Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 1312, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28465589

ABSTRACT

The development of metastases is a multistep process that requires the activation of physiological and biochemical processes that govern migration, invasion and entry of metastatic cells into blood vessels. The urokinase receptor (uPAR) promotes cell migration by interacting with the Formyl Peptide Receptors (FPRs). Since both uPAR and FPR1 are involved in tumor progression, the uPAR-FPR1 interaction is an attractive therapeutic target. We previously described peptide antagonists of the uPAR-FPR1 interaction that inhibited cell migration and angiogenesis. To develop enzyme-resistant analogues, we applied here the Retro-Inverso (RI) approach, whereby the topology of the side chains is maintained by inverting the sequence of the peptide and the chirality of all residues. Molecular dynamics suggests that peptide RI-3 adopts the turn structure typical of uPAR-FPR1 antagonists. Accordingly, RI-3 is a nanomolar competitor of N-formyl-Met-Leu-Phe for binding to FPR1 and inhibits migration, invasion, trans-endothelial migration of sarcoma cells and VEGF-triggered endothelial tube formation. When sarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density, circulating tumor cells and pulmonary metastases were significantly reduced in animals treated daily with 6 mg/Kg RI-3 as compared to animals treated with vehicle only. Thus, RI-3 represents a promising lead for anti-metastatic drugs.


Subject(s)
Neovascularization, Pathologic/drug therapy , Peptides/administration & dosage , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Sarcoma/drug therapy , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Mice , Molecular Dynamics Simulation , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Sarcoma/genetics , Sarcoma/pathology , Xenograft Model Antitumor Assays
2.
Oncotarget ; 7(34): 54474-54487, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27323409

ABSTRACT

The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility and angiogenesis by interacting with the formyl peptide receptor type 1 (FPR1). In this study, we present evidence that the cyclization of the uPAR88-92 sequence generates a new potent inhibitor of migration, and extracellular matrix invasion of human osteosarcoma and chondrosarcoma cells expressing comparable levels of FPR1 on cell surface. In vitro, the cyclized peptide [SRSRY] prevents formation of capillary-like tubes by endothelial cells co-cultured with chondrosarcoma cells and trans-endothelial migration of osteosarcoma and chondrosarcoma cells. When chondrosarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density and circulating tumor cells in blood samples collected before the sacrifice, were significantly reduced in animals treated daily with i.p-administration of 6 mg/Kg [SRSRY] as compared to animals treated with vehicle only. Our findings indicate that [SRSRY] prevents three key events occurring during the metastatic process of osteosarcoma and chondrosarcoma cells: the extracellular matrix invasion, the formation of a capillary network and the entry into bloodstream.


Subject(s)
Bone Neoplasms/blood supply , Chondrosarcoma/blood supply , Neovascularization, Pathologic/drug therapy , Osteosarcoma/blood supply , Peptides, Cyclic/therapeutic use , Receptors, Urokinase Plasminogen Activator/therapeutic use , Animals , Cell Line, Tumor , Cell Movement , Chondrosarcoma/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Osteosarcoma/pathology , Receptors, Formyl Peptide/physiology
3.
PLoS One ; 10(5): e0126172, 2015.
Article in English | MEDLINE | ID: mdl-25938482

ABSTRACT

The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility. We and others have previously documented that the uPAR88-92 sequence, even in the form of synthetic linear peptide (SRSRY), interacts with the formyl peptide receptor type 1 (FPR1), henceforth inducing cell migration of several cell lines, including monocytes. FPR1 is mainly expressed by mammalian phagocytic leukocytes and plays a crucial role in chemotaxis. In this study, we present evidence that the cyclization of the SRSRY sequence generates a new potent and stable inhibitor of monocyte trafficking. In rat basophilic leukaemia RBL-2H3/ETFR cells expressing high levels of constitutively activated FPR1, the cyclic SRSRY peptide ([SRSRY]) blocks FPR1 mediated cell migration by interfering with both internalization and ligand-uptake of FPR1. Similarly to RBL-2H3/ETFR cells, [SRSRY] competes with fMLF for binding to FPR1 and prevents agonist-induced FPR1 internalization in human monocyte THP-1 cells. Unlike scramble [RSSYR], [SRSRY] inhibits fMLF-directed migration of monocytes in a dose-dependent manner, with IC50 value of 0.01 nM. PMA-differentiated THP-1 cell exposure to fMLF gradient causes a marked cytoskeletal re-organization with the formation of F-actin rich pseudopodia that are prevented by the addition of [SRSRY]. Furthermore, [SRSRY] prevents migration of human primary monocytes and trans-endothelial migration of monocytes. Our findings indicate that [SRSRY] is a new FPR1 inhibitor which may suggest the development of new drugs for treating pathological conditions sustained by increased motility of monocytes, such as chronic inflammatory diseases.


Subject(s)
Monocytes/drug effects , Monocytes/physiology , Peptides/metabolism , Peptides/pharmacology , Receptors, Urokinase Plasminogen Activator/chemistry , Receptors, Urokinase Plasminogen Activator/metabolism , Transendothelial and Transepithelial Migration/drug effects , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclization , Macrophages/drug effects , Macrophages/physiology , Peptides/chemistry , Protein Binding , Rats
4.
Oncotarget ; 6(7): 5324-41, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25481874

ABSTRACT

Proteomic analysis identified differentially expressed proteins between zoledronic acid-resistant and aggressive DU145R80 prostate cancer (PCa) cells and their parental DU145 cells. Ingenuity Pathway Analysis (IPA) showed a strong relationship between the identified proteins within a network associated with cancer and with homogeneous cellular functions prevalently related with regulation of cell organization, movement and consistent with the smaller and reduced cell-cell contact morphology of DU145R80 cells. The identified proteins correlated in publically available human PCa genomic data with increased tumor expression and aggressiveness. DU145R80 exhibit also a clear increase of alpha-v-(αv) integrin, and of urokinase receptor (uPAR), both included within the same network of the identified proteins. Interestingly, the actin-rich structures localized at the cell periphery of DU145R80 cells are rich of Filamin A, one of the identified proteins and uPAR which, in turn, co-localizes with αv-integrin, in podosomes and/or invadopodia. Notably, the invasive feature of DU145R80 may be prevented by blocking anti-αv antibody. Overall, we unveil a signaling network that physically links the interior of the nucleus via the cytoskeleton to the extracellular matrix and that could dictate PCa aggressiveness suggesting novel potential prognostic markers and therapeutic targets for PCa patients.


Subject(s)
Bone Density Conservation Agents/pharmacology , Diphosphonates/pharmacology , Drug Resistance, Neoplasm , Imidazoles/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics/methods , Signal Transduction/drug effects , Blotting, Western , Cell Movement , Cell Proliferation , Electrophoresis, Gel, Two-Dimensional , Gene Ontology , Gene Regulatory Networks , Humans , Male , Neoplasm Invasiveness , Prostatic Neoplasms/drug therapy , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured , Zoledronic Acid
5.
Oncotarget ; 5(12): 4154-69, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24980826

ABSTRACT

The clinical relevance of the urokinase receptor (uPAR) as a prognostic marker in ovarian cancer is well documented. We had shown that the uPAR sequence corresponding to 84-95 residues, linking D1 and D2 domains (uPAR84-95), drives cell migration and angiogenesis in a protease-independent manner. This study was aimed at defining the contribution of uPAR84-95 sequence to invasion of ovarian cancer cells. Now, we provide evidence that the ability of uPAR-expressing ovarian cancer cells to cross extra-cellular matrix and mesothelial monolayers is prevented by specific inhibitors of the uPAR84-95 sequence. To specifically investigate uPAR84-95 function, uPAR-negative CHO-K1 cells were stably transfected with cDNAs coding for uPAR D2 and D3 regions exposing (uPARD2D3) or lacking (uPAR∆D2D3) the 84-95 sequence. CHO-K1/D2D3 cells were able to cross matrigel, mesothelial and endothelial monolayers more efficiently than CHO-K1/∆D2D3 cells, which behave as CHO-K1 control cells. When orthotopically implanted in nude mice, tumor nodules generated by CHO-K1/D2D3 cells spreading to peritoneal cavity were more numerous as compared to CHO-K1/∆D2D3 cells. Ovarian tumor size and intra-tumoral microvessel density were significantly reduced in the absence of uPAR84-95. Our results indicate that cell associated uPAR promotes growth and abdominal dissemination of ovarian cancer cells mainly through its uPAR84-95 sequence.


