• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 50
  • 31
  • 8
  • 8
  • 5
  • 3
  • 3
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 140
  • 64
  • 43
  • 23
  • 22
  • 21
  • 21
  • 20
  • 20
  • 15
  • 15
  • 14
  • 14
  • 14
  • 13
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
21

Expression and function of NG2/CSPG4 in human chondrocytes

Jamil, Nuor Sabah Mohammed January 2013 (has links)
Introduction: NG2/CSPG4 is a unique transmembrane chondroitin sulphate proteoglycan molecule expressed as a core protein and a chondroitin sulphate proteoglycan (CSPG) up to 400kD. NG2/CSPG4 mediates the communication between the extracellular and intracellular compartments through interactions with collagen VI, growth factors and the actin cytoskeleton. NG2/CSPG4 affects cell migration, spreading, apoptosis and proliferation processes. NG2/CSPG4 has been shown to be expressed in developing and adult cartilage where less is known of its function. I tested the hypothesis: NG2/CSPG4 is an important regulator of chondrocytes function and has the potential to be a therapeutic target for treatment of diseases of cartilage such as osteoarthritis and chondrosarcoma. To do this, I had the following aims: 1) investigate whether different types of chondrocytes show variation in the form or distribution of NG2/CSPG4 expression and 2) through a knockdown approach develop a model to study the functional roles of NG2/CSPG4 in human chondrocytes. Materials and Methods: JJ012, a chondrosarcoma cell line, chondrocytes derived from human articular cartilage and C20/A4 an immortalised chondrocyte cell line were used. NG2/CSPG4 expression was investigated by RT-PCR western blotting, flow-cytometry and immunocytochemistry. NG2/CSPG4 interaction with Golgi complex and endoplasmic reticulum (ER) was assessed by double immunofluorescence. Biochemical interactions were assessed by immunoprecipitation and mass spectroscopy. For NG2/CSPG knockdown, a viral transduction method was carried out using 5 different constructs. Different functional roles of NG2/CSPG4 were investigated. The role of NG2/CSPG4 in gene regulation was studied by shRNA knockdown of NG2/CSPG4 in JJ012 cells and RTPCR. Results: NG2/CSPG4 mRNA was detectable in all cells tested. Western blotting showed expression of only a 270kD core protein in JJ012 and C20A4 cells. Using two different anti NG2/CSPG4 antibodies human OA chondrocytes were seen to express multiple molecular weight forms differentially recognised with and without chondroitinase ABC pre-treatment. Expression of NG2/CSPG4 in JJ012 cells was predominantly membrane associated whilst in OA chondrocytes and C20A4 cells, additional, predominant punctuate cytoplasmic distribution was evident. In OA chondrocytes NG2/CSPG4 co-localised with the Golgi complex and ER. Immunoprecipitation and mass spectrometry data demonstrated associations between NG2/CSPG4 and both collagen VI and thrombopoietin in OA chondrocytes. A model of NG2/CSPG4 gene knockdown was achieved in JJ012 chondrosarcoma cell line, known as B3. B3 cells spread more and migrate less than JJ012 cells; with a significant difference observed in migration (after 10hours: the closed area was 81.4% for JJ012 and 54.6% for B3). There was no difference in cell adhesion to collagen I, II, VI and fibronectin. EGTA inhibited cell adhesion to fibronectin in dose dependent manner with no significant difference observed between both JJ012 and B3 cells. EDTA reduced adhesion of B3 cells but not JJ012 to fibronectin. A significant difference in cell proliferation was detected with no change in apoptosis. Following NG2/CSPG4 knockdown in JJ012 cells there was no difference in expression of aggrecan, collagen II and SOX-9. In contrast, B3 cells showed a decreased expression of MPP3 and ADAMTS-4, a complete loss of ADAMTS-5 and increased expression of MMP13. Conclusions: I have identified altered expression and multiple forms of NG2/CSPG4 in different types of chondrocytes and shown association of this molecule with type VI collagen and thrombopoietin. Creation of a chondrocyte cell line that has stable knockdown of NG2/CSPG4 allowed further investigation of NG2/CSPG function in chondrocytes. NG2/CSPG4 knockdown reduced the cellular migration and proliferation and increased the chondrocyte spreading. The adhesion mechanism in JJ012 appears to be calcium dependent. Loss of NG2/CSPG4 induced changes in expression of aggrecanases and MMPs. Altered expression or associations of NG2/CSPG4 with extracellular ligands or intracellular signalling cascades may be important in the pathogenesis of OA by regulating proteolytic activity or apoptosis related pathways. NG2/CSPG4 is a potential therapeutic target in degenerative and neoplastic diseases of cartilage.
22

Effects of glucocorticoids on chondrocytes and cartilage

Wallace, Chelsey 24 July 2018 (has links)
OBJECTIVE: Osteoarthritis (OA) is a leading cause of disability worldwide. This disease is characterized by the inflammation and degradation of the cartilage and surrounding tissue in a joint. The disease manifests as either a result of years of wear and tear or after a joint injury. Post-traumatic osteoarthritis, as this latter case is named, is frequently studied since the exact trigger of the disease is known. In addition to several changes within the joint space, a significant alteration is the degradation of cartilage caused primarily by the release of inflammatory cytokines including interleukin-1 and 6 and tumor necrosis factor α. One current pharmacological treatment for the pain caused by OA is an intra-articular injection of glucocorticoids such as dexamethasone. As this is a common treatment, the goal of this research was to determine if, at the cellular level, this treatment impacts cell viability in the presence of pro-inflammatory cytokines. Another goal was to investigate how such treatment affects the progression of cartilage degradation caused by cytokines. OA results in the loss of the key extracellular matrix molecule, aggrecan, which contains negatively charged glycosaminoglycan (GAG) chains. Measurement of the amount of GAGs lost is an early indicator of cartilage degradation. In addition, biosynthesis of GAG chains can be measured to estimate the overall metabolic health of the cells. We hypothesized that dexamethasone blunts the harmful effects of proinflammatory cytokines and improves GAG biosynthesis and chondrocyte viability. METHODS: Cylindrical cartilage explants were collected from bovine knee joints and trimmed to a uniform 3 millimeters in diameter and 1 millimeter thick. Each treatment group consisted of n=6 explants from the same knee joint. In one set of experiments, these explants were subjected to two different doses of interleukin-1α (1 ng/mL and 10 ng/mL) with and without dexamethasone at 100 nM. In another set of experiments, explants were subjected to both interleukin-1α and tumor necrosis factor-α (1 ng/mL and 25 ng/mL respectively). The explants were cultured in medium for 6 days and were digested for outcome measurements on the final day. On day 4, 35S-sulfate was added to the explant medium for later measurement of radiolabel incorporation as a measure of GAG biosynthesis. Cell viability was measured on day 5 using red/green fluorescent viability dyes fluorescein diacetate (FDA) which stains live cells green and propidium iodide (PI) which stains dead cells red. RESULTS: Compared with untreated controls, explants subjected to the pro-inflammatory cytokines interleukin-1α and tumor necrosis factor-α exhibited greater glycosaminoglycan loss and a decrease in GAG biosynthesis. These treatments also decreased cell viability. Addition of dexamethasone improved cell viability compared to treatment with the cytokines. In addition, dexamethasone prevented glycosaminoglycan loss and increased GAG biosynthesis in the presence of interleukin-1α. However, dexamethasone did not prevent tumor necrosis factor-α mediated loss of GAGs. CONCLUSION: These studies demonstrated that dexamethasone inhibited specific aspects of cartilage degradation associated with inflammation in early OA. This therapeutic counteracts the degradative changes initiated by inflammatory cytokines such as interleukin-1α without compromising cell viability. Future studies are needed to identify the mechanisms of dexamethasone action and the ideal concentration to use if it is to be used as a treatment for OA following acute joint injury.
23

The bioeffect of ultrasound on human chondrocytes

Cheng, Yi-Li 29 July 2005 (has links)
Animal and clinical studies have shown an acceleration of bone healing by the application of pulsed low-intensity ultrasound (PLIUS). Several studies have reported that pulsed low-intensity ultrasound increase the synthesis of proteoglycan and type II collagen of cultured animal chondrocytes. The objectives of this study were to exam the bioeffect of pulsed low-intensity ultrasound on in vitro cultured human chondrocytes. Human chondrocytes were isolated from the amputated polydactyly digit of six different 1 to 10 years patients and cultured in agarose suspension for 3 days before treatment. PLIUS with intensities of 3.6, 18, 48, 72 and 98 mW/cm2was respectively applied to human chondrocytes for a single 10-min per day treatment. A control group was treated without PLIUS. The results demonstrated that PLIUS-treated human chondrocytes increased the proteoglycan synthesis compared with the control in a time-dependent manner. It is shown that the effect of 48 mW/cm2 is the most potent among a variety of PLIUS intensities tested determined by ELISA method. PLIUS at 48 mW/cm2 also increased type II collagen synthesis by up to 48.5+8.0% of the control determined by western blotting analysis. However, PLIUS has no significant influence on the cell proliferation of human chondrocytes compared with the control. It revealed that the PLIUS can enhance extracellular matrix synthesis. The response to PLIUS of chondrocytes harvested from 1 year old donor was significantly better than that of chondrocytes of 10 years old patient. These observations may lead to a better understanding of the bioeffect of PLIUS on in vitro cultured human chondrovytes.
24

Morphological properties of articular chondrocytes in various experimental and clinical conditions

Karim, Asima January 2015 (has links)
Previous work has suggested that there exists a relationship between chondrocyte morphology and matrix metabolism. Changes to chondrocyte morphology have been reported in human cartilage however it is unclear if these are involved in the degenerative process associated with osteoarthritis (OA). In this work, the morphology of human and bovine chondrocytes has been characterised under a range of conditions. Bovine chondrocytes have been utilised in these experiments as bovine cartilage is non-degenerate and the chondrocytes have ‘normal’ morphology. However, if human cartilage have been used instead then there is possibility of having chondrocytes of mixed shapes i.e. both ‘normal’ and ‘abnormal’ cells. The thesis aimed at experimentally inducing morphological changes to chondrocytes to determine whether these changes resemble those observed in human cartilage. The ultimate aim is to model these changes to clarify the link between morphology and matrix metabolism by determining how morphological changes influence matrix metabolism. A classification system was developed for chondrocyte morphology allowing the quantification of chondrocyte shapes under different conditions permitting statistical comparisons. The different conditions utilised were (1) non-degenerate and mildly-degenerate human articular cartilage and (2) two in vitro models (a) weak 3D agarose gels to study the effect of gel strength and increasing concentrations of foetal calf serum (FCS) on morphology of bovine chondrocytes and (b) scalpel induced mechanically-injured bovine cartilage model to study in situ chondrocyte viability and morphology at the injured site in various culture conditions. Additionally, the effect of raised medium osmolarity on the response of chondrocytes to injury was studied to determine if the abnormal morphology could be reversed. Using fluorescence-mode confocal laser scanning microscopy (CLSM), chondrocyte viability, volume and morphology were determined and quantified by using VolocityTM 3D image analysis software. Histological evaluation of matrix by using Haematoxylin and eosin, Alcian blue and Masson’s trichrome staining of matrix produced by chondrocytes cultured in strong or weak agarose gels and in injured cartilage was determined. Additionally, immunohistochemical evaluation of matrix (collagen Types I & II) produced by chondrocytes was also performed. Results demonstrated that in non-degenerate human femoral head cartilage, ~83% chondrocytes were normal in morphology and 17±2% chondrocytes had cytoplasmic processes as compared to mildly-degenerate cartilage where 35±5% abnormal chondrocytes with cytoplasmic processes were present. In non-degenerate cartilage, 11±3% chondrocytes formed small sized clusters however clustering was quite evident in the superficial zone of mildly-degenerate human femoral head cartilage where 43±16% chondrocytes had formed large clusters. In mildly-degenerate cartilage the number of abnormal chondrocytes with processes, length of processes and number of processes per cell were greater in the superficial as compared to mid and deep zones. A model was developed to study the effect of external supporting agarose gel on chondrocyte morphology and also to determine the influence of FCS. Bovine chondrocytes cultured in weak gels after 7 days developed similar morphological changes as those observed in degenerate human cartilage. However, in the strong gels only few chondrocytes with morphological changes were present i.e. similar to non-degenerate cartilage. These morphological changes (development of clusters and processes) occurred more rapidly with increasing concentrations of FCS. Histology revealed less Alcian blue staining intensity around chondrocytes cultured in weak gels as compared to strong gels suggesting altered matrix produced by abnormal chondrocytes. FCS and gel strength were therefore proposed as related factors in regulating chondrocyte morphology. In the bovine injured cartilage explant model, after 14 days chondrocytes at the injury in the presence of FCS or synovial fluid (SF) produced morphological changes. These changes comprised cell enlargement, flattening, elongation and production of cytoplasmic processes. In the absence of FCS or SF, chondrocytes at the injury remained unaffected and were morphologically ‘normal’. Throughout the cartilage and even in the absence of subchondral bone, chondrocytes displayed morphological abnormalities in the presence of FCS or SF. These findings suggested that this is not the property of chondrocytes in the superficial layers alone rather it is due to the extent of penetration of the ‘factors’ into the matrix and there is no possibility of interference of injured site with osteocytes or bone factors. Histology revealed that these abnormal chondrocytes showed less staining with Alcian blue at the injury suggesting that these morphological changes might play a role in the changes to matrix metabolism. By raising the osmolarity of the culture medium these changes were inhibited and chondrocytes maintained their normal morphology. The results suggest that morphogenic/proliferative factors in FCS or SF and strength/damage to the matrix may be inter-related and act as potent controllers of chondrocyte morphology. Raised osmolarity was found to inhibit the morphological changes suggesting the possibly that hyperosmolarity can antagonise the effects of these factors. The key conclusions from the thesis were (a) in non-degenerate human femoral cartilage a large percentage of chondrocytes ~83% were normal in morphology and the rest were abnormal however in mildly-degenerate cartilage 35±5% abnormal chondrocytes with processes were present (b) the changes to chondrocyte morphology (development of clusters and processes) were exacerbated with cartilage degeneration (c) chondrocytes cultured in the weak gels produced morphological changes as compared to strong gels (d) chondrocytes at the injury displayed marked morphological changes in the presence of FCS or SF (e) by raising the medium osmolarity these morphological changes to chondrocytes at the injury were inhibited. These results show that chondrocyte morphology is complex and strongly dependent on the environmental settings. Experimental conditions were therefore identified which showed increased chondrocyte volume, abnormal morphology with cytoplasmic processes, enhanced proliferation/cluster formation and matrix changes. These changes to volume and morphology of chondrocytes in the models studied in this work had certain similarities to the changes observed in human cartilage suggesting that these shape changes may play a role in the changes to matrix metabolism occurring in OA. These findings may be of translational relevance in clinical and experimental research into cartilage injury and degeneration by providing new insights in understanding the role played by chondrocyte morphology in cartilage degeneration and injury.
25

Investigation of the role of Staphylococcus aureus toxins in a cartilage explant model of septic arthritis

Smith, Innes Donald Mackenzie January 2015 (has links)
Septic arthritis has the potential to be a highly destructive joint disease. Although numerous bacterial species are capable of inducing septic arthritis, Staphylococcus aureus is most commonly implicated, accounting for up to 65% of cases. Whilst this organism is known to produce a diverse array of potential virulence factors, studies investigating a variety of S. aureus-related infections have implicated alpha(Hla)-, beta(Hlb)- and gamma(Hlg)-haemolysins as key damaging toxins, with the ‘pore-forming’ Hla considered to be the most potent. The work presented in this study focused on gaining further insight into the interaction between S. aureus toxins and in situ chondrocytes during an episode of septic arthritis. An in vitro bovine osteochondral explant model of S. aureus-induced septic arthritis was developed in this study. Utilising fluorescence-mode confocal laser scanning microscopy (CLSM), the model, which avoided the complexities of a host immune response, permitted an assessment of the following: (1) the spatial and temporal quantification of in situ chondrocyte viability following exposure to both a laboratory ‘wild-type’ (S. aureus 8325-4) and clinical strains of S. aureus; (2) the influence of Hla, Hlb and Hlg on in situ chondrocyte viability through the use of specific ‘haemolysin-knockout’ mutant strains; (3) the influence of altered culture medium osmolarity and extracellular Ca2+ on Hla-induced in situ chondrocyte death; and (4) dynamic changes in intracellular Ca2+ within in situ chondrocytes following Hla exposure. S. aureus 8325-4 and S. aureus clinical strains rapidly reduced in situ chondrocyte viability ( > 45% chondrocyte death at 40hrs). The increased acidity, observed during bacterial culture, had a minimal effect on chondrocyte viability. Chondrocyte death commenced within the superficial zone (SZ) of cartilage and rapidly progressed to the deep zone (DZ). Simultaneous exposure of SZ and DZ chondrocytes to S. aureus 8325-4 toxins (achieved with the use of subchondral bone-free explants) demonstrated that SZ chondrocytes were more susceptible to the toxins than DZ chondrocytes. When explants were cultured in the presence of a selection of isogenic S. aureus mutants, with varying Hla, Hlb and Hlg production capabilities (all originating from S. aureus 8325-4), Hla-producing mutants induced significant in situ chondrocyte death compared to toxin deficient controls (Hla-Hlb-Hlg-). In contrast, mutants producing Hlb and Hlg in the absence of Hla were unable to induce significant chondrocyte death. Hla alone was therefore identified as the key damaging toxin to in situ chondrocyte viability. Raised culture medium osmolarity had no influence on Hla-induced in situ chondrocyte death. In the absence of Hla, a high extracellular Ca2+ concentration (20mM) had no influence on chondrocyte viability during the experimental period. Hla-induced chondrocyte death increased in the presence of raised extracellular Ca2+ concentrations thereby confirming a role of Ca2+ in the chondrocyte death pathway. There was no significant difference between S. aureus growth in high and low Ca2+ culture media. Finally, when live osteochondral explants stained with the Ca2+-sensitive fluorophore Fluo-4 were cultured with an Hla-containing S. aureus supernatant (S. aureus 8325-4 (Hla+Hlb+Hlg+)) there was a significant rise in intracellular Ca2+ in comparison to those explants exposed to a non-Hla-containing supernatant (S. aureus DU5938 (Hla- Hlb-Hlg-)). The Hla-induced Ca2+ transients were always followed by chondrocyte death. Thus, it is likely that Hla-induced chondrocyte death was associated with a rise in intracellular Ca2+. These findings are of translational relevance. Firstly, toxins released by S. aureus have a rapid and fatal action on in situ chondrocytes, thereby advocating the prompt and thorough removal of bacteria and their toxins during the treatment of septic arthritis. Secondly, the identification of Hla alone as the key damaging toxin to in situ chondrocyte viability, with its destructive action being associated with a rise in intracellular Ca2+, may enable the development of future targeted therapeutic strategies in order to reduce the extent of cartilage destruction during and after an episode of septic arthritis.
26

Characterization of the in vitro growth and differentiation capabilities of human adipose-derived mesenchymal progenitor cells

Skritakis, Pantos Angelo 14 June 2019 (has links)
BACKGROUND: Human mesenchymal progenitor cells are multipotent cells that can be harvested from various adult and fetal tissues. They exhibit the potential to differentiate into several cell lineages, most notably osteogenic, chondrogenic, and adipogenic lineages. Conditions such as osteoporosis, metabolic disease, and arthritis are examples of dysfunction of tissues formed by the mesenchyme. The inability of these conditions to be healed by the body’s own mechanisms has raised considerable interest in the potential of using mesenchymal progenitor cells as a therapeutic intervention. This concept opens the possibility of harvesting mesenchymal progenitor cells from an individual, growing them into the desired tissue, and implanting them back into the individual. Treatment of this nature is much less invasive than current methods, overcomes rejection by the immune system, and could potentially demonstrate better outcomes in individuals suffering from degenerative disease of the mesenchyme. AIMS/OBJECTIVES: The aims of this study were to determine and to characterize the differentiation of human adipose-derived mesenchymal progenitor cells into osteocytes, chondrocytes, and adipocytes. The differentiation capacity of the mesenchymal progenitor cells was evaluated through cell staining, immunofluorescence, and RNA sequencing. METHODS: Subcutaneous adipose tissue was collected from patients undergoing elective panniculectomies. The abdominal panniculus was liposuctioned, and small explants of fat were embedded in Matrigel. Mesenchymal progenitor cells were extracted from the explants and plated for differentiation into osteogenic, chondrogenic, and adipogenic lineages. Control cells were grown in parallel in basal media to confirm differentiation. Dye staining for differentiation was performed with Alizarin Red S, Alcian Blue, and Oil Red O, and immunofluorescence staining was performed to indicate lineage-specific markers for differentiation. RNA sequencing was also completed on the different cell lineages. RESULTS: Human adipose-derived mesenchymal progenitor cells displayed the capacity to differentiate into osteogenic, chondrogenic, and adipogenic lineages as evidenced by dye staining. Osteogenic differentiation was confirmed with Alizarin Red S staining of calcium deposits in the differentiated cells, whereas staining in the control resulted in no calcium deposits. Alcian Blue staining confirmed chondrogenic differentiation as glycoproteins secreted by the differentiated cells were evident and different in morphology compared with the control cells. Oil Red O staining indicated adipogenic differentiation by showing lipid droplets in the differentiated cells and no lipid droplets in the control. RNA sequencing provided support that lineage differentiation was successful. Immunofluorescence staining further proved that differentiated cells expressed lineage-specific proteins and demonstrated morphological differences. CONCLUSIONS: This study demonstrates that mesenchymal progenitor cells harvested from human adipose tissue have the potential to differentiate into adipogenic, chondrogenic, and osteogenic cell lineages when induced with differentiation media. The differentiation of these cells can be assessed with dye staining, RNA sequencing, and immunofluorescence staining methods. Further studies should be done to investigate the potential of mesenchymal progenitor cells for therapeutic interventions in the treatment of various illnesses related to the mesenchyme.
27

Scx[+]/Sox9[+] progenitors contribute to the establishment of the junction between cartilage and tendon/ligament / Scx[+]/Sox9[+]前駆細胞は軟骨と腱/靭帯の連結部の構築に寄与する

Sugimoto, Yuki 23 January 2014 (has links)
京都大学 / 0048 / 新制・論文博士 / 博士(医学) / 乙第12802号 / 論医博第2074号 / 新制||医||1001(附属図書館) / 80846 / 京都大学大学院医学研究科医学専攻 / (主査)教授 妻木 範行, 教授 戸口田 淳也, 教授 松田 秀一 / 学位規則第4条第2項該当 / Doctor of Medical Science / Kyoto University / DFAM
28

Caracterisations structurales et fonctionnelles des glycosaminoglycannes matriciels dans le cartilage humain : pour une utilisation spécifique de GAG et de cellules souches pour la réparation du cartilage dans l'Osteoarthrose. / Structural and functional characterization of matrix glycosaminoglycans of the human cartilage : For a specific use of GAGs and stem cells for cartilage repair in Osteoarthritis.

Shamdani, Sara 29 March 2018 (has links)
L'Osteoarthrose (OA) est la maladie articulaire la plus répandue avec un impact socio-économique croissant en raison du vieillissement de la population, de l’augmentation de l'obésité et surtout de l'absence d'un traitement efficace. En effet, l’OA est caractérisée par la dégradation inéluctable du cartilage articulaire, l'apoptose des chondrocytes, un remodelage osseux sous-chondral et une inflammation de la synovie. La matrice extracellulaire (MEC) du cartilage est constituée de collagènes et de protéoglycanes (PG) eux-mêmes composés de glycosaminoglycanes (GAG) liés à un corps proteique, présents dans l'ECM ou à la surface cellulaire. Les GAG sont des chaînes polysaccharidiques linéaires sulfatées comprenant les Héparine/Héparan Sulfate (Hep/HS), Chondroitin Sulfate (CS) et Keratan Sulfate (KS). L'acide hyaluronique (AH) est un GAG non sulfaté particulier, non associé à un corps proteique. Dans le cartilage, l'un des principaux composants de la MEC est l'aggrécan, un CS/KS PG qui forme des aggrégats par interaction avec de l’AH. Au cours du vieillissement, des changements dans la qualité des PG ouvrent la voie à l’OA et les études depuis 60 ans se concentrent sur les aggrécans et le catabolisme des CS. En effets, les niveaux d'expression des CS, la taille de leurs chaînes, leurs profils de sulfatation évoluent, affectant les propriétés mécaniques de la MEC. Cependant, les traitements actuels de visco-supplémentation à base d’injections locales de CS ont démontré leur limite puisque la réparation du cartilage n'est pas induite. Même si ils sont rares dans le cartilage adulte, les HSPG sont associés aux chondrocytes et leurs rôles a été démontrée lors du développement osseux. Or les HS sont des régulateurs de l’homéostasie très importants car ils peuvent lier et réguler l'activité de protéines liant l'héparine (HBP) (facteurs de croissance, cytokines, chimiokines, morphogènes), les protégeant contre la protéolyse et potentialisant leur liaison à leurs récepteurs. Tous ces effets sont contrôlés par les profils de sulfatation complexes des chaînes d’HS.Dans ce contexte les objectifs de cette thèse sont de caractériser l'évolution de la signature chimique et de la fonctionnalité des HS au cours de l’OA. En collaboration avec les Rhumatologistes et Orthopédistes de l’Hopital Henri Mondor, une évaluation quantitative des HS dans des échantillons de cartilage humain contrôle versus OA a été corrélée à la gravité des dommages. Grace à la plateforme glycomic du CRRET, des modifications dans les profils de sulfatation des disaccharides de HS ont été observées et confirmées par des analyses de l'expression des enzymes de la biosynthèse des GAG. Ces caractéristiques structurales ont été corrélées à des changements fonctionnels de l’affinité des GAG pour des HBP, telles que FGF-2, VEGF et PTN. Enfin, les GAG OA ont des capacités différentes à moduler les propriétés (prolifération, adhésion, phénotype) de cellules souches mésenchymateuses, chondrocytes, fibroblastes et cellules endothéliales. Ces résultats démontrent que des modifications des structures et fonctions des HS pourraient être impliquées dans l'évolution de l'homéostasie du cartilage vers des processus pathologiques au cours de l’OA. Ce projet se positionne clairement comme une recherche fondamentale et translationnelle qui permettra d'acquérir des connaissances sur les mécanismes régulant les interactions cellules/matrice au cours de l'OA. De plus, les outils développés au cours de ce projet ont permis de réaliser 2 projets collaboratifs sur l'hypertension artérielle pulmonaire et une pathologie éosophagique. Enfin, ces données confirment l'intérêt d’identifier de nouvelles cibles glycaniques basées sur la chimie des HS. Cela permettra de proposer une nouvelle stratégie thérapeutique basée sur des composes à même de contrôler les profils de sulfatation de la MEC, dans le but d'améliorer les propriétés de cellules souches thérapeutiques endogènes ou exogènes, associées. / Osteoarthritis (OA) is the most prevalent joint disease with increasing socio-economic impact due to population aging, obesity , and absence of an efficient medical treatment that can repair cartilage. OA is characterized by degradation of articular cartilage, hypertrophy and apoptosis of chondrocytes, subchondral bone remodeling and joint synovial inflammation. Cartilage extracellular matrix (ECM) consists of collagens, glycoproteins and proteoglycans (PGs) that are composed of Glycosaminoglycans (GAGs) linked to core proteins, presents in the ECM or at the cell surface. GAGs are linear polysaccharidic sulfated chains including Heparine/Heparan Sulfate (Hep/HS), Chondroitin Sulfate (CS) and Keratan Sulfate (KS) families. Hyaluronic acid (HA) is a particular un-sulfated GAG no associated to core protein. In cartilage, one of the major ECM component is aggrecan, a CS/KS PG that form aggregate through HA interaction. During the aging process, changes in PGs quality pave the way for OA and studies are focus on aggrecans and CS catabolism since 60 years. CS expression levels, chain size, sulfation patterns evolved during OA, affecting the mechanical properties of ECM. However, treatments based on visco-supplementation with CS local injections have demonstrated their limit since cartilage repair is not induced. Even if rare in adult cartilage, HSPG are present associated to chondrocytes also and their relevance was demonstrated mainly during bone development. HS chains are very important homeostatic regulators because they are able to bind and regulate the activity of several heparin binding proteins (HBP) (growth factors, cytokines, chemokines, morphogens), protecting them against proteolysis and potentiating their binding to their receptors. These interactions provide a stock of regulatory factors that can be release by selective degradation of the HS chains too. All these regulatory effects are mediated through the complex sulfation/acetylation pattern of HS chains but no data are available on this aspect during OA.In this context, the goals of this Thesis were to characterize the evolution of HS chemical signature and functionality during OA. In collaboration with Rheumatology and Orthopedic clinical teams from Henri Mondor Hospital, a quantitative evaluation of HS and CS amount in control versus OA human cartilage samples was correlated to the structural damage severity. According to the tools of the CRRET’s lab glycomic platform, structural changes on HS and CS sulfated disaccharides compositions was observed using HPLC, confirmed by RQ-PCR analyzes of the expression of enzymes involved in GAG biosynthesis. These structural features were correlated to functional changes on HBP affinities, such as FGF-2, VEGF and PTN, through ELISA based competition assay. Finally, GAGs from OA have different abilities to modulate properties (adhesion proliferation, phenotype…) of Mesenchymal Stem Cells, chondrocytes, fibroblast and endothelial cells. These results clearly make the proof that modifications of HS structures and functions could be involved in the evolution of cartilage homeostasis and pave the way for altered pathological processes during OA. This project is clearly positioned as a fundamental and translational research that will permit to gain knowledge on the mechanisms regulating cartilage cells/matrix interactions during OA. All these results are summarized in 2 scientific and 1 review articles. Moreover, all the tools developed during this project have permit to realize 2 collaborative projects and associated articles on Pulmonary Hypertension and Eosophagic pathology also. Finally, all these data confirmed the interest of the team to identify new glycanic targets based on HS chemistry. This will permit to propose new therapeutic strategy based on HS compounds associated to endogenous or exogenous therapeutic stem cells, with the aim of improving cell properties according to HS ability to control sulfation panels of ECM.
29

Expression and regulation of microsomal prostaglandin E synthase-1 in human osteoarthritic cartilage and chondrocytes

Xinfang, Li January 2005 (has links)
Mémoire numérisé par la Direction des bibliothèques de l'Université de Montréal.
30

Implication du miR-24 et du miR-199a-5p dans le vieillissement prématuré du chondrocyte au cours de l'arthrose / Implication of miR-24 et du miR-199a-5p in cartilage premature aging during osteoarthritis

Philipot, Didier 07 December 2012 (has links)
L'arthrose tardive est la plus répandue des maladies ostéo-articulaires dont la prévalence augmente avec l'âge. Dans le cartilage arthrosique, des changements spécifiques des chondrocytes s'opèrent. Ils présentent une diminution de leur propriété de synthèse de la matrice extracellulaire, une diminution de leur réponse aux facteurs de croissance anabolisants et une augmentation de la sénescence cellulaire. Elle est caractérisée par un arrêt irréversible du cycle cellulaire, une érosion des télomères, une activation de la voie de dommages à l'ADN (ATM/p53/p21), une activation de la voie p16INK4a/pRb, l'établissement d'un sécrétome associé à un phénotype sénescent/hypertrophique appelé SAPS. Le sujet de ma thèse porte sur l'identification de microARNs impliqués dans le vieillissement prématuré du chondrocyte. Les microARNs (miRs) sont des petits ARNs non codant endogènes qui contrôlent un certain nombre de fonctions biologiques comme la prolifération, la différenciation ou la sénescence. Deux études ont montré le rôle préventif des miRs dans l'induction de la sénescence et dans l'hypertrophie. Au cours de ma thèse, nous avons utilisé un modèle de chondrocytes arthrosiques en 3D traités à l'IL-1β afin de récapituler le phénotype sénescent observé dans la pathologie. Cela nous a permit d'identifier deux miRs réprimés en réponse à cette cytokine : miR-24 et miR-199a-5p. Nous montrons que la répression de miR-24 conduit à une induction de p16INK4a et MMP1 associé à un phénotype hypertrophique. De plus, nos données préliminaires montrent que le miR-199a-5p est potentiellement un régulateur négatif de l'hormone anti-vieillissement Klotho qui est retrouvée dérégulée dans notre modèle cellulaire / Osteoarthritis (OA) is an age-related disease whose prevalence increases with late life. In osteoarthritic cartilage, chondrocytes presents age-specific changes such as a decrease in synthesis properties, a decrease in their response to growth and anabolic factors and an increase of cellular senescence. Senescent chondrocytes are characterized by an irreversible cell cycle arrest, DNA damage response activation (ATM/p53/p21), p16INK4a/pRb signaling pathway activation and the establishment of SAPS triggering to hypertrophy. The aim of my PhD project consisting to identify microRNAs involved in chondrocyte premature aging. microRNAs are small endogenous RNAs controlling several biological processes such as proliferation, differentiation and senescence. Two studies show that microRNAs have a preventive role in senescence and hypertrophy. During my PhD, we perform a cellular model based on OA chondrocytes placed in 3D and treated with IL-1β. We identified two miRs: miR-24 and miR-199a-5p. Repression of miR-24 leads to the induction of p16INK4a and MMP1, associated with chondrocyte hypertrophy. Moreover, preliminary datas suggests that miR-199a-5p is a potential regulator of anti-aging hormone Klotho which is deregulated in our model.

Page generated in 0.047 seconds