Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Exp Cell Res ; 315(5): 795-808, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19073173

ABSTRACT

The formation of new blood vessels, or angiogenesis, is a necessary process during development but also for tumour growth and other pathologies. It is promoted by different growth factors that stimulate endothelial cells to proliferate, migrate, and generate new tubular structures. Syndecans, transmembrane heparan sulphate proteoglycans, bind such growth factors through their glycosaminoglycan chains and could transduce the signal to the cytoskeleton, thus regulating cell behaviour. We demonstrated that syndecan-2, the major syndecan expressed by human microvascular endothelial cells, is regulated by growth factors and extracellular matrix proteins, in both bidimensional and tridimensional culture conditions. The role of syndecan-2 in "in vitro" tumour angiogenesis was also examined by inhibiting its core protein expression with antisense phosphorothioate oligonucleotides. Downregulation of syndecan-2 reduces spreading and adhesion of endothelial cells, enhances their migration, but also impairs the formation of capillary-like structures. These results suggest that syndecan-2 has an important function in some of the necessary steps that make up the angiogenic process. We therefore propose a pivotal role of this heparan sulphate proteoglycan in the formation of new blood vessels.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Syndecan-2/genetics , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Culture Techniques , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/physiology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Microcirculation/drug effects , Microcirculation/genetics , Neovascularization, Physiologic/drug effects , Oligonucleotides, Antisense/pharmacology , Syndecan-2/antagonists & inhibitors , Syndecan-2/metabolism , Syndecan-2/physiology
2.
Int J Biochem Cell Biol ; 39(10): 1915-29, 2007.
Article in English | MEDLINE | ID: mdl-17616478

ABSTRACT

Platelet-derived growth factor (PDGF) has been implicated in vascular smooth muscle cell proliferation and migration, a key process in vascular disease. PDGF is a family of dimeric isoforms of structurally related A-, B-, C- and D-chains that bind to PDGF receptors. PDGF A- and B-chains occur with and without basic C-terminal amino acid extensions as long (A(L) and B(L)) and short (A(S) and B(S)) isoforms. This basic sequence has been implicated as a cell retention signal through binding to glycosaminoglycans, especially to heparan sulfate. The aim of this study was to evaluate the biological relevance of PDGF interaction with glycosaminoglycans on the PDGF function in human arterial smooth muscle cells (hASMC). Here, we show that long PDGF isoforms showed greater affinity for hASMC cell surface and that they also presented more colocalization with heparan and chondroitin sulfates present on hASMC cell membrane than did short isoforms. Furthermore, all PDGF isoforms colocalized more with heparan sulfate than with chondroitin sulfate and there was little colocalization between heparan and chondroitin sulfate. PDGF-stimulated hASMC activation of DNA synthesis and directed migration (chemotaxis) was also examined. The isoform PDGF-BB(S) induced maximal proliferation and migration of hASMC. Collagen-I coating significantly increased hASMC motility towards PDGF isoforms, and particularly toward PDGF-BB(S). These results strongly support the notion that cell surface glycosaminoglycans are not essential for receptor-mediated activity of PDGF and may contribute basically to the retention and accumulation of long PDGF isoforms.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Glycosaminoglycans/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Arteries/cytology , Arteries/drug effects , Arteries/metabolism , Cells, Cultured , DNA/biosynthesis , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Protein Binding , Protein Isoforms/metabolism , Tissue Distribution
3.
Biochem J ; 394(Pt 2): 449-57, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16232121

ABSTRACT

Claudin-1 is an integral membrane protein component of tight junctions. The Snail family of transcription factors are repressors that play a central role in the epithelial-mesenchymal transition, a process that occurs during cancer progression. Snail and Slug members are direct repressors of E-cadherin and act by binding to the specific E-boxes of its proximal promoter. In the present study, we demonstrate that overexpression of Slug or Snail causes a decrease in transepithelial electrical resistance. Overexpression of Slug and Snail in MDCK (Madin-Darby canine kidney) cells down-regulated Claudin-1 at protein and mRNA levels. In addition, Snail and Slug are able to effectively repress human Claudin-1-driven reporter gene constructs containing the wild-type promoter sequence, but not those with mutations in two proximal E-box elements. We also demonstrate by band-shift assay that Snail and Slug bind to the E-box motifs present in the human Claudin-1 promoter. Moreover, an inverse correlation in the levels of Claudin-1 and Slug transcripts were observed in breast cancer cell lines. E-box elements in the Claudin-1 promoter were found to play a critical negative regulatory role in breast cancer cell lines that expressed low levels of Claudin-1 transcript. Significantly, in invasive human breast tumours, high levels of Snail and Slug correlated with low levels of Claudin-1 expression. Taken together, these results support the hypothesis that Claudin-1 is a direct downstream target gene of Snail family factors in epithelial cells.


Subject(s)
Down-Regulation , Epithelial Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line , Claudin-1 , Dogs , Fibroblasts/metabolism , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Snail Family Transcription Factors
4.
Mol Biol Cell ; 14(7): 2844-60, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12857869

ABSTRACT

Beta-catenin is a multifunctional protein involved in cell-cell adhesion and Wnt signal transduction. Beta-catenin signaling has been proposed to act as inducer of cell proliferation in different tumors. However, in some developmental contexts and cell systems beta-catenin also acts as a positive modulator of apoptosis. To get additional insights into the role of beta-catenin in the regulation of the cell cycle and apoptosis, we have analyzed the levels and subcellular localization of endogenous beta-catenin and its relation with adenomatous polyposis coli (APC) during the cell cycle in S-phase-synchronized epithelial cells. Beta-catenin levels increase in S phase, reaching maximum accumulation at late G2/M and then abruptly decreasing as the cells enter into a new G1 phase. In parallel, an increased cytoplasmic and nuclear localization of beta-catenin and APC is observed during S and G2 phases. In addition, strong colocalization of APC with centrosomes, but not beta-catenin, is detected in M phase. Interestingly, overexpression of a stable form of beta-catenin, or inhibition of endogenous beta-catenin degradation, in epidermal keratinocyte cells induces a G2 cell cycle arrest and leads to apoptosis. These results support a role for beta-catenin in the control of cell cycle and apoptosis at G2/M in normal and transformed epidermal keratinocytes.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Apoptosis/physiology , Cytoskeletal Proteins/metabolism , G2 Phase/physiology , Mitosis/physiology , Trans-Activators/metabolism , Animals , Cell Adhesion/physiology , Cell Nucleus/metabolism , Cells, Cultured , Centrosome/metabolism , Dogs , Flow Cytometry , Humans , Mice , Microscopy, Confocal , beta Catenin
5.
Cardiovasc Res ; 65(2): 328-33, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15639471

ABSTRACT

OBJECTIVE: The low number of postnatal endothelial progenitor cells (EPC) in the circulation limits their therapeutic application in cardiovascular medicine. Processed lipoaspirate (PLA) cells differentiate into osteoid, adipose, muscle, and cartilaginous cells. This study examines the potential of PLA cells as a source of EPCs. METHODS: PLA cells obtained from human lipoaspirates were cultured for 1 week in serum-depleted medium to form three-dimensional cell clusters (3DCC). The phenotype of 3DCC-derived cells was assessed by immunofluorescence staining and FACS analysis. RESULTS: Flow cytometry showed that 45+/-5% of cells derived from the 3DCC expressed Flk-1, a marker of early EPC, whilst only 4+/-0.5% of freshly isolated PLA were Flk-1+. The proportion of Flk-1+ cells increased to 98+/-2% during culture in hematopoietic stem cell medium. When cultured in an endothelial cell (EC)-specific medium, Flk-1+ cells also expressed Ve-cadherin, von Willebrand's factor (vW), and a lectin receptor, and took up low-density lipoprotein. Incorporation into an endothelial cell tubular network confirmed their functional activity. CONCLUSION: This report describes the first isolation and culture of Flk-1+ cells from human adipose tissue. The feasibility of the extraction and culture of these cells in increased numbers suggests that such autologous cells will be useful for applications ranging from basic research to cell-based therapies.


