  • G-protein coupled hormone receptors
  • hypothalamic-pituitary-gonadal axis
  • hormones
  • endocrine-disrupting chemicals

1. Introduction

Public concern of endocrine-disrupting chemicals (EDCs) has been rising since the 1990s. EDCs are defined as “an exogenous substance or mixture that alters function(s) of the endocrine system and consequently causes adverse health effects in an intact organism, or its progeny, or (sub)populations” [1, 2, 3]. EDCs are found in many products comprising plasticizers, personal care products, pesticides… [1]. Humans are constantly exposed to several different EDCs by ingestion, inhalation, and dermal contact. Some classes of EDCs have been studied in detail. Here, we selected three classes of EDCs based on knowledge of their effects on Hypothalamo-Pituitary-Gonadal (HPG) axis: bisphenol A (BPA), phthalates and dichlorodiphenyltrichloroethane (DDT). BPA is one of the most massively produced EDC with over three million tons manufactured annually [4]. It is used in food packaging, toys, resins used in canned, and medical equipment. Because its incomplete polymerization and its release from polycarbonate at high temperature, exposure to BPA is important via food containers [5, 6, 7]. Phthalates are used as liquid plasticizers found in a wide range of products including plastics, coatings, toys, cosmetics, and medical tubing. They are classified in two groups: high molecular weight phthalates, such as diethylhexyl phthalte (DEHP), and low molecular weight phthalates, such as dibutyl phthalates (DBP) [8]. DDT, an organochlorine pesticide, was largely used after the Second World War for its insecticidal properties. Although it was banned in the 1970s in the Western World, it continues to be used in developing countries. DDT is a synthetic mixture of three isoforms: p,p’DDT, o,p’DDT and p,p’DDD. EDCs are originally thought to act through nuclear hormone receptors, such as estrogen receptor (ER) or androgen receptor (AR) [9]. During the last decade, we, and others, were interested in the effect of EDCs on G-protein-coupled hormone receptors (GPCRs). These studies have shown that there are chemical compounds in the environment capable of binding to GPCRs and disrupting the activity and intracellular signaling pathways of receptor. Moreover, EDCs may alter pathways involved in hormone biosynthesis and/or receptor signaling regulation. This review summarizes the effects of three classes of EDCs on hormones homeostasis and GPCRs signaling involved in the HPG axis. Several molecular mechanisms can be involved in the EDC effects on the HPG axis. All studies cited here were performed in human species.

Advertisement

2. GPCRs implicated in the HPG axis

The GPCRs are the largest family of cell-surface receptors with over 800 members accounting for 4% of the encoded human genome [10]. About half of them have sensory functions, mediating olfaction, taste, light perception, and pheromone signaling. The other half (~350–400) are called endo-receptors, i.e. receptors that interact with endogenous ligands [11]. These receptors are involved in the detection of many extracellular stimuli (from photons or ions to large hormones proteins). Thus, they have important roles in various physiological systems. Dysfunction of GPCRs contributes to many human diseases and GPCRs represent 34% of all Food and Drug Administration-approved drugs [12].

GPCRs are characterized by a common structure with seven transmembrane helices with an extracellular N terminus and an intracellular C terminus [13]. The N-terminal portion, or transmembrane domain, constitute the ligand binding site while the C-terminal portion and the intracellular loops form a coupling domain with the intracellular effectors [14].

In the classical GPCR signaling pathway, after ligand binding, activated-GPCR binds the intracellular heterotrimeric G proteins, promoting the release of GDP from the Gα subunit, exchanged for GTP and the dissociation of the GTP-bound α subunit from βγ dimers. The activated G proteins can then transduce and amplify GPCR signals via second messengers to produce a variety of cell responses [15]. Briefly, Gαs activates adenylyl cyclases to catalyze the conversion of ATP to cAMP. Members of the Gαi family primarily inhibit cAMP production. The Gαq/11 family converts phosphatidylinositol 4,5-bisphosphate to diacylglycerol and inositol 1,4,5-trisphosphate to activate Protein Kinase C and increases intracellular Ca2+ levels. Approximately 10% of GPCRs can be coupled with different types of Gs subunit depending on cell type and context [16]. The second messengers then target other enzymes such as cAMP-dependent protein kinase A (PKA), GMP-dependent protein kinase G (PKG), Ca2+-dependent protein kinase C (PKC) or calcium-sensitive enzymes. The Gβγ subunit can also activate a multitude of effectors (GRKs, ion channels, PI3K, phospholipases, MAP kinases) to induce a variety of cellular effects [17]. G protein-mediated signaling is discontinued when the Gα subunit hydrolyzes GTP to GDP, due to its intrinsic GTPase activity. This then leads to the reassociation of Gα with Gβγ to form the inactive heterotrimer [14]. In addition to canonical signaling through heterotrimeric G proteins, some of GPCRs can use alternative modes of GPCR activation and initiate G protein–independent pathway. The main independent pathway involves a coupling with β-arrestin. Originally, β-arrestin was identified as an essential factor in the endocytosis and arrest of GPCR signaling induced by heterotrimeric G proteins. Today, other functions associated with β-arrestins are being studied and coupling to β-arrestins is increasingly described as “scaffolding” proteins involved in multiple G protein-independent signaling pathways. Indeed, in addition to clathrin, β-arrestins are able to bind to many proteins involved in different signaling pathways (Src, ERK1/2 and JNK3 kinases protein phosphatases, ubiquitin ligases…) [18]. The activation of β-arrestins signaling pathways can take place at the membrane but also in intracellular after internalization [15]. Indeed, a growing amount of evidence suggests that several molecules have not been known to be regulated by G proteins, suggesting that β-arrestin-mediated signaling pathways may be functioning in parallel with G-protein-mediated pathways enhancing GPCR signaling pathways.

The Hypothalamo-Pituitary-Gonadal axis is active in the midgestational fetus and after birth at the minipuberty but is mainly reactivate at onset of puberty. Some receptors of the HPG axis belong to the subfamily of GPCR: gonadotropin-releasing hormone receptor (GnRHR), GPR54/Kisspeptin receptor, Neurokinin B receptor (NK3R), Prokineticin receptor (PROKR2), follicle stimulating hormone receptor (FSHR), human chorionic gonadotropin/luteinizing hormone receptor (hCG/LHR) and Relaxin Family Peptide Receptor 2 (RXFP2).

The GnRH, a neuropeptidic hormone, is secreted by hypothalamic GnRH-expressing neurons into the portal blood vessels in rhythmic pulses [19]. It binds to a membrane receptor, the GnRH receptor, also known as the luteinizing hormone releasing hormone receptor (LHRHR), on pituitary gonadotropic cells and stimulates the biosynthesis and secretion of LH and FSH [19]. GnRHR is predominantly coupled to the Gq-protein [20]. GnRH/GnRHR pathway constitutes the initial step in the HPG axis and controls reproduction in both sexes. GnRH loss-of-function mutations are associated to normosmic hypogonadotropic hypogonadism [21]. GnRH neurons appear to be directly regulated by Kisspeptin-1 (KISS1), with Neurokinin B (NKB) and Prokineticin 2 (PROK2). KISS1 is a peptidic hormone mostly expressed in the hypothalamus [22]. It activates GPR54/KISS1R, which results in the activation of phospholipase C via Gq [12]. GPR54 has been described in brain regions, including hypothalamus, but also in peripheral regions [22]. Kisspeptin/GPR54 pathway has a crucial role in the onset of puberty, the regulation of sex hormone mediated secretion of FSH/LH, and in the control of fertility [22, 23]. Inactivating and activating mutations in KISS1 or GPR54 genes have been associated with hypogonadotropic hypogonadism and precocious puberty, respectively [23].

Gonadal function is under pituitary control via the gonadotropin hormones: follicle stimulating hormone (FSH) and luteinizing hormone (LH) [24]. FSH and LH are synthesized and secreted by the pituitary gonadotropic cells and work together in the reproductive system. The human chorionic gonadotropin (hCG) is secreted by the placenta and controls ovarian function during gestation. LH and hCG share the same GPCR, the hCG/LHR. The FSH and hCG/LH receptor belong to the glycoprotein-hormone receptor family. Activation of the LH and FSH receptor results in the production of intracellular cyclic AMP (cAMP) vias proteins [25, 26]. However, FSHR and LHR can also couple to several other effectors such as Gαq and β-arrestin [26, 27, 28]. FSHR is expressed in Sertoli and granulosa cells in male and female gonads, respectively, and is required for normal spermatogenesis and growth and maturation of ovarian follicles, as well as for estrogen production [29]. In women, LHR induces luteinization of granulosa cells, progesterone synthesis and corpus luteum maintenance during the luteal phase [30]. In men, LH stimulates testosterone production by Leydig cells [30].

Steroid hormones (estrogen, progesterone, and testosterone) secreted by the gonads, bind, and activate nuclear receptors. However, a membrane associated estrogen receptor (GPER) has been identified 15 years ago [31, 32]. Activation of GPER induces intracellular calcium mobilization, cAMP production and phosphorylation cascade involving ERK1/2, PKA, PI3K [33]. This receptor is implicated in many physiological functions: uterine proliferation, metabolism, cardiovascular, immune, and neural system.

More recently, the INSL3/RXFP2 system pathway was identified for its role in reproduction. Insulin-like peptide-3 (INSL3) belongs to the insulin/relaxin family of peptidic hormones [34, 35]. This hormone is mainly produced by testicular Leydig cells and the production is dependent on the state of Leydig cell differentiation [34]. INSL3 is considered as a marker for Leydig cells function. Its best characterized role is in the control of testicular descent since INSL3 gene inactivation males have bilateral cryptorchidism with testis remaining in abdominal position [36, 37].

Advertisement

3. Effects of EDCs on signaling of HPG axis G-protein coupled receptors

Effects of EDCs on the activity of HPG axis GPCR identified in the literature search are summarized in Table 1.

GPCREDCStudy modelMain results
FSHRDBP
[10−7 to 10−4 M]
Human granulosa cellsDBP increases FSHR expression[38]
DDT
[10−7 to 10−4 M]
CHO-K1 - hFSHRDDT decreases cAMP production stimulated by FSH[39]
DDE
[10−7 to 10−4 M]
Human granulosa cells from IVFDDE potentiates the FSH induced aromatase activity[40]
DDT
[10−7 to 10−5 M]
CHO-K1 - hFSHRDDT is an FSHR positive allosteric modulator[41]
BPA
[10−5 M]
CHO-K1 - hFSHRBPA decreases cAMP production stimulated by FSH[41]
hCG/LHRBPA
[10−6 M]
Human endometrial stromal cellsBPA decreases hCG/LHR expression[42]
DDT
[10−7 to 10−5 M]
CHO-K1 - hCG/LHRDDT decreases cAMP production stimulated by hCG/LHR[41]
RXFP2DEHP
[10−9 to 10−5 M]
HEK293 - hRXFP2DEHP increases cAMP production stimulated by INSL3[43]
DBP
[10−9 to 10−5 M]
DBP increases cAMP production stimulated by INSL3
BPA
[10−11 to 10−7 M]
HEK293 - hRXFP2BPA increased cAMP production stimulated by INSL3
DEHP + DBP + BPA
[10−10 to 10−6 M]
HEK293 - hRXFP2DEHP+DBP + BPA mixture decreases cAMP production stimulated by INSL3
GPERBPA
[10−6 M]
Human breast cancer cellsBPA increases GPER expression[44]
BPA
[10−12 to 10−9 M]
Human testicular seminoma cellsBPA promotes cellular proliferation via GPER activation[45]
BPA
[10−9 to 10−5 M]
HEK293 - hGPERBPA is a GPER agonist and induces the Gs protein pathway[46]
BPA
[10−9 to 10−5 M]
Human breast and lung cancer cells; cancer-associated fibroblastsBPA induces ERK1/2 activation and gene expression through GPER leading to cellular proliferation and migration[47, 48]
BPA
[10−7 to 10−4 M]
Human granulosa cellsBPA induces apoptosis via GPER activation[49]
o,p’-DDE
[10−7 to 10−6 M]
Human breast cancer cellso,p’-DDE is a GPER agonist and induces the Gs protein pathway[32, 46]

Table 1.

Experimental studies studying the effect of EDC on HPG axis GPCR signaling.

3.1 Hypothalamic hormones receptor

Currently, there are no data on the effects of EDCs on the activity of human hypothalamic hormone receptors. However, some studies have been conducted with animal models. Exposure to phthalates leads to a modulation of GnRHR expression (positive or negative depending on the studies) [50, 51], as well as an increase in its expression in rat uterus [52].

3.2 Gonadotropin hormones receptor

EDCs, like phthalates, increase the FSHR expression in human granulosa cells [38]. DDT has been shown to disturb the FSH induced-cAMP accumulation [39] and aromatase activity in human granulosa cells [40]. Recently, we showed that DDT behaves as an FSHR positive allosteric modulator [41]. DDT interacts with the receptor in the minor binding pocket in the transmembrane domain. DDT acts on the early steps of activation of the FSHR and induces an increase in FSH-stimulated cAMP production. Moreover, the binding of DDT enhances the FSHR response to hCG. The increased response to FSH in the presence of DDT and the gain of sensitivity to hCG may therefore by deleterious. In opposite, BPA is a FSHR negative allosteric modulator [41].

As for FSHR, EDCs, like BPA, disturbes the expression of hCG/LHR in human endometrial stromal cells [42]. In CHO-K1 cells stably transfected with hCG/LHR, DDT reduced the cAMP accumulation induced by hCG [39, 41] and hLH (Munier et al., Arch Toxicol, in revision). Moreover, DDT decreases the hCG- and hLH-promoted β-arrestin 2 recruitment (Munier et al., Arch Toxicol, in revision). DDT seems to act as a negative allosteric modulator of the hCG/LHR signaling.

3.3 Insl3 receptor, RXFP2

Only one study has very recently focused on the effect if EDCs on receptor signaling to INSL3: RXFP2. In a cellular model of HEK293 transiently expressing human RXFP2, individually, BPA, DEHP and DBP potentiate the cAMP response to INSL3 [43]. Because of their ubiquity, BPA, DEHP and DBP are present in many human biological fluids, as the amniotic liquid. Furthermore, everyone is chronically exposed to mixtures of environmental chemical factors resulting in toxicological interactions that cannot be predicted by reprotoxicological studies of single molecules. The combination of these three molecules, at concentrations found in human amniotic fluid, decreases the basal activity of RXFP2 as well as the response to INSL3. The structural similarity between FSHR and RXFP2 suggests that small hydrophobic molecules, like phthalates and BPA, could use the same binding sites as DDT in FSHR. The binding of one or two compounds to this site could lead to a stabilization of the active state of the receptor driving an increase of agonist activity [53]. In contrast, the binding of three compounds (DEHP+DBP + BPA) likely leads to a steric hindrance that may prevent the conformational changes necessary for the activation of RXFP2 and probably stabilize an inactive state. This study shows that in addition to individual EDC targets, HPG axis GPCRs can also be targeted by EDC cocktails.

3.4 Membrane sexual steroid hormones receptor

The G protein-coupled receptor (GPER/GPR30) is a membrane estrogen receptor [31]. Gene inactivation of GPER in mice did not induce major modifications in reproductive function [54]. However, several studies show that this receptor has pro-oncogenic effects in hormone-dependent cancers. Although many EDCs exhibit low binding affinities to the nuclear ERs and often require relatively high concentrations (>1 μM) to affect genomic pathways, several studies have focused on non-genomic signaling mediated by GPER [55].

Various DDT derivatives and BPA bind to GPER with a Kd between 1 to 10 μM and are competitors of E2 [46]. The binding affinity of EDCs for GPER is higher than for the nuclear receptors. Nevertheless, low concentrations of o,p’DDE and BPA increased cAMP production by GPER [32, 45, 46]. BPA and phthalate (MEHP) also affect proliferation and migration in human cervical cancer cells [56], in human seminoma cells [45], human breast cancer cells and cancer-associated fibroblasts that lack nuclear ERs [47, 57] as well as the migration and invasion of lung cancer cells [48]. BPA modifies these cellular responses by modulating different intracellular signaling pathways (ERK1/2 or Akt phosphorylation, gene expression) through GPER activation. In opposite, GPER mediates BPA-induced intracellular stress generation (ROS production and calcium accumulation) and apoptosis (caspase activation and mitochondrial membrane potential decrease) in human granulosa cells [49]. Recently, it has also been shown that BPA increases GPER gene expression in breast cancer cell lines [44]. Finally, bisphenols AF and B, two substitutes of BPA, exert high estrogenic effects via GPER pathway at nanomolar concentrations [58, 59].

Advertisement

4. Effects of EDCs on the synthesis and secretion of HPG axis hormones

Effects of EDCs on the synthesis and secretion of HPG axis hormones identified in the literature search are summarized in Table 2.

Study populationEDC exposureMatrix/biomarkerMain results
192 mother–child pairs from e-waste recycling town and 70 from control areaFree BPA in cord blood serumKiss gene expression in placentaHigher BPA concentrations showed positive correlation with Kiss gene expression[60]
73 girls with central precocious puberty and 31 controlsSeven urinary phthalate metabolites concentrationsSerum kisspeptinPositive correlation between kisspeptin- and mono-n-butyl phthalate[61]
535 men (18–40 yr) living or not in pesticides contamined area.Lipid-adjusted DDE and DDT concentrationsSerum FSH, LH, T, E2Positive association between DDT or DDE with T[62]
749 Swedish (fishermen and their pregnant wife)p,p′-DDE serum levelSerum FSH, LH, T, E2Positive association between DDE and FSH or LH[63]
97 adult men living in nothern Thailandplasma levels of DDT and its metabolitesSerum FSH, LH, T, E2Negative association of E2 level with p,p’-DDE and positive association with o,p’-DDE[64]
107 males exposed to DDT in ItalyLipid-adjusted p,p’-DDE and p,p’-DDT serum concentrationSerum FSH, LH, T, E2No association with serum hormone levels[65]
604 adults (men and women) in Brazil areas exposed to pesticidesSerum concentrations of 19 pesticides including p,p’-DDT and o,p’-DDTSerum FSH, LH, T, E2In men, o,p’-DDT level was associated with lower T, in peri- and postmenopausal women, p,p’-DDT showed inverse associations with LH; No association in premenopausal women[66]
234 mothers and their sonsSerum o,p’- and p,p’-DDT, p,p’-DDE from mothers during pregnancy or at delivery and their sons at 9 years.Serum FSH, LH and T in sons at 12 yearsPrenatal maternal DDT and DDE levels were associated with decreases in LH[67]
45 girls with early breast development, 16 girls with early puberty, and 33 girls with no signs of puberty2,4-DDT and 4,4’-DDE in the serum and adipose tissue samples.Serum basal and stimulated LH and FSH levelBasal and stimulated LH were higher in girls with detectable serum DDE levels[68]
308 young menUrinary BPA concentrationsSerum LH, T, E2Higher urinary BPA concentrations were associated with increased serum T, E2, and LH[69]
215 healthy young men (18–23 yr)Urinary BPA concentrationsSerum FSH, LH, T, E2Positive association between urinary BPA and LH levels[70]
560 men aged 18–55 yearsUrinary BPA concentrationsSerum FSH, LH, TBPA was associated with increased serum levels of LH and FSH in male smokers, and with decreased serum levels of total T in men with BMI ≥ 25 kg/m2.[71]
167 men from an infertility clinicUrinary BPA concentrationsSerum FSH, LH, T, E2Positive association between urinary BPA and serum FSH[72]
244 mothers-child pairsSerum maternal total BPA concentration during second or third trimesterSerum FSH, LH, T, E2No association with serum hormone levels[73]
159 women with premature ovarian insufficiency and 186 controlsUrinary concentrations of BPASerum FSH, LHNo association with serum hormone levels[74]
106 BPA-exposed factories and 250 unexposed female workersUrinary concentrations of BPASerum FSH, LH, E2Inverse association between BPA and FSH in unexposed group[75]
143 healthy, premenopausal womenUrinary concentrations of BPASerum FSH, LH, E2No association with serum hormone levels[76]
172 peripubertal boysUrinary concentrations of BPASerum FSH, LH, TNo association with serum hormone levels[77]
130 children with Attention-Deficit/Hyperactivity Disorder and 68 controls (boys and girls)Urine levels of phthalates and BPASerum FSH, LH, T, E2Among boys with ADHD, MBzP and MEHP levels were positively correlated with T; among girls, MEP was positively correlated with LH and T[78]
136 girls (6–9 yr) with early puberty and 136 controlsUrinary BPA concentrationsSerum basal and stimulated LH and FSH level, E2In early puberty group, negative correlation between BPA and peak FSH levels[79]
479 pregnant women and their infants (boys and girls)Urinary 12 phthalate metabolites concentrations at gestational week 28Serum T, LH, FSH during mini pubertyNo association with serum hormone levels[80]
302 Korean children and adolescentsUrinary and serum concentrations of DEHP, MEHP, DBP, MBPSerum FSH, LH, T, E2Positive correlations between serum DBP or MEHP, and E2 and/or LH in children.[81]
106 males and females (11–88 yr)Urinary phthalate metabolitesSerum FSH, LH, T, E2Positive associations between MEHP and FSH or T, MEOHP and FSH, LH or T, negative associations between MEHHP and LH, FSH or T[82]
88 infertile menUrinary and serum concentrations of 11 phthalate metabolitesSerum FSH, LH, TNegative associations between FSH and MiBP and MCMHP; positive association between T and phthalates metabolites.[83]
599 infertile menUrinary concentrations of 8 phthalate metabolitesSerum FSH, LH, T, E2Inverse associations between T and MiBP, FSH and MEHHP, positive relationship between E2 and MEP, %MEHP and FSH and LH[84]
295 adult menUrinary concentrations of phthalate metabolitesSerum FSH, LH, T, E2Negative association between MBzP and FSH[85]
881 healthy menUrinary concentrations of 14 phthalate metabolitesSerum FSH, LH, T, E2%MEHP was negatively associated with T and FSH[86]
Male with cryptorchidism (421), hypospadias (109) or controls (425)5cx-MEPP, 7cx-MMEHP in amniotic fluid (11–21 weeks)INSL3, T in amniotic fluidNegative correlations between INSL3 and cx7-MMeHP and 5cxMEPP[87]
1066 Chinese men of reproductive ageUrinary concentrations of 14 phthalate metabolitesSerum levels of INSL3, FSH, LH, TNegative association between INSL3 and MEHP; negative association between MBP and MiBP with T and LH[88]
male partners of subfertile (n = 253) and fertile (n = 37) couples11 phthalate metabolites in urine and semenSerum levels of INSL3, FSH, LH, T, E2Negative association between INSL3 and some urinary and seminal phthalate metabolites[89]
case–control study of 176 men (fertile and infertile)Urinary concentrations of 11 phthalate metabolitesSerum levels of INSL3, FSH, LH, T, E2inverse association MMP, MiBP, MEHP, MEHP% and T; MBzP and MEHP% were negatively associated with serum INSL3 level[90]
102 mother–child pairsMaternal serum concentration of MEHP (23–35 weeks of gestationCord blood INSL3, FSH, LH, T, E2 levelsInverse associations between maternal MEHP and INSL3 in males[91]
52 boys with cryptorchidism and 128 control boysCord blood BPA concentration at birthCord blood INSL3 levelsHigher cord blood BPA concentrations were associated with reduced cord blood INSL3 levels[92]

Table 2.

Human biomonitoring studies addressing the relationship between EDC and hormones of HPG axis.

T: testosterone, E2: estradiol.

4.1 Hypothalamus level

4.1.1 Kisspeptin

No data are available on the impact of DDT on Kisspeptin in epidemiological studies in humans.

Interestingly, a study led on 262 mother–child pairs from China found a positive correlation between cord blood levels of BPA and KISS1 mRNA expression in placenta tissue [60].

For phthalates, linear regression analysis showed increasing trend for kisspeptin secretion with the concentration of urinary phthalates [61].

4.1.2 GnRH

No epidemiological or experimental studies are available on the possible link between EDC levels and GnRH concentration in human. This is probably explained by the pulsatile nature of its release and the lack of dosage in clinical practice. However, many effects of EDC on GnRH were observed in rodents [93].

4.2 Pituitary level

DDT is rapidly metabolized in the body to DDE. Thus, in epidemiological studies, DDE is dosed in the blood more often than DDT. In a cohort of men of reproductive age, statistically significant positive association was found between the serum level of DDE and LH or FSH [63]. However, others studies did not reveal any association between DDT and FSH or LH levels in adult men [62, 64, 65]. In peri and postmenopausal women, inverse correlation was found between serum DDT and LH [66]. Moreover, it has been shown that maternal exposure to DDT or DDE, assayed in prenatal serum, induced a reduction of plasma LH in teenage boys, not found for FSH [67]. A study also showed that the serum levels of LH (basal level and after GnRH stimulation) was significantly higher in girls with detectable serum DDE levels than in girls with undetectable DDE [68]. This difference was not found for FSH [68].

For BPA, studies found that higher urinary BPA concentration was associated with significantly higher concentrations of serum LH in healthy young men, with or without association with FSH [69, 70, 71]. However, these results were not confirmed in others cohorts of fertile men [73]. Conversely, another study found a positive correlation between urinary BPA concentration and FSH level, without change in LH level in a cohort of infertile men [72]. In women, no association was found between urinary bisphenol A and LH or FSH levels in premenopausal women [74, 75, 76]. Moreover, no association was found in healthy children for LH and FSH [77, 78]. A modest negative correlation was found between urinary BPA concentration and peak of GnRH-stimulated FSH levels in girls with idiopathic central precocious puberty, without difference for LH levels [79].

Maternal phthalates exposure (urinary samples collected during second trimester) was not associated with serum LH level or FSH in offspring during mini-puberty in boys and girls [80]. However, positive correlations were observed between different phthalates and serum LH in prepubescent Korean children (for serum DBP or MEHP) [81], in girls with attention-deficit/hyperactivity disorder (for urinary MEP) [78] and in Chinese population (11–88 years, males and females) (for urinary MEHHP levels) [82]. In the same populations, either negative [82, 83] or no effects [78, 81] were observed on FSH level. In men, urinary phthalate metabolites were positively associated with LH and FSH levels [84] in one study while negative association between urinary phthalates concentrations and levels of FSH was found in American men (for MBzP) [85] and in Danish men (for MEHP or %MiNP) [86] without impact on LH.

Altogether, epidemiological data have linked exposure to EDC and LH and/or FSH level but evidence were often inconclusive. The inconsistent findings may partly be due to differences in the characteristics and sizes of the cohorts and to the different EDC exposure levels among studies.

4.3 Gonadal level

4.3.1 Sexual steroid hormones

Many data are already available on the effect of endocrine disruptors on the secretion of sex steroids. Recent reviews list all available studies for DDT [93], BPA [93, 94] or phthalates [93, 95, 96, 97].

4.3.2 INSL3

No data are available on the impact of DDT on INSL3 in humans epidemiological studies.

Several studies showed that INSL3 was negatively impacted by putative phthalate metabolites. The Diisononyl phthalate (DiNP) metabolite, cx7-MMeHP, and the DEHP metabolite, 5cxMEPP, showed significant negative correlations with INSL3 in amniotic fluid for weeks 11–22 [87]. Moreover, serum levels of INSL3 was negatively associated with urinary concentration of mono-2-ethylhexyl phthalate (MEHP) and MBzP among large cohorts of chinese men of reproductive age [88, 89, 90]. In adjusted models, quartiles increases in phthalates metabolites correlated with significant decreases in plasma INSL3 levels [88, 89, 90]. It has also been shown that maternal serum MEHP concentration (from 23–35 weeks of gestation) was negatively correlated with INSL3 level in cord blood mainly in boys [91].

There is also an inverse correlation between BPA level and concentration of INSL3 [92]. Indeed, in a population of 180 boys born after 34 weeks of gestation (52 cryptorchid and 128 control), cord blood levels of free BPA correlated negatively with INSL3 [92]. In this study, cord blood INSL3 level was also significantly decreased in the cryptorchid group compared with the control group [92].

Ex vivo studies on human testicular explant were performed, to study more precisely the effect of endocrine disruptors on the secretion of INSL3.

No data are available on the impact of DDT on INSL3 in humans experimental studies.

The exposure of fetal testis (8–12 weeks) to BPA at 10−8 M and 10−5 M for 72 h [98], significantly depressed the basal INSL3 production compared with control. This treatment also reduced INSL3 mRNA level by more than 20% [99]. However, BPA did not modify hCG or hLH-stimulated INSL3 production [98]. Conversely, in human adult testes, BPA increased significantly INSL3 production by Leydig cells, at a low doses (10−9 M) [100]. Interestingly, its analogs, Bisphenol B and Bisphenol S also increased INSL3 production at 10−9 and 10−8 M. Moreover, BADGE, another bisphenol, dose dependently increased INSL3 after 48 h of exposure. In contrast, BPE dose dependently inhibited INSL3 levels [100].

For phthalates, di-(2-ethylhexyl) phthalate (DEHP) and mono-(2-ethylhexyl) phthalate (MEHP) exposition on organo-cultured adult human testis did not affect Leydig cell INSL3 concentrations [101].

Advertisement

5. Conclusions

Most epidemiological and experimental studies focus on the effect of EDCs on the expression and secretion of hormones, as well as on the activity of nuclear steroid receptors. However, a few experimental studies have shown that G protein-coupled membrane receptors of the HPG axis are targets of EDCs as well. It can be pointed out that most of the studies analyzing the effects of EDCs on GPCRs of HPG axis have been performed with cell culture systems. In vitro models are valuable tools because they are easily manipulated. But the comparison of the effects of EDCs in wild-type and GPCRs- inactivated animal models could provide additional informations on the mode of action of these compounds.

Mechanisms of GPCR disruption by EDCs include: (1) changes in the expression; (2) interaction with transmembrane domain receptor; (3) modulation of intracellular signaling pathways.

The GPCRs of HPG axis, involved in diverse physiological functions, should be considered as possible contributors of the adverse effects of EDCs on reproduction. How their modulation by EDCs contributes to these deleterious effects should be an important field of investigations in the near future.

Advertisement

Acknowledgments

V.S was supported by funding from La Société Française d’Endocrinologie. MM was supported by funding from La Société Française d’Endocrinologie et de Diabétologie Pédiatrique and Novo Nordisk.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Diamanti-Kandarakis E, Bourguignon J-P, Giudice LC, Hauser R, Prins GS, Soto AM, et al. Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr Rev. 2009 Jun;30(4):293-342
  2. 2. Zoeller RT, Brown TR, Doan LL, Gore AC, Skakkebaek NE, Soto AM, et al. Endocrine-Disrupting Chemicals and Public Health Protection: A Statement of Principles from The Endocrine Society. Endocrinology. 2012 Sep;153(9):4097-4110
  3. 3. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, et al. EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev. 2015 Dec;36(6):E1-150
  4. 4. Jiang D, Chen W-Q , Zeng X, Tang L. Dynamic Stocks and Flows Analysis of Bisphenol A (BPA) in China: 2000-2014. Environ Sci Technol. 2018 20;52(6):3706-15
  5. 5. Burstyn I, Martin JW, Beesoon S, Bamforth F, Li Q , Yasui Y, et al. Maternal exposure to bisphenol-A and fetal growth restriction: a case-referent study. Int J Environ Res Public Health. 2013 Dec 11;10(12):7001-7014
  6. 6. Carwile JL, Luu HT, Bassett LS, Driscoll DA, Yuan C, Chang JY, et al. Polycarbonate bottle use and urinary bisphenol A concentrations. Environ Health Perspect. 2009 Sep;117(9):1368-1372
  7. 7. Woodruff TJ, Zota AR, Schwartz JM. Environmental chemicals in pregnant women in the United States: NHANES 2003-2004. Environ Health Perspect. 2011 Jun;119(6):878-885
  8. 8. Radke EG, Braun JM, Meeker JD, Cooper GS. Phthalate exposure and male reproductive outcomes: A systematic review of the human epidemiological evidence. Environ Int. 2018;121(Pt 1):764-793
  9. 9. Hall JM, Greco CW. Perturbation of Nuclear Hormone Receptors by Endocrine Disrupting Chemicals: Mechanisms and Pathological Consequences of Exposure. Cells. 2019 19;9(1)
  10. 10. Fredriksson R, Lagerström MC, Lundin L-G, Schiöth HB. The G-Protein-Coupled Receptors in the Human Genome Form Five Main Families. Phylogenetic Analysis, Paralogon Groups, and Fingerprints. Mol Pharmacol. 2003 Jun 1;63(6):1256-1272
  11. 11. Rosenbaum DM, Rasmussen SGF, Kobilka BK. The structure and function of G-protein-coupled receptors. Nature. 2009 May 21;459(7245):356-363
  12. 12. Sriram K, Insel PA. G Protein-Coupled Receptors as Targets for Approved Drugs: How Many Targets and How Many Drugs? Mol Pharmacol. 2018 Apr 1;93(4):251-258
  13. 13. Bockaert J, Philippe Pin J. Molecular tinkering of G protein-coupled receptors: an evolutionary success. EMBO J. 1999 Apr 1;18(7):1723-1729
  14. 14. Weis WI, Kobilka BK. The Molecular Basis of G Protein–Coupled Receptor Activation. Annu Rev Biochem. 2018 Jun 20;87:897-919
  15. 15. Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci. 2018 Apr 1;39(4):367-386
  16. 16. Wong SK-F. G Protein Selectivity Is Regulated by Multiple Intracellular Regions of GPCRs. Neurosignals. 2003;12(1):1-12
  17. 17. Smrcka AV, Fisher I. G-protein βγ subunits as multi-functional scaffolds and transducers in G-protein-coupled receptor signaling. Cell Mol Life Sci. 2019 Nov 1;76(22):4447-4459
  18. 18. Peterson YK, Luttrell LM. The Diverse Roles of Arrestin Scaffolds in G Protein–Coupled Receptor Signaling. Pharmacol Rev. 2017 Jul;69(3):256-297
  19. 19. Kaprara A, Huhtaniemi IT. The hypothalamus-pituitary-gonad axis: Tales of mice and men. Metabolism. 2018 Sep 1;86:3-17
  20. 20. Heitman LH, IJzerman AP. G protein-coupled receptors of the hypothalamic–pituitary–gonadal axis: A case for gnrh, LH, FSH, and GPR54 receptor ligands. Med Res Rev. 2008;28(6):975-1011
  21. 21. Young J, Xu C, Papadakis GE, Acierno JS, Maione L, Hietamäki J, et al. Clinical Management of Congenital Hypogonadotropic Hypogonadism. Endocr Rev. 2019 Apr 1;40(2):669-710
  22. 22. Trevisan CM, Montagna E, de Oliveira R, Christofolini DM, Barbosa CP, Crandall KA, et al. Kisspeptin/GPR54 System: What Do We Know About Its Role in Human Reproduction? Cell Physiol Biochem. 2018;49(4):1259-1276
  23. 23. Franssen D, Tena-Sempere M. The kisspeptin receptor: A key G-protein-coupled receptor in the control of the reproductive axis. Best Pract Res Clin Endocrinol Metab. 2018 Apr 1;32(2):107-123
  24. 24. Cahoreau C, Klett D, Combarnous Y. Structure–Function Relationships of Glycoprotein Hormones and Their Subunits’ Ancestors. Front Endocrinol [Internet]. 2015 Feb 26 [cited 2020 Oct 13];6. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4341566/
  25. 25. Minegishi T, Igarashi S, Nakamura K, Nakamura M, Tano M, Shinozaki H, et al. Functional expression of the recombinant human FSH receptor. J Endocrinol. 1994 May;141(2):369-375
  26. 26. Gudermann T, Birnbaumer M, Birnbaumer L. Evidence for dual coupling of the murine luteinizing hormone receptor to adenylyl cyclase and phosphoinositide breakdown and Ca2+ mobilization. Studies with the cloned murine luteinizing hormone receptor expressed in L cells. J Biol Chem. 1992 May 3;267(7):4479-4488
  27. 27. Ayoub MA, Landomiel F, Gallay N, Jégot G, Poupon A, Crépieux P, et al. Assessing Gonadotropin Receptor Function by Resonance Energy Transfer-Based Assays. Front Endocrinol [Internet]. 2015 [cited 2020 Oct 14];6. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4550792/
  28. 28. Gloaguen P, Crépieux P, Heitzler D, Poupon A, Reiter E. Mapping the Follicle-Stimulating Hormone-Induced Signaling Networks. Front Endocrinol [Internet]. 2011 [cited 2020 Oct 14];2. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3364461/
  29. 29. Siegel ET, Kim H-G, Nishimoto HK, Layman LC. The Molecular Basis of Impaired Follicle-Stimulating Hormone Action. Reprod Sci. 2013 Mar;20(3):211-233
  30. 30. Casarini L, Lispi M, Longobardi S, Milosa F, Marca AL, Tagliasacchi D, et al. LH and hCG Action on the Same Receptor Results in Quantitatively and Qualitatively Different Intracellular Signalling. PLOS ONE. 2012 Oct 5;7(10):e46682
  31. 31. Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science. 2005 Mar 11;307(5715):1625-1630
  32. 32. Thomas P, Pang Y, Filardo EJ, Dong J. Identity of an Estrogen Membrane Receptor Coupled to a G Protein in Human Breast Cancer Cells. Endocrinology. 2005 Feb 1;146(2):624-632
  33. 33. Prossnitz ER, Maggiolini M. Mechanisms of estrogen signaling and gene expression via GPR30. Mol Cell Endocrinol. 2009 Sep 24;308(1-2):32-38
  34. 34. Bathgate R a. D, Halls ML, Westhuizen ET van der, Callander GE, Kocan M, Summers RJ. Relaxin Family Peptides and Their Receptors. Physiol Rev. 2013 Jan 1;93(1):405-480
  35. 35. Halls ML, Bathgate RAD, Sutton SW, Dschietzig TB, Summers RJ. International Union of Basic and Clinical Pharmacology. XCV. Recent Advances in the Understanding of the Pharmacology and Biological Roles of Relaxin Family Peptide Receptors 1-4, the Receptors for Relaxin Family Peptides. Pharmacol Rev. 2015 Jan 4;67(2):389-440
  36. 36. Nef S, Parada LF. Cryptorchidism in mice mutant for Insl3. Nat Genet. 1999 Jul;22(3):295-299
  37. 37. Zimmermann S, Steding G, Emmen JMA, Brinkmann AO, Nayernia K, Holstein AF, et al. Targeted Disruption of the Insl3 Gene Causes Bilateral Cryptorchidism. Mol Endocrinol. 1999 May 1;13(5):681-691
  38. 38. Ma Y, Zhang J, Zeng R, Qiao X, Cheng R, Nie Y, et al. Effects of the Dibutyl Phthalate (DBP) on the Expression and Activity of Aromatase in Human Granulosa Cell Line KGN. Ann Clin Lab Sci. 2019 Mar;49(2):175-182
  39. 39. Rossi M, Dimida A, Dell’anno MT, Trincavelli ML, Agretti P, Giorgi F, et al. The thyroid disruptor 1,1,1-trichloro-2,2-bis(p-chlorophenyl)-ethane appears to be an uncompetitive inverse agonist for the thyrotropin receptor. J Pharmacol Exp Ther. 2007 Jan;320(1):465-474
  40. 40. Younglai EV. Synergistic effects between FSH and 1,1-dichloro-2,2-bis(P-chlorophenyl)ethylene (P,P’-DDE) on human granulosa cell aromatase activity. Hum Reprod. 2004 Mar 25;19(5):1089-1093
  41. 41. Munier M, Grouleff J, Gourdin L, Fauchard M, Chantreau V, Henrion D, et al. In Vitro Effects of the Endocrine Disruptor p,p’DDT on Human Follitropin Receptor. Environ Health Perspect. 2016 Feb 19;
  42. 42. Mannelli C, Szóstek AZ, Lukasik K, Carotenuto C, Ietta F, Romagnoli R, et al. Bisphenol A modulates receptivity and secretory function of human decidual cells: an in vitro study. Reprod Camb Engl. 2015 Aug;150(2):115-125
  43. 43. Suteau V, Briet C, Lebeault M, Gourdin L, Henrion D, Rodien P, et al. Human amniotic fluid-based exposure levels of phthalates and bisphenol A mixture reduce INSL3/RXFP2 signaling. Environ Int. 2020 May;138:105585
  44. 44. Castillo-Sanchez R, Ramirez-Ricardo J, Martinez-Baeza E, Cortes-Reynosa P, Candanedo-Gonzales F, Gomez R, et al. Bisphenol A induces focal adhesions assembly and activation of FAK, Src and ERK2 via GPER in MDA-MB-231 breast cancer cells. Toxicol Vitro Int J Publ Assoc BIBRA. 2020 Aug;66:104871
  45. 45. Bouskine A, Nebout M, Brücker-Davis F, Benahmed M, Fenichel P. Low doses of bisphenol A promote human seminoma cell proliferation by activating PKA and PKG via a membrane G-protein-coupled estrogen receptor. Environ Health Perspect. 2009 Jul;117(7):1053-1058
  46. 46. Thomas P, Dong J. Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: a potential novel mechanism of endocrine disruption. J Steroid Biochem Mol Biol. 2006 Dec;102(1-5):175-9
  47. 47. Pupo M, Pisano A, Lappano R, Santolla MF, De Francesco EM, Abonante S, et al. Bisphenol A Induces Gene Expression Changes and Proliferative Effects through GPER in Breast Cancer Cells and Cancer-Associated Fibroblasts. Environ Health Perspect. 2012 Aug;120(8):1177-1182
  48. 48. Zhang K-S, Chen H-Q , Chen Y-S, Qiu K-F, Zheng X-B, Li G-C, et al. Bisphenol A stimulates human lung cancer cell migration via upregulation of matrix metalloproteinases by GPER/EGFR/ERK1/2 signal pathway. Biomed Pharmacother. 2014 Oct 1;68(8):1037-1043
  49. 49. Huang M, Huang M, Li X, Liu S, Fu L, Jiang X, et al. Bisphenol A induces apoptosis through GPER-dependent activation of the ROS/Ca2+-ASK1-JNK pathway in human granulosa cell line KGN. Ecotoxicol Environ Saf. 2021 Jan 15;208:111429
  50. 50. Chen X, Li L, Li H, Guan H, Dong Y, Li X, et al. Prenatal exposure to di-n-butyl phthalate disrupts the development of adult Leydig cells in male rats during puberty. Toxicology. 2017 Jul 1;386:19-27
  51. 51. Liu T, Li N, Zhu J, Yu G, Guo K, Zhou L, et al. Effects of di-(2-ethylhexyl) phthalate on the hypothalamus-pituitary-ovarian axis in adult female rats. Reprod Toxicol Elmsford N. 2014 Jul;46:141-147
  52. 52. Liu T, Jia Y, Zhou L, Wang Q , Sun D, Xu J, et al. Effects of Di-(2-ethylhexyl) Phthalate on the Hypothalamus-Uterus in Pubertal Female Rats. Int J Environ Res Public Health. 2016 12;13(11)
  53. 53. Thal DM, Glukhova A, Sexton PM, Christopoulos A. Structural insights into G-protein-coupled receptor allostery. Nature. 2018;559(7712):45-53
  54. 54. Prossnitz ER, Hathaway HJ. What have we learned about GPER function in physiology and disease from knockout mice? J Steroid Biochem Mol Biol. 2015 Sep;153:114-126
  55. 55. Prossnitz ER, Arterburn JB. International Union of Basic and Clinical Pharmacology. XCVII. G Protein–Coupled Estrogen Receptor and Its Pharmacologic Modulators. Pharmacol Rev. 2015 Jul;67(3):505-540
  56. 56. Yang W, Tan W, Zheng J, Zhang B, Li H, Li X. MEHP promotes the proliferation of cervical cancer via GPER mediated activation of Akt. Eur J Pharmacol. 2018 Apr 5;824:11-16
  57. 57. Castillo Sanchez R, Gomez R, Perez Salazar E. Bisphenol A Induces Migration through a GPER-, FAK-, Src-, and ERK2-Dependent Pathway in MDA-MB-231 Breast Cancer Cells. Chem Res Toxicol. 2016 Mar 21;29(3):285-295
  58. 58. Cao L-Y, Ren X-M, Li C-H, Zhang J, Qin W-P, Yang Y, et al. Bisphenol AF and Bisphenol B Exert Higher Estrogenic Effects than Bisphenol A via G Protein-Coupled Estrogen Receptor Pathway. Environ Sci Technol. 2017 Oct 3;51(19):11423-11430
  59. 59. Lei B, Sun S, Zhang X, Feng C, Xu J, Wen Y, et al. Bisphenol AF exerts estrogenic activity in MCF-7 cells through activation of Erk and PI3K/Akt signals via GPER signaling pathway. Chemosphere. 2019 Apr;220:362-370
  60. 60. Xu X, Chiung YM, Lu F, Qiu S, Ji M, Huo X. Associations of cadmium, bisphenol A and polychlorinated biphenyl co-exposure in utero with placental gene expression and neonatal outcomes. Reprod Toxicol. 2015 Apr 1;52:62-70
  61. 61. Chen C-Y, Chou Y-Y, Wu Y-M, Lin C-C, Lin S-J, Lee C-C. Phthalates may promote female puberty by increasing kisspeptin activity. Hum Reprod. 2013 Oct 1;28(10):2765-2773
  62. 62. Bornman M, Delport R, Farías P, Aneck-Hahn N, Patrick S, Millar RP, et al. Alterations in male reproductive hormones in relation to environmental DDT exposure. Environ Int. 2018 Apr 1;113:281-289
  63. 63. Giwercman Aleksander, Rignell-Hydbom Anna, Toft Gunnar, Rylander Lars, Hagmar Lars, Lindh Christian, et al. Reproductive Hormone Levels in Men Exposed to Persistent Organohalogen Pollutants: A Study of Inuit and Three European Cohorts. Environ Health Perspect. 2006 Sep 1;114(9):1348-1353
  64. 64. Asawasinsopon R, Prapamontol T, Prakobvitayakit O, Vaneesorn Y, Mangklabruks A, Hock B. Plasma levels of DDT and their association with reproductive hormones in adult men from northern Thailand. Sci Total Environ. 2006 Feb 15;355(1):98-105
  65. 65. Cocco P, Loviselli A, Fadda D, Ibba A, Melis M, Oppo A, et al. Serum sex hormones in men occupationally exposed to dichloro-diphenyl-trichloro ethane (DDT) as young adults. J Endocrinol. 2004 Sep 1;182(3):391-397
  66. 66. Freire C, Koifman RJ, Sarcinelli PN, Rosa ACS, Clapauch R, Koifman S. Association between serum levels of organochlorine pesticides and sex hormones in adults living in a heavily contaminated area in Brazil. Int J Hyg Environ Health. 2014 Mar 1;217(2):370-378
  67. 67. Eskenazi B, Rauch SA, Tenerelli R, Huen K, Holland NT, Lustig RH, et al. In utero and childhood DDT, DDE, PBDE and PCBs exposure and sex hormones in adolescent boys: The CHAMACOS study. Int J Hyg Environ Health. 2017 Apr 1;220(2, Part B):364-72
  68. 68. Ozen S, Darcan S, Bayindir P, Karasulu E, Simsek DG, Gurler T. Effects of pesticides used in agriculture on the development of precocious puberty. Environ Monit Assess. 2012 Jul 1;184(7):4223-4232
  69. 69. Lassen TH, Frederiksen H, Jensen TK, Petersen JH, Joensen UN, Main KM, et al. Urinary Bisphenol A Levels in Young Men: Association with Reproductive Hormones and Semen Quality. Environ Health Perspect. 2014 May;122(5):478
  70. 70. Adoamnei E, Mendiola J, Vela-Soria F, Fernández MF, Olea N, Jørgensen N, et al. Urinary bisphenol A concentrations are associated with reproductive parameters in young men. Environ Res. 2018 Feb 1;161:122-128
  71. 71. Liang H, Xu W, Chen J, Shi H, Zhu J, Liu X, et al. The Association between Exposure to Environmental Bisphenol A and Gonadotropic Hormone Levels among Men. PLOS ONE. 2017 Jan 13;12(1):e0169217
  72. 72. Meeker JD, Calafat AM, Hauser R. Urinary Bisphenol A Concentrations in Relation to Serum Thyroid and Reproductive Hormone Levels in Men from an Infertility Clinic. Environ Sci Technol. 2010 Feb 15;44(4):1458
  73. 73. Hart RJ, Doherty DA, Keelan JA, Minaee NS, Thorstensen EB, Dickinson JE, et al. The impact of antenatal Bisphenol A exposure on male reproductive function at 20-22 years of age. Reprod Biomed Online. 2018 Mar 1;36(3):340-347
  74. 74. Li C, Cao M, Qi T, Ye X, Ma L, Pan W, et al. The association of bisphenol A exposure with premature ovarian insufficiency: a case–control study. Climacteric. 2020 Jul 16;0(0):1-6
  75. 75. Miao M, Yuan W, Yang F, Liang H, Zhou Z, Li R, et al. Associations between Bisphenol A Exposure and Reproductive Hormones among Female Workers. Int J Environ Res Public Health. 2015 Oct;12(10):13240
  76. 76. Pollack AZ, Mumford SL, Krall JR, Carmichael AE, Sjaarda LA, Perkins NJ, et al. Exposure to bisphenol A, chlorophenols, benzophenones, and parabens in relation to reproductive hormones in healthy women: A chemical mixture approach. Environ Int. 2018 Nov;120:137
  77. 77. Mustieles V, Ocón-Hernandez O, Mínguez-Alarcón L, Dávila-Arias C, Pérez-Lobato R, Calvente I, et al. Bisphenol A and reproductive hormones and cortisol in peripubertal boys: The INMA-Granada cohort. Sci Total Environ. 2018 Mar 15;618:1046-1053
  78. 78. Tsai C-S, Chou W-J, Lee S-Y, Lee M-J, Chou M-C, Wang L-J. Phthalates, Para-Hydroxybenzoic Acids, Bisphenol-A, and Gonadal Hormones’ Effects on Susceptibility to Attention-Deficit/Hyperactivity Disorder. Toxics. 2020 Sep;8(3):57
  79. 79. Chen Y, Wang Y, Ding G, Tian Y, Zhou Z, Wang X, et al. Association between bisphenol a exposure and idiopathic central precocious puberty (ICPP) among school-aged girls in Shanghai, China. Environ Int. 2018 Jun 1;115:410-416
  80. 80. Muerköster A-P, Frederiksen H, Juul A, Andersson A-M, Jensen RC, Glintborg D, et al. Maternal phthalate exposure associated with decreased testosterone/LH ratio in male offspring during mini-puberty. Odense Child Cohort. Environ Int. 2020 Nov 1;144:106025
  81. 81. Hyun Kim D, Min Choi S, Soo Lim D, Roh T, Jun Kwack S, Yoon S, et al. Risk assessment of endocrine disrupting phthalates and hormonal alterations in children and adolescents. J Toxicol Environ Health A. 2018;81(21):1150-1164
  82. 82. Zhang J, Yin W, Li P, Hu C, Wang L, Li T, et al. Interaction between diet- and exercise-lifestyle and phthalates exposure on sex hormone levels. J Hazard Mater. 2019 May 5;369:290-298
  83. 83. Wang B, Qin X, Xiao N, Yao Y, Duan Y, Cui X, et al. Phthalate exposure and semen quality in infertile male population from Tianjin, China: Associations and potential mediation by reproductive hormones. Sci Total Environ. 2020 Nov 20;744:140673
  84. 84. Al-Saleh I, Coskun S, Al-Doush I, Al-Rajudi T, Abduljabbar M, Al-Rouqi R, et al. The relationships between urinary phthalate metabolites, reproductive hormones and semen parameters in men attending in vitro fertilization clinic. Sci Total Environ. 2019 Mar 25;658:982-995
  85. 85. Duty SM, Calafat AM, Silva MJ, Ryan L, Hauser R. Phthalate exposure and reproductive hormones in adult men. Hum Reprod. 2005 Mar 1;20(3):604-610
  86. 86. Joensen UN, Frederiksen H, Jensen MB, Lauritsen MP, Olesen IA, Lassen TH, et al. Phthalate Excretion Pattern and Testicular Function: A Study of 881 Healthy Danish Men. Environ Health Perspect. 2012 Oct;120(10):1397-1403
  87. 87. Anand-Ivell R, Cohen A, Nørgaard-Pedersen B, Jönsson BAG, Bonde J-P, Hougaard DM, et al. Amniotic Fluid INSL3 Measured During the Critical Time Window in Human Pregnancy Relates to Cryptorchidism, Hypospadias, and Phthalate Load: A Large Case–Control Study. Front Physiol [Internet]. 2018 Apr 24 [cited 2020 Oct 6];9. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5928321/
  88. 88. Pan Y, Jing J, Dong F, Yao Q , Zhang W, Zhang H, et al. Association between phthalate metabolites and biomarkers of reproductive function in 1066 Chinese men of reproductive age. J Hazard Mater. 2015 Dec 30;300:729-736
  89. 89. Chang W-H, Wu M-H, Pan H-A, Guo P-L, Lee C-C. Semen quality and insulin-like factor 3: Associations with urinary and seminal levels of phthalate metabolites in adult males. Chemosphere. 2017 Apr;173:594-602
  90. 90. Chang W-H, Li S-S, Wu M-H, Pan H-A, Lee C-C. Phthalates might interfere with testicular function by reducing testosterone and insulin-like factor 3 levels. Hum Reprod. 2015 Nov 1;30(11):2658-2670
  91. 91. Araki A, Mitsui T, Miyashita C, Nakajima T, Naito H, Ito S, et al. Association between Maternal Exposure to di(2-ethylhexyl) Phthalate and Reproductive Hormone Levels in Fetal Blood: The Hokkaido Study on Environment and Children’s Health. PLOS ONE. 2014 Oct 8;9(10):e109039
  92. 92. Chevalier N, Brucker-Davis F, Lahlou N, Coquillard P, Pugeat M, Pacini P, et al. A negative correlation between insulin-like peptide 3 and bisphenol A in human cord blood suggests an effect of endocrine disruptors on testicular descent during fetal development. Hum Reprod. 2015 Jan 2;30(2):447-453
  93. 93. Graceli JB, Dettogni RS, Merlo E, Niño O, da Costa CS, Zanol JF, et al. The impact of endocrine-disrupting chemical exposure in the mammalian hypothalamic-pituitary axis. Mol Cell Endocrinol. 2020 Aug 22;518:110997
  94. 94. Mustieles V, D’Cruz SC, Couderq S, Rodríguez-Carrillo A, Fini J-B, Hofer T, et al. Bisphenol A and its analogues: A comprehensive review to identify and prioritize effect biomarkers for human biomonitoring. Environ Int. 2020 Nov 1;144:105811
  95. 95. Pallotti F, Pelloni M, Gianfrilli D, Lenzi A, Lombardo F, Paoli D. Mechanisms of Testicular Disruption from Exposure to Bisphenol A and Phtalates. J Clin Med [Internet]. 2020 Feb 8 [cited 2020 Nov 12];9(2). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7074154/
  96. 96. Radke EG, Glenn BS, Braun JM, Cooper GS. Phthalate exposure and female reproductive and developmental outcomes: a systematic review of the human epidemiological evidence. Environ Int. 2019 Sep 1;130:104580
  97. 97. Radke EG, Braun JM, Meeker JD, Cooper GS. Phthalate exposure and male reproductive outcomes: A systematic review of the human epidemiological evidence. Environ Int. 2018 Dec 1;121:764-793
  98. 98. Maamar MB, Lesné L, Desdoits-Lethimonier C, Coiffec I, Lassurguère J, Lavoué V, et al. An Investigation of the Endocrine-Disruptive Effects of Bisphenol A in Human and Rat Fetal Testes. PLoS ONE [Internet]. 2015 Feb 23 [cited 2017 Jul 10];10(2). Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4338204/
  99. 99. N’Tumba-Byn T, Moison D, Lacroix M, Lecureuil C, Lesage L, Prud’homme SM, et al. Differential Effects of Bisphenol A and Diethylstilbestrol on Human, Rat and Mouse Fetal Leydig Cell Function. PLoS ONE [Internet]. 2012 Dec 17 [cited 2017 Jul 10];7(12). Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3524173/
  100. 100. Desdoits-Lethimonier C, Lesné L, Gaudriault P, Zalko D, Antignac JP, Deceuninck Y, et al. Parallel assessment of the effects of bisphenol A and several of its analogs on the adult human testis. Hum Reprod. 2017 Jul 1;32(7):1465-1473
  101. 101. Desdoits-Lethimonier C, Albert O, Le Bizec B, Perdu E, Zalko D, Courant F, et al. Human testis steroidogenesis is inhibited by phthalates. Hum Reprod. 2012 May 1;27(5):1451-1459

Written By

Valentine Suteau, Patrice Rodien and Mathilde Munier

Submitted: 23 November 2020 Reviewed: 27 January 2021 Published: 28 April 2021