Open Access

BPA disrupts 17‑estradiol‑mediated hepatic protection against ischemia/reperfusion injury in rat liver by upregulating the Ang II/AT1R signaling pathway

  • Authors:
    • Yili Zhang
    • Yu Shi
    • Zeyu Li
    • Liankang Sun
    • Mei Zhang
    • Liang Yu
    • Shengli Wu
  • View Affiliations

  • Published online on: April 16, 2020     https://doi.org/10.3892/mmr.2020.11072
  • Pages: 416-422
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bisphenol A (BPA), a xenoestrogen commonly used in plastics, may act as an endocrine disruptor, which indicates that BPA might be a public health risk. The present study aimed to investigate the effect of BPA on 17β‑estradiol (E2)‑mediated protection against liver ischemia/reperfusion (I/R) injury, and to identify the underlying mechanisms using a rat model. A total of 56 male Sprague Dawley rats were randomly divided into the following seven groups: i) Sham; ii) I/R; iii) Sham + BPA; iv) I/R + BPA; v) I/R + E2; vi) I/R + E2 + BPA; and vii) I/R + E2 + BPA + losartan [LOS; an angiotensin II (Ang II) type I receptor (ATIR) antagonist]. A rat model of hepatic I/R injury was established by inducing hepatic ischemia for 60 min followed by reperfusion for 24 h. When ischemia was induced, rats were treated with vehicle, E2, BPA or LOS. After 24 h of reperfusion, blood samples and hepatic tissues were collected for histopathological and biochemical examinations. The results suggested that 4 mg/kg BPA did not significantly alter the liver function, or Ang II and AT1R expression levels in the Sham and I/R groups. However, 4 mg/kg BPA inhibited E2‑mediated hepatic protection by enhancing hepatic necrosis, and increasing the release of alanine transaminase, alkaline phosphatase and total bilirubin (P<0.05). Moreover, BPA increased serum and hepatic Ang II levels, as well as AT1R protein expression levels in the E2‑treated rat model of liver I/R injury (P<0.05). LOS treatment reversed the negative effects of BPA on hepatic necrosis and liver serum marker levels, although it did not reverse BPA‑mediated upregulation of serum and hepatic Ang II levels, or hepatic AT1R expression. Therefore, the present study suggested that BPA disrupted E2‑mediated hepatic protection following I/R injury, but did not significantly affect healthy or I/R‑injured livers; therefore, the mechanism underlying the effects of BPA may be associated with upregulation of the Ang II/AT1R signaling pathway.

Introduction

Bisphenol A (BPA) is a xenoestrogen (XE) that is extensively produced for the manufacture of polycarbonate plastic (1). Ubiquitous exposure to BPA is associated with a series of health problems, including obesity, diabetes and disorders of the reproductive system (24). Previous studies have reported that BPA may also affect liver function, and high levels of urinary BPA are associated with non-alcoholic fatty liver disease in adults in the United States (58). Another study reported that serum aspartate transaminase and alanine transaminase (ALT) levels were not altered following exposure to low level BPA (5 mg/kg), but were significantly increased following exposure to high level BPA (50 mg/kg) in rats (9).

Liver ischemia/reperfusion (I/R) injury occurs during a number of clinical events, including liver transplantation, partial hepatectomy and hemorrhagic shock (10). The pathophysiological processes of liver I/R injury are multifactorial and characterized by various clinical manifestations ranging in severity from asymptomatic elevation of serum transaminases to hepatic failure (11). Despite improvements in perioperative management and surgical techniques, liver I/R injury continues to be an important clinical issue. Previous studies have demonstrated that 17-estradiol (E2) protects the liver against I/R injury by reducing microvascular dysfunction and inflammatory responses (12), increasing vasodilator nitric oxide (NO) production (13) and preventing impaired Kupffer cell activity (14). Furthermore, a previous study reported that E2 provides hepatoprotection against I/R injury by downregulating the angiotensin II (Ang II)/Ang II type I receptor (AT1R) signaling pathway (15). However, the role of BPA during liver I/R injury has not been previously reported.

Previous studies have demonstrated that BPA can attenuate the physiological function of estrogen and inhibit the production of testosterone (16,17). Moreover, BPA can reduce the activity of multiple hepatoprotective enzymes, including catalase, total glutathione S-transferase, total glutathione peroxidase and total superoxide dismutase (18). Therefore, the present study aimed to investigate whether BPA disrupted E2-mediated hepatic protection against I/R injury, and to identify the possible underlying mechanisms using a rat model.

