DOI QR코드

DOI QR Code

Regulation of chicken vanin1 gene expression by peroxisome proliferators activated receptor α and miRNA-181a-5p

  • Wang, Zhongliang (School of Biology and Food Engineering, Changshu Institute of Technology) ;
  • Yu, Jianfeng (School of Biology and Food Engineering, Changshu Institute of Technology) ;
  • Hua, Nan (School of Biology and Food Engineering, Changshu Institute of Technology) ;
  • Li, Jie (School of Biology and Food Engineering, Changshu Institute of Technology) ;
  • Xu, Lu (School of Biology and Food Engineering, Changshu Institute of Technology) ;
  • Yao, Wen (College of Animal Science and Technology, Nanjing Agricultural University) ;
  • Gu, Zhiliang (School of Biology and Food Engineering, Changshu Institute of Technology)
  • Received : 2019.12.31
  • Accepted : 2020.03.30
  • Published : 2021.02.01

Abstract

Objective: Vanin1 (VNN1) is a pantetheinase that can catalyze the hydrolysis of pantetheine to produce pantothenic acid and cysteamine. Our previous studies showed that VNN1 is specifically expressed in chicken liver. In this study, we aimed to investigate the roles of peroxisome proliferators activated receptor α (PPARα) and miRNA-181a-5p in regulating VNN1 gene expression in chicken liver. Methods: 5'-RACE was performed to identify the transcription start site of chicken VNN1. JASPAR and TFSEARCH were used to analyze the potential transcription factor binding sites in the promoter region of chicken VNN1 and miRanda was used to search miRNA binding sites in 3' untranslated region (3'UTR) of chicken VNN1. We used a knock-down strategy to manipulate PPARα (or miRNA-181a-5p) expression levels in vitro to further investigate its effect on VNN1 gene transcription. Luciferase reporter assays were used to explore the specific regions of VNN1 targeted by PPARα and miRNA-181a-5p. Results: Sequence analysis of the VNN1 promoter region revealed several transcription factor-binding sites, including hepatocyte nuclear factor 1α (HNF1α), PPARα, and CCAAT/enhancer binding protein α. GW7647 (a specific agonist of PPARα) increased the expression level of VNN1 mRNA in chicken primary hepatocytes, whereas knockdown of PPARα with siRNA increased VNN1 mRNA expression. Moreover, the predicted PPARα-binding site was confirmed to be necessary for PPARα regulation of VNN1 gene expression. In addition, the VNN1 3'UTR contains a sequence that is completely complementary to nucleotides 1 to 7 of miRNA-181a-5p. Overexpression of miR-181a-5p significantly decreased the expression level of VNN1 mRNA. Conclusion: This study demonstrates that PPARα is an important transcriptional activator of VNN1 gene expression and that miRNA-181a-5p acts as a negative regulator of VNN1 expression in chicken hepatocytes.

Keywords

References

  1. Dupre S, Graziani MT, Rosei MA, Fabi A, Del Grosso E. The enzymatic breakdown of pantethine to pantothenic acid and cystamine. Eur J Biochem 1970;16:571-8. https://doi.org/10.1111/j.1432-1033.1970.tb01119.x
  2. Sendo F, Araki Y. Regulation of leukocyte adherence and migration by glycosylphosphatidyl-inositol-anchored proteins. J Leukoc Biol 1999;66:369-74. https://doi.org/10.1002/jlb.66.3.369
  3. Berruyer C, Pouyet L, Millet V, et al. Vanin-1 licenses inflammatory mediator production by gut epithelial cells and controls colitis by antagonizing peroxisome proliferator-activated receptor γ activity. J Exp Med 2006;203:2817-27. https://doi.org/10.1084/jem.20061640
  4. Jansen PAM, Kamsteeg M, Rodijk-Olthuis D, et al. Expression of the vanin gene family in normal and inflamed human skin: induction by proinflammatory cytokines. J Investig Dermatol 2009;129:2167-74. https://doi.org/10.1038/jid.2009.67
  5. van Diepen JA, Jansen PA, Ballak DB, et al. PPAR-alpha dependent regulation of vanin-1 mediates hepatic lipid metabolism. J Hepatol 2014;61:366-72. https://doi.org/10.1016/j.jhep.2014.04.013
  6. Li Y, Wang X, Yu J, et al. MiR-122 targets the vanin 1 gene to regulate its expression in chickens. Poult Sci 2016;95:1145-50. https://doi.org/10.3382/ps/pew039
  7. Chen S, Zhang W, Tang C, Tang X, Liu L, Liu C. Vanin-1 is a key activator for hepatic gluconeogenesis. Diabetes 2014;63:2073-85. https://doi.org/10.2337/db13-0788
  8. Berruyer C, Martin FM, Castellano R, et al. Vanin-1-/- mice exhibit a glutathione-mediated tissue resistance to oxidative stress. Mol Cell Biol 2004;24:7214-24. https://doi.org/10.1128/MCB.24.16.7214-7224.2004
  9. Wang X, Shao F, Yu J, Jiang H, Gong D, Gu Z. MicroRNA-122 targets genes related to liver metabolism in chickens. Comp Biochem Physiol B Biochem Mol Biol 2015;184:29-35. https://doi.org/10.1016/j.cbpb.2015.02.002
  10. Lefebvre P, Chinetti G, Fruchart JC, Staels B. Sorting out the roles of PPARα in energy metabolism and vascular homeostasis. J Clin Invest 2006;116:571-80. https://doi.org/10.1172/JCI27989
  11. Hashimoto T, Cook WS, Qi C, Yeldandi AV, Reddy JK, Rao MS. Defect in peroxisome proliferator-activated receptor α-inducible fatty acid oxidation determines the severity of hepatic steatosis in response to fasting. J Biol Chem 2000;275:28918-28. https://doi.org/10.1074/jbc.M910350199
  12. Rommelaere S, Millet V, Gensollen T, et al. PPARalpha regulates the production of serum Vanin-1 by liver. FEBS Lett 2013;587:3742-8. https://doi.org/10.1016/j.febslet.2013.09.046
  13. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281-97. https://doi.org/10.1016/S0092-8674(04)00045-5
  14. Kaucsar T, Racz Z, Hamar P. Post-transcriptional gene-expression regulation by micro RNA (miRNA) network in renal disease. Adv Drug Deliv Rev 2010;62:1390-401. https://doi.org/10.1016/j.addr.2010.10.003
  15. Lewis BP, Shih I, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell 2003;115: 787-98. https://doi.org/10.1016/S0092-8674(03)01018-3
  16. Friedman RC, Farh KKH, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 2009;19:92-105. https://doi.org/10.1101/gr.082701.108
  17. Stefani G, Slack FJ. Small non-coding RNAs in animal development. Nat Rev Mol Cell Biol 2008;9:219-30. https://doi.org/10.1038/nrm2347
  18. Lovis P, Roggli E, Laybutt DR, et al. Alterations in microRNA expression contribute to fatty acid-induced pancreatic β-cell dysfunction. Diabetes 2008;57:2728-36. https://doi.org/10.2337/db07-1252
  19. Ramirez CM, Goedeke L, Rotllan N, et al. MicroRNA 33 regulates glucose metabolism. Mol Cell Biol 2013;33:2891-902. https://doi.org/10.1128/MCB.00016-13
  20. Fu X, Dong B, Tian Y, et al. MicroRNA-26a regulates insulin sensitivity and metabolism of glucose and lipids. J Clin Invest 2015;125:2497-509. https://doi.org/10.1172/JCI75438
  21. Deeb N, Lamont SJ. Genetic architecture of growth and body composition in unique chicken populations. J Hered 2002;93:107-18. https://doi.org/10.1093/jhered/93.2.107
  22. Jennen DGJ, Vereijken ALJ, Bovenhuis H, et al. Detection and localization of quantitative trait loci affecting fatness in broilers. Poult Sci 2004;83:295-301. https://doi.org/10.1093/ps/83.3.295
  23. Park TS, Park J, Lee JH, Park JW, Park BC. Disruption of G0/G1 switch gene 2 (G0S2) reduced abdominal fat deposition and altered fatty acid composition in chicken. FASEB J 2019; 33:1188-98. https://doi.org/10.1096/fj.201800784R
  24. Martin F, Penet MF, Malergue F, et al. Vanin-1-/- mice show decreased NSAID- and Schistosoma-induced intestinal inflammation associated with higher glutathione stores. J Clin Invest 2004;113:591-7. https://doi.org/10.1172/JCI19557
  25. Frith MC, Valen E, Krogh A, Hayashizaki Y, Carninci P, Sandelin A. A code for transcription initiation in mammalian genomes. Genome Res 2008;18:1-12. https://doi.org/10.1101/gr.6831208
  26. Gensollen T, Bourges C, Rihet P, et al. Functional polymorphisms in the regulatory regions of the VNN1 gene are associated with susceptibility to inflammatory bowel diseases. Inflamm Bowel Dis 2013;19:2315-25. https://doi.org/10.1097/MIB.0b013e3182a32b03
  27. Wilson MJ, Jeyasuria P, Parker KL, Koopman P. The transcription factors steroidogenic factor-1 and SOX9 regulate expression of Vanin-1 during mouse testis development. J Biol Chem 2005;280:5917-23. https://doi.org/10.1074/jbc.M412806200
  28. Rakhshandehroo M, Knoch B, Muller M, Kersten S. Peroxisome proliferator-activated receptor alpha target genes. PPAR Res 2010;2010:612089. https://doi.org/10.1155/2010/612089
  29. Li H, Ma Z, Jia L, et al. Systematic analysis of the regulatory functions of microRNAs in chicken hepatic lipid metabolism. Sci Rep 2016;6:31766. https://doi.org/10.1038/srep31766
  30. Ouyang YB, Lu Y, Yue S, Giffard RG. MiR-181 targets multiple Bcl-2 family members and influences apoptosis and mitochondrial function in astrocytes. Mitochondrion 2012;12:213-9. https://doi.org/10.1016/j.mito.2011.09.001
  31. Mintz PJ, Saetrom P, Reebye V, et al. MicroRNA-181a* targets nanog in a subpopulation of CD34+ cells isolated from peripheral blood. Mol Ther Nucleic Acids 2012;1:e34. https://doi.org/10.1038/mtna.2012.29
  32. Li H, Chen X, Guan L, et al. MiRNA-181a regulates adipogenesis by targeting tumor necrosis factor-α (TNF-α) in the porcine model. PLoS One 2013;8:e71568. https://doi.org/10.1371/journal.pone.0071568
  33. Ouyang D, Xu L, Zhang L, et al. MiR-181a-5p regulates 3T3-L1 cell adipogenesis by targeting Smad7 and Tcf7l2. Acta Biochim Biophys Sin 2016;48:1034-41. https://doi.org/10.1093/abbs/gmw100
  34. Zhang Z, Gao Y, Xu MQ, et al. MiR-181a regulate porcine preadipocyte differentiation by targeting TGFBR1. Gene 2019;681:45-51. https://doi.org/10.1016/j.gene.2018.09.046
  35. Chu B, Wu T, Miao L, Mei Y, Wu M. MiR-181a regulates lipid metabolism via IDH1. Sci Rep 2015;5:8801. https://doi.org/10.1038/srep08801