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










Publication year range
1.
Cell Adh Migr ; 15(1): 215-223, 2021 12.
Article in English | MEDLINE | ID: mdl-34308743

ABSTRACT

We previously demonstrated that F4 peptide (CNPEDCLYPVSHAHQR) from collagen XIX was able to inhibit melanoma cell migrationin vitro and cancer progression in a mouse melanoma model. The aim of the present work was to study the anti-angiogenic properties of F4 peptide. We demonstrated that F4 peptide inhibited VEGF-induced pseudo-tube formation on Matrigel by endothelial cells and endothelial sprouting in a rat aortic ring assay. By affinity chromatography, we identified αvß3 and α5ß1 integrins as potential receptors for F4 peptide on endothelial cell surface. Using solid phase assays, we proved the direct interaction between F4 and both integrins. Taken together, our results demonstrate that F4 peptide is a potent antitumor agent inhibiting both angiogenesis and tumor cell migration.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Collagen/metabolism , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic/drug therapy , Peptide Fragments/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/pharmacology , Endothelial Cells/metabolism , Humans , Integrin alpha5beta1/drug effects , Integrin alphaVbeta3/drug effects , Neovascularization, Pathologic/pathology , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley
2.
J Med Chem ; 61(6): 2490-2499, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29489355

ABSTRACT

Specific targeting of the integrin subtype α5ß1 possesses high potential in cancer diagnosis and therapy. Through sequential N-methylation, we successfully converted the biselective α5ß1/αvß6 peptide c(phg- isoDGR-k) into a potent peptidic RGD binding α5ß1 subtype selective ligand c(phg- isoDGR-( NMe)k). Nuclear magnetic resonance spectroscopy and molecular modeling clarified the molecular basis of its improved selectivity profile. To demonstrate its potential in vivo, c(phg- isoDGR-( NMe)k) was trimerized with the chelator TRAP and used as a positron-emission tomography tracer for monitoring α5ß1 integrin expression in a M21 mouse xenograft.


Subject(s)
Integrin alpha5beta1/drug effects , Neoplasms/diagnostic imaging , Peptides/chemical synthesis , Peptides/pharmacology , Animals , Female , Gallium Radioisotopes , Humans , Integrin alpha5beta1/biosynthesis , Ligands , Magnetic Resonance Spectroscopy , Melanoma, Experimental/diagnostic imaging , Methylation , Mice , Mice, SCID , Models, Molecular , Molecular Structure , Positron-Emission Tomography , Protein Binding , Radioactive Tracers , Tissue Distribution , Xenograft Model Antitumor Assays
3.
Am J Physiol Heart Circ Physiol ; 309(10): H1667-78, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26432845

ABSTRACT

Circulating angiogenic cells (CACs) are monocyte-derived cells with endothelial characteristics, which contribute to both angiogenesis and arteriogenesis in a paracrine way. Interferon-ß (IFN-ß) is known to inhibit these divergent processes in animals and patients. We hypothesized that IFN-ß might act by affecting the differentiation and function of CACs. CACs were cultured from peripheral blood mononuclear cells and phenotypically characterized by surface expression of monocytic and endothelial markers. IFN-ß significantly reduced the number of CACs by 18-64%. Apoptosis was not induced by IFN-ß, neither in mononuclear cells during differentiation, nor after maturation to CACs. Rather, IFN-ß impaired adhesion to, and spreading on, fibronectin, which was dependent on α5ß1 (VLA-5)-integrin. IFN-ß affected the function of VLA-5 in mature CACs, leading to rounding and detachment of cells, by induction of calpain 1 activity. Cell rounding and detachment was completely reversed by inhibition of calpain 1 activity in mature CACs. During in vitro capillary formation, CAC addition and calpain 1 inhibition enhanced sprouting of endothelial cells to a comparable extent, but were not sufficient to rescue tube formation in the presence of IFN-ß. We show that the IFN-ß-induced reduction of the numbers of in vitro differentiated CACs is based on activation of calpain 1, resulting in an attenuated adhesion to extracellular matrix proteins via VLA-5. In vivo, this could lead to inhibition of vessel formation due to reduction of the locally recruited CAC numbers and their paracrine angiogenic factors.


