header advert
Orthopaedic Proceedings Logo

Receive monthly Table of Contents alerts from Orthopaedic Proceedings

Comprehensive article alerts can be set up and managed through your account settings

View my account settings

Visit Orthopaedic Proceedings at:

Loading...

Loading...

Volume 100-B, Issue SUPP_15 November 2018 The European Orthopaedic Research Society (EORS) 2018 Meeting, PART 2, Galway, Ireland, September 2018.

D. Docheva

Tendon and ligament tissues are fascinating in their simplistic appearance of tissue architecture coupled with outstanding biomechanical properties. In the last decade, the mechanisms governing their development, degenerative disease progression and step-wise repair process are becoming better understood. In this talk, I will present an overview of our basic research work on these following points. (i) Tendon generation: I will discuss our finding on the role of growth and biomechanical factors influencing tendon stem/progenitor cells; (ii) Tendon degeneration: I will provide evidences how disturbed cell-cell and cell-matrix contacts are involved in loss of tissue integrity; (iii) Tendon regeneration: I will present in vivo data on the application and performance of various cell populations in tendon repair.


M. van Griensven

The structure and extracellular matrix composition of the interface are complex and allow for a gradual mechanical stress transfer between tendons and bone. In this study, biphasic silk fibroin scaffolds designed to mimic the gradient in collagen molecule alignment present at the interface. The scaffolds had two different pore alignments: anisotropic at the tendon side and isotropic at the bone side. Total porosity ranged from 50–80% and the majority of pores were <100–300 µm. Young's modulus varied from 689–1322 kPa. In addition, human adMSC were cultured on the scaffolds to evaluate the effect of pore morphology on cell proliferation and gene expression. Biphasic scaffolds supported cell attachment and influenced cytoskeleton organization depending on pore alignment. In addition, the gene expression of tendon, enthesis and cartilage markers significantly changed in each region of the scaffolds. We functionalized those scaffolds with heparin and explored their ability to deliver TGF-β2 and GDF5. TGF-β2 and pore anisotropy synergistically increased the expression of tendon/ligament markers and collagen I protein content. The combined delivery of TGF-β2 and GDF5 enhanced the expression of cartilage markers and collagen II protein content on substrates with isotropic porosity, whereas enthesis markers were enhanced in areas of mixed anisotropic/isotropic porosity.


D. Barritault

Matrix therapy is a newly coined name emphasizing the importance of the extracellular matrix in regenerative medicine. Heparan sulfates (HS) are key elements of the extracellular matrix (ECM) scaffold which store and protect most growth factors/cytokines controlling the cell migration and differentiation required for healing processes. We have engineered biodegradable nano-polymers (alpha 1–6 polyglucose carboxymethyl sulfate) mimicking (RGTA®) to replace destroyed HS in the damaged ECM scaffolding and to protect cytokines produced by healthy neighbouring cells, thereby restoring the ECM microenvironment and tissue homeostasis and, if needed, provide a homing niche for cell therapy. This matrix therapy approach has considerably improved the quality of healing in various animal models, including muscle and tendon, with reduction or absence of fibrosis resulting in a regeneration process. Over 50 000 patients have been treated in the last years for skin and corneal wounds with dedicated products based on this technology. A randomized controlled trial was performed on 22 racing French Standardbred Trotters (ST) horses to evaluate the efficacy of another polymer, OTR4131 Equitend®, to treat tendinopathies. We evaluated the effect versus placebo on acute superficial digital flexor tendonitis over 4 months by clinical and ultrasonographic measures and their racing performances followed up over the 2 years after treatment. A significant reduction on tendon cross section area was measured in treated animals, racing was 2–3 times more often than placebo, with 3.3 times fewer recurrences and pre-injury performance level was maintained. This study may pave the way for development in humans.


C. Perucca Orfei A.B. Lovati G. Lugano M. Viganò M. Bottagisio D. D'Arrigo S. Setti L. de Girolamo

Tendon-related pathologies such as tendinopathy represent a relevant clinical and socioeconomic issue. The most innovative and conservative therapeutic approaches are meant to stimulate the intrinsic healing capability of the tissue. In this study, the use of pulsed electromagnetic fields (PEMFs) was investigated in a rat model of Achilles tendinopathy as a potential therapy. Achilles tendinopathy was chemically induced in eighty-six Sprague Dawley rats by injecting collagenase Type I within the tendon fibers. Fifty-six of them were stimulated with PEMFs (8 hours/day, 1.5 ± 0.2 mT; 75 Hz), divided in different experimental groups basing on the starting-time of PEMFs exposure (after 0, 7, 15 after Collagenase injection) and its duration (7, 15 or 30 days). Thirty animals were left unstimulated (CTRL group). According to the different time points, explanted tendons were evaluated through histological and immunohistochemical analyses in term of matrix deposition, fiber re-organization, neovascularization and inflammatory reaction. The most effective PEMF stimulation was demonstrated in the 15 days of treatment. However, when PEMF were applied immediately after the collagenase injection, no significant therapeutic results were found. On the contrary, when PEMF were applied after 7 and 15 days from the collagenase injection, they promoted the deposition of extracellular matrix and tendon fiber re-organization, reducing both the inflammatory reaction and vascularization, with significant differences compared to the CTRL group (p<0.05). Therefore, these results suggest an effective activity of PEMFs stimulation that provides a satisfying restoration of the damaged tissue, although the most performing protocol of application still needs to be identified.


J. Huegel J.F. Boorman-Padgett C.A. Nuss M.C.C. Minnig J.J. Tucker J.M. Cirone T.R. Morris D.S. Choi A.F. Kuntz E.I. Waldorff N. Zhang J.T. Ryaby L.J. Soslowsky

PEMF is currently approved by the FDA for adjunctive treatment of lumbar/cervical spine fusion and for treatment of long-bone non-unions. Soft tissues are a potential new therapeutic application for PEMF due to pre-clinical studies showing a reduction of inflammatory markers following PEMF exposure. The aim was therefore to investigate the structural/functional effects of PEMFs on tendon-to-bone and tendon-to-tendon healing in a rotator-cuff (RC) and Achilles tendon (AT) repair model, respectively. RC study: Adult male rats (n=280), underwent bi-lateral supraspinatus tendon transections with immediate repair followed by cage activity until sacrifice (4, 8, and 16 weeks). Non-controls received PEMF for 1, 3, or 6 hours daily. AT study: Male rats underwent acute, complete transection and repair of the Achilles tendon (FULL, n=144) or full thickness, partial width injury (PART, n=160) followed by immobilization for 1 week. Sacrifice was at 1, 3, and 6 weeks. Outcome measures included passive joint mechanics, gait analysis, biomechanical assessments, histological analysis of the repair site and mCT (humerus) assessment (FULL only). RC study: Significant increases in modulus, stiffness, bone mineral content and improved collagen organization was observed for the PEMF groups. No differences in joint mechanics and ambulation were observed. AT study: A decrease in stiffness and limb-loading rate was observed for the PEMF groups for the FULL groups, whereas an increase in stiffness with no change in range-of-motion was seen for the PART groups. The combined studies show that PEMF can be effective for soft tissue repair but is dependent on the location of application.


S. Avnet

The stem cell fraction of a cell population is finely tuned by stimuli from the external microenvironment. Among these stimuli, a decrease of extracellular pH (pHe) may occur in a variety of physiological and pathological conditions, including hypoxia and inflammation. Also in bone, the maintenance of acid-base balance is fundamental for skeleton homeostasis. Bone cells are extremely sensitive to the effects of interstitial pH. Acidosis inhibits mineral deposition by osteoblasts and activates osteoclast-mediated bone resorption. Moreover, acidosis is associated with inflammation, and in case of bone injury, local short-term acidosis is a crucial regulator of the healing process. Evidence of the role of acidosis as an enhancer of MSC stemness and for their activation as sensors and switcher of inflammation will be discussed.


R. Ferracini

Human mesenchymal stem cells (MSCs) are multipotent stem cells with the ability to differentiate into mesoderm-type cells such as osteoblasts, chondroblast, tenocytes etc. They can be retrieved by different sources, but the number of cells obtained suggested the adipose tissue as a primary harvest site of MSCs. Cells can be harvested using the Coleman procedure, obtaining stromal vascular fraction (SVF), enriched with MSCs, after collagenase digestion. The availability of SVF storage has been envisioned for multiple treatments of the degenerated tissue. Indeed, the use of SVF has been introduced into clinical trials for tissue regeneration for orthopaedic patients. Difficulties of a selective delivery of SVF locally have been previously discussed. Thus, the use of biological scaffolds in order to better localize SVF in the tissue site has been studied. The methodological evolution for the use of SVF in the best possible biological conditions is a milestone for good clinical results.


R. Oreffo

Advances in our understanding of skeletal stem cells and their role in bone development and repair, offer the potential to open new frontiers in bone regeneration. However, the ability to harness these cells to replace or restore the function of traumatised or lost skeletal tissue as a consequence of age or disease remains a significant challenge. We have developed protocols for the isolation, expansion and translational application of skeletal cell populations with cues from developmental biology informed by in vitro and ex vivo models as well as, nanoscale architecture and biomimetic niche development informing our skeletal tissue engineering approaches. We demonstrate the importance of biomimetic cues and delivery strategies to directly modulate differentiation of human adult skeletal cells and, central to clinical application, translational studies to examine the efficacy of skeletal stem and cell populations in innovative scaffold compositions for orthopaedics. While a number of challenges remain multidisciplinary approaches that integrate developmental and engineering processes as well as cell, molecular and clinical techniques for skeletal tissue engineering offer significant promise. Harnessing such approaches across the hard tissue interface will ultimately improve the quality of life of an increasing ageing population.


G. Duffy

The clinical translation of regenerative therapies, whether in the form of mesenchymal cells, macromolecules or small molecules, is hampered by several factors: the poor retention and short biological half-life of the therapeutic agent, the adverse side effects from systemic delivery, and difficulties with the administration of multiple doses to a target site. We report the development and application of a therapeutic reservoir device that enables sustained and repeated administration of small molecules, macromolecules and cells directly to organs and tissues of interest via a polymer-based reservoir connected to a subcutaneous port. In a myocardial infarct rodent model, we show that repeated administration of cells over a four-week period using the reservoir provided functional compared to a single injection of cells and to no treatment. Recent advances of the system include a multi-port and multi-reservoir system that can be tailored to cargo and application need. The pre-clinical use of our therapeutic reservoir as a research model may enable insights into regenerative orthopaedic therapy, particularly those therapies that require multi-dose approaches.


P. Mancuso S. Raman F. Barry M.J. Murphy

Osteoarthritis (OA) is a degenerative disease with a strong inflammatory component. Intra-articular (IA) injections of mesenchymal stem cells (MSCs) modulate local inflammation, although the lack of engraftment suggests that they undergo apoptosis. The aim of this study is to investigate the fate of IA-delivered MSCs in an animal model of OA and to assess the role of apoptosis in vitro. Collagenase-induced OA (CIOA) was performed on C57BL/6 mice and 2×10∧5 GFP+ MSCs were IA-injected in the animals. 3 days later, knee joints were digested into a single-cell suspension and MSCs retrieved by cell sorting. Conditioned medium (CM) of retrieved cells was tested on murine macrophages and cytokine secretion was measured. Apoptosis of MSCs was induced in vitro with staurosporine (STS) and evaluated by Annexin V/Sytox Blue staining; activation of caspases was measured by FLICA assays. Murine lymphocytes were cocultured with apoptotic MSCs and their proliferation measured by quantification of Cell Trace Violet. 1.63% of injected cells were retrieved and proliferated in culture. Their CM significantly modulated activation of macrophages, with greater effects from OA-induced MSCs. STS induced apoptosis with activation of Caspase 3/7. Apoptotic MSCs significantly prevented the proliferation of murine lymphocytes. MSCs can be administered and retrieved from murine knees. Retrieval yield is low, consistent with previous studies. MSCs were licensed from the OA joint to produce an immunosuppressive milieu that modulated macrophages ex vivo. In vitro, apoptosis increased the immunomodulatory potential of MSCs. This suggests that apoptosis may contribute to the therapeutic effects of MSCs in OA.


S. Garnica-Galvez I. Skoufos A. Tzora N. Prassinos N. Diakakis D.I. Zeugolis

Cell-based scaffold-free tissue equivalents present a limited clinical translation as consequence of the delayed extracellular matrix (ECM) deposition due to the prolonged production time in vitro. Different combinations of media supplements such as ascorbic acid, growth factors, oxygen tension among others can modulate the cell fate or the ECM synthesis. New research lines are focusing on the use of macromolecular crowders (MMCs) as media supplement for cell sheet production due to their ability to increase ECM deposition by volume exclusion effect, pro-collagenases alosteric regulation, matrix self-assembly by confinement and diffusion limitation (most probably, modulating the interaction between the ECM, MMPs and TIMPs). Herein, different molecular weights and concentrations of a natural potential MMC (Crowder-A) have been tested in equine adipose-derived stem cell (eADSC) and human dermal fibroblast (hADF) cultures in comparison with other commonly used crowders such as carrageenan and the Ficoll™ cocktail 70 KDa and 400 KDa. The eADSCs were characterized according to the current criteria for horse MSCs. Tri-lineage and FACS analysis showed eADSC osteogenic and adipogenic potentials and the presence of the markers CD29, CD44, CD90. The screening of the aforementioned Crowder-A was performed in cultures of 15,000 cells / cm2 for the eADSCs and 25,000 cells / cm2 for the hADFs during 3, 5, and 7 days. Non-MMC conditions were used as negative controls. Collagen type I was analysed by SDS-PAGE. Other collagen types were studied by immunocytochemistry assays. Significant increase of some ECM components was observed in some concentrations and molecular weights of the Crowder-A.


D. Mason

Osteoarthritis (OA), characterised by pain, disability and joint degeneration, is common and has no cure. Prevalence of severe radiographic knee OA is 19% in over 45's and 50% in over 75's in the US and Europe. Abnormal joint loading, or injury, increase risk of OA. We have discovered that glutamatergic signalling is mechanically regulated and glutamate receptors (GluR) drive inflammation, degeneration and pain representing potential drug targets in osteoarthritic joints. Joints from OA and knee injured patients, and rodent models of arthritis, show increased synovial fluid glutamate concentrations and abundant GluR expression. Since AMPA/kainate GluRs regulate IL-6, a critical mediator of arthritic degeneration, we tested protective effects of the AMPA/KA GluR antagonist, NBQX in animal models of arthritis. In rodent antigen induced arthritis, and osteoarthritis (meniscal transection and anterior cruciate ligament rupture), NBQX reduced joint swelling, degeneration and pain, exceeding anti-degenerative effects of other drugs tested similarly. 3D osteocyte/osteoblast co-cultures and human bone samples taken from patients undergoing high tibial osteotomy joint realignment surgery, revealed underlying cellular mechanisms mediated by bone cells. Related drugs, already used in humans for epilepsy and migraine, represent a repurposing opportunity and are effective in our models of arthritis.


O. D. Kennedy

Osteoarthritis (OA) is the most common musculoskeletal disease in the EU and is characterized by cartilage degeneration, pain and restricted movement. Post-Traumatic OA (PTOA) is a specific disease subset that occurs subsequent to traumatic injury, such as ACL rupture and makes up 12% of the overall disease burden. Our current understanding PTOA is that initial injury affects multiple tissues, and many/all contribute to overall ‘joint failure.’ MRI scans show that subchondral bone marrow lesions (BMLs) are present in 80% of ACL rupture cases in the immediate aftermath of joint injury. Their presence indicates an acute consequence in subchondral bone. It has also been suggested that BMLs overlap with, or directly represent, bone microdamage. Microdamage is known to induce osteoclast-mediated remodelling in bone. Therefore, the inhibition of subchondral bone remodelling, particularly in the early phase post-injury, may be a candidate therapeutic approach for preventing PTOA. Finally, the contiguous link between subchondral bone and articular cartilage, can allow transport of small molecules across this boundary, this suggests that bone/cartilage crosstalk is likely to be a key factor in PTOA development after injury. This presentation will summarize recent advances in our understanding these phenomena in both animal and human studies.


H. Jahr

Onset and progression of osteoarthritis (OA) is affected by a plethora of factors, including joint injury, obesity, aging, and heredity. This multi-factorial etiology obstructs our understanding of driving molecular mechanisms, which likely comprise an interplay between systemic and local factors. Next to biomechanical factors and cytokines, the course of OA appears to be altered by microenvironmental oxidative stress: cumulative evidence now suggests a prominent participation of cell signalling mediated by nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a master regulator of cellular protective processes, in this process. Nrf2 activation through phosphorylation of mitogen-activated protein kinases (MAPKs) regulates Nrf2 target genes, like hemeoxygenase-1 (HO-1), superoxide dismutase 2 (SOD2), or NAD(P)H Quinone Dehydrogenase 1 (NQO1) in OA chondrocytes. Maintaining high levels of HO-1 appears to be beneficial against OA development. Experimental manipulation of putative antioxidant response element (ARE) binding sites alters the in vitro expression of key transcription factors of chondrocyte markers in promoter-reporter assays. Potentially, Nrf2 is involved in autophagy, intermediary metabolism and unfolded protein response. RNAi-mediated depletion of Nrf2 further significantly abrogated anti-inflammatory and chondroprotective effects and epigenetics link transcriptional pathways of ‘N-factors’, Nrf2 and NFATs, to micro-RNA signalling. Current findings thus reveal novel mechanisms regulating extracellular matrix synthesis by chondrocytes. A further understanding of these pathways and their regulation will lead to important novel targets to slow OA progression.


F.-S. Wang

Chondrocyte dysfunction is attributable to the development of osteoarthritis (OA). Deregulation of chondrogenic regulators and deleterious factors, e.g. proteinases, Wnt signalling components, and autophagy repressors lowers chondrogenic activities and ultimately deteriorates cartilage homeostasis. Emerging evidence is that epigenetic pathways, including non-coding microRNAs and histone remodelling switch on/off the expression of joint-deleterious factors. MicroRNAs reduces the expressions of mRNAs through binding to the 3'-untranslation regions of targets. The levels of microRNAs, e.g. miR-29a, miR-128a in serum, synovial fluid, synovium, and cartilage are correlated with the occurrence of OA. Mice overexpressing/deficient microRNAs of interest show minor responses to OA progression. Besides, acetylation and methylation statuses of histones regulate the factors detrimental to chondrocytes through altering the interactions between histones and promoters. Histone deacetylases and demethylases, e.g. HDAC4, SIRT1, and EZH2 contribute to the modification reactions of histones, which modulate cartilage matrix metabolism. An intricate nature is that reciprocal actions between microRNAs and histone deacetylase/demethylase are indispensable in chondrocyte survival and function. Administrations with specific inhibitor/agonists for microRNAs and histone deacetylases/demethylase enable joints to show minor responses to articular injury, which mitigate the pathogenesis of OA. This talk highlights the biological roles and therapeutic advantage of epigenetic microRNAs and histone remodelling in OA.


Y.S. Chen W.S. Lian J.Y. Ko F.S. Wang

Fatty marrow and bone loss are prominent pathologic features of osteoporosis. DNA hypermethylation shifts mesenchymal stem cells towards adipocytes impairing bone formation. Brown adipocytes produce growth factors advantageous to osteogenesis, whereas white adipocytes secrete pro-inflammatory cytokines deleterious to bone homeostasis. We assess DNA methylation inhibitor action to brown and white adipocyte formation in marrow fat of osteoporotic skeletons. Osteoporotic skeletons in mice were induced by glucocorticoid, ovariectomy or ageing. Marrow adipose volume and bone structure were quantified using OsO4 contrast-μCT imaging. Brown and white adipocytes were probed using immunostaining, RT-PCR and primary bone-marrow mesenchymal stem cell cultures. Abundant marrow fat and spare trabecular bone existed in osteoporotic skeletons. Osteoporosis increased expressions of general adipogenic markers PPARγ2 and FABP4 and white adipocyte markers TCF21 and HOXc9, whereas expressions of brown adipocyte markers PGC-1α and UCP-1 and osteogenic markers Runx2 and osteocalcin were significantly decreased. Number of UCP-1 immunostaining-positive brown adipocytes also reduced in osteoporotic bone. In vitro, DNA methylation inhibitor 5'-aza-deoxycystidine significantly increased brown adipocyte formation and osteogenic differentiation and mitigated dexamethasone-induced white adipocyte formation in mesenchymal stem cells. 5'-aza-deoxycystidine control of brown adipogenesis and white fat formation appeared to be regulated by increasing Wnt3a/β-catenin and reducing Dkk1. Disintegrated brown adipocyte and white fat cell differentiation contribute to osteoporosis pathogenesis. Maintaining DNA hypomethylation promotes Wnt signalling and brown adipocyte differentiation facilitating osteogenic differentiation. This study shed a new light to the contribution of brown adipocytic cells to bone metabolism during osteoporosis.


K. Dalgarno M. Benning S. Partridge A. Tulah S. Ahmed A. Dickinson P. Genever R. Pearson G. Feichtinger J. Loughlin A-M Ferreira-Duarte

This paper reports on a proof of concept project funded by the UK National Council for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), with the aim of developing an in vitro model to recapitulate the human osteoarthritic joint, based on a multiple human cell type co-culture system, for research and drug development in OA. The targets were: (i) the development of a cell culture platform that could produce a mixed stable cell culture of cell types that represent the key components of the human joint: synoviocytes – type I and type II; osteoblasts; osteoclasts; chondrocytes/cartilage or cartilage-like matrix; adipocytes; and immune cells. (ii) demonstration of cell phenotype stability and viability for at least 72 hours. In order to establish the cell culture platform we have developed an eight-channel cell printer, capable of accurately and reliably printing the required cell types to create osteochondral and synovial cell types within a transwell system. Two different sets of cells have been developed and processed using the cell printer: a set based on using an immortalised hTERT MSC line to create osteoblasts, chondrocytes and adipocytes, with commercial cells lines providing the other cell types, and a set obtained from tissue excised during orthopaedic surgery. This gives both a repeatable set of cells with which to undertake mode of action studies, and a bank of cell sets which will be representative of different stages of osteoarthritis. The co-cultures have been immunohistochemically assessed in order to demonstrate maintenance of phenotype.


G. Vadalà

Irisin is a hormone-like myokine released from skeletal muscle during exercise. It has also been reported that irisin levels in serum and synovial fluid of knee osteoarthritis (OA) patients were negatively correlated with OA severity. We hypothesized that irisin might play a role in the cartilage homeostasis mediated by physical activity. Therefore, this study aims to explore the cross talk between skeletal muscle and cartilage tissues in human with OA mediated by the myokine irisin. Human articular OA chondrocytes were isolated, expanded and cultured in micro-mass 3-D culture system. Pellets were cultured with or without r-Irisin, and then activated by protein inhibitors of p38-MAPK signalling pathway. After one week the amount of GAG content was evaluated. Quantitative gene expression of Coll-X and Coll-II was performed. WB was utilized to detect expressions of p38-MAPK signalling pathway and Coll-X and Coll-II. In the current study, chondrocytes cultured in r-Irisin showed a significant higher GAG/DNA content compared to control (p<0.05). Moreover, r-Irisin promoted a significant increase of the expression collagen type II and decrease of collagen type X in (p<0.05). This OA chondrocytes recovery was abrogated by the p38 MAPK and ERK signalling pathways. Our observation suggests that Irisin targets chondrocytes promoting GAG content, increasing Collagen Type II and decreasing Collagen type X gene expressions. The observed OA chondrocyte recovery mediated by irisin is obtained through the inactivation of p38/ERK MAP kinase signalling cascades in vitro. This is the first study that demonstrates a cross-talk between muscle and cartilage mediated by irisin.


S. Kearns

The goal of surgery for osteochondral lesions is to regenerate the damaged cartilage with ideally hyaline cartilage. The current gold standard treatment is bone marrow stimulation (BMS) by microfracture. In reality however BMS typically results in the generation of fibrocartilage. Orthobiologics including bone marrow aspirate, platelet rich plasma and hyaluronic acid products have been shown to promote cartilage healing and potentially increase the formation of hyaline cartilage in treated lesions. However the role of these products, the timing of their administration and frequency of application are still not clearly defined and their routine use is still not recommended. These issues and future directions for research and future clinical application will be discussed.


E. Kunisch A.K. Knauf E. Hesse F. Bothe S. Diederichs W. Richter

Engineered cartilage is poorly organized and fails to recapitulate physiologic organization in a hyaline upper and a mineralizing bottom zone deemed important for proper function. Objective was to grow bizonal human cartilage constructs in which in vivo mineralization is self-restricted to the bottom zone. Self-assembling biomaterial-free cell discs were generated from mesenchymal stroma cells and allowed to accumulate proteoglycans and collagen-type II over 3 weeks. In vitro mineralization of the cell discs with four mineralization media for up to 8 weeks showed that calcification was supported in all media containing ß-glycerophosphate. However, proteoglycans were retained only in media containing insulin. Bizonal cartilage constructs were made from 3-week non-mineralized cell discs overlaid with chondrocyte-seeded starPEG-heparin hydrogel or with a fibrin-gel layer to select the best design for upper zone development. Freshly prepared zonal constructs were implanted into subcutaneous pouches of immuno-deficient mice to compare in vivo development. After 6 weeks in vivo, both construct types were rich in collagen-type II in the upper zone and contained a mineralized bottom zone. However, solely for starPEG constructs, tissue volume of the upper zone remained high and alkaline phosphatase, alizarin red, and collagen-type X staining were restricted to the bottom zone. StarPEG zonal constructs were superior to fibrin constructs due to self-restriction of mineralization and hypertrophic markers to the bottom zone. This innovative design of bizonal constructs offers the successful generation of an organized cartilage resembling the native cartilage with the chance for immediate use of autogenous chondrocytes in a one-step surgical joint intervention.


S. Capar G.J.V.M. van Osch J.A.N. Verhaar Y.M. Bastiaansen-Jenniskens

Joint injuries often result in inflammation and cartilage defects. When inflamed, the synovium secretes factors that prevent successful cartilage repair by inhibiting chondrogenic differentiation of progenitor cells. In particular the pro-inflammatory macrophages in the synovium are indicated to contribute to this anti-chondrogenic effect. Thus, we aimed to counteract the anti-chondrogenic effect of inflamed synovium by modulating synovial inflammation and its macrophages. Synovium tissue obtained from osteoarthritic patients undergoing a total knee replacement was cut into explants and cultured for 72 hours +/− 1 µM of the anti-inflammatory drug triamcinolone acetonide (TAA) (Sigma Aldrich). TAA significantly decreased gene expression of TNFA, IL1β and IL6, and increased expression of CCL18, IL1RA in synovial explants (all with p < 0.001). On the other hand, TAA significantly decreased the percentages of pro-inflammatory CD14+/CD80+ and CD14+/CD86+ macrophages in the synovium (both p < 0.001) as assessed by flow cytometry analyses. The percentages of anti-inflammatory CD14+/CD163+ macrophages, is significantly increased (p < 0.001) in TAA treated synovium. Conditioned medium (CM) from synovium explants downregulated the gene expression of cartilage matrix components collagen type-2 and aggrecan expression in chondrogenic MSCs. This chondrogenesis inhibiting effect was reduced by treating synovium with TAA during the production of the CM. Our findings indicate that reducing synovial inflammation might improve the joint environment for better cartilage repair, possibly by modulation of macrophage phenotypes.


R.L. Boyanich J.P. Wu T.B. Kirk

High resolution imaging techniques such as atomic force microscopy, provide a platform to study the fibrillary architecture of biological tissues, but are not capable of imaging the internal microstructure of tissues in 3D. Conversely, multiphoton microscopes facilitate 3D imaging to study the spatial relationship of micro-components within tissues, but without the resolution of atomic force microscopy. The lamina splendens is the most superficial layer of articular cartilage. It is believed to play a crucial role in the health of the tissue. However, the precise form of this layer is uncertain as it has never been independently studied. Here, we use multiphoton microscopy and atomic force microscopy to demonstrate the anatomic form of the lamina splendens. The lamina splendens were peeled from the femoral condyles of healthy, adult sheep (n=20). Using atomic force microscopy, we show that the collagen and elastin form an interweaving fibrillary network at the surface of the lamina splendens and at the interface of the lamina splendens with the underlying cartilage. Moreover, using fluorescent stains; sulforhodamine B and acridine orange, multiphoton microscopy shows the heterogeneous distribution of collagen, elastin and chondrocytes throughout the depth of the lamina splendens. Our results demonstrate the fibrillary and component level architecture of the lamina splendens. We believe our findings provide the backbone of knowledge to advance tissue engineering techniques that will lead to more promising strategies to treat cartilage pathologies, including osteoarthritis. Furthermore, our results provide a starting point to determine the role of the lamina splendens in cartilage pathology.


G. Pattappa J. Zellner B. Johnstone D. Docheva P. Angele

Mesenchymal Stem Cells (MSCs) are a candidate cell type for treating osteoarthritic focal defects. In vivo, cartilage and bone marrow reside under a low oxygen tension, between 2–7% oxygen or physioxia, that has been shown to enhance MSC chondrogenesis. However, chondrogenesis is inhibited in the presence of IL-1. Here, it was hypothesized that physioxia reduces IL-1 inhibited chondrogenesis. Human MSCs (Mean age, 32 years; n = 9) were split equally for expansion under either 2% (physioxia) or 20% (hyperoxia) oxygen. Chondrogenic pellets (2 × 105 MSCs/pellet) were formed and cultured in the presence of 10 ng/ml TGF-b1 and in combination with either 0.1 or 0.5 ng/ml IL-1 under their respective expansion conditions. Pellets were assessed for their wet weight, GAG and collagen II content and evaluated histologically (Collagen X and MMP-13). Statistical analysis was performed using a Two-way ANOVA with Tukey post-hoc test, significant differences stated when p < 0.05. A significant dose-dependent IL-1 inhibition in chondrogenesis was observed for pellet wet weight and GAG content under hyperoxia (p < 0.05). Physioxia alone significantly increased wet weight, GAG and collagen II content (p < 0.05) compared to hyperoxia. A donor-dependant response was observed, whereby 80% of donors responded to physioxia and their analysis showed significant increases in wet weight and GAG content in the presence IL-1(p < 0.05). Furthermore, reduced hypertrophy marker expression (Collagen X and MMP-13) was observed under physioxia in the presence of IL-1. The molecular signalling mechanisms controlling these responses are to be investigated.


D. Mantovani

Over the last 50 years, biomaterials, prostheses and implants saved and prolonged the life of millions of humans around the globe. The main clinical complications for current biomaterials and artificial organs still reside in an interfacial mismatch between the synthetic surface and the natural living tissue surrounding it. Today, nanotechnology, nanomaterials and surface modifications provides a new insight to the current problem of biomaterial complications, and even allows us to envisage strategies for the organ shortage. Advanced tools and new paths towards the development of functional solutions for cardiovascular clinical applications are now available. In this talk, the potential of nanostructured metallic degradable metals to provide innovative solutions at medium term for the cardiovascular field will be depicted. Focus will be on Fe-based biodegradable metals with exceptional resistance, ductility and elasticity, for pushing innovative vascular applications. The intrinsic goal of this talk is to present an extremely personal look at how biodegradable metals can impact materials, surfaces and interfaces, and how the resulting unique properties allowed biomedical functional applications to progress, from their introduction, to the promising future that biodegradable metals may or may not hold for improving the quality of the life of millions worldwide.


Y. Zheng

In order to evaluate the feasibility of zinc alloys as future biodegradable bone implant materials, the mechanical properties, corrosion resistance, hemocompatibility, cell activity, proliferation and adhesion, in vivo animal implantation experiments have been employed. The experimental results show that the alloying element magnesium, calcium and strontium can significantly improve the mechanical properties of pure zinc, and further deformation processes can further improve the mechanical properties of zinc alloys. Alloying elements can effectively control the corrosion rates of zinc alloys, which are between the rates of magnesium alloys and iron alloys. Zinc and zinc alloys exhibit excellent hemocompatibility and the hemolysis rate is far lower than 5%. After adding alloying elements Mg, Ca and Sr, MG63 and ECV304 cell proliferation rate and activity increased significantly, while for VSMC cell, the influence of alloying elements effect is not obvious. Zinc alloy intramedullary pins can effectively promote the new bone formation, and after 2 months implanted in mice femur, they still maintained a relatively complete structure, indicating that they are able to provide enough mechanical strength and thus more conducive to bone tissue repair and healing.


S. Ribeiro V. Novacek E.M. Fernandes M.E. Gomes R.L. Reis Y. Bayon D.I. Zeugolis

To repair soft tissue, it is vital to ensure that the biomaterial is able to mimic the complex elasticity of the native tissue. It has been demonstrated that substrate stiffness has a huge influence on cellular growth, differentiation, motility and phenotype maintenance. The goal of the present study is to characterize extensively a set of polymeric films with variable mechanical profiles. A range of synthetic biodegradable polymers was selected according to the physico-chemical intrinsic properties of aliphatic polymers. They have similar chemistry (absorbable polyesters made from lactic acid, glycolic acid, trimethylene carbonate, dioxanone & β-caprolactone), however show different mechanical and degradation properties. The films were manufactured by thermal presser and then characterized by scanning electron microscopy (SEM), differential scanning calorimetry (DSC), nuclear magnetic resonance spectroscopy (NMR) and Fourier transform infrared spectroscopy (FTIR). The mechanical properties of the films were assessed by uniaxial tensile tests in wet conditions and also by atomic force microscopy (AFM) to assess the material's stiffness at a micro-level. In vitro assays were performed to assess the cell cytocompatibility, proliferation and differentiation potential of the films. The mechanical properties of the materials are within the range intended for musculoskeletal tissue repair. Biological assays showed good cell adhesion, cell proliferation and cell viability. Stem cells were able to differentiate into adipogenic, osteogenic, chondrogenic and tenogenic lineages. Overall the selection of polymers gave good options for a potential tissue repair scaffold. In the future, the combined effect of stiffness and topography will be assessed on cell phenotype maintenance.


H. Capella-Monsonís D.I. Zeugolis

Collagen materials are extensively used in regenerative medicine. However, they still present limitations such as a mono-domain composition and poor mechanical properties. On the other hand, tissue grafts overcome most of these limitations. In addition, the potential of tissue grafts in musculoskeletal tissue engineering has not been fully investigated. Herein, we ventured to assess the potential of a decellularised porcine peritoneum for musculoskeletal applications by comparing its characteristics with a commercial collagen scaffold employed in tendon. Results indicated that the porcine peritoneum had higher mechanical properties and a lower crosslinking ratio (p < 0.01). Furthermore, it presented a lower resistance to collagenase digestion, which suggests a faster remodelling in vivo of the tissue graft. Immunohistochemistry analysis showed a preserved and multicomponent structure in the porcine peritoneum contrary to the collagen matrix, confirming the multifunctional nature of the tissue graft. Regarding the cell-response assessment, tenocytes and ADSCs were able to grow on both materials, however, proliferation was enhanced by the porcine peritoneum (p<0.01). Immune response by THP-1 showed an acute inflammatory response by macrophages to the collagen matrix, contrary to that observed in the porcine peritoneum which triggered a mild reaction. The in-progress in vivo study in a rabbit tendon model will elucidate the potential of porcine peritoneum for tendon repair applications. The present study shows how the multifunctionality of the porcine peritoneum provides higher cytocompatibility than a mono-domain collagen matrix with human tenocytes and ADSC. Besides, its lower immune response in vitro suggests better remodelling after implantation.


Z. Bal T. Kaito H. Ishiguro P. Korkusuz E.C. Dede F. Korkusuz

Sustained release of BMP-2 is reported to be able to reduce the required dose of BMP-2 for bone induction. Nanohydroxyapatite (nHAp) has an osteoinduction capability which is lack in conventional hydroxyapatite. In this study, we combined PLA-PEG with nHAp and investigated the bone regenerative capacity of the newly established composite material of rhBMP-2/PLA-PEG/nHAp in a rat model of spinal fusion. The PLA-PEG was liquidized in acetone and mixed with nHAp and rhBMP-2. The sheet-shaped BMP-2/PLA-PEG (5mg)/nHAp (12.5mg) composites were prepared while evaporating the acetone. The release kinetics of rhBMP-2 from the composite was investigated by ELISA. In vivo bone formation was investigated by posterolateral spinal fusion in rats (the dosage of rhBMP-2; 0µg/ 0.5µg / 3µg). Bone formation was assessed by µCT and histology at post-op. 8 weeks. The composite showed the burst-release in the initial 24 hours (69% of total release) and the subsequent sustained-release for 25 days. According to µCT and histology of the spinal fusion experiment for all groups the bone formation was observed. While no bony bridging was observed in 0 µg and 0.5 µg BMP groups; in 3 µg group bony bridging and fusion were achieved. We developed a new technology for bone regeneration with rhBMP-2/PLA-PEG/nHAp composite. The reduction in the required dose of BMP-2 for bone induction was achieved. This result can be explained by the high bone induction ability of nHAp and sustainable release of BMP from PLA-PEG in the composite.


Y. Li P. Pavanram J. Zhou M.A. Leeflang B. Pouran K.-U. Schröder H. Weinans T. Pufe A.A. Zadpoor H. Jahr

The ideal bone substituting biomaterials should possess bone-mimicking mechanical properties; have of porous interconnected structure, and adequate biodegradation behaviour to enable full recovery of bony defects. Direct metal printed porous scaffolds hold potential to satisfy all these requirements and were additively manufactured (AM) from atomized WE43 magnesium alloy powder with grain sizes between 20 and 60 μm. Their micro-structure, mechanical properties, degradation behavior and biocompatibility was then evaluated in vitro. Firstly, post-processing values nicely followed design parameters. Next, Young's moduli were similar to that of trabecular bone (i.e., E = 700–800 MPa) even after 28 days of simulated in vivo-like corrosion by in vitro immersion. Also, a relatively moderate hydrogen evolution, corresponding to a calculated 19.2% of scaffold mass loss, was in good agreement with 20.7% volume reduction as derived from reconstructed μCT images. Finally, only moderate cytotoxicity (i.e., level 0, <25%), even after extensive ISO 10993-conform testing for 72 h using MG-63 cells, was determined using WE43 extracts (2 way ANOVA, post-hoc Tukey's multiple comparisons test; α = 0.05). Cytotoxicity was further evaluated by direct live-dead staining assays, revealing a higher cell death in static culture. However, intimate cell-metal contact was observed by SEM. In summary, while pure WE43 may not yet be an ideal surface for cell adhesion, this novel AM process allows for adjusting biodegradation through topological design. Our approach holds tremendous potential to develop functional and biodegradable implants for orthopaedic applications.


C. M. Coleman

Long bone fractures in patients with diabetes mellitus (DM) are slow to heal, often resulting in delayed reunion or non-union. It is reasonable to postulate that the underlying cause of these DM-associated complications is a reduced population of bone marrow progenitor cells and/or their dysfunction. With the hypothesis that the administration of healthy, allogeneic adult bone marrow-derived mesenchymal stromal cells (MSCs) can enhance DM fracture healing, the aim of this endeavour was to assess the efficacy of MSC administration to support fracture repair using two doses. Here 250,000 or 500,000 human bone marrow-derived MSCs were locally introduced to femoral fractures in diabetic mice, and the quality of de novo bone assessed 8 weeks later. Preliminary bone bridging was evident in all animals; however, a large circumferential reparative callus was consistently retained indicating non-union. Micro-CT analysis elucidated consistent callus dimensions, bone mineral density, bone volume/total volume in all groups, but an increase in bone surface area/bone volume in cell-treated fractures. Moreover, greater amounts of mature bone were identified in fractures treated with a low dose of MSCs. Four-point bending evaluation of the mechanical integrity of the repairing fracture indicated a statistically significant improvement in flexure strength and flexure modulus in DM fractures treated with 250,000 MSCs as compared to controls. An improvement in total energy required for failure was observed in both groups that received MSCs. Therefore, the administration of non-DM bone marrow-derived MSCs supported the development of more mature bone in the reparative callus, resulting in greater mechanical integrity.


J. P. O'Connor

Cyclooxygenase-2 (COX-2) activity is necessary for fracture healing to proceed normally. In most cell types, COX-2 is inductively expressed and acts in a coordinated pathway to produce prostaglandins, which affect many physiological processes including inflammation. In the fracture callus, however, COX-2 expression and the molecular and cellular processes affected by COX-2 activity remain poorly understood. Using LC-MS/MS and xMAP, we measured fracture callus prostaglandin and inflammatory cytokine levels. We found that inflammatory cytokines rapidly peaked after fracture before declining to normal levels by day 4 after fracture. However, callus prostaglandin levels did not peak until 4 days after fracture before returning to normal levels by day 10. We used immunohistochemistry to detected COX-2 expression in callus cells and found that COX-2 was expressed in callus chondrocytes and osteoclasts during endochondral ossification, including those osteoclasts at the callus chondro-osseous junction. Targeted deletion of the COX-2 gene (Ptgs2) in osteoclasts or in chondrocytes was found to delay fracture healing. Using cell-based experiments, we found that COX-2 expression could be induced in osteoclasts by osteopontin treatment, suggesting an integrin-dependent induction of COX-2 expression in osteoclasts. This was confirmed in vivo using mice lacking osteopontin or integrin ß3. Immunohistochemistry also showed abundant osteopontin expression at the callus chondro-osseous junction. The results indicate that COX-2 expression in osteoclasts is controlled by integrin-dependent signalling, that COX-2 expression in osteoclasts and chondrocytes is necessary for fracture healing to proceed normally, and that COX-2 expression in chondro-osseous junction osteoclasts may be induced by osteopontin-dependent signalling by chondrocytes.


C. Sanz-Nogués M. Mustafa H. Burke T. O'Brien C. M. Coleman

Patients living with type 1 diabetes mellitus (T1DM) can develop early onset osteoporosis and are exposed to an increased risk of fracture. Skeletal health can be influenced easily with diet and exercise. However, diabetes mellitus (DM)-related osteopathy is not emphasized in the public information campaigns on the American Diabetes Association, Diabetes UK, Diabetes Ireland or International Diabetes Federation websites. This investigation aims to assess the perceptions of patients regarding living with T1DM and their baseline knowledge on DM-related osteopathy. A survey was administered to 102 consenting individuals living with T1DM in attendance at the Galway University Hospital Diabetes Centre. Of the respondents, 44% were female and 56% male (mean age of 43). Respondents had T1DM for a mean of 21 years. Participants were asked to identify DM-related complications, including bone thinning and bone fractures. Respondents were primarily concerned about developing DM-related blindness, kidney damage and amputations, but not osteopathy. 49% of respondents did not identify osteopathy as a potential DM-related complication, 28% of respondents related DM with bone thinning and bone fractures, and 22% individuals only identified bone thinning or bone fractures. When asked for their primary source of DM-related information, endocrinologists and internet where identified. When comparable questions were asked of DM-related healthcare professionals, 56% did not recognize osteopathy as a complication of T1DM. This study demonstrated a low-level awareness of the impact living with T1DM has on bone health. The deployment of patient-interactive activities or educational modules may enhance the future health of individuals living with T1DM.


D.B. Raina I. Qayoom D. Larsson M.H. Zheng A. Kumar H. Isaksson L. Lidgren M. Tägil

Metaphyseal fracture healing is important in joint-adjacent fractures and appears to differ from diaphyseal healing. We recently found that a biomaterial delivering bone morphogenic protein-2 (BMP-2) and zoledronic acid (ZA) healed the metaphyseal bone in a tibial defect but failed closing the cortical defect. In this study we added a BMP-2 soaked collagen membrane to study cortical healing from the muscle tissue surrounding the bone. We used SD rats and a 4.5 mm metaphyseal circular tibial defect. In group 1 (G1), a porous gelatin-calcium sulphate-hydroxyapatite (GCH) biomaterial containing rhBMP-2 and ZA was used to fill the defect (GCH+5 μg BMP-2+10 μg ZA). In group 2 (G2), we used a collagen membrane (2 μg BMP-2) to cover the GCH filled defect (GCH+3μg BMP+10 μg ZA). Group 3 (G3) was an empty control. Animals were sacrificed after 8-weeks and bone regeneration was evaluated with micro-CT and histology. In both G1 (P<0.001) and G2 (p<0.001) a significantly higher mineralized volume was found in the defect compared to empty G3. In G2 higher mineralized volume was found in the cortical region compared to both G1 (p<0.01) and G3 (p<0.001) as seen via micro-CT. Histologically, G1 and G2 showed islands of trabecular bone in the defect peripherally but only G2 showed cortical healing. G3 was empty in the middle but showed healed cortex. In conclusion, GCH can be used to deliver BMP-2 and ZA to promote metaphyseal bone growth. A membrane (CM) doped with low dose BMP-2 improved cortical regeneration.


W.S. Lian J.Y. Ko F.S. Wang

Sclerostin (SOST) is an endogenous inhibitor of Wnt/β-catenin signalling pathway to impair osteogenic differentiation and bone anabolism. SOST immunotherapy like monoclonal antibody has been observed to control bone remodeling and regeneration. This study is aimed to develop a SOST vaccine and test its protective effects on estrogen deficiency-induced bone loss in mice. Gene sequences coded SOST peptide putative targeting Wnt co-receptor LRP5 were cloned and constructed into vectors expressing Fc fragment to produced SOST-Fc fusion protein. Mice were subcutaneously injected SOST-Fc to boost anti-SOST antibody. Bone mineral density, microstructure, and mechanical property were quantified using μCT scanning and material testing system. Serum bone formation and resorption markers and anti-SOST levels were measured using ELISA. SOST-Fc injections significantly increased serum anti-SOST antibody levels but reduced serum SOST concentrations. SOST-Fc vaccination significantly reduced estrogen deficiency-induced serum bone resorption markers CTX-1 increased serum bone formation marker osteocalcin. Of note, it significantly alleviated the severity of estrogen-induced loss of bone mineral density, trabecular morphometric properties, and biomechanical forces of bone tissue. Mechanistically, SOSF-Fc vaccination attenuated trabecular loss histopathology and restored immunostaining of Wnt pathway like Wnt3a, β-catenin, and TCF4 in bone tissue along with increased serum osteoclast inhibitor OPG levels but decreased serum osteoclast enhancer RANKL concentrations. Taken together, SOST-Fc vaccination boosts anti-SOST antibody to neutralize SOST and mitigates the estrogen deficiency-induced bone mass and microstructure deterioration through preserving Wnt signalling. This study highlights an innovative remedial potential of SOST vaccine for preventing osteoporosis.


F. Vincenzi S. Pasquini S. Setti M. Cadossi P.A. Borea R. Cadossi K. Varani

Several studies explored the biological effects of low frequency low energy pulsed electromagnetic fields (PEMFs, Igea Biophysics Laboratory, Carpi, Italy) on human body reporting different functional changes. In the orthopedic field, PEMFs have been shown to be effective in enhancing endogenous bone and osteochondral repair, incrementing bone mineral density, accelerating the process of osteogenic differentiation and limiting cartilage damage. Much research activity has focused on the mechanisms of interaction between PEMFs and membrane receptors such as adenosine receptors (ARs). In particular, PEMF exposure mediates a significant upregulation of A2A and A3ARs expressed in various cells or tissues involving a reduction of most of the pro-inflammatory cytokines. In tissue engineering for cartilage repair a double role for PEMFs could be hypothesized: in vitro by stimulating cell proliferation, colonization of the scaffold and production of tissue matrix; in vivo after surgical implantation of the construct by favoring the anabolic activities of the implanted cells and surrounding tissues and protecting the construct from the catabolic effects of inflammation. Of particular interest is the observation that PEMFs, through the increase of ARs, enhance the working efficiency of the endogenous modulator adenosine, producing a more physiological effect than the use of exogenous drugs. This observation suggests the hypothesis that PEMFs could be considered a non-invasive treatment with a low impact on daily life. In conclusion, PEMFs represent an important approach in the pharmacological field providing excellent therapeutic results in various inflammatory diseases and in particular in the functional recovery of the damaged joint tissues.


M. Dalby

In this presentation, the response of mesenchymal stem cells (MSCs) to nanoscale cues (e.g. topography, chemistry and vibrations) will be considered. In particular, control of MSC self-renewal and differentiation. A focus will be on a new bioreactor that has been developed, the nanokick, that delivers precise nanovibrational cues to MSC cultures in 2D and 3D, driving the cells to turn into mineralizing osteoblasts. Mechanotransductive signalling will be considered looking at ion channel mediated differentiation.


J. de Boer

Our lab uses computer-aided design to build in silico libraries of surface topographies, which we reproduce on polymeric chips and analyse for cellular responses using high content imaging and machine learning. In addition, we use transcriptomics and mass spectrometry to obtain a holistic view of biomaterial-mediated cellular responses and build gene regulatory networks thereof. This approach enables us to parameterize both the biomaterial properties as well as the cell response and to correlate them using computational tools. We think that this approach can be translated to other biomaterial platforms, such as polymer arrays, and foresee large scale crosstalk between them if we can standardize our methodology to describe the materials and to analyse the cells. To this end, we have started cBIT, the compendium for biomaterial-induced transcriptomics, an open-source database in which scientists can deposit and search material-induced transcriptomics data. The meta-analyses that cBIT enables, could lead to the identification of genes, pathways or expression profiles that can inform the design and development of new biomaterials. As such, by generating new information and simultaneously accumulating it in cBIT, we expect it is possible to one day predict cell responses to biomaterials.


H. Yin C. Popov M. Schieker M. Nerlich D. Docheva

Background: The exact pathways of collagen remodeling in tendon tissue are not well understood. Therefore, we have established a 3D collagen gel system and studied the remodeling capacity of two different TSPC lines: young, Y-TSPC and aged/degenerative, A-TSPC. We specifically investigated the involvement of integrin receptors in the remodeling process. Methods: Y- and A-TSPC were derived from human Achilles tendon. RT-PCR was used to assess the expression of collagen-binding integrins. Integrins a1 and a11 were silenced by lentiviral delivery of shRNA in the Y-TSPC. Control-shRNA, a1-shRNA and a11-shRNA virus was given for 24h and then cells were selected with zeocin for 10 days. The integrin knockdown (KD) efficiency was assessed by quantitative PCR and western blotting. Last, time-lapse recording of gel contraction of Y-TSPC+con, Y-TSPC+a1KD, Y-TSPC+a11KD, and A-TSPC were performed. Results: Integrin a1 and a11 were significantly downregulated in A-TSPC. Therefore, to mimic the A-TSPC we carried out a1 and a11 KD in Y-TSPC. PCR and western blot validated very efficient KD. Analyses of collagen contraction revealed that Y-TSPC+a11KD had significant reduction in collagen contractibility comparable to A-TSPC phenotype. Regarding integrin a1, we found that this receptor had no effect on the contraction rate of TSPC. Thus, to our knowledge we have now identified for the first time a novel role of a11 integrin in tendon matrix remodeling, and a follow up analyses of the exact downstream cascade are on the way.


L. Aubert L. Entz M. Dubus H. Rammal C. Mongaret J. Lesieur S. C. Gangloff C. Chaussain C. Mauprivez H. Kerdjoudj

Mesenchymal stem cells (MSCs) are tissue-resident stroma cells capable of modulating immune cells through the secretion of paracrine factors. However, the comparison of MSCs potential, from different sources and submitted to hypoxia within a 3D scaffold, in secreting pro-healing factors has never been investigated. With a chemical composition similar to type I collagen, a major component of connective tissues retrieved in dental pulp, bone and umbilical cord, Hemocollagene® haemostatic foam presented porous and interconnected structure (> 90%) and a relative low elastic modulus of around 60 kPa. All these criteria meet basic requirements for tissue engineering based material. Herein, we assessed and compared the effect of hypoxia (3% O2) on the regulation and release of pro-angiogenic factors (VEGF, b-FGF and IL-8) from bone marrow (BM), Wharton's jelly (WJ) and dental pulp (DP) derived MSCs cultured in Hemocollagene®. After 10 days of culture, qRT-PCR analysis showed an up-regulation of b-FGF and VEGF mRNA in BM- and WJ-derived MSCs, but not in DP-derived MSCs. Furthermore, hypoxia highly up-regulated IL-8 expression in WJ-derived MSCs and moderately in both BM and DP-derived MSCs. In contrast, ELISA analysis showed a higher amount of VEGF and IL-8 in supernatant provided from DP-derived MSCs culture compared to BM and WJ-derived MSCs. B-FGF was not detected whatever the experimental condition. In conclusion, MSCs derived from several tissues were able to release pro-angiogenic factors under hypoxic conditions. There was no clearly superior type of MSCs for therapeutic use, however DP-derived MSCs are likely to be more advantageous.


D. De Troy L. Hertzog S. Normand A. Tury R. Baron S. Pietri

Mesenchymal Stromal Cells (MSC) are promising therapies for fracture healing. However, undifferentiated MSC may act only through an inductive paracrine effect without direct bone formation. Here, we developed an injectable product constituted of human bone-forming cells derived from bone marrow (BM)-MSC (ALLO-P2) that display more potent bone repair properties not only by stimulating host osteoinduction but also by direct bone formation. In vitro, ALLO-P2 overexpressed markers such as ALP compared to BM-MSC isolated from the same donors, suggesting their engagement into the osteogenic lineage. In vivo, a single dose of ALLO-P2 significantly enhanced bone neoformation 14 days post-administration over the calvaria of NMRI-Nude mice compared to the control excipient. Histological analyses and mouse/human type I collagen double-immunolabelling revealed the presence of mineralized bone nodules of mixed host and donor origins in mice administered with ALLO-P2. Together, these results show that ALLO-P2 is a potential promising clinical candidate to promote bone repair, since it can be produced at high yields, is injectable and boosts ossification mechanisms involved in the physiological repair process.


S. Du D.I. Zeugolis T. O'Brien

Macromolecular crowding (MMC) is a biophysical phenomenon that accelerates thermodynamic activities and biological processes by several orders of magnitude. Herein, we ventured to identify the optimal crowder and to assess the influence of MMC in umbilical cord mesenchymal stem cell. 7 types of carrageenan (κ&λ, κ-LV1, κ-LV2, λ-MV, λ-HV, ι-MV, ι-HV) acted as crowder and biophysical properties were assessed respectively. Human umbilical cord mesenchymal stem cells were seeded at 15,000 cells/cm2 in 24 well plates and allowed to attach for 24 h. Subsequently, the medium was changed to medium with 7 types of carrageenan (10, 50, 100, 500 μg/ml) and 100 μM L-ascorbic acid phosphate (Sigma Aldrich). Medium without carrageenan was used as control. Cell morphology and SDS-PAGE analysis were conducted after 3, 5 and 7 days. Biophysical assessment showed 7 types of carrageenan have increased particle size with concentration, good polydispersity and negative charges. SDS-PAGE and densitometric analyses revealed significant increase (p < 0.001) in collagen deposition in the presence of 10 μg/ml carrageenan λ and ι at all the time points. SDS-PAGE and densitometric analysis also showed that the highest collagen deposition was observed in culture at 50 μg/ml carrageenan λ. No significant difference was observed in cell morphology between the groups. Collectively, these data primarily illustrate the beneficial effect of carrageenan λ in human umbilical cord mesenchymal stem cell culture.


B. Nolan

Increasingly more emphasis is being placed on Patient Reported Outcome Measures (PROMs). There are many used and reported in clinical studies, but there are no universally accepted or preferred measures. It is important for a researcher with a non-clinical background to understand how these assessments are performed, the type of information provided by each of the measures, and which diseases states are best reported by each measure.


Full Access
R. Aaron

OA pathophysiology has a vascular component consisting of venous stasis resulting in intraosseous hypertension and hypoxia. In response, osteoblasts change their cytokine expression, accelerating bone remodelling and cartilage breakdown consistent with OA. We have characterized circulatory kinetics in OA bone in animal models with dynamic contrast enhanced MRI (DCE-MRI) and 18F PET and have demonstrated venous stasis and reduced perfusion that temporally precede and spatially coincide with OA lesions. Osteoblast uptake of 18F is consistent with abnormal perfusion, bone remodelling, and severity of OA. Circulatory kinetics with DCE-MRI in humans with OA of the knee exhibit similar venous outflow obstruction. Venous stasis is associated with hypoxia in subchondral bone. As an example of the effects of hypoxia on OA osteoblasts, we have described upregulation of fibrinolytic peptides, but a deficiency in the upregulation of PAI-1, leading to the generation of plasmin by human OA osteoblasts exposed to hypoxia in vitro. Plasmin is a serine protease that has been shown to degrade cartilage in OA. Abnormal circulatory kinetics by DCE-MRI may be an imaging biomarker of OA. Pharmacologic modulation of venous stasis would have a salutary effect on the physicochemical microcirculation of subchondral osteoblasts and the pathophysiology of OA.


H. J. Meisel

Cervical and lumbar spine fusion procedures are increasing every year. Nonetheless, these procedures are associated with high infection rates, resulting in additional cost burden. The conundrum of achieving efficient spinal fusions with minimum complications requires an ideal bone graft with osteoconductive, osteoinductive, osteogenic and structural characteristics. Synthetic bone graft substitutes with or without autograft, allograft or synthetic bone substitutes have been commonly used for fusion procedures. We carried out a meta-analysis of comparative studies and prospective case series (n = 29) with cervical and lumbar fusion procedures using synthetic bone graft substitutes, autograft or allograft and other biologics. Synthetic bone graft substitutes analysed included HA (Hydroxyapatite), β-TPC (Tri Calcium Phosphate), β-TSC (Tri Calcium Sulfate), PMMA (Polymethylmetacrylate), Surgibone, BOP (Biocompatible Osteoconductive Polymer). The analysis revealed suboptimal evidence for the efficacy and safety of synthetic products used in spinal fusion procedures. Further studies are needed to determine beneficial effects of synthetic substitutes. However, the infection rate could be highly decreased with surface and composition modification of widely used polyether ether ketone (PEEK) implants. Laser modification of surface characteristics and collagen fleeces with micro and nano pore structures can prove to be excellent surface for increased osteoblasts cell proliferation and vitality.


C. Barlow E. Dominguez G. Dixon H. Crouch-Smith R. Wallace H. Simpson K. Al-Hourani

Femoral shaft fractures are potentially devastating injuries. Despite this, clinical studies of the biomechanics of this injury are lacking. We aimed to clinically evaluate bone behaviour under high and low energy trauma in paediatric, adult and older patients. Single-centre retrospective study identifying all diaphyseal femoral fractures between Feb 2015-Feb 2017. Peri-prosthetic and pathological fractures were excluded. Patients were subdivided into groups 1 (paediatric, <16yo), 2 (adult, 17–55yo) and 3 (older, >55yo) to reflect immature, peak bone age and osteoporotic bone respectively. Chi-Squared analysis assessed significance of bone age to degree of comminution and fracture pattern. A p-value <0.05 was significant. A total 4130 radiographs were analysed with 206 femoral shaft fractures identified. Forty-three patients were excluded with 163 remaining. Group 1, 2 and 3 included 38, 37 and 88 patients respectively. Mean age 50.8 (SD 32.8) with male-to-female ratio of 1:1.2. Groups 1 and 3 included majority simple fractures (35/38 and 62/88 respectively). Group 2 included more comminuted injuries (33/37). Bone age to degree of comminution proved significant (p<0.05) with a bimodal distribution of simple fractures noted in groups 1 and 3. Energy to fracture was significant in group 2, where a high energy injury was associated with comminution (p<0.05). This study is the first to demonstrate an association between fracture comminution and age. Simple femoral shaft fractures showed a bimodal age distribution in paediatric and older patients regardless of mechanism energy. High energy mechanism trauma was directly related to fracture comminution at peak bone age.


K. Yeung

Bone allograft is the most widely accepted approach in treating patients suffering from large segmental bone defect regardless of the advancement of synthetic bone substitutes. However, the long-term complications of allograft application in term of delayed union and nonunion were reported due to the stringent sterilization process. Our previous studies demonstrated that the incorporation of magnesium ions (Mg2+) into biomaterials could significantly promote the gene up-regulation of osteoblasts and new bone formation in animal model. Hence, our group has proposed to establish an Mg2+ enriched tissue microenvironment onto bone allograft so as to enhance the bone healing. The decellularization and gamma irradiation process were performed on bovine bone allograft and followed by magnesium plasma treatment. To evaluate the biocompatibility and bioactivity, materials characterizations, in vitro and in vivo studies were conducted, respectively. Mg composite layer on bone surface ranged from 500nm to ∼800nm thick. The cell viability on magnesium enriched allograft was significantly higher than that of the control. The ALP gene expression of hTMSCs in the group of PIII&D treated samples was highly up-regulated. The bone regeneration ability of Mg modified bone allograft implanted in animal model was significantly superior than the control after 2-month post-operation.


S. Hofmann

Bone tissue engineering has the intent to grow bone copies in the laboratory that could be used either for bone regeneration or as model systems to study bone physiology and pathology. Bone marrow- or adipose derived derived mesenchymal stromal cells are commonly used as they have been shown to be capable to differentiate into osteoblasts and depositing a calcium phosphate rich extracellular matrix. However, real bone is more than that: there are commonly three cell types described that are essential contributors to the tissue's native function: osteoblasts, osteocytes and osteoclasts. While all three cell types are being investigated separately, co-cultures of them including their precursors and inactive forms still provide a huge challenge these days, both in terms of culturing and (quantitative) evaluation. In addition, the matrix deposited by the osteoblasts in vitro is still far from bone's hierarchical organization in vivo that contributes to bone's impressive mechanical properties. Using a large set of microscopic tools (micro-computed tomography, SEM, 3D FIB/SEM, TEM and fluorescence), combined with spectroscopic (FTIR) and molecular tools (qPCR) we show that our 3D model system develops the main features of bone by human stromal cells differentiating first into osteoblasts who further embed themselves to become osteocytes. In their right environment and when stimulated mechanically, the cells are embedded within a collagenous matrix which is mineralized with carbonated hydroxyapatite. While this system still needs the addition of osteoclasts to represent ‘real’ bone, it allows to study the interaction between osteoblasts and osteocytes and to invest parameters contributing to collagen mineralization in high resolution and cryogenic conditions.


D.M. Devine J.S. Hayes D. Kotsougiani C.H. Evans

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.


K. Stuckensen J.M. Lamo-Espinosa E. Muiños-López P. Ripalda-Cemboráin T. López-Martínez I. Andreu R. Elizalde F. Prósper J. Groll F. Granero-Moltó

In the treatment of bone non-unions an alternative to bone autografts is the use of bone morphogenetic proteins (BMP-2, BMP-7) with powerful osteoinductive and osteogenic properties. In clinical settings, BMPs are applied using absorbable collagen sponges. Supraphysiological doses are needed and major side effects may occur as induce ectopic bone formation, chronic inflammation and excessive bone resorption. In order to increase the efficiency of the delivered for BMPs we designed cryostructured collagen scaffolds functionalized with hydroxyapatite, mimicking the structure of cortical bone (aligned porosity, anisotropic, ANI) or trabecular bone (random distributed porosity, isotropic, ISO). We hypothesize that anisotropic structure would enhance osteoconductive properties of the scaffolds increasing rhBMP-2 regenerative properties. In vitro, both scaffolds presented similar mechanical properties, rhBMP-2 retention and delivery capacity. For in vivo testing, a rat femoral critical size defect model was created. Four groups were assessed depending on the implant applied to the bone defect: ISO, unloaded isotropic sponge; ISO-BMP, isotropic sponge loaded with 5 μg of hrBMP-2; ANI, unloaded anisotropic sponge; and ANI-BMP, anisotropic sponge loaded with 5 μg of hrBMP-2. Regeneration was allowed for 10 weeks. X-ray, μCT, biomechanical testing and histology were used to evaluate repair. Independently of their structure, sponges loaded with rhBMP-2 demonstrate increased bone volume, and biomechanical properties than their controls (p<0.01 and p<0.05 respectively). Globally, ANI-BMP group demonstrated better bone regeneration outputs with increased defect bridging (p<0.05 when compared ANI-BMP vs ISO-BMP groups). In conclusion, anisotropic cryostructured collagen scaffolds improve the efficiency of rhBMP-2 in bone regeneration.


L.M. O'Sullivan H. Allison J. Schiavi K. Spanoudes E.E. Parle L.M. McNamara

Although osteoporosis reduces overall bone mass causing bone fragility, our recent studies have shown that bone tissue composition is altered at the microscopic level, which is undetectable by conventional diagnostic techniques (DEXA) but may contribute to bone fracture. However, the time sequence of changes in bone microarchitecture, mechanical environment and mineral distribution are not yet fully understood. This study quantified the longitudinal effects of estrogen deficiency on the trabecular microarchitecture and mineral distribution in the tibia of Female Wistar rats (6 months) that underwent ovariectomy (OVX, n=10) or sham surgery (SHAM, n=10). Weekly micro-CT scans of the proximal tibia were conducted at 15µm resolution for the first month of estrogen deficiency. Morphometric analysis was conducted to characterise the trabecular bone microarchitecture. The bone mineral composition was characterised with analysis of bone mineral density distributions (BMDD). There was significantly reduced trabecular bone volume fraction at 2 weeks in OVX rats compared to controls (p<0.01). There was no difference in mineral distribution between the OVX and control animals. This study provides the first evidence in uncovering the temporal nature of changes in bone microarchitecture and mineral distribution, showing that structure changes before composition. In-vivo µCT analysis for later time points (week 8, 14 and 34) is ongoing to comprehensively examine these longitudinal compositional changes. Moreover, we are conducting ex-vivo mechanical analysis (nanoindentation), and together these will uncover the time-sequence and respective contribution of changes in bone mass and composition to the integrity of the bone tissue at these stages of estrogen deficiency.


S. Negri D. Regis A. Sandri B. Magnan

Introduction. The management of periprosthetic pelvic bone loss is a challenging problem in hip revision surgery. This study evaluates the minimum 10-year clinical and radiographic outcome of major column structural allografts combined with the Burch-Schneider antiprotrusio cage for acetabular reconstruction. Methods. From January 1992 to August 2005, 106 hips with periprosthetic osteolysis underwent acetabular revision using massive allografts and the Burch-Schneider antiprotrusio cage. Forty-five patients (49 hips) died for unrelated causes without further surgery. Fifty-nine hips in 59 patients underwent clinical and radiographic evaluation at an average follow-up of 15.1 years. There were 17 male and 42 female patients, with age ranging from 29 to 83 years (mean 59). Results. Ten hips required rerevision because of infection (3), aseptic loosening (6), and flange breakage (1). Moreover, 4 cages showed x-ray signs of instability with severe bone resorption. The survivorship of the Burch-Schneider cage at 21.9 years with removal for any reason or radiographic migration and aseptic or radiographic failure as the end points were 76.3 and 81.4, respectively. The average Harris hip score improved from 33.2 points preoperatively to 75.7 points at the latest follow-up (p < 0.001). Discussion. In hip revision surgery, severe deficiency of pelvic bone stock is a critical concern because of the difficulty in providing a stable and durable fixation of the prosthesis. Although antiprotrusio cages have a limited role in acetabular revision, the use in association with massive allografts in extended bone loss demonstrated highly successful long-term results, enabling bone stock restoration and cup stability.


V. Glatt

The management of bone defects and impaired fracture healing remains one of the most challenging clinical problems. Several treatments exist to aid in the healing of large bone defects, including biologics such as recombinant human bone morphogenetic protein-2 (BMP-2), yet all have met with limited success. Regeneration of bone requires a coordinated network of molecular signals where the local mechanical environment plays a major role in the success of the healing process. The mechanical environment itself is determined by the stiffness of the implant used to stabilize the fracture and weight-bearing, and if fixation is either too flexible or too rigid the healing might fail. The hypothesis is that the healing of large-segmental bone defects and fractures can be accelerated by the imposition of an appropriate mechanical environment. An overview of the progress made in this research area on how the amount of rhBMP-2 could be reduced and its effectiveness increased by providing an optimized mechanical environment to achieve bone union will be presented. Additionally, the latest findings of improved fracture healing through the manipulation of fixation stability introducing a potential clinical strategy to improve the healing outcome of unstable fractures, particularly for non-unions through increased stabilization, will be discussed.


M. J. Stoddart

The unique properties of mesenchymal stem cells (MSCs) and their natural presence within the bone marrow make them an attractive source of cells for novel cartilage repair strategies. As mechanics play a critical role in vivo, a more physiological loading regime in vitro would be more appropriate to test novel therapies, and this can be achieved using bioreactors. Using a multiaxial load bioreactor system, we have investigated the effect of mechanical stimulation on human stem cell differentiation in the absence of growth factors, specifically transforming growth factor β (TGFβ). Our bioreactor system allows for the application of shear, compression or a combination of both stimuli to establish the phenotypic changes induced within MSCs. Neither compression alone, nor shear alone induces a change in MSC phenotype with a fibrin-based scaffold. However, we have demonstrated that a combination of compression and shear is able to induce chondrogenic differentiation and this is due to increased endogenous expression and activation of TGFβ. Using this multiaxial load bioreactor system, we can search for novel markers and potential therapeutic targets that only occur under physiological loads. In addition, potential rehabilitation protocols to be used after cell therapy in cartilage repair can also be investigated.


M. Riffault G.P. Johnson D.A. Hoey

Bone tissue experiences continued remodelling in response to changes in its biochemical and biophysical environment. Given the finite lifespan of osteoblasts, this continued bone formation requires replenishment from a progenitor population. Although this is largely believed to be from a skeletal stem cell population, given the limitation in in-vivo markers for this cell type, progress in demonstrating this mechanism is limited. Therefore, we characterized the LepR-Cre mouse strain and evaluated whether LepR positive cells are the progenitor population and if they contribute to the osteoblast population over time and in mechanically-induced bone formation in-vivo. Transgenic mouse strains; B6.129(Cg)-Leprtm2(cre)Rck/J to study LepR-expressing cells and B6.Cg-Gt(ROSA)26Sortm9(CAG-tdTomato)Hze/J as a reporter strain were obtained from Jackson Laboratories. Characterization studies were performed on LepR:tdTomato mice at embryonic stage (19.5dpc), 8 and 12 weeks old. Mice (12 weeks old) were subjected to compressive tibia loading with a 11N peak load for 40 cycles, every other day for 2 weeks. Histological analysis reveal that LepR is expressed from the embryonic stage in various organs including bones. LepR positive cells are found around blood vessels and on bone surfaces. Flow cytometry analysis show the amount of LepR positive cells negative for CD45 and Ter-119 markers inside the bone marrow increases over time and following tibial loading. Mechanical loading induces an increase in bone mass and bone parameters. This model allows us to track and evaluate the role of LepR positive cells as bone forming cells, and to decipher the role of these cells in mechanically-induced bone formation.


K. Moeller R. Gottardi S. Font Tellado R.S. Tuan M. van Griensven E. R. Balmayor

After surgical tendon repair, the tendon-to-bone enthesis often doesn't regenerate, which leads to high numbers of rupture recurrences. To remedy this, tissue engineering techniques are being pursued to strengthen the interface and improve regeneration. In this study, we used biphasic 3D printed PLGA scaffolds with aligned pores at the tendon side and random pores at the bone side to mimic the native enthesis. We seeded these with mesenchymal stem cells and inserted them into dual-flow bioreactors, allowing us to employ tenogenic and chondrogenic differentiation medium in separate flows. MTS assay demonstrated metabolism in dual-flow bioreactors at levels similar to tissue culture plate and rotating bioreactors. After 7, 14 and 21 days, samples were collected and analyzed by histology, RT-PCR and GAG production. H&E staining confirmed a compact cell layer attached to fibers and between porous cavities of scaffolds that increased with time of culture. Interestingly, cultured constructs in dual-flow bioreactors biased towards a chondrogenic fate regardless of which flow they were exposed to, possibly due to high porosity of the scaffold allowing for fluid mixture. Sox9 was upregulated at all timepoints (up to 30× compared to control), and by day 21 Col2A1 was also highly upregulated. Additionally, GAG production in treated constructs (serum-free) was able to match constructs exposed to 10% FBS in controls, demonstrating the functional matrix forming capabilities of this system. Overall, we have validated this dual-flow system as a potential platform to form the enthesis, and future studies will further optimize parameters to achieve distinctly biphasic constructs.


D. Gaspar D. I. Zeugolis

Current cell-based tissue engineering strategies have limited clinical applicability due to the need for large cell numbers and prolonged culture periods that lead to phenotypic drift. In vitro microenvironmental modulators have been proposed to mimic the native tendon. Standard in vitro culture conditions result in delayed extracellular matrix (ECM) deposition, impairing the development of scaffold-free approaches. ECM deposition can be enhanced by macromolecular crowding (MMC), a biophysical phenomenon that governs the milieu of multicellular organisms. We assessed a multifactorial biophysical approach, using MMC and mechanical loading, on different cell sources to determine their suitability for in vitro fabrication of tendon-like tissue. Human dermal fibroblasts (DFs), tenocytes (TCs) and bone marrow mesenchymal stem cells (BMSCs) were cultured with MMC under static and uniaxial strain culture conditions. TCs and DFs exhibited alignment perpendicular to the load, whilst BMSCs did not show preferential alignment. When MMC was used, DFs and BMSCs showed increased deposition of collagen I, the main component in tendon ECM. DFs presented ECM composition similar to TCs with collagen types III, V and VI present. Gene expression analysis revealed upregulation of tenogenic markers by TCs and DFs, such as scleraxis and thrombospondin-4, under both loading and MMC. The combined use of MMC and mechanical stimulation is suitable for TCs phenotype maintenance and can modulate the phenotype of DFs and BMSCs differentially. This study provides insight into response of different cell sources to biophysical cues and contributes to further development of cell therapies for tendon repair and regeneration.


T. Wang A. Wagner C. Thien R. Gehwolf N. Kunkel H. Tempfer Q. Jiang A. Traweger M. Zheng

Mechanical loading plays an essential role in both tendon development and degradation. However, the underlying mechanism of how tendons sense and response to mechanical loading remains largely unknown. SPARC, a multifunctional extracellular matrix glycoprotein, modulates cell extracellular matrix contact, cell-cell interaction, ECM deposition and cell migration. Adult mice with SPARC deficiency exhibited hypoplastic tendons in load-bearing zone. By investigating tendon maturation in different stages, we found that hypoplastic tendons developed at around postnatal 3 weeks when the mice became actively mobile. The in vitro experiments on primary tendon derived stem cells demonstrated that mechanical loading induced SPARC production and AKT/S6K signalling activation, which was disrupted by deleting SPARC causing reduced collagen type I production, suggesting that mechanical loading was harmful to tendon homeostasis without SPARC. In vivo treadmill training further confirmed that increased loading led to reduced Achilles tendon size and eventually caused tendon rupture in SPARC-/− mice, whereas no abnormality was seen in WT mice after training. We then investigate whether paralysing the hindlimb of SPARC-/− mice using BOTOX from postnatal 2 weeks to 5 weeks would delay the hypoplastic tendon development. Increased patellar tendon thickness was shown in SPARC-/− mice by reducing mechanical loading, whereas opposite effect was seen in WT mice. Finally, we identified a higher prevalence of a missense SNP in the SPARC gene in patients who suffered from a rotator cuff tear. In conclusion, SPARC is a mechano-sensor that regulates tendon development and homeostasis.


J. Dudhia

Tendon injuries in both the human and horse represent a challenge due to persistent inflammation combined with inadequate reparative cells and a poorly organised extracellular matrix. The potential of mesenchymal stem cells (MSCs) in regenerating tendon injuries remains to be fully realised. The main mechanism of action by MSCs is considered to be primarily mediated via paracrine mechanisms. This may involve the production and release of extracellular vesicles (EVs) by stem cells with a sub-fraction of these EVs (<100 nm diameter) called exosomes that appear to be the main paracrine effectors. EVs can be readily prepared from MSCs and offer a clinically relevant therapy. However, EVs for tendon repair need to be fully characterised. The horse represents a highly relevant model of tendon and ligament injuries as it shares many features of mechanical loading, function and aetiopathology with the human. We have isolated and characterised EVs from equine MSCs for modulating tendon cell phenotype in an in vitro tendon injury model using IL-1ß. EVs can be isolated from IL-1ß stimulated MSCs although their levels are not significantly increased over controls suggesting that the nature of the stimulated EV cargo may be more important than absolute levels of released EVs.


M. E. Gomes

Significant challenges remain to accomplishing the development of fully functional tendon tissue substitutes that can lead to clinically effective and successful applications. Scaffolding materials must meet demanding requirements such i) mimic the hierarchical and anisotropically aligned structure of tendon tissues from the nano- up to the macroscale, ii) meet tendon mechanical requirements and non-linear biomechanical behaviour, iii) provide the necessary biophysical/biochemical cues and mechanical responsiveness to induce the tenogenic differentiation of stem cells and potentiating the effects of biochemical supplementation. On the other side, tenogenic differentiation of stem cells is still to be established, as well as the role of such cells (either naïve or pre-differentiated) in promoting tissue regeneration. We have recently found evidences that magnetic actuation can provide means of mechanically stimulating cells in a contact-free manner and, more interestingly, can also modulate inflammatory response, a critical issue for achieving tissue regeneration instead of repair. In summary, synergies of scaffold design and magnetic responsiveness can impact significantly cells behaviour as well as in vivo response and thus widen the therapeutically range of cell-laden tissue engineered constructs in tendon regeneration.


D. Berdecka P.S. Babo R.L. Reis M.E. Gomes

Tendon and ligament injuries represent highly prevalent and unmet clinical challenge that may significantly benefit from tissue engineering therapeutic strategies, once optimal cell source and biomolecules regulating tendon homeostasis are properly defined. Herein, we aimed to evaluate the expression of tendon/ligament markers in two novel cell populations, namely human dental pulp stem cells (DPSCs) and periodontal ligament cells (PDLCs), in response to supplementation with TGF-β ligands relevant for tendon development and healing, as well as under standard tri-lineage differentiation conditions. DPSCs and PDLCs were isolated from sound human permanent molars removed for orthodontic reasons. Pulp tissue and periodontal ligament were minced and digested with collagenase (3mg/mL) and cells were expanded in α-MEM supplemented with 10% fetal bovine serum (basal medium). To evaluate the susceptibility of DPSCs and PDLCs to tenogenic induction, cells were seeded at density of 1000 cells/cm2 and cultured up to 21 days in basal medium or media supplemented with TGF-β3 (10ng/ml), or GDF-5 (50 ng/ml). Cell response was evaluated weakly by analysis of expression of tendon, bone and cartilage markers, employing real time RT-PCR and immunocytochemistry. A significant increase in collagen I and collagen III expression was observed with the culture progression in all conditions, with abundant matrix being deposited by day 14. A significant upregulation of scleraxis expression was demonstrated in response to supplementation with TGF-β3 in both cell populations, when compared to basal medium and medium with GDF-5. It was concluded that TGF-β3 may represent an effective inducer of stem cell tenogenic differentiation.


A. Djalali-Cuevas I. Skoufos A. Tzora N. Prassinos N. Diakakis D. I. Zeugolis

RNA-Seq or whole transcriptome shotgun sequencing has been adopted in the last years as a reference technique to determine the presence and the quantity of different species of RNA in determined biological samples, thanks to it allows the identification every single RNA species transcribed from a reference genome. Meta-profiling takes advantage of the public availability of an increasing set of RNA-Seq data produced by different laboratories to summarize the expression levels of the different RNA species of many samples according to their biological context, giving the opportunity to perform comparisons on the gene expression profiles of different tissues by integrating data derived from a high number of studies. By using Genevestigator™; a platform which integrates RNA-Seq data into meta-profiles, we have performed a comparison between the gene expression profiles of bone, cartilage, muscle tendon and skin by means of interrogating its database with different gene sets and families with relevance to the function of the tissues of the musculoskeletal system. The collagen gene family and genes coding for proteoglycans, matrix metalloproteinases and tissue inhibitors of metalloproteinases, mechanotransduction-related proteins and signalling pathways involved in tissue development and differentiation have been analysed. Hierarchical clustering for every gene set was performed for the understanding the differences and similarities between the different tissues included in the analyses. The results of this study will help to improve our understanding of the musculoskeletal system, and will help to identify new biomarkers and signalling pathways of specific relevance for the bone, cartilage, muscle and tendon.


O. K. Nwawka B. Lin B. Toresdahl A. Allen M. Drakos

This study of collegiate basketball players evaluated change over time (COT) in ultrasound shear wave (SW) elastography metrics across the basketball season, and correlated to morphologic changes on conventional ultrasound imaging, and VISA-P scores. In eleven male collegiate basketball players (mean age 19, age range 18–21), patella tendon (PT) ultrasound and SW elastography of both knees were performed at pre-season and post-season time points, and players reported their VISA-P scores throughout the season. Patella tendinopathy grade and SW metrics were correlated to VISA-P scores using Spearman correlation coefficients. Paired t-test was used to assess differences in mean SW metrics at pre-and post-season timepoints, accounting for leg dominance. 6 of 11 players (54.5%) had baseline patella tendinopathy on ultrasound progressing in 4 players. The mean change in VISA-P score was 15.18 (+/−8.55). No significant correlation was seen between ultrasound grades of tendinopathy and VISA-P. Pre-season SW velocities did not significantly correlate with baseline VISA-P scores. Post-season SW values and SW COT demonstrated strong correlation with change in VISA-P score in dominant and non-dominant knees. Although not statistically significant, there was a trend towards higher SW velocity for tendinopathy in both dominant and non-dominant knees at both study visits. SW metrics of the PT correlated to change in VISA-P scores in the dominant and non-dominant knees, whereas conventional ultrasound grades of patella tendinopathy did not. There was a trend towards higher SW velocities in patella tendinopathy which may indicate detection of change in intrinsic tissue stiffness.


L. Mercer D. Mercer R. Mercer M. Moneim L. Benjey E. Kamermans C. Salas

We hypothesized that the finger extensor mechanism has attachments along the dorsal surface of the entire length of the proximal phalanx and that this anatomy has not been clearly defined. The attachment along the dorsal aspect of the proximal phalanx of the index, middle, index and small fingers was dissected in 20 fresh-frozen cadavers. The lateral bands and attachments along the lateral and medial surface were released to appreciate the attachments along the dorsal aspect. We characterized the ligament attachments as very robust, moderately robust, and minimally robust at the distal, middle, and proximal portions. Three orthopaedic surgeons quantified the attachment, finding that 93% of specimens had tendinous attachments and the most robust attachment found at the most proximal and distal aspects adjacent to the articular cartilage. 87% of the specimens had very robust attachments at the proximal portion of the proximal phalanx. The middle portion of the proximal phalanx had moderate to minimally robust attachments. Greatest variability in attachment was found along the most distal portion of proximal phalanx adjacent to the proximal interphalangeal joint (26% of specimens had moderate to minimal robust attachment; 74% had robust attachments). The attachments along the proximal phalanx were attached on the dorsal half of the proximal phalanx, with no fibrous attachments extending past the lateral bands. In summary, we found tendinous attachment along the proximal phalanx that may assist in finger extension and may extend the digit at the metacarpal phalangeal joint without central band contribution.


J. Orbay

Advancements in treating the unstable elbow. We will review and discuss the kinematics and biomechanics of the forearm, concentrating on the role of soft tissue structures and how they affect forearm and elbow function. During this session, we will review the latest techniques for treating the terrible triad, including solutions to complex injuries of the olecranon, coronoid, and radial head. Techniques presented will address fixation, reconstruction, and salvaging of complex unstable elbow injuries.


N. Hoekzema

Advancements in treating distal humerus fractures. We will review and discuss approaches to the elbow to treat different types of fractures. We will discuss the role of soft tissue structures and how they affect elbow function. During this session, we will review the latest techniques for treating the complex articular fractures of the distal humerus to include capitellar and trochlear fractures. Techniques presented will address fixation, reconstruction, and salvaging of complex distal humerus fractures.


K. R. Jethwa A. Abdelhaq A. P. Sanghrajka

Supracondylar fractures of the humerus (SCFH) are the most common type of paediatric elbow fractures. Due to beliefs that non-operatively managed SCFH may displace further from the original position, they are monitored with repeated radiographs and a large number are unnecessarily surgically pinned. Very limited evidence currently exists to support these beliefs. This study aimed to determine the incidence of late “significant” displacement (requiring surgical management) of non-operatively managed paediatric SCFH, and whether they necessitate close radiographic follow-up. Patients aged ≤16, with a SCFH, were included in this retrospective cohort study. All were initially managed non-operatively with at least one follow-up radiograph within six weeks of injury. Data from four consecutive years (2013–2016) was collected using the hospital's radiology database. Two observers independently analysed patient radiographs and classified fractures by the Gartland and AO systems. The incidence of late displacement was determined using follow-up radiographs and clinic notes. Of the 164 patients included in the study, one patient (Gartland Type II, AO Type III) suffered late displacement at two weeks, requiring surgical fixation. One further patient (AO Type II) had a persistent cubitus varus deformity (Baumann's angle 90°), with no long-term functional deficit. Incidence of late displacement was 0.6% (n=1). Our findings suggest that stable Gartland Type I/AO Type I and II fractures do not require repeated radiographic follow-up. However, some Gartland Type II/AO Type III fractures require monitoring. This could considerably reduce the financial costs for the healthcare system, and inconvenience to families, associated with repeated follow-ups.


A. Juhdi A. Abdulkarim P. Harrington

The treatment of massive chronic tears is problematic. The re-tear rate following surgery for extensive cuff tears remains high, and there is little consensus regarding optimum treatment. To investigate the outcome of a cohort of patients who had open repair of an extensive cuff tear using the Leeds Kuff patch as an augment. A retrospective cohort study of consecutive patients with a massive cuff tear who had surgery in our regional elective orthopaedic centre over a two year period from January 2015 to Dec 2016. All patients followed identical rehabilitation protocols, supervised by physiotherapists with an interest in the shoulder. Outcomes assessment was undertaken at a minimum of 12 months by a registrar or physiotherapist who was not part of the treating team. Pre-op data collection included; range of motion, pain score, Oxford shoulder score (OSS), assessment of muscle atrophy on MRI. Data collection was completed in 15 patients. The mean age was 62 yrs (56 – 75). The mean pre-op OSS was 22, improving to a mean of 43. The range of motion and pain score improved. There were no intra-operative complications. One patient required a second surgery for evacuation of a haematoma at 10 days post op. One patient had an obvious re-tear at 4 months. Open rotator cuff repair with synthetic Kuff patch augmentation for chronic degenerative tears appears worthwhile when assessed at 12 months and they continuous to improve even at 18 months. This treatment method may be a useful option for patients > 70 years old.


T.C. Tsai W.S. Lian F.S. Wang J.Y. Ko

Subacromial bursa fibrosis are linked to rotator cuff lesion with shoulder stiffness; however, the mechanism underlying this shoulder disorder remain elusive. MicroRNA-29s (miR-29s) are emerging fibrosis inhibitor targeting fibrogenic matrices during tissue fibrosis. This study is aimed to investigate clinical relevance and function of miR-29 signalling to subacromial bursa homeostasis in shoulder stiffness. Subacromial bursa in patients with rotator cuff lesion with or without shoulder stiffness who required open acromioplasty were harvested for assessing fibrosis histopathology using Manson's trichrome staining. Expressions of proinflammatory cytokines, fibrotic matrices, and miR-29s were quantified using RT-PCR and in situ hybridization. Range of motion and pain scores of the stiffness group were higher than those of non-stiffness group. Upregulated proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and fibrotic matrices (collagen 1α1, 3α1, and 4α1) but decreased miR-29a and b expression existed in the stiffness group. Affected tissues exhibited severe fibrotic matrix accumulation, synovial hyperangiogenesis, hyperplasia, and strong miR-29a transcripts. In vitro, IL-1β rather than IL-6 and TNF-α decreased miR-29a expression of subacromial bursa fibroblasts. miR-29a knockdown escalated fibrotic matrix expression, whereas forced miR-29a expression alleviated the IL-1β-induced fibrotic matrix expression. Of interest, miR-29a transgenic mice displayed moderate responses to supraspinatus and infraspinatus tenotomy-induce fibrosis and gait irregularity of affected shoulders. Weak miR-29 signalling causes excessive fibrosis and remodelling in subacromial bursa and ultimately increases the prevalence of shoulder stiffness. This study reveals a new mechanistic underlying shoulder stiffness and highlights that sustained miR-29a potentially ameliorates the severity and function of stiff shoulder.


M. Zhu A. Thambyah D. Tuari K. Callon M.L. Tay D. Patel B. Coleman J. Cornish D.S. Musson

Surgical repair of rotator cuff tears have high failure rates (20–70%), often due to a lack of biological healing. Augmenting repairs with extracellular matrix-based scaffolds is a common option for surgeons, although to date, no commercially available product has proven to be effective. In this study, a novel collagen scaffold was assessed for its efficacy in augmenting rotator cuff repair. The collagen scaffold was assessed in vitro for cytocompatability and retention of tenocyte phenotype using alamarBLUE assays, confocal imaging and real-time PCR. Immunogenicity was assessed in vitro by the activation of pre-macrophage cells. In vivo, using a modified rat rotator cuff defect model, supraspinatus tendon repairs were carried out in 46 animals. Overlay augmentation with the collagen scaffold was compared to unaugmented repairs. At 6- and 12-weeks post-op the repairs were tested biomechanically to evaluate repair strength, and histologically for quality of healing. The collagen scaffold supported human tenocyte growth in vitro, with cells appearing morphologically tenocytic and expressing higher tendon gene markers compared to plastic controls. No immunogenic responses were provoked compared to suture material control. In vivo, augmentation with the scaffold improved the histological scores at 12 weeks (8.37/15 vs. 6.43/15, p=0.0317). However, no significant difference was detected on mechanical testing. While the collagen scaffold improved the quality of healing of the tendon, a meaningful increase in biomechanical strength was not achieved. This is likely due to its inability to affect the bone-tendon junction. Future materials/orthobiologics must target both the repaired tendon and the regenerating bone-tendon junction.


R. Cadossi

The purine nucleoside, adenosine regulates functions in every tissue and organ in the body acting via four G-protein-coupled receptors, A1, A2A, A2B, and A3 adenosine receptors (ARs). Electromagnetic field (EMF) stimulation is an innovative therapeutic technique able to increase cellular anabolic activity and limit the catabolic effects of inflammatory cytokines. The mechanisms of cell reception of EMFs are not well known and much research activity has focused on the interactions between EMFs and membrane receptors. Interestingly, links have been found between ARs and their modulation by such physical agents as pulsed EMFs. EMF exposure mediates a significant upregulation of A2A and A3ARs in chondrocytes, synoviocytes and osteoblasts, leading to the reduction of synthesis and release of pro-inflammatory cytokines. In cultured full-thickness cartilage explants, pulsed EMFs preserve the integrity of the extracellular matrix and antagonize the effect of catabolic cytokines, such as IL-1. Pulsed EMFs, through the increase of ARs, enhance the working efficiency of adenosine without the side effects, desensitization, and receptor down-regulation often related to the use of agonist drugs. Modulation of adenosine receptors by pulsed EMFs could be a mechanism of cell reception of EMFs and an innovative physiologic alternative to the use of adenosine agonists.


M. Thio

Electrospinning of (bio)polymers is a well acknowledged technology used by scientists all over the world to manufacture scaffolds for tissue engineering & 3D cell culture purposes. The ability to control key parameters such as fibre diameter and fibre orientation allow the generation of highly specific scaffolds that closely mimic the native extracellular matrix. Despite the popularity in the R&D lab, the technology itself has only recently seen acceptance as a method for manufacturing clinical-grade medical devices. Subsequently, never before have more electrospun materials obtained market approval (FDA/CE) and are in clinical trials. In this presentation, we share our experience as a manufacturer of clinical-grade medical devices via electrospinning and give insight into the possible applications in orthopaedics.


J. Racine

Exposure to electromagnetic energy has potent signalling effects upon articular cells including chondrocytes, synoviocytes and osteoblasts. Attention has focused on two actions – the altered synthesis of cytokines and enzymes, and the enhanced synthesis of bone and cartilage extracellular matrix (ECM) molecules. In vitro studies with human and bovine articular cartilage have shown increased aggregcan synthesis, glycosaminoglycan content, and biomechanical aggregate modulus with EMF exposure. Osteoarthritic (OA) cartilage responds similarly depending upon the severity of the OA with early OA cartilage responding more robustly. On these bases, two in vivo studies have been done with the Dunkin-Hartley guinea pig model of spontaneous OA. Both studies demonstrated preservation of ECM with increased aggrecan synthesis, matrix glycosaminoglycan and type 2 collagen content, and reduced histological-histochemical (Mankin) scores. Suppression of matrix metalloproteases and IL-1, together with increased TGFb were also observed. Responses to various EMF configurations, in terms of amplitude, frequency, and exposure duration have been described, indicating dose responsiveness. These studies suggest the conclusion that exposure to specific EMFs reduce the progression of early OA. A randomized clinical trial is underway. EMFs may be a disease-modifying therapy for OA, resulting in maintenance of ECM and improvement in the cytokine environment of OA joints.


F. F. Masieri J. R. Schofield C. P. Velloso J. Ballisat S. Setti R. Cadossi M. Abdel-Maguid

Pulsed Electromagnetic Fields (PEMFs) promote joint tissue anabolic activities, particularly in cartilage and bone. Here we investigated the effect of selected PEMFs (75Hz, 1.5mT, 1.3msec) in a differentiating model of murine myoblasts (C2C12) in vitro. C2C12 were seeded at 5×103 cells/cm2 in 4 well plates and left to adhere for 24h. Subsequently, cells were either maintained in growth medium (GM) or induced towards myogenic differentiation in low-serum conditions, with and without PEMF exposure, for 4 days. Morphological analysis, myotube formation and fusion index (FI) were assessed with fluorescence microscopy techniques. Metabolic activity was determined by MTT; moreover, a multiplex cytokine array (RayBiotech) allowed cell supernatant molecule quantification. Cells exposed to PEMFs in GM acquired a distinctive elongated morphology, with increased bi-nuclear figures (3.2-fold FI increase over PEMF-unexposed cells) and displayed a significantly higher metabolic activity (+31%, p<0.05 over PEMF-unexposed cells). PEMF exposure increased metabolic activity also under myogenic differentiation (+15% over PEMF-unexposed differentiating cells, p<0.05), with the formation of long, thick polynuclear myotubes, suggesting a role of PEMFs in enhancing myogenesis (7.7-fold FI increase over PEMF-unexposed cells). 4-day culture supernatants revealed the presence of several myokines (KC/CXCL1, LIX, MCP-1, TIMP-1). Preliminary analysis showed a 1.16-fold increase (n=2) of LIX and, notably, a 1.91-fold increase (n=2) of TNF-RI, in cell supernatants of PEMF-exposed over PEMF-unexposed cells. Collectively, these results suggest that PEMF may successfully be applied in models of muscle cell trauma to optimise muscle fibre repair, by fine-tuning the release of myokines, promoting myoblast proliferation and myotube formation.


K. F. Eichholz D. A. Hoey

The architecture within which cells reside is key to mediating their specific functions within the body. In this study, we use melt electrospinning writing (MEW), a recently developed 3D printing technology unique in its ability to generate ECM like fibres and control their deposition, to fabricate cell micro-environments with various fibrous architectures to study their effect on human stem cell behaviour. We designed, built and optimised a MEW apparatus and used it to fabricate four different platform designs of 10.4±2μm fibre diameter, with angles between fibres on adjacent layers of 90°, 45°, 10° and R (random). Characterisation was conducted via scanning electron microscopy (SEM) imaging and tensile testing, and human skeletal stem cells (hSSCs) were seeded to scaffolds to study the effect of architecture on cell morphology and mechanosensing. Cell morphology was significantly altered between groups, with cells on 90° scaffolds having a lower aspect ratio, greater spreading, greater cytoskeletal tension and nuclear YAP expression. Long term cell culture studies were then conducted to determine the differentiation potential of scaffolds in terms of alkaline phosphatase activity, collagen and mineral production. Across these studies, an increased cell spreading in 3-dimensions is seen, with decreasing alignment of architecture correlated with enhanced osteogenesis, as seen by significant fold increases in ALP (2.8), collagen (2.5) and calcium (3.6) in the 90° scaffold architecture compared to 10°. This study therefore highlights the critical role of fibrous architecture in regulating stem cell behaviour with implications for tissue engineering and disease progression.


A. Hammerl C. Diaz Cano E. De-Juan-Pardo M. van Griensven P.S.P. Poh

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.


T. Guise

Breast and other cancers commonly metastasize to bone to cause bone destruction, pain, fractures hypercalcemia and muscle weakness. Recently, we described a specific molecular mechanism by which bone-derived transforming growth factor (TGF)-beta, released as a consequence of tumor-induced bone destruction causes muscle dysfunction, before the loss of muscle mass. Circulating TGF-beta induces oxidation of the ryanodine receptor (RYR1) on the sarcoplasmic reticulum of skeletal muscle to induce calcium leak and muscle weakness. Blocking TGF-beta, or its release from bone (with bisphosphonates), preventing oxidation of or stabilizing RyR1 all prevented muscle weakness in mouse models of breast cancer bone metastases. In addition to these effects on skeletal muscle, circulating TGF-beta may act on beta cells of the pancreas to impair insulin secretion and result in glucose intolerance. These and other potential systemic effects of TGF-beta released from the tumor-bone microenvironment or from cancer treatment-induced bone destruction implicate bone as a major source of systemic effects of cancer and cancer treatment. Therapy to block the systemic effects of the bone microenvironment will improve morbidity associated with bone metastases and cancer treatment.


R. L. Reis

The selection of a proper material to be used as a scaffold or as a hydrogel to support, hold or encapsulate cells is both a critical and a difficult choice that will determine the success of failure of any tissue engineering and regenerative medicine (TERM) strategy. We believe that the use of natural origin polymers, including a wide range of marine origin materials, is the best option for many different approaches that allow for the regeneration of different tissues. In addition to the selection of appropriate material systems it is of outmost importance the development of processing methodologies that allow for the production of adequate scaffolds/matrices, in many cases incorporating bioactive/differentiation agents in their structures. An adequate cell source should be selected. In many cases efficient cell isolation, expansion and differentiation, and in many cases the selection of a specific sub-population, methodologies should be developed and optimized. We have been using different human cell sources namely: mesenchymal stem cells from bone marrow, mesenchymal stem cells from human adipose tissue, human cells from amniotic fluids and membranes and cells obtained from human umbilical cords. The development of dynamic ways to culture the cells and of distinct ways to stimulate their differentiation in 3D environments, as well as the use of nano-based systems to induce their differentiation and internalization into cells, is also a key part of some of the strategies that are being developed in our research group. The potential of each combination materials/cells, to be used to develop novel useful regeneration therapies will be discussed. The use of different cells and their interactions with different natural origin degradable scaffolds and smart hydrogels will be described. Several examples of TERM strategies to regenerate different types of musculoskeletal tissues will be presented. Relevance to orthopaedics will be highlighted.


G. Im

Osteoarthritis (OA) is the most common arthritis. Early OA is treated with pain-relieving medication while advanced diseases are treated with joint replacement. Intraarticular (IA) injection has been also used as a local therapy for OA. Only corticosteroids and hyaluronic acid has been clinically used for IA injection up to now. While these drugs are effective in alleviating pain relief and mitigating inflammation, they do not regenerate damaged cartilage. We have developed drug delivery system for OA treatment using a new molecule kartogenin which are known to have regenerative effects for cartilage. These systems include kartogenin-conjugated chitosan nano/microparticles, thermoresponsive nanospheres containing kartogenin and diclofenac, hyaluronic acid hydrogel containing PEGylated kartogenin micelles. We have found that injection of these systems arrested the progression of OA as well as inhibiting inflammation in surgically-induced OA model in rats. These data will be introduced in this talk.


G. McCarthy

Osteoarthritis (OA) is the most common cause of joint disease and associated disability. Despite this, its pathogenesis remains incompletely understood and no specific drug exists to prevent or reverse the structural changes in OA. Basic calcium phosphate (BCP) crystals are extremely common in OA. BCP crystals consist primarily of hydroxyapatite, with smaller amounts of octacalcium phosphate, tricalcium phosphate and magnesium whitlockite. They are present in 100% of joints at the time of knee and hip joint replacement surgery. Their presence strongly correlates with radiographic severity of osteoarthitis. In mice, intra-articular BCP crystals elicit synovial inflammation and cartilage degradation. The potential mechanisms by which calcium-containing crystals may promote articular damage have been studied in the laboratory setting and in vitro properties of BCP crystals have been observed that emphasise their pathogenic potential. BCP crystals interact with articular cells such as fibroblasts and chondrocytes to induce mitogenesis with resultant cellular proliferation likely leading to synovial lining hypertrophy. BCP crystals also upregulate production of cytokines such as tumour necrosis factor alpha (TNF-α), interleukin 1 (IL-1), increase prostaglandin E2 via the cyclooxygenase pathway, stimulate matrix metalloproteinases production and increase nitrous oxide production. Therefore, BCP crystals have potent biologic effects and represent a potential therapeutic target in OA.


N. Madhusudan U. Oppermann C. Bountra R.O.C. Oreffo M.C. de Andrés

Osteoarthritis (OA) is a leading cause of joint deformity and functional limitation. An imbalance of anabolic and catabolic activity results in destruction of the extracellular matrix of articular cartilage. There is evidence to support the role of DNA methylation in the pathogenesis of OA, but the effect of other epigenetic modifiers is yet to be described. This study looks at the effect of novel epigenetic modulators, PFI-1, a bromodomain inhibitor, and SGC707, a histone methytransferase inhibitor, and their effects on gene expression in the pathogenesis of OA. Chondrocytes were extracted from OA femoral heads (n=6), cultured and incubated. Samples were treated with media alone (control), interleukin 1-beta (IL-1β) plus oncostatin M (OSM) alone, or in combination with increasing concentrations of PFI-1 or SGC707. Levels of expression of iNOS, COX2, IL8, IL1B, matrix metalloproteinase-13 (MMP13), RUNX2 and COL9A1 were measured using qRT-PCR, and expressed relative to GAPDH. PFI-1 (0.5 and 5µM) suppressed expression of catabolic genes in OA chondrocytes, at basal levels and when co-stimulated with IL-1β+OSM. Catabolic gene expression decreased (iNOS, COX2, IL-8, IL-1β and MMP), and RUNX2 expression was also supressed. There was no effect on expression of the anabolic gene COL9A1. SGC707 (0.1 and 1µM) did not induce a reduction in expression of all the catabolic genes. This study has demonstrated that PFI-1 has a potent protective effect against cartilage degradation, by modulating the expression of catabolic genes in OA chondrocytes. This further validates the role of epigenetics in OA, with implications for therapeutic interventions in the future.


C. Dooley M. Murphy

Osteoarthritis (OA) is a debilitating joint disease that severely affects elderly populations. At present there are no effective treatments for OA and mechanisms of disease progression are poorly understood. Previous work has identified that neuronal-Interleukin-16 (nIL-16) was significantly up-regulated in cartilage during the later stages of OA. Preliminary investigations identified co-localisation of nIL-16 with the Transient Receptor Potential cation ion-channel sub-Family-V-member-4 (TRPV4) in the primary cilium and the pericellular matrix of human OA chondrocytes. Perturbation of both TRPV4 and cilia are strongly associated with OA. We hypothesised that nIL-16 and TRPV4 work in tandem in a pathway that leads to chondrocyte hypertrophy and calcification that is seen in late OA and contributes to the loss of joint integrity. This makes it a promising target for development of a gene therapy to combat the disease. With the aim of elucidating the mechanism involved, nIl-16 knock-out cell lines generated using the Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9 system will be used to knock out nIl-16 PDZ domains to investigate whether this is the mechanism in which nIL-16 functions to anchor TRPV4 to the membrane of chondrocytes at the primary cilium. This work will be carried out using an immortalized hTERT mesenchymal stromal cell (MSC) cell line and effects on terminal MSC chondrogenesis, where hypertrophy mimics the process of calcification seen in OA, will be used to define functional effects of the knockout. Cell lines will be made using the RALA peptide (Phion Therapeutics), a bioinspired nanoparticle, for delivery the CRISPR/Cas9 system.


L. Chen F. Yao C. James T. Wang J. Gao O Beaumont D. Wood M. Zheng

Osteoarthritis (OA) is traditionally believed to affect the osteochondral unit by wear-and-tear from the superficial zone to the deep zone of cartilage and extended to subchondral plate. Obesity is commonly considered as a risk of OA development and hence total knee replacement (TKR), but the mechanism remains unclear. We hypothesized that obesity accelerated OA development by deteriorating tidemarks and increasing bone remodelling. 616,495 cases of TKR for OA from Australia and British joint replacement registries were collected, and data indicated that patients with higher BMI had TKR at earlier age. Specifically, patients with BMI ≤25kg/m2 showed 8 years younger than patients with BMI ≥40kg/m2 (P<0.0001) when they received TKR. We next examined tibia plateaus of 88 knee OA patients by micro-CT and histomorphometry. Linear regression showed that less cartilage degradation was associated with increased BMI in the load-bear compartment (p<0.05), while 58.3% of patients with BMI≥40kg/m2 demonstrated a clear anatomical separation close to tidemarks filled with fibrosis, erythrocytes and bone fragments (compared to BMI ≤25kg/m2 group: 7.7%, p<0.01). In subchondral bone, elevated bone formation was associated with increased BMI, as higher thickness of osteoid (p<0.01), percent osteoid volume (p<0.01), percent osteoid surface (p<0.01) were found in obese patients. However, no alteration of bone resorption and microstructural parameters was found to be associated with BMI. We suspected that the abnormal loading in knee joint due to high BMI led to the direct deterioration of binding site of osteochondral unit, which might be the mechanism of the rapid progression in obesity-related OA.


S. Flynn M. O'Reilly I. Feeley E. Sheehan

Knee osteoarthritis is a common, debilitating condition. Intra articular corticosteroid injections are a commonly used non-operative treatment strategy. Intra articular hip injection with Ketorolac (an NSAID) has proven to be as efficacious as corticosteroids. No prior study compares the efficacy of Ketorolac relative to corticosteroids for relief of discomfort in knee osteoarthritis. The study design was a single centre double blinded RCT. Severity of osteoarthritic changes were graded on plain film weightbearing radiographs using the Kellgren and Lawrence system. Injection was with either 30mg Ketorolac or 40mg Methylprednisolone, given by intra-articular injection, in a syringe with 5mls 0.5% Marcaine. Pre-injection clinical outcomes were assessed using the Numerical Pain Score (NPS), WOMAC, and Oxford knee scores. Patients' NPS scores were assessed at Day 1 and Day 14 post-injection. An assessment of all clinical outcomes took place in clinic at six weeks. There were 72 participants (83 knees) in the study. No patients were lost to follow-up. Mean age was 62.66 years (Range 29–85). 42 knees received a corticosteroid injection, 41 a NSAID injection. Mean Kellgren and Lawrence score was 3.1. There was no significant difference in pre-injection clinical scores in either group. There was a significant improvement of NPS on Day 1 and 14 in both injection groups(p<0.05). These improved pain scores were sustained at 6 weeks in both groups. WOMAC and Oxford Knee Scores showed a statistically significant improvement in the corticosteroid group. WOMAC scores showed significant improvement in the NSAID group, however these improvements didn't achieve statistical significance using the Oxford Knee Score. Corticosteroid or NSAID injectate are a safe and effective non-operative treatment strategy in the patient with knee osteoarthritis. Ketorolac appears to provide effective medium-term improvement of pain and clinical scores. Further follow-up is recommended to investigate if this trend in sustained.


T. M. Grupp

Modular hip prostheses were introduced to optimize the intra-surgical adaptation of the implant design to the native anatomy und biomechanics of the hip. The downside of a modular implant design with an additional modular interface is the potential susceptibility to fretting, crevice corrosion and wear. For testing hip implants with proximal femoral modularity according to ISO & ASTM, sodium chloride solutions are frequently used to determine the fatigue strength and durability of the stem-neck connection. The present study illustrate that the expansion of standard requirements of biomechanical testing is necessary to simulate metal ion release as well as fretting and crevice corrosion by using alternative test fluids. To assess the primary stability of tibial plateaus in vitro, different approaches had been undergone: cement penetration depth analysis, static tension or compression loading until interface failure. However, these test conditions do not reflect the in vivo physiologic loading modes, where the tibial plateau is predominantly subjected to combined compression and shear forces. The objectives were to evaluate the impact of the tibial keel & stem length on the primary stability of a posterior-stabilised tibial plateau under dynamic compression-shear loading conditions in human tibiae.


N. Diakakis

The paramount importance of synovial fluid in lubrication and protection of articular joints has long been recognized. Synovial fluid, a dialysate of plasma, forms an interface with both the synovium and cartilage and plays a crucial role in joint lubrication and bearing functions. In an osteoarthritic joint, damage to the articular cartilage causes modifications in the rheological properties of synovial fluid and, reducing the viscoelasticity and increasing the friction between articular surfaces. Viscosupplementation is a treatment for osteoarthritis that uses hyaluronic acid as a (visco)supplement to the diseased joint. The aim of this treatment is to restore the rheological properties of synovial fluid. Osteoarthritis is the most common disease affecting the joints in human population and among the most important causes of pain, disability and economic loss. Therefore, innovative methods are needed to more effectively treat osteoarthritis, directly addressing the disease process. Among various locomotor mechanisms that could serve to illustrate the integrated nature of functional morphology, perhaps none is more complex than the equine locomotor system.

Confronting the need for evaluating the current methods to control joint disease, the horse provides an excellent animal model. As it suffers similar clinical manifestations to those seen in human, it may provide tentative biomedical extrapolations.


S. Di Paolo M. Wesseling M. Pastrama S. Van Rossom G. Valente I. Jonkers

In knee osteoarthritis (OA) patients, a focal cartilage defect is commonly found, especially in the medial compartment. In addition, cartilage softening is often observed at the defect rim. Both factors may alter the loading distribution and thereby the contact pressures, previously related to cartilage degeneration. To determine contact pressure in-vivo during motion, computational modelling can be used. The aim of this study was to analyse knee cartilage pressures during walking in healthy and damaged cartilage using a multi-scale modelling approach. Using 3D motion capture and musculoskeletal models, multi-body simulations of the stance phase of gait calculated knee kinematics and muscle, ligament and contact forces. These were subsequently imposed to a finite element (FE) model including tibial and femoral bones and cartilage. FE analyses were performed using intact cartilage as well as including a medial tibial cartilage defect, with and without softening of the defect rim. Specifically during loading response, a medial cartilage defect reduced the contact surface (−28%) and thereby increased the contact pressure (+33%) compared to intact cartilage, particularly on the medial compartment (+75% in contact pressure). Including softening of the cartilage rim increased the contact area (+22%) and decreased contact pressures (−9%) compared to the defect. This indicates that a focal defect increases the cartilage loading. This is partially compensated by softening of the cartilage rim. Therefore, the role of focal defects in altered cartilage loading and consequent OA development always needs to be discussed acknowledging the cartilage status at the defect rim.


T. Fujito T. Tomita T. Yamazaki M. Tamaki T. Ishibashi K. Kono S. Konda H. Yoshikawa K. Sugamoto

Our aim was to investigate whether it is possible to predict post-operative kinematics (Post-Ope) from intra-operative kinematics (Intra-Ope) after total knee arthroplasty. Our study were performed for 11 patients (14 knees) who underwent primary PS TKA using CT-based navigation system between Sept.2012 and Sept.2014. The mean subject age was 71.5 ± 5.5 years at the time of surgery. Intra-Ope was measured using the navigation system after implantation during passive full extension and flexion imposed by the surgeon. Under fluoroscopic surveillance, each patient was asked to perform sequential deep knee flexion under both non-weight bearing (NWB) and weight bearing (WB) conditions from full extension to maximum flexion. To estimate the spatial position and orientation, we used a 2- to 3- dimensional (2D3D) registration technique. Intra-Ope and Post-Ope had a common coordinate axis for bones. Evaluations were range of motion (ROM), external rotation angles (ER). The level of statistical significant difference was set at 0.05. Mean ROM in Intra-Ope(130°± 7.9°) was statistically larger than both NWB(121.1°±10.5°) and WB(124.0°±14.7°). No Statistically significant difference was found in the mean ER from 10° to 120° among Intra-Ope (11.2°± 8.5°) and NWB(7.1°±6.0°) and WB(5.3°±3.2°). It is suggested that we could predict Post-Ope from Intra-Ope by considering the increase of the range of motion due to the muscle relaxation condition and the amount of change in the ER.


M. T. A. Griffin A.H.R.W. Simpson D.F. Hamilton

The first three months following Total Knee Arthroplasty (TKA) provide an early window into a patient's functional outcomes, with the change of function in this time yielding valuable insight.

20 patients due to undergo primary TKA were recruited to the study. Data were recorded at three time points; pre-assessment clinic (PAC) before the operation, 6-weeks-post-operation (6WKs), at 12-weeks-post-operation (12WKs). Functional activity levels were monitored during early post-operative recovery for changes in early functional outcome, and allowed a comparison of metrics at each time point. This included direct functional testing of power output, timed functional performance in clinic, patient reported outcome measures, and multiday activity monitoring devices. Maximal power output symmetry (Power) was similar at 6WKs vs PAC (p = 0.37). At 12WKs, it had increased (p < 0.05). Timed functional performance (Performance) remained similar across all three time points (p = 0.27). Patient reported activities of daily living (ADL) performance significantly increased at 6WKs vs PAC (p < 0.05). At 12WKs, it remained similar (p = 0.10). Patient daily step count significantly decreased at 6WKs vs PAC (p < 0.05). By 12WKs, this had increased to similar levels to PAC (p = 0.30). Within the functional outcome measures, strong post-operative correlations were observed between Power and Performance (r = 0.62), Power and ADL (r = 0.49), and Performance and ADL (r = 0.61). Despite reduced measured step count and similar functional performance, patients report improved ADL at 6WKs. When symmetrical power output and measured step count have improved at 12WKs, patients report similar ADL to that at 6WKs. Multiple measures are required to get a full picture, however this highlights the different aspects measured by different tools.


S. Deo B. Lotz F. Thorne

The medical model of history, examination and investigation forms the bedrock of diagnosis and management of all patients. The essence is the recognition of patterns of symptoms and signs. In the modern era there are an increasing number of non-medical resources ranging from web-based information, computer diagnostic aids and non-specialist healthcare professionals to provide a diagnosis and commence management of a wide range of conditions, including knee problems. We analysed the quality and patterns of clinical presentation in order to answer the question how closely clinical symptoms and examination findings correlate to diagnosis based on MRI scan and/or arthroscopic findings. The analysis was a dataset of a consecutive series of patients, aged 18 to 45, with no past history of knee problems or end stage arthritis, presenting to a single specialist triage physiotherapist, working within an integrated knee service, who fully completed a standardised knee assessment proforma of presenting symptoms and signs at a large district general hospital. The study comprises 86 patients and 98 knees. We analysed this data based on diagnostic findings of MRI scan or arthroscopy to provide definitive intra-articular diagnosis. Based on standard textbook descriptions of common presentations, we went on to define the patients' presentation history and examination as typical or atypical, with typical meaning the symptoms and signs correlated with the diagnosis. The null hypothesis is that patients have a high chance of typical presentations for common knee conditions. In the 75% of patients with a significant intra-articular pathology we found the majority had chondral rather than meniscal tears 1.7 to 1. Forty four percent of patients had atypical symptoms and 71% had atypical clinical signs, 30% and only 26% of the cohort had both typical symptoms and signs together, reflecting a surprisingly low positive predictive probability of symptoms and signs in this group of patients, particularly those with chondral lesions which was 44%. In this cohort, 57% of the cohort has 3 or more multiple diagnoses. In the diagnostically normal group, 43% had symptoms and signs typical for a meniscal tear. We conclude that clinical symptoms and signs surprisingly inaccurate in guiding intra-articular pathology within the knee, even in a sub-set considered the easy and accurate to assess. The number of multiple diagnoses and the incidence of false positive results also means that simplistic interpretations of non-definitive diagnoses and linear causation of pain pathways should be treated cautiously.


A. Pandit

Biomaterials are no longer considered innate structures and using functionalisation strategies to modulate a desired response whether it is a host or implant is currently an important focus in current research paradigms. Fundamentally, a thorough understanding the host response will enable us to design proper functionalisation strategies. The input from the host response need to be weighed in depending on the host disease condition. In addition, biomaterials themselves provide immense therapeutic benefits which needs to be accounted for when using functionalisation strategies. Using strategies such as enzymatic and hyperbranched linking systems, we have been able to link biomolecules to different structural moieties. Our recent design efforts have harnessed the therapeutic effects of biomaterials and mapped the molecular fingerprint of this specific host response in a disease target. This approach allows us to rethink functionalisation strategies currently employed in the field. This talk will elucidate some of these ongoing strategies that have applications in the development of the next generation of orthopaedics devices.


D. Zhu

Biometals like Magnesium (Mg) and Zinc (Zn) are essential for life. Mg/Zn-deficiency has been linked to numerous diseases including cardiovascular, bone, diabetics, neurological and neurodegenerative disorders. Moreover, Mg/Zn-based biomaterials have recently emerged as innovative degradable medical implants, typically for cardiovascular and orthopedic application. We study the pathophysiological role of Mg2+/Zn2+ ion in vascular and bone diseases, as well as metallic Mg/Zn alloys for stent and bone implant applications. We demonstrated some interesting role and mechanism of Mg2+/Zn2+ ion in controlling cellular functions. Also, metallic Mg/Zn-based medical implants exhibited strong potential as stent and bone fixation device. They have sufficient mechanical strength, promotes tissue regeneration, and are fully bioresorbable with minimal toxicity. The beneficial or therapeutic role of biometals Mg/Zn in medicine and biomaterial applications is still not fully explored, our research aims to answer some fundamental questions and to inspire more future studies related to biometals in health.


A. Terzi E. Storelli S. Bettini T. Sibillano D. Altamura L. Salvatore M. Madaghiele A. Romano D. Siliqi M. Ladisa L. De Caro A. Quattrini L. Valli A. Sannino C. Giannini

The aim of this work was the structural investigation of different type I collagen isoforms at atomic and nanoscale, as well as the evaluation of the impact of different fabrication treatments on the structural, mechanical and biological properties of collagen-based films. Raw type-I collagens from bovine hide (Typ-BH, CS, SYM) and equine tendon (TypE, TypCH and OPO) were analyzed. Materials were then used for fabricating air-dried films, obtained by: 1) dissolution in distilled water (HH); 2) dissolution in acidic medium (AA); 3) homogenization of acid solubilized fibers (HOM). Crosslinking treatments (DHT, DHT+EDC) were also adopted and studied. Analysis by Wide Angle (WAXS) and Small Angle (SAXS) X-ray Scattering was carried out at the XMI L@b (CNR-IC-Bari); Fourier Transform-IR and biological analysis was performed at UniSalento. WAXS and SAXS data on raw materials demonstrated the preferential orientation of collagen molecules and the preservation of hierarchical nanoscale architecture in equine tendon-derived collagens, in particular in chemically extracted, while randomly oriented molecules were found in bovine dermis collagens, together with a certain degree of salt contamination. Concerning equine collagen, we found that TypCH structure is influenced by crosslinking procedures at atomic scale, whereas both processing conditions and crosslinking treatments affect TypE collagen structure at atomic and nanoscale. WAXS, SAXS and FT-IR analyses showed that the HOM processing was the one which ensures a high content of structural super-organization of collagen into triple helices and a high crystalline domainof the final material. Crosslinking of the films by DHT/EDC combined treatment was shown to affect their mechanical stiffness, the latter depending on the collagen source and the specific processing conditions.


L. Frapin J. Clouet N. Henry C. Chedeville J. Guicheux C. Le Visage

The recent description of progenitor/stem cells in degenerated intervertebral discs (IVDs) raised the possibility of harnessing their regenerative capacity for endogenous repair. The aim of this work is to develop an intradiscal polysaccharide microbead-based delivery system for the sequential release of chemokines and nucleopulpogenic factors. This delivery system would sequentially contribute to 1) the recruitment of resident progenitors (CXCL12 or CCL5), 2) the differentiation of the mobilized progenitors (TGF-β1 and GDF5), and 3) the subsequent regeneration of NP. To determine the effects of chemokines on in vitro cell recruitment, human mesenchymal stem cells (MSC) were cultured in Transwells for 4h, with or without CXCL12 or CCL5. In parallel, pullulan microbeads (PMBs) (100µm) were prepared by a simultaneous crosslinking protocol coupled to a water-in-oil emulsification process. Freeze-dried PMBs were loaded with biological factors then release assays were performed at 37°C for 21 days and supernatant concentrations were measured by ELISA. As compared to untreated MSC, MSC migration was improved with a 3.9 (CXCL12) and 7.5 (CCL5) fold increase, respectively. All factors were successfully adsorbed on PMBs and a burst release within the 1st day was observed. At day 7, 27.5% and 83% of CXCL12 and CCL5 were released, respectively and at day 21, 20% and 100% of TGF-β1 and GDF5 were released, respectively. Currently, released cytokine bioactivity is being analysed and an ex vivo ovine IVD model is developed to determine the repair potential of this controlled release approach.


J. Quintas Coentro D. I. Zeugolis

Complex pathophysiologies involve different signalling mechanisms, with a multitude of often interconnected potential therapeutic targets. Therefore, there is a need for the development of multi-compartment delivery vehicles for combinatorial and synergistic therapeutic approaches. In this study it was hypothesized that multi-compartment crosslinked collagen type I systems can deliver multiple bioactive agents in a controlled manner in an in vitro model condition of skin fibrosis. Multi-compartment collagen-based systems were made using solutions of dialyzed type I collagen mixed with 10× PBS, after which they were neutralised and crosslinked with 1 and 2.0 mM 4 arm-succinimidyl glutarate ester PEG (4 arm-PEG-SG), respectively, followed by incubation at 37ºC. The systems were characterised through swelling assessment, collagenase degradation assay and compression tests. The release of encapsulated drugs from the hydrogels was studied by ELISA and the effect of the delivered bioactive agents was assessed through imaging and quantification for fibrotic markers in an in vitro model. A pilot study using FITC-dextran proved that the inner compartment was capable of promoting a sustained release over a long period of time (7 days), which was further confirmed with drug release assays using a TNF-α antagonist and recombinant decorin, fitting the intended therapeutic release profile. Protein expression studies showed a decrease of endogenous collagen type I and α-smooth muscle actin expression (p<0.05) indicating amelioration of fibrosis. In summary, this indicates that this system is suitable for dual delivery of multiple bioactive agents, resulting in a controlled release in vitro and illustrating its potential in therapy.


Full Access
R. Gibney S. Matthyssen J. Steele J. Patterson N. Zakaria E. Ferraris

Collagen is a key component of the extracellular matrix in a variety of tissues and hence is widely used in tissue engineering research, yet collagen has had limited uptake in the field of 3D printing. In this study we successfully adapted an existing electronic printing method, aerosol jet printing (AJP), to print high resolution 3D constructs of recombinant collagen type III (RHCIII). Circular samples with a diameter of 4.5mm and 288 layers thick, or a diameter of 6.5mm and 400 layers thick were printed on glass cover slips with print lines of 60µm. Attenuated Total Reflectance Fourier-Transorm Infa-red (ATR-FTIR) spectroscopy performed on the 4 of the printed samples and dried non-printed RHCIII samples showed that no denaturation had occurred due to the printing process. Printed samples were crosslinked using EDC [N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride, Sigma Aldrich] to improve their stability and mechanical strength. Differential scanning calorimetry (DSC) performed showed a marked difference in the denaturation temperature between crosslinked printed samples and fibrillar non-printed samples and nano-indentation showed that the construct was relatively stiff. Previous results with similar samples have shown that mesenchymal stem cells (MSCs) align with and travel parallel to print direction. Results obtained from these samples show signs that they might be applied in other areas such as bone tissue engineering.


G. J. Atkins

Periprosthetic joint infections (PJI) are increasing in prevalence and are recognised as one of the most common modes of failure of joint replacements. Osteomyelitis arising from PJI is challenging to treat, difficult to cure and increases patient mortality 5-fold. PJI can have subtle symptoms and lie dormant or go undiagnosed for many years, suggesting persistent bacterial infection. Staphylococcus aureus is the most common pathogen causing PJI. Osteocytes are the most numerous and long-lived cell type in hard bone tissue. Our recent work has shown that S. aureus can infect and reside in human osteocytes without causing cell death, both experimentally and in bone samples from patients with PJI. Osteocytes respond to infection by the differential regulation of a large number of genes, suggesting previously unknown immune functions of this important cell type. S. aureus adapts during intracellular infection of osteocytes by adopting a quasi-dormant, small colony variant (SCV) phenotype, a property of several bacterial species known to cause PJI, which could contribute to persistent or silent infection. These findings shed new light on the aetiology of PJI and osteomyelitis in general. Further elucidation of the role of osteocytes in bone infection will hopefully lead to improved disease detection and management.


E. Greenfield

Considerable evidence exists that aseptic loosening is initiated by wear particles that recruit macrophages and stimulate their production of pro-inflammatory cytokines. The cytokines primarily act indirectly by inducing production of RANKL, which stimulates osteoclast differentiation, osteolysis, and inflammatory bone loss. There is also considerable evidence that activation of macrophage Toll-like Receptors (TLRs) contributes to this cascade of events. It is however controversial whether bacterially-derived immunostimulatory molecules known as Pathogen-Associated Molecular Patterns (PAMPs) can contribute to aseptic loosening by stimulating their cognate TLRs on macrophages. Priming and subsequent activation of the NLRP3 inflammasome is essential for macrophage production of mature, active IL-1β in response to wear particles. We recently confirmed that wear particles can activate pre primed NLRP3 inflammasomes in the absence of PAMPs. Thus, activation of the NLRP3 inflammasome is the only macrophage-based event in the aseptic loosening cascade that we have found to date is independent of PAMPs. In contrast, priming of the NLRP3 inflammasome by wear particles requires PAMPs as well as their cognate TLRs. These results add to the growing body of evidence that bacterially-derived PAMPs can contribute to aseptic loosening.


E. J. Ryan A. J. Ryan A. Philippart F. E. Ciraldo A. R. Boccaccini C. J. Kearney F. J. O'Brien

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.


B.G. Pijls I.M.J.G. Sanders E.J. Kuijper R.G.H.H. Nelissen

The main problem of infected orthopaedic implants is that the presence of microorganisms in an organized biofilm making them difficult accessible for antibiotics. This biofilm consists of a complex community of microorganisms embedded in an extracellular matrix that forms on surfaces such as an implant. Non-contact induction heating uses pulsed electromagnetic fields to induce so-called ‘eddy currents’ within metal objects which causes them to heat up. This heat causes thermal damage to the bacterial biofilm hence killing the bacteria on the metal implant. The purpose of this study is to determine the effectiveness of induction heating on killing Staphylococcus epidermidis in a biofilm. S. epidermidis biofilms were grown on Titanium alloy (Ti6Al4V) coupons and subsequently were heated with a custom-built induction heater to temperatures of 60°C, 70°C, 80°C and 90°C for 3.5 minutes. Temperature was controlled with an infra-red thermal sensor and micro-controller. We also included two control conditions without induction heating: C1 without induction heating and C2 with chlorhexidine 0.5% in 70% alcohol without induction heating. Experiments were repeated 5 times. In the C1 group (no induction heating), 1.3 * 10(7) colony forming units (CFU)/cm(−2) of S. epidermidis were observed. For 60°C, 70C, 80 C and 90C, a 3.9-log reduction, 5.3-log reduction, 5.5-log reduction and 6.1-log reduction in CFU/cm(−2) were observed, respectively. For the C2 (chlorhexidine) there was a 6.7-log reduction CFU/cm(-2). We concluded that induction heating of Titanium coupons is effective in reducing bacterial load in vitro for S. epidermidis biofilms.


F. Reffuveille J. Varin-Simon V. Vernet-Garnier J. Madoux S.C. Gangloff X. Ohl C. Mongaret

Prosthetic Joint Infections (PJIs) are increasing with the use of orthopedic devices on an ageing population. Cutibacterium acnes is a commensal organism that plays an important role in the ecosystem healthy human skin, yet this species is also recognized as a pathogen in foreign body infection: endocarditis, prostatitis and specifically in PJIs. C. acnes is able to escape the immune system. This phenomenon could reflect two bacterial behaviour: the bacterial internalization by host cells and the biofilm formation. In this study, we studied different clinical strains of C. acnes. We noticed that C. acnes isolated from PJIs form 2 fold-more biofilm than the strains isolated from a normal skin in two models (Crystal violet staining and fluorescent microscopy (p=0.04 and p=0.02, respectively, Mann-Whitney test). We did not observe any difference in the internalization rate of those strains by osteoblasts. However, the quantity of biofilm formed by C. acnes before and after the internalization was compared. A significant increase in biofilm formation was observed for the strains isolated from the skin (x2.3±0.07; p=0.008, Mann-Whitney test). However, the hydrophobicity of the skin strains is significantly less important than for the PJIs strains (24.8±13% vs 56.6±12% respectively; p=0.003, Mann-Whitney test) but this did not change after internalization suggesting that there is no cell wall evolution. In conclusion, we studied for the first time the impact of bacterial internalization by osteoblasts on the virulent behaviour of C. acnes, which could explain the hided pathogenicity of this commensal bacterium.


E.S. Veltman E. Lenguerrand D.J.F. Moojen M.R. Whitehouse R.G.H.H. Nelissen A.W. Blom R.W. Poolman

Administration of perioperative antibiotic prophylaxis (AP) reduces the risk of prosthetic joint infection (PJI) following primary total hip (THA) and knee (TKA) arthroplasty. The optimal type of antibiotic used, and duration of prophylaxis are subject to debate. We compared the risk of revision surgery for PJI in the first year following THA and TKA by AP regimen. A national survey collecting information on hospital-level AP regimen policy was conducted across the Netherlands and linked to data from the LROI arthroplasty registry for 2011–2015. PJI status was defined using the surgical indication reported at revision by surgeons in the registry form. Restricted cubic splines Poisson model adjusted for hospital clustering were used to conduct the comparisons on 130,712 THAs and 111,467 TKAs performed across 99 institutions. These included 399 THAs and 303 TKAs revised for an indication of PJI. Multiple shot of Cefazolin (MCZ), of cefuroxime (MCX) and single shot of Cefazolin (SCZ) were respectively administrated to 87%, 4% and 9% of patients. For THA, the rates of revision for PJI were respectively 31/10,000 person-years 95%CI[28, 35], 39[25, 59] and 23[15, 34] in the groups which received MCZ, MCX and SCZ; respectively, the rates for TKA were 27[24, 31], 40[24, 62] and 24[16, 36]. No evidence of difference between AP regimens was found in the unadjusted and adjusted model (age, gender, BMI and ASA grade). Further work is advocated to confirm whether there is an association between AP regimen collected at patient-level and the risk of subsequent revision for PJI.


J. Waddell

We have undertaken a series of clinical trials over the last 20 years to look at different bearing surface combinations in young adults. We continue to follow these patients well beyond the planned duration of the trials and new information is constantly becoming available. The first trial compared ceramic-on-ceramic with ceramic-on-standard-polyethylene. These patients have now been followed for 20 years with significant wear in the polyethylene group but virtually identical revision rates. The second trial ceramic-on-ceramic, cobalt-chrome-on-standard-polyethylene and cobalt-chrome-on-cross-linked-polyethylene. In this group the ceramic-on-ceramic patients have the lowest revision rate; the ceramic-on-polyethylene group demonstrates a lower wear rate than cobalt-chrome-on-polyethylene. The third trial looks at cobalt-chrome versus zirconium on either cross-linked polyethylene or conventional polyethylene. At 10 years there remains no evidence of improved performance from the zirconium surface as compared to cobalt-chrome. The cross-linked polyethylene group is clearly outperforming the conventional polyethylene in terms of wear rate but at 10 years the revision rates remain the same in all groups. Cross liked polyethylene appears to be the major determining factor in prosthetic longevity and appears to be more important than the counter face material.


R. H. S. Gill

Metal on metal hip replacements have been one of worst failures in recent years in terms of orthopaedic implants. Some of these devices have had catastrophic failure rates, with reports of 48% failure at 6 years. The failure of these devices has led to considerable suffering, pain and reduction in quality of life; consequently, they have given rise to high costs and multi-million-dollar legal cases. This talk will describe the history of the current metal on metal failure and discuss some of the reasons why might have occurred. It will also consider the reasons that wear debris arising from the trunnion is worse in terms of biological activity then that arising from the bearing surfaces.


C. Scholes M. Ebrahimi S.B. Farah C. Field D. Kerr L. Kohan

The aim of this study was to report the procedure survival and patient-reported outcomes in a consecutive series of patients <50yrs at the time of hip arthroplasty with a metal-on-metal hip resurfacing system who have progressed to a minimum of 10yrs follow-up. Patients presenting for treatment of degenerative conditions of the hip electing to undergo hip resurfacing were included in a clinical registry (N=226 patients; 238 procedures). Procedure survival was confirmed by crosschecking to the Australian Orthopaedic Association National Joint Replacement Registry and comparing to all procedures by other surgeons nationwide. Kaplan-meier survival curves with 95% confidence intervals were constructed, while patient-reported outcome measures were compared with t-tests and postoperative scores assessed with anchor analysis to age and gender-matched normative data. At mean follow up of 12 years, six cases were revised with a cumulative survival rate of 96.8% (95%CI 94.2–99.4) at 15 years. Majority of revisions were early (<3yrs) and occurred in females (N=4). Patient-reported general health, disease state, hip function and activity level maintained large improvements beyond 10 years post-implantation and were equal to or exceeded age and gender-matched normative data. Metal-on-metal hip resurfacing in males and females aged <50 years at time of surgery demonstrated a high rate of cumulative survival beyond 10 years follow up. The results demonstrate excellent outcomes in this age group.


Y. Kubo G. Motomura S. Ikemura H. Hatanaka J. Fukushi S. Hamai T. Yamamoto Y. Nakashima

Similar to the radiological findings in rapidly destructive arthrosis of the hip joint (RDA), subchondral insufficiency fracture of the femoral head (SIF) can result in progressive femoral head collapse of unknown etiology. We thus examined the osteoclast activity in hip joint fluid in SIF with progressive collapse in comparison to that in RDA. Twenty-nine hip joint fluid samples were obtained intraoperatively with whole femoral heads from 12 SIF patients and 17 RDA patients. SIF cases were classified into subgroups based on the presence of ≥2mm collapse on preoperative radiographs: SIF with progressive collapse (n=5) and SIF without progressive collapse (n=7). The levels of tartrate-resistant acid phosphatase (TRACP)-5b, interleukin-8, vascular endothelial growth factor (VEGF), and matrix metalloproteinase (MMP)-9 were measured. Numbers of multinuclear giant cells at the subchondral region were assessed histopathologically using mid-coronal slices of each femoral head specimen. Median levels of all markers and median numbers of multinuclear giant cells in SIF with progressive collapse were significantly higher than those in SIF without progressive collapse, while there were no significant differences in SIF with progressive collapse versus RDA. Regression analysis showed that the number of multinuclear giant cells correlated positively with the level of TRACP-5b in joint fluid. This study suggests an association of increased osteoclast activity with the existing condition of progressive collapse in SIF, which was quite similar to the findings in RDA. Therefore, high activation of osteoclast cell may reflect the condition of progressive collapse in SIF as well as RDA.


B. Hardy M.N. Armitage D.O. Khair N. Nandan E.R. Pettifor D. Lake A. Lingham A. Relwani

The World Health Organisation (WHO) Surgical Safety checklist is an evidence-based tool shown to reduce surgery-related morbidity and mortality. Despite audits showing 96% checklist compliance, our hospital had 3 surgical never events in 10 months, 2 of which were in orthopaedics. By March 2018, the authors aimed to achieve 100% compliance with all 5 sections of the WHO Five Steps to Safer Surgery bundle for all surgical patients. Additionally, the authors aimed to assess the impact of the quality of bundle delivery on preventable errors related to human factors. Quantitative assessment involved direct observations of compliance in theatres. Qualitative data in the form of rich, descriptive observations of events and discussions held during checklist delivery was analysed thematically. Interventions included trust-wide policy changes, awareness sessions, introduction of briefing and debrief proformas and documented prosthesis checks. For elective surgeries, checklist compliance increased to 100% in 4 of 5 sections of the bundle. The incidence of reported preventable critical incidents decreased from 6.7% to 2.4%. A chi-squared test of independence demonstrated a significant relationship between the implementation of changes and completion of the checklist, X2 (1, N = 1019) = 25.69, p < 0.0001. Thematic analysis identified leadership, accountability, engagement, empowerment, communication, and teamwork as factors promoting effective checklist use. Our findings highlight the benefits of a qualitative approach to auditing checklists. Exploring the role of human factors and promoting staff awareness and engagement improves checklist compliance and enhances its effectiveness in reducing surgery-related adverse outcomes.


G.A. Sheridan R.M. Kelly S.M. McDonnell P.J. Kenny

This was a retrospective study of registry data from a National Orthopaedic Hospital for all THRs with 10-year follow-up data. Inclusion criteria were all THRs with a minimum of 10-year follow-up data. All metal-on-metal (MoM) THRs and MoM resurfacings were excluded from the analysis due to the high rate of revision associated with these bearings. Univariate and multivariate analyses controlling for confounding variables were performed to compare outcomes. A total of 1,697 THRs were performed in 1,553 patients. The four significant predictors for revision were fixation type (p<0.01), surface bearing type (p<0.01), age (P<0.05) and head size (p<0.05). Gender, BMI and approach had no effect on revision rates. The lowest 10-year all-cause revision rates were seen in cemented THRs at 1.7%. Ceramic-on-poly bearings had the lowest revision rate at only 1.2%. Metal-on-poly bearings had a 1.7% revision rate. Ceramic on ceramic bearings had a 7.1% revision rate with 1 revision for squeak and 1 revision for ceramic head fracture. The causes for revision in order of decreasing frequency were as follows: Infection (n=13, 0.7%), dislocation (n=7, 0.4%), periprosthetic fracture (n=3, 0.2%) and aseptic loosening (n=2, 0.1%). There were 2 re-revisions at 10 years in total. The smaller 22.225mm head sizes had a significantly lower revision rate than other head sizes (p<0.05). Ceramic-on-poly bearings, cemented fixation and smaller head sizes perform better in the experience of this registry. However, with multivariate analysis, these differences were shown to be insignificant.


T. Spalding

Articular cartilage injury has a high prevalence in elite and recreational athletes. Articular cartilage repair remains a challenge due to cost effectiveness and clinical effectiveness issues. There are now several effective technologies and it is possible to return to competitive sports following many of the procedures available. The durability of repair tissue is variable and there remains extensive growth in the Scientific world. Evolving cartilage restoration technologies focus on increasing cartilage quality and quantity, while optimising surgery and rehabilitation. In UK ACI has undergone extensive cost effectiveness analysis and the in-depth review has shown that ACI is cost effective compared to microfracture. ACI is indicated for lesions >2cm sq but NICE has considered that it is not indicated for problems after microfracture. This presentation details the various options available to surgeons and examines the cost effectiveness.


Full Access
F. Barry

Osteoarthritis (OA) of the spine and diarthrodial joints is by far the most common cause of chronic disability in people over 50 years of age. The disease has a striking impact on quality of life and represents an enormous societal and economic cost, a burden that will increase greatly as populations age. OA is a complex condition with broad pathology. Damage to the articular cartilage is a consistent feature, accompanied by changes to the subchondral bone and synovium. Progression of the disease involves further degeneration of the articular cartilage, damage to the underlying bone and morphological changes that include subchondral bone thickening, development of cysts, osteophytes and inflammation of the synovium. Enhanced production of proinflammatory cytokines and matrix metalloproteinases accelerates degradation of the articular cartilage. It is striking that no approved pharmacological intervention, biological therapy or procedure prevents the progressive destruction of the OA joint. All current treatments, without exception, produce symptomatic rather than regenerative results. While there have been some exciting developments in the search for OA treatments in the last decade, including matrix metalloproteinase inhibitors, anti-TNF and anti-IL1 drugs for example, none of these has to date emerged as an effective medicinal product. There is thus an urgent and compelling need to identify, validate and test new biological therapeutics. Stromal cell therapy represents one such compelling approach. The results from several early clinical studies have indicated that this approach holds a great deal of promise for the treatment of OA. Most studies have involved direct intraarticular injection of a suspension of mesenchymal stromal cells (MSCs) for treatment of knee OA. Results from a number of controlled patient studies have suggested that this treatment results in an effective repair response. Although data regarding mechanism of action are limited, it appears that the cells have an anti-inflammatory effect, possibly targeting cells within the synovium, rather than a direct cartilage repair effect. Several recent reports have highlighted a dramatic and sustained response in patients receiving MSC treatment. For example, allogeneic expanded adipose-derived MSCs have been shown to be safe and effective in the treatment of complex perianal fistulas in Crohn's disease. Also, allogeneic bone marrow-derived MSCs has a been shown to have a positive effect in pediatric acute graft versus host disease. These observations point to a mechanism of action that involves host immunomodulation, but this needs further examination. Within the field of musculoskeletal disease effective translation of MSC technology has been hindered by a lack of randomized controlled patient studies, severe inconsistencies regarding the preparation and characterization of the cell product, and an incomplete understanding of the therapeutic mechanism. Direct to consumer clinics have flourished in some countries, providing cell treatments to OA patients. Most or all of these utilize unexpanded cell fractions from marrow or fat without even rudimentary product characterization and may report an exaggerated clinical outcome. Data from these clinics is not likely to yield information that will be useful. In fact, a recent systemic review of clinical trials involving MSC treatment in OA indicated that only a limited number of studies provided high quality evidence and long term follow up. Many suffered from a lack of consistency, including a diversity of methods for MSC preparation, and thus did not contribute to a supporting evidence base. There is a compelling need to provide clear and unambiguous clinical proof of concept relating to MSC treatment for OA. The ADIPOA2 study, currently active in Europe, will go some way towards achieving this. This is a 150 patient, phase 2b study designed to to assess the efficacy of a single injection of autologous adipose-derived MSCs in the treatment of mild to moderate OA of the knee, active and unresponsive to conservative therapy for at least 12 months.


S. Nürnberger

Organ and tissue decellularisation are promising approaches for the generation of scaffolds for tissue regeneration since these materials provides the accurate composition and architecture for the specific tissues. Repopulation of the devitalized matrixes is the most critical step and a challenge, especially in dense tissues such as cartilage. To overcome this difficulty, several chemical and mechanical strategies have been developed. Chemical extraction targeting specific matrix components such as elastin, makes auricular cartilage accessible for cells via channels originating from the elastic fiber network. However, chemical treatment for glycosaminoglycan removal is not sufficient to allow cell ingrowth in articular cartilage. As alternative, laser perforation has been developed allowing to engrave fine structures with controlled size, distance and depth, with reproducibility and high throughput. Two of the most commonly used laser technologies used in the medical field, the CO2 and femtosecond laser, were applied to hyaline cartilage with very different structural effect. Within this talk, the structuralizing possibilities of laser and enzymatic treatments, the effect on the matrix and the general advantages and disadvantages for tissue engineering are discussed. We believe that the optimal combination of chemical and laser treatment has high potential for a new generation of biomaterials for tissue engineering.


J. C. Silva C. Moura G. Borrecho A. Alves de Matos J. M. S. Cabral R. J. Linhardt F. C. Ferreira

Bioreactors have been used in articular cartilage tissue engineering (AC-TE) to apply different mechanical stimuli in an attempt to better mimic the native AC microenvironment. However, these systems are often highly complex, costly and not very versatile. In this work, we propose a simple and customizable perfusion bioreactor fabricated by 3D-extrusion to study the effect of shear stress in human bone-marrow mesenchymal stem cells (hBMSC) cultured in 3D porous polycaprolactone (PCL) scaffolds. Prototype models were designed in a CAD-software to perfectly fit the scaffolds and computational fluid dynamics analysis was used to predict the fluid velocities and shear stress forces inside the bioreactor. For the culture studies, hBMSC-PCL constructs were cultured under static expansion for 2 weeks and then transferred to the ABS-extruded bioreactors for continuous perfusion culture (0.2mL/min) under chondrogenic induction for additional 3 weeks. Perfused constructs showed similar cell proliferation and higher sGAG production in comparison to the static counterparts (bioreactor without perfusion). Constructs exposed to shear stress stimuli presented higher expressions of chondrogenic genes (COLII/Sox9/Aggrecan) and reduced expressions of COLI and Runx2 (osteogenic) than static group. However, the higher expression of COLX in the perfused constructs suggests a shear stress role in AC hypertrophy. Both conditions (perfused/static) stained positively for GAG deposition and for the presence of collagen II and aggrecan. Overall, the results provide a proof-of-concept of our customizable extruded bioreactor envisaging applications as a platform for AC-TE repair strategies and in the development of more reliable in vitro models for disease modelling and drug screening.


M. Lemoine J. O'Byrne D. J. Kelly F. J. O'Brien

Damage to articular cartilage is difficult to treat, as it has a low capacity to regenerate. Biomimetic natural polymer scaffolds can potentially be used to regenerate cartilage. Collagen hyaluronic acid (CHyA) scaffolds have been developed in our laboratory to promote cell infiltration and repair of articular cartilage. However, the low mechanical properties of such scaffolds potentially limit their use to the treatment of small cartilage defects. 3D-printed polymers can provide a reinforcing framework in these scaffolds, thus allowing their application in the treatment of larger defects. The aim of this study was to create mechanically functional biomaterial scaffolds by incorporating a CHyA matrix into 3D-printed polymer meshes resulting in an integrated porous material composite with improved mechanical properties for repair of large cartilage defects. 3D-printed meshes were developed to facilitate an architecture suitable for nutrient flow, cell infiltration, and even CHyA incorporation. And the meshes were freeze dried in custom made moulds to create a pore structure suitable for chondrogenesis. Uniaxial compressive testing of the scaffolds revealed improved mechanical properties following reinforcement with printed meshes, with the compressive modulus increasing from 0.8kPa (alone) to 0.5MPa (reinforced structure). The reinforced scaffolds maintained interconnected pores with the mean pore diameter increasing from 130 to 175µm. The reinforcement had no negative impact on MSC viability, with 90.1% viability in reinforced scaffolds at day 7. The compressive modulus of the reinforced CHyA scaffold is close to native articular cartilage, suggesting that this approach can be used for treatment of large cartilage defects.


M. Xu E.-L. Stattin G. Shaw D. Heinegård G. Sullivan I. Wilmut A. Colman P. Önnerfjord A. Khabut A. Aspberg P. Dockery M. Murphy F. Barry

Familial osteochondritis dissecans (FOCD) is an inherited defect of cartilage and bone characterized by development of large cartilage lesions in multiple joints, short stature and early onset osteoarthritis. We have studied a family from Northern Sweden with FOCD over five generations. All affected family members have a heterozygous missense mutation on exon 17 of the aggrecan gene, resulting in a Val-Met amino acid replacement in the G3 aggrecan C-type lectin domain (CLD). Aggrecan, a major proteoglycan of articular cartilage produced by chondrocytes, has a large protein core richly substituted with sulfated glycosaminoglycan chains. The unique structure, its high concentration within the cartilage extracellular matrix and its ability to form a supermolecular complex with hyaluronan and bind to other matrix proteins all profoundly influence the biomechanical properties of the tissue. Deletion of CLD in a chick aggrecan construct was found to influence its secretion from chondrocytes and human aggrecan constructs carrying the V2303M mutation showed diminished interactions with the ECM proteins tenascin-R, fibulin-1 and fibulin-2. To investigate the pathogenesis of FOCD, we studied chondrogenic differentiation of patient bone marrow mesenchymal stem cells and induced pluripotent stem cells. We demonstrated that the mutation results in accumulation of unfolded or misfolded aggrecan within the lumen of the chondrocyte endoplasmic reticulum. Associated with this is the failure to assemble a normal extracellular matrix. This explains the susceptibility of these patients to cartilage injury and the degenerative changes that lead to early onset osteoarthritis.


E. Murphy C. Fenelon C. Egan S. Kearns

Osteochondral lesions (OCLs) of the talus are a challenging and increasingly recognized problem in chronic ankle pain. Many novel techniques exist to attempt to treat this challenging entity. Difficulties associated with treating OCLs include lesion location, size, chronicity and problems associated with potential graft harvest sites. Matrix associated stem cell transplantation (MAST) is one such treatment described for larger lesions >15mm2 or failed alternative therapies. This cohort study describes a 5 year review of the outcomes of talar lesions treated with MAST. A review of all patients treated with MAST by a single surgeon was conducted. Pre-operative radiographs, MRIs and FAOS outcome questionnaire scores were conducted. Intraoperative classification was conducted to correlate with imaging. Post-operative outcomes included FAOS scores, return to sport, revision surgery/failure of treatment and progression to arthritis/fusion surgery. 32 patients were identified in this cohort. There were 10 females, 22 males, with an average age of 35. 01. 73% had returned and continued playing active sport. 23 patients underwent MAST in the setting of a failed previous operative attempt, with just 9 having MAST as a first option. 9 patients out of 32 had a further procedure. Two patients had a further treatment directed at their OCL. Two patients had a fusion, 2 had a cheilectomy at > 4 years for impingement, one had a debridement of their anterolateral gutter, one had debridement for arthrofibrosis, one patient had a re alignment calcaneal osteotomy with debridement of their posterior tibial tendon. MAST has demonstrated positive results in lesions which prove challenging to treat, even in a “failed microfracture” cohort.


P. Beaulé

Total hip arthroplasty (THA) is one of the most successful surgery. However, patients' expectations have increased over the last two decades in regards to hip function after joint replacement, the patients assume to return their daily and sport activities without major limitations. This presentation will examine the effect of surgical approaches and implant designs as well as rehabilitation protocol on the clinical and biomechanical outcomes after THA. The new implant designs for THA aim to improve joint function whereas the surgical approaches intend to reduce muscle damage to regain muscle strength. One important determinant measured from gait analysis is the hip abduction moment as the abductors play a key role in stabilizing the pelvis in the frontal plane, particularly in phases of transition, such as the single leg stance in walking or stair climbing. This showed that muscle strength needs to be preserved. To minimize the risk of hip joint instability, a strong focus of implant development has been carried out. To illustrate this important concept within the context of gait analysis, I will present two studies that examine the influence of surgical approach and biomechanical reconstruction; and the second, is a prospective RCT comparing a dual mobility implant to a standard total hip replacement.


P. McGarry

Several experimental studies demonstrate that controlled substrate micro-patterning has a significant impact on cell behaviour. Several experiments reveal cell spread area is dependent on both substrate rigidity and ligand density. The biomechanisms underlying such observations are not fully understood. We demonstrate that a thermodynamically consistent statistical mechanics model explains several of the key phenomena observed experimentally. We implement a steady-state thermodynamically consistent framework for stress-fibre formation and focal adhesion assembly. A Markov chain Monte-Carlo (MCMC) methodology is used to compute the distribution of cell spread states for a given substrate ligand density and stiffness. Several million spread states are considered by imposing a sequence of random trial moves on the cell. For each spread state, we compute quantities such as the cytoskeletal protein distribution, SF orientation, and FA distribution via a mixed finite element/boundary element method scheme. The free energy of all accepted states averaged equates to the homeostatic free energy. Following completion of the MCMC scheme we can construct the probability distribution for an observable of interest. For cells on a rigid substrate both the mean spread area and SD increase as the collagen density increases. A peak spread area is observed at a collagen density of 300 ng.cm-2, with an area A/A0≅2.7. Further increases in collagen density lead to a reduction in cell area, motivated by focal adhesion free energy. On a compliant elastic substrate, lower spread areas are observed (peak A/A0≅1.8). Our computed dependence of spread area on substrate stiffness and ligand density has been observed experimentally.


D.S. Catelli K.C.G. Ng E. Kowalski P.E. Beaulé M. Lamontagne

Cam-type femoroacetabular impingement (FAI) is a common cause for athletic hip injury and early hip osteoarthritis. Although corrective cam FAI surgery can improve patient reported outcome measures (PROMs), it is not clear how surgery affects muscle forces and hip joint loading. Surgery for FAI may redistribute muscle forces and contact forces at the hip joint during routine activities. The purpose of this study was to examine the muscle contributions and hip contact forces during gait in patients prior and after two years of undergoing surgery for cam FAI. Kinematics and kinetics were recorded in 11 patients with symptomatic cam FAI as they completed a gait task. Muscle and hip contact forces during the stance phase were estimated using musculoskeletal modelling and static optimization in OpenSim. All patients reported improvements in PROMs. Post-operatively, patients showed reduced forces in the long head of the biceps femoris at ipsilateral foot-strike and in the rectus femoris at the contralateral foot-strike. The reduced muscle forces decreased sagittal hip moment but did not change hip contact forces. This was the first study to evaluate hip muscle and contact forces in FAI patients post-operatively. Although hip contact forces are not altered following surgery, muscle forces are decreased even after two years. These findings can provide guidance in optimizing recovery protocols after FAI surgery to improve hip flexor and extensor muscle forces.


C. Greaney C. Duffy D. Hoey M.G. Monaghan

Recapitulating tissue elasticity can direct mesenchymal stromal cell (MSC) differentiation; however, it is unclear how substrate elasticity affects MSC metabolism. It is hypothesized MSCs subjected to stiffnesses, atypical of standard tissue culture plastic, display altered metabolic phenotypes during differentiation. In this study, such alterations in MSC metabolic profiles, based on the fluorescence lifetime of NAD(P)H, a critical co-factor in energy production, were monitored using Fluorescence lifetime imaging microscopy (FLIM) as an evaluation tool. Polyacrylamide substrates with varying stiffnesses were fabricated to model the native elasticity of cartilage and bone. MSCs cultured on these substrates exhibited potent alterations in their metabolic status over a 14-day period that were detectable as early as day 3 using FLIM. Overall, soft substrates induced a more glycolytic response after 10 days of culture that persisted at day 14 (as measured by protein-bound NAD(P)H contributions to the lifetime decay). Similarly, by day 10; MSCs on intermediate-stiffness substrates favoured glycolysis. MSCs on stiffer substrates initially displayed a glycolytic phenotype followed by a transition to oxidative phosphorylation by day 10. Staining for mineralised nodules and glycosaminoglycans verified MSCs on stiffer substrates differentiating towards an osteogenic lineage, while MSCs on intermediate substrates showed similarities with differentiated chondrocytes. Overall, it can be concluded that matrix stiffness can induce metabolic perturbations in MSCs for up to 14 days. From this research, ideal culture conditions in which the metabolics of MSCs could be manipulated to promote maximum potency could potentially be defined in the future.


M. Jalal R. Wallace H. Simpson

There is a growing trend towards using pre-clinical models of atrophic non-union. This study investigated different fixation devices, by comparing the mechanical stability at the fracture site of tibia bone fixed by either intramedullary nail, compression plate or external fixator. 40 tibias from adult male Wistar rats' cadavers were osteotomised at the mid-shaft and a gap of 1 mm was created and maintained at the fracture site to simulate criteria of atrophic non-union model. These were divided into five groups (n=8 in each): the first group was fixed with 20G intramedullary nail, the second group with 18G nail, the third group with 4-hole plate, the fourth group with 6-hole plate, and the fifth group with external fixator. Tibia was harvested by leg disarticulation from the knee and ankle joints, the soft tissues were carefully removed from the leg, and tibias were kept hydrated throughout the experiment. Each group was then subdivided into two subgroups for mechanical testing: one for axial loading (n=4) and one for 4-point bending (n=4). Statistical analysis was carried out by ANOVA with a fisher post-hoc comparison between groups. A p-value less than 0.05 was considered statistically significant. Axial load to failure data and stiffness data revealed that intramedullary nails are significantly stronger and stiffer than other devices, however there was no statistically significant difference axially between the nail thicknesses. In bending, load to failure revealed that 18G nails are significantly stronger than 20G. We concluded that 18G nail is superior to the other fixation devices, therefore it has been used for in-vivo experiments to create a novel model of atrophic non-union with stable fixation.


J. Pyszkowski B. Pouran G. Mattei E. Breel

The micro-mechanical properties of complex biomaterials play an important role in tissue engineering and regenerative medicine, by regulating cellular processes and signalling. Local characterization of complex tissues while immersed in liquids proves to be very difficult to perform. We therefore present a method to derive viscoelastic micro-mechanical properties via non-destructive nano-indentation measurements in liquid. This technique is featured with a fiber-optical ferrule-top micro-machined force transducer, enabling a wide range of mechanical tests: from quasi-static experiments to derive elastic moduli, to step-response tests (e.g. creep, stress-relaxation), dynamic mechanical analysis (DMA) and constant strain rate tests to characterize sample viscoelastic behaviour. As a complex application we here present the osteochondral (OC) interface, which gradually ranges from hard and stiff bone regions towards softer and viscoelastic articular cartilage covering joint surface. The osteochondral plugs were collected from medial femoral condyle of cadaveric knees and measured at 37°C to mimic in-vivo physiological-like conditions. The stiffness of articular cartilage was 1.58±0.06 MPa, whereas subchondral bone plate could be categorized in “softer” region with 68.24±37.43 MPa, and a “stiffer” region with 683.68±622.88 MPa. The high stiffness in the “hard” region could be attributed to the mineralized matrix in the contact area, whereas the contribution of gel-like material, containing cell processes, along with osteocytes was larger in the “soft” region of the subchondral bone plate, leading to lower stiffness. These results might correlate with differences in extracellular matrix (ECM) composition and micro-architecture and are essential for engineering functional gradient scaffolds to better understand cell-ECM interactions.


S.M. Naqvi J.A.P Perez L.M. McNamara

3D cell culture studies more accurately represent the complex in vivo mechanical environment of human bone and are, thus, superior to 2D studies when testing the efficacy of osteoporosis therapies. As such, the objective of this study was to use a 3D model to investigate the effect of sclerostin antibodies. Sclerostin is a protein, which inhibits osteoblasts and is downregulated under mechanical stimulation. It is not yet known how expression of sclerostin mediates the site-specific and temporal changes in mineralisation. To address this, we developed a 3D cellular niche of MC3T3 osteoblasts encapsulated within gelatin and applied mechanical loading to the constructs using a custom-designed compression bioreactor system (0.5% strain at 0.5 Hz, 1 hr/day) (VizStim) under continuous perfusion of cell culture media. Osteoblasts were pretreated with estrogen for 14 days, followed by estrogen withdrawal (EW) to simulate postmenopausal conditions. 3D constructs were successfully fabricated and actin staining revealed the formation of dendritic cells under both static and stimulated conditions indicative of osteocyte-like cells. Under static conditions, estrogen treatment enhanced production of calcium by osteoblasts when compared to the same cells cultured under estrogen deficient conditions. Overall, preliminary results propose a link between mechanical stimulation, estrogen deficiency and mineral production by osteoblasts. Ongoing studies are comparing the static and stimulated groups after a longer culture period of 21 days using sclerostin antibodies. This research aims to deliver further understanding of the mechanical regulation of bone formation, and will inform novel approaches for regeneration of bone tissue and treatment of osteoporosis.


E. Parle S.W. Tio A. Behre J. Carey C.G. Murphy T. O'Brien W. Curtin S. Kearns J. McCabe C.M. Coleman T. Vaughan L.M. McNamara

Recent studies have shown that bone mineral distribution is more heterogeneous in bone tissue from an animal model of osteoporosis and osteoporotic human vertebral trabeculae. These tissue alterations may play a role in bone fragility seen in osteoporosis, albeit that they are not detectable by current diagnostic techniques (dual-energy X-ray absorptiometry, DXA). Type II Diabetes Mellitus (T2DM) also increases a patient's fracture risk beyond what can be explained or diagnosed by DXA, and is associated with impaired bone cell function, compromised collagen structure and reduced mechanical properties. However, it is not currently known whether osteoporosis or T2DM leads to an increased mineral heterogeneity in the femoral head of humans, a common osteoporotic fracture site. In this study, we examine bone microarchitecture, mineralisation and mechanical properties of trabecular bone from osteoarthritic, diabetic and osteoporotic patients. We report that while osteoporotic trabecular bone has significantly deteriorated mechanical properties and microarchitecture compared to the other groups, there is also a significant increase in mean mineral content. Moreover, the heterogeneity of the mineral content in osteoporotic bone is significantly higher than osteoarthritic (+35%) and diabetic (+13%) groups. We propose that the compromised architecture following bone loss at the onset of osteoporosis alters the mechanical environment, which initiates compensatory changes in mineral content. We show for the first time that trabecular bone mineralisation is significantly more heterogeneous (+20%) in T2DM compared to osteoarthritic controls. Interestingly, bone microarchitecture and mechanical properties are not significantly different between diabetic and osteoarthritic groups despite this increase in mineral heterogeneity.


J. Moon Y. Jung J. Lee

Suture anchor have been used in surgical procedure of tendon or ligament repair. Recently, there has been developed an all suture anchor (soft anchor) which can be used even when the insertion area is narrow. But, the stability of soft anchors due to narrow zone has not been elucidated. This purpose of this study was to investigate stability of soft anchors with respect to their fixation intervals. Polyurethane foams with two different bone densities (10 pcf; 0.16g / cm³, 20 pcf; 0.32g / cm³) were used. All suture anchors and conventional suture anchors were fixed at 10mm, 5mm, and 2.5mm intervals. The failure load was measured using a mechanical testing machine. The average load to failure of conventional suture anchor were 200.4N, 200.2N, 184.7N in the 10mm, 5mm and 2.5mm interval with 10pcf foam bones and 200.4 N, 200.2 N and 184.7 N with the 20 pcf foam bone respectively. Average load to failure load of soft anchor was 97.3N, 93.9N and 76.9N with 10pcf foam bones and 200.4 N, 200.2 N and 184.7 N with 20 pcf foam bone. Suture screw spacing and bone density are important factors in anchor pullout strength. In osteoporotic bone density, insertion of the suture screw interval of 5 mm might be necessary.


J. Snedeker

Tendon tissue equilibrium very heavily depends on appropriate mechanical loading within a narrow, and still poorly defined, physiological range. We will present an overview of our recent work on the tendon cell-matrix interactions that drive tissue homeostasis, matrix remodelling and eventual tissue degeneration, and discuss a roadmap for unravelling these mechanically regulated signalling pathways for the development of effective treatment strategies. Our data suggest that tissue damage accumulates in the tendon until “intrinsic repair mechanisms” are overwhelmed. At this point, the metabolic cost of extracellular matrix remodeling exceeds the locally available nutrient supply. We hypothesize that upon reach S43.1 ing this “Metabolic Tipping Point”, the vascular system is recruited along with accompanying nerve supply (and pain) and the tissue enters into a chronic disease state characterized by high matrix turnover and increasingly poor tissue quality. In this paradigm, a delicate mechanically regulated balance exists between recruitment and suppression of the extrinsic vascular system by the resident tendon core cells. Upon injury or damage, this regulation in turn steers the tissue towards either functional remodeling or chronic tendon disease.


C. Zhao

A rotator cuff tear is one of the most common traumatic and degenerative tendon injuries resulting in over 4.5 million physician visits in the US alone. Functional restoration of rotator cuff defects usually requires surgical repair, estimated at 300,000 cased in the US annually. However, postoperative retear of repaired tendons ranges from 20% in small to medium tears to over 90% in large and massive tears. Recently, augmentation with grafting materials to strengthen a reparable tear or to bridge an unrepairable defect has become a common and attractive strategy to reduce the retear rate, especially for large or massive tears. Current graft materials, however, have encountered great challenges in achieving these goals. To meet these challenges, we have developed an engineered tendon with layered tendon-fibrocartilage-bone composite (TFBC) from patellar-tibia unit revitalized by seeding bone marrow derived stem cells (BMDSCs) within the slices, and then reassembled to an engineered tendon. Both in vitro and in vivo results have shown that engineered TFBC enhance the biomechanical strength and biological healing using canine model.


M. Delgado Cáceres D. Docheva

The establishment of a proper musculoskeletal system depends on the well-organized and synchronized development of muscle, tendon and cartilage/bone. In tendon biology, a great progress in identifying tendon-specific genes (Scleraxis, Mohawk, Tenomodulin) had been made in the last decade. However, there are many open questions regarding the exact function of genes in tendon development and homeostasis. The purpose of this study was to perform a systematic review of publications describing tendon-related genes, which were studied in-depth and characterized by using knockout technologies and the respectively generated transgenic mouse. Method: Literature search was carried out in Pubmed using “tendon” and “mouse knockout” and “phenotype” and was not limited to year. Results: We report in a tabular manner, that from a total of 25 tendon-related genes, in 23 of the respective knockout mouse models phenotypic changes were detected. Additionally, in some of the models it was described at which developmental stages these changes appeared and progressed. Interestingly, so far only loss of Scleraxis and TGFbeta signaling led to severe tendon developmental phenotypes, while mice deficient for various proteoglycans, Mohawk, EGR1 and 2, and Tenomodulin exhibited mild phenotypes. This suggests that in general the tendon developmental program is well backup and specifically that among the members of the proteoglycan family there are clear compensatory effects. In future, it will be of great importance to discover additional master tendon transcription factors as well as genes that play indispensable roles in tendon development.


Full Access
Z. Yan H. Yin M. Nerlich C. Pfeifer D. Docheva

Tendons are dense connective tissues and critical components of the musculoskeletal system with known long repair process. Tissue engineering is a promising approach for achieving complete recovery of ruptured tendons. The most studies have focused on the combination of cells with various carriers; however, frequent times the biomaterials do not match the tissue organization and strength. For this reason, we first reviewed the literature for an alternative scaffold-free strategy for tendon engineering and second, we compared the cell sheet formation of two different cell types: bone marrow-derived mesenchymal stem cells (BM-MSCs) and tendon stem/progenitor cells (TSPCs). Methods: Literature search was performed in Pubmed using “tendon tissue engineering” and “scaffold-free” keywords and was limited to the last ten years. By using a 3-step protocol, BM-MSCs and TSPCs were induced to form cell sheets in 5 weeks. The sheets were compared by analysis for weight, diameter, cell density, tissue morphology (H&E and scoring) and cartilaginous matrix (DMMB and S.O. staining). Results: Scaffold-free models (cell sheets and pellet cultures) are available; however, further optimization is needed. Comparison between the two cell types clearly demonstrated that TSPCs form more mature cell sheet, while BMSCs form larger but less organized and differentiated sheet as judged by higher cell density and lower scoring outcome. Future efforts will focus on identifying mechanisms to speed BM-MSC sheet formation and maturation, which can in turn lead to development of new methodology for scaffold-free tendon tissue engineering.


D. Stanco G. Soldati G. Ciardelli

Tendon injuries are common and current therapies often are unsuccessful. Cell-based therapy using mesenchymal stem cells (MSCs) seems to be the most promising approach to heal tendon. Moreover, providing safe and regulated cell therapy products to patients requires adherence to good manufacturing practices (GMP). Adipose-derived stem cells (n=4) were cultured in 6-well plates coated with type-I collagen in a chemically defined serum-free medium (SF) or a xenogenic-free human pooled platelet lysate medium (hPL). At passage 4, ASCs were induced to tendon lineage for 14 days using 100ng/ml CTGF, 10ng/ml TGFβ3, 50ng/ml BMP12 and 50µg/ml ascorbic acid in the SF (SF-TENO) or in the hPL (hPL-TENO) medium. Cells cultured without any supplements are used as control. Morphological appearance, cell viability and FACS were performed in undifferentiated cells to evaluate the xenogenic-free culture conditions; the gene and protein expression were performed by RT-PCR and immunofluorescence to evaluate to expression of stem cell- and tendon-related markers upon cell differentiation. SF-CTRL and hPL-CTRL showed similar viability and MSC's surface proteins and expressed the stemness markers NANOG, OCT4 and Ki67. Moreover, both SF-TENO and hPL-TENO expressed significant higher levels of SCX, COL1A1, COL3A1, COMP, MMP3 and MMP13 genes already at 3d (p<0.05) respect to CTRLs. Scleraxis and collagen were also detected in both SF-TENO and hPL-TENO at protein level in higher amount than CTRLs. In conclusion, ASCs exposed to CTGF, BMP12, TGFb3 and AA in both serum and xenogenic-free media possess similar tenogenic differentiation ability moving forward the GMP-compliant approaches for the clinical use of ASCs.


A. Dei W. Chang A. Sampaio D. Zeugolis

Mesenchymal stromal cells (MSCs) have been one of the most widely studied cell types in preclinical and clinical trials, due to their self-renewing, multipotent capacity, immunomodulatory properties and relative ease of isolation from multiple tissues. Despite limitations and safety concerns, fetal bovine serum (FBS) is still predominantly used for MSC expansion in clinical protocols. In addition, the undefined nature of serum composition and lot-to-lot variability have been linked to reduced reproducibility and efficiency of MSC bioprocessing. Moreover, use of animal serum in human cell culture increases the risk of contamination with adventitious pathogenic microorganisms, such as viruses, prions and bacteria. Hence, a defined serum-free formulation can provide increased safety, better control over physiological responsiveness, consistent performance and reproducible results. Here we present preliminary data on a prototype serum-free medium optimized for in vitro tenogenic differentiation of human bone marrow-derived MSCs. This serum-free formulation is capable of generating tenocyte-like cells in vitro expressing tenogenic markers such as Scx, Tnmd, TnC, Collagen I and Collagen III, whilst repressing expression of specific markers of other mesenchymal lineages.


M. Breborowicz P. Lubiatowski J. Jakubowski L. Romanowski

The retear of the rotator cuff (RC) repair is a significant problem. Usually it is the effect of poor quality of the tendon. The aim was to evaluate histologically two types of RC reconstruction with scaffold. We have chosen commercially available scaffold polycaprolactone based poly(urethane urea). Rat model of supraspinatus tendon injury was chosen. There were four study groups: RC tear (no repair) (n=10), RC repair (n=10), RC repair augmented with scaffold (n=10) and RC reconstruction with scaffold interposition between tendon and bone (n=10). The repairs were investigated histologically at 6 and 16 weeks. The results in two groups in which scaffold was used had significantly better scores at 6 weeks comparing to non-scaffold groups (16,4±3, 17,3± 2,8 vs. 12,5±4,4, 13,8±1,4 respectively) and 16 weeks (23±1,9, 22,8±1,6 vs. 13,8±3,3, 14,9± 3,8 respectively). Results in two scaffold groups improved between 6 and 16 weeks. Signs of foreign body reaction against scaffold were not observed. Application of scaffold to strengthen the repair site and bridging of the tendon defect improved healing of the RC repair in animal model at 6 and 16 weeks. The quality of reconstructed tendon improved over time. No such effect was observed in groups without repairs and isolated repairs were performed.


A. Rampin I. Skoufos A. Tzora N. Prassinos N. Diakakis D.I. Zeugolis

Tenocytes from several mammal species have been shown to be prone to phenotypic drift at early sub-culture passages. In the present study we compared allogenic and xenogenic serum supplementation suitability as a supplement for the in vitro expansion of equine tenocytes (eTCs), in combination with the presence or absence of crowding conditions. eTCs were isolated from superficial digital flexor tendon and expanded in normal growth medium containing DMEM, 10% appropriate serum, 1% penicillin/streptomycin solution. Isolation was performed by migration method in growth medium containing the selected serum. Silver staining, densitometry, zymography, immunofluorescence, metabolic activity, proliferation, viability and morphology were performed after 3, 5 and 7 days in culture with a seeding density of 10,000 cells/cm2. Treatment conditions were equine serum (ES) or foetal bovine serum (FBS), with or without 75 μg/mL of crowding agent carrageenan (CR). Viability and metabolic activity of eTCs were affected by FBS. eTCs in ES reached higher cell density than in FBS in day 7, especially with CR. Morphology of eTCs was maintained under different sera. Silver staining on pepsin digested cell layers shows that collagen type I deposition rate is remarkably enhanced in the presence of CR in all conditions. Immunofluorescence showed increased expression for collagen I, III, V and VI in both sera in the presence of CR. Deposition of all collagen types but type VI was increased by ES supplementation. We conclude that ES in combination with CR can represent a reliable choice for the ex vivo expansion of eTCs.


E. Argentieri P. Shah M. Koff B. Lin H. Potter O. Nwawka

Patellar tendinosis (PT) is common and can result in prolonged disability, especially in jumping athletes. Recently developed ultra-short-echo (UTE) MRI sequences allow for quantitative evaluation of tendon biostructure with T2* relaxometry. This study evaluated the relationships between changes over time (COT) in quantitative T2*-metrics, qualitative PT grades, and patient reported symptoms within 10 male basketball players from a single collegiate basketball team. All subjects completed weekly VISA-P symptomology questionnaires over the basketball season. Bilateral 3-Tesla MRIs (GE Healthcare) were obtained at pre- and post-season study visits. High-resolution, PD-weighted, FSE sequences were used to qualitatively grade PT. Quantitative T2*-metrics were evaluated using high-resolution, 3D, multi-echo, UTE-MRI sequences. Bilinear exponential fits of SI to corresponding echo time were used to calculate T2*-metrics. All qualitative and quantitative evaluations were region specific (proximal, middle, distal). Linear mixed effects models assessed associations of side and region with T2*-metrics. Spearman correlations evaluated relationships between outcome measures. Within and between study visits, significant side-to-side differences in T2*-metrics were found and were significantly impacted by leg dominance (p<0.05). Pre-season T2*-metrics correlated with COT in T2*-metrics, COT in T2*-metrics correlated with COT in qualitative PT grades, and post-season T2*-metrics correlated with max changes in VISA-P scores (ρ≥0.64). Quantitative T2*-metrics can detect PT and may be capable of predicting the onset of pathology. T2*-metrics could benefit the clinical management of PT: it is sensitive to changes in pathologic severity over time, and therefore can serve as a quantitative metric to guide treatment and evaluate intervention efficacy.


D. Mercer

Advancements in treating complications of operatively treated distal radius fractures. We will review tips and tricks to avoid complications associated with operative fixation of these complicated injuries. We will cover treatment of the distal radioulnar joint, associated distal ulna fracture, complications of malreduction and implant prominence. During this session, we will review the latest techniques for treating these complex distal radius fractures and how to avoid associated complications.


Full Access
R. Gray

Advancements in treating complex distal radius fractures. We will review tips and tricks in the treatment of complex articular distal radius fractures. We will discuss the treatment of carpal instability resulting from fracture of the volar marginal fragment. We will cover optimizing surgical exposure to address fractures extending from the radial styloid to the lunate facet. During this session, we will review the latest techniques for treating these complex distal radius fractures.


A.S. Galbraith S. Glynn C.M. Coleman C.G. Murphy

The international literature base demonstrates that individuals living with diabetes mellitus (DM) are at increased risk of mortality and post-operative complications following hip fracture surgery (HFS) than non-diabetics. Studies investigating databases in American, European or Asiatic populations highlight the impact geography can have on the resultant investigation. We aim to quantify the impact DM has on HFS patients in a single university hospital. The HIPE dataset of fragility fractures occurring in Galway University Hospital from 2014–2016 were analysed and cross referenced with hospital laboratory and public databases. A database of 759 individuals was created including 515 females and 237 males, with a mean age of 78+/−12.2 years, of which 110 patients had DM. The patient length-of-stay (PLOS) was comparable in all groups with patient age being the primary influencing factor. An extended PLOS correlated with an increased long-term mortality. A trend toward increased occurrence of sub-trochanteric fractures was observed in diabetics with fewer periprosthetic and intertrochanteric fractures. Patients with DM had a significant increased risk of post-operative mortality compared to non-diabetics. Males with DM where at a greater risk of death after HFS [HR 2.29, 95% CI 1.26–4.17. p=0.006] than females with DM [HR 1.69, 95% CI 0.99–2.91. p=0.056]. The presence of DM did not directly impact a patient's PLOS or increase the need for a re-operation. DM is associated with increased post-operative patient mortality and may influence the anatomical fracture pattern. This observation will support further investigation into the mechanical and biochemical changes occurring in the femur in individuals living with DM.