Next Article in Journal
Key Soybean Seedlings Drought-Responsive Genes and Pathways Revealed by Comparative Transcriptome Analyses of Two Cultivars
Next Article in Special Issue
Bisphenol A (BPA) Leading to Obesity and Cardiovascular Complications: A Compilation of Current In Vivo Study
Previous Article in Journal
A Novel CDK4/6 and PARP Dual Inhibitor ZC-22 Effectively Suppresses Tumor Growth and Improves the Response to Cisplatin Treatment in Breast and Ovarian Cancer
Previous Article in Special Issue
Behavioral Effects of Exposure to Phthalates in Female Rodents: Evidence for Endocrine Disruption?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Does Bisphenol A Confer Risk of Neurodevelopmental Disorders? What We Have Learned from Developmental Neurotoxicity Studies in Animal Models

1
Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
2
Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA 95819, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(5), 2894; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052894
Submission received: 29 January 2022 / Revised: 2 March 2022 / Accepted: 5 March 2022 / Published: 7 March 2022
(This article belongs to the Special Issue Advances in Endocrine Disruptors)

Abstract

:
Substantial evidence indicates that bisphenol A (BPA), a ubiquitous environmental chemical used in the synthesis of polycarbonate plastics and epoxy resins, can impair brain development. Clinical and epidemiological studies exploring potential connections between BPA and neurodevelopmental disorders in humans have repeatedly identified correlations between early BPA exposure and developmental disorders, such as attention deficit/hyperactivity disorder and autism spectrum disorder. Investigations using invertebrate and vertebrate animal models have revealed that developmental exposure to BPA can impair multiple aspects of neuronal development, including neural stem cell proliferation and differentiation, synapse formation, and synaptic plasticity—neuronal phenotypes that are thought to underpin the fundamental changes in behavior-associated neurodevelopmental disorders. Consistent with neuronal phenotypes caused by BPA, behavioral analyses of BPA-treated animals have shown significant impacts on behavioral endophenotypes related to neurodevelopmental disorders, including altered locomotor activity, learning and memory deficits, and anxiety-like behavior. To contextualize the correlations between BPA and neurodevelopmental disorders in humans, this review summarizes the current literature on the developmental neurotoxicity of BPA in laboratory animals with an emphasis on neuronal phenotypes, molecular mechanisms, and behavioral outcomes. The collective works described here predominantly support the notion that gestational exposure to BPA should be regarded as a risk factor for neurodevelopmental disorders.

1. Introduction

Bisphenol A (BPA, 2,2-bis (4′-hydroxyphenyl) propane), a ubiquitous chemical used in the synthesis of polycarbonate plastic and epoxy resins, is taking shape as a risk factor for neurodevelopmental disorders (NDDs). NDDs refer to a heterogeneous group of nervous system disorders caused by changes in brain development, including autism spectrum disorder (ASD), attention deficit/hyperactivity disorder (ADHD), intellectual disability (ID), learning disabilities, cerebral palsy, seizure disorders, and impairments in vision and hearing. Analysis of the prevalence of developmental disabilities in the United States (U.S.) between 2009 and 2017 indicated that 16.93% of children were diagnosed with an NDD [1]. Studies have also found that the prevalence of NDDs has increased significantly over the past several decades—most notably, the incidence ASD has increased by almost 300 percent in the last 20 years [2,3].
NDDs have complex etiologies and can result from both genetic susceptibilities and environmental factors [4,5]. Given the increasing prevalence of BPA in our environment [6], along with numerous studies showing an association between human BPA exposure and NDDs [7,8,9], BPA has garnered increasing attention over the past fifteen years for its ability to disrupt brain development. The aim of this review is to summarize studies that have delineated neurodevelopmental consequences of early (prenatal or perinatal) BPA exposure in animal models in an effort to illuminate the mechanisms by which it may impede human brain development.
BPA is categorized as an endocrine disrupting chemical (EDC) due to its ability to bind endogenous hormone receptors and cause adverse effects. Structurally similar to estradiol, BPA is most well-known for its ability to agonize and antagonize estrogen receptor (ER) subtypes and antagonize androgen receptors [10,11]. BPA also elicits non-estrogenic/non-androgenic impacts on development by binding other receptors, including the thyroid hormone receptor [12,13], glucocorticoid receptor [14,15,16], the G protein-coupled receptor, GPR30 [17] and Peroxisome Proliferator-Activated Receptor γ (PPARγ) [18,19]. The ability of BPA to influence hormonal signaling was first documented in the 1930s [20,21]. Despite awareness of its endocrine disrupting capability, BPA was adapted for use in the synthesis of polycarbonate plastics in the 1950s and quickly became one of the most prevalent synthetic compounds in the world [6,22]. An estimated 7.7 million metric tons of BPA were generated worldwide in 2015, and production is expected to rise to 10.6 million metric tons in 2022 [6].
BPA enters the body via ingestion, dermal absorption, and inhalation, with ingestion being the most common route of exposure [23,24,25]. A wide array of products used in everyday life contain BPA, including plastic containers, thermal papers, food cans, and beverage cans [26,27,28]. Residual BPA can leach from these products due to incomplete polymerization during production or from depolymerization when exposed to high temperatures or extreme pH conditions, which speed up the hydrolysis of ester bonds that link BPA monomers [6].
Due to concerns surrounding the endocrine disrupting capabilities of BPA, the European Union banned its use in all infant products beginning in 2011, and the U.S. Food and Drug Administration followed suit in 2012. However, BPA remains pervasive in our environments; of greatest concern, pregnant women are still persistently exposed to BPA in a variety of ways. The lipophilic structure of BPA allows it to readily cross cell membranes, as well as both placental and blood–brain barriers [29,30,31], enabling its ability to potentially affect the neurodevelopmental program of a growing embryo or fetus.
Despite vast evidence that BPA should be more tightly regulated, establishing safe exposure levels is complicated as is evidenced by the varied reference doses in different countries. For example, the current reference dose set by the Environmental Protection Agency in the U.S. is 50 mg/kg/day [32], the set tolerable dose intake (TDI) in Canada is 25 mg/kg/day [33], while the European Food Safety Authority (EFSA) set their TDI at 4 mg/kg/day [34].
The reference doses of BPA may be reasonable limits for adults, but the research summarized in this review and others suggest that doses in the ng/kg/day range may have deleterious impacts on the developing brains of laboratory animals [35]. In adult humans, BPA does not bioaccumulate and is metabolized and excreted within 48 h; however, given its environmental prevalence, many individuals experience chronic exposure. In addition, while BPA exposure levels are generally low in adults, averaging 0.043 mg/kg/day in Canada and 0.073 mg/kg/day in the U.S. [32], fetal, infant, and child exposure levels are much higher. In Canada, infants were found to consume up to 1.32 mg /kg/day [32], and in Europe the estimates for child BPA exposure were as high as 13 mg/kg/day [36,37]. Of particular concern regarding NDD pathophysiology, analysis of free versus conjugated BPA in human fetal samples demonstrated a reduced capacity of the fetus to metabolize BPA [38]. In this study, measurement of BPA in fetal liver samples indicated a geometric mean of 2.26 ng/g BPA/wet weight, with individual samples measuring as high as 50 ng/g (or 50 mg/kg) [38]—higher than the daily recommended exposure limitations for BPA in some countries.
Like many other EDCs, BPA can elicit non-monotonic dose responses [39,40], which yield U-shaped dose response curves instead of linear dose response curves. BPA may also cause distinct responses depending on the developmental time point of exposure. Thus, depending on the dose administered and duration of exposure, a higher dose of BPA can elicit a milder phenotype than a lower dose [22,40,41], meaning that, when a particular dose of BPA fails to elicit a negative health impact, it cannot be assumed that all lower doses are safe.
Consistent with the brain being a commonly reported target of EDCs [42], early BPA exposure is associated with behavioral impairment and NDDs in children [7,8,9]. Longitudinal studies that measured maternal BPA exposure levels during pregnancy and subsequently examined the children identified positive correlations between BPA and ADHD-related symptoms [43], learning deficits [44,45], externalizing behaviors [46], and anxiety and depression [47]. Cross-sectional and case-controlled studies examining concurrent BPA exposure levels found positive correlations between BPA exposure and both ASD [48,49] and ADHD [50,51]. A recent longitudinal study went a step further by examining both behavior and diffusion magnetic resonance images of children’s brains. In this study, analysis of 98 mother-child pairs revealed higher maternal urinary BPA concentrations at mid-gestation correlated with both internalizing problems and altered white matter microstructure when their children reached preschool age [52].
While studies examining the impact of BPA on brain structure in humans are rare, numerous studies have investigated the developmental neurotoxicity of BPA by examining the brains of animal models or cultured neurons. Here, we describe recent studies that have shed light on some of the cellular and molecular mechanisms by which BPA interferes with brain development to cause behavioral endophenotypes common to NDDs.

2. Neural Stem Cell Proliferation and Differentiation Are Affected by BPA

One of the first critical cellular processes of neurodevelopment is neurogenesis, a tightly regulated process that involves the migration, proliferation, and differentiation of neural precursor cells into functional units of circuitry [53,54]. During this process, neural stem cells (NSCs) and neural progenitor cells (NPCs) transition into neurons and glia [55]. Neurogenesis begins during embryonic brain development and continues in localized areas of the adult mammalian brain, including the subventricular zone and the dentate gyrus in the hippocampus [55,56].
Disruptions and abnormalities in neurogenesis have been implicated in various NDDs and neuropsychiatric illnesses [57,58]. Neurogenesis can be investigated at the cellular and molecular levels by evaluating proliferation rates and expression patterns of key genes and proteins during differentiation of NSCs/NPCs. Proper proliferation and differentiation of precursor cell types relies heavily on intrinsic factors, such as mitogens, small molecules, and cell signaling pathways, but are also influenced by extrinsic factors, such as environmental chemical exposures [59,60,61,62,63,64,65,66]. Indeed, maternal exposure to BPA has been studied in various model organisms and has been shown to influence the proliferative capacity of NSCs and neuronal differentiation (Table 1) [54,67,68,69,70,71,72,73,74].
As the developing nervous system is highly complex, in vitro assays using cell-based models of neurogenesis have become a valuable tool to study NSC/NPC proliferation and differentiation [75]. Following exposure to BPA, disruptions in rates of cell proliferation and neuronal differentiation have been observed in NSCs/NPCs and neuron cultures derived from the rat hippocampus [54,71], embryonic and fetal brain [68,69], the mouse brain [72], human umbilical cord blood [70], and human brain-derived cell lines [73,74] in response to various dosing regimens of BPA. In NSCs derived from the rat hippocampus and in human brain-derived cells, BPA was found to cause a significant decrease in BrdU-positive [71] and β-III tubulin-positive cells [71,74], which are biomarkers of neuronal proliferation and differentiation, respectively. In cells derived from rat embryonic and fetal brains, BPA was also found to have adverse effects on the processes of neuronal and glial maturation [68,69]. Upon exposure to concentrations of BPA ranging from 0.05 to 100 μM, decreases in neuron and oligodendrocyte maturation were observed as was an increase in astrocyte differentiation [68].
BPA exposure has also been found to affect regions of the brain important for neuronal and glial homeostasis. Rats chronically exposed to 100 μg/L BPA during prenatal development and postnatal weaning were found to have significantly enlarged lateral ventricles [76]. Lateral ventricle enlargement was speculated as being caused by a BPA-induced increase in cerebrospinal fluid (CSF) pressure and circulation [76], which could directly affect developing neurons and glia given the critical role CSF plays in maintaining brain homeostasis.
Investigators have also identified multiple molecular mechanisms by which BPA exposure disrupts the differentiation of cultured NSCs/NPCs. One such mechanism is the BPA-mediated disruption of the Wnt pathway, a signaling pathway that influences NSC/NPC development and a known contributor to the pathophysiology of NDDs and neuropsychiatric disorders [77,78]. BPA treatment was found to significantly reduce the activity of the Wnt pathway, demonstrated by altered expression of Wnt pathway genes, as well as reduced cellular β-catenin levels, reduced phosphorylation of GSK-3β (Glycogen Synthase Kinase 3 Beta), and reduced β-catenin translocation to the nucleus [54,71]. Transcription factors critical for neuronal differentiation are also impacted by BPA treatment. One study using cells derived from human umbilical cord blood showed decreased mRNA and protein levels of Sox1, Pax6, and Ngn1, and increased levels of Oct4 and Gdf3 [70]. Sox1, Pax6, and Ngn1 are actively expressed during neuronal differentiation, while expression of multipotential markers, such as Oct4 and Gdf3, is repressed [70,79]. Another analysis using human brain-derived NSCs found that concentrations of BPA as low as 1 μM resulted in a decrease in GFAP (Glial fibrillary acidic protein) and MAP2 (Microtubule associated protein 2) protein expression (markers of neuronal differentiation) as well as an increase in Nestin and Sox2 expression (markers of NSC maintenance) [73].
Although not as extensive as in vitro studies, there have also been studies investigating the impact of BPA on NSC/NPC proliferation and differentiation in vivo. For example, Tiwari et al. found that perinatal exposure in rats to 40 and 400 ug/kg/day caused a significant reduction in BrdU-positive NPCs in the hippocampus and subventricular zone (SVZ) of offspring [54]. Further, immunofluorescent labeling of lineage-specific markers showed BPA exposure caused a significant increase in GFAP-positive cells (a glial marker) and a concomitant decrease in nestin, b-tubulin, neuroD1, and doublecortin (DCX) expression (neuronal markers) [54]. This data suggests that early BPA exposure can increase glial differentiation and decrease neuronal differentiation, therein, disrupting the ratio of glia to neurons in the rat hippocampus. BPA was also shown to cause premature neurogenesis in the hypothalamus of zebrafish brains at concentrations as low as 0.0068 μM [67]. Another study using fruit flies demonstrated that embryonic and larval exposure to 1 mM BPA caused a reduction in the number of mitotically active cells in the larval brain [41].
The ability of NSCs/NPCs to proliferate and differentiate at appropriate rates is critical for neurogenesis and brain function. Both in vitro studies with cells derived from mammalian models and in vivo studies using vertebrate and invertebrate models have provided evidence that BPA exposure disrupts NSC/NPC proliferation. The delineated mechanisms involve BPA-mediated impairment of signaling pathways and gene expression critical for NSC/NPC development. Given the key role neurogenesis plays in brain development, this data demonstrates that BPA may confer a risk of NDDs by interrupting NSC/NPC development.
Table 1. Summary of studies on BPA-associated impacts on neural stem cell (NSC) development.
Table 1. Summary of studies on BPA-associated impacts on neural stem cell (NSC) development.
Neural Structure/
Cell Type
OrganismPhenotypeConcentrationReference
Central brain
(larval)
Drosophila
melanogaster
Reduced proliferation of neuroblasts1 mM
(developmental)
Nguyen et al., 2021 [41]
HypothalamusDanio rerioPremature neurogenesis0.0068 μM
(developmental)
Kinch et al., 2015 [67]
Neural progenitor cellsMus musculusHigh concentration (>100 μM) resulted in decrease in proliferation1 nM–500 μM (in vitro)Kim et al., 2007 [72]
Fetal neural stem cells Rattus
norvegicus
Increased cell proliferation;
decreased maturation of oligodendrocytes and neurons;
increased astrocyte differentiation and morphological changes;
reduced arborization by astrocytes,
oligodendrocytes, and neurons
0.05 μM, 0.25 μM,
10 μM, 50 μM, and 100 μM (in vitro)
Gill and
Kumara., 2021 [68]
Lateral ventriclesRattus
norvegicus
Enlargement of lateral ventricles100 μg/L; equivalent to 10 μg/kg/day
(perinatal)
Santoro et al., 2021 [76]
Primary neuronal cultures from embryonic rat brainsRattus
norvegicus
Reduced maturation of neural progenitor cells (at 200 µM)50, 100, or 200 μM (in vitro)Cho et al., 2018 [69]
Hippocampus and lateral ventricle (in vivo); hippocampal neural stem cells (in vitro)Rattus
norvegicus
Impaired neural stem cell proliferation and differentiation (hippocampus and subventricular zone); altered expression/protein levels of neurogenic genes (hippocampus); reduced Wnt pathway activity (hippocampus)4, 40, and 400 μg/kg/day
(perinatal)
Tiwari et al., 2015 [54]
Hippocampus (in vivo) and hippocampal neural stem cells (in vitro)Rattus
norvegicus
Inhibited hippocampal-derived neural stem cell proliferation and differentiation40 μg/kg/day
(perinatal)
Agarwal et al., 2016 [71]
Neural stem cellsHomo sapiensPromoted cell proliferation (0.1 and 1 µM); inhibited differentiation (1 µM); reduced GFAP and MAP2 expression (1 µM); increased expression of nestin and Sox2 (1 µM)0.1, 1, 5, and 10 µM
(in vitro)
Dong et al., 2021 [73]
Fetal brain-
derived neural progenitor cells
Homo sapiensReduced neuronal differentiation (decreased β III-tubulin mRNA levels and β III-tubulin-positive cells)10−16, 10−13, and 10−10 M (in vitro)Fujiwara et al., 2018 [74]
Neural stem cells from umbilical cord bloodHomo sapiensReduced NSC proliferation and differentiation50 and 100 μmol/L (in vitro)Huang et al., 2019 [70]

3. Synapse Formation Is Disrupted by BPA

The process of synapse formation, or synaptogenesis, occurs when a neuron responds to guidance cues by extending its axon toward a target cell (a neuron, gland, or muscle cell) and forms an adhesion that enables neuronal communication across a synapse. Throughout the central and peripheral nervous systems, synaptogenesis gives rise to the specialized neural networks responsible for receiving and integrating sensory stimuli in order to actuate functional responses [80,81,82]. Thus, the input–output mechanisms of the brain that facilitate appropriate behavioral responses are dependent on successful synapse formation [80,81,82].
NDDs, which are often behaviorally-defined, can be caused by disruptions in synaptogenesis [80]; therefore, elucidating the impacts of BPA on synapse formation is critical because of the implications for risk of neurodevelopmental impairment. Experimentally, synapse formation can be measured at the cellular and molecular level by evaluating axon growth and guidance [41,83,84,85], dendrite length and arborization patterns [86,87,88], and the expression levels of genes and proteins critical for the synaptogenic program [89]. BPA-associated deficits in synapse formation have been examined in the fruit fly [41,85], zebrafish [83,84], rodents [87,88,89], and embryonic stem cell-derived models from humans (Table 2) [86].
Early exposure to BPA has been found to dysregulate axon growth and guidance in fruit flies and zebrafish. In the fruit fly, developmental exposure to 0.1, 1, and 2 mM BPA caused axon guidance defects in a midbrain structure called the mushroom body [41], a neural structure critical for learning and memory [90,91]. Axonal branching has also been examined at the larval neuromuscular junction (NMJ)—a relatively accessible and highly specialized synapse between the nerve terminals of motor neurons and larval body wall muscle fibers [80,92]. Analysis of the NMJ in fruit fly larvae showed that exposure to 1 mM BPA can increase axonal branching, which corresponded to transcriptomic data indicating that BPA causes the misexpression of genes involved in axogenesis and synapse development [85]. In zebrafish, exposure to 15 mM BPA decreased the length of dorsal and ventral axons extending from secondary NMJ motoneurons [84]. A separate study using zebrafish also demonstrated a BPA-mediated inhibition of motor axon growth, this time showing a dose-dependent decrease in motor axon length following exposure to 15–90 mM during embryogenesis [83]. These studies indicate that developmental exposure to BPA can have adverse effects on axon growth and guidance.
BPA-associated impacts on synapse formation have also been measured in mammalian model organisms and cell lines [87,88,89]. Similar to the reduced axon outgrowth observed in zebrafish [83,84], BPA treatment of human embryonic stem cell-derived NSCs led to a reduction in neurite length [86]. In contrast, analysis of dendritogenesis in cultured rat hippocampal neurons found treatment with 10–100 nM BPA increased the motility and density of dendritic filipodia, as well as dendrite length [87]. Studies focused on synaptic morphology have revealed BPA can also affect synaptogenesis after the presynaptic and postsynaptic membranes come into contact. Analysis of the hippocampus in male mice following perinatal BPA exposure (0.4–4 mg/kg/day) revealed altered structural characteristics of synapses, including enlarged synaptic clefts, reduced active zones, thinned postsynaptic densities, and increased synaptic curvature [88]. Cultured mouse neuroblasts (Neuro-2a/N2a cells) exhibited cell shrinkage and a reduced number of synapses upon exposure to BPA ranging from 50 μM to 200 μM [89]. BPA also caused reduced expression of Dbn (Drebin), MAP2, and Tau, and increased expression of SYP (Synaptophysin) in N2a cells [90]. Dbn and SYP regulate synaptic morphology and MAP2 and Tau are critical for maintaining stability of the neuronal cytoskeleton. Thus, the BPA-induced misexpression of these critical synaptic proteins provides a molecular explanation for its deleterious impacts on synapse formation and synaptic integrity.
Studies using invertebrate and vertebrate models, in addition to cultured cells, provide evidence that BPA exposure can impair synapse formation by disrupting axogenesis, dendritogenesis, and synaptic structural integrity. BPA has been found to impact the expression of genes critical for synapse formation in invertebrate and mammalian systems [86,90], suggesting that BPA interferes with synaptogenesis at least in part through its dysregulation of neurodevelopmental gene expression.

4. Synaptic Plasticity Is Impaired by BPA

Synaptic plasticity—the ability of neurons to modify the strength of their connections in an activity-dependent manner—is the foundation of learning and memory, adaptive social, and emotional behavior and is thought to be a common feature of many NDDs [93,94,95,96,97]. Synaptic plasticity can be measured at the cellular level by assessing dendritic spines, small protrusions along the dendritic membrane where synapses form. The density of dendritic spines changes in response to neural activity; long-term potentiation (LTP) increases spine density [98], whereas long-term depression (LTD) reduces spine density [99].
Chemical exposures can disrupt synaptic plasticity by altering the expression or activity of proteins important for either structural changes in dendritic spines or neuronal signaling pathways that contribute to LTP and LTD. Maternal exposure to BPA has been found to affect the synaptic plasticity of offspring in various brain regions by reducing dendritic spine density [100,101,102], altering LTP and LTD [103,104], and dysregulating the expression of molecular regulators of plasticity (Table 3) [88,101,102,103,104,105,106].
The impact of BPA on synaptic plasticity in the hippocampus has been extensively investigated because of the key role hippocampal plasticity plays in learning and memory [96]. Reductions in spine density have been observed in the hippocampus of non-human primates [107], rats [100,101,105,106], and mice [88,108] following prenatal or perinatal exposure to BPA at levels ranging from a very low dose (30 μg/kg/day) up to a high dose (50 mg/kg/day). Analysis of potential molecular causes of reduced hippocampal spine density suggests BPA can downregulate critical synaptic proteins, including presynaptic synapsin I [88,104], postsynaptic density protein 95 (PSD-95) [88,105,106,109], N-methyl-D-aspartate (NMDA) receptor subunits [88,105,106,110], α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor subunits [105,106], and activity-regulated cytoskeleton-associated protein (Arc), as well as reduced PKC/ERK/CREB signaling [105]. While most of these studies exclusively used male animals, two studies that included both sexes reported different impacts in females and males (males were found to have a more robust decrease in spines) [100,105], one study found BPA-associated effects on plasticity were mediated by ER-a [105], while another study suggested plasticity phenotypes were sex-independent because BPA-associated impacts were indistinguishable in male and female brains [101]. Although there remains some debate about estrogen signaling being involved in BPA-mediated consequences in the central nervous system, all of the referenced studies found that exposure to BPA during development reduced plasticity in the hippocampus, therein, supporting the notion that maternal BPA exposure can cause learning and memory deficits in offspring.
BPA-associated changes in synaptic plasticity have also been identified in the primary visual cortex [102], basal ganglia [104], and basolateral amygdala (BLA) [103] of rats. In the primary visual cortex, reductions in spine density were observed in response to low BPA exposure levels (1 mg/kg/day) [102]. In this case, the reduced spine density was connected to diminished expression of the proinflammatory cytokine, interleukin 1β (IL-1β) [102], which plays a role in LTP [111]. In the basal ganglia, low dose BPA (20 μg/kg/day) caused deficits in the development of LTP and LTD at the dorsolateral striatum via enhancement of dopamine receptor (D1R) activity, a disruption suggested by the authors to diminish control of motor behaviors [104]; though, it is worth noting the basal ganglia is also critical for non-motor functions, such as emotion and executive function [112]. In the BLA, low dose BPA exposure also caused increased dopaminergic signaling, which triggered elevated LTP in the cortical-BLA pathway [103], neuronal excitability thought to underpin hyperactivity and attention deficits.
Although this review does not focus on adult exposure, numerous studies have found postnatal BPA treatment also has deleterious impacts on synaptic plasticity—typically in the form of reduced dendritic spine density—in the prefrontal cortex and hippocampus of non-human primates [113,114], rats [115,116,117,118,119], and mice [120]. The ability of BPA to consistently impair synaptic plasticity in a variety of brain structures critical for cognition and behavior in mammalian models, including non-human primates, rats, and mice, provides compelling evidence that BPA is a risk factor for NDDs in humans, given that aberrant neuroplasticity is a hallmark of these disorders.
Table 3. Summary of studies that have investigated the impact of BPA on synaptic plasticity.
Table 3. Summary of studies that have investigated the impact of BPA on synaptic plasticity.
Brain RegionOrganismPhenotypeExposureReference
HippocampusMus musculus
(males only)
Downregulated expression of PSD95 and synaptophysin; upregulated gephyrin (inhibitory); reduced excitatory to inhibitory protein ratio 50 μg/kg/day (perinatal)Kumar and Thakur, 2017 [109]
Mus musculus
(males only)
Reduced spine density40 or 400 μg/kg/day
(prenatal)
Kimura et al., 2016 [108]
Mus musculus
(males only)
Reduced synapsin I, PSD-95, NMDA receptor subunit NR1, AMPA receptor subunit GluR10.04, 0.4, or 4.0 mg/kg/day
(perinatal)
Xu et al., 2013 [88]
Mus musculus
(males only)
Downregulated NMDA receptor subunits NR1, NR2A, and 2B50, 5, 0.5, or 0.05 mg/kg/day (perinatal) Xu et al., 2010 [110]
Rattus norvegicusReduced spine density in males; increased spine density in females at estrus, but reduced spine density at proestrus30 ug/kg/day (perinatal) Kawato et al., 2021 [100]
Rattus norvegicusDownregulated expression of p-NR2B, NR2B, p-GluA1, GluA1, PSD-95, synapsin I, PKC, p-ERK and p-CREB in males and females (greater reduction in males)1 and 10 μg/mL, equivalent to 0.14 or 1.4 mg/kg/day (perinatal)Wu et al., 2020 [105]
Rattus norvegicusReduced spine density; increased mIPSC amplitude; reduced Arc (activity-regulated cytoskeleton-associated protein) expression 0.15–7.5 mg/kg/day (prenatal and postnatal, through PND 87)Liu et al., 2016 [101]
Rattus norvegicus (males only)Reduced expressions of synaptophysin, PSD-95, spinophilin, GluR1 and NMDAR10.05, 0.5, 5, or 50 mg/kg/day (perinatal)Wang et al., 2014 [106]
Macaca mulatta (females only)Reduced spine synapses in CA1, but not PFC125 mg delivered subcutaneously to pregnant females or 50 days (resulted in mean serum level of 0.91 ± 0.13 ng/mL)Elsworth et al., 2013 [107]
Primary visual cortex (V1)Rattus norvegicus (males only)Reduced spine density and maturity; decreased interleukin 1β (IL-1β) expression; reduced P38 phosphorylation1 mg/kg/day (perinatal and neonatal)Hu et al., 2020 [102]
Basal ganglia (dorsal striatum)Rattus norvegicus (males only)Caused deficits in development of LTP and LTD at dorsolateral striatum; dysregulated dopaminergic signaling (D1R and D2R)20 μg/kg/day (perinatal and neonatal)Zhou et al., 2009 [104]
Basolateral amygdala
(BLA)
Rattus norvegicus (males only)Increased neuronal excitability and facilitation of LTP induction in cortical-BLA pathway; GABAergic disinhibition; dopaminergic enhancement2 μg/kg/day
(perinatal)
Zhou et al., 2011 [103]

5. Behavior Is Impacted by BPA

Disruptions in neural development can have lasting effects on animal behavior. Consistent with the extensive impacts BPA elicits on the developing brain of laboratory animals, a variety of behavioral aberrations have been attributed to BPA exposure. Notably, many of the predominant BPA-associated behavioral outcomes are common endophenotypes of neurodevelopmental disorders, such as ADHD, ASD, and ID, including increased locomotor activity (hyperactivity), deficits in learning and memory, and increased anxiety-like behavior (Table 4). There are also studies indicating BPA causes contrasting phenotypes for each behavior; as with any toxicology study, these disparities may be due to differences in exposure regimen (dose, mode, and duration of exposure), genetic composition of the organism (even subtle genetic differences can alter an organism’s response to environmental toxicants [121]), or experimental design, including differences in the age at which behavioral analysis occurred.
Despite differences in animal model and experimental design, studies examining locomotor activity have largely found developmental exposure to BPA gives rise to hyperactive progeny. At least eight separate studies using fruit flies [41,122,123], zebrafish [67,124,125], mice [126], and rats [103], found that when developing organisms were exposed to BPA, they exhibited increased locomotor activity as larvae/juveniles and/or as adults. In one study, BPA-associated hyperactivity was sex-specific and only observed in female mice [126], yet another study that solely examined locomotor activity in male rats detected hyperactive behavior [101]. Of the investigations attempting to delineate the cellular or molecular underpinnings of locomotor changes, studies using Drosophila and zebrafish attributed hyperactivity to BPA-mediated transcriptional changes in neurodevelopmental [85,101] and metabolic pathways [101]. Another group attributed the BPA-associated hyperactivity in rats to increased neuronal excitability in the cortical-basolateral amygdala (BLA) pathway [103]. Studies of human brains found individuals with ADHD have altered connectivity between the amygdala and prefrontal cortex, in addition to smaller amygdalae [127]. Thus, the finding that BPA impacts the cortical-BLA pathway in rats points to a potential pathophysiological mechanism underlying the association between BPA exposure and ADHD in humans [51]. In contrast to the studies reporting BPA-associated hyperactivity, three studies using C. elegans (in a head thrashing test) [128], zebrafish (in swimming activity tests) [84], and rats (in an open-field test) [106] found BPA exposure caused hypoactivity.
Learning and memory involves the acquisition of information in response to environmental stimuli, which is encoded and stored in the brain for future retrieval. At least 14 studies have found early BPA exposure causes learning and memory deficits in offspring, in agreement with the many reports on BPA-associated diminishment of synaptogenesis and synaptic plasticity in the mammalian hippocampus described in preceding sections of this review. Avoidance learning and memory were found to be affected in female mice (only females were tested) [129], male mice (only males were tested) [110], and in male rats (both sexes were tested) [105]. Spatial learning and memory were disrupted by BPA in organisms ranging from zebrafish [125], mice [110], deer mice [130], and rats [101,105,106,131,132,133,134,135,136]; some studies reported sex-specific differences [130,131,135], some only tested one sex [84,132], while others found no difference on the impact in male and female rodents [101,105,125,126,133]. Object recognition learning and memory in rodents was also impaired by BPA [135,136]. In contrast, contextual fear memory was enhanced in female mice exposed to BPA [137]. In Drosophila, learning was found to be disrupted in males (only males were tested) in an associative learning paradigm called conditioned courtship suppression [85]; while flies do not have a hippocampus, this finding is consistent with BPA-mediated axon guidance defects of the mushroom body [41].
Some of the proposed underlying molecular mechanisms of BPA-mediated learning and memory impairments in rodents include modulation of PKC/ERK and BDNF/CREB signaling cascades [105,129,135], and altered expression of NMDA receptor subunits [105,106,110], AMPA receptor subunits [106], and critical pre/post-synaptic proteins [105,106], all of which can interfere with normal synaptic function. Finally, while the reasons for the disparities are unclear, some investigations found BPA had no impact on learning and memory in rodents, including two studies that reported no observed effects on spatial learning and memory [138,139], and another that found avoidance learning was intact [140].
Studies aimed at determining whether BPA causes anxiety have predominantly found developmental exposure leads to increased anxiety-like behaviors in offspring, often in a sex-specific manner. Three investigations using rodents of both sexes identified an increase in anxiety-like behaviors in males but not females [131,140,141]. Another study exclusively used male rodents and found similar BPA-mediated increases in anxiety-like behavior [109]. Only one study noted anxiety was increased in female rodents exposed to BPA but not males [133], and one investigation found that BPA led to a reduction in anxiety in both sexes [134]. These two studies used a Y-maze test, while all of the aforementioned studies used the open field test and/or elevated plus maze test to measure anxiety-like behavior; thus, the discrepancy may relate to the experimental modality and suggests using multiple paradigms within each study is warranted. Of the groups that identified BPA-associated anxiety-like behavior, Kumar et al. found perinatal exposure to BPA alters the ratio of excitatory to inhibitory synaptic densities [109]. Imbalances in neural excitation and inhibition have been reported in the pathophysiology of many neuropsychiatric disorders, including ASD, ADHD, schizophrenia, and epilepsy [142,143,144]. Thus, if BPA can indeed disrupt the excitation/inhibition balance, developmental exposure poses risk for numerous mental disorders.
Several recent reviews have also discussed neurobehavioral ramifications of BPA exposure in animal models [145,146,147,148,149]. Although not reviewed here, many other behaviors have been associated with early BPA exposure, including visual perception [85,102], depression-like behavior [140,150], and social and reproductive behaviors [141,146,151].
Table 4. Summary of studies that have investigated the impact of BPA on animal behavior.
Table 4. Summary of studies that have investigated the impact of BPA on animal behavior.
BehaviorOrganismPhenotypeExposureReference
Locomotor
Behavior
Caenorhabditis
elegans
Reduced activity0.01–10 mM
(developmental)
Zhou et al., 2016 [128]
Drosophila
melanogaster
Increased activity0.1–1 mM
(developmental)
Musachio et al., 2021 [122]
Drosophila
melanogaster
Increased activity0.1–1 mM
(developmental)
Nguyen et al., 2021 [41]
Drosophila
melanogaster
Increased activity0.1–1 mM
(developmental)
Kaur et al., 2015 [123]
Danio rerioIncreased activity
(specifically in
response to 0.001 μM BPA)
0.1 nM to 30 μM
(developmental)
Olsvik et al., 2019 [124]
Danio rerioIncreased activity0.01, 0.1, or 1 μM
(developmental)
Saili et al., 2012 [125]
Danio rerioIncreased activity0.1 or 1 μM BPA
(developmental)
Kinch et al., 2015 [67]
Danio rerioReduced activity1, 5, or 15 μM
(developmental)
Wang et al., 2013 [84]
Mus musculusIncreased activity
in females
(not affected in males)
50 ng, 50 μg, or 50 mg BPA/kg/day (perinatal)Anderson et al., 2013 [126]
Rattus norvegicus (males only)Increased activity2 μg/kg/day
(perinatal)
Zhou et al., 2011 [103]
Rattus norvegicus (males only)Reduced activity0.05, 0.5, 5, or 50 mg/kg/day
(perinatal)
Wang et al., 2014 [106]
Learning & MemoryDrosophila
melanogaster
(males only)
Impaired associative
learning
1 mM
(developmental)
Welch et al., 2022 [85]
Danio rerioImpaired learning0.01, 0.1, or 1 μM
(developmental)
Saili et al., 2012 [125]
Mus musculusEnhanced fear memory in
females; no observed effect in males
250 ng/kg/day
(perinatal)
Matsuda et al., 2013 [137]
Mus musculus
(females only)
Impaired memory retention0.1–10 mg/kg/day
(prenatal)
Jang et al., 2012 [129]
Mus musculusNo observed effect on spatial learning and memory20 μg/kg/day
(perinatal)
Nakamura et al., 2012 [138]
Mus musculus
(males only)
Impaired spatial and
avoidance memory
0.05–50 mg/kg/day
(perinatal)
Xu et al., 2010 [110]
Peromyscus
maniculatus
(deer mice)
Impaired spatial learning in males at 5 and 50 mg/kg,
no observed
effect in females
One or three doses of BPA at 50 μg, 5 mg, and 50 mg/kg feed weightJašarević et al., 2012 [130]
Rattus norvegicusImpaired spatial and
recognition memory
in males and females;
Impaired passive
avoidance memory in males
1 and 10 μg/mL, equivalent to 0.14 or 1.4 mg/kg/day
(perinatal)
Wu et al., 2020 [105]
Rattus norvegicus (males only)Impaired object
recognition memory
0.05, 0.5, 5, or 50 mg/kg/day (prenatal)Wang et al., 2016 [135]
Rattus norvegicusImpaired spatial memory in both males and females0.15–7.5 mg/kg/day
(prenatal and postnatal, through
PND 87)
Liu et al., 2016 [101]
Rattus norvegicusImpaired spatial recognition learning and memory in females at 2500 μg/kg/day2.5 μg, 25 μg, and 2500 μg/kg/day
(perinatal)
Johnson et al., 2016 [131]
Rattus norvegicusAltered spatial learning of
females at 25 μg/kg/day
(masculinization of
female brain)
0, 25 μg, 250 μg, 5 mg, or 50 mg/kg/day
(perinatal)
Hass et al., 2016 [136]
Rattus norvegicus (males only)Impaired working and
reference memory
0.05, 0.5, 5, or 50 mg/kg/day
(perinatal)
Wang et al., 2014 [106]
Rattus norvegicus (males only)Impaired spatial memory2.5 mg/kg/day
(perinatal)
Xu et al., 2014 [132]
Rattus norvegicusImpaired spatial memory in both males and females40 ug/kg/day
(perinatal)
Poimenova et al., 2010 [133]
Rattus norvegicusNo effect on avoidance
learning
15 μg/kg/day
(prenatal)
Fujimoto et al., 2006 [140]
Rattus norvegicusImpaired working
memory and object
recognition memory
100 or 500 μg/kg/day
(perinatal)
Tian et al., 2010 [134]
Anxiety-Like
Behavior
Mus musculus
(males only)
Increased50 μg/kg/day
(perinatal)
Kumar and Thakur, 2017 [109]
Mus musculusIncreased in males,
no effect in
females
50 mg/kg/day
(prenatal)
Cox et al., 2010 [141]
Peromyscus maniculatus
(deer mice)
Increased in males at
5 and 50 mg/kg,
no observed effect
in females
One or three doses of BPA at 50 μg, 5 mg, and 50 mg/kg feed weightJašarević et al., 2012 [130]
Rattus norvegicusIncreased in females,
no effect in males
40 ug/kg/day
(perinatal)
Poimenova et al., 2010 [133]
Rattus norvegicusNo observed effect0.15, 1.5, 75, 750, and 2250 ppm (perinatal)Stump et al., 2010 [139]
Rattus norvegicusReduced anxiety100 or 500 μg/kg/day (perinatal)Tian et al., 2010 [134]
Rattus norvegicusIncreased in males,
no effect in females
15 μg/kg/day
(prenatal)
Fujimoto et al., 2006 [140]

6. Conclusions

This review described the neurodevelopmental impacts—molecular, cellular, and behavioral—resulting from early exposure to BPA in organisms spanning the animal kingdom, from invertebrates to mammals. While the laboratory animals used in these studies largely served as models for understanding human impacts of BPA exposure, it should be emphasized that BPA clearly impaired the neurodevelopment of organisms throughout our environment, suggesting broad implications for the ecosystem.
Behavioral changes caused by BPA in animal models are largely consistent with human behaviors associated with BPA exposure, including hyperactivity, learning deficits, and anxiety-like behavior. In animal models, the neurodevelopmental impacts associated with both low and high dose exposure to BPA during development are vast, beginning with interruptions in NSC development and extending to axon growth and guidance defects, impaired dendritogenesis, and altered synaptic plasticity. The findings presented in this review collectively indicate that developmental exposure to BPA should be regarded as a risk factor for NDDs in humans.

Author Contributions

C.W. and K.M. both made substantial contributions to the writing and editing of this review. All authors have read and agreed to the published version of the manuscript.

Funding

Publication charges were supported by the National Institute of General Medical Sciences of the National Institutes of Health under award number 5 SC2 GM132005.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zablotsky, B.; Black, L.I.; Maenner, M.J.; Schieve, L.A.; Danielson, M.L.; Bitsko, R.H.; Blumberg, S.J.; Kogan, M.D.; Boyle, C.A. Prevalence and Trends of Developmental Disabilities among Children in the United States: 2009–2017. Pediatrics 2019, 144, e20190811. [Google Scholar] [CrossRef] [PubMed]
  2. Baio, J.; Wiggins, L.; Christensen, D.L.; Maenner, M.J.; Daniels, J.; Warren, Z.; Kurzius-Spencer, M.; Zahorodny, W.; Robinson, C.R.; Rosenberg, C.R.; et al. Prevalence of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2014. MMWR. Surveill. Summ. 2018, 67, 1–23. [Google Scholar] [CrossRef] [PubMed]
  3. Maenner, M.J.; Shaw, K.A.; Baio, J.; Washington, A.; Patrick, M.; DiRienzo, M.; Christensen, D.L.; Wiggins, L.D.; Pettygrove, S.; Andrews, J.G.; et al. Prevalence of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2016. Morb. Mortal. Wkly. Rep. Surveill. Summ. 2020, 69, 1–12. [Google Scholar] [CrossRef] [PubMed]
  4. Herbert, M.R. Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders. Curr. Opin. Neurol. 2010, 23, 103–110. [Google Scholar] [CrossRef]
  5. Matelski, L.; Van de Water, J. Risk factors in autism: Thinking outside the brain. J. Autoimmun. 2016, 67, 1–7. [Google Scholar] [CrossRef] [Green Version]
  6. Almeida, S.; Raposo, A.; Almeida-González, M.; Carrascosa, C. Bisphenol A: Food Exposure and Impact on Human Health. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1503–1517. [Google Scholar] [CrossRef] [Green Version]
  7. De Cock, M.; Maas, Y.G.; Van De Bor, M. Does perinatal exposure to endocrine disruptors induce autism spectrum and attention deficit hyperactivity disorders? Review. Acta Paediatr. 2012, 101, 811–818. [Google Scholar] [CrossRef]
  8. Minatoya, M.; Kishi, R. A Review of Recent Studies on Bisphenol A and Phthalate Exposures and Child Neurodevelopment. Int. J. Environ. Res. Public Health 2021, 18, 3585. [Google Scholar] [CrossRef]
  9. Mustieles, V.; Fernández, M.F. Bisphenol A shapes children’s brain and behavior: Towards an integrated neurotoxicity assessment including human data. Environ. Health 2020, 19, 66. [Google Scholar] [CrossRef]
  10. Wetherill, Y.B.; Akingbemi, B.T.; Kanno, J.; McLachlan, J.A.; Nadal, A.; Sonnenschein, C.; Watson, C.S.; Zoeller, R.T.; Belcher, S.M. In vitro molecular mechanisms of bisphenol A action. Reprod. Toxicol. 2007, 24, 178–198. [Google Scholar] [CrossRef]
  11. Bonefeld-Jørgensen, E.C.; Long, M.; Hofmeister, M.V.; Vinggaard, A.M. Endocrine-Disrupting Potential of Bisphenol A, Bisphenol A Dimethacrylate, 4-N-Nonylphenol, and 4-N-Octylphenolin Vitro: New Data and a Brief Review. Environ. Health Perspect. 2007, 115, 69–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Moriyama, K.; Tagami, T.; Akamizu, T.; Usui, T.; Saijo, M.; Kanamoto, N.; Hataya, Y.; Shimatsu, A.; Kuzuya, H.; Nakao, K. Thyroid Hormone Action Is Disrupted by Bisphenol A as an Antagonist. J. Clin. Endocrinol. Metab. 2002, 87, 5185–5190. [Google Scholar] [CrossRef] [PubMed]
  13. Sun, H.; Shen, O.-X.; Wang, X.-R.; Zhou, L.; Zhen, S.-Q.; Chen, X.-D. Anti-thyroid hormone activity of bisphenol A, tetrabromobisphenol A and tetrachlorobisphenol A in an improved reporter gene assay. Toxicol. Vitr. 2009, 23, 950–954. [Google Scholar] [CrossRef]
  14. Wang, J.; Sun, B.; Hou, M.; Pan, X.; Li, X. The environmental obesogen bisphenol A promotes adipogenesis by increasing the amount of 11β-hydroxysteroid dehydrogenase type 1 in the adipose tissue of children. Int. J. Obes. 2012, 37, 999–1005. [Google Scholar] [CrossRef] [PubMed]
  15. Prasanth, G.K.; Divya, L.M.; Sadasivan, C. Bisphenol-A can bind to human glucocorticoid receptor as an agonist: An in silico study. J. Appl. Toxicol. 2010, 30, 769–774. [Google Scholar] [CrossRef] [PubMed]
  16. Atlas, E.; Pope, L.; Wade, M.G.; Kawata, A.; Boudreau, A.; Boucher, J.G. Bisphenol A increases aP2 expression in 3T3L1 by enhancing the transcriptional activity of nuclear receptors at the promoter. Adipocyte 2014, 3, 170–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Thomas, P.; Dong, J. Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: A potential novel mechanism of endocrine disruption. J. Steroid Biochem. Mol. Biol. 2006, 102, 175–179. [Google Scholar] [CrossRef]
  18. Pereira-Fernandes, A.; Demaegdt, H.; Vandermeiren, K.; Hectors, T.L.M.; Jorens, P.G.; Blust, R.; Vanparys, C. Evaluation of a Screening System for Obesogenic Compounds: Screening of Endocrine Disrupting Compounds and Evaluation of the PPAR Dependency of the Effect. PLoS ONE 2013, 8, e77481. [Google Scholar] [CrossRef] [Green Version]
  19. Somm, E.; Schwitzgebel, V.M.; Toulotte, A.; Cederroth, C.R.; Combescure, C.; Nef, S.; Aubert, M.L.; Hüppi, P.S. Perinatal Exposure to Bisphenol A Alters Early Adipogenesis in the Rat. Environ. Health Perspect. 2009, 117, 1549–1555. [Google Scholar] [CrossRef] [Green Version]
  20. Vogel, S.A. The Politics of Plastics: The Making and Unmaking of Bisphenol A “Safety”. Am. J. Public Health 2009, 99, S559–S566. [Google Scholar] [CrossRef]
  21. Dodds, E.C.; Lawson, W. Synthetic strogenic Agents without the Phenanthrene Nucleus. Nature 1936, 137, 996. [Google Scholar] [CrossRef]
  22. Vandenberg, L.N.; Maffini, M.V.; Sonnenschein, C.; Rubin, B.S.; Soto, A.M. Bisphenol-A and the Great Divide: A Review of Controversies in the Field of Endocrine Disruption. Endocr. Rev. 2009, 30, 75–95. [Google Scholar] [CrossRef] [PubMed]
  23. Nerín, C.; Fernández, C.; Domeño, C.; Salafranca, J. Determination of Potential Migrants in Polycarbonate Containers Used for Microwave Ovens by High-Performance Liquid Chromatography with Ultraviolet and Fluorescence Detection. J. Agric. Food Chem. 2003, 51, 5647–5653. [Google Scholar] [CrossRef] [PubMed]
  24. Brede, C.; Fjeldal, P.; Skjevrak, I.; Herikstad, H. Increased migration levels of bisphenol A from polycarbonate baby bottles after dishwashing, boiling and brushing. Food Addit. Contam. 2003, 20, 684–689. [Google Scholar] [CrossRef] [PubMed]
  25. Konieczna, A.; Rutkowska, A.; Rachoń, D. Health risk of exposure to Bisphenol A (BPA). Rocz. Panstw. Zakl. Hig. 2015, 66, 5–11. [Google Scholar]
  26. Liao, C.; Kannan, K. Concentrations and Profiles of Bisphenol A and Other Bisphenol Analogues in Foodstuffs from the United States and Their Implications for Human Exposure. J. Agric. Food Chem. 2013, 61, 4655–4662. [Google Scholar] [CrossRef]
  27. Liao, C.; Kannan, K. A Survey of Alkylphenols, Bisphenols, and Triclosan in Personal Care Products from China and the United States. Arch. Environ. Contam. Toxicol. 2014, 67, 50–59. [Google Scholar] [CrossRef]
  28. Pivnenko, K.; Pedersen, G.A.; Eriksson, E.; Astrup, T. Bisphenol A and its structural analogues in household waste paper. Waste Manag. 2015, 44, 39–47. [Google Scholar] [CrossRef] [Green Version]
  29. Balakrishnan, B.; Henare, K.; Thorstensen, E.B.; Ponnampalam, A.P.; Mitchell, M.D. Transfer of bisphenol A across the human placenta. Am. J. Obstet. Gynecol. 2010, 202, 393.e1–393.e7. [Google Scholar] [CrossRef]
  30. Sun, Y.; Nakashima, M.N.; Takahashi, M.; Kuroda, N.; Nakashima, K. Determination of bisphenol A in rat brain by microdialysis and column switching high-performance liquid chromatography with fluorescence detection. Biomed. Chromatogr. 2002, 16, 319–326. [Google Scholar] [CrossRef]
  31. Nishikawa, M.; Iwano, H.; Yanagisawa, R.; Koike, N.; Inoue, H.; Yokota, H. Placental Transfer of Conjugated Bisphenol A and Subsequent Reactivation in the Rat Fetus. Environ. Health Perspect. 2010, 118, 1196–1203. [Google Scholar] [CrossRef] [PubMed]
  32. LaKind, J.S.; Levesque, J.; Dumas, P.; Bryan, S.N.; Clarke, J.; Naiman, D.Q. Comparing United States and Canadian population exposures from National Biomonitoring Surveys: Bisphenol A intake as a case study. J. Expo. Sci. Environ. Epidemiol. 2012, 22, 219–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Eladak, S.; Grisin, T.; Moison, D.; Guerquin, M.-J.; N’Tumba-Byn, T.; Pozzi-Gaudin, S.; Benachi, A.; Livera, G.; Rouiller-Fabre, V.; Habert, R. A new chapter in the bisphenol A story: Bisphenol S and bisphenol F are not safe alternatives to this compound. Fertil. Steril. 2015, 103, 11–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hessel, E.V.; Ezendam, J.; van Broekhuizen, F.A.; Hakkert, B.; DeWitt, J.; Granum, B.; Guzylack, L.; Lawrence, B.P.; Penninks, A.; Rooney, A.A.; et al. Assessment of recent developmental immunotoxicity studies with bisphenol A in the context of the 2015 EFSA t-TDI. Reprod. Toxicol. 2016, 65, 448–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Negri-Cesi, P. Bisphenol A Interaction with Brain Development and Functions. Dose-Response 2015, 13, 1559325815590394. [Google Scholar] [CrossRef] [PubMed]
  36. Geens, T.; Roosens, L.; Neels, H.; Covaci, A. Assessment of human exposure to Bisphenol-A, Triclosan and Tetrabromobisphenol-A through indoor dust intake in Belgium. Chemosphere 2009, 76, 755–760. [Google Scholar] [CrossRef] [PubMed]
  37. Corrales, J.; Kristofco, L.A.; Steele, W.B.; Yates, B.S.; Breed, C.S.; Williams, E.S.; Brooks, B.W. Global Assessment of Bisphenol A in the Environment: Review and Analysis of Its Occurrence and Bioaccumulation. Dose-Response 2015, 13, 1559325815598308. [Google Scholar] [CrossRef] [Green Version]
  38. Nahar, M.S.; Liao, C.; Kannan, K.; Dolinoy, D.C. Fetal Liver Bisphenol A Concentrations and Biotransformation Gene Expression Reveal Variable Exposure and Altered Capacity for Metabolism in Humans. J. Biochem. Mol. Toxicol. 2012, 27, 116–123. [Google Scholar] [CrossRef] [Green Version]
  39. Vandenberg, L.N. Non-Monotonic Dose Responses in Studies of Endocrine Disrupting Chemicals: Bisphenol a as a Case Study. Dose-Response 2013, 12, 259–276. [Google Scholar] [CrossRef]
  40. Villar-Pazos, S.; Martinez-Pinna, J.; Castellano-Muñoz, M.; Magdalena, P.A.; Marroqui, L.; Quesada, I.; Gustafsson, J.-A.; Nadal, A. Molecular mechanisms involved in the non-monotonic effect of bisphenol-a on Ca2+ entry in mouse pancreatic β-cells. Sci. Rep. 2017, 7, 11700. [Google Scholar] [CrossRef]
  41. Nguyen, U.; Tinsley, B.; Sen, Y.; Stein, J.; Palacios, Y.; Ceballos, A.; Welch, C.; Nzenkue, K.; Penn, A.; Murphy, L.; et al. Exposure to bisphenol A differentially impacts neurodevelopment and behavior in Drosophila melanogaster from distinct genetic backgrounds. Neuro Toxicol. 2020, 82, 146–157. [Google Scholar] [CrossRef] [PubMed]
  42. Gore, A.C. Neuroendocrine targets of endocrine disruptors. Hormones 2010, 9, 16–27. [Google Scholar] [CrossRef]
  43. Casas, M.; Forns, J.; Martínez, D.; Avella-García, C.; Valvi, D.; Ballesteros-Gómez, A.; Luque, N.; Rubio, S.; Julvez, J.; Sunyer, J.; et al. Exposure to bisphenol A during pregnancy and child neuropsychological development in the INMA-Sabadell cohort. Environ. Res. 2015, 142, 671–679. [Google Scholar] [CrossRef] [PubMed]
  44. Lin, C.-C.; Chien, C.-J.; Tsai, M.-S.; Hsieh, C.-J.; Hsieh, W.-S.; Chen, P.-C. Prenatal phenolic compounds exposure and neurobehavioral development at 2 and 7 years of age. Sci. Total Environ. 2017, 605–606, 801–810. [Google Scholar] [CrossRef] [PubMed]
  45. Jensen, T.K.; Mustieles, V.; Bleses, D.; Frederiksen, H.; Trecca, F.; Schoeters, G.; Andersen, H.R.; Grandjean, P.; Kyhl, H.B.; Juul, A.; et al. Prenatal bisphenol A exposure is associated with language development but not with ADHD-related behavior in toddlers from the Odense Child Cohort. Environ. Res. 2019, 170, 398–405. [Google Scholar] [CrossRef] [PubMed]
  46. Stacy, S.; Papandonatos, G.; Calafat, A.M.; Chen, A.; Yolton, K.; Lanphear, B.P.; Braun, J.M. Early life bisphenol A exposure and neurobehavior at 8 years of age: Identifying windows of heightened vulnerability. Environ. Int. 2017, 107, 258–265. [Google Scholar] [CrossRef]
  47. Perera, F.; Nolte, E.L.R.; Wang, Y.; Margolis, A.E.; Calafat, A.M.; Wang, S.; Garcia, W.; Hoepner, L.A.; Peterson, B.S.; Rauh, V.; et al. Bisphenol A exposure and symptoms of anxiety and depression among inner city children at 10–12 years of age. Environ. Res. 2016, 151, 195–202. [Google Scholar] [CrossRef] [Green Version]
  48. Stein, T.P.; Schluter, M.D.; Steer, R.A.; Guo, L.; Ming, X. Bisphenol A Exposure in Children With Autism Spectrum Disorders. Autism Res. 2015, 8, 272–283. [Google Scholar] [CrossRef] [Green Version]
  49. Rahbar, M.H.; Swingle, H.M.; Christian, M.A.; Hessabi, M.; Lee, M.; Pitcher, M.R.; Campbell, S.; Mitchell, A.; Krone, R.; Loveland, K.A.; et al. Environmental Exposure to Dioxins, Dibenzofurans, Bisphenol A, and Phthalates in Children with and without Autism Spectrum Disorder Living near the Gulf of Mexico. Int. J. Environ. Res. Public Health 2017, 14, 1425. [Google Scholar] [CrossRef] [Green Version]
  50. Li, Y.; Zhang, H.; Kuang, H.; Fan, R.; Cha, C.; Li, G.; Luo, Z.; Pang, Q. Relationship between bisphenol A exposure and attention-deficit/ hyperactivity disorder: A case-control study for primary school children in Guangzhou, China. Environ. Pollut. 2018, 235, 141–149. [Google Scholar] [CrossRef]
  51. Tewar, S.; Auinger, P.; Braun, J.M.; Lanphear, B.; Yolton, K.; Epstein, J.N.; Ehrlich, S.; Froehlich, T.E. Association of Bisphenol A exposure and Attention-Deficit/Hyperactivity Disorder in a national sample of U.S. children. Environ. Res. 2016, 150, 112–118. [Google Scholar] [CrossRef] [PubMed]
  52. Grohs, M.N.; the APrON Study Team; Reynolds, J.E.; Liu, J.; Martin, J.; Pollock, T.; Lebel, C.; Dewey, D. Prenatal maternal and childhood bisphenol a exposure and brain structure and behavior of young children. Environ. Health 2019, 18, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Sun, D.; Zhou, X.; Yu, H.-L.; He, X.-X.; Guo, W.-X.; Xiong, W.-C.; Zhu, X.-J. Regulation of neural stem cell proliferation and differentiation by Kinesin family member 2a. PLoS ONE 2017, 12, e0179047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tiwari, S.K.; Agarwal, S.; Seth, B.; Yadav, A.; Ray, R.S.; Mishra, V.N.; Chaturvedi, R.K. Inhibitory Effects of Bisphenol-A on Neural Stem Cells Proliferation and Differentiation in the Rat Brain Are Dependent on Wnt/β-Catenin Pathway. Mol. Neurobiol. 2015, 52, 1735–1757. [Google Scholar] [CrossRef] [PubMed]
  55. Urbã¡n, N.; Guillemot, F. Neurogenesis in the embryonic and adult brain: Same regulators, different roles. Front. Cell. Neurosci. 2014, 8, 396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Shohayeb, B.; Diab, M.; Ahmed, M.; Ng, D.C.H. Factors that influence adult neurogenesis as potential therapy. Transl. Neurodegener. 2018, 7, 4. [Google Scholar] [CrossRef]
  57. Ernst, C. Proliferation and Differentiation Deficits are a Major Convergence Point for Neurodevelopmental Disorders. Trends Neurosci. 2016, 39, 290–299. [Google Scholar] [CrossRef]
  58. Stevens, H.E.; Smith, K.M.; Rash, B.; Vaccarino, F.M. Neural stem cell regulation, fibroblast growth factors, and the developmental origins of neuropsychiatric disorders. Front. Neurosci. 2010, 4, 59. [Google Scholar] [CrossRef] [Green Version]
  59. Smith, I.; Nakashima, K. Mechanisms of Neural Stem Cell Fate Determination: Extracellular Cues and Intracellular Programs. Curr. Stem Cell Res. Ther. 2006, 1, 267–277. [Google Scholar] [CrossRef]
  60. Kong, S.-Y.; Park, M.-H.; Lee, M.; Kim, J.-O.; Lee, H.-R.; Han, B.W.; Svendsen, C.N.; Sung, S.H.; Kim, H.-J. Kuwanon V Inhibits Proliferation, Promotes Cell Survival and Increases Neurogenesis of Neural Stem Cells. PLoS ONE 2015, 10, e0118188. [Google Scholar] [CrossRef] [Green Version]
  61. Longo, F.M.; Yang, T.; Xie, Y.; Massa, S.M. Small Molecule Approaches for Promoting Neurogenesis. Curr. Alzheimer Res. 2006, 3, 5–10. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, H.-J.; Rosenfeld, M.G. Epigenetic control of stem cell fate to neurons and glia. Arch. Pharmacal Res. 2010, 33, 1467–1473. [Google Scholar] [CrossRef] [PubMed]
  63. Kim, H.-J.; Jin, C.Y. Stem Cells in Drug Screening for Neurodegenerative Disease. Korean J. Physiol. Pharmacol. 2012, 16, 1–9. [Google Scholar] [CrossRef] [Green Version]
  64. Yoon, H.J.; Kong, S.-Y.; Park, M.-H.; Cho, Y.; Kim, S.-E.; Shin, J.-Y.; Jung, S.; Lee, J.; Farhanullah; Kim, H.-J.; et al. Aminopropyl carbazole analogues as potent enhancers of neurogenesis. Bioorganic Med. Chem. 2013, 21, 7165–7174. [Google Scholar] [CrossRef]
  65. Kim, H.J.; Leeds, P.; Chuang, D.-M. The HDAC inhibitor, sodium butyrate, stimulates neurogenesis in the ischemic brain. J. Neurochem. 2009, 110, 1226–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Saxe, J.P.; Wu, H.; Kelly, T.K.; Phelps, M.E.; Sun, Y.E.; Kornblum, H.I.; Huang, J. A Phenotypic Small-Molecule Screen Identifies an Orphan Ligand-Receptor Pair that Regulates Neural Stem Cell Differentiation. Chem. Biol. 2007, 14, 1019–1030. [Google Scholar] [CrossRef] [Green Version]
  67. Kinch, C.D.; Ibhazehiebo, K.; Jeong, J.-H.; Habibi, H.R.; Kurrasch, D.M. Low-dose exposure to bisphenol A and replacement bisphenol S induces precocious hypothalamic neurogenesis in embryonic zebrafish. Proc. Natl. Acad. Sci. USA 2015, 112, 1475–1480. [Google Scholar] [CrossRef] [Green Version]
  68. Gill, S.; Kumara, V. Comparative Neurodevelopment Effects of Bisphenol A and Bisphenol F on Rat Fetal Neural Stem Cell Models. Cells 2021, 10, 793. [Google Scholar] [CrossRef]
  69. Cho, J.-H.; Kim, A.H.; Lee, S.; Lee, Y.; Lee, W.J.; Chang, S.-C.; Lee, J. Sensitive neurotoxicity assessment of bisphenol A using double immunocytochemistry of DCX and MAP2. Arch. Pharmacal Res. 2018, 41, 1098–1107. [Google Scholar] [CrossRef]
  70. Huang, M.; Li, Y.; Wu, K.; Hao, S.; Cai, Q.; Zhou, Z.; Yang, H. Effects of environmental chemicals on the proliferation and differentiation of neural stem cells. Environ. Toxicol. 2019, 34, 1285–1291. [Google Scholar] [CrossRef]
  71. Agarwal, S.; Yadav, A.; Tiwari, S.K.; Seth, B.; Chauhan, L.K.S.; Khare, P.; Ray, R.S.; Chaturvedi, R.K. Dynamin-related Protein 1 Inhibition Mitigates Bisphenol A-mediated Alterations in Mitochondrial Dynamics and Neural Stem Cell Proliferation and Differentiation. J. Biol. Chem. 2016, 291, 15923–15939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Kim, K.; Son, T.G.; Kim, S.J.; Kim, H.S.; Kim, T.S.; Han, S.Y.; Lee, J. Suppressive Effects of Bisphenol A on the Proliferation of Neural Progenitor Cells. J. Toxicol. Environ. Health Part A 2007, 70, 1288–1295. [Google Scholar] [CrossRef] [PubMed]
  73. Dong, P.; Ye, G.; Tu, X.; Luo, Y.; Cui, W.; Ma, Y.; Wei, L.; Tian, X.; Wang, Q. Roles of ERRα and TGF-β signaling in stemness enhancement induced by 1 µM bisphenol A exposure via human neural stem cells. Exp. Ther. Med. 2021, 23, 164. [Google Scholar] [CrossRef] [PubMed]
  74. Fujiwara, Y.; Miyazaki, W.; Koibuchi, N.; Katoh, T. The Effects of Low-Dose Bisphenol A and Bisphenol F on Neural Differentiation of a Fetal Brain-Derived Neural Progenitor Cell Line. Front. Endocrinol. 2018, 9, 24. [Google Scholar] [CrossRef] [Green Version]
  75. Azari, H.; Reynolds, B.A. In Vitro Models for Neurogenesis. Cold Spring Harb. Perspect. Biol. 2015, 8, a021279. [Google Scholar] [CrossRef] [Green Version]
  76. Santoro, A.; Scafuro, M.; Troisi, J.; Piegari, G.; Di Pietro, P.; Mele, E.; Cappetta, D.; Marino, M.; De Angelis, A.; Vecchione, C.; et al. Multi-Systemic Alterations by Chronic Exposure to a Low Dose of Bisphenol A in Drinking Water: Effects on Inflammation and NAD+-Dependent Deacetylase Sirtuin1 in Lactating and Weaned Rats. Int. J. Mol. Sci. 2021, 22, 9666. [Google Scholar] [CrossRef]
  77. Mulligan, K.A.; Cheyette, B.N. Neurodevelopmental Perspectives on Wnt Signaling in Psychiatry. Mol. Neuropsychiatry 2016, 2, 219–246. [Google Scholar] [CrossRef] [Green Version]
  78. Mulligan, K.A.; Cheyette, B.N.R. Wnt Signaling in Vertebrate Neural Development and Function. J. Neuroimmune Pharmacol. 2012, 7, 774–787. [Google Scholar] [CrossRef] [Green Version]
  79. Hirabayashi, Y.; Gotoh, Y. Epigenetic control of neural precursor cell fate during development. Nat. Rev. Neurosci. 2010, 11, 377–388. [Google Scholar] [CrossRef]
  80. Batool, S.; Raza, H.; Zaidi, J.; Riaz, S.; Hasan, S.; Syed, N.I. Synapse formation: From cellular and molecular mechanisms to neurodevelopmental and neurodegenerative disorders. J. Neurophysiol. 2019, 121, 1381–1397. [Google Scholar] [CrossRef]
  81. Poggio, T. A Theory of How the Brain Might Work. Cold Spring Harb. Symp. Quant. Biol. 1990, 55, 899–910. [Google Scholar] [CrossRef] [PubMed]
  82. Taverna, E.; Götz, M.; Huttner, W.B. The Cell Biology of Neurogenesis: Toward an Understanding of the Development and Evolution of the Neocortex. Annu. Rev. Cell Dev. Biol. 2014, 30, 465–502. [Google Scholar] [CrossRef] [PubMed]
  83. Morrice, J.R.; Gregory-Evans, C.Y.; Shaw, C.A. Modeling Environmentally-Induced Motor Neuron Degeneration in Zebrafish. Sci. Rep. 2018, 8, 4890. [Google Scholar] [CrossRef] [Green Version]
  84. Wang, X.; Dong, Q.; Chen, Y.; Jiang, H.; Xiao, Q.; Wang, Y.; Li, W.; Bai, C.; Huang, C.; Yang, D. Bisphenol A affects axonal growth, musculature and motor behavior in developing zebrafish. Aquat. Toxicol. 2013, 142–143, 104–113. [Google Scholar] [CrossRef] [PubMed]
  85. Welch, C.; Johnson, E.; Tupikova, A.; Anderson, J.; Tinsley, B.; Newman, J.; Widman, E.; Alfareh, A.; Davis, A.; Rodriguez, L.; et al. Bisphenol A affects neurodevelopment gene expression, cognitive function, and neuromuscular synaptic morphology in Drosophila melanogaster. Neurotoxicology 2022, 89, 67–78. [Google Scholar] [CrossRef] [PubMed]
  86. Liang, X.; Yin, N.; Liang, S.; Yang, R.; Liu, S.; Lu, Y.; Jiang, L.; Zhou, Q.; Jiang, G.; Faiola, F. Bisphenol A and several derivatives exert neural toxicity in human neuron-like cells by decreasing neurite length. Food Chem. Toxicol. 2020, 135, 111015. [Google Scholar] [CrossRef]
  87. Xu, X.; Lu, Y.; Zhang, G.; Chen, L.; Tian, D.; Shen, X.; Yang, Y.; Dong, F. Bisphenol A promotes dendritic morphogenesis of hippocampal neurons through estrogen receptor-mediated ERK1/2 signal pathway. Chemosphere 2014, 96, 129–137. [Google Scholar] [CrossRef]
  88. Xu, X.; Xie, L.; Hong, X.; Ruan, Q.; Lu, H.; Zhang, Q.; Zhang, G.; Liu, X. Perinatal exposure to bisphenol-A inhibits synaptogenesis and affects the synaptic morphological development in offspring male mice. Chemosphere 2013, 91, 1073–1081. [Google Scholar] [CrossRef]
  89. Yin, Z.; Hua, L.; Chen, L.; Hu, D.; Li, J.; An, Z.; Tian, T.; Ning, H.; Ge, Y. Bisphenol-A exposure induced neurotoxicity and associated with synapse and cytoskeleton in Neuro-2a cells. Toxicol. Vitr. 2020, 67, 104911. [Google Scholar] [CrossRef]
  90. Van Battum, E.Y.; Brignani, S.; Pasterkamp, R.J. Axon guidance proteins in neurological disorders. Lancet Neurol. 2015, 14, 532–546. [Google Scholar] [CrossRef]
  91. McFadden, K.; Minshew, N.J. Evidence for dysregulation of axonal growth and guidance in the etiology of ASD. Front. Hum. Neurosci. 2013, 7, 671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Cruz, P.M.R.; Cossins, J.; Beeson, D.; Vincent, A. The Neuromuscular Junction in Health and Disease: Molecular Mechanisms Governing Synaptic Formation and Homeostasis. Front. Mol. Neurosci. 2020, 13, 610964. [Google Scholar] [CrossRef] [PubMed]
  93. Zoghbi, H.Y.; Bear, M.F. Synaptic Dysfunction in Neurodevelopmental Disorders Associated with Autism and Intellectual Disabilities. Cold Spring Harb. Perspect. Biol. 2012, 4, a009886. [Google Scholar] [CrossRef] [Green Version]
  94. Washbourne, P. Synapse Assembly and Neurodevelopmental Disorders. Neuropsychopharmacology 2014, 40, 4–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bourgeron, T. From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat. Rev. Neurosci. 2015, 16, 551–563. [Google Scholar] [CrossRef]
  96. Stuchlik, A. Dynamic learning and memory, synaptic plasticity and neurogenesis: An update. Front. Behav. Neurosci. 2014, 8, 106. [Google Scholar] [CrossRef] [Green Version]
  97. Davidson, R.J.; McEwen, B.S. Social influences on neuroplasticity: Stress and interventions to promote well-being. Nat. Neurosci. 2012, 15, 689–695. [Google Scholar] [CrossRef] [Green Version]
  98. Segal, M. Dendritic spines: Morphological building blocks of memory. Neurobiol. Learn. Mem. 2017, 138, 3–9. [Google Scholar] [CrossRef]
  99. Hasegawa, S.; Sakuragi, S.; Tominaga-Yoshino, K.; Ogura, A. Dendritic spine dynamics leading to spine elimination after repeated inductions of LTD. Sci. Rep. 2015, 5, 7707. [Google Scholar] [CrossRef] [Green Version]
  100. Kawato, S.; Ogiue-Ikeda, M.; Soma, M.; Yoshino, H.; Kominami, T.; Saito, M.; Aou, S.; Hojo, Y. Perinatal Exposure of Bisphenol A Differently Affects Dendritic Spines of Male and Female Grown-Up Adult Hippocampal Neurons. Front. Neurosci. 2021, 15, 712261. [Google Scholar] [CrossRef]
  101. Liu, Z.-H.; Ding, J.-J.; Yang, Q.-Q.; Song, H.-Z.; Chen, X.-T.; Xu, Y.; Xiao, G.; Wang, H.-L. Early developmental bisphenol-A exposure sex-independently impairs spatial memory by remodeling hippocampal dendritic architecture and synaptic transmission in rats. Sci. Rep. 2016, 6, 32492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Hu, F.; Zhang, L.; Li, T.; Wang, H.; Liang, W.; Zhou, Y. Bisphenol-A Exposure during Gestation and Lactation Causes Visual Perception Deficits in Rat Pups Following a Decrease in Interleukin 1β Expression in the Primary Visual Cortex. Neuroscience 2020, 434, 148–160. [Google Scholar] [CrossRef] [PubMed]
  103. Zhou, R.; Bai, Y.; Yang, R.; Zhu, Y.; Chi, X.; Li, L.; Chen, L.; Sokabe, M.; Chen, L. Abnormal synaptic plasticity in basolateral amygdala may account for hyperactivity and attention-deficit in male rat exposed perinatally to low-dose bisphenol-A. Neuropharmacology 2011, 60, 789–798. [Google Scholar] [CrossRef] [PubMed]
  104. Zhou, R.; Zhang, Z.; Zhu, Y.; Chen, L.; Sokabe, M. Deficits in development of synaptic plasticity in rat dorsal striatum following prenatal and neonatal exposure to low-dose bisphenol A. Neuroscience 2009, 159, 161–171. [Google Scholar] [CrossRef]
  105. Wu, D.; Wu, F.; Lin, R.; Meng, Y.; Wei, W.; Sun, Q.; Jia, L. Impairment of learning and memory induced by perinatal exposure to BPA is associated with ERα-mediated alterations of synaptic plasticity and PKC/ERK/CREB signaling pathway in offspring rats. Brain Res. Bull. 2020, 161, 43–54. [Google Scholar] [CrossRef]
  106. Wang, C.; Niu, R.; Zhu, Y.; Han, H.; Luo, G.; Zhou, B.; Wang, J. Changes in memory and synaptic plasticity induced in male rats after maternal exposure to bisphenol A. Toxicology 2014, 322, 51–60. [Google Scholar] [CrossRef]
  107. Elsworth, J.D.; Jentsch, J.D.; VandeVoort, C.A.; Roth, R.H., Jr. Prenatal exposure to bisphenol A impacts midbrain dopamine neurons and hippocampal spine synapses in non-human primates. Neuro Toxicol. 2013, 35, 113–120. [Google Scholar] [CrossRef] [Green Version]
  108. Kimura, E.; Matsuyoshi, C.; Miyazaki, W.; Benner, S.; Hosokawa, M.; Yokoyama, K.; Kakeyama, M.; Tohyama, C. Prenatal exposure to bisphenol A impacts neuronal morphology in the hippocampal CA1 region in developing and aged mice. Arch. Toxicology 2016, 90, 691–700. [Google Scholar] [CrossRef] [Green Version]
  109. Kumar, D.; Thakur, M. Anxiety like behavior due to perinatal exposure to Bisphenol-A is associated with decrease in excitatory to inhibitory synaptic density of male mouse brain. Toxicology 2017, 378, 107–113. [Google Scholar] [CrossRef]
  110. Xu, X.-H.; Zhang, J.; Wang, Y.-M.; Ye, Y.-P.; Luo, Q.-Q. Perinatal exposure to bisphenol-A impairs learning-memory by concomitant down-regulation of N-methyl-d-aspartate receptors of hippocampus in male offspring mice. Horm. Behav. 2010, 58, 326–333. [Google Scholar] [CrossRef]
  111. Schneider, H.; Pitossi, F.; Balschun, D.; Wagner, A.; del Rey, A.; Besedovsky, H.O. A neuromodulatory role of interleukin-1 in the hippocampus. Proc. Natl. Acad. Sci. USA 1998, 95, 7778–7783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Eisinger, R.; Urdaneta, M.; Foote, K.; Okun, M.S.; Gunduz, A. Non-motor Characterization of the Basal Ganglia: Evidence From Human and Non-human Primate Electrophysiology. Front. Neurosci. 2018, 12, 385. [Google Scholar] [CrossRef] [PubMed]
  113. Leranth, C.; Hajszan, T.; Szigeti-Buck, K.; Bober, J.; MacLusky, N. Bisphenol A prevents the synaptogenic response to estradiol in hippocampus and prefrontal cortex of ovariectomized nonhuman primates. Proc. Natl. Acad. Sci. USA 2008, 105, 14187–14191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Elsworth, J.D.; Jentsch, J.D.; Groman, S.M.; Roth, R.H., Jr.; Leranth, C. Low circulating levels of bisphenol-A induce cognitive deficits and loss of asymmetric spine synapses in dorsolateral prefrontal cortex and hippocampus of adult male monkeys. J. Comp. Neurol. 2015, 523, 1248–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Leranth, C.; Szigeti-Buck, K.; MacLusky, N.; Hajszan, T. Bisphenol A Prevents the Synaptogenic Response to Testosterone in the Brain of Adult Male Rats. Endocrinology 2007, 149, 988–994. [Google Scholar] [CrossRef] [Green Version]
  116. Eilam-Stock, T.; Serrano, P.; Frankfurt, M.; Luine, V. Bisphenol-A impairs memory and reduces dendritic spine density in adult male rats. Behav. Neurosci. 2012, 126, 175–185. [Google Scholar] [CrossRef] [Green Version]
  117. Inagaki, T.; Frankfurt, M.; Luine, V. Estrogen-Induced Memory Enhancements Are Blocked by Acute Bisphenol A in Adult Female Rats: Role of Dendritic Spines. Endocrinology 2012, 153, 3357–3367. [Google Scholar] [CrossRef] [Green Version]
  118. Bowman, R.E.; Hagedorn, J.; Madden, E.; Frankfurt, M. Effects of adolescent Bisphenol-A exposure on memory and spine density in ovariectomized female rats: Adolescence vs. adulthood. Horm. Behav. 2019, 107, 26–34. [Google Scholar] [CrossRef]
  119. Huarui, G.; Li, T.; Gong, H.; Chen, Z.; Jin, Y.; Xu, G.; Wang, M. Bisphenol A Impairs Synaptic Plasticity by Both Pre- and Postsynaptic Mechanisms. Adv. Sci. 2017, 4, 1600493. [Google Scholar] [CrossRef] [Green Version]
  120. Lee, C.Y.; Hyun, S.-A.; Ko, M.Y.; Kim, H.R.; Rho, J.; Kim, K.K.; Kim, W.-Y.; Ka, M. Maternal Bisphenol A (BPA) Exposure Alters Cerebral Cortical Morphogenesis and Synaptic Function in Mice. Cereb. Cortex 2021, 31, 5598–5612. [Google Scholar] [CrossRef]
  121. Aldridge, J.E.; Gibbons, J.A.; Flaherty, M.M.; Kreider, M.L.; Romano, J.A.; Levin, E.D. Heterogeneity of Toxicant Response: Sources of Human Variability. Toxicol. Sci. 2003, 76, 3–20. [Google Scholar] [CrossRef] [PubMed]
  122. Musachio, E.A.S.; Couto, S.D.F.; Poetini, M.R.; Bortolotto, V.C.; Dahleh, M.M.M.; Janner, D.E.; Araujo, S.M.; Ramborger, B.P.; Rohers, R.; Guerra, G.P.; et al. Bisphenol A exposure during the embryonic period: Insights into dopamine relationship and behavioral disorders in Drosophila melanogaster. Food Chem. Toxicol. 2021, 157, 112526. [Google Scholar] [CrossRef] [PubMed]
  123. Kaur, K.; Simon, A.; Chauhan, V.; Chauhan, A. Effect of bisphenol A on Drosophila melanogaster behavior—A new model for the studies on neurodevelopmental disorders. Behav. Brain Res. 2015, 284, 77–84. [Google Scholar] [CrossRef] [PubMed]
  124. Olsvik, P.A.; Whatmore, P.; Penglase, S.J.; Skjærven, K.H.; D’Auriac, M.A.; Ellingsen, S. Associations Between Behavioral Effects of Bisphenol A and DNA Methylation in Zebrafish Embryos. Front. Genet. 2019, 10, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Saili, K.S.; Corvi, M.M.; Weber, D.N.; Patel, A.U.; Das, S.R.; Przybyla, J.; Anderson, K.A.; Tanguay, R.L. Neurodevelopmental low-dose bisphenol A exposure leads to early life-stage hyperactivity and learning deficits in adult zebrafish. Toxicology 2012, 291, 83–92. [Google Scholar] [CrossRef] [Green Version]
  126. Anderson, O.S.; Peterson, K.E.; Sanchez, B.N.; Zhang, Z.; Mancuso, P.; Dolinoy, D.C. Perinatal bisphenol A exposure promotes hyperactivity, lean body composition, and hormonal responses across the murine life course. FASEB J. 2013, 27, 1784–1792. [Google Scholar] [CrossRef] [Green Version]
  127. Plessen, K.J.; Bansal, R.; Zhu, H.; Whiteman, R.; Amat, J.; Quackenbush, G.A.; Martin, L.; Durkin, K.; Blair, C.; Royal, J.; et al. Hippocampus and Amygdala Morphology in Attention-Deficit/Hyperactivity Disorder. Arch. Gen. Psychiatry 2006, 63, 795–807. [Google Scholar] [CrossRef] [Green Version]
  128. Zhou, N.; Yang, J.; Li, H.; Lu, Q.; Liu, Y.-D.; Lin, K.-F. Ecotoxicological evaluation of low-concentration bisphenol A exposure on the soil nematode Caenorhabditis elegansand intrinsic mechanisms of stress response in vivo. Environ. Toxicol. Chem. 2016, 35, 2041–2047. [Google Scholar] [CrossRef]
  129. Jang, Y.J.; Park, H.R.; Kim, T.H.; Yang, W.-J.; Lee, J.-J.; Choi, S.Y.; Oh, S.B.; Lee, E.; Park, J.-H.; Kim, H.-P.; et al. High dose bisphenol A impairs hippocampal neurogenesis in female mice across generations. Toxicology 2012, 296, 73–82. [Google Scholar] [CrossRef]
  130. Jašarević, E.; Williams, S.A.; Vandas, G.M.; Ellersieck, M.R.; Liao, C.; Kannan, K.; Roberts, R.M.; Geary, D.C.; Rosenfeld, C.S. Sex and dose-dependent effects of developmental exposure to bisphenol A on anxiety and spatial learning in deer mice (Peromyscus maniculatus bairdii) offspring. Horm. Behav. 2012, 63, 180–189. [Google Scholar] [CrossRef] [Green Version]
  131. Johnson, S.A.; Javurek, A.B.; Painter, M.S.; Ellersieck, M.R.; Welsh, T.H.; Camacho, L.; Lewis, S.M.; Vanlandingham, M.M.; Ferguson, S.; Rosenfeld, C.S. Effects of developmental exposure to bisphenol A on spatial navigational learning and memory in rats: A CLARITY-BPA study. Horm. Behav. 2016, 80, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Xu, X.-B.; He, Y.; Song, C.; Ke, X.; Fan, S.-J.; Peng, W.-J.; Tan, R.; Kawata, M.; Matsuda, K.-I.; Pan, B.-X.; et al. Bisphenol a regulates the estrogen receptor alpha signaling in developing hippocampus of male rats through estrogen receptor. Hippocampus 2014, 24, 1570–1580. [Google Scholar] [CrossRef] [PubMed]
  133. Poimenova, A.; Markaki, E.; Rahiotis, C.; Kitraki, E. Corticosterone-regulated actions in the rat brain are affected by perinatal exposure to low dose of bisphenol A. Neuroscience 2010, 167, 741–749. [Google Scholar] [CrossRef] [PubMed]
  134. Tian, Y.-H.; Baek, J.-H.; Lee, S.-Y.; Jang, C.-G. Prenatal and postnatal exposure to bisphenol a induces anxiolytic behaviors and cognitive deficits in mice. Synapse 2010, 64, 432–439. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, C.; Li, Z.; Han, H.; Luo, G.; Zhou, B.; Wang, S.; Wang, J. Impairment of object recognition memory by maternal bisphenol A exposure is associated with inhibition of Akt and ERK/CREB/BDNF pathway in the male offspring hippocampus. Toxicology 2016, 341–343, 56–64. [Google Scholar] [CrossRef]
  136. Hass, U.; Christiansen, S.; Boberg, J.; Rasmussen, M.G.; Mandrup, K.; Axelstad, M. Low-dose effect of developmental bisphenol A exposure on sperm count and behaviour in rats. Andrology 2016, 4, 594–607. [Google Scholar] [CrossRef] [Green Version]
  137. Matsuda, S.; Matsuzawa, D.; Ishii, D.; Tomizawa, H.; Sajiki, J.; Shimizu, E. Perinatal exposure to bisphenol A enhances contextual fear memory and affects the serotoninergic system in juvenile female mice. Horm. Behav. 2013, 63, 709–716. [Google Scholar] [CrossRef]
  138. Nakamura, K.; Itoh, K.; Dai, H.; Han, L.; Wang, X.; Kato, S.; Sugimoto, T.; Fushiki, S. Prenatal and lactational exposure to low-doses of bisphenol A alters adult mice behavior. Brain Dev. 2012, 34, 57–63. [Google Scholar] [CrossRef]
  139. Stump, D.G.; Beck, M.J.; Radovsky, A.; Garman, R.H.; Freshwater, L.L.; Sheets, L.P.; Marty, M.S.; Waechter, J.M.; Dimond, S.S.; Van Miller, J.P.; et al. Developmental Neurotoxicity Study of Dietary Bisphenol A in Sprague-Dawley Rats. Toxicol. Sci. 2010, 115, 167–182. [Google Scholar] [CrossRef] [Green Version]
  140. Fujimoto, T.; Kubo, K.; Aou, S. Prenatal exposure to bisphenol A impairs sexual differentiation of exploratory behavior and increases depression-like behavior in rats. Brain Res. 2006, 1068, 49–55. [Google Scholar] [CrossRef]
  141. Cox, K.H.; Gatewood, J.D.; Howeth, C.; Rissman, E.F. Gestational exposure to bisphenol A and cross-fostering affect behaviors in juvenile mice. Horm. Behav. 2010, 58, 754–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Gao, R. Common Mechanisms of Excitatory and Inhibitory Imbalance in Schizophrenia and Autism Spectrum Disorders. Curr. Mol. Med. 2015, 15, 146–167. [Google Scholar] [CrossRef] [PubMed]
  143. Selten, M.; Van Bokhoven, H.; Kasri, N.N. Inhibitory control of the excitatory/inhibitory balance in psychiatric disorders. F1000Research 2018, 7, 23. [Google Scholar] [CrossRef] [Green Version]
  144. Mamiya, P.; Arnett, A.; Stein, M. Precision Medicine Care in ADHD: The Case for Neural Excitation and Inhibition. Brain Sci. 2021, 11, 91. [Google Scholar] [CrossRef] [PubMed]
  145. Mhaouty-Kodja, S.; Belzunces, L.P.; Canivenc, M.-C.; Schroeder, H.; Chevrier, C.; Pasquier, E. Impairment of learning and memory performances induced by BPA: Evidences from the literature of a MoA mediated through an ED. Mol. Cell. Endocrinol. 2018, 475, 54–73. [Google Scholar] [CrossRef] [PubMed]
  146. CLARITY-BPA Research Program. NTP Research Report on the Consortium Linking Academic and Regulatory Insights on Bisphenol A Toxicity (CLARITY-BPA): A Compendium of Published Findings: Research Report 18. 2021. Available online: https://pubmed.ncbi.nlm.nih.gov/34910417/ (accessed on 4 March 2022).
  147. Rebolledo-Solleiro, D.; Flores, L.Y.C.; Solleiro-Villavicencio, H. Impact of BPA on behavior, neurodevelopment and neurodegeneration Daniela. Front. Biosci. 2021, 26, 363–400. [Google Scholar] [CrossRef]
  148. Bakoyiannis, I.; Kitraki, E.; Stamatakis, A. Endocrine-disrupting chemicals and behaviour: A high risk to take? Best Pr. Res. Clin. Endocrinol. Metab. 2021, 35, 101517. [Google Scholar] [CrossRef]
  149. Nesan, D.; Kurrasch, D.M. Gestational Exposure to Common Endocrine Disrupting Chemicals and Their Impact on Neurodevelopment and Behavior. Annu. Rev. Physiol. 2020, 82, 177–202. [Google Scholar] [CrossRef] [Green Version]
  150. Xin, F.; Fischer, E.; Krapp, C.; Krizman, E.N.; Lan, Y.; Mesaros, C.; Snyder, N.W.; Bansal, A.; Robinson, M.B.; Simmons, R.A.; et al. Mice exposed to bisphenol A exhibit depressive-like behavior with neurotransmitter and neuroactive steroid dysfunction. Horm. Behav. 2018, 102, 93–104. [Google Scholar] [CrossRef]
  151. Santoro, A.; Chianese, R.; Troisi, J.; Richards, S.; Nori, S.L.; Fasano, S.; Guida, M.; Plunk, E.; Viggiano, A.; Pierantoni, R.; et al. Neuro-toxic and Reproductive Effects of BPA. Curr. Neuropharmacol. 2019, 17, 1109–1132. [Google Scholar] [CrossRef]
Table 2. Summary of studies that have investigated the impact of BPA on synapse formation.
Table 2. Summary of studies that have investigated the impact of BPA on synapse formation.
Neural Structure/Cell TypeOrganismPhenotypeExposureReference
Mushroom bodyDrosophila
melanogaster
Increased axon midline crossing (axon guidance
defect)
0.1 and 1 mM (developmental)Nguyen et al., 2021 [41]
Neuromuscular junction (NMJ)Drosophila
melanogaster
Increased axonal branches1 mM
(developmental)
Welch et al., 2022 [85]
Motor neuronDanio rerioReduced motor axon length and branching; reduced NMJ integrity 50 μM
(developmental)
Morrice et al., 2018 [83]
Motor neuronDanio rerioDecreased ventral and dorsal axons from secondary motoneurons (specifically at
15 μM)
1, 5, and 15 μM
(developmental)
Wang et al., 2013 [84]
Neuroblasts
(Neuro-2A cell line)
Mus musculusCell shrinkage, rounding, and reduced number of synapses; decreased relative protein and mRNA expression levels of Dbn, MAP2 and Tau; increased the relative protein and mRNA expression levels of SYP 50, 100, 150, or 200 μM (in vitro)Yin et al., 2020 [89]
Hippocampus
(CA1 area)
Mus musculus (males only)Inhibited synaptogenesis; altered synaptic structure0.04, 0.4, and 4.0 mg/kg/day
(perinatal)
Xu et al., 2013 [88]
Hippocampal
neurons
Rattus norvegicusIncreased total length of dendrites; increased motility and density of dendritic filipodia 1, 10, and 100 nM (in vitro) Xu et al., 2014 [87]
Embryonic stem cell-derived neural stem cellsHomo sapiensDecreased neurite outgrowth1, 10, and 100 nM (in vitro)Liang et al., 2020 [86]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Welch, C.; Mulligan, K. Does Bisphenol A Confer Risk of Neurodevelopmental Disorders? What We Have Learned from Developmental Neurotoxicity Studies in Animal Models. Int. J. Mol. Sci. 2022, 23, 2894. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052894

AMA Style

Welch C, Mulligan K. Does Bisphenol A Confer Risk of Neurodevelopmental Disorders? What We Have Learned from Developmental Neurotoxicity Studies in Animal Models. International Journal of Molecular Sciences. 2022; 23(5):2894. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052894

Chicago/Turabian Style

Welch, Chloe, and Kimberly Mulligan. 2022. "Does Bisphenol A Confer Risk of Neurodevelopmental Disorders? What We Have Learned from Developmental Neurotoxicity Studies in Animal Models" International Journal of Molecular Sciences 23, no. 5: 2894. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052894

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

Article Metrics

Back to TopTop