Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 114(Pt 21): 3905-14, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11719557

ABSTRACT

The type-1 inhibitor of plasminogen activator (PAI-1) is an important physiological regulator of extracellular matrix (ECM) homeostasis and cell motility. Various growth factors mediate temporal changes in the expression and/or focalization of PAI-1 and its protease target PAs, thereby influencing cell migration by barrier proteolysis and/or ECM adhesion modulation. TGF-beta1, in particular, is an effective inducer of matrix deposition/turnover, cell locomotion and PAI-1 expression. Therefore, the relationship between motility and PAI-1 induction was assessed in TGF-beta1-sensitive T2 renal epithelial cells. PAI-1 synthesis and its matrix deposition in response to TGF-beta1 correlated with a significant increase in cell motility. PAI-1 expression was an important aspect in cellular movement as PAI-1-deficient cells had significantly impaired basal locomotion and were unresponsive to TGF-beta1. However, the induced migratory response to this growth factor was complex. TGF-beta1 concentrations of 1-2 ng/ml were significantly promigratory, whereas lower levels (0.2-0.6 ng/ml) were ineffective and final concentrations > or =5 ng/ml inhibited T2 cell motility. This same growth factor range progressively increased PAI-1 transcript levels in T2 cells consistent with a bifunctional role for PAI-1 in cell migration. TGF-beta1 induced PAI-1 mRNA transcripts in quiescent T2 cells via an immediate-early response mechanism. Full TGF-beta1-stimulated expression required tyrosine kinase activity and involved MAPK/ERK kinase (MEK). MEK appeared to be a major mediator of TGF-beta1-dependent PAI-1 expression and T2 cell motility since PD98059 effectively attenuated both TGF-beta1-induced ERK1/2 activation and PAI-1 transcription as well as basal and growth factor-stimulated planar migration. Since MEK activation in response to growth factors is adhesion-dependent, it was important to determine whether cellular adhesive state influenced TGF-beta1-mediated PAI-1 expression in the T2 cell system. Cells maintained in suspension culture (i.e., over agarose underlays) in growth factor-free medium or treated with TGF-beta1 in suspension expressed relatively low levels of PAI-1 transcripts compared with the significant induction of PAI-1 mRNA evident in T2 cells upon stimulation with TGF-beta1 during adhesion to a fibronectin-coated substrate. Attachment to fibronectin alone (i.e., in the absence of added growth factor) was sufficient to initiate PAI-1 transcription, albeit at levels considerably lower than that induced by the combination of cell adhesion in the presence of TGF-beta1. T2 cells allowed to attach to vitronectin-coated surfaces also expressed PAI-1 transcripts but to a significantly reduced extent relative to cells adherent to fibronectin. Moreover, newly vitronectin-attached cells did not exhibit a PAI-1 inductive response to TGF-beta1, at least during the short 2 hour period of combined treatment. PAI-1 mRNA synthesis in response to substrate attachment, like TGF-beta1-mediated induction in adherent cultures, also required MEK activity as fibronectin-stimulated PAI-1 expression was effectively attenuated by the MEK inhibitor PD98059. These data indicate that cellular adhesive state modulates TGF-beta1 signaling to particular target genes (i.e., PAI-1) and that MEK is a critical mediator of the PAI-1(+)/promigratory phenotype switch induced by TGF-beta1 in T2 cells.


Subject(s)
Fibronectins/metabolism , Gene Expression/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Plasminogen Activator Inhibitor 1/genetics , Transforming Growth Factor beta/pharmacology , Vitronectin/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Mitogen-Activated Protein Kinase 3 , Transforming Growth Factor beta1
2.
Cell Adhes Commun ; 7(6): 477-90, 2000.
Article in English | MEDLINE | ID: mdl-11051458

ABSTRACT

Previous studies have shown that the adhesion protein, vitronectin, directs the localization of urokinase-type plasminogen activator (uPA) to areas of cell-substrate adhesion, where uPA is thought to regulate cell migration as well as pericellular proteolysis. In the present study, HT-1080 cell lines expressing either wild-type vitronectin or vitronectin containing a single amino-acid substitution in the integrin binding domain were used to assess whether ligation of the alphavbeta5 integrin was required for uPA localization to focal adhesions. The synthesis of wild-type vitronectin by HT-1080 cells adherent to either collagen or fibronectin resulted in the redistribution of both the alphavbeta5 integrin as well as uPA to focal adhesion structures. In contrast, cells synthesizing mutant vitronectin, containing the amino-acid substitution in the integrin binding domain, were unable to direct the redistribution of either alphavbeta5 or uPA to focal adhesions. Recombinant forms of wild-type and mutant vitronectin were prepared in a baculovirus system and compared for their ability to direct the redistribution of vitronectin integrin receptors as well as uPA on human skin fibroblasts. In the absence of vitronectin, fibroblast cells adherent to fibronectin assemble focal adhesions which contain the beta1 integrin but do not contain uPA. Addition of recombinant wild-type, but not mutant, vitronectin to fibroblasts adherent to fibronectin resulted in the redistribution of alphavbeta3, alphavbeta5, and uPA into focal adhesions. However, when cells were plated directly onto antibodies directed against either the alphavbeta3 or alphavbeta5 integrins, uPA was not localized on the cell surface. These data indicate that ligation of vitronectin integrin receptors is necessary but not sufficient for the localization of uPA to areas of cell matrix adhesion, and suggest that vitronectin may promote cell migration by recruiting vitronectin integrin receptors and components of the plasminogen activator system to areas of cell matrix contact.


