Browse > Article
http://dx.doi.org/10.5713/ajas.20.0292

Zearalenone exposure affects the Wnt/β-catenin signaling pathway and related genes of porcine endometrial epithelial cells in vitro  

Song, Tingting (Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University)
Yang, Weiren (Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University)
Huang, Libo (Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University)
Yang, Zaibin (Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University)
Jiang, Shuzhen (Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University)
Publication Information
Animal Bioscience / v.34, no.6, 2021 , pp. 993-1005 More about this Journal
Abstract
Objective: Zearalenone (ZEA) has estrogen-like effects. Our previous study has shown that ZEA (0.5 to 1.5 mg/kg) could induce abnormal uterine proliferation through transforming growth factor signaling pathway. To further study the other regulatory networks of uterine hypertrophy caused by ZEA, the potential mechanism of ZEA on porcine endometrial epithelial cells (PECs) was explored by the Illumina Hiseq 2000 sequencing system. Methods: The PECs were treated with ZEA at 0 (ZEA0), 5 (ZEA5), 20 (ZEA20), and 80 (ZEA80) µmol/L for 24 h. The collected cells were subjected to cell cycle, RNA-seq, real-time quantitative polymerase chain reaction, immunofluorescence, and western blot analysis. Results: The proportion of cells in the S and G2 phases decreased (p<0.05), but the proportion of cells in the G1 phase increased (p<0.05) in the ZEA80 treatment. Data analysis revealed that the expression of Wnt pathway-related genes, estrogen-related genes, and mitogen-activated protein kinase pathway-related genes increased (p<0.05), but the expression of genetic stability genes decreased (p<0.05) with increasing ZEA concentrations. The relative mRNA and protein expression of WNT1, β-catenin, glycogen synthase kinase 3β (GSK-3β) were increased (p<0.05) with ZEA increasing, while the relative mRNA and protein expression of cyclin D1 (CCND1) was decreased (p<0.05). Moreover, our immunofluorescence results indicate that β-catenin accumulated around the nucleus from the cell membrane and cytoplasm with increasing ZEA concentrations. Conclusion: In summary, ZEA can activate the Wnt/β-catenin signaling pathway by up-regulating WNT1 and β-catenin expression, to promote the proliferation and development of PECs. At the same time, the up-regulation of GSK-3β and down-regulation of CCND1, as well as the mRNA expression of other pathway related genes indicated that other potential effects of ZEA on the uterine development need further study.
Keywords
Zearalenone; Porcine Endometrial Epithelial Cells; WNT1; ${\beta}$-Catenin; GSK-$3{\beta}$; Cyclin D1;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Jiang SZ, Yang ZB, Yang WR, et al. Effects of purified zearalenone on growth performance, organ size, serum metabolites, and oxidative stress in postweaning gilts. J Anim Sci 2011;89:3008-15. https://doi.org/10.2527/jas.2010-3658   DOI
2 Jiang SZ, Yang ZB, Yang WR, et al. Effect of purified zearalenone with or without modified montmorillonite on nutrient availability, genital organs and serum hormones in post-weaning piglets. Livest Sci 2012;144:110-8. https://doi.org/10.1016/j.livsci.2011.11.004   DOI
3 Etienne M, Jemmali M. Effects of zearalenone (F2) on estrous activity and reproduction in gilts. J Anim Sci 1982;55:1-10. https://doi.org/10.2527/jas1982.5511   DOI
4 Chen XX, Yang CW, Huang LB, Niu QS, Jiang SZ, Chi F. Zearalenone altered the serum hormones, morphologic and apoptotic measurements of genital organs in post-weaning gilts. Asian-Australas J Anim Sci 2015;28:171-9. https://doi.org/10.5713/ajas.14.0329   DOI
5 Zhou M, Yang LJ, Yang WR, et al. Effects of zearalenone on the localization and expression of the growth hormone receptor gene in the uteri of post-weaning piglets. Asian-Australas J Anim Sci 2018;31:32-9. https://doi.org/10.5713/ajas.17.0526   DOI
6 Zhou M, Yang L, Chen Y, et al. Comparative study of stress response, growth and development of uteri in post-weaning gilts challenged with zearalenone and estradiol benzoate. J Anim Physiol Anim Nutr 2019;103:1885-94. https://doi.org/10.1111/jpn.13195   DOI
7 Duchartre Y, Kim YM, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol 2016;99:141-9. https://doi.org/10.1016/j.critrevonc.2015.12.005   DOI
8 Andl T, Reddy ST, Gaddapara T, Millar SE. Wnt signals are required for the initiation of hair follicle development. Dev Cell 2002;2:643-53. https://doi.org/10.1016/S1534-5807(02)00167-3   DOI
9 Lee EY, Choi EJ, Kim JA, et al. Malva verticillata seed extracts upregulate the wnt pathway in human dermal papilla cells. Int J Cosmet Sci 2016;38:148-54. https://doi.org/10.1111/ics.12268   DOI
10 Bai C, Zhang H, Zhang X, Yang W, Li X, Gao Y. MiR-15/16 mediate crosstalk between the MAPK and Wnt/β-catenin pathways during hepatocyte differentiation from amniotic epithelial cells. Biochim Biophys Acta Gene Regul Mech 2019;1862:567-81. https://doi.org/10.1016/j.bbagrm.2019.02.003   DOI
11 Othmen ZOB, Golli EE, Abid-Essefi S, Bacha H. Cytotoxicity effects induced by Zearalenone metabolites, α Zearalenol and β Zearalenol, on cultured Vero cells. Toxicology 2008;252:72-7. https://doi.org/10.1016/j.tox.2008.07.065   DOI
12 Rogowska A, Pomastowski P, Rafinska K, et al. A study of zearalenone biosorption and metabolisation by prokaryotic and eukaryotic cells. Toxicon 2019;169:81-90. https://doi.org/10.1016/j.toxicon.2019.09.008   DOI
13 Maragos C. Zearalenone occurrence and human exposure. World Mycotoxin J 2010;3:369-83. https://doi.org/10.3920/WMJ2010.1240   DOI
14 Zhang K, Li H, Dong S, et al. Establishment and evaluation of a PRRSV-sensitive porcine endometrial epithelial cell line by transfecting SV40 large T antigen. BMC Vet Res 2019;15:299. https://doi.org/10.1186/s12917-019-2051-1   DOI
15 Luna-Medina R, Cortes-Canteli M, Sanchez-Galiano S, et al. NP031112, a thiadiazolidinone compound, prevents inflammation and neurodegeneration under excitotoxic conditions: potential therapeutic role in brain disorders. J Neurosci 2007;27:5766-76. https://doi.org/10.1523/JNEUROSCI.1004-07.2007   DOI
16 Yang LJ, Zhou M, Huang LB, et al. Zearalenone-promoted follicle growth through modulation of Wnt-1/β-catenin signaling pathway and expression of estrogen receptor genes in ovaries of postweaning piglets. J Agric Food Chem 2018;66:7899-906. https://doi.org/10.1021/acs.jafc.8b02101   DOI
17 Vlad-Fiegen A, Langerak A, Eberth S, Muller O. The Wnt pathway destabilizes adherens junctions and promotes cell migration via β-catenin and its target gene cyclin D1. FEBS Open Bio 2012;2:26-31. https://doi.org/10.1016/j.fob.2012.02.004   DOI
18 Zeng X, Tamai K, Doble B, et al. A dual-kinase mechanism for Wnt co-receptor phosphorylation and activation. Nature 2005;438:873-7. https://doi.org/10.1038/nature04185   DOI
19 Huang P, Yan R, Zhang X, Wang L, Ke X, Qu Y. Activating Wnt/β-catenin signaling pathway for disease therapy: challenges and opportunities. Pharmacol Ther 2019;196:79-90. https://doi.org/10.1016/j.pharmthera.2018.11.008   DOI
20 Kumawat K, Koopmans T, Gosens R. β-catenin as a regulator and therapeutic target for asthmatic airway remodeling. Expert Opin Ther Targets 2014;18:1023-34. https://doi.org/10.1517/14728222.2014.934813   DOI
21 De Luca A, Maiello MR, D'Alessio A, Pergameno M, Normanno N. The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: role in cancer pathogenesis and implications for therapeutic approaches. Expert Opin Ther Targets 2012;16(Suppl 2):S17-27. https://doi.org/10.1517/14728222.2011.639361   DOI
22 Ye X, Guo Y, Zhang Q, et al. βKlotho suppresses tumor growth in hepatocellular carcinoma by regulating Akt/GSK-3β/cyclin D1 signaling pathway. PLoS One 2013;8:e55615. https://doi.org/10.1371/journal.pone.0055615   DOI
23 Zheng W, Wang B, Li X, et al. Zearalenone promotes cell proliferation or causes cell death? Toxins 2018;10:184. https://doi.org/10.3390/toxins10050184   DOI
24 Kunishige K, Kawate N, Inaba T, Tamada H. Exposure to zearalenone during early pregnancy causes estrogenic multitoxic effects in mice. Reprod Sci 2017;24:421-7. https://doi.org/10.1177/1933719116657194   DOI
25 Jiang H, Luo S, Li H. Cdk5 activator-binding protein C53 regulates apoptosis induced by genotoxic stress via modulating the G2/M DNA damage checkpoint. J Biol Chem 2005;280:20651-9. https://doi.org/10.1074/jbc.M413431200   DOI
26 Heneweer M, Houtman R, Poortman J, Groot M, Maliepaard C, Peijnenburg A. Estrogenic effects in the immature rat uterus after dietary exposure to ethinylestradiol and zearalenone using a systems biology approach. Toxicol Sci 2007;99:303-14. https://doi.org/10.1093/toxsci/kfm151   DOI
27 Pazhohan A, Amidi F, Akbari-Asbagh F, et al. The Wnt/β-catenin signaling in endometriosis, the expression of total and active forms of β-catenin, total and inactive forms of glycogen synthase kinase-3β, Wnt7a and Dickkopf-1. Eur J Obstet Gynecol Reprod Biol 2018;220:1-5. https://doi.org/10.1016/j.ejogrb.2017.10.025   DOI
28 Li J, He T, Yuan H. Expression of GSK-3β and E-cadherin in endometrial cancer and its significance. Chongqing Med J 2019;48:288-92.
29 Koehler A, Schlupf J, Schneider M, Kraft B, Winter C, Kashef J. Loss of Xenopus cadherin-11 leads to increased Wnt/β-catenin signaling and up-regulation of target genes c-myc and cyclin D1 in neural crest. Dev Biol 2013;383:132-45. https://doi.org/10.1016/j.ydbio.2013.08.007   DOI
30 Liu XL, Wu RY, Sun XF, et al. Mycotoxin zearalenone exposure impairs genomic stability of swine follicular granulosa cells in vitro. Int J Biol Sci 2018;14:294-305. https://doi.org/10.7150/ijbs.23898   DOI