Elsevier

Reproductive Toxicology

Volume 24, Issue 2, August–September 2007, Pages 199-224
Reproductive Toxicology

Review
In vivo effects of bisphenol A in laboratory rodent studies

https://doi.org/10.1016/j.reprotox.2007.06.004Get rights and content

Abstract

Concern is mounting regarding the human health and environmental effects of bisphenol A (BPA), a high-production-volume chemical used in synthesis of plastics. We have reviewed the growing literature on effects of low doses of BPA, below 50 mg/(kg day), in laboratory exposures with mammalian model organisms. Many, but not all, effects of BPA are similar to effects seen in response to the model estrogens diethylstilbestrol and ethinylestradiol. For most effects, the potency of BPA is approximately 10–1000-fold less than that of diethylstilbestrol or ethinylestradiol. Based on our review of the literature, a consensus was reached regarding our level of confidence that particular outcomes occur in response to low dose BPA exposure. We are confident that adult exposure to BPA affects the male reproductive tract, and that long lasting, organizational effects in response to developmental exposure to BPA occur in the brain, the male reproductive system, and metabolic processes. We consider it likely, but requiring further confirmation, that adult exposure to BPA affects the brain, the female reproductive system, and the immune system, and that developmental effects occur in the female reproductive system.

Introduction

Bisphenol A (BPA) is used as the monomer to manufacture polycarbonate plastic, the resin that lines most food and beverage cans, dental sealants, and as an additive in other plastics (Fig. 1, Table 1) [1]. BPA is one of the highest volume chemicals produced worldwide; global BPA production capacity in 2003 was 2.2 million metric tonnes (over 6.4 billion pounds), with a 6–10% growth in demand expected per year [2]. Heat and either acidic or basic conditions accelerate hydrolysis of the ester bond linking BPA monomers, leading to release of BPA and the potential for human and environmental exposure. Studies conducted in Japan [3] and in the USA [4] have shown that BPA accounts for the majority of estrogenic activity that leaches from landfills into the surrounding ecosystem.

BPA has been demonstrated in both in vivo and in vitro experiments to act as an endocrine disrupting chemical (EDC) (reviewed in [5], [6]). There is extensive evidence that BPA is an estrogen-mimicking chemical, although recent findings have revealed that BPA is a selective estrogen receptor modulator (SERM), since BPA and the potent endogenous estrogen 17β-estradiol (E2) do not always show identical effects, and in some studies BPA has been shown to antagonize the activity of E2 [7]. There is evidence that, similar to other estrogens, BPA can bind to androgen receptors and inhibit the action of androgen [8]. In addition, there is evidence for an anti-thyroid hormone effect of BPA [9]. However, effects of BPA mediated by binding to androgen and thyroid hormone receptors appear to require higher doses than those required to elicit estrogenic or antiestrogenic responses [7].

Chemicals classified as endocrine disruptors include not only hormone-mimics or antagonists that act via binding to receptors, but also chemicals that can interfere with hormone synthesis and clearance, as well as other aspects of tissue metabolism. Experiments have shown that BPA influences enzyme activity and thus metabolism in various tissues. Another mechanism of endocrine disruption is the alteration of hormone receptor expression, and experiments described below have shown that BPA alters hormone receptor numbers and hormone receptor gene activity in target tissues.

In this review, we summarize the recent literature on low dose effects of BPA in laboratory animals. The majority of the studies used rats and mice; only a few used other mammalian species. We conclude with a series of statements expressing our level of confidence concerning various effects of BPA in laboratory animals at low doses.

Endocrinology experiments with laboratory animals are particularly vulnerable to confounding effects. Among other difficulties, treatment effects can be masked by hormonally active components of feed, water, or caging; and species and strains differ greatly in their sensitivity to different hormonally active compounds. Therefore, proper reporting of experimental design is critical to evaluation of studies in the literature. Below, we discuss several of the most important aspects of experimental design, which we considered when evaluating the studies discussed in this review.

Published results must identify precisely the animal model and supplier being used. For example, Sprague–Dawley rats from different commercial breeders cannot be assumed to be the same, since it is an outbred stock. In particular, the outbred Sprague–Dawley CD rat from Charles River Laboratories [Crl:CD(SD)] has very low sensitivity to exogenous estrogens, and after more than 50 years of selective breeding by Charles River for large body size and litter size, it would be inappropriate to identify these rats as just Sprague–Dawley (Table 2). In contrast to the Crl:CD(SD) rat, male and female CD-1 (ICR) mice are highly sensitive to exposure to low doses of BPA during development as revealed by over 20 published studies (reviewed below) reporting significant effects of low doses of BPA in this outbred stock. This high sensitivity of the CD-1 mouse to BPA is predicted by the high responsiveness to positive control estrogens: E2, ethinylestradiol and diethylstilbestrol (DES), as revealed by both the in vivo studies discussed here and other studies of CD-1 cells and organs in primary culture [10], [11], [12].

The marked difference in sensitivity of different animal models used in toxicological, pharmacological and endocrinological research is just one of many reasons why it is essential that experiments include appropriate positive controls, which is discussed in more detail in following sections. With regard to examination of the in vivo estrogenic activity of BPA, which is the subject of this review, the sensitivity of the endpoint of interest in the chosen animal model should be characterized with a positive control such as E2 (appropriate if administration is by injection or subcutaneous capsule, due to very limited oral absorption of E2) or either DES or ethinylestradiol (appropriate if chemicals are administered orally, since they are orally active at very low doses). For example, in the CD-1 mouse, which is the animal model used by the U.S. National Toxicology Program, an appropriate positive control dose of ethinylestradiol or DES to detect a response to BPA within the low dose range (below 50 mg/(kg day)) would be an oral dose not greater than 5 μg/(kg day). This suggestion is based on numerous reports that for responses mediated by nuclear estrogen receptors, estimates of BPA potency in CD-1 mice range between 10 and 1000-fold less than either ethinylestradiol or DES, depending on the specific response being measured [10], [12], [13], [14], [15], [16]. Also, doses of DES above 5 μg/(kg day) can result in opposite effects relative to lower doses; this has been shown following developmental exposure for the prostate [16], [17] and uterus [18]. High doses of these positive control estrogenic chemicals are thus not appropriate as a positive control for low dose effects of BPA or other estrogenic endocrine disrupting chemicals.

The exact feed used must be identified. Ideally, the estrogenicity of the feed should be characterized, since estrogenic components have been demonstrated to occur in both soy-based and non-soy-based animal feeds. There is also the possibility of variation in estrogenic activity between different lots of feed commonly used in toxicological research [19]. If possible, the same lot of feed (same mill date) should be used throughout an experiment.

The type of caging should be carefully selected to avoid estrogenic contamination of experimental animals. In particular, polycarbonate cages and water bottles should not be used, since they will leach uncontrolled concentrations of BPA to the experimental animals [20]. Polypropylene cages have been used successfully in studies of estrogens in mice, and polysulfone cages are available to replace polycarbonate; polysulfone is a co-polymer containing BPA and sulfone, but it is reported to be more resistant to degradation at high temperatures relative to polycarbonate [20]. Similarly, the source of drinking water must be free of BPA and other estrogens; reverse osmosis and carbon filtration is often necessary to achieve this requirement. In addition, if BPA is delivered in drinking water, the water must be free of chlorine or other reactive ions. Adding BPA to chlorinated water results in formation of tetrachlorobisphenol A.

In all cases the precise method of dosing the animals and the time of dosing should be identified. Methods of administration of BPA include—(1) oral (p.o.): by gavage, by adding BPA to feed or drinking water, or by feeding the chemical in oil; (2) injection: subcutaneous (s.c.), intraperitoneal (i.p.), intracisternal, or intramuscular (i.m.) routes; and (3) implantation of Silastic® capsules or Alzet® minipumps that lead to steady-state exposures. The rationale for the dosing method must also be stated. For example, minipump implants model continuous exposure and avoid the first-pass metabolism of BPA in the liver that results from oral exposure.

The positive control chosen must be compatible with the selected route of exposure. For example, E2 has very low activity when administered orally. For studies in which BPA is administered orally, DES or ethinylestradiol are appropriate positive controls. When BPA is administered in constant release capsules, E2 would be an appropriate positive control, while an estrogen agonist with a long half-life would be inappropriate. Route of administration influences the rate of metabolism of BPA, at least in adults [21], and there is some evidence for a higher contribution from ingestion of BPA in humans relative to inhalation or absorption through the skin [22]. However, the report that BPA levels in plasma collected from pregnant women in Germany show a range encompassing two orders of magnitude, between 0.1 and 10 parts per billion [23], suggests the likely possibility of variable exposure to multiple sources of BPA. Thus, although oral delivery appears to be most relevant for extrapolation to humans, all delivery methods may reveal effects of BPA.

When animals are assigned to groups, the litter must be taken into account. It is well established that for both outbred stocks and inbred strains, the litter is a significant source of variation and needs to be accounted for in assigning animals to groups. Ideally, one animal per litter should be used. In cases when this is not possible, there are statistical methods (such as including litter as a main effect variable and dividing the F value for treatment effects by the F value for litter effects) that can be used when more than one animal per litter is used, or litter can be used as a covariate in ANCOVA. The method used to avoid confounding litter effects must be reported.

For all experiments, the positive and negative controls must be clearly identified. Experiments using a replicate block design should include a positive and a negative control in each block, if possible. Experiments showing no effect and lacking positive control data cannot be interpreted.

When taking measurements from adult animals, care must be taken to normalize the reproductive state of the experimental animals. Males should be singly housed for 2–4 weeks before collection, in order to avoid physiological differences arising from differences in dominance status of the males. Normalization of adult female reproductive status can be achieved by assaying their estrous cycles and collecting females at the same stage, or by using ovariectomized females with or without hormone replacement [24].

Low dose effects of environmental endocrine disrupting chemicals generally refer to effects being reported at doses lower than those used in traditional toxicological studies for risk assessment purposes. “Low dose” is also commonly used to refer to environmentally relevant doses, i.e., doses resulting in serum levels close to those observed in human serum. For BPA, prior to 1997, the lowest dose studied for risk assessment purposes was 50 mg/(kg day), which in the USA remains the currently accepted lowest observed adverse effect level (LOAEL) that was used to calculate the current EPA reference dose (and FDA acceptable daily intake or ADI dose) of 50 μg/(kg day); this presumed “safe” dose is estimated by dividing the LOAEL by three 10-fold safety factors (i.e. by 1000) [25]. Thus, we included in our analysis studies dosing with less than 50 mg/(kg day) BPA (Table 3, Table 4).

Exposures to endocrine disruptors have different effects depending on the life stage of the exposed animals. Effects resulting from adult exposure are generally reversible and are termed “activational”. Effects resulting from exposure during organ development (beginning during prenatal development and continuing in postnatal life through puberty) may result in persistent alterations of the affected systems, even in the absence of subsequent exposure; these effects are termed “organizational” [26]. Some organizational effects are measurable immediately upon exposure and persist throughout the life of the animal [16]. Other organizational effects are undetectable at the time of exposure, but they become apparent in subsequent adulthood [26], [27]. Windows of vulnerability, also known as critical periods, during which the developing system is most sensitive to exposure, are common features of organizational effects [28]. Exposures occurring outside the critical periods will not elicit organizational effects. There is evidence that organizational effects of estrogenic endocrine disruptors such as BPA are mediated by epigenetic alterations in DNA [29]. Organizational and activational effects on the same tissue often differ qualitatively as well as in duration and in the dose required to elicit effects. In this review, we will first discuss organizational effects of prenatal through pubertal BPA exposure, termed “developmental effects” and then activational effects of adult BPA exposure, termed “adult effects”.

Section snippets

Developmental effects of BPA due to exposure during gestation through puberty

Many studies have examined the effects of prenatal, neonatal (shortly after birth) and lactational (birth through weaning) exposure to low doses of BPA. These experiments involved examining effects of exposure to low doses of BPA during “critical periods” in the development of different tissues. These critical periods continue through puberty, the period of physiological transition to fertility.

Effects of BPA exposure during adulthood

BPA exposure at low doses has diverse activational effects. Some of these effects are predicted due to the affinity of BPA for ERα and ERβ, while other effects diverge from those observed in response to activation of estrogen receptors.

Comparison of findings of significant effects and no-significant effects in low dose BPA studies

As of the end of October 2006 we are aware of 27 in vivo studies reporting no significant effects in response to low doses of BPA. The variables that account for most of the studies that find no significant effects have recently been reviewed [5], [153]. Below we discuss a number of these variables.

Based on existing evidence, we are confident of the following

The criterion for an outcome being assessed as achieving this level (we are confident) is that multiple independent studies have shown the same or similar outcomes.

Acknowledgements

Financial support: This review was prepared in conjunction with the Bisphenol A Conference, Chapel Hill, NC, 28–29 November 2006. Support was provided by the National Institute of Environmental Health Sciences and the National Institute of Dental and Craniofacial Research, NIH, DHHS, the W.M. Keck Center for Behavioral Biology at NC State University, and from Commonweal.

References (199)

  • K. Kubo et al.

    Low dose effects of bisphenol A on sexual differentiation of the brain and behavior in rats

    Neurosci Res

    (2003)
  • H.B. Patisaul et al.

    Neonatal genistein or bisphenol-A exposure alters sexual differentiation of the AVPV

    Neurotoxicol Teratol

    (2006)
  • Y. Masuo et al.

    Motor hyperactivity caused by a deficit in dopaminergic neurons and the effects of endocrine disruptors: a study inspired by the physiological roles of PACAP in the brain

    Regul Pept

    (2004)
  • K. Mizuo et al.

    Prenatal and neonatal exposure to bisphenol-A affects the morphine-induced rewarding effect and hyperlocomotion in mice

    Neurosci Lett

    (2004)
  • A.M. Aloisi et al.

    Exposure to the estrogenic pollutant bisphenol A affects pain behavior induced by subcutaneous formalin injection in male and female rats

    Brain Res

    (2002)
  • B.C. Ryan et al.

    Developmental exposure to environmental estrogens alters anxiety and spatial memory in female mice

    Horm Behav

    (2006)
  • F. Farabollini et al.

    Perinatal exposure to the estrogenic pollutant bisphenol A affects behavior in male and female rats

    Pharmacol Biochem Behav

    (1999)
  • S. Porrini et al.

    Early exposure to a low dose of bisphenol A affects socio-sexual behavior of juvenile female rats.

    Brain Res Bull

    (2005)
  • T. Suzuki et al.

    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)
  • G. Laviola et al.

    d-Amphetamine-related reinforcing effects are reduced in mice exposed prenatally to estrogenic endocrine disruptors.

    Brain Res Bull

    (2005)
  • D. Della Seta et al.

    Pubertal exposure to estrogenic chemicals affects behavior in juvenile and adult male rats

    Horm Behav

    (2006)
  • T. Funabashi et al.

    Exposure to bisphenol A during gestation and lactation causes loss of sex difference in corticotropin-releasing hormone-immunoreactive neurons in the bed nucleus of the stria terminalis of rats

    Psychoneuroendocrinology

    (2004)
  • T. Fujimoto et al.

    Prenatal exposure to bisphenol A impairs sexual differentiation of exploratory behavior and increases depression-like behavior in rats

    Brain Res

    (2006)
  • T. Takeuchi et al.

    Serum bisphenol A concentrations showed gender differences, possibly linked to androgen levels

    Biochem Biophys Res Commun

    (2002)
  • N. Hanioka et al.

    Interaction of bisphenol A with rat hepatic cytochrome P450 enzymes

    Chemosphere

    (2000)
  • T.J. Murray et al.

    Induction of mammary gland ductal hyperplasias and carcinoma in situ following fetal bisphenol A exposure

    Reprod Toxicol

    (2007)
  • J.B. Colerangle et al.

    Profound effects of the weak environmental estrogen-like chemical bisphenol A on the growth of the mammary gland of Noble rats

    J Steroid Biochem Mol Biol

    (1997)
  • G. Schönfelder et al.