Subject(s)
Ovarian Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Prognosis , Receptors, Urokinase Plasminogen Activator , Transfection
6.
Mol Cancer Ther ; 13(5): 1092-104, 2014 May.
Article in English | MEDLINE | ID: mdl-24705350

ABSTRACT

This work is based on previous evidence showing that chemotactic sequence of the urokinase receptor (uPAR(88-92)) drives angiogenesis in vitro and in vivo in a protease-independent manner, and that the peptide Ac-Arg-Glu-Arg-Phe-NH(2) (RERF) prevents both uPAR(88-92)- and VEGF-induced angiogenesis. New N-acetylated and C-amidated peptide analogues containing α-methyl α-amino acids were designed and synthesized to optimize the biochemical properties for therapeutic applications. Among these, Ac-L-Arg-Aib-L-Arg-D-Cα(Me)Phe-NH2, named UPARANT, adopts in solution a turned conformation similar to that found for RERF, is stable to sterilization in 3 mg/mL sealed vials in autoclave for 20 minutes at 120°C, is stable in blood, and displays a long-time resistance to enzymatic proteolysis. UPARANT competes with N-formyl-Met-Leu-Phe (fMLF) for binding to the formyl-peptide receptor, inhibits VEGF-directed endothelial cell migration, and prevents cytoskeletal organization and αvß3 activation in endothelial cells exposed to VEGF. In vitro, UPARANT inhibits VEGF-dependent tube formation of endothelial cells at a 100× lower concentration than RERF. In vivo, UPARANT reduces to the basal level VEGF-dependent capillary sprouts originating from the host vessels that invaded Matrigel sponges implanted in mice, and completely prevents neovascularization induced by subcorneal implantation of pellets containing VEGF in rabbits. Both excellent stability and potency position UPARANT as a promising new therapeutic agent for the control of diseases fueled by excessive angiogenesis, such as cancer and inflammation.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Peptides/chemistry , Peptides/pharmacology , Angiogenesis Inhibitors/metabolism , Animals , Binding, Competitive , Cell Line , Cell Movement/drug effects , Cytoskeleton/metabolism , Drug Stability , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Models, Molecular , Molecular Conformation , Neovascularization, Physiologic/drug effects , Nuclear Magnetic Resonance, Biomolecular , Peptides/metabolism , Phosphorylation/drug effects , Protein Binding , Rabbits , Receptors, Formyl Peptide/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Mol Cancer Ther ; 12(10): 1981-93, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939376

ABSTRACT

The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration, and uPAR88₋92 is the minimal sequence required to induce cell motility. We previously showed that soluble forms of uPAR elicit angiogenic responses through their uPAR88₋92 chemotactic sequence and that the synthetic peptide SRSRY exerts similar effects. By a drug design approach, based on the conformational analysis of the uPAR88₋92 sequence, we developed peptides (pERERY, RERY, and RERF) that potently inhibit signaling triggered by uPAR88₋92. In this study, we present evidence that these peptides are endowed also with a clear-cut antiangiogenic activity, although to different extents. The most active, RERF, prevents tube formation by human endothelial cells exposed to SRSRY. RERF also inhibits VEGF-triggered endothelial cell migration and cord-like formation in a dose-dependent manner, starting in the femtomolar range. RERF prevents F-actin polymerization, recruitment of αvß3 integrin at focal adhesions, and αvß3/VEGFR2 complex formation in endothelial cells exposed to VEGF. At molecular level, the inhibitory effect of RERF on VEGF signaling is shown by the decreased amount of phospho-FAK and phospho-Akt in VEGF-treated cells. In vivo, RERF prevents VEGF-dependent capillary sprouts originating from the host vessels that invaded angioreactors implanted in mice and neovascularization induced by subcorneal implantation of pellets containing VEGF in rabbits. Consistently, RERF reduced the growth and vascularization rate of tumors formed by HT1080 cells injected subcutaneously in the flanks of nude mice, indicating that RERF is a promising therapeutic agent for the control of diseases fuelled by excessive angiogenesis such as cancer.


