Effects of rutin on osteoblast MC3T3-E1 differentiation, ALP activity and Runx2 protein expression

Submitted: 1 November 2020
Accepted: 17 December 2020
Published: 20 January 2021
Abstract Views: 1624
PDF: 770
HTML: 24
Publisher's note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

Authors

As a flavonoid, rutin has been found to have a wide range of biological functions, such as resisting inflammation and oxidation, and preventing cerebral hemorrhage and hypertension. It has been found to play an important role in osteoporosis and other orthopedic diseases in recent years. MC3T3-E1 cells were randomly divided into a control group, a rutin-1 group (0.01 mmol/L), a rutin-2 group (0.05 mmol/L) and a rutin-3 group (0.1 mmol/L). Osteogenic differentiation of cells was induced by osteogenic induction fluid. The control group was treated with the maximum dose of drug solvent. 2~3 days later, the solvent was replaced with fresh osteogenic induction fluid containing rutin. After a certain period of routine culture, the cells were collected for subsequent experiments. The expression of Runx2 gene in cells in all groups was detected by Real-time PCR; the expression of Runx2 protein was detected by Western blot and immunocytochemistry; the activity of ALP was detected by reagent kit method; osteogenic differentiation was analyzed by alizarin red staining. The results of Real-time PCR showed that, compared with the control group, the treatment of cells with rutin can significantly increase the expression of Runx2 gene (p<0.05); the higher the concentration, the higher the expression of Runx2 gene, and significant differences were found among groups in which different concentrations were used (p<0.05); the results of Western blot and IHC showed that the expression trend of Runx2 protein in each group was consistent with PCR results. In drug treatment groups, the activity of ALP was significantly higher than that in the control group (p<0.05); there were significant differences among groups in which different concentrations were used (p<0.05). The results of alizarin red staining showed that calcified nodules were formed in all groups and that the area of calcified nodules formed in groups treated with rutin was greater than that in the control group; the greater the concentration, the larger the area. Rutin can promote osteoblastic differentiation; and the greater the concentration, the more effective it is.

Dimensions

Altmetric

PlumX Metrics

Downloads

Download data is not yet available.

Citations

Solomon H. Rutin as a natural therapy for Alzheimer’s disease: Insights into its mechanisms of action. Curr Med Chem 2016;23:860-73. DOI: https://doi.org/10.2174/0929867323666160217124333
Solomon H, Abebech B. Natural therapies of the inflammatory bowel disease: The case of rutin and its aglycone, quercetin. Mini Rev Med Chem 2018;18:234-43.
Lee S C.A review on plant-based rutin extraction methods and its pharmacological activities. J Ethnopharmacol 2013;150:805-17. DOI: https://doi.org/10.1016/j.jep.2013.10.036
Fushimi S, Nohno T, Nagatsuka H, Katsuyama H. Involvement of miR-140-3p in Wnt3a and TGFβ3 signaling pathways during osteoblast differentiation in MC3T3-E1 cells. Genes Cells 2018;23:517-27. DOI: https://doi.org/10.1111/gtc.12591
Buo AM, Tomlinson RE, Eidelman ER, Chason M, Stains JP. Connexin43 and Runx2 interact to affect cortical bone geometry, skeletal development, and osteoblast and osteoclast function. J Bone Miner Res 2017;32:1727-38. DOI: https://doi.org/10.1002/jbmr.3152
Toshihisa K. Regulation of bone development and maintenance by Runx2. Front Biosci 2008;13:898-903. DOI: https://doi.org/10.2741/2730
Luca DC, Giulio I, Maria TV. Transcription factor Runx2 and its application to bone tissue engineering. Stem Cell Rev Rep 2012;8:891-7. DOI: https://doi.org/10.1007/s12015-011-9337-4
Kyung TW, Lee JE, Shin HH, Choi HS. Rutin inhibits osteoclast formation by decreasing reactive oxygen species and TNF-alpha by inhibiting activation of NF-kappaB. Exp Mol Med 2008;40:52-8. DOI: https://doi.org/10.3858/emm.2008.40.1.52
Toshihisa K. Roles of Runx2 in skeletal development. Adv Exp Med Biol 2017;962:83-93. DOI: https://doi.org/10.1007/978-981-10-3233-2_6
Birmingham E, Niebur GL, McHugh PE, Shaw G, Barry FP, McNamara LM. Osteogenic differentiation of mesenchymal stem cells is regulated by osteocyte and osteoblast cells in a simplified bone niche. Eur Cell Mater 2012;23:13-27. DOI: https://doi.org/10.22203/eCM.v023a02
Zhang L, Gong Z X. Clinical characteristics and prognostic factors in bone metastases from lung cancer. Med Sci Monit 2017;23:4087-94. DOI: https://doi.org/10.12659/MSM.902971
Ravi GS, Charyulu RN, Dubey A, Prabhu P, Hebbar S, Mathias AC. Nano-lipid complex of rutin: Development, characterisation and in vivo investigation of hepatoprotective, antioxidant activity and bioavailability study in rats. AAPS PharmSciTech 2018;19:3631-49. DOI: https://doi.org/10.1208/s12249-018-1195-9
Ghorbani A. Mechanisms of antidiabetic effects of flavonoid rutin. Biomed Pharmacother 2017;96:305-312. DOI: https://doi.org/10.1016/j.biopha.2017.10.001
Habtemariam S, Lentini G. The therapeutic potential of rutin for diabetes: an update. Mini Rev Med Chem 2015;15:524-8. DOI: https://doi.org/10.2174/138955751507150424103721
Abdel-Naim AB, Alghamdi AA, Algandaby MM, Al-Abbasi FA, Al-Abd AM, Eid BG, et al. Rutin isolated from chrozophora tinctoria enhances bone cell proliferation and ossification markers. Oxid Med Cell Longev 2018;2018:5106469. DOI: https://doi.org/10.1155/2018/5106469
Xiao Y, Wei R, Yuan Z, Lan X, Kuang J, Hu D, et al. Rutin suppresses FNDC1 expression in bone marrow mesenchymal stem cells to inhibit postmenopausal osteoporosis. Am J Transl Res 2019;11:6680-90.
Zhao B, Zhang W, Xiong Y, Zhang Y, Jia L, Xu X. Rutin protects human periodontal ligament stem cells from TNF-α induced damage to osteogenic differentiation through suppressing mTOR signaling pathway in inflammatory environment. Arch Oral Biol 2020;109:104584. DOI: https://doi.org/10.1016/j.archoralbio.2019.104584
Toshihisa K. Regulation of osteoblast differentiation by transcription factors. J Cell Biochem 2006;99:1233-9. DOI: https://doi.org/10.1002/jcb.20958
Ducy P, Starbuck M, Priemel M, Shen J, Pinero G, Geoffroy V, et al. A Cbfa1-dependent genetic pathway controls bone formation beyond embryonic development. Genes Dev 1999;13:1025-36. DOI: https://doi.org/10.1101/gad.13.8.1025
Toshihisa K. Regulation of proliferation, differentiation and functions of osteoblasts by Runx2. Int J Mol Sci 2019;20:1694. DOI: https://doi.org/10.3390/ijms20071694
Gowda PS, Wildman BJ, Trotter TN, Xu X, Hao X, Hassan MQ, et al. Runx2 suppression by miR-342 and miR-363 inhibits multiple myeloma progression. Mol Cancer Res 2018;16:1138-48. DOI: https://doi.org/10.1158/1541-7786.MCR-17-0606
Zhang J, Zhang W, Dai J, Wang X, Shen SG. Overexpression of Dlx2 enhances osteogenic differentiation of BMSCs and MC3T3-E1 cells via direct upregulation of osteocalcin and ALP. Int J Oral Sci 2019;11:12. DOI: https://doi.org/10.1038/s41368-019-0046-1
Liu L, Wang D, Qin Y, Xu M, Zhou L, Xu W, et al. Astragalin promotes osteoblastic differentiation in MC3T3-E1 cells and bone formation in vivo. Front Endocrinol (Lausanne) 2019;10:228. DOI: https://doi.org/10.3389/fendo.2019.00228

How to Cite

Liu, X.-W. ., Ma, B. ., Zi, Y. ., Xiang, L.-B. ., & Han, T.-Y. (2021). Effects of rutin on osteoblast MC3T3-E1 differentiation, ALP activity and Runx2 protein expression. European Journal of Histochemistry, 65(1). https://doi.org/10.4081/ejh.2021.3195