Subject(s)
Calpain/drug effects , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Endothelial Cells/drug effects , Interferon-beta/pharmacology , Leukocytes, Mononuclear/drug effects , Neovascularization, Physiologic/drug effects , Apoptosis/drug effects , Calpain/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Fibronectins , Humans , In Vitro Techniques , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/metabolism , Leukocytes, Mononuclear/metabolism , Neovascularization, Physiologic/physiology
4.
Gynecol Oncol ; 134(3): 624-30, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24995580

ABSTRACT

OBJECTIVE: Resveratrol (Res) is known to inhibit adhesion of numerous malignancies though its effect on an adherence of ovarian cancer cells to peritoneal mesothelium remains undefined. METHODS: To address this issue, ovarian cancer cells (A2780, OVCAR-3, SKOV-3) were subjected to Res (10, 50, 100 µM), and then their adhesion to omentum-derived human peritoneal mesothelial cells (HPMCs) was assayed. RESULTS: The study showed that Res inhibits adhesion of all ovarian cancer cell lines investigated. More importantly, this effect was evident either when cancer cells were directly treated with Res (cell-dependent activity) or when intact cancer cells were pretreated with conditioned medium (CM) generated by their counterparts subjected to Res (medium-dependent activity). Cell-dependent activity of Res has been recognized to be linked with decreased level of cellular α5ß1 integrins which decreased functionality corresponds with reduced efficiency of cancer cell adhesion. Medium-related effects have been, in turn, associated with up-regulated secretion of soluble HA to environment (CM). The experiments with exogenous HA revealed the inverse relation between HA concentration in CM and cancer cell adhesion. When the CM from cells subjected with Res (with elevated HA) was supplemented with hyaluronidase, the restoration of cell adhesive capabilities occurred. CONCLUSIONS: Our studies evidenced that Res affects ovarian cancer cell adhesion to HPMCs by decreasing cellular α5ß1 integrin level and by increasing the secretion of HA to environment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Hyaluronic Acid/biosynthesis , Integrin alpha5beta1/biosynthesis , Integrin alpha5beta1/drug effects , Ovarian Neoplasms/pathology , Stilbenes/pharmacology , Cell Adhesion/drug effects , Epithelial Cells , Epithelium , Female , Humans , Peritoneum , Resveratrol , Tumor Cells, Cultured
5.
Fitoterapia ; 82(6): 911-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21605636

ABSTRACT

Ginsenoside Rh1 has been reported to possess antiallergic and anti-inflammatory activities, but its effects on monocytes remain to be determined. Herein, we investigated the effects of Rh1 on the expression of MCP-1 and CCR2, activation of MAPK signaling, and chemotaxis of monocytes. Treatment of Rh1 decreased the levels of MCP-1 and CCR2 and the expression of VLA5 and activated ß1 integrin on the cell surface, and attenuated the phosphorylation of MAPKs. Based on these results, the inhibitory effects of Rh1 on monocyte function should be regarded as a promising new anti-inflammatory response with a potential therapeutic role against inflammation-dependent diseases.


Subject(s)
Cell Movement/drug effects , Ginsenosides/pharmacology , Leukemia, Monocytic, Acute/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Chemokine CCL2/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Dose-Response Relationship, Drug , Ginsenosides/chemistry , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Integrin beta1/drug effects , Integrin beta1/genetics , Integrin beta1/metabolism , Leukemia, Monocytic, Acute/genetics , Receptors, CCR2/drug effects , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
6.
Int J Pharm ; 366(1-2): 201-10, 2009 Jan 21.
Article in English | MEDLINE | ID: mdl-18835580

ABSTRACT

Integrin alpha(5)beta(1) is expressed on several types of cancer cells, including colon cancer, and plays an important role in tumor growth and metastasis. The ability to target the integrin alpha(5)beta(1) using an appropriate drug delivery nano-vector can significantly help in inhibiting tumor growth, reducing tumor metastasis, and decreasing deleterious side effects associated with different cancer therapies. Liposomes are nano-sized phospholipid bilayer vesicles that have been extensively studied as drug delivery carriers. The goal of this study is to design stealth liposomes (liposomes covered with polyethylene glycol (PEG)) that will target colon cancer cells that express the integrin alpha(5)beta(1). The PEG provides a steric barrier allowing the liposomes to circulate in the blood and the functionalizing moiety, PR_b peptide, will specifically recognize and bind to alpha(5)beta(1) expressing cells. PR_b is a novel peptide sequence that mimics the cell adhesion domain of fibronectin, and includes four building blocks, RGDSP (the primary recognition site for alpha(5)beta(1)), PHSRN (the synergy site for alpha(5)beta(1)), a (SG)(5) linker, and a KSS spacer. In this study we have demonstrated that by varying the amount of PEG (PEG750 or PEG2000) and PR_b on the liposomal interface we can engineer nano-vectors that bind to CT26.WT, HCT116, and RKO colon cancer cells in a specific manner and are internalized through most likely alpha(5)beta(1)-mediated endocytosis. GRGDSP-targeted stealth liposomes bind to colon cancer cells and internalize, but they have much lesser efficiency than PR_b-targeted stealth liposomes, and more importantly they are not as specific since many integrins bind to RGD peptides. PR_b-targeted stealth liposomes are as cytotoxic as free 5-Fluorouracil (5-FU) and exert the highest cytotoxicity on CT26.WT cells compared to GRGDSP-targeted stealth liposomes and non-targeted stealth liposomes. Thus, the proposed targeted delivery system has the great potential to deliver a therapeutic load directly to colon cancer cells, in an efficient and specific manner.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Integrin alpha5beta1/drug effects , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/physiopathology , Endocytosis , Fibronectins/chemistry , Fluorouracil/pharmacology , Humans , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Liposomes , Mice , Peptides/chemistry , Polyethylene Glycols/chemistry
7.
Clin Cancer Res ; 14(23): 7924-9, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047123

ABSTRACT

PURPOSE: This study aimed to assess the safety and feasibility of administering volociximab, a chimeric monoclonal antibody that specifically binds to alpha(5)beta(1) integrin, and to determine the pharmacokinetics, pharmacodynamics, and preliminary evidence of antitumor activity. EXPERIMENTAL DESIGN: Patients with advanced solid malignancies were treated with escalating doses of volociximab i.v. administered over 60 minutes. Blood samples were assayed to determine plasma pharmacokinetic parameters, detect human antichimeric antibody formation, and determine the saturation of alpha5beta1 sites on peripheral blood monocytes. RESULTS: Twenty-one patients received 223 infusions of volociximab at doses ranging from 0.5 to 15 mg/kg i.v. on days 1, 15, 22, 29, and 36; and weekly thereafter. Treatment was well tolerated, and dose-limiting toxicity was not identified over the range examined. Mild (grade 1 or 2), reversible fatigue was the principal toxicity of volociximab at the highest dose levels of 10 and 15 mg/kg. Nausea, fever, anorexia, headache, vomiting, and myalgias were mild and infrequent, and there was no hematologic toxicity. Volociximab had biexponential distribution; clearance was inversely related to increasing dose, and the half-life at 15 mg/kg was estimated as being 30 days. Three patients tested positive for anti-volociximab antibodies. Saturation of monocyte alpha5beta1 integrin sites was dose-dependent up to 15 mg/kg. There was one minor response (renal, 7 months) and one durable stable disease (melanoma, 14 months). CONCLUSIONS: Volociximab can be safely administered at 15 mg/kg i.v. per week. The absence of severe toxicities and preliminary activity at the highest dose level warrants further disease-directed studies.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Integrin alpha5beta1/drug effects , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Monocytes/drug effects , Monocytes/metabolism
8.
Arch Inst Pasteur Tunis ; 85(1-4): 69-80, 2008.
Article in French | MEDLINE | ID: mdl-19469418

ABSTRACT

Snake venoms are a rich natural source of bioactive molecules, such as peptides, proteins and enzymes, more and more used in biomedical research in diagnostic or therapeutic purposes. The protein components of snake venoms belong to diverse families such as serine proteases, phospholipases, disintegrins, metalloproteinases and C-type lectins. Due to their effects on various receptors such as GPIb, GPVI, alpha2beta1..., the C-type lectins were considered, in first time, as modulators of the platelet aggregation. Recently, some of them have been described for their anti-tumoral potential effect due to their capacity to inhibit adhesion, migration, proliferation and invasion of different cancer cell lines. Also, the C-type lectins have a powerful antiangiogenic effect in vivo and in vitro by interacting with integrins of endothelial cells.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Lectins, C-Type/therapeutic use , Neovascularization, Pathologic/prevention & control , Platelet Aggregation/drug effects , Snake Venoms/chemistry , Animals , Cell Line, Tumor/drug effects , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Humans , Integrin alpha2beta1/drug effects , Integrin alpha5beta1/drug effects , Lectins, C-Type/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects
9.
J Neurosci ; 27(28): 7447-58, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17626205

ABSTRACT

Previous studies demonstrated that cross-linking of GM1 ganglioside with multivalent ligands, such as B subunit of cholera toxin (CtxB), induced Ca2+ influx through an unidentified, voltage-independent channel in several cell types. Application of CtxB to undifferentiated NG108-15 cells resulted in outgrowth of axon-like neurites in a Ca2+ influx-dependent manner. In this study, we demonstrate that CtxB-induced Ca2+ influx is mediated by TRPC5 channels, naturally expressed in these cells and primary neurons. Both Ca2+ influx and neurite induction were blocked by TRPC5 small interfering RNA (siRNA). Pretreatment of NG108-15 cells with neuraminidase increased cell-surface GM1 and greatly enhanced the signal. GM1 was not directly associated with TRPC5 but rather with alpha5beta1 integrin, which opened the channel through a signaling sequence after cross-linking of the GM1/integrin complex. This cascade included autophosphorylation of focal adhesion kinase and subsequent activation of phospholipase Cgamma (PLCgamma) and phosphoinositide-3 kinase [PI(3)K]. Pharmacological blockers that inhibited tyrosine kinase, PLC, and PI(3)K suppressed both CtxB-induced Ca2+ influx and neurite outgrowth. These were also suppressed by SK&F96365, a nonspecific transient receptor potential channel blocker. Confocal immunocytochemistry revealed that GM1 cross-linking induced colocalization of GM1 with these signaling elements in sprouting regions of plasma membrane. In primary cerebellar granular neurons (CGNs), TRPC5 was detected at 2 d in vitro (2 DIV), a stage corresponding to CtxB-stimulated Ca2+ influx. Neurite outgrowth in CGNs, determined at 3 DIV, was accelerated by CtxB and suppressed by TRPC5 siRNA and the above blockers. The crucial role of GM1 was indicated with CGNs from ganglio-series null mice, in which growth of axons was significantly retarded.


Subject(s)
Calcium/metabolism , Cross-Linking Reagents/pharmacology , G(M1) Ganglioside/pharmacology , Integrin alpha5beta1 , Neurites/drug effects , Neurites/physiology , TRPC Cation Channels/metabolism , Animals , Axons/physiology , Cell Membrane/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Cholera Toxin/pharmacology , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/metabolism , Integrins/metabolism , Mice , Mice, Knockout , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Phosphorylation , Signal Transduction/drug effects , TRPC Cation Channels/deficiency , Tissue Distribution
10.
Br J Pharmacol ; 151(8): 1235-45, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17592510

ABSTRACT

BACKGROUND AND PURPOSE: Endothelial cell proliferation, migration and adhesion are necessary for the formation of new blood vessels. We reported previously that baicalein strongly inhibited proliferation of rat heart endothelial cells and here we assess effects on migration and adhesion of these cells. EXPERIMENTAL APPROACH: Effects of baicalein on endothelial migration and adhesion were determined by in vitro wound assays and in modified Boyden chambers. Protein expression and subcellular distribution in rat heart endothelial cells were analysed by immunoblots and immunofluorescence staining. RESULTS: Pretreatment with baicalein for 48 h resulted in a concentration-dependent inhibition of endothelial migration, with an IC(50) of approximately 20 microM. Adhesion assays revealed that baicalein stimulated endothelial cell adhesion to fibronectin and vitronectin, effects blocked by the synthetic peptide Arg-Gly-Asp (RGD). Moreover, treatment with a blocking antibody against integrin alpha5beta1 drastically attenuated baicalein-mediated endothelial adhesion to fibronectin, but not to vitronectin. Furthermore, baicalein-mediated anti-migration effect and adhesion promotion could be partially reversed by the addition of 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE). Western blot analysis indicated that baicalein increased expression levels of integrin-alpha5beta1, -alphavbeta3 and vinculin proteins. Immunofluorescence staining showed that baicalein induced a marked reorganization of actin stress fibres and the recruitment of vinculin and integrins to focal adhesion plaques, with consequently increased formation of focal adhesion contacts. CONCLUSIONS AND IMPLICATIONS: Baicalein markedly inhibited the migration and enhanced the adhesion of rat heart endothelial cells, possibly by up-regulation of the integrins (alpha5beta1 and alphavbeta3) and vinculin and by promotion of actin reorganization and focal adhesion contact formation.


Subject(s)
Cell Adhesion/drug effects , Enzyme Inhibitors/pharmacology , Flavanones/pharmacology , Up-Regulation/drug effects , Actins/metabolism , Animals , Blotting, Western , Cell Movement/drug effects , Dose-Response Relationship, Drug , Endothelial Cells , Enzyme Inhibitors/administration & dosage , Fibronectins/drug effects , Flavanones/administration & dosage , Fluorescent Antibody Technique , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/metabolism , Integrins/drug effects , Integrins/metabolism , Rats , Receptors, Vitronectin/drug effects , Receptors, Vitronectin/metabolism , Vinculin/drug effects , Vinculin/metabolism
11.
Int J Cancer ; 121(11): 2402-9, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17597104

ABSTRACT

Endostatin, a C-terminal fragment of collagen type XVIII, is one of the well-characterized endogenous inhibitors of angiogenesis. Endostatin is known to bind integrin alpha(5)beta(1), which is upregulated on tumor endothelium. Most of the ovarian cancer cells express significant amounts of alpha(5)beta(1) integrin, which is important for ovarian cancer cells to attach to the peritoneal wall. Therefore we investigated whether endostatin could directly bind ovarian cancer cells and inhibit tumor cell attachment to extracellular matrix. Binding of endostatin to ovarian cancer cells was characterized by preincubation with function blocking antibodies to integrin subunits. These studies showed that ovarian cancer cell attachment to fibronectin-coated wells can be inhibited by alpha(5)beta(1) integrin specific antibodies as well as endostatin. Downregulation of integrin alpha(5) and beta(1) by siRNA abrogated the binding of OVCAR5 and human umbilical vein endothelial cell to endostatin. Although endostatin treatment did not affect ovarian cancer cell migration, treated cells failed to attach mouse peritoneal wall preparations. These studies suggest an extra-antiangiogenic role for endostatin, which can be used prevent peritoneal attachment and dissemination of ovarian cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Endostatins/pharmacology , Integrin alpha5beta1/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/prevention & control , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endostatins/metabolism , Female , Fibronectins/metabolism , Flow Cytometry , Humans , Integrin alpha5beta1/drug effects , Integrins/metabolism , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , RNA, Small Interfering/metabolism
12.
J Periodontol ; 77(5): 883-90, 2006 May.
Article in English | MEDLINE | ID: mdl-16671882

ABSTRACT

BACKGROUND: Tumor necrosis factor-alpha (TNF-alpha) is associated with chronic gingival inflammation and is suspected to influence periodontal destruction. However, the exact roles of TNF-alpha in wound healing and periodontal tissue regeneration are largely unknown. In the present study, we examined the effects of TNF-alpha on migration and proliferation of human periodontal ligament (PDL) cells. METHODS: PDL cells were cultured in the presence of TNF-alpha to determine its effects on cellular migration and proliferation. The protein expression profiles of alpha5 and beta1 integrin subunits and their related molecules, paxillin and focal adhesion kinases (FAK), were investigated. Gene expression of fibronectin also was assayed. Further, the activation of Rho-family small guanosine triphosphate (GTP)-binding protein (RhoA) was evaluated using a GTP-loading pull-down assay, and focal adhesion formation by PDL cells after transfection with the expression vector of paxillin-fused green fluorescent protein (GFP) also was observed with confocal microscopy. RESULTS: Cellular migration was impaired by TNF-alpha and recovered following the addition of anti-TNF-alpha antibodies. In contrast, PDL cell proliferation was not affected by TNF-alpha. TNF-alpha upregulated the expression of the alpha5 and beta1 integrin subunits, whereas fibronectin was not overexpressed. Phosphorylation of paxillin and FAK by PDL cells was induced, and RhoA activation also was induced. Confocal microscopic analysis revealed that TNF-alpha induced focal adhesion and stress fiber formation in all parts of the cells. CONCLUSION: Our results suggested that TNF-alpha impairs cellular migration by enhancing cellular adhesive ability following significant focal adhesion and stress fiber formation.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Periodontal Ligament/cytology , Tumor Necrosis Factor-alpha/pharmacology , Adolescent , Female , Fibronectins/genetics , Focal Adhesion Protein-Tyrosine Kinases/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/metabolism , Paxillin/drug effects , Paxillin/metabolism , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/metabolism
13.
Biomaterials ; 27(9): 1907-16, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16310247

ABSTRACT

The ability of fibronectin (Fn) to mediate cell adhesion through binding to alpha(5)beta(1) integrins is dependent on the conditions of its adsorption to the surface. Using a model system of alkylsilane SAMs with different functional groups (X=OH, COOH, NH(2) and CH(3)) and an erythroleukemia cell line expressing a single integrin (alpha(5)beta(1)), the effect of surface properties on the cellular adhesion with adsorbed Fn layers was investigated. (125)I-labeled Fn, a modified biochemical cross-linking/extraction technique and a spinning disc apparatus were combined to quantify the Fn adsorption, integrin binding and adhesion strength, respectively. This methodology allows for a binding equilibrium analysis that more closely reflects cellular adhesion found in stable tissue constructs in vivo. Differences in detachment strength and integrin binding were explained in terms of changes in the adhesion constant (psi, related to affinity) and binding efficiency of the adsorbed Fn for the alpha(5)beta(1) integrins (CH(3) approximately NH(2)

Subject(s)
Biocompatible Materials/pharmacology , Cell Adhesion , Fibronectins/drug effects , Integrin alpha5beta1/drug effects , Silanes/pharmacology , Cell Line, Tumor , Cross-Linking Reagents/pharmacology , Fibronectins/metabolism , Humans , Integrin alpha5beta1/metabolism , Succinimides/pharmacology , Surface Properties
14.
BMC Urol ; 5: 8, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15890073