Subject(s)
Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Peptides/administration & dosage , Receptors, Urokinase Plasminogen Activator/genetics , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Movement/drug effects , Corneal Keratocytes/drug effects , Drug Design , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Peptides/chemical synthesis , Peptides/chemistry , Rabbits , Receptors, Urokinase Plasminogen Activator/administration & dosage , Receptors, Urokinase Plasminogen Activator/chemistry , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
8.
PLoS One ; 7(9): e44806, 2012.
Article in English | MEDLINE | ID: mdl-23049759

ABSTRACT

The receptor for urokinase-type plasminogen activator (uPAR) plays an important role in controlling cell migration. uPAR binds urokinase and vitronectin extracellular ligands, and signals in complex with transmembrane receptors such as Formyl-peptide Receptors (FPR)s and integrins. Previous work from this laboratory has shown that synthetic peptides, corresponding to the uPAR(88-92) chemotactic sequence, when carrying the S90P or S90E substitutions, up- or down-regulate cell migration, respectively. To gain mechanistic insights into these opposite cell responses, the functional consequences of S90P and S90E mutations in full-length uPAR were evaluated. First, (HEK)-293 embryonic kidney cells expressing uPAR(S90P) exhibit enhanced FPR activation, increased random and directional cell migration, long-lasting Akt phosphorylation, and increased adhesion to vitronectin, as well as uPAR/vitronectin receptor association. In contrast, the S90E substitution prevents agonist-triggered FPR activation and internalization, decreases binding and adhesion to vitronectin, and inhibits uPAR/vitronectin receptor association. Also, 293/uPAR(S90P) cells appear quite elongated and their cytoskeleton well organized, whereas 293/uPAR(S90E) cells assume a large flattened morphology, with random orientation of actin filaments. Interestingly, when HT1080 cells co-express wild type uPAR with uPAR S90E, the latter behaves as a dominant-negative, impairing uPAR-mediated signaling and reducing cell wound repair as well as lung metastasis in nude mice. In contrast, signaling, wound repair and in vivo lung metastasis of HT1080 cells bearing wild type uPAR are enhanced when they co-express uPAR(S90P). In conclusion, our findings indicate that Ser(90) is a critical residue for uPAR signaling and that the S90P and S90E exert opposite effects on uPAR activities. These findings may be accommodated in a molecular model, in which uPAR(S90E) and uPAR(S90P) are forced into inactive and active forms, respectively, suggesting important implications for the development of novel drugs targeting uPAR function.


Subject(s)
Amino Acid Substitution , Neoplasm Invasiveness/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Animals , Cell Adhesion/genetics , Cell Line, Tumor/transplantation , Cell Movement/genetics , Cell Shape/genetics , Gene Expression , HEK293 Cells , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Mice , Mice, Nude , Phosphorylation , Plasmids , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Transfection , Vitronectin/genetics , Vitronectin/metabolism
9.
Sarcoma ; 2011: 842842, 2011.
Article in English | MEDLINE | ID: mdl-21253510

ABSTRACT

High levels of urokinase receptor (uPAR) in tissue and serum of patients with chondrosarcoma correlate with poor prognosis. First, we analyzed the uPAR levels in tissues and plasma of five patients affected by chondrosarcoma. Interestingly, very high levels of uPAR and its soluble forms (SuPAR) were found on tumor cell surfaces and plasma, respectively, of two patients with lung metastases. Therefore, to investigate the role of SuPAR in chondrosaromas, we generated a primary cell culture from a chondrosarcoma tissue overexpressing uPAR on cell surfaces. We found that chondrosarcoma-like primary culture cells release a large amount of SuPAR in the medium. In vitro, SuPAR elicits chondrosarcoma cell migration likely through its uPAR(88-92) sequence, since the DII(88-183) or DIIDIIR(88-284) uPAR domains retain motogen effect whereas DI(1-87) or DIII(184-284) domains, both lacking the uPAR(88-92) sequence, are ineffective. Chondrosarcoma cells cross matrigel in response to SuPAR, and their invasion capability is abrogated by RERF peptide which inhibits uPAR(88-92) signalling. These findings assign a role to uPAR in mobilizing chondrosarcoma cells and suggest that RERF peptide may be regarded as a prototype to generate new therapeutics for the chondrosarcoma treatment.

10.
Cell Mol Life Sci ; 68(14): 2453-67, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20972812

ABSTRACT

The receptor (CXCR4) for the stromal-derived factor-1 (SDF1) and the urokinase-receptor (uPAR) are up-regulated in various tumors. We show that CXCR4-transfected cells migrate toward SDF1 on collagen (CG) and do not on vitronectin (VN). Co-expression of cell-surface uPAR, which is a VN receptor, impairs SDF1-induced migration on CG and allows migration on VN. Blocking fMLP receptors (fMLP-R), alpha-v integrins or the uPAR region capable to interact with fMLP-Rs, impairs migration of uPAR/CXCR4-transfected cells on VN and restores their migration on CG. uPAR co-expression also reduces the adherence of CXCR4-expressing cells to various components of the extracellular matrix (ECM) and influences the partitioning of beta1 and alpha-v integrins to membrane lipid-rafts, affecting ECM-dependent signaling. uPAR interference in CXCR4 activity has been confirmed in cells from prostate carcinoma. Our results demonstrate that uPAR expression regulates the adhesive and migratory ability of CXCR4-expressing cells through a mechanism involving fMLP receptors and alpha-v integrins.


Subject(s)
Receptor Cross-Talk , Receptors, CXCR4/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Chemokine CXCL12/metabolism , Collagen/metabolism , Enzyme Activation , HEK293 Cells , Humans , Integrin alpha5beta1/metabolism , Membrane Microdomains/metabolism , Microscopy, Confocal , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , RNA Interference , Receptors, CXCR4/genetics , Receptors, Formyl Peptide/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Transfection , Vitronectin/metabolism
11.
Mol Cancer Ther ; 8(9): 2708-17, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19706734

ABSTRACT

The urokinase-type plasminogen activator receptor (uPAR) plays a central role in sustaining the malignant phenotype and promoting tumor metastasis. The Ser(88)-Arg-Ser-Arg-Tyr(92) is the minimum chemotactic sequence of uPAR required to induce the same intracellular signaling as its ligand uPA. Here, we describe the generation of new peptide inhibitors of cell migration and invasion derived from SRSRY by a drug design approach. Ac-Arg-Glu-Arg-Phe-NH(2) (i.e., RERF), which adopts a turned structure in solution, was selected for its ability to potently prevent SRSRY-directed cell migration. Fluorescein-RERF associates with very high affinity to RBL-2H3 rat basophilic leukemia cells expressing the human formyl peptide receptor (FPR). Accordingly, femtomolar concentrations of RERF prevent agonist-dependent internalization of FPR and inhibit N-formyl-Met-Leu-Phe-dependent migration in a dose-dependent manner. In the absence of FPR, fluorescein-RERF binds to cell surface at picomolar concentrations in an alphav integrin-dependent manner. The involvement of vitronectin receptor is further supported by the findings that 100 pmol/L RERF selectively inhibits vitronectin-dependent RBL-2H3 cell migration and prevents SRSRY-triggered uPAR/alphav association. Furthermore, RERF reduces the speed of wound closure and the extent of Matrigel invasion by human fibrosarcoma HT1080 cells without affecting cell proliferation. Finally, a 3- to 5-fold reduction of lung metastasis number and size in nude mice following i.v. injection of green fluorescent protein-expressing HT1080 cells in the presence of 3.32 mg/kg RERF is observed. Our findings indicate that RERF effectively prevents malignant cell invasion in vivo with no signs of toxicity and may represent a promising prototype drug for anticancer therapy.


Subject(s)
Cell Movement/drug effects , Lung Neoplasms/secondary , Neoplasm Metastasis/prevention & control , Peptide Fragments/pharmacology , Receptors, Urokinase Plasminogen Activator/chemistry , Animals , Female , Fibrosarcoma/pathology , Humans , Immunoprecipitation , Mice , Mice, Nude , Microscopy, Fluorescence , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Rats
12.
Front Biosci (Landmark Ed) ; 14(10): 3782-94, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273310

ABSTRACT

Urokinase (uPA) is a serine protease which converts plasminogen to plasmin, a broad-spectrum protease active on extracellular matrix (ECM) components. Like many components of the blood coagulation, fibrinolytic and complement cascades, uPA has a modular structure, including three conserved domains: a growth factor-like domain (GFD, residues 1 - 49), a kringle domain (residues 50 - 131), linked by an interdomain linker or "connecting peptide" (CP, residues 132 - 158) to the serine protease domain (residues 159 - 411). Although direct molecular interactions with urokinase receptor and integrins have been extensively described, the function of single uPA domains is not completely understood. Because of the causal involvment of uPA in cancer invasion and metastasis, the blockade of uPA interactions and activity with specific inhibitors is of interest for novel strategies in cancer therapy. New inhibitors derived from the interdomain linker or "connecting peptide" are coming into focus. This review summarizes the recent findings on the uPA structure-function relationship and provides further information on existing inhibitors of uPA multiple functions.


Subject(s)
Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism , Catalytic Domain , Enzyme Inhibitors/pharmacology , Humans , Kringles , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
13.
Peptides ; 30(2): 267-72, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19041917

ABSTRACT

Human beta-defensin-2 (hBD-2) is an antimicrobial peptide which is released upon microbial invasion and contributes to mucosal and epithelial defense modulating both innate and adaptive immunity. We found that hBD-2 stimulates chemotaxis of human endothelial cells with an extent similar to that exerted by the vascular endothelial growth factor (VEGF). The hBD-2-dependent chemotaxis is dose-dependent, maximal effect being reached at 500 ng/ml concentration. In the absence of any growth factor, hBD-2 favors wound healing of endothelial cells, causing an about 2-fold increase in the speed of wound closure with respect to the control. Furthermore, hBD-2 promotes endothelial cell proliferation, although at a minor extent as compared to VEGF. When plated on matrigel enriched with angiogenic factors, endothelial cells form a three-dimensional network of tubes that gives rise to capillary-like structures. Similarly to VEGF, hBD-2 promotes capillary-like tube formation of human endothelial cells. Pro-angiogenic effect promoted by hBD-2 is dose-dependent, peaks at a 500 ng/ml hBD-2 concentration and is prevented by blocking anti-alphavbeta3 monoclonal antibody. However, hBD-2-induced pro-angiogenic activity is not due to endogenously produced VEGF because it is not prevented by neutralizing anti-VEGF antibodies. Overall, our findings suggest that hBD-2 could link inflammation and the host defense through its pro-angiogenic activity.


Subject(s)
Anti-Infective Agents/pharmacology , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Endothelium, Vascular/cytology , beta-Defensins/pharmacology , Cells, Cultured , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Humans , Umbilical Veins/cytology
14.
Tumori ; 94(4): 505-10, 2008.
Article in English | MEDLINE | ID: mdl-18822686

ABSTRACT

AIMS AND BACKGROUND: The prognosis of each individual patient affected by sarcoma, including those with low histopathologic grading, cannot be reliably predicted at the time of surgery. We have developed an in vitro cell invasion assay on early primary cell cultures derived from surgically removed sarcomas. METHODS: Primary cell cultures were subjected to in vitro cell invasion assays by using Boyden chambers, filters coated with matrigel and fetal bovine serum as a source of chemoattractant. For each primary cell culture, the sarcoma cell invasion index was determined in comparison with the percentage of human fibrosarcoma HT1080 cell invasion extent. The cell invasion index of 7 different sarcomas was evaluated in respect to the outcome of the disease, after a follow-up ranging from 14 to 48 months. RESULTS: Data evidenced that a low cell invasion index (39.7% +/- 8.9) was retained by tumor cells derived from patients with no progression of the disease and with a longer interval of disease-free survival (21 +/- 0.8 months). However, an increase in cell invasion index (61% +/- 5) was retained by tumor cells derived from patients with progression of the disease and with a shorter disease-free survival (9 +/- 3 months). Overall, although only 7 cases were analyzed, a statistically significant correlation was found between disease-free survival and cell invasion index (P = 0.003). CONCLUSIONS: Our data support the possibility that cell invasion assays performed in vitro on cells derived from human sarcomas may be predictive of a more aggressive form of the disease.


Subject(s)
Sarcoma/pathology , Adolescent , Adult , Aged , Chondrosarcoma/pathology , Collagen , Disease Progression , Disease-Free Survival , Drug Combinations , Female , Fibroma/pathology , Fibrosarcoma/pathology , Humans , Immunohistochemistry , Laminin , Liposarcoma, Myxoid/pathology , Male , Middle Aged , Neoplasm Invasiveness , Predictive Value of Tests , Prognosis , Proteoglycans , Sarcoma/mortality , Sarcoma/surgery , Tumor Cells, Cultured
15.
FEBS Lett ; 582(7): 1141-6, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18339322

ABSTRACT

Urokinase receptor (uPAR) plays a key role in physiological and pathological processes sustained by an altered cell migration. We have developed peptides carrying amino acid substitutions along the Ser(88)-Arg-Ser-Arg-Tyr(92) (SRSRY) uPAR chemotactic sequence. The peptide pyro glutamic acid (pGlu)-Arg-Glu-Arg-Tyr-NH2 (pERERY-NH(2)) shares the same binding site with SRSRY and competes with N-formyl-Met-Leu-Phe (fMLF) for binding to the G-protein-coupled N-formyl-peptide receptor (FPR). pERERY-NH(2) is a dose-dependent inhibitor of both SRSRY- and fMLF-directed cell migration, and prevents agonist-induced FPR internalization and fMLF-dependent ERK1/2 phosphorylation. pERERY-NH(2) is a new and potent uPAR inhibitor which may suggest the generation of new pharmacological treatments for pathological conditions involving increased cell migration.


Subject(s)
Chemotaxis/drug effects , Oligopeptides/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Formyl Peptide/antagonists & inhibitors , Animals , Cell Line , Humans , Oligopeptides/chemistry , Peptides/chemistry , Peptides/pharmacology , Rats , Receptors, Urokinase Plasminogen Activator , Signal Transduction/drug effects
16.
J Biol Chem ; 280(26): 25225-32, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15866865

ABSTRACT

The urokinase-type plasminogen activator receptor (uPAR) sustains cell migration through its capacity to promote pericellular proteolysis, regulate integrin function, and mediate chemotactic signaling in response to urokinase. We have characterized the early signaling events triggered by the Ser-Arg-Ser-Arg-Tyr (SRSRY) chemotactic uPAR sequence. Cell exposure to SRSRY peptide promotes directional migration on vitronectin-coated filters, regardless of uPAR expression, in a specific and dose-dependent manner, with maximal effect at a concentration level as low as 10 nm. A similar concentration profile is observed in a quantitative analysis of SRSRY-dependent cytoskeletal rearrangements, mostly consisting of filamentous structures localized in a single cell region. SRSRY analogues with alanine substitutions fail to drive F-actin formation and cell migration, indicating a critical role for each amino acid residue. As with ligand-dependent uPAR signaling, SRSRY stimulates protein kinase C activity and results in ERK1/2 phosphorylation. The involvement of the high affinity N-formyl-Met-Leu-Phe receptor (FPR) in this process is indicated by the finding that 100 nm N-formyl-Met-Leu-Phe inhibits binding of D2D3 to the cell surface, as well as SRSRY-stimulated cell migration and F-actin polarization. Moreover, cell exposure to SRSRY promotes FPR-dependent vitronectin release and increased uPAR.alphavbeta5 vitronectin receptor physical association, indicating that alphavbeta5 activity is regulated by the SRSRY uPAR sequence via FPR. Finally, we provide evidence that alphavbeta5 is required for SRSRY-dependent ERK1/2 phosphorylation, whereas it is not required for protein kinase C activation. The data indicate that the ability of uPAR to stimulate cell migration and cytoskeletal rearrangements is retained by the SRSRY peptide alone and that it is supported by cross-talk between FPR and alphavbeta5.


Subject(s)
Integrin alphaVbeta3/chemistry , Receptors, Cell Surface/metabolism , Receptors, Formyl Peptide/chemistry , Actins/chemistry , Actins/metabolism , Alanine/chemistry , Androstadienes/pharmacology , Blotting, Western , Cell Adhesion , Cell Line , Cell Movement , Chemotaxis , Chromones/pharmacology , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Flavonoids/pharmacology , Humans , Immunoprecipitation , Integrin alphaV/metabolism , Integrin beta4/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Morpholines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Naphthalenes/pharmacology , Peptides/chemistry , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/chemistry , Signal Transduction , Temperature , Time Factors , Vitronectin/chemistry , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...