Materials and methods

Animals

Male Sprague-Dawley rats (weight, 190–210 g; age, 9–10 weeks old) were purchased from the Animal Center of Xi'an Jiaotong University. Animals were kept in standard housing conditions at 22±2°C with 12-h light/dark cycles, 45–60% humidity, and free access to food and water. Animal experiments were conducted according to the National Institutes of Health Guidelines on the Use of Laboratory Animals (12). The present study was approved by the Xi'an Jiaotong University Health Science Center Ethics Committee.

Model of total hepatic I/R

Previous studies have demonstrated that short periods (60 min) of global liver ischemia result in reversible cell injury, in which liver oxygen consumption returns to control levels when the oxygen supply is reestablished following ischemia (19). However, reperfusion following prolonged warm ischemia (>120 min) results in irreversible cell damage (19). Therefore, the present study established a 60 min global liver ischemia model, which simulated the clinical situation of warm ischemia after the Pringle maneuver (20) in hepatic surgery. Rats were fasted for 16–18 h prior to the operation. Rats were anesthetized using 64% N2O, 32% O2 and 4% isoflurane and anesthesia was maintained using 65% N2O, 32% O2 and 3% isoflurane. Rats were ventilated endotracheally during the operation. A median incision was made in the upper abdomen and the hepatic pedicle was clamped using a non-invasive microvascular clip to induce total liver I/R. After 60 min, the clamp was removed to allow reperfusion. Sham controls received the laparotomy procedure without hepatic pedicle clamping. After closing the abdominal cavity, rats were allowed to recover with free access to food and water. At 24 h post-reperfusion, rats were euthanized by overdose with intravenous sodium pentobarbital (100 mg/kg) followed by exsanguination. Subsequently, the liver tissues and 5 ml venous blood drawn from the inferior vena cava were harvested for subsequent experiments.

Experimental design

A total of 56 rats were randomly divided into the following seven experimental groups (n=8/group): i) Sham; ii) I/R; iii) Sham + BPA; iv) I/R + BPA; v) I/R + E2; vi) I/R + E2 + BPA; and vii) I/R + E2 + BPA + Losartan (LOS). BPA (Tokyo Chemical Industry Co., Ltd.) and E2 (Sigma-Aldrich; Merck KGaA) were dissolved in DMSO, and diluted in saline. The final concentration of DMSO was <0.1%. BPA (4 mg/kg) or E2 (4 mg/kg) were administered intravenously when the laparotomy procedure began. LOS (30 mg/kg; Wuhan Boster Biological Technology, Ltd.) was administered intraperitoneally when the laparotomy procedure began. The Sham and I/R groups received only dilution vehicle (saline).

Histological assessment of I/R injury

Formalin-fixed (10%, 24 h, room temperature) paraffin-embedded liver tissues were cut into 5 µm-thick sections, and stained with hematoxylin (15 min) and eosin (5 min) at room temperature to estimate the severity of I/R injury. Sections were examined for the following signs: Nuclear pyknosis, loss of hepatocellular borders or areas of necrosis. Stained sections were observed in at least 10 randomly selected fields with a light microscope (magnification, ×400) and examined for the following signs: Nuclear pyknosis, loss of hepatocellular borders or areas of necrosis. Sections were scored using according to a previously described formula: Necrotic area/total area of the field (21), and analyzed using Image-Pro Plus software (version 6.0; Media Cybernetics, Inc.).

Measurement of serum hepatic damage markers

At 24 h post-reperfusion, harvested blood samples were centrifuged at 3,000 × g for 5 min at 4°C for serum separation and serum concentrations of ALT, alkaline phosphate (ALP) and total bilirubin (TBIL) were detected using the AU400 automated chemistry analyzer according to the manufacturer's protocol (Olympus Corporation).

Measurement of Ang II

Rat liver tissue was homogenized and total protein was extracted using T-PER™ tissue protein extraction buffer (Pierce; Thermo Fisher Scientific, Inc.) containing protease inhibitors. Protein concentration was determined using the Coomassie brilliant blue method. Serum and hepatic Ang II levels were measured using an Ang II ELISA kit (cat. no. ABIN416068; Uscn Life Science Inc.), according to the manufacturer's protocol.

Western blotting

The hepatic tissue expression levels of AT1R were detected using the western blotting. Total protein was extracted using T-PER™ tissue protein extraction buffer (Pierce; Thermo Fisher Scientific, Inc.) containing protease inhibitors. Protein concentration was determined using the Coomassie brilliant blue method. The mass of the proteins loaded in per lane was 50 µg. Protein was separated by 10% SDS-PAGE and transferred onto nitrocellulose membranes (GE Healthcare Life Sciences). The membranes were blocked with 5% bovine serum albumin (cat. no. CSB-NP009501B; CUSA Bio.) at 37°C for 1 h, and washed three times with ice-cold PBS. Subsequently, the membranes were incubated at 37°C for 2 h with the following primary antibodies: Anti-AT1R (cat. no. sc-31181; 1:1,000; Santa Cruz Biotechnology, Inc.) and anti-β-actin (cat. no. sc-130656; 1,000; Santa Cruz Biotechnology, Inc.). Following primary incubation, the membranes were incubated with a horseradish peroxidase-conjugated donkey anti-rabbit IgG secondary antibody (cat. no. NA934; 1:1,000; GE Healthcare Life Sciences) for 1 h at 37°C. Protein bands were visualized using an enhanced chemiluminescence detection system (GE Healthcare Life Sciences) and the UVP BioSpectrum500 imaging system (UVP, LLC). Protein expression was quantified using Quantity One software (version 4.6.9; Bio-Rad Laboratories, Inc.) with β-actin as the loading control.

Statistical analysis

Data are presented as the mean ± SD. Statistical analyses were performed using SPSS software (version 24.0; IBM Corp.). Comparisons among groups were analyzed using one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Histological changes

At 24 h post-reperfusion, liver tissues were harvested. No significant morphological changes were observed between the Sham and Sham + BPA groups. However, severe necrosis, extensive nuclear pyknosis and loss of intercellular borders were detected in the I/R and I/R + BPA groups. Compared with the I/R and I/R + BPA groups, the I/R + E2 group displayed a significantly reduced percentage of hepatic necrosis (52±6 vs. 22±3.2% and 50±5.5 vs. 22±3.2%, respectively; P<0.05; Fig. 1); however, BPA treatment decreased E2-mediated hepatoprotective effects (45±5.6%; P<0.05 vs. I/R + E2 group). Furthermore, it was demonstrated that LOS treatment significantly reversed the negative effect of BPA on E2-mediated hepatoprotection (25±3%; P<0.05 vs. I/R + E2+BPA group; Fig. 1).

Liver function

Serum concentrations of ALT, ALP and TBIL in the Sham + BPA group were not significantly different at 24 h post-reperfusion compared with the Sham group (all P>0.05). However, the I/R and I/R + BPA groups displayed significantly higher serum concentrations levels of ALT, ALP and TBIL compared with the Sham and Sham + BPA groups (P<0.05). Moreover, compared with the I/R and I/R + BPA groups, the I/R + E2 group displayed significantly reduced levels of the serum markers (all P<0.05); however, BPA treatment significantly inhibited the hepatoprotective activity of E2 against I/R injury (all P<0.05 vs. I/R + E2 group). In addition, the results suggested that LOS treatment reversed the negative effect of BPA on E2 in the rat model of liver I/R injury (Fig. 2).

Serum and hepatic Ang II levels

At 24 h post-reperfusion, there were no significant differences in serum and hepatic Ang II levels between the Sham and Sham + BPA groups (all P>0.05). By contrast, the levels of serum and hepatic Ang II in the I/R and I/R + BPA groups were significantly higher compared with the Sham and Sham + BPA groups (P<0.05). Furthermore, E2 treatment significantly reduced the levels of serum and hepatic Ang II compared with the I/R and I/R + BPA groups (all P<0.05), and BPA treatment inhibited the hepatoprotective activity of E2 against liver I/R injury (all P<0.05 vs. I/R + E2 group). However, LOS treatment did not significantly alter serum and hepatic Ang II levels in the I/R + E2 + BPA group (all P>0.05 vs. I/R+E2+BPA+LOS group) (Fig. 3).

Expression of hepatic AT1R

The hepatic tissue expression level of AT1R was determined by western blotting. At 24 h post-reperfusion, there were no significant differences in the expression of AT1R protein between the Sham and Sham + BPA groups (P>0.05). However, hepatic AT1R expression levels in the I/R and I/R + BPA groups were significantly higher compared with the Sham and Sham + BPA groups (P<0.05). The results indicated that E2 treatment significantly decreased hepatic AT1R expression levels compared with the I/R and I/R + BPA groups (all P<0.05); however, BPA treatment also inhibited E2-mediated effects on AT1R expression (P<0.05 vs. I/R+E2 group). Additionally, LOS treatment did not alter hepatic AT1R expression in I/R + E2 + BPA rats (P>0.05 vs. I/R+E2+BPA group) (Fig. 4).

Discussion

BPA is an endocrine disrupting chemical (EDC) widely used in various industries, including dentistry (1). BPA has attracted attention due to its threat to human health, as it can alter the expression of neural genes including oxytocin and vasopressin, leading to abnormal social behaviors (22).

