DOI QR코드

DOI QR Code

MiR-26a promotes apoptosis of porcine granulosa cells by targeting the 3β-hydroxysteroid-Δ24-reductase gene

  • Zhang, Xiaodong (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Tao, Qiangqiang (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Shang, Jinnan (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Xu, Yiliang (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Zhang, Liang (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Ma, Yingchun (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Zhu, Weihua (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Yang, Min (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Ding, Yueyun (College of Animal Science and Technology, Anhui Agricultural University) ;
  • Yin, Zongjun (College of Animal Science and Technology, Anhui Agricultural University)
  • Received : 2019.03.03
  • Accepted : 2019.05.31
  • Published : 2020.04.01

Abstract

Objective: Apoptosis of ovarian granulosa cells (GCs) affects mammalian follicular development and fecundity. This study aimed to explore the regulatory relationship between microRNA-26a (miR-26a) and the 3β-hydroxysteroid-Δ24-reductase gene (DHCR24) gene in porcine follicular granular cells (pGCs), and to provide empirical data for the development of methods to improve the reproductive capacity of pigs. Methods: The pGCs were transfected with miR-26a mimic, miR-26a inhibitor and DHCR24-siRNA in vitro. The cell apoptosis rate of pGCs was detected by the flow cytometry. The secretion levels of estradiol (E2) and progesterone (P) in pGCs were detected by enzyme-linked immunosorbent assay. Double luciferase validation system was used to detect the binding sites between miR-26a and DHCR24 3'-UTR region. Qualitative real-time polymerase chain reaction and Western blotting were used to verify the DHCR24 mRNA and protein expression in pGCs, respectively, after transfecting with miR-26a mimic and miR-26a inhibitor. Results: Results showed that enhancement of miR-26a promoted apoptosis, and inhibited E2 and P secretion in pGCs. Meanwhile, inhibition of DHCR24 also upregulated the Caspase-3 expression, reduced the BCL-2 expression, promoted pGCs apoptosis, and inhibited E2 and P secretion in pGCs. There were the binding sites of miR-26a located within DHCR24 3'-UTR. Up-regulation of miR-26a inhibited DHCR24 mRNA and protein expression in pGCs. Conclusion: This study demonstrates that miR-26a can promote cell apoptosis and inhibit E2 and P secretion by inhibiting the expression of DHCR24 in pGCs.

