You are on page 1of 13

Environmental Toxicology and Pharmacology 77 (2020) 103373

Contents lists available at ScienceDirect

Environmental Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/etap

Review or Mini-review

Bisphenol A-induced metabolic disorders: From exposure to mechanism of T


action
Muhammad Sajid Hamid Akasha,*, Shakila Sabira, Kanwal Rehmanb,*
a
Department of Pharmaceutical Chemistry, Government College University Faisalabad, Pakistan
b
Department of Pharmacy, University Agriculture, Faisalabad, Pakistan

A R T I C LE I N FO A B S T R A C T

Keywords: Bisphenol A (BPA) is considered as ubiquitous xenooestrogen and an endocrine disrupting chemical which has
Endocrine disrupting chemicals deleterious effects on endocrine functions. Human populations are continuously exposed to BPA as it is abundant
Metabolic programming in daily life. It has been found to be associated with wide range of metabolic disorders notably type 2 diabetes
Epigenetic modifications mellitus (DM). Numerous epidemiological studies have been conducted to find its role in development of DM.
Experimental studies have found that BPA exposure is associated with pathogenesis of DM and also considered as
a risk factor for gestational diabetes. Being a lipophilic compound, BPA is preferably accumulated in adipose
tissues where it alters the production of adipokines that play important roles in insulin resistance. BPA induces
apoptosis by caspase activation after mitochondrial damage and it impairs insulin signaling pathways by altering
associated ion channel activity especially potassium channels. Perinatal exposure of BPA makes offspring more
susceptible to develop DM in early years. Epigenetic modifications are the key mechanisms for BPA-induced
metabolic re-programming, where BPA alters the expression of DNA methyltransferases involved in methylation
of various genes. In this way, DNA methyltransferase controls the expression of numerous genes including genes
important for insulin secretion and signaling. Furthermore, BPA induces histone modifications and alters miRNA
expression. In this article, we have briefly described the sources of BPA exposure to human being and sum-
marized the evidence from epidemiological studies linking DM with BPA exposure. Additionally, we have also
highlighted the potential molecular pathways for BPA-induced DM.

1. Introduction contaminants which are known as endocrine disrupting chemicals


(EDCs) play a crucial role in pathogenesis of T2DM (Zheng et al., 2018).
Type 2 diabetes mellitus (T2DM) has been considered as a complex EDCs are defined as a chemical or mixture of chemicals that have the
metabolic disorder characterized by impaired metabolism of carbohy- potential to disrupt endocrine functions (Zoeller et al., 2012). They are
drates, lipids and proteins (Akash et al., 2013; Rehman and Akash, comprised of heterogeneous range of compounds that are extensively
2017). Hyperglycemia and dyslipidemia are collectively involved to used in daily consumed products, such as personal care products, food
provoke the multifactorial transcriptional, metabolic and enzymatic packaging, pesticides, building materials, and many others. Exposure to
pathways that once initiated, induce the generation of oxidative stress EDCs occurs through different routes such as oral, dermal or inhalation
and inflammatory responses (Akash et al., 2013; Donath and Shoelson, (Gore et al., 2015). It is well established EDCs may interrupt hormone
2011; Prasad et al., 2009; Rehman and Akash, 2017; Rehman et al., related signaling pathways by binding hormone receptors and mod-
2017). Chronic exposure of inflammatory response along with oxidative ifyiing either the production, metabolism or transport of hormones
stress may lead to the development of several causative factors that are (Casals-Casas and Desvergne, 2011; Gore et al., 2015). It has been re-
responsible for paghogenesis of T2DM. Among the causative factors, the vealed that EDCs are responsible for interrupting the key metabolic
most important and crucial factors are insulin resistance in peripheral processes that may lead to disruption of energy and/or glucose home-
tissues, impaired insulin secretion β-cells of pancreatic islets, abnormal ostasis and impaired carbohydrate, protein, and lipid metabolism
hepatic glucose production and decreased glucose uptake (Akash et al., (Alonso-Magdalena et al., 2011; Heindel et al., 2017; Nadal et al.,
2018; Donath and Shoelson, 2011; Rehman et al., 2017). 2017).
Along with the genetic background, a number of environmental Bisphenol A (BPA) is one of the most familiar examples of EDCs. It


Corresponding authors.
E-mail addresses: sajidakash@gcuf.edu.pk (M.S.H. Akash), kanwal.akash@uaf.edu.pk (K. Rehman).

https://doi.org/10.1016/j.etap.2020.103373
Received 16 December 2019; Received in revised form 10 March 2020; Accepted 12 March 2020
1382-6689/ © 2020 Elsevier B.V. All rights reserved.
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

was first synthesized by Dianin in 1891 but as early as the 1930s, it was to daily intake of BPA in human beings (Geens et al., 2012b; Valentino
reported that BPA has oestrogenic properties (Dodds and Lawson, et al., 2016). Besides these dietary sources, BPA may also be found in
1936). It is primarily a monomer that is utilized as a basic component various fresh foods, such as meat, eggs, and milk (Van Landuyt et al.,
for manufacturing of polycarbonate and plastics. It is found in coating 2011). Additionally, BPA has also been detected in food products that
materials of cans and jar caps that are used for food preservation are packed in cardboard boxes. Foodstuff canning processes such as
(Oldring et al., 2014), as well as in dental materials. BPA-based coatings sterilization are the major sourse of the leaching of BPA into food
are used to avoid the direct contact of metal with food and for thermal content from the can walls (Cooper et al., 2011). Heating temperature is
stability as well as to enhance the mechanical strength of jars for food more important, compared with heating duration, in terms of its impact
packing (Cao et al., 2010). This coating is usually comprised of epoxy on BPA migration from the can lining to the food. There is a maximum
resins. Nearly, 9% of total BPA produced is annually utilized for the leakage of BPA when products are heated for 90 min at 121 °C (Kang
manufacturing of can lining material (Ashcroft and Rorsman, 2013). and Kondo, 2003). The process of sterilization causes the leakage of
BPA is commonly used as a color developer in manufacturing of thermal 80–100% of unconjugated BPA and it may also depend upon the con-
printing paper that is widely used for payment and cash registers. ditions as well as food contents nature (Goodson et al., 2004). The
BPA has received special attention from researchers as a ubiquitous lower the pH and the greater the fat content, the more BPA migration
chemical since it was first introduced in plastic manufacturing in- will occur (Munguia-Lopez et al., 2002). Contamination may also
dustries in 1950. Currently, it is among the most abundantly produced happen due to the migration of BPA to the food from polycarbonate
chemicals with over 6 billion pounds produced every year (Lee et al., plastic container (reusable containers, drink dispensers and water/baby
2018). As human populations worldwide are routinely exposed to BPA, bottles) in which food is stored or heated by microwaves (Viñas et al.,
it can be detected in most individuals (Calafat et al., 2004; Vandenberg 2010).
et al., 2007). Keeping in view the regulatory issues, in the 1980s, the In thermal papers, BPA is present in free and unbound forms that
United States Environmental Protection Agency (EPA) set the lowest can be easily released (Geens et al., 2012c). Therefore, handling of
adverse effect level of BPA at 50 mg/kg/day for human populations thermal paper is a major source of human BPA exposure, when eating
(EPA, 2010). food with hands that are contaminated with thermal paper contact. For
Alternatively, the European Food Safety Authority (EFSA) declared instance, cashiers routinely handle thermal receipts, have higher urine
no adverse effect level of 5 mg BPA/kg/day for for rats. EFSA has re- BPA level in comparison with general population (Braun et al., 2010).
cently established the tolerable daily intake (TDI) (estimated amount of Thermal paper receipt is the second common source of BPA exposure
BPA in drinking water or food that may be ingested daily over a lifetime after the dietary intake (EFSA Panel on Food Contact Materials and
without considerable health risk) for BPA i.e 50 μg/kg/day (WHO, Aids, 2015).
2009). The TDI and reference dose are greater than the 95th percentile BPA may also be found in dust coming from the laminate flooring,
of BPA intake level (1.5 μg/kg/day) for the adults estimated by World paints, electronic equipment, and adhesives comprising of epoxy resins
Health Organization (WHO). This estimated safe dose is on the basis of (Hanaoka et al., 2002). BPA was present in 95 % of 56 different samples
published data of BPA exposure estimation in various countries and of dust with concentration ranges from 0.8−10 mg/g of dust sample
information available about BPA level in foods (Shelnutt et al., 2013; (Geens et al., 2009). Comparatively higher BPA levels were detected in
Tudurí et al., 2018). dust samples obtained from laboratories and offices, which is com-
According to European Union Commission, the migration of BPA mensurate with their reliance on many electronic devices and furniture.
into food from varnishes or coatings applied to containers should not Exposure among infants and children is linked with the oral or in-
exceed 0.05 mg of BPA per kg of food (mg/kg). Migration of BPA has halation routes (Calafat et al., 2007). Dust contributes only about 5% of
been prohibited from varnishes or coatings applied to containers in- total BPA exposure (Geens et al., 2012c). BPA exposure through in-
tended to use for infants and children (EU, 2018). BPA-induced ecolo- halation route is < 0.4 ng/kg/day in adults and 5.3 ng/kg/day in in-
gical risks has been investigated by the European Union (2008), Canada fants (Loganathan and Kannan, 2011).
(2008), and Japan (2007). The Japanese authories stated that “the Dental materials may comprise of BPA-containing monomers,
current exposure levels of BPA will not pose unacceptable risks to the mostly in the form of BPA-glycidyl methacrylate. BPA is frequently
local populations of aquatic life, particularly fish.” The European Union leached out from dental sealants, fillings and tooth crowns (Drozdz
view was that “although the predicted exposure concentrations were et al., 2011). The maximum BPA concentration was found in patient’s
significantly below its hazard values, there was a need for further in- saliva immediately after dental filling insertion,whilst the levels de-
formation and/or testing on other organisms as freshwater snails”. clined soon afterward. However, leaching of BPA is known to occur
Canada’s position was that “BPA concentrations in water have potential from artificial dental material (Fleisch et al., 2010). After the re-
to cause adverse effects on populations of pelagic organisms in Canada construction of a tooth crown, almost 13−30 mg/day BPA was released
and BPA concentrations in biota have the potential to cause adverse (Van Landuyt et al., 2011). These studies suggest that dental treatment
effects in populations of wildlife in Canada, but that there is a low risk may contribute significantly to BPA exposure. Therefore, patients with
of direct adverse effects to sediment organisms and to avian wildlife multiple dental fillings are at higher risk of BPA exposure (Geens et al.,
species in Canada” (EPA, 2010). 2012a). Lower BPA concentration (0.3 to 0.35 mg) may be leaked from
In view of these positions, the purpose of the present review is to number of medical devices, which are comprised of polysulfone or
summarize the potential sources of BPA exposure and its hazardous polycarbonate plasticizers, for example, contact lenses, probes, in-
effects on health. Moreover, we have also focused on potential mole- travenous cannulas, inhalers, catheters, hemodialysis apparatus or
cular pathways for BPA-induced metabolic disorders. neonatal incubators (Calafat et al., 2008; Geens et al., 2012c; Haishima
et al., 2001).
2. Sources of BPA exposure Another important source of chronic BPA exposure are toys and
other products that are used by young children and infants such as
As described previously, there are multiple routes for human BPA dummies (pacifiers) and teething aids that are usually held in the
exposure and many significant sources of the chemical. These include mouth for many hours on daily basis. It has been reported that saliva
thermal paper (54.000–79.0 ng/cm2), dust (0.8–10 μg/g), meat BPA level was detected from 0.14 to 2.1 mg/l after exposure to rattles
(17–602 ng/g) and meat products. Fish (5–109 ng/g), fruits and vege- and from 0.11 to 14 mg/l saliva for dummies (pacifiers), after 24 h
tables (9–76 ng/g), beverages (1–18 ng/g), dairy products (21–43 ng/ contact with these products (Konieczna et al., 2015). After 1 min ex-
g), dental material (0.013–30 mg), infant formula (0.1–13 ng/g) and posure to teethers and dummies (pacifiers), BPA saliva level was
plastic (0.2–26 ppb) have also been identified as important contributors 5.9 mg/l and 0.3 mg/l respectively (Viñas et al., 2010).

