@article {7727, title = {TNF-related apoptosis inducing ligand in ocular cancers and ocular diabetic complications.}, journal = {Biomed Res Int}, volume = {2015}, year = {2015}, month = {2015}, pages = {424019}, abstract = {

TNF-related apoptosis inducing ligand (TRAIL) is an intensively studied cytokine, in particular for its anticancer activity. The discovery that conjunctival sac fluid contains extremely high levels of soluble TRAIL as compared to other body fluids suggested important implications in the context of the immunological surveillance of the eye, in particular of the anterior surface. In this review, we discuss the potential physiopathologic and therapeutic role of the TRAIL/TRAIL receptor system in a variety of ocular cancers. Moreover, since an increasing amount of data has indicated the important biological activities of the TRAIL/TRAIL receptor systems also in a completely different pathologic context such as diabetes mellitus, in the second part of this review we summarize the currently available data on the involvement of TRAIL in the ocular complications of diabetes mellitus as modulator of the inflammatory and angiogenic response in the eye.

}, keywords = {Apoptosis, Diabetes Complications, Diabetes Mellitus, Eye Neoplasms, Humans, Receptors, TNF-Related Apoptosis-Inducing Ligand, TNF-Related Apoptosis-Inducing Ligand}, issn = {2314-6141}, doi = {10.1155/2015/424019}, author = {Perri, Paolo and Zauli, Giorgio and Gonelli, Arianna and Milani, Daniela and Celeghini, Claudio and Lamberti, Giuseppe and Secchiero, Paola} } @article {3527, title = {Soluble TRAIL is present at high concentrations in seminal plasma and promotes spermatozoa survival.}, journal = {Reproduction}, volume = {148}, year = {2014}, month = {2014 Aug}, pages = {191-8}, abstract = {

The expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL(TNFSF10)) and of its receptors (TRAILR1, TRAILR2, TRAILR3, and TRAILR4) have been documented in testis, but the presence of soluble TRAIL in seminal fluid, as well as the potential physiopathological role of the TRAIL/TRAILR system in spermatozoa, has not been previously investigated. Male donors (n=123) among couples presenting for infertility evaluation were consecutively enrolled in this study. The presence of soluble TRAIL was analyzed in seminal samples by ELISA, while the surface expression of TRAIL receptors was investigated by flow cytometry. High levels of soluble TRAIL were detected in seminal plasma (median, 11 621 pg/ml and mean{\textpm}s.d., 13 371{\textpm}8367 pg/ml) and flow cytometric analysis revealed a variable expression of TRAIL receptors in the sperm cellular fraction among different subjects. In addition, the effect of physiologically relevant concentrations of recombinant TRAIL was investigated on survival and motility of spermatozoa. Of interest, the in vitro exposure of capacitated spermatozoa to recombinant TRAIL (10 ng/ml) significantly preserved their overall survival. Therefore, the present study demonstrates for the first time the presence of elevated levels of the anti-inflammatory cytokine TRAIL in seminal fluids. Moreover, the demonstration that recombinant TRAIL promotes spermatozoa survival after capacitation suggests potential therapeutic implications.

}, keywords = {Adult, Apoptosis, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Humans, Infertility, Male, Male, Receptors, TNF-Related Apoptosis-Inducing Ligand, Semen, Sperm Capacitation, Sperm Motility, Spermatozoa, TNF-Related Apoptosis-Inducing Ligand}, issn = {1741-7899}, doi = {10.1530/REP-14-0144}, author = {Zauli, Giorgio and Celeghini, Claudio and Monasta, Lorenzo and Martinelli, Monica and Luppi, Stefania and Gonelli, Arianna and Grill, Vittorio and Ricci, Giuseppe and Secchiero, Paola} } @article {1769, title = {Activation of the p53 pathway induces α-smooth muscle actin expression in both myeloid leukemic cells and normal macrophages.}, journal = {J Cell Physiol}, volume = {227}, year = {2012}, month = {2012 May}, pages = {1829-37}, abstract = {

A range of cell types of mesenchymal origin express α-smooth muscle actin (α-SMA), a protein that plays a key role in controlling cell motility and differentiation along the fibrocyte and myofibroblast lineages. Although α-SMA is often expressed in stromal cells associated to a variety of cancers including hematological malignancies, up to now the role of anti-cancer drugs on α-SMA has not been deeply investigated. In this study, we demonstrated that Nutlin-3, the small molecule inhibitor of the MDM2/p53 interactions, significantly up-regulated the mRNA and protein levels of α-SMA in normal macrophages as well as in p53(wild-type) but not in p53(mutated/null) myeloid leukemic cells. The p53-dependence of α-SMA up-regulation induced by Nutlin-3 was demonstrated in experiments performed with siRNA for p53. Of note, Nutlin-3 mediated up-regulation of α-SMA in OCI leukemic cells was accompanied by cell adhesion to plastic substrate and by reduced cell migratory response in transwell assays. Notably, the role of α-SMA induction in the modulation of myeloid cell migration was clearly documented in α-SMA gene knockdown experiments. In addition, Nutlin-3 significantly up-regulated α-SMA expression in primary endothelial cells, but not in fibroblasts and mesenchymal stem cells (MSC). Conversely, transforming growth factor-β1 up-regulated α-SMA in fibroblasts and MSC, but not in macrophages and endothelial cells. Taken together, these data indicate that Nutlin-3 is a potent inducer of α-SMA in both normal and leukemic myeloid cells as well as in endothelial cells.

