Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Acta Biomater ; 13: 150-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25462844

ABSTRACT

Receptor activator of nuclear factor kappa-B (RANK) and RANK-ligand are relevant targets for the treatment of polyethylene particle-induced osteolysis. This study assessed the local administration of siRNA, targeting both human RANK and mouse Rank transcripts in a mouse model. Four groups of mice were implanted with polyethylene (PE) particles in the calvaria and treated locally with 2.5, 5 and 10 µg of RANK siRNA or a control siRNA delivered by the cationic liposome DMAPAP/DOPE. The tissues were harvested at day 9 after surgery and evaluated by micro-computed tomography, tartrate-resistant acid phosphatase (TRAP) immunohistochemistry for macrophages and osteoblasts, and gene relative expression of inflammatory and osteolytic markers. 10 µg of RANK siRNA exerted a protective effect against PE particle-induced osteolysis, decreasing the bone loss and the osteoclastogenesis, demonstrated by the significant increase in the bone volume (P<0.001) and by the reduction in both the number of TRAP(+) cells and osteoclast activity (P<0.01). A bone anabolic effect demonstrated by the formation of new trabecular bone was confirmed by the increased immunopositive staining for osteoblast-specific proteins. In addition, 5 and 10 µg of RANK siRNA downregulated the expression of pro-inflammatory cytokines (P<0.01) without depletion of macrophages. Our findings show that RANK siRNA delivered locally by a synthetic vector may be an effective approach for reducing osteolysis and may even stimulate bone formation in aseptic loosening of prosthetic implants.


Subject(s)
Gene Expression Regulation/drug effects , Genetic Vectors , Osteolysis , Polyethylene/toxicity , RNA, Small Interfering , Receptor Activator of Nuclear Factor-kappa B , Acid Phosphatase/metabolism , Animals , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/pharmacology , HEK293 Cells , Humans , Isoenzymes/metabolism , Liposomes , Mice , Osteoblasts/metabolism , Osteoblasts/pathology , Osteolysis/chemically induced , Osteolysis/genetics , Osteolysis/metabolism , Osteolysis/pathology , Osteolysis/therapy , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Receptor Activator of Nuclear Factor-kappa B/genetics , Tartrate-Resistant Acid Phosphatase
2.
Ann Rheum Dis ; 71(1): 150-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22039170

ABSTRACT

OBJECTIVES: Interleukin (IL) 34 is a new cytokine implicated in macrophage differentiation and osteoclastogenesis. This study assessed IL-34 expression in the tissue of patients with rheumatoid arthritis (RA). METHODS: Immunohistochemistry was performed in synovial biopsies from patients with RA (n=20), osteoarthritis (n=3) or other inflammatory arthritis (n=4). IL-34 was detected in the synovial fluid by ELISA and its messenger RNA expression was studied by quantitative PCR in rheumatoid synovial fibroblasts after stimulation by tumour necrosis factor α (TNFα) and IL-1ß. Wild-type, jnk1(-/-)-jnk2(-/-) and nemo(-/-) murine fibroblasts and pharmacological inhibition were used to determine the involvement of nuclear factor kappa B (NF-κB) and JNK in that effect. RESULTS: IL-34 was expressed in 24/27 biopsies, with three samples from RA patients being negative. A significant association was found between IL-34 expression and synovitis severity. Levels of IL-34 and the total leucocyte count in synovial fluid were correlated. TNFα and IL-1ß stimulated IL-34 expression by synovial fibroblasts in a dose/time-dependent manner through the NF-κB and JNK pathway. CONCLUSION: This work for the first time identifies IL-34 expression in the synovial tissue of patients with arthritis. This cytokine, as a downstream effector of TNFα and IL-1ß, may contribute to inflammation and bone erosions in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Interleukins/metabolism , Synovitis/metabolism , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/pharmacology , Interleukins/genetics , MAP Kinase Signaling System/physiology , Male , Middle Aged , NF-kappa B/physiology , Osteoarthritis/genetics , Osteoarthritis/metabolism , RNA, Messenger/genetics , Synovial Fluid/metabolism , Synovitis/etiology , Synovitis/genetics , Tumor Necrosis Factor-alpha/pharmacology
3.
J Cell Mol Med ; 12(3): 928-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18494934

ABSTRACT

We recently demonstrated original anti-tumor effects of zoledronic acid (Zol) on osteosarcoma cell lines independently of their p53 and Rb status. The present study investigated the potential Zol-resistance acquired by osteosarcoma cells after prolonged treatment. After 12 weeks of culture in the presence of 1 microm Zol, the effects of high doses of Zol (10-100 microm) were compared between the untreated rat (OSRGA, ROS) and human (MG63, SAOS2) osteosarcoma cells and Zol-pretreated cells in terms of cell proliferation, cell cycle analysis, migration assay and cytoskeleton organization. Long-term treatment with 1 microm Zol reduced the sensitivity of osteosarcoma cells to high concentrations of Zol. Furthermore, the Zol-resistant cells were sensitive to conventional anti-cancer agents demonstrating that this resistance process is independent of the multidrug resistance phenotype. However, as similar experiments performed in the presence of clodronate and pamidronate evidenced that this drug resistance was restricted to the nitrogen-containing bisphosphonates, we then hypothesized that this resistance could be associated with a differential expression of farnesyl diphos-phate synthase (FPPS) also observed in human osteosarcoma samples. The transfection of Zol-resistant cells with FPPS siRNA strongly increased their sensitivity to Zol. This study demonstrates for the first time the induction of metabolic resistance after prolonged Zol treatment of osteosarcoma cells confirming the therapeutic potential of Zol for the treatment of bone malignant pathologies, but points out the importance of the treatment regimen may be important in terms of duration and dose to avoid the development of drug metabolic resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Diphosphonates/pharmacology , Drug Resistance, Neoplasm/drug effects , Geranyltranstransferase/metabolism , Imidazoles/pharmacology , Osteosarcoma/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Osteosarcoma/diagnosis , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Small Interfering/pharmacology , Time Factors , Transfection , Zoledronic Acid
4.
Oncogene ; 26(46): 6653-64, 2007 Oct 11.
Article in English | MEDLINE | ID: mdl-17471233

ABSTRACT

Oncostatin M (OSM), a cytokine of the interleukin-6 family, induces growth arrest and differentiation of osteoblastic cells into glial-like/osteocytic cells. Here, we asked whether OSM regulates apoptosis of normal or transformed (osteosarcoma) osteoblasts. We show that OSM sensitizes cells to apoptosis induced by various death inducers such as staurosporine, ultraviolet or tumor necrosis factor-alpha. Apoptosis is mediated by the mitochondrial pathway, with release of cytochrome c from the mitochondria to the cytosol and activation of caspases-9 and -3. DNA micro-arrays revealed that OSM modulates the expression of Bax, Bad, Bnip3, Bcl-2 and Mcl-1. Pharmacological inhibitors, dominant-negative signal transducer and activator of transcriptions (STATs), stable RNA interference and knockout cells indicated that the transcription factors p53 and STAT5, which are activated by OSM, are implicated in the sensitization to apoptosis, being responsible for Bax induction and Bcl-2 reduction, respectively. These results indicate that, in addition to growth arrest and induced differentiation, OSM also sensitizes normal and transformed osteoblasts to apoptosis by a mechanism implicating (i) activation and nuclear translocation of STAT5 and p53 and (ii) an increased Bax/Bcl-2 ratio. Therefore, association of OSM with kinase inhibitors such as Sts represents new therapeutic opportunities for wild-type p53 osteosarcoma.


Subject(s)
Apoptosis , Oncostatin M/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indole Alkaloids/pharmacology , Osteosarcoma , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT5 Transcription Factor , Tumor Suppressor Protein p53 , bcl-2-Associated X Protein/metabolism
5.
J Pathol ; 211(5): 555-562, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17323424

ABSTRACT

RANK, RANK ligand (RANKL) and osteoprotegerin (OPG) are the key regulators of bone metabolism, both in normal and pathological conditions. Previous data have demonstrated that human osteosarcoma biopsies express RANKL as well as OPG, and functional RANK is expressed in a murine osteosarcoma cell line. As RANK expression in human osteosarcoma remains controversial, the aim of the present study was to analyse its expression in vitro in human osteosarcoma cell lines, ex vivo using pathological tissues, and then to determine its functionality in terms of signal transduction pathways modulated by RANKL. RT-PCR analysis and immunohistochemistry experiments revealed that RANK is expressed at both transcriptional and protein levels in MNNG/HOS, Saos-2 and MG-63 human osteosarcoma cell lines, in contrast to the U-2 OS osteosarcoma cell line and human osteoblasts, which were negative. RANK was also expressed in 57% of osteosarcoma biopsies. Furthermore, western blot experiments clearly demonstrated the functionality of RANK. Thus, RANKL significantly induced the phosphorylation of ERK1/2, p38 and IkappaB in RANK-positive osteosarcoma cells. This study is the first report of functional RANK expression in human osteosarcoma cells: this strengthens the involvement of the RANK-RANKL-OPG axis in primary bone tumour biology and identifies novel therapeutic approaches targeting RANK-positive osteosarcoma.


Subject(s)
Bone Neoplasms/chemistry , Neoplasm Proteins/analysis , Osteosarcoma/chemistry , Receptor Activator of Nuclear Factor-kappa B/analysis , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Child , Cross-Sectional Studies , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Osteoblasts/chemistry , Osteoprotegerin/analysis , Phosphorylation , RANK Ligand/metabolism , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/physiology
6.
Biochim Biophys Acta ; 1644(1): 1-7, 2004 Feb 02.
Article in English | MEDLINE | ID: mdl-14741739

ABSTRACT

Osteoprotegerin (OPG) is a decoy receptor for receptor activator of nuclear factor kappaB ligand (RANKL), an inducer of osteoclastogenesis via its receptor RANK. We recently demonstrated that OPG also exerts a direct effect in osteoclasts by regulating protease expression. Herein, we showed that OPG-induced pro-matrix metalloproteinase-9 activity was abolished by ras/MAPK inhibitors in purified osteoclasts. OPG induced the phosphorylation of p38 and ERK1/2 in RAW264.7 cells. Only p38 activation was totally abolished by a blocking anti-RANKL antibody or an excess of RANKL. Surface plasmon resonance experiments revealed that RANK, RANKL and OPG are able to form a tertiary complex. These results suggested a potential formation of a tertiary complex RANK-RANKL-OPG on osteoclasts. Thus, OPG is not only a soluble decoy receptor for RANKL but must be also considered as a direct effector of osteoclast functions.


Subject(s)
Glycoproteins/physiology , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/pharmacology , Cell Line , Cells, Cultured , Enzyme Activation , Glycoproteins/antagonists & inhibitors , Glycoproteins/pharmacology , Ligands , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/pharmacology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoprotegerin , Phosphorylation , RANK Ligand , Rabbits , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Tumor Necrosis Factor , Signal Transduction/physiology , Surface Plasmon Resonance , Transfection , p38 Mitogen-Activated Protein Kinases
7.
Gut ; 48(1): 53-61, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11115823

ABSTRACT

BACKGROUND: Dietary fibres have been proposed as protective agents against colon cancer but results of both epidemiological and experimental studies are inconclusive. AIMS: Hypothesising that protection against colon cancer may be restricted to butyrate producing fibres, we investigated the factors needed for long term stable butyrate production and its relation to susceptibility to colon cancer. METHODS: A two part randomised blinded study in rats, mimicking a prospective study in humans, was performed using a low fibre control diet (CD) and three high fibre diets: starch free wheat bran (WB), type III resistant starch (RS), and short chain fructo-oligosaccharides (FOS). Using a randomised block design, 96 inbred rats were fed for two, 16, 30, or 44 days to determine the period of adaptation to the diets, fermentation profiles, and effects on the colon, including mucosal proliferation on day 44. Subsequently, 36 rats fed the same diets for 44 days were injected with azoxymethane and checked for aberrant crypt foci 30 days later. RESULTS: After fermentation had stabilised (44 days), only RS and FOS produced large amounts of butyrate, with a trophic effect in the large intestine. No difference in mucosal proliferation between the diets was noted at this time. In the subsequent experiment one month later, fewer aberrant crypt foci were present in rats fed high butyrate producing diets (RS, p=0.022; FOS, p=0.043). CONCLUSION: A stable butyrate producing colonic ecosystem related to selected fibres appears to be less conducive to colon carcinogenesis.


Subject(s)
Butyrates/metabolism , Colon/metabolism , Colonic Neoplasms/prevention & control , Dietary Fiber/administration & dosage , Animals , Azoxymethane , Carcinogens , Colon/pathology , Fermentation , Intestinal Mucosa/pathology , Oligosaccharides/administration & dosage , Rats , Rats, Inbred Strains , Starch/administration & dosage
8.
Biol Cell ; 92(7): 545-54, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11229605

ABSTRACT

Parietal cells of the gastric fundic mucosa are small and contain only a few tiny mitochondria when they begin to differentiate from mucous neck cells. The canalicular ATPase activity characteristic of mature parietal cells is discrete in these young cells, whereas areas of very high activity are apparent in the Golgi complex, reticulum, nuclear envelope, mitochondrial wall, and plasma membrane. Close relations and contacts occur between mitochondria and these organelles, and the size and number of mitochondria increase progressively. These relations, as well as mitochondrial ATPase activity (a true differentiation marker), cease once the mitochondria become as numerous and large as those of a mature parietal cell. Our observations suggest that a secondary form of mitochondrial biogenesis, involving the massive participation of other organelles and independent of the classical mechanisms inherent in mitosis, occurs in parietal cells at the beginning of G1 phase during the 6 days of their maturation.


Subject(s)
Cell Differentiation , Gastric Mucosa/cytology , Mitochondria/ultrastructure , Parietal Cells, Gastric/ultrastructure , 4-Nitrophenylphosphatase/analysis , Animals , Humans , Microscopy, Electron , Parietal Cells, Gastric/cytology , Parietal Cells, Gastric/enzymology , Rabbits
9.
Biol Cell ; 91(3): 195-208, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10425706

ABSTRACT

Ultrasmall superparamagnetic iron oxide (USPIO) contrast agents for use in magnetic resonance imaging (MRI) are currently undergoing clinical evaluation. However, the images observed and the kinetic profiles obtained differ from one agent to another. In this study, BD IX rats received an intravenous penis injection of the USPIO contrast agents AMI-HS and AMI-227. A cytologic study of the liver was performed, and the data obtained were compared with those of MRI. Images acquired in light microscopy, transmission electron microscopy, microanalysis and electron diffraction provided data on the cell categories involved in the processing of these contrast agents, the importance and modalities of each category relative to this processing, and the modalities of agent elimination. AMI-HS was rapidly removed from the bloodstream by Kupffer's cells and hepatocytes and then eliminated through bile ducts. AMI-227 remained much longer in the blood compartment since it was processed very slowly by endothelial and Kuppfer's cells in the near absence of hepatocytic participation and thus of elimination by the bile ducts. These results allowed us to base our interpretation of MRI sequences on cytologic observations.


Subject(s)
Contrast Media/pharmacokinetics , Ferric Compounds/pharmacokinetics , Liver/metabolism , Magnetic Resonance Imaging , Microscopy, Electron , Animals , Dextrans , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Ferrosoferric Oxide , Iron/metabolism , Kupffer Cells/metabolism , Kupffer Cells/ultrastructure , Liver/ultrastructure , Magnetics , Magnetite Nanoparticles , Male , Myocardium/metabolism , Myocardium/ultrastructure , Oxides/metabolism , Particle Size , Rats
10.
J Histochem Cytochem ; 44(10): 1131-40, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8813078

ABSTRACT

Biodegradation of ceramics in vivo is achieved essentially by monocytes and multinuclear cells (osteoclasts). Monocytes are the key element in this process because they intervene first at the biomaterial implantation site during inflammatory reaction. In this work, in vitro studies were conducted on an ultrastructural scale to determine the specific behavior of these cells with regard to a calcium phosphate (CaP) ceramic. Two types of phagocytosis were observed when cells came into contact with the biomaterial: either CaP crystals were taken up alone and then dissolved in the cytoplasm after disappearance of the phagosome membrane or they were incorporated together with large quantities of culture medium, in which case dissolution occurred after the formation of heterophagosomes. Phagocytosis of CaP coincided with autophagy and the accumulation of residual bodies in the cells. Addition of HILDA/LIF factor to these cultures induced a very marked decrease in phagocytotic activity directed at the capture of CaP crystals and culture medium. Autophagy was reduced, and residual bodies were rare or absent. This study specifies the role of monocytes in CaP biodegradation and demonstrates for the first time that HILDA/LIF has a biological effect on this cell line.


Subject(s)
Ceramics/metabolism , Durapatite/metabolism , Foreign-Body Reaction/pathology , Growth Inhibitors/pharmacology , Interleukin-6 , Lymphokines/pharmacology , Monocytes/metabolism , Phagocytosis , Animals , Biodegradation, Environmental/drug effects , CHO Cells , Cell Differentiation/drug effects , Cells, Cultured , Cricetinae , Foreign-Body Reaction/metabolism , Humans , Leukemia Inhibitory Factor , Macrophages/drug effects , Macrophages/metabolism , Macrophages/ultrastructure , Microscopy, Electron , Monocytes/drug effects , Monocytes/ultrastructure , Phagosomes/metabolism , Phagosomes/ultrastructure , Recombinant Proteins/pharmacology
11.
Anticancer Drug Des ; 10(6): 491-505, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7575990

ABSTRACT

We considered the usefulness of an association of two cytokines interferon gamma (INF gamma) and tumor necrosis factor alpha (TNF alpha) in optimizing intraperitoneal (i.p.) radioimmunotherapy of ovarian cancer. Studies were performed in conditions similar to those observed in vivo, using tumor multicell spheroids of the SHIN-3 ovarian adenocarcinoma line which expresses CA125 and O3 antigens. Light and electron microscopy showed that this cytokine association caused considerable modification of the three-dimensional morphology of the spheroids and the cells composing them. The uptake and retention kinetics of 125I-labeled F(ab')2 fragments of OC125 and OVTL-3 antibodies indicated that the presence of the cytokines led to a 1.5-fold increase in the quantity of O3 antigen at the spheroid surface. These results were confirmed by autoradiographs showing that the INF gamma-TNF alpha association produced extensive direct antitumor action, with better penetration and uptake of OVTL-3 antibody. Thus, i.p. radioimmunotherapy of micrometastases could be optimized by an initial injection of the IFN gamma-TNF alpha combination.


Subject(s)
Interferon-gamma/administration & dosage , Ovarian Neoplasms/therapy , Tumor Necrosis Factor-alpha/administration & dosage , Antibodies, Monoclonal/metabolism , Antibodies, Neoplasm/metabolism , Antigens, Neoplasm/metabolism , Combined Modality Therapy , Female , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/metabolism , Organoids , Radioimmunotherapy/methods , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...