73 results on '"Coltrini D"'
Search Results
2. P2Y1 and P2Y2 Receptor-Operated Ca2+ Signals in Primary Cultures of Cardiac Microvascular Endothelial Cells
- Author
-
Moccia, F., Baruffi, S., Spaggiari, S., Coltrini, D., Berra-Romani, R., Signorelli, S., Castelli, L., Taglietti, V., and Tanzi, F.
- Published
- 2001
- Full Text
- View/download PDF
3. Role of cannabinoids in a multistage murine model of prostate carcinoma to investigate potential clinical applications
- Author
-
Ligresti A, Ronca R, Marolda V, Coltrini D, and Di Marzo V
- Subjects
Prostate carcinoma ,Cannabinoids - Published
- 2016
4. 307 Stromal delivery of long Pentraxin-3 impairs FGF/FGFR-dependent tumor growth and metastasis
- Author
-
Giacomini, A., primary, Di Salle, E., additional, Coltrini, D., additional, Rezzola, S., additional, Belleri, M., additional, Presta, M., additional, and Ronca, R., additional
- Published
- 2014
- Full Text
- View/download PDF
5. Long pentraxin 3/tumor necrosis factor-stimulated gene-6 interaction a biological rheostat for fibroblast growth factor 2-mediated angiogenesis
- Author
-
Leali, D., Inforzato, A., Ronca, R., Bianchi, R., Belleri, M., Coltrini, D., Di Salle, E., Sironi, M., Norata, Giuseppe, Bottazzi, B., Garlanda, C., Day, A., Presta, M., Leali, D., Inforzato, A., Ronca, R., Bianchi, R., Belleri, M., Coltrini, D., Di Salle, E., Sironi, M., Norata, Giuseppe, Bottazzi, B., Garlanda, C., Day, A., and Presta, M.
- Abstract
Objective-Angiogenesis is regulated by the balance between pro-and antiangiogenic factors and by extracellular matrix protein interactions. Fibroblast growth factor 2 (FGF2) is a major proangiogenic inducer inhibited by the interaction with the soluble pattern recognition receptor long pentraxin 3 (PTX3). PTX3 is locally coexpressed with its ligand tumor necrosis factor-stimulated gene-6 (TSG-6), a secreted glycoprotein that cooperates with PTX3 in extracellular matrix assembly. Here, we characterized the effect of TSG-6 on PTX3/FGF2 interaction and FGF2-mediated angiogenesis. Methods and Results-Solid phase binding and surface plasmon resonance assays show that TSG-6 and FGF2 bind the PTX3 N-terminal domain with similar affinity. Accordingly, TSG-6 prevents FGF2/PTX3 interaction and suppresses the inhibition exerted by PTX3 on heparan sulfate proteoglycan/FGF2/FGF receptor complex formation and on FGF2-dependent angiogenesis in vitro and in vivo. Also, endogenous PTX3 exerts an inhibitory effect on vascularization induced by FGF2 in a murine subcutaneous Matrigel plug assay, the inhibition being abolished in Ptx3-null mice or by TSG-6 treatment in wild-type animals. Conclusion-TSG-6 reverts the inhibitory effects exerted by PTX3 on FGF2-mediated angiogenesis through competition of FGF2/PTX3 interaction. This may provide a novel mechanism to control angiogenesis in those pathological settings characterized by the coexpression of TSG-6 and PTX3, in which the relative levels of these proteins may fine-tune the angiogenic activity of FGF2. © 2012 American Heart Association, Inc.
- Published
- 2012
6. Impact of fibroblast growth factor-2 on tumor microvascular architecture. A tridimensional morphometric study
- Author
-
Konerding, M. A., Fait, E., Dimitropoulou, C., Malkusch, W., Ferri, C., Giavazzi, R., Coltrini, D., and Marco PRESTA
- Subjects
Microcirculation ,Mice, Nude ,Adenocarcinoma ,Corrosion Casting ,Transfection ,Endometrial Neoplasms ,Specific Pathogen-Free Organisms ,Mice ,Tumor Cells, Cultured ,Animals ,Humans ,Female ,Fibroblast Growth Factor 2 ,Neoplasm Transplantation ,Research Article - Abstract
Three cell clones originated by transfection of human endometrial adenocarcinoma HEC-1-B cells with fibroblast growth factor-2 (FGF-2) cDNA and characterized by a different capacity to produce and secrete the growth factor were transplanted subcutaneously in nude mice. Corrosion casting of the tumor microvasculature of xenografts produced by injection of 2 x 10(6) or 10 x 10(6) FGF-2-B9 cells (which produce and secrete significant amounts of FGF-2), 10 x 10(6) FGF-2-A8 cells (which produce comparable amounts of FGF-2 but do not secrete it), or 10 x 10(6) control FGF-2-B8 cells (which produce only trace amounts of FGF-2) was performed after 14 days of growth. Interbranching distances, intervascular distances, branching angles, and vessel diameters were then determined using tridimensional stereo pairs of the casted tumor vascularity. When transplanted at the same concentration, FGF-2-B9 cells grew faster in nude mice compared with FGF-2-A8 and FGF-2-B8 clones. The total amount of new vessel formation was far higher in FGF-2-B9 tumors than in FGF-2-B8 or FGF-2-A8 tumors. Also, vessel courses were more irregular and blind-ending vessels and evasates were more frequent in FGF-2-B9 tumors. Moreover, FGF-2-B9 tumor microvasculature was characterized by a wider average vascular diameter and by an extreme variability of the diameter of each individual vessel along its course between two ramifications. No statistical differences were observed when the distribution curves of the values of intervascular distances, interbranching distances, and branching angles of the microvessel network were compared among the different experimental groups. The distinctive features of the microvasculature of FGF-2-B9 tumors were retained, at least in part, in the smaller lesions produced by injection of a limited number of cells. The data indicate that FGF-2 production and release confer to FGF-2-B9 cells the ability to stimulate the formation of new blood vessels with distinctive architectural features. Neovascularization of FGF-2-B9 lesions parallels the faster rate of growth of the neoplastic parenchyma. This does not affect the overall architecture of the microvessel network that appears to be primed by characteristics of the HEC-1-B tumor cell line and/or by the microenvironment of the host. To our knowledge, this work represents the first attempt to define the influence of a single, defined growth factor on the tridimensional tumor vascular pattern.
- Published
- 1998
7. The bone morphogenic protein antagonist Drm/gremlin is a novel pro- angiogenic factor
- Author
-
Stabile, H, Mitola, S, Moroni, E, Belleri, M, Nicoli, S, Coltrini, D, Peri, F, Pessi, A, Orsatti, L, Talamo, F, Castronovo, V, Waltregny, D, Cotelli, F, Ribatti, D, Marco, P, Marco Presta, PERI, FRANCESCO, Stabile, H, Mitola, S, Moroni, E, Belleri, M, Nicoli, S, Coltrini, D, Peri, F, Pessi, A, Orsatti, L, Talamo, F, Castronovo, V, Waltregny, D, Cotelli, F, Ribatti, D, Marco, P, Marco Presta, and PERI, FRANCESCO
- Abstract
Angiogenesis plays a key role in various physiological and pathological conditions, including tumor growth. Drm/gremlin, a member the Dan family of bone morphogenic protein (BMP) antagonists, is commonly thought to affect different processes during growth, differentiation, and development by heterodimerizing various BMPs. Here we identify Drm/gremlin as a novel pro-angiogenic factor expressed by endothelium. Indeed, Drm/gremlin was purified to homogeneity from the conditioned medium of transformed endothelial cells using an endothelial cell sprouting assay to follow protein isolation. Accordingly, recombinant Drm/gremlin stimulates endothelial cell migration and invasion in fibrin and collagen gels, binds with high-affinity to various endothelial cell types, and triggers tyrosine phosphorylation of intracellular signaling proteins. Also, Drm/gremlin induces neovascularization in the chick embryo chorioallantoic membrane. BMP4 does not affect Drm/gremlin interaction with endothelium and both molecules exert a pro-angiogenic activity in vitro and in vivo when administered alone or in combination. Finally, Drm/gremlin is produced by the stroma of human tumor xenografts in nude mice and it is highly expressed in endothelial cells of human lung tumor vasculature when compared to non-neoplastic lung. Our observations point to a novel, previously unrecognized capacity of Drm/gremlin to interact directly with target endothelial cells and to modulate angiogenesis.
- Published
- 2007
8. Interaction of HIV-1 Tat protein with heparin. Role of the backbone structure, sulfation, and size
- Author
-
Rusnati, M, Coltrini, D, Oreste, P, Zoppetti, G, Albini, A, Noonan, Douglas, Difagagna, Fd, Giacca, M, Presta, M., M., Rusnati, D., Coltrini, P., Oreste, G., Zoppetti, A., Albini, D., Noonan, F., d'Adda di Fagagna, Giacca, Mauro, and M., Presta
- Subjects
Transcriptional Activation ,Recombinant Fusion Proteins ,genetics/metabolism ,Binding Sites, Gene Products ,tat ,genetics/metabolism, Glycosaminoglycans ,chemistry/metabolism/pharmacology, HIV Long Terminal Repeat, HIV-1, Heparin ,chemistry/metabolism/pharmacology, Heparitin Sulfate ,chemistry/metabolism/pharmacology, Protein Binding, Recombinant Fusion Proteins ,metabolism, Structure-Activity Relationship, Sulfuric Acid Esters ,chemistry, Transcriptional Activation ,drug effects, tat Gene Products ,Human Immunodeficiency Virus ,Sulfuric Acid Esters ,chemistry ,Structure-Activity Relationship ,metabolism, Structure-Activity Relationship, Sulfuric Acid Ester ,Gene Products ,chemistry/metabolism/pharmacology ,Binding Sites, Gene Product ,Glycosaminoglycans ,HIV Long Terminal Repeat ,Binding Sites ,Heparin ,drug effects, tat Gene Product ,drug effects ,genetics/metabolism, Glycosaminoglycan ,Gene Products, tat ,HIV-1 ,tat Gene Products, Human Immunodeficiency Virus ,Heparitin Sulfate ,tat Gene Products ,metabolism ,chemistry/metabolism/pharmacology, Protein Binding, Recombinant Fusion Protein ,Protein Binding - Abstract
Human immunodeficiency virus type 1 (HIV-1) Tat protein is released from infected cells. Extracellular Tat enters the cell where it stimulates the transcriptional activity of HIV-long terminal repeat (LTR) and of endogenous genes. Heparin modulates the angiogenic (Albini, A., Benelli, R., Presta, M., Rusnati, M., Ziche, M., Rubartelli, A., Paglialunga, G., Bussolino, F., and Noonan, D. (1996) Oncogene 12, 289-297) and transcriptional (Mann, D. A., and Frankel, A. D. (1991) EMBO J. 10, 1733-1739) activity of extracellular Tat. Here we demonstrate that heparin binds specifically to recombinant HIV-1 Tat produced as glutathione S-transferase (GST) fusion protein and immobilized on glutathione-agarose beads. Heparin and heparan sulfate (HS), but not dermatan sulfate, chondroitin sulfates A and C, hyaluronic acid, and K5 polysaccharide, competed with 3H-labeled heparin for binding to immobilized GST-Tat and inhibited HIV-LTR transactivation induced by extracellular GST-Tat. Selective 2-O-, 6-O-, total-O-desulfation, or N-desulfation/N-acetylation dramatically reduced the capacity of heparin to bind GST-Tat. Totally-O-desulfated and 2-O-desulfated heparins also showed a reduced capacity to inhibit the transactivating activity of GST-Tat. Very low molecular weight heparins showed a significant decrease in their capacity to bind GST-Tat and to inhibit its LTR transactivating activity when compared with conventional 13.6-kDa heparin. However, when 3.0-kDa heparin was affinity chromatographed on immobilized GST-Tat to isolate binding and non-binding subfractions, the Tat-bound fraction was >/=1,000 times more potent than the unbound fraction in inhibiting the transactivating activity of GST-Tat. The results demonstrate that Tat interacts in a size-dependent manner with heparin/HS and that high affinity Tat-heparin interaction requires at least some 2-O-, 6-O-, and N-positions to be sulfated. The Tat binding activity of the glycosaminoglycans tested correlates with their capacity to affect the transactivating activity of extracellular Tat, indicating the possibility to design specific heparin/HS-like structures with Tat-antagonist activity.
