AccScience Publishing / IJB / Volume 10 / Issue 2 / DOI: 10.36922/ijb.1698
Cite this article
51
Download
624
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
RESEARCH ARTICLE

Effect of lattice type on biomechanical and osseointegration properties of 3D-printed porous Ti6Al4V scaffolds

Jiantao Liu1 Kao Wang2 Runqing Wang3 Zhanhai Yin1 Xiaoling Zhou1 Aofei Xu4 Xiwei Zhang4 Yiming Li4 Ruiyan Wang4 Shuyuan Zhang4 Jun Cheng5 Weiguo Bian1 Jia Li1 Zhiwei Ren1 Mengyuan Sun1 Yin Yang6* Dezhi Wang7* Jing Ren1*
Show Less
1 Department of Orthopedics, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
2 Medical School of Yan’an University, Yanan, Shaanxi, China
3 Lanzhou University, Lanzhou, Gansu, China
4 Xi’an Jiaotong University, Xi’an, Shaanxi, China
5 Shaanxi Key laboratory of Biomedical Metal Materials, Northwest Institute for Nonferrous Metal Research, Xi’an, Shaanxi, China
6 Department of Orthopedics, Xi’an Central Hospital, Xi’an, Shaanxi, China
7 Anesthesiology Department, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
IJB 2024, 10(2), 1698 https://doi.org/10.36922/ijb.1698
Submitted: 28 August 2023 | Accepted: 24 October 2023 | Published: 8 January 2024
(This article belongs to the Special Issue Advancements in 3D bioprinting applied to musculoskeletal tissues)
© 2024 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

Porous structure is an efficient tool for optimizing the elastic modulus and osseointegration properties of titanium alloy materials. However, the investigations on pore shape remain scarce. In this study, we created porous Ti6Al4V scaffolds with a pore size of 600 μm but different lattices (cubic pentagon, diamond, cuboctahedron). The mechanical and biological properties of the scaffolds were investigated in static simulation analysis, in vitro mechanical compression test, computational fluid dynamics, as well as cell and animal experiments. The results demonstrated that the calculated yield strength difference between the three Ti6Al4V porous scaffolds was negligible, at approximately 140 MPa, allowing them to match the strength requirements of human bones. The diamond scaffold has the lowest calculated elastic modulus (11.6 GPa), which is conducive for preventing stress shielding. The shear stress was largely concentrated in the diamond scaffold, and the stress range of 120–140 MPa accounted for the greatest share. The mouse MC3T3-E1 cells were found to attach to all three scaffolds, with the diamond scaffold displaying a higher degree of cell adherence. There was more proliferating cells on the diamond and cubic pentagon scaffolds than on the cuboctahedron scaffolds (P < 0.05). The diamond scaffold exhibited the highest alkaline phosphatase activity and calcium salt accumulation in cell differentiation tests. Besides, the expression of osteogenic genes on the diamond scaffold was higher than that on the cuboctahedron scaffold, the cubic pentagon scaffold displaying the lowest expression. The in vivo studies revealed that all three scaffolds fused well with the surrounding bone and that there was no loosening or movement of the prosthesis. Micro-computed tomography, corroborated by the staining results of hard tissues, revealed that the level of new bone formation was the highest in the diamond scaffold, followed by the cuboctahedron scaffold (P < 0.05). Taken together, the diamond scaffold is comparatively better at optimizing the elastic modulus and osseointegration properties of titanium alloy materials, and thus is a preferred choice for porous design.

Keywords
Titanium
Porous structure
Lattice
Biomechanics
Osseointegration
Funding
This work was financially supported by Key Research and Development Plan of Shaanxi Province-General Project (2022GY-390), Health and Family Planning Commission of Shaanxi Province (2022E001), General Cultivation Project of Xi’an Municipal Health Commission (2022ms02), Shaanxi Natural Science Basic Research Foundation (2021JM-276), Key Research and Development Program of Shaanxi (2023-YBGY-488), and National Natural Science Foundation of China (52271249).
