Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1227 word(s) 1227 2021-07-02 11:50:00 |
2 format correct Meta information modification 1227 2021-07-19 10:58:34 | |
3 def corrected Meta information modification 1227 2021-07-19 10:59:17 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Hsu, W. Bisphenol A in Female Rats. Encyclopedia. Available online: https://encyclopedia.pub/entry/12155 (accessed on 25 April 2024).
Hsu W. Bisphenol A in Female Rats. Encyclopedia. Available at: https://encyclopedia.pub/entry/12155. Accessed April 25, 2024.
Hsu, Wei-Hsuan. "Bisphenol A in Female Rats" Encyclopedia, https://encyclopedia.pub/entry/12155 (accessed April 25, 2024).
Hsu, W. (2021, July 19). Bisphenol A in Female Rats. In Encyclopedia. https://encyclopedia.pub/entry/12155
Hsu, Wei-Hsuan. "Bisphenol A in Female Rats." Encyclopedia. Web. 19 July, 2021.
Bisphenol A in Female Rats
Edit

Bisphenol A (BPA) exposure is the most prevalent in the environment. Rahman et al. (2021) summarized several confounding factors that may be directly or indirectly related to human BPA exposure and detailed the disparities between scientifically derived safe dosages of BPA and those designated as “safe” by government regulatory agencies. Exposure to BPA during early development has been associated with the prevalence of various cardiometabolic diseases including obesity, metabolic syndrome, type 2 diabetes, and cardiovascular diseases.

resveratrol butyrate esters (RBEs) perinatal exposure obesity bisphenol A (BPA) Firmicutes/Bacteroidetes (F/B) ratio

1. Overview

Resveratrol butyrate esters (RBE) are derivatives of resveratrol (RSV) and butyric acid and exhibit biological activity similar to that of RSV but with higher bioavailability. The aim of this study was designed as an animal experiment to explore the effects of RBE on the serum biochemistry, and fat deposits in the offspring rats exposed to bisphenol A (BPA), along with the growth and decline of gut microbiota. We constructed an animal model of perinatal Bisphenol A (BPA) exposure to observe the effects of RBE supplementation on obesity, blood lipids, and intestinal microbiota in female offspring rats. Perinatal exposure to BPA led to weight gain, lipid accumulation, high levels of blood lipids, and deterioration of intestinal microbiota in female offspring rats. RBE supplementation reduced the weight gain and lipid accumulation caused by BPA, optimised the levels of blood lipids, significantly reduced the Firmicutes/Bacteroidetes (F/B) ratio, and increased and decreased the abundance of S24-7 and Lactobacillus, respectively. The analysis of faecal short-chain fatty acid (SCFA) levels revealed that BPA exposure increased the faecal concentration of acetate, which could be reduced via RBE supplementation. However, the faecal concentrations of propionate and butyrate were not only significantly lower than that of acetate, but also did not significantly change in response to BPA exposure or RBE supplementation. Hence, RBE can suppress BPA-induced obesity in female offspring rats, and it demonstrates excellent modulatory activity on intestinal microbiota, with potential applications in perinatological research. 

2. Bisphenol A

Bisphenol A (BPA) exposure is the most prevalent in the environment [1]. Rahman et al. (2021) summarized several confounding factors that may be directly or indirectly related to human BPA exposure and detailed the disparities between scientifically derived safe dosages of BPA and those designated as “safe” by government regulatory agencies [2]. Exposure to BPA during early development has been associated with the prevalence of various cardiometabolic diseases [3] including obesity [4][5], metabolic syndrome [6], type 2 diabetes [5][7], and cardiovascular diseases [5][8]. The toxicokinetic studies of laboratory animals and humans after oral ingestion of BPA have similar results. BPA is rapidly absorbed from the gastrointestinal tract and undergoes first-pass conjugation to BPA-glucuronide (BPA-gluc) and BPA-sulfate (BPA-sulfate), which are biologically inactive metabolites [9][10][11][12]. The studies after oral intake of BPA or high-BPA diet in humans are consistent with animal data, confirming that the internal exposure of unbound BPA is lower after oral exposure, and BPA-gluc is the main metabolite [13][14][15][16][17][18].