- Published
- 1997
9. Growth advantage and vascularization induced by basic fibroblast growth factor overexpression in endometrial HEC-1-B cells: an export-dependent mechanism of action
- Author
-
Coltrini D, Gualandris A, Ee, Nelli, Parolini S, Mp, Molinari-Tosatti, Quarto N, Ziche M, Raffaella Giavazzi, and Presta M
- Subjects
Cultured ,Neovascularization, Pathologic ,Gene Expression ,Mice, Nude ,Neoplasms, Experimental ,In Vitro Techniques ,Adenocarcinoma ,pathology, Female, Fibroblast Growth Factor 2 ,pathology, Neovascularization ,Cultured, Urokinase-Type Plasminogen Activator ,Transfection ,Urokinase-Type Plasminogen Activator ,Endometrial Neoplasms ,Adenocarcinoma, Papillary ,Mice ,Tumor Cells, Cultured ,Animals ,Humans ,pathology ,Female ,Fibroblast Growth Factor 2 ,RNA, Messenger ,Cell Division ,Neoplasm Transplantation ,Neovascularization ,Aged - Abstract
The human endometrial adenocarcinoma HEC-1-B cell line was transfected with an expression vector harboring the human basic fibroblast growth factor (bFGF) cDNA under the control of the human beta-actin gene promoter. Stable transfectants were obtained in which a constitutive, limited overexpression of M(r) 24,000, 22,000, and 18,000 bFGF isoforms was observed. When transfectants were screened for the capacity to release the growth factor, significant amounts of bFGF were present in the conditioned medium and extracellular matrix of the bFGF-B9 clone but not of the bFGF-A8 clone, even though both cell lines produced similar levels of intracellular bFGF. When compared to parental cells, bFGF-B9 cells showed down-regulation of tyrosine kinase fibroblast growth factor receptors along with up-regulation of urokinase-type plasminogen activator expression which was abolished by incubation of the cell cultures with neutralizing anti-bFGF antibody. In vivo, bFGF-B9 cells formed highly vascularized tumors growing faster than parental cells when injected s.c. in nude mice. Also, they were more potent than nontransfected cells in inducing an angiogenic response in the rabbit cornea assay. In contrast, the bFGF-A8 cell phenotype was indistinguishable from parental cells both in vitro and in vivo. In conclusion, clonal differences exist within the HEC-1-B cell line in the capacity to release bFGF. bFGF export by human endometrial adenocarcinoma cells results in autocrine and paracrine effects that confer a growth advantage in vivo associated with increased neovascularization.
- Published
- 1995
10. Fibroblast Growth Factor-2 Antagonist and Antiangiogenic Activity of Long-Pentraxin 3-Derived Synthetic Peptides
- Author
-
Leali, D., primary, Alessi, P., additional, Coltrini, D., additional, Rusnati, M., additional, Zetta, L., additional, and Presta, M., additional
- Published
- 2009
- Full Text
- View/download PDF
11. Role of endothelial cell extracellular signal-regulated kinase1/2 in urokinase-type plasminogen activator upregulation and in vitro angiogenesis by fibroblast growth factor-2
- Author
-
Giuliani, R., primary, Bastaki, M., additional, Coltrini, D., additional, and Presta, M., additional
- Published
- 1999
- Full Text
- View/download PDF
12. Different effects of mucosal, bovine lung and chemically modified heparin on selected biological properties of basic fibroblast growth factor
- Author
-
Coltrini, D, primary, Rusnati, M, additional, Zoppetti, G, additional, Oreste, P, additional, Grazioli, G, additional, Naggi, A, additional, and Presta, M, additional
- Published
- 1994
- Full Text
- View/download PDF
13. Distinct Role of 2-O-, N-, and 6-O-Sulfate Groups of Heparin in the Formation of the Ternary Complex with Basic Fibroblast Growth Factor and Soluble FGF Receptor-1
- Author
-
Rusnati, M., primary, Coltrini, D., additional, Caccia, P., additional, Dellera, P., additional, Zoppetti, G., additional, Oreste, P., additional, Valsasina, B., additional, and Presta, M., additional
- Published
- 1994
- Full Text
- View/download PDF
14. P2Y1and P2Y2Receptor-Operated Ca2+Signals in Primary Cultures of Cardiac Microvascular Endothelial Cells
- Author
-
Moccia, F., Baruffi, S., Spaggiari, S., Coltrini, D., Berra-Romani, R., Signorelli, S., Castelli, L., Taglietti, V., and Tanzi, F.
- Abstract
Intracellular Ca2+signals elicited by nucleotide agonists were investigated in primary cultures of rat cardiac microvascular endothelial cells using the fura-2 technique. UTP increased the intracellular [Ca2+] in 94% of the cells, whereas 2MeSATP was active in 32%. The rank order of potency was ATP = UTP > 2MeSATP and the maximal response to 2MeSATP was lower compared to UTP and ATP. ATP and UTP showed strong homologous and heterologous desensitization. ATP fully inhibited the 2MeSATP response, while UTP abolished 2MeSATP-elicited transients in 25% of cells. 2MeSATP did not desensitize the UTP or ATP response. Adenosine 2′,5′-diphosphate inhibited the response to 2MeSATP, while it did not modify the response to ATP and UTP. 2MeSATP was more sensitive to suramin than UTP and ATP. These results indicate that P2Y1and P2Y2receptors may be coexpressed in CMEC. Nucleotide-induced Ca2+signals lacked a sustained plateau and were almost independent from extracellular Ca2+. ATP and UTP elicited Ca2+transients longer than 2MeSATP-evoked transients. The kinetics of Ca2+responses was not affected by bath solution stirring or ectonucleotidase inhibition. Furthermore, the nonhydrolyzable ATP analogue AMP-PNP induced Ca2+signals similar to those elicited by ATP and UTP. These results suggest that the distinct kinetics of nucleotide-evoked Ca2+responses do not depend on the activity of ectonucleotidases or ATP autocrine stimulation. The possibility that Ca2+signals with different time courses may modulate different cellular responses is discussed.
- Published
- 2001
- Full Text
- View/download PDF
15. Endothelial cells overexpressing basic fibroblast growth factor (FGF-2) induce vascular tumors in immunodeficient mice
- Author
-
Sola, F., Gualandris, A., Belleri, M., Giuliani, R., Coltrini, D., Bastaki, M., Tosatti, M. P. M., Bonardi, F., Vecchi, A., Fioretti, F., Giavazzi, R., Ciomei, M., Grandi, M., Alberto Mantovani, and Presta, M.
- Subjects
angiogenesis ,angiostatic compounds ,endothelium ,fibroblast growth factor ,immunity ,vascular tumors
16. Pentraxin 3 inhibits fibroblast growth factor 2-dependent activation of smooth muscle cells in vitro and neointima formation in vivo
- Author
-
Serena Zacchigna, Marco Presta, Mauro Giacca, Maura Camozzi, Alberto Mantovani, Genaro A. Ramirez-Correa, Marco Rusnati, Barbara Bottazzi, Daniela Coltrini, Camozzi, M, Zacchigna, Serena, Rusnati, M, Coltrini, D, RAMIREZ CORREA, G, Bottazzi, B, Mantovani, A, Giacca, Mauro, and Presta, M.
- Subjects
Male ,Neointima ,Cell type ,Cell Survival ,Cell ,Inflammation ,In Vitro Techniques ,Biology ,Fibroblast growth factor ,Muscle, Smooth, Vascular ,arterial injury ,Catheterization ,Iodine Radioisotopes ,Transduction, Genetic ,fibroblast growth factor ,medicine ,Animals ,Humans ,Rats, Wistar ,pentraxin 3 ,Receptor ,Cells, Cultured ,Hyperplasia ,Cell growth ,Chemotaxis ,PTX3 ,Coronary Vessels ,gene therapy ,smooth muscle cells ,Rats ,Cell biology ,Serum Amyloid P-Component ,C-Reactive Protein ,medicine.anatomical_structure ,Immunology ,Fibroblast Growth Factor 2 ,medicine.symptom ,Carotid Artery Injuries ,Tunica Intima ,Cardiology and Cardiovascular Medicine ,Cell Division - Abstract
Objective— The fibroblast growth factor (FGF)/FGF receptor system plays an important role in smooth muscle cell (SMC) activation. Long-pentraxin 3 (PTX3) is a soluble pattern recognition receptor with non-redundant functions in inflammation and innate immunity. PTX3 is produced by different cell types of the vessel wall, including SMCs. PTX3 binds FGF2 and inhibits its angiogenic activity on endothelial cells. We investigated the capacity of PTX3 to affect FGF2-dependent SMC activation in vitro and in vivo. Methods and Results— When added to human coronary artery SMCs, human PTX3 inhibits cell proliferation driven by endogenous FGF2 and the mitogenic and chemotactic activity exerted by exogenous recombinant FGF2. Accordingly, PTX3 prevents 125 I-FGF2 interaction with FGF receptors on the same cells. Also, PTX3 overexpression after recombinant adeno-associated virus- PTX3 gene transfer inhibits human coronary artery SMC proliferation and survival promoted by FGF2 in vitro. Consistently, a single local endovascular injection of recombinant adeno-associated virus- PTX3 gene inhibits intimal thickening after balloon injury in rat carotid arteries. Conclusions— PTX3 is a potent inhibitor of the autocrine and paracrine stimulation exerted by FGF2 on SMCs. Local PTX3 upregulation may modulate SMC activation after arterial injury.
- Published
- 2005
17. Iron supplementation enhances RSL3-induced ferroptosis to treat naïve and prevent castration-resistant prostate cancer.
- Author
-
Maccarinelli F, Coltrini D, Mussi S, Bugatti M, Turati M, Chiodelli P, Giacomini A, De Cillis F, Cattane N, Cattaneo A, Ligresti A, Asperti M, Poli M, Vermi W, Presta M, and Ronca R
- Abstract
Prostate cancer (PCa) is a leading cause of death in the male population commonly treated with androgen deprivation therapy that often relapses as androgen-independent and aggressive castration-resistant prostate cancer (CRPC). Ferroptosis is a recently described form of cell death that requires abundant cytosolic labile iron to promote membrane lipid peroxidation and which can be induced by agents that inhibit the glutathione peroxidase-4 activity such as RSL3. Exploiting in vitro and in vivo human and murine PCa models and the multistage transgenic TRAMP model of PCa we show that RSL3 induces ferroptosis in PCa cells and demonstrate for the first time that iron supplementation significantly increases the effect of RSL3 triggering lipid peroxidation, enhanced intracellular stress and leading to cancer cell death. Moreover, the combination with the second generation anti-androgen drug enzalutamide potentiates the effect of the RSL3 + iron combination leading to superior inhibition of PCa and preventing the onset of CRPC in the TRAMP mouse model. These data open new perspectives in the use of pro-ferroptotic approaches alone or in combination with enzalutamide for the treatment of PCa., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
18. Cannabidiol alters mitochondrial bioenergetics via VDAC1 and triggers cell death in hormone-refractory prostate cancer.
- Author
-
Mahmoud AM, Kostrzewa M, Marolda V, Cerasuolo M, Maccarinelli F, Coltrini D, Rezzola S, Giacomini A, Mollica MP, Motta A, Paris D, Zorzano A, Di Marzo V, Ronca R, and Ligresti A
- Subjects
- Humans, Male, Mice, Animals, Cell Death, Mitochondria metabolism, Oxidative Phosphorylation, Carcinogenesis metabolism, Hormones metabolism, Voltage-Dependent Anion Channel 1 metabolism, Cannabidiol pharmacology, Prostatic Neoplasms metabolism
- Abstract
In spite of the huge advancements in both diagnosis and interventions, hormone refractory prostate cancer (HRPC) remains a major hurdle in prostate cancer (PCa). Metabolic reprogramming plays a key role in PCa oncogenesis and resistance. However, the dynamics between metabolism and oncogenesis are not fully understood. Here, we demonstrate that two multi-target natural products, cannabidiol (CBD) and cannabigerol (CBG), suppress HRPC development in the TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model by reprogramming metabolic and oncogenic signaling. Mechanistically, CBD increases glycolytic capacity and inhibits oxidative phosphorylation in enzalutamide-resistant HRPC cells. This action of CBD originates from its effect on metabolic plasticity via modulation of VDAC1 and hexokinase II (HKII) coupling on the outer mitochondrial membrane, which leads to strong shifts of mitochondrial functions and oncogenic signaling pathways. The effect of CBG on enzalutamide-resistant HRPC cells was less pronounced than CBD and only partially attributable to its action on mitochondria. However, when optimally combined, these two cannabinoids exhibited strong anti-tumor effects in TRAMP mice, even when these had become refractory to enzalutamide, thus pointing to their therapeutical potential against PCa., Competing Interests: Conflict of interest The authors have no conflicts of interest to declare. All co-authors have seen and agree with the contents of the manuscript and there is no financial interest to report., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. β-Galactosylceramidase Deficiency Causes Upregulation of Long Pentraxin-3 in the Central Nervous System of Krabbe Patients and Twitcher Mice.
