Advertisement for orthosearch.org.uk
Results 1 - 20 of 129
Results per page:
Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 63 - 63
2 Jan 2024
Charbonnier B Guyon L Touya N Dutilleul M Véziers J Maitre P Gauthier O Corre P Weiss P
Full Access

Developments in the field of additive manufacturing have allowed significant improvements in the design and production of scaffolds with biologically relevant features to treat bone defects. Unfortunately, the workflow to generate personalized scaffolds is source of inaccuracies leading to a poor fit between the implant and patients' bone defects. In addition, scaffolds are often brittle and fragile, uneasing their handling by surgeons, with significant risks of fracture during their insertion in the defect. Consequently, we developed organo-mineral cementitious scaffolds displaying evolutive mechanical properties which are currently being evaluated to treat maxillofacial bone deformities in veterinary clinics. Treatment of dog patients was approved by ethic and welfare committees (CERVO-2022-14-V). To date, 8 puppies with cleft palate/lip deformities received the following treatment. Two weeks prior surgery, CT-scan of patient's skull was performed to allow for surgical planning and scaffold designing. Organo-mineral printable pastes were formulated by mixing an inorganic cement precursor (α-Ca3(PO4)2) to a self-reticulating hydrogel (silanized hyaluronic acid) supplemented with a viscosifier (hydroxymethylpropylcellulose). Scaffolds were produced by robocasting of these pastes. Surgical interventions included the reconstruction of soft tissues, and the insertion of the scaffold soaked with autologous bone marrow. Bone formation was monitored 3 and 6 months after reconstruction, and a biopsy at 6 months was performed for more detailed analyses. Scaffolds displayed great handling properties and were inserted within bone defects without significant issue with a relevant bone edges/scaffold contact. Osteointegration of the scaffolds was observed after 3 months, and regeneration of the defect at 6 months seemed quite promising. Preliminary results have demonstrated a potential of the set-up strategy to treat cleft lip/palate deformities in real, spontaneous clinical setting. Translation of these innovative scaffolds to orthopedics is planned for a near future


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_3 | Pages 2 - 2
1 Feb 2020
Shields D Llopis-Hernandez V Jayawarna V Gonzales-Garcia C Marshall W Dalby M Salmeron-Sanchez M
Full Access

Osteoinductive bone substitutes are in their developmental infancy and a paucity of effective grafts options persists despite clinical demand. Bone mineral substitutes such as hydroxyapatite cause minimal biological activity when compared to osteoinductive systems present biological growth factors in order to drive bone regeneration. We have previously demonstrated the in-vitro efficacy of a bioengineered system at presenting growth factors at ultra low-doses. This study aimed to translate this growth factor delivery system towards a clinically applicable implant. Osteoinductive surfaces were engineered using plasma polymerisation of poly(ethyl acrylate) onto base materials followed by adsorption of fibronectin protein and subsequently growth factor (BMP-2). Biological activity following ethylene oxide (EO) sterilisation was evaluated using ELISAs targeted against BMP-2, cell differentiation studies and atomic force microscopy. Scaffolds were 3D printed using polycaprolactone/hydroxyapatite composites and mechanically tested using a linear compression models to calculate stress/strain. In-vivo analysis was performed using a critical defect model in 23 mice over an 8 week period. Bone formation was assessed using microCT and histological analysis. Finally, a computer modelling process was developed to convert patient CT images into surface models, then formatted into 3D-printable scaffolds to fill critical defects. Following EO sterilisation, there was no change in scaffold surface and persistent availability of growth factors. Scaffolds showed adequate porosity for cell migration with mechanical stiffness similar to cancellous bone. Finally, the in vivo murine model demonstrated rapid bone formation with evidence of trabecular remodelling in samples presenting growth factors compared to controls


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_2 | Pages 103 - 103
2 Jan 2024
Cardona-Timoner M Bessa-Gonçalves M Nogueira F Barbosa M Santos S
Full Access

Bone defects and fractures, caused by injury, trauma or tumour resection require hospital treatment and temporary loss of mobility, representing an important burden for societies and health systems worldwide. Autografts are the gold standard for promoting new bone formation, but these may provide insufficient material and lead to donor site morbidity and pain. We previously showed that Fibrinogen (Fg) scaffolds promote bone regeneration in vivo (1), and that modifying them with 10mM of Magnesium (Mg) ions modulates macrophage response in vitro and in vivo (2). Also, we showed that Extracellular Vesicles (EV) secreted by Dendritic Cells (DC) recruit Mesenchymal Stem/Stromal Cells (MSC)(3). Herein, we aim to functionalize FgMg scaffolds with DC-EV, to promote recruitment and osteogenic differentiation of MSC. Scaffolds were produced by freeze-drying (2). Ethical permission was sought for all studies. Primary human peripheral blood monocyte-derived DC were cultured, their secreted EV were isolated by differential (ultra)-centrifugation and characterised by transmission electron microscopy and nanoparticle tracking analysis (3). Bone marrow MSC were used to determine the impact of EV-functionalized scaffolds through migration assays and their osteogenic differentiation was assessed by Alizarin Red staining. Fg and FgMg scaffolds functionalized with EV were characterized. Fg and FgMg scaffolds functionalized with DC-secreted EV were more efficient at recruiting MSC than scaffolds alone. MSC cultured on FgMg scaffolds showed significantly increased calcium deposits, in comparison with those cultured on Fg scaffolds. Fg scaffold modification by Mg promotes MSC osteogenic differentiation, while their functionalization with DC-secreted EV acts to promote MSC recruitment. This renders the FgMg-EV functionalized scaffolds an attractive material to promote new bone formation. Acknowledgments: Work funded by Orthoregeneration Network (ON Pilot Grant Spine 2021, EVS4Fusion). MCT supported by ERASMUS+ program


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_3 | Pages 85 - 85
23 Feb 2023
Flynn S Lemoine M Boland F O'Brien F O'Byrne J
Full Access

Restoration a joint's articular surface following degenerative or traumatic pathology to the osteochondral unit pose a significant challenge. Recent advances have shown the utility of collagen-based scaffolds in the regeneration of osteochondral tissue. To provide these collagen scaffolds with the appropriate superstructure novel techniques in 3D printing have been investigated. This study investigates the use of polyɛ-caprolactone (PCL) collagen scaffolds in a porcine cadaveric model to establish the stability of the biomaterial once implanted. This study was performed in a porcine cadaveric knee model. 8mm defects were created in the medial femoral trochlea and repaired with a PCL collagen scaffold. Scaffolds were secured by one of three designs; Press Fit (PF), Press Fit with Rings (PFR), Press Fit with Fibrin Glue (PFFG). Mobilisation was simulated by mounting the pig legs on a continuous passive motion (CPM) machine for either 50 or 500 cycles. Biomechanical tensile testing was performed to examine the force required to displace the scaffold. 18 legs were used (6 PF, 6 PFR, 6 PFFG). Fixation remained intact in 17 of the cohort (94%). None of the PF or PFFG scaffolds displaced after CPM cycling. Mean peak forces required to displace the scaffold were highest in the PFFG group (3.173 Newtons, Standard deviation = 1.392N). The lowest peak forces were observed in the PFR group (0.871N, SD = 0.412N), while mean peak force observed in the PF group was 2.436N (SD = 0.768). There was a significant difference between PFFG and PFR (p = 0.005). There was no statistical significance in the relationship between the other groups. PCL reinforcement of collagen scaffolds provide an innovative solution for improving stiffness of the construct, allowing easier handling for the surgeon. Increasing the stiffness of the scaffold also allows press fit solutions for reliable fixation. Press fit PCL collagen scaffolds with and without fibrin glue provide dependable stability. Tensile testing provides an objective analysis of scaffold fixation. Further investigation of PCL collagen scaffolds in a live animal model to establish quality of osteochondral tissue regeneration are required


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_15 | Pages 75 - 75
1 Nov 2018
Hammerl A Cano CD De-Juan-Pardo E van Griensven M Poh P
Full Access

Bone regeneration using a scaffold-based tissue engineering approach involves a spectrum of overlapping processes, which are driven by cell-to-cell, cell-to-extracellular matrix (ECM) and cell-to-biomaterials interactions. Traditionally, the study of osteogenesis potential of tissue-engineered constructs (TECs) in vitro only considers the osteoblasts- or mesenchymal cells (MSCs)-to-biomaterials interactions. However, this poorly recapitulates the process of bone regeneration under physiological conditions. In this study, a growth factors free co-culture model, comprising osteoblasts and monocytes was established to allow for the study of the osteogenesis potential of a TEC taking into consideration osteoblasts-to-monocytes and cells-to-biomaterials interactions. Scaffolds made of medical-grade polycaprolactone (mPCL) were fabricated by means of melt electrospinning writing technique. Subsequently, scaffolds were coated with a thin layer of calcium phosphate (CaP) by means of chemical deposition. Scaffolds with CaP coating were seeded with human-derived primary osteoblasts and monocytes and cultured for up to nine weeks. At several time-points, cells were evaluated for alkaline phosphatase and tartrate-resistant acid phosphatase activity. Additionally, cell morphology was observed through fluorescence microscopy and osteoblastic- and osteoclastic-related gene expression was analyzed by quantitative reverse transcription-polymerase chain reaction. The simultaneous presence of osteoblasts and monocytes and CaP accelerated cell matrix formation on scaffolds. Quantitative gene expression profile showed similar findings. Whereby, osteoblastic- and osteoclastic-related gene expression was highest in the PCL/CaP co-culture groups compared to other groups. This indicated synergistic effects of soluble factors secreted by cells and solubilized inorganic components from the scaffolds in promoting matrix deposition


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_15 | Pages 98 - 98
1 Nov 2018
Ryan EJ Ryan AJ Philippart A Ciraldo FE Boccaccini AR Kearney CJ O'Brien FJ
Full Access

The bone infection osteomyelitis (typically Staphylococcus aureus) requires a multistep treatment process including: surgical debridement, long-term systemic high-dose antibiotics, and often bone grafting. With antibiotic resistance becoming increasingly concerning, alternative approaches are urgently needed. Herein, we develop a one-step treatment for osteomyelitis that combines local, controlled release of non-antibiotic antibacterials (copper) within a proven regenerative scaffold. To maximise efficacy we utilised bioactive glass – an established material with immense osteogenic capacity – as a copper ion delivery reservoir. Copper ions have also been shown to stimulate angiogenesis and induce MSC differentiation down an osteogenic lineage. To eliminate grafting requirements, the copper-doped BG was incorporated into our previously developed collagen scaffolds to produce multifunctional antibacterial, osteogenic, and angiogenic scaffolds. Scaffolds were fabricated by freeze-drying a co-suspension of collagen and bioactive glass particles (+/− copper doping, referred to as CuBG and BG, respectively) at a range of different concentrations (0–300% w/w bioactive glass/collagen). Scaffolds demonstrated a 2.7-fold increase in compressive modulus (300% CuBG vs. 0%; p≤0.01), whilst maintaining >98% porosity. The 300% CuBG scaffolds showed significant antibacterial activity against Staphylococcus aureus (p≤0.001). In terms of osteogenesis, both 100% and 300% CuBG scaffolds increased cell-mediated calcium deposition on the scaffolds at day 14 and 28 (p≤0.05 and p≤0.001), as confirmed by alizarin red staining. 100% CuBG scaffolds significantly enhanced angiogenesis by increased tubule formation (p≤0.01) and VEGF protein production (p≤0.001) (all ≥n=3). In summary, this single-stage, off-the-shelf treatment for osteomyelitis shows potential to minimise bone grafting and antibiotic dependence, while reducing hospital stays and costs


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_11 | Pages 34 - 34
1 Dec 2020
Pugliese E Zeugolis D
Full Access

The enthesis is a tissue interface between tendon and bone, essential for adequate force transmission and composed by four distinct zones, namely tendon, fibrocartilage, mineralized fibrocartilage and bone. Given the avascularity of the tendon and the gradual change in tissue architecture and cell phenotype, the enthesis original tissue is often not re-established after chronic injuries, resulting in scar formation. Conservative treatments and surgical approaches are still far from a functional regeneration, whilst tissue engineering based scaffolds have recently showed great potential. In this work, we hypothesised that collagen-based scaffolds that mimic the basic architecture of the enthesis, will be able to spatially direct stem cell differentiation, providing an in vitro platform to study enthesis regeneration. A three-layer sponge composed of a tendon-like layer (collagen type I), a fibrocartilage-like layer (collagen type II) and a bone-like layer (collagen type I and hydroxyapatite) was fabricated by an iterative layering freeze-drying technique. Scaffold pore size and structural continuity at the interfaces were assessed by SEM and μ-CT analysis. Bone-marrow derived stem cells (BMSCs) were seeded on the scaffold and cultured in basal and differentiation media (chondrogenic, tenogenic and osteogenic). At day 7 and 21 the scaffolds were stained with Alizarin Red and Alcian Blue; alkaline phosphatase activity (ALP) and calcium and glycosaminoglycans (GAGs) were quantified in order to evaluate BMSC differentiation towards osteogenic and chondrogenic lineage. The presence of collagen I, III, tenascin and decorin in the scaffolds was evaluated by immunofluorescence staining in order to evaluate tenogenic differentiation of BMSCs. Scaffolds with three distinct but interconnected layers of collagen type I, collagen type II and collagen type I + hydroxyapatite were fabricated, with pore sizes in the range of 100–200 μm. Increased ALP and calcium levels were detected in a localised manner within the bone-like layer when scaffolds were cultured in basal medium (p<0.025 vs the other 2 layers). Similarly, proteoglycans were detected specifically in the fibrocartilage-like layer when scaffolds were cultured in the chondrogenic differentiation medium (p<0.03 vs the other 2 layers). Increased expression of tenogenic markers was observed in the tendon-like layer of scaffolds cultured in tenogenic media (p<0.045 vs the other 2 layers). In conclusion, the different collagen composition of each layer was able to spatially direct BMSC differentiation in a localized manner within the scaffold. Ongoing work is evaluating the synergistic effect between growth factor functionalized within the fibrocartilage and tendon-like layers for improved BMSC differentiation. Overall, these scaffolds hold promising potential in developing novel and more efficient strategy towards enthesis regeneration


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 193 - 193
1 Jul 2014
Tovar N Sobieraj M Witek L Smay J Coelho P
Full Access

