AccScience Publishing / IJB / Volume 8 / Issue 2 / DOI: 10.18063/ijb.v8i2.544
RESEARCH ARTICLE

3D-Printed β-Tricalcium Phosphate Scaffolds Promote Osteogenic Differentiation of Bone Marrow-Deprived Mesenchymal Stem Cells in an N6-methyladenosineDependent Manner

Xin Jiao1† Xin Sun1† Wentao Li1† Wenxiang Chu2 Yuxin Zhang3 Yiming Li1 Zengguang Wang1 Xianhao Zhou1 Jie Ma1 Chen Xu1 Kerong Dai1 Jinwu Wang1* Yaokai Gan1*
Show Less
1 Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, No. 639 Zhi Zao Ju Road, Huangpu District, Shanghai, China
2 Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Huangpu District, Shanghai, China
3 Department of Rehabilitation Medicine, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhi Zao Ju Road, Huangpu District, Shanghai, China
Submitted: 30 November 2021 | Accepted: 5 February 2022 | Published: 22 March 2022
© 2022 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Bone defect is a serious orthopedic disease which has been studied for a long time. Alternative degradable biomaterials are required for bone repairing and regeneration to address the limitation of autogenous bone. β-tricalcium phosphate (β-TCP) is an alternative material with good cytocompatibility and has been used in bone defect treatment. However, whether β-TCP contributes to osteogenesis of bone marrow stem cells (BMSCs) through N6-methyladenosine (m6A) modification remains unknown. To address this issue, we verified the effects of β-TCP on osteogenesis of BMSCs. We also studied the expression of m6A-related enzymes in BMSCs after β-TCP treatment. Furthermore, the m6A level and stability of Runt-related transcription factor 2 (RUNX2) mRNA were investigated after β-TCP treatment. Finally, rat calvarial defect models were performed to detect expression level of osteogenic factors and m6A-related enzymes after the stimulation of three-dimension (3D)-printed β-TCP scaffolds. We found that β-TCP showed good biocompatibility and was osteoinductive. Meanwhile, methyltransferase-like 3 (METTL3) increased, causing the elevation of m6A level of RUNX2, results in stabler RUNX2 mRNA level. At last, based on the animal experiments, we demonstrated that the increase of RUNX2 and METTL3 levels was induced by β-TCP. These findings suggest that METTL3 increases the m6A level of RUNX2 mRNA after β-TCP induction, contributing to its stability, and the results in vivo also confirmed the osteogenic and bone-repair properties of β-TCP.

Keywords
β-tricalcium phosphate
Osteogenic differentiation
Bone marrow stem cells
Runt-related transcription factor 2
N6-methyladenosine
References

1. Wang Z, Guo Z, Bai H, et al., 2013, Clinical Evaluation of β-TCP in the Treatment of Lacunar Bone Defects: A Prospective, Randomized Controlled Study. Mater Sci Eng C, 33:1894–9. https://doi.org/10.1016/j.msec.2012.12.041

2. Seebach C, Henrich D, Kähling C, et al., 2010, Endothelial Progenitor Cells and Mesenchymal Stem Cells Seeded onto β-TCP Granules Enhance Early Vascularization and Bone Healing in a Critical-Sized Bone Defect in Rats. Tissue Eng Part A, 16:1961–70. https://doi.org/10.1089/ten.tea.2009.0715

3. Chu W, Zhuang Y, Gan Y, et al., 2019, Comparison and Characterization of Enriched Mesenchymal Stem Cells Obtained by the Repeated Filtration of Autologous Bone Marrow Through Porous Biomaterials. J Transl Med, 17:377. https://doi.org/10.1186/s12967-019-02131-y

4. Chu W, Gan Y, Zhuang Y, et al., 2018, Mesenchymal Stem Cells and Porous β-tricalcium Phosphate Composites Prepared through Stem Cell Screen-enrich-combine (Biomaterials) Circulating System for the Repair of Critical Size Bone Defects in Goat Tibia. Stem Cell Res Ther, 9:157. https://doi.org/10.1186/s13287-018-0906-1

5. Chu W, Wang X, Gan Y, et al., 2019, Screen-enrich-combine Circulating System to Prepare MSC/β-TCP for Bone Repair in Fractures with Depressed Tibial Plateau. Regener Med, 14:555–69. https://doi.org/10.2217/rme-2018-0047

