Advertisement for orthosearch.org.uk
Results 1 - 20 of 20
Results per page:
The Bone & Joint Journal
Vol. 97-B, Issue 5 | Pages 582 - 589
1 May 2015
Brennan SA Ní Fhoghlú C Devitt BM O’Mahony FJ Brabazon D Walsh A

Implant-associated infection is a major source of morbidity in orthopaedic surgery. There has been extensive research into the development of materials that prevent biofilm formation, and hence, reduce the risk of infection. Silver nanoparticle technology is receiving much interest in the field of orthopaedics for its antimicrobial properties, and the results of studies to date are encouraging. Antimicrobial effects have been seen when silver nanoparticles are used in trauma implants, tumour prostheses, bone cement, and also when combined with hydroxyapatite coatings. Although there are promising results with in vitro and in vivo studies, the number of clinical studies remains small. Future studies will be required to explore further the possible side effects associated with silver nanoparticles, to ensure their use in an effective and biocompatible manner. Here we present a review of the current literature relating to the production of nanosilver for medical use, and its orthopaedic applications. Cite this article: Bone Joint J 2015; 97-B:582–9


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_3 | Pages 39 - 39
1 Mar 2021
Aziz M Rosenzweig D Weber M
Full Access

Great strides have been made in the early detection and treatment of cancer which is resulting in improved survivability and more Canadians living with cancer. Approximately 80% of primary breast, lung, and prostate cancers metastasize to the spine. Poly-methyl methacrylate (PMMA) bone cement is one of the most commonly used bone substitutes in spine surgery. In clinical practice it can be loaded with various drugs, such as antibiotics or chemotheraputic drugs, as a means of local drug delivery. However, studies have shown that drugs loaded into PMMA cement tend to release in small bursts in the first 48–72 hours, and the remaining drug is trapped without any significant release over time. The objective of this study is to develop a nanoparticle-functionalized PMMA cement for use as a sustained doxorubicin delivery device. We hypothesize that PMMA cement containing mesoporous silica nanoparticles will release more doxorubicin than regular PMMA. High viscosity SmartSet ™ PMMA cement by DePuy Synthes was used in this study. The experimental group consisted of 3 replicates each containing 0.24 g of mesoporous silica nanoparticles, 1.76 g of cement powder, 1ml of liquid cement monomer and 1 mg of doxorubicin. The control group consisted 3 replicates each containing 2.0 g of cement powder, 1ml of liquid cement monomer and 1 mg of doxorubicin. The experimental group contained an average of 8.18 ± 0.008 % (W/W) mesoporous silica nanoparticles. Each replicate was casted into a cylindrical block and incubated in a PBS solution which was changed at predetermined intervals for 45 days. The concentration of eluted doxorubicin in each solution was measured using a florescent plate reader. The mechanical properties of cement were assessed by unconfined compression testing. The effect of the doxorubicin released from cement on prostate and breast tumor cell metabolic activity was assessed using the Alamar Blue test. After 45 days the experimental group released 3.24 ± 0.25 % of the initially loaded doxorubicin which was more than the 2.12 ± 0.005% released by the control group (p 0.03). There was no statistically significant difference in Young's elasticity modulus between groups (p 0.53). Nanoparticle functionalized PMMA suppressed the metabolic activity of prostate cancer by more than 50 percent but did not reach statistical significance. Nanoparticle functionalized PMMA suppressed the metabolic activity of breast cancer cells by 69 % (p < 0.05). Nanoparticle-functionalized PMMA cement can release up to 1.53 times more doxorubicin than the standard PMMA. The use of mesoporous silica nanoparticles to improve drug release from PMMA cement shows promise. In the future, in vivo experiments are required to test the efficacy of released doxorubicin on tumor cell growth


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_4 | Pages 98 - 98
1 Feb 2017
Lal S Hall R Tipper J
Full Access

Introduction. Particle-induced oxidative stress in cells is a unifying factor that determines toxicity and carcinogenicity potential in biomaterials. A previous study by Bladen et al. showed the production of significant levels of reactive oxygen species (ROS) following the stimulation of phagocytes by UHMWPE and CoCr wear debris [1]. Latest generation bearing materials such as silicon nitride also need to be tested for potential generation of ROS in phagocytic cells. This study aimed to investigate the production of reactive oxygen species in L929 fibroblasts stimulated with clinically relevant doses of nanoscale and micron-sized silicon nitride (Si. 3. N. 4. ) particles, silica nanoparticles, and CoCr wear debris. Silica nanoparticles were included as a comparison material for situations where the Si. 3. N. 4. particle's surface are oxidised to silicon dioxide [2]. Materials and Methods. Si. 3. N. 4. particles (<50 nm and <1 µm, Sigma), silica nanopowder (<100 nm, Sigma) and clinically relevant CoCr wear particles were heat-treated at 180°C for 4 h to remove endotoxin. Particles were then re-suspended in sterile water by sonication. L929 murine fibroblasts were cultured with low doses (0.5 µm. 3. /cell) and high doses (50 µm. 3. /cell) of Si. 3. N. 4. particles, and high doses (50 µm. 3. /cell) of silica nanoparticles and CoCr wear debris. Cells were incubated for three and six days at 37°C with 5% (v/v) CO. 2. tert-Butyl hydroperoxide (TBHP) was used as a positive control for the production of ROS in the cells. Intracellular ROS was measured using Image-IT LIVE kit (Invitrogen). This assay is based on carboxy-2',7'-dichlorodihydro-fluorescein diacetate (carboxy-H2DCFDA), which forms a non-fluorescent derivative by intracellular esterases and then reacts with intracellular ROS to form green fluoroscence producing derivative carboxy- dichlorodihydro-fluorescein. Images were captured using a confocal microscope and analysed using ImageJ for corrected total cell fluorescence (CTCF). The results were expressed as mean ± 95% confidence limits and the data was analysed using one-way ANOVA and Tukey-Kramer post-hoc tests. Results and Discussion. Si. 3. N. 4. nanoparticles significantly reduced the ROS levels in L929 fibroblasts at low doses (0.5 μm. 3. /cell) and high doses (50 μm. 3. /cell) over a period of six days; whereas no significant change in the levels of ROS was observed in cells treated with micron-sized Si. 3. N. 4. particles [Figure 1]. Only a few cells treated with high doses of CoCr wear particles (50 μm. 3. /cell) survived for up to six days and produced significantly higher levels of ROS [Figure 1, 2]. Interestingly, cells challenged with high doses (50 μm. 3. /cell) of Si. 3. N. 4. and silica nanoparticles produced statistically similar levels of ROS in cells [Figure 1]. This might be due to the potential surface oxidation of Si. 3. N. 4. nanoparticles, which makes their surface chemistry and biological identity similar to silica nanoparticles. Conclusion. Unlike existing implant materials such as UHMWPE and CoCr, silicon nitride has demonstrated the capacity to reduce or maintain normal levels of ROS in macrophages depending on the particle size and dose. Acknowledgements. The research leading to these results has received funding from the European Union's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. GA-310477 LifeLongJoints


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXI | Pages 159 - 159
1 May 2012
P. NS B. Q R. L
Full Access

Aim. Ultrahigh molecular weight polyethylene (UHMWPE) has been used for many years as a bearing surface in total joint replacement (TJR). However, late-state failure in TJR is predominantly caused by osteolysis mediated by wear particles. We tested our hypothesis that UHMWPE nanoparticles are important determinants in activating dendritic cells (DCs). Methods. UHMWPE wear particles generated from a knee simulator were profiled using an atomic force microscopy and fractionated into six fractions: 0.05-0.2, 0.2-0.8, 0.8-1, 1-5, 5-10, and 10-20 micrometer. Effects of each fraction, a mixture of nano-sized fractions, and a mixture of all fractions on the activation of mice spleen DCs were determined using flow cytometry with specific antibodies of anti-CD11c-APC, anti-CD80-PE, anti-CD11b-PerCp, anti-CD86-Biotin and streptavidin-FITC. Supernatant from DCs treated with wear particles were assayed for IL-1beta, IL-6, IL-12/23, TNF-alpha and IFN-gamma. Activation of human osteoclasts (OCs) by wear particles were determined using TRAP stain. Results. DCs treated with a mixture of nanoparticles showed a significant increase in CD80 expression. A similar trend was not observed when DCs were treated with solvent or media, suggesting that the increased expression of CD80 was UHMWPE nanoparticle specific. Macrophages treated with nanoparticles did not show a significant increase in the expression of CD80, suggesting that DCs may be more sensitive to activation than macrophages. These results were further supported by the increased production of cytokines, IL-1beta and IL-6. Furthermore, the mixture of nanoparticles and the mixture of all fractions directly stimulated maturation of OCs. Conclusion. This study identifies a novel mechanism where UHMWPE nanoparticles activate DCs. The high proportion of nanoparticles from prosthetic joints would suggest this mechanism is a likely pathway for cytokine production and OCs maturation, all of which involve osteolysis. The nanoparticles as mediators of periprosthetic inflammation should be considered in developing biomaterials for bearing surfaces


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_5 | Pages 86 - 86
1 Apr 2018
Xu J Zeng L Knight M Shelton J
Full Access

Introduction. Wear debris and metal ions originating from metal on metal hip replacements have been widely shown to recruit and activate macrophages. These cells secrete chemokines and pro-inflammatory cytokines that lead to an adverse local tissue reaction (ALTR), frequently requiring early revision. The mechanism for this response is still poorly understood. It is well documented that cobalt gives rise to apoptosis, necrosis and reactive oxygen species generation. Additionally, cobalt stimulates T cell migration, although the effect on macrophage motility remains unknown. This study tests the hypothesis that cobalt ions and nanoparticles affect macrophage migration stimulating an ALTR. Methods. This study used Co. 2+. ions (200µM) and cobalt nanoparticles (CoNPs, 100µM, 2–60nm diameter). PMA differentiation of the U937 cell line was used as macrophage-like cells. The effect of cobalt on macrophage migration was investigated by live cell imaging. After 12 hours of each treatment, timelapse images of 20 cells were collected over a 6 hour period with images captured every 5 min. Migration of individual cells was tracked in 2D using ImageJ software. The transwell migration assay was also applied to study the effect of cobalt on macrophage directional migration. U937 cells in serum free medium were added to the upper chamber of a 8µm pore size Transwell insert in the presence of cobalt, whilst the lower chamber was filled with medium plus 10% FBS. After 6 hours treatment, cells remaining on the membrane were fixed, stained with crystal violet and counted. Cellular F-actin and podosomes were visualized by labeling with TRITCconjugated phalloidin and anti-vinculin antibody after 12 hours of cobalt exposure (Co. 2+. and CoNPs). Results. Cells incubated with cobalt ions and nanoparticles showed a substantial reduction in cell migration compared with control cells. The total migration path length of cells treated with Co. 2+. (362.4±96.6µm) and CoNPs (217.3±128.1µm) were significantly shorter than those for untreated cells (801.1±198.3µm). The ability of macrophages to migrate through the transwell membrane was significantly impaired by pre-treatment with cobalt, with 16±4 and 18± migrated cells/field for Co. 2+. and CoNPs respectively with the control at 42±7 migrated cells/field. In addition, cobalt influenced macrophage morphology and actin cytoskeletal organization with a dramatic increase in the presence of intracellular podosome-type adhesions structure. Discussion. Co. 2+. ions and nanoparticles dramatically inhibited the migration of U937 macrophages in contrast to the enhanced migration reported for T cells. We propose that macrophages recruited into the area of CoCr implants would lose their responsiveness to migration signals and be retained in situ due to cobalt-induced cytoskeleton rearrangement. This enhanced macrophage accumulation and cobalt-induced formation of podosomes may therefore represent a mechanism through which cobalt wear debris and metal ions from joint prostheses exacerbate the ALTR leading to revision surgery


Orthopaedic Proceedings
Vol. 101-B, Issue SUPP_5 | Pages 25 - 25
1 Apr 2019
Cazzola M Ferraris S Stella B Orlygsson G Ng CH Cempura G Scolaro C Prenesti E Yamaguchi S Pezzotti G Cochis A Rimondini L Spriano S
Full Access

In order to improve fast osseointegration, to modulate inflammatory response and to avoid biofilm formation, several attempts of surface modifications of titanium alloy in term of surface topography and chemistry have been performed over years, but this is still an open issue. In our research work, a patented chemical treatment was developed and tailored to improve fast osseointegration and to allow further surface functionalization in order to get a multifunctional surface. After the chemical treatment, Ti6Al4V shows a micro and nano-textured surface oxide layer with high density of hydroxyls groups, as summarized Figure 1: it is able to induce apatite precipitation (during soaking in Simulated Body Fluid), high wettability by blood, specific protein adsorption, positive osteoblast response and surface mechanical resistance to implantation friction. Hydroxyl groups exposed by the treated surface also allow binding natural biomolecules such as polyphenols, which can further improve the rate and quality of osseointegration by adding anti-inflammatory, antibacterial and antitumoral effects suitable for implants in critical situations. Polyphenols have the further added value of being a low cost and eco-sustainable product, extractable from byproducts of wine and food industry. On the chemically treated and functionalized samples, the surface characterization was performed using Folin&Ciocalteu test, fluorescence microscopy and XPS analysis in order to check the presence and activity of the grafted biomolecules (polyphenols from red grape pomace and green tea leaves). Cell tests were performed with Kusa A-1 cells highlighting the ability of polyphenols to improve osteoblasts differentiation and deposition of mineralized extracellular matrix. Surface functionalization can also be performed with chitin derived biomolecules to reduce inflammation. With the purpose of obtaining the antibacterial effect, during the chemical treatment a silver precursor can also be added to obtain in situ reduced silver nanoparticles embedded in the nano-structured oxide layer. The samples containing nanoparticles on the surface were characterized by means of TEM and FESEM observation highlighting the presence of well distributed and small-sized nanoparticles on the surface and through the thickness of the oxide layer. A long-lasting release in water was observed up to 14 days and antibacterial tests on Staphylococcus aureus showed the ability of the surface to reduce bacteria viability avoiding biofilm formation. The results showed that the patented chemical treatment can improve the response of osteoblasts to titanium alloy implants, but is also a promising way to obtain multifunctional surfaces with antibacterial, antioxidant, anti-inflammatory and antitumoral properties that can be the future of orthopedic implants


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_17 | Pages 28 - 28
24 Nov 2023
De Vecchi E Balzano V Bottagisio M Gavioli L
Full Access

Aim. Antibacterial activity of coatings based on metal and metal oxide nanoparticles (NPs) often depends on materials and biotic targets resulting in a material-specific killing activity of selected Gram-positive and Gram-negative bacteria, including drug-resistant strains. In this perspective, the NPs loading amount, the relative elemental concentration inside the nanogranular building blocks and the deposition method are of paramount importance when the goal is to widen the antimicrobial spectrum, but at the same time to avoid high levels of metal content to limit undesired toxic effects. Aim of the present study was evaluation of the antimicrobial properties of two multielement nanogranular coatings composed of Titanium-Silver and Copper and of Magnesium-Silver and Copper. Method. Ti-Ag-Cu and Mg-Ag-Cu NPs were deposited on circular cover glasses (VWR) by Supersonic Cluster Beam Deposition. Biofilm-producer strains of Staphylococcus aureus (methicillin susceptible and resistant), Staphylococcus epidermidis (methicillin susceptible and resistant), Escherichia coli (fully susceptible and producer of extended spectrum beta lactamases), and Pseudomonas aeruginosa (susceptible and multidrug-resistant) were selected. The abilities of the selected strains to adhere, colonize and produce biofilm on the discs coated with Ti-Ag-Cu or Mg-Ag-Cu NPs were compared to uncoated circular cover glasses which were used as growth control. Cytotoxicity was also evaluated in order to assess the biocompatibility of the newly synthesized NPs. Results. In comparison to uncoated controls, both coatings showed significant anti-adhesive properties against S. aureus, S. epidermidis, and E. coli. Reduction in adhesion to Mg-Ag-Cu coated discs was observed also for P. aeruginosa isolates, although differences vs uncoated controls did not reach statistical significance. Biofilm formation was reduced on discs coated with Mg-Ag-Cu compared to Ti-Ag-Cu and, again, coatings had a milder effect on P. aeruginosa, probably due to its exceptional capability of attachment and matrix production. These results were confirmed by the evaluation of bacterial colonization on nanoparticles-coated discs by means of confocal laser scanning microscopy. A viability of 95.8% and 89.4% of cells cultured in the presence of Ti-Ag-Cu and Mg-Ag-Cu discs, respectively, when compared to negative controls was observed, thus excluding cytotoxic effects on eukaryotic cells. Conclusions. The newly synthesized Ti-Ag-Cu and Mg-Ag-Cu coatings are able to limit bacterial adhesion colonization and biofilm production, thus highlighting the safe use of multi-element families of NPs as new strategies against bacterial attachment to the surface of biomedical implants. However, further studies addressing activity against P. aeruginosa and including a wide number of isolates are warranted


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_8 | Pages 44 - 44
1 Aug 2020
Li Z Geng Z Cui Z Wu S Zhu S Liang Y Yang X
Full Access

Surgical failure, mainly caused by loosening implants, causes great mental and physical trauma to patients. Improving the physicochemical properties of implants to achieve favourable osseointegration will continue to be the focus of future research. Strontium (Sr), a trace element, is often incorporated into hydroxyapatite (HA) to improve its osteogenic activity. Our previous studies have shown that miR-21 can promote the osteogenic differentiation of mesenchymal stem cells by the PI3K/β-catenin pathway. The aim of this study is to fabricate a SrHA and miR-21 composite coating and it is expected to have a favorable bone healing capability. Ti discs (20 mm diameter and one mm thickness for the in vitro section) and rods (four mm diameter and seven mm length for the in vivo section) were prepared by machining pure Ti. The Ti cylinders were placed in a Teflon-lined stainless-steel autoclave for treating at 150°C for 24 h to form SrHA layer. The miR-21 was encapsulated in nanocapsules. The miR-21 nanocapsules were mixed with CMCS powder to form a gel-like sample and uniformly coated on the SrHA modifed Ti. Osteoblast-like MG63 cells were cultured on SrHA and miR-21 modified Ti, Cell proliferation activity and osteogenesis-related gene expression were evaluated. A bone defect model was established with mature New Zealand to evaluate the osseointegration. Cylindrical holes (four mm in diameter) were created at the distal femur and tibial plateau. Each rabbit was implanted with four of the aforementioned rods (distal femur and tibial plateau of the hind legs). After implantation for one, two and three months, the rabbits were observed by X-ray and scanned using u-CT. Histological and Immunohistochemical analysis were performed to examine the osteogenic markers. A biomechanical push-in test was used to assess the bone-implant bonding strength. Both SrHA nanoparticles with good superhydrophilicity and miR-21 nanocapsules with uniform sizes were distributed evenly on the surface of the Ti. In vitro experiments revealed that the composite coating was beneficial to osteoblast proliferation, differentiation and mineralization. In vivo evaluations demonstrated that this coating could not only promote the expression of angiogenic factor CD31 but also enhance the expression of osteoblastic genes to facilitate angio-osteogenesis. In addition, the composite coating also showed a decreased RANKL expression compared with the miR-21 coating. As a result, the SrHA/miR-21 composite coating promoted new bone formation and mineralization and thus enhanced osseointegration and bone-implant bonding strength. A homogeneous SrHA and miR-21 composite coating was fabricated by generating pure Ti through a hydrothermal process, followed by adhering miR-21 nanocapsules. This coating combined the favorable physicochemical properties of SrHA and miR-21 that synergistically promoted angiogenesis, osteogenesis, osseointegration, bone mineralization and thus bone-implant bonding strength. This study provided a new strategy for surface modification of biomedical implants


Orthopaedic Proceedings
Vol. 98-B, Issue SUPP_8 | Pages 133 - 133
1 May 2016
Lal S Allinson L Hall R Tipper J
Full Access

Introduction. Silicon nitride (SiN) is a recently introduced bearing material for THR that has shown potential in its bulk form and as a coating material on cobalt-chromium (CoCr) substrates. Previous studies have shown that SiN has low friction characteristics, low wear rates and high mechanical strength. Moreover, it has been shown to have osseointegration properties. However, there is limited evidence to support its biocompatibility as an implant material. The aim of this study was to investigate the responses of peripheral blood mononuclear cells (PBMNCs) isolated from healthy human volunteers and U937 human histiocytes (U937s) to SiN nanoparticles and CoCr wear particles. Methods. SiN nanopowder (<50nm, Sigma UK) and CoCr wear particles (nanoscale, generated in a multidirectional pin-on-plate reciprocator) were heat-treated for 4 h at 180°C and dispersed by sonication for 10 min prior to their use in cell culture experiments. Whole peripheral blood was collected from healthy donors (ethics approval BIOSCI 10–108, University of Leeds). The PBMNCs were isolated using Lymphoprep® as a density gradient medium and incubated for 24 h in 5% (v/v) CO2at 37°C to allow attachment of mononuclear phagocytes. SiN and CoCr particles were then added to the phagocytes at a volume concentration of 50 µm3 particles per cell and cultured for 24 h in RPMI-1640 culture medium in 5% (v/v) CO2 at 37°C. Cells alone were used as a negative control and lipopolysaccharide (LPS; 200ng/ml) was used as a positive control. Cell viability was measured after 24 h by ATPLite assay and tumour necrosis factor alpha (TNF-α) release was measured by sandwich ELISA. U937s were co-cultured with SiN and CoCr particles at doses of 0.05, 0.5, 5 and 50 µm3 particles per cell for 24h in 5% (v/v) CO2 at 37 C. Cells alone were used as a negative control and camptothecin (2 µg/ml) was used as a positive control. Cell viability was measured after 0, 1, 3, 6 and 9 days. Results from cell viability assays and TNF-α response were expressed as mean ±95% confidence limits and the data was analysed using one-way ANOVA and Tukey-Kramer post-hoc analysis. Results and Discussion. At a high volume concentration of particles (50µm3 per cell), SiN did not affect the viability of PBMNCs, while CoCr significantly reduced the viability over a 24 h period [Figure 1A]. Similarly, SiN particles had no effect on the viability of U937s up to 9 days with a range of particle doses (0.05–50 µm3 per cell) [Figure 2A]. In contrast, CoCr particles significantly reduced the viability of U937s after 6 days [Figure 2B]. Additionally, CoCr particles caused significantly elevated levels of pro-inflammatory cytokine TNF-α, whereas no inflammation was associated with SiN particles [Figure 1B]. Conclusion. This study has demonstrated the in-vitro biocompatibility of SiN nanoparticles. Therefore, SiN is a promising orthopaedic bearing material not only due to its suitable mechanical and tribological properties, but also due to its biocompatibility. Acknowledgements. The research leading to these results has received funding from the European Union's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. GA-310477 LifeLongJoints


Introduction. Significant reduction in the wear of current orthopaedic bearing materials has made it challenging to isolate wear debris from simulator lubricants. Ceramics such as silicon nitride (SiN), as well as ceramic-like surface coatings on metal substrates have been explored as potential alternatives to conventional implant materials. Current isolation methods were designed for isolating conventional metal, UHMWPE and ceramic wear debris. The objective of this study was to develop methodology for isolation and characterisation of modern ceramic or ceramic-like coating particles and metal wear particles from serum lubricants under ultra-low wearing conditions. Sodium polytungstate (SPT) was used as a novel density gradient medium due to its properties, such as high water solubility, the fact that it is non-toxic and acts as a protein denaturant, coupled with a large density range of 1.1–3.0 g/cm3 in water. Methods. SiN nanoparticles (<50nm nanopowder, Sigma-Aldrich) and clinically relevant cobalt-chromium wear debris were added to 25% (v/v) bovine serum lubricant at concentrations of 0.03 and 0.3 mm3/ million cycles. The particles were isolated by a newly developed method using SPT gradients. The sample volume was reduced by centrifuging the lubricant at 160,000 g for 3 h at 20°C. Then, re-suspended pellet was digested twice with 0.5 mg/ml proteinse K for 18 hours at 50°C in the presence of 0.5% (w/v) SDS. Particles were then isolated from partially hydrolysed proteins by density gradient ultracentrifugation at 270,000 g for 4 h using SPT gradients [Figure 1]. At the end of centrifugation, particles were pelleted at the bottom of the centrifuge tube, leaving protein fragments and other impurities suspended higher up the tube. Isolated particles were then washed with pyrogen free water, dispersed by sonication and filtered through 15 nm polycarbonate membrane filters for SEM and EDX analysis. Results and Discussion. The morphology and size distribution of the SiN and cobalt-chromium particles was not altered after isolation [Figure 2] [Figure 3]. The mode size of the SiN particles was 30–40 nm, while the mode size of cobalt-chromium particles was 10–20 nm [Figure 3]. Unlike current isolation methods, the present study developed a highly sensitive method which uses cost effective commercially available reagents and components. Furthermore, the particles are recovered in solution and can be readily analysed using commercial size analysers, prior to use in cell studies. This study also confirmed the aggregation characteristics of silicon nitride particles in aqueous medium as observed in previous studies. The above method may also be used to isolate wear debris of materials that have density higher than 1.6 g/cm3. This includes the majority of ceramics, metals and ceramic-like coatings used in TJR components such as alumina, zirconia toughened alumina, titanium, chromium nitride coating, titanium nitride coating and chromium carbon nitride coating. Conclusions. The new isolation method successfully isolated silicon nitride nanoparticles and cobalt-chromium wear debris from serum lubricant at ultra-low concentrations of 0.03 mm3/million cycles. Acknowledgements. The research leading to these results has received funding from the European Union's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. GA-310477 LifeLongJoints


Orthopaedic Proceedings
Vol. 95-B, Issue SUPP_34 | Pages 165 - 165
1 Dec 2013
Russo A Panseri S Shelyakova T Sandri M Ortolani A Meikle S Lacey J Tampieri A Dediu V Santin M Marcacci M
Full Access

Introduction. Diaphyseal bone defect represents a significant problem for orthopaedic surgeons and patients. Bone is a complex tissue whose structure and function depend strictly on ultrastructural organization of its components: cells, organic (extracellular matrix, ECM) and inorganic components. The purpose of this study was to evaluate bone regeneration in a critical diaphyseal defect treated by implantation of a magnetic scaffold fixed by hybrid system (magnetic and mechanical), supplied through nanoparticle-magnetic (MNP) functionalized with Vascular Endothelial-Growth-Factor-(VEGF) and magnetic-guiding. Methods. A critical long bone defect was created in 8 sheep metatarsus diaphysis: it was 20.0 mm in length; the medullary canal was reamed till 8.00 mm of inner diameter. Then a 8.00 mm diameter magnetic rod was fitted into proximal medullary canal (10 mm in length). After that a scaffold made of Hydroxyapatite (outer diameter 17.00 mm) that incorporates magnetite (HA/Mgn 90/10) was implanted to fill critical long bone defect. A magnetic rod (6.00 mm diameter) was firmly incorporated at proximal side into the scaffold. Both magnets had 10 mm length. To give stability to the complex bone-scaffold-bone a plate was used as a bridge; it was fixed proximally by 2 screws and distally by 3 screws. Scaffolds biocompatibility was previously assessed in vitro using human osteoblast-like cells. Magnetic forces through scaffold were calculated by finite element software (COMSOL Multiphysics, AC/DC Model). One week after surgery, magnetic nanoparticles functionalized with VEGF were injected at the mid portion of the scaffold using a cutaneous marker positioned during surgery as reference point in 4 sheep; other sheep were used as control group. After sixteen weeks, sheep were sacrificed to analyze metatarsi. Macroscopical, radiological and microCT examinations were performed. Results. Samples obtained didn't show any inflammatory tissue around the scaffold and revealed bone tissue formation inside pores of the scaffolds and we could see also complete coverage of the scaffolds. Formation of new bone tissue was more evident at magnetized bone-scaffold interface. X-rays showed a good integration of the scaffold with a good healing process of critical bone defect: new cortical bone formation seemed to be present, recreating continuity of metatarsus diaphysis. No signs of scaffold mobilization was showed (Fig. 1). All these datas were confirmed by the microCT: new bone formation inside the scaffolds was evident, in particular at proximal bone-scaffold interface, where permanent magnet were present (Fig. 2). These preliminary results lead our research to exploiting magnetic forces to stimulate bone formation, as attested in both in vitro and in vivo models and to improve fixation at bone scaffold interface, as calculated by finite element software, and moreover to guide targeted drug delivery without functionalized magnetic nanoparticles dissemination in all body


Orthopaedic Proceedings
Vol. 97-B, Issue SUPP_15 | Pages 87 - 87
1 Dec 2015
Ballay R Landor I Suchý T Supová M Klapková E Horný L Rýglová S Zaloudková M Braun M Sucharda Z
Full Access

The aim of our project is to develop resorbable nanostructured composite layer with controlled elution of antibiotics for implants survival rate enhancement. The nanostructured layers are expected to be used especially in the case of known systemic or local (joint) inflammation. This layer can provide a bone tissue/implant (titanium alloy) bioactive interface improving the physiological healing process and eliminating the risk of bacterial orthopedic infections. The main aim of this study was to verify whether the local concentration of released vancomycin exceeded the minimum inhibitory concentration (MIC) for vancomycin-resistant Staphylococcus aureus (VRSA, >16 mg/l). The layer is composed of collagen (type I, isolated form calf skin), hydroxyapatite nanoparticles and vancomycin hydrochloride (10 wt%). The stability of collagen was enhanced by EDC/NHS cross-linking. The in vitro release of vancomycin and crystalline degradation products from optimally cross-linked layers was investigated. An elution method and a high performance liquid chromatographic assay were employed to characterize the in vitro release rates of the vancomycin and its crystalline degradation antibacterial inactive products over a 21-day period. During the whole experimental period, the level of released vancomycin was high above the MIC for VRSA. The maximum average concentration was obtained between day 4 and day 8 and it reached 265 mg/l. At the end of the experiment (day 21), an average concentration of 104 mg/l was detected. Our study confirmed the prophylactic effects of studied vancomycin-loaded nanostructured layers


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XLI | Pages 46 - 46
1 Sep 2012
To K
Full Access

Our previous study has revealed that silver nanoparticles (AgNPs) have potential to promote wound healing by accelerated re-epithelization and enhanced differentiation of fibroblasts. However, the effect of AgNPs on the functionality of repaired skin is unknown. The aim of this study was to explore the tensile properties of healed skin after treatment with AgNPs. Immunohistochemical staining, quantitative assay and scanning electron microscopy (SEM) were used to detect and compare collagen deposition, and the morphology and distribution of collagen fibers. Our results showed that AgNPs improved tensile properties and led to better fibril alignments in repaired skin, with a close resemblance to normal skin. Based on our findings, we concluded that AgNPs were predominantly responsible for regulating deposition of collagen and their use resulted in excellent alignment in the wound healing process. The exact signaling pathway by which AgNPs affect collagen regeneration is yet to be investigated


Orthopaedic Proceedings
Vol. 95-B, Issue SUPP_9 | Pages 23 - 23
1 Feb 2013
Parry M Bhabra G Sood A Figgitt M Case P Blom A
Full Access

Orthopaedic cobalt chromium particles and ions can induce indirect DNA damage and chromosome aberrations in human cells on the other side of a cellular barrier in tissue culture. This occurs by intercellular signalling across the barrier. We now show that the threshold for this effect depends on the metal form and the particle composition. Ionic cobalt and chromium induced single strand breaks at concentrations equivalent to those found in the blood of patients with well functioning metal on metal hip prostheses. However, they only caused double strand breaks if the chromium was present as chromium (VI), and did not induce chromosome aberrations. Nanoparticles of cobalt chromium alloy caused DNA double strand breaks and chromosome aberrations, of which the majority were tetraploidy. Ceramic nanoparticles induced only single strand breaks and/or alkaline labile sites when indirectly exposed to human fibroblasts. The assessment of reproductive risk from maternal exposure to biomaterials, especially those liberated by orthopaedic implants, is not yet possible with epidemiology. Whilst the barrier model used here differs from the in vivo situation in several respects, it may be useful as a framework to evaluate biomaterial induced damage across physiological barriers


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXI | Pages 27 - 27
1 May 2012
M. P G. B A. S L. C M. S A. B P. C
Full Access

Orthopaedic cobalt chromium particles and ions can induce indirect DNA damage and chromosome aberrations in human cells on the other side of a cellular barrier in tissue culture. This occurs by intercellular signalling across the barrier. We now show that the threshold for this effect depends on the metal form and the particle composition. Ionic cobalt and chromium induced single strand breaks at concentrations equivalent to those found in the blood of patients with well functioning metal on metal hip prostheses. However, they only caused double strand breaks if the chromium was present as chromium (VI), and did not induce chromosome aberrations. Nanoparticles of cobalt chromium alloy caused DNA double strand breaks and chromosome aberrations, of which the majority were tetraploidy. Ceramic nanoparticles induced only single strand breaks and/or alkaline labile sites when indirectly exposed to human fibroblasts. The assessment of reproductive risk from maternal exposure to biomaterials, especially those liberated by orthopaedic implants, is not yet possible with epidemiology. Whilst the barrier model used here differs from the in vivo situation in several respects, it may be useful as a framework to evaluate biomaterial induced damage across physiological barriers


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXIII | Pages 1 - 1
1 May 2012
Einoder B
Full Access

In 1823 J. White excised the head. In 1887 a German surgeon replaced the head with ivory. Interposition arthroplasties were common after WW1. Short-stemmed head replacing prosthesis were developed after WW2. Moores and Thompson designed a more stable intramedullary stem. Acetabular erosion was troublesome—and so replacing both surfaces started in the late 1950s using Teflon cup and metal femur. Unfortunately, these quickly became loose due to wear or sepsis. In 1960, Charnley used a polyethylene cup and stainless-steel femur and fixed both with dental cement. This ‘low friction arthroplast’ became a routine procedure after 1961. In the 1970s there were many ‘Charnley look-alike’ prosthesis with similar problems of poly-wear, granulomas and cysts causing bone loss, loosening, breakages and infection. Resurfacing with two thin shells was developed to reduce the foreign material, the bone resection and the cement used. Unfortunately, neck fractures, avascular necrosis and excessive wear of the poly shell were common. Despite operating theatres with laminar flow of sterile air, space suits and improved cementing techniques, the same problems occurred. To avoid poly and cement, Mittelmayer developed a ceramic screw cup, which did not require cement. Although some screws migrated, they did not wear. Because the un-cemented metal stem remained fixed solid to the femur, un-cemented metal cups and stems were developed. To avoid the poly-wear, ceramic liners became popular. To provide the active patients with a stable joint that requires no restriction in physical activity, a large head in a large cup is desirable. Unfortunately, the large metal-on-metal resurfacing prosthesis produce metal wear ions and nanoparticles which can form hypersensitivities, cysts and pseudotumours. Computer assisted navigation to ensure correct positioning of the prosthetic components is obviously useful for surgeons that use incisions too small to see enough to be certain of the cups position. Presently, articular cartilage research is progressing rapidly and by 2020 most arthritic hip joints will be arthroscopically debrided and resurfaced by an injection of genetically engineered articular cartilage stem cells


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXV | Pages 183 - 183
1 Jun 2012
Patella S Kon E Filardo G Martino AD D'Orazio L Matteo BD Zaffagnini S Marcacci M
Full Access

INTRODUCTION. Osteochondral defects are still a challenge for the orthopaedic surgeon, since most of the current surgical techniques lead to fibrocartilage formation and poor subchondral regeneration, often associated to joint stiffness and/or pain. Thinking of the ideal osteochondral graft from both the surgical an commercial point of view, it should be an off-the-shelf product; this is the research direction and the explanation for the new biomaterials recently proposed to repair osteochondral defect inducing an “in situ” cartilage regeneration starting from the time of the implantation into the defect site. For the clinical pilot study we performed, a newly developed nanostructured biomimetic scaffold was used to treat chondral and osteochondral lesions of the knee; its safety and manageability, as much as the surgical procedure reproducibility and the clinical outcome, were evaluated in order to test its intrinsic potential without any cells colture aid. MATERIALS AND METHODS. A new osteochondral scaffold was obtained by enucleating equine collagen type 1 fibrils with hydroxyapatite nanoparticles in 3 different layers with 3 different gradient ratios at physiological conditions. 30 patients (9F, 21M, mean age 29,3yy) affected by either chondral or osteochondral lesions of the knee (8 medial femoral condyles, 5 lateral femoral condyles, 12 patellae, 8 femoral throcleas) underwent the scaffold implantation from January to July 2007. The sizes of the lesions were in between 2 and 6 squared cm. All patients and their clinical outcome were analyzed prospectively at 6, 12, 24 and 36 months using the Cartilage standard Evaluation Form as proposed by ICRS and an high resolution MRI. RESULTS. We observed a statistically significant scores improvement and function recovery comparing the pre-operative to the follow-up parameters evaluated. Moreover, we noticed a better improvement from 12 to 24mm follow up while the good results gained at 2yy were confirmed at 3yy follow up evaluation. The MOCART scoring scale was used to analyze the MRIs. In 80% of cases we obtained a complete filling of the cartilage defect and in some patients we even appreciated articular surface congruency. In this series we report 1 failure followed by a re-operation with different technique. CONCLUSIONS. This new minimally invasive one-step surgical approach to osteochondral defects seems to be an easy and effective procedure. The results obtained are very encouraging and this procedure show satisfactory outcomes even in big osteochondral defects


The Bone & Joint Journal
Vol. 96-B, Issue 5 | Pages 569 - 573
1 May 2014
Sullivan MP McHale KJ Parvizi J Mehta S

Nanotechnology is the study, production and controlled manipulation of materials with a grain size < 100 nm. At this level, the laws of classical mechanics fall away and those of quantum mechanics take over, resulting in unique behaviour of matter in terms of melting point, conductivity and reactivity. Additionally, and likely more significant, as grain size decreases, the ratio of surface area to volume drastically increases, allowing for greater interaction between implants and the surrounding cellular environment. This favourable increase in surface area plays an important role in mesenchymal cell differentiation and ultimately bone–implant interactions.

Basic science and translational research have revealed important potential applications for nanotechnology in orthopaedic surgery, particularly with regard to improving the interaction between implants and host bone. Nanophase materials more closely match the architecture of native trabecular bone, thereby greatly improving the osseo-integration of orthopaedic implants. Nanophase-coated prostheses can also reduce bacterial adhesion more than conventionally surfaced prostheses. Nanophase selenium has shown great promise when used for tumour reconstructions, as has nanophase silver in the management of traumatic wounds. Nanophase silver may significantly improve healing of peripheral nerve injuries, and nanophase gold has powerful anti-inflammatory effects on tendon inflammation.

Considerable advances must be made in our understanding of the potential health risks of production, implantation and wear patterns of nanophase devices before they are approved for clinical use. Their potential, however, is considerable, and is likely to benefit us all in the future.

Cite this article: Bone Joint J 2014; 96-B: 569–73.


The Bone & Joint Journal
Vol. 97-B, Issue 9 | Pages 1162 - 1169
1 Sep 2015
George DA Gant V Haddad FS

The number of arthroplasties being undertaken is expected to grow year on year, and periprosthetic joint infections will be an increasing socioeconomic burden. The challenge to prevent and eradicate these infections has resulted in the emergence of several new strategies, which are discussed in this review.

Cite this article: Bone Joint J 2015;97-B:1162–9.


Bone & Joint Research
Vol. 4, Issue 5 | Pages 84 - 92
1 May 2015
Hamamura K Nishimura A Iino T Takigawa S Sudo A Yokota H

Objectives

Salubrinal is a synthetic agent that elevates phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) and alleviates stress to the endoplasmic reticulum. Previously, we reported that in chondrocytes, Salubrinal attenuates expression and activity of matrix metalloproteinase 13 (MMP13) through downregulating nuclear factor kappa B (NFκB) signalling. We herein examine whether Salubrinal prevents the degradation of articular cartilage in a mouse model of osteoarthritis (OA).

Methods

OA was surgically induced in the left knee of female mice. Animal groups included age-matched sham control, OA placebo, and OA treated with Salubrinal or Guanabenz. Three weeks after the induction of OA, immunoblotting was performed for NFκB p65 and p-NFκB p65. At three and six weeks, the femora and tibiae were isolated and the sagittal sections were stained with Safranin O.