2
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

3. BPA exposure and metabolic programming receptor 1 (Gpr30). On the other hand, Chronic BPA treatment upre-
gulated miR-338 by suppressing glucagon-like peptide 1 receptor
Barker and his colleagues were the first to propose that nutrition (Glp1r). BPA alters Gpr30/Glp1r activation to control miR-338 expres-
provided to children in early-life may be associated with hypertension, sion which in turn controls Pdx1-dependent insulin secretion (Wei
coronary heart disease, and DM (Barker, 1996, 1995; Hales and Barker, et al., 2017). Glucokinase, encoded by Gck, is an important enzyme to
1992). These observations became the foundation of foetal origins of maintain glucose homeostasis. It is involved in the production of gly-
adult disease hypothesis; it stated that events happening during early cogen (Chakera et al., 2018). Hypermethylation of Gck is involved in
developmental stages may enhance the risk for particular diseases in epigenetic pathogenesis of DM (Tang et al., 2014).
adult life (Gluckman and Hanson, 2004). It is well documented that Hepatic hypomethylation and upregulation of DNA methyl-
exposure of particular hazardous environmental agents during devel- transferase 3B have been observed in male progeny of BPA-treated
opmental stages may make a person more prone to develop non-com- rodents. Furthermore, studies have shown that BPA exposure may in-
municable diseases such as DM (Heindel and Vandenberg, 2015). The duce hypermethylation of Gck promoter in the male progeny of rats.
molecular mechanism behind this hypothesis is still controversial. Altered hepatic DNA methylation and Gck downregulation followed by
However, studies have proposed a number of mechanisms working BPA exposure resulted in insulin resistance along with with decreased
during this sensitivity window such as alteration in various gene ex- hepatic glycogen level in the offspring of BPA exposed rats. These
pression, hormone regulation, cell metabolism, or cellular plasticity, findings suggest that oral intake of BPA by female rats during their
etc. These molecular changes have the potential to permanently alter gestational and lactation time period may enhance the risk of DM in
the physiological and metabolic set points for homeostasis and cause their next generation (Ma et al., 2013). In the same study, second
the programming of an individual to develop metabolic diseases on- generation progeny of BPA exposed rats developed glucose intolerance
ward (Heindel et al., 2017). Epigenetic modifications have been sug- owing to insulin resistance in addition to decreased hepatic Gck level.
gested as key mechanisms behind the altered programming made Moreover, the altered methylation Gck gene has been noticed in the
during early developmental stage (Alavian‐Ghavanini and Rüegg, liver of F2 generation. Therefore, it is suggested that dysregulation of
2018). Besides early developmental exposure, it is also well docu- glucose homeostasis due to BPA treatment may be transmitted in sub-
mented that meternal dietry intakes and placental transfer during the sequent generations (Li et al., 2014). Obesity is considered as a major
prenatal life may have great influence on postnatal health (Barker, etiological factor for DM. A number of investigations have been made to
1998; Kim, 2009) which may also include meternal exposure to toxins evaluate the obesogenic effect induced by various EDCs like BPA ex-
like BPA. Epidemiological and experimental studies conducted on ani- posure via oral or parenteral route. BPA has an hyperlipidemic effect
mals and humans have suggested that birth weight may act as a risk through epigenetic modifications as underlying mechanisms (Bhandari
factor for developmental of DM in later life (Gilbert and Epel, 2009). et al., 2013; de Sousa et al., 2018; Liu et al., 2017).
There are various animal studies showing a negative impact of BPA in BPA may alter human lipid profiles and promote obesity, resulting
metabolic developmental programming. Numerous animal studies have in insulin resistance and/or DM (Alonso-Magdalena et al., 2016). It has
been conducted where pregnant females were exposed to BPA and al- been revealed that BPA exposure (0.5 μg of BPA/kg/day) in male mice
tered glucose homeostasis was observed in offspring during postnatal from its birth to 10 months, exhibited significant accumulation of tri-
life (Table 1). glycerides and cholesterol in liver. Moreover, liver cells exposed with
Epigenetics refers to heritable modifications that do not involve an BPA exhibited increased expression level of lipid biosynthesis genes via
altered DNA sequence. Rather, epigenetic modifications such as DNA increased expression levels of Srebf1 and Srebf2. BPA exposed liver cells
methylation, histone modification and expression of non-coding RNA, showed decreased the DNA methylation level of Srebf1 and Srebf2 genes
alter DNA accessibility and chromatin structure, thereby regulating the (Ke et al., 2016). This alteration in hepatic genes leads to the increased
patterns of gene expression (Handy et al., 2011; Loscalzo and Handy, lipid synthesis and accumulation in liver that may be due to the epi-
2014). It has been revealed that human insulin-like growth factor 2 genetic reprogramming of hepatic genes, involving DNA methylation
(Igf2) and insulin genes are found in same genomic region and strong pattern alteration, that influences lipid metabolism (Ke et al., 2016).
association has been found between DM and insulin/Igf2 locus. Over- During a study in which mice were treated with BPA for 3 months,
expression of Igf2 induces β-cell damage and makes them more sus- considerable reduction in hepatic miR-192 was recorded. Additionally,
ceptible to immune attack (Casellas et al., 2015). BPA exposure, during srebf1 gene overexpression was also observed in the hepatocytes. The
early developmental stages, induces DNA hyper-methylation with authors suggested that miR-192 might be an epigenetic modulator of
subsequent over-expression of Igf2 which impairs β-cell function in the srebf1 and thus lipid synthesis. BPA treatment might impair the normal
rodent offspring (Mao et al., 2017). Recently, a study has been con- functioning of miR-192 resulting in the srebf1 gene upregulation (Lin
ducted in order to evaluate the impact of BPA in both F1 and F2 gen- et al., 2017). Indeed, when rats were treated with BPA, their male
erations. This study showed that perinatal BPA exposure may induce offspring showed comperatively higher hepatic lipid accumulation in
Igf2 methylation in F2 male but this alteration was not observed in F2 comparison to female offspring, thus they were more susceptible to
female foetus. Furthermore, this study showed that BPA exposure develop fatty liver disease. It was observed that carnitine palmitoyl-
during pregnancy may induce Igf2 over-expression in a pregnant female transferase 1A (Cpt1a) expression, encodes a important enzyme for fatty
which resulted in glucose intolerance and GDM (Susiarjo et al., 2015). acid metabolism, was considerably reduced in male offspring, particu-
Pancreatic and duodenal homeobox-1(Pdx1) gene encoded tran- larly in the group fed with high-fat diet. In addition to hypermethyla-
scription factor plays a crucial role in transcription regulation. It con- tion of Cpt1a gene, decreased level of histone 3 lysine 4 dimethylation
tributes considerably in production of different types of pancreatic cells, (H3Me2K4) was the key epigenetic dysregulation detected following
β-cell development, and adult β-cell physiology. Altered Pdx1 expres- perinatal BPA exposure. Furthermore, Kmt2c expression, a key histone
sion is connected with hyperglycaemia and suppressed β-cell pro- methyltransferase enzyme that induces gene expression was con-
liferation (Spaeth et al., 2017). Chang et al. has observed in their study siderably decreased. On the basis of these outcomes, it was concluded
that maternal exposure of BPA during gestational period reduces Pdx1 that the chief epigenetic regulatory pathway of Cpt1a gene in BPA ex-
expression in offspring. Histones H3 and H4 deacetylation, demethy- posed rodents could be K4 dimethylation of H3 (Strakovsky et al.,
lation of histone 3 lysine 4 (H3K4) and methylation of histone 3 lysine 9 2015) (Fig. 3). Recently, it has also been found that epigenetic mod-
(H3K9) were found at the promoter of Pdx1. All these modifications ifications of few lipogenic genes followed by BPA treatment during
induced Pdx1 downregulation (Chang et al., 2016). miR-338 acts as developmental phases were evaluated. Female offspring of BPA treated
epigenetic regulatory machinery for Pdx1 gene. Acute BPA treatment mice exhibited hypomethylation of Fasn, Nrf2, and SREBP-1C genes
downregulated miR-338 by activating G-protein-coupled oestrogen with subsequent gene upregulation (Shimpi et al., 2017).

3
M.S.H. Akash, et al.

Table 1
Impact of BPA exposure on metabolic programming.
Animal model Dose/ route Duration Follow up Observations Ref

Pregnant Sheep 0.05, 0.5, 5 mg/kg/d GD30-GD90 21 months after delivery BPA disrupted the insulin sensitivity mediators. BPA elevated oxidative stress level in (Puttabyatappa et al., 2019)
S.c. injection metabolic tissues, dyslipidemia and lipotoxicity in liver and muscles.
NMRI male mice 100 μg/kg/d 4 weeks 13-13.5 weeks of age Elevated fasting blood glucose level, lowered level of insulin and adiponectin and HOMA-β (Veissi et al., 2018)
S.c. injection
Pregnant sheep 0.5 mg/kg/d GD30-GD90 21 months aftr delivery BPA upregulated genes associated with adipogenesis in visceral adipose tissues. (Dou et al., 2019)
S.c. injection
Pregnant OF-1 mice 10 μg/kg/d GD9-GD16 4 months after delivery Body weight gain in offspring, enhanced insulin secretion due to reduced insulin sensitivity, (Alonso-Magdalena et al.,
S.c. injection increased triglycerides, glycerol and leptin levels. Impaired glucose tolerance. 2010b)
Pregnant CD-1 mice 5 μg/kg/d GD9-GD18 18-19 weeks old (offspring) Increase in body weight and liver weight, abdominal adipocyte mass, number and volume, (Angle et al., 2013a)
Oral and in serum leptin and insulin.
Pregnant C57BL/6 10 μg/kg/d and 10 mg/kg/ From 2 weeks prior to 16-21 weeks old (male F1 Dose and sex specific effects, impaired insulin secretion, reduced β cell mass, increased β (Bansal et al., 2017)
d mating until weaning and F2 offspring) cell death, levels of pro-inflammatory cytokines and altered islet DNA methylation.
Oral

4
Pregnant NOD mice 0.1, 1, 10 mg/L GD0-PND21 7-28 weeks old female Increased α and β cells and tissue resident macrophage apoptosis, augmented number of (Bodin et al., 2013)
Drinking water offspring regulatory T cells and active caspase-3 positive cells in pancreatic islets. Increased severity
of insulitis and incidence of DM.
Pregnant OF-1 mice 10 μg/kg/d GD9-GD16 17 & 28 week Increased fasting blood glucose, insulin secretion and NEFA levels. In liver, increased (García-Arevalo et al., 2014)
S.c. inject mRNA expression of PPARγ and increased triglyceride content.
Pregnant OF-1 mice 10 or 100 μg/kg/d GD9-GD16 PND 0, 21, 30 and 120 Increased β cell mass and hyperinsulimenia during early PND but decreased in late PND. (García-Arévalo et al., 2016)
S.c. inject
Pregnant C3H/HeN 50 μg/kg/d Orally/0.1 % GD15-PND21 From weaning to PND170 Impaired glucose tolerance, decreased insulin sensitivity. (Malaisé et al., 2017)
mice ethanol in corn oil
CD1 male mice 5, 50, 500 or 5,000 μg/kg/ 28 days 10 weeks old Increased plasma insulin levels, hepatic mRNA and protein related to lipid biosynthesis and (Marmugi et al., 2012b)
d lipid content.
Diet
C57BL6 male mice 50 μg/kg/d 12 weeks 18 weeks old Glucose intolerance and decreased Akt phosphorylation in skeletal muscle. (Moon et al., 2015)
Drinking water
C57BL6 male mice 50 μg/kg/d 2 weeks 6 and 6.5 months old Diminished glucokinase activity after acute exposure. (Perreault et al., 2013)
Drinking water

PND; Post-natal days.


Environmental Toxicology and Pharmacology 77 (2020) 103373
M.S.H. Akash, et al.

Table 2
Clinical studies reported the association of BPA and DM.
Country Year Study design Population, Sample Technique BPA level Results Ref
Case/ control (Mean)

Saudi Arabia 2018 Case control 54/47 Urine HPLC 3.9* Urinary BPA is associated with increased risk for DM. (Li et al., 2018)
Missouri 2018 Cross over 8 Serum LC-MS 7.3 ng/ml Environmental Protection Agency–estimated “safe” daily dose alters the insulin/C-peptide in (Stahlhut et al., 2018)
response to glucose.
Turkey 2018 Case control 50/50 Urine HPLC 24.13 ng/ml No significant association between urinary BPA levels and T1DM in children aged between 5–18 (İnce et al., 2018)
years.
Denmark 2018 Cross sectional 107 Urine LC–MS 2.55 ng/ml A study on healthy normal-weight children suggests an inverse association between BPA and (Carlsson et al., 2018)
insulin resistance.
China 2018 Nested case 232/232 Serum ELISA 1.3 ng/ml BPA is not associated with a 5-year DM incidence. (Shu et al., 2018)
control
China 2018 Case control 251/251 Urine HPLC 0.503* μg/g Several urinary bisphenols were positively associated with DM. (Duan et al., 2018)
creatinine
Thai 2017 Cross sectional 240 Serum Competitive ELISA 1.1 ng/ml There is an association between BPA exposure and impaired glucose tolerance, but not DM. (Chailurkit et al., 2017)
United states 2008 Cross sectional 1455 Urine HPLC-MS 4.6 ng/ml Higher BPA level was positively associated with DM. (Lang et al., 2008)

5
Canada 2016 Cross sectional 4320 Urine GC-MS 1.21 μg/l* Urinary BPA was positively associated with adverse glucose homeostasis in men. (Tai and Chen, 2016)
Lebanon 2017 Cross sectional 501 Urine HPLC-MS 3.67 μg/l Urinary BPA is linked with metabolic syndrome, obesity and diabetes. (Mouneimne et al., 2017)
United states 2015 Cross sectional 382 Urine GC-MS-MS 3800 ng/l Urinary monochlorinated BPA derivative was significantly associated with DM, whereas the (Zhou et al., 2015)
parent compound (total BPA) was not.
China 2015 Cross sectional 3423 Urine HPLC-MS 0.95 ng/ml No association found between BPA and DM. (Ning et al., 2011)
Thai 2014 Cross sectional 2581 Serum ELISA 0.34 ng/ml* Serum BPA level was not associated with impaired fasting glucose, but positively associated with (Aekplakorn et al., 2015)
DM.
Iran 2014 Case control 239 Urine GC-MS 2.9 μg/l Urinary Bisphenol A levels are found to be associated with DM independent of traditional DM risk (Ahmadkhaniha et al.,
factors. 2014)
United States 2014 Case control 971/970 Urine LC-MS 4.4 μg/l BPA exposures may be associated with the risk of DM among middle-aged, but not older women. (Sun et al., 2014)
Korea 2013 Cross sectional 1210 Urine GC-MS 2.2 ng/ml* High body BPA burden may not be associated with an increased prevalence of DM. (Kim and Park, 2013)
United States 2013 Cross sectional 3516 Urine GC-MS 2.075 ng/ml Higher urinary BPA levels are found to be associated with prediabetes independent of traditional (Sabanayagam et al., 2013)
diabetes risk factors. association was stronger among women and obese subjects.
India 2017 Case control 30/30 Urine ELISA 1225.01 No co-relation was found. (Kumar et al., 2018)
pg/mg creatinine
United States 2011 Cross sectional 4792 Urine GC-MS 3.93 ng/ml Urinary BPA is associated with DM independent of traditional risk factors. (Shankar and Teppala,
2011)

* Geometrical mean.
Environmental Toxicology and Pharmacology 77 (2020) 103373
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

4. BPA exposure and DM: evidences from clinical studies

(Chiu et al., 2017)

(Robledo et al.,
(Bellavia et al.,

(Shapiro et al.,
Numerous clinical studies conducted around the globe have found
(Wang et al., that there is a strong association between BPA exposure and DM in-
2017b) dependent of traditional risk factors (Table 2). (Aekplakorn et al., 2015;

2018)

2015)

2013)
Ref

Chailurkit et al., 2017; Duan et al., 2018). However, few clinical based
studies fail to show correlations between BPA and metabolic disorders
BPA exposure during the second trimester may have adverse effect on blood

Moderately high BPA concentrations were associated with increased glucose


Higher maternal exposure to BPA may be associated with decreased risk of

(Ning et al., 2011; Shu et al., 2018). Data from US National Health and
Nutrition Examination Survey (NHANES) revealed that BPA was de-
tectable in almost 90 % of U.S. population aged ≥6 years old, with
concentration ranging from 0.4−149 μg/L. Adolescents and children
have greater urinary BPA concentration than adults. Important socio-
economic and ethnic differences has also been observed. It has been
noted that there is an inverse correlation between income and urinary
BPA excretion. BPA has also been found in foetal cord blood and breast
milk (Mirmira and Evans-Molina, 2014). Recently, a meta-analysis was
conducted on sixteen clinical based studies by Hwang and his collea-
levels among overweight/obese women

gues. They have concluded that BPA exposure to human showed posi-
glucose levels but not in 1st trimester

tive associations with onset of DM (OR 1.28, 95 % CI 1.14, 1.44)


(Hwang et al., 2018).
GDM and lower birth weight.

Lang et al. investigated the association between DM and urinary


BPA level by examining cross-sectional NHANES data from 2003 to
No association found

No association found

2004. They observed that higher BPA levels were positively associated
with self reported diagnosis of either DM or borderline diabetes (Lang
et al., 2008). Afterward, Melzer et al. analyzed NHANES data from
2003 to 2004 and 2005 to 2006 and observed a considerable associa-
Results

tion between DM and urinary BPA level in pooled data. However, the
association was not significant after analysis by using fully adjusted
models. The diagnosis of DM in clinical based study was self-reported in
1.23 & 1.01 μg/L* for 1st and
1.39 & 1.27 μg/L for 1 and

which DM and borderline DM were taken as a single outcome (Melzer


st

et al., 2010). Shankar and Teppala examined samples from NHANES


participants (adults) between 2003–2008. This study was superior to its
peers as the authors did a reliable assessment of DM cases, which were
2nd trimester

2nd trimester

diagnosed on the basis of standards provided by American Diabetes


1.32 μg/L*
BPA level

0.9 μg/L*

0.8 μg/L*

Association i.e. fasting serum glucose and HbA1c levels. The results of
(Mean)

this study also showed a positive correlation between urinary BPA level
and diagnosis of DM (Teppala et al., 2012). Moreover, Silver and col-
leagues also noted the similar findings. They also investigated pooled
HPLC-MS/MS
Technique

HPLC–MS

NHANES data from 2003 to 2004, 2005 to 2006, 2007 to 2008. In this
HPLC-MS

GC–MS

clinical based research, DM was diagnosed on the basis of HbA1c ≥ 6.5


HPLC

% or through the intake of medications for DM. Notably, when data of


three different time periods was separately analyzed, it was found that
Sample

Urine

Urine

Urine

Urine

Urine

the association was more pronounced during 2003–2004, where con-


centration of urinary BPA was highest as compared to that of other two
time periods (Silver et al., 2011). The same researchers conducted an-
Mean Maternal

other pooled analysis of cross-sectional NHANES data (2003–2008) and


cross examined the correlations between BPA level and metabolic
syndrome. Similary, they obtained a positive association between me-
Clinical studies reported the relationship of BPA and GDM.

≥25
age

tabolic syndrome and higher urinary level of BPA independent of other


31

35

32

32

confounders i.e age, race and diet. In this study, the diagnosis was made
1st and 2nd trimester

on the basis of revised Adult Treatment Panel III guidelines included:


Gestational week

higher waist circumference, glucose intolerance, increased blood pres-


1st trimester

sure, increased triglycerides level, and decreased HDL level (Teppala


24 to 28

et al., 2012). In contrast to the above, LaKind et al. conducted another


38.7

26.6

analysis on all NHANES data obtained during 2003–2010 and observed


no association between cross sectional urinary levels of BPA and DM
Population

diagnosis. DM was diagnosed on the bsis of either doctor diagnosed DM,


fasting glucose level > 126 mg/dl and two hours blood glucose
1274
620

245

350

level > 200 mg/dl after an oral glucose tolerance test (LaKind et al.,
94

* Geometrical mean.

2012). Carwile and Michels investigated cross sectional pooled data


2017

2017

2018

2015

2013
Year

from adult participants in 2003–04 and 2005–06 NHANES cohorts.


Results provided the evidence that participants falling in upper quar-
United states

United states
United state

tiles were more likely to be obese and presented with abdominal obesity
Country

Canada

(Carwile and Michels, 2011). Similar results were obtained from ana-
Table 3

China

lysis of NHANES data from 2003 to 2008, which investigated associa-


tion between obesity (durig childhood and adolescence) and urinary

6
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

BPA levels. Children from lower quartiles of urinary BPA level showed was verified that ERα knockout mice show adaptation of glucose uti-
the lowest rates of obesity (Carwile and Michels, 2011). These results lization by skeletal muscle and extreme hepatic insulin resistance
were confirmed by another group, and later analysis of NHANES pooled (Bryzgalova et al., 2006). In addition, oestrogen can control the process
data from 2003 to 2010 ha s revealed that higher BPA levels was in- of adipose tissue metabolism and body fat distribution through ERα and
dependently correlated with childhood obesity plus abdominal obesity ERβ (Cooke et al., 2001). Oestrogen also regulates the distribution of
(Eng et al., 2013). white adipose tissue (WAT) not only females but also males. It has also
Studies have also reported an increased rate of GDM from the last been confirmed that ERα deficiency will lead to hypertrophy in WAT
few years (Zhu and Zhang, 2016). Up-to some extent, exposure to en- and adipocyte hyperplasia along with insulin resistance and glucose
vironmental chemicals including BPA also contributes to this trend intolerance, but not in brown adipocyte tissue (BAT) (Cooke et al.,
(Table 3). A clinical study conducted in Mexico has revealed that higher 2001). Human data has also underlined the importance of both re-
urinary BPA level in women may increase the frequency of premature ceptors in fat metabolism and obesity. The relative amount of ERα/ERβ
deliveries (Cantonwine et al., 2010). Miao et al. reported low birth is related to obesity, high serum level and synthesis of leptin in omental
weight in new born of BPA-exposed mothers. Similar obervations were adipose tissue of females (Cooke et al., 2001). ERα disturbance in
also seen in infants of BPA-exposed fathers (Miao et al., 2011). It is ventromedial nucleus of the hypothalamus can cause hyperphagia,
evident from the literature that there is an increased risk of altered hyperglycaemia, weight gain, visceral adiposity, and imbalance of en-
glucose metabolism due to BPA exposure in obese pregnant women ergy utilization in female mice (Musatov et al., 2007). Furthermore ERβ
(Bellavia et al., 2018). Chiu et al. refined these findings, observing that has been found to produce an anorectic effect which is started from
the BPA effect on glucose tolerance is trimester specific. They reported central nervous system (CNS) (Liang et al., 2002). Also it has been
that BPA is positively associated with blood glucose level in second confirmed on mice treated with high fat diet that ERβ play main role in
trimester, but they found no association in first and third trimester adiposity regulation (Foryst-Ludwig et al., 2008).
(Chiu et al., 2017). Interestingly, contrast to these studies, Shapiro et al. Oestrogen-related receptors (ERRs) belong to a subfamily of orphan
showed that there is no association between BPA exposure and glucose nuclear receptors that are closely related to ER (ERα and ERβ). There
tolerance disorders during pregnancy (Shapiro et al., 2015). Similarly, are further three types of these ERRs (ERRα, ERRβ, and ERRγ).
Robledo et al. did not support the relationship of BPA and altered However, oestrogen does not show significant affinity to ERRs, but
glucose metabolism during pregnancy. these receptors possess significant homology with ERs, especially in the
Sugiura et al. conducted a study in order to evaluate the risk factors ligand binding and DNA binding domains (Ascenzi et al., 2006). The
working behind miscarriages during 1st trimester of pregnancy. They ERRs have constitutive transcriptional activity which is repressed by
analyzed serum BPA level alongwith other biological parameters. certain chemicals (such as dexamethasone and 4-hydroxytamoxifen).
Furthermore, immunoassays for antinuclear antibodies were also per- BPA shows strongly binding with ERR with a half maximal inhibitory
formed for each partner. The mean serum BPA concentration of female concentration (IC50) value of 13.1 nmol/L, which can preserve ERRγ
with recurrent pregnancy loss was 2.59 ± 5.23 ng/ml. On the other constitutive activity in the presence of 4-hydroxytamoxifen. It is evi-
hand, the mean serum BPA concentration of women in normal control dent from the studies that low-dose BPA effects may be mediated
group was 0.77 ± 0.38 ng/ml. The antinuclear antibodies-positive through these nuclear receptors (Acconcia et al., 2015).
women had considerably greater BPA concentration than the anti-
nuclear antibodies-negative women. The results of this clinical study 6. Mechanism of BPA-induced metabolic disorders
showed that high BPA exposure may be correlated with recurrent
pregnancy loss, especially in antinuclear antibody-positive women 6.1. Effect of BPA on adipocytes
(Sugiura-Ogasawara et al., 2005).
BPA is a lipophilic compound and is preferentially accumulated in
5. BPA and oestrogen receptors adipose tissues (Fernandez et al., 2007). It may promote the differ-
entiation of adipose stem/stromal cells. BPA may also alter the adipo-
The oestrogen receptors ERα and ERβ, play key roles in energy and genic genes expression including DLK, C/EBPα, IFG1, PPARγ, and LPL
glucose homeostasis (Alonso-Magdalena et al., 2008). However ERα is (Ariemma et al., 2016; Ohlstein et al., 2014b). When animals were
considered as the chief regulator. It has also been confirmed by Heine treated with oestrogen receptor (ER) antagonist following administra-
et al. that the rodents show obesity and insulin resistance which were tion of BPA, there was weakened BPA–induced adipogenic effect. These
ERα knockout. ERα has the ability to maintain physiological function of observations showed that BPA produces the adipogenic effect through
glucose metabolism in several tissues like adipose tissue, brain, skeletal an ER-dependent pathway (Ohlstein et al., 2014a). As BPA accumulates
muscles, pancreas and liver (Alonso-Magdalena et al., 2008; Barros in adipocytes, it may bring alterations in cell signaling pathways. BPA
et al., 2006). It has been found that insulin release, synthesis and β-cell has an effect on adipokines secretion. Adiponectin enhances the insulin
maintenance and regulation is depended on ERα. BPA and 17-estradiol sensitivity via adiponectin receptor signaling pathway; abundantly
(E2) activated ERα can enhance the production of insulin by non present in liver and skeletal muscles. Angle et al. observed that adi-
classical pathway of oestrogen activation through ERK1/2 phosphor- pocytes exposed to BPA reduces adiponectin secretion in human and
ylation. It is E2 concentration depended activation of ERα which has a adult mice (Angle et al., 2013b). PPARγ is considered as a major factor
significant role in changes of endocrine pancreas to pregnancy. Yet, stimulating the adiponectin expression. It is evident from the literature
ERα is upregulated by elevated concentrations of E2 or BPA, which that BPA may reduce adiponectin production by antagonizing PPARγ
increases insulin production unnecessarily. Hence it may promote is- (Ben-Jonathan et al., 2009; Volberg et al., 2013). Reduced serum leptin
sues related to T2DM, by negative effects on beta-cell health and insulin has been observed after BPA exposure (Angle et al., 2013b). Literature
resistance in liver and muscle (Nadal et al., 2009). has shown that BPA has the potential to activate Toll-like Receptors
GLUT4 (glucose transporter-4) is believed to be important in insulin (TLR) resulting in activation of JNK and NF-kB signaling pathways with
signaling and is considered as key regulator in peripheral utilization of subsequent up-regulation of some pro-inflammatory cytokines such as
glucose by muscles and adipocytes through insulin stimulation. Here IL-6 and IFN-γ (11). During a cohort study, prenatal BPA exposure
ERα promotes GLUT4 expression particularly in skeletal muscle cell stimulates TLR with subsequently enhanced secretion of TNF-α, IL-10
membrane which has been confirmed through analysis by Barros et al; and IL-6 (Liao et al., 2016). A cross-sectional study, exclusively with
upregulation of GLUT4 occurred in ArKO mice, was unchanged in male adults, has revealed that blood BPA level is positively associated
ERβKO mice and greatly reduced in ERαKO mice (Barros et al., 2006). with raised pro-inflammatory cytokines (TNF-α and IL-6) and they
ERα is the main influence over hepatic insulin sensitization. In fact, it contribute significantly towards insulin resistance development

7
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

(Lin et al., 2013a). Approximately 98 % of total ATP production in β-


cells is generated via mitochondrial oxidative mechanism (MacDonald
et al., 2005). A study conducted on rat offsprings that were perinatally
exposed to BPA at the dose of 50 mg/kg/day, showed structural defects
in mitochondria in β-cells resulting in altered glucose homeostasis (Wei
et al., 2011). Moreover, isolated rat islets were exposied to BPA, it
caused mitochondrial swelling and altered mitochondrial gene expres-
sion. In addition, there was decreased activity of the respiratory en-
zyme, COX IV, present in mitochondria as well as reduced ATP content
(Song et al., 2012). BPA eroded the structural integrity of mitochondria
and reduced mitochondrial mass of INS-1 cells. These structural ab-
normalities were accompanied by altered gene regulation that is pivotal
for proper functioning of mitochondria and energy metabolism (Lin
et al., 2013b; Shirani et al., 2019). For instance, Atp6 expression and
citrate synthase were considerably reduced in cells after the exposure of
BPA (0.020–2.0 mM), suggesting the decreased mitochondrial capacity
to produce ATP following BPA exposure at concentrations greater than
supraphysiological levels. α-ketoglutarate dehydrogenase or ox-
oglutarate dehydrate is an enzyme which plays a pivotal role in TCA
cycle. It is encoded by gene Ogdh. Similarly, mitochondrial uncoupling
protein-2 is a pprotein that is encoded by UCP2 gene. Lin et al. observed
Fig. 1. BPA promotes the adipogenesis and inflammation by activating ER and that downregulation of mRNA expression of Ogdh and upregulation of
TLR respectively. But BPA reduces the adiponectin level by antagonizing mRNA expression of UCP2 after BPA exposure (Lin et al., 2013b). Be-
PPARγ. All these pathological changes contribute to the development of insulin sides the effect on INS-1 cells, BPA also has potential to induce the
resistance. Abbreviations| BPA: Bisphenol A, DM: Diabetes mellitus, ER: apoptosis in other cell types including hepatocytes, germ cells and
Oestrogen receptor, PPARγ: Peroxisome proliferator-activated receptor-alpha, bronchial epithelial cells (George and Rupasinghe, 2018; Kourouma
TLR: Toll-like receptor.
et al., 2015; Wang et al., 2010). BPA may trigger apoptosis in germ cells
via upregulating the mitochondrial apoptotic pathway (Wang et al.,
(Savastano et al., 2015) (Fig. 1). Several genes, including PLIN2, Nie- 2010). Ogdh catalyzes the oxidative decarboxylation of α-ketoglutarate
mann-Pick 2 (NPC2), ACACA, apolipoprotein A1-binding protein to succinyl-CoA in mitochondria, while UCP2 regulates glucose-induced
(APOA1BP), FADS1 and PPAP2A were found to be upregulated through insulin secretion from β-cells of pancreatic islets and mitochondrial
microarray study with the application of BPA in TG and lipid metabo- membrane potential (Bo et al., 2016; Song et al., 2012; Zhang et al.,
lism (Skogsberg et al., 2008). In addition, BPA also improves the pro- 2001). Altered expression of these genes contributes to the BPA-induced
cess of lipid metabolism and accumulation through amplification in disrupted glucose-induced insulin secretion and ATP production. Tfam
expression of LDLR as it’s been clear that Low-density lipoproteins encodes a mitochondrial transcription factor that provides a molecular
(LDL) are known to reduce intracellular lipolysis. It has also been basis for the connection between environmental stimuli and mi-
confirmed by DAVID analysis that upregulated genes in cholesterol and tochondrial biogenesis. BPA treatment may also reduce the expression
sterol metabolic processes play a significant role in IPA. GDF15 and of Tfam (Hock and Kralli, 2009). Mitochondrial dysfunction may po-
MIR503HG genes show modulation in their expression by usage of BPA. tentially affect the viability of the cells and induce the apoptosis. It is
GDF15 being a part of TGFB family is associated with two important well documented that BPA may induce apoptosis in a number of cells
process adipogenesis and obesity, that is identified and produced in and tissues via activation of apoptosis signaling pathways (Gong et al.,
both human and 3T3L1 adipocytes (Ding et al., 2009). Atlas establishes 2017; Kaur et al., 2018; Wang et al., 2017a). Lin et al. have observed
a link between BPA and DEX on GDF15 expression in vice versa order as that BPA exposure may trigger apoptosis in INS-1 cells through acti-
BPA show increment while DEX show decline (Lee et al., 2011). It is vation of mitochondrial apoptotic pathways via involvement of cas-
evident that BPA application produce highest decline in MIR503HG pases and Bcl-2 family (Lin et al., 2013b). Bcl-2 family, including anti-
gene expression. In the differentiation of human preadipocytes apoptotic and pro-apoptotic members, plays a pivotal role in the mi-
MIR503HG is the gene which comprises of microRNA miR-503 se- tochondrial apoptotic pathway (Edlich, 2018).
quence and being able of down-regulation (Ortega et al., 2010). An- The pro-apoptotic protein Bax may enhance the mitochondrial
other important gene which shows upregulation by BPA was SREBF membrane permeability with subsequent release of apoptogenic factors
linked to adipogenesis and has characteristics of transcriptional acti- from the mitochondrial membrane which induces the cell death (Karch
vation in adipocyte differentiation, PPARG activation and lipid meta- et al., 2013). Unlike Bax, Bcl-2 has been reported as an anti-apoptotic
bolism (Kim et al., 1998). BPA-treated mice show SREBP1c and SREBP2 protein, which maintains the integrity of the outer membrane of mi-
gene expression which is upregulated and modulation in hepatocytes tochondria (Gross, 2016). BPA has been found to be involved in Bax
(Marmugi et al., 2012a). It has also been stated that SREBF1gene was expression upregulation and Bcl-2 expression downregulation resulting
expressed in human preadipocytes which was induced through DEX. It in altered permeability of the mitochondrial membrane. Thus mi-
is SREBF1which is identified in fatty acid synthase (FAS), ACACA, LPL tochondrial cytochrome c is released from mitochondria, thus inducing
and LDLR that has the ability to direct lot of protein sequence and their the caspase cascade activation in the cytosol (Hwang et al., 2013).
expression. BPA-treated preadipocytes show SREBF1-cleavage acti- Various caspases have an essential role in transduction of such apop-
vating protein (SCAP) capable of positive upstream regulation. BPA totic signals (Green and Llambi, 2015). Some of the caspases are con-
produce an important mechanism of SCAP activation which can sti- sidered as initiators(8, 9, and 10), whilst some act as effectors (3, 6, and
mulate PPARG, and/or SREBF1 in adipogenesis process (Boucher et al., 7). When INS-1 cells were exposed to BPA, these cells give response to
2014). Apaf-1 (apoptotic protease activating factor-1) activation at dose-de-
pendent manner. Apaf-1 and cytochrome c are combined to make
6.2. BPA-induced mitochondrial damage apoptosome complex. The apoptosome initiates the activation of a
series of caspases that is responsible for the induction of cell death
This can result in the dysfunctioning of β-cells of pancreatic islets (Lakhani et al., 2006). BPA may trigger the pro-caspase-9 activation, it

8
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

is evident from the considerable increase of cleaved caspase-9 (active)


(Barbonetti et al., 2016). Activated or cleaved caspase-9 is remained to
attach along with apoptosome, act like a holoenzyme, in order to sus-
tain the catalytic activity of this complex and further activates caspase-
3. Indeed, this caspase is among the most important effectors that
trigger apoptotic events (Estaquier et al., 2012) (Fig. 2). These findings
have shown that the mitochondrial apoptotic pathway may be involved
in BPA induced INS-1 cell death. The INS-1 cell line is considered very
important model for monitoring the insulin secretion and pancreatic β
cells function. Apoptosis of INS-1 cells reflected the impairment of
carbohydrate metabolism followed by the development of metabolic
disorders (Hectors et al., 2013).

6.3. BPA-induced altered ion channel activity in β-cells

At low concentration of glucose, KATP channels remain open in β-


cells of pancreatic islets. When glucose levels increase, ATP to ADP
ratio raises resulting in KATP channel closure. After that, there is a de-
polarization of membrane causing the opening of voltage-gated Ca2+
channels and an increase in the intracellular calcium level (Ashcroft
and Rorsman, 2013). Calcium triggers the insulin release from β cells
that counteract the raised serum glucose level. (Dyachok et al., 2008).
Thus KATP channel is considered as a key ion channel for insulin release
from pancreatic cells. There is a rapid closure of KATP channels in
pancreatic β cells by estradiol exposure. The maximum inhibition is
achieved within 3−7 min after estradiol exposure and the effect is re-
versible and returns to normal levels after 30 min (Nadal et al., 1998;
Fig. 2. BPA potentiates the pro-apoptotic protein (Bax) and suppresses the anti-
apoptotic protein (Bcl-2) followed by the activation of a series of caspases. Soriano et al., 2009). Thus there is an enhanced insulin secretion.
Activated caspases cause the induction of apoptosis in pancreatic β-cells. During an ex vivo experiment, the enhanced insulinotropic effect was
produced by co-administration of estradiol and glucose and this glucose
provides additional stimulation for insulin secretion (Nadal et al.,
2011). BPA has the potential to induce insulinotropic effect by

Fig. 3. Epigenetic mechanisms for BPA-induced me-


tabolic disorders. BPA is associated with altered DNMT
expression. DNMT controls the regulation of certain
important genes (Gck, Igf2, Srebf2, Nrf2 and Fasn) by
altering methylation pattern having critical role in the
development of metabolic disorder. Abbreviations|
BPA: Bisphenol A, DNMT; DNA methyltransferase.

9
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

modulating KATP channel activity in rodents and human β cells and BPA oestrogenic effect of bisphenol A disrupts pancreatic beta-cell function in vivo and
diminishes KATP channel activity at environmental exposure level induces insulin resistance. Environ. Health Perspect. 114, 106–112.
Alonso-Magdalena, P., Ropero, A.B., Carrera, M.P., Cederroth, C.R., Baquié, M., Gauthier,
(Soriano et al., 2012, 2016). Experimental studies have shown that BPA B.R., Nef, S., Stefani, E., Nadal, A., 2008. Pancreatic insulin content regulation by the
promotes glucose-induced calcium oscillations in β-cells just like es- oestrogen receptor ER alpha. PLoS One 3 e2069–e2069.
tradiol and nanomolar concentration of BPA is sufficient to trigger this Alonso-Magdalena, P., Ropero, A.B., Soriano, S., Quesada, I., Nadal, A., 2010a. Bisphenol-
A: a new diabetogenic factor? Hormones (Athens, Greece) 9, 118–126.
effect (Alonso-Magdalena et al., 2010a; Nadal et al., 2000). Although Alonso-Magdalena, P., Vieira, E., Soriano, S., Menes, L., Burks, D., Quesada, I., Nadal, A.,
the mechanism behind the BPA-induced insulinotropic effect is yet not 2010b. Bisphenol A exposure during pregnancy disrupts glucose homeostasis in
clear, it is most probable that it is through ER similar to estradiol. in vivo mothers and adult male offspring. Environ. Health Perspect. 118, 1243–1250.
Alonso-Magdalena, P., Quesada, I., Nadal, A., 2011. Endocrine disruptors in the etiology
experiments have shown that BPA rapidly reduces the hyperglycemia of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 7, 346.
by promoting the insulin secretion. Like estradiol, administration of Alonso-Magdalena, P., Rivera, F.J., Guerrero-Bosagna, C., 2016. Bisphenol-A and meta-
BPA (10 μg/kg) increases blood insulin level within 30 min. After four bolic diseases: epigenetic, developmental and transgenerational basis. Environ.
Epigenet. 2, dvw022.
day exposure of either estradiol or BPA, mice developed chronic hy-
Angle, B.M., Do, R.P., Ponzi, D., Stahlhut, R.W., Drury, B.E., Nagel, S.C., Welshons, W.V.,
perinsulinemia (Alonso-Magdalena et al., 2006). Similar to estradiol, Besch-Williford, C.L., Palanza, P., Parmigiani, S., 2013a. Metabolic disruption in male
BPA induces the activation of a ubiquitous transcription factor known mice due to fetal exposure to low but not high doses of bisphenol A (BPA): evidence
as cAMP response element binding protein (CREB) rapidly, just five for effects on body weight, food intake, adipocytes, leptin, adiponectin, insulin and
glucose regulation. Reprod. Toxicol. 42, 256–268.
minutes after the stimulation produced at low doses (Quesada et al., Angle, B.M., Do, R.P., Ponzi, D., Stahlhut, R.W., Drury, B.E., Nagel, S.C., Welshons, W.V.,
2002). This effect is of great interest as CREB activation is found to be Besch-Williford, C.L., Palanza, P., Parmigiani, S., vom Saal, F.S., Taylor, J.A., 2013b.
involved in insulin gene expression and β cell survival (Jhala et al., Metabolic disruption in male mice due to fetal exposure to low but not high doses of
bisphenol A (BPA): evidence for effects on body weight, food intake, adipocytes,
2003; Oetjen et al., 1994). leptin, adiponectin, insulin and glucose regulation. Reprod. Toxicol. 42, 256–268.
Ariemma, F., D’Esposito, V., Liguoro, D., Oriente, F., Cabaro, S., Liotti, A., Cimmino, I.,
7. Conclusion Longo, M., Beguinot, F., Formisano, P., Valentino, R., 2016. Low-dose Bisphenol-A
impairs adipogenesis and generates dysfunctional 3T3-L1 adipocytes. PLoS One 11
e0150762–e0150762.
BPA has been found in urine and blood samples of the general po- Ascenzi, P., Bocedi, A., Marino, M., 2006. Structure-function relationship of oestrogen
pulation which indicates that BPA exposure is a global issue that should receptor alpha and beta: impact on human health. Mol. Asp. Med. 27, 299–402.
Ashcroft, F.M., Rorsman, P., 2013. K(ATP) channels and islet hormone secretion: new
be urgently addressed. In addition to inducing endocrine disruption in insights and controversies. Nature reviews. Endocrinology 9, 660–669.
adults, BPA has a potential to accelerate the metabolic programming in Bansal, A., Rashid, C., Xin, F., Li, C., Polyak, E., Duemler, A., van der Meer, T., Stefaniak,
young children that is an actual matter of debate. BPA exposure may M., Wajid, S., Doliba, N., 2017. Sex-and dose-specific effects of maternal bisphenol A
exposure on pancreatic islets of first-and second-generation adult mice offspring.
even impair the neurological development of infants and children.
Environ. Health Perspect. 125, 097022.
Owing to these neurological effects, FDA has recently introduced new Barbonetti, A., Castellini, C., Di Giammarco, N., Santilli, G., Francavilla, S., Francavilla,
regulations for its control and mandated its complete removal from F., 2016. In vitro exposure of human spermatozoa to bisphenol A induces pro-oxi-
commonly used baby products. Although there are various pathways dative/apoptotic mitochondrial dysfunction. Reprod. Toxicol. 66, 61–67.
Barker, D.J., 1995. The fetal and infant origins of disease. Eur. J. Clin. Invest. 25,
through which BPA alters energy metabolism, impairment of insulin 457–463.
signaling pathways is the key factor for the disruption of carbohydrate Barker, D., 1996. The fetal origins of hypertension. Journal of hypertension. Supplement
metabolism. BPA exposure and its potential consequences should be 14, S117–120.
Barker, D.J., 1998. In utero programming of chronic disease. Clin. Sci. 95, 115–128.
taken more seriously and more regulatory action taken to control Barros, R.P.A., Machado, U.F., Gustafsson, J.-A., 2006. Oestrogen receptors: new players
human and environmental exposure to BPA. A focus on developing in diabetes mellitus. Trends Mol. Med. 12, 425–431.
safer alternatives to replace this commonly used chemical agent will Bellavia, A., Cantonwine, D.E., Meeker, J.D., Hauser, R., Seely, E.W., McElrath, T.F.,
James-Todd, T., 2018. Pregnancy urinary bisphenol-A concentrations and glucose
make significant progress in safeguarding future generations from the levels across BMI categories. Environ. Int. 113, 35–41.
numerous hazards and dangers posed by BPA and its fellow EDCs. Ben-Jonathan, N., Hugo, E.R., Brandebourg, T.D., 2009. Effects of bisphenol A on adi-
pokine release from human adipose tissue: implications for the metabolic syndrome.
Mol. Cell. Endocrinol. 304, 49–54.
Declaration of Competing Interest
Bhandari, R., Xiao, J., Shankar, A., 2013. Urinary bisphenol A and obesity in US children.
Am. J. Epidemiol. 177, 1263–1270.
Bo, J., Xie, S., Guo, Y., Zhang, C., Guan, Y., Li, C., Lu, J., Meng, Q.H., 2016. Methylglyoxal
Nothing to declare.
impairs insulin secretion of pancreatic beta-cells through increased production of
ROS and mitochondrial dysfunction mediated by upregulation of UCP2 and MAPKs.
Acknowledgement J. Diabetes Res. 2016, 2029854.
Bodin, J., Bølling, A.K., Becher, R., Kuper, F., Løvik, M., Nygaard, U.C., 2013.
Transmaternal bisphenol A exposure accelerates diabetes type 1 development in NOD
This work has been financially supported by the research grant mice. Toxicol. Sci. 137, 311–323.
(5661/Punjab/NRPU/R&D/HEC/2016) received from Higher Boucher, J.G., Husain, M., Rowan-Carroll, A., Williams, A., Yauk, C.L., Atlas, E., 2014.
Education Commission (HEC) of Pakistan. Identification of mechanisms of action of bisphenol a-induced human preadipocyte
differentiation by transcriptional profiling. Obesity (Silver Spring) 22, 2333–2343.
Braun, J.M., Kalkbrenner, A.E., Calafat, A.M., Bernert, J.T., Ye, X., Silva, M.J., Barr, D.B.,
References Sathyanarayana, S., Lanphear, B.P., 2010. Variability and predictors of urinary bi-
sphenol A concentrations during pregnancy. Environ. Health Perspect. 119, 131–137.
Bryzgalova, G., Gao, H., Ahren, B., Zierath, J.R., Galuska, D., Steiler, T.L., Dahlman-
Acconcia, F., Pallottini, V., Marino, M., 2015. Molecular mechanisms of action of BPA.
Wright, K., Nilsson, S., Gustafsson, J.A., Efendic, S., Khan, A., 2006. Evidence that
Dose Response 13 1559325815610582–1559325815610582.
ooestrogen receptor-alpha plays an important role in the regulation of glucose
Aekplakorn, W., Chailurkit, Lo., Ongphiphadhanakul, B., 2015. Relationship of serum
homeostasis in mice: insulin sensitivity in the liver. Diabetologia 49, 588–597.
bisphenol A with diabetes in the Thai population, N ational H ealth E xamination S
Calafat, A.M., Kuklenyik, Z., Reidy, J.A., Caudill, S.P., Ekong, J., Needham, L.L., 2004.
urvey IV, 2009. J. Diabetes 7, 240–249.
Urinary concentrations of bisphenol A and 4-nonylphenol in a human reference po-
Ahmadkhaniha, R., Mansouri, M., Yunesian, M., Omidfar, K., Jeddi, M.Z., Larijani, B.,
pulation. Environ. Health Perspect. 113, 391–395.
Mesdaghinia, A., Rastkari, N., 2014. Association of urinary bisphenol a concentration
Calafat, A.M., Ye, X., Wong, L.-Y., Reidy, J.A., Needham, L.L., 2007. Exposure of the US
with type-2 diabetes mellitus. J. Environ. Health Sci. Eng. 12, 64.
population to bisphenol A and 4-tertiary-octylphenol: 2003–2004. Environ. Health
Akash, M.S.H., Rehman, K., Chen, S., 2013. Role of inflammatory mechanisms in pa-
Perspect. 116, 39–44.
thogenesis of type 2 diabetes mellitus. J. Cell. Biochem. 114, 525–531.
Calafat, A.M., Weuve, J., Ye, X., Jia, L.T., Hu, H., Ringer, S., Huttner, K., Hauser, R., 2008.
Akash, M.S.H., Rehman, K., Liaqat, A., 2018. Tumor necrosis factor-alpha: role in de-
Exposure to bisphenol A and other phenols in neonatal intensive care unit premature
velopment of insulin resistance and pathogenesis of type 2 diabetes mellitus. J. Cell.
infants. Environ. Health Perspect. 117, 639–644.
Biochem. 119, 105–110.
Cantonwine, D., Meeker, J.D., Hu, H., Sánchez, B.N., Lamadrid-Figueroa, H., Mercado-
Alavian‐Ghavanini, A., Rüegg, J., 2018. Understanding epigenetic effects of endocrine
García, A., Fortenberry, G.Z., Calafat, A.M., Téllez-Rojo, M.M., 2010. Bisphenol a
disrupting chemicals: from mechanisms to novel test methods. Basic Clin. Pharmacol.
exposure in Mexico city and risk of prematurity: a pilot nested case control study.
Toxicol. 122, 38–45.
Environ. Health 9, 62.
Alonso-Magdalena, P., Morimoto, S., Ripoll, C., Fuentes, E., Nadal, A., 2006. The
Cao, X.-L., Corriveau, J., Popovic, S., 2010. Bisphenol A in canned food products from

10
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

Canadian markets. J. Food Prot. 73, 1085–1089. Belgium. Chemosphere 76, 755–760.
Carlsson, A., Sørensen, K., Andersson, A.-M., Frederiksen, H., Juul, A., 2018. Bisphenol A, Geens, T., Aerts, D., Berthot, C., Bourguignon, J.-P., Goeyens, L., Lecomte, P., Maghuin-
phthalate metabolites and glucose homeostasis in healthy normal-weight children. Rogister, G., Pironnet, A.-M., Pussemier, L., Scippo, M.-L., 2012a. A review of dietary
Endocr. Connect. 7, 232–238. and non-dietary exposure to bisphenol-A. Food Chem. Toxicol. 50, 3725–3740.
Carwile, J.L., Michels, K.B., 2011. Urinary bisphenol A and obesity: NHANES 2003–2006. Geens, T., Aerts, D., Berthot, C., Bourguignon, J.P., Goeyens, L., Lecomte, P., Maghuin-
Environ. Res. 111, 825–830. Rogister, G., Pironnet, A.M., Pussemier, L., Scippo, M.L., Van Loco, J., Covaci, A.,
Casals-Casas, C., Desvergne, B., 2011. Endocrine disruptors: from endocrine to metabolic 2012b. A review of dietary and non-dietary exposure to bisphenol-A. Food Chem.
disruption. Annu. Rev. Physiol. 73, 135–162. Toxicol. 50, 3725–3740.
Casellas, A., Mallol, C., Salavert, A., Jimenez, V., Garcia, M., Agudo, J., Obach, M., Geens, T., Goeyens, L., Kannan, K., Neels, H., Covaci, A., 2012c. Levels of bisphenol-A in
Haurigot, V., Vilà, L., Molas, M., 2015. Insulin-like growth factor 2 overexpression thermal paper receipts from Belgium and estimation of human exposure. Sci. Total
induces β-cell dysfunction and increases beta-cell susceptibility to damage. J. Biol. Environ. 435, 30–33.
Chem. 290, 16772–16785. George, V.C., Rupasinghe, H.P.V., 2018. DNA damaging and apoptotic potentials of
Chailurkit, L.-o., Tengpraettanakorn, P., Chanprasertyotin, S., Ongphiphadhanakul, B., Bisphenol A and Bisphenol S in human bronchial epithelial cells. Environ. Toxicol.
2017. Is bisphenol A exposure associated with the development of glucose intoler- Pharmacol. 60, 52–57.
ance and increased insulin resistance in Thais? Nutr. Health 23, 185–191. Gilbert, S.F., Epel, D., 2009. Ecological developmental biology: integrating epigenetics,
Chakera, A.J., Hurst, P.S., Spyer, G., Ogunnowo-Bada, E.O., Marsh, W.J., Riches, C.H., medicine, and evolution. Yale J. Biol. Med. 82, 231–232.
Yueh, C.-Y., Markkula, S.P., Dalley, J.W., Cox, R.D., 2018. Molecular reductions in Gluckman, P.D., Hanson, M.A., 2004. Living with the past: evolution, development, and
glucokinase activity increase counter-regulatory responses to hypoglycemia in mice patterns of disease. Science 305, 1733–1736.
and humans with diabetes. Mol. Metab. 17, 17–27. Gong, X., Xie, H., Li, X., Wu, J., Lin, Y., 2017. Bisphenol A induced apoptosis and tran-
Chang, H., Wang, D., Xia, W., Pan, X., Huo, W., Xu, S., Li, Y., 2016. Epigenetic disruption scriptome differences of spermatogonial stem cells in vitro. Acta Biochim. Biophys.
and glucose homeostasis changes following low-dose maternal bisphenol A exposure. Sin. 49, 780–791.
Toxicol. Res. 5, 1400–1409. Goodson, A., Robin, H., Summerfield, W., Cooper, I., 2004. Migration of bisphenol A from
Chiu, Y.-H., Mínguez-Alarcón, L., Ford, J.B., Keller, M., Seely, E.W., Messerlian, C., can coatings—effects of damage, storage conditions and heating. Food Addit.
Petrozza, J., Williams, P.L., Ye, X., Calafat, A.M., 2017. Trimester-specific urinary Contam. 21, 1015–1026.
bisphenol A concentrations and blood glucose levels among pregnant women from a Gore, A.C., Chappell, V., Fenton, S., Flaws, J.A., Nadal, A., Prins, G., Toppari, J., Zoeller,
fertility clinic. J. Clin. Endocrinol. Metab. 102, 1350–1357. R., 2015. EDC-2: the Endocrine Society’s second scientific statement on endocrine-
Cooke, P.S., Heine, P.A., Taylor, J.A., Lubahn, D.B., 2001. The role of oestrogen and disrupting chemicals. Endocr. Rev. 36, E1–E150.
oestrogen receptor-alpha in male adipose tissue. Mol. Cell. Endocrinol. 178, 147–154. Green, D.R., Llambi, F., 2015. Cell death signaling. Cold Spring Harb. Perspect. Biol. 7.
Cooper, J.E., Kendig, E.L., Belcher, S.M., 2011. Assessment of bisphenol A released from Gross, A., 2016. BCL-2 family proteins as regulators of mitochondria metabolism.
reusable plastic, aluminium and stainless steel water bottles. Chemosphere 85, Biochim. Biophys. Acta 1857, 1243–1246.
943–947. Haishima, Y., Hayashi, Y., Yagami, T., Nakamura, A., 2001. Elution of bisphenol‐A from
de Sousa, L.N., Marques, T.F., Bernardo, G.P., Bernardo, L.P., Parente, L.L., Parente, R.E., hemodialyzers consisting of polycarbonate and polysulfone resins. J. Biomed. Mater.
Telles, M.V., Malheiro, D.R., 2018. Endocrine disruptors: bisphenol A and its relation Res. 58, 209–215.
with obesity. Amadeus Int. Multidiscip. J. 2, 41–49. Hales, C.N., Barker, D.J., 1992. Type 2 (non-insulin-dependent) diabetes mellitus: the
Ding, Q., Mracek, T., Gonzalez-Muniesa, P., Kos, K., Wilding, J., Trayhurn, P., Bing, C., thrifty phenotype hypothesis. Diabetologia 35, 595–601.
2009. Identification of macrophage inhibitory cytokine-1 in adipose tissue and its Hanaoka, T., Kawamura, N., Hara, K., Tsugane, S., 2002. Urinary bisphenol A and plasma
secretion as an adipokine by human adipocytes. Endocrinology 150, 1688–1696. hormone concentrations in male workers exposed to bisphenol A diglycidyl ether and
Dodds, E.C., Lawson, W., 1936. Synthetic strogenic agents without the phenanthrene mixed organic solvents. Occup. Environ. Med. 59, 625–628.
nucleus. Nature 137, 996. Handy, D.E., Castro, R., Loscalzo, J., 2011. Epigenetic modifications: basic mechanisms
Donath, M.Y., Shoelson, S.E., 2011. Type 2 diabetes as an inflammatory disease. Nature and role in cardiovascular disease. Circulation 123, 2145–2156.
reviews. Immunology 11, 98–107. Hectors, T.L.M., Vanparys, C., Pereira-Fernandes, A., Martens, G.A., Blust, R., 2013.
Dou, J.F., Puttabyatappa, M., Padmanabhan, V., Bakulski, K., 2019. Developmental Evaluation of the INS-1 832/13 cell line as a beta-cell based screening system to
Programming: Transcriptional Regulation of Visceral and Subcutaneous Adipose by assess pollutant effects on beta-cell function. PLoS One 8 e60030–e60030.
Prenatal Bisphenol-A in Female Sheep. bioRxiv. Heindel, J.J., Vandenberg, L.N., 2015. Developmental origins of health and disease: a
Drozdz, K., Wysokinski, D., Krupa, R., Wozniak, K., 2011. Bisphenol A-glycidyl metha- paradigm for understanding disease etiology and prevention. Curr. Opin. Pediatr. 27,
crylate induces a broad spectrum of DNA damage in human lymphocytes. Arch. 248.
Toxicol. 85, 1453–1461. Heindel, J.J., Blumberg, B., Cave, M., Machtinger, R., Mantovani, A., Mendez, M.A.,
Duan, Y., Yao, Y., Wang, B., Han, L., Wang, L., Sun, H., Chen, L., 2018. Association of Nadal, A., Palanza, P., Panzica, G., Sargis, R., 2017. Metabolism disrupting chemicals
urinary concentrations of bisphenols with type 2 diabetes mellitus: a case-control and metabolic disorders. Reprod. Toxicol. 68, 3–33.
study. Environ. Pollut. 243, 1719–1726. Hock, M.B., Kralli, A., 2009. Transcriptional control of mitochondrial biogenesis and
Dyachok, O., Idevall-Hagren, O., Sagetorp, J., Tian, G., Wuttke, A., Arrieumerlou, C., function. Annu. Rev. Physiol. 71, 177–203.
Akusjarvi, G., Gylfe, E., Tengholm, A., 2008. Glucose-induced cyclic AMP oscillations Hwang, J.K., Min, K.H., Choi, K.H., Hwang, Y.C., Jeong, I.K., Ahn, K.J., Chung, H.Y.,
regulate pulsatile insulin secretion. Cell Metab. 8, 26–37. Chang, J.S., 2013. Bisphenol A reduces differentiation and stimulates apoptosis of
Edlich, F., 2018. BCL-2 proteins and apoptosis: recent insights and unknowns. Biochem. osteoclasts and osteoblasts. Life Sci. 93, 367–372.
Biophys. Res. Commun. 500, 26–34. Hwang, S., Lim, J.-E., Choi, Y., Jee, S.H., 2018. Bisphenol A exposure and type 2 diabetes
EFSA Panel on Food Contact Materials, E., Flavourings, Aids, P, 2015. Scientific Opinion mellitus risk: a meta-analysis. BMC Endocr. Disord. 18 81–81.
on the risks to public health related to the presence of bisphenol A (BPA) in food- İnce, T., Balcı, A., Yalçın, S.S., Özkemahlı, G., Erkekoglu, P., Kocer-Gumusel, B.,
stuffs. EFSA J. 13, 3978. Yurdakök, K., 2018. Urinary bisphenol-A levels in children with type 1 diabetes
Eng, D.S., Lee, J.M., Gebremariam, A., Meeker, J.D., Peterson, K., Padmanabhan, V., mellitus. J. Pediatr. Endocrinol. Metab. 31, 829–836.
2013. Bisphenol A and chronic disease risk factors in US children. Pediatrics 132, Jhala, U.S., Canettieri, G., Screaton, R.A., Kulkarni, R.N., Krajewski, S., Reed, J., Walker,
e637–e645. J., Lin, X., White, M., Montminy, M., 2003. cAMP promotes pancreatic β-cell survival
EPA, U.S, 2010. Bisphenol A Action Plan. via CREB-mediated induction of IRS2. Genes Dev. 17, 1575–1580.
Estaquier, J., Vallette, F., Vayssiere, J.L., Mignotte, B., 2012. The mitochondrial pathways Kang, J.-H., Kondo, F., 2003. Determination of bisphenol A in milk and dairy products by
of apoptosis. Adv. Exp. Med. Biol. 942, 157–183. high-performance liquid chromatography with fluorescence detection. J. Food Prot.
EU, 2018. COMMISSION REGULATION (EU) 2018/213 Of 12 February 2018 on the Use 66, 1439–1443.
of Bisphenol A in Varnishes and Coatings Intended to Come Into Contact With Food Karch, J., Kwong, J.Q., Burr, A.R., Sargent, M.A., Elrod, J.W., Peixoto, P.M., Martinez-
and Amending Regulation (EU) No 10/2011 As Regards the Use of That Substance in Caballero, S., Osinska, H., Cheng, E.H., Robbins, J., Kinnally, K.W., Molkentin, J.D.,
Plastic Food Contact. 2013. Bax and Bak function as the outer membrane component of the mitochondrial
Fernandez, M., Arrebola, J., Taoufiki, J., Navalón, A., Ballesteros, O., Pulgar, R., Vilchez, permeability pore in regulating necrotic cell death in mice. eLife 2, e00772.
J., Olea, N., 2007. Bisphenol-A and chlorinated derivatives in adipose tissue of Kaur, S., Saluja, M., Bansal, M.P., 2018. Bisphenol A induced oxidative stress and apop-
women. Reprod. Toxicol. 24, 259–264. tosis in mice testes: modulation by selenium. Andrologia 50.
Fleisch, A.F., Sheffield, P.E., Chinn, C., Edelstein, B.L., Landrigan, P.J., 2010. Bisphenol A Ke, Z.-H., Pan, J.-X., Jin, L.-Y., Xu, H.-Y., Yu, T.-T., Ullah, K., Rahman, T.U., Ren, J.,
and related compounds in dental materials. Pediatrics 126, 760. Cheng, Y., Dong, X.-Y., 2016. Bisphenol A exposure may induce hepatic lipid accu-
Foryst-Ludwig, A., Clemenz, M., Hohmann, S., Hartge, M., Sprang, C., Frost, N., Krikov, mulation via reprogramming the DNA methylation patterns of genes involved in lipid
M., Bhanot, S., Barros, R., Morani, A., 2008. Metabolic actions of oestrogen receptor metabolism. Sci. Rep. 6, 31331.
beta (ERβ) are mediated by a negative cross-talk with PPARγ. PLoS Genet. 4. Kim, Y.J., 2009. In utero programming of chronic disease. J. Womens Med. 2, 48–53.
García-Arevalo, M., Alonso-Magdalena, P., Dos Santos, J.R., Quesada, I., Carneiro, E.M., Kim, K., Park, H., 2013. Association between urinary concentrations of bisphenol A and
Nadal, A., 2014. Exposure to bisphenol-A during pregnancy partially mimics the ef- type 2 diabetes in Korean adults: a population-based cross-sectional study. Int. J. Hyg.
fects of a high-fat diet altering glucose homeostasis and gene expression in adult male Environ. Health 216, 467–471.
mice. PLoS One 9, e100214. Kim, J.B., Wright, H.M., Wright, M., Spiegelman, B.M., 1998. ADD1/SREBP1 activates
García-Arévalo, M., Alonso-Magdalena, P., Servitja, J.-M., Boronat-Belda, T., Merino, B., PPARgamma through the production of endogenous ligand. Proc. Natl. Acad. Sci. U.
Villar-Pazos, S., Medina-Gómez, G., Novials, A., Quesada, I., Nadal, A., 2016. S. A. 95, 4333–4337.
Maternal exposure to bisphenol-A during pregnancy increases pancreatic β-cell Konieczna, A., Rutkowska, A., Rachon, D., 2015. Health risk of exposure to Bisphenol A
growth during early life in male mice offspring. Endocrinology 157, 4158–4171. (BPA). Roczniki Państwowego Zakładu Higieny 66.
Geens, T., Roosens, L., Neels, H., Covaci, A., 2009. Assessment of human exposure to Kourouma, A., Quan, C., Duan, P., Qi, S., Yu, T., Wang, Y., Yang, K., 2015. Bisphenol A
Bisphenol-A, Triclosan and Tetrabromobisphenol-A through indoor dust intake in induces apoptosis in liver cells through induction of ROS. Adv. Toxicol. 2015.

