Advertisement for orthosearch.org.uk
Results 1 - 20 of 47
Results per page:
Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_11 | Pages 76 - 76
1 Dec 2020
Akdogan EK Baysan G Erkul G Cankurt U Havitcioglu H
Full Access

Meniscus has many important functions in the knee joint such as load bearing, shock absorption, joint stability, joint lubrication and proprioception. In the recent years, meniscus injuries have been the focus of orthopaedic surgeons and musculoskeletal tissue engineering applications because of its avascular nature. In this study, we aimed to compare the regeneration capacities of two composite scaffolds in a New Zealand Rabbit meniscal defect model. The first scaffold consists Poly-Lactic Acid (PLA) + chitosan + loofah and the second PLA + Hydroxyapatite (HAp) + loofah. In order to produce these scaffolds; 4% chitosan, 4% PLA and 4% HAp solutions were seperately prepared. The loofah pieces were saturated with these solutions and vacuum-dried for 14 days and sterilized with ethylene oxide. There were several characterizations performed such as Fourier Transform Infrared Spectroscopy (FTIR) for the investigation of chemical structure, Scanning Electron Microscopy (SEM) for morphological analysis, thermogravimetric differential thermal analysis (TGA/DTA) for thermal properties, mechanical compression and swelling ratio analysis. Moreover, in order to investigate biocompatibility of the scaffolds, WST-1 colorimetric assay at days 3, 7, 10, 14 and 21 was conducted. After these biocompatibility analysis, a 1.5-mm cylindrical defect was created in the avascular portion of the anterior horn of the medial meniscus in 14 New Zealand rabbits (2.5–3 kg weight) which were randomly grouped in two. The scaffolds were implanted at the defect site with the help of a freshly prepared fibrin glue. 8 weeks after the operation, the rabbits were sacrificed and their tissues were kept for further mechanical, radiological and histological analysis. In conclusion, we succeeded to produce a new meniscus scaffold. The proliferation ability of PLA + chitosan + loofah scaffold is higher than PLA + HAp + loofah scaffold. However, there was no statistically significant difference among them


The Journal of Bone & Joint Surgery British Volume
Vol. 88-B, Issue 2 | Pages 270 - 275
1 Feb 2006
Orhan Z Cevher E Mülazimoglu L Gürcan D Alper M Araman A Özsoy Y

Ciprofloxacin hydrochloride-loaded microspheres were prepared by a spray-drying method using pectin and chitosan. The effects of different polymers and drug ratios were investigated. The most appropriate carriers were selected by in vitro testing. A rat methicillin-resistant Staphylococcus aureus osteomyelitis model was used to evaluate the effects of the loaded microspheres. The drug was released rapidly from the pectin carrier but this was more sustained in the chitosan formulation. Chitosan microspheres loaded with ciprofloxacin hydrochloride were more effective for the treatment of osteomyelitis than equivalent intramuscular antibiotics


Orthopaedic Proceedings
Vol. 88-B, Issue SUPP_II | Pages 237 - 237
1 May 2006
Sprowson AP McCaskie AW Birch MA
Full Access

Background A biomaterial serves to support, organize and directly influence the behaviour of growing cells. Chitosan has the capability to be a very useful biomaterial in the speciality of orthopaedics, due to its excellent biocompatibility, and physical properties that allow topographical modification. Chitosan films have potential to be used to coat implant surfaces, regulating bone cells at the implant interface. Enhanced integration may therefore help towards solving problems such as aseptic loosening. Method 85% deacylated chitosan (Sigma) was dissolved in 2% acetic overnight. The viscous chitosan was then sterilized by autoclaving for 10 minutes. PDMS patterned stamps produced from a silicon mould were added to the viscous chitosan and as the chitosan film forms the topographic impression is left on the surface. The gel was then dried for 36 hours in a sterile system. The pH is neutralized with NaOH1M for 24 hours. The gel was washed in sterile hanks balanced salt solution until the pH was 7.4. Osteoblasts were then grown on these surfaces in a cell culture system and analysed by light microscopy and image analysis. Results We have successfully designed a protocol for the production of sterile topographically modified chitosan, with surface features that can be produced in the range of 1–100um. We have shown that cells on un-modified chitosan differentiate and form bone at a much slower rate than on chitosan with a modified surface. Findings supported by in-situ alkaline phosphatase levels. Control can be exerted on cell shape and inter-cellular interactions based upon shape and surface area between shapes; with a smaller surface area making adhesion more difficult. Conclusion. Our data shows that osteoblasts can be controlled by altering chitosans surface topography. Being able to influence biology by changing biomaterial surface features will enhance interaction at the bone implant interface, allowing greater implant integration


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_1 | Pages 77 - 77
1 Jan 2017
Crosio A Ronchi G Fornasari B Geuna S Gambarotta G Tos P
Full Access

