DOI QR코드

DOI QR Code

MiR-126-3p inhibits apoptosis and promotes proliferation by targeting phosphatidylinositol 3-kinase regulatory subunit 2 in porcine ovarian granulosa cells

  • Zhou, Xiaofeng (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • He, Yingting (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • Jiang, Yao (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • He, Bo (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • Deng, Xi (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • Zhang, Zhe (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • Yuan, Xiaolong (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University) ;
  • Li, Jiaqi (Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University)
  • 투고 : 2019.04.07
  • 심사 : 2019.08.14
  • 발행 : 2020.06.01

초록

Objective: Numerous studies have indicated that the apoptosis and proliferation of granulosa cells (GCs) are closely related to the normal growth and development of follicles and ovaries. Previous evidence has suggested that miR-126-3p might get involved in the apoptosis and proliferation of GCs, and phosphatidylinositol 3-kinase regulatory subunit 2 (PIK3R2) gene has been predicted as one target of miR-126-3p. However, the molecular regulation of miR-126-3p on PIK3R2 and the effects of PIK3R2 on porcine GCs apoptosis and proliferation remain virtually unexplored. Methods: In this study, using porcine GCs as a cellular model, luciferase report assay, mutation and deletion were applied to verify the targeting relationship between miR-126-3p and PIK3R2. Annexin-V/PI staining and 5-ethynyl-2'-deoxyuridine assay were applied to explore the effect of PIK3R2 on GCs apoptosis and proliferation, respectively. Real-time quantitative polymerase chain reaction and Western Blot were applied to explore the regulation of miR-126-3p on PIK3R2 expression. Results: We found that miR-126-3p targeted at PIK3R2 and inhibited its mRNA and protein expression. Knockdown of PIK3R2 significantly inhibited the apoptosis and promoted the proliferation of porcine GCs, and significantly down-regulated the mRNA expression of several key genes of PI3K pathway such as insulin-like growth factor 1 receptor (IGF1R), insulin receptor (INSR), pyruvate dehydrogenase kinase 1 (PDK1), and serine/threonine kinase 1 (AKT1). Conclusion: MiR-126-3p might target and inhibit the mRNA and protein expressions of PIK3R2, thereby inhibiting GC apoptosis and promoting GC proliferation by down-regulating several key genes of the PI3K pathway, IGF1R, INSR, PDK1, and AKT1. These findings would provide great insight into further exploring the molecular regulation of miR-126-3p and PIK3R2 on the functions of GCs during the folliculogenesis in female mammals.

키워드

참고문헌

  1. Pepling ME, Spradling AC. Mouse ovarian germ cell cysts undergo programmed breakdown to form primordial follicles. Dev Biol 2001;234:339-51. https://doi.org/10.1006/dbio.2001.0269
  2. 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
  3. Reddy P, Shen L, Ren C, et al. Activation of Akt (PKB) and suppression of FKHRL1 in mouse and rat oocytes by stem cell factor during follicular activation and development. Dev Biol 2005;281:160-70. https://doi.org/10.1016/j.ydbio.2005.02.013
  4. Fiedler SD, Carletti MZ, Hong XM, Christenson LK. Hormonal regulation of microRNA expression in periovulatory mouse mural granulosa cells. Biol Reprod 2008;79:1030-7. https://doi.org/10.1095/biolreprod.108.069690
  5. Eppig JJ. Oocyte control of ovarian follicular development and function in mammals. Reproduction 2001;122:829-38. https://doi.org/10.1530/rep.0.1220829
  6. Shimizu T, Kosaka N, Murayama C, Tetsuka M, Miyamoto A. Apelin and APJ receptor expression in granulosa and theca cells during different stages of follicular development in the bovine ovary: Involvement of apoptosis and hormonal regulation. Anim Reprod Sci 2009;116:28-37. https://doi.org/10.1016/j.anireprosci.2009.01.009
  7. Zhang P, Wang J, Lang H, et al. Knockdown of CREB1 promotes apoptosis and decreases estradiol synthesis in mouse granulosa cells. Biomed Pharmacother 2018;105:1141-6. https://doi.org/10.1016/j.biopha.2018.06.101
  8. Maneixa L, Antonsonb P, Humireb P, et al. Estrogen receptor $\beta$ exon 3-deleted mouse: The importance of non-ERE pathways in $ER{\beta}$ signaling. Proc Natl Acad Sci USA 2015;112: 5135-40. https://doi.org/10.1073/pnas.1504944112
  9. Woodruff TK, Mayo KE. To beta or not to beta: estrogen receptors and ovarian function. Endocrinology 2005;146:3244-6. https://doi.org/10.1210/en.2005-0630
  10. Rodriguez KF, Couse JF, Jayes FL, et al. Insufficient luteinizing hormone-induced intracellular signaling disrupts ovulation in preovulatory follicles lacking estrogen receptor-$\beta$. Endocrinology 2010;151:2826-34. https://doi.org/10.1210/en.2009-1446
  11. Lei L, Jin S, Gonzalez G, Behringer RR, Woodruff TK. The regulatory role of Dicer in folliculogenesis in mice. Mol Cell Endocrinol 2010;315:63-73. https://doi.org/10.1016/j.mce.2009.09.021
  12. Hamm ML, Bhat GK, Thompson WE, Mann DR. Folliculogenesis is impaired and granulosa cell apoptosis is increased in leptin deficient mice. Biol Reprod 2004;71:66-72. https://doi.org/10.1095/biolreprod.104.027292
  13. Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol 2009;10:126-39. https://doi.org/10.1038/nrm2632
  14. Hong X, Luense LJ, McGinnis LK, Nothnick WB, Christenson LK. Dicer1 is essential for female fertility and normal development of the female reproductive system. Endocrinology 2008;149:6207-12. https://doi.org/10.1210/en.2008-0294
  15. Nagaraja AK, Andreu-Vieyra C, Franco HL, et al. Deletion of Dicer in somatic cells of the female reproductive tract causes sterility. Mol Endocrinol 2008;22:2336-52. https://doi.org/10.1210/me.2008-0142
  16. Yao G, Yin M, Lian J, et al. MicroRNA-224 is involved in transforming growth factor-beta-mediated mouse granulosa cell proliferation and granulosa cell function by targeting Smad4. Mol Endocrinol 2010;24:540-51. https://doi.org/10.1210/me.2009-0432
  17. Lin F, Li R, Pan ZX, et al. miR-26b promotes granulosa cell apoptosis by targeting ATM during follicular atresia in porcine ovary. PLoS ONE 2012;7:e38640. https://doi.org/10.1371/journal.pone.0038640
  18. Bernstein C, Bernstein H, Payne CM, Garewal H. DNA repair/ pro-apoptotic dual-role proteins in five major DNA repair pathways: fail-safe protection against carcinogenesis. Mutat Res 2002;511:145-78. https://doi.org/10.1016/S1383-5742(02)00009-1
  19. Ma T, Jiang H, Gao Y, et al. Microarray analysis of differentially expressed microRNAs in non-regressed and regressed bovine corpus luteum tissue; microRNA-378 may suppress luteal cell apoptosis by targeting the interferon gamma receptor 1 gene. J Appl Genet 2011;52:481-6. https://doi.org/10.1007/s13353-011-0055-z
  20. Mao Z, Fan L, Yu Q, et al. Abnormality of klotho signaling is involved in polycystic ovary syndrome. Reprod Sci 2018;25:372-83. https://doi.org/10.1177/1933719117715129
  21. Yuan X, Deng X, Zhou X, 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
  22. Qu Y, Wu J, Deng JX, et al. MicroRNA-126 affects rheumatoid arthritis synovial fibroblast proliferation and apoptosis by targeting PIK3R2 and regulating PI3K-AKT signal pathway. Oncotarget 2016;7:74217-26. https://doi.org/10.18632/oncotarget.12487
  23. Wu XJ, Zhao ZF, Kang XJ, Wang HJ, Zhao J, Pu XM. MicroRNA-126-3p suppresses cell proliferation by targeting PIK3R2 in Kaposi's sarcoma cells. Oncotarget 2016;7:36614-21. https://doi.org/10.18632/oncotarget.9311
  24. Sessa R, Seano G, di Blasio L, et al. The miR-126 regulates angiopoietin-1 signaling and vessel maturation by targeting p85beta. Biochim Biophys Acta 2012;1823:1925-35. https://doi.org/10.1016/j.bbamcr.2012.07.011
  25. Gao J, Zhou XL, Kong RN, Ji LM, He LL, Zhao DB. microRNA -126 targeting PIK3R2 promotes rheumatoid arthritis synovial fibro-blasts proliferation and resistance to apoptosis by regulating PI3K/AKT pathway. Exp Mol Pathol 2016;100:192-8. https://doi.org/10.1016/j.yexmp.2015.12.015
  26. Rodrigues Alves APN, Fernandes JC, Fenerich BA, et al. IGF1R/IRS1 targeting has cytotoxic activity and inhibits PI3K/ AKT/mTOR and MAPK signaling in acute lymphoblastic leukemia cells. Cancer Lett 2019;456:59-68. https://doi.org/10.1016/j.canlet.2019.04.030
  27. Bai X, Chen XH, Liu YH, et al. Effects of water extract and crude polysaccharides from Liriope spicata var. prolifera on InsR/IRS-1/PI3K pathway and glucose metabolism in mice. J Ethnopharmacol 2009;125:482-6. https://doi.org/10.1016/j.jep.2009.07.007
  28. Sambandam V, Frederick MJ, Shen L, et al. PDK1 mediates NOTCH1-mutated head and neck squamous carcinoma vulnerability to therapeutic PI3K/mTOR inhibition. Clin Cancer Res 2019;25:3329-40. https://doi.org/10.1158/1078-0432.CCR-18-3276
  29. Wang B, Zhao CH, Sun G, et al. IL-17 induces the proliferation and migration of glioma cells through the activation of PI3K/Akt1/NF-kappa B-p65. Cancer Lett 2019;447:93-104. https://doi.org/10.1016/j.canlet.2019.01.008
  30. Dai L, Xu J, Liu S, et al. Characterization of miR-126-3p and its target talin2 in the bovine corpus luteum during the oestrus cycle. Reprod Domest Anim 2014;49:913-9. https://doi.org/10.1111/rda.12400
  31. Cui W, Li Q, Feng L, Ding W. MiR-126-3p regulates progesterone receptors and involves development and lactation of mouse mammary gland. Mol Cell Biochem 2011;355:17-25. https://doi.org/10.1007/s11010-011-0834-1
  32. Sirotkin AV, Laukova M, Ovcharenko D, Brenaut P, Mlyncek M. Identification of microRNAs controlling human ovarian cell proliferation and apoptosis. J Cell Physiol 2010;223:49-56. https://doi.org/10.1002/jcp.21999
  33. Sirotkin AV, Ovcharenko D, Grossmann R, Laukova M, Mlyncek M. Identification of MicroRNAs controlling human ovarian cell steroidogenesis via a genome-scale screen. J Cell Physiol 2009;219:415-20. https://doi.org/10.1002/jcp.21689
  34. Fan HY, Richards JS. Minireview: Physiological and pathological actions of RAS in the ovary. Mol Endocrinol 2010;24:286-98. https://doi.org/10.1210/me.2009-0251
  35. Zeleznik AJ, Saxena D, Little-Ihrig L. Protein kinase B is obligatory for follicle-stimulating hormone-induced granulosa cell differentiation. Endocrinology 2003;144:3985-94. https://doi.org/10.1210/en.2003-0293
  36. Gonzalez-Robayna IJ, Falender AE, Ochsner S, Firestone GL, Richards JS. Follicle-stimulating hormone (FSH) stimulates phosphorylation and activation of protein kinase B (PKB/Akt) and serum and glucocorticoid-induced kinase (Sgk): evidence for a kinaseindependent signaling by FSH in granulosa cells. Mol Endocrinol 2000;14:1283-300. https://doi.org/10.1210/mend.14.8.0500
  37. Zheng W, Nagaraju G, Liu Z, Liu K. Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary. Mol Cell Endocrinol 2012;356:24-30. https://doi.org/10.1016/j.mce.2011.05.027
  38. Reddy P, Adhikari D, Zheng W, et al. PDK1 signaling in oocytes controls reproductive aging and lifespan by manipulating the survival of primordial follicles. Hum Mol Genet 2009;18:2813-24. https://doi.org/10.1093/hmg/ddp217
  39. Brown C, LaRocca J, Pietruska J, et al. Subfertility caused by altered follicular development and oocyte growth in female mice lacking PKBalpha/Akt1. Biol Reprod 2010;82:246-56. https://doi.org/10.1095/biolreprod.109.077925
  40. Baker J, Hardy MP, Zhou J, et al. Effects of an Igf1 gene null mutation on mouse reproduction. Mol Endocrinol 1996;10:903-18. https://doi.org/10.1210/mend.10.7.8813730
  41. Bachelot A, Monget P, Imbert-Bollore P, et al. Growth hormone is required for ovarian follicular growth. Endocrinology 2002;143:4104-12. https://doi.org/10.1210/en.2002-220087
  42. Gareis NC, Huber E, Hein GJ, et al. Impaired insulin signaling pathways affect ovarian steroidogenesis in cows with COD. Anim Reprod Sci 2018;192:298-312. https://doi.org/10.1016/j.anireprosci.2018.03.031

피인용 문헌

  1. miR-126 Controls the Apoptosis and Proliferation of Immature Porcine Sertoli Cells by Targeting the PIK3R2 Gene through the PI3K/AKT Signaling Pathway vol.11, pp.8, 2020, https://doi.org/10.3390/ani11082260
  2. Genome-Wide Analysis of LncRNA in Bovine Mammary Epithelial Cell Injuries Induced by Escherichia Coli and Staphylococcus Aureus vol.22, pp.18, 2021, https://doi.org/10.3390/ijms22189719
  3. MicroRNAs: Potential biomarkers for reproduction, diagnosis, prognosis, and therapeutic in domestic animals vol.142, 2020, https://doi.org/10.1016/j.rvsc.2021.12.004