Ang II receptors are present in two forms: AT1R and AT2R (23). The physiological function of Ang II and AT1R, as well as their role in the pathogenesis of certain diseases are not completely understood. However, compared with AT1R, it is difficult to investigate the functions of AT2R, at least in part due to the relatively low expression levels in cells (23). Previous studies investigating Ang II and its receptors have primarily focused on the Ang II/AT1R axis (2427); therefore, the present study investigated the effects of BPA on hepatic damage and the Ang II/AT1R signaling pathway in a rat model of liver I/R injury. A previous study reported that 0.4 mg/kg/day BPA in rats is close to the current reference daily limit for human exposure by the U.S. Environmental Protection Agency (28). In another study assessing the effects of BPA on the cognitive function of rats, it was indicated that 0.4 mg/kg/day BPA caused a significant decline in spatial memory; however, anxiety-like behavior was only observed in the high-dose BPA group (4 mg/kg/day) (28). A previous study investigating the effects of perinatal maternal exposure to BPA on the behavior of rat offspring, it was reported that male offspring in the 4 mg/kg group displayed significantly lower responses compared with control rats (29); therefore, 4 mg/kg BPA was used in the present study.

It has been reported that 25 mg/kg/day BPA leads to increased serum levels of liver enzymes and defects in the morphology of the liver in rats (30). Moreover, Kazemi et al (31) indicated that 5 µg/kg BPA induced reactive oxygen species production and increased antioxidant gene expression in rats; however, the morphological and functional responses of the liver were not investigated. In the present study, 4 mg/kg BPA did not affect the liver microstructure and enzymes in the Sham or I/R groups. Furthermore, it was demonstrated that there were no significant differences in serum Ang II levels, hepatic Ang II levels and AT1R protein levels between rats treated with or without BPA alone. Therefore, the results suggested that 4 mg/kg BPA may not affect liver function or the Ang II/AT1R signaling pathway in healthy or I/R-injured livers.

The hepatoprotective effect of E2 against I/R injury has been previously reported in rodent models (3234). The possible mechanisms underlying the actions of E2 include: Apoptosis inhibition (21); increasing serum NO levels and decreasing serum tumor necrosis factor-α levels (35); regulating the expression of heat shock protein (36); modulating the activities of mitogen-activated protein kinase (37); and downregulating the Ang II/AT1R signaling pathway (16). Moreover, previous clinical studies have demonstrated that female livers are more tolerant to I/R injury compared with male livers, which may be explained by E2 (38,39). BPA, a well-characterized XE, interacts with estrogen receptors to act as an agonist or antagonist via estrogen receptor-dependent signaling pathways; therefore, BPA plays a role in the pathogenesis of several endocrine disorders, including female and male infertility, precocious puberty and hormone dependent tumors (40). It has been hypothesized that BPA may have a negative effect on the protective effect of E2 against hepatic I/R injury. The present study examined whether BPA disrupted E2-mediated hepatic protection against I/R injury, and the possible underlying mechanisms. The results suggested that E2 protected the liver against I/R injury by attenuating hepatic necrosis, and lowering serum levels of ALT, ALP and TBIL. Furthermore, BPA, as an EDC, abolished certain hepatoprotective activities of E2 by aggravating hepatic necrosis, and increasing the release of ALT, ALP and TBIL, as demonstrated by biochemical and histological analyses.

Ang II is the major effector or peptide of the renin-angiotensin system (41). Previous studies have revealed that Ang II can induce a series of proinflammatory responses by increasing adhesion molecule expression (42), leukocyte-endothelial interaction (43), activator protein 1 and NF-κB activation (44), reactive oxygen species production (45) and proinflammatory cytokine accumulation (46). Moreover, the role of Ang II and its primary receptor, AT1R, in the process of liver I/R injury has been previously reported. Alfany-Fernandez et al (47) reported that Ang II receptor antagonists protect non-steatotic liver grafts against I/R damage. Furthermore, Sabry et al (48) revealed that the hepatoprotective effect of Apelin-13 against I/R injury was related to suppression of the Ang II/AT1R signaling pathway. The results of the aforementioned studies were consistent with the results of the present study. In addition, our previous study indicated that E2 hepatoprotection against I/R injury occurs via downregulation of the Ang II/AT1R signaling pathway (15). However, whether BPA disrupts the hepatoprotective activity of E2 against I/R injury by modulating the Ang II/AT1R signaling pathway is not completely understood. A previous studies has demonstrated that oral administration of BPA induces high blood pressure in mice by upregulating Ang II (49). Another in vitro study reported that following BPA treatment, Ang II expression is upregulated in vascular smooth muscle cells, and the Ang II receptor antagonist LOS attenuates BPA-induced cell proliferation (50). The results of the present study suggested that although BPA did not significantly affect the Ang II/AT1R signaling pathway in the rat model of hepatic I/R injury without E2 treatment, BPA treatment significantly increased the levels of serum and hepatic Ang II, as well as the expression of AT1R protein in the liver in the ER-treated rat model of hepatic I/R injury.

Previous studies have indicated that chronic use of LOS can decrease the expression of AT1R. Abbasloo et al (51) reported that treatment with LOS (10 mg/kg; intraperitoneally) for 5 days reduces the expression of AT1R in cardiac tissue in a rat model of I/R injury. Furthermore, Panico et al (52) demonstrated that renal I/R-induced cardiac levels of AT1R were decreased following LOS treatment (10 mg/kg in drinking water) for 7 days. However, the results of the present study suggested that LOS treatment did not significantly alter serum and liver Ang II levels, or liver AT1R protein expression following hepatic I/R injury; however, LOS reversed the negative effect of BPA on certain E2-mediated hepatoprotective activities. A potential explanation for the discrepancies between the results of the present study and the results of previous studies could be the different doses and courses of treatments used. In the present study, LOS was administered once (30 mg/kg; intraperitoneally) at 24 h before tissue harvesting, which may alter AT1R function, but not AT1R expression. Therefore, further studies are required to identify the mechanisms underlying LOS-mediated regulation of the Ang II/AT1R axis.

In conclusion, to the best of our knowledge, the present study was the first to suggest that upregulation of the Ang II/AT1R signaling pathway may play an important role in BPA-mediated disruption of the hepatoprotective activities of E2 against I/R injury. Moreover, the Ang II/AT1R axis may serve as a promising target for the development of hepatoprotective strategies to prevent BPA-induced liver damage.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the Science and Technology Co-ordination and Innovation Project of Shaanxi (grant. no. 2011KTCL03-21) and the National Natural Science Foundation of China (grant. no. 81373157).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LY, YZ and YS analyzed and interpreted the experimental animal data. ZL and LS performed the histological examination of the liver. MZ performed the PCR and western blotting. LY was a major contributor in writing the manuscript. SW designed the experiments and reviewed the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Xi'an Jiaotong University Health Science Center Ethics Committee.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ehrlich S, Calafat AM, Humblet O, Smith T and Hauser R: Handling of thermal receipts as a source of exposure to bisphenol A. JAMA. 311:859–860. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Fernández M, Bianchi M, Lux-Lantos V and Libertun C: Neonatal exposure to bisphenol a alters reproductive parameters and gonadotropin releasing hormone signaling in female rats. Environ Health Perspect. 117:757–762. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Li DK, Zhou Z, Miao M, He Y, Wang J, Ferber J, Herrinton LJ, Gao E and Yuan W: Urine bisphenol-A (BPA) level in relation to semen quality. Fertil Steril. 95:625–630.e1-e4. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Mahalingam S, Ther L, Gao L, Wang W, Ziv-Gal A and Flaws JA: The effects of in utero bisphenol A exposure on ovarian follicle numbers and steroidogenesis in the F1 and F2 generations of mice. Reprod Toxicol. 74:150–157. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Kim D, Yoo ER, Li AA, Cholankeril G, Tighe SP, Kim W, Harrison SA and Ahmed A: Elevated urinary bisphenol A levels are associated with non-alcoholic fatty liver disease among adults in the United States. Liver Int. 39:1335–1342. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Eweda SM, Newairy AS, Abdou HM and Gaber AS: Bisphenol A-induced oxidative damage in the hepatic and cardiac tissues of rats: The modulatory role of sesame lignans. Exp Ther Med. 19:33–44. 2020.PubMed/NCBI

7 

Lin R, Jia Y, Wu F, Meng Y, Sun Q and Jia L: Combined exposure to fructose and bisphenol a exacerbates abnormal lipid metabolism in liver of developmental male rats. Int J Environ Res Public Health. 16:E41522019. View Article : Google Scholar : PubMed/NCBI

8 

Peerapanyasut W, Kobroob A, Palee S, Chattipakorn N and Wongmekiat O: N-Acetylcysteine attenuates the increasing severity of distant organ liver dysfunction after acute kidney injury in rats exposed to bisphenol a. Antioxidants (Basel). 8:E4972019. View Article : Google Scholar : PubMed/NCBI

9 

Peerapanyasut W, Kobroob A, Palee S, Chattipakorn N and Wongmekiat O: Activation of sirtuin 3 and maintenance of mitochondrial integrity by N-acetylcysteine protects against bisphenol A-induced kidney and liver toxicity in rats. Int J Mol Sci. 20:E2672019. View Article : Google Scholar : PubMed/NCBI

