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










Publication year range
1.
Angiogenesis ; 16(2): 353-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23254820

ABSTRACT

VEGF-A is a crucial growth factor for blood vessel homeostasis and pathological angiogenesis. Due to alternative splicing of its pre-mRNA, VEGF-A is produced under several isoforms characterized by the combination of their C-terminal domains, which determines their respective structure, availability and affinity for co-receptors. As controversies still exist about the specific roles of these exon-encoded domains, we systematically compared the properties of eight natural and artificial variants containing the domains encoded by exons 1-4 and various combinations of the domains encoded by exons 5, 7 and 8a or 8b. All the variants (VEGF111a, VEGF111b, VEGF121a, VEGF121b, VEGF155a, VEGF155b, VEGF165a, VEGF165b) have a similar affinity for VEGF-R2, as determined by Surface plasmon resonance analyses. They strongly differ however in terms of binding to neuropilin-1 and heparin/heparan sulfate proteoglycans. Data indicate that the 6 amino acids encoded by exon 8a must be present and cooperate with those of exons 5 or 7 for efficient binding, which was confirmed in cell culture models. We further showed that VEGF165b has inhibitory effects in vitro, as previously reported, but that the shortest VEGF variant possessing also the 6 amino acids encoded by exon 8b (VEGF111b) is remarkably proangiogenic, demonstrating the critical importance of domain interactions for defining the VEGF properties. The number, size and localization of newly formed blood vessels in a model of tumour angiogenesis strongly depend also on the C-terminal domain composition, suggesting that association of several VEGF isoforms may be more efficient for treating ischemic diseases than the use of any single variant.


Subject(s)
Neovascularization, Pathologic , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology , Alternative Splicing , Base Sequence , Blotting, Western , Capillary Permeability , Cloning, Molecular , DNA Primers , HEK293 Cells , Humans , Immunohistochemistry , Ligands , Phosphorylation , Protein Binding , Proteolysis , Surface Plasmon Resonance , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Chem Commun (Camb) ; 47(19): 5506-8, 2011 May 21.
Article in English | MEDLINE | ID: mdl-21455511

ABSTRACT

This communication reports the synthesis, characterization and in vivo evaluation in mice of a new tri-tyrosine conjugated MR contrast agent, which may help to identify vulnerable plaques in atherosclerosis by targeting the lipid core.


Subject(s)
Contrast Media/chemical synthesis , Dextrans/chemical synthesis , Magnetic Resonance Imaging/methods , Plaque, Atherosclerotic/diagnosis , Tyrosine/analogs & derivatives , Animals , Contrast Media/chemistry , Dextrans/chemistry , Mice , Tyrosine/chemistry
3.
Int J Cancer ; 126(5): 1095-108, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-19642141

ABSTRACT

The aim of our study was to investigate whether myofibroblasts and the chemokine monocyte chemoattractant protein-1 (MCP-1)/CCL2 may play a role in hepatocellular carcinoma progression. We observed that hepatic myofibroblast LI90 cells express MCP-1/CCL2 mRNA and secrete this chemokine. Moreover, myofibroblast LI90 cell-conditioned medium (LI90-CM) induces human hepatoma Huh7 cell migration and invasion. These effects are strongly reduced when a MCP-1/CCL2-depleted LI90-CM was used. We showed that MCP-1/CCL2 induces Huh7 cell migration and invasion through its G-protein-coupled receptor CCR2 and, to a lesser extent, through CCR1 only at high MCP-1/CCL2 concentrations. MCP-1/CCL2's chemotactic activities rely on tyrosine phosphorylation of focal adhesion components and depend on matrix metalloproteinase (MMP)-2 and MMP-9. Furthermore, we observed that Huh7 cell migration and invasion induced by the chemokine are strongly inhibited by heparin, by beta-D-xyloside treatment of cells and by anti-syndecan-1 and -4 antibodies. Finally, we developed a 3-dimensional coculture model of myofibroblast LI90 and Huh7 cells and demonstrated that MCP-1/CCL2 and its membrane partners, CCR1 and CCR2, may be involved in the formation of mixed hepatoma-myofibroblast spheroids. In conclusion, our data show that human liver myofibroblasts act on hepatoma cells in a paracrine manner to increase their invasiveness and suggest that myofibroblast-derived MCP-1/CCL2 could be involved in the pathogenesis of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Movement/physiology , Chemokine CCL2/metabolism , Fibroblasts/metabolism , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Liver/cytology , Liver/metabolism , Liver Neoplasms/pathology , Neoplasm Invasiveness/physiopathology , RNA Interference , Surface Plasmon Resonance
4.
Glycobiology ; 19(12): 1511-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19717493

ABSTRACT

We have recently reported that the CXC-chemokine stromal cell-derived factor-1 (SDF-1)/CXCL12 induces proliferation, migration, and invasion of the Huh7 human hepatoma cells through its G-protein-coupled receptor CXCR4 and that glycosaminoglycans (GAGs) are involved in these events. Here, we demonstrate by surface plasmon resonance that the chemokine binds to GAG mimetics obtained by grafting carboxylate, sulfate or acetate groups onto a dextran backbone. We also demonstrate that chemically modified dextrans inhibit SDF-1/CXCL12-mediated in vitro chemotaxis and anchorage-independent cell growth in a dose-dependent manner. The binding of GAG mimetics to the chemokine and their effects in modulating the SDF-1/CXCL12 biological activities are mainly related to the presence of sulfate groups. Furthermore, the mRNA expression of enzymes involved in heparan sulfate biosynthesis, such as exostosin-1 and -2 or N-deacetylase N-sulfotransferases remained unchanged, but heparanase mRNA and protein expressions in Huh7 cells were decreased upon GAG mimetic treatment. Moreover, decreasing heparanase-1 mRNA levels by RNA interference significantly reduced SDF-1/CXCL12-induced extracellular signal-regulated kinase 1/2 (ERK 1/2) phosphorylation. Therefore, we suggest that GAG mimetic effects on SDF-1/CXCL12-mediated hepatoma cell chemotaxis may rely on decreased heparanase expression, which impairs SDF-1/CXCL12's signaling. Altogether, these data suggest that GAG mimetics may compete with cellular heparan sulfate chains for the binding to SDF-1/CXCL12 and may affect heparanase expression, leading to reduced SDF-1/CXCL12 mediated in vitro chemotaxis and growth of hepatoma cells.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Chemokine CXCL12/antagonists & inhibitors , Glycosaminoglycans/pharmacology , Liver Neoplasms/pathology , Binding, Competitive/drug effects , Carcinoma, Hepatocellular/metabolism , Cell Adhesion/drug effects , Cell Communication/drug effects , Chemokine CXCL12/metabolism , Chemokine CXCL12/pharmacology , Glucuronidase/antagonists & inhibitors , Glucuronidase/biosynthesis , Glycosaminoglycans/agonists , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Humans , Liver Neoplasms/metabolism , Neoplasm Invasiveness , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
5.
Biochim Biophys Acta ; 1790(10): 1314-26, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19632304

ABSTRACT

BACKGROUND: We previously demonstrated that the CC-chemokine Regulated upon Activation, Normal T cell Expressed and Secreted (RANTES)/CCL5 exerts pro-tumoral effects on human hepatoma Huh7 cells through its G protein-coupled receptor, CCR1. Glycosaminoglycans play major roles in these biological events. METHODS: In the present study, we explored 1/ the signalling pathways underlying RANTES/CCL5-mediated hepatoma cell migration or invasion by the use of specific pharmacological inhibitors, 2/ the role of RANTES/CCL5 oligomerization in these effects by using a dimeric RANTES/CCL5, 3/ the possible involvement of two membrane heparan sulfate proteoglycans, syndecan-1 (SDC-1) and syndecan-4 (SDC-4) in RANTES/CCL5-induced cell chemotaxis and spreading by pre-incubating cells with specific antibodies or by reducing SDC-1 or -4 expression by RNA interference. RESULTS AND CONCLUSION: The present data suggest that focal adhesion kinase phosphorylation, phosphoinositide 3-kinase-, mitogen-activated protein kinase- and Rho kinase activations are involved in RANTES/CCL5 pro-tumoral effects on Huh7 cells. Interference with oligomerization of the chemokine reduced RANTES/CCL5-mediated cell chemotaxis. This study also indicates that SDC-1 and -4 may be required for HepG2, Hep3B and Huh7 human hepatoma cell migration, invasion or spreading induced by the chemokine. These results also further demonstrate the involvement of glycosaminoglycans as the glycosaminoglycan-binding deficient RANTES/CCL5 variant, in which arginine 47 was replaced by lysine, was devoid of effect. GENERAL SIGNIFICANCE: The modulation of RANTES/CCL5-mediated cellular effects by targeting the chemokine-syndecan interaction could represent a new therapeutic approach for hepatocellular carcinoma.


Subject(s)
Cell Movement/drug effects , Chemokine CCL5/pharmacology , Syndecan-1/metabolism , Syndecan-4/metabolism , Anthracenes/pharmacology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chemokine CCL5/chemistry , Chemotaxis/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Protein Multimerization , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Surface Plasmon Resonance , Syndecan-1/genetics , Syndecan-1/immunology , Syndecan-4/genetics , Syndecan-4/immunology
6.
Blood ; 113(21): 5176-85, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19270265

ABSTRACT

Human T-cell lymphotropic virus type 1 (HTLV-1) entry involves the interaction between the surface (SU) subunit of the Env proteins and cellular receptor(s). Previously, our laboratories demonstrated that heparan sulfate proteoglycans (HSPGs) and neuropilin-1 (NRP-1), a receptor of VEGF(165), are essential for HTLV-1 entry. Here we investigated whether, as when binding VEGF(165), HSPGs and NRP-1 work in concert during HTLV-1 entry. VEGF(165) binds to the b domain of NRP-1 through both HSPG-dependent and -independent interactions, the latter involving its exon 8. We show that VEGF(165) is a selective competitor of HTLV-1 entry and that HTLV-1 mimics VEGF(165) to recruit HSPGs and NRP-1: (1) the NRP-1 b domain is required for HTLV-1 binding; (2) SU binding to target cells is blocked by the HSPG-binding domain of VEGF(165); (3) the formation of Env/NRP-1 complexes is enhanced by HSPGs; and (4) the HTLV SU contains a motif homologous to VEGF(165) exon 8. This motif directly binds to NRP-1 and is essential for HTLV-1 binding to, internalization into, and infection of CD4(+) T cells and dendritic cells. These findings demonstrate that HSPGs and NRP-1 function as HTLV-1 receptors in a cooperative manner and reveal an unexpected mimicry mechanism that may have major implications in vivo.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Human T-lymphotropic virus 1/pathogenicity , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Virus Attachment , Binding, Competitive , Cells, Cultured , Gene Products, env/metabolism , HTLV-I Infections/virology , Humans , Molecular Mimicry , Protein Binding , Receptors, Virus/metabolism
7.
Biochim Biophys Acta ; 1790(2): 141-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19026722

ABSTRACT

BACKGROUND: P-selectin is an adhesion receptor expressed on activated platelets and endothelial cells. Its natural ligand, P-selectin glycoprotein ligand-1, is expressed on leucocytes and the P-selectin/PSGL-1 interaction is involved in leukocyte rolling. We have compared the interaction of P-selectin with several low molecular weight polysaccharides: fucoidan, heparin and dextran sulfate. METHODS: Binding assays were obtained from the interaction of the polysaccharides with Sialyl Lewis X and PSGL-1 based constructs onto microtiter plates coated with P-selectin. SELDI TOF mass spectrometry was performed with anionic chips arrays coated with P-selectin in the absence or in the presence of polysaccharides. Kd were obtained from surface plasmon resonance experiments with immobilized P-selectin constructs, polysaccharides being injected in the mobile phase. Human whole blood flow cytometry experiments were performed with fluorescein isothiocyanate labelled polysaccharides with or without platelets activators. RESULTS: The fucoidan prevented P-selectin binding to Sialyl Lewis X with an IC(50) of 20 nM as compared to 400 nM for heparin and <25000 nM for dextran sulfate. It exhibited the highest affinity for immobilized P-selectin with a KD of 1.2 nM, two orders of magnitude greater than the K(D) of the other polysaccharides. Mass spectrometry evidenced the formation of a complex between P-selectin and fucoidan. The intensity of the fucoidan binding to platelets was dependent on the level of platelet activation. Competition between fucoidan and an anti P-selectin antibody demonstrated the specificity of the interaction. GENERAL SIGNIFICANCE: Low molecular weight fucoidan is a promising therapeutic agent of natural origin for biomedical applications.


Subject(s)
Blood Platelets/metabolism , P-Selectin/metabolism , Polysaccharides/metabolism , Adult , Anticoagulants/chemistry , Anticoagulants/metabolism , Anticoagulants/pharmacology , Blood Platelets/drug effects , Dextran Sulfate/metabolism , Dextran Sulfate/pharmacology , Drug Evaluation, Preclinical , Heparin/metabolism , Heparin/pharmacology , Humans , Membrane Glycoproteins/metabolism , Molecular Weight , Oligosaccharides/pharmacology , Platelet Activation/drug effects , Polysaccharides/chemistry , Polysaccharides/pharmacology , Protein Binding/physiology , Sialyl Lewis X Antigen , Substrate Specificity
8.
Biochim Biophys Acta ; 1780(4): 723-32, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18325345

ABSTRACT

We previously demonstrated that a non sulfated analogue of heparin, phenylacetate carboxymethyl benzylamide dextran (NaPaC) inhibited angiogenesis. Here, we observed that NaPaC inhibited the VEGF165 binding to both VEGFR2 and NRP-1 and abolished VEGFR2 activity. Further, we explored the effects of NaPaC on VEGF165 interactions with its receptors, VEGFR2 and NRP-1, co-receptor of VEGFR2. Surface plasmon resonance and affinity gel electrophoresis showed that NaPaC interacted directly with VEGF165, VEGFR2 and NRP-1 but not with heparin-independent factor such as VEGF121. NaPaC completely inhibited the heparin binding to VEGF165, NRP-1 and VEGFR2. We found that NaPaC bound to all three molecules, VEGF165, VEGFR2 and NRP-1, but was more effective in inhibiting heparin binding to VEGF165. These results suggested that heparin binding sites of VEGFR2 and NRP-1 were different from those of VEGF165.


Subject(s)
Dextrans/metabolism , Heparin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Binding Sites , Binding, Competitive/drug effects , Blotting, Western , Cells, Cultured , Dextrans/pharmacology , Dose-Response Relationship, Drug , Heparin/pharmacology , Neuropilin-1/genetics , Neuropilin-1/metabolism , Protein Binding/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Swine , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
9.
Am J Pathol ; 172(1): 167-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18079435

ABSTRACT

Vascular endothelial growth factor (VEGF) is essential for breast cancer progression and is a relevant target in anti-angiogenesis. Although VEGF121 and VEGF165, the fully or partially secreted isoforms, respectively, have been the focus of intense studies, the role of the cell-associated VEGF189 isoform is not understood. To clarify the contribution of VEGF189 to human mammary carcinogenesis, we established several clones of MDA-MB-231 cells stably overexpressing VEGF189 (V189) and VEGF165 (V165). V189 and V165 clones increased tumor growth and angiogenesis in vivo. Remarkably, V165 induced the most rapid tumor uptake, whereas V189 increased vasodilation. In vitro overexpression of VEGF165 and VEGF189 increases the proliferation and chemokinesis of these cancer cells. Interestingly, overexpression of VEGF189 increased cell adhesion on fibronectin (1.9-fold) and vitronectin (1.6-fold), as compared to VEGF165, through alpha5beta1 and alphavbeta5 integrins. Using the BIACore system we demonstrated for the first time that VEGF189 binds directly to neuropilin-1, which is strongly expressed in MDA-MB-231 cells. In contrast, VEGF-R2 was not significantly expressed and VEGF-R1 was expressed at low level. Our in vitro results suggest an autocrine effect of VEGF189 on breast cancer cells, probably through neuropilin-1. In conclusion, our data indicate that VEGF189 participates in mammary tumor growth through both angiogenesis and nonangiogenic functions. Whether VEGF189 overexpression is correlated to prognosis in human breast tumors remains to be established.


Subject(s)
Breast Neoplasms/metabolism , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/physiology , Animals , Breast Neoplasms/blood supply , Carcinoma/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Mammary Neoplasms, Animal/blood supply , Mammary Neoplasms, Animal/metabolism , Mice , Mice, SCID , Neoplasm Transplantation , Vascular Endothelial Growth Factor A/metabolism
10.
Clin Exp Metastasis ; 25(1): 53-64, 2008.
Article in English | MEDLINE | ID: mdl-17952617

ABSTRACT

The integrin alpha(v)beta(3) is involved in multiple aspects of malignant cancer, including tumor angiogenesis and metastasis, which makes the receptor a key target for the development of anti-cancer therapies. We report here on the production, the characterization and the in vivo anti-angiogenic and anti-metastatic properties of a novel alpha(v)beta(3)-binding disintegrin, DisBa-01, isolated from a cDNA library made with RNAs from the venom gland of Bothrops alternatus. The 11,637 Da-recombinant monomeric form of DisBa-01 displayed an RGD motif and interacted with purified alpha(v)beta(3) integrin in surface plasmon resonance studies, in a dose-dependent and cation sensitive manner. A three-dimensional molecular model of DisBa-01 in complex with alpha(v)beta(3) predicted a large surface of contacts with the beta(3) subunit. DisBa-01 inhibited the adhesion of alpha(v)beta(3)-expressing human microvascular endothelial cell line-1 (HMEC-1) and murine melanoma cell line B16F10 to vitronectin (IC(50) = 555 nM and 225 nM, respectively), and transiently inhibited their proliferation without direct cell toxicity, but did not affect the binding nor the proliferation of a human breast cancer-derived cell line (MDA-MB-231) not expressing alpha(v)beta(3). In vivo, DisBa-01 dose-dependently decreased bFGF-induced angiogenesis in a matrigel plug assay in athymic nude mice (IC(50) = 83 nM). When injected intravenously to C57BL/6 mice together with B16F10 melanoma cells, DisBa-01 time- and dose-dependently inhibited lung metastasis monitored by bioluminescent imaging. We conclude that DisBa-01 is a potent new inhibitor of alpha(v)beta(3)-dependent adherence mechanisms involved in neo-vascularization and tumor metastasis processes.


Subject(s)
Crotalid Venoms/pharmacology , Disintegrins/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Amino Acid Motifs , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Base Sequence , Bothrops , Cell Adhesion/drug effects , Cloning, Molecular , Crotalid Venoms/chemistry , Disintegrins/chemistry , Disintegrins/genetics , Fibroblast Growth Factors/metabolism , Humans , Integrin alphaVbeta3/drug effects , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
11.
Peptides ; 28(12): 2397-402, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17983687

ABSTRACT

Heptapeptide ATWLPPR (A7R), identified in our laboratory by screening a mutated phage library, was shown to bind specifically to neuropilin-1 (NRP-1) and then to selectively inhibit VEGF(165) binding to this receptor. In vivo, treatment with A7R resulted in decreasing breast cancer angiogenesis and growth. The present work is focused on structural characterization of A7R. Analogs of the peptide, obtained by substitution of each amino acid with alanine (alanine-scanning) or by amino acid deletion, have been systematically assayed to determine the relative importance of the side chains of each residue with respect to the inhibitory effect of A7R on VEGF(165) binding to NRP-1. We show here the importance of the C-terminal sequence LPPR and particularly the key role of C-terminal arginine. In solution, A7R displays significant secondary structure of the backbone adopting an extended conformation. However, the functional groups of arginine are very flexible in the absence of NRP-1 pointing to an induced fit upon binding to the receptor. A MD trajectory of the A7R/NRP-1 complex in explicit water, based on the recent tuftsin/NRP-1 crystal structure, has revealed the hydrogen-bonding network that contributes to A7R's binding activity.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Neuropilin-1/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Animals , In Vitro Techniques , Models, Molecular , Multiprotein Complexes , Protein Binding , Protein Conformation , Rats , Recombinant Proteins/metabolism , Structure-Activity Relationship , Thermodynamics , Vascular Endothelial Growth Factor A/metabolism
12.
Mol Cancer Ther ; 6(11): 2948-58, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18025279

ABSTRACT

The CC-chemokine regulated on activation, normal T-cell expressed, and presumably secreted (RANTES)/CCL5 mediates its biological activities through activation of G protein-coupled receptors, CCR1, CCR3, or CCR5, and binds to glycosaminoglycans. This study was undertaken to investigate whether this chemokine is involved in hepatoma cell migration or invasion and to modulate these effects in vitro by the use of glycosaminoglycan mimetics. We show that the human hepatoma Huh7 and Hep3B cells express RANTES/CCL5 G protein-coupled receptor CCR1 but not CCR3 nor CCR5. RANTES/CCL5 binding to these cells depends on CCR1 and glycosaminoglycans. Moreover, RANTES/CCL5 strongly stimulates the migration and the invasion of Huh7 cells and to a lesser extent that of Hep3B cells. RANTES/CCL5 also stimulates the tyrosine phosphorylation of focal adhesion kinase and activates matrix metalloproteinase-9 in Huh7 hepatoma cells, resulting in increased invasion of these cells. The fact that RANTES/CCL5-induced migration and invasion of Huh7 cells are both strongly inhibited by anti-CCR1 antibodies and heparin, as well as by beta-d-xyloside treatment of the cells, suggests that CCR1 and glycosaminoglycans are involved in these events. We then show by surface plasmon resonance that synthetic glycosaminoglycan mimetics, OTR4120 or OTR4131, directly bind to RANTES/CCL5. The preincubation of the chemokine with each of these mimetics strongly inhibited RANTES-induced migration and invasion of Huh7 cells. Therefore, targeting the RANTES-glycosaminoglycan interaction could be a new therapeutic approach for human hepatocellular carcinoma.


Subject(s)
Biomimetic Materials/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/pharmacology , Glycosaminoglycans/pharmacology , Liver Neoplasms/pathology , Biomimetic Materials/chemical synthesis , Cell Line, Tumor , Chemotaxis/drug effects , Glycosaminoglycans/chemistry , Humans , Neoplasm Invasiveness/pathology
13.
Lung Cancer ; 58(2): 205-13, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17686546

ABSTRACT

INTRODUCTION: Investigation of the role of vascular endothelial growth factor-C (VEGF-C) and VEGF receptor-3 (VEGFR-3) in non-small-cell lung cancer (NSCLC) has mainly focused on lymph node (LN) metastasis related to lymphangiogenesis. However, the coexpression of VEGF-C/VEGFR-3 by tumour cells can independently play an important role. The present study was therefore designed to evaluate VEGF-C/VEGFR-3 coexpression in tumour cells from the primary tumour and corresponding LN metastases. METHODS: VEGF-C and VEGFR-3 expression in cancer cells were evaluated by immunohistochemistry in 92 NSCLC samples and 45 metastatic LNs. Ki67 expression and mitotic index (MI) in tumours and clinicopathological data were analysed concurrently. RESULTS: VEGFR-3 and VEGF-C expression were observed in 42% and 74% of tumours, respectively. Concurrent expression of VEGF-C and VEGFR-3, observed in 39% of tumours, was significantly associated with a higher proliferation rate and a higher incidence of LN metastases. VEGF-C expression in tumour cells was observed in 100% of metastatic LN and VEGF-C/VEGFR-3 coexpression was observed in 71% of metastatic LN. Finally, concurrent expression of VEGF-C/VEGFR-3 in the primary tumour was associated with poor disease-free survival on univariate analysis. CONCLUSION: In NSCLC cancer cells, VEGF-C/VEGFR-3 coexpression suggests an autocrine/paracrine loop responsible for a high proliferation rate in tumour cells. As VEGF-C/VEGFR-3 coexpression is very frequent in metastatic LN tumour cells, it can be hypothesised that this coexpression participates in the growth of LN metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymph Nodes/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Adult , Aged , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease-Free Survival , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Proportional Hazards Models
14.
J Drug Target ; 14(10): 694-706, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17162739

ABSTRACT

In this paper, liposomes containing a lipopeptide bearing a ligand specifically recognized by neuropilin-1 (NRP-1) have been used to target a human breast cancer cell line overexpressing this receptor. The synthesis of this lipopeptide, C16-A7R, formed by the sequence of 7 amino acids ATWLPPR, linked to a palmitoyl fatty chain by an amide bond was described. After the characterisation of cationic liposomes formulated with the lipopeptide, the results obtained using various techniques showed that the lipopeptide-based liposomes were well accumulated in cells of the human breast cancer line MDA-MB-231 overexpressing NRP-1. Delivery of reporter genes expressing either beta-galactosidase (beta-gal) or green fluorescent protein (GFP) was selectively enhanced in these cells when compared with NRP-1-negative cells. In MDA-MB-231 cells, an increase by 250% in beta-gal activity was observed when delivered by lipopeptide-based liposomes compared to cationic liposomes alone.


Subject(s)
DNA/administration & dosage , DNA/genetics , Liposomes/chemistry , Neuropilin-1/biosynthesis , Neuropilin-1/genetics , Peptides/chemistry , Blotting, Western , Cell Division/physiology , Cell Line , Cell Survival/physiology , Chromatography, High Pressure Liquid , Cytomegalovirus/genetics , Flow Cytometry , Genes, Reporter/genetics , Green Fluorescent Proteins/chemistry , Indicators and Reagents , Mass Spectrometry , Microscopy, Fluorescence , Peptides/isolation & purification , Plasmids/genetics , Tetrazolium Salts , Thiazoles , Transfection , beta-Galactosidase/genetics
15.
J Biol Chem ; 281(49): 37844-52, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17028197

ABSTRACT

Therapeutic induction of angiogenesis is a potential treatment for chronic ischemia. Heparan sulfate proteoglycans are known to play an important role by their interactions with proangiogenic growth factors such as vascular endothelial growth factor (VEGF). Low molecular weight fucoidan (LMWF), a sulfated polysaccharide from brown seaweeds that mimic some biological activities of heparin, has been shown recently to promote revascularization in rat critical hindlimb ischemia. In this report, we first used cultured human endothelial cells (ECs) to investigate the possible ability of LMWF to enhance the actions of VEGF(165). Data showed that LMWF greatly enhances EC tube formation in growth factor reduced matrigel. LMWF is a strong enhancer of VEGF(165)-induced EC chemotaxis, but not proliferation. In addition, LMWF has no effect on VEGF(121)-induced EC migration, a VEGF isoform that does not bind to heparan sulfate proteoglycans. Then, with binding studies using (125)I-VEGF(165), we observed that LMWF enhances the binding of VEGF(165) to recombinant VEGFR-2 and Neuropilin-1 (NRP1), but not to VEGFR-1. Surface plasmon resonance analysis showed that LMWF binds with high affinity to VEGF(165) (1.2 nm) and its receptors (5-20 nm), but not to VEGF(121). Pre-injection of LMWF on immobilized receptors shows that VEGF(165) has the highest affinity for VEGFR-2 and NRP1, as compared with VEGFR-1. Overall, the effects of LMWF were much more pronounced than those of LMW heparin. These findings suggested an efficient mechanism of action of LMWF by promoting VEGF(165) binding to VEGFR-2 and NRP1 on ECs that could help in stimulating therapeutic revascularization.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/physiology , Neuropilin-1/metabolism , Polysaccharides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Humans , Molecular Weight , Neovascularization, Physiologic/drug effects , Neuropilin-1/genetics , Polysaccharides/chemistry , Protein Binding , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
16.
Life Sci ; 79(25): 2370-81, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16959272

ABSTRACT

Neuropilin-1 (NRP-1), a non-tyrosine kinase receptor of vascular endothelial growth factor-165 (VEGF165), was found expressed on endothelial and some tumor cells. Since its overexpression is correlated with tumor angiogenesis and progression, the targeting of NRP-1 could be a potential anti-cancer strategy. To explore this hypothesis, we identified a peptide inhibiting the VEGF165 binding to NRP-1 and we tested whether it was able to inhibit tumor growth and angiogenesis. To prove the target of peptide action, we assessed its effects on binding of radiolabeled VEGF165 to recombinant receptors and to cultured cells expressing only VEGFR-2 (KDR) or NRP-1. Antiangiogenic activity of the peptide was tested in vitro in tubulogenesis assays and in vivo in nude mice xenotransplanted in fat-pad with breast cancer MDA-MB-231 cells. Tumor volumes, vascularity and proliferation indices were determined. The selected peptide, ATWLPPR, inhibited the VEGF165 binding to NRP-1 but not to tyrosine kinase receptors, VEGFR-1 (flt-1) and KDR; nor did it bind to heparin. It diminished the VEGF-induced human umbilical vein endothelial cell proliferation and tubular formation on Matrigel and in co-culture with fibroblasts. Administration of ATWLPPR to nude mice inhibited the growth of MDA-MB-231 xenografts, and reduced blood vessel density and endothelial cell area but did not alter the proliferation indices of the tumor. In conclusion, ATWLPPR, a previously identified KDR-interacting peptide, was shown to inhibit the VEGF165 interactions with NRP-1 but not with KDR and to decrease the tumor angiogenesis and growth, thus validating, in vivo, NRP-1 as a possible target for antiangiogenic and antitumor agents.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Neuropilin-1/metabolism , Peptide Fragments/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Blotting, Western , CHO Cells , Cells, Cultured , Coculture Techniques , Cricetinae , Cross-Linking Reagents , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoenzyme Techniques , Iodine Radioisotopes , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Protein Binding , Umbilical Veins , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
17.
Nucl Med Biol ; 31(5): 575-81, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219275

ABSTRACT

Neuropilins (NRP) are receptors of angiogenic vascular endothelial growth factor (VEGF). Their overexpression was correlated with tumor angiogenesis and growth suggesting that their specific targeting could provide a new marker of tumor progression. Here, we observed in vitro that new (99m)Tc-labeled derivative of anti-VEGF heptapeptide, ATWLPPR, binds to NRP1 but not to NRP2. Our radiotracer is stable up to 24 h in human serum and in cysteine challenge. But, its too low affinity and too fast extraction indicate further improvement to give a successful imaging of tumor in vivo.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Neuropilin-1/metabolism , Peptides/pharmacokinetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Female , Humans , Isotope Labeling/methods , Male , Metabolic Clearance Rate , Mice , Mice, Nude , Neuropilin-1/chemistry , Organ Specificity , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Reproducibility of Results , Sensitivity and Specificity , Technetium/chemistry , Technetium/pharmacology , Tissue Distribution
18.
Glycobiology ; 14(4): 311-23, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15033938

ABSTRACT

The stromal cell-derived factor-1 (SDF-1) is a CXC chemokine, which plays critical roles in migration, proliferation, and differentiation of leukocytes. SDF-1 is the only known ligand of CXCR4, the coreceptor of X4 HIV strains. We show that SDF-1 binds to high- and low-affinity sites on HeLa cells. Coimmunoprecipitation studies demonstrate that glycanated and oligomerized syndecan-4 but neither syndecan-1, syndecan-2, betaglycan, nor CD44 forms complexes with SDF-1 and CXCR4 on these cells as well as on primary lymphocytes or macrophages. Moreover, biotinylated SDF-1 directly binds in a glycosaminoglycans (GAGs)-dependent manner to electroblotted syndecan-4, and colocalization of SDF-1 with syndecan-4 was visualized by confocal microscopy. Glycosaminidases pretreatment of the HeLa cells or the macrophages decreases the binding of syndecan-4 to the complex formed by it and SDF-1. In addition, this treatment also decreases the binding of the chemokine to CXCR4 on the primary macrophages but not on the HeLa cells. Therefore GAGs-dependent binding of SDF-1 to the cells facilitates SDF-1 binding to CXCR4 on primary macrophages but not on HeLa cell line. Finally, an SDF-1-independent heteromeric complex between syndecan-4 and CXCR4 was visualized on HeLa cells by confocal microscopy as well as by electron microscopy. Moreover, syndecan-4 from lymphocytes, monocyte derived-macrophages, and HeLa cells coimmunoprecipitated with CXCR4. This syndecan-4/CXCR4 complex is likely a functional unit involved in SDF-1 binding. The role of these interactions in the pathophysiology of SDF-1 deserves further study.


Subject(s)
Chemokines, CXC/metabolism , Lymphocytes/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Proteoglycans/metabolism , Receptors, CXCR4/metabolism , Chemokine CXCL12 , Fluorescence , Gene Expression , HeLa Cells , Humans , Multiprotein Complexes/metabolism , Protein Binding , Proteoglycans/biosynthesis , Syndecan-4
19.
Biochim Biophys Acta ; 1617(1-2): 80-8, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14637022

ABSTRACT

Interaction of RANTES with its membrane ligands or receptors transduces multiple intracellular signals. Whether RANTES uses proteoglycans (PGs) belonging to the syndecan family to attach to primary cells expressing RANTES G-protein-coupled receptors (GPCRs) was investigated. We demonstrate that RANTES specifically binds to high and low affinity binding sites on human monocyte-derived macrophages (MDM). We show by co-immunoprecipitation experiments that RANTES is associated on these cells with syndecan-1 and syndecan-4, but neither with syndecan-2 nor with betaglycan, in addition to CD44 and its GPCRs, CCR5 and CCR1. Glycosaminidases pre-treatment of the monocyte derived-macrophages strongly decreases the binding of RANTES to syndecan-1 and syndecan-4 and also to CCR5, and abolishes RANTES binding to CD44. This suggests that glycosaminoglycans (GAGs) are involved in RANTES binding to the PGs and that such bindings facilitate the subsequent interaction of RANTES with CCR5, on the MDM, characterized by low membrane expression of CCR5. The role of these interactions in the pathophysiology of RANTES deserves further study.


Subject(s)
Cell Membrane/chemistry , Chemokine CCL5/chemistry , Macrophages/chemistry , Macrophages/metabolism , Membrane Glycoproteins/chemistry , Proteoglycans/chemistry , Cell Membrane/metabolism , Chemokine CCL5/metabolism , Humans , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/metabolism , Membrane Glycoproteins/biosynthesis , Protein Binding , Proteoglycans/biosynthesis , Syndecan-1 , Syndecan-4 , Syndecans
20.
Glycobiology ; 13(9): 623-34, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12773479

ABSTRACT

It is believed that proteoglycans influence biological properties of chemokines. We show that the CC chemokine RANTES binds not only to high-affinity binding sites on CCR5-positive HeLa cells but also to low-affinity binding sites on HeLa cells expressing or lacking RANTES G protein-coupled receptors. Coimmunoprecipitation studies demonstrate that RANTES forms complexes with glycanated syndecan (SD)-1 and -4, in addition to CCR5 on the CCR5-positive HeLa cells. Moreover, confocal microscopy analysis shows the colocalization of RANTES with SD-1 and -4. Glycosaminoglycans removal from the cells by glycosaminidases treatment prevented RANTES binding to SD-1 and -4 and decreased RANTES binding to CCR5 on the CCR5-positive HeLa cells. Removal of glycosaminoglycans by glycosaminidases treatment of the complexes, RANTES/SD-1/SD-4/+/-CCR5, immobilized on beads, reversed SD-1 and -4 bindings. Therefore, RANTES bindings to SD-1 and -4 depend on glycosaminoglycans and facilitate RANTES interaction with CCR5. Extracting plasma membrane cholesterol abolished the coimmunoprecipitation of SD-1 with RANTES, suggesting that rafts are involved in RANTES association to SD-1. Confocal microscopy analysis as well as coimmunoprecipitation experiments show a RANTES-independent heteromeric complex on the CCR5-positive HeLa cells, SD-1, SD-4, and CCR5. This complex is likely a functional unit in which proteoglycans may modulate RANTES binding to CCR5.


Subject(s)
Chemokine CCL5/metabolism , Membrane Glycoproteins/metabolism , Proteoglycans/metabolism , beta-Cyclodextrins , Cell Membrane/metabolism , Cyclodextrins , Fluorescent Antibody Technique , HeLa Cells , Humans , Precipitin Tests , Protein Binding , Receptors, CCR5/metabolism , Syndecan-1 , Syndecan-4 , Syndecans
SELECTION OF CITATIONS
SEARCH DETAIL
...