Open access peer-reviewed chapter

Biotransformation of Bisphenol A and Its Adverse Effects on the Next Generation

Written By

Hidetomo Iwano, Hiroki Inoue, Miyu Nishikawa, Jumpei Fujiki and Hiroshi Yokota

Submitted: 17 February 2018 Reviewed: 03 May 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.78275

From the Edited Volume

Endocrine Disruptors

Edited by Ahmed R. G.

Chapter metrics overview

1,799 Chapter Downloads

View Full Metrics

Abstract

Although we are exposed to many chemical substances in routine daily life, the body has metabolic systems capable of detoxifying and eliminating these chemicals. Bisphenol A (BPA) is an endocrine disrupter of great concern because of its estrogenic activity, but studies have indicated no severe adverse effects in adult rodents exposed to BPA due to metabolic detoxification. BPA is metabolized by glucuronidation mediated by phase II enzymes such as UDP-glucuronosyltransferase. Numerous recent studies in rodents have indicated that maternal BPA exposure causes adverse effects in offspring. It was also shown that bisphenol analogs are efficiently absorbed via the oral route and distributed to the reproductive tract in pregnant rats, with its residue capable of crossing the placental barrier in the late stage of gestation. Both animal and human studies have demonstrated that BPA and the BPA metabolite BPA-GA are detectable in fetal and amniotic fluid, suggesting the presence of a placental transfer mechanism. In this review, we discuss the pharmacokinetics of BPA, particularly its (1) metabolism and disposition in the intestine, (2) metabolism and disposition in the liver, and (3) transfer from maternal tissues to the fetus.

Keywords

  • bisphenol A
  • UDP-glucuronosyltransferase (UGT)
  • multidrug resistance-associated protein (MRP)
  • organic anion-transporting polypeptide (Oatp)
  • xenobiotic-metabolizing enzymes (XMEs)
  • liver perfusion
  • β-glucuronidase

1. Introduction

Bisphenol A (BPA; 2,2-bis[4-hydroxyphenyl]propane) is an industrial chemical widely used in the manufacture of polycarbonate plastics and epoxy resin liners for aluminum cans [1, 2, 3, 4, 5, 6, 7]. BPA is an endocrine-disrupting chemical (EDC) that has been demonstrated to affect reproductive organ development [7, 8, 9], brain development [10, 11, 12, 13, 14, 15], metabolic diseases [16], and postnatal behavior [17, 18, 19]. These adverse effects are thought to be due to disturbed signaling mechanisms involving estrogen, androgen, and thyroid hormone.

BPA introduced into the body orally must pass through the gastrointestinal tract and liver before arriving at target tissues such as the uterus, testes, or fetus. To elucidate the mechanism responsible for the adverse effects of BPA, it is essential to clarify the fate of the compound during its passage through the hepatointestinal pathway. The hepatointestinal pathway serves as a protective barrier against a variety of potentially harmful chemicals due to the activity of potent xenobiotic-metabolizing enzymes (XMEs), which can be classified into three main categories [20]. The first category consists of phase I enzymes, mainly of the cytochrome P450 (CYP) family [21]. Most drugs are metabolized by CYPs, either during detoxification or due to the activation of a pathway for an inactive prodrug. The second XME category consists of what are referred to as phase II enzymes [22, 23, 24]. These enzymes usually conjugate phase I products but can also conjugate other intermediate compounds and intracellular substrates, such as steroids and bilirubin. The third category of XMEs consists of drug transporters, which are membrane-bound proteins involved in drug uptake or excretion [25, 26].

Detoxification enzymes have been shown to play a pivotal role in the elimination of ingested chemicals from the intestinal wall and liver. In rat liver, BPA is metabolized by phase II enzymes via glucuronidation, which is mediated by UDP-glucuronosyltransferase (UGT, Enzyme Classification 2.4.1.17), primarily the UGT2B1 isoform [27]. Glucuronidation is a major elimination process that converts lipophilic substrates to hydrophilic molecules that are readily excreted via the bile and urine [20, 28]. Glucuronidation is the main pathway by which BPA is metabolized to a hydrophilic form lacking estrogenic activity. We were the first to report that BPA is highly glucuronidated in rat liver [27]. To clearly elucidate the metabolism and disposition of BPA in the body, we thought it was important to conduct an investigation at the tissue level, and we therefore carried out tissue perfusion experiments.

In this chapter, we discuss the metabolism and disposition of BPA, particularly in the gastrointestinal tract and liver, as well as why the fetus is so easily affected by BPA. The investigation specifically focused on the following: (1) metabolism and disposition of BPA in the intestine, (2) metabolism and disposition of BPA in the liver, and (3) transfer of BPA from maternal tissues to the fetus.

1.1. Metabolism and disposition of BPA in the intestine

This section focuses on the absorption and metabolism of BPA in the intestine. For a thorough investigation of the metabolism of BPA in the intestine, we adopted a method using a segment of everted intestine (Figure 1A) [29]. We find that in the intestine of Sprague-Dawley rats exposed to BPA, (1) most of the compounds absorbed by the intestine are glucuronidated within the intestinal wall and (2) the resulting GA is preferentially eliminated into the mucosal side of the small intestine and the serosal side of the colon (Figure 1B and C).

Figure 1.

Overview of the metabolism and disposition of BPA in the intestine. (A) Schema of method using a segment of everted intestine. (B) Metabolism and disposition of BPA in small intestinal mucosal cells. BPA was highly glucuronidated through the lumen of the rat intestine. (C) Summary of BPA metabolism in the intestinal tract. Mucosal excretion of BPA-GA greatly exceeded serosal excretion in the proximal intestine; however, the direction of elimination was reversed in the colon.

These results suggest that the proximal intestine plays a highly protective role against ingested BPA. BPA was highly glucuronidated during its passage through the lumen of the rat intestine, with most of the compounds being excreted to the mucosal side as BPA-GA, which is low in estrogenic activity (Figure 2B) [30]. This was particularly evident in the proximal jejunum, where mucosal excretion of BPA-GA greatly exceeded serosal excretion. Therefore, it appears that the proximal jejunum defends against the potentially adverse effects of orally introduced BPA by limiting entry of the free compound into the bloodstream and curtailing exposure to the middle and distal parts of the intestine. These results are in line with previous reports of low exposure to the compound in association with the oral intake of BPA [31, 32, 33, 34, 35, 36]. Comparing the concentration-time profiles of BPA in the blood of F344 rats exposed to the compound intraperitoneally with those of rats exposed orally, Pottenger et al. [31] found that oral administration results in lower exposure to unconjugated BPA. In light of these findings, the diminution of exposure to unconjugated BPA on oral administration may be ascribed to the high degree of glucuronidation of the compound in the proximal intestine, which is the foremost barrier to damage from oral administration. We previously reported that BPA glucuronidation in the liver is mediated by UGT2B1, an isoform of UDP-glucuronosyltransferase, and that this isoform is not expressed in rat intestine [27]. Generally, the UGT2B family glucuronidates steroid hormones [37]. In humans, several UGT isoforms are known to conjugate BPA. UGT2B15 reportedly has the highest activity, with lower activities reported for recombinant UGT1A1, UGT1A3, UGT1A9, UGT2B4, and UGT2B7 [38]. The results of a recent analysis of a mouse cell line in which all UGT 2B genes were deleted suggest that members of the UGT1 family play a major role in BPA glucuronidation [39], which in turn suggests that the UGT1 family also plays a major role in intestinal BPA glucuronidation.

Figure 2.

Overview of the metabolism and disposition of BPA in the liver. (A) Schema of the liver perfusion method. (B) Metabolism and disposition of BPA in hepatocytes. (C) Summary of BPA metabolism in the liver. BPA is conjugated primarily to monoglucuronide and partially BPA-glucuronide/sulfate diconjugate in males, but otherwise almost no diconjugate was detected in female rats. Most metabolites were excreted into the bile, but during pregnancy, bilious excretion of BPA-GA decreases, and reciprocally, venous excretion increases.

Although BPA-GA was excreted into the mucosal side of the small intestine, the direction of elimination was reversed in the colon, where excretion was into the serosal side (Figure 2C). ATP-dependent transporters have been described as mediating the transport of GA-conjugated compounds across the cell membrane [40]. In rat liver, a member of the ATP-binding cassette (ABC) transporter family, namely multidrug resistance-associated protein (MRP), is capable of mediating transmembrane excretion of a wide range of amphipathic compounds, including bilirubin-, estrogen-, and xenobiotic-GA [41]. In rat intestine, MRP2, localized in the apical domain of enterocytes, is distributed in the proximal intestine [42], and MRP3, localized in the basolateral domain, is distributed mainly in the ileum and colon [43]. Intriguingly, the apical and basolateral directions of BPA-GA excretion in the present study paralleled the distribution patterns of MRP2 and MRP3, respectively. Other reports have indicated that MRP2 is highly expressed in the proximal intestine, whereas MRP3 and MRP4 are highly expressed in the colon [44]. As MRP3 and MRP4 are expressed in the basolateral domain of the liver and intestine [45], the supposition may be made that the elimination direction of BPA-GA is governed by the distribution of an organic anion transporter system such as MRP.

As large amounts of BPA-GA are eliminated from the lumen, the excreted GA would presumably flow into the distal intestine with the luminal contents. In the colon, GA would most likely be deconjugated by lumen bacterial β-glucuronidase, an enzyme known to generate toxic and carcinogenic substances [46]. Deconjugation by lumen bacterial β-glucuronidase is known to be involved in the reactivation of an antitumor compound derived from irinotecan [47]. Furthermore, as excreted BPA-GA is deconjugated by bacterial β-glucuronidase in the cecum, free BPA is detected only in the colon and feces [48]. In light of these previous findings, the notable absorption and transport of unconjugated BPA to the serosal side of the rat colon observed in this study suggests that deconjugated BPA is eventually reabsorbed by the colon.

Generally, the paramount issue in studies of the adverse effects of BPA concerns oral exposure to the chemical in low doses [7, 49]. Although Rubin et al. [50] described adverse effects in rat offspring after maternal administration, other studies have found no adverse effects [51, 52]. Therefore, the toxicity of low doses of BPA remains controversial. We believe that an animal’s sensitivity to ingested BPA reflects the conditions inside the intestine (e.g., the luminal contents and composition of the bacterial flora). Further studies are required to clarify the correlation between the catalytic reactivation of BPA-GA by the luminal flora and any resulting adverse effects.

1.2. Metabolism and disposition of BPA in the liver

Because environmental estrogens introduced orally are absorbed by the gastrointestinal tract and consequently the liver before being distributed throughout the body, it is important to trace their fate before they reach the reproductive organs. This section focuses on the metabolism and disposition of BPA in the liver. To facilitate thorough investigation of the metabolism of BPA in the liver, we adopted a liver perfusion assay in a previous study (Figure 2A and B) [53, 54, 55]. The results showed that (1) in Sprague-Dawley rats, most BPA absorbed by the intestine is likely glucuronidated in the liver, (2) the resulting BPA-GA is excreted into the bile and venous blood, and (3) in pregnant rats, there is a slight but significant decrease in bilious excretion of BPA-GA, which results in a reciprocal increase in venous excretion (Figure 2C).

These findings are in line with those from a study reporting that BPA added to the culture medium of isolated hepatocytes is highly metabolized to BPA-GA [56]. Moreover, previous studies of BPA pharmacokinetics and metabolism in rats provided evidence that the major metabolite in the plasma is BPA monoglucuronide conjugates [31, 32, 33]. Therefore, glucuronidation is a major pathway of BPA metabolism in the liver. After glucuronidation, the conjugates must be excreted from the hepatocytes into the bile and venous blood. Intriguingly, in male rats, approximately one-fourth of infused BPA was eliminated as a GA/sulfate diconjugate, whereas this diconjugate was virtually absent in female rats (Figure 2). Suiko et al. reported that BPA is conjugated with sulfate by several forms of human sulfotransferase [57]. We recently reported the results of rat liver perfusion experiments in which we found that BPA is conjugated primarily to monoglucuronide; in males, we found that diconjugate (GA/sulfate diconjugate) production occurs under conditions of high-dose BPA exposure [53]. These findings agree with those from a previous study in which BPA added to the medium of isolated hepatocytes was metabolized into both monoglucuronide and diconjugate; moreover, almost no diconjugate was detected in female rats [56]. BPA sulfoconjugation is mediated by phenol sulfotransferase isoforms of the SULT1 family [58]. One member of the SULT1 family, SULT1A1, exhibits a high conjugation activity toward BPA [59]. The expression level of SULT1 family enzymes is estimated to be higher in male than in female rats [60].

During pregnancy, the bilious excretion of BPA-GA decreases, and reciprocally, venous excretion increases. A wide variety of drug conjugates are transported by members of the ABC transporter family known as glutathione-S-conjugate export pumps. MRP2, a pump that is expressed primarily in the canalicular membrane of hepatocytes, transports drug GA to the bile [45]. Both the hepatic expression and function of MRP2 decrease in pregnant rats [61]. Regarding sinusoidal excretion, MRP1 and MRP3 have been shown to mediate chemical-GA transport [62, 63, 64], and the expression of MRP3 is attenuated in pregnancy [61]. Together with our previous results regarding liver perfusion in Eisai hyperbilirubinemic rats [54], these findings give rise to the view that a low expression of MRP2 in pregnancy limits the transport rate of BPA-GA into the bile and that sinusoidal transport systems such as MRP1 and MRP3 compensate by transporting GA to the venous blood.

1.3. Transfer of BPA from the maternal side to the fetus

Venous BPA-GA excreted from the liver enters the systemic blood circulation. Pottenger et al. [31] showed that BPA-GA can be detected in the urine after administration of BPA; therefore, BPA-GA is excreted into the urine. However, certain organs, such as the lungs, small intestines, and placenta, show a high β-glucuronidase activity [65, 66]. BPA-GA can be cleaved in these organs, and it can be predicted that the resultant BPA moves to the lower organs supplied by the bloodstream. In the placenta, β-glucuronidase activity leads to fetal exposure to BPA. Kushari and Mukherjea [67] reported that placental β-glucuronidase activity is present during early gestation in humans, which is a highly vulnerable period for the developing fetus. An important concern, however, is that previous investigators reported that the placenta exhibits a minimal glucuronidation activity [68, 69]. We also demonstrated that BPA-GA is transported to the fetus following uterine perfusion and that BPA-GA and deconjugated BPA can be detected in the fetus and amniotic fluid due to a high deconjugation activity and vulnerable drug metabolism in the fetus [70]. After oral administration of 10 mg/kg 14C-BPA to GD16.0 rat mothers, Domoradzki et al. found that BPA-GA was concentrated in the fetus [32]. Kurebayashi et al. also detected radioactivity in GD18 fetal tissues 24 h after oral administration of 14C-BPA to pregnant rats, but they found no radioactivity in GD13 or GD15 fetuses [35]. Therefore, BPA-GA may be transferred across the placenta to the fetus by placental transporters that mediate the transfer of essential endogenous physiologic estrogenic compounds (Figure 3).

Figure 3.

Illustration of the metabolism and disposition of BPA during pregnancy.

BPA is highly glucuronidated through the lumen of the rat intestine. In the intestine, serosal excretion of BPA metabolite is probably BPA-GA, and partially free BPA may be also transported into the portal vein. In the liver, BPA is conjugated primarily to monoglucuronide and partially BPA-glucuronide/sulfate diconjugate in males. During pregnancy, bilious excretion of BPA-GA decreases, and reciprocally, venous excretion may increase through MRP. BPA-GA remaining in systemic blood circulation is metabolized by placental or fetal β-glucuronidase, and the resultant BPA would permeate the fetal tissues. MRP, multidrug resistance-associated protein; UGT, UDP-glucuronosyltransferase; ST, sulfotransferase.

Some members of the Oatp [71, 72, 73] and Mrp [74, 75] transporter families are known to transport conjugates of steroid hormones such as DHEAS and 17β-estradiol-GA, suggesting that BPA-GA is transported across the placenta by these transporters. In light of the studies cited above and our present results, we surmise that if BPA-GA remaining in systemic blood circulation is metabolized by placental or fetal β-glucuronidase, the resultant BPA would permeate the fetal tissues (Figure 3). Due to a low UGT2B1 expression in fetal rat liver, we also reported that this metabolic system is weak in the fetus [70, 76, 77]. Numerous recent studies in rodents have found that maternal BPA exposure causes adverse effects in the offspring [17, 78, 79, 80, 81, 82, 83, 84]. In light of these findings, the present results suggest that the risk of BPA exposure to the fetus is high, despite preservation of BPA glucuronidation in the maternal liver.

Advertisement

2. Conclusion(s)

Many reports have suggested that human health may be affected by exposure to even low levels of BPA, especially during the gestation period. However, the detailed mechanisms of BPA’s effects remain unknown. To further elucidate the mechanism governing the detrimental effects of EDCs on target organs, it is essential to clarify both the metabolism and elimination pathways of such chemicals in the body. However, BPA is highly glucuronidated in the intestine and liver, and the resultant formation of BPA-GA prevents a complete understanding of metabolism and disposition by facilitating deconjugation during enterohepatic circulation and systematic circulation in the body. Given that exposure to BPA could adversely affect the fetus in pregnant animals, it is critical that further work be done to determine the fate of venous GA compounds in the complete BPA pathway before excretion.

In modern society, we are continually exposed to many chemical substances. We have to deal with all of these chemicals to ensure good health. Many studies of the effects of chemical substances have focused only on terminal mechanisms. We originally developed the prominent drug metabolism systems to eliminate various chemicals in the process of evolution. The various mechanisms that determine the effects of EDCs can only be productively discussed after a more complete understanding of their metabolism systems is achieved. At that point, new precautions to avoid the risks of adverse effects could be developed.

