Stocktake Sale on now: wide range of books at up to 70% off!
Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

miR-18b regulates the function of rabbit ovary granulosa cells

Ze Li A , Junyi Jiang A , Xiaohua Yi A , Guoyan Wang A , Shuhui Wang A and Xiuzhu Sun https://orcid.org/0000-0002-9760-0584 B C
+ Author Affiliations
- Author Affiliations

A College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China.

B College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, PR China.

C Corresponding author. Email: sunxiuzhu208@163.com

Reproduction, Fertility and Development 33(5) 363-371 https://doi.org/10.1071/RD20237
Submitted: 18 September 2020  Accepted: 17 January 2021   Published: 1 March 2021

Abstract

MicroRNAs (miRNAs) have been determined to participate in the process of oestradiol production. Generally, there are two pathways by which oestradiol levels change, one being the state of cells (i.e. the status of enzymes involved in the synthesis of hormones such as oestradiol) and the other being the number of cells that secrete oestradiol. It is known that oestrogens are the main steroids produced by granulosa cells (GCs) of mature ovarian follicles. In this study we explored the function of miR-18b in rabbit GCs by overexpressing or inhibiting its activity. We found that miR-18b silencing promoted the secretion of oestradiol by significantly affecting the expression of steroidogenesis-related genes. Thus, miR-18b may act as a negative regulator of the production of enzymes related to oestradiol synthesis and affect oestradiol production. Furthermore, the effects of miR-18b on the proliferation, cell cycle and apoptosis of GCs were investigated using a cell counting kit (CCK-8) proliferation assay, detection of annexin V–fluorescein isothiocyanate apoptosis, flow cytometry and quantitative polymerase chain reaction. The results showed that miR-18b upregulated GC apoptosis (miR-18b overexpression decreases cell growth and stimulates apoptosis). These findings suggest that miR-18b and the oestrogen receptor 1 (ESR1) gene may be attractive targets to further explore the molecular regulation of GCs. The miR-18b may also explain, in part, the abnormal folliculogenesis in mammals caused by conditions such as polycystic ovary syndrome, primary ovarian insufficiency, and others.

Graphical Abstract Image

Keywords: miR-18b, granulosa cell, oestradiol synthesis, female mammalian fertility, ESR1.


References

Abdalla, M. O., Yamamoto, T., Maehara, K., Nogami, J., Ohkawa, Y., Miura, H., Poonperm, R., Hiratani, I., Nakayama, H., Nakao, M., and Saitoh, N. (2019). The Eleanor ncRNAs activate the topological domain of the ESR1 locus to balance against apoptosis. Nat. Commun. 10, 3778–3794.
The Eleanor ncRNAs activate the topological domain of the ESR1 locus to balance against apoptosis.Crossref | GoogleScholarGoogle Scholar | 31439835PubMed |

Basu, R. K., Wong, H. R., Krawczeski, C. D., Wheeler, D. S., Manning, P. B., and Chawla, L. S. (2014). Combining functional and tubular damage biomarkers improves diagnostic precision for acute kidney injury after cardiac surgery. J. Am. Coll. Cardiol. 64, 2753–2762.
Combining functional and tubular damage biomarkers improves diagnostic precision for acute kidney injury after cardiac surgery.Crossref | GoogleScholarGoogle Scholar | 25541128PubMed |

Berisha, B., Pfaffl, M. W., and Schams, D. (2002). Expression of estrogen and progesterone receptors in the bovine ovary during estrous cycle and pregnancy. Endocrine 17, 207–214.
Expression of estrogen and progesterone receptors in the bovine ovary during estrous cycle and pregnancy.Crossref | GoogleScholarGoogle Scholar | 12108521PubMed |

Blaschka, C., Sánchez-Guijo, A., Zimmer, B., Stöhr, J., Kotarski, F., and Grothmann, H. (2019). Temporal expression pattern of steroid-metabolizing enzymes in bovine COC during in vitro maturation employing different gonadotropin concentrations. Theriogenology 131, 182–192.
Temporal expression pattern of steroid-metabolizing enzymes in bovine COC during in vitro maturation employing different gonadotropin concentrations.Crossref | GoogleScholarGoogle Scholar | 30981973PubMed |

Cadoret, V., Frapsauce, C., Jarrier, P., Maillard, V., and Monget, P. (2017). Molecular evidence that follicle development is accelerated in vitro compared to in vivo. Reproduction 153, 493–508.
Molecular evidence that follicle development is accelerated in vitro compared to in vivo.Crossref | GoogleScholarGoogle Scholar | 28154111PubMed |

Calin, G. A., Sevignani, C., Dumitru, C. D., Hyslop, T., Noch, E., and Yendamuri, S. (2004). Human microrna genes are frequently located at fragile sites and genomic regions involved in cancers. Proc. Natl. Acad. Sci. USA 101, 2999–3004.
Human microrna genes are frequently located at fragile sites and genomic regions involved in cancers.Crossref | GoogleScholarGoogle Scholar | 14973191PubMed |

Chen, B., Xu, P., Wang, J., and Zhang, C. (2019). The role of MiRNA in polycystic ovary syndrome (PCOS). Gene 706, 91–96.
The role of MiRNA in polycystic ovary syndrome (PCOS).Crossref | GoogleScholarGoogle Scholar | 31054362PubMed |

Chimento, A., Sirianni, R., Casaburi, I., Ruggiero, C., Maggiolini, M., Andò, S., and Pezzi, V. (2012). 17β-Estradiol activates GPER-and ESR1-dependent pathways inducing apoptosis in GC-2 cells, a mouse spermatocyte-derived cell line. Mol. Cell. Endocrinol. 355, 49–59.
17β-Estradiol activates GPER-and ESR1-dependent pathways inducing apoptosis in GC-2 cells, a mouse spermatocyte-derived cell line.Crossref | GoogleScholarGoogle Scholar | 22306083PubMed |

Cookson, V. J., Bentley, M. A., Hogan, B. V., Horgan, K., Hayward, B. E., Hazelwood, L. D., and Hughes, T. A. (2012). Circulating microRNA profiles reflect the presence of breast tumours but not the profiles of microRNA within the tumours. Cell Oncol (Dordr) 35, 301–308.
Circulating microRNA profiles reflect the presence of breast tumours but not the profiles of microRNA within the tumours.Crossref | GoogleScholarGoogle Scholar | 22821209PubMed |

Dar, A. A., Majid, S., Rittsteuer, C., De Semir, D., Bezrookove, V., and Tong, S. (2013). The role of mir-18b in mdm2-p53 pathway signaling and melanoma progression. J. Natl. Cancer Inst. 105, 433–442.
The role of mir-18b in mdm2-p53 pathway signaling and melanoma progression.Crossref | GoogleScholarGoogle Scholar | 23365201PubMed |

Eyster, K. M. (2016). The Estrogen Receptors: An Overview from Different Perspectives. Methods Mol. Biol. 1366, 1–10.
The Estrogen Receptors: An Overview from Different Perspectives.Crossref | GoogleScholarGoogle Scholar | 26585122PubMed |

Fonseca-Sanchéz, M. A., Pérez-Plasencia, C., Fernández-Retana, J., Arechaga-Ocampo, E., Marchat, L. A., Rodríguez-Cuevas, S., et al. (2013). Microrna-18b is upregulated in breast cancer and modulates genes involved in cell migration. Oncol. Rep. 30, 2399–2410.
Microrna-18b is upregulated in breast cancer and modulates genes involved in cell migration.Crossref | GoogleScholarGoogle Scholar | 23970382PubMed |

Fu, X., He, Y., Wang, X., Peng, D., Chen, X., Li, X., and Wan, Q. (2018). Microrna-16 promotes ovarian granulosa cell proliferation and suppresses apoptosis through targeting pdcd4 in polycystic ovarian syndrome. Cell. Physiol. Biochem. 48, 670–682.
Microrna-16 promotes ovarian granulosa cell proliferation and suppresses apoptosis through targeting pdcd4 in polycystic ovarian syndrome.Crossref | GoogleScholarGoogle Scholar | 30025387PubMed |

Gao, H., Xue, Y., Cao, L., Liu, Q., and Liu, C. (2017). ESR1 and its antagonist fulvestrant in pituitary adenomas. Mol. Cell. Endocrinol. 443, 32–41.
ESR1 and its antagonist fulvestrant in pituitary adenomas.Crossref | GoogleScholarGoogle Scholar | 28043824PubMed |

Giguère, V., Tremblay, A., and Tremblay, G. B. (1998). Estrogen receptor β: re-evaluation of estrogen and antiestrogen signaling. Steroids 63, 335–339.
Estrogen receptor β: re-evaluation of estrogen and antiestrogen signaling.Crossref | GoogleScholarGoogle Scholar | 9618797PubMed |

Gong, W., Song, J., Chen, X., Li, S., Yu, J., Xia, W., Ding, G., Zhang, Y., Jia, Z., Zhang, A., and Huang, S. (2019). Estrogen-Related Receptor α Mediates Puromycin Aminonucleoside-induced Mesangial Cell apoptosis and Inflammatory Injury. Am. J. Physiol. Renal Physiol. 316, F906–F913.
Estrogen-Related Receptor α Mediates Puromycin Aminonucleoside-induced Mesangial Cell apoptosis and Inflammatory Injury.Crossref | GoogleScholarGoogle Scholar | 30698047PubMed |

Guo, J., Miao, Y., Xiao, B., Huan, R., and Wang, Y. (2009). Differential expression of microrna species in human gastric cancer versus non-tumorous tissues. J. Gastroenterol. Hepatol. 24, 652–657.
Differential expression of microrna species in human gastric cancer versus non-tumorous tissues.Crossref | GoogleScholarGoogle Scholar | 19175831PubMed |

Hamilton, K. J., Arao, Y., and Korach, K. S. (2014). Estrogen hormone physiology: Reproductive findings from estrogen receptor mutant mice. Reprod. Biol. 14, 3–8.
Estrogen hormone physiology: Reproductive findings from estrogen receptor mutant mice.Crossref | GoogleScholarGoogle Scholar | 24607249PubMed |

Huang, T., Wang, Y. D., Xue, M. M., Feng, X., Sun, C. X., Wang, A. S., Xie, S. Y., Zhang, M., Sun, G. R., and Li, M. (2018). Transcriptome analysis of differentially expressed genes in rabbits’ ovaries by digital gene-expression profiling. Genes Genomics 40, 687–700.
Transcriptome analysis of differentially expressed genes in rabbits’ ovaries by digital gene-expression profiling.Crossref | GoogleScholarGoogle Scholar | 29934810PubMed |

Hughes, F. M., and Gorospe, W. C. (1991). Biochemical identification of apoptosis (programmed cell death) in granulosa cells: evidence for a potential mechanism underlying follicular atresia. Endocrinology 129, 2415–2422.
Biochemical identification of apoptosis (programmed cell death) in granulosa cells: evidence for a potential mechanism underlying follicular atresia.Crossref | GoogleScholarGoogle Scholar | 1935775PubMed |

Hutt, K. J., McLaughlin, E. A., and Holland, M. K. (2006). Primordial follicle activation and follicular development in the juvenile rabbit ovary. Cell Tissue Res. 326, 809–822.
Primordial follicle activation and follicular development in the juvenile rabbit ovary.Crossref | GoogleScholarGoogle Scholar | 16830146PubMed |

Leivonen, S.-K., Mäkelä, R., Östling, P., Kohonen, P., Haapa-Paananen, S., Haapa-Paananen, S., Kleivi, K., Enerly, E., Aakula, A., Hellström, K., Sahlberg, N., Kristensen, V. N., Børresen-Dale, A.-L., Saviranta, P., Perälä, M., and Kallioniemi, O. (2009). Protein lysate microarray analysis to identify micrornas regulating estrogen receptor signaling in breast cancer cell lines. Oncogene 28, 3926–3936.
Protein lysate microarray analysis to identify micrornas regulating estrogen receptor signaling in breast cancer cell lines.Crossref | GoogleScholarGoogle Scholar | 19684618PubMed |

Li, Y., Chen, M., Liu, J., Li, L., Yang, X., Zhao, J., Wu, M., and Ye, M. (2017). Upregulation of microrna 18b contributes to the development of colorectal cancer by inhibiting cdkn2b. Mol. Cell. Biol. 37, e00391-17.
Upregulation of microrna 18b contributes to the development of colorectal cancer by inhibiting cdkn2b.Crossref | GoogleScholarGoogle Scholar | 28784723PubMed |

Li, Q., Du, X., Pan, Z., Zhang, L., and Li, Q. (2018). The transcription factor smad4 and mir-10b contribute to e2 release and cell apoptosis in ovarian granulosa cells by targeting cyp19a1. Mol. Cell. Endocrinol. 476, 84–95.
The transcription factor smad4 and mir-10b contribute to e2 release and cell apoptosis in ovarian granulosa cells by targeting cyp19a1.Crossref | GoogleScholarGoogle Scholar | 29723543PubMed |

Lubahn, D. B., Moyer, J. S., Golding, T. S., Couse, J. F., Korach, K. S., and Smithies, O. (1993). Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene. Proc. Natl. Acad. Sci. USA 90, 11162–11166.
Alteration of reproductive function but not prenatal sexual development after insertional disruption of the mouse estrogen receptor gene.Crossref | GoogleScholarGoogle Scholar | 8248223PubMed |

Mansoori, B, Mohammadi, A, Gjerstorff, M F, Shirjang, S, Asadzadeh, Z, Khaze, V, Holmskov, U, Kazemi, T, Duijf, P, and Baradaran, B (2019). miR-142–3p is a tumor suppressor that inhibits estrogen receptor expression in ER-positive breast cancer. Journal of Cellular Physiology 234, 16043–16053.
miR-142–3p is a tumor suppressor that inhibits estrogen receptor expression in ER-positive breast cancer.Crossref | GoogleScholarGoogle Scholar |

Matsuda, F., Inoue, N., Manabe, N., and Ohkura, S. (2012). Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J. Reprod. Dev. 58, 44–50.
Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells.Crossref | GoogleScholarGoogle Scholar | 22450284PubMed |

Murakami, Y., Tamori, A., Itami, S., Tanahashi, T., Toyoda, H., Tanaka, M., Wu, W., Brojigin, N., Kaneoka, Y., Maeda, A., Kumada, T., Kawada, N., Kubo, S., and Kuroda, M. (2013). The expression level of mir-18b in hepatocellular carcinoma is associated with the grade of malignancy and prognosis. BMC Cancer 13, 99–110.
The expression level of mir-18b in hepatocellular carcinoma is associated with the grade of malignancy and prognosis.Crossref | GoogleScholarGoogle Scholar | 23496901PubMed |

Ogino, Y., Tohyama, S., Kohno, S., Toyota, K., Yamada, G., et al. (2018). Functional distinctions associated with the diversity of sex steroid hormone receptors ESR and AR. J. Steroid Biochem. Mol. Biol. 184, 38–46.
Functional distinctions associated with the diversity of sex steroid hormone receptors ESR and AR.Crossref | GoogleScholarGoogle Scholar | 29885351PubMed |

Ow, S. H., Chua, P. J., and Bay, B. H. (2018). miR-149 as a Potential Molecular Target for Cancer. Curr. Med. Chem. 25, 1046–1054.
miR-149 as a Potential Molecular Target for Cancer.Crossref | GoogleScholarGoogle Scholar | 28721828PubMed |

Pérez-Cremades, D., Mompeón, A., Vidal-Gómez, X., Hermenegildo, C., and Novella, S. (2018). Mirna as a new regulatory mechanism of estrogen vascular action. Int. J. Mol. Sci. 19, 473–488.
Mirna as a new regulatory mechanism of estrogen vascular action.Crossref | GoogleScholarGoogle Scholar |

Pu, M., Chen, J., Tao, Z., Miao, L., Qi, X., Wang, Y., and Ren, J. (2019). Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell. Mol. Life Sci. 76, 441–451.
Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression.Crossref | GoogleScholarGoogle Scholar | 30374521PubMed |

Richards, J. S., Fitzpatrick, S. L., Clemens, J. W., Morris, J. K., Alliston, T., and Sirois, J. (1995). Ovarian cell differentiation: a cascade of multiple hormones, cellular signals, and regulated genes. Recent Prog. Horm. Res. 50, 223–254.
| 7740159PubMed |

Roth, L. W., Mccallie, B., Alvero, R., Schoolcraft, W. B., Minjarez, D., and Katz-Jaffe, M. G. (2014). Altered microrna and gene expression in the follicular fluid of women with polycystic ovary syndrome. J. Assist. Reprod. Genet. 31, 355–362.
Altered microrna and gene expression in the follicular fluid of women with polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 24390626PubMed |

Rovani, M. T., Gasperin, B., Ilha, G., Ferreira, R., Bohrer, R., and Duggavathi, R. (2014). Expression and molecular consequences of inhibition of estrogen receptors in granulosa cells of bovine follicles. J. Ovarian Res. 7, 96–102.
Expression and molecular consequences of inhibition of estrogen receptors in granulosa cells of bovine follicles.Crossref | GoogleScholarGoogle Scholar | 25339519PubMed |

Suzuki, T. (2004). Estrogen-related receptor alpha in human breast carcinoma as a potent prognostic factor. Cancer Res. 64, 4670–4676.
Estrogen-related receptor alpha in human breast carcinoma as a potent prognostic factor.Crossref | GoogleScholarGoogle Scholar | 15231680PubMed |

Tecalco-Cruz, AC, Ramírez-Jarquín, JO, and Cruz-Ramos, E (2019). Estrogen receptor alpha and its ubiquitination in breast cancer cells. Current drug targets 20, 690–704.
Estrogen receptor alpha and its ubiquitination in breast cancer cells.Crossref | GoogleScholarGoogle Scholar | 30324876PubMed |

Wang, L., Li, C., Li, R., Deng, Y., Tan, Y., and Tong, C. (2016). MicroRNA-764–3p regulates 17β-estradiol synthesis of mouse ovarian granulosa cells by targeting steroidogenic factor-1. In Vitro Cell. Dev. Biol. Anim. 52, 365–373.
MicroRNA-764–3p regulates 17β-estradiol synthesis of mouse ovarian granulosa cells by targeting steroidogenic factor-1.Crossref | GoogleScholarGoogle Scholar | 26676955PubMed |

Wang, S., Wang, X., Weng, Z., Zhang, S., Ning, H., and Li, B. (2017). Expression and role of microRNA 18b and hypoxia inducible factor-1α in placental tissues of preeclampsia patients. Exp. Ther. Med. 14, 4554–4560.
Expression and role of microRNA 18b and hypoxia inducible factor-1α in placental tissues of preeclampsia patients.Crossref | GoogleScholarGoogle Scholar | 29104664PubMed |

Wei, Z., Qiao, S., Zhao, J., Liu, Y., Li, Q., Wei, Z., Dai, Q., Kang, L., and Xu, B. (2019). miRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced inflammatory response after myocardial ischemia-reperfusion injury. Life Sci. 232, 116632.
miRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced inflammatory response after myocardial ischemia-reperfusion injury.Crossref | GoogleScholarGoogle Scholar | 31278944PubMed |

Worku, T., Rehman, Z. U., Talpur, H. S., Bhattarai, D., Ullah, F., Malobi, N., Kebede, T., and Yang, L. (2017). Micrornas: new insight in modulating follicular atresia: a review. Int. J. Mol. Sci. 18, 333–346.
Micrornas: new insight in modulating follicular atresia: a review.Crossref | GoogleScholarGoogle Scholar |

Wu, G., Song, D., Wei, Q., Xing, J., and Shi, X. (2018). Melatonin mitigates bisphenol A-induced estradiol production and proliferation by porcine ovarian granulosa cells in vitro. Anim. Reprod. Sci. 192, 91–98.
Melatonin mitigates bisphenol A-induced estradiol production and proliferation by porcine ovarian granulosa cells in vitro.Crossref | GoogleScholarGoogle Scholar | 29523361PubMed |

Yeung, C. K., Wang, G., Yao, Y., Liang, J., and Yang, X. (2017). Bre modulates granulosa cell death to affect ovarian follicle development and atresia in the mouse. Cell Death Dis. 8, e2697.
Bre modulates granulosa cell death to affect ovarian follicle development and atresia in the mouse.Crossref | GoogleScholarGoogle Scholar | 28333135PubMed |

Yin, Y., Song, M., Gu, B., Qi, X., and Huang, Z. (2015). Systematic analysis of key mirnas and related signaling pathways in colorectal tumorigenesis. Gene 578, 177–184.
Systematic analysis of key mirnas and related signaling pathways in colorectal tumorigenesis.Crossref | GoogleScholarGoogle Scholar | 26692142PubMed |

Yoshimoto, N., Toyama, T., Takahashi, S., Sugiura, H., Endo, Y., and Iwasa, M. (2011). Distinct expressions of micrornas that directly target estrogen receptor α in human breast cancer. Breast Cancer Res. Treat. 130, 331–339.
Distinct expressions of micrornas that directly target estrogen receptor α in human breast cancer.Crossref | GoogleScholarGoogle Scholar | 21755340PubMed |

Yu, Y. S., Sui, H. S., Han, Z. B., Li, W., Luo, M. J., and Tan, J. H. (2004). Apoptosis in granulosa cells during follicular atresia: relationship with steroids and insulin-like growth factors. Cell Res. 14, 341–346.
Apoptosis in granulosa cells during follicular atresia: relationship with steroids and insulin-like growth factors.Crossref | GoogleScholarGoogle Scholar | 15353131PubMed |

Yue, W., Berstein, L. M., Wang, J. P., Clark, G. M., and Santen, R. J. (2001). The potential role of estrogen in aromatase regulation in the breast. J. Steroid Biochem. Mol. Biol. 79, 157–164.
The potential role of estrogen in aromatase regulation in the breast.Crossref | GoogleScholarGoogle Scholar | 11850220PubMed |

Zhang, H., Luo, Q., Lu, X., Yin, N., Zhou, D., Zhang, L., Zhao, W., Wang, D., Du, P., Hou, Y., Zhang, Y., and Yuan, W. (2018). Effects of hPMSCs on granulosa cell apoptosis and AMH expression and their role in the restoration of ovary function in premature ovarian failure mice. Stem Cell Res. Ther. 9, 20–32.
Effects of hPMSCs on granulosa cell apoptosis and AMH expression and their role in the restoration of ovary function in premature ovarian failure mice.Crossref | GoogleScholarGoogle Scholar | 29386068PubMed |

Zhang, J., Xu, Y., Liu, H., and Pan, Z. (2019). MicroRNAs in ovarian follicular atresia and granulosa cell apoptosis. Reprod. Biol. Endocrinol. 17, 9–20.
MicroRNAs in ovarian follicular atresia and granulosa cell apoptosis.Crossref | GoogleScholarGoogle Scholar | 30630485PubMed |