References
  1. Neto MQ, Radice S, Hall DJ, et al. Microstructure and electrochemical behavior of contemporary Ti6Al4V implant alloys. J Bio Tribocorros. 2022;8(1). doi: 10.1007/s40735-021-00623-3
  2. Xi D, Wong L. Titanium and implantology: A review in dentistry. J Biol Regul Homeost Agents. 2021;35(1 Suppl. 1):63- 72. doi: 10.3390%2Fma15093150
  3. Gkiatas I, Sharma AK, Driscoll DA, McLawhorn AS, Chalmers BP, Sculco PK. Nonconcentric and irregular dislocations of total hip arthroplasties: Radiographic analysis and review of the literature. J Emerg Med. 2021;60(4): 451-459. doi: 10.1016/j.jemermed.2020.11.023
  4. Liu T, Hua X, Yu W, et al. Long-term follow-up outcomes for patients undergoing primary total hip arthroplasty with uncemented versus cemented femoral components: A retrospective observational study with a 5-year minimum follow-up. J Orthop Surg Res. 2019;14(1):371. doi: 10.1186/s13018-019-1415-3
  5. Raja BS, Gowda A, Singh S, Ansari S, Kalia RB, Paul S. Comparison of functional outcomes and complications of cemented vs uncemented total hip arthroplasty in the elderly neck of femur fracture patients: A systematic review and meta-analysis. J Clin Orthop Trauma. 2022;29:101876. doi: 10.1016%2Fj.jcot.2022.101876
  6. Bittredge O, Hassanin H, El-Sayed MA, et al. Fabrication and optimisation of Ti-6Al-4V lattice-structured total shoulder implants using laser additive manufacturing. Materials (Basel). 2022;15(9). doi: 10.3390/ma15093095
  7. Ito K, Mori Y, Kamimura M, et al. Beta-type TiNbSn alloy plates with low Young modulus accelerates osteosynthesis in Rabbit Tibiae. Clin Orthop Relat Res. 2022;480(9):1817-1832. doi: 10.1097/corr.0000000000002240
  8. Lei P, Qian H, Zhang T, et al. Porous tantalum structure integrated on Ti6Al4V base by laser powder bed fusion for enhanced bony-ingrowth implants: In vitro and in vivo validation. Bioact Mater. 2022;7:3-13. doi: 10.1016/j.bioactmat.2021.05.025
  9. Li L, Li Y, Yang L, et al. Polydopamine coating promotes early osteogenesis in 3D printing porous Ti6Al4V scaffolds. Ann Transl Med. 2019;7(11):240. doi: 10.21037/atm.2019.04.79
  10. Meena VK, Kalra P, Sinha RK. Finite element study on the influence of pore size and structure on stress shielding effect of additive manufactured spinal cage. Comput Methods Biomech Biomed Engin. 2022;25(5):566-577. doi: 10.1080/10255842.2021.1970142
  11. Al-Tamimi AA. 3D topology optimization and mesh dependency for redesigning locking compression plates aiming to reduce stress shielding. Int J Bioprint. 2021;7(3):339. doi: 10.18063/ijb.v7i3.339
  12. Han X, Ma J, Tian A, et al. Surface modification techniques of titanium and titanium alloys for biomedical orthopaedics applications: A review. Colloids Surf B Biointerfaces. 2023;227:113339. doi: 10.1016/j.colsurfb.2023.113339
  13. Xia C, Ma X, Zhang X, et al. Enhanced physicochemical and biological properties of C/Cu dual ions implanted medical titanium. Bioact Mater. 2020;5(2):377-386. doi: 10.1016/j.bioactmat.2020.02.017
  14. Wang Q, Zhou P, Liu S, et al. 2020, Multi-scale surface treatments of titanium implants for rapid osseointegration: A review. Nanomaterials (Basel), 10(6). doi: 10.3390%2Fnano10061244
  15. Wang L, Zhou W, Yu Z, et al. An in vitro evaluation of the hierarchical micro/nanoporous structure of a Ti3Zr2Sn3Mo25Nb alloy after surface dealloying. ACS Appl Mater Interfaces. 2021;13(13):15017-15030. doi: 10.1021/acsami.1c02140
  16. Wang R, Ni S, Ma L, Li M. Porous construction and surface modification of titanium-based materials for osteogenesis: A review. Front Bioeng Biotechnol. 2022; 10:973297. doi: 10.3389%2Ffbioe.2022.973297
  17. Liverani E, Rogati G, Pagani S, Brogini S, Fortunato A, Caravaggi P. Mechanical interaction between additive-manufactured metal lattice structures and bone in compression: Implications for stress shielding of orthopaedic implants. J Mech Behav Biomed Mater. 2021;121:104608. doi: 10.1016/j.jmbbm.2021.104608
  18. Arabnejad S, Johnston B, Tanzer M, Pasini D. Fully porous 3D printed titanium femoral stem to reduce stress-shielding following total hip arthroplasty. J Orthop Res. 2017; 35(8):1774-1783. doi: 10.1002/jor.23445
  19. Yao YT, Yang Y, Ye Q, et al. Effects of pore size and porosity on cytocompatibility and osteogenic differentiation of porous titanium. J Mater Sci Mater Med. 2021;32(6):72. doi: 10.1007/s10856-021-06548-0
  20. Cuan-Urquizo E, Silva RG. Fused filament fabrication of cellular, lattice and porous mechanical metamaterials: A review. Virtual Phys Prototyp. 2023;1(18):e2224300. doi: 10.1080/17452759.2023.2224300
  21. Yan L, Wu J, Zhang L, Liu X, Zhou K, Su B. Pore structures and mechanical properties of porous titanium scaffolds by bidirectional freeze casting. Mater Sci Eng C Mater Biol Appl. 2017;75:335-340. doi: 10.1016/j.msec.2016.12.044
  22. Dziaduszewska M, Zielinski A. Structural and material determinants influencing the behavior of porous ti and its alloys made by additive manufacturing techniques for biomedical applications. Materials (Basel). 2021;14(4). doi: 10.3390/ma14040712
  23. Xu Z, Zhang Y, Wu Y, et al. In vitro and in vivo analysis of the effects of 3D-printed porous titanium alloy scaffold structure on osteogenic activity. Biomed Res Int. 2022;2022:8494431. doi: 10.1155/2022/8494431
  24. Wang H, Su K, Su L, Liang P, Ji P, Wang C. Comparison of 3D-printed porous tantalum and titanium scaffolds on osteointegration and osteogenesis. Mater Sci Eng C Mater Biol Appl. 2019;104:109908. doi: 10.1016/j.msec.2019.109908
  25. Yang J, Li Y, Shi X, et al. Design and analysis of three-dimensional printing of a porous titanium scaffold. BMC Musculoskelet Disord. 2021;22(1):654. doi: 10.1186/s12891-021-04520-1
  26. Afrouzian A, Groden CJ, Field DP, Bose S, Bandyopadhyay A. Additive manufacturing of Ti-Ni bimetallic structures. Mater Des. 2022;215. doi: 10.1016%2Fj.matdes.2022.110461
  27. Huang G, Pan ST, Qiu JX. The osteogenic effects of porous Tantalum and Titanium alloy scaffolds with different unit cell structure. Colloids Surf B Biointerfaces. 2022;210:112229. doi: 10.1016/j.colsurfb.2021.112229
  28. Ciliveri S, Bandyopadhyay A. Influence of strut-size and cell-size variations on porous Ti6Al4V structures for load-bearing implants. J Mech Behav Biomed Mater. 2022;126:105023. doi: 10.1016/j.jmbbm.2021.105023
  29. Deng F, Liu L, Li Z, Liu J. 3D printed Ti6Al4V bone scaffolds with different pore structure effects on bone ingrowth. J Biol Eng. 2021;15(1):4. doi: 10.1186/s13036-021-00255-8
  30. Lei H, Yi T, Fan H, et al. Customized additive manufacturing of porous Ti6Al4V scaffold with micro-topological structures to regulate cell behavior in bone tissue engineering. Mater Sci Eng C Mater Biol Appl. 2021;120:111789. doi: 10.1016/j.msec.2020.111789
  31. Suresh S, Sun CN, Tekumalla S, Rosa V, Ling Nai SM, Wen Wong RC. Mechanical properties and in vitro cytocompatibility of dense and porous Ti-6Al-4V ELI manufactured by selective laser melting technology for biomedical applications. J Mech Behav Biomed Mater. 2021;123:104712. doi: 10.1016/j.jmbbm.2021.104712
  32. Ran Q, Yang W, Hu Y, et al. Osteogenesis of 3D printed porous Ti6Al4V implants with different pore sizes. J Mech Behav Biomed Mater. 2018;84:1-11. doi: 10.1016/j.jmbbm.2018.04.010
  33. Zaharin HA, Abdul RA, Azam FI, et al. Effect of unit cell type and pore size on porosity and mechanical behavior of additively manufactured Ti6Al4V scaffolds. Materials (Basel). 2018;11(12). doi: 10.3390%2Fma11122402
  34. Taniguchi N, Fujibayashi S, Takemoto M, et al. Effect of pore size on bone ingrowth into porous titanium implants fabricated by additive manufacturing: An in vivo experiment. Mater Sci Eng C Mater Biol Appl. 2016;59:690-701. doi: 10.1016/j.msec.2015.10.069
  35. Lu Y, Cheng L, Yang Z, Li J, Zhu H. Relationship between the morphological, mechanical and permeability properties of porous bone scaffolds and the underlying microstructure. PLoS One. 2020;15(9):e238471. doi: 10.1371%2Fjournal.pone.0238471
  36. Chen H, Han Q, Wang C, Liu Y, Chen B, Wang J. Porous scaffold design for additive manufacturing in orthopedics: A review. Front Bioeng Biotechnol. 2020;8:609. doi: 10.3389/fbioe.2020.00609
  37. Li X, Wang Y, Zhang B, et al. The design and evaluation of bionic porous bone scaffolds in fluid flow characteristics and mechanical properties. Comput Methods Programs Biomed. 2022;225:107059. doi: 10.1016/j.cmpb.2022.107059
  38. Omar AM, Hassan MH, Daskalakis E, et al. Geometry-based computational fluid dynamic model for predicting the biological behavior of bone tissue engineering scaffolds. J Funct Biomater. 2022;13(3). doi: 10.3390/jfb13030104
  39. Chao L, Jiao C, Liang H, Xie D, Shen L, Liu Z. Analysis of mechanical properties and permeability of trabecular-like porous scaffold by additive manufacturing. Front Bioeng Biotechnol. 2021;9:779854. doi: 10.3389%2Ffbioe.2021.779854
  40. Zhang C, Zhu H, Ren X, et al. Mechanics-driven nuclear localization of YAP can be reversed by N-cadherin ligation in mesenchymal stem cells. Nat Commun. 2021;12(1):6229. doi: 10.1038/s41467-021-26454-x
  41. Zhang Z, Sha B, Zhao L, et al. Programmable integrin and N-cadherin adhesive interactions modulate mechanosensing of mesenchymal stem cells by cofilin phosphorylation. Nat Commun. 2022;13(1):6854. doi: 10.1038/s41467-022-34424-0
  42. De Belly H, Paluch EK, Chalut KJ. Interplay between mechanics and signalling in regulating cell fate. Nat Rev Mol Cell Biol. 2022;23(7):465-480. doi: 10.1038/s41580-022-00472-z
  43. Truscello S, Kerckhofs G, Van Bael S, Pyka G, Schrooten J, Van Oosterwyck H. Prediction of permeability of regular scaffolds for skeletal tissue engineering: A combined computational and experimental study. Acta Biomater. 2012;8(4):1648-1658. doi: 10.1016/j.actbio.2011.12.021
  44. Winther NS, Jensen CL, Jensen CM, et al. Comparison of a novel porous titanium construct (Regenerex(R)) to a well proven porous coated tibial surface in cementless total knee arthroplasty - A prospective randomized RSA study with two-year follow-up. Knee. 2016;23(6): 1002-1011. doi: 10.1016/j.knee.2016.09.010
  45. Arts M, Torensma B, Wolfs J. Porous titanium cervical interbody fusion device in the treatment of degenerative cervical radiculopathy; 1-year results of a prospective controlled trial. Spine J. 2020;20(7):1065-1072. doi: 10.1016/j.spinee.2020.03.008
  46. Ibhadode O, Zhang Z, Sixt J, et al. Topology optimization for metal additive manufacturing: current trends, challenges, and future outlook. Virtual Phys Prototyp. 2023;1(18):e2181192. doi: 10.1080/17452759.2023.2181192
  47. Ahmadi SM, Yavari SA, Wauthle R, et al. Additively manufactured open-cell porous biomaterials made from six different space-filling unit cells: The mechanical and morphological properties. Materials (Basel). 2015;8(4):1871-1896. doi: 10.3390%2Fma8041871
  48. Hedayati R, Sadighi M, Mohammadi-Aghdam M, et al. Mechanics of additively manufactured porous biomaterials based on the rhombicuboctahedron unit cell. J Mech Behav Biomed Mater. 2016;53:272-294. doi: 10.1016/j.jmbbm.2015.07.013
  49. Matena J, Petersen S, Gieseke M, et al. SLM produced porous titanium implant improvements for enhanced vascularization and osteoblast seeding. Int J Mol Sci; 2015;16(4):7478-7492. doi: 10.3390%2Fijms16047478
  50. Gogolewski D, Kozior T, Zmarzly P, Gogolewski D. Morphology of models manufactured by SLM technology and the Ti6Al4V titanium alloy designed for medical applications. Materials (Basel). 2021;14(21). doi: 10.3390/ma14216249
  51. Ataee A, Li Y, Brandt M, Wen C. Ultrahigh-strength titanium gyroid scaffolds manufactured by selective laser melting (SLM) for bone implant applications. Acta Materialia. 2018;158:15. doi: 10.3390%2Fjfb14030125
  52. Li JL, Guo D, L J, et al. Irregular pore size of degradable bioceramic Voronoi scaffolds prepared by stereolithography: Osteogenesis and computational fluid dynamics analysis. Mater Des. 2022;224:111414. doi: 10.1016/j.matdes.2022.111414
  53. Li J, Chen D, Luan H, Zhang Y, Fan Y. Numerical evaluation and prediction of porous implant design and flow performance. Biomed Res Int. 2018;2018:1215021. doi: 10.1155%2F2018%2F1215021
  54. Li J, Chen D, Fan Y, Evaluation and prediction of mass transport properties for porous implant with different unit cells: A numerical study. Biomed Res Int. 2019;2019:3610785. doi: 10.1155/2019/3610785
  55. Porter B, Zauel R, Stockman H, Guldberg R, Fyhrie D. 3-D computational modeling of media flow through scaffolds in a perfusion bioreactor. J Biomech. 2005;38(3):543-549. doi: 10.1016/j.jbiomech.2004.04.011
  56. Cartmell SH, Porter BD, Garcia AJ, Guldberg RE. Effects of medium perfusion rate on cell-seeded three-dimensional bone constructs in vitro. Tissue Eng. 2003;9(6):1197-1203. doi: 10.1089/10763270360728107
  57. Raimondi MT, Boschetti F, Falcone L, et al. Mechanobiology of engineered cartilage cultured under aquantified fluid-dynamic environment. Biomechan Model Mechanobiol. 2002;1(1):14. doi: 10.1007/s10237-002-0007-y
  58. Van Bael S, Chai YC, Truscello S, et al. The effect of pore geometry on the in vitro biological behavior of human periosteum-derived cells seeded on selective laser-melted Ti6Al4V bone scaffolds. Acta Biomater. 2012;8(7):2824-2834. doi: 10.1016/j.actbio.2012.04.001
  59. 59. Rudrich U, Lasgorceix M, Champion E, et al. Pre-osteoblast cell colonization of porous silicon substituted hydroxyapatite bioceramics: Influence of microporosity and macropore design. Mater Sci Eng C Mater Biol Appl. 2019;97:510-528. doi: 10.1016/j.msec.2018.12.046
Conflict of interest
The authors declare they have no competing interest.
Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing