Korean J Physiol Pharmacol.  2019 May;23(3):171-179. 10.4196/kjpp.2019.23.3.171.

MicroRNA-186 targets SKP2 to induce p27(Kip1)-mediated pituitary tumor cell cycle deregulation and modulate cell proliferation

Affiliations
  • 1Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China. chenly11@163.com, tangjian@med.uestc.edu.cn

Abstract

Pituitary tumors are usually benign but can occasionally exhibit hormonal and proliferative behaviors. Dysregulation of the G1/S restriction point largely contributes to the over-proliferation of pituitary tumor cells. F-box protein S-phase kinase-interacting protein-2 (SKP2) reportedly targets and inhibits the expression of p27(Kip1), a well-known negative regulator of G1 cell cycle progression. In this study, SKP2 expression was found to be upregulated while p27(Kip1) expression was determined to be downregulated in rat and human pituitary tumor cells. Furthermore, SKP2 knockdown induced upregulation of p27(Kip1) and cell growth inhibition in rat and human pituitary tumor cells, while SKP2overexpression elicited opposite effects on p27(Kip1) expression and cell growth. The expression of microRNA-186 (miR-186) was reported to be reduced in pituitary tumors. Online tools predicted SKP2 to be a direct downstream target of miR-186, which was further confirmed by luciferase reporter gene assays. Moreover, miR-186 could modulate the cell proliferation and p27(Kip1)-mediated cell cycle alternation of rat and human pituitary tumor cells through SKP2. As further confirmation of these findings, miR-186 and p27(Kip1) expression were downregulated, while SKP2 expression was upregulated in human pituitary tumor tissue samples; thus, SKP2 expression negatively correlated with miR-186 and p27(Kip1) expression. In contrast, miR-186 expression positively associated with p27(Kip1) expression. Taken together, we discovered a novel mechanism by which miR-186/SKP2 axis modulates pituitary tumor cell proliferation through p27(Kip1)-mediated cell cycle alternation.

Keyword

Cell proliferation; Cyclin-dependent kinase inhibitor p27; microRNA-186; Pituitary neoplasms; S-phase kinase-interacting protein-2

MeSH Terms

Animals
Cell Cycle*
Cell Proliferation*
Cyclin-Dependent Kinase Inhibitor p27
Genes, Reporter
Humans
Luciferases
Pituitary Neoplasms*
Rats
Up-Regulation
Cyclin-Dependent Kinase Inhibitor p27
Luciferases

Figure

  • Fig. 1 The expression and combined function of SKP2 and p27Kip1 in pituitary tumor cell lines. (A, B) SKP2 expression in rat GH3 and human GH-secreting PA cells was regulated by transfection with si-SKP2 or SKP2 overexpressing vectors, as confirmed by immunoblot assays. Negative control small RNA interference (si-NC) or an empty vector (NC) served as a control. (C, D) Rat GH3 and human GH-secreting PA cells were transfected with si-SKP2 or SKP2 overexpressing vectors, and then DNA synthesis was examined using BrdU assays. (E, F) Cell proliferation was determined using CCK-8 assays. (G) Rat GH3 and human GH-secreting PA cells were cotransfected with si-SKP2 and si-p27Kip1, and then cell cycle analysis was performed using flow cytometer assays. The data are presented as the mean ± standard deviation of three independent experiments. *p < 0.05 and **p < 0.01, compared to the si-NC or SKP2 overexpression vector group. si, small interfering; NC, negative control; GH, growth hormone; PA, pituitary adenoma; BrdU, 5-bromo-2′-deoxyuridine; OD, optical density.

  • Fig. 2 SKP2 is a direct target of miR-186, which negatively regulates SKP2 expression. (A) Rat GH3 and human GH-secreting PA cells were transfected with miR-186 mimics or inhibitors to regulate miR-186 expression, as confirmed using real-time PCR. (B) SKP2 mRNA expression in response to miR-186 overexpression or inhibition in rat GH3 and human GH-secreting PA cells was examined using real-time PCR. (C) SKP2 protein levels under the same conditions were examined using immunoblot assays. (D) Luciferase reporter gene assays were performed using wt-SKP2 3′-UTR or mut-SKP2 3′-UTR vectors to verify the direct binding between miR-186 and SKP2 3′-UTR. The data are presented as mean ± standard deviation of three independent experiments. *p < 0.05, **p < 0.01. miR, microRNA; mRNA, messenger RNA; GH, growth hormone; PA, pituitary adenoma; PCR, polymerase chain reaction; UTR, untranslated region; NC, negative control.

  • Fig. 3 The miR-186 modulates pituitary tumor cell proliferation and the cell cycle through SKP2. (A) Rat GH3 and human GH-secreting PA cells were transfected with miR-186 mimics or inhibitors, and then, DNA synthesis was examined using BrdU assays. (B) Cell proliferation using CCK-8 assays. (C) Cell cycle analysis using flow cytometer assays. (D, E) Rat GH3 and human GH-secreting PA cells were cotransfected with miR-186 mimics and SKP2 overexpressing vectors, and then examined for the protein levels of SKP2 and p27Kip1 were examined using Immunoblot assays. (F) DNA synthesis was examined using BrdU assays. (G) Cell proliferation was examined using CCK-8 assays. The data are presented as the mean ± standard deviation of three independent experiments. *p < 0.05, **p < 0.01, compared to the control group; ##p < 0.01, compared to the SKP2 group. miR, microRNA; GH, growth hormone; PA, pituitary adenoma; BrdU, 5-bromo-2′-deoxyuridine; UTR, untranslated region; NC, negative control; OD, optical density.

