header advert
Results 1 - 20 of 51
Results per page:
Orthopaedic Proceedings
Vol. 104-B, Issue SUPP_10 | Pages 29 - 29
1 Oct 2022
Ibrahim M Mah T Abdelbary H
Full Access

Introduction. Gram-negative prosthetic joint infections (GN-PJI) present unique challenges in management due to their distinct pathogenesis of biofilm formation on implant surfaces. The purpose of this study is to establish a clinically representative GN-PJI model that can reliably recapitulate biofilm formation on titanium implant surface in vivo. We hypothesized that biofilm formation on an implant surface will affect its ability to osseointegrate. Methods. The model was developed using 3D-printed titanium hip implants, to replace the femoral head of male Sprague-Dawley rats. GN-PJI was induced using two bioluminescent Pseudomonas aeruginosa strains: a reference strain (PA14-lux) and a mutant biofilm-defective strain (ΔflgK-lux). Infection was monitored in real-time using the in vivo imaging system (IVIS) and Magnetic Resonance Imaging (MRI). Bacterial loads on implant surface and in periprosthetic tissues were quantified utilizing viable-colony-count. Field-emission scanning-electron-microscopy of the explanted implants was used to visualize the biofilm formation at the bone-implant-interface. The implant stability, as an outcome, was directly assessed by quantifying the osseointegration in vitro using microCT scan, and indirectly assessed by identifying the gait pattern changes using DigiGait. TM. system in vivo. Results. Localized infection was established within the hip joint and was followed by IVIS in real-time. There was a quantitative and qualitative difference in the bacterial load and biofilm formation between PA14-lux and ΔflgK-lux. This difference in the ability to persist in the model between the two strains was reflected in the gait pattern and implant osseointegration. Conclusions. We developed a novel uncemented hip hemiarthroplasty, GN-PJI rat model. To date, the proposed in vivo biofilm-based model is the most clinically representative for GN-PJI since animals can bear weight on the implant and poor osseointegration correlates with biofilm formation. In addition, localized PJI was detected by various modalities. Clinical Relevance. The proposed in vivo GN-PJI model will allow for more reliable testing of novel biofilm-targeting therapeutics


Orthopaedic Proceedings
Vol. 104-B, Issue SUPP_10 | Pages 19 - 19
1 Oct 2022
Schenk HM Sebillotte M Lomas J Taylor A Benavent E Murillo O Fernandez-Sampedro M Huotari K Aboltins C Trebse R Soriano A Wouthuyzen-Bakker M
Full Access

Aim. Patients with late acute periprosthetic joint infections (PJI) and treated with surgical debridement have a high failure rate. Previous studies have shown that rheumatoid arthritis (RA) is an independent risk factor for treatment failure. We conducted a case-control study to identify predictors for failure in late acute PJI treatment in RA patients. We hypothesize that patients with RA have a higher failure rate compared to controls due to the use of immunosuppressive drugs. Method. Data of an international multicenter retrospective observational study was used. Late acute PJI was defined as a sudden onset of symptoms and signs of a PJI, more than 3 months after implantation. Failure of treatment was defined as persistent signs of infection, relapse with the same or reinfection with a different micro-organism, need for prosthesis removal or death. Cases with RA were matched with cases without RA based on the affected joint. A Cox survival analyses, stratified for RA, was used to calculate hazard ratio's (HR) for failure. Subgroup analyses were used to explore other predictors for treatment failure in RA patients. Results. A total of 40 patients with RA and 80 controls without RA were included. Treatment failure occurred in 65% patients with RA compared to 45% for controls (p= .052). 68% of patients with RA used immunosuppressive drugs at time of PJI diagnosis. The use or continuation of immunosuppressive drugs in PJI was not associated with a higher failure rate; neither were the duration of symptoms and causative microorganism. The time between implantation of the prosthetic joint and diagnosis of infection was longer in RA patients: median 110 (IQR 41-171) vs 29 months (IQR 7.5–101.25). Exchange of mobile components was associated with a lower risk of treatment failure (HR 0.489, 95% CI 0.242–0.989, p-value .047). Conclusions. The use of immunosuppressive drugs does not seem to be associated with a higher failure rate in patients with RA. Mobile exchange in RA patients is associated with a lower risk of failure. This might be due to the significantly older age of the prosthesis in RA patients. Future studies are needed to explore these associations and its underlying pathogenesis


Orthopaedic Proceedings
Vol. 104-B, Issue SUPP_12 | Pages 16 - 16
1 Dec 2022
Ibrahim M Abdelbary H Mah T
Full Access

