1
|
Liu Z, Zhao MC, Yin D, Zhao YC, Atrens A. Bio-functional niobium-based metallic biomaterials: Exploring their physicomechanical properties, biological significance, and implant applications. Acta Biomater 2025; 192:1-27. [PMID: 39681153 DOI: 10.1016/j.actbio.2024.12.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
The significance of biomedical applications of bio-functional niobium (Nb)-based metallic biomaterials is underscored by their potential utilization in implant application. Nb-based metallic materials present reliable physicomechanical and biological properties, thus represent materials highly suitable for implant application. This review provides an overview on the advances of pure niobium and Nb-based metallic materials as implant materials over the past 20 years, and highlights the advantages of Nb-based metallic biomaterials for implant application in terms of their physicomechanical properties, corrosion resistance in biological media, magnetic resonance imaging (MRI) compatibility, cell compatibility, blood compatibility, osteogenesis, and bioactivity. An introduction is provided for the production and processing techniques for Nb-based metallic biomaterials, including traditional melting processes like vacuum arc remelting, additive manufacturing like selective laser melting (SLM), electron beam melting (EBM), spark plasma sintering (SPS), and severe plastic deformation like equal channel angular pressing (ECAP), multi-axial forging (MAF), high pressure torsion (HPT), as well as their physicomechanical properties and implant application. Also suggested are the critical issues, challenges, and prospects in the further development of Nb-based metallic biomaterials for implant applications. STATEMENT OF SIGNIFICANCE: Nb-based biomaterials have gained significant interest for bioimplantable scaffolds because of their appropriate mechanical characteristics and biocompatibility. No prior work has been published specifically reviewing bio-functional Nb-based biomaterials for exploring their physicomechanical properties, biological significance, and implant applications. This review provides an overview on the advances of niobium and Nb-based materials as implant materials over the past 20 years, and highlights the advantages of Nb-based biomaterials for implant application. An introduction is provided for the production and processing techniques for Nb-based biomaterials, as well as their physicomechanical properties and implant application. Also suggested are the critical issues, challenges, and prospects in the further development of Nb-based biomaterials for implant applications.
Collapse
Affiliation(s)
- Ziyuan Liu
- College of Mechanical Engineering, University of South China, Hengyang 421001, PR China; School of Materials Science and Engineering, Central South University, Changsha 410083, PR China
| | - Ming-Chun Zhao
- School of Materials Science and Engineering, Central South University, Changsha 410083, PR China
| | - Dengfeng Yin
- School of Materials Science and Engineering, Central South University, Changsha 410083, PR China
| | - Ying-Chao Zhao
- College of Mechanical Engineering, University of South China, Hengyang 421001, PR China.
| | - Andrej Atrens
- School of Mechanical and Mining Engineering, University of Queensland, Brisbane QLD4072, Australia
| |
Collapse
|
2
|
Emanuelli L, Babaei M, De Biasi R, du Plessis A, Trivisonno A, Agostinacchio F, Motta A, Benedetti M, Pellizzari M. Optimising β-Ti21S Alloy Lattice Structures for Enhanced Femoral Implants: A Study on Mechanical and Biological Performance. MATERIALS (BASEL, SWITZERLAND) 2025; 18:170. [PMID: 39795817 PMCID: PMC11722399 DOI: 10.3390/ma18010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025]
Abstract
The metastable β-Ti21S alloy exhibits a lower elastic modulus than Ti-6Al-4V ELI while maintaining high mechanical strength and ductility. To address stress shielding, this study explores the integration of lattice structures within prosthetics, which is made possible through additive manufacturing. Continuous adhesion between the implant and bone is essential; therefore, auxetic bow-tie structures with a negative Poisson's ratio are proposed for regions under tensile stress, while Triply Periodic Minimal Surface (TPMS) structures with a positive Poisson's ratio are recommended for areas under compressive stress. This research examines the manufacturability and quasi-static mechanical behaviour of two auxetic bow-tie (AUX 2.5 and AUX 3.5) and two TPMS structures (TPMS 2.5 and TPMS 1.5) in β-Ti21S alloy produced via laser powder bed fusion. Micro-CT reveals printability issues in TPMS 1.5, affecting pore size and reducing fatigue resistance compared to TPMS 2.5. AUX 3.5's low stiffness matches cancellous bone but shows insufficient yield strength and fatigue resistance for femoral implants. Biological tests confirm non-toxicity and enhanced cell activity in β-Ti21S structures. The study concludes that the β-Ti21S alloy, especially with TPMS 2.5 structures, demonstrates promising mechanical and biological properties for femoral implants. However, challenges like poor printability in TPMS 1.5 are acknowledged and should be addressed in future research.
Collapse
Affiliation(s)
- Lorena Emanuelli
- INSTM Operative Center, University of Trento, 38122 Trento, Italy;
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
| | - Melika Babaei
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
- BIOTech Research Center, University of Trento, 38123 Trento, Italy
| | - Raffaele De Biasi
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
| | - Anton du Plessis
- Research Group 3D Innovation, Stellenbosch University, Stellenbosch 7602, South Africa;
- Object Research Systems, Montreal, QC H3C 1M4, Canada
| | | | - Francesca Agostinacchio
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
- BIOTech Research Center, University of Trento, 38123 Trento, Italy
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
- BIOTech Research Center, University of Trento, 38123 Trento, Italy
| | - Matteo Benedetti
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
| | - Massimo Pellizzari
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy; (M.B.); (R.D.B.); (F.A.); (A.M.); (M.P.)
| |
Collapse
|
3
|
Zhang Y, Yang J, Wan W, Zhao Q, Di M, Zhang D, Liu G, Chen C, Sun X, Zhang W, Bian H, Liu Y, Tian Y, Xue L, Dou Y, Wang Z, Li Q, Yang Q. Evaluation of biological performance of 3D printed trabecular porous tantalum spine fusion cage in large animal models. J Orthop Translat 2025; 50:185-195. [PMID: 39895865 PMCID: PMC11786794 DOI: 10.1016/j.jot.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 02/04/2025] Open
Abstract
Background The materials for artificial bone scaffolds have long been a focal point in biomaterials research. Tantalum, with its excellent bioactivity and tissue compatibility, has gradually become a promising alternative material. 3D printing technology shows unique advantages in designing complex structures, reducing costs, and providing personalized customization in the manufacture of porous tantalum fusion cages. Here we report the pre-clinical large animal (sheep) study on the newly developed 3D printed biomimetic trabecular porous tantalum fusion cage for assessing the long-term intervertebral fusion efficacy and safety. Methods Porous tantalum fusion cages were fabricated using laser powder bed fusion (LPBF) and chemical vapor deposition (CVD) methods. The fusion cages were characterized using scanning electron microscopy (SEM) and mechanical compression tests. Small-Tailed Han sheep served as the animal model, and the two types of fusion cages were implanted in the C3/4 cervical segments and followed for up to 12 months. Imaging techniques, including X-ray, CT scans, and Micro CT, were used to observe the bone integration of the fusion cages. Hard tissue sections were used to assess osteogenic effects and bone integration. The range of motion (ROM) of the motion segments was evaluated using a biomechanical testing machine. Serum biochemical indicators and pathological analysis of major organs were conducted to assess biocompatibility. Results X-ray imaging showed that both the 3D-printed and chemical vapor deposition porous tantalum fusion cages maintained comparable average intervertebral disc heights. Due to the presence of metal artifacts, CT and Micro CT imaging could not effectively analyze bone integration. Histomorphology data indicated that both the 3D-printed and chemical vapor deposition porous tantalum fusion cages exhibited similar levels of bone contact and integration at 3, 6, and 12 months, with bone bridging observed at 12 months. Both groups of fusion cages demonstrated consistent mechanical stability across all time points. Serum biochemistry showed no abnormalities, and no significant pathological changes were observed in the heart, liver, spleen, lungs, and kidneys. Conclusion This study confirms that 3D-printed and chemical vapor deposition porous tantalum fusion cages exhibit comparable, excellent osteogenic effects and long-term biocompatibility. Additionally, 3D-printed porous tantalum fusion cages offer unique advantages in achieving complex structural designs, low-cost manufacturing, and personalized customization, providing robust scientific support for future clinical applications. The translational potential of this article The translational potential of this paper is to use 3D printed biomimetic trabecular porous tantalum spine fusion cage with bone trabecular structure and validating its feasibility in large animal models (sheep). This study provides a basis for further research into the clinical application of the 3D printed biomimetic trabecular porous tantalum spine fusion cage.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Jingzhou Yang
- Institute of Materials Research, Tsinghua Shenzhen International Graduate School, Tsinghua University, Guangdong, China
- Shenzhen Dazhou Medical Technology Co., Ltd., Guangdong, China
| | - Wentao Wan
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Qingqian Zhao
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Mingyuan Di
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Dachen Zhang
- Shenzhen Dazhou Medical Technology Co., Ltd., Guangdong, China
| | - Gang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Chao Chen
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Wei Zhang
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Hebei Medical University, Hebei, China
| | - Hanming Bian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Ye Tian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Lu Xue
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Zheng Wang
- Department of Orthopedics, No.1 Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qiulin Li
- Institute of Materials Research, Tsinghua Shenzhen International Graduate School, Tsinghua University, Guangdong, China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Farber SH, Oldham AJ, O'Neill LK, Sawa AGU, Ratliff AC, Doomi A, Pereira BDA, Uribe JS, Kelly BP, Turner JD. Optimization of 3D-printed titanium interbody cage design. Part 1: in vitro biomechanical study of subsidence. Spine J 2024:S1529-9430(24)01186-0. [PMID: 39694448 DOI: 10.1016/j.spinee.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND CONTEXT Cage subsidence is a complication of interbody fusion associated with poor clinical outcomes. 3D-printed titanium interbody cages allow for the alteration of features such as stiffness and porosity. However, the influence of these features on subsidence and their biological effects on fusion have not been rigorously evaluated. PURPOSE This 2-part study sought to determine how changes in 3D-printed titanium cage parameters affect subsidence using an in vitro bone model (Part 1) and biological fusion using an in vivo sheep model (Part 2). STUDY DESIGN Biomechanical foam block model. METHODS In Part 1 of this study, 9 implant types were tested (8 per implant type). The implant types included 7 3D-printed titanium interbody cages with various surface areas, porosities, and surface topographies, along with 1 standard polyetheretherketone (PEEK) cage and 1 solid titanium cage. Subsidence testing was performed in a standardized foam block model using 2 different densities of foam. Digital imaging correlation was used to determine the relative vertical displacement of the interbody cage-foam block construct. RESULTS Subsidence decreased as the surface contact area with the bone model increased (all p≤.01). Increased porous surface topography increased subsidence, while a nonporous surface significantly decreased subsidence (all p<.001). Subsidence did not differ based on changes in implant porosity (all p≥.35) or material property/modulus (all p≥.19). Subsidence was significantly decreased with the higher density foam (p<.001). The stiffness of the implant was affected by porosity (all p<.02) and smooth surface topography (p=.01) but not by lumen size (all p≥.15). Stiffness did not differ between porous titanium and PEEK implants (p=.96), which were both less stiff than solid titanium implants (both p<.001). Surface area negatively correlated with subsidence (r=-0.786, p=.012) but was not correlated with stiffness (r=0.560, p=.12). CONCLUSIONS Implant surface area and surface topography greatly influenced interbody subsidence. Apparent stiffness, implant porosity, and material property did not affect subsidence in this in vitro model. Higher foam density also led to lower subsidence than low-density foam. Biological response in the in vivo setting likely also influences clinical subsidence, which is evaluated in the companion study (Part 2). CLINICAL SIGNIFICANCE This study provides valuable information regarding the new 3D-printed titanium technology. We showed that cage surface area and surface topography were the implant design parameters that had the greatest influence on the development of interbody subsidence. Moreover, bone mineral density was the factor that had the greatest effect on subsidence prevention. These data support patient optimization before surgery and emphasize the importance of endplate protection during surgery.
Collapse
Affiliation(s)
- S Harrison Farber
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Alton J Oldham
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Luke K O'Neill
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Anna G U Sawa
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Alexis C Ratliff
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Ahmed Doomi
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Bernardo de Andrada Pereira
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Juan S Uribe
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Brian P Kelly
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Jay D Turner
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| |
Collapse
|
5
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
6
|
Lombardo JA, Wills D, Wang T, Pelletier M, Farber SH, Kelly BP, Uribe JS, Turner JD, Vizesi F, Walsh WR. Optimization of 3D-printed titanium interbody cage design. Part 2: An in vivo study of spinal fusion in sheep. Spine J 2024:S1529-9430(24)01194-X. [PMID: 39662684 DOI: 10.1016/j.spinee.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/25/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND CONTEXT 3D-printed titanium cage designs can incorporate complex, porous features for bone ingrowth and a greater surface area for minimizing subsidence. In a companion study (Part 1), we determined that increased surface area leads to decreased subsidence; however, it remains unclear how increasing the cage surface area, resulting in a smaller graft aperture, influences fusion. PURPOSE We evaluated the effects of surface area of 3D-printed titanium cages and the use of autologous bone grafts on spinal fusion in sheep. STUDY DESIGN In vivo large animal study in 12 sheep. METHODS Interbody fusion was performed in 12 adult sheep at 24 levels (L2-3 and L4-5) using 3D-printed titanium cages with bilateral pedicle screw fixation. The cage designs varied in aperture: standard (low endplate surface area), small (medium endplate surface area), or none (high endplate surface area). These cages were packed with autologous iliac crest bone grafts (ICBG). A fourth group was implanted without bone grafts, using the no-aperture cage. Fusion was evaluated at 16 weeks via manual palpation, microcomputed tomography (microCT), histology, and histomorphometry. RESULTS Standard, small, and no-aperture cages packed with ICBG resulted in high fusion rates (80%, 100%, and 83%, respectively) at 16 weeks by manual palpation, and these results were not significantly different. Implantation without ICBG was associated with a significantly lower fusion rate (33%, p<.05). Histological, histomorphometry, and microCT results supported the findings obtained by manual palpation; findings from these modalities showed new bone spanning the vertebral endplates in the spines graded as fused by manual palpation. CONCLUSIONS Similar fusion results for standard, small, and no-aperture cage designs packed with ICBG suggest that aperture size does not influence fusion results in the sheep model. However, without ICBG grafting, fusion was significantly decreased, suggesting that graft material is necessary to predictably obtain fusion in this model. When the in vitro subsidence data (companion study, Part 1) is considered with the in vivo fusion data described here, porous 3D-printed titanium cages with maximal surface endplate contact and bone grafting perform favorably, resulting in low subsidence and high fusion rates. CLINICAL SIGNIFICANCE 3D-printed porous titanium interbody cages are novel devices with increasing clinical use. The study results show that the aperture size of the interbody cage did not influence fusion in a large animal (sheep) model. The use of bone graft material was the most important variable affecting fusion. These data suggest that the clinical use of 3D Ti cages without graft material should be avoided.
Collapse
Affiliation(s)
| | - Dan Wills
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Tian Wang
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Matthew Pelletier
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - S Harrison Farber
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Brian P Kelly
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Juan S Uribe
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Jay D Turner
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| | - Frank Vizesi
- SeaSpine Inc., Carlsbad, California; Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - William R Walsh
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Karavasili C, Young T, Francis J, Blanco J, Mancini N, Chang C, Bernstock JD, Connolly ID, Shankar GM, Traverso G. Local drug delivery challenges and innovations in spinal neurosurgery. J Control Release 2024; 376:1225-1250. [PMID: 39505215 DOI: 10.1016/j.jconrel.2024.10.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
The development of novel therapeutics in the field of spinal neurosurgery faces a litany of translational challenges. Achieving precise drug targeting within the confined spaces associated with the spinal cord, canal and vertebra requires the development of next generation delivery systems and devices. These must be capable of overcoming inherent barriers related to drug diffusion, whilst concurrently ensuring optimal drug distribution and retention. In this review, we provide an overview of the most recent advances in the therapeutic management of diseases and disorders affecting the spine, including systems and devices capable of releasing small molecules and biopharmaceuticals that help eliminate pain and restore the mechanical function and stability of the spine. We highlight material-based approaches and minimally invasive techniques that can be employed to provide control over drug release kinetics and improve retention. We also seek to explore how the newest advancements in nanotechnology, biomaterials, additive manufacturing technologies and imaging modalities can be employed in this translational pursuit. Finally, we discuss the landscape of clinical trials and recently approved products aimed at overcoming the complexities associated with drug delivery to the spine.
Collapse
Affiliation(s)
- Christina Karavasili
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Thomas Young
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua Francis
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Julianna Blanco
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Nicholas Mancini
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Charmaine Chang
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua D Bernstock
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ian D Connolly
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ganesh M Shankar
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
8
|
Xue B, Cao W, Zhao H, Zhang B, Liu J, Zhang H, Qi H, Zhou Q. Nanocrystal hydroxyapatite carrying traditional Chinese medicine for osteogenic differentiation. Colloids Surf B Biointerfaces 2024; 244:114186. [PMID: 39226849 DOI: 10.1016/j.colsurfb.2024.114186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Developing biomaterials with high osteogenic properties is crucial for achieving rapid bone repair and regeneration. This study focuses on the application of nanocrystal hydroxyapatite (nHAp) as a drug carrier to load Fu Yuan Huo Xue Decoction (FYHXD), a traditional Chinese medicine derived from Angelica sinensis, aiming to achieve improved efficacy in treating bone diseases such as osteoporosis. Through a facile physical adsorption approach, the FTIR result emerges new characteristic absorption peaks in the range of 1200-950 cm-1, proving the successful absorption of FYHXD onto the nHAp with a loading efficiency of 39.76 %. The modified nHAp exhibits a similar shape to the bone-derived hydroxyapatite nanocrystals, and their diameter increases slightly after modification. The drug release assay implies the rapid release of FYHXD in the first 10 h, followed by a continuously slow release within 70 h. The developed nHAp effectively enhances the adhesion, spreading, and proliferation of MC3T3-E1 cells in vitro, and significantly promotes their osteogenic differentiation, as indicated by increased alkaline phosphatase activity. Overall, the biocomposites hold great promise as active ingredients for integration into bone-associated biomaterials, offering the potential to stimulate spontaneous osteogenesis without requiring exogenous osteogenic factors.
Collapse
Affiliation(s)
- Bo Xue
- Department of Bone, Huangdao District Central Hospital, Qingdao 266555, China; Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, Shandong Engineering Research Center for Tissue Rehabilitation Materials and Devices, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Wen Cao
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hong Zhao
- Department of Bone, Huangdao District Central Hospital, Qingdao 266555, China
| | - Bingqiang Zhang
- Qingdao Key Laboratory of Cancer and Immune Cells, Qingdao Restore Medical Testing Laboratory Co., Ltd., Qingdao, Shandong 266111, PR China
| | - Jia Liu
- Department of Bone, Huangdao District Central Hospital, Qingdao 266555, China
| | - Huixin Zhang
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, Shandong Engineering Research Center for Tissue Rehabilitation Materials and Devices, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Qihui Zhou
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, Shandong Engineering Research Center for Tissue Rehabilitation Materials and Devices, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| |
Collapse
|
9
|
Li J, Sun Z, Wei X, Tan Q, He X. Effect of Structure on Osteogenesis of Bone Scaffold: Simulation Analysis Based on Mechanobiology and Animal Experiment Verification. Bioengineering (Basel) 2024; 11:1120. [PMID: 39593780 PMCID: PMC11592375 DOI: 10.3390/bioengineering11111120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Porous scaffolds, whose mechanical and biological properties are greatly affected by structure, are new treatments for bone defects. Since bone repair is related to biomechanics, analyzing the osteogenesis in scaffolds based on mechanical stimulation may become a more effective method than traditional biological experiments. A tissue regeneration algorithm based on mechanical regulation theory was implemented in this study to evaluate the osteogenesis of classical scaffolds (Gyroid, I-WP, and Diamond). In vivo experiments were used to verify and supplement the simulation results. Different approaches to describing osteogenesis were discussed. Bone formation was more obvious inside the Gyroid scaffold and outside the I-WP scaffold, while the new bone was more sufficient and evenly distributed in the Diamond scaffold. The osteogenesis pattern of the bone scaffold in the simulation analysis was consistent with the results of animal experiments, and the bone volume calculated by the tissue fraction threshold method and the elastic modulus threshold method was very similar to the in vivo experiment. The mechanical responses mediated by structure affect the osteogenesis of bone scaffolds. This study provided and confirmed a simulation analysis method based on mechanical regulation theory, which is more efficient and economical for analyzing tissue healing in bioengineering.
Collapse
Affiliation(s)
- Jialiang Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Zhongwei Sun
- Jiangsu Key Laboratory of Engineering Mechanics, School of Civil Engineering, Southeast University, Nanjing 210096, China;
| | - Xinyu Wei
- Department of Health Management, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710014, China;
| | - Qinghua Tan
- Department of Orthopedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710014, China; (Q.T.); (X.H.)
| | - Xijing He
- Department of Orthopedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710014, China; (Q.T.); (X.H.)
| |
Collapse
|
10
|
Yao X, Gong Z, Yin W, Li H, Douroumis D, Huang L, Li H. Islet cell spheroids produced by a thermally sensitive scaffold: a new diabetes treatment. J Nanobiotechnology 2024; 22:657. [PMID: 39456025 PMCID: PMC11515210 DOI: 10.1186/s12951-024-02891-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The primary issues in treating type 1 diabetes mellitus (T1DM) through the transplantation of healthy islets or islet β-cells are graft rejection and a lack of available donors. Currently, the majority of approaches use cell encapsulation technology and transplant replacement cells that can release insulin to address transplant rejection and donor shortages. However, existing encapsulation materials merely serve as carriers for islet cell growth. A new treatment approach for T1DM could be developed by creating a smart responsive material that encourages the formation of islet cell spheroids to replicate their 3D connections in vivo and controls the release of insulin aggregates. In this study, we used microfluidics to create thermally sensitive porous scaffolds made of poly(N-isopropyl acrylamide)/graphene oxide (PNIPAM/GO). The material was carefully shrunk under near-infrared light, enriched with mouse insulinoma pancreatic β cells (β-TC-6 cells), encapsulated, and cultivated to form 3D cell spheroids. The controlled contraction of the thermally responsive porous scaffold regulated insulin release from the spheroids, demonstrated using the glucose-stimulated insulin release assay (GSIS), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence assay. Eventually, implantation of the spheroids into C57BL/6 N diabetic mice enhanced the therapeutic effect, potentially offering a novel approach to the management of T1DM.
Collapse
Affiliation(s)
- Xueting Yao
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, Zhejiang, 325000, P. R. China
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, P. R. China
| | - Zehua Gong
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hanbing Li
- Department of Pharmaceutical Sciences, Institute of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Dennis Douroumis
- Centre for Research Innovation, CRI, University of Greenwich, Kent, ME4 4TB, UK
| | - Lijiang Huang
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China.
| | - Huaqiong Li
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, Zhejiang, 325000, P. R. China.
| |
Collapse
|
11
|
Mathey E, Pelletier MH, Walsh WR, Gall K, Carpenter D. Implant Strength Contributes to the Osseointegration Strength of Porous Metallic Materials. J Biomech Eng 2024; 146:101005. [PMID: 38668718 DOI: 10.1115/1.4065405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Indexed: 05/14/2024]
Abstract
Creating the optimal environment for effective and long term osseointegration is a heavily researched and sought-after design criteria for orthopedic implants. A validated multimaterial finite element (FE) model was developed to replicate and understand the results of an experimental in vivo push-out osseointegration model. The FE model results closely predicted global force (at 0.5 mm) and stiffness for the 50-90% porous implants with an r2 of 0.97 and 0.98, respectively. In addition, the FE global force at 0.5 mm showed a correlation to the maximum experimental forces with an r2 of 0.90. The highest porosity implants (80-90%) showed lower stiffnesses and more equitable load sharing but also failed at lower a global force level than the low porosity implants (50-70%). The lower strength of the high porosity implants caused premature plastic deformation of the implant itself during loading as well as significant deformations in the ingrown and surrounding bone, resulting in lower overall osseointegration strength, consistent with experimental measurements. The lower porosity implants showed a balance of sufficient bony ingrowth to support osseointegration strength coupled with implant mechanical properties to circumvent significant implant plasticity and collapse under the loading conditions. Together, the experimental and finite element modeling results support an optimal porosity in the range of 60-70% for maximizing osseointegration with current structure and loading.
Collapse
Affiliation(s)
- Elizabeth Mathey
- Department of Mechanical Engineering, University of Colorado Denver, 1200 Larimer St, Denver, CO 80204
| | - Matthew H Pelletier
- Prince of Wales Clinical School UNSW Sydney, Surgical and Orthopaedic Research Laboratories (SORL), Kensington 2031, Australia
| | - William R Walsh
- Prince of Wales Clinical School UNSW Sydney, Surgical and Orthopaedic Research Laboratories (SORL), Kensington 2031, Australia
| | - Ken Gall
- Pratt School of Engineering, Duke University, Durham, NC 27708
| | - Dana Carpenter
- Department of Mechanical Engineering, University of Colorado Denver, Denver, CO 80217-3364
| |
Collapse
|
12
|
Hitchon S, Soltanmohammadi P, Milner JS, Holdsworth D, Willing R. Porous versus solid shoulder implants in humeri of different bone densities: A finite element analysis. J Orthop Res 2024; 42:1897-1906. [PMID: 38520665 DOI: 10.1002/jor.25840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
Porous metallic prosthesis components can now be manufactured using additive manufacturing techniques, and may prove beneficial for promoting bony ingrowth, for accommodating drug delivery systems, and for reducing stress shielding. Using finite element modeling techniques, 36 scenarios (three porous stems, three bone densities, and four held arm positions) were analysed to assess the viability of porous humeral stems for use in total shoulder arthroplasty, and their resulting mechanobiological impact on the surrounding humerus bone. All three porous stems were predicted to experience stresses below the yield strength of Ti6Al4V (880 MPa) and to be capable of withstanding more than 10 million cycles of each loading scenario before failure. There was an indication that within an 80 mm region of the proximal humerus, there would be a reduction in bone resorption as stem porosity increased. Overall, this study shows promise that these porous structures are mechanically viable for incorporation into permanent shoulder prostheses to combat orthopedic infections.
Collapse
Affiliation(s)
- Sydney Hitchon
- School of Biomedical Engineering, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
| | | | - Jaques S Milner
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - David Holdsworth
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Ryan Willing
- School of Biomedical Engineering, Western University, London, Ontario, Canada
- Bone and Joint Institute, Western University, London, Ontario, Canada
- Department of Mechanical and Materials Engineering, Western University, London, Ontario, Canada
| |
Collapse
|
13
|
Ma B, Chen F, Liu X, Zhang Y, Gou S, Meng Q, Liu P, Cai K. Modified Titanium Implants Satisfy the Demands of Diabetic Osseointegration via Sequential Regulation of Macrophages and Mesenchymal Stem Cells. Adv Healthc Mater 2024:e2401556. [PMID: 39138979 DOI: 10.1002/adhm.202401556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/21/2024] [Indexed: 08/15/2024]
Abstract
The application of titanium (Ti) implants for patients with diabetes mellitus (DM) is still facing a significant challenge due to obstacles such as hyperglycemia, reactive oxygen species (ROS), and chronic inflammation, which hinders osseointegration. To address this issue, a Ti implant with dual functions of regulating polarization of macrophages and facilitating osseointergration is developed via hydrothermal reaction and hydrogel coating. The reactive oxygen species (ROS) and glucose (Glu) responsive hydrogel coating can locally deliver adenosine (ADO) in the early stage of implantation. The controlled release of ADO regulated the phenotype of macrophages, restored oxidative balance, and enhanced mitochondrial function during the early stages of implantation. Subsequently, strontium (Sr) ions will be released to promote osteogenic differentiation and proliferation of mesenchymal stem cells (MSCs), as the hydrogel coating degraded. It eventually leads to bone reconstruction during the late stages, aligning with the biological cascade of bone healing. The modified Ti implants showed effective osteogenesis for bone defects in DM patients, shedding light on the design and biological mechanisms of surface modification. This research offers promising potential for improving the treatment of bone-related complications in diabetic patients.
Collapse
Affiliation(s)
- Bo Ma
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Fangye Chen
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xin Liu
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yang Zhang
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shuangquan Gou
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qianxiang Meng
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Peng Liu
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
14
|
Xu J, Bao G, Jia B, Wang M, Wen P, Kan T, Zhang S, Liu A, Tang H, Yang H, Yue B, Dai K, Zheng Y, Qu X. An adaptive biodegradable zinc alloy with bidirectional regulation of bone homeostasis for treating fractures and aged bone defects. Bioact Mater 2024; 38:207-224. [PMID: 38756201 PMCID: PMC11096722 DOI: 10.1016/j.bioactmat.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
Healing of fractures or bone defects is significantly hindered by overactivated osteoclasts and inhibited osteogenesis in patients with abnormal bone metabolism. Current clinical approaches using titanium alloys or stainless steel provide mechanical support but have no biological effects on bone regeneration. Therefore, designing and fabricating degradable metal materials with sufficient mechanical strength and bidirectional regulation of both osteoblasts and osteoclasts is a substantial challenge. Here, this study first reported an adaptive biodegradable Zn-0.8 Mg alloy with bidirectional regulation of bone homeostasis, which promotes osteogenic differentiation by activating the Pi3k/Akt pathway and inhibits osteoclast differentiation by inhibiting the GRB2/ERK pathway. The anti-osteolytic ability of the Zn-0.8 Mg alloy was verified in a mouse calvarial osteolysis model and its suitability for internal fracture fixation with high-strength screws was confirmed in the rabbit femoral condyle fracture model. Furthermore, in an aged postmenopausal rat femoral condyle defect model, 3D printed Zn-0.8 Mg scaffolds promoted excellent bone regeneration through adaptive structures with good mechanical properties and bidirectionally regulated bone metabolism, enabling personalized bone defect repair. These findings demonstrate the substantial potential of the Zn-0.8 Mg alloy for treating fractures or bone defects in patients with aberrant bone metabolism.
Collapse
Affiliation(s)
- Jialian Xu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Guo Bao
- Laboratory Animal centre, National Research Institute for Family Planning, Beijing, 100081, China
| | - Bo Jia
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Peng Wen
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Tianyou Kan
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Aobo Liu
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Kerong Dai
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| |
Collapse
|
15
|
Lewandrowski KU, Vira S, Elfar JC, Lorio MP. Advancements in Custom 3D-Printed Titanium Interbody Spinal Fusion Cages and Their Relevance in Personalized Spine Care. J Pers Med 2024; 14:809. [PMID: 39202002 PMCID: PMC11355268 DOI: 10.3390/jpm14080809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
3D-printing technology has revolutionized spinal implant manufacturing, particularly in developing personalized and custom-fit titanium interbody fusion cages. These cages are pivotal in supporting inter-vertebral stability, promoting bone growth, and restoring spinal alignment. This article reviews the latest advancements in 3D-printed titanium interbody fusion cages, emphasizing their relevance in modern personalized surgical spine care protocols applied to common clinical scenarios. Furthermore, the authors review the various printing and post-printing processing technologies and discuss how engineering and design are deployed to tailor each type of implant to its patient-specific clinical application, highlighting how anatomical and biomechanical considerations impact their development and manufacturing processes to achieve optimum osteoinductive and osteoconductive properties. The article further examines the benefits of 3D printing, such as customizable geometry and porosity, that enhance osteointegration and mechanical compatibility, offering a leap forward in patient-specific solutions. The comparative analysis provided by the authors underscores the unique challenges and solutions in designing cervical, and lumbar spine implants, including load-bearing requirements and bioactivity with surrounding bony tissue to promote cell attachment. Additionally, the authors discuss the clinical outcomes associated with these implants, including the implications of improvements in surgical precision on patient outcomes. Lastly, they address strategies to overcome implementation challenges in healthcare facilities, which often resist new technology acquisitions due to perceived cost overruns and preconceived notions that hinder potential savings by providing customized surgical implants with the potential for lower complication and revision rates. This comprehensive review aims to provide insights into how modern 3D-printed titanium interbody fusion cages are made, explain quality standards, and how they may impact personalized surgical spine care.
Collapse
Affiliation(s)
- Kai-Uwe Lewandrowski
- Center for Advanced Spine Care of Southern Arizona, Division Personalized Pain Research and Education, Tucson, AZ 85712, USA
- Department of Orthopaedics, Fundación Universitaria Sanitas Bogotá, Bogotá 111321, Colombia
| | - Shaleen Vira
- Orthopedic and Sports Medicine Institute, Banner-University Tucson Campus, 755 East McDowell Road, Floor 2, Phoenix, AZ 85006, USA;
| | - John C. Elfar
- Department of Orthopaedic Surgery, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Morgan P. Lorio
- Advanced Orthopedics, 499 East Central Parkway, Altamonte Springs, FL 32701, USA;
- Orlando College of Osteopathic Medicine, Orlando, FL 34787, USA
| |
Collapse
|
16
|
Ma C, de Barros NR, Zheng T, Gomez A, Doyle M, Zhu J, Nanda HS, Li X, Khademhosseini A, Li B. 3D Printing and Surface Engineering of Ti6Al4V Scaffolds for Enhanced Osseointegration in an In Vitro Study. Biomimetics (Basel) 2024; 9:423. [PMID: 39056864 PMCID: PMC11274417 DOI: 10.3390/biomimetics9070423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Ti6Al4V superalloy is recognized as a good candidate for bone implants owing to its biocompatibility, corrosion resistance, and high strength-to-weight ratio. While dense metal implants are associated with stress shielding issues due to the difference in densities, stiffness, and modulus of elasticity compared to bone tissues, the surface of the implant/scaffold should mimic the properties of the bone of interest to assure a good integration with a strong interface. In this study, we investigated the additive manufacturing of porous Ti6Al4V scaffolds and coating modification for enhanced osteoconduction using osteoblast cells. The results showed the successful fabrication of porous Ti6Al4V scaffolds with adequate strength. Additionally, the surface treatment with NaOH and Dopamine Hydrochloride (DOPA) promoted the formation of Dopamine Hydrochloride (DOPA) coating with an optimized coating process, providing an environment that supports higher cell viability and growth compared to the uncoated Ti6Al4V scaffolds, as demonstrated by the higher proliferation ratios observed from day 1 to day 29. These findings bring valuable insights into the surface modification of 3D-printed scaffolds for improved osteoconduction through the coating process in solutions.
Collapse
Affiliation(s)
- Changyu Ma
- Autonomy Research Center for STEAHM, California State University Northridge, Northridge, CA 91324, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | | | - Tianqi Zheng
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Gomez
- Autonomy Research Center for STEAHM, California State University Northridge, Northridge, CA 91324, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Marshall Doyle
- Autonomy Research Center for STEAHM, California State University Northridge, Northridge, CA 91324, USA
| | - Jianhao Zhu
- Autonomy Research Center for STEAHM, California State University Northridge, Northridge, CA 91324, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Himansu Sekhar Nanda
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
- Discipline of Mechanical Engineering, Indian Institute of Information Technology, Design and Manufacturing, Jabalpur 482005, India
| | - Xiaochun Li
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Bingbing Li
- Autonomy Research Center for STEAHM, California State University Northridge, Northridge, CA 91324, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| |
Collapse
|
17
|
Chen Y, Luo Z, Meng W, Liu K, Chen Q, Cai Y, Ding Z, Huang C, Zhou Z, Jiang M, Zhou L. Decoding the "Fingerprint" of Implant Materials: Insights into the Foreign Body Reaction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310325. [PMID: 38191783 DOI: 10.1002/smll.202310325] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Foreign body reaction (FBR) is a prevalent yet often overlooked pathological phenomenon, particularly within the field of biomedical implantation. The presence of FBR poses a heavy burden on both the medical and socioeconomic systems. This review seeks to elucidate the protein "fingerprint" of implant materials, which is generated by the physiochemical properties of the implant materials themselves. In this review, the activity of macrophages, the formation of foreign body giant cells (FBGCs), and the development of fibrosis capsules in the context of FBR are introduced. Additionally, the relationship between various implant materials and FBR is elucidated in detail, as is an overview of the existing approaches and technologies employed to alleviate FBR. Finally, the significance of implant components (metallic materials and non-metallic materials), surface CHEMISTRY (charge and wettability), and physical characteristics (topography, roughness, and stiffness) in establishing the protein "fingerprint" of implant materials is also well documented. In conclusion, this review aims to emphasize the importance of FBR on implant materials and provides the current perspectives and approaches in developing implant materials with anti-FBR properties.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zeyu Luo
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weikun Meng
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiqing Chen
- Department of Ultrasound, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Yongrui Cai
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zichuan Ding
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chao Huang
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongke Zhou
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Jiang
- Emergency and Trauma Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liqiang Zhou
- MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
18
|
Qi J, Yu M, Liu Y, Zhang J, Li X, Ma Z, Sun T, Liu S, Qiu Y. Polydopamine-Coated Copper-Doped Co 3O 4 Nanosheets Rich in Oxygen Vacancy on Titanium and Multimodal Synergistic Antibacterial Study. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2019. [PMID: 38730825 PMCID: PMC11084916 DOI: 10.3390/ma17092019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024]
Abstract
Medical titanium-based (Ti-based) implants in the human body are prone to infection by pathogenic bacteria, leading to implantation failure. Constructing antibacterial nanocoatings on Ti-based implants is one of the most effective strategies to solve bacterial contamination. However, single antibacterial function was not sufficient to efficiently kill bacteria, and it is necessary to develop multifunctional antibacterial methods. This study modifies medical Ti foils with Cu-doped Co3O4 rich in oxygen vacancies, and improves their biocompatibility by polydopamine (PDA/Cu-Ov-Co3O4). Under near-infrared (NIR) irradiation, nanocoatings can generate •OH and 1O2 due to Cu+ Fenton-like activity and a photodynamic effect of Cu-Ov-Co3O4, and the total reactive oxygen species (ROS) content inside bacteria significantly increases, causing oxidative stress of bacteria. Further experiments prove that the photothermal process enhances the bacterial membrane permeability, allowing the invasion of ROS and metal ions, as well as the protein leakage. Moreover, PDA/Cu-Ov-Co3O4 can downregulate ATP levels and further reduce bacterial metabolic activity after irradiation. This coating exhibits sterilization ability against both Escherichia coli and Staphylococcus aureus with an antibacterial rate of ca. 100%, significantly higher than that of bare medical Ti foils (ca. 0%). Therefore, multifunctional synergistic antibacterial nanocoating will be a promising strategy for preventing bacterial contamination on medical Ti-based implants.
Collapse
Affiliation(s)
- Jinteng Qi
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China;
| | - Miao Yu
- Key Laboratory of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China (S.L.)
| | - Yi Liu
- Key Laboratory of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China (S.L.)
| | - Junting Zhang
- Key Laboratory of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China (S.L.)
| | - Xinyi Li
- Key Laboratory of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China (S.L.)
| | - Zhuo Ma
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China;
| | - Shaoqin Liu
- Key Laboratory of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China (S.L.)
| | - Yunfeng Qiu
- Key Laboratory of Microsystems and Microstructures Manufacturing, School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China (S.L.)
| |
Collapse
|
19
|
Kelly C, Adams SB. 3D Printing Materials and Technologies for Orthopaedic Applications. J Orthop Trauma 2024; 38:S9-S12. [PMID: 38502597 DOI: 10.1097/bot.0000000000002765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2024] [Indexed: 03/21/2024]
Abstract
SUMMARY 3D printing technologies have evolved tremendously over the last decade for uses in orthopaedic surgical applications, including being used to manufacture implants for spine, upper extremity, foot and ankle, oncologic, and traumatic reconstructions. Materials used for 3D-printed orthopaedic devices include metals, degradable and nondegradable polymers, and ceramic composites. There are 2 primary advantages for use of 3D printing technologies for orthopaedics: first, the ability to create complex porous lattices that allow for osseointegration and improved implant stability and second, the enablement of complex geometric designs allowing for patient-specific devices based on preoperative imaging. Given continually evolving technology, and the relatively early stage of the materials and 3D printers themselves, the possibilities for continued innovation in orthopaedics are great.
Collapse
Affiliation(s)
| | - Samuel B Adams
- Department of Orthopedic Surgery, Duke University Medical Center, Durham, NC
| |
Collapse
|
20
|
Köksal B, Kartal RB, Günay US, Durmaz H, Yildiz AA, Yildiz ÜH. Fabrication of gelatin-polyester based biocomposite scaffold via one-step functionalization of melt electrowritten polymer blends in aqueous phase. Int J Biol Macromol 2024; 265:130938. [PMID: 38493814 DOI: 10.1016/j.ijbiomac.2024.130938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The rapid manufacturing of biocomposite scaffold made of saturated-Poly(ε-caprolactone) (PCL) and unsaturated Polyester (PE) blends with gelatin and modified gelatin (NCO-Gel) is demonstrated. Polyester blend-based scaffold are fabricated with and without applying potential in the melt electrowriting system. Notably, the applied potential induces phase separation between PCL and PE and drives the formation of PE rich spots at the interface of electrowritten fibers. The objective of the current study is to control the phase separation between saturated and unsaturated polyesters occurring in the melt electro-writing process and utilization of this phenomenon to improve efficiency of biofunctionalization at the interface of scaffold via Aza-Michael addition reaction. Electron-deficient triple bonds of PE spots on the fibers exhibit good potential for the biofunctionalization via the aza-Michael addition reaction. PE spots are found to be pronounced in which blend compositions are PCL-PE as 90:10 and 75:25 %. The biofunctionalization of scaffold is monitored through CN bond formation appeared at 400 eV via X-ray photoelectron spectroscopy (XPS) and XPS chemical mapping. The described biofunctionalization methodology suggest avoiding use of multi-step chemical modification on additive manufacturing products and thereby rapid prototyping of functional polymer blend based scaffolds with enhanced biocompatibility and preserved mechanical properties. Additionally one-step additive manufacturing method eliminates side effects of toxic solvents and long modification steps during scaffold fabrication.
Collapse
Affiliation(s)
- Büşra Köksal
- Department of Chemistry, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey
| | | | - Ufuk Saim Günay
- Department of Chemistry, Istanbul Technical University, 34469 Maslak, İstanbul, Turkey
| | - Hakan Durmaz
- Department of Chemistry, Istanbul Technical University, 34469 Maslak, İstanbul, Turkey
| | - Ahu Arslan Yildiz
- Department of Bioengineering, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey
| | - Ümit Hakan Yildiz
- Department of Chemistry, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey; Department of Polymer Science and Engineering, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey.
| |
Collapse
|
21
|
Honda S, Fujibayashi S, Shimizu T, Yamaguchi S, Okuzu Y, Takaoka Y, Masuda S, Takemoto M, Kawai T, Otsuki B, Goto K, Matsuda S. Strontium-loaded 3D intramedullary nail titanium implant for critical-sized femoral defect in rabbits. J Biomed Mater Res B Appl Biomater 2024; 112:e35393. [PMID: 38385959 DOI: 10.1002/jbm.b.35393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/23/2024]
Abstract
The treatment of critical-sized bone defects has long been a major problem for surgeons. In this study, an intramedullary nail shaped three-dimensional (3D)-printed porous titanium implant that is capable of releasing strontium ions was developed through a simple and cost-effective surface modification technique. The feasibility of this implant as a stand-alone solution was evaluated using a rabbit's segmental diaphyseal as a defect model. The strontium-loaded implant exhibited a favorable environment for cell adhesion, and mechanical properties that were commensurate with those of a rabbit's cortical bone. Radiographic, biomechanical, and histological analyses revealed a significantly higher amount of bone ingrowth and superior bone-bonding strength in the strontium-loaded implant when compared to an untreated porous titanium implant. Furthermore, one-year histological observations revealed that the strontium-loaded implant preserved the native-like diaphyseal bone structure without failure. These findings suggest that strontium-releasing 3D-printed titanium implants have the clinical potential to induce the early and efficient repair of critical-sized, load-bearing bone defects.
Collapse
Affiliation(s)
- Shintaro Honda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shunsuke Fujibayashi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayoshi Shimizu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seiji Yamaguchi
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Aichi, Japan
| | - Yaichiro Okuzu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yusuke Takaoka
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Soichiro Masuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mitsuru Takemoto
- Department of Orthopaedic Surgery, Kyoto City Hospital, Kyoto, Japan
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Bungo Otsuki
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
22
|
Kechagias S, Theodoridis K, Broomfield J, Malpartida-Cardenas K, Reid R, Georgiou P, van Arkel RJ, Jeffers JRT. The effect of nodal connectivity and strut density within stochastic titanium scaffolds on osteogenesis. Front Bioeng Biotechnol 2023; 11:1305936. [PMID: 38107615 PMCID: PMC10721980 DOI: 10.3389/fbioe.2023.1305936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023] Open
Abstract
Modern orthopaedic implants use lattice structures that act as 3D scaffolds to enhance bone growth into and around implants. Stochastic scaffolds are of particular interest as they mimic the architecture of trabecular bone and can combine isotropic properties and adjustable structure. The existing research mainly concentrates on controlling the mechanical and biological performance of periodic lattices by adjusting pore size and shape. Still, less is known on how we can control the performance of stochastic lattices through their design parameters: nodal connectivity, strut density and strut thickness. To elucidate this, four lattice structures were evaluated with varied strut densities and connectivity, hence different local geometry and mechanical properties: low apparent modulus, high apparent modulus, and two with near-identical modulus. Pre-osteoblast murine cells were seeded on scaffolds and cultured in vitro for 28 days. Cell adhesion, proliferation and differentiation were evaluated. Additionally, the expression levels of key osteogenic biomarkers were used to assess the effect of each design parameter on the quality of newly formed tissue. The main finding was that increasing connectivity increased the rate of osteoblast maturation, tissue formation and mineralisation. In detail, doubling the connectivity, over fixed strut density, increased collagen type-I by 140%, increased osteopontin by 130% and osteocalcin by 110%. This was attributed to the increased number of acute angles formed by the numerous connected struts, which facilitated the organization of cells and accelerated the cell cycle. Overall, increasing connectivity and adjusting strut density is a novel technique to design stochastic structures which combine a broad range of biomimetic properties and rapid ossification.
Collapse
Affiliation(s)
- Stylianos Kechagias
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | | | - Joseph Broomfield
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Kenny Malpartida-Cardenas
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Ruth Reid
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Pantelis Georgiou
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Richard J. van Arkel
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | | |
Collapse
|
23
|
Lee SS, Du X, Smit T, Bissacco EG, Seiler D, de Wild M, Ferguson SJ. 3D-printed LEGO®-inspired titanium scaffolds for patient-specific regenerative medicine. BIOMATERIALS ADVANCES 2023; 154:213617. [PMID: 37678088 DOI: 10.1016/j.bioadv.2023.213617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/08/2023] [Accepted: 09/02/2023] [Indexed: 09/09/2023]
Abstract
Despite the recent advances in 3D-printing, it is often difficult to fabricate implants that optimally fit a defect size or shape. There are some approaches to resolve this issue, such as patient-specific implant/scaffold designs based on CT images of the patients, however, this process is labor-intensive and costly. Especially in developing countries, affordable treatment options are required, while still not excluding these patient groups from potential material and manufacturing advances. Here, a selective laser melting (SLM) 3D-printing strategy was used to fabricate a hierarchical, LEGO®-inspired Assemblable Titanium Scaffold (ATS) system, which can be manually assembled in any shape or size with ease. A surgeon can quickly create a scaffold that would fit to the defect right before the implantation during the surgery. Additionally, the direct inclusion of micro- and macroporous structures via 3D-printing, as well as a double acid-etched surface treatment (ST) in the ATS, ensure biocompatibility, sufficient nutrient flow, cell migration and enhanced osteogenesis. Three different structures were designed (non-porous:NP, semi-porous:SP, ultra-porous:UP), 3D-printed with the SLM technique and then surface treated for the ST groups. After analyzing characteristics of the ATS such as printing quality, surface roughness and interconnected porosity, mechanical testing and finite element analysis (FEA) demonstrated that individual and stacked ATS have sufficient mechanical properties to withstand loading in a physiological system. All ATS showed high cell viability, and the SP and UP groups demonstrated enhanced cell proliferation rates compared to the NP group. Furthermore, we also verified that cells were well-attached and spread on the porous structures and successful cell migration between the ATS units was seen in the case of assemblies. The UP and SP groups exhibited higher calcium deposition and RT-qPCR proved higher osteogenic gene expression compared to NP group. Finally, we demonstrate a number of possible medical applications that reveal the potential of the ATS through assembly.
Collapse
Affiliation(s)
- Seunghun S Lee
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| | - Xiaoyu Du
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Thijs Smit
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Elisa G Bissacco
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Daniel Seiler
- Institute for Medical Engineering and Medical Informatics IM(2), FHNW, Muttenz, Switzerland
| | - Michael de Wild
- Institute for Medical Engineering and Medical Informatics IM(2), FHNW, Muttenz, Switzerland
| | - Stephen J Ferguson
- Institute for Biomechanics, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Zhang Y, Cheng Z, Liu Z, Shen X, Cai C, Li M, Luo Z. Functionally Tailored Metal-Organic Framework Coatings for Mediating Ti Implant Osseointegration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303958. [PMID: 37705110 PMCID: PMC10582459 DOI: 10.1002/advs.202303958] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/24/2023] [Indexed: 09/15/2023]
Abstract
Owing to their mechanical resilience and non-toxicity, titanium implants are widely applied as the major treatment modality for the clinical intervention against bone fractures. However, the intrinsic bioinertness of Ti and its alloys often impedes the effective osseointegration of the implants, leading to severe adverse complications including implant loosening, detachment, and secondary bone damage. Consequently, new Ti implant engineering strategies are urgently needed to improve their osseointegration after implantation. Remarkably, metalorganic frameworks (MOFs) are a class of novel synthetic material consisting of coordinated metal species and organic ligands, which have demonstrated a plethora of favorable properties for modulating the interfacial properties of Ti implants. This review comprehensively summarizes the recent progress in the development of MOF-coated Ti implants and highlights their potential utility for modulating the bio-implant interface to improve implant osseointegration, of which the discussions are outlined according to their physical traits, chemical composition, and drug delivery capacity. A perspective is also provided in this review regarding the current limitations and future opportunities of MOF-coated Ti implants for orthopedic applications. The insights in this review may facilitate the rational design of more advanced Ti implants with enhanced therapeutic performance and safety.
Collapse
Affiliation(s)
- Yuan Zhang
- Joint Disease & Sport Medicine CentreDepartment of OrthopaedicsXinqiao HospitalArmy Medical UniversityChongqing400038China
| | - Zhuo Cheng
- School of Life ScienceChongqing UniversityChongqing400044China
| | - Zaiyang Liu
- Joint Disease & Sport Medicine CentreDepartment of OrthopaedicsXinqiao HospitalArmy Medical UniversityChongqing400038China
| | - Xinkun Shen
- Department of OrthopaedicsRuian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Chunyuan Cai
- Department of OrthopaedicsRuian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Menghuan Li
- School of Life ScienceChongqing UniversityChongqing400044China
| | - Zhong Luo
- School of Life ScienceChongqing UniversityChongqing400044China
| |
Collapse
|
25
|
Lerch RJ, Gulati A, Highlander PD. Revision of Subtalar Joint Arthrodesis: Considerations for Bone Grafting, Fixation Constructs, and Three-Dimensional Printing. Clin Podiatr Med Surg 2023; 40:633-648. [PMID: 37716742 DOI: 10.1016/j.cpm.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
Abstract
Subtalar joint arthrodesis is a commonly used procedure for numerous pathologic conditions in the foot and ankle. Although rarely performed in isolation, this procedure can provide successful resolution of various lower-extremity complaints. There are traditional approaches to isolated subtalar joint arthrodesis; however, when these fail, the authors recommend alternatives to enhance the success of revisional surgery. These include the use of intramedullary nailing, larger harvest of autograft, and metallic wedge.
Collapse
Affiliation(s)
- Ryan J Lerch
- The Reconstruction Institute, The Bellevue Hospital, 1400 West Main Street, Bellevue, OH 44811, USA
| | - Amar Gulati
- Progressive Feet, 611 South Carlin Springs Road, Suite 508, Arlington, VA 22204, USA
| | - Peter D Highlander
- The Reconstruction Institute, The Bellevue Hospital, 1400 West Main Street, Bellevue, OH 44811, USA.
| |
Collapse
|
26
|
Zhang Z, Zhang X, Zheng Z, Xin J, Han S, Qi J, Zhang T, Wang Y, Zhang S. Latest advances: Improving the anti-inflammatory and immunomodulatory properties of PEEK materials. Mater Today Bio 2023; 22:100748. [PMID: 37600350 PMCID: PMC10432209 DOI: 10.1016/j.mtbio.2023.100748] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
Excellent biocompatibility, mechanical properties, chemical stability, and elastic modulus close to bone tissue make polyetheretherketone (PEEK) a promising orthopedic implant material. However, biological inertness has hindered the clinical applications of PEEK. The immune responses and inflammatory reactions after implantation would interfere with the osteogenic process. Eventually, the proliferation of fibrous tissue and the formation of fibrous capsules would result in a loose connection between PEEK and bone, leading to implantation failure. Previous studies focused on improving the osteogenic properties and antibacterial ability of PEEK with various modification techniques. However, few studies have been conducted on the immunomodulatory capacity of PEEK. New clinical applications and advances in processing technology, research, and reports on the immunomodulatory capacity of PEEK have received increasing attention in recent years. Researchers have designed numerous modification techniques, including drug delivery systems, surface chemical modifications, and surface porous treatments, to modulate the post-implantation immune response to address the regulatory factors of the mechanism. These studies provide essential ideas and technical preconditions for the development and research of the next generation of PEEK biological implant materials. This paper summarizes the mechanism by which the immune response after PEEK implantation leads to fibrous capsule formation; it also focuses on modification techniques to improve the anti-inflammatory and immunomodulatory abilities of PEEK. We also discuss the limitations of the existing modification techniques and present the corresponding future perspectives.
Collapse
Affiliation(s)
- Zilin Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Xingmin Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Zhi Zheng
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Jingguo Xin
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Song Han
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Jinwei Qi
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Tianhui Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Yongjie Wang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| | - Shaokun Zhang
- Department of Spine Surgery, Center of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
- Jilin Engineering Research Center for Spine and Spinal Cord Injury, Changchun, 130021, China
| |
Collapse
|
27
|
Ma L, Zhou J, Wu Q, Luo G, Zhao M, Zhong G, Zheng Y, Meng X, Cheng S, Zhang Y. Multifunctional 3D-printed scaffolds eradiate orthotopic osteosarcoma and promote osteogenesis via microwave thermo-chemotherapy combined with immunotherapy. Biomaterials 2023; 301:122236. [PMID: 37506512 DOI: 10.1016/j.biomaterials.2023.122236] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 06/04/2023] [Accepted: 07/02/2023] [Indexed: 07/30/2023]
Abstract
Tumor recurrence and a lack of bone-tissue integration are two critical concerns in the surgical treatment of osteosarcoma. Thus, an advanced multifunctional therapeutic platform capable of simultaneously eliminating residual tumor cells and promoting bone regeneration is urgently needed for efficient osteosarcoma treatment. Herein, to thoroughly eliminate tumors and simultaneously promote bone regeneration, an intelligent multifunctional therapeutic scaffold has been engineered by integrating microwave-responsive zeolitic imidazolate framework 8 (ZIF-8) nanomaterials loaded with a chemotherapeutic drug and an immune checkpoint inhibitor onto 3D-printed titanium scaffolds. The constructed scaffold features distinct microwave-thermal sensitization and tumor microenvironment-responsive characteristics, which can induce tumor immunogenic death by microwave hyperthermia and chemotherapy. Orthotopic implantation of the nanocomposite scaffold results in an enhanced immune response against osteosarcoma that may effectively inhibit tumor recurrence through synergistic immunotherapy. During long-term implantation, the zinc ions released from the degradation of ZIF-8 can induce the osteogenic differentiation of stem cells. The porous structure and mechanical properties of the 3D-printed titanium scaffolds provide a structural microenvironment for bone regeneration. This study provides a paradigm for the design of multifunctional microwave-responsive composite scaffolds for use as a therapy for osteosarcoma, which could lead to improved strategies for the treatment of the disease.
Collapse
Affiliation(s)
- Limin Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Jielong Zhou
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Qiong Wu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guowen Luo
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Manzhi Zhao
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Guoqing Zhong
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, PR China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| | - Shi Cheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, PR China.
| |
Collapse
|
28
|
Zhou H, Ye S, Xu M, Hao L, Chen J, Fang Z, Guo K, Chen Y, Wang L. Dynamic surface adapts to multiple service stages by orchestrating responsive polymers and functional peptides. Biomaterials 2023; 301:122200. [PMID: 37423184 DOI: 10.1016/j.biomaterials.2023.122200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
Control over the implant surface functions is highly desirable to enhance tissue healing outcomes but has remained unexplored to adapt to the different service stages. In the present study, we develop a smart titanium surface by orchestrating thermoresponsive polymer and antimicrobial peptide to enable dynamic adaptation to the implantation stage, normal physiological stage and bacterial infection stage. The optimized surface inhibited bacterial adhesion and biofilm formation during surgical implantation, while promoted osteogenesis in the physiological stage. The further temperature increase driven by bacterial infection induced polymer chain collapse to expose antimicrobial peptides by rupturing bacterial membranes, as well as protect the adhered cells from the hostile environment of infection and abnormal temperature. The engineered surface could inhibit infection and promote tissue healing in rabbit subcutaneous and bone defect infection models. This strategy enables the possibility to create a versatile surface platform to balance bacteria/cell-biomaterial interactions at different service stages of implants that has not been achieved before.
Collapse
Affiliation(s)
- Haiyan Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Silin Ye
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Mingjian Xu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Lihui Hao
- Department of Stomatology, Xingtai Medical College, Xingtai 054000, China
| | - Junjian Chen
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China.
| | - Zhou Fang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Kunzhong Guo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China.
| | - Lin Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
29
|
Moiduddin K, Mian SH, Elseufy SM, Alkhalefah H, Ramalingam S, Sayeed A. Polyether-Ether-Ketone (PEEK) and Its 3D-Printed Quantitate Assessment in Cranial Reconstruction. J Funct Biomater 2023; 14:429. [PMID: 37623673 PMCID: PMC10455463 DOI: 10.3390/jfb14080429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Three-dimensional (3D) printing, medical imaging, and implant design have all advanced significantly in recent years, and these developments may change how modern craniomaxillofacial surgeons use patient data to create tailored treatments. Polyether-ether-ketone (PEEK) is often seen as an attractive option over metal biomaterials in medical uses, but a solid PEEK implant often leads to poor osseointegration and clinical failure. Therefore, the objective of this study is to demonstrate the quantitative assessment of a custom porous PEEK implant for cranial reconstruction and to evaluate its fitting accuracy. The research proposes an efficient process for designing, fabricating, simulating, and inspecting a customized porous PEEK implant. In this study, a CT scan is utilized in conjunction with a mirrored reconstruction technique to produce a skull implant. In order to foster cell proliferation, the implant is modified into a porous structure. The implant's strength and stability are examined using finite element analysis. Fused filament fabrication (FFF) is utilized to fabricate the porous PEEK implants, and 3D scanning is used to test its fitting accuracy. The results of the biomechanical analysis indicate that the highest stress observed was approximately 61.92 MPa, which is comparatively low when compared with the yield strength and tensile strength of the material. The implant fitting analysis demonstrates that the implant's variance from the normal skull is less than 0.4436 mm, which is rather low given the delicate anatomy of the area. The results of the study demonstrate the implant's endurance while also increasing the patient's cosmetic value.
Collapse
Affiliation(s)
- Khaja Moiduddin
- Advanced Manufacturing Institute, King Saud University, Riyadh 11421, Saudi Arabia
| | - Syed Hammad Mian
- Advanced Manufacturing Institute, King Saud University, Riyadh 11421, Saudi Arabia
| | | | - Hisham Alkhalefah
- Advanced Manufacturing Institute, King Saud University, Riyadh 11421, Saudi Arabia
| | - Sundar Ramalingam
- Department of Oral and Maxillofacial Surgery, College of Dentistry and Dental University Hospital, King Saud University Medical City, Riyadh 11545, Saudi Arabia
| | - Abdul Sayeed
- Department of Mechanical Engineering, College of Engineering, King Saud University, Riyadh 11421, Saudi Arabia
| |
Collapse
|
30
|
Yarali E, Zadpoor AA, Staufer U, Accardo A, Mirzaali MJ. Auxeticity as a Mechanobiological Tool to Create Meta-Biomaterials. ACS APPLIED BIO MATERIALS 2023; 6:2562-2575. [PMID: 37319268 PMCID: PMC10354748 DOI: 10.1021/acsabm.3c00145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Mechanical and morphological design parameters, such as stiffness or porosity, play important roles in creating orthopedic implants and bone substitutes. However, we have only a limited understanding of how the microarchitecture of porous scaffolds contributes to bone regeneration. Meta-biomaterials are increasingly used to precisely engineer the internal geometry of porous scaffolds and independently tailor their mechanical properties (e.g., stiffness and Poisson's ratio). This is motivated by the rare or unprecedented properties of meta-biomaterials, such as negative Poisson's ratios (i.e., auxeticity). It is, however, not clear how these unusual properties can modulate the interactions of meta-biomaterials with living cells and whether they can facilitate bone tissue engineering under static and dynamic cell culture and mechanical loading conditions. Here, we review the recent studies investigating the effects of the Poisson's ratio on the performance of meta-biomaterials with an emphasis on the relevant mechanobiological aspects. We also highlight the state-of-the-art additive manufacturing techniques employed to create meta-biomaterials, particularly at the micrometer scale. Finally, we provide future perspectives, particularly for the design of the next generation of meta-biomaterials featuring dynamic properties (e.g., those made through 4D printing).
Collapse
Affiliation(s)
- Ebrahim Yarali
- Department
of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials
Engineering, Delft University of Technology
(TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Maritime
and Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Amir A. Zadpoor
- Department
of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials
Engineering, Delft University of Technology
(TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Urs Staufer
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Maritime
and Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Angelo Accardo
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Maritime
and Materials Engineering, Delft University
of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Mohammad J. Mirzaali
- Department
of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials
Engineering, Delft University of Technology
(TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| |
Collapse
|
31
|
Kovács ÁÉ, Csernátony Z, Csámer L, Méhes G, Szabó D, Veres M, Braun M, Harangi B, Serbán N, Zhang L, Falk G, Soósné Horváth H, Manó S. Comparative Analysis of Bone Ingrowth in 3D-Printed Titanium Lattice Structures with Different Patterns. MATERIALS (BASEL, SWITZERLAND) 2023; 16:ma16103861. [PMID: 37241487 DOI: 10.3390/ma16103861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023]
Abstract
In this study, metal 3D printing technology was used to create lattice-shaped test specimens of orthopedic implants to determine the effect of different lattice shapes on bone ingrowth. Six different lattice shapes were used: gyroid, cube, cylinder, tetrahedron, double pyramid, and Voronoi. The lattice-structured implants were produced from Ti6Al4V alloy using direct metal laser sintering 3D printing technology with an EOS M290 printer. The implants were implanted into the femoral condyles of sheep, and the animals were euthanized 8 and 12 weeks after surgery. To determine the degree of bone ingrowth for different lattice-shaped implants, mechanical, histological, and image processing tests on ground samples and optical microscopic images were performed. In the mechanical test, the force required to compress the different lattice-shaped implants and the force required for a solid implant were compared, and significant differences were found in several instances. Statistically evaluating the results of our image processing algorithm, it was found that the digitally segmented areas clearly consisted of ingrown bone tissue; this finding is also supported by the results of classical histological processing. Our main goal was realized, so the bone ingrowth efficiencies of the six lattice shapes were ranked. It was found that the gyroid, double pyramid, and cube-shaped lattice implants had the highest degree of bone tissue growth per unit time. This ranking of the three lattice shapes remained the same at both 8 and 12 weeks after euthanasia. In accordance with the study, as a side project, a new image processing algorithm was developed that proved suitable for determining the degree of bone ingrowth in lattice implants from optical microscopic images. Along with the cube lattice shape, whose high bone ingrowth values have been previously reported in many studies, it was found that the gyroid and double pyramid lattice shapes produced similarly good results.
Collapse
Affiliation(s)
- Ágnes Éva Kovács
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Zoltán Csernátony
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Loránd Csámer
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Dániel Szabó
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Mihály Veres
- Isotoptech Private Limited Company, H-4026 Debrecen, Hungary
| | - Mihály Braun
- Isotoptech Private Limited Company, H-4026 Debrecen, Hungary
| | - Balázs Harangi
- Department of Data Science and Visualization, Faculty of Informatics, University of Debrecen, H-4028 Debrecen, Hungary
| | - Norbert Serbán
- Department of Data Science and Visualization, Faculty of Informatics, University of Debrecen, H-4028 Debrecen, Hungary
| | - Lei Zhang
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - György Falk
- Varinex Private Limited Company, H-1141 Budapest, Hungary
| | - Hajnalka Soósné Horváth
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Sándor Manó
- Laboratory of Biomechanics, Department of Orthopaedic Surgery, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
32
|
Casanova EA, Rodriguez-Palomo A, Stähli L, Arnke K, Gröninger O, Generali M, Neldner Y, Tiziani S, Dominguez AP, Guizar-Sicairos M, Gao Z, Appel C, Nielsen LC, Georgiadis M, Weber FE, Stark W, Pape HC, Cinelli P, Liebi M. SAXS imaging reveals optimized osseointegration properties of bioengineered oriented 3D-PLGA/aCaP scaffolds in a critical size bone defect model. Biomaterials 2023; 294:121989. [PMID: 36628888 DOI: 10.1016/j.biomaterials.2022.121989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 12/01/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023]
Abstract
Healing large bone defects remains challenging in orthopedic surgery and is often associated with poor outcomes and complications. A major issue with bioengineered constructs is achieving a continuous interface between host bone and graft to enhance biological processes and mechanical stability. In this study, we have developed a new bioengineering strategy to produce oriented biocompatible 3D PLGA/aCaP nanocomposites with enhanced osseointegration. Decellularized scaffolds -containing only extracellular matrix- or scaffolds seeded with adipose-derived mesenchymal stromal cells were tested in a mouse model for critical size bone defects. In parallel to micro-CT analysis, SAXS tensor tomography and 2D scanning SAXS were employed to determine the 3D arrangement and nanostructure within the critical-sized bone. Both newly developed scaffold types, seeded with cells or decellularized, showed high osseointegration, higher bone quality, increased alignment of collagen fibers and optimal alignment and size of hydroxyapatite minerals.
Collapse
Affiliation(s)
- Elisa A Casanova
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | | | - Lisa Stähli
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Kevin Arnke
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Gröninger
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Melanie Generali
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Yvonne Neldner
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Simon Tiziani
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Ana Perez Dominguez
- Oral Biotechnology and Bioengineering, Department of Cranio-Maxillofacial and Oral Surgery, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | | | - Zirui Gao
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | - Christian Appel
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | - Leonard C Nielsen
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden
| | - Marios Georgiadis
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| | - Franz E Weber
- Oral Biotechnology and Bioengineering, Department of Cranio-Maxillofacial and Oral Surgery, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Wendelin Stark
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Hans-Christoph Pape
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Paolo Cinelli
- Department of Trauma Surgery, University of Zurich, University Hospital Zurich, Zurich, Switzerland; Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland.
| | - Marianne Liebi
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden; Centre for X-ray Analytics, Swiss Federal Laboratories for Materials Science and Technology (EMPA), St. Gallen, Switzerland
| |
Collapse
|
33
|
Han J, Park S, Kim JE, Park B, Hong Y, Lim JW, Jeong S, Son H, Kim HB, Seonwoo H, Jang KJ, Chung JH. Development of a Scaffold-on-a-Chip Platform to Evaluate Cell Infiltration and Osteogenesis on the 3D-Printed Scaffold for Bone Regeneration. ACS Biomater Sci Eng 2023; 9:968-977. [PMID: 36701173 DOI: 10.1021/acsbiomaterials.2c01367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Developing a scaffold for efficient and functional bone regeneration remains challenging. To accomplish this goal, a "scaffold-on-a-chip" device was developed as a platform to aid with the evaluation process. The device mimics a microenvironment experienced by a transplanted bone scaffold. The device contains a circular space at the center for scaffold insert and microfluidic channel that encloses the space. Such a design allows for monitoring of cell behavior at the blood-scaffold interphase. MC3T3-E1 cells were cultured with three different types of scaffold inserts to test its capability as an evaluation platform. Cellular behaviors, including migration, morphology, and osteogenesis with each scaffold, were analyzed through fluorescence images of live/dead assay and immunocytochemistry. Cellular behaviors, such as migration, morphology, and osteogenesis, were evaluated. The results revealed that our platform could effectively evaluate the osteoconductivity and osteoinductivity of scaffolds with various properties. In conclusion, our proposed platform is expected to replace current in vivo animal models as a highly relevant in vitro platform and can contribute to the fundamental study of bone regeneration.
Collapse
Affiliation(s)
- Jinsub Han
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea.,Convergence Major in Global Smart Farm, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Sangbae Park
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jae Eun Kim
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea
| | - Byeongjoo Park
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea
| | - Yeonggeol Hong
- Department of Bio-Systems Engineering, Institute of Smart Farm, Gyeongsang National University, Jinju 52828, Korea
| | - Jae Woon Lim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Seung Jeong
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Hyunmok Son
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Hong Bae Kim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Hoon Seonwoo
- Department of Convergent Biosystems Engineering, College of Life Sciences and Natural Resources, Sunchon National University, Suncheon 57922, Korea.,Interdisciplinary Program in IT-Bio Convergence System, Sunchon National University, Suncheon 57922, Korea
| | - Kyoung-Je Jang
- Department of Bio-Systems Engineering, Institute of Smart Farm, Gyeongsang National University, Jinju 52828, Korea.,Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52828, Korea
| | - Jong Hoon Chung
- Department of Biosystems Engineering, Seoul National University, Seoul 08826, Korea.,Convergence Major in Global Smart Farm, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.,Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
34
|
Jiao J, Hong Q, Zhang D, Wang M, Tang H, Yang J, Qu X, Yue B. Influence of porosity on osteogenesis, bone growth and osteointegration in trabecular tantalum scaffolds fabricated by additive manufacturing. Front Bioeng Biotechnol 2023; 11:1117954. [PMID: 36777251 PMCID: PMC9911888 DOI: 10.3389/fbioe.2023.1117954] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Porous tantalum implants are a class of materials commonly used in clinical practice to repair bone defects. However, the cumbersome and problematic preparation procedure have limited their widespread application. Additive manufacturing has revolutionized the design and process of orthopedic implants, but the pore architecture feature of porous tantalum scaffolds prepared from additive materials for optimal osseointegration are unclear, particularly the influence of porosity. We prepared trabecular bone-mimicking tantalum scaffolds with three different porosities (60%, 70% and 80%) using the laser powder bed fusing technique to examine and compare the effects of adhesion, proliferation and osteogenic differentiation capacity of rat mesenchymal stem cells on the scaffolds in vitro. The in vivo bone ingrowth and osseointegration effects of each scaffold were analyzed in a rat femoral bone defect model. Three porous tantalum scaffolds were successfully prepared and characterized. In vitro studies showed that scaffolds with 70% and 80% porosity had a better ability to osteogenic proliferation and differentiation than scaffolds with 60% porosity. In vivo studies further confirmed that tantalum scaffolds with the 70% and 80% porosity had a better ability for bone ingrowh than the scaffold with 60% porosity. As for osseointegration, more bone was bound to the material in the scaffold with 70% porosity, suggesting that the 3D printed trabecular tantalum scaffold with 70% porosity could be the optimal choice for subsequent implant design, which we will further confirm in a large animal preclinical model for better clinical use.
Collapse
Affiliation(s)
- Juyang Jiao
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qimin Hong
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dachen Zhang
- Shenzhen Dazhou Medical Technology Co., Ltd., Shenzhen, Guangdong, China
- Center of Biomedical Materials 3D Printing, National Engineering Laboratory for Polymer Complex Structure Additive Manufacturing, Baoding, Hebei, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingzhou Yang
- Shenzhen Dazhou Medical Technology Co., Ltd., Shenzhen, Guangdong, China
- Center of Biomedical Materials 3D Printing, National Engineering Laboratory for Polymer Complex Structure Additive Manufacturing, Baoding, Hebei, China
- School of Mechanical and Automobile Engineering, Qingdao University of Technology, Qingdao, Shandong, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
RANDHAWA AAYUSHI, DEB DUTTA SAYAN, GANGULY KEYA, V. PATIL TEJAL, LUTHFIKASARI RACHMI, LIM KITAEK. Understanding cell-extracellular matrix interactions for topology-guided tissue regeneration. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
36
|
Liu H, Wang C, Sun X, Zhan C, Li Z, Qiu L, Luo R, Liu H, Sun X, Li R, Zhang J. Silk Fibroin/Collagen/Hydroxyapatite Scaffolds Obtained by 3D Printing Technology and Loaded with Recombinant Human Erythropoietin in the Reconstruction of Alveolar Bone Defects. ACS Biomater Sci Eng 2022; 8:5245-5256. [PMID: 36336837 DOI: 10.1021/acsbiomaterials.2c00690] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The fast osteogenesis of the large alveolar fossa and the maintenance of the height of the alveolar ridge after tooth extraction have always been a clinical challenge. Therefore, this work describes the creation of innovative silk fibroin/collagen/hydroxyapatite (SCH) biological scaffolds by 3D printing technology, which are loaded with recombinant human erythropoietin (rh-EPO) for the reconstruction of bone defects. Low-temperature 3D printing can maintain the biological activity of silk fibroin and collagen. The SCH scaffolds showed the ideal water absorption and porosity, being a sustained-release carrier of rh-EPO. The optimized scaffolds had ideal mechanical properties in vitro, and MC3T3-E1 cells could easily adhere and proliferate on it. In vivo experiments in rabbits demonstrated that the composite scaffolds gradually degraded and promoted the accumulation and proliferation of osteoblasts and the formation of collagen fibers, significantly promoting the reconstruction of mandibular defects. In this study, a novel composite biological scaffold was prepared using 3D printing technology, and the scaffold was innovatively combined with the multifunctional growth factor rh-EPO. This provides a new optimized composite material for the reconstruction of irregular mandible defects, and this biomaterial is promising for clinical reconstruction of alveolar bone defects.
Collapse
Affiliation(s)
- Han Liu
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Chao Wang
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Xiaoqian Sun
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Chaojun Zhan
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Zixiao Li
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Lin Qiu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100034, China
| | - Rui Luo
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Hao Liu
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Xiaodi Sun
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Ruixin Li
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Jun Zhang
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| |
Collapse
|
37
|
Su Y, Gao Q, Deng R, Zeng L, Guo J, Ye B, Yu J, Guo X. Aptamer engineering exosomes loaded on biomimetic periosteum to promote angiogenesis and bone regeneration by targeting injured nerves via JNK3 MAPK pathway. Mater Today Bio 2022; 16:100434. [PMID: 36186848 PMCID: PMC9519612 DOI: 10.1016/j.mtbio.2022.100434] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 12/04/2022] Open
Abstract
Repairing critical bone defects is a complex problem in the clinic. The periosteum rich in nerve plays a vital role in initiating and regulating bone regeneration. However, current studies have paid little attention to repairing nerves in the periosteum to promote bone regeneration. Thus, it is essential to construct bionic periosteum with the targeted injured nerves in the periosteum. We coupled phosphatidylserine (PS) targeted aptamers with repair Schwann cell exosomes to construct exosome@aptamer (EA). Then through PEI, EA was successfully built on the surface of the electrospun fiber, which was PCL@PEI@exosome@aptamer (PPEA). Through SEM, TEM, and other technologies, PPEA was characterized. Experiments prove in vivo and in vitro that it has an excellent repair effect on damaged nerves and regeneration of vascular and bones. In vivo, we confirmed that biomimetic periosteum has an apparent ability to promote nerve and bone regeneration by using Microcomputer tomography, hematoxylin-eosin, Masson, and Immunofluorescence. In vitro, we used Immunofluorescence, Real-Time Quantitative PCR, Alkaline phosphatase staining, and other tests to confirm that it has central nerve, blood vessel, and bone regeneration ability. The PPEA biomimetic periosteum has apparent neurogenic, angiogenic, and osteogenic effects. The PPEA biomimetic periosteum will provide a promising method for treating bone defects. To construct a biomimetic periosteum that can target injured axons and bone regeneration. PS targeted aptamer is coupled with repair Schwann cell exosomes. PEI self-assembly was used for the PCL electrospun biomimetic membrane loading. It targeted and repaired the injured axons and promoted the secretion of CGRP and SP. Biomimetic periosteum promotes vascular regeneration and bone regeneration.
Collapse
Affiliation(s)
- Yanlin Su
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Qing Gao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Rongli Deng
- PCFM Lab, School of Chemistry and School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Lian Zeng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jingyi Guo
- College of Arts and Science of Hubei Normal University, Huangshi, Hubei 430022, China
| | - Bing Ye
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jialin Yu
- The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 430022, China
| | - Xiaodong Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- Corresponding author.
| |
Collapse
|
38
|
Li G, Yang L, Wu G, Qian Z, Li H. An update of interbody cages for spine fusion surgeries: from shape design to materials. Expert Rev Med Devices 2022; 19:977-989. [PMID: 36617696 DOI: 10.1080/17434440.2022.2165912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Discectomy and interbody fusion are widely used in the treatment of intervertebral disc-related diseases. Among them, the interbody cage plays a significant role. However, the complications related to the interbody cage, such as nonunion or pseudoarthrosis, subsidence, loosening, and prolapse of the cage, cannot be ignored. By changing the design and material of the interbody fusion cage, a better fusion effect can be obtained, the incidence of appeal complications can be reduced, and the quality of life of patients after interbody fusion can be improved. AREAS COVERED This study reviewed the research progress of cage design and material and discussed the methods of cage design and material to promote intervertebral fusion. EXPERT OPINION Current treatment of cervical and lumbar degenerative disease requires interbody fusion to maintain decompression and to promote fusion and reduce the incidence of fusion failure through improvements in implant material, design, internal structure, and function. However, interbody fusion is not an optimal solution for treating vertebral instability.Abbreviations: ACDF, Anterior cervical discectomy and fusion; ALIF, anterior lumbar interbody fusion; Axi-aLIF, axial lumbar interbody fusion; BAK fusion cage, Bagby and Kuslich fusion cage; CADR, cervical artificial disc replacement; DBM, decalcified bone matrix; HA, hydroxyapatite; LLIF/XLIF, lateral or extreme lateral interbody fusion; MIS-TLIF, minimally invasive transforaminal lumbar interbody fusion; OLIF/ATP, oblique lumbar interbody fusion/anterior to psoas; PEEK, Poly-ether-ether-ketone; PLIF, posterior lumbar interbody fusion; ROI-C, Zero-profile Anchored Spacer; ROM, range of motion; SLM, selective melting forming; TLIF, transforaminal lumbar interbody fusion or.
Collapse
Affiliation(s)
- Guangshen Li
- Nantong University Medical School, 226000, Nantong, Jiangsu, China.,Department of Orthopedics, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, 225300, Taizhou, China.,Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Lei Yang
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Gang Wu
- Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Zhanyang Qian
- School of Medicine, Southeast University, Nanjing, China; Spine Center, Zhongda Hospital of Southeast University, Nanjing, China
| | - Haijun Li
- Nantong University Medical School, 226000, Nantong, Jiangsu, China.,Department of Orthopedics, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, 225300, Taizhou, China.,Department of Orthopedics, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China.,Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|
39
|
Xue Y, Zhang L, Liu F, Zhao Y, Zhou J, Hou Y, Bao H, Kong L, Ma F, Han Y. Surface Bandgap Engineering of Nanostructured Implants for Rapid Photothermal Ion Therapy of Bone Defects. Adv Healthc Mater 2022; 11:e2200998. [PMID: 36064207 DOI: 10.1002/adhm.202200998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/31/2022] [Indexed: 01/28/2023]
Abstract
Bone defects are seriously threatening the health of orthopedics patients and it is difficult for implants to accelerate bone regeneration without using bone growth factors. Herein, a fast photothermal ion therapeutic strategy is developed based on the bandgap engineering of nanostructured TiO2 through (Si/P)-dual elemental doping by micro-arc oxidation treatment of titanium implants. The (Si/P)-dual doping can tune the surface bandgap structure of TiO2 by decreasing bandgap and broadening valence band simultaneously, which is confirmed by density functional theory calculations. It not only endows the implants with a mildly photothermal effect under near-infrared (NIR) light irradiation, but also creates an (Si/P) ion-rich microenvironment around the implants. This photothermal ion microenvironment can tune the behaviors of osteoblasts by promoting p38/Smad and ERK signaling pathways of osteoblasts, thus significantly upregulating the expression of osteogenesis genes by the synergistic action of mild photothermal stimulation and increased release of Si/P ions. The in vivo results are also in good agreement with in vitro tests, i.e., under NIR light irradiation, the photothermally responsive TiO2 enhances the bone formation and osteointegration with implants. Therefore, this kind of photothermal ion strategy is a promising remote and noninvasive therapeutic mode for promoting bone regeneration of Ti implants.
Collapse
Affiliation(s)
- Yang Xue
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lan Zhang
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yiwei Zhao
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jianhong Zhou
- Institute of Physics & Optoelectronics Technology, Advanced Titanium Alloys and Functional Coatings Cooperative Innovation Center, Baoji University of Arts and Sciences, Baoji, 721016, China
| | - Yan Hou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Han Bao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fei Ma
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yong Han
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
40
|
Murchio S, Benedetti M, Berto A, Agostinacchio F, Zappini G, Maniglio D. Hybrid Ti6Al4V/Silk Fibroin Composite for Load-Bearing Implants: A Hierarchical Multifunctional Cellular Scaffold. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6156. [PMID: 36079541 PMCID: PMC9458142 DOI: 10.3390/ma15176156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
Despite the tremendous technological advances that metal additive manufacturing (AM) has made in the last decades, there are still some major concerns guaranteeing its massive industrial application in the biomedical field. Indeed, some main limitations arise in dealing with their biological properties, specifically in terms of osseointegration. Morphological accuracy of sub-unital elements along with the printing resolution are major constraints in the design workspace of a lattice, hindering the possibility of manufacturing structures optimized for proper osteointegration. To overcome these issues, the authors developed a new hybrid multifunctional composite scaffold consisting of an AM Ti6Al4V lattice structure and a silk fibroin/gelatin foam. The composite was realized by combining laser powder bed fusion (L-PBF) of simple cubic lattice structures with foaming techniques. A combined process of foaming and electrodeposition has been also evaluated. The multifunctional scaffolds were characterized to evaluate their pore size, morphology, and distribution as well as their adhesion and behavior at the metal-polymer interface. Pull-out tests in dry and hydrated conditions were employed for the mechanical characterization. Additionally, a cytotoxicity assessment was performed to preliminarily evaluate their potential application in the biomedical field as load-bearing next-generation medical devices.
Collapse
Affiliation(s)
- Simone Murchio
- Department of Industrial Engineering–DII, University of Trento, 38123 Trento, Italy
- BIOtech Research Center, University of Trento, 38122 Trento, Italy
| | - Matteo Benedetti
- BIOtech Research Center, University of Trento, 38122 Trento, Italy
| | - Anastasia Berto
- BIOtech Research Center, University of Trento, 38122 Trento, Italy
| | - Francesca Agostinacchio
- Department of Industrial Engineering–DII, University of Trento, 38123 Trento, Italy
- BIOtech Research Center, University of Trento, 38122 Trento, Italy
| | | | - Devid Maniglio
- Department of Industrial Engineering–DII, University of Trento, 38123 Trento, Italy
- BIOtech Research Center, University of Trento, 38122 Trento, Italy
| |
Collapse
|
41
|
Howard MT, Wang S, Berger AG, Martin JR, Jalili-Firoozinezhad S, Padera RF, Hammond PT. Sustained release of BMP-2 using self-assembled layer-by-layer film-coated implants enhances bone regeneration over burst release. Biomaterials 2022; 288:121721. [PMID: 35981926 PMCID: PMC10396073 DOI: 10.1016/j.biomaterials.2022.121721] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
Abstract
Current clinical products delivering the osteogenic growth factor bone morphogenetic protein 2 (BMP-2) for bone regeneration have been plagued by safety concerns due to a high incidence of off-target effects resulting from bolus release and supraphysiological doses. Layer-by-layer (LbL) film deposition offers the opportunity to coat bone defect-relevant substrates with thin films containing proteins and other therapeutics; however, control of release kinetics is often hampered by interlayer diffusion of drugs throughout the film during assembly, which causes burst drug release. In this work, we present the design of different laponite clay diffusional barrier layer architectures in self-assembled LbL films to modulate the release kinetics of BMP-2 from the surface of a biodegradable implant. Release kinetics were tuned by incorporating laponite in different film arrangements and with varying deposition techniques to achieve release of BMP-2 over 2 days, 4 days, 14 days, and 30 days. Delivery of a low dose (0.5 μg) of BMP-2 over 2 days and 30 days using these LbL film architectures was then compared in an in vivo rat critical size calvarial defect model to determine the effect of BMP-2 release kinetics on bone regeneration. After 6 weeks, sustained release of BMP-2 over 30 days induced 3.7 times higher bone volume and 7.4 times higher bone mineral density as compared with 2-day release of BMP-2, which did not induce more bone growth than the uncoated scaffold control. These findings represent a crucial step in the understanding of how BMP-2 release kinetics influence treatment efficacy and underscore the necessity to optimize protein delivery methods in clinical formulations for bone regeneration. This work could be applied to the delivery of other therapeutic proteins for which careful tuning of the release rate is a key optimization parameter.
Collapse
Affiliation(s)
- MayLin T Howard
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Sheryl Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Adam G Berger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - John R Martin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Robert F Padera
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, United States.
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| |
Collapse
|
42
|
Wang Y, Ling C, Chen J, Liu H, Mo Q, Zhang W, Yao Q. 3D-printed composite scaffold with gradient structure and programmed biomolecule delivery to guide stem cell behavior for osteochondral regeneration. BIOMATERIALS ADVANCES 2022; 140:213067. [PMID: 35961187 DOI: 10.1016/j.bioadv.2022.213067] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
The fabrication of osteochondral scaffolds with both structural and biochemical cues to regulate endogenous bone marrow-derived mesenchymal stem cells (BMSCs) behavior for cartilage and subchondral bone regeneration is still a challenge. To this end, a composite scaffold (BE-PSA) with gradient structure and programmed biomolecule delivery was prepared by fused deposition modeling (FDM) 3D printing and multi-material-based modification. The 3D-printed polycaprolactone (PCL) scaffold included upper pores of 200 μm for cartilage regeneration and lower pores of 400 μm for bone regeneration. For a sequential modulation of BMSCs behavior, fast-degrading sodium alginate (SA) hydrogel was used to deliver a burst release of E7 peptide to enhance BMSCs migration within 72 h, while a slowly-degrading silk fibroin (SF) porous matrix was used to provide a sustained release of B2A peptide to improve BMSCs dual-lineage differentiation lasting for >300 h, depending on the different degradation rates of SA hydrogel and SF matrix. The BE-PSA scaffold had good biocompatibility and could improve the migration and osteogenic/chondrogenic differentiation of BMSCs. Benefiting from the synergistic effects of spatial structures and programmed biomolecule delivery, the BE-PSA scaffold showed enhanced cartilage and subchondral bone regeneration in rabbit osteochondral defect model. This work not only provides a promising scaffold to guide BMSCs behavior for osteochondral regeneration but also offers a method for the fabrication of tissue engineering biomaterials based on the structural and biochemical modification.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Chen Ling
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China; Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China; China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China; Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China; China Orthopedic Regenerative Medicine Group (CORMed), China.
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China; China Orthopedic Regenerative Medicine Group (CORMed), China.
| |
Collapse
|
43
|
Liu Y, Yang Q, Wang Y, Lin M, Tong Y, Huang H, Yang C, Wu J, Tang B, Bai J, Liu C. Metallic Scaffold with Micron-Scale Geometrical Cues Promotes Osteogenesis and Angiogenesis via the ROCK/Myosin/YAP Pathway. ACS Biomater Sci Eng 2022; 8:3498-3514. [PMID: 35834297 DOI: 10.1021/acsbiomaterials.2c00225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The advent of precision manufacturing has enabled the creation of pores in metallic scaffolds with feature size in the range of single microns. In orthopedic implants, pore geometries at the micron scale could regulate bone formation by stimulating osteogenic differentiation and the coupling of osteogenesis and angiogenesis. However, the biological response to pore geometry at the cellular level is not clear. As cells are sensitive to curvature of the pore boundary, this study aimed to investigate osteogenesis in high- vs low-curvature environments by utilizing computer numerical control laser cutting to generate triangular and circular precision manufactured micropores (PMpores). We fabricated PMpores on 100 μm-thick stainless-steel discs. Triangular PMpores had a 30° vertex angle and a 300 μm base, and circular PMpores had a 300 μm diameter. We found triangular PMpores significantly enhanced the elastic modulus, proliferation, migration, and osteogenic differentiation of MC3T3-E1 preosteoblasts through Yes-associated protein (YAP) nuclear translocation. Inhibition of Rho-associated kinase (ROCK) and Myosin II abolished YAP translocation in all pore types and controls. Inhibition of YAP transcriptional activity reduced the proliferation, pore closure, collagen secretion, alkaline phosphatase (ALP), and Alizarin Red staining in MC3T3-E1 cultures. In C166 vascular endothelial cells, PMpores increased the VEGFA mRNA expression even without an angiogenic differentiation medium and induced tubule formation and maintenance. In terms of osteogenesis-angiogenesis coupling, a conditioned medium from MC3T3-E1 cells in PMpores promoted the expression of angiogenic genes in C166 cells. A coculture with MC3T3-E1 induced tubule formation and maintenance in C166 cells and tubule alignment along the edges of pores. Together, curvature cues in micropores are important stimuli to regulate osteogenic differentiation and osteogenesis-angiogenesis coupling. This study uncovered key mechanotransduction signaling components activated by curvature differences in a metallic scaffold and contributed to the understanding of the interaction between orthopedic implants and cells.
Collapse
Affiliation(s)
- Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Qihao Yang
- The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, 510150 Guangzhou, China
| | - Yue Wang
- Department of Mechanical and Energy Engineering, College of Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Yanrong Tong
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Hanwei Huang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Jianqun Wu
- College of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Bin Tang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Jiaming Bai
- Department of Mechanical and Energy Engineering, College of Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Advanced Biomaterials, 1088 Xueyuan Avenue, 518055 Shenzhen, China
| |
Collapse
|
44
|
Corrosion Resistance of 3D Printed Ti6Al4V Gyroid Lattices with Varying Porosity. MATERIALS 2022; 15:ma15144805. [PMID: 35888273 PMCID: PMC9316743 DOI: 10.3390/ma15144805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023]
Abstract
Corrosion of medical implants is a possible failure mode via induced local inflammatory effects, systemic deposition and corrosion related mechanical failure. Cyclic potentiodynamic polarisation (CPP) testing was utilized to evaluate the effect of increased porosity (60% and 80%) and decreased wall thickness in gyroid lattice structures on the electrochemical behaviour of LPBF Ti6Al4V structures. The use of CPP allowed for the landmarks of breakdown potential, resting potential and vertex potential to be analysed, as well as facilitating the construction of Tafel plots and qualitative Goldberg analysis. The results indicated that 60% gyroid samples were most susceptible to the onset of pitting corrosion when compared to 80% gyroid and solid samples. This was shown through decreased breakdown and vertex potentials and were found to correlate to increased lattice surface area to void volume ratio. Tafel plots indicated that despite the earlier onset of pitting corrosion, both gyroid test groups displayed lower rates of corrosion per year, indicating a lower severity of corrosion. This study highlighted inherent tradeoffs between lattice optimisation and corrosion behaviour with a potential parabolic link between void volume, surface area and corrosion being identified. This potential link is supported by 60% gyroid samples having the lowest breakdown potentials, but investigation into other porosity ranges is suggested to support the hypothesis. All 3D printed materials studied here showed breakdown potentials higher than ASTM F2129's suggestion of 800 mV for evaluation within the physiological environment, indicating that under static conditions pitting and crevice corrosion should not initiate within the body.
Collapse
|
45
|
Zhou R, Xue H, Wang J, Wang X, Wang Y, Zhang A, Zhang J, Han Q, Zhao X. Improving the Stability of a Hemipelvic Prosthesis Based on Bone Mineral Density Screw Channel and Prosthesis Optimization Design. Front Bioeng Biotechnol 2022; 10:892385. [PMID: 35706507 PMCID: PMC9189365 DOI: 10.3389/fbioe.2022.892385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
In pelvic reconstruction surgery, the hemipelvic prosthesis can cause significant changes in stress distribution due to its high stiffness, and its solid structure is not suitable for osseointegration. The purpose of this study was to identify a novel bone mineral density screw channel and design the structure of the prosthesis so as to improve the distribution of stress, promote bone growth, and enhance the biomechanical properties of the prosthesis. The mechanical characteristics of bone mineral density screw and traditional screw were compared by finite element analysis method, and redesigned by topology optimization. The direction of the newly proposed screw channel was the posterolateral entrance of the auricular surface, ending at the contralateral sacral cape. Compared to the original group, the maximum stress of the optimized prosthesis was decreased by 24.39%, the maximum stress of the sacrum in the optimized group was decreased by 27.23%, and the average strain energy density of the sacrum in the optimized group was increased by 8.43%. On the surface of screw and connecting plate, the area with micromotion more than 28 μm is reduced by 12.17%. On the screw surface, the area with micromotion more than 28 μm is reduced by 22.9%. The newly determined screw channel and optimized prosthesis design can effectively improve the biomechanical properties of a prosthesis and the microenvironment of osseointegration. This method can provide a reference for the fixation of prostheses in clinical pelvic reconstruction.
Collapse
|
46
|
Ran T, Lin C, Ma T, Qin Y, Li J, Zhang Y, Xu Y, Li C, Wang M. Ultra-Pulsed CO 2 Laser Osteotomy: A New Method for the Bone Preparation of Total Knee Arthroplasty. Front Bioeng Biotechnol 2022; 10:858862. [PMID: 35573227 PMCID: PMC9096707 DOI: 10.3389/fbioe.2022.858862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Cementless total knee arthroplasty (TKA) can achieve long-term biological fixation, but its application is limited by the risk of early aseptic loosening. One of the important reasons for early aseptic loosening is that mechanical osteotomy tools cannot achieve ideal bone preparation because of poor accuracy and serious bone tissue damage produced by them. Therefore, we designed an ultra-pulsed CO2 laser osteotomy system to solve these problems. To reveal the safety at the tissue and cell levels of the ultra-pulsed CO2 laser osteotomy system, a series of experiments on distal femur osteotomy in animals were performed. Then, the bone surface characteristics were analyzed through scanning electron microscopy, and the bone thermal and mechanical damage was evaluated via histological analysis. Finally, mesenchymal stem cells (MSCs) were inoculated on the bone surfaces prepared by the two osteotomy tools, and the effect of cell adhesion was analyzed through a confocal laser scanning microscope (CLSM). We successfully achieved TKA bone preparation of animal knees with the ultra-pulsed CO2 laser osteotomy system. Moreover, the biological evaluation results indicated that compared with the traditional mechanical saw, the laser can preserve the natural bone structure and cause no thermal damage to the bone. In addition, CLSM examination results showed that the laser-cut bone surface was more conducive to cell adhesion and infiltration than the bone surface cut by a mechanical saw. Overall, these results indicate that ultra-pulsed CO2 laser can achieve non-invasive bone cutting, which can be a new option for TKA bone preparation and has the potential to lead in the future.
Collapse
Affiliation(s)
- Tianfei Ran
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Chuanchuan Lin
- Department of Blood Transfusion, Xinqiao Hospital, Amy Medical University (Third Military Medical University), Chongqing, China
| | - Tianying Ma
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yinyin Qin
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Xu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Min Wang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
47
|
Chen J, Xie L, Ruan Q, Gao A, Liao Q, Mo S, Lv Y, Tong L, Wang H, Chu PK, Li X. Diamond-like carbon coating and surface grafting of osteoprotegerin and alendronate on polyetheretherketone to ameliorate the mechanical performance and osseointegration simultaneously. COMPOSITES PART B: ENGINEERING 2022; 236:109815. [DOI: 10.1016/j.compositesb.2022.109815] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
48
|
Abstract
Ti-6Al-4V (Ti64) alloy is one of the most widely used orthopedic implant materials due to its mechanical properties, corrosion resistance, and biocompatibility nature. Porous Ti64 structures are gaining more research interest as bone implants as they can help in reducing the stress-shielding effect when compared to their solid counterpart. The literature shows that porous Ti64 implants fabricated using different additive manufacturing (AM) process routes, such as laser powder bed fusion (L-PBF) and electron beam melting (EBM) can be tailored to mimic the mechanical properties of natural bone. This review paper categorizes porous implant designs into non-gradient (uniform) and gradient (non-uniform) porous structures. Gradient porous design appears to be more promising for orthopedic applications due to its closeness towards natural bone morphology and improved mechanical properties. In addition, this paper outlines the details on bone structure and its properties, mechanical properties, fatigue behavior, multifunctional porous implant designs, current challenges, and literature gaps in the research studies on porous Ti64 bone implants.
Collapse
|
49
|
Yu F, Lian R, Liu L, Liu T, Bi C, Hong K, Zhang S, Ren J, Wang H, Ouyang N, Du LJ, Liu Y, Zhou L, Liu Y, Fang B, Li Y, Duan SZ, Xia L. Biomimetic Hydroxyapatite Nanorods Promote Bone Regeneration via Accelerating Osteogenesis of BMSCs through T Cell-Derived IL-22. ACS NANO 2022; 16:755-770. [PMID: 35005890 DOI: 10.1021/acsnano.1c08281] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Manipulations of morphological properties of nanobiomaterials have been demonstrated to modulate the outcome of osteoimmunomodulation and eventually osteogenesis through innate immune response. However, the functions and mechanisms of adaptive immune cells in the process of nanobiomaterials-mediated bone regeneration have remained unknown. Herein, we developed bone-mimicking hydroxyapatite (HAp) nanorods with different aspect ratios as model materials to investigate the impacts of the nanoshape features on osteogenesis and to explore the underlying mechanisms focusing on the functions of T cells and T cell-derived cytokines. HAp nanorods with different aspect ratios (HAp-0, HAp-30, and HAp-100) were implanted into mouse mandibular defect models. Micro-CT and hematoxylin and eosin staining demonstrated that HAp-100 had the best osteogenic effects. Flow cytometry analysis revealed that HAp-100 increased the percentage of T cells in injured mandibles. The osteogenic effects of HAp-100 were significantly blunted in injured mandibles of TCRβ-/- mice. The Luminex xMAP assay and ELISA showed that HAp-100 induced a marked increase of interleukin (IL)-22 in injured mandibles. In cultured T cells, HAp-100 manifested the best capacity to induce the production of IL-22. Conditioned media from HAp-100-primed T cells promoted osteogenesis and JAK1/STAT3 activation in bone marrow stromal cells, all of which were abolished by neutralizing antibodies against IL-22. In summary, bone-mimicking HAp nanorods with different aspect ratios could regulate osteogenesis through modulation of T cells and IL-22 in the bone regeneration process. These findings provided insights for mediation of the immune response of T cells by nanomaterials on osteogenesis and strategies for designing biomaterials with osteoimmunomodulative functions.
Collapse
Affiliation(s)
- Fei Yu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Ruixian Lian
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Lu Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Ting Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Chao Bi
- Department of Stomatology, First Affiliated Hospital, Anhui Medical University, Hefei 230061, China
| | - Kan Hong
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shuiquan Zhang
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiazi Ren
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lujun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Bing Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| | - Lunguo Xia
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai 200125, China
| |
Collapse
|
50
|
Gherasim O, Grumezescu AM, Grumezescu V, Andronescu E, Negut I, Bîrcă AC, Gălățeanu B, Hudiță A. Bioactive Coatings Loaded with Osteogenic Protein for Metallic Implants. Polymers (Basel) 2021; 13:4303. [PMID: 34960852 PMCID: PMC8703935 DOI: 10.3390/polym13244303] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoconductive and osteoinductive coatings represent attractive and tunable strategies towards the enhanced biomechanics and osseointegration of metallic implants, providing accurate local modulation of bone-to-implant interface. Composite materials based on polylactide (PLA) and hydroxyapatite (HAp) are proved beneficial substrates for the modulation of bone cells' development, being suitable mechanical supports for the repair and regeneration of bone tissue. Moreover, the addition of osteogenic proteins represents the next step towards the fabrication of advanced biomaterials for hard tissue engineering applications, as their regulatory mechanisms beneficially contribute to the new bone formation. In this respect, laser-processed composites, based on PLA, Hap, and bone morphogenetic protein 4(BMP4), are herein proposed as bioactive coatings for metallic implants. The nanostructured coatings proved superior ability to promote the adhesion, viability, and proliferation of osteoprogenitor cells, without affecting their normal development and further sustaining the osteogenic differentiation of the cells. Our results are complementary to previous studies regarding the successful use of chemically BMP-modified biomaterials in orthopedic and orthodontic applications.
Collapse
Affiliation(s)
- Oana Gherasim
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania; (O.G.); (A.M.G.); (E.A.); (A.C.B.)
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, RO-77125 Magurele, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania; (O.G.); (A.M.G.); (E.A.); (A.C.B.)
- Academy of Romanian Scientists, Ilfov No. 3, 50044 Bucharest, Romania
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Valentina Grumezescu
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, RO-77125 Magurele, Romania;
- Academy of Romanian Scientists, Ilfov No. 3, 50044 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania; (O.G.); (A.M.G.); (E.A.); (A.C.B.)
- Academy of Romanian Scientists, Ilfov No. 3, 50044 Bucharest, Romania
| | - Irina Negut
- Lasers Department, National Institute for Lasers, Plasma, and Radiation Physics, RO-77125 Magurele, Romania;
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania; (O.G.); (A.M.G.); (E.A.); (A.C.B.)
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independentei, 050095 Bucharest, Romania; (B.G.); (A.H.)
| | - Ariana Hudiță
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independentei, 050095 Bucharest, Romania; (B.G.); (A.H.)
| |
Collapse
|