Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Biochem J ; 478(16): 3145-3155, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34405859

ABSTRACT

Autophagy receptor p62/SQSTM1 signals a complex network that links autophagy-lysosomal system to proteasome. Phosphorylation of p62 on Serine 349 (P-Ser349 p62) is involved in a cell protective, antioxidant pathway. We have shown previously that P-Ser349 p62 occurs and is rapidly degraded during human synovial fibroblasts autophagy. In this work we observed that fingolimod (FTY720), used as a medication for multiple sclerosis, induced coordinated expression of p62, P-Ser349 p62 and inhibitory TFEB form, phosphorylated on Serine 211 (P-Ser211 TFEB), in human synovial fibroblasts. These effects were mimicked and potentiated by proteasome inhibitor MG132. In addition, FTY720 induced autophagic flux, LC3B-II up-regulation, Akt phosphorylation inhibition on Serine 473 but down-regulated TFEB, suggesting stalled autophagy. FTY720 decreased cytoplasmic fraction contained TFEB but induced TFEB in nuclear fraction. FTY720-induced P-Ser211 TFEB was mainly found in membrane fraction. Autophagy and VPS34 kinase inhibitor, autophinib, further increased FTY720-induced P-Ser349 p62 but inhibited concomitant expression of P-Ser211 TFEB. These results suggested that P-Ser211 TFEB expression depends on autophagy. Overexpression of GFP tagged TFEB in HEK293 cells showed concomitant expression of its phosphorylated form on Serine 211, that was down-regulated by autophinib. These results suggested that autophagy might be autoregulated through P-Ser211 TFEB as a negative feedback loop. Of interest, overexpression of p62, p62 phosphorylation mimetic (S349E) mutant and phosphorylation deficient mutant (S349A) in HEK293 cells markedly induced P-Ser211 TFEB. These results showed that p62 is involved in regulation of TFEB phosphorylation on Serine 211 but that this involvement does not depend on p62 phosphorylation on Serine 349.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fibroblasts/metabolism , Sequestosome-1 Protein/metabolism , Serine/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blotting, Western , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Fibroblasts/drug effects , Fingolimod Hydrochloride/pharmacology , HEK293 Cells , Humans , Immunosuppressive Agents/pharmacology , Leupeptins/pharmacology , Microscopy, Fluorescence , Mutation , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sequestosome-1 Protein/genetics , Serine/genetics , Synovial Membrane/cytology , Synovial Membrane/metabolism
2.
Oncotarget ; 9(88): 35830-35843, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-30533198

ABSTRACT

Phosphorylation of p62/SQSTM1 (p62) on Serine 349 (P-Ser349 p62) as well as proteasome dysfunction have been shown to activate the cell protective Keap1/Nrf2 pathway. We showed previously that BAY 11-7085-induced human synovial fibroblast cell death includes autophagy and p62 downregulation. In this work, we have studied expression of P-Ser349 p62 in human synovial fibroblasts. Results showed that P-Ser349 p62 was not detected in synovial cell extracts unless cells were cultured in the presence of proteasome inhibitor (MG132). MG132 revealed P-Ser349 p62 turnover, that was further increased by concomitant autophagy inhibition and markedly enhanced in serum starved cells. Starvation sensitized synovial fibroblasts to BAY 11-7085 while MG132 protected both non-starved and starved cells from BAY 11-7085-induced cell death. Lentivirus mediated overexpression of phosphorylation-mimetic p62 mutant S349E markedly protected synovial fibroblasts from BAY 11-7085. Inhibitor of Keap1-P-S349 p62 interaction, K67, had synergistic effect with MG132. Starvation increased p62 molecular weight, that was reversed by serum and bovine serum albumin re-feeding. Furthermore, starvation markedly induced RAD23B. Increased endo-ß-N-acetylglucosaminidase (ENGase) turnover was detected in starved synovial fibroblasts. PNGase F treatment produced faster migration p62 form in human synovial tissue extracts but starvation-like p62 form of higher molecular weight in synovial cell extracts. Co-transfection of NGLY1, with p62 or p62 mutants S349A and S349E markedly stabilized p62 expressions in HEK293 cells. Tunicamycin upregulated p62 and protected synovial fibroblasts from BAY 11-7085-induced cell death. These results showed that P-Ser349 p62 has pro-survival role in human synovial fibroblasts and that de-glycosylation events are involved in p62 turnover.

3.
Int J Mol Sci ; 17(12)2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27999417

ABSTRACT

Osteoarthritis (OA) is a joint pathology characterized by progressive cartilage degradation. Medical care is mainly based on alleviating pain symptoms. Compelling studies report the presence of empty lacunae and hypocellularity in cartilage with aging and OA progression, suggesting that chondrocyte cell death occurs and participates to OA development. However, the relative contribution of apoptosis per se in OA pathogenesis appears complex to evaluate. Indeed, depending on technical approaches, OA stages, cartilage layers, animal models, as well as in vivo or in vitro experiments, the percentage of apoptosis and cell death types can vary. Apoptosis, chondroptosis, necrosis, and autophagic cell death are described in this review. The question of cell death causality in OA progression is also addressed, as well as the molecular pathways leading to cell death in response to the following inducers: Fas, Interleukin-1ß (IL-1ß), Tumor Necrosis factor-α (TNF-α), leptin, nitric oxide (NO) donors, and mechanical stresses. Furthermore, the protective role of autophagy in chondrocytes is highlighted, as well as its decline during OA progression, enhancing chondrocyte cell death; the transition being mainly controlled by HIF-1α/HIF-2α imbalance. Finally, we have considered whether interfering in chondrocyte apoptosis or promoting autophagy could constitute therapeutic strategies to impede OA progression.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Cartilage, Articular/pathology , Chondrocytes/metabolism , Osteoarthritis/pathology , Aging , Apoptosis Regulatory Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Leptin/metabolism , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Arthritis Res Ther ; 18(1): 219, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27716396

ABSTRACT

BACKGROUND: Glucocorticoid-induced leucine zipper (GILZ) is a mediator of the anti-inflammatory activities of glucocorticoids. However, GILZ deletion does not impair the anti-inflammatory activities of exogenous glucocorticoids in mice arthritis models and GILZ could also mediate some glucocorticoid-related adverse events. Osteoarthritis (OA) is a metabolic disorder that is partly attributed to adipokines such as leptin, and we previously observed that glucocorticoids induced leptin secretion in OA synovial fibroblasts. The purpose of this study was to position GILZ in OA through its involvement in the anti-inflammatory activities of glucocorticoids and/or in the metabolic pathway of leptin induction. The influences of mineralocorticoids on GILZ and leptin expression were also investigated. METHODS: Human synovial fibroblasts were isolated from OA patients during knee replacement surgery. Then, the cells were treated with a glucocorticoid (prednisolone), a mineralocorticoid (aldosterone), a glucocorticoid receptor (GR) antagonist (mifepristone), a selective glucocorticoid receptor agonist (Compound A), mineralocorticoid receptor (MR) antagonists (eplerenone and spironolactone), TNF-α or transforming growth factor (TGF)-ß. Cells were transfected with shRNA lentiviruses for the silencing of GILZ and GR. The leptin, IL-6, IL-8 and matrix metalloproteinase (MMP)-1 levels were measured by ELISA. Leptin, the leptin receptor (Ob-R), GR and GILZ expression levels were analyzed by western blotting and/or RT-qPCR. RESULTS: (1) The glucocorticoid prednisolone and the mineralocorticoid aldosterone induced GILZ expression dose-dependently in OA synovial fibroblasts, through GR but not MR. Similar effects on leptin and Ob-R were observed: leptin secretion and Ob-R expression were also induced by prednisolone and aldosterone through GR; (2) GILZ silencing experiments demonstrated that GILZ was involved in the glucocorticoid-induced and mineralocorticoid-induced leptin secretion and Ob-R expression in OA synovial fibroblasts; and (3) GILZ inhibition did not alter the production of pro-inflammatory cytokines by OA synovial fibroblast or the anti-inflammatory properties of glucocorticoids. CONCLUSIONS: The absence of GILZ prevents corticoid-induced leptin and Ob-R expression without affecting the anti-inflammatory properties of glucocorticoids in OA synovial fibroblasts. Mineralocorticoids also induce leptin and Ob-R expression through GILZ.


Subject(s)
Fibroblasts/metabolism , Leptin/biosynthesis , Osteoarthritis, Knee/metabolism , Synoviocytes/metabolism , Transcription Factors/metabolism , Aged , Aged, 80 and over , Aldosterone/pharmacology , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Gene Knockdown Techniques , Glucocorticoids/pharmacology , Humans , Male , Middle Aged , Mineralocorticoids/pharmacology , Polymerase Chain Reaction , Prednisolone/pharmacology
5.
Oncotarget ; 7(17): 23370-82, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-26993765

ABSTRACT

Inhibition of proapoptotic pathways in synovial fibroblasts is one of the major causes of synovial proliferation and hyperplasia in rheumatic diseases. We have shown previously that NF-κB inhibitor BAY 11-7085, through inactivation of PPAR-γ, induces apoptosis in human synovial fibroblasts. In this work we showed that BAY 11-7085 induced autophagy that preceded BAY 11-7085-induced apoptosis. Of interest, BAY 11-7085 induced Serine 211 phosphorylation and degradation of glucocorticoid receptor (GR). Glucocorticoid prednisolone induced both activation and degradation of GR, as well as autophagy in synovial fibroblasts. BAY 11-7085-induced cell death was significantly decreased with glucocorticoid inhibitor mifepristone and with inhibitors of autophagy. Both BAY 11-7085-induced autophagy and GR activation were down regulated with PPAR-γ agonist, 15d-PGJ2 and MEK/ERK inhibitor UO126. Inhibition of autophagy markedly decreased endogenous and BAY 11-7085-induced ERK phosphorylation, suggesting a positive feed back loop between ERK activation and autophagy in synovial fibroblasts. Co-transfection of MEK1 with PPAR-γ1 in HEK293 cells caused known inhibitory phosphorylation of PPAR-γ1 (Serine 112) and enhanced GR degradation, in the absence or presence of prednisolone. Furthermore, GR was both phosphorylated on Serine 211 and down regulated in synovial fibroblasts during serum starvation induced autophagy. These results showed that GR activation and PPAR-γ inactivation mediated BAY 11-7085-induced autophagy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Fibroblasts/pathology , Nitriles/pharmacology , Osteoarthritis/pathology , Receptors, Glucocorticoid/metabolism , Sulfones/pharmacology , Synovial Membrane/pathology , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , HEK293 Cells , Humans , NF-kappa B/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , PPAR gamma/metabolism , Phosphorylation , Signal Transduction/drug effects , Synovial Membrane/drug effects , Synovial Membrane/metabolism
6.
PLoS One ; 10(4): e0122904, 2015.
Article in English | MEDLINE | ID: mdl-25849372

ABSTRACT

Osteoarthritis (OA) is associated with a local inflammatory process. Dyslipidemia is known to be an underlying cause for the development of OA. Therefore, lipid and inflammatory levels were quantified ex vivo in blood and synovial fluid of OA patients (n=29) and compared to those of rheumatoid arthritis (RA) patients (n=27) or healthy volunteers (HV) (n=35). The role of apolipoprotein A-I (ApoA1) was investigated in vitro on inflammatory parameters using human joint cells isolated from cartilage and synovial membrane obtained from OA patients after joint replacement. Cells were stimulated with ApoA1 in the presence or not of serum amyloid A (SAA) protein and/or lipoproteins (LDL and HDL) at physiological concentration observed in OA synovial fluid. In our ex vivo study, ApoA1, LDL-C and total cholesterol levels were strongly correlated to each other inside the OA joint cavity whereas same levels were not or weakly correlated to their corresponding serum levels. In OA synovial fluid, ApoA1 was not as strongly correlated to HDL as observed in OA serum or in RA synovial fluid, suggesting a dissociative level between ApoA1 and HDL in OA synovial fluid. In vitro, ApoA1 induced IL-6, MMP-1 and MMP-3 expression by primary chondrocytes and fibroblast-like synoviocytes through TLR4 receptor. HDL and LDL attenuated joint inflammatory response induced by ApoA1 and SAA in a ratio dependent manner. In conclusion, a dysregulated lipidic profile in the synovial fluid of OA patients was observed and was correlated with inflammatory parameters in the OA joint cavity. Pro-inflammatory properties of ApoA1 were confirmed in vitro.


Subject(s)
Apolipoprotein A-I/metabolism , Osteoarthritis/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Chondrocytes/immunology , Chondrocytes/metabolism , Female , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Middle Aged , Osteoarthritis/immunology , Primary Cell Culture , Synovial Fluid/metabolism , Toll-Like Receptor 4/metabolism , Transcriptional Activation , Young Adult
7.
Rheumatology (Oxford) ; 54(6): 1087-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25389358

ABSTRACT

OBJECTIVE: Glucocorticoids are powerful anti-inflammatory compounds that also induce the expression of leptin and leptin receptor (Ob-R) in synovial fibroblasts through TGF-ßsignalling and Smad1/5 phosphorylation. Compound A (CpdA), a selective glucocorticoid receptor agonist, reduces inflammation in murine arthritis models and does not induce diabetes or osteoporosis, thus offering an improved risk:benefit ratio in comparison with glucocorticoids. Due to the detrimental role of leptin in OA pathogenesis, we sought to determine whether CpdA also induced leptin and Ob-R protein expression as observed with prednisolone. METHODS: Human synovial fibroblasts and chondrocytes were isolated from the synovium and cartilage of OA patients after joint surgery. The cells were treated with prednisolone, TGF-ß1, TNF-α and/or CpdA. Levels of leptin, IL-6, IL-8, MMP-1 and MMP-3 were measured by ELISA and expression levels of Ob-R phospho-Smad1/5, phospho-Smad2, α-tubulin and glyceraldehyde 3-phosphate dehydrogenase were analysed by western blotting. RESULTS: CpdA, unlike prednisolone, did not induce leptin secretion or Ob-R protein expression in OA synovial fibroblasts. Moreover, CpdA decreased endogenous Ob-R expression and down-regulated prednisolone-induced leptin secretion and Ob-R expression. Mechanistically, CpdA, unlike prednisolone, did not induce Smad1/5 phosphorylation. CpdA, similarly to prednisolone, down-regulated endogenous and TNF-α-induced IL-6, IL-8, MMP-1 and MMP-3 protein secretion. The dissociative effect of CpdA was confirmed using chondrocytes with no induction of leptin secretion, but with a significant decrease in IL-6, IL-8, MMP-1 and MMP-3 protein secretion. CONCLUSION: CpdA, unlike prednisolone, did not induce leptin or Ob-R in human OA synovial fibroblasts, thereby demonstrating an improved risk:benefit ratio.


Subject(s)
Chondrocytes/metabolism , Fibroblasts/metabolism , Osteoarthritis/metabolism , Prednisolone/pharmacology , Receptors, Glucocorticoid/agonists , Synovial Membrane/metabolism , Aged , Aged, 80 and over , Blotting, Western , Chondrocytes/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Leptin/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Middle Aged , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Smad Proteins, Receptor-Regulated/drug effects , Smad Proteins, Receptor-Regulated/metabolism , Synovial Membrane/drug effects , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism , Tubulin/drug effects , Tubulin/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
World J Stem Cells ; 6(3): 367-70, 2014 Jul 26.
Article in English | MEDLINE | ID: mdl-25126385

ABSTRACT

In cell therapy protocols, many tissues were proposed as a source of mesenchymal stem cells (MSC) isolation. So far, bone marrow (BM) has been presented as the main source of MSC despite the invasive isolation procedure related to this source. During the last years, the umbilical cord (UC) matrix was cited in different studies as a reliable source from which long term ex vivo proliferating fibroblasts were isolated but with contradictory data about their immunophenotype, gene expression profile, and differentiation potential. Hence, an interesting question emerged: Are cells isolated from cord matrix (UC-MSC) different from other MSCs? In this review, we will summarize different studies that isolated and characterized UC-MSC. Considering BM-MSC as gold standard, we will discuss if UC-MSC fulfill different criteria that define MSC, and what remain to be done in this issue.

9.
PLoS One ; 8(6): e66769, 2013.
Article in English | MEDLINE | ID: mdl-23776697

ABSTRACT

OBJECTIVE: To determine if serum amyloid A (A-SAA) could be detected in human osteoarthritic (OA) joints and further clarify if high A-SAA level in joints result from a local production or from a diffusion process from abnormally elevated plasma concentration. Regulatory mechanism of A-SAA expression and its pro-inflammatory properties were also investigated. METHODS: A-SAA levels in serum and synovial fluid of OA (n = 29) and rheumatoid arthritis (RA) (n = 27) patients were measured and compared to matched-healthy volunteers (HV) (n = 35). In vitro cell cultures were performed on primary joint cells provided from osteoarthritis patients. Regulatory mechanisms were studied using Western-blotting, ELISA and lentiviral transfections. RESULTS: A-SAA was statistically increased in OA plasma patients compared to HV. Moreover, A-SAA level in OA plasma and synovial fluid increased with the Kellgren & Lauwrence grade. For all OA and RA patients, A-SAA plasma level was higher and highly correlated with its corresponding level in the synovial fluid, therefore supporting that A-SAA was mainly due to the passive diffusion process from blood into the joint cavity. However, A-SAA expression was also observed in vitro under corticosteroid treatment and/or under IL-1beta stimuli. A-SAA expression was down-regulated by PPAR-γ agonists (genistein and rosiglitazone) and up-regulated by TGF-ß1 through Alk1 (Smad1/5) pathway. RhSAA induced proinflammatory cytokines (IL-6, IL-8, GRO-α and MCP-1) and metalloproteinases (MMP-1, MMP-3 and MMP-13) expression in FLS and chondrocytes, which expression was downregulated by TAK242, a specific TLR4 inhibitor. CONCLUSION: Systemic or local A-SAA expression inside OA joint cavity may play a key role in inflammatory process seen in osteoarthritis, which could be counteracted by TLR4 inhibition.


Subject(s)
Acute-Phase Reaction/blood , Gene Expression Regulation/physiology , Joints/metabolism , Osteoarthritis/blood , Serum Amyloid A Protein/metabolism , Toll-Like Receptor 4/metabolism , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Genistein/pharmacology , Humans , Interleukin-1beta/pharmacology , Lentivirus , PPAR gamma/antagonists & inhibitors , Rosiglitazone , Sulfonamides/pharmacology , Synovial Fluid/metabolism , Thiazolidinediones/pharmacology
10.
Stem Cells Dev ; 21(11): 1948-55, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22087763

ABSTRACT

Leptin plays a central role in maintaining energy balance, with multiple other systemic effects. Despite leptin importance in peripheral regulation of mesenchymal stem cells (MSC) differentiation, little is known about its expression mechanism. Leptin is often described as adipokine, while it is expressed by other cell types. We have recently shown an in vitro leptin expression, enhanced by glucocorticoids in synovial fibroblasts (SVF). Here, we investigated leptin expression in MSC from bone marrow (BM-MSC) and umbilical cord matrix (UMSC). Results showed that BM-MSC, but not UMSC, expressed leptin that was strongly enhanced by glucocorticoids. Transforming growth factor ß1 (TGF-ß1) markedly inhibited the endogenous- and glucocorticoid-induced leptin expression in BM-MSC. Since TGF-ß1 was shown to signal via ALK-5-Smad2/3 and/or ALK-1-Smad1/5 pathways, we analyzed the expression of proteins from both pathways. In BM-MSC, TGF-ß1 increased phosphorylated Smad2 (p-Smad2) expression, while ALK-5 inhibitor (SB431542) induced leptin expression and significantly restored TGF-ß1-induced leptin inhibition. In addition, both prednisolone and SB431542 increased p-Smad1/5 expression. These results suggested the ALK-5-Smad2 pathway as an inhibitor of leptin expression, while ALK-1-Smad1/5 as an activator. Indeed, Smad1 expression silencing induced leptin expression inhibition. Furthermore, prednisolone enhanced the expression of TGF-ßRII while decreasing p-Smad2 in BM-MSC and SVF but not in UMSC. In vitro differentiation revealed differential osteogenic potential in SVF, BM-MSC, and UMSC that was correlated to their leptin expression potential. Our results suggest that ALK-1/ALK-5 balance regulates leptin expression in MSC. It also underlines UMSC as leptin nonproducer MSC for cell therapy protocols where leptin expression is not suitable.


Subject(s)
Activin Receptors, Type II/metabolism , Leptin/metabolism , Mesenchymal Stem Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Activin Receptors, Type II/genetics , Benzamides/pharmacology , Bone Marrow/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dioxoles/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Leptin/antagonists & inhibitors , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis , Phosphorylation , Prednisolone/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta1/pharmacology , Umbilical Cord/cytology , Umbilical Cord/metabolism
11.
Cell Biol Int ; 34(7): 693-701, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20187873

ABSTRACT

Many studies have drawn attention to the emerging role of MSC (mesenchymal stem cells) as a promising population supporting new clinical concepts in cellular therapy. However, the sources from which these cells can be isolated are still under discussion. Whereas BM (bone marrow) is presented as the main source of MSC, despite the invasive procedure related to this source, the possibility of isolating sufficient numbers of these cells from UCB (umbilical cord blood) remains controversial. Here, we present the results of experiments aimed at isolating MSC from UCB, BM and UCM (umbilical cord matrix) using different methods of isolation and various culture media that summarize the main procedures and criteria reported in the literature. Whereas isolation of MSC were successful from BM (10:10) and (UCM) (8:8), only one cord blood sample (1:15) gave rise to MSC using various culture media [DMEM (Dulbecco's modified Eagle's medium) +5% platelet lysate, DMEM+10% FBS (fetal bovine serum), DMEM+10% human UCB serum, MSCGM] and different isolation methods [plastic adherence of total MNC (mononuclear cells), CD3+/CD19+/CD14+/CD38+-depleted MNC and CD133+- or LNGFR+-enriched MNC]. MSC from UCM and BM were able to differentiate into adipocytes, osteocytes and hepatocytes. The expansion potential was highest for MSC from UCM. The two cell populations had CD90+/CD73+/CD105+ phenotype with the additional expression of SSEA4 and LNGFR for BM MSC. These results clearly exclude UCB from the list of MSC sources for clinical use and propose instead UCM as a rich, non-invasive and abundant source of MSC.


Subject(s)
Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Adipocytes/cytology , Adipocytes/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cattle , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Separation/methods , Cells, Cultured , Flow Cytometry , Hepatocytes/cytology , Hepatocytes/immunology , Humans , Immunophenotyping , Mesenchymal Stem Cells/immunology , Osteocytes/cytology , Osteocytes/immunology , Phenotype
12.
Lab Invest ; 89(7): 811-22, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19434061

ABSTRACT

It was shown recently that synovial fibroblast transformation into adipocytes reduced the expression of interleukin-6 (IL-6) and IL-8. However, the synovial fibroblast adipogenesis was inhibited in inflammatory conditions induced by the tumor necrosis factor-alpha (TNF-alpha). Furthermore, adipogenesis is often accompanied by leptin production, a proinflammatory adipokine in rheumatic diseases. In this study, we tested the phytohormone genistein for adipogenic and anti-inflammatory properties on human synovial fibroblasts. Results showed that genistein was able to transform synovial fibroblasts into adipocytes that expressed perilipin-A and produced adiponectin, but not leptin. Furthermore, genistein enhanced glucocorticoid-mediated synovial fibroblast adipogenesis and, in parallel, downregulated glucocorticoid-induced leptin and leptin receptor. Endogenous and TNF-alpha-induced expressions of IL-6, IL-8, p38, p65 and C/EBP-beta were also downregulated by genistein, showing its anti-inflammatory properties. Peroxisome proliferator- activated receptor-gamma (PPAR-gamma) agonist, rosiglitazone, had a synergic effect on genistein-induced adipogenesis, whereas the non-active tyrosine kinase inhibitor, daidzein, had a significantly inferior adipogenic activity than genistein. The Janus kinase-2 tyrosine kinase inhibitor, AG 490, mimicked the anti-leptin effect of genistein. These results showed that genistein-induced adipogenesis involves PPAR-gamma induction and tyrosine kinase inhibition. In conclusion, genistein, alone or coupled with glucocorticoids, have both adipogenic and anti-inflammatory effects on synovial fibroblasts.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/drug effects , Genistein/pharmacology , Leptin/biosynthesis , Synovial Membrane/cytology , Synovial Membrane/drug effects , Adipocytes/metabolism , Anti-Inflammatory Agents/pharmacology , Base Sequence , Cell Differentiation/drug effects , DNA Primers/genetics , Dexamethasone/pharmacology , Down-Regulation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , In Vitro Techniques , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Interleukin-8/genetics , PPAR gamma/biosynthesis , Phytoestrogens/pharmacology , Prostaglandin D2/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Rosiglitazone , Signal Transduction/drug effects , Synovial Membrane/metabolism , Thiazolidinediones/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
13.
J Biol Chem ; 281(32): 22597-604, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16766531

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) plays a central role in whole body metabolism by regulating adipocyte differentiation and energy storage. Recently, however, PPAR-gamma has also been demonstrated to affect proliferation, differentiation, and apoptosis of different cell types. As we have previously shown that BAY 11-7085-induced synovial fibroblast apoptosis is prevented by PPAR-gamma agonist 15d-PGJ2; the expression of PPAR-gamma in these cells was studied. Both PPAR-gamma1 and PPAR-gamma2 isoforms were cloned from synovial fibroblast RNA, but only PPAR-gamma1 was detected by Western blot, showing constitutive nuclear expression. Within minutes of BAY 11-7085 treatment, a PPAR-gamma1-specific band was shifted into a form of higher mobility, suggesting dephosphorylation, as confirmed by phosphatase treatment of cell extracts. Of interest, BAY 11-7085-induced PPAR-gamma1 dephosphorylation was followed by PARP and caspase-8 cleavage as well as by PPAR-gamma1 protein degradation. PPAR-gamma1 dephosphorylation was followed by the loss of PPAR-DNA binding activity ubiquitously present in synovial fibroblast nuclear extracts. Unlike the phosphorylated form, dephosphorylated PPAR-gamma1 was found in insoluble membrane cell fraction and was not ubiquitinated before degradation. PPAR-gamma1 dephosphorylation coincided with ERK1/2 phosphorylation that accompanies BAY 11-7085-induced synovial fibroblasts apoptosis. 15d-PGJ2, PGD2, and partially UO126, down-regulated ERK1/2 phosphorylation, protected cells from BAY 11-7085-induced apoptosis, and reversed both PPAR-gamma dephosphorylation and degradation. Furthermore, PPAR-gamma antagonist BADGE induced PPAR-gamma1 degradation, ERK1/2 phosphorylation, and synovial fibroblasts apoptosis. The results presented suggest an anti-apoptotic role for PPAR-gamma1 in synovial fibroblasts. Since apoptotic marker PARP is cleaved after PPAR-gamma1 dephosphorylation but before PPAR-gamma1 degradation, dephosphorylation event might be enough to mediate BAY 11-7085-induced apoptosis in synovial fibroblasts.


Subject(s)
Apoptosis , Fibroblasts/pathology , Nitriles/pharmacology , PPAR gamma/chemistry , Sulfones/pharmacology , Cell Line , Cell Nucleus/metabolism , Fibroblasts/metabolism , Gene Expression , Humans , Osteoarthritis/metabolism , PPAR gamma/metabolism , Phosphorylation , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Synovial Membrane/pathology , Transfection
14.
J Bacteriol ; 187(18): 6479-87, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16159781

ABSTRACT

Rhizobium (Sinorhizobium) sp. strain NGR234 contains three replicons, the smallest of which (pNGR234a) carries most symbiotic genes, including those required for nodulation and lipo-chito-oligosaccharide (Nod factor) biosynthesis. Activation of nod gene expression depends on plant-derived flavonoids, NodD transcriptional activators, and nod box promoter elements. Nod boxes NB6 and NB7 delimit six different types of genes, one of which (fixF) is essential for the formation of effective nodules on Vigna unguiculata. In vegetative culture, wild-type NGR234 produces a distinct, flavonoid-inducible lipopolysaccharide (LPS) that is not produced by the mutant (NGRomegafixF); this LPS is also found in nitrogen-fixing bacteroids isolated from V. unguiculata infected with NGR234. Electron microscopy showed that peribacteroid membrane formation is perturbed in nodule cells infected by the fixF mutant. LPSs were purified from free-living NGR234 cultured in the presence of apigenin. Structural analyses showed that the polysaccharide portions of these LPSs are specialized, rhamnose-containing O antigens attached to a modified core-lipid A carrier. The primary sequence of the O antigen is [-3)-alpha-L-Rhap-(1,3)-alpha-L-Rhap-(1,2)-alpha-L-Rhap-(1-]n, and the LPS core region lacks the acidic sugars commonly associated with the antigenic outer core of LPS from noninduced cells. This rhamnan O antigen, which is absent from noninduced cells, has the same primary sequence as the A-band O antigen of Pseudomonas aeruginosa, except that it is composed of L-rhamnose rather than the D-rhamnose characteristic of the latter. It is noteworthy that A-band LPS is selectively maintained on the P. aeruginosa cell surface during chronic cystic fibrosis lung infection, where it is associated with an increased duration of infection.


Subject(s)
Flavonoids/pharmacology , Gene Expression Regulation, Bacterial , Lipopolysaccharides/biosynthesis , Pseudomonas aeruginosa/drug effects , Rhizobium/drug effects , Gene Expression Regulation, Bacterial/drug effects , Lipopolysaccharides/chemistry , Nitrogen Fixation , O Antigens/chemistry , O Antigens/metabolism , Pseudomonas aeruginosa/metabolism , Rhizobium/genetics , Rhizobium/physiology
15.
Arthritis Rheum ; 52(1): 84-93, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15641051

ABSTRACT

OBJECTIVE: Interleukin-6 (IL-6) and soluble IL-6 receptor (sIL-6R) activation of gp130 represents an alternative pathway for osteoclast development in inflammatory conditions. The goal of the present study was to investigate changes in sIL-6R levels in response to the inflammatory cytokines IL-1beta and tumor necrosis factor alpha (TNFalpha) and to determine the role of TNFalpha-converting enzyme (TACE) in this process. METHODS: Levels of sIL-6R in the culture media of MG63 and SAOS-2 osteoblast-like cell lines after exposure to various agents were determined by immunoassay. TACE protein levels were measured by Western immunoblotting. Cells were transfected with small interfering RNA (siRNA) or with an expression plasmid for IL-6R and TACE to determine the potential involvement of TACE in IL-6R shedding. RESULTS: IL-1beta and TNFalpha increased the levels of sIL-6R in the culture media of MG63 osteoblast-like cells. This effect was not influenced by cycloheximide or 5,6-dichlorobenzimidazole riboside but was markedly inhibited by the calcium chelator EGTA and by the TACE and matrix metalloproteinase inhibitor hydroxamate (Ru36156). IL-1beta and TNFalpha had no influence on the alternatively spliced form of IL-6R RNA. Levels of sIL-6R were reduced when MG63 cells were transiently transfected with TACE siRNA. Transfection of SAOS-2 cells with expression plasmids for IL-6R and TACE produced a dose-dependent increase in sIL-6R levels. CONCLUSION: IL-1beta- and TNFalpha-mediated induction of IL-6R shedding in osteoblast-like cells is at least partly dependent on TACE activation.


Subject(s)
Interleukin-1/pharmacology , Metalloendopeptidases/metabolism , Osteoblasts/metabolism , Receptors, Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/pharmacology , ADAM Proteins , ADAM17 Protein , Cell Line, Tumor , Humans , Metalloendopeptidases/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/genetics , Recombinant Proteins/pharmacology , Solubility , Transfection
16.
J Biol Chem ; 279(21): 22399-403, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15004016

ABSTRACT

We have previously shown that nuclear factor-kappaB inhibition by adenovirus expressing mutated IkappaB-alpha or by proteasome inhibitor increases human articular chondrocytes sensibility to apoptosis. Moreover, the nuclear factor-kappaB inhibitor BAY11-7085, a potent anti-inflammatory drug in rat adjuvant arthritis, is itself a proapoptotic agent for chondrocytes. In this work, we show that BAY 11-7085 but not the proteasome inhibitor MG-132 induced a rapid and sustained phosphorylation of extracellular signal-regulated kinases (ERK1/2) in human articular chondrocytes. The level of ERK1/2 phosphorylation correlated with BAY 11-7085 concentration and chondrocyte apoptosis. 15-Deoxy-delta(12,14)-prostaglandin J2 (15d-PGJ2) and its precursor prostaglandin (PG) D2 but not PGE2 and PGF2alpha rescued chondrocytes from BAY 11-7085-induced apoptosis. 15d-PGJ2 markedly inhibited BAY 11-7085-induced phosphorylation of ERK1/2. BAY 11-7085 also induced ERK1/2 phosphorylation and apoptosis in human synovial fibroblasts, and these reactions were down-regulated by 15d-PGJ2. Further analysis in synovial fibroblasts showed that only molecules that suppressed BAY 11-7085-induced phosphorylation of ERK1/2 (i.e. 15d-PGJ2, PGD2, and to a lesser extent, MEK1/2 inhibitor UO126, but not prostaglandins E2 and F2alpha or peroxisome proliferator-activated receptor-gamma agonist ciglitazone) were able protect cells from apoptosis. These results suggested that the antiapoptotic effect of 15d-PGJ2 on chondrocytes and synovial fibroblasts might involve inhibition of ERK1/2 phosphorylation.


Subject(s)
Anti-Infective Agents/pharmacology , Apoptosis , Cartilage, Articular/cytology , Chondrocytes/metabolism , Fibroblasts/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostaglandin D2/physiology , Synovial Membrane/cytology , Annexin A5/pharmacology , Blotting, Western , Cartilage/cytology , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Coloring Agents/pharmacology , Cysteine Endopeptidases , Dinoprost/metabolism , Dinoprostone/metabolism , Down-Regulation , Humans , I-kappa B Proteins/metabolism , Immunologic Factors/physiology , Leupeptins/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Mutation , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Nitriles , Phosphorylation , Prostaglandin D2/analogs & derivatives , Proteasome Endopeptidase Complex , Receptors, Cytoplasmic and Nuclear/agonists , Sulfones , Thiazolidinediones/pharmacology , Transcription Factors/agonists
17.
Oncogene ; 23(8): 1631-5, 2004 Feb 26.
Article in English | MEDLINE | ID: mdl-14985703

ABSTRACT

The oncoprotein HER-2/neu is a prosurvival factor and its overexpression has been correlated with adverse prognosis in breast cancers. High levels of the cyclooxygenase-2 (COX-2), a proinflammatory and antiapoptotic enzyme, were detected in HER-2-positive tumors and this observation was linked to an HER-2-mediated induction of COX-2 gene transcription. Here, we report that COX-2 expression, and synthesis of its major enzymatic product, PGE2, leads in turn to an enhanced HER-2 expression. Moreover, COX-2 enzymatic inhibition dramatically reduced HER-2 protein levels, efficiently increased the cancer cells sensitility to chemotherapeutic treatment and acted in synergy with HER-2 inhibitor, trastuzumab. Therefore, we propose an original model where HER-2 and COX-2 transcriptionally regulate each other in a positive loop.


Subject(s)
Breast Neoplasms/metabolism , Dinoprostone/biosynthesis , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclooxygenase 2 , Female , Humans , Membrane Proteins , Models, Genetic , RNA, Messenger/analysis , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Oncogene ; 22(1): 90-7, 2003 Jan 09.
Article in English | MEDLINE | ID: mdl-12527911

ABSTRACT

The ubiquitous NF-kappaB transcription factor has been reported to inhibit apoptosis and to induce drug resistance in cancer cells. Drug resistance is the major reason for cancer therapy failure and neoplastic cells often develop multiple mechanisms of drug resistance during tumor progression. We observed that NF-kappaB or P-glycoprotein inhibition in the HCT15 colon cancer cells led to increased apoptotic cell death in response to daunomycin treatment. Interestingly, NF-kappaB inhibition through transfection of a plasmid coding for a mutated IkappaB-alpha inhibitor increased daunomycin cell uptake. Indeed, the inhibition of NF-kappaB reduced mdr1 mRNA and P-glycoprotein expression in HCT15 cells. We identified a consensus NF-kappaB binding site in the first intron of the human mdr1 gene and demonstrated that NF-kappaB complexes could bind with this intronic site. Moreover, NF-kappaB transactivates an mdr1 promoter luciferase construct. Our data thus demonstrate a role for NF-kappaB in the regulation of the mdr1 gene expression in cancer cells and in drug resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Colonic Neoplasms/genetics , Drug Resistance, Neoplasm/physiology , NF-kappa B/physiology , Antineoplastic Agents/pharmacology , Base Sequence , Colonic Neoplasms/pathology , DNA Primers , Daunorubicin/antagonists & inhibitors , Daunorubicin/pharmacokinetics , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic/physiology , Humans , Introns , Plasmids , Promoter Regions, Genetic , Transcriptional Activation/physiology , Tumor Cells, Cultured
19.
Lab Invest ; 82(12): 1661-72, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12480916

ABSTRACT

TNF-alpha plays a key role in rheumatoid arthritis, but its effect on chondrocyte survival is still conflicting. In the present study, we tested how TNF-alpha influences chondrocyte survival in response to nitric oxide (NO)-related apoptotic signals, which are abundant during rheumatoid arthritis. Human primary articular chondrocytes or cartilage explants were pretreated with TNF-alpha for 24 hours and then treated with the proapoptotic NO donor sodium-nitro-prusside (SNP) for an additional 24 hours. TNF-alpha pretreatment markedly protected chondrocytes from SNP-induced cell death. Preincubation of chondrocytes with TNF-alpha inhibited both SNP-induced high-molecular weight DNA fragmentation and annexin V-FITC binding. Of interest, TNF-alpha induced persistent nuclear factor-kappaB (NF-kappaB)-DNA binding activity even in the presence of SNP, mirroring apoptosis protection effects. Both the TNF-alpha antiapoptotic effect and NF-kappaB-DNA binding activity were significantly inhibited by NF-kappaB inhibitors, Bay 11-7085, MG-132, and adenovirus-expressing mutated IkappaB-alpha. Phosphatidylinositol-3 kinase inhibitor LY 294002 also markedly inhibited the antiapoptotic effect of TNF-alpha. In primary chondrocytes, TNF-alpha induced expression of the antiapoptotic protein Cox-2, which persisted in the presence of SNP, and a specific Cox-2 inhibitor significantly blocked the TNF-alpha protective effect. We therefore conclude that TNF-alpha-mediated protection of chondrocytes from NO-induced apoptosis acts through NF-kappaB and requires Cox-2 activity.


Subject(s)
Apoptosis/drug effects , Chondrocytes/drug effects , NF-kappa B/metabolism , Nitric Oxide/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Adenoviruses, Human/genetics , Adult , Aged , Anti-Infective Agents/pharmacology , Cartilage, Articular , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/pathology , Chromones/pharmacology , Cyclooxygenase 2 , Enzyme Inhibitors/pharmacology , Flow Cytometry , Genetic Vectors , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Leupeptins/pharmacology , Membrane Proteins , Middle Aged , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , Nitric Oxide Donors/pharmacology , Nitriles , Nitroprusside/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Sulfones
20.
J Cell Physiol ; 192(1): 93-101, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12115740

ABSTRACT

Interleukin (IL)-4, which exhibits potent anti-inflammatory activities, is of potential therapeutic value in destructive arthropathies. To further define the response of human joint cells to IL-4, we analyzed the ability of this cytokine to modulate the effects of IL-1beta and growth factors. Freshly isolated chondrocytes, dedifferentiated chondrocytes, and synoviocytes were treated with IL-4 before determination of nitric oxide (NO) and collagenase production in response to IL-1beta, or before proliferation assays in presence of IL-1beta, platelet-derived growth factor (PDGF), or transforming growth factor (TGF)-beta. IL-4 downregulated IL-1beta induced NO production in dedifferentiated chondrocytes and inhibited IL-1beta induced collagenase release, as well as IL-1beta and growth factor induced proliferation in dedifferentiated chondrocytes and synoviocytes. In contrast, IL-4 had no effect in freshly isolated primary chondrocytes and in cartilage explants. The lack of response to IL-4 in primary chondrocytes was associated with impaired signal transduction, as indicated by markedly decreased IL-4 dependent tyrosine phosphorylation of signal transducer and activator of transcription (STAT)-6. It also correlated with differences in the expression pattern of IL-4 receptor (IL-4R) subunits during chondrocyte dedifferentiation. Indeed, whereas the IL-4Ralpha and IL-13Ralpha' subunits were expressed in all cell types, expression of the common receptor gamma chain was restricted to freshly isolated chondrocytes. In conclusion, IL-4 downregulated IL-1beta-induced catabolic events and cell proliferation in dedifferentiated chondrocytes and synoviocytes, but had no effects in freshly isolated chondrocytes. The difference in IL-4 responsiveness between primary and dedifferentiated chondrocytes correlated with changes in proximal signaling events and in the expression pattern of IL-4R subunits during cell dedifferentiation.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrocytes/drug effects , Interleukin-4/pharmacology , Receptors, Interleukin-4/metabolism , Synovial Membrane/cytology , Synovial Membrane/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Chondrocytes/metabolism , Collagenases/metabolism , Humans , Interleukin-1/pharmacology , Nitric Oxide/biosynthesis , Phosphorylation/drug effects , Protein Isoforms/metabolism , STAT6 Transcription Factor , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...