  • Fig. 4 The expression of and correlations among miR-186, SKP2 and p27Kip1 in human tissue samples. (A–C) The mRNA expression of miR-186, SKP2 and p27kip1 in 5 normal human pituitary tissues and 20 human GH-producing pituitary tumor tissues was examined using real-time PCR. The data are presented as the mean ± standard deviation of three independent experiments. **p < 0.01. The protein levels of p27kip1 in normal pituitary tissues and pituitary tumor tissues were determined using immunoblot assays (D) and immunohistochemistry staining (E), respectively. The red arrows indicated p27kip1 positive cells. (F–H) The correlations among miR-186, SKP2 and p27Kip1 in tissue samples were analyzed using Spearman's rank correlation. mRNA, messenger RNA; GH, growth hormone; PCR, polymerase chain reaction.


Reference

1. Shimon I, Melmed S. Genetic basis of endocrine disease: pituitary tumor pathogenesis. J Clin Endocrinol Metab. 1997; 82:1675–1681.
2. Coire CI, Horvath E, Kovacs K, Smyth HS, Ezzat S. Cushing's syndrome from an ectopic pituitary adenoma with peliosis: a histological, immunohistochemical, and ultrastructural study and review of the literature. Endocr Pathol. 1997; 8:65–74.
Article
3. Asa SL, Ezzat S. The cytogenesis and pathogenesis of pituitary adenomas. Endocr Rev. 1998; 19:798–827.
Article
4. Palumbo T, Faucz FR, Azevedo M, Xekouki P, Iliopoulos D, Stratakis CA. Functional screen analysis reveals miR-26b and miR-128 as central regulators of pituitary somatomammotrophic tumor growth through activation of the PTEN-AKT pathway. Oncogene. 2013; 32:1651–1659.
Article
5. Tang FY, Shih CJ, Cheng LH, Ho HJ, Chen HJ. Lycopene inhibits growth of human colon cancer cells via suppression of the Akt signaling pathway. Mol Nutr Food Res. 2008; 52:646–654.
6. Li W, Zhang G, Wang HL, Wang L. Analysis of expression of cyclin E, p27kip1 and Ki67 protein in colorectal cancer tissues and its value for diagnosis, treatment and prognosis of disease. Eur Rev Med Pharmacol Sci. 2016; 20:4874–4879.
7. Berton S, Cusan M, Segatto I, Citron F, D'Andrea S, Benevol S, Avanzo M, Dall'Acqua A, Schiappacassi M, Bristow RG, Belletti B, Baldassarre G. Loss of p27kip1 increases genomic instability and induces radio-resistance in luminal breast cancer cells. Sci Rep. 2017; 7:595.
Article
8. Kim GJ, Kim DH, Min KW, Kim YH, Oh YH. Loss of p27kip1 expression is associated with poor prognosis in patients with taxane-treated breast cancer. Pathol Res Pract. 2018; 214:565–571.
9. Haddad NF, Teodoro AJ, Leite de Oliveira F, Soares N, de Mattos RM, Hecht F, Dezonne RS, Vairo L, Goldenberg RC, Gomes FC, de Carvalho DP, Gadelha MR, Nasciutti LE, Miranda-Alves L. Lycopene and beta-carotene induce growth inhibition and proapoptotic effects on ACTH-secreting pituitary adenoma cells. PLoS One. 2013; 8:e62773.
Article
10. Kulinski M, Achkar IW, Haris M, Dermime S, Mohammad RM, Uddin S. Dysregulated expression of SKP2 and its role in hematological malignancies. Leuk Lymphoma. 2018; 59:1051–1063.
Article
11. Ma Y, Yan M, Huang H, Zhang L, Wang Q, Zhao Y, Zhao J. Associations and prognostic significance of p27Kip1, Jab1 and Skp2 in non-Hodgkin lymphoma. Mol Clin Oncol. 2016; 5:357–364.
12. Borriello A, Naviglio S, Bencivenga D, Caldarelli I, Tramontano A, Speranza MC, Stampone E, Sapio L, Negri A, Oliva A, Sinisi AA, Spina A, Della Ragione F. Histone deacetylase inhibitors increase p27(Kip1) by affecting its ubiquitin-dependent degradation through Skp2 downregulation. Oxid Med Cell Longev. 2016; 2016:2481865.
13. Frescas D, Pagano M. Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP: tipping the scales of cancer. Nat Rev Cancer. 2008; 8:438–449.
14. Croce CM, Calin GA. miRNAs, cancer, and stem cell division. Cell. 2005; 122:6–7.
Article
15. Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, Ménard S, Palazzo JP, Rosenberg A, Musiani P, Volinia S, Nenci I, Calin GA, Querzoli P, Negrini M, Croce CM. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005; 65:7065–7070.
Article
16. Bandres E, Bitarte N, Arias F, Agorreta J, Fortes P, Agirre X, Zarate R, Diaz-Gonzalez JA, Ramirez N, Sola JJ, Jimenez P, Rodriguez J, Garcia-Foncillas J. microRNA-451 regulates macrophage migration inhibitory factor production and proliferation of gastrointestinal cancer cells. Clin Cancer Res. 2009; 15:2281–2290.
Article
17. Stilling G, Sun Z, Zhang S, Jin L, Righi A, Kovācs G, Korbonits M, Scheithauer BW, Kovacs K, Lloyd RV. MicroRNA expression in ACTH-producing pituitary tumors: up-regulation of microRNA-122 and -493 in pituitary carcinomas. Endocrine. 2010; 38:67–75.
Article
18. Bottoni A, Piccin D, Tagliati F, Luchin A, Zatelli MC, degli Uberti EC. miR-15a and miR-16-1 down-regulation in pituitary adenomas. J Cell Physiol. 2005; 204:280–285.
Article
19. Wierinckx A, Roche M, Legras-Lachuer C, Trouillas J, Raverot G, Lachuer J. MicroRNAs in pituitary tumors. Mol Cell Endocrinol. 2017; 456:51–61.
Article
20. Zhang QJ, Xu C. The role of microRNAs in the pathogenesis of pituitary tumors. Front Biosci (Landmark Ed). 2016; 21:1–7.
Article
21. Sapochnik M, Nieto LE, Fuertes M, Arzt E. Molecular mechanisms underlying pituitary pathogenesis. Biochem Genet. 2016; 54:107–119.
Article
22. Dweep H, Gretz N, Sticht C. miRWalk database for miRNA-target interactions. Methods Mol Biol. 2014; 1182:289–305.
Article
23. John B, Enright AJ, Aravin A, Tuschl T, Sander C, Marks DS. Human MicroRNA targets. PLoS Biol. 2004; 2:e363.
Article
24. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. Elife. 2015; 4:e05005.
Article
25. Molatore S, Marinoni I, Lee M, Pulz E, Ambrosio MR, degli Uberti EC, Zatelli MC, Pellegata NS. A novel germline CDKN1B mutation causing multiple endocrine tumors: clinical, genetic and functional characterization. Hum Mutat. 2010; 31:E1825–E1835.
Article
26. An J, Pei X, Zang Z, Zhou Z, Hu J, Zheng X, Zhang Y, He J, Duan L, Shen R, Zhang W, Zhu F, Li S, Yang H. Metformin inhibits proliferation and growth hormone secretion of GH3 pituitary adenoma cells. Oncotarget. 2017; 8:37538–37549.
Article
27. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995; 9:1149–1163.
Article
28. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999; 13:1501–1512.
Article
29. Muşat M, Vax VV, Borboli N, Gueorguiev M, Bonner S, Korbonits M, Grossman AB. Cell cycle dysregulation in pituitary oncogenesis. Front Horm Res. 2004; 32:34–62.
30. Craig C, Wersto R, Kim M, Ohri E, Li Z, Katayose D, Lee SJ, Trepel J, Cowan K, Seth P. A recombinant adenovirus expressing p27Kip1 induces cell cycle arrest and loss of cyclin-Cdk activity in human breast cancer cells. Oncogene. 1997; 14:2283–2289.
Article
31. Katayose Y, Kim M, Rakkar AN, Li Z, Cowan KH, Seth P. Promoting apoptosis: a novel activity associated with the cyclin-dependent kinase inhibitor p27. Cancer Res. 1997; 57:5441–5445.
32. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004; 116:281–297.
33. Liang HQ, Wang RJ, Diao CF, Li JW, Su JL, Zhang S. The PTTG1-targeting miRNAs miR-329, miR-300, miR-381, and miR-655 inhibit pituitary tumor cell tumorigenesis and are involved in a p53/PTTG1 regulation feedback loop. Oncotarget. 2015; 6:29413–29427.
Article
34. Renjie W, Haiqian L. MiR-132, miR-15a and miR-16 synergistically inhibit pituitary tumor cell proliferation, invasion and migration by targeting Sox5. Cancer Lett. 2015; 356:568–578.
Article
35. Cai J, Wu J, Zhang H, Fang L, Huang Y, Yang Y, Zhu X, Li R, Li M. miR-186 downregulation correlates with poor survival in lung adenocarcinoma, where it interferes with cell-cycle regulation. Cancer Res. 2013; 73:756–766.
Article
36. Yang J, Yuan D, Li J, Zheng S, Wang B. miR-186 downregulates protein phosphatase PPM1B in bladder cancer and mediates G1-S phase transition. Tumour Biol. 2016; 37:4331–4341.
Article
37. Hua X, Xiao Y, Pan W, Li M, Huang X, Liao Z, Xian Q, Yu L. miR-186 inhibits cell proliferation of prostate cancer by targeting GOLPH3. Am J Cancer Res. 2016; 6:1650–1660.
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr