Advertisement for orthosearch.org.uk
Results 1 - 14 of 14
Results per page:
Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 91 - 91
2 Jan 2024
Graça A Rodrigues M Domingues R Gomes M Gomez-Florit M
Full Access

Macrophages play a critical role in innate immunity by promoting or inhibiting tissue inflammation and repair. Classically, macrophages can differentiate into either pro-inflammatory (M1) or pro-reparative (M2) phenotypes in response to various stimuli. Therefore, this study aimed to address how extracellular vesicles (EVs) derived from polarized macrophages can affect the inflammatory response of tendon cells. For that purpose, human THP-1 cells were stimulated with lipopolysaccharide (LPS), and interleukins -4 and -13 (IL- 4, IL-13), to induce macrophages polarization into M1, M2, and hybrid M1/M2 phenotypes. Subsequently, the EVs were isolated from the culture medium by ultracentrifugation. The impact of these nanovesicles on the inflammation and injury scenarios of human tendon-derived cells (hTDCs), which had previously been stimulated with interleukin- 1 beta (IL-1ß) to mimic an inflammatory scenario, was assessed. We were able to isolate three different nanovesicles populations, showing the typical shape, size and surface markers of EVs. By extensively analyzing the proteomic expression profiles of M1, M2, and M1/M2, distinct proteins that were upregulated in each type of macrophage-derived EVs were identified. Notably, most of the detected pro- inflammatory cytokines and chemokines had higher expression levels in M1-derived EVs and were mostly absent in M2-derived EVs. Hence, by acting as a biological cue, we observed that M2 macrophage-derived EVs increased the expression of the tendon-related marker tenomodulin (TNMD) and tended to reduce the presence of pro-inflammatory markers in hTDCs. Overall, these preliminary results show that EVs derived from polarized macrophages might be a potential tool to modulate the immune system responses becoming a valuable asset in the tendon repair and regeneration fields worthy to be further explored


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_7 | Pages 6 - 6
4 Apr 2023
Jamieson S Mawdesley A Hyde P Kirby J Tyson-Capper A
Full Access

Total hip replacement (THR) is indicated for patients with osteoarthritis where conservative treatment has failed. Metal alloys used in THR implants such as cobalt-chromium (CoCr) have been known to cause pro-inflammatory reactions in patients, therefore leading to the need for costly revision surgery. This study therefore aimed to investigate the role of TLR4 in the activation of a human osteoblast model in response to CoCr particles in vitro. Human osteoblasts (MG-63 cell line) were seeded at a density of 100,000 cells and treated with 0.5, 5, 50mm3 CoCr particles per cell for 24-hours. Trypan blue and the XTT Cell Proliferation Kit II were then used in conjunction with the cells to assess CoCr-induced cytotoxicity. Cells were pre-treated with a commercially available TLR4-specific small molecule inhibitor (CLI-095) for 6 hours. Untreated cells were used as a negative control and lipopolysaccharide (LPS) was used as a positive control. Following treatment the cell supernatant was collected and used for enzyme-linked immunosorbant assay (ELISA) to measure the secretion of interleukin-8 (IL-8), CXCL10, and interleukin-6 (IL-6). Trypan blue and XTT analysis showed that there was no significant changes to cell viability or proliferation at any dose used of CoCr after 24 hours. There was a significant increase in protein secretion of IL-8 (p<0.001), CXCL10 (p<0.001), and IL-6 (p<0.001) in the cells which received the highest dosage of CoCr. This pro-inflammatory secretory response was ameliorated by TLR4 blockade (p<0.001). CoCr particles are not cytotoxic to osteoblasts but they do induce pro-inflammatory changes as characterised by increased secretion of chemokines IL-8, CXCL10, and IL-6. These responses occur via a TLR4-mediated pathway and upon inhibition they can be effectively ameliorated. This is particularly important as TLR4 could be a potential target for pharmacological intervention used in patients experiencing immunological reactions to metal implant debris


Orthopaedic Proceedings
Vol. 97-B, Issue SUPP_11 | Pages 24 - 24
1 Oct 2015
Saengsoi W Tew S Bing C Comerford E German A
Full Access