After big loos of substances of peripheral nerves, in order to connect proximal with distal stump, it is possible to use, in alternative to autologous grafting, different kind of conduits. The chitosan conduit and the muscle in vein technique showed very good results in pre clinical and clinical settings. We compared in this study the efficacy of empty chitosan conduit versus chitosan conduit enriched with fresh muscle fibbers (MIT) to improve peripheral nerve regeneration. The median nerve of rat was repaired by means of empty chitosan conduit or MIT (nerve gam 6mm, conduit length 10 mm). As control group we used auto grafting technique. We performed analysis at short term (7,14,28 days) and at long term (12 weeks) in order to register bimolecular modification (quantitative real time PCRand western blot), morphological modification (optic and electronic microscope) and functional changing (grasping test). Bimolecular analysis showed that muscle fibbers produced and released Neuregulin1, needed for regeneration and activity of Schwann cells. Otherwise also the autograft product Neuregulin1, instead no production was observed in empty conduit. So muscle fibbers compensate this fact. Morphological analysis showed that the first myelinc fibbers appear in MIT after 14 days, but not in empty tube. The results of our work are very interesting because can merge the easiness of the implantation of chitosan tube and the efficacy of fresh muscle fibbers, as previously demonstrated by muscle in vein technique. From a clinical point of view this procedure could be an alternative to auto grafting that is nowadays the gold standard for nerve repair, but present soma disadvantages


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 57 - 57
1 Jul 2020
Chevrier A Hurtig M Lacasse F Lavertu M Potter H Pownder S Rodeo S Buschmann M
Full Access

Surgical reattachment of torn rotator cuff tendons can lead to satisfactory clinical outcome but failures remain common. Ortho-R product is a freeze-dried formulation of chitosan (CS) that is solubilized in platelet-rich plasma (PRP) to form injectable implants. The purpose of the current pilot study was to determine Ortho-R implant acute residency, test safety of different implant doses, and assess efficacy over standard of care in a sheep model. The infraspinatus tendon (ISP) was detached and immediately repaired in 22 skeletally mature ewes. Repair was done with four suture anchors in a suture bridge configuration (n = 6 controls). Freeze-dried formulations containing 1% w/v chitosan (number average molar mass 35 kDa and degree of deacetylation 83%) with 1% w/v trehalose (as lyoprotectant) and 42.2 mM calcium chloride (as clot activator) were solubilized with autologous leukocyte-rich PRP and injected at the tendon-bone interface and on top of the repaired site (n = 6 with a 1 mL dose and n = 6 with a 2 mL dose). Acute implant residency was assessed histologically at 1 day (n = 2 with a 1 mL dose and n = 2 with a 2 mL dose). Outcome measures included MRI assessment at baseline, 6 weeks and 12 weeks, histopathology at 12 weeks and clinical pathology. MRI images and histological slides were scored by 2 blinded readers (veterinarian and human radiologist, and veterinarian pathologist) and averaged. The Generalized Linear Model task (SAS Enterprise Guide 7.1 and SAS 9.4) was used to compare the different groups with post-hoc analysis to test for pairwise differences. Ortho-R implants were detected near the enthesis, near the top of the anchors holes and at the surface of ISP tendon and muscle at 1 day. Numerous polymorphonuclear cells were recruited to the implant in the case of ISP tendon and muscle. On MRI, all repair sites were hyperintense compared to normal tendon at 6 weeks and only 1 out 18 repair sites was isointense at 12 weeks. The tendon repair site gap seen on MRI, which is the length of the hyperintense region between the greater tuberosity and tendon with normal signal intensity, was decreased by treatment with the 2 mL dose when compared to control at 12 weeks (p = 0.01). Histologically, none of the repair sites were structurally normal. A trend of improved structural organization of the tendon (p = 0.06) and improved structural appearance of the enthesis (p = 0.1) with 2 mL dose treatment compared to control was seen at 12 weeks. There was no treatment-specific effect on all standard safety outcome measures, which suggests high safety. Ortho-R implants (2 mL dose) modulated the rotator cuff healing processes in this large animal model. The promising MRI and histological findings may translate into improved mechanical performance, which will be assessed in a future study with a larger number of animals. This study provides preliminary evidence on the safety and efficacy of Ortho-R implants in a large animal model that could potentially be translated to a clinical setting


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_14 | Pages 129 - 129
1 Nov 2018
Sá MJC Segundo FAS Freitas VML Azevedo AS Silva ACF de Lima GG Devine DM
Full Access

The aim of this study was to evaluate the trochlear bone and cartilaginous regeneration of rabbits using a composite based on platelet rich plasma (PRP), chitosan and hydroxyapatite. The study was approved by the ethics committee of the Federal University of Campina Grande under number 72/2017. Surgical holes measuring four millimetres in diameter were performed in rabbit trochleae, one surgical hole in each animal remained empty and another one was filled with the composite. Clinical-orthopaedic and radiographic evaluations were carried out for 60 days, after which the animals were euthanized for histomorphometric evaluations. Clinical-evaluations exhibited lameness of two members of the treatment (T) group and one member of control (C) group. The radiographic evaluation of T group exhibited absence of subchondral bone reaction (33%); nonetheless, presence of moderate subchondral bone reaction was more frequently reported in group C with 67%. Microscopic evaluation revealed the presence of tissue neoformation, composed of dense connective tissue. Microscopic findings were similar in both groups, with a difference in the amount of neoformed tissue, which was confirmed after the morphometric analysis, revealing a significant difference in the quantity of newly formed tissue at the bone / cartilage / implant interface in the T group. The results indicate that the composite based on chitosan, hydroxyapatite and PRP enhanced bone and cartilage healing


Objectives. Previously, we reported the improved transfection efficiency of a plasmid DNA-chitosan (pDNA-CS) complex using a phosphorylatable nuclear localization signal-linked nucleic kinase substrate short peptide (pNNS) conjugated to chitosan (pNNS-CS). This study investigated the effects of pNNS-CS-mediated miR-140 and interleukin-1 receptor antagonist protein (IL-1Ra) gene transfection both in rabbit chondrocytes and a cartilage defect model. Methods. The pBudCE4.1-miR-140, pBudCE4.1-IL-1Ra, and negative control pBudCE4.1 plasmids were constructed and combined with pNNS-CS to form pDNA/pNNS-CS complexes. These complexes were transfected into chondrocytes or injected into the knee joint cavity. Results. High IL-1Ra and miR-140 expression levels were detected both in vitro and in vivo. In vitro, compared with the pBudCE4.1 group, the transgenic group presented with significantly increased chondrocyte proliferation and glycosaminoglycan (GAG) synthesis, as well as increased collagen type II alpha 1 chain (COL2A1), aggrecan (ACAN), and TIMP metallopeptidase inhibitor 1 (TIMP-1) levels. Nitric oxide (NO) synthesis was reduced, as were a disintegrin and metalloproteinase with thrombospondin type 1 motif 5 (ADAMTS-5) and matrix metalloproteinase (MMP)-13 levels. In vivo, the exogenous genes reduced the synovial fluid GAG and NO concentrations and the ADAMTS-5 and MMP-13 levels in cartilage. In contrast, COL2A1, ACAN, and TIMP-1 levels were increased, and the cartilage Mankin score was decreased in the transgenic group compared with the pBudCE4.1 group. Double gene combination produced greater efficacies than each single gene, both in vitro and in vivo. Conclusion. This study suggests that pNNS-CS is a good candidate for treating cartilage defects via gene therapy, and that IL-1Ra in combination with miR-140 produces promising biological effects on cartilage defects. Cite this article: R. Zhao, S. Wang, L. Jia, Q. Li, J. Qiao, X. Peng. Interleukin-1 receptor antagonist protein (IL-1Ra) and miR-140 overexpression via pNNS-conjugated chitosan-mediated gene transfer enhances the repair of full-thickness cartilage defects in a rabbit model. Bone Joint Res 2019;8:165–178. DOI: 10.1302/2046-3758.83.BJR-2018-0222.R1


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_16 | Pages 11 - 11
19 Aug 2024
Hamilton JL Gianotti S Impergre A AbuAlia M Markovics A Jacobs JJ Valle CJD Wimmer MA
Full Access

We created TiO. 2. nanotubes (TNTs) on the surface of titanium (Ti) implants with subsequential loading with gentamicin and chitosan, acting as a control release agent, by electrophoretic deposition (EPD). We hypothesized femoral implants with TNTs loaded with gentamicin and chitosan would localize antibiotic to the implant and surgical site and prevent PJI in a mouse model. Ti-6Al-4V ELI wires underwent TNT surface modification by two-step anodization. EPD was then used to load gentamicin and chitosan onto the Ti wire with surface TNTs. Control Ti wires contained TNTs with EPD of chitosan only. 12-week-old male C57BL/6J mice underwent received a right femoral intramedullary implant followed by inoculation at the surgical site with 1×10. 3. CFUs of bioluminescent Xen36 Staphylococcus aureus (S. aureus). Mice were randomly divided into two implant groups: 1) Gentamicin + Chitosan Group (n=7; experimental group); 2) Chitosan Group (n=7; control group). Outcomes included: 1) Relative S. aureus abundance by bioluminescence imaging; 2) Quantification of S. aureus at the implant and surrounding tissue by colony forming unit (CFU) analysis; 3) Scanning electron microscopy (SEM) for implant bacterial biofilm; 4) Radiographic signs of PJI. Over 14 days assessment following wire implantation and inoculation with S. aureus, the experimental group had no evidence of infection based on i) no increased Xen36 S. aureus bioluminescence signal and ii) no CFUs present. All control had increased bioluminescence signal, above baseline, at all time-points and presence of CFUs. Ti femoral implants modified with surface TNTs and coated with gentamicin and chitosan through EPD prevented PJI in all mice through 14 days. In comparison, all control mice demonstrated evidence of PJI over 14 days. Implants with TNTs and EPD of gentamicin were highly effective in this mouse PJI model


Bone & Joint Research
Vol. 12, Issue 12 | Pages 722 - 733
6 Dec 2023
Fu T Chen W Wang Y Chang C Lin T Wong C

Aims. Several artificial bone grafts have been developed but fail to achieve anticipated osteogenesis due to their insufficient neovascularization capacity and periosteum support. This study aimed to develop a vascularized bone-periosteum construct (VBPC) to provide better angiogenesis and osteogenesis for bone regeneration. Methods. A total of 24 male New Zealand white rabbits were divided into four groups according to the experimental materials. Allogenic adipose-derived mesenchymal stem cells (AMSCs) were cultured and seeded evenly in the collagen/chitosan sheet to form cell sheet as periosteum. Simultaneously, allogenic AMSCs were seeded onto alginate beads and were cultured to differentiate to endothelial-like cells to form vascularized bone construct (VBC). The cell sheet was wrapped onto VBC to create a vascularized bone-periosteum construct (VBPC). Four different experimental materials – acellular construct, VBC, non-vascularized bone-periosteum construct, and VBPC – were then implanted in bilateral L4-L5 intertransverse space. At 12 weeks post-surgery, the bone-forming capacities were determined by CT, biomechanical testing, histology, and immunohistochemistry staining analyses. Results. At 12 weeks, the VBPC group significantly increased new bone formation volume compared with the other groups. Biomechanical testing demonstrated higher torque strength in the VBPC group. Notably, the haematoxylin and eosin, Masson’s trichrome, and immunohistochemistry-stained histological results revealed that VBPC promoted neovascularization and new bone formation in the spine fusion areas. Conclusion. The tissue-engineered VBPC showed great capability in promoting angiogenesis and osteogenesis in vivo. It may provide a novel approach to create a superior blood supply and nutritional environment to overcome the deficits of current artificial bone graft substitutes. Cite this article: Bone Joint Res 2023;12(12):722–733


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_7 | Pages 115 - 115
4 Apr 2023
Wu H Ding Y Sun Y Liu Z Li C
Full Access

Intervertebral disc degeneration can lead to physical disability and significant pain, while the present therapeutics still fail to biochemically and biomechanically restore the tissue. Stem cell-based therapy in treating intervertebral disc (IVD) degeneration is promising while transplanting cells alone might not be adequate for effective regeneration. Recently, gene modification and 3D-printing strategies represent promising strategies to enhanced therapeutic efficacy of MSC therapy. In this regard, we hypothesized that the combination of thermosensitive chitosan hydrogel and adipose derived stem cells (ADSCs) engineered with modRNA encoding Interleukin − 4 (IL-4) can inhibit inflammation and promote the regeneration of the degenerative IVD. Rat ADSCs were acquired from adipose tissue and transfected with modRNAs. First, the kinetics and efficacy of modRNA-mediated gene transfer in mouse ADSCs were analyzed in vitro. Next, we applied an indirect co-culture system to analyze the pro-anabolic potential of IL-4 modRNA engineered ADSCs (named as IL-4-ADSCs) on nucleus pulposus cells. ModRNA transfected mouse ADSCs with high efficiency and the IL-4 modRNA-transfected ADSCs facilitated burst-like production of bio-functional IL-4 protein. In vitro, IL-4-ADSCs induced increased anabolic markers expression of nucleus pulposus cells in inflammation environment compared to untreated ADSCs. These findings collectively supported the therapeutic potential of the combination of thermosensitive chitosan hydrogel and IL-4-ADSCs for intervertebral disc degeneration management. Histological and in vivo validation are now being conducted


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 15 - 15
2 Jan 2024
Costa B Alves P Fonseca D Campos F Monteiro AC Pereira R Costa F Gomes P Martínez-de-Tejada G Monteiro C Martins M
Full Access

Orthopedic Device-Related Infections (ODRIs) are a major medical challenge, particularly due to the involvement of biofilm-encased and multidrug-resistant bacteria. Current treatments, based on antibiotic administration, have proven to be ineffective. Consequently, there is a need for antibiotic-free alternatives. Antimicrobial peptides (AMPs) are a promising solution due to their broad-spectrum of activity, high efficacy at very low concentrations, and low propensity to induce resistance. We aim to develop a new AMP-based chitosan nanogel to be injected during orthopedic device implantation to prevent ODRIs. Chitosan was functionalized with norbornenes (NorChit) through the reaction with carbic anhydride and then, a cysteine-modified AMP, Dhvar5, a peptide with potent antibacterial activity, even against methicillin-resistant Staphylococcus aureus (MRSA), was covalently conjugated to NorChit (NorChit- Dhvar5), through a thiol-norbornene photoclick chemistry (UV= 365 nm). For NorChit-Dhvar5 nanogels production, the NorChit-Dhvar5 solution (0.15% w/v) and Milli-Q water were injected separately into microfluidic system. The nanogels were characterized regarding size, concentration, and shape, using Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA) and Dynamic light scattering (DLS). The nanogels antibacterial properties were assessed in Phosphate Buffer (PBS) for 6 h, against four relevant microorganisms (Pseudomonas aeruginosa, S. aureus and MRSA, and in Muller- Hinton Broth (MHB), 50% (v/v) in PBS, supplemented with human plasma (1% (v/v)), for 6 and 24 h against MRSA. The obtained NorChit-Dhvar5 nanogels, presented a round-shaped and ∼100 nm. NorChit- Dhvar5 nanogels in a concentration of 10. 10. nanogels/mL in PBS were capable of reducing the initial inoculum of P. aeruginosa by 99%, S. aureus by 99%, and MRSA by 90%. These results were corroborated by a 99% MRSA reduction, after 24 h in medium. Furthermore, NorChit-Dhvar5 nanogels do not demonstrate signs of cytotoxicity against MC3T3-E1 cells (a pre-osteoblast cell line) after 14 days, having high potential to prevent antibiotic-resistant infection in the context of ODRIs


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_2 | Pages 72 - 72
2 Jan 2024
Agnes C Murshed M Willie B Tabrizian M
Full Access

Critical size bone defects deriving from large bone loss are an unmet clinical challenge1. To account for disadvantages with clinical treatments, researchers focus on designing biological substitutes, which mimic endogenous healing through osteogenic differentiation promotion. Some studies have however suggested that this notion fails to consider the full complexity of native bone with respect to the interplay between osteoclast and osteoblasts, thus leading to the regeneration of less functional tissue2. The objective of this research is to assess the ability of our laboratory's previously developed 6-Bromoindirubin-3’-Oxime (BIO) incorporated guanosine diphosphate crosslinked chitosan scaffold in promoting multilineage differentiation of myoblastic C2C12 cells and monocytes into osteoblasts and osteoclasts1, 3, 4. BIO addition has been previously demonstrated to promote osteogenic differentiation in cell cultures5, but implementation of a co-culture model here is expected to encourage crosstalk thus further supporting differentiation, as well as the secretion of regulatory molecules and cytokines2. Biocompatibility testing of both cell types is performed using AlamarBlue for metabolic activity, and nucleic acid staining for distribution. Osteoblastic differentiation is assessed through quantification of ALP and osteopontin secretion, as well as osteocalcin and mineralization staining. Differentiation into osteoclasts is verified using SEM and TEM, qPCR, and TRAP staining. Cellular viability of C2C12 cells and monocytes was maintained when cultured separately in scaffolds with and without BIO for 21 days. Both scaffold variations showed a characteristic increase in ALP secretion from day 1 to 7, indicating early differentiation but BIO-incorporated sponges yielded higher values compared to controls. SEM and TEM imaging confirmed initial aggregation and fusion of monocytes on the scaffold's surface, but BIO addition appeared to result in smoother cell surfaces indicating a change in morphology. Late-stage differentiation assessment and co-culture work in the scaffold are ongoing, but initial results show promise in the material's ability to support multilineage differentiation. Acknowledgements: The authors would like to acknowledge the financial support of the Collaborative Health Research Program (CHRP) through CIHR and NSERC, as well as Canada Research Chair – Tier 1 in Regenerative Medicine and Nanomedicine, and the FRQ-S


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_15 | Pages 48 - 48
1 Nov 2018
Devine D Hayes J Kotsougiani D Evans C
Full Access

Bone has a remarkable capacity to heal. However, in some instances the amount of bone which is needed to heal exceeds its healing capacity. Due to reported issues with current treatments there is continued research into alternative approaches with a view to producing an off the shelf alternative to the gold standard autologous bone transplants. The current investigated the use of a chitosan/hydroxyapatite scaffold, which was used to covalently bone morphogenetic protein and vascular endothelial growth factor using a UV crosslinking process. Results indicate that the incorporation of hydroxyapatite increased the mechanical properties of the scaffold compared to chitosan alone. Furthermore, crosslinking was confirmed using swelling studies and FTIR analysis. Elisa indicated that physiological doses of BMP were released after 10 days while in vitro testing did not indicate a cytotoxic response to the scaffold. In vivo testing in a rat femoral defect model indicated the efficacy of the treatment with scaffolds containing BMP and VEGF in combination resulting in more bone in the defect compared to the scaffold alone 8 weeks post-surgery


Orthopaedic Proceedings
Vol. 104-B, Issue SUPP_14 | Pages 5 - 5
1 Dec 2022
Lombardo MDM Mangiavini L Peretti GM
Full Access

Menisci are crucial structures for knee homeostasis: they provide increase of congruence between the articular surfaces of the distal femur and tibial plateau, bear loading, shock absorption, lubrication, and proprioception. After a meniscal lesion, the golden rule, now, is to save as much meniscus as possible: only the meniscus tissue which is identified as unrepairable should be excised and meniscal sutures find more and more indications. Several different methods have been proposed to improve meniscal healing. They include very basic techniques, such as needling, abrasion, trephination and gluing, or more complex methods, such as synovial flaps, meniscal wrapping, or the application of fibrin clots. Basic research of meniscal substitutes has also become very active in the last decades. The features needed for a meniscal scaffold are: promotion of cell migration, it should be biomimetic and biocompatible, it should resist forces applied and transmitted by the knee, it should slowly biodegrade and should be easy to handle and implant. Several materials have been tested, that can be divided into synthetic and biological. The first have the advantage to be manufactured with the desired shapes and sizes and with precise porosity dimension and biomechanical characteristics. To date, the most common polymers are polylactic acid (PGA); poly-(L)-lactic acid (PLLA); poly- (lactic-co-glycolic acid) (PLGA); polyurethane (PU); polyester carbon and polycaprolactone (PCL). The possible complications, more common in synthetic than natural polymers are poor cell adhesion and the possibility of developing a foreign body reaction or aseptic inflammation, leading to alter the joint architecture and consequently to worsen the functional outcomes. The biological materials that have been used over time are the periosteal tissue, the perichondrium, the small intestine submucosa (SIS), acellular porcine meniscal tissue, bacterial cellulose. Although these have a very high biocompatibility, some components are not suitable for tissue engineering as their conformation and mechanical properties cannot be modified. Collagen or proteoglycans are excellent candidates for meniscal engineering, as they maintain a high biocompatibility, they allow for the modification of the porosity texture and size and the adaptation to the patient meniscus shape. On the other hand, they have poor biomechanical characteristics and a more rapid degradation rate, compared to others, which could interfere with the complete replacement by the host tissue. An interesting alternative is represented by hydrogel scaffolds. Their semi-liquid nature allows for the generation of scaffolds with very precise geometries obtained from diagnostic images (i.e. MRI). Promising results have been reported with alginate and polyvinyl alcohol (PVA). Furthermore, hydrogel scaffolds can be enriched with growth factors, platelet-rich plasma (PRP) and Bone Marrow Aspirate Concentrate (BMAC). In recent years, several researchers have developed meniscal scaffolds combining different biomaterials, to optimize the mechanical and biological characteristics of each polymer. For example, biological polymers such as chitosan, collagen and gelatin allow for excellent cellular interactions, on the contrary synthetic polymers guarantee better biomechanical properties and greater reliability in the degradation time. Three-dimensional (3D) printing is a very interesting method for meniscus repair because it allows for a patient-specific customization of the scaffolds. The optimal scaffold should be characterized by many biophysical and biochemical properties as well as bioactivity to ensure an ECM-like microenvironment for cell survival and differentiation and restoration of the anatomical and mechanical properties of the native meniscus. The new technological advances in recent years, such as 3D bioprinting and mesenchymal stem cells management will probably lead to an acceleration in the design, development, and validation of new and effective meniscal substitutes


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 46 - 46
1 Jul 2020
Adoungotchodo A Lerouge S Alinejad Y Mwale F Grant M Epure L Antoniou J
Full Access

Intervertebral disc (IVD) degeneration plays a major role in low back pain which is the leading cause of disability. Current treatments in severe cases require surgical intervention often leading to adjacent segment degeneration. Injectable hydrogels have received much attention in recent years as scaffolds for seeding cells to replenish disc cellularity and restore disc properties and function. However, they generally present poor mechanical properties. In this study, we investigated several novel thermosensitive chitosan hydrogels for their ability to mimic the mechanical properties of the nucleus pulposus (NP) while being able to sustain the viability of NP cells, and retain proteoglycans. CH hydrogels were prepared by mixing the acidic chitosan solution (2% w/v) with various combinations of three gelling agents: sodium hydrogen carbonate (SHC) and/or beta-glycerophosphate (BGP) and/or phosphate buffer (PB) (either BGP0.4M, SHC0.075M-BGP0.1M, SHC0.075M-PB0.02M or SHC0.075M-PB0.04M). The gelation speed was assessed by following rheological properties within 1h at 37°C (strain 5% and 1Hz). The mechanical properties were characterized and compared with that of human NP tissues. Elastic properties of the hydrogels were studied by evaluating the secant modulus in unconfined compression. Equilibrium modulus was also measured, using an incremental stress-relaxation test 24h after gelation in unconfined compression (5% strain at 5%/s followed by 5min relaxation, five steps). Cells from bovine IVD were encapsulated in CH-based gels and maintained in culture for 14 days. Cytocompatibility was assessed by measuring cell viability, metabolism and DNA content. Glycosaminoglycan (GAG) synthesis (retained in the gel and released) was determined using DMMB assay. Finally injectability was tested using human cadaveric discs. Unconfined compression confirmed drastically enhanced mechanical properties compared to conventional CH-BGP hydrogels (secant Young modulus of 105 kPa for SHC0.075PB0.02 versus 3–6 kPa for BGP0.04). More importantly, SHC0.075PB0.02 and SHC0.075BGP0.1 hydrogels exhibited mechanical properties very similar to NP tissue. For instance, equilibrium modulus was 5.2±0.6 KPa for SHC0.075PB0.02 and 8±0.8 KPa for SHC0.075BGP0.1 compared to 6.1±1.7 KPa for human NP tissue. Rheological properties and gelation time (G′=G″ after less than 15 s at 37°C, and rapid increase of G') of these hydrogels also appear to be adapted to this application. Cell survival was greater than 80% in SHC0.075BGP0.1 and SHC0.075PB0.02 hydrogels. Cells encapsulated in the new formulations also showed significantly higher metabolic activity and DNA content after 14 days of incubation compared to cells encapsulated in BGP0.4 hydrogel. Cells encapsulated in SHC0.075BGP0.1 and SHC0.075PB0.02 produced significantly higher amounts of glycosaminoglycans (GAG) compared to cells encapsulated in SHC0.075PB0.04 and BGP0.4 hydrogels. The total amount of GAG was higher in SHC0.075BGP0.1 hydrogel compared to SHC0.075PB0.02. Interestingly, both the SHC0.075BGP0.1 and SHC0.075PB0.02 hydrogels retained similar amounts of GAG. Injectability through a 25G syringe, filling of nuclear clefts and good retention in human degenerated discs was demonstrated for SHC0.075PB0.02 hydrogel. SHC0.075BGP0.1 appears to be a particularly promising injectable scaffold for IVD repair by providing suitable structural environment for cell survival, ECM production and mechanical properties very similar to that of NP tissue


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_14 | Pages 44 - 44
1 Nov 2018
Kimna C Deger S Tamburaci S Tihminlioglu F
Full Access

There has been a significant increase in the demand of polymeric scaffolds with promising affects in bone regeneration. However, inflammation is still a problem in transplantations to overcome with local antibiotic therapy. In this study, it is aimed to develop a functional POSS nanocage reinforced chitosan scaffold (CS/POSS) coated with drug loaded chitosan composite nanospheres to provide a controlled antibianyiotic delivery at the defect site. Gentamicin and vancomycin were selected as model antibiotic drugs. Drug loaded nanospheres were fabricated with electrospray method and characterized in terms of morphology, hydrodynamic size, surface charge, FT-IR, in vitro drug release, antimicrobial activity and cytotoxicity. CS/POSS scaffolds were fabricated via lyophilisation and characterized with mechanic, swelling test, SEM and micro CT analyses. Positively charged nanospheres with uniform morphology were obtained. High drug encapsulation efficiency (80–95%) and sustained release profile up to 25 days were achieved with a cumulative release of 80–90%. In addition, the release media of the nanospheres (in 6 hours, 24 hours and 25 days of incubation period) showed a strong antimicrobial activity against S.aureus and E.coli, and did not show any cytotoxic effect to 3T3 and SaOS-2 cell lines. CS/POSS scaffolds were obtained with high porosity (89%) and 223.3±55.2μm average pore size. POSS reinforcement increased the compression modulus from 755.7 to 846.1Pa for 10 % POSS addition. In vitro studies of nanosphere coated bilayer scaffolds have showed high cell viability. Besides ALP activity results showed that POSS incorporation significantly increased the ALP activity of Saos-2 cells cultured on the scaffold. In conclusion, these composites can be considered as a potential candidate in view of its enhanced physico-chemical properties as well as biological activities for infection preventive bone tissue engineering applications


Orthopaedic Proceedings
Vol. 87-B, Issue SUPP_III | Pages 224 - 225
1 Sep 2005
Green D Leveque I Walsh D Partridge K Howard D Mann S Oreffo R
Full Access

Introduction: The clinical need for a biodegradable material with broad application is evidenced by the fact that tissue loss as a result of injury or disease provides reduced quality of life for many at significant socio-economic cost. The development of simple biodegradable materials, with broad applicability and tissue/ cell specificity has to date proved elusive. Natural biopolymers such as alginate and chitosan are structural biomaterials of increasing significance to tissue repair and regeneration due to their potential for fabrication, design and efficient, environmentally benign synthesis. We describe the development of innovative microcapsule scaffolds based on chitosan and alginate that can be tailored to a range of cell types for a variety of tissues. Methods: Semi-permeable polysaccharide microcapsules were produced by a one-step method, in which the deposition of a semi-permeable alginate/chitosan membrane around droplets of sodium alginate was coupled with in-situ precipitation of amorphous calcium phosphate as described by Leveque et al (2002). *. A variety of human cell types including mesenchymal stem cells, osteoprogenitors selected using the STRO-1 antibody by magnetically activated cell separation (MACS), osteoprogenitors transfected with adenovirus expressing Green Fluorescent Protein (GFP) and chondrocytes were mixed with sodium alginate and encapsulated within alginate/chitosan and calcium phosphate. Results: Hybrid spheres (750–10,000um) were generated encapsulating primary human osteoprogenitor cells, STRO-1 selected osteoprogenitors and AdGFP transfected osteoprogenitors. Encapsulated cells remain viable inside the polysaccharide microcapsules for 2 weeks as shown by positive alkaline phosphatase staining of encapsulated cells. Cells expressing GFP were observed within microspheres indicating the e ability to deliver cells/factors as well as the potential for gene therapy. Encapsulation and delivery of active BMP-2 was confirmed using the promyoblast cell line C2C12 known to be exquisitely sensitive to BMP-2. Nucleation of calcium phosphate occurred within the polysaccharide membrane and could be controlled by the phosphate concentration in the alginate droplets to produce hybrid microcapsules with enhanced mechanical strength. Thin walled capsules were shown to split and degrade in culture within 2–4 days releasing viable osteoprogenitor cells indicating the ability to manipulate the mechanical integrity and to programme degradation of the microspheres. Finally we have shown that aggregation of the microspheres into extended frameworks can be achieved using a designed droplet/vapour aerosol system resulting in foams of aggregated beads. Discussion and Conclusion: A variety of human skeletal cells have been encapsulated within polysaccharide/ calcium phosphate microspheres and extended frameworks with specifiable dimensions. These composite scaffolds offer stable mechanical and chemical biomimetic environments conducive to normal cell function. Natural polysaccharides are also highly amenable to complexation with a range of bioactive molecules and consequently offer tremendous potential in tissue engineering and regeneration of hard and soft tissues


Orthopaedic Proceedings
Vol. 90-B, Issue SUPP_III | Pages 501 - 501
1 Aug 2008
Bagnaninchi P Yang Y Maffulli N Wang R El Haj A
Full Access

Introduction: Tendon tissue engineering entails the generation of a highly ordered collagen matrix with several organization scales that confer the tendon its mechanical functionality. Endogenous production of proteoglycans account for the typical microscopic organization in bundles of the tendon extracellular matrix, as they prevent lateral fusion of collagen fibril by binding the shaft of the fibres and promoting tip to tip fusion. The approach developed in this study is to rely on this molecular endogenous production and to induce a supramolecular uniaxial alignment of collagen fibres bundles with the help of specially designed scaffolds under continuous fluid shear stress. Methods: Microchannel chitosan scaffolds were produced by casting 2% chitosan gel on a mould equipped with stainless steel needles array that was imaged by optical coherence tomography with a resolution at ~10microns. From OCT measurements, regularly spaced microchannels with clearly delimited boundaries are obtained inside a microporous core of chitosan. By varying the number and the diameter of needles (from 250 μm (microns)to 500 μm (microns)) different types of microstructure have been produced. Microchannels scaffolds were seeded with primary tenocytes explanted from pig tendons and cultured in static culture, as nonstimulated group, and in a perfusion bioreactor. Results: There was a general increase in the channels occupation ratio for the group stimulated by perfusion, and inversely proportional to the microchannel diameter. Tenocytes were able to proliferate and to produce collagen extracellular matrix from the inner surface of the microchannel up to the whole channel volume. Conclusion: The proposed microstructure was appropriate for tendon engineering and its channel structure is adequate for direct OCT monitoring


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_5 | Pages 64 - 64
1 Apr 2018
DesJardins J Bales C Helms S
Full Access

Introduction. The accumulation of proteins and bacteria on implant surfaces is a critical concern in the biomedical field, especially with respect to the potential of biofilm formation on implant surfaces. Material surface wettability is often used as a predictor of potential colonization of specific bacterial strains. Surface roughness has also been shown to have a strong relationship with biofilm formation, as rougher surfaces tend to have a stronger affinity to harbor bacterial colonies. The modification of implant surfaces to impart a biofilm resistant layer can come at the expense of increasing surface roughness however, and it is therefore important to determine how the variables of wettability and roughness are affected by any new surface coating technologies. In the current work, a novel CoBlast (C) process that impregnates alumina (A) at 50 μm grit (5) or 90 μm grit (9) sizes, with the possible addition of polytetrafluoroethylene (P) onto titanium surfaces, combined with a plasma coating process called BioDep, that coats the surface with chitosan (X) with the possible addition of vancomycin (V), were evaluated for wettability and surface roughness to determine their potential as biofilm resistant treatments on implants. Materials and Methods. N=65 titanium alloy samples (n=5 for 13 sample modification types as described above and in the figure legends below) were analyzed for surface roughness and wettability. Following cleaning in ethanol, roughness testing (Ra, Rq, Rt and Rz, Wyko NT-2000 optical profilometer @ 28.7× magnification, FOV of 164×215 μm) at 5 different surface locations per specimen, and contact angle analysis was performed (2 μL water drops, KRUSS EasyDrop). Statistical differences between groups was determined using ANOVA. Results and Discussion. Figure 1a summarizes the roughness results, with significant roughening being observed with between surface blanks and all surface modification techniques, especially the CoBlasted 90 μm grit treatments. As expected, wettability (shown in Figure 1b) was significantly affected by PTFE modifications and also by the introduction chitosan and vancomycin. Conclusions. As can be seen from these results, changing the coating of a material can change the surface topography and the wettability of the surface, which can be beneficial for different applications. The results from this work show that the CoBlast and BioDep processes significantly affect both wettability and roughness, and that the benefits and potential drawbacks of each must be considered when assessing their potential for biofilm resistance. PTFE-coated samples would be best used when wanting to prevent a hydrophobic substance from binding to the material, while the alumina-coated or blank samples would be best used to prevent a hydrophilic substance from binding. In the future, nonpolar liquid wettability will be assessed to better mimic in-vivo conditions and to determine surface energy to be able to make better conclusions about the relationship between surface roughness and wettability. For any figures or tables, please contact the authors directly


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_13 | Pages 13 - 13
1 Nov 2021
Dubus M Rammal H Scomazzon L Baldit A Braux J Mauprivez C Kerdjoudj H
Full Access

Introduction and Objective. Alveolar bone resorption following tooth extraction or periodontal disease compromises the bone volume required to ensure the stability of an implant. Guided bone regeneration (GBR) is one of the most attractive technique for restoring oral bone defects, where an occlusive membrane is positioned over the bone graft material, providing space maintenance required to seclude soft tissue infiltration and to promote bone regeneration. However, bone regeneration is in many cases impeded by a lack of an adequate tissue vascularization and/or by bacterial contamination. Using simultaneous spray coating of interacting species (SSCIS) process, a bone inspired coating made of calcium phosphate-chitosan-hyaluronic acid was built on one side of a nanofibrous GBR collagen membrane in order to improve its biological properties. Materials and Methods. First, the physicochemical characterizations of the resulting hybrid coating were performed by scanning electron microscopy, X-ray photoelectron, infrared spectroscopies and high-resolution transmission electron microscopy. Then human mesenchymal stem cells (MSCs) and human monocytes were cultured on those membranes. Biocompatibility and bioactivity of the hybrid coated membrane were respectively evaluated through MSCs proliferation (WST-1 and DNA quantification) and visualization; and cytokine release by MSCs and monocytes (ELISA and endothelial cells recruitment). Antibacterial properties of the hybrid coating were then tested against S. aureus and P. aeruginosa, and through MSCs/bacteria interactions. Finally, a preclinical in vivo study was conducted on rat calvaria bone defect. The newly formed bone was characterized 8 weeks post implantation through μCT reconstructions, histological characterizations (Masson's Trichrome and Von Kossa stain), immunohistochemistry analysis and second harmonic generation. Biomechanical features of newly formed bone were determined. Results. The resulting hybrid coating of about 1 μm in thickness is composed of amorphous calcium phosphate and carbonated poorly crystalline hydroxyapatite, wrapped within chitosan/hyaluronic acid polysaccharide complex. Hybrid coated membrane possesses excellent bioactivity and capability of inducing an overwhelmingly positive response of MSCs and monocytes in favor of bone regeneration. Furthermore, the antibacterial experiments showed that the hybrid coating provides contact-killing properties by disturbing the cell wall integrity of Gram-positive and Gram-negative bacteria. Its combination with MSCs, able to release antibacterial agents and mediators of the innate immune response, constitutes an excellent strategy for fighting bacteria. A preclinical in vivo study was therefore conducted in rat calvaria bone defect. μCT reconstructions showed that hybrid coated membrane favored bone regeneration, as we observed a two-fold increase in bone volume / total volume ratios vs. uncoated membrane. The histological characterizations revealed the presence of mineralized collagen (Masson's Trichrome and Von Kossa stain), and immunohistochemistry analysis highlighted a bone vascularization at 8 weeks post-implantation. However, second harmonic generation analysis showed that the newly formed collagen was not fully organized. Despite a significant increase in the elastic modulus of the newly formed bone with hybrid coated membrane (vs. uncoated membrane), the obtained values were lower than those for native bone (approximately 3 times less). Conclusions. These significant data shed light on the regenerative potential of such bioinspired hybrid coating, providing a suitable environment for bone regeneration and vascularization, as well as an ideal strategy to prevent bone implant-associated infections