Skip to main content
Log in

In vitro and in vivo investigation on biodegradable Mg-Li-Ca alloys for bone implant application

镁锂钙合金作为骨植入材料的体内外研究

  • Articles
  • Published:
Science China Materials Aims and scope Submit manuscript

Abstract

Magnesium alloys show promise for application in orthopedic implants, owing to their biodegradability and biocompatibility. In the present study, ternary Mg-(3.5, 6.5 wt%) Li-(0.2, 0.5, 1.0 wt%) Ca alloys were developed. Their mechanical strength, corrosion behavior and cytocompatibility were studied. These alloys showed improved mechanical strength than pure Mg and exhibited suitable corrosion resistance. Furthermore, Mg-3.5Li-0.5Ca alloys with the best in vitro performance were implanted intramedullary into the femurs of mice for 2 and 8 weeks. In vivo results revealed a significant increase in cortical bone thickness around the Mg-3.5Li-0.5Ca alloy rods, without causing any adverse effects. Western blotting and immunofluorescence staining of β-catenin illustrated that Mg-3.5Li-0.5Ca alloy extracts induced osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMMSCs) through the canonical Wnt/β-catenin pathway. Our studies demonstrate that Mg-3.5Li-0.5Ca alloys hold much promise as candidates for the facilitation of bone implant application.

摘要

本文制备了三元Mg-(3.5, 6.5 wt.%)Li-(0.2, 0.5, 1.0 wt.%)Ca合金, 并研究了其力学性能、 腐蚀性能与生物相容性. 此合金的力学性能较纯镁显著提高, 并具有良好的耐腐蚀性. 然后, 将体外性能最佳的Mg-3.5Li-0.5Ca合金植入小鼠股骨骨髓腔, 体内实验结果显示, Mg-3.5Li-0.5Ca合金周围的骨厚度增加, 未见不良反应. Western blot和免疫荧光染色结果显示, Mg-3.5Li-0.5Ca合金通过经典的Wnt/β-catenin信号通路促进了人骨髓间充质干细胞的成骨向分化. 研究结果表明, Mg-3.5Li-0.5Ca合金具有作为骨植入材料的巨大潜力.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Witte F, Kaese V, Haferkamp H, et al. In vivo corrosion of four magnesium alloys and the associated bone response. Biomaterials, 2005, 26: 3557–3563

    Article  Google Scholar 

  2. Witte F, Fischer J, Nellesen J, et al. In vitro and in vivo corrosion measurements of magnesium alloys. Biomaterials, 2006, 27: 1013–1018

    Article  Google Scholar 

  3. Cheng P, Han P, Zhao C, et al. High-purity magnesium interference screws promote fibrocartilaginous entheses regeneration in the anterior cruciate ligament reconstruction rabbit model via accumulation of BMP-2 and VEGF. Biomaterials, 2016, 81: 14–26

    Article  Google Scholar 

  4. Zhao D, Huang S, Lu F, et al. Vascularized bone grafting fixed by biodegradable magnesium screw for treating osteonecrosis of the femoral head. Biomaterials, 2016, 81: 84–92

    Article  Google Scholar 

  5. Staiger MP, Pietak AM, Huadmai J, et al. Magnesium and its alloys as orthopedic biomaterials: A review. Biomaterials, 2006, 27: 1728–1734

    Article  Google Scholar 

  6. Witte F. The history of biodegradable magnesium implants: A review. Acta Biomater, 2010, 6: 1680–1692

    Article  Google Scholar 

  7. Rössig C, Angrisani N, Helmecke P, et al. In vivo evaluation of a magnesium-based degradable intramedullary nailing system in a sheep model. Acta Biomater, 2015, 25: 369–383

    Article  Google Scholar 

  8. Witte F, Hort N, Vogt C, et al. Degradable biomaterials based on magnesium corrosion. Curr Opin Solid State Mater Sci, 2008, 12: 63–72

    Article  Google Scholar 

  9. Xu W, Birbilis N, Sha G, et al. A high-specific-strength and corrosion-resistant magnesium alloy. Nat Mater, 2015, 14: 1229–1235

    Article  Google Scholar 

  10. Geddes JR, Burgess S, Hawton K, et al. Long-term lithium therapy for bipolar disorder: systematic review and meta-analysis of randomized controlled trials. Am J Psych, 2004, 161: 217–222

    Article  Google Scholar 

  11. Baastrup PC, Poulsen JC, Schou M, et al. Prophylactic lithium: double blind discontinuation in manic-depressive and recurrentdepressive disorders. Lancet, 1970, 296: 326–330

    Article  Google Scholar 

  12. Clément-Lacroix P, Ai M, Morvan F, et al. Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice. Proc Natl Acad Sci USA, 2005, 102: 17406–17411

    Article  Google Scholar 

  13. Day TF, Guo X, Garrett-Beal L, et al. Wnt/ß-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell, 2005, 8: 739–750

    Article  Google Scholar 

  14. Zamani A, Omrani GR, Nasab MM. Lithium’s effect on bone mineral density. Bone, 2009, 44: 331–334

    Article  Google Scholar 

  15. Hirai K, Somekawa H, Takigawa Y, et al. Effects of Ca and Sr addition on mechanical properties of a cast AZ91 magnesium alloy at room and elevated temperature. Mater Sci Eng-A, 2005, 403: 276–280

    Article  Google Scholar 

  16. Erdmann N, Angrisani N, Reifenrath J, et al. Biomechanical testing and degradation analysis of MgCa0.8 alloy screws: A comparative in vivo study in rabbits. Acta Biomater, 2011, 7: 1421–1428

    Article  Google Scholar 

  17. Haferkamp H, Niemeyer M, Boehm R, et al. Development, processing and applications range of magnesium lithium alloys. Magnesium alloys, 2000, 350–351: 31–41

    Google Scholar 

  18. Haferkamp H, Boehm R, Holzkamp U, et al. Alloy development, processing and applications in magnesium lithium alloys. Mater Trans, 2001, 42: 1160–1166

    Article  Google Scholar 

  19. Johnston Jr. CC, Miller JZ, Slemenda CW, et al. Calcium supplementation and increases in bone mineral density in children. N Engl J Med, 1992, 327: 82–87

    Article  Google Scholar 

  20. Tang BM, Eslick GD, Nowson C, et al. Use of calcium or calcium in combination with vitamin D supplementation to prevent fractures and bone loss in people aged 50 years and older: a metaanalysis. Lancet, 2007, 370: 657–666

    Article  Google Scholar 

  21. Zeng R, Sun X, Song Y, et al. Influence of solution temperature on corrosion resistance of Zn-Ca phosphate conversion coating on biomedical Mg-Li-Ca alloys. Trans Nonferrous Met Soc China, 2013, 23: 3293–3299

    Article  Google Scholar 

  22. Zeng RC, Sun L, Zheng YF, et al. Corrosion and characterisation of dual phase Mg–Li–Ca alloy in Hank’s solution: The influence of microstructural features. Corrosion Sci, 2014, 79: 69–82

    Article  Google Scholar 

  23. ASTM E8/E8M-16a. Standard Test Methods for Tension Testing of Metallic Materials, Annual Book of ASTM standards. 2004

    Google Scholar 

  24. Liu Y, Wu Y, Bian D, et al. Study on the Mg-Li-Zn ternary alloy system with improved mechanical properties, good degradation performance and different responses to cells. Acta Biomater, 2017, 62: 418–433

    Article  Google Scholar 

  25. Kirkland NT, Birbilis N, Staiger MP. Assessing the corrosion of biodegradable magnesium implants: A critical review of current methodologies and their limitations. Acta Biomater, 2012, 8: 925–936

    Article  Google Scholar 

  26. ASTM G31-72. Standard Practice for Laboratory Immersion Corrosion Testing of Metals. 1990

    Google Scholar 

  27. Zeng RC, Cui L, Jiang K, et al. In vitro corrosion and cytocompatibility of a microarc oxidation coating and poly(l-lactic acid) composite coating on Mg–1Li–1Ca alloy for orthopedic implants. ACS Appl Mater Interfaces, 2016, 8: 10014–10028

    Article  Google Scholar 

  28. ISO 10993–5. Biological evaluation of medical devices—Part 5: Tests for in vitro cytotoxicity. International Organization for Standardisation, 2009

    Google Scholar 

  29. Liu Y, Zhang X, Liu Y, et al. Bi-functionalization of a calcium phosphate-coated titanium surface with slow-release simvastatin and metronidazole to provide antibacterial activities and proosteodifferentiation capabilities. PLoS ONE, 2014, 9: e97741

    Article  Google Scholar 

  30. Ge W, Shi L, Zhou Y, et al. Inhibition of osteogenic differentiation of human adipose-derived stromal cells by retinoblastoma binding protein 2 repression of RUNX2-activated transcription. Stem Cells, 2011, 29: 1112–1125

    Article  Google Scholar 

  31. Wang Y, Wei M, Gao J, et al. Corrosion process of pure magnesium in simulated body fluid. Mater Lett, 2008, 62: 2181–2184

    Article  Google Scholar 

  32. Zhang M, Elkin FM. Mg-Li Ultra-light Alloy. Beijing: Science Press, 2010

    Google Scholar 

  33. Cipriano AF, Sallee A, Guan RG, et al. Investigation of magnesium–zinc–calcium alloys and bone marrow derived mesenchymal stem cell response in direct culture. Acta Biomater, 2015, 12: 298–321

    Article  Google Scholar 

  34. Yang C, Yuan G, Zhang J, et al. Effects of magnesium alloys extracts on adult human bone marrow-derived stromal cell viability and osteogenic differentiation. Biomed Mater, 2010, 5: 045005

    Article  Google Scholar 

  35. Wu Y, Zhu S, Wu C, et al. A bi-lineage conducive scaffold for osteochondral defect regeneration. Adv Funct Mater, 2014, 24: 4473–4483

    Article  Google Scholar 

  36. Poitevin AA, Viezzer C, Machado DC, et al. Effect of standard and neutral-pH peritoneal dialysis solutions upon fibroblasts pro-liferation. J Bras Nefrol, 2014, 36: 150–154

    Article  Google Scholar 

  37. Nguyen TY, Liew CG, Liu H. An in vitro mechanism study on the proliferation and pluripotency of human embryonic stems cells in response to magnesium degradation. PLoS ONE, 2013, 8: e76547

    Article  Google Scholar 

  38. Wang J, Witte F, Xi T, et al. Recommendation for modifying current cytotoxicity testing standards for biodegradable magnesium-based materials. Acta Biomater, 2015, 21: 237–249

    Article  Google Scholar 

  39. Li Z, Gu X, Lou S, et al. The development of binary Mg–Ca alloys for use as biodegradable materials within bone. Biomaterials, 2008, 29: 1329–1344

    Article  Google Scholar 

  40. Zhang Y, Xu J, Ruan YC, et al. Implant-derived magnesium induces local neuronal production of CGRP to improve bone-fracture healing in rats. Nat Med, 2016, 22: 1160–1169

    Article  Google Scholar 

  41. Han P, Wu C, Chang J, et al. The cementogenic differentiation of periodontal ligament cells via the activation of Wnt/ß-catenin signalling pathway by Li+ ions released from bioactive scaffolds. Biomaterials, 2012, 33: 6370–6379

    Article  Google Scholar 

  42. Tang L, Chen Y, Pei F, et al. Lithium chloride modulates adipogenesis and osteogenesis of human bone marrow-derived mesenchymal stem cells. Cell Physiol Biochem, 2015, 37: 143–152

    Article  Google Scholar 

  43. Rude RK, Gruber HE. Magnesium deficiency and osteoporosis: animal and human observations. J Nutritional Biochem, 2004, 15: 710–716

    Article  Google Scholar 

  44. Yoshizawa S, Brown A, Barchowsky A, et al. Magnesium ion stimulation of bone marrow stromal cells enhances osteogenic activity, simulating the effect of magnesium alloy degradation. Acta Biomater, 2014, 10: 2834–2842

    Article  Google Scholar 

  45. Liu Z, Yao X, Yan G, et al. Mediator MED23 cooperates with RUNX2 to drive osteoblast differentiation and bone development. Nat Commun, 2016, 7: 11149

    Article  Google Scholar 

  46. Komori T. Regulation of osteoblast differentiation by Runx2. Adv Exp Med Biol, 2010, 658: 43–49

    Article  Google Scholar 

  47. Neve A, Corrado A, Cantatore FP. Osteocalcin: Skeletal and extraskeletal effects. J Cell Physiol, 2013, 228: 1149–1153

    Article  Google Scholar 

  48. Nakashima K, Zhou X, Kunkel G, et al. The novel zinc fingercontaining transcription factor osterix is required for osteoblast differentiation and bone formation. Cell, 2002, 108: 17–29

    Article  Google Scholar 

  49. Liu F, Kohlmeier S, Wang CY. Wnt signaling and skeletal development. Cellular Signalling, 2008, 20: 999–1009

    Article  Google Scholar 

  50. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol, 2004, 20: 781–810

    Article  Google Scholar 

  51. Sheng H. Nuclear translocation of beta-catenin in hereditary and carcinogen-induced intestinal adenomas. Carcinogenesis, 1998, 19: 543–549

    Article  Google Scholar 

  52. Zhou WR, Zheng YF, Leeflang MA, et al. Mechanical property, biocorrosion and in vitro biocompatibility evaluations of Mg–Li–(Al)–(RE) alloys for future cardiovascular stent application. Acta Biomater, 2013, 9: 8488–8498

    Article  Google Scholar 

  53. Cui L, Sun L, Zeng R, et al. In vitro degradation and biocompatibility of Mg-Li-Ca alloys—the influence of Li content. Sci China Mater, 2018, 61: 607–618

    Article  Google Scholar 

  54. Díaz-Tocados JM, Herencia C, Martínez-Moreno JM, et al. Magnesium chloride promotes osteogenesis through notch signaling activation and expansion of mesenchymal stem cells. Sci Rep, 2017, 7: 7839

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key Research and Development Program of China (2016YFC1102900 and 2016YFC1102402), the National Natural Science Foundation of China (81771039, 81470769 and 51431002), the Project for Culturing Leading Talents in Scientific and Technological Innovation of Beijing, China (Z171100001117169), the NSFC-RFBR Cooperation Project (51611130054), and the NSFC/RGC Joint Research Scheme (51361165101 and 5161101031).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Yunsong Liu  (刘云松) or Yufeng Zheng  (郑玉峰).

Additional information

Dandan Xia is currently a PhD student at the School and Hospital of Stomatology, Peking University. She received her Bachelor’s degree from Peking University in 2015. Her research focuses on the magnesium alloys and mesenchymal stem cells.

Yunsong Liu received his PhD in the School and Hospital of Stomatology from Peking University, China, in 2008. Since 2008, he has been a full professor at Peking University in Beijing, China. His research focuses on the magnesium alloys and mesenchymal stem cells.

Yufeng Zheng received his PhD in materials science from Harbin Institute of Technology, China, in 1998. Since 2004, he has been a full professor at Peking University in Beijing, China. His research focuses on the development of various new biomedical metallic materials (biodegradable Mg, Fe and Zn based alloys, β-Ti alloys with low elastic modulus, bulk metallic glass, ultra-fine grained metallic materials, etc.).

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xia, D., Liu, Y., Wang, S. et al. In vitro and in vivo investigation on biodegradable Mg-Li-Ca alloys for bone implant application. Sci. China Mater. 62, 256–272 (2019). https://doi.org/10.1007/s40843-018-9293-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40843-018-9293-8

Keywords

Navigation