Summary. A specialised 3D- printed scaffold, combined with fillers and bioactive molecules, can be designed and characterised to demonstrate the efficacy of synthetic, off-the-shelf and custom fabricated scaffolds for the repair of long bone defects. Introduction. Using specialised three-dimensional (3-D) printing technology, combined with fillers and bioactive molecules, 3-D scaffolds for bone repair of sizable defects can be manufactured with a level of design customization that other methods lack. Hydroxyapatite (HA)/Beta-Tri-Calcium Phosphate (β -TCP) scaffold components may be created that provide mechanical strength, guide osseo- conduction and integration, and remodel over time. Additionally, research suggests that bone morphogenic protein (BMP) stimulates growth and differentiation of new bone. Therefore, we hypothesise that with the addition of BMP, HA- β -TCP scaffolds will show improved regeneration of bone over critical sized bone defects in an in vivo model. Patients & Methods. Scaffolds were implanted in six New Zealand White rabbits with a 10mm radial defect for 2 and 8 weeks. The scaffolds, made from 15% HA: 85% β-TCP, were designed using ROBOCAD design software and fabricated using a 3-D printing Robocast machine. Scaffolds were sintered at 1100°C for 4 hours with a final composition of 5% HA: ∼95% β-TCP. Micro-CT, histological analysis, and nanoindentation were conducted to determine the degree of new bone formation and remodeling. Results. Reconstructed microCT images show increased bone formation, remodeling, and integration in HA/ β -TCP-BMP scaffolds compared to virgin HA/ β -TCP scaffolds. Histological analysis showed increased bone formation but decreased osteoconduction in HA/ β -TCP-BMP scaffolds. Nanoindentation showed no effect of BMP on hardness nor elastic modulus of bone formed on the scaffolds. Discussion/Conclusions. HA/ β -TCP scaffolds with/without BMP are highly biocompatible and can successfully augment and accelerate the regeneration and remodeling of bone in critically sized long bone defects in a rabbit model. However, the data in this study show both improvement and detriment with the addition of BMP. Therefore, further studies must be performed. Ideally, eventual translation of this research to humans would eliminate the need for allograft and/or autograft in large bony defects and allow for a customizable 3D scaffold material relative to patient needs


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_2 | Pages 45 - 45
1 Jan 2017
Manjubala I Ganesan P Narendrakumar U Madhan B
Full Access

Current strategy for orthopedic tissue engineering mainly focusses on the regeneration of the damaged tissue using cell-seeded three-dimensional scaffolds. Biocompatible scaffolds with controllable degradation and suitable mechanical property are required to support new tissue in-growth and regeneration . [1]. Porous composite scaffolds made from organic and inorganic materials are highly preferred, which can mimic the natural bone in their composition as well can enhance tissue repair . [2]. Scaffolds with optimum mechanical strength in both dry and wet state are more suitable for in vivo orthopedic application. Biphasic calcium phosphate (BCP), a biocompatible ceramic and carboxymethyl cellulose (CMC), a semi-natural polymer are used in the study to prepare composite scaffolds. Citric acid is used as a crosslinking agent for the polymer to improve its stability . [3]. Stability, mechanical property in dry and wet conditions and cytocompatibility of the scaffolds were investigated. Cellulose-BCP (BC25) and crosslinked cellulose-BCP (BC25CA) scaffolds are fabricated by freeze-drying method. The stability of the scaffolds was assessed in phosphate buffered saline (PBS) and compressive modulus was measured in dry and wet condition. Cytocompatibility was assessed by culturing pre-osteoblast cells at a density of 2.5×10. 4. on crosslinked scaffold and cell proliferation was measured by performing MTT assay on day 4 and 7. Crosslinked scaffold was more stable than non-crosslinked scaffold in aqueous environment as the latter disintegrated within few hours in the solution. Non-crosslinked scaffold showed higher compressive modulus of 116.3±14.8 kPa in dry condition but is reduced to 1.2±0.7 kPa in hydrated state. Though the crosslinked scaffold shows low compressive modulus of 37.67±6.7 kPa in dry state, it exhibited appreciable compressive moduli of 17.15±1.3 kPa in hydrated state. Thus, the crosslinking of the scaffolds improved the stability as well as the mechanical strength in wet condition. Cytocompatibility was assessed by culturing pre-osteoblast cells and from the MTT assay, it is shown that the cells are proliferating on the crosslinked scaffolds with time which indicates that the scaffolds are non-toxic and cytocompatible. Stability and optimum mechanical property for scaffold in aqueous environment are highly crucial for in vivo hard tissue regeneration. This study demonstrated the preparation of crosslinked scaffolds which exhibited good stability and mechanical strength in wet condition along with a porous architecture, controlled degradability and cytocompatibility, hence, crosslinked cellulose-BCP scaffold can be used for orthopedic application


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 194 - 194
1 Jul 2014
Poldervaart M Gremmels H van Deventer K Fledderus J Oner FC Verhaar M Dhert W Alblas J
Full Access

