Advertisement for orthosearch.org.uk
Results 1 - 20 of 39
Results per page:
Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXVIII | Pages 30 - 30
1 Jun 2012
Millar N Reilly J Leach W Rooney B Murrell G McInnes I
Full Access

The objective was to seek evidence of hypoxia in early human tendinopathy and thereafter, to explore mechanisms whereby tissue hypoxia may regulate apoptosis, inflammatory mediators and matrix regulation in human tenocytes. Fifteen torn supraspinatus tendon (established pathology) and matched intact subscapularis tendon (representing ‘early pathology’) biopsies were collected from patients undergoing arthroscopic shoulder surgery. Control samples of subscapularis tendon were collected from 10 patients undergoing arthroscopic stabilisation surgery. Markers of hypoxia were quantified by immunohistochemical methods. Human tendon-derived primary cells were derived from hamstring tendon tissue obtained during hamstring tendon ACL reconstruction. The impact of hypoxia upon tenocyte biology ex vivo was measured using quantitative RT-PCR, multiplex cytokine assays, apoptotic proteomic profiling, immunohistochemistry and annexin V FACS staining. Increased expression of HIF 1a, Bcl-2 and clusterin (hypoxic and apoptotic markers) was detected in subscapularis tendon samples compared to both matched torn samples and non matched control samples (p<0.01). Hypoxic tenocytes exhibited increased production of proinflammatory cytokines (p<0.001), altered matrix regulation (p<0.01) with increased production of Collagen type III operating through a MAPK dependent pathway. Finally, hypoxia increased expression of several mediators of apoptosis and thereby promoted tenocyte apoptosis. Hypoxia promotes expression of proinflammatory cytokines, key apoptotic mediators and drives matrix component synthesis towards a collagen type III profile by human tenocytes. We propose hypoxic cell injury as a critical pathophysiological mechanism in early tendinopathy offering novel therapeutic opportunities in the management of tendon disorders


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_2 | Pages 74 - 74
1 Jan 2017
Kuhn A Das R Pavanram P Pufe T Jahr H
Full Access

Adult chondrocytes experience a hypoxic environment in vivo. Culturing chondrocytes under oxygen tension that more closely resembles the in vivo situation, i.e. hypoxic conditions, has been shown to have positive effects on matrix synthesis. During redifferentiation of expanded chondrocytes, hypoxia increased collagen type II expression. However, the mechanism by which hypoxia enhances redifferentiation is still incompletely elucidated. We employed micro-bioreactor technology to elucidate the contribution of TGF-β superfamily ligands to the chondrocyte differentiation process under hypoxic conditions in vitro. Dedifferentiated chondrocytes in alginate were cultured for 48 hours under hypoxic (1% pO2) or normoxic (20%) conditions, using specialized bioreactor technology. Gene expression of chondrocyte-specific markers (SOX9, COL2A1, COL1A1, AGC1 and MMP13) as well as established hypoxia-controlled genes (GDF1-, PHD3, HAS2, VEGF, COX2) and components of the TGF-β superfamily signaling pathways were analyzed by qPCR and protein expression after 48 hours in combination with TGF-β superfamily ligand-specific siRNA as well as selected TGF-β superfamily receptor inhibitors. Hypoxic culture showed robust upregulation of the selected hypoxia-specific marker genes. In addition, well-established chondrocyte-specific markers like SOX9 and collagen type II were upregulated. TGF-β isoforms were selectively upregulated under hypoxia on both mRNA and protein level. In addition, both Activin receptor-like kinases, ALK1 and ALK5, were upregulated under hypoxia, while respective type II and III receptors were unresponsive. The hypoxia-induced COL2 expression was abrogated by TGF-β2 siRNA, as was ALK5 inhibition. Our data strongly indicates that TGF-β superfamily signaling pathways are involved in chondrocyte redifferentiation under low oxygen tension in vitro


The Journal of Bone & Joint Surgery British Volume
Vol. 92-B, Issue 3 | Pages 448 - 453
1 Mar 2010
Benson RT McDonnell SM Knowles HJ Rees JL Carr AJ Hulley PA

The aim of this study was to investigate the occurrence of tissue hypoxia and apoptosis at different stages of tendinopathy and tears of the rotator cuff. We studied tissue from 24 patients with eight graded stages of either impingement (mild, moderate and severe) or tears of the rotator cuff (partial, small, medium, large and massive) and three controls. Biopsies were analysed using three immunohistochemical techniques, namely antibodies against HIF-1α (a transcription factor produced in a hypoxic environment), BNip3 (a HIF-1α regulated pro-apoptotic protein) and TUNEL (detecting DNA fragmentation in apoptosis). The HIF-1α expression was greatest in mild impingement and in partial, small, medium and large tears. BNip3 expression increased significantly in partial, small, medium and large tears but was reduced in massive tears. Apoptosis was increased in small, medium, large and massive tears but not in partial tears. These findings reveal evidence of hypoxic damage throughout the spectrum of pathology of the rotator cuff which may contribute to loss of cells by apoptosis. This provides a novel insight into the causes of degeneration of the rotator cuff and highlights possible options for treatment


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_16 | Pages 9 - 9
17 Nov 2023
Lim JW Ball D Johnstone A
Full Access

Abstract. Objectives. Acute compartment syndrome (ACS) is a progressive form of muscle ischaemia that is a surgical emergency and can have detrimental outcomes for patients if not treated optimally. The current problem is that there is no clear diagnostic threshold for ACS or guidance as to when fasciotomies should be performed. A new diagnostic method(s) is necessary to provide real-time information about the extent of muscle ischaemia in ACS. Given that lactic acid is produced by cells through anaerobic respiration, it may be possible to measure H+ ion concentration and to use this as a measure of ischaemia within muscle. Although we are familiar with the key biochemical metabolites involved in ischaemia; and the use of viability dyes in cell culture to distinguish between living or dead cells is well recognised; research has not been undertaken to correlate the biochemical and histological findings of ischaemia in skeletal muscle biopsies. Our primary aim was to investigate the potential for viability dyes to be used on live skeletal muscle biopsies (explants). Our secondary aim was to correlate the intramuscular pH readings with muscle biopsy viability. Methods. Nine euthanised Wistar rats were used. A pH catheter was inserted into one exposed gluteus medius muscles to record real-time pH levels and muscle biopsies were taken from the contralateral gluteus medius at the start of experiment and subsequently at every 0.1 of pH unit drop. Prior to muscle biopsy, the surface of the gluteus medius was painted with a layer of 50µmol/l Brilliant blue FCF solution to facilitate biopsy orientation. A 4mm punch biopsy tool was used to take biopsies. Each muscle biopsy was placed in a base mould filled with 4% ultra-low melting point agarose. The agarose embedded tissue block was sectioned to generate 400 micron thick tissue slices with a vibratome. The tissue slices were then placed in the staining solution with Hoechst 33342, Ethidium homodimer-1 and Calcein am. The tissue slices were imaged with Zeiss LSM880 confocal microscope's Z stack function. A dead muscle control was created by adding TritonX-100 to other tissue slices. For quantitative analyses, the images were analysed in Image J using the selection tool. This permitted individual cells to be identified and the mean grey value of each channel to be defined. Using the dead control, we were able to identify the threshold value for living cells using the Calcein AM channel. Results. Viability dyes, used primarily for cell cultures, can be used with skeletal muscle explants. Our study also showed that despite a significant reduction in tissue pH concentration over time, that almost 100% of muscle cells were still viable at pH 6.0, suggesting that skeletal muscle cells are robust to hypoxic insult in the absence of reperfusion. Conclusions. Viability dyes can be used on skeletal muscle biopsies. Further research investigating the likely associations between direct measured pH using a pH catheter, the concentrations of key cellular metabolic markers, and muscle tissue histology using vitality dyes in response to ischaemia, rather than hypoxia, is warranted. Declaration of Interest. (b) declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported:I declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research project


Orthopaedic Proceedings
Vol. 105-B, Issue SUPP_16 | Pages 10 - 10
17 Nov 2023
Lim JW Ball D Johnstone A
Full Access

Abstract. Background. Progressive muscle ischaemia results in reduced aerobic respiration and increased anaerobic respiration, as cells attempt to survive in a hypoxic environment. Acute compartment syndrome (ACS) is a progressive form of muscle ischaemia that is a surgical emergency resulting in the production of Lactic acid by cells through anaerobic respiration. Our previous research has shown that it is possible to measure H+ ions concentration (pH) as a measure of progressive muscle ischaemia (in vivo) and hypoxia (in vitro). Our aim was to correlate intramuscular pH readings and cell viability techniques with the intramuscular concentration of key metabolic biomarkers [adenosine triphosphate (ATP), Phosphocreatine (PCr), lactate and pyruvate], to assess overall cell health in a hypoxic tissue model. Methods. Nine euthanised Wistar rats were used in a non-circulatory model. A pH catheter was used to measure real-time pH levels from one of the exposed gluteus medius muscles, while muscle biopsies were taken from the contralateral gluteus medius at the start of the experiment and subsequently at every 0.1 of a pH unit decline. The metabolic biomarkers were extracted from the snap frozen muscle biopsies and analyzed with standard fluorimetric method. Another set of biopsies were stained with Hoechst 33342, Ethidium homodimer-1 and Calcein am and imaged with a Zeiss LSM880 confocal microscope. Results. Our study shows that the direct pH electrode readings decrease with time and took an average of 69 minutes to drop to a pH of 6.0. The concentrations of ATP, pyruvate and PCr declined over time, and the concentration of lactate increased over time. At pH 6.0, both ATP and PCr concentrations had decreased by 20% and pyruvate has decreased by 50%, whereas lactate had increased 6-fold. The majority of cells were still viable at a pH of 6.0, suggesting that skeletal muscle cells are remarkably robust to hypoxic insult, although this was a hypoxic model where reperfusion was not possible. Conclusions. Our research suggests that histologically, skeletal muscle cells are remarkably robust to hypoxic insult despite the reduction in the total adenine nucleotide pool, but this may not reflect the full extent of cell injury and quite possibly irreversible injury. The timely restoration of blood flow in theory should halt the hypoxic insult, but late reperfusion results in cellular dysfunction and cell death due to localised free radical formation. Further research investigating the effects of reperfusion in vivo are warranted, as this may identify an optimal time for using pharmacological agents to limit reperfusion injury, around the time of fasciotomy to treat acute compartment syndrome. Declaration of Interest. (b) declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported:I declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research project


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 73 - 73
2 Jan 2024
Vinhas A Rodrigues M Gonçalves A Gomes M
Full Access

Common tendon injuries impair healing, leading to debilitation and an increased re-rupture risk. The impact of oxygen-sensing pathways on repair mechanisms, vital in regulating inflammation and fibrosis, remains unclear despite their relevance in tendon pathologies. Recent studies show that pulsed electromagnetic field (PEMF) reduce inflammation in human tendon cells (hTDCs) and in hypoxia-induced inflammation. We investigated the hypoxia's impact (1% and 2% oxygen tension) using magnetic cell sheet constructs (IL-1β-magCSs) primed with IL-1β. IL-1β-magCSs were exposed to low OT (1h, 4h,6h) in a hypoxic chamber. To confirm the role of PEMF (5Hz, 4mT, 50% duty cycle) on hypoxia modulation, IL-1β-magCSs, previously exposed to OT, were 1h-stimulated with PEMF. Our results show a significant increase in HIF- 1a and HIF-2a expression on IL-1β-magCSs after exposure to 2%-OT at all time points, compared to 1%- OT and normoxia. TNFa, IL-6, and IL-8 expression increased after 6 hours of 1%-OT exposure. PEMF stimulation of hypoxic IL-1β-magCSs led to decreased pro-inflammatory genes and increased anti-inflammatory (IL-4,IL-10) expression compared to unstimulated magCSs. IFN-g, TNF-α, and IL-6 release increased after 6 hours, regardless of %-OT, while IL-10 levels tended to rise after PEMF stimulation at 2%-OT. Also, NFkB expression was increased on IL-1β-magCSs exposed to 4 h and 6 h of 2%-OT, suggesting a link between NFkB and the production of pro-inflammatory factors. Moreover, PEMF stimulation showed a significantly decreased NFkB level in IL-1β-magCSs. Overall, low OT enhances expression of hypoxia-associated genes and inflammatory markers in IL-1β-magCSs with the involvement of NFkB. PEMF modulates the response of magCSs, previously conditioned to hypoxia and to inflammatory triggers, favouring expression of anti-inflammatory genes and proteins, supporting PEMF impact in pro-regenerative tendon strategies. Acknowledgements: ERC CoG MagTendon(No.772817), FCT under the Scientific Employment Stimulus-2020.01157.CEECIND. Thanks to Hospital da Prelada for providing tendon tissue samples (Portugal), and TERM. RES Hub (Norte-01-0145-FEDER-022190)


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_1 | Pages 107 - 107
2 Jan 2024
Park H
Full Access

The rotator cuff tendinopathy is one of the most common shoulder problems leading to full-thickness rotator cuff tendon tear and, eventually, to degenerative arthritis. Recent research on rotator cuff tendon degeneration has focused on its relationship to cell death. The types of cell death known to be associated with rotator cuff tendon degeneration are apoptosis, necrosis, and autophagic cell death. The increased incidence of cell death in degenerative tendon tissue may affect the rates of collagen synthesis and repair, possibly weakening tendon tissue and increasing the risk of tendon rupture. The biomolecular mechanisms of the degenerative changes leading to apoptotic cell death in rotator cuff tenofibroblasts have been identified as oxidative-stress-related cascade mechanisms. Furthermore, apoptosis, necrosis, and autophagic cell death are all known to be mediated by oxidative stress, a condition in which ROS (reactive oxygen species) are overproduced. Lower levels of oxidative stress trigger apoptosis; higher levels mediate necrosis. Although the signaltransduction pathway leading to autophagy has not yet been fully established, ROS are known to be essential to autophagy. A neuronal theory regarding rotator cuff degeneration has been developed from the findings that glutamate, a neural transmitter, is present in increased concentrations in tendon tissues with tendinopathy and that it induces rat supraspinatus tendon cell death. Recent studies have reported that hypoxia involved in rotator cuff tendon degeneration. Because antioxidants are known to scavenge for intracellular ROS, some studies have been conducted to determine whether antioxidants can reduce cell death in rotator cuff tendon-origin fibroblasts. The first study reported that an antioxidant has the ability to reduce apoptosis in oxidative-stressed rotator cuff tenofibroblasts. The second study reported that antioxidants have both antiapoptotic effects and antinecrotic effects on rotator cuff tendon-origin fibroblasts exposed to an oxidative stimulus. The third study reported that an antioxidant has antiautophagic-cell-death effects on rotator cuff tendon-origin fibroblasts exposed to an oxidative stimulus. The fourth study reported that glutamate markedly increases cell death in rotator cuff tendonorigin fibroblasts. The glutamate-induced cytotoxic effects were reduced by an antioxidant, demonstrating its cytoprotective effects against glutamate-induced tenofibroblast cell death. The fifth study reported that hypoxia significantly increases intracellular ROS and apoptosis. The hypoxia-induced cytotoxic effects were markedly attenuated by antioxidants, demonstrating their cytoprotective effects against hypoxia-induced tenofibroblast cell death. In conclusion, antioxidants have cytoprotective effects on tenofibroblasts exposed in vitro to an oxidative stressor, a neurotransmitter, or hypoxia. These cytoprotective effects result from antiapoptotic, antinecrotic, and antiautophagic actions involving the inhibition of ROS formation. These findings suggest that antioxidants may have therapeutic potential for rotator cuff tendinopathy. Further studies must be conducted in order to apply these in vitro findings to clinical situations


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_4 | Pages 113 - 113
1 Mar 2021
George A Ellis M Gill R
Full Access

Hypoxic Inducible Factor and Hypoxic mimicking agents (HMA) trigger the initiation and promotion of angiogenic-osteogenic cascade events. However, there has been paucity of studies investigating how HIF could be over expressed under chronic hypoxic conditions akin to that seen in sickle cell disease patients to help form a template for tackling the matter of macrocellular avascular necrosis. Angiogenesis and osteogenesis are tightly coupled during bone development and regeneration, and the hypoxia-inducible factor-1 alpha (HIF-1) pathway has been identified as a key component in this process studies have shown. There are still no established experimental models showing how this knowledge can be used for the evaluation of bone implant integration and suggest ways of improving osseointegration in sickle cell disease patients with hip arthroplasty and thereby prevent increased implant loosening. The aim of this study is to help develop an in vitro experimental model which would mimic the in vivo pathologic state in the bone marrow of sickle cell disease patients. It also seeks to establish if the hypoxic inducible factor (HIF) could be over expressed in vitro and thus enhancing osseointegration. MG63 osteoblastic cells were cultured under normoxia and hypoxic conditions (20%; and 1% oxygen saturation) for 48 and 72 hours. Cobalt chloride was introduced to the samples in order to mimic true hypoxia. Cells cultured under normoxic conditions and without cobalt chloride was used as the control in this study. The expression of the hypoxic inducible factor was assessed using the reverse transcriptase qualitative polymerase chain reaction (RT-qPCR). There was increased expression of HIF1-alpha at 72hours as compared to 48hours under the various conditions. The level of expression of HIF increased from 48hrs (mean rank= 4.60) to 72hrs (mean rank =5.60) but this difference was not statistically significant, X2(1) = 0.24, p =0.625. The mean rank fold change of HIF in hypoxic samples decreased compared to the normoxic samples but this difference was not statistically significant, X2(1) = 0.54, p= 0.462. Therefore, the expression of HIF is only increased with prolonged hypoxia as seen in the 72hours samples. The expression of HIF increased in samples with CoCl2 (mean rank=5.17), compared with samples without CoCl2 (mean rank 4.67), however this was not statistically significant, X2(1) = 0.067, p=0.796, p value > 0.05. The over expression of HIF was achieved within a few days (72hours) with the introduction of Cobalt Chloride, which is a mimetic for hypoxia similar to the in vivo environment in sickle cell disease patients. This is an in vitro model which could help investigate osseointergation in such pathologic bone conditions


Orthopaedic Proceedings
Vol. 103-B, Issue SUPP_13 | Pages 37 - 37
1 Nov 2021
Peretti GM
Full Access

In the last decades, significant effort has been attempted to salvage the meniscus following injury. Basic science approaches to meniscus repair include procedures for both meniscus regeneration and meniscus healing. Regeneration of meniscal tissue focuses on filling a defect with reparative tissue, which resembles the native structure and function of the meniscus. Procedures for meniscus healing, on the other hand, aim to accomplish adhesion between the margins of a meniscal lesion, with no attempt to regenerate or replace meniscal tissue. Regeneration studies of tissue to fill a defect in the meniscus have shown interesting results, but complete restoration of the native meniscus has not yet been accomplished. Healing of a meniscal lesion has been investigated in different models although none has demonstrated reproducible healing. Therefore, different paths of investigation must be undertaken, and one of these may be the cell-therapy / tissue engineering approach. In a study from our group, we showed the capacity of chondrocyte-seeded cartilaginous scaffold to repair a bucket-handle lesion of the knee meniscus orthotopically in a large animal study. Following studies were done in order to test the potential of other scaffolds and different cell sources for the repair of the meniscal tissue. We have also evaluated the role of hypoxia in meniscal development in vitro as basis for future research in this field, as hypoxia could be be considered as a promoter for meniscal cells maturation, and opens considerably opportunities in the field of meniscus tissue engineering


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_15 | Pages 43 - 43
1 Nov 2018
Aaron R
Full Access

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


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 115 - 115
1 Jul 2014
Buizer A Bulstra S Veldhuizen A Kuijer R
Full Access

Summary. Within hours after exposure to hypoxic circumstances hMSCs start producing AGFs. Initially hypoxia does not affect hMSC proliferation and metabolic activity, but after 7 days both are decreased, compared to hMSCs cultured under ambient oxygen conditions. Introduction. At the moment of implantation of a large cell seeded scaffold, usually a vascular network is lacking within the scaffold. Therefore, the cells seeded on the scaffold are exposed to hypoxic circumstances. Human mesenchymal stem cells (hMSCs) exposed to hypoxic circumstances, start to produce angiogenic factors (AGF). 1. and to proliferate faster than at ambient oxygen levels. 2. Under severe, continued hypoxia, hMSC metabolism slows down and ultimately stops. 3. We hypothesise that there is a threshold oxygen level above which hMSCs at hypoxia will both produce AGF and still proliferate, and below which cells slow down their metabolism. If hMSCs are provided with oxygen levels just above this threshold, effective tissue regeneration, which requires cell proliferation and vascular ingrowth, may be accomplished. Methods. Human mesenchymal stem cells (hMSCs) were isolated using Ficoll density gradient centrifugation from reaming debris that was collected during total hip replacement. Experiments were performed in a hypoxic chamber at 5% CO. 2. and 1%, 2%, 3%, 4%, 6%, 8% and 10% O. 2. , temperature was 37°C and humidity was 100%. The cells were transfected with a pCDNA\HRE-luciferase 2 plasmid to find the oxygen range at which hMSCs start producing AGF. Subsequently, AGF production was quantified using qPCR. Cell proliferation and cell metabolism rate under hypoxic circumstances was assayed using the CyQuant DNA quantification method and a (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, respectively. Results. In hMSCs cultured at 1% O. 2. , the hypoxia responsive element was activated. A selection of AGFs was expressed at 1%, 2% and 3% O. 2. AGF production started four hours after exposure to hypoxic circumstances commenced. Initially, at 1 day of exposure to hypoxia, cell proliferation and cell metabolic activity were not influenced by hypoxia. After 7 days, hMSC proliferation and metabolic activity were lower in cells grown under hypoxic circumstances than in cells grown under ambient oxygen circumstances. Discussion/Conclusion. The threshold level above which hMSCs at hypoxia will both produce AGF and still proliferate seems lower than 1% O. 2. Within hours after exposure to hypoxic circumstances hMSCs start producing AGFs. Initially, hMSC proliferation and metabolic activity were not affected by hypoxia, but after 7 days both proliferation and metabolic activity were decreased compared to hMSCs cultured under ambient oxygen circumstances


Orthopaedic Proceedings
Vol. 106-B, Issue SUPP_2 | Pages 93 - 93
2 Jan 2024
Bermudez-Lekerika P Tseranidou S Kanelis E Crump K Le Maitre C Wuertz-Kozak K Alexopoulos L Noailly J Gantenbein B
Full Access

Intervertebral disc (IVD) degeneration is a pathological process often associated with chronic back pain and considered a leading cause of disability worldwide. 1. During degeneration, progressive structural and biochemical changes occur, leading to blood vessel and nerve ingrowth and promoting discogenic pain. 2. In the last decades, several cytokines have been applied to IVD cells in vitro to investigate the degenerative cascade. Particularly, IL-10 and IL-4 have been predicted as important anabolic factors in the IVD according to a regulatory network model based in silico approach. 3. Thus, we aim to investigate the potential presence and anabolic effect of IL-10 and IL-4 in human NP cells (in vitro) and explants (ex vivo) under hypoxia (5% O2) after a catabolic induction. Primary human NP cells were expanded, encapsulated in 1.2% alginate beads (4 × 106 cells/ml) and cultured for two weeks in 3D for phenotype recovery while human NP explants were cultured for five days. Afterwards, both alginate and explant cultures were i) cultured for two days and subsequently treated with 10 ng/ml IL-10 or IL-4 (single treatments) or ii) stimulated with 0.1 ng/ml IL-1β for two days and subsequently treated with 10 ng/ml IL-10 or IL-4 (combined treatments). The presence of IL-4 receptor, IL-4 and IL-10 was confirmed in human intact NP tissue (Fig 1). Additionally, IL-4 single and combined treatments induced a significant increase of proinflammatory protein secretion in vitro (Fig. 2A-C) and ex vivo (Fig. 2D and E). In contrast, no significant differences were observed in the secretome between IL-10 single and combined treatments compared to control group. Overall, IL-4 containing treatments promote human NP cell and explant catabolism in contrast to previously reported IL-4 anti-inflammatory performance. 4. Thus, a possible pleiotropic effect of IL-4 could occur depending on the IVD culture and environmental condition. Acknowledgements: This project was supported by the Marie Skłodowska Curie International Training Network “disc4all” under the grant agreement #955735. For any figures and tables, please contact the authors directly


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 238 - 238
1 Jul 2014
Deschepper M Paquet J Manassero M Logeart-Avramoglou D Bensidhoum M Petite H
Full Access

Summary. In this study, we challenged the current paradigm of human Mesenchymal Stem Cells survival, which assigned a pivotal role to oxygen, by testing the hypothesis that exogenous glucose may be key to their survival. Introduction. The survival of human mesenchymal stem cells (hMSCs) has elicited a great deal of interest, because it is relevant to the efficacy of engineered tissues. However, to date, hMSCs have not met this promise, in part due to the high death rate of cells upon transplantation. In this study, we challenged the current paradigm of hMSC survival, which assigned a pivotal role to oxygen, by testing the hypothesis that exogenous glucose may be key to hMSC survival. Materials and methods. In vitro model of ischemia 2.10. 4. hMSCs from five donors, were seeded into individual wells of a 24-well plate, cultured overnight, washed twice with PBS and then maintained in hypoxia (0.1% oxygen) under serum (FBS) free αMEM medium in either the absence or in the presence (1 or 5 g/L) of glucose for 21 days. In vitro Cell viability: To assess the role of glucose on hMSCs viability, cells were cultured under hypoxia in the absence or in the presence of glucose (1 and 5g/L), At days 0, 3, 7, 14 and 21, cell viability was evaluated by flow cytometry and ATP content per cell quantified. In vivo effect of glucose supply on hMSCs viability. 3.10. 5. eGFG-luc hMSCs were seeded on a cylindrical AN-69 scaffolds. At the time of implantation, 100 µl of hyaluronic acide (HA) (2%) containing either 0g/L (negative control) or 10g/L of glucose was gently injected inside the construct. Cell- constructs were implanted subcutaneously in eight week-old mice (2 per animal) and were imaged by bioluminescence imaging (BLI) at day 1, 4, 7 and 14 until sacrifice. Results. hMSCs were able to survive and to maintain their ATP content 21 days under sustained hypoxia providing that they were cultured in the presence of a sufficient glucose supply (i.e. 5g/L). In contrast, hMSCs cultured without or with 1g/L of glucose failed to survive. These results established that glucose depletion but not sustained hypoxia affected cell survival. In vivo results showed a striking increase of cell viability in cell constructs loaded with glucose. At day 14, a five-fold increase in cell number was observed in cell constructs loaded with glucose when compared to the control cell constructs without glucose. Discussion. The present study challenge the current paradigm that gives a pivotal role to oxygen on hMSCs massive cell death. By using an in vitro model of hypoxia/ischemia, we demonstrated that in the presence of sufficient glucose, hMSCs were able to survive 21 days under sustained hypoxia. Most importantly, an appropriate glucose supply strongly increases cell viability of hMSCs implanted subcutaneously in a mice model. This study provides evidences that glucose depletion but not hypoxia affects hMSCs viability. Further investigations need to be performed to develop hydrogels that ensure continuous glucose delivery to the implanted cells. Theses findings are particularly relevant because they pave the way to the development of new delivery systems to ensure hMSCs viability in order to increase their therapeutical potential after implantation


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_15 | Pages 39 - 39
1 Nov 2018
Aubert L Entz L Dubus M Rammal H Mongaret C Lesieur J Gangloff SC Chaussain C Mauprivez C Kerdjoudj H
Full Access

Mesenchymal stem cells (MSCs) are tissue-resident stroma cells capable of modulating immune cells through the secretion of paracrine factors. However, the comparison of MSCs potential, from different sources and submitted to hypoxia within a 3D scaffold, in secreting pro-healing factors has never been investigated. With a chemical composition similar to type I collagen, a major component of connective tissues retrieved in dental pulp, bone and umbilical cord, Hemocollagene® haemostatic foam presented porous and interconnected structure (> 90%) and a relative low elastic modulus of around 60 kPa. All these criteria meet basic requirements for tissue engineering based material. Herein, we assessed and compared the effect of hypoxia (3% O. 2. ) on the regulation and release of pro-angiogenic factors (VEGF, b-FGF and IL-8) from bone marrow (BM), Wharton's jelly (WJ) and dental pulp (DP) derived MSCs cultured in Hemocollagene®. After 10 days of culture, qRT-PCR analysis showed an up-regulation of b-FGF and VEGF mRNA in BM- and WJ-derived MSCs, but not in DP-derived MSCs. Furthermore, hypoxia highly up-regulated IL-8 expression in WJ-derived MSCs and moderately in both BM and DP-derived MSCs. In contrast, ELISA analysis showed a higher amount of VEGF and IL-8 in supernatant provided from DP-derived MSCs culture compared to BM and WJ-derived MSCs. B-FGF was not detected whatever the experimental condition. In conclusion, MSCs derived from several tissues were able to release pro-angiogenic factors under hypoxic conditions. There was no clearly superior type of MSCs for therapeutic use, however DP-derived MSCs are likely to be more advantageous


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XXXVI | Pages 4 - 4
1 Aug 2012
McGuire C Walsh P Mulhall PK
Full Access

OBJECTIVES. Ischaemic preconditioning (IPC) is a phenomenon whereby tissues develop an increased tolerance to ischaemia and subsequent reperfusion if first subjected to sublethal periods of ischaemia. Despite extensive investigation of IPC, the molecular mechanism remains largely unknown. Our aim was to show genetic changes that occur in skeletal muscle cells in response to IPC. METHODS. Firstly, we established an in-vitro model of IPC using a human skeletal muscle cell line. Gene expression of both control and preconditioned cells at various time points was determined. The genes examined were HIF-1 alpha, EGR1, JUN, FOS, and DUSP1. HIF-1 alpha is a marker of hypoxia. EGR1, JUN, FOS and DUSP1 are early response genes and may play a role in the protective responses induced by IPC. Secondly, the expression of HSPB8 was examined in a cohort of preconditioned total knee arthroplasty patients. RESULTS. HIF-1 alpha was upregulated following 1 and 2 hours of simulated ischaemia (p = 0.076 and 0.841 respectively) verifying that hypoxic conditions were met using our model. Expression of EGR1, FOS and DUSP1 were upregulated and peaked after 1 hour of hypoxia (p = 0.001, <0.00, and 0.038 respectively). cFOS was upregulated at 2 and 3 hours of hypoxia. IPC prior to simulated hypoxia resulted in a greater level of upregulation of EGR1, JUN and FOS genes (p = <0.00, 0.047, and <0.00 respectively). HSPB8 was not significantly upregulated following IPC using the hypoxic model. It was, however, upregulated on an mRNA level in total knee arthroplasty patients (p = 0.15). CONCLUSION. This study has validated the use of our hypoxic model for studying IPC in-vitro. IPC results in a greater upregulation of protective genes in skeletal muscle cells exposed to hypoxia than in control cells. We have demonstrated hitherto unknown molecular mechanisms of IPC in cell culture and in patients undergoing TKA


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 117 - 117
1 Jul 2014
Deschepper M Paquet J Petite H
Full Access

Introduction. The use of mesenchymal stem cells in regenerative medicine remains a promising approach due to the ability of these cells to differentiate into a variety of cell types of mesodermal lineage. Today, however, it is not clear whether long-term differentiation of MSCs is necessary or alternatively whether the benefits of MSCs can be conferred by transitory paracrine effects (via secreted chemical compounds). Human MSCs secrete a broad variety of cytokines, chemokines and growth factors that may potentially be involved in tissue repair. Nevertheless, hMSCs secretome profile is closely related to cells biological and chemical environment (pO. 2. , inflammation, nutrients disponibility…). In the context of stem-cell-based regenerative medicine, upon implantation, hMSC are exposed to stresses such as ischemia, oxidative stress and inflammatory mediators. Knowledge of the paracrine properties of stem cells under hypoxic conditions is essential for planning appropriate strategies that overcome the potential negative impacts of all levels of low oxygen content (from hypoxiato anoxia) leading to ischemia and tissue necrosis pertinent to MSC-based tissue engineered constructs. Since the beneficial effects of stem cells may be confered predominantly indirectly through paracrine mechanisms, the present study was designed to characterise the hMSC secretome and to assess its biological effects considering oxygen level and nutrients disponibility. Methods. hMSCs were exposed in vitro either to sustain ischemic environment (pO. 2. ≤ 0.1%, serum deprived), to hypoxia (pO. 2. ≤ 0.1%, 5g/L glucose) or to normoxic conditions (pO. 2. =21%). We used an hypoxic station (Biospherix, US) to ensure sustained hypoxia during 21 days. Levels of angiogenic, chemo-attractant, inflammatory and immunomodulative mediators were assessed in supernatants using the luminex technology (milliplex KIT, Millipore, USA). The chemo-attractant potential of conditioned media (CM) was assessed in vitro using Boyden chambers (BD, USA). To assess angiogenic potential of CM, HEPC were seeded on matrigel for 18 hours with CM obtained from hMSCs. Functionnality of secreted mediators was also assessed in vivo: briefly, CM media were lyophilyzed on collagen sponges and ectopically implanted in nude mice. Chemo-attraction and vascularization of the implants were determined using histological and CTscan analysis. Results. This study shows that the release of mediators by hMSCs is a function of nutrient availability whatever the pO. 2. considered. Moreover, hypoxia promotes a specific secretome profile of chemo-attractive and pro-angiogenic mediators from hMSCs, inflammatory/immunomodulative mediators and growth factors are not expressed in a hypoxic environment. The bioactivity of these mediators was confirmed by in vitro and in vivo tests. In addition, hMSCs chemo-attractive and pro-angiogenic potential is increased by glucose supply. Discussion and Conclusion. By using an in vitro model of ischemia/hypoxia, we first demonstrated that hMSC secretome is deeply affected, quantitatively and qualitatively, by the presence or no of nutrients (ie glucose) under hypoxia. Moreover, the present study shows selectivity in mediators produced by hMSCs in a hypoxic environment. Immunomodulatives and inflammatory mediators currently described to be secreted by hMSCs, under “normoxic conditions”, are not expressed and secretome profile is focused on pro-angiogenic and chemo-attractive chemical compounds


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_16 | Pages 108 - 108
1 Nov 2018
Ivanovska A Grolli S Balogh L Conti V Ravanetti F Schneider A Pöstényi Z Gazza F Borghetti P Cacchioli A
Full Access

Mesenchymal stem/stromal cells (MSC) have the ability to home and migrate towards injured and inflamed tissues which can be useful as a minimally invasive systemic approach to deliver MSC to the site of damaged articular surface in arthritis in human and veterinary patients. From a molecular point of view, the CXCR4/SDF-1 plays an important role in this phenomenon and can be used as a target to enhance the therapeutic efficacy of culture expanded MSC. It has been demonstrated that extensive in vitro expansion down-regulates CXCR4 expression in human, murine and canine MSCs hindering their therapeutic efficacy. Therefore, the aim of the present study was to assess the effect of hypoxia and basic fibroblast growth factor (bFGF) pre-conditioning on CXCR4 and SDF-1 expression in canine adipose derived MSC (cAT-MSC). MSC were isolated from subcutaneous adipose tissue of two adult Beagle dogs (n=2; 3–5 years old, 9–12kg) and cultured under standard conditions (5%CO. 2. , 37°C). Cells at passage 3 were then cultured in hypoxia (2%O. 2. ) and normoxia, with supplementation of 1 and 5 ng/ml bFGF for 24h. MTT assay, flow cytometry, immunohistochemistry and qRT-PCR analysis were conducted to assess respectively the modulation effect on cell proliferation, CXCR4 protein expression and CXCR4 and SDF-1 gene expression. Cell proliferation increased proportionally with the increasing bFGF concentrations, with a statistically significant higher proliferative rate in normoxic conditions (p<0.05). The gene expression of CXCR4 and SDF-1 increased in hypoxic conditions with bFGF supplementation (p<0.05). bFGF supplementation increased cytoplasmatic expression of CXCR4 in hypoxic conditions (p<0.05), however the surface expression remained low in all culture conditions. The described pre-conditioning method can be used for the enhancement of the therapeutic potential of systemically administered canine AT-MSC and can have a relevant translational character for the optimization of culturing protocols of human adipose derived MSC


Orthopaedic Proceedings
Vol. 100-B, Issue SUPP_4 | Pages 83 - 83
1 Apr 2018
Hameister R Dheen ST Lohmann CH Kaur C Singh G
Full Access

Background. Mechanisms underlying implant failure remain incompletely described, though the presence of macrophage-mediated inflammatory reactions is well documented. Hypoxia has a critical role in many diseases and is known to be interdependent with inflammation. Metals used for joint replacements have also been reported to provoke hypoxia-like conditions. In view of this, we aim to investigate hypoxia-associated factors in aseptic loosening and osteoarthritis with a focus on macrophages. Methods. Western blotting, calorimetric assay, haematoxylin-eosin staining, immunohistochemistry, double-immunofluorescence and transmission electron microscopy were performed on capsular tissue obtained from patients undergoing primary implantation of a total hip replacement for osteoarthritis and from patients undergoing revision surgery for aseptic loosening to investigate the presence of hypoxia-associated factors. Results. Tissues from patients with osteoarthritis and aseptic loosening showed the presence of inflammatory cells, many of which were macrophages as confirmed with CD68 immunostaining. In aseptic loosening, macrophages containing metal particles were present in clusters. This was observed both at the light and electron microscopic levels. Under the electron microscope, endothelial cells appeared to be hypertrophied and some showed signs of degeneration. The presence of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and nitric oxide was demonstrated by western blotting and colorimetric assay. Macrophages were the predominant cell type to release HIF-1α, VEGF, inducible nitric oxide synthase (iNOS). This was confirmed by double-immunofluorescence showing co-localization of HIF-1α, VEGF, iNOS with the macrophage marker CD68. Endothelial cells were stained for endothelial nitric oxide synthase as assessed by immunohistochemistry. Conclusion. This study demonstrates the release of hypoxia-associated factors by macrophages. The presence of hypoxia-associated factors in both, osteoarthritis and aseptic loosening suggest that hypoxia may be a factor underlying both pathologic conditions. This study was supported by research grant (NMRC/CNIG/1147/2016) from National Medical Research Council (NMRC), Singapore


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 324 - 324
1 Jul 2014
Marmotti A Mattia S Bonasia DE Bruzzone M Terrando S Tarella C Ponzo E Blonna D Castoldi F Peretti GM Rossi R
Full Access

Summary Statement. Hypoxia enhances chondrocyte phenotype of cells migrating from cartilage fragments, thus supporting the use of chondral fragment as a potential cell source for one-stage cartilage repair. Introduction. Minced cartilage fragments are a viable cell source for one stage cartilage repair, as shown in both in preclinical and clinical studies. However, the joint microenvironment, in which the repair process takes place, is hypoxic and no evidences are present in literature regarding the behaviour of cartilage fragments in a hypoxic environment. Aim of the study is to verify if hypoxia could influence chondrocyte outgrowth from cartilage fragments into a Hyaluronic-Acid/fibrin scaffold and evaluate its effects on migrating chondrocyte behaviour, compared to normoxic condition. This could be significant in the perspective of a wide clinical application of human chondral fragments for single stage repair. Materials and methods. Constructs were prepared with minced cartilage fragments harvested from the trochlear region of 20 human young donors during ACL reconstruction, loaded onto a non-woven esterified Hyaluronic-Acid-derivative felt (Hyaff-11) and retained with a coating of fibrin glue (Tisseel). Constructs were cultured either in normoxic or in hypoxic (10% O. 2. ) condition. The growth medium contained DMEM-high-glucose (4500mg/l)with 10% fetal-bovine-serum. After 1 month, construct sections were stained with haematoxylin/eosin and Safranin-O and examined for cell counting. Expression of chondrocyte markers (SOX9, collagen-II, collagen-I), hypoxic markers(HIFs) and proliferative markers (beta-catenin, PCNA) was assessed using immunofluorescence. Results. Migrating cells predominantly showed a spindle-like shape when close to the fragments and a more roundish shape when embedded into the scaffold. A slight decrease in chondrocyte migration and proliferation was observed in hypoxic cultures, albeit not statistically significant. Conversely, an increase in the expression of SOX9, beta-catenin, HIFs, collagen-II (p<0.05) in migrating chondrocytes from hypoxic cultures was shown by immunofluorescence. Discussion/Conclusion. Hypoxia seems to improve the chondrocyte phenotype/behaviour of cell outgrowing from cartilage fragments onto a HA/fibrin scaffold. Moreover, hypoxic condition did not hamper the ability and the mechanisms by which chondrocytes migrate from cartilage fragments and proliferate into the surrounding environment. This is clinically relevant in order to validate one-step repair techniques by means of human cartilage fragments loaded into composite scaffolds


Orthopaedic Proceedings
Vol. 94-B, Issue SUPP_XVIII | Pages 17 - 17
1 May 2012
Khan W Dheerendra S Johnson D Andrew J Hardingham T
Full Access

INTRODUCTION. Bone marrow derived mesenchymal stem cells are a potential source of cells for the repair of articular cartilage defects. Hypoxia has been shown to improve chondrogenesis in adult stem cells. In this study we characterised bone marrow derived stem cells and investigated the effects of hypoxia on gene expression changes and chondrogenesis. MATERIALS AND METHODS. Adherent colony forming cells were isolated and cultured from the stromal component of bone marrow. The cells at passage 2 were characterised for stem cell surface epitopes, and then cultured as cell aggregates in chondrogenic medium under normoxic (20% oxygen) or hypoxic (5% oxygen) conditions for 14 days. Gene expression analysis, glycosoaminoglycan and DNA assays, and immunohistochemical staining were determined to assess chondrogenesis. RESULTS. Bone marrow derived adherent colony forming cells stained strongly for markers of adult mesenchymal stem cells including CD44, CD90 and CD105, and they were negative for the haematopoietic cell marker CD34 and for the neural and myogenic cell marker CD56. Interestingly, a high number of cells were also positive for the pericyte marker 3G5. Cell aggregates showed a chondrogenic response and in lowered oxygen there was increased matrix accumulation of proteoglycan, but less cell proliferation, which resulted in 3.2-fold more glycosoaminoglycan per DNA after 14 days of culture. In hypoxia there was increased expression of key transcription factor SOX6, and the expression of collagens II and XI, and aggrecan was also increased. DISCUSSION. Pericytes are a candidate stem cell in many tissue and our results show that bone marrow derived mesenchymal stem cells express the pericyte marker 3G5. The response to chondrogenic culture in these cells was enhanced by lowered oxygen tension, which up-regulated SOX6 and increased the synthesis and assembly of matrix during chondrogenesis. This has important implications for tissue engineering applications of bone marrow derived stem cells