ABSTRACT

BACKGROUND: Current models of the mechanism by which intravesical BCG induces an anti-tumor effect in urothelial carcinoma propose a secondary cellular immune response as principally responsible. Our group has demonstrated that BCG mediated cross-linking of alpha51 integrin receptors present on the tumor surface elicits a complex biologic response involving AP1 and NF-kappaB signaling as well as the transactivation of immediate early genes. This study evaluated the direct biologic effect of cross-linking alpha5beta1 integrin on cell cycle progression and apoptosis in two human urothelial carcinoma cell lines. METHODS: Two independent assays (MTT and Colony forming ability) were employed to measure the effect of alpha5beta1 cross-linking (antibody mediated or BCG) on cellular proliferation. Flow cytometry was employed to measure effect of BCG and alpha5beta1 antibody mediated cross-linking on cell cycle progression. Apoptosis was measured using assays for both DNA laddering and Caspase 3 activation. RESULTS: Results demonstrate that integrin cross-linking by BCG, or antibody mediated crosslinking of alpha5beta1 resulted in a decrease in proliferating cell number. BCG treatment or alpha5beta1 cross-linking increased the percentage of cells in G0/G1, in both 253J and T24 cell lines. Peptide mediated blockade of integrin binding site using RGDS reversed the effect BCG on both proliferation and cell cycle arrest. Apoptosis in response to BCG was not identified by either DNA laddering or Caspase 3 activation. CONCLUSION: These findings show that BCG exerts a direct cytostatic effect on human urothelial carcinoma cell lines. Cell cycle arrest at the G1/S interface is a mechanism by which BCG inhibits cellular proliferation. This effect is duplicated by antibody mediated cross-linking of alpha5beta1 and likely occurs as a consequence of crosslink-initiated signal transduction to cell cycle regulatory genes.


Subject(s)
Adjuvants, Immunologic/pharmacology , Apoptosis/drug effects , BCG Vaccine/pharmacology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Cell Cycle/drug effects , Integrin alpha5beta1/drug effects , Adjuvants, Immunologic/therapeutic use , BCG Vaccine/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Cell Line, Tumor , Humans , Integrin alpha5beta1/metabolism
15.
Transplant Proc ; 37(4): 1679-81, 2005 May.
Article in English | MEDLINE | ID: mdl-15919428

ABSTRACT

We tested a hypothesis that interactions between fibronectin (FN), a key extracellular matrix component, and its integrin alpha5beta1 receptor are important in the development of ischemia/reperfusion (I/R) injury of steatotic liver transplants. We examined the effect of a cyclic RGD peptide (cRGD), with high affinity for alpha5beta1 integrin, in a well-established steatotic rat liver model of ex vivo cold ischemia followed by isotransplantation. In this model, cRGD peptides were administered through the portal vein of steatotic Zucker rat livers prior to and after cold ischemic storage. Lean Zucker recipients of fatty orthotopic liver transplants (OLTs) received an additional course of cRGD peptides 1 hour posttransplantation. cRGD peptide therapy significantly inhibited the recruitment of monocyte/macrophages, and repressed the expression of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-alpha and interferon (IFN)-gamma. Moreover, it resulted in selective inhibition of inducible nitric oxide synthase (iNOS), and MMP-9 expression. Importantly, cRGD peptide therapy improved the function and histologic preservation of steatotic liver grafts, extending their 14-day survival in lean recipients from 50% in untreated to 100% in cRGD-treated OLTs. Thus, cRGD peptide-mediated blockade of FN-alpha5beta1 interaction protects against severe I/R injury otherwise experienced by steatotic OLTs.


Subject(s)
Fatty Liver/genetics , Integrin alpha5beta1/physiology , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Animals , Integrin alpha5beta1/drug effects , Male , Matrix Metalloproteinase 9/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Rats , Rats, Zucker
16.
Exp Biol Med (Maywood) ; 230(4): 271-80, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15792949

ABSTRACT

Soft tissue injury accounts for approximately 44% of all wounds in both the military and civilian populations. Following injury to soft tissue, Substance P (SP) and other neuropeptides are released by cutaneous neurons and modulate the function of immunocompetent and inflammatory cells, as well as epithelial and endothelial cells. The interaction between these components of the nervous system and multiple target cells affecting cutaneous repair is of increasing interest. In this report, we describe the effects of SP on wound repair in a novel, laser-induced, skin-wound model. Gross and histologic examination of laser-induced injury revealed that exogenously administered SP affects wound healing via neurite outgrowth, in addition to adhesion molecule and neurokinin-1 receptor involvement in vivo. All SP effects were decreased by pretreatment with Spantide II, an SP antagonist. The elucidation of SP-mediating mechanisms is crucial to firmly establishing the involvement and interaction of the peripheral nervous system and the immune system in cutaneous repair. Findings presented here suggest that SP participates in the complex network of mediators involved in cutaneous inflammation and wound healing.


Subject(s)
Skin Physiological Phenomena , Skin/pathology , Substance P/analogs & derivatives , Substance P/pharmacology , Wound Healing/drug effects , Animals , Disease Models, Animal , Immunohistochemistry , Injections, Subcutaneous , Integrin alpha5beta1/drug effects , Intercellular Adhesion Molecule-1/drug effects , Lasers , Male , Neurites/physiology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/drug effects , Substance P/administration & dosage , Substance P/antagonists & inhibitors , Time Factors
17.
Arthritis Res Ther ; 7(1): R118-26, 2005.
Article in English | MEDLINE | ID: mdl-15642131

ABSTRACT

Fibroblast-like synoviocytes (FLSs) play a major role in the pathogenesis of rheumatoid arthritis (RA) by secreting effector molecules that promote inflammation and joint destruction. How these cells become and remain activated is still elusive. Both genetic and environmental factors probably play a role in transforming FLSs into inflammatory matrix-degrading cells. As bacterial products have been detected in the joint and shown to trigger joint inflammation, this study was undertaken to investigate whether a bacterial ligand of integrin alpha5beta1, protein I/II, could contribute to the aggressive behavior of RA FLSs. Protein I/II is a pathogen-associated molecular pattern (PAMP) isolated from oral streptococci that have been identified in the joints of RA patients. The response of RA and osteoarthritis FLSs to protein I/II was analyzed using human cancer cDNA expression arrays. RT-PCR and pro-MMP-3 (pro-matrix metalloproteinase) assays were then performed to confirm the up-regulation of gene expression. Protein I/II modulated about 6% of all profiled genes. Three of these, those encoding IL-6, leukemia inhibitory factor, and MMP-3, showed a high expression level in all RA FLSs tested, whereas the expression of genes encoding other members of the cytokine or MMP-family was not affected. Furthermore, the up-regulation of MMP-3 gene expression was followed by an increase of pro-MMP-3 release. The expression of interferon regulatory factor 1 and fibroblast growth factor-5 was also up-regulated, although the expression levels were lower. Only one gene, that for insulin-like growth factor binding protein-4, was down-regulated in all RA FLSs. In contrast, in osteoarthritis FLSs only one gene, that for IL-6, was modulated. These results suggest that a bacterial ligand of integrin alpha5beta1 may contribute to the aggressive behavior of RA FLSs by inducing the release of pro-inflammatory cytokines and a cartilage-degrading enzyme, such as IL-6 and MMP-3, respectively.


Subject(s)
Arthritis, Rheumatoid/enzymology , Bacterial Proteins/pharmacology , Fibroblasts/enzymology , Integrin alpha5beta1/drug effects , Matrix Metalloproteinase 3/biosynthesis , Streptococcus mutans/chemistry , Synovial Membrane/pathology , Arthritis, Rheumatoid/pathology , Bacterial Proteins/genetics , Cell Wall/chemistry , Enzyme Induction/drug effects , Enzyme Precursors/biosynthesis , Enzyme Precursors/genetics , Fibroblast Growth Factor 5/biosynthesis , Fibroblast Growth Factor 5/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Integrin alpha5beta1/physiology , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Leukemia Inhibitory Factor , Ligands , Matrix Metalloproteinase 3/metabolism , Osteoarthritis/enzymology , Osteoarthritis/pathology , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/drug effects , Synovial Membrane/metabolism
18.
J Cell Sci ; 118(Pt 2): 291-300, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15615773

ABSTRACT

Integrin adhesion receptors are structurally dynamic proteins that adopt a number of functionally relevant conformations. We have produced a conformation-dependent anti-alpha5 monoclonal antibody (SNAKA51) that converts alpha5beta1 integrin into a ligand-competent form and promotes fibronectin binding. In adherent fibroblasts, SNAKA51 preferentially bound to integrins in fibrillar adhesions. Clustering of integrins expressing this activation epitope induced directional translocation of alpha5beta1, mimicking fibrillar adhesion formation. Priming of alpha5beta1 integrin by SNAKA51 increased the accumulation of detergent-resistant fibronectin in the extracellular matrix, thus identifying an integrin conformation that promotes matrix assembly. The SNAKA51 epitope was mapped to the calf-1/calf-2 domains. We propose that the action of the antibody causes the legs of the integrin to change conformation and thereby primes the integrin to bind ligand. These findings identify SNAKA51 as the first anti-integrin antibody to selectively recognize a subset of adhesion contacts, and they identify an integrin conformation associated with integrin translocation and fibronectin matrix formation.


Subject(s)
Extracellular Matrix/physiology , Fibronectins/physiology , Integrin alpha5beta1/physiology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/physiology , Biological Transport , Cell Adhesion/drug effects , Extracellular Matrix/drug effects , Fibronectins/drug effects , Humans , Integrin alpha5beta1/biosynthesis , Integrin alpha5beta1/drug effects , K562 Cells , Ligands , Molecular Mimicry , Protein Conformation , Protein Transport/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/physiology , Signal Transduction/physiology
19.
J Vasc Res ; 40(3): 234-43, 2003.
Article in English | MEDLINE | ID: mdl-12902636

ABSTRACT

NOV is a member of the CCN family of matricellular proteins. We have shown previously that NOV is strongly expressed by vascular smooth muscle cells (VSMCs) of the rat carotid artery. However, 7 days after injury, NOV expression is down-regulated, except near the luminal surface of the developing intima, where it is strongly expressed. These data suggested that NOV might be involved in the regulation of endothelial cell adhesion. NOV promoted the adhesion of human umbilical vein endothelial cells (HUVECs), which was abolished by anti-NOV antibody. HUVEC adhesion to NOV required divalent cations and was inhibited by GRGDS peptide, implicating integrins in the adhesion mechanism. Monoclonal antibodies (mAbs) against alphavbeta3 inhibited adhesion of HUVECs to NOV, and NOV was shown to bind to alphavbeta3. Anti-alpha5beta1 mAbs also inhibited HUVEC adhesion to NOV, but adhesion via alpha5beta1 was mediated by fibronectin. HUVEC adhesion to NOV caused intracellular signalling, as evidenced by increased phosphotyrosine content of focal adhesion kinase. Together with evidence that NOV expression in a variety of tissues is restricted to blood vessels containing VSMCs, these data are consistent with a role for NOV in endothelial cell adhesion in vascular homeostasis and in the response to injury.


Subject(s)
Endothelium, Vascular/physiology , Immediate-Early Proteins/physiology , Integrin alpha5beta1/physiology , Integrin alphaVbeta3/physiology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/physiology , Cells, Cultured , Connective Tissue Growth Factor , Endothelium, Vascular/cytology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/pharmacology , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/immunology , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Nephroblastoma Overexpressed Protein , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Tissue Distribution
20.
Int J Dermatol ; 41(12): 836-40, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12492965

ABSTRACT

BACKGROUND: Interferon-alpha and -gamma are glycoproteins with antiviral and immunoregulatory properties. In vitro studies have shown a role for these cytokines in the regulation of epidermal keratinocyte growth and differentiation. In the same way, integrins are adhesion molecules which regulate keratinocyte proliferation and differentiation. AIM: To determine whether the regulatory activity of interferons on keratinocyte proliferation and differentiation is related to a modulation of keratinocyte integrins. METHODS: Two different methods were used: monolayers and reconstituted skin, incubated either with 1,200 U/mL interferon-alpha or 500 U/mL interferon-gamma or control medium for 48 h. The integrin expression was assessed by flow cytometry and immunohistochemistry. RESULTS: In monolayers, only the alpha3 subunit was significantly inhibited by interferon-gamma. In reconstituted skin, where keratinocytes are differentiated, both interferons had an inductive effect on beta1 expression and interferon-alpha had an inhibitory effect on alpha6 expression. CONCLUSION: Interferon-alpha and -gamma induce a modulatory effect on alpha3, alpha6 and beta1 which appears to be related to the state of differentiation. Moreover, the decreased expression of alpha6 and alpha3 could be one of the mechanisms involved in the formation of bullous lesions during long-term interferon therapy.


Subject(s)
Antigenic Modulation/drug effects , Immunologic Factors/pharmacology , Integrins/analysis , Integrins/drug effects , Interferon-alpha/pharmacology , Keratinocytes/drug effects , Cell Differentiation/drug effects , Humans , In Vitro Techniques , Integrin alpha2beta1/analysis , Integrin alpha2beta1/drug effects , Integrin alpha3beta1/analysis , Integrin alpha3beta1/drug effects , Integrin alpha5beta1/analysis , Integrin alpha5beta1/drug effects , Integrin alpha6beta4/analysis , Integrin alpha6beta4/drug effects , Keratinocytes/pathology , Receptors, Vitronectin/analysis , Receptors, Vitronectin/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...