Subject(s)
Focal Adhesions/chemistry , Focal Adhesions/enzymology , Receptors, Vitronectin/metabolism , Urokinase-Type Plasminogen Activator/analysis , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosarcoma , Gene Expression/physiology , Humans , Integrins/analysis , Integrins/metabolism , Ligation , Oligopeptides/metabolism , Receptors, Vitronectin/analysis , Receptors, Vitronectin/genetics , Skin/cytology , Transfection , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism , Vitronectin/blood , Vitronectin/pharmacology
3.
Am J Physiol ; 277(6): G1189-99, 1999 12.
Article in English | MEDLINE | ID: mdl-10600816

ABSTRACT

Fibronectin (Fn) is a major adhesive protein found in the hepatic extracellular matrix (ECM). In adult rats, the in vivo turnover of plasma Fn (pFn) incorporated into the liver ECM is relatively rapid, i.e., <24 h, but the regulation of its turnover has not been defined. We previously reported that cellular Fn (cFn) and enzymatically desialylated plasma Fn (aFn), both of which have a high density of exposed terminal galactose residues, rapidly interact with hepatic asialoglycoprotein receptors (ASGP-R) in association with their plasma clearance after intravenous infusion. With the use of adult male rats (250-350 g) and measurement of the deoxycholate (DOC)-insoluble (125)I-labeled Fn in the liver, we determined whether the ASGP-R system can also influence the hepatic matrix retention of various forms of Fn. There was a rapid deposition of (125)I-pFn, (125)I-aFn, and (125)I-cFn into the liver ECM after their intravenous injection. Although (125)I-pFn was slowly lost from the liver matrix over 24 h, more than 90% of the incorporated (125)I-aFn and (125)I-cFn was cleared within 4 h (P < 0.01). Intravenous infusion of excess nonlabeled asialofetuin to competitively inhibit the hepatic ASGP-R delayed the rapid turnover of both aFn and cFn already incorporated within the ECM of the liver. ECM retention of both (125)I-aFn and (125)I-cFn was also less than (125)I-pFn (P < 0.01) as determined in vitro using liver slices preloaded in vivo with either tracer form of Fn. The hepatic ASGP-R appears to participate in the turnover of aFn and cFn within the liver ECM, whereas a non-ASGP-R-associated endocytic pathway apparently influences the removal of normal pFn incorporated within the hepatic ECM, unless it becomes locally desialylated.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibronectins/pharmacokinetics , Liver/metabolism , Receptors, Cell Surface/metabolism , Animals , Antirheumatic Agents/pharmacology , Asialoglycoprotein Receptor , Asialoglycoproteins/isolation & purification , Asialoglycoproteins/metabolism , Asialoglycoproteins/pharmacokinetics , Cell Fractionation , Chloroquine/pharmacology , Cross-Linking Reagents/metabolism , Cross-Linking Reagents/pharmacokinetics , Deoxycholic Acid , Detergents , Endocytosis/physiology , Fetuins , Fibroblasts/chemistry , Fibronectins/chemistry , Fibronectins/metabolism , Galactose/metabolism , Humans , Iodine Radioisotopes , Isomerism , Liver/chemistry , Liver/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , alpha-Fetoproteins/isolation & purification , alpha-Fetoproteins/metabolism , alpha-Fetoproteins/pharmacokinetics
4.
J Cell Sci ; 112 ( Pt 19): 3225-35, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10504328

ABSTRACT

Adhesion to the extracellular matrix modulates the cellular response to growth factors and is critical for cell cycle progression. The present study was designed to address the relationship between fibronectin matrix assembly and cell shape or shape dependent cellular processes. The binding of fibronectin's amino-terminal matrix assembly domain to adherent cells represents the initial step in the assembly of exogenous fibronectin into the extracellular matrix. When added to monolayers of pulmonary artery endothelial cells, the 70 kDa fragment of fibronectin (which contains the matrix assembly domain) stabilized both the extracellular fibronectin matrix as well as the actin cytoskeleton against cytochalasin D-mediated structural reorganization. This activity appeared to require specific fibronectin sequences as fibronectin fragments containing the cell adhesion domain as well as purified vitronectin were ineffective inhibitors of cytochalasin D-induced cytoarchitectural restructuring. Such pronounced morphologic consequences associated with exposure to the 70 kDa fragment suggested that this region of the fibronectin molecule may affect specific growth traits known to be influenced by cell shape. To assess this possibility, the 70 kDa fragment was added to scrape-wounded monolayers of bovine microvessel endothelium and the effects on two shape-dependent processes (i.e. migration and proliferation) were measured as a function of time after injury and location from the wound. The addition of amino-terminal fragments of fibronectin to the monolayer significantly inhibited (by >50%) wound closure. Staining of wounded monolayers with BrdU, moreover, indicated that either the 70 kDa or 25 kDa amino-terminal fragments of fibronectin, but not the 40 kDa collagen binding fragment, also inhibited cell cycle progression. These results suggest that the binding of fibronectin's amino-terminal region to endothelial cell layers inhibits cell cycle progression by stabilizing cell shape.


Subject(s)
Cell Cycle/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibronectins/metabolism , Actins/metabolism , Animals , Antimetabolites/pharmacology , Bromodeoxyuridine/pharmacology , Cattle , Cell Cycle/drug effects , Cell Movement/physiology , Cell Size/physiology , Cells, Cultured , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Fibronectins/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Polymers/metabolism , Protein Structure, Tertiary , Pulmonary Artery/cytology
5.
Am J Physiol ; 277(4): L749-59, 1999 10.
Article in English | MEDLINE | ID: mdl-10516216

ABSTRACT

Plasma fibronectin (pFN) can incorporate into the lung extracellular matrix (ECM) as well as enhance hepatic cell phagocytic removal of bloodborne microparticulate debris that can contribute to lung vascular injury. Treatment of human pFN (hFN) with N-ethylmaleimide (NEM) blocks its ECM incorporation but not its ability to augment phagocytosis. Using hFN purified from fresh human plasma cryoprecipitate, we compared the effect of NEM-treated hFN versus normal hFN on lung transvascular protein clearance (TVPC) in postoperative bacteremic sheep to determine whether the ability of hFN to attenuate the increase in lung endothelial permeability required its ECM incorporation. Sheep with lung lymph fistulas were infused with a sublethal dose of Pseudomonas aeruginosa (5 x 10(8)) 48 h after surgery. In the first study, sheep received either FN-rich human cryoprecipitate, FN-deficient cryoprecipitate, FN purified from cryoprecipitate (hFN), FN-deficient cryoprecipitate reconstituted with purified hFN, or the sterile saline diluent. In the second study, sheep received either 200 mg of purified hFN (group I), 200 mg of NEM-treated hFN (group II), or the saline diluent (group III). In the first study, the increase in TVPC after bacterial challenge was attenuated by FN-rich cryoprecipitate, hFN, or reconstituted FN-deficient cryoprecipitate (P < 0.05) but not by saline and FN-deficient cryoprecipitate. In the second study, TVPC increased by 2 h (P < 0.05) and peaked over 4-8 h (P < 0.05) at 380-420% above baseline in postoperative bacteremic sheep given the diluent (group III). In contrast, intravenous infusion of hFN, but not of NEM-treated hFN, significantly (P < 0.05) attenuated this increase of lung protein clearance. Thus the ability for the intravenously infused purified pFN to attenuate the increase in lung endothelial protein permeability in sheep during postsurgical bacteremia appears to require its ECM incorporation into the interstitial ECM of the lung.


