Open access peer-reviewed chapter

Bisphenol A: Understanding Its Health Effects from the Studies Performed on Model Organisms

Written By

Papiya Ghosh, Sohini Singha Roy, Morium Begum and Sujay Ghosh

Submitted: 28 January 2017 Reviewed: 04 April 2017 Published: 07 June 2017

DOI: 10.5772/intechopen.68971

From the Edited Volume

Bisphenol A Exposure and Health Risks

Edited by Pinar Erkekoglu and Belma Kocer-Gumusel

Chapter metrics overview

1,800 Chapter Downloads

View Full Metrics

Abstract

Bisphenol A [4,4′‐(propane‐2,2‐diyl)diphenol] (abbreviated as BPA) is a synthetic xenoestrogenic chemical and endocrine disruptor. It is a most common plasticizer that is used widely to produce epoxy resin and polycarbonate plastics, enters the living system through food and water contamination and generates health hazards. Researches are being conducted to explore the adversity that BPA exerts in living body, and for this reason, model organisms are of scientific choice. Rodents, zebrafish, Drosophila, nematodes, crustaceans and echinoderms are being used for monitoring the effect of BPA on their life history traits, nervous system, endocrine system, reproductive systems, behaviour, etc., which could help us to anticipate what kind of challenges BPA is putting in human life. This systematic review is focused on the latest research trend on BPA toxicity on different model organisms.

Keywords

  • bisphenol A
  • rodent
  • zebrafish
  • Drosophila melanogaster
  • invertebrates
  • reproductive system
  • life history traits
  • developmental defects
  • gene expression

1. Introduction

Bisphenol A [4,4′‐(propane‐2,2‐diyl)diphenol] (abbreviated as BPA) is a synthetic xenoestrogenic chemical and endocrine disruptor [13] widely used in dentistry, food packaging and as lacquers to coat food cans, bottle‐tops and water pipes since the 1960s. It is a most common plasticizer that is used widely to produce epoxy resin and polycarbonate plastics. It was first synthesized by Dianin in 1891 and was investigated for potential commercial use in the 1930s during a search for synthetic estrogens. BPA enters the living system inconspicuously through various routes, particularly through food and water contamination, and creates multitude of imperilments at cellular, molecular and genetic level. The EC50 and LC50 values of BPA range from 1.0 to 10 mg/L (Environment Canada 2008), and BPA is declared as ‘moderately toxic’ and ‘toxic’ to aquatic biota by the European Commission and the United States Environmental Protection Agency (US EPA), respectively [4], Commission of the European Communities 1996]. Moreover, environmentally relevant concentrations (12 mg/L or lower) of BPA were also found to be harmful as far as wildlife is concerned [5]. BPA exerts its effect through direct binding to estrogen receptor (ER) in a wide range of species that includes invertebrates, fish, amphibians, reptiles, birds and mammals [6]. BPA binds both ERα and ERβ receptors, with approximately 10‐fold higher affinity to ERβ [7].

The toxicokinetics of BPA exposure reveal that after oral administration in human, BPA is metabolized rapidly in the intestine and liver. BPA is not completely metabolized via Phase I reactions, but it is rapidly conjugated with glucuronic acid (Phase II metabolism) to produce non‐active BPA‐glucuronide in the gut wall and liver. Little amount of BPA also reacts with sulphate to form BPA‐sulphate compound. The formation of BPA conjugates with other chemical moieties is a detoxification process [8, 9]. The BPA conjugates formed in the liver reach the kidney through blood circulation and then excreted in the urine with terminal half‐lives of less than 6 hours [10, 11]. According to a declaration made in 2010 by U.S. Food and Drug Administration, exposure to BPA is alarming because of possible health hazards it exerts on brain, behaviour and prostate gland of foetuses, infants and children. The European Food Safety Authority (EFSA) reviewed new scientific information on BPA in the years 2008, 2009, 2010, 2011 and 2015, concluding on each occasion the known level of exposure to BPA to be hazardous. In February 2016, France announced that it intends to propose BPA as a REACH Regulation candidate substance of very high concern (SVHC).

Owing to difficulty in doing research on human subjects, researchers prefer to use model organisms to test the toxic effect of xenobiotic agents in living system. This approach is also popular in the research on BPA as the agent is ubiquitously present in our ‘plastic wrapped world’ and no perfect control subject could be obtained in natural environment. Several model organisms from different taxa are in use for studying the effects of BPA on their life history, morphological traits, reproductive functioning, neural functioning and behaviour. The outcome of these studies helps to anticipate the probable adversity that BPA inflicts in human body. Keeping all these factors in mind, a critical review on latest research works is presented here to understand the deleterious effects of BPA exposure on different vertebrate and invertebrate model organisms that could facilitate the understanding of human health hazards due to exposure to this xenoestrogen and endocrine disruptor BPA.

Advertisement

2. Vertebrate model animals

2.1. Studies on rodents

Owing to close taxonomic proximity, rodents, including rat, mice and hamster, resemble most with of humans among all other commonly used vertebrate models, and many symptoms of human conditions can be replicated in mice and rats. For that reason, rodents occupy the most preferred model animal in biomedical research, and keeping pace with this global trend, BPA researchers also rely on rodents to unravel the BPA effects on mammals.

2.1.1. Effects on reproductive system

Almost all xenobiotic agents have been tested for their toxicity in rodents to anticipate the probable effects on human body owing to taxonomic closeness of rodents and human as primate. There is extensive evidence that BPA imperils development of reproductive system in male rats and mice, although there appear to be species, strain and dose differences in the sensitivity of specific outcomes to BPA [3]. There are numerous studies of the effects of low doses of BPA on the development of the female and male reproductive organs in rats and mice. Findings include chromosomal abnormalities in oocytes in females [12, 13] and long‐term effects on accessory reproductive organs that are not observed until mid‐life, such as uterine fibroids and para‐ovarian cysts [14]. In Newbold’s study [14], outbred female CD‐1 mice were treated on days 1–5 with subcutaneous injections of BPA (10, 100 or 1000 μg/kg/day). At 18 months of age, ovaries and reproductive tract tissues exhibited significant increase in cystic ovaries and cystic endometrial hyperplasia in the BPA‐treated group. Progressive proliferative lesion of the oviduct and cystic mesonephric (Wolffian) duct remnants was also seen in BPA‐treated groups [15].

The effect of BPA on male reproductive organs and function includes decrease in testosterone secretion [16] and sperm production [17, 18]. Impacts on other reproductive structures include reduction in the size of the epididymis at a dose of 2 ng/g and enlargement of the size of prostate ducts in the male foetuses when pregnant females were exposed to a dose of 10 μg/kg BPA/day [19, 20]. These findings are consistent with effects of low doses of positive control chemicals, such as diethylstilbestrol (DES) and ethinyl estradiol. Moreover, the testicular function impairment includes germ cell sloughing, disruption of the blood‐testis‐barrier and germ cell apoptosis [21, 22].

Impairment in testicular function is also evident in other studies [23, 24, 25]. The effects of BPA resemble more or less the estrogenic effects on the testes [18, 26, 27] with reduction in daily sperm production [28], deformed acrosomal vesicles, acrosomal caps, acrosomes and nuclei of the spermatids. Tohei et al. [29] reported that plasma concentration of testosterone was decreased, and LH was increased in rats after administration of BPA. Testicular content of inhibin was decreased. The testicular response to human chorionic gonadotropin (hCG) for progesterone and testosterone release was also decreased in BPA‐treated rats. These results suggest that BPA directly inhibits testicular functions by disrupting the pathway of negative feedback regulation.

Studies have revealed that BPA exposure also affects the female systems, and it is found to be associated with a number of anomalies like polycystic ovarian syndrome [30], endometriosis [31] and anovulation. Studies have also been conducted to evaluate effects of BPA on development of mammary gland. In utero exposure to 25 and 250 μg BPA/kg body weight showed changes in the mammary glands of CD1 mice, including a significant increase in the percentage of gland ducts, terminal ducts, terminal end buds and alveolar buds at 6 months of age [32]. Perinatal exposure to 25 and 250 ng BPA/kg body weight showed increased area of terminal end buds relative to the gland ductal area [33]. Studies in both rats and mice have shown that BPA induces change in mammary gland morphology that may predispose animals to develop cancer [34, 35]. Table 1 shows the summary of results from experiments on reproductive system of laboratory rodents.

Affected area Model; time and route of exposure Effect Citation
Ovary Mice; developmental, pellet implantation Disruption of early oogenesis Susiarjo et al. [12]
Ovaries and reproductive tract tissues CD‐1 mice; developmental, injection Increase in cystic ovaries and cystic endometrial hyperplasia Newbold et al. [14]
Mammary gland Mice, Rats; developmental, injection, minipump Enhanced growth and differentiation Markey et al. [32]; Munoz‐de‐Toro et al. [33]; Soto et al. [13]; Durando et al. [34]; Murray et al. [35]
Testes Mice, rats ; developmental, adult, oral, injection Decreased testosterone secretion and sperm production ; deformed sperm with reduced motility Akingbemi et al. [16]; Aikawa et al. [17]; Toyama et al. [18]; Al‐Hiyasat et al. [23]; Chitra et al. [24]; Sakaue et al. [26]
Seminiferous tubules C57BL/6 mice; adult, oral Disrupted Takao et al. [25]
Blood Rats, adult, oral ↓ Plasma testosterone and ↑ LH Tohei et al. [29]
Prostate gland CF‐1 mice, CD‐1 mice; developmental, oral ↑ weight, ↑ prostate duct volume Thayer et al. [27]; Timms et al. [20]
Epididymis CF‐1 mice; developmental, oral ↓ size vom Saal et al. [19]

Table 1.

Summary table of the various effects of BPA exposure on reproductive system of laboratory rodents.

2.1.2. Effects on nervous system

BPA has both indirect and direct effects on the nervous system. Since gonadal hormones in conjunction with other neurotrophins regulate cell death, neuronal migration, neurogenesis and neurotransmitter plasticity [36], BPA, in disrupting sex hormone functions, can affect brain development. Estrogen plays a major role in development and differentiation of certain parts of male and female brains. Male and female brains are exposed to different amounts of estrogen during development, and this appears to shape some regions of the brain differently. One of these regions is the hypothalamus, which controls a variety of basic functions including hunger, mood and sex drive. Due to its estrogenic and antiandrogenic activities, BPA can interfere with the dimorphic development of the neuronal networks of male and female brain regulating [37] the activation of hypothalamic estrogen or androgen receptors, testosterone‐activating enzymes and hippocampal aromatase expression [38].

As BPA disrupts thyroid function, it can also affect the development of the nervous system because thyroid hormones regulate prenatal and neonatal development of the brain [39]. Juvenile hypothyroidism due to BPA exposure leads to diminutive dentritic growth in hippocampal neurons of rat brain, resulting in cognitive defects including impaired memory, defective perception and attention problems [40]. In a prenatal study [41] of brain development in mice treated with BPA in a dose 20 μg/kg, body revealed decrease in growth in the ventricular zone of the BPA‐treated offspring, whereas in the cortical plate, growth was increased. In addition, the expression of thyroid Receptor gene TRα (and other genes) was significantly upregulated in the cortical area of the BPA‐treated group. BPA induces cortical plate growth via upregulation of the thyroid pathway. In doing so, BPA might have disrupted normal neocortical development by accelerating neuronal differentiation and migration. BPA exposure may also interfere with the development and expression of normal sex differences in cognitive function, via inhibition of estrogen‐dependent hippocampal synapse formation in female rat [42] and testosterone‐induced hippocampal synapse formation in male mice [43].

In addition, BPA may directly cause neurodegeneration. BPA enhances hydroxyl radical formation in the rat brain [44], and it is induced by 1‐methyl‐4‐phenylpyridinium ion (MPP+) [45]. This leads to neurodegeneration of the substantia nigra and produces acute Parkinsons like symptoms. In this study, 10 μM BPA was infused into the rat striatum to generate OH radical, and in vivo micro‐dialysis technique was used for evaluating toxic effects on nervous tissues. In another study [46], BPA was shown to increase intracellular reactive oxygen species at a concentration of 1, 10, 25 and 50 μmol/L and induce apoptosis at a concentration of 100 μmol/L in mesencephalic neuronal cell culture. Besides, BPA has a significant impact on the dopaminergic system and hippocampal‐associated cognitive functions. Table 2 represents the various observations on the nervous system of laboratory rodents exposed to BPA.

Affected area Model; time and route of exposure Effect Citation
Brain Mice; developmental, injection ↓ growth of ventricular zone, Nakamura et al. [41]
↑ cortical plate growth
Hypothalamus Mice, rats; developmental, injection Affect sex differences in brain development Negri‐Cesi [38]
Hippocampus Sprague‐Dawley rats; adult, injection Inhibits synapse formation at CA1 area MacLusky et al. [42]; Leranth et al. [43]
Striatum Rat; adult, infusion Neurodegeneration of substantia nigra Obata and Kubota [44]

Table 2.

Summary table of the various effects of BPA exposure on nervous system of laboratory rodents.

2.1.3. Effects on chromosomes

Recently, researches have unravelled the fact that maternal exposure to a very low dose (20 ng/g body weight) of BPA disrupts alignment of chromosomes during meiosis in the embryonic oocyte during formation of the primary follicles. This abnormality was also observed in mice that were housed in polycarbonate cages and that were provided water in polycarbonate bottles that had been damaged by exposure to a harsh detergent during washing [47]. This finding suggests that exposure to BPA during the time that meiosis resumes in the mid‐cycle surge by luteinizing hormone (LH) can result in an increase in foetal aneuploidy and subsequent spontaneous abortion in humans [47]. The effect of BPA on aneuploidy has also been examined in cell culture [4851]. In the study by Tsutsui et al. [48, 49], treatment of Syrian hamster embryo cells with BPA (100 μM) for 48 hours resulted in statistically significant increases in the percentage of aneuploid metaphases with chromosome losses. Reports are also available that revealed delay in the meiotic cell cycle, possibly by a mechanism that degrades centrosomal proteins and thus perturbs the spindle microtubule organization and chromosome segregation in mouse oocyte during meiosis. When cultured cells were exposed to BPA during the transition from meiosis‐I to meiosis‐II, a delay in meiosis‐I had been observed. This transition phase usually lasts for 8–10 hours in mice, but for BPA‐exposed culture, 53% of cells remained in meiosis‐I. Insignificant counts of cells were found in anaphase [52].

2.1.4. Effects on behaviour

With inevitable effects of BPA on nervous system, behavioural patterns of rodents are reported to be affected by BPA exposure. An increase in defensive aggression was reported in the offspring of male Sprague‐Dawley rat whose mother was offered oral BPA dose (40 μg/kg/day) throughout gestation [53]. In addition, increased aggressiveness (using a composite score of aggression) in male CD‐1 mouse offspring was evident as a result of oral administration of low dose of BPA (2 and 20 ng/g of body weight) to pregnant females on gestation days 11–17 [54, 55].

A series of studies demonstrated that prenatal and neonatal exposure to BPA upregulates activities of the dopamine system and induced hyperactivity among the experimental rat [56]. Support to this primary report came from the study [57] that revealed prenatal and neonatal exposure of mice to BPA caused upregulation of dopamine D1 receptors, produced hyperlocomotion and increased rewarding responses induced by methamphetamine. Narita et al. [58] demonstrated that exposure of mice to BPA during either organogenesis or lactation, but not implantation and parturition, significantly enhanced the morphine‐induced hyperactivity and rewarding effects. In a rat model, Ishido et al. [59] demonstrated that neonatal exposure to BPA (87 nmol/10 μl/rat) caused significant hyperactivity at 4–5 weeks of age, and significantly decreased gene expression of dopamine transporter at 8 weeks.

Negishi et al. [60] demonstrated that BPA impaired both passive and active avoidance learning among offspring of Fisher 344 rats that were fed a low dose of BPA (0.1 mg/kg/day orally) during pregnancy and lactation. There are also evidences of depressed maternal behaviour in female exposed [61, 62]. There are also reports by Dessi-Fulgheri et al. [63] about decrease in play behaviour of juvenile Sprague‐Dawley rats due to exposure of BPA. Authors observed a masculinization of female behaviour in two behavioural categories, that is, play with females and sociosexual exploration, an effect probably mediated by the estrogenic activity of BPA in the central nervous system.

Foetal/neonatal exposure to low doses of BPA causes sex differences in brain structure, chemistry and behaviour. BPA interferes with the normal processes of sexual differentiation, with brain changes in both male and female rat and mice [61, 64]. Evidence of anatomical alterations in brain sexual differentiation was evident in male and female offspring born to mother exposed to 25 or 250 ng BPA/kg body weight per day [65]. In Fujimoto’s experiment, prenatal exposure to BPA affected male rats and abolished sex differences in rearing behaviour in the open‐field test and struggling behaviour in the forced swimming test. Table 3 shows the summary of the experimental results on the behavioural aspects of laboratory rodents.

Event Model; time and route of exposure Effect Citation
Defensive aggression in male Sprague‐Dawley rat, CD‐1 mice; developmental, oral Increased Farabollini et al. [53]; Kawai et al. [54]
Hyperactivity, hyperlocomotion and rewarding response Mice, rats; adult, developmental, oral, injection Increased Mizuo et al. [56]; Suzuki et al. [57]; Narita et al. [58]; Ishido et al. [59]
Passive and active avoidance learning Fisher 344 rats; developmental, oral Impaired Negishi et al. [60]
Maternal behaviour in females CD‐1 mice, rats; adult, developmental, oral Decreased Palanza et al. [61]; Della Seta et al. [62]
Play behaviour in juveniles Sprague‐Dawley rats; developmental, oral Decreased Farabollini et al. [63]
Sex differences in behaviour CD‐1 mice, rats; developmental, oral Lost Fujimoto et al. [64]; Palanza et al. [61]; Rubin et al. [65]

Table 3.

Summary table of the various effects of BPA exposure on behaviour of laboratory rodents.

2.1.5. Other miscellaneous effects

There are evidences on effects of BPA on subsequent activity of enzymes in tissues and thus metabolic processes [6669]. Study showed very low dose (10 μg/kg) of BPA stimulates insulin production and secretion, which is then followed by insulin resistance at a dose of 100 μg/kg in mice [70]. In the study by Sakurai et al. [71], a high dose of BPA has been revealed to stimulate an increase in the glucose transporter and glucose uptake into adipocytes in cell culture. Study showed that perinatal exposure to a low dose of BPA increased adipogenesis in female rats at weaning [72].

BPA appears to possess complex immuno‐modulating effects. It may stimulate or suppress the immune system. It may also alter immune response pathways. There is extensive evidence that BPA modulates both T helper 1 and T helper 2 cytokine production and alters antibody production [7375]. Yamashita et al. [76] used immune cells from BALB/c mice and demonstrated that BPA induces innate immune response by increasing cytokine synthesis, including tumour necrosis factor (TNF) and IL‐1 in macrophages, and stimulates both T and B cells in adaptive response pathway. Using IL‐2 and IFN‐γ as markers for Th1 response and IL‐4 for Th2 response, the authors found that BPA stimulated Th1 cells to produce IFN‐γ and Th2 cells to express IL‐4. The authors inferred that BPA does not selectively activate the Th1 or Th2 path. BPA also enhances Th1 or Th2 response in vivo, depending on the doses [74, 77]. In addition, prenatal exposure to BPA was shown to augment both Th1 and Th2 responses in adulthood [74]. BPA has been reported to modulate immune function at doses between 2.5 and 30 μg/kg/day [70, 73].

2.2. Studies on zebrafish

Zebrafish (Danio rerio) as vertebrate model system is popular for studying developmental events. The reasons for choosing zebrafish in developmental biology research include its easy maintenance and rearing, prolific fecundity, transparent embryo, absence of placenta that eases the study of morphological characters and even teratogenic effects on anatomy due to experimental exposure to xenotoxicants. Researchers have taken this opportunity to facilitate their understanding in the effects of BPA on vertebrate model. Summary of the results of experiments on zebrafish model is given in Table 4.

Effects on development and reproduction Endpoint Life stage and route of exposure Effect Citation
Hatching, axial curvature, tail morphology Fertilized eggs, directly in a plate Delayed hatching, altered axial curvature, tail malformation Hua and Lin [78]
Early dorso‐ventral patterning, segmentation and brain development Embryo, directly in a plate Altered William et al. [79]
Fertilization and egg production Breeding adult, in aquarium Reduced rate of fertilization, increased egg production Laing et al. [83]
Testes Adult, in aquarium Degenerated, increased number of sustentacular cells, decreased percentage of germ cells Lora et al. [81]
Ovary Adult, in aquarium Deteriorated ovarian tissues, increased number of atretic follicles, distorted and less developed oocytes Yon and Akbulut [82]
Transcription of genes involved in reproductive function Adult, Altered Laing et al. [83]
Oocyte maturation Adult, in aquarium Disrupted Fitzgerald et al. [84]
Effects on nervous system and behaviour Hypothalamus Embryo, directly in culture plate Increased neurogenesis and hyperactivity Kinch et al. [89]
Larval hyperactivity, Adult learning behaviour Embryo, directly in culture plate Increased activity, learning deficit Saili et al. [90]
Effects on chromosomes Oocyte maturation Adult, in aquarium Disrupted by chromatin modification Santangeli et al. [85]

Table 4.

Summary table of the various effects of BPA exposure on zebrafish (Danio rerio).

2.2.1. Effects on development and reproduction

Laboratory studies showed that BPA causes developmental and reproductive effects in zebrafish. There are evidences of delayed hatching, altered axial curvature and tail malformation in zebrafish embryos following exposure of fertilized eggs to BPA [78]. In a study by William et al. [79], BPA altered early dorso‐ventral patterning, segmentation and brain development in zebrafish embryos at a concentration of 50 μM within 24 hours of exposure. Perturbations in expression of cytochrome P450 aromatase activity have also been observed in zebrafish. Estrogen synthesized in the brain by the action of P450 aromatase is known to have organizing effects on the developing central nervous system. In fish, estrogen increases the predominant brain isoform (P450aromB), implying that xenoestrogens like BPA could act as neurodevelopmental toxicants by altering the expression of P450aromB [80].

Lora et al. [81] found several alterations in the zebrafish testes including a pronounced degeneration of all cellular components, an increase in the percentage of the Sertoli cells and a marked decrease in the percentage of germ cells due to exposure of BPA. Histological studies also showed severe deterioration of ovarian tissue such as disintegration of vesicular structures of mature oocytes, irregularities at cytoplasm, reduction in the number of primary and developing oocytes, deformation at the ooplasm and structure of the mature oocytes and irregularities at nucleolus. The number of the atretic oocytes increased due to BPA exposure. Structurally distorted and less developed oocytes were also observed [82]. A study by Laing et al. [83] documented significant increase in egg production, together with a reduced rate of fertilization in zebrafish exposed to BPA, associated with considerable alterations in the transcription of genes involved in reproductive function and epigenetic processes in both liver (vtg1, esr2b, hdac3, mbd2, mecp2 and dnmt1) and gonad tissue (esr2a, cyp19a1a and amh). Their study demonstrated how BPA disrupts reproductive processes in zebrafish. BPA can also disrupt zebrafish oocyte maturation by a novel nongenomic estrogenic mechanism [84]. BPA exerts this nongenomic estrogenic action on zebrafish oocytes directly through binding to the membrane estrogen receptor Gper and activating a Gper‐dependent Egfr/Mapk3/1 pathway. BPA activates this pathway by increasing phosphorylation of Mapk3/1and cAMP concentrations in zebrafish oocytes. Activation of this pathway prevents the resumption of meiotic maturation in fish oocytes [83]. Study showed that BPA downregulated oocyte maturation‐promoting signals through changes in the chromatin structure mediated by histone modifications in zebrafish [85].

2.2.2. Effects on nervous system and behaviour

Zebrafish has been used extensively to elucidate basic mechanisms underlying behavioural toxicology [86]. Zebrafish was also employed as a model for identifying sex‐specific effects on social interactions induced by developmental BPA exposure [87, 88]. A study by Kinch et al. [89] revealed that treatment of embryonic zebrafish with very low‐dose BPA (0.0068 μM, 1000‐fold lower than the accepted human daily exposure) resulted in 180% increase in neurogenesis within the hypothalamus. Fish embryos exposed to BPA exhibit hyperactivity with ontogenetic growth possibly due to the accelerated neural growth. The authors also found that these effects are probably not due to an effect on estrogen receptors (or estrogen‐like receptors) but may be due to its deleterious effects on the synthesis of key enzyme in steroid hormone synthesis, Aromatase B. This study also demonstrated that developmental BPA exposure led to larval hyperactivity or learning deficits in adult zebrafish [90]. There are evidences for temperature‐specific impairment of swimming performance, disturbances in muscle activity and gene expression in zebrafish due to exposure of BPA [91]. This result suggests that BPA toxicity is compounded with the effects of climate change.

2.2.3. Other miscellaneous effects

BPA can alter sex ratio of zebrafish by inducing feminization of the fry [92]. Zebrafish embryos exposed to BPA also showed signs of feminized brains [86]. Kinch et al. [93] investigated morphological changes to developing zebrafish caused by exposure to BPA including changes in body length, pericardia (heart) and the head. Na et al. [94] observed a significant damage in the liver of zebrafish after 96 hours of exposure to BPA. This result further confirmed that liver was the target organ of BPA.

Advertisement

3. Invertebrate model animals

3.1. Study on Drosophila melanogaster

Drosophila melanogaster remains as one of the popular organism in studying the effects of BPA on eukaryotic biological system. The study on Drosophila includes change in gene expression profile, change in behaviour and nervous system, alteration in juvenile growth and development, history traits and fecundity and metabolism.

3.1.1. Effects on life history traits and developmental event

In comparison to other studies on effects of BPA on biological aspects in Drosophila melanogaster, adequate references are available on the researches on Drosophila life history traits. The effects of BPA on growth and development in Drosophila were observed, which demonstrated a statistically significant increase in larval growth for the low‐dose treatment group (0.1 mg/L), but not in the high‐dose treatment group (10 mg/L). BPA exposure caused an increase in body size in treated flies at 48, 72 and 96 hours following egg laying (AEL), suggesting a non‐monotonic dose response. The increase in growth rate found for all treatment groups was associated with a statistically significant increase in food intake observed at 72‐hour AEL. Furthermore, it was observed that the increased growth rate was coupled with an earlier onset of pupariation and metamorphosis, resulting from increased activity of insulin/insulin growth factor signalling (IIS) in Drosophila. Thus, this suggests that BPA exerts its effects through disruption of endocrine signalling in Drosophila since the timing of the onset of pupariation in Drosophila is controlled through the complex interaction of the IIS and the ecdysone signalling pathways. All these observations suggest that the effect is probably due to disruption of insulin‐like signalling in cellular system [95].

Another study on life history traits of Drosophila [96] obtained some contradiction to the above‐mentioned observation. The author reported a delay in both the mean pupation and the mean maturation times in treated group. In that experiment, larvae of D. melanogaster were exposed to three different concentrations: 0.1, 1 and 10 mg/L BPA. In the 0.1 and 1 mg/L exposed groups, the mean offspring numbers were significantly less than that of the control groups, indicating that mean fecundity was significantly decreased. Thus, administration of BPA in both food and through body wall absorption resulted in altered fecundity [96]. Mean decrease in fecundity as compared to control in Drosophila exposed to BPA is also evident in the work of Atli et al. [96]. William et al. [97] have reported that BPA exposure causes inhibition of lipolysis during starvation, leading to significantly increased lipid content after 24 hours of fasting. Furthermore, it also suppresses the expression of insulin‐like peptide in Drosophila, indicating that BPA may inhibit lipid recruitment during starvation in Drosophila.

3.1.2. Effects on behaviour and nervous system

BPA causes [98] behavioural modifications in Drosophila melanogaster, which, in turn, suggests intuitively the role of environmental risk factors for the behavioural impairments like autism and attention deficit hyperactivity disorder (ADHD) in human. The study revealed disturbance in the locomotion patterns of BPA‐exposed Drosophila that may relate to the decision‐making and the motivational state of the animal. Furthermore, an increase in repetitive behaviour and disturbance in grooming behaviour and abnormal social interaction of Drosophila following BPA exposure were seen.

A recent study conducted by Streifel [99] shows that administration of BPA in the prenatal environment had significant impacts on some aspects of Drosophila behaviour, which includes increased time spent in seeking behaviour, increased numbers of peristaltic contractions, increased linear as well as angular movement, decrease in turn angle value as well as potentially significant impacts on motor nerve morphology. These findings suggest implication of BPA as ubiquitous neurotoxin that acts upon the delicate process of neurodevelopment.

3.1.3. Effects on global gene expression profile

Alteration in gene expression profile in Drosophila has been studied by Branco et al. [100]. The authors reported that the effects due to BPA on genome‐wide gene expression of D. melanogaster can be enhanced by the ingestion of high dietary sugar. The authors have found that acute and chronic exposure to BPA causes gross downfall in transcription of testis‐specific genes and overexpression of ribosome‐associated genes across tissues. In addition, it causes alteration of transposable elements that are specific to the ribosomal DNA loci, suggesting that nucleolar stress might implicate in BPA toxicity. This observation suggests that BPA and dietary sugar might functionally interact, with consequences to regulatory programmes in both reproductive and somatic tissues [100].

3.2. Study on other invertebrate model

As compared to vertebrates, the number of research works regarding BPA exposure on invertebrates is minimum. Invertebrates are frequently used as bioindicators for endocrine‐disrupting chemicals. Research suggests that some invertebrates appear to be quite sensitive to BPA, and effects have been documented even at environmentally relevant concentrations [101].

3.2.1. Effects on life history traits and developmental events

A study conducted by Lemos et al. [102] revealed that low BPA concentrations disrupt the endocrine function of terrestrial arthropod Porcellio scaber by causing a sex‐ratio shift. In this study, endocrine system‐related chronic effects were identified at a lower dose of BPA than the concentration having acute toxic effects on isopods, indicating impairment of molting, incomplete ecdysis.

The effects of various concentrations of BPA on the development of two sea urchin species Hemicentrotus pulcherrimus and Strongylocentrotus nudus were examined [103]. This study suggested that the sensitivity of sea urchin embryos and juveniles to endocrine disrupter chemicals changes during the stages of development. The development in the first 12 hours following fertilization up to the morphogenesis of embryo was found to be most sensitive. Even higher concentrations of BPA exposure (>300 mg/L) resulted in developmental arrest and mortality in the sea urchin Paracentrotus lividus [104].

Studies on lepidopteran corn stalk borer Sesamia nonagrioides revealed that BPA induces various developmental disorders through interfering effect in ecdysteroidal pathway [105] and over expression of heat‐shock proteins [106]. Study on freshwater insect Chironomus riparius showed that adult emergence times were significantly delayed on moderate BPA exposure [107]. Marcial et al. (2003) and Watts et al. [108, 109] found that the marine copepod Tigriopus japonicus showed developmental inhibition at a very low concentration of BPA (0.1 mg/L). However, it is unclear if these effects have any long‐term impacts in adult life. Experimental exposure to higher concentration of (11.4 mg/L) BPA for 1 hour caused premature larval metamorphosis in the marine polychaete worm Capitella capitata [110].

A study conducted on Hydra vulgaris by Pascoe et al. [111] pointed that the structure and physiology of polyps were adversely affected at concentrations greater than 42 μg/L BPA. Also, inhibition of regeneration ability was recorded above 460 μg/L BPA concentration. The results indicate that signalling processes necessary for the control and regulation of cell movement and differentiation during normal development, regeneration and sexual reproduction in H. vulgaris are not disrupted by BPA at low environmentally relevant concentrations.

3.2.2. Effects on reproductive system and fecundity

As far as published literatures are concerned, several studies have been conducted to unravel the adverse effects of BPA on reproductive systems and reproductive functioning in various invertebrate animals. In the study of Manshilha et al. [112], an increased fecundity (neonates per female), in comparison with the negative control group (100.3 ± 1.6%), was observed when daphnids were cultured and allowed to breed in the polycarbonate (PC) containers (145.1 ± 4.3%–264.7 ± 3.8%) for single and multiple generations. A strong dose‐dependent ecotoxicological effect was evident, and it was suggested that BPA leached from plastic materials acts as functional estrogen in vivo at very low concentrations. In contrast, neonate production by daphnids cultured in polypropylene and non‐PC bottles was slightly but not significantly enhanced (92.5 ± 2.0 to 118.8 ± 1.8%). Multigenerational tests also demonstrated magnification of the adverse effects, not only on fecundity but also on mortality of the species. Reproductive impairment in Daphnia due to exposure to BPA is also evident in the study by Tišler et al. [113].

Andersen et al. [6] found an increase in egg production in copepod Acartiatonsa exposed to 20 μg BPA/L. Moreover, inhibition in normal development at BPA concentrations above environmentally relevant levels (100 mg/L) was also evident. At extremely high exposures (16,000–80,000 mg/L), abnormal growth and inhibition of gemule germination was found in freshwater sponges Heteromyenia sp. and Eunapius fragilis [114]. A study conducted by Oehlmann et al. [115] on freshwater snail Marisa cornuarietis and of the marine prosobranch Nucella lapillus revealed that BPA affects the reproductive system and has a negative impact on snails even at nominal concentration, that is, 1 μg/L. Affected Marisa females were designated as ‘superfemales’ and were characterized by the presence of additional female organs, hyperplasia of the accessory pallial sex glands, malformations of the pallial oviduct causing increased female mortality and a strong stimulation of oocyte and spawning mass production. In these follow‐up studies, Oehlmann et al. [116] tried to bridge several gaps in knowledge by conducting additional experiments. Here, the authors confirm the previous results and additionally conclude that the occurrence of superfemales is associated with adverse effects on reproduction and survival, even at sub‐micrograms per litre concentrations of BPA (NOEC, 7.9 ng/L; EC10, 13.9 ng/L). However, if snails are exposed to BPA under conditions that maximize the reproductive output, particularly during the spawning season or at elevated temperatures, the induction of superfemales is at least partially masked. The superfemale induction is probably mediated by binding of BPA with estrogen receptor, because the response can completely be reversed by coexposure to potent estrogen inhibitors. Furthermore, the extreme BPA sensitivity of M. cornuarietis and other prosobranch snails probably due to higher affinity of the compound for the estrogen receptor in this species was compared. Overall, the results suggest that BPA imposes a potential hazard for prosobranch population in the field even at environmentally relevant concentrations. Experimentally determined EC50 values of BPA for different invertebrate model organisms have been given in Table 5.

Species EC50 (mg/L) NOEC (mg/L) Reference
Waterflea Daphnia magna 10.2 4.1 Alexander et al. [4]
Mysid Mysidopsis bahia 1.1 0.51 Surprenant [123]
Chironomid Chironomus tentans 2.7 1.4 Mihaich et al. [124]
Copepod Tigriopus japonicus 4.32 3.5 Marcial et al. [108]
Snail Marisa cornuarietis >4.03 (LC50) 1.32 Mihaich et al. [124]
Snail Marisa cornuarietis 2.24 (LC50) 1.18 Mihaich et al. [124]

Table 5.

Summarized presentation showing experimentally determined effective concentration (EC50) and no effect concentration (NOEC) of BPA on different invertebrate animals [103].

3.2.3. Effects on gene expression profile

Change in expression pattern of genes and alteration in RNA expression pattern due to BPA exposure are also within the scientific interest. Planelló et al. [117] studied the effects of BPA on the expression of some selected genes, including housekeeping, stress‐induced and hormone‐related genes in C. riparius larvae. They found that exposure to BPA at a concentration of 3 mg/L for 12–24‐hour exposure did not influence the levels of ribosomal RNA or those of mRNAs for both L11 or L13 ribosomal proteins which were selected as representative of housekeeping genes involved in ribosome biogenesis. Nonetheless, BPA treatment induced the transcription of the HSP70 gene. Interestingly, BPA causes significant increase in transcript of the ecdysone receptor (EcR), suggesting that BPA can selectively affect the expression of the ecdysone receptor gene suggesting a direct interaction with the insect endocrine system.

Significant level of DNA strand break has been detected in snail Potamopyrgus antipodarum under exposure to BPA [118]. DNA‐damaging effect of BPA on aquatic insect C. riparius has also been reported by Martinez‐Paz et al. [119].

Advertisement

4. Conclusion

Bisphenol‐A (BPA), found ubiquitously in our environment, has received a tremendous amount of attention from research scientists, government panels and the popular press. Extensive investigational work has been and is still being carried out in various fields like: (1) mechanisms of BPA action; (2) levels of human exposure; (3) routes of human exposure; (4) pharmacokinetic models of BPA metabolism; (5) effects of BPA on exposed animals and (6) links between BPA and cancer. BPA interferes with hormone signalling via two mechanisms: altering the availability of ovarian hormones and altering binding and activity of the hormone at the receptor level [120122].

Besides understanding the probable human health hazards, study of BPA effect on model organisms facilitates our concern to the issues like biodiversity loss, environmental degradation and overall imbalance in ecological functioning. Today’s world is extremely dependent on plastics, and this dependency inevitably brings the challenges of BPA exposure to the environment. Invertebrate and vertebrate fauna from terrestrial and aquatic ecosystems get affected equally, and the situation is going worse every day. Tantalizingly, the role of BPA in biodiversity loss is not being analysed when the issue comes on the table for discussion. So, mass awareness is to be build up among the people that include students, scholar, academician, conservationist, wildlife activist, NGOs working with environmental issues, policy‐makers and politicians across the nation. It is hard to make BPA free world, but the extent of its adverse effect could be mitigated by our concern and consciousness.

References

  1. 1. Bhandari RK, Deem SL, Holliday DK, Jandegian CM, Kassotis CD, Nagel SCH, Tillitt DE, vom Saal FS, Rosenfeld CS. Effects of the environmental estrogenic contaminants bisphenol A and 17α‐ethinyl estradiol on sexual development and adult behaviors in aquatic wildlife species. General and Comparative Endocrinology. 2015;214:195–219
  2. 2. Yoon K, Kwack SJ, Kim HS, Lee BM. Estrogenic endocrine‐disrupting chemicals: Molecular mechanisms of actions on putative human diseases. Journal of Toxicology and Environmental Health Part B Critical Reviews. 2014;17(3):127–174
  3. 3. Fenichel P, Chevalier N, Brucker‐Davis F. Bisphenol A: An endocrine and metabolic disruptor. Annales D Endocrinologie (Paris). 2013;74(3):211–220
  4. 4. Alexander HC, Dill DC, Smith LW, Guiney PD, Dorn P. Bisphenol A: Acute aquatic toxicity. Environmental Toxicology and Chemistry. 1988;7:19–26
  5. 5. Sohoni P, Tyler CR, Hurd K, Caunter J, Hetheridge M, Williams T, Woods C, Evans M, Toy R, Gargas M, Sumpter JP. Reproductive effects of long‐term exposure to bisphenol A in the fathead minnow (Pimephales promelas). Environmental Science & Technology. 2001;35:2917–2925
  6. 6. Andersen HR, Halling‐Sorensen B, Kusk KO. A parameter for detecting estrogenic exposure in the copepod Acartia tonsa. Ecotoxicology and Environmental Safety. 1999;44:56–61
  7. 7. Pellegrini M, Bulzomi P, Lecis M, Leone S, Campesi I, Franconi F, Marino M. Endocrine disruptors differently influence estrogen receptor β and androgen receptor in male and female rat VSMC. Journal of Cellular Physiology. 2014;229(8):1061–1068
  8. 8. Matthews JB, Twomey K, Zacharewski TR. In vitro and in vivo interactions of bisphenol A and its metabolite, bisphenol A glucuronide, withestrogen receptors alpha and beta. Chemical Research in Toxicology. 2001;14:149–157
  9. 9. Snyder RW, Maness SC, Gaido KW, Sumner SCJ, Fennell TR. Metabolism and disposition of bisphenol A in female rats. Toxicology and Applied Pharmacology. 2000;168:225–234
  10. 10. Volkel W, Colnot T, Csanady GA, Filser JG, Dekant W. Metabolism and kinetics of bisphenol A in humans at low doses following oral administration. Chemical Research in Toxicology. 2002;15:1281–1287
  11. 11. Volkel W, Bittner N, Dekant W. Quantitation of bisphenol A and bisphenol A glucuronide in biological samples by high performance liquid chromatography–tandem mass spectrometry. Drug Metabolism and Disposition. 2005;33:1748–1757
  12. 12. Susiarjo M, Hassold TJ, Freeman E, Hunt PA. Bisphenol A exposure in utero disrupts early oogenesis in the mouse. PLoS Genetics. 2007;3:63–70
  13. 13. Soto AM, Vandenberg LN, Maffini MV, Sonnenschein C. Does breast cancer start in the womb? Basic & Clinical Pharmacology & Toxicology. 2008;102:125–133
  14. 14. Newbold RR, Jefferson WN, Padilla‐Banks E. Long‐term adverse effects of neonatal exposure to bisphenol A on the murine female reproductive tract. Reproductive Toxicology. 2007;24:253–258
  15. 15. Manfo FP, Jubendradass R, Nantia EA, Moundipa PF, Mathur PP. Adverse effects of bisphenol A on male reproductive function. Reviews of Environmental Contamination and Toxicology. 2014;228:57–82
  16. 16. Akingbemi BT, Sottas CM, Koulova AI, Klinefelter GR, Hardy MP. Inhibition of testicular steroidogenesis by the xenoestrogenbisphenol A is associated with reduced pituitary luteinizing hormone secretion and decreased steroidogenic enzyme gene expression in rat Leydig cells. Endocrinology. 2004;145(2):592–603
  17. 17. Aikawa H, Koyama S, Matsuda M, Nakahashi K, Akazome Y, Mori T. Relief effect of vitamin A on the decreased motility of sperm and the increased incidence of malformed sperm in mice exposed neonatally to bisphenol A. Cell and Tissue Research. 2004;315(1):119–124
  18. 18. Toyama Y, Suzuki‐Toyota F, Maekawa M, Ito C, Toshimori K. Adverse effects of bisphenol A to spermiogenesis in mice and rats. Archives of Histology and Cytology. 2004;67(4):373–381
  19. 19. vomSaal FS, Cooke PS, Buchanan DL, Palanza P, Thayer KA, Nagel SC, et al. A physiologically based approach to the study of bisphenol A and other estrogenic chemicals on the size of reproductive organs, daily sperm production, and behavior. Toxicology and Industrial Health. 1998;14(12):239–260
  20. 20. Timms BG, Howdeshell KL, Barton L, Bradley S, Richter CA, vomSaal FS. Estrogenic chemicals in plastic and oral contraceptives disrupt development of the mouse prostate and urethra. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(19):7014–7019
  21. 21. Lagos‐Cabré R, Moreno RD. Contribution of environmental pollutants to male infertility: A working model of germ cell apoptosis induced by plasticizers. Biological Research. 2012;45(1):5–14
  22. 22. Wong EW, Cheng CY. Impacts of environmental toxicants on male reproductive dysfunction. Trends in Pharmacological Sciences. 2011;32(5):290–299
  23. 23. Al‐Hiyasat AS, Darmani H, Elbetieha AM. Effects of bisphenol A on adult male mouse fertility. European Journal of Oral Sciences. 2002;110(2):163–167
  24. 24. Chitra KC, Latchoumycandane C, Mathur PP. Induction of oxidative stress by bisphenol A in the epididymal sperm of rats. Toxicology. 2003;185(12):119–127
  25. 25. Takao T, Nanamiya W, Nagano I, Asaba K, Kawabata K, Hashimoto K. Exposure with the environmental estrogen bisphenol A disrupts the male reproductive tract in young mice. Life Science. 1999;65(22):2351–2357
  26. 26. Sakaue M, Ohsako S, Ishimura R, Kurosawa S, Kurohmaru M, Hayashi Y, et al. Bisphenol A affects spermatogenesis in the adult rat even at a low dose. Journal of Occupational Health. 2001;43:185–190
  27. 27. Thayer KA, Ruhlen RL, Howdeshell KL, Buchanan DL, Cooke PS, Preziosi D, et al. Altered prostate growth and daily sperm production in male mice exposed prenatally to subclinical doses of 17aethinyl oestradiol. Human Reproduction. 2001;16(5):988–996
  28. 28. Jeng HA. Exposure to endocrine disrupting chemicals and male reproductive health. Frontiers in Public Health. 2014;5;2:55
  29. 29. Tohei A, Suda S, Taya K, Hashimoto T, Kogo H. Bisphenol A inhibits testicular functions and increases luteinizing hormone secretion in adult male rats. ExpBiol Med 2001;226:216–221
  30. 30. Palioura E, Diamanti‐Kandarakis E. Industrial endocrine disruptors and polycystic ovary syndrome. Journal of Endocrinological Investigation. 2013;36(11):1105–1111
  31. 31. Benagiano G, Brosens I. In utero exposure and endometriosis. The Journal of Maternal Fetal & Neonatal Medicine. 2014;27(3):303–308
  32. 32. Markey CM, Luque EH, Munoz De Toro M, Sonnenschein C, Soto AM. In utero exposure to bisphenol A alters the development and tissue organization of the mouse mammary gland. Biology of Reproduction. 2001;65(4):1215–1223
  33. 33. Munoz‐de‐Toro M, Markey CM, Wadia PR, Luque EH, Rubin BS, Sonnenschein C, et al. Perinatal exposure to bisphenol‐A alters peripubertal mammary gland development in mice. Endocrinology. 2005;146(9):4138–4147
  34. 34. Durando M, Kass L, Piva J, Sonnenschein C, Soto AM, Luque E, et al. Prenatal bisphenol A exposure induces preneoplastic lesions in the mammary gland in Wistar rats. Environmental Health Perspectives. 2007;115(1):80–86
  35. 35. Murray TJ, Maffini MV, Ucci AA, Sonnenschein C, Soto AM. Induction of mammary gland ductal hyperplasias and carcinoma in situ following fetal bisphenolA exposure. Reproductive toxicology (Elmsford, NY). 2007;23(3):383–390. DOI: 10.1016/j.reprotox.2006.10.002
  36. 36. Simerly RB. Wired for reproduction: Organization and development of sexually dimorphic circuits in the mammalian forebrain. Annual Review of Neuroscience. 2002;25:507–536
  37. 37. Itoh K, Yaoi T, Fushiki S. Bisphenol A, an endocrine‐disrupting chemical, and brain development. Neuropathology. 2012;32(4):447–457
  38. 38. Negri‐Cesi P. Bisphenol A interaction with brain development and functions. Dose‐Response. 2015;13(2):1559325815590394. DOI: 10.1177/1559325815590394
  39. 39. Porterfield SP, Hendrich CE. The role of thyroid hormones in prenatal and neonatal neurological development—Current perspectives. Endocrine Reviews. 1993;14:94–106
  40. 40. Schantz SL, Widholm JJ. Cognitive effects of endocrine‐disrupting chemicals in animals. Environmental Health Perspectives. 2001;109:1197–1206
  41. 41. Nakamura K, Itoh K, et al. Murine neocortical histogenesis is perturbed by prenatal exposure to low doses of Bisphenol A. Journal of Neuroscience Research. 2006;84(6):1197–1205
  42. 42. MacLusky NJ, Hajszan T, Leranth C. The environmental estrogen bisphenol A inhibits estrogen induced hippocampal synaptogenesis. Environmental Health Perspectives. 2005;113:675–679
  43. 43. Leranth C, Szigeti‐Buck K, Maclusky NJ, Hajszan T. Bisphenol A prevents the synaptogenic response to testosterone in the brain of adult male rats. Endocrinology. 2007;149:988–994