Gram-negative prosthetic joint infections (GN-PJI) present unique challenges in management due to their distinct pathogenesis of biofilm formation on implant surfaces. To date, there are no animal models that can fully recapitulate how a biofilm is challenged in vivo in the setting of GN-PJI. The purpose of this study is to establish a clinically representative GN-PJI in vivo model that can reliably depict biofilm formation on titanium implant surface. We hypothesized that the biofilm formation on the implant surface would affect the ability of the implant to be osseointegrated. The model was developed using a 3D-printed, medical-grade titanium (Ti-6Al-4V), monoblock, cementless hemiarthroplasty hip implant. This implant was used to replace the femoral head of a Sprague-Dawley rat using a posterior surgical approach. To induce PJI, two bioluminescent Pseudomonas aeruginosa (PA) strains were utilized: a reference strain (PA14-lux) and a mutant strain that is defective in biofilm formation (DflgK-lux). PJI development and biofilm formation was quantitatively assessed in vivo using the in vivo imaging system (IVIS), and in vitro using the viable colony count of the bacterial load on implant surface. Magnetic Resonance Imaging (MRI) was acquired to assess the involvement of periprosthetic tissue in vivo, and the field emission scanning electron microscopy (FE-SEM) of the explanted implants was used to visualize the biofilm formation at the bone-implant interface. The implant stability, as an outcome, was directly assessed by quantifying the osseointegration using microCT scans of the extracted femurs with retained implants in vitro, and indirectly assessed by identifying the gait pattern changes using DigiGaitTM system in vivo. A localized prosthetic infection was reliably established within the hip joint and was followed by IVIS in real-time. There was a quantitative and qualitative difference in the bacterial load and biofilm formation between PA14 and DflgK. This difference in the ability to persist in the model between the two strains was reflected on the gait pattern and implant osseointegration. We developed a novel uncemented hip hemiarthroplasty GN-PJI rat model. This model is clinically representative since animals can bear weight on the implant. PJI was detected by various modalities. In addition, biofilm formation correlated with implant function and stability. In conclusion, the proposed in vivo GN-PJI model will allow for more reliable testing of novel biofilm-targeting therapetics


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. 105-B, Issue SUPP_17 | Pages 77 - 77
24 Nov 2023
Oehen L Morgenstern M Wetzel K Goldenberger D Kühl R Clauss M Sendi P
Full Access

Aim. One of the surgical therapeutic options for periprosthetic joint infection (PJI) includes debridement, antibiotics, and implant retention (DAIR). Prognostically favorable criteria for DAIR include short duration of symptoms, stable implant, pathogen susceptible to a ‘biofilm-active’ antimicrobial agent, and intact soft-tissue conditions. Despite this, there is a proportion of failures after DAIR, possibly because the duration of infection is underestimated. With the hypothesis that the duration of infection correlates with the bacterial load, and hence, the bacterial load is associated with failure after DAIR, we aimed to investigate the association of bacterial load in the sonication fluid of mobile parts and clinical outcome after DAIR. Method. From our PJI cohort (2010–2021), patients with DAIR (both palliative and curative approaches) were reviewed retrospectively. Patients with hip, knee or shoulder arthroplasties fulfilling infection definition, available sonication results, and ≥2 years follow-up were included. Sonication results were categorized in ≤ or >1000 cfu/mL. Univariate analysis was performed to identify predictors for DAIR failure. Results. Out of 209 PJIs, we identified 96 patients (100 PJIs, 47.8%) with DAIR. In 67 (69.8%) patients with 71 PJIs, there was a follow-up of ≥2 years. The mean age was 72.7 (SD 12.99) years, 50% were male. The infection affected 36 hips (50.7%), 32 knees (45.1%) and 3 shoulders (4.2%). At follow-up, there were 29 (40.8%) cured and 42 (59.2%) failed cases. When comparing failed and cured cases, we found no difference in comorbidities and previously defined risk factors for PJI, ASA score, Charlson score, anatomic location, no. of previous surgeries, pathogenesis of infection or laboratory values. The proportion of patients with high bacterial load on mobile parts (i.e. >1000 cfu/mL) was significantly higher in the failed DAIR group than it was in the cured group (61.9% vs 20.7%, p<0.001). Conclusions. In this study, a high bacterial load in sonication fluid of mobile parts was associated with failure after DAIR in patients with PJI. Sonication may help to differentiate acute hematogenous seeding to the implant and late reactivation of a previously silent implant-associated infection


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 89 - 89
1 Jul 2020
Chua K
Full Access

Osteosarcoma is the most common primary bone tumour worldwide. This disease presents a formidable challenge to the orthopaedic surgeon, with a mortality rate of 30 per cent, even after surgical clearance. Aberrant Wnt signalling has been implicated in the pathogenesis osteoblastic tumours. The objective of this study is 2 fold- to investigate if osteosarcoma does indeed demonstrate aberrant Wnt signaling, and if so, does osteosarcoma respond to a novel Wnt inhibitor(ETC159). This can potentially lead to the development of a new adjuvant treatment modality for osteosarcoma. A novel Wnt signaling pathway protein antibody (YJ5) was used in immunihistochemistry staining of clinical osteosarcoma samples. A Wnt high osteosarcoma cell line(SJSA-1) was then implanted subcutaneously in a mouse model. These mice were treated with a novel PORCN inhibitor, ETC 159 for a period of 4 weeks in a two-arm randomised control study. The results of treatment were evaulated by clinical outcome parameters as well as immunohstochemistry. 100 per cent of clinical osteosarcoma samples demonstrated increased WLS expression and Wnt protein expression. SJSA-1 showed no significant decrease in tumour volume after 30 days of drug treatment (3070 SD 625 mm3 vs 3480 SD 433 mm3 p= 0.605 and 2060 SD 209 vs 1677 SD 213 mm3 p=0.219 respectively). Significantly, SJSA-1 demonstrated increased tumour necrosis in the treatment arm(30–60 percent increase across all samples p < 0 .005) Treated tumours also demonstrated markedly less angiogenesis compared to the non treatment arm. Osteosarcoma demonstrates aberrant Wnt signaling in a large percentage of cases. The use of a novel PORCN inhibitor ETC 159 for the treatment of Osteosarcoma has a marked effect on tumour necrosis. Our results suggest that ETC159 may cause tumour necrosis by inhibiting angiogenesis within the tumour. Further evaluation and understanding of the mechanism of Wnt singaling in regulating tumour pathogenesis may hold the potential for developing a curative therapeutic drug for this deadly disease


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_15 | Pages 26 - 26
1 Dec 2021
Lang S Loibl M Gläsner J Simon M Rupp M Grad S Neumann C Alt V Gessner A Hanses F
Full Access

Aim. Vertebral osteomyelitis (VO) is an infection of the spine mostly caused by bacterial pathogens. The pathogenesis leading to destruction of intervertebral discs (IVD) and adjacent vertebral bodies (VB) is poorly described. We aimed to investigate the connection between infection, bone- and disc-metabolism in VO patients. Method. Fourteen patients with VO (infection group) and 14 patients with incomplete burst fractures of the spine (fracture group as controls) were included prospectively. Demographic data, treatment details, laboratory infection markers, and patient-reported outcome were assessed. Tissue biopsies from affected IVDs and adjacent VBs were analyzed for mRNA-expression levels of 18 target genes including chemokines, adipokines and genes involved in bone-metabolism by RT-qPCR. Results. The Receptor activator of NF-κB/Osteoprotegerin (RANK/OPG) expression ratio was elevated in VB and IVD of the infection group (p<0.001 and p=0.028, respectively). The RANK-ligand (RANKL)/OPG expression ratio was elevated in VB of the infection group (p<0.01). Expressions of the chemokines IL8 and CCL20 were higher in VB samples of the infection group. The expression of leptin was higher in IVD tissue, the mRNA expression of omentin and resistin was lower in VBs of the infection group. OPG mRNA expression was lower in infected VB and in IVD tissue compared to the fracture group. Conclusions. We identified similar expression patterns of pro-inflammatory cytokines and the RANK/RANKL/OPG axis in VBs and IVDs of patients with VO. This finding suggests that common immuno-metabolic pathways are involved in mechanisms leading to tissue degradation in VBs and IVDs during VO


Orthopaedic Proceedings
Vol. 101-B, Issue SUPP_8 | Pages 39 - 39
1 May 2019
Sharkey P
Full Access

Bone marrow lesions (BMLs), identified by MRI, are defined as a region of cancellous bone with high T2 and low T1 signal intensity. They are associated with various knee pathologies including spontaneous osteonecrosis of the knee (SPONK), AVN, trauma (fracture and bone contusion), following arthroscopy and secondary to overuse (i.e., after completing a marathon). They also are commonly recognised in patients with knee OA (referred to as OA-BMLs) and their substantial importance in knee OA pathogenesis has been recently identified. Depending upon the etiology (i.e., bone contusion, overuse, etc.) of the BML, these lesions can be “acute” in nature and spontaneously resolve over time. However, OA-BMLs generally are considered to be a “chronic” condition and overtime they have been shown to often persist and increase in size. Retrieval studies following THA and TKA, in patients with a preoperatively identified BML, have greatly expanded our understanding of OA – BMLs and these investigations consistently identify the critical role subchondral bone plays in OA disease progression. Histologic, histochemical and mechanical studies of OA-BMLs demonstrate significant alternations from healthy subchondral bone. The effected bone contains regions where fibrous tissue has replaced cancellous bone, microfractures are present and vascularity is increased. There is an increased concentration of inflammatory mediators and the bone structural integrity is compromised. Standard radiographs of the knee correlate only modestly with patient symptoms, but conversely, the presence of an OA-BML is an extremely strong predictor of pain and knee joint dysfunction. Felson et al. reported this relationship. In a large group of patients with painful knee OA, 77.5% of these patients had a BML. Both the presence and size of the BML, following multiregression analysis, were significant predictors of knee pain severity. Additionally, likely secondary to inadequate subchondral bone plate support, the presence of an OA-BML is associated with subchondral bone attrition (SBA). SBA leads to collapse of the subchondral bone plate and progressive joint deformity. Based on the association of an OA-BML with pain, joint dysfunction and deformity, it is not surprising that these lesions are prognostic for patients seeking knee arthroplasty. Several studies have demonstrated that the odds of knee arthroplasty performance are substantially higher in patents with an OA-BML. This enhanced understanding of knee OA pathogenesis and the critical role of subchondral bone in this process creates an opportunity for development of novel prevention and treatment strategies. Prevention of OA-BML formation has been considered and pharmacologic interventions proposed. Recent studies have reported positive results for treatment with bisphosphonates in patients with knee OA. One study reported significant pain and OA-BML size reduction in patients receiving a bisphosphonate for 4 months. A strategy aimed at repairing and/or enhancing subchondral bone compromised by an OA-BML has also been proposed. Early results reported with this intervention are encouraging, but preliminary


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_7 | Pages 36 - 36
1 Jul 2020
Lian WS Wang F Hsieh CK
Full Access

Aberrant infrapatellar fat metabolism is a notable feature provoking inflammation and fibrosis in the progression of osteoarthritis (OA). Irisin, a secretory subunit of fibronectin type III domain containing 5 (FNDC5) regulate adipose morphogenesis, energy expenditure, skeletal muscle, and bone metabolism. This study aims to characterize the biological roles of Irisin signaling in an infrapatellar fat formation and OA development. Injured articular specimens were harvested from 19 patients with end-stage knee OA and 11 patients with the femoral neck fracture. Knee joints in mice that overexpressed Irisin were subjected to intra-articular injection of collagenase to provoke OA. Expressions of Irisin, adipokines, and MMPs probed with RT-quantitative PCR. Infrapatellar adiposity, articular cartilage damage, and synovial integrity verified with histomorphometry and immunohistochemistry. Infrapatellar adipose and synovial tissues instead of articular cartilage exhibited Irisin immunostaining. Human OA specimens showed 40% decline in Irisin expression than the non-OA group. In vitro, the gain of Irisin function enabled synovial fibroblasts but not chondrocytes to display minor responses to the IL-1β provocation of MMP3 and MMP9 expression. Of note, Irisin signaling reduced adipogenic gene expression and adipocyte formation of mesenchymal progenitor cells. In collagenase-mediated OA knee pathogenesis, forced FNDC5 expression in articular compromised the collagenase-induced infrapatellar adipose hypertrophy, synovial hypercellularity, and membrane hyperplasia. These adipose-regulatory actions warded off the affected knees from cartilage destruction and gait aberrance. Likewise, intra-articular injection of Irisin recombinant protein mitigated the development of infrapatellar adiposity and synovitis slowing down the progression of cartilage erosion and walking profile irregularity. Affected joints and adipocytes responded to the Irisin recombinant protein treatment by reducing the expressions of cartilage-deleterious adipokines IL-6, leptin, and adiponectin through regulating PPAR&gamma, function. Irisin dysfunction is relevant to the existence of end-stage knee OA. Irisin signaling protects from excessive adipogenesis of mesenchymal precursor cells and diminished inflammation and cartilage catabolism actions aggravated by adipocytes and synovial cells. This study sheds emerging new light on the Irisin signaling stabilization of infrapatellar adipose homeostasis and the perspective of the therapeutic potential of Irisin recombinant protein for deescalating knee OA development


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 26 - 26
1 Jul 2020
Lemirre T Richard H Janes J Laverty S Fogarty U Girard C Santschi E
Full Access

Juvenile Osteochondritis dissecans (JOCD) in humans and subchondral cystic lesions (SCL) in horses (also termed radiolucencies) share similarities: they develop in skeletally immature individuals at the same location in the medial femoral condyle (MFC) and their etiology is only partially understood but trauma is suspected to be involved. JOCD is relatively uncommon in people whereas SCLs arise in 6% of young horses leading to lameness. Ischemic chondronecrosis is speculated to have a role in both osteochondrosis and SCL pathogenesis. We hypothesize that MFC radiolucencies develop very early in life following a focal internal trauma to the osteochondral junction. Our aims were to characterize early MFC radioluciencies in foals from 0 to 2 years old. Distal femurs (n=182) from Thoroughbred horses (n=91, 0–2 years old), presented for post-mortem examination for reasons unrelated to this study, were collected. Radiographs and clinical tomodensitometry were performed to identify lesions defined as a focal delay of ossification. Micro-tomodensitometry (m-CT) and histology was then performed on the MFCs (CT lesions and age-matched subset of controls). Images were constructed in 3D. The thawed condyles, following fixation, were sectioned within the region of interest, determined by CT lesion sites. Hematoxylin eosin phloxin and safran (HEPS) and Martius-Scarlet-Blue (MSB) stains were performed. Histological parameters assessed included presence of chondronecrosis, fibrin, fibroplasia and osteochondral fracture. An additional subset of CT control (lesion-free) MFCs (less 6 months old) were studied to identify early chondronecrosis lesions distant from the osteochondral junction. One MFC in clinical CT triages controls had a small lesion on m-CT and was placed in the lesion group. All m-CT and histologic lesions (n=23) had a focal delay of ossification located in the same site, a weight bearing area on craniomedial condyle. The youngest specimen with lesions was less than 2 months old. On m-CT 3D image analysis, the lesions seemed to progressively move in a craniolateral to caudomedial direction with advancing age and development. Seventy-four percent (n=17/23) of the lesions had bone-cartilage separation (considered to be osteochondral fractures) confirmed by the identification of fibrin/clot on MSB stains, representing an acute focal bleed. Fibroplasia, indicating chronicity, was also identified (74%, n=17/23). In four cases, the chondrocytes in the adjacent cartilage were healthy and no chondronecrosis was identified in any sections in the lesions. Nineteen cases had chondronecrosis and always on the surface adjacent to the bone, at the osteochondral junction. None of the subset of control specimens, less than 6 months old (n=44), had chondronecrosis within the growth cartilage. Early subchondral cystic lesions of the medial femoral condyle may arise secondary to focal internal trauma at the osteochondral junction. The presence of fibrin/clot is compatible with a recent focal bleed in the lesion. Medial femorotibial joint internal forces related to geometry could be the cause of repetitive trauma and lesion progression. In the juvenile horse, and potentially humans, the early diagnosis of MFC lesions and rest during the susceptible period may reduce progression and promote healing by prevention of repetitive trauma, but requires further study


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 102 - 102
1 Jul 2020
Mosley G Nasser P Lai A Charen D Evashwick-Rogler T Iatridis J
Full Access

Low back pain is more common in women than men, yet most studies of intervertebral disc (IVD) degeneration do not address sex differences. In humans, there are sex differences in spinal anatomy and degenerative changes in biomechanics, and animal models of chronic pain have demonstrated sex differences in pain transduction. However, there are few studies investigating sex differences in annular puncture IVD degeneration models. IVD puncture is known to result in progressive biomechanical alterations, but whether these IVD changes correlate with pain is unknown. This study used a rat IVD injury model to determine if sex differences exist in mechanical allodynia, biomechanics, and the relationship between them, six weeks after IVD injury. Procedures were IACUC approved. 24 male & 24 female four-month-old Sprague-Dawley rats underwent a sham or annular puncture injury surgery (n=12 male, 12 female). In injury groups, three lumbar IVDs were each punctured three times with a needle, and injected with tumor necrosis factor-alpha. Mechanical allodynia was tested biweekly using von Frey filaments. Six weeks after IVD injury, rats were euthanized and motion segments were dissected for non-destructive axial tension-compression and torsional rotation biomechanical testing. Two-way ANOVA with Bonferroni corrections identified statistically significant differences (p < 0 .05) and correlations used Pearson's coefficient. Annular puncture injury induced a significant increase in mechanical allodynia compared to sham in male but not female rats up to six weeks after injury. There was a significant sex effect on both torque range and torsional stiffness, with males exhibiting greater stiffness and torque range than females. Tensile stiffness, compressive stiffness, and axial range of motion showed no sex difference. Males and females showed similar patterns of correlation between variables when sham and injury groups were analyzed together, but correlations were stronger in males. Most correlations were clustered within testing approach: axial biomechanics negatively correlated, torsional biomechanics positively correlated, and von Frey thresholds positively correlated. Surprisingly, mechanical allodynia did not correlate with any biomechanics after injury, and the axial and torsional biomechanics showed little correlation. This study demonstrates that males and females respond to IVD injury differently. Given the absence of correlation between pain and biomechanics, pain cannot be attributed completely to biomechanical changes. This may explain why spinal fusion surgery, an intervention limited to the spine, has produced inconsistent results and is controversial for patients with low back pain. Thus, in addressing low back pain, we must consider both spinal tissues and the nervous system. Further, the limited correlation between axial and torsional biomechanics indicates that IVD injury may have distinct effects on nucleus pulposus and annulus fibrosus. Biomechanics did not differ between sham and injury at week six, suggesting healing after injury. It remains possible that acute biomechanical changes may initiate chronic pain pathogenesis. We conclude that the observed sex differences demonstrate the need for inclusion of both males and females in IVD injury and pain studies, and suggest that males and females may require different treatments for conditions that appear similar


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 123 - 123
1 Jul 2020
J.Dixon S Beaucage K Nagao M Lajoie G Veras M Fournier D Holdsworth D Bailey C Hammond J Séguin C
Full Access

Equilibrative nucleoside transporter 1 (ENT1) transfers nucleosides, such as adenosine, across plasma membranes. We reported previously that mice lacking ENT1 (ENT1-KO) exhibit progressive ectopic calcification of spinal tissues, including the annulus fibrosus (AF) of intervertebral discs (J Bone Miner Res 28:1135–49, 2013, Bone 90:37–49, 2016). Our purpose was twofold: (1) to compare ectopic calcifications in ENT1-KO mice with those in human DISH, and (2) to investigate the molecular pathways underlying pathological calcification in ENT1-KO mice. Studies were performed with age-matched wild-type (WT) and ENT1-KO mice, as well as human cadaveric vertebral columns meeting radiographic criteria for DISH. Mouse and human specimens were scanned using high-resolution, micro-computed tomography (micro-CT). As well, some samples were decalcified and processed for histological assessment. Calcified lesions in selected specimens were examined using energy dispersive X-ray spectroscopy (EDX) and X-ray diffraction (XRD). To investigate molecular changes associated with ectopic calcification, we isolated AF tissue from thoracic intervertebral discs of WT and ENT1-KO mice. Tissues were then subjected to transcriptomic and proteomic analyses. Micro-CT of ENT1-KO mice revealed ectopic calcification of spinal tissues, first appearing in the cervical-thoracic region and extending caudally with advancing age. Histological examination of calcified lesions in mice revealed accumulations of amorphous, eosinophilic, acellular material in paraspinal ligaments and entheses, intervertebral discs, mandibular symphysis, and sternocostal articulations. There was no evidence of inflammation associated with these lesions. EDX of calcified lesions revealed a high content of calcium and phosphorus in a molar ratio of ∼1.6, with hydroxyapatite detected by micro-XRD. Ten human cadaveric spines (three females and seven males, mean age 81 years) that met radiographic criteria for DISH were analysed in detail by micro-CT. Remarkable heterogeneity in the density and morphology of ectopic calcifications was observed. Analyses of calcifications by EDX and XRD again yielded a calcium/phosphorus ratio of ∼1.6 and a crystalline diffraction pattern matching hydroxyapatite. Histological examination of human lesions revealed regions of mature ossification and other areas of irregular amorphous calcification that resembled lesions in ENT1-KO mice. Microarray analysis of AF tissue from WT and ENT1-KO mice showed extensive dysregulation of transcription in affected tissues. Cell cycle-associated transcripts were the most affected, including the E2f family of transcription factors and proliferating cell nuclear antigen. In addition, expression of genes involved in the regulation of mineralization and bone development were dysregulated. Proteomic analyses confirmed transcriptomic changes and revealed alterations in known modulators of biomineralization such as matrix Gla-protein. Many of the characteristics of ectopic calcification in ENT1-KO mice resemble those of DISH in humans. Human lesions were found to be heterogeneous with regions of pathological ossification and amorphous calcification, the latter resembling lesions in the mouse model. Our studies of the molecular events associated with ectopic calcification in ENT1-KO mice may provide insights into the pathogenesis of DISH in humans. ENT1-KO mice may also be useful for evaluating therapeutics for the prevention of ectopic calcification in DISH and related disorders


Orthopaedic Proceedings
Vol. 102-B, Issue SUPP_6 | Pages 63 - 63
1 Jul 2020
Zhang J Zhao G Li F Wang JH
Full Access

Tendinopathy is one of the most common orthopaedic pathological conditions characterized by tendon degenerative changes. Excessive mechanical loading is considered as a major causative factor in the development of tendinopathy, but the mechanisms of pathogenesis remain unclear. High mobility group box-1 (HMGB1), a potent inflammatory mediator when released into the matrix, has been identified in the early stage tendinopathy patients. Since the release and contribution of HMGB1 in tendinopathy development due to mechanical overloading is unknown, we investigated the role of HMGB1 in tendinopathy using a mouse intensive treadmill running (ITR) model and injection of glycyrrhizin (GL), a specific inhibitor of HMGB1. A total of 48 mice were divided into four groups, Cage Control group: The animals were allowed to move freely in their cage, GL group: The animals were received daily IP injection of GL (50 mg/kg body weight) for 24 weeks, ITR group: The animals ran on treadmill at 15 meters/min for three h/ day, five days a week for 12 or 24 weeks, GL+ITR group: The animals ran the same protocol as that of ITR group plus daily IP injection of GL for 12 or 24 weeks. Six mice/group were sacrificed at 12 or 24 weeks and the Achilles and patellar tendon tissues were harvested and used for histochemical staining and immunostaining. Mechanical overloading induced HMGB1 released from the cell nuclei to the matrix (Fig. 1a, b) caused tendon inflammation (Fig. 1c, d) and led to tendon degenerative changes (Fig. 1e-j). After 12 weeks of ITR, the tendon tissue near the bone insertion site showed typical tendinopathic changes in cell shape, accumulation of glycosaminoglycans (GAG) (Fig. 1e, f), and increase in SOX-9 staining (Fig. 1g-j). After 24 weeks ITR, the distal site of Achilles tendon showed considerable changes in cell shape (Fig. 2A, g, arrows), which is round compared to more elongated in the control and GL groups (Fig. 2A, e, f). However, daily treatment with GL prior to ITR blocked the cell shape change (Fig. 2A, h) and, ITR induced extensive GAG accumulation in ITR group (Fig. 2B, bottom panel). Furthermore, GL inhibited ITR-induced expression of chondrogenic markers (SOX-9 and collagen II) in the tendons (Fig. 3). Our results showed that mechanical overloading-induced HMGB1 plays a critical role in the development of tendinopathy by initiating tendon inflammation and eventual degeneration characterized by the presence of chondrocyte-like cells, accumulation of proteoglycans, high levels of collagen type II production, and chondrogenic marker SOX-9 expression. These results provide the first evidence for the role of HMGB1 as a therapeutic target to prevent tendinopathy before its onset and block further development at its early inflammation stages. The inhibition of tendinopathy development by GL administration in this study also suggests the putative therapeutic potential of this natural triterpene that is already in clinical use to treat other inflammation-related diseases. For any figures or tables, please contact authors directly


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXIII | Pages 137 - 137
1 May 2012
Zheng M
Full Access

Autologous cell therapy using stem cells and progenitor cells is considered to be a popular approach in regenerative medicine for the repair and regeneration of tissue and organs. In orthopaedic practice, autologous cell therapy has become a major focus, particularly, as a feasible treatment for tendon injury. Tendons are dense connective tissue that bridge bone to muscle and transmit forces between muscle and bone to maintain mechanical movement. Tendons are poorly vascularised and have very little capacity to self-regenerate. Degeneration of tendon is often caused by injury. The pathogenesis of tendon injury, commonly known as tendinosis, is not an inflammatory condition but is secondary to degenerative changes, including disruption of the collagen matrix, calcification, vascularisation and adipogenesis. The aetiology of tendinosis is considered to be multifactorial and the pathogenesis is still unclear. Intrinsic factors such as a lack of blood and nutrition supply and extrinsic factors such as acute trauma and overuse injury caused by repetitive strain, have been implicated as contributors to the pathogenesis of tendinosis. More recent studies suggest that programmed tendon cell death (tenocyte apoptosis) may play a major role in the development of tendinosis. Such cellular abnormalities may influence the capacity of tendon to maintain its integrity. Traditional treatments such as anti-inflammatory drugs, steroid injections and physiotherapy are aimed at symptom relief and do not address the underlying pathological changes of degeneration. Here, we propose that autologous cell therapy may be an innovative and promising treatment for tendon injury. We will present evidence that suggest that autologous tendon cell therapy may be feasible to repair and regenerate tendon. We will also present data summarising the preclinical evaluation of autologous tendon cell therapy in animal models and the safety and tolerability of autologous tendon cell therapy in humans in studies, which are currently conducted at the Centre for Orthopaedic Research at the University of Western Australia


Orthopaedic Proceedings
Vol. 98-B, Issue SUPP_23 | Pages 13 - 13
1 Dec 2016
Hackl S Hellinger L Von Rüden C Friederichs J Bühren V Perl M Hierholzer C
Full Access

Aim. The pathogenesis of non-union is multifactorial. Path biological factors, mechanical factors, and low-grade-infection contribute to impaired bone healing. Aim of this study was to determine the rate of low-grade-infection in patients with long bone non-union of the lower extremity without signs of acute infection, the influence of CRP (C-reactive protein), and the outcome. Method. In a retrospective study (2003–2013), all patients who underwent surgery for treatment of tibial- or femoral-shaft-non-union without any clinical evidence of infection were assessed. Bacterial cultures harvested during non-union revision, the CRP and WBC (white blood cells) values at hospital admission, the outcome, and epidemiological data were analysed. Results. In 88 patients with tibial-shaft-non-union without any clinical signs of infection, bacterial samples remained negative in 51 patients (46 yr; 33% open fracture; 33% nicotine abuse; 8% diabetes mellitus; revision of non-union 10.9 months following primary osteosynthesis). In 37 patients (46 yr; 54% open fracture; 42% nicotine abuse; 11% diabetes mellitus; revision of non-union 15.2 months) microbiological diagnostic studies after long-term-culturing demonstrated positive bacterial cultures whereas after short-term-culturing for 2 days only 17 positive cultures were observed. Among patients with negative bacterial cultures bone healing was achieved after 13.2 months, whereas in 29% additional surgical interventions (1.3 procedures) were necessary. Non-union with positive bacterial cultures required 22.9 months (p-value<0.01) until bone healing, and even 57% of these patients required additional operations (2.9 procedures; p-value<0,01). Hematological studies performed at hospital admission demonstrated no significant difference regarding CRP (negative vs. positive culture: 0.8 mg/dl vs. 1.9 mg/dl) and WBC (negative vs. positive culture: 7.6/nl vs. 7.8/nl). Comparable results were observed in 86 patients with femoral-shaft-non-union (38 patients with positive bacterial cultures after long-term-culturing and 18 patients after short-term-culturing) with an increased number of required operations (0.8 vs. 1.6 procedures; p-value<0.05) and a longer time period until bone healing (18.2 months vs. 27.2 months; p-value<0.05) in the group with positive bacterial cultures. In contrast to tibial-shaft-non-union, a significant difference of the CRP level was observed (negative vs. positive culture: 0.8 mg/dl vs. 2.7 mg/dl; p-value<0.01). Conclusions. The pathogenesis of non-union may originate from low-grade-infection even in patients without any signs of infection and may result in increased number of required surgical interventions. Therefore, during any non-union revision surgery, multiple bacterial samples should be harvested for long-term-culturing. Possibly, increased CRP levels may be a predictor for low-grade-infection in femoral – but not in tibial-shaft-non-union


The Bone & Joint Journal
Vol. 106-B, Issue 6 | Pages 632 - 638
1 Jun 2024
Hart CM Kelley BV Mamouei Z Turkmani A Ralston M Arnold M Bernthal NM Sassoon AA

Aims

Delayed postoperative inoculation of orthopaedic implants with persistent wound drainage or bacterial seeding of a haematoma can result in periprosthetic joint infection (PJI). The aim of this in vivo study was to compare the efficacy of vancomycin powder with vancomycin-eluting calcium sulphate beads in preventing PJI due to delayed inoculation.

Methods

A mouse model of PJI of the knee was used. Mice were randomized into groups with intervention at the time of surgery (postoperative day (POD) 0): a sterile control (SC; n = 6); infected control (IC; n = 15); systemic vancomycin (SV; n = 9); vancomycin powder (VP; n = 21); and vancomycin bead (VB; n = 19) groups. Delayed inoculation was introduced during an arthrotomy on POD 7 with 1 × 105 colony-forming units (CFUs) of a bioluminescent strain of Staphylococcus aureus. The bacterial burden was monitored using bioluminescence in vivo. All mice were killed on POD 21. Implants and soft-tissue were harvested and sonicated for analysis of the CFUs.


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_17 | Pages 57 - 57
1 Dec 2018
Peng SH Lin YC
Full Access

Aim. As the populations of patients who have multiple prosthetic joints increase these years, the fate of a single joint periprosthetic joint infection in these patients is still unknown. Risk factors leading to a subsequent infection in another prosthetic joint are unclear. Our goal is to identify the risk factors of developing a subsequent infection in another prosthetic joint and describe the organism profile to the second prosthetic infection. Method. We performed a retrospective cohort study of all PJI cases underwent surgical intervention at our institute, a tertiary care referral center over 11 years, during January 2006 to December 2016. We identified 96 patients with periprosthetic joint infection who had another prosthetic joint in place at the time of presentation. The comorbidity, number of prosthetic joints, date and type of each arthroplasty, times of recurrent infection at each prosthetic joint with subsequent debridement or 2-stage resection arthroplasty, organisms from every infection episode, the outcome of each periprosthetic joint infection in these patients were analyzed. Results. During January 2006 to May 2017, we retrospective collected 294 PJI cases (159 hips, 135 knees) in our institute. Patients with single prosthetic joint were excluded and finally 96 patients were included. Of the 96 patients, 19 (19.79%) developed a periprosthetic joint infection in a second joint. The type of organism was the same as the first infection in 12 (63.16%) of 19 patients. The time to developing a second infection averaged 2.16 years (range, 0–9.3 years). The risk factors leading to a subsequent infection in another prosthetic joint are albumin level (< 3.5 mg/dl), long-term steroid usage (> 5mg/day, > 3 months), history of necrotizing fasciitis, history of invasive dental procedure (> Grade IV procedure), 3-stage resection arthroplasty or more, and PJI caused by vacomycin-resistent enterococcus (VRE). Conclusions. A PJI might predispose patients to subsequent PJI in another prosthesis. Patients and surgeons must be aware of the risk factors contribute to this devastating complication. Most organisms in the second PJI are identical to the first one, and we believe the bacteremia may be the pathogenesis, but need further proved. The preventive policy may be needed in the future for this population who has multiple prosthetic joints


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_17 | Pages 10 - 10
1 Dec 2018
Renz N Trebse R Akgün D Perka C Trampuz A
Full Access

Aim. Treatment of enterococcal periprosthetic joint infections (PJI) is challenging due to heterogeneous pathogenesis, non-standardized management strategies and lack of biofilm-active antibiotics. Previous studies report treatment success from 50–76%. We evaluated the characteristics and outcome of enterococcal PJI, in particular the influence of antimicrobial treatment regimens. Method. Consecutive patients with enterococcal PJI treated at two specialized orthopaedic institutions were retrospectively included from 2010 to 2017. PJI was defined by the proposed European Bone and Joint Infection Society (EBJIS) criteria. Adequate antimicrobial treatment was considered when the antibiotic was appropiate for the treatment of enterococcal bone infections (activity, dose, oral bioavailability, bone penetration). The treatment success (defined as no relapse of enteroccal infection) and clinical success(i.e. infection-free status) was evaluated and compared using Fishers exact test. Results. We included 75 episodes with enterococcal PJI, involving 41 hip, 30 knee, 2 elbow, 1 shoulder prosthesis. The median patient age was 76 years (range, 30–90 years), 48 (64%) were female. The infection occurred perioperatively in 61 episodes (81%), haematogenously in 13 (17%) and by contiguous spread in 1 case. Sinus tract was present in 16 patients (21%), predominantly in polymicrobial compared to monomicrobial infections (13 vs. 3 episodes, p= 0.01). Preoperative serum C-reactive protein level was elevated in 63/75 patients (84%) and synovial fluid leukocyte count was increased in 25/29 patients (86%). Enterococci grew in synovial fluid in 76%, in periprosthetic tissue in 78% and in sonication fluid in 73% of patients. Predominantly, E. faecalis was identified (n=64), followed by E. faecium (n=10) and E. casseliflavus (n=1); mixed infections were diagnosed in 38 patients (51%). Two-stage prosthesis exchange was performed in 44 (59%), debridement and retention in 13 (17%), resection arthroplasty in 11 (15%) and one-stage exchange in 10 patients (13%). Of 66 patients with available follow-up data (median, 31.8 months; range, 0.3–83.3 months), the treatment success was 85% (56/66), however, clinical success was only 68% (45/66). Treatment success was similar in monomicrobial and polymicrobial infections. Adequate antimicrobial treatment was associated with significant better outcome (91% vs. 38%, p=0.002). Treatment with fosfomycin (19/20, 95%) and combination therapy (45/50, 90%) was associated with better outcome, however, did not reach statistical significance (p >0.05). Conclusions. The treatment outcome of enterococcal PJI was high (85%), however, a second episode of PJI caused by a new pathogen was common in the later course. Adequate antimicrobial treatment was the only significant factor associated with better treatment success


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_III | Pages 49 - 49
1 Feb 2012
Wimsey S Lien C Sharma S Brennan P Harper G Gorecki D
Full Access

Introduction. Osteoarthritis (OA) has historically been thought of as a degenerative joint disease, but inflammation and angiogenesis are increasingly being recognised as contributing to the pathogenesis, symptoms and progression of OA. b-dystroglycan (b-DG) is a pivotal element of the transmembrane adhesion molecule involved in cell-extracellular matrix adhesion and angiogenesis. Matrix metalloproteinases (MMPs) are the main enzymes responsible for cartilage extracellular matrix breakdown and are also implicated in both angiogenesis and b-DG degradation in a number of malignancies. We aimed to investigate the expression and localisation of b-DG and MMP-3, -9, and -13 within cartilage, synovium and synovial fluid and establish their roles in the pathogenesis of OA. Methods. Following ethical committee approval, cartilage, synovium and synovial fluid were obtained from the hip joints of 5 osteoarthritic (patients undergoing total hip replacement) and 5 control hip joints (patients undergoing hemiarthroplasty for femoral neck fracture). The samples were analysed for b-DG expression using Western Blotting and for the distribution of b-DG, MMP-3, -9, and -13 using immunohistochemistry on paraffin embedded tissue. Results. Whilst no significant expression of b-DG was found in cartilage or synovial fluid, b-DG was expressed in the smooth muscle of both normal and osteoarthritic synovial blood vessels. Moreover, b-DG was expressed in endothelium of blood vessels of OA synovium, but not in the normal endothelium. In the endothelium of osteoarthritic synovial blood vessels, b-DG co-localised with MMP -3 and -9. Discussion. Our results demonstrate that b-DG does not act as a cell adhesion molecule binding chondrocytes to the ECM. However, specific immunolocalisation of b-DG within endothelium of inflamed OA blood vessels suggests that b-DG may play a role in angiogenesis associated with OA. Its co-localisation with MMP-3 and -9, previously reported to also have pro-angiogenic roles, may be linked. Further research is required to understand these roles more fully