- Author
-
Coltrini D, Chandran AMK, Belleri M, Poliani PL, Cominelli M, Pagani F, Capra M, Calza S, Prioni S, Mauri L, Prinetti A, Kofler JK, Escolar ML, and Presta M
- Subjects
- Animals, Central Nervous System metabolism, Disease Models, Animal, Humans, Mice, Mice, Transgenic, Psychosine, Up-Regulation, C-Reactive Protein genetics, Galactosylceramidase deficiency, Galactosylceramidase genetics, Leukodystrophy, Globoid Cell metabolism, Nerve Tissue Proteins genetics
- Abstract
Globoid cell leukodystrophy (GLD), or Krabbe disease, is a neurodegenerative sphingolipidosis caused by genetic deficiency of lysosomal β-galactosylceramidase ( GALC ), characterized by neuroinflammation and demyelination of the central (CNS) and peripheral nervous system. The acute phase protein long pentraxin-3 (PTX3) is a soluble pattern recognition receptor and a regulator of innate immunity. Growing evidence points to the involvement of PTX3 in neurodegeneration. However, the expression and role of PTX3 in the neurodegenerative/neuroinflammatory processes that characterize GLD remain unexplored. Here, immunohistochemical analysis of brain samples from Krabbe patients showed that macrophages and globoid cells are intensely immunoreactive for PTX3. Accordingly, Ptx3 expression increases throughout the course of the disease in the cerebrum, cerebellum, and spinal cord of GALC-deficient twitcher ( Galc
twi/twi ) mice, an authentic animal model of GLD. This was paralleled by the upregulation of proinflammatory genes and M1-polarized macrophage/microglia markers and of the levels of PTX3 protein in CNS and plasma of twitcher animals. Crossing of Galctwi/twi mice with transgenic PTX3 overexpressing animals ( hPTX3 mice) demonstrated that constitutive PTX3 overexpression reduced the severity of clinical signs and the upregulation of proinflammatory genes in the spinal cord of P35 hPTX3 / Galctwi/twi mice when compared to Galctwi/twi littermates, leading to a limited increase of their life span. However, this occurred in the absence of a significant impact on the histopathological findings and on the accumulation of the neurotoxic metabolite psychosine when evaluated at this late time point of the disease. In conclusion, our results provide the first evidence that PTX3 is produced in the CNS of GALC-deficient Krabbe patients and twitcher mice. PTX3 may exert a protective role by reducing the neuroinflammatory response that occurs in the spinal cord of GALC-deficient animals.- Published
- 2022
- Full Text
- View/download PDF
20. FGFR blockade by pemigatinib treats naïve and castration resistant prostate cancer.
- Author
-
Chiodelli P, Coltrini D, Turati M, Cerasuolo M, Maccarinelli F, Rezzola S, Grillo E, Giacomini A, Taranto S, Mussi S, Ligresti A, Presta M, and Ronca R
- Subjects
- Androgen Antagonists pharmacology, Animals, Humans, Male, Mice, Morpholines pharmacology, Pyrimidines pharmacology, Pyrroles pharmacology, Androgen Antagonists therapeutic use, Morpholines therapeutic use, Prostatic Neoplasms, Castration-Resistant drug therapy, Pyrimidines therapeutic use, Pyrroles therapeutic use, Receptor, Fibroblast Growth Factor, Type 2 antagonists & inhibitors
- Abstract
Prostate cancer (PCa) is a leading cause of cancer mortality in the male population commonly treated with androgen deprivation therapy (ADT) and relapsing as aggressive and androgen-independent castration-resistant prostate cancer (CRPC). In PCa the FGF/FGFR family of growth factors and receptors represents a relevant mediator of cancer growth, tumor-stroma interaction, and a driver of resistance and relapse to ADT. In the present work, we validate the therapeutic efficacy the FDA-approved FGFR inhibitor pemigatinib, in an integrated platform consisting of human and murine PCa cells, and the transgenic multistage TRAMP model of PCa that recapitulates both androgen-dependent and CRPC settings. Our results show for the first time that pemigatinib causes intracellular stress and cell death in PCa cells and prevents tumor growth in vivo and in the multistage model. In addition, the combination of pemigatinib with enzalutamide resulted in long-lasting tumor inhibition and prevention of CRPC relapse in TRAMP mice. These data are confirmed by the implementation of a stochastic mathematical model and in silico simulation. Pemigatinib represents a promising FDA-approved FGFR inhibitor for the treatment of PCa and CRPC alone and in combination with enzalutamide., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. An Orthotopic Model of Uveal Melanoma in Zebrafish Embryo: A Novel Platform for Drug Evaluation.
- Author
-
Tobia C, Coltrini D, Ronca R, Loda A, Guerra J, Scalvini E, Semeraro F, and Rezzola S
- Abstract
Uveal melanoma is a highly metastatic tumor, representing the most common primary intraocular malignancy in adults. Tumor cell xenografts in zebrafish embryos may provide the opportunity to study in vivo different aspects of the neoplastic disease and its response to therapy. Here, we established an orthotopic model of uveal melanoma in zebrafish by injecting highly metastatic murine B16-BL6 and B16-LS9 melanoma cells, human A375M melanoma cells, and human 92.1 uveal melanoma cells into the eye of zebrafish embryos in the proximity of the developing choroidal vasculature. Immunohistochemical and immunofluorescence analyses showed that melanoma cells proliferate during the first four days after injection and move towards the eye surface. Moreover, bioluminescence analysis of luciferase-expressing human 92.1 uveal melanoma cells allowed the quantitative assessment of the antitumor activity exerted by the canonical chemotherapeutic drugs paclitaxel, panobinostat, and everolimus after their injection into the grafted eye. Altogether, our data demonstrate that the zebrafish embryo eye is a permissive environment for the growth of invasive cutaneous and uveal melanoma cells. In addition, we have established a new luciferase-based in vivo orthotopic model that allows the quantification of human uveal melanoma cells engrafted in the zebrafish embryo eye, and which may represent a suitable tool for the screening of novel drug candidates for uveal melanoma therapy.
- Published
- 2021
- Full Text
- View/download PDF
22. Vitreous from idiopathic epiretinal membrane patients induces glial-to-mesenchymal transition in Müller cells.
- Author
-
Krishna Chandran AM, Coltrini D, Belleri M, Rezzola S, Gambicorti E, Romano D, Morescalchi F, Calza S, Semeraro F, and Presta M
- Subjects
- Aged, Biomarkers metabolism, Cell Transdifferentiation physiology, Cells, Cultured, Down-Regulation physiology, Ependymoglial Cells metabolism, Epiretinal Membrane metabolism, Female, Fibrosis metabolism, Fibrosis pathology, Humans, Male, Myofibroblasts metabolism, Myofibroblasts pathology, Neuroglia metabolism, Retina metabolism, Retina pathology, Up-Regulation physiology, Ependymoglial Cells pathology, Epiretinal Membrane pathology, Epithelial-Mesenchymal Transition physiology, Neuroglia pathology
- Abstract
Idiopathic epiretinal membranes (ERMs) are fibrocellular membranes containing extracellular matrix proteins and epiretinal cells of retinal and extraretinal origin. iERMs lead to decreased visual acuity and their pathogenesis has not been completely defined. Macroglial Müller cells appear to play a pivotal role in the pathogenesis of iERM where they may undergo glial-to-mesenchymal transition (GMT), a transdifferentiation process characterized by the downregulation of Müller cell markers, paralleled by the upregulation of pro-fibrotic myofibroblast markers. Previous observations from our laboratory allowed the molecular identification of two major clusters of iERM patients (named iERM-A and iERM-B), iERM-A patients being characterized by less severe clinical features and a more "quiescent" iERM gene expression profile when compared to iERM-B patients. In the present work, Müller MIO-M1 cells were exposed to vitreous samples obtained before membrane peeling from the same cohort of iERM-A and iERM-B patients. The results demonstrate that iERM vitreous induces proliferation, migration, and GMT in MIO-M1 cells, a phenotype consistent with Müller cell behavior during iERM progression. However, even though the vitreous samples obtained from iERM-A patients were able to induce a complete GMT in MIO-M1 cells, iERM-B samples caused only a partial GMT, characterized by the downregulation of Müller cell markers in the absence of upregulation of pro-fibrotic myofibroblast markers. Together, the results indicate that a relationship may exist among the ability of iERM vitreous to modulate GMT in Müller cells, the molecular profile of the corresponding iERMs, and the clinical features of iERM patients., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Gene expression analysis identifies two distinct molecular clusters of idiopatic epiretinal membranes.
- Author
-
Coltrini D, Belleri M, Gambicorti E, Romano D, Morescalchi F, Krishna Chandran AM, Calza S, Semeraro F, and Presta M
- Subjects
- Aged, Cluster Analysis, Epiretinal Membrane pathology, Female, Gene Expression Profiling, Humans, Male, Tomography, Optical Coherence, Epiretinal Membrane genetics
- Abstract
Idiopathic epiretinal membranes (ERMs) are fibrocellular membranes containing extracellular matrix proteins and epiretinal cells of retinal and extraretinal origin. iERMs lead to decreased visual acuity and their pathogenesis has not been completely defined. Aim of this study was to provide a molecular characterization of iERMs by gene expression analysis. To this purpose, 56 iERMs obtained by pars plana vitrectomy were analyzed for the expression levels of genes encoding biomarkers of the cellular and molecular events occurring in iERMs. RT-qPCR analysis showed significant differences in the levels of cell population, extracellular matrix and cytokine/growth factor biomarkers among the iERMs investigated. Hierarchical clustering of RT-qPCR data identified two distinct iERM clusters, Cluster B samples representing transcriptionally "activated" iERMs when compared to transcriptionally "quiescent" Cluster A specimens. Further, Cluster B could be subdivided in two subgroups, Cluster B1 iERMs, characterized by a marked glial cell activation, and Cluster B2 samples characterized by a more pro-fibrotic phenotype. Preoperative decimal best-corrected visual acuity and post-surgery inner segment/outer grading values were higher in Cluster A patients, that showed a prevalence of fovea-attached type iERMs with near-normal inner retina, than in Cluster B patients, that presented more severe clinical and spectral domain optical coherence tomography (SD-OCT) features. In conclusion, this molecular characterization has identified two major clusters of iERM specimens with distinct transcriptional activities that reflect different clinical and SD-OCT features of iERM patients. This retrospective work paves the way to prospective whole-genome transcriptomic studies to allow a molecular classification of iERMs and for the identification of molecular signature(s) of prognostic and therapeutic significance., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
24. β-Galactosylceramidase Promotes Melanoma Growth via Modulation of Ceramide Metabolism.
- Author
-
Belleri M, Paganini G, Coltrini D, Ronca R, Zizioli D, Corsini M, Barbieri A, Grillo E, Calza S, Bresciani R, Maiorano E, Mastropasqua MG, Annese T, Giacomini A, Ribatti D, Casas J, Levade T, Fabrias G, and Presta M
- Subjects
- Animals, Cell Differentiation genetics, Cell Line, Tumor, Cell Movement, Cell Proliferation, Down-Regulation, Galactosylceramidase genetics, Gene Silencing, Humans, Lung Neoplasms secondary, Melanocytes cytology, Melanocytes enzymology, Melanoma metabolism, Melanoma secondary, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, SCID, Neoplasm Invasiveness, Skin Neoplasms metabolism, Sphingolipids metabolism, Sphingomyelin Phosphodiesterase metabolism, Up-Regulation, Zebrafish, Ceramides metabolism, Galactosylceramidase metabolism, Melanoma pathology, Skin Neoplasms pathology
- Abstract
Disturbance of sphingolipid metabolism may represent a novel therapeutic target in metastatic melanoma, the most lethal form of skin cancer. β-Galactosylceramidase (GALC) removes β-galactose from galactosylceramide and other sphingolipids. In this study, we show that downregulation of galcb , a zebrafish ortholog of human GALC , affects melanoblast and melanocyte differentiation in zebrafish embryos, suggesting a possible role for GALC in melanoma. On this basis, the impact of GALC expression in murine B16-F10 and human A2058 melanoma cells was investigated following its silencing or upregulation. Galc knockdown hampered growth, motility, and invasive capacity of B16-F10 cells and their tumorigenic and metastatic activity when grafted in syngeneic mice or zebrafish embryos. Galc -silenced cells displayed altered sphingolipid metabolism and increased intracellular levels of ceramide, paralleled by a nonredundant upregulation of Smpd3 , which encodes for the ceramide-generating enzyme neutral sphingomyelinase 2. Accordingly, GALC downregulation caused SMPD3 upregulation, increased ceramide levels, and inhibited the tumorigenic activity of human melanoma A2058 cells, whereas GALC upregulation exerted opposite effects. In concordance with information from melanoma database mining, RNAscope analysis demonstrated a progressive increase of GALC expression from common nevi to stage IV human melanoma samples that was paralleled by increases in microphthalmia transcription factor and tyrosinase immunoreactivity inversely related to SMPD3 and ceramide levels. Overall, these findings indicate that GALC may play an oncogenic role in melanoma by modulating the levels of intracellular ceramide, thus providing novel opportunities for melanoma therapy. SIGNIFICANCE: Data from zebrafish embryos, murine and human cell melanoma lines, and patient-derived tumor specimens indicate that β-galactosylceramidase plays an oncogenic role in melanoma and may serve as a therapeutic target., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
25. Natural killer cell impairment in ovarian clear cell carcinoma.
- Author
-
Patrizi O, Rampinelli F, Coltrini D, Pesce S, Carlomagno S, Sivori S, Pascale A, Marcenaro E, Parolini S, and Tabellini G
- Subjects
- Antigens, Differentiation, T-Lymphocyte immunology, Carcinoma pathology, Female, Humans, K562 Cells, Killer Cells, Natural pathology, Neoplasm Proteins immunology, Ovarian Neoplasms pathology, Carcinoma immunology, Cell Communication immunology, Cytotoxicity, Immunologic, Killer Cells, Natural immunology, Ovarian Neoplasms immunology
- Abstract
In the present study, we report the analysis of NK cells derived from patients suffering from a rare ovarian cancer histotype of clear cell carcinoma (OCCC) resistant to conventional chemotherapies. We analyzed the phenotype of NK cells derived from peripheral blood (PB) and peritoneal fluid (PF) and evaluated cytotoxic interactions between NK cells and autologous tumor cells (ATC) derived from patients. We provided evidence of impaired degranulation capacity of NK cells derived from patients' PF in the presence of ATC. Analyzing tumor cell ligands recognized by NK cell receptors, we found that ATC are characterized by an HLA class I
+ phenotype (although the level of HLA-I expression varies among all patients) and by a heterogeneous expression of ligands for activating NK receptors (from normal to decreased expression of some markers). Furthermore, we observed a down-regulation of crucial NK cell activating receptors, primarily DNAX Accessory Molecule-1 (DNAM-1), on tumor-associated NK cells. Based on these results, we propose that this severe lysis defect may be due to both negative interactions between HLA-I-specific inhibitory NK cell receptors/HLA-I molecules and to defective interactions between activating NK receptors and cognate ligands. In conclusion, for the first time, the phenotypic and functional properties of tumor-associated NK cells and their ATC derived from PF of patients with advanced stage of OCCC were characterized. Taken together results indicate altered interactions between NK cells and ATC and shed light on the aggressive mechanisms of this cancer histotype. Further studies on this rare tumor will be helpful to improve and define more effective therapies., (©2020 Society for Leukocyte Biology.)- Published
- 2020
- Full Text
- View/download PDF
26. D-Peptide analogues of Boc-Phe-Leu-Phe-Leu-Phe-COOH induce neovascularization via endothelial N-formyl peptide receptor 3.
- Author
-
Nawaz MI, Rezzola S, Tobia C, Coltrini D, Belleri M, Mitola S, Corsini M, Sandomenico A, Caporale A, Ruvo M, and Presta M
- Subjects
- Humans, Oligopeptides chemical synthesis, Oligopeptides chemistry, Human Umbilical Vein Endothelial Cells metabolism, Neovascularization, Physiologic drug effects, Oligopeptides pharmacology, Receptors, Formyl Peptide agonists, Receptors, Formyl Peptide metabolism
- Abstract
N-formyl peptide receptors (FPRs) are G protein-coupled receptors involved in the recruitment and activation of immune cells in response to pathogen-associated molecular patterns. Three FPRs have been identified in humans (FPR1-FPR3), characterized by different ligand properties, biological function and cellular distribution. Recent findings from our laboratory have shown that the peptide BOC-FLFLF (L-BOC2), related to the FPR antagonist BOC2, acts as an angiogenesis inhibitor by binding to various angiogenic growth factors, including vascular endothelial growth factor-A
165 (VEGF). Here we show that the all-D-enantiomer of L-BOC2 (D-BOC2) is devoid of any VEGF antagonist activity. At variance, D-BOC2, as well as the D-FLFLF and succinimidyl (Succ)-D-FLFLF (D-Succ-F3) D-peptide variants, is endowed with a pro-angiogenic potential. In particular, the D-peptide D-Succ-F3 exerts a pro-angiogenic activity in a variety of in vitro assays on human umbilical vein endothelial cells (HUVECs) and in ex vivo and in vivo assays in chick and zebrafish embryos and adult mice. This activity is related to the capacity of D-Succ-F3 to bind FRP3 expressed by HUVECs. Indeed, the effects exerted by D-Succ-F3 on HUVECs are fully suppressed by the G protein-coupled receptor inhibitor pertussis toxin, the FPR2/FPR3 antagonist WRW4 and by an anti-FPR3 antibody. A similar inhibition was observed following WRW4-induced FPR3 desensitization in HUVECs. Finally, D-Succ-F3 prevented the binding of the anti-FPR3 antibody to the cell surface of HUVECs. In conclusion, our data demonstrate that the angiogenic activity of D-Succ-F3 is due to the engagement and activation of FPR3 expressed by endothelial cells, thus shedding a new light on the biological function of this chemoattractant receptor.- Published
- 2020
- Full Text
- View/download PDF
27. Modeling Acquired Resistance to the Second-Generation Androgen Receptor Antagonist Enzalutamide in the TRAMP Model of Prostate Cancer.
- Author
-
Cerasuolo M, Maccarinelli F, Coltrini D, Mahmoud AM, Marolda V, Ghedini GC, Rezzola S, Giacomini A, Triggiani L, Kostrzewa M, Verde R, Paris D, Melck D, Presta M, Ligresti A, and Ronca R
- Subjects
- Androgen Receptor Antagonists therapeutic use, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Benzamides, Cell Line, Tumor transplantation, Disease Models, Animal, Disease Progression, Humans, Male, Mice, Nitriles, Phenylthiohydantoin pharmacology, Phenylthiohydantoin therapeutic use, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen metabolism, Signal Transduction drug effects, Taxoids pharmacology, Taxoids therapeutic use, Androgen Receptor Antagonists pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Drug Resistance, Neoplasm drug effects, Phenylthiohydantoin analogs & derivatives, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Enzalutamide (MDV3100) is a potent second-generation androgen receptor antagonist approved for the treatment of castration-resistant prostate cancer (CRPC) in chemotherapy-naïve as well as in patients previously exposed to chemotherapy. However, resistance to enzalutamide and enzalutamide withdrawal syndrome have been reported. Thus, reliable and integrated preclinical models are required to elucidate the mechanisms of resistance and to assess therapeutic settings that may delay or prevent the onset of resistance. In this study, the prostate cancer multistage murine model TRAMP and TRAMP-derived cells have been used to extensively characterize in vitro and in vivo the response and resistance to enzalutamide. The therapeutic profile as well as the resistance onset were characterized and a multiscale stochastic mathematical model was proposed to link the in vitro and in vivo evolution of prostate cancer. The model showed that all therapeutic strategies that use enzalutamide result in the onset of resistance. The model also showed that combination therapies can delay the onset of resistance to enzalutamide, and in the best scenario, can eliminate the disease. These results set the basis for the exploitation of this "TRAMP-based platform" to test novel therapeutic approaches and build further mathematical models of combination therapies to treat prostate cancer and CRPC. Significance: Merging mathematical modeling with experimental data, this study presents the "TRAMP-based platform" as a novel experimental tool to study the in vitro and in vivo evolution of prostate cancer resistance to enzalutamide., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
28. β-Galactosylceramidase Deficiency Causes Bone Marrow Vascular Defects in an Animal Model of Krabbe Disease.
- Author
-
Belleri M, Coltrini D, Righi M, Ravelli C, Taranto S, Chiodelli P, Mitola S, Presta M, and Giacomini A
- Subjects
- Animals, Bone Marrow pathology, Brain metabolism, Cytokines metabolism, Disease Models, Animal, Endothelial Cells metabolism, Galactosylceramidase genetics, Hematopoiesis, Hematopoietic Stem Cell Transplantation, Leukodystrophy, Globoid Cell genetics, Leukodystrophy, Globoid Cell pathology, Mice, Mice, Inbred C57BL, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, Bone Marrow metabolism, Galactosylceramidase metabolism, Leukodystrophy, Globoid Cell metabolism
- Abstract
Krabbe disease (KD) is an autosomal recessive sphingolipidosis caused by the deficiency of the lysosomal hydrolase β-galactosylceramidase (GALC). Oligodendroglia degeneration and demyelination of the nervous system lead to neurological dysfunctions which are usually lethal by two years of age. At present, the only clinical treatment with any proven efficacy is hematopoietic stem-cell transplantation, which is more effective when administered in the neonatal period to presymptomatic recipients. Bone marrow (BM) sinusoidal endothelial cells (SECs) play a pivotal role in stem cell engraftment and reconstitution of hematopoiesis. Previous observations had shown significant alterations of microvascular endothelial cells in the brain of KD patients and in Galc mutant twitcher mice, an authentic model of the disease. In the present study, we investigated the vascular component of the BM in the femurs of symptomatic homozygous twitcher mice at postnatal day P36. Histological, immunohistochemical, and two-photon microscopy imaging analyses revealed the presence of significant alterations of the diaphyseal BM vasculature, characterized by enlarged, discontinuous, and hemorrhagic SECs that express the endothelial marker vascular endothelial growth factor receptor-2 (VEGFR2) but lack platelet/endothelial cell adhesion molecule-1 (CD31) expression. In addition, computer-aided image analysis indicates that twitcher CD31
- /VEGFR2+ SECs show a significant increase in lumen size and in the number and size of endothelial gaps compared to BM SECs of wild type littermates. These results suggest that morphofunctional defects in the BM vascular niche may contribute to the limited therapeutic efficacy of hematopoietic stem-cell transplantation in KD patients at symptomatic stages of the disease.- Published
- 2019
- Full Text
- View/download PDF
29. From Natural Killer Cell Receptor Discovery to Characterization of Natural Killer Cell Defects in Primary Immunodeficiencies.
- Author
-
Tabellini G, Patrizi O, Dobbs K, Lougaris V, Baronio M, Coltrini D, Plebani A, Badolato R, Notarangelo LD, and Parolini S
- Subjects
- History, 20th Century, History, 21st Century, Humans, Epstein-Barr Virus Infections genetics, Epstein-Barr Virus Infections history, Epstein-Barr Virus Infections immunology, Herpesvirus 4, Human genetics, Herpesvirus 4, Human immunology, Killer Cells, Natural immunology, Lymphoproliferative Disorders genetics, Lymphoproliferative Disorders history, Lymphoproliferative Disorders immunology, Primary Immunodeficiency Diseases genetics, Primary Immunodeficiency Diseases history, Primary Immunodeficiency Diseases immunology, Receptors, KIR genetics, Receptors, KIR history, Receptors, KIR immunology
- Abstract
Alessandro Moretta was Professor of Histology at University of Brescia from 1994 to 1997. It was in that period that we met and started a collaboration that continued in the years to follow. He immediately involved us in the production of monoclonal antibodies (mAbs) that allowed the identification and fine characterization of novel receptor molecules that were able to activate or inhibit human Natural Killer cell function, including several antibodies specific for Natural Cytotoxicity Receptor (NCR) and Killer-cell Immunoglobulin-like Receptor (KIR) molecules. These reagents, generated in our laboratory in Brescia, contributed to complete the studies aimed to characterize innate lymphoid NK cells, that had been initiated by Alessandro and his brother Lorenzo in Genoa. Soon, we identified an anti-KIR3DL2 that was subsequently shown to be helpful for the diagnosis and treatment of various forms of cutaneous T cell lymphoma. While in Brescia, Alessandro established a partnership with those of us who were working in the Department of Pediatrics; together, in short time we tackled the goal of studying the role of NK cells in patients with primary immunodeficiencies. This collaboration led to novel discoveries that shed light on the critical role played by NK cells in the immune response against virus and tumors in humans, as best exemplified by our characterization of the molecular mechanisms of impaired control of Epstein-Barr Virus (EBV) infection in patients with X-linked lymphoproliferative (XLP) disease. After Alessandro left Brescia to return to Genoa, our collaboration continued with the same enthusiasm, and even from a distance he remained an extraordinary example of an inspirational and generous mentor. This review is a sign of our gratitude to a mentor and a friend whom we deeply miss.
