Advertisement for orthosearch.org.uk
Results 1 - 16 of 16
Results per page:
Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_17 | Pages 25 - 25
24 Nov 2023
Parente R Possetti V Granata V Schiavone ML Strina D Davi F Menale C Palagano E Filipović M Grčević D Bottazzi B Mantovani A Sobacchi C Inforzato A
Full Access

Aim. Osteomyelitis (OM) is a debilitating infection of the bone that originates from hematogenous spreading of microbes or contamination after surgery/fracture. OM is mainly caused by the opportunistic bacterium Staphylococcus aureus (SA), which can evade the host immune response, acquire antibiotic resistance and chronically colonize the musculoskeletal tissue . 1,2. , yet the underlying molecular and cellular processes are largely unclear. This study aimed to characterize the pathogenetic mechanisms of SA-OM with a focus on the long pentraxin 3 (PTX3), a soluble pattern recognition molecule and bone tissue component that is emerging as a new player in osteoimmunology . 3. and a diagnostic marker of periprosthetic joint infections, a common form of OM. 4. . Method. A murine model of OM based on intra-bone injection of SA was developed that closely mimicked surgery/trauma-related OM in humans and allowed addressing the role of PTX3 in gene-modified (Ptx3-/-) animals. Local and systemic infection and inflammation were assessed via microbiology, flow cytometry, histochemistry and microCT techniques. Results. SA-injected mice developed chronic infection with measurable levels of viable bone-resident bacteria up until 30 days from microbial challenge. The infection was confined to the treated limbs only and accompanied by extensive tissue remodelling. The bacterial load was higher in WT than Ptx3. -/-. animals at 6 and 14 days from SA injection. Accordingly, WT mice had enhanced systemic inflammation with expanded innate immune compartment in the spleen and increased serum levels of inflammatory cytokines and chemokines. PTX3 levels were higher in SA- than vehicle (PBS)-injected WT animals both in the serum and bone tissue. Furthermore, administration of a PTX3-targeting antibody reduced the bacterial burden in the bones of SA-injected WT mice. Conclusions. In a mouse model of SA-OM, genetic deficiency of PTX3 protected from infection and inflammation, pointing to this pentraxin as a crucial player in OM pathogenesis and a novel therapeutic target in bone infections. The study was approved by the Italian Ministry of Health (approval n. 520/2019-PR issued on 19/07/2019) and supported by Fondazione Beppe and Nuccy Angiolini


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 59 - 59
1 Jul 2020
Chim Y Cheung W Chow SK
Full Access

It has been previously shown that Low-Magnitude High-Frequency Vibration (LMHFV) is able to enhance ovariectomy-induced osteoporotic fracture healing in rats. Fracture healing begins with the inflammatory stage, and all subsequent stages are regulated by the infiltration of immune cells such as macrophages and the release of inflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and interleukin-10 (IL-10). Therefore, the aim of this study was to investigate the effect of LMFHV treatment on the inflammatory response in osteoporotic fracture healing. In this study, ovariectomy-induced osteoporotic and sham-operated closed-femoral fracture SD-rats were randomized into three groups: sham control (SHAM), ovariectomized control (OVX-C) or ovariectomized vibration (OVX-V) (n=36, n=6 per group per time point). LMHFV (35Hz, 0.3g) was given 20 min/day and 5 days/week to OVX-V group. SHAM operation and ovariectomy were performed at 6-month and closed femoral fracture was performed at 9-month. Callus morphometry was determined by callus width from weekly radiography. Local expressions of inducible nitric oxide synthase (iNOS) (macrophage M1 marker), CD206 (macrophage M2 marker), TNF-α, IL-6 and IL-10 were detected by immunohistochemistry and quantified by colour threshold in ImageJ, assessed at weeks 1 and 2 post-fracture. Significant difference between groups was considered at p≤0.05 by one-way ANOVA. Callus formation was higher in OVX-V than that of OVX-C as shown by callus width at weeks 1 and 2 (p=0.054 and 0.028, respectively). Immunohistochemistry results showed that CD206 positive signal and the M2/M1 ratio which indicates the progression of macrophage polarization were significantly higher in OVX-V rats (p=0.053 and 0.049, respectively) when compared to OVX-C at week 1. Area fraction of TNF-α positive signal was significantly higher in SHAM and OVX-V rats at week 1 (p=0.01 and 0.033, respectively). IL-6 signal was also significantly higher in SHAM and OVX-V groups at week 1 (p=0.004 and 0.029, respectively). IL-10 expression was significantly lower in SHAM and OVX-V groups at week 1 (p=0.013 and 0.05, respectively). Here we have shown that LMHFV treatment promoted the shift from pro-inflammatory stage towards anti-inflammatory stage earlier. It has been reported that the polarization of pro-inflammatory macrophages M1 to anti-inflammatory macrophages M2 was indicative of the endochondral ossification process in the long bone fracture model. Besides, we found that LMHFV treatment enhanced pro-inflammatory markers of TNF-α and IL-6 and suppressed anti-inflammatory marker of IL-10 at week 1, showing that inflammatory response was enhanced at week 1 post-fracture. These inflammatory cytokines involved in fracture healing were shown to coordinate different fracture healing processes such as mesenchymal stem cell recruitment and angiogenesis. Our previous study has demonstrated that ovariectomized rats exhibit lower levels of inflammatory response after fracture creation. Therefore, we report that LMHFV treatment can modulate macrophage polarization from M1 to M2 at an earlier time-point and partly restore the impaired inflammatory response in OVX bones at the early stage of fracture healing that may lead to accelerated healing of osteoporotic fracture as shown by promoted callus formation


Orthopaedic Proceedings
Vol. 104-B, Issue SUPP_12 | Pages 76 - 76
1 Dec 2022
Eltit F Ng T Gokaslan Z Fisher C Dea N Charest-Morin R
Full Access

Giant cell tumors of bone (GCTs) are locally aggressive tumors with recurrence potential that represent up to 10% of primary tumors of the bone. GCTs pathogenesis is driven by neoplastic mononuclear stromal cells that overexpress receptor activator of nuclear factor kappa-B/ligand (RANKL). Treatment with specific anti-RANKL antibody (denosumab) was recently introduced, used either as a neo-adjuvant in resectable tumors or as a stand-alone treatment in unresectable tumors. While denosumab has been increasingly used, a percentage of patients do not improve after treatment. Here, we aim to determine molecular and histological patterns that would help predicting GCTs response to denosumab to improve personalized treatment. Nine pre-treatment biopsies of patients with spinal GCT were collected at 2 centres. In 4 patients denosumab was used as a neo-adjuvant, 3 as a stand-alone and 2 received denosumab as adjuvant treatment. Clinical data was extracted retrospectively. Total mRNA was extracted by using a formalin-fixed paraffin-embedded extraction kit and we determined the transcript profile of 730 immune-oncology related genes by using the Pan Cancer Immune Profiling panel (Nanostring). The gene expression was compared between patients with good and poor response to Denosumab treatment by using the nSolver Analysis Software (Nanostring). Immunohistochemistry was performed in the tissue slides to characterize cell populations and immune response in CGTs. Two out of 9 patients showed poor clinical response with tumor progression and metastasis. Our analysis using unsupervised hierarchical clustering determined differences in gene expression between poor responders and good responders before denosumab treatment. Poor responding lesions are characterized by increased expression of inflammatory cytokines as IL8, IL1, interferon a and g, among a myriad of cytokines and chemokines (CCL25, IL5, IL26, IL25, IL13, CCL20, IL24, IL22, etc.), while good responders are characterized by elevated expression of platelets (CD31 and PECAM), coagulation (CD74, F13A1), and complement classic pathway (C1QB, C1R, C1QBP, C1S, C2) markers, together with extracellular matrix proteins (COL3A1, FN1,. Interestingly the T-cell response is also different between groups. Poor responding lesions have increased Th1 and Th2 component, but good responders have an increased Th17 component. Interestingly, the checkpoint inhibitor of the immune response PD1 (PDCD1) is increased ~10 fold in poor responders. This preliminary study using a novel experimental approach revealed differences in the immune response in GCTs associated with clinical response to denosumab. The increased activity of checkpoint inhibitor PD1 in poor responders to denosumab treatment may have implications for therapy, raising the potential to investigate immunotherapy as is currently used in other neoplasms. Further validation using a larger independent cohort will be required but these results could potentially identify the patients who would most benefit from denosumab therapy


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 75 - 75
1 Jul 2020
Algate K Cantley M Fitzsimmons T Paton S Wagner F Zannettino A Holson E Fairlie D Haynes D
Full Access