Advertisement

Acknowledgments

This study was supported by the Rakuno Gakuen University Research Fund (No. 2018-04).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Brotons JA, Olea-Serrano MF, Villalobos M, Pedraza V, Olea N. Xenoestrogens released from lacquer coatings in food cans. Environmental Health Perspectives. 1995;103:608-612
  2. 2. Krishnan AV, Stathis P, Permuth SF, Tokes L, Feldman D. Bisphenol A: An estrogenic substance is released from polycarbonate flasks during autoclaving. Endocrinology. 1993;132:2279-2286. DOI: 10.1210/en.132.6.2279
  3. 3. vom Saal 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;14:239-260
  4. 4. vom Saal FS, Hughes C. An extensive new literature concerning low-dose effects of bisphenol A shows the need for a new risk assessment. Environmental Health Perspectives. 2005;113:926-933
  5. 5. European Food Safety Authority (EFSA). Scientific opinion on the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs: Executive summary EFSA Panel on Food Contact Materials, Enzymes, Flavourings and Processing Aids (CEF). EFSA Journal. 2015;13(1):3978-4599
  6. 6. Muhamad MS, Salim MR, Lau WJ, Yusop Z. A review on bisphenol A occurrences, health effects and treatment process via membrane technology for drinking water. Environmental Science and Pollution Research. 2016;23:11549-11567. DOI: 10.1007/s11356-016-6357-2
  7. 7. Peretz J, Vrooman L, Ricke WA, Hunt PA, Ehrlich S, Hauser R, et al. Bisphenol A and reproductive health: Update of experimental and human evidence, 2007-2013. Environmental Health Perspectives. 2014;122:775-786. DOI: 10.1289/ehp.1307728
  8. 8. Salian S, Doshi T, Vanage G. Impairment in protein expression profile of testicular steroid receptor coregulators in male rat offspring perinatally exposed to bisphenol A. Life Sciences. 2009;85:11-18. DOI: 10.1016/j.lfs.2009.04.005
  9. 9. Timms BG, Howdeshell KL, Barton L, Bradley S, Richter CA, vom Saal FS. Estrogenic chemicals in plastic and oral contraceptives disrupt development of the fetal mouse prostate and urethra. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:7014-7019. DOI: 10.1073/pnas.0502544102
  10. 10. Komada M, Asai Y, Morii M, Matsuki M, Sato M, Nagao T. Maternal bisphenol A oral dosing relates to the acceleration of neurogenesis in the developing neocortex of mouse fetuses. Toxicology. 2012;295:31-38. DOI: 10.1016/j.tox.2012.02.013
  11. 11. Patisaul HB, Polston EK. Influence of endocrine active compounds on the developing rodent brain. Brain Research Reviews. 2008;57:352-362. DOI: 10.1016/j.brainresrev.2007.06.008
  12. 12. 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. DOI: 10.1210/en.2006-0189
  13. 13. Ahmed RG. Maternal bisphenol A alters fetal endocrine system: Thyroid adipokine dysfunction. Food and Chemical Toxicology. 2016 Sep;95:168-174. DOI: 10.1016/j.fct.2016.06.017
  14. 14. Ahmed RG, Walaa GH, Asmaa FS. Suppressive effects of neonatal bisphenol A on the neuroendocrine system. Toxicology and Industrial Health. Jan 1, 2018;34:397-407. DOI: 10.1177/0748233718757082
  15. 15. Kimura E, Matsuyoshi C, Miyazaki W, Benner S, Hosokawa M, Yokoyama K, et al. Prenatal exposure to bisphenol A impacts neuronal morphology in the hippocampal CA1 region in developing and aged mice. Archives of Toxicology. 2016;90:691-700. DOI: 10.1007/s00204-015-1485-x
  16. 16. Liu J, Yu P, Qian W, Li Y, Zhao J, Huan F, et al. Perinatal bisphenol A exposure and adult glucose homeostasis: Identifying critical windows of exposure. PLoS One. 2013;8:e64143. DOI: 10.1371/journal.pone.0064143
  17. 17. Kubo K, Arai O, Ogata R, Omura M, Hori T, Aou S. Exposure to bisphenol A during the fetal and suckling periods disrupts sexual differentiation of the locus coeruleus and of behavior in the rat. Neuroscience Letters. 2001;304:73-76. DOI: 10.1016/s0304-3940(01)01760-8
  18. 18. De Coster S, van Larebeke N. Endocrine-disrupting chemicals: Associated disorders and mechanisms of action. Journal of Environmental and Public Health. 2012;2012:713696. DOI: 10.1155/2012/713696
  19. 19. Ohtani N, Suda K, Tsuji E, Tanemura K, Yokota H, Inoue H, et al. Late pregnancy is vulnerable period for exposure to BPA. The Journal of Veterinary Medical Science. 2018;80:536-543. DOI: 10.1292/jvms.17-0460
  20. 20. Bock KW. Homeostatic control of xeno- and endobiotics in the drug-metabolizing enzyme system. Biochemical Pharmacology. 2014;90:1-6. DOI: 10.1016/j.bcp.2014.04.009
  21. 21. Nelson DR, Kamataki T, Waxman DJ, Guengerich FP, Estabrook RW, Feyereisen R, et al. The P450 superfamily: Update on new sequences, gene mapping, accession numbers, early trivial names of enzymes, and nomenclature. DNA and Cell Biology. 1993;12:1-51. DOI: 10.1089/dna.1993.12.1
  22. 22. MacKenzie PI, Owens IS, Burchell B, Bock KW, Bairoch A, Belanger A, et al. The UDP glycosyltransferase gene superfamily: Recommended nomenclature update based on evolutionary divergence. Pharmacogenetics. 1997;7:255-269. DOI: 10.1097/00008571-199708000-00001
  23. 23. Mackenzie PI, Gregory PA, Gardner-Stephen DA, Lewinsky RH, Jorgensen BR, Nishiyama T, et al. Regulation of UDP glucuronosyltransferase genes. Current Drug Metabolism. 2003;4:249-257
  24. 24. Di Pietro G, Magno LA, Rios-Santos F. Glutathione S-transferases: An overview in cancer research. Expert Opinion on Drug Metabolism & Toxicology. 2010;6:153-170. DOI: 10.1517/17425250903427980
  25. 25. Hediger MA, Romero MF, Peng JB, Rolfs A, Takanaga H, Bruford EA. The ABCs of solute carriers: Physiological, pathological and therapeutic implications of human membrane transport proteins: Introduction. Pflügers Archiv. 2004;447:465-468. DOI: 10.1007/s00424-003-1192-y
  26. 26. Holland IB. ABC transporters, mechanisms and biology: An overview. Essays in Biochemistry. 2011;50:1-17. DOI: 10.1042/bse0500001
  27. 27. Yokota H, Iwano H, Endo M, Kobayashi T, Inoue H, S-i I, et al. Glucuronidation of the environmental oestrogen bisphenol A by an isoform of UDP-glucuronosyltransferase, UGT2B1, in the rat liver. The Biochemical Journal. 1999;340:405-409. DOI: 10.1042/0264-6021:3400405
  28. 28. Bock KW. The UDP-glycosyltransferase (UGT) superfamily expressed in humans, insects and plants: Animal-plant arms-race and co-evolution. Biochemical Pharmacology. 2016;99:11-17. DOI: 10.1016/j.bcp.2015.10.001
  29. 29. Inoue H, Yuki G, Yokota H, Kato S. Bisphenol A glucuronidation and absorption in rat intestine. Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2003;31:140-144
  30. 30. Matthews JB, Twomey K, Zacharewski TR. In vitro and in vivo interactions of bisphenol A and its metabolite, bisphenol A glucuronide, with estrogen receptors alpha and beta. Chemical Research in Toxicology. 2001;14:149-157
  31. 31. Pottenger LH, Domoradzki JY, Markham DA, Hansen SC, Cagen SZ, Waechter JM Jr. The relative bioavailability and metabolism of bisphenol A in rats is dependent upon the route of administration. Toxicological Sciences. 2000;54:3-18
  32. 32. Domoradzki JY, Pottenger LH, Thornton CM, Hansen SC, Card TL, Markham DA, et al. Metabolism and pharmacokinetics of bisphenol A (BPA) and the embryo-fetal distribution of BPA and BPA-monoglucuronide in CD Sprague-Dawley rats at three gestational stages. Toxicological Sciences. 2003;76:21-34. DOI: 10.1093/toxsci/kfg206
  33. 33. Domoradzki JY, Thornton CM, Pottenger LH, Hansen SC, Card TL, Markham DA, et al. Age and dose dependency of the pharmacokinetics and metabolism of bisphenol A in neonatal Sprague-Dawley rats following oral administration. Toxicological Sciences. 2004;77:230-242. DOI: 10.1093/toxsci/kfh054
  34. 34. Ginsberg G, Rice DC. Does rapid metabolism ensure negligible risk from bisphenol A? Environmental Health Perspectives. 2009;117:1639-1643. DOI: 10.1289/ehp.0901010
  35. 35. Kurebayashi H, Betsui H, Ohno Y. Disposition of a low dose of 14C-bisphenol A in male rats and its main biliary excretion as BPA glucuronide. Toxicological Sciences. 2003;73:17-25. DOI: 10.1093/toxsci/kfg040
  36. 36. Kurebayashi H, Nagatsuka S-I, Nemoto H, Noguchi H, Ohno Y. Disposition of low doses of C−14-bisphenol A in male, female, pregnant, fetal, and neonatal rats. Archives of Toxicology. 2005;79:243-252. DOI: 10.1007/s00204-004-0628-2
  37. 37. Turgeon D, Carrier JS, Levesque E, Hum DW, Belanger A. Relative enzymatic activity, protein stability, and tissue distribution of human steroid-metabolizing UGT2B subfamily members. Endocrinology. 2001;142:778-787. DOI: 10.1210/endo.142.2.7958
  38. 38. Hanioka N, Naito T, Narimatsu S. Human UDP-glucuronosyltransferase isoforms involved in bisphenol A glucuronidation. Chemosphere. 2008;74:33-36. DOI: 10.1016/j.chemosphere.2008.09.053
  39. 39. Fay MJ, Nguyen MT, Snouwaert JN, Dye R, Grant DJ, Bodnar WM, et al. Xenobiotic metabolism in mice lacking the UDP-glucuronosyltransferase 2 family. Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2015;43:1838-1846. DOI: 10.1124/dmd.115.065482
  40. 40. Oude Elferink RP, Meijer DK, Kuipers F, Jansen PL, Groen AK, Groothuis GM. Hepatobiliary secretion of organic compounds; molecular mechanisms of membrane transport. Biochimica et Biophysica Acta. 1995;1241:215-268
  41. 41. Yamazaki M, Suzuki H, Sugiyama Y. Recent advances in carrier-mediated hepatic uptake and biliary excretion of xenobiotics. Pharmaceutical Research. 1996;13:497-513
  42. 42. Mottino AD, Hoffman T, Jennes L, Cao J, Vore M. Expression of multidrug resistance-associated protein 2 in small intestine from pregnant and postpartum rats. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2001;280:G1261-G1273
  43. 43. Rost D, Mahner S, Sugiyama Y, Stremmel W. Expression and localization of the multidrug resistance-associated protein 3 in rat small and large intestine. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2002;282:G720-G726. DOI: 10.1152/ajpgi.00318.2001
  44. 44. Park S, Cheng SL, Cui JY. Characterizing drug-metabolizing enzymes and transporters that are bona fide CAR-target genes in mouse intestine. Acta Pharmaceutica Sinica B. 2016;6:475-491. DOI: 10.1016/j.apsb.2016.07.004
  45. 45. Kock K, Brouwer KL. A perspective on efflux transport proteins in the liver. Clinical Pharmacology and Therapeutics. 2012;92:599-612. DOI: 10.1038/clpt.2012.79
  46. 46. Reddy BS, Engle A, Simi B, Goldman M. Effect of dietary fiber on colonic bacterial enzymes and bile acids in relation to colon cancer. Gastroenterology. 1992;102:1475-1482
  47. 47. Kaneda N, Yokokura T. Nonlinear pharmacokinetics of CPT-11 in rats. Cancer Research. 1990;50:1721-1725
  48. 48. Sakamoto H, Yokota H, Kibe R, Sayama Y, Yuasa A. Excretion of bisphenol A-glucuronide into the small intestine and deconjugation in the cecum of the rat. Biochimica et Biophysica Acta. 2002;1573:171-176
  49. 49. Feldman D. Estrogens from plastic--are we being exposed? Endocrinology. 1997;138:1777-1779. DOI: 10.1210/endo.138.5.5213
  50. 50. Rubin BS, Murray MK, Damassa DA, King JC, Soto AM. Perinatal exposure to low doses of bisphenol A affects body weight, patterns of estrous cyclicity, and plasma LH levels. Environmental Health Perspectives. 2001;109:675-680
  51. 51. Cagen SZ, Waechter JM Jr, Dimond SS, Breslin WJ, Butala JH, Jekat FW, et al. Normal reproductive organ development in Wistar rats exposed to bisphenol A in the drinking water. Regulatory Toxicology and Pharmacology. 1999;30:130-139. DOI: 10.1006/rtph.1999.1340
  52. 52. Ema M, Fujii S, Furukawa M, Kiguchi M, Ikka T, Harazono A. Rat two-generation reproductive toxicity study of bisphenol A. Reproductive Toxicology. 2001;15:505-523
  53. 53. Inoue H, Kemanai S, Sano C, Kato S, Yokota H, Iwano H. Bisphenol A glucuronide/sulfate diconjugate in perfused liver of rats. The Journal of Veterinary Medical Science. 2016;78:733-737. DOI: 10.1292/jvms.15-0573
  54. 54. Inoue H, Tsuruta A, Kudo S, Ishii T, Fukushima Y, Iwano H, et al. Bisphenol A glucuronidation and excretion in liver of pregnant and nonpregnant female rats. Drug Metabolism and Disposition: the Biological Fate of Chemicals. 2005;33:55-59. DOI: 10.1124/dmd.104.001537
  55. 55. Inoue H, Yokota H, Makino T, Yuasa A, Kato S. Bisphenol a glucuronide, a major metabolite in rat bile after liver perfusion. Drug Metabolism and Disposition: : the Biological Fate of Chemicals. 2001;29:1084-1087
  56. 56. Pritchett JJ, Kuester RK, Sipes IG. Metabolism of bisphenol a in primary cultured hepatocytes from mice, rats, and humans. Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2002;30:1180-1185
  57. 57. Suiko M, Sakakibara Y, Liu MC. Sulfation of environmental estrogen-like chemicals by human cytosolic sulfotransferases. Biochemical and Biophysical Research Communications. 2000;267:80-84. DOI: 10.1006/bbrc.1999.1935
  58. 58. Shimizu M, Ohta K, Matsumoto Y, Fukuoka M, Ohno Y, Ozawa S. Sulfation of bisphenol A abolished its estrogenicity based on proliferation and gene expression in human breast cancer MCF-7 cells. Toxicology In Vitro. 2002;16:549-556
  59. 59. Nishiyama T, Ogura K, Nakano H, Kaku T, Takahashi E, Ohkubo Y, et al. Sulfation of environmental estrogens by cytosolic human sulfotransferases. Drug Metabolism and Pharmacokinetics. 2002;17:221-228
  60. 60. Klaassen CD, Liu L, Dunn RT 2nd. Regulation of sulfotransferase mRNA expression in male and female rats of various ages. Chemico-Biological Interactions. 1998;109:299-313
  61. 61. Cao J, Stieger B, Meier PJ, Vore M. Expression of rat hepatic multidrug resistance-associated proteins and organic anion transporters in pregnancy. The American Journal of Physiology. 2002;283:G757-G766
  62. 62. Soroka CJ, Lee JM, Azzaroli F, Boyer JL. Cellular localization and up-regulation of multidrug resistance-associated protein 3 in hepatocytes and cholangiocytes during obstructive cholestasis in rat liver. Hepatology. 2001;33:783-791. DOI: 10.1053/jhep.2001.23501
  63. 63. Ogawa K, Suzuki H, Hirohashi T, Ishikawa T, Meier PJ, Hirose K, et al. Characterization of inducible nature of MRP3 in rat liver. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2000;278:G438-G446
  64. 64. Roelofsen H, Muller M, Jansen PL. Regulation of organic anion transport in the liver. The Yale Journal of Biology and Medicine. 1997;70:435-445
  65. 65. Paigen K. Mammalian beta-glucuronidase: Genetics, molecular biology, and cell biology. Progress in Nucleic Acid Research and Molecular Biology. 1989;37:155-205
  66. 66. Sperker B, Backman JT, Kroemer HK. The role of beta-glucuronidase in drug disposition and drug targeting in humans. Clinical Pharmacokinetics. 1997;33:18-31
  67. 67. Kushari J, Mukherjea M. Studies on beta-glucuronidase of the developing human placenta. Gynecologic and Obstetric Investigation. 1980;11:119-127
  68. 68. Lucier GW, Sonawane BR, McDaniel OS. Glucuronidation and deglucuronidation reactions in hepatic and extrahepatic tissues during perinatal development. Drug Metabolism and Disposition. 1977;5:279-287
  69. 69. Juchau MR. Drug biotransformation in the placenta. Pharmacology & Therapeutics. 1980;8:501-524
  70. 70. Nishikawa M, Iwano H, Yanagisawa R, Koike N, Inoue H, Yokota H. Placental transfer of conjugated bisphenol A and subsequent reactivation in the rat fetus. Environmental Health Perspectives. 2010;118:1196-1203. DOI: 10.1289/ehp.0901575
  71. 71. Ugele B, St-Pierre MV, Pihusch M, Bahn A, Hantschmann P. Characterization and identification of steroid sulfate transporters of human placenta. American Journal of Physiology. Endocrinology and Metabolism. 2003;284:E390-E398. DOI: 10.1152/ajpendo.00257.2002
  72. 72. Cheng X, Maher J, Chen C, Klaassen CD. Tissue distribution and ontogeny of mouse organic anion transporting polypeptides (Oatps). Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2005;33:1062-1073. DOI: 10.1124/dmd.105.003640
  73. 73. Leazer TM, Klaassen CD. The presence of xenobiotic transporters in rat placenta. Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2003;31:153-167
  74. 74. Nagashige M, Ushigome F, Koyabu N, Hirata K, Kawabuchi M, Hirakawa T, et al. Basal membrane localization of MRP1 in human placental trophoblast. Placenta. 2003;24:951-958
  75. 75. Joshi AA, Vaidya SS, St-Pierre MV, Mikheev AM, Desino KE, Nyandege AN, et al. Placental ABC transporters: Biological impact and pharmaceutical significance. Pharmaceutical Research. 2016;33:2847-2878. DOI: 10.1007/s11095-016-2028-8
  76. 76. Matsumoto J, Yokota H, Yuasa A. Developmental increases in rat hepatic microsomal UDP-glucuronosyltransferase activities toward xenoestrogens and decreases during pregnancy. Environmental Health Perspectives. 2002;110:193-196
  77. 77. Yabusaki R, Iwano H, Tsushima S, Koike N, Ohtani N, Tanemura K, et al. Weak activity of UDP-glucuronosyltransferase toward bisphenol analogs in mouse perinatal development. The Journal of Veterinary Medical Science. 2015;77:1479-1484. DOI: 10.1292/jvms.15-0197
  78. 78. Farabollini F, Porrini S, Dessi-Fulgherit F. Perinatal exposure to the estrogenic pollutant bisphenol A affects behavior in male and female rats. Pharmacology, Biochemistry, and Behavior. 1999;64:687-694
  79. 79. Gioiosa L, Fissore E, Ghirardelli G, Parmigiani S, Palanza P. Developmental exposure to low-dose estrogenic endocrine disruptors alters sex differences in exploration and emotional responses in mice. Hormones and Behavior. 2007;52:307-316. DOI: 10.1016/j.yhbeh.2007.05.006
  80. 80. Jasarevic E, Sieli PT, Twellman EE, Welsh TH Jr, Schachtman TR, Roberts RM, et al. Disruption of adult expression of sexually selected traits by developmental exposure to bisphenol A. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:11715-11720. DOI: 10.1073/pnas.1107958108
  81. 81. Ryan BC, Vandenbergh JG. Developmental exposure to environmental estrogens alters anxiety and spatial memory in female mice. Hormones and Behavior. 2006;50:85-93. DOI: 10.1016/j.yhbeh.2006.01.007
  82. 82. Wolstenholme JT, Rissman EF, Connelly JJ. The role of bisphenol A in shaping the brain, epigenome and behavior. Hormones and Behavior. 2011;59:296-305. DOI: 10.1016/j.yhbeh.2010.10.001
  83. 83. Wolstenholme JT, Taylor JA, Shetty SR, Edwards M, Connelly JJ, Rissman EF. Gestational exposure to low dose bisphenol A alters social behavior in juvenile mice. PLoS One. 2011;6:e25448. DOI: 10.1371/journal.pone.0025448
  84. 84. Cox KH, Gatewood JD, Howeth C, Rissman EF. Gestational exposure to bisphenol A and cross-fostering affect behaviors in juvenile mice. Hormones and Behavior. 2010;58:754-761. DOI: 10.1016/j.yhbeh.2010.07.008

Written By

Hidetomo Iwano, Hiroki Inoue, Miyu Nishikawa, Jumpei Fujiki and Hiroshi Yokota

Submitted: 17 February 2018 Reviewed: 03 May 2018 Published: 05 November 2018