- Published
- 2019
- Full Text
- View/download PDF
30. Fibroblast growth factor modulates mast cell recruitment in a murine model of prostate cancer.
- Author
-
Ronca R, Tamma R, Coltrini D, Ruggieri S, Presta M, and Ribatti D
- Abstract
Mast cells are important modifiers of prostate tumor microenvironment. The fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) system plays a non-redundant autocrine/paracrine role in the growth, vascularization and progression of prostate tumors. Accordingly, the FGF antagonist long pentraxin-3 (PTX3) and the PTX3-derived small molecule FGF-trap NSC12 have been shown to inhibit the growth and vascularization of different FGF-dependent tumor types, including prostate cancer. In this study, we show that recombinant FGF2 is able to cause mast cell recruitment in vivo in the Matrigel plug assay. Conversely, PTX3 overexpression in transgenic mice or treatment with the FGF inhibitor NSC12 result in a significant inhibition of the growth and vascularization of TRAMP-C2 tumor grafts, a murine model of prostate cancer, that were paralleled by a decrease of mast cell infiltrate into the lesion. These data confirm and extend previous observations about the capacity of mast cells to respond chemotactically to FGF2 stimulation and provide evidence about a relationship among mast cell recruitment, angiogenesis, and tumor growth in human prostate adenocarcinoma., Competing Interests: CONFLICTS OF INTEREST The authors declare no competing financial interests.
- Published
- 2017
- Full Text
- View/download PDF
31. Inflammation and N-formyl peptide receptors mediate the angiogenic activity of human vitreous humour in proliferative diabetic retinopathy.
- Author
-
Rezzola S, Corsini M, Chiodelli P, Cancarini A, Nawaz IM, Coltrini D, Mitola S, Ronca R, Belleri M, Lista L, Rusciano D, De Rosa M, Pavone V, Semeraro F, and Presta M
- Subjects
- Aged, Aged, 80 and over, Animals, Cell Line, Diabetic Retinopathy immunology, Female, Human Umbilical Vein Endothelial Cells metabolism, Humans, Male, Mice, Middle Aged, Neovascularization, Pathologic immunology, Neovascularization, Pathologic metabolism, Diabetic Retinopathy metabolism, Inflammation metabolism, Receptors, Formyl Peptide metabolism, Vitreous Body metabolism
- Abstract
Aims/hypothesis: Angiogenesis and inflammation characterise proliferative diabetic retinopathy (PDR), a major complication of diabetes mellitus. However, the impact of inflammation on the pathogenesis of PDR neovascularisation has not been elucidated. Here, we assessed the capacity of PDR vitreous fluid to induce pro-angiogenic/proinflammatory responses in endothelium and the contribution of the inflammation-related pattern recognition N-formyl peptide receptors (FPRs) in mediating these responses., Methods: Pooled and individual pars plana vitrectomy-derived PDR vitreous fluid ('PDR vitreous') samples were assessed in endothelial cell proliferation, motility, sprouting and morphogenesis assays, and for the capacity to induce proinflammatory transcription factor activation, reactive oxygen species production, intercellular junction disruption and leucocyte-adhesion molecule upregulation in these cells. In vivo, the pro-angiogenic/proinflammatory activity of PDR vitreous was tested in murine Matrigel plug and chick embryo chorioallantoic membrane (CAM) assays. Finally, the FPR inhibitors Boc-Phe-Leu-Phe-Leu-Phe (Boc-FLFLF) and Ac-L-Arg-Aib-L-Arg-L-Cα(Me)Phe-NH
2 tetrapeptide (UPARANT) were evaluated for their capacity to affect the biological responses elicited by PDR vitreous., Results: PDR vitreous activates a pro-angiogenic/proinflammatory phenotype in endothelial cells. Accordingly, PDR vitreous triggers a potent angiogenic/inflammatory response in vivo. Notably, the different capacity of individual PDR vitreous samples to induce neovessel formation in the CAM correlates with their ability to recruit infiltrating CD45+ cells. Finally, the FPR inhibitor Boc-FLFLF and the novel FPR antagonist UPARANT inhibit neovessel formation and inflammatory responses triggered by PDR vitreous in the CAM assay., Conclusions/interpretation: This study provides evidence that inflammation mediates the angiogenic activity of PDR vitreous and paves the way for the development of FPR-targeting anti-inflammatory/anti-angiogenic approaches for PDR therapy.- Published
- 2017
- Full Text
- View/download PDF
32. β3 Integrin Promotes Long-Lasting Activation and Polarization of Vascular Endothelial Growth Factor Receptor 2 by Immobilized Ligand.
- Author
-
Ravelli C, Grillo E, Corsini M, Coltrini D, Presta M, and Mitola S
- Subjects
- Cells, Cultured, Endothelial Cells cytology, Endothelial Cells metabolism, Endothelium, Vascular cytology, Humans, Sensitivity and Specificity, Signal Transduction, Endothelium, Vascular metabolism, Integrin beta3 metabolism, Neovascularization, Physiologic physiology, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Objective: During neovessel formation, angiogenic growth factors associate with the extracellular matrix. These immobilized factors represent a persistent stimulus for the otherwise quiescent endothelial cells (ECs), driving directional EC migration and proliferation and leading to new blood vessel growth. Vascular endothelial growth factor receptor 2 (VEGFR2) is the main mediator of angiogenesis. Although VEGFR2 signaling has been deeply characterized, little is known about its subcellular localization during neovessel formation. Aim of this study was the characterization and molecular determinants of activated VEGFR2 localization in ECs during neovessel formation in response to matrix-immobilized ligand., Approach and Results: Here we demonstrate that ECs stimulated by extracellular matrix-associated gremlin, a noncanonical VEGFR2 ligand, are polarized and relocate the receptor in close contact with the angiogenic factor-enriched matrix both in vitro and in vivo. GM1 (monosialotetrahexosylganglioside)-positive planar lipid rafts, β3 integrin receptors, and the intracellular signaling transducers focal adhesion kinase and RhoA (Ras homolog gene family, member A) cooperate to promote VEGFR2 long-term polarization and activation., Conclusions: A ligand anchored to the extracellular matrix induces VEGFR2 polarization in ECs. Long-lasting VEGFR2 relocation is closely dependent on lipid raft integrity and activation of β3 integrin pathway. The study of the endothelial responses to immobilized growth factors may offer insights into the angiogenic process in physiological and pathological conditions, including cancer, and for a better engineering of synthetic tissue scaffolds to blend with the host vasculature., (© 2015 The Authors.)
- Published
- 2015
- Full Text
- View/download PDF
33. Brain angioarchitecture and intussusceptive microvascular growth in a murine model of Krabbe disease.
- Author
-
Giacomini A, Ackermann M, Belleri M, Coltrini D, Nico B, Ribatti D, Konerding MA, Presta M, and Righi M
- Subjects
- Animals, Disease Models, Animal, Humans, Intussusception genetics, Intussusception pathology, Leukodystrophy, Globoid Cell genetics, Leukodystrophy, Globoid Cell pathology, Mice, Brain blood supply, Cerebrovascular Circulation, Intussusception physiopathology, Leukodystrophy, Globoid Cell physiopathology, Microcirculation, Microvessels physiopathology
- Abstract
Defects of the angiogenic process occur in the brain of twitcher mouse, an authentic model of human Krabbe disease caused by genetic deficiency of lysosomal β-galactosylceramidase (GALC), leading to lethal neurological dysfunctions and accumulation of neurotoxic psychosine in the central nervous system. Here, quantitative computational analysis was used to explore the alterations of brain angioarchitecture in twitcher mice. To this aim, customized ImageJ routines were used to assess calibers, amounts, lengths and spatial dispersion of CD31(+) vessels in 3D volumes from the postnatal frontal cortex of twitcher animals. The results showed a decrease in CD31 immunoreactivity in twitcher brain with a marked reduction in total vessel lengths coupled with increased vessel fragmentation. No significant changes were instead observed for the spatial dispersion of brain vessels throughout volumes or in vascular calibers. Notably, no CD31(+) vessel changes were detected in twitcher kidneys in which psychosine accumulates at very low levels, thus confirming the specificity of the effect. Microvascular corrosion casting followed by scanning electron microscopy morphometry confirmed the presence of significant alterations of the functional angioarchitecture of the brain cortex of twitcher mice with reduction in microvascular density, vascular branch remodeling and intussusceptive angiogenesis. Intussusceptive microvascular growth, confirmed by histological analysis, was paralleled by alterations of the expression of intussusception-related genes in twitcher brain. Our data support the hypothesis that a marked decrease in vascular development concurs to the onset of neuropathological lesions in twitcher brain and suggest that neuroinflammation-driven intussusceptive responses may represent an attempt to compensate impaired sprouting angiogenesis.
- Published
- 2015
- Full Text
- View/download PDF
34. Long-Pentraxin 3 Derivative as a Small-Molecule FGF Trap for Cancer Therapy.
- Author
-
Ronca R, Giacomini A, Di Salle E, Coltrini D, Pagano K, Ragona L, Matarazzo S, Rezzola S, Maiolo D, Torrella R, Moroni E, Mazzieri R, Escobar G, Mor M, Colombo G, and Presta M
- Subjects
- Animals, Blotting, Western, C-Reactive Protein metabolism, Cell Cycle drug effects, Cell Cycle genetics, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Cells, Cultured, Female, Fibroblast Growth Factors metabolism, Fibroblast Growth Factors pharmacology, Humans, Kaplan-Meier Estimate, Male, Mice, Inbred C57BL, Mice, Nude, Mice, Transgenic, Molecular Structure, Neoplasms metabolism, Neoplasms therapy, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Reverse Transcriptase Polymerase Chain Reaction, Serum Amyloid P-Component metabolism, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Tumor Burden drug effects, Tumor Burden genetics, Xenograft Model Antitumor Assays methods, C-Reactive Protein genetics, Fibroblast Growth Factors genetics, Gene Expression Regulation, Neoplastic, Neoplasms genetics, Serum Amyloid P-Component genetics
- Abstract
The fibroblast growth factor (FGF)/FGF receptor (FGFR) system plays a crucial role in cancer by affecting tumor growth, angiogenesis, drug resistance, and escape from anti-angiogenic anti-vascular endothelial growth factor therapy. The soluble pattern recognition receptor long-pentraxin 3 (PTX3) acts as a multi-FGF antagonist. Here we demonstrate that human PTX3 overexpression in transgenic mice driven by the Tie2 promoter inhibits tumor growth, angiogenesis, and metastasis in heterotopic, orthotopic, and autochthonous FGF-dependent tumor models. Using pharmacophore modeling of the interaction of a minimal PTX3-derived FGF-binding pentapeptide with FGF2, we identified a small-molecule chemical (NSC12) that acts as an extracellular FGF trap with significant implications in cancer therapy., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape.
- Author
-
Pesce S, Tabellini G, Cantoni C, Patrizi O, Coltrini D, Rampinelli F, Matta J, Vivier E, Moretta A, Parolini S, and Marcenaro E
- Abstract
In this study the phenotype and function of tumor-associated NK cells from peritoneal fluids of a selected cohort of patients with seropapillary ovarian carcinoma were analyzed. In > 50% of these patients, the expression of the activating receptor NKp30 in tumor-associated NK cells was substantially reduced as compared to autologous peripheral blood (PB) NK cells. The impaired expression of this receptor was associated with the presence of one of its cellular ligands (B7-H6), which was detectable as a surface/cytosolic molecule in tumor cells and as a soluble molecule in the peritoneal fluid. NK cells from patients expressing this NKp30low phenotype displayed an impaired interferon-gamma (IFNγ) production and cytolytic function when tested against target cells expressing surface B7-H6. Our data also suggest that in these patients, the defective expression and function of NKp30 may be induced by the chronic engagement of this receptor by soluble B7-H6 or by tumor cells expressing this ligand. The impairment of NK cell functions described herein could represent a novel mechanism by which the tumor microenvironment may contribute to the escape from immune surveillance.
- Published
- 2015
- Full Text
- View/download PDF
36. Primitive neuroectodermal tumor in an ovarian cystic teratoma: natural killer and neuroblastoma cell analysis.
- Author
-
Tabellini G, Benassi M, Marcenaro E, Coltrini D, Patrizi O, Ricotta D, Rampinelli F, Moretta A, and Parolini S
- Abstract
In the present study, we report an extremely rare case of a 31-year-old woman with neuroblastoma arising in an ovarian cystic teratoma. We analyzed the expression of activating receptors on natural killer (NK) cells derived from the patient's peripheral blood and peritoneal fluid. In addition, we investigated the presence of specific ligands recognized by different NK cell receptors on tumor cells. We show that NK cells isolated from peritoneal fluid expressed certain triggering receptors including DNAM-1 (CD226) and CD16 with lower intensity as compared to peripheral blood NK cells. Remarkably, at variance with most cases of childhood neuroblastoma, the tumor cells from this patient expressed substantial amounts of HLA class-I molecules. These molecules are known to be protective against NK cell-mediated lysis. In addition, neuroblastoma cells expressed B7-H3 (CD276), another surface molecule that inhibits NK cell function. Finally, this tumor did not express the PVR (CD155) and nectin-2 (CD112) ligands for the DNAM-1 activating NK receptor, which plays a crucial role in NK/neuroblastoma interactions. Altogether, these findings indicate that the neuroblastoma cells of this patient express an NK-resistant surface phenotype, which is at least in part similar to that previously described in a fraction of childhood neuroblastoma.
- Published
- 2014
- Full Text
- View/download PDF
37. Long pentraxin-3 inhibits epithelial-mesenchymal transition in melanoma cells.
- Author
-
Ronca R, Di Salle E, Giacomini A, Leali D, Alessi P, Coltrini D, Ravelli C, Matarazzo S, Ribatti D, Vermi W, and Presta M
- Subjects
- Animals, C-Reactive Protein genetics, Chick Embryo, Epithelial-Mesenchymal Transition genetics, Female, Fibroblast Growth Factor 2 antagonists & inhibitors, Fibroblast Growth Factor 2 metabolism, Gene Expression, Humans, Melanoma genetics, Melanoma, Experimental, Mice, Neoplasm Invasiveness, Neoplasm Metastasis, Serum Amyloid P-Component genetics, Tumor Burden drug effects, Tumor Burden genetics, C-Reactive Protein metabolism, C-Reactive Protein pharmacology, Epithelial-Mesenchymal Transition drug effects, Melanoma metabolism, Melanoma pathology, Recombinant Proteins pharmacology, Serum Amyloid P-Component metabolism, Serum Amyloid P-Component pharmacology
- Abstract
During melanoma progression, malignant melanocytes are reprogrammed into mesenchymal-like cells through to an epithelial-mesenchymal transition (EMT) process associated with the acquisition of an invasive, prometastatic phenotype. The fibroblast growth factor-2 (FGF2)/FGF receptor (FGFR) system plays a pivotal role in melanoma, leading to autocrine/paracrine induction of tumor cell proliferation and angiogenesis. Long pentraxin-3 (PTX3) interacts with FGF2, and other FGF family members, inhibiting FGF-dependent neovascularization and tumor growth. Here, PTX3 protein and the PTX3-derived acetylated pentapeptide Ac-ARPCA-NH2 inhibit FGF2-driven proliferation and downstream FGFR signaling in murine melanoma B16-F10 cells. Moreover, human PTX3-overexpressing hPTX_B16-F10 cells are characterized by the reversed transition from a mesenchymal to an epithelial-like appearance, inhibition of cell proliferation, loss of clonogenic potential, reduced motility and invasive capacity, downregulation of various mesenchymal markers, and upregulation of the epithelial marker E-cadherin. Accordingly, PTX3 affects cell proliferation and EMT transition in human A375 and A2058 melanoma cells. Also, hPTX_B16-F10 cells showed a reduced tumorigenic and metastatic activity in syngeneic C57BL/6 mice. In conclusion, PTX3 inhibits FGF/FGFR-driven EMT in melanoma cells, hampering their tumorigenic and metastatic potential. These data represent the first experimental evidence about a nonredundant role of the FGF/FGFR system in the modulation of the EMT process in melanoma and indicate that PTX3 or its derivatives may represent the basis for the design of novel therapeutic approaches in FGF/FGFR-dependent tumors, including melanoma., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
38. Inhibition of angiogenesis by β-galactosylceramidase deficiency in globoid cell leukodystrophy.
- Author
-
Belleri M, Ronca R, Coltrini D, Nico B, Ribatti D, Poliani PL, Giacomini A, Alessi P, Marchesini S, Santos MB, Bongarzone ER, and Presta M
- Subjects
- Animals, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic metabolism, Aorta pathology, Aorta ultrastructure, Biocompatible Materials, Brain drug effects, Brain pathology, Brain ultrastructure, Cattle, Cell Movement drug effects, Cell Movement genetics, Chorioallantoic Membrane drug effects, Chorioallantoic Membrane metabolism, Collagen toxicity, Disease Models, Animal, Drug Combinations, Endothelial Cells drug effects, Endothelial Cells metabolism, Fibroblast Growth Factor 2 pharmacology, Green Fluorescent Proteins metabolism, Humans, Laminin toxicity, Leukodystrophy, Globoid Cell genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Electron, Transmission, Neovascularization, Pathologic prevention & control, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Proteoglycans toxicity, Psychosine metabolism, Psychosine pharmacology, RNA, Small Interfering administration & dosage, Time Factors, Transfection, Umbilical Veins cytology, Vascular Endothelial Growth Factor A pharmacology, Vascular Endothelial Growth Factor Receptor-2 genetics, Vascular Endothelial Growth Factor Receptor-2 metabolism, Zonula Occludens-1 Protein, Leukodystrophy, Globoid Cell complications, Neovascularization, Pathologic etiology, Neovascularization, Pathologic pathology
- Abstract
Globoid cell leukodystrophy (Krabbe disease) is a neurological disorder of infants caused by genetic deficiency of the lysosomal enzyme β-galactosylceramidase leading to accumulation of the neurotoxic metabolite 1-β-d-galactosylsphingosine (psychosine) in the central nervous system. Angiogenesis plays a pivotal role in the physiology and pathology of the brain. Here, we demonstrate that psychosine has anti-angiogenic properties by causing the disassembling of endothelial cell actin structures at micromolar concentrations as found in the brain of patients with globoid cell leukodystrophy. Accordingly, significant alterations of microvascular endothelium were observed in the post-natal brain of twitcher mice, an authentic model of globoid cell leukodystrophy. Also, twitcher endothelium showed a progressively reduced capacity to respond to pro-angiogenic factors, defect that was corrected after transduction with a lentiviral vector harbouring the murine β-galactosylceramidase complementary DNA. Finally, RNA interference-mediated β-galactosylceramidase gene silencing causes psychosine accumulation in human endothelial cells and hampers their mitogenic and motogenic response to vascular endothelial growth factor. Accordingly, significant alterations were observed in human microvasculature from brain biopsy of a globoid cell leukodystrophy case. Together these data demonstrate that β-galactosylceramidase deficiency induces significant alterations in endothelial neovascular responses that may contribute to central nervous system and systemic damages that occur in globoid cell leukodystrophy.
- Published
- 2013
- Full Text
- View/download PDF
39. Long pentraxin-3 as an epithelial-stromal fibroblast growth factor-targeting inhibitor in prostate cancer.
- Author
-
Ronca R, Alessi P, Coltrini D, Di Salle E, Giacomini A, Leali D, Corsini M, Belleri M, Tobia C, Garlanda C, Bonomi E, Tardanico R, Vermi W, and Presta M
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Cell Line, Tumor, Chick Embryo, Chorioallantoic Membrane blood supply, Chorioallantoic Membrane drug effects, Dihydrotestosterone pharmacology, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Nude, Mitogens antagonists & inhibitors, Neovascularization, Physiologic drug effects, Prostate metabolism, Prostate pathology, Prostatic Intraepithelial Neoplasia metabolism, Prostatic Intraepithelial Neoplasia pathology, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Recombinant Proteins pharmacology, Adenocarcinoma drug therapy, Antineoplastic Agents pharmacology, C-Reactive Protein pharmacology, Prostate drug effects, Prostatic Neoplasms drug therapy, Serum Amyloid P-Component pharmacology
- Abstract
Fibroblast growth factors (FGFs) exert autocrine/paracrine functions in prostate cancer by stimulating angiogenesis and tumour growth. Here dihydrotestosterone (DHT) up-regulates FGF2 and FGF8b production in murine TRAMP-C2 prostate cancer cells, activating a FGF-dependent autocrine loop of stimulation. The soluble pattern recognition receptor long pentraxin-3 (PTX3) acts as a natural FGF antagonist that binds FGF2 and FGF8b via its N-terminal domain. We demonstrate that recombinant PTX3 protein and the PTX3-derived pentapeptide Ac-ARPCA-NH2 abolish the mitogenic response of murine TRAMP-C2 cells and human LNCaP prostate cancer cells to DHT and FGFs. Also, PTX3 hampers the angiogenic activity of DHT-activated TRAMP-C2 cells on the chick embryo chorioallantoic membrane (CAM). Accordingly, human PTX3 overexpression inhibits the mitogenic activity exerted by DHT or FGFs on hPTX3_TRAMP-C2 cell transfectants and their angiogenic activity. Also, hPTX3_TRAMP-C2 cells show a dramatic decrease of their angiogenic and tumourigenic potential when grafted in syngeneic or immunodeficient athymic male mice. A similar inhibitory effect is observed when TRAMP-C2 cells overexpress only the FGF-binding N-terminal PTX3 domain. In keeping with the anti-tumour activity of PTX3 in experimental prostate cancer, immunohistochemical analysis of prostate needle biopsies from primary prostate adenocarcinoma patients shows that parenchymal PTX3 expression, abundant in basal cells of normal glands, is lost in high-grade prostatic intraepithelial neoplasia and in invasive tumour areas. These results identify PTX3 as a potent FGF antagonist endowed with anti-angiogenic and anti-neoplastic activity in prostate cancer., (Copyright © 2013 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
40. Matrigel plug assay: evaluation of the angiogenic response by reverse transcription-quantitative PCR.
- Author
-
Coltrini D, Di Salle E, Ronca R, Belleri M, Testini C, and Presta M
- Subjects
- Drug Combinations, Humans, Immunohistochemistry, Reverse Transcriptase Polymerase Chain Reaction, Collagen, Laminin, Neovascularization, Physiologic, Proteoglycans
- Abstract
The subcutaneous Matrigel plug assay in mice is a method of choice for the in vivo evaluation of pro- and anti-angiogenic molecules. However, quantification of the angiogenic response in the plug remains a problematic task. Here we report a simple, rapid, unbiased and reverse transcription-quantitative PCR (RT-qPCR) method to investigate the angiogenic process occurring in the Matrigel plug in response to fibroblast growth factor-2 (FGF2). To this purpose, a fixed amount of human cells were added to harvested plugs at the end of the in vivo experimentation as an external cell tracer. Then, mRNA levels of the pan-endothelial cell markers murine CD31 and vascular endothelial-cadherin were measured by species-specific RT-qPCR analysis of the total RNA and data were normalized for human GAPDH or β-actin mRNA levels. RT-qPCR was used also to measure the levels of expression in the plug of various angiogenesis/inflammation-related genes. The procedure allows the simultaneous, quantitative evaluation of the newly-formed endothelium and of non-endothelial/inflammatory components of the cellular infiltrate in the Matrigel implant, as well as the expression of genes involved in the modulation of the angiogenesis process. Also, the method consents the quantitative assessment of the effect of local or systemic administration of anti-angiogenic compounds on the neovascular response triggered by FGF2.
- Published
- 2013
- Full Text
- View/download PDF
41. Long pentraxin 3/tumor necrosis factor-stimulated gene-6 interaction: a biological rheostat for fibroblast growth factor 2-mediated angiogenesis.
- Author
-
Leali D, Inforzato A, Ronca R, Bianchi R, Belleri M, Coltrini D, Di Salle E, Sironi M, Norata GD, Bottazzi B, Garlanda C, Day AJ, and Presta M
- Subjects
- Animals, Binding, Competitive, C-Reactive Protein deficiency, C-Reactive Protein genetics, CHO Cells, Cattle, Cell Adhesion Molecules genetics, Chick Embryo, Cricetinae, Cricetulus, Female, Fibroblast Growth Factor 2 genetics, HEK293 Cells, Heparan Sulfate Proteoglycans metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins genetics, Protein Binding, Protein Interaction Domains and Motifs, Protein Interaction Mapping, Recombinant Proteins metabolism, Serum Amyloid P-Component genetics, Surface Plasmon Resonance, Transfection, C-Reactive Protein metabolism, Cell Adhesion Molecules metabolism, Fibroblast Growth Factor 2 metabolism, Neovascularization, Physiologic, Nerve Tissue Proteins metabolism, Serum Amyloid P-Component metabolism
- Abstract
Objective: Angiogenesis is regulated by the balance between pro- and antiangiogenic factors and by extracellular matrix protein interactions. Fibroblast growth factor 2 (FGF2) is a major proangiogenic inducer inhibited by the interaction with the soluble pattern recognition receptor long pentraxin 3 (PTX3). PTX3 is locally coexpressed with its ligand tumor necrosis factor-stimulated gene-6 (TSG-6), a secreted glycoprotein that cooperates with PTX3 in extracellular matrix assembly. Here, we characterized the effect of TSG-6 on PTX3/FGF2 interaction and FGF2-mediated angiogenesis., Methods and Results: Solid phase binding and surface plasmon resonance assays show that TSG-6 and FGF2 bind the PTX3 N-terminal domain with similar affinity. Accordingly, TSG-6 prevents FGF2/PTX3 interaction and suppresses the inhibition exerted by PTX3 on heparan sulfate proteoglycan/FGF2/FGF receptor complex formation and on FGF2-dependent angiogenesis in vitro and in vivo. Also, endogenous PTX3 exerts an inhibitory effect on vascularization induced by FGF2 in a murine subcutaneous Matrigel plug assay, the inhibition being abolished in Ptx3-null mice or by TSG-6 treatment in wild-type animals., Conclusion: TSG-6 reverts the inhibitory effects exerted by PTX3 on FGF2-mediated angiogenesis through competition of FGF2/PTX3 interaction. This may provide a novel mechanism to control angiogenesis in those pathological settings characterized by the coexpression of TSG-6 and PTX3, in which the relative levels of these proteins may fine-tune the angiogenic activity of FGF2.
- Published
- 2012
- Full Text
- View/download PDF
42. Long pentraxin-3 inhibits FGF8b-dependent angiogenesis and growth of steroid hormone-regulated tumors.
- Author
-
Leali D, Alessi P, Coltrini D, Ronca R, Corsini M, Nardo G, Indraccolo S, and Presta M
- Subjects
- Angiogenesis Inhibitors metabolism, Animals, C-Reactive Protein genetics, C-Reactive Protein metabolism, CHO Cells, Cattle, Cell Line, Cell Proliferation drug effects, Chick Embryo, Cricetinae, Fibroblast Growth Factor 8 metabolism, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, Mice, Inbred C57BL, Mice, SCID, Neoplasms metabolism, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Protein Binding, Serum Amyloid P-Component genetics, Serum Amyloid P-Component metabolism, Androgens physiology, Angiogenesis Inhibitors pharmacology, C-Reactive Protein pharmacology, Fibroblast Growth Factor 8 antagonists & inhibitors, Serum Amyloid P-Component pharmacology
- Abstract
Fibroblast growth factor-8b (FGF8b) exerts nonredundant autocrine/paracrine functions in steroid hormone-regulated tumors. Previous observations had shown that the soluble pattern recognition receptor long pentraxin-3 (PTX3) is a natural selective antagonist for a restricted number of FGF family members, inhibiting FGF2 but not FGF1 and FGF4 activity. Here, we assessed the capacity of PTX3 to antagonize FGF8b and to inhibit the vascularization and growth of steroid hormone-regulated tumors. Surface plasmon resonance analysis shows that PTX3 binds FGF8b with high affinity (K(d) = 30-90 nmol/L). As a consequence, PTX3 prevents the binding of FGF8b to its receptors, inhibits FGF8b-driven ERK1/2 activation, cell proliferation, and chemotaxis in endothelial cells, and suppresses FGF8b-induced neovascularization in vivo. Also, PTX3 inhibits dihydrotestosterone (DHT)- and FGF8b-driven proliferation of androgen-regulated Shionogi 115 (S115) mouse breast tumor cells. Furthermore, DHT-treated, PTX3 overexpressing hPTX3_S115 cell transfectants show a reduced proliferation rate in vitro and a limited angiogenic activity in the chick embryo chorioallantoic membrane and murine s.c. Matrigel plug assays. Accordingly, hPTX3_S115 cells show a dramatic decrease of their tumorigenic activity when grafted in immunodeficient male mice. These results identify PTX3 as a novel FGF8b antagonist endowed with antiangiogenic and antineoplastic activity with possible implications for the therapy of hormonal tumors.
- Published
- 2011
- Full Text
- View/download PDF
43. Antiangiogenic activity of a neutralizing human single-chain antibody fragment against fibroblast growth factor receptor 1.
- Author
-
Ronca R, Benzoni P, Leali D, Urbinati C, Belleri M, Corsini M, Alessi P, Coltrini D, Calza S, Presta M, and Dell'Era P
- Subjects
- Angiogenesis Inhibitors therapeutic use, Animals, Antibodies, Neutralizing therapeutic use, Antibody Specificity drug effects, Cattle, Cell Proliferation drug effects, Chick Embryo, Female, Humans, In Vitro Techniques, Mammary Neoplasms, Experimental blood supply, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental pathology, Mice, Neovascularization, Pathologic drug therapy, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Single-Chain Antibodies therapeutic use, Angiogenesis Inhibitors pharmacology, Antibodies, Neutralizing pharmacology, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Single-Chain Antibodies pharmacology
- Abstract
Fibroblast growth factor receptor-1 (FGFR-1) transduces proangiogenic and proliferative signals in human cancers. Thus, FGFR-1 may represent a target for the development of antiangiogenic/antineoplastic therapies. We screened a human single-chain fragment variable (scFv) antibody phage display library against the extracellular domain of the FGFR-1-IIIc isoform that harbors the FGF binding site. Several phages were isolated and tested for specificity and sensitivity, and the most promising antibody fragment RR-C2 was characterized for its biochemical and biological properties. ScFv RR-C2 specifically recognizes FGFR-1α and FGFR-1β isoforms in ELISA, Western blotting, and surface plasmon resonance analysis with a K(d) value of 300 and 144 nmol/L for the 2 receptor isoforms, respectively. The antibody fragment also recognizes FGFR-1 when the receptor is exposed on the cell surface, thus preventing the formation of the ternary complex among FGFR-1, its ligand FGF2, and cell surface heparan sulfate proteoglycans. Accordingly, scFv RR-C2 specifically inhibits FGF2-mediated mitogenic activity in endothelial cells of human, bovine, and murine origin in a nanomolar range of concentrations. Also, the antibody fragment prevents FGF2-triggered sprouting of both human umbilical vein endothelial cell spheroids and of murine endothelium from aortic rings. Finally, the antibody fragment hampers the angiogenic activity exerted both by FGF2 in the chick embryo chorioallantoic membrane assay and by S115 mouse mammary tumor cells in the Matrigel plug assay. Taken together, the data show that scFv RR-C2 recognizes and neutralizes FGFR-1 activity in different animal species, including humans, thus representing a novel tool for the development of antiangiogenic/antineoplastic therapies., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
44. Impact of VEGF-dependent tumour micro-environment on EDB fibronectin expression by subcutaneous human tumour xenografts in nude mice.
- Author
-
Coltrini D, Ronca R, Belleri M, Zardi L, Indraccolo S, Scarlato V, Giavazzi R, and Presta M
- Subjects
- Adenocarcinoma blood supply, Adenocarcinoma pathology, Angiogenesis Inhibitors therapeutic use, Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Bevacizumab, Endometrial Neoplasms blood supply, Endometrial Neoplasms pathology, Female, Fibroblast Growth Factor 2 metabolism, Fibronectins genetics, Gene Expression Regulation, Neoplastic, Humans, Kidney Neoplasms blood supply, Kidney Neoplasms pathology, Kidney Neoplasms prevention & control, Mice, Mice, Nude, Neoplasm Proteins metabolism, Neoplasm Proteins physiology, Neoplasm Transplantation, Neovascularization, Pathologic metabolism, Protein Isoforms metabolism, RNA, Messenger genetics, RNA, Neoplasm genetics, Reverse Transcriptase Polymerase Chain Reaction methods, Transplantation, Heterologous, Adenocarcinoma metabolism, Endometrial Neoplasms metabolism, Fibronectins metabolism, Vascular Endothelial Growth Factor A physiology
- Abstract
Fibronectin (FN) is an extracellular matrix cell-adhesive glycoprotein. The alternative spliced isoform EDB-FN (extra domain B containing FN) is highly expressed in tumour blood vessels and stroma and represents a candidate for tumour targeting. To investigate the impact of different angiogenic micro-environments on EDB-FN expression, we used a tumour model in which human endometrial adenocarcinoma Tet-FGF2 cells overexpressing fibroblast growth factor-2 (FGF2) driven by the tetracycline-responsive promoter were further transfected with a VEGF antisense cDNA, generating AS-VEGF/Tet-FGF2 cells. In this model, the expression of FGF2 plus VEGF results in fast-growing, highly vascularized Tet-FGF2 tumours. Down-regulation of FGF2 production by tetracycline administration and/or of VEGF production by AS-VEGF transduction inhibited tumour growth and vascularization, with profound changes in tumour micro-environment. Quantitative RT-PCR analysis using human EDB-FN primers shows that subcutaneous grafting in immunodeficient mice is per se sufficient to cause a dramatic up-regulation of EDB-FN expression by these cells, as well as by human oesophageal cancer KYSE 30 cells and renal carcinoma Caki-1 cells. However, in vivo down-regulation of VEGF expression, as occurs in AS-VEGF/Tet-FGF2 tumours, and to a lesser extent of FGF2 expression, as occurs in tetracycline-treated Tet-FGF2 tumour-bearing animals, causes significant inhibition of EDB-FN production in tumour grafts, as shown by immunohistochemistry and quantitative RT-PCR analysis. Accordingly, treatment of Tet-FGF2 tumour-bearing animals with the neutralizing anti-murine VEGF receptor-2 antibody DC101, or of Caki-1 tumour-bearing animals with the anti-VEGF antibody bevacizumab, inhibited EDB-FN expression in tumour grafts. EDB-FN down-regulation was paralleled by a decrease in vascularity, thus confirming EDB-FN as a marker of tumour angiogenesis. These data demonstrate that the angiogenic micro-environment, and in particular the VEGF/VEGFR-2 system, plays a key role in modulating EDB-FN expression by tumour cells in vivo. This may have implications for the design of therapeutic strategies targeting EDB-FN in combination with anti-angiogenic and/or cytotoxic drugs.
- Published
- 2009
- Full Text
- View/download PDF
45. A pro-inflammatory signature mediates FGF2-induced angiogenesis.
- Author
-
Andrés G, Leali D, Mitola S, Coltrini D, Camozzi M, Corsini M, Belleri M, Hirsch E, Schwendener RA, Christofori G, Alcamì A, and Presta M
- Subjects
- Animals, Base Sequence, Chick Embryo, DNA Primers, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases physiology, Polymerase Chain Reaction, Recombinant Proteins pharmacology, Fibroblast Growth Factor 2 pharmacology, Neovascularization, Physiologic drug effects
- Abstract
Fibroblast growth factor-2 (FGF2) is a potent angiogenic growth factor. Here, gene expression profiling of FGF2-stimulated microvascular endothelial cells revealed, together with a prominent pro-angiogenic profile, a pro-inflammatory signature characterized by the upregulation of pro-inflammatory cytokine/chemokines and their receptors, endothelial cell adhesion molecules and members of the eicosanoid pathway. Real-time quantitative PCR demonstrated early induction of most of the FGF2-induced, inflammation-related genes. Accordingly, chick embryo chorioallantoic membrane (CAM) and murine Matrigel plug angiogenesis assays demonstrated a significant monocyte/macrophage infiltrate in the areas of FGF2-driven neovascularization. Similar results were obtained when the conditioned medium (CM) of FGF2-stimulated endothelial cells was delivered onto the CAM, suggesting that FGF2-upregulated chemoattractants mediate the inflammatory response. Importantly, FGF2-triggered new blood vessel formation was significantly reduced in phosphatidylinositol 3-kinase-gamma null mice exhibiting defective leucocyte migration or in clodronate liposome-treated, macrophage-depleted mice. Furthermore, the viral pan-chemokine antagonist M3 inhibited the angiogenic and inflammatory responses induced by the CM of FGF2-stimulated endothelial cells and impaired FGF2-driven neovascularization in the CAM assay. These findings point to inflammatory chemokines as early mediators of FGF2-driven angiogenesis and indicate a non-redundant role for inflammatory cells in the neovascularization process elicited by the growth factor.
- Published
- 2009
- Full Text
- View/download PDF
46. Cardiac microvascular endothelial cells express a functional Ca+ -sensing receptor.
- Author
-
Berra Romani R, Raqeeb A, Laforenza U, Scaffino MF, Moccia F, Avelino-Cruz JE, Oldani A, Coltrini D, Milesi V, Taglietti V, and Tanzi F
- Subjects
- Animals, Calcium Signaling drug effects, Endothelial Cells drug effects, Estrenes pharmacology, Gadolinium pharmacology, Indoles pharmacology, Lanthanum pharmacology, Meglumine pharmacology, Neomycin pharmacology, Phenylalanine pharmacology, Pyrrolidinones pharmacology, RNA, Messenger metabolism, Rats, Rats, Wistar, Receptors, Calcium-Sensing agonists, Reverse Transcriptase Polymerase Chain Reaction, Sodium physiology, Spermine pharmacology, Tryptophan pharmacology, Type C Phospholipases physiology, Calcium Signaling physiology, Endothelial Cells metabolism, Myocytes, Cardiac metabolism, Receptors, Calcium-Sensing genetics
- Abstract
The mechanism whereby extracellular Ca(2+) exerts the endothelium-dependent control of vascular tone is still unclear. In this study, we assessed whether cardiac microvascular endothelial cells (CMEC) express a functional extracellular Ca(2+)-sensing receptor (CaSR) using a variety of techniques. CaSR mRNA was detected using RT-PCR, and CaSR protein was identified by immunocytochemical analysis. In order to assess the functionality of the receptor, CMEC were loaded with the Ca(2+)-sensitive fluorochrome, Fura-2/AM. A number of CaSR agonists, such as spermine, Gd(3+), La(3+) and neomycin, elicited a heterogeneous intracellular Ca(2+) signal, which was abolished by disruption of inositol 1,4,5-trisphosphate (InsP(3)) signaling and by depletion of intracellular stores with cyclopiazonic acid. The inhibition of the Na(+)/Ca(2+) exchanger upon substitution of extracellular Na(+) unmasked the Ca(2+) signal triggered by an increase in extracellular Ca(2+) levels. Finally, aromatic amino acids, which function as allosteric activators of CaSR, potentiated the Ca(2+) response to the CaSR agonist La(3+). These data provide evidence that CMEC express CaSR, which is able to respond to physiological agonists by mobilizing Ca(2+) from intracellular InsP(3)-sensitive stores., (Copyright 2008 S. Karger AG, Basel.)
- Published
- 2009
- Full Text
- View/download PDF
47. alphavbeta3 Integrin-dependent antiangiogenic activity of resveratrol stereoisomers.
- Author
-
Belleri M, Ribatti D, Savio M, Stivala LA, Forti L, Tanghetti E, Alessi P, Coltrini D, Bugatti A, Mitola S, Nicoli S, Vannini V, and Presta M
- Subjects
- Angiogenesis Inhibitors chemistry, Animals, Antineoplastic Agents, Phytogenic chemistry, Cattle, Chick Embryo, Endothelial Cells cytology, Female, Humans, Melanoma, Experimental drug therapy, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Resveratrol, Stereoisomerism, Stilbenes chemistry, Angiogenesis Inhibitors pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Integrin alphaVbeta3 metabolism, Neovascularization, Pathologic, Stilbenes pharmacology
- Abstract
Angiogenesis is target for antineoplastic and chemopreventive therapies. The natural phytoalexin resveratrol is found in grapes and red wine as cis and trans stereoisomers. trans-Resveratrol shows antiangiogenic activity, but its mechanism of action is not fully elucidated. Recently, trans-resveratrol has been shown to interact with the beta3 integrin subunit, raising the possibility that inhibition of endothelial alphavbeta3 integrin function may concur to its angiosuppressive activity. To get novel insights about the antiangiogenic activity of resveratrol, we compared cis- and trans-resveratrol stereoisomers for their effect on the angiogenesis process and endothelial alphavbeta3 integrin function. trans-Resveratrol inhibits endothelial cell proliferation and the repair of mechanically wounded endothelial cell monolayers. Also, it prevents endothelial cell sprouting in fibrin gel, collagen gel invasion, and morphogenesis on Matrigel. In vivo, trans-resveratrol inhibits vascularization of the chick embryo area vasculosa and murine melanoma B16 tumor growth and neovascularization. In all the assays, cis-resveratrol exerts a limited, if any, effect. In keeping with these observations, trans-resveratrol, but not cis-resveratrol, inhibits alphavbeta3 integrin-dependent endothelial cell adhesion and the recruitment of enhanced green fluorescent protein-tagged beta3 integrin in focal adhesion contacts. In conclusion, stereoisomery affects the antiangiogenic activity of resveratrol, the trans isomer being significantly more potent than the cis isoform. The different antiangiogenic potential of resveratrol stereoisomers is related, at least in part, to their different capacity to affect alphavbeta3 integrin function. This may have profound implications for the design of synthetic antiangiogenic/angiopreventive phytoalexin derivatives.
- Published
- 2008
- Full Text
- View/download PDF
48. Bone morphogenic protein antagonist Drm/gremlin is a novel proangiogenic factor.
- Author
-
Stabile H, Mitola S, Moroni E, Belleri M, Nicoli S, Coltrini D, Peri F, Pessi A, Orsatti L, Talamo F, Castronovo V, Waltregny D, Cotelli F, Ribatti D, and Presta M
- Subjects
- Amino Acid Sequence, Angiogenesis Inducing Agents chemistry, Angiogenesis Inducing Agents isolation & purification, Angiogenesis Inducing Agents metabolism, Animals, Bone Morphogenetic Proteins metabolism, Cells, Cultured, Chick Embryo, Cytokines, Endothelial Cells metabolism, Immunohistochemistry, Intercellular Signaling Peptides and Proteins chemistry, Intercellular Signaling Peptides and Proteins isolation & purification, Mice, Molecular Sequence Data, Neoplasms blood supply, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Protein Binding, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Angiogenesis Inducing Agents pharmacology, Bone Morphogenetic Proteins antagonists & inhibitors, Intercellular Signaling Peptides and Proteins metabolism, Intercellular Signaling Peptides and Proteins pharmacology
- Abstract
Angiogenesis plays a key role in various physiologic and pathologic conditions, including tumor growth. Drm/gremlin, a member the Dan family of bone morphogenic protein (BMP) antagonists, is commonly thought to affect different processes during growth, differentiation, and development by heterodimerizing various BMPs. Here, we identify Drm/gremlin as a novel proangiogenic factor expressed by endothelium. Indeed, Drm/gremlin was purified to homogeneity from the conditioned medium of transformed endothelial cells using an endothelial-cell sprouting assay to follow protein isolation. Accordingly, recombinant Drm/gremlin stimulates endothelial-cell migration and invasion in fibrin and collagen gels, binds with high affinity to various endothelial cell types, and triggers tyrosine phosphorylation of intracellular signaling proteins. Also, Drm/gremlin induces neovascularization in the chick embryo chorioallantoic membrane. BMP4 does not affect Drm/gremlin interaction with endothelium, and both molecules exert a proangiogenic activity in vitro and in vivo when administered alone or in combination. Finally, Drm/gremlin is produced by the stroma of human tumor xenografts in nude mice, and it is highly expressed in endothelial cells of human lung tumor vasculature when compared with non-neoplastic lung. Our observations point to a novel, previously unrecognized capacity of Drm/gremlin to interact directly with target endothelial cells and to modulate angiogenesis.
- Published
- 2007
- Full Text
- View/download PDF
49. Cutting edge: extracellular high mobility group box-1 protein is a proangiogenic cytokine.
- Author
-
Mitola S, Belleri M, Urbinati C, Coltrini D, Sparatore B, Pedrazzi M, Melloni E, and Presta M
- Subjects
- Animals, Cattle, Cell Proliferation, Chemotaxis physiology, Chick Embryo, Enzyme Activation physiology, MAP Kinase Kinase 1 metabolism, MAP Kinase Kinase 2 metabolism, Endothelial Cells metabolism, HMGB1 Protein metabolism, Neovascularization, Pathologic metabolism
- Abstract
The chromosomal high mobility group box-1 (HMGB1) protein acts as a proinflammatory cytokine when released in the extracellular environment by necrotic and inflammatory cells. In the present study, we show that HMGB1 exerts proangiogenic effects by inducing MAPK ERK1/2 activation, cell proliferation, and chemotaxis in endothelial cells of different origin. Accordingly, HMGB1 stimulates membrane ruffling and repair of a mechanically wounded endothelial cell monolayer and causes endothelial cell sprouting in a three-dimensional fibrin gel. In keeping with its in vitro properties, HMGB1 stimulates neovascularization when applied in vivo on the top of the chicken embryo chorioallantoic membrane whose blood vessels express the HMGB1 receptor for advanced glycation end products (RAGE). Accordingly, RAGE blockade by neutralizing Abs inhibits HMGB1-induced neovascularization in vivo and endothelial cell proliferation and membrane ruffling in vitro. Taken together, the data identify HMGB1/RAGE interaction as a potent proangiogenic stimulus.
- Published
- 2006
- Full Text
- View/download PDF
50. Pentraxin 3 inhibits fibroblast growth factor 2-dependent activation of smooth muscle cells in vitro and neointima formation in vivo.
- Author
-
Camozzi M, Zacchigna S, Rusnati M, Coltrini D, Ramirez-Correa G, Bottazzi B, Mantovani A, Giacca M, and Presta M
- Subjects
- Animals, C-Reactive Protein genetics, C-Reactive Protein pharmacology, Carotid Artery Injuries pathology, Catheterization adverse effects, Cell Division drug effects, Cell Division physiology, Cell Survival physiology, Cells, Cultured, Chemotaxis drug effects, Humans, Hyperplasia, In Vitro Techniques, Iodine Radioisotopes, Male, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular drug effects, Rats, Rats, Wistar, Serum Amyloid P-Component genetics, Serum Amyloid P-Component pharmacology, Transduction, Genetic, Tunica Intima cytology, Tunica Intima drug effects, Tunica Intima metabolism, C-Reactive Protein metabolism, Carotid Artery Injuries metabolism, Coronary Vessels cytology, Fibroblast Growth Factor 2 pharmacokinetics, Muscle, Smooth, Vascular metabolism, Serum Amyloid P-Component metabolism
- Abstract
Objective: The fibroblast growth factor (FGF)/FGF receptor system plays an important role in smooth muscle cell (SMC) activation. Long-pentraxin 3 (PTX3) is a soluble pattern recognition receptor with non-redundant functions in inflammation and innate immunity. PTX3 is produced by different cell types of the vessel wall, including SMCs. PTX3 binds FGF2 and inhibits its angiogenic activity on endothelial cells. We investigated the capacity of PTX3 to affect FGF2-dependent SMC activation in vitro and in vivo., Methods and Results: When added to human coronary artery SMCs, human PTX3 inhibits cell proliferation driven by endogenous FGF2 and the mitogenic and chemotactic activity exerted by exogenous recombinant FGF2. Accordingly, PTX3 prevents (125)I-FGF2 interaction with FGF receptors on the same cells. Also, PTX3 overexpression after recombinant adeno-associated virus-PTX3 gene transfer inhibits human coronary artery SMC proliferation and survival promoted by FGF2 in vitro. Consistently, a single local endovascular injection of recombinant adeno-associated virus-PTX3 gene inhibits intimal thickening after balloon injury in rat carotid arteries., Conclusions: PTX3 is a potent inhibitor of the autocrine and paracrine stimulation exerted by FGF2 on SMCs. Local PTX3 upregulation may modulate SMC activation after arterial injury.
- 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.