Growing evidence suggests that, due to the phenolic structure of BPA, it interacts with estrogen receptors and acts as either agonist or antagonist via estrogen receptor-dependent signaling pathways [19][20][21][22]. Previous studies have demonstrated the developmental origins of the health and disease (DOHaD) hypothesis [23][24], which highlights the links between the periconceptual, foetal, and early infant phases of life and the subsequent development of adult obesity and related metabolic disorders [25][26]. The lipophilicity of BPA facilitates its entry through placental and blood–brain barriers into the foetus, where it triggers oxidative damage and nerve injury in the brain, thereby affecting brain development and causing permanent foetal brain injury [27]. In addition, previous animal studies have demonstrated that maternal and foetal exposure to BPA also damages the immune system and affects the balance of intestinal microbiota in the offspring [28][29]. There is little doubt that poor diet and lack of exercise contribute to the obesity epidemic; recent studies have identified an estrogen endocrine disrupter chemical BPA that may act as an environmental obesogen and either directly or indirectly influence fat accrual [30]. The plausible explanation may involve sex hormones, genomic and non-genomic pathway involving nuclear estrogen receptors, differing developmental pattern and/or epigenetic influence [31][32][33]. Somm et al. [34] reported that exposure to 70 μg/kg/day of BPA during pregnancy can upregulate the expression of PPARγ, C/EBPα, and LPL in abdominal adipocytes of adult female rats. In addition, body weight only increased in females, in which parametrial white adipose tissue (PWAT) weight increased about threefold. Resveratrol (RSV) can prevent BPA-induced metabolic abnormalities in offspring. Darby et al. (2019) showed that RSV can improve the placental blood flow by increasing the bioavailability of NO, can increase the activity of antioxidant enzymes in foetuses and placentas, and can reduce the rates of inflammation and cell death in placentas to ensure adequate foetal nutrition [35]. Therefore, RSV is often employed in the treatment of metabolic abnormalities and oxidative stress attributed to gestational diabetes in offspring.
The pharmacokinetic study reported by Huang et al. (2019) showed that the blood concentration of RSV peaked within 0.5 to 2 h after oral administration, followed by a rapid decline [36], indicating that RSV is unable to induce adequate pharmacological responses in pregnant women; hence, it is not an ideal clinical drug. We reported novel resveratrol butyrate esters (RBEs) and indicated the esterification of RSV with butyrate that contained RSV (~17.1%), RBE monoester (~47.1%), and RBE diester (~35.0%), which also had better hydrogen peroxide scavenging activity than RSV [37] and could effectively inhibit fatty-acid-induced lipid accumulation in HepG2 cells, with effects similar to those of RSV, but achieved at a lower dose level [38]. Butyrate, one of the short-chain fatty acids (SCFAs), can selectively promote the growth of beneficial bacteria that improve the intestinal barrier’s function [39]. As RBE can be catabolised into RSV and butyrate in the body [40], we also attempted to further increase the concentration of butyrate in the intestinal tract to protect the colonic epithelial cells and nervous tissue in the brain.
Numerous studies have confirmed that intestinal microbiota and obesity are closely associated with metabolic abnormalities in hosts [41]. For example, the Firmicutes/Bacteroidetes (F/B) ratio, which is related to obesity, is lower in obese individuals than in normal individuals [42][43][44][45].
To date, there are still no obesity-related perinatological studies of RBE. Although the sex-specific effects of BPA are well documented, including the differential susceptibility of males and females to different doses of BPA [31][32][46][47][48][49], the underlying mechanism remains unclear [47]. Therefore, we performed perinatal BPA exposure studies to observe the effects of RBE supplementation on obesity-related indicators and intestinal microbiota in female rat offspring to better understand the potential applications of RBE.

3. Conclusions

Rats were exposed to BPA during the perinatal period to observe the effects of RBE supplementation on obesity-related indicators and intestinal microbiota in their female offspring to better understand the potential application of RBE. The results demonstrated that RBE supplementation can alleviate BPA-induced weight gain and body fat accumulation, optimise the concentration of blood-lipid-related markers, significantly reduce the Firmicutes/Bacteroidetes (F/B) ratio and the abundance of Lactobacillus, and increase the abundance of S24-7. In addition, RBE supplementation can also regulate the intestinal concentration of acetate in female offspring rats. In summary, RBE can suppress BPA-induced obesity in female offspring rats and exhibits excellent modulatory activity in intestinal microbiota, with potential applications in perinatological research. In the future, we will clarify the metabolism of RBE in the digestive systems of animals and its haemodynamics after absorption to further understand the potential of RBE in future applications.

References

  1. Pelch, K.; Wignall, J.A.; Goldstone, A.E.; Ross, P.K.; Blain, R.B.; Shapiro, A.J.; Holmgren, S.D.; Hsieh, J.H.; Svoboda, D.; Auerbach, S.S.; et al. A scoping review of the health and toxicological activity of bisphenol A (BPA) structural analogues and functional alternatives. Toxicology 2019, 424, 152235.
  2. Rahman, M.S.; Adegoke, E.O.; Pang, M.-G. Drivers of owning more BPA. J. Hazard. Mater. 2021, 417, 126076.
  3. Moon, S.; Yu, S.H.; Lee, C.B.; Park, Y.J.; Yoo, H.J.; Kim, D.S. Effects of bisphenol A on cardiovascular disease: An epidemiological study using National Health and Nutrition Examination Survey 2003–2016 and meta-analysis. Sci. Total Environ. 2021, 763.
  4. Engin, A.B.; Engin, A. The effect of environmental Bisphenol A exposure on breast cancer associated with obesity. Environ. Toxicol. Pharmacol. 2021, 81.
  5. Lee, I.; Park, Y.J.; Kim, M.J.; Kim, S.; Choi, S.; Park, J.; Cho, Y.H.; Hong, S.; Yoo, J.; Park, H.; et al. Associations of urinary concentrations of phthalate metabolites, bisphenol A, and parabens with obesity and diabetes mellitus in a Korean adult population: Korean National Environmental Health Survey (KoNEHS) 2015–2017. Environ. Int. 2021, 146, 106227.
  6. Angle, B.M.; Do, R.P.; Ponzi, D.; Stahlhut, R.W.; Drury, B.E.; Nagel, S.C.; Welshons, W.V.; Besch-Williford, C.L.; Palanza, P.; Parmigiani, S.; et al. Metabolic disruption in male mice due to fetal exposure to low but not high doses of bisphenol A (BPA): Evidence for effects on body weight, food intake, adipocytes, leptin, adiponectin, insulin and glucose regulation. Reprod. Toxicol. 2013, 42, 256–268.
  7. Rancière, F.; Botton, J.; Slama, R.; Lacroix, M.Z.; Debrauwer, L.; Charles, M.A.; Roussel, R.; Balkau, B.; Magliano, D.J.; Balkau, B.; et al. Exposure to bisphenol a and bisphenol s and incident type 2 diabetes: A case-cohort study in the French cohort D.E.S.I.R. Environ. Health Perspect. 2019, 127.
  8. Shu, L.; Meng, Q.; Diamante, G.; Tsai, B.; Chen, Y.-W.; Mikhail, A.; Luk, H.; Ritz, B.; Allard, P.; Yang, X. Prenatal bisphenol a exposure in mice induces multitissue multiomics disruptions linking to cardiometabolic disorders. Endocrinology 2019, 160, 409–429.
  9. Churchwell, M.I.; Camacho, L.; Vanlandingham, M.M.; Twaddle, N.C.; Sepehr, E.; Delclos, K.B.; Fisher, J.W.; Doerge, D.R. Comparison of life-stage-dependent internal dosimetry for bisphenol A, ethinyl estradiol, a reference estrogen, and endogenous estradiol to test an estrogenic mode of action in Sprague Dawley rats. Toxicol. Sci. 2014, 139, 4–20.
  10. Doerge, D.R.; Twaddle, N.C.; Vanlandingham, M.; Fisher, J.W. Pharmacokinetics of bisphenol A in neonatal and adult Sprague-Dawley rats. Toxicol. Appl. Pharmacol. 2010, 247, 158–165.
  11. Doerge, D.R.; Twaddle, N.C.; Vanlandingham, M.; Fisher, J.W. Pharmacokinetics of Bisphenol A in neonatal and adult CD-1 mice: Inter-species comparisons with Sprague-Dawley rats and rhesus monkeys. Toxicol. Lett. 2011, 207, 298–305.
  12. Doerge, D.R.; Twaddle, N.C.; Vanlandingham, M.; Fisher, J.W. Pharmacokinetics of bisphenol A in serum and adipose tissue following intravenous administration to adult female CD-1 mice. Toxicol. Lett. 2012, 211, 114–119.
  13. Thayer, K.A.; Doerge, D.R.; Hunt, D.; Schurman, S.H.; Twaddle, N.C.; Churchwell, M.I.; Garantziotis, S.; Kissling, G.E.; Easterling, M.R.; Bucher, J.R.; et al. Pharmacokinetics of bisphenol A in humans following a single oral administration. Environ. Int. 2015, 83, 107–115.
  14. Völkel, W.; Colnot, T.; Csanády, G.A.; Filser, J.G.; Dekant, W. Metabolism and kinetics of bisphenol a in humans at low doses following oral administration. Chem. Res. Toxicol. 2002, 15, 1281–1287.
  15. Völkel, W.; Kiranoglu, M.; Fromme, H. Determination of free and total bisphenol A in human urine to assess daily uptake as a basis for a valid risk assessment. Toxicol. Lett. 2008, 179, 155–162.
  16. Teeguarden, J.G.; Calafat, A.M.; Ye, X.; Doerge, D.R.; Churchwell, M.I.; Gunawan, R.; Graham, M.K. Twenty-four hour human urine and serum profiles of bisphenol A during high-dietary exposure. Toxicol. Sci. 2011, 123, 48–57.
  17. Teeguarden, J.G.; Twaddle, N.C.; Churchwell, M.I.; Yang, X.; Fisher, J.W.; Seryak, L.M.; Doerge, D.R. 24-h human urine and serum profiles of bisphenol A: Evidence against sublingual absorption following ingestion in soup. Toxicol. Appl. Pharmacol. 2015, 288, 131–142.
  18. Report on the two-phase public consultation on the draft EFSA scientific opinion on bisphenol A (BPA). EFSA Support. Publ. 2017, 12, 740E.
  19. Adegoke, E.O.; Rahman, M.S.; Pang, M.G. Bisphenols Threaten Male Reproductive Health via Testicular Cells. Front. Endocrinol. 2020, 11, 624.
  20. Kitamura, S.; Suzuki, T.; Sanoh, S.; Kohta, R.; Jinno, N.; Sugihara, K.; Yoshihara, S.; Fujimoto, N.; Watanabe, H.; Ohta, S. Comparative study of the endocrine-disrupting activity of bisphenol A and 19 related compounds. Toxicol. Sci. 2005, 84, 249–259.
  21. Rahman, M.S.; Kwon, W.S.; Yoon, S.J.; Park, Y.J.; Ryu, B.Y.; Pang, M.G. A novel approach to assessing bisphenol-A hazards using an in vitro model system. BMC Genom. 2016, 17, 1–12.
  22. Beausoleil, C.; Emond, C.; Cravedi, J.P.; Antignac, J.P.; Applanat, M.; Appenzeller, B.M.R.; Beaudouin, R.; Belzunces, L.P.; Canivenc-Lavier, M.C.; Chevalier, N.; et al. Regulatory identification of BPA as an endocrine disruptor: Context and methodology. Mol. Cell. Endocrinol. 2018, 475, 4–9.
  23. Tain, Y.-L.; Chan, J.Y.H.; Lee, C.-T.; Hsu, C.-N. Maternal melatonin therapy attenuates methyl-donor diet-induced programmed hypertension in male adult rat offspring. Nutrients 2018, 10, 1407.
  24. Tain, Y.-L.; Lin, Y.-J.; Chan, J.Y.H.; Lee, C.-T.; Hsu, C.-N. Maternal melatonin or agomelatine therapy prevents programmed hypertension in male offspring of mother exposed to continuous light. Biol. Reprod. 2017, 97, 636–643.
  25. Sergeyev, O.V.; Nikitin, A.I. Developmental origins of health and disease (DOHaD) and paternal origins of health and disease (POHaD). Multigenerational inheritance. Obstet. Gynecol. Reprod. 2019, 13, 326–336.
  26. Tain, Y.-L.; Lin, Y.-J.; Sheen, J.-M.; Yu, H.-R.; Tiao, M.-M.; Chen, C.-C.; Tsai, C.-C.; Huang, L.-T.; Hsu, C.-N. High Fat Diets Sex-Specifically Affect the Renal Transcriptome and Program Obesity, Kidney Injury, and Hypertension in the Offspring. Nutrients 2017, 9, 357.
  27. Mhaouty-Kodja, S.; Belzunces, L.P.; Canivenc, M.-C.; Schroeder, H.; Chevrier, C.; Pasquier, E. Impairment of learning and memory performances induced by BPA: Evidences from the literature of a MoA mediated through an ED. Mol. Cell. Endocrinol. 2018, 475, 54–73.
  28. Malaisé, Y.; Menard, S.; Cartier, C.; Gaultier, E.; Lasserre, F.; Lencina, C.; Harkat, C.; Geoffre, N.; Lakhal, L.; Castan, I.; et al. Gut dysbiosis and impairment of immune system homeostasis in perinatally-exposed mice to Bisphenol A precede obese phenotype development. Sci. Rep. 2017, 7.
  29. Reddivari, L.; Veeramachaneni, D.N.R.; Walters, W.A.; Lozupone, C.; Palmer, J.; Hewage, M.K.; Bhatnagar, R.; Amir, A.; Kennett, M.J.; Knight, R.; et al. Perinatal bisphenol a exposure induces chronic inflammation in rabbit offspring via modulation of gut bacteria and their metabolites. mSystems 2017, 2.
  30. Janesick, A.; Blumberg, B. Obesogens, stem cells and the developmental programming of obesity. Int. J. Androl. 2012, 35, 437–448.
  31. Vandenberg, L.N.; Colborn, T.; Hayes, T.B.; Heindel, J.J.; Jacobs, D.R.; Lee, D.H.; Shioda, T.; Soto, A.M.; vomSaal, F.S.; Welshons, W.V.; et al. Hormones and endocrine-disrupting chemicals: Low-dose effects and nonmonotonic dose responses. Endocr. Rev. 2012, 33, 378–455.
  32. Kundakovic, M.; Gudsnuk, K.; Franks, B.; Madrid, J.; Miller, R.L.; Perera, F.P.; Champagne, F.A. Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol a exposure. Proc. Natl. Acad. Sci. USA 2013, 110, 9956–9961.
  33. Susiarjo, M.; Sasson, I.; Mesaros, C.; Bartolomei, M.S. Bisphenol A Exposure Disrupts Genomic Imprinting in the Mouse. PLoS Genet. 2013, 9, e1003401.
  34. Somm, E.; Schwitzgebel, V.M.; Toulotte, A.; Cederroth, C.R.; Combescure, C.; Nef, S.; Aubert, M.L.; Hüppi, P.S. Perinatal exposure to bisphenol A alters early adipogenesis in the rat. Environ. Health Perspect. 2009.
  35. Darby, J.R.T.; Mohd Dollah, M.H.B.; Regnault, T.R.H.; Williams, M.T.; Morrison, J.L. Systematic review: Impact of resveratrol exposure during pregnancy on maternal and fetal outcomes in animal models of human pregnancy complications—Are we ready for the clinic? Pharmacol. Res. 2019, 144, 264–278.
  36. Huang, X.; Li, X.; Xie, M.; Huang, Z.; Huang, Y.; Wu, G.; Peng, Z.; Sun, Y.; Ming, Q.; Liu, Y.; et al. Resveratrol: Review on its discovery, anti-leukemia effects and pharmacokinetics. Chem. Biol. Interact. 2019, 306, 29–38.
  37. Tain, Y.-L.; Chang, S.K.C.; Liao, J.-X.; Chen, Y.-W.; Huang, H.-T.; Li, Y.-L.; Hou, C.-Y. Synthesis of Short-Chain-Fatty-Acid Resveratrol Esters and Their Antioxidant Properties. Antioxidants 2021, 10, 420.
  38. Tain, Y.-L.; Jheng, L.-C.; Chang, S.K.C.; Chen, Y.-W.; Huang, L.-T.; Liao, J.-X.; Hou, C.-Y. Synthesis and Characterization of Novel Resveratrol Butyrate Esters That Have the Ability to Prevent Fat Accumulation in a Liver Cell Culture Model. Molecules 2020, 25, 4199.
  39. Wang, M.; Wichienchot, S.; He, X.; Fu, X.; Huang, Q.; Zhang, B. In vitro colonic fermentation of dietary fibers: Fermentation rate, short-chain fatty acid production and changes in microbiota. Trends Food Sci. Technol. 2019, 88, 1–9.
  40. Hu, X.-P.; Yin, F.-W.; Zhou, D.-Y.; Xie, H.-K.; Zhu, B.-W.; Ma, X.-C.; Tian, X.-G.; Wang, C.; Shahidi, F. Stability of resveratrol esters with caprylic acid during simulated in vitro gastrointestinal digestion. Food Chem. 2019, 276, 675–679.
  41. Wilkins, A.T.; Reimer, R.A. Obesity, early life gut microbiota, and antibiotics. Microorganisms 2021, 9, 413.
  42. Li, X.; Yang, L.; Xu, M.; Qiao, G.; Li, C.; Lin, L.; Zheng, G. Smilax china L. polyphenols alleviates obesity and inflammation by modulating gut microbiota in high fat/high sucrose diet-fed C57BL/6J mice. J. Funct. Foods 2021, 77, 104332.
  43. Koliada, A.; Syzenko, G.; Moseiko, V.; Budovska, L.; Puchkov, K.; Perederiy, V.; Gavalko, Y.; Dorofeyev, A.; Romanenko, M.; Tkach, S.; et al. Association between body mass index and Firmicutes/Bacteroidetes ratio in an adult Ukrainian population. BMC Microbiol. 2017, 17, 120.
  44. Shen, Z.H.; Zhu, C.X.; Quan, Y.S.; Yang, Z.Y.; Wu, S.; Luo, W.W.; Tan, B.; Wang, X.Y. Relationship between intestinal microbiota and ulcerative colitis: Mechanisms and clinical application of probiotics and fecal microbiota transplantation. World J. Gastroenterol. 2018, 24, 5–14.
  45. Abenavoli, L.; Scarpellini, E.; Colica, C.; Boccuto, L.; Salehi, B.; Sharifi-Rad, J.; Aiello, V.; Romano, B.; De Lorenzo, A.; Lzzo, A.A. Gut Microbiota and Obesity: A Role for Probiotics. Nutrients 2019, 11, 2690.
  46. Strakovsky, R.S.; Wang, H.; Engeseth, N.J.; Flaws, J.A.; Helferich, W.G.; Pan, Y.X.; Lezmi, S. Developmental bisphenol A (BPA) exposure leads to sex-specific modification of hepatic gene expression and epigenome at birth that may exacerbate high-fat diet-induced hepatic steatosis. Toxicol. Appl. Pharmacol. 2015, 284, 101–112.
  47. Lejonklou, M.H.; Dunder, L.; Bladin, E.; Pettersson, V.; Rönn, M.; Lind, L.; Waldén, T.B.; Lind, P.M. Effects of low-dose developmental bisphenol a exposure on metabolic parameters and gene expression in male and female fischer 344 rat offspring. Environ. Health Perspect. 2017, 125.
  48. Yang, T.C.; Peterson, K.E.; Meeker, J.D.; Sánchez, B.N.; Zhang, Z.; Cantoral, A.; Solano, M.; Tellez-Rojo, M.M. Bisphenol A and phthalates in utero and in childhood: Association with child BMI z-score and adiposity. Environ. Res. 2017, 156, 326–333.
  49. Rubin, B.S.; Paranjpe, M.; DaFonte, T.; Schaeberle, C.; Soto, A.M.; Obin, M.; Greenberg, A.S. Perinatal BPA exposure alters body weight and composition in a dose specific and sex specific manner: The addition of peripubertal exposure exacerbates adverse effects in female mice. Reprod. Toxicol. 2017, 68, 130–144.
More
Information
Subjects: Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 342
Revisions: 3 times (View History)
Update Date: 19 Jul 2021
1000/1000