The inflammatory cascade associated with prosthetic implant wear debris, in addition to diseases such as rheumatoid arthritis and periodontitis, it is shown to drastically influence bone turnover in the local environment. Ultimately, this leads to enhanced osteoclastic resorption and the suppression of bone formation by osteoblasts causing implant failure, joint failure, and tooth loosening in the respective conditions if untreated. Regulation of this pathogenic bone metabolism can enhance bone integrity and the treatment bone loss. The current study used novel compounds that target a group of enzymes involved with the epigenetic regulation of gene expression and protein function, histone deacetylases (HDAC), to reduce the catabolism and improve the anabolism of bone material in vitro. Human osteoclasts were differentiated from peripheral blood monocytes and cultured over a 17 day period. In separate experiments, human osteoblasts were differentiated from human mesenchymal stem cells isolated from bone chips collected during bone marrow donations, and cultured over 21 days. In these assays, cells were exposed to the key inflammatory cytokine involved with the cascade of the abovementioned conditions, tumour necrosis factor-α (TNFα), to represent an inflammatory environment in vitro. Cells were then treated with HDAC inhibitors (HDACi) that target the individual isoforms previously shown to be altered in pathological bone loss conditions, HDAC-1, −2, −5 and −7. Analysis of bone turnover through dentine resorptive measurements and bone mineral deposition analyses were used to quantify the activity of bone cells. Immunohistochemistry of tartrate resistant acid phosphatase (TRAP), WST-assay and automated cell counting was used to assess cell formation, viability and proliferation rates. Real-time quantitative PCR was conducted to identify alterations in the expression of anti- and pro-inflammatory chemokines and cytokines, osteoclastic and osteoblastic factors, in addition to multiplex assays for the quantification of cytokine/chemokine release in cell supernatant in response to HDACi treatments in the presence or absence of TNFα. TNFα stimulated robust production of pro-inflammatory cytokines and chemokines by PBMCs (IL-1β, TNFα, MCP1 and MIP-1α) both at the mRNA and protein level (p < 0 .05). HDACi that target the isoforms HDAC-1 and −2 in combination significantly suppressed the expression or production of these inflammatory factors with greater efficacy than targeting these HDAC isoforms individually. Suppression of HDAC-5 and −7 had no effect on the inflammatory cascade induced by TNFα in monocytes. During osteoclastic differentiation, TNFα stimulated the size and number of active cells, increasing the bone destruction observed on dentine slices (p < 0 .05). Targeting HDAC-1 and −2 significantly reduced bone resorption through modulation of the expression of RANKL signalling factors (NFATc1, TRAF6, CatK, TRAP, and CTR) and fusion factors (DC-STAMP and β3-integerin). Conversely, the anabolic activity of osteoblasts was preserved with HDACi targeting HDAC-5 and −7, significantly increasing their mineralising capacity in the presence of TNFαthrough enhanced RUNX2, OCN and Coll-1a expression. These results identify the therapeutic potential of HDACi through epigenetic regulation of cell activity, critical to the processes of inflammatory bone destruction


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_7 | Pages 57 - 57
1 Jul 2020
Grant M Mwale F Antoniou J Bergeron S Karaplis A Panda D
Full Access

Osteoarthritis (OA) is a debilitating disease and the most common joint disorder worldwide. Although the development of OA is considered multifactorial, the mechanisms underlying its initiation and progression remain unclear. A prominent feature in OA is cartilage degradation typified by the progressive loss of extracellular matrix components - aggrecan and type II collagen (Col II). Cartilage homeostasis is maintained by the anabolic and catabolic activities of chondrocytes. Prolonged exposure to stressors such as mechanical loading and inflammatory cytokines can alter the phonotype of chondrocytes favoring cartilage catabolism, and occurs through decreased matrix protein synthesis and upregulation of catabolic enzymes such as aggrecanases (ADAMTS-) 4 and 5 and matrix metalloproteinases (MMPs). More recently, the endoplasmic reticulum (ER) stress response has been implicated in OA. The ER-stress response protects the cell from misfolded proteins however, excessive activation of this system can lead to chondrocyte apoptosis. Acute exposure of chondrocytes to IL-1β has been demonstrated to upregulate ER-stress markers (GADD153 and GRP78), however, it is unclear whether the ER-stress response plays a role on chronic IL-1β exposure. The purpose of this study was to determine whether modulating the ER stress response with tauroursodeoxycholic acid (TUDCA) in human OA chondrocytes during prolonged IL-1β exposure can alter its catabolic effects. Articular cartilage was isolated from donors undergoing total hip or knee replacement. Chondrocytes were recovered from the cartilage of each femoral head or knee by sequential digestion with Pronase followed by Collagenase, and expanded in DMEM-low glucose supplemented with 10% FBS. Chondrocytes were expanded in flasks for one passage before being prepared for micropellet culture. Chondrocyte pellets were cultured in regular growth medium (Control), medium supplemented with IL-1β [10 ng/mL], TUDCA [100 uM] or IL-1β + TUDCA for 12 days. Medium was replaced every three days. Cartilage explants were prepared from the donors undergoing knee replacement, and included cartilage with the cortical bone approximately 1 cm2 in dimension. Explants were cultured in the above mentioned media, however, the incubation period was extended to 21 days. RNA was extracted using Geneaid RNA Mini Kit for Tissue followed by cDNA synthesis. QPCR was performed using Cyber Green mastermix and primers for the following genes: ACAN (aggreacan), COL1A1, COL2A1, COL10A1, ADAMTS-4, ADAMTS-5, MMP-3, and MMP-13, on an ABI 7500 fast qPCR system. Although IL-1β did not significantly decrease the expression of matrix proteins, it did increase the expression of ADAMTS-4, −5, and MMP3 and −13 when compared to controls (Kruskal-Wallis, p < 0 .05, n=3). TUDCA treatment alone did not significantly increase the expression of catabolic enzymes but it did increase the expression of collagen type II. When IL-1β was coincubated with TUDCA, the expression of ADAMTS-4, ADAMTS-5, and MMP-13 significantly decreased by ∼40-fold, ∼10-fold, and ∼3-fold, respectfully. We provide evidence that the catabolic activities of IL-1β on human cartilage can be abrogated through modulation of the ER stress response


INTRODUCTION. Loosening is concerned to be the major cause of revision in the artificial prosthesis. Wear debris of UHMWPE dispersed into the implant-bone interface are phagocytosed by macrophages releasing inflammatory cytokines such as TNF-α which leads to osteolysis and loosening eventually. It is known that the size and structure [1] as well as attached substances on particle surface such as endotoxin could affect the amount of cytokines released [2]. An in vivo study using rat femurs showed that the presence of polyethylene particles around implants could result in accumulation of lipopolysaccharide (LPS) from exogenous sources that may affect bone remodeling around implants [3]. It is also reported that LPS is transported throughout the body with lipoproteins or LPS binding proteins [4] and Circulating LPS may originate from local sites of infection or via bloodborne bacteria [5]. In this study, we evaluated the effects of LPS that attached to UHMWPE particle surface by measuring TNF-α released from macrophages. MATERIALS AND METHODS. We cultured mouse macrophage cell line RAW 264 with spherical UHMWPE particles (8.7µm and 23µm diameter in average, Mitsui chemicals Co., LTD.) and LDPE particles (3.6µm and 5.8µm diameter in average, Sumitomo Seika Chemicals Co., LTD.) using the Inverse Culture Method for 24 hours before estimating the TNF-α generation by TNF- ALPHA QUANTIKINE ELISA KIT (R&D). Spherical UHMWPE particles (10µm diameter in average, Mitsui chemicals Co., LTD.) with E.coli original LPS (Enzo Life Sciences) attached to them were incubated with cells to see the effects of LPS on the bio-reactivity tests. REAULTS AND DISCUSSIONS. Figure 1 shows the TNF-α concentration of different materials and sizes of polyethylene particles. TNF-α concentration was shown to be dose-dependent to the total surface area of particles added regardless of the materials and sizes. Figure 2 shows TNF-α concentration relative to the particle surface area inverse and non-inverse cultured. No significant difference was observed in TNF-α concentration between particles that were attached to LPS and virgin particles in non-inverse culture method. However, when cultured inversely, the effect of LPS became more significant in higher surface area range in which dose-dependent relationship was not observed. The results suggest that saturation may occur caused by size exclusion, production limitation, etc. However, LPS attached to particle surface may alter the production limitation due to increased presence of particles around macrophages. For any figures or tables, please contact the authors directly


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_6 | Pages 32 - 32
1 Apr 2018
Zeng W Liu J Yang L
Full Access

Background. The reductions of perioperative blood loss and inflammatory response are important in total knee arthroplasty. Tranexamic acid reduced blood loss and the inflammatory response in several studies. However, the effect of epinephrine administration plus tranexamic acid has not been intensively investigated, to our knowledge. In this study, we evaluated whether the combined administration of low-dose epinephrine plus tranexamic acid reduced perioperative blood loss or inflammatory response further compared with tranexamic acid alone. Methods. This randomized placebo-controlled trial consisted of 179 consecutive patients who underwent primary total knee arthroplasty. Patients were randomized into 3 interventions: Group IV received intravenous low-dose epinephrine plus tranexamic acid, Group TP received topical diluted epinephrine plus tranexamic acid, and Group CT received tranexamic acid alone. The primary outcome was perioperative blood loss on postoperative day 1. Secondary outcomes included perioperative blood loss on postoperative day 3, coagulation and fibrinolysis parameters (measured by thromboelastography), inflammatory cytokine levels, transfusion values (rate and volume), thromboembolic complications, length of hospital stay, wound score, range of motion, and Hospital for Special Surgery (HSS) score. Results. The mean calculated total blood loss (and standard deviation) in Group IV was 348.1 ± 158.2 mL on postoperative day 1 and 458.0 ± 183.4 mL on postoperative day 3, which were significantly reduced (p < 0.05) compared with Group TP at 420.5 ± 188.4 mL on postoperative day 1 and 531.1 ± 231.4 mL on postoperative day 3 and Group CT at 520.4 ± 228.4 mL on postoperative day 1 and 633.7 ± 237.3 mL on postoperative day 3. Intravenous low-dose epinephrine exhibited a net anti-inflammatory activity in total knee arthroplasty and did not induce an obvious hypercoagulable status. Transfusion values were significantly (p=0.023 and 0.032) reduced in Group IV, but no significant differences were observed in the incidence of thromboembolic complications, wound score, range of motion, and HSS score among the 3 groups (p > 0.05). Conclusions. The combined administration of low-dose epinephrine and tranexamic acid demonstrated an increased effect in reducing perioperative blood loss and the inflammatory response compared with tranexamic acid alone, with no apparent increased incidence of thromboembolic and other complications


Orthopaedic Proceedings
Vol. 98-B, Issue SUPP_20 | Pages 48 - 48
1 Nov 2016
Albesher M Grant M Epure L Huk O Antoniou J Mwale F
Full Access

Osteoarthritis (OA) is a multifactorial disease that affects millions of Canadians. Although, there is not one specific mechanism that causes OA, the biological outcome is cartilage degradation. The articular cartilage in joints is composed primarily of the proteoglycan aggrecan and type II collagen (Col II) which together provide cartilage with functional properties. In OA, the imbalance of the anabolic and catabolic activities of chondrocytes favors cartilage catalysis. The main inflammatory cytokine involved in cartilage degradation is interleukin (IL) 1β. It has previously been demonstrated that Link N, a 16 residue peptide derived from proteolytic cleavage of link protein, can stimulate matrix proteins in normal cartilage and intervertebral discs (IVDs). Recently, we showed that a shorter sequence of Link N (sLink N), consisting of the first 8 residues of the peptide, has the potential to increase synthesis of matrix proteins in IVD cells in vitro and stimulate repair in ex vivo IVD organ culture. There are currently no treatments that actively repair cartilage in OA joints. In the present study, we aimed to evaluate the potential of sLink N as a therapeutic agent in the repair of OA cartilage. OA cartilage was isolated from four donors undergoing total knee replacement (50–70 y). Cells were recovered from the cartilage of each knee by sequential digestion with Pronase followed by Collagenase, and expanded in PrimeGrowth culture medium (Wisent Bioproducts, Canada; Cat# 319–510-CL, −S1, and −S2). After 7 days in culture, cells were treated for 24h with sLink N (0.5, 5, 50, 500 or 5000 ng/ml) or sLink N in combination with IL-1β (1 ng/ml) to mimic an inflammatory milieu. Conditioned media was collected and measured for proteoglycan (GAG) release using the safranin O and for Col II synthesis by Western blotting. Human articular cartilage explants including cartilage with subchondral bone were prepared from the same donors using the PrimeGrowth Isolation kit (Wisent, Canada) and cultured for 21 days in presence of IL-1β (1ng/ml) and sLink N (0.5, 5, 50, 500 or 5000 ng/ml). Aggrecan and Col II were extracted with guanidine buffer and measured by Western blotting. Treatment of OA chondrocytes significantly increased the GAG and Col II synthesis. The EC50 dose-response of sLink N on GAG synthesis was 67 ± 41 nM [65 ± 40 ng/ml] and the GAG synthesis reached a maximum of 194 ± 30% with the highest dose above control. When chondrocytes were cultured in the presence of IL-1β, GAG synthesis was also elevated by sLink N above control. Treatment of OA cartilage explants with sLink N increased the content of aggrecan and Col II even in the presence of IL-1β. Our results suggest that sLink N is a growth factor supplement that can increase cartilage matrix protein synthesis, and a chondroprotective agent, by modulating the catabolic effects of IL-1β. sLink N is the first small-peptide to demonstrate potential in cartilage repair of OA joints


Orthopaedic Proceedings
Vol. 95-B, Issue SUPP_7 | Pages 10 - 10
1 Feb 2013
Nunn T Horner A Pallister I
Full Access

Introduction. The ability of activated platelets to induce cellular proliferation is well recognised. In a previous diffusion model, platelets combined with Tri-calcium phosphate (TCP) led to an osteoprogenitor mitogenic response followed by cellular differentiation. This study was designed to look at osteoprogenitor responses when cultured directly onto TCP granules combined with activated platelets. Method. Human osteoprogenitors were loaded onto TCP with activated platelets at a low seeding density and high seeding densities. Cellular proliferation was assessed using the pico-green DNA content analysis. Differentiation towards osteoblastic phenotype was assessed using an alkaline phosphatase assay. RNA extraction, reverse transcription and quantitative real-time polymerase chain reaction was used to assess gene expression for type 1 collagen and osteocalcin. Histological assessment for live/dead staining and alkaline phosphatase was used on cultured granule samples. Results. In the low seeding density, platelets induced an early proliferative response compared with controls. After 14 days of culture the cells had not differentiated to an osteoblastic phenotype. When seeded at high densities, cellular differentiation was induced by the activated platelets. Significant cellular proliferation was not observed after seeding at high density. Conclusions. This study demonstrates that osteoprogenitors respond to the local environment which is modulated by both cell-cell contact factors and inflammatory cytokines from the platelets. This study helps to define the concentration of progenitors and platelets needed for further on-growth studies. It may also help define the optimal conditions for seeding cells and platelets for clinical use of composite bone graft substitutes


Orthopaedic Proceedings
Vol. 98-B, Issue SUPP_7 | Pages 6 - 6
1 May 2016
Abe S Nochi H Ito H
Full Access

Introduction. The systematic effects of joint replacement in rheumatoid arthritis (RA) patients are that inflamed synovium and pathological articular cartilage has dissipated. Expectations of total knee arthroplasty (TKA) are reduction of inflammatory cytokines, decreased disease activity and improvement of drug efficacy and ADL. Remission of rheumatoid arthritis is defined as having a Disease Activity Score DAS28 (ESR) of less than 2.6 and Health Assessment Questionnaire (HAQ) – Disability Index, less than 0.5. Purpose. We investigated whether TKA could reduce disease activity and improve ADL, and subsequent remission levels of DAS and HAQ or not. Material and Methods. We analyzed the Knee Society Score (KSS), KOOS score and DAS28 in 15 patients, 23 rheumatoid arthritic knees at pre-operation and 1 year after operation. Preoperatively patients had used non-steroidal anti-inflammatory drugs, prednisolone, disease-modifying anti-rheumatic drugs including methotrexate. TKA (Zimmer NexGen LPS Flex Knee implants and Stemmed Tibial component with stem) was performed with the modified gap technique or modified anatomical technique using original tensor with synovectomy. Results. Preoperative and postoperative KSS are 45.7±18.1 and 88.7±17.7 (P<0.01) respectively, and function scores were 40.1±21.7 and 74.8±24.0 (P<0.01) respectively (Figure 1). Preoperative and postoperative KOOS score (%) were ‘pain’ 50.6±37.8 and 95.4±19.3 (P<0.01), ‘symptom’ 56.6±32.8 and 94.7±18.6 (P<0.01), ‘ADL’ 60.6±27.9 and 89.5±32.4 (P<0.01), ‘QOL’ 28.4±32.1 and 63.6 ±22.9 (P<0.01) and ‘sport’ 20.56±29.52 and 47.10±33.9(P=0.06), respectively (Figure 2). Preoperative and postoperative DAS28(CRP) were 4.48±1.08 and 3.58±1.11(P<0.01), and DAS28 (ESR) were 4.90±1.02 and 4.13±0.99 (P=0.02) (Figure 3). Discussion. Each scores except ‘sport’ and DAS28 (ESR) improved statistically 1 year after operation. Function score, ‘ADL’ and ‘QOL’ scores also improved. HAQ score includes 2 categories related to walking ability and TKA was expected to improve the HAQ score, although the HAQ score is highly affected by upper arm function. The ‘sport’ score did not improve because almost all patients did not do sports preoperatively and postoperatively. DAS28 (ESR) and DAS28 (CRP) correlate strongly, but in this study there were statistical discrepancies in improvement. This might be because age, sex, disease duration, and existence of rheumatoid factors, anti-cyclic citrullinated peptide antibody and DLA-DRB1 shared epitope have been shown to influence ESR. DAS28 improved by a little less than 1.0, but there was limited control of disease activity. It was reported that the average DAS 28 didn't drop below 3.2 in 3 years follow-up after TKA. In this study we did not assess depending on preoperative disease activity, but it was reported that TKA had a systematic effect on severe or moderate RA activity, not low disease activity


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXI | Pages 88 - 88
1 May 2012
N.L. M A.J. H J.H. R Y. X U.G. F G.A. M I.B. M
Full Access

The cellular mechanisms of tendinopathy remain unclear, particularly with respect to the role of inflammation in early disease. We have previously identified increased levels of inflammatory cytokines in an early human model of tendinopathy and sought to extend these studies to the cellular analysis of tissue. Purpose. To characterise inflammatory cell subtypes in early human tendinopathy we explored the phenotype and quantification of inflammatory cells in torn and control tendon samples. Design. Controlled laboratory study. Methods. Torn supraspinatus tendon and matched intact subscapularis tendon samples were collected from twenty patients undergoing arthroscopic shoulder surgery. Control samples of subscapularis tendon were collected from ten patients undergoing arthroscopic stabilisation surgery. Tendon biopsies were evaluated immunohistochemically by quantifying the presence of macrophages (CD68 and CD206), T cells (CD3), mast cells (Mast cell tryptase) and vascular endothelium (CD34). Results. Subscapularis tendon biopsies obtained from patients with torn supraspinatus tendon exhibited significantly greater macrophage, mast cell and T cell expression compared to either torn supraspinatus samples or control subscapularis derived tissue (p< 0.01). Inflammatory cell infiltrate correlated inversely (r=0.5, p< 0.01) with rotator cuff tear size, with larger tears correlating with a marked reduction in all cell lineages. There was a modest but significant correlation between mast cells and CD 34 expression (r= 0.4, p< 0.01) in pre-rupture subscapularis tendon. Conclusion. We provide evidence for an inflammatory cell infiltrate in early mild/moderate human supraspinatus tendinopathy. In particular, we demonstrate significant infiltration of mast cells and macrophages suggesting a role for innate immune pathways in the events that mediate early tendinopathy. Further mechanistic studies to evaluate the net contribution and hence therapeutic utlity of these cellular lineages and their downstream processes may reveal novel therapeutic approaches to the management of early tendinopathy


Orthopaedic Proceedings
Vol. 95-B, Issue SUPP_15 | Pages 353 - 353
1 Mar 2013
Uetsuki K Sugimoto T Kawasaki T Turner A Tomita N
Full Access

Introduction. Vitamin-E (VE)-blended UHMWPE has been developed as a bearing-surface material due to the antioxidant ability of VE and has demonstrated a low wear rate in knee simulator [1]. Additionally, in vitro biological response testing has revealed that wear particles from VE blended UHMWPE induce the secretion of inflammatory cytokines at significantly lower levels compared to conventional UHMWPE [2]. However, as the joint kinematics are different between the knee and the hip, it is not guaranteed that these improvements will be repeated in the hip. In this study, the wear resistance of VE-blended UHMWPE was evaluated in knee and hip simulator tests and the effects of VE concentration and electron-beam irradiation were investigated. Materials and Methods. VE blended samples (GUR_VE xx%) were manufactured via direct compression molding following the blending of UHMWPE resin powder with VE at several concentrations (0, 0.1, 0.3, 1.0%). Cross-linking for the VE samples was achieved by 10 MeV electron beam at several irradiance doses (30, 90, 300 kGy) and annealed below the melting point of UHMWPE. Knee and hip simulator testing were carried out according to ISO 14243 and ISO 14242, respectively, and the volumetric wear was calculated. The gel fraction was determined by measuring the weight of the samples before and after soaking in decahydronaphthalene at 150°C. The oxidative resistance of the material was determined by measuring the Oxidation Index (OI) following ASTM F2102 before and after compulsory aging (ASTM2003). Radical measurements were made using high-sensitivity X-band ESR. Results & Discussion. In knee simulator testing, the volumetric wear for GUR_VE was lower than that for non-blended UHMWPE (GUR), with this difference amplified by aging. Conversely, in hip simulator testing, the wear rates were the same for the GUR and GUR_VE samples, while the GUR_XL samples showed almost no wear both before and after aging. Additionally, the wear for the non-XL samples (GUR and GUR_VE) actually decreased after aging. Treatment utilizing electron-beam irradiation and annealing below the melting point achieved a high degree of cross-linking in VE-blended UHMWPE, while also preserving the antioxidant ability of VE. These results suggest that electron-beam irradiated VE blended UHMWPE can be used as a bearing surface material for hip prostheses. However, the longevity of prosthesis is determined not only by its wear performance, but also by its biocompatibility. ESR measurements have revealed that VE radicals are formed during radiation crosslinking of VE blended UHMWPE, and it is thought that the VE radicals may reduce the anti-inflammatory effects of UHMWPE particles containing VE. We are now developing a multidirectional lift-off type aseptic wear simulation device and procedures to measure the biological response to wear particles produced therein [3]. Acknowledgement. A part of this study was supported by the “Super special consortia” for supporting the development of cutting edge medical care (Cabinet office, Government of Japan) and Innovation promotion program (NEDO)


Orthopaedic Proceedings
Vol. 95-B, Issue SUPP_34 | Pages 13 - 13
1 Dec 2013
Bechtel C Gebhart J Tatro J Schluchter M Wilkinson JM Greenfield E
Full Access

Introduction:. Wear particles cause aseptic loosening by stimulating macrophages to produce inflammatory cytokines. Recent studies indicate that Toll-like receptor 2 (TLR2) and TLR4 mediate macrophage responses to the wear particles [1–3]. TLR2 and TLR4 uniquely activate MyD88-dependent signaling via an additional adapter protein known as TIRAP/Mal [4]. Del Vescovo et al reported that three single nucleotide polymorphisms (SNPs) within the TIRAP/Mal gene associate with aseptic loosening in THA patients [5]. The goal of the current study was therefore to determine whether TIRAP/Mal mediates responses to orthopaedic wear particles. Methods:. Immortalized wild type (WT) and TIRAP/Mal knockout (KO) murine macrophages (Mfs) were incubated in the presence or absence of titanium (Ti) particles (1 × 10. 8. particles/cm. 2. [2]. Three types of particles were used as described previously [1,2]: Ti particles with adherent bacterial debris (38.3 Endotoxin Units/10. 9. particles), endotoxin-free Ti particles (<0.1 EU/10. 9. particles), and Ti particles with adherent lipopolysacharide (LPS, 32.8 EU/10. 9. particles). TNFa, IL-1b, and IL-6 mRNAs were measured by real-time PCR and the secreted cytokines were measured by ELISA. Particle-induced osteolysis in calvaria of TIRAP/Mal KO and WT mice was measured 7 days after particle implantation [1,2]. In vitro results are presented as mean ± SEM of 3–4 replicate experiments analyzed by two-way ANOVA with Bonferroni post-hoc corrections. In vivo results are presented as means of individual parietal bones ± SEM (n = 22) and analyzed by one-way ANOVA on ranks with Student Neuman-Keuls post-hoc corrections. * denotes p < 0.5, ** denotes p < 0.01, *** denotes p < 0. Results:. Ti particles with adherent bacterial debris induced substantial osteolysis and expression of TNFa, IL-1b, and IL-6 at both the mRNA and protein levels and all of those responses were significantly inhibited by TIRAP/Mal KO (Fig 1 & Fig 2). Endotoxin-free Ti particles had a small effect on osteolysis and cytokine mRNA expression that was not dependent on TIRAP/Mal (Fig 1 & data not shown). Adherence of highly purified LPS to the endotoxin-free particles reconstituted the stimulation of osteolysis and cytokine expression as well as the dependence on TIRAP/MAL (Fig 1 & data not shown). Specificity of the effects of TIRAP/Mal KO was demonstrated since responses induced by recombinant murine IL-1b were unaffected (data not shown). Discussion:. Our results are the first demonstration that TIRAP/Mal mediates effects of orthopaedic wear particles. TIRAP/Mal KO inhibited expression of TNFa by ∼50% and almost completely inhibited particle-induced osteolysis, as well as expression of IL-1b and IL-6. Our results, coupled with the genetic association of SNPs in human TIRAP/Mal with aseptic loosening [5], lead to two conclusions. First, activation of TIRAP/Mal likely contributes to aseptic loosening in patients. Second, pathogen-associated molecular patterns (PAMPs) also likely contribute to aseptic loosening since the results with endotoxin-free Ti particles demonstrate that adherent PAMPs are required for activation of TIRAP/Mal


Orthopaedic Proceedings
Vol. 98-B, Issue SUPP_9 | Pages 149 - 149
1 May 2016
Rieker C Hasgall P Ferrando M
Full Access

INTRODUCTION. Total joint arthroplasty continues to gain acceptance as the standard of care for the treatment of severe degenerative joint disease, and is considered one of the most successful surgical interventions in the history of medicine. However, infection of these implants, called Periprosthetic Joint Infection (PJI), remains one of the biggest challenges facing orthopaedics today. PJI can lead to additional surgeries, revision, fusion and amputation. Diagnosis of PJI. It is important to accurately diagnose PJI because its management differs from that of other causes of arthroplasty failure. In acute infection, the local signs and symptoms (e.g., severe pain, swelling, erythema, and warmth at the infected joint) of inflammation are generally present. On the other hand, chronic infection usually has a more subtle presentation, with pain alone, and is often accompanied by loosening of the prosthesis at the bone-implant interface. The diagnosis of PJI has proven quite challenging, as both acute and chronic infections can be difficult to differentiate from other forms of inflammation. The reported literature on the diagnosis of PJI has focused on evaluated laboratory tests that were never developed specifically for the diagnosis of PJI. These include the erythrocyte sedimentation rate (ESR), the serum C-reactive protein (CRP), the synovial fluid white blood cell count and the leukocyte differential. Because these tests were not made for the purpose of diagnosing PJI, it has been the responsibility of the orthopaedic community to evaluate and recommend their interpretation. This has resulted in significant confusion regarding the appropriate thresholds and optimal combination of these tests. These difficulties were the motivation for the development of a specific test for the detection of PJI. The Synovasure® Test for Periprosthetic Joint Infection (PJI). The promising diagnostic capabilities of synovial fluid biomarkers for PJI have already been reported in the literature. These biomarkers include inflammatory proteins, cytokines, and microbicidal peptides / proteins that are known to be involved in the host response to infection. Studies have demonstrated that the alpha-defensin microbicidal peptide present in human neutrophils is an ideal biomarker for PJI due to the distinct separation it achieves between positive and negative results. A specific test allowing to measure the concentration of the alpha-defensin in the synovial fluid has been developed. The specificity and the sensitivity of this test for the detection of a PJI are respectively 96% and 97%. This test has been proven to have also a high reproducibility, its results not being influenced by antibiotics. DISCUSSION. A lateral flow version of this test (Synovasure PJI, distributed exclusively in Europe by Zimmer GmbH) has been recently developed. It allows reading the results in 10 minutes and it doesn't require any laboratories for its interpretation. Currently, this test device is in clinical evaluation in more than 200 European hospitals. CONCLUSIONS. In case that the clinical evaluation of this test device is positive, this method will be a new paradigm for the diagnosis of periprosthetic joint infection


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

Objectives

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

Methods

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


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

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

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

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

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