6. Wang X, Chu W, Zhuang Y, et al., 2019, Bone Mesenchymal Stem Cell-Enriched β-Tricalcium Phosphate Scaffold Processed by the Screen-Enrich-Combine Circulating System Promotes Regeneration of Diaphyseal Bone Non-Union. Cell Transplant, 28:212–23. https://doi.org/10.1177/0963689718818096

7. Masaoka T, Yoshii T, Yuasa M, et al., 2016, Bone Defect Regeneration by a Combination of a β-Tricalcium Phosphate Scaffold and Bone Marrow Stromal Cells in a Non-Human Primate Model. Open Biomed Eng J, 10: 2-11.  https://doi.org/10.2174/1874120701610010002

8. Barradas AM, Monticone V, Hulsman M, et al., 2013, Molecular Mechanisms of Biomaterial-driven Osteogenic Differentiation in Human Mesenchymal Stromal Cells. Integr Biol, 5:920–31. https://doi.org/10.1039/c3ib40027a

9. Liu J, Zhao L, Ni L, et al., 2015, The Effect of Synthetic α-tricalcium Phosphate on Osteogenic Differentiation of Rat Bone Mesenchymal Stem Cells. Am J Transl Res, 7:1588–601.

10. Rittipakorn P, Thuaksuban N, Mai-Ngam K, et al., 2021, Bioactivity of a Novel Polycaprolactone-Hydroxyapatite Scaffold Used as a Carrier of Low Dose BMP-2: An In Vitro Study. Polymers (Basel), 13:466. https://doi.org/10.3390/polym13030466

11. Hesse E, Saito H, Kiviranta R, et al., 2010, Zfp521 Controls Bone Mass by HDAC3-dependent Attenuation of Runx2 Activity. J Cell Biol, 191:1271–83. https://doi.org/10.1083/jcb.201009107

12. Liu JC, Lengner CJ, Gaur T, et al., 2011, Runx2 Protein Expression Utilizes the Runx2 P1 Promoter to Establish Osteoprogenitor Cell Number for Normal Bone Formation. J Biol Chem, 286:30057–70. https://doi.org/10.1074/jbc.M111.241505

13. Kronenberg HM, 2004, Twist Genes Regulate Runx2 and Bone Formation. Dev Cell, 6:317–8. https://doi.org/10.1016/S1534-5807(04)00069-3

14. Yan G, Yuan Y, He M, et al., 2020, m6A Methylation of Precursor-miR-320/RUNX2 Controls Osteogenic Potential of Bone Marrow-Derived Mesenchymal Stem Cells. Mol Ther Nucleic Acids, 19:421–36. https://doi.org/10.1016/j.omtn.2019.12.001

15. Chen J, Tian Y, Zhang Q, et al., 2021, Novel Insights into the Role of N6-Methyladenosine RNA Modification in Bone Pathophysiology. Stem Cells Dev, 30:17–28. https://doi.org/10.1089/scd.2020.0157

16. Lee M, Kim B, Kim VN, 2014, Emerging Roles of RNA Modification: m 6 A and U-Tail. Cell, 158:980–7. https://doi.org/10.1016/j.cell.2014.08.005

17. Zhao BS, Roundtree IA, He C, 2017, Post-transcriptional Gene Regulation by mRNA Modifications. Nat Rev Mol Cell Biol, 18:31–42. https://doi.org/10.1038/nrm.2016.132

18. He C, 2010, Grand Challenge Commentary: RNA epigenetics? Nat Chem Biol, 6:863–65. https://doi.org/10.1038/nchembio.482

19. Yang Y, Hsu PJ, Chen YS, et al., 2018, Dynamic Transcriptomic m6A Decoration: Writers, Erasers, Readers and Functions in RNA Metabolism. Cell Res, 28:616–24. https://doi.org/10.1038/s41422-018-0040-8

20. Wu Y, Xie L, Wang M, et al., 2018, Mettl3-mediated m6A RNA Methylation regulates the fate of bone marrow mesenchymal stem cells and osteoporosis. Nat Commun, 9:4772. https://doi.org/10.1038/s41467-018-06898-4

21. Yu J, Shen L, Liu Y, et al., 2020, The m6A Methyltransferase METTL3 Cooperates with Demethylase ALKBH5 to Regulate Osteogenic Differentiation through NF-Κb Signaling. Mol Cell Biochem, 463:203–10. https://doi.org/10.1007/s11010-019-03641-5

22. Liu Y, Gu C, Li X, et al., 2021, Involvement of METTL3/m6Adenosine and TGFβ/Smad3 Signaling on Tenon’s Fibroblasts and in a Rabbit Model of Glaucoma Surgery. J Mol Histol, 52:1129–44. https://doi.org/10.1007/s10735-021-10028-8

23. Liu L, Yu F, Li L, et al., 2021, Bone Marrow Stromal Cells Stimulated by Strontium-substituted Calcium Silicate Ceramics: Release of Exosomal miR-146a Regulates Osteogenesis and Angiogenesis. Acta Biomater, 119:444–57. https://doi.org/10.1016/j.actbio.2020.10.038

24. Zhou P, Xia D, Ni Z, et al., 2021, Calcium Silicate Bioactive Ceramics Induce Osteogenesis through Oncostatin M. Bioact Mater, 6:810–22. https://doi.org/10.1016/j.bioactmat.2020.09.018

25. Huang Y, Jin X, Zhang X, et al., 2009, In Vitro and In Vivo Evaluation of Akermanite Bioceramics for Bone Regeneration. Biomaterials, 30:5041–8. https://doi.org/10.1016/j.biomaterials.2009.05.077

26. Sun X, Ma Z, Zhao X, et al., 2021, Three-dimensional Bioprinting of Multicell-laden Scaffolds Containing Bone Morphogenic Protein-4 for Promoting M2 Macrophage Polarization and Accelerating Bone Defect Repair in Diabetes Mellitus. Bioact Mater, 6:757–69. https://doi.org/10.1016/j.bioactmat.2020.08.030

27. Zheng M, Weng M, Zhang X, et al., 2021, Beta-tricalcium Phosphate Promotes Osteogenic Differentiation of Bone Marrow-derived Mesenchymal Stem Cells through Macrophages. Biomed Mater, 16:025005. https://doi.org/10.1088/1748-605X/abdbdc

28. El-Rashidy AA, Roether JA, Harhaus L, et al., 2017, Regenerating Bone with Bioactive Glass Scaffolds: A Review of In Vivo Studies in Bone Defect Models. Acta Biomater, 62:1–28. https://doi.org/10.1016/j.actbio.2017.08.030

29. Wu D, Wang Z, Wang J, et al., 2018, Development of a Micro-tissue-mediated Injectable Bone Tissue Engineering Strategy for Large Segmental Bone Defect Treatment. Stem Cell Res Ther, 9:331. https://doi.org/10.1186/s13287-018-1064-1

30. Stevens MM, 2008, Biomaterials for Bone Tissue Engineering. Mater Today, 11:18–25. https://doi.org/10.1016/S1369-7021(08)70086-5

31. Baldwin P, Li DJ, Auston DA, et al., 2019, Autograft, Allograft, and Bone Graft Substitutes: Clinical Evidence and Indications for Use in the Setting of Orthopaedic Trauma Surgery. J Orthop Trauma, 33:203–13. https://doi.org/10.1097/BOT.0000000000001420

32. Prakash J, Prema D, Venkataprasanna KS, et al., 2020, Nanocomposite Chitosan Film Containing Graphene Oxide/Hydroxyapatite/Gold for Bone Tissue Engineering. Int J Biol Macromol, 154:62–71. https://doi.org/10.1016/j.ijbiomac.2020.03.095

33. Kaushik N, Nguyen LN, Kim JH, et al., 2020, Strategies for Using Polydopamine to Induce Biomineralization of Hydroxyapatite on Implant Materials for Bone Tissue Engineering. Int J Mol Sci, 21:E6544. https://doi.org/10.3390/ijms21186544

34. Take Y, Mae T, Yoneda M, et al., 2020, On-lay Grafting of a Calcium Hydroxyapatite Bone Substitute: A Preliminary Animal Experimental Study. J Orthop Sci, 25:1101–6. https://doi.org/10.1016/j.jos.2019.12.012 

35. Sathiyavimal S, Vasantharaj S, Oscar FL, et al., 2019, Biosynthesis and Characterization of Hydroxyapatite and its Composite (Hydroxyapatite-gelatin-chitosan-fibrin-bone Ash) for Bone Tissue Engineering Applications. Int J Biol Macromol, 129:844–52. https://doi.org/10.1016/j.ijbiomac.2019.02.058

36. Beuriat PA, Lohkamp LN, Szathmari A, et al., 2019, Repair of Cranial Bone Defects in Children Using Synthetic Hydroxyapatite Cranioplasty (CustomBone). World Neurosurg, 129:e104–13. https://doi.org/10.1016/j.wneu.2019.05.052

37. Zhang H, Zhou Y, Yu N, et al., 2019, Construction of Vascularized Tissue-engineered Bone with Polylysine modified Coral Hydroxyapatite and a Double Cell-sheet Complex to Repair a Large Radius Bone Defect in Rabbits. Acta Biomater, 91:82–98. https://doi.org/10.1016/j.actbio.2019.04.024

38. Przekora A, Kazimierczak P, Wojcik M, 2021, Ex Vivo Determination of Chitosan/Curdlan/Hydroxyapatite Biomaterial Osseointegration with the Use of Human Trabecular Bone Explant: New Method for Biocompatibility Testing of Bone Implants Reducing Animal Tests. Mater Sci Eng C Mater Biol Appl, 119:111612. https://doi.org/10.1016/j.msec.2020.111612

39. Putri TS, Hayashi K, Ishikawa K, 2020, Bone Regeneration Using β-tricalcium Phosphate (β-TCP) block with Interconnected Pores Made by Setting Reaction of β-TCP Granules. J Biomed Mater Res A, 108:625–32. https://doi.org/10.1002/jbm.a.36842

40. Zhang D, Gao P, Li Q, et al., Engineering Biomimetic Periosteum with β-TCP Scaffolds to Promote Bone Formation in Calvarial Defects of Rats. Stem Cell Res Ther, 8:134. https://doi.org/10.1186/s13287-017-0592-4

41. Tian C, Huang Y, Li Q, et al., 2019, Mettl3 Regulates Osteogenic Differentiation and Alternative Splicing of Vegfa in Bone Marrow Mesenchymal Stem Cells. Int J Mol Sci, 20:551. https://doi.org/10.3390/ijms20030551

42. Son HE, Min HY, Kim EJ, et al., 2020, Fat Mass and Obesity-Associated (FTO) Stimulates Osteogenic Differentiation of C3H10T1/2 Cells by Inducing Mild Endoplasmic Reticulum Stress via a Positive Feedback Loop with p-AMPK. Mol Cells, 43:58–65.

43. Zhang C, Chen Y, Sun B, et al., 2017, m6A Modulates Haematopoietic Stem and Progenitor Cell Specification. Nature, 549:273–6. https://doi.org/10.1038/nature23883

44. Zhang B, Wu Q, Li B, et al., 2020, m6A Regulator-Mediated Methylation Modification Patterns and Tumor Microenvironment Infiltration Characterization in Gastric Cancer. Mol Cancer, 19:53. https://doi.org/10.1186/s12943-020-01170-0

45. Zhang Y, Gu X, Li D, et al., 2019, METTL3 Regulates Osteoblast Differentiation and Inflammatory Response via Smad Signaling and MAPK Signaling. Int J Mol Sci, 21:199. https://doi.org/10.3390/ijms21010199

46. Barbieri I, Tzelepis K, Pandolfini L, et al., 2017, Promoter bound METTL3 Maintains Myeloid Leukaemia by m6Adependent Translation Control. Nature, 552:126–31. https://doi.org/10.1038/nature24678

47. Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al., 2012, Topology of the Human and Mouse m6A RNA Methylomes Revealed by m6A-seq. Nature, 485:201–6. https://doi.org/10.1038/nature11112

48. Bartosovic M, Molares HC, Gregorova P, et al., 2017, N6-methyladenosine Demethylase FTO Targets pre-mRNAs and Regulates Alternative Splicing and 3’-end Processing. Nucleic Acids Res, 45:11356–70. https://doi.org/10.1093/nar/gkx778

49. Geula S, Moshitch-Moshkovitz S, Dominissini D, et al., 2015, Stem Cells. m6A mRNA Methylation Facilitates Resolution of Naïve Pluripotency toward Differentiation. Science, 347:1002–6. https://doi.org/10.1126/science.1261417

50. Kennedy EM, Bogerd HP, Kornepati AV, et al., 2017, Posttranscriptional m6A Editing of HIV-1 mRNAs Enhances Viral Gene Expression. Cell Host Microbe, 22:830. https://doi.org/10.1016/j.chom.2017.11.010

Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing