Advertisement for orthosearch.org.uk
Results 1 - 20 of 158
Results per page:
Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_7 | Pages 109 - 109
4 Apr 2023
Ouyang Z Ding Y Lin S Wen Z
Full Access

Aseptic inflammation is the main factor causing aseptic loosening of artificial joints. Studies have shown that inflammatory cells can activate STING (stimulator of interferon genes, STING) after being stressed. This study aims to explore the specific mechanism of STING in aseptic loosening of artificial joints, and provide new strategies for disease prevention. Titanium particles with a diameter of 1.2-10 μm were prepared to stimulate macrophages (RAW 264.7) to simulate the periprosthetic microenvironment. A lentiviral vector targeting the STING gene was designed and transfected into macrophages to construct a cell line targeting STING knockdown. The expression and secretion levels of TNF-α were detected by qPCR and ELISA, the activation levels of inflammatory pathways (NF-κB, IRF3, etc.) were detected by western blot, and the nucleus translocation of P65 and IRF3 was observed by cellular immunofluorescence. After titanium particles stimulated macrophages, qPCR and ELISA showed that the transcription and secretion levels of TNF-α were significantly increased. Western blot showed that titanium particle stimulation could increase the phosphorylation levels of NF-κB and IRF3 pathways. While knockdown of STING can significantly reduce titanium particle-induced TNF production, attenuate the activation levels of NF-κB and IRF3 pathways as well as the nucleus translocation of P65 and IRF3. Conclusions: STING positively regulates the level of inflammation in macrophages induced by titanium particles, and targeted inhibition of STING can reduce inflammation, which may delay the progression of aseptic loosening of artificial joints


Aims. This study intended to investigate the effect of vericiguat (VIT) on titanium rod osseointegration in aged rats with iron overload, and also explore the role of VIT in osteoblast and osteoclast differentiation. Methods. In this study, 60 rats were included in a titanium rod implantation model and underwent subsequent guanylate cyclase treatment. Imaging, histology, and biomechanics were used to evaluate the osseointegration of rats in each group. First, the impact of VIT on bone integration in aged rats with iron overload was investigated. Subsequently, VIT was employed to modulate the differentiation of MC3T3-E1 cells and RAW264.7 cells under conditions of iron overload. Results. Utilizing an OVX rat model, we observed significant alterations in bone mass and osseointegration due to VIT administration in aged rats with iron overload. The observed effects were concomitant with reductions in bone metabolism, oxidative stress, and inflammation. To elucidate whether these effects are associated with osteoclast and osteoblast activity, we conducted in vitro experiments using MC3T3-E1 cells and RAW264.7 cells. Our findings indicate that iron accumulation suppressed the activity of MC3T3-E1 while enhancing RAW264.7 function. Furthermore, iron overload significantly decreased oxidative stress levels; however, these detrimental effects can be mitigated by VIT treatment. Conclusion. Collectively, our data provide compelling evidence that VIT has the potential to reverse the deleterious consequences of iron overload on osseointegration and bone mass during ageing. Cite this article: Bone Joint Res 2024;13(9):427–440


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 45 - 45
2 Jan 2024
Riool M Li R Hofwegen L de Boer L Loontjens J Zaat S
Full Access

Infection of implanted medical devices (biomaterials), like titanium orthopaedic implants, can have disastrous consequences, including removal of the device. These so-called biomaterial-associated infections (BAI) are mainly caused by Staphylococcus aureus and Staphylococcus epidermidis. To prevent biofilm formation using a non-antibiotic based strategy, we aimed to develop a novel permanently fixed antimicrobial coating for titanium devices based on stable immobilized quaternary ammonium compounds (QACs). Medical grade titanium implants were dip-coated in subsequent solutions of hyperbranched polymer, polyethyleneimine and 10 mM sodium iodide, and ethanol. The QAC-coating was characterized using water contact angle measurements, scanning electron microscopy, FTIR, AFM and XPS. The antimicrobial activity of the coating was evaluated against S. aureus strain JAR060131 and S. epidermidis strain ATCC 12228 using the JIS Z 2801:2000 surface microbicidal assay. Lastly, we assessed the in vivo antimicrobial activity in a mouse subcutaneous implant infection model with S. aureus administered locally on the QAC-coated implants prior to implantation to mimic contamination during surgery. Detailed material characterization of the titanium samples showed the presence of a homogenous and stable coating layer at the titanium surface. Moreover, the coating successfully killed S. aureus and S. epidermidis in vitro. The QAC-coating strongly reduced S. aureus colonization of the implant surface as well as of the surrounding tissue, with no apparent macroscopic signs of toxicity or inflammation in the peri-implant tissue at 1 and 4 days after implantation. An antimicrobial coating with stable quaternary ammonium compounds on titanium has been developed which holds promise to prevent BAI. Non-antibiotic-based antimicrobial coatings have great significance in guiding the design of novel antimicrobial coatings in the present, post-antibiotic era. Acknowledgements: This research was financially supported by the Health∼Holland/LSH-TKI call 2021–2022, project 25687, NACQAC: ‘Novel antimicrobial coatings with stable non-antibiotic Quaternary Ammonium Compounds and photosensitizer technology'


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 59 - 59
2 Jan 2024
Depboylu F
Full Access

Production of porous titanium bone implants is a highly promising research and application area due to providing high osseointegration and achieving the desired mechanical properties. Production of controlled porosity in titanium implants is possible with laser powder bed fusion (L- PBF) technology. The main topics of this presentation includes the L-PBF process parameter optimization to manufacture thin walls of porous titanium structures with almost full density and good mechanical properties as well as good dimensional accuracy. Moreover, the cleaning and coating process of these structures to further increase osseointegration and then in-vitro biocompatibility will be covered


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_4 | Pages 70 - 70
1 Mar 2021
Stich T Krenek T Kovarik T Docheva D
Full Access

Numerous implanted hip and knee joint arthroplasties have to be replaced due to early or late loosening of the implant, a failure of osteointegration with fibrous tissue at the bone-implant-interface. This could be counteracted by ensuring that cells which attach to the implant surface differentiate towards bone cells afterwards. For this reason, human mesenchymal stem cells (hMSCs) will be included in this study. These cells are naturally available at the bone-implant-interface, multipotent and therefore ideal to study the osteoinductivity of a material. The goal of this pilot study was to test the cell response towards three different titanium grades with a novel surface structuring, as a first step towards achieving an improved implant surface for enhanced osteointegration. Disk-shaped titanium scaffolds with a diameter of 12 mm and a height of 1.2 mm were used. The surface topography (500 µm × 500 µm × 300 µm pores) was generated via laser treatment of the surface. By using nanosecond pulsed laser technique, a rough surface with micro- and nanostructural (titanium droplets) features was automatically formed. Three different batches made of commercially pure titanium grades 1 and 2 (Ti1/Ti2) or Ti6Al4V alloy grade 5 (Ti5) were produced. Four cell types were analysed on these batches: primary hMSCs from one donor (m, 25 y), periosteum derived cells (PDCs), human osteoblasts (hOBs) and periodontal ligament cells (PDLs). Cells were seeded on Ti1, Ti2 and Ti5 scaffolds in triplicates. Resazurin assay to examine cell viability was conducted with all cell types. Measurements were executed on several days after seeding, from day one up to day 14. Actin staining as well as live/dead staining was performed with hMSCs cultured on titanium for 1, 3, 5 or 7 days. The cell viability assay revealed early turning points of growth for osteogenic hOBs (day 3) and PDCs (day 7). HMSCs grew steadily on the material and non-osteogenic PDLs stayed in plateau throughout the cultivation period. With respect to the material, cells demonstrated better proliferation on Ti1 and Ti2 than on Ti5. Live/dead staining showed a high survival rate of hMSCs at each time point and on all three titanium grades, with a neglectable number of dead cells. Actin staining confirmed an enhanced spreading and stretching of hMSCs on Ti1 and Ti2 compared to hMSCs on Ti5. Our pilot data indicates that cells react to different titanium compositions, revealed by increased proliferation on commercially pure titanium (Ti1/2). Furthermore, our results demonstrate that osteogenic cells prefer the novel surface structuring in comparison to non-osteogenic PDL cells, which stayed in plateau. The turning points of growth (hOBs/PDCs) suggest an osteosupportiveness of the surface. Although hMSCs did not show a turning point in growth, their growth was steady and resulted in the highest number of cells along with a well stretched morphology. Due to their good proliferation and response to the material, hMSCs are currently being used for evaluating the osteogenic potential of the novel scaffolds


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_4 | Pages 70 - 70
1 Apr 2018
He X Milz S Michalke B Sprecher CM Gahlert M Röhling S Kniha H Högg C Reichl FX
Full Access

The aim of this study is to compare the release of titanium (Ti) and zirconium (Zr) into the tissue surrounding Ti- and ZrO. 2. -implants. Methyl methacrylate embedded mini pig maxillae with 6 Ti-implants and 4 ZrO2-implants were analysed after 12-weeks of implantation. The spatial distribution of elemental Ti and Zr in maxillae near implants was assessed with laser ablation (LA)-inductively coupled plasma (ICP)-mass spectrometry (MS). From each maxilla two bone slices adjacent to the implants were measured. The contents of Ti and Zr in these bone slices were determined by ICP-MS and ICP-optical emission spectrometry. Increased intensity of Ti and Zr could be detected in bone tissues at a distance of 891±398 µm (mean ± SD) from Ti-implants and 927±404 µm from ZrO2-implants. The increased intensity was mainly detected near implant screw threads. The average Ti content detected in 11 bone slices from samples with Ti-implants was 1.67 mg/kg, which is significantly higher than the Ti content detected in 8 slices from samples with ZrO. 2. -implants. The highest Ti content detected was 2.17 mg/kg. The average Zr content in 4 bone slices from samples with ZrO. 2. -implants is 0.59 mg/Kg, the other 4 bone slices showed Zr contents below the detection limit (. After 12-weeks of implantation, increased intensity of Ti and Zr can be detected in bone tissues near Ti- and ZrO. 2. -implants. The results show that Ti content released from Ti-implants is higher than the Zr content released from ZrO. 2. -implants


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 114 - 114
2 Jan 2024
Maglio M Tschon M Sartori M Martini L Rocchi M Dallari D Giavaresi G Fini M
Full Access

The use of implant biomaterials for prosthetic reconstructive surgery and osteosynthesis is consolidated in the orthopaedic field, improving the quality of life of patients and allowing for healthy and better ageing. However, there is the lack of advanced innovative methods to investigate the potentialities of smart biomaterials, particularly for the study of local effects of implant and osteointegration. Despite the complex process of osseointegration is difficult to recreate in vitro, the growing challenges in developing alternative models require to set-up and validate new approaches. Aim of the present study is to evaluate an advanced in vitro tissue culture model of osteointegration of titanium implants in human trabecular bone. Cubic samples (1.5×1.5 cm) of trabecular bone were harvested as waste material from hip arthroplasty surgery (CE AVEC 829/2019/Sper/IOR); cylindrical defects (2 mm Ø, 6 mm length) were created, and tissue specimens assigned to the following groups: 1) empty defects- CTR-; 2) defects implanted with a cytotoxic copper pin (Merck cod. 326429)- CTR+; 3) defects implanted with standard titanium pins of 6 µm-rough (ZARE S.r.l) -Ti6. Tissue specimens were cultured in mini rotating bioreactors in standard conditions, weekly assessing viability. At the 8-week-timepoint, immunoenzymatic, microtomographic, histological and histomorphometric analyses were performed. The model was able to simulate the effects of implantation of the materials, showing a drop in viability in CTR+, differently from Ti6 which appears to have a trophic effect on the bone. MicroCT and histological analysis supported the results, with lower BV/TV and Tb.Th values observed in CTR- compared to CTR+ and Ti6 and signs of matrix and bone deposition at the implant site. The collected data suggest the reliability of the tested model which can recreate the osseointegration process in vitro and can therefore be used for preliminary evaluations to reduce and refine in vivo preclinical models. Acknowledgment: This work was supported by Emilia-Romagna Region for the project “Sviluppo di modelli biologici in vitro ed in silico per la valutazione e predizione dell'osteointegrazione di dispositivi medici da impianto nel tessuto osseo”


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_7 | Pages 140 - 140
4 Apr 2023
Fry M Ren W Bou-Akl T Wu B Cizmic Z Markel D
Full Access

Extensor mechanism and abductor reconstructions in total joint arthroplasty are problematic. Growing tendon into a metallic implant would have great reconstructive advantages. With the introduction of porous metal implants, it was hoped that tendons could be directly attached to implants. However, the effects of the porous metal structure on tissue growth and pore penetration is unknown. In this rat model, we investigated the effect of pore size on tendon repair fixation using printed titanium implants with differing pore sizes. There were four groups of six Sprague Dawley rats (n = 28) plus control (n=4). Implants had pore sizes of 400µm (n=8), 700µm (n=8), and 1000µm (n=8). An Achilles tendon defect was created, and the implant positioned and sutured between the cut ends. Harvest occurred at 12-weeks. Half the specimens underwent tensile load to failure testing, the other half fixed and processed for hard tissue analysis. Average load to failure was 72.6N for controls (SD 10.04), 29.95N for 400µm (SD 17.95), 55.08N for 700µm (SD 13.47), and 63.08N for 1000µm (SD 1.87). The load to failure was generally better in the larger pore sizes. Histological evaluation showed that there was fibrous tendon tissue within and around the implant material, with collagen fibers organized in bundles. This increases as the pore diameter increases. Printing titanium implants allows for precise determination of pore size and structure. Our results showed that tendon repair utilizing implants with 700µm and 1000µm pores exhibited similar load to failure as controls. Using a defined pore structure at the attachment points of tendons to implants may allow predictable tendon to implant reconstruction at the time of revision arthroplasty


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_7 | Pages 108 - 108
4 Apr 2023
Wen Z Ding Y Lin S Li C Ouyang Z
Full Access

As peri-prosthetic aseptic loosening is one of the main causes of implant failure, inhibiting wear particles induced macrophages inflammation is considered as a promising therapy for AL to expand the lifespan of implant. Here, we aim at exploring the role of p110δ, a member of class IA PI3K family, and Krüppel-like factor 4 (KLF4) in titanium particles (TiPs) induced macrophages-inflammation and osteolysis. Firstly, IC87114, the inhibitor of p110δ and siRNA targeting p110δ were applied and experiments including ELISA and immunofluorescence assay were conducted to explore the role of p110δ. Sequentially, KLF4 was predicted as the transcription factor of p110δ and the relation was confirmed by dual luciferase reporter assay. Next, assays including RT-PCR, western blotting and flow cytometry were performed to ensure the specific role of KLF4. Finally, TiPs-induced mice cranial osteolysis model was established, and micro-CT scanning and immunohistochemistry assay were performed to reveal the role of p110δ and KLF4 in vivo. Here, we found that p110δ was upregulated in TiPs-stimulated macrophages. The inhibition of p110δ or knockdown of p110δ could significantly dampen the TiPs-induced secretion of TNFα and IL-6. Further mechanistic studies confirmed that p110δ was responsible for TNFα and IL-6 trafficking out of Golgi complex without affecting their expression in TiPs-treated macrophages. Additionally, we explored the upstream regulators and confirmed that Krüppel-like factor 4 (KLF4) was the transcription repressor of p110δ. Apart from that, KLF4, targeted by miR-92a, could also attenuate TiPs-induced inflammation by mediating NF-κB pathway and M1/M2 polarization. By the establishment of TiPs-induced mice cranial osteolysis model, we found that KLF4 knockdown exacerbated TiPs-induced osteolysis which was strikingly ameliorated by knockdown of p110δ. In summary, our study suggests the key role of miR-92a/KLF4/p110δ signal in TiPs-induced macrophages inflammation and osteolysis


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_1 | Pages 22 - 22
1 Jan 2017
Pacha-Olivenza M García-Alonso M Tejero R Escudero M Gallardo Moreno A González-Martín M
Full Access

Adhered bacteria on titanium surfaces are able to decrease its corrosion potential and impedance values at the lowest frequencies. This result points to the detrimental influence of the biofilm on the passive film formed on the surfaces, independently on the surface finishes. Titanium is one of the most used metallic biomaterials for biological and implant applications. The spontaneous formation of a protective passive film around 2–5 nm thick, make titanium unique as a biomaterial for implants. Its composition has been described by a three-layer model: TiO2/Ti2O3/TiO and its stability is ultimately responsible for the success of osseointegrated titanium implants. The cases of breakdown of the protective passive film are associated with highly acidic environments induced by bacterial biofilms and/or inflammatory processes that lead to localized corrosion of titanium and, in extreme cases, implant failure. Bearing in mind that the surface design of a titanium implant is a key element involved in the healing mechanisms at the bone-implant interface, the surface modifications have sought to enhance the biomechanical anchorage of the implant and promote osseointegration at the cell-biomolecular level. However, little attention has been paid to the effects of these surface modifications in the microbiologically induced corrosion (MIC). The aim of this work is to evaluate the potential for MIC of titanium in the short term under viable bacterial cells of Streptococcus mutansas a representative microorganism of oral biofilm considered to be a highly cariogenic pathogen. Discs of 64 mm. 2. surface area of commercially pure titanium, grade 4, were supplied by Biotechnology Institute (BTI, Vitoria, Spain). Four surface treatments were studied: two acid etchings (low roughness, opN and high roughness, opV). In addition, acid etched plus anodic oxidation (opNT). For comparative purposes, two surface finishes have been included: high roughness – corresponding with sandblasting-large grit plus acid (SLA); and, as-machined titanium (mach). The oral strain used for assessing the biofilm formation on the corrosion behavior of Ti surfaces was Streptococus mutansATCC 25175, obtained from the Spanish Type Culture Collection (CECT). The study of MIC from Streptococcus mutanson surfaces of Ti was carried out in an electrochemical cell specifically designed and patented by some of the present authors [1]. A three set up configuration of the electrochemical cell was used in the experiments. The measurement of the corrosion potential and electrochemical impedance was performed at different periods of incubation of bacteria: 2, 7, 15, 21 and 28 days. Out Slight but continuous decrease in the corrosion potential and impedance values at the lowest frequencies indicate the deleterious influence of the biofilm on the passive film formed on the surfaces, independently on the surface finishes. This research suggests that the most appropriate surface modification for the dental implant portion at the bone level would be the acid etched of high roughness (opV) surface


Bone & Joint Research
Vol. 1, Issue 6 | Pages 125 - 130
1 Jun 2012
Bøe BG Støen RØ Solberg LB Reinholt FP Ellingsen JE Nordsletten L

Objectives. An experimental rabbit model was used to test the null hypothesis, that there is no difference in new bone formation around uncoated titanium discs compared with coated titanium discs when implanted into the muscles of rabbits. Methods. A total of three titanium discs with different surface and coating (1, porous coating; 2, porous coating + Bonemaster (Biomet); and 3, porous coating + plasma-sprayed hydroxyapatite) were implanted in 12 female rabbits. Six animals were killed after six weeks and the remaining six were killed after 12 weeks. The implants with surrounding tissues were embedded in methyl methacrylate and grinded sections were stained with Masson-Goldners trichrome and examined by light microscopy of coded sections. Results. Small amounts of bone were observed scattered along the surface of five of the 12 implants coated with porous titanium, and around one out of 12 porous coated surfaces with Bonemaster. No bone formation could be detected around porous coated implants with plasma-sprayed hydroxyapatite. Conclusion. Porous titanium coating is to some degree osteoinductive in muscles


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 81 - 81
2 Jan 2024
Vautrin A Aw J Attenborough E Varga P
Full Access

Although 3D-printed porous dental implants may possess improved osseointegration potential, they must exhibit appropriate fatigue strength. Finite element analysis (FEA) has the potential to predict the fatigue life of implants and accelerate their development. This work aimed at developing and validating an FEA-based tool to predict the fatigue behavior of porous dental implants. Test samples mimicking dental implants were designed as 4.5 mm-diameter cylinders with a fully porous section around bone level. Three porosity levels (50%, 60% and 70%) and two unit cell types (Schwarz Primitive (SP) and Schwarz W (SW)) were combined to generate six designs that were split between calibration (60SP, 70SP, 60SW, 70SW) and validation (50SP, 50SW) sets. Twenty-eight samples per design were additively manufactured from titanium powder (Ti6Al4V). The samples were tested under bending compression loading (ISO 14801) monotonically (N=4/design) to determine ultimate load (F. ult. ) (Instron 5866) and cyclically at six load levels between 50% and 10% of F. ult. (N=4/design/load level) (DYNA5dent). Failure force results were fitted to F/F. ult. = a(N. f. ). b. (Eq1) with N. f. being the number of cycles to failure, to identify parameters a and b. The endurance limit (F. e. ) was evaluated at N. f. = 5M cycles. Finite element models were built to predict the yield load (F. yield. ) of each design. Combining a linear correlation between FEA-based F. yield. and experimental F. ult. with equation Eq1 enabled FEA-based prediction of F. e. . For all designs, F. e. was comprised between 10% (all four samples surviving) and 15% (at least one failure) of F. ult. The FEA-based tool predicted F. e. values of 11.7% and 12.0% of F. ult. for the validation sets of 50SP and 50SW, respectively. Thus, the developed FEA-based workflow could accurately predict endurance limit for different implant designs and therefore could be used in future to aid the development of novel porous implants. Acknowledgements: This study was funded by EU's Horizon 2020 grant No. 953128 (I-SMarD). We gratefully acknowledge the expert advice of Prof. Philippe Zysset


The Journal of Bone & Joint Surgery British Volume
Vol. 87-B, Issue 8 | Pages 1157 - 1163
1 Aug 2005
Peter B Zambelli P Guicheux J Pioletti DP

In an attempt to increase the life of cementless prostheses, an hydroxyapatite-coated implant which releases a bisphosphonate has been suggested as a drug-delivery system. Our in vitro study was designed to determine the maximum dose to which osteoblasts could be safely exposed. Our findings demonstrated that zoledronate did not impair the proliferation of human osteoblasts when used at concentrations below 1 μ. m. Murine cells can be exposed to concentrations as high as 10 μ. m. . A concentration of 0.01% of titanium particles did not impair the proliferation of either cell line. Zoledronate affected the alkaline phosphatase activity of murine osteoblasts through a chelation phenomenon. The presence of titanium particles strongly decreased the alkaline phosphatase activity of murine osteoblasts. We did not detect any synergic effect of zoledronate and titanium particles on the behaviour of both human and murine osteoblasts


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_2 | Pages 32 - 32
2 Jan 2024
Depboylu F Yasa E Poyraz Ö Korkusuz F
Full Access

Decreasing the bulk weight without losing the biomechanical properties of commercial pure titanium (Cp-Ti) medical implants is now possible by using Laser Powder Bed Fusion (L-PBF) technology. Gyroid lattice structures that have precious mechanical and biological advantages because of similarity to trabecular bone. The aim of the study was to design and develop L-PBF process parameter optimization for manufacturing gyroid lattice Cp-Ti structures. The cleaning process was then optimized to remove the non-melted powder from the gyroid surface without mechanical loss. Gyroid cubic designs were created with various relative densities by nTopology. L-PBF process parameter optimization was progressed using with Cp-Ti (EOS TiCP Grade2) powder in the EOS M290 machine to achieve parts that have almost full dense and dimensional accuracy. The metallography method was made for density. Dimensional accuracy at gyroid wall thicknesses was investigated between designed and manufactured via stereomicroscope, also mechanical tests were applied with real time experiment and numerical analysis (ANSYS). Mass loss and strut thickness loss were investigated for chemical etching cleaning process. Gyroid parts had 99,5% density. High dimensional accuracy was achieved during L-PBF process parameters optimization. Final L-PBF parameters gave the highest 19% elongation and 427 MPa yield strength values at tensile test. Mechanical properties of gyroid were controlled with changing relative density. A minute chemical etching provided to remove non-melted powders. Compression test results of gyroids at numerical and real-time analysis gave unrelated while deformation behaviors were compatible with each other. Gyroid Cp-Ti osteosynthesis mini plates will be produced with final L-PBF process parameters. MTT cytotoxicity test will be characterized for cell viability. Acknowledgements This project is granted by TUBITAK (120N943). Feza Korkusuz MD is a member of the Turkish Academy of Sciences (TÜBA)


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_9 | Pages 74 - 74
17 Apr 2023
Theodoridis K Hall T Munford M Van Arkel R
Full Access

The success of cementless orthopaedic implants relies on bony ingrowth and active bone remodelling. Much research effort is invested to develop implants with controllable surface roughness and internal porous architectures that encourage these biological processes. Evaluation of these implants requires long-term and costly animal studies, which do not always yield the desired outcome requiring iteration. The aim of our study is to develop a cost-effective method to prescreen design parameters prior to animal trials to streamline implant development and reduce live animal testing burden. Ex vivo porcine cancellous bone cylinders (n=6, Ø20×12mm) were extracted from porcine knee joints with a computer-numerically-controlled milling machine under sterile conditions within 4 hours of animal sacrifice. The bone discs were implanted with Ø6×12mm additive manufactured porous titanium implants and were then cultured for 21days. Half underwent static culture in medium (DMEM, 10% FBS, 1% antibiotics) at 37°C and 5% CO. 2. The rest were cultured in novel high-throughput stacked configuration in a bioreactor that simulated physiological conditions after surgery: the fluid flow and cyclic compression force were set at 10ml/min and 10–150 N (1Hz,5000 cycles/day) respectively. Stains were administered at days 7 and 14. Samples were evaluated with widefield microscopy, scanning electron microscopy (SEM) and with histology. More bone remodelling was observed on the samples cultured within the bioreactor: widefield imaging showed more remodelling at the boundaries between the implant-bone interface, while SEM revealed immature bone tissue integration within the pores of the implant. Histological analysis confirmed these results, with many more trabecular struts with new osteoid formation on the samples cultured dynamically compared to static ones. Ex vivo bone can be used to analyse new implant technologies with lower cost and ethical impact than animal trial. Physiological conditions (load and fluid flow) promoted bone ingrowth and remodelling


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_11 | Pages 33 - 33
1 Dec 2020
Diez-Escudero A Andersson BM Järhult JD Hailer NP
Full Access

Uncemented implants combining antimicrobial properties with osteoconductivity would be highly desirable in revision surgery due to periprosthetic joint infection (PJI). Silver coatings convey antibacterial properties, however, at the cost of toxicity towards osteoblasts. On the other hand, topological modifications such as increased surface roughness or porosity support osseointregation but simultaneously lead to enhanced bacterial colonization. In this study, we investigated the antibacterial and osteoconductive properties of silver-coated porous titanium (Ti) alloys manufactured by electron beam melting, rendering a macrostructure that mimics trabecular bone. Trabecular implants with silver coating (TR-Ag) or without coating (TR) were compared to grit-blasted Ti6Al4V (GB) and glass cover slips as internal controls. Physicochemical characterization was performed by X-ray diffraction (XRD) and energy dispersive X-rays (EDX) together with morphological characterization through electron scanning microscopy (SEM). Bacterial adherence after incubation of samples with Staphylococcus (S.) aureus and S. epidermidis strains harvested from PJI patients was quantitatively assessed by viable count after detachment of adherent bacteria by collagenase/dispase treatment. Primary human osteoblasts (hOB) were used to investigate the osteoconductive potential by lactate dehydrogenase (LDH) and alkaline phosphatase (ALP) activity. Cell morphology was investigated by fluorescence microscopy after staining with carboxifluorescein diacetate succinimidyl ester (CFDA-SE) and 4′,6-diamidino-2-phenylindole (DAPI). The trabecular implants depicted a porosity of 70% with pore sizes of 600µm. The amount of silver analyzed by EDX accounted for 35%wt in TR-Ag but nil in TR. Silver-coated TR-Ag implants had 24% lower S. aureus viable counts compared to non-coated TR analogues, and 9% lower compared to GB controls. Despite trabecular implants, both with and without silver, had higher viable counts than GB, the viable count of S. epidermidis was 42% lower on TR-Ag compared to TR. The percentage of viable hOB, measured by LDH and normalized to controls and area at 1 day, was lower on both TR-Ag (18%) and on TR (13%) when compared with GB (89%). However, after 1 week, cell proliferation increased more markedly on trabecular implants, with a 5-fold increase on TR-Ag, a 3.4-fold increase on TR, and a 1.7-fold increase on GB. Furthermore, after 2 weeks of hOB culture, proliferation increased 20-fold on TR-Ag, 29-fold on TR, and 3.9-fold for GB, compared to 1 day. The osteoconductive potential measured by ALP illustrated slightly higher values for TR-Ag compared to TR at 1 day and 2 weeks, however below those of GB samples. Cell morphology assessed by microscopy showed abundant growth of osteoblast-like cells confined to the pores of TR-Ag and TR. Overall, our findings indicate that the silver coating of trabecular titanium exerts limited cytotoxic effects on osteoblasts and confers antimicrobial effects on two PJI-relevant bacterial strains. We conclude that improving material design by mimicking the porosity and architecture of cancellous bone can enhance osteoconductivity while the deposition of silver confers potent antimicrobial properties


Bone & Joint Research
Vol. 6, Issue 2 | Pages 108 - 112
1 Feb 2017
Itabashi T Narita K Ono A Wada K Tanaka T Kumagai G Yamauchi R Nakane A Ishibashi Y

Objectives. The surface of pure titanium (Ti) shows decreased histocompatibility over time; this phenomenon is known as biological ageing. UV irradiation enables the reversal of biological ageing through photofunctionalisation, a physicochemical alteration of the titanium surface. Ti implants are sterilised by UV irradiation in dental surgery. However, orthopaedic biomaterials are usually composed of the alloy Ti6Al4V, for which the antibacterial effects of UV irradiation are unconfirmed. Here we evaluated the bactericidal and antimicrobial effects of treating Ti and Ti6Al4V with UV irradiation of a lower and briefer dose than previously reported, for applications in implant surgery. Materials and Methods. Ti and Ti6Al4V disks were prepared. To evaluate the bactericidal effect of UV irradiation, Staphylococcus aureus 834 suspension was seeded onto the disks, which were then exposed to UV light for 15 minutes at a dose of 9 J/cm. 2. To evaluate the antimicrobial activity of UV irradiation, bacterial suspensions were seeded onto the disks 0, 0.5, one, six, 24 and 48 hours, and three and seven days after UV irradiation as described above. In both experiments, the bacteria were then harvested, cultured, and the number of colonies were counted. Results. No colonies were observed when UV irradiation was performed after the bacteria were added to the disks. When the bacteria were seeded after UV irradiation, the amount of surviving bacteria on the Ti and Ti6Al4V disks decreased at 0 hours and then gradually increased. However, the antimicrobial activity was maintained for seven days after UV irradiation. Conclusion. Antimicrobial activity was induced for seven days after UV irradiation on both types of disk. Irradiated Ti6Al4V and Ti had similar antimicrobial properties. Cite this article: T. Itabashi, K. Narita, A. Ono, K. Wada, T. Tanaka, G. Kumagai, R. Yamauchi, A. Nakane, Y. Ishibashi. Bactericidal and antimicrobial effects of pure titanium and titanium alloy treated with short-term, low-energy UV irradiation. Bone Joint Res 2017;6:108–112. DOI: 10.1302/2046-3758.62.2000619


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_8 | Pages 93 - 93
1 Apr 2017
Karaaslan A Karakaşlı A Ertem F Aycan H
Full Access

Background. Intramedullary nailing is a widely accepted treatment method for femoral fractures. Failure of locking screws is often a threatening complication, particularly on comminuted fractures. For comminuted fractures, the locking nails are load-bearing devices. The load transfer between fractured fragments is made through especially the locking screws for these load bearing situations. Nonunion, malunion, delayed union, shortening, and nail migration are the expected results if early failure of locking screws is present with comminuted fractures. In this study our aim was to compare the bending resistance of titanium and stainless steel locking screws. Methods. We tested 60 locking screws in six groups (titanium, stainless steel, unthreaded, low threaded and high threaded) in a steel tube that has 30 mm inner diameter, which imitates the lesser trochanter level. We determined the yield points at three-point bending tests that permanent deformation started in the locking screws using an axial compression testing machine. Results. The three-point bending resistance of 5 mm low threaded titanium locking screws (bending at 1413 N loading) was 46.5 % less than the three-point bending resistance of 5 mm low threaded stainless steel locking screws (bending at 2171 N loading) (p < 0.001). Five mm stainless steel locking screws are 29–57 % more resistant to three-point bending deformation than titanium ones. Conclusions. Therefore, stainless steel locking screws instead of titanium ones must be preferred in comminuted femur shaft fractures. In intramedullary nailing of comminuted or long oblique femur fractures, a locking screw should be 5 mm low threaded or unthreaded stainless steel or 5 mm unthreadedtitanium. Five mm high threaded titanium or stainless steel screws must not ever be used as a locking screw. Level of Evidence. 5. Disclosure. Authors declare that there is no conflict of interest related to the present study


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_9 | Pages 76 - 76
1 May 2017
De Pasquale D Beraudi A Stea S Baleani M Guerra G Toni A
Full Access

Modular femoral stems for total hip arthroplasty (THA) were introduced to allow additional options for surgeons in controlling leg lengths, offset, and implant stability. This option is widely used in Region Emilia Romagna, Italy, where the study was conducted, having a modular neck stem nearly 35% of primary THA in 2013. Great majority of modular neck is made of Titanium alloy. The study was designed as a retrospective descriptive case series of 67 hips in patients who underwent revision of a THA. All had a Titanium modular neck. In 44 cases revision was due to breakage of the neck, in the remaining 23 it was due to different reasons unrelated to modular neck such as bone fracture, breakage of a ceramic component, cup loosening. Mean follow up was 3.5 yrs. For all patients excised capsule and surrounding tissue were graded for presence of necrosis, inflammatory exudate, lymphocytes, and wear particles using light microscopy of routine paraffin sections stained with hematoxylin and eosin. The retrieved modular neck-body and head-neck junctions were examined for evidence of fretting and corrosion. For some patient dosage of circulating Titanium was obtained. Approval was obtained from institutional review board. It resulted that a variable amount of wear was observed in the first group of patients, with no evidence of lymphocytic reaction, but with variable notes of necrosis. Broken necks showed different patterns of damage, with different degree of corrosion, beside the fatigue fracture. In the second group wear was less evident or absent and negativity of lymphocyte reaction was substantially confirmed. Circulating Titanium ions were one order of magnitude higher in first group (mean 35 micrograms /litre). It can be concluded that fracture of Titanium modular necks occurs progressively, wear does not induce lymphocytic reaction and circulating ions increase. Level of Evidence. III retrospective, comparative study. Acknowledgments. The research was funded by Ministry of Health, grant ‘Early diagnosis of pending failure…’RF 20091472961


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_8 | Pages 72 - 72
11 Apr 2023
Stich T Kovářík T Křenek T Alt V Docheva D
Full Access

The goal was to analyze the cellular response, specifically the osteogenic capacity, of titanium (Ti) implants harbouring a novel laserbased-surface-structure with the overall aim: augmented osteointegration. Surface micro-/nanoproperties greatly influence cell behaviour at the tissue-implant-interface and subsequent osteointegration. We investigated Ti-materials subjected to a specially developed shifted-Laser-Surface-Texturing (sLST) technology and compared them to a standard roughening-technique (sand-blasting-acid-etching, SLA). The biological response was evaluated with hMSCs, which are naturally available at the bone-implant-interface. We hypothesized: the novel surface is beneficial for our three different (young/healthy-YH; aged/healthy-AH;aged/osteoporotic-OP) cohorts. The sLST was performed using a SPI-G3-series laser (beam-wavelength=1064nm, pulse-duration=200ns). For the SLA surface, Ti was sandblasted, afterwards acid-etched (HCl/H2SO4). Three different hMSC cohorts were studied: YH: n=6,29±6; AH: n=5,79±5; OP: n=5,76±5 years (osteoporosis confirmed via DEXA-scan). OP hMSCs show e.g. ColI-deficient-matrix and decreased mineralization. Cells were examined for survival, cell proliferation and cytoskeleton arrangement. Osteogenic differentiation was carried out over 21 days, matrix mineralization was validated with Alizarin-Red-S-staining and quantification. Laser-texturing generated precisely the desired microgeometry. On nanostructural level, differently-sized Ti-droplets were formed stochastically by laser-induced-Ti-plasma. Live/dead-/Actin-stainings showed comparable results for all cohorts and surfaces in terms of survival and cell shape. On Ti-materials, cell growth showed no significant difference between the 3 cohorts. Alizarin quantification revealed the highest levels on laser-textured-surfaces; highest value for YH, followed by AH, lastly OP; no significance between AH/YH, but between OP/YH (p<0.0001). However, mineralization of all cohorts cultured on laser-textured-surfaces increased significantly (p<0.0001) compared to respective SLA-group, with >20fold higher value in the OP-cohort (AH:11fold, YH:6fold). The data proves the biocompatibility of the laser-structured-Ti for young+aged cohorts. Osteogenic differentiation was significantly augmented on laser-treated-Ti. Most intriguingly, OP-donors could reach manifold increased mineralization, suggesting the novel laser texturing can counteract the osteoporotic phenotype. As osteogenesis-enhancing capacities may be related to mechanisms controlling cellular shape/fate, further investigations referring to this are currently ongoing. In conclusion, our laser-textured-Ti-materials are safe, can have a demand-oriented designer-surface-topography and represent a great potential for development into next-generation-implants suitable for different patient-cohorts, especially osteoporosis patients