Next Article in Journal
A Reciprocal Link between Oral, Gut Microbiota during Periodontitis: The Potential Role of Probiotics in Reducing Dysbiosis-Induced Inflammation
Next Article in Special Issue
Developmental Exposure to DDT Disrupts Transcriptional Regulation of Postnatal Growth and Cell Renewal of Adrenal Medulla
Previous Article in Journal
Defining the Molecular Mechanisms of the Relaxant Action of Adiponectin on Murine Gastric Fundus Smooth Muscle: Potential Translational Perspectives on Eating Disorder Management
Previous Article in Special Issue
Early-Life Exposure to Traffic-Related Air Pollutants Induced Anxiety-like Behaviors in Rats via Neurotransmitters and Neurotrophic Factors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Application of In Vitro Models for Studying the Mechanisms Underlying the Obesogenic Action of Endocrine-Disrupting Chemicals (EDCs) as Food Contaminants—A Review

by
Monika Kowalczyk
1,†,
Jakub P. Piwowarski
2,*,
Artur Wardaszka
1,
Paulina Średnicka
1,
Michał Wójcicki
1 and
Edyta Juszczuk-Kubiak
1,*,†
1
Laboratory of Biotechnology and Molecular Engineering, Department of Microbiology, Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology—State Research Institute, 02-532 Warsaw, Poland
2
Microbiota Lab, Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, 02-097 Warsaw, Poland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(2), 1083; https://doi.org/10.3390/ijms24021083
Submission received: 5 December 2022 / Revised: 29 December 2022 / Accepted: 4 January 2023 / Published: 5 January 2023
(This article belongs to the Special Issue Advances in the Research of Endocrine Disrupting Chemicals 2023)

Abstract

:
Obesogenic endocrine-disrupting chemicals (EDCs) belong to the group of environmental contaminants, which can adversely affect human health. A growing body of evidence supports that chronic exposure to EDCs can contribute to a rapid increase in obesity among adults and children, especially in wealthy industrialized countries with a high production of widely used industrial chemicals such as plasticizers (bisphenols and phthalates), parabens, flame retardants, and pesticides. The main source of human exposure to obesogenic EDCs is through diet, particularly with the consumption of contaminated food such as meat, fish, fruit, vegetables, milk, and dairy products. EDCs can promote obesity by stimulating adipo- and lipogenesis of target cells such as adipocytes and hepatocytes, disrupting glucose metabolism and insulin secretion, and impacting hormonal appetite/satiety regulation. In vitro models still play an essential role in investigating potential environmental obesogens. The review aimed to provide information on currently available two-dimensional (2D) in vitro animal and human cell models applied for studying the mechanisms of obesogenic action of various industrial chemicals such as food contaminants. The advantages and limitations of in vitro models representing the crucial endocrine tissue (adipose tissue) and organs (liver and pancreas) involved in the etiology of obesity and metabolic diseases, which are applied to evaluate the effects of obesogenic EDCs and their disruption activity, were thoroughly and critically discussed.

1. Introduction

According to the World Health Organization (WHO), obesity is one of the top ten threats to human health [1]. The research of the Statistical Office of the European Union (Eurostat) shows that 52.7% of the adult population of the European Union (EU) was overweight in 2019, of which approximately 17% were obese [2]. Moreover, in the EU, every eighth child aged 7–8 is obese [3]. Nowadays, there are far more people in the world overweight or classified as obese than malnourished and obesity has become a serious health problem in both developed and developing countries [1]. Obesity is associated with comorbidities such as the increased risk of cardiovascular disease, insulin resistance, Type 2 diabetes mellitus (T2DM), hypertension, as well as non-alcoholic fatty liver disease (NAFLD), and hormone-sensitive cancers [4]. Obesity is characterized by an imbalance between energy intake and total energy expenditure resulting in increased lipid accumulation in adipocytes and excess fat storage in the body [5,6]. However, recent studies have demonstrated that consumption of a calorie-dense diet coupled with physical inactivity as well as genetics cannot explain rising obesity among adults and children [7,8]. Over the past decade, there is considerable evidence that a substantial increase in environmental chemical production may contribute to the rapid increase in human obesity and metabolic syndrome [7,8]. Especially, environmentally existing xenobiotic chemicals, such as endocrine-disrupting chemicals (EDCs) are the main candidates [4,9,10,11,12,13]. EDCs are a class of natural or synthetic exogenous chemical substances that may interfere with the function of the endocrine system by mimicking or blocking hormone biosynthesis, metabolism or action [4,7,9,10,13]. Their adverse effects on estrogen, androgen, and thyroid hormone signaling have been well documented [14,15,16,17]. Moreover, EDCs promote adverse effects during fetal life, infancy, puberty, and pregnancy via epigenetic mechanisms and some of these effects can be transmitted to the next generations [11,18,19,20]. Research shows that prenatal and perinatal exposure to EDCs may contribute to the greater storage of fat in the organism from the beginning of its life [13].
EDCs encompass a variety of chemical classes, including plasticizers, pesticides, industrial by-products, and pollutants [4,11,12]. The most common contaminants are bisphenols, phthalates, dioxins, pesticides, and polychlorinated biphenyls to which humans are exposed daily by eating, breathing polluted air, and drinking contaminated water [21]. However, new evidence has shown that some of them can stimulate lipid accumulation in target cells such as adipocytes and hepatocytes or disrupt sensitive metabolic processes, leading to obesity and metabolic syndrome [22]. These EDCs are referred to as “environmental obesogens” [9,18,23,24]. It has been reported that obesogens can promote obesity by increasing body weight [25], as well as causing hypertrophy and/or hyperplasia of adipocytes [26], stimulating the adipogenesis process [27], disturbing lipid and glucose metabolism [28], interfering with neuroendocrine regulation of satiety and appetite [7,28], inhibiting energy expenditure and/or brown adipose tissue thermogenesis [29], promoting inflammation [30], as well as changing in the taxonomic and metabolomic profiles of the gut microbiota [31]. The main source of human exposure to obesogenic EDCs is through diet, particularly the consumption of contaminated food such as meat, fish, fruit, vegetables, milk, and dairy products [21]. Such compounds can penetrate foods as a result of migration from food contact materials, including plastic containers for foodstuff and drinks and lining materials for food and beverage cans [32,33,34]. Exposure to these substances may also occur via inhalation and dermal absorption as they are widely used in personal care products (cosmetics, perfumes, lotions), detergents, medical devices, children’s toys, printing inks, the thermal paper used in cash register receipts as well as textiles [33,34,35]. Moreover, many chemicals that have been identified as obesogenic EDCs are pesticides, although some of them were withdrawn from general use many years ago but are still present in the environment [35,36]. EDCs’ concentration in food varies among countries and different foodstuffs as well as types of food packaging [37]. Moreover, socio-demographic, lifestyle and dietary habits can play important role in EDC exposure [38]. To date, there are more than 1000 chemicals reported to have endocrine effects [18,20,39] among which about 10 different classes of obesogenic chemicals including pesticides, in particular, organotins [40], industrial chemicals such as bisphenol A (BPA) [41] and di(2-ethylhexyl) phthalate (DEHP) [42], as well as parabens (alkyl esters of p-hydroxybenzoic acid) [43], have been characterized [44]. Several epidemiological trials have shown that high levels of these contaminants have been detected globally in human blood plasma [45], urine [46,47], breast milk [48,49], and adipose tissue [50]. Moreover, a significantly higher level of EDC exposure was observed in children compared to adults [38,51]. In Poland, results of a cohort study including children from the prospective Polish Mother and Child Cohort (REPRO_PL) showed a higher level of urinary phthalate metabolites in children at an age of 7 years than in the same children at age 2 and their mothers during pregnancy [51]. Moreover, phthalate exposures were much higher than exposures reported in other European children populations [51].
The dramatically increasing production of highly processed foods and evidence of health risks linked with food chemical contamination has led to the development of different chemical analogous and alternatives [52,53,54,55,56,57,58]. Nonetheless, results of several studies have suggested that some of the biological activities of chemical analogous are of a similar or even higher magnitude in comparison to initial compounds [58,59,60,61] but mechanistic aspects underlying the obesogenic effects remain largely unrecognized [62]. Therefore, in vitro models still play an essential role in identifying environmental obesogens and understanding obesogenic mechanisms for further insight into the in vivo action of these chemicals linked with the risk of developing various chronic diseases including obesity and metabolic disorders. Moreover, in vitro models provide a rapid approach to investigating hazards of exposure to EDCs and their toxicity potential, while reducing or eliminating the need for animal testing [63].
Hence, this review aimed to provide information from peer-reviewed literature on currently available 2D in vitro animal and human cell models applied for studying the mechanisms of action of the various obesogenic EDCs as food contaminants. We focused mainly on in vitro models representing the main tissues and organs involved in the etiology of obesity and metabolic diseases, such as adipose tissue, liver, and endocrine pancreas. These in vitro models are currently used to evaluate the chemical toxicity of EDCs and their metabolic disruption activity. We believe that this comprehensive insight could help scientists choose the appropriate in vitro model to study the mechanisms of action of environmental obesogens associated with the risk of human obesity and its comorbidities.

2. Adipogenesis and Obesogenic Action of Endocrine-Disrupting Chemicals (EDCs)

Adipose tissue (AT) is a metabolic active tissue located in the body under the skin (subcutaneous adipose tissue (SAT)) as well as around the internal organs (visceral adipose tissue (VAT)) [64]. AT contains different types of cells, such as preadipocytes, adipocytes, immune cells, fibroblasts, and endothelial cells [65]. In mammals, there are two types of AT, white (white adipose tissue (WAT)) and brown (brown adipose tissue (BAT)), which differ in anatomical location, morphology, functions, biochemical features, and gene expression patterns [66]. WAT accounts for over 95% of adipose mass, while BAT represents about 1–2% [66]. WAT is responsible for energy storage in triglycerides, formed from the esterification of glycerol-3-phosphate and free fatty acids (FFAs) [64]. The most dominant characteristic of BAT is non-shivering thermogenesis, where energy derived from fatty acid (FA) oxidation generates heat by mitochondrial uncoupling to maintain body temperature [29,66]. The magnitude of adipose tissue mass is determined by the enlargement of existing adipocytes (hypertrophy) and by an increase in preadipocyte number (hyperplasia) [64,67,68]. During childhood and adolescence, the number of adipocytes is determined and remains constant in the adult body, regardless of whether the individual is obese or lean [65]. In adulthood, the mass of WAT increases through hypertrophy [65]. In overnutrition, there is an increased accumulation of fat in the adipocytes and cells undergo cellular hypertrophy. In contrast, lipolysis occurs in the adipocytes during calorie restriction [68].
Adipogenesis is a multi-step process leading to the conversion of mesenchymal stem cells (MSCs) and preadipocytes into mature adipocytes and consists of three stages: commitment of MSCs to the adipocyte lineage, mitotic clonal expansion (replication of DNA and duplication of cells intensively takes place), and terminal differentiation [66]. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding proteins (C/EBP), C/EBPδ, and C/EBPβ, are key transcription factors during the early stages of differentiation [66]. PPARγ and C/EBPα cooperatively promote differentiation and the induction of adipocyte-specific genes including, inter alia, adipocyte protein 2 (AP2) and glucose transporter type 4 (GLUT4). At the terminal differentiation stage, the preadipocytes acquire the features of mature adipocytes, such as insulin sensitivity, lipid synthesis and transport, and secretion of adipocyte-specific proteins [68].
Recent evidence showed that chronic human exposure to obesogenic EDCs can be associated with inducing preadipocyte differentiation, increasing oxidative stress, and promoting a pro-inflammatory state leading to an increase in the risk of obesity and metabolic disorders [7,8,18,69]. Numerous obesogens such as tributyltin (TBT), bisphenol A (BPA) as well as mono-2-ethylhexyl phthalate (MEHP) activate adipogenesis by acting on nuclear receptors (NRs), in particular by activating retinoid X receptor (RXR)/PPARγ-dependent signalling [70,71,72,73]. Activation of RXR/PPARγ plays a crucial role in the regulation of the expression of genes involved in lipid droplet formation, glucose uptake and insulin responsiveness [74]. Furthermore, obesogenic EDCs can promote adipogenesis and lipid storage and fat deposition by interfering with steroid hormone receptors such as glucocorticoid receptors (GRs) and estrogen receptors (ERs) [75,76]. For example, BPA-binding GRs directly increase adipogenesis and lipid accumulation and indirectly via induction of the 11beta-hydroxysteroid dehydrogenase 1 (HSD11B1) mRNA expression involved in cortisone/cortisol conversion [76].
In vitro studies have shown that many obesogenic EDCs not only induce the differentiation of MSCs into adipocytes [55,77,78,79] but also alter the metabolism of mature adipocytes [8,62,80,81,82,83,84]. Exposure to EDCs can reduce the sensitivity of adipocytes to insulin, which causes an increase in blood glucose level and, consequently, may lead to insulin resistance in WAT of adulthood, potentially via a reduction in protein (serine/threonine) kinase B (PKB, also known as Akt) and GLUT4 levels [8,85]. Moreover, EDCs affect the expression of genes related to the de novo synthesis of free fatty acids, such as fatty acid synthase (FASN) or sterol regulatory element-binding protein 1c (SREBP1C), as well as the synthesis of triglycerides, such as diacylglycerol acyltransferase 1 and 2 (DGAT1 and DGAT2), leading to a disturbance in the adipose lipid metabolism [86]. Recent evidence showed that the obesogenic action of some EDCs is associated with disruption of appetite/satiety signaling and food preferences [18]. As an endocrine organ, WAT plays an integral role in maintaining global energy homeostasis by secretion of adipokines (leptin, adiponectin), which regulate global insulin sensitivity, satiety and inflammation [66,87]. Several EDCs have been shown to alter leptin levels in animal models, including dichlorodiphenyldichloroethylene (DDE) and DEHP [80,84]. The in vitro effect of EDCs on other adipokines such as resistin correlated with insulin signaling has also been reported [80]. The potential mechanisms of the obesogenic action of daily exposed EDCs on the development of obesity and metabolic syndrome are presented in Figure 1.

3. In vitro Models Used in the Study of the Obesogenic Effects of EDCs

Data from epidemiological trials are essential for evaluating of potential adverse effects of EDC exposure but usually provide only suggestive outcomes. Therefore, in vitro models including animal (Figure 2A) and human ones (Figure 2B) are used to investigate molecular mechanisms underlying the obesogenic action of EDCs prior to in vivo studies. The first group of in vitro models utilizes preadipocytes and mature adipocytes of humans and different animals such as rodents, although porcine or feline cells have also been used to a lesser extent. The second group of in vitro models is used to study the biotransformation of xenobiotics, assess their toxicity and identify the molecular mechanisms that lead to the disruption of the endocrine function of the crucial metabolic organs that control glucose and lipid homeostasis (e.g., the liver and pancreas).
The main advantage of animal in vitro models over in vivo research is the speed of the experiments, tight control of the environment, reduced cost, well-established protocols, higher throughput and reduced animal use [88]. Regarding adipogenesis, most of these models utilize mouse 3T3-L1 cells to elucidate the mechanisms of EDCs’ action during the preadipocyte differentiation processes. However, it is still unclear if the rodent’s in vitro models are suitable for studying adipogenic responses due to the maintained species specificity, which may have varying responses to obesogens and limit the application of results for human-based risk assessments [89]. Nonetheless, numerous human in vitro models including primary cells from different organs and tissues have been developed and are used to identify obesogens and elucidate the mechanisms of their action implicated in the adipogenic differentiation process, adipose function and hepatotoxicity [8,83,84,90]. Regarding the adipogenesis process, in recent years, mesenchymal stem cells (MSCs) derived from adipose tissue have been utilized as an alternative to animal and human preadipocyte cell models to investigate the mechanisms of obesogenic EDC action, particularly related to disrupting the programming of adipogenesis during a prenatal period, in combination with a Western diet, which leads to a higher risk of obesity in early life and adolescence [91].

4. Adipose Tissue Cell Models

4.1. Animal Preadipocytes

4.1.1. 3T3-L1 Cell Line

The 3T3-L1 cell line is a well-established in vitro system of white preadipocytes from murine Swiss 3T3 cells [92] and consists of unipotent preadipocytes, which can differentiate only into mature adipocytes [93]. Initiation of the adipogenesis in 3T3-L1 cells requires the treatment of several pro-differentiation agents after cell growth arrest, such as dexamethasone (DEX), insulin, and phosphodiesterase inhibitor 1-methyl-3-isobutyl xanthine (IBMX) [92]. The 3T3-L1 cells are easy to culture and tolerate a very large amount of passages [94]. An important feature of 3T3-L1 cells is their ability to differentiate into both white and brown adipocytes [95]. Unfortunately, 3T3-L1 cells differ between batches from different vendors, which makes them impossible to define as a universal test system [39]. Nonetheless, the 3T3-L1 cell line has been used widely to investigate the effects of various EDCs to establish the molecular mechanisms of adipogenesis and evaluate the potential effects on the risk of obesity [93]. 3T3-L1 cells have been used by Sun et al. [93] to evaluate the molecular mechanisms of a widely used surfactant, 4-hexylphenol (4-HP), as a potential EDC-impaired adipogenesis process. Results showed that 4-HP induced adipogenic differentiation via increasing the mRNA level of PPARγ and its target genes such as fatty acid-binding protein 4 (FABP4) (also known as adipocyte protein 2 (AP2)), fatty acid translocase (CD36), perilipin, and adiponectin, but did not disturb C/EBPα expression. Moreover, in 3T3-L1 cells exposed to 4-HP, a significant increase in lipid accumulation was observed [93]. Choi et al. [7] proved that exposure of 3T3-L1 cells to BPA and its analogous such as bisphenol S (BPS), and bisphenol F (BPF) resulted in increased both mRNA and protein levels of PPARγ, C/EBPα, and AP2. De Filippis et al. [96] reported that BPA had no impact on the PPARγ, FABP4, and FASN expression and adipocyte differentiation but increased mRNA levels of pro-inflammatory markers, tumor necrosis factor-α (TNFα) and interleukin 6 (IL-6) in 3T3-L1 cells. Moreover, Sargis et al. [97] showed that induction of the adipocyte differentiation in the 3T3-L1 cells was promoted by exposure to BPA, dicyclohexyl phthalate (DCHP), and tolylfluanid (TF) through stimulating glucocorticoid receptors (GR), without any significant activation of PPARγ expression. Meruvu et al. [98] evaluated the potential of benzyl butyl phthalate (BBP) on the epigenetic modification of genes involved in adipogenesis. BBP exposure induced miR-34a-5p expression and significantly decreased the expression level of its target genes, including nicotinamide phosphoribosyltransferase (NAMPT), sirtuin 1 (SIRT1), and sirtuin 3 (SIRT3), leading to an impairment in 3T3-L1 preadipocyte differentiation and an increase in adipogenesis [98]. Numerous studies using the 3T3-L1 model have confirmed the obesogenic potential of extensively used different classes of pesticides including quizalofop-p-ethyl (QpE) [99], dichlorodiphenyltrichloroethane (DDT) and dichlorodiphenyldichloroethylene (DDE) [100], diazinon [101], chlorpyrifos (CPF) [102], and tributyltin (TBT) [103], as well as zoxamide, spirodiclofen, flusilazole and acetamiprid [104]. For example, Mangum et al. [50] showed that exposure to 1,1-dichloro-2,2-bis(4-chlorophenyl)ethane (p,p’-DDE) at both the 10 and 20 μM concentrations increased intracellular lipid accumulation by 42% and 58% respectively, compared to the control. The induction of 3T3-L1 preadipocyte differentiation into mature, lipid-storing adipocytes after exposure to these agrochemicals was primarily regulated via PPARγ activation; nonetheless, multiple other obesogenic mechanisms including mitochondrial dysfunction or altered intracellular calcium levels have been also reported [29,105,106,107].

4.1.2. NIH3T3-L1 Cell Line

The NIH3T3-L1 cell line was derived from desegregated NIH Swiss mouse embryo fibroblasts [108]. These cells are adherent, exhibit many physiological similarities to primary adipocytes, and are relatively easy to culture, therefore they are a good model to study adipogenesis and adipocyte function [80,108]. Regarding the EDC exposure, Riu et al. [109] evaluated the effect of tetrabromobisphenol (TBBPA) and tetrachlorobisphenol A (TCBPA), the brominated analogues of BPA, on preadipocyte differentiation and showed that TBBPA and TCBPA, via PPARγ activation, promoted triglyceride accumulation in the NIH3T3-L1 cells. In turn, Howell et al. [80] showed that exposure to DDE did not affect adipogenesis in NIH3T3-L1 cells, but significantly increased the level of adipokines such as resistin, adiponectin, and leptin in mature adipocytes.

4.1.3. 3T3-F442A Cell Line

The 3T3-F442A cell line contains murine preadipocytes of WAT [81,110] which does not require stimulation with DEX and IBMX to differentiate into mature adipocytes [111]. The 3T3-F442A cell line was derived from fibroblasts isolated from disaggregated Swiss mouse embryos and is used to investigate the mechanism of adipogenesis [110,111]. This cell line was applied to investigate the adipogenic potential of DDT, belonging to the group of organochlorine (OC) insecticides that were used heavily during the 1950s and 1960s [112,113,114]. Several epidemiological studies have linked DDT exposure to T2D and obesity [115,116,117]. An in vitro study by Moreno-Aliaga and Matsumura [111] showed that 1,1,1-trichloro-2,2-bis (p-chlorophenyl)-ethane (p,p’-DDT) at a concentration of 20 μM caused 3T3-F442A cells to obtain the adipocyte-like morphology at day 2 of the differentiation process, but treated cells did not fully differentiate. The mouse 3T3-F442A cell line was also used to investigate the impact of BPA exposure on glucose transport [81] and demonstrated that BPA enhanced basal and insulin-stimulated glucose uptake partially via increased GLUT4 protein levels.

4.1.4. OP9 Cell Line

The murine OP9 is an adipocyte cell culture model established from the calvaria of newborn mice genetically deficient in functional macrophage colony-stimulating factor (M-CSF) [94,118]. OP9 cells are bone marrow-derived stromal preadipocytes of WAT characterized by fast adipogenic differentiation and the rapid accumulation of triacylglycerides in lipid droplets after only 72 h of adipogenic stimuli [39,94]. Moreover, OP9 cells can differentiate into adipocytes after reaching confluence and can be passaged for long periods in culture [39]. In comparison with 3T3-L1 cells, OP9 cells are more sensitive to the induction of adipogenesis by chemicals with the ability to activate PPARγ and RXR [19]. OP9 differentiation is a PPARγ-dependent process, and differentiating preadipocytes express C/EBPA, C/EBPβ, as well as perilipin-1 (PLIN1) and perilipin-4 (PLIN4), similar to other adipocyte models [119]. In the last years, the OP9 cell line has been used to evaluate the effects of various compounds on adipogenesis [39,120]. This cell line was used by Kassotis et al. [39] to evaluate the effect of TBT exposure on preadipocyte differentiation. Authors reported that TBT stimulated preadipocyte differentiation, and significantly enhanced triglyceride accumulation in cells on day 7 of differentiation at 100 nM concentration [39]. In turn, 36 potentially adipogenic chemicals identified by the Toxicological Priority Index (ToxPi) on preadipocyte differentiation and adipocyte metabolism in the OP9 cells were also reported by Andrews et al. [120]. Results showed that TBBPA significantly enhanced adipocyte differentiation and had high efficiency in inducing lipid accumulation in OP9 cells at 20 µM [120].

4.1.5. ST-13 Cell Line

The ST-13 cell line was derived from newborn mouse skin and consists of preadipocytes of WAT [82]. Differentiation of the ST-13 preadipocytes is stimulated via ciglitazone and during cell differentiation, the expression of the adipogenic markers such as PPARγ, C/EBPβ and AP2 is induced [121]. The ST-13 cell line has been used by Yamasaki et al. [82] to evaluate the effect of TBBPA exposure on the expression level of genes related to lipid metabolism in differentiated and undifferentiated adipocytes. Exposure to TBBPA at concentrations of 0.5 µM and 1 µM did not have any effects on lipid accumulation and mRNA level of the acetoacetyl-CoA synthetase (AACS) and succinyl-CoA-3-oxoacid CoA-transferase (SCOT) in mature adipocyte cell culture. However, a significantly increased gene expression of lipid and ketone body-utilizing factors such as AACS, PLIN1 and fatty acid synthase (FAS) in both 0.5 µM and 1 µM of TBBPA in ST-13 preadipocytes was noticed. Surprisingly, data showed that the BAT-related factors, uncoupling protein-1 and -3 (UCP-1 and UCP-3), PR domain-containing 16 (PRDM16), lysine-specific demethylase-1 (LSD-1), as well as cell death-inducing DNA fragmentation factor-alpha-like effector A (CIDEA), were overexpressed in ST-13 preadipocytes upon TBBPA treatment [82].

4.1.6. UCP-1 Cell Line

Immortalized UCP-1 reporter brown preadipocytes were generated from Ucp1-luciferase reporter mice and were used to determine the promoter activity of the UCP1 gene [29]. Only one study investigated the effect of EDC exposure on the expression of UCP-1 in immortalized brown adipocytes [29]. Wang et al. [29] investigated the effects of 34 chemicals commonly found in food due to food processing, packaging, and agriculture practices, and showed that only the organophosphate insecticide CPF suppressed UCP1 expression and mitochondrial respiration in BAT at a concentration of 1 pM. Moreover, RNA sequencing showed that at 1 pM CPF, after 4 h exposure, the mRNA levels of the carnitine palmitoyltransferase I A (CPT1A), carnitine palmitoyltransferase I B (CPT1B), and acetyl-coenzyme A acetyltransferase 3 (ACAT3) genes, important for regulating fatty acid oxidation, as well as the cytochrome C oxidase assembly factor (COX16) gene, were reduced [29].

4.2. Human Preadipocytes

4.2.1. Primary Human Preadipocytes

Primary human preadipocytes are often applied as in vitro models for the study of preadipocyte differentiation and adipocyte metabolism [122]. They are isolated from adipocyte tissue from different anatomical sites and different donors. Therefore, they reflect donor- and depot-specific characteristics which may lead to some unpredictable differences during experimental studies [94,122,123]. Even though they reflect the characteristics of the donor, they are useful in studies assessing differences between individuals (e.g., obesity, weight loss, age) [94].
Regarding the EDCs, primary subcutaneous human preadipocytes from healthy donors with body mass indices BMI ≤ 24.99 kg m−2 were used to investigate the mechanism of BPA-induced adipogenesis [75]. The study showed that BPA exposition increased the expression of C/EBPα and β, PPARγ as well as preadipocyte lipid accumulation in the absence of GR and GR agonist [75]. In another study [70], in vitro microarray analysis showed that human subcutaneous preadipocytes from donors with BMI ≤ 24.99 kg m−2 exposed to 50 µM BPA revealed 373 differentially expressed genes, with 235 of those upregulated and 138 genes downregulated. Several genes involved in triglyceride (TG) and lipid metabolism were upregulated after BPA exposure, including acetyl-CoA carboxylase α (ACACA), apolipoprotein A1-binding protein (APOA1BP), perilipin 2 (PLIN2), fatty acid desaturase 1 (FADS1), Niemann-Pick 2 (NPC2), and phosphatidic acid phosphatase type 2A (PPAP2A). In addition, for BPA-treated cells, an increase in mRNA levels was noticed for genes related to lipid metabolism such as sterol regulatory element-binding transcription factor 1 (SREBF1), low-density lipoprotein receptor (LDLR), lipoprotein lipase (LPL), and insulin-induced gene 1 (INSIG1), as well as for those related to adipogenesis, such as growth differentiation factor 15 (GDF15). Moreover, network interaction analysis identified the mammalian target of rapamycin (mTOR) signaling and the thyroid-receptor/retinoid X receptor (TR/RXR) activation as potentially involved in BPA-mediated adipogenesis [70]. Wang et al. [76] analyzed the effect of BPA on human visceral preadipocytes and adipocytes and showed that BPA in the lowest concentration tested (10 nM) increased the mRNA level of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) gene (encoding an enzyme essential in adipogenesis and lipid synthesis). In addition, an increase in PPARγ and lipoprotein lipase (LPL) mRNA levels in preadipocytes and adipocytes was also observed [76]. Currently, scientists are very interested in the molecular effects through which EDCs can disrupt adipose tissue function in children, leading to a risk of childhood obesity and related metabolic syndrome [124]. Therefore, the detrimental effect of BPA on adipogenesis in primary preadipocytes derived from children and modulating endocrine functions has been reported [124]. Menale et al. [124] investigated the molecular mechanisms by which environmentally relevant doses of BPA affect adipogenesis in preadipocytes derived from subcutaneous adipose tissue of non-obese children between 7 and 10 years old. BPA increased the expression of FABP4 and CD36, which are important for lipid metabolism, as well as the expression of proinflammatory cytokines, such as interleukin 1 beta (IL1B), interleukin 18 (IL18) and chemokine (C-C motif) ligand 20 (CCL20) [124].

4.2.2. PCS-210-010

PCS-210-010 cells are commercially available cells containing normal primary subcutaneous human preadipocytes derived from WAT after liposuction surgery. PCS-210-010 cells can proliferate in an undifferentiated state and possess a higher efficiency of adipogenesis than mesenchymal stem cells. Interestingly, PCS-210-010 preadipocytes can be differentiated down osteogenic and chondrogenic lineages [125]. Regarding the EDCs, only one study reported by El-Atta et al. [83] showed that BPA exposure induced the PPARγ, AP2, and peptidylprolyl isomerase A (PPIA) expression as well as increased adiponectin levels in PCS-210-010 cells.

4.2.3. SGBS

SGBS cells were isolated from the stromal vascular fraction of adipose tissue of a male infant with Simpson-Golabi-Behemel syndrome (SGBS) by Wabitsch et al. [122,126]. The SGBS cells exhibit a high capacity for adipose differentiation and were applied in a study of human adipocyte development and metabolism [126]. SGBS cells, unlike primary preadipocytes from healthy donors, retain the ability to differentiate over at least 50 generations [122].
Recently, several studies applied SGBS as a model to investigate the action of a variety of plasticizers such as BPA and phthalates on human adipocyte metabolism [8,84]. For example, Schaffert et al. [8] reported that BPA and its analogues such as BPS, BPF, bisphenol B (BPB) and bisphenol AF (BPAF) displayed significant binding to PPARγ during adipocyte differentiation, but not activated PPARγ at concentrations of 10 nM, 100 nM, 1 µM and 10 µM in SGBS cells. Interestingly, during the differentiation of SGBS preadipocytes, all bisphenols decreased lipid accumulation and BPS, BPB, BPF and BPAF decreased adiponectin levels. Moreover, 1 µM BPA, BPB and BPS considerably reduced insulin sensitivity in SGBS cells upon insulin stimulation [8]. On the other hand, Schaedlich et al. [84] reported that DEHP downregulated the FABP4, adipose triglyceride lipase (ATGL), LPL, lipase E (LIPE), and CD36 mRNA levels, as well as reduced TGs accumulation in lipid droplets of the SGBS cells. Moreover, decreased adiponectin levels and increased leptin secretion in SGBS cells were also observed [84]. Recently, the effects of 20 alternative plasticizers and their metabolites on SGBS preadipocyte differentiation, induction of adipogenic markers and lipid accumulation in mature adipocytes have been reported by Schaffert et al. [62]. The molecules 1,2-cyclohexanedicarboxylic acid mono 4-methyloctyl ester (MINCH), monohydroxy isononyl phthalate (MHINP) and 6-hydroxy monopropylheptyl phthalate (OH-MPHP), which are the metabolites of bis(7-methyloctyl) cyclohexane-1,2-dicarboxylate (DINCH), diisononyl phthalate (DINP), and bis(2-propylheptyl) phthalate (DPHP), respectively, exhibited the highest adipogenic potential by induction of the SGBS preadipocyte differentiation mediated by PPARγ binding and activation. In mature adipocytes, DINCH, DINP and DPHP as well as their metabolites induced oxidative stress and mitochondrial dysfunction, and disturbed lipid storage and adipokine secretion, which was linked to inflammation and insulin resistance [62].

4.2.4. SW 872 Cell Line

SW 872 is a human liposarcoma cell line that differentiates without the differentiation cocktail and they constitutively express PPARγ and C/EBPα, which are crucial to adipocyte development [127,128]. Campioli et al. [129] showed that exposure to MEHP at a concentration of 10 µM activated the SW 872 preadipocyte differentiation and increased the expression of the glucose transporter type 1 and 4 (GLUT1 and GLUT4), calcium-binding protein B (S100B), as well as adenosine triphosphate citrate lyase (ACLY) and ACACA, involved in de novo lipogenesis. Additionally, MEHP temporarily increased the translocator protein (TSPO) mRNA levels during SW 872 adipocyte differentiation [129].
The obesogenic activity of selected EDCs confirmed on adipose tissue cell models is summarized in Table 1.

5. Mesenchymal Stem Cells (MSCs)

Mesenchymal stem cells (MSCs) are multipotent and can differentiate into adipocytes, osteocytes, myocytes and chondrocytes [78,93,103,130]. In a variety of studies, MSCs were isolated from bone marrow, adipose tissue, periosteum, muscle tissue, blood vessels, blood, lymphoid organs, lung, skin and umbilical cord [131]. MSCs’ differentiation is controlled via several transcription factors such as octamer-binding transcription factor 4 (OCT4), SRY-box 2 (SOX2) and Nanog homeobox (NANOG) that are responsible for maintaining cells in an undifferentiated state [77]. In addition, MSCs adhere to plastic and express specific surface antigens such as CD19-, CD34-, CD45-, CD79-, CD14 or CD11b-, CD73+, CD90+, and HLA-DR [132]. MSCs are used to study the modulation of stem cell fate under environmental and nutritional factors, and various aspects of adipogenesis in vitro [93,103,129,130].

5.1. Animal Adipose-Derived Stem Cells (ADSCs)

The C3H10T1/2 cell line was established in 1973 and derived from C3H mouse embryos that were 14–17 days old [133,134]. C3H10T1/2 cells have a fibroblastic morphology and the capacity to differentiate into adipocytes, chondrocytes and osteocytes [133]. In recent years, C3H10T1/2 cells have been used to investigate the impact of various compounds on preadipocyte differentiation to investigate the molecular mechanisms associated with obesity [94,135,136]. Regarding the EDCs, the C3H10T1/2 cell line was applied to study the impact of parabens such as butylparaben on the disruption of the adipogenesis process. Results showed that exposure to butylparaben stimulated adipogenic differentiation via increased expression of PPARγ, C/EBPα and FABP4, as well as via decreased runt-related transcription factor 2 (RUNX2) mRNA levels, which plays an inhibitory role during adipocyte differentiation [130]. Other studies revealed that BBP induced adipocyte differentiation in C3H10T1/2 stem cells [137,138]. In turn, Zhang and Choudhury [138] showed that expression of the PPARγ and aP2 were significantly increased in C3H10T1/2 stem cells exposed to 50 µM BBP after 8 days of incubation. In addition, decreased SIRT1 mRNA levels, as well as increased β-catenin and forkhead box protein O1 (FoxO1) acetylation under BBP exposure, were also associated with increased adipogenesis [138]. Moreover, the same authors [137] reported that 50 µM BBP significantly downregulated the expression of long non-coding H19 RNA and increased the expression of miR-103/107/let-7 (a, b, c, d, f and g) on day 2 of C3H10T1/2 cell differentiation, which probably stimulated adipogenesis. An adipogenic effect of DEHP, BPA, and TBT as a single compound was also observed in C3H10T1/2 cells by Biemann et al. [139]. Moreover, Biemann, Fisher and Navarrete Santos [140] studied the effects of the EDC mixture at high concentrations (10 µM BPA, 100 µM DEHP, 100 nM TBT), and at environmentally relevant concentrations (10 nM BPA, 100 nM DEHP, 1 nM TBT) and demonstrated that the EDC mixture affects adipogenic differentiation of the C3H10T1/2 cells, but its impact on adipogenesis was dose-dependent. Moreover, in a previous study, Kirchner et al. [78] provided evidence that TBT induced PPARγ2, FABP4 and leptin (LEP) expression in mouse ADSCs while the mRNA level of the adipocyte differentiation-associated protein (PREF-1), an inhibitor of adipocyte differentiation, was decreased.
In recent years ADSCs from domestic animals have gained increased attention because they are a much better model for understanding adipogenesis in vitro and obesity-related diseases compared to rodent cell models [141]. For example, ADSCs isolated from porcine adipose tissue have been applied to study the obesogenic activity of EDCs. Gigante et al. [142] demonstrated that glyphosate (GLY) at the concentration of 4 μg/mL significantly decreased the viability of ADSCs and inhibited their adipogenic differentiation. Similar results were also observed by Berni et al. [143] who noticed a significantly decreased cell viability of proliferating porcine ADSCs after 48 and 72 h of 1 µM BPS exposure. Moreover, similar to GLY, BPS did not increase the PPARγ and LEP expression during the differentiation nor fat droplet formation in porcine ADSCs [143].

5.2. Human Adipose-Derived Stem Cells (hADSCs)

Human adipose-derived stem cells (hADSCs) are isolated from biopsies and liposuction specimens [144,145]. hADSCs are available from normal donors and patients with obesity (BMI > 30), Type 1 diabetes or Type 2 diabetes. Moreover, hADSCs have functional and phenotypic characteristics similar to bone marrow-derived mesenchymal stem cells (BMMSC) [145]. Normal hADSCs can differentiate into many different lineages including adipogenic, neural, osteogenic, and chondrogenic cells and can be used in research, including in stem cell differentiation [145]. An important feature of hADSCs is that they can be cultivated up to passage eight with no sign of decline [141]. hADSCs can be used as an alternative to human preadipocytes which have reduced proliferative ability and can exhibit physiological differences related to the fat depot of origin within the body [77]. A main advantage of hADSCs is the commitment of stem cells to preadipocytes and their differentiation to mature adipocytes [146]. hADSCs have been used to assess possible metabolic disruptors in vitro [77,78,147].
Regarding obesogenic EDCs, Valentino et al. [147] showed that the adipose tissue-derived stromal vascular fraction (SVF) exposed to 1 nM BPA exposure decreased insulin-stimulated glucose utilization and increased cytokine secretion such as IL6 and interferon-gamma (IFN-γ). However, no changes in mRNA levels of the adipogenic markers such as GLUT4 and PPARγ were found [147]. Ohlstein et al. [148] showed that BPA enhanced adipogenesis in human ADSCs obtained from subcutaneous abdominal tissue of three female donors with a BMI less than 25. BPA increased the expression of PPARγ, C/EBPα, LPL, insulin-like growth factor-1 (IGF1) and dual leucine zipper-bearing kinase (DLK). It has been reported that the adipogenic effect of BPA was compounded via ER activation, and this effect can be blocked by the ER antagonist ICI 182,780 [148]. De Filippis et al. [96] showed that exposure to 1 nM and 3 nM BPA did not affect the cellular commitment of hADSCs to the adipose lineage, nor did it affect PPARγ, C/EBPα and FABP4 expression or lipid accumulation. Recently, Cohen et al. [55] compared the effects of BPA and BPA replacements such as BPAF and tetramethyl bisphenol F (TMBPF) on adipogenesis and lipid accumulation in female hADSCs. BPA at 0.1 μM and BPAF at 0.1 nM increased adipogenesis and lipid accumulation but higher amounts of BPA (1 μM) and BPAF (10 nM) significantly decreased adipogenesis. Moreover, higher doses of BPA and BPAF were more toxic than lower doses, leading to an increase in cell apoptosis, thus contributing to a decreased level of adipogenesis and fat accumulation. In addition, TMBPF at a concentration of 0.01 µM and 0.1 µM also significantly lowered adipogenesis. This compound exhibited cytotoxic and anti-adipogenic effects on hADSCs, which resulted in increased levels of apoptosis and reduced lipid production [55]. Similar results were also obtained in the study of Harnett et al. [56] who demonstrated that BPA (1, 10 μM), BPAF (0.0003 µM, 0.003 µM, 0.03 µM, 0.3 µM) and TMBPF (0.01 µM, 0.1 µM, 1 µM, 10 µM, 50 µM) had high cytotoxicities and significantly decreased cell viability, leading to massive apoptosis in hADSCs. Reina-Pérez et al. [79] examined the impact of BPF and BPS on lipid accumulation and adipogenesis in hADSCs and showed that these BPA analogues at concentrations of 10 µM or 25 µM enhanced their capacity to differentiate into adipocytes and accumulate lipid droplets in a dose-dependent manner. Another study [77] showed that exposure to DDE maintained the undifferentiated state of hADSCs. DDE influenced the expression of genes involved in maintaining the pluripotent state of cells and differentiation (SOX2, OCT4, NANOG, peroxisome proliferator-activated receptor gamma, coactivator 1 beta (PPARγC1B)), lipid metabolism (FASN, sterol regulatory element-binding protein 1 (SREBP1), UCP3) and members of an insulin signaling pathway (homo sapiens v-akt murine thymoma viral oncogene homolog 2 (AKT2), insulin receptor (INSR)). In turn, hADSCs exposed to TBT showed increased cell differentiation via activation of PPARγ and downregulation of the PREF-1 expression, as an inhibitor of adipocyte differentiation [78].
The obesogenic activity of selected EDCs confirmed on mesenchymal stem cell models is summarized in Table 2.

6. Other In Vitro Models of Adipose Tissue and Mesenchymal Stem Cells That Can Be Used for Studying the Mechanism of Obesogenic Action of EDCs

Several immortalized human white adipocyte cell models such as the telomerase reverse transcriptase white preadipocyte cell line (TERT-hWA) [149], LiSa-2 [150], LS14 [151] and Chub-S7 [152], and brown adipocyte cell models such as TERT-hBA [149] and PAZ6 [153], have been generated. These cells, compared to primary cultures, maintain adipogenic potential over time and passages, and therefore have been used over the past few years to study adipocyte function [154].
TERT-hWA and TERT-hBA were isolated by Markussen et al. [149] upon the immortalization of white and brown stromal-vascular cell fractions from superficial and deep neck adipose tissue from a single donor. These cells maintain a fibroblast-like morphology during propagation and exhibit high proliferation and differentiation up to at least passage 20 [149]. The LiSa-2 cell line was isolated by Wabitsch et al. [150] from a poorly differentiated human pleomorphic liposarcoma and displays a high capacity for terminal adipose differentiation [150]. The LiSa-2 cells accumulate lipids and express adipocyte gene markers such as PPARγ, LPL, FASN, hormone-sensitive lipase (HSL), adipocyte most abundant gene transcript-1 (APM1), glycerol-3-phosphate-dehydrogenase (GPD1) and GLUT4 [150]. LS14 is an adipocyte cell line that was derived from a metastatic liposarcoma and shares many of the characteristics of primary preadipocytes that undergo terminal differentiation, with the expression of many adipose-associated genes [151]. The Chub-S7 cell line was derived from human subcutaneous primary preadipocytes which were transfected with human papillomavirus E7 oncoprotein and the human telomerase reverse transcriptase (hTERT) [152]. Chub-S7 adipocytes display expression of the adipocyte markers and the capacity to accumulate triglycerides [152]. Chub-S7 has been applied in the study of adipocyte differentiation, adipogenic miRNA regulation [154,155], as well as cellular metabolism [154,156].
PAZ6 is the first available immortalized human BAT cell line isolated from infant brown adipose tissue [153,154] and has been used both in white and brown preadipocyte models [154]. Differentiated PAZ6 adipocytes accumulate lipids and express brown/white markers including UCP1, β1, β2, and β3 adrenergic receptors (β-AR), adrenergic receptor α2A (α2A-AR), LPL, GLUT1 and GLUT4, as well as LEP [153,154]. Moreover, PAZ6 cells can be passaged for several months without losing their molecular markers and morphological characteristics [153]. Therefore, the above-mentioned cell lines can be potential in vitro models to study the obesogenic action of various EDCs classified as food contaminants [149].
Similarly, mouse white adipose cell lines such as Ob17 (epididymal fat cells), TA (embryo fibroblasts), or PFC6 (stromal-vascular fraction of epididymal fat) are used as practical rodent models for studying adipogenic mechanisms and adipocyte biology [110]. Additionally, mouse brown adipose cell lines such as BFC-1 (stromal-vascular fraction of interscapular of BAT), RBM-Ad (BAT/bone marrow, multipotent stem cell line), HB2 (stromal-vascular fraction of interscapular of BAT from p53-knock-out-mouse), HIB 1B and T37i (BAT/brown fat tumors) from different SV40T-transgenic mice have been established [110]. Moreover, porcine [157], feline [158], fetal and adult ovine preadipocytes [159] are used in adipogenesis research.

7. Hepatic Cellular Models

The liver is an important detoxification organ in the body and is responsible for the biotransformation and storage of toxic compounds such as exogenous xenobiotics [160]. More than 90% of orally exposed pollutants absorbed via the stomach and intestine are transported to the liver and removed from the body via biotransformation, catalyzed by UDP-glucuronosyltransferases (UGTs) and cytochromes P450 (CYPs) [161]. The liver plays a crucial role in lipid metabolism such as the synthesis and regulation of blood lipids, de novo lipogenesis, fatty acid oxidation, fatty acid uptake, and triacylglycerol export [68,162]. The excessive lipid storage in the liver may lead to lipotoxicity and NAFLD, which very often accompanies obesity [14,93]. Scientific evidence indicates that EDCs affect liver function and lipid accumulation and induce several metabolic syndromes, including hepatic steatosis and hyperlipidemia, but the mechanisms of their action still need to be explained [28,86,163,164]. Nonetheless, several studies have shown that bisphenols, including BPA, BPS and fluorene-9-bisphenol (BHPF), induce liver toxicity and hepatocyte necrosis at low dosages [165,166,167,168].

7.1. Animal Hepatocytes

7.1.1. Animal Primary Hepatocytes

Primary hepatocytes isolated from 5 week-old female CD-1 mice were used to evaluate the hepatic toxicity of BHPF [165]. Results showed that exposure to BHPF at 1 µM and 10 µM concentrations increased the lactate dehydrogenase activity (LDH) in the cell culture medium and reduced the hepatocyte numbers [165]. Cocci et al. [169] using hepatocytes from gilthead seabream (Sparus aurata L.) showed that diisodecyl phthalate (DIDP) at low concentrations (0.1 to 1 µM) upregulated genes involved in FA desaturation such as stearoyl-CoA desaturase 1A-1B (SCD1A, SCD1B) and fatty acid desaturase (FADS2), FA β-oxidation (CPT1A and CPT1B), and FA transport and metabolism (apolipoprotein A-I (APOA-I), FABP, FA synthesis and uptake (SREBP1), as well as TG and phospholipid hydrolysis (hepatic lipase (Hl) and LPL). In turn, Olsvik and Søfteland [170] reported that exposure to the 10 μM p,p’-DDE induced the expression of the markers of endocrine disruption such as Vitellogenin 1 (VTG1) and estrogen receptor 1 (ESR1) in Atlantic salmon hepatocytes. In addition, metabolomics profiling showed that 100 μM p,p’-DDE strongly affected diacylglycerol and sphingolipid metabolism, glucose and bile acid metabolism, as well as amino acid metabolism [170].

7.1.2. FaO Cell Line

The rat hepatoma (FaO) cell line is a well-characterized liver cell line used as an in vitro model of steatosis [171]. FaO cells characterize a low mRNA level of the estrogen receptor β (ERβ) and lack the estrogen receptor α (Erα) expression compared to rat liver [171]. A study by Grasselli et al. [171] showed that BPA at non-cytotoxic concentrations, 30 and 300 ng/mL, induced lipid droplet accumulation and triglyceride content in FaO cells. Additionally, decreased expression of PPARα, γ, β and δ, as well as decreased expression acyl-CoA oxidase (AOX) and CPT1 genes involved in lipid oxidation, was observed. BPA had no effect on the expression of lipogenic genes (FAS, glycerol-3-phosphate acyltransferase (GPAT). Moreover, it lowered the level of mRNA transcripts of apolipoprotein B (APOB) and extracellular triglycerides, which suggests that it may cause changes in lipid secretion [171].

7.1.3. BRL-3A Cell Line

The BRL-3A cell line is an epithelial cell line derived from buffalo rat liver [172]. Cells of this line are capable of division in the absence of serum [172]. Using BRL-3A, Zhang et al. [173] showed that MEHP induced cellular lipid accumulation and fatty acid synthesis through inhibition of the JAK2/STAT5 signaling.

7.1.4. AML12 Cell Line

The alpha mouse liver 12 (AML12) cell line is immortalized and not a tumorous cell line that is a widely used cellular model for the study of liver lipid metabolism [174]. Wu et al. [175] reported that polychlorinated biphenyls-153 (PCB-153) disturbed glucose and lipid metabolism via decreased hepatocyte nuclear factor 1b (HNF1b) expression, elevated reactive oxygen species (ROS) levels, and enhanced nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-mediated inflammation, resulting in an increased accumulation of lipid and inhibition of insulin-stimulated glucose uptake in AML-12 cells. A study by Le et al. [176] showed that chlorinated-organophosphorus flame retardants (OPFRs) such as tris (2-chloroethyl) phosphate (TCEP), tris (2-chloroisopropyl) phosphate (TCPP), tris-(2-chloro-1- (chloromethyl) ethyl) phosphate (TDCPP), triphenyl phosphate (TPhP) and tricresyl phosphate (TCP) increased the cell lipid area by 2.3-, 2.5-, 2.7-, 3.3- and 5.2-fold, respectively.

7.1.5. Hepa1-6 Cell Line

Hepa1-6 is a murine hepatoma obtained from the BW7756 hepatoma tumor that emerged spontaneously in C57L/J mice used as an in vitro clinical model for preclinical immunotherapy studies [177]. Regarding EDCs, Ke et al. [178] using Hepa1-6 hepatocytes investigated the effects of BPA at 0.001 µM, and 0.01 µM concentrations on the mRNA level of DNA methyltransferases and genes involved in lipid metabolism. BPA decreased the expression of DNA methyltransferase 1, 3-α and 3-β (DNMT1, DNMT3a, DNMT3b) but increased the FASN, 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR), SREBF1 and SREBF2 genes [178].

7.1.6. FL83B Cell Line

FL83B is a hepatocyte cell line derived from a liver of a 15–17 day-old fetal mouse [179,180]. FL83B hepatocytes actively synthesize cholesterol and store glycogen [181]. The FL83B cells have been used to study the effects of various compounds on hepatocytes’ function, including fucoxanthin [182], herbal tea extracts [183] and heavy metals such as cadmium (Cd) [184]. FL83B cells were used by Lo et al. [185] who showed that DEHP at different concentrations (125 µM, 250 µM, 500 µM, 1000 µM) injured liver FL83B cells by reducing cell viability, increasing LDH and alanine aminotransferase (ALT) release, as well as increasing cell populations of sub-G1 and S phase in a dose-dependent manner.

7.1.7. RTL-W1 Cell Line

RTL-W1 is the epithelial cell line derived from the liver of 4 year-old male rainbow trout [186,187]. This in vitro model was applied to study the ability of EDCs, namely TBT, terpyridine platinum(II) chloride (TPT), 4-nonylphenol (4-NP), BPA and DEHP, to alter the expression of markers of cellular lipid metabolism leading to steatosis in fish [186]. Results presented by Dimastrogiovanni et al. [186] showed that DEHP and BPA significantly increased the accumulation of lipids in RTL-W1 cells, whereas TBT, 4-NP, BPA and DEHP altered membrane lipids such as phosphatidylcholines (PCs) and plasmalogen PCs. Furthermore, RTL-W1 cells exposed to BPA, TBT, TPT, DEHP and 4-NP altered mRNA levels of ATP-binding cassette transporter A1 (ABCA1), CD36, fatty acid transport protein 1 (FATP1), FAS, LPL, PPARα and PPARβ [186].

7.1.8. PLHC-1 and ZFL Cell Line

The fish hepatoma PLHC-1 cell line has been derived from topminnow (Poeciliopsis lucida) [188,189] whereas the zebrafish liver cell line (ZFL) has been isolated from zebrafish (Danio rerio) [188]. These cells maintain several differentiated cell functions of hepatocytes and have been extensively used to assess the cytotoxicity and changes in gene transcription associated with xenobiotic exposure [188,189]. Regarding the effects of EDCs, Marqueño et al. [190] reported that exposure to BPA, BPF and bisphenol A bis(3-chloro-2-hydroxypropyl) ether (BADGE·2HCl) induced the accumulation of ether-triacylglycerides (ether-TGs) and dihydroceramides in hepatic ZFL cells. Moreover, BPA and BADGE·2HCl increased the level of saturated TGs and lowered the levels of unsaturated TGs. Concentrations of 20 µM BPA and 20 µM BPF led to an increase in the expression of the lipogenic genes such as SCD and ELOVL fatty acid elongase 6 (ELOVL6), while the PPARα mRNA level was down-regulated by 20 µM BPF and 5 µM BADGE·2HCl [190]. In the PLHC-1 cells, exposure to BADGE·2HCl induced a strong decrease of triacylglycerides (TGs), while DEHP and dibutyl phthalate (DBP) stimulated the accumulation of TGs [191]. Furthermore, the effect of TBT on the dysregulation of lipid metabolism in PLHC-1 and ZFL cells, as well as the alteration of the FASN, SCD, and ELOVL6 expression in ZFL cells, was also reported [188].

7.2. Human Hepatocytes

Since the liver is the main organ involved in the metabolism and the toxicity of xenobiotics, isolated primary human hepatocytes (PHHs) have been increasingly used as a model in pharmaco-toxicological studies for the detection of toxic chemicals and evaluating their mechanism of toxicity [192,193]. While PHHs represent a valuable tool for studying liver function, the main limitation of their utilization is the restricted accessibility, heterogeneity, phenotypic instability and limited time for cell proliferation in in vitro culture [193,194]. Therefore, alternative hepatocyte models have been explored and used, including cells from human liver tumors or immortalized adult or fetal human hepatic cells [193]. The advantage of these cell lines is their unlimited availability and rapid growth, but they are dedifferentiated, and compared to normal adult hepatic cells, they show less liver-specific metabolism [195].

7.2.1. Human Primary Hepatocytes

Primary hepatocytes isolated from cancer-free portions of the liver after resection were used to assess the level of FA accumulation upon 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure [90]. TCDD increased total FAs in hepatocytes, including stearate, palmitate, oleic and linoleic acids after 48 h exposure at a 10 nM concentration [90].

7.2.2. HepG2 Cell Line

HepG2 is the best-characterized human hepatoma cell line [194]. This cell-based model is cost-effective, easy to handle, and ensures the repeatability of the obtained results [194]. Regarding the differences between the HepG2 cell line and normal hepatocytes, HepG2 cells do not possess the complete set of xenobiotic-metabolizing enzymes (XMEs), especially UDP-glucuronosyltransferases (UGTs) and some cytochromes P450 (CYPs) such as CYP2A6, CYP2D6, CYP3A4, CYP2C9, CYP2C19, etc., that are involved during phase I of xenobiotic oxidation in the liver [164,196,197]. Despite this, HepG2 cells retain most of the metabolic functions performed by normal hepatocytes, which allows them to be used in studies of the toxic effects of drugs, nanoparticles, and heavy metals in vitro [198]. The HepG2 cell line has been applied in hepatotoxicity assessments [194] and has been used to evaluate the link between EDC exposure and fatty liver disease associated with an increased risk of obesity [93]. Regarding the surfactants, Sun et al. [93] reported that exposure to 4-HP increased lipid accumulation in oleic acid (OA)-treated HepG2 cells and inhibited de novo lipogenesis by decreasing the acetyl-CoA carboxylase (ACC) and SREBP1c expression as well as the fatty acid oxidation by decreasing the PPARα and CPT1A mRNA levels. Moreover, 4-HP accelerated the uptake process of OA in hepatocytes by an increase of the CD36 mRNA level [93]. In turn, Lu et al. [199] reported that 1,3-dichloro-2-propanol (1,3-DCP) at a 0.5 to 2 µg/mL concentration increased lipid droplet accumulation as well as total cholesterol (TC) and TGs content in HepG2 cells. The molecule 1,3-DCP considerably increased the mRNA level of LDLR, SREBP2 and HMGCR, associated with lipid metabolism [199]. Furthermore, an increase in the lipid accumulation in HepG2 cells exposed to pentabromotoluene (PBT), hexabromocyclododecane (HBCD), and tetrabromobenzoate (TBB) was also reported by Maia et al. [200]. Recently, Vasconcelosa, Silva and Louro [57] showed that DINCH as a non-phthalate plasticizer induced oxidative DNA damage in HepG2 cells, which can be correlated to numerous human diseases including diabetes and cardiovascular disease.

7.2.3. HepaRG Cell Line

HepaRG is an immortalized hepatic cell line that has a similar expression of nuclear receptors, key metabolic enzymes (XMEs), and drug transporters as primary human hepatocytes [201]. HepaRG is a good model used in the field of toxicology because HepaRG cells can enter into a differentiation program toward hepatocyte-like and biliary-like cells [164]. Regarding pesticides, Stossi et al. [202] showed that TBT induced lipid accumulation in HepaRG cells via increased mRNA levels of SREBF1 and FASN involved in de novo lipogenesis. On the other hand, HepaRG cells exposed to BPA had significantly greater cellular triglyceride and neutral lipid accumulation at a 2 nM concentration [164]. BPA induced hepatic lipid accumulation by increasing the apolipoprotein A4 (APOA4) mRNA level, whereas no effect on perilipin 3 (TIP47) and perilipin 2 (PLIN2) gene expression, involved in lipid droplets accumulation, nor for genes associated with carbohydrate homeostasis, was observed [164].

7.2.4. HPR116 Cell Line

HPR116 cells, which are differentiated HepaRG cells, are used to save time in the experiment because they are ready-to-use and easy to use. Cells from the same batch show a repeated differentiation level and have the same behaviour. In addition, HPR116 cells are long living, remaining viable and usable for at least two weeks. They exhibit responses and functions similar to those of primary hepatocytes [203].

7.2.5. Huh-7 Cell Line

Huh-7 (human hepatoma) is an immortalized cell line consisting of tumorigenic cells [204]. Wada et al. [205] showed that BPA and 4-NP exposure stimulated lipid accumulation in Huh-7 cells. Similarly, Lee et al. [206] confirmed that BPA increased intracellular lipid accumulation and fatty acid uptake in Huh-7 cells.

7.2.6. Huh-6 Cell Line

Fetal HuH6 hepatocytes from a hepatoblastoma of a 1 year-old male donor are also used in in vitro tests of the endocrine-disrupting effects of different substances [207,208]. The advantages of this cell line include availability, unlimited growth, high reproducibility of results, and the expression of enzymes involved in the metabolism of xenobiotics [209].

7.2.7. HHL-5 Cell Line

HHL-5 is an immortalized human hepatocyte cell line. Its phenotype resembles primary hepatocytes [210]. Martella et al. [210] demonstrated that exposure of HHL-5 cells to BPA induced FA accumulation in an endocannabinoid receptor type 1 (CB1)-dependent manner. BPA increased CB1 activity by stimulating the synthesis of anandamide (N-arachidonoylethanolamine; AEA) [210].

7.2.8. L02 Cell Line

The human normal liver cell line L02 is used to study the effects of various compounds on lipogenesis [211]. The L02 cell line was derived from primary normal human hepatocytes and immortalized in 1980 [212]. This cell line has found applications in the research of human hepatocellular functions such as drug hepatotoxicity, hepatic steatosis, and chemical carcinogenesis [212]. Zhang et al. [213] showed that triclosan (TCS), widely used as an antibacterial and antifungal agent, promoted the perturbation of intracellular lipids in L02 cells.
The obesogenic effects of selected EDCs confirmed in hepatic cellular models are summarized in Table 3.

8. Pancreatic Cellular Models

The α and β cells of the pancreas play an important role in blood glucose control through the secretion of glucagon and insulin [14]. Glucagon secreted by α-cells is involved in the synthesis and mobilization of glucose in the liver [214]. The insulin secreted by β-cells reduces blood glucose levels via increasing glucose uptake by insulin-sensitive tissues, such as the liver, adipose tissue and skeletal muscle, and inhibiting hepatic glucose production [68]. Type 2 diabetes mellitus (T2DM) is characterized by hyperglucagonemia and hypoinsulinemia, which results in increased blood glucose levels [14]. T2DM is comprised of a series of interrelated abnormalities such as insulin resistance (IR) and metabolic syndrome [28]. IR in WAT, skeletal muscle and liver combined with inappropriate insulin secretion from pancreatic β cells is the major cause of human T2DM [68]. Recently, an increasingly significant role in the development of T2DM is attributed to EDCs [215]. Moreover, EDCs may be of great importance in the pathogenesis of Type 1 diabetes, especially during the developmental period [23].

8.1. Animal Pancreatic Cells

8.1.1. Rat Pancreatic Islets

Pancreatic islets isolated from male Wistar rats at 8 weeks of age were used to investigate the effects of acute and long-term exposure to BPA and NP on insulin secretion [216]. Adachi et al. [216] showed that acute exposure (60 min) to BPA and NP at 0.1, 1, 10 and 100 µg/L did not affect insulin secretion in pancreatic cells with glucose stimulation. In turn, 24 h exposure to BPA (10 and 100 µg/L) or NP (0.1, 1, 10 and 100 µg/L) with 16.7 mM glucose significantly increased insulin secretion via cytosolic/nuclear estrogen receptors [216]. Ghaemmaleki et al. [217] reported that 10 µM TBT reduced the viability of the pancreatic islets of Langerhans isolated from 2–3 month-old male Wistar rats by 50%. In addition, increased insulin secretion at both basal (2.8 mM) and stimulatory (16.7 mM) concentrations of glucose after 10 µM TBT exposure was observed [217].

8.1.2. INS-1 Cell Line

The INS-1 cells were isolated from a rat insulinoma induced by X-ray irradiation and are applied in the studies of insulin secretory mechanisms. INS-1 cells are bi-hormonal and are capable of expressing both insulin and proglucagon proteins [218]. There is strong evidence that BPA in the concentration range from 0.002 to 2 µM lowers the viability of INS-1 cells and increases apoptosis via a mitochondria-mediated pathway in a dose-dependent manner [219]. Interestingly, BPA at higher concentrations (0.2 and 2 µM) significantly decreased insulin secretion in response to glucose stimulation, but at 0.002 µM slightly increased insulin secretion [219].

8.1.3. INS-1E Cell Line

The INS-1E cell line is a stable rat insulinoma pancreatic β-cell line cloned from the INS-1 cells, characterized as less heterogeneous than the INS-1 cell line [23]. INS-1E cells are widely used in studies of β cell function [23]. It has been reported that BPA (1 µM) and TBT (200 nM) decreased INS-1E cell viability by inducing apoptosis [23]. In addition, TBT caused a reduction in the expression of the MAFA, which is a transcription factor regulating the expression of genes involved in the biosynthesis and secretion of insulin (such as pancreatic and duodenal homeobox 1 (PDX1) and glucokinase (GCK)), as well as Pdx1. Moreover, triphenylphosphate (TPP), PFOA (perfluorooctanoic acid), TCS and DDE at a concentration of up to 1 µM did not affect the viability of INS-1E cells, nor the expression level of genes involved in insulin biosynthesis and secretion [23]. Another study showed that p,p’-DDT (10 µM) and its metabolite p,p’-DDE (10 µM) reduced the intracellular level of proinsulin (precursor of insulin) and insulin monomer (the active form of insulin), as well as decreased insulin 1 (INS1) and 2 (INS2) mRNA levels in INS-1E cells [220]. Additionally, p,p’-DDT decreased the intracellular level of hexameric insulin, which is a final form of insulin secreted by pancreatic β cells. p,p’-DDT decreased the expression of actin and mortalin/GRP75 and increased the expression of tubulin beta-5 chain, annexin A4, and vitamin D-binding protein (VDBP). In turn, p,p’-DDE also increased VDBP expression but decreased glucosidase 2 subunit beta (GLU2β) precursor expression [220].

8.1.4. RIN-m5F Cell Line

The RIN-m5F cell line is a rat pancreatic β-cell line that can produce and secrete insulin [106,221]. Chen et al. [106] demonstrated that TBT at the concentration between 0.05 and 0.2 µM did not affect the viability of RIN-m5F cells after 24 h incubation, but increased glucose-stimulated insulin secretion (GSIS) in β cells after 0.1 and 0.2 µM TBT treatment. In a study reported by Huang et al. [221], TBT at a 0.5 µM dose increased the number of apoptotic RIN-m5F cells after 24 h incubation, which was associated with the phosphorylation of mitogen-activated protein kinases (MAPKs)-c-Jun N-terminal kinase (JNK), as well as extracellular signal-regulated protein kinase (ERK1/2) and poly (ADP-ribose) polymerase (PARP) cleavage. Furthermore, TBT (0.5 µM, 1 µM) was found to significantly decrease GSIS after 24 h treatment [221]. In turn, Suh et al. [222] reported that PFOA at the 100–500 µM concentration range significantly decreased the viability of RIN-m5F cells and increased their apoptosis. It turned out that this EDC caused oxidative stress and mitochondrial dysfunction via the reduction of adenosine triphosphate (ATP) level, as well as induction of cardiolipin peroxidation, mitochondrial membrane potential collapse as well as cytochrome c release [222].

8.1.5. Mouse Pancreatic Islets

The mouse islets derived from adult male mice (12–14 weeks old) have been used by Dos Santos et al. [23] who demonstrated that BPA (1 nM, 1 µM) and TBT (20 nM, 200 nM) promoted apoptosis in dispersed mice islets. Carchia et al. [105] showed that BPA at a low dose (0.001 µM) changed the functioning of primary murine pancreatic islets and glucose homeostasis. BPA led to the dysfunction of the mitochondria and their destruction by inhibiting the expression of genes important in mitochondrial activity. This caused a decrease in insulin secretion by β cells after 1 h glucose stimulation (16 mM), as well as a decrease in the viability of these cells [105]. Soriano et al. [223] reported that BPA at a concentration of 1 nM increased insulin secretion and decreased ATP-sensitive K+ (KATP) channel activity in β cells from wild-type (WT) mice (C57), which was not recorded in cells from estrogen receptor β (ERβ) knockout (ERβ -/-) mice (BERKO mice). Recently, Marroqui et al. [224] showed that BPS and BPF at 1 nM and 1 µM concentrations increased insulin secretion and lowered KATP channel activity (1 nM BPS, 10 nM BPF) in pancreatic β cells from WT mice (C57BL/6J). In turn, Chen et al. [106] reported that 0.1 µM and 0.2 µM TBT increased GSIS in isolated mouse islets.

8.1.6. MIN-6 Cell Line

The MIN6 cell line is derived from a mouse insulinoma. This transformed β-cell line retains GSIS and is used to study insulin secretion [225,226]. Nonetheless, it should be noted that the long-term culture of MIN6 cells results in the loss of their insulin secretory capacity in response to glucose [227]. This is probably because β-cells dominating in the culture respond poorly to glucose or due to the increased expression of genes responsible for GSIS changes over time [228]. Al-Abdulla et al. [229] showed that 100 nM BPA increased GSIS. Moreover, upregulation of, e.g., MAFA, hepatocyte nuclear factor 4 alpha (HNF4α) and PDX1, which are important for insulin secretion and normal glucose sensing, was observed in cells treated with BPA. In turn, BPS, DEHP, perfluorooctanesulfonic acid (PFOS) and DDE decreased insulin release, while cadmium chloride (CdCl2) had no effect on GSIS in MIN-6 cells [229].

8.1.7. β-TC-6 Cell Line

The beta-tumor cell-6 (β-TC-6) cell line is a mouse islet β-cancer cell line derived from a transgenic mouse expressing genes encoding insulin, glucagon and somatostatin; β-TC-6 cells are capable of secreting insulin in response to glucose [230]. The β-TC-6 cell line was applied in the study reported by Qin et al. [231] who showed that exposure to 50 µM and 100 µM PFOS stimulated GSIS in β-TC-6 cells and increased intracellular calcium levels via G protein-coupled receptor 40 (GPR40) activation. In another study [232], 10 µM p,p’-DDE significantly increased basal and GSIS in β-TC-6 cells. Scientists speculated that this EDC does not affect insulin transcription because it does not increase the levels of PDX1 that regulates insulin gene transcription. Probably, DDE alters insulin translation by increasing the level of prohormone convertase (PC), which is involved in the cleavage of insulin to its mature form [232].
The literature also describes other animal insulin-secreting cell lines used in diabetes mellitus research such as murine cell lines (β-TC-1, β-TC-3, IgSC195, βHC, NIT-1), rat cell lines (RINm, RINr, BRIN-BG5, BRIN-BG7, BRIN-BD11, CRI-G1, CRI-G1-RS, In-111) and hamster pancreatic β-cell lines (HIT-T15) [227,233], but to the best of our knowledge, the effect of EDC exposure on the development of obesity and metabolic disorders using these in vitro models has not yet been investigated.

8.2. Human Pancreatic Cells

8.2.1. Human Pancreatic Islets

Studies conducted on β-cells from the islets of Langerhans from different human donors demonstrated that 1 nM BPA decreased KATP channel activity (closure of KATP channels), which contributed to an increase in GSIS [223]. Moreover, Chen et al. [106] reported that 0.1 µM TBT significantly increased GSIS in human islets from patients with benign pancreatic tumors.

8.2.2. EndoC-βH1 Cell Line

The EndoC-βH1 is a human cell line widely used in diabetes and islet biology research [23]. These cells express all genes that determine the primary β-cells phenotype. However, unlike primary cells, EndoC-βH1 cells may show a different expression of disallowed genes and some β-cell markers and contain approximately 5–10% of the insulin present in native β-cells [23]. Moreover, EndoC-βH1 cells have a greater ability to proliferate than adult human β-cells and present similar insulin secretion in response to glucose in human islets [23].
Using EndoC-βH1cells with BPA and TBT as positive controls, Dos Santos et al. [23] evaluated the adverse effects of PFOA, TPP, TCS, and DDE exposure on β-cell viability and GSIS. Results showed that 1 µM of the DDE, TCS, TPP, and PFOA did not affect the viability, whereas higher concentrations of PFOA (20 to 200 µM) induced apoptosis in the β-cells upon 24 h treatment. In contrast, 1 µM BPA and 200 nM TBT reduced the cell viability and induced the apoptosis of the β-cells. BPA and TCS did not affect GSIS whereas TPP, DDE, and TBT increased insulin secretion. Interestingly, PFOA decreased insulin secretion both at high and low glucose concentrations. All tested compounds, except TBT, did not modify the insulin content. PFOA, BPA, TCS, TPP, and DDE did not affect the expression of genes related to insulin biosynthesis and secretion in comparison to TBT, which increased the glucose transporter type 2 (GLUT2) expression in EndoC-βH1 cells [23]. Al-Abdulla et al. [229] investigated the effects of BPA, BPS, BPF, PFOS, DEHP, CdCl2 and DDE exposure at different concentrations ranging from 100 pM to 10 μM on human pancreatic β-cell function. BPA, PFOS and CdCl2 treatment resulted in a marked increase in GSIS, whereas a decrease in insulin secretion in EndoC-βH1 cells upon BPS and DEHP exposure was observed. BPF and DDE had no effect on insulin release. Regarding BPS, a significant decrease of the GLUT1, MAFA, MAFB, synaptosome-associated protein 25 (SNAP25) and KIR6.2 mRNA levels in pancreatic β-cells was also noticed [229].

8.2.3. NES2Y Cell Line

The NES2Y is a human pancreatic β-cell line characterized by a constitutive insulin release and possesses an insulin promoter unresponsive to changes in glucose levels [234]. NES2Y cells are proliferative, lack functional ATP-sensitive potassium channels (KATP), and also carry a defect in the insulin gene-regulatory transcription factor (PDX1) [234].
Regarding EDCs, Pavlikova et al. [234] verified that p,p’-DDT and p,p’-DDE at 100 μM concentrations induced a time-dependent inhibition of pancreatic β-cell proliferation and observed that 10 µM p,p’-DDT after 1 month of exposure downregulated the levels of the three cytoskeletal proteins (actin (ACTB), cytokeratin 18 (CK18) cytokeratin 8 (CK8)) and alpha-enolase (ENO1), involved in glycolysis. In turn, 10 µM of p,p’-DDE decreased the expression of heterogeneous nuclear ribonucleoprotein H1 (HNRH1) and CK18 [234]. Moreover, it has been shown that high concentrations of DDT (150 µM, 175 µM, 200 µM) reduced NES2Y cell viability after 24 h of exposure. In addition, NES2Y cells exposed to 150 μM DDT showed decreased levels of 22 proteins such as mitochondrial proteins (enoyl-CoA hydratase (ECHM), 75 kDa glucose-regulated protein (GRP75), NADH dehydrogenase (ubiquinone) iron-sulfur protein 3 and 1 (NDUS3 and NDUS1), proteins involved in the endoplasmic reticulum (ER) stress (endoplasmin, 78 kDa glucose-regulated protein (GRP78)), proteins associated with maintenance of the cell morphology (T-complex protein 1 subunit alpha (TCPA), ezrin, EF-hand domain-containing protein (D2EFHD2), N-myc downstream regulated 1 (NDRG1)), as well as other proteins such as heat shock protein 27 (HSP27), polysaccharide biosynthesis domain-containing 1 (PBDC1) and proliferating cell nuclear antigen (PCNA) [107].

8.2.4. PANC-1 Cell Line

The PANC-1 cell line is an epithelioid carcinoma cell line derived from the human pancreas [235] and is widely used as a human model of pancreatic cells because of the cells’ ability to secrete insulin in response to high amounts of glucose in the culture medium [236]. Using the PANC-1 line, Menale et al. [124] reported that 10 nM BPA effectively impaired insulin secretion in the exposed cells via downregulation of the proprotein convertase subtilisin/kexin type 1 (PCSK1) expression gene involved in insulin production. Other human pancreatic beta-cell lines such as CM, TRM-1, and Blox5 have also been used in adipogenic differentiation studies [227]. However, to the best of our knowledge, they have not yet been applied to study the impact of EDCs on the adipogenesis process.
The obesogenic effects of selected EDCs on pancreatic cellular models are summarized in Table 4.

9. Conclusions

In this review, we have made extensive insight into the available literature regarding animal and human 2D in vitro cell models applied to evaluate the obesogenic action of various environmental EDCs. According to the available literature, animal in vitro cell models especially from rodents are extensively used for assessing adipogenesis and applied to screen environmental obesogens. The main advantage of animal in vitro cell models is their commercial availability and well-established protocols for their cultivation. Nonetheless, the translation from animal models is limited by their metabolic heterogeneity, especially between murine and human types. To reduce this risk, human primary cells isolated from adipocyte tissue and crucial endocrine organs such as the liver and pancreas are increasingly used. The main advantage of these cells is comparable identity and functions to native tissue, but their maintenance under in vitro conditions still remains a challenge, because such cells are characterized by a limited number of divisions, after which they lose their biochemical functionality and undergo programmed cell death. Recently, there has been a growing interest in the derivation of hADSCs that does not require cell transformation. The main advantage of hADSCs is the possibility of studying the exposure of chemical compounds on adipo- and lipo-genesis mechanisms by omitting the extrapolation step.
It should be noted that in vitro models are a relatively simplified system compared to the complexity of a living organism and its response to exogenous factors. It is difficult to estimate what concentration of a given compound in vitro corresponds to an in vivo dose, as well as to analyze interactions between different cell types and simulate the effects of long-term exposure to a given compound in cellular models. In addition, it is also worth noting that in vitro tests can provide unreliable results, e.g., in the case of examining the obesogenic effect of phthalates, as they are present in some laboratory plastics. Moreover, in studies with the use of cell models, not only should individual EDCs be tested, but also EDC mixtures, which reflect the “real-life” obesogenic effect much better.
In summary, in vitro studies carried out mainly on cell cultures or isolated tissue samples are used extensively to investigate the mode of action of possible industrial obesogens. While in vitro models have limitations that must be resolved, they are generally simpler, more cost-effective and can be performed in a large series of experiments under the same conditions. Nonetheless, for a better understanding of the mechanisms of the obesogenic EDCs, all information from in vitro and in vivo models should be combined.

Author Contributions

Conceptualization, M.K. and E.J.-K.; investigation, M.K., E.J.-K., J.P.P., A.W., P.Ś. and M.W.; writing—original draft preparation, M.K. and E.J.-K.; writing—review and editing, E.J.-K.; visualization, A.W., M.K. and E.J.-K.; supervision, E.J.-K. and J.P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This article was done under statutory project “The effect of phthalates as environmental obesogens on epigenetic modifications of the endocrine activity of adipose tissue in the context of human obesity and metabolic disorders—in vitro study” no. ZM-103-01, supported by Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology—State Research Institute.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This manuscript constitutes part of Monika Kowalczyk’s doctoral thesis, performed in the “AgroBioTech PhD” Doctoral Program of the Prof. Wacław Dąbrowski Institute of Agricultural and Food Biotechnology—State Research Institute.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

1,3-DCP1,3-dichloro-2-propanol
11β-HSD111β-hydroxysteroid dehydrogenase type 1
4-HP4-hexylphenol
4-NP4-nonylphenol
AACSacetoacetyl-CoA synthetase
ABCA1ATP-binding cassette transporter A1
ACACAacetyl-CoA carboxylase α
ACAT3acetyl-coenzyme A acetyltransferase 3
ACCacetyl-CoA carboxylase
ACC2acetyl-CoA carboxylase 2
ACLYadenosine triphosphate citrate lyase
ACTBactin
Act-Dactinomycin-D
ADIPOQadiponectin, C1Q and collagen domain containing
ADIPOR2adiponectin receptor 2
ADPadenosine diphosphate
ADSCsanimal adipose-derived stem cells
AEAanandamide (N-arachidonoylethanolamine)
AKT2v-akt murine thymoma viral oncogene homolog 2
ALTalanine aminotransferase
AML12alpha mouse liver 12
AMPadenosine monophosphate
AMPKAMP-activated protein kinase
AMPKαAMP-activated protein kinase-α
AOXacyl-CoA oxidase
AP2adipocyte protein 2
APAF1apoptotic protease activating factor 1
APM1adipocyte most abundant gene transcript-1
APOA1BPapolipoprotein A1-binding protein
APOA2apolipoprotein A2
APOA4apolipoprotein A4
APOA-Iapolipoprotein A-I
APOBapolipoprotein B
APOEapolipoprotein E
ASCsadipose-derived stem cells
ASTaspartate aminotransferase
ATadipose tissue
ATGLadipose triglyceride lipase
ATPadenosine triphosphate
ATP1B1ATPase Na+/K+ transporting subunit beta 1
ATP6ATP synthase subunit 6
ATP6v1FATPase H+ transporting V1 subunit F
BADGE2HCl bisphenol A bis(3-chloro-2-hydroxypropyl) ether
BATbrown adipose tissue
BAXBCL2 associated X apoptosis regulator
BBPbenzyl butyl phthalate
BCL-2apoptosis regulator Bcl-2
BHPFfluorene-9-bisphenol
BIEAbiliverdin reductase
BMIbody mass index
BMMSCbone marrow-derived mesenchymal stem cells
BPAbisphenol A
BPAFbisphenol AF
BPBbisphenol B
BPFbisphenol F
BPSbisphenol S
C/EBPCCAAT/enhancer-binding protein
C/EBPαCCAAT/enhancer-binding protein α
C/EBPβCCAAT/enhancer-binding protein β
C/EBPδCCAAT/enhancer-binding protein δ
CACNA1Ecalcium voltage-gated channel subunit alpha1 E
CaMKIICa2+/calmodulin-dependent protein kinase II
cAMPcyclic adenosine monophosphate
CASP3Bcaspase 3B
CATcatalase
CB1endocannabinoid receptor type 1
CBScystathionine-beta-synthase
CCL20chemokine (C-C motif) ligand 20
Cdcadmium
CD36fatty acid translocase
CdCl2cadmium chloride
CDKN1Bcyclin-dependent kinase inhibitor 1B
Cerceramide
CHOPC/EBP homologous protein
CIDEAcell death-inducing DNA fragmentation factor-alpha-like effector A
CK18cytokeratin 18
CK8cytokeratin 8
CNR1cannabinoid receptor 1
COX16cytochrome C oxidase assembly factor
CPFchlorpyrifos
CPT1carnitine palmitoyltransferase 1
CPT1Acarnitine palmitoyltransferase 1 a
CPT1Bcarnitine palmitoyltransferase 1 b
CREBcAMP response element-binding protein
CYP1A1cytochrome P450 family 1 subfamily A member 1
CYP2C18cytochrome P450 family 2 subfamily C member 18
CYP2C19cytochrome P450 family 2 subfamily C member 19
CYP2C9cytochrome P450 family 2 subfamily C member 9
CYP3Acytochrome P450 family 3 subfamily A
CYP3A4cytochrome P450 family 3 subfamily A member 4
CYP3A65cytochrome P450 family 3 subfamily A polypeptide 65
CYPscytochromes
D2EFHD2EF-hand domain-containing protein
DBPdibutyl phthalate
DCHPdicyclohexyl phthalate
DDEdichlorodiphenyldichloroethylene
DDTdichlorodiphenyltrichloroethane
DEHPdi(2-ethylhexyl) phthalate
DEXdexamethasone
DGdiacylglycerol
DGAT1diacylglycerol acyltransferase 1
DGAT1Adiacylglycerol acyltransferase 1a
DGAT2diacylglycerol acyltransferase 2
DIDPdiisodecyl phthalate
DINCHbis(7-methyloctyl) cyclohexane-1,2-dicarboxylate
DINPdiisononyl phthalate
DLKdual leucine zipper-bearing kinase
DNMT1DNA (cytosine-5-)-methyltransferase 1
DNMT3ADNA methyltransferase 3 alpha
DNMT3AADNA (cytosine-5-)-methyltransferase 3A
DNMT3BDNA methyltransferase 3 beta
DPHPbis(2-propylheptyl) phthalate
ECHMenoyl-CoA hydratase
EDCsendocrine-disrupting chemicals
EF2elongation factor 2
EFHD2EF-hand domain family member D2
ELOVL6ELOVL fatty acid elongase 6
ENO1α-enolase
ERstress endoplasmic reticulum stress
ERK1/2extracellular signal-regulated protein kinase 1/2
ERRγestrogen-related receptor γ
ERsestrogen receptors
ERαestrogen receptor α
ERβ -/- (or BERKO) miceestrogen receptor β knockout mice
ERβestrogen receptor β
ESR1estrogen receptor 1
ESR2estrogen receptor 2
ESRRAestrogen related receptor alpha
ether-TGsether-triacylglycerides
EUEuropean Union
EurostatStatistical Office of the European Union
EZRIezrin
FAfatty acid
FAAHfatty acid amide hydrolase
FABPfatty acid binding protein
FABP4fatty acid binding protein 4
FABP5fatty acid binding protein 5
FADS1fatty acid desaturase 1
FADS2fatty acid desaturase
FASfatty acid synthase
FASNfatty acid synthase
FATP1fatty acid transport protein 1
FOSFos proto-oncogene, AP-1 transcription factor subunit
FOXO1forkhead box protein O1
FRILferritin light chain
FSP27fat-specific protein 27
G3BP1Ras GTPase-activating protein-binding 1
GCKglucokinase
GDF15growth differentiation factor 15
GLU2βglucosidase 2 subunit beta
GLUT1glucose transporter type 1
GLUT2glucose transporter type 2
GLUT4glucose transporter type 4
GLYglyphosate
GPATglycerol-3-phosphate acyltransferase
GPATglycerol-3-phosphate acyltransferase
GPAT3glycerol-3-phosphate acyltransferase 3
GPD1glycerol-3-phosphate-dehydrogenase
GPR109Bprotein-coupled receptor 109B
GPR30G protein-coupled receptor 30
GPR40G protein-coupled receptor 40
GPR41G protein-coupled receptor 41
GPR43G protein-coupled receptor 43
GPX1glutathione peroxidase 1
GPX3glutathione peroxidase 3
GPX4glutathione peroxidase 4
GPX8glutathione peroxidase 8
GRglucocorticoid receptor
GRP7575 kDa glucose-regulated protein
GRP7878 kDa glucose-regulated protein
GSHglutathione
GSISglucose-stimulated insulin secretion
GSRglutathione-disulfide reductase
GSSGoxidized glutathione
GSTA1/2glutathione S-transferase alpha ½
GSTA3glutathione S-transferase alpha 3
GSTO1glutathione S-transferase omega-1
GUSBglucuronidase beta
hADSCshuman adipose-derived stem cells
HBCDhexabromocyclododecane
HLhepatic lipase
HMGCR3-hydroxy-3-methylglutaryl CoA reductase
HMOX1heme oxygenase 1
HNF1Bhepatocyte nuclear factor 1b
HNF4αhepatocyte nuclear factor 4 alpha
HNRH1heterogenous nuclear ribonucleoprotein H1
HNRPFheterogeneous nuclear ribonucleoprotein F
HSD11B111beta-hydroxysteroid dehydrogenase 1
HSLhormone-sensitive lipase
HSP27heat shock protein 27
HSPA1Aheat shock protein family A (Hsp70) member 1A
HSPA8heat shock protein family A (Hsp70) member 8
hTERThuman telomerase reverse transcriptase
IBMXphosphodiesterase inhibitor 1-methyl-3-isobutyl xanthine
IDH3Aisocitrate dehydrogenase (NAD(+)) 3 catalytic subunit alpha
IFN-γinterferon gamma
IGF1insulin-like growth factor 1
IL18interleukin 18
IL1B(IL1β) interleukin 1 beta
IL1αinterleukin 1 alpha
IL6interleukin 6
INS1insulin 1
INS2insulin 2
INSIG1insulin induced gene 1
INSRinsulin receptor
IRinsulin resistance
IRS-1insulin receptor substrate 1
IRS-2insulin receptor substrate 2
IR-βinsulin receptor subunit β
JAK2Janus kinase 2
JNKc-Jun N-terminal kinase
K2C8keratin type II cytoskeletal 8
KATPchannel ATP-sensitive K+ channel
KCNIPpotassium voltage-gated channel interacting protein
KCNIP1potassium voltage-gated channel interacting protein 1
KCNMA1potassium calcium-activated channel subfamily M alpha 1
LAP3leucine aminopeptidase 3
LDHlactate dehydrogenase
LDLRlow-density lipoprotein receptor
LEPleptin
LEPRleptin receptor
LIPElipase E
LKB1serine-threonine liver kinase B1
LPClysophosphatidylcholine
LPElysophosphatidylethanolamine
LPLlipoprotein lipase
LSD-1lysine-specific demethylase-1
LXRliver X receptor
MAPKsmitogen-activated protein kinases
MAT1A2methionine adenosyltransferase 1A2
MCP1monocyte chemoattractant protein-1
MDAmalondialdehyde
MDCsmetabolism-disrupting chemicals
MEHPmono-2-ethylhexyl phthalate
MHINPmonohydroxy isononyl phthalate
MINCH1,2-cyclohexanedicarboxylic acid mono 4-methyloctyl ester
MMPmitochondrial membrane potential
MSCsmesenchymal stem cells
mTORmammalian target of rapamycin
MTTPmicrosomal triglyceride transfer protein
N6AMT2N-6 adenine-specific DNA methyltransferase 2
NAFLDnon-alcoholic fatty liver disease
NAMPTnicotinamide phosphoribosyltransferase
NANOGnanog homeobox
ND4LNADH-ubiquinone oxidoreductase subunit 4 L
NDRG1N-myc downstream regulated 1
NDUFS4NADH: ubiquinone oxidoreductase subunit S4
NDUS1NADH dehydrogenase [ubiquinone] iron-sulfur protein 1
NDUS3NADH dehydrogenase [ubiquinone] iron-sulfur protein 3
NF-κBnuclear factor kappa-light-chain-enhancer of activated B cells
NOnitric oxide
NPC2Niemann-Pick 2
NQO1NAD(P)H quinone dehydrogenase 1
NRF1nuclear respiratory factor 1
NRF2nuclear respiratory factor 2
NRsnuclear receptors
OAoleic acid
OCT4octamer-binding transcription factor 4
OEAoleoylethanolamide
OGDHoxoglutarate dehydrogenase
OH-MPHP6-hydroxy monopropylheptyl phthalate
OPFRschlorinated-organophosphorus flame retardants
p,p’-DDE1,1-dichloro-2,2-bis(4-chlorophenyl)ethane
p,p’-DDT1,1,1-trichloro-2,2-bis (p-chlorophenyl)-ethane
pACCphosphorylated acetyl-CoA carboxylase
p-AKTphosphorylated v-akt murine thymoma viral oncogene homolog
p-AMPKphosphorylated AMP-activated protein kinase
PARPpoly (ADP-ribose) polymerase
PBDC1polysaccharide biosynthesis domain containing 1
PBTpentabromotoluene
PCprohormone convertase
PCB-153polychlorinated biphenyls-153
PCK1phosphoenolpyruvate carboxykinase 1
PCNAproliferating cell nuclear antigen
PCsphosphatidylcholines
PCSK1proprotein convertase subtilisin/kexin type 1
PDK4pyruvate dehydrogenase kinase 4
PDX1pancreatic and duodenal homeobox 1
PEphosphatidylethanolamine
PEApalmitoylethanolamide
p-ERK1/2phosphorylated extracellular signal-regulated protein kinase ½
PFOAperfluorooctanoic acid
PFOSperfluorooctanesulfonic acid
PGphosphatidylglycerol
PHHsprimary human hepatocytes
PIpolyphosphoinositide
p-JNKphosphorylated c-Jun N-terminal kinase
PKAprotein kinase A
PKBprotein kinase B (also known as AKT)
PKCεprotein kinase C epsilon type
PLIN1perilipin-1
PLIN2perilipin 2
PLIN4perilipin-4
PNPLA2patatin like phospholipase domain containing 2
PNPLA3patatin like phospholipase domain containing 3
PPAP2Aphosphatidic acid phosphatase type 2A
PPARGC1A(or PGC1α) PPARG coactivator 1 alpha
PPARαperoxisome proliferator-activated receptor α
PPARβperoxisome proliferator-activated receptor β
PPARγperoxisome proliferator-activated receptor γ
PPARγ1peroxisome proliferator-activated receptor γ1
PPARγ2peroxisome proliferator-activated receptor γ2
PPARγC1Bperoxisome proliferator-activated receptor gamma, coactivator 1 beta
PPARδperoxisome proliferator-activated receptor δ
PPIApeptidylprolyl isomerase A
p-PKAphosphorylated protein kinase A
p-PKCphosphorylated protein kinase C
PRDM16PR domain containing 16
PREF-1adipocyte differentiation-associated protein
PTGS2prostaglandin-endoperoxide synthase 2
PXRpregnane X receptor
QpEquizalofop-p-ethyl
ROSreactive oxygen species
RUNX2runt-related transcription factor 2
RXRretinoid X receptor
RXRαretinoid X receptor alpha
S100Bcalcium binding protein B
SATsubcutaneous adipose tissue
SCDstearoyl-CoA desaturase
SCD1stearoyl-CoA desaturase 1
SCD1Astearoyl-CoA desaturase 1A
SCD1Bstearoyl-CoA desaturase 1B
SCN9Asodium voltage-gated channel alpha subunit 9
SCOTsuccinyl-CoA-3-oxoacid CoA-transferase
SDHDsuccinate dehydrogenase complex subunit D
SGBSSimpson-Golabi-Behemel syndrome
SIRT1sirtuin 1
SIRT2sirtuin 2
SIRT3sirtuin 3
SIRT5sirtuin 5
SIRT6sirtuin 6
SIRT7sirtuin 7
SMsphingomyelin
SNAP25synaptosome-associated protein 25
SODsuperoxide dismutase
SOD1superoxide dismutase 1
SOD2superoxide dismutase 2
SOX2SRY-box 2
SR-A1scavenger receptor A1
SR-B1scavenger receptor B1
SRCspare respiratory capacity
SREBF1sterol regulatory element binding transcription factor 1
SREBF2sterol regulatory element binding transcription factor 2
SREBP1sterol regulatory element-binding protein 1
SREBP1Csterol regulatory element-binding protein 1c
SREBP2sterol regulatory element-binding protein 2
STAT3signal transducer and activator of transcription 3
STAT5signal transducer and activator of transcription
STAT5Asignal transducer and activator of transcription 5A
STAT5Bsignal transducer and activator of transcription 5B
STSsteroid sulfatase
SULT1A1sulfotransferase family 1A member 1
SULT1A3/4sulfotransferase family 1A member 3
SURsulfonylurea receptor
SUR1sulfonylurea receptor 1
SVFstromal vascular fraction
T2DMtype 2 diabetes mellitus
TBBtetrabromobenzoate
TBBPAtetrabromobisphenol
TBTtributyltin
TCtotal cholesterol
TCBPAtetrachlorobisphenol A
TCDD2,3,7,8-tetrachlorodibenzo-p-dioxin
TCEPtris (2-chloroethyl) phosphate
TCPtricresyl phosphate
TCPAT-complex protein 1 subunit alpha
TCPPtris (2-chloroisopropyl) phosphate
TCStriclosan
TDCPPtris-(2-chloro-1- (chloromethyl) ethyl) phosphate
TERTtelomerase reverse transcriptase
TFtolylfluanid
TFAMtranscription factor A, mitochondrial
TGtriglyceride
THRSPthyroid hormone responsive
TIP47perilipin 3
TMBPFtetramethyl bisphenol F
TNFαtumor necrosis factor-α
ToxPitoxicological Priority Index
TPhPtriphenyl phosphate
TPPtriphenylphosphate
TPTterpyridine platinum(II) chloride
TR/RXRthyroid-receptor/retinoid X receptor
TRIB3tribbles pseudokinase 3
TSPOtranslocator protein
UCP-1uncoupling protein-1
UCP-2uncoupling protein-2
UCP-3uncoupling protein-3
UGT2B15UDP glucuronosyltransferase family 2 member B15
UGTsUDP-glucuronosyltransferases
UQCRBubiquinol-cytochrome C reductase binding protein
VAPAVAMP associated protein A
VATvisceral adipose tissue
VDBPvitamin D-binding protein
VTG1Vitellogenin 1
WATwhite adipose tissue
WHOWorld Health Organization
WTwild-type
XMEsxenobiotic-metabolizing enzymes
ZFAND2Azinc finger AN1-type containing 2A
α2A-ARadrenergic receptor α2A
β-ARβ adrenergic receptor
β-TC-6beta-tumor cell-6

References

  1. Zorena, K.; Jachimowicz-Duda, O.; Ślęzak, D.; Robakowska, M.; Mrugacz, M. Adipokines and Obesity. Potential Link to Metabolic Disorders and Chronic Complications. Int. J. Mol. Sci 2020, 21, 3570. [Google Scholar] [CrossRef]
  2. Overweight and Obesity—BMI Statistics. Available online: https://ec.europa.eu/eurostat/statistics-explained/index.php?title=Overweight_and_obesity_-_BMI_statistics#Education_level_and_overweight (accessed on 20 November 2022).
  3. OECD/EU. Health at a Glance: Europe 2018: State of Health in the EU Cycle; OECD Publishing: Paris, France, 2018; p. 124. [Google Scholar]
  4. Nappi, F.; Barrea, L.; Di Somma, C.; Savanelli, M.C.; Muscogiuri, G.; Orio, F.; Savastano, S. Endocrine Aspects of Environmental “Obesogen” Pollutants. Int. J. Res. Public Health 2016, 13, 765. [Google Scholar] [CrossRef] [Green Version]
  5. Peña-Romero, A.C.; Navas-Carrillo, D.; Marín, F.; Orenes-Piñero, E. The future of nutrition: Nutrigenomics and nutrigenetics in obesity and cardiovascular diseases. Crit. Rev. Food Sci. Nutr. 2018, 58, 3030–3041. [Google Scholar] [CrossRef] [PubMed]
  6. Engin, A.B. MicroRNA and Adipogenesis. Adv. Exp. Med. Biol. 2017, 960, 489–509. [Google Scholar] [CrossRef] [PubMed]
  7. Choi, S.-I.; Kwon, H.-Y.; Han, X.; Men, X.; Choi, Y.-E.; Jang, G.-W.; Park, K.-T.; Han, J.; Lee, O.-H. Environmental obesogens (bisphenols, phthalates and parabens) and their impacts on adipogenic transcription factors in the absence of dexamethasone in 3T3-L1 cells. J. Steroid Biochem. Mol. Biol. 2021, 214, 105994. [Google Scholar] [CrossRef]
  8. Schaffert, A.; Krieg, L.; Weiner, J.; Schlichting, R.; Ueberham, E.; Karkossa, I.; Bauer, M.; Landgraf, K.; Junge, K.M.; Wabitsch, M.; et al. Alternatives for the worse: Molecular insights into adverse effects of bisphenol a and substitutes during human adipocyte differentiation. Environ. Int. 2021, 156, 106730. [Google Scholar] [CrossRef] [PubMed]
  9. Heindel, J.J.; Blumberg, B. Environmental Obesogens: Mechanisms and Controversies. Annu. Rev. Pharmacol. Toxicol. 2019, 6, 89–106. [Google Scholar] [CrossRef]
  10. Kladnicka, I.; Bludovska, M.; Plavinowa, I.; Muller, L.; Mullerova, D. Obesogenes in food. Biomolecules 2022, 12, 680. [Google Scholar] [CrossRef]
  11. Diamanti-Kandarakis, E.; Bourguignon, J.-P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-disrupting chemicals: An Endocrine Society scientific statement. Endocr. Rev. 2009, 30, 293–342. [Google Scholar] [CrossRef]
  12. Kumar, M.; Sarma, D.K.; Shubham, S.; Kumawat, M.; Verma, V.; Prakash, A.; Tiwari, R. Environmental Endocrine-Disrupting Chemical Exposure: Role in Non-Communicable Diseases. Front. Public Health 2020, 8, 553850. [Google Scholar] [CrossRef] [PubMed]
  13. Janesick, A.; Blumberg, B. Obesogens, Stem Cells and the Developmental Programming of Obesity. Int. J. Androl. 2012, 35, 437–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Legler, J.; Zalko, D.; Jourdan, F.; Jacobs, M.; Fromenty, B.; Balaguer, P.; Bourguet, W.; Kos, V.M.; Nadal, A.; Beausoleil, C.; et al. The GOLIATH Project: Towards an Internationally Harmonised Approach for Testing Metabolism Disrupting Compounds. Int. J. Mol. Sci. 2020, 21, 3480. [Google Scholar] [CrossRef] [PubMed]
  15. Song, W.; Lu, H.; Wu, K.; Zhang, Z.; Shuk-Wa Lau, E.; Ge, W. Genetic evidence for estrogenicity of bisphenol A in zebrafish gonadal differentiation and its signalling mechanism. J. Hazard. Mater. 2020, 386, 121886. [Google Scholar] [CrossRef] [PubMed]
  16. Mattiske, D.M.; Pask, A.J. Endocrine disrupting chemicals in the pathogenesis of hypospadias; developmental and toxicological perspectives. Curr. Res. Toxicol. 2021, 2, 179–191. [Google Scholar] [CrossRef]
  17. Thambirajah, A.A.; Wade, M.G.; Verreault, J.; Buisine, N.; Alves, V.A.; Langlois, V.S.; Helbing, C.C. Disruption by stealth—Interference of endocrine disrupting chemicals on hormonal crosstalk with thyroid axis function in humans and other animals. Environ. Res. 2022, 203, 111906. [Google Scholar] [CrossRef]
  18. Gupta, R.; Kumar, P.; Fahmi, N.; Garg, B.; Dutta, S.; Sachar, S.; Matharu, A.S.; Vimaleswaran, K.S. Endocrine disruption and obesity: A current review on environmental obesogens. CRGSC 2020, 3, 100009. [Google Scholar] [CrossRef]
  19. Mohajer, N.; Du, C.Y.; Checkcinco, C.; Blumberg, B. Obesogens: How They Are Identified and Molecular Mechanisms Underlying Their Action. Front. Endocrinol. 2021, 12, 780888. [Google Scholar] [CrossRef]
  20. 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] [Green Version]
  21. Sokal, A.; Jarmakiewicz-Czaja, S.; Tabarkiewicz, J.; Filip, R. Dietary Intake of Endocrine Disrupting Substances Presents in Environment and Their Impact on Thyroid Function. Nutrients 2021, 13, 867. [Google Scholar] [CrossRef]
  22. Naomi, R.; Yazid, M.D.; Bahari, H.; Keong, T.T.; Rajandram, R.; Embomg, H.; Teoh, S.H.; Halim, S.; Othman, F. Bisphenol A (BPA) leading to obesity and cardiovascular complication: A complication of current in vivo study. Int. J. Mol. Sci. 2022, 23, 2969. [Google Scholar] [CrossRef]
  23. Dos Santos, R.S.; Medina-Gali, R.M.; Babiloni-Chust, I.; Marroqui, L.; Nadal, A. In vitro Assays to Identify Metabolism-Disrupting Chemicals with Diabetogenic Activity in a Human Pancreatic β-Cell Model. Int. J. Mol. Sci. 2022, 23, 5040. [Google Scholar] [CrossRef] [PubMed]
  24. Norgren, K.; Tuck, A.; Vieira Silva, A.; Burkhardt, P.; Öberg, M.; Munic Kos, V. High throughput screening of bisphenols and their mixtures under conditions of low-intensity adipogenesis of human mesenchymal stem cells (hMSCs). Food Chem. Toxicol. 2022, 161, 112842. [Google Scholar] [CrossRef]
  25. Hines, E.P.; White, S.S.; Stanko, J.P.; Gibbs-Flournoy, E.A.; Lau, H.; Fenton, S.E. Phenotypic dichotomy following developmental exposure to perfluorooctanoic acid (PFOA) in female CD-1 mice: Low doses induce elevated serum leptin and insulin, and overweight in mid-life. Mol. Cell Endocrinol. 2009, 304, 97–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bokobza, E.; Hinault, C.; Tiroille, V.; Clavel, S.; Bost, F.; Chevalier, N. The adipose tissue at the crosstalk between EDCs and cancer development. Front. Endocrinol. 2021, 12, 691658. [Google Scholar] [CrossRef]
  27. 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]
  28. Haq, M.E.U.; Akash, M.S.H.; Rehman, K.; Mahmood, M.H. Chronic exposure of bisphenol A impairs carbohydrate and lipid metabolism by altering corresponding enzymatic and metabolic pathways. Environ. Toxicol. Pharmacol. 2020, 78, 103387. [Google Scholar] [CrossRef]
  29. Wang, B.; Tsakiridis, E.E.; Zhang, S.; Llanos, A.; Desjardins, E.M.; Yabut, J.M.; Green, A.E.; Day, E.A.; Smith, B.K.; Lally, J.S.V.; et al. The pesticide chlorpyrifos promotes obesity by inhibiting diet-induced thermogenesis in brown adipose tissue. Nat. Commun. 2021, 12, 1. [Google Scholar] [CrossRef]
  30. Zhou, L.; Chen, H.; Xu, Q.; Han, X.; Zhao, Y.; Song, X.; Zhao, T.; Ye, L. The effect of di-2-ethylhexyl phthalate on inflammation and lipid metabolic disorder in rats. Ecotoxicol. Environ. Saf. 2019, 170, 391–398. [Google Scholar] [CrossRef] [PubMed]
  31. Amato, A.A.; Wheeler, H.B.; Blumberg, B. Obesity and endocrine-disrupting chemicals. Endocr. Connect. 2021, 10, R87–R105. [Google Scholar] [CrossRef] [PubMed]
  32. Callaghan, M.A.; Alatorre-Hinojosa, S.; Connors, L.T.; Singh, R.D.; Thompson, J.A. Plasticizers and Cardiovascular Health: Role of Adipose Tissue Dysfunction. Front. Pharmacol. 2021, 11, 626448. [Google Scholar] [CrossRef]
  33. Giuliani, A.; Zuccarini, M.; Cichelli, A.; Khan, H.; Reale, M. Critical Review on the Presence of Phthalates in Food and Evidence of Their Biological Impact. Int. J. Environ. Res. Public Health 2020, 17, 5655. [Google Scholar] [CrossRef] [PubMed]
  34. Almeida, S.; Raposo, A.; Almeida-González, M.; Carrascosa, C. Bisphenol A: Food Exposure and Impact on Human Health. CRFSFS 2018, 17, 1503–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mnif, W.; Hassine, A.I.; Bouaziz, A.; Bartegi, A.; Thomas, O.; Roig, B. Effect of endocrine disruptor pesticides: A review. Int. J. Environ. Res. Public Health 2011, 8, 2265–2303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ren, X.M.; Kuo, Y.; Blumberg, B. Agrochemicals and obesity. Mol. Cell. Endocrinol. 2020, 515, 110926. [Google Scholar] [CrossRef]
  37. Mukherjee, R.; Pandya, P.; Baxi, D.; Ramachandran, A.V. Endocrine Disruptors-‘Food’ for Thought. Proc. Zool. Soc. 2021, 74, 432–442. [Google Scholar] [CrossRef]
  38. Garí, M.; Moos, R.; Bury, D.; Kasper-Sonnenberg, M.; Jankowska, A.; Andysz, A.; Hanke, W.; Nowak, D.; Bose-O’Reilly, S.; Koch, H.M.; et al. Human-biomonitoring derived exposure and daily intakes of Bisphenol A and their associations with neurodevelopmental outcomes among children of the Polish Mother and Child Cohort Study. Environ. Health 2021, 20, 95. [Google Scholar] [CrossRef]
  39. Kassotis, C.D.; Masse, L.; Kim, S.; Schlezinger, J.J.; Webster, T.F.; Stapleton, H.M. Characterization of Adipogenic Chemicals in Three Different Cell Culture Systems: Implications for Reproducibility Based on Cell Source and Handling. Sci. Rep. 2017, 7, 1. [Google Scholar] [CrossRef] [Green Version]
  40. Grün, F.; Blumberg, B. Environmental obesogens: Organotins and endocrine disruption via nuclear receptor signaling. Endocrinology 2006, 147 (Suppl. 6), S50–S55. [Google Scholar] [CrossRef] [Green Version]
  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]
  42. Ernst, J.; Grabiec, U.; Falk, K.; Dehghani, F.; Schaedlich, K. The endocrine disruptor DEHP and the ECS: Analysis of a possible crosstalk. Endocr. Connect. 2020, 9, 101–110. [Google Scholar] [CrossRef]
  43. Lincho, J.; Martins, R.C.; Gomes, J. Paraben Compounds—Part I: An Overview of Their Characteristics, Detection, and Impacts. Appl. Sci. 2021, 1, 2307. [Google Scholar] [CrossRef]
  44. Metcalfe, C.D.; Bayen, S.; Desrosiers, M.; Muñoz, G.; Sauvé, S.; Yargeau, V. An introduction to the sources, fate, occurrence and effects of endocrine disrupting chemicals released into the environment. Environ. Res. 2022, 207, 112658. [Google Scholar] [CrossRef] [PubMed]
  45. Wan, H.T.; Leung, P.Y.; Zhao, Y.G.; Wei, X.; Wong, M.H.; Wong, C.K. Blood plasma concentrations of endocrine disrupting chemicals in Hong Kong populations. J. Hazard. Mater. 2013, 261, 763–769. [Google Scholar] [CrossRef]
  46. Arbuckle, T.E.; Davis, K.; Marro, L.; Fisher, M.; Legrand, M.; LeBlanc, A.; Gaudreau, E.; Foster, W.G.; Choeurng, V.; Fraser, W.D. MIREC Study Group. Phthalate and bisphenol A exposure among pregnant women in Canada—Results from the MIREC study. Environ. Int. 2014, 68, 55–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Ye, X.; Wong, L.Y.; Kramer, J.; Zhou, X.; Jia, T.; Calafat, A.M. Urinary Concentrations of Bisphenol A and Three Other Bisphenols in Convenience Samples of U.S. Adults during 2000–2014. Environ. Sci. Technol. 2015, 49, 11834–11839. [Google Scholar] [CrossRef] [PubMed]
  48. Desalegn, A.A.; Iszatt, N.; Stigum, H.; Jensen, T.K.; Eggesbø, M. A case-cohort study of perinatal exposure to potential endocrine disrupters and the risk of cryptorchidism in the Norwegian HUMIS study. Environ. Int. 2021, 157, 106815. [Google Scholar] [CrossRef]
  49. Cariou, R.; Antignac, J.P.; Zalko, D.; Berrebi, A.; Cravedi, J.P.; Maume, D.; Marchand, P.; Monteau, F.; Riu, A.; Andre, F.; et al. Exposure assessment of French women and their newborns to tetrabromobisphenol-A: Occurrence measurements in maternal adipose tissue, serum, breast milk and cord serum. Chemosphere 2008, 73, 1036–1041. [Google Scholar] [CrossRef]
  50. Mangum, L.H.; Howell, G.E., 3rd; Chambers, J.E. Exposure to p,p’-DDE enhances differentiation of 3T3-L1 preadipocytes in a model of sub-optimal differentiation. Toxicol. Lett. 2015, 238, 65–71. [Google Scholar] [CrossRef]
  51. Garí, M.; Koch, H.M.; Pälmke, C.; Jankowska, A.; Wesołowska, E.; Hanke, W.; Nowak, D.; Bose-O’Reilly, S.; Polańska, K. Determinants of phthalate exposure and risk assessment in children from Poland. Environ. Int. 2019, 127, 742–753. [Google Scholar] [CrossRef]
  52. Ravichandran, G.; Lakshmanan, D.K.; Arunachalam, A.; Thilagar, S. Food obesogens as emerging metabolic disruptors; A toxicological insight. J. Steroid Biochem. Mol. Biol. 2022, 217, 106042. [Google Scholar] [CrossRef]
  53. Lowell Center for Sustainable Production at the University of Massachusetts Lowell. Phthalates and Their Alternatives: Health and Environmental Concerns; University of Massachusetts: Lowell, MA, USA, 2011; pp. 1–24. [Google Scholar]
  54. Wang, Y.; Qian, H. Phthalates and Their Impacts on Human Health. Healthcare 2021, 9, 603. [Google Scholar] [CrossRef] [PubMed]
  55. 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]
  56. Harnett, K.G.; Chin, A.; Schuh, S.M. BPA and BPA alternatives BPS, BPAF, and TMBPF, induce cytotoxicity and apoptosis in rat and human stem cells. Ecotoxicol. Environ. Saf. 2021, 216, 112210. [Google Scholar] [CrossRef] [PubMed]
  57. Vasconcelos, A.L.; Silva, M.J.; Louro, H. In vitro exposure to the next-generation plasticizer diisononyl cyclohexane-1,2-dicarboxylate (DINCH): Cytotoxicity and genotoxicity assessment in human cells. J. Toxicol. Environ. Health A 2019, 82, 526–536. [Google Scholar] [CrossRef]
  58. Andújar, N.; Gálvez-Ontiveros, Y.; Zafra-Gómez, A.; Rodrigo, L.; Álvarez-Cubero, M.J.; Aguilera, M.; Monteagudo, C.; Rivas, A.A. Bisphenol A Analogues in Food and Their Hormonal and Obesogenic Effects: A Review. Nutrients 2019, 11, 2136. [Google Scholar] [CrossRef] [Green Version]
  59. Zughaibi, T.A.; Sheikh, I.A.; Beg, M.A. Insights into the Endocrine Disrupting Activity of Emerging Non-Phthalate Alternate Plasticizers against Thyroid Hormone Receptor: A Structural Perspective. Toxics 2022, 10, 263. [Google Scholar] [CrossRef]
  60. Schaffert, A.; Arnold, J.; Karkossa, I.; Blühe, 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]
  61. Campioli, E.; Lau, M.; Papadopoulos, V. Effect of subacute and prenatal DINCH plasticizer exposure on rat dams and male offspring hepatic function: The role of PPAR-α. Environ. Res. 2019, 179 Pt A, 108773. [Google Scholar] [CrossRef]
  62. 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]
  63. Doke, S.K.; Dhawale, S.C. Alternatives to animal testing: A review. Saudi Pharm. J. 2015, 23, 223–229. [Google Scholar] [CrossRef]
  64. Porro, S.; Genchi, V.A.; Cignarelli, A.; Natalicchio, A.; Laviola, L.; Giorgino, F.; Perrini, S. Dysmetabolic adipose tissue in obesity: Morphological and functional characteristics of adipose stem cells and mature adipocytes in healthy and unhealthy obese subjects. J. Endocrinol. Investig. 2021, 44, 921–941. [Google Scholar] [CrossRef]
  65. Jung, U.J.; Choi, M.S. Obesity and Its Metabolic Complications: The Role of Adipokines and the Relationship between Obesity, Inflammation, Insulin Resistance, Dyslipidemia and Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2014, 15, 6184–6223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Ahmad, B.; Serpell, C.J.; Fong, I.L.; Wong, E.H. Molecular Mechanisms of Adipogenesis: The Anti-adipogenic Role of AMP-Activated Protein Kinase. Front. Mol. Biosci. 2020, 7, 76. [Google Scholar] [CrossRef]
  67. Mentor, A.; Brunström, B.; Mattsson, A.; Jönsson, M. Developmental exposure to a human relevant mixture of endocrine disruptors alters metabolism and adipogenesis in zebrafish (Danio rerio). Chemosphere 2020, 238, 124584. [Google Scholar] [CrossRef] [PubMed]
  68. Longo, M.; Zatterale, F.; Naderi, J.; Parrillo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications. Int. J. Mol. Sci. 2019, 20, 2358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. González-Casanova, J.E.; Pertuz-Cruz, S.L.; Caicedo-Ortega, N.H.; Rojas-Gomez, D.M. Adipogenesis Regulation and Endocrine Disruptors: Emerging Insights in Obesity. Biomed. Res. Int. 2020, 2020, 7453786. [Google Scholar] [CrossRef]
  70. 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]
  71. Baker, A.H.; Watt, J.; Huang, C.K.; Gerstenfeld, L.C.; Schlezinger, J.J. Tributyltin engages multiple nuclear receptor pathways and suppresses osteogenesis in bone marrow multipotent stromal cells. Chem. Res. Toxicol. 2015, 28, 1156–1166. [Google Scholar] [CrossRef] [Green Version]
  72. Ahmed, S.; Atlas, E. Bisphenol S-and bisphenol A-induced adipogenesis of murine preadipocytes occurs through direct peroxisome proliferator-activated receptor gamma activation. Int. J. Obes. 2016, 40, 1566. [Google Scholar] [CrossRef]
  73. Feige, J.N.; Gelman, L.; Rossi, D.; Zoete, V.; Metivier, 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]
  74. Stark, J.M.; Coquet, J.M.; Tibbitt, C.A. The Role of PPAR-γ in Allergic Disease. Curr. Allergy Asthma Rep. 2021, 21, 45. [Google Scholar] [CrossRef]
  75. Boucher, J.G.; Boudreau, A.; Atlas, E. Bisphenol A induces differentiation of human preadipocytes in the absence of glucocorticoid and is inhibited by an estrogen-receptor antagonist. Nutr. Diabetes 2014, 4, e102. [Google Scholar] [CrossRef] [Green Version]
  76. Wang, J.; Sun, B.; Hou, M.; 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] [PubMed]
  77. Pesta, M.; Cedikova, M.; Dvorak, P.; Dvorakova, J.; Kulda, V.; Srbecka, K.; Muller, L.; Bouchalova, V.; Kralickova, M.; Babuska, V.; et al. Trends in gene expression changes during adipogenesis in human adipose derived mesenchymal stem cells under dichlorodiphenyldichloroethylene exposure. Mol. Cell. Toxicol. 2018, 14, 369–379. [Google Scholar] [CrossRef]
  78. Kirchner, S.; Kieu, T.; Chow, C.; Casey, S.; Blumberg, B. Prenatal exposure to the environmental obesogen tributyltin predisposes multipotent stem cells to become adipocytes. Mol. Endocrinol. 2010, 24, 526–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. 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]
  80. Howell, G., 3rd; Mangum, L. Exposure to bioaccumulative organochlorine compounds alters adipogenesis, fatty acid uptake, and adipokine production in NIH3T3-L1 cells. Toxicol. Vitr. 2011, 25, 394–402. [Google Scholar] [CrossRef] [Green Version]
  81. Sakurai, K.; Kawazuma, M.; Adachi, T.; Harigaya, T.; Saito, Y.; Hashimoto, N.; Mori, C. Bisphenol A affects glucose transport in mouse 3T3-F442A adipocytes. Br. J. Pharmacol. 2004, 141, 209–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. 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]
  83. El-Atta, H.M.; Ahmed, E.R. Study of the In-vitro Epigenetic Toxicity Effects of Malaoxon, Malathion Dicarboxylic Acid, Cadmium Chloride and Bisphenol-A on PPAR γ, PPIA and aP2 gene Expressions. J. Clin. Toxicol. 2018, 8, 3. [Google Scholar] [CrossRef]
  84. 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] [Green Version]
  85. Xue, P.; Hou, Y.; Zhang, Q.; Woods, C.G.; Yarborough, K.; Liu, H.; Sun, G.; Andersen, M.E.; Pi, J. Prolonged inorganic arsenite exposure suppresses insulin-stimulated AKT S473 phosphorylation and glucose uptake in 3T3-L1 adipocytes: Involvement of the adaptive antioxidant response. Biochem. Biophys. Res. Commun. 2011, 407, 360–365. [Google Scholar] [CrossRef] [Green Version]
  86. Jin, Y.; Lin, X.; Miao, W.; Wu, T.; Shen, H.; Chen, S.; Li, Y.; Pan, Q.; Fu, Z. Chronic exposure of mice to environmental endocrine-disrupting chemicals disturbs their energy metabolism. Toxicol. Lett. 2014, 225, 392–400. [Google Scholar] [CrossRef]
  87. Al-Suhaimi, E.A.; Shehzad, A. Leptin, resistin and visfatin: The missing link between endocrine metabolic disorders and immunity. Eur. J. Med. Res. 2013, 18, 12. [Google Scholar] [CrossRef] [Green Version]
  88. Graudejus, O.; Ponce Wong, R.D.; Varghese, N.; Wagner, S.; Morrison, B. Bridging the gap between in vivo and in vitro research: Reproducing in vitro the mechanical and electrical environment of cells in vivo. In Proceedings of the MEA Meeting 2018|11th International Meeting on Substrate Integrated Microelectrode Arrays, Reutlingen, Germany, 4–6 July 2018. [Google Scholar] [CrossRef]
  89. Griffin, M.; Pereira, S.R.; DeBari, M.K.; Abbott, R.D. Mechanisms of action, chemical characteristics, and model systems of obesogens. BMC Biomed. Eng. 2020, 2, 6. [Google Scholar] [CrossRef] [PubMed]
  90. Forgacs, A.L.; Dere, E.; Angrish, M.M.; Zacharewski, T.R. Comparative analysis of temporal and dose-dependent TCDD-elicited gene expression in human, mouse, and rat primary hepatocytes. Toxicol. Sci. 2013, 133, 54–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Janesick, A.S.; Blumberg, B. Obesogens: An emerging threat to public health. Am. J. Obstet. Gynecol. 2016, 214, 559–565. [Google Scholar] [CrossRef] [Green Version]
  92. Morrison, S.; McGee, S.L. 3T3-L1 adipocytes display phenotypic characteristics of multiple adipocyte lineages. Adipocyte 2015, 4, 295–302. [Google Scholar] [CrossRef] [Green Version]
  93. Sun, Z.; Cao, H.; Liu, Q.S.; Liang, Y.; Fiedler, H.; Zhang, J.; Zhou, Q.; Jiang, G. 4-Hexylphenol influences adipogenic differentiation and hepatic lipid accumulation in vitro. Environ. Pollut. 2021, 268, 115635. [Google Scholar] [CrossRef] [PubMed]
  94. Ruiz-Ojeda, F.J.; Rupérez, A.I.; Gomez-Llorente, C.; Gil, A.; Aguilera, C.M. Cell Models and Their Application for Studying Adipogenic Differentiation in Relation to Obesity: A Review. Int. J. Mol. Sci. 2016, 17, 1040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Vernochet, C.; Peres, S.B.; Davis, K.E.; McDonald, M.E.; Qiang, L.; Wang, H.; Scherer, P.E.; Farmer, S.R. C/EBPalpha and the corepressors CtBP1 and CtBP2 regulate repression of select visceral white adipose genes during induction of the brown phenotype in white adipocytes by peroxisome proliferator-activated receptor gamma agonists. Mol. Cell. Biol. 2009, 29, 4714–4728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. 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] [Green Version]
  97. 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] [PubMed] [Green Version]
  98. 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]
  99. Biserni, M.; Mesnage, R.; Ferro, R.; Wozniak, E.; Xenakis, T.; Mein, C.A.; Antoniou, M.N. Quizalofop-p-Ethyl Induces Adipogenesis in 3T3-L1 Adipocytes. Toxicol. Sci. 2019, 170, 452–461. [Google Scholar] [CrossRef] [Green Version]
  100. Kim, J.; Sun, Q.; Yue, Y.; Yoon, K.S.; Whang, K.Y.; Marshall Clark, J.; Park, Y. 4,4′-Dichlorodiphenyltrichloroethane (DDT) and 4,4′-dichlorodiphenyldichloroethylene (DDE) promote adipogenesis in 3T3-L1 adipocyte cell culture. Pestic. Biochem. Physiol. 2016, 131, 40–45. [Google Scholar] [CrossRef] [Green Version]
  101. Smith, A.; Yu, X.; Yin, L. Diazinon exposure activated transcriptional factors CCAAT-enhancer-binding proteins α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ) and induced adipogenesis in 3T3-L1 preadipocytes. Pestic. Biochem. Physiol. 2018, 150, 48–58. [Google Scholar] [CrossRef]
  102. Blanco, J.; Guardia-Escote, L.; Mulero, M.; Basaure, P.; Biosca-Brull, J.; Cabré, M.; Colomina, M.T.; Domingo, J.L.; Sánchez, D.J. Obesogenic effects of chlorpyrifos and its metabolites during the differentiation of 3T3-L1 preadipocytes. Food Chem. Toxicol. 2020, 137, 111171. [Google Scholar] [CrossRef]
  103. Regnier, S.M.; El-Hashani, E.; Kamau, W.; Zhang, X.; Massad, N.L.; Sargis, R.M. Tributyltin differentially promotes development of a phenotypically distinct adipocyte. Obesity 2015, 23, 1864–1871. [Google Scholar] [CrossRef]
  104. Janesick, A.S.; Dimastrogiovanni, G.; Vanek, L.; Boulos, C.; Chamorro-García, R.; Tang, W.; Blumberg, B. On the Utility of ToxCast™ and ToxPi as Methods for Identifying New Obesogens. Environ. Health Perspect. 2016, 124, 1214–1226. [Google Scholar] [CrossRef] [Green Version]
  105. Carchia, E.; Porreca, I.; Almeida, P.J.; D’Angelo, F.; Cuomo, D.; Ceccarelli, M.; De Felice, M.; Mallardo, M.; Ambrosino, C. Evaluation of low doses BPA-induced perturbation of glycemia by toxicogenomics points to a primary role of pancreatic islets and to the mechanism of toxicity. Cell Death Dis. 2015, 6, e1959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chen, Y.-W.; Lan, K.-C.; Tsai, J.-R.; Weng, T.-I.; Yang, C.-Y.; Liu, S.-H. Tributyltin exposure at noncytotoxic doses dysregulates pancreatic β-cell function in vitro and in vivo. Arch. Toxicol. 2017, 91, 3135–3144. [Google Scholar] [CrossRef]
  107. Pavlikova, N.; Sramek, J.; Jelinek, M.; Halada, P.; Kovar, J. Markers of acute toxicity of DDT exposure in pancreatic beta-cells determined by a proteomic approach. PLoS ONE 2020, 15, e0229430. [Google Scholar] [CrossRef]
  108. NIH3T3. NIH3T3 General Information. Available online: https://www.nih3t3.com/ (accessed on 20 November 2022).
  109. 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]
  110. Klein, J.; Fasshauer, M.; Klein, H.H.; Benito, M.; Kahn, C.R. Novel adipocyte lines from brown fat: A model system for the study of differentiation, energy metabolism, and insulin action. Bioessays 2002, 24, 382–388. [Google Scholar] [CrossRef] [PubMed]
  111. Moreno-Aliaga, M.J.; Matsumura, F. Effects of 1,1,1-trichloro-2,2-bis(p-chlorophenyl)-ethane (p,p′-DDT) on 3T3-L1 and 3T3-F442A adipocyte differentiation. Biochem. Pharmacol. 2002, 63, 997–1007. [Google Scholar] [CrossRef] [PubMed]
  112. Rylander, L.; Nilsson-Ehle, P.; Hagmar, L. A simplified precise method for adjusting serum levels of persistent organohalogen pollutants to total serum lipids. Chemosphere 2006, 62, 333–336. [Google Scholar] [CrossRef] [Green Version]
  113. Azzouz, A.; Hausler, R.; El-Akhrass, M. Pesticides and removal approaches. In Sorbents Materials for Controlling Environmental Pollution, Current State and Trends; Núñez-Delgado, A., Ed.; Elsevier: Amsterdam, The Netherlands, 2021; pp. 435–462. [Google Scholar]
  114. DDT—A Brief History and Status. Available online: https://www.epa.gov/ingredients-used-pesticide-products/ddt-brief-history-and-status (accessed on 21 November 2022).
  115. Tawar, N.; Banerjee, B.D.; Mishra, B.K.; Sharma, T.; Tyagi, S.; Madhu, S.V.; Agarwal, V.; Gupta, S. Adipose Tissue Levels of DDT as Risk Factor for Obesity and Type 2 Diabetes Mellitus. Indian J. Endocrinol. Metab. 2021, 25, 160–165. [Google Scholar] [CrossRef]
  116. La Merrill, M.A.; Johnson, C.L.; Smith, M.T.; Kandula, N.R.; Macherone, A.; Pennell, K.D.; Kanaya, A.M. Exposure to Persistent Organic Pollutants (POPs) and Their Relationship to Hepatic Fat and Insulin Insensitivity among Asian Indian Immigrants in the United States. Environ. Sci. Technol. 2019, 53, 13906–13918. [Google Scholar] [CrossRef]
  117. Tyagi, S.; Mishra, B.K.; Sharma, T.; Tawar, N.; Urfi, A.J.; Banerjee, B.D.; Madhu, S.V. Level of Organochlorine Pesticide in Prediabetic and Newly Diagnosed Diabetes Mellitus Patients with Varying Degree of Glucose Intolerance and Insulin Resistance among North Indian Population. Diabetes Metab. J. 2021, 45, 558–568. [Google Scholar] [CrossRef]
  118. Bahmad, H.F.; Daouk, R.; Azar, J.; Sapudom, J.; Teo, J.C.M.; Abou-Kheir, W.; Al-Sayegh, M. Modeling Adipogenesis: Current and Future Perspective. Cells 2020, 9, 2326. [Google Scholar] [CrossRef]
  119. Lane, J.M.; Doyle, J.R.; Fortin, J.P.; Kopin, A.S.; Ordovás, J.M. Development of an OP9 derived cell line as a robust model to rapidly study adipocyte differentiation. PLoS ONE 2014, 9, e112123. [Google Scholar] [CrossRef] [Green Version]
  120. Andrews, F.V.; Kim, S.M.; Edwards, L.; Schlezinger, J.J. Identifying adipogenic chemicals: Disparate effects in 3T3-L1, OP9 and primary mesenchymal multipotent cell models. Toxicol. Vitr. 2020, 67, 104904. [Google Scholar] [CrossRef]
  121. Yajima, Y.; Sato, M.; Sumida, M.; Kawashima, S. Mechanism of adult primitive mesenchymal ST-13 preadipocyte differentiation. Endocrinology 2003, 144, 2559–2565. [Google Scholar] [CrossRef] [Green Version]
  122. Wabitsch, M.; Brenner, R.E.; Melzner, I.; Braun, M.; Möller, P.; Heinze, E.; Debatin, K.M.; Hauner, H. Characterization of a human preadipocyte cell strain with high capacity for adipose differentiation. Int. J. Obes. Relat. Metab. Disord. 2001, 25, 8–15. [Google Scholar] [CrossRef] [Green Version]
  123. Fischer-Posovszky, P.; Newell, F.S.; Wabitsch, M.; Tornqvist, H.E. Human SGBS cells—A unique tool for studies of human fat cell biology. Obes. Facts 2008, 1, 184–189. [Google Scholar] [CrossRef]
  124. 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] [Green Version]
  125. Primary Subcutaneous Pre-Adipocytes; Normal, Human PCS-210-010™. Available online: https://www.atcc.org/products/pcs-210-010 (accessed on 20 November 2022).
  126. Yeo, C.R.; Agrawal, M.; Hoon, S.; Shabbir, A.; Shrivastava, M.K.; Huang, S.; Khoo, C.M.; Chhay, V.; Yassin, M.S.; Tai, E.S.; et al. SGBS cells as a model of human adipocyte browning: A comprehensive comparative study with primary human white subcutaneous adipocytes. Sci. Rep. 2017, 7, 1. [Google Scholar] [CrossRef] [Green Version]
  127. Wassef, H.; Bernier, L.; Davignon, J.; Cohn, J.S. Synthesis and secretion of apoC-I and apoE during maturation of human SW872 liposarcoma cells. J. Nutr. 2004, 134, 2935–2941. [Google Scholar] [CrossRef]
  128. Carmel, J.F.; Tarnus, E.; Cohn, J.S.; Bourdon, E.; Davignon, J.; Bernier, L. High expression of apolipoprotein E impairs lipid storage and promotes cell proliferation in human adipocytes. J. Cell. Biochem. 2009, 106, 608–617. [Google Scholar] [CrossRef] [PubMed]
  129. 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] [Green Version]
  130. Hu, P.; Overby, H.; Heal, E.; Wang, S.; Chen, J.; Shen, C.-L.; Zhao, L. Methylparaben and butylparaben alter multipotent mesenchymal stem cell fates towards adipocyte lineage. Toxicol. Appl. Pharmacol. 2017, 329, 48–57. [Google Scholar] [CrossRef]
  131. Bateman, M.E.; Strong, A.L.; McLachlan, J.A.; Burow, M.E.; Bunnell, B.A. The Effects of Endocrine Disruptors on Adipogenesis and Osteogenesis in Mesenchymal Stem Cells: A Review. Front. Endocrinol. 2017, 7, 171. [Google Scholar] [CrossRef] [Green Version]
  132. Casteilla, L.; Planat-Benard, V.; Laharrague, P.; Cousin, B. Adipose-derived stromal cells: Their identity and uses in clinical trials, an update. World J. Stem Cells 2011, 3, 25–33. [Google Scholar] [CrossRef]
  133. Zhao, L.; Li, G.; Chan, K.-M.; Wang, Y.; Tang, P.-F. Comparison of multipotent differentiation potentials of murine primary bone marrow stromal cells and mesenchymal stem cell line C3H10T1/2. Calcif. Tissue Int. 2009, 84, 56–64. [Google Scholar] [CrossRef]
  134. Reznikoff, C.A.; Bertram, J.S.; Brankow, D.W.; Heidelberger, C. Quantitative and qualitative studies of chemical transformation of cloned C3H mouse embryo cells sensitive to postconfluence inhibition of cell division. Cancer Res. 1973, 33, 3239–3249. [Google Scholar]
  135. Lee, N.; Kim, I.; Park, S.; Han, D.; Ha, S.; Kwon, M. Creatine inhibits adipogenesis by downregulating insulin-induced activation of the phosphatidylinositol 3-kinase signaling pathway. Stem Cells Dev. 2015, 24, 983–994. [Google Scholar] [CrossRef]
  136. Beg, M.; Chauhan, P.; Varshney, S.; Shankar, K.; Rajan, S.; Saini, D. A withanolide coagulin-L inhibits adipogenesis modulating Wnt/β-catenin pathway and cell cycle in mitotic clonal expansion. Phytomedicine 2014, 21, 406–414. [Google Scholar] [CrossRef]
  137. Zhang, J.; Choudhury, M. Benzyl Butyl Phthalate Induced Early lncRNA H19 Regulation in C3H10T1/2 Stem Cell Line. Chem. Res. Toxicol. 2021, 34, 54–62. [Google Scholar] [CrossRef]
  138. Zhang, J.; Choudhury, M. The plasticizer BBP selectively inhibits epigenetic regulator sirtuin during differentiation of C3H10T1/2 stem cell line. Toxicol. Vitr. 2017, 39, 75–83. [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] [PubMed]
  140. Biemann, R.; Fischer, B.; Navarrete Santos, A. Adipogenic effects of a combination of the endocrine-disrupting compounds bisphenol A, diethylhexylphthalate, and tributyltin. Obes. Facts 2014, 7, 48–56. [Google Scholar] [CrossRef]
  141. Bukowska, J.; Szóstek-Mioduchowska, A.Z.; Kopcewicz, M.; Walendzik, K.; Machcińska, S.; Gawrońska-Kozak, B. Adipose-Derived Stromal/Stem Cells from Large Animal Models: From Basic to Applied Science. SCRR 2021, 17, 719–738. [Google Scholar] [CrossRef] [PubMed]
  142. Gigante, P.; Berni, M.; Bussolati, S.; Grasselli, F.; Grolli, S.; Ramoni, R.; Basini, G. Glyphosate affects swine ovarian and adipose stromal cell functions. Anim. Reprod. Sci. 2018, 195, 185–196. [Google Scholar] [CrossRef] [PubMed]
  143. Berni, M.; Gigante, P.; Bussolati, S.; Grasselli, F.; Grolli, S.; Ramoni, R.; Basini, G. Bisphenol S, a Bisphenol A alternative, impairs swine ovarian and adipose cell functions. Domest. Anim. Endocrinol. 2019, 66, 48–56. [Google Scholar] [CrossRef]
  144. Dubois, S.G.; Floyd, E.Z.; Zvonic, S.; Kilroy, G.; Wu, X.; Carling, S.; Halvorsen, Y.D.; Ravussin, E.; Gimble, J.M. Isolation of human adipose-derived stem cells from biopsies and liposuction specimens. Methods Mol. Biol. 2008, 449, 69–79. [Google Scholar] [CrossRef]
  145. Human Adipose Derived Stem Cells (ADSCs, Type 1 Diabetes). Available online: https://www.ixcellsbiotech.com/human-primary-cells/human-adipose-derived-stem-cells-adscs-type-1-diabetes (accessed on 20 November 2022).
  146. Ejaz, A.; Hatzmann, F.M.; Hammerle, S.; Ritthammer, H.; Mattesich, M.; Zwierzina, M.; Waldegger, P.; Zwerschke, W. Fibroblast feeder layer supports adipogenic differentiation of human adipose stromal/progenitor cells. Adipocyte 2019, 8, 178–189. [Google Scholar] [CrossRef] [Green Version]
  147. 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] [Green Version]
  148. 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]
  149. Markussen, L.K.; Isidor, M.S.; Breining, P.; Andersen, E.S.; Rasmussen, N.E.; Petersen, L.I.; Pedersen, S.B.; Richelsen, B.; Hansen, J.B. Characterization of immortalized human brown and white pre-adipocyte cell models from a single donor. PLoS ONE 2017, 12, e0185624. [Google Scholar] [CrossRef]
  150. Wabitsch, M.; Brüderlein, S.; Melzner, I.; Braun, M.; Mechtersheimer, G.; Möller, P. LiSa-2, a novel human liposarcoma cell line with a high capacity for terminal adipose differentiation. Int. J. Cancer 2000, 88, 889–894. [Google Scholar] [CrossRef] [PubMed]
  151. Hugo, E.R.; Brandebourg, T.D.; Comstock, C.E.; Gersin, K.S.; Sussman, J.J.; Ben-Jonathan, N. LS14: A novel human adipocyte cell line that produces prolactin. Endocrinology 2006, 147, 306–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Darimont, C.; Zbinden, I.; Avanti, O.; Leone-Vautravers, P.; Giusti, V.; Burckhardt, P.; Pfeifer, A.M.; Macé, K. Reconstitution of telomerase activity combined with HPV-E7 expression allow human preadipocytes to preserve their differentiation capacity after immortalization. Cell Death Differ. 2003, 10, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
  153. Zilberfarb, V.; Piétri-Rouxel, F.; Jockers, R.; Krief, S.; Delouis, C.; Issad, T.; Strosberg, A.D. Human immortalized brown adipocytes express functional beta3-adrenoceptor coupled to lipolysis. J. Cell Sci. 1997, 110 Pt 7, 801–807. [Google Scholar] [CrossRef]
  154. Dufau, J.; Shen, J.X.; Couchet, M.; De Castro Barbosa, T.; Mejhert, N.; Massier, L.; Griseti, E.; Mouisel, E.; Amri, E.Z.; Lauschke, V.M.; et al. In vitro and ex vivo models of adipocytes. Am. J. Physiol. Cell Physiol. 2021, 320, C822–C841. [Google Scholar] [CrossRef]
  155. Alam, M.T.; Ott, S.; Kumar, S.; Saravanan, P. Low vitamin b12 in pregnancy is associated with adipose-derived circulating miRs targeting PPARgamma and insulin resistance. J. Clin. Endocrinol. Metab. 2017, 102, 4200–4209. [Google Scholar] [CrossRef] [Green Version]
  156. Jackisch, L.; Murphy, A.M.; Kumar, S.; Randeva, H.; Tripathi, G.; McTernan, P.G. Tunicamycin-Induced Endoplasmic Reticulum Stress Mediates Mitochondrial Dysfunction in Human Adipocytes. J. Clin. Endocrinol. Metab. 2020, 105, dgaa258. [Google Scholar] [CrossRef]
  157. Pan, S.; Cui, Y.; Fu, Z.; Zhang, L.; Xing, H. MicroRNA-128 is involved in dexamethasone-induced lipid accumulation via repressing SIRT1 expression in cultured pig preadipocytes. J. Steroid Biochem. Mol. Biol. 2018, 186, 185–195. [Google Scholar] [CrossRef]
  158. Riedel, J.; Badewien-Rentzsch, B.; Kohn, B.; Hoeke, L.; Einspanier, R. Characterization of key genes of the renin–angiotensin system in mature feline adipocytes and during in vitro adipogenesis. J. Anim. Physiol. Anim. Nutr. 2016, 100, 1139–1148. [Google Scholar] [CrossRef]
  159. Pu, Y.; Veiga-Lopez, A. PPARγ agonist through the terminal differentiation phase is essential for adipogenic differentiation of fetal ovine preadipocytes. Cell. Mol. Biol. Lett. 2017, 22, 1. [Google Scholar] [CrossRef]
  160. Jetter, A.; Kullak-Ublick, G.A. Drugs and hepatic transporters: A review. Pharmacol. Res. 2020, 154, 104234. [Google Scholar] [CrossRef] [PubMed]
  161. Cano, R.; Pérez, J.L.; Dávila, L.A.; Dávila, L.A.; Ortega, Á.; Gómez, Y.; Valero-Cedeño, N.J.; Parra, H.; Manzano, A.; Véliz Castro, T.I.; et al. Role of Endocrine-Disrupting Chemicals in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 4807. [Google Scholar] [CrossRef] [PubMed]
  162. Shi, H.; Jan, J.; Hardesty, J.E.; Falkner, K.C.; Prough, R.A.; Balamurugan, A.N.; Mokshagundam, S.P.; Chari, S.T.; Cave, M.C. Polychlorinated biphenyl exposures differentially regulate hepatic metabolism and pancreatic function: Implications for nonalcoholic steatohepatitis and diabetes. Toxicol. Appl. Pharmacol. 2019, 363, 22–33. [Google Scholar] [CrossRef] [PubMed]
  163. Neel, B.A.; Brady, M.J.; Sargis, R.M. The Endocrine Disrupting Chemical Tolylfluanid Alters Adipocyte Metabolism via Glucocorticoid Receptor Activation. Mol. Endocrinol. 2013, 27, 394–406. [Google Scholar] [CrossRef] [Green Version]
  164. 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]
  165. Yang, L.; Guo, X.; Mao, X.; Jia, X.; Zhou, Y.; Hu, Y.; Sun, L.; Guo, J.; Xiao, H.; Zhang, Z. Hepatic toxicity of fluorene-9-bisphenol (BHPF) on CD-1 mice. Ecotoxicol. Environ. Saf. 2021, 219, 112298. [Google Scholar] [CrossRef]
  166. Eweda, S.M.; Newairy, A.S.A.; Abdou, H.M.; Gaber, A.S. Bisphenol A-induced oxidative damage in the hepatic and cardiac tissues of rats: The modulatory role of sesame lignans. Exp. Ther. Med. 2020, 19, 33–44. [Google Scholar] [CrossRef] [Green Version]
  167. Meng, Z.; Wang, D.; Liu, W.; Li, R.; Yan, S.; Jia, M.; Zhang, L.; Zhou, Z.; Zhu, W. Perinatal exposure to Bisphenol S (BPS) promotes obesity development by interfering with lipid and glucose metabolism in male mouse offspring. Environ. Res. 2019, 173, 189–198. [Google Scholar] [CrossRef]
  168. Sun, Y.; Wang, X.; Zhou, Y.; Zhang, J.; Cui, W.; Wang, E.; Du, J.; Wei, B.; Xu, X. Protective effect of metformin on BPA-induced liver toxicity in rats through upregulation of cystathionine β synthase and cystathionine γ lyase expression. Sci. Total Environ. 2021, 750, 141685. [Google Scholar] [CrossRef]
  169. Cocci, P.; Mosconi, G.; Arukwe, A.; Mozzicafreddo, M.; Angeletti, M.; Aretusi, G.; Palermo, F.A. Effects of Diisodecyl Phthalate on PPAR:RXR-Dependent Gene Expression Pathways in Sea Bream Hepatocytes. Chem. Res. Toxicol. 2015, 28, 935–947. [Google Scholar] [CrossRef]
  170. Olsvik, P.A.; Søfteland, L. Metabolic effects of p,p′-DDE on Atlantic salmon hepatocytes. J. Appl. Toxicol. 2018, 38, 489–503. [Google Scholar] [CrossRef]
  171. 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] [PubMed]
  172. Boess, F.; Kamber, M.; Romer, S.; Gasser, R.; Muller, D.; Albertini, S.; Suter, L. Gene expression in two hepatic cell lines, cultured primary hepatocytes, and liver slices compared to the in vivo liver gene expression in rats: Possible implications for toxicogenomics use of in vitro systems. Toxicol. Sci. 2003, 73, 386–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. 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]
  174. Sefried, S.; Häring, H.U.; Weigert, C.; Eckstein, S.S. Suitability of hepatocyte cell lines HepG2, AML12 and THLE-2 for investigation of insulin signalling and hepatokine gene expression. Open Biol. 2018, 8, 180147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Wu, H.; Yu, W.; Meng, F.; Mi, J.; Peng, J.; Liu, J.; Zhang, X.; Hai, C.; Wang, X. Polychlorinated biphenyls-153 induces metabolic dysfunction through activation of ROS/NF-κB signaling via downregulation of HNF1b. Redox Biol. 2017, 12, 300–310. [Google Scholar] [CrossRef]
  176. Le, Y.; Shen, H.; Yang, Z.; Lu, D.; Wang, C. Comprehensive analysis of organophosphorus flame retardant-induced mitochondrial abnormalities: Potential role in lipid accumulation. Environ. Pollut. 2021, 274, 116541. [Google Scholar] [CrossRef]
  177. Hepa 1-6: A Murine Model of Hepatocellular Carcinoma. Available online: https://drugdevelopment.labcorp.com/industry-solutions/oncology/preclinical/tumor-spotlights/hepa-1-6-a-murine-model-of-hepatocellular-carcinoma.html (accessed on 20 November 2022).
  178. Ke, Z.-H.; Pan, J.-X.; Jin, L.-Y.; Xu, H.-Y.; Yu, T.-T.; Ullah, K.; Rahman, T.U.; Ren, J.; Cheng, Y.; Dong, X.-Y.; et al. Bisphenol A Exposure May Induce Hepatic Lipid Accumulation via Reprogramming the DNA Methylation Patterns of Genes Involved in Lipid Metabolism. Sci. Rep. 2016, 6, 31331. [Google Scholar] [CrossRef] [Green Version]
  179. Breslow, J.L.; Sloan, H.R.; Ferrans, V.J.; Anderson, J.L.; Levy, R.I. Characterization of the mouse liver cell line FL83B. Exp. Cell Res. 1973, 78, 441–453. [Google Scholar] [CrossRef]
  180. Liu, X.H.; Pan, J.P.; Bauman, W.A.; Cardozo, C.P. AdipoRon prevents myostatin-induced upregulation of fatty acid synthesis and downregulation of insulin activity in a mouse hepatocyte line. Physiol. Rep. 2019, 7, e14152. [Google Scholar] [CrossRef]
  181. ATCC. FL83B CRL-2390TM. Available online: https://www.atcc.org/products/crl-2390 (accessed on 20 November 2022).
  182. Chang, Y.-H.; Chen, Y.-L.; Huang, W.-C.; Liou, C.-J. Fucoxanthin attenuates fatty acid-induced lipid accumulation in FL83B hepatocytes through regulated Sirt1/AMPK signaling pathway. Biochem. Biophys. Res. Commun. 2018, 495, 197–203. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, G.W.; Chen, T.Y.; Yang, P.M. Differential effect of herbal tea extracts on free fatty acids-, ethanol- and acetaminophen-induced hepatotoxicity in FL83B hepatocytes. Drug Chem. Toxicol. 2022, 45, 347–352. [Google Scholar] [CrossRef] [PubMed]
  184. Wang, B.; Ji, K.; Wang, Y.; Li, Y.; Tang, Y.; Gu, J.; Cai, L. Exposure to low dose cadmium enhances FL83B cells proliferation through down-regulation of caspase-8 by DNA hypermethylation. Toxicol. Res. 2015, 4, 248–259. [Google Scholar] [CrossRef]
  185. Lo, D.; Wang, Y.T.; Wu, M.C. Hepatoprotective effect of silymarin on di(2-ethylhexyl)phthalate (DEHP) induced injury in liver FL83B cells. Environ. Toxicol. Pharmacol. 2014, 38, 112–118. [Google Scholar] [CrossRef]
  186. 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]
  187. Malhão, F.; Urbatzka, R.; Navas, J.M.; Cruzeiro, C.; Monteiro, R.A.; Rocha, E. Cytological, immunocytochemical, ultrastructural and growth characterization of the rainbow trout liver cell line RTL-W1. Tissue Cell 2013, 45, 159–174. [Google Scholar] [CrossRef] [PubMed]
  188. Marqueño, A.; Flores, C.; Casado, M.; Porte, C. Dysregulation of lipid metabolism in PLHC-1 and ZFL cells exposed to tributyltin an all-trans retinoic acid. Aquat. Toxicol. 2021, 231, 105733. [Google Scholar] [CrossRef]
  189. Fernandes, D.; Pujol, S.; Pérez-Albaladejo, E.; Tauler, R.; Bebianno, M.J.; Porte, C. Characterization of the environmental quality of sediments from two estuarine systems based on different in-vitro bioassays. Mar. Environ. Res. 2014, 96, 127–135. [Google Scholar] [CrossRef]
  190. 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]
  191. 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]
  192. Gomez-Lechon, M.; Donato, M.; Castell, J.; Jover, R. Human Hepatocytes as a Tool for Studying Toxicity and Drug Metabolism. Curr. Drug Metab. 2003, 4, 292–312. [Google Scholar] [CrossRef]
  193. Clayton, R.F.; Rinaldi, A.; Kandyba, E.E.; Edward, M.; Willberg, C.; Klenerman, P.; Patel, A.H. Liver cell lines for the study of hepatocyte functions and immunological response. Liver Int. 2005, 25, 389–402. [Google Scholar] [CrossRef]
  194. Tolosa, L.; Gómez-Lechón, M.J.; López, S.; Guzmán, C.; Castell, J.V.; Donato, M.T.; Jover, R. Human Upcyte Hepatocytes: Characterization of the Hepatic Phenotype and Evaluation for Acute and Long-Term Hepatotoxicity Routine Testing. Toxicol. Sci. 2016, 152, 214–229. [Google Scholar] [CrossRef] [Green Version]
  195. Baquerizo, A.; Bañares, R.; Saliba, F. Current Clinical Status of the Extracorporeal Liver Support Devices. Transplant. Liver 2015, 107, 1463–1487. [Google Scholar] [CrossRef]
  196. HepG2 Cell Line. Available online: https://encyclopedia.pub/entry/17273 (accessed on 20 November 2022).
  197. Kammerer, S.; Küpper, J.-H. Human hepatocyte systems for in vitro toxicology analysis. J. Cell. Biotechnol. 2018, 3, 85–93. [Google Scholar] [CrossRef] [Green Version]
  198. Arzumanian, V.A.; Kiseleva, O.I.; Poverennaya, E.V. The Curious Case of the HepG2 Cell Line: 40 Years of Expertise. Int. J. Mol. Sci. 2021, 22, 13135. [Google Scholar] [CrossRef] [PubMed]
  199. Lu, J.; Fang, B.; Zheng, Y.; Yu, X.; Huang, G.; Wang, Z.; Deng, X.; Guan, S. 1,3-dichloro-2-propanol induced lipid accumulation in HepG2 cells through cAMP/protein kinase A and AMP-activated protein kinase pathways via Gi/o-coupled receptors. Environ. Toxicol. Pharmacol. 2017, 55, 118–126. [Google Scholar] [CrossRef] [PubMed]
  200. Maia, M.L.; Sousa, S.; Pestana, D.; Faria, A.; Teixeira, D.; Delerue-Matos, C.; Domingues, V.F.; Calhau, C. Impact of brominated flame retardants on lipid metabolism: An in vitro approach. Environ. Pollut. 2022, 294, 118639. [Google Scholar] [CrossRef]
  201. ThermoFisher Scientific. HepaRG™ Cells, Cryopreserved. Available online: https://www.thermofisher.com/order/catalog/product/HPRGC10 (accessed on 20 November 2022).
  202. Stossi, F.; Dandekar, R.D.; Johnson, H.; Lavere, P.; Foulds, C.E.; Mancini, M.G.; Mancini, M.A. Tributyltin chloride (TBT) induces RXRA down-regulation and lipid accumulation in human liver cells. PLoS ONE 2019, 14, e0224405. [Google Scholar] [CrossRef] [PubMed]
  203. Differentiated HepaRG Cells—HPR116. Available online: https://www.heparg.com/rubrique-differentiated-heparg-cells-hpr116 (accessed on 20 November 2022).
  204. Huh-7 Cell Line Origins and Characteristics. Available online: https://huh7.com/ (accessed on 20 November 2022).
  205. Wada, K.; Sakamoto, H.; Nishikawa, K.; Sakuma, S.; Nakajima, A.; Fujimoto, Y.; Kamisaki, Y. Life style-related diseases of the digestive system: Endocrine disruptors stimulate lipid accumulation in target cells related to metabolic syndrome. J. Pharmacol. Sci. 2007, 105, 133–137. [Google Scholar] [CrossRef]
  206. 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]
  207. Lorenzetti, S.; Marcoccia, D.; Mantovani, A. Biomarkers of effect in endocrine disruption: How to link a functional assay to an adverse outcome pathway. Ann. Ist. Super. Sanita 2015, 51, 167–171. [Google Scholar] [CrossRef]
  208. La Rocca, C.; Tait, S.; Mantovani, A. Use of a combinedin vitroassay for effect-directed assessment of infant formulas. Int. J. Food Sci. 2014, 50, 77–83. [Google Scholar] [CrossRef]
  209. Štampar, M.; Breznik, B.; Filipič, M.; Žegura, B. Characterization of In vitro 3D Cell Model Developed from Human Hepatocellular Carcinoma (HepG2) Cell Line. Cells 2020, 9, 2557. [Google Scholar] [CrossRef] [PubMed]
  210. 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] [Green Version]
  211. Mardonna, F.; Carnevali, O. Lipid Metabolism Alteration by Endocrine Disruptors in Animal Models: An Overview. Front. Endocrinol. 2018, 9, 654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Liang, X.; Xu, G.; Gao, Q.; Tao, X. LKB1 expression reverses the tumorigenicity of L02 cells. Oncol. Rep. 2016, 36, 1055–1061. [Google Scholar] [CrossRef] [Green Version]
  213. Zhang, H.; Shao, X.; Zhao, H.; Li, X.; Wei, J.; Yang, C.; Cai, Z. Integration of Metabolomics and Lipidomics Reveals Metabolic Mechanisms of Triclosan-Induced Toxicity in Human Hepatocytes. Environ. Sci. Technol. 2019, 53, 5406–5415. [Google Scholar] [CrossRef]
  214. Alonso-Magdalena, P.; Laribi, O.; Ropero, A.B.; Fuentes, E.; Ripoll, C.; Soria, B.; Nadal, A. Low doses of bisphenol A and diethylstilbestrol impair Ca2+ signals in pancreatic alpha-cells through a nonclassical membrane estrogen receptor within intact islets of Langerhans. Environ. Health Perspect. 2005, 113, 969–977. [Google Scholar] [CrossRef] [Green Version]
  215. Stojanoska, M.M.; Milosevic, N.; Milic, N.; Abenavoli, L. The influence of phthalates and bisphenol A on the obesity development and glucose metabolism disorders. Endocrine 2017, 55, 666–681. [Google Scholar] [CrossRef]
  216. 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]
  217. Ghaemmaleki, F.; Mohammadi, P.; Baeeri, M.; Navaei-Nigjeh, M.; Abdollahi, M.; Mostafalou, S. Estrogens counteract tributyltin-induced toxicity in the rat islets of Langerhans. Heliyon 2020, 6, e03562. [Google Scholar] [CrossRef] [PubMed]
  218. Acosta-Montalvo, A.; Saponaro, C.; Kerr-Conte, J.; Prehn, J.H.M.; Pattou, F.; Bonner, C. Proglucagon-Derived Peptides Expression and Secretion in Rat Insulinoma INS-1 Cells. Front. Cell Dev. Biol. 2020, 8, 590763. [Google Scholar] [CrossRef] [PubMed]
  219. Lin, Y.; Sun, X.; Qiu, L.; Wei, J.; Huang, Q.; Fang, C.; Ye, T.; Kang, M.; Shen, H.; Dong, S. Exposure to bisphenol A induces dysfunction of insulin secretion and apoptosis through the damage of mitochondria in rat insulinoma (INS-1) cells. Cell Death Dis. 2013, 4, e460. [Google Scholar] [CrossRef] [Green Version]
  220. Pavlíková, N.; Daniel, P.; Šrámek, J.; Jelínek, M.; Šrámková, V.; Němcová, V.; Balušíková, K.; Halada, P.; Kovář, J. Upregulation of vitamin D-binding protein is associated with changes in insulin production in pancreatic beta-cells exposed to p,p′-DDT and p,p′-DDE. Sci. Rep. 2019, 9, 18026. [Google Scholar] [CrossRef] [Green Version]
  221. Huang, C.-F.; Yang, C.-Y.; Tsai, J.-R.; Wu, C.-T.; Liu, S.-H.; Lan, K.-C. Low-dose tributyltin exposure induces an oxidative stress-triggered JNK-related pancreatic β-cell apoptosis and a reversible hypoinsulinemic hyperglycemia in mice. Sci. Rep. 2018, 8, 5734. [Google Scholar] [CrossRef] [Green Version]
  222. Suh, K.S.; Choi, E.M.; Kim, Y.J.; Hong, S.M.; Park, S.Y.; Rhee, S.Y.; Oh, S.; Kim, S.W.; Pak, Y.K.; Choe, W.; et al. Perfluorooctanoic acid induces oxidative damage and mitochondrial dysfunction in pancreatic β-cells. Mol. Med. Rep. 2017, 15, 3871–3878. [Google Scholar] [CrossRef] [Green Version]
  223. Soriano, S.; Alonso-Magdalena, P.; García-Arévalo, M.; Novials, A.; Muhammed, S.J.; Salehi, A.; Gustafsson, J.A.; Quesada, I.; Nadal, A. Rapid insulinotropic action of low doses of bisphenol-A on mouse and human islets of Langerhans: Role of estrogen receptor β. PLoS ONE 2012, 7, e31109. [Google Scholar] [CrossRef] [Green Version]
  224. Marroqui, L.; Martinez-Pinna, J.; Castellano-Muñoz, M.; Dos Santos, R.S.; Medina-Gali, R.M.; Soriano, S.; Quesada, I.; Gustafsson, J.-A.; Encinar, J.A.; Nadal, A. Bisphenol-S and Bisphenol-F alter mouse pancreatic β-cell ion channel expression and activity and insulin release through an estrogen receptor ERβ mediated pathway. Chemosphere 2021, 265, 129051. [Google Scholar] [CrossRef]
  225. Nakashima, K.; Kanda, Y.; Hirokawa, Y.; Kawasaki, F.; Matsuki, M.; Kaku, K. MIN6 is not a pure beta cell line but a mixed cell line with other pancreatic endocrine hormones. Endocr. J. 2009, 56, 45–53. [Google Scholar] [CrossRef]
  226. Yamato, E.; Tashiro, F.; Miyazaki, J. Microarray analysis of novel candidate genes responsible for glucose-stimulated insulin secretion in mouse pancreatic β cell line MIN6. PLoS ONE 2013, 8, e61211. [Google Scholar] [CrossRef] [Green Version]
  227. Skelin, M.; Rupnik, M.; Cencic, A. Pancreatic beta cell lines and their applications in diabetes mellitus research. ALTEX 2010, 27, 105–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Miyazaki, S.; Tashiro, F.; Tsuchiya, T.; Sasaki, K.; Miyazaki, J.I. Establishment of a long-term stable β-cell line and its application to analyze the effect of Gcg expression on insulin secretion. Sci. Rep. 2021, 11, 477. [Google Scholar] [CrossRef] [PubMed]
  229. 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] [PubMed]
  230. ATCC. Beta-TC-6. Available online: https://www.atcc.org/products/crl-11506 (accessed on 12 January 2022).
  231. Qin, W.-P.; Cao, L.-Y.; Li, C.-H.; Guo, L.-H.; Colbourne, J.; Ren, X.-M. Perfluoroalkyl Substances Stimulate Insulin Secretion by Islet β Cells via G Protein-Coupled Receptor 40. Environ. Sci. Technol. 2020, 54, 3428–3436. [Google Scholar] [CrossRef]
  232. Ward, A.B.; Dail, M.B.; Chambers, J.E. In vitro effect of DDE exposure on the regulation of B-TC-6 pancreatic beta cell insulin secretion: A potential role in beta cell dysfunction and type 2 diabetes mellitus. Toxicol. Mech. Methods 2021, 31, 667–673. [Google Scholar] [CrossRef] [PubMed]
  233. Chandiramani, N.; Wang, X.; Margeta, M. Molecular basis for vulnerability to mitochondrial and oxidative stress in a neuroendocrine CRI-G1 cell line. PLoS ONE 2011, 6, e14485. [Google Scholar] [CrossRef]
  234. Pavlikova, N.; Smetana, P.; Halada, P.; Kovar, J. Effect of prolonged exposure to sublethal concentrations of DDT and DDE on protein expression in human pancreatic beta cells. Environ. Res. 2015, 142, 257–263. [Google Scholar] [CrossRef]
  235. Lieber, M.; Mazzetta, J.; Nelson-Rees, W.; Kaplan, M.; Todaro, G. Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrine pancreas. Int. J. Cancer 1975, 15, 741–747. [Google Scholar] [CrossRef]
  236. Hamil, L.; Benghuzzi, H.; Tucci, M. Evaluation of insulin secretion by pancreatic cells in response to increasing amounts of glucose. Biomed. Sci. Instrum. 2008, 44, 441–446. [Google Scholar]
Figure 1. Summary of the effects of daily exposed obesogenic EDCs on crucial organs (liver and pancreas) and tissues (adipose and brain) and the relationship of these effects with human obesity and diabetes. T2DM, Type 2 diabetes mellitus; NAFLD, non-alcoholic fatty liver disease.
Figure 1. Summary of the effects of daily exposed obesogenic EDCs on crucial organs (liver and pancreas) and tissues (adipose and brain) and the relationship of these effects with human obesity and diabetes. T2DM, Type 2 diabetes mellitus; NAFLD, non-alcoholic fatty liver disease.
Ijms 24 01083 g001
Figure 2. In vitro models applied in the study of the obesogenic action of endocrine-disrupting chemicals (EDCs): (A) animals, and (B) humans.
Figure 2. In vitro models applied in the study of the obesogenic action of endocrine-disrupting chemicals (EDCs): (A) animals, and (B) humans.
Ijms 24 01083 g002
Table 1. The obesogenic effects of selected EDCs confirmed on adipose tissue cell models.
Table 1. The obesogenic effects of selected EDCs confirmed on adipose tissue cell models.
Cell TypeOrganismIn vitro ModelEDCsMechanism of ActionConcentration *References
PreadipocytesAnimal3T3-L1 cell line4-HPadipogenic differentiation ↑
intracellular lipid accumulation ↑ (10 µM, 20 µM)
mRNA level of PPARγ, FABP4, CD36, perilipin, adiponectin ↑ (10 µM, 20 µM); C/EBPα (−) (10 µM, 20 µM)
10 µM, 20 µMSun et al.
[93]
BPAlipid accumulation ↑
mRNA and protein levels of PPARγ, C/EBPα, and AP2 ↑
20 µMChoi et al.
[7]
adipogenic differentiation (–) (1 nM, 10 nM), ↓ (100 nM)
mRNA level of PPARγ, FABP4, FASN (–) (1 nM, 10 nM),
↓ (100 nM); TNFα, IL6 ↑ (1 nM, 3 nM)
insulin-stimulated glucose uptake ↓ (1 nM)
1 nM, 3 nM,
10 nM, 100 nM
De Filippis et al. [96]
adipogenic differentiation ↑
lipid accumulation ↑ (100 nM)
glucocorticoid-like activity ↑ (1 µM)
PPARγ activity ↑ (however, less activation of PPARγ than GR) (1 µM)
protein expression of IR-β (insulin receptor subunit β) ↑; C/EBPα ↑ (1 µM–100 pM), adiponectin ↑ (10 nM, 100 nM)
100 pM, 1 nM,
10 nM, 100 nM,
1 µM
Sargis et al.
[97]
BPSlipid accumulation ↑
mRNA and protein levels of PPARγ, C/EBPα, and AP2 ↑
20 µMChoi et al.
[7]
BPFlipid accumulation ↑
mRNA and protein levels of PPARγ, C/EBPα, and AP2 ↑
20 µMChoi et al.
[7]
TBBPAAdipogenesis ↑
lipid accumulation ↑ (20 µM)
PPARγ activity ↑ (10, 20 µM)
20 µMAndrews et al. [120]
BBPadipogenic differention ↑
lipid accumulation ↑ (1 µM, 10 µM, 50 µM), (–) (0.001 µM, 0.01 µM, 0.1 µM)
miR-34a-5p expression level ↑ (1 µM, 10 µM, 50 µM), (−) (0.01 µM, 0.1 µM)
mRNA level of PPARγ ↑ (10 µM, 50 µM), (–) (0.01 µM, 0.1 µM, 1 µM);
AP2 ↑ (50 µM), (–) (0.01 µM, 0.1 µM, 1 µM);
NAMPT, SIRT1, SIRT3 ↓ (0.01 µM, 0.1 µM, 1 µM, 10 µM, 50 µM)
protein level of NAMPT ↓, SIRT1, SIRT3 (–) (0.1 µM, 50 µM)
NAD/NADH ratio ↓ (0.1 µM, 50 µM)
0.01 µM, 0.1 µM,
1 µM, 10 µM,
50 µM
Meruvu et al.
[98]
DCHPadipogenic differentiation ↑
lipid accumulation ↑ (100 nM)
glucocorticoid-like activity ↑ (1 µM)
PPARγ activity ↑ (however, less activation of PPARγ than GR) (1 µM)
protein expression of IR-β ↑, C/EBPα ↑ (1 µM–100 pM), adiponectin ↑ (1 µM–100 pM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µMSargis et al.
[97]
endrinadipogenic differentiation ↑
lipid accumulation ↑ (100 nM)
glucocorticoid-like activity ↑ (1 µM)
PPARγ activity ↑ (however, less activation of PPARγ than GR) (1 µM)
protein expression of IR-β ↑, C/EBPα ↑ (1 µM–100 pM), adiponectin ↑ (100 nM–100 pM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µMSargis et al.
[97]
TFadipogenic adifferentiation ↑
lipid accumulation ↑ (100 nM)
glucocorticoid-like activity ↑ (1 µM)
PPARγ activity ↑ (however, less activation of PPARγ than GR) (1 µM)
protein expression of IR-β ↑, C/EBPα ↑ (1 µM–100 pM), adiponectin ↑ (1 µM–100 pM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µMSargis et al.
[97]
QpEadipogenesis ↑
lipid accumulation ↑ (5–100 µM)
PPARγ activity ↑ (100 µM)
5–100 µMBiserni et al.
[99]
p,p’-DDTadipogenesis ↑
lipid accumulation ↑ (20 µM)
mRNA level of C/EBPα, PPARγ, FAS, ACC, ATGL, HSL, LEP ↑ (10 µM, 20 µM)
C/EBPα, PPARγ, AMPKα, ACC protein expression ↑ (10 µM, 20 µM)
phosphorylated forms of AMPKα, ACC protein expression ↓ (10 µM, 20 µM)
10 µM, 20 µMKim et al
[100]
adipogenesis ↑
lipid accumulation ↑ (10 µM, 20 µM, 30 µM, 50 µM)
protein level of C/EBPβ (–), C/EBPα ↑ (20 µM); PPARγ1, PPARγ2 ↑ (10 µM, 20 µM, 30 µM)
10 µM, 20 µM,
30 µM, 50 µM
Moreno-Aliaga and Matsumura [111]
p,p’-DDEadipogenesis ↑
lipid accumulation ↑ (10 µM, 20 µM)
mRNA level of C/EBPα, PPARγ, FAS, ACC, ATGL, HSL, LEP (10 µM, 20 µM), Lpl (20 µM) ↑
C/EBPα, PPARγ, AMPKα, Acc protein expression ↑ (10 µM, 20 µM)
phosphorylated forms of AMPKα, ACC protein expression ↓ (10 µM, 20 µM)
10 µM, 20 µMKim et al. [100]
adipogenesis ↑
lipid accumulation ↑ (10 µM, 20 µM)
mRNA level of SREBF1, FASN, PPARγ, LEP, FABP4 ↑ (2.5 µM, 10 µM, 20 µM)
2.5 µM, 10 µM,
20 µM
Mangum et al. [51]
diazinonadipogenesis ↑
lipid accumulation ↑ (1 µM, 10 µM, 25 µM, 50 µM, 100 µM)
mRNA level of C/EBPα, PPARγ, FASN ↑ (10 µM)
C/EBPα, PPARγ, FASN, ACC, adiponectin, perilipin protein expression ↑ (10 µM, 100 µM)
1 µM, 10 µM,
25 µM, 50 µM,
100 µM
Smith, Yu, Yin [101]
CPFadipogenesis ↑
lipid accumulation ↑ (10 µM, 50 µM)
mRNA level of C/EBPα, PPARγ, FABP4 ↑ (50 µM)
protein expression of C/EBPα, PPARγ, FABP4 ↑ (50 µM)
10 µM, 50 µMBlanco et al.
[102]
TBTadipogenesis ↑
lipid accumulation ↑ (50 nM)
PPARγ activity ↑ (5 nM, 50 nM, 100 nM)
basal glucose uptake (50 nM)
5 nM, 50 nM,
100 nM
Regnier et al. [103]
zoxamideadipogenesis ↑
lipid accumulation ↑ (0.02 µM)
PPARγ activity ↑ (EC50 = 1.31 µM, EC10 = 0.31 µM)
mRNA level of FABP4 ↑ (2 µM)
0.02 µM, 0.31 µM,
1.31 µM, 2 µM
Janesick et al. [104]
spirodiclofenadipogenesis ↑
lipid accumulation ↑ (0.02 µM, 0.2 µM, 2 µM, 10 µM, 20 µM)
PPARγ activity ↑ (EC50 = 12.76 µM, EC10 = 7.27 µM)
mRNA level of LPL ↑ (0.02 µM, 2 µM, 10 µM, 20 µM)
0.02 µM, 0.2 µM,
2 µM, 7.27 µM,
10 µM, 12.76 µM,
20 µM
Janesick et al. [104]
flusilazoleadipogenesis ↑
lipid accumulation ↑ (0.02 µM, 0.2 µM, 2 µM)
mRNA level of FABP4 ↑ (0.02 µM),
FSP27 ↑ (0.02 µM, 0.2 µM), LPL ↑ (0.02 µM)
0.02 µM, 0.2 µM, 2 µMJanesick et al. [104]
acetamipridadipogenesis ↑
lipid accumulation ↑ (0.2 µM)
mRNA level of FABP4, FSP27, LPL ↑ (0.2 µM)
0.2 µMJanesick et al. [104]
NIH3T3-L1 cell lineTBBPAadipogenesis ↑
lipid accumulation ↑
ERα, ERβ, PPARγ activity ↑
mRNA level of APOA2/FABP4 (AP2), PPARγ
10 µMRiu et al. [109]
TCBPAadipogenesis ↑
lipid accumulation ↑
ERα, ERβ, PPARγ activity ↑
mRNA level of APOA2/FABP4 (AP2), PPARγ
10 µMRiu et al. [109]
p,p’-DDEadipogenesis (−)
lipid accumulation (−) (2 µM, 20 µM)
basal fatty acid uptake ↑ (2 µM)
insulin-stimulated fatty acid uptake (−) (2 µM, 20 µM)
lipolysis (−) (2 µM, 20 µM)
leptin, resistin, adiponectin release ↑ (2 µM, 20 µM)
mRNA level of adiponectin, resistin ↑ (20 µM)
2 µM, 20 µMHowell et al. [80]
oxychlordaneadipogenesis (−)
lipid accumulation (−) (2 µM, 20 µM)
basal fatty acid uptake ↑ (20 µM)
insulin-stimulated fatty acid uptake (−) (2 µM, 20 µM)
lipolysis (−) (2 µM, 20 µM)
20 µMHowell et al.
[80]
dieldrinadipogenesis (−) (2 µM), ↓ (20 µM)
lipid accumulation (−) (2 µM), ↓ (20 µM)
basal fatty acid uptake ↑ (2 µM, 20 µM)
insulin-stimulated fatty acid uptake (−) (2 µM, 20 µM)
lipolysis (−) (2 µM, 20 µM)
adiponectin release ↑ (2 µM)
2 µM, 20 µMHowell et al
[80]
3T3-F442A cell lineBPAbasal glucose uptake ↑ (10−4 M)
insulin-stimulated glucose uptake ↑ (10−4 M, 10−6 M)
GLUT4 protein expression ↑ (10−4 M, 10−6 M)
10−4 M, 10−6 MSakurai et al.
[81]
p,p’-DDTadipogenesis ↓ (most cells did not differentiate completely)
C/EBPα protein level ↓
20 µMMoreno-Aliaga and Matsumura [111]
OP9 cell lineTBTpreadipocyte differentiation ↑
triglyceride accumulation ↑
100 nMKassotis et al. [39]
TBBPApreadipocyte differentiation ↑
triglyceride accumulation ↑
10 µMKassotis et al. [39]
adipogenesis ↑
lipid accumulation ↑ (20 µM)
PPARγ activity ↑ (10, 20 µM)
20 µMAndrews et al. [120]
TCBPApreadipocyte differentiation ↑
triglyceride accumulation ↑
10 µMKassotis et al. [39]
ST-13 cell lineTBBPAundifferentiated cells:
lipid accumulation (−) (0.5 µM, 1 µM)
mRNA level of AACS, PLIN1, FAS, CIDEA, LSD-1 ↑ (0.5 µM, 1 µM); UCP-1, UCP-3, PRDM16 ↑ (1 µM); SCOT, PPARγ (−) (0.5 µM, 1 µM)
mature adipocytes:
lipid accumulation (−) (0.5 µM, 1 µM)
mRNA level of AACS, SCOT (−) (0.5 µM, 1 µM)
0.5 µM, 1 µMYamasaki et al. [82]
UCP-1
cell line
CPFmRNA level of UCP1, CPT1A, CPT1B, ACAT3, COX16
mRNA level of PPARγ, PPARGC1A, PRDM16 (−)
mitochondrial respiration ↓
1 pMWang et al.
[29]
HumanPrimary preadipocytesBPAadipogenesis ↑
lipid accumulation ↑ (25 µM, 50 µM)
mRNA level of AP2, C/EBPα ↑ (25 µM, 50 µM), adipsin, PPARγ, C/EBPβ ↑ (50 µM)
protein level of AP2 ↑ (25 µM)
probable adipogenic action via a non-classical estrogen-receptor (ER) pathway rather than through the glucocorticoid-receptor (GR) activation
25 µM, 50 µMBoucher, Boudreau, and Atlas [75]
preadipocyte differentiation ↑
expression of genes ACACA, APOA1BP, PLIN2, FADS1, NPC2, PPAP2A
mRNA level of SREBF1, LDLR, LPL, INSIG1, GDF15
probable mechanism of action via mTOR signaling and TR/RXR activation
50 µMBoucher et al. [70]
adipogenesis ↑
lipid accumulation ↑ (10 nM, 1 µM, 80 µM)
mRNA level of 11β-HSD1, PPARγ ↑ (10 nM, 1 µM, 80 µM),
LPL ↑ (10 nM, 80 µM)
probable mechanism of action through GR pathway
10 nM,
1 µM, 80 µM
Wang et al
[76]
adipogenesis ↑
lipid accumulation ↑ (1 nM, 10 nM)
mRNA level of ERα ↑ (10 nM, 100 nM), ERRγ ↑ (10 nM), LEP ↑ (10 nM, 100 nM), ERβ (−) (1 nM, 10 nM, 100 nM), GPR30 ↓ (10 nM)
the expression of CD36, FABP4 ↑ (1 nM, 10 nM)
secretion of IL1B, IL18, CCL20 ↑ (10 nM)
1 nM,
10 nM, 100 nM
Menale et al. [124]
PCS-210-010 cellsBPAadipogenesis ↑
mRNA level of PPARγ, AP2, PPIA
adiponectin release ↑
0.059 µMEl-Atta et al.
[83]
SGBS cellsBPAlipid accumulation ↓ (10 nM, 100 nM, 1 µM, 10 µM)
binding to PPARγ ↑ (50 µM)
PPARγ activity (−) (10 nM, 100 nM, 1 µM, 10 µM)
protein level of FABP4 ↓ (10 nM, 100 nM, 1 µM), GPD1 ↓ (10 nM, 100 nM, 1 µM), LPL ↓ (100 nM, 1 µM), APOE ↓ (10 nM, 100 nM, 1 µM, 10 µM)
protein level of LIPE ↑ (10 nM, 100 nM, 1 µM, 10 µM), PNPLA2 ↑ (10 nM, 100 nM, 10 µM), CD36 ↑ (10 nM, 100 nM, 1 µM, 10 µM), ADIPOQ ↑ (10 nM, 100 nM, 10 µM)
release of MCP1 ↑ (1 µM), leptin ↑ (10 nM)
proteins related to oxidative stress level of CAT ↓ (100 nM, 1 µM), SOD2 ↓ (10 nM, 100 nM, 1 µM, 10 µM)
cellular ROS level ↓ (10 nM, 100 nM, 1 µM, 10 µM)
insulin sensitivity, pAKT/AKT ratio ↓ (1 µM)
10 nM, 100 nM,
1 µM, 10 µM,
50 µM
Schaffert et al.
[8]
BPSlipid accumulation ↓ (10 nM, 100 nM, 1 µM, 10 µM)
binding to PPARγ ↑ (50 µM)
PPARγ activity (−) (10 nM, 100 nM, 1 µM, 10 µM)
protein level of FABP4 ↓ (100 nM, 1 µM), GPD1 ↓ (10 nM, 100 nM, 1 µM, 10 µM), LPL ↓ (100 nM, 1 µM, 10 µM), APOE ↓ (10 nM, 100 nM, 1 µM, 10 µM)
protein level of LIPE ↑ (10 nM, 1 µM, 10 µM), PNPLA2 ↑ (10 nM, 100 nM, 1 µM, 10 µM), CD36 ↑ (10 nM, 1 µM, 10 µM), ADIPOQ ↑ (10 nM, 1 µM, 10 µM)
release of adiponectin ↓ (1 µM), LEP ↑ (10 nM)
proteins related to oxidative stress level of CAT ↓ (10 nM, 100 nM), SOD1 ↓ (10 µM), SOD2 ↓ (10 nM, 100 nM, 1 µM, 10 µM)
cellular ROS level ↓ (10 nM, 100 nM, 1 µM, 10 µM)
insulin sensitivity, pAKT/AKT ratio ↓ (1 µM)
10 nM, 100 nM, 1 µM, 10 µM, 50 µMSchaffert et al. [8]
BPBlipid accumulation ↓ (10 nM, 100 nM, 1 µM, 10 µM)
binding to PPARγ ↑ (50 µM)
PPARγ activity (−) (10 nM, 100 nM, 1 µM, 10 µM)
protein level of FABP4, GPD1, LPL, APOE, ADIPOQ ↓ (10 nM, 100 nM, 1 µM, 10 µM)
protein level of PNPLA2 ↑ (10 nM), CD36 ↑ (10 nM, 10 µM)
release of adiponectin ↓, MCP1 ↑, leptin ↓ (1 µM)
proteins related to oxidative stress level of CAT ↓ (10 nM, 100 nM, 1 µM, 10 µM), SOD1 ↓ (10 nM, 100 nM, 1 µM, 10 µM), SOD2 ↓ (10 nM, 100 nM, 1 µM)
cellular ROS level ↓ (10 nM, 100 nM, 1 µM, 10 µM)
insulin sensitivity, pAKT/AKT ratio ↓ (1 µM)
10 nM, 100 nM,
1 µM, 10 µM,
50 µM
Schaffert et al.
[8]
BPFlipid accumulation ↓ (10 nM, 100 nM, 1 µM, 10 µM)
binding to PPARγ ↑ (50 µM)
PPARγ activity (−) (10 nM, 100 nM, 1 µM, 10 µM)
protein level of FABP4, GPD1, LPL, APOE ↓ (10 nM, 100 nM, 1 µM, 10 µM)
protein level of LIPE ↑ (1 µM, 10 µM), PNPLA2 ↑ (10 µM), CD36 ↑ (10 nM, 100 nM, 1 µM, 10 µM), ADIPOQ ↑ (10 nM, 10 µM)
release of adiponectin ↓ (1 µM), MCP1 ↑ (1 µM), leptin ↑ (10 nM, 1 µM)
proteins related to oxidative stress level of CAT ↓ (10 nM, 100 nM, 1 µM, 10 µM), SOD1 ↓ (10 nM, 100 nM, 10 µM), SOD2 ↓ (100 nM, 1 µM, 10 µM)
cellular ROS level ↓ (10 nM, 100 nM, 1 µM, 10 µM)
10 nM, 100 nM,
1 µM, 10 µM
50 µM
Schaffert et al.
[8]
BPAFlipid accumulation ↓ (10 nM, 100 nM, 1 µM)
binding to PPARγ ↑ (50 µM)
PPARγ activity (−) (10 nM, 100 nM, 1 µM, 10 µM)
protein level of FABP4, GPD1, LPL, APOE ↓ (10 nM, 100 nM, 1 µM)
protein level of LIPE ↑ (10 nM, 100 nM), CD36 ↑ (10 nM, 1 µM), ADIPOQ ↑ (1 µM)
release of adiponectin ↓ (1 µM), leptin ↑ (10 nM)
proteins related to oxidative stress level of CAT ↓ (100 nM), SOD1 ↓ (10 nM, 100 nM, 1 µM), SOD2 ↓ 10 nM, 100 nM, 1 µM)
cellular ROS level ↓ (10 nM, 100 nM, 1 µM)
10 nM, 100 nM,
1 µM, 50 µM
Schaffert et al.
[8]
DEHPtriglyceride content ↓
mRNA level of ADIPOR2, GLUT4 (−), LEPR, CD36, FABP4, LPL, LIPE, ATGL
protein level of PPARα, PPARγ, SOD2, GPX1 (−)
secretion of adiponectin ↓, LEP ↑
ratio of pAMPK/AMPK, pSTAT3/STAT3 (−), pACC2/ACC2 ↑
phosphorylation of ERK1, ERK2 ↓
lipolysis ↑, level of free glycerol ↑
ROS level ↑
50 µg/mLSchaedlich et al. [84]
DINCHbinding to PPARγ ↑ (−) (6.25 µM, 12.5 µM, 25 µM, 50 µM, 100 µM, 200 µM, 400 µM)
PPARγ activation (−)
undifferentiated cells:
lipid accumulation (−) (10 nM, 1 µM, 10 µM, 25 µM, 50 µM, 100 µM)
secretion of adipsin ↑ (10 nM), MCP-1 ↓ (10 µM)
mature adipocytes:
lipid accumulation ↓ (10 µM, 25 µM, 50 µM, 100 µM)
secretion of LEP ↑ (10 nM, 10 µM), adipsin ↑ (10 nM, 10 µM), MCP-1 ↑ (10 nM, 10 µM), adiponectin ↓ (10 nM, 10 µM)
protein level of GPX1 ↑ (10 µM), GPX4 ↑ (10 µM), GSTO1 ↑ (10 nM, 10 µM), LAP3 ↑ (10 µM)
10 nM, 10 µM,
25 µM, 50 µM,
100 µM
Schaffert et al. [62]
DINPbinding to PPARγ ↑ (−) (6.25 µM, 12.5 µM, 25 µM, 50 µM, 100 µM, 200 µM, 400 µM, PPARγ activation (−)
undifferentiated cells:
lipid accumulation (−) (10 nM, 1 µM, 10 µM, 25 µM, 50 µM, 100 µM), secretion of adipsin ↑ (10 nM)
mature adipocytes:
lipid accumulation (−) (10 nM, 1 µM, 10 µM, 25 µM, 50 µM, 100 µM), secretion of LEP ↑ (10 nM, 10 µM), adipsin ↑ (10 µM), MCP-1 ↑ (10 µM), adiponectin ↓ (10 nM, 10 µM)
protein level of GPX1 ↑ (10 µM), GPX4 ↑ (10 nM, 10 µM), GPX8 ↑ (10 nM), GSR ↑ (10 nM), GSTO1 ↑ (10 nM, 10 µM), LAP3 ↑ (10 µM)
10 nM, 10 µMSchaffert et al. [62]
DPHPbinding to PPARγ ↑ (−) (6.25 µM, 12.5 µM, 25 µM, 50 µM, 100 µM, 200 µM, 400 µM), PPARγ activation (−)
undifferentiated cells:
lipid accumulation (−) (10 nM, 1 µM, 10 µM, 25 µM, 50 µM, 100 µM), secretion of MCP-1 ↓ (10 nM, 10 µM)
mature adipocytes:
lipid accumulation ↓ (10 µM, 25 µM, 50 µM, 100 µM)
secretion of LEP ↑ (10 µM), adipsin ↑ (10 µM), MCP-1 ↑ (10 µM), adiponectin ↓ (10 nM, 10 µM)
protein level of GPX1 ↑ (10 nM, 10 µM), GPX4 ↑ (10 µM), GPX8 ↑ (10 µM), GSR ↑ (10 nM), GSTO1 ↑ (10 nM, 10 µM), LAP3 ↑ (10 µM)
10 nM, 10 µM,
25 µM, 50 µM,
100 µM
Schaffert et al. [62]
MINCHbinding to PPARγ ↑ (25 µM, 50 µM, 100 µM, 200 µM, 400 µM)
PPARγ activation (−)
undifferentiated cells:
preadipocyte differentiation ↑
lipid accumulation ↑ (10 µM, 25 µM, 50 µM)
secretion of LEP ↑ (10 µM), adipsin ↑ (10 nM), MCP-1 ↓ (10 nM, 10 µM)
protein level of FABP4, FASN, FABP5, GPD1, PLIN1 ↑ (10 µM)
mature adipocytes:
lipid accumulation ↓ (10 µM, 25 µM, 50 µM)
secretion of LEP ↑ (10 nM, 10 µM), adipsin ↑ (10 µM), MCP-1 ↑ (10 nM, 10 µM), adiponectin ↓ (10 nM, 10 µM)
protein level of GPX1 ↑ (10 µM), GPX4 ↑ (10 µM), GPX8 ↑ (10 nM, 10 µM), GSTO1 ↑ (10 nM, 10 µM), LAP3 ↑ (10 µM)
10 nM, 10 µM,
25 µM, 50 µM,
100 µM, 200 µM,
400 µM
Schaffert et al. [62]
MHINPbinding to PPARγ ↑ (100 µM, 200 µM, 400 µM)
PPARγ activation ↑ (1 µM)
undifferentiated cells:
preadipocyte differentiation ↑
lipid accumulation ↑ (10 µM, 25 µM, 50 µM, 100 µM)
secretion of LEP ↑ (10 µM), adipsin ↑ (10 nM, 10 µM), MCP-1 ↓ (10 nM)
protein level of FABP4, FASN, FABP5, GPD1, PLIN1 ↑ (10 µM)
mature adipocytes:
lipid accumulation ↑ (1 µM)
secretion of LEP ↑ (10 nM, 10 µM), adipsin ↑ (10 µM), MCP-1 ↑ (10 nM, 10 µM), adiponectin ↓ (10 nM, 10 µM)
protein level of GPX1 ↑ (10 nM, 10 µM), GPX4 ↑ (10 nM, 10 µM), GPX8 ↑ (10 nM), GSR ↑ (10 nM), GSTO1 ↑ (10 nM, 10 µM), LAP3 ↑ (10 µM)
10 nM, 10 µM,
25 µM, 50 µM,
100 µM, 200 µM,
400 µM
Schaffert et al. [62]
OH-MPHPbinding to PPARγ ↑ (200 µM, 400 µM)
PPARγ activation ↑ (10 µM)
undifferentiated cells:
preadipocyte differentiation ↑
lipid accumulation ↑ (10 µM, 25 µM, 50 µM)
secretion of LEP ↑ (10 µM)
protein level of FABP4, FASN, FABP5, GPD1 ↑ (10 µM)
mature adipocytes:
lipid accumulation ↓ (10 nM, 10 µM, 25 µM, 50 µM, 100 µM)
secretion of LEP ↑ (10 nM, 10 µM), adipsin ↑ (10 µM), MCP-1 ↑ (10 nM, 10 µM), adiponectin ↓ (10 nM, 10 µM)
protein level of GPX1 ↑ (10 µM), GPX4 ↑ (10 µM), GPX8 ↑ (10 µM), GSR ↑ (10 nM, 10 µM), GSTO1 ↑ (10 nM, 10 µM), LAP3 ↑ (10 µM)
10 nM, 10 µM,
25 µM, 50 µM,
100 µM, 200 µM,
400 µM
Schaffert et al. [62]
SW 872
cell line
MEHPdifferentiating effect ↑
lipid accumulation (−)
mRNA level of TSPO, PKCε, PPARα, ACACA, ACLY, GLUT1, GLUT4, S100B ↑, PPARγ ↓, PPARβ/δ (−)
protein level of TSPO ↓
10 µMCampioli et al. [129]
Legend: ↑ increase; ↓ decrease; (−) no observed effects; * concentration (s) at which obesogenic effects were observed.
Table 2. The obesogenic effect of selected EDCs confirmed on mesenchymal stem cell models.
Table 2. The obesogenic effect of selected EDCs confirmed on mesenchymal stem cell models.
Cell TypeOrganismIn vitro ModelEDCsMechanism of ActionConcentration *References
Adipose-derived mesenchymal stem cells (ADSCs)AnimalC3H10T1/2
cell line
BPAadipogenic differentiation (− (1 nM, 3 nM)
lipid accumulation (−) (1 nM, 3 nM)
mRNA level of PPARγ, C/EBPα and FABP4 (−) (1 nM, 3 nM)
1 nM, 3 nMDe Filippis et al. [96]
whole period of adipogenic differentiation: amount of adipocytes ↓, triglyceride content ↓, mRNA level of FABP4, PPARγ, LPL, adiponectin ↓ (10 µM); amount of adipocytes (−), triglyceride content (−) (10 nM)
selective treatment during undifferentiated growth: amount of adipocytes ↓, triglyceride content ↓ (10 µM);
hormonal induction: amount of adipocytes (−), triglyceride content (−) (10 µM);
terminal differentiation: amount of adipocytes (−), triglyceride content (−) (10 µM)
10 µMBiemann et al. [139]
DEHPwhole period of adipogenic differentiation: amount of adipocytes ↑, triglyceride content ↑ (100 µM); amount of adipocytes (−), triglyceride content (−) (100 nM)
selective treatment during undifferentiated growth: amount of adipocytes (−), triglyceride content (−) (100 µM); hormonal induction: amount of adipocytes ↑, triglyceride content ↑ (100 µM); terminal differentiation: amount of adipocytes (−), triglyceride content (−) (100 µM)
100 µMBiemann et al. [139]
BBPadipogenesis ↑
lipid accumulation ↑
mRNA level of AP2, PPARγ ↑, SIRT1, SIRT7, PGC1α, NRF1, NRF2, TFAM ↓; SIRT2, SIRT3, SIRT4, SIRT5, SIRT6 (−)
protein level of FOXO1 ↑, SIRT1, SIRT3 ↓, SIRT7, β-catenin (−)
acetylation of FOXO1, β-catenin ↑
50 µMZhang and Choudhury [138]
expression of miR-103/107, miR-let7 (a, b, c, d, f, g) ↑, lncRNA H19 ↓, miR-let7e (−) at day 2 of differentiation
mRNA level of IRS-1 ↓, IR, IRS-2 (−) at day 2, 4, 6, 8 of differentiation
protein expression of phospho-Akt ↓ at day 4 of differentiation
50 µMZhang and Choudhury [137]
TBTwhole period of adipogenic differentiation: amount of adipocytes ↑, triglyceride content ↑ (100 nM); amount of adipocytes (−), triglyceride content (−) (1 nM)
selective treatment during undifferentiated growth: amount of adipocytes ↑, triglyceride content ↑ (100 nM);
hormonal induction: amount of adipocytes ↑, triglyceride content ↑ (100 nM);
terminal differentiation: amount of adipocytes ↑, triglyceride content ↑ (100 nM)
100 nMBiemann et al. [139]
butylparabenadipogenic differentiation ↑
lipid accumulation ↑
PPARγ activity ↑
GR activity (−)
mRNA level of PPARγ, C/EBPα, FABP4 ↑, RUNX2
100 µMHu et al.
[130]
mixture of
BPA, DEHP,
TBT
BPA (10 nM), DEHP (100 nM) and TBT (1 nM)
whole period of adipogenic differentiation, undifferentiated growth, hormonal induction and terminal differentiation: amount of adipocytes (−), triglyceride content (−), mRNA level of FABP4, adiponectin, LPL, PPARγ2 (−)
BPA (10 µM), DEHP (100 µM) and TBT (100 nM)
whole period of adipogenic differentiation: amount of adipocytes ↑, triglyceride content (−), mRNA level of FABP4, adiponectin ↑, LPL, PPARγ2 (−);
undifferentiated growth: amount of adipocytes ↑, triglyceride content (−), mRNA level of FABP4 ↑, adiponectin, LPL, PPARγ2 (−);
hormonal induction: amount of adipocytes ↑, triglyceride content (−), mRNA level of FABP4, adiponectin, LPL, PPARγ2 ↑;
terminal differentiation: amount of adipocytes ↑, triglyceride content (−); mRNA level of FABP4 ↑, adiponectin, Lpl, PPARγ2 (−)
1 nM, 10 nM,
100 nM, 10 µM,
100 µM
Biemann, Fisher and Navarrete Santos
[140]
Primary mouse ADSCsTBTadipogenesis ↑
lipid accumulation ↑
mRNA level of FABP4, PPARΓ2, LEP ↑, PREF-1 ↓, ADIPOQ (−)
50 nMKirchner et al. [78]
Primary porcine ADSCsBPSviability of cells ↓
lipid accumulation ↓
1 µMBerni et al.
[143]
GLYviability of cells ↓
adipogenic differentiation ↓
4 µg/mLGigante et al. [142]
HumanPrimary hADSCsBPAadipogenic differentiation (−) (1 nM, 3 nM)
lipid accumulation (−) (1 nM, 3 nM)
mRNA level of PPARγ, C/EBPα and FABP4 (−) (1 nM, 3 nM)
1 nM, 3 nMDe Filippis et al. [96]
mRNA level of PPARγ, GLUT4 (−)
insulin-stimulated glucose utilization ↓
insulin-activated insulin receptor (IR) tyrosine phosphorylation ↓, ERK1/2 phosphorylation ↓, PKB/Akt phosphorylation ↓
release of IL6, IFN-γ ↑
JAK/STAT, JNK, NFkB pathways activity ↑
1 nMValentino et al. [147]
adipogenesis ↑
lipid accumulation ↑
mRNA level of ERα ERβ, IGF1, PPARγ, LPL, C/EBPα, DLK ↑, AP2, SREBP1c, C/EBPβ (−)
protein level of LPL ↑
probable mechanism of action through ER-dependent pathway
1 µMOhlstein et al. [148]
adipogenesis ↑ (0.1 µM), ↓ (1 µM)
lipid production ↑ (0.1 µM), ↓ (1 µM)
0.1 µM, 1 µMCohen et al.
[55]
cell viability ↓ (1 µM, 10 µM)
apoptosis ↑ (1 µM)
caspase-6 activity (1 µM)
1 µM, 10 µMHarnett et al. [56]
BPFadipogenesis ↑
lipid accumulation ↑ (10 µM, 25 µM)
mRNA level of PPARγ, LPL, FABP4 ↑ (10 µM, 25 µM); C/EBPα ↑ (1 µM, 10 µM, 25 µM)
protein level of PPARγ, C/EBPα, LPL ↑ (10 µM, 25 µM)
probable mechanism of action via an ER-dependent pathway
1 µM,
10 µM, 25 µM
Reina-Pérez et al. [79]
BPSadipogenesis ↑
lipid accumulation ↑ (1 µM, 10 µM, 25 µM)
mRNA level of PPARγ, C/EBPα, LPL ↑ (1 µM, 10 µM, 25 µM); FABP4 ↑ (10 µM, 25 µM)
protein level of PPARγ ↑ (10 µM, 25 µM); C/EBPα, LPL ↓ (10 µM, 25 µM)
1 µM, 10 µM, 25 µMReina-Pérez et al. [79]
BPAFadipogenesis ↑ (0.1 nM), (−) (1 nM), ↓ (10 nM)
lipid production ↑ (0.1 nM), (−) (1 nM), ↓ (10 nM)
0.1 nM, 10 nMCohen et al. [55]
cell viability ↓ (0.0003 µM, 0.003 µM, 0.03 µM, 0.3 µM)
apoptosis ↑ (0.003 µM)
caspase-6 activity (0.003 µM)
0.0003 µM, 0.003 µM,
0.03 µM, 0.3 µM
Harnett et al. [56]
TMBPFadipogenesis ↓
lipid production ↓
0.01 µM, 0.1 µMCohen et al.
[55]
cell viability ↓ (0.01 µM, 0.1 µM, 1 µM, 10 µM, 50 µM)
apoptosis ↑ (1 µM)
caspase-6 activity (1 µM)
0.01 µM, 0.1 µM,
1 µM, 10 µM, 50 µM
Harnett et al. [56]
TBTadipogenesis ↑
lipid accumulation ↑
mRNA level of FABP4, PPARγ2, LEP ↑, PREF-1 ↓, ADIPOQ (−)
50 nMKirchner et al. [78]
p,p’-DDEmaintenance of cells in an undifferentiated state ↑
mRNA level of OCT4, PPARγ, SREBP1, FASN, INSR, AKT2, UCP-3 ↑ (0.1 µM, 1 µM, 10 µM); SOX2 ↑ (1 µM, 10 µM); PPARγC1B ↑ (0.1 µM, 1 µM); NANOG ↓ (0.1 µM, 1 µM, 10 µM)
0.1 µM,
1 µM, 10 µM
Pesta et al.
[77]
Legend: ↑ increase; ↓ decrease; (−) no observed effects; * concentration (s) at which biological effects were observed.
Table 3. The obesogenic effects of selected EDCs confirmed in hepatic cellular models.
Table 3. The obesogenic effects of selected EDCs confirmed in hepatic cellular models.
Cell TypeOrganismIn vitro
Model
EDCMechanism of ActionConcentration *References
HepatocytesAnimalPrimary mouse hepatocytesBHPFcell viability ↓
LDH activity ↑
1 µM, 10 µMYang et al.
[165]
Primary gilthead seabream hepatocytesDIDPbinding to PPARγ, PPARα, RXRα ↑
mRNA level of PPARA, PPARΒ, PPARγ, CPT1A, FADS2, SCD1A, SCD1B, LPL, HL, FABP, APOA-I, SREBP1 ↑ (0.1 µM, 1 µM), (−) (10 µM), CPT1B ↑ (1 µM), (−) (10 µM)
probable mechanism of action via PPAR:RXR signaling
0.1 µM, 1 µMCocci et al.
[169]
Primary Atlantic salmon hepatocytesp,p’-DDEviability of cells ↓ (100 μM), (−) (0.1 μM, 1 μM, 10 μM)
global DNA methylation (−) (0.1 μM, 1 μM, 10 μM, 100 μM)
mRNA level of stress-responsive genes such as estrogenic markers ESR1 ↑ (10 μM), (−) (0.1 μM, 1 μM, 100 μM); ESR2 (−) (0.1 μM, 1 μM, 10 μM, 100 μM); VTG1 ↑ (1 μM, 10 μM), (−) (0.1 μM, 100 μM); markers of detoxification CYP1A1, CYP3A (−) (0.1 μM, 1 μM, 10 μM, 100 μM); genes whose protein products are associated with cell death HSPA8, FOS ↑ (100 μM), (−) (0.1 μM, 1 μM, 10 μM); CASP3B, PTGS2 ↓ (100 μM), (−) (0.1 μM, 1 μM, 10 μM); CDKN1B,
INSIG1 (−) (0.1 μM, 1 μM, 10 μM, 100 μM)
mRNA level of DNA methylation-relevant genes DNMT3AA, CBS, N6AMT2, MAT1A2 ↓ (100 μM), (−) (0.1 μM, 1 μM, 10 μM); DNMT1 (−) (0.1 μM, 1 μM, 10 μM, 100 μM)
bile acid metabolism (level of glycocholate, glycochenodeoxycholate, taurocholate, taurochenodeoxycholate, deoxycholate, glycodeoxycholate, taurolithocholate ↑ (100 μM))
glucose metabolism (level of glycolytic intermediates such as 3-phosphoglycerate, phosphoenolpyruvate, glucose-6-phosphate, glucose ↓, pyruvate ↑, pentose sugars such as arabonate/xylonite, arabitol/xylitol, sedoheptulose ↓, pentose phosphate pathway intermediate such as 5-phosphogluconate ↓, glycogen hydrolysis products such as maltotriose, maltotetraose, maltose ↓ (100 μM))
amino acids metabolism (level of glutamine, N-acetylglutamine, glutamate, N-acetylglutamate ↓, gamma-aminobutyrate ↑, (100 μM))
lipid metabolism (level of monoacylglycerols such as 1-mirystoylglycerol ↑, diacylglycerols such as 1-palmitoyl-2-arachidononyl-GPE, phosphatidylethanolamine species such as oleoyl-oleoyl-glycerol ↓ (100 μM))
1 μM, 10 μM,
100 μM
Olsvik and Søfteland [170]
BRL-3A
cell line
MEHPlipid accumulation ↑ (100 µM, 200 µM)
mRNA level of FAS, PDK4, aP2 ↑ (10 µM, 50 µM, 100 µM, 200 µM); AOX, PPARγ ↑ (50 µM, 100 µM, 200 µM); JAK2, STAT5A ↓ (50 µM, 100 µM, 200 µM), STAT5B ↓ (10 µM, 50 µM, 100 µM, 200 µM)
protein level of AOX, PDK4, FAS ↑ (100 µM, 200 µM); aP2 ↑ (50 µM, 200 µM; PPARγ ↑ (200 µM); JAK2, STAT5A, STAT5B ↓ (10 µM, 50 µM, 100 µM, 200 µM);
JAK2/STAT5 signalling ↓
level of indicators of oxidative stress: SOD ↓, MDA ↑ (10 µM, 50 µM, 100 µM, 200 µM)
level of indicators of damage status of liver cells: ALT ↑ (50 µM, 100 µM, 200 µM), AST ↑ (10 µM, 50 µM, 100 µM, 200 µM)
10 µM, 50 µM,
100 µM, 200 µM
Zhang et al. [173]
FaO cell lineBPAintracellular lipid content ↑ (30 ng/mL, 300 ng/mL)
mRNA level of PPARα, PPARβ, PPARδ, PPARγ, AOX, CPT1, APOB ↓, FAS, GPAT (−) (300 ng/mL)
30 ng/mL and 300 ng/mL corresponding to 0.1 µM and 1 µMGrasselli et al.
[171]
AML12
cell line
PCB-153lipid accumulation ↑
mRNA level of p65 subunit of NFkB, IL1α, IL6 ↑, HNF1B, GPX1
nuclear protein expression of p65 subunit of NFkB ↑, cytoplasmic protein expression of p65 subunit of NFkB ↓
protein level of HNF1b, GPX1 ↓
ROS level ↑
ratio of glutathione (GSH)/ /oxidized GSH (GSSG) ↓
ratio of NADP+/NADPH ↑
insulin-stimulated glucose uptake ↓
0.5 µM, 1 µMWu et al.
[175]
TCEPlipid accumulation ↑ (10 µM)
disturbance of mitochondrial structure ↑ (1 µM, 10 µM)
mitoATP rate/glycoATP rate ↓ (1 µM, 10 µM)
1 µM, 10 µMLe et al
[176]
TCPPlipid accumulation ↑ (10 µM)
disturbance of mitochondrial structure ↑ (1 µM, 10 µM)
mitochondrial membrane potential (MMP) ↓ (10 µM)
mitoATP rate/glycoATP rate ↓ (10 µM)
1 µM, 10 µMLe et al.
[176]
TDCPPlipid accumulation ↑ (1 µM, 10 µM)
disturbance of mitochondrial structure ↑ (1 µM, 10 µM)
mitochondrial ROS production ↑ (1 µM, 10 µM)
MMP ↓ (10 µM)
mitochondrial basal respiration ↓ (10 µM)
proton leak ↓ (10 µM)
mitoATP production rate ↓ (10 µM)
mitoATP rate/glycoATP rate ↓ (10 µM)
1 µM, 10 µMLe et al.
[176]
TPhPlipid accumulation ↑ (1 µM, 10 µM)
disturbance of mitochondrial structure ↑ (1 µM, 10 µM)
mitochondrial ROS production ↑ (1 µM, 10 µM)
MMP ↓ (1 µM, 10 µM)
mitochondrial basal respiration ↓ (10 µM)
proton leak ↓ (10 µM)
spare respiratory capacity (SRC) ↑ (10 µM)
mitoATP production rate ↓ (10 µM)
mitoATP rate/glycoATP rate ↓ (10 µM)
1 µM, 10 µMLe et al.
[176]
TCPlipid accumulation ↑ (0.1 µM, 1 µM, 10 µM)
disturbance of mitochondrial structure ↑ (1 µM, 10 µM)
mitochondrial ROS production ↑ (1 µM, 10 µM)
MMP ↓ (1 µM, 10 µM)
mitochondrial basal respiration ↓ (10 µM)
proton leak ↓ (10 µM)
SRC ↑ (10 µM)
mitoATP production rate ↓ (10 µM)
mitoATP rate/glycoATP rate ↓ (10 µM)
0.1 µM, 1 µM, 10 µMLe et al.
[176]
Hepa1-6
cell line
BPAmRNA level of DNA methyltransferases: DNMT1, DNMT3A ↓ (0.001 µM, 0.01 µM); DNMT3B ↓ (0.001 µM)
mRNA level of SREBF1, SREBF2, FASN, HMGCR ↑ (0.001 µM, 0.01 µM)
0.001 µM, 0.01 µMKe et al.
[178]
FL83B
cell line
DEHPcell viability ↓ (250 µM, 500 µM, 1000 µM)
LDH release ↑ (125 µM, 250 µM, 500 µM, 1000 µM), ALT release ↑ (500 µM, 1000 µM)
cell populations of sub-G1 and S phase ↑ (250 µM, 500 µM, 1000 µM)
125 µM, 250 µM,
500 µM, 1000 µM
Lo et al.
[185]
RTL-W1
cell line
BPAlipid accumulation ↑
alteration of membrane lipids (phosphatidylcholines (PCs), plasmalogen PCs)
mRNA level of ABCA1, CD36, FATP1, FAS, LPL, PPARα, PPARβ
10 µMDimastrogiovanni et al. [186]
TBTalteration of membrane lipids (phosphatidylcholines (PCs), plasmalogen PCs)
mRNA level of ABCA1, CD36, FAS, LPL
100 nMDimastrogiovanni et al. [186]
TPTmRNA level of ABCA1, FATP1, FAS100 nMDimastrogiovanni et al. [186]
DEHPlipid accumulation ↑
alteration of membrane lipids (phosphatidylcholines (PCs), plasmalogen PCs)
mRNA level of CD36, FAS, LPL
5 µMDimastrogiovanni et al. [186]
4-NPalteration of membrane lipids (phosphatidylcholines (PCs), plasmalogen PCs)
mRNA level of ABCA1 ↑, CD36, FAS, LPL, PPARβ
20 µMDimastrogiovanni et al. [186]
PLHC-1
cell line
DBPtriacylglyceride accumulation ↑ (20 µM), (−) (5 µM)
ROS generation ↑ (5 µM, 20 µM, 50 µM, 100 µM)
5 µM, 20 µM,
50 µM, 100 µM
Pérez-Albaladejo et al. [191]
DEHPtriacylglyceride accumulation ↑ (5 µM, 10 µM)
ROS generation ↑ (100 µM)
5 µM, 10 µM,
100 µM
Pérez-Albaladejo et al. [191]
BADGE·2HCltriacylglyceride accumulation ↓ (1 µM, 5 µM)
ROS generation ↑ (100 µM)
1 µM, 5 µM,
100 µM
Pérez-Albaladejo et al. [191]
TBTintracellular accumulation of triglycerides and diglycerides ↑100 nM, 200 nMMarqueño et al.
[188]
ZFL cell lineBPAlipid accumulation: dihydroceramides ↑ (50 µM), ether-triacylglycerides (ether-TGs), saturated and lower unsaturated TGs ↑ (5 µM, 50 µM)
mRNA level of SCD, ELOVL6 ↑, LXR, FASN, GPAT3, DGAT1A (−) (20 µM)
ROS production ↑ (20 µM, 50 µM, 70 µM, 100 µM, 150 µM, 200 µM)
5 µM, 20 µM,
50 µM, 70 µM,
100 µM, 150 µM,
200 µM
Marqueño et al.
[190]
BPFlipid accumulation: dihydroceramides, ether-triacylglycerides (ether-TGs) ↑ (50 µM), TGs containing polyunsaturated fatty acids (PUFAs) ↓ (50 µM)
mRNA level of SCD, ELOVl6, ABCA1B, CYP3A65 ↑, PPARα ↓, LXR, FASN, GPAT3, DGAT1a (−) (20 µM)
ROS production ↑ (5 µM, 20 µM, 50 µM, 100 µM, 150 µM, 200 µM, 500 µM)
5 µM, 20 µM,
50 µM, 100 µM,
150 µM, 200 µM,
500 µM
Marqueño et al.
[190]
BADGE·2HCllipid accumulation: dihydroceramides, ether-TGs) ↑ (10 µM), saturated and lower unsaturated TGs ↑ (5 µM)
mRNA level of ABCA1b, PPARα ↓, LXR, FASN, GPAT3, DGAT1A (−) (5 µM)
ROS production (−) (20 µM, 50 µM, 60 µM, 70 µM, 80 µM, 100 µM)
5 µM, 10 µMMarqueño et al.
[190]
HumanPrimary hepatocytesTCDDfatty acids accumulation ↑10 nMForgacs et al.
[90]
HepG2
cell line
4-HPlipid deposition in OA (oleic acid)-treated cells ↑
de novo lipogenesis ↓
fatty acid oxidation ↓
mRNA level of CD36
mRNA level of PPARα, SREBP1c, CPT1A, ACC ↓, PPARγ (−)
20 µMSun et al.
[93]
1,3-DCPcell viability ↓ (250 µg/mL, 500 µg/mL),
(−) (0.1 µg/mL, 0.5 µg/mL, 1 µg/mL, 5 µg/mL, 25 µg/mL, 50 µg/mL)
intracellular TG and TC (total cholesterol) content ↑ (0.5 µg/mL, 1 µg/mL, 2 µg/mL)
level of cAMP, AMP, ADP ↓, ATP (−) (0.5 µg/mL, 1 µg/mL, 2 µg/mL)
intracellular calcium level (−) (0.5 µg/mL, 1 µg/mL, 2 µg/mL)
mRNA level of LDLR, SREBP2, HMGCR ↑ (0.5 µg/mL, 1 µg/mL, 2 µg/mL)
protein level of SREBP1c, SCD1, FAS, CD36, HMGCR, GPAT ↑, CREB, LKB1, HSL, p-AMPK, p-PKA, pACC, PGC1α, PPARα, SIRT1, CPT1, HNF4α ↓ (0.5 µg/mL, 1 µg/mL, 2 µg/mL), GPR41, GPR43 ↓, GPR109B (−) (2 µg/mL), Caldumolin 1, Calpain 1, Calpain 2, CaMKII, p-CaMKII (−) (0.5 µg/mL, 1 µg/mL, 2 µg/mL)
Gi/o expression ↑ (2 µg/mL)
probable mechanism of action through cAMP/PKA and AMPK signaling pathways via Gi/o-coupled receptor
0.5 µg/mL, 1 µg/mL,
2 µg/mL, 250 µg/mL, 500 µg/mL
Lu et al.
[199]
TCSlevel of diacylglycerol (DG) ↑ (1 µM, 10 µM), phosphatidylcholine (PC), sphingomyelin (SM), phosphatidylethanolamine (PE) ↑ (10 µM), ceramide (Cer) ↓ (1 µM, 10 µM), triglyceride (TG), polyphosphoinositide (PI), lysophosphatidylethanolamine (LPE), phosphatidylglycerol (PG), lysophosphatidylcholine (LPC) ↓ (10 µM)
ROS generation ↑ (10 µM), (−) (1 µM)
MDA content (−) (1 µM, 10 µM)
SOD activity ↑ (10 µM), (−) (1 µM)
CAT activity ↑ (10 µM), (−) (1 µM)
level of GSH ↑ (10 µM), (−) (1 µM), taurine (−) (1 µM, 10 µM)
1 µM, 10 µMZhang et al.
[213]
DINCHcell viability (−) (1 µg/mL, 5 µg/mL, 10 µg/mL, 100 µg/mL, 250 µg/mL, 500 µg/mL)
oxidative DNA damage at 3 h exposure ↑ (1 µg/mL, 10 µg/mL, 100 µg/mL, 250 µg/mL, 500 µg/mL), at 24 h exposure ↑ (100 µg/mL)
1 µg/mL, 10 µg/mL,
100 µg/mL, 250 µg/mL, 500 µg/mL
Vasconcelosa, Silva and Louroa
[57]
HepaRG
cell line
BPAtriglycerides and neutral lipids accumulation ↑ (2 nM)
mRNA level of lipid-responsive genes such as APOA4 ↑, PLIN2 (ADFP or ADRP), TIP47 (−) (0.2 nM, 2 nM, 20 nM, 200 nM, 2000 nM)
mRNA level of genes involved in lipid and carbohydrate homeostasis FASN, ACLY, ACACA, HMGCR, PPARγ, PNPLA3, THRSP (SPOT14), PDK4, APOB, GLUT2 (SLC2A2), MTTP, CPT1A (−) (0.2 nM, 2 nM, 20 nM, 200 nM, 2000 nM)
mRNA level of genes involved in oxidative stress NFKB1, HMOX1, GSTA1/2, GSTA3, NQO1, TRIB3, HSPA1A (HSP70-1A) (−) (0.2 nM, 2 nM, 20 nM, 200 nM, 2000 nM)
mRNA level of ERRγ target genes SDHD, PCK1, ESRRA (−) (0.2 nM, 2 nM, 20 nM, 200 nM, 2000 nM)
mRNA level of PXR target genes CD36, CYP2C9, CYP3A4 (−) (0.2 nM, 2 nM, 20 nM, 200 nM, 2000 nM)
mRNA level of genes involved in BPA biotransformation CYP2C19, CYP2C18, SULT1A1, SULT1A3/4, UGT2B15, STS, GUSB (−) (0.2 nM, 2 nM, 20 nM, 200 nM, 2000 nM)
2 nMBucher et al.
[164]
TBTlipid accumulation ↑ (5 nM, 10 nM, 50 nM)
mRNA level of SREBF1, FASN ↑ (50 nM)
protein level of RXRα ↓ (50 nM)
5 nM, 10 nM, 50 nMStossi et al.
[202]
Huh-7 cell lineBPAcell viability ↓ (200 µM, 400 µM), (−) (10 µM, 100 µM)
lipid accumulation ↑ (10 µM, 50 µM, 100 µM, 200 µM), (−) (1 µM)
fatty acid uptake ↑ (10 µM, 50 µM, 100 µM), (−) (1 µM)
mRNA level of CD36 ↑ (50 µM, 100 µM), SR-A1, SR-B1 (−) (10 µM, 50 µM, 100 µM)
protein level of CD36 ↑ (50 µM, 100 µM), SR-A1, SR-B1 (−) (10 µM, 50 µM, 100 µM)
intracellular ROS generation ↑ (10 µM, 50 µM, 100 µM, 200 µM), (−) (1 µM)
10 µM, 50 µM,
100 µM, 200 µM,
400 µM
Lee et al.
[206]
HHL-5
cell line
BPAlipid accumulation ↑ (10 nM, 100 nM, 1000 nM, 22.5 µM, 45 µM)
mRNA level of SREBP1c, ACACA, FASN ↑ (22.5 µM), CNR1 ↑ (45 µM)
level of AEA ↑, PEA, OEA ↓ (45 µM)
CB1 activity ↑ (45 µM)
FAAH activity ↓ (45 µM, 90 µM)
probable mechanism of action by endocannabinoid action at CB1 receptors
10 nM, 100 nM,
1000 nM, 22.5 µM,
45 µM, 90 µM
Martella et al.
[210]
L02 cell lineTCSlevel of TG, PG, LPC, LPE ↑ (1 µM, 2.5 µM), DG, PE, PC, SM, Cer ↑ (2.5 µM), PI ↓ (1 µM, 2.5 µM)
ROS generation ↑ (1 µM, 2.5 µM)
MDA content ↑ (1 µM, 2.5 µM)
SOD activity ↑ (2.5 µM), (−) (1 µM)
CAT activity ↑ (1 µM, 2.5 µM)
level of GSH ↓ (2.5 µM), (−) (1 µM), taurine ↓ (1 µM, 2.5 µM)
1 µM, 2.5 µMZhang et al.
[213]
Legend: ↑ increase; ↓ decrease; (−) no observed effects; * concentration (s) at which biological effects were observed.
Table 4. The obesogenic effect of selected EDCs confirmed on pancreatic cellular models.
Table 4. The obesogenic effect of selected EDCs confirmed on pancreatic cellular models.
Cell TypeOrganismIn Vitro ModelEDCsMechanism of ActionConcentration *References
Pancreatic cells/isletsAnimalRat pancreatic isletsBPAacute exposure (60 min)
insulin secretion with glucose (8.3 mM or 16.7 mM) stimulation (−) (0.1 µg/L, 1 µg/L, 10 µg/L, 100 µg/L, 1000 µg/L)
long-term exposure (24 h)
insulin secretion with glucose (16.7 mM) stimulation ↑ (10 µg/L, 100 µg/L)
co-incubation of BPA (10 µg/L) with actinomycin-D (Act-D) significantly suppressed insulin secretion
probable mechanism of action via cytosolic/nuclear estrogen receptors
10 µg/L, 100 µg/LAdachi et al. [216]
NPacute exposure (60 min)
insulin secretion with glucose (8.3 mM or 16.7 mM) stimulation (−) (0.1 µg/L, 1 µg/L, 10 µg/L, 100 µg/L, 1000 µg/L)
long-term exposure (24 h)
insulin secretion with glucose (16.7 mM) stimulation ↑ (0.1 µg/L, 1 µg/L, 10 µg/L, 100 µg/L)
co-incubation of NP (10 µg/L) with Act-D significantly suppressed insulin secretion
probable mechanism of action via cytosolic/nuclear estrogen receptors
0.1 µg/L, 1 µg/L,
10 µg/L, 100 µg/L
Adachi et al. [216]
TBTviability of cells ↓ (0.01 µM, 0.1 µM, 1 µM, 10 µM, 100 µM) ↑ (10 µM)
insulin secretion at both basal (2.8 mM) and stimulatory (16.7 mM) concentrations
ROS generation ↑ (10 µM)
0.01 µM, 0.1 µM,
1 µM, 10 µM,
100 µM
Ghaemmaleki et al. [217]
INS-1
cell line
BPAviability of cells after 48 h exposure ↓ (0.002 µM, 0.02 µM, 0.2 µM, 2 µM)
insulin secretion with basal glucose concentration (5.6 mM) ↑ (0.02 µM), (−) (0.002 µM, 0.2 µM, 2 µM)
insulin secretion with stimulatory glucose concentration (16.7 mM) ↑ (0.002 µM), ↓ (0.2 µM, 2 µM), (−) (0.02 µM)
mRNA level of insulin ↓ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM)
protein level of insulin ↓ (0.02 µM, 0.2 µM, 2 µM)
mRNA level of genes involved in GSIS pathway: GLUT2, GCK ↓ (0.2 µM, 2 µM), (−) (0.002 µM, 0.02 µM; KIR6.2, SUR ↑ (0.002 µM), ↓ (0.02 µM, 0.2 µM, 2 µM)
changes in mitochondrial morphology and mass ↑ (0.02 µM, 0.2 µM, 2 µM)
cellular ATP level ↓ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM)
mitochondrial potential ↓ (0.2 µM, 2 µM), (−) (0.002 µM, 0.02 µM)
mRNA level of genes involved in mitochondrial metabolism and function: UCP2 ↑ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM); ATP6, Citrate synthase ↓ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM); TFAM ↓ (0.2 µM, 2 µM), (−) (0.002 µM, 0.02 µM); OGDH ↓ (0.002 µM, 0.02 µM, 0.2 µM, 2 µM); ND4L (−) (0.002 µM, 0.02 µM, 0.2 µM, 2 µM)
apoptosis ↑ (0.2 µM, 2 µM)
release of cytochrome c from the mitochondria to the cytosol ↑ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM)
protein level of pro-apoptotic protein BAX, APAF1, 17-kDa cleaved form of caspase-9 ↑ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM), 17-kDa form of cleaved caspase-3 ↑ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM), 19-kDa form of cleaved caspase-3 ↑ (0.002 µM, 0.02 µM, 0.2 µM, 2 µM), anti-apoptotic protein Bcl-2 ↓ (0.02 µM, 0.2 µM, 2 µM), (−) (0.002 µM), 40-kDa cleaved form of caspase-9 (−) (0.002 µM, 0.02 µM, 0.2 µM, 2 µM)
0.002 µM, 0.02 µM,
0.2 µM, 2 µM
Lin et al. [219]
INS-1E cell lineBPAcell viability after 48 h exposure ↓ (10 pM, 1 nM, 1 µM), (−) (0.1 pM, 1 pM, 100 pM)
β-cell apoptosis ↑ (0.1 pM, 1 pM, 10 pM, 100 pM, 1 nM, 1 µM)
ERα and ERβ involved in BPA-induced apoptosis
ROS generation ↑ (1 nM, 1 µM)
mRNA level of MAFA, PDX1, INS1, INS2, GLUT2, GCK (−) (0.1 pM, 1 pM, 10 pM, 100 pM, 1 nM, 1 µM)
0.1 pM, 1 pM,
10 pM, 100 pM,
1 nM, 1 µM
Dos Santos et al. [23]
TBTcell viability after 48 h exposure ↓ (20 nM, 50 nM, 100 nM, 200 nM),
(−) (1 nM, 10 nM)
β-cell apoptosis ↑ (10 nM, 20 nM, 50 nM, 100 nM, 200 nM), (−) (1 nM)
PPARγ involved in TBT-induced apoptosis
ROS generation ↑ (20 nM, 200 nM)
mRNA level of MAFA, PDX1 ↓ (200 nM), (−) (1 nM, 10 nM, 20 nM, 50 nM, 100 nM), INS1, INS2, GLUT2, GCK (−) (1 nM, 10 nM, 20 nM, 50 nM, 100 nM, 200 nM)
10 nM, 20 nM,
50 nM, 100 nM,
200 nM
Dos Santos et al. [23]
PFOAcell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM,
10 nM, 100 nM, 1 µM)
β-cell apoptosisthe ↑ (10 µM, 20 µM, 50 µM, 10the 0 µM, 200 µM),
(−) (1 nM, 1 µM)
ROS generation (−) (1 nM, 1 µM)
mRNA level of MAFA, PDX1, INS1, INS2, GLUT2, GCK (−) (1 nM, 1 µM)
10 µM, 20 µM, the 50 µM, 100 µM, 200 µMDos Santos et al. [23]
TPPcell viability after 48 h exposure (−) (10 pM, 100 the pM, 1 nM, 10 nM, 100 nM, 1 µM)
β-cell apoptosis (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNAthe level of MAFA, PDX1, INS1, INS2, GLUT2, GCK (−) (1 nM, 1 µM)
none of the the doses tested resulted in a biological effectDos Santos et al. [23]
TCScell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM,
10 nM, 100 nM, 1 µM)
β-cell apoptosis (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFA, PDX1, INS1, INS2, GLUT2, GCK (−) (1 nM, 1 µM)
none of the doses tested resulted in a biological effectDos Santos et al. [23]
DDE
cell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM,
10 nM, 100 nM, 1 µM)
β-cell apoptosis (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFA, PDX1, INS1, INS2, GLUT2, GCK (−) (1 nM, 1 µM)
none of the doses tested resulted in a biological effectDos Santos et al. [23]
p,p’-DDEprotein expression of vitamin D-binding protein (VDBP) ↑, glucosidase 2 subunit beta precursor ↓
intracellular protein level of proinsulin, insulin monomer ↓, hexameric insulin (−)
mRNA level of INS1, INS2 ↓, VDBP (−)
10 µMPavlíková et al. [220]
p,p’-DDTprotein expression of tubulin beta-5 chain, annexin A4, vitamin D-binding protein (VDBP) ↑, actin, mortalin/GRP75 ↓
intracellular protein level of proinsulin, hexameric insulin, insulin monomer ↓
mRNA level of VDBP ↑, INS1, INS2
10 µMPavlíková et al. [220]
RIN-m5F cell lineTBTviability of cells (−) (0.05 µM, 0.1 µM, 0.2 µM)
GSIS (20 mM glucose) ↑ (0.1 µM, 0.2 µM), (−) (0.05 µM)
insulin secretion ↑ (0.1 µM)
intracellular calcium level ↑ (0.1 µM, 0.2 µM)
protein level of p-PKC, p-ERK1/2 ↑, p-Akt (−) (0.1 µM, 0.2 µM)
level of intracellular ROS ↑ (0.05 µM, 0.1 µM, 0.2 µM)
0.05 µM, 0.1 µM,
0.2 µM
Chen et al.
[106]
viability of cells ↓ (0.5 µM, 1 µM), (−) (0.1 µM, 0.2 µM)
GSIS (after 24 h exposure) ↓ (0.5 µM, 1 µM)
apoptosis ↑ (0.5 µM)
cleavage of PARP ↑ (0.5 µM, 1 µM)
level of p-JNK, p-ERK1/2 ↑, p-38 (−) (0.5 µM, 1 µM)
intracellular ROS generation ↑ (0.2 µM, 0.5 µM, 1 µM)
caspase-3 activity ↑ (0.5 µM)
0.2 µM, 0.5 µM, 1 µMHuang et al.
[221]
PFOAviability of cells ↓ (100 µM, 200 µM, 300 µM, 500 µM), (−) (1 µM)
apoptosis ↑ (100 µM, 300 µM, 500 µM), (−) (1 µM)
ROS generation ↑ (200 µM, 300 µM, 400 µM, 500 µM), (−) (1 µM, 10 µM, 50 µM, 100 µM)
mitochondrial superoxide accumulation ↑ (200 µM, 300 µM, 500 µM), (−) (1 µM, 10 µM, 50 µM, 100 µM)
NO (nitric oxide) production ↑ (50 µM, 100 µM, 200 µM, 300 µM, 400 µM, 500 µM), (−) (1 µM, 10 µM)
cytosolic level of TNFα ↑ (100 µM, 150 µM, 200 µM, 300 µM, 500 µM), (−) (10 µM), Il1β ↑ (200 µM, 300 µM, 500 µM), (−) (10 µM, 100 µM, 150 µM)
MMP collapse ↑ (300 µM, 500 µM), (−) (1 µM, 10 µM, 50 µM, 100 µM, 200 µM)
intracellular ATP level ↑ (10 µM, 50 µM), ↓ (200 µM, 300 µM, 500 µM), (−) (1 µM, 100 µM)
cytochrome c release ↑ (200 µM, 300 µM, 500 µM), (−) (10 µM, 100 µM)
cardiolipin peroxidation ↑ (200 µM, 300 µM, 500 µM), (−) (10 µM, 100 µM)
10 µM, 50 µM, 100 µM, 150 µM, 200 µM, 300 µM, 400 µM, 500 µMSuh et al.
[222]
Primary β-cells from wild type mouseBPAβ-cell apoptosis ↑ (1 nM, 1 µM)1 nM, 1 µMDos Santos et al. [23]
insulin secretion in the presence of 8 mM glucose ↑
KATP channel activity ↓
frequency of [Ca2+]i oscillations ↑
1 nMSoriano et al.
[223]
viability of cells (after 48 h exposure) ↓ (0.001 µM, 1 µM, 100 µM)
number of apoptotic cells (after 48 h exposure) ↑ (0.001 µM, 1 µM, 100 µM)
mRNA level of genes encoding components of respiratory chain complexes: NDUFS4, UQCRB, genes involved in ATP production and/or in insulin exocytosis process: VAPA, ATP1B1, ATP6V1F, genes involved in detoxification: SOD2, GPX3, ZFAND2A, anti-apoptotic gene BCL-2 ↓ (0.001 µM), pro-apoptotic gene Bax ↑ (0.001 µM)
ROS level ↑ (0.001 µM)
MMP ↓ (0.001 µM)
GSIS (16 mM glucose) ↓ (0.001 µM, 100 µM)
probable mechanism of action via activation of NF-kB pathway
0.001 µM, 1 µM, 100 µMCarchia et al.
[105]
BPSinsulin secretion in response to 16.7 mM concentration (during 1 h treatment of BPS) ↑ (1 nM, 1 µM)
insulin secretion in response to 8.3 mM concentration (during 48 h treatment of BPS) ↑ (1 nM, 1 µM)
insulin secretion in response to 16.7 mM concentration (during 48 h treatment of BPS) ↑ (1 nM), (−) (1 µM)
KATP channel activity ↓ (1 nM)
mRNA level of (after 48 h treatment of BPS) CACNA1E, KCNMa1, SCN9A ↓ (1 nM), (−) (100 nM, 1 µM), KCNIP ↑ (1 nM), (−) (100 nM, 1 µM)
Ca2+ currents ↓ (1 nM), (−) (100 nM, 1 µM)
1 nM, 1 µMMarroqui et al.
[224]
BPFinsulin secretion in response to 16.7 mM concentration (during 1 h treatment of BPF) ↑ (1 nM, 1 µM)
insulin secretion in response to 16.7 mM concentration (during 48 h treatment of BPF) ↑ (1 µM)
KATP channel activity ↓ (10 nM), (−) (1 nM)
mRNA level of (after 48 h treatment of BPF) CACNA1E ↓ (100 nM, 1 µM), (−) (1 nM), KCNMA1, SCN9A, KCNIP1 ↓ (1 µM), (−) (1 nM, 100 nM)
Ca2+ currents ↓ (1 µM), (−) (1 nM, 100 nM)
1 nM, 10 nM, 100 nM, 1 µMMarroqui et al.
[224]
TBTβ-cell apoptosis ↑ (20 nM, 200 nM)20 nM, 200 nMDos Santos et al. [23]
GSIS (20 mM glucose) ↑
ROS generation ↑
0.1 µM, 0.2 µMChen et al.
[106]
PFOAβ-cell apoptosis (−) (1 nM, 1 µM)none of the doses tested resulted in a biological effectDos Santos et al. [23]
TPPβ-cell apoptosis (−) (1 nM, 1 µM)none of the doses tested resulted in a biological effectDos Santos et al. [23]
TCSβ-cell apoptosis (−) (1 nM, 1 µM)none of the doses tested resulted in a biological effectDos Santos et al. [23]
DDEβ-cell apoptosis (−) (1 nM, 1 µM)none of the doses tested resulted in a biological effectDos Santos et al. [23]
Primary β-cells from ERβ-/- mouse (estrogen receptor β (ERβ) knockout (BERKO) mouse)BPAinsulin secretion in the presence of 8 mM glucose (−)
KATP channel activity (−)
frequency of [Ca2+]i oscillations ↓
1 nMSoriano et al. [223]
BPSKATP channel activity (−) (1 nM)
Ca2+ currents (−) (1 nM, 100 nM, 1 µM)
mRNA level of (after 48 h treatment of BPS) CACNA1E, KCNMA1, SCN9A (−) (1 nM)
none of the doses tested resulted in a biological effectMarroqui et al.
[224]
BPFKATP channel activity (−) (1 nM, 10 nM)
Ca2+ currents (−) (1 nM, 100 nM, 1 µM)
mRNA level of (after 48 h treatment of BPF) CACNA1E ↓, KCNMA1, SCN9A (−) (1 µM)
1 µMMarroqui et al.
[224]
MIN-6 cell lineBPAviability of cells (after 24 h exposure) ↓ (100 pM, 1 nM, 10 nM, 100 nM, 1 µM), (−) (10 µM)
mRNA level of PDX1 ↑ (100 pM, 10 nM, 1 µM), (−) (1 nM, 100 nM, 10 µM); HNF4α ↑ (100 pM, 10 nM, 1 µM, 10 µM), (−) (1 nM, 100 nM); KIR6.2 ↑ (10 nM), (−) (100 pM, 1 nM, 100 nM, 1 µM, 10 µM); MAFA ↑ (100 nM), (−) (100 pM, 1 nM, 10 nM, 1 µM, 10 µM); INS, SUR1, GLUT2, GCK (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
GSIS (20 mM glucose) ↑ (100 nM), (−) (100 pM, 1 nM, 10 nM, 1 µM, 10 µM)
insulin secretion in response to low glucose concentration (1.67 mM) ↑ (100 nM, 10 µM), (−) (100 pM, 1 nM, 10 nM, 1 µM)
insulin content ↑ (10 µM), (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µMAl-Abdulla et al. [229]
BPSviability of cells (after 24 h exposure) ↓ (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
mRNA level of MAFA ↑ (10 nM, 100 nM), (−) (100 pM, 1 nM, 1 µM, 10 µM); KIR6.2 ↑ (10 nM, 100 nM, 10 µM), (−) (100 pM, 1 nM, 1 µM); HNF4α ↓ (100 pM), (−) (1 nM, 10 nM, 100 nM, 1 µM, 10 µM); GlUT2 ↓ (1 nM, 10 nM, 100 nM, 1 µM), (−) (100 pM, 10 µM), INS, PDX1, SUR1, GCK (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
GSIS (20 mM) ↓ (100 pM, 10 nM, 1 µM, 10 µM), (−) (1 nM, 100 nM)
insulin secretion in response to low glucose concentration (1.67 mM) (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
insulin content ↓ (10 nM, 100 nM, 1 µM, 10 µM), (−) (100 pM, 1 nM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µMAl-Abdulla et al. [229]
DEHPviability of cells (after 24 h exposure) ↓ (1 µM), (−) (100 pM, 1 nM, 10 nM, 100 nM, 10 µM)
mRNA level of SUR1, GLUT2 ↑ (10 µM), (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM); INS, PDX1, HNF4α, MAFA, KIR6.2, GCK (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
GSIS (20 mM glucose) ↓ (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
insulin secretion in response to low glucose concentration (1.67 mM) (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
insulin content ↓ (1 µM), (−) (100 pM, 1 nM, 10 nM, 100 nM, 10 µM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µMAl-Abdulla et al. [229]
PFOSviability of cells (after 24 h exposure) ↓ (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
mRNA level of INS ↑ (1 nM), (−) (100 pM, 10 nM, 100 nM, 1 µM, 10 µM), HNF4α ↑ (10 nM), (−) (100 pM, 1 nM, 100 nM, 1 µM, 10 µM), MAFA ↓ (1 µM), (−) (100 pM, 1 nM, 10 nM, 100 nM, 10 µM), GLUT2 ↓ (10 nM, 100 nM, 1 µM, 10 µM), (−) (100 pM, 1 nM), PDX1, KIR6.2, SUR1, GCK (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
GSIS (20 mM) ↓ (100 pM, 100 nM, 10 µM), (−) (1 nM, 10 nM, 1 µM)
insulin secretion in response to low glucose concentration (1.67 mM) (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
insulin content (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µMAl-Abdulla et al. [229]
CdCl2viability of cells (after 24 h exposure) ↓ (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
mRNA level of INS ↑ (100 pM, 100 nM), (−) (1 nM, 10 nM); MAFA ↑ (100 nM), (−) (100 pM, 1 nM, 10 nM); PDX1, HNF4α, KIR6.2, SUR1, GLUT2, GCK (−) (100 pM, 1 nM, 10 nM, 100 nM)
GSIS (20 mM glucose) (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
insulin secretion in response to low glucose concentration (1.67 mM) ↓ (100 pM), (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin content (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µMAl-Abdulla et al. [229]
DDEviability of cells (after 24 h exposure) ↓ (10 nM), (−) (100 pM, 1 nM, 100 nM, 1 µM, 10 µM)
mRNA level of SUR1 ↑ (100 pM), (−) (1 nM, 10 nM, 100 nM, 1 µM, 10 µM); INS ↓ (1 nM, 1 µM, 10 µM), (−) (100 pM, 10 nM, 100 nM); MAFA ↓ (1 nM), (−) (1 nM, 10 nM, 100 nM, 1 µM, 10 µM); PDX1; HNF4α, KIR6.2, GLUT2, GCK (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
GSIS (20 mM) (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
insulin secretion in response to low glucose concentration (1.67 mM) (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
insulin content (−) (100 pM, 1 nM, 10 nM, 100 nM, 1 µM, 10 µM)
100 pM, 1 nM, 10 nM, 1 µM, 10 µMAl-Abdulla et al. [229]
β-TC-6 cell linePFOSGSIS (1.4 mM glucose) ↑ (50 µM, 100 µM)
insulin secretion in the absence of glucose ↑ (50 µM)
intracellular calcium level ↑ (5 µM, 10 µM, 50 µM, 100 µM)
probable mechanism of action via GPR40 activation
5 µM, 10 µM, 50 µM, 100 µMQin et al. [231]
p,p’-DDEbasal insulin secretion ↑ (10 µM)
GSIS (5 mM) ↑ (10 µM)
ROS level (−) (10 µM)
intracellular protein level of PDX1 (−) (1 µM, 10 µM, 100 µM)
PC enzyme activity ↑ (10 µM), (−) (100 µM)
10 µMWard et al. [232]
HumanPancreatic isletsBPAinsulin secretion in the presence of 8 mM glucose ↑
KATP channel activity ↓
1 nMSoriano et al. [223]
TBTGSIS (20 mM glucose) ↑0.1 µMChen et al. [106]
EndoC-βH1 cell lineBPAcell viability after 48 h exposure ↓ (1 pM, 10 pM, 100 pM, 1 nM, 1 µM), (−) (0.1 pM)
β-cell apoptosis ↑ (1 pM, 10 pM, 100 pM, 1 nM, 1 µM), (−) (0.1 pM)
caspase 3/7 activity ↑ (10 nM, 1 µM)
Erα and Erβ involved in BPA-induced apoptosis
ROS generation ↑ (1 nM, 1 µM)
GSIS (20 mM glucose) (−) (0.1 pM, 1 pM, 10 pM, 100 pM, 1 nM, 1 µM)
insulin content (−) (0.1 pM, 1 pM, 10 pM, 100 pM, 1 nM, 1 µM)
mRNA level of MAFA, PDX1, INS, GLUT2, GCK (−) (0.1 pM, 1 pM, 10 pM, 100 pM, 1 nM, 1 µM)
1 pM, 10 pM, 100 pM, 1 nM, 10 nM, 1 µMDos Santos et al. [23]
cell viability after 72 h exposure (−) (1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFB ↑ (10 nM), (−) (1 nM, 100 nM), SNAP25 ↑ (1 nM), (−) (10 nM, 100 nM), INS, PDX1, HNF4α, MAFA, KIR6.2, SUR1, GLUT1, GCK (−) (1 nM, 10 nM, 100 nM, 1 µM)
GSIS (20 mM glucose) ↑ (10 nM, 100 nM, 1 µM), (−) (1 nM)
basal insulin secretion ↑ (1 µM), (−) (1 nM, 10 nM, 100 nM)
insulin content ↑ (1 nM, 10 nM, 100 nM, 1 µM)
1 nM, 10 nM, 100 nM, 1 µMAl-Abdulla et al. [229]
BPScell viability after 48 h exposure ↓ (1 µM), (−) (1 nM, 10 nM, 100 nM)
mRNA level of HNF4α ↑ (1 nM), (−) (10 nM, 100 nM), MAFA ↓ (100 nM), (−) (1 nM, 10 nM), MAFB, GLUT1 ↓ (10 nM, 100 nM), (−) (1 nM), KIR6.2 ↓ (10 nM), (−) (1 nM, 100 nM), SNAP25 ↓ (1 nM), (−) (10 nM, 100 nM), INS, PDX1, SUR1, GCK (−) (1 nM, 10 nM, 100 nM)
GSIS (20 mM) ↓ (1 nM, 1 µM), (−) (10 nM, 100 nM)
basal insulin secretion (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin content (−) (1 nM, 10 nM, 100 nM, 1 µM)
1 nM, 10 nM, 100 nM, 1 µMAl-Abdulla et al. [229]
BPFcell viability after 72 h exposure ↓ (1 µM), (−) (1 nM, 10 nM, 100 nM)
GSIS (20 mM glucose) (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin secretion in response to low glucose concentration (2.8 mM) (−) (1 nM, 10 nM, 100 nM, 1 µM)
1 µMAl-Abdulla et al. [229]
DEHPcell viability after 7 days of exposure ↓ (1 nM, 10 nM, 100 nM), (−) (1 µM)
mRNA level of INS, PDX1, HNF4α, MAFA, MAFB, KIR6.2, SUR1, SNAP25, GLUT1, GCK (−) (1 nM, 10 nM, 100 nM)
GSIS (20 mM) ↑ (1 µM), ↓ (1 nM, 10 nM, 100 nM)
insulin secretion in response to low glucose concentration (2.8 mM) (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin content ↑ (1 µM), ↓ (1 nM, 10 nM, 100 nM)
1 nM, 10 nM, 100 nM, 1 µMAl-Abdulla et al. [229]
TBTcell viability after 48 h exposure ↓ (20 nM, 50 nM, 100 nM, 200 nM), (−) (1 nM, 10 nM)
β-cell apoptosis ↑ (1 nM, 20 nM, 50 nM, 100 nM, 200 nM), (−) (10 nM)
caspase 3/7 activity ↑ (20 nM, 200 nM)
PPARγ involved in TBT-induced apoptosis
ROS generation ↑ (20 nM, 200 nM)
GSIS ↑ (20 mM glucose), (−) (1 nM, 10 nM, 20 nM, 50 nM, 100 nM)
insulin content ↑ (20 nM), ↓ (200 nM), (−) (1 nM, 10 nM, 50 nM, 100 nM)
mRNA level of GLUT2 ↑ (200 nM), (−) (1 nM, 10 nM, 20 nM, 50 nM, 100 nM), MAFA ↓ (10 nM, 20 nM, 50 nM, 100 nM, 200 nM), (−) (1 nM), PDX1, INS, GCK (−) (1 nM, 10 nM, 20 nM, 50 nM, 100 nM, 200 nM)
1 nM, 20 nM, 50 nM, 100 nM, 200 nMDos Santos et al. [23]
PFOAcell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
β-cell apoptosis ↑ (20 µM, 50 µM, 100 µM, 200 µM), (−) (1 nM, 1 µM)
caspase 3/7 activity (−) (1 µM)
ROS generation (−) (1 nM, 1 µM)
GSIS (20 mM glucose) ↓ (10 pM, 1 nM, 10 nM, 100 nM), (−) (100 pM, 1 µM)
insulin secretion at low glucose concentration (0 mM) ↓ (1 nM), (−) (10 pM, 100 pM, 10 nM, 100 nM, 1 µM)
insulin content (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFA, PDX1, INS, GLUT2, GCK (−) (1 nM, 1 µM)
10 pM, 1 nM, 10 nM, 100 nM, 20 µM, 50 µM, 100 µM, 200 µMDos Santos et al. [23]
PFOScell viability after 72 h exposure (−) (1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of INS, PDX1, HNF4α, MAFA, MAFB, KIR6.2, SUR1, SNAP25, GLUT1, GCK (−) (10 nM, 100 nM)
GSIS (20 mM glucose) ↑ (10 nM, 100 nM), (−) (1 nM, 1 µM)
insulin secretion in response to low glucose concentration (2.8 mM) ↑ (10 nM, 1 µM), (−) (1 nM, 100 nM)
insulin content ↑ (1 nM, 10 nM, 1 µM), (−) (100 nM)
1 nM, 10 nM, 100 nM, 1 µMAl-Abdulla et al. [229]
TPPcell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
β-cell apoptosis (−) (1 nM, 1 µM)
caspase 3/7 activity (−) (1 µM)
GSIS (20 mM glucose) ↑ (1 µM), (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM)
insulin secretion at low glucose concentration (0 mM) ↑ (100 pM)
insulin content (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFA, PDX1, INS, GLUT2, GCK (−) (1 nM, 1 µM)
100 pM, 1 µMDos Santos et al. [23]
TCScell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
β-cell apoptosis (−) (1 nM, 1 µM)
caspase 3/7 activity (−) (1 µM)
GSIS (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
insulin content (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFA, PDX1, INS, GLUT2, GCK (−) (1 nM, 1 µM)
none of the doses tested resulted in a biological effectDos Santos et al. [23]
CdCl2cell viability after 72 h exposure ↓ (1 nM, 10 nM, 100 nM), (−) (1 µM)
mRNA level of HNF4α, SNAP25 ↓ (100 nM), (−) (10 nM), INS, PDX1, MAFA, MAFB, KIR6.2, SUR1, GLUT1, GCK (−) (10 nM, 100 nM)
GSIS (20 mM glucose) ↑ (10 nM), (−) (1 nM, 100 nM, 1 µM)
insulin secretion in response to low glucose concentration (2.8 mM) (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin content (−) (1 nM, 10 nM, 100 nM, 1 µM)
1 nM, 10 nM, 100 nMAl-Abdulla et al. [229]
DDEcell viability after 48 h exposure (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
β-cell apoptosis (−) (1 nM, 1 µM)
caspase 3/7 activity (−) (1 µM)
GSIS (20 mM glucose) ↑ (10 nM, 1 µM), (−) (10 pM, 100 pM, 1 nM, 100 nM)
insulin content (−) (10 pM, 100 pM, 1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of MAFA, PDX1, INS, GLUT2, GCK (−) (1 nM, 1 µM)
10 nM, 1 µMDos Santos et al. [23]
cell viability after 7 days of exposure ↓ (1 nM, 10 nM, 100 nM, 1 µM)
mRNA level of INS, PDX1, HNF4α, MAFA, MAFB, KIR6.2, SUR1, SNAP25, GLUT1, GCK (−) (1 nM, 10 nM, 100 nM)
GSIS (20 mM glucose) (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin secretion in response to low glucose concentration (2.8 mM) (−) (1 nM, 10 nM, 100 nM, 1 µM)
insulin content (−) (1 nM, 10 nM, 100 nM, 1 µM)
1 nM, 10 nM, 100 nM, 1 µMAl-Abdulla et al. [229]
NES2Y cell linep,p’-DDTviability of cells after 24 h and 48 h exposure ↓ (100 µM), (−) (0.1 nM, 1 nM, 10 nM, 0.1 µM, 1 µM, 10 µM)
protein expression of cytokeratin 8, cytokeratin 18, alpha-enolase, actin ↓ (10 µM)
10 µM, 100 µMPavlikova et al. [234]
viability of cells after 24 h exposure ↓ (150 µM, 175 µM, 200 µM), (−) (100 µM, 125 µM)
protein expression of cleaved caspase -6, -7, -8, -9, cleaved PARP, CHOP, GRP78 (BiP), GRP75 (mortalin), NDRG1, EFHD2 ↑, ECHM, vimentin, HSP27, IDH3A, K2C8, HNRPF, BIEA, EF2, EZRI, FRIL, G3BP1, HNRH1, NDUS3, NDUS1, PBDC1, PCNA, TCPA ↓ (150 µM)
150 µM, 175 µM, 200 µMPavlikova et al. [107]
p,p’-DDEviability of cells after 24 h and 48 h exposure ↓ (100 µM), (−) (0.1 nM, 1 nM, 10 nM, 0.1 µM, 1 µM, 10 µM)
protein expression of cytokeratin 18, HNRH1 ↓ (10 µM)
10 µM, 100 µMPavlikova et al. [234]
PANC-1 cell lineBPAmRNA level of PCSK1 ↓, insulin secretion ↓10 nMMenale et al.
[124]
Legend: ↑ increase; ↓ decrease; (−) no observed effects; * concentration(s) at which biological effects were observed.
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

Kowalczyk, M.; Piwowarski, J.P.; Wardaszka, A.; Średnicka, P.; Wójcicki, M.; Juszczuk-Kubiak, E. Application of In Vitro Models for Studying the Mechanisms Underlying the Obesogenic Action of Endocrine-Disrupting Chemicals (EDCs) as Food Contaminants—A Review. Int. J. Mol. Sci. 2023, 24, 1083. https://doi.org/10.3390/ijms24021083

AMA Style

Kowalczyk M, Piwowarski JP, Wardaszka A, Średnicka P, Wójcicki M, Juszczuk-Kubiak E. Application of In Vitro Models for Studying the Mechanisms Underlying the Obesogenic Action of Endocrine-Disrupting Chemicals (EDCs) as Food Contaminants—A Review. International Journal of Molecular Sciences. 2023; 24(2):1083. https://doi.org/10.3390/ijms24021083

Chicago/Turabian Style

Kowalczyk, Monika, Jakub P. Piwowarski, Artur Wardaszka, Paulina Średnicka, Michał Wójcicki, and Edyta Juszczuk-Kubiak. 2023. "Application of In Vitro Models for Studying the Mechanisms Underlying the Obesogenic Action of Endocrine-Disrupting Chemicals (EDCs) as Food Contaminants—A Review" International Journal of Molecular Sciences 24, no. 2: 1083. https://doi.org/10.3390/ijms24021083

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