Next Article in Journal
Epigenetic Abnormalities in Chondrosarcoma
Next Article in Special Issue
Endocrine-Disrupting Chemicals and Disease Endpoints
Previous Article in Journal
Chitosan as a Promising Support of a CDH Activity Preservation System for Biomedical and Industrial Applications
Previous Article in Special Issue
A Mixture of Chemicals Found in Human Amniotic Fluid Disrupts Brain Gene Expression and Behavior in Xenopus laevis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Critical Overview on Endocrine Disruptors in Diabetes Mellitus

by
Charlotte Hinault
1,2,3,
Philippe Caroli-Bosc
1,2,3,
Frédéric Bost
1,2 and
Nicolas Chevalier
1,2,3,*
1
Université Côte d’Azur, INSERM U1065, C3M, Bâtiment Universitaire Archimed, 151 Route de Saint-Antoine de Ginestière, BP 2 3194, CEDEX 3, 06204 Nice, France
2
Équipe Labelisée Ligue Nationale Contre le Cancer
3
Université Côte d’Azur, CHU, INSERM U1065, C3M, 06202 Nice, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(5), 4537; https://doi.org/10.3390/ijms24054537
Submission received: 11 January 2023 / Revised: 15 February 2023 / Accepted: 21 February 2023 / Published: 25 February 2023
(This article belongs to the Special Issue Advances in Endocrine Disruptors 2.0)

Abstract

:
Diabetes mellitus is a major public health problem in all countries due to its high human and economic burden. Major metabolic alterations are associated with the chronic hyperglycemia that characterizes diabetes and causes devastating complications, including retinopathy, kidney failure, coronary disease and increased cardiovascular mortality. The most common form is type 2 diabetes (T2D) accounting for 90 to 95% of the cases. These chronic metabolic disorders are heterogeneous to which genetic factors contribute, but so do prenatal and postnatal life environmental factors including a sedentary lifestyle, overweight, and obesity. However, these classical risk factors alone cannot explain the rapid evolution of the prevalence of T2D and the high prevalence of type 1 diabetes in particular areas. Among environmental factors, we are in fact exposed to a growing amount of chemical molecules produced by our industries or by our way of life. In this narrative review, we aim to give a critical overview of the role of these pollutants that can interfere with our endocrine system, the so-called endocrine-disrupting chemicals (EDCs), in the pathophysiology of diabetes and metabolic disorders.

1. Introduction

Diabetes mellitus is a group of metabolic disorders that is defined phenotypically by chronic elevations in blood glucose (i.e., hyperglycemia). According to the classical definition, it results mainly either from a deficiency in insulin secretion (type 1 diabetes mellitus [T1D]) and/or from a defect in insulin action, namely insulin resistance affecting liver and peripheral tissues (type 2 diabetes mellitus [T2D]). Today the distinction between T1D and T2D is equivocal, and the classification has been revisited with many other specific diabetes subtypes. The current prevalence of diabetes is unprecedented. Ten years ago, the World Health Organization (WHO) reported that 422 million people suffered from diabetes all over the world (95% of them with type 2 diabetes) [1], whereas the same organization predicted earlier in the 2000s that 330 million people would have diabetes in 2030. The latest report of the IDF (International Diabetes Federation) estimated this number as 537 million people in the world, representing 1 in 10 adults (20–79 years) [2]. Moreover, across the world, 240 million more adult people (20–79 years) have impaired glucose tolerance, which is a predictor for the development of diabetes [2]. Importantly, due to the chronic complications arising from uncontrolled chronic hyperglycemia, diabetes imposes significant economic burdens across the world. The IDF estimated that 10% of global health expenditures are spent on diabetes, with a total annual economic cost estimated to more than USD 760 billion in the world, including a notable annual cost of USD 330 billion in the United States for example [3].
The reasons for this rapid increase in diabetes remain unclear. At the same time, the prevalence of obesity has increased in the same proportion across the world. Excess caloric consumption and a sedentary lifestyle are undoubtedly key causal factors for obesity and diabetes. However, there is growing interest in the contribution of “non-traditional” risk factors such as environmental chemicals, micronutrients, or gut microbiome to the etiology of these health conditions. Indeed, concurrently with the escalation of diabetes prevalence, the world has witnessed the massive production and release of toxicants into the environment. Many of these can interfere with the endocrine system by altering the production, release, transport, and action of hormones; these are termed endocrine-disrupting chemicals (EDCs). Since the beginning of the 2010s, a growing body of both in vitro and in vivo evidence suggests that EDCs play a significant role in the etiology of diabetes and metabolic disorders. The US National Institute of Environmental Health Sciences (NIEHS) concluded that the existing literature provided plausibility, varying from suggestive to strong, that exposure to environmental chemicals may contribute to the actual epidemic of diabetes [4]. The present review will give a critical overview of the role of EDCs in the etiology of diabetes, by discussing available epidemiological data concerning T1D and T2D, experimental data demonstrating links between chemical exposure and changes in insulin action or secretion, and by highlighting the role of EDCs on metabolic fetal programming. We systematically searched animal and human studies available up to 15 September 2022. We conducted a literature search in Scopus, ISI Web of Science and PubMed using the following keywords to identify relevant articles: “endocrine disruptors”, “chemicals”, “pollutants”, “metabolic syndrome”, “insulin resistance”, “glucose intolerance”, and “diabetes”.

2. Human Implications of Diabetogenic Pollutants

2.1. Persistent EDCs and Type 2 Diabetes

The most representative EDCs showing a well-known causal relationship with obesity and diabetes are persistent organic pollutants (POPs), which contain two main groups of synthetic compounds: polycyclic aromatic hydrocarbons and halogenated hydrocarbons. All the known POPs are listed in the Stockholm Convention (available at http://chm.pops.int/TheConvention/ThePOPs/AllPOPs/tabid/2509/Default.aspx (accessed on, 10 February 2023)). Among these, the best known are organochlorine (OCs) pesticides (dichloro-diphenyl-trichloroethane (DDT) and its metabolites chlordane) and industrial chemicals (polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDE), dioxins (TCDD: 2,3,7,8-tétrachlorodibenzo-p-dioxine), and per- and polyfluoroalkyl substances (PFAs)). These anthropic chemicals are resistant to degradation, possess low solubility in water but high solubility in lipids and tend to accumulate in the adipose tissue with the highest concentrations in humans. They have long half-lives (exceeding decades in the environment) and are not well metabolized or excreted, explaining why even small amount that are daily ingested can accumulate to yield detectable amounts over time [5]. They were involved in several main health concerns, including cancer [6], damage to neurological and reproductive systems [7], but the awareness of their metabolic concern was the result of large-scale casualties demonstrated by mass poisoning incidents.
Indeed, a chemical plant explosion in Seveso, Italy, was responsible for a massive and acute release of dioxins in the summer of 1976. Several years after this environmental disaster, follow-up studies showed an increased risk of T2D in exposed people, especially in women [8]. Interestingly, this risk was higher in the youngest women, with a significant increased risk in women aged 12 and under at the explosion (adjusted odd ratio [OR] for metabolic syndrome in women = 1.05 [95% CI 0.78–1.43]; before 12 years, adjusted OR = 2.03 [95% CI 1.25–3.30]; above 12 years, adjusted OR = 0.96; [95% CI 0.68–1.35]; p interaction = 0.01) [9]. Similarly, the study of military troops exposed to dioxins contained in the “Agent Orange” herbicide used by the US Army Corps, Operation Ranch Hand, during the Vietnam War, revealed the same association between serum TCDD concentration and the prevalence of T2D (relative risk [RR] = 1.5; [95% CI 1.2–2.0]) [10]. The consumption of adulterated rice bran cooking oil (“Yusho” oil) in a group of Taiwanese, also known as the Yu-cheng incident, was related to a higher prevalence in T2D, especially in women (OR = 2.1; [95% CI 1.1–4.5]), due to a contamination with PCB ethers, dioxins and furans [11]. A cross-sectional study of Taiwanese living in a highly dioxin-contaminated area also reported a strong correlation between abdominal obesity and insulin resistance (determined by the HOMA index) in people with the highest serum levels of dioxins (OR = 5.23; [95% CI 3.53–7.77] in men; OR = 4.57; [95% CI 2.70–7.64] in women) [12]. However, those poisoning events are not characteristic of most environmental exposures.
To document the role of pollutants in diabetes burden, authors have mainly reported occupational case-control cohorts that in fact exhibit many limitations: the exposure occurs many years before the analysis is undertaken and the levels of EDCs are usually estimated by questionnaires or back calculation from serum or urine concentrations measured many years later. For example, in the D.E.S.I.R. cohort, incident T2D was not associated with POPs levels measured in plasma nine years before the T2D diagnosis [13]. In addition, most of these occupational studies focused on exposure to one EDC at a time, while the cohorts are usually exposed to a whole mixture of toxicants and studies usually focus on individuals who live in heavily polluted areas [14]. Levels of PCBs were associated with higher fasting plasma glucose and an increased risk of T2D in a Chinese population (OR = 1.558 when fourth quartile [Q4] was compared to first quartile [Q1] for PCB52 [95% CI 1.109–2.189; p = 0.025]; OR = 1.841 Q4 vs. Q1 for PCB153 [95% CI 1.275–2.656; p = 0.001]) [15]. In a cohort of obese adolescents and young adults (SOLAR and CHS cohorts), PCB118 and PBDE153 were associated with higher glucose concentrations in oral glucose tolerance tests [16]. Another critical point to consider is the exposure dose associated with the occurrence of T2D. Generally, people are exposed to low to very low doses of EDCs that are usually at least hundred times lower than no or low adverse effect levels according to regulatory guidelines [17]. However, these low to very low doses were related to incident T2D, especially in the case of exposure to PFAS, while higher doses were not related to adverse metabolic effects [18,19], suggesting a non-monotonic dose-response relationship following parabolic-shaped or inverted-U-shaped curves, as previously reported for bisphenol A [20].
Thus, all these results should be interpreted with caution and may not reflect the exact risk incurred by the general population. For this reason, the usual figures adapted from Neel et al. [21] and Baillie-Hamilton P. [22] illustrating the spectacular concordance between the respective evolution of diabetes and obesity prevalence and the national production of synthetic organic compounds in the United States should be carefully considered.
Finally, the association between POPs exposure and diabetes may be due to confounding factors such as fat mass, as most of them are stored in adipose tissue and/or as POPs are also considered as obesogens [23]. Thus, POPs exposure may have a synergistic effect with increased adipose tissue on the risk of T2D [24]. However, some studies reported that POPs were related to T2D only in lean patients but not in overweight or obese patients, suggesting a specific role of POPs in T2D pathophysiology [25], probably through modifications of insulin synthesis and/or insulin release [26].

2.2. Non-Persistent EDCs and Type 2 Diabetes

Since the beginning of the 2000s, several longitudinal studies have investigated the potential role of persistent and non-persistent EDCs, with a special concern for phthalates, especially bisphenol A (BPA), because more than 95% of all people worldwide have BPA in their bodies [27]. Indeed, BPA is probably one of the most extensively studied and well-known EDCs. First synthesized in 1891, BPA exhibits estrogenic properties that have aroused concern for human health, especially regarding developmental and reproductive effects [28], and cancers of hormone-dependent organs [29].
Among the population studies, the US National Health and Nutrition Examination Survey (NHANES), which is an American food consumption database program operating since 1960, is probably the most documented survey, monitoring 5000 to 10,000 representative Americans annually. Since 1999, it also includes the testing of an extensive number of chemicals and systematically reports detectable blood and/or urine levels of several chemicals in most participants. Moreover, diagnosed or self-reported diabetes was strongly associated with the exposure to POPs (PCBs, dioxins, p,p′-DDE) but also with non-persistent EDCs such as phthalates and BPA (OR = 2.74 [95%CI 1.44–5.23] after adjustment for age, gender, body mass index, and ethnicity) [30,31]. Other cross-sectional studies reported a suggestive to strong association between diabetes and pre-diabetes and EDCs exposure, especially for BPA exposure [32,33,34]. For example, Sun Q. et al. showed a positive correlation between urinary BPA and butyl-phthalate concentrations (assessed from blood and urine samples collected 5 to 10 years before) and the incident T2D in the participants of the Nurses’ Health Study II (NHSII), but not in the older NHS counterparts [35]. In a meta-analysis of all clinical studies on BPA published before mid-2014, urinary concentrations of BPA were associated with T2D risk (OR = 1.47 between highest and lowest urinary levels) [36]. This risk was confirmed by BPA CLARITY, a project begun by the NIEHS (National Institute of Environmental Health Sciences)/NTP (National Toxicology Project) and the FDA, which confirmed significant metabolic adverse effects at the lowest dose examined (2.5 μg/kg body weight per day), far lower than the maximum safe daily oral BPA dose over the lifetime (50 μg/kg body weight per day) established by the EPA and FDA (CLARITY-BPA Program (https://ntp.niehs.nih.gov/whatwestudy/topics/bpa/index.html?utm_source=direct&utm_medium=prod&utm_campaign=ntpgolinks&utm_term=clarity_bpa (accessed on 10 January 2023))).
The health concerns of BPA led to the progressive use of substitutes since about ten years ago. Urinary and blood levels of these substitutes (notably bisphenol S, F, M, B AP, AF, BADGE) are gradually rising in the global population. However, limited data are available concerning their innocuity for human health. One of the most investigated substitutes is bisphenol S (BPS) but only a few studies are available in the literature regarding the association of exposure to BPS with incident diabetes, fasting blood glucose, or insulin resistance [36,37,38,39,40]. Among them, two reported a significant association between exposure to BPS and incident or overt diabetes with variable odd ratios: OR 3.83 [95% CI 2.37–6.20; p < 0.001] [37] and adjusted hazard ratio (aHR) at year 3:2.81 [95% CI 1.74–4.53] [36]. As for BPA, this increased risk is not dependent on overweight or obesity, as urinary BPS concentrations were positively associated with an increase in insulin resistance evaluated by ln-transformed homeostatic model assessment for insulin resistance (ln-HOMA-IR) (p = 0.017) only in women not overweight or obese (BMI < 25 kg/m2) [41].
Phthalates are plasticizers with well-known EDC properties, especially in obesity determining. Recently, the evidence for a role in the determining of diabetes has been mixed. Indeed, some reported a positive correlation between phthalates exposure and insulin resistance determined by the HOMA index [42]. Moreover, in a meta-analysis of seven studies including more than 12,000 people, exposure to low and high molecular weight phthalate metabolites, but also to di-2-ethylhexyl phthalate (DEHP), was significantly associated with an increased risk of incident or overt type 2 diabetes, with an overall OR of 1.69 [95% CI 1.29–2.20], 1.71 [95% CI 1.43–2.05] and 2.15 [95% CI 1.48–3.13], respectively [43].
Other EDCs have been suggested in the diabetes burden. Triclosan and triclocarban are two biocides commonly used despite an FDA ban in 2016, especially in toothpastes. As for phthalates, evidence in human studies has been mixed. In a recent analysis of NHANES (2013–2014), a significant positive association was reported between exposure to triclocarban and incident or overt diabetes, but only in women (OR = 1.79 [95% CI 1.05–2.05] in women vs. 1.29 [95% CI 0.73–2.27] in men; p = 0.032), even after adjusting for potential confounding factors [44]. Heavy metals (cadmium, lead, and mercury), trace elements (such as zinc), and arsenic have also been suggested as risk factors for metabolic syndromes or overt diabetes. However, results remain conflicting [45,46,47,48,49]. More recently, some authors reported a possible role of the air pollution in the diabetes burden, including ozone [50], nitrogen dioxide [51], and particulate matter (PM), especially PM2.5, which were related to an increased incidence and prevalence of diabetes [52] and worsened glycemic control among people previously diagnosed with diabetes [53]. It is important to note that the impact of air pollution is enhanced by warm and humid weather, which raises important concerns about the possible impact of climate change on diabetes risk [54].
However, several limitations with these epidemiological studies should be highlighted. As in any observational study, the association between EDCs exposure and diabetes does not establish causality. Furthermore, many of the above studies were designed to correlate diabetes prevalence with current EDCs levels, while their current levels may strongly differ from concentrations during the disease development. Moreover, EDCs are usually highly correlated with each other, and it is not always possible to precisely determine the independent effect of each chemical compound when people are daily exposed to a whole mixture. Another point to consider is the potential individual differences in EDCs metabolism. Indeed, genetic polymorphisms could affect both bioavailability and adverse effects of EDCs, notably during critical period of exposure, such as in pregnancy and early childhood, as it was notably reported for BPA [55]. Finally, the association between EDCs exposure and diabetes may be due to confounding factors, as suggested above [13,56,57].

2.3. EDCs and Type 1 Diabetes

The link between environmental toxicant exposures and T1D has been relatively understudied compared to T2D. However, the prevalence of T1D is also increasing to a lesser extent but with notable geographic variations, especially in industrializing countries such as China, for example. Several cross-sectional studies reported conflicting results for EDCs: some studies were characterized by higher concentrations of EDCs at diagnosis of T1D in children [58], while other suggested a protective effect of some EDCs (especially perfluoroalkyls) on the development of T1D in children [59]. As T1D arises from autoimmune destruction of pancreatic beta cells, it has been suggested that EDCs could be involved by promoting beta cells apoptosis or necrosis and/or by disrupting the immune system (notably for BPA and its substitutes) [58]. Other epidemiological studies focused on the role of air pollution on T1D incidence and reported that exposure to air pollution (notably to ozone) during pregnancy is associated with an increased risk for early T1D in children (HR per interquartile increase = 2.00 [95% CI 1.04–3.86]) [60,61]. Those data were recently confirmed in an environment-wide association study conducted in the UK on about 14,000 children with T1D, which reported a spatial heterogeneity in disease mapping depending on 15 main risk factors including air pollutants (PM2.5, nitrogen dioxide), lead, radon, outdoor light at night, overcrowding, population density, and ethnicity [62]. Overall, more work is needed to better understand the contribution of EDCs and environment to T1D risk.

2.4. Maternal Exposure to EDCs and Diabetes

Another critical issue to consider is exposure to EDCs during pregnancy, which opens two lines of research, one in relation to the development of diabetes during pregnancy and the other one in relation to the development of diabetes in the offspring [63]. Indeed, the fetal period is a critical window, which can definitively influence the developmental origins of health and disease (DOHaD) through modifications in gene expression and unexpected effects on metabolism of progeny as proposed by David Barker [64]. Therefore, either gestational diabetes potentially induced by EDCs or the EDC exposure during this critical window may have an impact on progeny predisposition to develop diabetes. The data from human studies investigating the potential impact of prenatal exposure to EDCs on mother metabolic dysfunction are equivocal. Some studies have shown that exposure to phthalates and/or BPA was associated with increase plasma glucose and impaired glucose tolerance in pregnant women [65]. The same results were recently reported for BPS, which impaired blood glucose tolerance during pregnancy but was not associated with incident gestational diabetes [40]. Yan et al. have recently published a meta-analysis of 25 studies including 16 cohort studies, 7 case-control studies and 2 cross-sectional studies to conclude that PCBs, PBDE, PFAs and phthalates are critical gestational diabetes risk factors [66]. Other epidemiological studies have found associations between heavy metals [67,68], PFASs, POPs [69,70], or some bisphenol substitutes (notably bisphenol AF) [40] and an increased risk of gestational diabetes. Overall, there is some evidence for the role of EDCs in gestational diabetes, but further studies are needed to better characterize this risk.
Importantly, several studies have shown that mothers can directly expose their progeny to EDCs, for instance BPA, through the placenta but also by breast feeding [71,72]. Others have suggested associations between prenatal exposure to DDE and PFAs and predisposition to develop diabetes into progeny [73]. Moreover, prenatal exposure to pollutants is associated with an increased incidence of overweight and obesity in childhood and adolescence, with early catch-up growth as previously described by David Barker [64] in the DOHaD hypothesis. For example, in the CHAMACOS (Center for the Health Assessment of Mothers and Children of Salinas) study, prenatal exposure to mixtures of phthalates and phenols [74,75] or to DDT and its metabolites (i.e., DDE) [76] was associated with increased risks of overweight and obesity at the age of 5 and 12 years for both boys and girls. In this cohort, the risk of T2D in progeny has not yet been assessed but we can assume that such early metabolic disorders may lead to an increased prevalence of T2D.

3. In Vivo Evidence of Diabetogenic Pollutants

Epidemiological evidence is necessary to evaluate the potential impact of EDCs on human health, but although usually correlative, studies have not been sufficient to establish that a molecule is an EDC. In vivo and in vitro experiments are required to identify the doses, the kinetics, and the molecular mechanisms of the interferences with the endocrine systems even though they do not exactly replicate human physiology and variability/heterogeneity [77,78,79,80]. Using phenotypic and functional analyses in addition to complementary omics approaches, as carried out on humans [81], animal experiments specifically can study pathophysiological features from periconception to adult life up to progeny. Of note, some studies have been performed and well-reviewed for EDCs as obesogenic pollutants, showing their impact on adipose tissue hyperplasia and/or hypertrophy [23,78,82,83]. However, obesity is not the only cause of T2D and is not necessarily linked to diabetes. EDCs have also been implicated as diabetogenic pollutants as previously reviewed [56,82,84,85,86,87,88]. To show diabetogenic effects of EDCs in vivo experiments usually measure insulin secretion and glucose metabolism dysregulations due to insulin resistance. Of note, some studies have also started to explore the effects of EDCs on gut microbial ecology and physiology, and their subsequent impact on glucose metabolism as gut microbiota modifications have been associated with diabetes [89]. In vivo evidence is summarized in Figure 1.

3.1. Persistent EDCs and T2D

Global studies have been conducted through consumption of fat-containing food, considering that we are exposed to pollutants this way. By feeding rats [90] or mice [91] during a month with a high-fat diet along with natural source of a cocktail of persistent EDCs present in salmon oil, Ruzzin et al. were the first to show that these rats became insulin-resistant. This chronic exposure to low doses of POPs exacerbates obesity and hepatic steatosis with a dysregulation of key hepatic genes. When a similar type of diet, this time supplemented with a cocktail of commonly found EDCs (TCDD, PCB153, DEHP and BPA) instead of salmon oil, was maintained from periconception period throughout life in mice, this led to sex-specific metabolic alterations in the progeny [92]. A chronic exposure to a PCB mixture (Aroclor 1254) for 30 weeks led to hyperinsulinemia independent of body weight in both standard and high-fat diet-fed mice, while an aggravation of insulin resistance was observed in high-fat diet-fed mice [93]. Another study showed that a chronic exposure of this mixture every 3 days for 60 days increased body weight and insulinemia associated with insulin resistance development [94]. Even though a mixture is more representative of our everyday exposure, we are not exposed to the same cocktails and not in the same proportions. Therefore, animals were also exposed to EDCs one by one rather than in cocktails to identify the effects of each EDC to avoid potential synergic or antagonist activities of one EDC on the others. Regarding PCBs, a 2-week exposure either with PCB126 or PCB77, two coplanar PCBs, was sufficient to provoke an impairment of glucose tolerance associated with insulin resistance in standard diet-fed mice [95]. Similar data were obtained with chronic exposure of PCB-118 or PCB-138 in standard diet-fed mice [96], while for PCB153, a deterioration of glucose and insulin intolerance in high-fat diet-fed mice was shown [97]. Interestingly, PCB126 had a strong effect in inducing hepatic steatosis in mice compared to a PCB mixture (Aroclor 1260); conversely, the mixture had the strongest deleterious effect on pancreatic beta cell function. However, in both cases, insulin resistance was not measured [98]. Concerning TCCD, an acute exposure has been shown to impair glucose-stimulated insulin secretion without hyperglycemia in WT mice but not in AhR (Aryl hydrocarbon Receptor)-deficient mice, suggesting that this effect is mediated though this nuclear receptor [99]. A single exposure to TCDD can also abolish basal insulin levels up to six weeks post-injection in both sexes, but adverse effects on glucose homeostasis are sex-dependent, as they were observed only in males [100]. Moreover, chronic exposure to TCDD may induce insulin resistance in rats [101].
Adult exposure to other persistent EDCs such as DDT, and its metabolite DDE, or TBT were shown to have obesogenic effects, and subsequently potential diabetes risk factors rather than to have direct diabetogenic effects [23,82]. All these studies show that EDCs impact on glucose homeostasis and insulin secretion, but the phenotypes have a variable intensity dependent on the doses but also the exposure reflecting the complexity of the problem [102,103].

3.2. Non-Persistent EDCs and T2D

Studies of BPA exposure and the development of T2D in rodent data can appear contradictory because of different methods, the windows, the exposure, and the doses, but they provide convincing evidence of adverse effects of BPA contributing to the development of diabetes. Several reviews have recently outlined key publications studying mechanisms linking BPA and T2D development [55,71,82,104,105,106]. In summary, it is now clear that BPA exposure leads to perturbated insulin secretion with β cell dysfunction, liver and muscle insulin resistance but also increased adipogenesis. Recent studies have investigated the metabolic outcomes of chronic and prenatal exposure to BPA substitutes in rodents. Despite some conflicting results (notably between males and females, which seem less susceptible), BPA substitutes, in particular bisphenol S (BPS), exhibit the same metabolic adverse outcomes than BPA, involving similar signaling pathways, including especially PPARγ and extranuclear-initiated ER pathways [107,108,109].
Contrary to BPA, few studies have shown in vivo that phthalates/DEHP are linked to diabetes interfering with insulin secretion [110,111]. Phthalates have rather been linked to insulin resistance and obesity, notably increasing oxidative stress. Interestingly, Wei et al. have shown that chronic exposure of mice to DEHP deregulates oxidative stress in muscle though the miR-17/Keap1-Nrf2/miR-200a axis leading to insulin resistance [112]. Recently, Baralic et al. highlighted a potential additive effect for a mixture of DEHP, DBP and BPA regulating redox status parameters such as TOS, SOD, and SH groups in rat pancreatic tissue [113].
Importantly, as reviewed by Castriota et al. [114], recent studies in rodents have shown that arsenic exposure in adult life induced glucose intolerance and decreased glucose-stimulated insulin secretion in mice [115], and led to impaired glucose sensing and subsequently insulin release in rats [116]. In addition, chronic exposure to arsenic in drinking water have been shown to impact body weight and composition, fasting glycemia, and insulinemia [117]. These data have shown an increased HOMA index in several different outbred mice [117].

3.3. EDCs and T1D

Concerning the potential causal relation between EDC and T1D, some studies have been carried out mainly in non-obese diabetic (NOD) mice [118]. So far, no consensus has been obtained as too few studies have been performed, and because both immunosuppressive and immunoproliferative effects have been reported. Regarding PCBs, Kuiper et al. have shown that, either at high or low doses in short or chronic exposures, the development of T1D was decreased in NOD mice, associated with a decrease in T cell populations [119]. These data were obtained in the context of conflicting epidemiological studies [120,121,122], and there have been no further studies to corroborate. Likewise, for PFAs only one published work in NOD mice showed accelerated insulitis [123], and for inconsistent epidemiological studies regarding this, see [118]. While NOD mice exposed to DDE displayed increased incidence of T1D [124], contradictory results were previously obtained on the immune response in mice after DDT exposure [125]. The situation is not clearer for dioxins as TCDD. Indeed, the exposure of WT mice leads to impaired-second-phase of glucose-stimulated secretion [99], whereas the exposure of NOD mice reduced insulitis through the increase in Foxp3 T cells [126]. In both cases, the effect of TCDD has been shown to be mediated through AhR. Concerning non-persistent EDCs, the same observation can be made since there are so far not enough studies. Exposure to BPA, but not to phthalates or to mixed BPA and phthalates, accelerated the development of T1D in NOD mice [127], yet the developmental exposure to phthalates led to pancreatic beta cell dysfunction with impaired glucose tolerance in rats [58,110]. Importantly, for BPA, but also BPS, effects on T1D development depend on the exposure window, the diet, and the sex [128,129]. However, regarding BPA, data remain controversial. Few data were also obtained in NOD and STZ mice after exposure to arsenic and air pollution [118], reinforcing the conclusion that further in vivo experiments are required to obtain concrete evidence for the adverse impact of EDCs on autoimmune responses and the development of T1D. EDCs have mainly been investigated for immunomodulation effects and some studies have measured their impact on gut microbiota or vitamin D levels as other mechanisms involved in the development of T1D [89,118]. However, further experiments notably in vitro are required to clarify their action and to identify their precise action modes at the molecular level.

3.4. Maternal-Fetal Exposure to EDCs and Diabetes

While epidemiological studies focus on the potential impact of EDCs on the development of gestational diabetes, animal studies with EDC exposure during pregnancy usually investigate the effect on the development of diabetes in the offspring. Not many studies have been performed to study the effects of EDC developmental exposure. Interestingly, the rat dam exposure from day 8 to 15 with DDE led to transgenerational glucose intolerance and pancreatic impairment in F1 and F2 associated with modified genomic imprinting on igf2/H19 gene expression transmitted to F3 progeny through the male germ line [130,131]. Regarding BPA however, data remain contradictory [65,105,132]. Several studies have shown that developmental exposure to BPA leads to transgenerational abnormalities, but the degree of the phenotypes associated in progeny varies in terms of T1D or T2D development [105,129,133,134]. These conclusions are similar for other EDCs, with even counteracting effects when used as mixture.

4. In Vitro Evidence of Diabetogenic Pollutants

To complete animal experimental data, in vitro approaches are required to better decipher and identify the action modes of EDCs, but also to bring robustness in data with the possibility to work with complementary cellular models and in various species. Different approaches have been developed to set up in vitro screenings for EDCs, for instance with 96-well cell plates with isolated mice islets using ROS production measurement as a read out for damage in islet cells [135] or with human embryonic stem cell culture to study developmental origins of diabetes [136]. Other in vitro assays have been developed to study the impact of these EDCs on beta cell function [103,137]. However, the question is more than a screen and requires identifying how or at which level of the hormonal pathway EDCs act. Multiple mechanisms have been implicated for EDC interference action modes: through nuclear receptor hormone pathways (ER/AhR/PPAR) resulting in genomic effects, but also through insulin-signaling pathways (PI3K/ERK/AKT) implicating both genomic and non-genomic effects [82,87,88]. In both cases, they can induce or exacerbate an oxidative stress, a dysregulation of KATP channel or interference with other pathways involved in insulin exocytosis in pancreatic islets or adipose tissue inflammation and/or hepatic metabolism dysregulation [56,82,86,87]. Classically, among the main studies EDCs, PCB, PFOA and TCDD have been shown to interfere with PPARs, TCDD is also able to bind ER and AhR, and DDT with RXR /CAR. In vitro pathways involved in T2D are summarized in Figure 2.

4.1. Persistent EDCs and T2D

Concerning insulin secretion, an acute exposure to a PCB mixture (Aroclor 1254) was initially shown to stimulate glucose-induced insulin secretion of RINm5F cells potentially through calcium and CaM kinase [138,139]. Lee et al. showed that high concentrations of this same mixture decreased insulin release in Ins1E cells, while low concentrations had a stimulatory effect [140]. Ex-vivo analyses of tissues from mice chronically exposed to Aroclor 1254 showed an increased pancreatic beta cell mass associated with a decreased expression of key actors in the insulin-signaling pathway (IR/IRS/PI3K/AKT/GLUT4) in liver and muscle [94]. Interestingly, Kim et al. showed that in 3T3-L1 adipocytes and mice adipose tissue, the lipid droplet-associated protein, fat-specific protein 27 (Fsp27), mediated PCB-induced insulin resistance for both PCB118 (a dioxin-like PCB) and PCB138 (a non-dioxin-like PCB) through the downregulation of IRS1 associated with a phosphorylation decrease in PI3K and AKT [96].
Besides PCB, acute exposure of skeletal muscle (L6 cells) and pancreatic beta cells (Rin-m5F) to low doses of DDT (1 to 5 µM) impairs insulin-stimulated glucose uptake and insulin secretion, respectively, through increased production of intracellular reactive oxygen species (ROS) [141]. These subtoxic doses of DDT causes increased oxidative stress-induced activation of redox-sensitive kinases responsible for the impairment of insulin signaling with the decreased phosphorylation of IRS1/AKT [142]. Moreover, a proteomic approach revealed several markers of toxicity for acute but high doses of DDT (150 µM) in pancreatic beta cells, including oxidative stress proteins (GRP78, and endoplasmin), mitochondrial proteins (GRP75, ECHM, IDH3A, NDUS1, and NDUS3), proteins involved in the maintenance of the cell morphology (EFHD2, TCPA, NDRG1, and ezrin), and other proteins (vimentin, PBDC1, EF2, PCNA, and HSP27) [143]. Recently, it was reported that p,p′-DDE (10 µM) could also play a role in beta cell dysfunction by altering insulin translation, probably due to an increase in prohormone convertase levels [103,144] whereas low doses of TBT (0.1 µM) led to an increase in calcium-dependent insulin secretion through PKC and ERC phosphorylation [102,103].
Concerning dioxin, TCDD has been identified as an estrogen mimetic, which is a key regulator of glucose homeostasis by promoting energy homeostasis and improving insulin resistance, and beta cell function and survival [56,145,146]. Moreover, an acute exposure of pancreatic beta cells (Ins-1E) to nanomolar doses of TCDD has been shown to induce calcium influx involved in lysosomal and secretory granule exocytosis [147]. Kim et al. postulated that TCDD might exert adverse effects on beta cells inducing beta cell failure due to permanent insulin release. Inversely, morphological and functional alterations of pancreatic-isolated islets have been observed after acute exposure to nanomolar TCDD specifically in beta cells associated with decreased glucose-induced insulin secretion and dysregulation of key beta cell gene expression [148]. Besides diabetogen, TCDD has been identified as an obesogen interfering with insulin signaling (GLUT-4 [149]; TNF-a [150,151,152]) and adipocyte differentiation [80,153].
A perfluoroalkyls effect appears controversial in islets, for instance, for acute exposure in beta cells, while Qin et al. showed that PFOS stimulated calcium-induced insulin secretion through the activation of G protein-coupled receptor 40 [154]), He et al. showed that PFOA decreased insulin secretion through ER stress and the ATF4/CHOP/TRIB3 pathway [155] in hepatocytes [156]. Concerning effect in the liver, data are more consistent for involvement in steatosis [157,158,159].

4.2. Non-Persistent EDCs and T2D

Several reviews have largely discussed the link between BPA and diabetes covering the key studies bringing mechanistic and functional evidence reinforcing epidemiologic arguments [55,71,86,105]. BPA can act through several physiological receptors involved in glucose metabolism and insulin secretion, including nuclear receptors such as ER and PPAR but also membrane receptors such as GPR30 (membrane-bound estrogen receptors) or estrogen-related receptor (ERR) gamma. Recently, Park et al. showed that mixtures of BPA and its analog BPS and BPF have estrogen agonist and anti-androgen activities at lower doses than each one alone, highlighting a confounder variable in epidemiological approaches [160,161].
Concerning phthalates, in addition to activating oxidative stress in muscles [112], an acute exposure of pancreatic beta cells (Rin-m5F) to micromolar doses of phthalate induced a ROS-mediated PI3K/AKT/Bcl-2 pathway responsible for pancreatic beta cell apoptosis [162].
Regarding arsenic, a recent review robustly highlighted the potential molecular mechanisms mediating arsenic’s effects on glucose metabolism inhibiting SIRT3, AKT, and GLUT4 translocation [114]. Arsenic may interfere with calcium-mediated signaling necessary for insulin secretory granule exocytosis. Castriota et al. identified 16 genes commonly affected by arsenic, insulin resistance, and T2D (including insulin, IRS, adiponectin, and leptin) [114]. Arsenic has also been shown to modulate NLRP3 inflammasome activation involved in hepatic insulin resistance [163], or to be a master regulator of microRNA involved in pancreatic beta cell impairment [164]). Interestingly from a biophysical side, mercury and cadmium have the ability to modulate in a different manner the aggregation of human islet amyloid polypeptide (hIAPP) [165]. Cadmium promotes insoluble amyloid aggregation potentially leading to beta cell death, while mercury increases the formation of ion channel as an oligomer possibly involved in beta cell dysfunction [165].
Finally, many EDCs also exist in air as volatile or semi-volatile compounds in the gas phase or attached to PM. Hill et al. have recently demonstrated that chronic incubation of aortas from naive mice with plasma isolated from mice exposed to four times the recommended values of PM2.5 leads to a dyslipidemic phenotype, which could contribute to vascular inflammation and loss of insulin sensitivity [166].

5. Conclusions

Despite all these in vivo and in vitro studies, it remains difficult to form a comprehensive view on the causal relationship between EDCs and diabetes (resumed in Table 1). It is difficult to test, even for one molecule, different concentrations with various kinetics on different signaling targets. Even focusing on one pathway, we need thereby to multiply in vivo and in vitro approaches to obtain a complete analysis and we need to consider critical windows of exposition. Therefore, to definitely prove the role of EDCs in diabetes pathophysiology, further experiments are required. This would follow an experimental consensus with both acute and chronic exposures reflecting our daily exposure in accordance with observations from epidemiological data.

Author Contributions

C.H. and N.C. designed the narrative review, researched data and contributed to the discussions and manuscript. P.C.-B. designed data and contributed to discussions. F.B. contributed to the discussions. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by ITMO (Plan Cancer 2014–2019), ANSES (EST-17-086 Contract), Alpes-Maritimes General Council (Health Projects 2017; 2017-253 DGA-DSH) and the French government, through the UCAJEDI Investments in the Future project managed by the National Research Agency (ANR) with the reference number ANR-15-IDEX-01 (ProstAdiPE Project, Academy #3).

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.

References

  1. World Health Organization. Diabetes (Fact. Sheet N°312). Available online: http://www.who.int/mediacentre/factsheets/fs312/en/ (accessed on 15 September 2022).
  2. International Diabetes Federation. IDF Diabetes Atlas, 10th ed.; International Diabetes Federation: Brussels, Belgium, 2021; Available online: http://www.idf.org/diabetesatlas (accessed on 15 January 2023).
  3. American Diabetes Association. Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care 2018, 41, 917–928. [Google Scholar] [CrossRef] [Green Version]
  4. Thayer, K.A.; Heindel, J.J.; Bucher, J.R.; Gallo, M.A. Role of environmental chemicals in diabetes and obesity: A National Toxicology Program workshop review. Environ. Health Perspect. 2012, 120, 779–789. [Google Scholar] [CrossRef] [Green Version]
  5. UNEP. Stockholm Convention On Persistent Organic Pollutants (POPs). Texts and Annexes. Available online: http://chm.pops.int/TheConvention/Overview/TextoftheConvention/tabid/2232/Default.aspx (accessed on 15 September 2022).
  6. Vasseghian, Y.; Hosseinzadeh, S.; Khataee, A.; Dragoi, E.N. The concentration of persistent organic pollutants in water resources: A global systematic review, meta-analysis and probabilistic risk assessment. Sci. Total Environ. 2021, 796, 149000. [Google Scholar] [CrossRef] [PubMed]
  7. Guillotin, S.; Delcourt, N. Studying the Impact of Persistent Organic Pollutants Exposure on Human Health by Proteomic Analysis: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 14271. [Google Scholar] [CrossRef] [PubMed]
  8. Bertazzi, P.A.; Consonni, D.; Bachetti, S.; Rubagotti, M.; Baccarelli, A.; Zocchetti, C.; Pesatori, A.C. Health effects of dioxin exposure: A 20-year mortality study. Am. J. Epidemiol. 2001, 153, 1031–1044. [Google Scholar] [CrossRef]
  9. Warner, M.; Mocarelli, P.; Brambilla, P.; Wesselink, A.; Samuels, S.; Signorini, S.; Eskenazi, B. Diabetes, metabolic syndrome, and obesity in relation to serum dioxin concentrations: The Seveso women’s health study. Environ. Health Perspect. 2013, 121, 906–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Michalek, J.E.; Pavuk, M. Diabetes and cancer in veterans of Operation Ranch Hand after adjustment for calendar period, days of spraying, and time spent in Southeast Asia. J. Occup. Environ. Med. Am. Coll. Occup. Environ. Med. 2008, 50, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Wang, S.L.; Tsai, P.C.; Yang, C.Y.; Guo, Y.L. Increased risk of diabetes and polychlorinated biphenyls and dioxins: A 24-year follow-up study of the Yucheng cohort. Diabetes Care 2008, 31, 1574–1579. [Google Scholar] [CrossRef] [Green Version]
  12. Chang, J.W.; Chen, H.L.; Su, H.J.; Lee, C.C. Abdominal Obesity and Insulin Resistance in People Exposed to Moderate-to-High Levels of Dioxin. PLoS ONE 2016, 11, e0145818. [Google Scholar] [CrossRef] [Green Version]
  13. Magliano, D.J.; Ranciere, F.; Slama, R.; Roussel, R.; Kiviranta, H.; Coumoul, X.; Balkau, B.; Botton, J.; the D.E.S.I.R. Study Group. Exposure to persistent organic pollutants and the risk of type 2 diabetes: A case-cohort study. Diabetes Metab. 2021, 47, 101234. [Google Scholar] [CrossRef]
  14. Magliano, D.J.; Loh, V.H.; Harding, J.L.; Botton, J.; Shaw, J.E. Persistent organic pollutants and diabetes: A review of the epidemiological evidence. Diabetes Metab. 2014, 40, 1–14. [Google Scholar] [CrossRef] [PubMed]
  15. Tan, Q.; Wang, M.; Yu, L.; Liang, R.; Liu, W.; Dong, C.; Zhang, Y.; Li, M.; Ye, Z.; Wang, B.; et al. Associations of polychlorinated biphenyls exposure with plasma glucose and diabetes in general Chinese population: The mediating effect of lipid peroxidation. Environ. Pollut. 2022, 308, 119660. [Google Scholar] [CrossRef]
  16. Baumert, B.O.; Goodrich, J.A.; Hu, X.; Walker, D.I.; Alderete, T.L.; Chen, Z.; Valvi, D.; Rock, S.; Berhane, K.; Gilliland, F.D.; et al. Plasma concentrations of lipophilic persistent organic pollutants and glucose homeostasis in youth populations. Environ. Res. 2022, 212, 113296. [Google Scholar] [CrossRef] [PubMed]
  17. Zoeller, R.T.; Brown, T.R.; Doan, L.L.; Gore, A.C.; Skakkebaek, N.E.; Soto, A.M.; Woodruff, T.J.; Vom Saal, F.S. Endocrine-disrupting chemicals and public health protection: A statement of principles from the Endocrine Society. Endocrinology 2012, 153, 4097–4110. [Google Scholar] [CrossRef]
  18. Gui, S.Y.; Qiao, J.C.; Xu, K.X.; Li, Z.L.; Chen, Y.N.; Wu, K.J.; Jiang, Z.X.; Hu, C.Y. Association between per- and polyfluoroalkyl substances exposure and risk of diabetes: A systematic review and meta-analysis. J. Expo. Sci. Environ. Epidemiol. 2022, 33, 40–55. [Google Scholar] [CrossRef]
  19. Duan, Y.; Sun, H.; Yao, Y.; Li, Y.; Meng, Y.; Lu, Y.; Han, L.; Chen, L. Serum concentrations of per-/polyfluoroalkyl substances and risk of type 2 diabetes: A case-control study. Sci. Total Environ. 2021, 787, 147476. [Google Scholar] [CrossRef]
  20. Vandenberg, L.N. Non-monotonic dose responses in studies of endocrine disrupting chemicals: Bisphenol a as a case study. Dose Response 2014, 12, 259–276. [Google Scholar] [CrossRef]
  21. Neel, B.A.; Sargis, R.M. The paradox of progress: Environmental disruption of metabolism and the diabetes epidemic. Diabetes 2011, 60, 1838–1848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Baillie-Hamilton, P.F. Chemical toxins: A hypothesis to explain the global obesity epidemic. J. Altern. Complement. Med. 2002, 8, 185–192. [Google Scholar] [CrossRef]
  23. Heindel, J.J.; Blumberg, B. Environmental Obesogens: Mechanisms and Controversies. Annu. Rev. Pharm. Toxicol. 2019, 59, 89–106. [Google Scholar] [CrossRef]
  24. Airaksinen, R.; Rantakokko, P.; Eriksson, J.G.; Blomstedt, P.; Kajantie, E.; Kiviranta, H. Association between type 2 diabetes and exposure to persistent organic pollutants. Diabetes Care 2011, 34, 1972–1979. [Google Scholar] [CrossRef] [Green Version]
  25. Ngwa, E.N.; Kengne, A.P.; Tiedeu-Atogho, B.; Mofo-Mato, E.P.; Sobngwi, E. Persistent organic pollutants as risk factors for type 2 diabetes. Diabetol. Metab. Syndr. 2015, 7, 41. [Google Scholar] [CrossRef] [Green Version]
  26. Jorgensen, M.E.; Borch-Johnsen, K.; Bjerregaard, P. A cross-sectional study of the association between persistent organic pollutants and glucose intolerance among Greenland Inuit. Diabetologia 2008, 51, 1416–1422. [Google Scholar] [CrossRef] [Green Version]
  27. vom Saal, F.S.; Hughes, C. An extensive new literature concerning low-dose effects of bisphenol A shows the need for a new risk assessment. Environ. Health Perspect. 2005, 113, 926–933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Peretz, J.; Vrooman, L.; Ricke, W.A.; Hunt, P.A.; Ehrlich, S.; Hauser, R.; Padmanabhan, V.; Taylor, H.S.; Swan, S.H.; VandeVoort, C.A.; et al. Bisphenol a and reproductive health: Update of experimental and human evidence, 2007–2013. Environ. Health Perspect. 2014, 122, 775–786. [Google Scholar] [CrossRef]
  29. Shafei, A.; Ramzy, M.M.; Hegazy, A.I.; Husseny, A.K.; El-Hadary, U.G.; Taha, M.M.; Mosa, A.A. The molecular mechanisms of action of the endocrine disrupting chemical bisphenol A in the development of cancer. Gene 2018, 647, 235–243. [Google Scholar] [CrossRef]
  30. Lee, D.H.; Lee, I.K.; Song, K.; Steffes, M.; Toscano, W.; Baker, B.A.; Jacobs, D.R., Jr. A strong dose-response relation between serum concentrations of persistent organic pollutants and diabetes: Results from the National Health and Examination Survey 1999–2002. Diabetes Care 2006, 29, 1638–1644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Everett, C.J.; Frithsen, I.L.; Diaz, V.A.; Koopman, R.J.; Simpson, W.M., Jr.; Mainous, A.G., 3rd. Association of a polychlorinated dibenzo-p-dioxin, a polychlorinated biphenyl, and DDT with diabetes in the 1999–2002 National Health and Nutrition Examination Survey. Environ. Res. 2007, 103, 413–418. [Google Scholar] [CrossRef]
  32. Hong, Y.C.; Park, E.Y.; Park, M.S.; Ko, J.A.; Oh, S.Y.; Kim, H.; Lee, K.H.; Leem, J.H.; Ha, E.H. Community level exposure to chemicals and oxidative stress in adult population. Toxicol. Lett. 2009, 184, 139–144. [Google Scholar] [CrossRef] [PubMed]
  33. Ning, G.; Bi, Y.; Wang, T.; Xu, M.; Xu, Y.; Huang, Y.; Li, M.; Li, X.; Wang, W.; Chen, Y.; et al. Relationship of urinary bisphenol A concentration to risk for prevalent type 2 diabetes in Chinese adults: A cross-sectional analysis. Ann. Intern. Med. 2011, 155, 368–374. [Google Scholar] [CrossRef]
  34. Wang, T.; Li, M.; Chen, B.; Xu, M.; Xu, Y.; Huang, Y.; Lu, J.; Chen, Y.; Wang, W.; Li, X.; et al. Urinary bisphenol A (BPA) concentration associates with obesity and insulin resistance. J. Clin. Endocrinol. Metab. 2012, 97, E223–E227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Sun, Q.; Cornelis, M.C.; Townsend, M.K.; Tobias, D.K.; Eliassen, A.H.; Franke, A.A.; Hauser, R.; Hu, F.B. Association of urinary concentrations of bisphenol A and phthalate metabolites with risk of type 2 diabetes: A prospective investigation in the Nurses’ Health Study (NHS) and NHSII cohorts. Environ. Health Perspect. 2014, 122, 616–623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ranciere, F.; Botton, J.; Slama, R.; Lacroix, M.Z.; Debrauwer, L.; Charles, M.A.; Roussel, R.; Balkau, B.; Magliano, D.J.; the D.E.S.I.R. Study Group. Exposure to Bisphenol A and Bisphenol S and Incident Type 2 Diabetes: A Case-Cohort Study in the French Cohort D.E.S.I.R. Environ. Health Perspect. 2019, 127, 107013. [Google Scholar] [CrossRef] [Green Version]
  37. Duan, Y.; Yao, Y.; Wang, B.; Han, L.; Wang, L.; Sun, H.; Chen, L. Association of urinary concentrations of bisphenols with type 2 diabetes mellitus: A case-control study. Environ. Pollut. 2018, 243, 1719–1726. [Google Scholar] [CrossRef] [PubMed]
  38. Kataria, A.; Levine, D.; Wertenteil, S.; Vento, S.; Xue, J.; Rajendiran, K.; Kannan, K.; Thurman, J.M.; Morrison, D.; Brody, R.; et al. Exposure to bisphenols and phthalates and association with oxidant stress, insulin resistance, and endothelial dysfunction in children. Pediatr. Res. 2017, 81, 857–864. [Google Scholar] [CrossRef] [Green Version]
  39. Song, S.; Duan, Y.; Zhang, T.; Zhang, B.; Zhao, Z.; Bai, X.; Xie, L.; He, Y.; Ouyang, J.P.; Huang, X.; et al. Serum concentrations of bisphenol A and its alternatives in elderly population living around e-waste recycling facilities in China: Associations with fasting blood glucose. Ecotoxicol. Environ. Saf. 2019, 169, 822–828. [Google Scholar] [CrossRef]
  40. Zhang, W.; Xia, W.; Liu, W.; Li, X.; Hu, J.; Zhang, B.; Xu, S.; Zhou, Y.; Li, J.; Cai, Z.; et al. Exposure to Bisphenol a Substitutes and Gestational Diabetes Mellitus: A Prospective Cohort Study in China. Front. Endocrinol. 2019, 10, 262. [Google Scholar] [CrossRef] [Green Version]
  41. Lee, M.W.; Lee, M.; Oh, K.J. Adipose Tissue-Derived Signatures for Obesity and Type 2 Diabetes: Adipokines, Batokines and MicroRNAs. J. Clin. Med. 2019, 8, 854. [Google Scholar] [CrossRef] [Green Version]
  42. Shoshtari-Yeganeh, B.; Zarean, M.; Mansourian, M.; Riahi, R.; Poursafa, P.; Teiri, H.; Rafiei, N.; Dehdashti, B.; Kelishadi, R. Systematic review and meta-analysis on the association between phthalates exposure and insulin resistance. Environ. Sci. Pollut. Res. Int. 2019, 26, 9435–9442. [Google Scholar] [CrossRef]
  43. Zhang, H.; Ben, Y.; Han, Y.; Zhang, Y.; Li, Y.; Chen, X. Phthalate exposure and risk of diabetes mellitus: Implications from a systematic review and meta-analysis. Environ. Res. 2022, 204, 112109. [Google Scholar] [CrossRef]
  44. Xie, X.; Lu, C.; Wu, M.; Liang, J.; Ying, Y.; Liu, K.; Huang, X.; Zheng, S.; Du, X.; Liu, D.; et al. Association between triclocarban and triclosan exposures and the risks of type 2 diabetes mellitus and impaired glucose tolerance in the National Health and Nutrition Examination Survey (NHANES 2013–2014). Environ. Int. 2020, 136, 105445. [Google Scholar] [CrossRef]
  45. Planchart, A.; Green, A.; Hoyo, C.; Mattingly, C.J. Heavy Metal Exposure and Metabolic Syndrome: Evidence from Human and Model System Studies. Curr. Environ. Health Rep. 2018, 5, 110–124. [Google Scholar] [CrossRef]
  46. Dai, L.; Lv, X.; Chen, Z.; Huang, Z.; Li, B.; Xie, Y.; Duan, Y.; Zhao, H.; Wang, Y.; Yu, Q.; et al. Elevated whole blood arsenic level is associated with type 2 diabetes in coal-burning areas in Guizhou. Toxicol. Appl. Pharmacol. 2020, 403, 115135. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, Q.; Hou, Y.; Wang, D.; Xu, Y.; Wang, H.; Liu, J.; Xia, L.; Li, Y.; Tang, N.; Zheng, Q.; et al. Interactions of arsenic metabolism with arsenic exposure and individual factors on diabetes occurrence: Baseline findings from Arsenic and Non-Communicable disease cohort (AsNCD) in China. Environ. Pollut. 2020, 265, 114968. [Google Scholar] [CrossRef] [PubMed]
  48. Ye, Z.; Liang, R.; Wang, B.; Yu, L.; Liu, W.; Wang, X.; Xiao, L.; Ma, J.; Zhou, M.; Chen, W. Cross-sectional and longitudinal associations of urinary zinc with glucose-insulin homeostasis traits and type 2 diabetes: Exploring the potential roles of systemic inflammation and oxidative damage in Chinese urban adults. Environ. Pollut. 2022, 314, 120331. [Google Scholar] [CrossRef] [PubMed]
  49. Qu, Z.; Zhou, J.; Guo, P.; Wang, J.; Wang, P.; Liu, L.; Wu, M.; Wang, P.; Liu, N. Association between environmental lead/cadmium co-exposure in drinking water and soil and type 2 diabetes mellitus/obesity in Southern China. Front. Public Health 2022, 10, 941922. [Google Scholar] [CrossRef] [PubMed]
  50. Li, A.; Mei, Y.; Zhao, M.; Xu, J.; Seery, S.; Li, R.; Zhao, J.; Zhou, Q.; Ge, X.; Xu, Q. The effect of ambient ozone on glucose-homoeostasis: A prospective study of non-diabetic older adults in Beijing. Sci. Total Environ. 2021, 761, 143308. [Google Scholar] [CrossRef] [PubMed]
  51. Zhang, Q.; Liu, C.; Wang, Y.; Gong, J.; Wang, G.; Ge, W.; Chen, R.; Meng, X.; Zhao, Y.; Kan, H. Associations of long-term exposure to ambient nitrogen dioxide with indicators of diabetes and dyslipidemia in China: A nationwide analysis. Chemosphere 2021, 269, 128724. [Google Scholar] [CrossRef]
  52. Elbarbary, M.; Honda, T.; Morgan, G.; Kelly, P.; Guo, Y.; Negin, J. Ambient air pollution exposure association with diabetes prevalence and glycosylated hemoglobin (HbA1c) levels in China. Cross-sectional analysis from the WHO study of AGEing and adult health wave 1. J. Environ. Sci. Health Tox Hazard. Subst Environ. Eng. 2020, 55, 1149–1162. [Google Scholar] [CrossRef]
  53. Hwang, M.J.; Kim, J.H.; Koo, Y.S.; Yun, H.Y.; Cheong, H.K. Impacts of ambient air pollution on glucose metabolism in Korean adults: A Korea National Health and Nutrition Examination Survey study. Environ. Health 2020, 19, 70. [Google Scholar] [CrossRef]
  54. Lin, Y.; Zhou, S.; Liu, H.; Cui, Z.; Hou, F.; Feng, S.; Zhang, Y.; Liu, H.; Lu, C.; Yu, P. Risk Analysis of Air Pollution and Meteorological Factors Affecting the Incidence of Diabetes in the Elderly Population in Northern China. J. Diabetes Res. 2020, 2020, 3673980. [Google Scholar] [CrossRef] [PubMed]
  55. Chevalier, N.; Fenichel, P. Bisphenol A: Targeting metabolic tissues. Rev. Endocr. Metab. Disord. 2015, 16, 299–309. [Google Scholar] [CrossRef]
  56. Chevalier, N.; Fenichel, P. Endocrine disruptors: New players in the pathophysiology of type 2 diabetes? Diabetes Metab. 2015, 41, 107–115. [Google Scholar] [CrossRef]
  57. Berg, V.; Charles, D.; Bergdahl, I.A.; Nost, T.H.; Sandanger, T.M.; Tornevi, A.; Huber, S.; Fuskevag, O.M.; Rylander, C. Pre- and post-diagnostic blood profiles of chlorinated persistent organic pollutants and metabolic markers in type 2 diabetes mellitus cases and controls; a pilot study. Environ. Res. 2021, 195, 110846. [Google Scholar] [CrossRef] [PubMed]
  58. Howard, S.G. Developmental Exposure to Endocrine Disrupting Chemicals and Type 1 Diabetes Mellitus. Front. Endocrinol. 2018, 9, 513. [Google Scholar] [CrossRef] [PubMed]
  59. Conway, B.; Innes, K.E.; Long, D. Perfluoroalkyl substances and beta cell deficient diabetes. J. Diabetes Complicat. 2016, 30, 993–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Malarvannan, G.; Dirinck, E.; Dirtu, A.C.; Pereira-Fernandes, A.; Neels, H.; Jorens, P.G.; Gaal, L.V.; Blust, R.; Covaci, A. Distribution of persistent organic pollutants in two different fat compartments from obese individuals. Environ. Int. 2013, 55, 33–42. [Google Scholar] [CrossRef]
  61. Elten, M.; Donelle, J.; Lima, I.; Burnett, R.T.; Weichenthal, S.; Stieb, D.M.; Hystad, P.; van Donkelaar, A.; Chen, H.; Paul, L.A.; et al. Ambient air pollution and incidence of early-onset paediatric type 1 diabetes: A retrospective population-based cohort study. Environ. Res. 2020, 184, 109291. [Google Scholar] [CrossRef]
  62. Sheehan, A.; Freni Sterrantino, A.; Fecht, D.; Elliott, P.; Hodgson, S. Childhood type 1 diabetes: An environment-wide association study across England. Diabetologia 2020, 63, 964–976. [Google Scholar] [CrossRef] [Green Version]
  63. Sargis, R.M.; Heindel, J.J.; Padmanabhan, V. Interventions to Address Environmental Metabolism-Disrupting Chemicals: Changing the Narrative to Empower Action to Restore Metabolic Health. Front. Endocrinol. 2019, 10, 33. [Google Scholar] [CrossRef] [Green Version]
  64. Barker, D.J. The developmental origins of adult disease. Eur. J. Epidemiol. 2003, 18, 733–736. [Google Scholar] [CrossRef] [PubMed]
  65. 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] [Green Version]
  66. Yan, D.; Jiao, Y.; Yan, H.; Liu, T.; Yan, H.; Yuan, J. Endocrine-disrupting chemicals and the risk of gestational diabetes mellitus: A systematic review and meta-analysis. Environ. Health 2022, 21, 53. [Google Scholar] [CrossRef] [PubMed]
  67. Xing, Y.; Xia, W.; Zhang, B.; Zhou, A.; Huang, Z.; Zhang, H.; Liu, H.; Jiang, Y.; Hu, C.; Chen, X.; et al. Relation between cadmium exposure and gestational diabetes mellitus. Environ. Int. 2018, 113, 300–305. [Google Scholar] [CrossRef] [PubMed]
  68. Salmeri, N.; Villanacci, R.; Ottolina, J.; Bartiromo, L.; Cavoretto, P.; Dolci, C.; Lembo, R.; Schimberni, M.; Valsecchi, L.; Vigano, P.; et al. Maternal Arsenic Exposure and Gestational Diabetes: A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 3094. [Google Scholar] [CrossRef] [PubMed]
  69. Li, J.; Yao, J.; Xia, W.; Dai, J.; Liu, H.; Pan, Y.; Xu, S.; Lu, S.; Jin, S.; Li, Y.; et al. Association between exposure to per- and polyfluoroalkyl substances and blood glucose in pregnant women. Int J. Hyg. Environ. Health 2020, 230, 113596. [Google Scholar] [CrossRef] [PubMed]
  70. Xu, H.; Zhou, Q.; Zhang, J.; Chen, X.; Zhao, H.; Lu, H.; Ma, B.; Wang, Z.; Wu, C.; Ying, C.; et al. Exposure to elevated per- and polyfluoroalkyl substances in early pregnancy is related to increased risk of gestational diabetes mellitus: A nested case-control study in Shanghai, China. Environ. Int. 2020, 143, 105952. [Google Scholar] [CrossRef]
  71. Farrugia, F.; Aquilina, A.; Vassallo, J.; Pace, N.P. Bisphenol A and Type 2 Diabetes Mellitus: A Review of Epidemiologic, Functional, and Early Life Factors. Int. J. Environ. Res. Public Health 2021, 18, 716. [Google Scholar] [CrossRef]
  72. Vandenberg, L.N.; Chahoud, I.; Heindel, J.J.; Padmanabhan, V.; Paumgartten, F.J.; Schoenfelder, G. Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A. Environ. Health Perspect. 2010, 118, 1055–1070. [Google Scholar] [CrossRef] [Green Version]
  73. Kahn, L.G.; Philippat, C.; Nakayama, S.F.; Slama, R.; Trasande, L. Endocrine-disrupting chemicals: Implications for human health. Lancet Diabetes Endocrinol. 2020, 8, 703–718. [Google Scholar] [CrossRef]
  74. Berger, K.; Hyland, C.; Ames, J.L.; Mora, A.M.; Huen, K.; Eskenazi, B.; Holland, N.; Harley, K.G. Prenatal Exposure to Mixtures of Phthalates, Parabens, and Other Phenols and Obesity in Five-Year-Olds in the CHAMACOS Cohort. Int. J. Environ. Res. Public Health 2021, 18, 1796. [Google Scholar] [CrossRef] [PubMed]
  75. Harley, K.G.; Berger, K.; Rauch, S.; Kogut, K.; Claus Henn, B.; Calafat, A.M.; Huen, K.; Eskenazi, B.; Holland, N. Association of prenatal urinary phthalate metabolite concentrations and childhood BMI and obesity. Pediatr. Res. 2017, 82, 405–415. [Google Scholar] [CrossRef] [PubMed]
  76. Warner, M.; Ye, M.; Harley, K.; Kogut, K.; Bradman, A.; Eskenazi, B. Prenatal DDT exposure and child adiposity at age 12: The CHAMACOS study. Environ. Res. 2017, 159, 606–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. 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] [PubMed] [Green Version]
  78. Griffin, M.D.; Pereira, S.R.; DeBari, M.K.; Abbott, R.D. Mechanisms of action, chemical characteristics, and model systems of obesogens. BMC Biomed. Eng. 2020, 2, 6. [Google Scholar] [CrossRef] [PubMed]
  79. 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]
  80. Bokobza, E.; Hinault, C.; Tiroille, V.; Clavel, S.; Bost, F.; Chevalier, N. The Adipose Tissue at the Crosstalk Between EDCs and Cancer Development. Front. Endocrinol. 2021, 12, 691658. [Google Scholar] [CrossRef]
  81. Misra, B.B.; Misra, A. The chemical exposome of type 2 diabetes mellitus: Opportunities and challenges in the omics era. Diabetes Metab. Syndr. 2020, 14, 23–38. [Google Scholar] [CrossRef]
  82. Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev. 2015, 36, E1–E150. [Google Scholar] [CrossRef]
  83. Egusquiza, R.J.; Blumberg, B. Environmental Obesogens and Their Impact on Susceptibility to Obesity: New Mechanisms and Chemicals. Endocrinology 2020, 161, bqaa024. [Google Scholar] [CrossRef] [Green Version]
  84. Fenichel, P.; Chevalier, N. Environmental endocrine disruptors: New diabetogens? C. R. Biol. 2017, 340, 446–452. [Google Scholar] [CrossRef] [PubMed]
  85. Nadal, A.; Quesada, I.; Tuduri, E.; Nogueiras, R.; Alonso-Magdalena, P. Endocrine-disrupting chemicals and the regulation of energy balance. Nat. Rev. Endocrinol. 2017, 13, 536–546. [Google Scholar] [CrossRef] [PubMed]
  86. 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]
  87. Kim, Y.A.; Park, J.B.; Woo, M.S.; Lee, S.Y.; Kim, H.Y.; Yoo, Y.H. Persistent Organic Pollutant-Mediated Insulin Resistance. Int. J. Environ. Res. Public Health 2019, 16, 448. [Google Scholar] [CrossRef] [Green Version]
  88. Schulz, M.C.; Sargis, R.M. Inappropriately sweet: Environmental endocrine-disrupting chemicals and the diabetes pandemic. Adv. Pharm. 2021, 92, 419–456. [Google Scholar] [CrossRef]
  89. Velmurugan, G.; Ramprasath, T.; Gilles, M.; Swaminathan, K.; Ramasamy, S. Gut Microbiota, Endocrine-Disrupting Chemicals, and the Diabetes Epidemic. Trends Endocrinol. Metab. 2017, 28, 612–625. [Google Scholar] [CrossRef] [PubMed]
  90. Ruzzin, J.; Petersen, R.; Meugnier, E.; Madsen, L.; Lock, E.J.; Lillefosse, H.; Ma, T.; Pesenti, S.; Sonne, S.B.; Marstrand, T.T.; et al. Persistent organic pollutant exposure leads to insulin resistance syndrome. Environ. Health Perspect. 2010, 118, 465–471. [Google Scholar] [CrossRef] [Green Version]
  91. Ibrahim, M.M.; Fjaere, E.; Lock, E.J.; Naville, D.; Amlund, H.; Meugnier, E.; Le Magueresse Battistoni, B.; Froyland, L.; Madsen, L.; Jessen, N.; et al. Chronic consumption of farmed salmon containing persistent organic pollutants causes insulin resistance and obesity in mice. PLoS ONE 2011, 6, e25170. [Google Scholar] [CrossRef]
  92. Naville, D.; Pinteur, C.; Vega, N.; Menade, Y.; Vigier, M.; Le Bourdais, A.; Labaronne, E.; Debard, C.; Luquain-Costaz, C.; Begeot, M.; et al. Low-dose food contaminants trigger sex-specific, hepatic metabolic changes in the progeny of obese mice. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2013, 27, 3860–3870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Gray, S.L.; Shaw, A.C.; Gagne, A.X.; Chan, H.M. Chronic exposure to PCBs (Aroclor 1254) exacerbates obesity-induced insulin resistance and hyperinsulinemia in mice. J. Toxicol. Environ. Health A 2013, 76, 701–715. [Google Scholar] [CrossRef]
  94. Zhang, S.; Wu, T.; Chen, M.; Guo, Z.; Yang, Z.; Zuo, Z.; Wang, C. Chronic Exposure to Aroclor 1254 Disrupts Glucose Homeostasis in Male Mice via Inhibition of the Insulin Receptor Signal Pathway. Environ. Sci Technol. 2015, 49, 10084–10092. [Google Scholar] [CrossRef] [PubMed]
  95. Baker, N.A.; Karounos, M.; English, V.; Fang, J.; Wei, Y.; Stromberg, A.; Sunkara, M.; Morris, A.J.; Swanson, H.I.; Cassis, L.A. Coplanar polychlorinated biphenyls impair glucose homeostasis in lean C57BL/6 mice and mitigate beneficial effects of weight loss on glucose homeostasis in obese mice. Environ. Health Perspect. 2013, 121, 105–110. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, H.Y.; Kwon, W.Y.; Kim, Y.A.; Oh, Y.J.; Yoo, S.H.; Lee, M.H.; Bae, J.Y.; Kim, J.M.; Yoo, Y.H. Polychlorinated biphenyls exposure-induced insulin resistance is mediated by lipid droplet enlargement through Fsp27. Arch. Toxicol. 2017, 91, 2353–2363. [Google Scholar] [CrossRef] [PubMed]
  97. Wu, H.; Yu, W.; Meng, F.; Mi, J.; Peng, J.; Liu, J.; Zhang, X.; Hai, C.; Wang, X. Polychlorinated biphenyls-153 induces metabolic dysfunction through activation of ROS/NF-kappaB signaling via downregulation of HNF1b. Redox Biol. 2017, 12, 300–310. [Google Scholar] [CrossRef]
  98. Shi, H.; Jan, J.; Hardesty, J.E.; Falkner, K.C.; Prough, R.A.; Balamurugan, A.N.; Mokshagundam, S.P.; Chari, S.T.; Cave, M.C. Polychlorinated biphenyl exposures differentially regulate hepatic metabolism and pancreatic function: Implications for nonalcoholic steatohepatitis and diabetes. Toxicol. Appl. Pharmacol. 2019, 363, 22–33. [Google Scholar] [CrossRef] [PubMed]
  99. Kurita, H.; Yoshioka, W.; Nishimura, N.; Kubota, N.; Kadowaki, T.; Tohyama, C. Aryl hydrocarbon receptor-mediated effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on glucose-stimulated insulin secretion in mice. J. Appl. Toxicol. 2009, 29, 689–694. [Google Scholar] [CrossRef] [PubMed]
  100. Hoyeck, M.P.; Blair, H.; Ibrahim, M.; Solanki, S.; Elsawy, M.; Prakash, A.; Rick, K.R.C.; Matteo, G.; O’Dwyer, S.; Bruin, J.E. Long-term metabolic consequences of acute dioxin exposure differ between male and female mice. Sci. Rep. 2020, 10, 1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Wang, H.; Wang, J.; Zhu, Y.; Yan, H.; Lu, Y. Effects of Different Intensity Exercise on Glucose Metabolism and Hepatic IRS/PI3K/AKT Pathway in SD Rats Exposed with TCDD. Int. J. Environ. Res. Public Health 2021, 18, 13141. [Google Scholar] [CrossRef]
  102. Chen, Y.W.; Lan, K.C.; Tsai, J.R.; Weng, T.I.; Yang, C.Y.; Liu, S.H. Tributyltin exposure at noncytotoxic doses dysregulates pancreatic beta-cell function in vitro and in vivo. Arch. Toxicol. 2017, 91, 3135–3144. [Google Scholar] [CrossRef] [PubMed]
  103. Dos Santos, R.S.; Medina-Gali, R.M.; Babiloni-Chust, I.; Marroqui, L.; Nadal, A. In Vitro Assays to Identify Metabolism-Disrupting Chemicals with Diabetogenic Activity in a Human Pancreatic beta-Cell Model. Int. J. Mol. Sci. 2022, 23, 5040. [Google Scholar] [CrossRef]
  104. Le Magueresse-Battistoni, B.; Multigner, L.; Beausoleil, C.; Rousselle, C. Effects of bisphenol A on metabolism and evidences of a mode of action mediated through endocrine disruption. Mol. Cell. Endocrinol. 2018, 475, 74–91. [Google Scholar] [CrossRef]
  105. Akash, M.S.H.; Sabir, S.; Rehman, K. Bisphenol A-induced metabolic disorders: From exposure to mechanism of action. Environ. Toxicol. Pharm. 2020, 77, 103373. [Google Scholar] [CrossRef]
  106. Abulehia, H.F.S.; Mohd Nor, N.S.; Sheikh Abdul Kadir, S.H. The Current Findings on the Impact of Prenatal BPA Exposure on Metabolic Parameters: In Vivo and Epidemiological Evidence. Nutrients 2022, 14, 2766. [Google Scholar] [CrossRef] [PubMed]
  107. Beausoleil, C.; Le Magueresse-Battistoni, B.; Viguie, C.; Babajko, S.; Canivenc-Lavier, M.C.; Chevalier, N.; Emond, C.; Habert, R.; Picard-Hagen, N.; Mhaouty-Kodja, S. Regulatory and academic studies to derive reference values for human health: The case of bisphenol S. Environ. Res. 2022, 204, 112233. [Google Scholar] [CrossRef]
  108. Alharbi, H.F.; Algonaiman, R.; Alduwayghiri, R.; Aljutaily, T.; Algheshairy, R.M.; Almutairi, A.S.; Alharbi, R.M.; Alfurayh, L.A.; Alshahwan, A.A.; Alsadun, A.F.; et al. Exposure to Bisphenol A Substitutes, Bisphenol S and Bisphenol F, and Its Association with Developing Obesity and Diabetes Mellitus: A Narrative Review. Int. J. Environ. Res. Public Health 2022, 19, 15918. [Google Scholar] [CrossRef]
  109. Marroqui, L.; Martinez-Pinna, J.; Castellano-Munoz, M.; Dos Santos, R.S.; Medina-Gali, R.M.; Soriano, S.; Quesada, I.; Gustafsson, J.A.; Encinar, J.A.; Nadal, A. Bisphenol-S and Bisphenol-F alter mouse pancreatic beta-cell ion channel expression and activity and insulin release through an estrogen receptor ERbeta mediated pathway. Chemosphere 2021, 265, 129051. [Google Scholar] [CrossRef] [PubMed]
  110. Lin, Y.; Wei, J.; Li, Y.; Chen, J.; Zhou, Z.; Song, L.; Wei, Z.; Lv, Z.; Chen, X.; Xia, W.; et al. Developmental exposure to di(2-ethylhexyl) phthalate impairs endocrine pancreas and leads to long-term adverse effects on glucose homeostasis in the rat. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E527–E538. [Google Scholar] [CrossRef] [PubMed]
  111. Kloting, N.; Hesselbarth, N.; Gericke, M.; Kunath, A.; Biemann, R.; Chakaroun, R.; Kosacka, J.; Kovacs, P.; Kern, M.; Stumvoll, M.; et al. Di-(2-Ethylhexyl)-Phthalate (DEHP) Causes Impaired Adipocyte Function and Alters Serum Metabolites. PLoS ONE 2015, 10, e0143190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Wei, J.; Hao, Q.; Chen, C.; Li, J.; Han, X.; Lei, Z.; Wang, T.; Wang, Y.; You, X.; Chen, X.; et al. Epigenetic repression of miR-17 contributed to di(2-ethylhexyl) phthalate-triggered insulin resistance by targeting Keap1-Nrf2/miR-200a axis in skeletal muscle. Theranostics 2020, 10, 9230–9248. [Google Scholar] [CrossRef]
  113. Baralic, K.; Zivancevic, K.; Jorgovanovic, D.; Javorac, D.; Radovanovic, J.; Gojkovic, T.; Buha Djordjevic, A.; Curcic, M.; Mandinic, Z.; Bulat, Z.; et al. Probiotic reduced the impact of phthalates and bisphenol A mixture on type 2 diabetes mellitus development: Merging bioinformatics with in vivo analysis. Food Chem. Toxicol. 2021, 154, 112325. [Google Scholar] [CrossRef]
  114. Castriota, F.; Rieswijk, L.; Dahlberg, S.; La Merrill, M.A.; Steinmaus, C.; Smith, M.T.; Wang, J.C. A State-of-the-Science Review of Arsenic’s Effects on Glucose Homeostasis in Experimental Models. Environ. Health Perspect. 2020, 128, 16001. [Google Scholar] [CrossRef] [Green Version]
  115. Kirkley, A.G.; Carmean, C.M.; Ruiz, D.; Ye, H.; Regnier, S.M.; Poudel, A.; Hara, M.; Kamau, W.; Johnson, D.N.; Roberts, A.A.; et al. Arsenic exposure induces glucose intolerance and alters global energy metabolism. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2018, 314, R294–R303. [Google Scholar] [CrossRef] [PubMed]
  116. Khan, F.; Hodjat, M.; Rahimifard, M.; Nigjeh, M.N.; Azizi, M.; Baeeri, M.; Bayrami, Z.; Gholami, M.; Hassani, S.; Abdollahi, M. Assessment of arsenic-induced modifications in the DNA methylation of insulin-related genes in rat pancreatic islets. Ecotoxicol. Environ. Saf. 2020, 201, 110802. [Google Scholar] [CrossRef] [PubMed]
  117. Xenakis, J.G.; Douillet, C.; Bell, T.A.; Hock, P.; Farrington, J.; Liu, T.; Murphy, C.E.Y.; Saraswatula, A.; Shaw, G.D.; Nativio, G.; et al. An interaction of inorganic arsenic exposure with body weight and composition on type 2 diabetes indicators in Diversity Outbred mice. Mamm. Genome 2022, 33, 575–589. [Google Scholar] [CrossRef] [PubMed]
  118. Predieri, B.; Bruzzi, P.; Bigi, E.; Ciancia, S.; Madeo, S.F.; Lucaccioni, L.; Iughetti, L. Endocrine Disrupting Chemicals and Type 1 Diabetes. Int. J. Mol. Sci. 2020, 21, 2937. [Google Scholar] [CrossRef]
  119. Kuiper, J.; Moran, M.; Cetkovic-Cvrlje, M. Exposure to polychlorinated biphenyl-153 decreases incidence of autoimmune Type 1 diabetes in non-obese diabetic mice. J. Immunotoxicol. 2016, 13, 850–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Longnecker, M.P.; Daniels, J.L. Environmental contaminants as etiologic factors for diabetes. Environ. Health Perspect. 2001, 109 (Suppl. S6), 871–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Langer, P.; Tajtakova, M.; Guretzki, H.J.; Kocan, A.; Petrik, J.; Chovancova, J.; Drobna, B.; Jursa, S.; Pavuk, M.; Trnovec, T.; et al. High prevalence of anti-glutamic acid decarboxylase (anti-GAD) antibodies in employees at a polychlorinated biphenyl production factory. Arch. Environ. Health 2002, 57, 412–415. [Google Scholar] [CrossRef]
  122. Rignell-Hydbom, A.; Elfving, M.; Ivarsson, S.A.; Lindh, C.; Jonsson, B.A.; Olofsson, P.; Rylander, L. A nested case-control study of intrauterine exposure to persistent organochlorine pollutants in relation to risk of type 1 diabetes. PLoS ONE 2010, 5, e11281. [Google Scholar] [CrossRef] [Green Version]
  123. Bodin, J.; Groeng, E.C.; Andreassen, M.; Dirven, H.; Nygaard, U.C. Exposure to perfluoroundecanoic acid (PFUnDA) accelerates insulitis development in a mouse model of type 1 diabetes. Toxicol. Rep. 2016, 3, 664–672. [Google Scholar] [CrossRef] [Green Version]
  124. Cetkovic-Cvrlje, M.; Olson, M.; Schindler, B.; Gong, H.K. Exposure to DDT metabolite p,p′-DDE increases autoimmune type 1 diabetes incidence in NOD mouse model. J. Immunotoxicol. 2016, 13, 108–118. [Google Scholar] [CrossRef] [PubMed]
  125. Banerjee, B.D.; Ramachandran, M.; Hussain, Q.Z. Sub-chronic effect of DDT on humoral immune response in mice. Bull. Environ. Contam Toxicol. 1986, 37, 433–440. [Google Scholar] [CrossRef] [PubMed]
  126. Kerkvliet, N.I.; Steppan, L.B.; Vorachek, W.; Oda, S.; Farrer, D.; Wong, C.P.; Pham, D.; Mourich, D.V. Activation of aryl hydrocarbon receptor by TCDD prevents diabetes in NOD mice and increases Foxp3+ T cells in pancreatic lymph nodes. Immunotherapy 2009, 1, 539–547. [Google Scholar] [CrossRef] [PubMed]
  127. Bodin, J.; Kocbach Bolling, A.; Wendt, A.; Eliasson, L.; Becher, R.; Kuper, F.; Lovik, M.; Nygaard, U.C. Exposure to bisphenol A, but not phthalates, increases spontaneous diabetes type 1 development in NOD mice. Toxicol. Rep. 2015, 2, 99–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Xu, J.; Huang, G.; Guo, T.L. Bisphenol S Modulates Type 1 Diabetes Development in Non-Obese Diabetic (NOD) Mice with Diet- and Sex-Related Effects. Toxics 2019, 7, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Xu, J.; Huang, G.; Nagy, T.; Guo, T.L. Bisphenol A alteration of type 1 diabetes in non-obese diabetic (NOD) female mice is dependent on window of exposure. Arch. Toxicol. 2019, 93, 1083–1093. [Google Scholar] [CrossRef]
  130. Song, Y.; Yang, L. Transgenerational pancreatic impairment with Igf2/H19 epigenetic alteration induced by p,p′-DDE exposure in early life. Toxicol. Lett. 2017, 280, 222–231. [Google Scholar] [CrossRef]
  131. Robles-Matos, N.; Artis, T.; Simmons, R.A.; Bartolomei, M.S. Environmental Exposure to Endocrine Disrupting Chemicals Influences Genomic Imprinting, Growth, and Metabolism. Genes 2021, 12, 1153. [Google Scholar] [CrossRef]
  132. 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]
  133. Alonso-Magdalena, P.; Rivera, F.J.; Guerrero-Bosagna, C. Bisphenol-A and metabolic diseases: Epigenetic, developmental and transgenerational basis. Environ. Epigenet 2016, 2, dvw022. [Google Scholar] [CrossRef] [Green Version]
  134. Bansal, A.; Rashid, C.; Xin, F.; Li, C.; Polyak, E.; Duemler, A.; van der Meer, T.; Stefaniak, M.; Wajid, S.; Doliba, N.; et al. Sex- and Dose-Specific Effects of Maternal Bisphenol A Exposure on Pancreatic Islets of First- and Second-Generation Adult Mice Offspring. Environ. Health Perspect. 2017, 125, 097022. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, J.; Zhong, L.; Wu, J.; Ke, S.; Morpurgo, B.; Golovko, A.; Ouyang, N.; Sun, Y.; Guo, S.; Tian, Y. A Murine Pancreatic Islet Cell-based Screening for Diabetogenic Environmental Chemicals. J. Vis. Exp. 2018, 136, e57327. [Google Scholar] [CrossRef] [Green Version]
  136. Chen, A.C.; Lee, K.F.; Yeung, W.S.B.; Lee, Y.L. Human embryonic stem cells as an in vitro model for studying developmental origins of type 2 diabetes. World J. Stem Cells 2020, 12, 761–775. [Google Scholar] [CrossRef]
  137. Al-Abdulla, R.; Ferrero, H.; Soriano, S.; Boronat-Belda, T.; Alonso-Magdalena, P. Screening of Relevant Metabolism-Disrupting Chemicals on Pancreatic beta-Cells: Evaluation of Murine and Human In Vitro Models. Int. J. Mol. Sci. 2022, 23, 4182. [Google Scholar] [CrossRef]
  138. Fischer, L.J.; Wagner, M.A.; Madhukar, B.V. Potential involvement of calcium, CaM kinase II, and MAP kinases in PCB-stimulated insulin release from RINm5F cells. Toxicol. Appl. Pharmacol. 1999, 159, 194–203. [Google Scholar] [CrossRef] [PubMed]
  139. Fischer, L.J.; Zhou, H.R.; Wagner, M.A. Polychlorinated biphenyls release insulin from RINm5F cells. Life Sci. 1996, 59, 2041–2049. [Google Scholar] [CrossRef]
  140. Lee, Y.M.; Ha, C.M.; Kim, S.A.; Thoudam, T.; Yoon, Y.R.; Kim, D.J.; Kim, H.C.; Moon, H.B.; Park, S.; Lee, I.K.; et al. Low-Dose Persistent Organic Pollutants Impair Insulin Secretory Function of Pancreatic beta-Cells: Human and In Vitro Evidence. Diabetes 2017, 66, 2669–2680. [Google Scholar] [CrossRef] [Green Version]
  141. Park, C.M.; Kim, K.T.; Rhyu, D.Y. Low-concentration exposure to organochlorine pesticides (OCPs) in L6 myotubes and RIN-m5F pancreatic beta cells induces disorders of glucose metabolism. Toxicol. In Vitro 2020, 65, 104767. [Google Scholar] [CrossRef]
  142. Singh, V.K.; Sarkar, S.K.; Saxena, A.; Koner, B.C. Effect of Subtoxic DDT Exposure on Glucose Uptake and Insulin Signaling in Rat L6 Myoblast-Derived Myotubes. Int. J. Toxicol. 2019, 38, 303–311. [Google Scholar] [CrossRef]
  143. Pavlikova, N.; Sramek, J.; Jelinek, M.; Halada, P.; Kovar, J. Markers of acute toxicity of DDT exposure in pancreatic beta-cells determined by a proteomic approach. PLoS ONE 2020, 15, e0229430. [Google Scholar] [CrossRef]
  144. Ward, A.B.; Dail, M.B.; Chambers, J.E. In vitro effect of DDE exposure on the regulation of B-TC-6 pancreatic beta cell insulin secretion: A potential role in beta cell dysfunction and type 2 diabetes mellitus. Toxicol. Mech. Methods 2021, 31, 667–673. [Google Scholar] [CrossRef] [PubMed]
  145. Novelli, M.; Piaggi, S.; De Tata, V. 2,3,7,8-Tetrachlorodibenzo-p-dioxin-induced impairment of glucose-stimulated insulin secretion in isolated rat pancreatic islets. Toxicol. Lett. 2005, 156, 307–314. [Google Scholar] [CrossRef]
  146. 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]
  147. Kim, Y.H.; Shim, Y.J.; Shin, Y.J.; Sul, D.; Lee, E.; Min, B.H. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces calcium influx through T-type calcium channel and enhances lysosomal exocytosis and insulin secretion in INS-1 cells. Int. J. Toxicol. 2009, 28, 151–161. [Google Scholar] [CrossRef]
  148. Novelli, M.; Beffy, P.; Masini, M.; Vantaggiato, C.; Martino, L.; Marselli, L.; Marchetti, P.; De Tata, V. Selective beta-cell toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin on isolated pancreatic islets. Chemosphere 2021, 265, 129103. [Google Scholar] [CrossRef] [PubMed]
  149. Liu, P.C.; Matsumura, F. TCDD suppresses insulin-responsive glucose transporter (GLUT-4) gene expression through C/EBP nuclear transcription factors in 3T3-L1 adipocytes. J. Biochem. Mol. Toxicol. 2006, 20, 79–87. [Google Scholar] [CrossRef] [PubMed]
  150. Li, W.; Vogel, C.F.; Matsumura, F. Studies on the cell treatment conditions to elicit lipolytic responses from 3T3-L1 adipocytes to TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Cell Biochem. 2007, 102, 389–402. [Google Scholar] [CrossRef]
  151. Nishiumi, S.; Yoshida, M.; Azuma, T.; Yoshida, K.; Ashida, H. 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs an insulin signaling pathway through the induction of tumor necrosis factor-alpha in adipocytes. Toxicol. Sci. Off. J. Soc. Toxicol. 2010, 115, 482–491. [Google Scholar] [CrossRef] [Green Version]
  152. Kim, M.J.; Pelloux, V.; Guyot, E.; Tordjman, J.; Bui, L.C.; Chevallier, A.; Forest, C.; Benelli, C.; Clement, K.; Barouki, R. Inflammatory pathway genes belong to major targets of persistent organic pollutants in adipose cells. Environ. Health Perspect. 2012, 120, 508–514. [Google Scholar] [CrossRef] [Green Version]
  153. Arsenescu, V.; Arsenescu, R.I.; King, V.; Swanson, H.; Cassis, L.A. Polychlorinated biphenyl-77 induces adipocyte differentiation and proinflammatory adipokines and promotes obesity and atherosclerosis. Environ. Health Perspect. 2008, 116, 761–768. [Google Scholar] [CrossRef] [Green Version]
  154. Qin, W.P.; Cao, L.Y.; Li, C.H.; Guo, L.H.; Colbourne, J.; Ren, X.M. Perfluoroalkyl Substances Stimulate Insulin Secretion by Islet beta Cells via G Protein-Coupled Receptor 40. Environ. Sci. Technol. 2020, 54, 3428–3436. [Google Scholar] [CrossRef] [PubMed]
  155. He, X.; Wu, D.; Xu, Y.; Zhang, Y.; Sun, Y.; Chang, X.; Zhu, Y.; Tang, W. Perfluorooctanoic acid promotes pancreatic beta cell dysfunction and apoptosis through ER stress and the ATF4/CHOP/TRIB3 pathway. Environ. Sci. Pollut. Res. Int. 2022, 29, 84532–84545. [Google Scholar] [CrossRef] [PubMed]
  156. Bassler, J.; Ducatman, A.; Elliott, M.; Wen, S.; Wahlang, B.; Barnett, J.; Cave, M.C. Environmental perfluoroalkyl acid exposures are associated with liver disease characterized by apoptosis and altered serum adipocytokines. Environ. Pollut. 2019, 247, 1055–1063. [Google Scholar] [CrossRef] [PubMed]
  157. Costello, E.; Rock, S.; Stratakis, N.; Eckel, S.P.; Walker, D.I.; Valvi, D.; Cserbik, D.; Jenkins, T.; Xanthakos, S.A.; Kohli, R.; et al. Exposure to per- and Polyfluoroalkyl Substances and Markers of Liver Injury: A Systematic Review and Meta-Analysis. Environ. Health Perspect. 2022, 130, 46001. [Google Scholar] [CrossRef]
  158. Attema, B.; Janssen, A.W.F.; Rijkers, D.; van Schothorst, E.M.; Hooiveld, G.; Kersten, S. Exposure to low-dose perfluorooctanoic acid promotes hepatic steatosis and disrupts the hepatic transcriptome in mice. Mol. Metab. 2022, 66, 101602. [Google Scholar] [CrossRef]
  159. Qi, Q.; Niture, S.; Gadi, S.; Arthur, E.; Moore, J.; Levine, K.E.; Kumar, D. Per- and polyfluoroalkyl substances activate UPR pathway, induce steatosis and fibrosis in liver cells. Environ. Toxicol. 2023, 38, 225–242. [Google Scholar] [CrossRef]
  160. Park, C.; Song, H.; Choi, J.; Sim, S.; Kojima, H.; Park, J.; Iida, M.; Lee, Y. The mixture effects of bisphenol derivatives on estrogen receptor and androgen receptor. Environ. Pollut. 2020, 260, 114036. [Google Scholar] [CrossRef]
  161. Ferreira Azevedo, L.; Masiero, M.M.; Cherkaoui, S.; Hornos Carneiro, M.F.; Barbosa, F., Jr.; Zamboni, N. The alternative analog plasticizer BPS displays similar phenotypic and metabolomic responses to BPA in HepG2 and INS-1E cells. Food Chem. Toxicol. 2022, 167, 113266. [Google Scholar] [CrossRef]
  162. Li, L.; Wang, F.; Zhang, J.; Wang, K.; De, X.; Li, L.; Zhang, Y. Typical phthalic acid esters induce apoptosis by regulating the PI3K/Akt/Bcl-2 signaling pathway in rat insulinoma cells. Ecotoxicol. Environ. Saf. 2021, 208, 111461. [Google Scholar] [CrossRef]
  163. Qiu, T.; Wu, C.; Yao, X.; Han, Q.; Wang, N.; Yuan, W.; Zhang, J.; Shi, Y.; Jiang, L.; Liu, X.; et al. AS3MT facilitates NLRP3 inflammasome activation by m(6)A modification during arsenic-induced hepatic insulin resistance. Cell Biol. Toxicol. 2022, 1–17. [Google Scholar] [CrossRef]
  164. Todero, J.E.; Koch-Laskowski, K.; Shi, Q.; Kanke, M.; Hung, Y.H.; Beck, R.; Styblo, M.; Sethupathy, P. Candidate master microRNA regulator of arsenic-induced pancreatic beta cell impairment revealed by multi-omics analysis. Arch. Toxicol. 2022, 96, 1685–1699. [Google Scholar] [CrossRef]
  165. Meleleo, D.; Gerbino, A.; Mastrodonato, M. Evidence of the different effect of mercury and cadmium on the hIAPP aggregation process. Biophys. Chem. 2022, 290, 106880. [Google Scholar] [CrossRef] [PubMed]
  166. Hill, B.G.; Rood, B.; Ribble, A.; Haberzettl, P. Fine particulate matter (PM2.5) inhalation-induced alterations in the plasma lipidome as promoters of vascular inflammation and insulin resistance. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1836–H1850. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the main EDC in vivo effects related to diabetes occurrence (BPA: Bisphenol A; DEHP: di-2-ethylhexyl phthalate; DDE: dichlorodiphenyldichloroethylene; DDT: dichlorodiphenyltrichloroethane; EDCs: Endocrine-Disrupting Chemicals; F1/F2: Filial generation 1/2; PCB: polychlorinated biphenyls; TBT: Tributyltin; TCDD: 2,3,7,8-Tétrachlorodibenzo-p-dioxine).
Figure 1. Schematic representation of the main EDC in vivo effects related to diabetes occurrence (BPA: Bisphenol A; DEHP: di-2-ethylhexyl phthalate; DDE: dichlorodiphenyldichloroethylene; DDT: dichlorodiphenyltrichloroethane; EDCs: Endocrine-Disrupting Chemicals; F1/F2: Filial generation 1/2; PCB: polychlorinated biphenyls; TBT: Tributyltin; TCDD: 2,3,7,8-Tétrachlorodibenzo-p-dioxine).
Ijms 24 04537 g001
Figure 2. Schematic representation of the main EDC modes of action in relation to diabetes occurrence in adipocytes, hepatocytes or pancreatic beta cells. The figure summarizes the biological action in adipocytes, hepatocytes, or pancreatic beta cells. (AKT: Protein Kinase B; GPER: G Protein-coupled Estrogen Receptor; IRS: Insulin Receptor; ER: Estrogen Receptor; GLUT: Glucose Transporter; PI3K: Phosphatidylinositol 3 Kinase; PPAR: Peroxisome Proliferator-Activated Receptor; ROS: Reactive Oxygen Species; RXR: Retinoic X Receptor; SIRT3: Sirtuin 3).
Figure 2. Schematic representation of the main EDC modes of action in relation to diabetes occurrence in adipocytes, hepatocytes or pancreatic beta cells. The figure summarizes the biological action in adipocytes, hepatocytes, or pancreatic beta cells. (AKT: Protein Kinase B; GPER: G Protein-coupled Estrogen Receptor; IRS: Insulin Receptor; ER: Estrogen Receptor; GLUT: Glucose Transporter; PI3K: Phosphatidylinositol 3 Kinase; PPAR: Peroxisome Proliferator-Activated Receptor; ROS: Reactive Oxygen Species; RXR: Retinoic X Receptor; SIRT3: Sirtuin 3).
Ijms 24 04537 g002
Table 1. Evidence for EDCs as obesogenic and diabetogenic (T1D: type 1 diabetes; T2D: type 2 diabetes; GDM: gestational diabetes mellitus).
Table 1. Evidence for EDCs as obesogenic and diabetogenic (T1D: type 1 diabetes; T2D: type 2 diabetes; GDM: gestational diabetes mellitus).
EDCsIn HumansIn VivoIn Vitro
Non persistentBPAStrong (T2D)/Low (T1D)Strong (T2D)/Controversial (F1/F2)Strong
BPA substitutesLimited dataLimited dataLimited data
Phthalates/DEHPControversial (T2D)/Strong (GDM)/Strong (Obesity)Low (T2D)/Strong (Obesity)Strong
ArsenicLow (T2D)Strong (T2D)Strong
PersistentPCBsStrong (T2D)/Suggestive (T1D)/Strong (GDM)Strong (T2D)/Strong (Obesity)Strong
PFOALow (T2D)/Strong (GDM)Low (T2D)Low
TCDDStrong (T2D)Strong (T2D)Strong
DDT/DDEStrong (T2D)/Strong (Obesity)Strong (F1/F2)/Strong (Obesity)Strong
TBTLow (T2D, T1D)/Strong (Obesity)Suggestive (T2D)/Strong (Obesity)Low
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hinault, C.; Caroli-Bosc, P.; Bost, F.; Chevalier, N. Critical Overview on Endocrine Disruptors in Diabetes Mellitus. Int. J. Mol. Sci. 2023, 24, 4537. https://doi.org/10.3390/ijms24054537

AMA Style

Hinault C, Caroli-Bosc P, Bost F, Chevalier N. Critical Overview on Endocrine Disruptors in Diabetes Mellitus. International Journal of Molecular Sciences. 2023; 24(5):4537. https://doi.org/10.3390/ijms24054537

Chicago/Turabian Style

Hinault, Charlotte, Philippe Caroli-Bosc, Frédéric Bost, and Nicolas Chevalier. 2023. "Critical Overview on Endocrine Disruptors in Diabetes Mellitus" International Journal of Molecular Sciences 24, no. 5: 4537. https://doi.org/10.3390/ijms24054537

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