  44. 44. Obata T, Kubota S. Formation of hydroxy radicals by environmental estrogen‐like chemicals in rat striatum. Neuroscience Letters. 2000;296:41–44
  45. 45. Obata T. Imidaprilat, an angiotensin‐converting enzyme inhibitor exerts neuroprotective effect via decreasing dopamine efflux and hydroxyl radical generation induced by bisphenol A and MPP+ in rat striatum. Brain Research. 2006;1071:250–253
  46. 46. Lin Y, Zeng XG, Wu DS, Wang X. Study on bisphenol A induced primary cultured mesencephalicneuronal cell injury by oxidative stress. Wei Sheng Yan Jiu. 2006;35:419–422
  47. 47. Hunt PA, Koehler KE, Susiarjo M, Hodges CA, Hagan A, Voigt RC, et al. Bisphenol A causes meiotic aneuploidy in the female mouse. Current Biology. 2003;13:546–553
  48. 48. Tsutsui T, Tamura Y, Yagi E, Hasegawa K, Takahashi M, Maizumi N, et al. Bisphenol‐A induces cellular transformation, aneuploidy and DNA adduct formation in cultured Syrian hamster embryo cells. International Journal of Cancer. 1998;75(2):290–294
  49. 49. Tsutsui T, Tamura Y, Suzuki A, Hirose Y, Kobayashi M, Nishimura H, et al. Mammalian cell transformation and aneuploidy induced by five bisphenols. International Journal of Cancer. 2000;86(2):151–154
  50. 50. Parry EM, Parry JM, Corso C, Doherty A, Haddad F, Hermine TF, et al. Detection and characterization of mechanisms of action of aneugenic chemicals. Mutagenesis. 2002;17(6):509–521
  51. 51. Eichenlaub‐Ritter U, Sun F, Betzendahl I. Meiotic progression, spindle formation and chromosome segregation in in vitro maturing mouse oocytes exposed to bisphenol A. Environmental Research. 2005;98(3):405–406
  52. 52. Can A, Semiz O, Cinar O. Bisphenol‐A induces cell cycle delay and alters centrosome and spindle microtubular organization in oocytes during meiosis. Molecular Human Reproduction. 2005;11(6):389–396. DOI: 10.1093/molehr/gah179
  53. 53. Farabollini F, Porrini S, Della Seta D, Bianchi F, Dessi‐Fulgheri F. Effects of perinatal exposure to bisphenol A on sociosexual behavior of female and male rats. Environmental Health Perspectives. 2002;110(Suppl 3):409–414
  54. 54. Kawai K, Takehiro N, Nishikata H, Aou S, Takii M, Kubo C. Aggressive behavior and serum testosterone concentration during the maturation process of male mice: The effects of fetal exposure to bisphenol A. Environmental Health Perspectives. 2003;111:175–178
  55. 55. Kajta M, Wójtowicz AK. Impact of endocrine‐disrupting chemicals on neural development and the onset of neurological disorders. Pharmacological Reports. 2013;65(6):1632–1639
  56. 56. Mizuo K, Narita M, Miyagawa K, Okuno E, Suzuki T. Prenatal and neonatal exposure to bisphenolA affects the morphine‐induced rewarding effect and hyperlocomotion in mice. Neuroscience Letters. 2004;356(2):95–98
  57. 57. Suzuki T, Mizuo K, Nakazawa H, Funae Y, Fushiki S, Fukushima S, Shirai T, Narita M. Prenatal and neonatal exposure to bisphenol‐A enhances the central dopamine D1 receptor‐mediated action in mice: Enhancement of the methamphetamine‐induced abuse state. Neuroscience. 2003;117:639–644
  58. 58. Narita M, Miyagawa K, Mizuo K, Yoshida T, Suzuki T. Changes in central dopaminergic systems and morphine reward by prenatal and neonatal exposure to bisphenol‐A in mice: Evidence for the importance of exposure period. Addiction Biology. 2007;12:167–172
  59. 59. Ishido M, Morita M, Oka S, Masuo Y. Alteration of gene expression of G protein coupled receptors in endocrine disruptors‐caused hyperactive rats. Regulatory Peptides. 2005;126:145–153
  60. 60. Negishi T, Kawasaki K, Suzaki S, Maeda H, Ishii Y, Kyuwa S, et al. Behavioral alterations in response to fear-provoking stimuli and tranylcypromine induced by perinatal exposure to bisphenol A and nonylphenol in male rats. Environ Health Perspect 2004;112(11):1159–1164
  61. 61. Palanza PL, Gioiosa L, Parmigiani S, vomSaal FS. Effects of developmental exposure to bisphenolA on brain and behavior in mice. Environmental Research. 2008;108:150–157
  62. 62. Della Seta D, Minder I, Dessi-Fulgheri F, Farabollini F. Bisphenol-A exposure during pregnancy and lactation affects maternal behavior in rats. Brain Res Bull 2005;65(3):255–260
  63. 63. Dessi-Fulgheri F, Porrini S, Farabollini F. Effects of perrinatal exposure to bisphenol A on play behavior of female and male juvenile rats. Environ Health Perspect 110 Suppl 2002;3:403-407
  64. 64. 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 Research. 2006;1068:49–55
  65. 65. Rubin BS, Lenkowski JR, Schaeberle CM, Vandenberg LN, Ronsheim PM, Soto AM. Evidence of altered brain sexual differentiation in mice exposed perinatally to low, environmentally relevant levels of bisphenol A. Endocrinology. 2006;147:3681–3691
  66. 66. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor‐α knockout mice. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(23):12729–12734
  67. 67. Cooke PS, Heine PA, Taylor JA, Lubahn DB. The role of estrogen and estrogen receptor‐alpha in male adipose tissue. Molecular and Cellular Endocrinology. 2001;178(12):147–154
  68. 68. Kabuto H, Hasuike S, Minagawa N, Shishibori T. Effects of bisphenol A on the metabolisms of active oxygen species in mouse tissues. Environmental Research. 2003;93(1):31–35
  69. 69. Alonso‐Magdalena P, Morimoto S, Ripoll C, Fuentes E, Nadal A. The estrogenic effect of bisphenol A disrupts pancreatic β‐cell function in vivo and induces insulin resistance. Environmental Health Perspectives. 2006;114(1):106–112
  70. 70. Ropero AB, Alonso‐Magdalena P, Garcia‐Garcia E, Ripoll C, Fuentes E, Nadal A. Bisphenol‐A disruption of the endocrine pancreas and blood glucose homeostasis. International Journal of Andrology. 2008;31:194–200
  71. 71. Sakurai K, Kawazuma M, Adachi T, Harigaya T, Saito Y, Hashimoto N, et al. Bisphenol A affects glucose transport in mouse 3T3-F442A adipocytes. Br J Pharmacol. 2004;141(2):209–214
  72. 72. Somm E, Schwitzgebel VM, Toulotte A, Cederroth CR, Combescure C, Nef S, Aubert ML, Huppi PS. Perinatal exposure to bisphenol a alters early adipogenesis in the rat. Environmental Health Perspectives. 2009;117:1549–1555
  73. 73. Sawai C, Anderson K, Walser‐Kuntz D. Effect of bisphenolA on murine immune function: Modification of interferon‐γ, IgG2a, and disease symptoms in NZB × NZW F1 mice. Environmental Health Perspectives. 2003;111(16):1883–1887
  74. 74. Yoshino S, Yamaki K, Yanagisawa R, Takano H, Hayashi H, Mori Y. Effects of bisphenol A on antigen‐specific antibody production, proliferative responses of lymphoid cells, and TH1 and TH2 immune responses in mice. British Journal of Pharmacology. 2003;138(7):1271–1276
  75. 75. Ohshima Y, Yamada A, Tokuriki S, Yasutomi M, Omata N, Mayumi M. Transmaternal exposure to bisphenol a modulates the development of oral tolerance. Pediatric Research. 2007;62:60–64
  76. 76. Yamashita U, Sugiura T and Kuroda E. Effect of endocrine disrupters on immune responses in vitro. J UOEH 2002;24:1–10
  77. 77. Tian X, Takamoto M, Sugane K. Bisphenol A promotes IL‐4 production by Th2 cells. International Archives of Allergy and Immunology. 2003;132:240–247
  78. 78. Hua DZ, Lin Z. Toxicity of bisphenol A on the growth of zebrafish embryos. Acta Hydrobiologica Sinica. 2006;30(6):638–642
  79. 79. William KF, Yeung BHY, Wan HT, Wong CKC. Early embryogenesis in zebrafish is affected by bisphenolAexposure. Biology Open 2013;2(5):466-471. DOI:10.1242/bio.20134283
  80. 80. Kishida M, McLellan M, Miranda JA, Callard GV. Estrogen and xenoestrogens upregulate the brain aromatase isoform (P450aromB) and perturb markers of early 39 development in zebrafish (Danio rerio). Comparative Biochemistry and Physiology Part B Biochemistry & Molecular Biology. 2001;129:261–268
  81. 81. Lora AJ, Molina AM, Bellido C, Blanco A, Monterde JG, Moyano MR. Adverse effects of bisphenol A on the testicular parenchyma of zebrafish revealed using histomorphological methods. VeterinarniMedicina 2016;61(10):577–589 DOI: 10.17221/212/2015-VETMED
  82. 82. Yön ND, Akbulut C. Histological changes in zebrafish (Danio rerio) ovaries following administration of bisphenol A. Pakistan Journal of Zoology. 2014;46(4):1153–1159
  83. 83. Laing LV, Viana J, Dempster EL, et al. Bisphenol A causes reproductive toxicity, decreases dnmt1 transcription, and reduces global DNA methylation in breeding zebrafish (Danio rerio). Epigenetics. 2016;11(7):526–538. DOI: 10.1080/15592294.2016.1182272
  84. 84. Fitzgerald AC, Peyton C, Dong J, Thomas P. Bisphenol A and related alkylphenols exert nongenomic estrogenic actions through a G protein‐coupled estrogen receptor 1 (GPER)/epidermal growth factor receptor (EGFR) pathway to inhibit meiotic maturation of zebrafish oocytes. Biology of Reproduction. 2015;93(6):135. DOI: 10.1095/biolreprod.115.132316
  85. 85. Santangeli S, Maradonna F, Gioacchini G, Cobellis G, Piccinetti CC, Dalla Valle L, Carnevali O. BPA‐Induced deregulation of epigenetic patterns: Effects on female zebrafish reproduction. Scientific Reports. 2016;6:21982. DOI: http://doi.org/10.1038/srep21982
  86. 86. Bailey J, Oliveri A, Levin ED. Zebrafish model systems for developmental neurobehavioral toxicology. Birth Defects Research Part C Embryo Today. 2013;99:14–23
  87. 87. Dahlbom SJ, Backström T, Lundstedt‐Enkel K, Winberg S. Aggression and monoamines: Effects of sex and social rank in zebrafish (Danio rerio). Behavioural Brain Research. 2012;228:333–338
  88. 88. Weber DN, Hoffmann RG, Hoke ES, Tanguay RL. Bisphenol A exposure during early development induces Sex‐Specific changes in adult zebrafish social interactions. Journal of Toxicology and Environmental Health Part A. 2015;78(1):50–66. DOI: 10.1080/15287394.2015.958419
  89. 89. https://www.ncbi.nlm.nih.gov/pubmed/?term=Kinch%20CD%5BAuthor%5D&cauthor=true&cauthor_uid=27107150” Kinch CD, “https://www.ncbi.nlm.nih.gov/pubmed/?term=Kurrasch%20DM%5BAuthor%5D&cauthor=true&cauthor_uid=27107150” Kurrasch DM, “https://www.ncbi.nlm.nih.gov/pubmed/?term=Habibi%20HR%5BAuthor%5D&cauthor=true&cauthor_uid=27107150” Habibi HR. Adverse morphological development in embryonic zebrafish exposed to environmental concentrations of contaminants individually and in mixture. Aquat Toxicol 2016;175:286–298. doi: 10.1016/j.aquatox.2016.03.021.
  90. 90. Saili KS, Corvi MM, Weber DN, Patel AU, Das SR, Przybyla J, Anderson KA, Tanguay RL. Neurodevelopmental low‐dose bisphenolA exposure leads to early life‐stage hyperactivity and learning deficits in adult zebrafish. Toxicology. 2012;291:83–92
  91. 91. Little AG, SeebacherF. Temperature determines toxicity: Bisphenol A reduces thermal tolerance in fish. Environmental Pollution. 2015;197:84–89
  92. 92. Drastichová J, Svobodová Z, Groenland M, Dobsíková R, Îlábek V, Weissová D, Szotkowská M. Effect of exposure to bisphenol A and 17β‐estradiol on the sex differentiation in zebrafish (Danio rerio). Acta Veterinaria Brno. 2005;74:287–291
  93. 93. Kinch CD, Kurrasch DM, Habibi HR. Adverse morphological development in embryonic zebra fish exposed to environmental concentration of contaminants individually and in mixture. Aquat Toxicol 2015;175:286–298
  94. 94. Na CAO, Hua WEI, Ling-guang WU, Ting-ting WU, Guo-peng LI. Effects of bisphenol A on zebrafish (Daniorerio) liver and gonad.Chinese Journal of Ecology 2010;29(11):2192–2198
  95. 95. Weiner AK, Ramirez A, Zintel T, Rose RW, Wolff E, Parker AL, Bennett K, Johndreau K, Rachfalski C, Zhou J, Smith ST. Bisphenol A affects larval growth and advances the onset of metamorphosis in Drosophila melanogaster. Ecotoxicology and Environmental Safety. 2014;101:7–13
  96. 96. Atli E, Ünlü H. Developmental and reproductive effects of bisphenol A (Bpa) in Drosophila melanogaster. The Journal of Biological Chemistry. 2012;40(1):61–68
  97. 97. Williams MJ, Wang Y, Klockars A, Monica Lind P, Fredriksson R, Schioth HB. Exposure to Bisphenol A affects lipid metabolism in Drosophila melanogaster. Basic Clin Pharmacol Toxicol 2014;114(5):414–420
  98. 98. Kaur K, Simon AF, Chauhan V, Chauhan A. Effect of bisphenol A on Drosophila melanogaster behavior—A new model for the studies on neurodevelopmental disorders. Behavioural Brain Research. 2015;284:77–84
  99. 99. Streifel A M. Effect of bisphenol-A on neurodevelopment in Drosophila melanogaster larvae.2016. (Thesis)
  100. 100. Branco AT, Lemos B. Interaction between bisphenol A and dietary sugar affects global gene transcription in Drosophila melanogaster. Genomics Data. 2014;2:308–311
  101. 101. Oehlmann J, Schulte‐Oehlmann U, Kloas W, Jagnytsch O, Lutz I, Kusk KO, Wollenberger L, Santos EM, Paull GC, Van Look KJ, Tyler CR. A critical analysis of the biological impacts of plasticizers on wildlife. Philosophical Transactions of the Royal Society B. 2009;364:2047–2062
  102. 102. Lemos MFL, van Gestel, CAM, Soares AMVM. Endocrine disruption in a terrestrial isopod under exposure to bisphenol A and vinclozolin. J Soil Sediments 2009;9:492–500.
  103. 103. Kiyomoto M, Kikuchi A, Unuma T, Yokota Y. Effects of ethynylestradiol and bisphenol A on the development of sea urchin embryos and juveniles. Marine Biology. 2006;149:57–63
  104. 104. Arslan OC, Parlak H. Effects of bisphenol A on the embryonic development of sea urchin (Paracentrotus lividus). Environmental Toxicology. 2008;23:387–392
  105. 105. Kontogiannatos D, Swevers L, Zakasis G, Kourti A. The molecular and physiological impact of bisphenol A in Sesami anonagrioides (Lepidoptera: Noctuidae). Ecotoxicology. 2015;24(2):356–367
  106. 106. Michail X, Kontogiannatos D, Syriou V, Kourti A. Bisphenol‐A affects the developmental progression and expression of heat‐shock protein genes in the moth Sesamia nonagrioides. Ecotoxicology. 2012;21(8):2244–2253
  107. 107. Watts MM, Pascoe D. Comparison of Chironomus riparius Meigen and Chironomus tentansFabricius (Diptera: Chironomidae) for assessing the toxicity of sediments. Environmental Toxicology and Chemistry. 2000;19:1885–1892
  108. 108. Marcial HS, HagiwaraA, Snell TW. Estrogenic compounds affect development of harpacticoid copepod Tigriopus japonicus. Environ Toxicol Chem 2003;22(12):3025–3030
  109. 109. Watts MM, Pascoe D, Carroll K. Exposure to 17a-ethinylestradiol and bisphenol A e effects on larval moulting and mouthpart structure of Chironomus riparius. Ecotoxicol Environ Saf 2003;54:207–215
  110. 110. Biggers WJ, Laufer H. Identification of juvenile hormone‐active alkylphenols in the lobster Homarus americanus and in marine sediments. The Biological Bulletin. 2004;206:13–24
  111. 111. Pascoe D, Carroll K, Karntanut W, Watts MM. Toxicity of 17a-ethinylestradiol and bisphenol A to the freshwater cnidarian Hydra vulgaris. Arch Environ Contam Toxicol 2002;43:56–63
  112. 112. Mansilha C, Silva P, Rocha S, Gameiro P, Domingues V, Pinho C, Ferreira IM. Bisphenol A migration from plastic materials: direct insight of ecotoxicity in Daphnia magna. Environ Sci Pollut Res Int. 2013;20(9):6007–18
  113. 113. Tišler T, Krel A, Gerželj U, Erjavec B, Dolenc MS, Pintar A. Environ Pollut. Hazard identification and risk characterization of bisphenols A, F and AF to aquatic organisms.2016;212:472–479
  114. 114. Hill M, Stabile C, Steffen LK, Hill A. Toxic effects of endocrine disruptors on freshwater sponges: Common developmental abnormalities. Environmental Pollution. 2002;117(2):295–300
  115. 115. Oehlmann J, Schulte-Oehlmann U, Tillmann M, Markert B. Effects of endocrine disruptors on prosobranch snails (Mollusca: Gastropoda) in the laboratory. Part I: Bisphenol A and octylphenol as xeno-estrogens. Ecotoxicology 2000;9:383–397
  116. 116. Oehlmann J, Schulte-Oehlmann U, Bachmann J, Oetken M , Lutz I, Kloas W, Ternes TA. Bisphenol A induces superfeminization in the ramshorn snail Marisa cornuarietis (Gastropoda: Prosobranchia) at environmentally relevant concentrations. Environ Health Perspect 2006;114:127–133
  117. 117. Planelló R, Martínez-Guitarte JL, Morcillo G. “http://www.ncbi.nlm.nih.gov/pubmed/18313723”The endocrine disruptor bisphenol A increases the expression of HSP70 and ecdysone receptor genes in the aquatic larvae of Chironomus riparius.Chemosphere 2008;71(10):1870–6
  118. 118. Vincent‐Hubert F, Revel M, Garric J. DNA strand breaks detected in embryos of the adult snails, Potamopyrgus antipodarum, and in neonates exposed to genotoxic chemicals. Aquatic Toxicology. 2012;122–123:1–8
  119. 119. Martínez PP, Morales M, Martínez-Guitarte JS, Morcillo G. Genotoxic effects of environmental endocrine disruptors on the aquatic insect Chironomus riparius evaluated using the comet assay. Mutation Research/Genetic Toxicology and Environmental Mutagenesis 2013;758:41–47
  120. 120. Craig ZR, Wang W, Flaws JA. Endocrine‐disrupting chemicals in ovarian function: Effects on steroidogenesis, metabolism and nuclear receptor signalling. Reproduction. 2011;142(5):633–646
  121. 121. Prossnitz ER, Barton M. The G‐protein‐coupled estrogen receptor GPER in health and disease. Nature Reviews Endocrinology. 2011;7(12):715–726
  122. 122. Weiss B. The intersection of neurotoxicology and endocrine disruption. Neurotoxicology. 2012;33(6):1410–1419
  123. 123. Surprenant D. Acute toxicity of bisphenol A to the mysid (Mysidopsis bahia) under flow-through conditions. Report BW-85-8-1825, SpringbornBionomics Aquatic Toxicilogy Lab 1985, Wareham, MA, USA
  124. 124. Mihaich EM, Friederich U, Carpers N, Hall AT, Klecka GM, Dimond SS, Staples CA, Ortego LS, Hentges SG. Acute and chronic toxicity testing of bisphenol A with aquatic invertebrates and plants. Ecotoxicology and Environmental Safety. 2009;72:1392–1399

Written By

Papiya Ghosh, Sohini Singha Roy, Morium Begum and Sujay Ghosh

Submitted: 28 January 2017 Reviewed: 04 April 2017 Published: 07 June 2017