Keywords

References

  1. McGee EA, Hsueh AJW. Initial and cyclic recruitment of ovarian follicles. Endocr Rev 2000;21:200-14. https://doi.org/10.1210/edrv.21.2.0394
  2. Pan Z, Zhang J, Lin F, Ma X, Wang X, Liu H. Expression profiles of key candidate genes involved in steroidogenesis during follicular atresia in the pig ovary. Mol Biol Rep 2012;39:10823-32. https://doi.org/10.1007/s11033-012-1976-2
  3. Peralta LE, Olarte MR, Arganaraz M, Ciocca D, Miceli DC. Progesterone receptors: Their localization, binding activity and expression in the pig oviduct during follicular and luteal phases. Domest Anim Endocrinol 2005;28:74-84. https://doi.org/10.1016/j.domaniend.2004.05.008
  4. Nie M, Yu S, Peng S, Fang Y, Wang H, Yang X. miR-23a and miR-27a promote human granulosa cell apoptosis by targeting SMAD5. Biol Reprod 2015;93:98. https://doi.org/10.1095/biolreprod.115.130690
  5. Xiong F, Hu L, Zhang Y, Xiao X, Xiao J. miR-22 inhibits mouse ovarian granulosa cell apoptosis by targeting SIRT1. Biol Open 2016;5:367-71. https://doi.org/10.1242/bio.016907
  6. Liu J, Tu F, Yao W, et al. Conserved miR-26b enhances ovarian granulosa cell apoptosis through HAS2-HA-CD44-Caspase-3 pathway by targeting HAS2. Sci Rep 2016;6:21197. https://doi.org/10.1038/srep21197
  7. Song Q, Xu K. MicroRNA-26a and tumor. Zhongguo Fei Ai Za Zhi 2017;20:769-74. https://doi.org/10.3779/j.issn.1009-3419.2017.11.08
  8. Ma Y, Deng F, Li P, Chen G, Tao Y, Wang H. The tumor suppressive miR-26a regulation of FBXO11 inhibits proliferation, migration and invasion of hepatocellular carcinoma cells. Biomed Pharmacother 2018;101:648-55. https://doi.org/10.1016/j.biopha.2018.02.118
  9. Liu J, Mi B, Wang Y, et al. miR-26a suppresses osteosarcoma migration and invasion by directly targeting HMGA1. Oncol Lett 2018;15:8303-10. https://doi.org/10.3892/ol.2018.8359
  10. Gong Y, Wu W, Zou X, Liu F, Wei T, Zhu J. MiR-26a inhibits thyroid cancer cell proliferation by targeting ARPP19. Am J Cancer Res 2018;8:1030-9.
  11. Su X, Liao L, Shuai Y, et al. MiR-26a functions oppositely in osteogenic differentiation of BMSCs and ADSCs depending on distinct activation and roles of Wnt and BMP signaling pathway. Cell Death Dis 2015;6:e1851. https://doi.org/10.1038/cddis.2015.221
  12. Zhou J, Ju WQ, Yuan XP, Zhu XF, Wang DP, He XS. miR-26a regulates mouse hepatocyte proliferation via directly targeting the 3' untranslated region of CCND2 and CCNE2. Hepatobiliary Pancreat Dis Int 2016;15:65-72. https://doi.org/10.1016/S1499-3872(15)60383-6
  13. Zhang Y, Qin W, Zhang L, et al. MicroRNA-26a prevents endothelial cell apoptosis by directly targeting TRPC6 in the setting of atherosclerosis. Sci Rep 2015;5:9401. https://doi.org/10.1038/srep09401
  14. Ran M, Li Z, Cao R, et al. miR-26a suppresses autophagy in swine Sertoli cells by targeting ULK2. Reprod Domest Anim 2018;53:864-71. https://doi.org/10.1111/rda.13177
  15. Zhang B, Chen L, Feng G, et al. MicroRNA mediating networks in granulosa cells associated with ovarian follicular development. Biomed Res Int 2017;2017:4585213. https://doi.org/10.1155/2017/4585213
  16. Huang L, Yin ZJ, Feng YF, et al. Identification and differential expression of microRNAs in the ovaries of pigs (Sus scrofa) with high and low litter sizes. Anim Genet 2016;47:543-51. https://doi.org/10.1111/age.12452
  17. Tian M, Zhang X, Ye P, et al. MicroRNA-21 and microRNA-214 play important role in reproduction regulation during porcine estrous. Anim Sci J 2018;89:1398-405. https://doi.org/10.1111/asj.13087
  18. Zerenturk EJ, Sharpe LJ, Ikonen E, Brown AJ. Desmosterol and DHCR24: unexpected new directions for a terminal step in cholesterol synthesis. Prog Lipid Res 2013;52:666-80. https://doi.org/10.1016/j.plipres.2013.09.002
  19. Sarkar D, Imai T, Kambe F, et al. The human homolog of Diminuto/Dwarf1 gene (hDiminuto): a novel ACTH-responsive gene overexpressed in benign cortisol-producing adrenocortical adenomas. J Clin Endocrinol Metab 2001;86:5130-7. https://doi.org/10.1210/jcem.86.11.8032
  20. Greeve I, Hermans-Borgmeyer I, Brellinger C, et al. The human DIMINUTO/DWARF1 homolog seladin-1 confers resistance to Alzheimer's disease-associated neurodegeneration and oxidative stress. J Neurosci 2000;20:7345-52. https://doi.org/10.1523/JNEUROSCI.20-19-07345.2000
  21. Lu X, Kambe F, Cao X, et al. 3beta-Hydroxysteroid-delta24 reductase is a hydrogen peroxide scavenger, protecting cells from oxidative stress-induced apoptosis. Endocrinology 2008;149:3267-73. https://doi.org/10.1210/en.2008-0024
  22. Maalouf SW, Liu WS, Pate JL. MicroRNA in ovarian function. Cell Tissue Res 2016;363:7-18. https://doi.org/10.1007/s00441-015-2307-4
  23. Schreiber NB, Totty ML, Spicer LJ. Expression and effect of fibroblast growth factor 9 in bovine theca cells. J Endocrinol 2012;215:167-75. https://doi.org/10.1530/JOE-12-0293
  24. Franks S, Stark J, Hardy K. Follicle dynamics and anovulation in polycystic ovary syndrome. Hum Reprod Update 2008;14:367-78. https://doi.org/10.1093/humupd/dmn015
  25. Pan B, Toms D, Shen W, Li J. MicroRNA-378 regulates oocyte maturation via the suppression of aromatase in porcine cumulus cells. Am J Physiol Endocrinol Metab 2015;308:E525-34. https://doi.org/10.1152/ajpendo.00480.2014
  26. Liu J, Li X, Yao Y, Li Q, Pan Z, Li Q. miR-1275 controls granulosa cell apoptosis and estradiol synthesis by impairing LRH-1/CYP19A1 axis. Biochim Biophys Acta Gene Regul Mech 2018;1861:246-57. https://doi.org/10.1016/j.bbagrm.2018.01.009
  27. Liu J, Du X, Zhou J, Pan Z, Liu H, Li Q. MicroRNA-26b functions as a proapoptotic factor in porcine follicular Granulosa cells by targeting Sma-and Mad-related protein 4. Biol Reprod 2014;91:146. https://doi.org/10.1095/biolreprod.114.122788
  28. Zhang L, Liu X, Liu J, et al. miR-26a promoted endometrial epithelium cells (EECs) proliferation and induced stromal cells (ESCs) apoptosis via the PTEN-PI3K/AKT pathway in dairy goats. J Cell Physiol 2018;233:4688-706. https://doi.org/10.1002/jcp.26252
  29. Saito M, Takano T, Nishimura T, Kohara M, Tsukiyama-Kohara K. 3beta-hydroxysterol delta24-reductase on the surface of hepatitis C virus-related hepatocellular carcinoma cells can be a target for molecular targeting therapy. PLoS One 2015;10:e0124197. https://doi.org/10.1371/journal.pone.0124197
  30. Lu X, Jia D, Zhao C, et al. Recombinant adenovirus-mediated overexpression of 3beta-hydroxysteroid-Delta24 reductase. Neural Regen Res 2014;9:504-12. https://doi.org/10.4103/1673-5374.130074
  31. Luciani P, Deledda C, Rosati F, et al. Seladin-1 is a fundamental mediator of the neuroprotective effects of estrogen in human neuroblast long-term cell cultures. Endocrinology 2008;149: 4256-66. https://doi.org/10.1210/en.2007-1795
  32. Peri A, Benvenuti S, Luciani P, Deledda C, Cellai I. Membrane cholesterol as a mediator of the neuroprotective effects of estrogens. Neuroscience 2011;191:107-17. https://doi.org/10.1016/j.neuroscience.2011.03.011
  33. Juengel JL, Heath DA, Quirke LD, Mcnatty KP. Oestrogen receptor alpha and beta, androgen receptor and progesterone receptor mRNA and protein localisation within the developing ovary and in small growing follicles of sheep. Reproduction 2006;131:81-92. https://doi.org/10.1530/rep.1.00704
  34. Williams AH, Valdez G, Moresi V, et al. MicroRNA-206 delays ALS progression and promotes regeneration of neuromuscular synapses in mice. Science 2009;326:1549-54. https://doi.org/10.1126/science.1181046
  35. Hwang HW, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science 2007;315:97-100. https://doi.org/10.1126/science.1136235
  36. Lal A, Thomas MP, Altschuler G, et al. Capture of microRNA-bound mRNAs identifies the tumor suppressor miR-34a as a regulator of growth factor signaling. PLoS Genet 2011;7:e1002363. https://doi.org/10.1371/journal.pgen.1002363
  37. Mohr AM, Mott JL. Overview of microRNA biology. Semin Liver Dis 2015;35:3-11. https://doi.org/10.1055/s-0034-1397344
  38. Yuan XL, Deng X, Zhou XF, et al. MiR-126-3p promotes the cell proliferation and inhibits the cell apoptosis by targeting TSC1 in the porcine granulosa cells. In Vitro Cell Dev Biol Anim 2018;54:715-24. https://doi.org/10.1007/s11626-018-0292-0

Cited by

  1. Transcriptional analysis of deoxynivalenol‐induced apoptosis of sow ovarian granulosa cell vol.55, pp.2, 2020, https://doi.org/10.1111/rda.13610
  2. microRNA-26a shuttled by extracellular vesicles secreted from adipose-derived mesenchymal stem cells reduce neuronal damage through KLF9-mediated regulation of TRAF2/KLF2 axis vol.10, pp.1, 2020, https://doi.org/10.1080/21623945.2021.1938829
  3. circRNA-Mediated Inhibin-Activin Balance Regulation in Ovarian Granulosa Cell Apoptosis and Follicular Atresia vol.22, pp.17, 2020, https://doi.org/10.3390/ijms22179113