Introduction. Obesity is one of risk factors of anterior cruciate ligament tear in man or cranial cruciate ligament (CCL) tear in dog. Adipokines are biologically active mediators released from adipocytes, and correlate with changes in body mass index. In order to study the possibility that adipocytes play a role in the pathogenesis of CCL disease, we investigated alterations of the matrix degradation biomarker genes (matrix metalloproteinase-13 [MMP-13], aggrecan) in CCL cells after stimulating with adipokines. Materials and Methods. We collected CCLs from 6 dog cadavers that had been euthanased for reasons other than musculoskeletal disease. CCL cells were isolated and treated with key adipokines including of adiponectin, leptin and visfatin at different concentration (0.1 ng/mL, 1 ng/mL and 10 ng/mL), and at three different time points (1 h, 6 h and 24 h). Real-time PCR was used to determine gene expression for MMP-13 and aggrecan in CCL cells comparing with negative control. In addition, lipopolysaccharide was used as a positive control. The statistical significance of differences between groups was determined using non-parametric Friedman test, followed by the Conover post-hoc test, and data were considered statistically significant at P<0.05. Results. We observed a significant difference for MMP-13 gene expression when stimulated with lipopolysaccharide at different concentrations and different time points (P<0.001 and P=0.007, respectively). However, there was no difference for any of the other treatments. Discussion. The adipokines studied do not affect gene expression within CCL cells for MMP-13 at doses under 10 ng/mL. Further studies could involve more animals, different adipokine concentrations, other biomarkers, and also detection of matrix degradation products in cell culture media


Orthopaedic Proceedings
Vol. 101-B, Issue SUPP_2 | Pages 28 - 28
1 Jan 2019
Mawdesley A Tyson-Capper A Kirby J Tipper JL
Full Access

Increased revision rates and early failure of Metal-on-Metal (MoM) hip replacements are often due to adverse reaction to metal debris (ARMD). Cobalt is a major component of MoM joints and can initiate an immune response via activation of the innate immune receptor Toll-like receptor 4 (TLR4). This leads to increased secretion of inflammatory cytokines/chemokines e.g. CCL3 and CCL4. The aim of this study was to evaluate whether TLR4-specific neutralising antibodies can prevent cobalt-mediated activation of TLR4. MonoMac 6 (MM6) cells, a human macrophage cell line, were treated with two different TLR4-specific monoclonal antibodies followed by 0.75mM of cobalt chloride (CoCl2). Lipopolysaccharide (LPS), a known TLR4 agonist was used as a positive control. Enzyme-linked immunosorbent assay (ELISA) was used to assess CCL3/CCL4 protein secretion and real time- polymerase chain reaction (RT-PCR) allowed quantification of CCL3/CCL4 gene expression. MM6 cells treated with cobalt and LPS up-regulate CCL3 and CCL4 gene expression and protein secretion. MM6 cells pre-treated with both monoclonal antibodies prior to stimulation with 0.75mM CoCl2 for 16 hours demonstrated significant inhibition of both CCL3 and CCL4 secretion as well as gene expression (both p=<0.0001). One of the antibodies failed to inhibit chemokine expression and secretion in LPS treated cells. This study identifies for the first time the use of TLR4-specific monoclonal antibodies to prevent cobalt activation of TLR4 and subsequent inflammatory response. This finding demonstrates the potential to exploit TLR4 inhibition in the context of MoM joint replacements by contributing to the development of novel therapeutics designed to reduce the incidence of ARMD


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_14 | Pages 60 - 60
1 Nov 2018
Raman S Mancuso P Murphy M
Full Access

Human synovium harbours macrophages and T-cells that secrete inflammatory cytokines, stimulating chondrocytes to release proteinases like aggrecanases and matrix metalloproteinases (MMPs) during the development of Osteoarthritis (OA). Inflammation of the synovium is a key feature of OA, linked to several clinical symptoms and the disease progression. As a prelude to testing in an OA mouse model, we have used the tetracycline system (Tet) to modify mouse mesenchymal stem cells (mMSCs) to over-express viral interleukin 10 (vIL10), an anti-inflammatory cytokine, to modulate the osteoarthritic environment and prevent disease development. MSCs isolated from the marrow of C57BL/6J mice expressed CD90.2, SCA-1, CD105, CD140a, and were negative for CD34, CD45 and CD11b by flow cytometry. Adenoviral transduction of MSCs carrying CMVIL10 and TetON as test, and untransduced, AdNull and TetOFF as negative controls was successful and tightly controlled vIL10 production was demonstrated by CMVIL10 and TetON MSCs using a vIL10 ELISA kit. Co-incubation of vIL10MSC CM with lipopolysaccharide activated bone-marrow derived murine macrophages (BMDMs) resulted in reduction of TNF-α, IL-6 levels and elevated production of IL-10 by ELISA and high iNOS release by Griess assay. Co-culture of active macrophages with TetON MSCs, resulted in polarisation of macrophage cell population from M1 to M2 phase, with decrease in pro-inflammatory MHC-II (M1 marker) and increase in regulatory CD206 (M2 marker) expression over time. The PCR profiler array on MSC CM treated BMDMs, also showed changes in gene expression of critical pro-inflammatory cytokines and receptors involved in the TLR4 pathway. The biscistronic TetON transduced MSCs proved to be most immuno-suppressive and therefore feasible as efficient anti-inflammatory therapy that can utilised in vivo


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_9 | Pages 10 - 10
1 May 2017
Mawdesley A Anjum S Lawrence H Deehan D Kirby J Tyson-Capper A
Full Access

Background. Increased revision rates and early failure of Metal-on-Metal (MoM) hip replacements are often due to adverse reaction to metal debris (ARMD). ARMD describes numerous symptoms in patients such as pain, osteolysis and soft tissue damage. Cobalt is a major component of MoM joints and can initiate an immune response via activation of the innate immune receptor Toll-like receptor 4 (TLR4). This leads to increased secretion of inflammatory cytokines e.g. interleukin-8 (IL-8). This study investigates whether TLR4-specific antagonists inhibit the inflammatory response to cobalt using IL-8 gene expression and protein secretion as a marker of TLR4 activation. Methods. MonoMac 6 (MM6) cells, a human macrophage cell line, were treated with TLR4-specific antagonists followed by 0.75mM of cobalt chloride. Lipopolysaccharide (LPS), a known TLR4 agonist was used as a positive control. Enzyme-linked immunosorbent assay (ELISA) was used to assess IL-8 protein secretion and real time- polymerase chain reaction (RT-PCR) allowed quantification of IL-8 gene expression. Results. MM6 cells treated with cobalt and LPS up-regulate IL-8 gene expression and protein secretion (n=3). The addition of TLR4-specific antagonists significantly inhibits this up-regulation suggesting the observed effects are TLR4-mediated. MM6 cells stimulated with cobalt (0.75mM) for 16 hours demonstrated a 27-fold increase in IL-8 gene expression (p-value = < 0.0001). When pre-treated with 10μg/ml of a TLR4-specific antagonist fold increase decreased to 6-fold (p-value = < 0.0001). IL-8 secretion decreased from 5000pg/ml to 3000pg/ml (p-value = < 0.0001). Conclusion. TLR4-specific antagonists inhibit cobalt-mediated IL-8 gene expression and protein secretion in MM6 cells. This finding demonstrates the potential to exploit this inhibition in the context of MoM joint replacements by contributing to the development of novel therapeutics designed to improve MoM implant longevity, reduce the incidence of ARMD and prevent subsequent revision surgery


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_3 | Pages 23 - 23
1 Apr 2018
Geurts J Müller M Pagenstert G Netzer C Schären S
Full Access

Introduction. Elevated remodelling of subchondral bone and marrow tissues has been firmly established as diagnostic and prognostic radiological imaging marker for human osteoarthritis. While these tissues are considered as promising targets for disease-modifying OA drugs, the development of novel treatment approaches is complicated by the lack of knowledge whether similar tissue changes occur in rodent OA models and poor understanding of joint-specific molecular and cellular pathomechanisms in human OA. Here, we describe the establishment of a human OA explant model to address this crucial niche in translational preclinical OA research. Methods. Osteochondral (knee, spine) and bone (iliac crest) clinical specimens were acquired from patients undergoing total knee arthroplasty (n=4) or lumbar spine fusion using bone autografts (n=6). Fresh specimens were immediately cut in equal-sized samples (50–500 mg wet weight) and cultured in 8 mL osteogenic medium for one week. Samples were either left untreated (control) or stimulated with lipopolysaccharide (LPS, 100 ng/mL) in the absence and presence of transforming growth factor-beta inhibitor (SB-505124, 10 μm). Pro-collagen-I (Col-I), interleukin-6 (IL-6) and monocyte chemoattractant protein 1 (MCP-1) secretion was determined in conditioned medium by ELISA. Tissue viability was assessed using MTT and alkaline phosphatase (ALP) activity staining. Results. Explanted tissues remained viable after one week culture in control and treatment conditions. Osteocytes, subchondral marrow spaces and calcified cartilage stained positive for ALP activity without gross morphological differences between groups. Median basal secretion levels were Col-I (2.3 ng/mg), IL-6 (90 pg/mg) and MCP-1 (25 pg/mg). LPS treatment led to a significant increase of IL-6 (330 pg/mg) and MCP-1 (70 pg/mg), but not Col-I secretion. Interestingly, inhibition of TGF-beta signalling in osteochondral tissues specifically reduced Col-I levels (0.4 ng/mg) compared to controls and LPS-treated samples. LPS-induced IL-6 and MCP-1 levels were slightly reduced (−120 pg/mg, p=0.03) and increased (+50 pg/mg) by SB-505124 treatment, respectively. IL-6 and MCP-1 levels were strongly correlated under basal (r=0.80) and treatment conditions (r=0.62). Conclusion. In this study, we provided proof of concept for the first ex vivo explant model of human osteoarthritis. Osteochondral tissue specimens can readily be cultured without loss of tissue viability and mount a robust inflammatory response upon LPS challenge. Treatment with a potential disease-modifying agent (TGF-beta signalling inhibitor) reduced collagen metabolism in bone and marrow and modified cytokine and chemokine expression. The osteochondral explant model might be highly valuable to evaluate disease-modifying OA drugs


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 10 - 10
1 Jul 2014
Kim S Hong J Yoon H Kwon B Lee I Kim H
Full Access

Summary Statement. Obovatol inhibits receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclastogenesis and prevents inflammatory bone loss in mice. Introduction. Adult skeletal mass and integrity are maintained by balancing osteoclast-mediated bone resorption and osteoblast-induced bone formation during bone remodeling. Abnormal increases in osteoclastic bone resorption can lead to excessive bone destruction as observed in osteoporosis, rheumatoid arthritis, and metastatic cancers Therefore, Modulation of osteoclast formation and function is a promising strategy for the treatment of bone-destructive diseases. To search for compounds that inhibit osteoclast formation, we tested the effect of obovatol, a natural product isolated from the medicinal plant Magnolia obovata, on osteoclastogenesis and inflammatory bone loss. Methods. Osteoclastogenesis was assessed using bone marrow-derived macrophages. RANKL signaling was assessed by immunoblotting and apoptosis by cell death ELISA assay. Actin ring staining and resorption pit assay was performed. Bone morphometric parameters were determined using a microcomputed tomography system. Results. We found that obovatol strongly inhibited osteoclast formation from bone marrow-derived macrophages in a dose-dependent manner without cytotoxicity. Obovatol significantly suppressed RANKL-induced activation of NF-κB, JNK, and ERK signaling pathways. Obovatol also inhibited RANKL-induced expression of the genes c-Fos and nuclear factor of activated T cells c1, which are transcription factors important for osteoclastogenesis. In addition to osteoclast differentiation, obovatol blocked cytoskeletal organization and abrogated the bone resorbing activity of mature osteoclast. Obovatol also accelerated osteoclast apoptosis through the induction of caspase-3 activation. Consistent with its in vitro anti-resorptive effect, obovatol prevented bone loss induced by lipopolysaccharide (LPS) in vivo. Conclusion. Our findings demonstrate that obovatol, a natural compound isolated from Magnolia obovata, suppresses the differentiation, function, and survival of osteoclasts. Furthermore, obovatol protected against LPS-induced bone loss in vivo. Therefore, we suggest that obovatol may have therapeutic potential for the treatment of bone-destructive diseases characterised by increased osteoclast number and/or activity


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 293 - 293
1 Jul 2014
Yasuda T
Full Access

Summary. Hyaluronan suppressed lipopolysaccharide-stimulated prostaglandin E. 2. production via intercellular adhesion molecule-1 through down-regulation of nuclear factor-κB. Administration of hyaluronan into rheumatoid joints may decrease prostaglandin E. 2. production by activated macrophages, which could result in improvement of arthritic pain. Introduction. Prostaglandin E. 2. (PGE. 2. ) is one of the key mediators of inflammation in rheumatoid arthritis (RA) joints. Intra-articular injection of high molecular weight hyaluronan (HA) into RA knee joints relieves arthritic pain. Although HA has been shown to inhibit PGE. 2. production in cytokine-stimulated synovial fibroblasts, it remains unclear how HA suppresses PGE. 2. production in catabolically activated cells. Furthermore, HA effect on macrophages has rarely been investigated in spite of their contribution to RA joint pathology. Objectives. This study was aimed to investigate the inhibitory mechanism of HA on lipopolysaccharide (LPS)-stimulated PGE. 2. in U937 human macrophage culture system. Methods. With or without pretreatment with one of HA, NS-398, and BAY11-7085, differentiated U937 macrophages were stimulated with LPS. In another set of experiments, the cells were incubated with anti-ICAM-1 antibody or non-specific IgG before pretreatment with HA. PGE. 2. concentrations of the cell-free supernatants were determined using an enzyme-linked immunosorbent assay. The cell lysates and nuclear extracts were prepared for immunoblot analysis. HA binding to ICAM-1 was evaluated by fluorescence microscopic analysis. Results. Stimulation of U937 macrophages with LPS enhanced PGE. 2. production in association with increased protein levels of cyclooxygenase-2 (COX-2). Pretreatment with HA of 2,700 kDa resulted in suppression of LPS-induced COX-2, leading to a decrease in PGE. 2. production. While LPS activated NF-κB pathway, inhibition studies using BAY11-7085 revealed the requirement of NF-κB for LPS-stimulated PGE. 2. production. HA down-regulated the phosphorylation and nuclear translocation of NF-κB by LPS. Fluorescence cytochemistry demonstrated that HA bound to ICAM-1 on U937 macrophages. Anti-ICAM-1 antibody reversed the inhibitory effects of HA on LPS-activated PGE. 2. , COX-2, and NF-κB. Conclusion. These results clearly demonstrated that HA suppressed LPS-stimulated PGE2 production via ICAM-1 through down-regulation of NF-κB. Clinical administration of high molecular weight HA into RA joints may decrease PGE2 production by activated macrophages, which could result in improvement of arthritic pain


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 292 - 292
1 Jul 2014
Lawrence H Deehan D Holland J Kirby J Tyson-Capper A
Full Access

Summary. Metal-on-metal hip replacements have been associated with adverse reactions including inflammatory pseudotumours and soft tissue necrosis. We have shown that cobalt can directly activate toll-like receptor 4, an immune receptor causing pro-inflammatory interleukin-8 secretion. This may contribute to adverse reaction development. Introduction. Metal-on-metal hips have the highest failure rate of any joint arthroplasty material. Reasons for failure include the development of pseudotumours, soft tissue necrosis and pain around the affected joint. The adverse reactions appear to be inflammatory as failing joints are often infiltrated by immune cells such as lymphocytes. However the exact cellular and biological mechanisms underlying this inflammation are unknown. Toll-like receptor 4 (TLR4) is found on the surface of immune cells including macrophages and dendritic cells. It is activated by lipopolysaccharide (LPS) from Gram negative bacteria, inducing an immune response against the pathogen through increased secretion of pro-inflammatory cytokines. It has recently been shown that nickel can activate TLR4, causing inflammation. Cobalt, a component of many metal-on-metal joints, is adjacent to nickel in the periodic table and shares a number of nickel's properties. Consequently we hypothesised that cobalt ions from metal-on-metal joints can activate TLR4. Methods. An in vitro cell culture model was developed using human and murine TLR4 reporter cell lines to investigate the effects of metal ions, including cobalt, on TLR4. Real-time PCR was used to examine the effect of cobalt on inflammatory gene expression, including IL-8, CCL-2 and IRAK-2, while an ELISA assay was conducted to investigate IL-8 protein expression in a human macrophage cell line (MonoMac 6). The TLR4 agonist LPS was included as a positive control and as a negative control TLR4 activation was blocked using the chemical agonist CLI-095 (Invivogen, UK). Results. Using human TLR4 reporter cells we show that cobalt at clinically-relevant concentrations can activate human TLR4. This effect appears unique to humans as murine TLR4 is unresponsive to cobalt but still responds to LPS. We also demonstrate that in human macrophages physiologically-relevant concentrations of cobalt cause increased pro-inflammatory IL-8 secretion (p<0.001). IL-8 is involved in perpetuating the immune response by recruiting more inflammatory cells to the site of inflammation. Cobalt-induced IL-8 secretion can be blocked using a TLR4 antagonist (p<0.001) showing that the effect is due to cobalt activation. Cobalt ions also alter gene expression in human macrophages. Cobalt upregulates expression of IL-8 and IRAK2 genes; IRAK2 is a key component of the TLR4 signalling pathway. Interestingly, cobalt causes downregulation of the CCL2 gene whereas it is upregulated in response to LPS. Discussion. In this study we have demonstrated that cobalt ions can activate human TLR4 signalling and in human macrophages this can increase expression of pro-inflammatory IL-8. We have also developed a robust series of assays for determining the effects of metal ions and other orthopaedic materials on the TLR4 signalling pathway. These methods will be used to investigate the immunological effects of additional orthopaedic metals (e.g. chromium, titanium and molybdenum). This work has identified a key pathway involved in the immune response to metal ions which can now be investigated for genetic variability and as a potential therapeutic target


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_VIII | Pages 57 - 57
1 Mar 2012
Okazaki S Matsumoto H Nagoya S Kaya M Sasaki M Tateda K Kosukegawa I Yamashita T
Full Access

Introduction. Although osteonecrosis of the femoral head has been observed in young adult patients with autoimmune diseases such as SLE and MCTD that are treated by corticosteroids, the pathogenesis of the osteonecrosis remains unclear. We established a rat model with osteonecrosis of the femoral head by injecting lipopolysaccharide (LPS) and corticosteroid, and assessed consequences of the histopathological alteration of the femoral head, the systemic immune response, and the lipid synthesis. Methods. Male Wistar rats were given 2 mg/kg LPS intravenously on days 0 and 1 and intramuscularly 20 mg/kg methylprednisolone on days 2, 3, and 4. The animals were sacrificed 1, 2, 3, or 4 weeks after the last injection of the methylprednisolone. Histopathological and biochemical analyses were performed every week. The bone samples were then processed for routine hematoxylin and eosin staining to assess the general architecture and injury of the tissue. The triglyceride and the total cholesterol concentrations in the PRP were measured. The levels of various cytokines (IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, GM-CSF, IFN-γ, TNF-α) in blood samples were measured. Results. The body weight of the rats over time decreased for 2 weeks but had recovered by week 4. The plasma triglyceride concentrations had decreased significantly by weeks 2 and 3. The total plasma cholesterol concentrations had increased significantly by week 1 but then decreased significantly by week 4. The plasma concentrations of IL-1?α, IL-2, IL-4, IL-6, IL-10, GM-CSF, IFN-γ and TNF-α had increased significantly by week 1. These cytokines can all be induced by toll-like receptor 4 (TLR4) signaling. We defined osteonecrosis as the diffuse presence of empty lacunae or pyknotic nuclei of osteocytes in the bone trabeculae, accompanied by surrounding bone marrow cell necrosis. Osteonecrosis of the femoral head was observed only in the epiphysis of the femoral head in sacrificed specimen every week. Histological analysis revealed osteocytic death surrounded by necrotic bone marrow with or without repaired tissue. Conclusion. We established a new rat model of corticosteroid-induced femoral head osteonecrosis. The necrosis that is generated in this model is similar to that seen in patients treated with corticosteroid. In particular, the necrotic lesion was exclusively observed in the proximal epiphysis. LPS is known to activate the immune system via the TLR4 signaling pathway. It has been recognized that the unique immunogenic effects of LPS promote autoimmune disease . LPS and methylprednisolone induced osteonecrosis of the femoral head in rats and this was associated with a disruption of the innate immune system and lipid synthesis. These findings suggest that the TLR4 signaling pathway plays an important role in the pathogenesis for osteonecrosis of the femoral head


Bone & Joint Research
Vol. 6, Issue 3 | Pages 154 - 161
1 Mar 2017
Liu J Li X Zhang H Gu R Wang Z Gao Z Xing L

Objectives

Ubiquitin E3 ligase-mediated protein degradation regulates osteoblast function. Itch, an E3 ligase, affects numerous cell functions by regulating ubiquitination and proteasomal degradation of related proteins. However, the Itch-related cellular and molecular mechanisms by which osteoblast differentiation and function are elevated during bone fracture repair are as yet unknown.

Methods

We examined the expression levels of E3 ligases and NF-κB members in callus samples during bone fracture repair by quantitative polymerase chain reaction (qPCR) and the total amount of ubiquitinated proteins by Western blot analysis in wild-type (WT) mice. The expression levels of osteoblast-associated genes in fracture callus from Itch knockout (KO) mice and their WT littermates were examined by qPCR. The effect of NF-κB on Itch expression in C2C12 osteoblast cells was determined by a chromatin immunoprecipitation (ChIP) assay.


Bone & Joint Research
Vol. 1, Issue 11 | Pages 297 - 309
1 Nov 2012
McIlwraith CW Frisbie DD Kawcak CE

Osteoarthritis (OA) is an important cause of pain, disability and economic loss in humans, and is similarly important in the horse. Recent knowledge on post-traumatic OA has suggested opportunities for early intervention, but it is difficult to identify the appropriate time of these interventions. The horse provides two useful mechanisms to answer these questions: 1) extensive experience with clinical OA in horses; and 2) use of a consistently predictable model of OA that can help study early pathobiological events, define targets for therapeutic intervention and then test these putative therapies. This paper summarises the syndromes of clinical OA in horses including pathogenesis, diagnosis and treatment, and details controlled studies of various treatment options using an equine model of clinical OA.


The Journal of Bone & Joint Surgery British Volume
Vol. 93-B, Issue 4 | Pages 558 - 565
1 Apr 2011
Xie X Wang X Zhang G Liu Z Yao D Hung L Hung VW Qin L

Corticosteroids are prescribed for the treatment of many medical conditions and their adverse effects on bone, including steroid-associated osteoporosis and osteonecrosis, are well documented. Core decompression is performed to treat osteonecrosis, but the results are variable. As steroids may affect bone turnover, this study was designed to investigate bone healing within a bone tunnel after core decompression in an experimental model of steroid-associated osteonecrosis. A total of five 28-week-old New Zealand rabbits were used to establish a model of steroid-induced osteonecrosis and another five rabbits served as controls. Two weeks after the induction of osteonecrosis, core decompression was performed by creating a bone tunnel 3 mm in diameter in both distal femora of each rabbit in both the experimental osteonecrosis and control groups. An in vivo micro-CT scanner was used to monitor healing within the bone tunnel at four, eight and 12 weeks postoperatively. At week 12, the animals were killed for histological and biomechanical analysis.

In the osteonecrosis group all measurements of bone healing and maturation were lower compared with the control group. Impaired osteogenesis and remodelling within the bone tunnel was demonstrated in the steroid-induced osteonecrosis, accompanied by inferior mechanical properties of the bone.

We have confirmed impaired bone healing in a model of bone defects in rabbits with pulsed administration of corticosteroids. This finding may be important in the development of strategies for treatment to improve the prognosis of fracture healing or the repair of bone defects in patients receiving steroid treatment.