}, keywords = {Actins, Cell Movement, Cells, Cultured, Endothelial Cells, Fibroblasts, Humans, Imidazoles, Leukemia, Myeloid, Macrophages, Mesenchymal Stromal Cells, Piperazines, Proto-Oncogene Proteins c-mdm2, RNA, Small Interfering, Signal Transduction, Transforming Growth Factor beta1, Tumor Suppressor Protein p53}, issn = {1097-4652}, doi = {10.1002/jcp.22910}, author = {Secchiero, Paola and Rimondi, Erika and di Iasio, Maria Grazia and Voltan, Rebecca and Gonelli, Arianna and Zauli, Giorgio} } @article {1738, title = {Hydrogen sulfide down-regulates the expression and release of osteoprotegerin (OPG) by vascular endothelial cells.}, journal = {Invest New Drugs}, volume = {30}, year = {2012}, month = {2012 Aug}, pages = {1731-5}, keywords = {Cell Death, Cytostatic Agents, Down-Regulation, Human Umbilical Vein Endothelial Cells, Humans, Hydrogen Sulfide, Osteoprotegerin, Tumor Necrosis Factor-alpha}, issn = {1573-0646}, doi = {10.1007/s10637-011-9675-8}, author = {Rimondi, Erika and di Iasio, Maria Grazia and Gonelli, Arianna and Celeghini, Claudio and Secchiero, Paola and Zauli, Giorgio} } @article {1667, title = {Pegylated TRAIL retains anti-leukemic cytotoxicity and exhibits improved signal transduction activity with respect to TRAIL.}, journal = {Invest New Drugs}, volume = {30}, year = {2012}, month = {2012 Apr}, pages = {828-32}, abstract = {

To improve the pharmacokinetic profile of tumor necrosis factor-related apoptosis inducing ligand (TRAIL) an N-terminal specific pegylation was performed to generate pegylated TRAIL (PEG-TRAIL). In in vitro experiments, we found that although PEG-TRAIL was slightly less efficient than recombinant TRAIL in promoting leukemic cell apoptosis, it showed an improved ability to promote migration of bone-marrow mesenchymal stem cells and to elicit the ERK1/2 intracellular signal transduction pathway. Overall, these data suggest that TRAIL pegylation retains, or even enhances, the biological activities of TRAIL relevant for its therapeutic applications.

}, keywords = {Antineoplastic Agents, Apoptosis, Caspase 3, Cell Movement, Dose-Response Relationship, Drug, HL-60 Cells, Humans, Leukemia, Mesenchymal Stromal Cells, Mitogen-Activated Protein Kinase 1, Mitogen-Activated Protein Kinase 3, Phosphorylation, Polyethylene Glycols, Recombinant Fusion Proteins, Signal Transduction, Time Factors, TNF-Related Apoptosis-Inducing Ligand}, issn = {1573-0646}, doi = {10.1007/s10637-010-9599-8}, author = {Gonelli, Arianna and Radillo, Oriano and Drioli, Sara and Rimondi, Erika and Secchiero, Paola and Maria Bonora, Gian} } @article {1817, title = {Trail down-regulates the release of osteoprotegerin (OPG) by primary stromal cells.}, journal = {J Cell Physiol}, volume = {226}, year = {2011}, month = {2011 Sep}, pages = {2279-86}, abstract = {

The soluble member of the TNF-R superfamily osteoprotegerin (OPG) is abundantly released under basal conditions by both mesenchymal stem cells (MSC) and fibroblasts and by endothelial cells upon stimulation with inflammatory cytokines. Since MSC, fibroblasts and endothelial cells represent key elements of the normal and tumor microenvironment and express detectable levels of surface TRAIL receptors, we investigated the effect of TRAIL on OPG release. Unexpectedly, recombinant TRAIL decreased the spontaneous OPG release in all cell types examined. Moreover, TRAIL decreased OPG release also in stromal cells co-cultured with lymphoma cells and counteracted the OPG induction by TN-alpha in HUVEC and MSC. Such down-regulation was not due to a masking effect in the ELISA quantification of the OPG released in the culture supernatants due to binding of OPG to its ligands (TRAIL and RANKL), as demonstrated by competition experiments with recombinant TRAIL and by the lack of RANKL release/induction. In addition, OPG down-regulation was not due to induction of cytotoxic effects by TRAIL, since the degree of apoptosis in response to TRAIL was negligible in all primary cell types. With regards to the possible molecular mechanism accounting for the down-regulation of OPG release by TRAIL, we found that treatment of MSC with TRAIL significantly decreased the phosphorylation levels of p38/MAPK. There is a suggestion that this pathway is involved in the stabilization of OPG mRNA. In this respect, the ability of TRAIL to decrease the release of OPG, in the absence of cell cytotoxicity, was mimicked by the p38/MAPK inhibitor SB203580.

}, keywords = {Bone Marrow Cells, Cell Death, Cells, Cultured, Coculture Techniques, Down-Regulation, Endothelial Cells, Enzyme Activation, Enzyme-Linked Immunosorbent Assay, Fibroblasts, Humans, MAP Kinase Signaling System, Mesenchymal Stromal Cells, Osteoprotegerin, p38 Mitogen-Activated Protein Kinases, Protein Binding, Recombinant Proteins, Stromal Cells, TNF-Related Apoptosis-Inducing Ligand, Tumor Necrosis Factor-alpha}, issn = {1097-4652}, doi = {10.1002/jcp.22564}, author = {Corallini, Federica and Celeghini, Claudio and Rimondi, Erika and di Iasio, Maria Grazia and Gonelli, Arianna and Secchiero, Paola and Zauli, Giorgio} }