10 

Liao X, Zhou S, Zong J and Wang Z: Sevoflurane exerts protective effects on liver ischemia/reperfusion injury by regulating NFKB3 expression via miR-9-5p. Exp Ther Med. 17:2632–2640. 2019.PubMed/NCBI

11 

Song H, Du C, Wang X, Zhang J and Shen Z: [Erratum] MicroRNA-101 inhibits autophagy to alleviate liver ischemia/reperfusion injury via regulating the mTOR signaling pathway. Int J Mol Med. 43:25322019.PubMed/NCBI

12 

Burkhardt M, Slotta JE, Garcia P, Seekamp A, Menger MD and Pohlemann T: The effect of estrogen on hepatic microcirculation after ischemia/reperfusion. Int J Colorectal Dis. 23:113–119. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Lü P, Liu F, Wang CY, Chen DD, Yao Z, Tian Y, Zhang JH and Wu YH: Gender differences in hepatic ischemic reperfusion injury in rats are associated with endothelial cell nitric oxide synthase-derived nitric oxide. World J Gastroenterol. 11:3441–3445. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Yokoyama Y, Kuebler JF, Matsutani T, Schwacha MG, Bland KI and Chaudry IH: Mechanism of the salutary effects of 17beta-estradiol following trauma-hemorrhage: Direct downregulation of Kupffer cell proinflammatory cytokine production. Cytokine. 21:91–97. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Li W, Li D, Sun L, Li Z, Yu L and Wu S: The protective effects of estrogen on hepatic ischemia-reperfusion injury in rats by downregulating the Ang II/AT1R pathway. Biochem Biophys Res Commun. 503:2543–2548. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Viñas R and Watson CS: Mixtures of xenoestrogens disrupt estradiol-induced non-genomic signaling and downstream functions in pituitary cells. Environ Health. 12:262013. View Article : Google Scholar : PubMed/NCBI

17 

Desdoits-Lethimonier C, Lesné L, Gaudriault P, Zalko D, Antignac JP, Deceuninck Y, Platel C, Dejucq-Rainsford N, Mazaud-Guittot S and Jégou B: Parallel assessment of the effects of bisphenol A and several of its analogs on the adult human testis. Hum Reprod. 32:1465–1473. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Uzunhisarcikli M and Aslanturk A: Hepatoprotective effects of curcumin and taurine against bisphenol A-induced liver injury in rats. Environ Sci Pollut Res Int. 26:37242–37253. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Mendes-Braz M, Elias-Miró M, Jiménez-Castro MB, Casillas-Ramírez A, Ramalho FS and Peralta C: The current state of knowledge of hepatic ischemia-reperfusion injury based on its study in experimental models. J Biomed Biotechnol. 2012:2986572012. View Article : Google Scholar : PubMed/NCBI

20 

Wei X, Zheng W, Yang Z, Liu H, Tang T, Li X and Liu X: Effect of the intermittent Pringle maneuver on liver damage after hepatectomy: A retrospective cohort study. World J Surg Oncol. 17:1422019. View Article : Google Scholar : PubMed/NCBI

21 

Lin FS, Shen SQ, Chen ZB and Yan RC: 17β-estradiol attenuates reduced-size hepatic ischemia/reperfusion injury by inhibition apoptosis via mitochondrial pathway in rats. Shock. 37:183–190. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Nilsson EE, Sadler-Riggleman I and Skinner MK: Environmentally induced epigenetic transgenerational inheritance of disease. Environ Epigenet. 4:dvy0162018. View Article : Google Scholar : PubMed/NCBI

23 

Azushima K, Morisawa N, Tamura K and Nishiyama A: Recent research advances in renin-angiotensin-aldosterone system receptors. Curr Hypertens Rep. 22:222020. View Article : Google Scholar : PubMed/NCBI

24 

Feng P, Wu Z, Liu H, Shen Y, Yao X, Li X and Shen Z: Electroacupuncture improved chronic cerebral hypoperfusion-induced anxiety-like behavior and memory impairments in spontaneously hypertensive rats by downregulating the ACE/Ang II/AT1RAxis and upregulating the ACE2/Ang-(1–7)/MasR axis. Neural Plast. 2020:90760422020. View Article : Google Scholar : PubMed/NCBI