Subject(s)
Adipocytes/metabolism , Endothelial Cells/cytology , Stem Cells/cytology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cell Separation/methods , Flow Cytometry , Fluorescent Antibody Technique , Humans , Lipectomy
6.
Biochim Biophys Acta ; 1686(1-2): 37-49, 2004 Nov 08.
Article in English | MEDLINE | ID: mdl-15522821

ABSTRACT

Lipoprotein lipase (LPL) is produced by cells in the artery wall and can mediate binding of lipoproteins to cell surface heparan sulfate proteoglycans (HSPG), resulting in endocytosis (the bridging function). Active, dimeric LPL may dissociate to inactive monomers, the main form found in plasma. We have studied binding/internalization of human low density lipoprotein (LDL), mediated by bovine LPL, using THP-1 monocytes and macrophages. Uptake of (125)I-LDL was similar in monocytes and macrophages and was not affected by the LDL-receptor family antagonist receptor-associated protein (RAP) or by the phagocytosis inhibitor cytochalasin D. In contrast, uptake depended on HSPG and on membrane cholesterol. Incubation in the presence of dexamethasone increased the endogenous production of LPL by the cells and also increased LPL-mediated binding of LDL to the cell surfaces. Monomeric LPL was bound to the cells mostly in a heparin-resistant fashion. We conclude that the uptake of LDL mediated by LPL dimers is receptor-independent and involves cholesterol-enriched membrane areas (lipid rafts). Dimeric and monomeric LPL differ in their ability to mediate binding/uptake of LDL, probably due to different mechanisms for binding/internalization.


Subject(s)
Lipoprotein Lipase/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Membrane Microdomains/metabolism , Monocytes/metabolism , Animals , Cattle , Cells, Cultured , Cholesterol/physiology , Cytochalasin D/pharmacology , Dimerization , Heparin Lyase/metabolism , Heparin Lyase/pharmacology , Humans , LDL-Receptor Related Protein-Associated Protein/metabolism , LDL-Receptor Related Protein-Associated Protein/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/chemistry , Lipoproteins, LDL/pharmacology , Macrophages/ultrastructure , Monocytes/ultrastructure , Protein Binding
7.
Int J Biochem Cell Biol ; 36(11): 2226-41, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15313468

ABSTRACT

The platelet-derived growth factor (PDGF) family comprises important mitogens for mesenchymal cells. The active dimeric form of PDGF consists of four structurally related A, B, C, and D chains. All PDGF-variants bind to PDGF-receptors. The A and B chains occur with and without basic C-terminal amino acid extensions as long (A(L) and B(L)) and short (A(S) and B(S)) isoforms. PDGF-A and -B form homo- or heterodimers. The biological relevance of short and long isoforms is unknown, although it may relate to different affinities for glycosaminoglycans of the cell glycocalix and intercellular matrix. Commercially available anti-PDGF-A and anti-PDGF-B antibodies cannot discriminate between the short and the long isoforms. Thus, to investigate the function of the long and short isoforms, we raised antibodies specific for the long A and B chain isoforms. The antibodies were affinity-purified and their properties analysed by surface plasmon resonance. Inhibition studies with different PDGF homodimers and dot-blot studies proved their high specificity for the respective isoforms. Both antibodies recognised the target PDGF homodimers complexed to the glycocalix of human arterial smooth muscle cells and human monocyte-derived macrophages. By using these specific antibodies, we were able to confirm at the protein level the synthesis of PDGF-A and -B during differentiation of human monocyte-derived macrophages and to demonstrate the presence of the PDGF-A(L) and PDGF-B(L) isoforms in human arterial tissue.


Subject(s)
Carotid Arteries/metabolism , Cell Differentiation/physiology , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Antibodies/immunology , Cells, Cultured , Glycocalyx/metabolism , Humans , Monocytes/cytology , Myocytes, Smooth Muscle/cytology , Platelet-Derived Growth Factor/immunology , Protein Binding , Protein Isoforms/immunology , Protein Isoforms/metabolism , Rabbits , Receptors, Platelet-Derived Growth Factor/metabolism , Surface Plasmon Resonance
8.
FEBS Lett ; 547(1-3): 212-6, 2003 Jul 17.
Article in English | MEDLINE | ID: mdl-12860416

ABSTRACT

ERM (Ezrin/Radixin/Moesin) proteins are crosslinkers between plasma membrane proteins and the actin cytoskeleton, thereby involved in the formation of cell adhesion sites. Earlier work showed that Ezrin links syndecan-2 to the actin cytoskeleton. Here we provide evidence that the Ezrin N-terminal domain binds to the syndecan-2 cytoplasmic domain with an estimated K(D) of 0.71 microM and without the requirement of other proteins. We also studied the regions in the syndecan-2 cytoplasmic domain implicated in the binding to Ezrin. By truncating the syndecan-2 cytoplasmic domain and by oligopeptide competition assays we show that the Ezrin-binding sequence is not located in the positively charged juxtamembrane region (RMRKK), but in the neighboring sequence DEGSYD. We therefore conclude that the consensus sequence for Ezrin binding is unique among membrane proteins, suggesting a distinct regulation.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Proteoglycans/chemistry , Proteoglycans/metabolism , Amino Acid Sequence , Binding Sites , Binding, Competitive , Cloning, Molecular , Cytoskeletal Proteins , Humans , Kinetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Syndecan-2
9.
FEBS Lett ; 569(1-3): 105-11, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15225617

ABSTRACT

Transforming growth factor-beta (TGFbeta1) is a proangiogenic factor both, in vitro and in vivo, that is mainly involved in the later phases of angiogenesis. In an attempt to identify genes that participate in this effect, we found that TGFbeta1 down-regulates expression of adenylate cyclase VI. In addition, cAMP analogs (8-Bromo-cAMP) and forskolin (an adenylate cyclase activator) also reduced TGFbeta1-induced in vitro angiogenesis in mouse endothelial cell lines and in primary cultures of human umbilical vein endothelial cells on collagen gels. Induction of Ets-1 and plasminogen activator inhibitor-1 (PAI-1) by TGFbeta1 was blocked by these cAMP agonists and activators, in the absence of effects on endothelial cell viability. Moreover, the signal transduction pathways stimulated by TGFbeta1 were unaffected. Thus, Smad2 was normally phosphorylated and translocated to the nucleus in the presence of forskolin. In contrast, transfection studies using the PAI-1-promoter indicated that these cAMP analogues inhibit transcriptional stimulation by TGFbeta1. Electrophoretic mobility shift assay showed that Smad2/3 were bound normally to a TGFbeta1-response region in the presence of the cAMP analogs. In all, these data suggest that the cAMP pathway inhibits the transcriptional activity of Smads, that could be responsible for the block of the TGFbeta1-induced in vitro angiogenesis caused by this second messenger.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cyclic AMP/pharmacology , Endothelium, Vascular/physiology , Neovascularization, Physiologic/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Capillaries/drug effects , Capillaries/physiology , Cell Line , Endothelium, Vascular/drug effects , Humans , Mice , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1 , Umbilical Veins/drug effects , Umbilical Veins/physiology
10.
FEBS Lett ; 559(1-3): 77-83, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14960311

ABSTRACT

HERC1 is a very large protein involved in membrane traffic through both its ability to bind clathrin and its guanine nucleotide exchange factor (GEF) activity over ARF and Rab family GTPases. Herein, we show that HERC1 is recruited onto actin-rich surface protrusions in ARF6-transfected HeLa cells upon aluminum fluoride (AlF(4)(-)) treatment. Moreover, the fact that HERC1 overexpression does not stimulate protrusion formation in the absence of AlF(4)(-), in conditions where ARNO does, indicates that HERC1 is not acting as an ARF6-GEF in this system, but that instead its recruitment takes place downstream of ARF6 activation. Finally, we suggest a phosphoinositide-binding mechanism whereby HERC1 may translocate to these protrusions.


Subject(s)
Actins/physiology , Cell Surface Extensions , Guanine Nucleotide Exchange Factors/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/physiology , Aluminum Compounds/pharmacology , Cell Surface Extensions/drug effects , Cytoskeleton , Fluorides/pharmacology , GTPase-Activating Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , HeLa Cells , Humans , Phosphatidylinositols/metabolism , Protein Binding , Protein Transport , Transfection , Ubiquitin-Protein Ligases
11.
Oligonucleotides ; 14(2): 100-13, 2004.
Article in English | MEDLINE | ID: mdl-15294074

ABSTRACT

The antiapoptotic protein survivin is an attractive target in cancer therapy because it is expressed differently in tumors and normal tissues and it is potentially required for cancer cells to remain viable. Given that survivin is also overexpressed in endothelial cells (ECs) of newly formed blood vessels found in tumors, its RNA targeting might compromise EC viability and interfere with tumor angiogenesis. We used two antisense strategies against survivin expression, antisense oligonucleotides (aODN) and small interfering RNA (siRNA), to study in ECs the contribution of survivin in various steps leading to tumor angiogenesis. A 21-mer phosphorothioate aODN and two siRNA oligonucleotides against survivin mRNA were designed to downregulate survivin expression. Survivin targeting caused (1) a strong growth-inhibitory effect, (2) a 4-fold increase in apoptosis, (3) an accumulation of cells in the S phase and a decrease in G2/M phase, (4) a dose-dependent inhibition of EC migration on Vitronectin, and (5) a decrease in capillary formation. Control oligonucleotides, an unrelated oligonucleotide, and one with four mismatches, had no significant effect. All these results show that survivin is a suitable target in cancer therapy because its inhibition in EC causes both a proapoptotic effect and an interruption of tumor angiogenesis. The two strategies used, classic aODN and siRNA technology, were very effective. Moreover, the latter can be used in the low nanomolar range, thus increasing the sensitivity of the treatment.


Subject(s)
Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Neovascularization, Pathologic/drug therapy , Oligonucleotides, Antisense/therapeutic use , RNA, Small Interfering/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Apoptosis/drug effects , Cell Culture Techniques , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/analysis , Neoplasm Proteins/biosynthesis , Oligonucleotides, Antisense/pharmacology , RNA, Small Interfering/pharmacology , Survivin
12.
BMC Physiol ; 4(1): 13, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15544705

ABSTRACT

BACKGROUND: Lipoprotein lipase (LPL) is anchored at the vascular endothelium through interaction with heparan sulfate. It is not known how this enzyme is turned over but it has been suggested that it is slowly released into blood and then taken up and degraded in the liver. Heparin releases the enzyme into the circulating blood. Several lines of evidence indicate that this leads to accelerated flux of LPL to the liver and a temporary depletion of the enzyme in peripheral tissues. RESULTS: Rat livers were found to contain substantial amounts of LPL, most of which was catalytically inactive. After injection of heparin, LPL mass in liver increased for at least an hour. LPL activity also increased, but not in proportion to mass, indicating that the lipase soon lost its activity after being bound/taken up in the liver. To further study the uptake, bovine LPL was labeled with 125I and injected. Already two min after injection about 33 % of the injected lipase was in the liver where it initially located along sinusoids. With time the immunostaining shifted to the hepatocytes, became granular and then faded, indicating internalization and degradation. When heparin was injected before the lipase, the initial immunostaining along sinusoids was weaker, whereas staining over Kupffer cells was enhanced. When the lipase was converted to inactive before injection, the fraction taken up in the liver increased and the lipase located mainly to the Kupffer cells. CONCLUSIONS: This study shows that there are heparin-insensitive binding sites for LPL on both hepatocytes and Kupffer cells. The latter may be the same sites as those that mediate uptake of inactive LPL. The results support the hypothesis that turnover of endothelial LPL occurs in part by transport to and degradation in the liver, and that this transport is accelerated after injection of heparin.


Subject(s)
Heparin/pharmacology , Lipoprotein Lipase/metabolism , Liver/enzymology , Animals , Cattle , Heparin/administration & dosage , Injections , Lipoprotein Lipase/analysis , Lipoprotein Lipase/pharmacokinetics , Male , Protein Transport , Rats , Rats, Sprague-Dawley
13.
J Cell Sci ; 119(Pt 21): 4541-53, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17046996

ABSTRACT

Podoplanin is a small membrane mucin expressed in tumors associated with malignant progression. It is enriched at cell-surface protrusions where it colocalizes with members of the ERM (ezrin, radixin, moesin) protein family. Here, we found that human podoplanin directly interacts with ezrin (and moesin) in vitro and in vivo through a cluster of basic amino acids within its cytoplasmic tail, mainly through a juxtamembrane dipeptide RK. Podoplanin induced an epithelial-mesenchymal transition in MDCK cells linked to the activation of RhoA and increased cell migration and invasiveness. Fluorescence time-lapse video observations in migrating cells indicate that podoplanin might be involved in ruffling activity as well as in retractive processes. By using mutant podoplanin constructs fused to green fluorescent protein we show that association of the cytoplasmic tail with ERM proteins is required for upregulation of RhoA activity and epithelial-mesenchymal transition. Furthermore, expression of either a dominant-negative truncated variant of ezrin or a dominant-negative mutant form of RhoA blocked podoplanin-induced RhoA activation and epithelial-mesenchymal transition. These results provide a mechanistic basis to understand the role of podoplanin in cell migration or invasiveness.


Subject(s)
Cytoskeletal Proteins/metabolism , Epithelial Cells/pathology , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mesoderm/pathology , Microfilament Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Blotting, Western , Cell Line , Cell Movement , Dogs , Epithelial Cells/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Kidney/metabolism , Membrane Glycoproteins/genetics , Mesoderm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Wound Healing , rhoA GTP-Binding Protein/genetics
14.
Apoptosis ; 11(11): 2065-75, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17041758

ABSTRACT

To examine the function of syndecan-2, one of the most abundant heparan sulfate proteoglycans in fibroblasts, we obtained stably transfected Swiss 3T3 clones. We examined the effects of stable syndecan-2 overexpression on programmed cell death, finding that syndecan-2 transfected cells were more sensitive to apoptosis induced by serum-withdrawal than control cells. In addition, overexpression of syndecan-2 correlates with increased membrane levels of the Fas/CD95 receptor, suggesting that the increased serum-withdrawal apoptosis observed in Swiss 3T3 cells might be Fas receptor-dependent. Differences in Fas membrane levels between both control and syndecan-2 transfected cells result from a redistribution of the Fas receptor. Our data clearly demonstrate that increased Fas levels are primarily related to lipid rafts and that this increase is a key factor in Fas/CD95-mediated apoptosis. Moreover, disruption of lipid rafts with methyl-beta-cyclodextrin or filipin significantly reduced apoptosis in response to serum withdrawal. The differences in Fas/CD95 membrane distribution could explain why syndecan-2 transfected cells have a higher susceptibility to serum-withdrawal-induced apoptosis.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Membrane Microdomains/metabolism , Serum/metabolism , Syndecan-2/physiology , 3T3 Cells , Animals , Apoptosis/drug effects , Cattle , Cell Culture Techniques , DNA, Complementary , Dogs , Filipin/pharmacology , Humans , Mice , Syndecan-2/genetics , Transfection , beta-Cyclodextrins/pharmacology
15.
J Pept Sci ; 11(12): 789-804, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15942930

ABSTRACT

The use of high-throughput methods in drug discovery allows the generation and testing of a large number of compounds, but at the price of providing redundant information. Evolutionary combinatorial chemistry combines the selection and synthesis of biologically active compounds with artificial intelligence optimization methods, such as genetic algorithms (GA). Drug candidates for the treatment of central nervous system (CNS) disorders must overcome the blood-brain barrier (BBB). This paper reports a new genetic algorithm that searches for the optimal physicochemical properties for peptide transport across the blood-brain barrier. A first generation of peptides has been generated and synthesized. Due to the high content of N-methyl amino acids present in most of these peptides, their syntheses were especially challenging due to over-incorporations, deletions and DKP formations. Distinct fragmentation patterns during peptide cleavage have been identified. The first generation of peptides has been studied by evaluation techniques such as immobilized artificial membrane chromatography (IAMC), a cell-based assay, log Poctanol/water calculations, etc. Finally, a second generation has been proposed.


Subject(s)
Blood-Brain Barrier , Combinatorial Chemistry Techniques , Peptides/chemical synthesis , Algorithms , Animals , Artificial Intelligence , Biological Transport , Blood-Brain Barrier/metabolism , Cattle , Cells, Cultured , Peptide Library , Peptides/metabolism , Peptides/therapeutic use , Pharmaceutical Preparations/chemical synthesis , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Rats , Structure-Activity Relationship
16.
Am J Transplant ; 5(9): 2154-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16095494

ABSTRACT

Circulating endothelial progenitor cells (EPCs) promote vascular repair and maintain integrity of the endothelial monolayer. Reduced EPCs number has been associated with endothelial dysfunction in various cardiovascular diseases. Cardiovascular disease risk is higher in renal transplant patients (RT) than the general population. We studied EPCs number and proliferation in RT, and examined the association with other cardiovascular risk factors such as reduced glomerular filtration rate (GFR) and LDL cholesterol. EPCs concentration was determined in 94 RT and 39 control subjects (C) by flow cytometry. EPCs proliferation was also studied after 7 days in culture. EPCs concentration was significantly reduced in RT versus C (median 33.5 [5-177] vs. 53 [9-257] EPCs/10(5) PMN cells, p=0.006). EPCs proliferation was also reduced in RT versus C (mean+/-SD; 372.7+/-229.3 vs. 539.8+/-291.3 EPCs x field, p=0.003). In multiple regression analysis, GFR, HDL, LDL and body weight were independent predictors of EPCs concentration in RT (r2=0.25, p<0.001). EPCs number is reduced in RT, particularly in patients with reduced GFR. Moreover, EPCs from RT studied in vitro, showed reduced proliferation, which is a sign of functional impairment. These alterations may be involved in increased cardiovascular risk of RT.


Subject(s)
Endothelial Cells/cytology , Kidney Transplantation , Stem Cells/cytology , AC133 Antigen , Adult , Antigens, CD/biosynthesis , Antigens, CD34/biosynthesis , Body Weight , Cardiovascular Diseases/pathology , Cardiovascular System/pathology , Cell Proliferation , Female , Flow Cytometry , Glomerular Filtration Rate , Glycoproteins/biosynthesis , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Kidney Failure, Chronic , Leukocyte Common Antigens/biosynthesis , Linear Models , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Male , Middle Aged , Neutrophils/metabolism , Peptides , Regression Analysis , Sex Factors , Time Factors
17.
Cell Biol Int ; 27(12): 1005-10, 2003.
Article in English | MEDLINE | ID: mdl-14642532

ABSTRACT

Syndecans (heparan sulfate proteoglycans) participate in cell-cell and cell-matrix adhesion and are co- and low-affinity receptors for growth factors and enzymes, respectively. We examined the influence of stable syndecan-2 expression in Swiss 3T3 cells on cell-adhesion and proliferation. Higher syndecan-2 expression changed cell morphology and increased spreading and adhesion in these cells and proliferation induced by FCS and FGF-2. This emphasizes the role of syndecan-2 in the integration of signals from soluble and insoluble factors.


Subject(s)
Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/physiology , Proteoglycans/biosynthesis , Proteoglycans/physiology , Animals , Blotting, Western , Cell Adhesion , Cell Division , DNA, Complementary/metabolism , Densitometry , Fibroblast Growth Factor 2/metabolism , Mice , Swiss 3T3 Cells , Syndecan-2 , Time Factors , Transfection
18.
Cell Biol Int ; 28(11): 829-33, 2004.
Article in English | MEDLINE | ID: mdl-15563406

ABSTRACT

Syndecans, transmembrane heparan sulfate proteoglycans (HSPG), mediate cell-cell and cell-matrix adhesion thereby controlling cell movement and shape. Syndecan cytoplasmic domains are very short (ca. 30 amino acids) and divided into two constant regions (C1 and C2) separated by one variable (V) region. Here we attempted to map the cytoplasmic region responsible for the filopodia-inducing effect of syndecan-3. We found that only the C1-region was necessary for this effect. In addition, the deletion of the C2-region led to extensive membrane blebbing. Nevertheless, the elimination of the entire cytoplasmic region did not affect delivery of syndecan-3 to the plasma membrane. These results indicate that the different regions of syndecan-3 cytoplasmic domain have different functions probably by binding to distinct proteins.


Subject(s)
Cell Membrane/metabolism , Cytoplasm/metabolism , Membrane Glycoproteins/pharmacology , Proteoglycans/pharmacology , Pseudopodia/drug effects , Sequence Deletion , Animals , CHO Cells , Cell Cycle/drug effects , Cell Membrane/ultrastructure , Cricetinae , Pseudopodia/ultrastructure , Receptors, Fibroblast Growth Factor/metabolism , Syndecan-3
19.
Angiogenesis ; 5(3): 167-72, 2002.
Article in English | MEDLINE | ID: mdl-12831057

ABSTRACT

This study compares phenotypic changes of human umbilical endothelial vein cells cultured in three-dimensional collagen matrixes in the presence of basic fibroblast growth factor or on Matrigel coats. Under both conditions, endothelial cells rapidly assembled into an irregular network of tubular structures with a high frequency of intercellular or lumen-like spaces. Tubular structures were characterized and compared by phase-contrast, confocal and electron microscopy. The dominant mechanism of lumen-like formation was highly model-dependent. Ultrastructural analyses of capillary-like structures and the mechanism of lumen-like formation indicated that the in vivo angiogenesis was better reproduced in the collagen model.


Subject(s)
Capillaries/physiology , Collagen , Drug Combinations , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/pharmacology , Laminin , Neovascularization, Physiologic/physiology , Proteoglycans , Animals , Capillaries/drug effects , Capillaries/ultrastructure , Cell Culture Techniques/methods , Cells, Cultured , Endothelium, Vascular/ultrastructure , Humans , Microscopy, Electron , Microscopy, Electron, Scanning , Neovascularization, Physiologic/drug effects , Rats , Umbilical Veins
20.
Eur J Neurosci ; 18(9): 2538-44, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14622154

ABSTRACT

The blood-brain barrier (BBB) ensures the homeostasis of the brain microenvironment, mostly through complex tight junctions between brain endothelial cells that prevent the passage of hydrophilic molecules from blood to brain and vice versa. A recent study has shown in vivo that systemic administration of erythropoietin (Epo) protects against brain injury. Using an in vitro model of the bovine BBB, we observed that the expression of the Epo receptor is modulated by its ligand and hypoxic stimuli such as vascular endothelial growth factor (VEGF) treatment. In addition, Epo protects against the VEGF-induced permeability of the BBB, decreases the levels of endothelial nitric oxide synthase and restores junction proteins. The kinetic transport experiments revealed the capacity of Epo to cross the in vitro BBB in a saturable and specific way. Our results suggest a new mechanism for Epo-induced neuroprotection, in which circulating Epo controls and maintains the BBB through an Epo receptor signalling pathway and the re-establishment of cell junctions.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Erythropoietin/physiology , Receptors, Erythropoietin/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Astrocytes/physiology , Blood-Brain Barrier/metabolism , Blotting, Western , Brain/physiology , Capillary Permeability/physiology , Cattle , Coculture Techniques , Endothelial Cells/metabolism , Erythropoietin/pharmacology , Fluorescent Antibody Technique , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Rats , Receptors, Erythropoietin/metabolism , Tight Junctions/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL