Next Article in Journal
Programmed Cell Death Ligand 1 Immunohistochemical Expression and Cutaneous Melanoma: A Controversial Relationship
Previous Article in Journal
Breed-Related Differential microRNA Expression and Analysis of Colostrum and Mature Milk Exosomes in Bamei and Landrace Pigs
Previous Article in Special Issue
Gonadotropin-Releasing Hormone Receptor (GnRHR) and Hypogonadotropic Hypogonadism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Endocrine Disruptors Bisphenols and Phthalates in Obesity: Current Evidence, Perspectives and Controversies

by
Maria Dalamaga
1,*,
Dimitrios Kounatidis
2,†,
Dimitrios Tsilingiris
3,†,
Natalia G. Vallianou
2,
Irene Karampela
4,
Sotiria Psallida
5 and
Athanasios G. Papavassiliou
1,*
1
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Internal Medicine, ‘Evangelismos’ General Hospital, 10676 Athens, Greece
3
First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece
4
Second Department of Critical Care, ‘Attikon’ General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
5
Department of Microbiology, ‘KAT’ General Hospital of Attica, 14561 Athens, Greece
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(1), 675; https://doi.org/10.3390/ijms25010675
Submission received: 11 December 2023 / Revised: 31 December 2023 / Accepted: 3 January 2024 / Published: 4 January 2024
(This article belongs to the Special Issue Hormone Receptors: A 2023 Update)

Abstract

:
Excess body weight constitutes one of the major health challenges for societies and healthcare systems worldwide. Besides the type of diet, calorie intake and the lack of physical exercise, recent data have highlighted a possible association between endocrine-disrupting chemicals (EDCs), such as bisphenol A, phthalates and their analogs, and obesity. EDCs represent a heterogeneous group of chemicals that may influence the hormonal regulation of body mass and adipose tissue morphology. Based on the available data from mechanistic, animal and epidemiological studies including meta-analyses, the weight of evidence points towards the contribution of EDCs to the development of obesity, associated disorders and obesity-related adipose tissue dysfunction by (1) impacting adipogenesis; (2) modulating epigenetic pathways during development, enhancing susceptibility to obesity; (3) influencing neuroendocrine signals responsible for appetite and satiety; (4) promoting a proinflammatory milieu in adipose tissue and inducing a state of chronic subclinical inflammation; (5) dysregulating gut microbiome and immune homeostasis; and (6) inducing dysfunction in thermogenic adipose tissue. Critical periods of exposure to obesogenic EDCs are the prenatal, neonatal, pubertal and reproductive periods. Interestingly, EDCs even at low doses may promote epigenetic transgenerational inheritance of adult obesity in subsequent generations. The aim of this review is to summarize the available evidence on the role of obesogenic EDCs, specifically BPA and phthalate plasticizers, in the development of obesity, taking into account in vitro, animal and epidemiologic studies; discuss mechanisms linking EDCs to obesity; analyze the effects of EDCs on obesity in critical chronic periods of exposure; and present interesting perspectives, challenges and preventive measures in this research area.

1. Introduction

Over recent decades, the escalating global increase in overweight and obesity, also known as “globesity”, has constituted one of the major health challenges for societies and healthcare systems worldwide. Based on the World Obesity Atlas 2023 report, approximately 38% of the world population presents with excess body weight, having a body mass index (BMI) of more than 25 kg/m2 [1]. This global prevalence of overweight and obesity is expected to reach 51% by 2035, while 78% of US adults are estimated to live with excess body weight by 2030 [2,3]. More striking than the elevated obesity rates in adults is the higher prevalence of excess body weight among children and adolescents, which has doubled or tripled in children of school age in many developed regions of the world [3,4]. Moreover, obesity is associated with a plethora of comorbidities, including diabetes mellitus type 2 (T2DM), coronary heart disease, hypertension, stroke, dyslipidemia, sleep apnea, osteoarthritis and some types of cancer [5,6,7,8].
During the past few decades, several research teams have investigated the association between common chemical exposures and the occurrence of allergy, asthma, immune dysfunction, cancer and other entities, including obesity [9,10]. The contemporary industrialized environment in developed countries is a constant source of a variety of chemical substances, presenting persistence and bioaccumulation potency in the food chain. Moreover, unrecognized or little-recognized environmental chemicals, such as industrial endocrine-disrupting chemicals (EDCs) or endocrine disruptors, have the potential to disrupt the actions of hormones, causing adverse health effects. Particularly, some EDCs that have been termed “obesogens” may also influence adipogenesis and regulatory metabolic pathways, leading to an imbalance in the regulation of body weight resulting in weight gain and obesity [11]. The most important EDCs that have been suspected in the development of obesity and obesity-associated metabolic disorders are the plastic additive bisphenol A (BPA) and the phthalate-based plasticizers. The aim of this narrative review is to summarize the available evidence on the role of EDCs, specifically BPA and phthalates, in the development of obesity, taking into account mechanistic, animal and epidemiologic studies; discuss mechanisms linking EDCs to obesity; analyze the effects of EDCs on obesity in critical chronic periods of exposure; and present interesting perspectives and challenges in the research area.

2. Methods of Literature Search and Review Criteria

Although this manuscript is not a systematic review, for its preparation, we applied the MESH search terms “obesity” and “endocrine-disrupting chemicals” in the PubMed NIH database from inception until 20 October 2023, which yielded 670 outputs. Among the 670 manuscripts, we excluded 19 manuscripts that were associated with brain and/or neurogenerative disorders, 14 manuscripts that were related to environmental factors only, 11 manuscripts that were referring to cancer, 4 articles related to rheumatic/bone diseases, 3 articles related to infectious diseases and 1 item that was a book and not an article. Furthermore, five manuscripts were written in French, two in German, one in Chinese and one in Japanese. Therefore, out of the 670 outputs, 61 were excluded for the aforementioned reasons, leaving a total of 609 manuscripts included in this literature search. Finally, we acknowledge that all these manuscripts cannot be covered in the context of this review.

3. Genetic and Environmental Factors in Obesity

Obesity is a multifactorial disorder characterized by abnormal or excessive fat accumulation that results from either an increase in the adipocyte size (hypertrophia) or an increase in the number of adipocytes (hyperplasia) and presents several risks to health. In clinical practice, BMI, which is closely associated with body fatness, is recommended by the WHO as a population-level measure of overweight and obesity, defined as a BMI of or above 25 kg/m2 and 30 kg/m2, respectively. [12,13,14]. Body fat percentage (BF%) as a % of total body weight presents some advantages over BMI in evaluating fat mass. The BF% cut-off points for obesity proposed by the WHO are 25% for men and 35% for women, corresponding to a BMI of 30 kg/m2 in young Caucasians [15].
Obesity represents a complex disorder due to a multifactorial etiology comprising genetic, epigenetic, environmental, societal and behavioral factors. Increased calorie diet, sedentary lifestyle and decreased energy expenditure play an important role [16,17]. Obesity has a strong genetic background demonstrated by family, twin and adoption studies that have shown heritability rates from 40 to 70% for BMI [18,19,20,21]. Nevertheless, non-syndromic monogenic obesity involves less than 5% of the obese population. More than 95% of subjects with obesity present common polygenic obesity, which is multifactorial and still poorly explained [22]. In particular, the relative contribution of polygenic scores from genome-wide association studies (GWASs) has been estimated to be between 2 and 8%, with the greater part of the BMI variation being unexplained [23]. The dominant challenge is a combination of genetic and environmental parameters such as physical exercise, energy intake and tobacco smoking, also including gene–gene and gene–environment interactions that underlie the complex and dynamic etiopathogenesis of obesity [24,25]. Furthermore, the accelerated global spread of excess body weight cannot be attributed to genetic factors only [26]. However, the availability of high-calorie food, the type of diet and physical inactivity are not sufficient to explain the striking increase in excess body weight. Therefore, environmental parameters may be responsible for the elevated rates of increased body weight during the last few decades [27]. Recent data have highlighted the role of other environmental factors contributing to the increase in BMI, such as EDCs.

4. The Spectrum of Endocrine Disruptors

Recent data from observational, animal and experimental studies have shown that certain chemical substances, such as EDCs, may have an impact on the endocrine system, being involved in the development and rapid propagation of obesity [28]. Based on the definition by the “Global assessment on the state of the science of endocrine disruptors” of the World Health Organization (WHO) in 2002, an endocrine disruptor has been described as “an exogenous substance or mixture that alters functions of the endocrine system and consequently causes adverse health effects in an intact organism, or its progeny, or (sub)populations” [29]. Certain EDCs are found in nature, such as phytoestrogens; however, the majority of EDCs are synthetic compounds that have been released through human activities into the ecosystem. Humans are exposed to endocrine disruptors through various sources in their daily lives, in outdoor and indoor environments and via the use of personal care and household products, industrial chemicals, pharmaceuticals, pesticides, herbicides, fungicides and flame retarders; air pollution; and dietary habits. EDCs constitute a highly heterogeneous group of chemical substances that can be categorized according to their chemical structure and properties, their occurrence and intended use, their mechanism of action, the potential direct or indirect impact on the endocrine system, the accumulation in the organism, their environmental persistence and the described or suggested clinical implications [30]. The catalog of EDCs encompasses agrochemicals, such as pesticides, herbicides and fungicides; industrial organic solvents/lubricants and their byproducts (dioxins, polychlorinated bisphenyls, polybrominated bisphenyls); pharmaceutical substances; catalysts; and plastic contaminants and plasticizers, such as bisphenols and phthalates [31]. In comparison to other EDCs, bisphenols and phthalates are metabolized and excreted relatively quickly (i.e., half-lives less than 24 h) and hence are considered nonpersistent [32]. Despite their nonpersistent nature, their ubiquitous and frequent use in a wide variety of consumer products throughout life leads to a chronic exposure, reported to be global [33].
Potential targets of EDCs include any endocrine organ, hormonal system and/or hormonally affected pathway [34]. Some of these EDCs may interfere with the regulation of metabolism, energy balance and the storage of fat in the organism, leading to the development of obesity by affecting the function of adipose tissue and disrupting metabolic endocrine signaling [35]. Figure 1 depicts some EDCs that have been implicated in obesity. Some of the best-documented groups of EDCs with obesogenic properties and widespread exposure in the general population are bisphenols and phthalates, which are mainly found in plastic products.
Globally, the production of plastics has increased significantly since the mid-20th century (Figure 2). After a stagnation in 2020 due to the emergence of the COVID-19 pandemic, this production reached 390.7 million tons in 2021 [36]. Interestingly, there was a parallel striking epidemic rise in obesity/overweight globally, as depicted in Figure 2, mainly attributed to the international food production and supply system [3,37]. Since 1970, food production and supply with ameliorated manufacturing and distribution systems have radically shifted in the direction of elevated energy availability [3].
Worldwide, more people have access to palatable, cheap and ultra-processed foods of lower nutritional quality, and a number of obesogenic EDCs have gradually reached the global food chain, with a potential influence on human metabolism. Moreover, plasticizers are the main additives in the manufacture of plastic products, commonly used in the production of food packaging materials to improve flexibility, durability, processing and resistance to heat, fire and UV radiation. All these chemicals may be detached from polymer compounds and leak into the surrounding environment after being degraded. Humans may be exposed to those EDCs through various routes including food ingestion, the respiratory tract and dermal exposure [10].

4.1. Bisphenol A

Bisphenol A (BPA), or 4,4 -isopropylidenediphenol 2,2-bis (4-hydroxy-phenyl)- propane, was first reported by the chemist A.P. Dianin in 1891. It is a functional diphenyl compound that possesses two hydroxyl groups in the “para” position (Figure 3), which allows it to bind with androgen and estrogen receptors (classical nuclear ERα and ERβ, ERγ and membrane-associated GPR30) as an antagonist or agonist [38]. BPA is classified as a xenoestrogen due to its similarity with diethylstilbestrol, a synthetic estrogen, and as an EDC which enhances the ER with lower affinity compared to 17β-estradiol [39,40].
Due to its elastic, cross-linking, polymer-forming and intrinsic heat resistance properties, BPA represents one of the world’s most heavily produced synthetic industrial chemical compounds listed by the Organization for Economic Cooperation and Development, with more than 15 billion pounds produced worldwide annually [41,42,43] and over 1 million pounds leached into the environment [44]. BPA is used worldwide in the synthesis of polycarbonate plastics, plastic consumer products, drink and food packaging, plastic bags, water bottles, epoxy resin linings of beverage containers and canned food, dental materials such as sealants, electronic equipment, toys, optical lenses, paper coatings, adhesives, dye developers and thermal papers [45].
BPA can be released into the surrounding environment through various means including exposure to heat or acidic conditions, hydrolysis or degradation of the polymer and constant diffusion of residual BPA that remains on the polymer [46]. The quantity of released BPA is affected by duration of exposure, processing methodology and environmental conditions such as temperature and PH [47]. Human exposure to nanomolar concentrations of environmental BPA is continuous and widespread via oral, skin and respiratory absorption from atmospheric exposure, and dust particles from commercial and residential environments [48]. Exposure to BPA in utero and through lactation is also important for the developing fetus and the neonate, respectively [49,50]. However, the main route of exposure originates from water and diet, as BPA may migrate from beverage and food packaging, as well as from dental sealants [46,51]. Interestingly, skin exposure is more severe than oral intake due to the sustained presence of BPA in the organism and the elevated plasma concentrations of unconjugated toxic BPA [52]. However, this kind of exposure does not reflect a natural setting. Moreover, Christensen et al. reported that the majority of BPA exposure was food-related (i.e., via leaching from food packaging materials and containers) after a fasting study to exclude food intake as a potential route of BPA exposure [53].
BPA and its conjugates have been detected in various body fluids and tissues, including urine, saliva, plasma, feces, amniotic fluid and breast milk; however, urine samples are mainly examined for human monitoring. Epidemiologic studies have confirmed the widespread exposure to BPA with 95 to 99.8% of adults, adolescents, children and infants presenting detectable concentrations of BPA and its metabolites in urine, independently of gender, income, educational level or BMI [54,55]. Additionally, circulating free unconjugated BPA, which is the active form of BPA, is usually determined at concentrations of nanograms per millimeter in serum or plasma [56]. BPA kinetics analyses have shown that the rate of excretion of BPA is not highly influenced by fasting, suggesting a slower rate of BPA excretion or a potential bioaccumulation of BPA in human tissues, particularly in adipose tissue [45,49]. Interestingly, in a study of the urinary BPA profile in five individuals over a 48-h period of fasting (bottled water only), BPA levels increased after the pre-fast meal, decreased over the next 24 h, fluctuated at lower levels during the second day, and then rose after the post-fast meal [53]. This rise may be attributed to non-food sources that could be still present, such as dust, or the release of BPA from lipid reservoirs from past exposures [53].
Exposure to BPA has been associated with a plethora of disorders such as obesity, T2DM, cardiovascular disease, infertility, neurodegenerative diseases and cancer, particularly breast cancer [57]. Moreover, BPA is classified as a potentially toxic and harmful substance to reproduction and eyes, respectively, and a possibly irritating substance to the skin and respiratory tract [58].
Due to increasing concerns about the safety of BPA and evidence of its relationship with human health, BPA has been banned in the manufacture of baby bottles since 2011 in the European Union (EU). The U.S. Food and Drug Administration (FDA) banned the use of BPA in baby bottles, spill-proof cups and infant formula packaging materials in 2012–2013. In April 2023, the European Food Safety Authority (EFSA) published a re-evaluation of BPA’s safety, drastically decreasing the tolerable daily intake (TDI) for BPA from 4 μg/kg of body weight per day in 2015 to 0.2 ng/kg (around 20,000 times lower than before) [59]. These restrictions were mainly based on toxicological animal studies that appraised BPA side effects on renal, hepatic and immune function [46,60]. Nevertheless, since there is an absence of international standardization regarding a tolerable BPA limit, BPA is actually used in the production of polycarbonate plastic food materials. Following the bans and restrictions, the industry has gradually employed a variety of less-studied BPA analogs with toxicological characteristics that are not fully elucidated. However, like BPA, alternatives such as bisphenols S (BPS), F (BPF), B (BPB), C (BPC), E (BPE), AF (BPAF), P (BPP) and Z (BPZ) and 4-cumylphenol (HPP) have also been identified as EDCs, presenting androgenic, estrogenic and obesogenic activity in vitro as well as neurotoxic, genotoxic and cytotoxic potential [46,57,60,61,62,63,64]. BPA analogs may bind to nuclear receptors due to their phenyl moiety and hydrophobic structure that play a role in endocrine-disrupting activity [65].

4.2. Phthalates

Phthalates are a group of chemical compounds in the family of esters derived from phthalic acid. The common chemical structure of phthalates involves a benzene ring with two carboxylic acid groups (ortho-phthalates, Figure 3) or one carboxylic acid group (para-phthalates) attached. The length and structure of the alkyl side chains attached to these carboxylic acid groups vary, resulting in different types of phthalates with specific properties [66]. Ortho-phthalates represent the most common type of phthalates and include diethyl phthalate (DEP), dibutyl phthalate (DBP), diisobutyl phthalate (DIBP), diisononyl phthalate (DINP) and di(2-ethylhexyl) phthalate (DEHP). Phthalates first appeared in the 1920s, and their widespread use as plasticizers destined primarily to soften polyvinyl chloride (PVC) started in the 1930s. Currently, the total production of phthalates is approximately 5.5 million tons per year with an increasing rise attributed to the use of PVC [67]. Phthalates constitute a large fraction of polymer additives due to the presence of various beneficial properties, such as flexibility, compatibility with polymers, chemical stability and durability, low water solubility, resistance to heat and weather conditions, electrical resistivity and transparency. They are primarily used as plasticizers in the production of flexible PVC products (vinyl flooring, PVC cables and wires, etc.); personal care products in cosmetics, perfumes and lotions to enhance fragrance and texture; toys and children’s products; adhesives and sealants; medical devices; building materials; coatings and inks; textiles and carpets; and food packaging [67,68].
Because phthalates are not covalently bound to their compounds, they may easily migrate and leak into the environment, where they can accumulate in the food chain due to their lipophilic nature [69]. Hence, humans are exposed to phthalates through dietary exposure, which represents the major route for intake; skin absorption; and inhalation of indoor and outdoor air comprising dust [30,70]. Infants, toddlers and children have a higher exposure to phthalate esters than adults through their mouthing activity and contact with toys, carpets and floors [30]. Furthermore, certain phthalates have the ability to pass through the placenta, potentially exposing the developing fetus to these chemicals during pregnancy [71,72]. Epidemiological studies have shown that phthalates, particularly the commonly used DEHP and its breakdown products, may be found in a plethora of human body fluids such as urine, which is the most commonly studied and used matrix for assessing human exposure to phthalates; plasma; saliva; follicular fluid; amniotic fluid; and breast milk [72,73,74,75]. Phthalate esters present endocrine-disrupting properties associated with detrimental reproductive and neurodevelopmental effects as well as obesity and T2DM [30,76,77,78]. In response to concerns about the potential harmful effects of phthalates, the use of certain phthalates including DEHP has been restricted in concentrations exceeding 0.1% by weight of the plasticized products in Europe and elsewhere [79]; nevertheless, there are no international acceptable limits for phthalate esters. Efforts have been made to reduce or replace their use in various applications with non-phthalate and bio-based plasticizers, polymer blends and formulations, and alternative materials. Interestingly, exposure to most EDCs, including DEHP metabolites and BPA, decreased between 2009 and 2016 in a sample of individuals with impaired fasting glucose from the Dutch population [80]. However, there is a need to assess the use of less toxic substitute chemical compounds for their metabolic and endocrine consequences.

5. Mechanisms Linking Endocrine Disruptors to Obesity

The hormonal system plays a crucial role in the regulation of carbohydrate, lipid and protein metabolism as well as in the regulation of body mass. Moreover, adipose tissue is not considered a passive storage depot for energy; it is also recognized as a large endocrine organ secreting a plethora of various hormones and bioactive molecules (adipokines) that play a critical role in regulating metabolism and body mass as well as other functions [81]. EDCs are xenobiotics exhibiting endocrine disruptor properties by binding to various nuclear receptors, altering specific cellular responses. These actions are pleiotropic depending on the cell and receptor types, receptor density, critical chronic periods of exposure, the presence of other cofactors, etc. Some important properties of EDCs include the following: (1) their effects at very low concentrations in a similar fashion to hormonal effects; (2) the exhibition of U-shaped nonmonotonic responses where decreased levels may cause phenotypic alterations not necessarily seen with increased doses; (3) agonistic or antagonistic actions after interacting with hormonal receptors resulting in complex unpredictable biological responses with stimulation, mimicking or inhibition of hormonal effects; (4) their impact on the number of cellular receptors and the level of serum hormones; and (5) their accumulation in adipose tissue due to their lipophilic nature which further increases the retention of other lipophilic chemicals [82].
BPA and phthalates may interact with several nuclear hormone receptors including ERα and ERβ, with an affinity being 103–104 times lower compared to 17β-estradiol (E2), androgen receptor (AR), progesterone receptor (PR), glucocorticoid receptor (GR), thyroid hormone receptors (TRα and TRβ), G-protein-coupled receptor 30 (GPR30), estrogen-related γ receptor (ERR-γ), mineralocorticoid receptor (MR), peroxisome-proliferator-activated receptors (PPARs) and retinoid receptors [83,84,85,86,87]. Obesogenic EDCs such as BPA and phthalates may be implicated in the etiopathogenesis of obesity and associated metabolic disorders by (1) increasing the number and size of adipocytes through the regulation of genes involved in adipogenesis; (2) modulating epigenetic pathways during development, enhancing susceptibility to obesity; (3) impacting neuroendocrine signals responsible for appetite and satiety; (4) promoting a proinflammatory milieu in adipose tissue and inducing a state of chronic subclinical inflammation; (5) dysregulating gut microbiome and immune homeostasis; and (6) inducing dysfunction in thermogenic adipose tissues. Figure 4 depicts the main mechanisms linking EDCs to excess body weight.
Based on animal and epidemiological studies, critical periods of exposure to obesogenic EDCs are in utero and the neonatal period. Moreover, EDCs can promote epigenetic transgenerational inheritance of obesity in adulthood in subsequent generations [88]. Fetal development is sensitive to maternal exposure to infectious agents, alcohol, drugs and toxins including EDCs [89]. The majority of EDCs cross the placenta barrier through active or passive transportation and may be found in the amniotic fluid or the placenta. Another critical period for exposure to EDCs is puberty due to the activation of the hypothalamic–pituitary–adrenal and gonadal axes. Certain animal studies have shown precocity or delay of puberty after exposure to EDCs with implications in body mass [89].

5.1. Effects on Adipogenesis

Obesity is characterized by the expansion of fat tissue, known as adipogenesis, attributed to hyperplasia via the differentiation of resident precursors or their increase in size (hypertrophy). Current data have shown that the number of adipocytes is set by the end of childhood, constituting a major predictor of fat mass in adulthood [90]. EDCs may influence both the linear commitment from mesenchymal stem cells (MSCs) to pre-adipocytes and the differentiation of pre-adipocytes into mature adipocytes, regulated by several adipogenic transcription factors such as the group of CCAAT/enhancer-binding family of proteins (C/EBPα, β, δ) that stimulate the nuclear [35] receptor PPAR-γ which is a critical regulator of adipocyte differentiation [91].
Mechanistic studies performed in 3T3-L1 cells (type of mouse fibroblast-like cell line that can differentiate into adipocytes), human adipose-derived stem/stromal cells and uncommitted NIH/3T3 cells have shown that exposure to BPA and its analogs upregulates the expression of C/EBPα and PPARγ, inducing adipocyte differentiation and lipid accumulation [61,92,93,94]. Although some phthalates and related metabolites exhibit greater effects on PPARα, particularly in mice, they can be potent and selective activators of PPARγ promoting adipogenesis [86,95,96,97]. Additive effects of a mixture of EDCs on adipogenesis may also be observed [35]. Interestingly, exposure to environmentally relevant doses of BPA, phthalates or their analogs in early life may result in elevated body weight and fat tissue mass in male and female mice by altering the recruitment and differentiation of adipocytes [98,99]. Finally, in human studies, an association between higher urinary BPA levels and obesity, metabolic syndrome, T2DM and cardiovascular disease has been reported in the general adult US population [100,101].
The exact molecular mechanism through which BPA and phthalate induce adipocyte differentiation is unclear; however, many mechanisms are unraveled and synopsized in Section 6 and Section 7.

5.2. Effects on Epigenetic Regulation

Out of the many potential mechanisms modulating gene expression in adipose tissue, epigenetic alterations have been of particular interest in the last few years. Epigenetics is defined as alterations in gene function, such as DNA methylation, histone modifications and microRNA (miRNA) interference, that occur without any modification in the DNA sequence.
EDCs may affect epigenetic pathways during early development and childhood, resulting in epigenetic alterations and predisposition to obesity. The interconnection of obesogenic EDCs and epigenetic alterations involves DNA methylation affecting the activity of DNA methyltransferases and their cofactors such as methyl donor S-adenosylmethionine or the modulation of locus-specific epigenetic patterns, histone modifications and miRNA expression, with most data focusing on DNA methylation [88].
BPA was shown to reduce global DNA methylation and activate adipocyte differentiation in 3T3-L1 cells [102]. Thus, altered epigenetic gene regulation could play a role in the association between BPA exposure and obesity development. In another study, prolonged low-dose BPA exposure in committed 3T3L1 and uncommitted NIH3T3 pre-adipocytes showed that its effects on adipogenesis are mediated through epigenetic mechanisms via a reduction in the PPAR-γ promoter methylation in pre-adipocytes mainly [103]. Interestingly, the ending of the BPA exposure may reverse the PPAR-γ promoter methylation and inflammatory pattern of the 3T3L1 cells [103]. Therefore, preventing BPA exposure is important for metabolic health. Moreover, butyl benzyl phthalate (BBP) may induce differentiation of 3T3-L1 cells and C3H10T1/2 stem cell lines, with increased expression levels of miR-34a-5p. MiR-34a-5p is involved in adipogenesis, obesity and potentially the epigenetic regulation of insulin signaling [104,105].
Developmental BPA exposure may modify and reprogram the liver β-oxidation function in male rats via the epigenetic regulation of genes (DNA methylation and histone modifications) implicated in β-oxidation, such as the carnitine palmitoyltransferase (Cpt1α) gene [106]. BPA changed DNA methylation and histone marks (H3Ac, H4Ac, H3Me2K4, H3Me3K36) and reduced the binding of several transcription factors (Pol II, C/EBPβ, SREBP1) in the Cpt1a gene, which is critical for β-oxidation [106]. Hence, BPA toxicity is characterized by DNA methylation and histone modifications.
Exposure to phthalate and its metabolites in utero may affect DNA methylation, leading to long-term implications in body weight [88]. Prenatal exposure to DEHP may impact fetal development [107]. A cord blood epigenome-wide study identified elevated methylation changes in genes associated with metabolism, the endocrine system and signaling pathways that were positively linked to maternal blood mono-(2-ethylhexyl) phthalate (MEHP) levels [107].
Larger and longitudinal multi-omics studies are needed to elucidate the effects of prenatal exposure to EDCs on fetal growth and long-term metabolic outcomes.

5.3. Effects on Neuroendocrine Signals of Appetite and Satiety

Obesogenic EDCs may influence the neuroendocrine control of appetite, satiety and food preference. Bisphenols, including BPA, have been detected in post-mortem human hypothalamic and white matter brain material [108]. In this study, the majority of bisphenol concentrations were associated with obesity. The accumulation of bisphenols in white-matter-enriched brain tissue may underscore their ability to cross the blood–brain barrier [108].
Exposure to BPA in utero and in adulthood may cause the arcuate nucleus of the hypothalamus to generate elevated levels of orexigenic neuropeptides such as neuropeptide Y (NPY) and Agouti-related peptide (AgRP), resulting in an increase in appetite [109,110,111].
Interestingly, NPY can also enhance lipogenic enzymes in fat tissues, leading to the development of obesity [112]. On the contrary, proopiomelanocortin (POMC), which generates the anorexigenic neuropeptide α-melanocyte-stimulating hormone (α-MSH), is diminished [109]. Moreover, exposure to BPA and its analog tetrabromobisphenol A (TBBPA) at environmental levels resulted in hyperphagia and obesity in adult male zebrafish by stimulating the endocannabinoid receptor type 1 (CB1), which plays an important role in the gut–brain axis control of appetite and satiety [113]. Phthalates such as DEHP may upregulate the expression levels of NPY in male rats [114]. Finally, in a prospective epidemiological study, BPA was strongly linked to the orexigenic gut hormones ghrelin and leptin particularly in women, suggesting that BPA could be implicated in the hormonal control of appetite and satiety [115].

5.4. Effects on Proinflammatory Pathways and Oxidative Stress

BPA and phthalates may induce the expression and secretion of certain proinflammatory cytokines such as interleukin (IL)-6 and tumor necrosis factor (TNF)-α and downregulate the expression of anti-inflammatory adipokines such as adiponectin, leading to a sustained low-grade inflammation both locally and systematically, adipose dysfunction and insulin resistance [84,116,117]. Nevertheless, the mechanisms implicated in metabolic inflammation are still under investigation. The majority of in vitro studies in 3T3-L1 cells and in adipocytes derived from subcutaneous tissues have shown that exposure to 1 nM BPA induced an increase in IL-6 and interferon (IFN)-γ while prolonged exposure upregulated the mRNA levels of IL-6, IL-1β, IFN-γ and monocyte chemoattractant protein-1 (MCP1), which were reversed after BPA removal [84,118]. These results were confirmed in some animal studies; however, the effect of BPA depends on caloric intake, diet composition and gender [84]. Furthermore, BPA and its substitutes could promote macrophage polarization toward the proinflammatory M1 subtype, which may be reversed by ERα inhibition in the case of BPF [119]. BPA and certain phthalates may also enhance the generation of reactive oxygen species (ROS), which is crucial in cellular stress and the progression of inflammation [120,121]. Finally, in many epidemiological studies, urinary and plasma BPA correlated positively with somatometric indices, such as BMI and waist circumference (WC); proinflammatory biomarkers, including IL-17, a cytokine implicated in chronic inflammation; and glycemic indices (insulin and glucose) [84,122,123,124].

5.5. Effects on Gut Microbiome

BPA and phthalates that accumulate in the gut may influence the gut microbiome by altering its composition, resulting in gut dysbiosis, immune system imbalance and altered glucose metabolism [60]. Exposure to BPA during gestation may promote obesity phenotypes in murine models by altering gut microbiota [109]. BPA exposure decreases the richness and diversity of the intestinal microbiota and beneficial small-chain fatty acid (SCFAs), induces leaky gut and elevates systemic lipopolysaccharide (LPS), resulting in chronic subclinical inflammation and altered lipid and glucose homeostasis [125,126]. In a mouse multi-omics study, prenatal exposure to low-dose BPA altered the expression of hepatic genes implicated in oxidative phosphorylation, fatty acid metabolism and PPAR signaling and impacted intestinal bacterial diversity in an age- and gender-dependent fashion [127]. Exposure to phthalate esters such as DEHP elevated cardiovascular risk in obese mice by dysregulating the arachidonic acid metabolism of intestinal flora [128], while prolonged exposure increased weight and liver lipogenesis in mice by inducing the uptake of fatty acids and dysregulating the metabolism of phospholipids and choline [129].

5.6. Effects on Thermogenic Adipose Tissue

Certain natural and synthetic EDCs, including BPA and phthalates, may induce dysfunction not only in white adipocytes but also in thermogenic brown and beige adipocytes, which regulate thermogenesis, fat metabolism and energy balance [130]. Brown and beige adipocytes share common features that comprise the existence of multiple dense mitochondria, lipid droplets and the expression of uncoupling protein 1 (UCP1) which permits the production of heat at the expense of ATP production through oxidative phosphorylation [131]. Therefore, these cells can prevent hypothermia without shivering and metabolize excess fat via UCP1.
Although data on the effects of EDCs on thermogenic adipose tissue are scant, growing evidence suggests that bisphenols and phthalates may target and regulate the activity of thermogenic adipocytes. BPA and DEHP preferentially accumulate in mice thermogenic adipose tissues at greater levels than those determined in serum, white adipose tissue (WAT) and the brain [132,133]. Nevertheless, their effects depend on the animal used, gender, life stage, type of exposure, and concentrations of specific chemical compounds. For example, there is a sexual dimorphism observed after gestational exposure to BPA with increased brown adipose tissue (BAT) depots and UCP1 in female offspring and decreased BAT activity and adipogenesis in male offspring [134]. The same applies to developmental exposure to DEHP where a decrease in BAT activity accompanied by hypothermia, hyperphagia and weight gain was observed in male mice [135] and hyperplastic brown fat was seen in female offspring [133]. These actions may be attributed to agonistic or antagonistic effects of EDCs on ERs, the impaired thyroid signaling that impacts the beiging of WAT and the macrophage subpopulation located in the WAT [130,136]. Finally, differential effects of bisphenols and phthalates are observed in macrophage polarization promoting an environment permissive for the beiging of WAT [130]. For example, BPA and BPF induce classical macrophage M1 polarization with increased secretion of proinflammatory cytokines that affect adipocyte beiging while DEHP may promote macrophage M2 polarization facilitating beiging [130]. More studies are needed to delineate the effects of EDCs on thermogenic adipose tissues, which are critical in the regulation of energy balance.

6. Evidence from Mechanistic Studies Linking BPA and Phthalates to Obesity

Existing experimental evidence has shown that BPA and phthalates may exert their obesogenic properties by promoting the differentiation of pre-adipocytes or MSC stems into mature adipocytes (Table 1). In addition, these endocrine disruptors may predispose individuals to obesity through metabolic alterations in mature adipocytes. Prolonged BPA exposure at low, environmentally relevant, concentrations may induce pre-adipocyte proliferation and differentiation due to the increased expression of adipogenic transcriptional factors, including PPARγ. It is worth noting that these effects of BPA exposure are relevant to the environmental concentrations, resulting in adipocyte metabolic dysfunction and proinflammatory cytokine production [137]. Moreover, MEHP exposure at the dose of 10 µM activates undifferentiated pre-adipocytes, leading to de novo lipogenesis, through the augmented expression of glucose transporter (GLUT)1, GLUT4 and S100B [138]. Interestingly, BPA and DEHP may promote the differentiation of murine MSCs into adipocytes at different concentrations and stages of cell determination and differentiation [139]. Furthermore, BBP potentially impairs pre-adipocyte differentiation through epigenetic changes of genes implicated in adipogenesis since BBP exposure augments miR-34a-5p expression and attenuates the expression level of its target genes [104].
On the other hand, low BPA doses may provoke adipose tissue dysfunction, without markedly interfering with adipocyte differentiation or the activation of adipogenic factors. In these cases, adipogenesis is suggested to be mediated by inflammatory and insulin signaling pathways. Alternatively, the activation of specific cellular receptors has been proposed. Thus, BPA and 2,4-dichlorophenol (DCP) may promote pre-adipocyte differentiation in the 3T3-L1 cells, through activating GRs, without increasing PPARγ expression [85]. In addition, the increased expression of insulin-like growth factor 1 (IGF-1) under BPA exposure may enhance adipogenesis via ER stimulation [140]. In mature adipocytes, DINP and DPHP exposure may lead to insulin resistance and inflammation as a result of oxidative stress, mitochondrial dysfunction and augmented adipokine secretion [141]. In turn, BPA may diminish insulin-stimulated glucose utilization. Moreover, exposure to BPA may stimulate intracellular signaling pathways that promote apoptosis (c-Jun N-terminal kinases/JNKs) or activate transcriptional factors that induce the expression of proinflammatory cytokines (NF-kB) [118]. Interestingly, daily BPA exposure has been reported to increase insulin secretion in response to glucose stimulation in pancreatic cells via ERs, in contrast to acute (60 min) exposure, which has no impact [142]. De Filippis et al. have shown that BPA exposure had no impact on the expression of PPARγ, fatty-acid-binding protein 4 (FABP4) and fatty acid synthase (FASN) and pre-adipocyte differentiation, highlighting the implication of the increased expression of IL-6 and TNF-α [143]. Notably, BPA may induce adipogenesis through mTOR signaling and TR/RXR stimulation [144].
Multiple in vitro studies have indicated that exposure to BPA and phthalates may disrupt cellular lipid homeostasis through a variety of underlying mechanisms, including the regulation of specific signaling pathways. Thus, MEHP may induce lipid accumulation by inhibiting the Janus kinase (JAK)2/signal transducer and activator of transcription (STAT)5 signaling [145]. In addition, in vitro evidence in hepatic cellular models supports that BPA dysregulates lipid homeostasis through the enhanced expression of apolipoprotein A4 (APOA4) or via the activation of the CB1 receptor [146,147]. BPA in the lowest doses may promote the expression of the mRNA level of the 11β-HSD1 gene, resulting in adipogenesis and lipid synthesis [148]. In addition, BPA seems to be involved in the upregulation of several genes related to lipid metabolism, like lipoprotein lipase (LPL) and SREBF1. As shown by Grasselli et al., non-cytotoxic levels of BPA reduced the expression of genes involved in lipid oxidation but had no impact on the expression of lipogenic genes [149]. Furthermore, BPA and DEHP exposure may alter the expression of lipid metabolism markers, consequently resulting in steatosis in RTL-W1 cells [150]. Five years ago, Schaedlich et al. reported that DEHP exposure attenuates triacylglycerol (TG) accumulation in lipid droplets of human Simpson–Golabi–Behmel syndrome pre-adipocyte cells (SGBSs), in which reduced adiponectin levels and increased leptin levels were present [151].
In conclusion, EDCs may exert in vitro obesogenic effects through a plethora of mechanisms involving the disruption of adipocyte differentiation and inflammation as well as changes in insulin metabolism and sensitivity. Although they are of clinical importance, further studies reflecting obesogenic features in real life are needed to elucidate their potential association with obesity.
Table 1. Major in vitro studies portraying associations between BPA and phthalates, and obesity.
Table 1. Major in vitro studies portraying associations between BPA and phthalates, and obesity.
Authors, YearType of Cell CultureMain FindingsRemarks
Bisphenol A and obesity
Riu et al., 2011 [152]NIH3T3-L1 cell line
(pre-adipocytes)
1. ↑ adipogenesis
2. ↑ lipid accumulation
3. ↑ mRNA level of PPARγ
4. ↑ PPARγ activity
1. Animal in vitro model
2. ED: TBBPA
3. Obesogenic effects at 10 µM
Valentino et al., 2013 [118]Primary hADSCs1. (-) mRNA level of PPARγ, GLUT4
2. ↓ of glucose utilization
3. ↓ tyrosine phosphorylation of
insulin receptor (IR)
3. ↓ of PKB/Akt phosphorylation
4. ↑ of IL-6, IFN-γ
5. ↑ of JAK/STAT, JNK
6. ↑ activity NF-kB pathway
1. Human in vitro model
2. ED: BPA
3. Biological effects at 1 nM
Bastos Sales et al., 2013 [102]Murine N2A, human SK-N-AS neuroblastoma cells and murine pre-adipocyte fibroblasts (3T3-L1)1. Modest ↓ in global DNA methylation in murine N2A cells
2. No changes in global DNA methylation in human SK-N-AS cells.
3. ↑ adipocyte differentiation in murine 3T3-L1 pre-adipocytes
1. Animal and human in vitro model
2. ED: BPA and a range of several EDCs not belonging to bisphenols
3. Biological effects at ≥ 10 μΜ
Menale et al., 2015 [137]Primary pre-adipocytes 1. ↑ adipogenesis
2. ↑ lipid accumulation
3. ↑ mRNA level of ERα (10 nM, 100 nM)
4. (-) mRNA level of ERβ
5. ↑ production of IL1B, IL18, CCL20 (10 nM)
1. Human in vitro model
2. ED: BPA
3. Obesogenic effects at 1 nM, 10 nM, 100 nM
Ariemma et al., 2016 [92]3T3-L1
Pre-adipocytes
1. Undifferentiated cells:
- ↑ proliferation
- ↑ differentiation
- ↑ expression of PPARγ, C/EBPα and FABP4/AP2
2. Mature adipocytes:
- Hypertrophy
- ↑ lipid accumulation
- ↑ mRNA of leptin, IL6, IFNγ
- ↓ glucose utilization
1. Animal in vitro model
2. ED: BPA
3. Obesogenic effects at 1 nM
Longo et al., 2020 [103]3T3L1 and NIH3T3 (committed and uncommitted pre-adipocytes, respectively) - ↓ DNA methylation at PPARγ promoter, without affecting mRNA expression in pre-adipocytes
- Transient ↑ in PPARγ expression and lipid accumulation at D4 of differentiation in 3T3L1 cells
- Ending BPA exposure restores the PPARγ promoter methylation and inflammatory profile of 3T3L1 cells.
- Expression of PPARγ is barely detectable and its promoter is completely methylated in NIH3T3 cells
- ↑ PPARγ expression is more evident both in pre-adipocytes and during the adipocyte differentiation
1. Animal in vitro model
2. ED: BPA
3. Biological effects at low doses: 1 nM
Cohen et al., 2021 [153]Primary hADSCs1. ↑ adipogenesis and lipid production at 0.1 nM
2. ↓ adipogenesis and lipid production at 10 nM
1. Human in vitro model
2. ED: BPA
3. Biological effects at 0.1 nM, 10 nM
Yamasaki et al., 2021 [154]ST-13 cell line (pre-adipocytes) - Undifferentiated cells:
1. (-) lipid accumulation
2. (-) mRNA level of PPARγ
3. ↑ mRNA level of AACS, PLIN1, FAS, CIDEA, LSD-1
- Mature adipocytes:
1. (-) lipid accumulation
2. (-) mRNA level of AACS, SCOT
1. Animal in vitro model
2. ED: TBBPA
3. Obesogenic effects at 0.5 µM, 1 µM
Schaffert et al., 2021 [121]SGBSs (pre-adipocytes) 1. ↑ binding to PPARγ (50 µM)
2. (-) PPARγ activity (10 nM, 100 nM, 1 µM, 10 µM)
3. ↓ lipid accumulation (10 nM, 100 nM, 1 µM, 10 µM)
4. ↑ leptin (10 nM)
5. ↓ cellular ROS level (10 nM, 100 nM, 1 µM, 10 µM)
6. ↓ insulin sensitivity (1 µM)
1. Human in vitro model
2. ED: BPA
3. Obesogenic effects at 10 nM, 100 nM, 1 µM, 10 µM, 50 µM
Marqueno et al., 2021 [155]ZFL cell line (primary mouse hepatocytes) 1. ↑ lipid accumulation (5 µM, 50 µM)
2. ↑ ROS generation (20 µM, 50 µM, 70 µM, 100 µM, 150 µM, 200 µM)
1. Animal in vitro model
2. ED: BPA
3. Biological effects at 5 µM, 20 µM, 50 µM, 70 µM, 100 µM, 150 µM, 200 µM
Lee et al., 2022 [156]Huh-7 cell line (primary hepatocytes) 1. ↓ cell viability (200 µM, 400 µM)
2. ↑ lipid accumulation (10 µM, 50 µM, 100 µM, 200 µM)
3. Fatty acid uptake ↑ (10 µM, 50 µM, 100 µM)
4. ↑ intracellular ROS formation (10 µM, 50 µM, 100 µM, 200 µM)
1. Human in vitro model
2. ED: BPA
3. Biological effects at 10 µM, 50 µM, 100 µM, 200 µM, 400 µM
Phthalates and obesity
Sargis et al., 2010 [85]3T3-L1 cell line (pre-adipocytes) 1. ↑ adipogenic differentiation
2. ↑ lipid accumulation (100 nM)
3. ↑ PPARγ and glucocorticoid-like activity (1 µM)
4. ↑ adiponectin and protein expression of IR-β (1 µM–100 pM)
1. Animal in vitro model
2. Pthalate: DCHP
3. Obesogenic effects at 100 pM, 1 nM, 10 nM, 100 nM, 1 µM
Dimastrogiovanni et al., 2015 [150]RTL-W1 cell line
(hepatocytes)
1. ↑ lipid accumulation
2. ↓ alteration of membrane lipids
3. ↓ mRNA level of CD36, FAS, LPL
1. Animal in vitro model
2. Pthalate: DEHP
3. Biological effects at 5 μΜ
Zhang et al., 2017 [105]C3H10T1/2 cell line (MSCs)1. ↑ adipogenesis
2. ↑ mRNA level of AP2, PPARγ
3. ↑ lipid accumulation
4. ↑ protein level of FOXO1
5. ↑ acetylation of FOXO1,
β-catenin
6. ↓ protein level of SIRT1, SIRT3
1. Animal in vitro model
2. Pthalate: BBP
3. Biological effects at 50 μΜ
Schaedlich et al., 2018 [151]SGBSs (pre-adipocytes) 1. ↓ TGsaccumulation
2. ↓ adiponectin production
3. ↓ protein level of PPARα, PPARγ
4. ↓ phosphorylation of ERK1, ERK2
5. ↑ lipolysis
6. ↑ ROS formation
1. Human in vitro model
2. Pthalate: DEHP
3. Obesogenic effects at 50 µg/mL
Zhang et al., 2019 [145]BRL-3A cell line
(hepatocytes)
1. ↑ lipid accumulation (100 µM, 200 µM)
2. ↑ mRNA level of FAS, PDK4, AP2 (10 µM, 50 µM, 100 µM, 200 µM)
3. ↑ mRNA level of PPARγ (50 µM, 100 µM, 200 µM)
4. ↓ JAK2/STAT5 signaling
5. ↓ level of indicators of oxidative stress: SOD ↓, MDA ↑ (10 µM, 50 µM, 100 µM, 200 µM)
1. Animal in vitro model
2. Pthalate: MEHP
3. Biological effects at 10 µM, 50 µM, 100 µM, 200 µM
Perez-Albaladejo et al., 2021 [157]PLHC-1 cell line (hepatocytes) - DBP:
1. ↑ TG accumulation (20 µM)
2. ↑ ROS formation (5 µM, 20 µM, 50 µM, 100 µM)
- DEHP:
1. ↑ TG accumulation (5 µM, 10 µM)
2. ↑ ROS formation (100 µM)
1. Animal in vitro model
2. Phthalates: DBP and DEHP
3. Biological effects at
- DBP: 5 µM, 20 µM, 50 µM, 100 µM
- DEHP: 5 µM, 10 µM, 100 µM
Meruvu et al., 2021 [104]3T3-L1 cells- ↑ miR-34a-5p expression
- ↑ adipogenesis
- ↓ Nampt, Sirt1 and Sirt3 gene expression levels; ↓ Nampt protein
- ↓ adipogenesis, ↑ Nampt protein and NAD+ after miR-34a-5p knockdown in the presence of BBP
1. Animal in vitro model
2. Phthalate: BBP
2. Biological effects at various doses of BBP without exogenous adipogenic stimuli
Al-Abdulla et al., 2022 [158]MIN-6 cell line (pancreatic cells) 1. ↓ viability of cells after 24 exposure at 1 μΜ
2. ↑ mRNA level of SUR1, GLUT2 at 10 μΜ
3. ↓ GSIS (20 μΜ glucose)
4. ↓ insulin content at 1 μΜ
1. Animal in vitro model
2. Pthalate: DEHP
3. Dose: 100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM
Schaffert et al., 2022 [141]SGBSs
(pre-adipocytes)
1. DINP:
- ↑ binding to PPARγ
- (-) PPARγ activation
- (-) lipid accumulation
- ↑ adipsin (10 µM)
- Mature adipocytes:
* 10 µM: ↑ MCP-1, LAP3, GPX1
* 10 nM: ↑ GPX8, GSR
* 10 nM, 10 µM: ↑ LEP, GPX4
* 10 nM, 10 µM: ↓ adiponectin
2. DPHP:
- ↑ binding to PPARγ
- (-) PPARγ activation
- Undifferentiated cells:
* (-) lipid accumulation
* ↓ MCP-1 (10 nM, 10 µM)
- Mature adipocytes:
* ↓ lipid accumulation (10 µM, 25 µM, 50 µM, 100 µM)
* 10 µM: ↑ LEP, MCP-1, LAP-3, GPX4, GPX8, adipsin
* 10 nM: ↑ GSR
* 10 nM, 10 µM: ↑ GPX1, GSTO1
* 10 nM, 10 µM: ↓ adiponectin
3. MHINP:
- ↑ binding to PPARγ (100 µM, 200 µM, 400 µM)
- ↑ PPARγ activation (1 µM)
- Undifferentiated cells:
* ↑ pre-adipocyte differentiation, lipid accumulation (10 µM, 25 µM, 50 µM, 100 µM)
* 10 µM: ↑ LEP, PLIN1, GPD1, FASN, FABP4, FABP5
* 10 nM: ↓ MCP-1
* 10 nM, 10 µM: ↑ adipsin
- Mature adipocytes:
* 1 µM: ↑ lipid accumulation
* 10 µM: ↑ LAP3, adipsin
* 10 nM: ↑ GSR, GPX8
* 10 nM, 10 µM: ↑LEP, MCP-1, GPX1, GPX4, GSTO1
* 10 nM, 10 µM: ↓ adiponectin
4. OH-MPHP:
- ↑ binding to PPARγ
- ↑ PPARγ activation
- Undifferentiated cells:
* ↑ pre-adipocyte differentiation, lipid accumulation (10 µM, 25 µM, 50 µM)
* ↑ LEP, GPD1, FASN, FABP4, FABP5 (10 µM)
- Mature adipocytes:
* 10 µM: ↑ LAP3, GPX1, GPX4, GPX8, adipsin
* 10 nM, 10 µM: ↑ LEP, GSR, MCP-1, GSTO1
* 10 nM, 10 µM: ↓ adiponectin
* 10 nM, 10 µM, 25 µM, 50 µM, 100 µM: ↓ lipid accumulation
1. Human in vitro model
2. Phthalates: DINP, DPHP, MHINP, OH-MPHP
2. Obesogenic effects at
- DINP: 10 nM, 10 µM,
- DPHP: 10 nM, 10 µM, 25 µM, 50 µM, 100 µM
- MHINP: 10 nM, 10 µM, 25 µM, 50 µM, 100 µM, 200 µM, 400 µM
- OH-MPHP: 10 nM, 10 µM, 25 µM, 50 µM, 100 µM, 200 µM, 400 µM
Abbreviations: AACS: acetoacetyl-CoA synthetase; AP2: adipocyte protein 2; BPA: bisphenol A; BBP: benzyl butyl phthalate; CCL20: chemokine (C-C motif) ligand 20; C/EBPα: CCAAT/enhancer-binding protein α; CD36: fatty acid translocase; CIDEA: cell-death-inducing DNA fragmentation factor-alpha-like effector A; DCHP: bis(2-propylheptyl) phthalate; DBP: dibutyl phthalate; DEHP: di(2-ethylhexyl) phthalate; DINP: diisononyl phthalate; DPHP: bis(2-propylheptyl) phthalate; ED: endocrine disruptor; ERα: estrogen receptor α; ERβ: estrogen receptor β; ERK1/2: extracellular-signal-regulated protein kinase 1/2; FABP4: fatty-acid-binding protein 4; FABP5: fatty-acid-binding protein 5; FABP4/AP2: fatty-acid-binding protein 4/adipocyte protein 2; FASN: fatty acid synthase; FOXO1: forkhead box protein O1; GLUT2: glucose transporter type 2; GLUT4: glucose transporter type 4; GPD1: glycerol-3-phosphate-dehydrogenase; GPX1: glutathione peroxidase 1; GPX4: glutathione peroxidase 4; GPX8: glutathione peroxidase 8; GSIS: glucose-stimulated insulin secretion; GSR: glutathione-disulfide reductase; GSTO1: glutathione S-transferase omega-1; IFN-γ: interferon-γ; IL: interleukin; IR-β: insulin receptor subunit β; JAK/STAT: Janus kinase/signal transducer and activator of transcription; JNK: c-Jun N-terminal kinase;LAP3: leucine aminopeptidase 3; LEP: leptin; LPL: lipoprotein lipase; LSD-1: lysine-specific demethylase-1; MCP-1: monocyte chemoattractant protein-1; MDA: malondialdehyde; MEHP: mono-2-ethylhexyl phthalate; MHINP: monohydroxy isononyl phthalate; N2A: Neuro-2A cells; NAD: nicotinamide adenine dinucleotide; Nampt: nicotinamide phosphoribosyltransferase; NF-kB: nuclear factor kappa-light-chain-enhancer of activated B cells; OH-MPHP: 6-hydroxy monopropylheptyl phthalate; PDK4: pyruvate dehydrogenase kinase 4; PPARα: peroxisome-proliferator-activated receptor α; PPARγ: peroxisome-proliferator-activated receptor γ; PKB/Akt: protein kinase B/AKT; PLIN1: perilipin-1; ROS: reactive oxygen species; SCOT: succinyl-CoA-3-oxoacid CoA-transferase; SIRT1: sirtuin 1; SIRT3: sirtuin 3; SOD: superoxide dismutase; SUR1: sulfonylurea receptor 1; TBBPA: tetrabromobisphenol A; TGs: triglycerides. ↑ increase, ↓ decrease.

7. Evidence from Animal Studies Linking BPA and Phthalates to Obesity

A significant number of in vivo studies in animals propose an association of BPA exposure with adipogenesis (Table 2). In fact, BPA exposure during pregnancy and lactation may provoke weight gain in the postnatal period, as well as an increase in the adipose tissue mass in the offspring. Interestingly, these features occur at low maternal BPA concentrations in comparison to the environmental levels of BPA. According to Wei et al., these unfavorable outcomes may become more evident when animals are exposed to a high-fat diet, and this is thought to be attributed to impaired glucose tolerance [159]. Surprisingly, elevated BPA levels in plasma do not seem to lead to maternal weight gain and increased body fat, due to the augmented secretion of α-fetoprotein during gestation. BPA’s binding to α-fetoprotein may abrogate its protective effects on the developing fetus, which are normally achieved through its binding to estrogen [160]. On the other hand, several in vivo studies have focused upon the potential adverse effects of different phthalates regarding obesity, implicating a wide range of proposed mechanisms. Most of these obesogenic features are investigated using rodents or zebrafish animal models and are thought to be dose-dependent or gender-related, respectively. Notably, effects on body weight and visceral adipose tissue mass have been reported at environmentally relevant doses, suggesting that phthalate exposure may be associated with obesity risk in humans.
Obesogenic features of BPA are correlated with its ability to exhibit hormone-like properties by interfering with specific receptors. In particular, BPA resembles the effects of estrogen, via binding to ERα and Erβ receptors. In such cases, attenuated adiponectin secretion, as well as increased adipocyte differentiation and lipid accumulation, may be seen [109]. In addition, BPA exposure may disable the beneficial impact of some adipokines in lipolysis, resulting in impaired lipid metabolism and difficulty in maintaining normal body mass. Existing in vivo experimental evidence has shown the adipogenic effects of BPA exposure due to its ability to interact with PPAR. BPA modulates PPAR-γ signaling, resulting in augmented expression of genes that induce adiposity [161]. On the other hand, BPA seems to inhibit the expression of PPAR-α, thus promoting liver TG deposition via alterations in lipid oxidation [113]. PPAR-γ activation is suggested to play an essential role in adipogenesis induced by phthalate exposure as well. According to Hao et al., DEHP injection in C57BL/6J mice increased adipose tissue weight due to the enhanced expression of adipogenic transcriptional factors PPAR-γ, C/EBP and SREBF1 [162]. In addition, DEHP exposure in female mice, for a period of 10 weeks, is associated with weight gain and increased adipose tissue mass, whereas insulin sensitivity impairment and maladaptive adipose tissue function were observed. Changes in circulating and tissue adiponectin, as well as the augmented expression of ERs and the activation of PPAR-γ, may be involved [163]. PPAR-γ activation seems to be also involved in adipogenesis, following MEHP exposure, resulting in the differentiation of pre-adipocyte-like adipocytes into lipid-laden and insulin-responsive adipocytes [164]. Interestingly, Zhuang et al. pointed out the role of the transforming growth factor (TGF)-β signaling pathway in adipocyte differentiation, via the stimulation of the adipogenic factors PPAR-γ and CEBPα, in contrast to the estrogen signaling pathway, which had no effect [165].
Recently, Tian et al. reported that the activation of CB1 may be a potential sequela of BPA exposure, resulting in increased appetite and ultimately obesity. The CB1 receptor is expressed in a variety of tissues, including nervous system and fat tissues, and may have an essential role in regulating food intake and energy storage [113]. Furthermore, BPA exerts its harmful effects on the central nervous system through the stimulation of orexigenic hypothalamic neuropeptides, AgRP and NPY [166]. Similar data suggest that DEHP, following 5 weeks of exposure, favors food intake and increased body weight in male C3H/He mice, probably due to the augmented expression of hypothalamic neuropeptides involved in the appetite regulation and the synergistic effects of hypothyroidism and hypothalamic leptin resistance [135]. Evidence from animal studies has underscored that exposure to phthalates may lead to alterations in lipid metabolism, resulting in dysregulation of energy homeostasis (Table 2). In zebrafish, DEHP at low doses alters the expression of liver genes associated with fatty acid metabolism, consequently leading to non-alcoholic fatty liver disease (NAFLD). Notably, these effects were absent following 17α-ethinylestradiol exposure, which typically lacks estrogen features [167]. On the other hand, high concentrations of DEHP may weaken diet-related obesity in mice, potentially via PPAR-α-dependent stimulation of hepatic fatty acid catabolism. Notably, this effect is observed in mice, but not in humans [97]. In rats, DEHP at the dose of 5–200 mg/kg daily alters the lipid profile, resulting in reduced concentrations of high-density lipoproteins (HDLs) and increased levels of total cholesterol and triglycerides [168]. Furthermore, MEHP has been proposed to boost the pathological progression of liver steatosis in a zebrafish animal model, upon co-administration with ethanol. In such cases, DNA damage and apoptosis seem to be mediated via CYP4A and alcohol dehydrogenase (ADH) involvement [169].
Exposure to BPA may also have an impact on the intestinal microbiota due to its ability to alter LPS and gut SCFA levels. Consequently, impaired lipid homeostasis and chronic low-grade activation of the inflammatory cascade may develop [109]. In accordance with this observation, long-term DEHP exposure may provoke gastrointestinal dysbiosis, thus increasing fat storage in zebrafish, as a result of the disrupted expression of genes associated with lipid metabolism [170]. Early-life BPA exposure could account for transgenerational epigenetic alterations, which may have an impact on obesity risk, across several generations. These effects are believed to occur due to specific changes in the DNA methylation pattern of genes modulating metabolic pathways. Susiarjo et al. showed that maternal exposure to BPA in C57BL/6 mice may induce metabolic abnormalities due to the overexpression of the imprinted insulin-like growth factor 2 (IGF-2) gene and increased DNA methylation at the IGF-2 locus [171]. Similar to BPA, phthalates also seem to facilitate epigenetic transgenerational effects on obesity. Prenatal phthalate exposure may modify DNA methylation at loci near genes, resulting in alterations in metabolic hormone signaling pathways. Specifically, Bis(2-ethyhexyl) tetrabromophthalate (TBPH) and its metabolite TBMEHP may induce demethylation of the PPAR-γ promoter DNA, leading to changes in lipid metabolism in early larval stages of zebrafish. As a result, lipid lipolysis and energy homeostasis may be disrupted [172].
In agreement with in vitro experimental data, potential in vivo obesogenic effects of BPA and phthalates seem to be mediated by mechanisms involving the activation of adipogenic transcriptional factors, stimulation of specific receptors and enhancement of the inflammatory cascade. In addition, disruption of gut dysbiosis, as well as transcriptional epigenetic alterations associated with DNA methylation modification, may also be involved.
Table 2. Major animal studies showing associations between BPA and phthalates and obesity.
Table 2. Major animal studies showing associations between BPA and phthalates and obesity.
Author, YearType of Animal UsedMain FindingsRemarks
Bisphenol A and obesity
Pu et al., 2017 [161]Primiparous female sheep1. ↑ differentiation rate in adipocytes
2. ↑ mRNA expression of PPARγ in fetal adipose tissue
3. ↑ expression of FABP4, GLUT4 and SOX6 in the offspring
4. ↑ gene expression of GR, ESR1, ESR2 and ERRα
1. Type of exposure: sc
2. Exposure duration: 147 days
3. Daily BPA dose: 0.5 mg/kg
Desai et al., 2018 [160]12-week-old female Sprague–Dawley rats1. ↑ body weight
2. ↑ mass of adipose tissue
3. Hypertophic adipocytes in male offspring
4. ↑ expression of PPARγ
5. ↑ TNF-α and CD68 in adipose tissue
1. Findings occurred in the offspring
2. Type of exposure: drinking water
3. Exposure duration: 2 weeks before mating up to weaning
4. Daily BPA dose: 5 mg/L
Stoker et al., 2019 [173]90-day-old female Wistar rats1. ↑ food intake
2. ↑ epididymal and perirenal fat deposition
3. ↑ fasting serum glucose and leptin in male mice
4. ↑ expression of hypothalamic orexigenic neuropeptides in male mice
1. Findings occurred in the offspring
2. Type of exposure: drinking water
3. Exposure duration: pregnancy day 9 to weaning
4. Daily BPA dose: 50 µg/kg
Lin et al., 2019 [174]3-week-old male Wistar rats1. ↑ fat deposition (visceral, liver)
2. ↑ TCHOL, LDL, TGs
3. ↓ HDL
4. ↑ TNF-α, IL-17
5. ↑ mRNA of SREBP1 and ACC1
6. ↑ TLR4 and NF-κB in the liver
7. ↓ HSL, ERα and ZAG in the liver
1. Type of exposure: drinking water
2. Exposure duration: 8 weeks
3. Daily BPA dose: 1 µg/mL
Tian et al., 2021 [113]5-month-old wild-type adult male Danio rerio1. ↑ weight gain, length, food intake
2. ↑ lipid accumulation in liver
3. Microvesicular fatty changes, hepatocyte ballooning, infiltration with inflammatory cells
4. ↑ of CB1
5. ↑ of insulin signaling pathways
6. ↓ expression of PPARα in adipose tissue and liver
7. ↓ gpr55
1. Type of exposure: water in static system
2. Exposure duration: 28 days
3. Daily BPA dose: 20, 100 and 500 μg/L
Shih et al., 2021 [175]15-week-old female Sprague–Dawley rats1. ↑ abdominal lipid weight up to 77% in female offspring
2. ↑ TCHOL, LDL, TGs
3. ↓ HDL
4. ↑ leptin
5. ↑ of Prevotella, C. perfringens, C. ruminantius in feces
1. Type of exposure: oral gavage
2. Exposure duration: 6th-36th day after pregnancy
3. Daily BPA dose: 50 µg/kg
Zhuang et al., 2023 [165]7-week-old male and female ICR mice1. ↑ weight gain in the offspring
2. ↑ size of adipocytes
3. ↓ insulin sensitivity
4. No obesogenic effects via estrogen signaling pathway
5. Obesogenic effects via TGF-β signaling pathway
1. Type of exposure: drinking water
2. Exposure duration: 7 days treatment up to delivery
3. Daily BPA dose: 0.5 μg/kg
Phthalates and obesity
Hao et al., 2013 [162]C57BL/6J mice1. ↑ expression of PPARγ, aP2, LPL and FAS
2. ↑ expression of C/EBP, Srebf1
3. ↑ glucose, TCHOL, TGs in serum
4. Obese phenotype only at the dose of 0.25 mg/kg in female offspring
5. ↑ weight gain in male offspring
1. Type of phthalate: DEHP
2. Ip DEHP at the dose of 0.5 mg/kg in six-week-old male mice
3. Female mice:
- Type of exposure: gavage
- Exposure duration: from day 12 of
gestation until day 7 of lactation
- Daily DEHP dose: 0.05, 0.25 or 0.5 mg/kg
Klöting et al., 2015 [163]Obesity-resistant 129S6 miceIn female (but not in male) mice:
1. ↑ weight gain
2. ↑ fat mass
3. ↓ insulin tolerance
4. ↓ Pparg and adiponectin in scAT
5. ↑↑ Esr1 protein levels in SC and visceral adipose tissue
6. (-) TCHOL, TGs in serum
7. ↑ in phospholipid and carnitine
1. Type of phthalate: DEHP
2. Type of exposure: oral
3. Exposure duration: 10 weeks
4. Daily DEHP dose: 0.05 mg/kg
Lv et al., 2016 [135]Male C3H/He mice1. ↑ food intake, adipogenesis and weight gain in all exposure groups except for 0.05 mg/kg
2. Interruption in hypothalamic appetite-related neuropeptides:
- ↑ expression of AgRP in all groups
- ↑ expression of NPY at 50 and 200 mg/kg
- ↓ expression of POMC at 200 mg/kg
3. Hypothalamic leptin resistance resulting in hypothyroidism
4. ↓ WAT lipid metabolism at 0.5 mg/kg
5. ↑ WAT lipid metabolism at 50 and 200 mg/kg
1. Type of phthalate:
DEHP
2. Type of exposure: gavage
3. Exposure duration: 5 weeks
4. Daily DEHP dose: 0.05, 0.5, 5, 50 and 200 mg/kg
Zhang et al., 2020 [176]C57BL/6 J male and female mice1. Weight gain in male mice on HFD at 3 mg/kg/d
2. At the dose of 3 mg/kg/d:
- ↑ activation of SREBP1
- (-) of SREBP2, PPARγ
- ↑ expression of downstream regulatory genes of SREBP1 (FAS, ACC, HMGCR)
3. ↓ insulin tolerance in male mice between HFD + BBP3 and HFD groups
1. Type of phthalate: BBP
2. Type of exposure: oral
3. Exposure duration: 16 weeks
4. Daily BBP dose:
4 μg/kg, 169 μg/kg,
3 mg/kg, 50 mg/kg
Guo et al., 2020 [172]Zebrafish embryos 0.75 hpf - ↑ expression of PPARγ due to the following:
1. Significant regional DNA demethylation
2. Upregulation of tet1 and tet2 gene
transcription)
- As a result,
↓ TCHOL and TGs due to ↑ expression of downstream genes involved in lipid metabolism
1. Type of phthalate:
TBPH and TBMEHP
2. Type of exposure: glass Petri dish containing 100 mL of TBPH or TBMEHP
3. Exposure duration: until 72 hpf
4. Daily TBPH and TBMEHP dose: 0.2–2000 nM
Buerger et al., 2020 [177]Zebrafish (Danio rerio)GI dysbiosis in the OF + DEHP group as a result of the following:
- ↑ of Bacteroidetes
- ↑ of UFAs
- ↑ lipid metabolism
- ↓ carbohydrate metabolism
- ↓ glycerolipid metabolism
- ↓ glycerophospholipid metabolism
- ↓ carbohydrate, galactose, inositol phosphate, taurine and hypotaurine metabolism
1. Type of phthalate: DEHP
2. Type of exposure: oral
3. Exposure duration: 60 days
4. Daily DEHP dose: 3 mg/kg
Abbreviations: ACC1: acetyl-CoA carboxylase 1; AgRP: Agouti-related protein; AP2: adipocyte protein 2; BPA: bisphenol A; CB1: endocannabinoid receptor type 1; CD68: cluster of differentiation 68; C/EBP: CCAAT/enhancer-binding protein; ESR1: estrogen receptor 1; ESR2: estrogen receptor 1; ERRα: estrogen-related receptor α; FABP4: fatty-acid-binding protein 4; FAS: fatty acid synthase; GLUT4: glucose transporter type 4; GR: glucocorticoid receptor; HDL: high-density lipoprotein cholesterol; HSL: hormone-sensitive lipase; IL-17: interleukin-17; LDL: low-density lipoprotein cholesterol; LPL: lipoprotein lipase; NPY: neuropeptide Y; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; PPARγ: peroxisome-proliferator-activated receptor γ; POMC: proopiomelanocortin; SC: subcutaneous; SOX6: SRY-box transcription factor 6; SREBP1: sterol regulatory element-binding transcription factor 1; TNF-α: tumor necrosis factor-α; TCHOL: total cholesterol; TGF-β: transforming growth factor-β; TGs: triglycerides; TLR4: toll-like receptor 4; WAT: white adipose tissue; ZAG: ZAG adiponectin. ↑ increase, ↓ decrease.

8. Evidence from Human Studies Linking BPA and Phthalates to Obesity

There is an abundance of epidemiological studies assessing the effects of BPA and phthalates on human adiposity (Table 3). Urinary and, to a lesser extent, serum concentrations of BPA and different phthalate metabolites have been used as surrogates of exposure. In several studies, urinary measurements have been adjusted for creatinine concentration to account for the confounding effect of urine dilution due to the individual hydration state.

8.1. Bisphenol A and Obesity

In general, the majority of human cross-sectional epidemiological studies in adult patients demonstrate a direct association between BPA measurements in urine and generalized (indexed by the BMI) as well as abdominal (based on WC measurements) obesity [182,189,191,194,196,197]. This observation is further strengthened by the findings of longitudinal studies [190,192] demonstrating accelerated weight gain and higher incidence of central obesity among individuals with higher urinary or serum BPA concentrations [181]. The findings regarding this relationship are not equivocal, since other studies with similar methodologies have failed to demonstrate any substantial associations [183,187]; however, existing meta-analyses of the available clinical studies point towards a consistent association of higher BPA measurements with the prevalence of overweight and generalized or central obesity [178,185,186], while one study has demonstrated the presence of an inverse U-shaped dose–effect relationship between serum BPA and incident overweight/obesity in individuals with normal weight [181]. Similar associations have also been found between BPA and childhood/adolescent obesity [179,184,187,188,193,195] and are additionally supported by relevant meta-analyses [215] despite the presence of studies with neutral results [183]. Importantly, even though some disparity among studies exists in this regard, the overall association of BPA exposure with obesity risk likely affects both genders to a significant degree. The results of a recent umbrella meta-analysis have strengthened this notion; however, a considerable heterogeneity among studies in males was noted, rendering the association between BPA and obesity more convincing for females [178]. Furthermore, the available observations have been conducted in ethnically diverse populations, allowing for the generalizability of the findings. Few observational studies have pointed towards race-dependent effects of BPA on obesity risk [193,195], while in others, the risk appears to be consistent across different ethnic backgrounds [194]. The studies on the impact of BPA exposure during gestation on infantile birth weight have yielded inconclusive results, demonstrating either neutral effects, restricted fetal growth or abnormally high birth weight [216,217], with most data favoring a relationship with growth restriction, especially when exposure during early pregnancy is taken into account [218]. These findings do not contradict the bulk of evidence favoring an obesogenic effect of BPA, but rather demonstrate the overall deleterious endocrine-disrupting properties of the compound.
Apart from measures of adiposity, exposure to BPA has been associated with obesity-related metabolic perturbations and a more adverse overall metabolic profile, including insulin resistance, hyperglycemia and overt T2DM [219,220,221] independently of traditional risk factors, arterial hypertension [222] and dyslipidemia [223,224]. It is unclear whether these observations emerge as direct consequences of BPA exposure or are indirectly mediated by its putative obesogenic effects.
Due to the overall adverse safety profile of BPA, the use of other bisphenols such as BPS, BPF or BPAF in the manufacture of plastics has been increasing. However, these compounds are not free of endocrine-disrupting properties [225]. With respect to obesity, there are increasing reports of an association with BPF, PBS or PBAF exposure [180,182,187,188,189], occasionally more robust than that of BPA [180,187], although other studies have not ascertained significant effects of BPF or BPS exposure on obesity risk [183].

8.2. Phthalates and Obesity

Most available cross-sectional studies have demonstrated a direct association between one or more of the numerous measured phthalate metabolites and measures of adiposity [78,203,204,208,212,214]. Supportive evidence has also been provided by prospective observational studies, which have generally demonstrated associations between phthalate urine [192,202,205,209] or serum [213] concentrations and prospective weight or fat mass over follow-up durations as prolonged as approximately 10 years. Another prospective study ascertained a significant association of baseline urine phthalates with weight gain in the short (after 3 years) but not long term (6 years) [208], suggesting that the effect of phthalates on weight gain may be short-lived and may vary with changing levels of exposure over time. Interestingly, increased baseline phthalate concentrations were also associated with impaired weight loss during a dietary intervention, suggesting obesogenic properties [226]. It should be noted that the sum of available prospective studies has based their associations on momentary urinary measurements at baseline, which arguably do not necessarily reflect the cumulative exposure over time.
Disparities also remain as to whether the effects of specific phthalate compounds are sex- or age-group-dependent [210,213,214], while evidence also shows that higher adiposity at the time of phthalate measurement facilitates the obesogenic effects of phthalates [201]; it is unclear, however, whether the latter is an actual priming effect of already-existing obesity or if it could merely reflect a confounding effect of higher cumulative phthalate exposure and obesogenic effect also in the time period preceding the measurement. Available meta-analyses indicate that the presence of an association between phthalate exposure and obesity is likely, although the reported results are highly heterogeneous. Furthermore, the presence of sex-specific effects or publication bias in available reports cannot be excluded [200,207].
Certain reports have also demonstrated similar associations in adolescents [214] while the evidence for an association with childhood obesity are conflicting [204,214]—at least regarding the presence of effects independent of other confounding factors pertinent to diet and/or physical activity [182]. Similarly, conflicting results appear in prospective studies investigating the effects of intrauterine exposure to phthalates (based on concentrations in maternal urine during pregnancy) and infantile obesity, with some demonstrating an adverse relationship between maternal exposure and offspring weight gain [206], some demonstrating neutral [210] or even beneficial effects [210,211] and others demonstrating phthalate-compound-specific effects [209]. A recent meta-analysis indicated an overall association of prenatal exposure to the most broadly used di-(2-ethylhexyl)-phthalate with decreased BMI z-scores in infants but no impact on fat mass, suggesting that the correlation between phthalate exposure and BMI is mediated by decreased muscle growth [227].

9. Perspectives, Controversies and Challenges

Even though the bulk of evidence tends to suggest overall adverse effects of exposure to BPA and phthalates on obesity-related endpoints, the methodological limitations of available observations should also be considered. The majority of available clinical studies are of cross-sectional nature, hence not allowing for concrete conclusion, although existing prospective epidemiological analyses have generally yielded compatible results. Furthermore, practically, the majority of studies have based their results on single momentary measurements of these pollutants, particularly in urine. The use of single measurements for nonpersistent chemicals is poor [228]. In observational studies, multiple repeated specimens need to be collected for EDC determination over an extended chronic period of toxicological relevance, i.e., before and during at-risk temporal windows for a disease, and with consideration of exposure patterns [80,228]. The probability of background contamination of samples during handling, conservation or even laboratory measurement constitutes another source of potential bias [229,230,231]. However, despite these theoretical considerations, it appears that, at least in the case of BPA assays, contamination is in fact negligible in most laboratories [232], and in any case, this does not itself present a sufficient ground for discrediting the ascertained associations between BPA or phthalate measurements and adverse health outcomes, including obesity. Another factor that should be considered when examining the effects of exposure on adiposity and other metabolic readouts is that BPA and phthalate exposure also occurs during the consumption of fat- and sugar-rich, highly caloric, palatable meals of low nutritional value (“junk food”) [233]; hence, the epidemiological associations could potentially be mere reflections of an unhealthy dietary lifestyle. Certain studies have taken dietary factors into account as potential confounders [179,192,199]; nevertheless, this remains a considerable potential source of bias that should not be overlooked in epidemiological studies. Lastly, exposure to multiple chemicals can occur from single sources; therefore, a causal nature of the observed correlations cannot be readily assumed, while the estimation of combined effects of multiple exposures becomes particularly challenging for studies of observational nature.
In any case, and even though available data from human studies are not unanimous for an unequivocal obesogenic effect of BPA or phthalate exposure, the majority of available evidence, coupled with the results of preclinical and in vitro studies, collectively present a strong argument in support of this notion. Furthermore, the fact that both compound groups have been associated with an adverse profile of other relevant cardiometabolic risk determinants such as insulin resistance and T2DM [234,235,236,237,238], dyslipidemia [223,239,240] and hypertension [241,242] renders the exposure to these ubiquitous chemicals a reasonable source of public health concern.
A gradual replacement of both BPA and phthalates by alternative compounds in the production of plastic has been undertaken on a large scale in order to diminish the proven and putative adverse health effects of exposure. For example, Canada has banned the marketing of baby bottles containing BPA since 2010 [243]. This has led to the increasing use of bisphenols other than BPA, which are, however, not free of endocrine-disrupting pharmacological actions [225]. On the contrary, obesogenic properties [187,188,189] and possible links to an increased cardiovascular risk [244,245,246] have also been attributed to BPF and BPS. Collectively, all these issues mandate the displacement of the sum of bisphenol compounds and their replacement by alternative plasticizers, such as those derived from plant biomass [243]. Accordingly, viable alternatives to phthalates include adipates, diisononyl cyclohexane-1,2 dicarboxylate (DINCH) and bis-2-ethylhexyl terephthalate, which exhibit a less hazardous profile, although human exposure has been steadily increasing in the latest years [247,248].
Currently, the replacement of phthalates and bisphenols constitutes a field of active applied research. Due to the ubiquitous use of plastics and the subsequent exposure hazard to humans, there is a pressing need for meticulous testing of candidate compounds in preclinical and epidemiological studies in order to establish their safety with regard not only to potential endocrine disruption but also to overall long-term health effects.

10. Conclusions

Based on the available data from mechanistic, animal and epidemiological human studies including meta-analyses, the weight of evidence points towards the contribution of EDCs to the etiopathogenesis of obesity. Moreover, these ubiquitous chemicals such as BPA and phthalates as well as their analogs have been linked to obesity-associated disorders, namely insulin resistance, T2DM, dyslipidemia, hypertension and, similar to advanced glycation end products (AGEs), female hormone imbalances [249,250]. Undoubtedly, the type of diet, caloric intake and the lack of somatic exercise are crucial factors in the obesity pandemic; however, the decrease in exposure to obesogenic EDCs, particularly during susceptible time windows such as prenatal, neonatal, pubertal and reproductive periods, may also contribute to decreasing excess body weight and its associated implications in the population. It is important to underscore that similar to hormones, EDCs may act in several organs at very low levels promoting time- and tissue-specific effects, including metabolic, endocrine, neurological, reproductive and transgenerational implications. Besides known EDCs, there are many chemical compounds or complex mixtures presenting hormonal/growth factor signaling (metabolism, carcinogenesis)-disrupting actions, such as (anti)estrogenic or antiandrogenic actions and deregulation of the IGF-1–IGF-1 receptor axis [251,252], particularly in plastic bottled water [253,254]. It is of paramount importance that all chemical compounds should undergo testing for their endocrine-disrupting properties at low levels by using in silico, mechanistic and animal studies before marketing. International standardization is required for the biomonitoring and evaluation of EDCs to improve policymaking and understanding regarding the actual exposure of people to EDCs and their potential implications for health. Limitations of their use or substitution as well as educational programs in maternity clinics about the use of EDCs and their obesogenic and metabolic consequences represent important preventive measures. An example of substitution of EDCs in water bottles could be the use of alternative materials such as glass, aluminum or stainless steel. Although difficult to use, glass is a safe reusable bottle material for storing both food and liquids because it is chemical-free, constructed from natural products and dishwasher-safe. Before any regulatory action, subjects should be informed so that they may decrease or avoid exposure to EDCs as much as possible for themselves and their children.

Author Contributions

M.D. conceived the idea of the review, organized its plan and reviewed the study; M.D., D.K. and D.T. wrote major parts of the manuscript; N.G.V., I.K., D.K. and D.T. performed literature search and prepared the tables; S.P. made the figures and prepared all references; M.D. and A.G.P. reviewed, edited and supervised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ADH: alcohol dehydrogenase; AGEs: advanced glycation end products; AgRP: Agouti-related peptide; AhR: aryl hydrocarbon receptor; APOA4: apolipoprotein A4; AR: androgen receptor; BAT: brown adipose tissue; BF%: body fat percentage; BMI: body mass index; BPA: bisphenol A; BPB: bisphenol B; BPC: bisphenol C; BPE: bisphenol E; BPAF: bisphenol AF; BPP: bisphenol P; BPZ: bisphenol Z; CB1: endocannabinoid receptor type 1; C/EBP: CCAAT/enhancer-binding family of proteins; CPT1α: carnitine palmitoyltransferase α; CRP: C-reactive protein; DBP: dibutyl phthalate; DCP: 2,4-dichlorophenol; DEHP: di(2-ethylhexyl) phthalate; DEP: diethyl phthalate; DIBP: diisobutyl phthalate; DINP: diisononyl phthalate; DPHP: di(2-propylheptyl) phthalate; E2: 17β-estradiol; EDCs: endocrine-disrupting chemicals; EFSA: European Food Safety Authority; ER: estrogen receptor; ERR-γ: estrogen-related γ receptor; EU: European Union; FABP4: fatty-acid-binding protein 4; FASN: fatty acid synthase; FDA: Food and Drug Administration; GPR30: G-protein-coupled receptor 30; GLUT: glucose transporter; GR: glucocorticoid receptor; GWAS: genome-wide association study; HDL: high-density lipoprotein; HOMA-IR: Homeostatic Model Assessment for Insulin Resistance; HPP: 4-cumylphenol; IFN: interferon; IGF: insulin-like growth factor; IL: interleukin; IR: insulin resistance; JAK: Janus kinase; JNK: c-Jun N-terminal kinase; LBP: lipopolysaccharide-binding protein; LPL: lipoprotein lipase; LPS: lipopolysaccharide; MCP: monocyte chemoattractant protein-1; MEHP: mono-(2-ethylhexyl) phthalate; MR: mineralocorticoid receptor; mTOR: mammalian target of rapamycin; NAFLD: non-alcoholic fatty liver disease; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; NPY: neuropeptide Y; PPAR: peroxisome-proliferator-activated receptors; PR: progesterone receptor; PVC: polyvinyl chloride; ROS: reactive oxygen species; SGBSs: Simpson–Golabi–Behmel syndrome pre-adipocyte cells; SREBF1: sterol regulatory element-binding factor 1; STAT: signal transducer and activator of transcription; T2DM: diabetes mellitus type 2; TBBPA: tetrabromobisphenol A; TBPH: Bis(2-ethyhexyl) tetrabromophthalate; TDI: tolerable daily intake; TG: triacylglycerol; TGF: transforming growth factor; TNF-α: tumor necrosis factor-α; Treg: T regulatory cell; TR: thyroid hormone receptors; TSH: thyroid-stimulating hormone; VEGFA: vascular endothelial growth factor A; WAT: white adipose tissue; WHO: World Health Organization.

References

  1. World Obesity Federation. World Obesity Atlas 2023. Available online: https://data.worldobesity.org/publications/?cat=19 (accessed on 14 November 2023).
  2. Wang, Y.; Beydoun, M.A.; Min, J.; Xue, H.; Kaminsky, L.A.; Cheskin, L.J. Has the prevalence of overweight, obesity and central obesity levelled off in the United States? Trends, patterns, disparities, and future projections for the obesity epidemic. Int. J. Epidemiol. 2020, 49, 810–823. [Google Scholar] [CrossRef] [PubMed]
  3. Koliaki, C.; Dalamaga, M.; Liatis, S. Update on the Obesity Epidemic: After the Sudden Rise, Is the Upward Trajectory Beginning to Flatten? Curr. Obes. Rep. 2023, 12, 514–527. [Google Scholar] [CrossRef] [PubMed]
  4. NCD-RisC. Height and body-mass index trajectories of school-aged children and adolescents from 1985 to 2019 in 200 countries and territories: A pooled analysis of 2181 population-based studies with 65 million participants. Lancet 2020, 396, 1511–1524. [Google Scholar] [CrossRef] [PubMed]
  5. Papadavid, E.; Vlami, K.; Dalamaga, M.; Giatrakou, S.; Theodoropoulos, K.; Gyftopoulos, S.; Stavrianeas, N.; Papiris, S.; Rigopoulos, D. Sleep apnea as a comorbidity in obese psoriasis patients: A cross-sectional study. Do psoriasis characteristics and metabolic parameters play a role? J. Eur. Acad. Dermatol. Venereol. 2013, 27, 820–826. [Google Scholar] [CrossRef] [PubMed]
  6. Papavasileiou, G.; Tsilingiris, D.; Spyrou, N.; Vallianou, N.G.; Karampela, I.; Magkos, F.; Dalamaga, M. Obesity and main urologic cancers: Current systematic evidence, novel biological mechanisms, perspectives and challenges. Semin. Cancer Biol. 2023, 91, 70–98. [Google Scholar] [CrossRef] [PubMed]
  7. Hroussalas, G.; Kassi, E.; Dalamaga, M.; Delimaris, I.; Zachari, A.; Dionyssiou-Asteriou, A. Leptin, soluble leptin receptor, adiponectin and resistin in relation to OGTT in overweight/obese postmenopausal women. Maturitas 2008, 59, 339–349. [Google Scholar] [CrossRef] [PubMed]
  8. Pavlidou, A.; Dalamaga, M.; Kroupis, C.; Konstantoudakis, G.; Belimezi, M.; Athanasas, G.; Dimas, K. Survivin isoforms and clinicopathological characteristics in colorectal adenocarcinomas using real-time qPCR. World J. Gastroenterol. 2011, 17, 1614–1621. [Google Scholar] [CrossRef] [PubMed]
  9. Hassan, S.; Thacharodi, A.; Priya, A.; Meenatchi, R.; Hegde, T.A.; Thangamani, R.; Nguyen, H.T.; Pugazhendhi, A. Endocrine disruptors: Unravelling the link between chemical exposure and Women’s reproductive health. Environ. Res. 2023, 241, 117385. [Google Scholar] [CrossRef]
  10. Emfietzoglou, R.; Spyrou, N.; Mantzoros, C.S.; Dalamaga, M. Could the endocrine disruptor bisphenol-A be implicated in the pathogenesis of oral and oropharyngeal cancer? Metabolic considerations and future directions. Metabolism 2019, 91, 61–69. [Google Scholar] [CrossRef]
  11. Heindel, J.J.; Alvarez, J.A.; Atlas, E.; Cave, M.C.; Chatzi, V.L.; Collier, D.; Corkey, B.; Fischer, D.; Goran, M.I.; Howard, S.; et al. Obesogens and Obesity: State-of-the-Science and Future Directions Summary from a Healthy Environment and Endocrine Disruptors Strategies Workshop. Am. J. Clin. Nutr. 2023, 118, 329–337. [Google Scholar] [CrossRef]
  12. Panuganti, K.K.; Nguyen, M.; Kshirsagar, R.K. Obesity. [Updated 2023 Aug 8]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 January. Available online: https://www.ncbi.nlm.nih.gov/books/NBK459357/ (accessed on 2 December 2023).
  13. WHO. Obesity. Available online: https://www.who.int/health-topics/obesity (accessed on 26 December 2023).
  14. Liu, J.; Tsilingiris, D.; Dalamaga, M. The non-linear relationship between muscle mass and BMI calls into question the use of BMI as a major criterion for eligibility for bariatric surgery. Metabol. Open 2022, 13, 100164. [Google Scholar] [CrossRef] [PubMed]
  15. WHO. Physical status: The use and interpretation of anthropometry. Report of a WHO Expert Committee. World Health Organ. Tech. Rep. Ser. 1995, 854, 1–452. [Google Scholar]
  16. Larson, E.A.; Dalamaga, M.; Magkos, F. The role of exercise in obesity-related cancers: Current evidence and biological mechanisms. Semin. Cancer Biol. 2023, 91, 16–26. [Google Scholar] [CrossRef] [PubMed]
  17. Dalamaga, M.; Christodoulatos, G.S.; Karampela, I.; Vallianou, N.; Apovian, C.M. Understanding the Co-Epidemic of Obesity and COVID-19: Current Evidence, Comparison with Previous Epidemics, Mechanisms, and Preventive and Therapeutic Perspectives. Curr. Obes. Rep. 2021, 10, 214–243. [Google Scholar] [CrossRef] [PubMed]
  18. Katzmarzyk, P.T.; Pérusse, L.; Rao, D.C.; Bouchard, C. Familial risk of overweight and obesity in the Canadian population using the WHO/NIH criteria. Obes. Res. 2000, 8, 194–197. [Google Scholar] [CrossRef] [PubMed]
  19. Pietiläinen, K.H.; Kaprio, J.; Rissanen, A.; Winter, T.; Rimpelä, A.; Viken, R.J.; Rose, R.J. Distribution and heritability of BMI in Finnish adolescents aged 16y and 17y: A study of 4884 twins and 2509 singletons. Int. J. Obes. Relat. Metab. Disord. 1999, 23, 107–115. [Google Scholar] [CrossRef] [PubMed]
  20. Stunkard, A.J.; Sørensen, T.I.; Hanis, C.; Teasdale, T.W.; Chakraborty, R.; Schull, W.J.; Schulsinger, F. An adoption study of human obesity. N. Engl. J. Med. 1986, 314, 193–198. [Google Scholar] [CrossRef]
  21. Locke, A.E.; Kahali, B.; Berndt, S.I.; Justice, A.E.; Pers, T.H.; Day, F.R.; Powell, C.; Vedantam, S.; Buchkovich, M.L.; Yang, J.; et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 2015, 518, 197–206. [Google Scholar] [CrossRef]
  22. Keller, M.; Svensson, S.I.A.; Rohde-Zimmermann, K.; Kovacs, P.; Böttcher, Y. Genetics and Epigenetics in Obesity: What Do We Know so Far? Curr. Obes. Rep. 2023, 12, 482–501. [Google Scholar] [CrossRef]
  23. Kim, R.; Lippert, A.M.; Wedow, R.; Jimenez, M.P.; Subramanian, S.V. The Relative Contributions of Socioeconomic and Genetic Factors to Variations in Body Mass Index Among Young Adults. Am. J. Epidemiol. 2020, 189, 1333–1341. [Google Scholar] [CrossRef]
  24. Wang, Z.; Emmerich, A.; Pillon, N.J.; Moore, T.; Hemerich, D.; Cornelis, M.C.; Mazzaferro, E.; Broos, S.; Ahluwalia, T.S.; Bartz, T.M.; et al. Genome-wide association analyses of physical activity and sedentary behavior provide insights into underlying mechanisms and roles in disease prevention. Nat. Genet. 2022, 54, 1332–1344. [Google Scholar] [CrossRef]
  25. Yengo, L.; Sidorenko, J.; Kemper, K.E.; Zheng, Z.; Wood, A.R.; Weedon, M.N.; Frayling, T.M.; Hirschhorn, J.; Yang, J.; Visscher, P.M. Meta-analysis of genome-wide association studies for height and body mass index in ∼700000 individuals of European ancestry. Hum. Mol. Genet. 2018, 27, 3641–3649. [Google Scholar] [CrossRef] [PubMed]
  26. Argyrakopoulou, G.; Fountouli, N.; Dalamaga, M.; Kokkinos, A. Revisiting Resting Metabolic Rate: What is the Relation to Weight Fluctuations? Curr. Obes. Rep. 2023, 12, 502–513. [Google Scholar] [CrossRef] [PubMed]
  27. Fogarasi, A.; Gonzalez, K.; Dalamaga, M.; Magkos, F. The Impact of the Rate of Weight Loss on Body Composition and Metabolism. Curr. Obes. Rep. 2022, 11, 33–44. [Google Scholar] [CrossRef] [PubMed]
  28. Kiess, W.; Haeusler, G. Endocrine-disrupting chemicals. Best. Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101566. [Google Scholar] [CrossRef]
  29. WHO. Global Assessment on the State of the Science of Endocrine Disruptors. Available online: https://www.who.int/publications/i/item/WHO-PSC-EDC-02.2 (accessed on 28 November 2023).
  30. Nidens, N.; Vogel, M.; Körner, A.; Kiess, W. Prenatal exposure to phthalate esters and its impact on child development. Best. Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101478. [Google Scholar] [CrossRef]
  31. National Institute of Environmental Health Sciences. Endocrine Disruptors. Available online: https://www.niehs.nih.gov/health/topics/agents/endocrine/index.cfm (accessed on 28 November 2023).
  32. van der Meer, T.P.; van Faassen, M.; van Beek, A.P.; Snieder, H.; Kema, I.P.; Wolffenbuttel, B.H.R.; van Vliet-Ostaptchouk, J.V. Exposure to Endocrine Disrupting Chemicals in the Dutch general population is associated with adiposity-related traits. Sci. Rep. 2020, 10, 9311. [Google Scholar] [CrossRef]
  33. Frederiksen, H.; Jensen, T.K.; Jørgensen, N.; Kyhl, H.B.; Husby, S.; Skakkebæk, N.E.; Main, K.M.; Juul, A.; Andersson, A.M. Human urinary excretion of non-persistent environmental chemicals: An overview of Danish data collected between 2006 and 2012. Reproduction 2014, 147, 555–565. [Google Scholar] [CrossRef]
  34. Kiess, W.; Häussler, G.; Vogel, M. Endocrine-disrupting chemicals and child health. Best. Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101516. [Google Scholar] [CrossRef]
  35. Darbre, P.D. Endocrine Disruptors and Obesity. Curr. Obes. Rep. 2017, 6, 18–27. [Google Scholar] [CrossRef]
  36. Plastics—The Facts 2022. Available online: https://plasticseurope.org/wp-content/uploads/2022/10/PE-PLASTICS-THE-FACTS_V7-Tue_19-10-1.pdf (accessed on 28 November 2023).
  37. Ng, M.; Fleming, T.; Robinson, M.; Thomson, B.; Graetz, N.; Margono, C.; Mullany, E.C.; Biryukov, S.; Abbafati, C.; Abera, S.F.; et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014, 384, 766–781. [Google Scholar] [CrossRef] [PubMed]
  38. Michałowicz, J. Bisphenol A--sources, toxicity and biotransformation. Environ. Toxicol. Pharmacol. 2014, 37, 738–758. [Google Scholar] [CrossRef] [PubMed]
  39. Vandenberg, L.N.; Maffini, M.V.; Sonnenschein, C.; Rubin, B.S.; Soto, A.M. Bisphenol-A and the great divide: A review of controversies in the field of endocrine disruption. Endocr. Rev. 2009, 30, 75–95. [Google Scholar] [CrossRef]
  40. Kim, M.J.; Park, Y.J. Bisphenols and Thyroid Hormone. Endocrinol. Metab. 2019, 34, 340–348. [Google Scholar] [CrossRef] [PubMed]
  41. Rubin, B.S. Bisphenol A: An endocrine disruptor with widespread exposure and multiple effects. J. Steroid Biochem. Mol. Biol. 2011, 127, 27–34. [Google Scholar] [CrossRef] [PubMed]
  42. Dueñas-Mas, M.J.; Ballesteros-Gómez, A.; Rubio, S. Supramolecular solvent-based microextraction of emerging bisphenol A replacements (colour developers) in indoor dust from public environments. Chemosphere 2019, 222, 22–28. [Google Scholar] [CrossRef] [PubMed]
  43. Darbre, P.D. Chemical components of plastics as endocrine disruptors: Overview and commentary. Birth Defects Res. 2020, 112, 1300–1307. [Google Scholar] [CrossRef] [PubMed]
  44. Seachrist, D.D.; Bonk, K.W.; Ho, S.M.; Prins, G.S.; Soto, A.M.; Keri, R.A. A review of the carcinogenic potential of bisphenol A. Reprod. Toxicol. 2016, 59, 167–182. [Google Scholar] [CrossRef]
  45. Robinson, L.; Miller, R. The Impact of Bisphenol A and Phthalates on Allergy, Asthma, and Immune Function: A Review of Latest Findings. Curr. Environ. Health Rep. 2015, 2, 379–387. [Google Scholar] [CrossRef]
  46. Biemann, R.; Blüher, M.; Isermann, B. Exposure to endocrine-disrupting compounds such as phthalates and bisphenol A is associated with an increased risk for obesity. Best. Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101546. [Google Scholar] [CrossRef]
  47. Hoekstra, E.J.; Simoneau, C. Release of bisphenol A from polycarbonate: A review. Crit. Rev. Food Sci. Nutr. 2013, 53, 386–402. [Google Scholar] [CrossRef] [PubMed]
  48. Fenichel, P.; Chevalier, N.; Brucker-Davis, F. Bisphenol A: An endocrine and metabolic disruptor. Ann. Endocrinol. 2013, 74, 211–220. [Google Scholar] [CrossRef] [PubMed]
  49. Xu, J.; Huang, G.; Guo, T.L. Developmental Bisphenol A Exposure Modulates Immune-Related Diseases. Toxics 2016, 4, 23. [Google Scholar] [CrossRef] [PubMed]
  50. Di Credico, A.; Gaggi, G.; Bucci, I.; Ghinassi, B.; Di Baldassarre, A. The Effects of Combined Exposure to Bisphenols and Perfluoroalkyls on Human Perinatal Stem Cells and the Potential Implications for Health Outcomes. Int. J. Mol. Sci. 2023, 24, 15018. [Google Scholar] [CrossRef] [PubMed]
  51. Hassan, R.; Aslam Khan, M.U.; Abdullah, A.M.; Abd Razak, S.I. A Review on Current Trends of Polymers in Orthodontics: BPA-Free and Smart Materials. Polymers 2021, 13, 1409. [Google Scholar] [CrossRef]
  52. Liu, J.; Martin, J.W. Prolonged Exposure to Bisphenol A from Single Dermal Contact Events. Environ. Sci. Technol. 2017, 51, 9940–9949. [Google Scholar] [CrossRef]
  53. Christensen, K.L.; Lorber, M.; Koslitz, S.; Brüning, T.; Koch, H.M. The contribution of diet to total bisphenol A body burden in humans: Results of a 48 h fasting study. Environ. Int. 2012, 50, 7–14. [Google Scholar] [CrossRef]
  54. 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]
  55. Olsén, L.; Lampa, E.; Birkholz, D.A.; Lind, L.; Lind, P.M. Circulating levels of bisphenol A (BPA) and phthalates in an elderly population in Sweden, based on the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS). Ecotoxicol. Environ. Saf. 2012, 75, 242–248. [Google Scholar] [CrossRef]
  56. Kandaraki, E.; Chatzigeorgiou, A.; Livadas, S.; Palioura, E.; Economou, F.; Koutsilieris, M.; Palimeri, S.; Panidis, D.; Diamanti-Kandarakis, E. Endocrine disruptors and polycystic ovary syndrome (PCOS): Elevated serum levels of bisphenol A in women with PCOS. J. Clin. Endocrinol. Metab. 2011, 96, E480–E484. [Google Scholar] [CrossRef]
  57. Catenza, C.J.; Farooq, A.; Shubear, N.S.; Donkor, K.K. A targeted review on fate, occurrence, risk and health implications of bisphenol analogues. Chemosphere 2021, 268, 129273. [Google Scholar] [CrossRef] [PubMed]
  58. European Chemicals Agency, 2022. The use of bisphenol A and its alternatives in thermal paper in the EU during 2014–2022. Echa. Available online: https://echa.europa.eu/documents/10162/2564887/bpa_thermal_paper_report_2020_en.pdf/59eca269-c788-7942-5c17-3bd822d9cba0 (accessed on 2 December 2023).
  59. Bisphenol, A. Available online: https://www.efsa.europa.eu/en/topics/topic/bisphenol (accessed on 28 November 2023).
  60. Kodila, A.; Franko, N.; Sollner Dolenc, M. A review on immunomodulatory effects of BPA analogues. Arch. Toxicol. 2023, 97, 1831–1846. [Google Scholar] [CrossRef] [PubMed]
  61. Martínez, M.; Blanco, J.; Rovira, J.; Kumar, V.; Domingo, J.L.; Schuhmacher, M. Bisphenol A analogues (BPS and BPF) present a greater obesogenic capacity in 3T3-L1 cell line. Food Chem. Toxicol. 2020, 140, 111298. [Google Scholar] [CrossRef]
  62. Ramskov Tetzlaff, C.N.; Svingen, T.; Vinggaard, A.M.; Rosenmai, A.K.; Taxvig, C. Bisphenols B, E, F, and S and 4-cumylphenol induce lipid accumulation in mouse adipocytes similarly to bisphenol A. Environ. Toxicol. 2020, 35, 543–552. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, D.; Kannan, K.; Tan, H.; Zheng, Z.; Feng, Y.L.; Wu, Y.; Widelka, M. Bisphenol Analogues Other Than BPA: Environmental Occurrence, Human Exposure, and Toxicity-A Review. Environ. Sci. Technol. 2016, 50, 5438–5453. [Google Scholar] [CrossRef] [PubMed]
  64. McDonough, C.M.; Xu, H.S.; Guo, T.L. Toxicity of bisphenol analogues on the reproductive, nervous, and immune systems, and their relationships to gut microbiome and metabolism: Insights from a multi-species comparison. Crit. Rev. Toxicol. 2021, 51, 283–300. [Google Scholar] [CrossRef] [PubMed]
  65. Usman, A.; Ahmad, M. Computational study suggesting reconsideration of BPA analogues based on their endocrine disrupting potential estimated by binding affinities to nuclear receptors. Ecotoxicol. Environ. Saf. 2019, 171, 154–161. [Google Scholar] [CrossRef]
  66. U.S. Environmental Protection Agency. Phthalates. Available online: https://www.epa.gov/assessing-and-managing-chemicals-under-tsca/phthalates (accessed on 1 December 2023).
  67. Geyer, R.; Jambeck, J.R.; Law, K.L. Production, use, and fate of all plastics ever made. Sci. Adv. 2017, 3, e1700782. [Google Scholar] [CrossRef]
  68. Barnes, S.J. Understanding plastics pollution: The role of economic development and technological research. Environ. Pollut. 2019, 249, 812–821. [Google Scholar] [CrossRef]
  69. Wormuth, M.; Scheringer, M.; Vollenweider, M.; Hungerbühler, K. What are the sources of exposure to eight frequently used phthalic acid esters in Europeans? Risk Anal. 2006, 26, 803–824. [Google Scholar] [CrossRef]
  70. Serrano, S.E.; Braun, J.; Trasande, L.; Dills, R.; Sathyanarayana, S. Phthalates and diet: A review of the food monitoring and epidemiology data. Environ. Health 2014, 13, 43. [Google Scholar] [CrossRef] [PubMed]
  71. Vogel, N.; Lange, R.; Schmidt, P.; Rodriguez Martin, L.; Remy, S.; Springer, A.; Puklová, V.; Černá, M.; Rudnai, P.; Középesy, S.; et al. Exposure to Phthalates in European Children, Adolescents and Adults since 2005: A Harmonized Approach Based on Existing HBM Data in the HBM4EU Initiative. Toxics 2023, 11, 241. [Google Scholar] [CrossRef] [PubMed]
  72. Xia, B.; Zhu, Q.; Zhao, Y.; Ge, W.; Zhao, Y.; Song, Q.; Zhou, Y.; Shi, H.; Zhang, Y. Phthalate exposure and childhood overweight and obesity: Urinary metabolomic evidence. Environ. Int. 2018, 121, 159–168. [Google Scholar] [CrossRef] [PubMed]
  73. Govarts, E.; Gilles, L.; Rodriguez Martin, L.; Santonen, T.; Apel, P.; Alvito, P.; Anastasi, E.; Andersen, H.R.; Andersson, A.M.; Andryskova, L.; et al. Harmonized human biomonitoring in European children, teenagers and adults: EU-wide exposure data of 11 chemical substance groups from the HBM4EU Aligned Studies (2014–2021). Int. J. Hyg. Environ. Health 2023, 249, 114119. [Google Scholar] [CrossRef] [PubMed]
  74. Li, Y.; Xiao, N.; Liu, M.; Liu, Y.; He, A.; Wang, L.; Luo, H.; Yao, Y.; Sun, H. Dysregulation of steroid metabolome in follicular fluid links phthalate exposure to diminished ovarian reserve of childbearing-age women. Environ. Pollut. 2023, 330, 121730. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, Y.; Xiao, M.; Huang, K.; Cui, J.; Liu, H.; Yu, Y.; Ma, S.; Liu, X.; Lin, M. Phthalate metabolites in breast milk from mothers in Southern China: Occurrence, temporal trends, daily intake, and risk assessment. J. Hazard. Mater. 2023, 464, 132895. [Google Scholar] [CrossRef]
  76. Ferguson, K.K.; Rosen, E.M.; Rosario, Z.; Feric, Z.; Calafat, A.M.; McElrath, T.F.; Vélez Vega, C.; Cordero, J.F.; Alshawabkeh, A.; Meeker, J.D. Environmental phthalate exposure and preterm birth in the PROTECT birth cohort. Environ. Int. 2019, 132, 105099. [Google Scholar] [CrossRef]
  77. Choi, G.; Villanger, G.D.; Drover, S.S.M.; Sakhi, A.K.; Thomsen, C.; Nethery, R.C.; Zeiner, P.; Knudsen, G.P.; Reichborn-Kjennerud, T.; Øvergaard, K.R.; et al. Prenatal phthalate exposures and executive function in preschool children. Environ. Int. 2021, 149, 106403. [Google Scholar] [CrossRef]
  78. Stahlhut, R.W.; van Wijngaarden, E.; Dye, T.D.; Cook, S.; Swan, S.H. Concentrations of urinary phthalate metabolites are associated with increased waist circumference and insulin resistance in adult U.S. males. Environ. Health Perspect. 2007, 115, 876–882. [Google Scholar] [CrossRef]
  79. Commission Regulation (EU) 2018/2005 of 17 December 2018 amending Annex XVII to Regulation (EC) No 1907/2006 of the European Parliament and of the Council concerning the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH) as regards bis(2-ethylhexyl) phthalate (DEHP), dibutyl phthalate (DBP), benzyl butyl phthalate (BBP) and diisobutyl phthalate (DIBP) (Text with EEA relevance.). Available online: https://eur-lex.europa.eu/eli/reg/2018/2005/oj (accessed on 1 December 2023).
  80. van der Meer, T.P.; Chung, M.K.; van Faassen, M.; Makris, K.C.; van Beek, A.P.; Kema, I.P.; Wolffenbuttel, B.H.R.; van Vliet-Ostaptchouk, J.V.; Patel, C.J. Temporal exposure and consistency of endocrine disrupting chemicals in a longitudinal study of individuals with impaired fasting glucose. Environ. Res. 2021, 197, 110901. [Google Scholar] [CrossRef]
  81. Karampela, I.; Christodoulatos, G.S.; Dalamaga, M. The Role of Adipose Tissue and Adipokines in Sepsis: Inflammatory and Metabolic Considerations, and the Obesity Paradox. Curr. Obes. Rep. 2019, 8, 434–457. [Google Scholar] [CrossRef] [PubMed]
  82. Vandenberg, L.N.; Colborn, T.; Hayes, T.B.; Heindel, J.J.; Jacobs, D.R., Jr.; Lee, D.H.; Shioda, T.; Soto, A.M.; vom Saal, F.S.; Welshons, W.V.; et al. Hormones and endocrine-disrupting chemicals: Low-dose effects and nonmonotonic dose responses. Endocr. Rev. 2012, 33, 378–455. [Google Scholar] [CrossRef]
  83. Mohammadi, H.; Ashari, S. Mechanistic insight into toxicity of phthalates, the involved receptors, and the role of Nrf2, NF-κB, and PI3K/AKT signaling pathways. Environ. Sci. Pollut. Res. Int. 2021, 28, 35488–35527. [Google Scholar] [CrossRef] [PubMed]
  84. González-Casanova, J.E.; Bermúdez, V.; Caro Fuentes, N.J.; Angarita, L.C.; Caicedo, N.H.; Rivas Muñoz, J.; Rojas-Gómez, D.M. New Evidence on BPA’s Role in Adipose Tissue Development of Proinflammatory Processes and Its Relationship with Obesity. Int. J. Mol. Sci. 2023, 24, 8231. [Google Scholar] [CrossRef] [PubMed]
  85. Sargis, R.M.; Johnson, D.N.; Choudhury, R.A.; Brady, M.J. Environmental endocrine disruptors promote adipogenesis in the 3T3-L1 cell line through glucocorticoid receptor activation. Obesity 2010, 18, 1283–1288. [Google Scholar] [CrossRef]
  86. Feige, J.N.; Gelman, L.; Rossi, D.; Zoete, V.; Métivier, R.; Tudor, C.; Anghel, S.I.; Grosdidier, A.; Lathion, C.; Engelborghs, Y.; et al. The endocrine disruptor monoethyl-hexyl-phthalate is a selective peroxisome proliferator-activated receptor gamma modulator that promotes adipogenesis. J. Biol. Chem. 2007, 282, 19152–19166. [Google Scholar] [CrossRef]
  87. Esteban, J.; Serrano-Maciá, M.; Sánchez-Pérez, I.; Alonso-Magdalena, P.; Pellín, M.C.; García-Arévalo, M.; Nadal, Á.; Barril, J. In utero exposure to bisphenol-A disrupts key elements of retinoid system in male mice offspring. Food Chem. Toxicol. 2019, 126, 142–151. [Google Scholar] [CrossRef]
  88. Nettore, I.C.; Franchini, F.; Palatucci, G.; Macchia, P.E.; Ungaro, P. Epigenetic Mechanisms of Endocrine-Disrupting Chemicals in Obesity. Biomedicines 2021, 9, 1716. [Google Scholar] [CrossRef]
  89. Lopez-Rodriguez, D.; Franssen, D.; Heger, S.; Parent, A.S. Endocrine-disrupting chemicals and their effects on puberty. Best. Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101579. [Google Scholar] [CrossRef]
  90. Spalding, K.L.; Arner, E.; Westermark, P.O.; Bernard, S.; Buchholz, B.A.; Bergmann, O.; Blomqvist, L.; Hoffstedt, J.; Näslund, E.; Britton, T.; et al. Dynamics of fat cell turnover in humans. Nature 2008, 453, 783–787. [Google Scholar] [CrossRef]
  91. Rosen, E.D.; Spiegelman, B.M. PPARgamma: A nuclear regulator of metabolism, differentiation, and cell growth. J. Biol. Chem. 2001, 276, 37731–37734. [Google Scholar] [CrossRef] [PubMed]
  92. Ariemma, F.; D’Esposito, V.; Liguoro, D.; Oriente, F.; Cabaro, S.; Liotti, A.; Cimmino, I.; Longo, M.; Beguinot, F.; Formisano, P.; et al. Low-Dose Bisphenol-A Impairs Adipogenesis and Generates Dysfunctional 3T3-L1 Adipocytes. PLoS ONE 2016, 11, e0150762. [Google Scholar] [CrossRef] [PubMed]
  93. Dong, H.; Yao, X.; Liu, S.; Yin, N.; Faiola, F. Non-cytotoxic nanomolar concentrations of bisphenol A induce human mesenchymal stem cell adipogenesis and osteogenesis. Ecotoxicol. Environ. Saf. 2018, 164, 448–454. [Google Scholar] [CrossRef] [PubMed]
  94. Reina-Pérez, I.; Olivas-Martínez, A.; Mustieles, V.; Ruiz-Ojeda, F.J.; Molina-Molina, J.M.; Olea, N.; Fernández, M.F. Bisphenol F and bisphenol S promote lipid accumulation and adipogenesis in human adipose-derived stem cells. Food Chem. Toxicol. 2021, 152, 112216. [Google Scholar] [CrossRef] [PubMed]
  95. Hurst, C.H.; Waxman, D.J. Activation of PPARalpha and PPARgamma by environmental phthalate monoesters. Toxicol. Sci. 2003, 74, 297–308. [Google Scholar] [CrossRef]
  96. Yin, L.; Yu, K.S.; Lu, K.; Yu, X. Benzyl butyl phthalate promotes adipogenesis in 3T3-L1 preadipocytes: A High Content Cellomics and metabolomic analysis. Toxicol. Vitro 2016, 32, 297–309. [Google Scholar] [CrossRef]
  97. Yang, Q.; Nagano, T.; Shah, Y.; Cheung, C.; Ito, S.; Gonzalez, F.J. The PPAR alpha-humanized mouse: A model to investigate species differences in liver toxicity mediated by PPAR alpha. Toxicol. Sci. 2008, 101, 132–139. [Google Scholar] [CrossRef]
  98. Schmidt, J.S.; Schaedlich, K.; Fiandanese, N.; Pocar, P.; Fischer, B. Effects of di(2-ethylhexyl) phthalate (DEHP) on female fertility and adipogenesis in C3H/N mice. Environ. Health Perspect. 2012, 120, 1123–1129. [Google Scholar] [CrossRef]
  99. 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]
  100. Lang, I.A.; Galloway, T.S.; Scarlett, A.; Henley, W.E.; Depledge, M.; Wallace, R.B.; Melzer, D. Association of urinary bisphenol A concentration with medical disorders and laboratory abnormalities in adults. Jama 2008, 300, 1303–1310. [Google Scholar] [CrossRef]
  101. Teppala, S.; Madhavan, S.; Shankar, A. Bisphenol A and Metabolic Syndrome: Results from NHANES. Int. J. Endocrinol. 2012, 2012, 598180. [Google Scholar] [CrossRef]
  102. Bastos Sales, L.; Kamstra, J.H.; Cenijn, P.H.; van Rijt, L.S.; Hamers, T.; Legler, J. Effects of endocrine disrupting chemicals on in vitro global DNA methylation and adipocyte differentiation. Toxicol. Vitro 2013, 27, 1634–1643. [Google Scholar] [CrossRef] [PubMed]
  103. Longo, M.; Zatterale, F.; Naderi, J.; Nigro, C.; Oriente, F.; Formisano, P.; Miele, C.; Beguinot, F. Low-dose Bisphenol-A Promotes Epigenetic Changes at Pparγ Promoter in Adipose Precursor Cells. Nutrients 2020, 12, 3498. [Google Scholar] [CrossRef] [PubMed]
  104. Meruvu, S.; Zhang, J.; Choudhury, M. Butyl Benzyl Phthalate Promotes Adipogenesis in 3T3-L1 Cells via the miRNA-34a-5p Signaling Pathway in the Absence of Exogenous Adipogenic Stimuli. Chem. Res. Toxicol. 2021, 34, 2251–2260. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, J.; Choudhury, M. The plasticizer BBP selectively inhibits epigenetic regulator sirtuin during differentiation of C3H10T1/2 stem cell line. Toxicol. Vitro 2017, 39, 75–83. [Google Scholar] [CrossRef] [PubMed]
  106. Strakovsky, R.S.; Wang, H.; Engeseth, N.J.; Flaws, J.A.; Helferich, W.G.; Pan, Y.X.; Lezmi, S. Developmental bisphenol A (BPA) exposure leads to sex-specific modification of hepatic gene expression and epigenome at birth that may exacerbate high-fat diet-induced hepatic steatosis. Toxicol. Appl. Pharmacol. 2015, 284, 101–112. [Google Scholar] [CrossRef] [PubMed]
  107. Miura, R.; Ikeda-Araki, A.; Ishihara, T.; Miyake, K.; Miyashita, C.; Nakajima, T.; Kobayashi, S.; Ishizuka, M.; Kubota, T.; Kishi, R. Effect of prenatal exposure to phthalates on epigenome-wide DNA methylations in cord blood and implications for fetal growth: The Hokkaido Study on Environment and Children’s Health. Sci. Total Environ. 2021, 783, 147035. [Google Scholar] [CrossRef] [PubMed]
  108. Charisiadis, P.; Andrianou, X.D.; van der Meer, T.P.; den Dunnen, W.F.A.; Swaab, D.F.; Wolffenbuttel, B.H.R.; Makris, K.C.; van Vliet-Ostaptchouk, J.V. Possible Obesogenic Effects of Bisphenols Accumulation in the Human Brain. Sci. Rep. 2018, 8, 8186. [Google Scholar] [CrossRef]
  109. Naomi, R.; Yazid, M.D.; Bahari, H.; Keong, Y.Y.; Rajandram, R.; Embong, H.; Teoh, S.H.; Halim, S.; Othman, F. Bisphenol A (BPA) Leading to Obesity and Cardiovascular Complications: A Compilation of Current In Vivo Study. Int. J. Mol. Sci. 2022, 23, 2969. [Google Scholar] [CrossRef]
  110. McIlwraith, E.K.; Lieu, C.V.; Belsham, D.D. Bisphenol A induces miR-708-5p through an ER stress-mediated mechanism altering neuronatin and neuropeptide Y expression in hypothalamic neuronal models. Mol. Cell Endocrinol. 2022, 539, 111480. [Google Scholar] [CrossRef]
  111. Loganathan, N.; McIlwraith, E.K.; Belsham, D.D. Bisphenol A Induces Agrp Gene Expression in Hypothalamic Neurons through a Mechanism Involving ATF3. Neuroendocrinology 2021, 111, 678–695. [Google Scholar] [CrossRef] [PubMed]
  112. Yi, M.; Li, H.; Wu, Z.; Yan, J.; Liu, Q.; Ou, C.; Chen, M. A Promising Therapeutic Target for Metabolic Diseases: Neuropeptide Y Receptors in Humans. Cell Physiol. Biochem. 2018, 45, 88–107. [Google Scholar] [CrossRef] [PubMed]
  113. Tian, S.; Yan, S.; Meng, Z.; Huang, S.; Sun, W.; Jia, M.; Teng, M.; Zhou, Z.; Zhu, W. New insights into bisphenols induced obesity in zebrafish (Danio rerio): Activation of cannabinoid receptor CB1. J. Hazard. Mater. 2021, 418, 126100. [Google Scholar] [CrossRef] [PubMed]
  114. Khalifa, M.; Fayed, R.H.; Sedik, A.A.; Khalil, H.M.A. Dose-dependent toxic effects of di-(2-ethylhexyl) phthalate in male rats: Focus on behavioral alterations and inducing TLR4/NF-κB signaling pathway. Toxicol. Appl. Pharmacol. 2023, 468, 116515. [Google Scholar] [CrossRef] [PubMed]
  115. Rönn, M.; Lind, L.; Örberg, J.; Kullberg, J.; Söderberg, S.; Larsson, A.; Johansson, L.; Ahlström, H.; Lind, P.M. Bisphenol A is related to circulating levels of adiponectin, leptin and ghrelin, but not to fat mass or fat distribution in humans. Chemosphere 2014, 112, 42–48. [Google Scholar] [CrossRef] [PubMed]
  116. Mondal, S.; Basu, S.; Ghosh, S.; Guria, S.; Mukherjee, S. Diethyl phthalate, a plasticizer, induces adipocyte inflammation and apoptosis in mice after long-term dietary administration. J. Biochem. Mol. Toxicol. 2023, e23561. [Google Scholar] [CrossRef] [PubMed]
  117. Schaedlich, K.; Beier, L.S.; Kolbe, J.; Wabitsch, M.; Ernst, J. Pro-inflammatory effects of DEHP in SGBS-derived adipocytes and THP-1 macrophages. Sci. Rep. 2021, 11, 7928. [Google Scholar] [CrossRef] [PubMed]
  118. Valentino, R.; D’Esposito, V.; Passaretti, F.; Liotti, A.; Cabaro, S.; Longo, M.; Perruolo, G.; Oriente, F.; Beguinot, F.; Formisano, P. Bisphenol-A impairs insulin action and up-regulates inflammatory pathways in human subcutaneous adipocytes and 3T3-L1 cells. PLoS ONE 2013, 8, e82099. [Google Scholar] [CrossRef]
  119. Shi, M.; Lin, Z.; Ye, L.; Chen, X.; Zhang, W.; Zhang, Z.; Luo, F.; Liu, Y.; Shi, M. Estrogen receptor-regulated SOCS3 modulation via JAK2/STAT3 pathway is involved in BPF-induced M1 polarization of macrophages. Toxicology 2020, 433–434, 152404. [Google Scholar] [CrossRef]
  120. Piao, X.; Liu, Z.; Li, Y.; Yao, D.; Sun, L.; Wang, B.; Ma, Y.; Wang, L.; Zhang, Y. Investigation of the effect for bisphenol A on oxidative stress in human hepatocytes and its interaction with catalase. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2019, 221, 117149. [Google Scholar] [CrossRef]
  121. Schaffert, A.; Arnold, J.; Karkossa, I.; Blüher, M.; von Bergen, M.; Schubert, K. The Emerging Plasticizer Alternative DINCH and Its Metabolite MINCH Induce Oxidative Stress and Enhance Inflammatory Responses in Human THP-1 Macrophages. Cells 2021, 10, 2367. [Google Scholar] [CrossRef] [PubMed]
  122. Hong, X.; Zhou, Y.; Zhu, Z.; Li, Y.; Li, Z.; Zhang, Y.; Hu, X.; Zhu, F.; Wang, Y.; Fang, M.; et al. Environmental endocrine disruptor Bisphenol A induces metabolic derailment and obesity via upregulating IL-17A in adipocytes. Environ. Int. 2023, 172, 107759. [Google Scholar] [CrossRef] [PubMed]
  123. Jain, J.; Gupta, N.; Mathur, R.; Nimesh, S.; Mathur, S.K. A Study on Impact of BPA in the Adipose Tissue Dysfunction (Adiposopathy) in Asian Indian Type 2 Diabetes Mellitus Subjects. Indian. J. Clin. Biochem. 2020, 35, 451–457. [Google Scholar] [CrossRef] [PubMed]
  124. Yang, Y.J.; Hong, Y.C.; Oh, S.Y.; Park, M.S.; Kim, H.; Leem, J.H.; Ha, E.H. Bisphenol A exposure is associated with oxidative stress and inflammation in postmenopausal women. Environ. Res. 2009, 109, 797–801. [Google Scholar] [CrossRef] [PubMed]
  125. Reddivari, L.; Veeramachaneni, D.N.R.; Walters, W.A.; Lozupone, C.; Palmer, J.; Hewage, M.K.K.; Bhatnagar, R.; Amir, A.; Kennett, M.J.; Knight, R.; et al. Perinatal Bisphenol A Exposure Induces Chronic Inflammation in Rabbit Offspring via Modulation of Gut Bacteria and Their Metabolites. mSystems 2017, 2, e00093-17. [Google Scholar] [CrossRef] [PubMed]
  126. Gálvez-Ontiveros, Y.; Páez, S.; Monteagudo, C.; Rivas, A. Endocrine Disruptors in Food: Impact on Gut Microbiota and Metabolic Diseases. Nutrients 2020, 12, 1158. [Google Scholar] [CrossRef] [PubMed]
  127. Diamante, G.; Cely, I.; Zamora, Z.; Ding, J.; Blencowe, M.; Lang, J.; Bline, A.; Singh, M.; Lusis, A.J.; Yang, X. Systems toxicogenomics of prenatal low-dose BPA exposure on liver metabolic pathways, gut microbiota, and metabolic health in mice. Environ. Int. 2021, 146, 106260. [Google Scholar] [CrossRef] [PubMed]
  128. Chai, X.; Wen, L.; Song, Y.; He, X.; Yue, J.; Wu, J.; Chen, X.; Cai, Z.; Qi, Z. DEHP exposure elevated cardiovascular risk in obese mice by disturbing the arachidonic acid metabolism of gut microbiota. Sci. Total Environ. 2023, 875, 162615. [Google Scholar] [CrossRef]
  129. Su, H.; Yuan, P.; Lei, H.; Zhang, L.; Deng, D.; Zhang, L.; Chen, X. Long-term chronic exposure to di-(2-ethylhexyl)-phthalate induces obesity via disruption of host lipid metabolism and gut microbiota in mice. Chemosphere 2022, 287, 132414. [Google Scholar] [CrossRef]
  130. Francis, C.E.; Allee, L.; Nguyen, H.; Grindstaff, R.D.; Miller, C.N.; Rayalam, S. Endocrine disrupting chemicals: Friend or foe to brown and beige adipose tissue? Toxicology 2021, 463, 152972. [Google Scholar] [CrossRef]
  131. Himms-Hagen, J. Brown adipose tissue thermogenesis: Interdisciplinary studies. Faseb J. 1990, 4, 2890–2898. [Google Scholar] [CrossRef] [PubMed]
  132. Nunez, A.A.; Kannan, K.; Giesy, J.P.; Fang, J.; Clemens, L.G. Effects of bisphenol A on energy balance and accumulation in brown adipose tissue in rats. Chemosphere 2001, 42, 917–922. [Google Scholar] [CrossRef] [PubMed]
  133. Lee, K.I.; Chiang, C.W.; Lin, H.C.; Zhao, J.F.; Li, C.T.; Shyue, S.K.; Lee, T.S. Maternal exposure to di-(2-ethylhexyl) phthalate exposure deregulates blood pressure, adiposity, cholesterol metabolism and social interaction in mouse offspring. Arch. Toxicol. 2016, 90, 1211–1224. [Google Scholar] [CrossRef] [PubMed]
  134. van Esterik, J.C.; Dollé, M.E.; Lamoree, M.H.; van Leeuwen, S.P.; Hamers, T.; Legler, J.; van der Ven, L.T. Programming of metabolic effects in C57BL/6JxFVB mice by exposure to bisphenol A during gestation and lactation. Toxicology 2014, 321, 40–52. [Google Scholar] [CrossRef] [PubMed]
  135. Lv, Z.; Cheng, J.; Huang, S.; Zhang, Y.; Wu, S.; Qiu, Y.; Geng, Y.; Zhang, Q.; Huang, G.; Ma, Q.; et al. DEHP induces obesity and hypothyroidism through both central and peripheral pathways in C3H/He mice. Obesity 2016, 24, 368–378. [Google Scholar] [CrossRef] [PubMed]
  136. Stratigou, T.; Dalamaga, M.; Antonakos, G.; Marinou, I.; Vogiatzakis, E.; Christodoulatos, G.S.; Karampela, I.; Papavassiliou, A.G. Hyperirisinemia is independently associated with subclinical hypothyroidism: Correlations with cardiometabolic biomarkers and risk factors. Endocrine 2018, 61, 83–93. [Google Scholar] [CrossRef] [PubMed]
  137. Menale, C.; Piccolo, M.T.; Cirillo, G.; Calogero, R.A.; Papparella, A.; Mita, L.; Del Giudice, E.M.; Diano, N.; Crispi, S.; Mita, D.G. Bisphenol A effects on gene expression in adipocytes from children: Association with metabolic disorders. J. Mol. Endocrinol. 2015, 54, 289–303. [Google Scholar] [CrossRef]
  138. Campioli, E.; Batarseh, A.; Li, J.; Papadopoulos, V. The endocrine disruptor mono-(2-ethylhexyl) phthalate affects the differentiation of human liposarcoma cells (SW 872). PLoS ONE 2011, 6, e28750. [Google Scholar] [CrossRef]
  139. Biemann, R.; Navarrete Santos, A.; Navarrete Santos, A.; Riemann, D.; Knelangen, J.; Blüher, M.; Koch, H.; Fischer, B. Endocrine disrupting chemicals affect the adipogenic differentiation of mesenchymal stem cells in distinct ontogenetic windows. Biochem. Biophys. Res. Commun. 2012, 417, 747–752. [Google Scholar] [CrossRef]
  140. Ohlstein, J.F.; Strong, A.L.; McLachlan, J.A.; Gimble, J.M.; Burow, M.E.; Bunnell, B.A. Bisphenol A enhances adipogenic differentiation of human adipose stromal/stem cells. J. Mol. Endocrinol. 2014, 53, 345–353. [Google Scholar] [CrossRef]
  141. Schaffert, A.; Karkossa, I.; Ueberham, E.; Schlichting, R.; Walter, K.; Arnold, J.; Blüher, M.; Heiker, J.T.; Lehmann, J.; Wabitsch, M.; et al. Di-(2-ethylhexyl) phthalate substitutes accelerate human adipogenesis through PPARγ activation and cause oxidative stress and impaired metabolic homeostasis in mature adipocytes. Environ. Int. 2022, 164, 107279. [Google Scholar] [CrossRef] [PubMed]
  142. Adachi, T.; Yasuda, K.; Mori, C.; Yoshinaga, M.; Aoki, N.; Tsujimoto, G.; Tsuda, K. Promoting insulin secretion in pancreatic islets by means of bisphenol A and nonylphenol via intracellular estrogen receptors. Food Chem. Toxicol. 2005, 43, 713–719. [Google Scholar] [CrossRef] [PubMed]
  143. De Filippis, E.; Li, T.; Rosen, E.D. Exposure of adipocytes to bisphenol-A in vitro interferes with insulin action without enhancing adipogenesis. PLoS ONE 2018, 13, e0201122. [Google Scholar] [CrossRef]
  144. Boucher, J.G.; Husain, M.; Rowan-Carroll, A.; Williams, A.; Yauk, C.L.; Atlas, E. Identification of mechanisms of action of bisphenol a-induced human preadipocyte differentiation by transcriptional profiling. Obesity 2014, 22, 2333–2343. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, Y.; Wang, S.; Zhao, T.; Yang, L.; Guo, S.; Shi, Y.; Zhang, X.; Zhou, L.; Ye, L. Mono-2-ethylhexyl phthalate (MEHP) promoted lipid accumulation via JAK2/STAT5 and aggravated oxidative stress in BRL-3A cells. Ecotoxicol. Environ. Saf. 2019, 184, 109611. [Google Scholar] [CrossRef] [PubMed]
  146. Bucher, S.; Jalili, P.; Le Guillou, D.; Begriche, K.; Rondel, K.; Martinais, S.; Zalko, D.; Corlu, A.; Robin, M.A.; Fromenty, B. Bisphenol a induces steatosis in HepaRG cells using a model of perinatal exposure. Environ. Toxicol. 2017, 32, 1024–1036. [Google Scholar] [CrossRef] [PubMed]
  147. Martella, A.; Silvestri, C.; Maradonna, F.; Gioacchini, G.; Allarà, M.; Radaelli, G.; Overby, D.R.; Di Marzo, V.; Carnevali, O. Bisphenol A Induces Fatty Liver by an Endocannabinoid-Mediated Positive Feedback Loop. Endocrinology 2016, 157, 1751–1763. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, J.; Sun, B.; Hou, M.; Pan, X.; Li, X. The environmental obesogen bisphenol A promotes adipogenesis by increasing the amount of 11β-hydroxysteroid dehydrogenase type 1 in the adipose tissue of children. Int. J. Obes 2013, 37, 999–1005. [Google Scholar] [CrossRef]
  149. Grasselli, E.; Cortese, K.; Voci, A.; Vergani, L.; Fabbri, R.; Barmo, C.; Gallo, G.; Canesi, L. Direct effects of Bisphenol A on lipid homeostasis in rat hepatoma cells. Chemosphere 2013, 91, 1123–1129. [Google Scholar] [CrossRef]
  150. Dimastrogiovanni, G.; Córdoba, M.; Navarro, I.; Jáuregui, O.; Porte, C. Alteration of cellular lipids and lipid metabolism markers in RTL-W1 cells exposed to model endocrine disrupters. Aquat. Toxicol. 2015, 165, 277–285. [Google Scholar] [CrossRef]
  151. Schaedlich, K.; Gebauer, S.; Hunger, L.; Beier, L.S.; Koch, H.M.; Wabitsch, M.; Fischer, B.; Ernst, J. DEHP deregulates adipokine levels and impairs fatty acid storage in human SGBS-adipocytes. Sci. Rep. 2018, 8, 3447. [Google Scholar] [CrossRef] [PubMed]
  152. Riu, A.; Grimaldi, M.; le Maire, A.; Bey, G.; Phillips, K.; Boulahtouf, A.; Perdu, E.; Zalko, D.; Bourguet, W.; Balaguer, P. Peroxisome proliferator-activated receptor γ is a target for halogenated analogs of bisphenol A. Environ. Health Perspect. 2011, 119, 1227–1232. [Google Scholar] [CrossRef] [PubMed]
  153. Cohen, I.C.; Cohenour, E.R.; Harnett, K.G.; Schuh, S.M. BPA, BPAF and TMBPF Alter Adipogenesis and Fat Accumulation in Human Mesenchymal Stem Cells, with Implications for Obesity. Int. J. Mol. Sci. 2021, 22, 5363. [Google Scholar] [CrossRef] [PubMed]
  154. Yamasaki, M.; Hasegawa, S.; Imai, M.; Fukui, T.; Takahashi, N. Browning Effect of Brominated Flame Retardant, TBBP-A, on Undifferentiated Adipocytes. BPB Rep. 2021, 4, 41–46. [Google Scholar] [CrossRef] [PubMed]
  155. Marqueño, A.; Pérez-Albaladejo, E.; Denslow, N.D.; Bowden, J.A.; Porte, C. Untargeted lipidomics reveals the toxicity of bisphenol A bis(3-chloro-2- hydroxypropyl) ether and bisphenols A and F in zebrafish liver cells. Ecotoxicol. Environ. Saf. 2021, 219, 112311. [Google Scholar] [CrossRef] [PubMed]
  156. Lee, J.L.; Wang, Y.C.; Hsu, Y.A.; Chen, C.S.; Weng, R.C.; Lu, Y.P.; Chuang, C.Y.; Wan, L. Bisphenol A Coupled with a High-Fat Diet Promotes Hepatosteatosis through Reactive-Oxygen-Species-Induced CD36 Overexpression. Toxics 2022, 10, 208. [Google Scholar] [CrossRef]
  157. Pérez-Albaladejo, E.; Solís, A.; Bani, I.; Porte, C. PLHC-1 topminnow liver cells: An alternative model to investigate the toxicity of plastic additives in the aquatic environment. Ecotoxicol. Environ. Saf. 2021, 208, 111746. [Google Scholar] [CrossRef]
  158. Al-Abdulla, R.; Ferrero, H.; Soriano, S.; Boronat-Belda, T.; Alonso-Magdalena, P. Screening of Relevant Metabolism-Disrupting Chemicals on Pancreatic β-Cells: Evaluation of Murine and Human In Vitro Models. Int. J. Mol. Sci. 2022, 23, 4182. [Google Scholar] [CrossRef]
  159. Wei, J.; Lin, Y.; Li, Y.; Ying, C.; Chen, J.; Song, L.; Zhou, Z.; Lv, Z.; Xia, W.; Chen, X.; et al. Perinatal exposure to bisphenol A at reference dose predisposes offspring to metabolic syndrome in adult rats on a high-fat diet. Endocrinology 2011, 152, 3049–3061. [Google Scholar] [CrossRef]
  160. Desai, M.; Ferrini, M.G.; Jellyman, J.K.; Han, G.; Ross, M.G. In vivo and in vitro bisphenol A exposure effects on adiposity. J. Dev. Orig. Health Dis. 2018, 9, 678–687. [Google Scholar] [CrossRef]
  161. Pu, Y.; Gingrich, J.D.; Steibel, J.P.; Veiga-Lopez, A. Sex-Specific Modulation of Fetal Adipogenesis by Gestational Bisphenol A and Bisphenol S Exposure. Endocrinology 2017, 158, 3844–3858. [Google Scholar] [CrossRef] [PubMed]
  162. Hao, C.; Cheng, X.; Guo, J.; Xia, H.; Ma, X. Perinatal exposure to diethyl-hexyl-phthalate induces obesity in mice. Front. Biosci. 2013, 5, 725–733. [Google Scholar] [CrossRef] [PubMed]
  163. Klöting, 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]
  164. Hao, C.; Cheng, X.; Xia, H.; Ma, X. The endocrine disruptor mono-(2-ethylhexyl) phthalate promotes adipocyte differentiation and induces obesity in mice. Biosci. Rep. 2012, 32, 619–629. [Google Scholar] [CrossRef] [PubMed]
  165. Zhuang, Y.; Sun, X.; Deng, S.; Wen, Y.; Xu, Q.; Guan, Q. In vivo effects of low dose prenatal bisphenol A exposure on adiposity in male and female ICR offspring. Ecotoxicol. Environ. Saf. 2023, 257, 114946. [Google Scholar] [CrossRef] [PubMed]
  166. Oliviero, F.; Marmugi, A.; Viguié, C.; Gayrard, V.; Picard-Hagen, N.; Mselli-Lakhal, L. Are BPA Substitutes as Obesogenic as BPA? Int. J. Mol. Sci. 2022, 23, 4238. [Google Scholar] [CrossRef]
  167. Huff, M.; da Silveira, W.A.; Carnevali, O.; Renaud, L.; Hardiman, G. Systems Analysis of the Liver Transcriptome in Adult Male Zebrafish Exposed to the Plasticizer (2-Ethylhexyl) Phthalate (DEHP). Sci. Rep. 2018, 8, 2118. [Google Scholar] [CrossRef]
  168. Amara, I.; Timoumi, R.; Annabi, E.; Neffati, F.; Najjar, M.F.; Bouaziz, C.; Abid-Essefi, S. Di (2-ethylhexyl) phthalate induces cardiac disorders in BALB/c mice. Environ. Sci. Pollut. Res. Int. 2019, 26, 7540–7549. [Google Scholar] [CrossRef]
  169. Tête, A.; Gallais, I.; Imran, M.; Legoff, L.; Martin-Chouly, C.; Sparfel, L.; Bescher, M.; Sergent, O.; Podechard, N.; Lagadic-Gossmann, D. MEHP/ethanol co-exposure favors the death of steatotic hepatocytes, possibly through CYP4A and ADH involvement. Food Chem. Toxicol. 2020, 146, 111798. [Google Scholar] [CrossRef]
  170. Jia, P.P.; Junaid, M.; Xin, G.Y.; Wang, Y.; Ma, Y.B.; Pei, D.S. Disruption of Intestinal Homeostasis Through Altered Responses of the Microbial Community, Energy Metabolites, and Immune System in Zebrafish after Chronic Exposure to DEHP. Front. Microbiol. 2021, 12, 729530. [Google Scholar] [CrossRef]
  171. Susiarjo, M.; Xin, F.; Bansal, A.; Stefaniak, M.; Li, C.; Simmons, R.A.; Bartolomei, M.S. Bisphenol a exposure disrupts metabolic health across multiple generations in the mouse. Endocrinology 2015, 156, 2049–2058. [Google Scholar] [CrossRef] [PubMed]
  172. Guo, W.; Han, J.; Wu, S.; Shi, X.; Wang, Q.; Zhou, B. Bis(2-ethylhexyl)-2,3,4,5-tetrabromophthalate Affects Lipid Metabolism in Zebrafish Larvae via DNA Methylation Modification. Environ. Sci. Technol. 2020, 54, 355–363. [Google Scholar] [CrossRef]
  173. Stoker, C.; Andreoli, M.F.; Kass, L.; Bosquiazzo, V.L.; Rossetti, M.F.; Canesini, G.; Luque, E.H.; Ramos, J.G. Perinatal exposure to bisphenol A (BPA) impairs neuroendocrine mechanisms regulating food intake and kisspetin system in adult male rats. Evidences of metabolic disruptor hypothesis. Mol. Cell Endocrinol. 2020, 499, 110614. [Google Scholar] [CrossRef] [PubMed]
  174. Lin, R.; Jia, Y.; Wu, F.; Meng, Y.; Sun, Q.; Jia, L. Combined Exposure to Fructose and Bisphenol A Exacerbates Abnormal Lipid Metabolism in Liver of Developmental Male Rats. Int. J. Environ. Res. Public. Health 2019, 16, 4152. [Google Scholar] [CrossRef] [PubMed]
  175. Shih, M.K.; Tain, Y.L.; Chen, Y.W.; Hsu, W.H.; Yeh, Y.T.; Chang, S.K.C.; Liao, J.X.; Hou, C.Y. Resveratrol Butyrate Esters Inhibit Obesity Caused by Perinatal Exposure to Bisphenol A in Female Offspring Rats. Molecules 2021, 26, 4010. [Google Scholar] [CrossRef] [PubMed]
  176. Zhang, J.; Powell, C.A.; Kay, M.K.; Park, M.H.; Meruvu, S.; Sonkar, R.; Choudhury, M. A moderate physiological dose of benzyl butyl phthalate exacerbates the high fat diet-induced diabesity in male mice. Toxicol. Res. 2020, 9, 353–370. [Google Scholar] [CrossRef] [PubMed]
  177. Buerger, A.N.; Dillon, D.T.; Schmidt, J.; Yang, T.; Zubcevic, J.; Martyniuk, C.J.; Bisesi, J.H., Jr. Gastrointestinal dysbiosis following diethylhexyl phthalate exposure in zebrafish (Danio rerio): Altered microbial diversity, functionality, and network connectivity. Environ. Pollut. 2020, 265, 114496. [Google Scholar] [CrossRef]
  178. Lin, M.H.; Lee, C.Y.; Chuang, Y.S.; Shih, C.L. Exposure to bisphenol A associated with multiple health-related outcomes in humans: An umbrella review of systematic reviews with meta-analyses. Environ. Res. 2023, 237, 116900. [Google Scholar] [CrossRef]
  179. Deodati, A.; Bottaro, G.; Germani, D.; Carli, F.; Tait, S.; Busani, L.; Della Latta, V.; Pala, A.P.; Maranghi, F.; Tassinari, R.; et al. Urinary Bisphenol-A (BPA) and Bis(2-ethylhexyl)phthalate (DEHP) metabolite concentrations in children with obesity: A case-control study. Horm. Res. Paediatr. 2023. [Google Scholar] [CrossRef]
  180. Chen, M.; Lv, C.; Zhang, S.; Tse, L.A.; Hong, X.; Liu, X.; Ding, Y.; Xiao, P.; Tian, Y.; Gao, Y. Bisphenol A substitutes and childhood obesity at 7 years: A cross-sectional study in Shandong, China. Environ. Sci. Pollut. Res. Int. 2023, 30, 73174–73184. [Google Scholar] [CrossRef]
  181. Bi, J.; Wang, F.; Wei, Y.; Zhang, Y.; Jia, C.; He, J.; Yao, J.; Zhang, Z.; Li, Z.; Li, P.; et al. Association of serum bisphenol A levels with incident overweight and obesity risk and the mediating effect of adiponectin. Chemosphere 2022, 308, 136287. [Google Scholar] [CrossRef] [PubMed]
  182. Choi, J.Y.; Lee, J.; Huh, D.A.; Moon, K.W. Urinary bisphenol concentrations and its association with metabolic disorders in the US and Korean populations. Environ. Pollut. 2022, 295, 118679. [Google Scholar] [CrossRef] [PubMed]
  183. Gajjar, P.; Liu, Y.; Li, N.; Buckley, J.P.; Chen, A.; Lanphear, B.P.; Kalkwarf, H.J.; Cecil, K.M.; Yolton, K.; Braun, J.M. Associations of mid-childhood bisphenol A and bisphenol S exposure with mid-childhood and adolescent obesity. Environ. Epidemiol. 2022, 6, e187. [Google Scholar] [CrossRef] [PubMed]
  184. Wu, B.; Jiang, Y.; Jin, X.; He, L. Using three statistical methods to analyze the association between exposure to 9 compounds and obesity in children and adolescents: NHANES 2005-2010. Environ. Health 2020, 19, 94. [Google Scholar] [CrossRef] [PubMed]
  185. Ribeiro, C.M.; Beserra, B.T.S.; Silva, N.G.; Lima, C.L.; Rocha, P.R.S.; Coelho, M.S.; Neves, F.A.R.; Amato, A.A. Exposure to endocrine-disrupting chemicals and anthropometric measures of obesity: A systematic review and meta-analysis. BMJ Open 2020, 10, e033509. [Google Scholar] [CrossRef]
  186. Wu, W.; Li, M.; Liu, A.; Wu, C.; Li, D.; Deng, Q.; Zhang, B.; Du, J.; Gao, X.; Hong, Y. Bisphenol A and the Risk of Obesity a Systematic Review With Meta-Analysis of the Epidemiological Evidence. Dose Response 2020, 18, 1559325820916949. [Google Scholar] [CrossRef]
  187. Jacobson, M.H.; Woodward, M.; Bao, W.; Liu, B.; Trasande, L. Urinary Bisphenols and Obesity Prevalence Among U.S. Children and Adolescents. J. Endocr. Soc. 2019, 3, 1715–1726. [Google Scholar] [CrossRef]
  188. Liu, B.; Lehmler, H.J.; Sun, Y.; Xu, G.; Sun, Q.; Snetselaar, L.G.; Wallace, R.B.; Bao, W. Association of Bisphenol A and Its Substitutes, Bisphenol F and Bisphenol S, with Obesity in United States Children and Adolescents. Diabetes Metab. J. 2019, 43, 59–75. [Google Scholar] [CrossRef]
  189. Zhang, Y.; Dong, T.; Hu, W.; Wang, X.; Xu, B.; Lin, Z.; Hofer, T.; Stefanoff, P.; Chen, Y.; Wang, X.; et al. Association between exposure to a mixture of phenols, pesticides, and phthalates and obesity: Comparison of three statistical models. Environ. Int. 2019, 123, 325–336. [Google Scholar] [CrossRef]
  190. Hao, M.; Ding, L.; Xuan, L.; Wang, T.; Li, M.; Zhao, Z.; Lu, J.; Xu, Y.; Chen, Y.; Wang, W.; et al. Urinary bisphenol A concentration and the risk of central obesity in Chinese adults: A prospective study. J. Diabetes 2018, 10, 442–448. [Google Scholar] [CrossRef]
  191. Do, M.T.; Chang, V.C.; Mendez, M.A.; de Groh, M. Urinary bisphenol A and obesity in adults: Results from the Canadian Health Measures Survey. Health Promot. Chronic Dis. Prev. Can. 2017, 37, 403–412. [Google Scholar] [CrossRef] [PubMed]
  192. Song, Y.; Hauser, R.; Hu, F.B.; Franke, A.A.; Liu, S.; Sun, Q. Urinary concentrations of bisphenol A and phthalate metabolites and weight change: A prospective investigation in US women. Int. J. Obes. 2014, 38, 1532–1537. [Google Scholar] [CrossRef] [PubMed]
  193. Bhandari, R.; Xiao, J.; Shankar, A. Urinary bisphenol A and obesity in U.S. children. Am. J. Epidemiol. 2013, 177, 1263–1270. [Google Scholar] [CrossRef] [PubMed]
  194. Shankar, A.; Teppala, S.; Sabanayagam, C. Urinary bisphenol a levels and measures of obesity: Results from the national health and nutrition examination survey 2003-2008. ISRN Endocrinol. 2012, 2012, 965243. [Google Scholar] [CrossRef] [PubMed]
  195. Trasande, L.; Attina, T.M.; Blustein, J. Association between urinary bisphenol A concentration and obesity prevalence in children and adolescents. Jama 2012, 308, 1113–1121. [Google Scholar] [CrossRef] [PubMed]
  196. 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]
  197. Carwile, J.L.; Michels, K.B. Urinary bisphenol A and obesity: NHANES 2003–2006. Environ. Res. 2011, 111, 825–830. [Google Scholar] [CrossRef] [PubMed]
  198. Stevens, D.R.; Starling, A.P.; Bommarito, P.A.; Keil, A.P.; Nakiwala, D.; Calafat, A.M.; Adgate, J.L.; Dabelea, D.; Ferguson, K.K. Midpregnancy Phthalate and Phenol Biomarkers in Relation to Infant Body Composition: The Healthy Start Prospective Cohort. Environ. Health Perspect. 2023, 131, 87017. [Google Scholar] [CrossRef]
  199. Li, D.; Yao, Y.; Chen, D.; Wu, Y.; Liao, Y.; Zhou, L. Phthalates, physical activity, and diet, which are the most strongly associated with obesity? A case-control study of Chinese children. Endocrine 2023, 82, 69–77. [Google Scholar] [CrossRef]
  200. Wu, Q.; Li, G.; Zhao, C.Y.; Na, X.L.; Zhang, Y.B. Association between phthalate exposure and obesity risk: A meta-analysis of observational studies. Environ. Toxicol. Pharmacol. 2023, 102, 104240. [Google Scholar] [CrossRef]
  201. Boyer, T.M.; Bommarito, P.A.; Welch, B.M.; Meeker, J.D.; James-Todd, T.; Cantonwine, D.E.; McElrath, T.F.; Ferguson, K.K. Maternal exposure to phthalates and total gestational weight gain in the LIFECODES birth cohort. Reprod. Toxicol. 2023, 117, 108354. [Google Scholar] [CrossRef] [PubMed]
  202. Vieyra, G.; Hankinson, S.E.; Oulhote, Y.; Vandenberg, L.N.; Tinker, L.; Manson, J.E.; Shadyab, A.H.; Thomson, C.A.; Bao, W.; Allison, M.; et al. Association between urinary phthalate biomarker concentrations and adiposity among postmenopausal women. Environ. Res. 2023, 222, 115356. [Google Scholar] [CrossRef]
  203. Milankov, A.; Milanović, M.; Milošević, N.; Sudji, J.; Pejaković, S.; Milić, N.; Bjelica, A.; Medić Stojanoska, M. The effects of phthalate exposure on metabolic parameters in polycystic ovary syndrome. Clin. Chim. Acta 2023, 540, 117225. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, Z.H.; Gao, D.; Zou, Z.Y. The association of phthalate metabolites with childhood waist circumference and abdominal obesity. Eur. J. Pediatr. 2023, 182, 803–812. [Google Scholar] [CrossRef] [PubMed]
  205. Peng, M.Q.; Karvonen-Gutierrez, C.A.; Herman, W.H.; Mukherjee, B.; Park, S.K. Phthalate exposure is associated with more rapid body fat gain in midlife women: The Study of Women’s Health Across the Nation (SWAN) Multi-Pollutant Study. Environ. Res. 2023, 216, 114685. [Google Scholar] [CrossRef] [PubMed]
  206. Kupsco, A.; Wu, H.; Calafat, A.M.; Kioumourtzoglou, M.A.; Cantoral, A.; Tamayo-Ortiz, M.; Pantic, I.; Pizano-Zárate, M.L.; Oken, E.; Braun, J.M.; et al. Prenatal maternal phthalate exposures and trajectories of childhood adiposity from four to twelve years. Environ. Res. 2022, 204, 112111. [Google Scholar] [CrossRef] [PubMed]
  207. Ribeiro, C.; Mendes, V.; Peleteiro, B.; Delgado, I.; Araújo, J.; Aggerbeck, M.; Annesi-Maesano, I.; Sarigiannis, D.; Ramos, E. Association between the exposure to phthalates and adiposity: A meta-analysis in children and adults. Environ. Res. 2019, 179, 108780. [Google Scholar] [CrossRef]
  208. Díaz Santana, M.V.; Hankinson, S.E.; Bigelow, C.; Sturgeon, S.R.; Zoeller, R.T.; Tinker, L.; Manson, J.A.E.; Calafat, A.M.; Meliker, J.R.; Reeves, K.W. Urinary concentrations of phthalate biomarkers and weight change among postmenopausal women: A prospective cohort study. Environ. Health 2019, 18, 20. [Google Scholar] [CrossRef]
  209. Rodríguez-Carmona, Y.; Cantoral, A.; Trejo-Valdivia, B.; Téllez-Rojo, M.M.; Svensson, K.; Peterson, K.E.; Meeker, J.D.; Schnaas, L.; Solano, M.; Watkins, D.J. Phthalate exposure during pregnancy and long-term weight gain in women. Environ. Res. 2019, 169, 26–32. [Google Scholar] [CrossRef]
  210. Buckley, J.P.; Engel, S.M.; Mendez, M.A.; Richardson, D.B.; Daniels, J.L.; Calafat, A.M.; Wolff, M.S.; Herring, A.H. Prenatal Phthalate Exposures and Childhood Fat Mass in a New York City Cohort. Environ. Health Perspect. 2016, 124, 507–513. [Google Scholar] [CrossRef]
  211. Valvi, D.; Casas, M.; Romaguera, D.; Monfort, N.; Ventura, R.; Martinez, D.; Sunyer, J.; Vrijheid, M. Prenatal Phthalate Exposure and Childhood Growth and Blood Pressure: Evidence from the Spanish INMA-Sabadell Birth Cohort Study. Environ. Health Perspect. 2015, 123, 1022–1029. [Google Scholar] [CrossRef]
  212. Yaghjyan, L.; Sites, S.; Ruan, Y.; Chang, S.H. Associations of urinary phthalates with body mass index, waist circumference and serum lipids among females: National Health and Nutrition Examination Survey 1999-2004. Int. J. Obes. 2015, 39, 994–1000. [Google Scholar] [CrossRef]
  213. Lind, P.M.; Roos, V.; Rönn, M.; Johansson, L.; Ahlström, H.; Kullberg, J.; Lind, L. Serum concentrations of phthalate metabolites are related to abdominal fat distribution two years later in elderly women. Environ. Health 2012, 11, 21. [Google Scholar] [CrossRef]
  214. Hatch, E.E.; Nelson, J.W.; Qureshi, M.M.; Weinberg, J.; Moore, L.L.; Singer, M.; Webster, T.F. Association of urinary phthalate metabolite concentrations with body mass index and waist circumference: A cross-sectional study of NHANES data, 1999–2002. Environ. Health 2008, 7, 27. [Google Scholar] [CrossRef]
  215. Kim, K.Y.; Lee, E.; Kim, Y. The Association between Bisphenol A Exposure and Obesity in Children-A Systematic Review with Meta-Analysis. Int. J. Environ. Res. Public. Health 2019, 16, 2521. [Google Scholar] [CrossRef]
  216. Zhou, Z.; Lei, Y.; Wei, W.; Zhao, Y.; Jiang, Y.; Wang, N.; Li, X.; Chen, X. Association between prenatal exposure to bisphenol a and birth outcomes: A systematic review with meta-analysis. Medicine 2019, 98, e17672. [Google Scholar] [CrossRef]
  217. Hu, C.Y.; Li, F.L.; Hua, X.G.; Jiang, W.; Mao, C.; Zhang, X.J. The association between prenatal bisphenol A exposure and birth weight: A meta-analysis. Reprod. Toxicol. 2018, 79, 21–31. [Google Scholar] [CrossRef]
  218. Vrachnis, N.; Loukas, N.; Vrachnis, D.; Antonakopoulos, N.; Zygouris, D.; Kοlialexi, A.; Pergaliotis, V.; Iavazzo, C.; Mastorakos, G.; Iliodromiti, Z. A Systematic Review of Bisphenol A from Dietary and Non-Dietary Sources during Pregnancy and Its Possible Connection with Fetal Growth Restriction: Investigating Its Potential Effects and the Window of Fetal Vulnerability. Nutrients 2021, 13, 2426. [Google Scholar] [CrossRef]
  219. Soundararajan, A.; Prabu, P.; Mohan, V.; Gibert, Y.; Balasubramanyam, M. Novel insights of elevated systemic levels of bisphenol-A (BPA) linked to poor glycemic control, accelerated cellular senescence and insulin resistance in patients with type 2 diabetes. Mol. Cell Biochem. 2019, 458, 171–183. [Google Scholar] [CrossRef]
  220. Shankar, A.; Teppala, S. Relationship between urinary bisphenol A levels and diabetes mellitus. J. Clin. Endocrinol. Metab. 2011, 96, 3822–3826. [Google Scholar] [CrossRef] [PubMed]
  221. Kawa, I.A.; Masood, A.; Ganie, M.A.; Fatima, Q.; Jeelani, H.; Manzoor, S.; Rizvi, S.M.; Muzamil, M.; Rashid, F. Bisphenol A (BPA) acts as an endocrine disruptor in women with Polycystic Ovary Syndrome: Hormonal and metabolic evaluation. Obes. Med. 2019, 14, 100090. [Google Scholar] [CrossRef]
  222. Jiang, S.; Liu, H.; Zhou, S.; Zhang, X.; Peng, C.; Zhou, H.; Tong, Y.; Lu, Q. Association of bisphenol A and its alternatives bisphenol S and F exposure with hypertension and blood pressure: A cross-sectional study in China. Environ. Pollut. 2020, 257, 113639. [Google Scholar] [CrossRef] [PubMed]
  223. Wang, B.; Wang, S.; Zhao, Z.; Chen, Y.; Xu, Y.; Li, M.; Xu, M.; Wang, W.; Ning, G.; Bi, Y.; et al. Bisphenol A exposure in relation to altered lipid profile and dyslipidemia among Chinese adults: A repeated measures study. Environ. Res. 2020, 184, 109382. [Google Scholar] [CrossRef] [PubMed]
  224. Moghaddam, H.S.; Samarghandian, S.; Farkhondeh, T. Effect of bisphenol A on blood glucose, lipid profile and oxidative stress indices in adult male mice. Toxicol. Mech. Methods 2015, 25, 507–513. [Google Scholar] [CrossRef]
  225. Rochester, J.R.; Bolden, A.L. Bisphenol S and F: A Systematic Review and Comparison of the Hormonal Activity of Bisphenol A Substitutes. Environ. Health Perspect. 2015, 123, 643–650. [Google Scholar] [CrossRef]
  226. van der Meer, T.P.; Thio, C.H.L.; van Faassen, M.; van Beek, A.P.; Snieder, H.; van Berkum, F.N.R.; Kema, I.P.; Makris, K.C.; Wolffenbuttel, B.H.R.; van Vliet-Ostaptchouk, J.V. Endocrine disrupting chemicals during diet-induced weight loss-A post-hoc analysis of the LOWER study. Environ. Res. 2021, 192, 110262. [Google Scholar] [CrossRef]
  227. Lee, D.W.; Lim, H.M.; Lee, J.Y.; Min, K.B.; Shin, C.H.; Lee, Y.A.; Hong, Y.C. Prenatal exposure to phthalate and decreased body mass index of children: A systematic review and meta-analysis. Sci. Rep. 2022, 12, 8961. [Google Scholar] [CrossRef]
  228. LaKind, J.S.; Idri, F.; Naiman, D.Q.; Verner, M.A. Biomonitoring and Nonpersistent Chemicals-Understanding and Addressing Variability and Exposure Misclassification. Curr. Environ. Health Rep. 2019, 6, 16–21. [Google Scholar] [CrossRef]
  229. Guo, Y.; Kannan, K. Challenges encountered in the analysis of phthalate esters in foodstuffs and other biological matrices. Anal. Bioanal. Chem. 2012, 404, 2539–2554. [Google Scholar] [CrossRef]
  230. Ye, X.; Zhou, X.; Hennings, R.; Kramer, J.; Calafat, A.M. Potential external contamination with bisphenol A and other ubiquitous organic environmental chemicals during biomonitoring analysis: An elusive laboratory challenge. Environ. Health Perspect. 2013, 121, 283–286. [Google Scholar] [CrossRef]
  231. Cerkvenik-Flajs, V. Bisphenol A background contamination encountered during advanced blood sampling and laboratory analysis. Int. J. Environ. Anal. Chem. 2022, 102, 6602–6612. [Google Scholar] [CrossRef]
  232. vom Saal, F.S.; Welshons, W.V. Evidence that bisphenol A (BPA) can be accurately measured without contamination in human serum and urine, and that BPA causes numerous hazards from multiple routes of exposure. Mol. Cell Endocrinol. 2014, 398, 101–113. [Google Scholar] [CrossRef] [PubMed]
  233. Zota, A.R.; Phillips, C.A.; Mitro, S.D. Recent Fast Food Consumption and Bisphenol A and Phthalates Exposures among the U.S. Population in NHANES, 2003-2010. Environ. Health Perspect. 2016, 124, 1521–1528. [Google Scholar] [CrossRef] [PubMed]
  234. Dales, R.E.; Kauri, L.M.; Cakmak, S. The associations between phthalate exposure and insulin resistance, β-cell function and blood glucose control in a population-based sample. Sci. Total Environ. 2018, 612, 1287–1292. [Google Scholar] [CrossRef] [PubMed]
  235. Ko, N.Y.; Lo, Y.C.; Huang, P.C.; Huang, Y.C.; Chang, J.L.; Huang, H.B. Changes in insulin resistance mediate the associations between phthalate exposure and metabolic syndrome. Environ. Res. 2019, 175, 434–441. [Google Scholar] [CrossRef] [PubMed]
  236. 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] [PubMed]
  237. 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]
  238. Rancière, F.; Botton, J.; Slama, R.; Lacroix, M.Z.; Debrauwer, L.; Charles, M.A.; Roussel, R.; Balkau, B.; Magliano, D.J. 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]
  239. Yao, J.; Wang, F.; Zhang, Y.; Zhang, Z.; Bi, J.; He, J.; Li, P.; Han, X.; Wei, Y.; Zhang, X.; et al. Association of serum BPA levels with changes in lipid levels and dyslipidemia risk in middle-aged and elderly Chinese. Ecotoxicol. Environ. Saf. 2022, 241, 113819. [Google Scholar] [CrossRef]
  240. Gao, D.; Zou, Z.; Li, Y.; Chen, M.; Ma, Y.; Chen, L.; Wang, X.; Yang, Z.; Dong, Y.; Ma, J. Association between urinary phthalate metabolites and dyslipidemia in children: Results from a Chinese cohort study. Environ. Pollut. 2022, 295, 118632. [Google Scholar] [CrossRef]
  241. Han, C.; Hong, Y.C. Bisphenol A, Hypertension, and Cardiovascular Diseases: Epidemiological, Laboratory, and Clinical Trial Evidence. Curr. Hypertens. Rep. 2016, 18, 11. [Google Scholar] [CrossRef] [PubMed]
  242. Shankar, A.; Teppala, S. Urinary bisphenol A and hypertension in a multiethnic sample of US adults. J. Environ. Public Health 2012, 2012, 481641. [Google Scholar] [CrossRef]
  243. Liguori, F.; Moreno-Marrodan, C.; Barbaro, P. Biomass-derived chemical substitutes for bisphenol A: Recent advancements in catalytic synthesis. Chem. Soc. Rev. 2020, 49, 6329–6363. [Google Scholar] [CrossRef] [PubMed]
  244. Moreno-Gómez-Toledano, R. Relationship between emergent BPA-substitutes and renal and cardiovascular diseases in adult population. Environ. Pollut. 2022, 313, 120106. [Google Scholar] [CrossRef] [PubMed]
  245. Lu, Y.; Chen, S.; Jin, H.; Tang, L.; Xia, M. Associations of bisphenol F and S, as substitutes for bisphenol A, with cardiovascular disease in American adults. J. Appl. Toxicol. 2023, 43, 500–507. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, R.; Fei, Q.; Liu, S.; Weng, X.; Liang, H.; Wu, Y.; Wen, L.; Hao, G.; Cao, G.; Jing, C. The bisphenol F and bisphenol S and cardiovascular disease: Results from NHANES 2013–2016. Environ. Sci. Eur. 2022, 34, 4. [Google Scholar] [CrossRef]
  247. Langsch, A.; David, R.M.; Schneider, S.; Sperber, S.; Haake, V.; Kamp, H.; Leibold, E.; Ravenzwaay, B.V.; Otter, R. Hexamoll® DINCH: Lack of in vivo evidence for obesogenic properties. Toxicol. Lett. 2018, 288, 99–110. [Google Scholar] [CrossRef] [PubMed]
  248. Bui, T.T.; Giovanoulis, G.; Cousins, A.P.; Magnér, J.; Cousins, I.T.; de Wit, C.A. Human exposure, hazard and risk of alternative plasticizers to phthalate esters. Sci. Total Environ. 2016, 541, 451–467. [Google Scholar] [CrossRef]
  249. Diamanti-Kandarakis, E.; Piouka, A.; Livadas, S.; Piperi, C.; Katsikis, I.; Papavassiliou, A.G.; Panidis, D. Anti-mullerian hormone is associated with advanced glycosylated end products in lean women with polycystic ovary syndrome. Eur. J. Endocrinol. 2009, 160, 847–853. [Google Scholar] [CrossRef]
  250. Saleh, A.C.; Sabry, R.; Mastromonaco, G.F.; Favetta, L.A. BPA and BPS affect the expression of anti-Mullerian hormone (AMH) and its receptor during bovine oocyte maturation and early embryo development. Reprod. Biol. Endocrinol. 2021, 19, 119. [Google Scholar] [CrossRef]
  251. Talia, C.; Connolly, L.; Fowler, P.A. The insulin-like growth factor system: A target for endocrine disruptors? Environ. Int. 2021, 147, 106311. [Google Scholar] [CrossRef] [PubMed]
  252. Papatsoris, A.G.; Karamouzis, M.V.; Papavassiliou, A.G. Novel insights into the implication of the IGF-1 network in prostate cancer. Trends Mol. Med. 2005, 11, 52–55. [Google Scholar] [CrossRef] [PubMed]
  253. Heindel, J.J.; Blumberg, B.; Cave, M.; Machtinger, R.; Mantovani, A.; Mendez, M.A.; Nadal, A.; Palanza, P.; Panzica, G.; Sargis, R.; et al. Metabolism disrupting chemicals and metabolic disorders. Reprod. Toxicol. 2017, 68, 3–33. [Google Scholar] [CrossRef] [PubMed]
  254. Wagner, M.; Schlüsener, M.P.; Ternes, T.A.; Oehlmann, J. Identification of putative steroid receptor antagonists in bottled water: Combining bioassays and high-resolution mass spectrometry. PLoS ONE 2013, 8, e72472. [Google Scholar] [CrossRef]
Figure 1. Obesogenic endocrine-disrupting chemicals. Abbreviations: PBDEs: polybrominated diphenyl ethers; PCBs: polychlorinated biphenyls; PFAs: perfluoroalkyl substances. All images are originated from the free medical site http://smart.servier.com/ (accessed on 1 December 2023) by Servier licensed under a Creative Commons Attribution 3.0 Unported License.
Figure 1. Obesogenic endocrine-disrupting chemicals. Abbreviations: PBDEs: polybrominated diphenyl ethers; PCBs: polychlorinated biphenyls; PFAs: perfluoroalkyl substances. All images are originated from the free medical site http://smart.servier.com/ (accessed on 1 December 2023) by Servier licensed under a Creative Commons Attribution 3.0 Unported License.
Ijms 25 00675 g001
Figure 2. Worldwide plastic production in million metric tons and prevalence of excess body weight (BMI: >25 kg/m2) in percentage (%). Figure is based on data from [1,36,37].
Figure 2. Worldwide plastic production in million metric tons and prevalence of excess body weight (BMI: >25 kg/m2) in percentage (%). Figure is based on data from [1,36,37].
Ijms 25 00675 g002
Figure 3. Chemical structure of bisphenol A (left) and phthalates (right).
Figure 3. Chemical structure of bisphenol A (left) and phthalates (right).
Ijms 25 00675 g003
Figure 4. Main mechanisms linking endocrine-disrupting chemicals such as BPA and phthalates to obesity. Abbreviations: AgRP: Agouti-related peptide; α-MSH: α-melanocyte-stimulating hormone; BPA: bisphenol A; C/EBP: CCAAT/enhancer-binding family of proteins; EDCs: endocrine-disrupting chemicals; IL: interleukin; LPS: lipopolysaccharide; MSCs: mesenchymal stem cells; NPY: neuropeptide Y; POMC: proopiomelanocortin; ROS: reactive oxygen species; SCFA: small-chain fatty acid; TNF-α: tumor necrosis factor-α. All images are originated from the free medical site http://smart.servier.com/ (accessed on 1 December 2023) by Servier licensed under a Creative Commons Attribution 3.0 Unported License.
Figure 4. Main mechanisms linking endocrine-disrupting chemicals such as BPA and phthalates to obesity. Abbreviations: AgRP: Agouti-related peptide; α-MSH: α-melanocyte-stimulating hormone; BPA: bisphenol A; C/EBP: CCAAT/enhancer-binding family of proteins; EDCs: endocrine-disrupting chemicals; IL: interleukin; LPS: lipopolysaccharide; MSCs: mesenchymal stem cells; NPY: neuropeptide Y; POMC: proopiomelanocortin; ROS: reactive oxygen species; SCFA: small-chain fatty acid; TNF-α: tumor necrosis factor-α. All images are originated from the free medical site http://smart.servier.com/ (accessed on 1 December 2023) by Servier licensed under a Creative Commons Attribution 3.0 Unported License.
Ijms 25 00675 g004
Table 3. Major studies depicting associations between endocrine disruptors and obesity.
Table 3. Major studies depicting associations between endocrine disruptors and obesity.
Author, YearStudy Design/PopulationMain FindingsComments
Bisphenol A and obesity
Lin et al. (2023) [178]Umbrella review of systematic reviews with meta-analyses on the association of BPA exposure with multiple outcomes, including obesity- Higher BPA exposure significantly associated with obesity risk in both sexes (females: OR 1.51; males: OR 1.88)
- Significant associations with generalized and abdominal obesity (OR 1.22 and 1.41, respectively) as well as overweight in adults (OR 1.25)
- Significantly increased risk for type 2 DM (OR 1.28)
Higher BPA exposure associated with obesity in children and adults, with less heterogeneity among studies in females
Deodati et al. (2023) [179]Case–control study among n = 122 children (n = 66 and n = 56 with and without obesity, respectively) matched for age and gender- Significantly higher creatinine-adjusted urinary BPA concentrations in obesity than normal weight (10.77 vs. 5.50 µg/g, respectively) among girls, but not boys.
- Significantly higher risk of obesity in children with BPA levels above the median eating packaged food (OR = 11.09)
Potential gender-specific relationship between BPA exposure and higher odds of childhood obesity in girls
Chen M et al. (2023) [180]Cross-sectional study among n = 426 children aged 7 years old- Urinary concentrations of BPA substitutes BPS, BPAF exhibit a significant positive association with BMI, WC, overweight/obesity only among boys
- No associations between adiposity measures and BPA or other substitute compounds
Associations between BPS, BPAF, but not BPA exposure and obesity in boys
Bi J et al. (2022) [181]Prospective observational study including n = 796 individuals with normal weight, among whom 133 developed overweight or obesity during follow-up- Presence of a statistically significant inverted U-shaped relationship between serum BPA and incident overweight/obesity
- Significant positive correlation between log10-BPA and increase in waist-to-hip ratio
- Serum adiponectin mediates 46% of association between BPA and incident overweight/obesity
Non-monotonic relationship between baseline BPA and incident overweight/obesity among individuals with normal weight, potentially indirectly mediated by adiponectin
Choi et al. (2022) [182]Cross-sectional study including 1046 adult participants in NHANES (2013–2016) and 3268 adult participants of the Korean National Environmental Health Survey (2015–2017) Those in the higher urinary BPA tertiles had significantly higher odds for obesity (OR = 1.58 and 1.41 for 3rd and 2nd vs. 1st tertile, respectively)
Similar associations for urinary BPF and BPS
Exposure not only to BPA but also to substitutes BPF and BPS is associated with adult obesity
Gajjar et al. (2021) [183]Prospective observational cohort of n = 212 children with urinary BPA and BPS measurements at 8 years and body composition assessments at 8 years (bioimpedance) and 12 years (DXA)No evidence for a synchronous or prospective association of urinary BPA or BPS with increased adiposity
Wu et al. (2020) [184]Cross-sectional study including n = 2372 children and adolescents (aged 6–19) participating in NHANESBPA levels significantly associated with higher weight in a statistical approach implementing weighted quantile sum statistical model but not in other approachesEvidence for an association between BPA exposure and childhood/adolescent obesity
Ribeiro et al. (2020) [185]Meta-analysis of studies investigating BPA exposure and multiple adverse health outcomes, including obesity- BPA is significantly associated with overweight (OR 1.254), obesity (OR 1.503) and increased WC (OR 1.503) in adults
- OR 1.8 for childhood obesity
Positive association between BPA exposure and generalized as well as abdominal obesity
Wu et al. (2020) [186]Meta-analysis of 10 observational studies - Statistically significant dose–response positive relationship between BPA and overweight/obesity risk in both sexes
- 11% increase in obesity risk for every 1 ng/mL of BPA
Continuous positive relationship between BPA and obesity irrespective of sex
Jacobson et al. (2019) [187]Cross-sectional study including n = 1831 children and adolescents (aged 6–19) participating in NHANESUrinary BPS and BPF but not total bisphenols or BPA were significantly associated with, particularly abdominal obesity Substitute bisphenol exposure may predispose individuals to childhood/adolescent obesity
Liu et al. (2019) [188]Cross-sectional study including n = 745 children and adolescents (aged 6–17 years) participating in NHANESUrinary BPA (OR 1.74) and BPF (OR 1.54) are significantly associated with obesity, with stronger associations between boys
Similar findings for abdominal obesity
Urinary BPA and its substitute BPF associated with obesity, particularly in boys
Zhang et al. (2019) [189]Cross-sectional study including n = 1269 adults participating in NHANESAmong other chemicals, increased urinary BPA and BPS are significantly associated with higher obesity prevalence Exposure to BPA, and BPS may predispose individuals to adulthood obesity, although the authors recommend considering the joint effects of different chemical exposures
Hao et al. (2018) [190]Prospective study (mean follow-up: 4 years) among 888 Chinese adults without abdominal obesity at baseline OR = 2.30 for incident abdominal obesity each unit increase in log [BPA] urinary concentration after adjustment for confounding factors
Individuals in the lowest tertile of BPA concentrations had the lowest risk for incident central obesity (ORs 1.73 and 1.81 for those in the 2nd and 3rd tertiles, respectively)
Prospective association of BPA exposure with incident central obesity in Chinese adults
Do et al. (2017) [191]Cross-sectional analysis of data from n = 4733 adults aged (18 to 79 years)For each natural-log unit increase in urinary BPA concentration, significant increase of 0.33 kg/m2 in BMI and 1.00 cm in waist circumferenceDose–response relationship between BPA exposure and generalized as well as abdominal obesity
Song et al. (2014) [192]Prospective (10 years) cohort study of 977 women with baseline measurements of urinary BPA and 9 phthalate biomarkersAfter adjustment for dietary and lifestyle variables, those in the highest BPA quartile gained on average an additional 0.23 kg/year (0.07–0.38) of body weight during follow-upBPA exposure is associated with greater longitudinal weight gain in women
Bhandari et al. (2013) [193]Cross-sectional analysis of data from n = 2200 children and adolescents (aged 6 to 18 years) from NHANES (2003–2008)- OR for obesity = 2.55 for children in the highest vs. lowest quartile of urinary BPA
- Associations more robust among males and non-Hispanic Whites
BPA is associated with childhood/adolescent obesity, with potential gender- and race-specific effects
Shankar et al. (2012) [194]Cross-sectional analysis of data from n = 3967 adult participants in NHANES (2003–2008)ORs = 1.69 and 1.59 for generalized and abdominal obesity for the 4th vs. 1st quartile of urinary BPA concentrations, persistent after adjustment for several confounders and consistent among gender and race–ethnic groups BPA exposure is associated with central and abdominal obesity in both genders and all race groups, irrespectively of traditional risk factors
Trasande et al. (2012) [195]Cross-sectional analysis of data from n = of 2838 children and adolescents (aged 6–19 years) participating in NHANES- Lowest prevalence of obesity in the 1st vs. 2nd-4th quartiles of ascending urinary BPA concentrations (10.3% vs. 20.1%, 19.0% and 22.3%, respectively)
- Association of BPA and obesity significant in Whites but not Blacks or Hispanics
Association between BPA exposure and obesity likely exhibits race-specific effects
Wang et al. (2012) [196]Cross-sectional study of n = 3390, aged >40 years- Those in the highest quartile of urinary BPA concentrations showed significantly higher prevalence of generalized (OR = 1.50) and abdominal obesity (OR = 1.28)
- Among participants without overweight or obesity, higher BPA was significantly associated with IR (OR 1.94)
Evidence for a positive association between BPA exposure and obesity, as well as IR among lean individuals
Carwile et al. (2011) [197]Cross-sectional study including n = 2747 adults (aged 18–74) participating in NHANES (2003–2006)Higher risk of general (OR 1.74) and abdominal (OR 1.58) obesity among individuals in the highest vs. lowest quartile of urinary BPA concentration BPA exposure is associated with general and abdominal obesity in US adults
Phthalates and obesity
Deodati et al. (2023) [179]Case–control study among n = 122 children (n = 66 and n = 56 with and without obesity, respectively) matched for age and gender- Early downstream metabolites of Di(2-ethylhexyl) phthalate in urine significantly higher in girls with obesity than normal weight
- Significant positive correlation of Di(2-ethylhexyl) phthalate metabolites with serum leptin levels
Significant correlation of certain phthalate metabolites with increased adiposity in girls
Stevens DR et al. (2023) [198]Prospective study among n = 438 infants from the Healthy Start prospective pregnancy cohort.- Significant inverse association between maternal urinary mono-benzyl and di-
n-butyl phthalate at 28th gestational week and percentage fat mass at birth in male infants
Maternal phthalate exposure in pregnancy is inversely associated with fat mass in male, but not female, infants at birth
Li et al. (2023) [199]Case–control study among n = 240 children with overweight/obesity (OBE) and n = 240 age- and gender-matched controlsAmong 9 phthalates, monomethyl phthalate and monobutyl phthalate were significantly higher in controls than children with overweight/obesity but not after adjustment for physical activity and caloric intake.No significant differences in phthalate concentrations between OBE and controls
Wu et al. (2022) [200]Meta-analysis of observational studies for the association between phthalate compounds and obesity in adult and pediatric populations - Mono-n-butyl-, monobutyl-, monoisobutyl-, monoethyl- and mono(2-ethyl-5-carboxypentyl) phthalate significantly associated with obesity, specific compounds more strongly correlate with general or abdominal obesity
- Stronger associations in women and in studies from the United States and Europe
Compound-specific effects on general and abdominal obesity, with potential gender- and study-site-specific effects
Boyer et al. (2023) [201]Measurement of the concentrations of 9 phthalates in n = 379 pregnant women, in relation to gestational weight gain (difference between pre-pregnancy and median 35.1 weeks weight)Significant direct association between mono-(3-carboxypropyl) phthalate and mono-n-butyl phthalate was positively associated with gestational weight gain (1.81 kg and 0.77 kg at 35 weeks) interquartile range increase among women with obesityPhthalate exposure is associated with greater weight gain in pregnancy, particularly among women with obesity at baseline
Vieyra et al. (2023) [202]Prospective observational study n = 1125 participants of the Woman Health Initiative (WHI) with available urine phthalate measurements and DXA-based estimations of VAT and SAT Significant positive associations of baseline di-isobutyl phthalate biomarkers, monocarboxy-isononly phthalate, and di(2-ethylhexyl) phthalate with VAT three years later, which persisted after adjustment for SATHigher levels of certain urinary phthalate compounds are longitudinally associated with higher VAT over time in postmenopausal women
Milankov et al. (2023) [203]Cross-sectional study among n = 60 women with PCOSTotal urinary phthalate concentrations significantly positively correlate with BMI, waist circumference, waist-to-height ratio, VAI, FPG and HOMA-RIncreased phthalate exposure associated with obesity, insulin resistance and hyperglycemia in women with PCOS
Wang et al. (2023) [204]Cross-sectional study among n = 798 students (7–10 years)Significantly increased risk of abdominal obesity for the fourth vs. first quartile (OR = 5.29 and 3.73) and 273% (OR = 3.73; 95% CI: 1.57, 8.86) of urinary concentrations of monoethyl phthalate and monoisobutyl phthalateMonoethyl-phthalate and monoisobutyl-phthalate exposure are associated with abdominal obesity in children
Peng et al. (2023) [205]Prospective observational analysis of n = 1369 women in the Study of Women’s Health Across the Nation Multi-Pollutant StudySignificantly higher levels of spot urinary phthalates (except mono-carboxy-isononyl phthalate) were associated with faster increases in body fat percentage and fat mass, but not total body weight change over time Urinary phthalate concentrations positively correlated with fat gain in middle-aged women
Kupsko et al. (2022) [206]Prospective observational study of 514 mother–child pairs in pregnancy until twelve years post-termHigher maternal urine di (2-ethylhexyl) phthalate metabolites significantly associated with greater odds of high and increasing weight in infants
Higher di-isononyl phthalate metabolites significantly associated with greater odds increasing weight in infants
Exposure to certain phthalates during pregnancy exerts a significant impact in infant weight trajectories during childhood
Ribeiro et al. (2019) [207]Meta-analysis of 29 studies for the association between phthalate compounds and obesity in adult and pediatric populations - The low number of studies for many phthalate compounds precludes meta-analysis
- Statistically significant association solely between mono(2-ethyl-5-carboxypentyl) phthalate and obesity in adults (OR = 1.67)
Positive association between many phthalate compounds and adiposity measures, most formally non-significant; possible publication-bias-related effects
Díaz Santana et al. (2019) [208]Cross-sectional (n = 997) and prospective (n = 660) observational study among participants of the Woman Health Initiative (WHI)- Significant positive associations between urinary phthalate biomarker concentrations and obesity in cross-sectional analysis
- Baseline urinary mono-(2-ethyl-5-oxohexyl)-, monoethyl-, mono-hydroxybutyl- and mono-hydroxyisobutyl phthalate significantly correlate with weight gain after 3 years
- No associations with weight changes at 6 years
Exposure to certain phthalates may predispose individuals to obesity and short-term weight gain
Rodriguez-Carmona, et al. (2019) [209]Prospective cohort study among n = 178 pregnant womenHigher urinary mono-3-carboxypropyl pthalate is significantly associated with moderately increased weight gain over the next 5.2–10.7 years
Higher mono-benzyl phthalate significantly associated with lower weight gain in the same timeframe
Prospective association between phthalate exposure in pregnancy and prospective weight changes in women
Buckley et al. (2016) [210]Prospective cohort study assessing the fat mass of n = 180 children (4–9 years) in relation to maternal third-trimester urinary phthalate concentrations in pregnancy- No continuous associations between maternal urinary phthalate concentrations and fat mass in offspring, without apparent gender-specific effects
- 3.06% lower fat mass in children in the highest vs. lowest quartile of summed di(2-ethylhexyl) phthalate metabolites
No evidence for an impact of maternal phthalate exposure and increased infantile fat mass
Valvi et al. (2015) [211]Prospective cohort study of n = 391 mothers with creatinine-adjusted measurements of urinary phthalates in the 1st and 3rd trimesters of pregnancyHigh-molecular-weight phthalate metabolites in maternal urine significantly associated with lower BMI z-scores in boys and higher in girls 4–7 years of agePotential gender-specific effects of maternal phthalate exposure and infantile BMI trajectories
Yaghjyan et al. (2015) [212]Cross-sectional analysis including n = 6005 women without diabetes participating in NHANES (1999–2004)- Significant positive associations between monobutylphthalate, mono-2-ethylhexyl- to mono(2-ethyl-5-hydroxyhexyl) phthalate ratio and BMI, WCAmong the observed associations, the higher mono-2-ethylhexyl- to mono(2-ethyl-5-hydroxyhexyl) phthalate ratio may be reflective of slower oxidative metabolism of mono-2-ethylhexyl-pthalate
Song et al. (2014) [192]Prospective cohort study of n = 977 women with baseline measurements of urinary BPA and 9 phthalate biomarkersAfter adjustment for dietary and lifestyle variables, significant albeit moderate positive dose–response relationship between phthalic acid, monobenzyl- and monobutyl-phthalate and weight gain over 10 yearsExposure to certain phthalates is associated with accelerated weight gain in women
Lind et al. (2012) [213]Prospective cohort study among n = 1016 individuals of 70 years of ageBaseline serum concentrations of mono-isobutyl phthalate and mono-methyl phthalate significantly positively associated with DXA- and abdominal MRI-derived indices of adiposity in women two years later, but not in menCirculating concentrations of certain phthalates are associated with increased adiposity only in women, suggesting possible sex-specific associations of phthalates with obesity
Hatch et al. (2008) [214]Cross-sectional analysis of data of n = 4369 NHANES participants (1999–2002)- Positive trends of BMI and WC across quartiles of concentrations of mono-benzyl, mono-2-ethyl-5-oxohexyl, mono-ethyl, mono-n-butyl, mono-2-ethyl-5-hydroxyhexyl, particularly among males 20–59 years old
- Similar trends across mono-ethyl phthalate quartiles in adolescent girls, less strong in adult women
- Inverse trend for mono-2-ethylhexyl phthalate in adolescent girls and several inverse associations in adults 60–80 years
- No associations in children
Exposure to several phthalates may be associated with increased adiposity, but age-group- and gender-specific effects likely exist
Stahlhut et al. (2007) [78]Cross-sectional analysis of data of n = 1443 men participating in NHANES (1999–2002)- Urinary concentrations of monobenzylphthalate, mono(2-ethyl-5-hydroxyhexyl) phthalate, mono(2-ethyl-5-oxohexyl) phthalate and monoethylphthalate are significantly associated with increased waist circumference, after adjustment for confounders
- Monobutylphthalate, monobenzylphthalate and monoethylphthalate concentrations exhibit significant positive correlations with HOMA-R
Exposure to certain phthalates is associated with higher abdominal obesity and insulin resistance
Abbreviations: BMI: body mass index; BPA/BPF/BPS: bisphenol A, F and S; DXA: dual X-ray absorptiometry; FPG: fasting plasma glucose; HOMA-R: Homeostatic Model Assessment for Insulin Resistance; NHANES: National Health and Nutrition Examination Survey; PCOS: polycystic ovary syndrome; SAT: subcutaneous adipose tissue; VAI: visceral adiposity index; VAT: visceral adipose tissue, WC: waist circumference.
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

Dalamaga, M.; Kounatidis, D.; Tsilingiris, D.; Vallianou, N.G.; Karampela, I.; Psallida, S.; Papavassiliou, A.G. The Role of Endocrine Disruptors Bisphenols and Phthalates in Obesity: Current Evidence, Perspectives and Controversies. Int. J. Mol. Sci. 2024, 25, 675. https://doi.org/10.3390/ijms25010675

AMA Style

Dalamaga M, Kounatidis D, Tsilingiris D, Vallianou NG, Karampela I, Psallida S, Papavassiliou AG. The Role of Endocrine Disruptors Bisphenols and Phthalates in Obesity: Current Evidence, Perspectives and Controversies. International Journal of Molecular Sciences. 2024; 25(1):675. https://doi.org/10.3390/ijms25010675

Chicago/Turabian Style

Dalamaga, Maria, Dimitrios Kounatidis, Dimitrios Tsilingiris, Natalia G. Vallianou, Irene Karampela, Sotiria Psallida, and Athanasios G. Papavassiliou. 2024. "The Role of Endocrine Disruptors Bisphenols and Phthalates in Obesity: Current Evidence, Perspectives and Controversies" International Journal of Molecular Sciences 25, no. 1: 675. https://doi.org/10.3390/ijms25010675

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