11
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

Kumar, A., Verma, R., Agrawal, N., 2018. A study of urinary bisphenol A levels in en- peppers and an acid food simulant. J. Agric. Food Chem. 50, 7299–7302.
docrine disorders. Int. J. Res. Med. Sci. 6, 696–700. Musatov, S., Chen, W., Pfaff, D.W., Mobbs, C.V., Yang, X.-J., Clegg, D.J., Kaplitt, M.G.,
Lakhani, S.A., Masud, A., Kuida, K., Porter Jr., G.A., Booth, C.J., Mehal, W.Z., Inayat, I., Ogawa, S., 2007. Silencing of oestrogen receptor alpha in the ventromedial nucleus of
Flavell, R.A., 2006. Caspases 3 and 7: key mediators of mitochondrial events of hypothalamus leads to metabolic syndrome. Proc. Natl. Acad. Sci. U. S. A. 104,
apoptosis. Science 311, 847–851. 2501–2506.
LaKind, J.S., Goodman, M., Naiman, D.Q., 2012. Use of NHANES data to link chemical Nadal, A., Rovira, J.M., Laribi, O., Leon-quinto, T., Andreu, E., Ripoll, C., Soria, B., 1998.
exposures to chronic diseases: a cautionary tale. PLoS One 7 e51086–e51086. Rapid insulinotropic effect of 17beta-estradiol via a plasma membrane receptor.
Lang, I.A., Galloway, T.S., Scarlett, A., Henley, W.E., Depledge, M., Wallace, R.B., Melzer, FASEB J. 12, 1341–1348.
D., 2008. Association of urinary bisphenol A concentration with medical disorders Nadal, A., Ropero, A.B., Laribi, O., Maillet, M., Fuentes, E., Soria, B., 2000. Nongenomic
and laboratory abnormalities in adults. JAMA 300, 1303–1310. actions of oestrogens and xenooestrogens by binding at a plasma membrane receptor
Lee, M.-J., Gong, D.-W., Burkey, B.F., Fried, S.K., 2011. Pathways regulated by gluco- unrelated to oestrogen receptor alpha and oestrogen receptor beta. Proc. Natl. Acad.
corticoids in omental and subcutaneous human adipose tissues: a microarray study. Sci. U. S. A. 97, 11603–11608.
Am. J. Physiol. Endocrinol. Metab. 300, E571–E580. Nadal, A., Alonso-Magdalena, P., Soriano, S., Quesada, I., Ropero, A.B., 2009. The pan-
Lee, I., Kim, S., Kim, K.-T., Kim, S., Park, S., Lee, H., Jeong, Y., Lim, J.-E., Moon, H.-B., creatic beta-cell as a target of oestrogens and xenooestrogens: implications for blood
Choi, K., 2018. Bisphenol A exposure through receipt handling and its association glucose homeostasis and diabetes. Mol. Cell. Endocrinol. 304, 63–68.
with insulin resistance among female cashiers. Environ. Int. 117, 268–275. Nadal, A., Alonso-Magdalena, P., Soriano, S., Ripoll, C., Fuentes, E., Quesada, I., Ropero,
Li, G., Chang, H., Xia, W., Mao, Z., Li, Y., Xu, S., 2014. F0 maternal BPA exposure induced A.B., 2011. Role of oestrogen receptors alpha, beta and GPER1/GPR30 in pancreatic
glucose intolerance of F2 generation through DNA methylation change in Gck. beta-cells. Front. Biosci. (Landmark Ed) 16, 251–260.
Toxicol. Lett. 228, 192–199. Nadal, A., Quesada, I., Tudurí, E., Nogueiras, R., Alonso-Magdalena, P., 2017. Endocrine-
Li, A.J., Xue, J., Lin, S., Al-Malki, A.L., Al-Ghamdi, M.A., Kumosani, T.A., Kannan, K., disrupting chemicals and the regulation of energy balance. Nat. Rev. Endocrinol. 13,
2018. Urinary concentrations of environmental phenols and their association with 536.
type 2 diabetes in a population in Jeddah, Saudi Arabia. Environ. Res. 166, 544–552. Ning, G., Bi, Y., Wang, T., Xu, M., Xu, Y., Huang, Y., Li, M., Li, X., Wang, W., Chen, Y.,
Liang, Y.Q., Akishita, M., Kim, S., Ako, J., Hashimoto, M., Iijima, K., Ohike, Y., Watanabe, 2011. Relationship of urinary bisphenol A concentration to risk for prevalent type 2
T., Sudoh, N., Toba, K., Yoshizumi, M., Ouchi, Y., 2002. Oestrogen receptor beta is diabetes in Chinese adults: a cross-sectional analysis. Ann. Intern. Med. 155,
involved in the anorectic action of oestrogen. Int. J. Obes. Relat. Metab. Disord. 26, 368–374.
1103–1109. Oetjen, E., Diedrich, T., Eggers, A., Eckert, B., Knepel, W., 1994. Distinct properties of the
Liao, S.L., Tsai, M.H., Lai, S.H., Yao, T.C., Hua, M.C., Yeh, K.W., Chiang, C.H., Huang, cAMP-responsive element of the rat insulin I gene. J. Biol. Chem. 269, 27036–27044.
S.Y., Huang, J.L., 2016. Prenatal exposure to bisphenol-A is associated with Toll-like Ohlstein, J.F., Strong, A.L., McLachlan, J.A., Gimble, J.M., Burow, M.E., Bunnell, B.A.,
receptor-induced cytokine suppression in neonates. Pediatr. Res. 79, 438–444. 2014a. Bisphenol A enhances adipogenic differentiation of human adipose stromal/
Lin, Y., Sun, X., Qiu, L., Wei, J., Huang, Q., Fang, C., Ye, T., Kang, M., Shen, H., Dong, S., stem cells. J. Mol. Endocrinol. 53, 345–353.
2013a. Exposure to bisphenol A induces dysfunction of insulin secretion and apop- Ohlstein, J.F., Strong, A.L., McLachlan, J.A., Gimble, J.M., Burow, M.E., Bunnell, B.A.,
tosis through the damage of mitochondria in rat insulinoma (INS-1) cells. Cell Death 2014b. Bisphenol A enhances adipogenic differentiation of human adipose stromal/
Dis. 4 e460–e460. stem cells. J. Mol. Endocrinol. 53, 345–353.
Lin, Y., Sun, X., Qiu, L., Wei, J., Huang, Q., Fang, C., Ye, T., Kang, M., Shen, H., Dong, S., Oldring, P., Castle, L., O’Mahony, C., Dixon, J., 2014. Estimates of dietary exposure to
2013b. Exposure to bisphenol A induces dysfunction of insulin secretion and apop- bisphenol A (BPA) from light metal packaging using food consumption and packaging
tosis through the damage of mitochondria in rat insulinoma (INS-1) cells. Cell Death usage data: a refined deterministic approach and a fully probabilistic (FACET) ap-
Dis. 4, e460. proach. Food Addit. Contam. Part A 31, 466–489.
Lin, Y., Ding, D., Huang, Q., Liu, Q., Lu, H., Lu, Y., Chi, Y., Sun, X., Ye, G., Zhu, H., 2017. Ortega, F.J., Moreno-Navarrete, J.M., Pardo, G., Sabater, M., Hummel, M., Ferrer, A.,
Downregulation of miR-192 causes hepatic steatosis and lipid accumulation by in- Rodriguez-Hermosa, J.I., Ruiz, B., Ricart, W., Peral, B., Fernández-Real, J.M., 2010.
ducing SREBF1: novel mechanism for bisphenol A-triggered non-alcoholic fatty liver MiRNA expression profile of human subcutaneous adipose and during adipocyte
disease. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1862, 869–882. differentiation. PLoS One 5 e9022–e9022.
Liu, B., Lehmler, H.J., Sun, Y., Xu, G., Liu, Y., Zong, G., Sun, Q., Hu, F.B., Wallace, R.B., Perreault, L., McCurdy, C., Kerege, A.A., Houck, J., Færch, K., Bergman, B.C., 2013.
Bao, W., 2017. Bisphenol A substitutes and obesity in US adults: analysis of a po- Bisphenol A impairs hepatic glucose sensing in C57BL/6 male mice. PLoS One 8,
pulation-based, cross-sectional study. Lancet Planet. Health 1, e114–e122. e69991.
Loganathan, S.N., Kannan, K., 2011. Occurrence of bisphenol A in indoor dust from two Prasad, S.K., Kulshreshtha, A., Qureshi, T.N., 2009. Antidiabetic activity of some herbal
locations in the eastern United States and implications for human exposures. Arch. plants in streptozotocin induced diabetic albino rats. Pak. J. Nutr. 8, 551–557.
Environ. Contam. Toxicol. 61, 68–73. Puttabyatappa, M., Martin, J.D., Andriessen, V., Stevenson, M., Zeng, L., Pennathur, S.,
Loscalzo, J., Handy, D.E., 2014. Epigenetic modifications: basic mechanisms and role in Padmanabhan, V., 2019. Developmental programming: changes in mediators of in-
cardiovascular disease (2013 Grover Conference series). Pulm. Circ. 4, 169–174. sulin sensitivity in prenatal bisphenol A-treated female sheep. Reprod. Toxicol. 85,
Ma, Y., Xia, W., Wang, D., Wan, Y., Xu, B., Chen, X., Li, Y., Xu, S., 2013. Hepatic DNA 110–122.
methylation modifications in early development of rats resulting from perinatal BPA Quesada, I., Fuentes, E., Viso-León, M.C., Soria, B., Ripoll, C., Nadal, A., 2002. Low doses
exposure contribute to insulin resistance in adulthood. Diabetologia 56, 2059–2067. of the endocrine disruptor bisphenol-A and the native hormone 17β-estradiol rapidly
MacDonald, P.E., Joseph, J.W., Rorsman, P., 2005. Glucose-sensing mechanisms in pan- activate transcription factor CREB. FASEB J. 16, 1671–1673.
creatic beta-cells. Philos. Trans. R. Soc. Lond., B, Biol. Sci. 360, 2211–2225. Rehman, K., Akash, M.S.H., 2017. Mechanism of generation of oxidative stress and pa-
Malaisé, Y., Menard, S., Cartier, C., Gaultier, E., Lasserre, F., Lencina, C., Harkat, C., thophysiology of type 2 diabetes mellitus: how are they interlinked? J. Cell. Biochem.
Geoffre, N., Lakhal, L., Castan, I., 2017. Gut dysbiosis and impairment of immune 118, 3577–3585.
system homeostasis in perinatally-exposed mice to Bisphenol A precede obese phe- Rehman, K., Akash, M.S.H., Liaqat, A., Kamal, S., Qadir, M.I., Rasul, A., 2017. Role of
notype development. Sci. Rep. 7, 14472. Interleukin-6 in development of insulin resistance and type 2 diabetes mellitus. Crit.
Mao, Z., Xia, W., Huo, W., Zheng, T., Bassig, B.A., Chang, H., Chen, T., Li, F., Pan, Y., Rev. Eukaryot. Gene Expr. 27, 229–236.
Peng, Y., 2017. Pancreatic impairment and Igf2 hypermethylation induced by de- Robledo, C., Peck, J.D., Stoner, J.A., Carabin, H., Cowan, L., Koch, H.M., Goodman, J.R.,
velopmental exposure to bisphenol A can be counteracted by maternal folate sup- 2013. Is bisphenol-A exposure during pregnancy associated with blood glucose levels
plementation. J. Appl. Toxicol. 37, 825–835. or diagnosis of gestational diabetes? J. Toxicol. Environ. Health Part A 76, 865–873.
Marmugi, A., Ducheix, S., Lasserre, F., Polizzi, A., Paris, A., Priymenko, N., Bertrand- Sabanayagam, C., Teppala, S., Shankar, A., 2013. Relationship between urinary bisphenol
Michel, J., Pineau, T., Guillou, H., Martin, P.G.P., Mselli-Lakhal, L., 2012a. Low doses A levels and prediabetes among subjects free of diabetes. Acta Diabetol. 50, 625–631.
of bisphenol A induce gene expression related to lipid synthesis and trigger trigly- Savastano, S., Tarantino, G., D’Esposito, V., Passaretti, F., Cabaro, S., Liotti, A., Liguoro,
ceride accumulation in adult mouse liver. Hepatology 55, 395–407. D., Perruolo, G., Ariemma, F., Finelli, C., Beguinot, F., Formisano, P., Valentino, R.,
Marmugi, A., Ducheix, S., Lasserre, F., Polizzi, A., Paris, A., Priymenko, N., 2015. Bisphenol-A plasma levels are related to inflammatory markers, visceral obe-
Bertrand‐Michel, J., Pineau, T., Guillou, H., Martin, P.G., 2012b. Low doses of bi- sity and insulin-resistance: a cross-sectional study on adult male population. J. Transl.
sphenol A induce gene expression related to lipid synthesis and trigger triglyceride Med. 13, 169.
accumulation in adult mouse liver. Hepatology 55, 395–407. Shankar, A., Teppala, S., 2011. Relationship between urinary bisphenol A levels and
Melzer, D., Rice, N.E., Lewis, C., Henley, W.E., Galloway, T.S., 2010. Association of ur- diabetes mellitus. J. Clin. Endocrinol. Metab. 96, 3822–3826.
inary bisphenol A concentration with heart disease: evidence from NHANES 2003/ Shapiro, G., Dodds, L., Arbuckle, T., Ashley-Martin, J., Fraser, W., Fisher, M., Taback, S.,
06. PLoS One 5 e8673–e8673. Keely, E., Bouchard, M., Monnier, P., 2015. Exposure to phthalates, bisphenol A and
Miao, M., Yuan, W., Zhu, G., He, X., Li, D.-K., 2011. In utero exposure to bisphenol-A and metals in pregnancy and the association with impaired glucose tolerance and ge-
its effect on birth weight of offspring. Reprod. Toxicol. 32, 64–68. stational diabetes mellitus: the MIREC study. Environ. Int. 83, 63–71.
Mirmira, P., Evans-Molina, C., 2014. Bisphenol A, obesity, and type 2 diabetes mellitus: Shelnutt, S., Kind, J., Allaben, W., 2013. Bisphenol A: update on newly developed data
genuine concern or unnecessary preoccupation? Transl. Res. 164, 13–21. and how they address NTP’s 2008 finding of “some concern”. Food Chem. Toxicol.
Moon, M.K., Jeong, I.-K., Jung Oh, T., Ahn, H.Y., Kim, H.H., Park, Y.J., Jang, H.C., Park, 57, 284–295.
K.S., 2015. Long-term oral exposure to bisphenol A induces glucose intolerance and Shimpi, P.C., More, V.R., Paranjpe, M., Donepudi, A.C., Goodrich, J.M., Dolinoy, D.C.,
insulin resistance. J. Endocrinol. 226, 35–42. Rubin, B., Slitt, A.L., 2017. Hepatic lipid accumulation and Nrf2 expression following
Mouneimne, Y., Nasrallah, M., Khoueiry-Zgheib, N., Nasreddine, L., Nakhoul, N., Ismail, perinatal and peripubertal exposure to bisphenol A in a mouse model of nonalcoholic
H., Abiad, M., Koleilat, L., Tamim, H., 2017. Bisphenol A urinary level, its correlates, liver disease. Environ. Health Perspect. 125.
and association with cardiometabolic risks in Lebanese urban adults. Environ. Monit. Shirani, M., Alizadeh, S., Mahdavinia, M., Dehghani, M.A., 2019. The ameliorative effect
Assess. 189, 517. of quercetin on bisphenol A-induced toxicity in mitochondria isolated from rats.
Munguia-Lopez, E.M., Peralta, E., Gonzalez-Leon, A., Vargas-Requena, C., Soto-Valdez, Environ. Sci. Pollut. Res. Int.
H., 2002. Migration of bisphenol A (BPA) from epoxy can coatings to jalapeno Shu, X., Tang, S., Peng, C., Gao, R., Yang, S., Luo, T., Cheng, Q., Wang, Y., Wang, Z., Zhen,

12
M.S.H. Akash, et al. Environmental Toxicology and Pharmacology 77 (2020) 103373

Q., 2018. Bisphenol A is not associated with a 5-year incidence of type 2 diabetes: a Front. Endocrinol. 9.
prospective nested case–control study. Acta Diabetol. 55, 369–375. Valentino, R., D’Esposito, V., Ariemma, F., Cimmino, I., Beguinot, F., Formisano, P., 2016.
Silver, M.K., O’Neill, M.S., Sowers, M.R., Park, S.K., 2011. Urinary bisphenol A and type-2 Bisphenol A environmental exposure and the detrimental effects on human metabolic
diabetes in U.S. adults: data from NHANES 2003–2008. PLoS One 6 e26868–e26868. health: is it necessary to revise the risk assessment in vulnerable population? J.
Skogsberg, J., Dicker, A., Rydén, M., Åström, G., Nilsson, R., Bhuiyan, H., Vitols, S., Endocrinol. Invest. 39, 259–263.
Mairal, A., Langin, D., Alberts, P., 2008. ApoB100-LDL acts as a metabolic signal from Van Landuyt, K., Nawrot, T., Geebelen, B., De Munck, J., Snauwaert, J., Yoshihara, K.,
liver to peripheral fat causing inhibition of lipolysis in adipocytes. PLoS One 3. Scheers, H., Godderis, L., Hoet, P., Van Meerbeek, B., 2011. How much do resin-based
Song, L., Xia, W., Zhou, Z., Li, Y., Lin, Y., Wei, J., Wei, Z., Xu, B., Shen, J., Li, W., Xu, S., dental materials release? A meta-analytical approach. Dent. Mater. 27, 723–747.
2012. Low-level phenolic oestrogen pollutants impair islet morphology and beta-cell Vandenberg, L.N., Hauser, R., Marcus, M., Olea, N., Welshons, W.V., 2007. Human ex-
function in isolated rat islets. J. Endocrinol. 215, 303–311. posure to bisphenol A (BPA). Reprod. Toxicol. 24, 139–177.
Soriano, S., Ropero, A.B., Alonso-Magdalena, P., Ripoll, C., Quesada, I., Gassner, B., Kuhn, Veissi, M., Jafarirad, S., Ahangarpour, A., Mohaghegh, S.M., Malehi, A.S., 2018. Co-ex-
M., Gustafsson, J.A., Nadal, A., 2009. Rapid regulation of K(ATP) channel activity by posure to endocrine disruptors: effect of bisphenol A and soy extract on glucose
17{beta}-estradiol in pancreatic {beta}-cells involves the oestrogen receptor {beta} homeostasis and related metabolic disorders in male mice. Endocr. Regul. 52, 76–84.
and the atrial natriuretic peptide receptor. Mol. Endocrinol. 23, 1973–1982. Viñas, P., Campillo, N., Martínez-Castillo, N., Hernández-Córdoba, M., 2010. Comparison
Soriano, S., Alonso-Magdalena, P., Garcia-Arevalo, M., Novials, A., Muhammed, S.J., of two derivatization-based methods for solid-phase microextraction–gas chromato-
Salehi, A., Gustafsson, J.A., Quesada, I., Nadal, A., 2012. Rapid insulinotropic action graphy–mass spectrometric determination of bisphenol A, bisphenol S and biphenol
of low doses of bisphenol-A on mouse and human islets of Langerhans: role of oes- migrated from food cans. Anal. Bioanal. Chem. 397, 115–125.
trogen receptor beta. PLoS One 7, e31109. Volberg, V., Harley, K., Calafat, A.M., Davé, V., McFadden, J., Eskenazi, B., Holland, N.,
Soriano, S., Ripoll, C., Alonso-Magdalena, P., Fuentes, E., Quesada, I., Nadal, A., 2013. Maternal bisphenol a exposure during pregnancy and its association with
Martinez-Pinna, J., 2016. Effects of Bisphenol A on ion channels: experimental evi- adipokines in Mexican‐American children. Environ. Mol. Mutagen. 54, 621–628.
dence and molecular mechanisms. Steroids 111, 12–20. Wang, Q., Zhao, X.-F., Ji, Y.-L., Wang, H., Liu, P., Zhang, C., Zhang, Y., Xu, D.-X., 2010.
Spaeth, J.M., Gupte, M., Perelis, M., Yang, Y.-P., Cyphert, H., Guo, S., Liu, J.-H., Guo, M., Mitochondrial signaling pathway is also involved in bisphenol A induced germ cell
Bass, J., Magnuson, M.A., 2017. Defining a novel role for the Pdx1 transcription apoptosis in testes. Toxicol. Lett. 199, 129–135.
factor in islet β-cell maturation and proliferation during weaning. Diabetes 66, Wang, C., Zhang, J., Li, Q., Zhang, T., Deng, Z., Lian, J., Jia, D., Li, R., Zheng, T., Ding, X.,
2830–2839. Yang, F., Ma, C., Wang, R., Zhang, W., Guo Wen, J., 2017a. Low concentration of BPA
Stahlhut, R.W., Myers, J.P., Taylor, J.A., Nadal, A., Dyer, J.A., vom Saal, F.S., 2018. induces mice spermatocytes apoptosis via GPR30. Oncotarget 8, 49005–49015.
Experimental BPA exposure and glucose-stimulated insulin response in adult men and Wang, X., Wang, X., Chen, Q., Luo, Z.-C., Zhao, S., Wang, W., Zhang, H.-J., Zhang, J.,
women. J. Endocr. Soc. 2, 1173–1187. Ouyang, F., 2017b. Urinary bisphenol A concentration and gestational diabetes
Strakovsky, R.S., Wang, H., Engeseth, N.J., Flaws, J.A., Helferich, W.G., Pan, Y.-X., Lezmi, mellitus in Chinese women. Epidemiology 28, S41–S47.
S., 2015. Developmental bisphenol A (BPA) exposure leads to sex-specific modifica- Wei, J., Lin, Y., Li, Y., Ying, C., Chen, J., Song, L., Zhou, Z., Lv, Z., Xia, W., Chen, X., Xu, S.,
tion of hepatic gene expression and epigenome at birth that may exacerbate high-fat 2011. Perinatal exposure to bisphenol A at reference dose predisposes offspring to
diet-induced hepatic steatosis. Toxicol. Appl. Pharmacol. 284, 101–112. metabolic syndrome in adult rats on a high-fat diet. Endocrinology 152, 3049–3061.
Sugiura-Ogasawara, M., Ozaki, Y., Sonta, S.-i., Makino, T., Suzumori, K., 2005. Exposure Wei, J., Ding, D., Wang, T., Liu, Q., Lin, Y., 2017. MiR-338 controls BPA-triggered pan-
to bisphenol A is associated with recurrent miscarriage. Hum. Reprod. 20, creatic islet insulin secretory dysfunction from compensation to decompensation by
2325–2329. targeting Pdx-1. FASEB J. 31, 5184–5195.
Sun, Q., Cornelis, M.C., Townsend, M.K., Tobias, D.K., Eliassen, A.H., Franke, A.A., WHO, 2009. BISPHENOL A (BPA) - Current State of Knowledge and Future Actions by
Hauser, R., Hu, F.B., 2014. Association of urinary concentrations of bisphenol A and WHO and FAO.
phthalate metabolites with risk of type 2 diabetes: a prospective investigation in the Zhang, C.Y., Baffy, G., Perret, P., Krauss, S., Peroni, O., Grujic, D., Hagen, T., Vidal-Puig,
Nurses’ Health Study (NHS) and NHSII cohorts. Environ. Health Perspect. 122, A.J., Boss, O., Kim, Y.B., Zheng, X.X., Wheeler, M.B., Shulman, G.I., Chan, C.B.,
616–623. Lowell, B.B., 2001. Uncoupling protein-2 negatively regulates insulin secretion and is
Susiarjo, M., Xin, F., Bansal, A., Stefaniak, M., Li, C., Simmons, R.A., Bartolomei, M.S., a major link between obesity, beta cell dysfunction, and type 2 diabetes. Cell 105,
2015. Bisphenol A exposure disrupts metabolic health across multiple generations in 745–755.
the mouse. Endocrinology 156, 2049–2058. Zheng, Y., Ley, S.H., Hu, F.B., 2018. Global aetiology and epidemiology of type 2 diabetes
Tai, X., Chen, Y., 2016. Urinary bisphenol A concentrations positively associated with mellitus and its complications. Nat. Rev. Endocrinol. 14, 88.
glycated hemoglobin and other indicators of diabetes in Canadian men. Environ. Res. Zhou, Y., Chen, M., Zhao, F., Mu, D., Zhang, Z., Hu, J., 2015. Ubiquitous occurrence of
147, 172–178. chlorinated byproducts of bisphenol A and nonylphenol in bleached food contacting
Tang, L., Ye, H., Hong, Q., Wang, L., Wang, Q., Wang, H., Xu, L., Bu, S., Zhang, L., Cheng, papers and their implications for human exposure. Environ. Sci. Technol. 49,
J., 2014. Elevated CpG island methylation of GCK gene predicts the risk of type 2 7218–7226.
diabetes in Chinese males. Gene 547, 329–333. Zhu, Y., Zhang, C., 2016. Prevalence of gestational diabetes and risk of progression to
Teppala, S., Madhavan, S., Shankar, A., 2012. Bisphenol A and metabolic syndrome: re- type 2 diabetes: a global perspective. Curr. Diab. Rep. 16, 7.
sults from NHANES. Int. J. Endocrinol. 2012 598180–598180. Zoeller, R.T., Brown, T.R., Doan, L.L., Gore, A.C., Skakkebaek, N., Soto, A., Woodruff, T.,
Tudurí, E., Marroqui, L., Dos Santos, R.S., Quesada, I., Fuentes, E., Alonso-Magdalena, P., Vom Saal, F., 2012. Endocrine-disrupting chemicals and public health protection: a
2018. Timing of exposure and bisphenol-A: implications for diabetes development. statement of principles from the Endocrine Society. Endocrinology 153, 4097–4110.

13

You might also like