Subject(s)
Bacteremia/physiopathology , Capillary Permeability/physiology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Lung/metabolism , Postoperative Complications , Animals , Bacteremia/metabolism , Blood Proteins/metabolism , Capillary Permeability/drug effects , Ethylmaleimide/pharmacology , Fibronectins/blood , Fibronectins/drug effects , Fibronectins/pharmacology , Humans , Infusions, Intravenous , Male , Pseudomonas Infections/metabolism , Pseudomonas Infections/physiopathology , Sheep
6.
J Biol Chem ; 274(38): 27257-64, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480945

ABSTRACT

The deposition of fibronectin into the extracellular matrix is an integrin-dependent, multistep process that is tightly regulated in order to ensure controlled matrix deposition. Reduced fibronectin deposition has been associated with altered embryonic development, tumor cell invasion, and abnormal wound repair. In one of the initial steps of fibronectin matrix assembly, the amino-terminal region of fibronectin binds to cell surface receptors, termed matrix assembly sites. The present study was undertaken to investigate the role of extracellular signals in the regulation of fibronectin deposition. Our data indicate that the interaction of cells with the extracellular glycoprotein, vitronectin, specifically inhibits matrix assembly site expression and fibronectin deposition. The region of vitronectin responsible for the inhibition of fibronectin deposition was localized to the heparin-binding domain. Vitronectin's heparin-binding domain inhibited both beta(1) and non-beta(1) integrin-dependent matrix assembly site expression and could be overcome by treatment of cells with lysophosphatidic acid, an agent that promotes actin polymerization. The interaction of cells with the heparin-binding domain of vitronectin resulted in changes in actin microfilament organization and the subcellular distribution of the actin-associated proteins alpha-actinin and talin. These data suggest a mechanism whereby the heparin-binding domain of vitronectin regulates the deposition of fibronectin into the extracellular matrix through alterations in the organization of the actin cytoskeleton.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Heparin/metabolism , Vitronectin/metabolism , Animals , Binding Sites , Cells, Cultured , Cytoskeleton/metabolism , Down-Regulation , Humans , Microscopy, Fluorescence , Rats
7.
Biochim Biophys Acta ; 1404(3): 353-66, 1998 Sep 16.
Article in English | MEDLINE | ID: mdl-9739164

ABSTRACT

The plasma protein vitronectin is thought to be an important regulator of extravascular plasminogen activation. In previous studies we have shown that a disulfide stabilized multimeric form of vitronectin is endocytosed and degraded by fibroblast cells (T.S. Panetti, P.J. McKeown-Longo, J. Biol. Chem. 268 (1993) 11988-11993; P.J. McKeown-Longo, T.S. Panetti, in: K.T. Preissner, S. Rosenblatt, C. Kost, J. Wegerhoff, D.F. Mosher (Eds.), Biology of Vitronectins and their Receptors, Elsevier Science Publishers, Amsterdam, 1993, pp. 111-118). The preparation of multimeric vitronectin used in these earlier studies was in the form of high molecular weight disulfide-bonded aggregates which were stable in sodium dodecyl sulfate (SDS). To address the question of whether vitronectin needed to be in the form of disulfide stabilized multimers in order to be endocytosed, a multimeric vitronectin, which was not disulfide stabilized, was prepared from vitronectin that had been treated with reducing agent and alkylated with iodoacetamide. The resulting protein migrated as a 65/75 kDa protein on SDS gels in the absence of reducing agent, confirming that this form of vitronectin was no longer stabilized into disulfide-bonded aggregates. However, the protein was still multimeric when analyzed by native gels and could be converted to SDS stable multimers by cross-linking agents. This result demonstrated that reduced and alkylated vitronectin aggregates into multimeric forms which are not stable in SDS. Similar to disulfide stabilized multimers, alkylated multimers of vitronectin bound to sulfated proteoglycans in the extracellular matrix and were endocytosed and degraded. Degradation of both forms of vitronectin was inhibited with arginine-glycine-aspartic acid peptides, an anti-alphavbeta5 antibody and heparin. Chloroquine and wortmannin were also able to inhibit degradation of both forms of vitronectin, indicating that both multimeric forms were following the same endocytic and degradative pathway. These results suggest that the organization of vitronectin into a multimeric form which will be recognized for endocytosis does not require disulfide bond stabilization. This study further suggests that recognition of vitronectin for endocytosis is dependent upon its conversion from a monomeric to a multivalent form (C.E. Wilkins-Port, P.J. McKeown-Longo, Mol. Biol. Cell 8:S:64A (1997).


Subject(s)
Fibroblasts/metabolism , Receptors, Vitronectin , Vitronectin/metabolism , Alkylation , Androstadienes/pharmacology , Ascorbic Acid/pharmacology , Cells, Cultured , Chloroquine/pharmacology , Endocytosis , Heparin/pharmacology , Humans , Integrins/metabolism , Lysosomes/metabolism , Oligopeptides/pharmacology , Proteoglycans/metabolism , Vitronectin/analysis , Vitronectin/chemistry , Wortmannin
8.
Hepatology ; 28(2): 475-85, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9696014

ABSTRACT

It has been postulated that the in vivo removal of many plasma glycoproteins after desialylation is mediated by their interaction with a specific endocytic receptor on hepatocytes called the asialoglycoprotein receptor (ASGP-R), which is known to have a high affinity for specific carbohydrate residues, such as galactose. However, this mechanism has never been proven in vivo, nor has a naturally occurring ligand for the ASGP-R been identified. We investigated the influence of the terminal galactose residues on plasma fibronectin (pFn) on its liver deposition and turnover in adult rats, using neuraminidase to remove sialic acid residues to expose galactose residues. We also tested the hypothesis that the normal presence of a large amount of terminal galactose residues in cellular Fn (cFn) may allow cFn to serve as a natural ligand readily able to interact with the ASGP-R. In contrast to the slow clearance of normal pFn from the blood, cFn and desialylated pFn (aFn) displayed a rapid plasma clearance (P < .001) with greater than 50% of both the 125I-cFn or 125I-aFn depositing in the liver within 15 minutes. The enhanced plasma removal and liver deposition of both 125I-cFn and 125I-aFn was competitively inhibited (P < .01) by prior intravenous infusion of excess asialofetuin, which can selectively bind to the ASGP-R. The enzymatic addition of terminal sialic acid residues onto cFn to "mask" or "cap" the normally exposed galactose residues delayed the rapid plasma removal of cFn. Accelerated degradation of 125I-aFn and 125I-cFn as compared with 125I-pFn was demonstrated in vitro by both primary cultures of normal rat hepatocytes or incubated (37 degrees C) tissue slices of livers harvested from normal rats after in vivo preloading with tracer 125I-Fn forms. Thus, the ASGP-R appears to directly participate in the rapid in vivo removal of cFn from the blood, while native pFn may be removed by an alternative pathway unless it can become desialylated in vivo. These findings suggest that cFn may be a naturally occurring ligand that does not require desialylation before removal by the ASGP-R on hepatocytes.


Subject(s)
Fibronectins/blood , Liver/metabolism , Receptors, Cell Surface/metabolism , Animals , Asialoglycoprotein Receptor , Asialoglycoproteins/metabolism , Bile/metabolism , Binding, Competitive , Fetuins , Fibronectins/metabolism , Fibronectins/pharmacokinetics , Humans , In Vitro Techniques , Ligands , Rats , alpha-Fetoproteins/metabolism
9.
Am J Physiol ; 275(1): L126-38, 1998 07.
Article in English | MEDLINE | ID: mdl-9688944

ABSTRACT

Exposure of confluent pulmonary arterial endothelial monolayers to tumor necrosis factor (TNF)-alpha causes both a reorganization and/or disruption of fibronectin (Fn) in the extracellular matrix and an increase in transendothelial protein permeability. However, the factors initiating this response to TNF-alpha have not been defined. Because TNF-alpha can induce proteinase expression in endothelial cells, we determined whether proteinases cause both the alteration of the Fn matrix and the permeability increase as is often speculated. Incubation of calf pulmonary arterial endothelial monolayers with TNF-alpha (200 U/ml) for 18 h caused a disruption of the Fn matrix and an increase in transendothelial protein permeability. A reduced colocalization of cell-surface alpha5beta1-Fn integrins with the Fn fibers in focal contacts was also observed. TNF-alpha treatment of endothelial monolayers with matrices prelabeled with 125I-human Fn (hFn) did not cause the release of Fn fragments or alter the content of Fn antigen in the medium as analyzed by SDS-PAGE coupled with autoradiography. Both the content and fragmentation pattern of Fn within the cell layer and the insoluble Fn matrix also appeared unchanged after TNF-alpha exposure as confirmed by Western immunoblot. Fn-substrate zymography revealed that TNF-alpha increased the expression of two proteinases within the conditioned medium in which activity could be blocked by aprotinin but not by EDTA, 1,10-phenanthroline, leupeptin, or pepstatin. However, inhibition of the Fn proteolytic activity of these two serine proteinases did not prevent either the TNF-alpha-induced disruption of the Fn matrix or the increase in permeability. Thus the reorganization and/or disruption of the Fn matrix and the temporally associated increase in endothelial permeability caused by TNF-alpha appear not to be due to proteolytic degradation of Fn within the extracellular matrix. In contrast, decreased alpha5beta1-Fn integrin interaction with Fn fibers in the matrix may be important in the response to TNF-alpha exposure.


Subject(s)
Endothelium, Vascular/cytology , Fibronectins/drug effects , Receptors, Fibronectin/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Aprotinin/pharmacology , Cattle , Cell Membrane Permeability/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Fibronectins/metabolism , Humans , Iodine Radioisotopes , Pulmonary Artery , Receptors, Fibronectin/drug effects
10.
J Cell Biol ; 141(1): 241-53, 1998 Apr 06.
Article in English | MEDLINE | ID: mdl-9531562

ABSTRACT

The interaction of cells with fibronectin generates a series of complex signaling events that serve to regulate several aspects of cell behavior, including growth, differentiation, adhesion, and motility. The formation of a fibronectin matrix is a dynamic, cell-mediated process that involves both ligation of the alpha5beta1 integrin with the Arg-Gly-Asp (RGD) sequence in fibronectin and binding of the amino terminus of fibronectin to cell surface receptors, termed "matrix assembly sites," which mediate the assembly of soluble fibronectin into insoluble fibrils. Our data demonstrate that the amino-terminal type I repeats of fibronectin bind to the alpha5beta1 integrin and support cell adhesion. Furthermore, the amino terminus of fibronectin modulates actin assembly, focal contact formation, tyrosine kinase activity, and cell migration. Amino-terminal fibronectin fragments and RGD peptides were able to cross-compete for binding to the alpha5beta1 integrin, suggesting that these two domains of fibronectin cannot bind to the alpha5beta1 integrin simultaneously. Cell adhesion to the amino-terminal domain of fibronectin was enhanced by cytochalasin D, suggesting that the ligand specificity of the alpha5beta1 integrin is regulated by the cytoskeleton. These data suggest a new paradigm for integrin-mediated signaling, where distinct regions within one ligand can modulate outside-in signaling through the same integrin.


Subject(s)
Cell Adhesion/physiology , Extracellular Matrix/physiology , Fibronectins/physiology , Receptors, Fibronectin/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement , Cycloheximide/pharmacology , Cytochalasin D/pharmacology , Cytoskeletal Proteins/metabolism , Extracellular Matrix/chemistry , Fibroblasts/cytology , Fibroblasts/physiology , Fibronectins/chemistry , Fibronectins/isolation & purification , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/physiology , Kinetics , Male , Oligopeptides , Paxillin , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Skin
11.
J Cell Sci ; 111 ( Pt 4): 425-33, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9443892

ABSTRACT

Endocytosis and degradation of vitronectin by human skin fibroblasts are regulated by the beta5 integrin. To determine whether the beta5 integrin is directly mediating the internalization of vitronectin, both vitronectin and the beta5 integrin were localized by indirect immunofluorescence during the endocytic process. This analysis showed that both vitronectin and beta5 were found in intracellular vesicles within 5 minutes of the addition of exogenous vitronectin to fibroblast cell layers. By 15 minutes, approximately 20% of the vitronectin-containing vesicles stained positively for beta5. In contrast, the beta3 integrin was not found in any intracellular vesicles. Within 30 minutes, more than 50% of vitronectin-containing vesicles also stained for lamp-1, indicating that internalized vitronectin traveled to lysosomes. Inhibition of clathrin assembly by either potassium depletion or hypertonic buffer inhibited vitronectin internalization, suggesting that vitronectin internalization occurred through coated pits. Confocal analysis confirmed the colocalization of vitronectin and alphavbeta5 in intracellular compartments and further demonstrated that the highest colocalization of the two proteins occurred within 1.8 microm from the ventral surface of the cell, suggesting endocytosis occurred at the substrate level. Pretreatment of cells with the PI-3 kinase inhibitor, wortmannin, resulted in a marked increase in the coincidence of vitronectin and beta5 staining within vesicles and prevented the accumulation of vitronectin within lysosomes. This suggests that following internalization, vitronectin and the alphavbeta5 integrin are segregated to different cellular compartments. This study provides the first evidence that the alphavbeta5 vitronectin receptor directly mediates the internalization of vitronectin.


Subject(s)
Endocytosis/physiology , Integrin beta Chains , Integrins/analysis , Receptors, Vitronectin/analysis , Androstadienes/pharmacology , Antigens, CD/analysis , Cell Adhesion , Cells, Cultured , Coated Pits, Cell-Membrane , Endosomes/chemistry , Enzyme Inhibitors/pharmacology , Fibroblasts , Humans , Integrin beta3 , Lysosomal Membrane Proteins , Lysosomes/chemistry , Membrane Glycoproteins/analysis , Phosphoinositide-3 Kinase Inhibitors , Platelet Membrane Glycoproteins/analysis , Potassium/physiology , Skin , Vitronectin/analysis , Vitronectin/metabolism , Wortmannin
12.
J Cell Sci ; 110 ( Pt 5): 569-81, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9092939

ABSTRACT

Polymerization of soluble fibronectin into extracellular matrix fibers occurs through the interaction between the amino terminus of fibronectin contained within a 70 kDa fragment and 'matrix assembly sites' on the cell surface. The present studies were performed to localize the 'matrix assembly sites' (defined by 70 kDa binding sites) on newly adherent cells and on cells containing preformed fibronectin matrix. Matrix nucleation sites on newly spread cells were visualized using Texas Red conjugated 70 kDa fragment and were found to colocalize with vinculin and substrate fibronectin fibrils. Cells plated onto vitronectin coated coverslips did not exhibit any 70 kDa binding sites although these cells were well-spread with fully developed focal adhesions. Time course studies indicated that 70 kDa binding sites could be detected on newly adherent cells within 30-40 minutes following cell plating onto fibronectin coated coverslips, prior to the reorganization of substrate fibronectin into fibrils. Similarly, exogenous fibronectin conjugated with Texas Red was also colocalized with vinculin when added to newly adherent cells. The disruption of actin filaments with cytochalasin D both prevented the expression of 70 kDa binding sites and also resulted in the loss of established 70 kDa binding sites on newly spread cells. After 3 days in culture, cells organized an extensive fibronectin matrix and 70 kDa was colocalized with two distinct types of matrix fibronectin fibers: fine linear cell-associated fibers which co-stained with the beta1 integrin and coarse extracellular fibers which did not stain for the beta1 integrin. There was also a third type of fibronectin fiber which was organized into a meshwork structure. There was no localization of either beta1 or 70 kDa to these structures. Treatment of 3-day cells with cytochalasin D resulted in the disruption of cell-matrix fibers and cell-associated 70 kDa binding sites. In contrast, the coarse extracellular matrix fibers as well as the meshwork fibers were unaffected by cytochalasin. In the presence of cytochalasin D, 70 kDa bound to sites which colocalized with the coarse extracellular matrix fibers. These data suggest that de novo assembly of fibronectin matrix occurs at sites of focal adhesion and as fibronectin polymerization proceeds, matrix nucleation sites colocalize along cell associated fibronectin fibers. At later times 70 kDa is localized to a subset of more mature fibronectin-containing fibers. These results suggest that there are at least three morphologically distinct 70 kDa binding sites on adherent cells: one which colocalizes with beta1 to focal adhesions, a second which colocalizes with beta1 and fibronectin in matrix contacts, and a third which localizes to extracellular matrix fibers.


Subject(s)
Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Actins/metabolism , Binding Sites , Cell Adhesion , Cells, Cultured , Endothelium, Vascular/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Humans
13.
J Cell Biol ; 133(2): 431-44, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8609174

ABSTRACT

Fibronectin matrix assembly is a cell-dependent process which is upregulated in tissues at various times during development and wound repair to support the functions of cell adhesion, migration, and differentiation. Previous studies have demonstrated that the alpha 5 beta 1 integrin and fibronectin's amino terminus and III-1 module are important in fibronectin polymerization. We have recently shown that fibronectin's III-1 module contains a conformationally sensitive binding site for fibronectin's amino terminus (Hocking, D.C., J. Sottile, and P.J. McKeown-Longo. 1994. J. Biol. Chem. 269: 19183-19191). The present study was undertaken to define the relationship between the alpha 5 beta 1 integrin and fibronectin polymerization. Solid phase binding assays using recombinant III-10 and III-1 modules of human plasma fibronectin indicated that the III-10 module contains a conformation-dependent binding site for the III-1 module of fibronectin. Unfolded III-10 could support the formation of a ternary complex containing both III-1 and the amino-terminal 70-kD fragment, suggesting that the III-1 module can support the simultaneous binding of III-10 and 70 kD. Both unfolded III-10 and unfolded III-1 could support fibronectin binding, but only III-10 could promote the formation of disulfide-bonded multimers of fibronectin in the absence of cells. III-10-dependent multimer formation was inhibited by both the anti-III-1 monoclonal antibody, 9D2, and amino-terminal fragments of fibronectin. A fragment of III-10, termed III-10/A, was able to block matrix assembly in fibroblast monolayers. Similar results were obtained using the III-10A/RGE fragment, in which the RGD site had been mutated to RGE, indicating that III-I0/A was blocking matrix assembly by a mechanism distinct from disruption of integrin binding. Texas red-conjugated recombinant III-1,2 localized to beta 1-containing sites of focal adhesions on cells plated on fibronectin or the III-9,10 modules of fibronectin. Monoclonal antibodies against the III-1 or the III-9,10 modules of fibronectin blocked binding of III-1,2 to cells without disrupting focal adhesions. These data suggest that a role of the alpha 5 beta 1 integrin in matrix assembly is to regulate a series of sequential self-interactions which result in the polymerization of fibronectin.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Receptors, Fibronectin/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Adhesion , Cell Line , Cell Membrane/chemistry , Fibroblasts , Fibronectins/analysis , Fibronectins/chemistry , Hot Temperature , Humans , Molecular Sequence Data , Molecular Weight , Polymers , Protein Binding , Protein Conformation , Protein Denaturation , Receptors, Fibronectin/analysis , Recombinant Fusion Proteins/metabolism
14.
Biochem Cell Biol ; 74(6): 887-97, 1996.
Article in English | MEDLINE | ID: mdl-9164657

ABSTRACT

Vitronectin, a 75-kDa plasma protein is also found in the extracellular matrix, where it is believed to promote cell adhesion and migration. In addition to its role in adhesion, matrix vitronectin is also believed to function as an opsonin promoting the clearance of thrombin-serpin complexes from the matrix. Vitronectin is cleared from the matrix by receptor-mediated endocytosis followed by lysosomal degradation, suggesting that cells can regulate the levels of vitronectin present in the matrix. However, the mechanism by which plasma vitronectin associates with the extracellular matrix remains unclear. Studies were conducted to define the binding site(s) for vitronectin in fibroblast cell layers. Sodium chlorate, a competitive inhibitor of proteoglycan sulfation, produced a dose-dependent decrease in both binding and degradation of vitronectin. This inhibition was reversible in that removal of chlorate returned both binding and degradation of vitronectin to near control levels within 24 h. The binding of vitronectin to cell layers was not dependent on cells because vitronectin bound directly to isolated matrix. Isolated matrices prepared from cell layers treated with sodium chlorate also exhibited a dose-dependent decrease in vitronectin binding, consistent with the binding site for vitronectin in the matrix being sulfated proteoglycans. Binding and degradation of vitronectin were also sensitive to the addition of exogenous heparin, suggesting that the heparin binding domain of vitronectin was mediating binding to the matrix. Incubating fibroblast monolayers with heparinase III resulted in a 40% decrease in binding and degradation of vitronectin. Taken together, the above findings suggest that vitronectin's binding to the matrix and its subsequent degradation are dependent on heparan sulfate proteoglycans.


Subject(s)
Fibroblasts/metabolism , Heparitin Sulfate/physiology , Proteoglycans/physiology , Vitronectin/metabolism , Binding Sites , Cells, Cultured , Chlorates/pharmacology , Extracellular Matrix , Heparan Sulfate Proteoglycans , Heparin/metabolism , Heparitin Sulfate/pharmacology , Humans , Male
15.
Biochem Cell Biol ; 74(6): 899-910, 1996.
Article in English | MEDLINE | ID: mdl-9164658

ABSTRACT

Cell surface plasminogen activators have been proposed to participate in cell migration and invasion by activating both intracellular signaling pathways and extracellular proteolysis. Urokinase-type plasminogen activator (uPA) is secreted from many cell types and localizes to focal contact areas when cells are seeded onto the plasma protein vitronectin. Induction of vitronectin synthesis during migration of neural crest cells and growth of certain tumors suggests that the de novo synthesis and deposition of vitronectin into the tissue matrix may remodel the matrix to provide an environment suitable for cell migration and (or) tumor invasion. To investigate the effects of vitronectin secretion and matrix deposition on the localization and activity of cell-associated uPA, HT-1080 fibrosarcoma cells were transfected with the Rc/CMV expression vector containing a vitronectin cDNA insert and stable cell lines expressing vitronectin were selected. Vitronectin-secreting cells were allowed to attach and spread on collagen- and fibronectin-coated substrates. Within 6 h, vitronectin was detected on the substrate; vitronectin synthesis was accompanied by the clustering of both the alpha v beta 5 vitronectin receptor and uPA into vinculin-containing focal adhesions. Although mock transfected cells formed small focal adhesions on both collagen and fibronectin, no co-localization of uPA or alpha v beta 5 to focal adhesions was evident in these cells. Vitronectin-secreting cells also exhibited decreased levels of plasminogen activation and increased levels of cell adhesion as compared with the mock transfected cells. These data demonstrate that the synthesis of vitronectin and its matrix association by transfected HT-1080 fibrosarcoma cells results in localization of uPA to alpha v beta 5 containing focal adhesions, decreased cell surface uPA activity, and an increase in cell adhesion.


Subject(s)
Cell Adhesion , Fibrosarcoma/metabolism , Urokinase-Type Plasminogen Activator/analysis , Vitronectin/biosynthesis , Extracellular Matrix/metabolism , Fibrinolysin/metabolism , Humans , Microscopy, Fluorescence , Protein Conformation , Receptors, Cell Surface/analysis , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/biosynthesis , Transfection , Tumor Cells, Cultured , Vitronectin/genetics , Vitronectin/metabolism
16.
Am J Physiol ; 269(6 Pt 1): G902-12, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8572222

ABSTRACT

The incorporation of plasma fibronectin (pFn) into the extracellular matrix (ECM) is believed to influence tissue integrity, wound repair, and vascular permeability. In vitro, matrix assembly of Fn requires the binding of soluble Fn to cell-associated matrix assembly sites. Alkylation of human pFn (HFn) with N-ethylmaleimide (NEM) prevents the initial binding of Fn to matrix assembly sites as well as its in vitro incorporation into the ECM as reflected by detergent-insoluble 125I-labeled Fn (pool II Fn). We determined the kinetics of Fn matrix incorporation in tissue and whether NEM treatment of rat pFn (NEM-RFn) would limit its in vivo incorporation into ECM by analysis of pool I [deoxycholate (DOC) soluble] and pool II (DOC insoluble) 125I-Fn in tissues after its intravenous injection into rats. After intravenous injection, tissue incorporation of normal rat 125I-pFn was especially intense in liver and spleen, in agreement with the large amount of endogenous Fn detected in the matrices of these organs. Tissue deposition of plasma-derived 125I-RFn in liver and spleen peaked by 4 h, with significant (P < 0.01) loss over 24 h, indicating turnover of matrix Fn. Tissue localization of normal 125I-RFn in liver, lung, spleen, heart, and intestine was greater (P < 0.05) than 125I-NEM-RFn at 4 h. Normal HFn, but not NEM-HFn, was incorporated into tissues and colocalized with endogenous Fn in the matrix. To identify the cells mediating the intense incorporation of pFn into liver ECM, we compared matrix assembly of 125I-HFn by cultured fibroblasts, hepatocytes, and hepatic Kupffer cells. With fibroblasts, 125I-HFn in pool I reached steady state by 3 h, whereas 125I-HFn in pool II exceeded that in pool I by 6 h and continued to increase over 24 h. With hepatocytes, pool I 125I-HFn reached steady state by 1 h, and a progressive increase (P < 0.05) of 125I-HFn in pool II was observed over 24 h. Kupffer cells were not able to incorporate significant amounts of 125I-HFn into matrix. NEM-HFn displayed limited incorporation into ECM by both fibroblast and hepatocyte cultures. These novel observations suggest that the interaction of soluble pFn with matrix assembly sites is necessary to its in vivo incorporation into the ECM.


Subject(s)
Ethylmaleimide/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Liver/metabolism , Spleen/metabolism , Alkylation , Animals , Cells, Cultured , Fibroblasts/metabolism , Humans , Kupffer Cells/metabolism , Liver/cytology , Rats , Reference Values , Time Factors , Tissue Distribution
17.
Circ Res ; 77(4): 651-9, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7554109

ABSTRACT

Complement-mediated pulmonary edema results from increases in lung capillary hydraulic conductivity (Lp), possibly by receptor-mediated mechanisms. We considered the Lp effects of vitronectin and the vitronectin-containing complement complex SC5b-9, which ligate the integrin alpha v beta 3. Vitronectin, SC5b-9, and SC5b-9-enriched zymosan-activated serum all rapidly increased Lp, as determined by the split-drop technique in single lung capillaries of rat lung. The Lp increases were inhibited by a monospecific (LM609) and a polyclonal (R838) antibody against the alpha v beta 3 integrin but not by an irrelevant monoclonal antibody isotype matched with LM609, by a monoclonal antibody against the alpha v beta 5 integrin, or by preimmune rabbit serum. Vitronectin monomers failed to increase Lp. The tyrosine kinase blockers genistein and methyl 2,5-dihydroxycinnamate caused significant concentration-dependent inhibitions of Lp increases due to vitronectin and zymosan-activated serum. By contrast, the protein kinase C blocker calphostin C had no major effect. We conclude that (1) multivalent ligation of the luminally located alpha v beta 3 integrin of lung capillary endothelium increases transcapillary liquid flux, and (2) the dominant signal transduction pathway for this effect occurs through tyrosine kinase activation.


Subject(s)
Endothelium, Vascular/physiology , Lung/blood supply , Microcirculation/physiology , Receptors, Vitronectin/physiology , Animals , Capillary Permeability , Complement Membrane Attack Complex , Complement System Proteins/pharmacology , Glycoproteins/pharmacology , Rabbits , Rats , Vitronectin/pharmacokinetics , Vitronectin/physiology
18.
Am J Physiol ; 269(2 Pt 1): L248-60, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7544537

ABSTRACT

Endothelial permeability can be altered by tumor necrosis factor-alpha (TNF-alpha), a cytokine released in association with inflammation-induced tissue injury. In the subendothelial matrix, fibronectin (Fn) influences endothelial cell adhesion by the interaction of integrins with RGD and non-RGD attachment sites in Fn. We compared the effect of TNF-alpha, RGD-containing peptides (GRGDSP), or antibody to alpha 5 beta 1-integrins on the protein permeability of bovine lung endothelial monolayers as assessed by transendothelial 125I-labeled albumin clearance. We also examined the influence of purified human plasma fibronectin (hFn) on this permeability response. TNF-alpha, RGD peptides, and antibodies to alpha 5 beta 1-integrins elicited a dose- and time-dependent increase in protein permeability as well as a reorganization and/or disruption of the endogenous Fn matrix. A control RGE peptide (GRGESP) as well as immunoglobulin G purified from nonimmune rabbit serum did not increase endothelial protein permeability or disrupt the endogenous fibrillar Fn pattern in the matrix. Likewise, a LDV peptide derived from the alternatively spliced type III connecting segment (IIICS) within bovine Fn (bFn) was unable to increase permeability of the bovine endothelial monolayer. Co-incubation of purified soluble hFn (300 or 600 micrograms/ml) with either TNF-alpha, the RGD peptide, or the antibody to alpha 5 beta 1-integrins prevented the increase in endothelial permeability. This protective effect was also observed when the purified hFn (600 micrograms/ml) was added after the TNF-alpha-induced increase in endothelial permeability had taken place. Immunofluorescent analysis confirmed the incorporation of the hFn into the subendothelial matrix and its co-localization with the endogenous bFn. The similar alteration of the subendothelial matrix after exposure to RGD peptides, anti-alpha 5 beta 1-antibodies, or TNF-alpha, coupled with the ability for hFn to attenuate the permeability increase typically elicited by all three agents, suggests that disruption of cell-matrix interactions may be the mechanism by which TNF-alpha alters endothelial permeability.


Subject(s)
Capillary Permeability/drug effects , Endothelium, Vascular/metabolism , Fibronectins/pharmacology , Integrins/antagonists & inhibitors , Oligopeptides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies/immunology , Cattle , Endothelium, Vascular/cytology , Humans , Integrins/immunology , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Receptors, Fibronectin , Serum Albumin/pharmacokinetics
19.
J Biol Chem ; 270(31): 18593-7, 1995 Aug 04.
Article in English | MEDLINE | ID: mdl-7543105

ABSTRACT

Previous studies have demonstrated that the alpha v beta 5 integrin receptor functions in the endocytosis and degradation of matrix-bound vitronectin by human skin fibroblasts (Panetti, T. S., and McKeown-Longo, P. J. (1993) J. Biol. Chem. 268, 11988-11993; Panetti, T. S., and McKeown-Longo, P. J. (1993) J. Biol. Chem. 268, 11492-11495). These earlier studies demonstrated that vitronectin degradation was inhibited by either antibodies to the beta 5 integrin or exogenous heparin, suggesting that both integrin receptors and cell surface heparan sulfate proteoglycans are involved in the endocytosis and degradation of vitronectin. The present study was done to define intracellular signaling pathways involved in endocytosis of vitronectin and to evaluate the relative contribution of cell surface heparan sulfate proteoglycans and the alpha v beta 5 integrin in the activation of these signaling pathways. The addition of the phorbol ester phorbol 12-myristate 13-acetate (PMA), a protein kinase C activator, to monolayers of human skin fibroblasts, increased vitronectin degradation. Staurosporine and calphostin C, inhibitors of protein kinase C, blocked internalization and subsequent degradation of vitronectin, while KT5720, an inhibitor of protein kinase A, had no effect on the degradation of vitronectin. PMA was also able to reverse the inhibition of vitronectin degradation seen when cells were pretreated with heparinase or incubated with exogenous heparin. In contrast, the inhibitory effect of either RGD peptides or anti-alpha v beta 5 antibodies on vitronectin degradation were not overcome by the addition of PMA. These data suggest that the internalization of vitronectin from the matrix is mediated by the alpha v beta 5 integrin following activation of protein kinase C.


Subject(s)
Endocytosis/physiology , Glycoproteins/metabolism , Integrins/metabolism , Protein Kinase C/metabolism , Receptors, Vitronectin , Signal Transduction , Biological Transport , Cells, Cultured , Fibroblasts/cytology , Heparan Sulfate Proteoglycans , Heparitin Sulfate/metabolism , Humans , Naphthalenes/pharmacology , Oligopeptides/pharmacology , Proteoglycans/metabolism , Skin Physiological Phenomena , Tetradecanoylphorbol Acetate/pharmacology , Vitronectin
20.
J Biol Chem ; 269(29): 19183-7, 1994 Jul 22.
Article in English | MEDLINE | ID: mdl-8034677

ABSTRACT

Cultured fibroblasts express binding sites for the amino-terminal region of fibronectin on their cell surface that mediate the assembly of soluble fibronectin into disulfide-stabilized fibrils. These binding sites have been termed matrix assembly sites and have been studied in binding assays using a 125I-labeled 70-kDa fragment derived from the amino terminus of fibronectin. In an attempt to isolate the protein(s) responsible for binding the 70-kDa fragment, cell surface proteins were cleaved from fibroblast monolayers by mild trypsinization. Trypsinization of monolayers generated a series of fibronectin fragments that bound the 125I-labeled 70-kDa fragment by ligand blot assay and affinity chromatography. All of the fibronectin fragments that bound the 70-kDa fragment contained the III-1 module. In solid phase binding assays, the 125I-labeled 70-kDa fragment bound preferentially to reduced fibronectin as compared with unreduced fibronectin fragments. Binding of the 125I-labeled 70-kDa fragment to reduced fibronectin was inhibited by a monoclonal antibody directed against the III-1 domain. Isolated III-1, however, did not bind the 125I-labeled 70-kDa fragment when adsorbed to plastic tissue culture wells. Heat denaturation of III-1 prior to adsorption conferred 70-kDa fragment binding properties on the isolated module. The 125I-labeled 70-kDa fragment did not bind to heat-denatured III-2, suggesting that 70-kDa fragment binding was a property of the III-1 module and not a general characteristic of all type III modules. The binding of 125I-labeled 70-kDa fragment to III-1 was of high affinity (KD = 1.8 x 10(-8) M). These results indicate that a binding site for the 70-kDa amino terminus of fibronectin is contained within a cryptic site found in the first type III module of fibronectin. Unfolding of the III-1 module on the cell surface may control matrix assembly site expression and represent an important step in the initiation of cell-dependent fibronectin polymerization.


Subject(s)
Fibronectins/metabolism , Base Sequence , Binding Sites , DNA Primers/chemistry , Extracellular Matrix/ultrastructure , Fibronectins/chemistry , Hot Temperature , Humans , Macromolecular Substances , Molecular Sequence Data , Oxidation-Reduction , Peptide Fragments/metabolism , Protein Conformation , Protein Denaturation
SELECTION OF CITATIONS
SEARCH DETAIL
...