91 results on '"Taraboletti G"'
Search Results
2. Soluble stroma-related biomarkers of pancreatic cancer
- Author
-
Resovi, A, Bani, M, Porcu, L, Anastasia, A, Minoli, L, Allavena, P, Cappello, P, Novelli, F, Scarpa, A, Morandi, E, Falanga, A, Torri, V, Taraboletti, G, Belotti, D, Giavazzi, R, Resovi A, Bani MR, Porcu L, Anastasia A, Minoli L, Allavena P, Cappello P, Novelli F, Scarpa A, Morandi E, Falanga A, Torri V, Taraboletti G, Belotti D, Giavazzi R, Resovi, A, Bani, M, Porcu, L, Anastasia, A, Minoli, L, Allavena, P, Cappello, P, Novelli, F, Scarpa, A, Morandi, E, Falanga, A, Torri, V, Taraboletti, G, Belotti, D, Giavazzi, R, Resovi A, Bani MR, Porcu L, Anastasia A, Minoli L, Allavena P, Cappello P, Novelli F, Scarpa A, Morandi E, Falanga A, Torri V, Taraboletti G, Belotti D, and Giavazzi R
- Abstract
The clinical management of pancreatic ductal adenocarcinoma (PDAC) is hampered by the lack of reliable biomarkers. This study investigated the value of soluble stroma-related molecules as PDAC biomarkers. In the first exploratory phase, 12 out of 38 molecules were associated with PDAC in a cohort of 25 PDAC patients and 16 healthy subjects. A second confirmatory phase on an independent cohort of 131 PDAC patients, 30 chronic pancreatitis patients, and 131 healthy subjects confirmed the PDAC association for MMP7, CCN2, IGFBP2, TSP2, sICAM1, TIMP1, and PLG Multivariable logistic regression model identified biomarker panels discriminating respectively PDAC versus healthy subjects (MMP7 + CA19.9, AUC = 0.99, 99% CI = 0.98-1.00) (CCN2 + CA19.9, AUC = 0.96, 99% CI = 0.92-0.99) and PDAC versus chronic pancreatitis (CCN2 + PLG+FN+Col4 + CA19.9, AUC = 0.94, 99% CI = 0.88-0.99). Five molecules were associated with PanIN development in two GEM models of PDAC (PdxCre/LSL-KrasG12D and PdxCre/LSL-KrasG12D/+/LSL-Trp53R172H/+), suggesting their potential for detecting early disease. These markers were also elevated in patient-derived orthotopic PDAC xenografts and associated with response to chemotherapy. The identified stroma-related soluble biomarkers represent potential tools for PDAC diagnosis and for monitoring treatment response of PDAC patients
- Published
- 2018
3. Non-peptidic Thrombospondin-1 Mimics as Fibroblast Growth Factor-2 Inhibitors: AN INTEGRATED STRATEGY FOR THE DEVELOPMENT OF NEW ANTIANGIOGENIC COMPOUNDS*
- Author
-
Colombo G., Margosio B., Ragona L., Neves M., Bonifacio S., Annis D.S., Stravalaci M., Tomaselli S., Giavazzi R., Rusnati M., Presta M., Zetta L., Mosher D.F., Ribatti D., Gobbi M., and Taraboletti G.
- Subjects
Magnetic Resonance Spectroscopy ,Protein Conformation ,Angiogenesis Inhibitors ,Chorion ,Chorioallantoic Membrane ,Recombinant Proteins ,Thrombospondin 1 ,Kinetics ,Protein Structure and Folding ,Animals ,Humans ,Fibroblast Growth Factor 2 ,Peptides ,Chickens ,Cell Proliferation ,Protein Binding - Abstract
Endogenous inhibitors of angiogenesis, such as thrombospondin-1 (TSP-1), are promising sources of therapeutic agents to treat angiogenesis-driven diseases, including cancer. TSP-1 regulates angiogenesis through different mechanisms, including binding and sequestration of the angiogenic factor fibroblast growth factor-2 (FGF-2), through a site located in the calcium binding type III repeats. We hypothesized that the FGF-2 binding sequence of TSP-1 might serve as a template for the development of inhibitors of angiogenesis. Using a peptide array approach followed by binding assays with synthetic peptides and recombinant proteins, we identified a FGF-2 binding sequence of TSP-1 in the 15-mer sequence DDDDDNDKIPD-DRDN. Molecular dynamics simulations, taking the full flexibility of the ligand and receptor into account, and nuclear magnetic resonance identified the relevant residues and conformational determinants for the peptide-FGF interaction. This information was translated into a pharmacophore model used to screen the NCI2003 small molecule databases, leading to the identification of three small molecules that bound FGF-2 with affinity in the submicromolar range. The lead compounds inhibited FGF-2-induced endothelial cell proliferation in vitro and affected angiogenesis induced by FGF-2 in the chicken chorioallantoic membrane assay. These small molecules, therefore, represent promising leads for the development of antiangiogenic agents. Altogether, this study demonstrates that new biological insights obtained by integrated multidisciplinary approaches can be used to develop small molecule mimics of endogenous proteins as therapeutic agents.
- Published
- 2010
- Full Text
- View/download PDF
4. The tyrosine kinase inhibitor E-3810 combined with paclitaxel inhibits the growth of advanced-stage triple-negative breast cancer xenografts
- Author
-
Bello, E, Taraboletti, G, Colella, G, Zucchetti, M, Forestieri, D, Licandro, S, Berndt, A, Richter, P, D'Incalci, M, Cavalletti, E, Giavazzi, R, Camboni, G, Damia, G, Bello E., Taraboletti G., Colella G., Zucchetti M., Forestieri D., Licandro S. A., Berndt A., Richter P., D'Incalci M., Cavalletti E., Giavazzi R., Camboni G., Damia G., Bello, E, Taraboletti, G, Colella, G, Zucchetti, M, Forestieri, D, Licandro, S, Berndt, A, Richter, P, D'Incalci, M, Cavalletti, E, Giavazzi, R, Camboni, G, Damia, G, Bello E., Taraboletti G., Colella G., Zucchetti M., Forestieri D., Licandro S. A., Berndt A., Richter P., D'Incalci M., Cavalletti E., Giavazzi R., Camboni G., and Damia G.
- Abstract
E-3810 is a novel small molecule that inhibits VEGF receptor-1, -2, and -3 and fibroblast growth factor receptor-1 tyrosine kinases at nmol/L concentrations currently in phase clinical II. In preclinical studies, it had a broad spectrum of antitumor activity when used as monotherapy in a variety of human xenografts. We here investigated the activity of E-3810 combined with different cytotoxic agents in a MDA-MB-231 triple-negative breast cancer xenograft model. The molecule could be safely administered with 5-fluorouracil, cisplatin, and paclitaxel. The E-3810-paclitaxel combination showed a striking activity with complete, lasting tumor regressions; the antitumor activity of the combination was also confirmed in another triple-negative breast xenograft, MX-1. The activity was superior to that of the combinations paclitaxel+brivanib and paclitaxel+sunitinib. Pharmacokinetics studies suggest that the extra antitumor activity of the combination is not due to higher paclitaxel tumor levels, which in fact were lower in mice pretreated with all three kinase inhibitors, and the paclitaxel plasma levels excluded reduced drug availability. Pharmacodynamic studies showed that E-3810, brivanib, and sunitinib given as single agents or in combination with paclitaxel reduced the number of vessels, but did not modify vessel maturation. Reduced tumor collagen IV and increased plasma collagen IV, associated with increased matrix metalloproteinases (MMP), particularly host MMP-9, indicate a proteolytic remodeling of the extracellular matrix caused by E-3810 that in conjunction with the cytotoxic effect of paclitaxel on the tumor cells (caspase-3/7 activity) may contribute to the striking activity of their combination. These data support the therapeutic potential of combining E-3810 with conventional chemotherapy. © 2012 American Association for Cancer Research.
- Published
- 2013
5. Expression levels of vascular endothelial growth factor, matrix metalloproteinases 2 and 9 and tissue inhibitor of metalloproteinases 1 and 2 in the plasma of patients with ovarian carcinoma
- Author
-
Manenti, L, Paganoni, P, Floriani, I, Landoni, F, Torri, V, Buda, A, Taraboletti, G, Labianca, R, Belotti, D, Giavazzi, R, Manenti L., Paganoni P., Floriani I., Landoni F., Torri V., Buda A., Taraboletti G., Labianca R., Belotti D., Giavazzi R., Manenti, L, Paganoni, P, Floriani, I, Landoni, F, Torri, V, Buda, A, Taraboletti, G, Labianca, R, Belotti, D, Giavazzi, R, Manenti L., Paganoni P., Floriani I., Landoni F., Torri V., Buda A., Taraboletti G., Labianca R., Belotti D., and Giavazzi R.
- Abstract
We measured the levels of the vascular endothelial growth factor (VEGF), matrix metalloproteinases type 2 and type 9 (MMP-2 and MMP-9) and tissue inhibitors of matrix metalloproteinase 1 and 2 (TIMP-1 and TIMP-2) in the plasma of patients with ovarian carcinoma (n=40), in other gynaecological pathologies (n=30) and in the plasma of healthy volunteers (n=26). MMP-2 and MMP-9 (pro and active forms) gelatinolytic activity was measured by zymography. Enzyme-linked immunosorbent assays (ELISA) were used to assay soluble VEGF and TIMPs. Preoperative plasma VEGF levels were significantly higher in patients with ovarian cancer than in healthy volunteers (P<0.0001) or patients with a benign gynaecological pathology (P<0.0001). The expression of pro-MMP-9 was higher in the plasma of ovarian cancer patients than in the plasma of women with non-malignant disease (P=0.01) or healthy women (P<0.0002). Pro-MMP-2 was detected in the plasma of ovarian cancer patients, but levels did not differ from those in non-malignant disease or healthy donor samples. Plasma TIMP-1 and TIMP-2 levels were significantly higher in patients with ovarian carcinomas than in healthy volunteers (P<0.0001 and P=0.006, respectively) or in the patients with a non-malignant pathology (P<0.0001 and P=0.002, respectively). Sub-group analysis showed that VEGF and pro-MMP-9 were higher in the plasma of patients with serous carcinomas than other histological types. Furthermore, plasma VEGF and pro-MMP-9 levels were higher in the plasma of cancer patients with thrombocytosis. Throughout the study, and in the univariate analysis, no correlation was found between the VEGF, MMP and TIMP levels. Only TIMP-1 was associated with a poor survival and mortality risk
- Published
- 2003
6. Thrombospondin/HIV-1 Tat protein interaction: modulation of the biological activity of extracellular Tat
- Author
-
RUSNATI M., TARABOLETTI G., URBINATI G., TULIPANO R., GIULIANI R., MOLINARI TOSATTI M. P., SENNINO B., ALBINI A., NOONAN D., GIAVAZZI R., PRESTA M., GIACCA, MAURO, Rusnati, M., Taraboletti, G., Urbinati, G., Tulipano, R., Giuliani, R., MOLINARI TOSATTI, M. P., Sennino, B., Giacca, Mauro, Albini, A., Noonan, D., Giavazzi, R., and Presta, M.
- Published
- 2000
7. The FGF2-binding domain of thrombospondin-1: functional characterization and exploitation to design antiangiogenic compounds
- Author
-
Taraboletti, G., Rusnati, Marco, Ragona, L., and Colombo, G.
- Subjects
angiogenesis ,endothelium ,FGF2 ,thrombospondin-1 - Published
- 2010
8. Hepatocyte growth factor (HGF) downregulates thrombospondin I (TSP-I) expression in thyroid papillary carcinoma cells
- Author
-
Scarpino, S, DI NAPOLI, Arianna, Taraboletti, G, Cancrini, A, and Ruco, Luigi
- Published
- 2005
9. Vascular endothelial growth factor c promotes ovarian carcinoma progression through paracrine and autocrine mechanisms
- Author
-
Decio, A, Taraboletti, G, Patton, V, Alzani, R, Perego, P, Fruscio, R, Jürgensmeier, J, Giavazzi, R, Belotti, D, FRUSCIO, ROBERT, Belotti, D., Decio, A, Taraboletti, G, Patton, V, Alzani, R, Perego, P, Fruscio, R, Jürgensmeier, J, Giavazzi, R, Belotti, D, FRUSCIO, ROBERT, and Belotti, D.
- Abstract
Vascular endothelial growth factor C (VEGFC) has been reported to promote tumor progression in several tumor types, mainly through the stimulation of lymphangiogenesis and lymphatic metastasis. However, the expression and biological significance of the VEGFC/VEGF receptor (VEGFR)-3 pathway in ovarian cancer growth and dissemination are unclear, and have been investigated in this study. Soluble VEGFC was detected in the plasma and ascites of patients with ovarian carcinoma, and VEGFR3 expression was found in their tumor tissues. In human ovarian carcinoma xenograft models, high levels of soluble VEGFC in ascites and serum were detected, in association with disease progression, tumor burden, and volume of ascites. Peak VEGFC expression preceded para-aortic lymph node infiltration by HOC8 neoplastic cells. Histological detection of tumor cells in blood and lymphatic vessels indicated both hematogenous and lymphatic dissemination. Overexpression of VEGFC in the VEGFR3-positive and luciferase-expressing IGROV1 cells promoted carcinoma dissemination after orthotopic transplantation in the ovary of immunodeficient mice. In vitro, VEGFC released by the tumor cells stimulated tumor cell migration in an autocrine manner. Cediranib, an inhibitor of VEGFR1-3 and c-kit, inhibited in vivo metastasis of VEGFC-overexpressing IGROV1 and in vitro autocrine effects. These findings suggest that the VEGFC/VEGFR3 pathway acts as an enhancer of ovarian cancer progression through autocrine and paracrine mechanisms, hence offering a potential target for therapy.
- Published
- 2014
10. Increased tumorigenicity and invasiveness of C6 rat glioma cells transfected with the human alpha-2,8 sialyltransferase cDNA
- Author
-
Sottocornola, E., Colombo, I., Vergani, V., Taraboletti, G., and Berra, B.
- Subjects
DNA, Complementary ,Reverse Transcriptase Polymerase Chain Reaction ,Chemotaxis ,Recombinant Fusion Proteins ,Mice, Nude ,Glioma ,Transfection ,Polymerase Chain Reaction ,Sialyltransferases ,Rats ,Mice ,Settore BIO/10 - Biochimica ,Gangliosides ,Tumor Cells, Cultured ,Animals ,Humans ,Neoplasm Invasiveness ,Chromatography, Thin Layer ,C6 rat glioma cells ,Invasion ,Proliferation rate ,Sialyltransferase ,beta-D-Galactoside alpha 2-6-Sialyltransferase ,Chromatography, High Pressure Liquid - Abstract
Gangliosides are thought to be involved in tumor cell proliferation, migration and invasiveness as so far demonstrated by the addition of exogenous gangliosides to the culture medium. To better understand the direct influence that alterations in ganglioside synthesis can exert on these functional aspects of cell biology, in the present study, we investigated the behaviour of C6 rat glioma cells after stable transfection with the human CMP-NeuAc:NeuAcalpha2-3Galbeta1-4GlcCer alpha2,8-sialyltransferase (SAT-II, EC 2.4.99.8) gene. The enzyme synthesizes ganglioside GD(3) by adding a sialic acid residue to ganglioside GM(3). Stable transfection of the constructs into C6 cells and expression of the human SAT-II gene were evaluated using PCR and RT-PCR amplification, respectively. Qualitative and quantitative analysis of the ganglioside profile was performed by conventional HP-TLC and identity of de novo synthesized species was assessed by TLC immunostaining. Results show that whereas C6 parental cells and C6 cells transfected with the empty expression vector synthesize, almost exclusively, ganglioside GM(3), de novo synthesis of GD(3) is clearly observed in clones expressing the alpha2,8-sialyltransferase. Subcutaneous grafting in athymic nude mice of cells expressing high levels of GD(3) induces tumors growing faster and more aggressively than controls. In in vitro assays, the same cells demonstrate increased proliferation rate, motility and invasiveness. Chemotaxis and chemoinvasion were assayed using the modified Boyden chamber. Data obtained suggest that endogenously neosynthesized GD(3) is able to modify proliferation rate, motility and invasion of C6 rat glioma cells, enhancing the features of malignancy of this tumor cell line.
- Published
- 1999
11. Paclitaxel (Taxol(R)) inhibits motility of paclitaxel-resistant human ovarian carcinoma cells
- Author
-
Belotti D, Rieppi M, Mi, Nicoletti, Am, Casazza, Fojo T, Taraboletti G, and Raffaella Giavazzi
- Subjects
Ovarian Neoplasms ,Paclitaxel ,Cell Movement ,Drug Resistance, Neoplasm ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,Female ,Antineoplastic Agents, Phytogenic ,Cell Division - Abstract
The effect of paclitaxel on the adhesive and motility properties of human ovarian carcinoma cell lines was investigated. Paclitaxel significantly inhibited the motility of OVCAR 5, SK-OV-3, and HOC-1OTC ovarian carcinoma cell lines (IC50 = 2.1 x 10(-8), 2 x 10(-9), and 1.9 x 10(-8) m, respectively) but did not affect the adhesion of these cells to the subendothelial matrix. The association between inhibition of motility and cytotoxic activity was investigated using an A2780 subclone (1A9) and three paclitaxel-resistant variants (designated 1A9/PTX22, 1A9/PTX10, and 1A9/PTX18). Although paclitaxel did not significantly affect the adhesion to subendothelial matrix of the sublines, it completely inhibited their migration. Inhibition of migration was similar in 1A9 cells and the resistant sublines, with an IC50 of 1 x 10(-8) for 1A9 cells and 5.4 x 10(-9), 1.1 x 10(-8), and 5.2 x 10(-9) m for 1A9/PTX22, 1A9/PTX10, and 1A9/PTX18, respectively. Paclitaxel inhibited motility induced by soluble attractant (chemotaxis) and immobilized attractant (haptotaxis). Inhibition of cell motility occurred in the absence of an antiproliferative effect, because higher concentrations of paclitaxel were required to inhibit tumor cell proliferation (IC50 = 1.9 x 10(-7) and 4.6 x 10(-6), 1 x 10(-5), and 3.1 x 10(-6) m for 1A9 and 1A9/PTX22, 1A9/PTX10, and 1A9/PTX18, respectively). These data show that paclitaxel is a potent inhibitor of ovarian carcinoma cell motility and that this activity is independent of its cytotoxic activity.
- Published
- 1996
12. Sequence dependent antitumour efficacy of the vascular disrupting agent ZD6126 in combination with paclitaxel
- Author
-
Martinelli, M, primary, Bonezzi, K, additional, Riccardi, E, additional, Kuhn, E, additional, Frapolli, R, additional, Zucchetti, M, additional, Ryan, A J, additional, Taraboletti, G, additional, and Giavazzi, R, additional
- Published
- 2007
- Full Text
- View/download PDF
13. Antiangiogenic activity of aplidine, a new agent of marine origin
- Author
-
Taraboletti, G, primary, Poli, M, additional, Dossi, R, additional, Manenti, L, additional, Borsotti, P, additional, Faircloth, G T, additional, Broggini, M, additional, D'Incalci, M, additional, Ribatti, D, additional, and Giavazzi, R, additional
- Published
- 2004
- Full Text
- View/download PDF
14. Functional domains of the 67-kDa laminin receptor precursor.
- Author
-
Castronovo, V., primary, Taraboletti, G., additional, and Sobel, M.E., additional
- Published
- 1991
- Full Text
- View/download PDF
15. Platelet thrombospondin modulates endothelial cell adhesion, motility, and growth: a potential angiogenesis regulatory factor.
- Author
-
Taraboletti, G, primary, Roberts, D, additional, Liotta, L A, additional, and Giavazzi, R, additional
- Published
- 1990
- Full Text
- View/download PDF
16. Sulfatide-binding domain of the laminin A chain.
- Author
-
Taraboletti, G, primary, Rao, C N, additional, Krutzsch, H C, additional, Liotta, L A, additional, and Roberts, D D, additional
- Published
- 1990
- Full Text
- View/download PDF
17. la antigen expression and IL-1 activity in murine tumour-associated macrophages.
- Author
-
Peri, G., Rossi, V., Taraboletti, G., Erroi, A., and Mantovani, A.
- Subjects
MACROPHAGES ,RETICULO-endothelial system ,TUMORS ,LYMPHOKINES ,SARCOMA ,T cells - Abstract
Tumour-associated macrophages (TAM) isolated from five murine sarcomas had a relatively high frequency of I-A
+ cells, with mean values of 27% (mFS6), 52% (MN/MCA1 ), 68% (N3), 62% (N4) and 98% (J3) for TAM compared to 12% for resident peritoneal macrophages. Expression of I-E in TAM was also high (29%) in the only sarcoma (N4) examined in this respect. Expression of I-A by TAM declined in culture but exposure to lymphokine supernatants maintained and increased the frequency of I-A+ cells in TAM. Transplantation of tumours into nude mice caused a marked decrease in the percentage of I-A+ TAM in the case of the N4 sarcoma (8% compared to 48%), whereas for the MN/MCA1 sarcoma the diminution was only marginal (from 53 to 41%), TAM from murine sarcomas did not constitutively release appreciable levels of interleukin-1 (IL-1) activity. Upon stimulation with bacterial lipopolysaccharides or silica, TAM showed a limited capacity to produce and release IL-1 activity compared to peritoneal macrophages. Thus the expression of I-A antigens and the IL-1-producing capacity are uncoupled in TAM from murine sarcomas. These properties of TAM could play an important role in the generation of anti-tumour immunity and/or of suppressive T-cell circuits. [ABSTRACT FROM AUTHOR]- Published
- 1986
18. Preclinical development of metalloproteasis inhibitors in cancer therapy
- Author
-
Giavazzi, R. and Taraboletti, G.
- Published
- 2001
- Full Text
- View/download PDF
19. Thrombospondin-induced tumor cell migration: haptotaxis and chemotaxis are mediated by different molecular domains.
- Author
-
Taraboletti, G, Roberts, D D, and Liotta, L A
- Abstract
Thrombospondin induces the migration of human melanoma and carcinoma cells. Using a modified Boyden chamber assay, tumor cells migrated to a gradient of soluble thrombospondin (chemotaxis). Checkerboard analysis indicated that directional migration was induced 27-fold greater than stimulation of random motility. Tumor cells also migrated in a dose-dependent manner to a gradient of substratum-bound thrombospondin (haptotaxis). A series of human melanoma and carcinoma cells were compared for their relative motility stimulation by thrombospondin haptotaxis vs. chemotaxis. Some cell lines exhibited a stronger haptotactic response compared to their chemotactic response while other lines exhibited little or no migration response to thrombospondin. Human A2058 melanoma cells which exhibit a strong haptotactic and chemotactic response to thrombospondin were used to study the structural domains of thrombospondin required for the response. Monoclonal antibody C6.7, which binds to the COOH-terminal region of thrombospondin, inhibited haptotaxis in a dose-dependent optimal manner. C6.7 had no significant effect on thrombospondin chemotaxis. In contrast, monoclonal antibody A2.5, heparin, and fucoidan, which bind to the NH2-terminal heparin-binding domain of thrombospondin, inhibited thrombospondin chemotaxis but not haptotaxis. Monoclonal antibody A6.1 directed against the internal core region of thrombospondin had no significant effect on haptotaxis or chemotaxis. Synthetic peptides GRGDS (50 micrograms/ml), but not GRGES, blocked tumor cell haptotaxis on fibronectin, but had minimal effect on thrombospondin or laminin haptotaxis. The 140-kD fragment of thrombospondin lacking the heparin-binding amino-terminal region retained the property to fully mediate haptotaxis but not chemotaxis. When the COOH region of the 140-kD fragment, containing the C6.7-binding site, was cleaved off, the resulting 120-kD fragment (which retains the RGDA sequence) failed to induce haptotaxis. Separate structural domains of thrombospondin are therefore required for tumor cell haptotaxis vs. chemotaxis. This may have implications during hematogenous cancer metastases formation.
- Published
- 1987
- Full Text
- View/download PDF
20. Role of laminin receptor in tumor cell migration
- Author
-
Um, Wewer, Taraboletti G, Me, Sobel, Reidar Albrechtsen, and La, Liotta
- Subjects
Receptors, Laminin ,Cell Movement ,Immune Sera ,Neoplasms ,Cell Adhesion ,Humans ,Enzyme-Linked Immunosorbent Assay ,Neoplasm Invasiveness ,Laminin ,Receptors, Immunologic ,Melanoma ,Fibronectins - Abstract
Polyclonal antisera were made against biochemically purified laminin receptor protein as well as against synthetic peptides deduced from a complementary DNA clone corresponding to the COOH-terminal end of the laminin receptor (U.M. Wewer et al., Proc. Natl. Acad. Sci. USA, 83: 7137-7141, 1986). These antisera were used to study the potential role of laminin receptor in laminin-mediated attachment and haptotactic migration of human A2058 melanoma cells. The anti-laminin receptor antisera reacted with the surface of suspended, nonpermeabilized melanoma and carcinoma cells. The anti-laminin receptor antisera blocked the surface interaction of A2058 cells with endogenous laminin, resulting in the inhibition of laminin-mediated cell attachment. The A2058 melanoma cells migrated toward a gradient of solid phase laminin or fibronectin (haptotaxis). Anti-laminin antiserum abolished haptotaxis on laminin but not on fibronectin. Synthetic peptide GRGDS corresponding to the fibronectin cell-binding domain inhibited haptotaxis on fibronectin but not on laminin. Both types of anti-laminin receptor antisera inhibited haptotaxis on laminin but not on fibronectin. Using immunohistochemistry, invading human carcinoma cells in vivo exhibited a marked cytoplasmic immunoreactivity for the receptor antigen. Together these findings indicate a specific role for the laminin receptor in laminin-mediated migration and that the ligand binding of the laminin receptor is encompassed in the COOH-terminal end of the protein.
- Published
- 1987
21. Tumor-derived chemotactic factor(s) from human ovarian carcinoma: evidence for a role in the regulation of macrophage content of neoplastic tissues
- Author
-
Bottazzi, B, Ghezzi, P, Taraboletti, G, Salmona, M, Colombo, N, Bonazzi, C, Mangioni, C, Mantovani, A, Mantovani, A., COLOMBO, NICOLETTA, Bottazzi, B, Ghezzi, P, Taraboletti, G, Salmona, M, Colombo, N, Bonazzi, C, Mangioni, C, Mantovani, A, Mantovani, A., and COLOMBO, NICOLETTA
- Abstract
Supernatants from freshly disaggregated human ovarian carcinomas maintained in vitro for 24 hr, from primary ovarian carcinoma cultures (4-6 days in culture) and from established ovarian cancer cell lines were examined for chemotactic activity on blood monocytes in blind-well chemotaxis chambers. Tumor-cell culture supernatants induced migration of peripheral blood monocytes across polycarbonate filters with considerable heterogeneity among different tumors. Induction of migration occurred only in the presence of a gradient between the lower and upper compartments of the chamber. Chemotactic activity was characterized by means of supernatants from primary ovarian carcinoma cultures. Chemotactic factor(s) was (were) produced in serum-free conditions and the production was inhibited by emetine but not by mitomycin C. The activity was destroyed by exposure to proteolytic enzymes and by heating at 100 degrees C but was unaffected by RNase, DNase, lipase and exposure to extreme pH values or heating at 56 degrees C. Upon fractionation on Sephadex G 75, the activity eluted as a single peak in the cytochrome C region, corresponding to an apparent molecular weight of about 12 kd. The percentage of macrophages was assessed in 25 freshly disaggregated tumor specimens. Ovarian carcinomas were heterogeneous in their macrophage content with values ranging from 4 to 36%. A significant (r = 0.62; p = 0.00097), though far from absolute, correlation was found between chemotactic activity of culture supernatants and percentage of tumor-associated macrophages. Tumor-derived chemotactic factor(s) could be one of the mechanisms involved in the regulation of the macrophage content of human ovarian carcinomas.
- Published
- 1985
22. Role of laminin receptor in tumor cell migration.
- Author
-
Wewer, U M, Taraboletti, G, Sobel, M E, Albrechtsen, R, Liotta, L A, Wewer, U M, Taraboletti, G, Sobel, M E, Albrechtsen, R, and Liotta, L A
- Abstract
Udgivelsesdato: 1987-Nov-1, Polyclonal antisera were made against biochemically purified laminin receptor protein as well as against synthetic peptides deduced from a complementary DNA clone corresponding to the COOH-terminal end of the laminin receptor (U.M. Wewer et al., Proc. Natl. Acad. Sci. USA, 83: 7137-7141, 1986). These antisera were used to study the potential role of laminin receptor in laminin-mediated attachment and haptotactic migration of human A2058 melanoma cells. The anti-laminin receptor antisera reacted with the surface of suspended, nonpermeabilized melanoma and carcinoma cells. The anti-laminin receptor antisera blocked the surface interaction of A2058 cells with endogenous laminin, resulting in the inhibition of laminin-mediated cell attachment. The A2058 melanoma cells migrated toward a gradient of solid phase laminin or fibronectin (haptotaxis). Anti-laminin antiserum abolished haptotaxis on laminin but not on fibronectin. Synthetic peptide GRGDS corresponding to the fibronectin cell-binding domain inhibited haptotaxis on fibronectin but not on laminin. Both types of anti-laminin receptor antisera inhibited haptotaxis on laminin but not on fibronectin. Using immunohistochemistry, invading human carcinoma cells in vivo exhibited a marked cytoplasmic immunoreactivity for the receptor antigen. Together these findings indicate a specific role for the laminin receptor in laminin-mediated migration and that the ligand binding of the laminin receptor is encompassed in the COOH-terminal end of the protein.
- Published
- 1987
23. Shedding of membrane vesicles by tumor and endothelial cells
- Author
-
Vincenza Dolo, D Ascenzo, S., Giusti, I., Millimaggi, D., Taraboletti, G., and Pavan, A.
- Subjects
Ovarian Neoplasms ,Umbilical Veins ,tumor ,Carcinoma, Hepatocellular ,Neovascularization, Pathologic ,Secretory Vesicles ,Carcinoma ,Cell Membrane ,angiogenesis ,endothelial cells ,membrane vesicles ,shedding ,Tissue Inhibitor of Metalloproteinases ,Exocytosis ,Matrix Metalloproteinases ,Cell Line, Tumor ,Neoplasms ,Microscopy, Electron, Scanning ,Humans ,Female ,Neoplasm Invasiveness ,Extracellular Space - Abstract
Shedding of membrane vesicles is a vital phenomenon frequently observed in tumor cells and suggested to be involved in several aspects of tumor progression. Our previous studies have shown that human breast tumor cells rapidly shed membrane vesicles containing matrix metalloproteinases (MMPs). In this study we present that human umbilical vein endothelial cells (HUVEC) as well as different tumor cell lines (human ovarian cancer, CABA I and A2780, and hepatocarcinoma cell line, SK-Hep 1) shed vesicles in the extracellular medium. These vesicles carry MMPs and their inhibitors TIMPs. We conclude that tumor and endothelial cells shed MMP-containing vesicles and this may represent a mechanism for regulating focalized proteolytic activity and a way to interact with microenvironment during tumor angiogenesis.
24. Soluble stroma-related biomarkers of pancreatic cancer
- Author
-
Luca Porcu, Maria Rosa Bani, Francesco Novelli, Paola Cappello, Alessia Anastasia, Anna Falanga, E. Morandi, Paola Allavena, Aldo Scarpa, Lucia Minoli, Andrea Resovi, Dorina Belotti, Valter Torri, Raffaella Giavazzi, Giulia Taraboletti, Resovi, A, Bani, M, Porcu, L, Anastasia, A, Minoli, L, Allavena, P, Cappello, P, Novelli, F, Scarpa, A, Morandi, E, Falanga, A, Torri, V, Taraboletti, G, Belotti, D, and Giavazzi, R
- Subjects
0301 basic medicine ,Oncology ,Male ,Medicine (General) ,endocrine system diseases ,medicine.medical_treatment ,pancreatic cancer ,QH426-470 ,medicine.disease_cause ,MMP7 ,Cohort Studies ,0302 clinical medicine ,Tumor Microenvironment ,Research Articles ,TIMP1 ,Cancer ,Aged, 80 and over ,circulating biomarkers ,Middle Aged ,Intercellular Adhesion Molecule-1 ,Prognosis ,early diagnosi ,early diagnosis ,treatment evaluation ,tumor microenvironment ,Molecular Medicine ,030220 oncology & carcinogenesis ,Matrix Metalloproteinase 7 ,Biomarker (medicine) ,Female ,KRAS ,Carcinoma, Pancreatic Ductal ,Research Article ,Adult ,medicine.medical_specialty ,03 medical and health sciences ,R5-920 ,Internal medicine ,Pancreatic cancer ,Genetics ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Aged ,Biomarkers & Diagnostic Imaging ,Tumor microenvironment ,Chemotherapy ,Tissue Inhibitor of Metalloproteinase-1 ,circulating biomarker ,business.industry ,Connective Tissue Growth Factor ,Post-translational Modifications, Proteolysis & Proteomics ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,030104 developmental biology ,Solubility ,Pancreatitis ,Stromal Cells ,business ,Thrombospondins - Abstract
The clinical management of pancreatic ductal adenocarcinoma (PDAC) is hampered by the lack of reliable biomarkers. This study investigated the value of soluble stroma‐related molecules as PDAC biomarkers. In the first exploratory phase, 12 out of 38 molecules were associated with PDAC in a cohort of 25 PDAC patients and 16 healthy subjects. A second confirmatory phase on an independent cohort of 131 PDAC patients, 30 chronic pancreatitis patients, and 131 healthy subjects confirmed the PDAC association for MMP7, CCN2, IGFBP2, TSP2, sICAM1, TIMP1, and PLG. Multivariable logistic regression model identified biomarker panels discriminating respectively PDAC versus healthy subjects (MMP7 + CA19.9, AUC = 0.99, 99% CI = 0.98–1.00) (CCN2 + CA19.9, AUC = 0.96, 99% CI = 0.92–0.99) and PDAC versus chronic pancreatitis (CCN2 + PLG+FN+Col4 + CA19.9, AUC = 0.94, 99% CI = 0.88–0.99). Five molecules were associated with PanIN development in two GEM models of PDAC (PdxCre/LSL‐Kras G12D and PdxCre/LSL‐Kras G12D/+ /LSL‐Trp53 R172H/+ ), suggesting their potential for detecting early disease. These markers were also elevated in patient‐derived orthotopic PDAC xenografts and associated with response to chemotherapy. The identified stroma‐related soluble biomarkers represent potential tools for PDAC diagnosis and for monitoring treatment response of PDAC patients.
- Published
- 2018
25. The tyrosine kinase inhibitor E-3810 combined with paclitaxel inhibits the growth of advanced-stage triple-negative breast cancer xenografts
- Author
-
Maurizio D'Incalci, Giovanna Damia, Gennaro Colella, Ezia Bello, Daniele Forestieri, Simonetta Andrea Licandro, Giulia Taraboletti, Gabriella Camboni, Ennio Cavalletti, Petra Richter, Alexander Berndt, Raffaella Giavazzi, Massimo Zucchetti, Bello, E, Taraboletti, G, Colella, G, Zucchetti, M, Forestieri, D, Licandro, S, Berndt, A, Richter, P, D'Incalci, M, Cavalletti, E, Giavazzi, R, Camboni, G, and Damia, G
- Subjects
Cancer Research ,Indoles ,Paclitaxel ,medicine.drug_class ,Alanine, Animals, Antineoplastic Combined Chemotherapy Protocols, Cell Line, Tumor, Drug Synergism, Female, Humans, Indoles, Mice, nude, Paclitaxel, Protein Kinase Inhibitors, Pyrroles, Rabeprazole, Random Allocation, Sunitinib, Triazines, Triple Negative Breast Neoplasms, Xenograft Model Antitumor Assays ,Mice, Nude ,Triple Negative Breast Neoplasms ,Pharmacology ,Tyrosine-kinase inhibitor ,chemistry.chemical_compound ,Mice ,Random Allocation ,Pharmacokinetics ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Sunitinib ,Animals ,Humans ,Pyrroles ,Protein Kinase Inhibitors ,Triple-negative breast cancer ,Cisplatin ,Alanine ,Kinase ,Chemistry ,Triazines ,Drug Synergism ,Xenograft Model Antitumor Assays ,Oncology ,Rabeprazole ,Female ,Tyrosine kinase ,medicine.drug - Abstract
E-3810 is a novel small molecule that inhibits VEGF receptor-1, -2, and -3 and fibroblast growth factor receptor-1 tyrosine kinases at nmol/L concentrations currently in phase clinical II. In preclinical studies, it had a broad spectrum of antitumor activity when used as monotherapy in a variety of human xenografts. We here investigated the activity of E-3810 combined with different cytotoxic agents in a MDA-MB-231 triple-negative breast cancer xenograft model. The molecule could be safely administered with 5-fluorouracil, cisplatin, and paclitaxel. The E-3810–paclitaxel combination showed a striking activity with complete, lasting tumor regressions; the antitumor activity of the combination was also confirmed in another triple-negative breast xenograft, MX-1. The activity was superior to that of the combinations paclitaxel+brivanib and paclitaxel+sunitinib. Pharmacokinetics studies suggest that the extra antitumor activity of the combination is not due to higher paclitaxel tumor levels, which in fact were lower in mice pretreated with all three kinase inhibitors, and the paclitaxel plasma levels excluded reduced drug availability. Pharmacodynamic studies showed that E-3810, brivanib, and sunitinib given as single agents or in combination with paclitaxel reduced the number of vessels, but did not modify vessel maturation. Reduced tumor collagen IV and increased plasma collagen IV, associated with increased matrix metalloproteinases (MMP), particularly host MMP-9, indicate a proteolytic remodeling of the extracellular matrix caused by E-3810 that in conjunction with the cytotoxic effect of paclitaxel on the tumor cells (caspase-3/7 activity) may contribute to the striking activity of their combination. These data support the therapeutic potential of combining E-3810 with conventional chemotherapy. Mol Cancer Ther; 12(2); 131–40. ©2012 AACR.
- Published
- 2012
26. Human immunodeficiency virus-1 (HIV-1)-Tat protein promotes migration of acquired immunodeficiency syndrome-related lymphoma cells and enhances their adhesion to endothelial cells
- Author
-
Federico Bussolino, Giulia Taraboletti, Mauro Giacca, Luca Barra, Giampiero Piccinini, Renato G. S. Chirivi, Raffaella Giavazzi, Maria Rosa Bani, Chirivi, R. G., Taraboletti, G., Bani, M. R., Barra, L., Piccinini, G., Giacca, Mauro, Bussolino, F., and Giavazzi, R.
- Subjects
Cell type ,Immunology ,Integrin ,Intercellular Adhesion Molecule-1 ,Biology ,Biochemistry ,immune system diseases ,Cell Movement ,hemic and lymphatic diseases ,Cell Adhesion ,Tumor Cells, Cultured ,Humans ,Cell adhesion ,Lymphoma, AIDS-Related ,Cell adhesion molecule ,Cell migration ,Cell Biology ,Hematology ,Endothelial stem cell ,Cell culture ,Gene Products, tat ,Cancer research ,biology.protein ,HIV-1 ,tat Gene Products, Human Immunodeficiency Virus ,Endothelium, Vascular - Abstract
Human immunodeficiency virus-1 (HIV-1)-Tat, the transactivating gene product of HIV-1, has been shown to interact with different cell types, inducing gene expression, altering their growth and migratory behavior. In this study we examined whether Tat might affect functions of acquired immunodeficiency syndrome (AIDS)-related non-Hodgkin’s lymphoma (NHL), relevant to the in vivo dissemination. Our results show that Tat significantly augmented the motility of the two AIDS-related Burkitt’s lymphoma cell lines (AS283 and PA682PB) and AIDS-primary effusion lymphoma cell line (HBL-6-AIDS-PEL). Mutations in RGD or basic domain of Tat (KGE-MBP and LxI-MBP, respectively) sharply reduced migration compared with wild type, suggesting that both domains are required for migration. In contrast, a Tat protein mutation outside the active domains (NH2-TAT-GST) did not reduce lymphoma cell migration. The treatment of lymphoma cells with Tat did not influence their adhesion to matrix proteins or to human vascular endothelial cells, but endothelial cells treated with Tat became more adhesive to lymphoma cells. Flow cytometric analysis showed that treatment of endothelial cells with Tat induced the cell surface expression of the adhesion molecules vascular cell adhesion molecule-1 (VCAM-1) and E-selectin and increased the expression of intercellular adhesion molecule-1 (ICAM-1). Only antibodies against VCAM-1 on endothelial cells or against the VLA-4 integrin expressed on AS283 cells inhibited the increment of adhesion, indicating the relevance of this pathway in the adhesion of lymphoma cells to vascular endothelium. In our work, we show for the first time that Tat can enhance the migration of lymphoma cells and their adhesion to endothelial cells, two processes that may contribute to the malignant behavior of NHL in patients with AIDS.
27. Fibronectin fragments generated by pancreatic trypsin act as endogenous inhibitors of pancreatic tumor growth.
- Author
-
Resovi A, Persichitti P, Brunelli L, Minoli L, Borsotti P, Garattini G, Tironi M, Dugnani E, Redegalli M, De Simone G, Pastorelli R, Bani MR, Piemonti L, Mosher DF, Giavazzi R, Taraboletti G, and Belotti D
- Subjects
- Animals, Humans, Mice, Acute Disease, Cell Line, Tumor, Cell Proliferation, Pancreas pathology, Proteomics, Trypsin metabolism, Tumor Microenvironment, Carcinoma, Pancreatic Ductal pathology, Fibronectins metabolism, Pancreatic Neoplasms pathology, Pancreatitis
- Abstract
Background: The pancreatic microenvironment has a defensive role against cancer but it can acquire tumor-promoting properties triggered by multiple mechanisms including alterations in the equilibrium between proteases and their inhibitors. The identification of proteolytic events, targets and pathways would set the basis for the design of new therapeutic approaches., Methods and Results: Here we demonstrate that spheroids isolated from human and murine healthy pancreas and co-transplanted orthotopically with pancreatic ductal adenocarcinoma (PDAC) in mouse pancreas inhibited tumor growth. The effect was mediated by trypsin-generated fibronectin (FN) fragments released by pancreatic spheroids. Tumor inhibition was observed also in a model of acute pancreatitis associated with trypsin activation. Mass spectrometry proteomic analysis of fragments and mAb against different FN epitopes identified the FN type III domain as responsible for the activity. By inhibiting integrin α5β1, FAK and FGFR1 signaling, the fragments induced tumor cell detachment and reduced cell proliferation. Consistent with the mutual relationship between the two pathways, FGF2 restored both FGFR1 and FAK signaling and promoted PDAC cell adhesion and proliferation. FAK and FGFR inhibitors additively inhibited PDAC growth in vitro and in orthotopic in vivo models., Conclusions: This study identifies a novel role for pancreatic trypsin and fibronectin cleavage as a mechanism of protection against cancer by the pancreatic microenvironment. The finding of a FAK-FGFR cross-talk in PDAC support the combination of FAK and FGFR inhibitors for PDAC treatment to emulate the protective effect of the normal pancreas against cancer., (© 2023. Italian National Cancer Institute ‘Regina Elena’.)
- Published
- 2023
- Full Text
- View/download PDF
28. Identification of a novel extracellular inhibitor of FGF2/FGFR signaling axis by combined virtual screening and NMR spectroscopy approach.
- Author
-
Pagano K, Listro R, Linciano P, Rossi D, Longhi E, Taraboletti G, Molinari H, Collina S, and Ragona L
- Subjects
- Humans, Magnetic Resonance Spectroscopy, Receptors, Fibroblast Growth Factor antagonists & inhibitors, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction, Resorcinols chemistry, Resorcinols pharmacology, Fibroblast Growth Factor 2 antagonists & inhibitors, Neoplasms
- Abstract
The aberrant activation of the fibroblast growth factor 2 (FGF2)/fibroblast growth factor receptor (FGFR) signalling pathway drives severe pathologies, including cancer development and angiogenesis-driven pathologies. The perturbation of the FGF2/FGFR axis via extracellular allosteric small inhibitors is a promising strategy for developing FGFR inhibitors with improved safety and efficacy for cancer treatment. We have previously investigated the role of new extracellular inhibitors, such as rosmarinic acid (RA), which bind the FGFR-D2 domain and directly compete with FGF2 for the same binding site, enabling the disruption of the functional FGF2/FGFR interaction. To select ligands for the previously identified FGF2/FGFR RA binding site, NMR data-driven virtual screening has been performed on an in-house library of non-commercial small molecules and metabolites. A novel drug-like compound, a resorcinol derivative named RBA4 has been identified. NMR interaction studies demonstrate that RBA4 binds the FGF2/FGFR complex, in agreement with docking prediction. Residue-level NMR perturbations analysis highlights that the mode of action of RBA4 is similar to RA in terms of its ability to target the FGF2/FGFR-D2 complex, inducing perturbations on both proteins and triggering complex dissociation. Biological assays proved that RBA4 inhibited FGF2 proliferative activity at a level comparable to the previously reported natural product, RA. Identification of RBA4 chemical groups involved in direct interactions represents a starting point for further optimization of drug-like extracellular inhibitors with improved activity., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper: [Collina Simona reports financial support was provided by University of Pavia.]., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
29. Inhibition of FGFR Signaling by Targeting FGF/FGFR Extracellular Interactions: Towards the Comprehension of the Molecular Mechanism through NMR Approaches.
- Author
-
Pagano K, Longhi E, Molinari H, Taraboletti G, and Ragona L
- Subjects
- Adenosine Triphosphate pharmacology, Comprehension, Fibroblast Growth Factors metabolism, Humans, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction, Fibroblast Growth Factor 2 pharmacology, Neoplasms metabolism
- Abstract
NMR-based approaches play a pivotal role in providing insight into molecular recognition mechanisms, affording the required atomic-level description and enabling the identification of promising inhibitors of protein-protein interactions. The aberrant activation of the fibroblast growth factor 2 (FGF2)/fibroblast growth factor receptor (FGFR) signaling pathway drives several pathologies, including cancer development, metastasis formation, resistance to therapy, angiogenesis-driven pathologies, vascular diseases, and viral infections. Most FGFR inhibitors targeting the intracellular ATP binding pocket of FGFR have adverse effects, such as limited specificity and relevant toxicity. A viable alternative is represented by targeting the FGF/FGFR extracellular interactions. We previously identified a few small-molecule inhibitors acting extracellularly, targeting FGFR or FGF. We have now built a small library of natural and synthetic molecules that potentially act as inhibitors of FGF2/FGFR interactions to improve our understanding of the molecular mechanisms of inhibitory activity. Here, we provide a comparative analysis of the interaction mode of small molecules with the FGF2/FGFR complex and the single protein domains. DOSY and residue-level NMR analysis afforded insights into the capability of the potential inhibitors to destabilize complex formation, highlighting different mechanisms of inhibition of FGF2-induced cell proliferation.
- Published
- 2022
- Full Text
- View/download PDF
30. Alternative Vascularization Mechanisms in Tumor Resistance to Therapy.
- Author
-
Belotti D, Pinessi D, and Taraboletti G
- Abstract
Blood vessels in tumors are formed through a variety of different mechanisms, each generating vessels with peculiar structural, molecular, and functional properties. This heterogeneity has a major impact on tumor response or resistance to antineoplastic therapies and is now emerging as a promising target for strategies to prevent drug resistance and improve the distribution and efficacy of antineoplastic treatments. This review presents evidence of how different mechanisms of tumor vessel formation (vasculogenesis, glomeruloid proliferation, intussusceptive angiogenesis, vasculogenic mimicry, and vessel co-option) affect tumor responses to antiangiogenic and antineoplastic therapies, but also how therapies can promote alternative mechanisms of vessel formation, contributing to tumor recurrence, malignant progression, and acquired drug resistance. We discuss the possibility of tailoring treatment strategies to overcome vasculature-mediated drug resistance or to improve drug distribution and efficacy.
- Published
- 2021
- Full Text
- View/download PDF
31. Thrombospondins in bone remodeling and metastatic bone disease.
- Author
-
Carminati L and Taraboletti G
- Subjects
- Animals, Bone Neoplasms pathology, Humans, Osteoblasts metabolism, Osteoblasts pathology, Osteoclasts metabolism, Osteoclasts pathology, Osteogenesis physiology, Bone Neoplasms metabolism, Bone Remodeling physiology, Thrombospondins metabolism
- Abstract
Thrombospondins (TSPs) are a family of five multimeric matricellular proteins. Through a wide range of interactions, TSPs play pleiotropic roles in embryogenesis and in tissue remodeling in adult physiology as well as in pathological conditions, including cancer development and metastasis. TSPs are active in bone remodeling, the process of bone resorption (osteolysis) and deposition (osteogenesis) that maintains bone homeostasis. TSPs are particularly involved in aberrant bone remodeling, including osteolytic and osteoblastic skeletal cancer metastasis, frequent in advanced cancers such as breast and prostate carcinoma. TSPs are major players in the bone metastasis microenvironment, where they finely tune the cross talk between tumor cells and bone resident cells in the metastatic niche. Each TSP family member has different effects on the differentiation and activity of bone cells-including the bone-degrading osteoclasts and the bone-forming osteoblasts-with different outcomes on the development and growth of osteolytic and osteoblastic metastases. Here, we overview the involvement of TSP family members in the bone tissue microenvironment, focusing on their activity on osteoclasts and osteoblasts in bone remodeling, and present the evidence to date of their roles in bone metastasis establishment and growth.
- Published
- 2020
- Full Text
- View/download PDF
32. CCN-Based Therapeutic Peptides Modify Pancreatic Ductal Adenocarcinoma Microenvironment and Decrease Tumor Growth in Combination with Chemotherapy.
- Author
-
Resovi A, Borsotti P, Ceruti T, Passoni A, Zucchetti M, Berndt A, Riser BL, Taraboletti G, and Belotti D
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Mice, Tumor Microenvironment, Adenocarcinoma drug therapy, Carcinoma, Pancreatic Ductal drug therapy, Peptides metabolism
- Abstract
The prominent desmoplastic stroma of pancreatic ductal adenocarcinoma (PDAC) is a determinant factor in tumor progression and a major barrier to the access of chemotherapy. The PDAC microenvironment therefore appears to be a promising therapeutic target. CCN2/CTGF is a profibrotic matricellular protein, highly present in the PDAC microenvironment and associated with disease progression. Here we have investigated the therapeutic value of the CCN2-targeting BLR100 and BLR200, two modified synthetic peptides derived from active regions of CCN3, an endogenous inhibitor of CCN2. In a murine orthotopic PDAC model, the two peptides, administered as monotherapy at low doses (approximating physiological levels of CCN3), had tumor inhibitory activity that increased with the dose. The peptides affected the tumor microenvironment, inhibiting fibrosis and vessel formation and reducing necrosis. Both peptides were active in preventing ascites formation. An increased activity was obtained in combination regimens, administering BLR100 or BLR200 with the chemotherapeutic drug gemcitabine. Pharmacokinetic analysis indicated that the improved activity of the combination was not mainly determined by the substantial increase in gemcitabine delivery to tumors, suggesting other effects on the tumor microenvironment. The beneficial remodeling of the tumor stroma supports the potential value of these CCN3-derived peptides for targeting pathways regulated by CCN2 in PDAC.
- Published
- 2020
- Full Text
- View/download PDF
33. Antimetastatic and antiangiogenic activity of trabectedin in cutaneous melanoma.
- Author
-
Carminati L, Pinessi D, Borsotti P, Minoli L, Giavazzi R, D'Incalci M, Belotti D, and Taraboletti G
- Subjects
- Animals, Cell Line, Cell Line, Tumor, Female, Macrophages drug effects, Macrophages metabolism, Melanoma metabolism, Melanoma, Experimental drug therapy, Melanoma, Experimental metabolism, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, NIH 3T3 Cells, Skin Neoplasms metabolism, Tissue Inhibitor of Metalloproteinase-1 metabolism, Tumor Microenvironment drug effects, Melanoma, Cutaneous Malignant, Angiogenesis Inhibitors pharmacology, Melanoma drug therapy, Neoplasm Metastasis drug therapy, Skin Neoplasms drug therapy, Trabectedin pharmacology
- Abstract
Trabectedin is a marine-derived antineoplastic drug. Besides targeting the cancer cells, trabectedin has a peculiar activity on the tumor microenvironment with marked effects on the vasculature and the immune response. Because a favorable microenvironment is a key factor in the progression of cutaneous melanoma, we hypothesized that trabectedin might affect the growth and metastasis of this highly aggressive cancer. This study shows that trabectedin inhibited the subcutaneous growth of the murine melanoma B16-BL6 and K1735-M2. In line with its known activities on the environment of other tumor types, it caused a significant reduction of tumor blood vessel density and tumor-associated macrophages. Trabectedin had a significant antimetastatic activity, inhibiting the formation of lung colonies following intravenous injection of B16-BL6 or K1735-M2 cells. The drug was also active in a clinically relevant spontaneous metastasis assay, where it inhibited lung metastasis when administered before (neoadjuvant) or after (adjuvant) surgical removal of the primary tumor. Relevant to its antimetastatic activity, trabectedin inhibited melanoma cell invasiveness in vitro, associated with increased tissue inhibitor of metalloproteinase-1 production and alteration in cell shape and cytoskeleton organization. This study shows that trabectedin affects melanoma growth and metastasis, acting with tumor-dependent mechanisms on both the tumor cells and the vascular and the inflammatory tumor microenvironment., (© The Author(s) 2018. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
34. Pentraxin 3 regulates synaptic function by inducing AMPA receptor clustering via ECM remodeling and β1-integrin.
- Author
-
Fossati G, Pozzi D, Canzi A, Mirabella F, Valentino S, Morini R, Ghirardini E, Filipello F, Moretti M, Gotti C, Annis DS, Mosher DF, Garlanda C, Bottazzi B, Taraboletti G, Mantovani A, Matteoli M, and Menna E
- Subjects
- Animals, Astrocytes metabolism, Brain growth & development, Brain metabolism, C-Reactive Protein genetics, CHO Cells, Cells, Cultured, Cricetinae, Cricetulus, Extracellular Matrix genetics, Mice, Mice, Inbred C57BL, Nerve Tissue Proteins genetics, Neuronal Plasticity genetics, Protein Transport genetics, Thrombospondin 1 metabolism, C-Reactive Protein physiology, Extracellular Matrix metabolism, Integrin beta1 metabolism, Nerve Tissue Proteins physiology, Receptors, AMPA metabolism, Synapses physiology
- Abstract
Control of synapse number and function in the developing central nervous system is critical to the formation of neural circuits. Astrocytes play a key role in this process by releasing factors that promote the formation of excitatory synapses. Astrocyte-secreted thrombospondins (TSPs) induce the formation of structural synapses, which however remain post-synaptically silent, suggesting that completion of early synaptogenesis may require a two-step mechanism. Here, we show that the humoral innate immune molecule Pentraxin 3 (PTX3) is expressed in the developing rodent brain. PTX3 plays a key role in promoting functionally-active CNS synapses, by increasing the surface levels and synaptic clustering of AMPA glutamate receptors. This process involves tumor necrosis factor-induced protein 6 (TSG6), remodeling of the perineuronal network, and a β1-integrin/ERK pathway. Furthermore, PTX3 activity is regulated by TSP1, which directly interacts with the N-terminal region of PTX3. These data unveil a fundamental role of PTX3 in promoting the first wave of synaptogenesis, and show that interplay of TSP1 and PTX3 sets the proper balance between synaptic growth and synapse function in the developing brain., (© 2018 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
35. ADAMTS13 Deficiency Shortens the Life Span of Mice With Experimental Diabetes.
- Author
-
Cassis P, Cerullo D, Zanchi C, Corna D, Lionetti V, Giordano F, Novelli R, Conti S, Casieri V, Matteucci M, Locatelli M, Taraboletti G, Villa S, Gastoldi S, Remuzzi G, Benigni A, and Zoja C
- Subjects
- ADAMTS13 Protein deficiency, ADAMTS13 Protein genetics, Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Connexin 43 metabolism, Dobutamine pharmacology, Immunohistochemistry, Male, Mice, Mice, Knockout, Phosphorylation drug effects, Reverse Transcriptase Polymerase Chain Reaction, Shear Strength, Thrombospondin 1 metabolism, ADAMTS13 Protein metabolism, Diabetes Mellitus, Experimental metabolism
- Abstract
In patients with diabetes, impaired activity of ADAMTS13 (a disintegrin and metalloprotease with thrombospondin type 1 repeats, member 13), the plasma metalloprotease that cleaves highly thrombogenic von Willebrand factor multimers, is a major risk factor of cardiovascular events. Here, using Adamts13
-/- mice made diabetic by streptozotocin, we investigated the impact of the lack of ADAMTS13 on the development of diabetes-associated end-organ complications. Adamts13-/- mice experienced a shorter life span than their diabetic wild-type littermates. It was surprising that animal death was not related to the occurrence of detectable thrombotic events. The lack of ADAMTS13 drastically increased the propensity for ventricular arrhythmias during dobutamine-induced stress in diabetic mice. Cardiomyocytes of diabetic Adamts13-/- mice exhibited an aberrant distribution of the ventricular gap junction connexin 43 and increased phosphorylation of Ca2+ /calmodulin-dependent kinase II (CaMKII), and with the consequent CaMKII-induced disturbance in Ca2+ handling, which underlie propensity for arrhythmia. In vitro, thrombospondin 1 (TSP1) promoted, in a paracrine manner, CaMKII phosphorylation in murine HL-1 cardiomyocytes, and ADAMTS13 acted to inhibit TSP1-induced CaMKII activation. In conclusion, the deficiency of ADAMTS13 may underlie the onset of lethal arrhythmias in diabetes through increased CaMKII phosphorylation in cardiomyocytes. Our findings disclose a novel function for ADAMTS13 beyond its antithrombotic activity., (© 2018 by the American Diabetes Association.)- Published
- 2018
- Full Text
- View/download PDF
36. Soluble stroma-related biomarkers of pancreatic cancer.
- Author
-
Resovi A, Bani MR, Porcu L, Anastasia A, Minoli L, Allavena P, Cappello P, Novelli F, Scarpa A, Morandi E, Falanga A, Torri V, Taraboletti G, Belotti D, and Giavazzi R
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Carcinoma, Pancreatic Ductal blood, Carcinoma, Pancreatic Ductal drug therapy, Cohort Studies, Connective Tissue Growth Factor biosynthesis, Connective Tissue Growth Factor blood, Female, Humans, Intercellular Adhesion Molecule-1 biosynthesis, Intercellular Adhesion Molecule-1 blood, Male, Matrix Metalloproteinase 7 biosynthesis, Matrix Metalloproteinase 7 blood, Middle Aged, Pancreatic Neoplasms blood, Pancreatic Neoplasms drug therapy, Prognosis, Solubility, Stromal Cells metabolism, Thrombospondins biosynthesis, Thrombospondins blood, Tissue Inhibitor of Metalloproteinase-1 biosynthesis, Tissue Inhibitor of Metalloproteinase-1 blood, Tumor Microenvironment physiology, Biomarkers, Tumor blood, Carcinoma, Pancreatic Ductal diagnosis, Pancreatic Neoplasms diagnosis
- Abstract
The clinical management of pancreatic ductal adenocarcinoma (PDAC) is hampered by the lack of reliable biomarkers. This study investigated the value of soluble stroma-related molecules as PDAC biomarkers. In the first exploratory phase, 12 out of 38 molecules were associated with PDAC in a cohort of 25 PDAC patients and 16 healthy subjects. A second confirmatory phase on an independent cohort of 131 PDAC patients, 30 chronic pancreatitis patients, and 131 healthy subjects confirmed the PDAC association for MMP7, CCN2, IGFBP2, TSP2, sICAM1, TIMP1, and PLG Multivariable logistic regression model identified biomarker panels discriminating respectively PDAC versus healthy subjects (MMP7 + CA19.9, AUC = 0.99, 99% CI = 0.98-1.00) (CCN2 + CA19.9, AUC = 0.96, 99% CI = 0.92-0.99) and PDAC versus chronic pancreatitis (CCN2 + PLG+FN+Col4 + CA19.9, AUC = 0.94, 99% CI = 0.88-0.99). Five molecules were associated with PanIN development in two GEM models of PDAC (PdxCre/LSL-Kras
G12D and PdxCre/LSL-KrasG12D/+ /LSL-Trp53R172H/+ ), suggesting their potential for detecting early disease. These markers were also elevated in patient-derived orthotopic PDAC xenografts and associated with response to chemotherapy. The identified stroma-related soluble biomarkers represent potential tools for PDAC diagnosis and for monitoring treatment response of PDAC patients., (© 2018 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2018
- Full Text
- View/download PDF
37. Snail levels control the migration mechanism of mesenchymal tumor cells.
- Author
-
Belgiovine C, Chiesa G, Chiodi I, Frapolli R, Bonezzi K, Taraboletti G, D'Incalci M, and Mondello C
- Abstract
Cancer cells use two major types of movement: Mesenchymal, which is typical of cells of mesenchymal origin and depends on matrix metalloproteinase (MMP) activity, and amoeboid, which is characteristic of cells with a rounded shape and relies on the activity of Rho-associated kinase (ROCK). The present authors previously demonstrated that, during neoplastic transformation, telomerase-immortalized human fibroblasts (cen3tel cells) acquired a ROCK-dependent/MMP independent mechanism of invasion, mediated by the downregulation of the ROCK cellular inhibitor Round (Rnd)3/RhoE. In the present study, cen3tel transformation was also demonstrated to be paralleled by downregulation of Snail, a major determinant of the mesenchymal movement. To test whether Snail levels could determine the type of movement adopted by mesenchymal tumor cells, Snail was ectopically expressed in tumorigenic cells. It was observed that ectopic Snail did not increase the levels of typical mesenchymal markers, but induced cells to adopt an MMP-dependent mechanism of invasion. In cells expressing ectopic Snail, invasion became sensitive to the MMP inhibitor Ro 28-2653 and insensitive to the ROCK inhibitor Y27632, suggesting that, once induced by Snail, the mesenchymal movement prevails over the amoeboid one. Snail-expressing cells had a more aggressive behavior in vivo , and exhibited increased tumor growth rate and metastatic ability. These results confirm the high plasticity of cancer cells, which can adopt different types of movement in response to changes in the expression of specific genes. Furthermore, the present findings indicate that Rnd3 and Snail are possible regulators of the type of invasion mechanism adopted by mesenchymal tumor cells.
- Published
- 2016
- Full Text
- View/download PDF
38. Integrating computational and chemical biology tools in the discovery of antiangiogenic small molecule ligands of FGF2 derived from endogenous inhibitors.
- Author
-
Foglieni C, Pagano K, Lessi M, Bugatti A, Moroni E, Pinessi D, Resovi A, Ribatti D, Bertini S, Ragona L, Bellina F, Rusnati M, Colombo G, and Taraboletti G
- Subjects
- Computational Biology, Humans, Ligands, Magnetic Resonance Spectroscopy, Angiogenesis Inhibitors pharmacology, Drug Discovery, Fibroblast Growth Factor 2 metabolism
- Abstract
The FGFs/FGFRs system is a recognized actionable target for therapeutic approaches aimed at inhibiting tumor growth, angiogenesis, metastasis, and resistance to therapy. We previously identified a non-peptidic compound (SM27) that retains the structural and functional properties of the FGF2-binding sequence of thrombospondin-1 (TSP-1), a major endogenous inhibitor of angiogenesis. Here we identified new small molecule inhibitors of FGF2 based on the initial lead. A similarity-based screening of small molecule libraries, followed by docking calculations and experimental studies, allowed selecting 7 bi-naphthalenic compounds that bound FGF2 inhibiting its binding to both heparan sulfate proteoglycans and FGFR-1. The compounds inhibit FGF2 activity in in vitro and ex vivo models of angiogenesis, with improved potency over SM27. Comparative analysis of the selected hits, complemented by NMR and biochemical analysis of 4 newly synthesized functionalized phenylamino-substituted naphthalenes, allowed identifying the minimal stereochemical requirements to improve the design of naphthalene sulfonates as FGF2 inhibitors.
- Published
- 2016
- Full Text
- View/download PDF
39. Vascular endothelial growth factor c promotes ovarian carcinoma progression through paracrine and autocrine mechanisms.
- Author
-
Decio A, Taraboletti G, Patton V, Alzani R, Perego P, Fruscio R, Jürgensmeier JM, Giavazzi R, and Belotti D
- Subjects
- Animals, Carcinoma, Ovarian Epithelial, Disease Progression, Enzyme-Linked Immunosorbent Assay, Female, Heterografts, Humans, Immunohistochemistry, Mice, Mice, Nude, Real-Time Polymerase Chain Reaction, Autocrine Communication physiology, Neoplasms, Glandular and Epithelial metabolism, Neoplasms, Glandular and Epithelial pathology, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Paracrine Communication physiology, Vascular Endothelial Growth Factor C metabolism, Vascular Endothelial Growth Factor Receptor-3 metabolism
- Abstract
Vascular endothelial growth factor C (VEGFC) has been reported to promote tumor progression in several tumor types, mainly through the stimulation of lymphangiogenesis and lymphatic metastasis. However, the expression and biological significance of the VEGFC/VEGF receptor (VEGFR)-3 pathway in ovarian cancer growth and dissemination are unclear, and have been investigated in this study. Soluble VEGFC was detected in the plasma and ascites of patients with ovarian carcinoma, and VEGFR3 expression was found in their tumor tissues. In human ovarian carcinoma xenograft models, high levels of soluble VEGFC in ascites and serum were detected, in association with disease progression, tumor burden, and volume of ascites. Peak VEGFC expression preceded para-aortic lymph node infiltration by HOC8 neoplastic cells. Histological detection of tumor cells in blood and lymphatic vessels indicated both hematogenous and lymphatic dissemination. Overexpression of VEGFC in the VEGFR3-positive and luciferase-expressing IGROV1 cells promoted carcinoma dissemination after orthotopic transplantation in the ovary of immunodeficient mice. In vitro, VEGFC released by the tumor cells stimulated tumor cell migration in an autocrine manner. Cediranib, an inhibitor of VEGFR1-3 and c-kit, inhibited in vivo metastasis of VEGFC-overexpressing IGROV1 and in vitro autocrine effects. These findings suggest that the VEGFC/VEGFR3 pathway acts as an enhancer of ovarian cancer progression through autocrine and paracrine mechanisms, hence offering a potential target for therapy., (Copyright © 2014 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. The tyrosine kinase inhibitor E-3810 combined with paclitaxel inhibits the growth of advanced-stage triple-negative breast cancer xenografts.
- Author
-
Bello E, Taraboletti G, Colella G, Zucchetti M, Forestieri D, Licandro SA, Berndt A, Richter P, D'Incalci M, Cavalletti E, Giavazzi R, Camboni G, and Damia G
- Subjects
- Alanine administration & dosage, Alanine analogs & derivatives, Animals, Cell Line, Tumor, Drug Synergism, Female, Humans, Indoles administration & dosage, Mice, Mice, Nude, Paclitaxel administration & dosage, Paclitaxel pharmacokinetics, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors pharmacokinetics, Pyrroles administration & dosage, Rabeprazole administration & dosage, Rabeprazole pharmacokinetics, Random Allocation, Sunitinib, Triazines administration & dosage, Triple Negative Breast Neoplasms pathology, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Protein Kinase Inhibitors pharmacology, Rabeprazole pharmacology, Triple Negative Breast Neoplasms drug therapy
- Abstract
E-3810 is a novel small molecule that inhibits VEGF receptor-1, -2, and -3 and fibroblast growth factor receptor-1 tyrosine kinases at nmol/L concentrations currently in phase clinical II. In preclinical studies, it had a broad spectrum of antitumor activity when used as monotherapy in a variety of human xenografts. We here investigated the activity of E-3810 combined with different cytotoxic agents in a MDA-MB-231 triple-negative breast cancer xenograft model. The molecule could be safely administered with 5-fluorouracil, cisplatin, and paclitaxel. The E-3810-paclitaxel combination showed a striking activity with complete, lasting tumor regressions; the antitumor activity of the combination was also confirmed in another triple-negative breast xenograft, MX-1. The activity was superior to that of the combinations paclitaxel+brivanib and paclitaxel+sunitinib. Pharmacokinetics studies suggest that the extra antitumor activity of the combination is not due to higher paclitaxel tumor levels, which in fact were lower in mice pretreated with all three kinase inhibitors, and the paclitaxel plasma levels excluded reduced drug availability. Pharmacodynamic studies showed that E-3810, brivanib, and sunitinib given as single agents or in combination with paclitaxel reduced the number of vessels, but did not modify vessel maturation. Reduced tumor collagen IV and increased plasma collagen IV, associated with increased matrix metalloproteinases (MMP), particularly host MMP-9, indicate a proteolytic remodeling of the extracellular matrix caused by E-3810 that in conjunction with the cytotoxic effect of paclitaxel on the tumor cells (caspase-3/7 activity) may contribute to the striking activity of their combination. These data support the therapeutic potential of combining E-3810 with conventional chemotherapy., (©2012 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
41. Inhibition of SIRT2 potentiates the anti-motility activity of taxanes: implications for antineoplastic combination therapies.
- Author
-
Bonezzi K, Belotti D, North BJ, Ghilardi C, Borsotti P, Resovi A, Ubezio P, Riva A, Giavazzi R, Verdin E, and Taraboletti G
- Subjects
- Acetylation drug effects, Cell Line, Tumor, Cell Movement genetics, Gene Silencing, Humans, Microtubules metabolism, Protein Binding, Protein Transport, RNA Interference, Sirtuin 2 genetics, Sirtuin 2 metabolism, Taxoids administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cell Movement drug effects, Sirtuin 2 antagonists & inhibitors, Taxoids pharmacology
- Abstract
Taxanes are potent inhibitors of cell motility, a property implicated in their antiangiogenic and antimetastatic activity and unrelated to their antiproliferative effect. The molecular mechanism of this anti-motility activity is poorly understood. In this study, we found that paclitaxel induced tubulin acetylation in endothelial and tumor cells, at concentrations that affected cell motility but not proliferation (10(-8) to 10(-9) M, for 4 hours). Induction of tubulin acetylation correlated with inhibition of motility but not proliferation based on a comparison of highly and poorly cytotoxic taxanes (paclitaxel and IDN5390) and tumor cell lines sensitive and resistant to paclitaxel (1A9 and 1A9 PTX22). Consistent with the hypothesis that tubulin deacetylase activity might affect cell response to the anti-motility activity of taxanes, we found that overexpression of the tubulin deacetylase SIRT2 increased cell motility and reduced cell response to the anti-motility activity of paclitaxel. Conversely, the SIRT2 inhibitor splitomicin reduced cell motility and potentiated the anti-motility activity of paclitaxel. The inhibitory effect was further potentiated by the addition of the HDAC6 inhibitor trichostatin A. Paclitaxel and splitomicin promoted translocation into the nucleus--and hence activation--of FOXO3a, a negative regulator of cell motility. This study indicates a role for SIRT2 in the regulation of cell motility and suggests that therapies combining sirtuin inhibitors and taxanes could be used to treat cell motility-based pathologic processes such as tumor angiogenesis, invasion, and metastasis.
- Published
- 2012
- Full Text
- View/download PDF
42. Direct and allosteric inhibition of the FGF2/HSPGs/FGFR1 ternary complex formation by an antiangiogenic, thrombospondin-1-mimic small molecule.
- Author
-
Pagano K, Torella R, Foglieni C, Bugatti A, Tomaselli S, Zetta L, Presta M, Rusnati M, Taraboletti G, Colombo G, and Ragona L
- Subjects
- Allosteric Regulation drug effects, Angiogenesis Inhibitors chemistry, Humans, In Vitro Techniques, Models, Molecular, Molecular Dynamics Simulation, Molecular Mimicry, Multiprotein Complexes antagonists & inhibitors, Multiprotein Complexes chemistry, Naphthalenesulfonates chemistry, Naphthalenesulfonates pharmacology, Nuclear Magnetic Resonance, Biomolecular, Protein Interaction Mapping, Surface Plasmon Resonance, Thrombospondin 1 chemistry, Thrombospondin 1 pharmacology, Angiogenesis Inhibitors pharmacology, Fibroblast Growth Factor 2 antagonists & inhibitors, Fibroblast Growth Factor 2 chemistry, Heparan Sulfate Proteoglycans antagonists & inhibitors, Heparan Sulfate Proteoglycans chemistry, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 1 chemistry
- Abstract
Fibroblast growth factors (FGFs) are recognized targets for the development of therapies against angiogenesis-driven diseases, including cancer. The formation of a ternary complex with the transmembrane tyrosine kinase receptors (FGFRs), and heparan sulphate proteoglycans (HSPGs) is required for FGF2 pro-angiogenic activity. Here by using a combination of techniques including Nuclear Magnetic Resonance, Molecular Dynamics, Surface Plasmon Resonance and cell-based binding assays we clarify the molecular mechanism of inhibition of an angiostatic small molecule, sm27, mimicking the endogenous inhibitor of angiogenesis, thrombospondin-1. NMR and MD data demonstrate that sm27 engages the heparin-binding site of FGF2 and induces long-range dynamics perturbations along FGF2/FGFR1 interface regions. The functional consequence of the inhibitor binding is an impaired FGF2 interaction with both its receptors, as demonstrated by SPR and cell-based binding assays. We propose that sm27 antiangiogenic activity is based on a twofold-direct and allosteric-mechanism, inhibiting FGF2 binding to both its receptors.
- Published
- 2012
- Full Text
- View/download PDF
43. Reduced expression of the ROCK inhibitor Rnd3 is associated with increased invasiveness and metastatic potential in mesenchymal tumor cells.
- Author
-
Belgiovine C, Frapolli R, Bonezzi K, Chiodi I, Favero F, Mello-Grand M, Dei Tos AP, Giulotto E, Taraboletti G, D'Incalci M, and Mondello C
- Subjects
- Animals, Blotting, Western, Cell Line, Transformed, Cell Movement genetics, Cell Shape genetics, Female, Fibroblasts metabolism, Fibroblasts pathology, Gene Expression Profiling, Humans, Matrix Metalloproteinases genetics, Matrix Metalloproteinases metabolism, Mesoderm pathology, Mice, Mice, Nude, Mice, SCID, NIH 3T3 Cells, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Oligonucleotide Array Sequence Analysis, Reverse Transcriptase Polymerase Chain Reaction, rho GTP-Binding Proteins metabolism, Gene Expression, Mesoderm metabolism, Neoplasms, Experimental genetics, rho GTP-Binding Proteins genetics
- Abstract
Background: Mesenchymal and amoeboid movements are two important mechanisms adopted by cancer cells to invade the surrounding environment. Mesenchymal movement depends on extracellular matrix protease activity, amoeboid movement on the RhoA-dependent kinase ROCK. Cancer cells can switch from one mechanism to the other in response to different stimuli, limiting the efficacy of antimetastatic therapies., Methodology and Principal Findings: We investigated the acquisition and molecular regulation of the invasion capacity of neoplastically transformed human fibroblasts, which were able to induce sarcomas and metastases when injected into immunocompromised mice. We found that neoplastic transformation was associated with a change in cell morphology (from fibroblastic to polygonal), a reorganization of the actin cytoskeleton, a decrease in the expression of several matrix metalloproteases and increases in cell motility and invasiveness. In a three-dimensional environment, sarcomagenic cells showed a spherical morphology with cortical actin rings, suggesting a switch from mesenchymal to amoeboid movement. Accordingly, cell invasion decreased after treatment with the ROCK inhibitor Y27632, but not with the matrix protease inhibitor Ro 28-2653. The increased invasiveness of tumorigenic cells was associated with reduced expression of Rnd3 (also known as RhoE), a cellular inhibitor of ROCK. Indeed, ectopic Rnd3 expression reduced their invasive ability in vitro and their metastatic potential in vivo., Conclusions: These results indicate that, during neoplastic transformation, cells of mesenchymal origin can switch from a mesenchymal mode of movement to an amoeboid one. In addition, they point to Rnd3 as a possible regulator of mesenchymal tumor cell invasion and to ROCK as a potential therapeutic target for sarcomas.
- Published
- 2010
- Full Text
- View/download PDF
44. Targeting tumor angiogenesis with TSP-1-based compounds: rational design of antiangiogenic mimetics of endogenous inhibitors.
- Author
-
Taraboletti G, Rusnati M, Ragona L, and Colombo G
- Subjects
- Angiogenesis Inhibitors chemistry, Animals, Biomimetics methods, Biomimetics trends, Humans, Models, Biological, Models, Molecular, Neoplasms blood supply, Rationalization, Thrombospondin 1 physiology, Angiogenesis Inhibitors chemical synthesis, Angiogenesis Inhibitors therapeutic use, Drug Design, Molecular Targeted Therapy methods, Neoplasms drug therapy, Neovascularization, Pathologic drug therapy, Thrombospondin 1 chemistry
- Abstract
Inhibitors of angiogenesis are an important addition to conventional chemotherapy. Among different "druggable" angiogenic factors, fibroblast growth factor-2 (FGF-2) is an attractive target for novel therapies because of its intricated involvement in tumor neovascularization, tumor cell proliferation and migration, and the acquisition of resistance to antiangiogenic therapies. FGF-2 bioavailability and activity is affected by several natural ligands, including the endogenous inhibitor of angiogenesis thrombospondin-1 (TSP-1). We hypothesized that the FGF-2-binding sequence of TSP-1 might serve as a template for the development of non-peptide inhibitors of angiogenesis. Computational biology and nuclear magnetic resonance spectroscopy approaches, major investigative tools in the characterizations of protein-protein interaction (PPI), were used to map the residues at the TSP-1/FGF-2 interface. The translation of this three-dimensional information into a pharmacophore model allowed screening a small molecule databases, identifying three FGF-2-binding, antiangiogenic small molecules, mimetic of TSP-1. Pharmacophore-based approaches are thus feasible tools to exploit naturally occurring PPI, by generating a set of lead compounds mimetic of endogenous proteins, as a starting point for the development of novel therapeutic agents.
- Published
- 2010
- Full Text
- View/download PDF
45. Thrombospondin-1 as a Paradigm for the Development of Antiangiogenic Agents Endowed with Multiple Mechanisms of Action.
- Author
-
Rusnati M, Urbinati C, Bonifacio S, Presta M, and Taraboletti G
- Abstract
Uncontrolled neovascularization occurs in several angiogenesis-dependent diseases, including cancer. Neovascularization is tightly controlled by the balance between angiogenic growth factors and antiangiogenic agents. The various natural angiogenesis inhibitors identified so far affect neovascularization by different mechanisms of action. Thrombospondin-1 (TSP-1) is a matricellular modular glycoprotein that acts as a powerful endogenous inhibitor of angiogenesis. It acts both indirectly, by sequestering angiogenic growth factors and effectors in the extracellular environment, and directly, by inducing an antiangiogenic program in endothelial cells following engagement of specific receptors including CD36, CD47, integrins and proteoglycans (all involved in angiogenesis ). In view of its central, multifaceted role in angiogenesis, TSP-1 has served as a source of antiangiogenic tools, including TSP-1 fragments, synthetic peptides and peptidomimetics, gene therapy strategies, and agents that up-regulate TSP-1 expression. This review discusses TSP-1-based inhibitors of angiogenesis, their mechanisms of action and therapeutic potential, drawing our experience with angiogenic growth factor-interacting TSP-1 peptides, and the possibility of exploiting them to design novel antiangiogenic agents.
- Published
- 2010
- Full Text
- View/download PDF
46. Cathepsin B mediates the pH-dependent proinvasive activity of tumor-shed microvesicles.
- Author
-
Giusti I, D'Ascenzo S, Millimaggi D, Taraboletti G, Carta G, Franceschini N, Pavan A, and Dolo V
- Subjects
- Cathepsin B genetics, Cell Line, Tumor, Cell Movement, Culture Media, Conditioned pharmacology, Enzyme Activation, Female, Humans, Hydrogen-Ion Concentration, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Neoplasm Invasiveness, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering pharmacology, Reverse Transcriptase Polymerase Chain Reaction, Cathepsin B metabolism, Membrane Microdomains metabolism, Membrane Microdomains pathology, Ovarian Neoplasms enzymology
- Abstract
Vesicles shed by cancer cells are known to mediate several tumor-host interactions. Tumor microenvironment may, in turn, influence the release and the activity of tumor-shed microvesicles. In this study, we investigated the molecular mediators of the pH-dependent proinvasive activity of tumor-shed vesicles. Gelatinase zymography showed increased microvesicle activity of matrix metalloproteinases 9 and 2 as a result of acid exposure (pH 5.6) compared to pH 7.4. Thus, we reasoned that the cysteine protease cathepsin B might play a role in mediating the pH-dependent activation of gelatinases. Cathepsin B expression in tumor-shed microvesicles was confirmed by Western blot analysis and zymography. The activity of vesicle-associated cathepsin B measured using Z-Arg-Arg-pNA as substrate was significantly increased at acidic pH values. Inhibition of protease activity by the cysteine protease inhibitor, E-64, and treatment of ovarian cancer cells with small interfering RNA against cathepsin B suppressed the ability of tumor-shed microvesicles to stimulate both gelatinase activation and the invasiveness of endothelial cells observed at low pH values. We conclude that microvesicle shedding is a major secretory pathway for cathepsin B release from tumor cells. Hence, the acidic microenvironment found in most solid tumors may contribute to cathepsin B-mediated proinvasive capabilities of tumor-shed vesicles.
- Published
- 2008
- Full Text
- View/download PDF
47. Vascular endothelial growth factor stimulates organ-specific host matrix metalloproteinase-9 expression and ovarian cancer invasion.
- Author
-
Belotti D, Calcagno C, Garofalo A, Caronia D, Riccardi E, Giavazzi R, and Taraboletti G
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Bevacizumab, Culture Media, Conditioned, Endothelial Cells drug effects, Endothelial Cells enzymology, Endothelial Cells pathology, Female, Humans, Immunohistochemistry, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Transplantation, Organ Specificity drug effects, Ovary drug effects, Ovary enzymology, Ovary pathology, Vascular Endothelial Growth Factor A metabolism, Matrix Metalloproteinase 9 metabolism, Ovarian Neoplasms enzymology, Ovarian Neoplasms pathology, Vascular Endothelial Growth Factor A pharmacology
- Abstract
Vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMP) regulate each other, contributing to tumor progression. We have previously reported that MMP9 induces the release of tumor VEGF, promoting ascites formation in human ovarian carcinoma xenografts. The aim of this study was to investigate whether tumor-derived VEGF regulated the expression of gelatinase by the stroma, influencing the invasive properties of ovarian tumors. Tumor variants derived from 1A9 human ovarian carcinoma, stably expressing VEGF(121) in the sense (1A9-VS-1) and antisense orientations (1A9-VAS-3), were used. In vivo, zymographic analysis of tumors from 1A9-VS-1 implanted in the peritoneal cavity of nude mice showed higher levels of gelatinases, particularly murine MMP9, indicating that VEGF stimulates host expression of the matrix-degrading enzyme. Murine MMP9 expression was also high in the ovaries of mice bearing 1A9-VS-1 tumors. The effect on host MMP9 activity was organ-specific. The levels of host pro-MMP9 in ovaries correlated with the plasma levels of tumor VEGF and with the selective invasion of the ovaries. Induction of host MMP9 expression in tumors and ovaries was independent of the site of tumor growth as it was seen in mice carrying both intraperitoneal and subcutaneous tumors. The anti-VEGF antibody bevacizumab (Avastin) inhibited MMP9 expression and tumor invasion in the ovaries of mice bearing 1A9-VS-1 tumors. These findings point to a complex cross-talk between VEGF and MMPs in the progression of ovarian tumor and suggest the possibility of using VEGF inhibitors to affect MMP-dependent tumor invasion.
- Published
- 2008
- Full Text
- View/download PDF
48. Bioavailability of VEGF in tumor-shed vesicles depends on vesicle burst induced by acidic pH.
- Author
-
Taraboletti G, D'Ascenzo S, Giusti I, Marchetti D, Borsotti P, Millimaggi D, Giavazzi R, Pavan A, and Dolo V
- Subjects
- Animals, Biological Availability, Cattle, Cell Line, Tumor, Cell Movement, Cells, Cultured, Culture Media, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Endothelium, Vascular physiopathology, Female, Humans, Neoplasm Invasiveness, Umbilical Veins, Vascular Endothelial Growth Factor A metabolism, Endothelium, Vascular pathology, Hydrogen-Ion Concentration, Neovascularization, Pathologic pathology, Ovarian Neoplasms blood supply, Ovarian Neoplasms pathology, Vascular Endothelial Growth Factor A pharmacokinetics
- Abstract
Tumor angiogenesis is regulated by a dynamic cross-talk between tumor cells and the host microenvironment. Because membrane vesicles shed by tumor cells are known to mediate several tumor-host interactions, we determined whether vesicles might also stimulate angiogenesis. Vesicles shed by human ovarian carcinoma cell lines CABA I and A2780 stimulated the motility and invasiveness of endothelial cells in vitro. Enzyme-linked immunosorbent assay and Western blot analysis revealed relevant amounts of vascular endothelial growth factor (VEGF) and the two matrix metalloproteinases MMP-2 and MMP-9, but not fibroblast growth factor-2, contained in shed vesicles. An A2780 cell-derived clone transfected to overexpress VEGF shed the same amount of vesicles as did a control clone, but contained significantly more VEGF within the vesicles. Despite a greater amount of VEGF in vesicles of the overexpressing clone, vesicles of both clones stimulated endothelial cell motility to comparable levels, suggesting that VEGF was stored within the vesicle and was unavailable. Only following vesicle burst induced by acidic pH (a characteristic of the tumor microenvironment) was VEGF released, leading to significantly higher stimulation of cell motility. Thus, tumor-shed membrane vesicles carry VEGF and release it in a bioactive form in conditions typical of the tumor microenvironment.
- Published
- 2006
- Full Text
- View/download PDF
49. Gorham-Stout syndrome: a monocyte-mediated cytokine propelled disease.
- Author
-
Colucci S, Taraboletti G, Primo L, Viale A, Roca C, Valdembri D, Geuna M, Pagano M, Grano M, Pogrel AM, Harris AL, Athanasou NN, Mantovani A, Zallone A, and Bussolino F
- Subjects
- Animals, Antigens, Surface analysis, Bone and Bones immunology, Bone and Bones pathology, Cell Adhesion, Cells, Cultured, Female, Humans, Mice, Mice, Inbred Strains, Neovascularization, Pathologic, Syndrome, Cell Movement, Cytokines metabolism, Monocytes immunology, Osteoclasts cytology, Osteolysis, Essential immunology, Osteolysis, Essential pathology
- Abstract
Unlabelled: We studied the biological features and the immunophenotype of a cell culture established from the lesion of soft tissues of a woman affected by Gorham-Stout syndrome. We found that these cells belonged to a monocytic lineage with some characteristics of immature osteoclasts and were able to release large amounts of osteoclastogenic and angiogenic molecules that may contribute to disease progression., Introduction: Gorham-Stout syndrome is a rare disease characterized by osteolysis and proliferation of vascular or lymphatic vessels, with a severe outcome. Its etiology and the identification of the cell types involved are completely unknown., Materials and Methods: A cell culture from a lesion of soft tissues was established, and its behavior in vitro and in immunodeficient mice was studied. We analyzed (1) the cell phenotype by flow cytometry; (2) the adhesive and migratory properties on different substrates; (3) the ability to differentiate into mature osteoclasts; (4) the production of osteclastogenic and angiogenic molecules; (5) the in vivo angiogenic activity of the cells subcutaneously implanted in mouse in a Matrigel plug; and (6) the ability to recapitulate the disease when transplanted in nude mice., Results and Conclusions: The established culture consisted of a morphologically homogeneous cell population belonging to a monocytic lineage having some features of an osteoclast-like cell type. Cells had an invasive phenotype, were angiogenic, and produced osteoclastogenic (IL-6, TGF-beta1, IL-1beta) and angiogenic (vascular endothelial growth factor-A [VEGF-A], CXCL-8) molecules when challenged with inflammatory cytokines. Immunodeficient mice injected with these cells did not show any bone lesions or vascular alteration, but had high amounts of circulating human IL-6 and VEGF-A. Cells isolated from a cutaneous lymphangiomatosis did not show any of these findings. These data suggest that cells of monocyte-macrophage lineage play an essential role in the pathogenesis of Gorham-Stout disease, whose progression is propelled by cytokine circuits that accelerate angiogenesis and osteoclastogenesis.
- Published
- 2006
- Full Text
- View/download PDF
50. Potential antagonism of tubulin-binding anticancer agents in combination therapies.
- Author
-
Taraboletti G, Micheletti G, Dossi R, Borsotti P, Martinelli M, Fiordaliso F, Ryan AJ, and Giavazzi R
- Subjects
- Angiogenesis Inhibitors administration & dosage, Angiogenesis Inhibitors pharmacology, Animals, Cell Adhesion drug effects, Cell Line, Cell Line, Tumor, Cell Proliferation drug effects, Cisplatin administration & dosage, Cisplatin pharmacology, Colchicine administration & dosage, Colchicine metabolism, Cytoskeleton drug effects, Cytoskeleton metabolism, Docetaxel, Dose-Response Relationship, Drug, Doxorubicin administration & dosage, Doxorubicin pharmacology, Drug Interactions, Endothelial Cells drug effects, Endothelial Cells metabolism, Female, Humans, Mice, Mice, Inbred C57BL, Mice, Nude, Microscopy, Fluorescence, Necrosis prevention & control, Organophosphorus Compounds administration & dosage, Organophosphorus Compounds pharmacology, Paclitaxel administration & dosage, Paclitaxel pharmacology, Protein Binding, Taxoids administration & dosage, Taxoids pharmacology, Umbilical Veins cytology, Vincristine administration & dosage, Vincristine pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colchicine analogs & derivatives, Colchicine pharmacology, Tubulin metabolism, Xenograft Model Antitumor Assays methods
- Abstract
ZD6126 is a vascular targeting agent, developed for the treatment of solid tumors. In vivo, ZD6126 is rapidly converted into the tubulin-binding agent N-acetylcolchinol. We have previously reported that in vitro N-acetylcolchinol disrupts microtubules and induces rapid changes in endothelial cell morphology, which in a tumor would lead to a rapid loss of tumor vessel integrity and subsequent extensive tumor necrosis. The aim of this study was to investigate the effect of cytotoxic antineoplastic drugs-cisplatin, doxorubicin, vincristine, paclitaxel, and docetaxel-on endothelial cell response to N-acetylcolchinol. We found that cisplatin and doxorubicin did not interfere with the ability of N-acetylcolchinol to cause morphologic changes in human umbilical vein endothelial cells, whereas vincristine showed additive effects. In contrast, the microtubule-stabilizing agents paclitaxel (1-10 micromol/L) and docetaxel (0.1-1 micromol/L) prevented the morphologic changes induced by N-acetylcolchinol in human umbilical vein endothelial cells. The effect was observed when cells were exposed to paclitaxel and N-acetylcolchinol together or when paclitaxel was given shortly before N-acetylcolchinol. Paclitaxel and N-acetylcolchinol interacted at the level of microtubule organization, as shown in immunofluorescence analysis of the cytoskeleton. The protective effect was reversible because 4 hours after paclitaxel wash out, cells recovered the sensitivity to N-acetylcolchinol. In vivo, pretreatment of mice with paclitaxel inhibited the vascular targeting activity of ZD6126 on newly formed vessels in the Matrigel plug assay and ZD6126-induced necrosis in tumors. These findings indicate that paclitaxel, depending on the timing and schedule of administration, can affect the vascular targeting activity of ZD6126, which may have an effect on the optimal scheduling of therapies based on the combined use of microtubule-stabilizing and microtubule-destabilizing agents.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.