Yonsei Med J.  2019 Aug;60(8):751-759. 10.3349/ymj.2019.60.8.751.

Silencing of LncRNA-ANCR Promotes the Osteogenesis of Osteoblast Cells in Postmenopausal Osteoporosis via Targeting EZH2 and RUNX2

Affiliations
  • 1Department of Respiratory, Qingdao Eighth People's Hospital, Qingdao, Shandong, China.
  • 2Department of Orthopedic Surgery, Qingdao University, Qingdao, Shandong, China.
  • 3Department of Tramotology and Orthopedics, Pingyi Hospital of Traditional Chinese Medicine, Linyi, Shandong, China.
  • 4Department of Sports Medicine, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China. wangfuke25@163.com

Abstract

PURPOSE
This study aimed to explore the effects and mechanisms of long non-coding RNA (lncRNA) anti-differentiation non-coding RNA (ANCR) on the osteogenesis of osteoblast cells in postmenopausal osteoporosis (PMOP).
MATERIALS AND METHODS
Mice models of PMOP were established. ANCR expression and intracellular calcium ions were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and laser confocal microscopy, respectively. ANCR was silenced in osteoblast cells from PMOP mice by the transfection of siRNA-ANCR (si-ANCR). The proliferation and apoptosis of osteoblast cells was analyzed by MTT and flow cytometry, respectively. Alkaline phosphatase (ALP) activity and calcium nodules were examined by ALP and alizarin red staining assay, respectively. The expression of enhancer of zeste homolog 2 (EZH2), runt related transcription factor 2 (RUNX2), and OSTERIX was detected by qRT-PCR and Western blot. Furthermore, an osteogenesis model was constructed in mice, and osteoid formation was observed by hematoxylin-eosin (HE) staining. The interaction between lncRNA-ANCR and EZH2 was further identified by RNA pull-down assay.
RESULTS
ANCR expression and intracellular calcium ions were increased in PMOP mice. Si-ANCR significantly increased the proliferation, ALP activity, calcium deposition of osteoblast cells and decreased apoptosis. ANCR and EZH2 were down-regulated by si-ANCR, while RUNX2 and OSTERIX were upregulated. Si-ANCR also promoted osteoid formation in mice treated with hydroxyapatite-tricalcium phosphate. In addition, ANCR specifically bound to EZH2.
CONCLUSION
Silencing ANCR promotes the osteogenesis of PMOP osteoblast cells. The specific binding of ANCR with EZH2 suppressed RUNX2, thereby inhibiting osteogenesis.

Keyword

Postmenopausal osteoporosis; osteoblast; lncRNA-ANCR; siRNA; EZH2; RUNX2

MeSH Terms

Alkaline Phosphatase
Animals
Apoptosis
Blotting, Western
Calcium
Female
Flow Cytometry
Humans
Ions
Mice
Microscopy, Confocal
Osteoblasts*
Osteogenesis*
Osteoporosis, Postmenopausal*
Real-Time Polymerase Chain Reaction
RNA
RNA, Long Noncoding
RNA, Small Interfering
RNA, Untranslated
Transcription Factors
Transfection
Alkaline Phosphatase
Calcium
Ions
RNA
RNA, Long Noncoding
RNA, Small Interfering
RNA, Untranslated
Transcription Factors

Figure

  • Fig. 1 Long non-coding RNA anti-differentiation noncoding RNA (lncRNA-ANCR) expression analysis based on a mouse postmenopausal osteoporosis (PMOP) model. The PMOP model was constructed using three groups: osteoporosis group, control group, and sham group (n=15 each group). (A) Bone mineral density comparison using microcomputed tomography. (B) Laser confocal microscopy detection for calcium level (×100, Fluo8 fluorescent probes are labeled). (C) Quantitative real-time polymerase chain reaction (qRT-PCR) analysis for lncRNA-ANCR mRNA and protein expression in osteoblast cells of the PMOP model. X-axis in C represents lncRNA-ANCR relative mRNA expression levels; Y-axis presents different groups in the PMOP model. *p<0.001 vs. Control group and Sham group.

  • Fig. 2 mRNA expression of long non-coding RNA anti-differentiation noncoding RNA (lncRNA-ANCR) in osteoblast cells transfected with siRNA-ANCR (si-ANCR). X-axis presents relative lncRNA-ANCR expression; Y-axis presents the three groups in postmenopausal osteoporosis model. *p<0.001 vs. NC-ANCR (negative control group, cell line was transfected with lncRNA-ANCR negative control) group and BLANK group (blank control group, cells without treatment).

  • Fig. 3 Effects of long non-coding RNA anti-differentiation noncoding RNA (lncRNA-ANCR) transfected with siRNA-ANCR (si-ANCR). (A) The effect of lncRNA-ANCR on proliferation evaluated by MTT assay. (B) The effect of lncRNA-ANCR on apoptosis determined by flow cytometry. (C) The effect of lncRNA-ANCR on alkaline phosphatase (ALP) activity detected by ALP staining. X-axis represents the relative lncRNA-ANCR expression; Y-axis presents the three groups in the postmenopausal osteoporosis model. (D) The effect of lncRNA-ANCR on calcium deposition upon alizarin red staining. *p<0.05, †p<0.01, ‡p<0.001 vs. NC-ANCR group (negative control group, cell line was transfected with lncRNA-ANCR negative control) and BLANK group (blank control group, cells without treatment). PI, propidium iodide; FITC, fluoresceine isothiocyanate; OD, optical density.

  • Fig. 4 In vivo experiments show the regulatory effect of long non-coding RNA anti-differentiation noncoding RNA (lncRNA-ANCR) on osteoid formation. (A) Hematoxylin-eosin (HE) staining of osteoid formation in mice injected with siRNA-ANCR (si-ANCR)-transfected osteoblast cells (bar=200 µm). Black and gray arrows indicate hydroxyapatite-tricalcium phosphate and osteoblast cells, respectively. (B) Osteoid/Total area (%). *p<0.01 vs. NC-ANCR group (negative control group, cell line was transfected with lncRNA-ANCR negative control) and BLANK group (blank control group, cells without treatment).

  • Fig. 5 Expression of enhancer of zeste homolog 2 (EZH2), runt related transcription factor 2 (RUNX2), and OSTERIX in osteoblast cells transfected with siRNA-anti-differentiation noncoding RNA (si-ANCR). (A) Quantitative real-time polymerase chain reaction (qRT-PCR) results for the mRNA and protein expression of EZH2, Runx2, and Osterix. (B and C) Western blot results for mRNA and protein expression of EZH2, RUNX2, and OSTERIX. *p<0.001 vs. NC-ANCR group (negative control group, cell line was transfected with lncRNA-ANCR negative control) and BLANK group (blank control group, cells without treatment). GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

  • Fig. 6 The interaction between long non-coding RNA anti-differentiation noncoding RNA (lncRNA-ANCR) and enhancer of zeste homolog 2 (EZH2). (A) Expression of EZH2 in ANCR and ANCR-Mut detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. (B) The expression of ANCR in EZH2 and EZH2-control detected by qRT-PCR and Western blot. *p<0.001, lncRNA-ANCR group vs. lncRNA-ANCR-Mut group, EZH2 group vs. EZH2-control group.


Reference

1. Black DM, Rosen CJ. Postmenopausal osteoporosis. N Engl J Med. 2016; 374:2096–2097.
Article
2. Svedbom A, Hernlund E, Ivergård M, Compston J, Cooper C, Stenmark J, et al. Osteoporosis in the European Union: a compendium of country-specific reports. Arch Osteoporos. 2013; 8:137.
Article
3. Wade SW, Strader C, Fitzpatrick LA, Anthony MS, O'Malley CD. Estimating prevalence of osteoporosis: examples from industrialized countries. Arch Osteoporos. 2014; 9:182.
Article
4. Gennari L, Rotatori S, Bianciardi S, Nuti R, Merlotti D. Treatment needs and current options for postmenopausal osteoporosis. Expert Opin Pharmacother. 2016; 17:1141–1152.
Article
5. Alami S, Hervouet L, Poiraudeau S, Briot K, Roux C. Barriers to effective postmenopausal osteoporosis treatment: a qualitative study of patients' and practitioners' views. Plos One. 2016; 11:e0158365.
Article
6. Chang CY, Tang CH, Chen KC, Huang KC, Huang KC. The mortality and direct medical costs of osteoporotic fractures among postmenopausal women in Taiwan. Osteoporos Int. 2016; 27:665–676.
Article
7. Baum R, Gravallese EM. Bone as a target organ in rheumatic disease: impact on osteoclasts and osteoblasts. Clin Rev Allergy Immunol. 2016; 51:1–15.
Article
8. Shen F, Ruddy MJ, Plamondon P, Gaffen SL. Cytokines link osteoblasts and inflammation: microarray analysis of interleukin-17-and TNF-α-induced genes in bone cells. J Leukoc Biol. 2005; 77:388–399.
Article
9. Marie PJ, Kassem M. Osteoblasts in osteoporosis: past, emerging, and future anabolic targets. Eur J Endocrinol. 2011; 165:1–10.
Article
10. Trošt Z, Trebše R, Preželj J, Komadina R, Logar DB, Marc J. A microarray based identification of osteoporosis-related genes in primary culture of human osteoblasts. Bone. 2010; 46:72–80.
Article
11. Kretz M, Webster DE, Flockhart RJ, Lee CS, Zehnder A, Lopez-Pajares V, et al. Suppression of progenitor differentiation requires the long noncoding RNA ANCR. Genes Dev. 2012; 26:338–343.
Article
12. Jia Q, Jiang W, Ni L. Down-regulated non-coding RNA (lncRNA-ANCR) promotes osteogenic differentiation of periodontal ligament stem cells. Arch Oral Biol. 2015; 60:234–241.
Article
13. Zhu L, Xu PC. Downregulated LncRNA-ANCR promotes osteoblast differentiation by targeting EZH2 and regulating Runx2 expression. Biochem Biophys Res Commun. 2013; 432:612–617.
Article
14. Yang ZY, Yang F, Zhang YL, Liu B, Wang M, Hong X, et al. LncRNA-ANCR down-regulation suppresses invasion and migration of colorectal cancer cells by regulating EZH2 expression. Cancer Biomark. 2017; 18:95–104.
Article
15. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001; 25:402–408.
Article
16. Harada S, Rodan GA. Control of osteoblast function and regulation of bone mass. Nature. 2003; 423:349–355.
Article
17. Ali AA, Weinstein RS, Stewart SA, Parfitt AM, Manolagas SC, Jilka RL. Rosiglitazone causes bone loss in mice by suppressing osteoblast differentiation and bone formation. Endocrinology. 2005; 146:1226–1235.
Article
18. Logothetis CJ, Lin SH. Osteoblasts in prostate cancer metastasis to bone. Nat Rev Cancer. 2005; 5:21–28.
Article
19. Giuliani N, Rizzoli V, Roodman GD. Multiple myeloma bone disease: pathophysiology of osteoblast inhibition. Blood. 2006; 108:3992–3996.
Article
20. Xiao X, Zhou T, Guo S, Guo C, Zhang Q, Dong N, et al. LncRNA MALAT1 sponges miR-204 to promote osteoblast differentiation of human aortic valve interstitial cells through up-regulating Smad4. Int J Cardiol. 2017; 243:404–412.
Article
21. McManus MT, Sharp PA. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet. 2002; 3:737–747.
Article
22. Li G, Zou L, Xie W, Wen S, Xie Q, Gao Y, et al. The effects of NONRATT021972 lncRNA siRNA on PC12 neuronal injury mediated by P2X 7 receptor after exposure to oxygen-glucose deprivation. Purinergic Signal. 2016; 12:479–487.
Article
23. Shao Z, Zhang W, Fu R, Li L, Wang H. TET2 gene expression in bone marrow cells in myelodysplastic syndromes patients and the effect of silencing TET2 by siRNA on biological characteristics of healthy CD34+ cells. Blood. 2012; 120:4934.
Article
24. Tan JZ, Yan Y, Wang XX, Jiang Y, Xu HE. EZH2: biology, disease, and structure-based drug discovery. Acta Pharmacol Sin. 2014; 35:161–174.
Article
25. Ding X, Wang X, Sontag S, Qin J, Wanek P, Lin Q, et al. The polycomb protein Ezh2 impacts on induced pluripotent stem cell generation. Stem Cells Dev. 2014; 23:931–940.
Article
26. Liu L, Luo Q, Sun J, Ju Y, Morita Y, Song G. Chromatin organization regulated by EZH2-mediated H3K27me3 is required for OPN-induced migration of bone marrow-derived mesenchymal stem cells. Int J Biochem Cell Biol. 2018; 96:29–39.
Article
27. Chen YH, Chung CC, Liu YC, Yeh SP, Hsu JL, Hung MC, et al. Enhancer of zeste homolog 2 and histone deacetylase 9c regulate age-dependent mesenchymal stem cell differentiation into osteoblasts and adipocytes. Stem Cells. 2016; 34:2183–2193.
Article
28. Dudakovic A, Camilleri ET, Paradise CR, Samsonraj RM, Gluscevic M, Paggi CA, et al. Enhancer of zeste homolog 2 (Ezh2) controls bone formation and cell cycle progression during osteogenesis in mice. J Biol Chem. 2018; 293:12894–12907.
Article
29. Wang D, Ding L, Wang L, Zhao Y, Sun Z, Karnes RJ, et al. LncRNA MALAT1 enhances oncogenic activities of EZH2 in castration-resistant prostate cancer. Oncotarget. 2015; 6:41045–41055.
Article
30. Li Z, Hou P, Fan D, Dong M, Ma M, Li H, et al. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer. Cell Death Differ. 2017; 24:59–71.
Article
31. Zhang F, Peng H. LncRNA-ANCR regulates the cell growth of osteosarcoma by interacting with EZH2 and affecting the expression of p21 and p27. J Orthop Surg Res. 2017; 12:103.
Article
32. Lucero CM, Vega OA, Osorio MM, Tapia JC, Antonelli M, Stein GS, et al. The cancer-related transcription factor Runx2 modulates cell proliferation in human osteosarcoma cell lines. J Cell Physiol. 2013; 228:714–723.
Article
33. Fujita T, Azuma Y, Fukuyama R, Hattori Y, Yoshida C, Koida M, et al. Runx2 induces osteoblast and chondrocyte differentiation and enhances their migration by coupling with PI3K-Akt signaling. J Cell Biol. 2004; 166:85–95.
Article
34. Yu C, Li L, Xie F, Guo S, Liu F, Dong N, et al. LncRNA TUG1 sponges miR-204-5p to promote osteoblast differentiation through upregulating Runx2 in aortic valve calcification. Cardiovasc Res. 2018; 114:168–179.
Article
Full Text Links
  • YMJ
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