Summary Statement. Prolonged presence of VEGF (released from gelatin microspheres) led to a significant increase in scaffold vascularization when applied in vivo. Bioprinted scaffolds with regional VEGF presence retained their architecture and regional vessel formation occurred. Introduction. Tissue-engineered bone constructs need timely vascularization for optimal performance in regeneration. A potent stimulus of vascularization is vascular endothelial growth factor (VEGF), a factor with a short half-life time. Controlled release of VEGF from gelatin microparticles (GMPs) was investigated as a means to prolong VEGF presence at the preferred location within bioprinted scaffolds, and study subsequent vascularization. Methods. Release of VEGF from GMPs was measured with ELISA and bioactivity was assessed using human endothelial progenitor cells (EPC) in Transwell and real-time migration assays. Matrigel scaffolds containing EPCs and VEGF, which was released either in a fast or sustained fashion by application of GMPs, were investigated for their in vivo vasculogenic capacity. In addition, regional differences with respect to VEGF release were introduced in 3D-printed EPC-laden scaffolds. Scaffolds were implanted in subcutaneous pockets in mice for 1 week and analyzed for vessel formation. Results. Release of VEGF from GMPs was continuous for 3 weeks. VEGF bioactivity was confirmed, EPC migration in the presence of GMP-released VEGF was indistinguishable from VEGF added to the medium. Implantation in subcutaneous pockets in mice demonstrated that vessel formation was significantly higher in the VEGF sustained release group when compared to fast release or control groups. In addition, the different regions in the bioprinted scaffolds were retained and vessel formation occurred analogous with the results seen in the Matrigel plugs. Discussion/Conclusion. We conclude that GMPs are suitable to generate sustained release profiles of bioactive VEGF, and that they can be used to generate defined differentiation regions in 3D printed heterogeneous constructs. The prolonged presence of VEGF led to a significant increase in scaffold vascularization when applied in vivo


Orthopaedic Proceedings
Vol. 87-B, Issue SUPP_I | Pages 65 - 65
1 Mar 2005
Brovarone CV Vernè E Bosetti M Cannas M
Full Access

Aims: The aim of this research work was the realization of an inorganic bioactive scaffold for bone regeneration. This biomaterial should be macroporous, in order to allow the bone in-growth, and bioactive aiming to promote the bone regeneration and healing. Methods: The macroporous biomaterial was prepared by consolidation of a suspension of starch and SiO2-CaO-Na2O-MgO glass powders. Starch powders were used as both pore former and consolidation agent. Starch-glass green bodies were prepared by uniaxial pressing and, after drying, they were heated to remove the organic phase and to sinter the inorganic one. The sintered scaffolds were characterized by X-Ray diffraction, scanning electron microscopy and mercury intrusion porosimetry. The scaffolds bioactivity was evaluated soaking the samples in a simulated body fluid for periods up to 4 weeks. On the most representative samples, in vitro tests of adhesion and proliferation were performed using human primary osteoblast-like cells. Results: The obtained scaffolds showed an interconnected macroporosity of 50–100 B5m and a satisfactory degree of sintering. The sintering treatment induced the nucleation and growth of Na2Ca2(SiO2)3 crystals which is a phase that possess a very high bioactivity index. By soaking the scaffolds in SBF for period up to 1 month, an extensive precipitation of hydroxylapatite, with the typical globular morphology, occurred both inside and outside the pores. The adhesion and proliferation tests showed a remarkable spreading of the osteoblasts on the scaffold surface and thus a good biological response. Conclusions: Scaffolds with interconnected porosity were successfully obtained. The pores are highly interconnected and homogenously distributed in the samples. The chosen thermal treatment and the use of starch powders led to a final macroporous glass-ceramic structure. The obtained scaffolds showed a very high in vitro bioactivity with precipitation of HAp. Moreover, preliminary biological tests, showed a satisfactory cellular interaction with the proposed biomaterials. For the above-mentioned reasons, the starch consolidation method, the optimized processing parameters and the tailored glass composition can be used to produce scaffolds suitable for bone substitutions and tissue engineering


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 248 - 248
1 Jul 2014
Hakimi O Mouthuy P Yapp C Wali A Baboldashti NZ Carr A
Full Access

Summary Statement. The aim of this study was to compare patterns (aligned, random and grid) of electrospun polydioxanone scaffolds for tendon repair. The aligned design was optimal, directing cell shape, orientation and protein expression. Moreover, it naturally crimped, presenting tendon-like morphology. Introduction. Nanofibrous electrospun materials have been previously proposed as potential scaffolds for tendon repair, with emphasis on biomimetic design, postulated to encourage tissue regeneration. In this study, we characterised the interaction of primary tendon-derived cells with polydioxanone (PDO) scaffolds. PDO is a polymer with an excellent in vitro and in vivo biocompatibility, and is specifically compatible with tendon-derived cells. Here, we designed electrospun PDO scaffolds with different fibre orientations, namely aligned, random and grid-like patterns. To evaluate their potential as patches for tendon repair, we grew primary tendon derived cells on these scaffolds, and tested different aspects of cell behavior, including cell shape, proliferation and protein expression. Methods. Scaffolds with different orientations were produced using a single nozzle electrospinning set-up. Human tendon cells were extracted from rotator cuff tissue resected during surgical repair, with appropriate ethical approval. Cells were grown on different scaffolds for at least 14 days. Multiphoton microscopy (MPM) was used to image cells (green, calcein AM, and red, actin-phalloidin). Cell growth was monitored using AlamarBlue assay. Cell length, width and orientation were manually measured from images acquired by fluorescence microscopy. RNA was extracted by Trizol homogenisation in a GentleMACS (Miltenyi Biotec). RNA samples were reversibly transcribed to cDNA and RT QPCR were performed using a ViiA7 (Life Technologies) with QuantiTect primer assays (QIAGEN). Results are in relative expression to GAPDH. Results. MPM enabled the visualization of the scaffolds and viable cells grown for at least 14 days. Images demonstrate the distinct appearance of cells grown on highly aligned scaffold compared to random, grid-orientation and glass control. They also show the crimp-like appearance of the oriented scaffold as well as the cells on these crimped fibres. Interestingly, proliferation was not significantly effected by scaffold pattern. However, cell shape was clearly affected, and cells grown on oriented scaffolds showed higher anisotropy and elongated, narrower cell shape compared to all other groups, presenting a more tendon-like phenotype. This was further supported by a strong increase in the expression of β-actin on the aligned compared to the randomly oriented scaffold, correlating with observed changes in cell length and shape. Conclusion. The aim of this study was to compare patterns (aligned, random and grid) of electrospun polydioxanone scaffolds as templates for tendon repair. Aligned PDO scaffolds significantly affected cell length, width, and orientation. We also found that β-actin expression was significantly increased on aligned polydioxanone scaffolds. Moreover, PDO scaffolds fabricated as aligned/oriented were observed to present crimp-like morphology, similar to native tendon. Taken together, these findings suggest an aligned morphology of polydioxanone may hold the potential to improve tendon healing


Orthopaedic Proceedings
Vol. 92-B, Issue SUPP_I | Pages 201 - 201
1 Mar 2010
Zreiqat H
Full Access

Scaffolds and Biomaterials used for skeletal tissue regeneration need to be biocompatible, osteo-inductive, osteo-conductive and mechanically compatible with bone to meet the requirements for bone tissue engineering. The aim of our research is to deliver. a new generation of stable, life-long orthopedic/dental implants that offer strong bone–implant anchorage. Novel smart scaffolds to permit greater control over the location and quality of bone regeneration, allowing faster healing. Currently available modalities for treating large bone defects, are limited in their success. Developing synthetic scaffolds that promote bone growth and adequate vascularization is vital in orthopaedic and maxillofacial surgeries. The current generation of synthetic scaffolds, does not combine the required posorsity, mechanical properties and bioactivity. This presentation will highlight some of our newly developed novel highly porous and mechanically strong scaffolds that promote the migration, proliferation and differentiation of bone and endothelial cells for effective skeletal tissue integration and vascularization. Despite major advances in prosthetic technologies, implants have a finite life of 1015 years, due to their premature failure. Novel micro-engineered surfaces are required to anchor prosthetic implants to the surrounding bony skeleton. Various surface chemical modifications have been applied to prosthetic devices to enhance osseointegration. To-date none have resulted in a stable interface strong enough to support functional loading for the lifetime of the implant. Our group demonstrated that surface chemisrty modification of biomaterials with bioactive molecules have the potential to provide a surface on a prosthesis that is conducive to normal bone metabolism


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_2 | Pages 18 - 18
2 Jan 2024
Ferreira S Tallia F Heyraud A Walker S Salzlechner C Jones J Rankin S
Full Access

For chondral damage in younger patients, surgical best practice is microfracture, which involves drilling into the bone to liberate the bone marrow. This leads to a mechanically inferior fibrocartilage formed over the defect as opposed to the desired hyaline cartilage that properly withstands joint loading. While some devices have been developed to aid microfracture and enable its use in larger defects, fibrocartilage is still produced and there is no clear clinical improvement over microfracture alone in the long term. Our goal is to develop 3D printed devices, which surgeons can implant with a minimally invasive technique. The scaffolds should match the functional properties of cartilage and expose endogenous marrow cells to suitable mechanobiological stimuli in-situ, in order to promote healing of articular cartilage lesions before they progress to osteoarthritis, and rapidly restore joint health and mobility. Importantly, scaffolds should direct a physiological host reaction, instead of a foreign body reaction, associated with chronic inflammation and fibrous capsule formation, negatively influencing the regenerative outcome.

Our novel silica/polytetrahydrofuran/polycaprolactone hybrids were prepared by sol-gel synthesis and scaffolds were 3D printed by direct ink writing. 3D printed hybrid scaffolds with pore channels of ~250 µm mimic the compressive behaviour of cartilage. Our results show that these scaffolds support human bone marrow stem/stromal cell (hMSC) differentiation towards chondrogenesis in vitro under hypoxic conditions to produce markers integral to articular cartilage-like matrix evaluated by immunostaining and gene expression analysis. Macroscopic and microscopic evaluation of subcutaneously implanted scaffolds in mice showed that scaffolds caused a minimal resolving inflammatory response. Our findings show that 3D printed hybrid scaffolds have the potential to support cartilage regeneration.

Acknowledgements: Authors acknowledge funding provided by EPSRC grant EP/N025059/1.


Orthopaedic Proceedings
Vol. 88-B, Issue SUPP_II | Pages 316 - 316
1 May 2006
Woodfield T Miot S Martin I Riesle J van Blitterswijk C
Full Access

Tissue engineering techniques, combining autologous chondrocytes with biodegradable biomaterials, may offer significant advantages over current articular cartilage repair strategies. We present a series of experiments investigating the effect of 3D scaffold architecture and biomaterial composition on cartilage tissue formation in vitro and in vivo. Porous polymer (PEGT/PBT) scaffolds with low (300/55/45) or high (1000/70/30) PEG molecular weight (MW) compositions were produced using novel solid free-form fabrication (3DF) techniques, allowing precise control over pore architecture, and conventional compression moulding (CM) foam techniques. Scaffolds were seeded with expanded human nasal chondrocytes, and cultured in vitro or implanted subcutaneously in vivo in nude mice for 4 weeks and cartilage tissue formation accessed. 3DF scaffolds contained highly accessible networks of large interconnecting pores (Ø525 μm) compared to CM scaffolds, containing complex networks of small interconnecting pores (Ø182 μm). 3DF scaffold architectures enhanced cell re-differentiation (GAG/DNA) and cartilaginous matrix accumulation compared to CM scaffolds, but only if 1000/70/30 compositions were used. Collagen type-II mRNA was significantly increased in 3DF architectures irrespective of scaffold composition. These effects were likely mediated by preferential protein adsorption to 1000/70/30 materials, promoting a spherical chondrocyte-like morphology, as well as efficient nutrient/waste exchange throughout interconnecting pores within 3DF architectures. We observed synergistic effects of both composition and 3D scaffold architecture on human chondrocyte re-differentiation capacity, however, our data suggests that scaffold composition has a more significant influence than architecture alone. Such design criteria could be included in future scaffold architectures for repairing articular cartilage defects


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_8 | Pages 18 - 18
11 Apr 2023
Kühl J Gorb S Klüter T Naujokat H Seekamp A Fuchs S
Full Access

Critical-sized bone defects can result from trauma, inflammation, and tumor resection. Such bone defects, often have irregular shapes, resulting in the need for new technologies to produce suitable implants. Bioprinting is an additive manufacturing method to create complex and individualised bone constructs, which can already include vital cells.

In this study, we established an extrusion-based printing technology to produce osteoinductive scaffolds based on polycaprolactone (PCL) combined with calcium phosphate, which is known to induce osteogenic differentiation of stem cells.

The model was created in python based on the signed distance functions. The shape of the 3D model is a ring with a diameter of 20 mm and a height of 10 mm with a spongiosa-like structure. The interconnected irregular pores have a diameter of 2 mm +/− 0.2 mm standard deviation.

Extrusion-based printing was performed using the BIO X6. To produce the bioink, PCL (80 kDa) was combined with calcium phosphate nanopowder (> 150 nm particle size) under heating. After printing, 5 × 106 hMSC were seeded on the construct using a rotating incubator.

We were able to print a highly accurate ring construct with an interconnected pore structure. The PCL combined with calcium phosphate particles resulted in a precise printed construct, which corresponded to the 3D model. The bioink containing calcium phosphate nanoparticles had a higher printing accuracy compared to PCL alone. We found that hMSC cultured on the construct settled in close proximity to the calcium phosphate particles. The hMSC were vital for 22 days on the construct as demonstrated by life/dead staining.

The extrusion printing technology enables to print a mechanically stable construct with a spongiosa-like structure. The porous PCL ring could serve as an outer matrix for implants, providing the construct the stability of natural bone. To extend this technology and to improve the implant properties, a biologised inner structure will be integrated into the scaffold in the future.


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXXVIII | Pages 65 - 65
1 Sep 2012
Adesida A Sernik J Croutze R Laouar L Secretan C Jomha NM
Full Access

Purpose. Traumatic articular cartilage (AC) defects are common in young adults and frequently progresses to osteoarthritis. Matrix-Induced Autologous Chondrocyte Implantation (MACI) is a recent advancement in cartilage resurfacing techniques and is a variant of ACI, which is considered by some surgeons to be the gold standard in AC regeneration. MACI involves embedding cultured chondrocytes into a scaffold that is then surgically implanted into an AC defect. Unfortunately, chondrocytes cultured in a normoxic environment (conventional technique) tend to de-differentiate resulting in decreased collagen II and increased collagen I producing in a fibrocartilagous repair tissue that is biomechanically inferior to AC and incapable of withstanding physiologic loads over prolonged periods. The optimum conditions for maintenance of chondrocyte phenotype remain elusive. Normal oxygen tension within AC is <7%. We hypothesized that hypoxic conditions would induce gene expression and matrix production that more closely characterizes normal articular chondrocytes than that achieved under normoxic conditions when chondrocytes are cultured in a collagen scaffold. Method. Chondrocytes were isolated from Outerbridge grade 0 and 1 AC from four patients undergoing total knee arthroplasty and embedded within 216 bovine collagen I scaffolds. Scaffolds were incubated in hypoxic (3% O2) or normoxic (21% O2) conditions for 1hr, 21hr and 14 days. Gene expression was determined using Q-rt-PCR for col I/II/X, COMP, SOX9, aggrecan and B actin. Matrix production was determined using glycosaminoglycan (GAG) content relative to cell count determined by DNA quantification. Cell viability and location within the matrix was determined by Live/Dead assay and confocal microscopy. Statistical analysis was performed using a two-tailed T-test. Results. Chondrocytes cultured under hypoxic conditions showed an upregulation of all matrix related genes compared to normoxic conditions noted most markedly in col II, COMP and SOX9 expression. There were similar numbers of chondrocytes between hypoxic and normoxic groups (P=0.68) but the chondrocytes in the hypoxic group produced more GAG per cell (P= 0.052). Viable cells were seen throughout the matrix in both groups. Conclusion. Important matrix related genes (col II, COMP, SOX9) were most significantly upregulated in hypoxic conditions compared to normoxic conditions. This was supported by an increase in GAG production per cell in hypoxic conditions. The results indicate that hypoxia induces an upregulation in the production of extracellular matrix components typical of AC with only modest increases in col I (possibly related to the col I based scaffold used in this experiment). These results indicate that hypoxic conditions are important for the maintenance of chondrocyte phenotype even when the cells are cultured in a 3D environment. In conclusion, hypoxic culture conditions should be used to help maintain chondrocyte phenotype even when culturing these cells in a 3D scaffold


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. 106-B, Issue SUPP_2 | Pages 58 - 58
2 Jan 2024
Camarero-Espinosa S
Full Access

The anterior cruciate ligament (ACL) is the connective tissue located at the end of long bones providing stability to the knee joint. After tear or rupture clinical reconstruction of the tissue remains a challenge due to the particular mechanical properties required for proper functioning of the tissue. The outstanding mechanical properties of the ACL are characterized by a viscoelastic behavior responsible of the dissipation of the loads that are transmitted to the bone. These mechanical properties are the result of a very specialized graded extracellular matrix that transitions smoothly between the heterotypic cells, stiffness and composition of the ACL and the adjacent bone. Thus, mimicking the zonal biochemical composition, cellular phenotype and organization are key to reset the proper functioning of the ACL.

We have previously shown how the biochemical composition presented to cells in electrospun scaffolds results in haptokinesis, reverting contact-guidance effects.[1] Here, we demonstrate that contact guidance can also be disrupted by structural parameters in aligned wavy scaffolds. The presentation of a wavy fiber arrangement affected the cell organization and the deposition of a specific ECM characteristic of fibrocartilage. Cells cultured in wavy scaffolds grew in aggregates, deposited an abundant ECM rich in fibronectin and collagen II, and expressed higher amounts of collagen II, X and tenomodulin as compared to aligned scaffolds. In-vivo implantation in rabbits of triphasic scaffolds accounting for aligned-wavy-aligned zones showed a high cellular infiltration and the formation of an oriented ECM, as compared to traditional aligned scaffolds.[2]


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_2 | Pages 17 - 17
2 Jan 2024
Ramos-Díez S Camarero-Espinosa S
Full Access

Articular cartilage is a multi-zonal tissue that coats the epiphysis of long bones and avoids its wear during motion. An unusual friction could micro-fracture this connective membrane and progress into an osteochondral defect (OD), where the affected cartilage suffers inflammation, fibrillation, and forfeiture of its anisotropic structure.

Clinical treatment for ODs has been focused on micro-fracture techniques, where the defect area is removed and small incisions are performed in the subchondral bone, which allows the exudation of mesenchymal stem cells (hMSCs) to the abraded zone. However, hMSCs represent less than 0.01% of the total cell population and are not able to self-organise coherently, so the treatments fail in the long term. To select, support and steer hMSCs from the bone marrow into a specific differentiation stage, and recreate the cartilage anisotropic microenvironment, multilayer dual-porosity 3D-printed scaffolds were developed.

Dual-porosity scaffolds were printed using prepared inks, containing specific ratios of poly-(d,l)lactide-co-caprolactone copolymer and gelatine microspheres of different diameters, which acted as sacrificial micro-pore templates and were leached after printing. The cell adhesion capability was investigated showing an increased cell number in dual-porosity scaffolds as compared to non-porous ones. To mimic the stiffness of the three cartilage zones, several patterns were designed, printed, and checked by dynamic-mechanical analysis under compression at 37 ºC. Three patterns with specific formulations were chosen as candidates to recreate the mechanical properties of the cartilage layers. Differentiation studies in the selected scaffolds showed the formation of mature cartilage by gene expression, protein deposition and biomolecular analysis. Given the obtained results, designed scaffolds were able to guide hMSC behaviour.

In conclusion, biocompatible, multilayer and dual-porosity scaffolds with cell entrapment capability were manufactured. These anisotropic scaffolds were able to recreate the physical microenvironment of the natural cartilage, which in turn stimulated cell differentiation and the formation of mature cartilage.

Acknowledgments: This work was supported by the EMAKIKER grant.