Next Article in Journal
Therapeutic Effects of hiPSC-Derived Glial and Neuronal Progenitor Cells-Conditioned Medium in Experimental Ischemic Stroke in Rats
Next Article in Special Issue
Mutant p53 Mediates Sensitivity to Cancer Treatment Agents in Oesophageal Adenocarcinoma Associated with MicroRNA and SLC7A11 Expression
Previous Article in Journal
The Role of LGR4 (GPR48) in Normal and Cancer Processes
Previous Article in Special Issue
Dosage Compensation in Females with X-Linked Metabolic Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Modifications Associated with Exposure to Endocrine Disrupting Chemicals in Patients with Gestational Diabetes Mellitus

by
Mateusz Kunysz
*,†,
Olimpia Mora-Janiszewska
and
Dorota Darmochwał-Kolarz
Department of Obstetrics and Gynecology, Medical College, University of Rzeszow, 35-959 Rzeszow, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(9), 4693; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094693
Submission received: 5 April 2021 / Revised: 22 April 2021 / Accepted: 26 April 2021 / Published: 29 April 2021
(This article belongs to the Special Issue Epigenetic Mechanisms and Human Pathology 2.0)

Abstract

:
Gestational diabetes mellitus (GDM) remains a significant clinical and public health issue due to its increasing prevalence and the possibility for numerous short- and long-term complications. The growing incidence of GDM seems to coincide with the widespread use of endocrine disrupting chemicals (EDCs). The extensive production and common use of these substances in everyday life has resulted in constant exposure to harmful substances from the environment. That may result in epigenetic changes, which may manifest themselves also after many years and be passed on to future generations. It is important to consider the possible link between environmental exposure to endocrine disrupting chemicals (EDCs) during pregnancy, epigenetic mechanisms and an increased risk for developing gestational diabetes mellitus (GDM). This manuscript attempts to summarize data on epigenetic changes in pregnant women suffering from gestational diabetes in association with EDCs. There is a chance that epigenetic marks may serve as a tool for diagnostic, prognostic, and therapeutic measures.

1. Introduction

Diabetes is one of the most common metabolic diseases globally. The prevalence of pregestational and gestational diabetes continues to increase and is now at the upper end of previous estimates in Europe [1]. Gestational diabetes is detected in approximately 15% of pregnancies [2]. Although GDM usually resolves after delivery, it may have numerous, long-lasting health consequences, such as increased risk for type II diabetes mellitus (T2DM) and cardiovascular disease in the mother, and future metabolic and cardiovascular complications such as increased adiposity or even obesity, impaired glucose metabolism, hypertension, hyperlipidemia, and nonalcoholic fatty liver disease in the offspring, as well as preterm puberty, but only in girls [3,4,5,6,7]. The increasing incidence of GDM seems to coincide with the growing use of endocrine disrupting chemicals (EDCs). EDCs are a group of chemical substances that, due to their structural similarities with steroid hormones, interact with receptors for estrogens, androgens, and progesterone, thereby increasing the risk of endocrinopathy and diseases of civilization [8]. Exposure of pregnant women to EDCs might be a cause of the growing incidence of gestational diabetes, and as a consequence, increased risk of epigenetic “diabetogenic” and “obesogenic” changes in the offspring DNA, thereby escalating the risk of developing civilization diseases in subsequent generations. There are many examples in the literature that provide proofs of concept that EDCs should be treated as possible risk factors for poor fetal and maternal outcomes [9]. On the basis of existing evidence concerning the widespread effects of EDCs as well as the potential diabetogenic actions of particular EDCs, it is important to consider the possible association between environmental exposure to EDCs during pregnancy and an increased risk for developing gestational diabetes mellitus (GDM). Exposure of the maternal-fetal unit to EDCs may also, and perhaps more likely, result in epigenetic changes, which may manifest themselves immediately or after many years and be passed on to future generations. This sets in motion a vicious intergenerational cycle of metabolic diseases that influence the health of the human population [10]. In our review we attempted to emphasize that EDCs contribute to metabolic complications not only in affected mothers, but also in their offspring. In this way, pathologic changes can be transferred to subsequent generations, increasing the frequency of obesity, diabetes, and polycystic ovary syndrome, concerning even younger people. Given that some epigenetic changes are reversible, identified epigenetic marks may become important therapeutic targets, as well as potential biomarkers.

2. Patophysiological Aspects of Gestational Diabetes Mellitus

In early gestation, insulin sensitivity increases, promoting the uptake and storage of glucose to meet the energy demands of later pregnancy [11]. Increasing insulin resistance instead is one of the most important metabolic adaptations in the course of pregnancy, emerging at 14 weeks’ gestation and increasing up to two-fold by late pregnancy [12]. This process is caused by a rise in the concentration of pregnancy hormones—estrogen, progesterone, leptin, cortisol, placental lactogen, and placental growth hormone, which enable an adequate supply of glucose to the developing fetus. In most women, the increase in insulin requirements is compensated for by the growing insulin production. Evidence in animals suggests that this process results from hypertrophy and hyperplasia of pancreatic β -cells, as well as increased glucose-stimulated insulin secretion [13]. Maternal insulin sensitivity returns to pre-pregnancy levels within a few days after delivery [14].
Physiologic metabolic adaptations to pregnancy do not always function adequately during pregnancy, and both β -cell impairment (reduced β -cell mass, reduced β -cell number, β -cell dysfunction, or a mix of all three), and tissue insulin resistance are critical components of the pathophysiology of GDM. Additional factors contributing to the development of gestational diabetes have also been identified: adipose expandability, low-grade chronic inflammation, and oxidative stress [15,16,17]. Increased gut permeability is also thought to facilitate the movement of inflammatory mediators from the gut into the circulation, promoting systemic insulin resistance [18].
GDM is defined as glucose intolerance that is first diagnosed in pregnancy. Maternal hyperglycemia increases transplacental glucose transfer to the fetal circulation, resulting in overstimulation of the fetal pancreas. Physiologically insulin does not pass the placenta and the fetus begins to produce its own insulin around 9 weeks of age. Fetal hyperinsulinemia intensifies the metabolism and growth of the fetus (an overgrowth of muscle tissue, including the heart muscle, adipose tissue, and liver), increasing the demand for oxygen, especially in the last stage of pregnancy. As a consequence, fetuses from pregnancies complicated by GDM are more likely to suffer from intrauterine hypoxia and perinatal injuries resulting from excessive birth weight. The offspring of mothers with GDM may also have an increased risk for long-term sequelae. GDM increases the risk of maternal and fetal complications during pregnancy and long-lasting health consequences, such as amplifying the risk of T2DM and cardiovascular disease in the mother, and future metabolic and cardiovascular complications such as increased adiposity or even obesity [3,4,5,19], impaired glucose metabolism [6], hypertension [7], hyperlipidemia, and nonalcoholic fatty liver disease in the offspring, as well as preterm puberty, but only in girls [20,21,22,23]. GDM is associated with an enlarged risk for pre-eclampsia, birth complications such as shoulder dystocia and neonatal hypoglycemia, and an extended rate of cesarean section [24,25]. Mentioned GDM complications may represent intergenerational epigenetic inheritance, but embryonic exposure to an altered intrauterine environment also might cause an epigenetic transgenerational effect. It seems more and more likely that epigenetic factors have an important role in the complex interplay between genes and the environment. These interactions may result in the activation or deactivation of genes by epigenetic mechanisms, facilitating adaptation to environmental changes [26].
Extensive primary and secondary prophylaxis allows for early diagnosis of the disease, but a better screening strategy could result in diagnosis and treatment at a much earlier stage of pregnancy. Thus, there is strong a need to identify currently overlooked root causes of GDM and new and more accurate GDM risk biomarkers [27].

3. Overview of Epigenetics

A change in gene activity without a change in the nucleotide sequence is known as an epigenetic modification. Epigenetic modifications can be transmitted through cell division (mitotic inheritance) and through subsequent generations (meiotic inheritance) [28]. These modifications can be induced by the occurrence of some environmental factors influencing biologic systems, making them important pathogenic mechanisms in the development of complex multifactorial diseases. Intergenerational epigenetic inheritance is considered when direct environmental influence cannot be ruled out, and transgenerational epigenetic inheritance is defined as germ line-mediated inheritance of epigenetic information between generations in the absence of a direct stressor that leads to phenotypic variation. Exposure during pregnancy has a direct influence on the mother, fetus (intergenerational), and developing primordial germ cells of the growing fetus (transgenerational inheritance) [29,30], as seen on Figure 1. DNA methylation, histone modifications, noncoding RNA regulation, and chromatin remodeling are the main processes of epigenetic reprogramming, whose basic role in the milieu of the genome is to react to external and internal factors by dynamic, reversible changes in chromatin structure and gene expression [31]. Disturbances to this complementary process are believed to be the cause of a growing incidence of several multifactorial diseases, including GDM [32].
DNA methylation is the most frequently studied modification. Methylation turns off repressor elements and increases gene expression, while methylation at the promoter regions decreases gene expression. Moreover, increased expression or alternative splicing may be caused by methylation in the gene body [33]. It is mainly regulated through methylation of CpG islets. Histone modification is another type of epigenetic regulation that can influence chromatin packing and subsequently gene expression [34]. Yet another type of epigenetic regulation is micro RNA (miRNA), which is involved in the post-transcriptional regulation of gene expression. Micro RNA is a short non-coding RNA that can affect both the translation and stability of mRNA. Most often, miRNA binding to mRNA leads to translational inhibition or destabilization [35].

4. Epigenetic Regulations in Gestational Diabetes Mellitus

An aberrant intrauterine environment may induce changes in gene expression by epigenetic mechanisms, thereby affecting the development of the fetus and altering the offspring’s long-term risk for obesity and metabolic diseases such as diabetes in adulthood [36]. In utero, maternal glucose freely crosses the placenta, whereas maternal insulin does not, resulting in overstimulation of the fetal pancreas that is exposed to high glucose levels in GDM pregnancies. Increased need for insulin in the early stage of life is a likely trigger of epigenetic changes involving genes crucial for pancreatic development; β -cell development, differentiation, and function; peripheral glucose uptake; and insulin sensitivity. The transgenerational persistence of the insulin-resistant phenotype suggests that the epigenotype can be transmitted to the next generation [37,38,39].
Epigenetic modifications, although they do not change the genome as a whole, can be mitotically stable over time, causing long-term changes in gene expression [40]. The importance of epigenetic marks emerging at a very early stage of human development as potential modulators and predictors of future health and diseases was recently mentioned [40,41,42]. Periconceptional and intrauterine periods are crucial for fetal programming [43]. Epigenetic changes seem to take part in the complex interplay between genes and the environment that is related to insulin resistance, T2DM, and GDM. In fact, a growing number of studies have identified specific gene variants for susceptibility to GDM [44,45,46,47], as well as epigenetic alterations [40,41,42,48]. One of the largest groups among genes being investigated for their connection to diabetes are those linked to β -cell function and insulin secretion. The predisposition might be increased in the presence of transcription factor 7-like 2 (TCF7L2), hepatocyte nuclear factor 4 alpha (HNF4a) polymorphisms, and variants among glucokinase and glucokinase regulatory protein genes. However, the presented inherited genetic variants may be under the influence of the mentioned epigenetic mechanism. Some of the most significant studies investigating the epigenetic background of GDM focused on mechanisms concerning gene silencing or augmentation are presented below. The ATP-binding cassette transporter A1 is another gene whose demethylation reduces high-density lipoprotein concentrations and raises glucose levels in OGTT [49].
In the EPOCH study, Yang et al. [50] divided patients into two groups: offspring of GDM and non-GDM mothers. Samples of cord and peripheral blood were obtained to identify GDM-associated DNA methylation areas and assess their possible association with child metabolic outcomes. The researchers identified significant changes in 51 areas of the genome and demonstrated that the methylation of five genes is linked to GDM. They demonstrated that methylation at the differentially methylated position in SH3PXD2A was significantly positively correlated with adiposity-related outcomes: BMI; waist circumference; triceps, suprailiac, and subscapular skinfold thickness; subcutaneous adipose tissue quantity; and leptin levels. DNA methylation in E2F6 was also associated with fasting insulin and the homeostatic model assessment for insulin resistance (Homa-IR). According to the researchers, these findings suggest that DNA methylation is affected by GDM exposure in utero and epigenetic changes may represent a significant link between this exposure and childhood obesity. SH3PXD2A hypomethylation was associated with GDM exposure in cord blood at birth [51], and with T2DM in pancreatic islets [52]. Furthermore, a CpG site within its homologue SH3PXD2B is also hypomethylated in severe childhood obesity [53]. Although the biologic function of the SH3PXD2A involvement in diabetes and obesity remains unclear, these multiple studies strongly suggest that it plays a crucial role in the molecular basis of metabolic disorders. As reported by Weng et al., 37 methylated CpGs (representing 20 genes) between the GDM and healthy groups were identified and showed potential as clinical biomarkers for GDM, suggesting it has epigenetic effects on genes that are preferentially involved in the Type I diabetes mellitus pathway, immune MHC-related pathways and neuron development-related pathways. The analysis was performed on umbilical cord blood [54]. Ruchat et al. demonstrated that differentially methylated genes identified in the placenta and in cord blood were also correlated with newborn weight [48]. A recent study conducted by Chen et al. identified differentially methylated CpGs in 39 genomic regions influenced by in utero exposure to GDM in offspring’s peripheral blood [55]. Methylation at three sites was also nominally associated with insulin secretion, while a fourth site was associated with a future risk of T2DM. Complex interactions between genetic (probably polygenic) susceptibility, unfavorable fetal surroundings, and the environmental impact of chemicals such as EDCs may lead to activation or deactivation of genes by epigenetic mechanisms, enabling adaptation (to some extent) to miscellaneous environmental situations, but sometimes bringing about the development of various disorders.
Epigenetic modifications have become a probable link between the in utero exposure to a diabetic environment and poor outcomes of the offspring. The association of GDM and diabetes during pregnancy with the epigenetic changes detected in in utero-exposed children has been confirmed by many others [40,41,42,55,56,57].
Wu et al. found significant differences in methylation patterns among women who developed GDM compared with those who did not [58]. Moreover, another study conducted by Reichetzeder et al. revealed that patients who develop GDM had higher levels of DNA methylation in placental tissues [59]. According to Michalczyk et al., the histone H3K27 and H3K4 demethylation levels are correlated with GDM progression to T2DM. The percentage of H3K27 and H3K4 methylation was lower in women who develop T2DM later in life than in women who are unaffected by T2DM post-GDM [60]. Another study performed by Wander et al. showed that miR-155-5p and miR-21-3p plasma levels in early pregnancy are associated with a higher risk of GDM. The miRNAs miR-21-3p and miR-210-3p were also linked to GDM, but only in overweight women. Moreover, pregnant women with male fetuses had a higher risk of GDM because of interferences in the plasma levels of miR-29a-3p, miR-223-3p, miR146b-5p, and miR-517-5p [61]. According to Zhao et al., pregnant women with considerably decreased expression levels of miRNAs miR-29a, miR-132, and miR-222 later developed GDM. miR-29a is involved in glucose metabolism, but the roles of miR-222 and miR-132 have yet to be determined [62].
Wu et al. [58] reported DNA methylation modifications in the blood of gravid women even before GDM was detected. They recognized a set of diversely methylated genes shared by the blood, umbilical cord, and placenta: retinol dehydrogenase 12, hook microtubule tethering protein 2, phosphoinositide-3-kinase regulatory subunit 5, constitutive photomorphogenic homolog subunit 8, coiled-coil domain containing 124, 3- hydroxyanthranilate 3,4-dioxygenase, and chromosome 5 open-reading frame 34.
Cardenas et al. [63] carried out an epigenome-wide association study on samples of placenta and plasma glucose, matching them up with 2 h post-OGTT plasma glucose levels. They discovered that plasma glucose at 2 h OGTT positively correlates with reduced DNA methylation of four CpG sites within the phosphodiesterase 4b gene. Moreover, three other CpG sites in the TNFRSF1B, LDLR, and BLM genes were found to be differentially methylated in association with maternal glucose.
Martinez-Ibarra et al. [64] explored the connection between GDM, EDCs, and miRNA. They observed higher levels of miR-9-5p, miR-29a-3p, and miR-330-3p in sera of patients with GDM compared to non-diabetic subjects. Moreover, according to Li et al., expression levels of miR-9-5p were significantly decreased in placental villous tissues and cytotrophoblast of GDM patients [65]. miR-9-5p directly targets hexokinase-2 (HK-2) and affects its expression. HK-2 was upregulated in both placental villous tissues and cytotrophoblasts from GDM patients compared with healthy women. Zhang et al. demonstrated that miR-9-5p significantly reduces the expression of glucose transporter 1 and glycolytic enzymes (HK-2, phosphofructokinase, and lactate dehydrogenase) [66]. Another study conducted by Kong et al. showed that overexpression of miR-9-5p in the serum of newly diagnosed T2DM patients was also found to reduce the expression of glucose transporter 1, hexokinase-2 (HK-2), phosphofructokinase, and lactate dehydrogenase, compared with a control group [67]. Sebastiani et al. also presented that miR-29a-3p modifies the expression of various genes involved in the insulin signaling pathway, such as HK2, and negatively regulates fatty acid oxidation through peroxisome proliferator-activated receptor γ coactivator 1- α expression. The expression levels of miR-330-3p, which is related to the proliferation and differentiation of β -cells and insulin secretion, are were high [68]. Table 1 summarizes some of the studies.

5. General Aspects of Endocrine Disrupting Chemicals

Endocrine disrupting chemicals (EDCs) are a group of chemical substances that, due to their structural similarities with steroid hormones, interact with receptors for estrogens, androgens, and progesterone, thereby increasing the risk of endocrinopathy and diseases of civilization [8]. Currently, the EDC list includes hundreds of chemical compounds and it is constantly growing. As reported by the European Union, from a total of 564 chemicals that have been suggested by various organizations in published papers or reports as being suspected EDCs, 147 are considered likely to be either persistent in the environment or produced at high volumes. A reliable estimation of the number of EDCs is practically impossible, however, due to the constant introduction of new chemical substances and the formation of their active derivatives.
EDCs comprise a heterogeneous group of synthetic and natural chemical compounds, most of which have phenol groups in their structure, conferring their affinity to the steroid hormone receptors: estrogens, progesterone, and androgens. EDCs have agonistic or antagonistic effects on nuclear receptors, which are their major targets [69]. Among them, the greatest health concern is linked with plasticizers (phthalates and bisphenol A (BPA) and its derivative bisphenol S) and pesticides: DDT (dichlorodiphenyltrichloroethane), chlorpyrifos, methoxychlor, fungicides (vinclozolin), herbicides, polychlorinated biphenyls, brominated flame retardants, and per- and poly-fluoroalkyl substances, the health effects of which are described in numerous publications [70]. These compounds enter the body by ingestion, inhalation, and absorption through the skin. EDCs were detected in body fluids: blood serum, saliva, tears, urine, stool, milk, semen, amniotic fluid, placenta, meconium, and fat tissue in tested populations around the world [71,72,73,74,75,76,77]. Leaching into the soil and groundwater, they enter into the food chain by accumulating in fish, animals, and plants. Some consumer products such as household chemicals, fabrics enriched with flame retardants, cosmetics, lotions, products with fragrance, and anti-bacterial soaps contain EDCs or are packaged in containers that can leach EDCs. Processed food can accumulate traces of EDCs that leach out of materials used in manufacturing and storage. EDCs such as lead, flame retardants, and polychlorinated biphenyls from furniture pollute household dust. Lipophilic EDCs can remain in the human body for years and be secreted from adipocytes, and then, after binding with the appropriate receptors, modify the hormonal response, whereas others are removed from the organism relatively quickly.
Daily, constant exposure to EDC mixtures in concentrations even below the established threshold tolerated by the human body for individual substances can significantly increase the risk of developing hormonal and metabolic disorders, such as obesity, diabetes, polycystic ovary syndrome, and cardiovascular disease, as well as malformations, infertility, and hormone-dependent cancers in both women and men [78,79,80,81,82,83,84,85,86,87].
Civilizational development and the growing demand for new chemical substances increases our exposure to endocrine active chemicals. The widespread production and common use of these substances in everyday life has resulted in constant exposure to harmful substances from the environment, including electronic equipment, furniture, paints, floors, and toys. Additional daily exposure occurs through the release of these substances from such commonly used items as food packaging, bottled drinks, cosmetics, receipts, clothes, food, contact lenses, and dental seals [88,89,90]. EDCs are absorbed through the skin, as well as by the consumption of contaminated food and breathing contaminated air [91]. Some EDCs may be even more prevalent in newborns and children than in adults in connection with greater consumption of specific foods and water. Additionally, ventilation rates, intestinal absorption, surface area to volume ratios, and hand-to-mouth activity are higher during infancy and puberty [92]. Breastfeeding is also associated with greater infants’ exposure to EDCs [93].
The effect of EDCs on adult organisms is different from that on developing organisms. In adult organisms, high doses of EDCs are needed for an effect, and the effect disappears when the exposure is discontinued. By contrast, in developing organisms, exposure to EDCs may often produce long-lasting effects. One possible reason that fetal exposure leads to more detrimental effects than adult exposure is the absence of adequate defense and detoxification mechanisms in prenatal life [94]. Compared with adults, the developing fetus has lower levels of cytochrome P450 enzymes, which are used by the body to metabolize environmental pharmaceuticals and chemicals [95,96].
After the identification of EDCs in the bodies of the pregnant mother and fetus [97,98,99,100], studies of their effects on fetal development and future health were initiated [101]. Several studies using model systems have demonstrated that external environmental stimuli such as EDCs can induce epigenetic mutations during gametogenesis, embryogenesis, and fetal development [31,102,103].
The pre- and perinatal period, infancy, childhood, and puberty are among the so-called critical periods of development during which the susceptibility to hormonal disorders induced by chemical stimuli is particularly high. A hostile intrauterine environment related to an adverse maternal lifestyle may be a causative factor of several metabolic adult-onset conditions, such as metabolic syndrome, type 2 diabetes mellitus (T2DM), and cardiovascular disease. Exposure of the fetus to EDCs may also, and perhaps more likely, result in epigenetic changes, which may manifest themselves after many years and be passed on to future generations. This sets in motion a vicious intergenerational cycle of metabolic diseases that influence the health of the human population [94,104,105,106].
There is a growing body of evidence that EDCs target several pathophysiologic features of GDM, such as weight gain, insulin resistance, and pancreatic β -cell function [84,107,108]. EDCs influence the activity of peroxisome proliferator-activated receptors, which are crucially involved in glucose metabolism and energy homeostasis. Regarding T2DM, animal studies indicate that some EDCs directly affect cells in the pancreas, adipocytes, and liver, and induce insulin resistance and hyperinsulinemia. These can also be associated with modified levels of adiponectin and leptin, often in the absence of obesity. This diabetogenic activity enhances the risk for cardiovascular diseases, and hyperinsulinemia augments the probability of diet-induced obesity. Exposure of pregnant women to EDCs can be a cause of the growing incidence of gestational diabetes, and as a consequence, increased risk of epigenetic “diabetogenic” and “obesogenic” changes in the offspring DNA, thereby increasing the risk of developing civilization diseases in subsequent generations. Many EDCs have been linked to metabolic effects in cell-based, animal, and epidemiologic studies.
One the most investigated EDCs in relation to its effects on the feto-maternal compartment is BPA (bisphenol A). BPA has been used worldwide since the 1960s to produce polycarbonate plastics (hard, transparent, with high thermal and mechanical resistance), but it is also used in epoxy resins and thermal paper. BPA can be found in everyday items such as compact discs and digital video discs, electronic equipment, and toys. Exposure already occurs in the prenatal period, then continues during lactation and through food consumed by the newborn. It is commonly detected in human blood and urine; regardless of age or sex, BPA enters the human body through the respiratory, digestive, and transdermal routes [109].
BPA is endocrine-active. Even small amounts of BPA may be toxic to the endocrine system [110]. The best known and documented BPA-related disorders are: increased likelihood of breast, uterine, and prostate cancer; impaired fertility, premature births, and miscarriages; and in vitro fertilization failure [111,112,113,114,115,116,117]. Exposure to BPA may also increase the risk of developing diabetes and obesity [118,119,120]. BPA exerts an estrogenic effect by activating estrogen receptors α and β . These receptors are ubiquitous in tissues and are involved in the regulation of blood glucose levels [121].
Phthalates are diesters of 1,2-benzendicarboxylic acid. They are widely used primarily in PVC, as general-purpose plasticizers in polymers. Day-to-day products containing phthalates are wall coverings, sealants, cables, floorings, coatings, adhesive paints, packaging materials, toys, roofings lacquers, and clothing [122].
Their absorption occurs through inhalation, ingestion or dermal absorption. Moreover, phthalates can cross the placenta, resulting in exposure to the fetus [123,124].
Parabens are another group of chemicals with endocrine activity. Chemically, they are esters of p-hydroxybenzoic acid with alkyl substituents ranging from methyl to butyl or benzyl groups. The most frequently applied are methylparaben (MeP) and propylparaben (PrP), ethylparaben (EtP), butylparaben (BtP), and benzylparaben (BenzylP). They are generally used as powerful antimicrobial substances and preservatives involved mainly in personal care products and pharmaceutic products, but also in a number of industrial products and foodstuffs. Parabens can be absorbed through ingestion, inhalation, and the skin. Parabens may cumulate in the body as a result of day-to-day use of paraben-containing products [125,126,127,128,129].

6. Association between Endocrine Disrupting Chemicals and Gestational Diabetes Mellitus

BPA may increase the risk of diabetes by altering the function of pancreatic β -cells and the action of insulin in target tissues. BPA disrupts pancreatic cell function, which leads to a failure of compensatory mechanisms and the development of hyperglycemia [130,131,132]. Wei et al. [133] showed that BPA promotes disturbances in insulin secretion in pancreatic cells that are associated with inhibition of pancreatic and duodenal homeobox 1 and decreased expression of miR-338.
Martinez-Ibarra et al. found high unadjusted mono(2-ethylhexyl) phthalate (MEHP) levels and high creatinine-adjusted BPA levels in non-diabetics compared to women with GDM. These findings could be explained by the similarities in the molecular structures of BPA and 17 β -estradiol. They found no correlation, however, between MEHP and GDM.
Moreover, their results showed a significant positive correlation between adjusted urinary levels of mono-benzyl phthalate and expression levels of miR-16-5p in non-diabetic women. Expression levels of miR-29a-3p negatively correlated with non-adjusted and adjusted urinary mono-n-butyl phthalate concentrations and unadjusted urinary mono isobutyl phthalate concentrations, and positively correlated with adjusted urinary MEHP concentrations. Unadjusted urinary BPA concentrations correlated with pre-gestational BMI in women without GDM, but not in those with GDM.
Schaffer et al. [134] found that MEP is significantly associated with increased odds of developing GDM. Moreover, they detected a positive association between mono-n-butyl phthalate and impaired glucose tolerance, and between both mono-carboxy-iso-octyl phthalate and blood glucose. By contrast, mono-(3-carboxypropyl) phthalate was inversely associated with GDM. Li et al. [135] showed that among overweight/obese pregnant women, who represent a subgroup more prone to GDM, moderately higher levels of propylparaben and total estrogenic activity of parabens were significantly associated with increasing GDM prevalence.
Moreover, a study conducted by Bellavia et al. [136] found that first trimester butylparaben and propylparaben urinary concentrations are associated with glucose levels in a cohort of pregnant women at high risk of GDM, even after adjusting for potential confounders.
In a recent study, Hou et al. [137] demonstrated that exposure to 2-tert-octylphenol (2-t-OP) is associated with a higher risk of GDM. However, higher NP exposure is associated with lower GDM risk.
On the other hand, some studies have demonstrated no adverse effects in humans. A recent meta-analysis of eight studies revealed no noteworthy association between BPA exposure and birth weight [138]. Moreover, no correlation between prenatal and early-life levels of BPA and childhood adiposity was detected [123,139]. Furthermore, there was no evidence for an association among urinary BPA concentrations and in vitro fertilization outcomes [140]. Table 2 summarizes some of the studies.

7. Discussion

As presented above, several studies have demonstrated negative effects of EDCs on glucose homeostasis in pregnant women, some leading to metabolic changes in the offspring. Unfortunately, however, many of the findings are inconsistent. The limited evidence and inconsistent results do not allow us to draw a firm conclusion. The inconsistencies in the literature may be due to differences in maternal age, BMI, the set of probes, race and ethnicity, socioeconomic status, and educational background of the study participants. Among the three pregnancy terms, the middle term seems to be the most metabolically susceptible to external influences, and therefore, the timing of sample collection must be taken into consideration.
In addition, urinary levels of EDCs might not reflect levels in the placenta or fetus, which are believed to be affected by tissue accumulation, subsequent release, and clearance. Many studies have not taken into account factors such as maternal pre-pregnancy diseases, gestational maternal weight gain, parity, hemodilution of plasma samples, dietary choices, and medications, all of which can affect the maternal environment and offspring health. Exposures to various mixtures of substances also differ across human beings, and the need for costly diagnostic technologies often limits studies of the thousands of natural and synthetic compounds with endocrine effects.
Despite these constraints, analysis of the available literature provides proofs of concept that EDCs should be treated as possible risk factors for poor fetal and maternal outcomes. Some studies provide important clues to help spot the consequences of EDCs more rapidly—mainly disorders with extended latency phases.
In general, the evidence analyzed in this essay indicates that EDCs contribute to metabolic complications not only in affected mothers, but also in their offspring. In this way, pathologic changes can be transferred to subsequent generations. These assumptions are supported by the constantly increasing frequency of obesity, diabetes, and polycystic ovary syndrome, affecting even younger people. Unfortunately, there is a lack of studies analyzing mixtures of many chemicals.
Because adverse effects on the maternal-fetal compartment are highly probable, implementation of preventive measures is crucial. Education of medical staff and the general population, particularly pregnant women, putting pressure on governments and politicians to reduce plastic production to the necessary minimum, and setting appropriate methods for the purification of industrial and municipal sewage are actions that will be beneficial for human health. There is a pressing need to minimize the occurrence of these illnesses, which can be managed by studies matching exposure to EDCs with future outcomes. The expanding evidence regarding these environmental contributors to non-communicable diseases suggests that synthetic chemicals are often ignored.

8. Conclusions

A growing body of evidence has shown that the increasing incidence and long-term metabolic consequences of GDM may be mediated by epigenetic modifications. Epigenetic marks found in GDM mothers and their offspring might be considered as future targets for potential management or early diagnostic predictors of some metabolic diseases and even tools for personalized preventive healthcare strategies. Despite inconsistent results of some studies on EDCs, it is highly probable that substances with proven negative health effects also have a detrimental impact on the maternal-fetal unit. There is a need for further multi-directional interventional and longitudinal research studies on epigenetic modifications being induced by environmental pollutants such as EDCs. An increasing number of large-scale genome-wide researches might be able to provide a large amount of essential information. Bearing in mind that these studies are complex and expensive, while waiting for their results, it is worth trying to modify the probable risk factors. Concerning that the periconceptual period, intrauterine development, and early postnatal lifetime are the most crucial for programming future human health, it seems to be valuable to undertake education, especially among women of reproductive age and in particular among pregnant women. Because epigenetic changes are modifiable, it is a chance for a reduction of intergenerational inheritance of metabolic traits and reversing the unfavorable growth trend of metabolic diseases such as diabetes, obesity, and PCOS and their long-lasting consequences.

Author Contributions

Writing—review and editing, M.K., O.M.-J. and D.D.-K.; supervision, M.K., O.M.-J. and D.D.-K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
2-t-OP2-tert-octylphenol
BPABisphenol A
EDCsEndocrine disrupting chemicals/chemicals
DDTDichlorodiphenyltrichloroethane
HNF4aHepatocyte nuclear factor 4 alpha
Homa-IRHomeostatic model assessment for insulin resistance
MCOPmono-carboxy-iso-octyl phthalate
MCPPMono-(3-carboxypropyl) phthalate
MEHPMono(2-ethylhexyl) phthalate
MEPMonoethyl phthalate
MiBPMono-isobutyl phthalate
miRNAMicroRNA
MBPMono-n-butyl phthalate
MBzPMono-benzyl phthalate
NPNonylphenol
OGTTOral glucose tolerance test
T2DMType 2 diabetes mellitus
TCF7L2Transcription factor 7-like 2

References

  1. Eades, C.E.; Cameron, D.M.; Evans, J.M. Prevalence of gestational diabetes mellitus in Europe: A meta-analysis. Diabetes Res. Clin. Pract. 2017, 129, 173–181. [Google Scholar] [CrossRef]
  2. Coustan, D.R.; Lowe, L.P.; Metzger, B.E.; Dyer, A.R. The Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study: Paving the way for new diagnostic criteria for gestational diabetes mellitus. Am. J. Obstet. Gynecol. 2010, 202, 654.e1–654.e6. [Google Scholar] [CrossRef] [Green Version]
  3. Lowe, W.L.; Scholtens, D.M.; Lowe, L.P.; Kuang, A.; Nodzenski, M.; Talbot, O.; Catalano, P.M.; Linder, B.; Brickman, W.J.; Clayton, P.; et al. Association of gestational diabetes with maternal disorders of glucose metabolism and childhood adiposity. JAMA 2018, 320, 1005–1016. [Google Scholar] [CrossRef] [PubMed]
  4. Liang, Z.; Liu, H.; Wang, L.; Song, Q.; Sun, D.; Li, W.; Leng, J.; Gao, R.; Hu, G.; Qi, L. Maternal Gestational Diabetes Mellitus Modifies the Relationship Between Genetically Determined Body Mass Index During Pregnancy and Childhood Obesity. Mayo Clin. Proc. 2020, 95, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
  5. Lowe, W.L.; Lowe, L.P.; Kuang, A.; Catalano, P.M.; Nodzenski, M.; Talbot, O.; Tam, W.H.; Sacks, D.A.; McCance, D.; Linder, B.; et al. Maternal glucose levels during pregnancy and childhood adiposity in the Hyperglycemia and Adverse Pregnancy Outcome Follow-up Study. Diabetologia 2019, 62, 598–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Davis, J.N.; Gunderson, E.P.; Gyllenhammer, L.E.; Goran, M.I. Impact of gestational diabetes mellitus on pubertal changes in adiposity and metabolic profiles in Latino offspring. J. Pediatr. 2013, 162, 741–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Miranda, J.O.; Cerqueira, R.J.; Barros, H.; Areias, J.C. Maternal diabetes mellitus as a risk factor for high blood pressure in late childhood: A prospective birth cohort study. Hypertension 2019, 73, e1–e7. [Google Scholar] [CrossRef]
  8. Rutkowska, A.; Rachoń, D.; Milewicz, A.; Ruchała, M.; Bolanowski, M.; Jędrzejuk, D.; Zgliczyński, W. Stanowisko Polskiego Towarzystwa Endokrynologicznego dotyczące związków endokrynnie czynnych (EDC). Endokrynol. Pol. 2015, 66, 276–285. [Google Scholar] [CrossRef] [Green Version]
  9. Ehrlich, S.; Lambers, D.; Baccarelli, A.; Khoury, J.; Macaluso, M.; Ho, S.M. Endocrine disruptors: A potential risk factor for gestational diabetes mellitus. Am. J. Perinatol. 2016, 33, 1313–1318. [Google Scholar]
  10. Street, M.E.; Angelini, S.; Bernasconi, S.; Burgio, E.; Cassio, A.; Catellani, C.; Cirillo, F.; Deodati, A.; Fabbrizi, E.; Fanos, V.; et al. Current knowledge on endocrine disrupting chemicals (EDCs) from animal biology to humans, from pregnancy to adulthood: Highlights from a national Italian meeting. Int. J. Mol. Sci. 2018, 19, 1647. [Google Scholar] [CrossRef] [Green Version]
  11. Di Cianni, G.; Miccoli, R.; Volpe, L.; Lencioni, C.; Del Prato, S. Intermediate metabolism in normal pregnancy and in gestational diabetes. Diabetes Metab. Res. Rev. 2003, 19, 259–270. [Google Scholar] [CrossRef] [PubMed]
  12. Catalano, P.M.; Tyzbir, E.D.; Roman, N.M.; Amini, S.B.; Sims, E.A. Longitudinal changes in insulin release and insulin resistance in nonobese pregnant women. Am. J. Obstet. Gynecol. 1991, 165, 1667–1672. [Google Scholar] [CrossRef]
  13. Parsons, J.A.; Brelje, T.C.; Sorenson, R.L. Adaptation of islets of Langerhans to pregnancy: Increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology 1992, 130, 1459–1466. [Google Scholar]
  14. Ryan, E.A.; O’Sullivan, M.J.; Skyler, J.S. Insulin action during pregnancy: Studies with the euglycemic clamp technique. Diabetes 1985, 34, 380–389. [Google Scholar] [CrossRef]
  15. Rojas-Rodriguez, R.; Lifshitz, L.M.; Bellve, K.D.; Min, S.Y.; Pires, J.; Leung, K.; Boeras, C.; Sert, A.; Draper, J.T.; Corvera, S.; et al. Human adipose tissue expansion in pregnancy is impaired in gestational diabetes mellitus. Diabetologia 2015, 58, 2106–2114. [Google Scholar] [CrossRef] [Green Version]
  16. Pantham, P.; Aye, I.L.H.; Powell, T.L. Inflammation in maternal obesity and gestational diabetes mellitus. Placenta 2015, 36, 709–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Lappas, M.; Hiden, U.; Desoye, G.; Froehlich, J.; Mouzon, S.H.D.; Jawerbaum, A. The role of oxidative stress in the pathophysiology of gestational diabetes mellitus. Antioxid. Redox Signal. 2011, 15, 3061–3100. [Google Scholar] [CrossRef]
  18. Caricilli, A.M.; Saad, M.J. The role of gut microbiota on insulin resistance. Nutrients 2013, 5, 829–851. [Google Scholar] [CrossRef] [Green Version]
  19. Lowe, W.L.; Scholtens, D.M.; Kuang, A.; Linder, B.; Lawrence, J.M.; Lebenthal, Y.; McCance, D.; Hamilton, J.; Nodzenski, M.; Talbot, O.; et al. Hyperglycemia and adverse pregnancy outcome follow-up study (HAPO FUS): Maternal gestational diabetes mellitus and childhood glucose metabolism. Diabetes Care 2019, 42, 372–380. [Google Scholar] [CrossRef] [Green Version]
  20. Grunnet, L.G.; Hansen, S.; Hjort, L.; Madsen, C.M.; Kampmann, F.B.; Thuesen, A.C.B.; Granstrømi, C.; Strøm, M.; Maslova, E.; Frikke-Schmidt, R.; et al. Adiposity, dysmetabolic traits, and earlier onset of female puberty in adolescent offspring of women with gestational diabetes mellitus: A clinical study within the Danish National Birth Cohort. Diabetes Care 2017, 40, 1746–1755. [Google Scholar] [CrossRef] [Green Version]
  21. Lauridsen, L.L.; Arendt, L.H.; Ernst, A.; Brix, N.; Parner, E.T.; Olsen, J.; Ramlau-Hansen, C.H. Maternal diabetes mellitus and timing of pubertal development in daughters and sons: A nationwide cohort study. Fertil. Steril. 2018, 110, 35–44. [Google Scholar] [CrossRef] [PubMed]
  22. Aksglaede, L.; Juul, A.; Olsen, L.W.; Sørensen, T.I. Age at puberty and the emerging obesity epidemic. PLoS ONE 2009, 4, e8450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kampmann, F.B.; Thuesen, A.C.B.; Hjort, L.; Olsen, S.F.; Pires, S.M.; Tetens, I.; Grunnet, L.G. Exposure to gestational diabetes is a stronger predictor of dysmetabolic traits in children than size at birth. J. Clin. Endocrinol. Metab. 2019, 104, 1766–1776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hunt, K.J.; Logan, S.L.; Conway, D.L.; Korte, J.E. Postpartum screening following GDM: How well are we doing? Curr. Diabetes Rep. 2010, 10, 235–241. [Google Scholar] [CrossRef]
  25. Kessous, R.; Shoham-Vardi, I.; Pariente, G.; Sherf, M.; Sheiner, E. An association between gestational diabetes mellitus and long-term maternal cardiovascular morbidity. Heart 2013, 99, 1118–1121. [Google Scholar] [CrossRef]
  26. Symonds, M.E.; Sebert, S.P.; Hyatt, M.A.; Budge, H. Nutritional programming of the metabolic syndrome. Nat. Rev. Endocrinol. 2009, 5, 604–610. [Google Scholar] [CrossRef]
  27. Vandorsten, J.P.; Dodson, W.C.; Espeland, M.A.; Grobman, W.A.; Guise, J.M.; Mercer, B.M.; Minkoff, H.L.; Poindexter, B.; Prosser, L.A.; Sawaya, G.F.; et al. NIH consensus development conference: Diagnosing gestational diabetes mellitus. NIH Consens. State-Statements 2013, 29, 1–31. [Google Scholar]
  28. Kimmins, S.; Sassone-Corsi, P. Chromatin remodelling and epigenetic features of germ cells. Nature 2005, 434, 583–589. [Google Scholar] [CrossRef]
  29. Blake, G.E.; Rakoczy, J.; Watson, E.D. Epigenetics of transgenerational inheritance of disease. In Epigenetics in Human Disease; Elsevier: Amsterdam, The Netherlands, 2018; pp. 805–836. [Google Scholar]
  30. Skinner, M.K. Environmental epigenetic transgenerational inheritance and somatic epigenetic mitotic stability. Epigenetics 2011, 6, 838. [Google Scholar] [PubMed] [Green Version]
  31. Baccarelli, A.; Bollati, V. Epigenetics and environmental chemicals. Curr. Opin. Pediatr. 2009, 21, 243. [Google Scholar] [CrossRef] [Green Version]
  32. Fernandez-Morera, J.; Rodriguez-Rodero, S.; Menendez-Torre, E.; Fraga, M. The possible role of epigenetics in gestational diabetes: Cause, consequence, or both. Obstet. Gynecol. Int. 2010, 2010, 605163. [Google Scholar] [CrossRef] [PubMed]
  33. Hernando-Herraez, I.; Garcia-Perez, R.; Sharp, A.J.; Marques-Bonet, T. DNA methylation: Insights into human evolution. PLoS Genet. 2015, 11, e1005661. [Google Scholar] [CrossRef] [PubMed]
  34. Martin, C.; Zhang, Y. The diverse functions of histone lysine methylation. Nat. Rev. Mol. Cell Biol. 2005, 6, 838–849. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, S.; Liu, M.F. Mechanisms of microRNA-mediated gene regulation. Sci. China Ser. C Life Sci. 2009, 52, 1111–1116. [Google Scholar] [CrossRef]
  36. Hanson, M.A.; Gluckman, P.D. Developmental origins of health and disease: New insights. Basic Clin. Pharmacol. Toxicol. 2008, 102, 90–93. [Google Scholar] [CrossRef]
  37. Fraga, M.F. Genetic and epigenetic regulation of aging. Curr. Opin. Immunol. 2009, 21, 446–453. [Google Scholar] [CrossRef]
  38. Pinney, S.E.; Simmons, R.A. Epigenetic mechanisms in the development of type 2 diabetes. Trends Endocrinol. Metab. 2010, 21, 223–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Simmons, R.A. Role of metabolic programming in the pathogenesis of β-cell failure in postnatal life. Rev. Endocr. Metab. Disord. 2007, 8, 95–104. [Google Scholar] [CrossRef] [PubMed]
  40. Franzago, M.; Fraticelli, F.; Stuppia, L.; Vitacolonna, E. Nutrigenetics, epigenetics and gestational diabetes: Consequences in mother and child. Epigenetics 2019, 14, 215–235. [Google Scholar] [CrossRef] [Green Version]
  41. Allard, C.; Desgagne, V.; Patenaude, J.; Lacroix, M.; Guillemette, L.; Battista, M.; Doyon, M.; Ménard, J.; Ardilouze, J.; Perron, P.; et al. Mendelian randomization supports causality between maternal hyperglycemia and epigenetic regulation of leptin gene in newborns. Epigenetics 2015, 10, 342–351. [Google Scholar] [CrossRef] [Green Version]
  42. Lesseur, C.; Armstrong, D.A.; Paquette, A.G.; Li, Z.; Padbury, J.F.; Marsit, C.J. Maternal obesity and gestational diabetes are associated with placental leptin DNA methylation. Am. J. Obstet. Gynecol. 2014, 211, 654.e1–654.e9. [Google Scholar] [CrossRef] [Green Version]
  43. Yajnik, C.S. Transmission of obesity-adiposity and related disorders from the mother to the baby. Ann. Nutr. Metab. 2014, 64, 8–17. [Google Scholar] [CrossRef] [PubMed]
  44. Schwenk, R.W.; Vogel, H.; Schürmann, A. Genetic and epigenetic control of metabolic health. Mol. Metab. 2013, 2, 337–347. [Google Scholar] [CrossRef] [PubMed]
  45. Franzago, M.; Fraticelli, F.; Nicolucci, A.; Celentano, C.; Liberati, M.; Stuppia, L.; Vitacolonna, E. Molecular analysis of a genetic variants panel related to nutrients and metabolism: Association with susceptibility to gestational diabetes and cardiometabolic risk in affected women. J. Diabetes Res. 2017, 2017, 4612623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. McCarthy, M.I. Genomics, type 2 diabetes, and obesity. N. Engl. J. Med. 2010, 363, 2339–2350. [Google Scholar] [CrossRef] [Green Version]
  47. Frayling, T.M. Genome–wide association studies provide new insights into type 2 diabetes aetiology. Nat. Rev. Genet. 2007, 8, 657–662. [Google Scholar] [CrossRef]
  48. Ruchat, S.M.; Houde, A.A.; Voisin, G.; St-Pierre, J.; Perron, P.; Baillargeon, J.P.; Gaudet, D.; Hivert, M.F.; Brisson, D.; Bouchard, L. Gestational diabetes mellitus epigenetically affects genes predominantly involved in metabolic diseases. Epigenetics 2013, 8, 935–943. [Google Scholar] [CrossRef] [Green Version]
  49. Houde, A.A.; Guay, S.P.; Desgagné, V.; Hivert, M.F.; Baillargeon, J.P.; St-Pierre, J.; Perron, P.; Gaudet, D.; Brisson, D.; Bouchard, L. Adaptations of placental and cord blood ABCA1 DNA methylation profile to maternal metabolic status. Epigenetics 2013, 8, 1289–1302. [Google Scholar] [CrossRef] [Green Version]
  50. Yang, I.V.; Zhang, W.; Davidson, E.J.; Fingerlin, T.E.; Kechris, K.; Dabelea, D. Epigenetic marks of in utero exposure to gestational diabetes and childhood adiposity outcomes: The EPOCH study. Diabet. Med. J. Br. Diabet. Assoc. 2018, 35, 612–620. [Google Scholar] [CrossRef] [PubMed]
  51. Finer, S.; Mathews, C.; Lowe, R.; Smart, M.; Hillman, S.; Foo, L.; Sinha, A.; Williams, D.; Rakyan, V.K.; Hitman, G.A. Maternal gestational diabetes is associated with genome-wide DNA methylation variation in placenta and cord blood of exposed offspring. Hum. Mol. Genet. 2015, 24, 3021–3029. [Google Scholar] [CrossRef] [PubMed]
  52. Dayeh, T.; Volkov, P.; Salö, S.; Hall, E.; Nilsson, E.; Olsson, A.H.; Kirkpatrick, C.L.; Wollheim, C.B.; Eliasson, L.; Rönn, T.; et al. Genome-wide DNA methylation analysis of human pancreatic islets from type 2 diabetic and non-diabetic donors identifies candidate genes that influence insulin secretion. PLoS Genet. 2014, 10, e1004160. [Google Scholar] [CrossRef]
  53. Huang, R.C.; Garratt, E.; Pan, H.; Wu, Y.; Davis, E.; Barton, S.; Burdge, G.; Godfrey, K.; Holbrook, J.; Lillycrop, K. Genome-wide methylation analysis identifies differentially methylated CpG loci associated with severe obesity in childhood. Epigenetics 2015, 10, 995–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Weng, X.; Liu, F.; Zhang, H.; Kan, M.; Wang, T.; Dong, M.; Liu, Y. Genome-wide DNA methylation profiling in infants born to gestational diabetes mellitus. Diabetes Res. Clin. Pract. 2018, 142, 10–18. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, P.; Piaggi, P.; Traurig, M.; Bogardus, C.; Knowler, W.C.; Baier, L.J.; Hanson, R.L. Differential methylation of genes in individuals exposed to maternal diabetes in utero. Diabetologia 2017, 60, 645–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. West, N.A.; Kechris, K.; Dabelea, D. Exposure to maternal diabetes in utero and DNA methylation patterns in the offspring. Immunometabolism 2013, 1, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Côté, S.; Gagné-Ouellet, V.; Guay, S.P.; Allard, C.; Houde, A.A.; Perron, P.; Baillargeon, J.P.; Gaudet, D.; Guérin, R.; Brisson, D.; et al. PPARGC1α gene DNA methylation variations in human placenta mediate the link between maternal hyperglycemia and leptin levels in newborns. Clin. Epigenet. 2016, 8, 1–13. [Google Scholar] [CrossRef] [Green Version]
  58. Wu, P.; Farrell, W.E.; Haworth, K.E.; Emes, R.D.; Kitchen, M.O.; Glossop, J.R.; Hanna, F.W.; Fryer, A.A. Maternal genome-wide DNA methylation profiling in gestational diabetes shows distinctive disease-associated changes relative to matched healthy pregnancies. Epigenetics 2018, 13, 122–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Reichetzeder, C.; Putra, S.D.; Pfab, T.; Slowinski, T.; Neuber, C.; Kleuser, B.; Hocher, B. Increased global placental DNA methylation levels are associated with gestational diabetes. Clin. Epigenet. 2016, 8, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Michalczyk, A.A.; Dunbar, J.A.; Janus, E.D.; Best, J.D.; Ebeling, P.R.; Ackland, M.J.; Asproloupos, D.; Ackland, M.L. Epigenetic markers to predict conversion from gestational diabetes to type 2 diabetes. J. Clin. Endocrinol. Metab. 2016, 101, 2396–2404. [Google Scholar] [CrossRef] [Green Version]
  61. Wander, P.L.; Boyko, E.J.; Hevner, K.; Parikh, V.J.; Tadesse, M.G.; Sorensen, T.K.; Williams, M.A.; Enquobahrie, D.A. Circulating early-and mid-pregnancy microRNAs and risk of gestational diabetes. Diabetes Res. Clin. Pract. 2017, 132, 1–9. [Google Scholar] [CrossRef]
  62. Zhao, C.; Dong, J.; Jiang, T.; Shi, Z.; Yu, B.; Zhu, Y.; Chen, D.; Xu, J.; Huo, R.; Dai, J.; et al. Early second-trimester serum miRNA profiling predicts gestational diabetes mellitus. PLoS ONE 2011, 6, e23925. [Google Scholar] [CrossRef]
  63. Cardenas, A.; Gagné-Ouellet, V.; Allard, C.; Brisson, D.; Perron, P.; Bouchard, L.; Hivert, M.F. Placental DNA methylation adaptation to maternal glycemic response in pregnancy. Diabetes 2018, 67, 1673–1683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Martínez-Ibarra, A.; Martínez-Razo, L.D.; Vázquez-Martínez, E.R.; Martínez-Cruz, N.; Flores-Ramírez, R.; García-Gómez, E.; López-López, M.; Ortega-González, C.; Camacho-Arroyo, I.; Cerbón, M. Unhealthy levels of phthalates and bisphenol A in Mexican pregnant women with gestational diabetes and its association to altered expression of miRNAs involved with metabolic disease. Int. J. Mol. Sci. 2019, 20, 3343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, J.; Song, L.; Zhou, L.; Wu, J.; Sheng, C.; Chen, H.; Liu, Y.; Gao, S.; Huang, W. A microRNA signature in gestational diabetes mellitus associated with risk of macrosomia. Cell. Physiol. Biochem. 2015, 37, 243–252. [Google Scholar] [CrossRef]
  66. Zhang, M.; Zhu, X. miR-9-5p plays an important role in gestational diabetes mellitus (GDM) progression by targeting HK-2. Int. J. Clin. Exp. Med. 2018, 11, 6694–6701. [Google Scholar]
  67. Kong, L.; Zhu, J.; Han, W.; Jiang, X.; Xu, M.; Zhao, Y.; Dong, Q.; Pang, Z.; Guan, Q.; Gao, L.; et al. Significance of serum microRNAs in pre-diabetes and newly diagnosed type 2 diabetes: A clinical study. Acta Diabetol. 2011, 48, 61–69. [Google Scholar] [CrossRef] [PubMed]
  68. Sebastiani, G.; Guarino, E.; Grieco, G.E.; Formichi, C.; Delli Poggi, C.; Ceccarelli, E.; Dotta, F. Circulating microRNA (miRNA) expression profiling in plasma of patients with gestational diabetes mellitus reveals upregulation of miRNA miR-330-3p. Front. Endocrinol. 2017, 8, 345. [Google Scholar] [CrossRef] [Green Version]
  69. Skinner, M.K. Role of epigenetics in developmental biology and transgenerational inheritance. Birth Defects Res. Part C Embryo Today Rev. 2011, 93, 51–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. List of Potential Endocrine Disruptors. Available online: https://endocrinedisruption.org/interactive-tools/tedx-list-of-potential-endocrine-disruptors/search-the-tedx-list (accessed on 25 March 2021).
  71. Cobellis, L.; Latini, G.; Felice, C.D.; Razzi, S.; Paris, I.; Ruggieri, F.; Mazzeo, P.; Petraglia, F. High plasma concentrations of di-(2-ethylhexyl)-phthalate in women with endometriosis. Hum. Reprod. 2003, 18, 1512–1515. [Google Scholar] [CrossRef]
  72. Ikezuki, Y.; Tsutsumi, O.; Takai, Y.; Kamei, Y.; Taketani, Y. Determination of bisphenol A concentrations in human biological fluids reveals significant early prenatal exposure. Hum. Reprod. 2002, 17, 2839–2841. [Google Scholar] [CrossRef] [Green Version]
  73. Kandaraki, E.; Chatzigeorgiou, A.; Livadas, S.; Palioura, E.; Economou, F.; Koutsilieris, M.; Palimeri, S.; Panidis, D.; Diamanti-Kandarakis, E. Endocrine disruptors and polycystic ovary syndrome (PCOS): Elevated serum levels of bisphenol A in women with PCOS. J. Clin. Endocrinol. Metab. 2011, 96, E480–E484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Main, K.M.; Mortensen, G.K.; Kaleva, M.M.; Boisen, K.A.; Damgaard, I.N.; Chellakooty, M.; Schmidt, I.M.; Suomi, A.M.; Virtanen, H.E.; Petersen, J.H.; et al. Human breast milk contamination with phthalates and alterations of endogenous reproductive hormones in infants three months of age. Environ. Health Perspect. 2006, 114, 270–276. [Google Scholar] [CrossRef] [PubMed]
  75. Barr, D.B.; Bishop, A.; Needham, L.L. Concentrations of xenobiotic chemicals in the maternal-fetal unit. Reprod. Toxicol. 2007, 23, 260–266. [Google Scholar] [CrossRef] [PubMed]
  76. Jensen, M.S.; Nørgaard-Pedersen, B.; Toft, G.; Hougaard, D.M.; Bonde, J.P.; Cohen, A.; Thulstrup, A.M.; Ivell, R.; Anand-Ivell, R.; Lindh, C.H.; et al. Phthalates and perfluorooctanesulfonic acid in human amniotic fluid: Temporal trends and timing of amniocentesis in pregnancy. Environ. Health Perspect. 2012, 120, 897–903. [Google Scholar] [CrossRef] [Green Version]
  77. Coakley, J.; Mueller, J.F.; Harden, F.; Toms, L.M.; Douwes, J. Partitioning of persistent organic pollutants (POPs) between human serum and breast milk: A literature review. Chemosphere 2012, 89, 911–918. [Google Scholar]
  78. Boisen, K.A.; Kaleva, M.; Main, K.M.; Virtanen, H.; Haavisto, A.M.; Schmidt, I.M.; Chellakooty, M.; Damgaard, I.N.; Mau, C.; Reunanen, M.; et al. High and increasing prevalence of cryptorchidism in Denmark. Ugeskr. Laeger 2004, 166, 4372–4375. [Google Scholar]
  79. Vafeiadi, M.; Roumeliotaki, T.; Myridakis, A.; Chalkiadaki, G.; Fthenou, E.; Dermitzaki, E.; Karachaliou, M.; Sarri, K.; Vassilaki, M.; Stephanou, E.G.; et al. Association of early life exposure to bisphenol A with obesity and cardiometabolic traits in childhood. Environ. Res. 2016, 146, 379–387. [Google Scholar] [CrossRef]
  80. Al-Daghri, N.M.; Abd-Alrahman, S.H.; Bukhari, I.; Panigrahy, A.; Khattak, M.N.; Alokail, M. BPA exposure is related to metabolic changes in obese Saudi children. Int. J. Clin. Exp. Pathol. 2017, 10, 9910. [Google Scholar]
  81. Rutkowska, A.Z.; Diamanti-Kandarakis, E. Polycystic ovary syndrome and environmental toxins. Fertil. Steril. 2016, 106, 948–958. [Google Scholar] [CrossRef] [Green Version]
  82. Soave, I.; Occhiali, T.; Assorgi, C.; Marci, R.; Caserta, D. Environmental toxin exposure in polycystic ovary syndrome women and possible ovarian neoplastic repercussion. Curr. Med. Res. Opin. 2020, 36, 693–703. [Google Scholar] [CrossRef]
  83. Sargis, R.M.; Simmons, R.A. Environmental neglect: Endocrine disruptors as underappreciated but potentially modifiable diabetes risk factors. Diabetologia 2019, 62, 1811–1822. [Google Scholar] [CrossRef] [Green Version]
  84. Papalou, O.; Kandaraki, E.A.; Papadakis, G.; Diamanti-Kandarakis, E. Endocrine disrupting chemicals: An occult mediator of metabolic disease. Front. Endocrinol. 2019, 10, 112. [Google Scholar] [CrossRef] [Green Version]
  85. Lauretta, R.; Sansone, A.; Sansone, M.; Romanelli, F.; Appetecchia, M. Endocrine disrupting chemicals: Effects on endocrine glands. Front. Endocrinol. 2019, 10, 178. [Google Scholar] [CrossRef] [Green Version]
  86. Menezo, Y.; Dale, B.; Elder, K. The negative impact of the environment on methylation/epigenetic marking in gametes and embryos: A plea for action to protect the fertility of future generations. Mol. Reprod. Dev. 2019, 86, 1273–1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Yilmaz, B.; Terekeci, H.; Sandal, S.; Kelestimur, F. Endocrine disrupting chemicals: Exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev. Endocr. Metab. Disord. 2020, 21, 127–147. [Google Scholar] [CrossRef] [PubMed]
  88. Pałczyński, C. Związki hormonalnie czynne: źródła narażenia i skutki zdrowotne. Alergia 2018, 3, 47–50. [Google Scholar]
  89. Żwierełło, W.; Maruszewska, A.; Skórka-Majewicz, M.; Goschorska, M.; Baranowska-Bosiacka, I.; Dec, K.; Styburski, D.; Nowakowska, A.; Gutowska, I. The influence of polyphenols on metabolic disorders caused by compounds released from plastics-Review. Chemosphere 2020, 240, 124901. [Google Scholar] [CrossRef] [PubMed]
  90. Beszterda, M.; Frański, R. Endocrine disruptor compounds in environment: As a danger for children health. Pediatr. Endocrinol. Diabetes Metab. 2018, 24, 88–95. [Google Scholar] [CrossRef] [PubMed]
  91. Rudel, R.A.; Perovich, L.J. Endocrine disrupting chemicals in indoor and outdoor air. Atmos. Environ. 2009, 43, 170–181. [Google Scholar] [CrossRef] [Green Version]
  92. Selevan, S.G.; Kimmel, C.A.; Mendola, P. Identifying critical windows of exposure for children’s health. Environ. Health Perspect. 2000, 108, 451–455. [Google Scholar] [PubMed]
  93. Grandjean, P.; Jensen, A.A. Breastfeeding and the weanling’s dilemma. Am. J. Public Health 2004, 94, 1075. [Google Scholar] [CrossRef]
  94. Bianco-Miotto, T.; Craig, J.M.; Gasser, Y.P.; van Dijk, S.J.; Ozanne, S. Epigenetics and DOHaD: From basics to birth and beyond. J. Dev. Orig. Health Dis. 2017, 8, 513–519. [Google Scholar] [CrossRef]
  95. Cresteil, T. Onset of xenobiotic metabolism in children: Toxicological implications. Food Addit. Contam. 1998, 15, 45–51. [Google Scholar] [CrossRef]
  96. Hakkola, J.; Tanaka, E.; Pelkonen, O. Developmental expression of cytochrome P450 enzymes in human liver. Pharmacol. Toxicol. 1998, 82, 209–217. [Google Scholar] [CrossRef]
  97. Lee, J.; Choi, K.; Park, J.; Moon, H.B.; Choi, G.; Lee, J.J.; Suh, E.; Kim, H.J.; Eun, S.H.; Kim, G.H.; et al. Bisphenol A distribution in serum, urine, placenta, breast milk, and umbilical cord serum in a birth panel of mother–neonate pairs. Sci. Total Environ. 2018, 626, 1494–1501. [Google Scholar] [CrossRef]
  98. Jeong, Y.; Lee, S.; Kim, S.; Park, J.; Kim, H.J.; Choi, G.; Choi, S.; Kim, S.; Kim, S.Y.; Kim, S.; et al. Placental transfer of persistent organic pollutants and feasibility using the placenta as a non-invasive biomonitoring matrix. Sci. Total Environ. 2018, 612, 1498–1505. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, X.; Wu, X.; Lei, B.; Jing, Y.; Zhang, X.; Fang, X.; Yu, Y. Transplacental transfer characteristics of organochlorine pesticides in paired maternal and cord sera, and placentas and possible influencing factors. Environ. Pollut. 2018, 233, 446–454. [Google Scholar] [CrossRef]
  100. Foster, W.G.; Gregorovich, S.; Morrison, K.M.; Atkinson, S.A.; Kubwabo, C.; Stewart, B.; Teo, K. Human maternal and umbilical cord blood concentrations of polybrominated diphenyl ethers. Chemosphere 2011, 84, 1301–1309. [Google Scholar] [CrossRef] [PubMed]
  101. Li, C.; Cheng, Y.; Tang, Q.; Lin, S.; Li, Y.; Hu, X.; Nian, J.; Gu, H.; Lu, Y.; Tang, H.; et al. The association between prenatal exposure to organochlorine pesticides and thyroid hormone levels in newborns in Yancheng, China. Environ. Res. 2014, 129, 47–51. [Google Scholar] [CrossRef] [PubMed]
  102. Tiffon, C. The impact of nutrition and environmental epigenetics on human health and disease. Int. J. Mol. Sci. 2018, 19, 3425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Uzumcu, M.; Zama, A.M.; Oruc, E. Epigenetic mechanisms in the actions of endocrine-disrupting chemicals: Gonadal effects and role in female reproduction. Reprod. Domest. Anim. 2012, 47, 338–347. [Google Scholar] [CrossRef] [PubMed]
  104. Ho, S.M.; Cheong, A.; Adgent, M.A.; Veevers, J.; Suen, A.A.; Tam, N.N.; Leung, Y.K.; Jefferson, W.N.; Williams, C.J. Environmental factors, epigenetics, and developmental origin of reproductive disorders. Reprod. Toxicol. 2017, 68, 85–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. de Aguiar Greca, S.C.; Kyrou, I.; Pink, R.; Randeva, H.; Grammatopoulos, D.; Silva, E.; Karteris, E. Involvement of the endocrine-disrupting chemical bisphenol A (BPA) in human placentation. J. Clin. Med. 2020, 9, 405. [Google Scholar] [CrossRef] [Green Version]
  106. Lee, J.E.; Jung, H.W.; Lee, Y.J.; Lee, Y.A. Early-life exposure to endocrine-disrupting chemicals and pubertal development in girls. Ann. Pediatr. Endocrinol. Metab. 2019, 24, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Filardi, T.; Panimolle, F.; Lenzi, A.; Morano, S. Bisphenol A and phthalates in diet: An emerging link with pregnancy complications. Nutrients 2020, 12, 525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Rotondo, E.; Chiarelli, F. Endocrine-Disrupting Chemicals and Insulin Resistance in Children. Biomedicines 2020, 8, 137. [Google Scholar] [CrossRef] [PubMed]
  109. Uzumcu, M.; Zachow, R. Developmental exposure to environmental endocrine disruptors: Consequences within the ovary and on female reproductive function. Reprod. Toxicol. 2007, 23, 337–352. [Google Scholar] [CrossRef] [Green Version]
  110. Sekizawa, J. Low-dose effects of bisphenol A: A serious threat to human health? J. Toxicol. Sci. 2008, 33, 389–403. [Google Scholar] [CrossRef] [Green Version]
  111. O’Connor, J.C.; Chapin, R.E. Critical evaluation of observed adverse effects of endocrine active substances on reproduction and development, the immune system, and the nervous system. Pure Appl. Chem. 2003, 75, 2099–2123. [Google Scholar] [CrossRef]
  112. Tomza-Marciniak, A.; Stępkowska, P.; Kuba, J.; Pilarczyk, B. Effect of bisphenol A on reproductive processes: A review of in vitro, in vivo and epidemiological studies. J. Appl. Toxicol. 2018, 38, 51–80. [Google Scholar] [CrossRef]
  113. Cariati, F.; D’Uonno, N.; Borrillo, F.; Iervolino, S.; Galdiero, G.; Tomaiuolo, R. Bisphenol a: An emerging threat to male fertility. Reprod. Biol. Endocrinol. 2019, 17, 1–8. [Google Scholar] [CrossRef] [Green Version]
  114. Wang, B.; Zhou, W.; Zhu, W.; Chen, L.; Wang, W.; Tian, Y.; Shen, L.; Zhang, J. Associations of female exposure to bisphenol A with fecundability: Evidence from a preconception cohort study. Environ. Int. 2018, 117, 139–145. [Google Scholar] [CrossRef]
  115. Shen, J.; Kang, Q.; Mao, Y.; Yuan, M.; Le, F.; Yang, X.; Xu, X.; Jin, F. Urinary bisphenol A concentration is correlated with poorer oocyte retrieval and embryo implantation outcomes in patients with tubal factor infertility undergoing in vitro fertilisation. Ecotoxicol. Environ. Saf. 2020, 187, 109816. [Google Scholar] [CrossRef] [PubMed]
  116. Du, Y.Y.; Guo, N.; Wang, Y.X.; Hua, X.; Deng, T.R.; Teng, X.M.; Yao, Y.C.; Li, Y.F. Urinary phthalate metabolites in relation to serum anti-Müllerian hormone and inhibin B levels among women from a fertility center: A retrospective analysis. Reprod. Health 2018, 15, 1–12. [Google Scholar] [CrossRef] [PubMed]
  117. Mínguez-Alarcón, L.; Messerlian, C.; Bellavia, A.; Gaskins, A.J.; Chiu, Y.H.; Ford, J.B.; Azevedo, A.R.; Petrozza, J.C.; Calafat, A.M.; Hauser, R.; et al. Urinary concentrations of bisphenol A, parabens and phthalate metabolite mixtures in relation to reproductive success among women undergoing in vitro fertilization. Environ. Int. 2019, 126, 355–362. [Google Scholar] [CrossRef] [PubMed]
  118. Trasande, L.; Attina, T.M.; Blustein, J. Association between urinary bisphenol A concentration and obesity prevalence in children and adolescents. JAMA 2012, 308, 1113–1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Tudurí, E.; Marroqui, L.; Dos Santos, R.S.; Quesada, I.; Fuentes, E.; Alonso-Magdalena, P. Timing of exposure and bisphenol-A: Implications for diabetes development. Front. Endocrinol. 2018, 9, 648. [Google Scholar] [CrossRef] [Green Version]
  120. Howdeshell, K.L.; Peterman, P.H.; Judy, B.M.; Taylor, J.A.; Orazio, C.E.; Ruhlen, R.L.; Vom Saal, F.S.; Welshons, W.V. Bisphenol A is released from used polycarbonate animal cages into water at room temperature. Environ. Health Perspect. 2003, 111, 1180–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Rettberg, J.R.; Yao, J.; Brinton, R.D. Estrogen: A master regulator of bioenergetic systems in the brain and body. Front. Neuroendocrinol. 2014, 35, 8–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Lorz, P.M.; Towae, F.K.; Enke, W.; Jäckh, R.; Bhargava, N.; Hillesheim, W. Phthalic acid and derivatives. In Ullmann’s Encyclopedia of Industrial Chemistry; Wiley-VCH: Weinheim, Germany, 2000. [Google Scholar]
  123. Buckley, J.P.; Herring, A.H.; Wolff, M.S.; Calafat, A.M.; Engel, S.M. Prenatal exposure to environmental phenols and childhood fat mass in the Mount Sinai Children’s Environmental Health Study. Environ. Int. 2016, 91, 350–356. [Google Scholar] [CrossRef] [Green Version]
  124. Rudel, R.A.; Gray, J.M.; Engel, C.L.; Rawsthorne, T.W.; Dodson, R.E.; Ackerman, J.M.; Rizzo, J.; Nudelman, J.L.; Brody, J.G. Food packaging and bisphenol A and bis (2-ethyhexyl) phthalate exposure: Findings from a dietary intervention. Environ. Health Perspect. 2011, 119, 914–920. [Google Scholar] [CrossRef]
  125. Liao, C.; Chen, L.; Kannan, K. Occurrence of parabens in foodstuffs from China and its implications for human dietary exposure. Environ. Int. 2013, 57, 68–74. [Google Scholar] [CrossRef]
  126. Błędzka, D.; Gromadzińska, J.; Wąsowicz, W. Parabens. From environmental studies to human health. Environ. Int. 2014, 67, 27–42. [Google Scholar] [CrossRef] [PubMed]
  127. Soni, M.; Taylor, S.; Greenberg, N.; Burdock, G. Evaluation of the health aspects of methyl paraben: A review of the published literature. Food Chem. Toxicol. 2002, 40, 1335–1373. [Google Scholar] [CrossRef]
  128. Ye, X.; Tao, L.J.; Needham, L.L.; Calafat, A.M. Automated on-line column-switching HPLC–MS/MS method for measuring environmental phenols and parabens in serum. Talanta 2008, 76, 865–871. [Google Scholar] [CrossRef]
  129. Boberg, J.; Taxvig, C.; Christiansen, S.; Hass, U. Possible endocrine disrupting effects of parabens and their metabolites. Reprod. Toxicol. 2010, 30, 301–312. [Google Scholar] [CrossRef]
  130. Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. Flaws JA Nadal A Prins GS Toppari J Zoeller RT. Executive summary to EDC-2: The Endocrine Society’s second Scientific Statement on endocrine-disrupting chemicals. Endocr. Rev. 2015, 36, 593–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Alonso-Magdalena, P.; Quesada, I.; Nadal, A. Endocrine disruptors in the etiology of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2011, 7, 346–353. [Google Scholar] [CrossRef] [PubMed]
  132. Mimoto, M.S.; Nadal, A.; Sargis, R.M. Polluted pathways: Mechanisms of metabolic disruption by endocrine disrupting chemicals. Curr. Environ. Health Rep. 2017, 4, 208–222. [Google Scholar] [CrossRef] [PubMed]
  133. Wei, J.; Ding, D.; Wang, T.; Liu, Q.; Lin, Y. MiR-338 controls BPA-triggered pancreatic islet insulin secretory dysfunction from compensation to decompensation by targeting Pdx-1. FASEB J. 2017, 31, 5184–5195. [Google Scholar] [CrossRef] [Green Version]
  134. Shaffer, R.M.; Ferguson, K.K.; Sheppard, L.; James-Todd, T.; Butts, S.; Chandrasekaran, S.; Swan, S.H.; Barrett, E.S.; Nguyen, R.; Bush, N.; et al. Maternal urinary phthalate metabolites in relation to gestational diabetes and glucose intolerance during pregnancy. Environ. Int. 2019, 123, 588–596. [Google Scholar] [CrossRef]
  135. Li, Y.; Xu, S.; Li, Y.; Zhang, B.; Huo, W.; Zhu, Y.; Wan, Y.; Zheng, T.; Zhou, A.; Chen, Z.; et al. Association between urinary parabens and gestational diabetes mellitus across prepregnancy body mass index categories. Environ. Res. 2019, 170, 151–159. [Google Scholar] [CrossRef]
  136. Bellavia, A.; Chiu, Y.H.; Brown, F.M.; Mínguez-Alarcón, L.; Ford, J.B.; Keller, M.; Petrozza, J.; Williams, P.L.; Ye, X.; Calafat, A.M.; et al. Urinary concentrations of parabens mixture and pregnancy glucose levels among women from a fertility clinic. Environ. Res. 2019, 168, 389–396. [Google Scholar] [CrossRef] [PubMed]
  137. Hou, Y.; Li, S.; Xia, L.; Yang, Q.; Zhang, L.; Zhang, X.; Liu, H.; Huo, R.; Cao, G.; Huang, C.; et al. Associations of urinary phenolic environmental estrogens exposure with blood glucose levels and gestational diabetes mellitus in Chinese pregnant women. Sci. Total Environ. 2021, 754, 142085. [Google Scholar] [CrossRef]
  138. Hu, C.Y.; Li, F.L.; Hua, X.G.; Jiang, W.; Mao, C.; Zhang, X.J. The association between prenatal bisphenol A exposure and birth weight: A meta-analysis. Reprod. Toxicol. 2018, 79, 21–31. [Google Scholar] [CrossRef] [PubMed]
  139. Braun, J.M.; Lanphear, B.P.; Calafat, A.M.; Deria, S.; Khoury, J.; Howe, C.J.; Venners, S.A. Early-life bisphenol a exposure and child body mass index: A prospective cohort study. Environ. Health Perspect. 2014, 122, 1239–1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Mínguez-Alarcón, L.; Gaskins, A.J.; Chiu, Y.H.; Williams, P.L.; Ehrlich, S.; Chavarro, J.E.; Petrozza, J.C.; Ford, J.B.; Calafat, A.M.; Hauser, R. Urinary bisphenol A concentrations and association with in vitro fertilization outcomes among women from a fertility clinic. Hum. Reprod. 2015, 30, 2120–2128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Exposure during pregnancy means direct influence on mother and fetus (intergenerational) and developing primordial germ cells of growing fetus (transgenerational inheritance).
Figure 1. Exposure during pregnancy means direct influence on mother and fetus (intergenerational) and developing primordial germ cells of growing fetus (transgenerational inheritance).
Ijms 22 04693 g001
Table 1. Epigenetic studies on gestational diabetes mellitus.
Table 1. Epigenetic studies on gestational diabetes mellitus.
StudyEffect
Weng et al. [54]Thirty-seven methylated CpGs linked to GDM
Yang et al. [50]Significant changes in 51 areas of the genome and demonstration that the methylation of 5 genes is linked to GDM
Finer et al. [51]SH3PXD2A hypomethylation is associated with GDM exposure in cord blood at birth
Chen et al. [55]Differentially methylated CpGs in 39 genomic regions influenced by in utero exposure to GDM in offspring peripheral blood
Reichetzeder et al. [59]Higher levels of DNA methylation in placental tissues in patients with gestational diabetes mellitus
Michalczyk et al. [60]Histone H3K27 and H3K4 demethylation levels are correlated with GDM progression to T2DM.
Zhao et al. [62]Decreased expression levels of miRNAs miR-29a, miR-132, and miR-222 associated with later development of GDM
Wander et al. [61]miR-155-5p and miR-21-3p plasma levels in early pregnancy are associated with a higher risk for GDM
Wu et al. [58]Differentially methylated genes shared by the blood, umbilical cord, and placenta
Cardenas et al. [63]Three CpG sites in the TNFRSF1B, LDLR, and BLM genes are differentially methylated in association with maternal glucose
Plasma glucose at 2 h OGTT positively correlates with reduced DNA methylation of 4 CpG sites within the phosphodiesterase 4b gene
Sebastiani et al. [68]Increased expression levels of miR-330-3p
Li et al. [65]miR-9-5p were significantly decreased in placental villous tissues and cytotrophoblast of GDM patients
Martinez-Ibarra et al. [64]Higher levels of miR-9-5p, miR-29a-3p, and miR-330-3p in sera of patients with GDM compared to non-diabetic subjects
Table 2. Studies on the epigentic effect on patients with gestational diabetes mellitus.
Table 2. Studies on the epigentic effect on patients with gestational diabetes mellitus.
StudyEDCs ExposureEffect
Hou et al. [137]Urinary 2-t-OPHigher risk of GDM
Urinary NPLower GDM risk
Urinary BPANo significant association with risk of GDM
Martínez-Ibarra et al. [64]Unadjusted urinary MiBP concentrationDecreased miR-29a-3p expression levels
Urinary MBP concentrationsDecreased miR-29a-3p expression levels
Urinary MEHP concentrationIncreased miR-29a-3p expression levels
Urinary MBzPIncreased miR-16-5p expression levels
Shaffer et al. [134]Urinary MCPPInversely associated with GDM
Urinary MCOPIncreased blood glucose
Urinary MBPIncreased impaired glucose tolerance
Urinary MEPIncreased blood glucose
Li et al. [135]Urinary propylparabenIncreased GDM prevalence in overweight/obese pregnant women
Bellavia et al. [136]Urinary butylparaben and propylparabenIncreased glucose levels
Hu et al. [138]Urinary BPAN/C birth weight
Braun et al. [139]Urinary BPAN/C BMI at 2–5 but accelerated growth
Buckley et al. [123]Urinary phenolsN/C influence on the development of childhood adiposity
Mínguez-Alarcón et al. [140]Urinary BPAN/C IVF outcomes
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kunysz, M.; Mora-Janiszewska, O.; Darmochwał-Kolarz, D. Epigenetic Modifications Associated with Exposure to Endocrine Disrupting Chemicals in Patients with Gestational Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 4693. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094693

AMA Style

Kunysz M, Mora-Janiszewska O, Darmochwał-Kolarz D. Epigenetic Modifications Associated with Exposure to Endocrine Disrupting Chemicals in Patients with Gestational Diabetes Mellitus. International Journal of Molecular Sciences. 2021; 22(9):4693. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094693

Chicago/Turabian Style

Kunysz, Mateusz, Olimpia Mora-Janiszewska, and Dorota Darmochwał-Kolarz. 2021. "Epigenetic Modifications Associated with Exposure to Endocrine Disrupting Chemicals in Patients with Gestational Diabetes Mellitus" International Journal of Molecular Sciences 22, no. 9: 4693. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22094693

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop