Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Curr Pharm Des ; 15(30): 3577-89, 2009.
Article in English | MEDLINE | ID: mdl-19860702

ABSTRACT

Angiogenesis and inflammation are closely integrated processes. Fibroblast growth factor-2 (FGF2) is a prototypic angiogenesis inducer belonging to the family of the heparin-binding FGF growth factors. FGF2 exerts its pro-angiogenic activity by interacting with various endothelial cell surface receptors, including tyrosine kinase receptors, heparan-sulfate proteoglycans, and integrins. A tight cross-talk exists between FGF2 and the inflammatory response in the modulation of blood vessel growth. Pentraxins act as soluble pattern recognition receptors with a wide range of functions in various pathophysiological conditions. The long-pentraxin PTX3 shares the C-terminal pentraxin-domain with short-pentraxins and possesses a unique N-terminal domain. These structural features indicate that PTX3 may have distinct biological/ligand recognition properties when compared to short-pentraxins. Co-expression of PTX3 and FGF2 has been observed in different inflammation/angiogenesis-dependent diseases. PTX3 binds FGF2 with high affinity and specificity. The interaction prevents the binding of FGF2 to its cognate tyrosine kinase receptors, leading to inhibition of the angiogenic activity of the growth factor. This suggests that PTX3 may exert a modulatory function by limiting the angiogenic activity of FGF2. An integrated approach that utilized PTX3 fragments, monoclonal antibodies, and surface plasmon resonance analysis has identified the FGF2-binding domain in the unique N-terminal extension of PTX3. On this basis, PTX3-derived synthetic peptides have been designed endowed with a significant antiangiogenic activity in vitro and in vivo. They may provide the basis for the development of novel antiangiogenic FGF2 antagonists.


Subject(s)
Angiogenesis Inhibitors/pharmacology , C-Reactive Protein/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Nerve Tissue Proteins/pharmacology , Peptides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , C-Reactive Protein/chemistry , Humans , Models, Molecular , Molecular Conformation , Neovascularization, Pathologic/drug therapy , Nerve Tissue Proteins/chemistry , Peptides/chemistry
2.
J Biol Chem ; 276(41): 37900-8, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11473122

ABSTRACT

The angiogenic basic fibroblast growth factor (FGF2) interacts with tyrosine kinase receptors (FGFRs) and heparan sulfate proteoglycans (HSPGs) in endothelial cells. Here, we report the FGF2 antagonist and antiangiogenic activity of novel sulfated derivatives of the Escherichia coli K5 polysaccharide. K5 polysaccharide was chemically sulfated in N- and/or O-position after N-deacetylation. O-Sulfated and N,O-sulfated K5 derivatives with a low degree and a high degree of sulfation compete with heparin for binding to 125I-FGF2 with different potency. Accordingly, they abrogate the formation of the HSPG.FGF2.FGFR ternary complex, as evidenced by their capacity to prevent FGF2-mediated cell-cell attachment of FGFR1-overexpressing HSPG-deficient Chinese hamster ovary (CHO) cells to wild-type CHO cells. They also inhibited 125I-FGF2 binding to FGFR1-overexpressing HSPG-bearing CHO cells and adult bovine aortic endothelial cells. K5 derivatives also inhibited FGF2-mediated cell proliferation in endothelial GM 7373 cells and in human umbilical vein endothelial (HUVE) cells. In all these assays, the N-sulfated K5 derivative and unmodified K5 were poorly effective. Also, highly O-sulfated and N,O-sulfated K5 derivatives prevented the sprouting of FGF2-transfected endothelial FGF2-T-MAE cells in fibrin gel and spontaneous angiogenesis in vitro on Matrigel of FGF2-T-MAE and HUVE cells. Finally, the highly N,O-sulfated K5 derivative exerted a potent antiangiogenic activity on the chick embryo chorioallantoic membrane. These data demonstrate the possibility of generating FGF2 antagonists endowed with antiangiogenic activity by specific chemical sulfation of bacterial K5 polysaccharide. In particular, the highly N,O-sulfated K5 derivative may provide the basis for the design of novel angiostatic compounds.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Escherichia coli/metabolism , Fibroblast Growth Factor 2/antagonists & inhibitors , Polysaccharides/pharmacology , Animals , CHO Cells , Carbohydrate Sequence , Cattle , Cells, Cultured , Chick Embryo , Cricetinae , Endothelium, Vascular/cytology , Humans , Polysaccharides/chemistry , Recombinant Proteins/antagonists & inhibitors
3.
Microvasc Res ; 61(3): 240-52, 2001 May.
Article in English | MEDLINE | ID: mdl-11336535

ABSTRACT

Intracellular Ca2+ signals elicited by nucleotide agonists were investigated in primary cultures of rat cardiac microvascular endothelial cells using the fura-2 technique. UTP increased the intracellular [Ca2+] in 94% of the cells, whereas 2MeSATP was active in 32%. The rank order of potency was ATP = UTP > 2MeSATP and the maximal response to 2MeSATP was lower compared to UTP and ATP. ATP and UTP showed strong homologous and heterologous desensitization. ATP fully inhibited the 2MeSATP response, while UTP abolished 2MeSATP-elicited transients in 25% of cells. 2MeSATP did not desensitize the UTP or ATP response. Adenosine 2',5'-diphosphate inhibited the response to 2MeSATP, while it did not modify the response to ATP and UTP. 2MeSATP was more sensitive to suramin than UTP and ATP. These results indicate that P(2Y1) and P(2Y2) receptors may be coexpressed in CMEC. Nucleotide-induced Ca2+ signals lacked a sustained plateau and were almost independent from extracellular Ca2+. ATP and UTP elicited Ca2+ transients longer than 2MeSATP-evoked transients. The kinetics of Ca2+ responses was not affected by bath solution stirring or ectonucleotidase inhibition. Furthermore, the nonhydrolyzable ATP analogue AMP-PNP induced Ca2+ signals similar to those elicited by ATP and UTP. These results suggest that the distinct kinetics of nucleotide-evoked Ca2+ responses do not depend on the activity of ectonucleotidases or ATP autocrine stimulation. The possibility that Ca2+ signals with different time courses may modulate different cellular responses is discussed.


Subject(s)
Calcium Signaling , Endothelium, Vascular/metabolism , Myocardium/metabolism , Receptors, Purinergic P2/metabolism , Animals , Calcium Signaling/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Kinetics , Microcirculation/cytology , Microcirculation/drug effects , Microcirculation/metabolism , Myocardium/cytology , Nucleotides/pharmacology , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Rats , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y2
4.
Cancer Res ; 61(1): 309-17, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196179

ABSTRACT

Fibroblast growth factor-2 (FGF2) is a pleiotropic heparin-binding growth factor endowed with a potent angiogenic activity in vitro and in vivo. To investigate the impact of the modulation of FGF2 expression on the neovascularization at different stages of tumor growth, we generated stable transfectants (Tet-FGF2) from the human endometrial adenocarcinoma HEC-1-B cell line in which FGF2 expression is under the control of the tetracycline-responsive promoter (Tet-off system). After transfection, independent clones were obtained in which FGF2 mRNA and protein were up-regulated compared with parental cells. Also, the conditioned medium of Tet-FGF2 transfectants caused proliferation, urokinase-type plasminogen activator up-regulation, migration, and sprouting of cultured endothelial cells. A 3-day treatment of Tet-FGF2 cell cultures with tetracycline abolished FGF2 overexpression and the biological activity of the conditioned medium without affecting their proliferative capacity. Tet-FGF2 cells formed tumors when nude mice received s.c. injections. The administration of 2.0 mg/ml tetracycline in the drinking water before cell transplantation, continued throughout the whole experiment, inhibited FGF2 expression in Tet-FGF2 tumor lesions. This was paralleled by a significant decrease in the rate of tumor growth and vascularization to values similar to those observed in lesions generated by parental HEC-1-B cells. Tetracycline administration 20 days after tumor cell implant, although equally effective in reducing FGF2 expression and inhibiting tumor vascularity, only minimally impaired the growth of established Tet-FGF2 tumors. The results indicate that FGF2 expression deeply affects the initial tumor growth and neovascularization of HEC-1-B human endometrial adenocarcinoma in nude mice. On the contrary, the growth of established tumors appears to be independent of the inhibition of FGF2 expression and decreased vascular density. The possibility that a significant reduction of angiogenesis may not affect the progression of large tumors points to the use of antiangiogenic therapy in early tumor stage.


Subject(s)
Adenocarcinoma/blood supply , Endometrial Neoplasms/blood supply , Fibroblast Growth Factor 2/biosynthesis , Neovascularization, Pathologic/metabolism , Tetracycline/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Division/drug effects , Cell Division/physiology , DNA, Complementary/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Fibroblast Growth Factor 2/genetics , Gene Expression/drug effects , Genetic Vectors , Humans , Mice , Mice, Nude , Promoter Regions, Genetic , Time Factors , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Cell Sci ; 112 ( Pt 15): 2597-606, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10393815

ABSTRACT

Downstream signaling triggered by the binding of fibroblast growth factor-2 (FGF2) to its tyrosine-kinase receptors involves the activation of mitogen-activated protein kinase kinase (MEK) with consequent phosphorylation of extracellular signal-regulated kinases (ERKs). Here we demonstrate that FGF2 induces ERK1/2 activation in bovine aortic endothelial (BAE) cells and that the continuous presence of the growth factor is required for sustained ERK1/2 phosphorylation. This is prevented by the MEK inhibitors PD 098059 and U0126, which also inhibit FGF2-mediated upregulation of urokinase-type plasminogen activator (uPA) and in vitro formation of capillary-like structures in three-dimensional type I collagen gel. Various FGF2 mutants originated by deletion or substitution of basic amino acid residues in the amino terminus or in the carboxyl terminus of FGF2 retained the capacity to induce a long-lasting activation of ERK1/2 in BAE cells. Among them, K128Q/R129Q-FGF2 was also able to stimulate uPA production and morphogenesis whereas R129Q/K134Q-FGF2 caused uPA upregulation only. In contrast, K27, 30Q/R31Q-FGF2, K128Q/K138Q-FGF2 and R118,129Q/K119,128Q-FGF2 exerted a significant uPA-inducing and morphogenic activity in an ERK1/2-dependent manner only in the presence of heparin. Furthermore, no uPA upregulation and morphogenesis was observed in BAE cells treated with the deletion mutant (delta)27-32-FGF2 even in the presence of soluble heparin. Thus, mutational analysis of FGF2 dissociates the capacity of the growth factor to induce a persistent activation of ERK1/2 from its ability to stimulate uPA upregulation and/or in vitro angiogenesis. In conclusion, the data indicate that ERK1/2 phosphorylation is a key step in the signal transduction pathway switched on by FGF2 in endothelial cells. Nevertheless, a sustained ERK1/2 activation is not sufficient to trigger uPA upregulation and morphogenesis. FGF2 mutants may represent useful tools to dissect the signal transduction pathway(s) mediating the complex response elicited by an angiogenic stimulus in endothelial cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Endothelium, Vascular/physiology , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Mitogen-Activated Protein Kinases , Neovascularization, Physiologic , Urokinase-Type Plasminogen Activator/genetics , Amino Acid Sequence , Animals , Aorta , Butadienes/pharmacology , Capillaries , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/physiology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mutagenesis, Site-Directed , Neovascularization, Physiologic/drug effects , Nitriles/pharmacology , Recombinant Proteins/pharmacology , Sequence Deletion
6.
Am J Pathol ; 152(6): 1607-16, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626064

ABSTRACT

Three cell clones originated by transfection of human endometrial adenocarcinoma HEC-1-B cells with fibroblast growth factor-2 (FGF-2) cDNA and characterized by a different capacity to produce and secrete the growth factor were transplanted subcutaneously in nude mice. Corrosion casting of the tumor microvasculature of xenografts produced by injection of 2 x 10(6) or 10 x 10(6) FGF-2-B9 cells (which produce and secrete significant amounts of FGF-2), 10 x 10(6) FGF-2-A8 cells (which produce comparable amounts of FGF-2 but do not secrete it), or 10 x 10(6) control FGF-2-B8 cells (which produce only trace amounts of FGF-2) was performed after 14 days of growth. Interbranching distances, intervascular distances, branching angles, and vessel diameters were then determined using tridimensional stereo pairs of the casted tumor vascularity. When transplanted at the same concentration, FGF-2-B9 cells grew faster in nude mice compared with FGF-2-A8 and FGF-2-B8 clones. The total amount of new vessel formation was far higher in FGF-2-B9 tumors than in FGF-2-B8 or FGF-2-A8 tumors. Also, vessel courses were more irregular and blind-ending vessels and evasates were more frequent in FGF-2-B9 tumors. Moreover, FGF-2-B9 tumor microvasculature was characterized by a wider average vascular diameter and by an extreme variability of the diameter of each individual vessel along its course between two ramifications. No statistical differences were observed when the distribution curves of the values of intervascular distances, interbranching distances, and branching angles of the microvessel network were compared among the different experimental groups. The distinctive features of the microvasculature of FGF-2-B9 tumors were retained, at least in part, in the smaller lesions produced by injection of a limited number of cells. The data indicate that FGF-2 production and release confer to FGF-2-B9 cells the ability to stimulate the formation of new blood vessels with distinctive architectural features. Neovascularization of FGF-2-B9 lesions parallels the faster rate of growth of the neoplastic parenchyma. This does not affect the overall architecture of the microvessel network that appears to be primed by characteristics of the HEC-1-B tumor cell line and/or by the microenvironment of the host. To our knowledge, this work represents the first attempt to define the influence of a single, defined growth factor on the tridimensional tumor vascular pattern.


Subject(s)
Adenocarcinoma/blood supply , Endometrial Neoplasms/blood supply , Fibroblast Growth Factor 2/physiology , Animals , Corrosion Casting , Female , Fibroblast Growth Factor 2/metabolism , Humans , Mice , Mice, Nude , Microcirculation/pathology , Neoplasm Transplantation , Specific Pathogen-Free Organisms , Transfection , Tumor Cells, Cultured
7.
AIDS ; 11(6): 727-36, 1997 May.
Article in English | MEDLINE | ID: mdl-9143604

ABSTRACT

OBJECTIVE: To demonstrate that Tat modulates the plasminogen-dependent proteolytic activity of tumour cell lines derived from BK virus (BKV)/tat-transgenic mice by affecting the production of plasminogen activators (PA) and the PA inhibitor (PAI)-1 and to demonstrate that this occurs through mechanism(s) that are distinct from those responsible for transactivating activity of extracellular Tat. DESIGN AND METHODS: To assess whether endogenous Tat is responsible for PA activity in T53 adenocarcinoma cells, cell cultures were transfected with antisense Tat cDNA and evaluated for cell-associated PA activity by a plasmin chromogenic assay. The assay was also used to evaluate PA activity in T53 cells and T111 leiomyosarcoma cells stimulated by extracellular Tat. The type(s) of PA produced were identified by sodium dodecyl sulphate-polyacrylamide gel electrophoresis zymography. The levels of PAI-1 were evaluated by Western blotting. Tat transactivating activity was measured by a chloramphenicol acetyltransferase (CAT) enzyme-linked immunosorbent assay in HL3T1 cells containing integrated copies of an HIV-1 long terminal repeat (LTR)-CAT plasmid. RESULTS: Transfection of T53 cells with antisense Tat cDNA results in the decrease of Tat production and PA activity. Exogenously added Tat increases PA levels in T53 and in T111 cells. PA activity was identified as urokinase-type PA (uPA). Tat also increases the production of PAI-1 in T111 but not in T53 cells. Chloroquine and heparin have different affects on the LTR-CAT-transactivating and the PA-inducing activities of Tat. The fusion protein glutathione-S-transferase-Tat and the mutant Tat-1e, lacking the second Tat exon, cause LTR-CAT transactivation without stimulating uPA upregulation. CONCLUSIONS: Tat affects the fibrinolytic activity of tumour cell lines derived from BKV/tat-transgenic mice by modulating the production of both uPA and PAI-1 via autocrine and paracrine mechanisms of action. The capacity of Tat to modulate the plasminogen-dependent proteolytic activity of these tumour cell lines may contribute to their metastatic potential. The uPA-inducing activity of Tat depends upon specific biological and structural features of the Tat protein that are distinct from those responsible for its LTR-CAT-transactivating activity, suggesting distinct mechanisms of induction for the two biological responses.


Subject(s)
Gene Products, tat/metabolism , Up-Regulation , Urokinase-Type Plasminogen Activator/analysis , Animals , BK Virus/genetics , Chloramphenicol O-Acetyltransferase/genetics , Gene Products, tat/genetics , Gene Products, tat/pharmacology , Genetic Vectors , HIV Long Terminal Repeat , HIV-1/chemistry , Humans , Mice , Mice, Transgenic , Plasminogen Activator Inhibitor 1/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured , tat Gene Products, Human Immunodeficiency Virus
8.
J Biol Chem ; 272(17): 11313-20, 1997 Apr 25.
Article in English | MEDLINE | ID: mdl-9111037

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Tat protein is released from infected cells. Extracellular Tat enters the cell where it stimulates the transcriptional activity of HIV-long terminal repeat (LTR) and of endogenous genes. Heparin modulates the angiogenic (Albini, A., Benelli, R., Presta, M., Rusnati, M., Ziche, M., Rubartelli, A., Paglialunga, G., Bussolino, F., and Noonan, D. (1996) Oncogene 12, 289-297) and transcriptional (Mann, D. A., and Frankel, A. D. (1991) EMBO J. 10, 1733-1739) activity of extracellular Tat. Here we demonstrate that heparin binds specifically to recombinant HIV-1 Tat produced as glutathione S-transferase (GST) fusion protein and immobilized on glutathione-agarose beads. Heparin and heparan sulfate (HS), but not dermatan sulfate, chondroitin sulfates A and C, hyaluronic acid, and K5 polysaccharide, competed with 3H-labeled heparin for binding to immobilized GST-Tat and inhibited HIV-LTR transactivation induced by extracellular GST-Tat. Selective 2-O-, 6-O-, total-O-desulfation, or N-desulfation/N-acetylation dramatically reduced the capacity of heparin to bind GST-Tat. Totally-O-desulfated and 2-O-desulfated heparins also showed a reduced capacity to inhibit the transactivating activity of GST-Tat. Very low molecular weight heparins showed a significant decrease in their capacity to bind GST-Tat and to inhibit its LTR transactivating activity when compared with conventional 13.6-kDa heparin. However, when 3.0-kDa heparin was affinity chromatographed on immobilized GST-Tat to isolate binding and non-binding subfractions, the Tat-bound fraction was >/=1,000 times more potent than the unbound fraction in inhibiting the transactivating activity of GST-Tat. The results demonstrate that Tat interacts in a size-dependent manner with heparin/HS and that high affinity Tat-heparin interaction requires at least some 2-O-, 6-O-, and N-positions to be sulfated. The Tat binding activity of the glycosaminoglycans tested correlates with their capacity to affect the transactivating activity of extracellular Tat, indicating the possibility to design specific heparin/HS-like structures with Tat-antagonist activity.


Subject(s)
Gene Products, tat/metabolism , HIV-1 , Heparin/metabolism , Binding Sites , Gene Products, tat/genetics , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Glycosaminoglycans/pharmacology , HIV Long Terminal Repeat , Heparin/chemistry , Heparin/pharmacology , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Heparitin Sulfate/pharmacology , Protein Binding , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Sulfuric Acid Esters/chemistry , Transcriptional Activation/drug effects , tat Gene Products, Human Immunodeficiency Virus
9.
Cancer Res ; 55(20): 4729-38, 1995 Oct 15.
Article in English | MEDLINE | ID: mdl-7553656

ABSTRACT

The human endometrial adenocarcinoma HEC-1-B cell line was transfected with an expression vector harboring the human basic fibroblast growth factor (bFGF) cDNA under the control of the human beta-actin gene promoter. Stable transfectants were obtained in which a constitutive, limited overexpression of M(r) 24,000, 22,000, and 18,000 bFGF isoforms was observed. When transfectants were screened for the capacity to release the growth factor, significant amounts of bFGF were present in the conditioned medium and extracellular matrix of the bFGF-B9 clone but not of the bFGF-A8 clone, even though both cell lines produced similar levels of intracellular bFGF. When compared to parental cells, bFGF-B9 cells showed down-regulation of tyrosine kinase fibroblast growth factor receptors along with up-regulation of urokinase-type plasminogen activator expression which was abolished by incubation of the cell cultures with neutralizing anti-bFGF antibody. In vivo, bFGF-B9 cells formed highly vascularized tumors growing faster than parental cells when injected s.c. in nude mice. Also, they were more potent than nontransfected cells in inducing an angiogenic response in the rabbit cornea assay. In contrast, the bFGF-A8 cell phenotype was indistinguishable from parental cells both in vitro and in vivo. In conclusion, clonal differences exist within the HEC-1-B cell line in the capacity to release bFGF. bFGF export by human endometrial adenocarcinoma cells results in autocrine and paracrine effects that confer a growth advantage in vivo associated with increased neovascularization.


Subject(s)
Adenocarcinoma, Papillary/pathology , Endometrial Neoplasms/pathology , Fibroblast Growth Factor 2/metabolism , Neovascularization, Pathologic/pathology , Aged , Animals , Cell Division , Female , Fibroblast Growth Factor 2/genetics , Gene Expression , Humans , In Vitro Techniques , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/pathology , RNA, Messenger/genetics , Transfection , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
10.
Biochem J ; 303 ( Pt 2): 583-90, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7980421

ABSTRACT

Heparins from bovine mucosa and lung, and chemically modified heparins were assayed for their capacity to: (i) protect human recombinant basic fibroblast growth factor (bFGF) from tryptic cleavage; (ii) prevent 125I-bFGF binding to heparan sulphate proteoglycans present in the extracellular matrix and on the cell surface of fetal bovine aortic endothelial GM 7373 cell cultures; (iii) affect 125I-bFGF binding to high-affinity tyrosine kinase FGF receptors present on the cell membrane of GM 7373 cells; (iv) inhibit the mitogenic activity exerted by bFGF in the same cells. The results demonstrate that the potency shown by mucosal heparins in the different assays is a direct function of size, very-low-molecular-mass heparin (2.0 kDa) being significantly less effective on a molar basis than unfractionated heparin (13.6 kDa). Increased flexibility of the backbone structure, as observed in reduced/oxidized heparins of different size, does not affect the capacity of the polysaccharide to interact with bFGF. In contrast, selective 2-O-desulphation, but not 6-O-desulphation, drastically reduced the capacity of heparin to protect bFGF from proteolytic cleavage, to affect its interaction with low- and high-affinity sites, and to inhibit its mitogenic activity. Two preparations of bovine lung heparin, differing in molecular mass, were as effective as mucosal heparin in the bFGF-tryptic-digestion assay and the endothelial-cell proteoglycan-binding assay, but they were highly inefficient at inhibiting the capacity of bFGF to interact with its tyrosine kinase receptors. Bovine lung heparins were also less effective than mucosal heparin as bFGF antagonists in GM 7373-cell-proliferation assays. N-Desulphated/N-acetylated bovine lung heparin retained only a significant capacity to protect bFGF from tryptic cleavage. The results demonstrate that different chemical features of the heparin molecule, including decrease in molecular mass, selective desulphation, disaccharide composition and clustering, affect differently the capacity of the glycosaminoglycan to interact with bFGF and to influence its biological behaviour in different assays in vitro and in endothelial cell cultures. Our findings should aid the design of synthetic oligosaccharides aimed at improving the bioavailability of bFGF when administered in vivo as a therapeutic agent.


Subject(s)
Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/drug effects , Heparin/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Biological Availability , Cattle , Cell Division/drug effects , Cell Line , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/metabolism , Heparin/chemistry , Heparitin Sulfate/metabolism , Humans , Lung/metabolism , Molecular Weight , Mucous Membrane/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism
11.
Biochem Biophys Res Commun ; 203(1): 450-8, 1994 Aug 30.
Article in English | MEDLINE | ID: mdl-8074689

ABSTRACT

Interaction of basic fibroblast growth factor (bFGF) with heparan sulfate proteoglycans (HSPGs) plays an important role in the binding of bFGF to its tyrosine kinase receptor (FGFR). The molecular bases of this interaction were investigated by evaluating the capacity of conventional and selectively desulfated heparins i) to affect the binding of bFGF to FGFR and HSPGs of NIH 3T3 cells transfected with FGFR-1/flg cDNA, ii) to facilitate the interaction of bFGF with a recombinant soluble form of the extracellular domain of FGFR-1/flg (xcFGFR-1), and iii) to protect xcFGFR-1 from tryptic cleavage. 6-O-desulfated (6-O-DS) heparin, but not 2-O-desulfated (2-O-DS) and N-desulfated/N-acetylated (N-DS/N-Ac) heparins, retains the capacity to bind bFGF, as assessed by its ability to inhibit bFGF-binding to cell-associated FGFR-1 and HSPGs. On the other hand, at variance with conventional heparin, 2-O-DS, N-DS/N-Ac, and 6-O-DS heparins are all ineffective in potentiating the binding of bFGF to xcFGFR-1 and protecting xcFGFR-1 from tryptic cleavage. The data indicate that 6-O-sulfate groups are not essential for the interaction of heparin with bFGF but are involved in the interaction with xcFGFR-1. Our findings support the hypothesis that HSPGs modulate the binding of bFGF to FGFR through the formation of a ternary complex in which the glycosaminoglycan chains interact with bFGF via 2-O- and N-sulfate groups and with FGFR also via 6-O-sulfate groups.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Proteoglycans/chemistry , Proteoglycans/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Fibroblast Growth Factor 2/isolation & purification , Filaggrin Proteins , Heparan Sulfate Proteoglycans , Heparitin Sulfate/pharmacology , Humans , Kinetics , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Proteoglycans/pharmacology , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/biosynthesis , Receptors, Fibroblast Growth Factor/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sulfuric Acids , Transfection
12.
Eur J Biochem ; 214(1): 51-8, 1993 May 15.
Article in English | MEDLINE | ID: mdl-8508806

ABSTRACT

In the present study we have attempted a characterization of the biochemical bases of the interaction of human basic fibroblast growth factor (bFGF) with glycosaminoglycans (GAGs) in solution. This interaction has been evidenced as the capacity of different GAGs and various sulfated compounds to protect bFGF and different bFGF mutants from tryptic cleavage. Heparin protects bFGF from trypsin digestion in a dose-dependent fashion. Substitution by site-directed mutagenesis of two or more basic residues with neutral glutamine residues in the amino-terminal region bFGF(27-32) or in the carboxyl-terminal region bFGF(118-129) does not significantly affect the protective effect exerted by heparin. In contrast, heparin protection is abolished when the full region bFGF(27-32) is deleted. The capacity of different GAGs to protect bFGF from proteolytic cleavage decreases in the following order: heparin > heparan sulfate > dermatan sulfate = chondroitin sulfates A and C > hyaluronic acid = K5 polysaccharide, indicating that both the degree of sulfation and the backbone structure of GAG modulate its interaction with bFGF. This is confirmed by the different capacity of various sulfated compounds (including dextran sulfates, suramin, trypan blue, and sulfate ion) to protect bFGF from tryptic digestion. Moreover, tryptic digestion studies performed with various heparin molecules and dextran sulfates of different size, ranging from 2.0 kDa to 500 kDa, indicate that the number of bFGF molecules which interact with a single molecule of polysaccharide is related to the molecular mass of the GAG and that six hexose residues are sufficient to protect 1-2 molecules bFGF. In conclusion, our findings indicate that the capacity of GAGs to protect bFGF from tryptic cleavage depends upon their size, sulfation, distribution of the anionic sites along the chain, and structural requirements of the bFGF molecule. These studies will help to design synthetic oligosaccharides endowed with different bFGF agonist and/or antagonist activities.


Subject(s)
Fibroblast Growth Factor 2/chemistry , Glycosaminoglycans/chemistry , Heparin/chemistry , Amino Acid Sequence , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Dermatan Sulfate/chemistry , Dermatan Sulfate/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Glycosaminoglycans/metabolism , Heparin/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Trypsin/metabolism
13.
Growth Factors ; 8(1): 49-60, 1993.
Article in English | MEDLINE | ID: mdl-8448039

ABSTRACT

Immunolocalization of basic fibroblast growth factor (bFGF) was investigated in NIH 3T3 cells transfected with a cDNA encoding for the 18 kD form of human bFGF (18 kD-bFGF) or with a bFGF cDNA encoding for both 18 kD-bFGF and NH2-terminal extended high molecular weight forms of bFGF (HMW-bFGFs). Nuclear and cytoplasmic bFGF-immunoreactivity was observed in both transfectants. Nuclear bFGF immunoreactivity was evenly distributed during interfase and associated with condensed chromosomes throughout the mitotic cycle. Cell fractionation, followed by Western blot analysis, confirmed the presence of 18 kD-bFGF and of HMW-bFGFs in the nucleus of transfected cells. Also, both 18-kD bFGF and HMW-bFGFs copurified with nuclear chromatin. After trypsin digestion, chromatin-bound bFGFs showed a rapid degradation of the nuclear-targeting NH2-terminal extension of HMW-bFGFs which were converted to the 18 kD form. On the contrary, 18 kD-bFGF appeared to be trypsin-resistant when bound to nuclear chromatin or to isolated eukaryote DNA. Thus, our data indicate that: i) both 18 kD-bFGF and HMW-bFGFs localize into the nucleus of transfected NIH 3T3 cells and bind to nuclear chromatin; ii) the interaction of all bFGF isoforms with nuclear chromatin is mediated by one or more sequences present within the 18 kD form; iii) the chromatin-binding domain of HMW-bFGFs is distinct from their nuclear-targeting domain.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/metabolism , 3T3 Cells , Animals , Blotting, Western , DNA/genetics , Electrophoresis, Polyacrylamide Gel , Fibroblast Growth Factor 2/genetics , Mice , Molecular Weight , Peptide Fragments/metabolism , Transfection , Trypsin/metabolism
14.
Growth Factors ; 9(4): 269-78, 1993.
Article in English | MEDLINE | ID: mdl-8148156

ABSTRACT

Residues 27-31 (Lys-Asp-Pro-Lys-Arg) of the 155-amino acid form of basic fibroblast growth factor (bFGF) are in good agreement with a consensus sequence for nuclear translocation. To evaluate the role of this sequence in mediating the intracellular localization and biological activity of bFGF, basic residues Lys-27, Lys-30, and Arg-31 were changed to neutral glutamine residues by site-directed mutagenesis of the human bFGF cDNA. The bFGF mutant (M1Q-bFGF) was expressed in eukaryotic cells and in prokaryotic cells, from which it was purified to homogeneity. Transient expression of bFGF cDNA and of M1Q-bFGF cDNA in simian COS-1 cells followed by immunolocalization and by subcellular fractionation indicated that both molecules localize in the nucleus, as well as in the cytoplasm of transfected cells, and interact with nuclear chromatin and with eukaryote DNA in a similar manner. Prokaryotic expression of M1Q-bFGF cDNA yields a polypeptide endowed with a receptor-binding capacity and mitogenic activity similar to that exerted by wild-type bFGF. However, recombinant M1Q-bFGF showed a drastically reduced capacity to induce the production of urokinase-type plasminogen activator (uPA) in endothelial cells. The uPA-inducing activity of M1Q-bFGF was fully restored by the presence of soluble heparin in the culture medium. In conclusion, the sequence bFGF(27-31) does not appear to represent a nuclear translocation and/or retention sequence for bFGF. However, neutralization of its basic residues seems to modify the tertiary structure of the growth factor, thus affecting some of its biological properties.


Subject(s)
Cell Nucleus/metabolism , Fibroblast Growth Factor 2/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Biological Transport , Cattle , Cell Line , Cloning, Molecular , Consensus Sequence , Fibroblast Growth Factor 2/genetics , Humans , Immunoblotting , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...