25 

Kawabe Y, Mori J, Morimoto H, Yamaguchi M, Miyagaki S, Ota T, Tsuma Y, Fukuhara S, Nakajima H, Oudit GY and Hosoi H: ACE2 exerts anti-obesity effect via stimulating brown adipose tissue and induction of browning in white adipose tissue. Am J Physiol Endocrinol Metab. 317:E1140–E1149. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Figueiredo VP, Barbosa MA, de Castro UGM, Zacarias AC, Bezerra FS, de Sá RG, de Lima WG, Dos Santos RAS and Alzamora AC: Antioxidant effects of oral Ang-(1–7) restore insulin pathway and RAS components ameliorating cardiometabolic disturbances in rats. Oxid Med Cell Longev. 2019:58689352019. View Article : Google Scholar : PubMed/NCBI

27 

Wang D, Chai XQ, Magnussen CG, Zosky GR, Shu SH, Wei X and Hu SS: Renin-angiotensin-system, a potential pharmacological candidate, in acute respiratory distress syndrome during mechanical ventilation. Pulm Pharmacol Ther. 58:1018332019. View Article : Google Scholar : PubMed/NCBI

28 

Chen Z, Li T, Zhang L, Wang H and Hu F: Bisphenol A exposure remodels cognition of male rats attributable to excitatory alterations in the hippocampus and visual cortex. Toxicology. 410:132–141. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Negishi T, Kawasaki K, Takatori A, Ishii Y, Kyuwa S, Kuroda Y and Yoshikawa Y: Effects of perinatal exposure to bisphenol A on the behavior of offspring in F344 rats. Environ Toxicol Pharmacol. 14:99–108. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Korkmaz A, Ahbab MA, Kolankaya D and Barlas N: Influence of vitamin C on bisphenol A, nonylphenol and octylphenol induced oxidative damages in liver of male rats. Food Chem Toxicol. 48:2865–2871. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Kazemi S, Mousavi SN, Aghapour F, Rezaee B, Sadeghi F and Moghadamnia AA: Induction effect of bisphenol A on gene expression involving hepatic oxidative stress in rat. Oxid Med Cell Longev. 2016:62985152016. View Article : Google Scholar : PubMed/NCBI

32 

Guo Y, Hu B, Huang H, Tsung A, Gaikwad NW, Xu M, Jiang M, Ren S, Fan J, Billiar TR, et al: Estrogen sulfotransferase is an oxidative stress-responsive gene that gender-specifically affects liver ischemia/reperfusion injury. J Biol Chem. 290:14754–14764. 2015. View Article : Google Scholar : PubMed/NCBI

33 

de Vries HA, Ponds FA, Nieuwenhuijs VB, Morphett A, Padbury RT and Barritt GJ: Evidence that estrogen receptors play a limited role in mediating enhanced recovery of bile flow in female rats in the acute phase of liver ischemia reperfusion injury. Ann Hepatol. 12:130–137. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Yang X, Qin L, Liu J, Tian L and Qian H: 17β-Estradiol protects the liver against cold ischemia/reperfusion injury through the Akt kinase pathway. J Surg Res. 178:996–1002. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Eckhoff DE, Bilbao G, Frenette L, Thompson JA and Contreras JL: 17-Beta-estradiol protects the liver against warm ischemia/reperfusion injury and is associated with increased serum nitric oxide and decreased tumor necrosis factor-alpha. Surgery. 132:302–309. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Shen SQ, Zhang Y and Xiong CL: The protective effects of 17beta-estradiol on hepatic ischemia-reperfusion injury in rat model, associated with regulation of heat-shock protein expression. J Surg Res. 140:67–76. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Vilatoba M, Eckstein C, Bilbao G, Frennete L, Eckhoff DE and Contreras JL: 17beta-estradiol differentially activates mitogen-activated protein-kinases and improves survival following reperfusion injury of reduced-size liver in mice. Transplant Proc. 37:399–403. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Ng IO, Ng M and Fan ST: Better survival in women with resected hepatocellular carcinoma is not related to tumor proliferation or expression of hormone receptors. Am J Gastroenterol. 92:1355–1358. 1997.PubMed/NCBI

39 

Yokoyama Y, Nagino M and Nimura Y: Which gender is better positioned in the process of liver surgery? Male or female? Surg Today. 37:823–830. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Konieczna A, Rutkowska A and Rachoń D: Health risk of exposure to Bisphenol A (BPA). Rocz Panstw Zakl Hig. 66:5–11. 2015.PubMed/NCBI

41 

Mehta PK and Griendling KK: Angiotensin II cell signaling: Physiological and pathological effects in the cardiovascular system. Am J Physiol Cell Physiol. 292:C82–C97. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Pueyo ME, Gonzalez W, Nicoletti A, Savoie F, Arnal JF and Michel JB: Angiotensin II stimulates endothelial vascular cell adhesion molecule-1 via nuclear factor-kappaB activation induced by intracellular oxidative stress. Arterioscler Thromb Vasc Biol. 20:645–651. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Yusof M, Kamada K, Gaskin FS and Korthuis RJ: Angiotensin II mediates postischemic leukocyte-endothelial interactions: Role of calcitonin gene-related peptide. Am J Physiol Heart Circ Physiol. 292:H3032–H3037. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Ruiz-Ortega M, Lorenzo O, Rupérez M, Blanco J and Egido J: Systemic infusion of angiotensin II into normal rats activates nuclear factor-kappaB and AP-1 in the kidney: Role of AT(1) and AT(2) receptors. Am J Pathol. 158:1743–1756. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Harrison DG, Cai H, Landmesser U and Griendling KK: Interactions of angiotensin II with NAD(P)H oxidase, oxidant stress and cardiovascular disease. J Renin Angiotensin Aldosterone Syst. 4:51–61. 2003. View Article : Google Scholar : PubMed/NCBI

46 

Li X, Cai W, Xi W, Sun W, Shen W, Wei T, Chen X, Sun L, Zhou H, Sun Y, et al: MicroRNA-31 Regulates immunosuppression in Ang II (Angiotensin II)-induced hypertension by targeting Ppp6C (Protein Phosphatase 6c). Hypertension. 73:e14–e24. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Alfany-Fernandez I, Casillas-Ramirez A, Bintanel-Morcillo M, Brosnihan KB, Ferrario CM, Serafin A, Rimola A, Rodés J, Roselló-Catafau J and Peralta C: Therapeutic targets in liver transplantation: Angiotensin II in nonsteatotic grafts and angiotensin-(1–7) in steatotic grafts. Am J Transplant. 9:439–451. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Sabry MM, Ramadan NM, Al Dreny BA, Rashed LA and Abo El Enein A: Protective effect of apelin preconditioning in a rat model of hepatic ischemia reperfusion injury; possible interaction between the apelin/APJ system, Ang II/AT1R system and eNOS. United European Gastroenterol J. 7:689–698. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Saura M, Marquez S, Reventun P, Olea-Herrero N, Arenas MI, Moreno-Gómez-Toledano R, Gómez-Parrizas M, Muñóz-Moreno C, González-Santander M, Zaragoza C and Bosch RJ: Oral administration of bisphenol A induces high blood pressure through angiotensin II/CaMKII-dependent uncoupling of eNOS. FASEB J. 28:4719–4728. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Gao F, Huang Y, Zhang L and Liu W: Involvement of estrogen receptor and GPER in bisphenol A induced proliferation of vascular smooth muscle cells. Toxicol In Vitro. 56:156–162. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Abbasloo E, Najafipour H and Vakili A: Chronic treatment with apelin, losartan and their combination reduces myocardial infarct size and improves cardiac mechanical function. Clin Exp Pharmacol Physiol. 47:393–402. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Panico K, Abrahão MV, Trentin-Sonoda M, Muzi-Filho H, Vieyra A and Carneiro-Ramos MS: Cardiac inflammation after ischemia-reperfusion of the kidney: Role of the sympathetic nervous system and the renin-angiotensin system. Cell Physiol Biochem. 53:587–605. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 22 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Shi Y, Li Z, Sun L, Zhang M, Yu L and Wu S: BPA disrupts 17‑estradiol‑mediated hepatic protection against ischemia/reperfusion injury in rat liver by upregulating the Ang II/AT1R signaling pathway. Mol Med Rep 22: 416-422, 2020
APA
Zhang, Y., Shi, Y., Li, Z., Sun, L., Zhang, M., Yu, L., & Wu, S. (2020). BPA disrupts 17‑estradiol‑mediated hepatic protection against ischemia/reperfusion injury in rat liver by upregulating the Ang II/AT1R signaling pathway. Molecular Medicine Reports, 22, 416-422. https://doi.org/10.3892/mmr.2020.11072
MLA
Zhang, Y., Shi, Y., Li, Z., Sun, L., Zhang, M., Yu, L., Wu, S."BPA disrupts 17‑estradiol‑mediated hepatic protection against ischemia/reperfusion injury in rat liver by upregulating the Ang II/AT1R signaling pathway". Molecular Medicine Reports 22.1 (2020): 416-422.
Chicago
Zhang, Y., Shi, Y., Li, Z., Sun, L., Zhang, M., Yu, L., Wu, S."BPA disrupts 17‑estradiol‑mediated hepatic protection against ischemia/reperfusion injury in rat liver by upregulating the Ang II/AT1R signaling pathway". Molecular Medicine Reports 22, no. 1 (2020): 416-422. https://doi.org/10.3892/mmr.2020.11072