    Developmental effects of prenatal exposure to bisphenol A on the uterus of rat offspring

    Neoplasia

    (2004)
  • G. Schönfelder et al.

    In utero exposure to low doses of bisphenol A lead to long-term deleterious effects in the vagina

    Neoplasia

    (2002)
  • P.A. Hunt et al.

    Bisphenol A causes meiotic aneuploidy in the female mouse

    Curr Biol

    (2003)
  • R.G. Berger et al.

    Influence of oral and subcutaneous bisphenol-A on intrauterine implantation of fertilized ova in inseminated female mice

    Reprod Toxicol

    (2007)
  • G.S. Prins et al.

    Developmental estrogen exposures predispose to prostate carcinogenesis with aging

    Reprod Toxicol

    (2007)
  • B.G. Timms et al.

    Prostate gland growth during development is stimulated in both male and female rat fetuses by intrauterine proximity to female fetuses

    J Urol

    (1999)
  • E. Burridge

    Bisphenol A: product profile

    Eur Chem News

    (2003)
  • Y. Kawagoshi et al.

    Estrogenic chemicals and estrogenic activity in leachate from municipal waste landfill determined by yeast two-hybrid assay

    J Environ Monitor

    (2003)
  • A. Coors et al.

    Removal of estrogenic activity from municipal waste landfill leachate assessed with a bioassay based on reporter gene expression

    Environ Sci Technol

    (2003)
  • F.S. vom Saal et al.

    An extensive new literature concerning low dose effects of bisphenol A shows the need for a new risk assessment

    Environ Health Perspect

    (2005)
  • W.V. Welshons et al.

    Large effects from small exposures. I. Mechanisms for endocrine-disrupting chemicals with estrogenic activity

    Environ Health Perspect

    (2003)
  • W.V. Welshons et al.

    Large effects from small exposures. III. Endocrine mechanisms mediating effects of bisphenol A at levels of human exposure

    Endocrinology

    (2006)
  • H.J. Lee et al.

    Antiandrogenic effects of bisphenol A and nonylphenol on the function of androgen receptor

    Toxicol Sci

    (2003)
  • R.T. Zoeller et al.

    Bisphenol-A, an environmental contaminant that acts as a thyroid hormone receptor antagonist in vitro, increases serum thyroxine, and alters RC3/neurogranin expression in the developing rat brain

    Endocrinology

    (2005)
  • C. Gupta

    Reproductive malformation of the male offspring following maternal exposure to estrogenic chemicals

    Proc Soc Exp Biol Med

    (2000)
  • C. Gupta

    The role of estrogen receptor, androgen receptor and growth factors in diethylstilbestrol-induced programming of prostate differentiation

    Urol Res

    (2000)
  • C.A. Richter et al.

    Estradiol and bisphenol A stimulate androgen receptor and estrogen receptor gene expression in fetal mouse prostate mesenchyme cells

    Environ Health Perspect

    (2007)
  • B.G. Timms et al.

    Estrogenic chemicals in plastic and oral contraceptives disrupt development of the mouse prostate and urethra

    Proc Natl Acad Sci USA

    (2005)
  • F.S. vom Saal et al.

    Prostate enlargement in mice due to fetal exposure to low doses of estradiol or diethylstilbestrol and opposite effects at high doses

    Proc Natl Acad Sci USA

    (1997)
  • J.E. Thigpen et al.

    Dietary phytoestrogens accelerate the time of vaginal opening in immature CD-1 mice

    Comp Med

    (2003)
  • K.L. Howdeshell et al.

    Bisphenol A is released from used polycarbonate animal cages into water at room temperature

    Environ Health Perspect

    (2003)
  • L.H. Pottenger et al.

    The relative bioavailability and metabolism of bisphenol A in rats is dependent upon the route of administration

    Toxicol Sci

    (2000)
  • G. Schönfelder et al.

    Parent bisphenol A accumulation in human maternal-fetal-placental unit

    Environ Health Perspect

    (2002)
  • Cited by (0)

    The information in this document has been subjected to review by the National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, and approved for publication. Approval does not signify that the contents reflect the views of the Agency, nor does mention of trade names or commercial products constitute endorsement or recommendation for use.

    View full text